Generic placeholder image

Current Nanomedicine

Editor-in-Chief

ISSN (Print): 2468-1873
ISSN (Online): 2468-1881

Review Article

Phytoniosomes: An Emergent Strategy for Herbal Drug Delivery System

Author(s): Priya Kumari, Shaweta Sharma*, Pramod Kumar Sharma and Mohd Aftab Alam

Volume 11, Issue 3, 2021

Published on: 22 December, 2021

Page: [149 - 167] Pages: 19

DOI: 10.2174/2468187311666210921103858

Price: $65

conference banner
Abstract

Background and Objective: In the current era of advancement in the field of pharmaceutics, there is a growing interest in applying nanomedicine technology for active phytoconstituents and herbal extracts. This revolution in the area of herbal medicine has led to the growth of different technological approaches for delivering poorly soluble active herbal constituents or phytoconstituents, and herbal extract to enhance the safety, bioavailability, efficacy as well as receptor binding selectivity of the active entity. These nano-medicinal approaches have shown to be a more effective and reliable delivery system for herbal drugs. Niosomes are one of the novel drug delivery system approaches that have shown promising results when employed in the delivery of many drugs, including herbal drugs. The term ‘phytoniosomes’ as mentioned in various research papers, defines phyto/herbal drugs encapsulated in a non-ionic vesicular system. During the past decade, several research articles have been published on the development and characterization of phytoniosome nano-vesicle along with their in vivo studies as well as delivery approaches via different routes.

Methods: During the writing of this document, the data was derived from existing original research papers gathered from a variety of sources such as PubMed, Google Scholar, Science Direct, etc.

Conclusion: This review discusses various aspects of phytoniosomes covering different areas such as techniques and methods involved in their preparation, various factors affecting their physicochemical properties while developing phytoniosomes, their characterization, and several applications and advantages.

Keywords: Phytoniosomes, herbal niosomes, niosome, phytochemicals, targeted drug delivery, nanotechnology.

