500
Views
33
CrossRef citations to date
0
Altmetric
Reviews

Intranasal delivery: circumventing the iron curtain to treat neurological disorders

, &

Bibliography

  • O’Collins VE, Macleod MR, Donnan GA, et al. 1,026 experimental treatments in acute stroke. Ann Neurol 2006;59:467–77
  • Turner RC, Dodson SC, Rosen CL, et al. The science of cerebral ischemia and the quest for neuroprotection: navigating past failure to future success. J Neurosurg 2013;118:1072–85
  • Jiang Y, Zhu J, Xu G, et al. Intranasal delivery of stem cells to the brain. Expert Opin Drug Deliv 2011;8:623–32
  • Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev 2012;64:614–28
  • Fisher M, Feuerstein G, Howells DW, et al. Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke 2009;40:2244–50
  • Zhu J, Jiang Y, Xu G, et al. Intranasal administration: a potential solution for cross-BBB delivering neurotrophic factors. Histol Histopathol 2012;27:537–48
  • Zhao H, Wang J, Gao L, et al. MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation. Stroke 2013;44:1706–13
  • Fousteri G, Chan JR, Zheng Y, et al. Virtual optimization of nasal insulin therapy predicts immunization frequency to be crucial for diabetes protection. Diabetes 2010;59:3148–58
  • Law S, Derry S, Moore RA. Triptans for acute cluster headache. Cochrane Database Syst Rev 2010;CD008042
  • Frey WH. Neurologic agents for nasal administration to the brain. Chiron Corporation; US: 1991
  • Topkoru BC, Altay O, Duris K, et al. Nasal administration of recombinant osteopontin attenuates early brain injury after subarachnoid hemorrhage. Stroke 2013;44:3189–94
  • Hashizume R, Ozawa T, Gryaznov SM, et al. New therapeutic approach for brain tumors: Intranasal delivery of telomerase inhibitor GRN163. Neuro Oncol 2008;10:112–20
  • Jiang Y, Wei N, Zhu J, et al. A new approach with less damage: intranasal delivery of tetracycline-inducible replication-defective herpes simplex virus type-1 vector to brain. Neuroscience 2012;201:96–104
  • Han IK, Kim MY, Byun HM, et al. Enhanced brain targeting efficiency of intranasally administered plasmid DNA: an alternative route for brain gene therapy. J Mol Med 2007;85:75–83
  • Kim ID, Shin JH, Kim SW, et al. Intranasal delivery of HMGB1 siRNA confers target gene knockdown and robust neuroprotection in the postischemic brain. Mol Ther 2012;20:829–39
  • Danielyan L, Schafer R, von Ameln-Mayerhofer A, et al. Intranasal delivery of cells to the brain. Eur J Cell Biol 2009;88:315–24
  • van Velthoven CT, Kavelaars A, van Bel F, et al. Nasal administration of stem cells: a promising novel route to treat neonatal ischemic brain damage. Pediatr Res 2010;68:419–22
  • Albertsson AM, Zhang X, Leavenworth J, et al. The effect of osteopontin and osteopontin-derived peptides on preterm brain injury. J Neuroinflammation 2014;11:197
  • Alex AT, Joseph A, Shavi G, et al. Development and evaluation of carboplatin-loaded PCL nanoparticles for intranasal delivery. Drug Deliv 2014;1–10
  • Kanazawa T. Brain delivery of small interfering ribonucleic acid and drugs through intranasal administration with nano-sized polymer micelles. Med Devices (Auckl) 2015;8:57–64
  • Mustafa G, Ahuja A, Al Rohaimi AH, et al. Nano-ropinirole for the management of Parkinsonism: blood-brain pharmacokinetics and carrier localization. Expert Rev Neurother 2015;15:695–710
  • Florence K, Manisha L, Kumar BA, et al. Intranasal clobazam delivery in the treatment of status epilepticus. J Pharm Sci 2011;100:692–703
  • Balyasnikova IV, Prasol MS, Ferguson SD, et al. Intranasal delivery of mesenchymal stem cells significantly extends survival of irradiated mice with experimental brain tumors. Mol Ther 2014;22:140–8
  • De Rosa R, Garcia AA, Braschi C, et al. Intranasal administration of nerve growth factor (NGF) rescues recognition memory deficits in AD11 anti-NGF transgenic mice. Proc Natl Acad Sci USA 2005;102:3811–16
  • Martinez JA, Francis GJ, Liu WQ, et al. Intranasal delivery of insulin and a nitric oxide synthase inhibitor in an experimental model of amyotrophic lateral sclerosis. Neuroscience 2008;157:908–25
