N. Kuznetsova, V. Gretsova, O. Derkacheva, E. Kaliya, and . Lukyanets, Sulfonated phthalocyanines: aggregation and singlet oxygen quantum yield in aqueous solutions, Journal of Porphyrins and Phthalocyanines, vol.67, issue.03, pp.147-154, 2003.
DOI : 10.1111/j.1751-1097.1988.tb02778.x

P. Trindade, N. O-'brien, and . Pickett, Nanocrystalline Semiconductors:?? Synthesis, Properties, and Perspectives, Chemistry of Materials, vol.13, issue.11, pp.3843-3858, 2001.
DOI : 10.1021/cm000843p

A. Torchilin, Tumor delivery of macromolecular drugs based on the EPR effect, Advanced Drug Delivery Reviews, vol.63, issue.3, pp.131-135, 2011.
DOI : 10.1016/j.addr.2010.03.011

P. Lassalle, D. Dumas, S. Grafe, M. A. D-'hallewin, F. Guillemin et al., Correlation between in vivo pharmacokinetics, intratumoral distribution and photodynamic efficiency of liposomal mTHPC, Journal of Controlled Release, vol.134, issue.2, pp.118-124, 2009.
DOI : 10.1016/j.jconrel.2008.11.016

URL : https://hal.archives-ouvertes.fr/hal-00338449

S. Derycke and P. A. De-witte, Liposomes for photodynamic therapy, Advanced Drug Delivery Reviews, vol.56, issue.1, pp.17-30, 2004.
DOI : 10.1016/j.addr.2003.07.014

S. Preuss, M. Hackbarth, T. Wacker, K. Knobloch, B. Langer et al., Comprehensive in vitro investigations on biodegradable photosensitizer-nanoparticle delivery systems, Journal of Controlled Release, vol.148, issue.1, pp.117-118, 2010.
DOI : 10.1016/j.jconrel.2010.07.089

S. Battah, A. Balaratnam, S. O. Casas, C. Neill, A. Edwards et al., Macromolecular delivery of 5-aminolaevulinic acid for photodynamic therapy using dendrimer conjugates, Molecular Cancer Therapeutics, vol.6, issue.3, pp.876-885, 2007.
DOI : 10.1158/1535-7163.MCT-06-0359

V. Synatschke, T. Nomoto, H. Cabral, M. Fortsch, K. Toh et al., Photodynamic Therapy, ACS Nano, vol.8, issue.2, pp.1161-1172, 2014.
DOI : 10.1021/nn4028294

H. Guo, Y. Mao, A. Li, H. Zhu, H. He et al., Dual imaging-guided photothermal/photodynamic therapy using micelles, Biomaterials, vol.35, issue.16, pp.4656-4666, 2014.
DOI : 10.1016/j.biomaterials.2014.02.018

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4568826

O. Gary-bobo, D. Hocine, M. Brevet, L. Maynadier, S. Raehm et al., Cancer therapy improvement with mesoporous silica nanoparticles combining targeting, drug delivery and PDT, International Journal of Pharmaceutics, vol.423, issue.2, pp.509-515, 2012.
DOI : 10.1016/j.ijpharm.2011.11.045

URL : https://hal.archives-ouvertes.fr/hal-00656891

V. Hackbarth, A. Horneffer, F. Wiehe, B. Hillenkamp, and . Röder, Photophysical properties of pheophorbide-a-substituted diaminobutane poly-propylene-imine dendrimer, Chemical Physics, vol.269, issue.1-3, pp.339-346, 2001.
DOI : 10.1016/S0301-0104(01)00337-8

C. Lee, J. A. Mackay, J. M. Frechet, and F. C. Szoka, Designing dendrimers for biological applications, Nature Biotechnology, vol.124, issue.12, pp.1517-1526, 2005.
DOI : 10.1007/3-540-12796-8_9

M. Kaminskas, B. J. Boyd, and C. J. Porter, Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties, Nanomedicine, vol.6, issue.6, pp.1063-1084, 2011.
DOI : 10.2217/nnm.11.67

H. R. Nishiyama, G. D. Stapert, D. Zhang, D. L. Takasu, T. Jiang et al., Light-Harvesting Ionic Dendrimer Porphyrins as New Photosensitizers for Photodynamic Therapy, Bioconjugate Chemistry, vol.14, issue.1, pp.58-66, 2003.
DOI : 10.1021/bc025597h

Y. Kojima, A. Toi, K. Harada, and . Kono, Preparation of Poly(ethylene glycol)-Attached Dendrimers Encapsulating Photosensitizers for Application to Photodynamic Therapy, Bioconjugate Chemistry, vol.18, issue.3, pp.663-670, 2007.
DOI : 10.1021/bc060244u

K. Trester-zedlitz, S. K. Kamada, D. Burley, B. T. Fenyo, T. W. Chait et al., A Modular Cross-Linking Approach for Exploring Protein Interactions, Journal of the American Chemical Society, vol.125, issue.9, pp.2416-2425, 2003.
DOI : 10.1021/ja026917a

M. Sweet, R. B. Kolhatkar, A. Ray, P. Swaan, and H. Ghandehari, Transepithelial transport of PEGylated anionic poly(amidoamine) dendrimers: Implications for oral drug delivery, Journal of Controlled Release, vol.138, issue.1, pp.78-85, 2009.
DOI : 10.1016/j.jconrel.2009.04.022

W. P. Wilkinson, A. B. Helman, and . Ross, Quantum Yields for the Photosensitized Formation of the Lowest Electronically Excited Singlet State of Molecular Oxygen in Solution, Journal of Physical and Chemical Reference Data, vol.22, issue.1, pp.113-262, 1993.
DOI : 10.1063/1.555934

G. Malgady and D. B. Krebs, Understanding Correlation Coefficients and Regression, Physical Therapy, vol.66, issue.1, pp.110-120, 1986.
DOI : 10.1093/ptj/66.1.110

J. Taylor and . Diagn, Interpretation of the Correlation Coefficient: A Basic Review, Journal of Diagnostic Medical Sonography, vol.73, issue.1, pp.35-39, 1990.
DOI : 10.1161/01.CIR.73.1.100

M. I. Megow, M. Rohr, . Schmidt, T. Busch, R. Renger et al., Site-dependence of van der Waals interaction explains exciton spectra of double-walled tubular J-aggregates, Phys. Chem. Chem. Phys., vol.11, issue.10, pp.6741-6747, 2015.
DOI : 10.1002/jcc.540110311

R. Kay, M. Humphry-baker, and . Grätzel, Artificial Photosynthesis. 2. Investigations on the Mechanism of Photosensitization of Nanocrystalline TiO2 Solar Cells by Chlorophyll Derivatives, The Journal of Physical Chemistry, vol.98, issue.3, pp.952-959, 1994.
DOI : 10.1021/j100054a035

G. Li, X. Zhou, G. Zhu, P. Li, L. Yan et al., Photodynamic therapy with hyperbranched poly(ether-ester) chlorin(e6) nanoparticles on human tongue carcinoma CAL-27 cells, Photodiagnosis and Photodynamic Therapy, vol.9, issue.1, pp.76-82, 2012.
DOI : 10.1016/j.pdpdt.2011.08.001

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3292741

K. Park and . Na, Conjugation of the photosensitizer Chlorin e6 to pluronic F127 for enhanced cellular internalization for photodynamic therapy, Biomaterials, vol.34, issue.28, pp.6992-7000, 2013.
DOI : 10.1016/j.biomaterials.2013.05.070

