Intranasal treatment of central nervous system dysfunction in humans

Colin D Chapman, William H Frey 2nd, Suzanne Craft, Lusine Danielyan, Manfred Hallschmid, Helgi B Schiöth, Christian Benedict, Colin D Chapman, William H Frey 2nd, Suzanne Craft, Lusine Danielyan, Manfred Hallschmid, Helgi B Schiöth, Christian Benedict

Abstract

One of the most challenging problems facing modern medicine is how to deliver a given drug to a specific target at the exclusion of other regions. For example, a variety of compounds have beneficial effects within the central nervous system (CNS), but unwanted side effects in the periphery. For such compounds, traditional oral or intravenous drug delivery fails to provide benefit without cost. However, intranasal delivery is emerging as a noninvasive option for delivering drugs to the CNS with minimal peripheral exposure. Additionally, this method facilitates the delivery of large and/or charged therapeutics, which fail to effectively cross the blood-brain barrier (BBB). Thus, for a variety of growth factors, hormones, neuropeptides and therapeutics including insulin, oxytocin, orexin, and even stem cells, intranasal delivery is emerging as an efficient method of administration, and represents a promising therapeutic strategy for the treatment of diseases with CNS involvement, such as obesity, Alzheimer's disease, Parkinson's disease, Huntington's disease, depression, anxiety, autism spectrum disorders, seizures, drug addiction, eating disorders, and stroke.

Figures

Fig. 1
Fig. 1
A scheme illustrating the mechanism of nose-to-brain delivery. Unlike the olfactory nerve which terminates in the olfactory bulb, the trigeminal nerve enters the brain through both the pons and the cribriform plate, which allows for drug delivery to both the anterior and posterior regions of the brain (9,12). Transport of substances along the olfactory and trigeminal nerve pathways can happen through both intracellular and extracellular mechanisms (9). However, intracellular transport is a slow process, requiring at best several hours and at worst several days (17,20). Extracellular transport, on the other hand, is rapid and likely accounts for much of the rapid delivery and onset of action observed with intranasal CNS therapeutics (8,21). Abbreviations: ECS, extracellular space.

References

    1. van Sorge NM, Doran KS. Defense at the border: the blood-brain barrier versus bacterial foreigners. Future Microbiol. 2012;7(3):383–394. doi: 10.2217/fmb.12.1.
    1. Bitter C, Suter-Zimmermann K, Surber C. Nasal drug delivery in humans. Curr Probl Dermatol. 2011;40:20–35. doi: 10.1159/000321044.
    1. Lindup WE, Orme MC. Clinical pharmacology: plasma protein binding of drugs. Br Med J (Clin Res Ed) 1981;282(6259):212–4. doi: 10.1136/bmj.282.6259.212.
    1. Hanson LR, Frey WH., II Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008;9(Suppl 3):S5. doi: 10.1186/1471-2202-9-S3-S5.
    1. Jiang Y, Zhu J, Xu G, Liu X. Intranasal delivery of stem cells to the brain. Expert Opin Drug Deliv. 2011;8(5):623–32. doi: 10.1517/17425247.2011.566267.
    1. Frey WH, II, Liu J, Chen X, Thorne RG, Fawcett JR, Ala TA, et al. Delivery of 125I-NGF to the brain via the olfactory route. Drug Delivery. 1997;4:87–92. doi: 10.3109/10717549709051878.
    1. Chen XQ, Fawcett JR, Rahman YE, Ala TA, Frey WH., II Delivery of nerve growth factor to the brain via the olfactory pathway. J Alzheimers Dis. 1998;1(1):35–44.
    1. Dhuria SV, Hanson LR, Frey WH., II Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J Pharm Sci. 2010;99(4):1654–73.
    1. Lochhead J, Thorne R. Intranasal delivery of biologics to the central nervous system. Advanced Drug Delivery Reviews. 2012;64:614–628. doi: 10.1016/j.addr.2011.11.002.
