Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Lipid Nanoparticles as a Skin Wound Healing Drug Delivery System: Discoveries and Advances

Author(s): Myla Lôbo de Souza, Widson Michael dos Santos, André Luiz Moreira Domingues de Sousa, Victor de Albuquerque Wanderley Sales, Fernanda Pontes Nóbrega, Marcos Victor Gregorio de Oliveira and Pedro José Rolim-Neto*

Volume 26, Issue 36, 2020

Page: [4536 - 4550] Pages: 15

DOI: 10.2174/1381612826666200417144530

Price: $65

Abstract

Chronic wounds are a remarkable cause of morbidity, requiring long-time treatments with a significant impact on the quality of life and high costs for public health. Although there are a variety of topical skin preparations commercially available, they have several limitations that frequently impair wound healing, such as drug instability, toxicity, limited time of action and ineffective skin permeation. In recent years, researchers have focused on the development of new effective treatments for wound healing and shown frequent interest in nanometric drug delivery systems to overcome such obstacles. In dermatology, lipid nanoparticles (LNPs) have received great attention from researchers due to their great functionalities, greater adhesion to the skin and film formation, enabling the hydration and maintenance of skin integrity, as well as present a more effective penetration through the skin barrier. This review provides an update on topical formulations based on Solid Lipid Nanoparticles (SLNs) and Nanostructured Lipid Carriers (NLCs) as wound healing treatments. Both SLNs and NLCs are able to increase solubility and stability of active pharmaceutical ingredients and increase skin penetration compared to the free drugs. Additionally, SLNs and NLCs can increase pharmacological activity, increase the release profile of the drugs, promote synergistic effects and improve the sensory properties of the final formulation. Topical dosage forms containing nanoparticles have been extensively evaluated for wound healing activity, mainly the dressings, films and scaffolds. Therefore, lipid nanoparticles have contributed in improving wound healing therapies when incorporated into other dosage forms with better efficacy and lesser adverse effects than conventional formulations.

Keywords: Nanocarriers, lipid nanoparticles, cicatrization, wound healing, topical use, skin penetration.

[1]
Cañedo-Dorantes L, Cañedo-Ayala M. Skin acute wound healing: A comprehensive review. Int J Inflamm 2019; 2019: 3706315.
[http://dx.doi.org/10.1155/2019/3706315 ] [PMID: 31275545]
[2]
Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res 2012; 49(1): 35-43.
[http://dx.doi.org/10.1159/000339613 ] [PMID: 22797712]
[3]
Sorg H, Tilkorn DJ, Hager S, Hauser J, Mirastschijski U. Skin wound healing: An update on the current knowledge and concepts. Eur Surg Res 2017; 58(1-2): 81-94.
[http://dx.doi.org/10.1159/000454919 ] [PMID: 27974711]
[4]
Wong VW, Gurtner GC. Tissue engineering for the management of chronic wounds: current concepts and future perspectives. Exp Dermatol 2012; 21(10): 729-34.
[http://dx.doi.org/10.1111/j.1600-0625.2012.01542.x ] [PMID: 22742728]
[5]
Zeng R, Lin C, Lin Z, et al. Approaches to cutaneous wound healing: basics and future directions. Cell Tissue Res 2018; 374(2): 217-32.
[http://dx.doi.org/10.1007/s00441-018-2830-1 ] [PMID: 29637308]
[6]
Takeo M, Lee W, Ito M. Wound healing and skin regeneration. Cold Spring Harb Perspect Med 2015; 5(1): a023267.
[http://dx.doi.org/10.1101/cshperspect.a023267 ] [PMID: 25561722]
[7]
Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol 2015; 173(2): 370-8.
[http://dx.doi.org/10.1111/bjd.13954 ] [PMID: 26175283]
[8]
Scotton MF, Miot HA, Abbade LPF. Factors that influence healing of chronic venous leg ulcers: a retrospective cohort. An Bras Dermatol 2014; 89(3): 414-22.
[http://dx.doi.org/10.1590/abd1806-4841.20142687 ] [PMID: 24937814]
[9]
Lanau-Roig A, Fabrellas N, Sáez-Rubio G, Wilson K. Time of chronic wound healing, as part of a prevalence and incidence study. Enferm Glob 2017; 16: 454-63.
[http://dx.doi.org/10.6018/eglobal.16.2.251311]
[10]
Lazarus G, Valle MF, Malas M, et al. Chronic venous leg ulcer treatment: future research needs. Wound Repair Regen 2014; 22(1): 34-42.
[http://dx.doi.org/10.1111/wrr.12102 ] [PMID: 24134795]
[11]
Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci 2016; 73(20): 3861-85.
[http://dx.doi.org/10.1007/s00018-016-2268-0 ] [PMID: 27180275]
[12]
Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration. Science 2017; 356: 1026-30.
[http://dx.doi.org/10.1126/science.aam7928]
[13]
Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med 2014; 6(265): 265sr6.
[http://dx.doi.org/10.1126/scitranslmed.3009337 ] [PMID: 25473038]
[14]
Sarabahi S. Recent advances in topical wound care. Indian J Plast Surg 2012; 45(2): 379-87.
