1
|
Lee D, Shen AM, Shah M, Garbuzenko OB, Minko T. In Vivo Evaluation of Nose-to-Brain Delivery of Liposomal Donepezil, Memantine, and BACE-1 siRNA for Alzheimer's Disease Therapy. Int J Mol Sci 2024; 25:10357. [PMID: 39408684 PMCID: PMC11476875 DOI: 10.3390/ijms251910357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Our study took an innovative approach by evaluating, in vivo, the efficacy of intranasal (IN) administration of liposomal formulations of donepezil, memantine, and beta-site amyloid precursor protein-cleaving enzyme (BACE-1) siRNA, and their combination as a "triple-drug therapy" in treating Alzheimer's disease (AD). Female APP/PS1 homozygous, transgenic mice were used as an AD model. The spatial short-term memory of the APP/PS1 mice was evaluated by a Y-maze behavioral test. IN-administered formulations demonstrated better short-term memory recovery than oral administration. Triple-drug therapy induced short-term memory recovery and lowered beta-amyloid (Aβ) 40 and 42 peptide levels and BACE-1 mRNA expression. Additionally, inflammatory cytokine mRNA expression was downregulated. This innovative approach opens new possibilities for Alzheimer's disease treatment and nose-to-brain delivery.
Collapse
Affiliation(s)
- David Lee
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Andrew M. Shen
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Milin Shah
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Olga B. Garbuzenko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Environmental and Occupational Health Science Institute, Piscataway, NJ 08854, USA
| |
Collapse
|
2
|
Wang J, Qiao Z, Li Q, Yu X, Zhang L, Ma C. Exploring the oral processing mechanisms for saltiness enhancement with NaCl nanocrystals in oleogel systems based on black pepper oleoresin and sunflower seed oil. Food Chem 2024; 463:141258. [PMID: 39298855 DOI: 10.1016/j.foodchem.2024.141258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Reducing salt content in processed foods while maintaining flavor is crucial for human health. This study investigates the sensory interaction between the pungency of black pepper oleoresin and saltiness within an oleogel matrix by incorporating NaCl nanocrystals into a black pepper oleoresin formulated with glyceryl monostearate and soy lecithin (G-L-P). Psychophysical results revealed that the salt content in the G-L-P and G-L systems (G-L-P without black pepper oleoresin) was reduced by up to 66.77 % and 37.08 % compared to the G system (G-L without soy lecithin), respectively. During oral processing, the G-L and G-L-P systems formed oil-in-water emulsions with smaller and more numerous oil droplets. Within 5 s, the Na+ concentration was lowest in the G system (2.2 g/L), while the G-L and G-L-P systems showed higher concentrations (3.1 g/L and 3.0 g/L, respectively). This research provides a theoretical basis for the food industry to develop low-salt products without compromising flavor.
Collapse
Affiliation(s)
- Jiajie Wang
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China
| | - Zeyao Qiao
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China
| | - Qianqian Li
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China
| | - Xinyu Yu
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China
| | - Lulu Zhang
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China; State Key Laboratory of Efficient Production of Forest Resources, Beijing Forestry University, Beijing 100083, China.
| | - Chao Ma
- Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Key Laboratory of Forest Food Processing and Safety, Beijing Forestry University, Beijing 100083, China; State Key Laboratory of Efficient Production of Forest Resources, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
3
|
Villegas-Novoa C, Wang Y, Sims CE, Allbritton NL. Creation of a spatially complex mucus bilayer on an in vitro colon model. Sci Rep 2024; 14:16849. [PMID: 39039235 PMCID: PMC11263341 DOI: 10.1038/s41598-024-67591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
The colonic epithelium is comprised of three-dimensional crypts (3D) lined with mucus secreted by a heterogeneous population of goblet cells. In this study, we report the formation of a long-lived, and self-renewing replica of human 3D crypts with a mucus layer patterned in the X-Y-Z dimensions. Primary colon cells were cultured on a shaped scaffold under an air-liquid interface to yield architecturally accurate crypts with a mucus bilayer (605 ± 180 μm thick) possessing an inner (149 ± 50 μm) and outer (435 ± 111 μm) region. Lectins with distinct carbohydrate-binding preferences demonstrated that the mucus in the intercrypt regions was chemically distinct from that above and within the crypts replicating in vivo chemical patterning. Constitutive mucus secretion ejected beads from crypt lumens in 8-10 days, while agonist-stimulated secretion increased mucus thickness by 17-fold in 8 h. The tissue was long-lived, > 50 days, the longest time assessed. In conclusion, the in vitro mucus replicated key physiology of the human mucus, including the bilayer (Z) structure and intercrypt-crypt (X-Y) zones, constitutive mucus flow, spatially complex chemical attributes, and mucus secretion response to stimulation, with the potential to reveal local and global determinants of mucus function and its breakdown in disease.
Collapse
Affiliation(s)
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Christopher E Sims
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
4
|
Oral docetaxel delivery with cationic polymeric core-shell nanocapsules: In vitro and in vivo evaluation. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
5
|
Magnetically Driven Muco-Inert Janus Nanovehicles for Enhanced Mucus Penetration and Cellular Uptake. Molecules 2022; 27:molecules27217291. [DOI: 10.3390/molecules27217291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
One of the main challenges of transmucosal drug delivery is that of enabling particles and molecules to move across the mucosal barrier of the mucosal epithelial surface. Inspired by nanovehicles and mucus-penetrating nanoparticles, a magnetically driven, mucus-inert Janus-type nanovehicle (Janus-MMSN-pCB) was fabricated by coating the zwitterionic polymer poly(carboxybetaine methacrylate) (pCB) on the mesoporous silica nanorod, which was grown on one side of superparamagnetic Fe3O4 nanoparticle using the sol–gel method. X-ray diffraction, transmission electron microscopy, vibrating sample magnetometry, and Fourier infrared spectroscopy were used to characterize the structure and morphology of the nanovehicles, proving the success of each synthesis step. The in vitro cell viability assessment of these composites using Calu-3 cell lines indicates that the nanovehicles are biocompatible in nature. Furthermore, the multiparticle tracking, Transwell® system, and cell imaging experimental results demonstrate that both the modification of pCB and the application of a magnetic field effectively accelerated the diffusion of the nanovehicles in the mucus and improved the endocytosis through Calu-3. The favorable cell uptake performance of Janus-MMSN-pCB in mucus systems with/without magnetic driving proves its potential role in the diagnosis, treatment, and imaging of mucosal-related diseases.
