. .. , 174 II. 2. 1. Modèle in vitro d'évaluation des propriétés anti-inflammatoires des matériaux à base d'associations chitosan/alginate et ?CD-chitosan/alginate

. , Évaluation de l'effet des CoPEC chitosan/alginate et ?CD-chitosan/alginate sur l'expression du marqueur d'activation CD40 par des macrophages activés

. , Évaluation de l'effet des CoPEC chitosan/alginate et ?CD-chitosan/alginate sur l'expression du marqueur de prolifération Ki-67 par des macrophages activés

. , Détermination du rôle des polyélectrolytes dans l'effet anti-inflammatoire des CoPEC chitosan/alginate et ?CD-chitosan/alginate

. , Évaluation de l'impact de l'incorporation de piroxicam sur les propriétés antiinflammatoires du CoPEC ?CD-chitosan/alginate, incorporation de molécules actives anti-inflammatoires sur les propriétés du CoPEC ?CD-chitosan/alginate vis-à-vis de l'inflammation

. .. , 2. 4. Évaluation de l'impact de l'inclusion de piroxicam dans le CoPEC ?CDchitosan/alginate sur la production d'oxyde nitrique par des macrophages activés, 3. Extraction et dosage du piroxicam inclus dans les CoPEC

. , 3. 4. Évaluation de l'impact de l'inclusion de prednisolone dans le CoPEC ?CDchitosan/alginate sur la production d'oxyde nitrique par des macrophages activés

N. R. Patel and P. Gohil, A Review on Biomaterials: Scope, Applications & Human Anatomy Significance, International Journal of Emerging Technology and Advanced Engineering, vol.2012, issue.4, pp.91-101

Q. Chen and G. A. Thouas, Metallic Implant Biomaterials. Materials Science and Engineering: R: Reports, vol.87, pp.1-57, 2015.

J. Schijve, Fatigue of Structures and Materials in the 20th Century and the State of the Art, International Journal of Fatigue, vol.25, issue.8, pp.679-702, 2003.

R. K. Singh-raman, S. Jafari, and S. E. Harandi, Corrosion Fatigue Fracture of Magnesium Alloys in Bioimplant Applications: A Review, Engineering Fracture Mechanics, vol.137, pp.97-108, 2015.

M. Dehurtevent, L. Robberecht, J. C. Hornez, A. Thuault, E. Deveaux et al., Stereolithography: A New Method for Processing Dental Ceramics by Additive ComputerAided Manufacturing, Dental Materials, vol.33, issue.5, pp.477-485, 2017.

A. J. Teo, A. Mishra, I. Park, Y. Kim, W. Park et al., Polymeric Biomaterials for Medical Implants and Devices, ACS Biomaterials Science & Engineering, vol.2016, issue.4, pp.454-472

J. Yin and S. Luan, Opportunities and Challenges for the Development of PolymerBased Biomaterials and Medical Devices, Regenerative Biomaterials, vol.2016, issue.2, pp.129-135

J. E. Mark, Physical Properties of Polymers Handbook Second Edition, 2007.

S. Li and A. H. Burstein, Ultra-High Molecular Weight Polyethylene. The Material and its Use in Total Joint Implants, The Journal of Bone and Joint Surgery American, vol.1994, issue.7, pp.1080-1090

J. Zhou, X. Huang, D. Zheng, H. Li, T. Herrler et al., Oriental Nose Elongation using an L-Shaped Polyethylene Sheet Implant for Combined Septal Spreading and Extension, Aesthetic Plastic Surgery, vol.38, issue.2, pp.295-302, 2014.

C. Terrada, K. Julian, N. Cassoux, A. M. Prieur, M. Debre et al., Cataract Surgery with Primary Intraocular Lens Implantation in Children with Uveitis: Long-Term Outcomes, Journal of Cataract and Refractive Surgery, vol.37, issue.11, pp.1977-1983, 2011.

A. Rivkin, A Prospective Study of Non-Surgical Primary Rhinoplasty using a

, Polymethylmethacrylate Injectable Implant. Dermatological Surgery, vol.40, issue.3, pp.305-313, 2014.

B. J. Kim, K. S. Hong, K. J. Park, D. H. Park, Y. G. Chung et al., In Vivo Studies Comparing the Biocompatibility of Various Polypropylene Meshes and their Handling Properties during Endoscopic Total Extraperitoneal (TEP) Patchplasty: An Experimental Study in Pigs, Journal of Korean Neurosurgical Society, vol.2012, issue.6, pp.211-220, 2004.

F. Zheng, L. Xu, L. Verbiest, E. Verbeken, D. De-ridder et al., Cytokine Production Following Experimental Implantation of Xenogenic Dermal Collagen and Polypropylene Grafts in Mice, Neurourology and Urodynamics, vol.26, issue.2, pp.280-289, 2007.

H. K. Na, H. Y. Song, J. H. Kim, J. H. Park, M. K. Kang et al., How to Design the Optimal Self-Expandable Oesophageal Metallic Stents: 22 Years of Experience in 645 Patients with Malignant Strictures, European Radiology, vol.23, issue.3, pp.786-796, 2013.

H. K. Na, H. Y. Song, H. J. Yeo, J. H. Park, J. H. Kim et al., Retrospective Comparison of Internally and Externally Covered Retrievable Stent Placement for Patients with Benign Urethral Strictures caused by Traumatic Injury, American Journal of Roentgenology, vol.2012, issue.1, pp.55-61

G. T. Howard, Biodegradation of Polyurethane: A Review. International Biodeterioration and Biodegradation, vol.49, pp.245-252, 2002.

J. A. De-la-peña-salcedo, M. A. Soto-miranda, and J. F. Lopez-salguero, Back to the Future: A 15-Year Experience with Polyurethane Foam-Covered Breast Implants using the Partial-Subfascial Technique, Aesthetic Plastic Surgery, vol.2012, issue.2, pp.331-338

G. A. Georgeu and J. D. Frame, Conical Polyurethane Implants: An Uplifting Augmentation, Aesthetic Surgery Journal, vol.33, issue.8, pp.1116-1128, 2013.

R. A. Hachem, E. Perez, I. Bueno, . Van-de, T. R. Water et al., Comparison of Packing Material in Middle Ear Mucosal Trauma, Otolaryngology-Head and Neck Surgery, vol.2012, issue.2_suppl, pp.83-84

H. F. Guo, Z. S. Li, S. W. Dong, W. J. Chen, L. Deng et al., Piezoelectric PU/PVDF Electrospun Scaffolds for Wound Healing Applications. Colloids and Surfaces B Biointerfaces, vol.96, pp.29-36, 2012.

N. E. Yousef and A. A. Shaman, Reduction of Microbial Contamination along Medical Polymeric Implants, Journal of American Science, vol.2013, issue.12, pp.92-99

M. Hasegawa and K. Horie, Photophysics, Photochemistry, and Optical Properties of Polyimides. Progress in Polymer Science, vol.26, pp.259-335, 2001.

G. C. Hakan, P. Jiansheng, A. Georgiev, D. Dimov, E. Spassova et al., Chemical and Physical Properties of Polyimides: Biomedical and Engineering Applications, High Performance Polymers-Polyimides Based-From Chemistry to Applications, Abadie, M., Ed. IntechOpen: 2012. 27. Hasenkamp, vol.15, pp.831-839, 2008.

A. K. Thota, S. Kuntaegowdanahalli, A. K. Starosciak, J. J. Abbas, J. Orbay et al., A System and Method to Interface with Multiple Groups of Axons in Several Fascicles of Peripheral Nerves, Journal of Neuroscience Methods, vol.244, pp.78-84, 2015.

K. Y. Kwon, B. Sirowatka, A. Weber, and W. Li, Opto-?ECoG Array: A Hybrid Neural Interface with Transparent ?ECoG Electrode Array and Integrated LEDs for Optogenetics, IEEE Transactions on Biomedical Circuits and Systems, vol.7, issue.5, pp.593-600, 2013.

K. K. Yong, S. Brenton, L. Wen, and W. Arthur, Opto-?ECoG Array: Transparent ?ECoG Electrode Array and Integrated LEDs for Optogenetics, IEEE Biomedical Circuits and Systems Conference, vol.2012, pp.164-167, 2012.

E. Yilgör and I. Yilgör, Silicone Containing Copolymers: Synthesis, Properties and Applications. Progress in Polymer Science, vol.39, pp.1165-1195, 2014.

R. F. Willis, Thermal Decomposition of Silicone Fluids at Metal Surfaces, Nature, vol.221, pp.1134-1135, 1969.

N. Van-ardenne, J. Vanderwegen, and G. Van-nuffelen,

P. Heyning, Medialization Thyroplasty: Vocal Outcome of Silicone and Titanium Implant, European Archives of Oto-Rhino-Laryngology, vol.268, issue.1, pp.101-107, 2011.

D. S. Borah, S. S. Pal, R. Aakanksha, and C. Report, Silicone Implant in Augmentation of Saddle Nose, International Journal of Recent Scientific Research, vol.2013, issue.4, pp.1661-1662

A. Unkovskiy, S. Spintzyk, J. Brom, F. Huettig, and C. Keutel, Direct 3D Printing of Silicone Facial Prostheses: A Preliminary Experience in Digital Workflow, The Journal of Prosthetic Dentistry, 2018.

C. R. Dandekar and Y. C. Shin, Modeling of Machining of Composite Materials: A Review, International Journal of Machine Tools and Manufacture, vol.57, pp.102-121, 2012.

E. Salernitano, C. Migliaresi, G. Gourav, K. Ankur, T. Rahul et al., Application and Future of Composite Materials: A Review, Composite Materials for Biomedical Applications: A Review. Journal of Applied Biomaterials and Biomechanics, vol.1, issue.1, pp.6907-6911, 2003.

N. Björk, K. Ekstrand, and I. E. Ruyter,

, Graphite Fibre Reinforced Poly (Methyl Methacrylate), Biomaterials, vol.7, issue.1, pp.73-75, 1986.

J. Jancar, A. T. Dibenedetto, and A. J. Goldberg, Thermoplastic Fibre-Reinforced Composites for Dentistry. Part II Effect of Moisture on Flexural Properties of Unidirectional Composites, Journal of Materials Science Materials in Medicine, vol.4, issue.6, pp.562-568, 1993.

T. Chen, C. Yao, H. Wang, G. Chou, T. Lee et al., Evaluation of a Novel Malleable, Biodegradable Osteoconductive Composite in a Rabbit Cranial Defect Model. Materials Chemistry and Physics, vol.55, pp.44-50, 1998.

Y. Shikinami and M. Okuno, Bioresorbable Devices made of Forged Composites of

, Hydroxyapatite (HA) Particles and Poly L-Lactide (PLLA). Part II: Practical Properties of Miniscrews and Miniplates, vol.22, pp.3197-3211, 2001.

S. Kim, M. Park, O. Jeon, C. Yong-choi, and B. Kim,

. Glycolide, Hydroxyapatite Composite Scaffolds for Bone Tissue Engineering, vol.27, pp.1399-1409, 2006.

X. Liu, L. A. Smith, J. Hu, and P. X. Ma, Biomimetic Nanofibrous Gelatin/Apatite Composite Scaffolds for Bone Tissue Engineering, Biomaterials, vol.30, issue.12, pp.2252-2258, 2009.

N. T. Dai, M. R. Williamson, N. Khammo, E. F. Adams, and A. G. Coombes, Composite Cell Support Membranes based on Collagen and Polycaprolactone for Tissue Engineering of Skin, Biomaterials, vol.25, issue.18, pp.4263-4271, 2004.

A. Wang, R. Lin, V. K. Polineni, A. Essner, C. Stark et al., Carbon Fiber Reinforced Polyether Ether Ketone Composite as a Bearing Surface for Total Hip Replacement, Tribology International, vol.31, issue.11, pp.661-667, 1998.

