. Department-of-health and S. Human, , 2013.

, Agence nationale de recherches sur le sida et les hépatites virales, Ministère des affaires sociales et de la Santé, 2013.

G. Kumari and R. K. Singh, Anti-HIV Drug Development: Structural Features and Limitations of Present Day Drugs and Future Challenges in the Successful HIV/AIDS Treatment, Curr. Pharm. Design, vol.19, pp.1767-1783, 2013.

H. B. Krentz, I. Cosman, K. Lee, J. M. Ming, and M. J. Gill, Pill burden in HIV infection: 20 years of experience, Antivir. Ther, vol.17, pp.833-840, 2012.

A. Mocroft, M. Youle, A. Moore, C. A. Sabin, S. Madge et al., Reasons for modification and discontinuation of antiretrovirals: results from a single treatment centre, Aids, vol.15, pp.185-194, 2001.

S. G. Deeks, B. Autran, B. Berkhout, M. Benkirane, S. Cairns et al., for The International AIDS Society Scientific Working Group on HIV Cure, Towards an HIV cure: a global scientific strategy, vol.12, pp.607-614, 2012.

H. K. Han, R. L. De-vrueh, J. K. Rhie, K. M. Covitz, P. L. Smith et al., 5 '-amino acid esters of antiviral nucleosides, acyclovir, and AZT are absorbed by the intestinal PEPT1 peptide transporter, Pharm. Res, vol.15, pp.1154-1159, 1998.

K. F. Croom and S. J. Keam, Tipranavir -A ritonavir-boosted protease inhibitor, Drugs, vol.65, pp.1669-1677, 2005.

D. A. Chiappetta, C. Hocht, C. Taira, and A. Sosnik, Efavirenz-loaded polymeric micelles for pediatric anti-HIV pharmacotherapy with significantly higher oral bioavailaibility, Nanomedicine, vol.5, pp.11-23, 2010.

K. Parang, L. I. Wiebe, and E. E. Knaus, Novel approaches for designing 5 '-O-ester prodrugs of 3'-azido-2 ',3 '-dideoxythymidine (AZT), Curr. Med. Chem, vol.7, pp.995-1039, 2000.

A. P. Sharma, A. P. Ollapally, and H. J. Lee, Synthesis and Anti-Hiv Activity of Prodrugs of Azidothymidine, Antivir. Chem. Chemother, vol.4, pp.93-96, 1993.

N. Tsuzuki, T. Hama, M. Kawada, A. Hasui, R. Konishi et al., Adamantane as a Brain-Directed Drug Carrier for Poorly Absorbed Drug .2. Azt Derivatives Conjugated with the 1-Adamantane Moiety, J. Pharm. Sci, vol.83, pp.481-484, 1994.

T. Kawaguchi, K. Ishikawa, T. Seki, and K. Juni, Ester Prodrugs of Zidovudine, J. Pharm. Sci, vol.79, pp.531-533, 1990.

J. Balzarini, P. Herdewijn, and E. Declercq, Differential Patterns of Intracellular Metabolism of 2',3'-Didehydro-2',3'-Dideoxythymidine and 3'-Azido-2',3'-Dideoxythymidine, 2 Potent Anti-Human Immunodeficiency Virus Compounds, J. Biol. Chem, vol.264, pp.6127-6133, 1989.

J. Bourdais, R. Biondi, S. Sarfati, C. Guerreiro, I. Lascu et al., Cellular phosphorylation of Anti-HIV nucleosides -Role of nucleoside diphosphate kinase, J. Biol. Chem, vol.271, pp.7887-7890, 1996.

A. Tsotinis, T. Calogeropoulou, M. Koufaki, C. Souli, J. Balzarini et al., Synthesis and antiretroviral evaluation of new alkoxy and aryloxy phosphate derivatives of 3'-azido-3'-deoxythymidine, J. Med. Chem, vol.39, pp.3418-3422, 1996.