    1. Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5(6):514–516. doi: 10.1038/nn0602-849.
    1. Thorne RG, Pronk GJ, Padmanabhan V, Frey WH., II Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127:481–496. doi: 10.1016/j.neuroscience.2004.05.029.
    1. Renner DB, Svitak AL, Gallus NG, Ericson ME, Frey WH II, Hanson LR. Intranasal delivery of insulin via the olfactory nerve pathway. J Pharm Pharmacol. 2012; doi:10.1111/j.2042-7158.2012.01555.x.
    1. Thorne RG, Hanson LR, Ross TM, Tung D, Frey WH., II Delivery of interferon-b to the monkey nervous system following intranasal administration. Neuroscience. 2008;152:785–797. doi: 10.1016/j.neuroscience.2008.01.013.
    1. Graff CL, Zhao R, Pollack GM. Pharmacokinetics of substrate uptake and distribution in murine brain after nasal instillation. Pharm Res. 2005;22:235–244. doi: 10.1007/s11095-004-1191-5.
    1. Hanson LR, Martinez PM, Taheri S, Kamsheh L, Mignot E, Frey WH., II Intranasal administration of hypocretin 1 (orexin A) bypasses the blood-brain barrier & targets the brain: A new strategy for the treatment of narcolepsy. Drug Del Tech. 2004;4:66–70.
    1. Scranton RA, Fletcher L, Sprague S, Jimenez DF, Digicaylioglu M. The rostral migratory stream plays a key role in intranasal delivery of drugs into the CNS. PLoS One. 2011;6(4):e18711. doi: 10.1371/journal.pone.0018711.
    1. Broadwell RD, Balin BJ. Endocytic and exocytic pathways of the neuronal secretory process and trans-synaptic transfer of wheat germ agglutininhorseradish peroxidase in vivo. J Comp Neurol. 1985;242:632–650. doi: 10.1002/cne.902420410.
    1. Baker H, Spencer RF. Transneuronal transport of peroxidase-conjugated wheat germ agglutinin (WGA-HRP) from the olfactory epithelium to the brain of the adult rat. Exp Brain Res. 1986;63:461–473. doi: 10.1007/BF00237470.
    1. Kristensson K, Olsson Y. Uptake of exogenous proteins in mouse olfactory cells. Acta Neuropathol (Berl) 1971;19:145–154. doi: 10.1007/BF00688493.
    1. Thorne RG, Emory CR, Ala TA, Frey WH., II Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain Res. 1995;692(1–2):278–82. doi: 10.1016/0006-8993(95)00637-6.
    1. Thorne RG, Frey WH., II Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations. Clin Pharmacokinet. 2001;40(12):907–946. doi: 10.2165/00003088-200140120-00003.
    1. Balin BJ, Broadwell RD, Salcman M, el-Kalliny M. Avenues for entry of peripherally administered protein to the central nervous system in mouse, rat, and squirrel monkey. J Comp Neurol. 1986;251:260–280. doi: 10.1002/cne.902510209.
    1. Charlton ST, Whetstone J, Fayinka ST, Read KD, Illum L, Davis SS. Evaluation of direct transport pathways of glycine receptor antagonists and an Angiotensin antagonist from the nasal cavity to the central nervous system in therat model. Pharm Res. 2008;25:1531–1543. doi: 10.1007/s11095-008-9550-2.
    1. Nonaka N, Farr SA, Kageyama H, Shioda S, Banks WA. Delivery of galanin-like peptide to the brain: targeting with intranasal delivery and cyclodextrins. J Pharmacol Exp Ther. 2008;325:513–519. doi: 10.1124/jpet.107.132381.
    1. Banks WA, During MJ, Niehoff ML. Brain uptake of the glucagon-like peptide-1 antagonist exendin(9-39) after intranasal administration. J Pharmacol Exp Ther. 2004;309:469–475. doi: 10.1124/jpet.103.063222.
