353
Views
0
CrossRef citations to date
0
Altmetric
Review Articles

Overcoming challenges in dermal and transdermal delivery of herbal therapeutics with polymeric microneedles

, , , , , , & ORCID Icon show all
Pages 364-396 | Received 19 May 2023, Accepted 16 Nov 2023, Published online: 24 Nov 2023

References

  • Raj GM, Raveendran R. Introduction to basics of pharmacology and toxicology: volume 1: general and molecular pharmacology: principles of drug action. Singapore: Springer Nature; 2019. DOI: 10.1007/978-981-32-9779-1
  • Benet LZ, Hosey CM, Ursu O, et al. BDDCS, the rule of 5 and drugability. Adv Drug Deliv Rev. 2016;101:89–98. doi: 10.1016/j.addr.2016.05.007.
  • Hughes JP, Rees S, Kalindjian SB, et al. Principles of early stages of drug discovery. Br J Pharmacol. 2011;162(6):1239–1249. doi: 10.1111/j.1476-5381.2010.01127.x.
  • Marschütz MK, Bernkop-Schnürch A. Oral peptide drug delivery: polymer–inhibitor conjugates protecting insulin from enzymatic degradation in vitro. Biomaterials. 2000;21(14):1499–1507. doi: 10.1016/s0142-9612(00)00039-9.
  • Dahan A, Miller JM, Amidon GL. Prediction of solubility and permeability class membership: provisional BCS classification of the world’s top oral drugs. Aaps J. 2009;11(4):740–746. doi: 10.1208/s12248-009-9144-x.
  • Kaur G, Kaur P, Madaan P, et al. Herbal bioactives in transdermal drug delivery system. In Herbal bioactive-based drug delivery systems. Chennai, India: Academic Press; 2022. doi: 10.1016/B978-0-12-824385-5.00016-9.
  • Chen RP, Chavda VP, Patel AB, et al. Phytochemical delivery through transferosome (phytosome): an advanced transdermal drug delivery for complementary medicines. Front Pharmacol. 2022;13:850862. doi: 10.3389/fphar.2022.850862.
  • Rosenstein ED. Topical agents in the treatment of rheumatic disorders. Rheum Dis Clin North Am. 1999;25(4):899–918, viii. doi: 10.1016/s0889-857x(05)70109-5.
  • Murthy SN, Shivakumar HN. Topical and transdermal drug delivery. In Handbook of non-invasive drug delivery systems. New York: Elsevier Inc.; 2010. doi: 10.1016/B978-0-8155-2025-2.10001-0.
  • Kolarsick PAJ, Kolarsick MA, Goodwin C. Anatomy and physiology of the skin. J Dermatol Nurses Assoc. 2011;3(4):203–213. doi: 10.1097/JDN.0b013e3182274a98.
  • Arunkumar N., Drug Delivery Aspects of Herbal Medicines. In: Arumugam S, Watanabe K.,  editors. Japanese Kampo medicines for the treatment of common diseases: focus on inflammation. Academic Press; 2017. p. 143–164.
  • Quinn HL, Kearney M-C, Courtenay AJ, et al. The role of microneedles for drug and vaccine delivery. Expert Opin Drug Deliv. 2014;11(11):1769–1780. doi: 10.1517/17425247.2014.938635.
  • Ramadon D, McCrudden MTC, Courtenay AJ, et al. Enhancement strategies for transdermal drug delivery systems: current trends and applications. Drug Deliv Transl Res. 2022;12(4):758–791. doi: 10.1007/s13346-021-00909-6.
  • Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008;26(11):1261–1268. doi: 10.1038/nbt.1504.
  • Bariya SH, Gohel MC, Mehta TA, et al. Microneedles: an emerging transdermal drug delivery system. J Pharm Pharmacol. 2012;64(1):11–29. doi: 10.1111/j.2042-7158.2011.01369.x.
  • Matriano JA, Cormier M, Johnson J, et al. Macroflux® microprojection array patch technology: a new and efficient approach for intracutaneous immunization. Pharm Res. 2002;19(1):63–70. doi: 10.1023/a:1013607400040.
  • Swathi HP, Anusha MV, Paul GJ, et al. Effect of gamma sterilization on the properties of microneedle array transdermal patch system. Drug Dev Ind Pharm. 2020;46(4):606–620. doi: 10.1080/03639045.2020.1742144.