Graphical Abstract

[1]
Li F-S, Weng J-K. Demystifying traditional herbal medicine with modern approach. Nat Plants 2017; 3(8): 17109.
[http://dx.doi.org/10.1038/nplants.2017.109] [PMID: 28758992]
[2]
Rates SM. Plants as source of drugs. Toxicon 2001; 39(5): 603-13.
[http://dx.doi.org/10.1016/S0041-0101(00)00154-9] [PMID: 11072038]
[3]
OMS. Informe mundial de la OMS sobre medicina tradicional y complementaria 2019. Organización Mundial de la Salud 2019.
[4]
Mendoza N, Silva EME. Introduction to phytochemicals: secondary metabolites from plants with active principles for pharmacological importance. Asao, Toshiki. Phytochemicals - Source of Antioxidants and Role in Disease Prevention. In Tech 2018; pp. 25-47.
[http://dx.doi.org/10.5772/intechopen.78226]
[5]
Mujeeb F, Bajpai P, Pathak N. Phytochemical evaluation, antimicrobial activity, and determination of bioactive components from leaves of Aegle marmelos. BioMed Res Int 2014; 2014: 497606.
[http://dx.doi.org/10.1155/2014/497606] [PMID: 24900969]
[6]
Marianecci C, Rinaldi F, Mastriota M, et al. Anti-inflammatory activity of novel ammonium glycyrrhizinate/niosomes delivery system: human and murine models. J Control Release 2012; 164(1): 17-25.
[http://dx.doi.org/10.1016/j.jconrel.2012.09.018] [PMID: 23041542]
[7]
Kesarwani K, Gupta R, Mukerjee A. Bioavailability enhancers of herbal origin: an overview. Asian Pac J Trop Biomed 2013; 3(4): 253-66.
[http://dx.doi.org/10.1016/S2221-1691(13)60060-X] [PMID: 23620848]
[8]
Bonifácio BV, Silva PB, Ramos MA, Negri KM, Bauab TM, Chorilli M. Nanotechnology-based drug delivery systems and herbal medicines: a review. Int J Nanomedicine 2014; 9: 1-15.
[http://dx.doi.org/10.2147/IJN.S52634] [PMID: 24363556]
[9]
Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology 2018; 16(1): 71.
[http://dx.doi.org/10.1186/s12951-018-0392-8] [PMID: 30231877]
[10]
Vital R. WHO global report on traditional and complementary medicine 2019 vital. 2019.
[11]
Jahangirian H, Lemraski EG, Webster TJ, Rafiee-Moghaddam R, Abdollahi Y. A review of drug delivery systems based on nanotechnology and green chemistry: green nanomedicine. Int J Nanomedicine 2017; 12: 2957-78.
[http://dx.doi.org/10.2147/IJN.S127683] [PMID: 28442906]
[12]
Nalini T, Kumari SBK. Novel nanosystems for herbal drug delivery. World J Pharm Pharm Sci 2017; 6(5): 1447-63.
[http://dx.doi.org/10.20959/wjpps201705-9183]
[13]
Khogta S, Patel J, Barve K, Londhe V. Herbal nano-formulations for topical delivery. J Herb Med 2020; 20: 100300.
[http://dx.doi.org/10.1016/j.hermed.2019.100300]
[14]
Bhat M, Pukale S, Singh S, Mittal A, Chitkara D. Nano-enabled topical delivery of anti-psoriatic small molecules. J Drug Deliv Sci Technol 2021; 62: 102328.
[http://dx.doi.org/10.1016/j.jddst.2021.102328]
[15]
Sandhiya V, Ubaidulla U. A review on herbal drug loaded into pharmaceutical carrier techniques and its evaluation process. Futur J Pharm Sci 2020; 6(1): 1-6.
[http://dx.doi.org/10.1186/s43094-020-00050-0]
[16]
Rahman HS, Othman HH, Hammadi NI, et al. Novel drug delivery systems for loading of natural plant extracts and their biomedical applications. Int J Nanomedicine 2020; 15: 2439-83.
[http://dx.doi.org/10.2147/IJN.S227805] [PMID: 32346289]
[17]
Chakraborty K, Shivakumar A, Ramachandran S. Nano-technology in herbal medicines: A review. Int J Herb Med 2016; 4(3): 21-7.
[http://dx.doi.org/10.22271/flora.2016.v4.i3.05]
[18]
Elzoghby A. Thematic Issue: Nanocarriers based on natural polymers as platforms for drug and gene delivery applications. Curr Pharm Des 2016; 22(22): 3303-4.
[http://dx.doi.org/10.2174/1381612822999160511151612] [PMID: 27165162]
[19]
Masjedi M, Montahaei T. An illustrated review on nonionic surfactant vesicles (niosomes) as an approach in modern drug delivery: Fabrication, characterization, pharmaceutical, and cosmetic applications. J Drug Deliv Sci Technol 2021; 61: 102234.
[http://dx.doi.org/10.1016/j.jddst.2020.102234]
[20]
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]
