Generic placeholder image

Current Drug Therapy

Editor-in-Chief

ISSN (Print): 1574-8855
ISSN (Online): 2212-3903

Review Article

Drug/Vehicle Impacts and Formulation Centered Stratagems for Enhanced Transdermal Drug Permeation, Controlled Release and Safety: Unparalleled Past and Recent Innovations-An Overview

Author(s): Balamurugan Manickam*, Rajesh Sreedharan and Kumarappan Chidambaram

Volume 14, Issue 3, 2019

Page: [192 - 209] Pages: 18

DOI: 10.2174/1574885514666190212113754

Abstract

Transdermal drug delivery systems (TDDS) are one of the fascinating unconventional drug delivery systems offering plentiful advantages of which patient compliance is of paramount importance. However, as a matter of fact, the transdermal delivery of drug molecules is absolutely a tedious job which is precisely influenced by a number of factors including penetration barrier properties of the skin, drug characteristics formulation allied issues, etc. Over the years, innumerable tremendous efforts have been made in transporting the drugs through the skin into the systemic circulation by noteworthy tactics. This paper discusses such revolutionary formulation based techniques that have been endeavored in achieving the enhanced skin permeation of drugs, controlled release, and safety.

Keywords: Formulation strategies, drug and vehicle impacts, transdermal drug delivery, enhanced drug permeation, controlled release, safety.

Graphical Abstract
[1]
Alkilani AZ, McCrudden MT, Donnelly RF. Transdermal drug delivery: Innovative pharmaceutical developments based on disruption of the barrier properties of the stratum corneum. Pharmaceutics 2015; 7(4): 438-70.
[2]
Yie WC. Novel Drug Delivery System, 2nd revised and expanded Marcel Dekker:. New York. 1992.
[3]
Ita K. Transdermal drug delivery: progress and challenges. J Drug Deliv Sci Technol 2014; 24(3): 245-50.
[4]
Singh MC, Naik AS, Sawant S. Transdermal drug delivery systems with major emphasis on transdermal patches: a review. J Pharm Res 2010; 3(10): 2537-43.
[5]
Han T, Das DB. Potential of combined ultrasound and microneedles for enhanced transdermal drug permeation: a review. Eur J Pharm Biopharm 2015; 89: 312-28.
[6]
Pastore MN, Kalia YN, Horstmann M, Roberts MS. Transdermal patches: history, development and pharmacology. Br J Pharmacol 2015; 172(9): 2179-209.
[7]
Thakur G, Singh A, Singh I. Formulation and evaluation of transdermal composite films of chitosan-montmorillonite for the delivery of curcumin. Int J Pharm Investig 2016; 6(1): 23-31.
[8]
Schäfer-Korting M, Mehnert W, Korting HC. Lipid nanoparticles for improved topical application of drugs for skin diseases. Adv Drug Deliv Rev 2007; 59(6): 427-43.
[9]
Chandrashekar NS, Shobha Rani RH. Physicochemical and pharmacokinetic parameters in drug selection and loading for transdermal drug delivery. Indian J Pharm Sci 2008; 70(1): 94-6.
[10]
N’Da DD. Prodrug strategies for enhancing the percutaneous absorption of drugs. Molecules 2014; 19(12): 20780-807.
[11]
Hadgraft J, Guy RH. Transdermal drug delivery. Marcel Dekker 2003.
[12]
Aliyar H, Schalau II, G. Recent developments in silicones for topical and transdermal drug delivery 2015.
[13]
Zhu Z, Zhai Y, Zhang N, Leng D, Ding P. The development of polycarbophil as a bioadhesive material in pharmacy. Asian J Pharm Sci 2013; 8(4): 218-27.
[14]
Balamurugan M. Chitosan: a perfect polymer used in fabricating gene delivery and novel drug delivery systems. Int J Pharm Pharm Sci 2012; 4(3): 54-6.
[15]
Tu Y, Wang X, Lu Y, et al. Promotion of the transdermal delivery of protein drugs by N-trimethyl chitosan nanoparticles combined with polypropylene electret. Int J Nanomedicine 2016; 11: 5549-61.
