R. , J. , C. Hatziantoniou, S. Demetzos, C. Harendra et al., Nanoliposomes and their applications in food nanotechnology [3] Bangham, A.D. A correlation between surface charge and coagulant action of phospholipids Production and Characterization of Liposome Systems for Pharmaceutical Applications Kinetic and thermodynamic effects in the formation of amphiphilic colloidal particles, Mortazavi M.S. Nanoliposomes: From Fundamentals to Recent Developments Liposomal nanocapsules in food science and agriculture, pp.902-912, 1961.

R. Langer, J. M. Metselaar, P. Bruin, L. W. De-boer, T. De-vringer et al., A novel family of L-amino acid-based biodegradable polymer-lipid conjugates for the development of long-circulating liposomes with effective drug-targeting capacity [10] Cevc, G. Transfersomes, liposomes and other lipid suspensions on the skin: Permeation enhancement, vesicle penetration, and transdermal drug delivery From bugs to drugs: Therapeutic immunomodulation with oligodeoxynucleotides containing CpG sequences from bacterial DNA Recent trends in the lipid-based nanoencapsulation of antioxidants and their role in foods, Biodegradable long-circulating polymeric nanospheres. Sci. Cult, pp.1600-1603, 1994.

C. Garti14-]-tardi, M. Drechsler, K. H. Bauer, M. Brandl, and R. J. Mannino, Steam sterilisation of vesicular phospholipid gels. Inter Cochleates: New lipidbased drug delivery system, J. Pharm. J.R.; Perlin, D J. Liposome Res, vol.21716, issue.104, pp.161-172, 2000.

L. Zarif, Elongated supramolecular assemblies in drug delivery, Journal of Controlled Release, vol.81, issue.1-2, pp.7-23, 2002.
DOI : 10.1016/S0168-3659(02)00010-X

M. R. Mozafari, N. Nanomaterials, N. J. For-biomedical-applications-clifton, L. Ed-aarons, K. Petrak et al., The Netherlands Efficiency of drug targeting: steady-state considerations using a three-compartment model Prodrugs and site-specific drug delivery, [21] Samuni, A.M.; Barenholz, Y. In: Liposomes, pp.29-50, 1980.

N. Düzgünee, E. Shah, S. P. Misra, and A. , Liposomal amikacin dry powder inhaler: effect of fines on in vitro performance, AAPS Pharm.Sci.Tech, vol.5, issue.4, pp.299-314, 2004.

T. Selhorst, P. Schuster, A. Vos, and U. Wenzel, Kinetics of maternal immunity against rabies in fox cubs

G. Lopez-berestein, R. Mehta, R. L. Hopfer, G. Lopez-berestein, V. Fainstein et al., Treatment and prophylaxis of disseminated infection due to Candida albicans in mice with liposomeencapsulated amphotericin B Liposomal amphotericin B for the treatment of systemic fungal infections in patients with cancer: A preliminary study Enhanced intracellular killing of Staphylococcus aureus by canine monocytes treated with liposomes containing amikacin, gentamicin, kanamycin, and tobramycin, Cullis, P.R. Techniques for encapsulating bioactive agents into liposomes, pp.11-16, 1981.

A. Bodor and J. Kutas, Liposomes in gene therapy, Acta. Pharm. Hung, vol.73, issue.2, pp.115-122, 2003.

J. Manosroi, D. D. Lasic, D. Papahadjopoulos, A. C. Allison, G. Gregoriadis et al., Vaccination with BLP25 liposome vaccine to treat nonsmall cell lung and prostate cancers Medical application of liposomes Liposomes as immunological adjuvants Protein-liposome conjugates using cysteine-lipids and native chemical ligation, Arzneim-Forsch. Expert. Re. Vaccin. Nature. Bioconjug. Chem, vol.4, issue.18, pp.58-485, 1974.

R. Aitken, R. Watkins, and B. C. Keller, Applications and implications of nanotechnologies for the food sector Liposomes in nutrition Use of liposomes for proteinase addition to Cheddar cheese, Food Addit. Contam. Trend. Food Sci. Tech. J. Dairy Res, vol.2537, issue.52, pp.241-258, 1985.

J. Magee, E. L. Olson, N. F. March, S. Shi, W. Rao et al., Microencapsulation of cheese ripening systems: Stability of microcapsules Liposomes: From concept to application. Food New Zealand FEB Microscopical investigations of nisin-loaded nanoliposomes prepared by Mozafari method and their bacterial targeting Acceleration of cheddar cheese lipolysis by using liposome-entrapped lipases Accelerated Cheddar cheese ripening with encapsulated proteinases Seung-Cheol-Lee, S. Encapsulation of bromelain in liposome The effect of cholesterol content of phospholipid vesicles on the encapsulation and acid resistance of Îgalactosidase from E. coli. Inter Liposomal encapsulation of beta-galactosidase: Comparison of two methods of encapsulation and in vitro lactose digestibility, J. Dairy Sci Micron. J. Food Sci. Int. J. Food Sci. Technol. J. Food Sci. Nutr. J. Food Sci. Technol. C.B. J. Food Biochem, vol.64, issue.184, pp.611-615, 1981.

M. A. Livreaa, Synergistic Interactions between Vitamin A and Vitamin

C. Banville, J. C. Vuillemard, C. Lacroix, R. Van-der-meer, C. G. De-kruif et al., Microencapsulation: Its application in nutrition Food applications of liposome-encapsulated antimicrobial peptides. Trend. Food Sci [49] Kirby, C. Microencapsulation and controlled delivery of food ingredients. Food Sci Influence of beef tallow and muscle on the antilisterial activity of pediocin AcH and liposome-encapsulated pediocin AcH Fliss, I. Impact of nisin producing culture and liposome-encapsulated nisin on ripening of Cactobacillus added- Cheddar cheese Modifications of membrane phospholipid composition in nisin-resistant Listeria monocytogenes Scott A Fliss, I. Inhibition of Listeria innocua in cheddar cheese by addition of nisin Z in liposomes or by in situ production in mixed culture.; Fliss, I. Liposome encapsulated nisin Z: Optimization, stability and release during milk fermentation Liposomal delivery systems for encapsulation of ferrous sulfate: Preparation and characterization, against Lipid Peroxidation in Phosphatidylcholine Liposomes. Acta Biochem. Biophys. Size, stability, and entrapment efficiency of phospholipid nanocapsules containing polypeptide antimicrobials, pp.57-63, 1991.

A. Albaldawi, C. S. Brennan, K. Alobaidy, W. Alammar, and D. Aljumaily, Effect of flour fortification with haem liposome on bread and bread doughs, International Journal of Food Science and Technology, vol.18, issue.8, pp.40-825, 2005.
DOI : 10.1016/0309-1740(89)90005-3

P. Walde, S. Ichikawa, J. M. Rodriguez-nogales, M. Pe-rez?mateos, and M. D. Busto, Enzymes inside lipid vesicles: Preparation, reactivity and applications Application of experimental design to the formulation of glucose oxidase encapsulation by liposomes, 60] Kellaway, I.W.; Farr, S.J. Liposomes as drug delivery systems to the lung, pp.143-177, 2001.

A. Smith, L. J. Anderson, J. Shamsuddin, M. Suntres, Z. E. Shek et al., Role of antioxidants in paraquat toxicity Incorporation of alpha-tocopherol in liposomes promotes the retention of liposome-encapsulated glutathione in the rat lung Incorporation of -tocopherol in marine lipid-based liposomes: In vitro and in vivo studies Controlled delivery of functional food ingredients: Opportunities for liposomes in the food industry Ultrastructural architecture of liposome-entrapped glutathione: A Cryo-SEM study Testing the effectiveness of nutrient delivery systems. In: Delivery and controlled release of bioactives in foods and nutraceuticals Liposomes as vehicles for enhancing drug delivery via skin routes In vivo comparison of various liposome formulations for cosmetic application marine lipidbased liposomes: Physical characterization and potential application in cosmetics . Inter Metabolism of polyunsaturated fatty acids by skin epidermal enzymes: generation of antiinflammatory and antiproliferative metabolites, Glutathione localization and distribution after intratracheal instillation: Implications for treatment 361S-366S. [72] Giacomoni, P.U. Advancement in skin aging: the future cosmeceuticals Swarnlata S. Novel approaches in herbal cosmetics vitro biological activity of gemcitabine-loaded liposomes, pp.149-161, 1990.

N. Düzgüne, I. F. Uchegbu, A. Florence, S. P. Vyas, A. Nagume et al., Non-ionic surfactant vesicles (niosomes)physical and pharmaceutical chemistry Nonionic surfactant based vesicles (niosomes) in drug delivery Electron spin resonance studies of phosphatidylcholine interacted with cholesterol and with a hopanoid in liposomal membrane Influence of cholesterol on liposome fluidity by EPR. Relationship with percutaneous absorption Intracellular targeting of sodium mercaptoundecahydrododecaborate (BSH) to solid tumors by transferrin-PEG liposomes, for boron neutron-capture therapy (BNCT) Lipid raft components cholesterol and sphingomyelin increase H +/OH-permeability of phosphatidylcholine membranes Liposome drugs' loading efficiency: A working model based on loading conditions and drug's physicochemical properties, Brazel, C.S. On the importance and mechanisms of burst release in matrix-controlled drug delivery systems, pp.40-57, 1991.

L. A. Meure, N. R. Foster, F. Dehghani, J. Lasch, V. Weissig et al., Liposomes: A practical approach Diffusion of univalent ions across the lamellae of swollen phospholipids The action of steroids and streptolysin S on the permeability of phospholipid structures to cations Single bilayer vesicles prepared without sonication: physico chemical properties Liposomes: A practical approach Single bilayer liposomes prepared without sonication, Conventional and dense gas techniques for the production of liposomes: A review Bangham, A.D. Large volume liposomes by an ether vaporization method. [92] Deamer, D.W. Preparation and properties of ether injection liposomes, pp.798-809, 1965.

C. Jaafar-maalej, R. Diab, V. Andrieu, A. Elaissari, H. Fessi et al., Ethanol injection method for hydrophilic and lipophilic drug-loaded liposome preparation, Journal of Liposome Research, vol.347, issue.3, pp.228-243, 2010.
DOI : 10.1016/j.bbrc.2006.06.152

R. Schubert, S. Förster, F. Szoka-jr, D. Papahadjopoulos, W. N. Vreeland et al., Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation Microfluidic directed formation of liposomes of controlled size Preparation of liposomal gene therapy vectors by a scalable method without using volatile solvents or detergents Development of non-toxic liposomal formulations for gene and drug delivery to the lung, Proc. Natl. Acad. Sci., USA, pp.1177-1180, 1978.

