N. Abed and P. Couvreur, Nanocarriers for antibiotics: A promising solution to treat intracellular bacterial infections, International Journal of Antimicrobial Agents, vol.43, issue.6, pp.485-496, 2014.
DOI : 10.1016/j.ijantimicag.2014.02.009

R. Aebersold, G. Bader, A. Edwards, J. Van-eyk, M. Kussmann et al., The Biology/Disease-driven Human Proteome Project (B/D-HPP): Enabling Protein Research for the Life Sciences Community, Disease-driven Human Proteome ProjectHPP): Enabling Protein Research for the Life Sciences Community, pp.23-27, 2013.
DOI : 10.1021/pr301151m

R. Aebersold and M. Mann, Mass spectrometry-based proteomics, Nature, vol.12, issue.6928, pp.198-207, 2003.
DOI : 10.1016/S0960-9822(01)00632-7

Y. Akiyama, T. Mori, Y. Katayama, and T. Niidome, The effects of PEG grafting level and injection dose on gold nanorod biodistribution in the tumor-bearing mice, Journal of Controlled Release, vol.139, issue.1, pp.81-84, 2009.
DOI : 10.1016/j.jconrel.2009.06.006

J. Alexander, History of the Medical Use of Silver, Surgical Infections, vol.10, issue.3, pp.289-292, 2009.
DOI : 10.1089/sur.2008.9941

M. Alibolandi, M. Mohammadi, S. Taghdisi, M. Ramezani, and K. Abnous, Fabrication of aptamer decorated dextran coated nano-graphene oxide for targeted drug delivery, Carbohydrate Polymers, vol.155, pp.218-229, 2017.
DOI : 10.1016/j.carbpol.2016.08.046

E. Allémann, R. Gurny, E. Doelker, F. Skinner, and H. Schütz, Distribution, kinetics and elimination of radioactivity after intravenous and intramuscular injection of 14C-savoxepine loaded poly(D,L-lactic acid) nanospheres to rats, Journal of Controlled Release, vol.29, issue.1-2, pp.97-104, 1994.
DOI : 10.1016/0168-3659(94)90125-2

T. Allen, Ligand-targeted therapeutics in anticancer therapy, Nature Reviews Cancer, vol.99, issue.1, pp.750-763, 2002.
DOI : 10.1073/pnas.122157899

B. Ames and L. Gold, Paracelsus to parascience: the environmental cancer distraction, Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, vol.447, issue.1, pp.3-13, 2000.
DOI : 10.1016/S0027-5107(99)00194-3

S. Antherieu, C. Chesne, R. Li, S. Camus, A. Lahoz et al., Stable Expression, Activity And Inductibility of Cytochromes P450 in Differentiated HepaRG Cells. Drug Metabolism and Disposition, 2009.

S. Anthérieu, A. Garat, N. Beauval, M. Soyez, D. Allorge et al., Comparison of cellular and transcriptomic effects between electronic cigarette vapor and cigarette smoke in human bronchial epithelial cells, Toxicology in Vitro, vol.45, 2017.
DOI : 10.1016/j.tiv.2016.12.015

J. Atienza, J. Zhu, X. Wang, X. Xu, and Y. Abassi, Dynamic Monitoring of Cell Adhesion and Spreading on Microelectronic Sensor Arrays, Journal of Biomolecular Screening, vol.292, issue.8, pp.795-805, 2005.
DOI : 10.1016/j.jim.2004.06.022

A. Baeza, E. Guisasola, E. Ruiz-hernández, and M. Vallet-regí, Magnetically Triggered Multidrug Release by Hybrid Mesoporous Silica Nanoparticles, Chemistry of Materials, vol.24, issue.3, pp.517-524, 2012.
DOI : 10.1021/cm203000u

E. Bajak, M. Fabbri, J. Ponti, S. Gioria, I. Ojea-jiménez et al., Changes in Caco-2 cells transcriptome profiles upon exposure to gold nanoparticles, Toxicology Letters, vol.233, issue.2, pp.187-199, 2015.
DOI : 10.1016/j.toxlet.2014.12.008

Y. Barenholz, Doxil?? ??? The first FDA-approved nano-drug: Lessons learned, Journal of Controlled Release, vol.160, issue.2, pp.117-134, 2012.
DOI : 10.1016/j.jconrel.2012.03.020

L. Barraud, P. Merle, E. Soma, L. Lefrançois, S. Guerret et al., Increase of doxorubicin sensitivity by doxorubicin-loading into nanoparticles for hepatocellular carcinoma cells in vitro and in vivo, Journal of Hepatology, vol.42, issue.5, pp.736-743, 2005.
DOI : 10.1016/j.jhep.2004.12.035

M. Barrow, A. Taylor, P. Murray, M. Rosseinsky, and D. Adams, Design considerations for the synthesis of polymer coated iron oxide nanoparticles for stem cell labelling and tracking using MRI, Chem. Soc. Rev., vol.9, issue.8, pp.6733-6748, 2015.
DOI : 10.1002/cmmi.1547

D. Bazile, C. Ropert, P. Huve, T. Verrecchia, M. Mariard et al., Body distribution of fully biodegradable [14C]-poly(lactic acid) nanoparticles coated with albumin after parenteral administration to rats, Biomaterials, vol.13, issue.15, pp.1093-1102, 1992.
DOI : 10.1016/0142-9612(92)90142-B

A. Beduneau, Z. Ma, C. Grotepas, A. Kabanov, B. Rabinow et al., Facilitated Monocyte-Macrophage Uptake and Tissue Distribution of Superparmagnetic Iron-Oxide Nanoparticles, PLoS ONE, vol.265, issue.2, p.4343, 2009.
DOI : 10.1371/journal.pone.0004343.g008

N. Biswas, Modified mesoporous silica nanoparticles for enhancing oral bioavailability and antihypertensive activity of poorly water soluble valsartan, European Journal of Pharmaceutical Sciences, vol.99, pp.152-160, 2017.
DOI : 10.1016/j.ejps.2016.12.015

T. Boissenot, A. Bordat, E. Fattal, and N. Tsapis, Ultrasound-triggered drug delivery for cancer treatment using drug delivery systems: From theoretical considerations to practical applications, Journal of Controlled Release, vol.241, pp.144-163, 2016.
DOI : 10.1016/j.jconrel.2016.09.026

P. Borm and W. Kreyling, Toxicological Hazards of Inhaled Nanoparticles???Potential Implications for Drug Delivery, Journal of Nanoscience and Nanotechnology, vol.4, issue.5, pp.521-531, 2004.
DOI : 10.1166/jnn.2004.081

K. Briley-saebo, A. Bjørnerud, D. Grant, H. Ahlstrom, T. Berg et al., Hepatic cellular distribution and degradation of iron oxide nanoparticles following single intravenous injection in rats: implications for magnetic resonance imaging, Cell and Tissue Research, vol.316, issue.3, pp.315-323, 2004.
DOI : 10.1007/s00441-004-0884-8

P. Browne, R. Judson, W. Casey, N. Kleinstreuer, and R. Thomas, Screening Chemicals for Estrogen Receptor Bioactivity Using a Computational Model, Environmental Science & Technology, vol.49, issue.14, pp.8804-8814, 2015.
DOI : 10.1021/acs.est.5b02641

A. Burns, J. Vider, H. Ow, E. Herz, O. Penate-medina et al., Fluorescent Silica Nanoparticles with Efficient Urinary Excretion for Nanomedicine, Nano Letters, vol.9, issue.1, pp.442-448, 2009.
DOI : 10.1021/nl803405h

F. Busquet, M. Palopoli, and T. Hartung, Regulatory Toxicology: Progress in Law, Science and the Law: Analytical Data in Support of Regulation in Health, Food, and the Environment, pp.51-69, 2014.

C. Caltagirone, A. Bettoschi, A. Garau, and R. Montis, Silica-based nanoparticles: a versatile tool for the development of efficient imaging agents, Chemical Society Reviews, vol.24, issue.126, pp.4645-4671, 2015.
DOI : 10.1002/adma.201104714

E. Casals, T. Pfaller, A. Duschl, G. Oostingh, and V. Puntes, Time Evolution of the Nanoparticle Protein Corona, ACS Nano, vol.4, issue.7, pp.3623-3632, 2010.
DOI : 10.1021/nn901372t

T. Cedervall, I. Lynch, S. Lindman, T. Berggård, E. Thulin et al., Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles, Proceedings of the National Academy of Sciences, vol.5, issue.3, pp.2050-2055, 2007.
DOI : 10.1021/pr050421l

W. Chan, Nanomedicine 2.0, Accounts of Chemical Research, vol.50, issue.3, pp.627-632, 2017.
DOI : 10.1021/acs.accounts.6b00629

B. Chang, X. Sha, J. Guo, Y. Jiao, C. Wang et al., Thermo and pH dual responsive, polymer shell coated, magnetic mesoporous silica nanoparticles for controlled drug release, Journal of Materials Chemistry, vol.3, issue.148, pp.9239-9247, 2011.
DOI : 10.1002/smll.200700005

X. Chen, H. Sun, J. Hu, X. Han, H. Liu et al., Transferrin gated mesoporous silica nanoparticles for redox-responsive and targeted drug delivery, Colloids and Surfaces B: Biointerfaces, vol.152, pp.77-84, 2017.
DOI : 10.1016/j.colsurfb.2017.01.010

J. Cheng, B. Teply, I. Sherifi, J. Sung, G. Luther et al., Formulation of functionalized PLGA???PEG nanoparticles for in vivo targeted drug delivery, Biomaterials, vol.28, issue.5, pp.869-876, 2007.
DOI : 10.1016/j.biomaterials.2006.09.047

V. Cheung, M. Morley, F. Aguilar, A. Massimi, R. Kucherlapati et al., Making and reading microarrays, Nature Genetics, vol.21, pp.15-24, 1999.
DOI : 10.1038/4439

N. Chiannilkulchai, Z. Driouich, J. Benoit, A. Parodi, and P. Couvreur, Doxorubicin-Loaded Nanoparticles: Increased Efficiency in Murine Hepatic Metastases, Selective Cancer Therapeutics, vol.5, issue.1, pp.1-11, 1989.
DOI : 10.1089/sct.1989.5.1

K. Cho, X. Wang, G. Kim, A. Gjyrezi, P. Giannakakou et al., Investigation of taxolresistance using folate-targeted ternary therapeutic nanoparticle, Cancer Research, vol.67, pp.2311-2311, 2007.

