C. Lecoutey, D. Hedou, T. Freret, P. Giannoni, F. Gaven et al., Design of donecopride, a dual serotonin subtype 4 receptor agonist/acetylcholinesterase inhibitor with potential interest for Alzheimer's disease treatment, Proc Natl Acad Sci, vol.111, issue.36, pp.3825-3830, 2009.

C. Gloaguen, A. S. Voisin-chiret, J. Sopkova-de-oliveira-santos, J. Fogha, F. Gautier et al., First evidence that oligopyridines, ?-helix foldamers, inhibit Mcl-1 and sensitize ovarian carcinoma cells to Bcl-xL-targeting strategies, J Med Chem, vol.58, issue.4, pp.1644-68, 2015.

, World Alzheimer Report, 2015.

L. Buée, A. Delacourte, and . La, Alzheimer : une tauopathie parmi d'autres ? médecine/sciences, vol.18, pp.727-763, 2002.

P. T. Francis, A. M. Palmer, M. Snape, and G. K. Wilcock, The cholinergic hypothesis of Alzheimer's disease: a review of progress, J Neurol Neurosurg Psychiatry, vol.66, issue.2, pp.137-184, 1999.

O. Cherif, F. Allouche, F. Chabchoub, M. Chioua, E. Soriano et al., Isoxazolotacrines as non-toxic and selective butyrylcholinesterase inhibitors for Alzheimer's disease, Future Med Chem, vol.6, issue.17, pp.1883-91, 2014.

S. Darvesh, D. A. Hopkins, and C. Geula, Neurobiology of butyrylcholinesterase, Nat Rev Neurosci, vol.4, issue.2, pp.131-139, 2003.

M. S. Quraishi, N. S. Jones, and J. Mason, The rheology of nasal mucus: a review, Clin Otolaryngol Allied Sci, vol.23, issue.5, pp.403-413, 1998.

N. A. Peppas, P. Bures, W. Leobandung, and H. Ichikawa, Hydrogels in pharmaceutical formulations, Eur J Pharm Biopharm Off J Arbeitsgemeinschaft Pharm Verfahrenstechnik EV, vol.50, issue.1, pp.27-46, 2000.

H. Jacqmin-gadda, A. Alperovitch, C. Montlahuc, D. Commenges, K. Leffondre et al., 20-Year prevalence projections for dementia and impact of preventive policy about risk factors, Eur J Epidemiol, vol.28, issue.6, pp.493-502, 2013.

R. Camicioli, Distinguer les différents types de démences. Rev Can Mal D'Alzheimer Autres Démence, vol.8, pp.4-11, 2006.

C. Paterson, World Alzheimer Report, 2018.

J. F. Dartigues, M. Gagnon, P. Michel, L. Letenneur, D. Commenges et al., The Paquid research program on the epidemiology of dementia. Methods and initial results, Rev Neurol, vol.147, issue.3, pp.225-255, 1991.

L. Carcaillon-bentata, Peut-on estimer la prévalence de la maladie d'Alzheimer et autres démences à partir des bases de données médico-administratives ?, Comparaison aux données de cohortes populationnelles

C. Gallez and . Rapport, Alzheimer et les maladies apparentées au nom de l'Office parlementaire d'évaluation des politiques de santé

, Espérance de vie -Mortalité ? Tableaux de l'économie française | Insee

A. Bérard, C. Gervès, and J. Aquino, Combien coûte la maladie, p.98

, Annexe rapport d'évaluation des médicaments

N. L. Graham, T. Emery, and J. R. Hodges, Distinctive cognitive profiles in Alzheimer's disease and subcortical vascular dementia, J Neurol Neurosurg Psychiatry, vol.75, issue.1, pp.61-71, 2004.

E. Helmes and T. Østbye, Beyond memory impairment: cognitive changes in Alzheimer's disease, Arch Clin Neuropsychol, vol.17, issue.2, pp.179-193, 2002.

D. M. Jacobs, M. Sano, G. Dooneief, K. Marder, K. L. Bell et al., Neuropsychological detection and characterization of preclinical Alzheimer's disease, Neurology, vol.45, issue.5, pp.957-62, 1995.

A. R. Price, G. Xu, Z. B. Siemienski, L. A. Smithson, D. R. Borchelt et al., Comment on "ApoE-directed therapeutics rapidly clear ?-amyloid and reverse deficits in AD mouse models, Science, vol.340, issue.6135, p.924, 2013.

V. Senanarong, J. L. Cummings, L. Fairbanks, M. Mega, D. M. Masterman et al., Agitation in Alzheimer's disease is a manifestation of frontal lobe dysfunction, Dement Geriatr Cogn Disord, vol.17, issue.1-2, pp.14-20, 2004.

J. Chen, Y. Stern, M. Sano, and R. Mayeux, Cumulative Risks of Developing Extrapyramidal Signs, Psychosis, or Myoclonus in the Course of Alzheimer's Disease, Arch Neurol, vol.48, issue.11, pp.1141-1144, 1991.

D. J. Hart, D. Craig, S. A. Compton, S. Critchlow, B. M. Kerrigan et al., A retrospective study of the behavioural and psychological symptoms of mid and late phase Alzheimer's disease, Int J Geriatr Psychiatry, vol.18, issue.11, pp.1037-1079, 2003.

, Les stades de la maladie d'Alzheimer | Société Alzheimer du Canada

K. I. Shulman, Clock-drawing: is it the ideal cognitive screening test?, Int J Geriatr Psychiatry, vol.15, issue.6, pp.548-61, 2000.

, Les 5 mots", épreuve simple et sensible pour le diagnostic de la maladie

I. Gélinas, L. Gauthier, M. Mcintyre, and S. Gauthier, Development of a functional measure for persons with Alzheimer's disease: the disability assessment for dementia, Am J Occup Ther Off Publ Am Occup Ther Assoc, vol.53, issue.5, pp.471-81, 1999.