[21]
Mozafari MR. Nanomaterials and nanosystems for biomedical applications dordrecht. Springer Netherlands 2007; pp. 1-159.
[http://dx.doi.org/10.1007/978-1-4020-6289-6]
[22]
Mozafari MR, Khosravi-Darani K. An Overview of liposome-derived nanocarrier technologies Nanomaterials and nanosystems for biomedical applications. Dordrecht: Springer Netherlands 2007; pp. 113-23.
[http://dx.doi.org/10.1007/978-1-4020-6289-6_7]
[23]
Hoque M, Agarwal S, Gupta S, Garg S, Syed I, Rupesh A. Lipid nanostructures in food applications Innovative food processing technologies. Elsevier 2021; pp. 565-79.
[http://dx.doi.org/10.1016/B978-0-08-100596-5.23047-3]
[24]
Muzzalupo R, Tavano L. Niosomal drug delivery for transdermal targeting: recent advances. Res Reports Transdermal Drug Deliv 2015; 4: 23-33.
[http://dx.doi.org/10.2147/RRTD.S64773]
[25]
Singh B, Saini S, Lohan S, Beg S. Systematic development of nanocarriers employing quality by design paradigms Nanotechnology-based approaches for targeting and delivery of drugs and genes. Elsevier 2017; pp. 110-48.
[http://dx.doi.org/10.1016/B978-0-12-809717-5.00003-8]
[26]
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]
[27]
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]
[28]
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]
[29]
Rigano L, Lionetti N. Nanobiomaterials in galenic formulations and cosmetics Nanobiomaterials in galenic formulations and cosmetics. Elsevier 2016; pp. 121-48.
[http://dx.doi.org/10.1016/B978-0-323-42868-2.00006-1]
[30]
Badwaik HR, Kumari L, Nakhate K, Verma VS, Sakure K. Phytoconstituent plumbagin: Chemical, biotechnological and pharmaceutical aspects. Stud Nat Prod Chem 2019; 63: 415-60.
[http://dx.doi.org/10.1016/B978-0-12-817901-7.00013-7]
[31]
Isnan AP, Jufri M. Formulation of niosomal gel containing green tea extract (Camellia Sinensis L. kuntze) using thin-layer hydration. Int J Appl Pharm 2017; 9: 38.
[http://dx.doi.org/10.22159/ijap.2017.v9s1.23_28]
[32]
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: 6847971.
[http://dx.doi.org/10.1155/2018/6847971] [PMID: 30651728]
[33]
Alyami H, Abdelaziz K, Dahmash EZ, Iyire A. Nonionic surfactant vesicles (niosomes) for ocular drug delivery: Development, evaluation and toxicological profiling. J Drug Deliv Sci Technol 2020; 60: 102069.
[http://dx.doi.org/10.1016/j.jddst.2020.102069]
[34]
Wilson DK, Shyamala G, Paulpandi M, Narayanasamy A, Siram K, Karuppaiah A. Development and characterization of phytoniosome nano vesicle loaded with aqueous leaf extracts of justicia adhatoda and psidium guajoava against dengue virus (DEN-2). J Cluster Sci 2021; 32(2): 297-304.
[http://dx.doi.org/10.1007/s10876-020-01788-6]
[35]
Raeiszadeh M, Pardakhty A, Sharififar F, Mehrabani M, Nejat-Mehrab-Kermani H, Mehrabani M. Phytoniosome: A novel drug delivery for myrtle extract. Iran J Pharm Res 2018; 17(3): 804-17.
[PMID: 30127807]
[36]
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]
[37]
Barani M, Mirzaei M, Torkzadeh-Mahani M, Nematollahi MH. Lawsone-loaded Niosome and its antitumor activity in MCF-7 breast Cancer cell line: a Nano-herbal treatment for Cancer. Daru 2018; 26(1): 11-7.
[http://dx.doi.org/10.1007/s40199-018-0207-3] [PMID: 30159762]
[38]
Assadpour E, Jafari SM. An overview of lipid-based nanostructures for encapsulation of food ingredients Lipid-based nanostructures for food encapsulation purposes. Elsevier Inc. 2019; pp. 1-34.
[http://dx.doi.org/10.1016/B978-0-12-815673-5.00001-5]
[39]
Orafidiya LO, Fakoya F, Agbani E, Iwalewa E. Vascular permeability-increasing effect of the leaf essential oil of Ocimum gratissimum Linn as a mechanism for its wound healing property. Afr J Tradit Complement Altern Med 2005; 2(3): 253-8.
[http://dx.doi.org/10.4314/ajtcam.v2i3.31123]
[40]
Patil DN, Kulkarni AR, Shahapurka AA, Hatappakki BC. Natural cumin seeds for wound healing activity in albino rats. Int J Biol Chem 2009; 3(4): 148-52.
