1
|
El-Housiny S, Fouad AG, El-Bakry R, Zaki RM, Afzal O, El-Ela FIA, Ghalwash MM. In Vitro and in vivo characterization of nasal pH-Responsive in-situ hydrogel of Candesartan-loaded invasomes as a potential stroke treatment. Drug Deliv Transl Res 2025; 15:1626-1645. [PMID: 39259459 DOI: 10.1007/s13346-024-01700-z] [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] [Accepted: 08/13/2024] [Indexed: 09/13/2024]
Abstract
Candesartan (CDN) is a useful anti-stroke medication because it lowers blood pressure, inflammation, oxidative stress, angiogenesis and apoptosis. However, CDN has limited efficacy due to its low solubility and poor bioavailability. This study set out to develop nasal pH-responsive in situ hydrogel of CDN-loaded invasomes a (PRHCLI) for enhancing CDN's release, penetration, bioavailability, and effectiveness as a possible treatment for stroke. Based on the results of the pre-formulation investigation, the optimum CLI formulation for intravasomal delivery of CDN was determined to be 3% of phospholipid, 0.16% of cholesterol, 3% of ethanol, and 1% of cineole. The optimum formulation significantly enhanced CDN permeation and release by 2.06-fold and 59.06%, respectively. The CLI formulation was added to a mixture of chitosan (0.67%w/v) and glyceryl monooleate (0.27%v/v) to develop PRHCLI. The PRHCLI formulation enhanced the release and permeation of CDN relative to free CDN by 2.15 and 2.76 folds, respectively. An experimental rat stroke model was utilized for in vivo studies to evaluate the bioavailability, effectiveness, and toxicity of the PRHCLI formulation. The nasal PRHCLI drops increased the CDN's bioavailability by 3.20-fold compared to oral free CDN. Increased grip strength and decreased flexion, spontaneous motor activity, and Morris Water Maze scores in comparison to oral free CDN showed that nasal PRHCLI drops have better anti-stroke activity. The toxicity evaluation revealed the safety of nasal PRHCLI. Hence, nasal PRHCLI drops may represent a promising avenue as a stroke therapy.
Collapse
Affiliation(s)
- Shaimaa El-Housiny
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Amr Gamal Fouad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
- Faculty of Pharmacy, Beni-Suef University, El-Shahid/Shehata Ahmed Hijaz St, Beni-Suef, Egypt.
| | - Rana El-Bakry
- Department of Pharmacology and Toxicology, EL Saleheya EL Gadida University, EL Saleheya El Gadida, Sharkia, Egypt
| | - Randa Mohammed Zaki
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, Saudi Arabia
| | - Fatma I Abo El-Ela
- Department of Pharmacology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Maha M Ghalwash
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
2
|
Papakyriakopoulou P, Valsami G. The nasal route for nose-to-brain drug delivery: advanced nasal formulations for CNS disorders. Expert Opin Drug Deliv 2025:1-17. [PMID: 40189901 DOI: 10.1080/17425247.2025.2489553] [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: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION The nasal route offers a feasible alternative to oral and/or parenteral administration, providing a noninvasive route to achieve nose-to-brain drug delivery involving the olfactory and trigeminal nerves, and facilitating local or systemic drug action. Conventional liquid nasal dosage forms have not managed to bridge the gaps of precise dosing and targeted central nervous system (CNS) delivery, while more sophisticated formulation approaches are being explored for brain targeting, aiming to enhance bioavailability and therapeutic efficacy. AREAS COVERED This review focuses on preclinical and clinical evaluation of microemulsions, in-situ gels, nasal powders, and nanocarrier-based formulations. Key pharmacokinetic and pharmacodynamic findings are discussed to evaluate their potential and limitations in improving drug bioavailability and CNS targeting. The existing regulatory framework for approval of products for nose-to-brain drug delivery is also addressed and relative hurdles are discussed. EXPERT OPINION While nasal drug delivery holds great promise for CNS therapeutics, key challenges remain, including formulation stability, mucosal permeability, patient adherence. Future research should prioritize improving targeting efficiency, overcoming mucociliary clearance, developing user-friendly pharmaceutical products. Personalized medicine and smart delivery systems could further enhance drug targeting and minimize side effects. Continued research and regulatory advancements are essential to fully realize nasal delivery's perspective in CNS therapeutics.
Collapse
Affiliation(s)
- Paraskevi Papakyriakopoulou
- Laboratory of Biopharmaceutics and Pharmacokinetics, Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - Georgia Valsami
- Laboratory of Biopharmaceutics and Pharmacokinetics, Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
3
|
Rajbanshi A, Hilton E, Atkinson E, Phillips JB, Vanukuru S, Khutoryanskiy VV, Gibbons A, Falloon S, Dreiss CA, Murnane D, Cook MT. Thermoresponsive engineered emulsions stabilised with branched copolymer surfactants for nasal drug delivery of molecular therapeutics. Int J Pharm 2025; 676:125506. [PMID: 40189166 DOI: 10.1016/j.ijpharm.2025.125506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Novel branched copolymer surfactants (BCS) allow the formation of oil-in-water emulsions that exhibit a temperature-induced liquid-to-gel transition. If the temperature of this transition is between room and body temperature (ca 25 and 37 °C, respectively), then the emulsions form a gel in situ upon contact with the body. A major advantage of this in situ gelation is the potential to manipulate the materials at room temperature in the low viscosity liquid state, then administer them to the body to initiate a switch to a retentive gel state, which could be used to deliver drugs to challenging sites such as the nasal mucosa. There are, however, several important factors which have not been explored for thermoresponsive BCS-stabilised emulsions to progress their use towards this application. Neither the delivery of drugs from the materials, the retention on tissue, nor the impact of co-formulated drugs on the thermoresponsive behaviours, are known. Furthermore, it has not been demonstrated that the materials are compatible with devices to generate sprays of the correct profiles for nasal administration. In this study we investigate the potential of thermoresponsive BCS-stabilised emulsions for the nasal delivery of licensed molecular therapeutics to examine the potential of BCS emulsion systems as a carrier for medicines. It was found that thermoresponsive behaviours can be maintained in the presence of drug substances, and that the liberation of the incorporated drugs occurs in a sustained manner. The BCS appear to have comparable cytotoxicity to common excipients and significantly enhanced retention on nasal tissue compared to even well-established mucoadhesives. The emulsions were incorporated into a spray device to demonstrate that the materials can be atomised with a plume appropriate for nasal administration prior to in situ gelation.
Collapse
Affiliation(s)
- Abhishek Rajbanshi
- School of Life and Medical Science, University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, UK; UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Eleanor Hilton
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Emily Atkinson
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - James B Phillips
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Shiva Vanukuru
- School of Life and Medical Science, University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, UK; School of Pharmacy, University of Reading, Reading, Berkshire RG6 6UR, UK
| | | | - Adam Gibbons
- Bespak, Bergen Way, King's Lynn, Norfolk PE30 2JJ, UK
| | | | - Cecile A Dreiss
- Institute of Pharmaceutical Science, King's College London, London SE1 8WA, UK
| | - Darragh Murnane
- School of Life and Medical Science, University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, UK
| | - Michael T Cook
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| |
Collapse
|
4
|
Khot S, Mahajan U, Jadhav A, Vaishampayan P, Bagul U, Gadhave D, Gorain B, Kokare C. Nose-to-brain delivery of sorafenib-loaded lipid-based poloxamer-carrageenan nanoemulgel: Formulation and therapeutic investigation in glioblastoma-induced orthotopic rat model. Int J Biol Macromol 2025; 309:142861. [PMID: 40188927 DOI: 10.1016/j.ijbiomac.2025.142861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Glioblastoma multiforme (GBM) has a poor clinical prognosis, where conventional treatment offers therapeutic limitations. Therefore, the current study introduces a first-of-its-kind sorafenib (SOR) nanoemulsion (SNE) loaded with poloxamer-carrageenan nanoemulgel (SPCNEG), a novel dual-functional and natural polymer-based payload system for effective intranasal chemotherapeutic administration. The nanoformulation was developed using carrageenan (a natural gelling agent), poloxamer (a mucoadhesive agent), glyceryl caprate as lipid, and Cremophor EL:PEG 400 blend as surfactant system. The improved biopharmaceutical attributes of developed formulations were confirmed from the release experiments, revealing augmentation in drug release from SNE (84.56 ± 3.78 %) and SPCNEG (68.62 ± 4.11 %) up to 3.41- and 8.12-fold compared to plain SOR. The ex vivo experiments showed a similar enhancement in drug permeation. Moreover, the SNE also showed superior performance on glioma cell lines, as indicated by lower IC50 (2.23 μg/mL) than plain SOR (16.61 μg/mL). The pharmacokinetic study revealed a 2.52- and 3.24-fold increase in SNE and SPCNEG brain concentration, respectively, compared to Soranib®. Additionally, a high correlation was also observed between in vitro drug release and in vivo absorption at prespecified time intervals for developed formulations. In conclusion, the current research promising and non-invasive alternative to existing interventions for enhanced brain targeting potential.
Collapse
Affiliation(s)
- Shubham Khot
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Unmesh Mahajan
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Amol Jadhav
- Institute of Applied Biological Research and Development, (IABRD), A division of Nirav Biosolutions Pvt Ltd, Aundh, Pune 411007, Maharashtra, India
| | | | - Uddhav Bagul
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Dnyandev Gadhave
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Chandrakant Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India.
| |
Collapse
|
5
|
Kuhn AJ, Outlaw VK, Marcink TC, Yu Z, Mears MC, Cajimat MN, Kreitler DF, Cleven PR, Mook JC, Bente DA, Porotto M, Gellman SH, Moscona A. Enhancing the solubility of SARS-CoV-2 inhibitors to increase future prospects for clinical development. J Virol 2025; 99:e0215924. [PMID: 39902960 PMCID: PMC11915835 DOI: 10.1128/jvi.02159-24] [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: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/06/2025] Open
Abstract
SARS-CoV-2 poses an ongoing threat to human health as variants continue to emerge. Several effective vaccines are available, but a diminishing number of Americans receive the updated vaccines (only 22% received the 2023 update). Public hesitancy towards vaccines and common occurrence of "breakthrough" infections (i.e., infections of vaccinated individuals) highlight the need for alternative methods to reduce viral transmission. SARS-CoV-2 enters cells by fusing its envelope with the target cell membrane in a process mediated by the viral spike protein, S. The S protein operates via a Class I fusion mechanism in which fusion between the viral envelope and host cell membrane is mediated by structural rearrangements of the S trimer. We previously reported lipopeptides derived from the C-terminal heptad repeat (HRC) domain of SARS-CoV-2 S that potently inhibit fusion by SARS-CoV-2, both in vitro and in vivo. These lipopeptides bear an attached cholesterol unit to anchor them in the membrane. Here, to improve prospects for experimental development and future clinical utility, we employed structure-guided design to incorporate charged residues at specific sites in the peptide to enhance aqueous solubility. This effort resulted in two new, potent lipopeptide inhibitors. IMPORTANCE Despite the existence of vaccines for SARS-CoV-2, the constant evolution of new variants and the occurrence of breakthrough infections highlight the need for new and effective antiviral approaches. We have shown that lipopeptides designed to bind a conserved region on the SARS-CoV-2 spike protein can effectively block viral entry into cells and thereby block infection. To support the feasibility of using this approach in humans, we re-designed these lipopeptides to be more soluble, using information about the structure of the spike protein interacting with the peptides to modify the peptide chain. The new peptides are effective against both SARS-CoV-2 and MERS. The lipopeptides described here could serve as treatment for people who are unvaccinated or who experience breakthrough infections, and the approach to increasing solubility can be applied in a broad spectrum approach to treating infections with emerging viruses.
