EFFECT OF SURFACTANTS ON THE FORMULATION OF NOVEL NANO VESICULAR FATTY ACID STRUCTURES OF MICONAZOLE; ENHANCED TOPICAL AND ANTIFUNGAL ACTIVITY

Main Article Content

Zainab Nisa
Aisha Sethi
Mudassar Mazher
Rida Sadiqque
Muhammad Mehboob-ur-Rehman

Keywords

Abstract

Background: Transcutaneous drug delivery is the most desirable method to improve efficacy and increase patient tolerance. Infections have increased over the past two decades. Superficial skin infections are treatable with conventional herbs while deep root infections cannot be treated due to disruption of the stratum corneum. the skin.


Objective: This study aimed to encapsulate the antifungal miconazole nitrate (MN) in advanced novasomes to improve skin penetration and clinically transform therapeutic improvement. Method : Novasomes with free fatty acid (FFA) as an internalization promoter were prepared by ethanol injection method and novasomes were characterized by percentage entrapment efficiency (EE%), particle size (PS), polydispersity index (PDI) and zeta potential (ZP), Scanning Electron Microscopy (SEM), Fourier Transform Infrared Spectroscopy (FTIR), Thermogravimetric Analysis (TGA), Differential Microscopy Calorimetry (DSC), MIC (Minimum Control Concentration), Agar Diffusion Method have been extensively studied. 


Result: The optimized MN7 formulation with 30 mg of lipid components and Span 60 oleic acid at a ratio of 2:1 (w/v) shows EE% = 97.45%, PS = 154 nm, PDI = 0.019, and ZP = ± 14 mV. In addition, MN7 showed greater inhibition of Candida albicans growth compared to MN suspension using the resazurin reduction test, The drug MN7 novasomes had significant results against C.albicans with a maximum zone of inhibition of 23.667±0.667mm. The MIC of MN7 was lower than that of unloaded novasomes (12.5 and 25 mg/mL respectively). The cell capacity remained above 85% which shows that it is non-toxic

