Generic placeholder image

Pharmaceutical Nanotechnology

Editor-in-Chief

ISSN (Print): 2211-7385
ISSN (Online): 2211-7393

Review Article

A Comprehensive Review on Niosomes as a Tool for Advanced Drug Delivery

Author(s): Shivani Sharma, Akash Garg*, Rutvi Agrawal, Himansu Chopra and Devender Pathak

Volume 12, Issue 3, 2024

Published on: 08 September, 2023

Page: [206 - 228] Pages: 23

DOI: 10.2174/2211738511666230726154557

Price: $65

Abstract

Over the past few decades, advancements in nanocarrier-based therapeutic delivery have been significant, and niosomes research has recently received much interest. The self-assembled nonionic surfactant vesicles lead to the production of niosomes. The most recent nanocarriers, niosomes, are self-assembled vesicles made of nonionic surfactants with or without the proper quantities of cholesterol or other amphiphilic molecules. Because of their durability, low cost of components, largescale production, simple maintenance, and high entrapment efficiency, niosomes are being used more frequently. Additionally, they enhance pharmacokinetics, reduce toxicity, enhance the solubility of poorly water-soluble compounds, & increase bioavailability. One of the most crucial features of niosomes is their controlled release and targeted diffusion, which is utilized for treating cancer, infectious diseases, and other problems. In this review article, we have covered all the fundamental information about niosomes, including preparation techniques, niosomes types, factors influencing their formation, niosomes evaluation, applications, and administration routes, along with recent developments.

Keywords: Niosomes, non-ionic surfactants, proniosomes, bioavailability, targeted delivery, cholesterol.