K. Cho, X. Wang, S. Nie, Z. Chen, and D. Shin, Therapeutic Nanoparticles for Drug Delivery in Cancer, Clinical Cancer Research, vol.14, issue.5, pp.1310-1316, 2008.
DOI : 10.1158/1078-0432.CCR-07-1441

URL : http://clincancerres.aacrjournals.org/content/clincanres/14/5/1310.full.pdf

M. Cho, W. Cho, M. Choi, S. Kim, B. Han et al., The impact of size on tissue distribution and elimination by single intravenous injection of silica nanoparticles, Toxicology Letters, vol.189, issue.3, pp.177-183, 2009.
DOI : 10.1016/j.toxlet.2009.04.017

J. Cottrell and U. London, Probability-based protein identification by searching sequence databases using mass spectrometry data, Electrophoresis, vol.20, pp.3551-3567, 1999.

F. Danhier, E. Ansorena, J. Silva, R. Coco, L. Breton et al., PLGA-based nanoparticles: An overview of biomedical applications, Journal of Controlled Release, vol.161, issue.2, pp.505-522, 2012.
DOI : 10.1016/j.jconrel.2012.01.043

S. Dave and X. Gao, Monodisperse magnetic nanoparticles for biodetection, imaging, and drug delivery: a versatile and evolving technology, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, vol.5, issue.3, pp.583-609, 2009.
DOI : 10.1038/nrc1566

J. Derksen, H. Morselt, D. Kalicharan, C. Hulstaert, and G. Scherphof, Interaction of immunoglobulin-coupled liposomes with rat liver macrophages in vitro, Experimental Cell Research, vol.168, issue.1, pp.105-115, 1987.
DOI : 10.1016/0014-4827(87)90420-4

A. Des-rieux, V. Fievez, M. Garinot, Y. Schneider, and V. Préat, Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach, Journal of Controlled Release, vol.116, issue.1, pp.1-27, 2006.
DOI : 10.1016/j.jconrel.2006.08.013

M. Djouina, N. Esquerre, P. Desreumaux, and C. Vignal, Toxicological consequences of experimental exposure to aluminum in human intestinal epithelial cells, Food and Chemical Toxicology, vol.91, pp.108-116
DOI : 10.1016/j.fct.2016.03.008

P. Durfee, Y. Lin, D. Dunphy, A. Muñiz, K. Butler et al., , 2016.

C. Fu, T. Liu, L. Li, H. Liu, D. Chen et al., The absorption, distribution, excretion and toxicity of mesoporous silica nanoparticles in mice following different exposure routes, Biomaterials, vol.34, issue.10, pp.2565-2575, 2013.
DOI : 10.1016/j.biomaterials.2012.12.043

K. Furumoto, K. Ogawara, M. Yoshida, Y. Takakura, M. Hashida et al., Biliary excretion of polystyrene microspheres depends on the type of receptor-mediated uptake in rat liver, BBA) -General Subjects, pp.221-226, 2001.
DOI : 10.1016/S0304-4165(01)00132-5

N. Gamage, A. Barnett, N. Hempel, R. Duggleby, K. Windmill et al., Human Sulfotransferases and Their Role in Chemical Metabolism, Toxicological Sciences, vol.90, issue.1, pp.5-22, 2006.
DOI : 10.1016/1357-2725(95)00164-6

F. Gentile, M. Ferrari, and P. Decuzzi, The Transport of Nanoparticles in Blood Vessels: The Effect of Vessel Permeability and Blood Rheology, Annals of Biomedical Engineering, vol.219, issue.1137, pp.254-261, 2008.
DOI : 10.1007/978-1-4419-6856-2

H. Gerets, E. Hanon, M. Cornet, S. Dhalluin, O. Depelchin et al., Selection of cytotoxicity markers for the screening of new chemical entities in a pharmaceutical context: A preliminary study using a multiplexing approach, Toxicology in Vitro, vol.23, issue.2, pp.319-332, 2009.
DOI : 10.1016/j.tiv.2008.11.012

H. Gerets, K. Tilmant, B. Gerin, H. Chanteux, B. Depelchin et al., Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins, Cell Biology and Toxicology, vol.105, issue.2, pp.69-87, 2012.
DOI : 10.1093/toxsci/kfn109

D. Goren, A. Horowitz, D. Tzemach, M. Tarshish, S. Zalipsky et al., Nuclear Delivery of Doxorubicin via Folate-targeted Liposomes with Bypass of Multidrug-resistance Efflux Pump, Clinical Cancer Research, vol.6, pp.1949-1957, 2000.

E. Gottmann, S. Kramer, B. Pfahringer, and C. Helma, Data Quality in Predictive Toxicology: Reproducibility of Rodent Carcinogenicity Experiments, Environmental Health Perspectives, vol.109, issue.5, pp.509-514, 2001.
DOI : 10.1289/ehp.01109509

T. Graham, Liquid Diffusion Applied to Analysis, Philosophical Transactions of the Royal Society of London, vol.151, issue.0, pp.183-224, 1861.
DOI : 10.1098/rstl.1861.0011

R. Gref, A. Domb, P. Quellec, T. Blunk, R. Müller et al., The controlled intravenous delivery of drugs using PEG-coated sterically stabilized nanospheres, Advanced Drug Delivery Reviews, vol.16, issue.2-3, pp.215-233, 1995.
DOI : 10.1016/0169-409X(95)00026-4

R. Gref, M. Lück, P. Quellec, M. Marchand, E. Dellacherie et al., ???Stealth??? corona-core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption, Colloids and Surfaces B: Biointerfaces, vol.18, issue.3-4, pp.301-313, 2000.
DOI : 10.1016/S0927-7765(99)00156-3

R. Gref, Y. Minamitake, M. Peracchia, V. Trubetskoy, V. Torchilin et al., Biodegradable long-circulating polymeric nanospheres, Science, vol.263, issue.5153, pp.1600-1603, 1994.
DOI : 10.1126/science.8128245

P. Gripon, S. Rumin, S. Urban, L. Seyec, J. Glaise et al.,

C. Guillouzo, Infection of a human hepatoma cell line by hepatitis B virus, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.15655-60, 2002.

E. Grzincic, J. Yang, J. Drnevich, P. Falagan-lotsch, and C. Murphy, Global transcriptomic analysis of model human cell lines exposed to surface-modified gold nanoparticles: the effect of surface chemistry, Nanoscale, vol.20, issue.4, pp.1349-1362, 2015.
DOI : 10.1002/adma.200701853

A. Guillouzo, A. Corlu, C. Aninat, D. Glaise, F. Morel et al., The human hepatoma HepaRG cells: A highly differentiated model for studies of liver metabolism and toxicity of xenobiotics, Chemico-Biological Interactions, vol.168, issue.1, pp.66-73, 2007.
DOI : 10.1016/j.cbi.2006.12.003

W. Hagens, A. Oomen, W. De-jong, F. Cassee, and A. Sips, What do we (need to) know about the kinetic properties of nanoparticles in the body?, Regulatory Toxicology and Pharmacology, vol.49, issue.3, pp.217-229, 2007.
DOI : 10.1016/j.yrtph.2007.07.006

I. Hamad, O. Hanbali, A. Hunter, K. Rutt, T. Andresen et al., Distinct Polymer Architecture Mediates Switching of Complement Activation Pathways at the Nanosphere???Serum Interface: Implications for Stealth Nanoparticle Engineering, ACS Nano, vol.4, issue.11, pp.6629-6638, 2010.
DOI : 10.1021/nn101990a

URL : http://eprints.brighton.ac.uk/7916/2/Hunter_Rutt_Distinct_polymer.pdf

J. Hardman and L. Limbird, Goodman & Gilman's The Pharmacological Basis of Therapeutics, 10th, 2001.

T. Hartung, Toxicology for the twenty-first century, Nature, vol.109, issue.7252, pp.208-220, 2009.
DOI : 10.1038/460208a

T. Hartung, Food for thought ??? on alternative methods for nanoparticle safety testing, ALTEX, vol.27, pp.87-95, 2010.
DOI : 10.14573/altex.2010.2.87

URL : https://www.altex.org/index.php/altex/article/download/557/566

T. Hartung, Evolution of toxicological science: the need for change, International Journal of Risk Assessment and Management, vol.20, issue.1/2/3, pp.21-45, 2017.
DOI : 10.1504/IJRAM.2017.082570

URL : http://www.inderscience.com/filter.php?aid=82570

T. Hartung, Opinion versus evidence for the need to move away from animal testing, ALTEX, vol.34, pp.193-200, 2017.
DOI : 10.14573/altex.1703291

URL : http://www.altex.ch/resources/altex_2017_2_193_200_FFT_Hartung.pdf

T. Hartung, Utility of the adverse outcome pathway concept in drug development, Expert Opinion on Drug Metabolism & Toxicology, vol.32, issue.1, pp.1-3, 2017.
DOI : 10.14573/altex.2013.2.119

X. He, H. Nie, K. Wang, W. Tan, X. Wu et al., In Vivo Study of Biodistribution and Urinary Excretion of Surface-Modified Silica Nanoparticles, Analytical Chemistry, vol.80, issue.24, pp.9597-9603, 2008.
DOI : 10.1021/ac801882g

B. Hicke, A. Stephens, T. Gould, Y. Chang, C. Lynott et al., Tumor targeting by an aptamer, Journal of nuclear medicine : official publication, Society of Nuclear Medicine, vol.47, pp.668-78, 2006.