A. Pompon, I. Lefebvre, J. L. Imbach, S. Kahn, and D. Farquhar, Decomposition Pathways of the Mono-(Pivaloyl-Oxymethyl) and Bis(Pivaloyl-Oxymethyl) Esters of Azidothymidine 5'-Monophosphate in Cell Extract and in Tissue-Culture Medium -an Application of the Online Isrp-Cleaning Hplc Technique, Antivir. Chem. Chemother, vol.5, pp.91-98, 1994.

C. Mcguigan, K. G. Devine, T. J. Oconnor, and D. Kinchington, Synthesis and Anti-Hiv Activity of Some Haloalkyl Phosphoramidate Derivatives of 3'-Azido-3'-Deoxythymidine (Azt) -Potent Activity of the Trichloroethyl Methoxyalaninyl Compound, Antiviral Res, vol.15, pp.255-263, 1991.

M. R. Blum, S. H. Liao, S. S. Good, and P. Demiranda, Pharmacokinetics and Bioavailability of Zidovudine in Humans, Am. J. Med, vol.85, pp.189-194, 1988.

A. P. Lea and D. Faulds, Stavudine -A review of its pharmacodynamic and pharmacokinetic properties and clinical potential in HIV infection, Drugs, vol.51, pp.846-864, 1996.

R. W. Klecker, J. M. Collins, R. C. Yarchoan, R. Thomas, N. Mcatee et al., Pharmacokinetics of 2',3'-Dideoxycytidine in Patients with Aids and Related Disorders, J. Clin. Pharmacol, vol.28, pp.837-842, 1988.

G. Y. Kwei, L. B. Novak, L. A. Hettrick, E. R. Reiss, D. Ostovic et al., Regiospecific Intestinal-Absorption of the Hiv Protease Inhibitor L-735,524 in Beagle Dogs, Pharm. Res, vol.12, pp.884-888, 1995.

S. D. Portsmouth, J. Osorio, K. Mccormick, B. G. Gazzard, and G. J. Moyle, Better maintained adherence on switching from twice-daily to once-daily therapy for HIV: a 24-week randomized trial of treatment simplification using stavudine prolonged-release capsules, HIV Med, vol.6, pp.185-190, 2005.

M. G. Mateo, M. D. Gutierrez, F. Vidal, and P. Domingo, Stavudine extended release (oncedaily, Bristol-Myers Squibb) for the treatment of HIV/AIDS, Expert Opin. Pharmacother, vol.14, pp.1055-1064, 2013.

A. Kuksal, A. K. Tiwary, N. K. Jain, and S. Jain, Formulation and in vitro, in vivo evaluation of extended-release matrix tablet of zidovudine: Influence of combination of hydrophilic and hydrophobic matrix formers, Aaps Pharmscitech, p.7, 2006.

J. V. Santos, M. E. Pina, M. P. Marques, and L. De-carvalho, New sustained release of Zidovudine Matrix tablets -cytotoxicity toward Caco-2 cells, Drug Dev. Ind. Pharm, vol.39, pp.1154-1166, 2013.

J. Gathe, J. Andrade-villanueva, S. Santiago, A. Horban, M. Nelson et al., Efficacy and safety of nevirapine extended-release once daily versus nevirapine immediaterelease twice-daily in treatment-naive HIV-1-infected patients, Antivir. Ther, vol.16, pp.759-769, 2011.

F. Bonina, C. Puglia, M. G. Rimoli, L. Avallone, E. Abignente et al., Synthesis and in vitro chemical and enzymatic stability of glycosyl 3 '-azido-3 '-deoxythymidine derivatives as potential anti-HIV agents, Eur. J. Pharm. Sci, vol.16, pp.167-174, 2002.

S. Wannachaiyasit, P. Chanvorachote, and U. Nimmannit, A Novel Anti-HIV Dextrin-Zidovudine Conjugate Improving the Pharmacokinetics of Zidovudine in Rats, Aaps Pharmscitech, vol.9, pp.840-850, 2008.

S. Kamboj, V. Saini, and S. Bala, Formulation and Characterization of Drug Loaded Nonionic Surfactant Vesicles (Niosomes) for Oral Bioavailability Enhancement, Sci. World J, 2014.

