Journal of Pharmaceutical and Biomedical Sciences

Transdermal Pharmacotherapy: Traditions and Innovations

Yurieva Eleonora A, Kushnareva Maria V*, Vozdvizhenskaya Ekaterina S

Abstract


The review is focused on the development questions and transdermal medicines usage. The advantages and limitations for transdermal drug administration are presented. The analysis of 70 scientific sources is performed. The morphological and functional structure of the skin as a factor of transdermal drug delivery into the body is described. The review provides information on transdermal therapeutic systems (advantages, disadvantages, structure, technology, scope). The possibility of using transdermal therapeutic systems for the vaccination and local anesthesia, treatment of arthritis, Alzheimer's disease,
contact dermatitis, burn wounds, infectious diseases, osteoporosis, oncology and other pathologies is discussed.

Keywords


transdermal therapeutic systems, pharmacotherapy, medicines

Full Text:

References


Caruthers SD, Wickline SA, Lanza GM. Nanotechnological applications in medicine. Curr Opin Biotechnol. 2007;18:26–30.

Dutta RC. Drug carriers in pharmaceutical design: promises and progress. Curr Pharm Des. 2007;13:761–769.

Kharkevitch DA. Pharmacology. Textbook for medical students. M.: GEOTAR-Media. 2008:672. http://www.rosmedlib.ru/book/ISBN5970402648.html

Li J, Zeng M, Shan H, Tong C. Microneedle patches as drug and vaccine delivery platform. Curr Med Chem. 2017;24:2413–2422.

Allen Jr, Nicholas G. Popovich, Howard CA. Pharmaceutical dosage forms and drug delivery systems, 8th Edition., Wolter Kluwer Publishers, New Delhi. 2005;298–299.

Gannu R, Vamshi YV, Kishan V, Rao YM. Development of nitrendipine transdermal patches: in vitro and ex vivo characterization.Curr Drug Deliv. 2007;4:69–76.

Kaza R, Pitchaimani R. Formulation of transdermal drug delivery system: matrix type and selection of polymer- their evaluation. Curr Drug Discov Technol. 2006;3:279–285.

Ning YM, Rao YF, Liang WQ. Influence of permeation enhancers on transdermal permeation of anemonin. Zhonqquo Zhong Yao Za Zhi. 2007;32:393–396.

Amsden BG, Goosen MF. Transdermal delivery of peptide and protein drugs: an overview. AIChE J. 2004;41:1972–1997.

Boix A, Peraire C, Obach R, Domenech J. Estimation of transdermal permeation parameters non-stationary diffusion experiments. Application to pre-treatment studies with terpenes. Pharm Res. 2005;22:94–102.

Foco A, Hadziabdic J, Becic F. Transdermal drug delivery systems. Med Arh. 2004;58:230–234.

Knutson K, Krill SL, Zhang J. Solvent-mediated alteration of the stratum corneum. J Control Rel. 1990;11:93–103.

Hatanaka T, Shimoyama M, Sugibayashi K, Morimoto Y. Effect of vehicle on the skin permeability of drug. J Control Rel.1993;23:247–260.

Megrab NA, Williams AC, Barn BW. Oestradiol permeation across human skin, silastic and snake skin membranes: the effects of ethanol/water co-solvent systems. Int J Pharm. 1995;116:1:101–112.

Jain AK, Panchagnula R. Transdermal delivery of imipramine hydrochloride: development and evaluation (in vitro and in vivo) of reservoir gel formulation. Biopharm Drag Dispos.2005;26:41–49.

Amirova I. To the problem of pre-clinical diagnostics of hyperproliferative diseases among oil workers. South Caucasian Dermatology and Venerology News January – June 2007;1:32–http://medportal.ge/eml/dermatology/2007_1n.pdf.

Fichtner CG, Braun BG. Hyperphagia and weight loss during fluoxetine treatment. Ann Pharmacother. 1994;28:1350–1352.

The Complete Drug Reference: The Extra Pharmacopoeia, 31st Revised Edition . William Martindale (Author): James. E. F. Reynolds (Editor). Pharmaceutical Press, Royal Pharmaceutical Society of Great Britain, London; 1996. pp. 2363. ISBN-100-85369-342-0, ISBN -13 978- 0-85369-342-0

Kusum Devi V, Saisivam S, Maria GR, Deepti PU. Design and evaluation of matrix diffusion controlled transdermal patches of verapamil hydrochloride. Drug Dev Ind Pharm. 2003;5:495– 503.

Siafaka I, Rellia P, Argyra E, Iakovidou N. Pharmacokinetic profile and efficacy of a fentanyl transdermal delivery system for acute postoperative pain after intra-abdominal gynecologic surgery for cancer. Pain Pract. 2004;4:98–102.

Yokota K, Johyama Y, Matsumoto N, Yamaguchi K. Occupational dermatitis from a one-component naphthalene type epoxy adhesive. Ind Health. 2002;40:63–65.

Leahy LG. From immediate release to long acting drug delivery systems: what do they mean and why do they matter? J Psychosoc Nurs Ment Health Serv. 2017;55:19–23.