[16]
Smith J, Wood E, Dornish M. Effect of chitosan on epithelial cell tight junctions. Pharm Res 2004; 21(1): 43-9.
[17]
He W, Guo X, Xiao L, Feng M. Study on the mechanisms of chitosan and its derivatives used as transdermal penetration enhancers. Int J Pharm 2009; 382(1-2): 234-43.
[18]
Can AS, Erdal MS, Güngör S, Özsoy Y. Optimization and characterization of chitosan films for transdermal delivery of ondansetron. Molecules 2013; 18(5): 5455-71.
[19]
Park YJ, Lee YM, Lee JY, Seol YJ, Chung CP, Lee SJ. Controlled release of platelet-derived growth factor-BB from chondroitin sulfate-chitosan sponge for guided bone regeneration. J Control Release 2000; 67(2-3): 385-94.
[20]
Ramanathan S, Block LH. The use of chitosan gels as matrices for electrically-modulated drug delivery. J Control Release 2001; 70(1-2): 109-23.
[21]
Vishwanath B, Shivakumar HR, Sheshappa RK, et al. In vitro release study of metoprolol succinate from the bioadhesive films of pullulan-polyacrylamide blends. Int J Polym Mater 2012; 61(4): 300-7.
[22]
Kanikkannan N, Andega S, Burton S, Babu RJ, Singh M. Formulation and in vitro evaluation of transdermal patches of melatonin. Drug Dev Ind Pharm 2004; 30(2): 205-12.
[23]
Banerjee S, Chattopadhyay P, Ghosh A, Datta P, Veer V. Aspect of adhesives in transdermal drug delivery systems. Int J Adhes Adhes 2014; 50: 70-84.
[24]
Bhattacharya SS, Banerjee S. Skin permeation of buflomedil form adhesive matrix patches. J Adhes Sci Technol 2015; 29(10): 925-42.
[25]
Hua L-l, Li Y, Wang Q, Hu Y-n, Zhao Z-f. Fabrication of amphiphilic hot-melt pressure sensitive adhesives for transdermal drug delivery. J Adhes Sci Technol 2012; 26(8-9): 1109-22.
[26]
Wang C, Liu R, Tang X, Han W. A drug-in-adhesive matrix based on thermoplastic elastomer: evaluation of percutaneous absorption, adhesion, and skin irritation. AAPS PharmSciTech 2012; 13(4): 1179-89.
[27]
Yu Z, Liang Y, Liang W. Development and evaluation of α-asarone transdermal patches based on hot-melt pressure-sensitive adhesives. AAPS PharmSciTech 2013; 14(1): 294-300.
[28]
Ma J, Wang C, Luo H, Zhu Z, Wu Y, Wang H. Design and evaluation of a monolithic drug-in-adhesive patch for testosterone based on styrene-isoprene-styrene block copolymer. J Pharm Sci 2013; 102(7): 2221-34.
[29]
Karande P, Jain A, Ergun K, Kispersky V, Mitragotri S. Design principles of chemical penetration enhancers for transdermal drug delivery. Proc Natl Acad Sci USA 2005; 102(13): 4688-93.
[30]
Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol 2008; 26(11): 1261-8.
[31]
Kim Y-C, Ludovice PJ, Prausnitz MR. Transdermal delivery enhanced by magainin pore-forming peptide. J Control Release 2007; 122(3): 375-83.
[32]
Chen Y, Quan P, Liu X, Wang M, Fang L. Novel chemical permeation enhancers for transdermal drug delivery. ‎. Asian J Pharm Sci 2014; 9(2): 51-64.
[33]
Chen J, Jiang Q-D, Wu Y-M, et al. Potential of essential oils as penetration enhancers for transdermal administration of ibuprofen to treat dysmenorrhoea. Molecules 2015; 20(10): 18219-36.
[34]
Herman A, Herman AP. Essential oils and their constituents as skin penetration enhancer for transdermal drug delivery: a review. J Pharm Pharmacol 2015; 67(4): 473-85.
[35]
Gao XC, Tong Y. Effect of wintergreen oil on in vitro transdermal permeation of osthole and geniposide. Zhongguo Zhongyao Zazhi 2017; 42(7): 1338-43.