  • Yuki Y, Byun Y, Fujita M, et al. Production of a recombinant hybrid molecule of cholera toxin-B-subunit and proteolipid-protein-peptide for the treatment of experimental encephalomyelitis. Biotechnol Bioeng 2001;74:62–9
  • Jiang Y, Wei N, Lu T, et al. Intranasal brain-derived neurotrophic factor protects brain from ischemic insult via modulating local inflammation in rats. Neuroscience 2011;172:398–405
  • Chen M, Wu B, Ye X, et al. Association between plasma homocysteine levels and obstructive sleep apnoea in patients with ischaemic stroke. J Clin Neurosci 2011;18:1454–7
  • Pardeshi CV, Belgamwar VS. Direct nose to brain drug delivery via integrated nerve pathways bypassing the blood-brain barrier: an excellent platform for brain targeting. Expert Opin Drug Deliv 2013;10:957–72
  • Pires A, Fortuna A, Alves G, et al. Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci 2009;12:288–311
  • Jansson B, Bjork E. Visualization of in vivo olfactory uptake and transfer using fluorescein dextran. J Drug Target 2002;10:379–86
  • Dhuria SV, Hanson LR, Frey WHII. Novel vasoconstrictor formulation to enhance intranasal targeting of neuropeptide therapeutics to the central nervous system. J Pharmacol Exp Ther 2009;328:312–20
  • Thorne RG, Pronk GJ, Padmanabhan V, et al. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience 2004;127:481–96
  • Mitchell FL, Marks GE, Bichenkova EV, et al. Molecular probes: insights into design and analysis from computational and physical chemistry. Biochem Soc Trans 2008;36:46–50
  • Hadaczek P, Yamashita Y, Mirek H, et al. The “perivascular pump” driven by arterial pulsation is a powerful mechanism for the distribution of therapeutic molecules within the brain. Mol Ther 2006;14:69–78
  • Skipor J, Grzegorzewski W, Einer-Jensen N, et al. Local vascular pathway for progesterone transfer to the brain after nasal administration in gilts. Reprod Biol 2003;3:143–59
  • Lochhead JJ, Wolak DJ, Pizzo ME, et al. Rapid transport within cerebral perivascular spaces underlies widespread tracer distribution in the brain after intranasal administration. J Cereb Blood Flow Metab 2015;35:371–81
  • Holscher C. Diabetes as a risk factor for Alzheimer’s disease: insulin signalling impairment in the brain as an alternative model of Alzheimer’s disease. Biochem Soc Trans 2011;39:891–7
  • De Felice FG, Vieira MN, Bomfim TR, et al. Protection of synapses against Alzheimer’s-linked toxins: insulin signaling prevents the pathogenic binding of Abeta oligomers. Proc Natl Acad Sci USA 2009;106:1971–6
  • de la Monte SM. Brain insulin resistance and deficiency as therapeutic targets in Alzheimer’s disease. Curr Alzheimer Res 2012;9:35–66
  • Yang Y, Ma D, Wang Y, et al. Intranasal insulin ameliorates tau hyperphosphorylation in a rat model of type 2 diabetes. J Alzheimers Dis 2013;33:329–38
  • Wallum BJ, Taborsky GJJr, Porte DJr, et al. Cerebrospinal fluid insulin levels increase during intravenous insulin infusions in man. J Clin Endocrinol Metab 1987;64:190–4
  • Porte DJr, Woods SC. Regulation of food intake and body weight in insulin. Diabetologia 1981;20(Suppl):274–80
  • Park CR, Seeley RJ, Craft S, et al. Intracerebroventricular insulin enhances memory in a passive-avoidance task. Physiol Behav 2000;68:509–14
  • Kern W, Born J, Schreiber H, et al. Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes 1999;48:557–63
  • Born J, Lange T, Kern W, et al. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci 2002;5:514–16
  • Benedict C, Hallschmid M, Hatke A, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinology 2004;29:1326–34
  • Benedict C, Hallschmid M, Schmitz K, et al. Intranasal insulin improves memory in humans: superiority of insulin aspart. Neuropsychopharmacology 2007;32:239–43
  • Krug R, Benedict C, Born J, et al. Comparable sensitivity of postmenopausal and young women to the effects of intranasal insulin on food intake and working memory. J Clin Endocrinol Metab 2010;95:E468–72
  • Benedict C, Kern W, Schultes B, et al. Differential sensitivity of men and women to anorexigenic and memory-improving effects of intranasal insulin. J Clin Endocrinol Metab 2008;93:1339–44
  • Hallschmid M, Benedict C, Schultes B, et al. Obese men respond to cognitive but not to catabolic brain insulin signaling. Int J Obes (Lond) 2008;32:275–82