T. N. Tombaugh and N. J. Mcintyre, The mini-mental state examination: a comprehensive review, J Am Geriatr Soc, vol.40, issue.9, pp.922-957, 1992.

R. A. Ruchinskas and K. J. Curyto, Cognitive Screening in Geriatric Rehabilitation, p.10

M. Crum-r, C. Anthony, J. Bassett, S. , F. Folstein et al., Population-based norms for the Mini-Mental State Examination by age and education level, JAMA, vol.269, pp.2386-2391, 1993.

G. B. Frisoni, C. Geroldi, A. Beltramello, A. Bianchetti, G. Binetti et al., Radial width of the temporal horn: a sensitive measure in Alzheimer disease, AJNR Am J Neuroradiol, vol.23, issue.1, pp.35-47, 2002.

, Les marqueurs biochimiques du LCR : un outil diagnostique de la maladie d'Alzheimer. | Base documentaire | BDSP

S. Schraen-maschke, C. M. Dhaenens, S. Bombois, V. Deramecourt, E. V. Brussel et al., Les marqueurs biologiques de la maladie d'Alzheimer : quel intérêt pour un diagnostic moins tardif ?

H. Hippius and G. Neundörfer, The discovery of Alzheimer's disease, Dialogues Clin Neurosci, vol.5, issue.1, pp.101-109, 2003.

M. Goedert, C. M. Wischik, R. A. Crowther, J. E. Walker, and A. Klug, Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau, Proc Natl Acad Sci, vol.85, issue.11, pp.4051-4056, 1988.

I. Grundke-iqbal, K. Iqbal, Y. C. Tung, M. Quinlan, H. M. Wisniewski et al., Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology, Proc Natl Acad Sci, vol.83, issue.13, pp.4913-4920, 1986.

M. Kidd, Paired Helical Filaments in Electron Microscopy of Alzheimer's Disease, Nature, vol.197, issue.4863, pp.192-195, 1963.

M. D. Weingarten, A. H. Lockwood, S. Y. Hwo, and M. W. Kirschner, A protein factor essential for microtubule assembly, Proc Natl Acad Sci U S A, vol.72, issue.5, pp.1858-62, 1975.

J. Biernat, N. Gustke, G. Drewes, E. M. Mandelkow, and E. Mandelkow, Phosphorylation of Ser262 strongly reduces binding of tau to microtubules: distinction between PHF-like immunoreactivity and microtubule binding, Neuron, vol.11, issue.1, pp.153-63, 1993.

H. Braak and E. Braak, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol (Berl), vol.82, issue.4, pp.239-59, 1991.

H. Braak and E. Braak, Staging of Alzheimer's disease-related neurofibrillary changes, Neurobiol Aging, vol.16, issue.3, pp.271-279, 1995.

S. Tanaka, S. Shiojiri, Y. Takahashi, N. Kitaguchi, H. Ito et al., Tissue-specific expression of three types of beta-protein precursor mRNA: enhancement of protease inhibitor-harboring types in Alzheimer's disease brain, Biochem Biophys Res Commun, vol.165, issue.3, pp.1406-1420, 1989.

G. R. Dawson, G. R. Seabrook, H. Zheng, D. W. Smith, S. Graham et al., Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the beta-amyloid precursor protein, Neuroscience, vol.90, issue.1, pp.1-13, 1999.

L. Dumery, F. Bourdel, Y. Soussan, A. Fialkowsky, S. Viale et al., beta-Amyloid protein aggregation: its implication in the physiopathology of Alzheimer's disease, Pathol Biol, vol.49, issue.1, pp.72-85, 2001.

F. S. Esch, P. S. Keim, E. C. Beattie, R. W. Blacher, A. R. Culwell et al., Cleavage of amyloid beta peptide during constitutive processing of its precursor, Science, vol.248, issue.4959, pp.1122-1126, 1990.

R. Vassar, B. D. Bennett, S. Babu-khan, S. Kahn, E. A. Mendiaz et al., Betasecretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE, Science, vol.286, issue.5440, pp.735-776, 1999.

, Les plaques amyloïdes -Site des ressources d'ACCES pour enseigner la Science de la Vie et de la Terre

J. A. Hardy and G. A. Higgins, Alzheimer's disease: the amyloid cascade hypothesis, Science, vol.256, issue.5054, pp.184-189, 1992.

E. Karran, M. Mercken, and B. D. Strooper, The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics, Nat Rev Drug Discov, vol.10, issue.9, pp.698-712, 2011.

D. R. Thal, U. Rüb, M. Orantes, and H. Braak, Phases of A beta-deposition in the human brain and its relevance for the development of AD, Neurology, vol.58, issue.12, pp.1791-800, 2002.

D. M. Mann, B. Marcyniuk, P. O. Yates, D. Neary, and J. S. Snowden, The progression of the pathological changes of Alzheimer's disease in frontal and temporal neocortex examined both at biopsy and at autopsy, Neuropathol Appl Neurobiol, vol.14, issue.3, pp.177-95, 1988.

G. G. Glenner and C. W. Wong, Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein, Biochem Biophys Res Commun, vol.122, issue.3, pp.1131-1136, 1984.

A. Alzheimer, Uber eine eigenartige Erkrankung der Hirnrinde. eitschrift fur Pshychiatrie und Physchisch-Gerichtlich Medizine, pp.146-154, 1907.

B. Permanne, C. Perez, C. Soto, B. Frangione, and T. Wisniewski, Detection of Apolipoprotein E/Dimeric Soluble Amyloid ? Complexes in Alzheimer's Disease Brain Supernatants, Biochem Biophys Res Commun, vol.240, issue.3, pp.715-735, 1997.

