1
|
Hu K, Xiao M, Chen S, Huang Y, Hou Z, Li X, Yang L. Innovative applications of natural polysaccharide polymers in intravesical therapy of bladder diseases. Carbohydr Polym 2025; 354:123307. [PMID: 39978897 DOI: 10.1016/j.carbpol.2025.123307] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/22/2025]
Abstract
Natural polysaccharide polymers, characterized by their remarkable biocompatibility, biodegradability, and structural versatility, hold great promise for intravesical therapy in treating of bladder diseases. Conditions such as bladder cancer and interstitial cystitis compromise drug efficacy by affecting the permeability of the bladder wall. Traditional therapeutic approaches are often hindered by physiological challenges, including rapid drug clearance and the intrinsic permeability barrier of the bladder. Polysaccharides like hyaluronic acid (HA) and chitosan (CS) have emerged as promising materials for intravesical drug delivery systems (IDDS), owing to their ability to repair tight junctions in the bladder wall, mitigate inflammation, and enhance permeability. This review provides a comprehensive overview of the mechanisms through which polysaccharide-based natural polymers regulate bladder wall permeability and highlights their advancements in delivery platforms, including nanoparticles, hydrogels, floating systems, and composite materials. By improving drug retention, enhancing bioavailability, and promoting patient adherence, these materials offer a solid foundation for the development of innovative therapeutic strategies for bladder diseases.
Collapse
Affiliation(s)
- Ke Hu
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Miaomiao Xiao
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Siwen Chen
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Yuanbing Huang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| | - Xiancheng Li
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| |
Collapse
|
2
|
Alsheikh R, Haimhoffer Á, Nemes D, Ujhelyi Z, Fehér P, Józsa L, Vasvári G, Pető Á, Kósa D, Nagy L, Horváth L, Balázs B, Bácskay I. Formulation of Thermo-Sensitive In Situ Gels Loaded with Dual Spectrum Antibiotics of Azithromycin and Ofloxacin. Polymers (Basel) 2024; 16:2954. [PMID: 39518163 PMCID: PMC11548455 DOI: 10.3390/polym16212954] [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: 09/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
In situ gels have been developed as an innovative strategy to prolong corneal residence time and enhance drug absorption compared to traditional eye drops. Our study aimed to formulate an ophthalmic in situ gel with a combination of two thermosensitive poloxamers, P407 and P188, in an optimal ratio not only to increase the time of action but also to increase the solubility of selected antibiotics for the treatment of ophthalmic infections. Two BSC II class substances, Azithromycin and Ofloxacin, with different mechanisms of action, have been incorporated into the in situ gel system after determining their solubility. The antibiotics-loaded in situ gel formulation was evaluated for its clarity, pH, rheological properties, and gel characteristics of gelling time, temperature, and capacity. The formulation demonstrated satisfactory clarity, appropriate pH, effective gelation properties in simulated tear fluid, and suitable rheological characteristics. In addition, APIs release insight has been studied through a dissolution test, and the effectivity against sensitive and resistant bacterial strains has been proved through the antimicrobial study. Therefore, our in situ gel system based on thermosensitive poloxamers, with two hydrophobic antibiotics, AZM and OFX, can be considered a valuable approach for ophthalmic drug delivery with an enhancement of the antibiotics bioavailability through increasing the contact time with the ocular surface and enhancing patient compliance.
Collapse
Affiliation(s)
- Raghad Alsheikh
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
- Doctorate School of Pharmaceutical Sciences, University of Debrecen, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, 4032 Debrecen, Hungary
| | - Ádám Haimhoffer
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Dániel Nemes
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Zoltán Ujhelyi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Pálma Fehér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Liza Józsa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Gábor Vasvári
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Ágota Pető
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Dóra Kósa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
| | - Lajos Nagy
- Department of Applied Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary;
| | - László Horváth
- Department of Pharmaceutical Surveillance and Economics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary;
| | - Bence Balázs
- Institute of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (R.A.); (Á.H.); (D.N.); (Z.U.); (P.F.); (L.J.); (G.V.)
