Generic placeholder image

Current Nanomedicine

Editor-in-Chief

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

Research Article

Development and Evaluation of Topical Nanoemulgel Formulation of Tazarotene for Effective Treatment of Excision Wounds

Author(s): Shanta Mahajan, Kirandeep Kaur, Navdeep Saini, Tajeshwar Kumar Chaudhary, Lovedeep Nim and Neena Bedi*

Volume 12, Issue 2, 2022

Published on: 06 September, 2022

Page: [137 - 149] Pages: 13

DOI: 10.2174/2468187312666220820161123

Price: $65

Abstract

Aim: The aim of the present investigation entails the development and evaluation of a topical nanoemulgel formulation of tazarotene for the effective treatment of excision wounds.

Background: Tazarotene (TZR) is a retinoid derivative marketed for the treatment of acne and psoriasis and recent investigations indicate its excellent wound healing potential. Despite having magnificent wound healing potential, it suffers from pharmacokinetic limitations of low aqueous solubility and local bioavailability. Conventional marketed formulations of TZR have been reported to cause significant skin irritation and redness after its topical application. Keeping this in view, the present investigation was designed to develop nanoemulsion based gel with the intention to resolve the above mentioned pharmacokinetic and pharmacodynamic issues.

Objective: The goal of the study was to develop a stable and effective tazarotene-loaded nanoemulgel for treating excision wounds, which can also overcome the issues associated with the native drug.

Methods: Various oils, surfactants and co-surfactants were selected on the basis of their solubilisation potential for tazarotene. Pseudoternary phase plots were constructed to obtain a stable nanoemulsion region for various proportions of oil, surfactant: co-surfactant (Smix) and water and to determine optimized concentration ranges for the robust formulation of tazarotene. Various characterization parameters were studied to investigate the optimized formulation having desired attributes of nanoemulsion. Reducing power assay and phamacodymanic studies were conducted to determine the wound healing potential of developed nanoemulgel.

Results: TZR loaded nanoemulsion was successfully developed with nano-sized globules using tea tree oil with Tween 20 as surfactant and PEG 400 as co-surfactant. Prepared nanoemulsions were thermodynamically stable and were further gellified using Carbopol 940 as a biodegradable gelling polymer. Prepared gels showed good spreadability, drug permeation and stability. In vivo investigations demonstrated that optimized formulation successfully masked the irritant nature of tazarotene. Moreover, the pharmacodynamic evaluation of the developed nanoemulsion gel revealed its significant analgesic, anti-inflammatory properties in addition to its markable wound contraction as compared to control.

Conclusion: The results demonstrated the potential of developed nanoemulgel over marketed formulation to overcome the drawbacks like redness and erythema. It also showed significant healing efficacy over other tested formulations owing to the synergistic activity of tazarotene and tea tree oil encapsulated in the nanoemulsion system.

Keywords: Tazarotene, wounds, anti-inflammatory, nanoemulsion, nanoemulgel, antioxidant.

