1
|
Almutairy BK, Khafagy ES, Aldawsari MF, Alshetaili A, Alotaibi HF, Abu Lila AS. Tailoring of Bilosomal Nanogel for Augmenting the Off-Label Use of Sildenafil Citrate in Pediatric Pulmonary Hypertension. ACS OMEGA 2024; 9:19536-19547. [PMID: 38708263 PMCID: PMC11064047 DOI: 10.1021/acsomega.4c01133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/09/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Pediatric pulmonary hypertension is a serious syndrome with significant morbidity and mortality. Sildenafil is widely used off-label in pediatric patients with pulmonary arterial hypertension. In this study, bile salt-stabilized nanovesicles (bilosomes) were screened for their efficacy to enhance the transdermal delivery of the phosphodiesterase type 5 inhibitor, sildenafil citrate, in an attempt to augment its therapeutic efficacy in pediatric pulmonary hypertension. A response surface methodology was implemented for fabricating and optimizing a bilosomal formulation of sildenafil (SDF-BS). The optimized SDF-BS formulation was characterized in terms of its entrapment efficiency (EE), zeta potential, vesicle size, and in vitro release profile. The optimized formula was then loaded onto hydroxypropyl methyl cellulose (HPMC) hydrogel and assessed for skin permeation, in vivo pharmacokinetics, and pharmacodynamic studies. The optimized SDF-BS showed the following characteristic features; EE of 88.7 ± 1.1%, vesicle size of 185.0 + 9.2 nm, zeta potential of -20.4 ± 1.1 mV, and efficiently sustained SDF release for 12 h. Skin permeation study revealed a remarkable improvement in SDF penetration from bilosomal gel compared to plain SDF gel. In addition, pharmacokinetic results revealed that encapsulating SDF within bilosomal vesicles significantly enhanced its systemic bioavailability (∼3 folds), compared to SDF oral suspension. In addition, pharmacodynamic investigation revealed that, compared to plain SDF gel or oral drug suspension, SDF-BS gel applied topically triggered a significant elevation (p < 0.05) in cGMP serum levels, underscoring the superior therapeutic efficacy of SDF-BS gel. Conclusively, bilosomes can be viewed as a promising nanocarrier for transdermal delivery of SDF that would grant higher therapeutic efficiency while alleviating the limitations encountered with SDF oral administration.
Collapse
Affiliation(s)
- Bjad K. Almutairy
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
| | - El-Sayed Khafagy
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department
of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Mohammed F. Aldawsari
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
| | - Abdullah Alshetaili
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
| | - Hadil Faris Alotaibi
- Department
of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint AbdulRahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Amr Selim Abu Lila
- Department
of Pharmaceutics, College of Pharmacy, University
of Hail, Hail 81442, Saudi Arabia
- Medical
and Diagnostic Research Center, University
of Hail, Hail 81442, Saudi Arabia
- Department
of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
2
|
Mao Y, Yuan W, Gai J, Zhang Y, Wu S, Xu EY, Wang L, Zhang X, Guan J, Mao S. Enhanced brain distribution of Ginsenoside F1 via intranasal administration in combination with absorption enhancers. Int J Pharm 2024; 654:123930. [PMID: 38387820 DOI: 10.1016/j.ijpharm.2024.123930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Ginsenoside F1 (GF1) is a potential drug candidate for the treatment of Alzheimer's disease. Nevertheless, its low oral bioavailability and poor solubility limit clinical application. By utilizing either a direct or indirect approach, intranasal administration is a non-invasive drug delivery method that can deliver drugs to the brain rapidly. But large molecule drug delivered to the brain through intranasal administration may be insufficient to reach required concentration for therapeutic effect. In this study, using GF1 as a model drug, the feasibility of intranasal administration in combination with absorption enhancers to increase brain distribution of GF1 was explored. First of all, the appropriate absorption enhancers were screened by in situ nasal perfusion study. GF1-HP-β-CD inclusion complex was prepared and characterized. Thereafter, in vivo absorption of GF1 after intranasal or intravenous administration of its inclusion complex with/without absorption enhancers was investigated, and safety of the formulations was evaluated. The results showed that 2% Solutol HS 15 was a superior absorption enhancer. HP-β-CD inclusion complex improved GF1 solubility by 150 fold. Following intranasal delivery, the absolute bioavailability of inclusion complex was 46%, with drug brain targeting index (DTI) 247% and nose-to-brain direct transport percentage (DTP) 58%. Upon further addition of 2% Solutol HS 15, the absolute bioavailability was increased to 75%, with DTI 315% and DTP 66%. Both nasal cilia movement and biochemical substances (total protein and lactate dehydrogenase) leaching studies demonstrated 2% Solutol HS 15 was safe to the nasal mucosa. In conclusion, intranasal administration combining with safe absorption enhancers is an effective strategy to enhance drug distribution in the brain, showing promise for treating disorders related to the central nervous system.
Collapse
Affiliation(s)
- Ying Mao
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Weihua Yuan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiayi Gai
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yixuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | | | - En-Yu Xu
- Department of Forensic Toxicological Analysis, School of Forensic Medicine, China Medical University, Shenyang 110122, China.
| | - Luyao Wang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jian Guan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
3
|
Chen Y, Zhang C, Huang Y, Ma Y, Song Q, Chen H, Jiang G, Gao X. Intranasal drug delivery: The interaction between nanoparticles and the nose-to-brain pathway. Adv Drug Deliv Rev 2024; 207:115196. [PMID: 38336090 DOI: 10.1016/j.addr.2024.115196] [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: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
Intranasal delivery provides a direct and non-invasive method for drugs to reach the central nervous system. Nanoparticles play a crucial role as carriers in augmenting the efficacy of brain delivery. However, the interaction between nanoparticles and the nose-to-brain pathway and how the various biopharmaceutical factors affect brain delivery efficacy remains unclear. In this review, we comprehensively summarized the anatomical and physiological characteristics of the nose-to-brain pathway and the obstacles that hinder brain delivery. We then outlined the interaction between nanoparticles and this pathway and reviewed the biomedical applications of various nanoparticulate drug delivery systems for nose-to-brain drug delivery. This review aims at inspiring innovative approaches for enhancing the effectiveness of nose-to-brain drug delivery in the treatment of different brain disorders.
Collapse
Affiliation(s)
- Yaoxing Chen
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Chenyun Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yuxiao Ma
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201210, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
4
|
Luo Z, Klein Cerrejon D, Römer S, Zoratto N, Leroux JC. Boosting systemic absorption of peptides with a bioinspired buccal-stretching patch. Sci Transl Med 2023; 15:eabq1887. [PMID: 37756378 DOI: 10.1126/scitranslmed.abq1887] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/29/2022] [Accepted: 06/28/2023] [Indexed: 09/29/2023]
Abstract
Biopharmaceuticals, including proteins and peptides, have revolutionized the treatment of a wide range of diseases, from diabetes and cardiovascular disorders to virus infections and cancer. Despite their efficacy, most of these macromolecular drugs require parenteral administration because of their high molecular weight and relative instability. Over the past 40 years, only a few oral peptide drugs have entered clinical trials, even when formulated with substantial amounts of permeation enhancers. To overcome the epithelial barrier, devices that inject drugs directly into the gastrointestinal mucosa have been proposed recently. However, the robustness and safety of those complex systems are yet to be assessed. In this study, we introduced an innovative technology to boost drug absorption by synergistically combining noninvasive stretching of the buccal mucosa with permeation enhancers. Inspired by the unique structural features of octopus suckers, a self-applicable suction patch was engineered, enabling strong adhesion to and effective mechanical deformation of the mucosal tissue. In dogs, this suction patch achieved bioavailability up to two orders of magnitude higher than those of the commercial tablet formulation of desmopressin, a peptide drug known for its poor oral absorption. Moreover, systemic exposure comparable to that of the approved oral semaglutide tablet was achieved without further optimization. Last, a first-in-human study involving 40 healthy participants confirmed the dosage form's acceptability, thereby supporting the clinical translatability of this simple yet effective platform technology.
Collapse
Affiliation(s)
- Zhi Luo
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, P.R. China
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - David Klein Cerrejon
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Simon Römer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Nicole Zoratto
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
5
|
Silva S, Bicker J, Falcão A, Fortuna A. Air-liquid interface (ALI) impact on different respiratory cell cultures. Eur J Pharm Biopharm 2023; 184:62-82. [PMID: 36696943 DOI: 10.1016/j.ejpb.2023.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/24/2022] [Accepted: 01/19/2023] [Indexed: 01/23/2023]
Abstract
The intranasal route has been receiving greater attention from the scientific community not only for systemic drug delivery but also for the treatment of pulmonary and neurological diseases. Along with it, drug transport and permeability studies across the nasal mucosa have exponentially increased. Nevertheless, the translation of data from in vitro cell lines to in vivo studies is not always reliable, due to the difficulty in generating an in vitro model that resembles respiratory human physiology. Among all currently available methodologies, the air-liquid interface (ALI) method is advantageous to promote cell differentiation and optimize the morphological and histological characteristics of airway epithelium cells. Cells grown under ALI conditions, in alternative to submerged conditions, appear to provide relevant input for inhalation and pulmonary toxicology and complement in vivo experiments. Different methodologies and a variety of materials have been used to induce ALI conditions in primary cells and numerous cell lines. Until this day, with only exploratory results, no consensus has been reached regarding the validation of the ALI method, hampering data comparison. The present review describes the most adequate cell models of airway epithelium and how these models are differently affected by ALI conditions. It includes the evaluation of cellular features before and after ALI, and the application of the method in primary cell cultures, commercial 3D primary cells, cell lines and stem-cell derived models. A variety of these models have been recently applied for pharmacological studies against severe acute respiratory syndrome-coronavirus(-2) SARS-CoV(-2), namely primary cultures with alveolar type II epithelium cells and organotypic 3D models. The herein compiled data suggest that ALI conditions must be optimized bearing in mind the type of cells (nasal, bronchial, alveolar), their origin and the objective of the study.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
6
|
Development and evaluation of novel krill oil-based clomiphene microemulsion as a therapeutic strategy for PCOS treatment. Drug Deliv Transl Res 2023:10.1007/s13346-023-01304-z. [PMID: 36821036 DOI: 10.1007/s13346-023-01304-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
Polycystic ovary syndrome (PCOS) is frequently diagnosed hormonal disorder with reproductive and metabolic complications. The most common symptoms include cyst in ovaries, anovulation, insulin resistance, and obesity. Clomiphene citrate, an ovulating agent, is the first-line drug used to treat PCOS. We hypothesized that clomiphene citrate, by stimulating ovarian function, with krill oil used as an oil phase to improve solubility, by addressing PCOS-associated symptoms might be effective in PCOS. Hence, our goal was to target hormonal imbalance along with PCOS-associated symptoms using a single formulation. The concentration of water (X1), oil (X2), and Smix (surfactant-cosurfactant mixture) (X3) were selected as independent variables, in a simplex lattice design, from microemulsion area derived from a pseuodoternary phase diagram while the globule size (Y1) was selected as a dependent parameter. The optimized microemulsion showed good sphericity having 41 nm globule size, 0.32 poly dispersibility index and + 31 mV zeta potential. The optimized microemulsion was further evaluated in-vivo using letrozole-induced PCOS rats. Formulation treated group reversed the effect of letrozole on body weight and estrus cycle in comparison to the disease control group (p < 0.001). The formulation was also effective in reducing insulin resistance, cholesterol and serum testosterone level (p < 0.001). The in vivo results were supported by histopathological studies where the formulation-treated group showed a marked decrease in the number of cystic follicles and a remarkable increase in the number of growing follicles at variable stages, similar to the normal control group. Thus, the results confirmed that novel krill oil-based clomiphene microemulsion may become a promising therapeutic choice for the treatment of PCOS.
