8,028
Views
98
CrossRef citations to date
0
Altmetric
Review Article

Nasal-nanotechnology: revolution for efficient therapeutics delivery

, &
Pages 671-683 | Received 01 Apr 2014, Accepted 29 Apr 2014, Published online: 05 Jun 2014

References

  • Aggarwal R, Cardozo A, Homer JJ. (2004). The assessment of topical nasal drug distribution. Clin Otolaryngol Allied Sci 29:201–5
  • Al-Ghananeem AM, Malkawi AH, Crooks PA. (2011). Bioavailability of Delta(9)-tetrahydrocannabinol following intranasal administration of a mucoadhesive gel spray delivery system in conscious rabbits. Drug Dev Ind Pharm 37:329–34
  • Al-Ghananeem AM, Saeed H, Florence R, et al. (2010). Intranasal drug delivery of didanosine-loaded chitosan nanoparticles for brain targeting; an attractive route against infections caused by AIDS viruses. J Drug Target 18:381–8
  • Ali J, Ali M, Baboota S, et al. (2010). Potential of nanoparticulate drug delivery systems by intranasal administration. Curr Pharm Des 16:1644–53
  • Almeida AJ, Alpar HO. (1996). Nasal delivery of vaccines. J Drug Target 3:455–67
  • Alsarra IA, Hamed AY, Mahrous GM, et al. (2009). Mucoadhesive polymeric hydrogels for nasal delivery of acyclovir. Drug Dev Ind Pharm 35:352–62
  • Amidi M, Romeijn SG, Verhoef JC, et al. (2007). N-trimethyl chitosan (TMC) nanoparticles loaded with influenza subunit antigen for intranasal vaccination: biological properties and immunogenicity in a mouse model. Vaccine 25:144–53
  • Arora P, Sharma S, Garg S. (2002). Permeability issues in nasal drug delivery. Drug Discov Today 7:967–75
  • Bacon A, Makin J, Sizer PJ, et al. (2000). Carbohydrate biopolymers enhance antibody responses to mucosally delivered vaccine antigens. Infect Immun 68:5764–70
  • Bahadur S, Pathak K. (2012a). Buffered nanoemulsion for nose to brain delivery of ziprasidone hydrochloride: preformulation and pharmacodynamic evaluation. Curr Drug Deliv 9:596–607
  • Bahadur S, Pathak K. (2012b). Physicochemical and physiological considerations for efficient nose-to-brain targeting. Expert Opin Drug Deliv 9:19–31
  • Balle VH, Illum P, Meistrup-Larsen U, Olesen KF. (1979). Disodium cromoglycate in the treatment of allergic conditions of the nose. Ugeskr Laeger 141:1906–8
  • Belgamwar VS, Patel HS, Joshi AS, et al. (2011). Design and development of nasal mucoadhesive microspheres containing tramadol HCl for CNS targeting. Drug Deliv 18:353–60
  • Bielinska AU, Janczak KW, Landers JJ, et al. (2007). Mucosal immunization with a novel nanoemulsion-based recombinant anthrax protective antigen vaccine protects against Bacillus anthracis spore challenge. Infect Immun 75:4020–9
  • Bijani C, Arnarez C, Brasselet S, et al. (2012). Stability and structure of protein-lipoamino acid colloidal particles: toward nasal delivery of pharmaceutically active proteins. Langmuir 28:5783–94
  • Bitter C, Suter-Zimmermann K, Surber C. (2011). Nasal drug delivery in humans. Curr Probl Dermatol 40:20–35
  • Boek WM, Romeijn SG, Graamans K, et al. (1999). Validation of animal experiments on ciliary function in vitro. II. The influence of absorption enhancers, preservatives and physiologic saline. Acta Otolaryngol 119:98–101
  • Bommer R. (1999). Latest advances in nasal drug-delivery technology. Med Device Technol 10:22–8
  • Boogaard R, de Jongste JC, Merkus PJ. (2007). Pharmacotherapy of impaired mucociliary clearance in non-CF pediatric lung disease. A review of the literature. Pediatr Pulmonol 42:989–1001
  • Bragagni M, Mennini N, Ghelardini C, Mura P. (2012). Development and characterization of niosomal formulations of doxorubicin aimed at brain targeting. J Pharm Pharm Sci 15:184–96
  • Brave A, Hallengard D, Schroder U, et al. (2008). Intranasal immunization of young mice with a multigene HIV-1 vaccine in combination with the N3 adjuvant induces mucosal and systemic immune responses. Vaccine 26:5075–8