L. N. Ramana, S. Sharma, S. Sethuraman, U. Ranga, and U. M. Krishnan, Investigation on the stability of saquinavir loaded liposomes: Implication on stealth, release characteristics and cytotoxicity, Int. J. Pharm, vol.431, pp.120-129, 2012.

L. Kinman, S. J. Brodie, C. C. Tsai, T. Bui, K. Larsen et al., Lipid-drug association enhanced HIV-1 protease inhibitor indinavir localization in lymphoid tissues and viral load reduction: A proof of concept study in HIV-2(287)-infected macaques, Jaids, vol.34, pp.387-397, 2003.

S. J. Snedecor, S. M. Sullivan, and R. J. Ho, Feasibility of weekly HIV drug delivery to enhance drug localization in lymphoid tissues based on pharmacokinetic models of lipid-associated indinavir, Pharm. Res, vol.23, pp.1750-1755, 2006.

S. Swindells, C. Flexner, C. V. Fletcher, and J. M. Jacobson, The Critical Need for Alternative Antiretroviral Formulations, and Obstacles to Their Development, J. Infect. Dis, vol.204, pp.669-674, 2011.

L. Baert, L. Schueller, Y. Tardy, D. Macbride, G. Van't-klooster et al., Development of an implantable infusion pump for sustained anti-HIV drug administration, Int. J. Pharm, vol.355, pp.38-44, 2008.

D. N. Kapoor, O. P. Katare, and S. Dhawan, In situ forming implant for controlled delivery of an anti-HIV fusion inhibitor, Int. J. Pharm, vol.426, pp.132-143, 2012.

J. N. Blankson, D. Persaud, and R. F. Siliciano, The challenge of viral reservoirs in HIV-1 infection, Annu. Rev. Med, vol.53, pp.557-593, 2002.

R. J. Pomerantz, Reservoirs, sanctuaries, and residual disease: The hiding spots of HIV-1, HIV Clin. Trials, vol.4, pp.137-143, 2003.

S. M. Crowe, Macrophages and residual HIV infection, Curr. Opin. HIV AIDS, vol.1, pp.129-133, 2006.

G. Molema, R. W. Jansen, R. Pauwels, E. Declercq, and D. K. Meijer, Targeting of Antiviral Drugs to Lymphocytes-T4 -Anti-Hiv Activity of Neoglycoprotein Aztmp Conjugates Invitro, vol.40, pp.2603-2610, 1990.

M. Garg and N. K. Jain, Reduced hematopoietic toxicity, enhanced cellular uptake and altered pharmacokinetics of azidothymidine loaded galactosylated liposomes, J. Drug Target, vol.14, pp.1-11, 2006.

J. H. Hu, H. Liu, and L. L. Wang, Enhanced delivery of AZT to macrophages via acetylated LDL, J. Control. Release, vol.69, pp.327-335, 2000.

P. Calvo, C. Remuñán-lópez, J. L. Vila-jato, and M. J. Alonso, Novel hydrophilic chitosanpolyethylene oxide nanoparticles as protein carriers, J Appl Polym Sci, vol.63, pp.125-132, 1997.

M. Garcia-fuentes and M. J. Alonso, Chitosan-based drug nanocarriers: where do we stand?, J Control Release, vol.161, pp.496-504, 2012.

S. Rodrigues, M. Dionísio, C. R. López, and A. Grenha, Biocompatibility of Chitosan Carriers with Application in Drug Delivery, J Funct Biomater, vol.3, pp.615-641, 2012.

H. Zhang, M. Oh, C. Allen, and E. Kumacheva, Monodisperse chitosan nanoparticles for mucosal drug delivery, Biomacromolecules, vol.5, pp.2461-2469, 2004.

J. Peng, X. Xing, K. Wang, W. Tan, X. He et al., Influence of anions on the formation and properties of chitosan-DNA nanoparticles, J Nanosci Nanotechnol, vol.5, pp.713-720, 2005.

H. Katas and H. O. Alpar, Development and characterisation of chitosan nanoparticles for siRNA delivery, J Control Release, vol.115, pp.216-241, 2006.