Collapse
|
7
|
Preparation and Evaluation of Mucus-Penetrating Inhalable Microparticles of Tiotropium Bromide Containing Sodium Glycocholate. Pharmaceutics 2022; 14:pharmaceutics14071409. [PMID: 35890304 PMCID: PMC9321333 DOI: 10.3390/pharmaceutics14071409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 12/10/2022] Open
Abstract
This study aimed to prepare mucus-penetrating inhalable microparticles for dry powder inhalers and to evaluate their applicability in an asthma-induced rat model. Microparticles were prepared from water solutions containing tiotropium bromide, L-leucine, and sodium glycocholate (NaGc) as permeation enhancers using the spray drying method. Four formulations (SDL1, SDL2, SDL3, and SDL4) were used, depending on the various NaGc concentrations. Tiotropium microparticles were characterized by standard methods. Additionally, an asthma-induced rat model was used to confirm the effects of the formulations on lung function. Tiotropium microparticles with NaGc resulted in formulations with a more corrugated morphology and smaller particle size distribution than those without NaGc. SDL 1 had a rough surface with irregular morphology, and SDL 2, 3, and 4 had a corrugated morphology. All SDL formulations had an aerodynamic size of <3 µm. The microparticles with a corrugated morphology aerosolized better than SDL1 microparticles. The apparent permeability coefficient (Papp) values of SDL3 and SDL4 were significantly higher than those for raw tiotropium. In an in vivo study using an asthma-induced rat model, the specific airway resistance (Sraw), airway wall thickness, and mean alveolus size recovered to those of the negative control group in the SDL4 formulation.
Collapse
|
8
|
Shirsath K, Agrawal YO. Intranasal Nanoemulsions A Potential Strategy for Targeting The Neurodegenerative Disorder: Parkinson's. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-124720. [PMID: 35733314 DOI: 10.2174/1871527321666220622163403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/11/2022] [Accepted: 04/26/2022] [Indexed: 06/15/2023]
Abstract
Intranasal delivery has great potential to cross the blood-brain barrier and deliver the drug molecule into the central nervous system faster than traditional methods. The olfactory neuronal and trigeminal pathways both are involved in intranasal delivery. The nano-technology is an innovative strategy for the nose to brain delivery. The mucoadhesive nanoemulsion formulation is a modified technology that increases the duration of drug accumulation and provides prolonged delivery at a targeted site. The nanoemulsion formulation oil, surfactant, and co-surfactant components maintain lower surface tension and particle coalescence. The globule dimension and zeta potential are affected in brain targeting. The globule size of the innovative formulation should be < 200 nm for drug permeation because, in humans, the average axon magnitude ranges from around 100 to 700. Furthermore, modified technology of nanoemulsion like nanogel and nanoemulsion in-situ gel provide a great advantage to cure neurodegenerative disorders. Therefore, focusing on the innovative pharmaceutical approaches of nanoemulsion in intranasal drug delivery, the current review provides insight into the applications of nanoemulsion in neurodegenerative disorders like Parkinson's disease, which are due to the depletion of dopamine in substania nigra resulting in cardinal motor activity bradykinesia and tremors. The review also touches upon the pathways for intranasal delivery of nanoemulsion, the pathogenesis of Parkinson's disease, and the future direction of the research on intranasal nanoemulsion.
Collapse
Affiliation(s)
- Krushna Shirsath
- Department of Pharmaceutics R. C. Patel Institute of Pharmaceutical Education and Research India
| | - Yogeeta O Agrawal
- Department of Pharmaceutics R. C. Patel Institute of Pharmaceutical Education and Research India
| |
Collapse
|
9
|
Cavanagh R, Shubber S, Vllasaliu D, Stolnik S. Enhanced permeation by amphiphilic surfactant is spatially heterogenous at membrane and cell level. J Control Release 2022; 345:734-743. [PMID: 35367276 DOI: 10.1016/j.jconrel.2022.03.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 11/25/2022]
Abstract
In the context of increased interest in permeability enhancement technologies to achieve mucosal delivery of drugs and biologics, we report our study on effects of the amphiphilic surfactant at cell membrane and cell population levels. Our results show that modulation in membrane order and fluidity initially occurs on insertion of individual surfactant molecules into the outer leaflet of membrane lipid bilayer; a process occurring at concentrations below surfactant's critical micellar concentration. The surfactant insertion, and consequent increase in membrane fluidity, are observed to be spatially heterogenous, i.e. manifested as 'patches' of increased membrane fluidity. At the cell population level, spatially heterogeneous activity of surfactant is also manifested, with certain cells displaying high permeability amongst a 'background' population. We propose that this heterogeneity is further manifested in a broad profile of intracellular and nuclear exposure levels to a model drug (doxorubicin) observed in cell population. The study points to heterogeneous nature of surfactant effects at cell membrane and cells in population levels.
Collapse
Affiliation(s)
- Robert Cavanagh
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Saif Shubber
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Driton Vllasaliu
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Snjezana Stolnik
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| |
Collapse
|
10
|
Phongpradist R, Thongchai W, Thongkorn K, Lekawanvijit S, Chittasupho C. Surface Modification of Curcumin Microemulsions by Coupling of KLVFF Peptide: A Prototype for Targeted Bifunctional Microemulsions. Polymers (Basel) 2022; 14:443. [PMID: 35160433 PMCID: PMC8838555 DOI: 10.3390/polym14030443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 01/12/2023] Open
Abstract
Curcumin is one of the most promising natural therapeutics for use against Alzheimer's disease. The major limitations of curcumin are its low oral bioavailability and difficulty in permeating the blood-brain barrier. Therefore, designing a delivery system of curcumin to overcome its limitations must be employed. KLVFF, a peptide known as an amyloid blocker, was used in this study as a targeting moiety to develop a targeted drug delivery system. A prototype of transnasal KLVFF conjugated microemulsions containing curcumin (KLVFF-Cur-ME) for the nose-to-brain delivery was fabricated. The KLVFF-Cur-ME was developed by a titration method. A conjugation of KLVFF was performed through a carbodiimide reaction, and the conjugation efficiency was confirmed by FTIR and DSC technique. KLVFD-Cur-ME was characterized for the drug content, globule size, zeta potential, and pH. A transparent and homogeneous KLVFF-Cur-ME is achieved with a drug content of 80.25% and a globule size of 76.1 ± 2.5 nm. The pH of KLVFF-Cur-ME is 5.33 ± 0.02, indicating non-irritation to nasal tissues. KLVFD-Cur-ME does not show nasal ciliotoxicity. An ex vivo diffusion study revealed that KLVFF-Cur-ME partitions the porcine nasal mucosa through diffusion, following the Higuchi model. This investigation demonstrates the successful synthesis of a bifunctional KLVFF-Cur-ME as a novel prototype to deliver anti-Aβ aggregation via an intranasal administration.
Collapse
Affiliation(s)
- Rungsinee Phongpradist
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Center of Excellence for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wisanu Thongchai
- Chemistry Program, Faculty of Science and Technology, Pibulsongkram Rajabhat University, Phitsanuloke 65000, Thailand;
| | - Kriangkrai Thongkorn
- Department of Companion Animals and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
| | - Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| |
Collapse
|
11
|
Clementino AR, Pellegrini G, Banella S, Colombo G, Cantù L, Sonvico F, Del Favero E. Structure and Fate of Nanoparticles Designed for the Nasal Delivery of Poorly Soluble Drugs. Mol Pharm 2021; 18:3132-3146. [PMID: 34259534 PMCID: PMC8335725 DOI: 10.1021/acs.molpharmaceut.1c00366] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoparticles are promising mediators to enable nasal systemic and brain delivery of active compounds. However, the possibility of reaching therapeutically relevant levels of exogenous molecules in the body is strongly reliant on the ability of the nanoparticles to overcome biological barriers. In this work, three paradigmatic nanoformulations vehiculating the poorly soluble model drug simvastatin were addressed: (i) hybrid lecithin/chitosan nanoparticles (LCNs), (ii) polymeric poly-ε-caprolactone nanocapsules stabilized with the nonionic surfactant polysorbate 80 (PCL_P80), and (iii) polymeric poly-ε-caprolactone nanocapsules stabilized with a polysaccharide-based surfactant, i.e., sodium caproyl hyaluronate (PCL_SCH). The three nanosystems were investigated for their physicochemical and structural properties and for their impact on the biopharmaceutical aspects critical for nasal and nose-to-brain delivery: biocompatibility, drug release, mucoadhesion, and permeation across the nasal mucosa. All three nanoformulations were highly reproducible, with small particle size (∼200 nm), narrow size distribution (polydispersity index (PI) < 0.2), and high drug encapsulation efficiency (>97%). Nanoparticle composition, surface charge, and internal structure (multilayered, core-shell or raspberry-like, as assessed by small-angle neutron scattering, SANS) were demonstrated to have an impact on both the drug-release profile and, strikingly, its behavior at the biological interface. The interaction with the mucus layer and the kinetics and extent of transport of the drug across the excised animal nasal epithelium were modulated by nanoparticle structure and surface. In fact, all of the produced nanoparticles improved simvastatin transport across the epithelial barrier of the nasal cavity as compared to a traditional formulation. Interestingly, however, the permeation enhancement was achieved via two distinct pathways: (a) enhanced mucoadhesion for hybrid LCN accompanied by fast mucosal permeation of the model drug, or (b) mucopenetration and an improved uptake and potential transport of whole PCL_P80 and PCL_SCH nanocapsules with delayed boost of permeation across the nasal mucosa. The correlation between nanoparticle structure and its biopharmaceutical properties appears to be a pivotal point for the development of novel platforms suitable for systemic and brain delivery of pharmaceutical compounds via intranasal administration.