K. Anton, P. Van-hoogevest, L. Frederiksen, K. Otake, T. Imura et al., Development of a new preparation method of liposomes using supercritical carbon dioxide Methods and apparatus for making liposomes containing hydrophobic drugs Recent applications of supercritical fluid technology to pharmaceutical powder systems Preparation of liposomes encapsulating water-soluble compounds using supercritical carbon dioxide Use of supercritical carbon dioxide for preparation of pharmaceutical formulations Preparation of liposomes using an improved supercritical reverse phase evaporation method Factors involved in the production of liposomes with a high-pressure homogenizer Interactions between pH-sensitive liposomes and model membranes Dual asymmetric centrifugation (DAC)-A new technique for liposome preparation The use of transmembrane pH gradient-driven drug encapsulation in the pharmacodynamic evaluation of liposomal doxorubicin, 921-928. [105] Frederiksen 3rd International Symposium on Supercritical Fluids Liposome Technology, Liposome Preparation and Related Techniques III. [112] Brandl, M.; Massing, U. In: Liposome Technology Liposome Preparation and Related Technique Liposome Technology, Liposome Preparation and Related Techniques, pp.17-3898, 1994.

C. Li, Y. J. Deng, J. Cui, G. Gregoriadis, . Ed et al., Influence of liposome size on accumulation in tumor and therapeutic efficiency of liposomal near-IR photosensitizer for PDT based on aluminum hydroxide tetra-3-phenylthiophthalocyanine. Technical Proceedings of the NSTI Nanotechnology Conference and Trade Show Scanning Electron Microscopy: Physics of Image Formation and Microanalysis Springer Series in Optical Sciences Atomic force microscopy: A tool to study the structure, dynamics and stability of liposomal drug delivery systems A review of analytical methods for the identification and characterization of nano delivery systems in food Analysis of liposomes Gel permeation chromatography: A review Characterization of loaded liposomes by size exclusion chromatography Thermal analysis of phase transition behaviour in liposomes Different effects of long-And short-chain ceramides on the gel-fluid and lamellar-hexagonal transitions of phospholipids: A calorimetric, NMR, and x-ray diffraction study Synthesis , liposomal formulation and thermal effects on phospholipid bilayers of leuprolide Toxicity of cationic liposome-DNA complex in lung isografts Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells Entrapment of ovalbumin into liposomes -Factors affecting entrapment efficiency, liposome size, and zeta potential The influence of temperature, cholesterol content and pH on liposome stability Mechanism for enhancement effect of lipid disperse system on percutaneous absorption Calcein permeation across phosphatidylcholine bilayer membrane: Effects of membrane fluidity, liposome size, and immobilization Production of large unilamellar vesicles by a rapid extrusion procedure. Characterization of size distribution, trapped volume and ability to maintain a membrane potential, Liposome Technology Liposome Preparation and Related Techniques 43-50. [125] Nagahiro Korn, E.D. Single bilayer liposomes Cullis, P.R. Vesicles of variable sizes produced by a rapid extrusion procedure Release of drugs from liposomes varies with particle size, pp.35-53, 1970.

B. Ruozi, G. Tosi, E. Leo, and M. A. Vandelli, Application of atomic force microscopy to characterize liposomes as drug and gene carriers, Liposome drug-loading method and composition, pp.12-22, 1997.
DOI : 10.1016/j.talanta.2007.03.031

R. Laridi, E. E. Kheadr, R. O. Benech, J. C. Vuillermard, C. Lacroix et al., A convenient and sensitive fluorescence assay for phospholipid vesicles using diphenylhexatriene Determination of liposomal encapsulation efficiency using proton NMR spectroscopy Filter extrusion of liposomes using different devices: Comparison of liposome size, encapsulation efficiency, and process characteristics Factors affecting physicochemical properties of liposomes prepared with hydrogenated purified egg yolk lecithins by the microencapsulation vesicle method Influence of physicochemical properties of fluoroquinolones on encapsulation efficiency in liposomes Generation of contrastcarrying liposomes of defined size with a new continuous high pressure extrusion method, Macdonald, R.C. A simple procedure for the determination of the trapped volume of liposomes, pp.325-336, 1978.

K. Anzai, M. Yoshida, Y. Kirino, D. P. Hinton, . Johnson et al., Change in intravesicular volume of liposomes by freeze-thaw treatment as studied by the ESR stopped-flow technique Simultaneous measurement of vesicle diffusion coefficients and trapping efficiencies by means of diffusion ordered 2D NMR spectroscopy, Biochim. Biophys. Acta -Biomembr. Chem. Phys. Lipids, vol.1021147, issue.692, pp.21-26, 1990.

H. I. Hauser, G. Gregoriadis, and . Ed, Liposome Technology, Liposome Preparation and Related Techniques, pp.49-197, 1993.

F. Tablin, The trehalose myth revisited: introduction to a symposium on stabilization of cells in the dry state, Cryobiology, vol.43150, pp.89-105, 2001.

L. M. Crowe, C. Womersley, J. H. Crowe, D. Reid, L. Appel et al., Prevention of fusion and leakage in freeze-dried liposomes by carbohydrates, Biochim. Biophys. Acta -Biomembr, vol.151, pp.861-131, 1986.

N. I. Payne, P. Timmins, and C. V. Ambrose, Proliposomes: A Novel Solution to an Old Problem, Journal of Pharmaceutical Sciences, vol.75, issue.4, pp.75-325, 1986.
DOI : 10.1002/jps.2600750402

N. I. Payne, I. Browning, C. A. Hynes, S. Perrett, M. Golding et al., Characterization of proliposomes Sterilization of liposomes by heat treatment Characterization of liposomes Efficient method for (lyso)phospholipid class separation by high-performance liquid chromatography using an evaporative light-scattering detector Rapid determination of acyl chain position in egg phosphatidylcholine by high performance liquid chromatography/electrospray mass spectrometry A structural study of interfacial phospholipid and lung surfactant layers by transmission electron microscopy after Blodgett sampling: Influence of surface pressure and temperature [160] Düzgüne, N. Liposomes, Methods in Enzymolog [161] Lasic, D.D. Liposomes, From Physics to Applications In: Boylan Encyclopedia of Pharmaceutical Technology Dekker: USA Damage to liposomal lipids: Protection by antioxidants and cholesterol-mediated dehydration Free Radicals in Biology and Medicine Vigo-Pelfrey C. Quality control of liposomal lipids with special emphasis on peroxidation of phospholipids and cholesterol Novel functions and applications of trehalose Role of trehalose phosphate synthase and trehalose during hypoxia: From flies to mammals Role of trehalose phosphate synthase in anoxia tolerance and development in Drosophila melanogaster Low-cariogenicity of trehalose as a substrate Study of trehalose addition on aroma retention in dehydrated strawberry puree Analysis and hydrolysis kinetics of phospholipids in aqueous liposome dispersions Chemical stability of liposomes: Implications for their physical stability Stability and storage of liposomes. Optimization of Drug Delivery Hydrolysis of saturated soybean phosphatidylcholine in aqueous liposome dispersions A newly developed immunoliposome -an egg phosphatidylcholine liposome coated with pullulan bearing both a cholesterol moiety and an IgMs fragment Laser triggered drug release from magnetoliposomes Ultrasound in drug and gene delivery A mathematical model of drug release from liposomes by low frequency ultrasound, Liposomes Nanocarrier Technologies: Frontiers of Nanotherapy, pp.330-333, 1982.

J. Bacri, S. Lesieur, O. Clément, J. Lu, L. Deng et al., Preparation and characterization of magnetic cationic liposome in gene delivery Development of monodispersed and functional magnetic polymeric liposomes via simple liposome method Fabrication of magnetic nanocomposite spheres for targeted drug delivery ASME International Mechanical Engineering Congress and Exposition: Proceeding Recent advances in nanoneurology for drug delivery to the brain Synthesis, characterization, and controllable drug release of pH-sensitive hybrid magnetic nanoparticles Liposomes from novel photolabile phospholipids: Light-induced unloading of small molecules as monitored by PFG NMR Visible light-induced destabilization of endocytosed liposomes Photoregulated potassium ion permeation through dihexadecyl phosphate bilayers containing azobenzene and stilbene surfactants, Magnetic targeting of magnetoliposomes to solid tumors with MR imaging monitoring in mice: Feasibility. Radiology. [193] Huth, U.S.; Schubert, R.; Peschka-Suss, R. Investigating the uptake and intracellular fate of pH-sensitive liposomes by flow cytometry and spectral bio-imaging, pp.415-424, 0194.

D. H. Hee, W. K. Tae, C. S. Byung, and S. C. Ho, Release of calcein from temperature-sensitive liposomes in a poly(N-isopropylacrylamide) hydrogel, Thermosensitive polymer-modified liposomes, pp.54-61, 0196.

K. Kono and T. Takagishi, Temperature-Sensitive Liposomes, Methods Enzymol, vol.387, pp.73-82, 0197.
DOI : 10.1016/S0076-6879(04)87005-8

J. Li, B. Wang, P. Liu, L. Nobs, F. Buchegger et al., Possibility of active targeting to tumor by local hyperthermia with temperature-sensitive nanoparticles Current methods for attaching targeting ligands to liposomes and nanoparticles Nanosystems in drug targeting: opportunities and challenges Essential properties of drug-targeting delivery systems, Med. Hypotheses J. Pharm. Sci. Curr. Nanosci. Drug Discov. Today, vol.71, issue.10, pp.249-521, 2004.

J. D. Byrne, T. Betancourt, L. Brannon-peppas, J. D. Almeida, C. M. Brand et al., Active targeting schemes for nanoparticle systems in cancer therapeutics Formation of virosomes from influenza subunits and liposomes, Cerna, H. Replication of rous sarcoma virus and the biosynthesis of the oncogenic subviral ribonucleoprotein particles, pp.60-1615, 1975.

. Maherani, somes") in the mitochondria isolated from rous sarcoma tissue, Biochem

. Biophys, . Res, M. Commun-adamina, U. Guller, L. Bracci et al., Clinical applications of virosomes in cancer immunotherapy Immunoliposome labeling: A sensitive and specific method for cell surface labeling, Expert Opin. Biol. Ther. J. Immunol. Methods, vol.44206, issue.1 6112, pp.162-170, 1971.

