93
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Key Transdermal Patch Using Cannabidiol-Loaded Nanocarriers with Better Pharmacokinetics in vivo

ORCID Icon, , , & ORCID Icon
Pages 4321-4337 | Received 14 Dec 2023, Accepted 29 Apr 2024, Published online: 16 May 2024

References

  • Yu L, Madsen FB, Eriksen SH, Andersen AJC, Skov AL. A reliable quantitative method for determining CBD content and release from transdermal patches in Franz cells. Phytochem Anal. 2022;33(8):1257–1265. doi:10.1002/pca.3188
  • Gaston TE, Friedman D. Pharmacology of cannabinoids in the treatment of epilepsy. Epilepsy Behav. 2017;70:313–318. doi:10.1016/j.yebeh.2016.11.016
  • Huntsman RJ, Tang-Wai R, Shackelford AE. Cannabis for pediatric epilepsy. J Clin Neurophysiol. 2020;37(1):2–8. doi:10.1097/wnp.0000000000000641
  • Watt G, Karl T. In vivo evidence for therapeutic properties of cannabidiol (CBD) for Alzheimer’s disease. Front Pharmacol. 2017;8:20. doi:10.3389/fphar.2017.00020
  • Crippa JA, Guimarães FS, Campos AC, Zuardi AW. Translational investigation of the therapeutic potential of cannabidiol (CBD): toward a new age. Front Immunol. 2018;9:2009. doi:10.3389/fimmu.2018.02009
  • Zuardi AW, Crippa JA, Hallak JE, et al. Cannabidiol for the treatment of psychosis in Parkinson’s disease. J Psychopharmacol. 2009;23(8):979–983. doi:10.1177/0269881108096519
  • Shannon S, Lewis N, Lee H, Hughes S. Cannabidiol in anxiety and sleep: a large case series. Perm J. 2019;23:18–041. doi:10.7812/tpp/18-041
  • Chagas MH, Eckeli AL, Zuardi AW, et al. Cannabidiol can improve complex sleep-related behaviours associated with rapid eye movement sleep behaviour disorder in Parkinson’s disease patients: a case series. J Clin Pharm Ther. 2014;39(5):564–566. doi:10.1111/jcpt.12179
  • García-Gutiérrez MS, Navarrete F, Gasparyan A, Austrich-Olivares A, Sala F, Manzanares J. Cannabidiol: a potential new alternative for the treatment of anxiety, depression, and psychotic disorders. Biomolecules. 2020;10(11):1575. doi:10.3390/biom10111575
  • McGuire P, Robson P, Cubala WJ, et al. Cannabidiol (CBD) as an adjunctive therapy in Schizophrenia: a multicenter randomized controlled trial. Am J Psychiatry. 2018;175(3):225–231. doi:10.1176/appi.ajp.2017.17030325
  • Millar SA, Maguire RF, Yates AS, O’Sullivan SE. Towards better delivery of cannabidiol (CBD). Pharmaceuticals. 2020;13(9):219. doi:10.3390/ph13090219
  • Hossain KR, Alghalayini A, Valenzuela SM. Current challenges and opportunities for improved cannabidiol solubility. Int J Mol Sci. 2023;24(19):14514. doi:10.3390/ijms241914514
  • Grifoni L, Vanti G, Donato R, Sacco C, Bilia AR. Promising nanocarriers to enhance solubility and bioavailability of cannabidiol for a plethora of therapeutic opportunities. Molecules. 2022;27(18):6070. doi:10.3390/molecules27186070
  • Millar SA, Stone NL, Yates AS, O’Sullivan SE. A systematic review on the pharmacokinetics of cannabidiol in humans. Front Pharmacol. 2018;9:1365. doi:10.3389/fphar.2018.01365
  • Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers. 2007;4(8):1770–1804. doi:10.1002/cbdv.200790152
  • Kumeria T, Wang J, Kim B, et al. Enteric polymer-coated porous silicon nanoparticles for site-specific oral delivery of IgA antibody. ACS Biomater Sci Eng. 2022;8(10):4140–4152. doi:10.1021/acsbiomaterials.0c01313