[http://dx.doi.org/10.4103/0970-0358.101321 ] [PMID: 23162238]
[15]
Zhao R, Liang H, Clarke E, Jackson C, Xue M. Inflammation in chronic wounds. Int J Mol Sci 2016; 17(12): E2085.
[http://dx.doi.org/10.3390/ijms17122085 ] [PMID: 27973441]
[16]
Dhingra GA, Kaur M, Singh M, Aggarwal G, Nagpal M. Lock stock and barrel of wound healing. Curr Pharm Des 2019; 25(38): 4090-107.
[http://dx.doi.org/10.2174/1381612825666190926163431 ] [PMID: 31556852]
[17]
Powers JG, Morton LM, Phillips TJ. Dressings for chronic wounds. Dermatol Ther (Heidelb) 2013; 26(3): 197-206.
[http://dx.doi.org/10.1111/dth.12055 ] [PMID: 23742280]
[18]
Han G, Ceilley R. Chronic wound healing: A review of current management and treatments. Adv Ther 2017; 34(3): 599-610.
[http://dx.doi.org/10.1007/s12325-017-0478-y ] [PMID: 28108895]
[19]
Nicholas MN, Yeung J. Current status and future of skin substitutes for chronic wound healing. J Cutan Med Surg 2017; 21(1): 23-30.
[http://dx.doi.org/10.1177/1203475416664037 ] [PMID: 27530398]
[20]
Harding K, Queen D. Innovation in wound healing. Int Wound J 2017; 14(1): 5-5.
[http://dx.doi.org/10.1111/iwj.12718 ] [PMID: 28054467]
[21]
Fonseca-Santos B, Silva PB, Rigon RB, Sato MR, Chorilli M. Formulating SLN and NLC as innovative drug delivery systems for non-invasive routes of drug administration. Curr Med Chem 2019; 27(22): 3623-56.
[http://dx.doi.org/10.2174/0929867326666190624155938 ] [PMID: 31232233]
[22]
Amasya G, Sandri G, Onay-Besikci A, et al. Skin Localization of Lipid Nanoparticles (SLN/NLC): Focusing the influence of formulation parameters. Curr Drug Deliv 2016; 13(7): 1100-10.
[http://dx.doi.org/10.2174/1567201813666160104130505 ] [PMID: 26725723]
[23]
Xie G, Lu W, Lu D. Penetration of titanium dioxide nanoparticles through slightly damaged skin in vitro and in vivo. J Appl Biomater Funct Mater 2015; 13(4): e356-61.
[http://dx.doi.org/10.5301/jabfm.5000243 ] [PMID: 26616753]
[24]
Mauro M, Crosera M, Monai M, et al. Cerium oxide nanoparticles absorption through intact and damaged human skin. Molecules 2019; 24(20): 1-10.
[http://dx.doi.org/10.3390/molecules24203759 ] [PMID: 31635398]
[25]
Shirodkar RK, Kumar L, Mutalik S, Lewis S. Solid lipid nanoparticles and nanostructured lipid carriers: Emerging lipid based drug delivery systems. Pharm Chem J 2019; 53: 440-53.
[http://dx.doi.org/10.1007/s11094-019-02017-9]
[26]
Thakur K, Sharma G, Singh B, Katare OP. Topical drug delivery of anti-infectives employing lipid-based nanocarriers: dermatokinetics as an important tool. Curr Pharm Des 2018; 24(43): 5108-28.
[http://dx.doi.org/10.2174/1381612825666190118155843 ] [PMID: 30657036]
[27]
Vogt A, Wischke C, Neffe AT, Ma N, Alexiev U, Lendlein A. Nanocarriers for drug delivery into and through the skin - Do existing technologies match clinical challenges? J Control Release 2016; 242: 3-15.
[http://dx.doi.org/10.1016/j.jconrel.2016.07.027 ] [PMID: 27449743]
[28]
Garcês A, Amaral MH, Sousa Lobo JM, Silva AC. Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. Eur J Pharm Sci 2018; 112: 159-67.
[http://dx.doi.org/10.1016/j.ejps.2017.11.023 ] [PMID: 29183800]
[29]
Nava-Arzaluz MG, Piñón-Segundo E, Ganem-Rondero A. Lipid nanocarriers as skin drug delivery systems Nanoparticles Pharmacother Elsevier 2019; 311-90.
[30]
Bandopadhyay S, Manchanda S, Chandra A, Ali J, Deb PK. Overview of different carrier systems for advanced drug delivery Drug Deliv Syst 2020; 179-233.
[31]
Chenthamara D, Subramaniam S, Ramakrishnan SG, et al. Therapeutic efficacy of nanoparticles and routes of administration. Biomater Res 2019; 23: 20.
[http://dx.doi.org/10.1186/s40824-019-0166-x ] [PMID: 31832232]
[32]
Pandey A. An overview on advances in the nanocarriers drug delivery systems Adv Struct Mater Springer Verlag 2017; 62: 65-76.
[http://dx.doi.org/10.1007/978-81-322-3655-9_3]
[33]
Sanna V, Roggio AM, Siliani S, et al. Development of novel cationic chitosan-and anionic alginate-coated poly(D,L-lactide-co-glycolide) nanoparticles for controlled release and light protection of resveratrol. Int J Nanomedicine 2012; 7: 5501-16.