    1. Dhuria SV, Hanson LR, Frey WH., 2nd Novel vasoconstrictor formulation to enhance intranasal targeting of neuropeptide therapeutics to the central nervous system. J Pharmacol Exp Ther. 2009;328(1):312–20. doi: 10.1124/jpet.108.145565.
    1. Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinology. 2004;29(10):1326–1334. doi: 10.1016/j.psyneuen.2004.04.003.
    1. Benedict C, Hallschmid M, Schmitz K, Schultes B, Ratter F, Fehm HL, et al. Intranasal insulin improves memory in humans: superiority of insulin aspart. Neuropsychopharmacology. 2007;32(1):239–43. doi: 10.1038/sj.npp.1301193.
    1. Reger MA, Watson GS, Frey WH, II, Baker LD, Cholerton B, Keeling ML, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging. 2006;27(3):451–8. doi: 10.1016/j.neurobiolaging.2005.03.016.
    1. Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology. 2008;70(6):440–8. doi: 10.1212/01.WNL.0000265401.62434.36.
    1. Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J Alzheimers Dis. 2008;13(3):323–31.
    1. Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012;69(1):29–38. doi: 10.1001/archneurol.2011.233.
    1. Benedict C, Hallschmid M, Schultes B, Born J, Kern W. Intranasal insulin to improve memory function in humans. Neuroendocrinology. 2007;86(2):136–42. doi: 10.1159/000106378.
    1. Benedict C, Frey WH, II, Schiöth HB, Schultes B, Born J, Hallschmid M. Intranasal insulin as a therapeutic option in the treatment of cognitive impairments. Exp Gerontol. 2011;46(2–3):112–5. doi: 10.1016/j.exger.2010.08.026.
    1. Schiöth HB, Frey WH, II, Brooks SJ, Benedict C. Insulin to treat Alzheimer's disease: just follow your nose? Expert Rev Clin Pharmacol. 2012;5(1):17–20. doi: 10.1586/ecp.11.70.
    1. Schiöth HB, Craft S, Brooks SJ, Frey WH II, Benedict C. Brain insulin signaling and Alzheimer's disease: current evidence and future directions. Mol Neurobiol. 2012;46(1):4–10.
    1. Craft S. Alzheimer disease: insulin resistance and AD-extending the translational path. Nat Rev Neurol. 2012;8(7):360–2. doi: 10.1038/nrneurol.2012.112.
    1. Schmidt H, Kern W, Giese R, Hallschmid M, Enders A. Intranasal insulin to improve developmental delay in children with 22q13 deletion syndrome: an exploratory clinical trial. J Med Genet. 2009;46(4):217–22. doi: 10.1136/jmg.2008.062141.
    1. Benedict C, Kern W, Schultes B, Born J, Hallschmid M. Differential sensitivity of men and women to anorexigenic and memory-improving effects of intranasal insulin. J Clin Endocrinol Metab. 2008;93(4):1339–44. doi: 10.1210/jc.2007-2606.
    1. Hallschmid M, Benedict C, Schultes B, Fehm HL, Born J, Kern W. Intranasal insulin reduces body fat in men but not in women. Diabetes. 2004;53(11):3024–9. doi: 10.2337/diabetes.53.11.3024.
    1. Hallschmid M, Higgs S, Thienel M, Ott V, Lehnert H. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Diabetes. 2012;61(4):782–9. doi: 10.2337/db11-1390.
    1. Benedict C, Brede S, Schiöth HB, Lehnert H, Schultes B, Born J, et al. Intranasal insulin enhances postprandial thermogenesis and lowers postprandial serum insulin levels in healthy men. Diabetes. 2011;60(1):114–118. doi: 10.2337/db10-0329.
