.. .. Greffage,

. Nanoparticulaires-coeur/coquille-modifie-par-l'af-charges and . .. En-dox,

S. Bhide and C. Nutting, Recent Advances in Radiotherapy, BMC Med, vol.8, issue.1, 2010.

,

K. Moghissi, R. J. Parsons, and K. Dixon, Photodynamic Therapy (PDT) for Bronchial Carcinoma with the Use of Rigid Bronchoscope, Lasers Med. Sci, vol.1992, issue.1-4, pp.381-385

T. J. Dougherty, C. J. Gomer, B. W. Henderson, G. Jori, D. Kessel et al., Photodynamic Therapy. J. Natl. Cancer Inst, vol.90, issue.12, pp.889-905, 1998.

N. P. Brodin, C. Guha, and W. A. Tomé, Photodynamic Therapy and Its Role in Combined Modality Anticancer Treatment, Technol. Cancer Res. Treat, vol.14, issue.4, pp.355-368, 2015.

X. Liang, C. Chen, Y. Zhao, P. C. Wang, J. Zhou et al., Circumventing Tumor Resistance to Chemotherapy by Nanotechnology. In Multi-Drug Resistance in Cancer, vol.596, pp.467-488, 2010.

M. Arruebo, N. Vilaboa, B. Sáez-gutierrez, J. Lambea, A. Tres et al., Assessment of the Evolution of Cancer Treatment Therapies, Cancers, vol.2011, issue.3, pp.3279-3330

,

A. Coates, S. Abraham, S. B. Kaye, T. Sowerbutts, C. Frewin et al., On the Receiving End-Patient Perception of the Side-Effects of Cancer Chemotherapy, Eur. J. Cancer Clin. Oncol, vol.19, issue.2, pp.203-208, 1983.

,

. Maruyama, . Ishida, . Takizawa, and . Moribe, Possibility of Active Targeting to Tumor Tissues with Liposomes, Adv. Drug Deliv. Rev, vol.40, issue.1-2, pp.89-102, 1999.

P. Couvreur and C. Vauthier, Nanotechnology: Intelligent Design to Treat Complex Disease, Pharm. Res, vol.23, issue.7, pp.1417-1450, 2006.

M. J. Embleton, G. F. Rowland, R. G. Simmonds, E. Jacobs, C. H. Marsden et al., Selective Cytotoxicity against Human Tumour Cells by a Vindesine-Monoclonal Antibody Conjugate, Br. J. Cancer, vol.47, issue.1, pp.43-49, 1983.

,

K. Tamarov, S. Näkki, W. Xu, and V. Lehto, Approaches to Improve the Biocompatibility and Systemic Circulation of Inorganic Porous Nanoparticles, J. Mater. Chem. B, vol.2018, issue.22, pp.3632-3649

M. Mir, S. Ishtiaq, S. Rabia, M. Khatoon, A. Zeb et al., Nanotechnology: From In Vivo Imaging System to Controlled Drug Delivery, Nanoscale Res. Lett, vol.2017, issue.1, p.12

,

J. Lin, Y. Li, Y. Li, H. Wu, F. Yu et al., Drug/Dye-Loaded, Multifunctional PEG-Chitosan-Iron Oxide Nanocomposites for Methotraxate Synergistically Self-Targeted Cancer Therapy and Dual Model Imaging, ACS Appl. Mater. Interfaces, vol.7, issue.22, pp.11908-11920, 2015.

,

H. Maeda, J. Wu, T. Sawa, Y. Matsumura, and K. Hori, Tumor Vascular Permeability and the EPR Effect in Macromolecular Therapeutics: A Review, J. Control. Release Off. J. Control. Release Soc, vol.65, issue.1-2, pp.271-284, 2000.

, , pp.248-253

Y. Zhao, B. G. Trewyn, I. I. Slowing, V. S. Lin, and .. , Mesoporous Silica Nanoparticle-Based Double Drug Delivery System for Glucose-Responsive Controlled Release of Insulin and Cyclic AMP, J. Am. Chem. Soc, vol.131, issue.24, pp.8398-8400, 2009.

,

A. K. Iyer, G. Khaled, J. Fang, and H. Maeda, Exploiting the Enhanced Permeability and Retention Effect for Tumor Targeting, Drug Discov. Today, vol.11, pp.812-818, 2006.

N. Hoshyar, S. Gray, H. Han, and G. Bao, The Effect of Nanoparticle Size on in Vivo Pharmacokinetics and Cellular Interaction, Nanomed, vol.2016, issue.6, pp.673-692

K. Iwai, H. Maeda, and T. Konno, Use of Oily Contrast Medium for Selective Drug Targeting to Tumor: Enhanced Therapeutic Effect and X-Ray Image, Cancer Res, vol.44, issue.5, pp.2115-2121, 1984.

A. , F. Pridgen, E. Molnar, L. K. Farokhzad, and O. C. , Factors Affecting the Clearance and Biodistribution of Polymeric Nanoparticles, Mol. Pharm, vol.5, issue.4, pp.505-515, 2008.

E. Ruoslahti, S. N. Bhatia, and M. J. Sailor, Targeting of Drugs and Nanoparticles to Tumors, J. Cell Biol, issue.6, pp.759-768, 2010.

,

J. Jiang, S. Yang, J. Wang, L. Yang, Z. Xu et al., Sequential Treatment of Drug-Resistant Tumors with RGD-Modified Liposomes Containing SiRNA or Doxorubicin, Eur. J. Pharm. Biopharm, vol.76, issue.2, pp.170-178, 2010.

,

G. D. Bothun, A. Lelis, Y. Chen, K. Scully, L. E. Anderson et al., Multicomponent Folate-Targeted Magnetoliposomes: Design, Characterization, and Cellular Uptake, Nanomedicine Nanotechnol. Biol. Med, vol.7, issue.6, pp.797-805, 2011.

A. Etzerodt, M. B. Maniecki, J. H. Graversen, H. J. Møller, V. P. Torchilin et al., Efficient Intracellular Drug-Targeting of Macrophages Using Stealth Liposomes Directed to the Hemoglobin Scavenger Receptor CD163, J. Controlled Release, vol.2012, issue.1, pp.72-80

R. E. Eliaz, S. Nir, C. Marty, and F. C. Szoka, Determination and Modeling of Kinetics of Cancer Cell Killing by Doxorubicin and Doxorubicin Encapsulated in Targeted Liposomes, Cancer Res, vol.64, issue.2, pp.711-718, 2004.

S. Wagner, F. Rothweiler, M. G. Anhorn, D. Sauer, I. Riemann et al., Enhanced Drug Targeting by Attachment of an Anti ?v Integrin Antibody to Doxorubicin Loaded Human Serum Albumin Nanoparticles, Biomaterials, vol.2010, issue.8, pp.2388-2398

,

A. Lowery, H. Onishko, D. E. Hallahan, and Z. Han, Tumor-Targeted Delivery of Liposome-Encapsulated Doxorubicin by Use of a Peptide That Selectively Binds to Irradiated Tumors, J. Controlled Release, vol.150, issue.1, pp.117-124, 2011.

,

G. Mani, S. Kim, and K. Kim, Development of Folate-Thioglycolate-Gold Nanoconjugates by Using Citric Acid-PEG Branched Polymer for Inhibition of MCF-7 Cancer Cell Proliferation, Biomacromolecules, vol.19, issue.8, pp.3257-3267, 2018.

,

D. Luong, S. Sau, P. Kesharwani, and A. K. Iyer, Polyvalent Folate-Dendrimer-Coated Iron Oxide Theranostic Nanoparticles for Simultaneous Magnetic Resonance Imaging and Precise Cancer Cell Targeting, Biomacromolecules, vol.2017, issue.4, pp.1197-1209

,

K. Siwowska, R. Schmid, S. Cohrs, R. Schibli, and C. Müller, Folate Receptor-Positive Gynecological Cancer Cells: In Vitro and In Vivo Characterization, Pharmaceuticals, vol.2017, issue.4

J. Xu, B. Xu, D. Shou, F. Qin, Y. Xu et al., Characterization and Evaluation of a Folic Acid Receptor-Targeted Cyclodextrin Complex as an Anticancer Drug Delivery System, Eur. J. Pharm. Sci, vol.83, pp.132-142, 2016.

,

J. Lin, Y. Li, Y. Li, H. Wu, F. Yu et al., Drug/Dye-Loaded, Multifunctional PEG-Chitosan-Iron Oxide Nanocomposites for Methotraxate Synergistically Self-Targeted Cancer Therapy and Dual Model Imaging, ACS Appl. Mater. Interfaces, vol.7, issue.22, pp.11908-11920, 2015.

,

J. P. May and S. Li, Hyperthermia-Induced Drug Targeting. Expert Opin. Drug Deliv, vol.10, issue.4, pp.511-527, 2013.

,

J. A. Vassie, J. M. Whitelock, and M. S. Lord, Targeted Delivery and Redox Activity of Folic Acid-Functionalized Nanoceria in Tumor Cells, Mol. Pharm, vol.15, issue.3, pp.994-1004, 2018.

L. E. Woodard, C. L. Dennis, J. A. Borchers, A. Attaluri, E. Velarde et al., Nanoparticle Architecture Preserves Magnetic Properties during Coating to Enable Robust Multi-Modal Functionality, Sci. Rep, vol.8, issue.1, 2018.