D. Hobson, Nanotoxicology, International Journal of Toxicology, vol.35, issue.1, pp.3-4, 2016.
DOI : 10.1177/1091581816631729

X. Huang and C. Brazel, On the importance and mechanisms of burst release in matrix-controlled drug delivery systems, Journal of Controlled Release, vol.73, issue.2-3, pp.121-136, 2001.
DOI : 10.1016/S0168-3659(01)00248-6

X. Huang, I. El-sayed, W. Qian, and M. El-sayed, Cancer Cell Imaging and Photothermal Therapy in the Near-Infrared Region by Using Gold Nanorods, Journal of the American Chemical Society, vol.128, issue.6, pp.2115-2120, 2006.
DOI : 10.1021/ja057254a

X. Huang, P. Jain, I. El-sayed, and M. El-sayed, Plasmonic photothermal therapy (PPTT) using gold nanoparticles, Lasers in Medical Science, vol.297, issue.6, p.217, 2007.
DOI : 10.1177/153303460300200602

A. Irfan, M. Cauchi, W. Edmands, N. Gooderham, J. Njuguna et al., Assessment of Temporal Dose-Toxicity Relationship of Fumed Silica Nanoparticle in Human Lung A549 Cells by Conventional Cytotoxicity and 1H-NMR-Based Extracellular Metabonomic Assays, Toxicological Sciences, vol.134, issue.2, pp.354-364, 2014.
DOI : 10.1021/ja304907c

T. Ishida and H. Kiwada, Accelerated blood clearance (ABC) phenomenon upon repeated injection of PEGylated liposomes, International Journal of Pharmaceutics, vol.354, issue.1-2, pp.56-62, 2008.
DOI : 10.1016/j.ijpharm.2007.11.005

I. Ivens, W. Achanzar, A. Baumann, A. Brändli-baiocco, J. Cavagnaro et al., PEGylated Biopharmaceuticals, Toxicologic Pathology, vol.45, issue.7, pp.959-983, 2015.
DOI : 10.1111/j.2042-7158.1995.tb05835.x

URL : http://journals.sagepub.com/doi/pdf/10.1177/0192623315591171

A. Iyer, G. Khaled, J. Fang, and H. Maeda, Exploiting the enhanced permeability and retention effect for tumor targeting, Drug Discovery Today, vol.11, issue.17-18, pp.812-818, 2006.
DOI : 10.1016/j.drudis.2006.07.005

A. Jain and S. Jain, In vitro and cell uptake studies for targeting of ligand anchored nanoparticles for colon tumors, European Journal of Pharmaceutical Sciences, vol.35, issue.5, pp.404-416, 2008.
DOI : 10.1016/j.ejps.2008.08.008

P. Jain, X. Huang, I. El-sayed, and M. El-sayed, Noble Metals on the Nanoscale: Optical and Photothermal Properties and Some Applications in Imaging, Sensing, Biology, and Medicine, Accounts of Chemical Research, vol.41, issue.12, pp.1578-1586, 2008.
DOI : 10.1021/ar7002804

R. Jain, The manufacturing techniques of various drug loaded biodegradable poly(lactide-co-glycolide) (PLGA) devices, Biomaterials, vol.21, issue.23, pp.2475-2490, 2000.
DOI : 10.1016/S0142-9612(00)00115-0

P. Jancova, P. Anzenbacher, and E. Anzenbacherova, PHASE II DRUG METABOLIZING ENZYMES, Biomedical Papers, vol.154, issue.2, pp.103-116, 2010.
DOI : 10.5507/bp.2010.017

URL : http://biomed.papers.upol.cz/doi/10.5507/bp.2010.017.pdf

Z. Jin, Y. Lv, H. Cao, J. Yao, J. Zhou et al., Core-shell nanocarriers with high paclitaxel loading for passive and active targeting, Scientific Reports, vol.125, issue.1, p.27559, 2016.
DOI : 10.1242/jcs.109520

URL : http://www.nature.com/articles/srep27559.pdf

V. Kagan, H. Bayir, and A. Shvedova, Nanomedicine and nanotoxicology: two sides of the same coin, Nanomedicine: Nanotechnology, Biology and Medicine, vol.1, issue.4, pp.313-316, 2005.
DOI : 10.1016/j.nano.2005.10.003

G. Kandasamy and D. Maity, Recent advances in superparamagnetic iron oxide nanoparticles (SPIONs) for in vitro and in vivo cancer nanotheranostics, International Journal of Pharmaceutics, vol.496, issue.2, pp.191-218, 2015.
DOI : 10.1016/j.ijpharm.2015.10.058

S. Khanna, T. Jecklin, and P. Speiser, Bead Polymerization Technique for Sustained-Release Dosage Form, Journal of Pharmaceutical Sciences, vol.59, issue.5, pp.614-618, 1970.
DOI : 10.1002/jps.2600590508

S. Khanna and P. Speiser, Epoxy resin beads as a pharmaceutical dosage form I: Method of preparation, Journal of Pharmaceutical Sciences, vol.58, issue.9, pp.1114-1117, 1969.
DOI : 10.1002/jps.2600580916

H. Kim, K. Andrieux, C. Delomenie, H. Chacun, M. Appel et al., Analysis of plasma protein adsorption onto PEGylated nanoparticles by complementary methods: 2-DE, CE and Protein Lab-on-chip?? system, ELECTROPHORESIS, vol.19, issue.13, pp.2252-2261, 2007.
DOI : 10.1016/0304-4157(92)90038-C

J. Kim, H. Kim, N. Lee, T. Kim, H. Kim et al., Multifunctional Uniform Nanoparticles Composed of a Magnetite Nanocrystal Core and a Mesoporous Silica Shell for Magnetic Resonance and Fluorescence Imaging and for Drug Delivery, Angewandte Chemie International Edition, vol.54, issue.44, pp.8438-8441, 2008.
DOI : 10.1002/anie.200802469

J. Kim, J. Lee, J. Lee, J. Yu, B. Kim et al., Magnetic Fluorescent Delivery Vehicle Using Uniform Mesoporous Silica Spheres Embedded with Monodisperse Magnetic and Semiconductor Nanocrystals, Journal of the American Chemical Society, vol.128, issue.3, pp.688-689, 2006.
DOI : 10.1021/ja0565875

J. Kim, A. Salvati, C. Aberg, and K. Dawson, Suppression of nanoparticle cytotoxicity approaching in vivo serum concentrations: limitations of in vitro testing for nanosafety, Nanoscale, vol.8, issue.23, pp.14180-14184, 2014.
DOI : 10.1021/nn502356f

R. Kim, J. Choi, B. Kim, and W. Kim, Comparative Analysis of Transcriptional Profile Changes in Larval Zebrafish Exposed to Zinc Oxide Nanoparticles and Zinc Sulfate, Bulletin of Environmental Contamination and Toxicology, vol.20, issue.137, pp.183-189, 2017.
DOI : 10.3762/bjnano.6.160

T. Kim, E. Momin, J. Choi, K. Yuan, H. Zaidi et al., Contrast Agents for Labeling and MRI Tracking of Adipose-Derived Mesenchymal Stem Cells, Journal of the American Chemical Society, vol.133, issue.9, pp.2955-2961, 2011.
DOI : 10.1021/ja1084095

A. Klibanov, K. Maruyama, A. Beckerleg, V. Torchilin, and L. Huang, Activity of amphipathic poly(ethylene glycol) 5000 to prolong the circulation time of liposomes depends on the liposome size and is unfavorable for immunoliposome binding to target, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1062, issue.2, pp.142-148, 1991.
DOI : 10.1016/0005-2736(91)90385-L

N. Knezevic, E. Ruiz-hernandez, W. Hennink, and M. Vallet-regi, Magnetic mesoporous silica-based core/shell nanoparticles for biomedical applications, RSC Advances, vol.18, issue.25, pp.9584-9593, 2013.
DOI : 10.1002/chem.201200030

K. Knop, R. Hoogenboom, D. Fischer, and U. Schubert, Poly(ethylene glycol) in Drug Delivery: Pros and Cons as Well as Potential Alternatives, Angewandte Chemie International Edition, vol.61, issue.36, pp.6288-6308, 2010.
DOI : 10.3109/10731199009117287

R. Kodama, Magnetic nanoparticles, Journal of Magnetism and Magnetic Materials, vol.200, issue.1-3, pp.359-372, 1999.
DOI : 10.1016/S0304-8853(99)00347-9

D. Kohane and R. Langer, Biocompatibility and drug delivery systems, Chem. Sci., vol.22, issue.4, pp.441-446, 2010.
DOI : 10.1038/nbt981

J. Kreuter, Nanoparticles???a historical perspective, International Journal of Pharmaceutics, vol.331, issue.1, pp.1-10, 2007.
DOI : 10.1016/j.ijpharm.2006.10.021

J. Kreuter, U. Täuber, and V. Illi, Distribution and Elimination of Poly(methyl-2-14C-methacrylate) Nanoparticle Radioactivity after Injection in Rats and Mice, Journal of Pharmaceutical Sciences, vol.68, issue.11, pp.1443-1447, 1979.
DOI : 10.1002/jps.2600681129

K. Krishnan, A. Pakhomov, Y. Bao, P. Blomqvist, Y. Chun et al., Nanomagnetism and spin electronics: materials, microstructure and novel properties, Journal of Materials Science, vol.97, issue.3, pp.793-815, 2006.
DOI : 10.1103/PhysRevB.65.161201

M. Krukemeyer, V. Krenn, F. Huebner, W. Wagner, and R. Resch, History and Possible Uses of Nanomedicine Based on Nanoparticles and Nanotechnological Progress, Journal of Nanomedicine & Nanotechnology, vol.6, p.1, 2015.