[http://dx.doi.org/10.3923/ijbc.2009.148.152]
[41]
Shetty S, Udupa S, Udupa L. Evaluation of antioxidant and wound healing effects of alcoholic and aqueous extract of ocimum sanctum linn in rats. Evid Based Complement Alternat Med 2008; 5(1): 95-101.
[http://dx.doi.org/10.1093/ecam/nem004] [PMID: 18317555]
[42]
Azwanida Nn. A review on the extraction methods use in medicinal plants, principle, strength and limitation. Med Aromat Plants 2015; 04(03): 1-6.
[http://dx.doi.org/10.4172/2167-0412.1000196]
[43]
Nayak BS, Kanhai J, Milne DM, Pinto Pereira L, Swanston WH. Experimental evaluation of ethanolic extract of Carapa guianensis L. Leaf for its wound healing activity using three wound models. Evid Based Complement Alternat Med 2011; 2011: 419612.
[http://dx.doi.org/10.1093/ecam/nep160] [PMID: 19825872]
[44]
Tuhin RH, Begum MM, Rahman MS, et al. Wound healing effect of Euphorbia hirta linn. (Euphorbiaceae) in alloxan induced diabetic rats. BMC Complement Altern Med 2017; 17(1): 423.
[http://dx.doi.org/10.1186/s12906-017-1930-x] [PMID: 28836990]
[45]
Redfern J, Kinninmonth M, Burdass D, Verran J. Using soxhlet ethanol extraction to produce and test plant material (essential oils) for their antimicrobial properties. J Microbiol Biol Educ 2014; 15(1): 45-6.
[http://dx.doi.org/10.1128/jmbe.v15i1.656] [PMID: 24839520]
[46]
Zhang Q-W, Lin L-G, Ye W-C. Techniques for extraction and isolation of natural products: a comprehensive review. Chin Med 2018; 13(1): 20.
[http://dx.doi.org/10.1186/s13020-018-0177-x] [PMID: 29692864]
[47]
Matos M, Pando D, Gutiérrez G. Nanoencapsulation of food ingredients by niosomes Lipid-based nanostructures for food encapsulation purposes. Elsevier 2019; pp. 447-81.
[http://dx.doi.org/10.1016/B978-0-12-815673-5.00011-8]
[48]
Khan DH, Bashir S, Figueiredo P, Santos HA, Khan MI, Peltonen L. Process optimization of ecological probe sonication technique for production of rifampicin loaded niosomes. J Drug Deliv Sci Technol 2019; 50: 27-33.
[http://dx.doi.org/10.1016/j.jddst.2019.01.012]
[49]
Baillie AJ, Florence AT, Hume LR, Muirhead GT, Rogerson A. The preparation and properties of niosomes--non-ionic surfactant vesicles. J Pharm Pharmacol 1985; 37(12): 863-8.
[http://dx.doi.org/10.1111/j.2042-7158.1985.tb04990.x] [PMID: 2868092]
[50]
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]
[51]
Kazi KM, Mandal AS, Biswas N, 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]
[52]
Ag Seleci D, Seleci M, Walter J-G, 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]
[53]
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]
[54]
Kiwada H, Niimura H, Fujisaki Y, Yamada S, Kato Y. Application of synthetic alkyl glycoside vesicles as drug carriers. I. Preparation and physical properties. Chem Pharm Bull (Tokyo) 1985; 33(2): 753-9.
[http://dx.doi.org/10.1248/cpb.33.753] [PMID: 4017121]
[55]
Budhiraja A, Dhingra G. Development and characterization of a novel antiacne niosomal gel of rosmarinic acid. Drug Deliv 2015; 22(6): 723-30.
[http://dx.doi.org/10.3109/10717544.2014.903010] [PMID: 24786487]
[56]
Abdallah M, Sammour O. Summary for policymakers. In: Intergovernmental Panel on Climate Change. Cambridge: Cambridge University Press 2013; pp. 1-30.
[http://dx.doi.org/10.1017/CBO9781107415324.004]
[57]
Rogerson A, Cummings J, Willmott N, Florence AT. The distribution of doxorubicin in mice following administration in niosomes. J Pharm Pharmacol 1988; 40(5): 337-42.
[http://dx.doi.org/10.1111/j.2042-7158.1988.tb05263.x] [PMID: 2899629]
[58]
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]
[59]
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]
[60]
Rangasamy M, Ayyasamy B, Raju S, Gummadevelly S, Shaik S. Formulation and in vitro evaluation of Niosome encapsulated acyclovir multiparticulate drug delivery systems: Pellet and pelletization technique view project formulation and in vitro evaluation of niosome encapsulated acyclovir. J Pharm Res 2008; 1(2): 163-6.
[61]