  • Zhang Y, Wang Y, Li X, Nie D, Liu C, Gan Y. Ligand-modified nanocarriers for oral drug delivery: challenges, rational design, and applications. J Control Release. 2022;352:813–832. doi:10.1016/j.jconrel.2022.11.010
  • Ouyang J, Zhang Z, Deng B, et al. Oral drug delivery platforms for biomedical applications. Mater. 2023;62:296–326. doi:10.1016/j.mattod.2023.01.002
  • Durán-Lobato M, Niu Z, Alonso MJ. Oral delivery of biologics for precision medicine. Adv Mater. 2020;32(13):1901935. doi:10.1002/adma.201901935
  • Asad S, Jacobsen A-C, Teleki A. Inorganic nanoparticles for oral drug delivery: opportunities, barriers, and future perspectives. Curr Opin Chem Eng. 2022;38:100869. doi:10.1016/j.coche.2022.100869
  • Hyung Kang R, Hee Kim N, Kim D. A transformable and biocompatible polymer series using ring-opening polymerization of cyclic silane for more effective transdermal drug delivery. Chem Eng J. 2022;440:135989. doi:10.1016/j.cej.2022.135989
  • Li B, Lu G, Liu W, Liao L, Ban J, Lu Z. Formulation and evaluation of PLGA nanoparticulate-based microneedle system for potential treatment of neurological diseases. Int J Nanomed. 2023;18:3745–3760. doi:10.2147/ijn.S415728
  • Tanner EEL, Curreri AM, Balkaran JPR, et al. Design principles of Ionic liquids for transdermal drug delivery. Adv Mater. 2019;31(27):1901103. doi:10.1002/adma.201901103
  • Hu H, Ruan H, Ruan S, et al. Acid-responsive PEGylated branching PLGA nanoparticles integrated into dissolving microneedles enhance local treatment of arthritis. Chem Eng J. 2022;431:134196. doi:10.1016/j.cej.2021.134196
  • Singh V, Kesharwani P. Recent advances in microneedles-based drug delivery device in the diagnosis and treatment of cancer. J Control Release. 2021;338:394–409. doi:10.1016/j.jconrel.2021.08.054
  • Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008;26(11):1261–1268. doi:10.1038/nbt.1504
  • Chen Y, Wang M, Fang L. Biomaterials as novel penetration enhancers for transdermal and dermal drug delivery systems. Drug Delivery. 2013;20(5):199–209. doi:10.3109/10717544.2013.801533
  • Li H, Peng Z, Song Y, et al. Study of the permeation-promoting effect and mechanism of solid microneedles on different properties of drugs. Drug Delivery. 2023;30(1):2165737. doi:10.1080/10717544.2023.2165737
  • Zhao W, Zheng L, Yang J, Ma Z, Tao X, Wang Q. Dissolving microneedle patch-assisted transdermal delivery of methotrexate improve the therapeutic efficacy of rheumatoid arthritis. Drug Delivery. 2023;30(1):121–132. doi:10.1080/10717544.2022.2157518
  • Jamaledin R, Yiu CKY, Zare EN, et al. Advances in antimicrobial microneedle patches for combating infections. Adv Mater. 2020;32(33):2002129. doi:10.1002/adma.202002129
  • Adin SN, Gupta I, Rashid MA, Alhamhoom Y, Aqil M, Mujeeb M. Nanotransethosomes for enhanced transdermal delivery of mangiferin against rheumatoid arthritis: formulation, characterization, invivo pharmacokinetic and pharmacodynamic evaluation. Drug Delivery. 2023;30(1):2173338. doi:10.1080/10717544.2023.2173338
  • Qindeel M, Ullah MH, Fakhar Ud D, Ahmed N, Rehman A. Recent trends, challenges and future outlook of transdermal drug delivery systems for rheumatoid arthritis therapy. J Control Release. 2020;327:595–615. doi:10.1016/j.jconrel.2020.09.016
  • Yu P, Zhang X, Cheng G, et al. Construction of a new multifunctional insomnia drug delivery system. Chem Eng J. 2022;430:132633. doi:10.1016/j.cej.2021.132633