  • Reger MA, Watson GS, Green PS, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology 2008;70:440–8
  • Reger MA, Watson GS, Frey WHII, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging 2006;27:451–8
  • Claxton A, Baker LD, Wilkinson CW, et al. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or Alzheimer’s disease. J Alzheimers Dis 2013;35:789–97
  • Reger MA, Watson GS, Green PS, et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J Alzheimers Dis 2008;13:323–31
  • Freiherr J, Hallschmid M, Frey WHII, et al. Intranasal insulin as a treatment for Alzheimer’s disease: a review of basic research and clinical evidence. CNS Drugs 2013;27:505–14
  • Craft S, Baker LD, Montine TJ, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol 2012;69:29–38
  • Benedict C, Frey WHII, Schioth HB, et al. Intranasal insulin as a therapeutic option in the treatment of cognitive impairments. Exp Gerontol 2011;46:112–15
  • Shemesh E, Rudich A, Harman-Boehm I, et al. Effect of intranasal insulin on cognitive function: a systematic review. J Clin Endocrinol Metab 2012;97:366–76
  • Schilling TM, Ferreira de Sa DS, Westerhausen R, et al. Intranasal insulin increases regional cerebral blood flow in the insular cortex in men independently of cortisol manipulation. Hum Brain Mapp 2014;35:1944–56
  • Jauch-Chara K, Friedrich A, Rezmer M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes 2012;61:2261–8
  • Kullmann S, Frank S, Heni M, et al. Intranasal insulin modulates intrinsic reward and prefrontal circuitry of the human brain in lean women. Neuroendocrinology 2013;97:176–82
  • Renner DB, Svitak AL, Gallus NJ, et al. Intranasal delivery of insulin via the olfactory nerve pathway. J Pharm Pharmacol 2012;64:1709–14
  • Watt JL, Olson IA, Johnston AW, et al. A familial pericentric inversion of chromosome 22 with a recombinant subject illustrating a ’pure’ partial monosomy syndrome. J Med Genet 1985;22:283–7
  • Schmidt H, Kern W, Giese R, et al. Intranasal insulin to improve developmental delay in children with 22q13 deletion syndrome: an exploratory clinical trial. J Med Genet 2009;46:217–22
  • McIntyre RS, Soczynska JK, Woldeyohannes HO, et al. A randomized, double-blind, controlled trial evaluating the effect of intranasal insulin on neurocognitive function in euthymic patients with bipolar disorder. Bipolar Disord 2012;14:697–706
  • Fan X, Copeland PM, Liu EY, et al. No effect of single-dose intranasal insulin treatment on verbal memory and sustained attention in patients with schizophrenia. J Clin Psychopharmacol 2011;31:231–4
  • Li J, Li X, Liu E, et al. No effect of adjunctive, repeated dose intranasal insulin treatment on body metabolism in patients with schizophrenia. Schizophr Res 2013;146:40–5
  • Frey WHII. Intranasal insulin to treat and protect against posttraumatic stress disorder. J Nerv Ment Dis 2013;201:638–9
  • Fehm-Wolfsdorf G, Born J. Behavioral effects of neurohypophyseal peptides in healthy volunteers: 10 years of research. Peptides 1991;12:1399–406
  • Merkus P, Guchelaar HJ, Bosch DA, et al. Direct access of drugs to the human brain after intranasal drug administration? Neurology 2003;60:1669–71
  • Pietrowsky R, Struben C, Molle M, et al. Brain potential changes after intranasal vs. intravenous administration of vasopressin: evidence for a direct nose-brain pathway for peptide effects in humans. Biol Psychiatry 1996;39:332–40
  • Perras B, Pannenborg H, Marshall L, et al. Beneficial treatment of age-related sleep disturbances with prolonged intranasal vasopressin. J Clin Psychopharmacol 1999;19:28–36
  • Eames P, Wood RL. Lysine vasopressin in post-traumatic memory disorders: an uncontrolled pilot study. Brain Inj 1999;13:255–60
  • Tsikunov SG, Belokoskova SG. Psychophysiological analysis of the influence of vasopressin on speech in patients with post-stroke aphasias. Span J Psychol 2007;10:178–88
  • Thompson R, Gupta S, Miller K, et al. The effects of vasopressin on human facial responses related to social communication. Psychoneuroendocrinology 2004;29:35–48
  • Brunnlieb C, Munte TF, Kramer U, et al. Vasopressin modulates neural responses during human reactive aggression. Soc Neurosci 2013;8:148–64