Goyal G, Garg T, Malik B, Chauhan G, Rath G, Goyal AK. Development and characterization of niosomal gel for topical delivery of benzoyl peroxide. Drug Deliv 2015; 22(8): 1027-42.
[http://dx.doi.org/10.3109/10717544.2013.855277] [PMID: 24251352]
[62]
Singh G, Dwivedi H, Saraf S, Saraf S. Niosomal delivery of isoniazid - development and characterization. Trop J Pharm Res 2011.
[http://dx.doi.org/10.4314/tjpr.v10i2.66564]
[63]
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 1985; 816(2): 294-302.
[http://dx.doi.org/10.1016/0005-2736(85)90497-3] [PMID: 3839135]
[64]
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]
[65]
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]
[66]
Talsma H, van Steenbergen MJ, Borchert JCH, Crommelin DJA. A novel technique for the one-step preparation of liposomes and nonionic surfactant vesicles without the use of organic solvents. Liposome formation in a continuous gas stream: the ‘bubble’ method. J Pharm Sci 1994; 83(3): 276-80.
[http://dx.doi.org/10.1002/jps.2600830303] [PMID: 8207668]
[67]
Khan R, Irchhaiya R. Niosomes: A potential tool for novel drug delivery. J Pharm Investig 2016; 46(3): 195-204.
[http://dx.doi.org/10.1007/s40005-016-0249-9]
[68]
Liposome preparation by single-pass process Google Patents EP1920765A1, 2008.
[69]
G DB, P VL. Recent advances of non-ionic surfactant-based nano-vesicles (niosomes and proniosomes): A brief review of these in enhancing transdermal delivery of drug. Futur J Pharm Sci 2020; 6(1): 100.
[http://dx.doi.org/10.1186/s43094-020-00117-y]
[70]
Azmin MN, Florence AT, Handjani-Vila RM, Stuart JFB, 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 1985; 37(4): 237-42.
[http://dx.doi.org/10.1111/j.2042-7158.1985.tb05051.x] [PMID: 2860220]
[71]
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 (Basel) 2019; 12(2): E46.
[http://dx.doi.org/10.3390/ph12020046] [PMID: 30934834]
[72]
Sezgin-Bayindir Z, Yuksel N. Investigation of formulation variables and excipient interaction on the production of niosomes. AAPS PharmSciTech 2012; 13(3): 826-35.
[http://dx.doi.org/10.1208/s12249-012-9805-4] [PMID: 22644706]
[73]
Talsma H, Özer AY, van Bloois L, Crommelin DJA. The size reduction of liposomes with a high pressure homogenizer (microfluidizerTM characterization of prepared dispersions and comparison with conventional methods. Drug Dev Ind Pharm 1989; 15(2): 197-207.
[http://dx.doi.org/10.3109/03639048909040205]
[74]
Sorgi FL, Huang L. Large scale production of DC-Chol cationic liposomes by microfluidization. Int J Pharm 1996; 144(2): 131-9.
[http://dx.doi.org/10.1016/S0378-5173(96)04733-3]
[75]
Guo P, Huang J, Zhao Y, Martin CR, Zare RN, Moses MA. Nanomaterial preparation by extrusion through nanoporous membranes. Small 2018; 14(18): e1703493.
[http://dx.doi.org/10.1002/smll.201703493] [PMID: 29468837]
[76]
Ong SGM, Chitneni M, Lee KS, Ming LC, Yuen KH. Evaluation of extrusion technique for nanosizing liposomes. Pharmaceutics 2016; 8(4): 36.
[http://dx.doi.org/10.3390/pharmaceutics8040036] [PMID: 28009829]
[77]
Uchegbu IF, Double JA, Turton JA, Florence AT. Distribution, metabolism and tumoricidal activity of doxorubicin administered in sorbitan monostearate (Span 60) niosomes in the mouse. Pharm Res 1995; 12(7): 1019-24.
[http://dx.doi.org/10.1023/A:1016210515134] [PMID: 7494796]
[78]
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 Advances 2018; 8(57): 32621-36.
[http://dx.doi.org/10.1039/C8RA06362A]
[79]
Hua W, Liu T. Preparation and properties of highly stable innocuous niosome in Span 80/PEG 400/H2O system. Colloids Surf A Physicochem Eng Asp 2007; 302(1–3): 377-82.
[http://dx.doi.org/10.1016/j.colsurfa.2007.02.068]
[80]
Tavano L, Aiello R, Ioele G, Picci N, Muzzalupo R. Niosomes from glucuronic acid-based surfactant as new carriers for cancer therapy: preparation, characterization and biological properties. Colloids Surf B Biointerfaces 2014; 118: 7-13.
[http://dx.doi.org/10.1016/j.colsurfb.2014.03.016] [PMID: 24709252]
[81]