T. Mizoue, T. Horibe, K. Maruyama, T. Takizawa, M. Iwatsuru et al., Targetability and intracellular delivery of anti- BCG antibody-modified, pH-sensitive fusogenic immunoliposomes to tumor cells Long circulating microparticulate drug carriers, Int. J. Pharm. Adv. Drug Deliv. Rev. J.W, vol.237208, issue.162-3, pp.129-137, 1995.

S. Roberge, R. K. Jain, and D. M. Mcdonald, Openings between defective endothelial cells explain tumor vessel leakiness Passive targeting of doxorubicin with polymer coated liposomes in tumor bearing rats, Am. J. Pathol. Biol. Pharm. Bull, vol.156210, issue.47, pp.1363-1380, 2000.

T. M. Allen, Long-circulating (sterically stabilized) liposomes for targeted drug delivery, Trends in Pharmacological Sciences, vol.15, issue.7, pp.215-220, 1994.
DOI : 10.1016/0165-6147(94)90314-X

S. K. Lim, H. J. Park, E. K. Choi, J. S. Kim, . Long-circulating et al., EGFR-targeted liposomes for drugs delivery Study on Azithromycin liposomes obtained by the ether injection method Physicochemical properties of amphotericin B liposomes prepared by reverse-phase evaporation method. Drug Dev Liposome stability and formation: Experimental parameters and theories on the size distribution Preparation and properties of liposomes composed of various phospholipids with different hydrophobic chains using a supercritical reverse phase evaporation method, etal. Processing Pharmaceutical Compounds Using Dense Gas Technology, pp.342-343, 0218.

Z. Li, B. Qin, Z. Q. Zhang, L. Y. Zhang, and J. Yao, Effect of liposome encapsulation on lrinotecan lactone ring and on drug release in vitro, Chin. Pharm. J, issue.13, pp.43-1003, 2008.

M. Mozafari, C. Johnson, S. Hatziantoniou, and C. Demetzos, Nanoliposomes and Their Applications in Food Nanotechnology, Journal of Liposome Research, vol.347, issue.1, pp.309-336, 2008.
DOI : 10.1016/j.ijpharm.2007.06.048

M. Mozafari and M. Mortazavi, Nanoliposomes: From Fundamentals to Recent Developments, 2005.

J. Mathai, G. Sprott, and M. Zeidel, Molecular Mechanisms of Water and Solute Transport across Archaebacterial Lipid Membranes, Journal of Biological Chemistry, vol.101, issue.29, pp.27266-71, 2001.
DOI : 10.1093/oxfordjournals.jbchem.a121942

M. Mehiri, W. Chen, V. Janout, R. S. Barich, H. Munson et al., Molecular umbrella transport: Exceptions to the classic size/Lipophilicity rule Physicochemical properties, formulation, and drug delivery Drug delivery: principles and applications, J Am Chem Soc, vol.1316, issue.4, pp.1338-1347, 2005.

X. Huang, C. Brazel, G. Acharya, and K. Park, On the importance and mechanisms of burst release in matrix-controlled drug delivery systems Mechanisms of controlled drug release from drug-eluting stents, J Contr Rel Mashkevich BO. Drug Delivery Research Advances. Adv Drug Del Rev, vol.7379, issue.583, pp.121-157, 2001.

A. Desai, H. Caldwell, A. Amber, K. Kono, T. Ozawa et al., Sustained release of phenylpropanolamine hydrochloride using a novel controlled release drug delivery system Highly temperature-sensitive liposomes based on a thermosensitive block copolymer for tumorspecific chemotherapy [11] Brazel C, Peppas N. Temperature-and pH-sensitive hydrogels for controlled release of heparin and streptokinase Liposomes and virosomes as immunoadjuvant and antigen-carrier systems in vaccine formulations, Medical Applications of Liposomes, pp.298-307, 1994.

R. Langer, B. Oraceska, U. Jalil, R. Nixon, R. Narasimhan et al., Polymeric delivery systems for controlled drug release A comparison of dissolution properties from matrix tablets prepared from microcapsules and mixtures containing phenobarbitone and poly(DL-lactic acid) In: I Controlled Release of Biologically Active Agents Zero-order release of micro-and macromolecules from polymeric devices: The role of the burst effect, Chem Eng Commun J Control Rel, vol.61617, issue.471, pp.1-48, 1978.

P. Mura, G. Bamanti, L. Fabbri, M. Valleri, R. Harland et al., Controlled release matrix tablets of ketoprofen Pharmaceutical Technology, Controlled Drug Release. Chichester: Ellis Horwood Limited Drug/polymer matrix swelling and dissolution, Pharm Res, vol.18, issue.58, pp.71-80, 1988.

C. Hwang, D. Wu, and E. Edelman, Impact of transport and drug properties on the local pharmacology of drug-eluting stents, International Journal of Cardiovascular Interventions, vol.103, issue.1, pp.7-12, 2003.
DOI : 10.1161/01.CIR.103.2.192

B. Anand, S. Dey, M. A. Oliveira, J. Pereira, J. Canduri et al., Current prodrug strategies via membrane transporters/receptors Crystallographic and presteady-state kinetics studies on binding of NADH to wild-type and isoniazid-resistant enoyl-ACP(CoA) reductase enzymes from mycobacterium tuberculosis, Expert Opin Biol Ther J Mol Biol, vol.2212223, issue.3593, pp.607-627, 2002.

J. Li, B. Wang, P. Liu, M. Nieh, J. Katsaras et al., Possibility of active targeting to tumor by local hyperthermia with temperature-sensitive nanoparticles Controlled release mechanisms of spontaneously forming unilamellar vesicles Ultrasound in drug and gene delivery Magnetic targeting of magnetoliposomes to solid tumors with MR imaging monitoring in mice: Feasibility Preparation and characterization of magnetic cationic liposome in gene delivery, Med Hypotheses Biochim Biophys Acta Biomembr Adv Drug Del Rev Radiology Int J Pharm, vol.7124252627, issue.36612, pp.249-51, 2006.

C. Irli, O. Hasirci, V. Miller, C. Clapp, P. et al., UV-induced drug release from photoactive REV sensitized by suprofen Visible light-induced destabilization of endocytosed liposomes Investigating the uptake and intracellular fate of pH-sensitive liposomes by flow cytometry and spectral bio-imaging, J Control Rel FEBS Lett J Control Rel, vol.962930, issue.1103, pp.85-96, 2000.

D. Hee, W. Tae, C. Byung, and S. Ho, Release of calcein from temperature-sensitive liposomes in a poly(N-isopropylacrylamide) hydrogel [32] Yekta OA, Farivar M, Atilla Hincal A. Temperature-and pHsensitive liposomes [33] Inskeep PB, Darrington RT. Utilization of biopharmaceutical and pharmacokinetic principles in the development of veterinary controlled release drug delivery systems. Controlled Release Veterinary Drug Delivery: Biological and Pharmaceutical Considerations, Macromol Re Eur J Pharm Biopharm, vol.1334, issue.393, pp.54-61, 1993.

M. Yamauchi, K. Tsutsumi, M. Abe, Y. Uosaki, M. Nakakura et al., Release of drugs from liposomes varies with particle size Effect of vesicle size on in vivo release of daunorubicin from hydrogenated egg phosphatidylcholine-based liposomes into blood circulation Enhanced permeability of freeze-dried liposomal bilayers upon rehydration Influence of chain ordering on the selectivity of dipalmitoylphosphatidylcholine bilayer membranes for permeant size and shape al. Effects of lipid composition and preparation conditions on physical-chemical properties, technological parameters and in vitro biological activity of gemcitabine-loaded liposomes, Biol Pharm Bull Biol Pharm Bull Cryobiology Biophys J Curr Drug Del, vol.30353839, issue.41, pp.963-969, 1995.

M. Mozafari, Nanocarrier Technologies: Frontiers of Nanotherapy . The Netherlands, 2006.
DOI : 10.1007/978-1-4020-5041-1

C. Dai, B. Wang, H. Zhao, and B. Li, Factors affecting protein release from microcapsule prepared by liposome in alginate, Colloids and Surfaces B: Biointerfaces, vol.42, issue.3-4, pp.253-261, 2005.
DOI : 10.1016/j.colsurfb.2004.12.020

L. Gregoriadis and . Technology, Liposome Preparation and Related Techniques, 2007.

D. Lasic and Y. Barenholz, A Handbook of Nonmedical Applications of Liposomes: Models for Biological Phenomena, 1996.

Y. Lichtenberg and . Barenholz, Liposomes: Preparation, Characterization, and Preservation, 1998.
DOI : 10.1016/0005-2736(81)90550-2

A. Berger, J. Sachse, R. Bender, M. Schubert, and . Brandl, Filter extrusion of liposomes using different devices: comparison of liposome size, encapsulation efficiency, and process characteristics, International Journal of Pharmaceutics, vol.223, issue.1-2, pp.55-68, 2001.
DOI : 10.1016/S0378-5173(01)00721-9

R. Mozafari, Nanomaterials and Nanosystems for Biomedical Applications The Netherlands, 2007.

D. Zucker, Y. Marcus, A. Barenholz, and . Goldblum, Liposome drugs' loading efficiency: A working model based on loading conditions and drug's physicochemical properties, Journal of Controlled Release, vol.139, issue.1, pp.73-80, 2009.
DOI : 10.1016/j.jconrel.2009.05.036

R. Mozafari and M. S. Mortazavi, Nanoliposomes: from Fundamentals to Recent Developments, 2005.

A. Betz, N. Aeppli, H. Menshutina, and . Leuenberger, In vivo comparison of various liposome formulations for cosmetic application???, International Journal of Pharmaceutics, vol.296, issue.1-2, pp.44-54, 2005.
DOI : 10.1016/j.ijpharm.2005.02.032

G. Calvagno, C. Celia, D. Paolino, D. Cosco, M. Iannone et al., Effects of Lipid Composition and Preparation Conditions on Physical-Chemical Properties, Technological Parameters and In Vitro Biological Activity of Gemcitabine-Loaded Liposomes, Current Drug Delivery, vol.4, issue.1, pp.89-101, 2007.
DOI : 10.2174/156720107779314749

H. Shimanouchi, N. Ishii, H. Yoshimoto, R. Umakoshi, and . Kuboi, Calcein permeation across phosphatidylcholine bilayer membrane: Effects of membrane fluidity, liposome size, and immobilization, Colloids and Surfaces B: Biointerfaces, vol.73, issue.1, pp.156-160, 2009.
DOI : 10.1016/j.colsurfb.2009.05.014