  • Yang H, Mu W, Wei D, et al. A novel targeted and high-efficiency nanosystem for combinational therapy for Alzheimer’s disease. Adv Sci. 2020;7(19):1902906. doi:10.1002/advs.201902906
  • Fattahi N, Shahbazi M-A, Maleki A, Hamidi M, Ramazani A, Santos HA. Emerging insights on drug delivery by fatty acid mediated synthesis of lipophilic prodrugs as novel nanomedicines. J Control Release. 2020;326:556–598. doi:10.1016/j.jconrel.2020.07.012
  • Sun J, Wei C, Liu Y, et al. Progressive release of mesoporous nano-selenium delivery system for the multi-channel synergistic treatment of Alzheimer’s disease. Biomaterials. 2019;197:417–431. doi:10.1016/j.biomaterials.2018.12.027
  • Liu Y, Shen J, Shi J, et al. Functional polymeric core–shell hybrid nanoparticles overcome intestinal barriers and inhibit breast cancer metastasis. Chem Eng J. 2022;427:131742. doi:10.1016/j.cej.2021.131742
  • Mou Y, Zhang P, Lai W-F, Zhang D. Design and applications of liposome-in-gel as carriers for cancer therapy. Drug Delivery. 2022;29(1):3245–3255. doi:10.1080/10717544.2022.2139021
  • Srivastav AK, Karpathak S, Rai MK, Kumar D, Misra DP, Agarwal V. Lipid based drug delivery systems for oral, transdermal and parenteral delivery: recent strategies for targeted delivery consistent with different clinical application. J Drug Deliv Sci Technol. 2023;85:104526. doi:10.1016/j.jddst.2023.104526
  • Kansız S, Elçin YM. Advanced liposome and polymersome-based drug delivery systems: considerations for physicochemical properties, targeting strategies and stimuli-sensitive approaches. Adv Colloid Interface Sci. 2023;102930. doi:10.1016/j.cis.2023.102930
  • Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: key to more efficient oral drug delivery systems. Adv Colloid Interface Sci. 2023;313:102848. doi:10.1016/j.cis.2023.102848
  • Battistella C, Liang Y, Gianneschi NC. Innovations in disease state responsive soft materials for targeting extracellular stimuli associated with cancer, cardiovascular disease, diabetes, and beyond. Adv Mater. 2021;33(46):2007504. doi:10.1002/adma.202007504
  • Li M, Li M, Li X, et al. Preparation, characterization and ex vivo skin permeability evaluation of type I collagen-loaded liposomes. Int J Nanomed. 2023;18:1853–1871. doi:10.2147/ijn.S404494
  • Hassan AS, Hofni A, Abourehab MAS, Abdel-Rahman IAM. Ginger extract-loaded transethosomes for effective transdermal permeation and anti-inflammation in rat model. Int J Nanomed. 2023;18:1259–1280. doi:10.2147/ijn.S400604
  • Constantinou AP, Tall A, Li Q, Georgiou TK. Liquid–liquid phase separation in aqueous solutions of poly(ethylene glycol) methacrylate homopolymers. J Polym Sci. 2022;60(2):188–198. doi:10.1002/pol.20210714
  • Mandrioli M, Tura M, Scotti S, Gallina Toschi T. Fast detection of 10 cannabinoids by RP-HPLC-UV method in cannabis sativa l. Molecules. 2019;24(11). doi:10.3390/molecules24112113
  • Citti C, Pacchetti B, Vandelli MA, Forni F, Cannazza G. Analysis of cannabinoids in commercial hemp seed oil and decarboxylation kinetics studies of cannabidiolic acid (CBDA). J Pharm Biomed Anal. 2018;149:532–540. doi:10.1016/j.jpba.2017.11.044
  • Opuni KFM, Boadu JA, Amponsah SK, Okai CA. High performance liquid chromatography: a versatile tool for assaying antiepileptic drugs in biological matrices. J Chromatogr B. 2021;1179:122750. doi:10.1016/j.jchromb.2021.122750