Abstract 120 | PDF Downloads 61

References

1. Al-Maghrabi, P.M., et al., Influence of formulation variables on miconazole nitrate–loaded lipid based nanocarrier for topical delivery. Colloids and Surfaces B: Biointerfaces, 2020. 193: p. 111046.
2. Kumar, L., et al., Eradication of superficial fungal infections by conventional and novel approaches: a comprehensive review. Artificial cells, nanomedicine, and biotechnology, 2014. 42(1): p. 32-46.
3. Sinico, C. and A.M. Fadda, Vesicular carriers for dermal drug delivery. Expert opinion on drug delivery, 2009. 6(8): p. 813-825.
4. Tejada, G., et al., Nanoparticulated Systems Based on Natural Polymers Loaded with Miconazole Nitrate and Lidocaine for the Treatment of Topical Candidiasis. AAPS PharmSciTech, 2020. 21(7): p. 1-13.
5. Elmoslemany, R.M., et al., Propylene glycol liposomes as a topical delivery system for miconazole nitrate: comparison with conventional liposomes. AAPS PharmSciTech, 2012. 13(2): p. 723-731.
6. Ofokansi, K.C., F.C. Kenechukwu, and N.N. Ogwu, Design of novel miconazole nitrate transdermal films based on Eudragit RS100 and HPMC hybrids: preparation, physical characterization, in vitro and ex vivo studies. Drug delivery, 2015. 22(8): p. 1078-1085.
7. Sanna, V., G. Caria, and A. Mariani, Effect of lipid nanoparticles containing fatty alcohols having different chain length on the ex vivo skin permeability of Econazole nitrate. Powder Technology, 2010. 201(1): p. 32-36.
8. Jain, S., et al., Quality by design approach for formulation, evaluation and statistical optimization of diclofenac-loaded ethosomes via transdermal route. Pharmaceutical development and technology, 2015. 20(4): p. 473-489.
9. Jain, S., et al., Recent advances in lipid-based vesicles and particulate carriers for topical and transdermal application. Journal of pharmaceutical sciences, 2017. 106(2): p. 423-445.
10. Lee, C.M. and H.I. Maibach, Deep percutaneous penetration into muscles and joints. Journal of pharmaceutical sciences, 2006. 95(7): p. 1405-1413.
11. Agarwal, R. and O. Katare, Miconazole nitrate-loaded topical liposomes. Pharmaceutical technology, 2002. 26(11): p. 48-60.
12. Singh, A., et al., Comparative studies on skin permeation of miconazole using different novel carriers. Int J Pharm Sci Res, 2010. 1(9): p. 61-66.
13. Peira, E., et al., Positively charged microemulsions for topical application. International journal of pharmaceutics, 2008. 346(1-2): p. 119-123.
14. Müller, R.H., K. Mäder, and S. Gohla, Solid lipid nanoparticles (SLN) for controlled drug delivery–a review of the state of the art. European journal of pharmaceutics and biopharmaceutics, 2000. 50(1): p. 161-177.
15. Singh, R.P. and P. Ramarao, Accumulated polymer degradation products as effector molecules in cytotoxicity of polymeric nanoparticles. toxicological sciences, 2013. 136(1): p. 131-143.
16. Madsen, J.T., et al., Ethosome formulations of known contact allergens can increase their sensitizing capacity. Acta dermato-venereologica, 2010. 90(4): p. 374-378.
17. Mukherjee, S., S. Ray, and R. Thakur, Solid lipid nanoparticles: a modern formulation approach in drug delivery system. Indian journal of pharmaceutical sciences, 2009. 71(4): p. 349.
18. Bardania, H., et al., Encapsulation of eptifibatide in RGD-modified nanoliposomes improves platelet aggregation inhibitory activity. Journal of thrombosis and thrombolysis, 2017. 43(2): p. 184-193.
19. Singh, A., R. Malviya, and P.K. Sharma, Novasome-a breakthrough in pharmaceutical technology a review article. Adv Biol Res, 2011. 5(4): p. 184-189.
20. Gregoriadis, G., Engineering liposomes for drug delivery: progress and problems. Trends in biotechnology, 1995. 13(12): p. 527-537.
21. Chambers, M.A., et al., A single dose of killed Mycobacterium bovis BCG in a novel class of adjuvant (Novasome™) protects guinea pigs from lethal tuberculosis. Vaccine, 2004. 22(8): p. 1063-1071.
22. Kakkar, S. and I.P. Kaur, Spanlastics—a novel nanovesicular carrier system for ocular delivery. International journal of pharmaceutics, 2011. 413(1-2): p. 202-210.
23. Al-Mahallawi, A.M., O.M. Khowessah, and R.A. Shoukri, Nano-transfersomal ciprofloxacin loaded vesicles for non-invasive trans-tympanic ototopical delivery: in-vitro optimization, ex-vivo permeation studies, and in-vivo assessment. International journal of pharmaceutics, 2014. 472(1-2): p. 304-314.
24. Anjum, S., et al., Development of antimicrobial and scar preventive chitosan hydrogel wound dressings. International journal of pharmaceutics, 2016. 508(1-2): p. 92-101.
25. Abdelbary, A.A. and M.H. AbouGhaly, 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. International journal of pharmaceutics, 2015. 485(1-2): p. 235-243.
26. Abd-Elal, R.M., et al., Trans-nasal zolmitriptan novasomes: in-vitro preparation, optimization and in-vivo evaluation of brain targeting efficiency. Drug delivery, 2016. 23(9): p. 3374-3386.
27. Zeb, A., et al., Improved skin permeation of methotrexate via nanosized ultradeformable liposomes. International journal of nanomedicine, 2016. 11: p. 3813.
28. Tunney, M.M., et al., Rapid colorimetric assay for antimicrobial susceptibility testing of Pseudomonas aeruginosa. Antimicrobial agents and chemotherapy, 2004. 48(5): p. 1879-1881.
29. Riss, T.L., et al., Cell viability assays. Assay Guidance Manual [Internet], 2016.
30. Balouiri, M., M. Sadiki, and S.K. Ibnsouda, Methods for in vitro evaluating antimicrobial activity: A review. Journal of pharmaceutical analysis, 2016. 6(2): p. 71-79.

Most read articles by the same author(s)

<< < 1 2