A. Kumar, H. Ma, X. Zhang, K. Huang, S. Jin et al., Gold Nanoparticles Functionalized with Therapeutic and Targeted Peptides for Cancer -39 -Treatment, Biomaterials, vol.2012, issue.4, pp.1180-1189

,

F. Porta, G. E. Lamers, J. Morrhayim, A. Chatzopoulou, M. Schaaf et al., Folic Acid-Modified Mesoporous Silica Nanoparticles for Cellular and Nuclear Targeted Drug Delivery, Adv. Healthc. Mater, vol.2013, issue.2, pp.281-286

D. Pissuwan, C. H. Cortie, S. M. Valenzuela, and M. B. Cortie, Functionalised Gold Nanoparticles for Controlling Pathogenic Bacteria, Trends Biotechnol, vol.28, issue.4, pp.207-213, 2010.

J. Zhou, J. Ralston, R. Sedev, and D. A. Beattie, Functionalized Gold Nanoparticles: Synthesis, Structure and Colloid Stability, J. Colloid Interface Sci, vol.331, issue.2, pp.251-262, 2009.

E. Amstad, M. Textor, and E. Reimhult, Stabilization and Functionalization of Iron Oxide Nanoparticles for Biomedical Applications, Nanoscale, vol.2011, issue.7, p.2819

R. Hao, R. Xing, Z. Xu, Y. Hou, S. Gao et al., Synthesis, Functionalization, and Biomedical Applications of Multifunctional Magnetic Nanoparticles, Adv. Mater, issue.25, pp.2729-2742, 2010.

,

A. Ito, M. Shinkai, H. Honda, and T. Kobayashi, Medical Application of Functionalized Magnetic Nanoparticles, J. Biosci. Bioeng, vol.100, issue.1, pp.1-11, 2005.

,

B. G. Trewyn, I. I. Slowing, S. Giri, H. Chen, V. S. Lin et al., Synthesis and Functionalization of a Mesoporous Silica Nanoparticle Based on the Sol-Gel Process and Applications in Controlled Release, Acc. Chem. Res, vol.40, issue.9, pp.846-853, 2007.

L. Feng, C. Zhu, H. Yuan, L. Liu, F. Lv et al., Polymer Nanoparticles: Preparation, Properties, Functionalization and Biological Applications, Chem. Soc. Rev, vol.42, issue.16, p.6620, 2013.

N. D. Thorat, R. A. Bohara, M. R. Noor, D. Dhamecha, T. Soulimane et al., Effective Cancer Theranostics with Polymer Encapsulated Superparamagnetic Nanoparticles: Combined Effects of Magnetic Hyperthermia and Controlled Drug Release, ACS Biomater. Sci. Eng, vol.2017, issue.7, pp.1332-1340

,

P. Mohan and N. Rapoport, Doxorubicin as a Molecular Nanotheranostic Agent: Effect of Doxorubicin Encapsulation in Micelles or Nanoemulsions on the Ultrasound-Mediated Intracellular Delivery and Nuclear Trafficking, Mol. Pharm, vol.7, issue.6, pp.1959-1973, 2010.

B. T. Mai, P. B. Balakrishnan, M. J. Barthel, F. Piccardi, D. Niculaes et al., Thermoresponsive Iron Oxide Nanocubes for an Effective Clinical Translation of Magnetic Hyperthermia and Heat-Mediated Chemotherapy, ACS Appl. Mater. Interfaces, vol.11, issue.6, pp.5727-5739, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02415100

D. Luong, S. Sau, P. Kesharwani, and A. K. Iyer, Polyvalent Folate-Dendrimer-Coated Iron Oxide Theranostic Nanoparticles for Simultaneous Magnetic Resonance Imaging and Precise Cancer Cell Targeting, Biomacromolecules, vol.2017, issue.4, pp.1197-1209

, , p.43

Y. Maeda, T. Kubota, H. Yamauchi, T. Nakaji, and H. Kitano, Hydration Changes of Poly(2-(2-Methoxyethoxy)Ethyl Methacrylate) during Thermosensitive Phase Separation in Water, Langmuir, vol.23, issue.22, pp.11259-11265, 2007.

,

T. Ishizone, A. Seki, M. Hagiwara, S. Han, H. Yokoyama et al., Anionic Polymerizations of Oligo(Ethylene Glycol) Alkyl Ether Methacrylates: Effect of Side Chain Length and ?-Alkyl Group of Side Chain on Cloud Point in Water, Macromolecules, issue.8, pp.2963-2967, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00340433

,

J. Lutz, Ö. Akdemir, and A. Hoth, Point by Point Comparison of Two Thermosensitive Polymers Exhibiting a Similar LCST: Is the Age of Poly(NIPAM) Over?, J. Am. Chem. Soc, vol.128, issue.40, pp.13046-13047, 2006.

,

H. Y. Liu and X. X. Zhu, Lower Critical Solution Temperatures of N -Substituted Acrylamide Copolymers in Aqueous Solutions, Polymer, vol.40, issue.25, pp.858-859, 1999.

J. Seuring, F. M. Bayer, K. Huber, and S. Agarwal, Upper Critical Solution Temperature of Poly( N -Acryloyl Glycinamide) in Water: A Concealed Property, Macromolecules, vol.2012, issue.1, pp.374-384

,

B. Pelaz, P. Del-pino, P. Maffre, R. Hartmann, M. Gallego et al., Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake, ACS Nano, vol.9, issue.7, pp.6996-7008, 2015.

,

K. Maier-hauff, F. Ulrich, D. Nestler, H. Niehoff, P. Wust et al., Efficacy and Safety of Intratumoral Thermotherapy Using Magnetic Iron-Oxide Nanoparticles Combined with External Beam Radiotherapy on Patients with Recurrent Glioblastoma Multiforme, J. Neurooncol, vol.103, issue.2, pp.317-324, 2011.

P. Masson, G. Beinert, E. Franta, and P. Rempp, Synthesis of Polyethylene Oxide Macromers, Polym. Bull, vol.7, issue.1, 1982.

,

K. Ito, H. Tsuchida, A. Hayashi, T. Kitano, E. Yamada et al., Reactivity of Poly(Ethylene Oxide) Macromonomers in Radical Copolymerization, Polym. J, vol.17, p.827, 1985.

J. Lutz, Polymerization of Oligo(Ethylene Glycol) (Meth)Acrylates: Toward New Generations of Smart Biocompatible Materials, J. Polym. Sci. Part Polym. Chem, vol.46, issue.11, pp.3459-3470, 2008.

,

H. Lee, J. Pietrasik, and K. Matyjaszewski, Phototunable Temperature-Responsive Molecular Brushes Prepared by ATRP, Macromolecules, vol.39, issue.11, pp.3914-3920, 2006.

F. J. Xu and W. T. Yang, Polymer Vectors via Controlled/Living Radical Polymerization for Gene Delivery, Prog. Polym. Sci, vol.36, issue.9, pp.1099-1131, 2011.

,

M. S. Reeve, S. P. Mccarthy, M. J. Downey, R. A. Gross, and . Stereochemistry, Effect on Enzymic Degradability. Macromolecules, vol.27, pp.825-831, 1994.

H. C. Kolb, M. G. Finn, and K. B. Sharpless, Click Chemistry: Diverse Chemical Function from a Few Good Reactions, Angew. Chem. Int. Ed Engl, vol.40, issue.11, pp.2004-2021, 2001.

J. Israelachvili, The Different Faces of Poly(Ethylene Glycol), Proc. Natl. Acad. Sci, vol.94, pp.8378-8379, 1997.

,

J. Lutz, N. Jahed, and K. Matyjaszewski, Preparation and Characterization of Graft Terpolymers with Controlled Molecular Structure, J. Polym. Sci. Part Polym. Chem, issue.8, pp.1939-1952, 2004.

,

S. Han, M. Hagiwara, and T. Ishizone, Synthesis of Thermally Sensitive Water-Soluble Polymethacrylates by Living Anionic Polymerizations of Oligo(Ethylene Glycol) Methyl Ether Methacrylates, Macromolecules, vol.36, issue.22, pp.8312-8319, 2003.

,

J. Lutz, Polymerization of Oligo(Ethylene Glycol) (Meth)Acrylates: Toward New Generations of Smart Biocompatible Materials, J. Polym. Sci. Part Polym. Chem, vol.46, issue.11, pp.3459-3470, 2008.

,

S. Minko, Grafting to" and "Grafting from, Methods. In Polymer Surfaces and Interfaces

M. Stamm and . Ed, , pp.215-234, 2008.

,

, Polymer Surfaces and Interfaces

M. Stamm and . Ed, , 2008.

Z. Zhang, A. E. Berns, S. Willbold, and J. Buitenhuis, Synthesis of Poly(Ethylene Glycol) (PEG)-Grafted Colloidal Silica Particles with Improved Stability in Aqueous Solvents, J. Colloid Interface Sci, vol.310, issue.2, pp.446-455, 2007.

R. Matsuno, K. Yamamoto, H. Otsuka, and A. Takahara, Polystyrene-and Poly(3-Vinylpyridine)-Grafted Magnetite Nanoparticles Prepared through Surface-Initiated Nitroxide-Mediated Radical Polymerization, Macromolecules, vol.37, issue.6, pp.2203-2209, 2004.