  • Yang J, Liu X, Fu Y, et al. Recent advances of microneedles for biomedical applications: drug delivery and beyond. Acta Pharm Sin B. 2019;9(3):469–483. doi: 10.1016/j.apsb.2019.03.007.
  • Wang Q, Yao G, Dong P, et al. Investigation on fabrication process of dissolving microneedle arrays to improve effective needle drug distribution. Eur J Pharm Sci. 2015;66:148–156. doi: 10.1016/j.ejps.2014.09.011.
  • DeMuth PC, Su X, Samuel RE, et al. Nanolayered microneedles for transcutaneous delivery of polymer nanoparticles and plasmid DNA. Adv Mater. 2010;22(43):4851–4856. doi: 10.1002/adma.201001525.
  • Jakka D, Matadh AV, Shankar VK, et al. Polymer coated polymeric (PCP) microneedles for controlled delivery of drugs (dermal and intravitreal). J Pharm Sci. 2022;111(10):2867–2878. doi: 10.1016/j.xphs.2022.05.023.
  • Anusha VM, Jakka D, Pragathi SG, et al. Polymer coated polymeric (PCP) microneedles for controlled dermal delivery of curcumin. AAPS PharmSciTech. 2023;24:9.
  • Anusha VM, Jakka D, Pragathi SG, et al. Polymer coated polymeric microneedles for intravitreal delivery of dexmethasone. Exp Eye Res. 2023;231:109467.
  • Jakka D, Matadh AV, Shivakumar HN, et al. Polymer coated polymeric (PCP) microneedles for sampling of drugs and biomarkers from tissues. Eur J Pharm Sci. 2022;175:106203. doi: 10.1016/j.ejps.2022.106203.
  • Walters KA, Lane ME. Dermal and transdermal drug delivery systems: an overview and recent advancements. In Tapash K Ghosh, editor. Dermal Drug Delivery 1st ed., CRC Press; 2020. pp 1–60.
  • Panda A, Matadh VA, Suresh S, et al. Non-dermal applications of microneedle drug delivery systems. Drug Deliv Transl Res. 2021;12(1):67–78. doi: 10.1007/s13346-021-00922-9.
  • Larrañeta E, McCrudden MTC, Courtenay AJ, et al. Microneedles: a new frontier in nanomedicine delivery. Pharm Res. 2016;33(5):1055–1073. doi: 10.1007/s11095-016-1885-5.
  • Gittard SD, Miller PR, Boehm RD, et al. Multiphoton microscopy of transdermal quantum dot delivery using two photon polymerization-fabricated polymer microneedles. Faraday Discuss. 2011;149:171–185. doi: 10.1039/c005374k.
  • Ganeson K, Alias AH, Murugaiyah V, et al. Microneedles for efficient and precise drug delivery in cancer therapy. Pharmaceutics. 2023;15(3):744. doi: 10.3390/pharmaceutics15030744.
  • Chen J, Ren H, Zhou P, et al. Microneedle mediated drug delivery for cutaneous disease. Front Bioeng Biotechnol. 2022;10:1032041. doi: 10.3389/fbioe.2022.1032041.
  • Guo L, Shi M, Song N, et al. Anchorage of curcumin onto PVP enhances anti-tumor effect of curcumin. Med Chem Res. 2019;28(5):646–656. doi: 10.1007/s00044-019-02319-3.
  • Sun M, Zhang Y, He Y, et al. Green synthesis of carrier-free curcumin nanodrugs for light-activated breast cancer photodynamic therapy. Colloids Surf B Biointerfaces. 2019;180:313–318. doi: 10.1016/j.colsurfb.2019.04.061.
  • Wahlang B, Pawar YB, Bansal AK. Identification of permeability-related hurdles in oral delivery of curcumin using the caco-2 cell model. Eur J Pharm Biopharm. 2011;77(2):275–282. doi: 10.1016/j.ejpb.2010.12.006.
  • Abdel-Hafez SM, Hathout RM, Sammour OA. Attempts to enhance the anti-cancer activity of curcumin as a magical oncological agent using transdermal delivery. Adv Tradit Med (ADTM). 2021;21(1):15–29. doi: 10.1007/s13596-020-00439-5.