- Doctorate School of Pharmaceutical Sciences, University of Debrecen, 4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
3
|
Wang P, Hong S, Cao C, Guo S, Wang C, Chen X, Wang X, Song P, Li N, Xu R. Ethosomes-mediated tryptanthrin delivery as efficient anti-psoriatic nanotherapy by enhancing topical drug absorption and lipid homeostasis. J Nanobiotechnology 2024; 22:584. [PMID: 39334378 PMCID: PMC11438247 DOI: 10.1186/s12951-024-02860-3] [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: 03/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Psoriasis is a chronic, relapsing, and refractory immune-mediated skin disease with the etiology and pharmaceutical targets remaining unsatisfactorily addressed. Topical herbal-derived compounds, such as tryptanthrin (Tryp), have been considered as an alternative therapy for psoriasis due to their lower costs and fewer side effects compared to other therapies. However, the effectiveness of topically administered drugs is substantially limited by the thickened pathological skin barrier and the low bioavailability of drugs in the deeper layers of the lesion. Ethosomes, being a novel phospholipid-based vesicle system with high content of ethanol, have been implicated in enhancing topical drug absorption and restoring psoriatic lesions. In this study, taking advantages of ethosomes as a soft and malleable drug carrier, we constructed the Tryp-loaded ethosome (Tryp-ES) through a one-step microfluidics-based technique. The optimal formulation of Tryp-ES was achieved by adding amino-acid-derived surfactant sodium lauroyl glutamate, and Tryp-ES exhibited homogeneous particle size and favorable stability at room temperature. In vitro evaluations showed that Tryp of Tryp-ES could be easily internalized into cells and accumulated in cell nuclei, hence inhibited the abnormally proliferated keratinocytes by inducing apoptosis. In vivo and in vitro assessment using psoritic skin of mice revealed that Tryp-ES had preferred skin retention and permeation of loaded drugs within the initial 1 h of topical administration, which could be attributed to transient disintegrations of cell membranes by ethosomes, thus improved cellular fluidity and permeability. Notably, a synergistic effect of ethosomes and Tryp was found in psoriatic mice. Tryp-ES-treated mice showed substantially ameliorated symptoms of psoriasis and reduced pathological alterations due to hyperplasia, inflammation and angiogenesis, without detectable local or systemic toxicities. Interestingly, lipidomics analysis confirmed that the supplementation of phospholipids, as in the form of ethosome vehicles, was an alterantive strategy to relieve psoriatic pathologies. Taken together, this study provides a novel impact for ethosomal topical delivery of Tryp and underlines their potential as an effective therapy for the management of psoriasis.
Collapse
Affiliation(s)
- Pengyu Wang
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Shihao Hong
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Can Cao
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shijie Guo
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chen Wang
- Central Instrument Facility, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xi Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xinnan Wang
- Central Instrument Facility, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ping Song
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
4
|
Mushtaq RY, Naveen NR, Rolla KJ, Al Shmrany H, Alshehri S, Salawi A, Kurakula M, Alghamdi MA, Rizg WY, Bakhaidar RB, Abualsunun WA, Hosny KM, Alamoudi AJ. Design and evaluation of magnetic-targeted bilosomal gel for rheumatoid arthritis: flurbiprofen delivery using superparamagnetic iron oxide nanoparticles. Front Pharmacol 2024; 15:1433734. [PMID: 39246659 PMCID: PMC11377347 DOI: 10.3389/fphar.2024.1433734] [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: 05/16/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction The study aimed to systematically enhance the fabrication process of flurbiprofen-loaded bilosomes (FSB) using Quality by Design (QbD) principles and Design of Experiments (DOE). The objective was to develop an optimized formulation with improved entrapment efficiency and targeted drug delivery capabilities. Methods The optimization process involved applying QbD principles and DOE to achieve the desired formulation characteristics. Superparamagnetic iron oxide nanoparticles (SPIONs) were incorporated to impart magnetic responsiveness. The size, entrapment efficiency, morphology, and in vitro release patterns of the FSB formulation were evaluated. Additionally, an in situ forming hydrogel incorporating FSB was developed, with its gelation time and drug release kinetics assessed. In vivo studies were conducted on osteoarthritic rats to evaluate the efficacy of the FSB-loaded hydrogel. Results The optimized FSB formulation yielded particles with a size of 453.60 nm and an entrapment efficiency of 91.57%. The incorporation of SPIONs enhanced magnetic responsiveness. Morphological evaluations and in vitro release studies confirmed the structural integrity and sustained release characteristics of the FSB formulation. The in situ forming hydrogel exhibited a rapid gelation time of approximately 40 ± 1.8 s and controlled drug release kinetics. In vivo studies demonstrated a 27.83% reduction in joint inflammation and an 85% improvement in locomotor activity in osteoarthritic rats treated with FSB-loaded hydrogel. Discussion This comprehensive investigation highlights the potential of FSB as a promising targeted drug delivery system for the effective management of osteoarthritis. The use of QbD and DOE in the formulation process, along with the integration of SPIONs, resulted in an optimized FSB formulation with enhanced entrapment efficiency and targeted delivery capabilities. The in situ forming hydrogel further supported the formulation's applicability for injectable applications, providing rapid gelation and sustained drug release. The in vivo results corroborate the formulation's efficacy, underscoring its potential for improving the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Rayan Y Mushtaq