A. Kunzmann, B. Andersson, T. Thurnherr, H. Krug, A. Scheynius et al., Toxicology of engineered nanomaterials: Focus on biocompatibility, biodistribution and biodegradation, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1810, issue.3, pp.361-73, 2011.
DOI : 10.1016/j.bbagen.2010.04.007

T. Laakso, P. Artursson, and I. Sjöholm, Biodegradable Microspheres IV: Factors Affecting the Distribution and Degradation of Polyacryl Starch Microparticles, Journal of Pharmaceutical Sciences, vol.75, issue.10, pp.962-967, 1986.
DOI : 10.1002/jps.2600751011

H. Labouta and M. Schneider, Interaction of inorganic nanoparticles with the skin barrier: current status and critical review, Nanomedicine: Nanotechnology, Biology and Medicine, vol.9, issue.1, pp.39-54, 2013.
DOI : 10.1016/j.nano.2012.04.004

S. Lai, Y. Wang, and J. Hanes, Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues, Advanced Drug Delivery Reviews, vol.61, issue.2, pp.158-171, 2009.
DOI : 10.1016/j.addr.2008.11.002

URL : http://europepmc.org/articles/pmc2667119?pdf=render

C. Lambert, C. Spire, C. N. Guillouzo, and A. , Dose- and time-dependent effects of phenobarbital on gene expression profiling in human hepatoma HepaRG cells, Toxicology and Applied Pharmacology, vol.234, issue.3, pp.345-360, 2009.
DOI : 10.1016/j.taap.2008.11.008

T. Lammers, F. Kiessling, W. Hennink, and G. Storm, Drug targeting to tumors: Principles, pitfalls and (pre-) clinical progress, Journal of Controlled Release, vol.161, issue.2, pp.175-187, 2012.
DOI : 10.1016/j.jconrel.2011.09.063

A. Lamprecht, U. Schäfer, and C. Lehr, Size-Dependent Bioadhesion of Micro-and Nanoparticulate Carriers to the Inflamed Colonic Mucosa, Pharmaceutical Research, vol.18, issue.6, pp.788-793, 2001.
DOI : 10.1023/A:1011032328064

E. Lander, L. Linton, B. Birren, C. Nusbaum, M. Zody et al., Initial sequencing and analysis of the human genome, Nature, vol.6, issue.6822, pp.860-921, 2001.
DOI : 10.1089/cmb.1999.6.91

L. Filon, F. Mauro, M. Adami, G. Bovenzi, M. Crosera et al., Nanoparticles skin absorption: New aspects for a safety profile evaluation, Regulatory Toxicology and Pharmacology, vol.72, issue.2, pp.310-322, 2015.
DOI : 10.1016/j.yrtph.2015.05.005

L. Ray, A. Vert, M. Gautier, J. Benoît, and J. , Fate of [14C]poly(DL-lactide-co-glycolide) nanoparticles after intravenous and oral administration to mice, International Journal of Pharmaceutics, vol.106, issue.3, pp.201-211, 1994.
DOI : 10.1016/0378-5173(94)90003-5

C. Leamon and J. Reddy, Folate-targeted chemotherapy, Advanced Drug Delivery Reviews, vol.56, issue.8, pp.1127-1141, 2004.
DOI : 10.1016/j.addr.2004.01.008

H. Lee, T. Leavens, S. Mason, N. Monteiro-riviere, and J. Riviere, Comparison of Quantum Dot Biodistribution with a Blood-Flow-Limited Physiologically Based Pharmacokinetic Model, Nano Letters, vol.9, issue.2, pp.794-799, 2009.
DOI : 10.1021/nl803481q

H. Lee, S. Yeo, and S. Jeong, Antibacterial effect of nanosized silver colloidal solution on textile fabrics, Journal of Materials Science, vol.38, issue.10, pp.2199-2204, 2003.
DOI : 10.1023/A:1023736416361

M. Leist, N. Hasiwa, C. Rovida, M. Daneshian, D. Basketter et al., Consensus report on the future of animal-free systemic toxicity testing, 2014.

A. Lesniak, F. Fenaroli, M. Monopoli, C. Åberg, K. Dawson et al., Effects of the Presence or Absence of a Protein Corona on Silica Nanoparticle Uptake and Impact on Cells, ACS Nano, vol.6, issue.7, pp.5845-5857, 2012.
DOI : 10.1021/nn300223w

A. Lesniak, A. Salvati, M. Santos-martinez, M. Radomski, K. Dawson et al., Nanoparticle Adhesion to the Cell Membrane and Its Effect on Nanoparticle Uptake Efficiency, Journal of the American Chemical Society, vol.135, issue.4, pp.1438-1444, 2013.
DOI : 10.1021/ja309812z

M. Li, K. Jamal, K. Kostarelos, and J. Reineke, Physiologically Based Pharmacokinetic Modeling of Nanoparticles, ACS Nano, vol.4, issue.11, pp.6303-6317, 2010.
DOI : 10.1021/nn1018818

Z. Li, J. Barnes, A. Bosoy, J. Stoddart, and J. Zink, Mesoporous silica nanoparticles in biomedical applications, Chemical Society Reviews, vol.5, issue.7, pp.2590-2605, 2012.
DOI : 10.1021/nn200809t

URL : http://onlinelibrary.wiley.com/doi/10.1002/jbm.a.35654/pdf

H. Liang, C. Chen, S. Chen, A. Kulkarni, Y. Chiu et al., Paclitaxel-loaded poly(??-glutamic acid)-poly(lactide) nanoparticles as a targeted drug delivery system for the treatment of liver cancer, Biomaterials, vol.27, issue.9, pp.2051-2059, 2006.
DOI : 10.1016/j.biomaterials.2005.10.027

J. Liu, C. Detrembleur, D. Pauw-gillet, M. Mornet, S. Elst et al., Heat-triggered drug release systems based on mesoporous silica nanoparticles filled with a maghemite core and phase-change molecules as gatekeepers, J. Mater. Chem. B, vol.7, issue.1, pp.59-70, 2014.
DOI : 10.1002/cmmi.1473

URL : https://hal.archives-ouvertes.fr/hal-00923633

J. Liu, T. Wei, J. Zhao, Y. Huang, H. Deng et al., Multifunctional aptamer-based nanoparticles for targeted drug delivery to circumvent cancer resistance, Biomaterials, vol.91, pp.44-56, 2016.
DOI : 10.1016/j.biomaterials.2016.03.013

J. Liu, J. Xu, J. Zhou, Y. Zhang, D. Guo et al., Fe<sub>3</sub>O<sub>4</sub>-based PLGA nanoparticles as MR contrast agents for the detection of thrombosis, International Journal of Nanomedicine, vol.12, pp.1113-1126, 2017.
DOI : 10.2147/IJN.S123228

T. Liu, L. Li, X. Teng, X. Huang, H. Liu et al., Single and repeated dose toxicity of mesoporous hollow silica nanoparticles in intravenously exposed mice, Biomaterials, vol.32, issue.6, pp.1657-68, 2011.
DOI : 10.1016/j.biomaterials.2010.10.035

W. Liu, I. Worms, N. Herlin-boime, D. Truffier-boutry, I. Michaud-soret et al.,

F. Genetet, Interaction of silver nanoparticles with metallothionein and ceruloplasmin: impact on metal substitution by Ag(i), corona formation and enzymatic activity, Nanoscale, vol.9, pp.6581-6594, 2017.
URL : https://hal.archives-ouvertes.fr/cea-01510783

Y. Liu, Z. Chen, C. Liu, D. Yu, Z. Lu et al., Gadolinium-loaded polymeric nanoparticles modified with Anti-VEGF as multifunctional MRI contrast agents for the diagnosis of liver cancer, Biomaterials, vol.32, issue.22, pp.5167-5176, 2011.
DOI : 10.1016/j.biomaterials.2011.03.077

Z. Liu, S. Tang, Z. Xu, Y. Wang, X. Zhu et al., Preparation and in vitro evaluation of a multifunctional iron silicate@ liposome nanohybrid for pH-sensitive doxorubicin delivery and photoacoustic imaging, Journal of Nanomaterials, vol.16, p.44, 2015.