Collapse
|
6
|
Kurano T, Kanazawa T, Ooba A, Masuyama Y, Maruhana N, Yamada M, Iioka S, Ibaraki H, Kosuge Y, Kondo H, Suzuki T. Nose-to-brain/spinal cord delivery kinetics of liposomes with different surface properties. J Control Release 2022; 344:225-234. [PMID: 35296406 DOI: 10.1016/j.jconrel.2022.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
The administration of liposomes via nose-to-brain delivery is expected to become a strategy for efficient drug delivery to the central nervous system. Efficient nose-to-brain delivery and the kinetics of drugs administered in this manner depend on the properties of liposomes. However, there is a lack of basic knowledge of which liposomes are suitable for this purpose. Here, a qualitative study of intranasally administered liposomes (positively charged, neutral, and negatively charged, with or without polyethylene glycol [PEG] modification; particle size <100 nm) was performed to elucidate their dynamics in the brain and spinal cord. Additionally, a quantitative investigation was performed to ascertain their distribution in each part of the brain and spinal cord. The effects of liposome surface charge and PEG modification on the kinetics and distribution post intranasal administration were investigated via two experiments. Qualitative evaluation was performed via ex vivo observation after intranasal administration of fluorescently labeled liposomes. Neutral PEG-modified liposomes were distributed throughout the brain and spinal cord 60 min after administration, and the fluorescence intensity increased with time. By contrast, non-PEG-modified neutral liposomes showed particularly strong fluorescence in the olfactory bulb, and the fluorescence was localized in the anterior part of the brain. Positively charged liposomes showed low fluorescence around the lateral part of the brain and lumbar spinal cord 60 min after administration. Low fluorescence was observed in the whole brain and spinal cord, with strong fluorescence being observed in the olfactory bulb after 120 min of administration. Negatively charged liposomes showed no fluorescence at 60 min after administration, but low fluorescence was observed throughout the brain and spinal cord 120 min after administration. We quantified the radioactivity in the brain and spinal cord after intranasal administration of radioisotope-labeled liposomes. Neutral liposomes showed the highest distribution by area under the drug concentration-time curve (AUC(60-120)) in the brain and spinal cord compared to other liposomes. Compared with negatively charged liposomes, positively charged liposomes had a higher distribution in the olfactory bulb and forebrain, while negatively charged liposomes had a higher distribution in the hindbrain and bulbospinal tract cord. In addition, the distribution of PEG-modified neutral liposomes in the brain and spinal cord was significantly enhanced compared to that of non-PEG-modified neutral liposomes after 90 min of intranasal administration. These results indicate that surface charge and PEG modification strongly affect the efficiency of nose-to-brain delivery kinetics, and that PEG-modified neutral liposomes are excellent carriers for drug delivery to a wide area of the brain and spinal cord.
Collapse
Affiliation(s)
- Takumi Kurano
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Takanori Kanazawa
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan; School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Aoi Ooba
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Yudai Masuyama
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Nao Maruhana
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Mayu Yamada
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Shingo Iioka
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hisako Ibaraki
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yasuhiro Kosuge
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Hiromu Kondo
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Toyofumi Suzuki
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| |
Collapse
|
7
|
Dave RS, Goostrey TC, Ziolkowska M, Czerny-Holownia S, Hoare T, Sheardown H. Ocular drug delivery to the anterior segment using nanocarriers: A mucoadhesive/mucopenetrative perspective. J Control Release 2021; 336:71-88. [PMID: 34119558 DOI: 10.1016/j.jconrel.2021.06.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022]
Abstract
There is a growing demand for effective treatments for ocular conditions that improve patient compliance and reduce side-effects. While methods such as implants and injections have proven effective, topical administration remains the method of choice for the delivery of therapeutics to the anterior segment of the eye. However, topical administration suffers from multiple drawbacks including low bioavailability of the target therapeutic, systemic toxicity, and the requirement for high therapeutic doses due to the effective clearance mechanisms that exist in the eye. Nanoparticles that have tunable mucoadhesion and/or mucopenetration offer outstanding potential to overcome the anatomical and physiological barriers present to improve ocular bioavailability, reduce toxicity, and increase ocular retention, among other benefits. The current review highlights recent advances in the field of developing nanocarriers with tunable mucoadhesion and mucopenetration for drug delivery to the eye.
Collapse
Affiliation(s)
- Ridhdhi S Dave
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Taylor C Goostrey
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Maya Ziolkowska
- Department of Integrated Biomedical Engineering & Health Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Sofia Czerny-Holownia
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.
| |
Collapse
|
8
|
Lebreton V, Legeay S, Saulnier P, Lagarce F. Specificity of pharmacokinetic modeling of nanomedicines. Drug Discov Today 2021; 26:2259-2268. [PMID: 33892140 DOI: 10.1016/j.drudis.2021.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 01/08/2023]
Abstract
Nanomedicines have been developed for more than four decades to optimize the pharmacokinetics (PK) of drugs, especially absorption, distribution, and stability in vivo. Unfortunately, only a few drug products have reached the market. One reason among others is the lack of proper PK modeling and evaluation, which impedes the optimization of these promising drug delivery systems. In this review, we discuss the specificity of nanomedicines and propose key parameters to take into account for future accurate PK evaluation of nanomedicine. We believe that this could help these innovative drug products to reach to market and change the fate of many diseases.
Collapse
Affiliation(s)
- Vincent Lebreton
- University of Angers, MINT Inserm 1066 CNRS 6021, Angers, France; CHU Angers, 4 Rue Larrey, 49033 Angers, France
| | - Samuel Legeay
- University of Angers, MINT Inserm 1066 CNRS 6021, Angers, France
| | - Patrick Saulnier
- University of Angers, MINT Inserm 1066 CNRS 6021, Angers, France; CHU Angers, 4 Rue Larrey, 49033 Angers, France
| | - Frederic Lagarce
- University of Angers, MINT Inserm 1066 CNRS 6021, Angers, France; CHU Angers, 4 Rue Larrey, 49033 Angers, France.
| |
Collapse
|
9
|
Bhattacharjee S, Brayden DJ. Addressing the challenges to increase the efficiency of translating nanomedicine formulations to patients. Expert Opin Drug Discov 2020; 16:235-254. [PMID: 33108229 DOI: 10.1080/17460441.2021.1826434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nanotechnology is in a growth phase for drug delivery and medical imaging. Nanomaterials with unique properties present opportunities for encapsulation of therapeutics and imaging agents, along with conjugation to ligands for targeting. Favorable chemistry of nanomaterials can create formulations that address critical challenges for therapeutics, such as insolubility and a low capacity to cross the blood-brain-barrier (BBB) and intestinal wall. AREAS COVERED The authors investigate challenges faced during translation of nanomedicines while suggesting reasons as to why some nanoformulations have under-performed in clinical trials. They assess physiological barriers such as the BBB and gut mucus that nanomedicines must overcome to deliver cargos. They also provide an overview with examples of how nanomedicines can be designed to improve localization and site-specific delivery (e.g., encapsulation, bioconjugation, and triggered-release). EXPERT OPINION There are examples where nanomedicines have demonstrated improved efficacy of payload in humans; however, most of the advantages conferred were in improved pharmacokinetics and reduced toxicity. Problematic data show susceptibility of nanoformulations against natural protective mechanisms present in the body, including distribution impediment by physiological barriers and activation of the reticuloendothelial system. Further initiatives should address current challenges while expanding the scope of nanomedicine into advanced biomedical imaging and antibiotic delivery.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - David J Brayden