Collapse
Affiliation(s)
- Ariel J Kuhn
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Victor K Outlaw
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Tara C Marcink
- Center for Host-Pathogen Interaction, Columbia University Medical Center, New York, New York, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Zhen Yu
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Megan C Mears
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Maria N Cajimat
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Dale F Kreitler
- Center for BioMolecular Structure, NSLS-II, Brookhaven National Laboratory, Upton, New York, USA
| | - Payton R Cleven
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Jee Ching Mook
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Dennis A Bente
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Experimental Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Matteo Porotto
- Center for Host-Pathogen Interaction, Columbia University Medical Center, New York, New York, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Samuel H Gellman
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Anne Moscona
- Center for Host-Pathogen Interaction, Columbia University Medical Center, New York, New York, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, New York, USA
- Department of Physiology & Cellular Biophysics, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
6
|
Meirinho SA, José de Abreu Marques Rodrigues M, Lourenço Alves G. Intranasal administration of antiseizure drugs using new formulation trends: one step closer to reach clinical trials. Expert Opin Drug Deliv 2025; 22:329-346. [PMID: 39826097 DOI: 10.1080/17425247.2025.2454476] [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: 11/08/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION Although there are numerous options for epilepsy treatment, its effective control continues unsatisfactory. Thus, search for alternative therapeutic options to improve the efficacy/safety binomial of drugs becomes very attractive to investigate. In this context, intranasal administration of antiseizure drugs formulated on state-of-the-art nanosystems can be a promising strategy. AREAS COVERED This work gives a comprehensive overview of different intranasal nanosystems for antiseizure drug administration developed and evaluated on preclinical studies over the last 10 years and published in 'PubMed' and 'Web of Science' databases. Additionally, it highlights their pharmaceutical critical quality attributes and in vivo pharmacological outputs that might infer possible results when transposing to clinical trials. EXPERT OPINION Research into optimized nanosystems encapsulating antiseizure drugs to enhance direct nose-to-brain delivery has increased over the last years. Particularly, the interest in formulating first- and second-generation antiseizure drugs in nanoparticles is here highlighted, having demonstrated its in vivo safety and improvement on pharmacokinetic and efficacy outputs. Still, none of them were brought to clinical trials. Thus, considering the existing barriers between preclinical and clinical trials, if supported by robust and targeted quality by design approaches, intranasal drug delivery can be presented as a valid and superior alternative for epilepsy treatment.
Collapse
Affiliation(s)
- Sara Alexandra Meirinho
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Márcio José de Abreu Marques Rodrigues
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- BRIDGES - Biotechnology Research, Innovation and Design for Health Products, Polytechnic Institute of Guarda, Guarda, Portugal
| | | |
Collapse
|
7
|
Wang E, Qi Z, Cao Y, Li R, Wu J, Tang R, Gao Y, Du R, Liu M. Gels as Promising Delivery Systems: Physicochemical Property Characterization and Recent Applications. Pharmaceutics 2025; 17:249. [PMID: 40006616 PMCID: PMC11858892 DOI: 10.3390/pharmaceutics17020249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Gels constitute a versatile class of materials with considerable potential for applications in both technical and medical domains. Physicochemical property characterization is a critical evaluation method for gels. Common characterization techniques include pH measurement, structural analysis, mechanical property assessment, rheological analysis, and phase transition studies, among others. While numerous research articles report characterization results, few reviews comprehensively summarize the appropriate numerical ranges for these properties. This lack of standardization complicates harmonized evaluation methods and hinders direct comparisons between different gels. To address this gap, it is essential to systematically investigate characterization methods and analyze data from the extensive body of literature on gels. In this review, we provide a comprehensive summary of general characterization methods and present a detailed analysis of gel characterization data to support future research and promote standardized evaluation protocols.
Collapse
Affiliation(s)
- Enzhao Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhaoying Qi
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuzhou Cao
- School of Science, National University of Singapore, Singapore 119077, Singapore;
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
| | - Jing Wu
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo 315100, China;
| | - Rongshuang Tang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Ruofei Du
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Minchen Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (E.W.); (Z.Q.); (R.L.); (R.T.)
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
8
|
Jang H, Kim N, Jin SG. Development of a Carvedilol-Loaded Solid Self-Nanoemulsifying System with Increased Solubility and Bioavailability Using Mesoporous Silica Nanoparticles. Int J Mol Sci 2025; 26:1592. [PMID: 40004060 PMCID: PMC11855893 DOI: 10.3390/ijms26041592] [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: 12/22/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
This study developed a solid self-nanoemulsifying drug delivery system (S-SNEDDS) to improve the oral bioavailability of poorly soluble carvedilol using mesoporous silica nanoparticles (MSNs). The liquid self-nanoemulsifying drug delivery system (L-SNEDDS) consisted of carvedilol, Peceol, Tween 80, and Labrasol in a weight ratio of 10:25:50:25. The liquid SNEDDS was suspended in MSN at various ratios and spray-dried to produce S-SNEDDS. The emulsion size, PDI, solubility, and dissolution of various ratios of MSN were evaluated to make the optimal S-SNEDDS. The optimal S-SNEDDS, manufactured using a ratio of MSN to L-SNEDDS 1000 at 500, formed a nanoemulsion and achieved efficient supersaturation compared to carvedilol alone, which significantly improved drug solubility (approximately 400 times), dissolution (approximately 5.7 times at 60 min), area under the curve (AUC) (21.7 times), and maximum plasma concentration (Cmax) (15.7 times). In addition, the physicochemical properties of the optimal S-SNEDDS were evaluated by differential scanning calorimetry (DSC), X-ray powder diffraction (XRD), Fourier transform infrared (FT-IR), particle size, and scanning electron microscopy (SEM) images. S-SNEDDS showed a smaller particle size than MSN alone, and the crystalline drug was transformed into an amorphous substance, resulting in encapsulation in MSN. These results suggest that MSN can be a novel biocompatible carrier contributing to a safer and more effective delivery system.
Collapse
Affiliation(s)
| | | | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea; (H.J.)
| |
Collapse
|
9
|
Komal K, Ghosh R, Sil D, Sharma R, Kumar S, Pandey P, Kumar M. Advancements in nose-to-brain drug targeting for Alzheimer's disease: a review of nanocarriers and clinical insights. Inflammopharmacology 2025; 33:605-626. [PMID: 39776027 DOI: 10.1007/s10787-024-01636-3] [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: 11/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is a type of neurodegenerative disease that describes cognitive decline and memory loss resulting in disability in movement, memory, speech etc. Which first affects the hippocampal and entorhinal cortex regions of brain. Pathogenesis of AD depends on Amyloid-β, hyper-phosphorylation of tau protein, mitochondrial dysfunction, cholinergic hypothesis and oxidative stress. In comparison with males, females are more prone to AD due to reduced estrogen level. Some of the FDA-approved drugs and their conventional formulations available in the market are discussed in this review. Nose-to-brain delivery system provides the target specific drug delivery via olfactory and trigeminal nerve (active and passive drug targeting strategies) and bypassing the Blood Brain Barrier. Mucoadhesive agents and permeation enhancers are mostly utilized to enhance the retention time and bioavailability of the drugs. Liposomes, niosomes, cubosomes, solid lipid nanoparticles, nanoemulsions, micelles, and many more nanocarriers for nose-to-brain delivery of drugs are also described thoroughly in this review. It also covers the clinical trials and patents for nose-to-brain delivery. In this article, we investigate the nose-to-brain pathways for AD treatment strategies.
Collapse
Affiliation(s)
- Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Debayan Sil
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Prachi Pandey
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
10
|
Gama F, Meirinho S, Pires PC, Tinoco J, Martins Gaspar MC, Baltazar G, Alves G, Santos AO. Simvastatin is delivered to the brain by high-strength intranasal cationic SMEDDS and nanoemulsions. Drug Deliv Transl Res 2025:10.1007/s13346-024-01769-6. [PMID: 39747745 DOI: 10.1007/s13346-024-01769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/04/2025]
Abstract
The repurposing of statins as neuroprotective agents and/or anti-brain tumor drugs is limited by challenges in brain bioavailability and systemic off-target effects. Therefore, improved and targeted delivery of statins to the brain is necessary. This study aimed to develop a high-strength liquid formulation of the poorly soluble prodrug simvastatin for intranasal administration, as a strategy to achieve high brain concentrations of the prodrug and/or its active form, tenivastatin. Cationic simvastatin nanoemulsions (c-NE) and self-microemulsifying drug delivery systems (c-SMEDDS) were screened for composition, extensively characterized, and the viscosity of the nanoemulsion was further optimized. The optimized c-NE and c-SMEDDS formulations achieved high drug strengths, approximately 5.5% and 9% (w/w), respectively. They formed highly homogeneous aqueous dispersions (polydispersity index < 0.1) with small droplet sizes (< 120 nm and ~ 25 nm, respectively) and remained stable for at least four months under refrigeration. Neither the c-NE nor the c-SMEDDS induced hemolysis up to concentrations of 40 µg/mL and 450 µg/mL of simvastatin, respectively. The zero-shear viscosity of the c-NE was increased to 186 mPa·s by incorporating 0.25% (w/w) polyvinylpyrrolidone, which approached the viscosity of the c-SMEDDS (~ 126 mPa·s). Following intranasal administration of the optimized formulations to Wistar rats at a dose of 10 mg/kg, simvastatin levels in the brain reached 50 to 150 ng/g between 15 and 60 min post-administration. These findings indicate that the developed c-NE and c-SMEDDS formulations hold promise for simvastatin intranasal delivery and brain targeting, potentially paving the way for the realization of simvastatin's neuroprotective potential.
Collapse
Affiliation(s)
- Francisco Gama
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Sara Meirinho
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Patrícia C Pires
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
- Group of Pharmaceutical Technology, Faculty of Pharmacy, REQUIMTE/LAQV, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Johann Tinoco
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Maria Carolina Martins Gaspar
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Graça Baltazar
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Gilberto Alves
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Adriana O Santos
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
| |
Collapse
|
11
|
Jabir SA, Rajab NA. Preparation, In-vitro, Ex-vivo, and Pharmacokinetic Study of Lasmiditan as Intranasal Nanoemulsion-based In Situ Gel. Pharm Nanotechnol 2025; 13:239-253. [PMID: 38173066 DOI: 10.2174/0122117385285009231222072303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Lasmiditan (LAS) is a recently developed antimigraine drug and was approved in October, 2019 for the treatment of acute migraines; however, it suffers from low oral bioavailability, which is around 40%. OBJECTIVES This study aimed to improve the LAS bioavailability via formulation as nanoemulsionbased in situ gel (NEIG) given intranasally and then compare the traditional aqueous-LASsuspension (AQS) with the two successful intranasal prepared formulations (NEIG 2 and NEIG 5) in order to determine its relative bioavailability (F-relative) via using rabbits. METHODS Two successfully prepared nanoemulsion (NE) formulas, a and b, were selected for the incorporation of different percentages of pH-sensitive in situ gelling polymer (Carbopol 934) to prepare NEIGs 1, 2, 3, 4, 5, and 6. The pH, gelation capacity, gel strength, and viscosity were predicted for the prepared NEIGs. The release (in vitro) and the nasal permeation (ex vivo) were determined for NEIG 2 and 5, and then both were subjected to pharmacokinetics in vivo studies. Eighteen male rabbits weighing 2.0 to 2.5 kg were employed in the parallel design study. The body surface area (BSA) normalization method was applied for LAS dose calculation. Serial blood samples were taken out and subjected to drug analysis using the HPLC method previously developed and validated by Kumar et al. Primary pharmacokinetics parameters, including maximum drug concentration in plasma (Cmax), time to reach C-max (T-max), and area under the concentration-time curve from time zero to affinity (AUCt0-∞) were calculated. Both NE (a and b), together with NEIG (2 and 5) formulas, were subjected to the stability study. Finally, a nasal ciliotoxicity study was carried out to evaluate the nasal toxicity of developed NEIGs 2 and 5. RESULTS The results showed that NEIGs 2 and 5 could be selected as the optimized NEIGs as both achieved 100% permeation within 20 min and then released within 25 and 35 min, respectively, thus achieving 3.3 folds with higher permeation percentages as compared to the AQS. Both NEIGs 2 and 5 exerted comparable release and permeation values as the corresponding NE a and b with more residence time in order to overcome the normal nasal physiological clearance. The values of C-max, Tmax, and AUC0- ∞ for NEIG 2 and NEIG 5 were 8066 ± 242 ng/ml, 0.75 ± 0.05 h, 19616.86 ± 589 ng. h/ml, and 7975.67 ± 239 ng/ml, 1.0 ± 0.05 h, 17912.36 ± 537 ng. h/ml, respectively, compared to the traditional AQS, which is equal to 4181.09 ± 125 ng/ml, 2 ± 0.2 h, and 8852.27 ± 266 ng. h/ml, respectively. It was discovered that NEIGs 2 and 5 had better intranasal delivery of LAS and could significantly (p < 0.05) achieve a higher value of permeability coefficient (3.3 folds) and 2.5 folds improvement in bioavailability when compared to AQS. The NE a, NE b, NEIG2, and NEIG5 formulations showed good stability at various temperatures. According to the nasal ciliotoxicity study, the nasal mucosal membrane, which was treated with NEIG 5, showed irritation with a bit of damage. However, damage was not observed when it was treated with NEIG 2, indicating the biocompatibility of the last one to be selected as the optimum formula. CONCLUSION NEIG 2 and NEIG 5 are promising new intranasal formulas with a faster onset of action and greater bioavailability than the oral dosage form (AQS). Finally, the selected optimum gold formula that will be ready for further clinical study is NEIG 2.