M. Longmire, P. Choyke, and H. Kobayashi, Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats, Nanomedicine, vol.3, issue.5, pp.703-717, 2008.
DOI : 10.1021/ja710193c

J. Lu, M. Liong, Z. Li, J. Zink, and F. Tamanoi, Biocompatibility, Biodistribution, and Drug-Delivery Efficiency of Mesoporous Silica Nanoparticles for Cancer Therapy in Animals, Small, vol.25, issue.16, pp.1794-1805, 2010.
DOI : 10.1002/smll.201000538

T. Luechtefeld, A. Maertens, D. Russo, C. Rovida, H. Zhu et al., Analysis of Draize eye irritation testing and its prediction by mining publicly available 2008-2014 REACH data. Alternatives to Animal Experimentation, ALTEX, vol.33, pp.123-134, 2016.

T. Luechtefeld, A. Maertens, D. Russo, C. Rovida, H. Zhu et al., Analysis of publically available skin sensitization data from REACH registrations Alternatives to Animal Experimentation, ALTEX, vol.33, pp.135-148, 2008.

B. Lundberg, G. Griffiths, and H. Hansen, Cellular association and cytotoxicity of anti-CD74-targeted lipid drug-carriers in B lymphoma cells, Journal of Controlled Release, vol.94, issue.1, pp.155-161, 2004.
DOI : 10.1016/j.jconrel.2003.09.016

M. Lundqvist, J. Stigler, T. Cedervall, T. Berggård, M. Flanagan et al., The Evolution of the Protein Corona around Nanoparticles: A Test Study, ACS Nano, vol.5, issue.9, pp.7503-7509, 2011.
DOI : 10.1021/nn202458g

M. Lundqvist, J. Stigler, E. G. Lynch, I. Cedervall, T. Dawson et al., Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts, Proceedings of the National Academy of Sciences, vol.67, issue.8, pp.14265-14270, 2008.
DOI : 10.1021/ac00104a020

O. Lunov, T. Syrovets, C. Loos, J. Beil, M. Delacher et al., Differential Uptake of Functionalized Polystyrene Nanoparticles by Human Macrophages and a Monocytic Cell Line, ACS Nano, vol.5, issue.3, pp.1657-1669, 2011.
DOI : 10.1021/nn2000756

J. Macdonald and R. Robertson, Toxicity Testing in the 21st Century: A View from the Pharmaceutical Industry, Toxicological Sciences, vol.165, issue.1, pp.40-46, 2009.
DOI : 10.1001/archinte.165.12.1363

H. Maeda, The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting, Advances in Enzyme Regulation, vol.41, issue.1, pp.189-207, 2001.
DOI : 10.1016/S0065-2571(00)00013-3

H. Maeda, G. Bharate, and J. Daruwalla, Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect, European Journal of Pharmaceutics and Biopharmaceutics, vol.71, issue.3, pp.409-419, 2009.
DOI : 10.1016/j.ejpb.2008.11.010

P. Maincent, P. Thouvenot, C. Amicabile, M. Hoffman, J. Kreuter et al., Lymphatic Targeting of Polymeric Nanoparticles After Intraperitoneal Administration in Rats, Pharmaceutical Research, vol.09, issue.12, pp.1534-1539, 1992.
DOI : 10.1023/A:1015895804597

Y. Malam, M. Loizidou, and A. Seifalian, Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer, Trends in Pharmacological Sciences, vol.30, issue.11, pp.592-599, 2009.
DOI : 10.1016/j.tips.2009.08.004

V. Mamaeva, C. Sahlgren, and M. Lindén, Mesoporous silica nanoparticles in medicine???Recent advances, Advanced Drug Delivery Reviews, vol.65, issue.5, pp.689-702, 2013.
DOI : 10.1016/j.addr.2012.07.018

H. Marie, L. Lemaire, F. Franconi, S. Lajnef, Y. Frapart et al., Superparamagnetic Liposomes for MRI Monitoring and External Magnetic Field-Induced Selective Targeting of Malignant Brain Tumors, Advanced Functional Materials, vol.103, issue.8, pp.1258-1269, 2015.
DOI : 10.1007/s11060-010-0389-0

URL : https://hal.archives-ouvertes.fr/hal-01392430

F. Martín-saavedra, E. Ruíz-hernández, A. Boré, D. Arcos, M. Vallet-regí et al., Magnetic mesoporous silica spheres for hyperthermia therapy, Acta Biomaterialia, vol.6, issue.12, pp.4522-4531, 2010.
DOI : 10.1016/j.actbio.2010.06.030

M. Martina, J. Fortin, L. Fournier, C. Ménager, F. Gazeau et al., Magnetic Targeting of Rhodamine-Labeled Superparamagnetic Liposomes to Solid Tumors: In Vivo Tracking by Fibered Confocal Fluorescence Microscopy, Molecular Imaging, vol.6, issue.2, p.729000004, 2007.
DOI : 10.2310/7290.2007.00004

URL : https://hal.archives-ouvertes.fr/hal-00162290

C. Mathe, S. Devineau, J. Aude, G. Lagniel, S. Chedin et al., Structural Determinants for Protein adsorption/non-adsorption to Silica Surface, PLoS ONE, vol.55, issue.11, p.81346, 2013.
DOI : 10.1371/journal.pone.0081346.s003

Y. Matsumura and H. Maeda, A New Concept for Macromolecular Therapeutics in Cancer Chemotherapy: Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs, Cancer Research, vol.46, pp.6387-6392, 1986.

A. Maynard, R. Aitken, T. Butz, V. Colvin, K. Donaldson et al., Safe handling of nanotechnology, Nature, vol.67, issue.7117, pp.267-269, 2006.
DOI : 10.1016/S0003-4878(98)00044-1

URL : http://www.nature.com/nature/journal/v444/n7117/pdf/444267a.pdf

R. Mcdonald, J. Mcdonald, D. Kallmes, M. Jentoft, D. Murray et al., Intracranial Gadolinium Deposition after Contrast-enhanced MR Imaging, Radiology, vol.275, issue.3, pp.772-782, 2015.
DOI : 10.1148/radiol.15150025

J. Mcquillan and A. Shaw, : A full transcriptomic profile, Nanotoxicology, vol.86, issue.sup1, pp.177-184, 2014.
DOI : 10.1021/nl301934w

H. Meng, T. Xia, S. George, and A. Nel, A Predictive Toxicological Paradigm for the Safety Assessment of Nanomaterials, ACS Nano, vol.3, issue.7, pp.1620-1627, 2009.
DOI : 10.1021/nn9005973

M. Miller and Y. Tang, Basic Concepts of Microarrays and Potential Applications in Clinical Microbiology, Clinical Microbiology Reviews, vol.22, issue.4, pp.611-633, 2009.
DOI : 10.1128/CMR.00019-09

URL : http://cmr.asm.org/content/22/4/611.full.pdf

V. Mirshafiee, R. Kim, S. Park, M. Mahmoudi, and M. Kraft, Impact of protein pre-coating on the protein corona composition and nanoparticle cellular uptake, Biomaterials, vol.75, pp.295-304, 2016.
DOI : 10.1016/j.biomaterials.2015.10.019

V. Mirshafiee, M. Mahmoudi, K. Lou, J. Cheng, and M. Kraft, Protein corona significantly reduces active targeting yield, Chemical Communications, vol.3, issue.25, pp.2557-2559, 2013.
DOI : 10.1039/C2RA22093H

URL : http://europepmc.org/articles/pmc3671387?pdf=render

S. Moghimi, Modulation of lymphatic distribution of subcutaneously injected poloxamer 407-coated nanospheres: the effect of the ethylene oxide chain configuration, FEBS Letters, vol.14, issue.1-3, pp.241-244, 2003.
DOI : 10.1023/A:1012145410859

S. Moghimi, A. Hunter, and J. Murray, Long-Circulating and Target-Specific Nanoparticles: Theory to Practice, Pharmacological Reviews, vol.53, pp.283-318, 2001.

M. Monopoli, C. Aberg, A. Salvati, and K. Dawson, Biomolecular coronas provide the biological identity of nanosized materials, Nature Nanotechnology, vol.7, issue.12, pp.779-786, 2012.
DOI : 10.1016/j.chemosphere.2010.07.062

URL : http://researchrepository.ucd.ie/bitstream/10197/4572/1/NNANO-12030435B-Dawson-AL_1350526345_2.pdf

M. Monopoli, D. Walczyk, A. Campbell, G. Elia, I. Lynch et al., Biological Impacts of Nanoparticles, Journal of the American Chemical Society, vol.133, issue.8, pp.2525-2559, 2011.
DOI : 10.1021/ja107583h

J. Mosafer, K. Abnous, M. Tafaghodi, H. Jafarzadeh, and M. Ramezani, Preparation and characterization of uniform-sized PLGA nanospheres encapsulated with oleic acid-coated magnetic-Fe 3 O 4 nanoparticles for simultaneous diagnostic and therapeutic applications, Colloids and Surfaces A: Physicochemical and Engineering Aspects, vol.514, pp.146-154, 2017.
DOI : 10.1016/j.colsurfa.2016.11.056

Q. Mu, S. Zhai, and Y. B. , Real-Time Monitoring of Cellular Responses to Carbon Nanotubes, Methods Mol Biol, vol.625, pp.85-94, 2010.
DOI : 10.1007/978-1-60761-579-8_8

S. Mura, N. J. Couvreur, and P. , Stimuli-responsive nanocarriers for drug delivery, Nature Materials, vol.9, issue.11, pp.991-1003, 2013.
DOI : 10.1021/mp300165s