  • Uchida T, Kadhum WR, Kanai S, Todo H, Oshizaka T, Sugibayashi K. Prediction of skin permeation by chemical compounds using the artificial membrane, Strat-M™. Eur J Pharm Sci. 2015;67:113–118. doi:10.1016/j.ejps.2014.11.002
  • Wang C, Wang J, Sun Y, et al. Enhanced stability and oral bioavailability of cannabidiol in zein and whey protein composite nanoparticles by a modified anti-solvent approach. Foods. 2022;11(3). doi:10.3390/foods11030376
  • Ujváry I, Hanuš L. Human metabolites of cannabidiol: a review on their formation, biological activity, and relevance in therapy. Cannabis Cannabinoid Res. 2016;1(1):90–101. doi:10.1089/can.2015.0012
  • Pichini S, Malaca S, Gottardi M, et al. UHPLC-MS/MS analysis of cannabidiol metabolites in serum and urine samples. application to an individual treated with medical cannabis. Talanta. 2021;223:121772. doi:10.1016/j.talanta.2020.121772
  • Meng Q, Buchanan B, Zuccolo J, Poulin -M-M, Gabriele J, Baranowski DC. A reliable and validated LC-MS/MS method for the simultaneous quantification of 4 cannabinoids in 40 consumer products. PLoS One. 2018;13(5):e0196396. doi:10.1371/journal.pone.0196396
  • Manca A, Chiara F, Mula J, et al. A new UHPLC-MS/MS method for cannabinoids determination in human plasma: a clinical tool for therapeutic drug monitoring. Biomed Pharmacother. 2022;156:113899. doi:10.1016/j.biopha.2022.113899
  • Citti C, Linciano P, Panseri S, et al. Cannabinoid profiling of hemp seed oil by liquid chromatography coupled to high-resolution mass spectrometry. Original Research Front Plant Sci. 2019;2019:10. doi:10.3389/fpls.2019.00120
  • Palazzoli F, Citti C, Licata M, et al. Development of a simple and sensitive liquid chromatography triple quadrupole mass spectrometry (LC–MS/MS) method for the determination of cannabidiol (CBD), Δ9-tetrahydrocannabinol (THC) and its metabolites in rat whole blood after oral administration of a single high dose of CBD. J Pharm Biomed Anal. 2018;150:25–32. doi:10.1016/j.jpba.2017.11.054
  • Deiana S, Watanabe A, Yamasaki Y, et al. Plasma and brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Δ9-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) in rats and mice following oral and intraperitoneal administration and CBD action on obsessive-compulsive behaviour. Psychopharmacology. 2012;219(3):859–873. doi:10.1007/s00213-011-2415-0
  • Cherniakov I, Izgelov D, Domb AJ, Hoffman A. The effect of Pro nanolipospheres (PNL) formulation containing natural absorption enhancers on the oral bioavailability of delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) in a rat model. Eur J Pharm Sci. 2017;109:21–30. doi:10.1016/j.ejps.2017.07.003
  • Williams AC, Barry BW. Penetration enhancers. Adv Drug Deliv Rev. 2012;64:128–137. doi:10.1016/j.addr.2012.09.032
  • Dragicevic-Curic N, Scheglmann D, Albrecht V, Fahr A. Temoporfin-loaded invasomes: development, characterization and in vitro skin penetration studies. J Control Release. 2008;127(1):59–69. doi:10.1016/j.jconrel.2007.12.013
  • Waheed A, Aqil M, Ahad A, et al. Improved bioavailability of raloxifene hydrochloride using limonene containing transdermal nano-sized vesicles. J Drug Deliv Sci Technol. 2019;52:468–476. doi:10.1016/j.jddst.2019.05.019
  • Lu B, Bo Y, Yi M, et al. Enhancing the solubility and transdermal delivery of drugs using ionic liquid-In-oil microemulsions. Adv Funct Mater. 2021;31(34):2102794. doi:10.1002/adfm.202102794