Collapse
Affiliation(s)
- Saba Abdulhadi Jabir
- Department of Pharmaceutics, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | - Nawal A Rajab
- Department of Pharmaceutics, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
12
|
Nyavanandi D, Mandati P, Vidiyala N, Parupathi P, Kolimi P, Mamidi HK. Enhancing Patient-Centric Drug Development: Coupling Hot Melt Extrusion with Fused Deposition Modeling and Pressure-Assisted Microsyringe Additive Manufacturing Platforms with Quality by Design. Pharmaceutics 2024; 17:14. [PMID: 39861666 PMCID: PMC11769097 DOI: 10.3390/pharmaceutics17010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
In recent years, with the increasing patient population, the need for complex and patient-centric medications has increased enormously. Traditional manufacturing techniques such as direct blending, high shear granulation, and dry granulation can be used to develop simple solid oral medications. However, it is well known that "one size fits all" is not true for pharmaceutical medicines. Depending on the age, sex, and disease state, each patient might need a different dose, combination of medicines, and drug release pattern from the medications. By employing traditional practices, developing patient-centric medications remains challenging and unaddressed. Over the last few years, much research has been conducted exploring various additive manufacturing techniques for developing on-demand, complex, and patient-centric medications. Among all the techniques, nozzle-based additive manufacturing platforms such as pressure-assisted microsyringe (PAM) and fused deposition modeling (FDM) have been investigated thoroughly to develop various medications. Both nozzle-based techniques involve the application of thermal energy. However, PAM can also be operated under ambient conditions to process semi-solid materials. Nozzle-based techniques can also be paired with the hot melt extrusion (HME) process for establishing a continuous manufacturing platform by employing various in-line process analytical technology (PAT) tools for monitoring critical process parameters (CPPs) and critical material attributes (CMAs) for delivering safe, efficacious, and quality medications to the patient population without compromising critical quality attributes (CQAs). This review covers an in-depth discussion of various critical parameters and their influence on product quality, along with a note on the continuous manufacturing process, quality by design, and future perspectives.
Collapse
Affiliation(s)
- Dinesh Nyavanandi
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, MA 02141, USA;
| | - Preethi Mandati
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA; (P.M.); (P.K.)
| | - Nithin Vidiyala
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, MA 02141, USA;
| | - Prashanth Parupathi
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA;
| | - Praveen Kolimi
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA; (P.M.); (P.K.)
| | | |
Collapse
|
13
|
Maisto N, Mango D. Nose to brain strategy coupled to nano vesicular system for natural products delivery: Focus on synaptic plasticity in Alzheimer's disease. J Pharm Anal 2024; 14:101057. [PMID: 39802402 PMCID: PMC11718335 DOI: 10.1016/j.jpha.2024.101057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/28/2024] [Accepted: 07/30/2024] [Indexed: 01/16/2025] Open
Abstract
A wide number of natural molecules demonstrated neuroprotective effects on synaptic plasticity defects induced by amyloid-β (Aβ) in ex vivo and in vivo Alzheimer's disease (AD) models, suggesting a possible use in the treatment of this neurodegenerative disorder. However, several compounds, administered parenterally and orally, are unable to reach the brain due to the presence of the blood-brain barrier (BBB) which prevents the passage of external substances, such as proteins, peptides, or phytocompounds, representing a limit to the development of treatment for neurodegenerative diseases, such as AD. The combination of nano vesicular systems, as colloidal systems, and nose to brain (NtB) delivery depicts a new nanotechnological strategy to overtake this limit and to develop new treatment approaches for brain diseases, including the use of natural molecules in combination therapy for AD. Herein, we will provide an updated overview, examining the literature of the last 20 years and using specific keywords that provide evidence on natural products with the ability to restore synaptic plasticity alterations in AD models, and the possible application using safe and non-invasive strategies focusing on nano vesicular systems for NtB delivery.
Collapse
Affiliation(s)
- Nunzia Maisto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, 00185, Italy
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, 00161, Italy
| | - Dalila Mango
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, 00161, Italy
- School of Pharmacy, Department of Biology, University of Rome Tor Vergata, Rome, 00133, Italy
| |
Collapse
|
14
|
Idoudi S, Saleh A, Akkbik M, Amine L, Alansari K, Rachid O, Alkilany AM. Investigating Strategies to Enhance the Aqueous Solubility of Ketamine HCl for Intranasal Delivery. Pharmaceutics 2024; 16:1502. [PMID: 39771482 PMCID: PMC11677332 DOI: 10.3390/pharmaceutics16121502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Ketamine HCl, an FDA-approved therapeutic, is administered through various routes, including intranasal delivery. Administering an adequate therapeutic dose of intranasal ketamine HCl is challenging due to the limited volume that can be delivered intranasally given the current commercially available concentrations. Objectives: This study investigates solubilizing strategies to enhance the aqueous solubility of ketamine HCl for intranasal administration. Methods: We assessed the solubility profile of ketamine HCl by evaluating factors such as pH, co-solvents, and surfactants. Additionally, we developed and validated a UV-Vis spectroscopy method for ketamine HCl analysis. Results: Our solubility screening in various organic co-solvents revealed the following order of effectiveness in enhancing solubility: methanol > water > propylene glycol > ethanol > dimethyl sulfoxide (DMSO) > N-methyl-2-pyrrolidone (NMP). Despite methanol's superior solubility, its potential toxicity, coupled with the relatively lower effectiveness of other solvents compared to water, suggests that a co-solvency approach is not advantageous for ketamine HCl. We found that ketamine HCl solubility increased with medium acidity, with pH 3.5 being the optimal for further formulation studies. The impact of pharmaceutical surfactants on ketamine HCl solubility at an acidic pH was also evaluated. Surfactants tested included SDS, PEG 400, PVP, Tween 20, poloxamer 188, and lecithin. Notably, PEG 400 and PVP reduced solubility due to a salting-out effect, whereas Tween 80, lecithin, and poloxamer 188 slightly improved solubility through micelle formation. Among the surfactants tested, 1% SDS emerged as the most effective in enhancing ketamine HCl solubility. Conclusions: These outcomes highlight the potential of these solubilization strategies to address the solubility limitations of ketamine HCl, enabling the preparation of highly concentrated ketamine HCl formulations for intranasal delivery.
Collapse
Affiliation(s)
- Sourour Idoudi
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha P.O. Box 2713, Qatar; (S.I.); (M.A.); (O.R.)
| | - Alaaeldin Saleh
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Mohammed Akkbik
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha P.O. Box 2713, Qatar; (S.I.); (M.A.); (O.R.)
- Central Laboratories Unit, Office of VP for Research & Graduate Studies, Qatar University, Doha P.O. Box 2713, Qatar
| | - Leena Amine
- Department of Emergency, Sidra Medicine, Doha P.O. Box 26999, Qatar; (L.A.); (K.A.)
| | - Khalid Alansari
- Department of Emergency, Sidra Medicine, Doha P.O. Box 26999, Qatar; (L.A.); (K.A.)
| | - Ousama Rachid
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha P.O. Box 2713, Qatar; (S.I.); (M.A.); (O.R.)
| | - Alaaldin M. Alkilany
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha P.O. Box 2713, Qatar; (S.I.); (M.A.); (O.R.)
| |
Collapse
|
15
|
Corazza E, Pizzi A, Parolin C, Giordani B, Abruzzo A, Bigucci F, Cerchiara T, Luppi B, Vitali B. Orange Peel Lactiplantibacillus plantarum: Development of A Mucoadhesive Nasal Spray with Antimicrobial and Anti-inflammatory Activity. Pharmaceutics 2024; 16:1470. [PMID: 39598593 PMCID: PMC11597421 DOI: 10.3390/pharmaceutics16111470] [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: 10/30/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Due to the high frequency and severity of upper respiratory bacterial infections, probiotics could offer a new medical approach. We explored the antibacterial and anti-inflammatory properties of the new strain Lactiplantibacillus plantarum BIA and formulated a nasal spray. Methods:L. plantarum BIA was isolated from orange peel and taxonomically identified through 16S rRNA gene sequencing. Its antibacterial activity was tested against Pseudomonas aeruginosa, Streptococcus pyogenes, Bacillus subtilis, Escherichia coli, and Staphylococcus aureus, while anti-inflammatory potential was evaluated by Griess assay. BIA genome was fully sequenced and analyzed to assess its safety. BIA was formulated in a freeze-dried matrix, containing prebiotics and cryoprotectants, to be reconstituted with a polymer solution. Solutions containing two types of hydroxypropyl methylcellulose (HPMC) and hyaluronic acid were evaluated as resuspending media and compared in terms of pH, viscosity, and mucoadhesion ability. The biological activity of BIA formulated as nasal spray was verified together with the stability of the selected formulations. Results:L. plantarum BIA inhibited human pathogens' growth and showed anti-inflammatory activity and a safe profile. In the best-performing formulation, the probiotic is lyophilized in 10% fructooligosaccharides, 0.1% ascorbic acid, and 0.5% lactose and reconstituted with HPMC high viscosity 1% w/v. This composition ensured the probiotic's viability for up to six months in its dried form and one week after reconstitution. It also allowed interaction with the nasal mucosa, preserving its antimicrobial and anti-inflammatory activities. Conclusion: The developed nasal spray could become a promising formulation in the field of nasal infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Elisa Corazza
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Asia Pizzi
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Carola Parolin
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Barbara Giordani
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| | - Angela Abruzzo
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Federica Bigucci
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Teresa Cerchiara
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Barbara Luppi
- Drug Delivery Research Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (E.C.); (A.A.); (F.B.); (T.C.)
| | - Beatrice Vitali
- Beneficial Microbes Laboratory, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (A.P.); (C.P.); (B.G.); (B.V.)
| |
Collapse
|
16
|
Farouk HO, Nagib MM, Fouad AG, Naguib DM, Khalil SFA, Belal A, Miski SF, Albezrah NKA, Al-Ziyadi SH, Kim GH, Hassan AHE, Lee KT, Hamad DS. Fabrication of an In Situ pH-Responsive Raloxifene-Loaded Invasome Hydrogel for Breast Cancer Management: In Vitro and In Vivo Evaluation. Pharmaceuticals (Basel) 2024; 17:1518. [PMID: 39598429 PMCID: PMC11597612 DOI: 10.3390/ph17111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Raloxifene (RLF) is a therapeutic option for invasive breast cancer because it blocks estrogen receptors selectively. Low solubility, limited targeting, first-pass action, and poor absorption are some of the challenges that make RLF in oral form less effective. This study aimed to create an intra-tumoral in situ pH-responsive formulation of RLF-invasome (IPHRLI) for breast cancer treatment, with the goals of sustaining RLF release, minimizing adverse effects, and enhancing solubility, bioavailability, targeting, and effectiveness. METHODS Numerous RLF-invasome formulations were optimized using design expert software (version 12.0.6.0, StatEase Inc., Minneapolis, MN, USA). Integrating an optimal formulation with an amalgam of chitosan and glyceryl monooleate resulted in the IPHRLI formulation. In vivo testing of the IPHRLI formulation was conducted utilizing the Ehrlich cancer model. RESULTS Requirements for an optimum RLF-invasome formulation were met by a mixture of phospholipids (2.46%), ethanol (2.84%), and cineole (0.5%). The IPHRLI formulation substantially sustained its release by 75.41% after 8 h relative to free RLF. The bioavailability of intra-tumoral IPHRLI was substantially raised by 4.07-fold compared to oral free RLF. Histopathological and tumor volume analyses of intra-tumoral IPHRLI confirmed its efficacy and targeting effect. CONCLUSIONS the intra-tumoral administration of the IPHRLI formulation may provide a potential strategy for breast cancer management.
