309
Views
0
CrossRef citations to date
0
Altmetric
Review Article

Exploring the use of niosomes in cosmetics for efficient dermal drug delivery

ORCID Icon, , &
Pages 708-718 | Received 19 Mar 2023, Accepted 02 Jul 2023, Published online: 14 Jul 2023

References

  • Abd-Elbary A, El-Laithy HM, Tadros MI. 2008. Sucrose stearate-based proniosome-derived niosomes for the nebulisable delivery of cromolyn sodium. Int J Pharm. 357(1–2):189–198. doi: 10.1016/j.ijpharm.2008.01.056.
  • Abu-Huwaij R, Al-Assaf S, Mousli F, Kutkut M, Al-Bashtawi A. 2020. Perceptive review on properties of iron oxide nanoparticles and their antimicrobial and anticancer activity. Sys Rev Pharm. 11(8):418–431.
  • Abu-Huwaij R, Al-Assaf SF, Hamed R. 2022. Recent exploration of nanoemulsions for drugs and cosmeceuticals delivery. J Cosmet Dermatol. 21(9):3729–3740. doi: 10.1111/jocd.14704.
  • Ahmed S, Kassem MA, Sayed S. 2020. Bilosomes as promising nanovesicular carriers for improved transdermal delivery: construction, in vitro optimization, ex vivo permeation and in vivo evaluation. Int J Nanomedicine. 15:9783–9798. PMID: 33324052; PMCID: PMC7733410. doi: 10.2147/IJN.S278688.
  • Agirre M, Zarate J, Puras G, Ojeda E, Pedraz JL. 2015. Improving transfection efficiency of ultrapure oligochitosan/DNA polyplexes by medium acidification. Drug Deliv. 22(1):100–110. doi: 10.3109/10717544.2013.871373.
  • Ag Seleci D, Seleci M, Walter J-G, Stahl F, Scheper T. 2016. Niosomes as nanoparticular drug carriers: fundamentals and recent applications. J Nanomater. 2016:1–13. doi: 10.1155/2016/7372306.
  • Ag Seleci D, Maurer V, Stahl F, Scheper T, Garnweitner G. 2019. Rapid microfluidic preparation of niosomes for targeted drug delivery. IJMS. 20(19):4696. doi: 10.3390/ijms20194696.
  • Akbari J, Saeedi M, Zangabad PS, Rahimzadeh P, Kouhi M, Akhtari J, Karimi M. 2019. Niosomes as a modern and promising drug delivery system: recent patents and emerging drugs. Recent Pat Drug Deliv Formul. 13(3):188–200.
  • Alagawany M, Abd El-Hack ME, Farag MR, Gopi M, Karthik K, Malik YS, Dhama K. 2017. Rosmarinic acid: modes of action, medicinal values and health benefits. Anim Health Res Rev. 18(2):167–176. doi: 10.1017/S1466252317000081.
  • Alam MA, Al-Jenoobi FI, Al-Mohizea AM, Ali R. 2019. Niosomes as a potential carrier for topical delivery of bioactives for management of skin disorders. Curr Pharm Des. 25(17):1916–1930.
  • Alhassan A, Ali M, Tariq M, Ali M, Khalid N. 2017. Niosomes: a review of preparation, characterization, and applications. Drug Dev Ind Pharm. 43(9):1514–1530.
  • Ali M, Alhassan A, Tariq M, Ali M, Khalid N. 2017. Niosomes: a promising drug delivery system. Drug Deliv. 24(1):14–26.
  • Aljuffali IA, Lin C-H, Yang S-C, Alalaiwe A, Fang J-Y. 2022. Nanoencapsulation of tea catechins for enhancing skin absorption and therapeutic efficacy. AAPS PharmSciTech. 23(6):187. doi: 10.1208/s12249-022-02344-3.
  • Alqahtani MA. 2017. Niosomes as novel drug delivery system: a review. Int J Pharm Sci Res. 8(6):2320–2330.
  • Al-Mahallawi AM, Abdelbary AA, Aburahma MH. 2015. Investigating the potential of employing bilosomes as a novel vesicular carrier for transdermal delivery of tenoxicam. Int J Pharm. 485(1–2):329–340. doi: 10.1016/j.ijpharm.2015.03.033.