[36]
Zhu XF, Luo J, Guan YM, et al. Effects of Frankincense and Myrrh essential oil on transdermal absorption in vitro of Chuanxiong and penetration mechanism of skin blood flow. Zhongguo Zhongyao Zazhi 2017; 42(4): 680-5.
[37]
Anirudhan T, Nair AS, Gopika S. The role of biopolymer matrix films derived from carboxymethyl cellulose, sodium alginate and polyvinyl alcohol on the sustained transdermal release of diltiazem. Int J Biol Macromol 2018; 107: 779-89.
[38]
Siddaramaiah Kumar P, Divya K, Mhemavathi B, Manjula D. Chitosan/HPMC polymer blends for developing transdermal drug delivery systems. J Macromol Sci A 2006; 43(3): 601-7.
[39]
Wahid A, Sridhar BK, Shivakumar S. Preparation and evaluation of transdermal drug delivery system of etoricoxib using modified chitosan. Indian J Pharm Sci 2008; 70(4): 455-60.
[40]
Suksaeree J, Monton C, Madaka F, et al. Formulation, physicochemical characterization, and in vitro study of chitosan/HPMC blends-based herbal blended patches. AAPS PharmSciTech 2015; 16(1): 171-81.
[41]
Baviskar DT, Parik VB, Gupta HN, Maniyar AH, Jain DK. Design and evaluation of patches for transdermal delivery of losartan potassium. PDA J Pharm Sci Technol 2012; 66(2): 126-35.
[42]
Dragicevic N, Maibach HI. Percutaneous Penetration Enhancers Chemical Methods in Penetration Enhancement. Springer 2015.
[43]
Kulkarni RV, Wagh YJ. Crosslinked alginate films as rate controlling membranes for transdermal drug delivery application. J Macromol Sci A 2010; 47(7): 732-7.
[44]
Garala KC, Shah PH. Influence of crosslinking agent on the release of drug from the matrix transdermal patches of HPMC/Eudragit RL 100 polymer blends. J Macromol Sci A 2010; 47(3): 273-81.
[45]
Manickam B, Sreedharan R, Elumalai M. ‘Genipin’ - the natural water soluble cross-linking agent and its importance in the modified drug delivery systems: an overview. Curr Drug Deliv 2014; 11(1): 139-45.
[46]
Mayet N, Kumar P, Choonara YE, et al. Synthesis of a semi-interpenetrating polymer network as a bioactive curcumin film. AAPS PharmSciTech 2014; 15(6): 1476-89.
[47]
Abbasi A, Eslamian M, Heyd D, Rousseau D. Controlled release of DSBP from genipin-crosslinked gelatin thin films. Pharm Dev Technol 2008; 13(6): 549-57.
[48]
Wong RSH, Dodou K. Effect of drug loading method and drug physicochemical properties on the material and drug release properties of poly (ethylene oxide) hydrogels for transdermal delivery. Polymers (Basel) 2017; 9(7): 286.
[49]
Ita KB. Prodrugs for transdermal drug delivery - trends and challenges. J Drug Target 2016; 24(8): 671-8.
[50]
Jornada DH, dos Santos Fernandes GF, Chiba DE, de Melo TRF, dos Santos JL, Chung MC. The prodrug approach: a successful tool for improving drug solubility. Molecules 2015; 21(1): 42.
[51]
Sloan KB, Devarajan-Ketha H, Wasdo SC. Dermal and transdermal delivery: prodrugs. Ther Deliv 2011; 2(1): 83-105.
[52]
Lobo S, Li H, Farhan N, Yan G. Evaluation of diclofenac prodrugs for enhancing transdermal delivery. Drug Dev Ind Pharm 2014; 40(3): 425-32.
[53]
Liu K-S, Sung KC, Al-Suwayeh SA, et al. Enhancement of transdermal apomorphine delivery with a diester prodrug strategy. Eur J Pharm Biopharm 2011; 78(3): 422-31.
[54]
Gala U, Pham H, Chauhan H. Pharmaceutical applications of eutectic mixtures. J Dev Drugs 2013; 2: 1-2.
[55]
Tuntarawongsa S, Phaechamud T. Polymeric eutectic drug delivery system. J Met Mater Minerals 2012; 22: 2.