Collapse
Affiliation(s)
- Hanan O. Farouk
- Department of Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni-Suef 62511, Egypt; (H.O.F.); (D.M.N.)
| | - Marwa M. Nagib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Cairo 11435, Egypt;
| | - Amr Gamal Fouad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Demiana M. Naguib
- Department of Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni-Suef 62511, Egypt; (H.O.F.); (D.M.N.)
| | | | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Samar F. Miski
- Pharmacology and Toxicology Department, College of Pharmacy, Taibah University, Medina 42278, Saudi Arabia;
| | - Nisreen Khalid Aref Albezrah
- Department of Obstetric & Gynecology, College of Medicine, Taif University, Taif 21944, Saudi Arabia; (N.K.A.A.); (S.H.A.-Z.)
| | - Shatha Hallal Al-Ziyadi
- Department of Obstetric & Gynecology, College of Medicine, Taif University, Taif 21944, Saudi Arabia; (N.K.A.A.); (S.H.A.-Z.)
| | - Gi-Hui Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ahmed H. E. Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Doaa S. Hamad
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Nile Valley University, Fayoum 63518, Egypt;
| |
Collapse
|
17
|
Wang H, Gong F, Liu J, Xiang L, Hu Y, Che W, Li R, Yang S, Zhuang Q, Teng X. Engineering Docetaxel Micelles for Enhanced Cancer Therapy Through Intermolecular Forces. Bioengineering (Basel) 2024; 11:1078. [PMID: 39593738 PMCID: PMC11591415 DOI: 10.3390/bioengineering11111078] [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: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Docetaxel has exhibited excellent therapeutic effects in cancer treatment; however, its hydrophobicity, short blood circulation time, and high blood toxicity restrict its clinical application. The use of mPEG-PLA micelles to deliver docetaxel into the body has been verified as an effective approach to enhance its therapeutic efficacy. However, mPEG-PLA micelles are easily disassembled in the bloodstream, which can easily lead to premature drug release. To broaden the application scenarios of mPEG PLA micelles, we utilized the π-π stacking effect as an intermolecular force to design a novel mPEG-PLA-Lys(Fmoc) micelle to enhance the blood stability and permeability of drug-loaded micelles. The result showed that drug-loaded micelles for injection did not alter the tissue selectivity of docetaxel. Intravenous injection of the micelles in nude mice showed better antitumor efficacy than docetaxel injection and tumor recurrence rate is 0%, which is significantly lower than that of docetaxel injection (100%). The micelles designed by this research institute are anticipated to improve the clinical therapeutic effect of docetaxel.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xin Teng
- National Material Experimental Teaching Demonstration Center, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China (Q.Z.)
| |
Collapse
|
18
|
Torres J, Silva R, Farias G, Sousa Lobo JM, Ferreira DC, Silva AC. Enhancing Acute Migraine Treatment: Exploring Solid Lipid Nanoparticles and Nanostructured Lipid Carriers for the Nose-to-Brain Route. Pharmaceutics 2024; 16:1297. [PMID: 39458626 PMCID: PMC11510892 DOI: 10.3390/pharmaceutics16101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Migraine has a high prevalence worldwide and is one of the main disabling neurological diseases in individuals under the age of 50. In general, treatment includes the use of oral analgesics or non-steroidal anti-inflammatory drugs (NSAIDs) for mild attacks, and, for moderate or severe attacks, triptans or 5-HT1B/1D receptor agonists. However, the administration of antimigraine drugs in conventional oral pharmaceutical dosage forms is a challenge, since many molecules have difficulty crossing the blood-brain barrier (BBB) to reach the brain, which leads to bioavailability problems. Efforts have been made to find alternative delivery systems and/or routes for antimigraine drugs. In vivo studies have shown that it is possible to administer drugs directly into the brain via the intranasal (IN) or the nose-to-brain route, thus avoiding the need for the molecules to cross the BBB. In this field, the use of lipid nanoparticles, in particular solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC), has shown promising results, since they have several advantages for drugs administered via the IN route, including increased absorption and reduced enzymatic degradation, improving bioavailability. Furthermore, SLN and NLC are capable of co-encapsulating drugs, promoting their simultaneous delivery to the site of therapeutic action, which can be a promising approach for the acute migraine treatment. This review highlights the potential of using SLN and NLC to improve the treatment of acute migraine via the nose-to-brain route. First sections describe the pathophysiology and the currently available pharmacological treatment for acute migraine, followed by an outline of the mechanisms underlying the nose-to-brain route. Afterwards, the main features of SLN and NLC and the most recent in vivo studies investigating the use of these nanoparticles for the treatment of acute migraine are presented.
Collapse
Affiliation(s)
- Joana Torres
- UCIBIO, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | | | - José Manuel Sousa Lobo
- UCIBIO, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Domingos Carvalho Ferreira
- UCIBIO, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Catarina Silva
- UCIBIO, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- FP-BHS (Biomedical and Health Sciences Research Unit), FP-I3ID (Instituto de Investigação, Inovação e Desenvolvimento), Faculty of Health Sciences, University Fernando Pessoa, 4200-150 Porto, Portugal
| |
Collapse
|
19
|
Papakyriakopoulou P, Balafas E, Kostomitsopoulos N, Rekkas DM, Dev KK, Valsami G. Pharmacokinetic Study of Fingolimod Nasal Films Administered via Nose-to-Brain Route in C57BL/6 J Mice as Potential Treatment for Multiple Sclerosis. Pharm Res 2024; 41:1951-1963. [PMID: 39470941 DOI: 10.1007/s11095-024-03745-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/09/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Fingolimod hydrochloride (FH) has emerged as a vital medication for managing Multiple Sclerosis (MS). Despite its high oral bioavailability of 93%, it is plagued by slow oral absorption (Tmax = 8-12 h) and extensive hepatic metabolism. Intranasal administration has emerged as an alternative to address these limitations, ensuring efficient central nervous system delivery and minimizing peripheral exposure and first-pass metabolism. OBJECTIVE This study aims to develop and evaluate FH nasal films for enhanced drug delivery. METHODS A Design of Experiments approach was employed to formulate FH nasal films, utilizing HPMC E50 as a film-forming polymer, PEG 400 as a plasticizer, and Me-β-CD as a permeation enhancer. Two formulations with superior in vitro and ex vivo performance were selected for in vivo evaluation. A comparative pharmacokinetic study was conducted in C57BL/6 J mice in the brain and serum after administration of nasal films and oral FH solution, respectively. Sparse sampling and non-compartmental analysis were used. RESULTS FH nasal films efficiently delivered the drug to both serum (Cmax(F3) = 0.35 ± 0.021, Cmax(F4) = 0.38 ± 0.029 μg/mL) and brain (Cmax(F3) = 0.39 ± 0.05, Cmax(F4) = 0.44 ± 0.048 μg/mL), achieving higher levels than oral delivery. Brain relative bioavailability (% Frel (0-6 h)) was 519% and 534%, while serum % Frel (0-6 h) was 295% and 343%. CONCLUSIONS The rapid nose-to-brain delivery within 30 min, in contrast to 10-h Tmax of the oral solution, indicates the potential of a combined IN and oral treatment regimen. This approach could expedite the attainment of steady-state concentrations, offering a promising method for managing multiple sclerosis (MS).
Collapse
Affiliation(s)
- Paraskevi Papakyriakopoulou
- Department of Pharmacy, School of Health Sciences Sector of Pharmaceutical Technology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Evangelos Balafas
- Laboratory Animal Facility, Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Nikolaos Kostomitsopoulos
- Laboratory Animal Facility, Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Dimitrios M Rekkas
- Department of Pharmacy, School of Health Sciences Sector of Pharmaceutical Technology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Kumlesh K Dev
- Department of Physiology, Drug Development Research Group, School of Medicine, Trinity College Dublin, Dublin, D18 DH50, Ireland.
| | - Georgia Valsami
- Department of Pharmacy, School of Health Sciences Sector of Pharmaceutical Technology, National and Kapodistrian University of Athens, Athens, 15784, Greece.
| |
Collapse
|
20
|
Popescu R, Dinu-Pîrvu CE, Ghica MV, Anuța V, Popa L. Physico-Chemical Characterization and Initial Evaluation of Carboxymethyl Chitosan-Hyaluronan Hydrocolloid Systems with Insulin Intended for Intranasal Administration. Int J Mol Sci 2024; 25:10452. [PMID: 39408782 PMCID: PMC11476560 DOI: 10.3390/ijms251910452] [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: 09/13/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The nasal route of administration can bypass the blood-brain barrier in order to obtain a higher concentration in the brain, thus offering a feasible alternative route of administration for diseases associated with the central nervous system. The advantages of the intranasal administration and the potential favorable therapeutic effects of intranasally administered insulin led to the formulation of carboxymethyl chitosan (CMC) and sodium hyaluronate (NaHA) hydrocolloidal systems with insulin for nasal administration, targeting nose-to-brain delivery and the initial assessment of these systems. The influence of the formulation variables on the response parameters defined as surface properties, rheology, and in vitro release of insulin were analyzed using experimental design and statistical programs (Modde and Minitab software). The systems recorded good wetting and adhesion capacity, allowing the spread of the hydrocolloidal systems on the nasal mucosa. The samples had a pseudoplastic flow and the rapid release of the insulin was according to our objective. According to the physico-chemical characterization and preliminary assessment, these formulations are appropriate for administration on the nasal mucosa, but further studies are necessary to demonstrate the beneficial therapeutic actions and the safety of using intranasal insulin.