  • Aparajay P, Dev A. 2022. Functionalized niosomes as a smart delivery device in cancer and fungal infection. Eur J Pharm Sci. 168:106052. doi: 10.1016/j.ejps.2021.106052.
  • Arafa MG, Ghalwash D, El-Kersh DM, Elmazar MM. 2018. Propolis-based niosomes as oromuco-adhesive films: a randomized clinical trial of a therapeutic drug delivery platform for the treatment of oral recurrent aphthous ulcers. Sci Rep. 8(1):18056. doi: 10.1038/s41598-018-37157-7.
  • Bhavani GD, Lakshmi PV. 2020. 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. 6(1):100. doi: 10.1186/s43094-020-00117-y.
  • Balakrishnan P, Shanmugam S, Lee WS, Lee WM, Kim JO, Oh DH, Kim DD, Kim JS, Yoo BK, Choi HG, et al. 2009. Formulation and in vitro assessment of minoxidil niosomes for enhanced skin delivery. Int J Pharm. 377(1–2):1–8. doi: 10.1016/j.ijpharm.2009.04.020.
  • Bartelds R, Nematollahi MH, Pols T, Stuart MCA, Pardakhty A, Asadikaram G, Poolman B. 2018. Niosomes, an alternative for liposomal delivery. PLOS One. 13(4):e0194179. doi: 10.1371/journal.pone.0194179.
  • Bhardwaj P, Tripathi P, Gupta R, Pandey S. 2020. Niosomes: a review on niosomal research in the last decade. J Drug Delivery Sci Technol. 56(Part A):101581. doi: 10.1016/j.jddst.2020.101581.
  • Budhiraja A, Dhingra G. 2015. Development and characterization of a novel antiacne niosomal gel of rosmarinic acid. Drug Deliv. 22(6):723–730. doi: 10.3109/10717544.2014.903010.
  • Carballo-Pedrares N, Kattar A, Concheiro A, Alvarez-Lorenzo C, Rey-Rico A. 2021. Niosomes-based gene delivery systems for effective transfection of human mesenchymal stem cells. Mater Sci Eng C Mater Biol Appl. 128:112307. doi: 10.1016/j.msec.2021.112307.
  • Chauhan MK, Sahoo PK, Rawat AS, Singh A, Bamrara A, Sharma D. 2015. Bilosomes: A novel approach to meet the challenges in oral immunization. Recent Pat Drug Deliv Formul. 9(3):201–212. doi: 10.2174/1872211309666150615111803.
  • Chen RP, Chavda VP, Patel AB, Chen ZS. 2022. Phytochemical delivery through transfersome (Phytosome): an advanced transdermal drug delivery for complementary medicines. Front Pharmacol. 13:850862. doi: 10.3389/fphar.2022.850862.
  • Chen S, Hanning S, Falconer J, Locke M, Wen J. 2019. Recent advances in non-ionic surfactant vesicles (niosomes): fabrication, characterization, pharmaceutical and cosmetic applications. Eur J Pharm Biopharm. 144:18–39. doi: 10.1016/j.ejpb.2019.08.015.
  • Cheng R, Xu T, Wang C, Gan C. 2021. The stabilization and antioxidant performances of coenzyme Q10-loaded niosomes coated by PEG and chitosan. J Mol Liq. 325:115194. doi: 10.1016/j.molliq.2020.115194.
  • Choi MH, Yang SH, Kim DS, Kim ND, Shin HJ, Liu K. 2021. Novel quercetin derivative of 3,7-dioleylquercetin shows less toxicity and highly potent tyrosinase inhibition activity. IJMS. 22(8):4264. doi: 10.3390/ijms22084264.
  • Danhui Li, Nataly Martini, Mengyang Liu, James R. Falconer, Michelle Locke, Zimei Wu, Jingyuan Wen. 2021. Non-ionic surfactant vesicles as a carrier system for dermal delivery of (+)-Catechin and their antioxidant effects. J Drug Target. 29(3):310–322. doi: 10.1080/1061186X.2020.1835923.