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]
[82]
Manosroi A, Wongtrakul P, Manosroi J, Sakai H, Sugawara F, Yuasa M. 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]
[83]
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]
[84]
Pando D, Gutiérrez G, Coca J, Pazos C. Preparation and characterization of niosomes containing resveratrol. J Food Eng 2013; 117(2): 227-34.
[http://dx.doi.org/10.1016/j.jfoodeng.2013.02.020]
[85]
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]
[86]
Ghafoorianfar S, Ghorani-Azam A, Mohajeri SA, Farzin D. Efficiency of nanoparticles for treatment of ocular infections: Systematic literature review. J Drug Deliv Sci Technol 2020; 57: 101765.
[http://dx.doi.org/10.1016/j.jddst.2020.101765]
[87]
B’Hymer C. Residual solvent testing: a review of gas-chromatographic and alternative techniques. Pharm Res 2003; 20(3): 337-44.
[http://dx.doi.org/10.1023/A:1022693516409] [PMID: 12669951]
[88]
Puranik S, Pai PNS, Rao G. Organic volatile impurities in pharmaceuticals. Indian J Pharm Sci 2007; 69(3): 352-9.
[http://dx.doi.org/10.4103/0250-474X.34542]
[89]
Anil M. Dwivedi Residual solvent analysis in pharmaceuticals. Pharmaceutical Technology Europe 2002.
[90]
Zhang S, Morris ME. Efflux transporters in drug excretion Drug Delivery. Hoboken, NJ, USA: John Wiley & Sons, Inc. 2005; pp. 381-410.
[http://dx.doi.org/10.1002/0471475734.ch18]
[91]
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]
[92]
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]
[93]
Vora B, Khopade AJ, Jain NK. Proniosome based transdermal delivery of levonorgestrel for effective contraception. J Control Release 1998; 54(2): 149-65.
[http://dx.doi.org/10.1016/S0168-3659(97)00100-4] [PMID: 9724902]
[94]
Hao YM, Li K. Entrapment and release difference resulting from hydrogen bonding interactions in niosome. Int J Pharm 2011; 403(1-2): 245-53.
[http://dx.doi.org/10.1016/j.ijpharm.2010.10.027] [PMID: 20971171]
[95]
Hu C, Rhodes DG. Proniosomes: a novel drug carrier preparation. Int J Pharm 1999; 185(1): 23-35.
[http://dx.doi.org/10.1016/S0378-5173(99)00122-2] [PMID: 10425362]
[96]
Biswal S, Murthy PN, Sahu J, Sahoo P. Vesicles of non-ionic surfactants (niosomes) and drug delivery potential. Int J Pharm Sci Nanotechnol 2008; 1(1): 1-8.
[http://dx.doi.org/10.37285/ijpsn.2008.1.1.1]
[97]
Florence AT, Cable C, Cassidy J, Kaye SB. Non-ionic surfactant vesicles as carriers of doxorubicintargeting of drugs 2. Springer US 1990; pp. 117-26.
[http://dx.doi.org/10.1007/978-1-4684-9001-5_11]
[98]
Hunter CA, Dolan TF, Coombs GH, Baillie AJ. Vesicular systems (niosomes and liposomes) for delivery of sodium stibogluconate in experimental murine visceral leishmaniasis. J Pharm Pharmacol 1988; 40(3): 161-5.
[http://dx.doi.org/10.1111/j.2042-7158.1988.tb05210.x] [PMID: 2899143]
[99]
Yoshioka T, Sternberg B, Florence A. Preparation and properties of vesicles (niosomes) of sorbitan monoesters (Span 20, 40, 60 and 80) and a sorbitan triester (Span 85). Int J Pharm 1994; 105(1): 1-6.
[http://dx.doi.org/10.1016/0378-5173(94)90228-3]
[100]
Toshimitsu Y, Florence AT. Vesicle (niosome)-in-water-in-oil (v/w/o) emulsions: An in vitro study. Int J Pharm 1994; 108(2): 117-23.
[http://dx.doi.org/10.1016/0378-5173(94)90322-0]
[101]
Shahiwala A, Misra A. Studies in topical application of niosomally entrapped Nimesulide. J Pharm Pharm Sci 2002; 5(3): 220-5.
[PMID: 12553889]
[102]
Radhi AA. Benazepril hydrochloride loaded niosomal formulation for oral delivery: Formulation and characterization. Int J Appl Pharm 2018; 10(5): 66-70.
[http://dx.doi.org/10.22159/ijap.2018v10i5.27564]
[103]
Arunothayanun P, Bernard M-S, 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]
[104]
Agarwal S, Tempta P, Kaushik P, Kaushik K. Role of Temperature on the Size of Niosomes Formed by the Ether Injection Method, Int. J Pharm Sci Nanotechnol 2019; 12: 4559-64.
[http://dx.doi.org/10.37285/ijpsn.2019.12.3.7]
[105]