  • Brooking J, Davis SS, Illum L. (2001). Transport of nanoparticles across the rat nasal mucosa. J Drug Target 9:267–79
  • Caban S, Capan Y, Couvreur P, Dalkara T. (2012). Preparation and characterization of biocompatible chitosan nanoparticles for targeted brain delivery of peptides. Methods Mol Biol 846:321–32
  • Cai Z, Song X, Sun F, et al. (2011). Formulation and evaluation of in situ gelling systems for intranasal administration of gastrodin. AAPS PharmSciTech 12:1102–9
  • Cao SL, Ren XW, Zhang QZ, et al. (2009). In situ gel based on gellan gum as new carrier for nasal administration of mometasone furoate. Int J Pharm 365:109–15
  • Cerchiara T, Luppi B, Chidichimo G, et al. (2005). Chitosan and poly(methyl vinyl ether-co-maleic anhydride) microparticles as nasal sustained delivery systems. Eur J Pharm Biopharm 61:195–200
  • Chandler SG, Thomas NW, Illum L. (1994). Nasal absorption in the rat. III. Effect of lysophospholipids on insulin absorption and nasal histology. Pharm Res 11:1623–30
  • Charlton ST, Davis SS, Illum L. (2007). Nasal administration of an angiotensin antagonist in the rat model: effect of bioadhesive formulations on the distribution of drugs to the systemic and central nervous systems. Int J Pharm 338:94–103
  • Chen J, Zhang C, Liu Q, et al. (2012). Solanum tuberosum lectin-conjugated PLGA nanoparticles for nose-to-brain delivery: in vivo and in vitro evaluations. J Drug Target 20:174–84
  • Chen W, Zhan C, Gu B, et al. (2011). Targeted brain delivery of itraconazole via RVG29 anchored nanoparticles. J Drug Target 19:228–34
  • Chiou CJ, Tseng LP, Deng MC, et al. (2009). Mucoadhesive liposomes for intranasal immunization with an avian influenza virus vaccine in chickens. Biomaterials 30:5862–8
  • Chung SW, Hil-lal TA, Byun Y. (2012). Strategies for non-invasive delivery of biologics. J Drug Target 20:481–501
  • Cokcaliskan C, Ozyoruk F, Gursoy RN, et al. (2014). Chitosan-based systems for intranasal immunization against foot-and-mouth disease. Pharm Dev Technol 19:181–8
  • Costantino HR, Illum L, Brandt G, et al. (2007). Intranasal delivery: physicochemical and therapeutic aspects. Int J Pharm 337:1–24
  • Critchley H, Davis SS, Farraj NF, Illum L. (1994). Nasal absorption of desmopressin in rats and sheep. Effect of a bioadhesive microsphere delivery system. J Pharm Pharmacol 46:651–6
  • Cros CD, Toth I, Blanchfield JT. (2011). Lipophilic derivatives of leu-enkephalinamide: in vitro permeability, stability and in vivo nasal delivery. Bioorg Med Chem 19:1528–34
  • Csaba N, Garcia-Fuentes M, Alonso MJ. (2009). Nanoparticles for nasal vaccination. Adv Drug Deliv Rev 61:140–57
  • Davis SS, Illum L. (2003). Absorption enhancers for nasal drug delivery. Clin Pharmacokinet 42:1107–28
  • Deurloo MJ, Hermens WA, Romeyn SG, et al. (1989). Absorption enhancement of intranasally administered insulin by sodium taurodihydrofusidate (STDHF) in rabbits and rats. Pharm Res 6:853–6
  • Djupesland PG, Docekal P. (2010). Intranasal sumatriptan powder delivered by a novel breath-actuated bi-directional device for the acute treatment of migraine: a randomised, placebo-controlled study. Cephalalgia 30:933–42
  • Djupesland PG, Skretting A, Winderen M, Holand T. (2004). Bi-directional nasal delivery of aerosols can prevent lung deposition. J Aerosol Med 17:249–59
  • Douglas SJ, Davis SS, Illum L. (1987). Nanoparticles in drug delivery. Crit Rev Ther Drug Carrier Syst 3:233–61
  • Duchateau GS. (1987). Studies on nasal drug delivery. Pharm Weekbl Sci 9:326–8
  • Duchateau GS, Zuidema J, Merkus FW. (1986a). Bioavailability of propranolol after oral, sublingual, and intranasal administration. Pharm Res 3:108–11
  • Duchateau GS, Zuidema J, Merkus FW. (1986b). The in vitro and in vivo effect of a new non-halogenated corticosteroid – budesonide – aerosol on human ciliary epithelial function. Allergy 41:260–5