Graphical Abstract
[1]
Kumar GP, Rajeshwarrao P. Nonionic surfactant vesicular systems for effective drug delivery—an overview. Acta Pharm Sin B 2011; 1(4): 208-19.
[http://dx.doi.org/10.1016/j.apsb.2011.09.002]
[2]
Kuotsu K, Karim KM, Mandal AS, et al. Niosome: A future of targeted drug delivery systems. J Adv Pharm Technol Res 2010; 1(4): 374-80.
[http://dx.doi.org/10.4103/0110-5558.76435] [PMID: 22247876]
[3]
Roy Biswas G, Majee S. Niosomes in ocular drug delivery. Eur J Pharm Med Res 2017; 4: 813-9.
[4]
Abdelkader H, Alani AWG, Alany RG. Recent advances in non-ionic surfactant vesicles (niosomes): Self-assembly, fabrication, characterization, drug delivery applications and limitations. Drug Deliv 2014; 21(2): 87-100.
[http://dx.doi.org/10.3109/10717544.2013.838077] [PMID: 24156390]
[5]
Rajera R, Nagpal K, Singh SK, Mishra DN. Niosomes: A controlled and novel drug delivery system. Biol Pharm Bull 2011; 34(7): 945-53.
[http://dx.doi.org/10.1248/bpb.34.945] [PMID: 21719996]
[6]
Mahale NB, Thakkar PD, Mali RG, Walunj DR, Chaudhari SR. Niosomes: Novel sustained release nonionic stable vesicular systems - An overview. Adv Colloid Interface Sci 2012; 183-184: 46-54.
[http://dx.doi.org/10.1016/j.cis.2012.08.002] [PMID: 22947187]
[7]
Marianecci C, Di Marzio L, Rinaldi F, et al. Niosomes from 80s to present: The state of the art. Adv Colloid Interface Sci 2014; 205: 187-206.
[http://dx.doi.org/10.1016/j.cis.2013.11.018] [PMID: 24369107]
[8]
Bhardwaj P, Tripathi P, Gupta R, Pandey S. Niosomes: A review on niosomal research in the last decade. J Drug Deliv Sci Technol 2020; 56: 101581.
[http://dx.doi.org/10.1016/j.jddst.2020.101581]
[9]
Ag Seleci D, Seleci M, Walter JG, Stahl F, Scheper T. Niosomes as nanoparticular drug carriers: Fundamentals and recent applications. J Nanomater 2016; 2016: 1-13.
[http://dx.doi.org/10.1155/2016/7372306]
[10]
Pham TT, Jaafar-Maalej C, Charcosset C, Fessi H. Liposome and niosome preparation using a membrane contactor for scale-up. Colloids Surf B Biointerfaces 2012; 94: 15-21.
[http://dx.doi.org/10.1016/j.colsurfb.2011.12.036] [PMID: 22326648]
[11]
Mehta SK, Jindal N. Formulation of Tyloxapol niosomes for encapsulation, stabilization and dissolution of anti-tubercular drugs. Colloids Surf B Biointerfaces 2013; 101: 434-41.
[http://dx.doi.org/10.1016/j.colsurfb.2012.07.006] [PMID: 23010052]
[12]
Chen S, Hanning S, Falconer J, Locke M, Wen J. Recent advances in non-ionic surfactant vesicles (niosomes): Fabrication, characterization, pharmaceutical and cosmetic applications. Eur J Pharm Biopharm 2019; 144: 18-39.
[http://dx.doi.org/10.1016/j.ejpb.2019.08.015] [PMID: 31446046]
[13]
Azmin MN, Florence AT, Handjani-Vila RM, Stuart J F B, Vanlerberghe G, Whittaker JS. The effect of non-ionic surfactant vesicle (niosome) entrapment on the absorption and distribution of methotrexate in mice. J Pharm Pharmacol 2011; 37(4): 237-42.
[http://dx.doi.org/10.1111/j.2042-7158.1985.tb05051.x] [PMID: 2860220]
[14]
Sahin NO. Niosomes as nanocarrier systems. In: Mozafari MR, Ed. Nanomaterials and Nanosystems for Biomedical Applications. Dordrecht: Springer Netherlands 2007; pp. 67-81.
[http://dx.doi.org/10.1007/978-1-4020-6289-6_4]
[15]
Moghassemi S, Hadjizadeh A. Nano-niosomes as nanoscale drug delivery systems: An illustrated review. J Control Release 2014; 185: 22-36.
[http://dx.doi.org/10.1016/j.jconrel.2014.04.015] [PMID: 24747765]
[16]
Kaur IP, Garg A, Singla AK, Aggarwal D. Vesicular systems in ocular drug delivery: An overview. Int J Pharm 2004; 269(1): 1-14.
[http://dx.doi.org/10.1016/j.ijpharm.2003.09.016] [PMID: 14698571]
[17]
Sudershan B, Murthy PN, Sahu J, Sahoo P, Amir F. Vesicles of non-ionic surfactants (Niosomes) and drug delivery potential. Int J Pharmaceut Sci Nanotechnol 2008; 1(1)
[18]
Pardakhty A, Moazeni E. Nano-niosomes in drug, vaccine and gene delivery: A rapid overview. Nanomed J 2012; 1: 1-13.
[19]
Steed JW, Turner DR, Wallace KJ. Core concepts in supramolecular chemistry and nanochemistry. John Wiley & Sons 2007.
[20]
Zeng W, Li Q, Wan T, et al. Hyaluronic acid-coated niosomes facilitate tacrolimus ocular delivery: Mucoadhesion, precorneal retention, aqueous humor pharmacokinetics, and transcorneal permeability. Colloids Surf B Biointerfaces 2016; 141: 28-35.
[http://dx.doi.org/10.1016/j.colsurfb.2016.01.014] [PMID: 26820107]
[21]
Yuksel N, Bayindir ZS, Aksakal E, Ozcelikay AT. In situ niosome forming maltodextrin proniosomes of candesartan cilexetil: In vitro and In vivo evaluations. Int J Biol Macromol 2016; 82: 453-63.
[http://dx.doi.org/10.1016/j.ijbiomac.2015.10.019] [PMID: 26455402]
[22]
Mokhtar M, Sammour OA, Hammad MA, Megrab NA. Effect of some formulation parameters on flurbiprofen encapsulation and release rates of niosomes prepared from proniosomes. Int J Pharm 2008; 361(1-2): 104-11.
[http://dx.doi.org/10.1016/j.ijpharm.2008.05.031] [PMID: 18577437]
[23]
Elhissi A, Hidayat K, Phoenix DA, et al. Air-jet and vibrating-mesh nebulization of niosomes generated using a particulate-based proniosome technology. Int J Pharm 2013; 444(1-2): 193-9.
[http://dx.doi.org/10.1016/j.ijpharm.2012.12.040] [PMID: 23299083]
[24]
Abd-Elbary A, El-laithy HM, Tadros MI. Sucrose stearate-based proniosome-derived niosomes for the nebulisable delivery of cromolyn sodium. Int J Pharm 2008; 357(1-2): 189-98.
[http://dx.doi.org/10.1016/j.ijpharm.2008.01.056] [PMID: 18339494]
[25]
Ibrahim MM, Shehata TM. The enhancement of transdermal permeability of water soluble drug by niosome-emulgel combination. J Drug Deliv Sci Technol 2012; 22(4): 353-9.
[http://dx.doi.org/10.1016/S1773-2247(12)50059-6]
[26]
Csongradi C, du Plessis J, Aucamp ME, Gerber M. Topical delivery of roxithromycin solid-state forms entrapped in vesicles. Eur J Pharm Biopharm 2017; 114: 96-107.
[http://dx.doi.org/10.1016/j.ejpb.2017.01.006] [PMID: 28119103]
[27]
Manosroi A, Jantrawut P, Manosroi J. Anti-inflammatory activity of gel containing novel elastic niosomes entrapped with diclofenac diethylammonium. Int J Pharm 2008; 360(1-2): 156-63.
[http://dx.doi.org/10.1016/j.ijpharm.2008.04.033] [PMID: 18539416]
[28]
Manosroi A, Chankhampan C, Manosroi W, Manosroi J. Transdermal absorption enhancement of papain loaded in elastic niosomes incorporated in gel for scar treatment. Eur J Pharm Sci 2013; 48(3): 474-83.
[http://dx.doi.org/10.1016/j.ejps.2012.12.010] [PMID: 23266464]
[29]
Abdelkader H, Ismail S, Kamal A, Alany RG. Design and evaluation of controlled-release niosomes and discomes for naltrexone hydrochloride ocular delivery. J Pharm Sci 2011; 100(5): 1833-46.
[http://dx.doi.org/10.1002/jps.22422] [PMID: 21246556]
[30]
Abdelkader H, Wu Z, Al-Kassas R, Alany RG. Niosomes and discomes for ocular delivery of naltrexone hydrochloride: Morphological, rheological, spreading properties and photo-protective effects. Int J Pharm 2012; 433(1-2): 142-8.