H. J. Früh and G. Willmann, Tribological Investigations of the Wear Couple AluminaCFRP for Total Hip Replacement, Biomaterials, vol.19, issue.13, pp.1145-1150, 1998.

J. Kolarík, C. Migliaresi, M. Stol, and L. Nicolais, Mechanical Properties of Model Synthetic Tendons, Journal of Biomedical Materials Research, vol.15, issue.2, pp.147-157, 1981.

P. A. Davis, S. J. Huang, L. Ambrosio, D. Ronca, and L. Nicolais, A Biodegradable Composite Artificial Tendon, Journal of Materials Science: Materials in Medicine, vol.1992, issue.5, pp.359-364
DOI : 10.1007/bf00705368

D. Huguet, J. Delecrin, N. Passuti, and G. Daculsi, Ovine Anterior Cruciate Ligament Reconstruction using a Synthetic Prosthesis and a Collagen Inductor, Journal of Materials Science Materials in Medicine, vol.8, issue.2, pp.67-73, 1997.

F. Mollica, M. Ventre, F. Sarracino, L. Ambrosio, and L. Nicolais, Mechanical Properties and Modelling of a Hydrophilic Composite used as a Biomaterial, Composites Science and Technology, vol.66, issue.1, pp.92-101, 2006.

M. Hasegawa, A. Sudo, Y. Shikinami, and A. Uchida, Biological Performance of a Three-Dimensional Fabric as Artificial Cartilage in the Repair of Large Osteochondral Defects in Rabbit, Biomaterials, vol.20, issue.20, pp.1969-1975, 1999.

G. Wu, B. Su, W. Zhang, and C. Wang, In Vitro Behaviors of Hydroxyapatite Reinforced Polyvinyl Alcohol Hydrogel Composite, Materials Chemistry and Physics, vol.107, issue.2, pp.364-369, 2008.

M. Hess, R. G. Jones, J. Kahovec, T. Kitayama, P. Kratochvil et al.,

, Polymers Containing Ionizable or Ionic Groups and of Polymers Containing Ions, Pure and Applied Chemistry, vol.78, issue.11, pp.2067-2074, 2006.

S. Lankalapalli and V. R. Kolapalli, Polyelectrolyte Complexes: A Review of their Applicability in Drug Delivery Technology, Indian Journal of Pharmaceutical Sciences, issue.5, pp.481-487, 2009.

P. Schaaf and J. B. Schlenoff, Saloplastics: Processing Compact Polyelectrolyte Complexes, Advanced Materials, vol.27, issue.15, pp.2420-2432, 2015.
DOI : 10.1002/adma.201500176

H. G. Bungenberg-de-jong and H. R. Kruyt, Coacervation (Partial Miscibility in Colloid Systems), Proceedings of the Koninklijke Nederlandse Akademie van Wetenschappen, vol.32, pp.849-856, 1929.

A. S. Michaels and R. G. Miekka, Polycation-Polyanion Complexes: Preparation and Properties of Poly-(Vinylbenzyltrimethylammonium) Poly-(Styrenesulfonate). The Journal of Physical Chemistry, vol.65, pp.1765-1773, 1961.

A. S. Michaels and P. Complexes, Industrial and Engineering Chemistry, vol.57, issue.10, pp.32-40, 1965.

J. B. Schlenoff, H. Ly, and M. Li, Charge and Mass Balance in Polyelectrolyte Multilayers, Journal of the American Chemical Society, vol.120, issue.30, pp.7626-7634, 1998.

C. B. Bucur, Z. Sui, and J. B. Schlenoff, Ideal Mixing in Polyelectrolyte Complexes and Multilayers: Entropy Driven Assembly, Journal of the American Chemical Society, vol.128, issue.42, pp.13690-13691, 2006.
DOI : 10.1021/ja064532c

J. B. Schlenoff, A. H. Rmaile, and C. B. Bucur, Hydration Contributions to Association in Polyelectrolyte Multilayers and Complexes: Visualizing Hydrophobicity, Journal of the American Chemical Society, vol.130, issue.41, pp.13589-13597, 2008.

V. A. Kabanov and A. B. Zezin, Soluble Interpolymeric Complexes as a New Class of Synthetic Polyelectrolytes, Pure and Applied Chemistry, vol.56, issue.3, pp.343-354, 1984.

A. Zintchenko, G. Rother, and H. Dautzenberg, Transition Highly Aggregated Complexes-Soluble Complexes via Polyelectrolyte Exchange Reactions: Kinetics, Structural Changes, and Mechanism, Langmuir, vol.19, issue.6, pp.2507-2513, 2003.
DOI : 10.1021/la0265427

M. A. Mintzer and E. E. Simanek, Nonviral Vectors for Gene Delivery, Chemical Reviews, vol.109, issue.2, pp.259-302, 2009.
DOI : 10.1021/cr800409e

G. Decher, J. D. Hong, and J. Schmitt, Buildup of Ultrathin Multilayer Films by a SelfAssembly Process: III. Consecutively Alternating Adsorption of Anionic and Cationic Polyelectrolytes on Charged Surfaces, Thin Solid Films, pp.831-835, 1992.

G. Decher, Fuzzy Nanoassemblies: Toward Layered Polymeric Multicomposites, Science, vol.277, issue.5330, pp.1232-1237, 1997.

K. Sato, Y. Imoto, J. Sugama, S. Seki, H. Inoue et al., Sugar-Induced Disintegration of Layer-by-Layer Assemblies Composed of Concanavalin A and Glycogen, Langmuir, vol.21, issue.2, pp.797-799, 2005.

J. A. Jaber and J. B. Schlenoff, Recent Developments in the Properties and Applications of Polyelectrolyte Multilayers. Current Opinion in Colloid and Interface Science, vol.11, pp.324-329, 2006.

T. Boudou, T. Crouzier, K. Ren, G. Blin, and C. Picart, Multiple Functionalities of
URL : https://hal.archives-ouvertes.fr/hal-00670215

P. Multilayer-films, New Biomedical Applications. Advanced Materials, vol.22, issue.4, pp.441-467, 2010.

J. B. Schlenoff, S. T. Dubas, and T. Farhat, Sprayed Polyelectrolyte Multilayers, Langmuir, vol.16, issue.26, pp.9968-9969, 2000.

S. Lee, J. Hong, C. H. Kim, K. Kim, J. P. Koo et al., Layer-byLayer Deposited Multilayer Assemblies of Ionene-Type Polyelectrolytes Based on the SpinCoating Method, Macromolecules, vol.34, issue.16, pp.5358-5360, 2001.

O. Mermut, J. Lefebvre, D. G. Gray, and C. J. Barrett, Structural and Mechanical Properties of Polyelectrolyte Multilayer Films Studied by AFM, Macromolecules, vol.36, issue.23, pp.8819-8824, 2003.

J. D. Mendelsohn, S. Y. Yang, J. Hiller, A. I. Hochbaum, and M. F. Rubner, Rational Design of Cytophilic and Cytophobic Polyelectrolyte Multilayer Thin Films, Biomacromolecules, vol.4, issue.1, pp.96-106, 2003.

J. A. Jaber and J. B. Schlenoff, Mechanical Properties of Reversibly Cross-Linked Ultrathin Polyelectrolyte Complexes, Journal of the American Chemical Society, vol.128, issue.9, pp.2940-2947, 2006.

L. Richert, F. Boulmedais, P. Lavalle, J. Mutterer, E. Ferreux et al., Improvement of Stability and Cell Adhesion Properties of Polyelectrolyte Multilayer Films by Chemical Cross-Linking, Biomacromolecules, vol.5, issue.2, pp.284-294, 2004.

L. Richert, A. J. Engler, D. E. Discher, and C. Picart, Elasticity of Native and CrossLinked Polyelectrolyte Multilayer Films, Biomacromolecules, vol.5, issue.5, pp.1908-1916, 2004.

G. Francius, J. Hemmerlé, J. Ohayon, P. Schaaf, J. C. Voegel et al., Effect of Crosslinking on the Elasticity of Polyelectrolyte Multilayer Films Measured by Colloidal Probe AFM. Microscopy Research and Technique, vol.69, pp.84-92, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00846392

B. Li, D. T. Haynie, and M. Biomimetics, Reversible Covalent Stabilization of a Nanostructured Biofilm, Biomacromolecules, vol.5, issue.5, pp.1667-1670, 2004.

S. Y. Yang and M. F. Rubner, Micropatterning of Polymer Thin Films with pH-Sensitive and Cross-Linkable Hydrogen-Bonded Polyelectrolyte Multilayers, Journal of the American Chemical Society, vol.124, issue.10, pp.2100-2101, 2002.

S. C. Olugebefola, W. A. Kuhlman, M. F. Rubner, A. M. Mayes, S. Deng et al., Photopatterned Nanoporosity in Polyelectrolyte Multilayer Films, Journal of the American Chemical Society, vol.24, issue.9, pp.13723-13731, 2004.

C. P. Vázquez, T. Boudou, V. Dulong, C. Nicolas, C. Picart et al., Variation of Polyelectrolyte Film Stiffness by Photo-Cross-Linking: A New Way to Control Cell Adhesion, Langmuir, vol.25, issue.6, pp.3556-3563, 2009.

B. Woltmann, B. Torger, M. Müller, and U. Hempel, Interaction Between Immobilized Polyelectrolyte Complex Nanoparticles and Human Mesenchymal Stromal Cells, International Journal of Nanomedicine, vol.9, pp.2205-2215, 2014.

M. T. Thompson, M. C. Berg, I. S. Tobias, M. F. Rubner, and K. J. Van-vliet, Tuning Compliance of Nanoscale Polyelectrolyte Multilayers to Modulate Cell Adhesion, Biomaterials, vol.26, issue.34, pp.6836-6845, 2005.

D. S. Salloum, S. G. Olenych, T. C. Keller, and J. B. Schlenoff, Vascular Smooth Muscle Cells on Polyelectrolyte Multilayers: Hydrophobicity-Directed Adhesion and Growth, Biomacromolecules, vol.6, issue.1, pp.161-167, 2005.

O. Etienne, C. Gasnier, C. Taddei, J. C. Voegel, D. Aunis et al., Antifungal Coating by

, Biofunctionalized Polyelectrolyte Multilayered Films, Biomaterials, vol.26, issue.33, pp.6704-6712, 2005.

J. A. Lichter and M. F. Rubner, Polyelectrolyte Multilayers with Intrinsic Antimicrobial Functionality: The Importance of Mobile Polycations, Langmuir, vol.25, issue.13, pp.7686-7694, 2009.

P. Schultz, D. Vautier, L. Richert, N. Jessel, Y. Haikel et al., Polyelectrolyte Multilayers Functionalized by a Synthetic Analogue of an Anti-Inflammatory Peptide, Alpha-MSH, for Coating a Tracheal Prosthesis, Biomaterials, vol.26, issue.15, pp.2621-2630, 2005.

C. Picart, A. Schneider, O. Etienne, J. Mutterer, P. Schaaf et al., Controlled Degradability of Polysaccharide Multilayer Films In Vitro and In Vivo, Advanced Functional Materials, vol.15, issue.11, pp.1771-1780, 2005.

J. Zhang, N. J. Fredin, J. F. Janz, B. Sun, and D. M. Lynn, Structure/Property Relationships in Erodible Multilayered Films: Influence of Polycation Structure on Erosion Profiles and the Release of Anionic Polyelectrolytes, vol.22, pp.239-245, 2006.

O. Etienne, A. Schneider, C. Taddei, L. Richert, P. Schaaf et al., Degradability of Polysaccharides Multilayer Films in the Oral Environment: an In Vitro and In Vivo Study, Biomacromolecules, vol.6, issue.2, pp.726-733, 2005.

C. H. Porcel and J. B. Schlenoff, Compact Polyelectrolyte Complexes, Candidates for Biomaterials. Biomacromolecules, vol.10, issue.11, pp.2968-2975, 2009.