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]
[106]
Oh YK, Nix DE, Straubinger RM. Formulation and efficacy of liposome-encapsulated antibiotics for therapy of intracellular Mycobacterium avium infection. Antimicrob Agents Chemother 1995; 39(9): 2104-11.
[http://dx.doi.org/10.1128/AAC.39.9.2104] [PMID: 8540724]
[107]
Malhotra M, Jain NK. Niosomes as drug carriers. Indian Drugs 1994; 31: 81.
[108]
Khandare: NIOSOMES-novel drug delivery system - Google Scholar. East Pharm 1994; 37: 61.
[109]
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]
[110]
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]
[111]
Parthasarathi G, Udupa N, Umadevi P, Pillai GK. 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]
[112]
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]
[113]
Zhang W, Ronca S, Mele E. Electrospun nanofibres containing antimicrobial plant extracts. Nanomaterials. MDPI AG 2017.
[http://dx.doi.org/10.3390/nano7020042]
[114]
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]
[115]
Velpandian T, Singh A, Jayasundar R. Herbal drugs for ophthalmic use pharmacology of ocular therapeutics. Springer International Publishing 2016; pp. 517-35.
[http://dx.doi.org/10.1007/978-3-319-25498-2_20]
[116]
Handjani RM, Ribier A, Vanlerberghe G, Zabotto A, Griat J. Cosmetic and pharmaceutical compositions containing niosomes and a water-soluble polyamide, and a process for preparing these compositions. US4830857A, 1985.
[117]
Kaul S, Gulati N, Verma D, Mukherjee S, Nagaich U. Role of Nanotechnology in Cosmeceuticals: A Review of Recent Advances. J Pharm (Cairo) 2018; 2018: 3420204.
[http://dx.doi.org/10.1155/2018/3420204] [PMID: 29785318]
[118]
Czajkowska-Kośnik A, Szekalska M, Winnicka K. Nanostructured lipid carriers: A potential use for skin drug delivery systems. Pharmacol Rep 2019; 71(1): 156-66.
[http://dx.doi.org/10.1016/j.pharep.2018.10.008] [PMID: 30550996]
[119]
Anghore D, Kulkarni GT. Development of novel nano niosomes as drug delivery system of spermacoce hispida extract and in vitro antituberculosis activity. Curr Nanomater 2017; 2(1): 17-23.
[http://dx.doi.org/10.2174/2405461502666170314151949]
[120]
Shijila P, Gowtham M. Formulation and evaluation of herbal niosomal gel for psoriasis like effect. World J Pharm Pharm Sci 2019; 8(4): 1052-79.
[121]
Rameshk M, Sharififar F, Mehrabani M, Pardakhty A, Farsinejad A, Mehrabani M. Proliferation and in vitro wound healing effects of the microniosomes containing narcissus tazetta L. Bulb Extract on Primary Human Fibroblasts (HDFs). Daru 2018; 26(1): 31-42.
[http://dx.doi.org/10.1007/s40199-018-0211-7] [PMID: 30209758]
[122]
Leelarungrayub J, Manorsoi J, Manorsoi A. Anti-inflammatory activity of niosomes entrapped with Plai oil (Zingiber cassumunar Roxb.) by therapeutic ultrasound in a rat model. Int J Nanomedicine 2017; 12: 2469-76.
[http://dx.doi.org/10.2147/IJN.S129131] [PMID: 28408818]
[123]
Desnita R, Hichmah HN, Luliana S. Effect of surfactant concentration on the entrapment efficiency niosomes aqueous extract of cassava leaves (Manihot esculenta Crantz). Asian J Pharm 2019; 13(4): 281.
[http://dx.doi.org/10.22377/ajp.v13i04.3397]
[124]
Priprem A, Janpim K, Nualkaew S, Mahakunakorn P. Topical niosome gel of zingiber cassumunar roxb. AAPS PharmSciTech 2016; 17(3): 631-9.
[http://dx.doi.org/10.1208/s12249-015-0376-z] [PMID: 26292930]
[125]
Un RN, Barlas FB, Yavuz M, Ag Seleci D, Seleci M, Gumus ZP. Phyto-Niosomes: in vitro assessment of the novel nanovesicles containing marigold extract. Int J Polym Mater Polym Biomater 2015; 64(17): 927-37.
[http://dx.doi.org/10.1080/00914037.2015.1030663]
[126]
Ali M, Abdel Motaal A, Ahmed MA, Alsayari A, El-Gazayerly ON. An in vivo study of Hypericum perforatum in a niosomal topical drug delivery system. Drug Deliv 2018; 25(1): 417-25.
[http://dx.doi.org/10.1080/10717544.2018.1431977] [PMID: 29382233]
[127]
PK S. Novel encapsulation of lycopene in niosomes and assessment of its anticancer activity. J Bioequivalence Bioavailab 2016; 8(5): 224-32.