Collapse
Affiliation(s)
- Adryana Rocha Clementino
- National Council for Scientific and Technological Development-CNPq, Brazilian Government, Brasília DF, 70311-000, Brazil.,Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 20090 Parma, Italy
| | - Giulia Pellegrini
- Department of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Via Fratelli Cervi 93, Segrate, 20122 Milan, Italy
| | - Sabrina Banella
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Gaia Colombo
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Laura Cantù
- Department of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Via Fratelli Cervi 93, Segrate, 20122 Milan, Italy
| | - Fabio Sonvico
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 20090 Parma, Italy.,Biopharmanet-TEC, University of Parma, Parco Area delle Scienze 27/A, 20090 Parma, Italy
| | - Elena Del Favero
- Department of Medical Biotechnologies and Translational Medicine, LITA, University of Milan, Via Fratelli Cervi 93, Segrate, 20122 Milan, Italy
| |
Collapse
|
12
|
Wagner C, Kesisoglou F, Pepin XJH, Parrott N, Emami Riedmaier A. Use of Physiologically Based Pharmacokinetic Modeling for Predicting Drug-Food Interactions: Recommendations for Improving Predictive Performance of Low Confidence Food Effect Models. AAPS JOURNAL 2021; 23:85. [PMID: 34142242 DOI: 10.1208/s12248-021-00601-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/20/2021] [Indexed: 11/30/2022]
Abstract
Food can alter drug absorption and impact safety and efficacy. Besides conducting clinical studies, in vitro approaches such as biorelevant solubility and dissolution testing and in vivo dog studies are typical approaches to estimate a drug's food effect. The use of physiologically based pharmacokinetic models has gained importance and is nowadays a standard tool for food effect predictions at preclinical and clinical stages in the pharmaceutical industry. This manuscript is part of a broader publication from the IQ Consortium's food effect physiologically based pharmacokinetic model (PBPK) modeling working group and complements previous publications by focusing on cases where the food effect was predicted with low confidence. Pazopanib-HCl, trospium-Cl, and ziprasidone-HCl served as model compounds to provide insights into why several food effect predictions failed in the first instance. Furthermore, the manuscript depicts approaches whereby PBPK-based food effect predictions may be improved. These improvements should focus on the PBPK model functionality, especially better reflecting fasted- and fed-state gastric solubility, gastric re-acidification, and complex mechanisms related to gastric emptying of drugs. For improvement of in vitro methodologies, the focus should be on the development of more predictive solubility, supersaturation, and precipitation assays. With regards to the general PBPK modeling methodology, modelers should account for the full solubility profile when modeling ionizable compounds, including common ion effects, and apply a straightforward strategy to account for drug precipitation.
Collapse
Affiliation(s)
- Christian Wagner
- Pharmaceutical Technologies, Chemical and Pharmaceutical Development, Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany.
| | | | - Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
13
|
Miyazawa T, Itaya M, Burdeos GC, Nakagawa K, Miyazawa T. A Critical Review of the Use of Surfactant-Coated Nanoparticles in Nanomedicine and Food Nanotechnology. Int J Nanomedicine 2021; 16:3937-3999. [PMID: 34140768 PMCID: PMC8203100 DOI: 10.2147/ijn.s298606] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Surfactants, whose existence has been recognized as early as 2800 BC, have had a long history with the development of human civilization. With the rapid development of nanotechnology in the latter half of the 20th century, breakthroughs in nanomedicine and food nanotechnology using nanoparticles have been remarkable, and new applications have been developed. The technology of surfactant-coated nanoparticles, which provides new functions to nanoparticles for use in the fields of nanomedicine and food nanotechnology, is attracting a lot of attention in the fields of basic research and industry. This review systematically describes these "surfactant-coated nanoparticles" through various sections in order: 1) surfactants, 2) surfactant-coated nanoparticles, application of surfactant-coated nanoparticles to 3) nanomedicine, and 4) food nanotechnology. Furthermore, current progress and problems of the technology using surfactant-coated nanoparticles through recent research reports have been discussed.
Collapse
Affiliation(s)
- Taiki Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| | - Mayuko Itaya
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Gregor C Burdeos
- Institute for Animal Nutrition and Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Kiyotaka Nakagawa
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
14
|
Shah B. Microemulsion as a promising carrier for nose to brain delivery: journey since last decade. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00528-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Bahadur S, Pardhi DM, Rautio J, Rosenholm JM, Pathak K. Intranasal Nanoemulsions for Direct Nose-to-Brain Delivery of Actives for CNS Disorders. Pharmaceutics 2020; 12:E1230. [PMID: 33352959 PMCID: PMC7767046 DOI: 10.3390/pharmaceutics12121230] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
The treatment of various central nervous system (CNS) diseases has been challenging, despite the rapid development of several novel treatment approaches. The blood-brain barrier (BBB) is one of the major issues in the treatment of CNS diseases, having major role in the protection of the brain but simultaneously constituting the main limiting hurdle for drugs targeting the brain. Nasal drug delivery has gained significant interest for brain targeting over the past decades, wherein the drug is directly delivered to the brain by the trigeminal and olfactory pathway. Various novel and promising formulation approaches have been explored for drug targeting to the brain by nasal administration. Nanoemulsions have the potential to avoid problems, including low solubility, poor bioavailability, slow onset of action, and enzymatic degradation. The present review highlights research scenarios of nanoemulsions for nose-to-brain delivery for the management of CNS ailments classified on the basis of brain disorders and further identifies the areas that remain unexplored. The significance of the total dose delivered to the target region, biodistribution studies, and long-term toxicity studies have been identified as the key areas of future research.
Collapse
Affiliation(s)
- Shiv Bahadur
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India;
| | - Dinesh M. Pardhi
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (D.M.P.); (J.R.)
| | - Jarkko Rautio
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (D.M.P.); (J.R.)
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| |
Collapse
|
16
|
Paprikar A, Soni A, Kaushal N, Lin S. Sublingual insulin administration: application of hydroxypropyl beta-cyclodextrin and poloxamer188 as permeation enhancers. Pharm Dev Technol 2020; 26:233-242. [PMID: 33258391 DOI: 10.1080/10837450.2020.1858319] [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: 10/22/2022]
Abstract
The objective of this investigation is to investigate the feasibility of sublingual insulin administration. Insulin solutions formulated with permeation enhancers (HPβCD/poloxamer 188) and their in-vitro and in-vivo performances were evaluated. Thereafter, insulin fast-dissolving film was further developed to have similar properties, upon dissolving the film, of the optimized insulin solution. In-vitro performance was evaluated via effect of HPβCD and/or poloxamer 188 concentration across cellulose acetate membrane and porcine esophagus. In-vivo performance was evaluated via pharmacodynamic and pharmacokinetic profiles of insulin solution administered. Cumulative amounts of insulin permeated at 60 min formulated with HPβCD (5%), poloxamer 188 (0.5%), and their combination were 1.31, 3.23, and 4.99 IU/cm2, respectively, indicating an additive effect of combination of HPβCD and poloxamer 188. Insulin-induced hypoglycemic effect was observed for insulin solutions with combination of HPβCD and poloxamer 188 after sublingual administration to Sprague-Dawley rats. Microscopic evaluation of porcine oesophageal tissue indicates that HPβCD and poloxamer 188 are safe. Furthermore, the cumulative amount permeated across cellulose acetate membrane at 30 min was 1.13 and 1.00 IU/cm2 for insulin solution and fast-dissolving film, respectively, demonstrating to be similar. In conclusion, the use of HPβCD/poloxamer 188 is feasible for the development of sublingual insulin solutions/films.
Collapse
Affiliation(s)
- Anuja Paprikar
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Ankit Soni
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Neeraj Kaushal
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Senshang Lin
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| |
Collapse
|
17
|
Abou Assi R, M. Abdulbaqi I, Seok Ming T, Siok Yee C, A. Wahab H, Asif SM, Darwis Y. Liquid and Solid Self-Emulsifying Drug Delivery Systems (SEDDs) as Carriers for the Oral Delivery of Azithromycin: Optimization, In Vitro Characterization and Stability Assessment. Pharmaceutics 2020; 12:E1052. [PMID: 33158058 PMCID: PMC7693798 DOI: 10.3390/pharmaceutics12111052] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Azithromycin (AZM) is a macrolide antibiotic used for the treatment of various bacterial infections. The drug is known to have low oral bioavailability (37%) which may be attributed to its relatively high molecular weight, low solubility, dissolution rate, and incomplete intestinal absorption. To overcome these drawbacks, liquid (L) and solid (S) self-emulsifying drug delivery systems (SEDDs) of AZM were developed and optimized. Eight different pseudo-ternary diagrams were constructed based on the drug solubility and the emulsification studies in various SEDDs excipients at different surfactant to co-surfactant (Smix) ratios. Droplet size (DS) < 150 nm, dispersity (Đ) ≤ 0.7, and transmittance (T)% > 85 in three diluents of distilled water (DW), 0.1 mM HCl, and simulated intestinal fluids (SIF) were considered as the selection criteria. The final formulations of L-SEDDs (L-F1(H)), and S-SEDDs (S-F1(H)) were able to meet the selection requirements. Both formulations were proven to be cytocompatible and able to open up the cellular epithelial tight junctions (TJ). The drug dissolution studies showed that after 5 min > 90% and 52.22% of the AZM was released from liquid and solid SEDDs formulations in DW, respectively, compared to 11.27% of the pure AZM, suggesting the developed SEDDs may enhance the oral delivery of the drug. The formulations were stable at refrigerator storage conditions.
Collapse
Affiliation(s)
- Reem Abou Assi
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
- The Discipline of Pharmaceutical Technology, College of Pharmacy, Al-Kitab University, Altun kupri, Kirkuk 36001, Iraq
| | - Ibrahim M. Abdulbaqi
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
- The Discipline of Pharmaceutical Technology, College of Pharmacy, Al-Kitab University, Altun kupri, Kirkuk 36001, Iraq
| | - Toh Seok Ming
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
| | - Chan Siok Yee
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
| | - Habibah A. Wahab
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
| | - Shaik Mohammed Asif
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
- Pharma Research, Wockhardt Research Center, Aurangabad 431002, India
| | - Yusrida Darwis
- The Discipline of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (R.A.A.); (I.M.A.); (T.S.M.); (S.M.A.)
| |
Collapse
|
18
|
Intranasal delivery of tetrabenazine nanoemulsion via olfactory region for better treatment of hyperkinetic movement associated with Huntington’s disease: Pharmacokinetic and brain delivery study. Chem Phys Lipids 2020; 230:104917. [DOI: 10.1016/j.chemphyslip.2020.104917] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/20/2020] [Accepted: 05/03/2020] [Indexed: 12/31/2022]
|
19
|
Novel and revisited approaches in nanoparticle systems for buccal drug delivery. J Control Release 2020; 320:125-141. [DOI: 10.1016/j.jconrel.2020.01.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/02/2020] [Accepted: 01/04/2020] [Indexed: 12/15/2022]
|
20
|
Komati S, Swain S, Rao MEB, Jena BR, Dasi V. Mucoadhesive Multiparticulate Drug Delivery Systems: An Extensive Review of Patents. Adv Pharm Bull 2019; 9:521-538. [PMID: 31857957 PMCID: PMC6912179 DOI: 10.15171/apb.2019.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 11/09/2022] Open
Abstract
Innovations in pharmaceutical research are striving for designing newer drug therapies to eradicate deadly diseases. Strategies for such inventions always flourish with keys and objectives of minimal adverse effects and effective treatment. Recent trends in pharmaceutical technology specify that mucoadhesive drug delivery system is particularly appropriate than oral control release, for getting local systematic delivery of drugs in GIT for an extended interval of time at a predetermined rate. However, it is somehow expensive and unpleasant sensation for some patients, but still it is needful for getting short enzymatic activity, simple administration without pain and evasion of fast pass metabolism. Usually the vehicles employed in drug delivery of mucoadhesive system have a significant impact that draws further attention to potential benefits like improved bioavailability of therapeutic agents, extensive drug residence time at the site of administration and a comparatively faster drug uptake into the systemic circulation. The drug release from mucoadhesive multiparticulates is contingent on several types of factors comprising carrier need to produce the multiparticles and quantity of medication drug contained in them. Mucoadhesion is characterized by selected theories and mechanisms. Various strategies emergent in mucoadhesive multiparticulate drug delivery system (MMDDS) by in-vitro as well as ex-vivo description and characterization are also critically discussed. Apart from these, the primary focus during this review is to highlight current patents, clinical status, and regulatory policy for enhancement of mucoadhesive multi-particulate drug delivery system in the present scenario.