[http://dx.doi.org/10.1016/j.ijpharm.2012.05.011] [PMID: 22595640]
[31]
Paolino D, Cosco D, Muzzalupo R, Trapasso E, Picci N, Fresta M. Innovative bola-surfactant niosomes as topical delivery systems of 5-fluorouracil for the treatment of skin cancer. Int J Pharm 2008; 353(1-2): 233-42.
[http://dx.doi.org/10.1016/j.ijpharm.2007.11.037] [PMID: 18191509]
[32]
Saini N, Sodhi RK, Bajaj L, et al. Intravaginal administration of metformin hydrochloride loaded cationic niosomes amalgamated with thermosensitive gel for the treatment of polycystic ovary syndrome: In vitro and in vivo studies. Colloids Surf B Biointerfaces 2016; 144: 161-9.
[http://dx.doi.org/10.1016/j.colsurfb.2016.04.016] [PMID: 27085048]
[33]
Gopinath D, Ravi D, Rao BR, Apte SS, Renuka D, Rambhau D. Ascorbyl palmitate vesicles (Aspasomes): Formation, characterization and applications. Int J Pharm 2004; 271(1-2): 95-113.
[http://dx.doi.org/10.1016/j.ijpharm.2003.10.032] [PMID: 15129977]
[34]
Hu C, Rhodes DG. Proniosomes: A novel drug carrier preparation. Int J Pharm 2000; 206(1-2): 109-22.
[http://dx.doi.org/10.1016/S0378-5173(00)00513-5] [PMID: 11202988]
[35]
Uchegbu IF, Schätzlein A, Vanlerberghe G, Morgatini N, Florence AT. Polyhedral non-ionic surfactant vesicles. J Pharm Pharmacol 2011; 49(6): 606-10.
[http://dx.doi.org/10.1111/j.2042-7158.1997.tb06853.x] [PMID: 9330200]
[36]
Uchegbu IF, Vyas SP. Non-ionic surfactant based vesicles (niosomes) in drug delivery. Int J Pharm 1998; 172(1-2): 33-70.
[http://dx.doi.org/10.1016/S0378-5173(98)00169-0]
[37]
Uchegbu IF, Florence AT. Non-ionic surfactant vesicles (niosomes): Physical and pharmaceutical chemistry. Adv Colloid Interface Sci 1995; 58(1): 1-55.
[http://dx.doi.org/10.1016/0001-8686(95)00242-I]
[38]
Laouini A, Jaafar-Maalej C, Sfar S, Charcosset C, Fessi H. Liposome preparation using a hollow fiber membrane contactor—Application to spironolactone encapsulation. Int J Pharm 2011; 415(1-2): 53-61.
[http://dx.doi.org/10.1016/j.ijpharm.2011.05.034] [PMID: 21641982]
[39]
He RX, Ye X, Li R, et al. PEGylated niosomes-mediated drug delivery systems for Paeonol: Preparation, pharmacokinetics studies and synergistic anti-tumor effects with 5-FU. J Liposome Res 2017; 27(2): 161-70.
[http://dx.doi.org/10.1080/08982104.2016.1191021] [PMID: 27184460]
[40]
Lin T, Fang Q, Peng D, et al. PEGylated non-ionic surfactant vesicles as drug delivery systems for Gambogenic acid. Drug Deliv 2013; 20(7): 277-84.
[http://dx.doi.org/10.3109/10717544.2013.836618] [PMID: 24044645]
[41]
Alemi A, Zavar Reza J, Haghiralsadat F, et al. Paclitaxel and curcumin coadministration in novel cationic PEGylated niosomal formulations exhibit enhanced synergistic antitumor efficacy. J Nanobiotechnol 2018; 16(1): 28.
[http://dx.doi.org/10.1186/s12951-018-0351-4] [PMID: 29571289]
[42]
Manosroi A, Chutoprapat R, Abe M, Manosroi J. Characteristics of niosomes prepared by supercritical carbon dioxide (scCO2) fluid. Int J Pharm 2008; 352(1-2): 248-55.
[http://dx.doi.org/10.1016/j.ijpharm.2007.10.013] [PMID: 18036754]
[43]
Bayindir ZS, Yuksel N. Characterization of niosomes prepared with various nonionic surfactants for paclitaxel oral delivery. J Pharm Sci 2010; 99(4): 2049-60.
[http://dx.doi.org/10.1002/jps.21944] [PMID: 19780133]
[44]
Gandhi A, Sen SO, Paul A. Current trends in niosome as vesicular drug delivery system. Asian J Pharm Life Sci 2012; 2(2): 339-53.
[45]
Baillie AJ, Florence AT, Hume LR, Muirhead GT, Rogerson A. The preparation and properties of niosomes—non-ionic surfactant vesicles. J Pharm Pharmacol 2011; 37(12): 863-8.
[http://dx.doi.org/10.1111/j.2042-7158.1985.tb04990.x] [PMID: 2868092]
[46]
Isnan AP, Jufri M. Formulation of niosomal gel containing green tea extract (Camellia sinensis L. Kuntze) using thin-layer hydration. Int J App Pharmaceut 2017; 9: 38.
[http://dx.doi.org/10.22159/ijap.2017.v9s1.23_28]
[47]
Onochie ITO, Nwakile C, Umeyor C, et al. Formulation and evaluation of niosomes of benzyl penicillin. J Appl Pharm Sci 2013; 3: 66-71.
[48]
Manosroi A, Khanrin P, Lohcharoenkal W, et al. Transdermal absorption enhancement through rat skin of gallidermin loaded in niosomes. Int J Pharm 2010; 392(1-2): 304-10.
[http://dx.doi.org/10.1016/j.ijpharm.2010.03.064] [PMID: 20381599]
[49]
Bini KB, Dubey A, Prabhu P, Kamath J. Development and characterization of non-ionic surfactant vesicles (niosomes) for oral delivery of lornoxicam. Int J Drug Develop Res 2012; 4: 157-64.
[50]
Gaafar PME, Abdallah OY, Farid RM, Abdelkader H. Preparation, characterization and evaluation of novel elastic nano-sized niosomes (ethoniosomes) for ocular delivery of prednisolone. J Liposome Res 2014; 24(3): 204-15.
[http://dx.doi.org/10.3109/08982104.2014.881850] [PMID: 24484536]
[51]
Ruckmani K, Sankar V. Formulation and optimization of Zidovudine niosomes. AAPS PharmSciTech 2010; 11(3): 1119-27.
[http://dx.doi.org/10.1208/s12249-010-9480-2] [PMID: 20635228]
[52]
Kamboj S, Saini V, Bala S. Formulation and characterization of drug loaded nonionic surfactant vesicles (niosomes) for oral bioavailability enhancement. ScientificWorldJournal 2014; 2014: 1-8.
[http://dx.doi.org/10.1155/2014/959741] [PMID: 24672401]
[53]
Umbarkar MG. Niosome as a novel pharmaceutical drug delivery: A brief review highlighting formulation, types, composition and application. Indian J Pharmaceut Edu Res 2021; 55(1s): s11-28.
[http://dx.doi.org/10.5530/ijper.55.1s.34]
[54]
Marwa A, Sammour O, Hanaa E, Mohammed A. Preparation and in-vitro evaluation of diclofenac sodium niosomal formulations. Int J Pharm Sci Res 2013; 4: 1757-65.
[55]
Verma S, Singh S, Syan N, Mathur P, Valecha V. Nanoparticle vesicular systems: A versatile tool for drug delivery. J Chem Pharm Res 2010; 2: 496-509.
[56]
Shreedevi HM, Jino Nesalin JA, Mani TT. Development and evaluation of stavudine niosome by ether injection method. Int J Pharma Sci Res 2016; 7(1): 38-46.
[57]
Bansal S, Aggarwal G, Chandel P, Harikumar SL. Design and development of cefdinir niosomes for oral delivery. J Pharm Bioallied Sci 2013; 5(4): 318-25.
[http://dx.doi.org/10.4103/0975-7406.120080] [PMID: 24302841]
[58]
Moghassemi S, Hadjizadeh A, Omidfar K. Formulation and characterization of bovine serum albumin-loaded niosome. AAPS PharmSciTech 2017; 18(1): 27-33.
[http://dx.doi.org/10.1208/s12249-016-0487-1] [PMID: 26817764]
[59]
Anchal SPP. Recent trends in niosome as vesicular drug delivery system. J Appl Pharm Sci 2012; 2(6): 20-32.
[60]
Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. Niosome: A promising nanocarrier for natural drug delivery through blood-brain barrier. Adv Pharmacol Sci 2018; 2018: 1-15.
[http://dx.doi.org/10.1155/2018/6847971] [PMID: 30651728]
[61]