F. Shamoun-rabih, A. Reisch, and B. Schlenoff-joseph, Extruded Saloplastic Polyelectrolyte Complexes, Advanced Functional Materials, vol.2012, issue.9, pp.1923-1931

R. A. Ghostine and J. B. Schlenoff, Ion Diffusion Coefficients through Polyelectrolyte Multilayers: Temperature and Charge Dependence, Langmuir, vol.27, issue.13, pp.8241-8247, 2011.
DOI : 10.1021/la2015258

J. Fu, Q. Wang, and J. B. Schlenoff, Extruded Superparamagnetic Saloplastic Polyelectrolyte Nanocomposites, ACS Applied Materials and Interfaces, vol.7, issue.1, pp.895-901, 2015.
DOI : 10.1021/am5074694

R. R. Costa, A. M. Costa, S. G. Caridade, and J. F. Mano, Compact Saloplastic Membranes of Natural Polysaccharides for Soft Tissue Engineering, Chemistry of Materials, vol.27, pp.7490-7502, 2015.

M. N. Rodrigues, M. B. Oliveira, R. R. Costa, and J. F. Mano, Chitosan/Chondroitin Sulfate Membranes Produced by Polyelectrolyte Complexation for Cartilage Engineering, Biomacromolecules, vol.17, issue.6, pp.2178-2188, 2016.
DOI : 10.1021/acs.biomac.6b00399

P. Tirado, A. Reisch, E. Roger, F. Boulmedais, L. Jierry et al., Catalytic Saloplastics: Alkaline Phosphatase Immobilized and Stabilized in Compacted Polyelectrolyte Complexes, Advanced Functional Materials, vol.23, issue.38, pp.4785-4792, 2013.
DOI : 10.1002/adfm.201300117

URL : https://hal.archives-ouvertes.fr/hal-01389162

H. H. Hariri and J. B. Schlenoff, Saloplastic Macroporous Polyelectrolyte Complexes: Cartilage Mimics, Macromolecules, vol.2010, issue.20, pp.8656-8663
DOI : 10.1021/ma1012978

URL : https://doi.org/10.1021/ma1012978

H. H. Hariri, A. M. Lehaf, and J. B. Schlenoff, Mechanical Properties of Osmotically Stressed Polyelectrolyte Complexes and Multilayers: Water as a Plasticizer, Macromolecules, vol.2012, issue.23, pp.9364-9372

A. Reisch, P. Tirado, E. Roger, F. Boulmedais, D. Collin et al., Compact Saloplastic Poly(Acrylic Acid)/Poly(Allylamine) Complexes: Kinetic Control Over Composition, Microstructure, and Mechanical Properties, Advanced Functional Materials, vol.2012, issue.6, pp.673-682
DOI : 10.1002/adfm.201201413

URL : https://hal.archives-ouvertes.fr/hal-00966799

A. Reisch, E. Roger, T. Phoeung, C. Antheaume, C. Orthlieb et al., On the Benefits of Rubbing Salt in the Cut: Self-Healing of Saloplastic PAA/PAH Compact Polyelectrolyte Complexes, Advanced Materials, vol.26, issue.16, pp.2547-2551, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01389168

P. G. Agache, C. Monneur, J. L. Leveque, and J. De-rigal, Mechanical Properties and Young's Modulus of Human Skin In Vivo, Archives of Dermatological Research, vol.269, issue.3, pp.221-232, 1980.

T. Phoeung, M. V. Spanedda, E. Roger, B. Heurtault, S. Fournel et al., Alginate/Chitosan Compact Polyelectrolyte Complexes: A Cell and Bacterial Repellent Material, Chemistry of Materials, vol.29, issue.24, pp.10418-10425, 2017.
DOI : 10.1021/acs.chemmater.7b03863

URL : https://hal.archives-ouvertes.fr/hal-02142349

M. Rinaudo, Main Properties and Current Applications of Some Polysaccharides as Biomaterials, Polymer International, vol.57, issue.3, pp.397-430, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00303873

K. Y. Lee and D. J. Mooney, Alginate: Properties and Biomedical Applications. Progress in Polymer Science, vol.37, pp.106-126, 2012.

S. N. Pawar and K. J. Edgar, Alginate Derivatization: A Review of Chemistry, Properties and Applications, Biomaterials, vol.2012, issue.11, pp.3279-3305

N. H. Chamberlain, G. E. Cunningham, and J. B. Speakman, Alginic Acid Diacetate, Nature, vol.1946, issue.4016, pp.553-553

A. Wassermann, Alginic Acid Acetate, Journal of the Chemical Society, vol.1948, issue.0, pp.197-198

R. G. Schweiger, Acetylation of Alginic Acid. I. Preparation and Viscosities of Algin Acetates, The Journal of Organic Chemistry, vol.27, issue.5, pp.1786-1789, 1962.

R. G. Schweiger, Acetylation of Alginic Acid. II. Reaction of Algin Acetates with Calcium and Other Divalent Ions. The Journal of Organic Chemistry, vol.27, pp.1789-1791, 1962.

G. Skjåk-braek, S. Paoletti, and T. Gianferrara, Selective Acetylation of Mannuronic Acid Residues in Calcium Alginate Gels, Carbohydrate Research, vol.185, issue.1, pp.119-129, 1989.

S. N. Pawar and K. J. Edgar, Chemical Modification of Alginates in Organic Solvent Systems, Biomacromolecules, vol.12, issue.11, pp.4095-4103, 2011.

R. J. Coleman, G. Lawrie, L. K. Lambert, M. Whittaker, K. S. Jack et al., Phosphorylation of Alginate: Synthesis, Characterization, and Evaluation of In Vitro Mineralization Capacity, Biomacromolecules, vol.12, issue.4, pp.889-897, 2011.

H. Ronghua, D. Yumin, and Y. Jianhong, Preparation and In Vitro Anticoagulant Activities of Alginate Sulfate and its Quaterized Derivatives, Carbohydrate Polymers, vol.52, issue.1, pp.19-24, 2003.

I. Freeman, A. Kedem, and S. Cohen, The Effect of Sulfation of Alginate Hydrogels on the Specific Binding and Controlled Release of Heparin-Binding Proteins, Biomaterials, vol.29, issue.22, pp.3260-3268, 2008.

L. Fan, L. Jiang, Y. Xu, Y. Zhou, Y. Shen et al., Synthesis and Anticoagulant Activity of Sodium Alginate Sulfates, Carbohydrate Polymers, vol.83, issue.4, pp.1797-1803, 2011.

M. Carré, C. Delestre, P. Hubert, and E. Dellacherie, Covalent Coupling of a Short Polyether on Sodium Alginate: Synthesis and Characterization of the Resulting Amphiphilic Derivative, Carbohydrate Polymers, vol.16, issue.4, pp.367-379, 1991.

A. Sinquin, P. Hubert, and E. Dellacherie, Amphiphilic Derivatives of Alginate: Evidence for Intra-and Intermolecular Hydrophobic Associations in Aqueous Solution, Langmuir, vol.9, issue.12, pp.3334-3337, 1993.

A. Sinquin, P. Hubert, and E. Dellacherie, Intermolecular Associations in Hydrophobically Modified Derivatives of Propyleneglycol Alginate, Polymer, vol.35, issue.16, pp.3557-3560, 1994.

A. Sinquin, M. C. Houzelle, P. Hubert, L. Choplin, M. L. Viriot et al., Amphiphilic Derivatives of Propylene Glycol Alginate: A Revisit of Their Physicochemical Behavior in Dilute Aqueous Solution, Langmuir, vol.12, issue.16, pp.3779-3782, 1996.

V. G. Babak, E. A. Skotnikova, I. G. Lukina, S. Pelletier, P. Hubert et al., Hydrophobically Associating Alginate Derivatives: Surface Tension Properties of their Mixed Aqueous Solutions with Oppositely Charged Surfactants, Journal of Colloid and Interface Science, vol.225, issue.2, pp.505-510, 2000.

C. Galant, A. L. Kjøniksen, G. T. Nguyen, K. D. Knudsen, and B. Nyström, Altering Associations in Aqueous Solutions of a Hydrophobically Modified Alginate in the Presence of Beta-Cyclodextrin Monomers, The Journal of Physical Chemistry B, vol.110, issue.1, pp.190-195, 2006.

I. Donati, A. Vetere, A. Gamini, G. Skjåk-braek, A. Coslovi et al., Galactose-Substituted Alginate: Preliminary Characterization and Study of Gelling Properties, Biomacromolecules, vol.4, issue.3, pp.624-631, 2003.

I. Donati, K. I. Draget, M. Borgogna, S. Paoletti, and G. Skjåk-braek, Tailor-Made Alginate Bearing Galactose Moieties on Mannuronic Residues: Selective Modification Achieved by a Chemoenzymatic Strategy, vol.6, pp.88-98, 2005.

J. Yang, M. Goto, H. Ise, C. S. Cho, and T. Akaike, Galactosylated Alginate as a Scaffold for Hepatocytes Entrapment, Biomaterials, vol.23, issue.2, pp.471-479, 2002.

J. A. Rowley, G. Madlambayan, and D. J. Mooney, Alginate Hydrogels as Synthetic Extracellular Matrix Materials, Biomaterials, vol.20, issue.1, pp.45-53, 1999.

S. B. Shah, C. P. Patel, and H. C. Trivedi, Ceric-Induced Grafting of Acrylate Monomers onto Sodium Alginate, Carbohydrate Polymers, vol.26, issue.1, pp.61-67, 1995.

S. B. Shah, C. P. Patel, and H. C. Trivedi, Fenton's Reagent-Initiated Graft Copolymerization of Acrylonitrile onto Sodium Alginate, Journal of Applied Polymer Science, vol.51, issue.8, pp.1421-1426, 1994.

T. Tripathy, S. R. Pandey, N. C. Karmakar, R. P. Bhagat, and R. P. Singh, Novel Flocculating Agent based on Sodium Alginate and Acrylamide, European Polymer Journal, vol.35, issue.11, pp.2057-2072, 1999.

S. Yang, G. Liu, Y. Cheng, and Y. Zheng, Electroresponsive Behavior of Sodium Alginate-g-Poly (acrylic acid) Hydrogel under DC Electric Field, Journal of Macromolecular Science, Part A, vol.46, issue.11, pp.1078-1082, 2009.

P. Ma, C. Xiao, L. Li, H. Shi, and M. Zhu, Facile Preparation of Ferromagnetic Alginate-g-Poly(vinyl alcohol) Microparticles, European Polymer Journal, vol.44, issue.11, pp.3886-3889, 2008.

Y. Omagari, Y. Kaneko, and J. Kadokawa, Chemoenzymatic Synthesis of AmyloseGrafted Alginate and its Formation of Enzymatic Disintegratable Beads, Carbohydrate Polymers, vol.82, issue.2, pp.394-400, 2010.

J. H. Kim, S. B. Lee, S. J. Kim, and Y. M. Lee, Rapid Temperature/pH Response of Porous Alginate-g-Poly(N-isopropylacrylamide) Hydrogels. Polymer, vol.43, pp.7549-7558, 2002.

C. Gao, M. Liu, S. Chen, S. Jin, and J. Chen, dimethylamino) ethyl methacrylate) Gel Beads and In Vitro Controlled Release Behavior of BSA, International Journal of Pharmaceutics, vol.371, issue.2, pp.16-24, 2009.

J. C. Sun and H. P. Tan, Alginate-Based Biomaterials for, Regenerative Medicine Applications. Materials, vol.6, issue.4, pp.1285-1309, 2013.

Y. Suzuki, M. Tanihara, K. Suzuki, A. Saitou, W. Sufan et al., Alginate Hydrogel Linked with Synthetic Oligopeptide Derived from BMP-2 Allows Ectopic Osteoinduction In Vivo, Journal of Biomedical Materials Research, vol.50, issue.3, pp.405-409, 2000.

J. Leor, S. Tuvia, V. Guetta, F. Manczur, D. Castel et al., Intracoronary Injection of In Situ Forming Alginate Hydrogel Reverses Left Ventricular Remodeling After Myocardial Infarction in Swine, Journal of the American College of Cardiology, vol.54, issue.11, pp.1014-1023, 2009.