[http://dx.doi.org/10.2147/ijn.s36684 ] [PMID: 23093904]
[34]
Wu L, Zhang J, Watanabe W. Physical and chemical stability of drug nanoparticles. Adv Drug Deliv Rev 2011; 63(6): 456-69.
[http://dx.doi.org/10.1016/j.addr.2011.02.001 ] [PMID: 21315781]
[35]
Kovačević AB, Müller RH, Keck CM. Formulation development of lipid nanoparticles: improved lipid screening and development of tacrolimus loaded nanostructured lipid carriers (NLC). Int J Pharm 2019.118918.
[http://dx.doi.org/10.1016/j.ijpharm.2019.118918 ] [PMID: 31870954]
[36]
Lauterbach A, Müller-Goymann CC. Applications and limitations of lipid nanoparticles in dermal and transdermal drug delivery via the follicular route Eur J Pharm Biopharm 2015; 97(Pt A): 152-63.
[http://dx.doi.org/10.1016/j.ejpb.2015.06.020] [PMID: 26144664]
[37]
Rajpoot K. Solid Lipid Nanoparticles: A Promising nanomaterial in drug delivery. Curr Pharm Des 2019; 25(37): 3943-59.
[http://dx.doi.org/10.2174/1381612825666190903155321 ] [PMID: 31481000]
[38]
Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: Structure preparation and application. Adv Pharm Bull 2015; 5(3): 305-13.
[http://dx.doi.org/10.15171/apb.2015.043 ] [PMID: 26504751]
[39]
Gordillo-Galeano A, Mora-Huertas CE. Solid lipid nanoparticles and nanostructured lipid carriers: A review emphasizing on particle structure and drug release. Eur J Pharm Biopharm 2018; 133: 285-308.
[http://dx.doi.org/10.1016/j.ejpb.2018.10.017 ] [PMID: 30463794]
[40]
Müller RH, Alexiev U, Sinambela P, Keck CM. Nanostructured lipid carriers (NLC): The second generation of solid lipid nanoparticles Percutaneous Penetration Enhanc Chem Methods Penetration Enhanc Nanocarriers, Springer Berlin Heidelberg 2016; 161-85.
[41]
Rawal SU, Patel MM. Lipid nanoparticulate systems: Modern versatile drug carriers Elsevier Grumezescu, AM 2018; 49-138.
[42]
Das S, Ng WK, Tan RBH. Are nanostructured lipid carriers (NLCs) better than solid lipid nanoparticles (SLNs): development, characterizations and comparative evaluations of clotrimazole-loaded SLNs and NLCs? Eur J Pharm Sci 2012; 47(1): 139-51.
[http://dx.doi.org/10.1016/j.ejps.2012.05.010 ] [PMID: 22664358]
[43]
Rigon RB, Gonçalez ML, Severino P, et al. Solid lipid nanoparticles optimized by 22 factorial design for skin administration: Cytotoxicity in NIH3T3 fibroblasts. Colloids Surf B Biointerfaces 2018; 171: 501-5.
[http://dx.doi.org/10.1016/j.colsurfb.2018.07.065 ] [PMID: 30081382]
[44]
Ridolfi DM, Marcato PD, MacHado D, Silva RA, Justo GZ, Durán N. In vitro cytotoxicity assays of solid lipid nanoparticles in epithelial and dermal cells. J Phys Conf Ser 2011; 304.
[http://dx.doi.org/10.1088/1742-6596/304/1/012032]
[45]
Eiras F, Amaral MH, Silva R, Martins E, Lobo JMS, Silva AC. Characterization and biocompatibility evaluation of cutaneous formulations containing lipid nanoparticles. Int J Pharm 2017; 519(1-2): 373-80.
[http://dx.doi.org/10.1016/j.ijpharm.2017.01.045 ] [PMID: 28131849]
[46]
Newton AMJ, Kaur S. Solid lipid nanoparticles for skin and drug delivery Nanoarchitectonics Biomed Elsevier 2019; 295-334.
[47]
Shah R, Eldridge D, Palombo E, Harding I. Compos Struct 2015; 11-22.
[http://dx.doi.org/10.1007/978-3-319-10711-0_2]
[48]
Rawal SU, Patel MM. Lipid nanoparticulate systems. Elsevier Inc. 2018.
[http://dx.doi.org/10.1016/B978-0-12-813687-4.00002-5]
[49]
Dwivedi D, Dwivedi M, Malviya S, Singh V. Evaluation of wound healing, anti-microbial and antioxidant potential of Pongamia pinnata in wistar rats. J Tradit Complement Med 2016; 7(1): 79-85.
[http://dx.doi.org/10.1016/j.jtcme.2015.12.002 ] [PMID: 28053891]
[50]
Ghayempour S, Montazer M, Mahmoudi Rad M. Encapsulation of Aloe Vera extract into natural Tragacanth Gum as a novel green wound healing product Int J Biol Macromol 2016; 93(Pt A): 344-9.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.08.076] [PMID: 27590536]
[51]
Miranda M, Cruz MT, Vitorino C, Cabral C. Nanostructuring lipid carriers using Ridolfia segetum (L.) Moris essential oil. Mater Sci Eng C 2019; 103: 109804.