  • Durak S, Esmaeili Rad M, Alp Yetisgin A, Eda Sutova H, Kutlu O, Cetinel S, Zarrabi A. 2020. Niosomal drug delivery systems for ocular disease-recent advances and future prospects. Nanomaterials . 10(6):1191. doi: 10.3390/nano10061191.
  • El-Malah Y, Elnaggar YS, El-Refaie WM. 2017. A promising approach to improve skin penetration and retard inflammation: niosomal gel of piroxicam. Artif Cells Nanomed Biotechnol. 45(4):729–737.
  • Ertekin ZC, Bayindir ZS, Yuksel N. 2015. Stability studies on piroxicam encapsulated niosomes. Curr Drug Deliv. 12(2):192–199. doi: 10.2174/1567201811666140723115852.
  • Falconer RJ. 2016. Applications of isothermal titration calorimetry—the research and technical developments from 2011 to 2015. J Mol Recognit. 29(10):504–515. doi: 10.1002/jmr.2550.
  • Gallego I, Villate-Beitia I, Martínez-Navarrete G, Menéndez M, López-Méndez T, Soto-Sánchez C, Zárate J, Puras G, Fernández E, Pedraz JL. 2019. Non-viral vectors based on cationic niosomes and minicircle DNA technology enhance gene delivery efficiency for biomedical applications in retinal disorders. Nanomedicine. 17:308–318. doi: 10.1016/j.nano.2018.12.018.
  • Ge X, Wei M, He S, Yuan W-E. 2019. Advances of non-ionic surfactant vesicles (Niosomes) and their application in drug delivery. Pharmaceutics. 11(2):55. doi: 10.3390/pharmaceutics11020055.
  • Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. 2018. Niosome: a promising nanocarrier for natural drug delivery through blood-brain barrier. Adv Pharmacol Sci. 2018:6847971. doi: 10.1155/2018/6847971.
  • Ghasemiyeh P, Mohammadi-Samani S. 2020. Potential of nanoparticles as permeation enhancers and targeted delivery options for skin: advantages and disadvantages. Drug Des Devel Ther. 14:3271–3289. doi: 10.2147/DDDT.S264648.
  • Gogoi P, Gogoi M. 2020. Niosomal drug delivery system: a promising approach for skin drug delivery. JDrug DelivTher. 10(5):120–127.
  • Grijalvo S, Puras G, Zárate J, Sainz-Ramos M, Qtaish NAL, López T, Mashal M, Attia N, Díaz D, Pons R, et al. 2019. Cationic niosomes as non-viral vehicles for nucleic acids: challenges and opportunities in gene delivery. Pharmaceutics. 11(2):50. doi: 10.3390/pharmaceutics11020050.
  • Hajhashemi V, Rajabi P, Mardani M. 2019. Beneficial effects of pumpkin seed oil as a topical hair growth promoting agent in a mice model. Avicenna J Phytomed. 9(6):499–504. doi: 10.22038/AJP.2019.13463.
  • Hamimed S, Jabberi M, Chatti A. 2022. Nanotechnology in drug and gene delivery. Naunyn Schmiedebergs Arch Pharmacol. 395(7):769–787. Jul doi: 10.1007/s00210-022-02245-z.
  • Hasan AA, Madkor H, Wageh S. 2013. Formulation and evaluation of metformin hydrochloride-loaded niosomes as controlled release drug delivery system. Drug Delivery. 20(3–4):120–126. doi: 10.3109/10717544.2013.779332.
  • Ho CH, Sood T, Zito PM. 2022. Androgenetic Alopecia. Treasure Island. StatPearls Publishing LLC; https://www.ncbi.nlm.nih.gov/books/NBK430924/.
  • Huang L, Teng W, Cao J, Wang J. 2022. Liposomes as delivery system for applications in meat products. Foods. 11(19):3017. doi: 10.3390/foods11193017.
  • Islam MA, Firdous J, Choi YJ, Yun CH, Cho CS. 2014. Regulation of endocytosis by non-viral vectors for efficient gene activity. J Biomed Nanotechnol. 10(1):67–80. doi: 10.1166/jbn.2014.1682.