K. T. Campbell, D. J. Hadley, D. L. Kukis, and E. A. Silva, Alginate Hydrogels Allow for Bioactive and Sustained Release of VEGF-C and VEGF-D for Lymphangiogenic Therapeutic Applications, PLoS One, vol.12, issue.7, p.181484, 2017.

X. Hao, E. A. Silva, A. Månsson-broberg, K. H. Grinnemo, A. J. Siddiqui et al., Angiogenic Effects of Sequential Release of VEGF-A165 and PDGF-BB with Alginate Hydrogels after Myocardial Infarction, Cardiovascular Research, vol.75, issue.1, pp.178-185, 2007.

S. J. Bidarra, C. C. Barrias, and P. L. Granja, Injectable Alginate Hydrogels for Cell Delivery in Tissue Engineering, Acta Biomaterialia, vol.10, issue.4, pp.1646-1662, 2014.

R. Rastogi, Y. Sultana, M. Aqil, A. Ali, S. Kumar et al., Alginate Microspheres of Isoniazid for Oral Sustained Drug Delivery, International Journal of Pharmaceutics, vol.334, issue.1-2, pp.71-77, 2007.

M. L. Soni, M. Kumar, and K. P. Namdeo, Sodium Alginate Microspheres for Extending Drug Release: Formulation and In Vitro Evaluation, International Journal of Drug Delivery, vol.2010, issue.1, pp.64-68

P. S. Rajinikanth, C. Sankar, and B. Mishra, Sodium Alginate Microspheres of Metoprolol Tartrate for Intranasal Systemic Delivery: Development and Evaluation, Drug Delivery, vol.10, issue.1, pp.21-28, 2003.

M. K. Das and P. C. Senapati, Furosemide-Loaded Alginate Microspheres Prepared by Ionic Cross-Linking Technique: Morphology and Release Characteristics, Indian Journal of Pharmaceutical Sciences, vol.70, issue.1, pp.77-84, 2008.

J. S. Patil, K. Devi, K. Devi, and S. Suresh, Formulation and Evaluation of Novel Spray-Dried Alginate Microspheres as Pulmonary Delivery Systems of Rifampicin in Rats, Indian Journal of Pharmaceutical Education and Research, vol.49, issue.4, pp.320-328, 2015.

K. Nagasree, G. V. Chowdary, C. B. Mahendra-kumar, T. Rama-mohan-reddy, and D. V. Bhikshapathi, Design and Evaluation of Sodium Alginate Microspheres Loaded with Gatifloxacin, Der Pharmacia Lettre, vol.8, issue.4, pp.361-370, 2016.

E. Bulut and M. Dilek, Development and Characterization of pH-Sensitive Locust Bean Gum-Alginate Microspheres for Controlled Release of Ibuprofen, Journal of Drug Delivery Science and Technology, vol.24, issue.6, pp.613-619, 2014.

F. Maestrelli, P. Mura, M. L. González-rodríguez, M. J. Cózar-bernal, A. M. Rabasco et al., Calcium Alginate Microspheres Containing Metformin Hydrochloride Niosomes and Chitosomes Aimed for Oral Therapy of Type 2

, Diabetes Mellitus. International Journal of Pharmaceutics, vol.2017, issue.1-2, pp.430-439

S. Martins, B. Sarmento, E. B. Souto, and D. C. Ferreira, Insulin-Loaded Alginate Microspheres for Oral Delivery-Effect of Polysaccharide Reinforcement on Physicochemical Properties and Release Profile, Carbohydrate Polymers, vol.69, issue.4, pp.725-731, 2007.

G. Ciofani, V. Raffa, A. Menciassi, S. Micera, and P. Dario, A Drug Delivery System Based on Alginate Microspheres: Mass-Transport Test and In Vitro Validation, Biomedical Microdevices, vol.9, issue.3, pp.395-403, 2007.

J. Yu, K. T. Du, Q. Fang, Y. Gu, S. S. Mihardja et al., The Use of Human Mesenchymal Stem Cells Encapsulated in RGD Modified Alginate Microspheres in the Repair of Myocardial Infarction in the Rat, Biomaterials, vol.2010, issue.27, pp.7012-7020

S. J. Bidarra, C. C. Barrias, M. A. Barbosa, R. Soares, and P. L. Granja, Immobilization of Human Mesenchymal Stem Cells within RGD-Grafted Alginate Microspheres and Assessment of their Angiogenic Potential, Biomacromolecules, vol.2010, issue.8, pp.1956-1964

A. Moshaverinia, S. Ansari, C. Chen, X. Xu, K. Akiyama et al., Co-Encapsulation of Anti-BMP2 Monoclonal Antibody and Mesenchymal Stem Cells in Alginate Microspheres for Bone Tissue Engineering, Biomaterials, vol.2013, issue.28, pp.6572-6579

R. G. Thomas, A. R. Unnithan, M. J. Moon, S. P. Surendran, T. Batgerel et al., Electromagnetic Manipulation Enabled Calcium Alginate Janus Microsphere for Targeted Delivery of Mesenchymal Stem Cells, International Journal of Biological Macromolecules, vol.110, pp.465-471, 2018.

N. Aggarwal, H. Hogenesch, P. Guo, A. North, M. Suckow et al., Biodegradable Alginate Microspheres as a Delivery System for Naked DNA, Canadian Journal of Veterinary Research, vol.63, issue.2, pp.148-152, 1999.

J. Y. Tian, X. Q. Sun, and X. G. Chen, Formation and Oral Administration of Alginate Microspheres Loaded with pDNA Coding for Lymphocystis Disease Virus (LCDV) to

, Japanese Flounder. Fish and Shellfish Immunology, vol.24, issue.5, pp.592-599, 2008.

C. C. Ribeiro, C. C. Barrias, and M. A. Barbosa, Calcium Phosphate-Alginate Microspheres as Enzyme Delivery Matrices, Biomaterials, vol.25, issue.18, pp.4363-4373, 2004.

X. Qi, J. Ye, and Y. Wang, Alginate/Poly (Lactic-co-Glycolic Acid)/Calcium Phosphate Cement Scaffold with Oriented Pore Structure for Bone Tissue Engineering, Journal of Biomedical Materials Research A, vol.89, issue.4, pp.980-987, 2009.

D. Singh, A. Tripathi, S. Zo, and S. S. Han, Synthesis of Composite GelatinHyaluronic Acid-Alginate Porous Scaffold and Evaluation for In Vitro Stem Cell Growth and In Vivo Tissue Integration, Colloids and Surfaces B Biointerfaces, vol.116, pp.502-509, 2014.

C. C. Wang, K. C. Yang, K. H. Lin, H. C. Liu, and F. H. Lin, A Highly Organized Three-Dimensional Alginate Scaffold for Cartilage Tissue Engineering Prepared by Microfluidic Technology, Biomaterials, vol.32, issue.29, pp.7118-7126, 2011.

M. Rinaudo, Chitin and Chitosan: Properties and Applications. Progress in Polymer Science, vol.31, pp.603-632, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00305792

F. Croisier and C. Jérôme, Chitosan-Based Biomaterials for Tissue Engineering, European Polymer Journal, vol.49, issue.4, pp.780-792, 2013.

M. N. Kumar, R. A. Muzzarelli, C. Muzzarelli, H. Sashiwa, and A. J. Domb, Chitosan Chemistry and Pharmaceutical Perspectives, Chemical Reviews, vol.104, issue.12, pp.6017-6084, 2004.
DOI : 10.1002/chin.200511296

I. K. Park, J. Yang, H. J. Jeong, H. S. Bom, I. Harada et al., Galactosylated Chitosan as a Synthetic Extracellular Matrix for Hepatocytes Attachment, Biomaterials, vol.24, issue.13, pp.2331-2337, 2003.

E. R. Welsh and R. R. Price, Chitosan Cross-Linking with a Water-Soluble, Blocked Diisocyanate. 2. Solvates and Hydrogels, Biomacromolecules, vol.4, issue.5, pp.1357-1361, 2003.

A. Chenite, C. Chaput, D. Wang, C. Combes, M. D. Buschmann et al., Novel Injectable Neutral Solutions of Chitosan Form Biodegradable Gels In Situ, Biomaterials, vol.21, issue.21, pp.2155-2161, 2000.

M. Moattari, H. M. Kouchesfehani, G. Kaka, S. H. Sadraie, M. Naghdi et al., Chitosan-Film Associated with Mesenchymal Stem Cells Enhanced Regeneration of Peripheral Nerves: A Rat Sciatic Nerve Model, Journal of Chemical Neuroanatomy, vol.88, pp.46-54, 2018.

S. N. Mohd and A. K. Mat, Composite Film of Chitosan Loaded Norfloxacin with Improved Flexibility and Antibacterial Activity for Wound Dressing Application, Oriental Journal of Chemistry, vol.33, issue.2, pp.628-636, 2017.

D. K. Khajuria, O. N. Patil, D. Karasik, and R. Razdan, Development and Evaluation of Novel Biodegradable Chitosan Based Metformin Intrapocket Dental Film for the Management of Periodontitis and Alveolar Bone Loss in a Rat Model, Archives of Oral Biology, vol.85, pp.120-129, 2018.

P. Ghosh, A. P. Rameshbabu, D. Das, N. K. Francis, H. S. Pawar et al., Covalent Cross-Links in Polyampholytic Chitosan Fibers Enhances Bone Regeneration in a Rabbit Model, Colloids and Surfaces B Biointerfaces, vol.125, pp.160-169, 2015.

P. Yilgor, K. Tuzlakoglu, R. L. Reis, N. Hasirci, and V. Hasirci, Incorporation of a Sequential BMP-2/BMP-7 Delivery System into Chitosan-Based Scaffolds for Bone Tissue Engineering, Biomaterials, vol.30, issue.21, pp.3551-3559, 2009.

S. Jia, X. Yang, W. Song, L. Wang, K. Fang et al., Incorporation of Osteogenic and Angiogenic Small Interfering RNAs into Chitosan Sponge for Bone Tissue Engineering, International Journal of Nanomedicine, vol.9, pp.5307-5316, 2014.

A. Echazú, M. I. Olivetti, C. E. Anesini, C. Perez, C. J. Alvarez et al., Development and Evaluation of Thymol-Chitosan Hydrogels with Antimicrobial-Antioxidant Activity for Oral Local Delivery, Materials Science and Engineering C, vol.81, pp.588-596, 2017.

D. Aycan and N. Alemdar, Development of pH-Responsive Chitosan-Based Hydrogel Modified with Bone Ash for Controlled Release of Amoxicillin, Carbohydrate Polymers, vol.184, pp.401-407, 2018.

M. Lu, Y. Liu, Y. C. Huang, C. J. Huang, and W. B. Tsai, Fabrication of PhotoCrosslinkable Glycol Chitosan Hydrogel as a Tissue Adhesive, Carbohydrate Polymers, vol.181, pp.668-674, 2018.

T. Fatmanur, Development of In Situ Poloxamer-Chitosan Hydrogels for Vaginal Drug Delivery of Benzydamine Hydrochloride: Textural, Mucoadhesive and In Vitro Release Properties, Marmara Pharmaceutical Journal, vol.21, issue.4, pp.762-770, 2017.

W. Xie, Q. Gao, Z. Guo, D. Wang, F. Gao et al., Injectable and Self-Healing Thermosensitive Magnetic Hydrogel for Asynchronous Control Release of Doxorubicin and Docetaxel to Treat Triple-Negative Breast Cancer, ACS Applied Materials and Interfaces, vol.2017, issue.39, pp.33660-33673

X. Qiao, X. Peng, J. Qiao, Z. Jiang, B. Han et al., Evaluation of a Photocrosslinkable Hydroxyethyl Chitosan Hydrogel as a Potential Drug Release System for Glaucoma Surgery, Journal of Materials Science Materials in Medicine, vol.2017, issue.10, p.149

K. Bankoti, A. P. Rameshbabu, S. Datta, P. P. Maity, P. Goswami et al., Accelerated Healing of Full Thickness Dermal Wounds by Macroporous Waterborne Polyurethane-Chitosan Hydrogel Scaffolds, Materials Science and Engineering C, vol.81, pp.133-143, 2017.