  • Duquesnoy C, Mamet JP, Sumner D, Fuseau E. (1998). Comparative clinical pharmacokinetics of single doses of sumatriptan following subcutaneous, oral, rectal and intranasal administration. Eur J Pharm Sci 6:99–104
  • Elshafeey AH, Bendas ER, Mohamed OH. (2009). Intranasal microemulsion of sildenafil citrate: in vitro evaluation and in vivo pharmacokinetic study in rabbits. AAPS PharmSciTech 10:361–7
  • Eskandari S, Varshosaz J, Minaiyan M, Tabbakhian M. (2011). Brain delivery of valproic acid via intranasal administration of nanostructured lipid carriers: in vivo pharmacodynamic studies using rat electroshock model. Int J Nanomedicine 6:363–71
  • Eyles JE, Sharp GJ, Williamson ED, et al. (1998a). Intra nasal administration of poly-lactic acid microsphere co-encapsulated Yersinia pestis subunits confers protection from pneumonic plague in the mouse. Vaccine 16:698–707
  • Eyles JE, Spiers ID, Williamson ED, Alpar HO. (1998b). Analysis of local and systemic immunological responses after intra-tracheal, intra-nasal and intra-muscular administration of microsphere co-encapsulated Yersinia pestis sub-unit vaccines. Vaccine 16:2000–9
  • Fazil M, Md S, Haque S, et al. (2012). Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur J Pharm Sci 47:6–15
  • Gaillard PJ, Appeldoorn CC, Rip J, et al. (2012). Enhanced brain delivery of liposomal methylprednisolone improved therapeutic efficacy in a model of neuroinflammation. J Control Release 164:364–9
  • Gavini E, Rassu G, Haukvik T, et al. (2009). Mucoadhesive microspheres for nasal administration of cyclodextrins. J Drug Target 17:168–79
  • Gavini E, Rassu G, Muzzarelli C, et al. (2008). Spray-dried microspheres based on methylpyrrolidinone chitosan as new carrier for nasal administration of metoclopramide. Eur J Pharm Biopharm 68:245–52
  • Gizurarson S. (2012). Anatomical and histological factors affecting intranasal drug and vaccine delivery. Curr Drug Deliv 9:566–82
  • Gizurarson S, Rasmussen SN, Larsen F. (1991). Pharmacokinetics of intranasal drug administration: the influence of some biological factors. J Pharm Sci 80:505–6
  • Greimel A, Bernkop-Schnurch A, Del Curto MD, D'Antonio M. (2007). Transport characteristics of a beta sheet breaker peptide across excised bovine nasal mucosa. Drug Dev Ind Pharm 33:71–7
  • Gungor S, Okyar A, Erturk-Toker S, et al. (2010a). Ondansetron-loaded biodegradable microspheres as a nasal sustained delivery system: in vitro/in vivo studies. Pharm Dev Technol 15:258–65
  • Gungor S, Okyar A, Erturk-Toker S, et al. (2010b). Ondansetron-loaded chitosan microspheres for nasal antiemetic drug delivery: an alternative approach to oral and parenteral routes. Drug Dev Ind Pharm 36:806–13
  • Hafner A, Filipovic-Grcic J, Voinovich D, Jalsenjak I. (2007). Development and in vitro characterization of chitosan-based microspheres for nasal delivery of promethazine. Drug Dev Ind Pharm 33:427–36
  • Hall MA, Stroop SD, Hu MC, et al. (2004). Intranasal immunization with multivalent group A streptococcal vaccines protects mice against intranasal challenge infections. Infect Immun 72:2507–12
  • Harikarnpakdee S, Lipipun V, Sutanthavibul N, Ritthidej GC. (2006). Spray-dried mucoadhesive microspheres: preparation and transport through nasal cell monolayer. AAPS PharmSciTech 7:E79–88
  • Hasegawa A, Fu Y, Koyama K. (2002). Nasal immunization with diphtheria toxoid conjugated-CD52 core peptide induced specific antibody production in genital tract of female mice. Am J Reprod Immunol 48:305–11
  • He P, Davis SS, Illum L. (1999). Sustained release chitosan microspheres prepared by novel spray drying methods. J Microencapsul 16:343–55
  • Hehar SS, Mason JD, Stephen AB, et al. (1999). Twenty-four hour ambulatory nasal pH monitoring. Clin Otolaryngol Allied Sci 24:24–5
  • Hinchcliffe M, Illum L. (1999). Intranasal insulin delivery and therapy. Adv Drug Deliv Rev 35:199–234