I. Garcia, A. Tercjak, N. E. Zafeiropoulos, M. Stamm, and I. Mondragon, Generation of Core/Shell Iron Oxide Magnetic Nanoparticles with Polystyrene Brushes by Atom Transfer Radical Polymerization, J. Polym. Sci. Part Polym. Chem, issue.20, pp.4744-4750, 2007.

P. Liu, W. Zhong, H. Shi, and D. Xue, Polymer-Grafted Magnetite Nanoparticles via a Facile in Situ Solution Radical Polymerisation, J. Exp. Nanosci, vol.4, issue.4, pp.323-329, 2009.

T. Pintauer and K. Matyjaszewski, Atom Transfer Radical Addition and Polymerization Reactions Catalyzed by Ppm Amounts of Copper Complexes, Chem. Soc. Rev, vol.37, issue.6, p.1087, 2008.

N. V. Tsarevsky and K. Matyjaszewski, Atom Transfer Radical Polymerization: From Process Design to Preparation of Well-Defined Environmentally Friendly Polymeric Materials, Chem. Rev, vol.107, issue.6, pp.2270-2299, 2007.

,

W. Jakubowski and K. Matyjaszewski, Activator Generated by Electron Transfer for Atom Transfer Radical Polymerization, Macromolecules, vol.38, issue.10, pp.4139-4146, 2005.

B. T. Mai, P. B. Balakrishnan, M. J. Barthel, F. Piccardi, D. Niculaes et al., Thermoresponsive Iron Oxide Nanocubes for an Effective Clinical Translation of Magnetic Hyperthermia and Heat-Mediated Chemotherapy, ACS Appl. Mater. Interfaces, vol.11, issue.6, pp.5727-5739, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02415100

J. Al-dine, E. Ferjaoui, Z. Ghanbaja, J. Roques-carmes, T. Meftah et al., Thermo-Responsive Magnetic Fe 3 O 4 @P(MEO 2 MA X -OEGMA 100-X ) NPs and Their Applications as Drug Delivery Systems, Int. J. Pharm, vol.2017, issue.2, pp.738-747
URL : https://hal.archives-ouvertes.fr/hal-01650405

,

J. Al-dine, E. Ferjaoui, Z. Ghanbaja, J. Roques-carmes, T. Meftah et al., Thermo-Responsive Magnetic Fe3O4@P(MEO2MAX-OEGMA100-X) NPs and Their Applications as Drug Delivery Systems, Int. J. Pharm, vol.2017, issue.2, pp.738-747
URL : https://hal.archives-ouvertes.fr/hal-01650405

J. Lutz, . Polymerization, and . Oligo, Ethylene Glycol) (Meth)Acrylates: Toward New Generations of Smart Biocompatible Materials. J. Polym. Sci. Part Polym. Chem, vol.46, issue.11, pp.3459-3470, 2008.

X. Gao, N. Ku?erka, M. Nieh, J. Katsaras, S. Zhu et al., Chain Conformation of a New Class of PEG-Based Thermoresponsive Polymer Brushes Grafted on Silicon as Determined by Neutron Reflectometry, Langmuir, vol.25, issue.17, pp.10271-10278, 2009.

N. Nath, A. Chilkoti, and . Creating, Smart" Surfaces Using Stimuli Responsive Polymers, Adv. Mater, vol.14, issue.17, pp.1243-1247, 2002.

D. Roy, W. L. Brooks, and B. S. Sumerlin, New Directions in Thermoresponsive Polymers, Chem. Soc. Rev, vol.42, issue.17, pp.7214-7243, 2013.

,

Z. Ferjaoui, R. Schneider, A. Meftah, E. Gaffet, and H. Alem, Functional Responsive Superparamagnetic Core/Shell Nanoparticles and Their Drug Release Properties, vol.7, pp.26243-26249, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01523829

E. J. Dine, Z. Ferjaoui, T. Roques-carmes, A. Schjen, and A. Meftah,

J. Toufaily, R. Schneider, E. Gaffet, and H. Alem, Efficient Synthetic Access to Thermo-Responsive Core/Shell Nanoparticles, Nanotechnology, vol.2017, issue.12, p.125601
URL : https://hal.archives-ouvertes.fr/hal-01461773

,

N. P. Mortensen, G. B. Hurst, W. Wang, C. M. Foster, P. D. Nallathamby et al., Dynamic Development of the Protein Corona on Silica Nanoparticles: Composition and Role in Toxicity, Nanoscale, vol.2013, issue.14, p.6372

W. Yeeling, B. Sonali, ?. B. Nata?a, and P. C. Massimilian, Interpreting non-linear drug diffusion data: Utilizing Korsmeyer-Peppas model to study drug release from liposomes, European Journal of Pharmaceutical Sciences, vol.138, 2019.

K. Duygu, K. Kurtul, and A. Neslihan, Modeling the drug release from reduced graphene oxide reinforced hyaluronic acid/gelatin/poly (ethylene oxide) polymeric films. Carbohydrate Polymers, vol.215, pp.189-197, 2019.

J. F. Martínez-lópez, S. Schneider, D. Salavera, A. M. Mainar, J. S. Urieta et al., Molar Heat Capacities of the Mixture {1,8-Cineole + Ethanol} at Several Temperatures and Atmospheric Pressure, J. Chem. Thermodyn, vol.92, pp.146-151, 2016.

,

E. Abumandour, F. Mutelet, and D. Alonso, Performance of an Absorption Heat Transformer Using New Working Binary Systems Composed of {ionic Liquid and Water}, Appl. Therm. Eng, vol.94, pp.579-589, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01934461

S. Shen, Y. Wu, Y. Liu, and D. Wu, High Drug-Loading Nanomedicines: Progress, Current Status, and Prospects, Int. J. Nanomedicine, vol.12, pp.4085-4109, 2017.

S. Mura, J. Nicolas, and P. Couvreur, Stimuli-Responsive Nanocarriers for Drug Delivery, Nat. Mater, vol.12, issue.11, pp.991-1003, 2013.

N. Andhariya, B. Chudasama, R. V. Mehta, and R. V. Upadhyay, Biodegradable Thermoresponsive Polymeric Magnetic Nanoparticles: A New Drug Delivery Platform for Doxorubicin, J Nanopart Res, vol.13, issue.4, pp.1677-1688, 2011.

B. T. Mai, P. B. Balakrishnan, M. J. Barthel, F. Piccardi, D. Niculaes et al., Thermoresponsive Iron Oxide Nanocubes for an Effective Clinical Translation of Magnetic Hyperthermia and Heat-Mediated Chemotherapy, ACS Applied Materials & Interfaces, vol.11, issue.6, pp.5727-5739, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02415100

E. A. Périgo, G. Hemery, O. Sandre, D. Ortega, E. Garaio et al., Fundamentals and Advances in Magnetic Hyperthermia, Appl. Phys. Rev, vol.2015, issue.4

K. Inder, E. Laura-jayne, R. Isabella, T. Ratna, C. Marie et al., Dispersion of Nanomaterials in Aqueous Media/ Towards Protocol Optimization, Journal of Visualized Experiments, issue.130, p.56074, 2017.

Y. Chen, K. Ai, J. Liu, G. Sun, Q. Yin et al., Multifunctional Envelope-Type Mesoporous Silica Nanoparticles for PH-Responsive Drug Delivery and Magnetic Resonance Imaging, Biomaterials, vol.60, pp.111-120, 2015.

J. Xu, B. Xu, D. Shou, F. Qin, Y. Xu et al., Characterization and Evaluation of a Folic Acid Receptor-Targeted Cyclodextrin Complex as an Anticancer Drug Delivery System, Eur. J. Pharm. Sci, vol.83, pp.132-142, 2016.

C. S. Herrington, E. Robert, R. H. Scully, P. B. Young, and . Clement, Armed Forces Institute of Pathology, Tumors of the Ovary, Maldeveloped Gonads, Fallopian Tube and Broad Ligament, vol.189, pp.145-145, 1998.

W. Zoli, P. Ulivi, A. Tesei, F. Fabbri, M. Rosetti et al., Addition of 5-Fluorouracil to Doxorubicin-Paclitaxel Sequence Increases Caspase-Dependent Apoptosis in Breast Cancer Cell Lines, Breast Cancer Res, vol.7, issue.5, 2005.

G. Mani, S. Kim, and K. Kim, Development of Folate-Thioglycolate-Gold Nanoconjugates by Using Citric Acid-PEG Branched Polymer for Inhibition of MCF-7 Cancer Cell Proliferation, Biomacromolecules, vol.19, issue.8, pp.3257-3267, 2018.

A. E. Kayl and C. A. Meyers, Side-Effects of Chemotherapy and Quality of Life in Ovarian and Breast Cancer Patients, Curr. Opin. Obstet. Gynecol, vol.18, issue.1, pp.24-28, 2006.