Collapse
Affiliation(s)
- Someshwar Komati
- Department of Pharmaceutics, University College of Pharmaceutical Sciences, Palamuru University, Mahaboobnagar, Telangana-509001, India
| | - Suryakanta Swain
- Southern Institute of Medical Sciences, College of Pharmacy, Mangaldas Nagar, Vijyawada Road, Guntur-522 001, Andhra Pradesh, India
| | - Muddana Eswara Bhanoji Rao
- Department of Pharmaceutics, Roland Institute of Pharmaceutical Sciences, Khodasinghi, Berhampur-760 010, Ganjam, Odisha, India
| | - Bikash Ranjan Jena
- Southern Institute of Medical Sciences, College of Pharmacy, Mangaldas Nagar, Vijyawada Road, Guntur-522 001, Andhra Pradesh, India
| | - Vishali Dasi
- Department of Pharmaceutics, University College of Pharmaceutical Sciences, Palamuru University, Mahaboobnagar, Telangana-509001, India
| |
Collapse
|
21
|
Chatterjee B, Gorain B, Mohananaidu K, Sengupta P, Mandal UK, Choudhury H. Targeted drug delivery to the brain via intranasal nanoemulsion: Available proof of concept and existing challenges. Int J Pharm 2019; 565:258-268. [PMID: 31095983 DOI: 10.1016/j.ijpharm.2019.05.032] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/10/2019] [Accepted: 05/11/2019] [Indexed: 12/15/2022]
Abstract
Intranasal delivery has shown to circumvent blood-brain-barrier (BBB) and deliver the drugs into the CNS at a higher rate and extent than other conventional routes. The mechanism of drug transport from nose-to-brain is not fully understood yet, but several neuronal pathways are considered to be involved. Intranasal nanoemulsion for brain targeting is investigated extensively. Higher brain distribution of drug after administering intranasal nanoemulsion was established by many researchers. Issues with nasomucosal clearance are solved by formulating modified nanoemulsion; for instance, mucoadhesive nanoemulsion or in situ nanoemulgel. However, no intranasal nanoemulsion for brain targeted drug delivery has been able to cross the way from 'benches to bed-side' of patients. Possibilities of toxicity by repeated administration, irregular nasal absorption during the diseased condition, use of a high amount of surfactants are few of the persisting challenges that need to overcome in coming days. Understanding the ways how current developments has solved some challenges is necessary. At the same time, the future direction of the research on intranasal nanoemulsion should be figured out based on existing challenges. This review is focused on the current developments of intranasal nanoemulsion with special emphasis on the existing challenges that would help to set future research direction.
Collapse
Affiliation(s)
- Bappaditya Chatterjee
- Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Malaysia.
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor 47500, Malaysia.
| | - Keithanchali Mohananaidu
- Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Malaysia
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 382355, India.
| | - Uttam Kumar Mandal
- Department of Pharmaceutical Science & Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, India.
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
22
|
Antioxidant and Photoprotective Activity of Apigenin and its Potassium Salt Derivative in Human Keratinocytes and Absorption in Caco-2 Cell Monolayers. Int J Mol Sci 2019; 20:ijms20092148. [PMID: 31052292 PMCID: PMC6539602 DOI: 10.3390/ijms20092148] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 02/01/2023] Open
Abstract
Ultraviolet (UV) radiation, especially types A (UVA) and B (UVB), is one of the main causes of skin disorders, including photoaging and skin cancer. Ultraviolent radiation causes oxidative stress, inflammation, p53 induction, DNA damage, mutagenesis, and oxidation of various molecules such as lipids and proteins. In recent decades, the use of polyphenols as molecules with an antioxidant and anti-inflammatory capacity has increased. However, some of these compounds are poorly soluble, and information regarding their absorption and bioavailability is scarce. The main objective of this study was to compare the intestinal absorption and biological activity of apigenin and its more soluble potassium salt (apigenin-K) in terms of antioxidant and photoprotective capacity. Photoprotective effects against UVA and UVB radiation were studied in human keratinocytes, and antioxidant capacity was determined by different methods, including trolox equivalent antioxidant capacity (TEAC), ferric reducing antioxidant power (FRAP) and oxygen radical absorbance capacity (ORAC) assays. Finally, the intestinal absorption of both apigenins was determined using an in vitro Caco-2 cell model. Apigenin showed a slightly higher antioxidant capacity in antioxidant activity assays when compared with apigenin-K. However, no significant differences were obtained for their photoprotective capacities against UVA or UVB. Results indicated that both apigenins protected cell viability in approximately 50% at 5 J/m2 of UVA and 90% at 500 J/m2 of UVB radiation. Regarding intestinal absorption, both apigenins showed similar apparent permeabilities (Papp), 1.81 × 10-5 cm/s and 1.78 × 10-5 cm/s, respectively. Taken together, these results suggest that both apigenins may be interesting candidates for the development of oral (nutraceutical) and topical photoprotective ingredients against UVA and UVB-induced skin damage, but the increased water solubility of apigenin-K makes it the best candidate for further development.
Collapse
|
23
|
Ullio-Gamboa G, Udobi KC, Dezard S, Perna MK, Miles KN, Costa N, Taran F, Pruvost A, Benoit JP, Skelton MR, Lonlay PD, Mabondzo A. Dodecyl creatine ester-loaded nanoemulsion as a promising therapy for creatine transporter deficiency. Nanomedicine (Lond) 2019; 14:1579-1593. [PMID: 31038003 DOI: 10.2217/nnm-2019-0059] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Creatine transporter (CrT) deficiency is an X-linked intellectual disability caused by mutations of CrT. Aim: This work focus on the preclinical development of a new therapeutic approach based on a microemulsion (ME) as drug delivery system for dodecyl creatine ester (DCE). Materials & methods: DCE-ME was prepared by titration method. Novel object recognition (NOR) tests were performed before and after DCE-ME treatment on Slc6a8-/y mice. Results: Intranasal administration with DCE-ME improved NOR performance in Slc6a8-/y mice. Slc6a8-/y mice treated with DCE-ME had increased striatal ATP levels mainly in the striatum compared with vehicle-treated Slc6a8-/y mice which was associated with increased expression of synaptic markers. Conclusion: These results highlight the potential value of DCE-ME as promising therapy for creatine transporter deficiency.
Collapse
Affiliation(s)
- Gabriela Ullio-Gamboa
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Kenea C Udobi
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Sophie Dezard
- Service de Chimie Bio Organique et de Marquage CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Marla K Perna
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Keila N Miles
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Narciso Costa
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Frédéric Taran
- Service de Chimie Bio Organique et de Marquage CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Alain Pruvost
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Jean-Pierre Benoit
- LUNAM Université-Micro et Nanomédecines Biomimétiques, F-49933 Angers, France, INSERM U1066, IBS-CHU, 4 rue Larrey, F-49933 Angers Cedex 9, France
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Pascale de Lonlay
- Centre de Référence des Maladies Héréditaires du Métabolisme de l'Enfant et de l'Adulte, Hôpital Necker-Enfants Malades, APHP, Université Paris Descartes, Imagine, INEM, Filière G2M, metabERN, Paris, France
| | - Aloïse Mabondzo
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| |
Collapse
|
24
|
Pérez-Sánchez A, Cuyàs E, Ruiz-Torres V, Agulló-Chazarra L, Verdura S, González-Álvarez I, Bermejo M, Joven J, Micol V, Bosch-Barrera J, Menendez JA. Intestinal Permeability Study of Clinically Relevant Formulations of Silibinin in Caco-2 Cell Monolayers. Int J Mol Sci 2019; 20:E1606. [PMID: 30935093 PMCID: PMC6480586 DOI: 10.3390/ijms20071606] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
An ever-growing number of preclinical studies have investigated the tumoricidal activity of the milk thistle flavonolignan silibinin. The clinical value of silibinin as a bona fide anti-cancer therapy, however, remains uncertain with respect to its bioavailability and blood⁻brain barrier (BBB) permeability. To shed some light on the absorption and bioavailability of silibinin, we utilized the Caco-2 cell monolayer model of human intestinal absorption to evaluate the permeation properties of three different formulations of silibinin: silibinin-meglumine, a water-soluble form of silibinin complexed with the amino-sugar meglumine; silibinin-phosphatidylcholine, the phytolipid delivery system Siliphos; and Eurosil85/Euromed, a milk thistle extract that is the active component of the nutraceutical Legasil with enhanced bioavailability. Our approach predicted differential mechanisms of transport and blood⁻brain barrier permeabilities between the silibinin formulations tested. Our assessment might provide valuable information about an idoneous silibinin formulation capable of reaching target cancer tissues and accounting for the observed clinical effects of silibinin, including a recently reported meaningful central nervous system activity against brain metastases.
Collapse
Affiliation(s)
- Almudena Pérez-Sánchez
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Verónica Ruiz-Torres
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Luz Agulló-Chazarra
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Isabel González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Marival Bermejo
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Vicente Micol
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), 07122 Palma de Mallorca, Spain.
| | - Joaquim Bosch-Barrera
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Department of Medical Sciences, Medical School University of Girona, 17003 Girona, Spain.
- Medical Oncology, Catalan Institute of Oncology (ICO), Dr. Josep Trueta University Hospital, 17007 Girona, Spain.