- Department of Pharmaceutics, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nimbagal Raghavendra Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, Mandya, Karnataka, India
| | - Krishna Jayanth Rolla
- Department of Data Analytics, Global Technical Software Service, Inc. (GTSS), Hamilton, NJ, United States
| | - Humood Al Shmrany
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Sameer Alshehri
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | - Majed A Alghamdi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdualziz University, Jeddah, Saudi Arabia
| | - Waleed Y Rizg
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdualziz University, Jeddah, Saudi Arabia
| | - Rana B Bakhaidar
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdualziz University, Jeddah, Saudi Arabia
| | - Walaa A Abualsunun
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdualziz University, Jeddah, Saudi Arabia
| | - Khaled M Hosny
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdualziz University, Jeddah, Saudi Arabia
| | - Abdulmohsin J Alamoudi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
Ibrahiem B, Shamma R, Salama A, Refai H. Magnetic targeting of lornoxicam/SPION bilosomes loaded in a thermosensitive in situ hydrogel system for the management of osteoarthritis: Optimization, in vitro, ex vivo, and in vivo studies in rat model via modulation of RANKL/OPG. Drug Deliv Transl Res 2024; 14:1982-2002. [PMID: 38158473 PMCID: PMC11153292 DOI: 10.1007/s13346-023-01503-8] [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] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
Osteoarthritis is a bone and joint condition characterized pathologically by articular cartilage degenerative damage and can develop into a devastating and permanently disabling disorder. This investigation aimed to formulate the anti-inflammatory drug lornoxicam (LOR) into bile salt-enriched vesicles loaded in an in situ forming hydrogel as a potential local treatment of osteoarthritis. This was achieved by formulating LOR-loaded bilosomes that are also loaded with superparamagnetic iron oxide nanoparticles (SPIONs) for intra-muscular (IM) administration to improve joint targeting and localization by applying an external magnet to the joint. A 31.22 full factorial design was employed to develop the bilosomal dispersions and the optimized formula including SPION (LSB) was loaded into a thermosensitive hydrogel. Moreover, in vivo evaluation revealed that the IM administration of LSB combined with the application of an external magnet to the joint reversed carrageen-induced suppression in motor activity and osteoprotegerin by significantly reducing the elevations in mitogen-activated protein kinases, extracellular signal-regulated kinase, and receptor activator of nuclear factor kappa beta/osteoprotegerin expressions. In addition, the histopathological evaluation of knee joint tissues showed a remarkable improvement in the injured joint tissues. The results proved that the developed LSB could be a promising IM drug delivery system for osteoarthritis management.
Collapse
Affiliation(s)
- Basma Ibrahiem
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt
| | - Rehab Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, El-Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Abeer Salama
- Department of Pharmacology, National Research Centre (NRC), Giza, 12622, Egypt
| | - Hanan Refai
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt.
| |
Collapse
|
6
|
Salem HF, Nafady MM, Eissa EM, Abdel-Sattar HH, Khallaf RA. Assembly of In-Situ Gel Containing Nano-Spanlastics of an Angiotensin II Inhibitor as a Novel Epitome for Hypertension Management: Factorial Design Optimization, In-vitro Gauging, Pharmacokinetics, and Pharmacodynamics Appraisal. AAPS PharmSciTech 2024; 25:115. [PMID: 38755324 DOI: 10.1208/s12249-024-02823-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
More than 1 billion people worldwide suffer from hypertension; therefore, hypertension management has been categorized as a global health priority. Losartan potassium (LP) is an antihypertensive drug with a limited oral bioavailability of about 33% since it undergoes the initial metabolic cycle. Thus, nasal administration is a unique route to overcome first-pass metabolism. The investigation focused on the potential effects of LP-loaded spanlastic vesicles (SNVs) on LP pharmacodynamics and pharmacokinetic parameters, utilizing a thin-film hydration methodology established on a 3122 full factorial design. Entrapment efficiency (EE%) ranged from 39.8 ± 3.87.8 to 83.8 ± 2.92% for LP-SNVs. Vesicle size (VS) varied from 205.5 ± 6.5.10 to 445.1 ± 13.52 nm, and the percentage of LP released after 8 h (Q8h) ranged from 30.8 ± 3.10 to 68.8 ± 1.45%. LP permeated through the nasal mucosa during 24 h and flocculated from 194.1 ± 4.90 to 435.3 ± 13.53 µg/cm2. After twenty-four hours, the optimal LP-SNVs in-situ gel showed 2.35 times more permeation through the nasal mucosa than the LP solution. It also lowered systolic blood pressure, so it is thought to be better than the reference formulation in terms of pharmacodynamics. The pharmacokinetics studies demonstrated that the intranasal LP-SNVs gel boosted its bioavailability approximately 6.36 times compared to the oral LP solution. Our research showed that intranasal LP-SNVs could be a good nanoplatform because they are well-tolerated and have possible pharmacokinetics and pharmacodynamics.