S. J. Yoon, H. Hyun, D. W. Lee, and D. H. Yang, Visible Light-Cured Glycol Chitosan Hydrogel Containing a Beta-Cyclodextrin-Curcumin Inclusion Complex Improves Wound Healing In Vivo, Molecules, vol.2017, issue.9, p.1513

Q. Q. Ouyang, Z. Hu, Z. P. Lin, W. Y. Quan, Y. F. Deng et al., Chitosan Hydrogel in Combination with Marine Peptides from Tilapia for Burns Healing, International Journal of Biological Macromolecules, vol.112, pp.1191-1198, 2018.

M. Zhai, Y. Xu, B. Zhou, and W. Jing, Keratin-Chitosan/n-ZnO Nanocomposite Hydrogel for Antimicrobial Treatment of Burn Wound Healing: Characterization and Biomedical Application, Journal of Photochemistry and Photobiology, B: Biology, vol.180, pp.253-258, 2018.

M. Kashi, F. Baghbani, F. Moztarzadeh, H. Mobasheri, and E. Kowsari, Green Synthesis of Degradable Conductive Thermosensitive Oligopyrrole/Chitosan Hydrogel Intended for Cartilage Tissue Engineering, International Journal of Biological Macromolecules, vol.107, pp.1567-1575, 2018.

Z. Li, H. Shim, M. O. Cho, I. S. Cho, J. H. Lee et al., Thermo-Sensitive Injectable Glycol Chitosan-Based Hydrogel for Treatment of Degenerative Disc Disease, Carbohydrate Polymers, vol.184, pp.342-353, 2018.

B. Xu, Y. Li, B. Deng, X. Liu, L. Wang et al., Chitosan Hydrogel Improves Mesenchymal Stem Cell Transplant Survival and Cardiac Function Following Myocardial Infarction in Rats, vol.13, pp.588-594, 2017.

J. Huang, Y. Deng, J. Ren, G. Chen, G. Wang et al., Novel In Situ Forming Hydrogel based on Xanthan and Chitosan Re-Gelifying in Liquids for Local Drug Delivery, Carbohydrate Polymers, vol.186, pp.54-63, 2018.

K. Kiene, F. Porta, B. Topacogullari, P. Detampel, and J. Huwyler, Self-Assembling Chitosan Hydrogel: A Drug-Delivery Device Enabling the Sustained Release of Proteins, Journal of Applied Polymer Science, vol.2017, issue.1, p.45638

A. B. Dhanikula and R. Panchagnula, Development and Characterization of Biodegradable Chitosan Films for Local Delivery of Paclitaxel, AAPS Journal, vol.6, issue.3, pp.88-99, 2004.

L. B. Rodrigues, H. F. Leite, M. I. Yoshida, J. B. Saliba, A. S. Cunha et al., In Vitro Release and Characterization of Chitosan Films as Dexamethasone Carrier, International Journal of Pharmaceutics, vol.368, issue.1-2, pp.1-6, 2009.

J. K. Smith, J. D. Bumgardner, H. S. Courtney, M. S. Smeltzer, and W. O. Haggard, Antibiotic-Loaded Chitosan Film for Infection Prevention: A Preliminary In Vitro Characterization, Journal of Biomedical Materials Research B, vol.2010, issue.1, pp.203-211

S. P. Noel, H. Courtney, J. D. Bumgardner, and W. O. Haggard, Chitosan Films: A Potential Local Drug Delivery System for Antibiotics, Clinical Orthopaedics and Related Research, vol.466, issue.6, pp.1377-1382, 2008.

N. Bavarsad, M. Kouchak, P. Mohamadipour, and B. Sadeghi-nejad, Preparation and Physicochemical Characterization of Topical Chitosan-Based Film Containing GriseofulvinLoaded Liposomes, Journal of Advanced Pharmaceutical Technology and Research, vol.7, issue.3, pp.91-98, 2016.

Y. Y. Wu, Y. P. Jiao, L. L. Xiao, M. M. Li, H. W. Liu et al., Experimental Study on Effects of Adipose-Derived Stem Cell-Seeded Silk Fibroin Chitosan Film on Wound Healing of a Diabetic Rat Model, Annals of Plastic Surgery, vol.80, issue.5, pp.572-580, 2018.

A. Abarrategi, A. Civantos, V. Ramos, J. V. Sanz-casado, and J. L. López-lacomba, Chitosan Film as rhBMP2 Carrier: Delivery Properties for Bone Tissue Application, Biomacromolecules, vol.9, issue.2, pp.711-718, 2008.

H. Liu, X. Qu, E. Kim, M. Lei, K. Dai et al., Bio-Inspired Redox-Cycling Antimicrobial Film for Sustained Generation of Reactive Oxygen Species, Biomaterials, vol.162, pp.109-122, 2018.

M. Ignatova, N. Manolova, and I. Rashkov, Electrospun Antibacterial Chitosan-Based Fibers, Macromolecular Bioscience, vol.13, issue.7, pp.860-872, 2013.

M. Ignatova, N. Manolova, and I. Rashkov, Novel Antibacterial Fibers of Quaternized Chitosan and Poly(vinyl pyrrolidone) Prepared by Electrospinning, European Polymer Journal, vol.43, issue.4, pp.1112-1122, 2007.

M. Arvand, E. Mirzaei, . ;-derakhshan, A. Mohammad, S. Kharrazi et al., Fabrication of Antibacterial Silver Nanoparticle-Modified Chitosan Fibers using Eucalyptus Extract as a Reducing Agent, Journal of Applied Polymer Science, vol.132, issue.25, p.42133, 2015.

Z. Zhou, D. Yan, X. Cheng, M. Kong, Y. Liu et al., Biomaterials Based on N,N,N-Trimethyl Chitosan Fibers in Wound Dressing Applications, International Journal of Biological Macromolecules, vol.89, pp.471-476, 2016.

Y. Zhou, H. Yang, X. Liu, J. Mao, S. Gu et al., Potential of QuaternizationFunctionalized Chitosan Fiber for Wound Dressing, International Journal of Biological Macromolecules, vol.52, pp.327-332, 2013.

A. Cooper, N. Bhattarai, and M. Zhang, Fabrication and Cellular Compatibility of Aligned Chitosan-PCL Fibers for Nerve Tissue Regeneration, Carbohydrate Polymers, vol.85, issue.1, pp.149-156, 2011.

T. Funakoshi, T. Majima, N. Iwasaki, S. Yamane, T. Masuko et al., Novel Chitosan-Based Hyaluronan Hybrid Polymer Fibers as a Scaffold in Ligament Tissue Engineering, Journal of Biomedical Materials Research A, vol.74, issue.3, pp.338-346, 2005.

Q. Lian, D. Li, Z. Jin, J. Wang, A. Li et al., Fabrication and In Vitro Evaluation of Calcium Phosphate Combined with Chitosan Fibers for Scaffold Structures, Journal of Bioactive and Compatible Polymers, vol.24, issue.1_suppl, pp.113-124, 2009.

K. Tuzlakoglu, C. M. Alves, J. F. Mano, and R. L. Reis, Production and Characterization of Chitosan Fibers and 3-D Fiber Mesh Scaffolds for Tissue Engineering Applications, Macromolecular Bioscience, vol.4, issue.8, pp.811-819, 2004.

X. Huang, X. Bao, Z. Wang, and Q. Hu, A Novel Silver-Loaded Chitosan Composite Sponge with Sustained Silver Release as a Long-Lasting Antimicrobial Dressing, vol.7, pp.34655-34663, 2017.

D. T. Vo and C. K. Lee, Antimicrobial Sponge Prepared by Hydrophobically Modified Chitosan for Bacteria Removal, Carbohydrate Polymers, vol.187, pp.1-7, 2018.

D. Revi, W. Paul, T. V. Anilkumar, and C. P. Sharma, Chitosan Scaffold Co-Cultured with Keratinocyte and Fibroblast Heals Full Thickness Skin Wounds in Rabbit, Journal of Biomedical Materials Research A, vol.2014, issue.9, pp.3273-3281

B. Lu, T. Wang, Z. Li, F. Dai, L. Lv et al., Healing of Skin Wounds with a Chitosan-Gelatin Sponge Loaded with Tannins and PlateletRich Plasma, International Journal of Biological Macromolecules, vol.82, pp.884-891, 2016.

D. Liang, Z. Lu, H. Yang, J. Gao, and R. Chen, Novel Asymmetric Wettable AgNPs/Chitosan Wound Dressing: In Vitro and In Vivo Evaluation, ACS Applied Materials and Interfaces, vol.8, issue.6, pp.3958-3968, 2016.

F. Han, Y. Dong, Z. Su, R. Yin, A. Song et al., Preparation, Characteristics and Assessment of a Novel Gelatin-Chitosan Sponge Scaffold as Skin Tissue Engineering Material, International Journal of Pharmaceutics, vol.476, issue.1-2, pp.124-133, 2014.

Z. Lu, J. Gao, Q. He, J. Wu, D. Liang et al., Enhanced Antibacterial and Wound Healing Activities of Microporous Chitosan-Ag/ZnO Composite Dressing, Carbohydrate Polymers, vol.156, pp.460-469, 2017.

A. C. Parker, K. E. Beenken, J. A. Jennings, L. Hittle, M. E. Shirtliff et al., Characterization of Local Delivery with

B. Amphotericin, Vancomycin from Modified Chitosan Sponges and Functional Biofilm Prevention Evaluation, Journal of Orthopaedic Research, vol.33, issue.3, pp.439-447, 2015.

J. A. Jennings, K. E. Beenken, A. C. Parker, J. K. Smith, H. S. Courtney et al., Polymicrobial Biofilm Inhibition Effects of AcetateBuffered Chitosan Sponge Delivery Device, Macromolecular Bioscience, vol.16, issue.4, pp.591-598, 2016.

M. A. Kassem, A. N. Elmeshad, and A. R. Fares, Lyophilized Sustained Release Mucoadhesive Chitosan Sponges for Buccal Buspirone Hydrochloride Delivery: Formulation and In Vitro Evaluation, AAPS PharmSciTech, vol.16, issue.3, pp.537-547, 2015.

J. Chen, Y. Li, B. Wang, J. Yang, B. C. Heng et al., TGFBeta1 Affinity Peptides Incorporated within a Chitosan Sponge Scaffold can Significantly Enhance Cartilage Regeneration, Journal of Materials Chemistry B, vol.6, issue.4, pp.675-687, 2018.

G. Crini, Review: A History of Cyclodextrins, Chemical Reviews, vol.114, issue.21, pp.10940-10975, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01089582

D. Valle and E. M. , Cyclodextrins and their Uses: A Review, Process Biochemistry, vol.39, issue.9, pp.1033-1046, 2004.

T. A. Reineccius, G. A. Reineccius, and T. L. Peppard, Encapsulation of Flavors using Cyclodextrins: Comparison of Flavor Retention in Alpha, Beta, and Gamma Types, Journal of Food Science, vol.67, issue.9, pp.3271-3279, 2006.

P. E. Shaw, J. H. Tatum, and C. W. Wilson, Improved Flavor of Navel Orange and Grapefruit Juices by Removal of Bitter Components with Beta-Cyclodextrin Polymer, Journal of Agricultural and Food Chemistry, vol.32, issue.4, pp.832-836, 1984.

U. Numano?lu, T. Sen, N. Tarimci, M. Kartal, O. M. Koo et al., Use of Cyclodextrins as a Cosmetic Delivery System for Fragrance Materials: Linalool and Benzyl Acetate, AAPS PharmSciTech, vol.8, issue.4, p.85, 2007.