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| |
Collapse
|
25
|
Breakdown of the Gut Barrier in Patients with Multiple Organ Dysfunction Syndrome is Attenuated by Continuous Blood Purification: Effects on Tight Junction Structural Proteins. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300102] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Breakdown of the gut barrier increases intestinal permeability and allows movement of intraluminal contents across the mucosa, which can lead to distant organ injury and multiple organ dysfunction syndrome (MODS). Intestinal permeability is associated with alterations in cellular tight junctions involving the structural proteins occludin and zonula occludens-1 (ZO-1). The aim of this study was to investigate the effect of continuous blood purification (CBP) on gut barrier function in patients with MODS. Method Serum diamine oxidase (DAO) and endotoxin, epithelial monolayer permeability, and transepithelial electrical resistance (TER) were used as markers for the assessment of gut barrier function in 22 patients with MODS who underwent continuous venovenous hemofiltration (CVVH) for 24 hours. Blood samples were taken from patients at 0, 6, 12, and 24 hours during CVVH therapy. Serum DAO and endotoxin were determined by spectrophotography. Permeability and TER were assessed using Caco-2 cell monolayers. Occludin and ZO-1 protein levels were analyzed by immunoblotting and immunofluorescence staining. And inducible nitric oxide synthase (iNOS) mRNA levels and nitric oxide (NO) production were determined by real-time PCR and spectrophotography, respectively. Result Gut barrier dysfunction was evident in patients with MODS compared with normal controls. Serum DAO, endotoxin levels, and epithelial permeability were elevated, while TER was decreased in patients with MODS, and this change was more pronounced in nonsurvivors. Breakdown and reorganization of occludin and ZO-1 away from tight junctions was found in all MODS patients. After CBP treatment, APACHE II and MODS scores improved significantly. Serum DAO and endotoxin levels and epithelial permeability also diminished, while TER increased in all patients; CBP significantly attenuated breakdown and reorganization of tight junction proteins, and also attenuated the inflammation-induced increase in iNOS mRNA expression and NO production. Conclusion Breakdown of gut barrier function is present in patients with MODS and may be correlated with poor outcomes in the disease. CBP can not only improve the general conditions, as measured by the APACHE II score, but also improve gut barrier dysfunction by attenuating the breakdown and reorganization of occludin and ZO-1. This beneficial effect of CBP on gut barrier dysfunction is associated with down-regulation of iNOS.
Collapse
|
26
|
Zhang J, Yuan C, Hua G, Tong R, Luo X, Ying Z. Early Gut Barrier Dysfunction in Patients with Severe Acute Pancreatitis: Attenuated by Continuous Blood Purification Treatment. Int J Artif Organs 2018. [DOI: 10.1177/039139881003301003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objectives The aim of this study was to investigate the effect of continuous blood purification (CBP) on early gut mucosal dysfunction in patients with severe acute pancreatitis (SAP). Methods Patients with SAP were randomized to receive 24 hours of continuous veno-venous hemofiltration (CVVH; n=33) or no CVVH (n=30). Blood samples were taken from the patients at 0, 6, 12, and 24 hours during CVVH therapy. Serum diamine oxidase (DAO) and endotoxin, epithelial permeability, transepithelial electrical resistance (TER) and F-actin rearrangement of the epithelial monolayer were used as the markers for the assessment of gut barrier function and the effect of CBP therapy in patients with SAP. Results Patients with SAP had increased levels of serum DAO, endotoxin, and epithelial permeability when compared with normal controls, and the increase was more pronounced in patients with organ dysfunction (p<0.01). F-actin rearrangement, loose cell-cell junction, and iNOS mRNA upregulation were found in all patients. After CBP treatment, Acute Physiology and Chronic Health Evaluation II score and SOFA score improved significantly; levels of serum DAO, endotoxin, and epithelial permeability decreased (p<0.05). CBP also significantly attenuated reorganization of actin and downregulated iNOS mRNA expression and NO production (p<0.05). Conclusions CBP can not only improve the general conditions but also effectively improve gut barrier dysfunction. The beneficial effect of CBP on gut barrier dysfunction is associated with the improvement of cytoskeletal instability, by downregulating iNOS through the removal of excess proinflammatory factors.
Collapse
Affiliation(s)
- JianBin Zhang
- Departments of Nephrology Diseases, The Third Affiliated Hospital of Nanchang University, Nanchang, JiangXi - China
| | - Chen Yuan
- Departments of Nephrology Diseases, The Third Affiliated Hospital of Nanchang University, Nanchang, JiangXi - China
| | - Gan Hua
- Departments of Nephrology Diseases, First Affiliated Hospital of Chongqing University of Medical Sciences, Chongqing - China
| | - RuYan Tong
- Departments of Nephrology Diseases, The Third Affiliated Hospital of Nanchang University, Nanchang, JiangXi - China
| | - XiangFeng Luo
- Departments of Nephrology Diseases, The Third Affiliated Hospital of Nanchang University, Nanchang, JiangXi - China
| | - Zhou Ying
- Departments of Nephrology Diseases, The Third Affiliated Hospital of Nanchang University, Nanchang, JiangXi - China
| |
Collapse
|
27
|
Enhanced Intestinal Permeability of Bufalin by a Novel Bufalin-Peptide-Dendrimer Inclusion through Caco-2 Cell Monolayer. Molecules 2017; 22:molecules22122088. [PMID: 29186041 PMCID: PMC6149814 DOI: 10.3390/molecules22122088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 11/17/2017] [Accepted: 11/23/2017] [Indexed: 01/06/2023] Open
Abstract
Bufalin (BFL) has excellent physiological activities such as defending tumors, improving cardiac function, and so on. However, due to its poor water-solubility and bioavailability, the clinical application of BFL remains limited. In order to improve bioavailability of BFL, in our previous research, a novel peptide-dendrimer (PD) was synthesized and applied to encapsulate BFL. In the present study, we investigate the absorption property and mechanism of BFL in free form and BFL-peptide-dendrimer inclusion (BPDI) delivery system by using the Caco-2 cell monolayer model in vitro. The apparent permeability coefficient (Papp) values of BFL in free or BPDI form were over 1.0 × 10−6 cm/s. Meanwhile, their almost equal bi-directional transport and linear transport percentage with time and concentration course indicated that BFL in both forms was absorbed mainly through passive diffusion. The most important result is that the Papp values of BFL increased about three-fold more BPDI than those of its free form, which indicated the intestinal permeability of BFL could be improved while BFL was encapsulated in BPDI form. Therefore, PD encapsulation may be a potential delivery system to increase the bioavailability of BFL.
Collapse
|
28
|
Pérez-Sánchez A, Borrás-Linares I, Barrajón-Catalán E, Arráez-Román D, González-Álvarez I, Ibáñez E, Segura-Carretero A, Bermejo M, Micol V. Evaluation of the intestinal permeability of rosemary (Rosmarinus officinalis L.) extract polyphenols and terpenoids in Caco-2 cell monolayers. PLoS One 2017; 12:e0172063. [PMID: 28234919 PMCID: PMC5325326 DOI: 10.1371/journal.pone.0172063] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/30/2017] [Indexed: 11/18/2022] Open
Abstract
Rosemary (Rosmarinus officinalis) is grown throughout the world and is widely used as a medicinal herb and to season and preserve food. Rosemary polyphenols and terpenoids have attracted great interest due to their potential health benefits. However, complete information regarding their absorption and bioavailability in Caco-2 cell model is scarce. The permeation properties of the bioactive compounds (flavonoids, diterpenes, triterpenes and phenylpropanoids) of a rosemary extract (RE), obtained by supercritical fluid extraction, was studied in Caco-2 cell monolayer model, both in a free form or liposomed. Compounds were identified and quantitated by liquid chromatography coupled to quadrupole time-of-flight with electrospray ionization mass spectrometry analysis (HPLC-ESI-QTOF-MS), and the apparent permeability values (Papp) were determined, for the first time in the extract, for 24 compounds in both directions across cell monolayer. For some compounds, such as triterpenoids and some flavonoids, Papp values found were reported for the first time in Caco-2 cells.Our results indicate that most compounds are scarcely absorbed, and passive diffusion is suggested to be the primary mechanism of absorption. The use of liposomes to vehiculize the extract resulted in reduced permeability for most compounds. Finally, the biopharmaceutical classification (BCS) of all the compounds was achieved according to their permeability and solubility data for bioequivalence purposes. BCS study reveal that most of the RE compounds could be classified as classes III and IV (low permeability); therefore, RE itself should also be classified into this category.
Collapse
Affiliation(s)
- Almudena Pérez-Sánchez
- Institute of Molecular and Cell Biology, Miguel Hernández University, Avda. Universidad s/n, Elche, Spain
| | - Isabel Borrás-Linares
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avda Fuentenueva s/n, Granada, Spain
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Avda. del Conocimiento n° 37, Armilla, Spain
| | - Enrique Barrajón-Catalán
- Institute of Molecular and Cell Biology, Miguel Hernández University, Avda. Universidad s/n, Elche, Spain
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain
- INVTROTECNIA S.L., Santiago Grisolía 2, Tres Cantos, Madrid, Spain
| | - David Arráez-Román
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avda Fuentenueva s/n, Granada, Spain
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Avda. del Conocimiento n° 37, Armilla, Spain
| | - Isabel González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain
| | - Elena Ibáñez
- Laboratory of Foodomics, Institute of Food Science Research-CIAL (CSIC-UAM), Nicolás Cabrera 9, Campus Cantoblanco, Madrid, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avda Fuentenueva s/n, Granada, Spain
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Avda. del Conocimiento n° 37, Armilla, Spain
| | - Marival Bermejo
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain
| | - Vicente Micol
- Institute of Molecular and Cell Biology, Miguel Hernández University, Avda. Universidad s/n, Elche, Spain
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), Spain
- * E-mail:
| |
Collapse
|
29
|
Investigation of betahistine dihydrochloride biocompatibility and nasal permeability in vitro. J Appl Biomed 2016. [DOI: 10.1016/j.jab.2016.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
30
|
Enhanced oral bioavailability of naringenin administered in a mixed micelle formulation with Pluronic F127 and Tween 80 in rats. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2015. [DOI: 10.1007/s40005-015-0216-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
31
|
Moghimipour E, Ameri A, Handali S. Absorption-Enhancing Effects of Bile Salts. Molecules 2015; 20:14451-73. [PMID: 26266402 PMCID: PMC6332414 DOI: 10.3390/molecules200814451] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/28/2015] [Accepted: 07/31/2015] [Indexed: 12/16/2022] Open
Abstract
Bile salts are ionic amphiphilic compounds with a steroid skeleton. Among the most important physiological properties of bile salts are lipid transport by solubilization and transport of some drugs through hydrophobic barriers. Bile salts have been extensively studied to enhance transepithelial permeability for different marker molecules and drugs. They readily agglomerate at concentrations above their critical micelle concentration (CMC). The mechanism of absorption enhancement by bile salts appears to be complex. The aim of the present article was to review bile salt structure and their application as absorption enhancers and the probable mechanism for increasing permeation based on previous studies.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-33184, Iran.
| | - Abdulghani Ameri
- Department of Drug and Food Control, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-33184, Iran.