Collapse
Affiliation(s)
- Roxana Popescu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Str., 020950 Bucharest, Romania; (R.P.); (C.-E.D.-P.); (V.A.); (L.P.)
| | - Cristina-Elena Dinu-Pîrvu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Str., 020950 Bucharest, Romania; (R.P.); (C.-E.D.-P.); (V.A.); (L.P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Mihaela Violeta Ghica
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Str., 020950 Bucharest, Romania; (R.P.); (C.-E.D.-P.); (V.A.); (L.P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Valentina Anuța
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Str., 020950 Bucharest, Romania; (R.P.); (C.-E.D.-P.); (V.A.); (L.P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Lăcrămioara Popa
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Str., 020950 Bucharest, Romania; (R.P.); (C.-E.D.-P.); (V.A.); (L.P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
21
|
Mahmoud DM, El-Ela FIA, Fouad AG, Belal A, Ali MAM, Ghoneim MM, Almeheyawi RN, Attia ME, Mahmoud TM. Improving the bioavailability and therapeutic efficacy of felodipine for the control of diabetes-associated atherosclerosis: In vitro and in vivo characterization. Int J Pharm 2024; 661:124395. [PMID: 38945465 DOI: 10.1016/j.ijpharm.2024.124395] [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: 04/30/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Felodipine has proven to be effective as an atherosclerosis therapy because it increases blood flow to the vessel wall. However, the poor solubility, low bioavailability, and hepatic first-pass metabolism of oral felodipine compromise its therapeutic effectiveness. The study's goal is to create a nasal pH-sensitive hydrogel of felodipine-loaded invasomes (IPHFI) that will improve felodipine's release, permeation, bioavailability, and efficacy as a potential diabetes-associated atherosclerosis therapy. According to the pre-formulation study, the felodipine-loaded invasomes formulation composed of phospholipid (3%w/v), cholesterol (0.16%w/v), ethanol (3%v/v) and cineole (1%v/v) was chosen as the optimum formulation. The optimum formulation was characterized in vitro and then mixed with a mixture of chitosan and glyceryl monooleate to make the IPHFI formulation. The IPHFI formulation enhanced the release and permeation of felodipine by 2.99 and 3-fold, respectively. To assess the efficacy and bioavailability of the IPHFI formulation, it was studied in vivo using an experimental atherosclerosis rat model. Compared to oral free felodipine, the nasal administration of the IPHFI formulation increased the bioavailability by 3.37-fold and decreased the serum cholesterol, triglycerides, LDL, and calcification score by 1.56, 1.53, 1.80, and 1.18 ratios, respectively. Thus, nasal IPHFI formulation may represent a promising diabetes-associated atherosclerosis therapy.
Collapse
Affiliation(s)
- Dina M Mahmoud
- Department of Pharmaceutics, Faculty of Pharmacy, El Saleheya El Gadida University, El Saleheya El Gadida, Sharkia, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.
| | - Fatma I Abo El-Ela
- Department of Pharmacology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Amr Gamal Fouad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), 11623, Riyadh, Saudi Arabia; Department of Biochemistry, Faculty of Science, Ain Shams University, Abbassia 11566, Cairo, Egypt.
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Riyadh 13713, Saudi Arabia.
| | - Rania N Almeheyawi
- Department of Physical therapy, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia.
| | - Mary Eskander Attia
- Pharmacology department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Tamer M Mahmoud
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
22
|
Muntu CM, Avanti C, Hayun, Surini S. Promising brain biodistribution of insulin via intranasal dry powder for nose-to-brain delivery. Heliyon 2024; 10:e33657. [PMID: 39027498 PMCID: PMC11255508 DOI: 10.1016/j.heliyon.2024.e33657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Nose-to-brain delivery (NTBD) offering potential benefits for treating Alzheimer's disease. In previous research, insulin dry powder (IDP) formulation for NTBD was developed, exhibiting favorable stability. This study aims to conduct in vitro and ex vivo assessment of release, permeation, mucoadhesion and histopathology, as well as an in vivo biodistribution study to produce IDP for NTBD and evaluate brain biodistribution. Spray-freeze-dried IDP formulations with varying weight ratios of trehalose-to-inulin were produced and analyzed. The release study was carried out in PBS with a pH of 5.8 stirred at 50 rpm and maintained at 37 °C ± 0.5 °C. Goat nasal mucosa was used for ex vivo permeation and mucoadhesion testing under similar conditions. An ex vivo histopathological examination and an in vivo study using enzyme-linked immunosorbent assay, were also performed. The IDP dissolution study demonstrated complete release of all IDPs within 120 min. The permeation study indicated that steady-state conditions were observed between 30 and 240 min. The mucoadhesion study unveiled that IDP F5 exhibited the fastest mucoadhesion time and the least force required within the fastest time of 43.60 ± 2.57 s. The histopathological study confirmed that none of the tested IDPs induced irritation in the nasal mucosa. Furthermore, the biodistribution study demonstrated the absence of detectable insulin in the plasma, while IDP F3 exhibited the highest deposited concentration of insulin within both the olfactory bulb and the whole brain. The extensive evaluation of the IDP formulations through in vitro, ex vivo, and in vivo studies implies their strength non-invasive NTBD. IDP F3, with a 1:1 wt ratio of trehalose to inulin, exhibited favorable brain biodistribution outcomes and was recommended for further investigation and development in the context of NTBD.
Collapse
Affiliation(s)
- Cynthia Marisca Muntu
- Laboratory of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Indonesia, Depok 16424, West Java, Indonesia
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Surabaya, Surabaya 60293, East Java, Indonesia
| | - Christina Avanti
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Surabaya, Surabaya 60293, East Java, Indonesia
| | - Hayun
- Laboratory of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmacy, Universitas Indonesia, Depok 16424, West Java, Indonesia
| | - Silvia Surini
- Laboratory of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Indonesia, Depok 16424, West Java, Indonesia
| |
Collapse
|
23
|
Cimino C, Bonaccorso A, Tomasello B, Alberghina GA, Musumeci T, Puglia C, Pignatello R, Marrazzo A, Carbone C. W/O/W Microemulsions for Nasal Delivery of Hydrophilic Compounds: A Preliminary Study. J Pharm Sci 2024; 113:1636-1644. [PMID: 38281664 DOI: 10.1016/j.xphs.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
The administration of hydrophilic therapeutics has always been a great challenge because of their low bioavailability after administration. For this purpose, W/O/W microemulsion resulted to be a potential successful strategy for the delivery of hydrophilic compounds, interesting for the nasal mucosal therapy. Herein, an optimized biphasic W/O microemulsion was designed, through a preliminary screening, and it was inverted in a triphasic W/O/W microemulsion, intended for the nasal administration. In order to enhance the mucosal retention, surface modification of the biphasic W/O microemulsion was performed adding didodecyldimethylammonium bromide, and then converting the system into a cationic triphasic W/O/W microemulsion. The developed samples were characterized in terms of droplet size, polydispersity, zeta potential, pH and osmolality. The physical long-term stability was analyzed storing samples at accelerated conditions (40 ± 2 °C and 75 ± 5 % RH) for 6 months in a constant climate chamber, following ICH guidelines Q1A (R2). In order to verify the potential retention on the nasal mucosa, the two triphasic systems were analyzed in terms of mucoadhesive properties, measuring the in vitro interaction with mucin over time. Furthermore, fluorescein sodium salt was selected as a model hydrophilic drug to be encapsulated into the inner core of the two triphasic W/O/W microemulsions, and its release was analyzed compared to the free probe solution. The cytocompatibility of the two platforms was assessed on two cell lines, human fibroblasts HFF1 and Calu-3 cell lines, chosen as pre-clinical models for nasal and bronchial/tracheal airway epithelium.
Collapse
Affiliation(s)
- Cinzia Cimino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Angela Bonaccorso
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Barbara Tomasello
- Section of Biochemistry, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Giovanni Anfuso Alberghina
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Teresa Musumeci
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Carmelo Puglia
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Rosario Pignatello
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Agostino Marrazzo
- NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; Medicinal Chemistry Laboratory, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy
| | - Claudia Carbone
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95124 Catania, Italy; NANOMED, Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Viale A. Doria 6, 95124 Catania, Italy.
| |
Collapse
|
24
|
Xu D, Song XJ, Chen X, Wang JW, Cui YL. Advances and future perspectives of intranasal drug delivery: A scientometric review. J Control Release 2024; 367:366-384. [PMID: 38286336 DOI: 10.1016/j.jconrel.2024.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
Intranasal drug delivery is as a noninvasive and efficient approach extensively utilized for treating the local, central nervous system, and systemic diseases. Despite numerous reviews delving into the application of intranasal drug delivery across biomedical fields, a comprehensive analysis of advancements and future perspectives remains elusive. This review elucidates the research progress of intranasal drug delivery through a scientometric analysis. It scrutinizes several challenges to bolster research in this domain, encompassing a thorough exploration of entry and elimination mechanisms specific to intranasal delivery, the identification of drugs compatible with the nasal cavity, the selection of dosage forms to surmount limited drug-loading capacity and poor solubility, and the identification of diseases amenable to the intranasal delivery strategy. Overall, this review furnishes a perspective aimed at galvanizing future research and development concerning intranasal drug delivery.
Collapse
Affiliation(s)
- Dong Xu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi' an 710032, China
| | - Xu-Jiao Song
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Xue Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
| | - Jing-Wen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi' an 710032, China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
25
|
Koo J, Lim C, Oh KT. Recent Advances in Intranasal Administration for Brain-Targeting Delivery: A Comprehensive Review of Lipid-Based Nanoparticles and Stimuli-Responsive Gel Formulations. Int J Nanomedicine 2024; 19:1767-1807. [PMID: 38414526 PMCID: PMC10898487 DOI: 10.2147/ijn.s439181] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
Addressing disorders related to the central nervous system (CNS) remains a complex challenge because of the presence of the blood-brain barrier (BBB), which restricts the entry of external substances into the brain tissue. Consequently, finding ways to overcome the limited therapeutic effect imposed by the BBB has become a central goal in advancing delivery systems targeted to the brain. In this context, the intranasal route has emerged as a promising solution for delivering treatments directly from the nose to the brain through the olfactory and trigeminal nerve pathways and thus, bypassing the BBB. The use of lipid-based nanoparticles, including nano/microemulsions, liposomes, solid lipid nanoparticles, and nanostructured lipid carriers, has shown promise in enhancing the efficiency of nose-to-brain delivery. These nanoparticles facilitate drug absorption from the nasal membrane. Additionally, the in situ gel (ISG) system has gained attention owing to its ability to extend the retention time of administered formulations within the nasal cavity. When combined with lipid-based nanoparticles, the ISG system creates a synergistic effect, further enhancing the overall effectiveness of brain-targeted delivery strategies. This comprehensive review provides a thorough investigation of intranasal administration. It delves into the strengths and limitations of this specific delivery route by considering the anatomical complexities and influential factors that play a role during dosing. Furthermore, this study introduces strategic approaches for incorporating nanoparticles and ISG delivery within the framework of intranasal applications. Finally, the review provides recent information on approved products and the clinical trial status of products related to intranasal administration, along with the inclusion of quality-by-design-related insights.