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
10
|
Liu J, Leng P, Liu Y. Oral drug delivery with nanoparticles into the gastrointestinal mucosa. Fundam Clin Pharmacol 2020; 35:86-96. [PMID: 32749731 DOI: 10.1111/fcp.12594] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022]
Abstract
The oral route of protein and peptide drugs has been a popular method of drug delivery in recent years, although it is often a challenge to achieve effective drug release and minimize the barrier functions of the gastrointestinal tract. Gastrointestinal mucosa can capture and remove harmful substances; similarly, it can limit the absorption of drugs. Many drugs are effectively captured by the mucus and rapidly removed, making it difficult to control the release of drugs in the gastrointestinal tract. The use of drug carrier systems can overcome the mucosal barrier and significantly improve bioavailability. Nanoparticle drug carriers can protect the drug from degradation, transporting it to a predetermined location in the gastrointestinal tract to achieve more efficient and sustained drug delivery. It is becoming clearer that the characteristics of nanoparticles, such as particle size, charge, and hydrophobicity, are related to permeability of the mucosal barrier. This review focuses on the latest research progress of nanoparticles to penetrate the mucosal barrier, including the delivery methods of nanoparticles on the surface of gastrointestinal mucosa, and aims to summarize how successful oral nanoparticle delivery systems can overcome this biological barrier in the human body. In addition, the in vitro model based on gastrointestinal mucus is an important tool for drug research and development. Here, we discuss different types of drug delivery systems and their advantages and disadvantages in design and potential applications. Similarly, we reviewed and summarized various methods for evaluating oral nanoparticles in in vitro and in vivo models.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Ping Leng
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Yujun Liu
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| |
Collapse
|
11
|
Poovi G, Damodharan N. Development of tamoxifen-loaded surface-modified nanostructured lipid carrier using experimental design: in vitro and ex vivo characterisation. IET Nanobiotechnol 2020; 14:261-274. [PMID: 32463016 DOI: 10.1049/iet-nbt.2019.0276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The present study aimed to develop a surface-modified biocompatible nanostructured lipid carrier (NLCs) system using polyoxyethylene (40) stearate (POE-40-S) to improve the oral bioavailability of poorly water-soluble Biopharmaceutics Classification System class-II drug like tamoxifen (TMX). Also aimed to screen the most influential factors affecting the particle size (PS) using Taguchi (L12 (211)) orthogonal array design (TgL12OA). Then, to optimize the TMX loaded POE-40-S (P) surface-modified NLCs (TMX-loaded-PEG-40-S coated NLC (PNLCs) or PNLCs) by central composite design (CCD) using a four-factor, five-level model. The most influential factors affecting the PS was screened and optimized. The in-vitro study showed that increased drug-loading (DL) and encapsulation efficiency (EE), decreased PS and charge, sustained drug release for the prolonged period of the time with good stability and suppressed protein adsorption. The Ex-vivo study showed that decreased mucous binding with five-fold enhanced permeability of PNLC formulation after surface modification with POE-40-S. The in-vitro cytotoxicity study showed that the blank carrier is biocompatible and cytotoxicity of the formulation was dependent on the concentration of the drug. Finally, it can be concluded that the surface-modified PNLCs formulation was an effective, biocompatible, stable formulation in the enhancement of dissolution rate, solubility, stability with reduced mucus adhesion and increased permeability thereby which indicates its enhanced oral bioavailability.
Collapse
Affiliation(s)
- Ganesan Poovi
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Narayanasamy Damodharan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
12
|
Adnet T, Groo AC, Picard C, Davis A, Corvaisier S, Since M, Bounoure F, Rochais C, Le Pluart L, Dallemagne P, Malzert-Fréon A. Pharmacotechnical Development of a Nasal Drug Delivery Composite Nanosystem Intended for Alzheimer's Disease Treatment. Pharmaceutics 2020; 12:pharmaceutics12030251. [PMID: 32168767 PMCID: PMC7151011 DOI: 10.3390/pharmaceutics12030251] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
Direct nose-to-brain delivery has been raised as a non-invasive powerful strategy to deliver drugs to the brain bypassing the blood-brain barrier (BBB). This study aimed at preparing and characterizing an innovative composite formulation, associating the liposome and hydrogel approaches, suitable for intranasal administration. Thermosensitive gel formulations were obtained based on a mixture of two hydrophilic polymers (Poloxamer 407, P407 and Poloxamer 188, P188) for a controlled delivery through nasal route via liposomes of an active pharmaceutical ingredient (API) of potential interest for Alzheimer’s disease. The osmolarity and the gelation temperature (T° sol-gel) of formulations, defined in a ternary diagram, were investigated by rheometry and visual determination. Regarding the issue of assays, a mixture composed of P407/P188 (15/1%, w/w) was selected for intranasal administration in terms of T° sol-gel and for the compatibility with the olfactory mucosal (280 ± 20 mOsmol, pH 6). Liposomes of API were prepared by the thin film hydration method. Mucoadhesion studies were performed by using mucin disc, and they showed the good natural mucoadhesive characteristics of in situ gel formulations, which increased when liposomes were added. The study demonstrated successful pharmacotechnical development of a promising API-loaded liposomes in a thermosensitive hydrogel intended for nasal Alzheimer’s disease treatment.
Collapse
Affiliation(s)
- Thomas Adnet
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
- CHU, 14000 Caen, France
| | - Anne-Claire Groo
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
- Correspondence: (A.-C.G.); (A.M.-F.); Tel.: +33-231-566819 (A.M.-F.)
| | - Céline Picard
- UNILEHAVRE, FR 3038 CNRS, URCOM, EA 3221, Normandie University,76063 Le Havre, France;
| | - Audrey Davis
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
| | - Sophie Corvaisier
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
| | - Marc Since
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
| | - Frédéric Bounoure
- UFR of Health, Laboratory of Pharmaceutical & Biopharmaceutical technology, UNIROUEN, Normandy University, 76183 Rouen CEDEX, France;
| | - Christophe Rochais
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
| | - Loïc Le Pluart
- LCMT, UMR CNRS 6507, EnsiCaen UniCaen, 14000 Caen, France;
| | - Patrick Dallemagne
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
| | - Aurélie Malzert-Fréon
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France; (T.A.); (A.D.); (S.C.); (M.S.); (C.R.); (P.D.)
- Correspondence: (A.-C.G.); (A.M.-F.); Tel.: +33-231-566819 (A.M.-F.)
| |
Collapse
|
13
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
14
|
Alp G, Aydogan N. Lipid-based mucus penetrating nanoparticles and their biophysical interactions with pulmonary mucus layer. Eur J Pharm Biopharm 2020; 149:45-57. [PMID: 32014491 DOI: 10.1016/j.ejpb.2020.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/25/2019] [Accepted: 01/29/2020] [Indexed: 12/27/2022]
Abstract
Lungs are critical organs that are continuously exposed to exogeneous matter. The presence of the mucus layer helps to protect them via its adhesive structure and filtering mechanisms. Mucus also acts as a strong barrier against the drugs and nanocarriers in drug delivery. In this study, solid lipid nanoparticles (SLNs), at different sizes and surface properties, were prepared and their spreading/penetration ability was tested for their use in pulmonary drug delivery. The biophysical interactions of SLNs have been studied via light scattering (LS) and zeta potential analyses by incubating the SLNs in mucin solution and forming a model mucus layer using a Langmuir-Blodgett (LB) trough. In addition, the penetration performance of the particles was evaluated using Franz diffusion cell and rotating diffusion tubes. It was determined that 36% of SLNs can penetrate through a 1.2 ± 0.2-mm-thick mucus layer. Finally, the spreading behavior of the particles on a mucus-mimicking subphase was characterized and enhanced using a catanionic surfactant mixture. Overall, the current study was the first to investigates both the spreading and penetration performance of SLNs. The developed systems offer a drug delivery system that is able to achieve high penetration rates through a thick mucus layer.