[http://dx.doi.org/10.1016/j.msec.2019.109804 ] [PMID: 31349527]
[52]
Pires FQ, da Silva JKR, Sa-Barreto LL, Gratieri T, Gelfuso GM, Cunha-Filho M. Lipid nanoparticles as carriers of cyclodextrin inclusion complexes: A promising approach for cutaneous delivery of a volatile essential oil. Colloids Surf B Biointerfaces 2019; 182: 110382.
[http://dx.doi.org/10.1016/j.colsurfb.2019.110382 ] [PMID: 31352250]
[53]
Gad HA, Abd El-Rahman FAA, Hamdy GM. Chamomile oil loaded solid lipid nanoparticles: A naturally formulated remedy to enhance the wound healing. J Drug Deliv Sci Technol 2019; 50: 329-38.
[http://dx.doi.org/10.1016/j.jddst.2019.01.008]
[54]
Bhaskar Rao A, Prasad E, Deepthi SS, et al. Wound healing: a new perspective on glucosylated tetrahydrocurcumin. Drug Des Devel Ther 2015; 9: 3579-88.
[http://dx.doi.org/10.2147/DDDT.S85041 ] [PMID: 26203224]
[55]
Trivedi MK, Gangwar M, Mondal SC, Jana S. Protective effects of tetrahydrocurcumin (THC) on fibroblast and melanoma cell lines in vitro: it’s implication for wound healing. J Food Sci Technol 2017; 54(5): 1137-45.
[http://dx.doi.org/10.1007/s13197-017-2525-8 ] [PMID: 28416863]
[56]
He P, Yan H, Zhao J, Gou M, Li X. An evaluation of the wound healing potential of tetrahydrocurcumin-loaded MPEG-PLA nanoparticles. J Biomater Appl 2019; 34(3): 315-25.
[http://dx.doi.org/10.1177/0885328219851195 ] [PMID: 31104542]
[57]
Kakkar V, Kaur IP, Kaur AP, Saini K, Singh KK. Topical delivery of tetrahydrocurcumin lipid nanoparticles effectively inhibits skin inflammation: in vitro and in vivo study. Drug Dev Ind Pharm 2018; 44(10): 1701-12.
[http://dx.doi.org/10.1080/03639045.2018.1492607 ] [PMID: 29938544]
[58]
Alihosseini F, Azarmi S, Ghaffari S, Haghighat S, Rezayat Sorkhabadi SM. Synergic antibacterial effect of curcumin with ampicillin; free drug solutions in comparison with SLN dispersions. Adv Pharm Bull 2016; 6(3): 461-5.
[http://dx.doi.org/10.15171/apb.2016.060 ] [PMID: 27766232]
[59]
Ghaffari S, Alihosseini F, Rezayat Sorkhabadi SM, et al. Nanotechnology in wound healing; Semisolid dosage forms containing curcumin-ampicillin solid lipid nanoparticles, in-vitro, ex-vivo and in-vivo characteristics. Adv Pharm Bull 2018; 8(3): 395-400.
[http://dx.doi.org/10.15171/apb.2018.046 ] [PMID: 30276135]
[60]
Ramos R, Silva JP, Rodrigues AC, et al. Wound healing activity of the human antimicrobial peptide LL37. Peptides 2011; 32(7): 1469-76.
[http://dx.doi.org/10.1016/j.peptides.2011.06.005 ] [PMID: 21693141]
[61]
Chereddy KK, Her C-H, Comune M, et al. PLGA nanoparticles loaded with host defense peptide LL37 promote wound healing. J Control Release 2014; 194: 138-47.
[http://dx.doi.org/10.1016/j.jconrel.2014.08.016 ] [PMID: 25173841]
[62]
Fumakia M, Ho EA. Nanoparticles encapsulated with LL37 and serpin A1 promotes wound healing and synergistically enhances antibacterial activity. Mol Pharm 2016; 13(7): 2318-31.
[http://dx.doi.org/10.1021/acs.molpharmaceut.6b00099 ] [PMID: 27182713]
[63]
Sandri G, Bonferoni MC, D’Autilia F, et al. 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]
[64]
Patel KK, Surekha DB, Tripathi M, et al. Antibiofilm potential of silver sulfadiazine-loaded nanoparticle formulations: A study on the effect of DNAse-i on microbial biofilm and wound healing activity. Mol Pharm 2019; 16(9): 3916-25.
[http://dx.doi.org/10.1021/acs.molpharmaceut.9b00527 ] [PMID: 31318574]
[65]
Silva EL, Carneiro G, De Araújo LA, et al. Solid lipid nanoparticles loaded with retinoic acid and lauric acid as an alternative for topical treatment of acne vulgaris. J Nanosci Nanotechnol 2015; 15(1): 792-9.
[http://dx.doi.org/10.1166/jnn.2015.9184 ] [PMID: 26328443]
[66]
Charoenputtakhun P, Opanasopit P, Rojanarata T, Ngawhirunpat T. All-trans retinoic acid-loaded lipid nanoparticles as a transdermal drug delivery carrier. Pharm Dev Technol 2014; 19(2): 164-72.