A. D. Snow and T. N. Wight, Proteoglycans in the pathogenesis of Alzheimer's disease and other amyloidoses, Neurobiol Aging, vol.10, issue.5, pp.481-97, 1989.

M. Stoltenberg, M. Bruhn, C. Søndergaard, P. Doering, M. J. West et al., Immersion autometallographic tracing of zinc ions in Alzheimer beta-amyloid plaques, Histochem Cell Biol, vol.123, issue.6, pp.605-616, 2005.

J. F. Collingwood, R. Chong, T. Kasama, L. Cervera-gontard, R. E. Dunin-borkowski et al., Three-dimensional tomographic imaging and characterization of iron compounds within Alzheimer's plaque core material, J Alzheimers Dis JAD, vol.14, issue.2, pp.235-280, 2008.

P. J. Whitehouse, D. L. Price, A. W. Clark, J. T. Coyle, and M. R. Delong, Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis, Ann Neurol, vol.10, issue.2, pp.122-128, 1981.

J. T. Coyle, D. L. Price, and M. R. Delong, Alzheimer's disease: a disorder of cortical cholinergic innervation, Science, vol.219, issue.4589, pp.1184-90, 1983.

J. A. Court and E. K. Perry, Dementia: The neurochemical basis of putative transmitter orientated therapy, Pharmacol Ther, vol.52, issue.3, pp.423-466, 1991.

N. R. Sims, D. M. Bowen, S. J. Allen, C. C. Smith, D. Neary et al., Presynaptic cholinergic dysfunction in patients with dementia, J Neurochem, vol.40, issue.2, pp.503-512, 1983.

E. K. Perry, R. H. Perry, P. H. Gibson, G. Blessed, and B. E. Tomlinson, A cholinergic connection between normal aging and senile dementia in the human hippocampus, Neurosci Lett, vol.6, issue.1, pp.85-94, 1977.

S. T. Dekosky, R. E. Harbaugh, F. A. Schmitt, R. A. Bakay, H. C. Chui et al., Cortical biopsy in Alzheimer's disease: diagnostic accuracy and neurochemical, neuropathological, and cognitive correlations, Intraventricular Bethanecol Study Group. Ann Neurol, vol.32, issue.5, pp.625-657, 1992.

J. Rinne, V. Kaasinen, T. Jarvenpaa, K. Nagren, A. Roivainen et al., Brain acetylcholinesterase activity in mild cognitive impairment and early Alzheimer's disease, J Neurol Neurosurg Psychiatry, vol.74, issue.1, pp.113-118, 2003.

R. Bordet and . Maladie, Alzheimer : au-delà de la seule hypothèse cholinergique -Autres systèmes de neurotransmission et modulation des récepteurs nicotiniques, vol.17, p.9, 2003.

J. Court, C. Martin-ruiz, M. Piggott, D. Spurden, M. Griffiths et al., Nicotinic receptor abnormalities in Alzheimer's disease, Biol Psychiatry, vol.49, issue.3, pp.175-84, 2001.

A. Nordberg, Nicotinic receptor abnormalities of Alzheimer's disease: therapeutic implications, Biol Psychiatry, vol.49, issue.3, pp.200-210, 2001.

R. Cacabelos, M. Takeda, and B. Winblad, The glutamatergic system and neurodegeneration in dementia: preventive strategies in Alzheimer's disease, Int J Geriatr Psychiatry, vol.14, issue.1, pp.3-47, 1999.

P. T. Francis, Glutamatergic systems in Alzheimer's disease, Int J Geriatr Psychiatry, vol.18, issue.1, pp.15-21, 2003.

R. Bullock, A. Zauner, J. J. Woodward, J. Myseros, S. C. Choi et al., Factors affecting excitatory amino acid release following severe human head injury, J Neurosurg, vol.89, issue.4, pp.507-525, 1998.

M. Schumacher, S. Camp, Y. Maulet, M. Newton, K. Macphee-quigley et al., Primary structure of Torpedo californica acetylcholinesterase deduced from its cDNA sequence, Nature, vol.319, issue.6052, pp.407-416, 1986.

M. Mesulam, A. Guillozet, P. Shaw, and B. Quinn, Widely spread butyrylcholinesterase can hydrolyze acetylcholine in the normal and Alzheimer brain, Neurobiol Dis, vol.9, issue.1, pp.88-93, 2002.

J. Birks, Cholinesterase inhibitors for Alzheimer's disease. Cochrane Database Syst Rev, p.5593, 2006.

E. Giacobini, Cholinesterases: new roles in brain function and in Alzheimer's disease, Neurochem Res, vol.28, issue.3-4, pp.515-537, 2003.

R. M. Lane, S. G. Potkin, and A. Enz, Targeting acetylcholinesterase and butyrylcholinesterase in dementia, Int J Neuropsychopharmacol, vol.9, issue.1, pp.101-125, 2006.

T. Darreh-shori, O. Almkvist, Z. Z. Guan, A. Garlind, B. Strandberg et al., Sustained cholinesterase inhibition in AD patients receiving rivastigmine for 12 months, Neurology, vol.59, issue.4, pp.563-72, 2002.

. Tacrine, Un médicament dépassé à rejeter, Rev. Prescrire, vol.18, pp.736-738, 1998.

Y. Masuda, Cardiac effect of cholinesterase inhibitors used in Alzheimer's disease--from basic research to bedside, Curr Alzheimer Res, vol.1, issue.4, pp.315-336, 2004.

G. Th, H. Lôo, and . Mémantine, Ebixa®) : une nouvelle stratégie thérapeutique dans le traitement des formes modérément sévères à sévères de la maladie d'Alzheimer. L'Encéphale, vol.30, pp.69-79, 2004.