[56]
Yuan X, Capomacchia AC. Physicochemical studies of binary eutectic of ibuprofen and ketoprofen for enhanced transdermal drug delivery. Drug Dev Ind Pharm 2010; 36(10): 1168-76.
[57]
Yuan X, Capomacchia AC. Influence of physicochemical properties on the in vitro skin permeation of the enantiomers, racemate, and eutectics of ibuprofen for enhanced transdermal drug delivery. J Pharm Sci 2013; 102(6): 1957-69.
[58]
Liu C, Hu J, Sui H, Zhao Q, Zhang X, Wang W. Enhanced skin permeation of glabridin using eutectic mixture-based nanoemulsion. Drug Deliv Transl Res 2017; 7(2): 325-32.
[59]
Gajbhiye V, Rai K, Jadon R, Jain A, Nayak A. Elastic liposomes mediated transdermal delivery of aceclofenac. Pharmacist 2008; 3(1): 9.
[60]
Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett 2013; 8(1): 102.
[61]
Ashtikar M, Nagarsekar K, Fahr A. Transdermal delivery from liposomal formulations - evolution of the technology over the last three decades. J Control Release 2016; 242: 126-40.
[62]
Hussain A, Singh S, Sharma D, Webster TJ, Shafaat K, Faruk A. Elastic liposomes as novel carriers: recent advances in drug delivery. Int J Nanomedicine 2017; 12: 5087-108.
[63]
Grumezescu A, Oprea AE. Nanotechnology Applications in Food: Flavor, Stability, Nutrition and Safety. Academic Press 2017.
[64]
Rizwan M, Aqil M, Azeem A, Talegaonkar S, Sultana Y, Ali A. Enhanced transdermal delivery of carvedilol using nanoemulsion as a vehicle. J Exp Nanosci 2010; 5(5): 390-411.
[65]
Lococo D, Mora-Huertas CE, Fessi H, Zaanoun I, Elaissari A. Argan oil nanoemulsions as new hydrophobic drug-loaded delivery system for transdermal application. J Biomed Nanotechnol 2012; 8(5): 843-8.
[66]
Shakeel F, Baboota S, Ahuja A, Ali J, Aqil M, Shafiq S. Nanoemulsions as vehicles for transdermal delivery of aceclofenac. AAPS PharmSciTech 2007; 8(4)E104
[67]
Ledet G, Pamujula S, Walker V, Simon S, Graves R, Mandal TK. Development and in vitro evaluation of a nanoemulsion for transcutaneous delivery. Drug Dev Ind Pharm 2014; 40(3): 370-9.
[68]
Bennet D, Kim S. A transdermal delivery system to enhance quercetin nanoparticle permeability. J Biomater Sci Polym Ed 2013; 24(2): 185-209.
[69]
Donnelly RF, Singh TRR. Novel delivery systems for transdermal and intradermal drug delivery. John Wiley & Sons 2015.
[70]
Chevalier Y, Bolzinger M-A, Briançon S. Pickering Emulsions for Controlled Drug Delivery to the SkinPercutaneous Penetration Enhancers Chemical Methods in Penetration Enhancement. Springer 2015; pp. 267-81.
[71]
Marto J, Gouveia L, Goncalves L, et al. Starch pickering emulsion: a safe vehicle for topical drug delivery. Athens J Sci 2015; 2: 77-87.
[72]
Marku D, Wahlgren M, Rayner M, Sjöö M, Timgren A. Characterization of starch Pickering emulsions for potential applications in topical formulations. Int J Pharm 2012; 428(1-2): 1-7.
[73]
Gaur PK, Purohit S, Mishra S. Development of aceclofenac nanovesicular system using biomaterial for transdermal delivery: physical characterization, ex vivo, in vivo, and anti-inflammatory studies. J Biomater Sci Polym Ed 2013; 24(18): 2126-41.
[74]
Gaur PK, Mishra S, Aeri V. Formulation and evaluation of guggul lipid nanovesicles for transdermal delivery of aceclofenac. ScientificWorldJournal 2014; 2014534210
[75]
Kumar Gaur P, Mishra S, Purohit S. Nanovesicles of nitrendipine with lipid complex for transdermal delivery: pharmacokinetic and pharmacodynamic studies. Artif Cells Nanomed Biotechnol 2016; 44(7): 1684-93.