M. Nag, V. Gajbhiye, P. Kesharwani, and N. Jain, Transferrin functionalized chitosan-PEG nanoparticles for targeted delivery of paclitaxel to cancer cells, Colloids and Surfaces B: Biointerfaces, vol.148, pp.363-370, 2016.
DOI : 10.1016/j.colsurfb.2016.08.059

D. Napierska, L. Thomassen, D. Lison, J. Martens, and P. Hoet, The nanosilica hazard: another variable entity, Particle and Fibre Toxicology, vol.7, issue.1, p.39, 2010.
DOI : 10.1186/1743-8977-7-39

URL : https://particleandfibretoxicology.biomedcentral.com/track/pdf/10.1186/1743-8977-7-39?site=particleandfibretoxicology.biomedcentral.com

A. Nel, L. Madler, D. Velegol, T. Xia, E. Hoek et al., Understanding biophysicochemical interactions at the nano???bio interface, Nature Materials, vol.20, issue.7, pp.543-557, 2009.
DOI : 10.1289/ehp.6000

D. Nelson, D. Zeldin, S. Hoffman, L. Maltais, H. Wain et al., Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants, Pharmacogenetics, vol.14, issue.1, pp.1-18, 2004.
DOI : 10.1097/00008571-200401000-00001

L. Nelson, K. Morgan, P. Treskes, K. Samuel, C. Henderson et al., Human Hepatic HepaRG Cells Maintain an Organotypic Phenotype with High Intrinsic CYP450 Activity/Metabolism and Significantly Outperform Standard HepG2/C3A Cells for Pharmaceutical and Therapeutic Applications, Basic & Clinical Pharmacology & Toxicology, vol.285, issue.1, pp.30-37, 2017.
DOI : 10.1126/science.285.5430.1028

URL : http://onlinelibrary.wiley.com/doi/10.1111/bcpt.12631/pdf

Y. Nishioka and H. Yoshino, Lymphatic targeting with nanoparticulate system, Advanced Drug Delivery Reviews, vol.47, issue.1, pp.55-64, 2001.
DOI : 10.1016/S0169-409X(00)00121-6

A. Nyström and B. Fadeel, Safety assessment of nanomaterials: Implications for nanomedicine, Journal of Controlled Release, vol.161, issue.2, pp.403-408, 2012.
DOI : 10.1016/j.jconrel.2012.01.027

G. Oberdörster, Safety assessment for nanotechnology and nanomedicine: concepts of nanotoxicology, Journal of Internal Medicine, vol.11, issue.Sl, pp.89-105, 2010.
DOI : 10.1165/ajrcmb.12.6.7539275

G. Oberdörster, V. Stone, and K. Donaldson, Toxicology of nanoparticles: A historical perspective, Nanotoxicology, vol.39, issue.7, pp.2-25, 2007.
DOI : 10.1021/es050265j

O. Günter, O. , and O. Jan, Nanotoxicology: An Emerging Discipline Evolving from Studies of Ultrafine Particles, Environmental Health Perspectives, vol.113, pp.823-839, 2005.

G. Orts-gil, K. Natte, D. Drescher, H. Bresch, A. Mantion et al., Characterisation of silica nanoparticles prior to in vitro studies: from primary particles to agglomerates, Journal of Nanoparticle Research, vol.139, issue.1, pp.1593-1604, 2011.
DOI : 10.1016/0021-9797(90)90441-P

O. Iii, D. Peppas, and N. , Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles, International Journal of Pharmaceutics, vol.307, pp.93-102, 2006.

Y. Paik, S. Jeong, G. Omenn, M. Uhlen, S. Hanash et al., The Chromosome-Centric Human Proteome Project for cataloging proteins encoded in the genome, Nature Biotechnology, vol.9, issue.3, pp.221-223, 2012.
DOI : 10.1083/jcb.201004052

D. Peer, J. Karp, S. Hong, O. Farokhzad, R. Margalit et al., Nanocarriers as an emerging platform for cancer therapy, Nature Nanotechnology, vol.8, issue.12, pp.751-760, 2007.
DOI : 10.1038/bjc.1996.587

M. Peracchia, S. Harnisch, H. Pinto-alphandary, A. Gulik, J. Dedieu et al., Visualization of in vitro protein-rejecting properties of PEGylated stealth?? polycyanoacrylate nanoparticles, Biomaterials, vol.20, issue.14, pp.1269-1275, 1999.
DOI : 10.1016/S0142-9612(99)00021-6

S. Perrault, C. Walkey, T. Jennings, H. Fischer, and W. Chan, Mediating Tumor Targeting Efficiency of Nanoparticles Through Design, Nano Letters, vol.9, issue.5, pp.1909-1915, 2009.
DOI : 10.1021/nl900031y

J. Perry, K. Reuter, M. Kai, K. Herlihy, S. Jones et al., PEGylated PRINT Nanoparticles: The Impact of PEG Density on Protein Binding, Macrophage Association, Biodistribution, and Pharmacokinetics, Nano Letters, vol.12, issue.10, pp.5304-5310, 2012.
DOI : 10.1021/nl302638g

C. Pisani, J. Gaillard, V. Nouvel, M. Odorico, J. Armengaud et al., High-throughput, quantitative assessment of the effects of low-dose silica nanoparticles on lung cells: Grasping complex toxicity with a great depth of field, Toxicology Letters, vol.238, issue.2, p.229, 2015.
DOI : 10.1016/j.toxlet.2015.08.678

M. Powell, M. Newman, and J. Burdman, Vaccine design: the subunit and adjuvant approach, 1995.
DOI : 10.1007/978-1-4615-1823-5

D. Pozzi, V. Colapicchioni, G. Caracciolo, S. Piovesana, A. Capriotti et al., Effect of polyethyleneglycol (PEG) chain length on the bio???nano-interactions between PEGylated lipid nanoparticles and biological fluids: from nanostructure to uptake in cancer cells, Nanoscale, vol.65, issue.5, pp.2782-2792, 2014.
DOI : 10.1016/0022-1759(83)90303-4

P. Pradhan, R. Banerjee, D. Bahadur, C. Koch, O. Mykhaylyk et al., Targeted Magnetic Liposomes Loaded with Doxorubicin, Liposomes: Methods and Protocols, pp.257-272, 2017.

V. Puddu and C. Perry, Peptide Adsorption on Silica Nanoparticles: Evidence of Hydrophobic Interactions, ACS Nano, vol.6, issue.7, pp.6356-63, 2012.
DOI : 10.1021/nn301866q

Y. Qiu, Y. Liu, L. Wang, L. Xu, R. Bai et al., Surface chemistry and aspect ratio mediated cellular uptake of Au nanorods, Biomaterials, vol.31, issue.30, pp.7606-7619, 2010.
DOI : 10.1016/j.biomaterials.2010.06.051

D. Nogueira-librelotto, F. Codevilla, C. Farooqi, A. , and M. B. , Rolim C (2017) Transferrin-Conjugated Nanocarriers as Active-Targeted Drug Delivery Platforms for Cancer Therapy, Current Pharmaceutical Design, vol.23, pp.454-466

T. Ramezanli, Z. Zhang, and B. Michniak-kohn, ????????????????????????Development and characterization of polymeric nanoparticle-based formulation of adapalene for topical acne therapy?????????????????????????????????????????????????????????????????????????????????????????????????????????, Nanomedicine: Nanotechnology, Biology and Medicine, vol.13, issue.1, pp.143-152, 2017.
DOI : 10.1016/j.nano.2016.08.008

S. Raymond, L. Chang, W. Jui-pin, T. Ming-hsien, W. Hsiu-jen et al., Persistent Tissue Kinetics and Redistribution of Nanoparticles, Quantum Dot 705 Mice: ICP-MS Quantitative Assessment, pp.1339-1343, 2007.

M. Reibold, N. Pätzke, A. Levin, W. Kochmann, I. Shakhverdova et al., Structure of several historic blades at nanoscale, Crystal Research and Technology, vol.479, issue.10, pp.1139-1146, 2009.
DOI : 10.2320/matertrans1960.5.115

P. Reuzel, J. Bruijntjes, V. Feron, and R. Woutersen, Subchronic inhalation toxicity of amorphous silicas and quartz dust in rats, Food and Chemical Toxicology, vol.29, issue.5, pp.341-354, 1991.
DOI : 10.1016/0278-6915(91)90205-L

R. Riley and E. Day, Gold nanoparticle-mediated photothermal therapy: applications and opportunities for multimodal cancer treatment, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, vol.5, issue.4863, 2017.
DOI : 10.3390/nano5041853

J. Rojas, H. Gavilán, V. Del-dedo, E. Lorente-sorolla, L. Sanz-ortega et al., Time-course assessment of the aggregation and metabolization of magnetic nanoparticles, Acta Biomaterialia, vol.58, 2017.
DOI : 10.1016/j.actbio.2017.05.047

J. Ross, P. Chaudhuri, and M. Ratnam, Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications, Cancer, vol.51, issue.9, pp.2432-2475, 1994.
DOI : 10.1016/B978-0-12-372180-8.50013-5

B. Rothen-rutishauser, S. Schürch, B. Haenni, N. Kapp, and P. Gehr, Environmental Science & Technology, vol.40, issue.14, pp.4353-4359, 2006.
DOI : 10.1021/es0522635

E. Ruiz-hernández, A. Baeza, and M. Vallet-regí, Smart Drug Delivery through DNA/Magnetic Nanoparticle Gates, ACS Nano, vol.5, issue.2, pp.1259-1266, 2011.
DOI : 10.1021/nn1029229

T. Rushmore, T. Kong, and A. , Pharmacogenomics, Regulation and Signaling Pathways of Phase I and II Drug Metabolizing Enzymes, Current Drug Metabolism, vol.3, issue.5, pp.481-490, 2002.
DOI : 10.2174/1389200023337171

W. Russell and R. Burch, The Principles of Humane Experimental Technique, 1959.

. Saba-n, X. Wang, M. Tighiouart, S. Muller, K. Cho et al., Examining expression of folate receptors in squamous cell carcinoma of the head and neck (SCCHN) as a target for a novel nanotherapeutic drug, Cancer Research, vol.67, pp.174-174, 2007.