| | - Somayeh Handali
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-33184, Iran.
| |
Collapse
|
32
|
Hazzah HA, Farid RM, Nasra MMA, Hazzah WA, El-Massik MA, Abdallah OY. Gelucire-Based Nanoparticles for Curcumin Targeting to Oral Mucosa: Preparation, Characterization, and Antimicrobial Activity Assessment. J Pharm Sci 2015. [PMID: 26202796 DOI: 10.1002/jps.24590] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The purpose of the study was to prepare and characterize curcumin (Cur) solid lipid nanoparticles (CurSLN) with a high-loading capacity and chemical stability for the treatment of oral mucosal infection. CurSLN were formulated using different lipids, namely, Gelucire 39/01, Gelucire 50/13, Precirol, Compritol, and poloxamer 407 as a surfactant. Formulae were evaluated for their entrapment efficiency, particle size, and ex vivo mucoadhesion test. Microbiological evaluation was carried out on six microorganisms, five of which are the most commonly affecting oral cavity in terms of determination of minimum inhibitory concentration (MIC), and minimum bactericidal concentration. Transmission electron microscopy was conducted for ultrathin section for Candida albicans-treated with formulated Cur. The results showed high entrapment efficiency and stability enhancement for Cur powder. Significant amount of Cur was retained onto the mucosal tissue indicating preferential mucosal uptake. CurSLN showed higher antimicrobial activity as compared with Cur raw material and chemically stabilized Cur where it showed MIC (0.185, 0.09375, 0.75, 3, 1.5, and 0.1875 mg/mL) against Staphylococcus aureus, Streptococcus mutans, Viridansstrept, Escherichia coli, Lactobacillus acidophilus, and Candida albicans, respectively. The prepared lipid nanoparticles maintained Cur chemical stability and microbiological activity. The lack of local antimicrobial therapeutics with minimum side effects augments the importance of studying natural products for this purpose.
Collapse
Affiliation(s)
- Heba A Hazzah
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt.
| | - Ragwa M Farid
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Maha M A Nasra
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Walaa A Hazzah
- Department of Microbiology, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Magda A El-Massik
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
33
|
Hazzah HA, Farid RM, Nasra MMA, Zakaria M, Gawish Y, El-Massik MA, Abdallah OY. A new approach for treatment of precancerous lesions with curcumin solid-lipid nanoparticle-loaded gels: in vitro and clinical evaluation. Drug Deliv 2015; 23:1409-19. [PMID: 26146889 DOI: 10.3109/10717544.2015.1065524] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Preparation and characterization of curcumin solid-lipid nanoparticle (CurSLN)-loaded mucoadhesive gel for local treatment of oral precancerous lesions with low dose. METHODOLOGY The formulated CurSLNs were dispersed in a mucoadhesive gel matrix to be applied to the buccal mucosa. Conventional mucoadhesive gel using binary system was adopted. The prepared gels were evaluated for in vitro drug dialysis, ex vivo mucoadhesion test and ex vivo permeation study using chicken buccal mucosa. Short-term clinical evaluation was carried out on 10 patients suffering oral erythroplakia in terms of pain index and lesion size measurement. (1) RESULTS: The results showed that the loaded gel with CurSLN showed good mucoadhesion property and 25 min in vivo residence time. In addition to stability enhancement for the Cur powder. All formulae did not show any drug permeated, however, significant amount of Cur was retained within the chicken buccal mucosal tissue confirmed by histological examination. Significant reduction in pain, and complete healing was observed after 6 weeks of treatment. CONCLUSION The local use of Cur in low dose is a promising option for treatment of precancerous lesions. The lack of local anti-inflammatory compounds with reduced side effects intensifies the importance of studying natural products for this purpose.
Collapse
Affiliation(s)
- Heba A Hazzah
- a Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing , Pharos University in Alexandria , Alexandria , Egypt
| | - Ragwa M Farid
- a Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing , Pharos University in Alexandria , Alexandria , Egypt
| | - Maha M A Nasra
- b Department of Pharmaceutics, Faculty of Pharmacy , and
| | - Mennatallah Zakaria
- c Department of Oral Medicine, Periodontology, Oral Diagnosis and Radiology, Faculty of Dentistry , Alexandria University , Alexandria , Egypt
| | - Yousria Gawish
- c Department of Oral Medicine, Periodontology, Oral Diagnosis and Radiology, Faculty of Dentistry , Alexandria University , Alexandria , Egypt
| | - Magda A El-Massik
- a Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing , Pharos University in Alexandria , Alexandria , Egypt
| | | |
Collapse
|
34
|
Acharya SP, Pundarikakshudu K, Panchal A, Lalwani A. Development of carbamazepine transnasal microemulsion for treatment of epilepsy. Drug Deliv Transl Res 2015; 3:252-9. [PMID: 25788134 DOI: 10.1007/s13346-012-0126-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Carbamazepine is widely preferred therapy for the treatment of epilepsy. However, oral therapy results in slower brain uptake and systemic side effects. Intranasal route can achieve faster brain uptake, but poor aqueous solubility of carbamazepine is the main obstacle for administration by nasal route. The purpose of this study was to prepare and evaluate intranasal oil in water microemulsion of carbamazepine to improve its solubility and enhance the brain uptake. Intranasal microemulsion of carbamazepine was prepared by water titration method using oleic acid as oil, Tween 80 as surfactant and Transcutol® as cosurfactant. Microemulsions were evaluated for various physical parameters including globule size, viscosity, pH and conductivity. Toxicity study of microemulsion was carried out by employing sheep nasal mucosa. The microemulsion was also evaluated by maximal electric shock, and the brain uptake study was done using HPLC method. The microemulsion was stable and transparent with average globule size of 21.03 nm and did not show any toxic symptoms. It showed reduction in the hind limb extension phase and faster recovery from seizures in comparison to oral microemulsion and nasal solution. Higher brain/plasma ratio was obtained with nasal microemulsion in comparison to ratio obtained after intraperitoneal injection of carbamazepine solution.
Collapse
|
35
|
Choi YA, Yoon YH, Choi K, Kwon M, Goo SH, Cha JS, Choi MK, Lee HS, Song IS. Enhanced Oral Bioavailability of Morin Administered in Mixed Micelle Formulation with PluronicF127 and Tween80 in Rats. Biol Pharm Bull 2015; 38:208-17. [DOI: 10.1248/bpb.b14-00508] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yeon Ah Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | - You Hyun Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | - Kwangik Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | - Mihwa Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | - Soo Hyeon Goo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| | | | | | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea
| | - Im-Sook Song
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University
| |
Collapse
|
36
|
Li Y, Li J, Zhang X, Ding J, Mao S. Non-ionic surfactants as novel intranasal absorption enhancers: in vitro and in vivo characterization. Drug Deliv 2014; 23:2272-2279. [PMID: 25347689 DOI: 10.3109/10717544.2014.971196] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To explore the potential of non-ionic surfactants as novel intranasal absorption enhancers. METHODS Taking sumatriptan succinate (SMS) as a model drug, influence of different non-ionic surfactants, including laurate sucrose ester (SE), cremophor EL and poloxamer 188, on the intranasal absorption of SMS was investigated using an in situ nasal perfusion technique in rats. Ciliotoxicity of the non-ionic surfactants was evaluated using an in situ toad palate model. In vivo behavior of the selected formulations was studied in rats. RESULTS All the non-ionic surfactants investigated increased the intranasal absorption of SMS remarkably but with varied extent and trend. Moreover, it was revealed that at the same concentration, laurate SE had better permeation-enhancing effect than that of cremophor EL and poloxamer 188. The ciliotoxicity results showed that all the non-ionic surfactants were regarded as safe at selected concentrations. Based on the in situ absorption data and ciliotoxicity results, the following three samples, 0.5% laurate SE, 0.1% cremophor EL and 0.5% poloxamer 188 were selected for in vivo absorption studies in rats. Among them, 0.5% laurate SE group presented the highest enhancing effect, followed by 0.1% cremophor EL and 0.5% poloxamer 188 group, with absolute bioavailability 29.99%, 22.64% and 20.90%, respectively. CONCLUSIONS Laurate SE is a promising intranasal absorption enhancer.
Collapse
Affiliation(s)
- Ying Li
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , People's Republic of China
| | - Jinfeng Li
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , People's Republic of China
| | - Xin Zhang
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , People's Republic of China
| | - Jiaojiao Ding
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , People's Republic of China
| | - Shirui Mao
- a School of Pharmacy , Shenyang Pharmaceutical University , Shenyang , People's Republic of China
| |
Collapse
|
37
|
Shubber S, Vllasaliu D, Rauch C, Jordan F, Illum L, Stolnik S. Mechanism of mucosal permeability enhancement of CriticalSorb® (Solutol® HS15) investigated in vitro in cell cultures. Pharm Res 2014; 32:516-27. [PMID: 25190006 PMCID: PMC4300420 DOI: 10.1007/s11095-014-1481-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/15/2014] [Indexed: 02/02/2023]
Abstract
Purpose CriticalSorb™, with the principal component Solutol® HS15, is a novel mucosal drug delivery system demonstrated to improve the bioavailability of selected biotherapeutics. The intention of this study is to elucidate mechanism(s) responsible for the enhancement of trans-mucosal absorption of biological drugs by Solutol® HS15. Methods Micelle size and CMC of Solutol® HS15 were determined in biologically relevant media. Polarised airway Calu-3 cell layers were used to measure the permeability of a panel of biological drugs, and to assess changes in TEER, tight junction and F-actin morphology. The rate of cell endocytosis was measured in vitro in the presence of Solutol® HS15 using a membrane probe, FM 2–10. Results This work initially confirms surfactant-like behaviour of Solutol® HS15 in aqueous media, while subsequent experiments demonstrate that the effect of Solutol® HS15 on epithelial tight junctions is different from a ‘classical’ tight junction opening agent and illustrate the effect of Solutol® HS15 on the cell membrane (endocytosis rate) and F-actin cytoskeleton. Conclusion Solutol® HS15 is the principle component of CriticalSorb™ that has shown an enhancement in permeability of medium sized biological drugs across epithelia. This study suggests that its mechanism of action arises primarily from effects on the cell membrane and consequent impacts on the cell cytoskeleton in terms of actin organisation and tight junction opening.
