E. Allard, F. Hindre, C. Passirani, L. Lemaire, N. Lepareur et al., 188Re-loaded lipid nanocapsules as a promising radiopharmaceutical carrier for internal radiotherapy of malignant gliomas, European Journal of Nuclear Medicine and Molecular Imaging, vol.112, issue.10, pp.1838-1846, 2008.
DOI : 10.1007/s00259-008-0735-z

URL : https://hal.archives-ouvertes.fr/inserm-00343438

M. Baker, Whole-animal imaging: The whole picture, Nature, vol.15, issue.7283, pp.977-980, 2010.
DOI : 10.1038/463977a

S. Ballot, N. Noiret, F. Hindré, B. Denizot, E. Garin et al., 99mTc/188Re-labelled lipid nanocapsules as promising radiotracers for imaging and therapy: formulation and biodistribution, European Journal of Nuclear Medicine and Molecular Imaging, vol.242, issue.5, pp.602-607, 2006.
DOI : 10.1007/s00259-005-0007-0

A. Béduneau, F. Hindré, A. Clavreul, J. Leroux, P. Saulnier et al., Brain targeting using novel lipid nanovectors, Journal of Controlled Release, vol.126, issue.1, pp.44-49, 2008.
DOI : 10.1016/j.jconrel.2007.11.001

A. Béduneau, P. Saulnier, F. Hindré, A. Clavreul, J. Leroux et al., Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab??? fragments, Biomaterials, vol.28, issue.33, pp.4978-4990, 2007.
DOI : 10.1016/j.biomaterials.2007.05.014

E. Bourseau-guilmain, J. Béjaud, A. Griveau, N. Lautram, F. Hindré et al., Development and characterization of immuno-nanocarriers targeting the cancer stem cell marker AC133, International Journal of Pharmaceutics, vol.423, issue.1, pp.93-101, 2012.
DOI : 10.1016/j.ijpharm.2011.06.001

H. Cabral, Y. Matsumoto, K. Mizuno, Q. Chen, M. Murakami et al., Accumulation of sub-100??nm polymeric micelles in poorly permeable tumours depends on size, Nature Nanotechnology, vol.705, issue.12, pp.815-823, 2011.
DOI : 10.1038/nnano.2011.166

C. H. Choi, J. E. Zuckerman, P. Webster, and M. E. Davis, Targeting kidney mesangium by nanoparticles of defined size, Proceedings of the National Academy of Sciences, vol.108, issue.16, pp.6656-6661, 2011.
DOI : 10.1073/pnas.1103573108

H. S. Choi, L. Wenhao, P. Misra, E. Tanaka, J. P. Zimmer et al., Renal clearance of quantum dots, Nature Biotechnology, vol.361, issue.10, pp.1165-1170, 2007.
DOI : 10.1038/nbt1340

Y. Cirpanli, E. Allard, C. Passirani, E. Bilensoy, L. Lemaire et al., Antitumoral activity of camptothecin-loaded nanoparticles in 9L rat glioma model, International Journal of Pharmaceutics, vol.403, issue.1-2, pp.201-206, 2011.
DOI : 10.1016/j.ijpharm.2010.10.015

D. Jong, W. H. Hagens, W. I. Krystek, P. Burger, M. C. Sips et al., Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials, vol.29, issue.12, pp.1912-1919, 2008.
DOI : 10.1016/j.biomaterials.2007.12.037

H. K. De-wolf, C. J. Snel, F. J. Verbaan, R. M. Schiffelers, W. E. Hennink et al., Effect of cationic carriers on the pharmacokinetics and tumor localization of nucleic acids after intravenous administration, International Journal of Pharmaceutics, vol.331, issue.2, pp.167-175, 2007.
DOI : 10.1016/j.ijpharm.2006.10.029

S. Dufort, L. Sancey, C. Wenk, V. Josserand, and J. L. Coll, Optical small animal imaging in the drug discovery process, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1798, issue.12, pp.2266-2273, 2010.
DOI : 10.1016/j.bbamem.2010.03.016

URL : https://hal.archives-ouvertes.fr/inserm-00559533

O. C. Farokhzad and R. Langer, Impact of Nanotechnology on Drug Delivery, ACS Nano, vol.3, issue.1, pp.16-20, 2009.
DOI : 10.1021/nn900002m

E. Garcion, A. Lamprecht, B. Heurtault, A. Paillard, A. Aubert-pouessel et al., A new generation of anticancer, drug-loaded, colloidal vectors reverses multidrug resistance in glioma and reduces tumor progression in rats, Molecular Cancer Therapeutics, vol.5, issue.7, pp.1710-1722, 2006.
DOI : 10.1158/1535-7163.MCT-06-0289