C. Tan, Y. Wang, H. Tan, M. Meng, X. Wang et al., Efficacy and Safety of Donepezil, Galantamine, Rivastigmine, and Memantine for the Treatment of Alzheimer's Disease: A Systematic Review and Meta-Analysis, J Alzheimers Dis, vol.41, issue.2, pp.615-646, 2014.

D. Knez, N. Coquelle, A. Pi?lar, S. ?akelj, M. Juki? et al., Multi-targetdirected ligands for treating Alzheimer's disease: Butyrylcholinesterase inhibitors displaying antioxidant and neuroprotective activities, Eur J Med Chem, vol.156, pp.598-617, 2018.

J. Reis, F. Cagide, M. E. Valencia, J. Teixeira, D. Bagetta et al., Multi-targetdirected ligands for Alzheimer's disease: Discovery of chromone-based monoamine oxidase/cholinesterase inhibitors, Eur J Med Chem, vol.158, pp.781-800, 2018.

H. Liu, L. Wang, W. Su, and X. Xie, Advances in recent patent and clinical trial drug development for Alzheimer's disease. Pharm Pat Anal, vol.3, pp.429-476, 2014.

E. Giacobini, X. Zhu, W. E. Sherman, and K. A. , The effect of the selective reversible acetylcholinesterase inhibitor E2020 on extracellular acetylcholine and biogenic amine levels in rat cortex, Neuropharmacology, vol.35, issue.2, pp.205-216, 1996.

Z. Wang, P. Cai, Q. Liu, D. Xu, Y. Wu et al., Rational modification of donepezil as multifunctional acetylcholinesterase inhibitors for the treatment of Alzheimer's disease, Eur J Med Chem, vol.123, pp.282-97, 2016.

, Acetylcholine receptors: muscarinic and nicotinic | CME at Pharmacology Corner

D. B. Vieira and L. F. Gamarra, Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier, Int J Nanomedicine, vol.11, pp.5381-414, 2016.

B. Cribier, Particularités anatomiques et histologiques du nez

J. M. Klossek, La physiologie naso-sinusienne, Rev Fr Allergol Immunol Clin, vol.38, issue.7, pp.579-83, 1998.

. Blausen, com staff, staff B com. Medical gallery of Blausen Medical, WikiJournal Med, vol.1, issue.2, p.10, 2014.

I. I. Frey, . Wh, J. Liu, and X. Chen, Intranasal delivery of 125 I-NGF to the brain via the olfactory route, Research advances in Alzheimer's disease and related disorders, pp.329-335, 1995.

L. Kozlovskaya, M. Abou-kaoud, and D. Stepensky, Quantitative analysis of drug delivery to the brain via nasal route, J Controlled Release, vol.189, pp.133-173, 2014.

E. Samaridou and M. J. Alonso, Nose-to-brain peptide delivery -The potential of nanotechnology, Bioorg Med Chem, vol.26, issue.10, pp.2888-905, 2018.

D. F. Proctor, I. Andersen, and G. R. Lundqvist, Human nasal mucosal function at controlled temperatures, Respir Physiol, vol.30, issue.1, pp.109-133, 1977.

H. Wu, K. Hu, and X. Jiang, From nose to brain: understanding transport capacity and transport rate of drugs, Expert Opin Drug Deliv, vol.5, issue.10, pp.1159-68, 2008.

A. Cuschieri, Enzyme histochemistry of the olfactory mucosa and vomeronasal organ in the mouse, J Anat, vol.118, pp.477-89, 1974.

N. Moniaux, F. Escande, N. Porchet, J. P. Aubert, and S. K. Batra, Structural organization and classification of the human mucin genes, Front Biosci J Virtual Libr, vol.6, pp.1192-1206, 2001.

X. Yang and H. Nauwynck, Interactions of pseudorabies virus and swine influenza virus with porcine respiratory mucus, 2015.

D. F. Proctor, I. Andersen, and G. Lundqvist, Clearance of Inhaled Particles From the Human Nose, Arch Intern Med, vol.131, issue.1, pp.132-141, 1973.

C. A. Lipinski, F. Lombardo, B. W. Dominy, and P. J. Feeney, Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings, Adv Drug Deliv Rev, vol.23, issue.1, pp.3-25, 1997.

D. F. Veber, S. R. Johnson, H. Cheng, B. R. Smith, K. W. Ward et al., Molecular properties that influence the oral bioavailability of drug candidates, J Med Chem, vol.45, issue.12, pp.2615-2638, 2002.

P. Européenne, , pp.940-942

, Formes pharmaceutiques transmucosales -1.4 -Les formes de la voie nasale. In: 9e Édition de la Pharmacopée Européenne

, Physiologie et physiopathologie de l'épuration du mucus des voies aériennes -Encyclopédie médicale Medix

K. Inthavong, Z. F. Tian, J. Y. Tu, W. Yang, and C. Xue, Optimising nasal spray parameters for efficient drug delivery using computational fluid dynamics, Comput Biol Med, vol.38, issue.6, pp.713-739, 2008.

K. Inthavong, Q. Ge, C. Se, W. Yang, and J. Y. Tu, Simulation of sprayed particle deposition in a human nasal cavity including a nasal spray device, J Aerosol Sci, vol.42, issue.2, pp.100-113, 2011.

K. Edsman, J. Carlfors, and R. Petersson, Rheological evaluation of poloxamer as an in situ gel for ophthalmic use, Eur J Pharm Sci, vol.6, issue.2, pp.105-112, 1998.

S. Salatin, J. Barar, M. Barzegar-jalali, K. Adibkia, M. A. Milani et al., Hydrogel nanoparticles and nanocomposites for nasal drug/vaccine delivery, Arch Pharm Res, vol.39, issue.9, pp.1181-92, 2016.