Collapse
Affiliation(s)
- Saif Shubber
- Division of Drug Delivery and Tissue Engineering, School of Pharmacy Boots Science Building, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | | | | | | | | | | |
Collapse
|
38
|
Salimi A, Motaharitabar E, Goudarzi M, Rezaie A, Kalantari H. Toxicity evaluation of microemulsion (nano size) of sour cherry kernel extract for the oral bioavailability enhancement. Jundishapur J Nat Pharm Prod 2014; 9:16-23. [PMID: 24644434 PMCID: PMC3957138 DOI: 10.17795/jjnpp-14370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/20/2013] [Accepted: 11/20/2013] [Indexed: 12/24/2022] Open
Abstract
Background: In the recent years nanostructured materials have been the focus of researches due to their wide-spread possibilities to provide new shapes and structures for some materials. Microemulsions can provide uniform nano-sized droplets for templating. Microemulsions are isotropic, thermodynamically-stable systems of oil, water and surfactant with a 20-200 nm droplet size. They can be prepared as oil-in-water (o/w), water-in-oil (w/o) or bicontinuous systems, depending on the equilibrium spontaneous curvature of the surfactant layer at the oil-water interface. Objectives: The aim of this study was to introduce a system designed to improve and enhance the bioavailability of bioflavonoids in the Prunus cerasus (sour cherry) seed kernel extract by developing a novel delivery system, i.e. microemulsion (nanosized particles). Materials and Methods: Microemulsion formulations were prepared by mixing appropriate amounts of surfactants (Tween 80 and Span 20), cosurfactant (propylene glycol) (3:1 ratio), and oil phase (olive oil). The prepared microemulsions were evaluated regarding their mean droplet size, transparency, viscosity, and pH. Sour cherry kernel extract microemulsion was orally administered to mice at doses of 2.5%, 5%, and 10% for 10 days. On the last day, their blood as well as their liver and kidney were used for biochemical and histopathological analyses, respectively. Results: Biochemical factors levels and the pathological study indicated that there were not significant differences in microemulsion extracts compared with the control group (P > 0.05). Conclusions: Not only no toxicity evidence of this product was observed in the dose range used in foods or healthcare, but also it improved the cardiac function recovery.
Collapse
Affiliation(s)
- Anayatollah Salimi
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Eisa Motaharitabar
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, IR Iran
| | - Mehdi Goudarzi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, IR Iran
| | - Annahita Rezaie
- Department of pathobiology, School of Veterinary Medicine, Shahid Chamran University, Ahvaz, IR Iran
| | - Heibatullah Kalantari
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, IR Iran
| |
Collapse
|
39
|
White JA, Blum JS, Hosken NA, Marshak JO, Duncan L, Zhu C, Norton EB, Clements JD, Koelle DM, Chen D, Weldon WC, Steven Oberste M, Lal M. Serum and mucosal antibody responses to inactivated polio vaccine after sublingual immunization using a thermoresponsive gel delivery system. Hum Vaccin Immunother 2014; 10:3611-21. [PMID: 25483682 PMCID: PMC4514067 DOI: 10.4161/hv.32253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/23/2014] [Accepted: 08/04/2014] [Indexed: 01/27/2023] Open
Abstract
Administering vaccines directly to mucosal surfaces can induce both serum and mucosal immune responses. Mucosal responses may prevent establishment of initial infection at the port of entry and subsequent dissemination to other sites. The sublingual route is attractive for mucosal vaccination, but both a safe, potent adjuvant and a novel formulation are needed to achieve an adequate immune response. We report the use of a thermoresponsive gel (TRG) combined with a double mutant of a bacterial heat-labile toxin (dmLT) for sublingual immunization with a trivalent inactivated poliovirus vaccine (IPV) in mice. This TRG delivery system, which changes from aqueous solution to viscous gel upon contact with the mucosa at body temperature, helps to retain the formulation at the site of delivery and has functional adjuvant activity from the inclusion of dmLT. IPV was administered to mice either sublingually in the TRG delivery system or intramuscularly in phosphate-buffered saline. We measured poliovirus type-specific serum neutralizing antibodies as well as polio-specific serum Ig and IgA antibodies in serum, saliva, and fecal samples using enzyme-linked immunosorbent assays. Mice receiving sublingual vaccination via the TRG delivery system produced both mucosal and serum antibodies, including IgA. Intramuscularly immunized animals produced only serum neutralizing and binding Ig but no detectable IgA. This study provides proof of concept for sublingual immunization using the TRG delivery system, comprising a thermoresponsive gel and dmLT adjuvant.
Collapse
Key Words
- CT, cholera toxin
- DPBS, Dulbecco's phosphate-buffered saline
- DU, D-antigen units
- ELISA, enzyme-linked immunosorbent assay
- IM, intramuscular
- IPV, inactivated poliovirus vaccine
- IgA, immunoglobulin A
- IgG, immunoglobulin G
- OPV, oral poliovirus vaccine
- PBS, phosphate-buffered saline
- RT, room temperature
- SL, sublingual
- SSI, Staten Serum Institute
- TMB, tetramethylbenzidine
- TRG, thermoresponsive gel
- adjuvants
- dmLT
- dmLT, double mutant heat-labile toxin
- mucosal immune response
- poliovirus
- sublingual immunization
- thermoresponsive gel
- vaccine delivery
Collapse
Affiliation(s)
| | | | - Nancy A Hosken
- Department of Medicine; University of Washington; Seattle, WA USA
| | - Joshua O Marshak
- Department of Medicine; University of Washington; Seattle, WA USA
| | | | | | - Elizabeth B Norton
- Department of Microbiology and Immunology; Tulane University School of Medicine; New Orleans, LA USA
| | - John D Clements
- Department of Microbiology and Immunology; Tulane University School of Medicine; New Orleans, LA USA
| | - David M Koelle
- Department of Medicine; University of Washington; Seattle, WA USA
- Department of Laboratory Medicine; University of Washington; Seattle, WA USA
- Vaccine and Infectious Diseases Division; Fred Hutchinson Cancer Research Institute; Seattle, WA USA
- Department of Global Health; University of Washington; Seattle, WA USA
| | | | - William C Weldon
- Division of Viral Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | - M Steven Oberste
- Division of Viral Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | | |
Collapse
|
40
|
Gelatin nanostructured lipid carriers-mediated intranasal delivery of basic fibroblast growth factor enhances functional recovery in hemiparkinsonian rats. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:755-64. [PMID: 24200526 DOI: 10.1016/j.nano.2013.10.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 10/03/2013] [Accepted: 10/23/2013] [Indexed: 11/23/2022]
Abstract
UNLABELLED Lipid nanoparticles with solid matrix have been given increasing attention due to their biodegradable status and ability to entrap a variety of biologically active compounds. In this study, new phospholipid-based gelatin nanoparticles encapsulating basic fibroblast growth factor (bFGF) were developed to target the brain via nasal administration. Treatment effects were assessed by quantifying rotational behavior, monoamine neurotransmitter levels and tyrosine hydroxylase expression in 6-hydroxydopamine induced hemiparkinsonian rats. The gelatin nanostructured lipid carriers (GNLs) were prepared by a water-in-water emulsion method and then freeze-dried. The GNLs possessed better profile than gelatin nanoparticles (GNs), with particle size 143±1.14nm and Zeta potential -38.2±1.2mV. The intranasal GNLs efficiently enriched exogenous bFGF in olfactory bulb and striatum without adverse impact on the integrity of nasal mucosa and showed obvious therapeutic effects on hemiparkinsonian rats. Thus, GNLs are attractive carriers for nose-to-brain drug delivery, especially for unstable macromolecular drugs such as bFGF. FROM THE CLINICAL EDITOR This team of authors reports the development of phospholipid-based gelatin nanoparticles encapsulating basic fibroblast growth factor to target the brain via intranasal administration. A rat model of hemiparkinsonism was applied demonstrating a good safety profile and an obvious therapeutic effect.
Collapse
|
41
|
Chitosan and cyclodextrin in intranasal microemulsion for improved brain buspirone hydrochloride pharmacokinetics in rats. Carbohydr Polym 2013; 99:297-305. [PMID: 24274510 DOI: 10.1016/j.carbpol.2013.08.027] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/13/2013] [Accepted: 08/13/2013] [Indexed: 12/21/2022]
Abstract
The aim of this study was to develop buspirone hydrochloride microemulsion formulations for intranasal administration to improve the drug bioavailability and provide high drug brain levels. For the purpose, chitosan aspartate, and hydroxypropyl-β-cyclodextrin were incorporated in the microemulsions. The prepared formulations were characterized. Biological investigations including pharmacokinetic studies, brain drug targeting efficiency determinations and histopathological examinations were performed on rats. The results showed that safe and stable mucoadhesive microemulsion suitable for nasal administration were successfully prepared. Ex vivo drug permeation revealed high drug permeation from microemulsions. Absolute bioavailability after intranasal administration of buspirone mucoadhesive microemulsion increased significantly and plasma concentration peaked at 15 min. The AUC0-360(brain) was 3 times that obtained after intravenous administration. A high brain targeting efficiency (86.6%) and a direct nose to brain transport (88%) confirmed the direct nose to brain transport of buspirone following nasal administration of the microemulsions.
Collapse
|
42
|
Ali H, Kalashnikova I, White MA, Sherman M, Rytting E. Preparation, characterization, and transport of dexamethasone-loaded polymeric nanoparticles across a human placental in vitro model. Int J Pharm 2013; 454:149-57. [PMID: 23850397 DOI: 10.1016/j.ijpharm.2013.07.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 06/25/2013] [Accepted: 07/02/2013] [Indexed: 01/16/2023]
Abstract
The purpose of this study was to prepare dexamethasone-loaded polymeric nanoparticles and evaluate their potential for transport across human placenta. Statistical modeling and factorial design was applied to investigate the influence of process parameters on the following nanoparticle characteristics: particle size, polydispersity index, zeta potential, and drug encapsulation efficiency. Dexamethasone and nanoparticle transport was subsequently investigated using the BeWo b30 cell line, an in vitro model of human placental trophoblast cells, which represent the rate-limiting barrier for maternal-fetal transfer. Encapsulation efficiency and drug transport were determined using a validated high performance liquid chromatography method. Nanoparticle morphology and drug encapsulation were further characterized by cryo-transmission electron microscopy and X-ray diffraction, respectively. Nanoparticles prepared from poly(lactic-co-glycolic acid) were spherical, with particle sizes ranging from 140 to 298 nm, and encapsulation efficiency ranging from 52 to 89%. Nanoencapsulation enhanced the apparent permeability of dexamethasone from the maternal compartment to the fetal compartment more than 10-fold in this model. Particle size was shown to be inversely correlated with drug and nanoparticle permeability, as confirmed with fluorescently labeled nanoparticles. These results highlight the feasibility of designing nanoparticles capable of delivering medication to the fetus, in particular, potential dexamethasone therapy for the prenatal treatment of congenital adrenal hyperplasia.