A. Cornell, Experiments with Mixtures, 2002.

L. Gregoriadis and . Technology, Liposome Preparation and Related Techniques, 2007.

Y. Ishii and . Nagasaka, Simple and Convenient Method for Estimation of Marker Entrapped in Liposomes, Journal of Dispersion Science and Technology, vol.415, issue.1, pp.97-101, 2001.
DOI : 10.1016/0005-2736(90)90111-Z

M. G. Taylor and R. M. Morris, Thermal analysis of phase transition behaviour in liposomes, Thermochimica Acta, vol.248, pp.289-301, 1995.
DOI : 10.1016/0040-6031(94)01884-J

T. Bunjes and A. Unruh, Characterization of lipid nanoparticles by differential scanning calorimetry, X-ray and neutron scattering???, Advanced Drug Delivery Reviews, vol.59, issue.6, pp.379-402, 2007.
DOI : 10.1016/j.addr.2007.04.013

H. Jousma, F. Talsma, and . Spies, Characterization of liposomes. The influence of extrusion of multilamellar vesicles through polycarbonate membranes on particle size, particle size distribution and number of bilayers, International Journal of Pharmaceutics, vol.35, issue.3, pp.263-274, 1987.
DOI : 10.1016/0378-5173(87)90139-6

S. Ferreri, A. Pierotti, L. Barbieri, L. Zambonin, S. Landi et al., Comparison of Phosphatidylcholine Vesicle Properties Related to Geometrical Isomerism???, Photochemistry and Photobiology, vol.33, issue.1, pp.274-280, 2006.
DOI : 10.1016/0005-2736(91)90295-J

R. F. Jurima-romet, J. Barber, P. N. Demeester, and . Shek, Distribution studies of liposome-encapsulated glutathione administered to the lung, International Journal of Pharmaceutics, vol.63, issue.3, pp.227-235, 1990.
DOI : 10.1016/0378-5173(90)90129-R

R. C. New, Liposomes: a Practical Approach, 1990.

X. Xiang and B. D. Anderson, Influence of Chain Ordering on the Selectivity of Dipalmitoylphosphatidylcholine Bilayer Membranes for Permeant Size and Shape, Biophysical Journal, vol.75, issue.6, pp.2658-2671, 1998.
DOI : 10.1016/S0006-3495(98)77711-2

A. Nii, K. Takamura, F. Mohri, and . Ishii, Factors affecting physicochemical properties of liposomes prepared with hydrogenated purified egg yolk lecithins by the microencapsulation vesicle method, Colloids and Surfaces B: Biointerfaces, vol.27, issue.4, pp.323-332, 2003.
DOI : 10.1016/S0927-7765(02)00097-8

B. N. Nagahiro, C. H. Mora, R. K. Boasquevisque, G. A. Scheule, and . Patterson, TOXICITY OF CATIONIC LIPOSOME-DNA COMPLEX IN LUNG ISOGRAFTS, Transplantation, vol.69, issue.9, pp.1802-1805, 2000.
DOI : 10.1097/00007890-200005150-00012

D. Brgles, M. D. Jurasin, R. Sikiri-c, J. Frkanec, and . Tomasi-c, Entrapment of Ovalbumin into Liposomes???Factors Affecting Entrapment Efficiency, Liposome Size, and Zeta Potential, Journal of Liposome Research, vol.42, issue.3, pp.235-248, 2008.
DOI : 10.1016/j.colsurfb.2005.01.008

S. Grabielle-madelmont, M. Lesieur, and . Ollivon, Characterization of loaded liposomes by size exclusion chromatography, Journal of Biochemical and Biophysical Methods, vol.56, issue.1-3, pp.189-217, 2003.
DOI : 10.1016/S0165-022X(03)00059-9

A. Chandaroy, S. W. Sen, and . Hui, Temperature-controlled content release from liposomes encapsulating Pluronic F127, Journal of Controlled Release, vol.76, issue.1-2, pp.27-37, 2001.
DOI : 10.1016/S0168-3659(01)00429-1

A. Schneider, G. Sachse, M. Robling, and . Brandl, Generation of contrast-carrying liposomes of defined size with a new continuous high pressure extrusion method, International Journal of Pharmaceutics, vol.117, issue.1, pp.1-12, 1995.
DOI : 10.1016/0378-5173(94)00245-Z

E. E. Laridi, R. O. Kheadr, J. C. Benech, C. Vuillemard, I. Lacroix et al., Liposome encapsulated nisin Z: optimization, stability and release during milk fermentation, International Dairy Journal, vol.13, issue.4, pp.325-336, 2003.
DOI : 10.1016/S0958-6946(02)00194-2

P. Y. Lohse, D. Bolinger, and . Stamou, Encapsulation Efficiency Measured on Single Small Unilamellar Vesicles, Journal of the American Chemical Society, vol.130, issue.44, pp.14372-14373, 2008.
DOI : 10.1021/ja805030w

E. Maherani, M. R. Arab-tehrany, C. Mozafari, M. Gaiani, and . Linder, Liposomes: A Review of Manufacturing Techniques and Targeting Strategies, Current Nanoscience, vol.7, issue.3, pp.436-452, 2011.
DOI : 10.2174/157341311795542453

E. Maherani, M. Arab-tehrany, and . Linder, Mechanism of Bioactive Transfer through Liposomal Bilayers, Current Drug Targets, vol.12, issue.4, pp.531-545, 2011.
DOI : 10.2174/138945011794751582

M. A. Luykx, R. J. Peters, S. M. Van-ruth, and H. Bouwmeester, A Review of Analytical Methods for the Identification and Characterization of Nano Delivery Systems in Food, Journal of Agricultural and Food Chemistry, vol.56, issue.18, pp.8231-8247, 2008.
DOI : 10.1021/jf8013926

G. Fr?-uhwirth, N. Fritz, O. Freiberger, and . Glatter, Structure and order in lamellar phases determined by small-angle scattering, Journal of Applied Crystallography, vol.37, issue.5, pp.703-710, 2004.
DOI : 10.1107/S0021889804012956

H. Agashe, P. Lagisetty, S. Awasthi, and V. Awasthi, Improved formulation of liposome-encapsulated hemoglobin with an anionic non-phospholipid, Colloids and Surfaces B: Biointerfaces, vol.75, issue.2, pp.75-573, 2010.
DOI : 10.1016/j.colsurfb.2009.09.038

M. A. Babizhayev, Biological activities of the natural imidazole-containing peptidomimetics n-acetylcarnosine, carcinine and l-carnosine in ophthalmic and skin care products, Life Sciences, vol.78, issue.20, pp.78-2343, 2006.
DOI : 10.1016/j.lfs.2005.09.054

H. Bunjes and T. Unruh, Characterization of lipid nanoparticles by differential scanning calorimetry, X-ray and neutron scattering???, Advanced Drug Delivery Reviews, vol.59, issue.6, pp.379-402, 2007.
DOI : 10.1016/j.addr.2007.04.013

M. G. Calvagno, C. Celia, D. Paolino, D. Cosco, M. Iannone et al., Effects of Lipid Composition and Preparation Conditions on Physical-Chemical Properties, Technological Parameters and In Vitro Biological Activity of Gemcitabine-Loaded Liposomes, Current Drug Delivery, vol.4, issue.1, pp.89-101, 2007.
DOI : 10.2174/156720107779314749

J. C. Colas, W. Shi, V. S. Rao, A. Omri, M. R. Mozafari et al., Microscopical investigations of nisin-loaded nanoliposomes prepared by Mozafari method and their bacterial targeting, Micron, vol.38, issue.8, pp.38-841, 2007.
DOI : 10.1016/j.micron.2007.06.013

P. Da-silva-malheiros, D. J. Daroit, and A. Brandelli, Food applications of liposome-encapsulated antimicrobial peptides, Trends in Food Science & Technology, vol.21, issue.6, pp.284-292, 2010.
DOI : 10.1016/j.tifs.2010.03.003

K. G. Desai and H. J. Park, Recent Developments in Microencapsulation of Food Ingredients, Drying Technology, vol.26, issue.7, pp.1361-1394, 2005.
DOI : 10.1081/PDT-100101357

R. Dong and L. E. Yu, Investigation of Surface Changes of Nanoparticles Using TM-AFM Phase Imaging, Environmental Science & Technology, vol.37, issue.12, pp.2813-2819, 2003.
DOI : 10.1021/es034071k

T. Fruhwirth, G. Fritz, N. Freiberger, and O. Glatter, Structure and order in lamellar phases determined by small-angle scattering, Journal of Applied Crystallography, vol.37, issue.5, pp.703-710, 2004.
DOI : 10.1107/S0021889804012956

C. Gasparovic, I. Barba, J. Born, S. Barton, C. Arús et al., A Study of Imidazole-Based Nuclear Magnetic Resonance Probes of Cellular pH, Analytical Biochemistry, vol.261, issue.1, pp.64-72, 1998.
DOI : 10.1006/abio.1998.2698

S. Gemili, A. Yemeniciog-?lu, and S. A. Altinkaya, Development of antioxidant food packaging materials with controlled release properties, Journal of Food Engineering, vol.96, issue.3, pp.325-332, 2010.
DOI : 10.1016/j.jfoodeng.2009.08.020

G. Gregoriadis, Liposome technology. Liposome preparation and related techniques, 2007.

A. R. Hipkiss, Could carnosine suppress zinc-mediated proteasome inhibition and neurodegeneration? Therapeutic potential of a non-toxic but non-patentable dipeptide, Biogerontology, vol.133, issue.supplement, pp.147-149, 2005.
DOI : 10.1089/rej.1.1999.2.337

R. Holliday and G. A. Mcfarland, Inhibition of the growth of transformed and neoplastic cells by the dipeptide carnosine, British Journal of Cancer, vol.73, issue.8, pp.966-971, 1996.
DOI : 10.1038/bjc.1996.189

S. Hupfeld, A. M. Holsaeter, M. Skar, C. B. Frantzen, and M. Brandl, Liposome size analysis by dynamic/static light scattering upon size exclusion-/field flowfractionation, Journal of Nanoscience and Nanotechnology, vol.6, pp.9-10, 2006.

H. Jousma, H. Talsma, and F. Spies, Characterization of liposomes. The influence of extrusion of multilamellar vesicles through polycarbonate B. Maherani et al, Food Chemistry, vol.134, pp.632-640, 1987.