C. Cho, Q. Zhang, and Y. Xia, The effect of sedimentation and diffusion on cellular uptake of gold nanoparticles, Nature Nanotechnology, vol.97, issue.6, pp.385-391, 2011.
DOI : 10.1016/j.colsurfb.2008.02.013

M. Li, W. K. Kroger, and . Liu, Endocytosis of PEGylated nanoparticles accompanied by structural and free energy changes of the grafted polyethylene glycol, Biomaterials, vol.35, issue.30, pp.8467-8478, 2014.
DOI : 10.1016/j.biomaterials.2014.06.032

H. Hatakeyama, H. Akita, and A. Harashima, A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: A strategy for overcoming the PEG dilemma, Advanced Drug Delivery Reviews, vol.63, issue.3, pp.152-160, 2011.
DOI : 10.1016/j.addr.2010.09.001

D. Kessel and Y. Luo, Mitochondrial photodamage and PDT-induced apoptosis, Journal of Photochemistry and Photobiology B: Biology, vol.42, issue.2, pp.89-95, 1998.
DOI : 10.1016/S1011-1344(97)00127-9

H. Teiten, L. Bezdetnaya, P. Morliere, R. Santus, and F. Guillemin, Endoplasmic reticulum and Golgi apparatus are the preferential sites of Foscan?? localisation in cultured tumour cells, British Journal of Cancer, vol.88, issue.1, pp.146-152, 2003.
DOI : 10.1038/sj.bjc.6600664

H. Schmidt-erfurth, R. Diddens, T. Birngruber, and . Hasan, Photodynamic targeting of human retinoblastoma cells using covalent low-density lipoprotein conjugates, British Journal of Cancer, vol.75, issue.1, pp.54-61, 1997.
DOI : 10.1038/bjc.1997.9

M. S. Soukos, T. Hamblin, and . Hasan, Conjugates, Photochemistry and Photobiology, vol.48, issue.4, pp.723-729, 1997.
DOI : 10.1080/09553009014551501

D. Spikes and J. , New trends in photobiology, Journal of Photochemistry and Photobiology B: Biology, vol.6, issue.3, pp.259-274, 1990.
DOI : 10.1016/1011-1344(90)85096-F

G. Feldman, N. Hahner, P. Spencer, M. Harder, and . Grunze, Probing Resistance to Protein Adsorption of Oligo(ethylene glycol)-Terminated Self-Assembled Monolayers by Scanning Force Microscopy, Journal of the American Chemical Society, vol.121, issue.43, pp.10134-10141, 1999.
DOI : 10.1021/ja991049b

B. Yuan, J. Yang, Y. Wu, X. Hu, and . Ming, Dendritic nanoconjugates of photosensitizer for targeted photodynamic therapy, Acta Biomaterialia, vol.21, pp.63-73, 2015.
DOI : 10.1016/j.actbio.2015.04.014

J. Shcharbin, M. Mazurb, M. Szwedzka, B. Wasiak, M. Palecz et al., Interaction between PAMAM 4.5 dendrimer, cadmium and bovine serum albumin: A study using equilibrium dialysis, isothermal titration calorimetry, zeta-potential and fluorescence, Colloids and Surfaces B: Biointerfaces, vol.58, issue.2, pp.286-289, 2007.
DOI : 10.1016/j.colsurfb.2007.04.003

J. Lundqvist, G. Stigler, I. Elia, T. Lynch, K. A. Cedervall 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

L. Perry, K. G. Reuter, M. P. Kai, K. P. Herlihy, S. W. 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

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, Journal of the American Chemical Society, vol.134, issue.4, pp.2139-2147, 2011.
DOI : 10.1021/ja2084338

P. Perumal, R. Inapagolla, S. Kannan, and R. M. Kannan, The effect of surface functionality on cellular trafficking of dendrimers, Biomaterials, vol.29, issue.24-25, pp.3469-3476, 2008.
DOI : 10.1016/j.biomaterials.2008.04.038

D. Trono, K. Mizuno, N. Yusa, T. Matsukawa, K. Yokoyama et al., Size, Concentration and Incubation Time Dependence of Gold Nanoparticle Uptake into Pancreas Cancer Cells and its Future Application to X-ray Drug Delivery System, Journal of Radiation Research, vol.52, issue.1, pp.103-109, 2011.
DOI : 10.1269/jrr.10068

R. Maxfield and T. E. Mcgraw, Endocytic recycling, Nature Reviews Molecular Cell Biology, vol.5, issue.2, pp.121-132, 2004.
DOI : 10.1038/nrm1315

G. Parton and K. Simons, The multiple faces of caveolae, Nature Reviews Molecular Cell Biology, vol.278, issue.3, pp.185-194, 2007.
DOI : 10.1212/WNL.62.4.538

T. Kress, R. Meier, F. Steiner, R. Dolp, U. Erdmann et al., Time-resolved microspectrofluorometry and fluorescence lifetime imaging of photosensitizers using picosecond pulsed diode lasers in laser scanning microscopes, Journal of Biomedical Optics, vol.8, issue.1, pp.26-32, 2003.
DOI : 10.1117/1.1528595

]. T. Dougherty, C. J. Gomer, B. W. Henderson, G. Jori, D. Kessel et al., Photodynamic therapy The Photodynamic Properties of a Particular Hematoporphyrin Derivative Further Evaluation of the Use of Hematoporphyrin Derivative as a New Aid for the Endoscopic Detection of Malignant Disease Photoradiation Therapy for Treatment of Malignant-Tumors An update on photodynamic therapy applications Photodynamic therapy for localized infections-State of the art Photodynamic Nanomedicine in the Treatment of Solid Tumors: Perspectives and Challenges The cost-effectiveness of Foscan mediated photodynamic therapy (Foscan-PDT) compared with extensive palliative surgery and palliative chemotherapy for patients with advanced head and neck cancer in the UK, J. Natl. Cancer Inst. Arch. Dermatol. Dis. Chest Cancer Res. J. Clin. Laser Med. Surg. Photodiagnosis Photodyn. Ther. J. Control. Release Off. J. Control. Release Soc. Oral Oncol, vol.90, issue.40 4, pp.889-905, 1960.

J. Wilson, . M. Golab10-]-w, C. M. Sharman, J. E. Allen, and . Van-lier, Photodynamic therapy of cancer: An update Role of activated oxygen species in photodynamic therapy, Singlet Oxygen, Uv-a, and Ozone Antioxidant defence mechanisms: from the beginning to the end, pp.250-281, 2000.

]. J. Van-lier, J. D. Spikes13-]-n, R. L. Oleinick, T. Morris, and . Belichenko, The chemistry, photophysics and photosensitizing properties of phthalocyanines The role of apoptosis in response to photodynamic therapy: what, where, why, and how Photodynamic therapy for cancer Intracellular signaling mechanisms in photodynamic therapy, Free Radic. Res. Ciba Found. Symp. Photochem. Photobiol. Sci. Nat. Rev. Cancer Biochim. Biophys. Acta BBA -Rev. Cancer, vol.3115, issue.1704 2, pp.261-272, 1989.

]. S. Marchal, L. Bezdetnaya, F. Guillemin, S. Marchal, A. Fadloun et al., Modality of cell death induced by Foscan-based photodynamic treatment in human colon adenocarcinoma cell line HT29 Necrotic and apoptotic features of cell death in response to Foscan® photosensitization of HT29 monolayer and multicell spheroids Signaling pathways in cell death and survival after photodynamic therapy, Biochem. Biokhimii?a Biochem. Pharmacol. J, vol.6917, issue.69 8, pp.45-49, 2004.