  • Cappel MJ, Kreuter J. Effect of nonionic surfactants on transdermal drug delivery: i. Polysorbates. Int J Pharm. 1991;69(2):143–153. doi:10.1016/0378-5173(91)90219-E
  • Phatale V, Vaiphei KK, Jha S, Patil D, Agrawal M, Alexander A. Overcoming skin barriers through advanced transdermal drug delivery approaches. J Control Release. 2022;351:361–380. doi:10.1016/j.jconrel.2022.09.025
  • Zhu Y, Xu W, Zhang J, et al. Self-microemulsifying Drug Delivery System for Improved Oral Delivery of Limonene: preparation, Characterization, in vitro and in vivo Evaluation. AAPS Pharm Sci Tech. 2019;20(4):153. doi:10.1208/s12249-019-1361-8
  • W-C L, Chiang B-H, Huang D-W, P-H L. Skin permeation of d-limonene-based nanoemulsions as a transdermal carrier prepared by ultrasonic emulsification. Ultrason Sonochem. 2014;21(2):826–832. doi:10.1016/j.ultsonch.2013.10.013
  • Juang R-S, Lin K-H. Ultrasound-assisted production of W/O emulsions in liquid surfactant membrane processes. Colloids Surf a Physicochem Eng Asp. 2004;238(1):43–49. doi:10.1016/j.colsurfa.2004.02.028
  • Capek I. Degradation of kinetically-stable o/w emulsions. Adv Colloid Interface Sci. 2004;107(2):125–155. doi:10.1016/S0001-8686(03)00115-5
  • Haq A, Goodyear B, Ameen D, Joshi V, Michniak-Kohn B. Strat-M® synthetic membrane: permeability comparison to human cadaver skin. Int J Pharm. 2018;547(1):432–437. doi:10.1016/j.ijpharm.2018.06.012
  • Bolla PK, Clark BA, Juluri A, Cheruvu HS, Renukuntla J. Evaluation of formulation parameters on permeation of Ibuprofen from topical formulations using Strat-M® membrane. Pharmaceutics. 2020;12(2):151. doi:10.3390/pharmaceutics12020151
  • Czajkowska-Kośnik A, Szymańska E, Winnicka K. Nanostructured lipid carriers (NLC)-based gel formulations as etodolac delivery: from gel preparation to permeation study. Molecules. 2023;28(1). doi:10.3390/molecules28010235
  • Pulsoni I, Lubda M, Aiello M, et al. Comparison between Franz diffusion cell and a novel micro-physiological system for in vitro penetration assay using different skin models. SLAS Technol. 2022;27(3):161–171. doi:10.1016/j.slast.2021.12.006
  • Shukla T, Upmanyu N, Agrawal M, Saraf S, Saraf S, Alexander A. Biomedical applications of microemulsion through dermal and transdermal route. Biomed Pharmacother. 2018;108:1477–1494. doi:10.1016/j.biopha.2018.10.021
  • Stinchcomb Audra L, Banks SL; inventors; ALLTRANZ INC, assignee. Formulations of cannabidiol and prodrugs of cannabidiol and methods of using the same. US patent US 2010/0273895 A1. patent application US 76951910 A. 2010.
  • Stinchcomb Audra L, Banks SL; inventors; ALLTRANZ INC, assignee. Transdermal formulations of cannabidiol comprising a penetration enhancer and methods of using the same. CA patent CA 2760460 C. patent application CA 2760460 A; 2019.
  • Stinchcomb Audra L, Nalluri Buchi N; Inventors; STINCHCOMB AUDRA L, NALLURI BUCHI N, ALLTRANZ LLC, assignee. Transdermal delivery of cannabidiol. US patent US 8435556 B2. patent application US 51122609 A; 2013.
  • Stinchcomb Audra L, Banks SL, Golinski Miroslaw J, Howard Jeffery L, Hammell Dana C; inventors ZYNERBA PHARMACEUTICALS INC, assignee. Use of cannabidiol prodrugs in topical and transdermal administration with microneedles. US patent US 9533942 B2. patent application US 201414539824 A; 2017.