Collapse
Affiliation(s)
- Gokce Alp
- Department of Chemical Engineering, Hacettepe University, Beytepe 06800, Ankara, Turkey
| | - Nihal Aydogan
- Department of Chemical Engineering, Hacettepe University, Beytepe 06800, Ankara, Turkey.
| |
Collapse
|
15
|
A novel microfluidic mucus-chip for studying the permeation of compounds over the mucus barrier. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
16
|
In vitro investigation on the impact of airway mucus on drug dissolution and absorption at the air-epithelium interface in the lungs. Eur J Pharm Biopharm 2019; 141:210-220. [DOI: 10.1016/j.ejpb.2019.05.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 01/07/2023]
|
17
|
O'Driscoll CM, Bernkop-Schnürch A, Friedl JD, Préat V, Jannin V. Oral delivery of non-viral nucleic acid-based therapeutics - do we have the guts for this? Eur J Pharm Sci 2019; 133:190-204. [PMID: 30946964 DOI: 10.1016/j.ejps.2019.03.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/22/2022]
Abstract
Gene therapy with RNA and pDNA-based drugs is limited by poor enzymatic stability and poor cellular permeation. The delivery of nucleic acids, in particular by the oral route, remains a major hurdle. This review will focus on the barriers to the oral delivery of nucleic acids and the strategies, in particular formulation strategies, which have been developed to overcome these barriers. Due to their very low oral bioavailability, the most obvious and most investigated biomedical applications for their oral delivery are related to the local treatment of inflammatory bowel diseases and colorectal cancers. Preclinical data but not yet clinical studies support the potential use of the oral route for the local delivery of formulated nucleic acid-based drugs.
Collapse
Affiliation(s)
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Julian D Friedl
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Véronique Préat
- Universite catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1.73.12, 1200 Brussels, Belgium.
| | - Vincent Jannin
- Gattefossé SAS, 36 chemin de Genas, 69804 Saint-Priest cedex, France.
| |
Collapse
|
18
|
Poinard B, Kamaluddin S, Tan AQQ, Neoh KG, Kah JCY. Polydopamine Coating Enhances Mucopenetration and Cell Uptake of Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4777-4789. [PMID: 30694045 DOI: 10.1021/acsami.8b18107] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Mucus is an endogenous viscoelastic biopolymer barrier that limits the entry of foreign pathogens and therapeutic carriers to the underlying mucosal cells. This could be overcome with a hydrophilic and nonpositively charged carrier surface that minimizes interactions with the mucin glycoprotein fibers. Although PEGylation remains an attractive surface strategy to enhance mucopenetration, cell uptake of PEGylated nanoparticles (NPs) often remains poor. Here, we demonstrated polydopamine (PDA) coating to enhance both mucopenetration and cell uptake of NPs. PDA was polymerized on carboxylated polystyrene (PS) NPs to form a PDA coating, and the resulting PS-PDA achieved a similar level of mucopenetration as our PEGylated PS (PS-PEG) positive control in three separate studies: NP-mucin interaction test, transwell assay, and multiple particle tracking. Compared to water, the diffusions of PS-PDA and PS-PEG in reconstituted mucus solution were only 3.5 and 2.4 times slower, respectively, whereas the diffusion of bare PS was slowed by up to 250 times. However, the uptake of PS-PDA (61.2 ± 6.1%) was almost three times higher than PS-PEG (24.6 ± 5.4%) in T24 cells, which were used as a model for underlying mucosal cells. Our results showed a novel unreported functionality of PDA coating in enhancing both mucopenetration and cell uptake of NPs for mucosal drug delivery applications, not possible with conventional PEGylation strategies.
Collapse
Affiliation(s)
- Barbara Poinard
- NUS Graduate School of Integrative Sciences and Engineering , National University of Singapore , 117456 Singapore
| | - Syafiqah Kamaluddin
- Department of Biomedical Engineering , National University of Singapore , 117583 Singapore
| | - Angeline Qiao Qi Tan
- School of Life Sciences & Chemical Technology , Ngee Ann Polytechnic , 599489 Singapore
| | - Koon Gee Neoh
- NUS Graduate School of Integrative Sciences and Engineering , National University of Singapore , 117456 Singapore
- Department of Chemical and Biomolecular Engineering , National University of Singapore , 117585 Singapore
| | - James Chen Yong Kah
- NUS Graduate School of Integrative Sciences and Engineering , National University of Singapore , 117456 Singapore
- Department of Biomedical Engineering , National University of Singapore , 117583 Singapore
| |
Collapse
|
19
|
Tran TT, Yu H, Vidaillac C, Lim AYH, Abisheganaden JA, Chotirmall SH, Hadinoto K. An evaluation of inhaled antibiotic liposome versus antibiotic nanoplex in controlling infection in bronchiectasis. Int J Pharm 2019; 559:382-392. [PMID: 30731256 DOI: 10.1016/j.ijpharm.2019.01.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/15/2019] [Accepted: 01/26/2019] [Indexed: 01/28/2023]
Abstract
Inhaled antibiotic nanoparticles have emerged as an effective strategy to control infection in bronchiectasis lung owed to their mucus-penetrating ability. Using ciprofloxacin (CIP) as the model antibiotic, we evaluated dry powder inhaler (DPI) formulations of two classes of antibiotic nanoparticles (i.e. liposome and nanoplex) in their (1) physical characteristics (i.e. size, zeta potential, CIP payload, preparation efficiency), (2) dissolution in artificial sputum medium, (3) ex vivo mucus permeability, (4) antimicrobial activity against Pseudomonas aeruginosa in mucus, (5) cytotoxicity towards human lung epithelium cells, and (6) in vitro aerosolization efficiency. The results showed that the CIP nanoplex exhibited fast dissolution with CIP supersaturation generation, in contrast to the slower release of the liposome (80 versus 30% dissolution after 1 h). Both nanoparticles readily overcame the mucus barrier attributed to their nanosize and mucus-inert surface (50% permeation after 1 h), leading to their similarly high antipseudomonal activity. The CIP liposome, however, possessed much lower CIP payload than the nanoplex (84% versus 3.5%), resulting in high lipid contents in its DPI formulation that led to higher cytotoxicity and lower aerosolization efficiency. The CIP nanoplex thus represented a superior formulation owed to its simpler preparation, higher CIP payload hence lower dosage, better aerosolization, and lower cytotoxicity.
Collapse
Affiliation(s)
- The-Thien Tran
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Hong Yu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Celine Vidaillac
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Albert Y H Lim
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - John A Abisheganaden
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Kunn Hadinoto
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore.
| |
Collapse
|
20
|
Li Y, Wan Z, Yang X. Salt reduction in liquid/semi-solid foods based on the mucopenetration ability of gum arabic. Food Funct 2019; 10:4090-4101. [DOI: 10.1039/c8fo02593b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gum arabic enhances the saltiness perception of liquid/semi-solid foods via a mucopenetration effect.