Graphical Abstract
[1]
Sen, C.K. Human wounds and its burden: An updated compendium of estimates. Adv. Wound Care (New Rochelle), 2019, 8(2), 39-48.
[http://dx.doi.org/10.1089/wound.2019.0946] [PMID: 30809421]
[2]
Lodhi, S.; Pawar, R.S.; Jain, A.P.; Singhai, A.K. Wound healing potential of Tephrosia purpurea (Linn.) Pers. in rats. J. Ethnopharmacol., 2006, 108(2), 204-210.
[http://dx.doi.org/10.1016/j.jep.2006.05.011] [PMID: 16806763]
[3]
Sandri, G.; Bonferoni, M.C.; D’Autilia, F. Wound dressings based on silver sulfadiazine solid lipid nanoparticles for tissue repairing. Eur. J. Pharm. Biopharm., 2013, 84(1), 84-90.
[http://dx.doi.org/10.1016/j.ejpb.2012.11.022] [PMID: 23207329]
[4]
Lopes, C.M.; Silva, J.; Oliveira, M.E.; Lúcio, M. Lipid-based colloidal carriers for topical application of antiviral drugs. In: In: Design of Nanostructures for Versatile Therapeutic Applications; Elsevier, 2018; pp. 565-622.
[http://dx.doi.org/10.1016/B978-0-12-813667-6.00014-0]
[5]
Indora, N.; Kaushik, D. Design, development and evaluation of ethosomal gel of fluconazole for topical fungal infection. Int J Eng Sci Invent Res Dev, 2015, 1(8), 280-306.
[6]
Ogarami, R.; Jain, P.; Sharma, S. Validated UV spectrophotometric method development for simultaneous estimation of tazarotene and hydroquinone in gel preparation. J. Pharm. Res., 2012, 5(4), 2273-2275.
[7]
Al Haj Zen, A.; Nawrot, D.A.; Howarth, A. The retinoid agonist tazarotene promotes angiogenesis and wound healing. Mol. Ther., 2016, 24(10), 1745-1759.
[http://dx.doi.org/10.1038/mt.2016.153] [PMID: 27480772]
[8]
Fischer-Levancini, C.; Sánchez-Regaña, M.; Llambí, F.; Collgros, H.; Expósito-Serrano, V.; Umbert-Millet, P. Nail psoriasis: Treatment with tazarotene 0.1% hydrophilic ointment. Actas Dermo. Sifiliográficas (English Edition) , 2012, 103(8), 725-728.
[http://dx.doi.org/10.1016/j.adengl.2012.09.014] [PMID: 22818395]
[9]
Marks, R. Pharmacokinetics and safety review of tazarotene. J. Am. Acad. Dermatol., 1998, 39(4), S134-S138.
[http://dx.doi.org/10.1016/S0190-9622(98)70310-X]
[10]
Sawant, K.; Mundada, P.; Sodani, D. Physicochemical characterization and clinical evaluation of a microemulsion system for topical delivery of tazarotene in Psoriasis. Micro Nanosyst., 2015, 7(2), 98-107.
[http://dx.doi.org/10.2174/1876402907666151103210822]
[11]
Dolgachev, V.A.; Ciotti, S.M.; Eisma, R. Nanoemulsion therapy for burn wounds is effective as a topical antimicrobial against gram negative and gram positive bacteria. J. Burn Care Res., 2016, 37(2), e104-e114.
[http://dx.doi.org/10.1097/BCR.0000000000000217] [PMID: 26182074]
[12]
McNeill, S.C.; Potts, R.O.; Francoeur, M.L. Local enhanced topical delivery (LETD) of drugs: Does it truly exist? Pharm. Res., 1992, 9(11), 1422-1427.
[http://dx.doi.org/10.1023/A:1015854728278] [PMID: 1475228]
[13]
Date, A.A.; Desai, N.; Dixit, R.; Nagarsenker, M. Self-nanoemulsifying drug delivery systems: Formulation insights, applications and advances. Nanomedicine (Lond.), 2010, 5(10), 1595-1616.
[http://dx.doi.org/10.2217/nnm.10.126] [PMID: 21143036]
[14]
Baroli, B.; López-Quintela, M.A.; Delgado-Charro, M.B.; Fadda, A.M.; Blanco-Méndez, J. Microemulsions for topical delivery of 8-methoxsalen. J. Control. Release, 2000, 69(1), 209-218.
[http://dx.doi.org/10.1016/S0168-3659(00)00309-6] [PMID: 11018558]
[15]
Aboalnaja, K.O.; Yaghmoor, S.; Kumosani, T.A.; McClements, D.J. Utilization of nanoemulsions to enhance bioactivity of pharmaceuticals, supplements, and nutraceuticals: Nanoemulsion delivery systems and nanoemulsion excipient systems. Expert Opin. Drug Deliv., 2016, 13(9), 1327-1336.
[http://dx.doi.org/10.1517/17425247.2016.1162154] [PMID: 26984045]
[16]
Abd-Allah, F.I.; Dawaba, H.M.; Mansour, A.; Samy, A.M. Evaluation of the anti-inflammatory and analgesic effects of piroxicam loaded microemulsion in topical formulations. Int. J. Pharm. Pharm. Sci., 2011, 3(2), 66-70.
[17]
Burt, S. Essential oils: Their antibacterial properties and potential applications in foods—a review. Int. J. Food Microbiol., 2004, 94(3), 223-253.