S. Scalia, A. Molinari, A. Casolari, and A. Maldotti, Complexation of the Sunscreen Agent, Phenylbenzimidazole Sulphonic Acid with Cyclodextrins: Effect on Stability and Photo-Induced Free Radical Formation, European Journal of Pharmaceutical Sciences, vol.22, issue.4, pp.241-249, 2004.

S. Scalia, S. Villani, A. Scatturin, M. A. Vandelli, and F. Forni, Complexation of the Sunscreen Agent, Butyl-Methoxydibenzoylmethane, with Hydroxypropyl-?-Cyclodextrin, International Journal of Pharmaceutics, vol.175, issue.2, pp.205-213, 1998.

S. Scalia, R. Tursilli, and V. Iannuccelli, Complexation of the Sunscreen Agent, 4Methylbenzylidene Camphor with Cyclodextrins: Effect on Photostability and Human Stratum Corneum Penetration, Journal of Pharmaceutical and Biomedical Analysis, vol.44, issue.1, pp.29-34, 2007.

M. Tanaka, Y. Iwata, Y. Kouzuki, K. Taniguchi, H. Matsuda et al., Effect of 2-Hydroxypropyl-Beta-Cyclodextrin on Percutaneous Absorption of Methyl Paraben, The Journal of Pharmacy and Pharmacology, vol.47, issue.11, pp.897-900, 1995.

A. Cireli and B. Yurdakul, Application of Cyclodextrin to the Textile Dyeing and Washing Processes, Journal of Applied Polymer Science, vol.100, issue.1, pp.208-218, 2006.

P. Savarino, G. Viscardi, P. Quagliotto, E. Montoneri, and E. Barni, Reactivity and Effects of Cyclodextrins in Textile Dyeing, Dyes and Pigments, vol.42, issue.2, pp.143-147, 1999.

P. Lo-nostro, L. Fratoni, and P. Baglioni, Modification of a Cellulosic Fabric with ?Cyclodextrin for Textile Finishing Applications, Journal of Inclusion Phenomena and Macrocyclic Chemistry, vol.44, issue.1, pp.423-427, 2002.

C. Ortiz-mellet, J. M. García-fernández, and J. M. Benito, Cyclodextrin-Based Gene Delivery Systems, Chemical Society Reviews, vol.40, issue.3, pp.1586-1608, 2011.

F. J. Otero-espinar, J. J. Torres-labandeira, C. Alvarez-lorenzo, and J. Blanco-méndez, Cyclodextrins in Drug Delivery Systems, Journal of Drug Delivery Science and Technology, vol.20, issue.4, pp.289-301, 2010.

S. Hwang-pun, H. Gonzalez, M. E. Davis, N. C. Bellocq, and J. Cheng, Complexing Agents for Compositions Containing Inclusion Complexes, p.20150031753, 2015.

G. Tripodo, C. Wischke, A. T. Neffe, and A. Lendlein, Efficient Synthesis of Pure Monotosylated Beta-Cyclodextrin and its Dimers, Carbohydrate Research, vol.381, pp.59-63, 2013.

M. Xu, S. Wu, F. Zeng, and C. Yu, Cyclodextrin Supramolecular Complex as a WaterSoluble Ratiometric Sensor for Ferric Ion Sensing, Langmuir, issue.6, pp.4529-4534, 2010.

P. Gonil, W. Sajomsang, U. R. Ruktanonchai, N. Pimpha, I. Sramala et al., Novel Quaternized Chitosan Containing ?-Cyclodextrin Moiety: Synthesis, Characterization and Antimicrobial Activity, Carbohydrate Polymers, vol.83, issue.2, pp.905-913, 2011.

J. S. Yang, S. Y. Han, L. Yang, and H. C. Zheng, Synthesis of Beta-CyclodextrinGrafted-Alginate and its Application for Removing Methylene Blue from Water Solution, Journal of Chemical Technology & Biotechnology, vol.91, issue.3, pp.618-623, 2014.

L. Tan, Y. Liu, W. Ha, L. Ding, S. Peng et al., StimuliInduced Gel-Sol Transition of Multi-Sensitive Supramolecular Beta-Cyclodextrin Grafted

, Alginate/Ferrocene Modified Pluronic Hydrogel, Soft Matter, vol.8, issue.21, pp.5746-5749, 2012.

W. Pluemsab, N. Sakairi, and T. Furuike, Synthesis and Inclusion Property of AlphaCyclodextrin-Linked Alginate, Polymer, vol.46, issue.23, pp.9778-9783, 2005.

J. Kohn and M. Wilchek, Mechanism of Activation of Sepharose and Sephadex by Cyanogen Bromide, Enzyme and Microbial Technology, vol.1982, issue.3, pp.161-163

C. Jaime, J. Redondo, F. Sánchez-ferrando, and A. Virgili, ?-Cyclodextrin Inclusion Complex with Adamantane Intermolecular 1H{1H} NOE Determinations and Molecular Mechanics Calculations, Journal of Molecular Structure, vol.248, issue.3, pp.317-329, 1991.

K. Sadrerafi, E. Moore, and M. Lee, Association Constant of ?-Cyclodextrin with Carboranes, Adamantane, and their Derivatives using Displacement Binding Technique, Journal of Inclusion Phenomena and Macrocyclic Chemistry, vol.83, issue.1-2, pp.159-166, 2015.

L. Li, X. Guo, J. Wang, P. Liu, R. K. Prud'homme et al., Polymer Networks Assembled by Host?Guest Inclusion between Adamantyl and ?Cyclodextrin Substituents on Poly(acrylic acid) in Aqueous Solution, Macromolecules, issue.22, pp.8677-8681, 2008.

C. Koopmans and H. Ritter, Formation of Physical Hydrogels via Host?Guest Interactions of ?-Cyclodextrin Polymers and Copolymers Bearing Adamantyl Groups, Macromolecules, issue.20, pp.7418-7422, 2008.

A. Charlot and R. Auzély-velty, Synthesis of Novel Supramolecular Assemblies Based on Hyaluronic Acid Derivatives Bearing Bivalent ?-Cyclodextrin and Adamantane Moieties, Macromolecules, vol.40, issue.4, pp.1147-1158, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00305582

E. Christoforides, A. Papaioannou, and K. Bethanis, Crystal Structure of the Inclusion Complex of Cholesterol in ?-Cyclodextrin and Molecular Dynamics Studies, Beilstein Journal of Organic Chemistry, vol.14, pp.838-848, 2018.

R. Breslow and B. Zhang, Cholesterol Recognition and Binding by Cyclodextrin Dimers, Journal of the American Chemical Society, vol.118, issue.35, pp.8495-8496, 1996.

F. Van-de-manakker, M. Van-der-pot, T. Vermonden, C. F. Van-nostrum, and W. E. Hennink, Self-Assembling Hydrogels Based on ?-Cyclodextrin/Cholesterol Inclusion Complexes, Macromolecules, issue.5, pp.1766-1773, 2008.

H. S. Kwak, S. H. Kim, J. H. Kim, H. J. Choi, and J. Kang, Immobilized BetaCyclodextrin as a Simple and Recyclable Method for Cholesterol Removal in Milk, Archives of Pharmaceutical Research, vol.27, issue.8, pp.873-877, 2004.

S. Kim, E. Han, J. Ahn, and H. Kwak, Effect of Crosslinked ?-Cyclodextrin on Quality of Cholesterol-Reduced Cream Cheese. Asian-Australasian Journal of Animal Sciences, vol.18, issue.4, pp.584-589, 2005.

R. J. Hooley, S. M. Biros, J. Rebek, and . Deep, Water-Soluble Cavitand Acts as a Phase-Transfer Catalyst for Hydrophobic Species, Angewandte Chemie, vol.45, issue.21, pp.3517-3519, 2006.

X. A. Ton, . Tse-sum, B. Bui, M. Resmini, P. Bonomi et al., A Versatile Fiber-Optic Fluorescence Sensor Based on Molecularly Imprinted Microstructures Polymerized In Situ, Angewandte Chemie, vol.52, issue.32, pp.8317-8321, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00866486

D. B. Yang, J. B. Zhu, Z. J. Huang, H. X. Ren, and Z. J. Zheng, Synthesis and Application of Poly(Ethylene Glycol)-Cholesterol (Chol-PEGm) Conjugates in Physicochemical Characterization of Nonionic Surfactant Vesicles, Colloids and Surfaces B Biointerfaces, vol.63, issue.2, pp.192-199, 2008.

T. L. Foley, A. Yasgar, C. J. Garcia, A. Jadhav, A. Simeonov et al., Preparation of FRET Reporters to Support Chemical Probe Development. Organic and Biomolecular Chemistry, vol.8, pp.4601-4606, 2010.

D. Silva, L. F. Pinto, D. Bozukova, L. F. Santos, A. P. Serro et al., Chitosan/Alginate Based Multilayers to Control Drug Release from Ophthalmic Lens, Colloids and Surfaces B Biointerfaces, vol.147, pp.81-89, 2016.

K. Kyung, S. Kim, and S. Shiratori, Preparation and Characterization of Antithrombogenic Chitosan/Alginate Films with Enhanced Physical Stability by CrossLinking Using Layer-By-Layer Method, MATEC Web of Conferences, vol.4, p.5008, 2013.

O. L. Shanmugasundaram, Development and Characterization of Cotton and Organic Cotton Gauze Fabric Coated with Biopolymers and Antibiotic Drugs for Wound Healing, Indian Journal of Fibre and Textile Research, vol.37, pp.146-150, 2012.

O. L. Shanmugasundaram and R. V. Mahendra-gowda, Development and Characterization of Cotton, Organic Cotton Flat Knit Fabrics Coated with Chitosan, Sodium Alginate, Calcium Alginate Polymers, and Antibiotic Drugs for Wound Healing, Journal of Industrial Textiles, vol.42, issue.2, pp.156-175, 2011.

W. Feng, W. Nie, C. He, X. Zhou, L. Chen et al., Effect of pH-Responsive Alginate/Chitosan Multilayers Coating on Delivery Efficiency, Cellular Uptake and Biodistribution of Mesoporous Silica Nanoparticles Based Nanocarriers, ACS Applied Materials and Interfaces, vol.6, issue.11, pp.8447-8460, 2014.

F. M. Kievit, S. J. Florczyk, M. C. Leung, K. Wang, J. D. Wu et al., Proliferation and Enrichment of CD133(+) Glioblastoma Cancer Stem Cells on 3D Chitosan-Alginate Scaffolds, Biomaterials, vol.35, issue.33, pp.9137-9143, 2014.

F. M. Kievit, K. Wang, A. E. Erickson, S. K. Lan-levengood, R. G. Ellenbogen et al., Modeling the Tumor Microenvironment Using Chitosan-Alginate Scaffolds to Control the Stem-Like State of Glioblastoma Cells, Biomaterials Science, vol.4, issue.4, pp.610-613, 2016.

S. J. Florczyk, F. M. Kievit, K. Wang, A. E. Erickson, R. G. Ellenbogen et al., 3D Porous Chitosan-Alginate Scaffolds Promote Proliferation and Enrichment of Cancer Stem-Like Cells, Journal of Materials Chemistry B, vol.2016, issue.38, pp.6326-6334

K. Wang, F. M. Kievit, S. J. Florczyk, Z. R. Stephen, and M. Zhang, 3D Porous Chitosan-Alginate Scaffolds as an In Vitro Model for Evaluating Nanoparticle-Mediated Tumor Targeting and Gene Delivery to Prostate Cancer, Biomacromolecules, vol.2015, issue.10, pp.3362-3372

T. W. Chung, J. Yang, T. Akaike, K. Y. Cho, J. W. Nah et al., Preparation of Alginate/Galactosylated Chitosan Scaffold for Hepatocyte Attachment, Biomaterials, vol.23, issue.14, pp.2827-2834, 2002.