  • Hu KL, Mei N, Feng L, Jiang XG. (2009). Hydrophilic nasal gel of lidocaine hydrochloride. 2nd communication: improved bioavailability and brain delivery in rats with low ciliotoxicity. Arzneimittelforschung 59:635–40
  • Hughes R, Watterson J, Dickens C, et al. (2008). Development of a nasal cast model to test medicinal nasal devices. Proc Inst Mech Eng H 222:1013–22
  • Illum L. (1991). The nasal delivery of peptides and proteins. Trends Biotechnol 9:284–9
  • Illum L. (2000). Transport of drugs from the nasal cavity to the central nervous system. Eur J Pharm Sci 11:1–18
  • Illum L. (2002). Nasal drug delivery: new developments and strategies. Drug Discov Today 7:1184–9
  • Illum L. (2003). Nasal drug delivery--possibilities, problems and solutions. J Control Release 87:187–98
  • Illum L. (2004). Is nose-to-brain transport of drugs in man a reality? J Pharm Pharmacol 56:3–17
  • Illum L. (2006). Nasal clearance in health and disease. J Aerosol Med 19:92–9
  • Illum L. (2007). Nanoparticulate systems for nasal delivery of drugs: a real improvement over simple systems? J Pharm Sci 96:473–83
  • Illum L. (2012). Nasal drug delivery – recent developments and future prospects. J Control Release 161:254–63
  • Illum L, Davis SS. (1991). Drug delivery. Curr Opin Biotechnol 2:254–9
  • Illum L, Davis SS. (2001). Nasal vaccination: a non-invasive vaccine delivery method that holds great promise for the future. Adv Drug Deliv Rev 51:1–3
  • Illum L, Jabbal-Gill I, Hinchcliffe M, et al. (2001). Chitosan as a novel nasal delivery system for vaccines. Adv Drug Deliv Rev 51:81–96
  • Illum L, Watts P, Fisher AN, et al. (2002). Intranasal delivery of morphine. J Pharmacol Exp Ther 301:391–400
  • Illum P, Balle V. (1978). Immunoglobulins in nasal secretions and nasal mucosa in perennial rhinitis. Acta Otolaryngol 86:135–41
  • Iqbal M, Lin W, Jabbal-Gill I, et al. (2003). Nasal delivery of chitosan-DNA plasmid expressing epitopes of respiratory syncytial virus (RSV) induces protective CTL responses in BALB/c mice. Vaccine 21:1478–85
  • Jain N, Akhter S, Jain GK, et al. (2011). Antiepileptic intranasal Amiloride loaded mucoadhesive nanoemulsion: development and safety assessment. J Biomed Nanotechnol 7:142–3
  • Jain R, Nabar S, Dandekar P, et al. (2010a). Formulation and evaluation of novel micellar nanocarrier for nasal delivery of sumatriptan. Nanomedicine (Lond) 5:575–87
  • Jain R, Nabar S, Dandekar P, Patravale V. (2010b). Micellar nanocarriers: potential nose-to-brain delivery of zolmitriptan as novel migraine therapy. Pharm Res 27:655–64
  • Jayachandra BR, Dayal PP, Pawar K, Singh M. (2011). Nose-to-brain transport of melatonin from polymer gel suspensions: a microdialysis study in rats. J Drug Target 19:731–40
  • Jones N. (2001). The nose and paranasal sinuses physiology and anatomy. Adv Drug Deliv Rev 51:5–19
  • Jones NS, Quraishi S, Mason JD. (1997). The nasal delivery of systemic drugs. Int J Clin Pract 51:308–11
  • Joshi AS, Patel HS, Belgamwar VS, et al. (2012). Solid lipid nanoparticles of ondansetron HCl for intranasal delivery: development, optimization and evaluation. J Mater Sci Mater Med 23:2163–75
  • Jug M, Becirevic-Lacan M. (2008). Development of a cyclodextrin-based nasal delivery system for lorazepam. Drug Dev Ind Pharm 34:817–26
  • Kakkar V, Mishra AK, Chuttani K, et al. (2011). Delivery of sesamol-loaded solid lipid nanoparticles to the brain for menopause-related emotional and cognitive central nervous system derangements. Rejuvenation Res 14:597–604
  • Kang ML, Cho CS, Yoo HS. (2009). Application of chitosan microspheres for nasal delivery of vaccines. Biotechnol Adv 27:857–65
  • Kantar A. (2010). Targeting the nasal cavity in rhinitis and sinusitis. Int J Immunopathol Pharmacol 23:67–9
  • Karasulu HY, Sanal ZE, Sozer S, et al. (2008). Permeation studies of indomethacin from different emulsions for nasal delivery and their possible anti-inflammatory effects. AAPS PharmSciTech 9:342–8