Collapse
Affiliation(s)
- Jain Koo
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Chaemin Lim
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Marcotte H, Cao Y, Zuo F, Simonelli L, Sammartino JC, Pedotti M, Sun R, Cassaniti I, Hagbom M, Piralla A, Yang J, Du L, Percivalle E, Bertoglio F, Schubert M, Abolhassani H, Sherina N, Guerra C, Borte S, Rezaei N, Kumagai-Braesch M, Xue Y, Su C, Yan Q, He P, Grönwall C, Klareskog L, Calzolai L, Cavalli A, Wang Q, Robbiani DF, Hust M, Shi Z, Feng L, Svensson L, Chen L, Bao L, Baldanti F, Xiao J, Qin C, Hammarström L, Yang X, Varani L, Xie XS, Pan-Hammarström Q. Conversion of monoclonal IgG to dimeric and secretory IgA restores neutralizing ability and prevents infection of Omicron lineages. Proc Natl Acad Sci U S A 2024; 121:e2315354120. [PMID: 38194459 PMCID: PMC10801922 DOI: 10.1073/pnas.2315354120] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 01/11/2024] Open
Abstract
The emergence of Omicron lineages and descendent subvariants continues to present a severe threat to the effectiveness of vaccines and therapeutic antibodies. We have previously suggested that an insufficient mucosal immunoglobulin A (IgA) response induced by the mRNA vaccines is associated with a surge in breakthrough infections. Here, we further show that the intramuscular mRNA and/or inactivated vaccines cannot sufficiently boost the mucosal secretory IgA response in uninfected individuals, particularly against the Omicron variant. We thus engineered and characterized recombinant monomeric, dimeric, and secretory IgA1 antibodies derived from four neutralizing IgG monoclonal antibodies (mAbs 01A05, rmAb23, DXP-604, and XG014) targeting the receptor-binding domain of the spike protein. Compared to their parental IgG antibodies, dimeric and secretory IgA1 antibodies showed a higher neutralizing activity against different variants of concern (VOCs), in part due to an increased avidity. Importantly, the dimeric or secretory IgA1 form of the DXP-604 antibody significantly outperformed its parental IgG antibody, and neutralized the Omicron lineages BA.1, BA.2, and BA.4/5 with a 25- to 75-fold increase in potency. In human angiotensin converting enzyme 2 (ACE2) transgenic mice, a single intranasal dose of the dimeric IgA DXP-604 conferred prophylactic and therapeutic protection against Omicron BA.5. Thus, dimeric or secretory IgA delivered by nasal administration may potentially be exploited for the treatment and prevention of Omicron infection, thereby providing an alternative tool for combating immune evasion by the current circulating subvariants and, potentially, future VOCs.
Collapse
Affiliation(s)
- Harold Marcotte
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Yunlong Cao
- Changping Laboratory, Beijing102206, People’s Republic of China
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing100871, People’s Republic of China
| | - Fanglei Zuo
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Luca Simonelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Josè Camilla Sammartino
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia27100, Italy
| | - Mattia Pedotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Rui Sun
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Irene Cassaniti
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia27100, Italy
| | - Marie Hagbom
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden
| | - Antonio Piralla
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia27100, Italy
| | - Jinxuan Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming650023, People’s Republic of China
| | - Likun Du
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Elena Percivalle
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia27100, Italy
| | - Federico Bertoglio
- Department of Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig38106, Germany
| | - Maren Schubert
- Department of Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig38106, Germany
| | - Hassan Abolhassani
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Natalia Sherina
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Concetta Guerra
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, Leipzig04129, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, Leipzig04129, Germany
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran14194, Iran
| | - Makiko Kumagai-Braesch
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm14186, Sweden
| | - Yintong Xue
- Department of Immunology, Peking University Health Science Center, Beijing100191, People’s Republic of China
| | - Chen Su
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing100871, People’s Republic of China
| | - Qihong Yan
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences,Guangzhou510530, People’s Republic of China
| | - Ping He
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences,Guangzhou510530, People’s Republic of China
| | - Caroline Grönwall
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm17176, Sweden
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm17176, Sweden
- Rheumatology Unit, Karolinska University Hospital, Stockholm17176, Sweden
| | - Luigi Calzolai
- European Commission, Joint Research Centre, Ispra21027, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, 200032 Shanghai200032, People’s Republic of China
| | - Davide F. Robbiani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Michael Hust
- Department of Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig38106, Germany
| | - Zhengli Shi
- State Key laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei430071, People’s Republic of China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences,Guangzhou510530, People’s Republic of China
| | - Lennart Svensson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden
- Division of Infectious Diseases, Department of Medicine, Karolinska Institute, Stockholm17177, Sweden
| | - Ling Chen
- Guangzhou Laboratory, Guangzhou510005, People’s Republic of China
| | - Linlin Bao
- Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, National Health Commission Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing100021, People’s Republic of China
- National Center of Technology Innovation for Animal Model, Beijing102206, People’s Republic of China
| | - Fausto Baldanti
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia27100, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia27100, Italy
| | - Junyu Xiao
- Changping Laboratory, Beijing102206, People’s Republic of China
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing100871, People’s Republic of China
| | - Chuan Qin
- Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, National Health Commission Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing100021, People’s Republic of China
- National Center of Technology Innovation for Animal Model, Beijing102206, People’s Republic of China
| | - Lennart Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Xinglou Yang
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming650023, People’s Republic of China
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona6500, Switzerland
| | - Xiaoliang Sunney Xie
- Changping Laboratory, Beijing102206, People’s Republic of China
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing100871, People’s Republic of China
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| |
Collapse
|
27
|
Abdelmonem R, El-Enin HAA, Abdelkader G, Abdel-Hakeem M. Formulation and characterization of lamotrigine nasal insert targeted brain for enhanced epilepsy treatment. Drug Deliv 2023; 30:2163321. [PMID: 36579655 PMCID: PMC9809415 DOI: 10.1080/10717544.2022.2163321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lamotrigine. (LMT) is a triazine drug has an antiepileptic effect but with low water solubility, dissolution rate and thus therapeutic effect. Spanlastics are nano-vesicular carriers' act as site-specific drug delivery system. Intranasal route could direct the drug from nose to brain and provide a faster and more specific therapeutic effect. Therefore, this study aimed to upload lamotrigine onto nano-vesicles using spanlastic nasal insert delivery for effective epilepsy treatment via overcoming lamotrigine's low solubility and improving its bioavailability. Lamtrigine-loaded nano-spanlastic vesicles were prepared by ethanol injection method. To study different formulation factor's effect on formulations characters; particle size (PS), Zeta potential (ZP), polydispersity index (PDI), entrapment efficiency percentage (EE%) and LMT released amount after 6 h (Q6h); 2^1 and 3^1 full factorial designs were employed. Optimized formula was loaded in lyophilized nasal inserts formulation which were characterized for LMT release and mucoadhesion. Pharmacokinetics studies in plasma and brain were performed on rats to investigate drug targeting efficiency. The optimal nano-spanlastic formulation (F4; containing equal Span 60 amount (100 mg) and edge activator; Tween 80) exhibited nano PS (174.2 nm), high EE% (92.75%), and Q6h > 80%. The prepared nasal inserts (S4) containing 100 mg HPMC has a higher mucoadhesive force (9319.5 dyne/cm2) and dissolution rate (> 80% within 10 min) for rapid in vivo bio-distribution. In vivo studies showed considerable improvement brain and plasma's rate and extent absorption after intranasal administration indicating a high brain targeting efficiency. The results achieved indicate that nano-spanlastic nasal-inserts offer a promising LMT brain targeting in order to maximize its antiepileptic effect.
Collapse
Affiliation(s)
- Rehab Abdelmonem
- Department of Industrial Pharmacy, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, 12566, Egypt
| | - Hadel A. Abo El-Enin
- Department of Pharmaceutics, National organization of drug Control and Research (NODCAR), Giza, Egypt,CONTACT Hadel A. Abo El-Enin Department of Pharmaceutics, National organization of drug Control and Research (NODCAR), Giza, Egypt
| | - Ghada Abdelkader
- College of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt
| | - Mohamed Abdel-Hakeem
- Department of pharmaceutical biotechnology, College of biotechnology, Misr University For Science and Technology (MUST), 6th of October City, Giza, Egypt
| |
Collapse
|
28
|
Pires PC, Paiva-Santos AC, Veiga F. Liposome-Derived Nanosystems for the Treatment of Behavioral and Neurodegenerative Diseases: The Promise of Niosomes, Transfersomes, and Ethosomes for Increased Brain Drug Bioavailability. Pharmaceuticals (Basel) 2023; 16:1424. [PMID: 37895895 PMCID: PMC10610493 DOI: 10.3390/ph16101424] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Psychiatric and neurodegenerative disorders are amongst the most prevalent and debilitating diseases, but current treatments either have low success rates, greatly due to the low permeability of the blood-brain barrier, and/or are connected to severe side effects. Hence, new strategies are extremely important, and here is where liposome-derived nanosystems come in. Niosomes, transfersomes, and ethosomes are nanometric vesicular structures that allow drug encapsulation, protecting them from degradation, and increasing their solubility, permeability, brain targeting, and bioavailability. This review highlighted the great potential of these nanosystems for the treatment of Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, anxiety, and depression. Studies regarding the encapsulation of synthetic and natural-derived molecules in these systems, for intravenous, oral, transdermal, or intranasal administration, have led to an increased brain bioavailability when compared to conventional pharmaceutical forms. Moreover, the developed formulations proved to have neuroprotective, anti-inflammatory, and antioxidant effects, including brain neurotransmitter level restoration and brain oxidative status improvement, and improved locomotor activity or enhancement of recognition and working memories in animal models. Hence, albeit being relatively new technologies, niosomes, transfersomes, and ethosomes have already proven to increase the brain bioavailability of psychoactive drugs, leading to increased effectiveness and decreased side effects, showing promise as future therapeutics.
Collapse
Affiliation(s)
- Patrícia C. Pires
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Cláudia Paiva-Santos
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
29
|
Abla KK, Mehanna MM. The battle of lipid-based nanocarriers against blood-brain barrier: a critical review. J Drug Target 2023; 31:832-857. [PMID: 37577919 DOI: 10.1080/1061186x.2023.2247583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Central nervous system integrity is the state of brain functioning across sensory, cognitive, emotional-social behaviors, and motor domains, allowing a person to realise his full potential. Thus, brain disorders seriously affect patients' quality of life. Efficient drug delivery to treat brain disorders remains a crucial challenge due to numerous brain barriers, particularly the blood-brain barrier (BBB), which greatly impacts the ultimate drug therapeutic efficacy. Lately, nanocarrier technology has made huge progress in overcoming these barriers by improving drug solubility, ameliorating its retention, reducing its toxicity, and targeting the encapsulated agents to different brain tissues. The current review primarily offers an overview of the different components of BBB and the progress, strategies, and contemporary applications of the nanocarriers, specifically lipid-based nanocarriers (LBNs), in treating various brain disorders.
Collapse
Affiliation(s)
- Kawthar K Abla
- Pharmaceutical Nanotechnology Research Lab, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Mohammed M Mehanna
- Faculty of Pharmacy, Industrial Pharmacy Department, Alexandria University, Alexandria, Egypt
| |
Collapse
|
30
|
Özkan B, Altuntaş E, Ünlü Ü, Doğan HH, Özsoy Y, Çakır Koç R. Development of an Antiviral Ion-Activated In Situ Gel Containing 18β-Glycyrrhetinic Acid: A Promising Alternative against Respiratory Syncytial Virus. Pharmaceutics 2023; 15:2055. [PMID: 37631269 PMCID: PMC10458153 DOI: 10.3390/pharmaceutics15082055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 08/27/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) is a major cause of serious lower respiratory infections and poses a considerable risk to public health globally. Only a few treatments are currently used to treat RSV infections, and there is no RSV vaccination. Therefore, the need for clinically applicable, affordable, and safe RSV prevention and treatment solutions is urgent. In this study, an ion-activated in situ gelling formulation containing the broad-spectrum antiviral 18β-glycyrrhetinic acid (GA) was developed for its antiviral effect on RSV. In this context, pH, mechanical characteristics, ex vivo mucoadhesive strength, in vitro drug release pattern, sprayability, drug content, and stability were all examined. Rheological characteristics were also tested using in vitro gelation capacity and rheological synergism tests. Finally, the cytotoxic and antiviral activities of the optimized in situ gelling formulation on RSV cultured in the human laryngeal epidermoid carcinoma (HEp-2) cell line were evaluated. In conclusion, the optimized formulation prepared with a combination of 0.5% w/w gellan gum and 0.5% w/w sodium carboxymethylcellulose demonstrated good gelation capacity and sprayability (weight deviation between the first day of the experiment (T0) and the last day of the experiment (T14) was 0.34%), desired rheological synergism (mucoadhesive force (Fb): 9.53 Pa), mechanical characteristics (adhesiveness: 0.300 ± 0.05 mJ), ex vivo bioadhesion force (19.67 ± 1.90 g), drug content uniformity (RSD%: 0.494), and sustained drug release over a period of 6 h (24.56% ± 0.49). The optimized formulation demonstrated strong anti-hRSV activity (simultaneous half maximal effective concentration (EC50) = 0.05 µg/mL; selectivity index (SI) = 306; pre-infection EC50 = 0.154 µg/mL; SI = 100), which was significantly higher than that of ribavirin (EC50 = 4.189 µg/mL; SI = 28) used as a positive control against hRSV, according to the results of the antiviral activity test. In conclusion, this study showed that nasal in situ gelling spray can prevent viral infection and replication by directly inhibiting viral entry or modulating viral replication.