R. Kohen, Y. Yamamoto, K. C. Cundy, and B. N. Ames, Antioxidant activity of carnosine, homocarnosine, and anserine present in muscle and brain., Proceedings of the National Academy of Sciences, vol.85, issue.9, pp.3175-3179, 1988.
DOI : 10.1073/pnas.85.9.3175

L. Krapf, M. Dezi, W. Reichstein, J. Köhler, and S. Oellerich, AFM characterization of spin-coated multilayered dry lipid films prepared from aqueous vesicle suspensions, Colloids and Surfaces B: Biointerfaces, vol.82, issue.1, pp.25-32, 2011.
DOI : 10.1016/j.colsurfb.2010.08.006

S. B. Kulkarni, G. V. Betageri, and M. Singh, Factors affecting microencapsulation of drugs in liposomes, Journal of Microencapsulation, vol.19, issue.3, pp.229-246, 1995.
DOI : 10.1016/0024-3205(76)90082-5

P. Leclère, R. Lazzaroni, J. L. Brédas, J. M. Yu, P. Dubois et al., Microdomain Morphology Analysis of Block Copolymers by Atomic Force Microscopy with Phase Detection Imaging, Langmuir, vol.12, issue.18, pp.12-4317, 1996.
DOI : 10.1021/la9600964

S. Lesieur, C. Grabielle-madelmont, M. T. Paternostre, and M. Ollivon, Size analysis and stability study of lipid vesicles by high-performance gel exclusion chromatography, turbidity, and dynamic light scattering, Analytical Biochemistry, vol.192, issue.2, pp.334-343, 1991.
DOI : 10.1016/0003-2697(91)90545-5

X. Liang, G. Mao, and K. Y. Ng, Mechanical properties and stability measurement of cholesterol-containing liposome on mica by atomic force microscopy, Journal of Colloid and Interface Science, vol.278, issue.1, pp.53-62, 2004.
DOI : 10.1016/j.jcis.2004.05.042

X. Liang, G. Mao, and K. Y. Ng, Probing small unilamellar EggPC vesicles on mica surface by atomic force microscopy, Colloids and Surfaces B: Biointerfaces, vol.34, issue.1, pp.41-51, 2004.
DOI : 10.1016/j.colsurfb.2003.10.017

D. Lichtenberg and Y. Barenholz, Liposomes: Preparation, Characterization, and Preservation, 1998.
DOI : 10.1016/0005-2736(81)90550-2

B. Lohse, P. Y. Bolinger, and D. Stamou, Encapsulation Efficiency Measured on Single Small Unilamellar Vesicles, Journal of the American Chemical Society, vol.130, issue.44, pp.14372-14373, 2008.
DOI : 10.1021/ja805030w

D. M. Luykx, R. J. Peters, S. M. Van-ruth, and H. Bouwmeester, A Review of Analytical Methods for the Identification and Characterization of Nano Delivery Systems in Food, Journal of Agricultural and Food Chemistry, vol.56, issue.18, pp.56-8231, 2008.
DOI : 10.1021/jf8013926

R. C. Macdonald, R. I. Macdonald, B. P. Menco, K. Takeshita, N. K. Subbarao et al., Small-volume extrusion apparatus for preparation of large, unilamellar vesicles, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1061, issue.2, pp.297-303, 1991.
DOI : 10.1016/0005-2736(91)90295-J

B. Maherani, E. Arab-tehrany, M. R. Mozafari, C. Gaiani, and M. Linder, Liposomes: A Review of Manufacturing Techniques and Targeting Strategies, Current Nanoscience, vol.7, issue.3, pp.436-452, 2011.
DOI : 10.2174/157341311795542453

M. R. Mozafari, Nanoliposomes: Preparation and analysis Liposomes: Methods and protocols, p.pharmaceutical nanocarriers, 2010.
DOI : 10.1007/978-1-60327-360-2_2

M. R. Mozafari, C. Johnson, S. Hatziantoniou, and C. Demetzos, Nanoliposomes and Their Applications in Food Nanotechnology, Journal of Liposome Research, vol.347, issue.1, pp.309-327, 2008.
DOI : 10.1016/j.ijpharm.2007.06.048

M. R. Mozafari, K. Khosravi-darani, G. G. Borazan, J. Cui, A. Pardakhty et al., Encapsulation of Food Ingredients Using Nanoliposome Technology, International Journal of Food Properties, vol.6, issue.4, pp.833-844, 2008.
DOI : 10.1016/j.jconrel.2006.04.015

M. R. Mozafari and M. S. Mortazavi, Nanoliposomes: From fundamentals to recent developments, 2005.

K. Nagai, A. Niijima, T. Yamano, H. Otani, N. Okumra et al., Possible Role of L-Carnosine in the Regulation of Blood Glucose through Controlling Autonomic Nerves, Experimental Biology and Medicine, vol.227, issue.10, pp.228-1138, 2003.
DOI : 10.2337/diabetes.48.12.2286

K. Nakano, Y. Tozuka, H. Yamamoto, Y. Kawashima, and H. Takeuchi, A novel method for measuring rigidity of submicron-size liposomes with atomic force microscopy, International Journal of Pharmaceutics, vol.355, issue.1-2, pp.203-209, 2008.
DOI : 10.1016/j.ijpharm.2007.12.018

T. Nii and F. Ishii, Encapsulation efficiency of water-soluble and insoluble drugs in liposomes prepared by the microencapsulation vesicle method, International Journal of Pharmaceutics, vol.298, issue.1, pp.198-205, 2005.
DOI : 10.1016/j.ijpharm.2005.04.029

T. Nii, A. Takamura, K. Mohri, and F. Ishii, Factors affecting physicochemical properties of liposomes prepared with hydrogenated purified egg yolk lecithins by the microencapsulation vesicle method, Colloids and Surfaces B: Biointerfaces, vol.27, issue.4, pp.323-332, 2003.
DOI : 10.1016/S0927-7765(02)00097-8

W. R. Perkins, S. R. Minchey, P. L. Ahl, and A. S. Janoff, The determination of liposome captured volume, Chemistry and Physics of Lipids, vol.64, issue.1-3, pp.1-3, 1993.
DOI : 10.1016/0009-3084(93)90066-C

B. Ruozi, G. Tosi, E. Leo, and M. A. Vandelli, Application of atomic force microscopy to characterize liposomes as drug and gene carriers, Talanta, vol.73, issue.1, pp.12-22, 2007.
DOI : 10.1016/j.talanta.2007.03.031

B. Ruozi, G. Tosi, M. Tonelli, L. Bondioli, A. Mucci et al., AFM phase imaging of soft-hydrated samples: A versatile tool to complete the chemical-physical study of liposomes, Journal of Liposome Research, vol.290, issue.1, pp.59-67, 2009.
DOI : 10.1007/s003390051340

P. Schön, K. Bagdi, K. Molnár, P. Markus, B. Pukánszky et al., Quantitative mapping of elastic moduli at the nanoscale in phase separated polyurethanes by AFM, European Polymer Journal, vol.47, issue.4, pp.692-698, 2011.
DOI : 10.1016/j.eurpolymj.2010.09.029

T. Shimanouchi, H. Ishii, N. Yoshimoto, H. Umakoshi, and R. Kuboi, Calcein permeation across phosphatidylcholine bilayer membrane: Effects of membrane fluidity, liposome size, and immobilization, Colloids and Surfaces B: Biointerfaces, vol.73, issue.1, pp.156-160, 2009.
DOI : 10.1016/j.colsurfb.2009.05.014

T. M. Taylor, P. M. Davidson, B. D. Bruce, and J. Weiss, Liposomal Nanocapsules in Food Science and Agriculture, Critical Reviews in Food Science and Nutrition, vol.44, issue.7-8, pp.7-8, 2005.
DOI : 10.1093/pcp/pcg005

K. M. Taylor and R. M. Morris, Thermal analysis of phase transition behaviour in liposomes, Thermochimica Acta, vol.248, issue.C, pp.289-301, 1995.
DOI : 10.1016/0040-6031(94)01884-J

L. M. Were, B. D. Bruce, P. M. Davidson, and J. Weiss, Size, Stability, and Entrapment Efficiency of Phospholipid Nanocapsules Containing Polypeptide Antimicrobials, Journal of Agricultural and Food Chemistry, vol.51, issue.27, pp.51-8073, 2003.
DOI : 10.1021/jf0348368

X. M. Zhang, A. B. Patel, R. A. De-graaf, and K. L. Behar, Determination of liposomal encapsulation efficiency using proton NMR spectroscopy, Chemistry and Physics of Lipids, vol.127, issue.1, pp.113-120, 2004.
DOI : 10.1016/j.chemphyslip.2003.09.013

B. Maherani, Influence of lipid composition on physicochemical properties of nanoliposomes encapsulating natural dipeptide antioxidant l-carnosine, Food Chemistry, vol.134, issue.2, pp.632-640, 2012.
DOI : 10.1016/j.foodchem.2012.02.098

I. Résultats, &. Discussions-chapitre, I. R. Ii-]-m, M. S. Mozafari, and . Mortazavi, Interaction moléculaire de la calcéine avec les membranes References Nanoliposomes: From Fundamentals to Recent Developments The Netherlands, Mozafari, Nanomaterials and Nanosystems for Biomedical Applications Maherani, E. Arab-Tehrany, M. Linder, Mechanism of bioactive transfer through liposomal bilayers, pp.531-545, 2005.

]. K. Gardikis, S. Hatziantoniou, K. Viras, M. Wagner, and C. Demetzos, A DSC and Raman spectroscopy study on the effect of PAMAM dendrimer on DPPC model lipid membranes, International Journal of Pharmaceutics, vol.318, issue.1-2, pp.318-118, 2006.
DOI : 10.1016/j.ijpharm.2006.03.023

. Fresta, Effects of lipid composition and preparation conditions on physical-chemical properties, technological parameters and in vitro biological activity of gemcitabineloaded liposomes, Current Drug Delivery, vol.4, pp.89-101, 2007.