Collapse
Affiliation(s)
- Yanlei Li
- Laboratory of Food Proteins and Colloids
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety
- Department of Food Science and Technology
- South China University of Technology
- Guangzhou 510640
| | - Zhili Wan
- Laboratory of Food Proteins and Colloids
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety
- Department of Food Science and Technology
- South China University of Technology
- Guangzhou 510640
| | - Xiaoquan Yang
- Laboratory of Food Proteins and Colloids
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety
- Department of Food Science and Technology
- South China University of Technology
- Guangzhou 510640
| |
Collapse
|
21
|
Amara RO, Ramadan AA, El-Moslemany RM, Eissa MM, El-Azzouni MZ, El-Khordagui LK. Praziquantel-lipid nanocapsules: an oral nanotherapeutic with potential Schistosoma mansoni tegumental targeting. Int J Nanomedicine 2018; 13:4493-4505. [PMID: 30122922 PMCID: PMC6084080 DOI: 10.2147/ijn.s167285] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Lipid nanocapsules (LNCs) have shown potential to increase the bioavailability and efficacy of orally administered drugs. However, their intestinal translocation to distal target sites and their implication in pharmacokinetic (PK)–pharmacodynamic (PD) relationships are yet to be elucidated. In this study, the effect of LNCs on the PD activity and pharmacokinetics of praziquantel (PZQ), the mainstay of schistosomiasis chemotherapy, was investigated. Materials and methods The composition of LNCs was modified to increase PZQ payload and to enhance membrane permeability. PZQ–LNCs were characterized in vitro for colloidal properties, entrapment efficiency (EE%), and drug release. PD activity of the test formulations was assessed in Schistosoma mansoni-infected mice 7 days post-oral administration of a single 250 mg/kg oral dose. Pharmacokinetics of the test formulations and their stability in simulated gastrointestinal (GI) fluids were investigated to substantiate in vivo data. Results PZQ–LNCs exhibited good pharmaceutical attributes in terms of size (46–62 nm), polydispersity index (0.01–0.08), EE% (>95%), and sustained release profiles. Results indicated significant efficacy enhancement by reduction in worm burden, amelioration of liver pathology, and extensive damage to the fluke suckers and tegument. This was partly explained by PK data determined in rats. In addition, oral targeting of the worms was supported by the stability of PZQ–LNCs in simulated GI fluids and scanning electron microscopy (SEM) visualization of nanostructures on the tegument of worms recovered from mesenteric/hepatic veins. Cytotoxicity data indicated tolerability of PZQ–LNCs. Conclusion Data obtained provide evidence for the ability of oral LNCs to target distal post-absorption sites, leading to enhanced drug efficacy. From a practical standpoint, PZQ–LNCs could be suggested as a potential tolerable single lower dose oral nanomedicine for more effective PZQ mass chemotherapy.
Collapse
Affiliation(s)
- Rokaya O Amara
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt, .,Biotechnology Research Center, Tripoli, Libya
| | - Alyaa A Ramadan
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt,
| | - Riham M El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt,
| | - Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mervat Z El-Azzouni
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Labiba K El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt,
| |
Collapse
|
22
|
Vllasaliu D, Thanou M, Stolnik S, Fowler R. Recent advances in oral delivery of biologics: nanomedicine and physical modes of delivery. Expert Opin Drug Deliv 2018; 15:759-770. [PMID: 30033780 DOI: 10.1080/17425247.2018.1504017] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Research into oral delivery of biologics has a long and rich history but has not produced technologies used in the clinic. The area has evolved in terms of strategies to promote oral biologics delivery from early chemical absorption enhancers to nanomedicine to devices. Continued activity in this area is justifiable considering the remarkable proliferation of biologics. AREAS COVERED The article discusses some physiological barriers to oral delivery of biologics, with a special focus on less characterized barriers such as the basement membrane. Recent progress in oral delivery of biologics via nanomedicine is subsequently covered. Finally, the emerging field of device-mediated gastrointestinal delivery of biotherapeutics is discussed EXPERT OPINION Oral delivery of biologics is considered a 'panacea' in drug delivery. Almost century-old approaches of utilizing chemical absorption enhancers have not produced clinically translated technologies. Nanomedicine for oral biologics delivery has demonstrated potential, but the field is relatively new, and technologies have not progressed to the clinic. Device-mediated oral biologics delivery (e.g. ultrasound or microneedles) is in its infancy. However, this space is likely to intensify owing to advances in electronics and materials, as well as the challenges and history related to clinical translation of alternative approaches.
Collapse
Affiliation(s)
- Driton Vllasaliu
- a School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine , King's College London , London , United Kingdom
| | - Maya Thanou
- a School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine , King's College London , London , United Kingdom
| | - Snjezana Stolnik
- b Division of Drug Delivery and Tissue Engineering, Boots Science Building , University of Nottingham , Nottingham , United Kingdom
| | - Robyn Fowler
- c SuccinctChoice Medical Communications , London , United Kingdom
| |
Collapse
|
23
|
Lechanteur A, das Neves J, Sarmento B. The role of mucus in cell-based models used to screen mucosal drug delivery. Adv Drug Deliv Rev 2018; 124:50-63. [PMID: 28751201 DOI: 10.1016/j.addr.2017.07.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/12/2017] [Accepted: 07/22/2017] [Indexed: 12/23/2022]
Abstract
The increasing interest in developing tools to predict drug absorption through mucosal surfaces is fostering the establishment of epithelial cell-based models. Cell-based in vitro techniques for drug permeability assessment are less laborious, cheaper and address the concerns of using laboratory animals. Simultaneously, in vitro barrier models that thoroughly simulate human epithelia or mucosae may provide useful data to speed up the entrance of new drugs and new drug products into the clinics. Nevertheless, standard cell-based in vitro models that intend to reproduce epithelial surfaces often discard the role of mucus in influencing drug permeation/absorption. Biomimetic models of mucosae in which mucus production has been considered may not be able to fully reproduce the amount and architecture of mucus, resulting in biased characterization of permeability/absorption. In these cases, artificial mucus may be used to supplement cell-based models but still proper identification and quantification are required. In this review, considerations regarding the relevance of mucus in the development of cell-based epithelial and mucosal models mimicking the gastro-intestinal tract, the cervico-vaginal tract and the respiratory tract, and the impact of mucus on the permeability mechanisms are addressed. From simple epithelial monolayers to more complex 3D structures, the impact of the presence of mucus for the extrapolation to the in vivo scenario is critically analyzed. Finally, an overview is provided on several techniques and methods to characterize the mucus layer over cell-based barriers, in order to intimately reproduce human mucosal layer and thereby, improve in vitro/in vivo correlation.
Collapse
|
24
|
García-Díaz M, Birch D, Wan F, Nielsen HM. The role of mucus as an invisible cloak to transepithelial drug delivery by nanoparticles. Adv Drug Deliv Rev 2018; 124:107-124. [PMID: 29117511 DOI: 10.1016/j.addr.2017.11.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 01/05/2023]
Abstract
Mucosal administration of drugs and drug delivery systems has gained increasing interest. However, nanoparticles intended to protect and deliver drugs to epithelial surfaces require transport through the surface-lining mucus. Translation from bench to bedside is particularly challenging for mucosal administration since a variety of parameters will influence the specific barrier properties of the mucus including the luminal fluids, the microbiota, the mucus composition and clearance rate, and the condition of the underlying epithelia. Besides, after administration, nanoparticles interact with the mucosal components, forming a biomolecular corona that modulates their behavior and fate after mucosal administration. These interactions are greatly influenced by the nanoparticle properties, and therefore different designs and surface-engineering strategies have been proposed. Overall, it is essential to evaluate these biomolecule-nanoparticle interactions by complementary techniques using complex and relevant mucus barrier matrices.