    1. Guthoff M, Grichisch Y, Canova C, Tschritter O, Veit R, Hallschmid M, et al. Insulin modulates food-related activity in the central nervous system. J Clin Endocrinol Metab. 2010;95(2):748–55. doi: 10.1210/jc.2009-1677.
    1. Bohringer A, Schwabe L, Richter S, Schachinger H. Intranasal insulin attenuates the hypothalamic-pituitary-adrenal axis response to psychosocial stress. Psychoneuroendocrinology. 2008;33:1394–1400. doi: 10.1016/j.psyneuen.2008.08.002.
    1. Schwartz MW, Woods SC, Porte D, Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404(6778):661–71.
    1. Halaas J, Boozer C, Blair-West J, Fidahusein N, Denton DA, Friedman JM. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci USA. 1997;94:8878–8883. doi: 10.1073/pnas.94.16.8878.
    1. Ramsey JJ, Kemnitz JW, Colman RJ, Cunningham D, Swick AG. Different central and peripheral responses to leptin in rhesus monkeys: brain transport may be limited. J Clin Endocrinol Metab. 1998;83:3230–3235. doi: 10.1210/jc.83.9.3230.
    1. Banks WA, Kastin AJ, Huang W, Jaspan JB, Maness LM. Leptin enters the brain by a saturable system independent of insulin. Peptides. 1996;17(2):305–11. doi: 10.1016/0196-9781(96)00025-3.
    1. Fliedner S, Schulz C, Lehnert H. Brain uptake of intranasally applied radioiodinated leptin in Wistar rats. Endocrinology. 2006;147(5):2088–94. doi: 10.1210/en.2005-1016.
    1. Schulz C, Paulus K, Jöhren O, Lehnert H. Intranasal leptin reduces appetite and induces weight loss in rats with diet-induced obesity (DIO) Endocrinology. 2009;153(1):143–53. doi: 10.1210/en.2011-1586.
    1. MacVicar J. Acceleration and augmentation of labour. Scott Med J. 1973;18(6):201–214.
    1. McGonigle P. Peptide therapeutics for CNS indications. Biochem Pharmacol. 2012;83(5):559–566. doi: 10.1016/j.bcp.2011.10.014.
    1. Kosfeld M, Heinrichs M, Zak PJ, Fischbacher U, Fehr E. Oxytocin increases trust in humans. Nature. 2005;435(7042):673–6. doi: 10.1038/nature03701.
    1. Domes G, Heinrichs M, Michel A, Berger C, Herpertz SC. Oxytocin improves “mind-reading” in humans. Biol Psychiatry. 2007;61:731–3. doi: 10.1016/j.biopsych.2006.07.015.
    1. Guastella AJ, Einfeld SL, Gray KM, Rinehart NJ, Tonge BJ, Lambert TJ, et al. Intranasal oxytocin improves emotion recognition for youth with autism spectrum disorders. Biol Psychiatry. 2010;67(7):692–694. doi: 10.1016/j.biopsych.2009.09.020.
    1. Guastella AJ, Howard AL, Dadds MR, Mitchell P, Carson DS. A randomized controlled trial of intranasal oxytocin as an adjunct to exposure therapy for social anxiety disorder. Psychoneuroendocrinology. 2009;34(6):917–923. doi: 10.1016/j.psyneuen.2009.01.005.
    1. Simeon D, Bartz J, Hamilton H, Crystal S, Braun A, Ketay S, et al. Oxytocin administration attenuates stress reactivity in borderline personality disorder: a pilot study. Psychoneuroendocrinology. 2011;36(9):1418–1421. doi: 10.1016/j.psyneuen.2011.03.013.
    1. Pedersen CA, Gibson CM, Rau SW, Salimi K, Smedley KL, Casey RL, et al. Intranasal oxytocin reduces psychotic symptoms and improves Theory of Mind and social perception in schizophrenia. Schizophr Res. 2011;132(1):50–53. doi: 10.1016/j.schres.2011.07.027.