Collapse
Affiliation(s)
- Burcu Özkan
- Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul 34220, Turkey;
| | - Ebru Altuntaş
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, Istanbul 34116, Turkey;
| | - Ümmühan Ünlü
- Elderly Care Program, Ataturk Health Services Vocational School, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey;
| | - Hasan Hüseyin Doğan
- Department of Biology, Science Faculty, Alaeddin Keykubat Campus, Selcuk University, Konya 42130, Turkey;
| | - Yıldız Özsoy
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, Istanbul 34116, Turkey;
| | - Rabia Çakır Koç
- Faculty of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey;
| |
Collapse
|
31
|
Pires PC, Fernandes M, Nina F, Gama F, Gomes MF, Rodrigues LE, Meirinho S, Silvestre S, Alves G, Santos AO. Innovative Aqueous Nanoemulsion Prepared by Phase Inversion Emulsification with Exceptional Homogeneity. Pharmaceutics 2023; 15:1878. [PMID: 37514064 PMCID: PMC10384498 DOI: 10.3390/pharmaceutics15071878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Formulating low-solubility or low-permeability drugs is a challenge, particularly with the low administration volumes required in intranasal drug delivery. Nanoemulsions (NE) can solve both issues, but their production and physical stability can be challenging, particularly when a high proportion of lipids is necessary. Hence, the aim of the present work was to develop a NE with good solubilization capacity for lipophilic drugs like simvastatin and able to promote the absorption of drugs with low permeability like fosphenytoin. Compositions with high proportion of two lipids were screened and characterized. Surprisingly, one of the compositions did not require high energy methods for high droplet size homogeneity. To better understand formulation factors important for this feature, several related compositions were evaluated, and their relative cytotoxicity was screened. Optimized compositions contained a high proportion of propylene glycol monocaprylate NF, formed very homogenous NE using a low-energy phase inversion method, solubilized simvastatin at high drug strength, and promoted a faster intranasal absorption of the hydrophilic prodrug fosphenytoin. Hence, a new highly homogeneous NE obtained by a simple low-energy method was successfully developed, which is a potential alternative for industrial application for the solubilization and protection of lipophilic actives, as well as (co-)administration of hydrophilic molecules.
Collapse
Affiliation(s)
- Patrícia C Pires
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mariana Fernandes
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Francisca Nina
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Francisco Gama
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Maria F Gomes
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Lina E Rodrigues
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Sara Meirinho
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Samuel Silvestre
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Sciences, University of Beira Interior, Rua Marquês d'Ávila e Bolama, 6201-001 Covilhã, Portugal
| | - Gilberto Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Adriana O Santos
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
32
|
De Martini LB, Sulmona C, Brambilla L, Rossi D. Cell-Penetrating Peptides as Valuable Tools for Nose-to-Brain Delivery of Biological Drugs. Cells 2023; 12:1643. [PMID: 37371113 DOI: 10.3390/cells12121643] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Due to their high specificity toward the target and their low toxicity, biological drugs have been successfully employed in a wide range of therapeutic areas. It is yet to be mentioned that biologics exhibit unfavorable pharmacokinetic properties, are susceptible to degradation by endogenous enzymes, and cannot penetrate biological barriers such as the blood-brain barrier (i.e., the major impediment to reaching the central nervous system (CNS)). Attempts to overcome these issues have been made by exploiting the intracerebroventricular and intrathecal routes of administration. The invasiveness and impracticality of these procedures has, however, prompted the development of novel drug delivery strategies including the intranasal route of administration. This represents a non-invasive way to achieve the CNS, reducing systemic exposure. Nonetheless, biotherapeutics strive to penetrate the nasal epithelium, raising the possibility that direct delivery to the nervous system may not be straightforward. To maximize the advantages of the intranasal route, new approaches have been proposed including the use of cell-penetrating peptides (CPPs) and CPP-functionalized nanosystems. This review aims at describing the most impactful attempts in using CPPs as carriers for the nose-to-brain delivery of biologics by analyzing their positive and negative aspects.
Collapse
Affiliation(s)
- Lisa Benedetta De Martini
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Claudia Sulmona
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| |
Collapse
|
33
|
To Quoc T, Bíró K, Pető Á, Kósa D, Sinka D, Lekli I, Kiss-Szikszai A, Budai I, Béres M, Vecsernyés M, Fehér P, Bácskay I, Ujhelyi Z. Development and Evaluation of an FDM Printed Nasal Device for CPZ Solid Nanoparticles. Molecules 2023; 28:molecules28114406. [PMID: 37298882 DOI: 10.3390/molecules28114406] [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: 04/14/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Nasal drug delivery has been a focus of scientific interest for decades. A number of drug delivery systems and devices are available and have been highly successful in providing better and more comfortable therapy. The benefits of nasal drug delivery are not in question. The nasal surface provides an excellent context for the targeted delivery of active substances. In addition to the large nasal surface area and intensive absorption, the active substances delivered through the nose overcome the blood-brain barrier and can be delivered directly to the central nervous system. Formulations for nasal administration are typically solutions or liquid dispersed systems such as emulsions or suspensions. Formulation techniques for nanostructures have recently undergone intensive development. Solid-phase heterogeneous dispersed systems represent a new direction in pharmaceutical formulations. The wide range of possible examples and the variety of excipients allow for the delivery of a wide range of active ingredients. The aim of our experimental work was to develop a solid drug delivery system that possesses all of the above-mentioned advantageous properties. In developing solid nanosystems, we not only exploited the advantages of size but also the adhesive and penetration-enhancing properties of excipients. During formulation, several amphiphilic compounds with adhesion properties and penetration enhancing effects were incorporated. We used chlorpromazine (CPZ), which is mainly used in the treatment of psychotic disorders such as schizophrenia and bipolar disorder. Chlorpromazine has been previously investigated by our team in other projects. With the availability of previous methods, the analytical characterization of the drug was carried out effectively. Due to the frequent and severe side effects of the drug, the need for therapeutic dose reduction is indisputable. In this series of experiments, we succeeded in constructing drug delivery systems. Finely divided Na nanoparticles were formed using a Büchi B90 nanospray dryer. An important step in the development of the drug carrier was the selection of suitable inert carrier compounds. Particle size determination and particle size distribution analysis were performed to characterize the prepared nanostructures. As safety is the most important aspect of any drug formulation, all components and systems were tested with different biocompatibility assays. The tests performed demonstrated the safe applicability of our systems. The bioavailability of chlorpromazine was studied as a function of the ratio of the active ingredient administered nasally and intravenously. As described above, most nasal formulations are liquids, but our system is solid, so there is currently no tool available to accurately target this system. As a supplement of the project, a nasal dosing device was developed, corresponding to the anatomical structure; a prototype of the device was made using 3D FDM technology. Our results lay the foundation for the design and industrial scaling of a new approach to the design and production of a high-bioavailability nasal medicinal product.
Collapse
Affiliation(s)
- Thinh To Quoc
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Krisztina Bíró
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Hospital Pharmacy at the University of Debrecen, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Ágota Pető
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Dóra Kósa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Dávid Sinka
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - István Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Attila Kiss-Szikszai
- Department of Organic Chemistry, University of Debrecen, Egyetem tér 1, 4010 Debrecen, Hungary
| | - István Budai
- Department of Engineering Management and Enterprise, Institute of Industrial Process Management, Faculty of Engineering, University of Debrecen, Ótemető utca 2, 4028 Debrecen, Hungary
| | - Mónika Béres
- Department of Medical Imaging, Department of Radiology and Imaging Science, Faculty of General Medicine, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Miklós Vecsernyés
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Pálma Fehér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| | - Zoltán Ujhelyi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei körút 98, 4032 Debrecen, Hungary
| |
Collapse
|
34
|
Changsan N, Sawatdee S, Suedee R, Chunhachaichana C, Srichana T. Aqueous cannabidiol β-cyclodextrin complexed polymeric micelle nasal spray to attenuate in vitro and ex-vivo SARS-CoV-2-induced cytokine storms. Int J Pharm 2023; 640:123035. [PMID: 37182795 PMCID: PMC10181874 DOI: 10.1016/j.ijpharm.2023.123035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/23/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Cannabidiol (CBD) has a number of biological effects by acting on the cannabinoid receptors CB1 and CB2. CBD may be involved in anti-inflammatory processes via CB1 and CB2 receptors, resulting in a decrease of pro-inflammatory cytokines. However, CBD's poor aqueous solubility is a major issue in pharmaceutical applications. The aim of the present study was to develop and evaluate a CBD nasal spray solution. A water-soluble CBD was prepared by complexation with β-cyclodextrin (β-CD) at a stoichiometric ratio of 1:1 and forming polymeric micelles using poloxamer 407. The mixture was then lyophilized and characterized using FT-IR, DSC, and TGA. CBD-β-CD complex-polymeric micelles were formulated for nasal spray drug delivery. The physicochemical properties of the CBD-β-CD complex-polymeric micelle nasal spray solution (CBD-β-CDPM-NS) were assessed. The results showed that the CBD content in the CBD-β-CD complex polymeric micelle powder was 102.1 ± 0.5%. The CBD-β-CDPM-NS was a clear colorless isotonic solution. The particle size, zeta potential, pH value, and viscosity were 111.9 ± 0.7 nm, 0.8 ± 0.1 mV, 6.02 ± 0.02, and 12.04 ± 2.64 cP, respectively. This formulation was stable over six months at ambient temperature. The CBD from CBD-β-CDPM-NS rapidly released to 100% within 1 min. Ex-vivo permeation studies of CBD-β-CDPM-NS through porcine nasal mucosa revealed a permeation rate of 4.8 μg/cm2/min, which indicated that CBD was effective in penetrating nasal epithelial cells. CBD-β-CDPM-NS was tested for its efficacy and safety in terms of cytokine production from nasal immune cells and toxicity to nasal epithelial cells. The CBD-β-CDPM-NS was not toxic to nasal epithelial at the concentration of CBD equivalent to 3.125-50 μg/mL. When the formulation was subjected to bioactivity testing against monocyte-like macrophage cells, it proved that the CBD-β-CDPM-NS has the potential to inhibit inflammatory cytokines. CBD-β-CDPM-NS demonstrated the formulation's ability to reduce the cytokine produced by S-RBD stimulation in ex vivo porcine nasal mucosa in both preventative and therapeutic modes.