[http://dx.doi.org/10.3109/10837450.2013.763261 ] [PMID: 23356887]
[67]
Arantes VT, Faraco AAG, Ferreira FB, et al. Retinoic acid-loaded solid lipid nanoparticles surrounded by chitosan film support diabetic wound healing in in vivo study. Colloids Surf B Biointerfaces 2020; 188: 110749.
[http://dx.doi.org/10.1016/j.colsurfb.2019.110749 ] [PMID: 31927466]
[68]
Schwarz JC, Weixelbaum A, Pagitsch E, Löw M, Resch GP, Valenta C. Nanocarriers for dermal drug delivery: influence of preparation method, carrier type and rheological properties. Int J Pharm 2012; 437(1-2): 83-8.
[http://dx.doi.org/10.1016/j.ijpharm.2012.08.003 ] [PMID: 22903049]
[69]
Shukla T, Upmanyu N, Prakash Pandey S, Gosh D. Lipid nanocarriers. Elsevier Inc. 2018.
[http://dx.doi.org/10.1016/B978-0-12-813687-4.00001-3]
[70]
Borges RS, Keita H, Ortiz BLS, et al. Anti-inflammatory activity of nanoemulsions of essential oil from Rosmarinus officinalis L.: in vitro and in zebrafish studies. Inflammopharmacology 2018; 26(4): 1057-80.
[http://dx.doi.org/10.1007/s10787-017-0438-9 ] [PMID: 29404883]
[71]
Khezri K, Farahpour MR, Mounesi Rad S. Accelerated infected wound healing by topical application of encapsulated Rosemary essential oil into nanostructured lipid carriers. Artif Cells Nanomed Biotechnol 2019; 47(1): 980-8.
[http://dx.doi.org/10.1080/21691401.2019.1582539 ] [PMID: 30857435]
[72]
Khezri K, Reza M, Mounesi S. Efficacy of Mentha pulegium essential oil encapsulated into nanostructured lipid carriers as an in vitro antibacterial and infected wound healing agent. Colloids Surf A Physicochem Eng Asp 2020; 589: 124414.
[http://dx.doi.org/10.1016/j.colsurfa.2020.124414]
[73]
Ghodrati M, Farahpour MR, Hamishehkar H. Encapsulation of Peppermint essential oil in nanostructured lipid carriers: In-vitro antibacterial activity and accelerative effect on infected wound healing. Colloids Surf A Physicochem Eng Asp 2019; 564: 161-9.
[http://dx.doi.org/10.1016/j.colsurfa.2018.12.043]
[74]
Modarresi M, Farahpour M-R, Baradaran B. Topical application of Mentha piperita essential oil accelerates wound healing in infected mice model. Inflammopharmacology 2019; 27(3): 531-7.
[http://dx.doi.org/10.1007/s10787-018-0510-0 ] [PMID: 29980963]
[75]
Brahmi F, Abdenour A, Bruno M, et al. Chemical composition and in vitro antimicrobial, insecticidal and antioxidant activities of the essential oils of Mentha pulegium L. and Mentha rotundifolia (L.) Huds growing in Algeria. Ind Crops Prod 2016; 88: 96-105.
[http://dx.doi.org/10.1016/j.indcrop.2016.03.002]
[76]
Mahboubi M, Haghi G. Antimicrobial activity and chemical composition of Mentha pulegium L. essential oil. J Ethnopharmacol 2008; 119(2): 325-7.
[http://dx.doi.org/10.1016/j.jep.2008.07.023 ] [PMID: 18703127]
[77]
Alonso G, Brandão C, Pham KBT, Doust J. Aloe vera for treating acute and chronic wounds. Sao Paulo Med J 2014; 132(6): 382.
[http://dx.doi.org/10.1590/1516-3180.20141326T1 ] [PMID: 25351761]
[78]
Garcia-Orue I, Gainza G, Garcia-Garcia P, et al. Composite nanofibrous membranes of PLGA/Aloe vera containing lipid nanoparticles for wound dressing applications. Int J Pharm 2019; 556: 320-9.
[http://dx.doi.org/10.1016/j.ijpharm.2018.12.010 ] [PMID: 30553008]
[79]
Garcia-Orue I, Gainza G, Gutierrez FB, et al. Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. Int J Pharm 2017; 523(2): 556-66.
[http://dx.doi.org/10.1016/j.ijpharm.2016.11.006 ] [PMID: 27825864]
[80]
Tejada S, Manayi A, Daglia M, et al. Wound healing effects of curcumin: a short review. Curr Pharm Biotechnol 2016; 17(11): 1002-7.
[http://dx.doi.org/10.2174/1389201017666160721123109 ] [PMID: 27640646]
[81]
Akbik D, Ghadiri M, Chrzanowski W, Rohanizadeh R. Curcumin as a wound healing agent. Life Sci 2014; 116(1): 1-7.