  • Jain AK, et al. 2013. Bilosomes: a novel approach to the targeted delivery of drugs and vaccines. J Drug Targeting. 21(10):911–922.
  • Jeong WY, Kwon M, Choi HE, Kim KS. 2021. Recent advances in transdermal drug delivery systems: a review. Biomater Res. 25(1):24. doi: 10.1186/s40824-021-00226-6.
  • Jiang X, Zhao H, Li W. 2022. Microneedle-mediated transdermal delivery of drug-carrying nanoparticles. Front Bioeng Biotechnol. 10:840395. doi: 10.3389/fbioe.2022.840395.
  • Jigar V, Puja V, Dhaval R, Paresh P. 2011. Development of topical niosomal gel of benzoyl peroxide. Int Sch Res Notices. 2011(6):1–6. doi: 10.5402/2011/503158.
  • Joseph MA, Nikolai AS, Stephen MM. 2020. The applied anatomy of human skin: a model for regeneration. Wound Med. 28:100179. doi: 10.1016/j.wndm.2020.100179.
  • Joy C, Nair SK, Krishna Kumar K, Dineshkumar B. 2021. Niosomes as nano-carrier based targeted drug delivery system. J Drug Delivery Ther. 11(4-S):166–170. doi: 10.22270/jddt.v11i4-S.4907.
  • Junyaprasert VB, Teeranachaideekul V, Supaperm T. 2008. Effect of charged and non-ionic membrane additives on physicochemical properties and stability of niosomes. AAPS PharmSciTech. 9(3):851–859. doi: 10.1208/s12249-008-9121-1.
  • Kar M, Saquib M, Jain DK. 2020. Formulation development and evaluation of aspasomes containing skin whitening agent. Manipal J Pharm Sci 6(1): 47–53, article 8. https://impressions.manipal.edu/mjps/vol6/iss1/8.
  • Karimi M, Sahandi Zangabad P, Baghaee-Ravari S, Ghazadeh M, Mirshekari H, Hamblin MR. 2017. Smart nanostructures for cargo delivery: uncaging and activating by light. J Am Chem Soc. 139(13):4584–4610. doi: 10.1021/jacs.6b08313.
  • Kaul S, Gulati N, Verma D, Mukherjee S, Nagaich U. 2018. Role of nanotechnology in cosmeceuticals: a review of recent advances. J Pharm. 2018:3420204. doi: 10.1155/2018/3420204.
  • Kauslya A, Borawake PD, Shinde JV, Chavan RS. 2021. Niosomes: a novel carrier drug delivery system. J Drug Delivery Ther. 11(1):162–170. doi: 10.22270/jddt.v11i1.4479.
  • Keck CM, Müller RH. 2013. Nanotoxicological classification system (NCS) - a guide for the risk-benefit assessment of nanoparticulate drug delivery systems. Eur J Pharm Biopharm. 84(3):445–448. doi: 10.1016/j.ejpb.2013.01.001.
  • Leung AWY, Amador C, Wang LC, Mody UV, Bally MB. 2019. What drives innovation: the Canadian touch on liposomal therapeutics. Pharmaceutics. 11(3):124. doi: 10.3390/pharmaceutics11030124.
  • Limongi T, Susa F, Marini M, Allione M, Torre B, Pisano R, di Fabrizio E. 2021. Lipid-based nanovesicular drug delivery systems. Nanomaterials. 11(12):3391. doi: 10.3390/nano11123391.
  • Li J, Xiang H, Zhang Q, Miao X. 2022. Polysaccharide-based transdermal drug delivery. Pharmaceuticals. 15(5):602. doi: 10.3390/ph15050602.
  • Li Z, Fang X, Yu D. 2021. Transdermal drug delivery systems and their use in obesity treatment. IJMS. 22(23):12754. doi: 10.3390/ijms222312754.
  • Lu B, Huang Y, Chen Z, Ye J, Xu H, Chen W, Long X. 2019. Niosomal nanocarriers for enhanced skin delivery of quercetin with functions of anti-tyrosinase and antioxidant. Molecules. 24(12):2322. doi: 10.3390/molecules24122322.