S. Bonora, M. D. Foggia, and M. Iafisco, DSC and Raman study on the interaction of DDT [1,1,1-trichloro-2,2-bis(p-chlorophenyl)-ethane] with liposomal phospholipids, Pesticide Biochemistry and Physiology, vol.92, issue.3, pp.144-149, 2008.
DOI : 10.1016/j.pestbp.2008.07.008

K. Gardikis, S. Hatziantoniou, K. Viras, and C. Demetzos, Effect of a bioactive curcumin derivative on DPPC membrane: A DSC and Raman spectroscopy study, Thermochimica Acta, vol.447, issue.1, pp.1-4, 2006.
DOI : 10.1016/j.tca.2006.03.007

]. T. Hata, H. Matsuki, and S. Kaneshina, Effect of local anesthetics on the bilayer membrane of dipalmitoylphosphatidylcholine: interdigitation of lipid bilayer and vesicle???micelle transition, Biophysical Chemistry, vol.87, issue.1, pp.25-36, 2000.
DOI : 10.1016/S0301-4622(00)00175-7

S. J. Bae, S. Kitamura, L. G. Herbette, and J. M. Sturtevant, The effects of calcium channel blocking drugs on the thermotropic behavior of dimyristoylphosphatidylcholine, Chemistry and Physics of Lipids, vol.51, issue.1, pp.1-7, 1989.
DOI : 10.1016/0009-3084(89)90059-5

. Kinnunen, Characterization of mixed monolayers of phosphatidylcholine and a dicationic gemini surfactant SS-1 with a langmuir balance: Effects of DNA, Biophysical Journal, vol.81, pp.2135-2143, 2001.

M. Vankann, J. Möllerfeld, H. Ringsdorf, and H. Höcker, Amphiphilic Model Peptides: Circular Dichroism Measurements and Investigations by a Langmuir Balance, Journal of Colloid and Interface Science, vol.178, issue.1, pp.241-250, 1996.
DOI : 10.1006/jcis.1996.0112

M. R. Mozafari, Nanoliposomes: Preparation and Analysis, Liposomes: Methods and Protocols, Pharmaceutical Nanocarriers, 2010.
DOI : 10.1007/978-1-60327-360-2_2

. Hu, Small-volume extrusion apparatus for preparation of large unilammelar vesicles, Biochim. Biophys. Acta, vol.1061, pp.297-303, 1991.

K. Czapla, B. Korchowiec, M. Orlof, J. R. Magnieto, and E. Rogalska, Enzymatic Probing of Model Lipid Membranes: Phospholipase A2 Activity toward Monolayers Modified by Oxicam NSAIDs, The Journal of Physical Chemistry B, vol.115, issue.29, pp.9290-9298, 2011.
DOI : 10.1021/jp202716k

D. I. Bower and W. F. Maddams, The Vibrational Spectroscopy of Polymers, 1989.
DOI : 10.1017/CBO9780511623189

E. Smith and . Dent, Modern Raman Spectroscopy: A Practical Approach, 2005.
DOI : 10.1002/0470011831

URL : http://cds.cern.ch/record/1137935/files/9780471497943_TOC.pdf

I. W. Levin, T. E. Thompson, Y. Barenholz, and C. Huang, Two types of hydrocarbon chain interdigitation in sphingomyelin bilayers, Biochemistry, vol.24, issue.22, pp.6282-6286, 1985.
DOI : 10.1021/bi00343a036

I. W. Levin, E. Keihn, and W. C. Harris, A Raman spectroscopic study on the effect of cholesterol on lipid packing in diether phosphatidylcholine bilayer dispersions, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.820, issue.1, pp.40-47, 1985.
DOI : 10.1016/0005-2736(85)90213-5

A. Csiszár, E. Koglin, R. J. Meier, and E. Klumpp, The phase transition behavior of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) model membrane influenced by 2,4-dichlorophenol???an FT-Raman Spectroscopy Study, Chemistry and Physics of Lipids, vol.139, issue.2, pp.115-124, 2006.
DOI : 10.1016/j.chemphyslip.2005.11.005

H. Matsui and S. Pan, Distribution of DNA in Cationic Liposome Complexes Probed by Raman Microscopy, Langmuir, vol.17, issue.3, pp.571-573, 2001.
DOI : 10.1021/la000437k

P. C. Painter, M. M. Coleman, and J. L. Koenig, The theory of vibrational spectroscopy and its application to polymeric materials, 1982.

X. M. Li, B. Zhao, D. Q. Zhao, J. Z. Ni, Y. Wu et al., Interaction of La3+ and cholesterol with dipalmitoylphosphatidylglycerol bilayers by FT-Raman spectroscopy, Thin Solid Films, pp.284-285, 1996.

E. Koglin and R. J. Meier, Conformational dependence of Raman frequencies and intensities in alkanes and polyethylene, Computational and Theoretical Polymer Science, vol.9, issue.3-4, pp.327-333, 1999.
DOI : 10.1016/S1089-3156(99)00022-7

H. Diehl and U. L. Ellingboe, Indicator for Titration of Calcium in Presence of Magnesium Using Disodium Dihydrogen Ethylenediamine Tetraacetate, Analytical Chemistry, vol.28, issue.5, pp.882-884, 1956.
DOI : 10.1021/ac60113a030

G. M. Maghraby, A. C. Williams, and B. W. Barry, Drug interaction and location in liposomes: correlation with polar surface areas, International Journal of Pharmaceutics, vol.292, issue.1-2, pp.292-179, 2005.
DOI : 10.1016/j.ijpharm.2004.11.037

T. Shimanouchi, H. Ishii, N. Yoshimoto, H. Umakoshi, and R. Kuboi, Calcein permeation across phosphatidylcholine bilayer membrane: Effects of membrane fluidity, liposome size, and immobilization, Colloids and Surfaces B: Biointerfaces, vol.73, issue.1, pp.156-160, 2009.
DOI : 10.1016/j.colsurfb.2009.05.014

B. Maherani, E. Arab-tehrany, A. Kheirolomoom, M. J. Stebe, and M. Linder, Optimization and characterization of liposome formulation by mixture design, The Analyst, vol.50, issue.3, pp.773-786, 2012.
DOI : 10.1103/PhysRevE.50.5047

URL : https://hal.archives-ouvertes.fr/hal-00703284

C. H. Huang and I. W. Levin, Effect of lipid chain length inequivalence on the packing characteristics of bilayer assemblies. Raman spectroscopic study of phospholipid dispersions in the gel state, The Journal of Physical Chemistry, vol.87, issue.9, pp.1509-1513, 1983.
DOI : 10.1021/j100232a012

B. J. Litman, Packing characteristics of highly unsaturated bilayer lipids: Raman spectroscopic studies of multilamellar phosphatidylcholine dispersions, Biochemistry, vol.30, issue.2, pp.313-319, 1991.
DOI : 10.1021/bi00216a001

B. P. Gaber and W. L. Peticolas, On the quantitative interpretation of biomembrane structure by Raman spectroscopy, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.465, issue.2, pp.260-274, 1977.
DOI : 10.1016/0005-2736(77)90078-5

J. A. Cornell, Experiments with Mixtures, 2002.

G. Zaccai, G. Büldt, A. Seelig, and J. Seelig, Neutron diffraction studies on phosphatidylcholine model membranes, Journal of Molecular Biology, vol.134, issue.4, pp.693-706, 1979.
DOI : 10.1016/0022-2836(79)90480-7

G. Büldt, H. U. Gally, J. Seelig, and G. Zaccai, Neutron diffraction studies on phosphatidylcholine model membranes, Journal of Molecular Biology, vol.134, issue.4, pp.673-691, 1979.
DOI : 10.1016/0022-2836(79)90479-0

J. T. Mason, C. Huang, and R. L. Biltonen, Calorimetric investigations of saturated mixed-chain phosphatidylcholine bilayer dispersions, Biochemistry, vol.20, issue.21, pp.6086-6092, 1981.
DOI : 10.1021/bi00524a026

J. T. Mason and C. H. Huang, Chain length dependent thermodynamics of saturated symmetric-chain phosphatidylcholine bilayers, Lipids, vol.424, issue.8, pp.604-608, 1981.
DOI : 10.1016/0005-2760(76)90051-5

J. Hernandez-borrell, F. Mas, and J. Puy, A theoretical approach to describe monolayerliposome lipid interaction, Biophysical Chemistry, pp.36-47, 1990.

L. Zhao, S. S. Feng, and M. L. Go, Investigation of molecular interactions between paclitaxel and DPPC by langmuir film balance and differential scanning calorimetry, Journal of Pharmaceutical Sciences, vol.93, issue.1, pp.86-98, 2004.
DOI : 10.1002/jps.10523

F. Pétriat, E. Roux, J. C. Leroux, and S. Giasson, Study of Molecular Interactions between a Phospholipidic Layer and a pH-Sensitive Polymer Using the Langmuir Balance Technique, Langmuir, vol.20, issue.4, pp.1393-1400, 2004.
DOI : 10.1021/la035583f

K. M. Taylor and R. M. Morris, Thermal analysis of phase transition behaviour in liposomes, Thermochimica Acta, vol.248, pp.289-301, 1995.
DOI : 10.1016/0040-6031(94)01884-J

Y. Corvis, W. Barzyk, G. Brezesinski, N. Mrabet, M. Badis et al., Interactions of a Fungistatic Antibiotic, Griseofulvin, with Phospholipid Monolayers Used as Models of Biological Membranes, Langmuir, vol.22, issue.18, pp.7701-7711, 2006.
DOI : 10.1021/la060998x

. Nowakowska, Interaction of curcumin with lipid monolayers and liposomal bilayers, Colloids and Surfaces B: Biointerfaces, 2011.