Singh G, Dwivedi H, Saraf SK, Saraf SA. Niosomal delivery of isoniazid - development and characterization. Trop J Pharm Res 2011; 10(2): 203-10.
[http://dx.doi.org/10.4314/tjpr.v10i2.66564]
[62]
Junyaprasert VB, Singhsa P, Suksiriworapong J, Chantasart D. Physicochemical properties and skin permeation of Span 60/Tween 60 niosomes of ellagic acid. Int J Pharm 2012; 423(2): 303-11.
[http://dx.doi.org/10.1016/j.ijpharm.2011.11.032] [PMID: 22155414]
[63]
Sahoo RK, Biswas N, Guha A, Sahoo N, Kuotsu K. Nonionic surfactant vesicles in ocular delivery: Innovative approaches and perspectives. BioMed Res Int 2014; 2014: 1-12.
[http://dx.doi.org/10.1155/2014/263604] [PMID: 24995280]
[64]
Mayer LD, Bally MB, Hope MJ, Cullis PR. Uptake of antineoplastic agents into large unilamellar vesicles in response to a membrane potential. Biochim Biophys Acta Biomembr 1985; 816(2): 294-302.
[http://dx.doi.org/10.1016/0005-2736(85)90497-3] [PMID: 3839135]
[65]
Martin JF. Pharmaceutical manufacturing of liposomes. New York: Marcel Dekker 1990.
[66]
Manosroi A, Ruksiriwanich W, Abe M, Sakai H, Manosroi W, Manosroi J. Biological activities of the rice bran extract and physical characteristics of its entrapment in niosomes by supercritical carbon dioxide fluid. J Supercrit Fluids 2010; 54(2): 137-44.
[http://dx.doi.org/10.1016/j.supflu.2010.05.002]
[67]
Sanklecha VM, Pande VV, Pawar SS, Pagar OB, Jadhav AC. Review on Niosomes. Austin Pharmacol Pharm 2018; 3(2): 1016.
[68]
Debnath A, Kumar A. Structural and functional significance of niosome and proniosome in drug delivery system. A Debnath 2022; 3(3): 621-37.
[69]
Radhi AA. Benazepril hydrochloride loaded niosomal formulation for oral delivery: Formulation and characterization. Int J App Pharma 2018; 10(5): 66.
[http://dx.doi.org/10.22159/ijap.2018v10i5.27564]
[70]
Raymond CR, Paul JS, Sian CO. Polyoxyethylene Alkyl ethers. (5th ed.). London: Pharmaceutical Press 2006; p. 8.
[71]
Manosroi A, Wongtrakul P, Manosroi J, et al. Characterization of vesicles prepared with various non-ionic surfactants mixed with cholesterol. Colloids Surf B Biointerfaces 2003; 30(1-2): 129-38.
[http://dx.doi.org/10.1016/S0927-7765(03)00080-8]
[72]
Bouwstra JA, van Hal DA, Hofland HEJ, Junginger HE. Preparation and characterization of nonionic surfactant vesicles. Colloids Surf A Physicochem Eng Asp 1997; 123-124: 71-80.
[http://dx.doi.org/10.1016/S0927-7757(96)03800-9]
[73]
Yeo LK, Chaw CS, Elkordy AA. The effects of hydration parameters and co-surfactants on methylene blue-loaded niosomes prepared by the thin film hydration method. Pharmaceuticals 2019; 12(2): 46.
[http://dx.doi.org/10.3390/ph12020046]
[74]
Hao Y, Zhao F, Li N, Yang Y, Li K. Studies on a high encapsulation of colchicine by a niosome system. Int J Pharm 2002; 244(1-2): 73-80.
[http://dx.doi.org/10.1016/S0378-5173(02)00301-0] [PMID: 12204566]
[75]
Marianecci C, Paolino D, Celia C, Fresta M, Carafa M, Alhaique F. Non-ionic surfactant vesicles in pulmonary glucocorticoid delivery: Characterization and interaction with human lung fibroblasts. J Control Release 2010; 147(1): 127-35.
[http://dx.doi.org/10.1016/j.jconrel.2010.06.022] [PMID: 20603167]
[76]
Arunothayanun P, Bernard MS, Craig DQM, Uchegbu IF, Florence AT. The effect of processing variables on the physical characteristics of non-ionic surfactant vesicles (niosomes) formed from a hexadecyl diglycerol ether. Int J Pharm 2000; 201(1): 7-14.
[http://dx.doi.org/10.1016/S0378-5173(00)00362-8] [PMID: 10867260]
[77]
Rogerson A, Cummings J, Willmott N, Florence AT. The distribution of doxorubicin in mice following administration in niosomes. J Pharm Pharmacol 2011; 40(5): 337-42.
[http://dx.doi.org/10.1111/j.2042-7158.1988.tb05263.x] [PMID: 2899629]
[78]
Bartelds R, Nematollahi MH, Pols T, et al. Niosomes, an alternative for liposomal delivery. PLoS One 2018; 13(4): e0194179.
[http://dx.doi.org/10.1371/journal.pone.0194179] [PMID: 29649223]
[79]
Parthasarathi G, Udupa N, Umadevi P, Pillai G. Niosome encapsulated of vincristine sulfate: Improved anticancer activity with reduced toxicity in mice. J Drug Target 1994; 2(2): 173-82.
[http://dx.doi.org/10.3109/10611869409015907] [PMID: 8069596]
[80]
Agarwal S, Mohamed MS, Raveendran S, Rochani AK, Maekawa T, Kumar DS. Formulation, characterization and evaluation of morusin loaded niosomes for potentiation of anticancer therapy. RSC Adv 2018; 8(57): 32621-36.
[http://dx.doi.org/10.1039/C8RA06362A] [PMID: 35547672]
[81]
Aboul-Einien MH, Kandil SM, Abdou EM, Diab HM, Zaki MSE. Ascorbic acid derivative-loaded modified aspasomes: Formulation, In vitro, ex vivo and clinical evaluation for melasma treatment. J Liposome Res 2020; 30(1): 54-67.
[http://dx.doi.org/10.1080/08982104.2019.1585448] [PMID: 30821553]
[82]
Rinaldi F, Hanieh P, Chan L, et al. Chitosan glutamate-coated niosomes: A proposal for nose-to-brain delivery. Pharmaceutics 2018; 10(2): 38.
[http://dx.doi.org/10.3390/pharmaceutics10020038] [PMID: 29565809]
[83]
Manvi SR, Gupta VRM, Srikanth K, Devanna N. Formulation and evaluation of candesartan niosomal suspension. Res J Pharma Technol 2012; 5(12): 1570-2.
[84]
Vyas S, Singh R, Jain S, et al. Non-ionic surfactant based vesicles (niosomes) for non-invasive topical genetic immunization against hepatitis B. Int J Pharm 2005; 296(1-2): 80-6.
[http://dx.doi.org/10.1016/j.ijpharm.2005.02.016] [PMID: 15885458]
[85]
Gupta PN, Mishra V, Rawat A, et al. Non-invasive vaccine delivery in transfersomes, niosomes and liposomes: A comparative study. Int J Pharm 2005; 293(1-2): 73-82.
[http://dx.doi.org/10.1016/j.ijpharm.2004.12.022] [PMID: 15778046]
[86]
Singh P, Prabakaran D, Jain S, Mishra V, Jaganathan KS, Vyas SP. Cholera toxin B subunit conjugated bile salt stabilized vesicles (bilosomes) for oral immunization. Int J Pharm 2004; 278(2): 379-90.
[http://dx.doi.org/10.1016/j.ijpharm.2004.03.014] [PMID: 15196642]
[87]
Sharma D, Aara A, Aate J. Niosomes as Novel Drug Delivery System: Review Article. Pharmatutor 2018; p. 6.
[88]
Shilpa S, Srinivasan BP, Chauhan MK. Niosomes as vesicular carriers for delivery of proteins and biologicals. Int J Drug Deliv 2011; 3(1): 14-24.
[http://dx.doi.org/10.5138/ijdd.2010.0975.0215.03050]
[89]
Parekh F, Patel B, Vyas K, Patani P. Recent advances and scopes in niosomes. J Pharm Negat Results 2022; 13(8): 2093-102.
[http://dx.doi.org/10.47750/pnr.2022.13.S08.260]
[90]
De A, Venkatesh N, Senthil M, Sanapalli BKR, Shanmugham R, Karri VVSR. Smart niosomes of temozolomide for enhancement of brain targeting. Nanobiomedicine 2018; 5: 1849543518805355.
[http://dx.doi.org/10.1177/1849543518805355] [PMID: 30344765]
[91]
Ammar HO, Haider M, Ibrahim M, El Hoffy NM. In vitro and In vivo investigation for optimization of niosomal ability for sustainment and bioavailability enhancement of diltiazem after nasal administration. Drug Deliv 2017; 24(1): 414-21.