J. Renukuntla, A. D. Vadlapudi, A. Patel, S. Boddu, and A. K. Mitra, Approaches for enhancing oral bioavailability of peptides and proteins, Int J Pharm, vol.447, issue.1-2, pp.75-93, 2013.

O. Witcherle, Hydrophilic Gels for Biological Use, vol.185, 1960.

J. Kope?ek and Y. J. Revie, Hydrogels as smart biomaterials, vol.56, 2007.

S. Van-vlierberghe, P. Dubruel, and E. Schacht, Biopolymer-based hydrogels as scaffolds for tissue engineering applications: a review, Biomacromolecules, vol.12, issue.5, pp.1387-408, 2009.

A. S. Hoffman, Hydrogels for biomedical applications, vol.54, pp.3-12, 2002.

N. Dahl and N. Mygind, Anatomy, physiology and function of the nasal cavities in health and disease. Adv Drug Deliv Rev, vol.29, pp.3-12, 1998.

A. Patel, K. Mequanint, and . Biomaterials, Biomed Eng -Front Chall

T. R. Hoare and D. S. Kohane, Hydrogels in drug delivery: Progress and challenges. Polymer, vol.49, pp.1993-2007, 2008.

T. H. Shubhra, Q. Toth, J. Gyenis, J. Feczkó, and T. , Poloxamers for Surface Modification of Hydrophobic Drug Carriers and Their Effects on Drug Delivery, vol.54, 2014.

R. Hitesh and P. Rakesh, Poloxamers: A pharmaceutical excipients with therapeutic behaviors, Int J PharmTech Res, vol.1, issue.2, pp.299-303, 2009.

S. D. Singh-joy and V. C. Mclain, poloxamer 105 benzoate, and poloxamer 182 dibenzoate as used in cosmetics, Int J Toxicol, vol.105, issue.2, pp.93-128, 2008.

A. Lucia, A. C. Toloza, E. Guzmán, F. Ortega, and R. G. Rubio, Novel polymeric micelles for insect pest control: encapsulation of essential oil monoterpenes inside a triblock copolymer shell for head lice control, PeerJ, vol.5, p.3171, 2017.

G. Dumortier, J. L. Grossiord, F. Agnely, and J. C. Chaumeil, A Review of Poloxamer 407 Pharmaceutical and Pharmacological Characteristics, Pharm Res, vol.23, issue.12, pp.2709-2737, 2006.

S. Mansuri, P. Kesharwani, K. Jain, R. K. Tekade, and N. K. Jain, Mucoadhesion: A promising approach in drug delivery system, React Funct Polym, vol.100, pp.151-72, 2016.

G. P. Andrews, L. Donnelly, D. S. Jones, R. M. Curran, R. J. Morrow et al., Characterization of the rheological, mucoadhesive, and drug release properties of highly structured gel platforms for intravaginal drug delivery, Biomacromolecules, vol.10, issue.9, pp.2427-2462, 2009.

J. D. Smart, The basics and underlying mechanisms of mucoadhesion, Adv Drug Deliv Rev, vol.57, issue.11, pp.1556-68, 2005.

G. S. Asane, S. A. Nirmal, K. B. Rasal, A. A. Naik, M. S. Mahadik et al., Polymers for mucoadhesive drug delivery system: a current status, Drug Dev Ind Pharm, vol.34, issue.11, pp.1246-66, 2008.

I. A. Sogias, A. C. Williams, and V. V. Khutoryanskiy, Why is chitosan mucoadhesive?, Biomacromolecules, vol.9, issue.7, pp.1837-1879, 2008.

S. Rossi, F. Ferrari, M. C. Bonferoni, and C. Caramella, Characterization of chitosan hydrochloride-mucin interaction by means of viscosimetric and turbidimetric measurements, Eur J Pharm Sci Off J Eur Fed Pharm Sci, vol.10, issue.4, pp.251-258, 2000.

M. H. Mahdi, B. R. Conway, and A. M. Smith, Development of mucoadhesive sprayable gellan gum fluid gels, Int J Pharm, vol.488, issue.1-2, pp.12-21, 2015.

A. Bacon, J. Makin, P. J. Sizer, I. Jabbal-gill, M. Hinchcliffe et al., Carbohydrate Biopolymers Enhance Antibody Responses to Mucosally Delivered Vaccine Antigens, Infect Immun, vol.68, issue.10, pp.5764-70, 2000.

B. S. Pattni, V. V. Chupin, and V. P. Torchilin, New Developments in Liposomal Drug Delivery, Chem Rev, vol.115, pp.10938-66, 2015.

M. K. Riaz, M. A. Riaz, X. Zhang, C. Lin, K. H. Wong et al., Surface Functionalization and Targeting Strategies of Liposomes in Solid Tumor Therapy: A Review, Int J Mol Sci, vol.19, issue.1, 2009.

G. Rassu, E. Soddu, M. Cossu, A. Brundu, G. Cerri et al., Solid microparticles based on chitosan or methyl-?-cyclodextrin: a first formulative approach to increase the nose-to-brain transport of deferoxamine mesylate, J Control Release Off J Control Release Soc, vol.201, pp.68-77, 2015.

E. Muntimadugu, R. Dhommati, A. Jain, V. Challa, M. Shaheen et al., Intranasal delivery of nanoparticle encapsulated tarenflurbil: A potential brain targeting strategy for Alzheimer's disease, Eur J Pharm Sci Off J Eur Fed Pharm Sci, vol.92, pp.224-258, 2016.

X. Zheng, X. Shao, C. Zhang, Y. Tan, Q. Liu et al., Intranasal H102 Peptide-Loaded Liposomes for Brain Delivery to Treat Alzheimer's Disease, Pharm Res, vol.32, issue.12, pp.3837-3886, 2015.