Collapse
Affiliation(s)
- Heba F Salem
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed M Nafady
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Essam M Eissa
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hend Hassan Abdel-Sattar
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Rasha A Khallaf
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
7
|
Gugleva V, Mihaylova R, Momekov G, Kamenova K, Forys A, Trzebicka B, Petrova M, Ugrinova I, Momekova D, Petrov PD. pH-responsive niosome-based nanocarriers of antineoplastic agents. RSC Adv 2024; 14:11124-11140. [PMID: 38606056 PMCID: PMC11008427 DOI: 10.1039/d4ra01334d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024] Open
Abstract
Differences in pH between the tumour interstitium and healthy tissues can be used to induce conformational changes in the nanocarrier structure, thereby triggering drug release at the desired site. In the present study, novel pH-responsive nanocarriers were developed by modifying conventional niosomes with hexadecyl-poly(acrylic acid)n copolymers (HD-PAAn). Niosomal vesicles were prepared by the thin film hydration method using Span 60, Span 60/Tween 60 and cholesterol as main constituents, and HD-PAA modifiers of different concentrations (0.5, 1, 2.5, 5 mol%). Next, two model substances, a water-soluble fluorescent dye (calcein) and a hydrophobic agent with pronounced antineoplastic activity (curcumin), were loaded in the aqueous core and hydrophobic membrane of the elaborated niosomes, respectively. Physicochemical properties of blank and loaded nanocarriers such as hydrodynamic diameter (Dh), size distribution, zeta potential, morphology and pH-responsiveness were investigated in detail. The cytotoxicity of niosomal curcumin was evaluated against human malignant cell lines of different origins (MJ, T-24, HUT-78), and the mechanistic aspects of proapoptotic effects were elucidated. The formulation composed of Span 60/Tween 60/cholesterol/2.5% HD-PAA17 exhibited optimal physicochemical characteristics (Dh 302 nm; ζ potential -22.1 mV; high curcumin entrapment 83%), pH-dependent drug release and improved cytotoxic and apoptogenic activity compared to free curcumin.
Collapse
Affiliation(s)
- Viliana Gugleva
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna "Prof. Dr Paraskev Stoyanov" 84 Tsar Osvoboditel Str. 9000 Varna Bulgaria
| | - Rositsa Mihaylova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia 2 Dunav Str. 1000 Sofia Bulgaria
| | - Georgi Momekov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia 2 Dunav Str. 1000 Sofia Bulgaria
| | - Katya Kamenova
- Institute of Polymers, Bulgarian Academy of Sciences bl.103 Akad. G. Bonchev Str.,1113 Sofia Bulgaria
| | - Aleksander Forys
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences ul. M. Curie-Skłodowskiej 34 Zabrze Poland
| | - Barbara Trzebicka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences ul. M. Curie-Skłodowskiej 34 Zabrze Poland
| | - Maria Petrova
- Institute of Molecular Biology "Akad. Roumen Tsanev", Bulgarian Academy of Sciences Acad. G. Bonchev str., bl 21 Sofia 1113 Bulgaria
| | - Iva Ugrinova
- Institute of Molecular Biology "Akad. Roumen Tsanev", Bulgarian Academy of Sciences Acad. G. Bonchev str., bl 21 Sofia 1113 Bulgaria
| | - Denitsa Momekova
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical University of Sofia 2 Dunav Str. 1000 Sofia Bulgaria
| | - Petar D Petrov
- Institute of Polymers, Bulgarian Academy of Sciences bl.103 Akad. G. Bonchev Str.,1113 Sofia Bulgaria
| |
Collapse
|
8
|
Roostaee M, Derakhshani A, Mirhosseini H, Banaee Mofakham E, Fathi-Karkan S, Mirinejad S, Sargazi S, Barani M. Composition, preparation methods, and applications of nanoniosomes as codelivery systems: a review of emerging therapies with emphasis on cancer. NANOSCALE 2024; 16:2713-2746. [PMID: 38213285 DOI: 10.1039/d3nr03495j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Nanoniosome-based drug codelivery systems have become popular therapeutic instruments, demonstrating tremendous promise in cancer therapy, infection treatment, and other therapeutic domains. An emerging form of vesicular nanocarriers, niosomes are self-assembling vesicles composed of nonionic surfactants, along with cholesterol or other amphiphilic molecules. This comprehensive review focuses on how nanosystems may aid in making anticancer and antibacterial pharmaceuticals more stable and soluble. As malleable nanodelivery instruments, the composition, types, preparation procedures, and variables affecting the structure and stability of niosomes are extensively investigated. In addition, the advantages of dual niosomes for combination therapy and the administration of multiple medications simultaneously are highlighted. Along with categorizing niosomal drug delivery systems, a comprehensive analysis of various preparation techniques, including thin-layer injection, ether injection, and microfluidization, is provided. Dual niosomes for cancer treatment are discussed in detail regarding the codelivery of two medications and the codelivery of a drug with organic, plant-based bioactive compounds or gene agents. In addition, niogelosomes and metallic niosomal carriers for targeted distribution are discussed. The review also investigates the simultaneous delivery of bioactive substances and gene agents, including siRNA, microRNA, shRNA, lncRNA, and DNA. Additional sections discuss the use of dual niosomes for cutaneous drug delivery and treating leishmanial infections, Pseudomonas aeruginosa, and Mycobacterium tuberculosis. The study concludes by delineating the challenges and potential routes for nanoniosome-based pharmaceutical codelivery systems, which will be useful for nanomedicine practitioners and researchers.