  • Morakul B, Junyaprasert VB, Sakchaisri K, Teeranachaideekul V. Cannabidiol-Loaded Nanostructured Lipid Carriers (NLCs) for Dermal Delivery: enhancement of Photostability, Cell Viability, and Anti-Inflammatory Activity. Pharmaceutics. 2023;15(2):537. doi:10.3390/pharmaceutics15020537
  • Franzè S, Angelo L, Casiraghi A, Minghetti P, Cilurzo F. Design of Liposomal Lidocaine/Cannabidiol Fixed Combinations for Local Neuropathic Pain Treatment. Pharmaceutics. 2022;14(9):1915. doi:10.3390/pharmaceutics14091915
  • Franzè S, Ricci C, Del Favero E, Rama F, Casiraghi A, Cilurzo F. Micelles-in-liposome systems obtained by proliposomal approach for cannabidiol delivery: structural features and skin penetration. Mol Pharm. 2023;20(7):3393–3402. doi:10.1021/acs.molpharmaceut.3c00044
  • Demisli S, Galani E, Goulielmaki M, et al. Encapsulation of cannabidiol in oil-in-water nanoemulsions and nanoemulsion-filled hydrogels: a structure and biological assessment study. J Colloid Interface Sci. 2023;634:300–313. doi:10.1016/j.jcis.2022.12.036
  • Sharkawy A, Silva AM, Rodrigues F, Barreiro F, Rodrigues A. Pickering emulsions stabilized with chitosan/collagen peptides nanoparticles as green topical delivery vehicles for cannabidiol (CBD). Colloids Surf a Physicochem Eng Asp. 2021;631:127677. doi:10.1016/j.colsurfa.2021.127677
  • Pimenta AFR, Vieira AP, Colaço R, et al. Controlled release of moxifloxacin from intraocular lenses modified by Ar plasma-assisted grafting with AMPS or SBMA: an in vitro study. Colloids Surf B. 2017;156:95–103. doi:10.1016/j.colsurfb.2017.04.060
  • Naeem A, Yu C, Liu Y, Feng Y, Fan J, Guan Y. Study of Gelatin-grafted-2-Acrylamido-2-methylpropane sulfonic acid hydrogels as a controlled release vehicle for amorphous solid dispersion of Tripterygium Wilfordii bioactive constituents. Arab J Chem. 2023;16(10):105139. doi:10.1016/j.arabjc.2023.105139
  • Cheng F-M, Chen H-X, Li H-D. Recent advances in tough and self-healing nanocomposite hydrogels for shape morphing and soft actuators. Eur Polym J. 2020;124:109448. doi:10.1016/j.eurpolymj.2019.109448
  • Brookes A, Jewell A, Feng W, Bradshaw TD, Butler J, Gershkovich P. Oral lipid-based formulations alter delivery of cannabidiol to different anatomical regions in the brain. Int J Pharm. 2023;635:122651. doi:10.1016/j.ijpharm.2023.122651
  • Zgair A, Wong JCM, Sabri A, et al. Development of a simple and sensitive HPLC–UV method for the simultaneous determination of cannabidiol and Δ9-tetrahydrocannabinol in rat plasma. J Pharm Biomed Anal. 2015;114:145–151. doi:10.1016/j.jpba.2015.05.019
  • Siemens AJ, Walczak D, Buckley FE. Characterization of blood disappearance and tissue distribution of [3H]cannabidiol. Biochem Pharmacol. 1980;29(3):462–464. doi:10.1016/0006-2952(80)90532-8
  • Muresan P, Woodhams S, Smith F, et al. Evaluation of cannabidiol nanoparticles and nanoemulsion biodistribution in the central nervous system after intrathecal administration for the treatment of pain. Nanomedicine. 2023;49:102664. doi:10.1016/j.nano.2023.102664
  • Beal R, Fitzsimmons N, McMahon A, Sand B, Martinez K; inventors; AMPERSAND BIOPHARMACEUTICALS INC, assignee. Transdermal penetrant formulations containing cannabidiol. US patent US 10842758 B1. patent application US 201916546269 A; 2020.
  • Fitzsimmons N, Beal R, McMahon A, Sand B, Martinez K; Inventors; AMPERSAND BIOPHARMACEUTICALS INC, DYVE BIOSCIENCES INC, assignee. Transdermal penetrant formulations containing cannabidiol. US patent US 11026896 B2. patent application US 201916546270 A; 2021.
  • Fitzsimmons N, Beal R, McMahon A, Sand B, Martinez K; Inventors; DYVE BIOSCIENCES INC, assignee. Transdermal penetrant formulations containing cannabidiol. US patent US 2023/0038462 A1. patent application US 202017422434 A; 2023.