[76]
Choi MJ, Maibach HI. Liposomes and niosomes as topical drug delivery systems. Skin Pharmacol Physiol 2005; 18(5): 209-19.
[77]
Szura D, Ozimek Ł, Przybyło M, et al. The impact of liposomes on transdermal permeation of naproxen-in vitro studies. Acta Pol Pharm 2014; 71(1): 145-51.
[78]
Roslan N Z I, Ghani S M A, Yusof N B. Liposome as Transdermal carrier for Labisia Pumila and Ficus Deltoidea water extracts. J teknologi 2017; 71(1): 161-6.
[79]
Nounou MI, El-Khordagui LK, Khalafallah NA, Khalil SA. Liposomal formulation for dermal and transdermal drug delivery: past, present and future. Recent Pat Drug Deliv Formul 2008; 2(1): 9-18.
[80]
Moghimipour E, Salami A, Monjezi M. Formulation and evaluation of liposomes for transdermal delivery of celecoxib. Jundishapur J Nat Pharm Prod 2015; 10(1)e17653
[81]
Yamazaki N, Yamakawa S, Sugimoto T, et al. Carboxylated phytosterol derivative-introduced liposomes for skin environment-responsive transdermal drug delivery system. J Liposome Res 2017; 1-10.
[82]
Benson HA. Elastic liposomes for topical and transdermal drug delivery. Curr Drug Deliv 2009; 6(3): 217-26.
[83]
Chen J, Lu WL, Gu W, Lu SS, Chen ZP, Cai BC. Skin permeation behavior of elastic liposomes: role of formulation ingredients. Expert Opin Drug Deliv 2013; 10(6): 845-56.
[84]
Benson HA. Elastic liposomes for topical and transdermal drug delivery. Methods Mol Biol 2017; 107-17.
[85]
Gupta A, Aggarwal G, Singla S, Arora R. Transfersomes: a novel vesicular carrier for enhanced transdermal delivery of sertraline: development, characterization, and performance evaluation. Sci Pharm 2012; 80(4): 1061-80.
[86]
Malakar J, Sen SO, Nayak AK, Sen KK. Formulation, optimization and evaluation of transferosomal gel for transdermal insulin delivery. Saudi Pharm J 2012; 20(4): 355-63.
[87]
Ahad A, Al-Saleh AA, Al-Mohizea AM, et al. Formulation and characterization of novel soft nanovesicles for enhanced transdermal delivery of eprosartan mesylate. Saudi Pharm J 2017; 25(7): 1040-6.
[88]
Ascenso A, Raposo S, Batista C, et al. Development, characterization, and skin delivery studies of related ultradeformable vesicles: transfersomes, ethosomes, and transethosomes. Int J Nanomedicine 2015; 10: 5837-51.
[89]
Ramadon D, Goldie AW, Anwar E. Novel transdermal ethosomal gel containing green tea (Camellia sinensis L. Kuntze) Leaves Extract: Formulation and In vitro Penetration Study. J Young Pharm 2017; 9(3): 336.
[90]
Verma P, Pathak K. Therapeutic and cosmeceutical potential of ethosomes: An overview. J Adv Pharm Technol Res 2010; 1(3): 274-82.
[91]
Bhosale SS, Avachat AM. Design and development of ethosomal transdermal drug delivery system of valsartan with preclinical assessment in Wistar albino rats. J Liposome Res 2013; 23(2): 119-25.
[92]
Song CK, Balakrishnan P, Shim CK, Chung SJ, Chong S, Kim D-D. A novel vesicular carrier, transethosome, for enhanced skin delivery of voriconazole: characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces 2012; 92: 299-304.
[93]
Garg V, Singh H, Bhatia A, et al. Systematic development of transethosomal gel system of piroxicam: formulation optimization, in vitro evaluation, and ex vivo assessment. AAPS PharmSciTech 2017; 18(1): 58-71.
[94]
Chen ZX, Li B, Liu T, et al. Evaluation of paeonol-loaded transethosomes as transdermal delivery carriers. Eur J Pharm Sci 2017; 99: 240-5.