  • Mahmoud K, Mohamed M, Amr I, Dina L. 2018. An overview on niosomes: a drug nanocarrier. Drug Des Int Prop Int J. 1(5):143–151. DDIPIJ. MS.ID.000125.
  • Mali N, Darandale S, Vavia P. 2013. Niosomes as a vesicular carrier for topical administration of minoxidil: formulation and in vitro assessment. Drug Deliv Transl Res. 3(6):587–592. doi: 10.1007/s13346-012-0083-1.
  • Manconi M, Sinico C, Valenti D, Lai F, Fadda AM. 2006. Niosomes as carriers for tretinoin. III. a study into the in vitro cutaneous delivery of vesicle-incorporated tretinoin. Int J Pharm. 311(1–2):11–19. doi: 10.1016/j.ijpharm.2005.11.045.
  • Manosroi A, Jantrawut P, Manosroi J. 2008. Anti-inflammatory activity of gel containing novel elastic niosomes entrapped with diclofenac diethylammonium. Int J Pharm. 360(1–2):156–163. doi: 10.1016/j.ijpharm.2008.04.033.
  • Marianecci C, Di Marzio L, Rinaldi F, Celia C, Paolino D, Alhaique F, Esposito S, Carafa M. 2014. Niosomes from 80s to present: the state of the art. Adv Colloid Interface Sci. 205:187–206. doi: 10.1016/j.cis.2013.11.018.
  • Mawazi SM, Ann TJ, Widodo RT. 2022. Application of niosomes in cosmetics: a systematic review. Cosmetics. 9(6):127. doi: 10.3390/cosmetics9060127.
  • McLaughlin J, Watterson S, Layton AM, Bjourson AJ, Barnard E, McDowell A. 2019. Propionibacterium acnes and acne vulgaris: new Insights from the integration of population genetic, multi-omic, biochemical and host-microbe studies. Microorganisms. 7(5):128. doi: 10.3390/microorganisms7050128.
  • Mehrarya M, Gharehchelou B, Haghighi Poodeh S, Jamshidifar E, Karimifard S, Farasati Far B, Akbarzadeh I, Seifalian A. 2022. Niosomal formulation for antibacterial applications. J Drug Target. 30(5):476–493. doi: 10.1080/1061186X.2022.2032094.
  • Miyazawa T, Itaya M, Burdeos GC, Nakagawa K, Miyazawa T. 2021. a critical review of the use of surfactant-coated nanoparticles in nanomedicine and food nanotechnology. Int J Nanomedicine. 16:3937–3999. doi: 10.2147/IJN.S298606.
  • Moghassemi S, Hadjizadeh A. 2014. Nano-niosomes as nanoscale drug delivery systems: an illustrated review. J Control Release. 185:22–36. doi: 10.1016/j.jconrel.2014.04.015.
  • Mohamadi N, Soltanian S, Raeiszadeh M, Moeinzadeh M, Ohadi M, Sharifi F, Pardakhty A, Sharififar F. 2022. Characteristics and in vitro anti skin aging activity and UV radiation protection of morin loaded in niosomes. J Cosmet Dermatol. 21(11):6326–6335. doi: 10.1111/jocd.15273.
  • Mohammadi S, Pardakhty A, Khalili M, Fathi R, Rezaeizadeh M, Farajzadeh S, Mohebbi A, Aflatoonian M. 2019. Niosomal benzoyl peroxide and clindamycin lotion versus niosomal clindamycin lotion in treatment of acne vulgaris: a randomized clinical trial. Adv Pharm Bull. 9(4):578–583. doi: 10.15171/apb.2019.066.
  • Mohsen AM, AbouSamra MM, ElShebiney SA. 2017. Enhanced oral bioavailability and sustained delivery of glimepiride via niosomal encapsulation: in-vitro characterization and in-vivo evaluation. Drug Dev Ind Pharm. 43(8):1254–1264. doi: 10.1080/03639045.2017.1310224.
  • Momekova DB, Gugleva VE, Petrov PD. 2021. Nanoarchitectonics of multifunctional niosomes for advanced drug delivery. ACS Omega. 6(49):33265–33273. doi: 10.1021/acsomega.1c05083.