Collapse
Affiliation(s)
- Maryam Roostaee
- Department of Chemistry, Faculty of Sciences, Vali-e-Asr University of Rafsanjan, Rafsanjan, Iran.
| | - Atefeh Derakhshani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hadiseh Mirhosseini
- Department of Chemistry, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Elmira Banaee Mofakham
- Department of Nanotechnology and Advanced Materials Research, Materials & Energy Research Center, Karaj, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd 9414974877, Iran.
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 7616913555, Iran.
| |
Collapse
|
9
|
Fernandes SCM, Aguirre G. Biopolymer Micro/Nanogel Particles as Smart Drug Delivery and Theranostic Systems. Pharmaceutics 2023; 15:2060. [PMID: 37631274 PMCID: PMC10457921 DOI: 10.3390/pharmaceutics15082060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
In recent years, micro/nanogels have become an important topic of interdisciplinary research, especially in the fields of polymer chemistry and material science, with a focus on their use in drug delivery applications [...].
Collapse
Affiliation(s)
- Susana C. M. Fernandes
- CNRS, IPREM-UMR 5254, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64000 Pau, France
- MANTA—Marine Materials Research Group, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64600 Anglet, France
| | - Garbine Aguirre
- CNRS, IPREM-UMR 5254, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64000 Pau, France
- Bio-Inspired Materials Group: Functionalities & Self-Assembly, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64000 Pau, France
| |
Collapse
|
10
|
Omidian H, Wilson RL, Chowdhury SD. Enhancing Therapeutic Efficacy of Curcumin: Advances in Delivery Systems and Clinical Applications. Gels 2023; 9:596. [PMID: 37623051 PMCID: PMC10453486 DOI: 10.3390/gels9080596] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Curcumin, a potent active compound found in turmeric and Curcuma xanthorrhiza oil, possesses a wide range of therapeutic properties, including antibacterial, anti-inflammatory, antioxidant, and wound healing activities. However, its clinical effectiveness is hindered by its low bioavailability and rapid elimination from the body. To overcome these limitations, researchers have explored innovative delivery systems for curcumin. Some promising approaches include solid lipid nanoparticles, nanomicelle gels, and transdermal formulations for topical drug delivery. In the field of dentistry, curcumin gels have shown effectiveness against oral disorders and periodontal diseases. Moreover, Pickering emulsions and floating in situ gelling systems have been developed to target gastrointestinal health. Furthermore, curcumin-based systems have demonstrated potential in wound healing and ocular medicine. In addition to its therapeutic applications, curcumin also finds use as a food dye, contraception aid, corrosion-resistant coating, and environmentally friendly stain. This paper primarily focuses on the development of gel compositions of curcumin to address the challenges associated with its clinical use.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (R.L.W.); (S.D.C.)