URL : https://hal.archives-ouvertes.fr/hal-00358958

M. Goutayer, S. Dufort, V. Josserand, A. Royère, E. Heinrich et al., Tumor targeting of functionalized lipid nanoparticles: Assessment by in vivo fluorescence imaging, European Journal of Pharmaceutics and Biopharmaceutics, vol.75, issue.2, pp.137-147, 2010.
DOI : 10.1016/j.ejpb.2010.02.007

C. He, Y. Hu, L. Yin, C. Tang, and C. Yin, Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles, Biomaterials, vol.31, issue.13, pp.3657-3666, 2010.
DOI : 10.1016/j.biomaterials.2010.01.065

B. Heurtault, P. Saulnier, B. Pech, J. Proust, and J. Benoit, A novel phase inversionbased process for the preparation of lipid nanocarriers, Pharmaceutical Research, vol.19, issue.6, pp.875-880, 2002.
DOI : 10.1023/A:1016121319668

B. Heurtault, P. Saulnier, B. Pech, M. Venier-julienne, J. Proust et al., The influence of lipid nanocapsule composition on their size distribution, European Journal of Pharmaceutical Sciences, vol.18, issue.1, pp.55-61, 2003.
DOI : 10.1016/S0928-0987(02)00241-5

S. Hirn, M. Semmler-behnke, C. Schleh, A. Wenk, J. Lipka et al., Particle size-dependent and surface charge-dependent biodistribution of gold nanoparticles after intravenous administration, European Journal of Pharmaceutics and Biopharmaceutics, vol.77, issue.3, pp.407-416, 2011.
DOI : 10.1016/j.ejpb.2010.12.029

S. Hirsjärvi, S. Dufort, J. Gravier, I. Texier, Y. Qiao et al., Influence of size, surface coating and fine chemical composition on the in vitro reactivity and in vivo biodistribution of lipid nanocapsules versus lipid nanoemulsions in cancer models, Nanomedicine: Nanotechnology, Biology and Medicine, vol.9, issue.3, pp.375-387, 2013.
DOI : 10.1016/j.nano.2012.08.005

S. Hirsjärvi, C. Passirani, and J. Benoit, Passive and Active Tumour Targeting with Nanocarriers, Current Drug Discovery Technologies, vol.8, issue.3, pp.188-196, 2011.
DOI : 10.2174/157016311796798991

D. Hoarau, P. Delmas, S. David, E. Roux, and J. Leroux, Novel Long-Circulating Lipid Nanocapsules, Pharmaceutical Research, vol.21, issue.10, pp.1783-1789, 2004.
DOI : 10.1023/B:PHAM.0000045229.87844.21

Z. Jin, V. Josserand, S. Foillard, D. Boturyn, P. Dumy et al., In vivo optical imaging of integrin ??V-??3 in mice using multivalent or monovalent cRGD targeting vectors, Molecular Cancer, vol.6, issue.1, p.41, 2007.
DOI : 10.1186/1476-4598-6-41

J. Kim, W. I. Choi, Y. H. Kim, G. Tae, S. Lee et al., In-vivo tumor targeting of pluronic-based nano-carriers, Journal of Controlled Release, vol.147, issue.1, pp.109-117, 2010.
DOI : 10.1016/j.jconrel.2010.06.010

R. Kircheis, E. Ostermann, M. F. Wolschek, C. Lichtenberger, C. Magin-lachmann et al., Tumor-targeted gene delivery of tumor necrosis factor-?? induces tumor necrosis and tumor regression without systemic toxicity, Cancer Gene Therapy, vol.9, issue.8, pp.673-680, 2002.
DOI : 10.1038/sj.cgt.7700487

A. Kumari, S. K. Yadav, and S. C. Yadav, Biodegradable polymeric nanoparticles based drug delivery systems, Colloids and Surfaces B: Biointerfaces, vol.75, issue.1, pp.1-18, 2010.
DOI : 10.1016/j.colsurfb.2009.09.001

M. Kursa, G. F. Walker, V. Roessler, M. Ogris, W. Roedl et al., Novel Shielded Transferrin???Polyethylene Glycol???Polyethylenimine/DNA Complexes for Systemic Tumor-Targeted Gene Transfer, Bioconjugate Chemistry, vol.14, issue.1, pp.222-231, 2002.
DOI : 10.1021/bc0256087

F. Lacoeuille, F. Hindré, F. Moal, J. Roux, C. Passirani et al., In vivo evaluation of lipid nanocapsules as a promising colloidal carrier for paclitaxel, International Journal of Pharmaceutics, vol.344, issue.1-2, pp.143-149, 2007.
DOI : 10.1016/j.ijpharm.2007.06.014

URL : https://hal.archives-ouvertes.fr/inserm-00258366

A. Lamprecht, J. Saumet, J. Roux, and J. Benoit, Lipid nanocarriers as drug delivery system for ibuprofen in pain treatment, International Journal of Pharmaceutics, vol.278, issue.2, pp.407-414, 2004.
DOI : 10.1016/j.ijpharm.2004.03.018