A. P. Bagre, K. Jain, and N. K. Jain, Alginate Coated Chitosan Core Shell Nanoparticles for Oral Delivery of Enoxaparin: In Vitro and In Vivo Assessment, International Journal of Pharmaceutics, vol.456, issue.1, pp.31-40, 2013.

P. Li, Y. N. Dai, J. P. Zhang, A. Q. Wang, and Q. Wei, Chitosan-Alginate Nanoparticles as a Novel Drug Delivery System for Nifedipine, International Journal of Biomedical Science, vol.4, issue.3, pp.221-228, 2008.

R. C. Nagarwal, R. Kumar, and J. K. Pandit, Chitosan Coated Sodium AlginateChitosan Nanoparticles Loaded with 5-FU for Ocular Delivery: In Vitro Characterization and In Vivo Study in Rabbit Eye, European Journal of Pharmaceutical Sciences, vol.2012, issue.4, pp.678-685

S. Biswas, M. Chattopadhyay, K. K. Sen, and M. K. Saha, Development and Characterization of Alginate Coated Low Molecular Weight Chitosan Nanoparticles as New Carriers for Oral Vaccine Delivery in Mice, Carbohydrate Polymers, vol.121, pp.403-410, 2015.

K. Neeraj and C. Shatakshee, Alginate Coated Chitosan Nanoparticles as Potential Oral Vaccine Carriers: Synthesis and In-Vitro Characterization, Nano Science and Nano Technology, vol.5, issue.1, pp.59-62, 2011.

M. A. Azevedo, A. I. Bourbon, A. A. Vicente, and M. A. Cerqueira, Alginate/Chitosan Nanoparticles for Encapsulation and Controlled Release of Vitamin B2, International Journal of Biological Macromolecules, vol.71, pp.141-146, 2014.

F. Maestrelli, M. Jug, M. Cirri, I. Kosalec, and P. Mura, Characterization and Microbiological Evaluation of Chitosan-Alginate Microspheres for Cefixime Vaginal Administration, Carbohydrate Polymers, vol.192, pp.176-183, 2018.

A. El-rasoul, S. Ahmed, and M. , Chitosan Polymer as a Coat of Calcium Alginate Microcapsules Loaded by Non-Steroidal Antiinflammatory Drug, Bulletin of Pharmaceutical Sciences, vol.33, pp.179-187, 2010.

S. ;. Petta, V. R. Kasa, and R. R. , Design and Evaluation of Controlled Release Chitosan-Calcium Alginate Microcapsules of Anti Tubercular Drugs for Oral Use, International Journal of ChemTech Research, vol.2, issue.1, pp.88-98, 2003.

R. Shukla, J. Gupta, P. Shukla, P. Dwivedi, P. Tripathi et al., Chitosan Coated Alginate Micro Particles for the Oral Delivery of Antifilarial Drugs and Combinations for Intervention in Brugia Malayi Induced Lymphatic Filariasis, RSC Advances, vol.5, issue.85, pp.69047-69056, 2015.

V. Araujo, A. Gamboa, N. Caro, L. Abugoch, M. Gotteland et al., Release of Prednisolone and Inulin from a New Calcium-Alginate Chitosan-Coated Matrix System for Colonic Delivery, Journal of Pharmaceutical Sciences, issue.8, pp.2748-2759, 2013.

C. Deka, D. Deka, M. M. Bora, D. K. Jha, and D. K. Kakati, Synthesis of Peppermint Oil-Loaded Chitosan/Alginate Polyelectrolyte Complexes and Study of their Antibacterial Activity, Journal of Drug Delivery Science and Technology, vol.35, pp.314-322, 2016.

C. H. Zheng, W. Q. Liang, F. Li, Y. P. Zhang, and W. J. Fang, Optimization and Characterization of Chitosan-Coated Alginate Microcapsules Containing Albumin, Pharmazie, vol.60, issue.6, pp.434-438, 2005.

A. M. Omer, T. M. Tamer, M. A. Hassan, P. Rychter, M. S. Mohy-eldin et al., Development of Amphoteric Alginate/Aminated Chitosan Coated Microbeads for Oral Protein Delivery, International Journal of Biological Macromolecules, vol.92, pp.362-370, 2016.

H. Onishi, K. Koyama, O. Sakata, and Y. Machida, Preparation of Chitosan/Alginate/Calcium Complex Microparticles Loaded with Lactoferrin and Their Efficacy on Carrageenan-Induced Edema in Rats. Drug Development and Industrial Pharmacy, vol.36, pp.879-884, 2010.

F. Q. Wang, P. Li, J. P. Zhang, A. Q. Wang, and Q. Wei, A Novel pH-Sensitive Magnetic Alginate-Chitosan Beads for Albendazole Delivery. Drug Development and Industrial Pharmacy, vol.36, pp.867-877, 2010.

H. J. Liu, P. Li, and Q. Wei, Magnetic N-Succinyl Chitosan/Alginate Beads for Carbamazepine Delivery. Drug Development and Industrial Pharmacy, vol.36, pp.1286-1294, 2010.

T. W. Wong and H. Nurulaini, Sustained-Release Alginate-Chitosan Pellets Prepared by Melt Pelletization Technique. Drug Development and Industrial Pharmacy, vol.38, pp.1417-1427, 2012.

L. Li, L. Wang, Y. Shao, R. Ni, T. Zhang et al., Drug Release Characteristics from Chitosan-Alginate Matrix Tablets Based on the Theory of Self-Assembled Film, International Journal of Pharmaceutics, vol.450, issue.1-2, pp.197-207, 2013.

H. Kaygusuz, E. Torlak, G. Ak?n-evingür, ?. Özen, R. Von-klitzing et al., Antimicrobial Cerium Ion-Chitosan Crosslinked Alginate Biopolymer Films: A Novel and Potential Wound Dressing, International Journal of Biological Macromolecules, pp.1161-1165, 2017.

L. Wang, E. Khor, A. Wee, and L. Y. Lim, Chitosan-Alginate PEC Membrane as a Wound Dressing: Assessment of Incisional Wound Healing, Journal of Biomedical Materials Research, vol.63, issue.5, pp.610-618, 2002.

Z. Hu, Q. Q. Ouyang, Y. Cheng, P. Z. Hong, M. N. Liao et al., Optimization of Preparation Process and Characterization of Carboxymethyl Chitosan/Sodium Alginate Hemostatic Sponge, IOP Conference Series: Materials Science and Engineering, vol.213, p.12045, 2017.

E. Öztürk, C. A?alar, K. Keçeci, and B. Denkbas?-emir, Preparation and Characterization of Ciprofloxacin-Loaded Alginate/Chitosan Sponge as a Wound Dressing Material, Journal of Applied Polymer Science, vol.101, issue.3, pp.1602-1609, 2006.

M. Dumont, R. Villet, M. Guirand, A. Montembault, T. Delair et al., Processing and Antibacterial Properties of Chitosan-Coated Alginate Fibers, Carbohydrate Polymers, vol.190, pp.31-42, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01917826

S. Reed and B. M. Wu, Biological and Mechanical Characterization of ChitosanAlginate Scaffolds for Growth Factor Delivery and Chondrogenesis, Journal of Biomedical Materials Research B, vol.2017, issue.2, pp.272-282

Z. Li and M. Zhang, Chitosan-Alginate as Scaffolding Material for Cartilage Tissue Engineering, Journal of Biomedical Materials Research A, vol.75, issue.2, pp.485-493, 2005.

N. Iwasaki, S. Yamane, T. Majima, Y. Kasahara, A. Minami et al., Feasibility of Polysaccharide Hybrid Materials for Scaffolds in Cartilage Tissue Engineering: Evaluation of Chondrocyte Adhesion to Polyion Complex Fibers Prepared from Alginate and Chitosan, Biomacromolecules, vol.5, issue.3, pp.828-833, 2004.

S. J. Seo, I. Y. Kim, Y. J. Choi, T. Akaike, and C. S. Cho, Enhanced Liver Functions of Hepatocytes Cocultured with NIH 3T3 in the Alginate/Galactosylated Chitosan Scaffold, Biomaterials, vol.27, issue.8, pp.1487-1495, 2006.

J. Yang, T. Chung, M. Nagaoka, M. Goto, C. Cho et al., Hepatocyte-Specific Porous Polymer-Scaffolds of Alginate/Galactosylated Chitosan Sponge for Liver-Tissue Engineering, Biotechnology Letters, vol.23, issue.17, pp.1385-1389, 2001.

F. Chen, M. Tian, D. Zhang, J. Wang, Q. Wang et al., Preparation and Characterization of Oxidized Alginate Covalently Cross-Linked Galactosylated Chitosan Scaffold for Liver Tissue Engineering, Materials Science and Engineering C, vol.2012, issue.2, pp.310-320

N. L. Francis, P. M. Hunger, A. E. Donius, B. W. Riblett, A. Zavaliangos et al., An Ice-Templated, Linearly Aligned Chitosan-Alginate Scaffold for Neural Tissue Engineering, Journal of Biomedical Materials Research A, vol.2013, issue.12, pp.3493-3503

L. A. Pfister, M. Papaloïzos, H. P. Merkle, and B. Gander, Hydrogel Nerve Conduits Produced from Alginate/Chitosan Complexes, Journal of Biomedical Materials Research A, vol.80, issue.4, pp.932-937, 2007.

Z. Li, H. R. Ramay, K. D. Hauch, D. Xiao, and M. Zhang, Chitosan-Alginate Hybrid Scaffolds for Bone Tissue Engineering, Biomaterials, vol.26, issue.18, pp.3919-3928, 2005.

S. J. Florczyk, M. Leung, Z. Li, J. I. Huang, R. A. Hopper et al., Evaluation of Three-Dimensional Porous Chitosan-Alginate Scaffolds in Rat Calvarial Defects for Bone Regeneration Applications, Journal of Biomedical Materials Research A, vol.2013, issue.10, pp.2974-2983

J. Venkatesan, I. Bhatnagar, and S. K. Kim, Chitosan-Alginate Biocomposite Containing Fucoidan for Bone Tissue Engineering, Marine Drugs, vol.12, issue.1, pp.300-316, 2014.

X. Liu, W. Peng, Y. Wang, M. Zhu, T. Sun et al., Synthesis of an RGD-Grafted Oxidized Sodium Alginate-N-Succinyl Chitosan Hydrogel and an In Vitro Study of Endothelial and Osteogenic Differentiation, Journal of Materials Chemistry B, vol.2013, issue.35, pp.4484-4492

D. J. Park, B. H. Choi, S. J. Zhu, J. Y. Huh, B. Y. Kim et al., Injectable Bone Using Chitosan-Alginate Gel/Mesenchymal Stem Cells/BMP-2 Composites, Journal of Cranio-Maxillofacial Surgery, vol.33, issue.1, pp.50-54, 2005.

T. Majima, T. Funakosi, N. Iwasaki, S. T. Yamane, K. Harada et al., Alginate and Chitosan Polyion Complex Hybrid Fibers for Scaffolds in Ligament and Tendon Tissue Engineering, Journal of Orthopaedic Science, vol.10, issue.3, pp.302-307, 2005.

K. Ziv, H. Nuhn, Y. Ben-haim, L. S. Sasportas, P. J. Kempen et al., A Tunable Silk-Alginate Hydrogel Scaffold for Stem Cell Culture and Transplantation, Biomaterials, vol.35, issue.12, pp.3736-3743, 2014.

K. Mladenovska, O. Cruaud, P. Richomme, E. Belamie, R. S. Raicki et al., 5-ASA Loaded Chitosan-CaAlginate Microparticles: Preparation and Physicochemical Characterization, International Journal of Pharmaceutics, vol.345, issue.1-2, pp.59-69, 2007.

J. A. Cury, C. Bragotto, and L. Valdrighi, The Demineralizing Efficiency of EDTA Solutions on Dentin. I. Influence of pH. Oral Surgery, Oral Medicine, Oral Pathology, vol.52, pp.446-448, 1981.