]. Q. Peng, J. Moan, J. M. Nesland, J. Usuda, S. Chiu et al., Correlation of subcellular and intratumoral photosensitizer localization with ultrastructural features after photodynamic therapy Domain-dependent Photodamage to Bcl-2 a membrane anchorage region is needed to form the target of phtalocyanine photosensitization, Relationship between subcellular localisation of Foscan® and caspase activation in photosensitised MCF-7 cells, pp.1-13, 1996.

]. S. Bonneau and C. Vever-bizet, Tetrapyrrole photosensitisers, determinants of subcellular localisation and mechanisms of photodynamic processes in therapeutic approaches, Expert Opinion on Therapeutic Patents, vol.58, issue.9, pp.944-951, 2007.
DOI : 10.1002/ijc.2910580620

. P. Ther, R. Kessel, M. G. Luguya, P. Maes, . J. Agostinis-]-j et al., ROS-mediated mechanisms of autophagy stimulation and their relevance in cancer therapy Assessing autophagy in the context of photodynamic therapy Destruction of rat mammary tumor and normal tissue microcirculation by hematoporphyrin derivative photoradiation observed in vivo in sandwich observation chambers Photodynamic Therapy of Tumors Can Lead to Development of Systemic Antigen-Specific Immune Response Photodynamic therapy and anti-tumour immunity Photodynamic therapy enhancement of anti-tumor immunity Photodynamic therapy for enhancing antitumour immunity, Localization and Photodynamic Efficacy of Two Cationic Porphyrins Varying in Charge Distribution Direct and indirect photodynamic therapy effects on the cellular and molecular components of the tumor microenvironment. [32] M. Korbelik and I. Cecic Contribution of myeloid and lymphoid host cells to the curative outcome of mouse sarcoma treatment by photodynamic therapy, pp.1011-1025, 1986.

P. C. Kousis, B. W. Henderson, P. G. Maier, S. O. Gollnick, M. Korbelik et al., The Role of Host Lymphoid Populations in the Response of Mouse EMT6 Tumor to Photodynamic Therapy The physics, biophysics and technology of photodynamic therapy State of the art in the delivery of photosensitizers for photodynamic therapy Effect of Sulfonation on the Cell and Tissue Distribution of the Photosensitizer Aluminum Phthalocyanine Dynamics of interactions of photosensitizers with lipoproteins and membrane-models: correlation with cellular incorporation and subcellular distribution Acid Ph in Tumors and Its Potential for Therapeutic Exploitation Acid?base properties of chlorin e 6: relation to cellular uptake Structure and Biodistribution Relationships of Photodynamic Sensitizers*, Photodynamic Therapy Enhancement of Antitumor Immunity Is Regulated by Neutrophils40] B. Cunderl??kováCunderl??ková, L. Gangeskar, and J. Moan Photoluminescence of Singlet Oxygen in Pigment Solutions, pp.10501-10510, 1989.

H. Photobiol, S. Mojzisova, C. Bonneau, D. Vever-bizet, E. Brault et al., Cellular uptake and subcellular distribution of chlorin e6 as functions of pH and interactions with membranes and lipoproteins Photophysical and photochemical properties of potential porphyrin and chlorin photosensitizers for PDT Photophysical Properties of Novel PDT Photosensitizer Radachlorin in Different Media Photosensitizer Radachlorin®: Skin cancer PDT phase II clinical trials Photodynamic therapy of early esophageal cancer In vitro and in vivo evaluation of Radachlorin® sensitizer for photodynamic therapy In vitro evaluation of Radachlorin® sensitizer for photodynamic therapy, Modulation of EGFR and ROS induced cytochrome c release by combination of photodynamic therapy and carboplatin in human cultured head and neck cancer cells and tumor xenograft in nude mice, pp.29-36, 1979.

S. Bae, Y. Kim, J. Lee, S. Namkoong, S. Han et al., The effect of Radachlorin® PDT in advanced NSCLC: A pilot study Efficacy and Safety of Photodynamic Therapy for Recurrent, High Grade Nonmuscle Invasive Bladder Cancer Refractory or Intolerant to Bacille Calmette-Guérin Immunotherapy The efficacy of Radachlorin-mediated photodynamic therapy in human hepatocellular carcinoma cells, 54] H. Mirzaei, G. E. Djavid, M. Hadizadeh, M. Jahanshiri-Moghadam, and P. Hajian, pp.389-394, 2004.

L. E. Gerweck, K. Seetharaman, Z. Li, C. Wang, L. Cheng et al., Cellular pH gradient in tumor versus normal tissue: Potential exploitation for the treatment of cancer Conjugation of the photosensitizer Chlorin e6 to pluronic F127 for enhanced cellular internalization for photodynamic therapy PEG-functionalized iron oxide nanoclusters loaded with chlorin e6 for targeted, NIR light induced, photodynamic therapy Photosensitizer-conjugated silica-coated gold nanoclusters for fluorescence imagingguided photodynamic therapy Physical mechanisms of generation and deactivation of singlet oxygen Direct measurement of the lifetime of 1? oxygen in solution Singlet oxygen-mediated damage to proteins and its consequences, Molecular Spectra and Molecular Structure. Volume I: Spectra of Diatomic Molecules. Second Edition, pp.86-91, 1950.

. Biophys, . Res, A. Commun, J. R. Baker, F. Kanofsky et al., Quenching of Singlet Oxygen by Biomolecules from L1210 Leukemia Cells Rate Constants for the Decay and Reactions of the Lowest Electronically Excited Singlet-State of Molecular-Oxygen in Solution -an Expanded and Revised Compilation Chain-Reaction Wheel -an Approach to Free-Radical Reactions, Photochem. Photobiol. J. Phys. Chem. Ref. Data J, vol.30564, issue.24 2, pp.761-770, 1992.

H. Mojzisova, S. Bonneau, P. Maillard, K. Berg, and D. Brault, Photosensitizing properties of chlorins in solution and in membrane-mimicking systems, Verteporfin, photofrin II, and merocyanine 540 as PDT photosensitizers against melanoma cells, pp.351-371, 2002.
DOI : 10.1042/bj2610277

URL : https://hal.archives-ouvertes.fr/hal-00394568

. Biophys, . Res, H. Commun, Q. Wu, G. Song et al., Recent developments in the detection of singlet oxygen with molecular spectroscopic methods Singlet Oxygen Detection During Photosensitization Fluorescent and luminescent probes for detection of reactive oxygen and nitrogen species Rational design of fluorescein-based fluorescence probes, mechanism-based design of a maximum fluorescence probe for singlet oxygen Novel Fluorescent Probes for Singlet Oxygen Mechanisms of Bacteriophage Inactivation via Singlet Oxygen Generation in UV Illuminated Fullerol Suspensions Photodegradation of 5- methyltetrahydrofolate in the presence of Uroporphyrin Indirect imaging of singlet oxygen generation from a single cell, Monitoring Singlet Oxygen and Hydroxyl Radical Formation with Fluorescent Probes During Photodynamic Therapy, pp.549-555, 1999.

P. Zhang, K. Aslan, M. J. Previte, C. D. Geddes, S. Nonell et al., Plasmonic engineering of singlet oxygen generation, Proceedings of the National Academy of Sciences, vol.50, issue.6, pp.662-666, 2008.
DOI : 10.1109/TAP.2002.804571

N. R. Wilson and . Baker, Imaging the production of singlet oxygen in vivo using a new fluorescent sensor, Singlet Oxygen Sensor Green (R) A comparative study of fluorescent singlet oxygen probes in plant leaves, J. Exp. Bot, vol.57, issue.8, pp.1725-1734, 2006.