[http://dx.doi.org/10.4172/jbb.1000300]
[128]
Barani M, Mirzaei M, Torkzadeh-Mahani M, Adeli-Sardou M. Evaluation of carum-loaded niosomes on breast cancer cells: Physicochemical properties, in vitro cytotoxicity, flow cytometric, DNA fragmentation and cell migration assay. Sci Rep 2019; 9(1): 7139.
[http://dx.doi.org/10.1038/s41598-019-43755-w] [PMID: 31073144]
[129]
Akbari J, Saeedi M, Enayatifard R, Morteza-Semnani K, Hassan Hashemi SM, Babaei A. Curcumin Niosomes (curcusomes) as an alternative to conventional vehicles: A potential for efficient dermal delivery. J Drug Deliv Sci Technol 2020; 60: 102035.
[http://dx.doi.org/10.1016/j.jddst.2020.102035]
[130]
Waddad AY, Abbad S, Yu F, et al. Formulation, characterization and pharmacokinetics of Morin hydrate niosomes prepared from various non-ionic surfactants. Int J Pharm 2013; 456(2): 446-58.
[http://dx.doi.org/10.1016/j.ijpharm.2013.08.040] [PMID: 23998955]
[131]
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]
[132]
Junyaprasert VB, Singhsa P, Jintapattanakit A. Influence of chemical penetration enhancers on skin permeability of ellagic acid-loaded niosomes. Asian J Pharm Sci 2013; 8(2): 110-7.
[http://dx.doi.org/10.1016/j.ajps.2013.07.014]
[133]
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]
[134]
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]
[135]
Sabry S. El hakim Ramadan A, Abd elghany M, Okda T, Hasan A. Formulation, characterization, and evaluation of the anti-tumor activity of nanosized galangin loaded niosomes on chemically induced hepatocellular carcinoma in rats. J Drug Deliv Sci Technol 2021; 61: 102163.
[http://dx.doi.org/10.1016/j.jddst.2020.102163]
[136]
Silindir M, Özer AY. Sterilization methods and the comparison of e-beam sterilization with gamma radiation sterilization. Fabad J Pharm Sci 2009; 34(1): 43.
[137]
Hasirci N. Micro and nano systems in biomedicine and drug deliverynanomaterials and nanosystems for biomedical applications. Dordrecht: Springer Netherlands 2007; pp. 1-26.
[http://dx.doi.org/10.1007/978-1-4020-6289-6_1]
[138]
Hofland HEJ, Bouwstra JA, Verhoef JC, et al. Safety aspects of non-ionic surfactant vesicles: a toxicity study related to the physicochemical characteristics of non-ionic surfactants. J Pharm Pharmacol 1992; 44(4): 287-94.
[http://dx.doi.org/10.1111/j.2042-7158.1992.tb03608.x] [PMID: 1355538]
[139]
Pape WJ, Pfannenbecker U, Hoppe U. Validation of the red blood cell test system as in vitro assay for the rapid screening of irritation potential of surfactants. Mol Toxicol 1987-1988-1988; 1(4): 525-36.
[PMID: 3509700]
[140]
Tavano L, Infante MR, Riya MA, Pinazo A, Vinardell MP, Mitjans M. Role of aggregate size in the hemolytic and antimicrobial activity of colloidal solutions based on single and gemini surfactants from arginine. Soft Matter 2013; 9(1): 306-19.
[http://dx.doi.org/10.1039/C2SM26670A]
[141]
Paolino D, Muzzalupo R, Ricciardi A, Celia C, Picci N, Fresta M. In vitro and in vivo evaluation of Bola-surfactant containing niosomes for transdermal delivery. Biomed Microdevices 2007; 9(4): 421-33.
[http://dx.doi.org/10.1007/s10544-007-9046-6] [PMID: 17252206]
[142]
Dov T, Enba Z, Yohan H, David S. Non surface active agent non polymeric agent hydro-alcoholic foamable compositions, breakable foams and their uses. US20200022912A1, 2020.
[143]
Stephen T, Andreu H. Composition comprising a polymer aggregate of lipid and surfactant. Google Patents JP6567575B2, 2019.
[144]
Chawrai S, Prasad S, Bajaj K. Coated particles and compositions comprising same. US20160128944A1, 2016.
[145]
Tamarkin D, Gazal E, Papiashvili I, Hazot Y, Schuz D, Keynan R. Surfactant-free water-free foamable compositions, breakable foams and gels and their uses. CA2776366C, 2010.
[146]
Cho BG, Kim DM, Kim SR. Stabilized niosomes of herbal extracts and color cosmetic materials containing them. KR1020020065958, 2007.

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