G. Giacalone, A. Bochot, E. Fattal, and H. Hillaireau, Drug-Induced Nanocarrier Assembly as a Strategy for the Cellular Delivery of Nucleotides and Nucleotide Analogues, Biomacromolecules, vol.14, pp.737-742, 2013.

T. López-león, E. L. Carvalho, B. Seijo, J. L. Ortega-vinuesa, and D. Bastos-gonzález, Physicochemical characterization of chitosan nanoparticles: electrokinetic and stability behavior, J Colloid Interface Sci, vol.283, pp.344-51, 2005.

O. Leppänen, T. Björnheden, M. Evaldsson, J. Boren, O. Wiklund et al., ATP depletion in macrophages in the core of advanced rabbit atherosclerotic plaques in vivo, Atherosclerosis, vol.188, pp.323-330, 2006.

W. Martinet, D. M. Schrijvers, and G. R. De-meyer, Necrotic cell death in atherosclerosis, Basic Research in Cardiology, vol.106, pp.749-760, 2011.

G. Y. Lee, J. H. Kim, G. T. Oh, B. H. Lee, I. C. Kwon et al., Molecular targeting of atherosclerotic plaques by a stabilin-2-specific peptide ligand, J. Control. Release, vol.155, pp.211-217, 2011.

O. Borges, G. Borchard, J. C. Verhoef, A. De-sousa, and H. E. Junginger, Preparation of coated nanoparticles for a new mucosal vaccine delivery system, Int J Pharm, vol.299, pp.155-166, 2005.

A. Nasti, N. M. Zaki, P. De-leonardis, S. Ungphaiboon, P. Sansongsak et al., Chitosan/TPP and chitosan/TPP-hyaluronic acid nanoparticles: systematic optimisation of the preparative process and preliminary biological evaluation, Pharm Res, vol.26, pp.1918-1948, 2009.

F. Schmitt, L. Lagopoulos, P. Kauper, N. Rossi, N. Busso et al., Chitosan-based nanogels for selective delivery of photosensitizers to macrophages and improved retention in and therapy of articular joints, J Control Release, vol.144, pp.242-50, 2010.

F. L. Mi, H. W. Sung, S. S. Shyu, C. C. Su, and C. K. Peng, Synthesis and characterization of biodegradable TPP/genipin co-crosslinked chitosan gel beads, Polymer, vol.44, pp.6521-6530, 2003.

H. W. Sung, I. L. Liang, C. N. Chen, R. N. Huang, and H. F. Liang, Stability of a biological tissue fixed with a naturally occurring crosslinking agent (genipin), J. Biomed. Mater. Res, vol.55, pp.538-546, 2001.

S. C. Bhatia and N. Ravi, A magnetic study of an Fe-chitosan complex and its relevance to other biomolecules, Biomacromolecules, vol.1, pp.413-420, 2000.

F. Reynaud, N. Tsapis, M. Deyme, T. G. Vasconcelos, C. Gueutin et al., Spray-dried chitosan-metal microparticles for ciprofloxacin adsorption: Kinetic and equilibrium studies, Soft Matter, vol.7, pp.7304-7312, 2011.

L. Lijklema, Interaction of orthophosphate with iron(III) and aluminum hydroxides, Environ Sci Technol, vol.14, pp.537-541, 1980.

K. Spengler, H. Follmann, K. S. Boos, D. Seidel, and F. Maywald, Characterization and extracorporeal application of a new phosphate-binding agent, Eur J Clin Chem Clin Biochem, vol.32, pp.733-742, 1994.

W. B. Fortune and M. G. Mellon, Determination of Iron with o-Phenanthroline: A Spectrophotometric Study, Ind Eng Chem Anal Ed, vol.10, pp.60-64, 1938.

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays, J Immunol Methods, vol.65, pp.55-63, 1983.

D. Fu, P. G. Keech, X. L. Sun, and J. C. Wren, Iron oxyhydroxide nanoparticles formed by forced hydrolysis: dependence of phase composition on solution concentration, Phys Chem Chem Phys, vol.13, pp.18523-18529, 2011.

S. V. Vinogradov, A. D. Zeman, E. V. Batrakova, and A. V. Kabanov, Polyplex Nanogel formulations for drug delivery of cytotoxic nucleoside analogs, J Control Release, vol.107, pp.143-157, 2005.