[http://dx.doi.org/10.1016/j.lfs.2014.08.016 ] [PMID: 25200875]
[82]
Hussain Z, Thu HE, Ng SF, Khan S, Katas H. Nanoencapsulation, an efficient and promising approach to maximize wound healing efficacy of curcumin: A review of new trends and state-of-the-art. Colloids Surf B Biointerfaces 2017; 150: 223-41.
[http://dx.doi.org/10.1016/j.colsurfb.2016.11.036 ] [PMID: 27918967]
[83]
Mohanty C, Sahoo SK. Curcumin and its topical formulations for wound healing applications. Drug Discov Today 2017; 22(10): 1582-92.
[http://dx.doi.org/10.1016/j.drudis.2017.07.001 ] [PMID: 28711364]
[84]
Chen P, Zhang H, Cheng S, Zhai G, Shen C. Development of curcumin loaded nanostructured lipid carrier based thermosensitive in situ gel for dermal delivery. Colloids Surf A Physicochem Eng Asp 2016; 506: 356-62.
[http://dx.doi.org/10.1016/j.colsurfa.2016.06.054]
[85]
Pivetta TP, Simões S, Araújo MM, Carvalho T, Arruda C, Marcato PD. Development of nanoparticles from natural lipids for topical delivery of thymol: Investigation of its anti-inflammatory properties. Colloids Surf B Biointerfaces 2018; 164: 281-90.
[http://dx.doi.org/10.1016/j.colsurfb.2018.01.053 ] [PMID: 29413607]
[86]
Riella KR, Marinho RR, Santos JS, et al. Anti-inflammatory and cicatrizing activities of thymol, a monoterpene of the essential oil from Lippia gracilis, in rodents. J Ethnopharmacol 2012; 143(2): 656-63.
[http://dx.doi.org/10.1016/j.jep.2012.07.028 ] [PMID: 22885071]
[87]
Zhang EY, Gao B, Shi HL, et al. 20(S)-Protopanaxadiol enhances angiogenesis via HIF-1α-mediated VEGF secretion by activating p70S6 kinase and benefits wound healing in genetically diabetic mice. Exp Mol Med 2017; 49(10): e387.
[http://dx.doi.org/10.1038/emm.2017.151 ] [PMID: 29075038]
[88]
Kim MH, Kim KT, Sohn SY, et al. Formulation and evaluation of nanostructured lipid carriers (NLCs) of 20(s)-protopanaxadiol (PPD) by box-behnken design. Int J Nanomedicine 2019; 14: 8509-20.
[http://dx.doi.org/10.2147/IJN.S215835 ] [PMID: 31749618]
[89]
Sun D. Silicone elastomer gel impregnated with 20(S)-protopanaxadiol-loaded nanostructured lipid carriers for ordered diabetic ulcer recovery. Acta Pharmacol Sin 2019.
[http://dx.doi.org/10.1038/s41401-019-0288-7 ] [PMID: 31534201]
[90]
Niculae G, Lacatusu I, Badea N, Meghea A, Stan R. Influence of vegetable oil on the synthesis of bioactive nanocarriers with broad spectrum photoprotection. Cent Eur J Chem. Versita 2014; 12: 837-50.
[http://dx.doi.org/10.2478/s11532-014-0503-9]
[91]
Lacatusu I, Badea N, Badea G, et al. Advanced bioactive lipid nanocarriers loaded with natural and synthetic anti-inflammatory actives. Chem Eng Sci 2019; 200: 113-26.
[http://dx.doi.org/10.1016/j.ces.2019.01.044]
[92]
Istrati D, Lacatusu I, Bordei N, et al. Phyto-mediated nanostructured carriers based on dual vegetable actives involved in the prevention of cellular damage. Mater Sci Eng C 2016; 64: 249-59.
[http://dx.doi.org/10.1016/j.msec.2016.03.087 ] [PMID: 27127051]
[93]
Lacatusu I, Istrati D, Bordei N, et al. Synergism of plant extract and vegetable oils-based lipid nanocarriers: Emerging trends in development of advanced cosmetic prototype products. Mater Sci Eng C 2020; 108: 110412.
[http://dx.doi.org/10.1016/j.msec.2019.110412 ] [PMID: 31923989]
[94]
Garcia-Orue I, Gainza G, Girbau C, et al. LL37 loaded nanostructured lipid carriers (NLC): A new strategy for the topical treatment of chronic wounds. Eur J Pharm Biopharm 2016; 108: 310-6.
[http://dx.doi.org/10.1016/j.ejpb.2016.04.006 ] [PMID: 27080206]
[95]
Romić MD, Klarić MŠ, Lovrić J, et al. Melatonin-loaded chitosan/Pluronic® F127 microspheres as in situ forming hydrogel: An innovative antimicrobial wound dressing. Eur J Pharm Biopharm 2016; 107: 67-79.
[http://dx.doi.org/10.1016/j.ejpb.2016.06.013 ] [PMID: 27329001]
[96]
Duvnjak Romić M, Špoljarić D, Šegvić Klarić M, Cetina-Čižmek B, Filipović-Grčić J, Hafner A. Melatonin loaded lipid enriched chitosan microspheres - Hybrid dressing for moderate exuding wounds. J Drug Deliv Sci Technol 2019; 52: 431-9.