Collapse
Affiliation(s)
- Narumon Changsan
- College of Pharmacy, Rangsit University, Pathumtani 12000, Thailand
| | - Somchai Sawatdee
- Drug and Cosmetics Excellence Center and School of Pharmacy, Walailak University, Thasala, Nakhon Si Thammarat, 80161, Thailand
| | - Roongnapa Suedee
- Molecular Recognition Materials Research Unit, Nanotec-PSU Center of Excellence on Drug Delivery System Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University Hat Yai, Songkhla 90112, Thailand
| | - Charisopon Chunhachaichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand.
| |
Collapse
|
35
|
Mehta PP, Fröhlich E, Khan R, Parihar A, Kumar CMS. Editorial: Advances in orally inhaled and nasal drug products (OINDPs). Front Pharmacol 2023; 14:1185609. [PMID: 37063274 PMCID: PMC10102658 DOI: 10.3389/fphar.2023.1185609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Affiliation(s)
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Raju Khan
- CSIR - Advanced Materials and Processes Research Institute (AMPRI), Bhopal, India
| | - Arpana Parihar
- CSIR - Advanced Materials and Processes Research Institute (AMPRI), Bhopal, India
| | - C. M. Santosh Kumar
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
36
|
Antunes JL, Amado J, Veiga F, Paiva-Santos AC, Pires PC. Nanosystems, Drug Molecule Functionalization and Intranasal Delivery: An Update on the Most Promising Strategies for Increasing the Therapeutic Efficacy of Antidepressant and Anxiolytic Drugs. Pharmaceutics 2023; 15:998. [PMID: 36986859 PMCID: PMC10054777 DOI: 10.3390/pharmaceutics15030998] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Depression and anxiety are high incidence and debilitating psychiatric disorders, usually treated by antidepressant or anxiolytic drug administration, respectively. Nevertheless, treatment is usually given through the oral route, but the low permeability of the blood-brain barrier reduces the amount of drug that will be able to reach it, thus consequently reducing the therapeutic efficacy. Which is why it is imperative to find new solutions to make these treatments more effective, safer, and faster. To overcome this obstacle, three main strategies have been used to improve brain drug targeting: the intranasal route of administration, which allows the drug to be directly transported to the brain by neuronal pathways, bypassing the blood-brain barrier and avoiding the hepatic and gastrointestinal metabolism; the use of nanosystems for drug encapsulation, including polymeric and lipidic nanoparticles, nanometric emulsions, and nanogels; and drug molecule functionalization by ligand attachment, such as peptides and polymers. Pharmacokinetic and pharmacodynamic in vivo studies' results have shown that intranasal administration can be more efficient in brain targeting than other administration routes, and that the use of nanoformulations and drug functionalization can be quite advantageous in increasing brain-drug bioavailability. These strategies could be the key to future improved therapies for depressive and anxiety disorders.
Collapse
Affiliation(s)
- Jéssica L. Antunes
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana Amado
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
37
|
Ferreira MD, Duarte J, Veiga F, Paiva-Santos AC, Pires PC. Nanosystems for Brain Targeting of Antipsychotic Drugs: An Update on the Most Promising Nanocarriers for Increased Bioavailability and Therapeutic Efficacy. Pharmaceutics 2023; 15:pharmaceutics15020678. [PMID: 36840000 PMCID: PMC9959474 DOI: 10.3390/pharmaceutics15020678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Orally administered antipsychotic drugs are the first-line treatment for psychotic disorders, such as schizophrenia and bipolar disorder. Nevertheless, adverse drug reactions jeopardize clinical outcomes, resulting in patient non-compliance. The design formulation strategies for enhancing brain drug delivery has been a major challenge, mainly due to the restrictive properties of the blood-brain barrier. However, recent pharmacokinetic and pharmacodynamic in vivo assays confirmed the advantage of the intranasal route when compared to oral and intravenous administration, as it allows direct nose-to-brain drug transport via neuronal pathways, reducing systemic side effects and maximizing therapeutic outcomes. In addition, the incorporation of antipsychotic drugs into nanosystems such as polymeric nanoparticles, polymeric mixed micelles, solid lipid nanoparticles, nanostructured lipid carriers, nanoemulsions, nanoemulgels, nanosuspensions, niosomes and spanlastics, has proven to be quite promising. The developed nanosystems, having a small and homogeneous particle size (ideal for nose-to-brain delivery), high encapsulation efficiency and good stability, resulted in improved brain bioavailability and therapeutic-like effects in animal models. Hence, although it is essential to continue research in this field, the intranasal delivery of nanosystems for the treatment of schizophrenia, bipolar disorder and other related disorders has proven to be quite promising, opening a path for future therapies with higher efficacy.
Collapse
Affiliation(s)
- Maria Daniela Ferreira
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana Duarte
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence: (A.C.P.-S.); or (P.C.P.)
| | - Patrícia C. Pires
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Correspondence: (A.C.P.-S.); or (P.C.P.)
| |
Collapse
|
38
|
Nano and Microemulsions for the Treatment of Depressive and Anxiety Disorders: An Efficient Approach to Improve Solubility, Brain Bioavailability and Therapeutic Efficacy. Pharmaceutics 2022; 14:pharmaceutics14122825. [PMID: 36559318 PMCID: PMC9783281 DOI: 10.3390/pharmaceutics14122825] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Most drugs used for the treatment of depression, anxiety and related disorders have low absorption, high metabolism, low brain targeting and/or low water solubility, which can make it hard to formulate them at high strength and can also lead to decreased bioavailability. Incorporating these drugs into nanometric emulsions can solve these issues. Hence, the aim of the present review was to assess the potential of nano and micro emulsions for the delivery of antidepressant and anxiolytic drugs. The results from several studies showed that nanometric emulsions were able to increase drug strength up to 20,270-fold (compared to aqueous solubility). Moreover, in general, the formulations showed droplet size, polydispersity index, zeta potential, viscosity, osmolality, pH, in vitro drug release and ex vivo drug permeation as adequate for the intended effect and administration route. In vivo animal pharmacokinetic experiments showed that nanometric emulsions improved systemic drug bioavailability and/or brain targeting, and in vivo pharmacodynamic studies showed that they had antidepressant and/or anxiolytic effects, also being apparently safe. Hence, the current review provides proof of the potential of nano and microemulsions for improving solubilization and increasing the overall bioavailability of antidepressant and/or anxiolytic drugs, providing evidence of a possible useful strategy for future therapies.
Collapse
|
39
|
Tan MSA, Pandey P, Lohman RJ, Falconer JR, Siskind DJ, Parekh HS. Fabrication and Characterization of Clozapine Nanoemulsion Sol-Gel for Intranasal Administration. Mol Pharm 2022; 19:4055-4066. [PMID: 36149013 DOI: 10.1021/acs.molpharmaceut.2c00513] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Clozapine is the most effective antipsychotic for treatment-resistant schizophrenia. However, it causes many adverse drug reactions (ADRs), which lead to poor treatment outcomes. Nose-to-brain (N2B) drug delivery offers a promising approach to reduce peripheral ADRs by minimizing systemic drug exposure. The aim of the present study was to develop and characterize clozapine-loaded nanoemulsion sol-gel (CLZ-NESG) for intranasal administration using high energy sonication method. A range of oils, surfactants, and cosurfactants were screened with the highest clozapine solubility selected for the development of nanoemulsion. Pseudoternary phase diagrams were constructed using a low-energy (spontaneous) method to identify the microemulsion regions (i.e., where mixtures were transparent). The final formulation, CLZ-NESG (pH 5.5 ± 0.2), comprising 1% w/w clozapine, 1% w/w oleic acid, 10% w/w polysorbate 80/propylene glycol (3:1), and 20% w/w poloxamer 407 (P407) solution, had an average globule size of ≤30 nm with PDI 0.2 and zeta potential of -39.7 ± 1.5 mV. The in vitro cumulative drug release of clozapine from the nanoemulsion gel at 34 °C (temperature of nasal cavity) after 72 h was 38.9 ± 4.6% compared to 84.2 ± 3.9% with the control solution. The permeation study using sheep nasal mucosa as diffusion barriers confirmed a sustained release of clozapine with 56.2 ± 2.3% cumulative drug permeated after 8 h. Additionally, the histopathological examination found no severe nasal ciliotoxicity on the mucosal tissues. The thermodynamic stability studies showed that the gel strength and viscosity of CLZ-NESG decreased after temperature cycling but was still seen to be in "gel" form at nasal temperature. However, the accelerated storage stability study showed a decrease in drug concentration after 3 months, which can be expected at elevated stress conditions. The formulation developed in this study showed desirable physicochemical properties for intranasal administration, highlighting the potential value of a nanoemulsion gel for improving drug bioavailability of clozapine for N2B delivery.
Collapse
Affiliation(s)
- Madeleine S A Tan
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia.,Medicines Management Unit, Department of Health, Northern Territory Government, Royal Darwin Hospital, 105 Rocklands Drive, Tiwi, Northern Territory 0810, Australia
| | - Preeti Pandey
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - Rink-Jan Lohman
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - James R Falconer
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - Dan J Siskind
- Faculty of Medicine, The University of Queensland, 20 Weightman Street, Herston, Queensland 4006, Australia.,Metro South Addiction and Mental Health Service, Level 2 Mental Health, Woolloongabba Community Health Centre, 228 Logan Road, Woolloongabba, Queensland 4102, Australia
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland, 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
40
|
Corazza E, di Cagno MP, Bauer-Brandl A, Abruzzo A, Cerchiara T, Bigucci F, Luppi B. Drug delivery to the brain: In situ gelling formulation enhances carbamazepine diffusion through nasal mucosa models with mucin. Eur J Pharm Sci 2022; 179:106294. [PMID: 36116696 DOI: 10.1016/j.ejps.2022.106294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/26/2022]
Abstract
The objective of this work was to optimize a thermosensitive in situ gelling formulation to improve intranasal and nose-to-brain delivery of the antiepileptic drug carbamazepine (CBZ). A preliminary procedure of vehicles obtained just mixing different fractions of poloxamer 407 (P407) and poloxamer 188 (P188) revealed preparations with phase transition temperatures, times to gelation and pH values suitable for nasal delivery. Subsequently, the mucoadhesive properties of the most promising formulations were tuned by adding hydroxypropylmethylcellulose types of different viscosity grades, and the effect of the adhesive polymers was evaluated by testing in vitro time and strength of mucoadhesion on specimens of sheep nasal mucosa. The formulation that showed the greatest mucoadhesive potential in vitro, with a time and force of mucoadhesion equal to 1746,75 s and 3.66 × 10-4 N, respectively, was that composed of 22% P407, 5% P188 and 0.8% HPMC low-viscous and it was further investigated for its ability to increase drug solubility and to control the release of the drug. Lastly, the capability of the candidate vehicle to ensure drug permeation across the biomimetic membrane Permeapad®, an artificial phospholipid-based barrier with a stratified architecture, and the same barrier enriched with a mucin layer was verified. The final formulation was characterized by a pH value of 6.0, underwent gelation at 32.33°C in 37.85 s, thus showing all the features required by in situ gelling thermosensitive preparations designed for nasal delivery and, more notably, it conserved the ability to favor drug permeation in the presence of mucin. These findings suggest that the optimized gelling system could be a promising and easy to realize strategy to improve CBZ delivery to the brain exploiting both a direct and indirect pathway.
Collapse
Affiliation(s)
- Elisa Corazza
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, Bologna 40127, Italy
| | - Massimiliano Pio di Cagno
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, Oslo 0371, Norway.
| | - Annette Bauer-Brandl
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Angela Abruzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, Bologna 40127, Italy
| | - Teresa Cerchiara
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, Bologna 40127, Italy
| | - Federica Bigucci
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, Bologna 40127, Italy
| | - Barbara Luppi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, Bologna 40127, Italy
| |
Collapse
|
41
|
Rodrigues M, Ali J, Alves G. Editorial: Intranasal Delivery of Central Nervous System Active Drugs: Opportunities and Challenges. Front Pharmacol 2022; 13:927812. [PMID: 35774602 PMCID: PMC9237567 DOI: 10.3389/fphar.2022.927812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/27/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Márcio Rodrigues
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- CPIRN-UDI-IPG—Center for Potential and Innovation of Natural Resources, Research Unit for Inland Development, Polytechnic Institute of Guarda, Guarda, Portugal
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Gilberto Alves
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- *Correspondence: Gilberto Alves,
| |
Collapse
|