U. Sakulkhu, M. Mahmoudi, L. Maurizi, J. Salaklang, and H. Hofmann, Protein Corona Composition of Superparamagnetic Iron Oxide Nanoparticles with Various Physico-Chemical Properties and Coatings, Scientific Reports, vol.139, issue.135, p.5020, 2014.
DOI : 10.1039/c3an02153j

URL : https://hal.archives-ouvertes.fr/hal-02132125

S. Sassa, O. Sugita, R. Galbraith, and A. Kappas, Drug metabolism by the human hepatoma cell, Hep G2, Biochemical and Biophysical Research Communications, vol.143, issue.1, pp.52-57, 1987.
DOI : 10.1016/0006-291X(87)90628-0

M. Schena, D. Shalon, R. Davis, and P. Brown, Quantitative Monitoring of Gene Expression Patterns with a Complementary DNA Microarray, Science, vol.270, issue.5235, pp.467-70, 1995.
DOI : 10.1126/science.270.5235.467

C. Schütz, L. Juillerat-jeanneret, H. Mueller, I. Lynch, and M. Riediker, Therapeutic nanoparticles in clinics and under clinical evaluation, Nanomedicine, vol.3, issue.3, pp.449-467, 2013.
DOI : 10.1016/S0140-6736(10)60357-1

T. Seki, J. Fang, and H. Maeda, Tumor-Targeted Macromolecular Drug Delivery Based on the Enhanced Permeability and Retention Effect in Solid Tumor, Pharmaceutical Perspectives of Cancer, pp.93-120, 2009.
DOI : 10.1007/978-1-4419-0131-6_3

B. Semete, L. Booysen, Y. Lemmer, L. Kalombo, L. Katata et al., In vivo evaluation of the biodistribution and safety of PLGA nanoparticles as drug delivery systems, Nanomedicine: Nanotechnology, Biology and Medicine, vol.6, issue.5, pp.662-671, 2010.
DOI : 10.1016/j.nano.2010.02.002

R. Serda, J. Gu, R. Bhavane, X. Liu, C. Chiappini et al., The association of silicon microparticles with endothelial cells in drug delivery to the vasculature, Biomaterials, vol.30, issue.13, pp.2440-2448, 2009.
DOI : 10.1016/j.biomaterials.2009.01.019

S. Shah, A. Khan, M. Arshad, M. Awan, S. Hashmi et al., Doxorubicin-loaded photosensitive magnetic liposomes for multi-modal cancer therapy, Colloids and Surfaces B: Biointerfaces, vol.148, pp.157-164, 2016.
DOI : 10.1016/j.colsurfb.2016.08.055

D. Shalon, S. Smith, and P. Brown, A DNA microarray system for analyzing complex DNA samples using two-color fluorescent probe hybridization., Genome Research, vol.6, issue.7, pp.639-684, 1996.
DOI : 10.1101/gr.6.7.639

W. Shim, M. Paik, D. Nguyen, J. Lee, Y. Lee et al., Analysis of Changes in Gene Expression and Metabolic Profiles Induced by Silica-Coated Magnetic Nanoparticles, ACS Nano, vol.6, issue.9, pp.7665-80, 2012.
DOI : 10.1021/nn301113f

V. Shubayev, P. Ii, T. , and J. S. , Magnetic nanoparticles for theragnostics, Advanced Drug Delivery Reviews, vol.61, issue.6, pp.467-477, 2009.
DOI : 10.1016/j.addr.2009.03.007

N. Singh, A. Karambelkar, L. Gu, K. Lin, J. Miller et al., Bioresponsive Mesoporous Silica Nanoparticles for Triggered Drug Release, Journal of the American Chemical Society, vol.133, issue.49, pp.19582-19585, 2011.
DOI : 10.1021/ja206998x

R. Singh, M. Norret, M. House, Y. Galabura, M. Bradshaw et al., Dose-Dependent Therapeutic Distinction between Active and Passive Targeting Revealed Using Transferrin-Coated PGMA Nanoparticles, Small, vol.2, issue.3, pp.351-359, 2016.
DOI : 10.1021/mp050032z

S. Singhal, S. Singh, P. Singhal, D. Horne, J. Singhal et al., Antioxidant role of glutathione S-transferases: 4-Hydroxynonenal, a key molecule in stress-mediated signaling, Toxicology and Applied Pharmacology, vol.289, issue.3, pp.361-370, 2015.
DOI : 10.1016/j.taap.2015.10.006

B. Sivakumar, R. Aswathy, R. Romero-aburto, T. Mitcham, K. Mitchel et al., Highly versatile SPION encapsulated PLGA nanoparticles as photothermal ablators of cancer cells and as multimodal imaging agents, Biomaterials Science, vol.107, issue.3, pp.432-443, 2017.
DOI : 10.1021/jp027048e

H. Slanina, A. König, H. Claus, M. Frosch, and A. Schubert-unkmeir, Real-time impedance analysis of host cell response to meningococcal infection, Journal of Microbiological Methods, vol.84, issue.1, pp.101-108, 2011.
DOI : 10.1016/j.mimet.2010.11.004

S. Choi, H. Liu, W. Misra, P. Tanaka, E. Zimmer et al., Renal clearance of quantum dots, Nature Biotechnology, vol.361, issue.10, pp.1165-1170, 2007.
DOI : 10.1152/ajprenal.00316.2002

H. Steen and M. Mann, The abc's (and xyz's) of peptide sequencing, Nature Reviews Molecular Cell Biology, vol.20, issue.Suppl., pp.699-711, 2004.
DOI : 10.1093/bioinformatics/btg484

B. Stella, S. Arpicco, R. F. Marsaud, V. Renoir, J. Cattel et al., Encapsulation of gemcitabine lipophilic derivatives into polycyanoacrylate nanospheres and nanocapsules, International Journal of Pharmaceutics, vol.344, issue.1-2, pp.71-77, 2007.
DOI : 10.1016/j.ijpharm.2007.06.006

J. Stolwijk, K. Matrougui, C. Renken, and M. Trebak, Impedance analysis of GPCR-mediated changes in endothelial barrier function: overview and fundamental considerations for stable and reproducible measurements, Pfl??gers Archiv - European Journal of Physiology, vol.748, issue.6, pp.2193-2218, 2015.
DOI : 10.1111/j.1749-6632.1994.tb17311.x

V. Stone, B. Nowack, A. Baun, N. Van-den-brink, V. Der-kammer et al., Nanomaterials for environmental studies: Classification, reference material issues, and strategies for physico-chemical characterisation, Science of The Total Environment, vol.408, issue.7, pp.1745-1754, 2010.
DOI : 10.1016/j.scitotenv.2009.10.035

K. Strebhardt and A. Ullrich, Paul Ehrlich's magic bullet concept: 100 years of progress, Nature Reviews Cancer, vol.309, issue.6, pp.473-480, 2008.
DOI : 10.1097/00002371-199611000-00006

M. Stubbs, P. Mcsheehy, J. Griffiths, and C. Bashford, Causes and consequences of tumour acidity and implications for treatment, Molecular Medicine Today, vol.6, issue.1, pp.15-19, 2000.
DOI : 10.1016/S1357-4310(99)01615-9

I. Szleifer, Protein Adsorption on Surfaces with Grafted Polymers, Biophysical Journal, vol.72, issue.2, pp.595-612, 1997.
DOI : 10.1016/S0006-3495(97)78698-3

K. Takahashi, K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka et al., Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors, Cell, vol.131, issue.5, pp.861-872, 2007.
DOI : 10.1016/j.cell.2007.11.019

J. Teeguarden, P. Hinderliter, G. Orr, B. Thrall, and J. Pounds, Particokinetics In Vitro: Dosimetry Considerations for In Vitro Nanoparticle Toxicity Assessments, Toxicological Sciences, vol.34, issue.2, pp.300-312, 2007.
DOI : 10.1111/j.1423-0410.1978.tb02890.x

S. Tenzer, D. Docter, J. Kuharev, A. Musyanovych, V. Fetz et al., Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology, Nature Nanotechnology, vol.12, issue.10, pp.772-781, 2013.
DOI : 10.1021/pr400193j

S. Tenzer, D. Docter, S. Rosfa, A. Wlodarski, J. Kuharev et al., Nanoparticle Size Is a Critical Physicochemical Determinant of the Human Blood Plasma Corona: A Comprehensive Quantitative Proteomic Analysis, ACS Nano, vol.5, issue.9, pp.7155-67, 2011.
DOI : 10.1021/nn201950e

L. Thomassen, A. Aerts, V. Rabolli, D. Lison, L. Gonzalez et al., Cytotoxicity Testing, Langmuir, vol.26, issue.1, pp.328-335, 2010.
DOI : 10.1021/la902050k

K. Tsoi, S. Macparland, X. Ma, V. Spetzler, J. Echeverri et al., Mechanism of hard-nanomaterial clearance by the??liver, Nature Materials, vol.205, issue.11, pp.1212-1221, 2016.
DOI : 10.1084/jem.20080108

R. Tukey and C. Strassburg, Human UDP-Glucuronosyltransferases: Metabolism, Expression, and Disease, Annual Review of Pharmacology and Toxicology, vol.40, issue.1, pp.581-616, 2000.
DOI : 10.1146/annurev.pharmtox.40.1.581

L. Unsworth, H. Sheardown, and J. Brash, Protein-Resistant Poly(ethylene oxide)-Grafted Surfaces:?? Chain Density-Dependent Multiple Mechanisms of Action, Langmuir, vol.24, issue.5, pp.1924-1929, 2008.
DOI : 10.1021/la702310t

M. Uz and V. Bulmus, Alsoy Altinkaya S (2016) Effect of PEG Grafting Density and Hydrodynamic Volume on Gold Nanoparticle?Cell Interactions: An Investigation on Cell Cycle, Apoptosis, and DNA Damage Glutathione conjugation as a bioactivation reaction, Langmuir van Bladeren PJ Chemico-Biological Interactions, vol.129, pp.61-76, 2000.