[http://dx.doi.org/10.1016/j.jddst.2019.05.004]
[97]
Romić MD, Sušac A, Lovrić J, Cetina-Čižmek B, Filipović-Grčić J, Hafner A. Evaluation of stability and in vitro wound healing potential of melatonin loaded (lipid enriched) chitosan based microspheres. Acta Pharm 2019; 69(4): 635-48.
[http://dx.doi.org/10.2478/acph-2019-0049 ] [PMID: 31639097]
[98]
Tezgel Ö, Distasio N, Laghezza-masci V, Taddei A, Szarpak-jankowska A, Auzély-velty R, et al. Collagen scaffold-mediated delivery of NLC / siRNA as wound healing materials. J Drug Deliv Sci Technol 2020; 55: 101421.
[http://dx.doi.org/10.1016/j.jddst.2019.101421]
[99]
Tezgel Ö, Szarpak-Jankowska A, Arnould A, Auzély-Velty R, Texier I. Chitosan-lipid nanoparticles (CS-LNPs): Application to siRNA delivery. J Colloid Interface Sci 2018; 510: 45-56.
[http://dx.doi.org/10.1016/j.jcis.2017.09.045 ] [PMID: 28934610]
[100]
Czech T, Lalani R, Oyewumi MO. Delivery systems as vital tools in drug repurposing. AAPS PharmSciTech 2019; 20(3): 116.
[http://dx.doi.org/10.1208/s12249-019-1333-z ] [PMID: 30771030]
[101]
El-Nahas M, Gawish H, Tarshoby M, State O. The impact of topical phenytoin on recalcitrant neuropathic diabetic foot ulceration. J Wound Care 2009; 18(1): 33-7.
[http://dx.doi.org/10.12968/jowc.2009.18.1.32146 ] [PMID: 19131916]
[102]
Shaw J, Hughes CM, Lagan KM, Stevenson MR, Irwin CR, Bell PM. The effect of topical phenytoin on healing in diabetic foot ulcers: a randomized controlled trial. Diabet Med 2011; 28(10): 1154-7.
[http://dx.doi.org/10.1111/j.1464-5491.2011.03309.x ] [PMID: 21480976]
[103]
Motawea A, Borg T, Abd El-Gawad AEGH. Topical phenytoin nanostructured lipid carriers: design and development Taylor & Francis 2018; 44.
[104]
Motawea A, Abd El-Gawad AEH, Borg T, Motawea M, Tarshoby M. The impact of topical phenytoin loaded nanostructured lipid carriers in diabetic foot ulceration. Foot 2019; 40: 14-21.
[http://dx.doi.org/10.1016/j.foot.2019.03.007 ] [PMID: 30999080]
[105]
Rezvanian M, Amin MCIM, Ng SF. Development and physicochemical characterization of alginate composite film loaded with simvastatin as a potential wound dressing. Carbohydr Polym 2016; 137: 295-304.
[http://dx.doi.org/10.1016/j.carbpol.2015.10.091 ] [PMID: 26686133]
[106]
Varshosaz J, Taymouri S, Minaiyan M, Rastegarnasab F, Baradaran A. Development and in vitro/in vivo evaluation of HPMC/chitosan gel containing simvastatin loaded self-assembled nanomicelles as a potent wound healing agent. Drug Dev Ind Pharm 2018; 44(2): 276-88.
[http://dx.doi.org/10.1080/03639045.2017.1391832 ] [PMID: 29043860]
[107]
Orgul D, Eroglu H, Hekimoglu S. Formulation and characterization of tissue scaffolds containing simvastatin loaded nanostructured lipid carriers for treatment of diabetic wounds. J Drug Deliv Sci Technol 2017; 41: 280-92.
[http://dx.doi.org/10.1016/j.jddst.2017.08.001]
[108]
Zhang J, Huang X, Wang L. Pioglitazone inhibits the expression of matrix metalloproteinase-9, a protein involved in diabetes-associated wound healing. Mol Med Rep 2014; 10(2): 1084-8.
[http://dx.doi.org/10.3892/mmr.2014.2277 ] [PMID: 24890117]
[109]
Vijay SK, Mishra M, Kumar H, Tripathi K. Effect of pioglitazone and rosiglitazone on mediators of endothelial dysfunction, markers of angiogenesis and inflammatory cytokines in type-2 diabetes. Acta Diabetol 2009; 46(1): 27-33.
[http://dx.doi.org/10.1007/s00592-008-0054-7 ] [PMID: 18758684]
[110]
Silva-Abreu M, Espinoza LC, Rodríguez-Lagunas MJ, et al. Human skin permeation studies with PPARγ agonist to improve its permeability and efficacy in inflammatory processes. Int J Mol Sci 2017; 18(12): E2548.
[http://dx.doi.org/10.3390/ijms18122548 ] [PMID: 29182532]
[111]
Sakai S, Sato K, Tabata Y, Kishi K. Local release of pioglitazone (a peroxisome proliferator-activated receptor γ agonist) accelerates proliferation and remodeling phases of wound healing. Wound Repair Regen 2016; 24(1): 57-64.