H. Lee, H. Fonge, B. Hoang, R. M. Reilly, and C. Allen, The Effects of Particle Size and Molecular Targeting on the Intratumoral and Subcellular Distribution of Polymeric Nanoparticles, Molecular Pharmaceutics, vol.7, issue.4, pp.1195-1208, 2010.
DOI : 10.1021/mp100038h

H. Meng, M. Xue, T. Xia, Z. Ji, D. Y. Tarn et al., Use of Size and a Copolymer Design Feature To Improve the Biodistribution and the Enhanced Permeability and Retention Effect of Doxorubicin-Loaded Mesoporous Silica Nanoparticles in a Murine Xenograft Tumor Model, ACS Nano, vol.5, issue.5, pp.4131-4144, 2011.
DOI : 10.1021/nn200809t

F. Mévellec, F. Tisato, F. Refosco, A. Roucoux, N. Noiret et al., Synthesis and Characterization of the ???Sulfur-Rich??? Bis(perthiobenzoato)(dithiobenzoato)technetium(III) Heterocomplex, Inorganic Chemistry, vol.41, issue.3, pp.598-601, 2002.
DOI : 10.1021/ic0107577

M. Morille, T. Montier, P. Legras, N. Carmoy, P. Brodin et al., Long-circulating DNA lipid nanocapsules as new vector for passive tumor targeting, Biomaterials, vol.31, issue.2, pp.440-321, 2010.
DOI : 10.1016/j.biomaterials.2009.09.044

URL : https://hal.archives-ouvertes.fr/inserm-00491402

M. Morille, C. Passirani, S. Dufort, G. Bastiat, B. Pitard et al., Tumor transfection after systemic injection of DNA lipid nanocapsules, Biomaterials, vol.32, issue.9, pp.2327-2333, 2011.
DOI : 10.1016/j.biomaterials.2010.11.063

R. H. Müller, R. Shegokar, and C. M. Keck, 20 Years of Lipid Nanoparticles (SLN & NLC): Present State of Development & Industrial Applications, Current Drug Discovery Technologies, vol.8, issue.3, pp.207-227, 2011.
DOI : 10.2174/157016311796799062

J. H. Na, H. Koo, S. Lee, K. H. Min, K. Park et al., Real-time and non-invasive optical imaging of tumor-targeting glycol chitosan nanoparticles in various tumor models, Biomaterials, vol.32, issue.22, pp.5252-5261, 2011.
DOI : 10.1016/j.biomaterials.2011.03.076

A. Paillard, F. Hindré, C. Vignes-colombeix, J. Benoit, and E. Garcion, The importance of endo-lysosomal escape with lipid nanocapsules for drug subcellular bioavailability, Biomaterials, vol.31, issue.29, pp.7542-7554, 2010.
DOI : 10.1016/j.biomaterials.2010.06.024

S. Peltier, J. Oger, F. Lagarce, W. Couet, and J. Benoit, Enhanced Oral Paclitaxel Bioavailability After Administration of Paclitaxel-Loaded Lipid Nanocapsules, Pharmaceutical Research, vol.47, issue.6, pp.1243-1250, 2006.
DOI : 10.1007/s11095-006-0022-2

S. D. Perrault, C. Walkey, T. Jennings, H. C. Fischer, and W. C. Chan, Mediating Tumor Targeting Efficiency of Nanoparticles Through Design, Nano Letters, vol.9, issue.5, 1909.
DOI : 10.1021/nl900031y

J. Razkin, V. Josserand, D. Boturyn, Z. Jin, P. Dumy et al., Activatable Fluorescent Probes for Tumour-Targeting Imaging in Live Mice, ChemMedChem, vol.5704, issue.10, pp.1069-1072, 2006.
DOI : 10.1002/cmdc.200600118

URL : https://hal.archives-ouvertes.fr/inserm-00176684

L. Sancey, S. Dufort, V. Josserand, M. Keramidas, C. Righini et al., Drug development in oncology assisted by noninvasive optical imaging, International Journal of Pharmaceutics, vol.379, issue.2, pp.309-316, 2009.
DOI : 10.1016/j.ijpharm.2009.05.034

URL : https://hal.archives-ouvertes.fr/inserm-00390608

A. Schädlich, H. Caysa, T. Mueller, F. Tenambergen, C. Rose et al., Tumor Accumulation of NIR Fluorescent PEG???PLA Nanoparticles: Impact of Particle Size and Human Xenograft Tumor Model, ACS Nano, vol.5, issue.11, pp.8710-8720, 2011.
DOI : 10.1021/nn2026353