| | | | | |
Collapse
|
11
|
Gopalakrishna PK, Jayaramu RA, Boregowda SS, Eshwar S, Suresh NV, Abu Lila AS, Moin A, Alotaibi HF, Obaidullah AJ, Khafagy ES. Piperine-Loaded In Situ Gel: Formulation, In Vitro Characterization, and Clinical Evaluation against Periodontitis. Gels 2023; 9:577. [PMID: 37504456 PMCID: PMC10378769 DOI: 10.3390/gels9070577] [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: 06/21/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
Periodontitis is an inflammatory disorder associated with dysbiosis and characterized by microbiologically related, host-mediated inflammation that leads to the damage of periodontal tissues including gingiva, connective tissues, and alveolar bone. The aim of this study was to develop an in situ gel consisting of piperine. Eight in situ gel formulations were designed by varying the concentration of deacylated gellan gum cross-linked with sodium tripolyphosphate, and poloxamer-407. The prepared gels were evaluated for gelation temperature, gelation time, viscosity, piperine-loading efficiency, and piperine release. Finally, the optimized formula was evaluated for anti-inflammatory effectiveness among human patients during a 14-day follow-up. The optimized in situ gel formulation exhibited a gelation temperature of 35 ± 1 °C, gelling of 36 ± 1 s, excellent syringeability, and piperine loading of 95.3 ± 2.3%. This formulation efficiently sustained in vitro drug release for up to 72 h. In vivo studies revealed an efficient sol-to-gel transformation of optimized in situ gel formulation at physiological conditions, permitting an efficient residence time of the formulation within a periodontitis pocket. Most importantly, a clinical study revealed that treatment with the optimized formulation elicited a significant reduction in the mean plaque score (p = 0.001), gingival index (p = 0.003), and pocket depth (p = 0.002), and exerted a potent anti-inflammatory potential, compared to the control group. Collectively, piperine-loaded in situ gel might represent a viable therapeutic approach for the management of gingival and periodontal diseases.
Collapse
Affiliation(s)
| | | | | | - Shruthi Eshwar
- KLE Society's Institute of Dental Sciences, Bengaluru 560022, India
| | - Nikhil V Suresh
- KLE Society's Institute of Dental Sciences, Bengaluru 560022, India
| | - Amr Selim Abu Lila
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Hadil Faris Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint AbdulRahman University, Riyadh 11671, Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
12
|
Fahmy S, Nasr S, Ramzy A, Dawood AS, Abdelnaser A, Azzazy HMES. Cytotoxic and Antioxidative Effects of Geranium Oil and Ascorbic Acid Coloaded in Niosomes against MCF-7 Breast Cancer Cells. ACS OMEGA 2023; 8:22774-22782. [PMID: 37396262 PMCID: PMC10308595 DOI: 10.1021/acsomega.3c01681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/12/2023] [Indexed: 07/04/2023]
Abstract
Geranium oil (GO) has antiproliferative, antiangiogenic, and anti-inflammatory properties. Ascorbic acid (AA) is reported to inhibit the formation of reactive oxygen species, sensitize cancer cells, and induce apoptosis. In this context, AA, GO, and AA-GO were loaded into niosomal nanovesicles to ameliorate the physicochemical properties of GO and improve its cytotoxic effects using the thin-film hydration technique. The prepared nanovesicles had a spherical shape with average diameters ranging from 200 to 300 nm and exhibited outstanding surface negative charges, high entrapment efficiencies, and a controlled sustained release over 72 h. Entrapping AA and GO in niosomes resulted in a lower IC50 value than free AA and GO when tested on MCF-7 breast cancer cells. In addition, flow cytometry analysis showed higher apoptotic cells in the late apoptotic stage upon treating the MCF-7 breast cancer cells with AA-GO niosomal vesicles compared to treatments with free AA, free GO, and AA or GO loaded into niosomal nanovesicles. Assessing the antioxidant effect of the free drugs and loaded niosomal nanovesicles showed enhanced antioxidant activity of AA-GO niosomal vesicles. These findings suggest the AA-GO niosomal vesicles as a potential treatment strategy against breast cancer, possibly through scavenging free radicals.
Collapse
Affiliation(s)
- Sherif
Ashraf Fahmy
- Chemistry
Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative
Capital, Cairo 11835, Egypt
- Department
of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Soad Nasr
- Institute
of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Asmaa Ramzy
- Department
of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Abdelhameed S. Dawood
- Institute
of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Anwar Abdelnaser
- Institute
of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
| | - Hassan Mohamed El-Said Azzazy
- Department
of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt
- Department
of Nanobiophotonics, Leibniz Institute for
Photonic Technology, Albert Einstein Str. 9, Jena 07745, Germany
| |
Collapse
|
13
|
Uboldi M, Perrotta C, Moscheni C, Zecchini S, Napoli A, Castiglioni C, Gazzaniga A, Melocchi A, Zema L. Insights into the Safety and Versatility of 4D Printed Intravesical Drug Delivery Systems. Pharmaceutics 2023; 15:pharmaceutics15030757. [PMID: 36986618 PMCID: PMC10057729 DOI: 10.3390/pharmaceutics15030757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
This paper focuses on recent advancements in the development of 4D printed drug delivery systems (DDSs) for the intravesical administration of drugs. By coupling the effectiveness of local treatments with major compliance and long-lasting performance, they would represent a promising innovation for the current treatment of bladder pathologies. Being based on a shape-memory pharmaceutical-grade polyvinyl alcohol (PVA), these DDSs are manufactured in a bulky shape, can be programmed to take on a collapsed one suitable for insertion into a catheter and re-expand inside the target organ, following exposure to biological fluids at body temperature, while releasing their content. The biocompatibility of prototypes made of PVAs of different molecular weight, either uncoated or coated with Eudragit®-based formulations, was assessed by excluding relevant in vitro toxicity and inflammatory response using bladder cancer and human monocytic cell lines. Moreover, the feasibility of a novel configuration was preliminarily investigated, targeting the development of prototypes provided with inner reservoirs to be filled with different drug-containing formulations. Samples entailing two cavities, filled during the printing process, were successfully fabricated and showed, in simulated urine at body temperature, potential for controlled release, while maintaining the ability to recover about 70% of their original shape within 3 min.