[http://dx.doi.org/10.1016/j.ijfoodmicro.2004.03.022] [PMID: 15246235]
[18]
Kim, H.J.; Chen, F.; Wu, C.; Wang, X.; Chung, H.Y.; Jin, Z. Evaluation of antioxidant activity of Australian tea tree (Melaleuca alternifolia) oil and its components. J. Agric. Food Chem., 2004, 52(10), 2849-2854.
[http://dx.doi.org/10.1021/jf035377d] [PMID: 15137824]
[19]
Pazyar, N.; Yaghoobi, R.; Bagherani, N.; Kazerouni, A. A review of applications of tea tree oil in dermatology. Int. J. Dermatol., 2013, 52(7), 784-790.
[http://dx.doi.org/10.1111/j.1365-4632.2012.05654.x] [PMID: 22998411]
[20]
Carson, C.F.; Mee, B.J.; Riley, T.V. Mechanism of action of Melaleuca alternifolia (tea tree) oil on Staphylococcus aureus determined by time-kill, lysis, leakage, and salt tolerance assays and electron microscopy. Antimicrob. Agents Chemother., 2002, 46(6), 1914-1920.
[http://dx.doi.org/10.1128/AAC.46.6.1914-1920.2002] [PMID: 12019108]
[21]
Chellapa, P.; Mohamed, A.T.; Keleb, E.I. Nanoemulsion and nanoemulgel as a topical formulation. IOSR J. Pharm., 2015, 5(10), 43-47.
[22]
Alves, D.A.; Machado, D.; Melo, A. Preparation of thermosensitive gel for controlled release of levofloxacin and their application in the treatment of multidrug-resistant bacteria. BioMed Res. Int., 2016, 2016, 1-10.
[http://dx.doi.org/10.1155/2016/9702129] [PMID: 27689094]
[23]
Craig, D.; Barker, S.A.; Banning, D.; Booth, S.W. An investigation into the mechanisms of self-emulsification using particle size analysis and low frequency dielectric spectroscopy. Int. J. Pharm., 1995, 114(1), 103-110.
[http://dx.doi.org/10.1016/0378-5173(94)00222-Q]
[24]
Shrivastav, A.; Mishra, A.K.; Ali, S.S.; Ahmad, A.; Abuzinadah, M.F.; Khan, N.A. In vivo models for assesment of wound healing potential: A systematic review. Wound Medicine, 2018, 20, 43-53.
[http://dx.doi.org/10.1016/j.wndm.2018.01.003]
[25]
Patel, T.; Soni, T.; Suhagia, B. Preparation and characterization of oxcarbazepine microemulsion. Egypt Pharm J, 2016, 15(3), 173.
[http://dx.doi.org/10.4103/1687-4315.197586]
[26]
Bali, V.; Ali, M.; Ali, J. Novel nanoemulsion for minimizing variations in bioavailability of ezetimibe. J. Drug Target., 2010, 18(7), 506-519.
[http://dx.doi.org/10.3109/10611860903548362] [PMID: 20067438]
[27]
Thomas, L.; Zakir, F.; Mirza, M.A.; Anwer, M.K.; Ahmad, F.J.; Iqbal, Z. Development of Curcumin loaded chitosan polymer based nanoemulsion gel: In vitro, ex vivo evaluation and In vivo wound healing studies. Int. J. Biol. Macromol., 2017, 101, 569-579.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.03.066] [PMID: 28322948]
[28]
Elmataeeshy, M.E.; Sokar, M.S.; Bahey-El-Din, M.; Shaker, D.S. Enhanced transdermal permeability of Terbinafine through novel nanoemulgel formulation; Development, in vitro and In vivo characterization. Fut J Pharm Sci, 2018, 4(1), 18-28.
[http://dx.doi.org/10.1016/j.fjps.2017.07.003]
[29]
Prakash, A.; Vadivel, V.; Rubini, D.; Nithyanand, P. Antibacterial and antibiofilm activities of linalool nanoemulsions against Salmonella typhimurium. Food Biosci., 2019, 28, 57-65.
[http://dx.doi.org/10.1016/j.fbio.2019.01.018]
[30]
Yıldırım A, Mavi A, Kara AA. Determination of antioxidant and antimicrobial activities of Rumex crispus L. extracts. J. Agric. Food Chem., 2001, 49(8), 4083-4089.
[http://dx.doi.org/10.1021/jf0103572] [PMID: 11513714]
[31]
Yogesh, K.; Jha, S.N.; Ahmad, T. Antioxidant potential of aqueous extract of some food grain powder in meat model system. J. Food Sci. Technol., 2014, 51(11), 3446-3451.
[http://dx.doi.org/10.1007/s13197-012-0804-y] [PMID: 26396344]
[32]
Naseri, N.; Valizadeh, H.; Zakeri-Milani, P. Solid lipid nanoparticles and nanostructured lipid carriers: Structure preparation and application. Adv. Pharm. Bull., 2015, 5(3), 305-313.
[http://dx.doi.org/10.15171/apb.2015.043] [PMID: 26504751]
[33]
Sharma, S.; Sahni, J.K.; Ali, J.; Baboota, S. Effect of high-pressure homogenization on formulation of TPGS loaded nanoemulsion of rutin-Pharmacodynamic and antioxidant studies. Drug Deliv., 2015, 22(4), 541-551.