  • Guastella AJ, Kenyon AR, Alvares GA, et al. Intranasal arginine vasopressin enhances the encoding of happy and angry faces in humans. Biol Psychiatry 2010;67:1220–2
  • Pietrowsky R, Thiemann A, Kern W, et al. A nose-brain pathway for psychotropic peptides: evidence from a brain evoked potential study with cholecystokinin. Psychoneuroendocrinology 1996;21:559–72
  • Denecke H, Czehak N, Pietrowsky R. Dose-response relationships of intranasal cholecystokinin and the P300 event-related brain potential. Pharmacol Biochem Behav 2002;73:593–600
  • Denecke H, Meyer F, Feldkamp J, et al. Repetitive intranasal administration of cholecystokinin potentiates its central nervous effects. Physiol Behav 2004;83:39–45
  • Smolnik R, Fischer S, Hagenah J, et al. Brain potential signs of slowed stimulus processing following cholecystokinin in Parkinson’s disease. Psychopharmacology (Berl) 2002;161:70–6
  • Striepens N, Kendrick KM, Maier W, et al. Prosocial effects of oxytocin and clinical evidence for its therapeutic potential. Front Neuroendocrinol 2011;32:426–50
  • Newton M, Egli GE. The effect of intranasal administration of oxytocin on the let-down of milk in lactating women. Am J Obstet Gynecol 1958;76:103–7
  • Kirsch P, Esslinger C, Chen Q, et al. Oxytocin modulates neural circuitry for social cognition and fear in humans. J Neurosci 2005;25:11489–93
  • Baumgartner T, Heinrichs M, Vonlanthen A, et al. Oxytocin shapes the neural circuitry of trust and trust adaptation in humans. Neuron 2008;58:639–50
  • Domes G, Heinrichs M, Kumbier E, et al. Effects of intranasal oxytocin on the neural basis of face processing in autism spectrum disorder. Biol Psychiatry 2013;74:164–71
  • Domes G, Heinrichs M, Michel A, et al. Oxytocin improves “mind-reading” in humans. Biol Psychiatry 2007;61:731–3
  • Opar A. Search for potential autism treatments turns to ’trust hormone’. Nat Med 2008;14:353
  • Guastella AJ, Einfeld SL, Gray KM, et al. Intranasal oxytocin improves emotion recognition for youth with autism spectrum disorders. Biol Psychiatry 2010;67:692–4
  • Naber FB, Poslawsky IE, van Ijzendoorn MH, et al. Brief report: oxytocin enhances paternal sensitivity to a child with autism: a double-blind within-subject experiment with intranasally administered oxytocin. J Autism Dev Disord 2013;43:224–9
  • Tachibana M, Kagitani-Shimono K, Mohri I, et al. Long-term administration of intranasal oxytocin is a safe and promising therapy for early adolescent boys with autism spectrum disorders. J Child Adolesc Psychopharmacol 2013;23:123–7
  • Sauer C, Montag C, Worner C, et al. Effects of a common variant in the CD38 gene on social processing in an oxytocin challenge study: possible links to autism. Neuropsychopharmacology 2012;37:1474–82
  • Montag C, Sauer C, Reuter M, et al. An interaction between oxytocin and a genetic variation of the oxytocin receptor modulates amygdala activity toward direct gaze: evidence from a pharmacological imaging genetics study. Eur Arch Psychiatry Clin Neurosci 2013;263(Suppl 2):S169–75
  • Drolet BC, Lorenzi NM. Translational research: understanding the continuum from bench to bedside. Transl Res 2011;157:1–5
  • Zerhouni EA. Translational and clinical science--time for a new vision. N Engl J Med 2005;353:1621–3
  • Liu X. Clinical trials of intranasal delivery for treating neurological disorders--a critical review. Expert Opin Drug Deliv 2011;8:1681–90
  • da Rocha Lindner G, Bonfanti Santos D, Colle D, et al. Improved neuroprotective effects of resveratrol-loaded polysorbate 80-coated poly(lactide) nanoparticles in MPTP-induced Parkinsonism. Nanomedicine (Lond) 2015;10:1127–38
  • Lu CT, Jin RR, Jiang YN, et al. Gelatin nanoparticle-mediated intranasal delivery of substance P protects against 6-hydroxydopamine-induced apoptosis: an in vitro and in vivo study. Drug Des Devel Ther 2015;9:1955–62
  • Fazil M, Md S, Haque S, et al. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur J Pharm Sci 2012;47:6–15
  • Haque S, Md S, Fazil M, et al. Venlafaxine loaded chitosan NPs for brain targeting: pharmacokinetic and pharmacodynamic evaluation. Carbohydr Polym 2012;89:72–9

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.