[95]
Garg G, Saraf S, Saraf S. Cubosomes: an overview. Biol Pharm Bull 2007; 30(2): 350-3.
[96]
Han S, Shen JQ, Gan Y, et al. Novel vehicle based on cubosomes for ophthalmic delivery of flurbiprofen with low irritancy and high bioavailability. Acta Pharmacol Sin 2010; 31(8): 990-8.
[97]
Karami Z, Hamidi M. Cubosomes: remarkable drug delivery potential. Drug Discov Today 2016; 21(5): 789-801.
[98]
Peng X, Zhou Y, Han K, et al. Characterization of cubosomes as a targeted and sustained transdermal delivery system for capsaicin. Drug Des Devel Ther 2015; 9: 4209-18.
[99]
Kwon TK, Kim JC. Preparation and in vitro skin permeation of cubosomes containing hinokitiol. J Dispers Sci Technol 2010; 31(7): 1004-9.
[100]
Kwon TK, Lee HY, Dai Kim J, Shin WC, Park SK, Kim JC. In vitro skin permeation of cubosomes containing water soluble extracts of Korean barberry. Colloid J 2010; 72(2): 205-10.
[101]
Salah S, Mahmoud AA, Kamel AO. Etodolac transdermal cubosomes for the treatment of rheumatoid arthritis: ex vivo permeation and in vivo pharmacokinetic studies. Drug Deliv 2017; 24(1): 846-56.
[102]
Dudala TB, Yalavarthi PR, Vadlamudi HC, et al. A perspective overview on lipospheres as lipid carrier systems. Int J Pharm Investig 2014; 4(4): 149-55.
[103]
Nasr M, Mansour S, Mortada ND, El Shamy AA. Lipospheres as carriers for topical delivery of aceclofenac: preparation, characterization and in vivo evaluation. AAPS PharmSciTech 2008; 9(1): 154-62.
[104]
Jain S, Patel N, Shah MK, Khatri P, Vora N. Recent advances in lipid-based vesicles and particulate carriers for topical and transdermal application. J Pharm Sci 2017; 106(2): 423-45.
[105]
Jain A, Pooladanda V, Bulbake U, et al. Liposphere mediated topical delivery of thymoquinone in the treatment of psoriasis. Nanomedicine (Lond) 2017; 13(7): 2251-62.
[106]
Desai P, Patlolla RR, Singh M. Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery. Mol Membr Biol 2010; 27(7): 247-59.
[107]
Mukherjee S, Ray S, Thakur RS. Solid lipid nanoparticles: a modern formulation approach in drug delivery system. Indian J Pharm Sci 2009; 71(4): 349-58.
[108]
Naseri N, Valizadeh H, Zakeri-Milani P. 108.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.
[109]
Elnaggar YS, El-Massik MA, Abdallah OY. Fabrication, appraisal, and transdermal permeation of sildenafil citrate-loaded nanostructured lipid carriers versus solid lipid nanoparticles. Int J Nanomedicine 2011; 6: 3195-205.
[110]
Abbasi E, Aval SF, Akbarzadeh A, et al. Dendrimers: synthesis, applications, and properties. Nanoscale Res Lett 2014; 9(1): 247.
[111]
Madaan K, Kumar S, Poonia N, Lather V, Pandita D. Dendrimers in drug delivery and targeting: Drug-dendrimer interactions and toxicity issues. J Pharm Bioallied Sci 2014; 6(3): 139-50.
[112]
Yang Y, Sunoqrot S, Stowell C, et al. Effect of size, surface charge, and hydrophobicity of poly(amidoamine) dendrimers on their skin penetration. Biomacromolecules 2012; 13(7): 2154-62.
[113]
Winnicka K, Wroblewska M, Sosnowska K, Car H, Kasacka I. Evaluation of cationic polyamidoamine dendrimers’ dermal toxicity in the rat skin model. Drug Des Devel Ther 2015; 9: 1367-77.
[114]
Duan X-D, Ji C-J, Nie L. Formulation and development of dendrimer-based transdermal patches of meloxicam for the management of arthritis. Trop J Pharm Res 2015; 14(4): 583-90.