Collapse
Affiliation(s)
- Marco Uboldi
- Sezione di Tecnologia e Legislazione Farmaceutiche “Maria Edvige Sangalli”, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via Giuseppe Colombo 71, 20133 Milano, Italy
| | - Cristiana Perrotta
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, via Giovanni Battista Grassi 74, 20157 Milano, Italy
| | - Claudia Moscheni
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, via Giovanni Battista Grassi 74, 20157 Milano, Italy
| | - Silvia Zecchini
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, via Giovanni Battista Grassi 74, 20157 Milano, Italy
| | - Alessandra Napoli
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, via Giovanni Battista Grassi 74, 20157 Milano, Italy
| | - Chiara Castiglioni
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Andrea Gazzaniga
- Sezione di Tecnologia e Legislazione Farmaceutiche “Maria Edvige Sangalli”, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via Giuseppe Colombo 71, 20133 Milano, Italy
| | - Alice Melocchi
- Sezione di Tecnologia e Legislazione Farmaceutiche “Maria Edvige Sangalli”, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via Giuseppe Colombo 71, 20133 Milano, Italy
- Correspondence: ; Tel.: +39-02-50324654
| | - Lucia Zema
- Sezione di Tecnologia e Legislazione Farmaceutiche “Maria Edvige Sangalli”, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via Giuseppe Colombo 71, 20133 Milano, Italy
| |
Collapse
|
14
|
Development of potent nanosized carbonic anhydrase inhibitor for targeted therapy of hypoxic solid tumors. Int J Pharm 2023; 631:122537. [PMID: 36572260 DOI: 10.1016/j.ijpharm.2022.122537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Overexpression of two carbonic anhydrase (CA) isoforms, CA IX and XII, in several hypoxic solid tumors provides an extracellular hypoxic microenvironment, interferes with extra- and intracellular pH regulation, thus favoring hypoxic tumor cell survival, proliferation and metastasis. In the current study, a selective inhibitor for human CA isoforms IX and XII (isatin-bearing sulfonamide, WEG-104), was incorporated into nanosized spherical niosomes at high encapsulation efficiency to allow for an enhanced and sustained antitumor activity. In vivo, administration of WEG-104 that is either free (10 mg/kg) or loaded into niosomes (5 mg/kg) into a mice model of Ehrlich ascites solid tumor resulted in comparable efficacy in terms of reduction of tumor weight and volume. Administration of WEG-104-loaded niosomes (10 mg/kg) exhibited superior antitumor activity compared to the free drug, evidenced by reduced tumor weight and volume, marked reduction in the activity of CA IX and XII, and suppression of HIF-1α and MMP-2. Moreover, prominent increase of caspase 3 and pronounced decrease in VEGF immune expression were observed in the treated animals. Hence, loading of molecularly designed compounds that targets CAs in hypoxic solid tumors into nanosized delivery systems provided an auspicious strategy for limiting solid tumor progression and malignancy.