  • Cheng Z, Lin H, Wang Z, et al. Preparation and characterization of dissolving hyaluronic acid composite microneedles loaded micelles for delivery of curcumin. Drug Deliv Transl Res. 2020;10(5):1520–1530. doi: 10.1007/s13346-020-00735-2.
  • Shan Y, Tan B, Zhang M, et al. Restorative biodegradable two-layered hybrid microneedles for melanoma photothermal/chemo co-therapy and wound healing. J Nanobiotechnol. 2022;20(1):238. doi: 10.1186/s12951-022-01426-5.
  • Zhou X, Luo Z, Baidya A, et al. Biodegradable β cyclodextrin conjugated gelatin methacryloyl microneedle for delivery of water insoluble drug. Adv Healthc Mater. 2020;9:2000527.
  • Uddin MJ, Scoutaris N, Klepetsanis P, et al. Inkjet printing of transdermal microneedles for the delivery of anticancer agents. Int J Pharm. 2015;494(2):593–602. doi: 10.1016/j.ijpharm.2015.01.038.
  • Sofroniou C, Baglioni M, Mamusa M, et al. Self-Assembly of soluplus in aqueous solutions: characterization and prospectives on perfume encapsulation. ACS Appl Mater Interfaces. 2022;14(12):14791–14804. doi: 10.1021/acsami.2c01087.
  • Abd-El-Azim H, Tekko IA, Ali A, et al. Hollow microneedle assisted intradermal delivery of hypericin lipid nanocapsules with light enabled photodynamic therapy against skin cancer. J Control Release. 2022;348:849–869. doi: 10.1016/j.jconrel.2022.06.027.
  • Kacerovská D, Pizinger K, Majer F, et al. Photodynamic therapy of nonmelanoma skin cancer with topical hypericum perforatum extract-a pilot study. Photochem Photobiol. 2008;84(3):779–785. doi: 10.1111/j.1751-1097.2007.00260.x.
  • Muresan P, McCrorie P, Smith F, et al. Development of nanoparticle loaded microneedles for drug delivery to a brain tumor resection site. Eur J Pharm Biopharm. 2023;182:53–61. doi: 10.1016/j.ejpb.2022.11.016.
  • Śledziński P, Zeyland J, Słomski R, et al. The current state and future perspectives of cannabinoids in cancer biology. Cancer Med. 2018;7(3):765–775. doi: 10.1002/cam4.1312.
  • Khodadadi H, Salles EP, et al. Inhalent cannabidiol inhibits glioblastoma progression through regulation of tumor environment. Cannabis Cannabinoid Res. 2021;14:14791–14804.
  • Gu Y, Bian Q, Zhou Y, et al. Hair follicle-targeting drug delivery strategies for the management of hair follicle-associated disorders. Asian J Pharm Sci. 2022;17(3):333–352. doi: 10.1016/j.ajps.2022.04.003.
  • Park H, Lee J, Jeong S, et al. Lipase sensitive transfersomes based on photosensitizer/polymerizable lipid conjugate for selective antimicrobial photodynamic therapy of acne. Adv Healthc Mater. 2016;5(24):3139–3147. doi: 10.1002/adhm.201600815.
  • Pornpattananangkul D, Fu V, Thamphiwatana S, et al. In vivo treatment of Propionibacterium acnes infection with liposomal lauric acids. Adv Healthc Mater. 2013;2(10):1322–1328. doi: 10.1002/adhm.201300002.
  • Zhang T, Sun B, Guo J, et al. Active pharmaceutical ingredient poly (ionic liquid)-based microneedles for the treatment of skin acne infection. Acta Biomater. 2020;115:136–147. doi: 10.1016/j.actbio.2020.08.023.
  • Ning X, Wiraja C, Chew WTS, et al. Transdermal delivery of Chinese herbal medicine extract using dissolvable microneedles for hypertrophic scar treatment. Acta Pharm Sin B. 2021;11(9):2937–2944. doi: 10.1016/j.apsb.2021.03.016.
  • Wang R, Yin R, Zhou W, et al. Shikonin and its derivatives: a patent review. Expert Opin Ther Pat. 2012;22(9):977–997. doi: 10.1517/13543776.2012.709237.