  • Kim TK, Kang W, Chun IK, et al. (2009). Pharmacokinetic evaluation and modeling of formulated levodopa intranasal delivery systems. Eur J Pharm Sci 38:525–32
  • Kimbell JS, Segal RA, Asgharian B, et al. (2007). Characterization of deposition from nasal spray devices using a computational fluid dynamics model of the human nasal passages. J Aerosol Med 20:59–74
  • Knuschke T, Sokolova V, Rotan O, et al. (2013). Immunization with biodegradable nanoparticles efficiently induces cellular immunity and protects against influenza virus infection. J Immunol 190:6221–9
  • Kumar A, Sharma P, Chaturvedi A, et al. (2009). Formulation development of sertraline hydrochloride microemulsion for intranasal delivery. Int J ChemTech Res 1:941–7
  • Kumar M, Misra A, Babbar AK, et al. (2008). Intranasal nanoemulsion based brain targeting drug delivery system of risperidone. Int J Pharm 358:285–91
  • Kumar R, Dhanawat M, Kumar S, et al. (2014). Carbon nanotubes: a potential concept for drug delivery applications. Recent Pat Drug Deliv Formul 8:12–26
  • Kundoor V, Dalby RN. (2010). Assessment of nasal spray deposition pattern in a silicone human nose model using a color-based method. Pharm Res 27:30–6
  • Kundu P, Mohanty C, Sahoo SK. (2012). Antiglioma activity of curcumin-loaded lipid nanoparticles and its enhanced bioavailability in brain tissue for effective glioblastoma therapy. Acta Biomater 8:2670–87
  • Laurie E, Hopkins ES, Patchin PC, et al. (2014). Nose-to-brain transport of aerosolized quantum dots following acute exposure. Nanotoxicology 8:885–93
  • Lee KR, Maeng HJ, Chae JB, et al. (2010). Lack of a primary physicochemical determinant in the direct transport of drugs to the brain after nasal administration in rats: potential involvement of transporters in the pathway. Drug Metab Pharmacokinet 25:430–41
  • Lemoine D, Deschuyteneer M, Hogge F, Preat V. (1999). Intranasal immunization against influenza virus using polymeric particles. J Biomater Sci Polym Ed 10:805–25
  • Lifeng Q, Xiaohu G. (2008). Emerging application of quantum dots for drug delivery and therapy. Expert Opin Drug Deliv 5:263–7
  • Lipworth BJ, Jackson CM. (2000). Safety of inhaled and intranasal corticosteroids: lessons for the new millennium. Drug Saf 23:11–33
  • Litvyakova LI, Baraniuk JN. (2001). Nasal provocation testing: a review. Ann Allergy Asthma Immunol 86:355–64
  • Liu Q, Shen Y, Chen J, et al. (2012). Nose-to-brain transport pathways of wheat germ agglutinin conjugated PEG-PLA nanoparticles. Pharm Res 29:546–58
  • Luppi B, Bigucci F, Corace G, et al. (2011). Albumin nanoparticles carrying cyclodextrins for nasal delivery of the anti-Alzheimer drug tacrine. Eur J Pharm Sci 44:559–65
  • Mahajan HS, Tyagi V, Lohiya G, Nerkar P. (2012). Thermally reversible xyloglucan gels as vehicles for nasal drug delivery. Drug Deliv 19:270–6
  • Majithiya RJ, Ghosh PK, Umrethia ML, Murthy RS. (2006). Thermoreversible-mucoadhesive gel for nasal delivery of sumatriptan. AAPS PharmSciTech 7:E80–6
  • Makidon PE, Nigavekar SS, Bielinska AU, et al. (2010). Characterization of stability and nasal delivery systems for immunization with nanoemulsion-based vaccines. J Aerosol Med Pulm Drug Deliv 23:77–89
  • Malerba F, Paoletti F, Capsoni S, Cattaneo A. (2011). Intranasal delivery of therapeutic proteins for neurological diseases. Expert Opin Drug Deliv 8:1277–96
  • Marazuela EG, Rodriguez R, Fernandez-Garcia H, et al. (2008). Intranasal immunization with a dominant T-cell epitope peptide of a major allergen of olive pollen prevents mice from sensitization to the whole allergen. Mol Immunol 45:438–45
  • Marttin E, Romeijn SG, Verhoef JC, Merkus FW. (1997). Nasal absorption of dihydroergotamine from liquid and powder formulations in rabbits. J Pharm Sci 86:802–7