[http://dx.doi.org/10.1111/wrr.12376 ] [PMID: 26710090]
[112]
Natarajan J, Sanapalli BKR, Bano M, Singh SK, Gulati M, Karri VVSR. Nanostructured lipid carriers of pioglitazone loaded collagen/chitosan composite scaffold for diabetic wound healing. Adv Wound Care (New Rochelle) 2019; 8(10): 499-513.
[http://dx.doi.org/10.1089/wound.2018.0831 ] [PMID: 31737408]
[113]
Doktorovová S, Kovačević AB, Garcia ML, Souto EB. Preclinical safety of solid lipid nanoparticles and nanostructured lipid carriers: Current evidence from in vitro and in vivo evaluation. Eur J Pharm Biopharm 2016; 108: 235-52.
[http://dx.doi.org/10.1016/j.ejpb.2016.08.001 ] [PMID: 27519829]
[114]
Vairo C, Collantes M, Quincoces G, et al. Preclinical safety of topically administered nanostructured lipid carriers (NLC) for wound healing application: biodistribution and toxicity studies. Int J Pharm 2019; 569: 118484.
[http://dx.doi.org/10.1016/j.ijpharm.2019.118484 ] [PMID: 31260785]
[115]
Saporito F, Sandri G, Bonferoni MC, et al. Essential oil-loaded lipid nanoparticles for wound healing. Int J Nanomedicine 2017; 13: 175-86.
[http://dx.doi.org/10.2147/IJN.S152529 ] [PMID: 29343956]
[116]
Bruschi ML. de A Pereira RR, de Francisco LM. The use of propolis in micro/nanostructured pharmaceutical formulations. Recent Pat Drug Deliv Formul 2016; 10(2): 130-40.
[http://dx.doi.org/10.2174/1872211310666151230112616 ] [PMID: 26715146]
[117]
Rosseto HC, Toledo LAS, Francisco LMB, et al. Nanostructured lipid systems modified with waste material of propolis for wound healing: Design, in vitro and in vivo evaluation. Colloids Surf B Biointerfaces 2017; 158: 441-52.
[http://dx.doi.org/10.1016/j.colsurfb.2017.07.029 ] [PMID: 28728086]
[118]
Islam MT, Ali ES, Uddin SJ, et al. Andrographolide, a diterpene lactone from Andrographis paniculata and its therapeutic promises in cancer. Cancer Lett 2018; 420: 129-45.
[http://dx.doi.org/10.1016/j.canlet.2018.01.074 ] [PMID: 29408515]
[119]
Jia Y, Zhang H, Yang S, et al. Electrospun PLGA membrane incorporated with andrographolide-loaded mesoporous silica nanoparticles for sustained antibacterial wound dressing. Nanomedicine (Lond) 2018; 13(22): 2881-99.
[http://dx.doi.org/10.2217/nnm-2018-0099 ] [PMID: 30427768]
[120]
Sanad RAB, Abdel-Bar HM. Chitosan–hyaluronic acid composite sponge scaffold enriched with Andrographolide-loaded lipid nanoparticles for enhanced wound healing Elsevier Ltd 2017; 173.
[121]
Esposito E, Pecorelli A, Sguizzato M, et al. Production and characterization of nanoparticle based hyaluronate gel containing retinyl palmitate for wound healing. Curr Drug Deliv 2018; 15(8): 1172-82.
[http://dx.doi.org/10.2174/1567201815666180518123926 ] [PMID: 29779480]
[122]
Clares B, Calpena AC, Parra A, et al. Nanoemulsions (NEs), liposomes (LPs) and solid lipid nanoparticles (SLNs) for retinyl palmitate: effect on skin permeation. Int J Pharm 2014; 473(1-2): 591-8.
[http://dx.doi.org/10.1016/j.ijpharm.2014.08.001 ] [PMID: 25102113]
[123]
García-Honduvilla N, Cifuentes A, Ortega MA, et al. Immunomodulatory effect of local rhEGF treatment during tissue repair in diabetic ulcers. Endocr Connect 2018; 7(4): 584-94.
[http://dx.doi.org/10.1530/EC-18-0117 ] [PMID: 29592858]
[124]
Gainza G, Pastor M, Aguirre JJ, et al. A novel strategy for the treatment of chronic wounds based on the topical administration of rhEGF-loaded lipid nanoparticles: In vitro bioactivity and in vivo effectiveness in healing-impaired db/db mice. J Control Release 2014; 185: 51-61.
[http://dx.doi.org/10.1016/j.jconrel.2014.04.032 ] [PMID: 24794895]
[125]
Gainza G, Chu WS, Guy RH, et al. Development and in vitro evaluation of lipid nanoparticle-based dressings for topical treatment of chronic wounds. Int J Pharm 2015; 490(1-2): 404-11.
[http://dx.doi.org/10.1016/j.ijpharm.2015.05.075 ] [PMID: 26043822]
[126]
Gainza G, Bonafonte DC, Moreno B, et al. The topical administration of rhEGF-loaded nanostructured lipid carriers (rhEGF-NLC) improves healing in a porcine full-thickness excisional wound model. J Control Release 2015; 197: 41-7.
[http://dx.doi.org/10.1016/j.jconrel.2014.10.033 ] [PMID: 25449803]

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