P. L. Azcona, M. G. Schneider, M. Grünhut, and V. L. Lassalle, Stimuli-Responsive Nanotheranostics Intended for Oncological Diseases: In Vitro Evaluation of Their Target, Diagnostic and Drug Release Capabilities, New J. Chem, vol.2019, issue.5, pp.2126-2133

D. E. Igartúa, P. L. Azcona, C. S. Martinez, S. Alonso, V. Lassalle et al., Folic Acid Magnetic Nanotheranostics for Delivering Doxorubicin: Toxicological and Biocompatibility Studies on Zebrafish Embryo and Larvae, Toxicol. Appl. Pharmacol, vol.358, pp.23-34, 2018.

H. Zhang, J. Li, Y. Hu, M. Shen, X. Shi et al., Folic Acid-Targeted Iron Oxide Nanoparticles as Contrast Agents for Magnetic Resonance Imaging of Human Ovarian Cancer

, J. Ovarian Res, vol.2016, issue.1, p.9

J. Zhu, H. Li, Z. Xiong, M. Shen, P. S. Conti et al., Polyethyleneimine-Coated Manganese Oxide Nanoparticles for Targeted Tumor PET/MR Imaging, ACS Appl. Mater. Interfaces, vol.10, issue.41, pp.34954-34964, 2018.

G. Mani, S. Kim, and K. Kim, Development of Folate-Thioglycolate-Gold Nanoconjugates by Using Citric Acid-PEG Branched Polymer for Inhibition of MCF-7 Cancer Cell Proliferation, Biomacromolecules, vol.19, issue.8, pp.3257-3267, 2018.

H. Chen, S. He, and . Pla-peg, Coated Multifunctional Imaging Probe for Targeted Drug Delivery, Mol. Pharm, vol.12, issue.6, pp.1885-1892, 2015.

T. J. Daou, G. Pourroy, S. Bégin-colin, J. M. Grenèche, C. Ulhaq-bouillet et al., Hydrothermal Synthesis of Monodisperse Magnetite Nanoparticles, Chem. Mater, vol.18, issue.18, pp.4399-4404, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00206075

W. Baaziz, B. P. Pichon, S. Fleutot, Y. Liu, C. Lefevre et al., Begin-Colin, S. Magnetic Iron Oxide Nanoparticles: Reproducible Tuning of the Size and Nanosized-Dependent Composition, Defects, and Spin Canting, J. Phys. Chem. C, issue.7, pp.3795-3810, 2014.

J. P. May and S. Li, Hyperthermia-Induced Drug Targeting, Expert Opin. Drug Deliv, vol.10, issue.4, pp.511-527, 2013.

Y. Chen, K. Ai, J. Liu, G. Sun, Q. Yin et al., Multifunctional Envelope-Type Mesoporous Silica Nanoparticles for PH-Responsive Drug Delivery and Magnetic Resonance Imaging, Biomaterials, vol.60, pp.111-120, 2015.

D. Luong, S. Sau, P. Kesharwani, and A. K. Iyer, Polyvalent Folate-Dendrimer-Coated Iron Oxide Theranostic Nanoparticles for Simultaneous Magnetic Resonance Imaging and Precise Cancer Cell Targeting, Biomacromolecules, vol.2017, issue.4, pp.1197-1209

Z. Ferjaoui, R. Schneider, A. Meftah, E. Gaffet, and H. Alem, Functional Responsive Superparamagnetic Core/Shell Nanoparticles and Their Drug Release Properties, vol.7, pp.26243-26249, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01523829

Y. Cao, J. He, J. Liu, M. Zhang, and P. Ni, Folate-Conjugated Polyphosphoester with Reversible Cross-Linkage and Reduction Sensitivity for Drug Delivery, ACS Appl. Mater. Interfaces, vol.10, issue.9, pp.7811-7820, 2018.

M. Xu, C. Y. Zhang, J. Wu, H. Zhou, R. Bai et al., PEG-Detachable Polymeric Micelles Self-Assembled from Amphiphilic Copolymers for Tumor-Acidity-Triggered Drug Delivery and Controlled Release, ACS Appl. Mater. Interfaces, vol.11, issue.6, pp.5701-5713, 2019.

S. Shen, Y. Wu, Y. Liu, and D. Wu, High Drug-Loading Nanomedicines: Progress, Current Status, and Prospects, Int. J. Nanomedicine, vol.12, pp.4085-4109, 2017.

M. Vallet-regi, A. Rámila, R. P. Del-real, and J. Pérez-pariente, A New Property of MCM-41: Drug Delivery System, Chem. Mater, vol.13, issue.2, pp.308-311, 2001.

J. Wu, Y. Zhu, S. Cao, and F. Chen, Hierachically Nanostructured Mesoporous Spheres of Calcium Silicate Hydrate: Surfactant-Free Sonochemical Synthesis and Drug-Delivery System with Ultrahigh Drug-Loading Capacity, Adv. Mater, issue.6, pp.749-753, 2010.

Y. Dai, D. Yang, P. Ma, X. Kang, X. Zhang et al., Doxorubicin Conjugated NaYF4:Yb3+/Tm3+ Nanoparticles for Therapy and Sensing of Drug Delivery by Luminescence Resonance Energy Transfer, Biomaterials, vol.2012, issue.33, pp.8704-8713

C. Fu, R. Yang, L. Wang, N. Li, M. Qi et al., Surface Functionalization of Superparamagnetic Nanoparticles by an Acid-Liable Polysaccharide-Based Prodrug for Combinatorial Monitoring and Chemotherapy of Hepatocellular Carcinoma, vol.7, pp.41919-41928, 2017.

V. Chapitre, Études des propriétés de la furtivité des Fe3-?O4 @ P(MEO2MAX-OEGMA100-X)

I. Hamad, O. Al-hanbali, A. C. Hunter, K. J. Rutt, T. L. 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.

A. E. Nel, L. Mädler, D. Velegol, T. Xia, E. M. Hoek et al., Understanding Biophysicochemical Interactions at the Nano-Bio Interface, Nat. Mater, vol.8, issue.7, pp.543-557, 2009.

I. Banerjee, R. C. Pangule, and R. S. Kane, Antifouling Coatings: Recent Developments in the Design of Surfaces That Prevent Fouling by Proteins, Bacteria, and Marine Organisms, Adv. Mater, vol.23, issue.6, pp.690-718, 2011.

S. Nir and M. Reches, Bio-Inspired Antifouling Approaches: The Quest towards Non-Toxic and Non-Biocidal Materials, Curr. Opin. Biotechnol, vol.39, pp.48-55, 2016.

P. Aggarwal, J. B. Hall, C. B. Mcleland, M. A. Dobrovolskaia, and S. E. Mcneil, Nanoparticle Interaction with Plasma Proteins as It Relates to Particle Biodistribution, Biocompatibility and Therapeutic Efficacy, Adv. Drug Deliv. Rev, issue.6, pp.428-437, 2009.

J. Müller, J. Simon, P. Rohne, C. Koch-brandt, V. Mailänder et al., Denaturation via Surfactants Changes Composition of Protein Corona, Biomacromolecules, vol.19, issue.7, pp.2657-2664, 2018.

B. Pelaz, P. Del-pino, P. Maffre, R. Hartmann, M. Gallego et al., Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake, ACS Nano, vol.9, issue.7, pp.6996-7008, 2015.

C. Rodríguez-nogales, Y. González-fernández, A. Aldaz, P. Couvreur, and M. J. Blanco-prieto, Nanomedicines for Pediatric Cancers, ACS, vol.12, issue.8, pp.7482-7496, 2018.

G. Mani, S. Kim, and K. Kim, Development of Folate-Thioglycolate-Gold Nanoconjugates by Using Citric Acid-PEG Branched Polymer for Inhibition of MCF-7 Cancer Cell Proliferation, Biomacromolecules, vol.19, issue.8, pp.3257-3267, 2018.

S. Winzen, J. C. Schwabacher, J. Müller, K. Landfester, and K. Mohr, Small Surfactant Concentration Differences Influence Adsorption of Human Serum Albumin on Polystyrene Nanoparticles, Biomacromolecules, vol.2016, issue.11, pp.3845-3851

L. Guerrini, R. Alvarez-puebla, and N. Pazos-perez, Surface Modifications of Nanoparticles for Stability in Biological Fluids, Materials, vol.2018, issue.7, p.1154

Q. He, J. Zhang, J. Shi, Z. Zhu, L. Zhang et al., The Effect of PEGylation of Mesoporous Silica Nanoparticles on Nonspecific Binding of Serum Proteins and Cellular Responses, Biomaterials, vol.2010, issue.6, pp.1085-1092

M. Moros, B. Pelaz, P. López-larrubia, M. L. García-martin, V. Grazú et al., Engineering Biofunctional Magnetic Nanoparticles for Biotechnological Applications, Nanoscale, vol.2010, issue.9, p.1746

P. Rivera-gil, D. Jimenez-de-aberasturi, V. Wulf, B. Pelaz, P. Del-pino et al., The Challenge To Relate the Physicochemical Properties of Colloidal Nanoparticles to Their Cytotoxicity, Acc. Chem. Res, vol.46, issue.3, pp.743-749, 2013.

R. A. Sperling, T. Liedl, S. Duhr, S. Kudera, M. Zanella et al., Size Determination of (Bio) Conjugated Water-Soluble Colloidal Nanoparticles: A Comparison of Different Techniques, J. Phys. Chem. C, issue.31, pp.11552-11559, 2007.