  • Marttin E, Verhoef JC, Merkus FW. (1998). Efficacy, safety and mechanism of cyclodextrins as absorption enhancers in nasal delivery of peptide and protein drugs. J Drug Target 6:17–36
  • Mason JD, Aspden TJ, Adler J, et al. (1995). Measurement of nasal mucociliary transport rates on the isolated human inferior turbinate. Clin Otolaryngol Allied Sci 20:530–5
  • McDonough JA, Persyn JT, Nino JA, et al. (2007). Microcapsule-gel formulation of promethazine HCl for controlled nasal delivery: a motion sickness medication. J Microencapsul 24:109–16
  • McNeela EA, Jabbal-Gill I, Illum L, et al. (2004). Intranasal immunization with genetically detoxified diphtheria toxin induces T cell responses in humans: enhancement of Th2 responses and toxin-neutralizing antibodies by formulation with chitosan. Vaccine 22:909–14
  • McNeela EA, O'Connor D, Jabbal-Gill I, et al. (2000). A mucosal vaccine against diphtheria: formulation of cross reacting material (CRM(197)) of diphtheria toxin with chitosan enhances local and systemic antibody and Th2 responses following nasal delivery. Vaccine 19:1188–98
  • Merkus FW, Schusler-van Hees MT. (1992). Influence of levocabastine suspension on ciliary beat frequency and mucociliary clearance. Allergy 47:230–3
  • Merkus FW, van den Berg MP. (2007). Can nasal drug delivery bypass the blood-brain barrier?: questioning the direct transport theory. Drugs R D 8:133–44
  • Merkus FW, Verhoef JC, Marttin E, et al. (1999a). Cyclodextrins in nasal drug delivery. Adv Drug Deliv Rev 36:41–57
  • Merkus FW, Verhoef JC, Romeijn SG, Schipper NG. (1991a). Absorption enhancing effect of cyclodextrins on intranasally administered insulin in rats. Pharm Res 8:588–92
  • Merkus FW, Verhoef JC, Romeijn SG, Schipper NG. (1991b). Interspecies differences in the nasal absorption of insulin. Pharm Res 8:1343 (PMID: 1796056)
  • Merkus FW, Verhoef JC, Schipper NG, Marttin E. (1998). Nasal mucociliary clearance as a factor in nasal drug delivery. Adv Drug Deliv Rev 29:13–38
  • Merkus P, Guchelaar HJ, Bosch DA, Merkus FW. (2003). Direct access of drugs to the human brain after intranasal drug administration? Neurology 60:1669–71
  • Merkus P, Romeijn SG, Verhoef JC, et al. (2001). Classification of cilio-inhibiting effects of nasal drugs. Laryngoscope 111:595–602
  • Merkus PJ, de Jongste JC. (2006). Inhaled corticosteroids and long term outcome in adults with asthma. Thorax 61:1011 (PMCID: PMC2121169)
  • Merkus PJ, Hazelzet JA, de Jongste JC. (1999b). Inhalation corticosteroids and the growth of asthmatic children. Ned Tijdschr Geneeskd 143:2547 (PMID: 10627761)
  • Mistry A, Glud SZ, Kjems J, et al. (2009a). Effect of physicochemical properties on intranasal nanoparticle transit into murine olfactory epithelium. J Drug Target 17:543–52
  • Mistry A, Stolnik S, Illum L. (2009b). Nanoparticles for direct nose-to-brain delivery of drugs. Int J Pharm 379:146–57
  • Muramatsu K, Maitani Y, Takayama K, Nagai T. (1999). The relationship between the rigidity of the liposomal membrane and the absorption of insulin after nasal administration of liposomes modified with an enhancer containing insulin in rabbits. Drug Dev Ind Pharm 25:1099–105
  • Myc A, Kukowska-Latallo JF, Bielinska AU, et al. (2003). Development of immune response that protects mice from viral pneumonitis after a single intranasal immunization with influenza A virus and nanoemulsion. Vaccine 21:3801–14
  • Nakamura K, Maitani Y, Lowman AM, et al. (1999). Uptake and release of budesonide from mucoadhesive, pH-sensitive copolymers and their application to nasal delivery. J Control Release 61:329–35
  • Nazar H, Fatouros DG, van der Merwe SM, et al. (2011). Thermosensitive hydrogels for nasal drug delivery: the formulation and characterisation of systems based on N-trimethyl chitosan chloride. Eur J Pharm Biopharm 77:225–32
  • Ohwaki T, Ando H, Watanabe S, Miyake Y. (1985). Effects of dose, pH, and osmolarity on nasal absorption of secretin in rats. J Pharm Sci 74:550–2