M. M. Elsayed and G. Cevc, Turbidity spectroscopy for characterization of submicroscopic drug carriers, such as nanoparticles and lipid vesicles: size determination, Pharm Res, vol.28, pp.2204-2226, 2011.

K. Buyens, B. Lucas, K. Raemdonck, K. Braeckmans, J. Vercammen et al., A fast and sensitive method for measuring the integrity of siRNA-carrier complexes in full human serum, J Control Release, vol.126, pp.67-76, 2008.

E. Geiringer, Intimal vascularisation and atherosclerosis, The Journal of Pathology and Bacteriology, vol.63, pp.201-211, 1951.

S. Parathath, S. L. Mick, J. E. Feig, V. Joaquin, L. Grauer et al., Hypoxia Is Present in Murine Atherosclerotic Plaques and Has Multiple Adverse Effects on Macrophage Lipid Metabolism, vol.109, pp.1141-1152, 2011.

T. J. Cory, T. W. Schacker, M. Stevenson, and C. V. Fletcher, Overcoming pharmacologic sanctuaries, Curr. Opin. HIV AIDS, issue.8, pp.190-195, 2013.

C. M. Coleman and L. Wu, HIV interactions with monocytes and dendritic cells: viral latency and reservoirs, Retrovirology, p.6, 2009.

A. Zalar, M. I. Figueroa, B. Ruibal-ares, P. Bare, P. Cahn et al., Macrophage HIV-1 infection in duodenal tissue of patients on long term HAART, Antiviral Res, vol.87, pp.269-271, 2010.

D. Mascio, M. Srinivasula, S. Bhattacharjee, A. Cheng, L. Martiniova et al., Antiretroviral Tissue Kinetics: In Vivo Imaging Using Positron Emission Tomography, Antimicrob. Agents Chemother, vol.53, pp.4086-4095, 2009.

T. W. North, J. Higgins, J. D. Deere, T. L. Hayes, A. Villalobos et al., Viral sanctuaries during highly active antiretroviral therapy in a nonhuman primate model for AIDS, J Virol, vol.84, pp.2913-2935, 2010.

J. Balzarini, P. Herdewijn, and E. Declercq, Differential Patterns of Intracellular Metabolism of 2',3'-Didehydro-2',3'-Dideoxythymidine and 3'-Azido-2',3'-Dideoxythymidine, 2 Potent Anti-Human Immunodeficiency Virus Compounds, J. Biol. Chem, vol.264, pp.6127-6133, 1989.

J. Bourdais, R. Biondi, S. Sarfati, C. Guerreiro, I. Lascu et al., Cellular phosphorylation of Anti-HIV nucleosides -Role of nucleoside diphosphate kinase, J. Biol. Chem, vol.271, pp.7887-7890, 1996.

R. Lobenberg, L. Araujo, and J. Kreuter, Body distribution of azidothymidine bound to nanoparticles after oral administration, Eur. J. Pharm. Biopharm, vol.44, pp.127-132, 1997.

R. Lobenberg, L. Araujo, H. Von-briesen, E. Rodgers, and J. Kreuter, Body distribution of azidothymidine bound to hexyl-cyanoacrylate nanoparticles after i.v. injection to rats, J. Control. Release, vol.50, pp.21-30, 1998.

M. Garg and N. K. Jain, Reduced hematopoietic toxicity, enhanced cellular uptake and altered pharmacokinetics of azidothymidine loaded galactosylated liposomes, J. Drug Target, vol.14, pp.1-11, 2006.

A. Desormeaux, P. Harvie, S. Perron, B. Makabipanzu, D. Beauchamp et al., Antiviral Efficacy, Intracellular Uptake and Pharmacokinetics of Free and Liposome-Encapsulated 2',3'-Dideoxyinosine, Aids, vol.8, pp.1545-1553, 1994.

P. Harvie, A. Desormeaux, N. Gagne, M. Tremblay, L. Poulin et al., Lymphoid-Tissues Targeting of Liposome-Encapsulated 2',3'-Dideoxyinosine, Aids, vol.9, pp.701-707, 1995.