  • Fan C, Xie Y, Dong Y, et al. Investigating the potential of Shikonin as a novel hypertrophic scar treatment. J Biomed Sci. 2015;22(1):70. doi: 10.1186/s12929-015-0172-9.
  • Patel P, Patel H, Panchal S, et al. Formulation strategies for drug delivery of tacrolimus: an overview. Int J Pharm Investig. 2012;2(4):169–175. doi: 10.4103/2230-973X.106981.
  • Yu K, Yu X, Cao S, et al. Layered dissolving microneedles as a need-based delivery system to simultaneously alleviate skin and joint lesions in psoriatic arthritis. Acta Pharm Sin B. 2021;11(2):505–519. doi: 10.1016/j.apsb.2020.08.008.
  • Chen DI, Shen J, Zhao W, et al. Osteoarthritis: toward a comprehensive understanding of pathological mechanism. Bone Res. 2017;5(1):16044. doi: 10.1038/boneres.2016.44.
  • Gerwin N, Hops C, Lucke A. Intraarticular drug delivery in osteoarthritis. Adv Drug Deliv Rev. 2006;58(2):226–242. doi: 10.1016/j.addr.2006.01.018.
  • Li E, Zhang J. Therapeutic effects of triptolide from Tripterygium wilfordii hook. f. on interleukin-1-beta-induced osteoarthritis in rats. Eur J Pharmacol. 2020;883:173341. doi: 10.1016/j.ejphar.2020.173341.
  • Zeng H, Zhu X, Tian Q, et al. In vivo antitumor effects of carboxymethyl chitosan-conjugated triptolide after oral administration. Drug Deliv. 2020;27(1):848–854. doi: 10.1080/10717544.2020.1770370.
  • Chen H, Chang X, Weng T, et al. A study of microemulsion systems for transdermal delivery of triptolide. J Control Release. 2004;98(3):427–436. doi: 10.1016/j.jconrel.2004.06.001.
  • Zhou P, Chen C, Yue X, et al. Strategy for osteoarthritis therapy: improved the delivery of triptolide using liposome-loaded dissolving microneedle arrays. Int J Pharm. 2021;609:121211. doi: 10.1016/j.ijpharm.2021.121211.
  • Jamaledin R, Yiu CK, Zare EN, et al. Advances in antimicrobial microneedle patches for combating infections. Adv Mater. 2020;32:2002129.
  • Yao S, Luo Y, Wang Y. Engineered microneedles arrays for wound healing. Eng Regen. 2022;3(3):232–240. doi: 10.1016/j.engreg.2022.05.003.
  • Barnum L, Samandari M, Schmidt TA, et al. Microneedle arrays for the treatment of chronic wounds. Expert Opin Drug Deliv. 2020;17(12):1767–1780. doi: 10.1080/17425247.2020.1819787.
  • Chi J, Sun L, Cai L, et al. Chinese herb microneedle patch for wound healing. Bioact Mater. 2021;6(10):3507–3514. doi: 10.1016/j.bioactmat.2021.03.023.
  • Wu Q, Dong L, Liu J, et al. Transdermal treatment with Chinese herb medicine: theory and clinical applications. Science, 2015;350:582–583.
  • Hashim P, Sidek H, Helan MHM, et al. Triterpene composition and bioactivities of Centella asiatica. Molecules. 2011;16(2):1310–1322. doi: 10.3390/molecules16021310.
  • Arribas-Lopez E, Zand N, Ojo O, et al. A systematic review of the effect of Centella asiatica on wound healing. Int J Environ Res Public Health. 2022;19(6):3266. doi: 10.3390/ijerph19063266.
  • Frydman GH, Olaleye D, Annamalai D, et al. Manuka honey microneedles for enhanced wound healing and the prevention and/or treatment of methicillin-resistant Staphylococcus aureus (MRSA) surgical site infection. Sci Rep. 2020;10(1):13229. doi: 10.1038/s41598-020-70186-9.
  • Park SY, Lee HU, Lee Y-C, et al. Wound healing potential of antibacterial microneedles loaded with green tea extracts. Mater Sci Eng C Mater Biol Appl. 2014;42:757–762. doi: 10.1016/j.msec.2014.06.021.
  • Bao C, Li Z, Liang S, et al. Microneedle patch delivery of capsaicin containing α lactalbumin nanomicelles to adipocytes achieves potent anti obesity effects. Adv Funct Mater. 2021;31:2011130.
  • Feng Y, Chang S, Jing Z, et al. Transdermal delivery of sinapine thiocyanate by gelatin microspheres and hyaluronic acid microneedles for allergic asthma in Guinea pigs. Int J Pharm. 2022;623:121899. doi: 10.1016/j.ijpharm.2022.121899.
  • Tang XY, Dai ZQ, Zeng JX, et al. Pharmacokinetics, hepatic disposition, and heart tissue distribution of 14 compounds in rat after oral administration of Qi Li Qiang Xin capsule via ultrahigh performance liquid chromatography coupled with triple quadrupole tandem mass spectrometry. J Sep Sci. 2022;45(13):2177–2189. doi: 10.1002/jssc.202101008.
  • Wentao F, Yuwei H, Lisheng W, et al. Effect of stimulating the acupoints Feishu (BL 13) and Dazhui (GV 14) on transdermal uptake of sinapine thiocyanate in asthma gel. J Tradit Chin Med. 2017;37(4):503–509. doi: 10.1016/S0254-6272(17)30157-7.
  • Association A. Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2018;14:367–429.
  • Nguyen TT, Van GV, et al. Current advances in transdermal delivery of drugs for Alzheimer’s disease. Indian J Pharmacol. 2017;49:145–154.
  • Ferreira A, Rodrigues M, Fortuna A, et al. Huperzine a from Huperzia serrata: a review of its sources, chemistry, pharmacology and toxicology. Phytochem Rev. 2016;15(1):51–85. doi: 10.1007/s11101-014-9384-y.
  • Wang R, Yan H, Tang X. Progress in studies of huperzine A, a natural cholinesterase inhibitor from Chinese herbal medicine. Acta Pharmacol Sin. 2006;27(1):1–26. doi: 10.1111/j.1745-7254.2006.00255.x.
  • Yan Q, Wang W, Weng J, et al. Dissolving microneedles for transdermal delivery of huperzine a for the treatment of Alzheimer’s disease. Drug Deliv. 2020;27(1):1147–1155. doi: 10.1080/10717544.2020.1797240.
  • Jeon KI, Xu X, Aizawa T, et al. Vinpocetine inhibits NF-κB–dependent inflammation via an IKK-dependent but PDE-independent mechanism. Proc Natl Acad Sci U S A. 2010;107(21):9795–9800. doi: 10.1073/pnas.0914414107.
  • Srivastava PK, Thakkar HP. Vinpocetine loaded ultradeformable liposomes as fast dissolving microneedle patch: tackling treatment challenges of dementia. Eur J Pharm Biopharm. 2020;156:176–190. doi: 10.1016/j.ejpb.2020.09.006.
  • Hua L, Weisan P, Jiayu L, et al. Preparation, evaluation, and NMR characterization of vinpocetine microemulsion for transdermal delivery. Drug Dev Ind Pharm. 2004;30(6):657–666. doi: 10.1081/ddc-120039183.
  • Rossi BB, Falcao CAB, Zanchetta B, et al. Performance of elastic liposomes for topical treatment of cutaneous leishmaniasis. In Nanocosmetics Nanomedicines New Approaches Skin Care. Berlin, Heidelberg: Springer, 2011; pp. 181–196. doi: 10.1007/978-3-642-19792-5_9.
  • Joy D, Jose J, Bibi S, et al. Development of microneedle patch loaded with Bacopa monnieri solid lipid nanoparticles for the effective management of Parkinson’s disease. Bioinorg Chem Appl. 2022;2022:9150205–9150217. doi: 10.1155/2022/9150205.
  • Chandran R, Mohd Tohit ER, Stanslas J, et al. Investigation and optimization of hydrogel microneedles for transdermal delivery of caffeine. Tissue Eng Part C Methods. 2022;28(10):545–556. doi: 10.1089/ten.TEC.2022.0045.
  • Luo L, Lane ME. Topical and transdermal delivery of caffeine. Int J Pharm. 2015;490(1-2):155–164. doi: 10.1016/j.ijpharm.2015.05.050.