H. Chem, Y. Lin, D. Shen, L. Chen, B. C. Lin et al., Singlet Oxygen Imaging in Polymeric Nanofibers by Delayed Fluorescence FCLA chemiluminescence from sonodynamic action in vitro and in vivo Mechanism of photosensitized chemiluminescence of 2-methyl-6-phenylimidazo[1,2-a]pyrazin-3(7H)-one (CLA) in aqueous solution Evaluation of the degree of medical radiation damage with a highly sensitive chemiluminescence method Theoretical and experimental analysis of the luminescence signal of singlet oxygen for different photosensitizers [Photosensitized luminescence of singlet oxygen in solution] Observation of collision-induced near-IR emission of singlet oxygen O2 a1?g generated by visible light excitation of gaseous O2 dimol Time-resolved detection of singlet oxygen luminescence in red cell ghost suspensions Time-resolved detection of singlet oxygen luminescence in red-cell ghost suspensions: concerning a signal component that can be attributed to O-1(2) luminescence from the inside of a native membrane New insights to primary photodynamic effects -Singlet oxygen kinetics in living cells Singlet oxygen production by lipophilic photosensitizers in liposomes studies by time and spectral resolved phosphorescence Spectroscopic study of singlet oxygen photogeneration by lipophilic photosensitiser in liposomes, Feasibility Study on Quantitative Measurements of Singlet Oxygen Generation Using Singlet Oxygen Sensor Green. [98] A. Molnar, R. Dedic, A. Svoboda, and J. Hala. [99] L. M. Andersen, Z. Gao, P. R. Ogilby, L. Poulsen, and I. Zebger, " A singlet oxygen image with 2, pp.161-165, 1976.

I. Zebger, L. Poulsen, Z. Gao, L. K. Andersen, and P. R. Ogilby, Singlet Oxygen Images of Heterogeneous Samples:?? Examining the Effect of Singlet Oxygen Diffusion across the Interfacial Boundary in Phase-Separated Liquids and Polymers, Langmuir, vol.19, issue.21, pp.8927-8933, 2003.
DOI : 10.1021/la0301487

J. Schlothauer, S. Hackbarth, and B. Roeder, A new benchmark for time-resolved detection of singlet oxygen luminescence - revealing the evolution of lifetime in living cells with low dose illumination, Laser Physics Letters, vol.6, issue.3, pp.216-221, 2009.
DOI : 10.1002/lapl.200810116

S. Okazaki, T. Tomo, and M. Mimuro, Direct measurement of singlet oxygen produced by four chlorin-ringed chlorophyll species in acetone solution, Chemical Physics Letters, vol.485, issue.1-3, pp.1-3, 2010.
DOI : 10.1016/j.cplett.2009.12.055

S. Hackbarth, J. Schlothauer, A. Preuss, and B. Roeder, Highly sensitive time resolved singlet oxygen luminescence detection using LEDs as the excitation source, Laser Physics Letters, vol.10, issue.12, p.125702, 2013.
DOI : 10.1088/1612-2011/10/12/125702

M. Niedre, M. S. Patterson, and B. C. Wilson, Direct Near-infrared Luminescence Detection of Singlet Oxygen Generated by Photodynamic Therapy in Cells In Vitro and Tissues In Vivo??, Photochemistry and Photobiology, vol.75, issue.4, pp.382-391, 2002.
DOI : 10.1562/0031-8655(2002)0750382DNILDO2.0.CO2

J. C. Schlothauer, J. Falckenhayn, T. Perna, S. Hackbarth, and B. Roeder, Luminescence investigation of photosensitizer distribution in skin: correlation of singlet oxygen kinetics with the microarchitecture of the epidermis, Journal of Biomedical Optics, vol.18, issue.11, p.115001, 2013.
DOI : 10.1117/1.JBO.18.11.115001

S. Hackbarth and B. Roeder, Singlet oxygen luminescence kinetics in a heterogeneous environment -identification of the photosensitizer localization in small unilamellar vesicles Photochem

L. Lin, H. Lin, D. Chen, L. Chen, M. Wang et al., Direct imaging of singlet oxygen luminescence generated in blood vessels during photodynamic therapy, Biophotonics Photonic Solut. Better Health Care Iv, vol.9129, p.912920, 2014.

B. C. Wilson, M. S. Patterson, B. Li, and M. T. Jarvi, tumor response and singlet oxygen luminescence detection in mTHPC-mediated photodynamic therapy, Journal of Innovative Optical Health Sciences, vol.92, issue.01
DOI : 10.1111/j.1464-410X.2008.07753.x

A. Wicki, D. Witzigmann, V. Balasubramanian, and J. Huwyler, Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications, Journal of Controlled Release, vol.200, pp.138-157, 2015.
DOI : 10.1016/j.jconrel.2014.12.030

E. Pérez-herrero and A. Fernández-medarde, Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy, European Journal of Pharmaceutics and Biopharmaceutics, vol.93, pp.52-79, 2015.
DOI : 10.1016/j.ejpb.2015.03.018

R. Solomon and A. A. Gabizon, Clinical Pharmacology of Liposomal Anthracyclines: Focus on Pegylated Liposomal Doxorubicin, Clinical Lymphoma and Myeloma, vol.8, issue.1, pp.21-32, 2008.
DOI : 10.3816/CLM.2008.n.001

A. N. Fader and P. G. Rose, Abraxane for the Treatment of Gynecologic Cancer Patients With Severe Hypersensitivity Reactions to Paclitaxel, International Journal of Gynecological Cancer, vol.17, issue.7, pp.1281-1283, 2009.
DOI : 10.1111/IGC.0b013e3181a38e2f

T. Stylianopoulos and R. K. Jain, Design considerations for nanotherapeutics in oncology, Nanomedicine: Nanotechnology, Biology and Medicine, vol.11, issue.8, 2015.
DOI : 10.1016/j.nano.2015.07.015

S. Karve, M. E. Werner, R. Sukumar, N. D. Cummings, J. A. Copp et al., Revival of the abandoned therapeutic wortmannin by nanoparticle drug delivery, Proc. Natl. Acad. Sci, pp.8230-8235, 2012.
DOI : 10.1101/gad.1015202

S. Thangavel, T. Yoshitomi, M. K. Sakharkar, and Y. Nagasaki, Redox nanoparticles inhibit curcumin oxidative degradation and enhance its therapeutic effect on prostate cancer, Journal of Controlled Release, vol.209, pp.110-119, 2015.
DOI : 10.1016/j.jconrel.2015.04.025

I. Levacheva, O. Samsonova, E. Tazina, M. Beck-broichsitter, S. Levachev et al., Optimized thermosensitive liposomes for selective doxorubicin delivery: Formulation development, quality analysis and bioactivity proof, Colloids and Surfaces B: Biointerfaces, vol.121, pp.248-256, 2014.
DOI : 10.1016/j.colsurfb.2014.02.028

Y. Li, J. Wang, M. G. Wientjes, and J. L. Au, Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor, Advanced Drug Delivery Reviews, vol.64, issue.1, pp.29-39
DOI : 10.1016/j.addr.2011.04.006

T. Lammers, F. Kiessling, W. E. 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

Z. Popovic, W. Liu, V. P. Chauhan, J. Lee, C. Wong et al., A Nanoparticle Size Series for In???Vivo Fluorescence Imaging, Angewandte Chemie International Edition, vol.25, issue.46, pp.8649-8652, 2010.
DOI : 10.1002/anie.201003142

J. Kim, H. Yoon, J. Sim, S. Ju, and W. Jang, The effects of dendrimer size and central metal ions on photosensitizing properties of dendrimer porphyrins, Journal of Drug Targeting, vol.3, issue.7, pp.610-618, 2014.
DOI : 10.1007/BF00613067