Collapse
Affiliation(s)
- María García-Díaz
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Ditlev Birch
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Feng Wan
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
25
|
Leal J, Smyth HDC, Ghosh D. Physicochemical properties of mucus and their impact on transmucosal drug delivery. Int J Pharm 2017; 532:555-572. [PMID: 28917986 PMCID: PMC5744044 DOI: 10.1016/j.ijpharm.2017.09.018] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023]
Abstract
Mucus is a selective barrier to particles and molecules, preventing penetration to the epithelial surface of mucosal tissues. Significant advances in transmucosal drug delivery have recently been made and have emphasized that an understanding of the basic structure, viscoelastic properties, and interactions of mucus is of great value in the design of efficient drug delivery systems. Mucins, the primary non-aqueous component of mucus, are polymers carrying a complex and heterogeneous structure with domains that undergo a variety of molecular interactions, such as hydrophilic/hydrophobic, hydrogen bonds and electrostatic interactions. These properties are directly relevant to the numerous mucin-associated diseases, as well as delivering drugs across the mucus barrier. Therefore, in this review we discuss regional differences in mucus composition, mucus physicochemical properties, such as pore size, viscoelasticity, pH, and ionic strength. These factors are also discussed with respect to changes in mucus properties as a function of disease state. Collectively, the review seeks to provide a state of the art roadmap for researchers who must contend with this critical barrier to drug delivery.
Collapse
Affiliation(s)
- Jasmim Leal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave., Austin, TX 78712, USA
| | - Hugh D C Smyth
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave., Austin, TX 78712, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave., Austin, TX 78712, USA.
| |
Collapse
|
26
|
Schattling P, Taipaleenmäki E, Zhang Y, Städler B. A Polymer Chemistry Point of View on Mucoadhesion and Mucopenetration. Macromol Biosci 2017; 17. [PMID: 28675773 DOI: 10.1002/mabi.201700060] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/07/2017] [Indexed: 12/20/2022]
Abstract
Although oral is the preferred route of administration of pharmaceutical formulations, the long-standing challenge for medically active compounds to efficiently cross the mucus layer barrier limits its wider applicability. Efforts in nanomedicine to overcome this hurdle consider mucoadhesive and mucopenetrating drug carriers by selectively designing (macromolecular) building blocks. This review highlights and critically discusses recent strategies developed in this context including poly(ethylene glycol)-based modifications, cationic and thiolated polymers, as well as particles with high charge density, zeta-potential shifting ability, or mucolytic properties. The latest advances in ex vivo test platforms are also reviewed.
Collapse
Affiliation(s)
- Philipp Schattling
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav-Wieds Vej 14, 8000, Aarhus, Denmark
| | - Essi Taipaleenmäki
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav-Wieds Vej 14, 8000, Aarhus, Denmark
| | - Yan Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav-Wieds Vej 14, 8000, Aarhus, Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav-Wieds Vej 14, 8000, Aarhus, Denmark
| |
Collapse
|
27
|
Ciprofloxacin-loaded lipid-core nanocapsules as mucus penetrating drug delivery system intended for the treatment of bacterial infections in cystic fibrosis. Int J Pharm 2017; 527:92-102. [PMID: 28499793 DOI: 10.1016/j.ijpharm.2017.05.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 11/22/2022]
Abstract
Treatment of bacterial airway infections is essential for cystic fibrosis therapy. However, effectiveness of antibacterial treatment is limited as bacteria inside the mucus are protected from antibiotics and immune response. To overcome this biological barrier, ciprofloxacin was loaded into lipid-core nanocapsules (LNC) for high mucus permeability, sustained release and antibacterial activity. Ciprofloxacin-loaded LNC with a mean size of 180nm showed a by 50% increased drug permeation through mucus. In bacterial growth assays, the drug in the LNC had similar minimum inhibitory concentrations as the free drug in P. aeruginosa and S. aureus. Interestingly, formation of biofilm-like aggregates, which were observed for S. aureus treated with free ciprofloxacin, was avoided by exposure to LNC. With the combined advantages over the non-encapsulated drug, ciprofloxacin-loaded LNC represent a promising drug delivery system with the prospect of an improved antibiotic therapy in cystic fibrosis.
Collapse
|
28
|
Roger E, Gimel JC, Bensley C, Klymchenko AS, Benoit JP. Lipid nanocapsules maintain full integrity after crossing a human intestinal epithelium model. J Control Release 2017; 253:11-18. [PMID: 28274740 DOI: 10.1016/j.jconrel.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 01/02/2023]
Abstract
Lipid nanocapsules (LNCs) have demonstrated great potential for the oral delivery of drugs having very limited oral bioavailability (BCS class II, III and IV molecules). It has been shown previously that orally-administered LNCs can permeate through mucus, increase drug absorption by the epithelial tissue, and finally, increase drug bioavailability. However, even if transport mechanisms through mucus and the intestinal barrier have already been clarified, the preservation of particle integrity is still not known. The aim of the present work is to study in vitro the fate of LNCs after their transportation across an intestinal epithelium model (Caco-2 cell model). For this, two complementary techniques were employed: Förster Resonance Energy Transfer (FRET) and Nanoparticle Tracking Analysis (NTA). Results showed, after 2h, the presence of nanoparticles in the basolateral side of the cell layer and a measurable FRET signal. This provides very good evidence for the transcellular intact crossing of the nanocarriers.
Collapse
Affiliation(s)
- Emilie Roger
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, IBS- CHU, 4 rue Larrey, 49933 Angers, France.
| | - Jean-Christophe Gimel
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, IBS- CHU, 4 rue Larrey, 49933 Angers, France
| | - Conor Bensley
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, IBS- CHU, 4 rue Larrey, 49933 Angers, France
| | - Andrey S Klymchenko
- University of Strasbourg, CNRS UMR7213, Laboratoire de Biophotonique et Pharmacologie, 74 Route du Rhin, 67401 Illkirch, Cedex, France
| | - Jean-Pierre Benoit
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, IBS- CHU, 4 rue Larrey, 49933 Angers, France
| |
Collapse
|
29
|
Micro- and nano-carrier systems: The non-invasive and painless local administration strategies for disease therapy in mucosal tissues. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:153-171. [DOI: 10.1016/j.nano.2016.08.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/05/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
|
30
|
Groo AC, De Pascale M, Voisin-Chiret AS, Corvaisier S, Since M, Malzert-Fréon A. Comparison of 2 strategies to enhance pyridoclax solubility: Nanoemulsion delivery system versus salt synthesis. Eur J Pharm Sci 2016; 97:218-226. [PMID: 27916693 DOI: 10.1016/j.ejps.2016.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/07/2016] [Accepted: 11/27/2016] [Indexed: 11/19/2022]
Abstract
Pyridoclax is an original oligopyridine lead, very promising in treatment of chemoresistant cancers. However, from solubility measurement and permeability evaluation, it appeared that this compound can be considered as a BCS II drug, with a poor water solubility. To overcome this unfavorable property, two strategies were proposed and compared: pyridoclax di-hydrochloride salt synthesis and formulation of pyridoclax-loaded nanoemulsions (PNEs) efficiently performed by transposing the spontaneous emulsification process previously developed by our team. Whereas the salt improved the thermodynamic solubility of the drug by a factor 4, the apparent solubility of the encapsulated pyridoclax was 1000-fold higher. Their stability was assessed upon dilution in various complex biomimetic media relevant for oral administration (SGF, FaSSIF-V2, FeSSIF-V2) or for the intravenous route (PBS). The solubility of the salt was affected by the nature of the medium, indicating that it could precipitate after administration, negatively impacting its bioavailability and its efficiency in vivo. On the contrary, in all media, PNEs remained stable in terms of granulometric properties (determined by DLS), ζ-potential and encapsulation efficiency (measured by HPLC). Thus, such nanomedicines appear as a valuable option to perform preclinical studies on the promising pyridoclax.