    1. Brüne M. Theory of mind and the role of IQ in chronic disorganized schizophrenia. Schizophr Res. 2003;60(1):57–64. doi: 10.1016/S0920-9964(02)00162-7.
    1. Adolphs R, Tranel D, Damasio AR. The human amygdala in social judgment. Nature. 1998;393(6684):470–4. doi: 10.1038/30982.
    1. Arletti R, Benelli A, Bertolini A. Oxytocin inhibits food and fluid intake in rats. Physiol Behav. 1990;48(6):825–30. doi: 10.1016/0031-9384(90)90234-U.
    1. Olson BR, Drutarosky MD, Chow MS, Hruby VJ, Stricker EM, Verbalis JG. Oxytocin and an oxytocin agonist administered centrally decrease food intake in rats. Peptides. 1991;12(1):113–118. doi: 10.1016/0196-9781(91)90176-P.
    1. Morton GJ, Thatcher BS, Reidelberger RD, Ogimoto K, Wolden-Hanson T, Baskin DG, et al. Peripheral oxytocin suppresses food intake and causes weight loss in diet-induced obese rats. Am J Physiol Endocrinol Metab. 2012;302(1):E134–144. doi: 10.1152/ajpendo.00296.2011.
    1. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. Genet Med. 2012;14(1):10–26. doi: 10.1038/gim.0b013e31822bead0.
    1. Swaab DF. Prader-Willi syndrome and the hypothalamus. Acta Paediatr Suppl. 1997;423:50–54. doi: 10.1111/j.1651-2227.1997.tb18369.x.
    1. Deadwyler SA, Porrino L, Siegel JM, Hampson RE. Systemic and nasal delivery of orexin-A (Hypocretin-1) reduces the effects of sleep deprivation on cognitive performance in nonhuman primates. J Neurosci. 2007;27(52):14239–14247. doi: 10.1523/JNEUROSCI.3878-07.2007.
    1. Baier PC, Weinhold SL, Huth V, Gottwald B, Ferstl R, Hinze-Selch D. Olfactory dysfunction in patients with narcolepsy with cataplexy is restored by intranasal Orexin A (Hypocretin-1) Brain. 2008;131(10):2734–2741. doi: 10.1093/brain/awn193.
    1. Baier PC, Hallschmid M, Seeck-Hirschner M, Weinhold SL, Burkert S, Diessner N, et al. Effects of intranasal hypocretin-1 (orexin A) on sleep in narcolepsy with cataplexy. Sleep Med. 2011;12(10):941–946. doi: 10.1016/j.sleep.2011.06.015.
    1. Dhuria SV, Hanson LR, Frey WH., II Intranasal drug targeting of hypocretin-1 (orexin-A) to the central nervous system. J Pharm Sci. 2009;98(7):2501–2515. doi: 10.1002/jps.21604.
    1. Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich S, et al. A comparison of lorazepam, diazepam, and placebo for the treatment of out-of-hospital status epilepticus. N Engl J Med. 2001;345(9):631–637. doi: 10.1056/NEJMoa002141.
    1. Bassin S, Smith TL, Bleck TP. Clinical review: status epilepticus. Crit Care. 2002;6(2):137–142. doi: 10.1186/cc1472.
    1. Pang T, Hirsch LJ. Treatment of convulsive and nonconvulsive status epilepticus. Curr Treat Options Neurol. 2005;7(4):247–259. doi: 10.1007/s11940-005-0035-x.
    1. Wermling DP. Intranasal delivery of antiepileptic medications for treatment of seizures. Neurotherapeutics. 2009;6(2):352–8. doi: 10.1016/j.nurt.2009.01.002.
    1. Jeannet PY, Roulet E, Maeder-Ingvar M, Gehri M, Jutzi A, Deonna T. Home and hospital treatment of acute seizures in children with nasal midazolam. Eur J Paediatr Neurol. 1999;3(2):73–7. doi: 10.1016/S1090-3798(99)80016-6.