H. Jeong, M. Huh, S. J. Lee, H. Koo, I. C. Kwon et al., Photosensitizer-Conjugated Human Serum Albumin Nanoparticles for Effective Photodynamic Therapy, Theranostics, vol.1, p.230, 2011.
DOI : 10.7150/thno/v01p0230

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3092446/pdf

P. Skupin-mrugalska, J. Piskorz, T. Goslinski, J. Mielcarek, K. Konopka et al., Current status of liposomal porphyrinoid photosensitizers, Drug Discovery Today, vol.18, issue.15-16, pp.15-16, 2013.
DOI : 10.1016/j.drudis.2013.04.003

C. Wong, T. Stylianopoulos, J. Cui, J. Martin, V. P. Chauhan et al., Multistage nanoparticle delivery system for deep penetration into tumor tissue, Proceedings of the National Academy of Sciences, vol.7, issue.7, pp.2426-2431, 2011.
DOI : 10.1038/89997

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3038705

U. Prabhakar, H. Maeda, R. K. Jain, E. M. Sevick-muraca, W. Zamboni et al., Challenges and Key Considerations of the Enhanced Permeability and Retention Effect for Nanomedicine Drug Delivery in Oncology, Cancer Research, vol.73, issue.8, pp.2412-2417, 2013.
DOI : 10.1158/0008-5472.CAN-12-4561

D. Peer, J. M. Karp, S. Hong, O. C. 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

H. Tang, X. Chen, M. Rui, W. Sun, J. Chen et al., Effects of Surface Displayed Targeting Ligand GE11 on Liposome Distribution and Extravasation in Tumor, Molecular Pharmaceutics, vol.11, issue.10, pp.3242-3250, 2014.
DOI : 10.1021/mp5001718

N. Oh and J. Park, Endocytosis and exocytosis of nanoparticles in mammalian cells, Int. J. Nanomedicine, p.51, 2014.

G. J. Doherty and H. T. Mcmahon, Mechanisms of Endocytosis, Annual Review of Biochemistry, vol.78, issue.1, pp.857-902, 2009.
DOI : 10.1146/annurev.biochem.78.081307.110540

I. Canton and G. Battaglia, Endocytosis at the nanoscale, Chemical Society Reviews, vol.26, issue.1, pp.2718-2739, 2012.
DOI : 10.1016/j.jconrel.2008.10.020

Z. Wang, C. Tiruppathi, R. D. Minshall, and A. B. Malik, Size and Dynamics of Caveolae Studied Using Nanoparticles in Living Endothelial Cells, ACS Nano, vol.3, issue.12, pp.4110-4116, 2009.
DOI : 10.1021/nn9012274

L. Pelkmans, J. Kartenbeck, and A. Helenius, Caveolar endocytosis of simian virus 40 reveals a new two-step vesicular-transport pathway to the ER, Nature Cell Biology, vol.3, issue.5, pp.473-483, 2001.
DOI : 10.1038/35074539

A. Yuan, B. Yang, J. Wu, Y. Hu, and X. Ming, Dendritic nanoconjugates of photosensitizer for targeted photodynamic therapy, Acta Biomaterialia, vol.21, pp.63-73, 2015.
DOI : 10.1016/j.actbio.2015.04.014

T. Okamoto, A. Schlegel, P. E. Scherer, and M. P. Lisanti, Caveolins, a Family of Scaffolding Proteins for Organizing ???Preassembled Signaling Complexes??? at the Plasma Membrane, Journal of Biological Chemistry, vol.87, issue.10
DOI : 10.1074/jbc.271.51.32975

S. D. Conner and S. L. Schmid, Regulated portals of entry into the cell, Nature, vol.277, issue.6927, pp.37-44, 2003.
DOI : 10.1091/mbc.12.9.2578

T. Iversen, T. Skotland, and K. Sandvig, Endocytosis and intracellular transport of nanoparticles: Present knowledge and need for future studies, Nano Today, vol.6, issue.2, pp.176-185, 2011.
DOI : 10.1016/j.nantod.2011.02.003

K. Sandvig, M. L. Torgersen, H. A. Raa, and B. Van-deurs, Clathrin-independent endocytosis: from nonexisting to an extreme degree of complexity, Histochemistry and Cell Biology, vol.13, issue.3, pp.267-276, 2008.
DOI : 10.1091/mbc.11.1.325

S. Grimmer, B. Van-deurs, and K. Sandvig, Membrane ruffling and macropinocytosis in A431 cells require cholesterol, J. Cell Sci, vol.115, pp.2953-2962, 2002.

S. K. Rodal, G. Skretting, O. Garred, F. Vilhardt, B. Van-deurs et al., Extraction of Cholesterol with Methyl-beta -Cyclodextrin Perturbs Formation of Clathrin-coated Endocytic Vesicles, Molecular Biology of the Cell, vol.10, issue.4
DOI : 10.1091/mbc.10.4.961

D. Vercauteren, R. E. Vandenbroucke, A. T. Jones, J. Rejman, J. Demeester et al., The Use of Inhibitors to Study Endocytic Pathways of Gene Carriers: Optimization and Pitfalls, Molecular Therapy, vol.18, issue.3, pp.561-569, 2010.
DOI : 10.1038/mt.2009.281

H. Damke, T. Baba, A. Vanderbliek, and S. Schmid, Clathrin-independent pinocytosis is induced in cells overexpressing a temperature-sensitive mutant of dynamin, The Journal of Cell Biology, vol.131, issue.1, pp.69-80, 1995.
DOI : 10.1083/jcb.131.1.69

D. Bechet, P. Couleaud, C. Frochot, M. Viriot, F. Guillemin et al., Nanoparticles as vehicles for delivery of photodynamic therapy agents, Trends in Biotechnology, vol.26, issue.11, pp.612-621, 2008.
DOI : 10.1016/j.tibtech.2008.07.007

URL : https://hal.archives-ouvertes.fr/hal-00323569

R. Bachor, C. R. Shea, R. Gillies, and T. Hasan, Photosensitized destruction of human bladder carcinoma cells treated with chlorin e6-conjugated microspheres., Proc. Natl. Acad. Sci, pp.1580-1584, 1991.
DOI : 10.1073/pnas.88.4.1580

G. Shim, S. Lee, Y. B. Kim, C. Kim, and Y. Oh, Enhanced tumor localization and retention of chlorin e6 in cationic nanolipoplexes potentiate the tumor ablation effects of photodynamic therapy, Nanotechnology, vol.22, issue.36, p.365101, 2011.
DOI : 10.1088/0957-4484/22/36/365101

A. Yuan, X. Tang, X. Qiu, K. Jiang, J. Wu et al., Activatable photodynamic destruction of cancer cells by NIR dye/photosensitizer loaded liposomes, Chem. Commun., vol.7, issue.16, pp.3340-3342, 2015.
DOI : 10.1021/nn403202w

B. Tian, C. Wang, S. Zhang, L. Feng, and Z. Liu, Photothermally Enhanced Photodynamic Therapy Delivered by Nano-Graphene Oxide, ACS Nano, vol.5, issue.9, pp.7000-7009, 2011.
DOI : 10.1021/nn201560b

S. Y. Park, H. J. Baik, Y. T. Oh, K. T. Oh, Y. S. Youn et al., A Smart Polysaccharide/Drug Conjugate for Photodynamic Therapy, Angewandte Chemie International Edition, vol.61, issue.7, pp.1644-1647, 2011.
DOI : 10.1211/jpp.61.06.0003

S. Kimani, G. Ghosh, A. Ghogare, B. Rudshteyn, D. Bartusik et al., Synthesis and Characterization of Mono-, Di-, and Tri-Polyethylene Glycol Chlorin e6 Conjugates for the Photokilling of Human Ovarian Cancer Cells, J. Org. Chem, vol.77, issue.23, 2012.