C. Godugu, A. R. Patel, U. Desai, T. Andey, A. Sams et al., AlgiMatrix? Based 3D Cell Culture System as an In-Vitro Tumor Model for Anticancer Studies, PLoS ONE, vol.2013, issue.1, p.53708

L. M. Coussens and Z. Werb, Inflammation and Cancer, Nature, vol.420, pp.860-867, 2002.

D. Richard, P. Chevalet, S. Fournel, N. Giraud, F. Gros et al., , 2012.

J. V. Sarma and P. A. Ward, The Complement System, Cell and Tissue Research, vol.343, issue.1, pp.227-235, 2011.

C. Golias, A. Charalabopoulos, D. Stagikas, K. Charalabopoulos, and A. Batistatou, The Kinin System-Bradykinin: Biological Effects and Clinical Implications. Multiple Role of the Kinin System-Bradykinin, Hippokratia, vol.11, issue.3, pp.124-128, 2007.

J. N. Sharma, A. Al-omran, and S. S. Parvathy, Role of Nitric Oxide in Inflammatory Diseases, Inflammopharmacology, vol.15, issue.6, pp.252-259, 2007.

R. Kumar, G. Clermont, Y. Vodovotz, and C. C. Chow, The Dynamics of Acute Inflammation, Journal of Theoretical Biology, vol.230, issue.2, pp.145-155, 2004.

J. G. Merrell, S. W. Mclaughlin, L. Tie, C. T. Laurencin, A. F. Chen et al., Curcumin-Loaded Poly(Epsilon-Caprolactone) Nanofibres: Diabetic Wound Dressing with Anti-Oxidant and Anti-Inflammatory Properties, Clinical and Experimental Pharmacology and Physiology, vol.36, issue.12, pp.1149-1156, 2009.

J. A. Méndez, M. Fernández, A. González-corchón, M. Salvado, F. Collía et al., Injectable Self-Curing Bioactive Acrylic-Glass Composites Charged with Specific AntiInflammatory/Analgesic Agent, Biomaterials, vol.25, issue.12, pp.2381-2392, 2004.

T. Ito, I. P. Fraser, Y. Yeo, C. B. Highley, E. Bellas et al., AntiInflammatory Function of an In Situ Cross-Linkable Conjugate Hydrogel of Hyaluronic Acid and Dexamethasone, Biomaterials, vol.28, issue.10, pp.1778-1786, 2007.

M. J. Webber, J. B. Matson, V. K. Tamboli, and S. I. Stupp, Controlled Release of Dexamethasone from Peptide Nanofiber Gels to Modulate Inflammatory Response, Biomaterials, vol.2012, issue.28, pp.6823-6832

C. Lee, L. Lo, C. Mou, and C. Yang, Synthesis and Characterization of Positive-Charge Functionalized Mesoporous Silica Nanoparticles for Oral Drug Delivery of an Anti-Inflammatory Drug, Advanced Functional Materials, vol.18, issue.20, pp.3283-3292, 2008.

K. K. Wong, S. O. Cheung, L. Huang, J. Niu, C. Tao et al., Further Evidence of the Anti-Inflammatory Effects of Silver Nanoparticles, ChemMedChem, vol.4, issue.7, pp.1129-1135, 2009.

L. David, B. Moldovan, A. Vulcu, L. Olenic, M. Perde-schrepler et al., Green Synthesis, Characterization and Anti-Inflammatory Activity of Silver Nanoparticles using European Black Elderberry Fruits Extract, Colloids and Surfaces B Biointerfaces, vol.122, pp.767-777, 2014.

B. Moldovan, L. David, A. Vulcu, L. Olenic, M. Perde-schrepler et al., Vitro and In Vivo Anti-Inflammatory Properties of Green Synthesized Silver Nanoparticles Using Viburnum Opulus L. Fruits Extract. Materials Science and Engineering C 2017, vol.79, pp.720-727

S. M. Hirst, A. S. Karakoti, R. D. Tyler, N. Sriranganathan, S. Seal et al., Anti-Inflammatory Properties of Cerium Oxide Nanoparticles, Small, vol.5, issue.24, pp.2848-2856, 2009.

M. X. Xu, Y. F. Zhu, H. F. Chang, and Y. Liang, Nanoceria Restrains PM2.5-Induced Metabolic Disorder and Hypothalamus Inflammation by Inhibition of Astrocytes Activation Related NF-?B Pathway in Nrf2 Deficient Mice, Free Radical Biology and Medicine, vol.99, pp.259-272, 2016.

H. Wang, H. Zhu, W. Fu, Y. Zhang, B. Xu et al., A High Strength Self-Healable Antibacterial and Anti-Inflammatory Supramolecular Polymer Hydrogel. Macromolecular Rapid Communications, vol.38, p.1600695, 2017.

I. L. Isa, A. Srivastava, D. Tiernan, P. Owens, P. Rooney et al., Hyaluronic Acid Based Hydrogels Attenuate Inflammatory Receptors and Neurotrophins in Interleukin-1? Induced Inflammation Model of Nucleus Pulposus Cells, Biomacromolecules, vol.2015, issue.6, pp.1714-1725

Y. Rosenfeld and Y. Shai, Lipopolysaccharide (Endotoxin)-Host Defense Antibacterial Peptides Interactions: Role in Bacterial Resistance and Prevention of Sepsis, Biochimica et Biophysica Acta-Biomembranes, vol.1758, issue.9, pp.1513-1522, 2006.

M. Kelm, Nitric Oxide Metabolism and Breakdown, Biochimica et Biophysica Acta, vol.1411, issue.2-3, pp.273-289, 1999.

N. S. Bryan and M. B. Grisham, Methods to Detect Nitric Oxide and its Metabolites in Biological Samples. Free Radical Biology and Medicine, vol.43, pp.645-657, 2007.

S. Galdiero, A. Falanga, M. Cantisani, R. Tarallo, M. E. Della-pepa et al., Microbe-Host Interactions: Structure and Role of Gram-Negative Bacterial Porins, Current Protein and Peptide Science, vol.2012, issue.8, pp.843-854

M. J. Eck and S. R. Sprang, The Structure of Tumor Necrosis Factor-Alpha at 2.6 A Resolution. Implications for Receptor Binding, The Journal of Biological Chemistry, vol.264, issue.29, pp.17595-17605, 1989.

H. Qin, C. A. Wilson, S. J. Lee, X. Zhao, and E. N. Benveniste, LPS Induces, p.40

, Gene Expression Through the Activation of NF-KappaB and STAT-1alpha in Macrophages and Microglia, Blood, vol.106, issue.9, pp.3114-3122, 2005.

T. Scholzen and J. Gerdes, The Ki-67 Protein: From the Known and the Unknown, Journal of Cellular Physiology, vol.182, issue.3, pp.311-322, 2000.

H. J. Yoon, M. E. Moon, H. S. Park, S. Y. Im, and Y. H. Kim, Chitosan Oligosaccharide (COS) Inhibits LPS-Induced Inflammatory Effects in RAW, vol.264

, Macrophage Cells. Biochemical and Biophysical Research Communications, vol.358, issue.3, pp.954-959, 2007.

Z. Jie, Z. Yuanyuan, W. Guoyou, X. Zhen, Z. Huanqin et al., Inhibitory Effects of Oligochitosan on TNF-?, IL-1? and Nitric Oxide Production in Lipopolysaccharide-Induced RAW264.7 Cells, Molecular Medicine Reports, vol.11, issue.1, pp.729-733, 2015.

G. Tejashri, B. Amrita, and J. Darshana, Cyclodextrin Based Nanosponges for Pharmaceutical Use: A Review, Acta Pharmaceutica, vol.63, issue.3, pp.335-358, 2013.

Y. Landry and J. Gies, Pharmacologie : Des Cibles Vers L'Indication Thérapeutique, 2003.

S. Rozou, A. Voulgari, and E. Antoniadou-vyza, The Effect of pH Dependent Molecular Conformation and Dimerization Phenomena of Piroxicam on the Drug:Cyclodextrin Complex Stoichiometry and its Chromatographic Behaviour. A New Specific HPLC Method for Piroxicam:Cyclodextrin Formulations, European Journal of Pharmaceutical Science, vol.21, issue.5, pp.661-669, 2004.

M. Jug and M. Be?irevi?-la?an, Multicomponent Complexes of Piroxicam with Cyclodextrins and Hydroxypropyl Methylcellulose. Drug Development and Industrial Pharmacy, vol.30, pp.1051-1060, 2004.

X. Deroubaix, A. Stockis, A. M. Allemon, E. Lebacq, D. Acerbi et al., Oral Bioavailability of CHF1194, an Inclusion Complex of Piroxicam and Beta-Cyclodextrin, in Healthy Subjects under Single Dose and Steady-State Conditions, European Journal of Clinical Pharmacology, vol.47, issue.6, pp.531-536, 1995.

B. G. Woodcock, D. Acerbi, P. G. Merz, S. Rietbrock, and N. Rietbrock, Supermolecular Inclusion of Piroxicam with Beta-Cyclodextrin: Pharmacokinetic Properties in Man, European Journal of Rheumatology and Inflammation, vol.12, issue.4, pp.12-28, 1993.

P. Osadebe, L. E. Onugwu, and A. Attama, Energetics of the Interaction Between Piroxicam and Beta-Cyclodextrin (Beta-CD) in Inclusion Complexes, Scientific Research and Essay, vol.3, issue.3, pp.86-93, 2008.

M. V. Nagabhushanam, Formulation Studies on Cyclodextrin Complexes of Piroxicam, Rasayan Journal of Chemistry, vol.2010, issue.2, pp.314-320

H. S. Chiong, Y. K. Yong, Z. Ahmad, M. R. Sulaiman, Z. A. Zakaria et al., Cytoprotective and Enhanced Anti-Inflammatory Activities of Liposomal Piroxicam Formulation in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages, International Journal of Nanomedicine, vol.8, pp.1245-1255, 2013.

S. Kirkiacharian, Guide de Chimie Médicinale et Médicaments, 2010.

F. Djedaïni and B. Perly, Nuclear Magnetic Resonance Investigation of the Stoichiometries in Beta-Cyclodextrin:Steroid Inclusion Complexes, Journal of Pharmaceutical Science, vol.80, issue.12, pp.1157-1161, 1991.

K. Uekama, T. Fujinaga, F. Hirayama, M. Otagiri, and M. Yamasaki, Inclusion Complexations of Steroid Hormones with Cyclodextrins in Water and in Solid Phase, International Journal of Pharmaceutics, vol.10, issue.1, pp.1-15, 1982.

K. Uekama, M. Otagiri, Y. Uemura, T. Fujinaga, K. Arimori et al., Improvement of Oral Bioavailability of Prednisolone by BetaCyclodextrin Complexation in Humans, Journal of Pharmacobio-dynamics, vol.1983, issue.2, pp.124-127

R. Ashok, P. P. Prakash, and R. Selvan, Development and Validation of Analytical Method for Estimation of Prednisolone in Bulk and Tablets using UV-Visible Spectroscopy, International Journal of Pharmacy and Pharmaceutical Sciences, vol.2011, issue.4, pp.184-186

M. Ali, S. K. Kumar, and S. Parthiban, Design and Evaluation of pH Triggered Osmotically Controlled System for Ileo-Colonic Drug Delivery, Asian Journal of Pharmaceutical Research, vol.2013, issue.2, pp.66-74

G. Gasparini, E. K. Bang, G. Molinard, D. V. Tulumello, S. Ward et al., Cellular Uptake of Substrate-Initiated Cell-Penetrating Poly(disulfide)s, Journal of the American Chemical Society, vol.136, issue.16, pp.6069-6074, 2014.

D. P. Nair, M. Podgórski, S. Chatani, T. Gong, W. Xi et al., The Thiol-Michael Addition Click Reaction: A Powerful and Widely Used Tool in Materials Chemistry, Chemistry of Materials, vol.26, issue.1, pp.724-744, 2014.