    1. Fişgin T, Gurer Y, Teziç T, Senbil N, Zorlu P, Okuyaz C, et al. Effects of intranasal midazolam and rectal diazepam on acute convulsions in children: prospective randomized study. J Child Neurol. 2002;17(2):123–126. doi: 10.1177/088307380201700206.
    1. Holsti M, Dudley N, Schunk J, Adelgais K, Greenberg R, Olsen C, et al. Intranasal midazolam vs rectal diazepam for the home treatment of acute seizures in pediatric patients with epilepsy. Arch Pediatr Adolesc Med. 2010;164(8):747–53. doi: 10.1001/archpediatrics.2010.130.
    1. de Haan GJ, van der Geest P, Doelman G, Bertram E, Edelbroek P. A comparison of midazolam nasal spray and diazepam rectal solution for the residential treatment of seizure exacerbations. Epilepsia. 2010;51(3):478–482. doi: 10.1111/j.1528-1167.2009.02333.x.
    1. Kerr D, Kelly AM, Dietze P, Jolley D, Barger B. Randomized controlled trial comparing the effectiveness and safety of intranasal and intramuscular naloxone for the treatment of suspected heroin overdose. Addiction. 2009;104(12):2067–74. doi: 10.1111/j.1360-0443.2009.02724.x.
    1. Robertson TM, Hendey GW, Stroh G, Shalit M. Intranasal naloxone is a viable alternative to intravenous naloxone for prehospital narcotic overdose. Prehosp Emerg Care. 2009;13(4):512–5. doi: 10.1080/10903120903144866.
    1. Ashton H, Hassan Z. Best evidence topic report. Intranasal naloxone in suspected opioid overdose. Emerg Med J. 2006;23(3):221–3. doi: 10.1136/emj.2005.034322.
    1. Lightlake Sinclare Ltd. Clinical Trial on Binge Eating Disorder, Treatment With Naloxone Spray (BED). Available at: .
    1. Lindvall O. Stem cells for cell therapy in Parkinson's disease. Pharmacol Res. 2003;47(4):279–287. doi: 10.1016/S1043-6618(03)00037-9.
    1. Joyce N, Annett G, Wirthlin L, Olson S, Bauer G, Nolta JA. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen Med. 2010;5(6):933–46. doi: 10.2217/rme.10.72.
    1. Babaei P, Soltani Tehrani B, Alizadeh A. Transplanted bone marrow mesenchymal stem cells improve memory in rat models of Alzheimer's Disease. Stem Cells Int. 2012; 369417.
    1. Sinden JD, Muir KW. Stem cells in stroke treatment: the promise and the challenges. Int J Stroke. 2012;7(5):426–34. doi: 10.1111/j.1747-4949.2012.00840.x.
    1. Danielyan L, Schäfer R, von Ameln-Mayerhofer A, Buadze M, Geisler J, Klopfer T, et al. Intranasal delivery of cells to the brain. Eur J Cell Biol. 2009;88(6):315–324. doi: 10.1016/j.ejcb.2009.02.001.
    1. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Nasal administration of stem cells: a promising novel route to treat neonatal ischemic brain damage. Pediatr Res. 2010;68(5):419–422.
    1. Danielyan L, Schäfer R, von Ameln-Mayerhofer A, Bernhard F, Verleysdonk S, Buadze M, et al. Therapeutic efficacy of intranasally delivered mesenchymal stem cells in a rat model of Parkinson disease. Rejuvination Res. 2011;14(1):3–16. doi: 10.1089/rej.2010.1130.
    1. Bossolasco P, Cova L, Levandis G, Diana V, Cerri S, Lambertenghi Deliliers G, et al. Noninvasive near-infrared live imaging of human adult mesenchymal stem cells transplanted in a rodent model of Parkinson's disease. Int J Nanomedicine. 2012;7:435–47.