[http://dx.doi.org/10.1080/10717544.2016.1259371] [PMID: 28165822]
[92]
Madhav NVS, Saini A. Niosomes: A novel drug delivery system. Int J Res Pharm Chem 2011; 1(3): 498-511.
[93]
Kopermsub P, Mayen V, Warin C. Potential use of niosomes for encapsulation of nisin and EDTA and their antibacterial activity enhancement. Food Res Int 2011; 44(2): 605-12.
[http://dx.doi.org/10.1016/j.foodres.2010.12.011]
[94]
Moghassemi S, Parnian E, Hakamivala A, et al. Uptake and transport of insulin across intestinal membrane model using trimethyl chitosan coated insulin niosomes. Mater Sci Eng C 2015; 46: 333-40.
[http://dx.doi.org/10.1016/j.msec.2014.10.070] [PMID: 25491995]
[95]
Yadav R, Chanana A, Chawra H, Pal R. Recent advances in niosomal drug delivery: A review. Int J Multidiscip Res 2023; 5(1): 1-10.
[96]
Badri SPS, Annagowni NR, et al. A review on niosomes as novel drug delivary system. Int J Indigenous Herbs Drugs 2022; 7(5): 87.
[http://dx.doi.org/10.46956/ijihd.v7i5.352]
[97]
Tavano L, Muzzalupo R, Mauro L, Pellegrino M, Andò S, Picci N. Transferrin-conjugated pluronic niosomes as a new drug delivery system for anticancer therapy. Langmuir 2013; 29(41): 12638-46.
[http://dx.doi.org/10.1021/la4021383] [PMID: 24040748]
[98]
Kim TH, Jo YG, Jiang HH, et al. PEG-transferrin conjugated TRAIL (TNF-related apoptosis-inducing ligand) for therapeutic tumor targeting. J Control Release 2012; 162(2): 422-8.
[http://dx.doi.org/10.1016/j.jconrel.2012.07.021] [PMID: 22824780]
[99]
Srivastav AK, Das P. To study the formulation of niosome of ofloxacin and its evaluation for efficacy of anti-microbial activity. Int J Innov Res Sci Eng Technol 2014; 3(12): 17958-65.
[http://dx.doi.org/10.15680/IJIRSET.2014.0312028]
[100]
Saharkhiz S, Zarepour A, Zarrabi A. A new theranostic pH-responsive niosome formulation for doxorubicin delivery and bio-imaging against breast cancer. Int J Pharm 2023; 637: 122845.
[http://dx.doi.org/10.1016/j.ijpharm.2023.122845] [PMID: 36958608]
[101]
Barani M, Hajinezhad MR, Zargari F, et al. Preparation, characterization, cytotoxicity and pharmacokinetics of niosomes containing gemcitabine: In vitro, in vivo, and simulation studies. J Drug Deliv Sci Technol 2023; 84: 104505.
[http://dx.doi.org/10.1016/j.jddst.2023.104505]
[102]
Hosseini F, Mirzaei Chegeni M, Bidaki A, et al. 3D-printing-assisted synthesis of paclitaxel-loaded niosomes functionalized by cross-linked gelatin/alginate composite: Large-scale synthesis and in-vitro anti-cancer evaluation. Int J Biol Macromol 2023; 242(Pt 1): 124697.
[http://dx.doi.org/10.1016/j.ijbiomac.2023.124697] [PMID: 37156313]
[103]
Ferreira N, Gonçalves NP, Saraiva MJ, Almeida MR. Curcumin: A multi-target disease-modifying agent for late-stage transthyretin amyloidosis. Sci Rep 2016; 6(1): 26623.
[http://dx.doi.org/10.1038/srep26623] [PMID: 27197872]
[104]
Yang X, Li Z, Wang N, et al. Curcumin-encapsulated polymeric micelles suppress the development of colon cancer In vitro and In vivo. Sci Rep 2015; 5(1): 10322.
[http://dx.doi.org/10.1038/srep10322] [PMID: 25980982]
[105]
Zaman MS, Chauhan N, Yallapu MM, et al. Curcumin nanoformulation for cervical cancer treatment. Sci Rep 2016; 6(1): 20051.
[http://dx.doi.org/10.1038/srep20051] [PMID: 26837852]
[106]
Sharma V, Anandhakumar S, Sasidharan M. Self-degrading niosomes for encapsulation of hydrophilic and hydrophobic drugs: An efficient carrier for cancer multi-drug delivery. Mater Sci Eng C 2015; 56: 393-400.
[http://dx.doi.org/10.1016/j.msec.2015.06.049] [PMID: 26249606]
[107]
Naderinezhad S, Amoabediny G, Haghiralsadat F. Co-delivery of hydrophilic and hydrophobic anticancer drugs using biocompatible pH-sensitive lipid-based nano-carriers for multidrug-resistant cancers. RSC Adv 2017; 7(48): 30008-19.
[http://dx.doi.org/10.1039/C7RA01736G]
[108]
Ma H, Guo D, Fan Y, Wang J, Cheng J, Zhang X. Paeonol-loaded ethosomes as transdermal delivery carriers: Design, preparation and evaluation. Molecules 2018; 23(7): 1756.
[http://dx.doi.org/10.3390/molecules23071756] [PMID: 30018278]
[109]
Xu Y, Zhu J, Lei Z, et al. Anti-proliferative effects of paeonol on human prostate cancer cell lines DU145 and PC-3. J Physiol Biochem 2017; 73(2): 157-65.
[http://dx.doi.org/10.1007/s13105-016-0537-x] [PMID: 27834040]
[110]
Zong S, Pu Y, Li S, et al. Beneficial anti-inflammatory effect of paeonol self-microemulsion-loaded colon-specific capsules on experimental ulcerative colitis rats. Artif Cells Nanomed Biotechnol 2018; 46(S1): 324-35.
[http://dx.doi.org/10.1080/21691401.2017.1423497]
[111]
Lau CH, Chan CM, Chan YW, et al. Pharmacological investigations of the anti-diabetic effect of Cortex Moutan and its active component paeonol. Phytomedicine 2007; 14(11): 778-84.
[http://dx.doi.org/10.1016/j.phymed.2007.01.007] [PMID: 17298878]
[112]
Kim SH, Kim SA, Park MK, et al. Paeonol inhibits anaphylactic reaction by regulating histamine and TNF-α. Int Immunopharmacol 2004; 4(2): 279-87.
[http://dx.doi.org/10.1016/j.intimp.2003.12.013] [PMID: 14996419]
[113]
Wu J, Xue X, Zhang B, et al. Enhanced antitumor activity and attenuated cardiotoxicity of Epirubicin combined with Paeonol against breast cancer. Tumour Biol 2016; 37(9): 12301-13.
[http://dx.doi.org/10.1007/s13277-016-5088-9] [PMID: 27272157]
[114]
Chen ZX, Li B, Liu T, et al. Evaluation of paeonol-loaded transethosomes as transdermal delivery carriers. Eur J Pharm Sci 2017; 99: 240-5.
[http://dx.doi.org/10.1016/j.ejps.2016.12.026] [PMID: 28039091]
[115]
Zong S, Pu Y, Xu B, Zhang T, Wang B. Study on the physicochemical properties and anti-inflammatory effects of paeonol in rats with TNBS-induced ulcerative colitis. Int Immunopharmacol 2017; 42: 32-8.
[http://dx.doi.org/10.1016/j.intimp.2016.11.010] [PMID: 27863299]
[116]
Ionescu A, Aprodu I, Pascaru G. Effect of papain and bromelin on muscle and collagen proteins in beef meat. The Annals of the University Dunarea De Jos of Galati Fascicle VI - Food Technology 2008; II: 31.
[117]
Roslan NZI, Aziz AA, Sarmidi MR, Aziz RA. Anti - oxidant coated liposome as the delivery system for papain based natural cosmetics. International Conference on Enabling Science and Nanotechnology (ESciNano). Kuala Lumpur, Malaysia. 2010; pp. 01-03.
[118]
Taghizadeh F, Mehryab F, Mortazavi SA, Rabbani S, Haeri A. Thiolated chitosan hydrogel-embedded niosomes: A promising crocin delivery system toward the management of aphthous stomatitis. Carbohydr Polym 2023; 318: 121068.
[http://dx.doi.org/10.1016/j.carbpol.2023.121068]
[119]
Aghazadeh H, Taheri P, Aboulhassanzadeh S, et al. An herbal bioactive drug compound with a delayed release curve in PEGylated cationic nano-niosomes formulation. Biocatal Agric Biotechnol 2023; 51: 102704.