J. Deng, Z. Li, M. Yao, and C. Gao, Influence of Albumin Configuration by the Chiral Polymer-Grafted Gold Nanoparticles, Langmuir, vol.2016, issue.22, pp.5608-5616

B. Pelaz, P. Del-pino, P. Maffre, R. Hartmann, M. Gallego et al., Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake, ACS Nano, vol.9, issue.7, pp.6996-7008, 2015.

C. D. Walkey, J. B. Olsen, H. Guo, A. Emili, and W. C. Chan, Nanoparticle Size and Surface Chemistry Determine Serum Protein Adsorption and Macrophage Uptake, J. Am. Chem. Soc, vol.2012, issue.4, pp.2139-2147

A. C. Weiss, K. Krüger, Q. A. Besford, M. Schlenk, K. Kempe et al., Situ Characterization of Protein Corona Formation on Silica Microparticles Using Confocal Laser Scanning Microscopy Combined with Microfluidics, ACS Appl. Mater. Interfaces, vol.2019, issue.2, pp.2459-2469

C. R. Anderson, Y. D. Gnopo, F. Gambinossi, S. E. Mylon, and J. K. Ferri, Modulation of Cell Responses to Ag-(MeO 2 MA-Co -OEGMA): Effects of Nanoparticle Surface Hydrophobicity and Serum Proteins on Cellular Uptake and Toxicity: CELL RESPONSES TO POLYMERIC AgNPs, J. Biomed. Mater. Res. A, vol.106, issue.4, pp.1061-1071, 2018.

M. Nazarenus, Q. Zhang, M. G. Soliman, P. Pino, B. Pelaz et al., In Vitro Interaction of Colloidal Nanoparticles with Mammalian Cells: What Have We Learned Thus Far?, Beilstein J. Nanotechnol, vol.5, pp.1477-1490, 2014.

C. Bernhard, K. N. Bauer, M. Bonn, F. R. Wurm, and G. Gonella, Interfacial Conformation of Hydrophilic Polyphosphoesters Affects Blood Protein Adsorption, ACS Appl. Mater. Interfaces, vol.2019, issue.1, pp.1624-1629

H. Zhang, T. Wu, W. Yu, S. Ruan, Q. He et al., Ligand Size and Conformation Affect the Behavior of Nanoparticles Coated with in Vitro and in Vivo Protein Corona, ACS Appl. Mater. Interfaces, vol.10, issue.10, pp.9094-9103, 2018.

J. Y. Oh, H. S. Kim, L. Palanikumar, E. M. Go, B. Jana et al., Cloaking Nanoparticles with Protein Corona Shield for Targeted Drug Delivery, Nat. Commun, vol.9, issue.1, p.4548, 2018.

C. Chen, J. Sun, S. Chen, Y. Liu, S. Zhu et al., A Multifunctional-Targeted Nanoagent for Dual-Mode Image-Guided Therapeutic Effects on Ovarian Cancer Cells, Int. J. Nanomedicine, vol.14, pp.753-769, 2019.

I. Vergote, F. Amant, G. Kristensen, T. Ehlen, N. S. Reed et al., Primary Surgery or Neoadjuvant Chemotherapy Followed by Interval Debulking Surgery in Advanced Ovarian Cancer, European Journal of Cancer, vol.47, issue.11, pp.70152-70158, 2011.

S. M. Eisenkop, N. M. Spirtos, and W. Lin, Optimal" Cytoreduction for Advanced Epithelial Ovarian Cancer: A Commentary, Gynecologic Oncology, vol.103, issue.1, pp.329-335, 2006.

S. Bhide and C. Nutting, Recent Advances in Radiotherapy, BMC Medicine, vol.8, issue.1, 2010.

K. Moghissi, R. J. Parsons, and K. Dixon, Photodynamic Therapy (PDT) for Bronchial Carcinoma with the Use of Rigid Bronchoscope. Lasers in Medical Science, vol.7, pp.381-385, 1992.

A. Coates, S. Abraham, S. B. Kaye, T. Sowerbutts, C. Frewin et al., On the Receiving End-Patient Perception of the Side-Effects of Cancer Chemotherapy, European Journal of Cancer and Clinical Oncology, vol.19, issue.2, pp.203-208, 1983.

C. Rodríguez-nogales, Y. González-fernández, A. Aldaz, P. Couvreur, and M. J. Blanco-prieto, Nanomedicines for Pediatric Cancers, ACS, vol.12, issue.8, pp.7482-7496, 2018.

A. Hervault, A. E. Dunn, M. Lim, C. Boyer, D. Mott et al., Doxorubicin Loaded Dual PH-and Thermo-Responsive Magnetic Nanocarrier for Combined Magnetic Hyperthermia and Targeted Controlled Drug Delivery Applications, Nanoscale, vol.8, issue.24, pp.12152-12161, 2016.

C. A. Quinto, P. Mohindra, S. Tong, and G. Bao, Multifunctional Superparamagnetic Iron Oxide Nanoparticles for Combined Chemotherapy and Hyperthermia Cancer Treatment, Nanoscale, vol.2015, issue.29, pp.12728-12736

J. Y. Oh, H. S. Kim, L. Palanikumar, E. M. Go, B. Jana et al., Cloaking Nanoparticles with Protein Corona Shield for Targeted Drug Delivery, Nature Communications, vol.9, issue.1, p.4548, 2018.

Z. Ferjaoui, R. Schneider, A. Meftah, E. Gaffet, and H. Alem, Functional Responsive Superparamagnetic Core/Shell Nanoparticles and Their Drug Release Properties, vol.7, pp.26243-26249, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01523829

V. A. Milichko, A. I. Nechaev, V. A. Valtsifer, V. N. Strelnikov, Y. N. Kulchin et al., Photo-Induced Electric Polarizability of Fe3O4 Nanoparticles in Weak Optical Fields, Nanoscale Research Letters, vol.2013, issue.1

D. Maity and D. C. Agrawal, Synthesis of Iron Oxide Nanoparticles under Oxidizing Environment and Their Stabilization in Aqueous and Non-Aqueous Media, Journal of Magnetism and Magnetic Materials, vol.308, issue.1, pp.46-55, 2007.

M. Mascolo, Y. Pei, and T. Ring, Room Temperature Co-Precipitation Synthesis of Magnetite Nanoparticles in a Large PH Window with Different Bases, Materials, vol.2013, issue.12, pp.5549-5567

A. Ali, H. Zafar, M. Zia, I. Haq, A. R. Phull et al., Synthesis, Characterization, Applications, and Challenges of Iron Oxide Nanoparticles, Nanotechnology, Science and Applications, vol.9, pp.49-67, 2016.

J. Al-dine, E. Ferjaoui, Z. Ghanbaja, J. Roques-carmes, T. Meftah et al., Thermo-Responsive Magnetic Fe 3 O 4 @P(MEO 2 MA X -OEGMA 100-X ) NPs and Their Applications as Drug Delivery Systems, International Journal of Pharmaceutics, vol.532, issue.2, pp.738-747, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01650405

L. M. Kaminskas, V. M. Mcleod, B. D. Kelly, G. Sberna, B. J. Boyd et al., A Comparison of Changes to Doxorubicin Pharmacokinetics, Antitumor Activity, and Toxicity Mediated by PEGylated Dendrimer and PEGylated Liposome Drug Delivery Systems, Nanomedicine: Nanotechnology, Biology and Medicine, vol.8, issue.1, pp.103-111, 2012.

A. Hervault, A. E. Dunn, M. Lim, C. Boyer, D. Mott et al., Doxorubicin Loaded Dual PH-and Thermo-Responsive Magnetic Nanocarrier for Combined Magnetic Hyperthermia and Targeted Controlled Drug Delivery Applications, Nanoscale, vol.8, issue.24, pp.12152-12161, 2016.

B. T. Mai, P. B. Balakrishnan, M. J. Barthel, F. Piccardi, D. Niculaes et al., Thermoresponsive Iron Oxide Nanocubes for an Effective Clinical Translation of Magnetic Hyperthermia and Heat-Mediated Chemotherapy, ACS Applied Materials & Interfaces, vol.11, issue.6, pp.5727-5739, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02415100

Y. Tian, S. Bian, and W. Yang, A Redox-Labile Poly(Oligo(Ethylene Glycol)Methacrylate)-Based Nanogel with Tunable Thermosensitivity for Drug Delivery, Polymer Chemistry, vol.7, issue.10, pp.1913-1921, 2016.

W. Pon-on, T. Tithito, W. Maneeprakorn, T. Phenrat, and I. Tang, Investigation of Magnetic Silica with Thermoresponsive Chitosan Coating for Drug Controlled Release and Magnetic Hyperthermia Application, Materials Science and Engineering: C, vol.97, pp.23-30, 2019.

Z. Tian, X. Yu, Z. Ruan, M. Zhu, Y. Zhu et al., Magnetic Mesoporous Silica Nanoparticles Coated with Thermo-Responsive Copolymer for Potential Chemo-and Magnetic Hyperthermia Therapy, Microporous and Mesoporous Materials, vol.256, pp.1-9, 2018.

M. E. Peralta, S. A. Jadhav, G. Magnacca, D. Scalarone, D. O. Mártire et al., Synthesis and in Vitro Testing of Thermoresponsive Polymer-Grafted Core-Shell Magnetic Mesoporous Silica Nanoparticles for Efficient Controlled and Targeted Drug Delivery, Journal of Colloid and Interface Science, vol.544, pp.198-205, 2019.