[http://dx.doi.org/10.3109/10717544.2014.893382] [PMID: 24625264]
[34]
Bhattacharya, S.; Prajapati, B.G. Formulation and optimization of celecoxib nanoemulgel. Asian J. Pharm. Clin. Res., 2017, 10(8), 353.
[http://dx.doi.org/10.22159/ajpcr.2017.v10i8.19510]
[35]
Juma’a, K.M.; Ahmed, Z.A.; Numan, I.T.; Hussain, S.A.R. Dose-dependent anti-inflammatory effect of silymarin in experimental animal model of chronic inflammation. Afr. J. Pharm. Pharmacol., 2009, 3(5), 242-247.
[36]
Kaur, L.; Jain, S.K.; Manhas, R.K.; Sharma, D. Nanoethosomal formulation for skin targeting of amphotericin B: An in vitro and In vivo assessment. J. Liposome Res., 2015, 25(4), 294-307.
[http://dx.doi.org/10.3109/08982104.2014.995670] [PMID: 25547800]
[37]
Eyarefe, D.O.; Kuforiji, D.I.; Jarikre, T.A.; Emikpe, B.O. Enhanced electroscalpel incisional wound healing potential of honey in wistar rats. Int. J. Vet. Sci. Med., 2017, 5(2), 128-134.
[http://dx.doi.org/10.1016/j.ijvsm.2017.10.002] [PMID: 30255061]
[38]
Kommuru, T.R.; Gurley, B.; Khan, M.A.; Reddy, I.K. Self-emulsifying drug delivery systems (SEDDS) of coenzyme Q10: Formulation development and bioavailability assessment. Int. J. Pharm., 2001, 212(2), 233-246.
[http://dx.doi.org/10.1016/S0378-5173(00)00614-1] [PMID: 11165081]
[39]
Singh, Y.; Meher, J.G.; Raval, K. Nanoemulsion: Concepts, development and applications in drug delivery. J. Control. Release, 2017, 252, 28-49.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.008] [PMID: 28279798]
[40]
Pandya, B.R.; Chotai, N.P.; Suthar, R.M.; Patel, H.K. Formulation, in-vitro evaluation and optimization of nanoemulsion of raloxifene hydrochloride. Int J Pharm Res, 2017, 9(2), 39.
[41]
Algahtani, M.S.; Ahmad, M.Z.; Shaikh, I.A.; Abdel-Wahab, B.A.; Nourein, I.H.; Ahmad, J. Thymoquinone loaded topical nanoemulgel for wound healing: Formulation design and in-vivo evaluation. Molecules, 2021, 26(13), 3863.
[http://dx.doi.org/10.3390/molecules26133863] [PMID: 34202733]
[42]
Gokhale, J.P.; Mahajan, H.S.; Surana, S.J. Quercetin loaded nanoemulsion-based gel for rheumatoid arthritis: In vivo and in vitro studies. Biomed. Pharmacother., 2019, 112, 108622.
[http://dx.doi.org/10.1016/j.biopha.2019.108622] [PMID: 30797146]
[43]
Bakshi, P.; Jiang, Y.; Nakata, T.; Akaki, J.; Matsuoka, N.; Banga, A.K. Formulation development and characterization of nanoemulsion-based formulation for topical delivery of heparinoid. J. Pharm. Sci., 2018, 107(11), 2883-2890.
[http://dx.doi.org/10.1016/j.xphs.2018.07.015] [PMID: 30055224]
[44]
Arora, D.S.; Nim, L.; Kaur, H. Antimicrobial potential of callistemon lanceolatus seed extract and its statistical optimization. Appl. Biochem. Biotechnol., 2016, 180(2), 289-305.
[http://dx.doi.org/10.1007/s12010-016-2099-3] [PMID: 27179517]
[45]
Alhakamy, N.A.; Caruso, G.; Privitera, A. Fluoxetine ecofriendly nanoemulsion enhances wound healing in diabetic rats: In vivo efficacy assessment. Pharmaceutics, 2022, 14(6), 1133.
[http://dx.doi.org/10.3390/pharmaceutics14061133] [PMID: 35745706]
[46]
Madison, K.C. Barrier function of the skin: “la raison d’être” of the epidermis. J. Invest. Dermatol., 2003, 121(2), 231-241.
[http://dx.doi.org/10.1046/j.1523-1747.2003.12359.x] [PMID: 12880413]
[47]
Erol İ Üstündağ Okur N, Orak D, Sipahi H, Aydın A, Özer Ö. Tazarotene-loaded in situ gels for potential management of psoriasis: Biocompatibility, anti-inflammatory and analgesic effect. Pharm. Dev. Technol., 2020, 25(8), 909-918.
[http://dx.doi.org/10.1080/10837450.2020.1765180] [PMID: 32364036]
[48]
Berman, B.; Maderal, A.; Raphael, B. Keloids and hypertrophic scars: Pathophysiology, classification, and treatment. Dermatol. Surg., 2017, 43(1), S3-S18.
[http://dx.doi.org/10.1097/DSS.0000000000000819] [PMID: 27347634]
[49]
Gauglitz, G.G.; Korting, H.C.; Pavicic, T.; Ruzicka, T.; Jeschke, M.G. Hypertrophic scarring and keloids: Pathomechanisms and current and emerging treatment strategies. Mol. Med., 2011, 17(1-2), 113-125.
[http://dx.doi.org/10.2119/molmed.2009.00153] [PMID: 20927486]

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