J. Gravier, B. Korchowiec, R. Schneider, and E. Rogalska, Interaction of amphiphilic chlorin-based photosensitizers with 1,2-dipalmitoyl-sn-glycero-3-phosphocholine monolayers, Chemistry and Physics of Lipids, vol.158, issue.2, pp.102-109, 2009.
DOI : 10.1016/j.chemphyslip.2009.01.004

URL : https://hal.archives-ouvertes.fr/hal-00371629

Y. Corvis, G. Brezesinski, R. Rink, A. Walcarius, A. Van-der-heyden et al., Analytical Investigation of the Interactions between SC3 Hydrophobin and Lipid Layers:?? Elaborating of Nanostructured Matrixes for Immobilizing Redox Systems, Analytical Chemistry, vol.78, issue.14, pp.78-4850, 2006.
DOI : 10.1021/ac0602064

B. Korchowiec, M. Paluch, Y. Corvis, and E. Rogalska, A Langmuir film approach to elucidating interactions in lipid membranes: 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine/cholesterol/metal cation systems, Chemistry and Physics of Lipids, vol.144, issue.2, pp.127-136, 2006.
DOI : 10.1016/j.chemphyslip.2006.08.005

R. G. Snyder, H. L. Strauss, and C. A. Elliger, Carbon-hydrogen stretching modes and the structure of n-alkyl chains. 1. Long, disordered chains, The Journal of Physical Chemistry, vol.86, issue.26, pp.5145-5150, 1982.
DOI : 10.1021/j100223a018

R. A. Macphail, H. L. Strauss, R. G. Snyder, and C. A. Eiliger, Carbon-hydrogen stretching modes and the structure of n-alkyl chains. 2. Long, all-trans chains, 139 III. Résultats & Discussions Chapitre III, pp.334-341, 1984.
DOI : 10.1021/j150647a002

V. B. Leite, A. Cavalli, and O. N. Oliveira-jr, Hydrogen-bond control of structure and conductivity of Langmuir films, Physical Review E, vol.170, issue.6, pp.6835-6839, 1998.
DOI : 10.1016/0301-0104(93)85117-Q

X. Du and Y. Liang, -alaninate Langmuir???Blodgett Films, The Journal of Physical Chemistry B, vol.104, issue.43, pp.10047-10052, 2000.
DOI : 10.1021/jp002016h

A. Dicko, H. Bourque, and M. Pézolet, Study by infrared spectroscopy of the conformation of dipalmitoylphosphatidylglycerol monolayers at the air???water interface and transferred on solid substrates, Chemistry and Physics of Lipids, vol.96, issue.1-2, pp.125-139, 1998.
DOI : 10.1016/S0009-3084(98)00084-X

R. N. Lewis, R. N. Mcelhaney, W. Pohle, and H. H. Mantsch, Components of the carbonyl stretching band in the infrared spectra of hydrated 1,2-diacylglycerolipid bilayers: a reevaluation, Biophysical Journal, vol.67, issue.6, pp.67-2367, 1994.
DOI : 10.1016/S0006-3495(94)80723-4

A. Blume, W. Huebner, G. Messner, and F. Transform-infrared, Fourier transform infrared spectroscopy of 13C:O labeled phospholipids hydrogen bonding to carbonyl groups, Biochemistry, vol.27, issue.21, pp.8239-8249, 1988.
DOI : 10.1021/bi00421a038

L. Y. Zhao, S. S. Feng, N. Kocherginsky, and I. Kostetski, DSC and EPR investigations on effects of cholesterol component on molecular interactions between paclitaxel and phospholipid within lipid bilayer membrane, International Journal of Pharmaceutics, vol.338, issue.1-2, pp.338-258, 2007.
DOI : 10.1016/j.ijpharm.2007.01.045

I. Résultats, &. Discussion-chapitre, and I. , III: Mécanisme de transfert de molécules hydrophiles References: [1] B. Maherani, E. Arab-Tehrany, M. Linder, Mechanism of bioactive transfer through liposomal bilayers, Curr. Drug Targets, vol.12, pp.531-545, 2011.

]. N. Bergstrand, M. C. Arfvidsson, J. M. Kim, D. H. Thompson, and K. Edwards, Interactions between pH-sensitive liposomes and model membranes, Biophysical Chemistry, vol.104, issue.1, pp.361-379, 2003.
DOI : 10.1016/S0301-4622(03)00011-5

D. H. Qualls, S. Thompson-gardikis, K. Hatziantoniou, M. Viras, C. Wagner et al., Chloroaluminum phthalocyanine tetrasulfonate delivered via acid-labile diplasmenylcholine-folate liposomes: Intracellular localization and synergistic phototoxicity, International Journal of Cancer, vol.59, issue.3, pp.384-392, 2001.
DOI : 10.1038/bjc.1989.73

K. Gardikis, S. Hatziantoniou, K. Viras, and C. Demetzos, Effect of a bioactive curcumin derivative on DPPC membrane: A DSC and Raman spectroscopy study, Thermochimica Acta, vol.447, issue.1, pp.1-4, 2006.
DOI : 10.1016/j.tca.2006.03.007

]. D. Needham and M. W. Dewhirst, The development and testing of a new temperaturesensitive drug delivery system for the treatment of solid tumors, Adv. Drug Del. Rev, issue.6, pp.53-285, 2001.

G. Acharya and K. Park, Mechanisms of controlled drug release from drug-eluting stents, Advanced Drug Delivery Reviews, vol.58, issue.3, pp.387-401, 2006.
DOI : 10.1016/j.addr.2006.01.016

R. W. Baker-narasimhan and R. Langer, Controlled Release of Biologically Active Agents Zero-order release of micro-and macromolecules from polymeric devices: The role of the burst effect, J Contr Rel, pp.47-60, 1978.

B. Maherani, E. Arab-tehrany, A. Kheirolomoom, F. Cleymand, and M. Linder, Influence of lipid composition on physicochemical properties of nanoliposomes encapsulating natural dipeptide antioxidant l-carnosine, Food Chemistry, vol.134, issue.2
DOI : 10.1016/j.foodchem.2012.02.098

B. Maherani, E. Arab-tehrany, A. Kheirolomoom, M. J. Stebe, and M. Linder, Optimization and characterization of liposome formulation by mixture design, The Analyst, vol.50, issue.3, pp.773-786, 2012.
DOI : 10.1103/PhysRevE.50.5047

URL : https://hal.archives-ouvertes.fr/hal-00703284

M. R. Mozafari, Nanoliposomes: Preparation and Analysis, Liposomes: Methods and Protocols, 2010.
DOI : 10.1007/978-1-60327-360-2_2

R. C. Macdonald, R. I. Macdonald, B. P. Menco, K. Takeshita, N. K. Subbarao et al., Small-volume extrusion apparatus for preparation of large, unilamellar vesicles, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1061, issue.2, pp.297-303, 1991.
DOI : 10.1016/0005-2736(91)90295-J

G. Gregoriadis and L. Technology, Third ed., Volume I, Liposome Preparation and Related Techniques, 2007.

T. Shimanouchi, H. Ishii, N. Yoshimoto, H. Umakoshi, and R. Kuboi, Calcein permeation across phosphatidylcholine bilayer membrane: Effects of membrane fluidity, liposome size, and immobilization, Colloids and Surfaces B: Biointerfaces, vol.73, issue.1, pp.156-160, 2009.
DOI : 10.1016/j.colsurfb.2009.05.014

S. Hamann, J. F. Kiilgaard, T. Litman, F. J. Alvarez-leefmans, B. R. Winther et al., Measurement of Cell Volume Changes by Fluorescence Self-Quenching, Journal of Fluorescence, vol.12, issue.2, pp.139-145, 2002.
DOI : 10.1023/A:1016832027325

T. Shimanouchi, P. Walde, J. Gardiner, Y. R. Mahajan, D. Seebach et al., Permeation of a beta-heptapeptide derivative across phospholipid bilayers, Biochimica Et Biophysica Acta-Biomembranes, pp.1768-2726, 2007.

S. Mourtas, S. Fotopoulou, S. Duraj, V. Sfika, C. Tsakiroglou et al., Liposomal drugs dispersed in hydrogels, Colloids and Surfaces B: Biointerfaces, vol.55, issue.2, pp.212-221, 2007.
DOI : 10.1016/j.colsurfb.2006.12.005

M. Corzo-martinez, M. Avila, F. J. Moreno, T. Requena, and M. Villamiel, Effect of milk protein glycation and gastrointestinal digestion on the growth of bifidobacteria and lactic acid bacteria, International Journal of Food Microbiology, vol.153, issue.3, pp.420-427, 2012.
DOI : 10.1016/j.ijfoodmicro.2011.12.006

M. Hamoudi, E. Fattal, C. Gueutin, V. Nicolas, and A. Bochot, Beads made of cyclodextrin and oil for the oral delivery of lipophilic drugs: In vitro studies in simulated gastro-intestinal fluids, International Journal of Pharmaceutics, vol.416, issue.2, pp.507-514, 2011.
DOI : 10.1016/j.ijpharm.2011.01.062

G. Mandalari, K. Adel-patient, V. Barkholt, C. Baro, L. Bennett et al., In vitro digestibility of ??-casein and ??-lactoglobulin under simulated human gastric and duodenal conditions: A multi-laboratory evaluation, Regulatory Toxicology and Pharmacology, vol.55, issue.3, pp.372-381, 2009.
DOI : 10.1016/j.yrtph.2009.08.010

S. W. Hell and J. Wichmann, Breaking the diffraction resolution limit by stimulated emission: stimulated-emission-depletion fluorescence microscopy, Optics Letters, vol.19, issue.11, pp.780-782, 1994.
DOI : 10.1364/OL.19.000780

J. Tønnesen, F. Nadrigny, K. I. Willig, R. Wedlich-söldner, and U. V. , Two-Color STED Microscopy of Living Synapses Using A Single Laser-Beam Pair, Biophysical Journal, vol.101, issue.10, pp.2545-2552, 2011.
DOI : 10.1016/j.bpj.2011.10.011

E. Rittweger, K. Y. Han, S. E. Irvine, C. Eggeling, and S. W. , STED microscopy reveals crystal colour centres with nanometric resolution, Nature Photonics, vol.442, issue.3, pp.144-147, 2009.
DOI : 10.1038/nphoton.2009.2

K. X. Ngo, H. Umakoshi, T. Shimanouchi, and R. Kuboi, Characterization of heat-induced interaction of neutral liposome with lipid membrane of Streptomyces griseus cell, Colloids and Surfaces B: Biointerfaces, vol.73, issue.2, pp.399-407, 2009.
DOI : 10.1016/j.colsurfb.2009.06.015

K. X. Ngo, H. Umakoshi, T. Shimanouchi, H. S. Jung, S. Morita et al., Heat-enhanced production of chitosanase from Streptomyces griseus in the presence of liposome, Journal of Bioscience and Bioengineering, vol.100, issue.5, pp.495-501, 2005.
DOI : 10.1263/jbb.100.495

J. G. Wijmans and R. W. Baker, The solution-diffusion model: a review, Journal of Membrane Science, vol.107, issue.1-2, pp.1-21, 1995.
DOI : 10.1016/0376-7388(95)00102-I

W. D. Stein, Transport and Diffusion Across Cell Membranes, 1986.

C. Pidgeon, S. Ong, H. Liu, X. Qiu, M. Pidgeon et al., IAM chromatography: an in vitro screen for predicting drug membrane permeability, Journal of Medicinal Chemistry, vol.38, issue.4, pp.38-590, 1995.
DOI : 10.1021/jm00004a004

B. De-castro, P. Gameiro, J. L. Lima, C. Matos, and S. Reis, Location and partition coefficients of anti-inflammatory drugs in EPC liposomes. A fluorescence quenching study using n-(9-anthroyloxy)-stearic probes, Colloids and Surfaces A: Physicochemical and Engineering Aspects, vol.190, issue.1-2, pp.205-212, 2001.
DOI : 10.1016/S0927-7757(01)00680-X