  • Muzzalupo R, Tavano L. 2015. Niosomal drug delivery for transdermal targeting: recent advances. RRTD. 4:23–33. doi: 10.2147/RRTD.S64773.
  • Muzzalupo R, Mazzotta E. 2019. Do niosomes have a place in the field of drug delivery? Expert Opin Drug Deliv. 16(11):1145–1147. doi: 10.1080/17425247.2019.1663821.
  • Nasseri B. 2005. Effect of cholesterol and temperature on the elastic properties of niosomal membranes. Int J Pharm. 300(1–2):95–101. doi: 10.1016/j.ijpharm.2005.05.009.
  • Nowroozi F, Almasi A, Javidi J, Haeri A, Dadashzadeh S. 2018. Effect of surfactant type, cholesterol content and various downsizing methods on the particle size of niosomes. Iran J Pharm Res. 17(Suppl2):1–11.
  • Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. 2022. Liposomes: structure, composition, types, and clinical applications. Heliyon. 8(5):e09394. doi: 10.1016/j.heliyon.2022.e09394.
  • Obeid M, Khadra I, Aljabali A, Amawi H, Ferro V. 2022. Characterisation of niosome nanoparticles prepared by microfluidic mixing for drug delivery. Int J Pharm X. 4:100137. doi: 10.1016/j.ijpx.2022.100137.
  • Ojeda E, Agirre M, Villate-Beitia I, Mashal M, Puras G, Zarate J, Pedraz JL. 2016a. Elaboration and physicochemical characterization of niosome-based nioplexes for gene delivery purposes. Methods Mol Biol. 1445:63–75.
  • Ojeda E, Puras G, Agirre M, Zarate J, Grijalvo S, Eritja R, DiGiacomo L, Caracciolo G, Pedraz JL. 2016b. The role of helper lipids in the intracellular disposition and transfection efficiency of niosome formulations for gene delivery to retinal pigment epithelial cells. Int J Pharm. 503(1–2):115–126. doi: 10.1016/j.ijpharm.2016.02.043.
  • Ojeda E, Puras G, Agirre M, Zárate J, Grijalvo S, Pons R, Eritja R, Martinez-Navarrete G, Soto-Sanchez C, Fernández E, et al. 2015. Niosomes based on synthetic cationic lipids for gene delivery: the Influence of polar head-groups on the transfection efficiency in HEK-293, ARPE-19 and MSC-D1 cells. Org Biomol Chem. 13(4):1068–1081. doi: 10.1039/c4ob02087a.
  • O'Leary EI, Jiang Z, Strub MP, Lee JC. 2018. Effects of phosphatidylcholine membrane fluidity on the conformation and aggregation of N-terminally acetylated α-synuclein. J Biol Chem. 293(28):11195–11205. doi: 10.1074/jbc.RA118.002780.
  • Oswald M, Platscher M, Geissler S, Goepferich A. 2016. HPLC analysis as a tool for assessing targeted liposome composition. Int J Pharm. 497(1–2):293–300. doi: 10.1016/j.ijpharm.2015.11.014.
  • Opatha SAT, Titapiwatanakun V, Chutoprapat R. 2020. Transfersomes: a promising nanoencapsulation technique for transdermal drug delivery. Pharmaceutics. 12(9):855. doi: 10.3390/pharmaceutics12090855.
  • Paluszkiewicz P, Martuszewski A, Zaręba N, Wala K, Banasik M, Kepinska M. 2021. The application of nanoparticles in diagnosis and treatment of kidney diseases. IJMS. 23(1):131. doi: 10.3390/ijms23010131.
  • Pandey M, Choudhury H, Gorain B, Tiong SQ, Wong GYS, Chan KX, They X, Chieu WS. 2021. Site-specific vesicular drug delivery system for skin cancer: a novel approach for targeting. Gels. 7(4):218. doi: 10.3390/gels7040218.
  • Pandey R, Bhairam M, Shukla SS, Gidwani B. 2021. Colloidal and vesicular delivery system for herbal bioactive constituents. Daru. 29(2):415–438. doi: 10.1007/s40199-021-00403-x.