[http://dx.doi.org/10.1016/j.bcab.2023.102704]
[120]
Masotti A. Niosomes as candidate bioconjugates for imaging and pH-sensitive drug delivery nanocarriers for rare pediatric tumors. J Drug Deliv Sci Technol 2013; 23(1): 22-4.
[http://dx.doi.org/10.1016/S1773-2247(13)50003-7]
[121]
Azeem A, Anwer MK, Talegaonkar S. Niosomes in sustained and targeted drug delivery: Some recent advances. J Drug Target 2009; 17(9): 671-89.
[http://dx.doi.org/10.3109/10611860903079454] [PMID: 19845484]
[122]
Gulia M, Nishal S, Maddiboyina B, et al. Physiological pathway, diagnosis and nanotechnology based treatment strategies for ovarian cancer: A review. Medicine in Omics 2023; 8: 100020.
[http://dx.doi.org/10.1016/j.meomic.2023.100020]
[123]
Wang S, Hu X, Wei W, Ma G. Transformable vesicles for cancer immunotherapy. Adv Drug Deliv Rev 2021; 179: 113905.
[http://dx.doi.org/10.1016/j.addr.2021.113905] [PMID: 34331988]
[124]
Lv Y, Wei W, Ma G. Recent advances in platelet engineering for anti-cancer therapies. Particuology 2022; 64: 2-13.
[http://dx.doi.org/10.1016/j.partic.2021.09.006]
[125]
Kusdemir BC, Kozgus Guldu O, Yurt Kilcar A, Medine EI. Preparation and In vitro investigation of prostate-specific membrane antigen targeted lycopene loaded niosomes on prostate cancer cells. Int J Pharm 2023; 640: 123013.
[http://dx.doi.org/10.1016/j.ijpharm.2023.123013] [PMID: 37149111]
[126]
Akbarzadeh I, Rezaei N, Bazzazan S, et al. In silico and In vitro studies of GENT-EDTA encapsulated niosomes: A novel approach to enhance the antibacterial activity and biofilm inhibition in drug-resistant Klebsiella pneumoniae. Biomater Adv 2023; 149: 213384.
[http://dx.doi.org/10.1016/j.bioadv.2023.213384] [PMID: 37060635]
[127]
Gurrapu A, Jukanti R, Bobbala SR, Kanuganti S, Jeevana JB. Improved oral delivery of valsartan from maltodextrin based proniosome powders. Adv Powder Technol 2012; 23(5): 583-90.
[http://dx.doi.org/10.1016/j.apt.2011.06.005]
[128]
Azmin SNHM, Yunus NA, Mustaffa AA, Alwi SRW, Chua LS. Computer-aided approach for designing solvents blend for herbal phytochemical extraction. In: Gernaey KV, Huusom JK, Gani R, Eds. Computer Aided Chemical Engineering. Elsevier 2015; vol. 37: pp. 1427-32.
[129]
Jain S, Singh P, Mishra V, Vyas SP. Mannosylated niosomes as adjuvant–carrier system for oral genetic immunization against Hepatitis B. Immunol Lett 2005; 101(1): 41-9.
[http://dx.doi.org/10.1016/j.imlet.2005.04.002] [PMID: 15869802]
[130]
Khan MI, Madni A, Peltonen L. Development and in-vitro characterization of sorbitan monolaurate and poloxamer 184 based niosomes for oral delivery of diacerein. Eur J Pharm Sci 2016; 95: 88-95.
[http://dx.doi.org/10.1016/j.ejps.2016.09.002] [PMID: 27600819]
[131]
El Sharazly BM, Aboul Asaad IA, Yassen NA, et al. Mefloquine loaded niosomes as a promising approach for the treatment of acute and chronic toxoplasmosis. Acta Trop 2023; 239: 106810.
[http://dx.doi.org/10.1016/j.actatropica.2022.106810] [PMID: 36581225]
[132]
Lin YK, Hsiao CY, Alshetaili A, Aljuffali IA, Chen EL, Fang JY. Lipid-based nanoformulation optimization for achieving cutaneous targeting: Niosomes as the potential candidates to fulfill this aim. Eur J Pharm Sci 2023; 186: 106458.
[http://dx.doi.org/10.1016/j.ejps.2023.106458] [PMID: 37137418]
[133]
Erdogan S, Ozer Y, Ercan M. Eryılmaz M, Hincal A İn vivo studies on iopromide radiopaque niosomes. STP Pharma Sciences 1996; p. 6.
[134]
Citrin DE. Recent developments in radiotherapy. N Engl J Med 2017; 377(11): 1065-75.
[http://dx.doi.org/10.1056/NEJMra1608986] [PMID: 28902591]
[135]
Shewaiter MA, Selim AA, Moustafa YM, Gad S, Rashed HM. Radioiodinated acemetacin loaded niosomes as a dual anticancer therapy. Int J Pharm 2022; 628: 122345.
[http://dx.doi.org/10.1016/j.ijpharm.2022.122345] [PMID: 36349611]
[136]
Mashal M, Attia N, Puras G, Martínez-Navarrete G, Fernández E, Pedraz JL. Retinal gene delivery enhancement by lycopene incorporation into cationic niosomes based on DOTMA and polysorbate 60. J Control Release 2017; 254: 55-64.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.386] [PMID: 28347807]
[137]
Li Q, Li Z, Zeng W, et al. Proniosome-derived niosomes for tacrolimus topical ocular delivery: In vitro cornea permeation, ocular irritation, and in vivo anti-allograft rejection. Eur J Pharm Sci 2014; 62: 115-23.
[http://dx.doi.org/10.1016/j.ejps.2014.05.020] [PMID: 24905830]
[138]
Zubairu Y, Negi LM, Iqbal Z, Talegaonkar S. Design and development of novel bioadhesive niosomal formulation for the transcorneal delivery of anti-infective agent: In-vitro. and ex-vivo investigations. Asian J Pharmaceut Sci 2015; 10(4): 322-30.
[http://dx.doi.org/10.1016/j.ajps.2015.02.001]
[139]
Sankhyan A, Pawar P. Recent trends in niosome as vesicular drugdelivery system. J Appl Pharmaceut Sci 2012; 2(6): 20-32.
[140]
Alsaadi M, Italia JL, Mullen AB, et al. The efficacy of aerosol treatment with non-ionic surfactant vesicles containing amphotericin B in rodent models of leishmaniasis and pulmonary aspergillosis infection. J Control Release 2012; 160(3): 685-91.
[http://dx.doi.org/10.1016/j.jconrel.2012.04.004] [PMID: 22516093]
[141]
Moazeni E, Gilani K, Sotoudegan F, et al. Formulation and In vitro evaluation of ciprofloxacin containing niosomes for pulmonary delivery. J Microencapsul 2010; 27(7): 618-27.
[http://dx.doi.org/10.3109/02652048.2010.506579] [PMID: 20681747]
[142]
Rajizadeh MA, Nematollahi MH, Jafari E, et al. Niosome nanocarrier enhances the ameliorating effects of myrtenol in the lungs of rats with experimental asthma. OpenNano 2023; 11: 100129.
[http://dx.doi.org/10.1016/j.onano.2023.100129]
[143]
Yeo PL, Lim CL, Chye SM, Kiong Ling AP, Koh RY. Niosomes: A review of their structure, properties, methods of preparation, and medical applications. Asian Biomed 2018; 11(4): 301-14.
[http://dx.doi.org/10.1515/abm-2018-0002]
[144]
Wu X, Guy RH. Applications of nanoparticles in topical drug delivery and in cosmetics. J Drug Deliv Sci Technol 2009; 19(6): 371-84.
[http://dx.doi.org/10.1016/S1773-2247(09)50080-9]
[145]
Limphapayom W, Loylerd K, Leabwan N, Sukhasem S. Encapsulation of alpha-mangostin in cosmetic production by using nanotechnology. Acta Hortic 2017; (1186): 189-92.
[146]
Manosroi A, Boonpisuttinant K, Winitchai S, Manosroi W, Manosroi J. Free radical scavenging and tyrosinase inhibition activity of physic nut (jatropha curcas linn.) seed oil entrapped in niosomes. Curr Nanosci 2011; 7(5): 825-9.
[http://dx.doi.org/10.2174/157341311797483709]
[147]
Mawazi SM, Ann TJ, Widodo RT. Application of niosomes in cosmetics: A systematic review. Cosmetics 2022; 9(6): 127.
[http://dx.doi.org/10.3390/cosmetics9060127]
[148]
Bhosale A, Paul G, Mazahir F, Yadav AK. Theoretical and applied concepts of nanocarriers for the treatment of Parkinson’s diseases. OpenNano 2023; 9: 100111.