N. Andhariya, B. Chudasama, R. V. Mehta, and R. V. Upadhyay, Biodegradable Thermoresponsive Polymeric Magnetic Nanoparticles: A New Drug Delivery Platform for Doxorubicin, J Nanopart Res, vol.13, issue.4, pp.1677-1688, 2011.

B. Sahoo, K. S. Devi, R. Banerjee, T. K. Maiti, P. Pramanik et al., Thermal and PH Responsive Polymer-Tethered Multifunctional Magnetic Nanoparticles for Targeted Delivery of Anticancer Drug, ACS Appl. Mater. Interfaces, vol.2013, issue.9, pp.3884-3893

J. L. Dormann and . Le-phénomène-de-superparamagnétisme, Revue de Physique Appliquée, vol.16, pp.275-301, 1981.

M. Kallumadil, M. Tada, T. Nakagawa, M. Abe, P. Southern et al., Suitability of Commercial Colloids for Magnetic Hyperthermia, Journal of Magnetism and Magnetic Materials, vol.321, issue.10, pp.1509-1513, 2009.

C. Blanco-andujar, D. Ortega, P. Southern, Q. A. Pankhurst, and N. T. Thanh, High Performance Multi-Core Iron Oxide Nanoparticles for Magnetic Hyperthermia: Microwave Synthesis, and the Role of Core-to-Core Interactions, Nanoscale, vol.7, issue.5, pp.1768-1775, 2015.

C. Blanco-andujar, D. Ortega, P. Southern, Q. A. Pankhurst, and N. T. Thanh, High Performance Multi-Core Iron Oxide Nanoparticles for Magnetic Hyperthermia: Microwave Synthesis, and the Role of Core-to-Core Interactions, Nanoscale, vol.7, issue.5, pp.1768-1775, 2015.

A. E. Dunn, D. J. Dunn, A. Macmillan, R. Whan, T. Stait-gardner et al., Spatial and Temporal Control of Drug Release through PH and Alternating Magnetic Field Induced Breakage of Schiff Base Bonds, Polym. Chem, vol.5, issue.10, pp.3311-3315, 2014.

S. Sene, M. T. Marcos-almaraz, N. Menguy, J. Scola, J. Volatron et al., Maghemite-NanoMIL-100(Fe) Bimodal Nanovector as a Platform for Image-Guided Therapy, vol.2017, pp.303-322
URL : https://hal.archives-ouvertes.fr/hal-01868345

M. Terracciano, M. Shahbazi, A. Correia, I. Rea, A. Lamberti et al., Surface Bioengineering of Diatomite Based Nanovectors for Efficient Intracellular Uptake and Drug Delivery, Nanoscale, vol.7, issue.47, pp.20063-20074, 2015.

H. Chen, S. He, and . Pla-peg, Coated Multifunctional Imaging Probe for Targeted Drug Delivery, Molecular Pharmaceutics, vol.12, issue.6, pp.1885-1892, 2015.

G. Mani, S. Kim, and K. Kim, Development of Folate-Thioglycolate-Gold Nanoconjugates by Using Citric Acid-PEG Branched Polymer for Inhibition of MCF-7 Cancer Cell Proliferation, Biomacromolecules, vol.19, issue.8, pp.3257-3267, 2018.

S. ;. and P. , Multi-Functional Core-Shell Fe3O4@Au Nanoparticles for Cancer Diagnosis and Therapy, Colloids and Surfaces B: Biointerfaces, vol.174, pp.252-259, 2019.

D. Bhattacharya, B. Behera, S. K. Sahu, R. Ananthakrishnan, T. K. Maiti et al., Design of Dual Stimuli Responsive Polymer Modified Magnetic Nanoparticles for Targeted Anti-Cancer Drug Delivery and Enhanced MR Imaging, New J. Chem, vol.40, issue.1, pp.545-557, 2016.

S. J. Rezaei, M. R. Nabid, H. Niknejad, and A. A. Entezami, Folate-Decorated Thermoresponsive Micelles Based on Star-Shaped Amphiphilic Block Copolymers for Efficient Intracellular Release of Anticancer Drugs, International Journal of Pharmaceutics, vol.2012, issue.1-2, pp.70-79

V. Sundaresan, J. U. Menon, M. Rahimi, K. T. Nguyen, and A. S. Wadajkar, Dual-Responsive Polymer-Coated Iron Oxide Nanoparticles for Drug Delivery and Imaging Applications, International Journal of Pharmaceutics, vol.466, issue.1-2, p.119, 2014.

H. Zhang, J. Li, Y. Hu, M. Shen, X. Shi et al., Folic Acid-Targeted Iron Oxide Nanoparticles as Contrast Agents for Magnetic Resonance Imaging of Human Ovarian Cancer, Journal of Ovarian Research, vol.2016, issue.1

,

Y. Cao, J. He, J. Liu, M. Zhang, and P. Ni, Folate-Conjugated Polyphosphoester with Reversible Cross-Linkage and Reduction Sensitivity for Drug Delivery, ACS Applied Materials & Interfaces, vol.10, issue.9, pp.7811-7820, 2018.

K. Kaaki, K. Hervé-aubert, M. Chiper, A. Shkilnyy, M. Soucé et al., Magnetic Nanocarriers of Doxorubicin Coated with Poly(Ethylene Glycol) and Folic Acid: Relation between Coating Structure, Surface Properties, Colloidal Stability, and Cancer Cell Targeting, Langmuir, vol.2012, issue.2, pp.1496-1505
URL : https://hal.archives-ouvertes.fr/hal-01387921

D. Luong, S. Sau, P. Kesharwani, and A. K. Iyer, Polyvalent Folate-Dendrimer-Coated Iron Oxide Theranostic Nanoparticles for Simultaneous Magnetic Resonance Imaging and Precise Cancer Cell Targeting, Biomacromolecules, vol.2017, issue.4, pp.1197-1209

W. J. Lin and . Liu, Polymeric Nanoparticles Conjugate a Novel Heptapeptide as an Epidermal Growth Factor Receptor-Active Targeting Ligand for Doxorubicin, International Journal of Nanomedicine, vol.4749, 2012.

J. A. Vassie, J. M. Whitelock, and M. S. Lord, Targeted Delivery and Redox Activity of Folic Acid-Functionalized Nanoceria in Tumor Cells, Molecular Pharmaceutics, vol.15, issue.3, pp.994-1004, 2018.

R. Sakhtianchi, F. Atyabi, P. Yousef, E. Vasheghani-farahani, M. Movahedi et al., Targeted Delivery of Doxorubicin-Utilizing Chitosan Nanoparticles Surface-Functionalized with Anti-Her2 Trastuzumab, International Journal of Nanomedicine, 1977.

A. K. Iyer, G. Khaled, J. Fang, and H. Maeda, Exploiting the Enhanced Permeability and Retention Effect for Tumor Targeting, Drug Discovery Today, vol.11, pp.812-818, 2006.

J. Deng, Z. Li, M. Yao, and C. Gao, Influence of Albumin Configuration by the Chiral Polymer-Grafted Gold Nanoparticles, Langmuir, vol.2016, issue.22, p.120

D. Thassu, M. Deleers, and Y. V. Pathak, Nanoparticulate Drug Delivery Systems, 2007.

H. M. Duvernoy, The Human Brain: Surface, Three-Dimensional Sectional Anatomy with MRI, and Blood Supply, 2012.

L. Li, W. Jiang, K. Luo, H. Song, F. Lan et al., Theranostics, issue.3, pp.595-615, 2013.

P. B. Santhosh and N. P. Ulrih, Cancer Lett, vol.336, pp.8-17, 2013.

R. , G. Chaudhuri, and S. Paria, Chem. Rev, vol.112, pp.2373-2433, 2012.

N. Lee, D. Yoo, D. Ling, M. H. Cho, T. Hyeon et al., Chem. Rev, vol.115, pp.10637-10689, 2015.

S. Laurent, D. Forge, M. Port, A. Roch, C. Robic et al., Chem. Rev, vol.108, pp.2064-2110, 2008.

P. Kucheryavy, J. He, V. T. John, P. Maharjan, L. Spinu et al., Langmuir, vol.29, pp.710-716, 2013.

M. Mahmoudi, H. Hofmann, B. Rothen-rutishauser, and A. Petri-fink, Chem. Rev, vol.112, pp.2323-2338, 2012.

A. K. Gupta and M. Gupta, Biomaterials, vol.26, pp.3995-4021, 2005.

S. Ganta, H. Devalapally, A. Shahiwala, and M. Amiji, J. Controlled Release, vol.126, pp.187-204, 2008.

M. A. Stuart, W. T. Huck, J. Genzer, M. Müller, C. Ober et al., Nat. Mater, vol.9, pp.101-113, 2010.

I. Y. Galaev and B. Mattiasson, Trends Biotechnol, vol.17, pp.335-340, 1999.

D. Schmaljohann and A. , Drug Delivery Rev, vol.58, pp.1655-1670, 2006.

E. Wischerhoff, K. Uhlig, A. Lankenau, H. G. Börner, A. Laschewsky et al., Angew. Chem., Int. Ed, vol.47, pp.5666-5668, 2008.