  • Patil S, Sandberg A, Heckert E, Self W, Seal S. 2007. Protein adsorption and cellular uptake of cerium oxide nanoparticles as a function of zeta potential. Biomaterials. 28(31):4600–4607. doi: 10.1016/j.biomaterials.2007.07.029.
  • Perez Ruiz de Garibay A. 2016. Endocytosis in gene therapy with non-viral vectors. Wien Med Wochenschr. 166(7–8):227–235. doi: 10.1007/s10354-016-0450-5.
  • Puras G, Martínez-Navarrete G, Mashal M, Zárate J, Agirre M, Ojeda E, Grijalvo S, Eritja R, Diaz-Tahoces A, Avilés-Trigueros M, et al. 2015. Protamine/DNA/Niosome ternary nonviral vectors for gene delivery to the retina: the role of protamine. Mol Pharm. 12(10):3658–3671. doi: 10.1021/acs.molpharmaceut.5b00422.
  • Puras G, Mashal M, Zárate J, Agirre M, Ojeda E, Grijalvo S, Eritja R, Diaz-Tahoces A, Martínez Navarrete G, Avilés-Trigueros M, et al. 2014. A novel cationic niosome formulation for gene delivery to the retina. J Control Release. 174:27–36. doi: 10.1016/j.jconrel.2013.11.004.
  • Puras G, Zarate J, Diaz-Tahoces A, Aviles-Trigueros M, Fernandez E, Pedraz JL. 2013. Oligochitosan polyplexes as carriers for retinal gene delivery. Eur J Pharm Sci. 48(1–2):323–331. doi: 10.1016/j.ejps.2012.11.009.
  • Qu F, Geng R, Liu Y, Zhu J. 2022. Advanced nanocarrier- and microneedle-based transdermal drug delivery strategies for skin diseases treatment. Theranostics. 12(7):3372–3406. doi: 10.7150/thno.69999.
  • Radmard A, Saeedi M, Morteza-Semnani K, Hashemi SMH, Nokhodchi A. 2021. An eco-friendly and green formulation in lipid nanotechnology for delivery of a hydrophilic agent to the skin in the treatment and management of hyperpigmentation complaints: arbutin noisome (Arbusome). Colloids Surf B Biointerfaces. 201:111616. doi: 10.1016/j.colsurfb.2021.111616.
  • Ramadon D, McCrudden MTC, Courtenay AJ, Donnelly RF. 2022. Enhancement strategies for transdermal drug delivery systems: current trends and applications. Drug Deliv Transl Res. 12(4):758–791. doi: 10.1007/s13346-021-00909-6.
  • Salvioni L, Morelli L, Ochoa E, Labra M, Fiandra L, Palugan L, Prosperi D, Colombo M. 2021. The emerging role of nanotechnology in skin care. Adv Colloid Interface Sci. 293:102437. doi: 10.1016/j.cis.2021.102437.
  • Khoee S, Yaghoobian M. 2017. Chapter 6 - Niosomes: a novel approach in modern drug delivery systems, editor(s): Ecaterina Andronescu, Alexandru Mihai Grumezescu, in micro and nano technologies, nanostructures for drug delivery, Elsevier; p. 207-237. ISBN 9780323461436 doi: 10.1016/B978-0-323-46143-6.00006-3.
  • Shah VM, Nguyen DX, Patel P, Cote B, Al-Fatease A, Pham Y, Huynh MG, Woo Y, Alani AW. 2019. Liposomes produced by microfluidics and extrusion: a comparison for scale-up purposes. Nanomedicine. 18:146–156. doi: 10.1016/j.nano.2019.02.019.
  • Sharma D, Ali AAE, Aate JR. 2018. Niosomes as novel drug delivery system: review article. PT. 6(3):58–65. doi: 10.29161/PT.v6.i3.2018.58.
  • Sharma A, Kuhad A, Bhandari R. 2022a. Novel nanotechnological approaches for treatment of skin-aging. J Tissue Viability. 31(3):374–386. doi: 10.1016/j.jtv.2022.04.010.