H. Hillaireau, N. Dereuddre-bosquet, R. Skanji, F. Bekkara-aounallah, J. Caron et al., Anti-HIV efficacy and biodistribution of nucleoside reverse transcriptase inhibitors delivered as squalenoylated prodrug nanoassemblies, Biomaterials, vol.34, pp.4831-4838, 2013.

S. V. Vinogradov, E. Kohli, and A. D. Zeman, Cross-linked polymeric nanogel formulations of 5 '-triphosphates of nucleoside analogues: Role of the cellular membrane in drug release, Mol. Pharm, vol.2, pp.449-461, 2005.

H. Hillaireau, T. Le-doan, M. Appel, and P. Couvreur, Hybrid polymer nanocapsules enhance in vitro delivery of azidothymidine-triphosphate to macrophages, J. Control. Release, vol.116, pp.346-352, 2006.

V. Agostoni, T. Chalati, P. Horcajada, H. Willaime, R. Anand et al., Towards an Improved anti-HIV Activity of NRTI via Metal-Organic Frameworks Nanoparticles, Adv. Health. Mater, issue.2, pp.1630-1637, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00935744

M. Magnani, L. Rossi, G. Brandi, G. F. Schiavano, M. Montroni et al., Targeting Antiretroviral Nucleoside Analogs in Phosphorylated Form to Macrophages -Invitro and Invivo Studies, Proc. Natl. Acad. Sci. U. S. A, vol.89, pp.6477-6481, 1992.

C. Oussoren, M. Magnani, A. Fraternale, A. Casabianca, L. Chiarantini et al., Liposomes as carriers of the antiretroviral agent dideoxycytidine-5'-triphosphate, Int J Pharm, vol.180, pp.261-270, 1999.

Y. Yang, J. L. Chen, H. Li, Y. Y. Wang, Z. Xie et al., Porcine interleukin-2 gene encapsulated in chitosan nanoparticles enhances immune response of mice to piglet paratyphoid vaccine, Comp. Immunol. Microbiol. Infect. Dis, vol.30, pp.19-32, 2007.

J. Benesch and P. Tengvall, Blood protein adsorption onto chitosan, Biomaterials, vol.23, pp.2561-2568, 2002.

S. Gummuluru, V. N. Kewalramani, and M. Emerman, Dendritic cell-mediated viral transfer to T cells is required for human immunodeficiency virus type 1 persistence in the face of rapid cell turnover, J. Virol, vol.76, pp.10692-10701, 2002.

G. Giacalone, H. Hillaireau, P. Capiau, H. Chacun, F. Reynaud et al., Stabilization and cellular delivery of chitosan-polyphosphate nanoparticles by incorporation of iron, J Control Release, vol.194, pp.211-219, 2014.

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays, J Immunol Methods, vol.65, pp.55-63, 1983.

M. A. Jenabian, H. Saidi, C. Charpentier, Y. Van-herrewege, J. C. Son et al., In vitro synergistic activity against CCR5-tropic HIV-1 with combinations of potential candidate microbicide molecules HHA, KRV2110 and enfuvirtide (T20), J. Antimicrob. Chemother, vol.64, pp.1192-1195, 2009.

H. Lecoeur, M. T. Melki, H. Saidi, and M. L. Gougeon, Programmed Cell Death, General Principles for Studying Cell Death, Pt A, vol.442, pp.51-82, 2008.

K. Konopka and N. Duzgunes, Expression of CD4 controls the susceptibility of THP-1 cells to infection by R5 and X4 HIV type 1 isolates, Aids Res. Hum. Retrovir, vol.18, pp.123-131, 2002.

L. Margolis and R. Shattock, Selective transmission of CCR5-utilizing HIV-1: the 'gatekeeper' problem resolved?, Nat. Rev. Microbiol, vol.4, pp.312-317, 2006.

H. Koppensteiner, R. Brack-werner, and M. Schindler, Macrophages and their relevance in Human Immunodeficiency Virus Type I infection, Retrovirology, p.9, 2012.