[115]
Bahadoran A, Moeini H, Bejo MH, Hussein MZ, Omar AR. Development of tat-conjugated dendrimer for transdermal DNA vaccine delivery. J Pharm Pharm Sci 2016; 19(3): 325-38.
[116]
Uchechi O, Ogbonna JD, Attama AA. Nanoparticles for dermal and transdermal drug delivery Application of Nanotechnology in Drug Delivery. InTech 2014.
[117]
Martins M, Azoia NG, Melle‐Franco M, Ribeiro A, Cavaco‐Paulo A. Permeation of skin with (C60) fullerene dispersions. Eng Life Sci 2017.
[118]
Degim IT, Burgess DJ, Papadimitrakopoulos F. Carbon nanotubes for transdermal drug delivery. J Microencapsul 2010; 27(8): 669-81.
[119]
Gupta R, Rai B. Molecular dynamics simulation study of translocation of fullerene C60 through skin bilayer: effect of concentration on barrier properties. Nanoscale 2017; 9(12): 4114-27.
[120]
Chu M, Wu Q, Wang J, et al. In vitro and in vivo transdermal delivery capacity of quantum dots through mouse skin. Nanotechnology 2007; 18(45)455103
[121]
Matea CT, Mocan T, Tabaran F, et al. Quantum dots in imaging, drug delivery and sensor applications. Int J Nanomedicine 2017; 12: 5421-31.
[122]
Franke D, Harris DK, Chen O, et al. Continuous injection synthesis of indium arsenide quantum dots emissive in the short-wavelength infrared. Nat Commun 2016; 7: 12749.
[123]
Surana K, Singh PK, Rhee H-W, Bhattacharya B. Synthesis, characterization and application of CdSe quantum dots. J Ind Eng Chem 2014; 20(6): 4188-93.
[124]
Degim IT, Kadioglu D. Cheap, suitable, predictable and manageable nanoparticles for drug delivery: quantum dots. Curr Drug Deliv 2013; 10(1): 32-8.
[125]
Liang K, Xu K, Bessarab D, Obaje J, Xu C. Arbutin encapsulated micelles improved transdermal delivery and suppression of cellular melanin production. BMC Res Notes 2016; 9(1): 254.
[126]
Tong Y-C, Yu T-Y, Chang S-F, Liaw J. Nanopolymeric micelle effect on the transdermal permeability, the bioavailability and gene expression of plasmid. Mol Pharm 2012; 9(1): 111-20.
[127]
Lee RW, Shenoy DB, Sheel R. Micellar nanoparticles: applications for topical and passive transdermal drug delivery Handbook of Non-Invasive Drug Delivery Systems. Burlington, MA: Elsevier Inc 2010; pp. 37-58.
[128]
Fernandes RFM. Penetration of gold nanoparticles through the skin. University of Southampton 2014.
[129]
Khayrullin RM, Terentyuk GS, Savenkova MV, Genina EA. The technique of enhancing the transdermal penetration for the gold nanoparticles and perspectives of application. J Cancer Ther 2013; 4(06): 48.
[130]
Hsiao PF, Peng S, Tang T-C, Lin S-Y, Tsai H-C. Enhancing the in vivo transdermal delivery of gold nanoparticles using poly(ethylene glycol) and its oleylamine conjugate. Int J Nanomedicine 2016; 11: 1867-78.
[131]
Ibrahim M M, Hafez S A, Mahdy M M. Organogels, hydrogels and bigels as transdermal delivery systems for diltiazem hydrochloride. asian journal of pharmaceutical sciences 2013; 8(1): 48-57.
[132]
Allen L, Ansel HC. Ansel’s pharmaceutical dosage forms and drug delivery systems. Lippincott Williams & Wilkins 2013.
[133]
Sahoo S, Kumar N, Bhattacharya C, et al. Organogels: properties and applications in drug delivery. Des Monomers Polym 2011; 14(2): 95-108.
[134]
Agrawal V, Gupta V, Ramteke S, Trivedi P. Preparation and evaluation of tubular micelles of pluronic lecithin organogel for transdermal delivery of sumatriptan. AAPS PharmSciTech 2010; 11(4): 1718-25.