  • Ozsoy Y, Gungor S, Cevher E. (2009). Nasal delivery of high molecular weight drugs. Molecules 14:3754–79
  • Patel GB, Ponce A, Zhou H, Chen W. (2008). Structural characterization of archaeal lipid mucosal vaccine adjuvant and delivery (AMVAD) formulations prepared by different protocols and their efficacy upon intranasal immunization of mice. J Liposome Res 18:127–43
  • Patel S, Chavhan S, Soni H, et al. (2011). Brain targeting of risperidone-loaded solid lipid nanoparticles by intranasal route. J Drug Target 19:468–74
  • Patil SB, Kaul A, Babbar A, et al. (2012). In vivo evaluation of alginate microspheres of carvedilol for nasal delivery. J Biomed Mater Res B Appl Biomater 100:249–55
  • Patil SB, Sawant KK. (2011). Chitosan microspheres as a delivery system for nasal insufflation. Colloids Surf B Biointerfaces 84:384–9
  • Pires A, Fortuna A, Alves G, Falcao A. (2009). Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci 12:288–311
  • Pisal PB, Patil SS, Pokharkar VB. (2012). Rheological investigation and its correlation with permeability coefficient of drug loaded carbopol gel: influence of absorption enhancers. Drug Dev Ind Pharm 39:593–9
  • Pisal S, Shelke V, Mahadik K, Kadam S. (2004a). Effect of organogel components on in vitro nasal delivery of propranolol hydrochloride. AAPS PharmSciTech 5:92–100
  • Pisal SS, Paradkar AR, Mahadik KR, Kadam SS. (2004b). Pluronic gels for nasal delivery of Vitamin B12. Part I: preformulation study. Int J Pharm 270:37–45
  • Quraishi MS, Jones NS, Mason JD. (1997). The nasal delivery of drugs. Clin Otolaryngol Allied Sci 22:289–301
  • Rajadhyaksha M, Boyden T, Liras J, et al. (2011). Current advances in delivery of biotherapeutics across the blood-brain barrier. Curr Drug Discov Technol 8:87–101
  • Read RC, Naylor SC, Potter CW, et al. (2005). Effective nasal influenza vaccine delivery using chitosan. Vaccine 23:4367–74
  • Ruan Y, Yao L, Zhang B, et al. (2011). Antinociceptive properties of nasal delivery of neurotoxin-loaded nanoparticles coated with polysorbate-80. Peptides 32:1526–9
  • Salama HA, Mahmoud AA, Kamel AO, et al. (2012). Brain delivery of olanzapine by intranasal administration of transfersomal vesicles. J Liposome Res 22:336–45
  • Scheerlinck JP, Gekas S, Yen HH, et al. (2006). Local immune responses following nasal delivery of an adjuvanted influenza vaccine. Vaccine 24:3929–36
  • Schipper NG, Romeijn SG, Verhoef J, Merkus FW. (1994). Hypocalcemic effect of salmon calcitonin following single and repeated nasal and intravenous administration in young rabbits. Calcif Tissue Int 54:50–5
  • Schipper NG, Verhoef JC, Merkus FW. (1991). The nasal mucociliary clearance: relevance to nasal drug delivery. Pharm Res 8:807–14
  • Seju U, Kumar A, Sawant KK. (2011). Development and evaluation of olanzapine-loaded PLGA nanoparticles for nose-to-brain delivery: in vitro and in vivo studies. Acta Biomater 7:4169–76
  • Shahiwala A, Misra A. (2004). Nasal delivery of levonorgestrel for contraception: an experimental study in rats. Fertil Steril 81:893–8
  • Shahiwala A, Misra A. (2006). Preliminary investigation of the nasal delivery of liposomal leuprorelin acetate for contraception in rats. J Pharm Pharmacol 58:19–26
  • Slomkowski S, Gosecki M. (2011). Progress in nanoparticulate systems for peptide, proteins and nucleic acid drug delivery. Curr Pharm Biotechnol 12:1823–39
  • Slot WB, Merkus FW, Van Deventer SJ, Tytgat GN. (1997). Normalization of plasma vitamin B12 concentration by intranasal hydroxocobalamin in vitamin B12-deficient patients. Gastroenterology 113:430–3
  • Smith JH, Papania M, Knaus D, et al. (2012). Nebulized live-attenuated influenza vaccine provides protection in ferrets at a reduced dose. Vaccine 30:3026–33
  • Soane RJ, Carney AS, Jones NS, et al. (2001). The effect of the nasal cycle on mucociliary clearance. Clin Otolaryngol Allied Sci 26:9–15