L. Dai, K. B. Lidie, Q. Chen, J. W. Adelsberger, X. Zheng et al., IL-27 inhibits HIV-1 infection in human macrophages by down-regulating host factor SPTBN1 during monocyte to macrophage differentiation, J. Exp. Med, vol.210, pp.517-534, 2013.

T. Kamala, Hock immunization: A humane alternative to mouse footpad injections, J. Immunol. Methods, vol.328, pp.204-214, 2007.

M. Wacker, Nanocarriers for intravenous injection -The long hard road to the market, Int. J. Pharm, vol.457, pp.50-62, 2013.

R. L. Dunn, J. P. , D. R. Cowsar, and D. P. Vanderbilt, , 1990.

M. Parent, C. Nouvel, M. Koerber, A. Sapin, P. Maincent et al., PLGA in situ implants formed by phase inversion: Critical physicochemical parameters to modulate drug release, J. Control. Release, vol.172, pp.292-304, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01273194

P. D. Graham, K. J. Brodbeck, and A. J. Mchugh, Phase inversion dynamics of PLGA solutions related to drug delivery, J. Control. Release, vol.58, pp.233-245, 1999.

J. M. Anderson and M. S. Shive, Biodegradation and biocompatibility of PLA and PLGA microspheres, Adv. Drug Deliv. Rev, vol.28, pp.5-24, 1997.

K. P. Lee, N. C. Chromey, R. Culik, J. R. Barnes, and P. W. Schneider, Toxicity of N-Methyl-2

. Pyrrolidone, Teratogenic, Subchronic, and 2-Year Inhalation Studies, Fundam. Appl. Toxicol, vol.9, pp.222-235, 1987.

Y. Tang and J. Singh, Controlled delivery of aspirin: Effect of aspirin on polymer degradation and in vitro release from PLGA based phase sensitive systems, Int. J. Pharm, vol.357, pp.119-125, 2008.

W. J. Lambert and K. D. Peck, Development of an in-Situ Forming Biodegradable Poly-Lactide-Co-Glycolide System for the Controlled-Release of Proteins, J. Control. Release, vol.33, pp.189-195, 1995.

K. J. Brodbeck, S. Pushpala, and A. J. Mchugh, Sustained release of human growth hormone from PLGA solution depots, Pharm. Res, vol.16, pp.1825-1829, 1999.

O. Sartor, Eligard: Leuprolide acetate in a novel sustained-release delivery system, Urology, vol.61, pp.25-31, 2003.

P. E. Le-renard, F. Buchegger, A. Petri-fink, F. Bosman, D. Rufenacht et al., Local moderate magnetically induced hyperthermia using an implant formed in situ in a mouse tumor model, Int. J. Hyperthermia, vol.25, pp.229-239, 2009.

V. Burckbuchler, G. Mekhloufi, A. P. Giteau, J. L. Grossiord, S. Huille et al., Rheological and syringeability properties of highly concentrated human polyclonal immunoglobulin solutions, Eur. J. Pharm. Biopharm, vol.76, pp.351-356, 2010.

A. J. Mchugh, The role of polymer membrane formation in sustained release drug delivery systems, J. Control. Release, vol.109, pp.211-221, 2005.

M. Adler, Challenges in the Development of Pre-filled Syringes for Biologics from a Formulation Scientist's Point of View, Am. Pharm. Rev, vol.15, 2012.

K. J. Brodbeck, J. R. Desnoyer, and A. J. Mchugh, Phase inversion dynamics of PLGA solutions related to drug delivery -Part II. The role of solution thermodynamics and bath-side mass transfer, J. Control. Release, vol.62, pp.333-344, 1999.

M. A. Royals, S. M. Fujita, G. L. Yewey, J. Rodriguez, P. C. Schultheiss et al., Biocompatibility of a biodegradable in situ forming implant system in rhesus monkeys, J. Biomed. Mater. Res, vol.45, pp.231-239, 1999.

S. Kempe and K. Mäder, In situ forming implants -an attractive formulation principle for parenteral depot formulations, J. Control. Release, vol.161, pp.668-679, 2012.