R. Narayan, M. Singh, O. Ranjan, Y. Nayak, S. Garg et al., Development of risperidone liposomes for brain targeting through intranasal route, Life Sci, vol.163, pp.38-45, 2016.

I. A. Alsarra, A. Y. Hamed, and F. K. Alanazi, Acyclovir Liposomes for Intranasal Systemic Delivery: Development and Pharmacokinetics Evaluation. Drug Deliv, vol.15, pp.313-334, 2008.

S. L. Law, K. J. Huang, and H. Y. Chou, Preparation of desmopressin-containing liposomes for intranasal delivery, J Controlled Release, vol.70, issue.3, pp.375-382, 2001.

A. K. Jain, K. B. Chalasani, R. K. Khar, F. J. Ahmed, and P. V. Diwan, Muco-adhesive multivesicular liposomes as an effective carrier for transmucosal insulin delivery, J Drug Target, vol.15, issue.6, pp.417-444, 2007.

M. L. Pinzón-daza, I. Campia, J. Kopecka, R. Garzón, D. Ghigo et al., Nanoparticle-and liposome-carried drugs: new strategies for active targeting and drug delivery across blood-brain barrier, Curr Drug Metab, vol.14, issue.6, pp.625-665, 2013.

A. D. Bangham, M. M. Standish, and J. C. Watkins, Diffusion of univalent ions across the lamellae of swollen phospholipids, J Mol Biol, vol.13, issue.1, pp.238-52, 1965.

I. Motta, , 2015.

B. De-kruijff, P. R. Cullis, and G. K. Radda, Outside-inside distributions and sizes of mixed phosphatidylcholine-cholesterol vesicles, Biochim Biophys Acta BBA -Biomembr, vol.436, issue.4, pp.729-769, 1976.

S. C. Semple, A. Chonn, and P. R. Cullis, Influence of cholesterol on the association of plasma proteins with liposomes, Biochemistry, vol.35, issue.8, pp.2521-2526, 1996.

M. S. El-samaligy, N. N. Afifi, and E. A. Mahmoud, Increasing bioavailability of silymarin using a buccal liposomal delivery system: preparation and experimental design investigation, Int J Pharm, vol.308, issue.1-2, pp.140-148, 2006.

H. Rongen, A. Bult, and W. P. Van-bennekom, Liposomes and immunoassays, J Immunol Methods, vol.204, issue.2, pp.105-138, 1997.

A. Sharma and U. S. Sharma, Liposomes in drug delivery: Progress and limitations, Int J Pharm, vol.154, issue.2, pp.123-163, 1997.

J. Stetefeld, S. A. Mckenna, and T. R. Patel, Dynamic light scattering: a practical guide and applications in biomedical sciences, Biophys Rev, vol.8, issue.4, pp.409-436, 2016.

V. P. Torchilin, Recent advances with liposomes as pharmaceutical carriers, Nat Rev Drug Discov, vol.4, issue.2, pp.145-60, 2005.

T. J. Franz, Percutaneous absorption on the relevance of in vitro data, J Invest Dermatol, vol.64, issue.3, pp.190-195, 1975.

J. Lindemann, R. Leiacker, G. Rettinger, and T. Keck, Nasal mucosal temperature during respiration, Clin Otolaryngol Allied Sci, vol.27, issue.3, pp.135-144, 2002.

L. Dombu-youta and C. , Utilisation de nanoparticules pour délivrer des protéines dans les épithéliums respiratoires : caractérisation des mécanismes impliqués, 2012.

G. L. Ellman, K. D. Courtney, V. Andres, and R. M. Featherstone, A new and rapid colorimetric determination of acetylcholinesterase activity, Biochem Pharmacol, vol.7, issue.2, pp.88-95, 1961.

M. L. Bolognesi, V. Andrisano, M. Bartolini, R. Banzi, and C. Melchiorre, Propidiumbased polyamine ligands as potent inhibitors of acetylcholinesterase and acetylcholinesterase-induced amyloid-beta aggregation, J Med Chem, vol.48, issue.1, pp.24-31, 2005.

O. Soga, C. F. Van-nostrum, M. Fens, C. Rijcken, R. M. Schiffelers et al., Thermosensitive and biodegradable polymeric micelles for paclitaxel delivery, J Control Release Off J Control Release Soc, vol.103, issue.2, pp.341-53, 2005.

S. R. Raghavan and B. H. Cipriano, Gel formation: phase diagrams using tabletop rheology and calorimetry, Molecular Gels, pp.241-252, 2006.

G. Bastiat, C. O. Pritz, C. Roider, F. Fouchet, E. Lignières et al., A new tool to ensure the fluorescent dye labeling stability of nanocarriers: A real challenge for fluorescence imaging, J Controlled Release, vol.170, issue.3, pp.334-376, 2013.

A. Martinac, J. Filipovi?-grci?, D. Voinovich, B. Perissutti, and E. Franceschinis, Development and bioadhesive properties of chitosan-ethylcellulose microspheres for nasal delivery, Int J Pharm, vol.291, issue.1-2, pp.69-77, 2005.

G. G. Pereira, F. A. Dimer, S. S. Guterres, C. P. Kechinski, J. E. Granada et al., Formulation and characterization of poloxamer 407®: thermoreversible gel containing polymeric microparticles and hyaluronic acid, Quím Nova, vol.36, issue.8, pp.1121-1125, 2013.

J. Wu, Y. Tian, S. Wang, M. Pistolozzi, Y. Jin et al., Design, synthesis and biological evaluation of bambuterol analogues as novel inhibitors of butyrylcholinesterase, Eur J Med Chem, vol.126, pp.61-71, 2017.