Collapse
Affiliation(s)
- A-C Groo
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France
| | - M De Pascale
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France
| | - A-S Voisin-Chiret
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France.
| | - S Corvaisier
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France
| | - M Since
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France
| | - A Malzert-Fréon
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), F-14000 Caen, France.
| |
Collapse
|
31
|
Beloqui A, des Rieux A, Préat V. Mechanisms of transport of polymeric and lipidic nanoparticles across the intestinal barrier. Adv Drug Deliv Rev 2016; 106:242-255. [PMID: 27117710 DOI: 10.1016/j.addr.2016.04.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/27/2016] [Accepted: 04/16/2016] [Indexed: 01/02/2023]
Abstract
Unraveling the mechanisms of nanoparticle transport across the intestinal barrier is essential for designing more efficient nanoparticles for oral administration. The physicochemical parameters of the nanoparticles (e.g., size, surface charge, chemical composition) dictate nanoparticle fate across the intestinal barrier. This review aims to address the most important findings regarding polymeric and lipidic nanoparticle transport across the intestinal barrier, including the evaluation of critical physicochemical parameters of nanoparticles that affect nanocarrier interactions with the intestinal barrier.
Collapse
|
32
|
Malhaire H, Gimel JC, Roger E, Benoît JP, Lagarce F. How to design the surface of peptide-loaded nanoparticles for efficient oral bioavailability? Adv Drug Deliv Rev 2016; 106:320-336. [PMID: 27058155 DOI: 10.1016/j.addr.2016.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/17/2016] [Accepted: 03/28/2016] [Indexed: 01/22/2023]
Abstract
The oral administration of proteins is a current challenge to be faced in the field of therapeutics. There is currently much interest in nanocarriers since they can enhance oral bioavailability. For lack of a clear definition, the key characteristics of nanoparticles have been highlighted. Specific surface area is one of these characteristics and represents a huge source of energy that can be used to control the biological fate of the carrier. The review discusses nanocarrier stability, mucus interaction and absorption through the intestinal epithelium. The protein corona, which has raised interest over the last decade, is also discussed. The universal ideal surface is a myth and over-coated carriers are not a solution either. Besides, common excipients can be useful on several targets. The suitable design should rather take into account the composition, structure and behavior of unmodified nanomaterials.
Collapse
|
33
|
Lakkireddy HR, Urmann M, Besenius M, Werner U, Haack T, Brun P, Alié J, Illel B, Hortala L, Vogel R, Bazile D. Oral delivery of diabetes peptides - Comparing standard formulations incorporating functional excipients and nanotechnologies in the translational context. Adv Drug Deliv Rev 2016; 106:196-222. [PMID: 26964477 DOI: 10.1016/j.addr.2016.02.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/23/2016] [Accepted: 02/28/2016] [Indexed: 12/12/2022]
Abstract
While some orally delivered diabetes peptides are moving to late development with standard formulations incorporating functional excipients, the demonstration of the value of nanotechnology in clinic is still at an early stage. The goal of this review is to compare these two drug delivery approaches from a physico-chemical and a biopharmaceutical standpoint in an attempt to define how nanotechnology-based products can be differentiated from standard oral dosage forms for oral bioavailability of diabetes peptides. Points to consider in a translational approach are outlined to seize the opportunities offered by a better understanding of both the intestinal barrier and of nano-carriers designed for oral delivery.
Collapse
Affiliation(s)
- Harivardhan Reddy Lakkireddy
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Matthias Urmann
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Melissa Besenius
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Ulrich Werner
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Torsten Haack
- Diabetes Division, Sanofi Research and Development, Frankfurt, Germany
| | - Priscilla Brun
- Disposition Safety and Animal Research, Sanofi Research and Development, Montpellier, France
| | - Jean Alié
- Analytical Sciences, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Brigitte Illel
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Laurent Hortala
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Rachel Vogel
- Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Montpellier, France
| | - Didier Bazile
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Operations, Lead Generation and Candidate Realization, Sanofi Research and Development, Vitry-sur-Seine, France.
| |
Collapse
|
34
|
Biopolymeric Mucin and Synthetic Polymer Analogs: Their Structure, Function and Role in Biomedical Applications. Polymers (Basel) 2016; 8:polym8030071. [PMID: 30979166 PMCID: PMC6432556 DOI: 10.3390/polym8030071] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 12/17/2022] Open
Abstract
Mucin networks are viscoelastic fibrillar aggregates formed through the complex self-association of biopolymeric glycoprotein chains. The networks form a lubricious, hydrated protective shield along epithelial regions within the human body. The critical role played by mucin networks in impacting the transport properties of biofunctional molecules (e.g., biogenic molecules, probes, nanoparticles), and its effect on bioavailability are well described in the literature. An alternate perspective is provided in this paper, presenting mucin’s complex network structure, and its interdependent functional characteristics in human physiology. We highlight the recent advances that were achieved through the use of mucin in diverse areas of bioengineering applications (e.g., drug delivery, biomedical devices and tissue engineering). Mucin network formation is a highly complex process, driven by wide variety of molecular interactions, and the network possess structural and chemical variations, posing a great challenge to understand mucin’s bulk behavior. Through this review, the prospective potential of polymer based analogs to serve as mucin mimic is suggested. These analog systems, apart from functioning as an artificial model, reducing the current dependency on animal models, can aid in furthering our fundamental understanding of such complex structures.
Collapse
|
35
|
Eissa MM, El-Moslemany RM, Ramadan AA, Amer EI, El-Azzouni MZ, El-Khordagui LK. Miltefosine Lipid Nanocapsules for Single Dose Oral Treatment of Schistosomiasis Mansoni: A Preclinical Study. PLoS One 2015; 10:e0141788. [PMID: 26574746 PMCID: PMC4648507 DOI: 10.1371/journal.pone.0141788] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 10/13/2015] [Indexed: 01/09/2023] Open
Abstract
Miltefosine (MFS) is an alkylphosphocholine used for the local treatment of cutaneous metastases of breast cancer and oral therapy of visceral leishmaniasis. Recently, the drug was reported in in vitro and preclinical studies to exert significant activity against different developmental stages of schistosomiasis mansoni, a widespread chronic neglected tropical disease (NTD). This justified MFS repurposing as a potential antischistosomal drug. However, five consecutive daily 20 mg/kg doses were needed for the treatment of schistosomiasis mansoni in mice. The present study aims at enhancing MFS efficacy to allow for a single 20mg/kg oral dose therapy using a nanotechnological approach based on lipid nanocapsules (LNCs) as oral nanovectors. MFS was incorporated in LNCs both as membrane-active structural alkylphospholipid component and active antischistosomal agent. MFS-LNC formulations showed high entrapment efficiency (EE%), good colloidal properties, sustained release pattern and physical stability. Further, LNCs generally decreased MFS-induced erythrocyte hemolytic activity used as surrogate indicator of membrane activity. While MFS-free LNCs exerted no antischistosomal effect, statistically significant enhancement was observed with all MFS-LNC formulations. A maximum effect was achieved with MFS-LNCs incorporating CTAB as positive charge imparting agent or oleic acid as membrane permeabilizer. Reduction of worm load, ameliorated liver pathology and extensive damage of the worm tegument provided evidence for formulation-related efficacy enhancement. Non-compartmental analysis of pharmacokinetic data obtained in rats indicated independence of antischistosomal activity on systemic drug exposure, suggesting possible gut uptake of the stable LNCs and targeting of the fluke tegument which was verified by SEM. The study findings put forward MFS-LNCs as unique oral nanovectors combining the bioactivity of MFS and biopharmaceutical advantages of LNCs, allowing targeting via the oral route. From a clinical point of view, data suggest MFS-LNCs as a potential single dose oral nanomedicine for enhanced therapy of schistosomiasis mansoni and possibly other diseases.