P. Calvo, C. Remuñán-lópez, J. L. Vila-jato, and M. J. Alonso, Novel hydrophilic chitosanpolyethylene oxide nanoparticles as protein carriers, J Appl Polym Sci, vol.63, pp.125-132, 1997.

H. Zhang, M. Oh, C. Allen, and E. Kumacheva, Monodisperse chitosan nanoparticles for mucosal drug delivery, Biomacromolecules, vol.5, pp.2461-2469, 2004.

J. Peng, X. Xing, K. Wang, W. Tan, X. He et al., Influence of anions on the formation and properties of chitosan-DNA nanoparticles, J Nanosci Nanotechnol, vol.5, pp.713-720, 2005.

H. Katas and H. O. Alpar, Development and characterisation of chitosan nanoparticles for siRNA delivery, J Control Release, vol.115, pp.216-241, 2006.

S. V. Vinogradov, E. Kohli, and A. D. Zeman, Cross-linked polymeric nanogel formulations of 5 '-triphosphates of nucleoside analogues: Role of the cellular membrane in drug release, Mol. Pharm, vol.2, pp.449-461, 2005.

E. Kohli, H. Han, A. D. Zeman, and S. V. Vinogradov, Formulations of biodegradable Nanogel carriers with 5'-triphosphates of nucleoside analogs that display a reduced cytotoxicity and enhanced drug activity, J Control Release, vol.121, pp.19-27, 2007.

H. Koppensteiner, R. Brack-werner, and M. Schindler, Macrophages and their relevance in Human Immunodeficiency Virus Type I infection, Retrovirology, p.9, 2012.

T. López-león, E. L. Carvalho, B. Seijo, J. L. Ortega-vinuesa, and D. Bastos-gonzález, Physicochemical characterization of chitosan nanoparticles: electrokinetic and stability behavior, J Colloid Interface Sci, vol.283, pp.344-51, 2005.

S. C. Bhatia and N. Ravi, A magnetic study of an Fe-chitosan complex and its relevance to other biomolecules, Biomacromolecules, vol.1, pp.413-420, 2000.

F. Reynaud, N. Tsapis, M. Deyme, T. G. Vasconcelos, C. Gueutin et al., Spray-dried chitosan-metal microparticles for ciprofloxacin adsorption: Kinetic and equilibrium studies, Soft Matter, vol.7, pp.7304-7312, 2011.

L. Lijklema, Interaction of orthophosphate with iron(III) and aluminum hydroxides, Environ Sci Technol, vol.14, pp.537-541, 1980.

K. Spengler, H. Follmann, K. S. Boos, D. Seidel, and F. Maywald, Characterization and extracorporeal application of a new phosphate-binding agent, Eur J Clin Chem Clin Biochem, vol.32, pp.733-742, 1994.

K. Buyens, B. Lucas, K. Raemdonck, K. Braeckmans, J. Vercammen et al., A fast and sensitive method for measuring the integrity of siRNA-carrier complexes in full human serum, J Control Release, vol.126, pp.67-76, 2008.

D. A. Rao, M. L. Forrest, A. W. Alani, G. S. Kwon, and J. R. Robinson, Biodegradable PLGA Based Nanoparticles for Sustained Regional Lymphatic Drug Delivery, J. Pharm. Sci, vol.99, 2009.

T. Kamala, Hock immunization: A humane alternative to mouse footpad injections, J. Immunol. Methods, vol.328, pp.204-214, 2007.

C. B. Packhaeuser, J. Schnieders, C. G. Oster, and T. Kissel, In situ forming parenteral drug delivery systems: an overview, Eur. J. Pharm. Biopharm, vol.58, pp.445-455, 2004.

. Department-of-health and S. Human, , 2013.

D. M. Margolis, Histone deacetylase inhibitors and HIV latency, Curr. Opin. HIV AIDS, vol.6, pp.25-29, 2011.

T. Courant, V. G. Roullin, C. Cadiou, M. Callewaert, M. C. Andry et al., Hydrogels Incorporating GdDOTA: Towards Highly Efficient Dual T1/T2 MRI Contrast Agents, vol.51, pp.9119-9122, 2012.