[135]
Ibrahim MM, Nair AB, Aldhubiab BE, Shehata TM. Hydrogels and Their Combination with Liposomes, Niosomes, or Transfersomes for Dermal and Transdermal Drug Delivery In:. Liposomes. InTech 2017.
[136]
Rehman K, Zulfakar MH. Recent advances in gel technologies for topical and transdermal drug delivery. Drug Dev Ind Pharm 2014; 40(4): 433-40.
[137]
Boddé HE, Van Aalten EA, Junginger HE. Hydrogel patches for transdermal drug delivery; in vivo water exchange and skin compatibility. J Pharm Pharmacol 1989; 41(3): 152-5.
[138]
Wang W, Wat E, Hui PC, et al. Dual-functional transdermal drug delivery system with controllable drug loading based on thermosensitive poloxamer hydrogel for atopic dermatitis treatment. Sci Rep 2016; 6: 24112.
[139]
Ritika A, Geeta A, Harikumar SL, Kirandeep K. Nanoemulsion based hydrogel for enhanced transdermal delivery of Ketoprofen. Adv Pharmaceutics 2014; p. 468456.
[140]
Moreira TSA, de Sousa VP, Pierre MBR. A novel transdermal delivery system for the anti-inflammatory lumiracoxib: influence of oleic acid on in vitro percutaneous absorption and in vivo potential cutaneous irritation. AAPS PharmSciTech 2010; 11(2): 621-9.
[141]
Karande P, Mitragotri S. Enhancement of transdermal drug delivery via synergistic action of chemicals. Biochim Biophys Acta 2009; 1788(11): 2362-73.
[142]
Walters KA, Hadgraft J. Pharmaceutical skin penetration enhancement. Informa Health Care 1993; Vol. 59.
[143]
Marwah H, Garg T, Goyal AK, Rath G. Permeation enhancer strategies in transdermal drug delivery. Drug Deliv 2016; 23(2): 564-78.
[144]
Rai V, Raghavan L. Transdermal Drug Delivery Systems Using SupersaturationPercutaneous Penetration Enhancers Chemical Methods in Penetration Enhancement. Berlin, Heidelberg: Springer 2015; pp. 151-61.
[145]
Pellett M, Raghavan SL, Hadgraft J, Davis A. The application of supersaturated systems to percutaneous drug delivery. Transdermal Drug Deliv 2002; 123: 305-26.
[146]
Bharkatiya M, Nema R. Skin penetration enhancement techniques. J Young Pharm 2009; 1(2): 110.
[147]
Dias MM, Raghavan SL, Pellett MA, Hadgraft J. The effect of β-cyclodextrins on the permeation of diclofenac from supersaturated solutions. Int J Pharm 2003; 263(1-2): 173-81.
[148]
Iervolino M, Cappello B, Raghavan SL, Hadgraft J. Penetration enhancement of ibuprofen from supersaturated solutions through human skin. Int J Pharm 2001; 212(1): 131-41.
[149]
Stojančević M, Pavlović N, Goločorbin-Kon S, Mikov M. Application of bile acids in drug formulation and delivery. Front Life Sci 2013; 7(3-4): 112-22.
[150]
Cheng CY, Oh H, Wang TY, Raghavan SR, Tung SH. Mixtures of lecithin and bile salt can form highly viscous wormlike micellar solutions in water. Langmuir 2014; 30(34): 10221-30.
[151]
Moghimipour E, Tabassi SS, Kouchak M, Varghaei H. Combination strategies for enhancing transdermal absorption of theophylline through shed snake skin. Asian J Pharm Clin Res 2012; 5(Suppl.): 30-4.
[152]
Moghimipour E, Ameri A, Handali S. Absorption-enhancing effects of bile salts. Molecules 2015; 20(8): 14451-73.
[153]
Fetih G, Ibrahim MA, Amin MA. Design and characterization of transdermal films containing ketorolac tromethamine. Int J Pharm Tech Res 2011; 3(1): 449-58.
[154]
Cannon JB. Lipids in transdermal and topical drug delivery. Am Pharm Rev 2014; 17: 7.

© 2024 Bentham Science Publishers | Privacy Policy