Sharma PK, Panda A, Pradhan A, Zhang J, Thakkar R, Whang CH, et al. Solid-state stability issues of drugs in transdermal patch formulations. AAPS Pharm Sci Tech. 2017;19:27–35.

Banga Ajay K. Transdermal devices and patches—enhancement techniques and disposal of residual drug. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Peterson T, Burton S, Englund B, Grosh S. In vitro permeability of poly (ethylene vinyl acetate) and microporous polyethylene membranes. Proc Int Symp Control Res Bioactive Mater. 1990;17:411–412.

Ghosh TK, Banga A?. Methods of enhancement of transdermal drug delivery: Part II A. Chemical permeation enhancers. Pharm Technol. 1993;17:68–90.

Rajadhyaksha VJ, Sharma K, Pfister WR. Oxazolidinones: A new class of permeation enhancer. Ghosh TK, Pfister WR, Yum S ed., Transdermal and Topical Drug Delivery Systems, Interpharm Press: Illinois, Buffalo Crave. 1997;477–509.

Dreher F, Walde P, Walther P, Wehrli E. Interaction of a lecithin microemulsion gel with human stratum corneum and its effect on transdermal transport. J Cont Rel. 1997;45:131–140.

Ogiso T, Iwaki M, Paku T. Effect of various enhancers on transdermal penetration on indometacin and urea and relationship between penetration parameters and enhancement factors. J Pharm Sci. 1995;84:482–488.

Gallarate M, Gasco MR, Trotta M, Chetoni P, Saettone MF. Preparation and evaluation in vitro of solution and o/w microemulsions containing levobunolol as ion-pair. J Pharm.1993;100:219– 225.

Hadgraft J, Peck J, Williams DG, Pugh W J, Allan G. Mechanisms of action of skin penetration enhancers/retarders: azone and analogs. Int J Pharm. 1996;141:17–25.

Takahshi Y, Furuya K, Iwata M, Onishi H. Trial for transdermal administration of sulfonylureas. Yakugaku Zasshi. 1997;117:1022–1027.

Mutalik S, Udupa N. Glibenclamide transdermal patches: physico-chemical, pharmacodynamic, and pharmacokinetic evaluations. J Pharm Sci. 2004;93:1577–1594.

Zhao H, Liu C, Quan P, Wan X, Shen M, Fang L. Mechanism study on ion-pair complexes controlling skin permeability: effect of ion-pair dissociation in the viable epidermis on transdermal permeation of bisoprolol. Int J Pharm. 2017;532:29–36.

Parikh DK, Ghosh TK. Feasibility of transdermal delivery of fluoxetine. AAPS Pharm Sci Tech. 2005;6:E144–E149.

Garland MJ, Caffarel-Salvador E, Migalska K, Woolfson AD, Donnelly RF. Dissolving polymeric microneedle arrays for electrically assisted transdermal drug delivery. J Con Rel. 2012;159:52–59.

Raghavan Lakshmi. Commercialization challenges and strategies for active delivery systems. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Alur HH, Beal JD, Pather SI, Mitra AK, Johnston TP. Evaluation of novel, natural oligosaccharide dum as a sustained-release and mucoadhesive component of calcitonin buccal tablets. J Pharm Sci. 2000;88:1313–1319.

Zobrist RH, Quan D, Thomas HM, Stanworth S, Sanders SW. Pharmacokinetics and metabolism of transdermal oxibutynin: in vitro and in vivo performance of a novel delivery system. Pharm Res. 2003;20:103–109.

Lin SY. Chronotherapeutic approach to design a thermoresponsive membrane for transdermal drug delivery. Curr Drug Deliv.2004;1:249–263.

Arora P, Mukherjee B. Design, development, physicochemical, and in vitro and in vivo evaluation of transdermal patches containing diclofenac diethylammonium salt. J Pharm Sci.2002;91:2076–2089.

Gore AV, Liang AC, Chien YW. Comparative biomembrane permeation of tacrine using Yucatan minipigs and domestic pigs as the animal model. J Pharm Sci. 1998;87:441–447.

Mutalik S, Udupa N. Formulation development, in vitro and in vivo evaluation of membrane controlled transdermal systems of glibenclamide. J Pharm Sci. 2005;21:26–38.

Hammell DC, Zhang LP, Ma F, Abshire SM, McIlwrath SL, Stinchcomb AL, et al. Transdermal cannabidiol reduces inflammation and pain-related behaviours in a rat model of arthritis. Eur J Pain. 2016;6:936–948.

Stinchcomb L. Microneedle enhanced transdermal delivery:academic bench to clinical trials audra. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Kulkarni RV, Mutalik S, Hiremath D. Effect of plasticizers on permeability and properties of Eudragit films for transdermal application.Indian J Pharm Sci. 2002;64:28–31.

Bohlke Allan. Development and scale-up of microneedle based drug delivery systems. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Murthy Narasimha. Dermal absorption during short-term exposure. Transdermal and intradermal drug delivery systems.Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA.2015;11–12; http://www.pharmaedresources.com/brochures/ Transdermal2015.pdf

Yamazaki N, Yamakawa S, Sugimoto T, Yoshizaki Y, Teranishi R, Hayashi T, et al. Carboxylated phytosterol derivative-introduced liposomes for skin environment-responsive transdermal drug delivery system. J Liposome Res. 2017;1–10.