  • Sharma R, Dua JS, Parsad DN. 2022b. An overview on niosomes: novel pharmaceutical drug delivery system. J Drug Delivery Ther. 12(2-s):171–177. doi: 10.22270/jddt.v12i2-S.5264.
  • Shatalebi MA, Mostafavi SA, Moghaddas A. 2010. Niosome as a drug carrier for topical delivery of N-acetyl glucosamine. Res Pharm Sci. 5(2):107–117.
  • Slocum M, Garcia SF, McKoy JM. 2019. Cancer drug toxicity: moving from patient to Survivor. In: McKoy, J., West, D. editors. Cancer policy: pharmaceutical safety. cancer treatment and research, vol 171. Cham: Springer; p. 107–118. doi: 10.1007/978-3-319-43896-2_8.
  • Sultana N, Ahmed M. 2021. Niosomes: an innovative drug delivery system. J Controlled Release. 330:205–216.
  • Tan Y, Li X, Zhang L, Chen L. 2021. Niosomes as promising drug delivery systems for topical and transdermal administration. J Drug Deliv Sci Technol. 63:102371.
  • Teeranachaideekul V, Parichatikanond W, Junyaprasert VB, Morakul B. 2022. Pumpkin seed oil-loaded niosomes for topical application: 5α-reductase inhibitory, anti-inflammatory, and in vivo anti-hair loss effects. Pharmaceuticals. 15(8):930. doi: 10.3390/ph15080930.
  • Toncic RJ, Jakasa I, Hadzavdic SL, Goorden SM, Vlugt KJG, Stet FS, Balic A, Petkovic M, Pavicic B, Zuzul K, et al. 2020. Altered Levels of Sphingosine, Sphinganine and their ceramides in atopic dermatitis are related to skin barrier function, disease severity and local cytokine Milieu. IJMS. 21(6):1958. PMID: 32183011; PMCID: PMC7139865. doi: 10.3390/ijms21061958.
  • Tristram-Nagle S, Nagle JF. 2004. Lipid bilayers: thermodynamics, structure, fluctuations, and interactions. Chem Phys Lipids. 127(1):3–14. doi: 10.1016/j.chemphyslip.2003.09.002.
  • Villate-Beitia I, Gallego I, Martínez-Navarrete G, Zárate J, López-Méndez T, Soto-Sánchez C, Santos-Vizcaíno E, Puras G, Fernández E, Pedraz JL. 2018. Polysorbate 20 non-ionic surfactant enhances retinal gene delivery efficiency of cationic niosomes after intravitreal and subretinal administration. Int J Pharm. 550(1–2):388–397. doi: 10.1016/j.ijpharm.2018.07.035.
  • Wen H, Jung H, Li X. 2015. Drug delivery approaches in addressing clinical pharmacology-related issues: opportunities and challenges. Aaps J. 17(6):1327–1340. doi: 10.1208/s12248-015-9814-9.
  • Yasamineh S, Yasamineh P, Kalajahi HG, Gholizadeh O, Yekanipour Z, Afkhami H, Eslami M, Kheirkhah AH, Taghizadeh M, Yazdani Y, et al. 2022. A state-of-the-art review on the recent advances of niosomes as a targeted drug delivery system. Int J Pharm. 624:121878. doi: 10.1016/j.ijpharm.2022.121878.
  • Yeo PL, Lim CL, Chye SM, Anna L, Koh YK. 2018. Niosomes: a review of their structure, properties, methods of preparation, and medical applications. Asian Biomedicine. 11(4):301–314. doi: 10.1515/abm-2018-0002.
  • Zaid Alkilani A, Abu-Zour H, Alshishani A, Abu-Huwaij R, Basheer HA, Abo-Zour H. 2022. Formulation and evaluation of niosomal alendronate sodium encapsulated in polymeric microneedles: in vitro studies, stability study and cytotoxicity study. Nanomaterials. 12(20):3570. PMID: 36296760; PMCID: PMC9611853. doi: 10.3390/nano12203570.
  • Zarei A. 2020. Niosomes as novel drug delivery systems: potential applications and toxicity evaluations. J Biomed Sci. 27(1):39.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.