W. Chin, P. Heng, P. Thong, R. Bhuvaneswari, W. Hirt et al., Improved formulation of photosensitizer chlorin e6 polyvinylpyrrolidone for fluorescence diagnostic imaging and photodynamic therapy of human cancer, European Journal of Pharmaceutics and Biopharmaceutics, vol.69, issue.3, pp.1083-1093, 2008.
DOI : 10.1016/j.ejpb.2008.02.013

W. W. Chin, T. Praveen, P. W. Heng, and M. Olivo, Effect of polyvinylpyrrolidone on the interaction of chlorin e6 with plasma proteins and its subcellular localization, European Journal of Pharmaceutics and Biopharmaceutics, vol.76, issue.2, pp.245-252, 2010.
DOI : 10.1016/j.ejpb.2010.06.005

S. Kruspe, C. Meyer, and U. Hahn, Chlorin e6 Conjugated Interleukin-6 Receptor Aptamers Selectively Kill Target Cells Upon Irradiation, Molecular Therapy - Nucleic Acids, vol.3, issue.1, p.143, 2014.
DOI : 10.1038/mtna.2013.70

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3910004

P. Huang, Z. Li, J. Lin, D. Yang, G. Gao et al., Photosensitizer-conjugated magnetic nanoparticles for in vivo simultaneous magnetofluorescent imaging and targeting therapy, Biomaterials, vol.32, issue.13, pp.3447-3458, 2011.
DOI : 10.1016/j.biomaterials.2011.01.032

J. Wang, G. Zhu, M. You, E. Song, M. I. Shukoor et al., Assembly of Aptamer Switch Probes and Photosensitizer on Gold Nanorods for Targeted Photothermal and Photodynamic Cancer Therapy, ACS Nano, vol.6, issue.6, pp.5070-5077, 2012.
DOI : 10.1021/nn300694v

J. Kim, W. I. Choi, M. Kim, and G. Tae, Tumor-targeting nanogel that can function independently for both photodynamic and photothermal therapy and its synergy from the procedure of PDT followed by PTT, Journal of Controlled Release, vol.171, issue.2, pp.113-121, 2013.
DOI : 10.1016/j.jconrel.2013.07.006

Z. Zhao, S. Shi, Y. Huang, S. Tang, and X. Chen, Simultaneous Photodynamic and Photothermal Therapy Using Photosensitizer-Functionalized Pd Nanosheets by Single Continuous Wave Laser, ACS Applied Materials & Interfaces, vol.6, issue.11
DOI : 10.1021/am501608c

D. Zhang, M. Wu, Y. Zeng, L. Wu, Q. Wang et al., Chlorin e6 Conjugated Poly(dopamine) Nanospheres as PDT/PTT Dual-Modal Therapeutic Agents for Enhanced Cancer Therapy, ACS Applied Materials & Interfaces, vol.7, issue.15, pp.8176-8187, 2015.
DOI : 10.1021/acsami.5b01027

L. Lin, Z. Cong, J. Cao, K. Ke, Q. Peng et al., @Polydopamine Core???Shell Nanocomposites for Intracellular mRNA Detection and Imaging-Guided Photothermal Therapy, ACS Nano, vol.8, issue.4, pp.3876-3883, 2014.
DOI : 10.1021/nn500722y

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4564054

M. Wu, D. Zhang, Y. Zeng, L. Wu, X. Liu et al., Nanocluster of superparamagnetic iron oxide nanoparticles coated with poly (dopamine) for magnetic field-targeting, highly sensitive MRI and photothermal cancer therapy, Nanotechnology, vol.26, issue.11, p.115102, 2015.
DOI : 10.1088/0957-4484/26/11/115102

P. Huang, J. Lin, X. Wang, Z. Wang, C. Zhang et al., Light-Triggered Theranostics Based on Photosensitizer-Conjugated Carbon Dots for Simultaneous Enhanced-Fluorescence Imaging and Photodynamic Therapy, Advanced Materials, vol.130, issue.37, pp.5104-5110, 2012.
DOI : 10.1021/ja802913f

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3657566

Y. I. Park, H. M. Kim, J. H. Kim, K. C. Moon, B. Yoo et al., Theranostic Probe Based on Lanthanide-Doped Nanoparticles for Simultaneous In Vivo Dual-Modal Imaging and Photodynamic Therapy, Theranostic Probe Based on Lanthanide-Doped Nanoparticles for Simultaneous In Vivo Dual- Modal Imaging and Photodynamic Therapy, pp.5755-5761, 2012.
DOI : 10.1038/nnano.2011.95

X. Wang, K. Liu, G. Yang, L. Cheng, L. He et al., Near-infrared light triggered photodynamic therapy in combination with gene therapy using upconversion nanoparticles for effective cancer cell killing, Nanoscale, vol.8, issue.15, p.9198, 2014.
DOI : 10.1021/nn406197c

P. A. Wooster, M. R. Futreal, and . Stratton, Patterns of somatic mutation in human cancer genomes, Nature, vol.446, issue.7132, pp.153-158, 2007.

H. Park, K. E. Bachman, N. Papadopoulos, B. Vogelstein, K. W. Kinzler et al., The consensus coding sequences of human breast and colorectal cancers, Science, vol.314, issue.5797, pp.268-274, 2006.

L. D. Wood, D. W. Parsons, S. Jones, J. Lin, T. Sjoblom et al., The Genomic Landscapes of Human Breast and Colorectal Cancers, Kinzler, V. E. Velculescu, and B. Vogelstein, pp.1108-1113, 2007.
DOI : 10.1002/jcc.20084

E. Buhleier, W. Wehner, and F. Vogtle, "Cascade"- and "Nonskid-Chain-like" Syntheses of Molecular Cavity Topologies, Synthesis, vol.1978, issue.02, pp.155-158, 1978.
DOI : 10.1055/s-1978-24702

J. Wolinsky and M. Grinstaff, Therapeutic and diagnostic applications of dendrimers for cancer treatment???, Advanced Drug Delivery Reviews, vol.60, issue.9, pp.1037-1055, 2008.
DOI : 10.1016/j.addr.2008.02.012

A. Caminade, R. Laurent, and J. Majoral, Characterization of dendrimers, Advanced Drug Delivery Reviews, vol.57, issue.15, pp.2130-2146, 2005.
DOI : 10.1016/j.addr.2005.09.011

U. Boas and P. M. Heegaard, Dendrimers in drug research, Chemical Society Reviews, vol.33, issue.1, p.43, 2004.
DOI : 10.1039/b309043b

R. Duncan and L. Izzo, Dendrimer biocompatibility and toxicity, Advanced Drug Delivery Reviews, vol.57, issue.15, pp.2215-2237, 2005.
DOI : 10.1016/j.addr.2005.09.019

D. A. Tomalia, Starburstr?? dendrimers - Nanoscopic supermolecules according to dendritic rules and principles, Macromolecular Symposia, vol.40, issue.1, pp.243-255, 1996.
DOI : 10.1021/bc00005a001

C. Hawker and J. Frechet, Preparation of polymers with controlled molecular architecture. A new convergent approach to dendritic macromolecules, Journal of the American Chemical Society, vol.112, issue.21, pp.7638-7647, 1990.
DOI : 10.1021/ja00177a027

K. J. Narendra and K. T. Rakesh, Dendrimers for enhanced drug solubilization, " in Drug delivery strategies for poorly water-soluble drugs, Advances in pharmaceutical technology, p.37, 2013.