E. Blatt and W. H. Sawyer, Depth-dependent flourescent quenching in micelles and membranes, Biochimica et Biophysica Acta (BBA) - Reviews on Biomembranes, vol.822, issue.1, pp.43-62, 1985.
DOI : 10.1016/0304-4157(85)90003-6

V. Joguparthi, T. X. Xiang, and B. D. Anderson, Liposome Transport of Hydrophobic Drugs: Gel Phase Lipid Bilayer Permeability and Partitioning of the Lactone Form of a Hydrophobic Camptothecin, DB???67, Journal of Pharmaceutical Sciences, vol.97, issue.1, pp.97-400, 2008.
DOI : 10.1002/jps.21125

M. A. Raviolo, J. M. Sanchez, M. C. Brinon, and M. A. Perillo, Determination of liposome permeability of ionizable carbamates of zidovudine by steady state fluorescence spectroscopy, Colloids and Surfaces B: Biointerfaces, vol.61, issue.2, pp.61-188, 2008.
DOI : 10.1016/j.colsurfb.2007.08.004

W. Zhang, E. C. Van-winden, J. A. Bouwstra, and D. J. Crommelin, Enhanced Permeability of Freeze-Dried Liposomal Bilayers upon Rehydration, Cryobiology, vol.35, issue.3, pp.277-289, 1997.
DOI : 10.1006/cryo.1997.2050

M. Yamauchi, K. Tsutsumi, M. Abe, Y. Uosaki, M. Nakakura et al., Release of Drugs from Liposomes Varies with Particle Size, Biological & Pharmaceutical Bulletin, vol.30, issue.5, pp.963-966, 2007.
DOI : 10.1248/bpb.30.963

Z. Yi, M. Nagao, and D. P. Bossev, Bending elasticity of saturated and monounsaturated phospholipid membranes studied by the neutron spin echo technique, Journal of Physics: Condensed Matter, vol.21, issue.15, 2009.
DOI : 10.1088/0953-8984/21/15/155104

V. Aguilella, M. Belaya, and V. Levadny, Ion Permeability of a Membrane with Soft Polar Interfaces. 1. The Hydrophobic Layer as the Rate-Determining Step, Langmuir, vol.12, issue.20, pp.4817-4827, 1996.
DOI : 10.1021/la960174n

H. P. Duwe and E. Sackmann, Bending elasticity and thermal excitations of lipid bilayer vesicles: Modulation by solutes, Physica A: Statistical Mechanics and its Applications, vol.163, issue.1, pp.163-410, 1990.
DOI : 10.1016/0378-4371(90)90349-W

K. Olbrich, W. Rawicz, D. Needham, and E. Evans, Water Permeability and Mechanical Strength of Polyunsaturated Lipid Bilayers, Biophysical Journal, vol.79, issue.1, pp.321-327, 2000.
DOI : 10.1016/S0006-3495(00)76294-1

J. H. Lpsen, O. G. Mouritsen, and M. Bloom, Relationships between lipid membrane area, hydrophobic thickness, and acyl-chain orientational order. The effects of cholesterol, Biophys. J, pp.57-405, 1990.

W. Rawicz, K. C. Olbrich, T. Mcintosh, D. Needham, and E. Evans, Effect of Chain Length and Unsaturation on Elasticity of Lipid Bilayers, Biophysical Journal, vol.79, issue.1, pp.79-328, 2000.
DOI : 10.1016/S0006-3495(00)76295-3

G. Illya, R. Lipowsky, and J. C. Shillcock, Effect of chain length and asymmetry on material properties of bilayer membranes, The Journal of Chemical Physics, vol.62, issue.24, p.122, 2005.
DOI : 10.1016/S0006-3495(03)74637-2

J. M. Ali, M. M. Smaby, H. L. Momsen, R. E. Brockman, and . Brown, Acyl Chain-Length Asymmetry Alters the Interfacial Elastic Interactions of Phosphatidylcholines, Biophysical Journal, vol.74, issue.1, pp.74-338, 1998.
DOI : 10.1016/S0006-3495(98)77791-4

T. X. Xiang, A computer simulation of free-volume distributions and related structural properties in a model lipid bilayer, Biophysical Journal, vol.65, issue.3, pp.1108-1120, 1993.
DOI : 10.1016/S0006-3495(93)81156-1

T. X. Xiang, X. Chen, and B. D. Anderson, Transport methods for probing the barrier domain of lipid bilayer membranes, Biophysical Journal, vol.63, issue.1, pp.78-88, 1992.
DOI : 10.1016/S0006-3495(92)81581-3

D. Papahadjopoulos, K. Jacobson, S. Nir, and T. Isac, Phase transitions in phospholipid vesicles Fluorescence polarization and permeability measurements concerning the effect of temperature and cholesterol, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.311, issue.3, pp.311-330, 1973.
DOI : 10.1016/0005-2736(73)90314-3

T. X. Xiang and B. D. Anderson, Influence of Chain Ordering on the Selectivity of Dipalmitoylphosphatidylcholine Bilayer Membranes for Permeant Size and Shape, Biophysical Journal, vol.75, issue.6, pp.2658-2671, 1998.
DOI : 10.1016/S0006-3495(98)77711-2

M. G. Calvagno, C. Celia, D. Paolino, D. Cosco, M. Iannone et al., Effects of Lipid Composition and Preparation Conditions on Physical-Chemical Properties, Technological Parameters and In Vitro Biological Activity of Gemcitabine-Loaded Liposomes, Current Drug Delivery, vol.4, issue.1, pp.89-101, 2007.
DOI : 10.2174/156720107779314749

J. A. Gazzara, M. C. Phillips, S. Lund-katz, M. N. Palgunachari, J. P. Segrest et al., Effect of vesicle size on their interaction with class A amphipathic helical peptides, J. Lipid Res, vol.38, pp.2147-2154, 1997.

T. X. Xiang and B. D. Anderson, The relationship between permeant size and permeability in lipid bilayer membranes, The Journal of Membrane Biology, vol.140, issue.2, pp.111-122, 1994.
DOI : 10.1007/BF00232899

T. X. Xiang and B. D. Anderson, Permeability of acetic acid across gel and liquidcrystalline lipid bilayers conforms to free-surface-area theory, Biophys. J, pp.72-223, 1997.

A. Nagayasu, T. Shimooka, and H. Kiwada, Effect of Vesicle Size on in vivo Release of Daunorubicin from Hydrogenated Egg Phosphatidylcholine-Based Liposomes into Blood Circulation., Biological & Pharmaceutical Bulletin, vol.18, issue.7, pp.1020-1023, 1995.
DOI : 10.1248/bpb.18.1020

J. Li, B. Wang, and P. Liu, Possibility of active targeting to tumor by local hyperthermia with temperature-sensitive nanoparticles, Medical Hypotheses, vol.71, issue.2, pp.249-251, 2008.
DOI : 10.1016/j.mehy.2008.03.023

A. Tronde, B. Nordén, A. Jeppsson, P. Brunmark, E. Nilsson et al., Drug Absorption from the Isolated Perfused Rat Lung???Correlations with Drug Physicochemical Properties and Epithelial Permeability, Journal of Drug Targeting, vol.4, issue.1, pp.61-74, 2003.
DOI : 10.1021/jm9810102

P. Ertl, B. Rohde, and P. Selzer, Fast Calculation of Molecular Polar Surface Area as a Sum of Fragment-Based Contributions and Its Application to the Prediction of Drug Transport Properties, Journal of Medicinal Chemistry, vol.43, issue.20, pp.43-3714, 2000.
DOI : 10.1021/jm000942e

G. M. Maghraby, A. C. Williams, and B. W. Barry, Drug interaction and location in liposomes: correlation with polar surface areas, International Journal of Pharmaceutics, vol.292, issue.1-2, pp.292-179, 2005.
DOI : 10.1016/j.ijpharm.2004.11.037

A. Breindl, B. Beck, T. Clark, and R. C. Glen, Prediction of the n-Octanol/Water Partition Coefficient, logP, Using a Combination of Semiempirical MO-Calculations and a Neural Network, Journal of Molecular Modeling, vol.3, issue.3, pp.142-155, 1997.
DOI : 10.1007/s008940050027

F. Kamp and H. A. Hamilton, pH gradients across phospholipid membranes caused by fast flip-flop of un-ionized fatty acids., Proc. Natl. Acad. Sci, pp.89-11367, 1992.
DOI : 10.1073/pnas.89.23.11367

T. Fichert, M. Yazdanian, and J. R. Proudfoot, A structure???Permeability study of small drug-like molecules, Bioorganic & Medicinal Chemistry Letters, vol.13, issue.4, pp.719-722, 2003.
DOI : 10.1016/S0960-894X(02)01035-1

G. E. Flaten, A. B. Dhanikula, K. Luthman, and M. Brandl, Drug permeability across a phospholipid vesicle based barrier: A novel approach for studying passive diffusion, Eur, J. Pharm. Sci, pp.27-80, 2006.

H. Vandewaterbeemd, G. Camenisch, G. Folkers, and O. A. Raevsky, Estimation of Caco-2 Cell Permeability using Calculated Molecular Descriptors, Quantitative Structure-Activity Relationships, vol.9, issue.6, pp.480-490, 1996.
DOI : 10.1111/j.2042-7158.1994.tb03858.x

D. A. Bahnick, Use of Huckel Molecular Orbital Theory in Interpreting the Visible Spectra of Polymethine Dyes: An Undergraduate Physical Chemistry Experiment, Journal of Chemical Education, vol.71, issue.2, pp.71-171, 1994.
DOI : 10.1021/ed071p171

T. B. Fulton, Diffusion and Transport Across Cell Membranes, Prologue Course, pp.65-85, 2009.

C. Y. Dai, B. C. Wang, H. W. Zhao, and B. Li, Factors affecting protein release from microcapsule prepared by liposome in alginate, Colloids and Surfaces B: Biointerfaces, vol.42, issue.3-4, pp.253-258, 2005.
DOI : 10.1016/j.colsurfb.2004.12.020

A. M. Krieg, From Bugs to Drugs: Therapeutic Immunomodulation with Oligodeoxynucleotides Containing CpG Sequences from Bacterial DNA, Antisense and Nucleic Acid Drug Development, vol.11, issue.3, pp.181-188, 2001.
DOI : 10.1089/108729001300338717

T. A. Klar, M. Dyba, and S. W. Hell, Stimulated emission depletion microscopy with an offset depleting beam, Applied Physics Letters, vol.78, issue.4, pp.393-395, 2001.
DOI : 10.1063/1.127056