[http://dx.doi.org/10.1016/j.onano.2022.100111]
[149]
Javid-Naderi MJ, Mahmoudi A, Kesharwani P, Jamialahmadi T, Sahebkar A. Recent advances of nanotechnology in the treatment and diagnosis of polycystic ovary syndrome. J Drug Deliv Sci Technol 2023; 79: 104014.
[http://dx.doi.org/10.1016/j.jddst.2022.104014]
[150]
Nasr M. In vitro and In vivo evaluation of proniosomes containing celecoxib for oral administration. AAPS PharmSciTech 2010; 11(1): 85-9.
[http://dx.doi.org/10.1208/s12249-009-9364-5] [PMID: 20058106]
[151]
Akhter S, Kushwaha S, Warsi MH, et al. Development and evaluation of nanosized niosomal dispersion for oral delivery of Ganciclovir. Drug Dev Ind Pharm 2012; 38(1): 84-92.
[http://dx.doi.org/10.3109/03639045.2011.592529] [PMID: 21726136]
[152]
Muzzalupo R, Tavano L, La Mesa C. Alkyl glucopyranoside-based niosomes containing methotrexate for pharmaceutical applications: Evaluation of physico-chemical and biological properties. Int J Pharm 2013; 458(1): 224-9.
[http://dx.doi.org/10.1016/j.ijpharm.2013.09.011] [PMID: 24060370]
[153]
Katare R, Gupta PN, Mahor S, et al. Development of polysaccharide-capped niosomes for oral immunization of tetanus toxoid. J Drug Deliv Sci Technol 2006; 16(3): 167-72.
[http://dx.doi.org/10.1016/S1773-2247(06)50031-0]
[154]
Ibrahim MM, Shehata TM. Tramadol HCl encapsulated niosomes for extended analgesic effect following oral administration. J Drug Deliv Sci Technol 2018; 46: 14-8.
[http://dx.doi.org/10.1016/j.jddst.2018.04.011]
[155]
Abd El-Alim SH, Kassem AA, Basha M. Proniosomes as a novel drug carrier system for buccal delivery of benzocaine. J Drug Deliv Sci Technol 2014; 24(5): 452-8.
[http://dx.doi.org/10.1016/S1773-2247(14)50087-1]
[156]
Chattaraj SC, Das SK. Physicochemical characterization of influenza viral vaccine loaded surfactant vesicles. Drug Deliv 2003; 10(2): 73-7.
[http://dx.doi.org/10.1080/713840363] [PMID: 12746052]
[157]
Abdelrahman FE, Elsayed I, Gad MK, Elshafeey AH, Mohamed MI. Response surface optimization, Ex vivo and In vivo investigation of nasal spanlastics for bioavailability enhancement and brain targeting of risperidone. Int J Pharm 2017; 530(1-2): 1-11.
[http://dx.doi.org/10.1016/j.ijpharm.2017.07.050] [PMID: 28733244]
[158]
Zidan AS, Habib MJ. Maximized mucoadhesion and skin permeation of anti-AIDS-loaded niosomal gels. J Pharm Sci 2014; 103(3): 952-64.
[http://dx.doi.org/10.1002/jps.23867] [PMID: 24464823]
[159]
Mukherjee B, Patra B, Layek B, Mukherjee A. Sustained release of acyclovir from nano-liposomes and nano-niosomes: An In vitro study. Int J Nanomedicine 2007; 2(2): 213-25.
[PMID: 17722549]
[160]
Pardakhty A, Shakibaie M, Daneshvar H, Khamesipour A, Mohammadi-Khorsand T, Forootanfar H. Preparation and evaluation of niosomes containing autoclaved Leishmania major: A preliminary study. J Microencapsul 2012; 29(3): 219-24.
[http://dx.doi.org/10.3109/02652048.2011.642016] [PMID: 22150018]
[161]
Yang H, Deng A, Zhang J, Wang J, Lu B. Preparation, characterization and anticancer therapeutic efficacy of cisplatin-loaded niosomes. J Microencapsul 2013; 30(3): 237-44.
[http://dx.doi.org/10.3109/02652048.2012.717116] [PMID: 23272761]
[162]
Wang M, Yuan Y, Gao Y, et al. Preparation and characterization of 5-fluorouracil pH-sensitive niosome and its tumor-targeted evaluation: In vitro and In vivo. Drug Dev Ind Pharm 2012; 38(9): 1134-41.
[http://dx.doi.org/10.3109/03639045.2011.641565] [PMID: 22182601]
[163]
Mullaicharam AR, Murthy RSR. Lung accumulation of niosome-entrapped gentamicin sulfate follows intravenous and intratracheal administration in rats. J Drug Deliv Sci Technol 2006; 16(2): 109-13.
[http://dx.doi.org/10.1016/S1773-2247(06)50016-4]
[164]
Hong M, Zhu S, Jiang Y, Tang G, Pei Y. Efficient tumor targeting of hydroxycamptothecin loaded PEGylated niosomes modified with transferrin. J Control Release 2008; 133(2): 96-102.
[http://dx.doi.org/10.1016/j.jconrel.2008.09.005] [PMID: 18840485]
[165]
El-Ridy MS, Abdelbary A, Essam T. Abd EL-Salam RM, Aly Kassem AA. Niosomes as a potential drug delivery system for increasing the efficacy and safety of nystatin. Drug Dev Ind Pharm 2011; 37(12): 1491-508.
[http://dx.doi.org/10.3109/03639045.2011.587431] [PMID: 21707323]
[166]
Mullaicharam AR, Murthy RSR. Lung accumulation of niosome-entrapped rifampicin following intravenous and intratracheal administration in the rat. J Drug Deliv Sci Technol 2004; 14(2): 99-104.
[http://dx.doi.org/10.1016/S1773-2247(04)50020-5]
[167]
Shaker DS, Shaker MA, Hanafy MS. Cellular uptake, cytotoxicity and in-vivo evaluation of Tamoxifen citrate loaded niosomes. Int J Pharm 2015; 493(1-2): 285-94.
[http://dx.doi.org/10.1016/j.ijpharm.2015.07.041] [PMID: 26200748]
[168]
Kumar K, Rai AK. Proniosomal formulation of curcumin having anti-inflammatory and anti-arthritic activity in different experimental animal models. Pharmazie 2012; 67(10): 852-7.
[PMID: 23136720]
[169]
Jin Y, Wen J, Garg S, et al. Development of a novel niosomal system for oral delivery of Ginkgo biloba extract. Int J Nanomedicine 2013; 8: 421-30.
[http://dx.doi.org/10.2147/IJN.S37984] [PMID: 23378764]
[170]
Alam MS, Ahad A, Abidin L, Aqil M, Mir SR, Mujeeb M. Embelin-loaded oral niosomes ameliorate streptozotocin-induced diabetes in Wistar rats. Biomed Pharmacother 2018; 97: 1514-20.
[http://dx.doi.org/10.1016/j.biopha.2017.11.073] [PMID: 29793314]
[171]
Abdelkader H, Ismail S, Hussein A, Wu Z, Al-Kassas R, Alany RG. Conjunctival and corneal tolerability assessment of ocular naltrexone niosomes and their ingredients on the hen’s egg chorioallantoic membrane and excised bovine cornea models. Int J Pharm 2012; 432(1-2): 1-10.
[http://dx.doi.org/10.1016/j.ijpharm.2012.04.063] [PMID: 22575752]
[172]
Ojeda E, Puras G, Agirre M, et al. The influence of the polar head-group of synthetic cationic lipids on the transfection efficiency mediated by niosomes in rat retina and brain. Biomaterials 2016; 77: 267-79.
[http://dx.doi.org/10.1016/j.biomaterials.2015.11.017] [PMID: 26610076]
[173]
El-Menshawe SF. A novel approach to topical acetazolamide/PEG 400 ocular niosomes. J Drug Deliv Sci Technol 2012; 22(4): 295-9.
[http://dx.doi.org/10.1016/S1773-2247(12)50049-3]
[174]
Abdel-Mageed HM, El-Laithy HM, Mahran LG, Fahmy AS, Mäder K, Mohamed SA. Development of novel flexible sugar ester vesicles as carrier systems for the antioxidant enzyme catalase for wound healing applications. Process Biochem 2012; 47(7): 1155-62.
[http://dx.doi.org/10.1016/j.procbio.2012.04.008]
[175]
Tavano L, Alfano P, Muzzalupo R, de Cindio B. Niosomes vs microemulsions: New carriers for topical delivery of Capsaicin. Colloids Surf B Biointerfaces 2011; 87(2): 333-9.