  • Suman JD. (2003). Nasal drug delivery. Expert Opin Biol Ther 3:519–23
  • Tafaghodi M, Abolghasem Sajadi TS, Jaafari MR, et al. (2004). Evaluation of the clearance characteristics of various microspheres in the human nose by gamma-scintigraphy. Int J Pharm 280:125–35
  • Tafaghodi M, Jaafari MR, Tabassi SA. (2008). Nasal immunization studies by cationic, fusogenic and cationic-fusogenic liposomes encapsulated with tetanus toxoid. Curr Drug Deliv 5:108–13
  • Tas C, Ozkan CK, Savaser A, et al. (2006). Nasal absorption of metoclopramide from different Carbopol 981 based formulations: in vitro, ex vivo and in vivo evaluation. Eur J Pharm Biopharm 64:246–54
  • Teijeiro-Osorio D, Remunan-Lopez C, Alonso MJ. (2009). New generation of hybrid poly/oligosaccharide nanoparticles as carriers for the nasal delivery of macromolecules. Biomacromolecules 10:243–9
  • van Asselt DZ, Merkus FW, Russel FG, Hoefnagels WH. (1998). Nasal absorption of hydroxocobalamin in healthy elderly adults. Br J Clin Pharmacol 45:83–6
  • van der Kuy PH, Lohman JJ, Hooymans PM, et al. (1999). Bioavailability of intranasal formulations of dihydroergotamine. Eur J Clin Pharmacol 55:677–80
  • Varshosaz J, Sadrai H, Heidari A. (2006). Nasal delivery of insulin using bioadhesive chitosan gels. Drug Deliv 13:31–8
  • Veronesi MC, Kubek DJ, Kubek MJ. (2011). Intranasal delivery of neuropeptides. Methods Mol Biol 789:303–12
  • Visor GC, Bajka E, Benjamin E. (1986). Intranasal delivery of nicardipine in the rat. J Pharm Sci 75:44–6
  • Vujanic A, Sutton P, Snibson KJ, et al. (2012). Mucosal vaccination: lung versus nose. Vet Immunol Immunopathol 148:172–7
  • Wang D, Christopher ME, Nagata LP, et al. (2004). Intranasal immunization with liposome-encapsulated plasmid DNA encoding influenza virus hemagglutinin elicits mucosal, cellular and humoral immune responses. J Clin Virol 31:S99–106
  • Wang J, Tabata Y, Morimoto K. (2006). Aminated gelatin microspheres as a nasal delivery system for peptide drugs: evaluation of in vitro release and in vivo insulin absorption in rats. J Control Release 113:31–7
  • Wang SH, Fan Y, Makidon PE, et al. (2012a). Induction of immune tolerance in mice with a novel mucosal nanoemulsion adjuvant and self-antigen. Nanomedicine (Lond) 7:867–76
  • Wang SH, Kirwan SM, Abraham SN, et al. (2012b). Stable dry powder formulation for nasal delivery of anthrax vaccine. J Pharm Sci 101:31–47
  • Wang X, Zheng C, Wu Z, et al. (2009). Chitosan-NAC nanoparticles as a vehicle for nasal absorption enhancement of insulin. J Biomed Mater Res B Appl Biomater 88:150–61
  • Wearley LL. (1991). Recent progress in protein and peptide delivery by noninvasive routes. Crit Rev Ther Drug Carrier Syst 8:331–94
  • Wen Z, Yan Z, He R, et al. (2011). Brain targeting and toxicity study of odorranalectin-conjugated nanoparticles following intranasal administration. Drug Deliv 18:555–61
  • Witschi C, Mrsny RJ. (1999). In vitro evaluation of microparticles and polymer gels for use as nasal platforms for protein delivery. Pharm Res 16:382–90
  • Wu H, Li J, Zhang Q, et al. (2012). A novel small Odorranalectin-bearing cubosomes: preparation, brain delivery and pharmacodynamic study on amyloid-beta(2)(5)(-)(3)(5)-treated rats following intranasal administration. Eur J Pharm Biopharm 80:368–78
  • Xu W, Shen Y, Jiang Z, et al. (2004). Intranasal delivery of chitosan-DNA vaccine generates mucosal SIgA and anti-CVB3 protection. Vaccine 22:3603–12
  • Yang ZZ, Zhang YQ, Wu K, et al. (2012). Tissue distribution and pharmacodynamics of rivastigmine after intranasal and intravenous administration in rats. Curr Alzheimer Res 9:315–25
  • Zhang X, Wang Y, Zheng C, Li C. (2012). Phenylboronic acid-functionalized glycopolymeric nanoparticles for biomacromolecules delivery across nasal respiratory. Eur J Pharm Biopharm 82:76–84

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.