J. J. Buccafusco, Cognitive Enhancing Drugs. Birkhäuser, vol.213, 2012.

S. N. Dighe, G. S. Deora, E. De-la-mora, F. Nachon, S. Chan et al., Discovery and Structure-Activity Relationships of a Highly Selective Butyrylcholinesterase Inhibitor by Structure-Based Virtual Screening, J Med Chem, vol.59, issue.16, pp.7683-7692, 201625.

K. Augustinsson, T. Bartfai, and B. Mannervik, A steady-state kinetic model of butyrylcholinesterase from horse plasma, Biochem J, vol.141, issue.3, pp.825-859, 1974.

H. Sun, K. Nguyen, E. Kerns, Z. Yan, K. R. Yu et al., Highly Predictive and Interpretable Models for PAMPA Permeability, Bioorg Med Chem, vol.25, issue.3, pp.1266-76, 2017.

T. Terasaki, S. Ohtsuki, S. Hori, H. Takanaga, E. Nakashima et al., New approaches to in vitro models of blood-brain barrier drug transport, Drug Discov Today, vol.8, issue.20, pp.944-54, 2003.

G. L. Amidon, H. Lennernäs, V. P. Shah, and J. R. Crison, A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability, Pharm Res, vol.12, issue.3, pp.413-433, 1995.

G. Center, The effect of central insulin on insulin sensitivity and energy metabolism (Centrin), ClinicalTrials.gov Identifier: NCT01479075

J. Freiherr, M. Hallschmid, W. H. Frey, Y. F. Brünner, C. D. Chapman et al., Intranasal Insulin as a Treatment for Alzheimer's Disease: A Review of Basic Research and Clinical Evidence, CNS Drugs, vol.27, issue.7, pp.505-519, 2013.

I. Gozes, B. H. Morimoto, J. Tiong, A. Fox, K. Sutherland et al., NAP: research and development of a peptide derived from activity-dependent neuroprotective protein (ADNP), CNS Drug Rev, vol.11, issue.4, pp.353-68, 2005.

Y. Ozsoy and S. Güngör, Nasal route: an alternative approach for antiemetic drug delivery, Expert Opin Drug Deliv, vol.8, issue.11, pp.1439-53, 2011.

P. N. Schlegel and H. Study-group, Efficacy and safety of histrelin subdermal implant in patients with advanced prostate cancer, J Urol, vol.175, issue.4, pp.1353-1361, 2006.

F. U. Momoh, J. S. Boateng, S. Richardson, B. Z. Chowdhry, and J. C. Mitchell, Development and functional characterization of alginate dressing as potential protein delivery system for wound healing, Int J Biol Macromol, vol.81, pp.137-50, 2015.

P. C. Bessa, M. Casal, and R. L. Reis, Bone morphogenetic proteins in tissue engineering: the road from laboratory to clinic, part II (BMP delivery), J Tissue Eng Regen Med, vol.2, issue.2-3, pp.81-96, 2008.

R. Thorn, J. Greenman, and A. Austin, An in vitro study of antimicrobial activity and efficacy of iodine-generating hydrogel dressings, J Wound Care, vol.15, issue.7, pp.305-315, 2006.

J. Su, B. Hu, W. L. Lowe, D. B. Kaufman, and P. B. Messersmith, Anti-Inflammatory Peptide Functionalized Hydrogels for Insulin-Secreting Cell Encapsulation, Biomaterials, vol.31, issue.2, pp.308-322, 2010.

J. M. Barichello, M. Morishita, K. Takayama, and T. Nagai, Absorption of insulin from pluronic F-127 gels following subcutaneous administration in rats, Int J Pharm, vol.184, issue.2, pp.189-98, 1999.

Y. Liu, Y. Zhu, G. Wei, and W. Lu, Effect of carrageenan on poloxamer-based in situ gel for vaginal use: Improved in vitro and in vivo sustained-release properties, Eur J Pharm Sci Off J Eur Fed Pharm Sci, vol.37, issue.3-4, pp.306-318, 2009.

P. Mura, N. Mennini, C. Nativi, and B. Richichi, In situ mucoadhesive-thermosensitive liposomal gel as a novel vehicle for nasal extended delivery of opiorphin, Eur J Pharm Biopharm Off J Arbeitsgemeinschaft Pharm Verfahrenstechnik EV, vol.122, pp.54-61, 2018.

H. Choi, J. Ryu, J. Yoon, S. Oh, Y. Kim et al., Development of in situ-gelling and mucoadhesive acetaminophen liquid suppository, Int J Pharm, vol.165, issue.1, pp.33-44, 1998.

M. Kapoor, J. C. Cloyd, and R. A. Siegel, A review of intranasal formulations for the treatment of seizure emergencies, J Controlled Release, vol.237, pp.147-59, 2016.

Y. Feng, H. He, F. Li, Y. Lu, J. Qi et al., An update on the role of nanovehicles in nose-to-brain drug delivery, Drug Discov Today, vol.23, issue.5, pp.1079-88, 2018.

A. Laouini, C. Jaafar-maalej, I. Limayem-blouza, S. Sfar, C. Charcosset et al., Preparation, Characterization and Applications of Liposomes: State of the Art, J Colloid Sci Biotechnol, vol.1, issue.2, pp.147-68, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02002202

G. Bozzuto and A. Molinari, Liposomes as nanomedical devices, Int J Nanomedicine, vol.10, pp.975-99, 2015.

U. Bulbake, S. Doppalapudi, N. Kommineni, and W. Khan, Liposomal Formulations in Clinical Use: An Updated Review, Pharmaceutics, vol.9, issue.2, 2017.