P. Van-pham, N. Nguyen, H. Nguyen, L. Khuat, P. Le et al., A simple in vitro method for evaluating dendritic cell-based vaccinations, OncoTargets & Therapy, vol.7, 2014.

J. Venter, M. Adams, E. Myers, P. Li, R. Mural et al., The Sequence of the Human Genome, Science, vol.6, issue.5507, pp.1304-1351, 2001.
DOI : 10.1093/nar/28.1.33

URL : https://hal.archives-ouvertes.fr/hal-00465088

M. Videira, M. Botelho, A. Santos, L. Gouveia, P. De-lima et al., Lymphatic Uptake of Pulmonary Delivered Radiolabelled Solid Lipid Nanoparticles, Journal of Drug Targeting, vol.10, issue.8, pp.607-613, 2002.
DOI : 10.1080/1061186021000054933

M. Vinken, The adverse outcome pathway concept: A pragmatic tool in toxicology, Toxicology, vol.312, pp.158-165, 2013.
DOI : 10.1016/j.tox.2013.08.011

M. Vinken, B. Landesmann, M. Goumenou, S. Vinken, I. Shah et al., Development of an Adverse Outcome Pathway From Drug-Mediated Bile Salt Export Pump Inhibition to Cholestatic Liver Injury, Toxicological Sciences, vol.126, issue.1, pp.97-106, 2013.
DOI : 10.1016/j.pharmthera.2010.03.005

. Wahajuddin and S. Arora, Superparamagnetic iron oxide nanoparticles: magnetic nanoplatforms as drug carriers, International Journal of Nanomedicine, vol.7, pp.3445-3471, 2012.
DOI : 10.2147/IJN.S30320

D. Walczyk, F. Bombelli, M. Monopoli, I. Lynch, and K. Dawson, What the Cell ???Sees??? in Bionanoscience, Journal of the American Chemical Society, vol.132, issue.16, pp.5761-5768, 2010.
DOI : 10.1021/ja910675v

C. Walkey and W. Chan, Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment, Chem. Soc. Rev., vol.14, issue.135, pp.2780-99, 2012.
DOI : 10.1002/elps.11501401214

Y. Wang and H. Gu, Core-Shell-Type Magnetic Mesoporous Silica Nanocomposites for Bioimaging and Therapeutic Agent Delivery, Advanced Materials, vol.22, issue.3, pp.576-585, 2015.
DOI : 10.1039/c2jm34364a

Y. Wang, Q. Zhao, N. Han, L. Bai, J. Li et al., Mesoporous silica nanoparticles in drug delivery and biomedical applications, Nanomedicine: Nanotechnology, Biology and Medicine, vol.11, issue.2, pp.313-327, 2015.
DOI : 10.1016/j.nano.2014.09.014

M. Waters and J. Fostel, Toxicogenomics and systems toxicology: aims and prospects, Nature Reviews Genetics, vol.22, issue.182, pp.936-948, 2004.
DOI : 10.1093/bioinformatics/bth130

W. Westerink and W. Schoonen, Phase II enzyme levels in HepG2 cells and cryopreserved primary human hepatocytes and their induction in HepG2 cells, Toxicology in Vitro, vol.21, issue.8, pp.1592-1602, 2007.
DOI : 10.1016/j.tiv.2007.06.017

P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Gellermann et al., Hyperthermia in combined treatment of cancer, The Lancet Oncology, vol.3, issue.8, pp.487-497, 2002.
DOI : 10.1016/S1470-2045(02)00818-5

B. Xi, T. Wang, N. Li, W. Ouyang, W. Zhang et al., Functional Cardiotoxicity Profiling and Screening Using the xCELLigence RTCA Cardio System, Journal of Laboratory Automation, vol.13, issue.6, pp.415-421, 2011.
DOI : 10.1016/j.drudis.2008.06.011

Z. Xi, S. Khare, A. Cheung, B. Huang, T. Pan et al., Mode of action classification of chemicals using multi-concentration time-dependent cellular response profiles, Computational Biology and Chemistry, vol.49, pp.23-35, 2014.
DOI : 10.1016/j.compbiolchem.2013.12.004

X. Xie, F. Li, H. Zhang, Y. Lu, S. Lian et al., EpCAM aptamer-functionalized mesoporous silica nanoparticles for efficient colon cancer cell-targeted drug delivery, European Journal of Pharmaceutical Sciences, vol.83, pp.28-35, 2016.
DOI : 10.1016/j.ejps.2015.12.014

J. Xu, D. Diaz, and P. O-'brien, Applications of cytotoxicity assays and pre-lethal mechanistic assays for assessment of human hepatotoxicity potential, Chemico-Biological Interactions, vol.150, issue.1, pp.115-128, 2004.
DOI : 10.1016/j.cbi.2004.09.011

H. Yang, Y. Chen, Z. Chen, Y. Geng, X. Xie et al., Chemo-photodynamic combined gene therapy and dual-modal cancer imaging achieved by pH-responsive alginate/chitosan multilayer-modified magnetic mesoporous silica nanocomposites, Biomaterials Science, vol.11, issue.5, pp.1001-1013, 2017.
DOI : 10.1002/smll.201500856

H. Yang, Y. Zhuang, Y. Sun, A. Dai, X. Shi et al., Targeted dual-contrast T1- and T2-weighted magnetic resonance imaging of tumors using multifunctional gadolinium-labeled superparamagnetic iron oxide nanoparticles, Biomaterials, vol.32, issue.20, pp.4584-4593, 2011.
DOI : 10.1016/j.biomaterials.2011.03.018

L. Yang, F. Cui, D. Cun, A. Tao, K. Shi et al., Preparation, characterization and biodistribution of the lactone form of 10-hydroxycamptothecin (HCPT)-loaded bovine serum albumin (BSA) nanoparticles, International Journal of Pharmaceutics, vol.340, issue.1-2, pp.163-172, 2007.
DOI : 10.1016/j.ijpharm.2007.03.028

W. Yang, J. Peters, and R. Williams, Inhaled nanoparticles???A current review, International Journal of Pharmaceutics, vol.356, issue.1-2, pp.239-247, 2008.
DOI : 10.1016/j.ijpharm.2008.02.011

Y. Yim, J. Choi, S. Jang, G. Kim, K. Park et al., Pharmacokinetic properties and tissue storage of FITC conjugated SA-MnMEIO nanoparticles in mice, Current Applied Physics, vol.9, issue.4, pp.304-307, 2009.
DOI : 10.1016/j.cap.2009.06.033

U. Zanger and M. Schwab, Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation, Pharmacology & Therapeutics, vol.138, issue.1, pp.103-141, 2013.
DOI : 10.1016/j.pharmthera.2012.12.007

J. Zhang, X. Li, J. Rosenholm, and H. Gu, Synthesis and characterization of pore size-tunable magnetic mesoporous silica nanoparticles, Journal of Colloid and Interface Science, vol.361, issue.1, pp.16-24, 2011.
DOI : 10.1016/j.jcis.2011.05.038

J. Zhang, W. Sun, L. Bergman, J. Rosenholm, M. Lindén et al., Magnetic mesoporous silica nanospheres as DNA/drug carrier, Materials Letters, vol.67, issue.1, pp.379-382, 2012.
DOI : 10.1016/j.matlet.2011.09.086

L. Zhang, H. Gao, L. Chen, B. Wu, Y. Zheng et al., Tumor targeting of vincristine by mBAFF-modified PEG liposomes in B lymphoma cells, Cancer Letters, vol.269, issue.1, pp.26-36, 2008.
DOI : 10.1016/j.canlet.2008.04.024

X. Zhao, Q. Chen, W. Liu, Y. Li, H. Tang et al., Codelivery of doxorubicin and curcumin with lipid nanoparticles results in improved efficacy of chemotherapy in liver cancer, International journal of nanomedicine, vol.10, pp.257-70, 2015.

Y. Zhu, W. Li, Q. Li, Y. Li, Y. Li et al., Effects of serum proteins on intracellular uptake and cytotoxicity of carbon nanoparticles, Carbon, vol.47, issue.5, pp.1351-1358, 2009.
DOI : 10.1016/j.carbon.2009.01.026

Z. Zhu, T. Posati, D. Moyano, R. Tang, B. Yan et al., The Interplay of Monolayer Structure and Serum Protein Interactions on the Cellular Uptake of Gold Nanoparticles, Small, vol.46, issue.135, pp.2659-2663, 2012.
DOI : 10.1002/anie.200700465