[http://dx.doi.org/10.1016/j.colsurfb.2011.05.041] [PMID: 21684725]
[176]
Moghddam SRM, Ahad A, Aqil M, Imam SS, Sultana Y. Formulation and optimization of niosomes for topical diacerein delivery using 3-factor, 3-level Box-Behnken design for the management of psoriasis. Mater Sci Eng C 2016; 69: 789-97.
[http://dx.doi.org/10.1016/j.msec.2016.07.043] [PMID: 27612773]
[177]
Fang JY, Hong CT, Chiu WT, Wang YY. Effect of liposomes and niosomes on skin permeation of enoxacin. Int J Pharm 2001; 219(1-2): 61-72.
[http://dx.doi.org/10.1016/S0378-5173(01)00627-5] [PMID: 11337166]
[178]
Singh S, Parashar P, Kanoujia J, Singh I, Saha S, Saraf SA. Transdermal potential and anti-gout efficacy of Febuxostat from niosomal gel. J Drug Deliv Sci Technol 2017; 39: 348-61.
[http://dx.doi.org/10.1016/j.jddst.2017.04.020]
[179]
Bendas ER, Abdullah H, El-Komy MHM, Kassem MAA. Hydroxychloroquine niosomes: A new trend in topical management of oral lichen planus. Int J Pharm 2013; 458(2): 287-95.
[http://dx.doi.org/10.1016/j.ijpharm.2013.10.042] [PMID: 24184035]
[180]
Khazaeli P, Sharifi I, Talebian E, Heravi G, Moazeni E, Mostafavi M. Anti-leishmanial effect of itraconazole niosome on in vitro susceptibility of Leishmania tropica. Environ Toxicol Pharmacol 2014; 38(1): 205-11.
[http://dx.doi.org/10.1016/j.etap.2014.04.003] [PMID: 24956400]
[181]
Qumbar M. Ameeduzzafar, Imam SS, Ali J, Ahmad J, Ali A. Formulation and optimization of lacidipine loaded niosomal gel for transdermal delivery: In-vitro characterization and in-vivo activity. Biomed Pharmacother 2017; 93: 255-66.
[http://dx.doi.org/10.1016/j.biopha.2017.06.043] [PMID: 28738502]
[182]
Abdelbary AA, AbouGhaly MHH. Design and optimization of topical methotrexate loaded niosomes for enhanced management of psoriasis: Application of Box–Behnken design, in-vitro evaluation and in-vivo skin deposition study. Int J Pharm 2015; 485(1-2): 235-43.
[http://dx.doi.org/10.1016/j.ijpharm.2015.03.020] [PMID: 25773359]
[183]
Sohrabi S, Haeri A, Mahboubi A, Mortazavi A, Dadashzadeh S. Chitosan gel-embedded moxifloxacin niosomes: An efficient antimicrobial hybrid system for burn infection. Int J Biol Macromol 2016; 85: 625-33.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.01.013] [PMID: 26794314]
[184]
Manosroi J, Lohcharoenkal W, Götz F, Werner RG, Manosroi W, Manosroi A. Transdermal absorption enhancement of N-terminal Tat-GFP fusion protein (TG) loaded in novel low-toxic elastic anionic niosomes. J Pharm Sci 2011; 100(4): 1525-34.
[http://dx.doi.org/10.1002/jps.22355] [PMID: 20891012]
[185]
Patel J, Ketkar S, Patil S, Fearnley J, Mahadik KR, Paradkar AR. Potentiating antimicrobial efficacy of propolis through niosomal-based system for administration. Integr Med Res 2015; 4(2): 94-101.
[http://dx.doi.org/10.1016/j.imr.2014.10.004] [PMID: 28664114]
[186]
Negi P, Aggarwal M, Sharma G, et al. Niosome-based hydrogel of resveratrol for topical applications: An effective therapy for pain related disorder(s). Biomed Pharmacother 2017; 88: 480-7.
[http://dx.doi.org/10.1016/j.biopha.2017.01.083] [PMID: 28126673]
[187]
Pando D, Matos M, Gutiérrez G, Pazos C. Formulation of resveratrol entrapped niosomes for topical use. Colloids Surf B Biointerfaces 2015; 128: 398-404.
[http://dx.doi.org/10.1016/j.colsurfb.2015.02.037] [PMID: 25766923]
[188]
Sambhakar S, Paliwal S, Sharma S, Singh B. Formulation of risperidone loaded proniosomes for effective transdermal delivery: An in-vitro and in-vivo study. Bull Fac Pharm Cairo Univ 2017; 55(2): 239-47.
[http://dx.doi.org/10.1016/j.bfopcu.2017.09.003]
[189]
Zhang Y, Zhang K, Wu Z, et al. Evaluation of transdermal salidroside delivery using niosomes via in vitro cellular uptake. Int J Pharm 2015; 478(1): 138-46.
[http://dx.doi.org/10.1016/j.ijpharm.2014.11.018] [PMID: 25448576]
[190]
Ammar HO, Ghorab M, El-Nahhas SA, Higazy IM. Proniosomes as a carrier system for transdermal delivery of tenoxicam. Int J Pharm 2011; 405(1-2): 142-52.
[http://dx.doi.org/10.1016/j.ijpharm.2010.11.003] [PMID: 21129461]
[191]
Manconi M, Sinico C, Valenti D, Lai F, Fadda AM. Niosomes as carriers for tretinoin. Int J Pharm 2006; 311(1-2): 11-9.
[http://dx.doi.org/10.1016/j.ijpharm.2005.11.045] [PMID: 16439071]
[192]
Jamal M, Imam SS, Aqil M, Amir M, Mir SR, Mujeeb M. Transdermal potential and anti-arthritic efficacy of ursolic acid from niosomal gel systems. Int Immunopharmacol 2015; 29(2): 361-9.
[http://dx.doi.org/10.1016/j.intimp.2015.10.029] [PMID: 26545446]
[193]
Kong M, Park H, Feng C, Hou L, Cheng X, Chen X. Construction of hyaluronic acid noisome as functional transdermal nanocarrier for tumor therapy. Carbohydr Polym 2013; 94(1): 634-41.
[http://dx.doi.org/10.1016/j.carbpol.2013.01.091] [PMID: 23544584]
[194]
Kassem AA, Abd El-Alim SH, Asfour MH. Enhancement of 8-methoxypsoralen topical delivery via nanosized niosomal vesicles: Formulation development, In vitro and In vivo evaluation of skin deposition. Int J Pharm 2017; 517(1-2): 256-68.
[http://dx.doi.org/10.1016/j.ijpharm.2016.12.018] [PMID: 27956194]
[195]
Tavano L, Gentile L, Oliviero Rossi C, Muzzalupo R. Novel gel-niosomes formulations as multicomponent systems for transdermal drug delivery. Colloids Surf B Biointerfaces 2013; 110: 281-8.
[http://dx.doi.org/10.1016/j.colsurfb.2013.04.017] [PMID: 23732806]
[196]
Shehata T, Kono Y, Higaki K, Kimura T, Ogawara K. In vivo distribution characteristics and anti-tumor effects of doxorubicin encapsulated in PEG-modified niosomes in solid tumor-bearing mice. J Drug Deliv Sci Technol 2023; 80: 104122.
[http://dx.doi.org/10.1016/j.jddst.2022.104122]
[197]
Hatami Nemati S, Bigdeli MR, Mortazavi Moghadam F, Sharifi K. Neuroprotective effects of niosomes loaded with thymoquinone in the cerebral ischemia model of male Wistar rats. Nanomedicine 2023; 48: 102637.
[http://dx.doi.org/10.1016/j.nano.2022.102637] [PMID: 36549552]
[198]
Bhardwaj P, Tripathi P, Pandey S, Gupta R, Khar RK, Patil PR. Improved dermal delivery of pentoxifylline niosomes for the management of psoriasis: Development, optimization and in-vivo studies in imiquimod induced psoriatic plaque model. J Drug Deliv Sci Technol 2022; 75: 103643.
[http://dx.doi.org/10.1016/j.jddst.2022.103643]
[199]
Kulkarni P, Rawtani D, Rajpurohit S, Vasvani S, Barot T. Self-assembly based aerosolized hyaluronic acid (HA) loaded niosomes for lung delivery: An in-vitro and in-vivo evaluation. J Drug Deliv Sci Technol 2022; 75: 103627.
[http://dx.doi.org/10.1016/j.jddst.2022.103627]
[200]
Andishmand H, Azadmard-damirchi S, Hamishekar H, et al. Nano-delivery systems for encapsulation of phenolic compounds from pomegranate peel. Adv Colloid Interface Sci 2023; 311: 102833.
[http://dx.doi.org/10.1016/j.cis.2022.102833] [PMID: 36610103]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy