532
Views
19
CrossRef citations to date
0
Altmetric
Review Article

Multifunctional nanoparticles for targeting cancer and inflammatory diseases

, , &
Pages 888-903 | Received 09 Jun 2013, Accepted 04 Aug 2013, Published online: 12 Sep 2013

References

  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA: Cancer J Clin 2013;63:11–30
  • Balkwill F, Coussens LM. Cancer: an inflammatory link. Nature 2004;431:405–6
  • Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860–7
  • Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet 2001;357:539–45
  • Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 1986;315:1650–9
  • Sica A, Allavena P, Mantovani A. Cancer related inflammation: the macrophage connection. Cancer Lett 2008;267:204–15
  • Mantovani A, Schioppa T, Porta C, et al. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev 2006;25:315–22
  • Allen TM, Cullis PR. Drug delivery systems: entering the mainstream. Science 2004;303:1818–22
  • Iyer AK, Khaled G, Fang J, et al. Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2006;11:812–18
  • Cho K, Wang X, Nie S, et al. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 2008;14:1310–16
  • Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 1986;46:6387–92
  • Maeda H, Wu J, Sawa T, et al. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release 2000;65:271–84
  • Senger DR, Galli SJ, Dvorak AM, et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983;219:983–5
  • Dvorak HF, Nagy JA, Feng D, et al. Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol 1999;237:97–132
  • Senger DR, Galli SJ, Dvorak AM, et al. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983;219:983–5
  • Maeda H, Bharate GY, Daruwalla J. Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect. Eur J Pharm Biopharm 2009;71:409–19
  • Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010;148:135–46
  • Jang SH, Wientjes MG, Lu D, et al. Drug delivery and transport to solid tumors. Pharm Res 2003;20:1337–50
  • Maeda H, Bharate GY, Daruwalla J. Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect. Eur J Pharm Biopharm 2009;71:409–19
  • Torchilin VP, Rammohan R, Weissig V, et al. TAT peptide on the surface of liposomes affords their efficient intracellular delivery even at low temperature and in the presence of metabolic inhibitors. Proc Natl Acad Sci USA 2001;98:8786–91
  • Ganesh S, Iyer AK, Morrissey DV, et al. Hyaluronic acid based self-assembling nanosystems for CD44 target mediated siRNA delivery to solid tumors. Biomaterials 2013;34:3489--502
  • Torchilin VP. Recent approaches to intracellular delivery of drugs and DNA and organelle targeting. Ann Rev Biomed Eng 2006;8:343–75
  • Ganesh S, Iyer AK, Morrissey DV, et al. Hyaluronic acid based self-assembling nanosystems for CD44 target mediated siRNA delivery to solid tumors. Biomaterials 2013;34:3489–502
  • Iyer AK, Su Y, Feng J, et al. The effect of internalizing human single chain antibody fragment on liposome targeting to epithelioid and sarcomatoid mesothelioma. Biomaterials 2011;32:2605–13
  • Farokhzad OC, Karp JM, Langer R. Nanoparticle-aptamer bioconjugates for cancer targeting. Expert Opin Drug Deliv 2006;3:311–24
  • Rihova B. Receptor-mediated targeted drug or toxin delivery. Adv Drug Deliv Rev 1998;29:273–89
  • Lue N, Ganta S, Hammer DX, et al. Preliminary evaluation of a nanotechnology-based approach for the more effective diagnosis of colon cancers. Nanomedicine 2010;5:1467–79
  • Duncan R. The dawning era of polymer therapeutics. Nat Rev Drug Discov 2003;2:347–60
  • Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 2005;5:161–71
  • Jabr-Milane LS, Van Vlerken LE, Yadav S, et al. Multi-functional nanocarriers to overcome tumor drug resistance. Cancer Treat Rev 2008;34:592–602
  • Veronese FM, Mero A. The impact of PEGylation on biological therapies. BioDrugs 2008;22:315–29
  • Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today 2005;10:1451–8
  • Zhang L, Radovic-Moreno AF, Alexis F, et al. Co-delivery of hydrophobic and hydrophilic drugs from nanoparticle-aptamer bioconjugates. ChemMedChem 2007;2:1268–71
  • Xiong XB, Lavasanifar A. Traceable multifunctional micellar nanocarriers for cancer-targeted co-delivery of MDR-1 siRNA and doxorubicin. ACS Nano 2011;5:5202–13
  • Janib SM, Moses AS, Mackay JA. Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev 2010;62:1052–63
  • Boddapati SV, D’souza GGM, Erdogan S, et al. Organelle-targeted nanocarriers: specific delivery of liposomal ceramide to mitochondria enhances its cytotoxicity in vitro and in vivo. Nano Lett 2008;8:2559–63
  • Ganta S, Devalapally H, Shahiwala A, et al. A review of stimuli-responsive nanocarriers for drug and gene delivery. J Control Release 2008;126:187–204
  • Shen M, Shi X. Dendrimer-based organic/inorganic hybrid nanoparticles in biomedical applications. Nanoscale 2010;2:1596–610
  • Abeylath SC, Ganta S, Iyer AK, et al. Combinatorial-designed multifunctional polymeric nanosystems for tumor-targeted therapeutic delivery. Acc Chem Res 2011;44:1009–17
  • Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2012;2:3–44
  • Salvador-Morales C, Valencia PM, Thakkar AB, et al. Recent developments in multifunctional hybrid nanoparticles: opportunities and challenges in cancer therapy. Front Biosci (Elite Ed) 2012;4:529–45
  • Iyer AK, He J, Amiji MM. Image-guided nanosystems for targeted delivery in cancer therapy. Curr Med Chem 2012;19:3230–40
  • Van Vlerken LE, Duan Z, Little SR, et al. Biodistribution and pharmacokinetic analysis of Paclitaxel and ceramide administered in multifunctional polymer-blend nanoparticles in drug resistant breast cancer model. Mol Pharm 2008;5:516–26
  • Mccarron PA, Marouf WM, Quinn DJ, et al. Antibody targeting of camptothecin-loaded PLGA nanoparticles to tumor cells. Bioconjug Chem 2008;19:1561–69
  • Ahmed N, Fessi H, Elaissari A. Theranostic applications of nanoparticles in cancer. Drug Discov Today 2012;17:928–34
  • Yu MK, Kim D, Lee IH, et al. Image-guided prostate cancer therapy using aptamer-functionalized thermally cross-linked superparamagnetic iron oxide nanoparticles. Small 2011;7:2241–9
  • Koo H, Lee H, Lee S, et al. In vivo tumor diagnosis and photodynamic therapy via tumoral pH-responsive polymeric micelles. Chem Commun (Camb) 2010;46:5668–70
  • Shegokar R, Al Shaal L, Mishra PR. SiRNA delivery: challenges and role of carrier systems. Pharmazie 2011;6:313–18
  • Yadav S, Van Vlerken LE, Little SR, et al. Evaluations of combination MDR-1 gene silencing and paclitaxel administration in biodegradable polymeric nanoparticle formulations to overcome multidrug resistance in cancer cells. Cancer Chemother Pharmacol 2009;63:711–22
  • Hood JD, Bednarski M, Frausto R, et al. Tumor regression by targeted gene delivery to the neovasculature. Science 2002;296:2404–7
  • Yoshizawa T, Hattori Y, Hakoshima M, et al. Folate-linked lipid-based nanoparticles for synthetic siRNA delivery in KB tumor xenografts. Eur J Pharm Biopharm 2008;70:718–25
  • Huh MS, Lee SY, Park S, et al. Tumor-homing glycol chitosan/polyethylenimine nanoparticles for the systemic delivery of siRNA in tumor-bearing mice. J Control Release 2010;144:134–43
  • Medarova Z, Pham W, Farrar C, et al. In vivo imaging of siRNA delivery and silencing in tumors. Nat Med 2007;13:372–7
  • Allavena P, Sica A, Solinas G, et al. The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol 2008;66:1–9
  • Rogers TL, Holen I. Tumour macrophages as potential targets of bisphosphonates. J Transl Med 2011;9:177
  • Jin-Yuan AY, Jeremy J-W Chen, Pan-Chyr Yang. Tumor-associated macrophage: its role in cancer invasion and metastasis. J Cancer Mol 2006;2:101–6
  • Yang J, Zeng Y, Li Y, et al. Intravascular site-specific delivery of a therapeutic antisense for the inhibition of restenosis. Eur J Pharm Sci 2008;35:427–34
  • Sica A, Schioppa T, Mantovani A, et al. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 2006;42:717–27
  • Zeisberger SM, Odermatt B, Marty C, et al. Clodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approach. Br J Cancer 2006;95:272–81
  • Schiffelers RM, Storm G. Liposomal nanomedicines as anticancer therapeutics: beyond targeting tumor cells. Int J Pharm 2008;364:258–64
  • Meng H, Xing G, Blanco E, et al. Gadolinium metallofullerenol nanoparticles inhibit cancer metastasis through matrix metalloproteinase inhibition: imprisoning instead of poisoning cancer cells. Nanomedicine 2012;8:136–46
  • Turk MJ, Waters DJ, Low PS. Folate-conjugated liposomes preferentially target macrophages associated with ovarian carcinoma. Cancer Lett 2004;213:165–72
  • Hallam S, Hagemann T. TAM: a moving clinical target. In: Lawrence T, Hagemann T, eds. Tumour-associated macrophages. Springer Publishers; 2012:63--73
  • Alizadeh D, Zhang L, Hwang J, et al. Tumor-associated macrophages are predominant carriers of cyclodextrin-based nanoparticles into gliomas. Nanomedicine 2010;6:382–90
  • Fujiwara N, Kobayashi K. Macrophages in inflammation. Curr Drug Targets Inflamm Allergy 2005;5:281–6
  • Ross JA, Auger MJ. The Biology of the macrophage. In: Bernard Burke, Lewis CE, eds. The macrophage. New York: Oxford University Press; 2002:16–23
  • Lopes MF, Freire-De-Lima CG, Dosreis GA. The macrophage haunted by cell ghosts: a pathogen grows. Immunol Today 2000;21:489–494
  • Jain S, Amiji M. Macrophage-targeted nanoparticle delivery systems. In: Svenson S, Prud’homme R, eds. Multifunctional nanoparticles for drug delivery applications imaging, targeting, and delivery. New York: Springer; 2012:48–55
  • Hamilton TA. Molecular basis of macrophage activation: from gene expression to phenotypic diversity. In: Burke B, Lewis CE, eds. The macrophage. New York: Oxford University Press; 2002:74–5
  • Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677–86
  • Gordon S. Alternative activation of macrophages. Nat Rev 2003;3:23–35
  • Wilson HM, Barker RN, Erwig LP. Macrophages: promising targets for the treatment of atherosclerosis. Curr Vasc Pharmacol 2009;7:234–43
  • Mosser DM. The many faces of macrophage activation. J Leukoc Biol 2003;73:209–12
  • Owais M, Gupta CM. Targeted drug delivery to macrophages in parasitic infections. Curr Drug Deliv 2005;2:311–18
  • Germann WJ, Stanfield CL, Cannon JG, et al. The immune system. In: Brassert C, ed. Principles of human physiology. San Francisco: Benjamin Cummings Publishing Co.; 2002:708–40
  • Tanner AR, Arthur MJ, Wright R. Macrophage activation, chronic inflammation and gastrointestinal disease. Gut 1984;25:760–83
  • Nie S, Xing Y, Kim GJ, et al. Nanotechnology applications in cancer. Ann Rev Biomed Eng 2007;9:257–88
  • Mukhopadhyay A, Basu SK. Intracellular delivery of drugs to macrophages. Adv Biochem Eng Biotechnol 2003;84:183–209
  • Brown GD, Taylor PR, Reid DM, et al. Dectin-1 is a major beta-glucan receptor on macrophages. J Exp Med 2002;196:407–12
  • Herre J, Gordon S, Brown GD. Dectin-1 and its role in the recognition of beta-glucans by macrophages. Mol Immunol 2004;40:869–76
  • Najjar VA. Tuftsin, a natural activator of phagocyte cells: an overview. Ann NY Acad Sci 1983;419:1–11
  • Fridkin M, Najjar VA. Tuftsin: its chemistry, biology, and clinical potential. Crit Rev Biochem Mol Biol 1989;24:1–40
  • Naor D, Nedvetzki S. CD44 in rheumatoid arthritis. Arthritis Res Ther 2003;5:105–15
  • Vachon E, Martin R, Plumb J, et al. CD44 is a phagocytic receptor. Blood 2006;107:4149–58
  • Ahsan F, Rivas IP, Khan MA, et al. Targeting to macrophages: role of physicochemical properties of particulate carriers – liposomes and microspheres – on the phagocytosis by macrophages. J Control Release 2002;79:29–40
  • Kaur A, Jain S, Tiwary AK. Mannan-coated gelatin nanoparticles for sustained and targeted delivery of didanosine: in vitro and in vivo evaluation. Acta Pharm 2008;58:61–74
  • Jain SK, Gupta Y, Jain A, et al. Mannosylated gelatin nanoparticles bearing an anti-HIV drug didanosine for site-specific delivery. Nanomedicine 2008;4:41–8
  • Schmitt F, Lagopoulos L, Kauper P, et al. Chitosan-based nanogels for selective delivery of photosensitizers to macrophages and improved retention in and therapy of articular joints. J Control Release 2010;144:242–50
  • Ryan SM, Mcmorrow J, Umerska A, et al. An intra-articular salmon calcitonin-based nanocomplex reduces experimental inflammatory arthritis. J Control Release 2013;167:120–9
  • Coco R, Plapied L, Pourcelle V, et al. Drug delivery to inflamed colon by nanoparticles: comparison of different strategies. Int J Pharm 2013;440:3–12
  • Chan JM, Rhee JW, Drum CL, et al. In vivo prevention of arterial restenosis with paclitaxel-encapsulated targeted lipid-polymeric nanoparticles. Proc Natl Acad Sci USA 2011;108:19347–52
  • Markovsky E, Koroukhov N, Golomb G. Additive-free albumin nanoparticles of alendronate for attenuating inflammation through monocyte inhibition. Nanomedicine (Lond) 2007;2:545–53
  • Zhou HF, Yan H, Senpan A, et al. Suppression of inflammation in a mouse model of rheumatoid arthritis using targeted lipase-labile fumagillin prodrug nanoparticles. Biomaterials 2012;33:8632–40
  • Cyrus T, Zhang H, Allen JS, et al. Intramural delivery of rapamycin with alphavbeta3-targeted paramagnetic nanoparticles inhibits stenosis after balloon injury. Arterioscler Thromb Vasc Biol 2008;28:820–6
  • Joner M, Morimoto K, Kasukawa H, et al. Site-specific targeting of nanoparticle prednisolone reduces in-stent restenosis in a rabbit model of established atheroma. Arterioscler Thromb Vasc Biol 2008;28:1960–6
  • Koo OM, Rubinstein I, Onyuksel H. Actively targeted low-dose camptothecin as a safe, long-acting, disease-modifying nanomedicine for rheumatoid arthritis. Pharm Res 2011;28:776–87
  • Kamaly N, Fredman G, Subramanian M, et al. Development and in vivo efficacy of targeted polymeric inflammation-resolving nanoparticles. Proc Natl Acad Sci USA 2013;110:6506--11
  • Thomas TP, Goonewardena SN, Majoros IJ, et al. Folate-targeted nanoparticles show efficacy in the treatment of inflammatory arthritis. Arthritis Rheum 2011;63:2671–80
  • Richards PJ, Williams BD, Williams AS. Suppression of chronic streptococcal cell wall-induced arthritis in Lewis rats by liposomal clodronate. Rheumatology (Oxford) 2001;40:978–87
  • Mccarthy J, O’neill MJ, Bourre L, et al. Gene silencing of TNF-alpha in a murine model of acute colitis using a modified cyclodextrin delivery system. J Control Release 2013;168:28–34
  • Scheinman RI, Trivedi R, Vermillion S, Kompella UB. Functionalized STAT1 siRNA nanoparticles regress rheumatoid arthritis in a mouse model. Nanomedicine (Lond) 2011;6:1669–82
  • Aouadi M, Tesz GJ, Nicoloro SM, et al. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature 2009;458:1180--4
  • Wilson DS, Dalmasso G, Wang L, et al. Orally delivered thioketal nanoparticles loaded with TNF-α--siRNA target inflammation and inhibit gene expression in the intestines. Nat Mater 2010;9:923--8
  • Shi Q, Wang H, Tran C, et al. Hydrodynamic delivery of chitosan-folate-DNA nanoparticles in rats with adjuvant-induced arthritis. J Biomed Biotechnol 2011;2011:148763
  • Attarwala H, Amiji M. Multi-compartmental nanoparticles-in-emulsion formulation for macrophage-specific anti-inflammatory gene delivery. Pharm Res 2012;29:1637–49
  • Beilvert F, Tissot A, Langelot M, et al. DNA/amphiphilic block copolymer nanospheres reduce asthmatic response in a mouse model of allergic asthma. Hum Gene Ther 2012;23:597–608
  • Te Boekhorst BC, Jensen LB, Colombo S, et al. MRI-assessed therapeutic effects of locally administered PLGA nanoparticles loaded with anti-inflammatory siRNA in a murine arthritis model. J Control Release 2012;16:772–80
  • Kumar M, Kong X, Behera AK, et al. Chitosan IFN-gamma-pDNA nanoparticle (CIN) therapy for allergic asthma. Genet Vaccines Ther 2003;1:3
  • Laroui H, Theiss AL, Yan Y, et al. Functional TNFα gene silencing mediated by polyethyleneimine/TNFalpha siRNA nanocomplexes in inflamed colon. Biomaterials 2011;32:1218–28
  • Chaudhury A, Das S, Bunte RM, et al. Potent therapeutic activity of folate receptor-targeted liposomal carboplatin in the localized treatment of intraperitoneally grown human ovarian tumor xenograft. Int J Nanomed 2012;7:739–51
  • Meng H, Mai WX, Zhang H, et al. Codelivery of an optimal drug/siRNA combination using mesoporous silica nanoparticles to overcome drug resistance in breast cancer in vitro and in vivo. ACS Nano 2013;7:994--1005
  • Roy A, Singh MS, Upadhyay P, Bhaskar S. Nanoparticle mediated co-delivery of paclitaxel and a TLR-4 agonist results in tumor regression and enhanced immune response in the tumor microenvironment of a mouse model. Int J Pharm 2013;445:171--80
  • Gao H, Qian J, Cao S, et al. Precise glioma targeting of and penetration by aptamer and peptide dual-functioned nanoparticles. Biomaterials 2012;33:5115–23
  • Sahoo SK, Ma W, Labhasetwar V. Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer. Int J Cancer 2004;112:335–40
  • Xin H, Jiang X, Gu J, et al. Angiopep-conjugated poly(ethylene glycol)-co-poly(epsilon-caprolactone) nanoparticles as dual-targeting drug delivery system for brain glioma. Biomaterials 2011;32:4293–305
  • Hood JD, Bednarski M, Frausto R, et al. Tumor regression by targeted gene delivery to the neovasculature. Science 2002;296:2404–7
  • Yoo HS, Lee KH, Oh JE, Park TG. In vitro and in vivo anti-tumor activities of nanoparticles based on doxorubicin-PLGA conjugates. J Control Release 2000;68:419–31
  • Mitra S, Gaur U, Ghosh PC, et al. Tumour targeted delivery of encapsulated dextran-doxorubicin conjugate using chitosan nanoparticles as carrier. J Control Release 2001;74:317–23
  • Williams J, Lansdown R, Sweitzer R, et al. Nanoparticle drug delivery system for intravenous delivery of topoisomerase inhibitors. J Control Release 2003;91:167–72
  • Jiang G, Park K, Kim J, et al. Target specific intracellular delivery of siRNA/PEI-HA complex by receptor mediated endocytosis. Mol Pharm 2009;6:727–37
  • Zhang J, Tang C, Yin C. Galactosylated trimethyl chitosan-cysteine nanoparticles loaded with Map4k4 siRNA for targeting activated macrophages. Biomaterials 2013;34:3667--77
  • Yu W, Liu C, Liu Y, et al. Mannan-modified solid lipid nanoparticles for targeted gene delivery to alveolar macrophages. Pharm Res 2010;27:1584–96
  • Nasti TH, Khan MA, Owais M. Enhanced efficacy of pH-sensitive nystatin liposomes against Cryptococcus neoformans in murine model. J Antimicrob Chemother 2006;57:349–52
  • Owais M, Gupta CM. Targeted drug delivery to macrophages in parasitic infections. Curr Drug Deliv 2005;2:311–18
  • He C, Yin L, Tang C, Yin C. Multifunctional polymeric nanoparticles for oral delivery of TNF-alpha siRNA to macrophages. Biomaterials 2013;34:2843–54
  • Chono S, Tauchi Y, Deguchi Y, Morimoto K. Efficient drug delivery to atherosclerotic lesions and the antiatherosclerotic effect by dexamethasone incorporated into liposomes in atherogenic mice. J Drug Target 2005;13:267–76
  • Chono S, Tanino T, Seki T, Morimoto K. Uptake characteristics of liposomes by rat alveolar macrophages: influence of particle size and surface mannose modification. J Pharm Pharmacol 2007;59:75–80
  • Prajapati VK, Awasthi K, Gautam S, et al. Targeted killing of Leishmania donovani in vivo and in vitro with amphotericin B attached to functionalized carbon nanotubes. J Antimicrob Chemother 2011;66:874–9
  • Skidan IN, Gel’perina SE, Severin SE, Guliaev AE. Enhanced activity of rifampicin loaded with polybutyl cyanoacrylate nanoparticles in relation to intracellularly localized bacteria. Antibiot Khimioter 2003;48:23–6
  • Mukherjee S, Das L, Kole L, et al. Targeting of parasite-specific immunoliposome-encapsulated doxorubicin in the treatment of experimental visceral leishmaniasis. J Infect Dis 2004;189:1024–34
  • Hrkach J, Von Hoff D, Mukkaram Ali M, et al. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile. Sci Transl Med 2012;4:128–39
  • Ganesh S, Iyer AK, Weiler J, et al. Combination of siRNA-directed gene silencing with cisplatin reverses drug resistance in human non-small cell lung cancer. Mol Ther Nucleic Acids 2013;2:e110

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.