A. Sahebkar, A. Badiee, M. Ghayour-mobarhan, S. R. Goldouzian, and M. R. Jaafari, A simple and effective approach for the treatment of dyslipidemia using anionic nanoliposomes, Colloids Surf B Biointerfaces, vol.122, pp.645-52, 2014.

D. Sotto, A. Paolicelli, P. Nardoni, M. Abete, L. Garzoli et al., SPC Liposomes as Possible Delivery Systems for Improving Bioavailability of the Natural Sesquiterpene ?-Caryophyllene: Lamellarity and Drug-Loading as Key Features for a Rational Drug Delivery Design, Pharmaceutics, vol.10, issue.4, 2018.

E. Ahmad, Y. Feng, J. Qi, W. Fan, Y. Ma et al., Evidence of nose-to-brain delivery of nanoemulsions: cargoes but not vehicles, vol.9, pp.1174-83, 2017.

A. Mistry, S. Stolnik, and L. Illum, Nanoparticles for direct nose-to-brain delivery of drugs, Int J Pharm, vol.379, issue.1, pp.146-57, 2009.

E. E. Morrison and R. M. Costanzo, Morphology of olfactory epithelium in humans and other vertebrates, Microsc Res Tech, vol.23, issue.1, pp.49-61, 1992.

M. M. Wen, Olfactory targeting through intranasal delivery of biopharmaceutical drugs to the brain: current development, Discov Med, vol.11, issue.61, pp.497-503, 2011.

A. S. Hanafy, R. M. Farid, and S. S. Elgamal, Complexation as an approach to entrap cationic drugs into cationic nanoparticles administered intranasally for Alzheimer's disease management: preparation and detection in rat brain, Drug Dev Ind Pharm, vol.41, issue.12, pp.2055-68, 2015.

S. K. Lai, J. S. Suk, A. Pace, Y. Wang, Y. M. Mert et al., Drug carrier nanoparticles that penetrate human chronic rhinosinusitis mucus, Biomaterials, vol.32, issue.26, pp.6285-90, 2011.

M. Van-woensel, N. Wauthoz, R. Rosière, K. Amighi, V. Mathieu et al., Formulations for Intranasal Delivery of Pharmacological Agents to Combat Brain Disease: A New Opportunity to Tackle GBM? Cancers, vol.5, pp.1020-1068, 2013.

K. Taylor, G. Taylor, I. W. Kellaway, and J. Stevens, Drug entrapment and release from multilamellar and reverse-phase evaporation liposomes, Int J Pharm, vol.58, issue.1, pp.49-55, 1990.

J. Li, X. Wang, T. Zhang, C. Wang, Z. Huang et al., A review on phospholipids and their main applications in drug delivery systems, Asian J Pharm Sci, vol.10, issue.2, pp.81-98, 2015.

X. Xu, M. A. Khan, and D. J. Burgess, A quality by design (QbD) case study on liposomes containing hydrophilic API: II. Screening of critical variables, and establishment of design space at laboratory scale, Int J Pharm, vol.423, issue.2, pp.543-53, 2012.

X. Xu, M. A. Khan, and D. J. Burgess, Predicting hydrophilic drug encapsulation inside unilamellar liposomes, Int J Pharm, vol.423, issue.2, pp.410-418, 2012.

E. Ruel-gariepy, G. Leclair, P. Hildgen, A. Gupta, and J. Leroux, Thermosensitive chitosan-based hydrogel containing liposomes for the delivery of hydrophilic molecules, J Controlled Release, vol.82, issue.2, pp.373-383, 2002.

S. Mourtas, S. Fotopoulou, S. Duraj, V. Sfika, C. Tsakiroglou et al., Liposomal drugs dispersed in hydrogels. Effect of liposome, drug and gel properties on drug release kinetics, Colloids Surf B Biointerfaces, vol.55, issue.2, pp.212-233, 2007.

M. Ruigrok, D. Lange, and E. , Emerging Insights for Translational Pharmacokinetic and Pharmacokinetic-Pharmacodynamic Studies: Towards Prediction of Nose-to-Brain Transport in Humans, AAPS J, vol.17, issue.3, pp.493-505, 2015.

M. Briuglia, C. Rotella, A. Mcfarlane, and D. A. Lamprou, Influence of cholesterol on liposome stability and on in vitro drug release, Drug Deliv Transl Res, vol.5, issue.3, pp.231-273, 2015.

R. G. Thorne and W. H. Frey, Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations, Clin Pharmacokinet, vol.40, issue.12, pp.907-953, 2001.

R. G. Thorne, C. R. Emory, T. A. Ala, and W. H. Frey, Quantitative analysis of the olfactory pathway for drug delivery to the brain, Brain Res, vol.692, issue.1-2, pp.278-82, 1995.

C. Karavasili and D. G. Fatouros, Smart materials: in situ gel-forming systems for nasal delivery, Drug Discov Today, vol.21, issue.1, pp.157-66, 2016.

J. Ds, B. An, A. Rb, S. Ss, A. Pn et al., Thermosensitive PLA based nanodispersion for targeting brain tumor via intranasal route, Mater Sci Eng C Mater Biol Appl, vol.63, pp.411-432, 2016.

S. Vaka, S. M. Sammeta, L. B. Day, and S. N. Murthy, Delivery of nerve growth factor to brain via intranasal administration and enhancement of brain uptake, J Pharm Sci, vol.98, issue.10, pp.3640-3646, 2009.

S. Vaka, S. N. Murthy, A. Balaji, and M. A. Repka, Delivery of brain-derived neurotrophic factor via nose-to-brain pathway, Pharm Res, vol.29, issue.2, pp.441-448, 2012.

. Fda, Safety Evaluation of Reformulated Drug Products and Products Intended for Administration by an Alternate Route -Guidance for Industry and Review Staff Good Review Practice, Pharmacology/Toxicology, 2015.