Collapse
Affiliation(s)
- Maha M. Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Riham M. El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Alyaa A. Ramadan
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Eglal I. Amer
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mervat Z. El-Azzouni
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Labiba K. El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- * E-mail:
| |
Collapse
|
36
|
Grießinger J, Dünnhaupt S, Cattoz B, Griffiths P, Oh S, Gómez SBI, Wilcox M, Pearson J, Gumbleton M, Abdulkarim M, Pereira de Sousa I, Bernkop-Schnürch A. Methods to determine the interactions of micro- and nanoparticles with mucus. Eur J Pharm Biopharm 2015; 96:464-76. [DOI: 10.1016/j.ejpb.2015.01.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 02/01/2023]
|
37
|
Groo AC, Bossiere M, Trichard L, Legras P, Benoit JP, Lagarce F. In vivo evaluation of paclitaxel-loaded lipid nanocapsules after intravenous and oral administration on resistant tumor. Nanomedicine (Lond) 2015; 10:589-601. [DOI: 10.2217/nnm.14.124] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim & methods: The aim of the present work was to encapsulate paclitaxel (Ptx) in various lipid nanocapsules (LNCs) formulations and then to compare their pharmacokinetics and efficacy on a subcutaneous isograft model in rats. Results: Three different Ptx formulations were obtained. Drug payloads ranged from 1.32 to 3.62 mg Ptx/g of formulation. After oral administration the area under concentration–time curve was higher (p < 0.05) if Ptx was encapsulated, (1,2 Distearoyl-sn-glycero-3–phosphoethanolamine-N-[amino(PEG)] (DSPE-PEG-NH2)) LNCs displaying the highest area under concentration–time curve (p < 0.05). Efficacy was better than control for standard LNCs after oral administration (p < 0.05) and for (DSPE-PEG-NH2) LNCs after intravenous administration. Despite good absorption, (DSPE-PEG-NH2) LNCs failed to remain efficient after oral route. Conclusion: This study highlights the importance of efficacy studies paired to pharmacokinetic studies for nanomedicines.
Collapse
Affiliation(s)
- AC Groo
- INSERM U1066 MINT, Micro et Nanomédecines Biomimétiques, LUNAM Université, 4 Rue Larrey, 49033 Angers, CEDEX 09, France
- Ethypharm SA, Grand-Quevilly, Chemin de la Poudrière, 76120 Grand Quevilly, France
| | - M Bossiere
- INSERM U1066 MINT, Micro et Nanomédecines Biomimétiques, LUNAM Université, 4 Rue Larrey, 49033 Angers, CEDEX 09, France
| | - L Trichard
- Ethypharm SA, Grand-Quevilly, Chemin de la Poudrière, 76120 Grand Quevilly, France
| | - P Legras
- SCAHU, LUNAM Université, Pavillon Ollivier, UFR Sciences médicales, Rue Haute de Reculée, 49045 Angers, CEDEX 01, France
| | - JP Benoit
- INSERM U1066 MINT, Micro et Nanomédecines Biomimétiques, LUNAM Université, 4 Rue Larrey, 49033 Angers, CEDEX 09, France
- Pharmacy Department, Angers University Hospital, CHU Angers, 4 rue Larrey, 49033 Angers, CEDEX 09, France
| | - F Lagarce
- INSERM U1066 MINT, Micro et Nanomédecines Biomimétiques, LUNAM Université, 4 Rue Larrey, 49033 Angers, CEDEX 09, France
- Pharmacy Department, Angers University Hospital, CHU Angers, 4 rue Larrey, 49033 Angers, CEDEX 09, France
| |
Collapse
|
38
|
Boegh M, Nielsen HM. Mucus as a Barrier to Drug Delivery - Understanding and Mimicking the Barrier Properties. Basic Clin Pharmacol Toxicol 2014; 116:179-86. [DOI: 10.1111/bcpt.12342] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/08/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Marie Boegh
- Department of Pharmacy; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
39
|
Choi SG, Lee SE, Kang BS, Ng CL, Davaa E, Park JS. Thermosensitive and mucoadhesive sol-gel composites of paclitaxel/dimethyl-β-cyclodextrin for buccal delivery. PLoS One 2014; 9:e109090. [PMID: 25275485 PMCID: PMC4183572 DOI: 10.1371/journal.pone.0109090] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/07/2014] [Indexed: 11/18/2022] Open
Abstract
The purpose of this study was to develop a buccal paclitaxel delivery system using the thermosensitive polymer Pluronic F127 (PF127) and the mucoadhesive polymer polyethylene oxide (PEO). The anticancer agent paclitaxel is usually used to treat ovarian, breast, and non-small-cell lung cancer. To improve its aqueous solubility, paclitaxel was incorporated into an inclusion complex with (2,6-di-O-methyl)-β-cyclodextrin (DMβCD). The formation of the paclitaxel inclusion complex was evaluated using various techniques, including x-ray diffractometry (XRD), Fourier-transform infrared (FT-IR) spectrophotometry, differential scanning calorimetry (DSC), and scanning electron microscopy (SEM). Hydrogels were prepared using a cold method. Concentrations of 18, 20, and 23% (w/v) PF127 were dissolved in distilled water including paclitaxel and stored overnight in a refrigerator at 4 °C. PEO was added at concentrations of 0.1, 0.2, 0.4, 0.8, and 1% (w/v). Each formulation included paclitaxel (0.5 mg/mL). The sol-gel transition temperature of the hydrogels was measured using the tube-inverting method. Drug release from the hydrogels was measured using a Franz diffusion cell containing pH 7.4 phosphate-buffered solution (PBS) buffer at 37 °C. The cytotoxicity of each formulation was measured using the MTT assay with a human oral cancer cell (KB cell). The sol-gel transition temperature of the hydrogel decreased when PF127 was present and varied according to the presence of mucoadhesive polymers. The in vitro release was sustained and the release rate was slowed by the addition of the mucoadhesive polymer. The cytotoxicity of the blank formulation was low, although the drug-loaded hydrogel showed acceptable cytotoxicity. The results of our study suggest that the combination of a PF 127-based mucoadhesive hydrogel formulation and inclusion complexes improves the in vitro release and cytotoxic effect of paclitaxel.
Collapse
Affiliation(s)
- Soon Gil Choi
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Eun Lee
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Bong-Seok Kang
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Choon Lian Ng
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Enkhzaya Davaa
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Sook Park
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
40
|
Groo AC, Lagarce F. Mucus models to evaluate nanomedicines for diffusion. Drug Discov Today 2014; 19:1097-108. [DOI: 10.1016/j.drudis.2014.01.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/20/2013] [Accepted: 01/24/2014] [Indexed: 01/25/2023]
|