Singh Bobby. Expanding the transdermal universe with next generation technologies. Transdermal and intradermal drug delivery systems. Advanced Design, Development, and Delivery of Skin-Mediated Therapies and Vaccines Racquet Club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.

com/brochures/Transdermal2015.pdf

Ciotti Susan. Mechanism of a novel antimicrobial Na-noemulsion for the treatment of burn wounds. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Moffatt K, Wang Y, Raj Singh TR, Donnelly RF. Microneedles for enhanced transdermal and intraocular drug delivery. Curr Opin Pharmacol. 2017;36:14–21.

Hattersley Gary. A phase 2 clinical study of transdermal patch delivering the PTHrP analog, abaloparatide, for treatment of postmenopausal women with osteoporosis. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Dede AD, Makras P, Anastasilakis AD. Investigational anabolic agents for the treatment of osteoporosis: an update on recent developments. Expert Opin Investig Drugs.2017;26:1137–1144.

Gill Harvinder. Microneedles for allergen immunotherapy and cancer treatment. Transdermal and intradermal drug delivery systems. Advanced design, development, and delivery of skin-mediated therapies and vaccines. Racquet club of Philadelphia, PA. 2015;11–12; http://www.pharmaedresources.com/brochures/Transdermal2015.pdf

Yu J, Wan K, Sun X. Improved transdermal delivery of morin efficiently inhibits allergic contact dermatitis. Int J Pharm.2017;530:145–154.

Chen C, You P. A novel local anesthetic system: transcriptional transactivator peptide-decorated nanocarriers for skin delivery of ropivacaine. Drug Des Devel Ther. 2017;11:1941–1949.

Yousefshahi F, Predescu O, Francisco Asenjo J. The efficacy of systemic lidocaine in the management of chronic pain: a literature review. Anesth Pain Med. 2017;3:e44732

Sood J, Sapra B, Tiwary AK. Microemulsion transdermal formulation for simultaneous delivery of valsartan and nifedipine: formulation by design. AAPS Pharm Sci Tech. 2017;6:1901–1916.

Kwon KM, Lim SM, Choi S, Kim DH, Jin HE, Jee G, et al. Microneedles: quick and easy delivery methods of vaccines. Clin Exp Vaccine Res.2017;2:156–159.

Rouphael NG, Paine M, Mosley R, Henry S, McAllister DV, Kalluri H, et al. TIV-MNP 2015 Study Group. The safety, immunogenicity, and acceptability of inactivated influenza vaccine delivered by microneedle patch (TIV-MNP 2015): a randomised, partly blinded, placebo-controlled, phase 1 trial. Lancet. 2017;390:649–658.

Li J, Zeng M, Shan H, Tong C. Microneedle patches as drug and vaccine delivery platform. Curr Med Chem. 2017;24:2413–2422.

Wildner P, Selmaj KW. Multiple sclerosis: skin-induced antigen-specific immune tolerance. J Neuroimmunol. 2017;311:49–58.

Wu P, Liang Q, Feng P, Li C, Yang C, Liang H. A novel brucine gel transdermal delivery system designed for anti-inflammatory and analgesic activities. Int J Mol Sci. 2017;18: pii E757.

Bhidayasiri R, Sringean J, Chaiwong S, Anan C, Penkeaw N, Leaknok A, et al. Rotigotine for nocturnal hypokinesia in Parkinson’s disease: quantitative analysis of efficacy from a randomized, placebo-

controlled trial using an axial inertial sensor.R Parkinsonism Relat Disord. 2017; https://www.ncbi.nlm.nih.gov/labs/articles/28818560/

Nguyen TT, Giau VV, Vo TK. Current advances in transdermal delivery of drugs for Alzheimer’s disease. Indian J Pharmacol.2017;49:145–154.

Yoon SJ, Choi SH, Na HR, Park KW, Kim EJ, Han HJ, et al. Effects on agitation with rivastigmine patch monotherapy and combination therapy with memantine in mild to moderate Alzheimer’s disease: a multicenter 24-week prospective randomized open-label study (the Korean EXelon Patch and combination with mEmantine

Comparative Trial study). Geriatr Gerontol Int. 2017;3:494–499.

Wais M, Aqil M, Sharma AK, Agnihotri J. Nanoemulsion-based transdermal drug delivery system for the treatment of tuberculosis. Recent Pat Antiinfect Drug Discov. 2017.

Rastogi V, Yadav P, Verma A, Pandit JK. Ex vivo and in vivo evaluation of microemulsion based transdermal delivery of E. coli specific T4 bacteriophage: a rationale approach to treat bacterial infection. Eur J Pharm Sci. 2017;107:168–182.

European Pharmacopoeia (Ph.Eur.) 9th edn, Pharmeuropa; useful information: June 2017. http://www.edqm.eu/node/15048.


Refbacks

  • There are currently no refbacks.


Copyright (c) 2018 Journal of Pharmaceutical and Biomedical Sciences

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.