  • Khan SU, Ullah M, Saeed S, et al. Nanotherapeutic approaches for transdermal drug delivery systems and their biomedical applications. Eur Polym J. 2024;207:112819. doi:10.1016/j.eurpolymj.2024.112819
  • Leong MY, Kong YL, Burgess K, Wong WF, Sethi G, Looi CY. Recent Development of Nanomaterials for Transdermal Drug Delivery. Biomedicines. 2023;11(4):1124. doi:10.3390/biomedicines11041124
  • Münch S, Wohlrab J, Neubert RHH. Dermal and transdermal delivery of pharmaceutically relevant macromolecules. Eur J Pharm Biopharm. 2017;119:235–242. doi:10.1016/j.ejpb.2017.06.019
  • Heuschkel S, Goebel A, Neubert RHH. Microemulsions—modern colloidal carrier for dermal and transdermal drug delivery. J Pharmaceut Sci. 2008;97(2):603–631. doi:10.1002/jps.20995
  • Sintov AC, Shapiro L. New microemulsion vehicle facilitates percutaneous penetration in vitro and cutaneous drug bioavailability in vivo. J Control Release. 2004;95(2):173–183. doi:10.1016/j.jconrel.2003.11.004
  • Yuan Y, S-m L, F-k M, D-f Z. Investigation of microemulsion system for transdermal delivery of meloxicam. Int J Pharm. 2006;321(1):117–123. doi:10.1016/j.ijpharm.2006.06.021
  • Obradović S, Poša M. The influence of the structure of selected Brij and Tween homologues on the thermodynamic stability of their binary mixed micelles. J Chem Thermodyn. 2017;110:41–50. doi:10.1016/j.jct.2017.01.020
  • Ćirin D, Krstonošić V, Poša M. Properties of poloxamer 407 and polysorbate mixed micelles: influence of polysorbate hydrophobic chain. J Ind Eng Chem. 2017;47:194–201. doi:10.1016/j.jiec.2016.11.032
  • Wang X, Gao Y. Effects of length and unsaturation of the alkyl chain on the hydrophobic binding of curcumin with Tween micelles. Food Chem. 2018;246:242–248. doi:10.1016/j.foodchem.2017.11.024
  • Chen S-Y, Kokalari I, Parnell SR, et al. Structure property relationship of micellar waterborne poly(urethane-urea): tunable mechanical properties and controlled release profiles with amphiphilic triblock copolymers. Langmuir. 2023;39(29):10033–10046. doi:10.1021/acs.langmuir.3c00921
  • 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):438–470. doi:10.3390/pharmaceutics7040438
  • Mo R, Zhang H, Xu Y, et al. Transdermal drug delivery via microneedles to mediate wound microenvironment. Adv Drug Deliv Rev. 2023;195:114753. doi:10.1016/j.addr.2023.114753
  • Yang D, Chen M, Sun Y, et al. Microneedle-mediated transdermal drug delivery for treating diverse skin diseases. Acta Biomater. 2021;121:119–133. doi:10.1016/j.actbio.2020.12.004
  • Sabbagh F, Kim BS. Recent advances in polymeric transdermal drug delivery systems. J Control Release. 2022;341:132–146. doi:10.1016/j.jconrel.2021.11.025
  • An H, Gu Z, Huang Z, et al. Novel microneedle platforms for the treatment of wounds by drug delivery: a review. Colloids Surf B. 2024;233:113636. doi:10.1016/j.colsurfb.2023.113636
  • Zheng L, Chen Y, Gu X, et al. Co-delivery of drugs by adhesive transdermal patches equipped with dissolving microneedles for the treatment of rheumatoid arthritis. J Control Release. 2024;365:274–285. doi:10.1016/j.jconrel.2023.11.029
  • Liu R, Li A, Lang Y, et al. Stimuli-responsive polymer microneedles: a rising transdermal drug delivery system and Its applications in biomedical. J Drug Deliv Sci Technol. 2023;88:104922. doi:10.1016/j.jddst.2023.104922
  • Sartawi Z, Blackshields C, Faisal W. Dissolving microneedles: applications and growing therapeutic potential. J Control Release. 2022;348:186–205. doi:10.1016/j.jconrel.2022.05.045
  • Zhang X, Hasani-Sadrabadi MM, Zarubova J, et al. Immunomodulatory microneedle patch for periodontal tissue regeneration. Matter. 2022;5(2):666–682. doi:10.1016/j.matt.2021.11.017