  • Tao SL, Desai TA. Gastrointestinal patch systems for oral drug delivery. Drug Discov Today. 2005;10(13):909–915. doi: 10.1016/S1359-6446(05)03489-6.
  • Dangol M, Kim S, Li CG, et al. Anti-obesity effect of a novel caffeine-loaded dissolving microneedle patch in high-fat diet-induced obese C57BL/6J mice. J Control Release. 2017;265:41–47. doi: 10.1016/j.jconrel.2017.03.400.
  • Masclaux C, Valadier MH, Brugière N, et al. Characterization of the sink/source transition in tobacco (Nicotiana tabacum L.) shoots in relation to nitrogen management and leaf senescence. Planta. 2000;211(4):510–518. doi: 10.1007/s004250000310.
  • Panda A, Sharma PK, Shivakumar HN, et al. Nicotine loaded dissolving microneedles for nicotine replacement therapy. J Drug Deliv Sci Technol. 2021;61:102300. doi: 10.1016/j.jddst.2020.102300.
  • Le HJ. Role of nicotine pharmacokinetics in nicotine addiction and nicotine replacement therapy: a review. Int J Tuberc Lung Dis. 2003;7:811–819.
  • Hou A, Cohen B, Haimovic A, et al. Microneedling: a comprehensive review. Dermatol Surg. 2017;43(3):321–339. doi: 10.1097/DSS.0000000000000924.
  • Kim JH, Shim SE, Kim JY, et al. A literature review of the microneedle therapy system for hair loss. J Acupunct Res. 2020;37(4):203–208. doi: 10.13045/jar.2020.00234.
  • Yoon HJ. A case study of androgenetic alopecia in woman improved by pharmacopuncture therapy and needle-embedding therapy. J Korean Med Ophthalmol Otolaryngol Dermatol. 2014;27(3):162–170. doi: 10.6114/jkood.2014.27.3.162.
  • Han SK, Lee SJ, Ha HY. Skin moisturizing effects of a microneedle patch containing hyaluronic acid and loniceraeflos. Processes. 2021;9(2):321. doi: 10.3390/pr9020321.
  • Ruiz PA, Braune A, Hölzlwimmer G, et al. Quercetin inhibits TNF-induced NF-κ B transcription factor recruitment to proinflammatory gene promoters in murine intestinal epithelial cells. J Nutr. 2007;137(5):1208–1215. doi: 10.1093/jn/137.5.1208.
  • Vicentini FTMC, Fonseca YM, Pitol DL, et al. Evaluation of protective effect of a water-in-oil microemulsion incorporating quercetin against UVB-induced damage in hairless mice skin. J Pharm Pharm Sci. 2010;13(2):274–285. doi: 10.18433/j3830g.
  • Bose S, Du Y, Takhistov P, et al. Formulation optimization and topical delivery of quercetin from solid lipid based nanosystems. Int J Pharm. 2013;441(1-2):56–66. doi: 10.1016/j.ijpharm.2012.12.013.
  • Paleco R, Vucen SR, Crean AM, et al. Enhancement of the in vitro penetration of quercetin through pig skin by combined microneedles and lipid microparticles. Int J Pharm. 2014;472(1-2):206–213. doi: 10.1016/j.ijpharm.2014.06.010.
  • Paul R, Saville AC, Hansel JC, et al. Extraction of plant DNA by microneedle patch for rapid detection of plant diseases. ACS Nano. 2019;13(6):6540–6549. doi: 10.1021/acsnano.9b00193.
  • Guglani L. Changing the paradigm–treating the basic defect in cystic fibrosis. Indian J Pediatr. 2015;82(8):727–736. doi: 10.1007/s12098-015-1786-3.
  • Mishra A, Greaves R, Massie J. The relevance of sweat testing for the diagnosis of cystic fibrosis in the genomic era. Clin Biochem Rev Assoc Clin Biochem. 2005;26:135.
  • Fatima T, Ajjarapu S, Shankar VK, et al. Topical pilocarpine formulation for diagnosis of cystic fibrosis. J Pharm Sci. 2020;109(5):1747–1751. doi: 10.1016/j.xphs.2020.01.030.
  • Li S, Hart K, Norton N, et al. Administration of pilocarpine by microneedle patch as a novel method for cystic fibrosis sweat testing. Bioeng Transl Med. 2021;6:e10222.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.