    1. Chartoff EH, Damez-Werno D, Sonntag KC, Hassinger L, Kaufmann DE, Peterson J, et al. Detection of intranasally delivered bone marrow-derived mesenchymal stromal cells in the lesioned mouse brain: a cautionary report. Stem Cells International. 2011. doi:10.4061/2011/586586.
    1. Yu SP, Wei N, Chau TC, Deveau L, Wei L. Distribution and therapeutic benefits of intranasally administered hypoxia-preconditioned bone marrow stem cells after barrel cortex stroke. 25th International Symposium on Cerebral Blood Flow, Metabolism, and Function. Barcelona, Spain. 2011, Available at: .
    1. Wei N, Yu SP, Gu X, Taylor TM, Song D, Liu XF, et al. Delayed intranasal delivery of hypoxic-preconditioned bone marrow mesenchymal stem cells enhanced cell homing and therapeutic benefits after ischemic stroke in mice. Cell Transplant. 2012. doi:10.3727/096368912X657251.
    1. MacDonald E, Dadds MR, Brennan JL, Williams K, Levy F, Cauchi AJ. A review of safety, side-effects and subjective reactions to intranasal oxytocin in human research. Psychoneuroendocrinology. 2011;36(8):1114–1126. doi: 10.1016/j.psyneuen.2011.02.015.
    1. Nathan RA. Intranasal steroids in the treatment of allergy-induced rhinorrhea. Clin Rev Allergy Immunol. 2011;41(1):89–101. doi: 10.1007/s12016-010-8206-2.
    1. Shemesh E, Rudich A, Harman-Boehm I, Cukierman-Yaffe T. Effect of intranasal insulin on cognitive function: a systematic review. J Clin Endocrinol Metab. 2012;97(2):366–76. doi: 10.1210/jc.2011-1802.
    1. Wolfe TR, Macfarlane TC. Intranasal midazolam therapy for pediatric status epilepticus. Am J Emerg Med. 2006;24(3):343–6. doi: 10.1016/j.ajem.2005.11.004.
    1. Woods SC, Seeley RJ, Baskin DG, Schwartz MW. Insulin and the blood-brain barrier. Curr Pharm Des. 2003;9(10):795–800. doi: 10.2174/1381612033455323.
    1. Kern W, Peters A, Born J, Fehm HL, Schultes B. Changes in blood pressure and plasma catecholamine levels during prolonged hyperinsulinemia. Metabolism. 2005;54(3):391–396. doi: 10.1016/j.metabol.2004.10.005.
    1. Fruehwald-Schultes B, Kern W, Born J, Fehm HL, Peters A. Hyperinsulinemia causes activation of the hypothalamus-pituitary-adrenal axis in humans. Int J Obes Relat Metab Disord. 2001;25(Suppl 1):S38–40. doi: 10.1038/sj.ijo.0801695.
    1. Benedict C, Dodt C, Hallschmid M, Lepiorz M, Fehm HL, Born J, et al. Immediate but not long-term intranasal administration of insulin raises blood pressure in human beings. Metabolism. 2005;54(10):1356–61. doi: 10.1016/j.metabol.2005.04.026.
    1. Frey WH II. Method of administering neurologic agents to the brain. US Patent 5,624,898 filed 1989 and issued April 29, 1997.
    1. Frey WH II. Neurologic agents for nasal administration to the brain. PCT International Patent WO91/07947 filed 1990 and issued June 13, 1991.
    1. Frey WH II. Method for administering insulin to the brain. Patent 6,313,093 B1 filed 1999 and issued November 6, 2001.
    1. Jogani V, Jinturkar K, Vyas T, Misra A. Recent patents review on intranasal administration for CNS drug delivery. Recent Pat Drug Deliv Formul. 2008;2(1):25–40. doi: 10.2174/187221108783331429.

Source: PubMed

3
Se inscrever