A. Schädlich, S. Hoffmann, T. Mueller, H. Caysa, C. Rose et al., Accumulation of nanocarriers in the ovary: A neglected toxicity risk?, Journal of Controlled Release, vol.160, issue.1, pp.105-112, 2012.
DOI : 10.1016/j.jconrel.2012.02.012

A. Schädlich, C. Rose, J. Kuntsche, H. Caysa, T. Mueller et al., How Stealthy are PEG-PLA Nanoparticles? An NIR In Vivo Study Combined with Detailed Size Measurements, Pharmaceutical Research, vol.48, issue.8, pp.1995-2007, 2011.
DOI : 10.1007/s11095-011-0426-5

G. Sonavane, K. Tomoda, and K. Makino, Biodistribution of colloidal gold nanoparticles after intravenous administration: Effect of particle size, Colloids and Surfaces B: Biointerfaces, vol.66, issue.2, pp.274-280, 2008.
DOI : 10.1016/j.colsurfb.2008.07.004

X. Sun, R. Rossin, J. L. Turner, M. L. Becker, M. J. Joralemon et al., An Assessment of the Effects of Shell Cross-Linked Nanoparticle Size, Core Composition, and Surface PEGylation on in Vivo Biodistribution, Biomacromolecules, vol.6, issue.5, pp.2541-2554, 2005.
DOI : 10.1021/bm050260e

V. Torchilin, Micellar Nanocarriers: Pharmaceutical Perspectives, Pharmaceutical Research, vol.277, issue.1, pp.1-16, 2007.
DOI : 10.1007/s11095-006-9132-0

V. P. Torchilin, Recent advances with liposomes as pharmaceutical carriers, Nature Reviews Drug Discovery, vol.103, issue.2, pp.145-160, 2005.
DOI : 10.1081/LPR-120017488

C. Vanpouille-box, F. Lacoeuille, C. Belloche, N. Lepareur, L. Lemaire et al., Tumor eradication in rat glioma and bypass of immunosuppressive barriers using internal radiation with 188Re-lipid nanocapsules, Biomaterials, vol.32, issue.28, pp.6781-6790, 2011.
DOI : 10.1016/j.biomaterials.2011.05.067

URL : https://hal.archives-ouvertes.fr/inserm-00638699

C. Vanpouille-box, F. Lacoeuille, J. Roux, C. Aubé, E. Garcion et al., Lipid Nanocapsules Loaded with Rhenium-188 Reduce Tumor Progression in a Rat Hepatocellular Carcinoma Model, PLoS ONE, vol.359, issue.3, p.490, 2011.
DOI : 10.1371/journal.pone.0016926.t004

URL : https://hal.archives-ouvertes.fr/hal-00741696

M. Weyland, F. Manero, A. Paillard, D. Grée, G. Viault et al., Mitochondrial targeting by use of lipid nanocapsules loaded with SV30, an analogue of the small-molecule Bcl-2 inhibitor HA14-1, Journal of Controlled Release, vol.151, issue.1, pp.74-82, 2011.
DOI : 10.1016/j.jconrel.2010.11.032

URL : https://hal.archives-ouvertes.fr/hal-00842655

A. Vonarbourg, C. Passirani, P. Saulnier, and J. Benoit, Parameters influencing the stealthiness of colloidal drug delivery systems, Biomaterials, vol.27, issue.24, pp.4356-4373, 2006.
DOI : 10.1016/j.biomaterials.2006.03.039

A. Vonarbourg, P. Saulnier, C. Passirani, and J. Benoit, Electrokinetic properties of noncharged lipid nanocapsules: Influence of the dipolar distribution at the interface, ELECTROPHORESIS, vol.15, issue.11, pp.2066-2075, 2005.
DOI : 10.1002/elps.200410145

Z. Yang, J. Leon, M. Martin, J. W. Harder, R. Zhang et al., Pharmacokinetics and biodistribution of near-infrared fluorescence polymeric nanoparticles, Nanotechnology, vol.20, issue.16, 2009.
DOI : 10.1088/0957-4484/20/16/165101

Z. Yang, S. Zheng, W. J. Harrison, J. Harder, X. Wen et al., Long-Circulating Near-Infrared Fluorescence Core-Cross-Linked Polymeric Micelles:?? Synthesis, Characterization, and Dual Nuclear/Optical Imaging, Biomacromolecules, vol.8, issue.11, pp.3422-3428, 2007.
DOI : 10.1021/bm7005399

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2542943

J. Zou, P. Saulnier, T. Perrier, Y. Zhang, T. Manninen et al., Distribution of lipid nanocapsules in different cochlear cell populations after round window membrane permeation, Journal of Biomedical Materials Research Part B: Applied Biomaterials, vol.100, issue.1/2, pp.10-18, 2008.
DOI : 10.1002/jbm.b.31058