L. Wu, M. Ficker, J. B. Christensen, P. N. Trohopoulos, and S. M. Moghimi, Dendrimers in Medicine: Therapeutic Concepts and Pharmaceutical Challenges, Bioconjugate Chemistry, vol.26, issue.7, p.150213085013008, 2015.
DOI : 10.1021/acs.bioconjchem.5b00031

D. Fischer, Y. Li, B. Ahlemeyer, J. Krieglstein, and T. Kissel, In vitro cytotoxicity testing of polycations: influence of polymer structure on cell viability and hemolysis, Biomaterials, vol.24, issue.7, pp.1121-1131, 2003.
DOI : 10.1016/S0142-9612(02)00445-3

R. Jevprasesphant, J. Penny, R. Jalal, D. Attwood, N. B. Mckeown et al., The influence of surface modification on the cytotoxicity of PAMAM dendrimers, International Journal of Pharmaceutics, vol.252, issue.1-2, pp.263-266, 2003.
DOI : 10.1016/S0378-5173(02)00623-3

W. Wang, W. Xiong, J. Wan, X. Sun, H. Xu et al., The decrease of PAMAM dendrimer-induced cytotoxicity by PEGylation via attenuation of oxidative stress, Nanotechnology, vol.20, issue.10, p.105103, 2009.
DOI : 10.1088/0957-4484/20/10/105103

M. Ciolkowski, J. F. Petersen, M. Ficker, A. Janaszewska, J. B. Christensen et al., Surface modification of PAMAM dendrimer improves its biocompatibility, Nanomedicine: Nanotechnology, Biology and Medicine, vol.8, issue.6
DOI : 10.1016/j.nano.2012.03.009

I. J. Majoros, T. P. Thomas, C. B. Mehta, and J. R. Baker, Poly(amidoamine) Dendrimer-Based Multifunctional Engineered Nanodevice for Cancer Therapy, Journal of Medicinal Chemistry, vol.48, issue.19, pp.5892-5899, 2005.
DOI : 10.1021/jm0401863

I. J. Majoros, A. Myc, T. Thomas, C. B. Mehta, and J. R. Baker, PAMAM Dendrimer-Based Multifunctional Conjugate for Cancer Therapy:?? Synthesis, Characterization, and Functionality, Biomacromolecules, vol.7, issue.2, pp.572-579, 2006.
DOI : 10.1021/bm0506142

A. Mecke, I. J. Majoros, A. K. Patri, J. R. Baker, M. M. Holl et al., Lipid Bilayer Disruption by Polycationic Polymers:?? The Roles of Size and Chemical Functional Group, Langmuir, vol.21, issue.23, pp.10348-10354, 2005.
DOI : 10.1021/la050629l

L. M. Kaminskas, B. J. Boyd, and C. J. Porter, Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties, Nanomedicine, vol.6, issue.6, pp.1063-1084, 2011.
DOI : 10.2217/nnm.11.67

S. S. Nigavekar, L. Y. Sung, M. Llanes, A. El-jawahri, T. S. Lawrence et al., H Dendrimer Nanoparticle Organ/Tumor Distribution, Pharmaceutical Research, vol.21, issue.3, pp.476-483, 2004.
DOI : 10.1023/B:PHAM.0000019302.26097.cc

URL : https://deepblue.lib.umich.edu/bitstream/2027.42/41504/1/11095_2004_Article_482250.pdf

N. Malik, R. Wiwattanapatapee, R. Klopsch, K. Lorenz, H. Frey et al., Dendrimers:, Journal of Controlled Release, vol.65, issue.1-2, pp.133-148, 2000.
DOI : 10.1016/S0168-3659(99)00246-1

L. M. Kaminskas, B. J. Boyd, P. Karellas, S. A. Henderson, M. P. Giannis et al., -lysine Dendrimers with Anionic Arylsulfonate or Succinate Groups on Intravenous Pharmacokinetics and Disposition, Molecular Pharmaceutics, vol.4, issue.6, pp.949-961, 2007.
DOI : 10.1021/mp070047s

H. Kobayashi, S. Kawamoto, T. Saga, N. Sato, A. Hiraga et al., Micro-MR angiography of normal and intratumoral vessels in mice using dedicated intravascular MR contrast agents with high generation of polyamidoamine dendrimer core: Reference to pharmacokinetic properties of dendrimer-based MR contrast agents, Journal of Magnetic Resonance Imaging, vol.46, issue.6
DOI : 10.1016/S0168-3659(99)00248-5

H. Kobayashi, S. Kawamoto, T. Saga, N. Sato, A. Hiraga et al., Positive effects of polyethylene glycol conjugation to generation, p.4

J. Lim, Y. Guo, C. L. Rostollan, J. Stanfield, J. Hsieh et al., The Role of the Size and Number of Polyethylene Glycol Chains in the Biodistribution and Tumor Localization of Triazine Dendrimers, Molecular Pharmaceutics, vol.5, issue.4, pp.540-547, 2008.
DOI : 10.1021/mp8000292

J. M. Harris and R. B. Chess, Effect of pegylation on pharmaceuticals, Nature Reviews Drug Discovery, vol.18, issue.3, pp.214-221, 2003.
DOI : 10.1023/A:1010910523202

K. Greish, G. Thiagarajan, H. Herd, R. Price, H. Bauer et al., Size and surface charge significantly influence the toxicity of silica and dendritic nanoparticles, Nanotoxicology, vol.21, issue.7, pp.713-723, 2012.
DOI : 10.1016/j.blre.2006.11.001

J. C. Roberts, M. K. Bhalgat, and R. T. Zera, Preliminary biological evaluation of polyamidoamine (PAMAM) StarburstTM dendrimers, Journal of Biomedical Materials Research, vol.30, issue.1, pp.53-65, 1996.
DOI : 10.1002/(SICI)1097-4636(199601)30:1<53::AID-JBM8>3.0.CO;2-Q

H. Chen, M. F. Neerman, A. R. Parrish, and E. E. Simanek, Cytotoxicity, Hemolysis, and Acute in Vivo Toxicity of Dendrimers Based on Melamine, Candidate Vehicles for Drug Delivery, Journal of the American Chemical Society, vol.126, issue.32
DOI : 10.1021/ja048548j

M. F. Neerman, W. Zhang, A. R. Parrish, and E. E. Simanek, In vitro and in vivo evaluation of a melamine dendrimer as a vehicle for drug delivery, International Journal of Pharmaceutics, vol.281, issue.1-2, pp.129-132, 2004.
DOI : 10.1016/j.ijpharm.2004.04.023

A. S. Chauhan, N. K. Jain, and P. V. Diwan, Pre-clinical and behavioural toxicity profile of PAMAM dendrimers in mice, Proc. R. Soc. -Math, pp.1535-1550, 2010.
DOI : 10.1016/S1359-6446(01)01757-3

J. B. Pryor, B. J. Harper, and S. L. Harper, Comparative toxicological assessment of PAMAM and thiophosphoryl dendrimers using embryonic zebrafish, Int. J. Nanomedicine, vol.9, pp.1947-1956, 2014.

T. C. King-heiden, E. Dengler, W. J. Kao, W. Heideman, and R. E. Peterson, Developmental toxicity of low generation PAMAM dendrimers in zebrafish, Toxicology and Applied Pharmacology, vol.225, issue.1, pp.70-79, 2007.
DOI : 10.1016/j.taap.2007.07.009

K. Winnicka, M. Wroblewska, K. Sosnowska, H. Car, and I. Kasacka, Evaluation of cationic polyamidoamine dendrimers&rsquo; dermal toxicity in the rat skin model, Drug Des. Devel. Ther, p.1367, 2015.