Collapse
|
15
|
Pourmadadi M, Abbasi P, Eshaghi MM, Bakhshi A, Ezra Manicum AL, Rahdar A, Pandey S, Jadoun S, Díez-Pascual AM. Curcumin delivery and co-delivery based on nanomaterials as an effective approach for cancer therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
16
|
Fahmy SA, Ramzy A, Sawy AM, Nabil M, Gad MZ, El-Shazly M, Aboul-Soud MAM, Azzazy HMES. Ozonated Olive Oil: Enhanced Cutaneous Delivery via Niosomal Nanovesicles for Melanoma Treatment. Antioxidants (Basel) 2022; 11:antiox11071318. [PMID: 35883809 PMCID: PMC9312098 DOI: 10.3390/antiox11071318] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/27/2023] Open
Abstract
Ozonated olive oil (OL) combines the therapeutic effects of both ozone and olive oil. However, it suffers from limited water solubility and poor transdermal permeation, which hinder its application in melanoma treatment. Nanocarrier host molecules, such as niosomes, were used to improve the water solubility, transdermal permeation, and anticancer effect of hydrophobic compounds. This study aims to design and optimize a niosomal vesicular nanoplatform loaded with OL (OL/NSs) to improve OL’s skin permeation and anti-melanoma effect. In this regard, OL was prepared and characterized by evaluating its chemical properties (acid, peroxide, and iodine values) and fatty acid composition using gas chromatography. Then, OL/NSs were developed using the thin film hydration method employing cholesterol, Span 60, and Tween 60 at five different molar ratios. The optimized niosomes had an average diameter of 125.34 ± 13.29 nm, a surface charge of −11.34 ± 4.71 mV, and a spherical shape. They could entrap 87.30 ± 4.95% of the OL. OL/NSs showed a 75% sustained oil release over 24 h. The skin permeation percentage of OL/NSs was 36.78 ± 3.31 and 53.44 ± 6.41% at 12 and 24 h, respectively, three times higher than that of the free OL (11.50 ± 1.3 and 17.24 ± 2.06%, at 12 and 24 h, respectively). Additionally, the anticancer activity of the developed niosmal formulation, when tested on human melanoma cells (A375), was double that of the free OL; the IC50 of the OL/NSs was 8.63 ± 2.8 μg/mL, and that of the free OL was 17.4 ± 3.7 μg/mL. In conclusion, the encapsulation of ozonated olive oil in niosomes enhanced its water solubility, skin permeation, and anticancer activity and thus may represent potent natural chemotherapy in treating melanoma.
Collapse
Affiliation(s)
- Sherif Ashraf Fahmy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt; (S.A.F.); (A.R.); (A.M.S.); (M.N.)
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative Capital, AL109AB, Cairo 11835, Egypt
| | - Asmaa Ramzy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt; (S.A.F.); (A.R.); (A.M.S.); (M.N.)
| | - Amany M. Sawy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt; (S.A.F.); (A.R.); (A.M.S.); (M.N.)
- Department of Physics, Faculty of Science, Fayoum University, Fayoum 63514, Egypt
| | - Mohamed Nabil
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt; (S.A.F.); (A.R.); (A.M.S.); (M.N.)
| | - Mohamed Z. Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, The German University in Cairo, Cairo 11835, Egypt;
| | - Mohamed El-Shazly
- Pharmacognosy Department, Faculty of Pharmacy, Ain-Shams University, Organization of African Unity Street, Abassia, Cairo 11566, Egypt;
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, The German University in Cairo, Cairo 11835, Egypt
| | - Mourad A. M. Aboul-Soud
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
- Biochemistry Department, Cairo University Research Park, Cairo University, Giza 12613, Egypt
| | - Hassan Mohamed El-Said Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt; (S.A.F.); (A.R.); (A.M.S.); (M.N.)
- Correspondence: ; Tel.: +20-2-2615-2559
| |
Collapse
|
17
|
Yasamineh S, Yasamineh P, Ghafouri Kalajahi H, Gholizadeh O, Yekanipour Z, Afkhami H, Eslami M, Hossein Kheirkhah A, Taghizadeh M, Yazdani Y, Dadashpour M. A state-of-the-art review on the recent advances of niosomes as a targeted drug delivery system. Int J Pharm 2022; 624:121878. [PMID: 35636629 DOI: 10.1016/j.ijpharm.2022.121878] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/14/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023]
Abstract
The buildup of nonionic surfactants in the aqueous environment produces niosomes. The usage of niosomes is becoming increasingly frequent due to their sustainability, low cost of components and assembly, large-scale manufacture, and, finally, easy maintenance of the niosomes to the other. Because of their nonionic characteristics, niosomes play a critical role in medication delivery systems. Controlled release and targeted distribution of niosomes to treat cancer, infectious illnesses, and other disorders are one of their most important properties. Niosomes can also be injected by ocular and transdermal routes, which are less common than oral and parenteral administration. Using niosomes to manufacture biotechnology goods and novel vaccines is one of the most exciting research fields today. The molecular structure of niosomes, the physicochemical characteristics of nonionic surfactants in their formulation, the influence of external stimuli on niosomes, the many methods of niosomes administration, and their diverse therapeutic qualities are all explored in this study.
Collapse
Affiliation(s)
- Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Pooneh Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Omid Gholizadeh
- Department of Virology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Yekanipour
- Department of Microbiology, Marand Branch, Islamic Azad University, Marand, Iran
| | - Hamed Afkhami
- Department of Medical Microbiology, Faculty of Medicine, Shahed University of Medical Science, Tehran, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Amir Hossein Kheirkhah
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Milad Taghizadeh
- Department of Laboratory Sciences, Faculty of Paramedical, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Yazdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|