M. Chanana, S. Jahn, R. Georgieva, J. Lutz, H. Bäumler et al., Chem. Mater, vol.21, pp.1906-1914, 2009.

A. M. Jonas, K. Glinel, R. Oren, B. Nysten, and W. T. Huck, Macromolecules, vol.40, pp.4403-4405, 2007.

R. Barbey, L. Lavanant, D. Paripovic, N. Schüwer, C. Sugnaux et al., Chem. Rev, vol.109, pp.5437-5527, 2009.

S. Edmondson, V. L. Osborne, and W. T. Huck, Chem. Soc. Rev, vol.33, pp.14-22, 2004.

E. J. Dine, Z. Ferjaoui, T. Roques-carmes, A. Schjen, A. Me?ah et al., Nanotechnology, 2017.

K. Matyjaszewski, W. Jakubowski, K. Min, W. Tang, J. Huang et al., Proc. Natl. Acad. Sci. U. S. A, vol.103, pp.15309-15314, 2006.

K. Matyjaszewski, H. Dong, W. Jakubowski, J. Pietrasik, and A. Kusumo, Langmuir, vol.23, pp.4528-4531, 2007.

H. Alem, A. Schejn, T. Roques-carmes, J. Ghanbaja, and R. Schneider, Nanotechnology, p.335605, 2015.

V. M. Gaspar, E. C. Costa, J. A. Queiroz, C. Pichon, F. Sousa et al., Pharm. Res, vol.32, pp.562-577, 2015.

N. Li, T. Li, C. Liu, S. Ye, J. Liang et al., J. Biomed. Nanotechnol, vol.12, pp.878-893, 2016.

B. Sahoo, K. S. Devi, R. Banerjee, T. K. Maiti, P. Pramanik et al., ACS Appl. Mater. Interfaces, issue.5, pp.3884-3893, 2013.

H. Maeda, K. Tsukigawa, and J. Fang, Microcirculation, vol.23, pp.173-182, 2016.

H. Maeda, Adv. Drug Delivery Rev, vol.91, pp.3-6, 2015.

Y. J. Wang, S. M. Hussain, and G. P. Krestin, Eur. Radiol, vol.11, pp.2319-2331, 2001.

A. Gloria, T. Russo, U. D'amora, S. Zeppetelli, T. D'alessandro et al., J. R. Soc., Interface, issue.10, 2013.

K. Hagane, T. Akera, and J. R. Berlin, J. Pharmacol. Exp. Ther, vol.246, pp.655-661, 1988.

A. Hervault, A. E. Dunn, M. Lim, C. Boyer, D. Mott et al., Nanoscale, vol.8, pp.12152-12161, 2016.

D. Missirlis, R. Kawamura, N. Tirelli, and J. A. Hubbell, Eur. J. Pharm. Sci, vol.29, pp.120-129, 2006.

S. Louguet, B. Rousseau, R. Epherre, N. Guidolin, G. Goglio et al., Polym. Chem, vol.3, pp.1408-1417, 2012.

S. Z. Moghaddam, E. Thormann, and . Adv, , vol.6, pp.27969-27973, 2016.

J. Iwahara, A. Esadze, and L. Zandarashvili, Biomolecules, vol.5, pp.2435-2463, 2015.

, This journal is © The Royal Society of Chemistry 2017 RSC Adv, vol.7, pp.26243-26249, 2017.

, Doxorubicin-Loaded Thermoresponsive Superparamagnetic Nanocarriers for Controlled Drug Delivery and Magnetic Hyperthermia Applications Zied Ferjaoui, vol.7198, p.54011

, All reagents were purchased from Sigma-Aldrich, except for ((chloromethyl)phenylethyl)trimethoxysilane (CMPETMS) (Gelest, >95%). iron(III) chloride hexahydrate (Lancaster, 98%), iron(II) sulfate heptahydrate (Merck, 99.5%), ammonia solution (NH 3 , 28? 30% w/v), and sodium citrate (99.8%), which were all purchased from Sigma-Aldrich. To graft the polymerization initiator, ((chloromethyl) phenylethyl)trimethoxysilane (CMPETMS) (Gelest, >95%), tetramethylammonium hydroxide pentahydrate (TMAOH) (VWR chemicals, 99.8%), and toluene (Laboratory Reagent, >99.the 2-(2-methoxy)ethyl methacrylate

N. , dimethyl sulfoxide (DMSO) (>99.8%), and Milli-Q water from Sigma-Aldrich, were used. The biological studies were performed in Dulbecco's modified eagle's medium high glucose (DMEM; Sigma-Aldrich, Saint-Louis) for the growth of the cells, DMF) (>99.8%)

. Sigma-aldrich,

C. Janko, T. Ratschker, K. Nguyen, L. Zschiesche, R. Tietze et al., Functionalized Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as Platform for the Targeted Multimodal Tumor Therapy, Front. Oncol, vol.9, p.59, 2019.

G. Kandasamy and D. Maity, Recent Advances in Superparamagnetic Iron Oxide Nanoparticles (SPIONs) for in Vitro and in Vivo Cancer Nanotheranostics, Int. J. Pharm, vol.496, pp.191-218, 2015.

E. Cazares-cortes, A. Espinosa, J. Guigner, A. Michel, N. Griffete et al., Doxorubicin Intracellular Remote Release from Biocompatible Oligo(Ethylene Glycol) Methyl Ether Methacrylate-Based Magnetic Nanogels Triggered by Magnetic Hyperthermia, ACS Appl. Mater. Interfaces, vol.9, pp.25775-25788, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01586118

D. Mertz and O. Sandre, Be?in-Colin, S. Drug Releasing Nanoplatforms Activated by Alternating Magnetic Fields, Biochim. Biophys. Acta, 1617.

G. K. Thirunavukkarasu, K. Cherukula, H. Lee, Y. Y. Jeong, I. Park et al., Magnetic Field-Inducible Drug-Eluting Nanoparticles for Image-Guided Thermo, Chemotherapy. Biomaterials, vol.180, pp.240-252, 2018.

W. Xue, X. Liu, H. Ma, W. Xie, S. Huang et al., AMF Responsive DOX-Loaded Magnetic Microspheres: Transmembrane Drug Release Mechanism and Multimodality Postsurgical Treatment of Breast Cancer, J. Mater. Chem. B, vol.6, pp.2289-2303, 2018.

E. A. Pe?igo, G. Hemery, O. Sandre, D. Ortega, E. Garaio et al., J. Fundamentals and Advances in Magnetic Hyperthermia. Appl. Phys. Rev, vol.2, p.41302, 2015.

S. Annamaneni, S. K. Vishwakarma, P. B. Meka, A. A. Khan, and P. Nallari, Regulation of Heat Shock Protein-70 Gene Transcripts in Breast Cancer Cells during Hypo and Hyperthermia Exposure, Meta Gene, vol.20, 2019.

P. Das, M. Colombo, and D. Prosperi, Recent Advances in Magnetic Fluid Hyperthermia for Cancer Therapy, Colloids Surf, vol.174, pp.42-55, 2019.

T. Saliev, L. B. Feril, D. Begimbetova, D. Baiskhanova, A. Klodzinskyi et al., Hyperthermia Enhances Bortezomib-Induced Apoptosis in Human White Blood Cancer Cells, J. Therm. Biol, vol.67, pp.9-14, 2017.

W. Andra and H. Nowak, Magnetism in Medicine: A Handbook, 2007.

M. Banõbre-lo?ez, A. Teijeiro, and J. Rivas, Magnetic Nanoparticle-Based Hyperthermia for Cancer Treatment, Rep. Pract. Oncol. Radiother, vol.18, pp.397-400, 2013.

N. D. Thorat, R. A. Bohara, M. R. Noor, D. Dhamecha, T. Soulimane et al., Effective Cancer Theranostics with Polymer Encapsulated Superparamagnetic Nanoparticles: Combined Effects of Magnetic Hyperthermia and Controlled Drug Release, ACS Biomater. Sci. Eng, vol.3, pp.1332-1340, 2017.

X. Guo, W. Li, L. Luo, Z. Wang, Q. Li et al., External Magnetic Field-Enhanced Chemo-Photothermal Combination Tumor Therapy via Iron Oxide Nanoparticles, ACS Appl. Mater. Interfaces, vol.9, pp.16581-16593, 2017.

A. Hervault, A. E. Dunn, M. Lim, C. Boyer, D. Mott et al., Doxorubicin Loaded Dual PH-and Thermo-Responsive Magnetic Nanocarrier for Combined Magnetic Hyperthermia and Targeted Controlled Drug Delivery Applications, Nanoscale, vol.8, pp.12152-12161, 2016.

C. Blanco-andujar, A. Walter, G. Cotin, C. Bordeianu, D. Mertz et al., Begin-Colin, S. Design of Iron Oxide-Based Nanoparticles for MRI and Magnetic Hyperthermia, Nanomedicine, vol.11, 1889.

S. Lara, A. Perez-potti, L. M. Herda, L. Adumeau, K. A. Dawson et al., Differential Recognition of Nanoparticle Protein Corona and Modified Low-Density Lipoprotein by Macrophage Receptor with Collagenous Structure, ACS, vol.12, pp.4930-4937, 2018.