Collapse
Affiliation(s)
- Hazem Ali
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | |
Collapse
|
43
|
Pepić I, Lovrić J, Filipović-Grčić J. How do polymeric micelles cross epithelial barriers? Eur J Pharm Sci 2013; 50:42-55. [PMID: 23619286 DOI: 10.1016/j.ejps.2013.04.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/27/2013] [Accepted: 04/07/2013] [Indexed: 12/22/2022]
Abstract
Non-parenteral delivery of drugs using nanotechnology-based delivery systems is a promising non-invasive way to achieve effective local or systemic drug delivery. The efficacy of drugs administered non-parenterally is limited by their ability to cross biological barriers, and epithelial tissues particularly present challenges. Polymeric micelles can achieve transepithelial drug delivery because of their ability to be internalized into cells and/or cross epithelial barriers, thereby delivering drugs either locally or systematically following non-parenteral administration. This review discusses the particular characteristics of various epithelial barriers and assesses their potential as non-parenteral routes of delivery. The material characteristics of polymeric micelles (e.g., size, surface charge, and surface decoration) and of unimers dissociated from polymeric micelles determine their interactions (non-specific and/or specific) with mucus and epithelial cells as well as their intracellular fate. This paper outlines the mechanisms governing the major modes of internalization of polymeric micelles into epithelial cells, with an emphasis on specific recent examples of the transport of drug-loaded polymeric micelles across epithelial barriers.
Collapse
Affiliation(s)
- Ivan Pepić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia.
| | | | | |
Collapse
|
44
|
Acharya SP, Pundarikakshudu K, Panchal A, Lalwani A. Preparation and evaluation of transnasal microemulsion of carbamazepine. Asian J Pharm Sci 2013. [DOI: 10.1016/j.ajps.2013.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
45
|
Doh HJ, Jung Y, Balakrishnan P, Cho HJ, Kim DD. A novel lipid nanoemulsion system for improved permeation of granisetron. Colloids Surf B Biointerfaces 2013; 101:475-80. [DOI: 10.1016/j.colsurfb.2012.07.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/07/2012] [Accepted: 07/13/2012] [Indexed: 11/24/2022]
|
46
|
Phospholipid based colloidal poloxamer–nanocubic vesicles for brain targeting via the nasal route. Colloids Surf B Biointerfaces 2012; 100:146-54. [DOI: 10.1016/j.colsurfb.2012.05.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/07/2012] [Accepted: 05/11/2012] [Indexed: 11/20/2022]
|
47
|
Nesseem DI, Michel CG. Development and characterization of local anti-inflammatory implantation for the controlled release of the hexane extract of the flower-heads of Euryops pectinatus L. (Cass.). Drug Discov Ther 2012; 5:96-106. [PMID: 22466147 DOI: 10.5582/ddt.2011.v5.2.96] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A hexane extract of the flower-heads of Euryops pectinatus L. (Cass.) was formulated into local anti-inflammatory implantation patches with controlled release. Cross-linked sodium hyaluronate patches (F1-F3) and chitosan patches (F4-F6) were prepared by a casting/solvent evaporation technique. Morphological and mechanical characterizations including the components ratio, surfactant and the loaded amount of the hexane extract (50, 100, and 200 mg/kg b.wt.) were investigated. Release studies were performed during 24 h using a diffusion cell. Films with optimum in vitro release rate have been investigated for testing the anti-inflammatory activity and the sustaining effect of the formulations. The sustained anti-inflammatory effect of the hexane extract of E. pectinatus flower-heads from the selected films was studied by inducing paw edema in rats with 1% (w/v) carrageenan solution. The results indicated the compatibility of hexane extract with both sodium hyaluronate and chitosan patches forming yellowish transparent films. Based on variations in drug release profiles throughout the 24-h among the formulations (F1-F6) studies, F3 and F6 were selected for further investigation. When the films were applied 1 h before the subplantar injection of carrageenan in the hind paw of male Albino rats, formulation (F3) provided its maximum inhibition of paw edema in rats (91.3%) 4 h after edema induction whereas, formulation (F6) showed less inhibition after 4 h (70.6%). The previous two formulations (F3 and F6) produced potent results (95.3 and 89.5%, respectively) after 24 h when compared with a local market preparation containing 25% β-sitosterol used as positive control. Histophathological investigation was conducted for 1, 4, and 12 weeks to study the tissue response for the two formulations (F3 and F6) at the implantation site. Chemical investigation of the hexane extract was achieved for both unsaponifiable matter (USM) and fatty acid methyl esters (FAME) using gas liquid chromatography (GLC). The USM was dominated by n-pentacosane (14.40%), phytosterols (Cholesterol, Campesterol, Stigmasterol, β-sitosterol, α-amyrin) reached 33.44% and the FAME was dominated by Linoleinic (49.97%). Quality control of the local implantation was evaluated by GLC using cholesterol as an analytical marker and phytosterols as an active marker compared to the plain extract.
Collapse
Affiliation(s)
- D I Nesseem
- Pharmaceutics Department, National Organization for Drug Control & Research, Cairo, Egypt
| | | |
Collapse
|
48
|
Salama HA, Mahmoud AA, Kamel AO, Abdel Hady M, Awad GAS. Brain delivery of olanzapine by intranasal administration of transfersomal vesicles. J Liposome Res 2012; 22:336-45. [PMID: 22881283 DOI: 10.3109/08982104.2012.700460] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study was to investigate the presence of a possible direct correlation between vesicle elasticity and the amount of drug reaching the brain intranasally. Therefore, transfersomes were developed using phosphatidylcholine (PC) as the lipid matrix and sodium deoxycholate (SDC), Span® 60, Cremophor® EL, Brij® 58, and Brij® 72 as surfactants. The influence of the type of surfactant and PC-to-surfactant ratio on vesicle morphology, size, membrane elasticity, drug entrapment, and in vitro drug release was studied. The prepared transfersomes were mainly spherical in shape, with diameters ranging from 310 to 885 nm. Transfersomes containing SDC and Span 60 with optimum lipid-to-surfactant molar ratio showed suitable diameters (410 and 380 nm, respectively) and deformability indices (17.68 and 20.76 mL/sec, respectively). Values for absolute drug bioavailability in rat plasma for transfersomes containing SDC and those containing Span 60 were 24.75 and 51.35%, whereas AUC(0-360 min) values in rat brain were 22,334.6 and 36,486.3 ng/mL/min, respectively. The present study revealed that the deformability index is a parameter having a direct relation with the amount of the drug delivered to the brain by the nasal route.
Collapse
Affiliation(s)
- Hamed A Salama
- Department of Pharmaceutical Technology, National Research Center, Dokki, Cairo, Egypt
| | | | | | | | | |
Collapse
|
49
|
Ujhelyi Z, Fenyvesi F, Váradi J, Fehér P, Kiss T, Veszelka S, Deli M, Vecsernyés M, Bácskay I. Evaluation of cytotoxicity of surfactants used in self-micro emulsifying drug delivery systems and their effects on paracellular transport in Caco-2 cell monolayer. Eur J Pharm Sci 2012; 47:564-73. [PMID: 22841998 DOI: 10.1016/j.ejps.2012.07.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 07/05/2012] [Accepted: 07/05/2012] [Indexed: 10/28/2022]
Abstract
The objective of this study was to examine the cellular effects of the members of two non-ionic amphiphilic tenside groups and their mixtures on human Caco-2 cell monolayers as dependent upon their chemical structures and physicochemical properties. The first group of polyethylene glycol esters is represented by Polysorbates and Labrasol alone and in blends, while the members of the second group. Capryol 90, Capryol PGMC, Lauroglycol 90 and Lauroglycol FCC were used as propylene glycol esters. They are increasingly used in SMEDDS as recent tensides or co-tensides to increase hydrophobic bioavailability of a drug. Critical micelle concentration was measured by determination of surface tension. CMC refers to the ability of solubilization of surfactants. Cytotoxicity tests were performed on Caco-2 cell monolayers by MTT and LDH methods. Paracellular permeability as a marker of the integrity of cell monolayers, was examined with Lucifer yellow assays combined with TransEpithelial Electrical Resistance (TEER) measurements. The effect of these surfactants on tight junctions as evidence for paracellular pathway was also characterized. The results of cytotoxicity assays were in agreement, and showed significant differences among the cytotoxic properties of surfactants in a concentration-dependent manner. Polysorbates 20, 60, 80 are the most toxic compounds. In the case of Labrasol, the degree of esterification and lack of sorbit component decreased cytotoxicity. If the hydrophyl head was changed from polyethylene glycol to propylene glycol the main determined factor of cytotoxicity was the monoester content and the length of carbon chain. In our CMC experiments, we found that only Labrasol showed expressed cytotoxicity above the CMC. It refers to good ability of micelle solubilization of Labrasol. In our paracellular transport experiments each of polyethylene glycol surfactants (Polysorbates and Labrasol) altered TEER values, but propylene glycol esters did not modify the monolayer integrity. Polyethylene glycol esters alone and in blends (0.05% Labrasol--0.001% Polysorbates 20, 60, 80) were able to increase Lucifer yellow permeability significantly below the IC₅₀ concentration. On the other hand Labrasol and Polysorbates 20 have expressed effect on tight junctions of Caco-2 monolayer. It could be concluded that polyethylene glycol ester-type tensides were able to enhance the paracellular permeability by the redistribution of junctional proteins. Our results might ensure useful data for selection of suitable tensides, co-tensides and tenside mixtures for SMEDDS formulations.
Collapse
Affiliation(s)
- Zoltán Ujhelyi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Medical and Health Science Center, University of Debrecen, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang J, Liu D, Huang Y, Gao Y, Qian S. Biopharmaceutics classification and intestinal absorption study of apigenin. Int J Pharm 2012; 436:311-7. [PMID: 22796171 DOI: 10.1016/j.ijpharm.2012.07.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/30/2012] [Accepted: 07/02/2012] [Indexed: 11/28/2022]
Abstract
The aim of the study was to characterize the biopharmaceutics classification system (BCS) category of apigenin (AP) using intrinsic dissolution rate (IDR) and rat intestinal permeability, and to investigate the intestinal absorption mechanism of AP in rats. In the present investigation, equilibrium solubility and intrinsic dissolution rate (IDR) of AP were estimated in phosphate buffers. Effective intestinal permeability (P(eff)) of AP was determined using single-pass intestinal perfusion (SPIP) technique in four segments (duodenum, jejunum, ileum and colon) of rat intestine at three concentrations (10, 50 and 100 μg/ml). The aqueous solubility of AP in tested phosphate buffers was very poor with maximum solubility of 2.16 μg/ml at pH 7.5. The IDR of AP was very low with a value of 0.006 mg/min/cm(2). The minimum and maximum P(eff)s determined by SPIP were 0.198×10(-4) and 0.713×10(-4) cm/s at jejunum and duodenum site, respectively. In addition, the concentration-dependent permeability behavior was observed in the duodenum and jejunum, which suggested that AP was transported by both passive and active carrier-mediated saturable mechanism in these two intestinal segments. However, the observed concentration-independent permeability behavior in ileum and colon indicated primarily passive transport mechanism of absorption of AP in the last two intestinal segments. AP was classified as class II drug of the BCS due to its low solubility and high intestinal permeability. AP could be well absorbed in the whole intestine with the main absorption site at duodenum. The absorption of AP in four intestinal segments exhibited different transport mechanisms.
Collapse
Affiliation(s)
- Jianjun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | |
Collapse
|