N. Zhao, L. Yan, X. Zhao, X. Chen, A. Li et al., Versatile Types of Organic/Inorganic Nanohybrids: From Strategic Design to Biomedical Applications, Chem. Rev, vol.119, pp.1666-1762, 2019.

N. Nath, A. Chilkoti, and . Creating, Smart" Surfaces Using Stimuli Responsive Polymers, Adv. Mater, vol.14, pp.1243-1247, 2002.

A. K. Teotia, H. Sami, and A. Kumar, 1 -Thermo-Responsive Polymers: Structure and Design of Smart Materials, Switchable and Responsive Surfaces and Materials for Biomedical Applications

Z. Zhang and . Ed, , 2015.

R. Hoogenboom, 2 -Temperature-Responsive Polymers: Properties, Synthesis and Applications, Smart Polymers and their Applications

M. R. Aguilar and J. San-roma?, , pp.15-44, 2014.

D. Schmaljohann, Thermo-and PH-Responsive Polymers in Drug Delivery, Adv. Drug Delivery Rev, vol.58, pp.1655-1670, 2006.

D. Roy, W. L. Brooks, and B. S. Sumerlin, New Directions in Thermoresponsive Polymers, Chem. Soc. Rev, vol.42, pp.7214-7243, 2013.

Z. Ferjaoui, R. Schneider, A. Meftah, E. Gaffet, and H. Alem, Functional Responsive Superparamagnetic Core/Shell Nanoparticles and Their Drug Release Properties, vol.7, pp.26243-26249, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01523829

J. Al-dine, E. Ferjaoui, Z. Ghanbaja, J. Roques-carmes, T. Meftah et al., Thermo-Responsive Magnetic Fe3O4@P(MEO2MAX-OEGMA100-X) NPs and Their Applications as Drug Delivery Systems, Int. J. Pharm, vol.532, pp.738-747, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01650405

E. J. Dine, Z. Ferjaoui, T. Roques-carmes, A. Schjen, A. Meftah et al., Efficient Synthetic Access to Thermo-Responsive Core/Shell Nanoparticles, Nanotechnology, vol.28, p.125601, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01461773

K. N. Plunkett, X. Zhu, J. S. Moore, and D. E. Leckband, PNIPAM Chain Collapse Depends on the Molecular Weight and Grafting Density, Langmuir, vol.22, pp.4259-4266, 2006.

H. Vihola, A. Laukkanen, L. Valtola, H. Tenhu, and J. Hirvonen, Cytotoxicity of Thermosensitive Polymers Poly(N-Isopropylacrylamide), Poly(N-Vinylcaprolactam) and Amphiphilically Modified Poly(N-Vinylcaprolactam), vol.26, pp.3055-3064, 2005.

L. Li, C. Zhang, R. Zhang, Z. Xu, Z. Xu et al., Multifunctional Magnetized Porous Silica Covered with Poly(2-Dimethylaminoethyl Methacrylate) for PH Controllable Drug Release and Magnetic Resonance Imaging, ACS Appl. Nano Mater, vol.1, pp.5027-5034, 2018.

M. A. Macchione, C. Biglione, M. Strumia, and . Design, Synthesis and Architectures of Hybrid Nanomaterials for Therapy and Diagnosis Applications, Polymers, vol.10, p.527, 2018.

M. S. Shim and Y. J. Kwon, Stimuli-Responsive Polymers and Nanomaterials for Gene Delivery and Imaging Applications, Adv. Drug Delivery Rev, vol.64, pp.1046-1059, 2012.

P. James, H. John, R. Alex, A. Anoop, and K. R. , Smart Polymers for the Controlled Delivery of Drugs ? a Concise Overview, Acta Pharm. Sin. B, vol.4, pp.120-127, 2014.

N. P. Mortensen, G. B. Hurst, W. Wang, C. M. Foster, P. D. Nallathamby et al., Dynamic Development of the Protein Corona on Silica Nanoparticles: Composition and Role in Toxicity, Nanoscale, vol.5, pp.6372-6380, 2013.

R. García-a?lvarez, M. Hadjidemetriou, A. Sa?chez-iglesias, L. M. Liz-marza?, and K. Kostarelos, In Vivo Formation of Protein Corona on Gold Nanoparticles. The Effect of Their Size and Shape, Nanoscale, vol.10, pp.1256-1264, 2018.

Z. S. Al-ahmady, M. Hadjidemetriou, J. Gubbins, and K. Kostarelos, Formation of Protein Corona in Vivo Affects Drug Release from Temperature-Sensitive Liposomes, J. Controlled Release, vol.276, pp.157-167, 2018.

G. Caracciolo, O. C. Farokhzad, and M. Mahmoudi, Biological Identity of Nanoparticles In Vivo: Clinical Implications of the Protein Corona, Trends Biotechnol, vol.35, pp.257-264, 2017.

S. Tenzer, D. Docter, J. Kuharev, A. Musyanovych, V. Fetz et al., Rapid Formation of Plasma Protein Corona Critically Affects Nanoparticle Pathophysiology, Nat. Nanotechnol, vol.8, pp.772-781, 2013.

C. Corbo, R. Molinaro, M. Tabatabaei, O. C. Farokhzad, and M. Mahmoudi, Personalized Protein Corona on Nanoparticles and Its Clinical Implications, Biomater. Sci, vol.5, pp.378-387, 2017.

T. Gillich, C. Acikgoz, L. Isa, A. D. Schlu?er, N. D. Spencer et al., PEG-Stabilized Core?Shell Nanoparticles: Impact of Linear versus Dendritic Polymer Shell Architecture on Colloidal Properties and the Reversibility of Temperature-Induced Aggregation, ACS Nano, vol.7, pp.316-329, 2013.

Q. Sun, D. Cheng, X. Yu, Z. Zhang, J. Dai et al., A PH-Sensitive Polymeric Nanovesicle Based on Biodegradable Poly(Ethylene Glycol)-b-Poly(2-(Diisopropylamino)Ethyl Aspartate) as a MRI-Visible Drug Delivery System, J. Mater. Chem, pp.21-15316, 2011.

J. Lutz, Polymerization of Oligo(Ethylene Glycol) (Meth)-Acrylates: Toward New Generations of Smart Biocompatible Materials

, J. Polym. Sci., Part A: Polym. Chem, vol.46, pp.3459-3470, 2008.

X. Gao, N. Kuc?rka, M. Nieh, J. Katsaras, S. Zhu et al., Chain Conformation of a New Class of PEG-Based Thermoresponsive Polymer Brushes Grafted on Silicon as Determined by Neutron Reflectometry, Langmuir, vol.25, pp.10271-10278, 2009.

H. Alem, A. Schejn, T. Roques-carmes, J. Ghanbaja, and R. Schneider, Thermo-Responsive and Aqueous Dispersible ZnO/ PNIPAM Core/Shell Nanoparticles, Nanotechnology, vol.26, issue.335605, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01266251

J. F. Martínez-lo?ez, S. Schneider, D. Salavera, A. M. Mainar, J. S. Urieta et al., Molar Heat Capacities of the Mixture {1,8-Cineole+ethanol} at Several Temperatures and Atmospheric Pressure

, J. Chem. Thermodyn, vol.92, pp.146-151, 2016.

E. Abumandour, F. Mutelet, and D. Alonso, Performance of an Absorption Heat Transformer Using New Working Binary Systems Composed of {ionic Liquid and Water}, Appl. Therm. Eng, vol.94, pp.579-589, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01934461

T. J. Daou, G. Pourroy, S. Be?in-colin, J. M. Greneche, C. Ulhaq-bouillet et al., Hydrothermal Synthesis of Monodisperse Magnetite Nanoparticles, Chem. Mater, vol.18, pp.4399-4404, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00206075

W. Baaziz, B. P. Pichon, S. Fleutot, Y. Liu, C. Lefevre et al., Begin-Colin, S. Magnetic Iron Oxide Nanoparticles: Reproducible Tuning of the Size and Nanosized-Dependent Composition, Defects, and Spin Canting, J. Phys. Chem. C, vol.118, pp.3795-3810, 2014.

Y. Guo, H. Liu, J. Chen, Y. Shang, and H. Liu, Synthesis of P(MEO2MA-co-OEGMA) Random Copolymers and Thermally Induced Phase Transition Behaviors in Aqueous Solutions, p.27, 2018.

N. B. Colthup, L. H. Daly, and S. E. Wiberley, Introduction to Infrared and Raman Spectroscopy, 1964.

A. Vigevani, M. Ballabio, E. Gandini, S. Penco, and . Spectra-of-antitumour-anthracyclines, Effect of the Substitution Pattern on the Chemical Shift Values of the Phenolic Protons and on IR Absorptions of the Quinone System, Magn. Reson. Chem, vol.23, pp.344-352, 1985.

L. Guo and W. Ding, Immobilized Transferrin Fe3O4@SiO2 Nanoparticle with High Doxorubicin Loading for Dual-Targeted Tumor Drug Delivery, Int. J. Nanomed, issue.8, p.4631, 2013.

S. Mura, J. Nicolas, and P. Couvreur, Stimuli-Responsive Nanocarriers for Drug Delivery, Nat. Mater, vol.12, pp.991-1003, 2013.