P. Rajananthanan, G. S. Attard, N. A. Sheikh, and W. J. Morrow, Evaluation of novel aggregate structures as adjuvants: composition, toxicity studies and humoral responses, Vaccine, vol.17, issue.7-8, pp.715-730, 1999.
DOI : 10.1016/S0264-410X(98)00256-4

H. B. Agashe, T. Dutta, M. Garg, and N. K. Jain, Investigations on the toxicological profile of functionalized fifth-generation poly(propylene imine) dendrimer, Journal of Pharmacy and Pharmacology, vol.19, issue.7, pp.1491-1498, 2006.
DOI : 10.1023/A:1016458104359

N. Malik, E. G. Evagorou, and R. Duncan, Dendrimer-platinate, Anti-Cancer Drugs, vol.10, issue.8, pp.767-776, 1999.
DOI : 10.1097/00001813-199909000-00010

M. T. Morgan, M. A. Carnahan, C. E. Immoos, A. A. Ribeiro, S. Finkelstein et al., Dendritic Molecular Capsules for Hydrophobic Compounds, Journal of the American Chemical Society, vol.125, issue.50, pp.15485-15489, 2003.
DOI : 10.1021/ja0347383

M. T. Morgan, Y. Nakanishi, D. J. Kroll, A. P. Griset, M. A. Carnahan et al., Dendrimer-Encapsulated Camptothecins: Increased Solubility, Cellular Uptake, and Cellular Retention Affords Enhanced Anticancer Activity In vitro, Cancer Research, vol.66, issue.24, pp.11913-11921, 2006.
DOI : 10.1158/0008-5472.CAN-06-2066

L. Wang, X. Xu, Y. Zhang, Y. Zhang, Y. Zhu et al., Encapsulation of curcumin within poly(amidoamine) dendrimers for delivery to cancer cells, Journal of Materials Science: Materials in Medicine, vol.74, issue.4
DOI : 10.1016/0092-8674(93)90508-N

L. M. Kaminskas, V. M. Mcleod, C. J. Porter, and B. J. Boyd, Association of Chemotherapeutic Drugs with Dendrimer Nanocarriers: An Assessment of the Merits of Covalent Conjugation Compared to Noncovalent Encapsulation, Molecular Pharmaceutics, vol.9, issue.3, pp.355-373, 2012.
DOI : 10.1021/mp2005966

R. S. Dhanikula and P. Hildgen, Influence of molecular architecture of polyether-co-polyester dendrimers on the encapsulation and release of methotrexate, Biomaterials, vol.28, issue.20, pp.3140-3152, 2007.
DOI : 10.1016/j.biomaterials.2007.03.012

L. M. Kaminskas, B. D. Kelly, V. M. Mcleod, G. Sberna, D. J. Owen et al., Characterisation and tumour targeting of PEGylated polylysine dendrimers bearing doxorubicin via a pH labile linker, Journal of Controlled Release, vol.152, issue.2, pp.241-248, 2011.
DOI : 10.1016/j.jconrel.2011.02.005

S. Khatri, N. G. Das, and S. K. Das, Effect of methotrexate conjugated PAMAM dendrimers on the viability of MES-SA uterine cancer cells, J. Pharm. Bioallied Sci, vol.6, issue.4, pp.297-302, 2014.

E. Abbasi, S. F. Aval, A. Akbarzadeh, M. Milani, H. T. Nasrabadi et al., Dendrimers: synthesis, applications, and properties, Nanoscale Research Letters, vol.9, issue.1
DOI : 10.1186/1556-276X-9-247

URL : http://doi.org/10.1186/1556-276x-9-247

P. Ma, X. Zhang, L. Ni, J. Li, F. Zhang et al., Targeted delivery of polyamidoamine-paclitaxel conjugate functionalized with anti-human epidermal growth factor receptor 2 trastuzumab, International Journal of Nanomedicine, p.2173, 2015.
DOI : 10.2147/IJN.S77152

B. Klajnert, M. Rozanek, and M. Bryszewska, Dendrimers in Photodynamic Therapy, Current Medicinal Chemistry, vol.19, issue.29, pp.4903-4912, 2012.
DOI : 10.2174/0929867311209024903

C. Kojima, Y. Toi, A. Harada, and K. Kono, Preparation of Poly(ethylene glycol)-Attached Dendrimers Encapsulating Photosensitizers for Application to Photodynamic Therapy, Bioconjugate Chemistry, vol.18, issue.3
DOI : 10.1021/bc060244u

O. Taratula, C. Schumann, M. A. Naleway, A. J. Pang, K. J. Chon et al., A Multifunctional Theranostic Platform Based on Phthalocyanine-Loaded Dendrimer for Image-Guided Drug Delivery and Photodynamic Therapy, Molecular Pharmaceutics, vol.10, issue.10, p.130910152412007, 2013.
DOI : 10.1021/mp400397t

O. Taratula, O. Taratula, M. Patel, C. Schumann, M. Naleway et al., Phthalocyanine-loaded graphene nanoplatform for&nbsp;imaging-guided combinatorial phototherapy, Int. J. Nanomedicine, p.2347, 2015.
DOI : 10.2147/ijn.s81097

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4378304

O. Taratula, C. Schumann, T. Duong, K. L. Taylor, and O. Taratula, Dendrimer-encapsulated naphthalocyanine as a single agent-based theranostic nanoplatform for near-infrared fluorescence imaging and combinatorial anticancer phototherapy, Nanoscale, vol.105, issue.9, pp.3888-3902, 2015.
DOI : 10.1073/pnas.0801349105

N. Nishiyama, H. R. Stapert, G. Zhang, D. Takasu, D. Jiang et al., Light-Harvesting Ionic Dendrimer Porphyrins as New Photosensitizers for Photodynamic Therapy, Bioconjugate Chemistry, vol.14, issue.1, pp.58-66, 2003.
DOI : 10.1021/bc025597h

G. Zhang, Polyion complex micelles entrapping cationic dendrimer porphyrin: effective photosensitizer for photodynamic therapy of cancer, Journal of Controlled Release, vol.93, issue.2, pp.141-150, 2003.
DOI : 10.1016/j.jconrel.2003.05.002

H. R. Stapert, N. Nishiyama, D. L. Jiang, T. Aida, and K. Kataoka, Polyion Complex Micelles Encapsulating Light-Harvesting Ionic Dendrimer Zinc Porphyrins, Langmuir, vol.16, issue.21, pp.8182-8188, 2000.
DOI : 10.1021/la000423e

W. D. Jang, N. Nishiyama, G. D. Zhang, A. Harada, D. L. Jiang et al., Supramolecular Nanocarrier of Anionic Dendrimer Porphyrins with Cationic Block Copolymers Modified with Polyethylene Glycol to Enhance Intracellular Photodynamic Efficacy, Angewandte Chemie International Edition, vol.28, issue.3, pp.419-423, 2005.
DOI : 10.1038/bjc.1958.52

W. Jang, N. Nishiyama, and K. Kataoka, Supramolecular Assembly of Photofunctional Dendrimers for Biomedical Nano-Devices, Supramolecular Chemistry, vol.102, issue.4-5, pp.4-5, 2007.
DOI : 10.1016/j.jconrel.2006.03.009

Y. Li, W. Jang, N. Nishiyama, A. Kishimura, S. Kawauchi et al., Dendrimer Generation Effects on Photodynamic Efficacy of Dendrimer Porphyrins and Dendrimer-Loaded Supramolecular Nanocarriers, Chemistry of Materials, vol.19, issue.23
DOI : 10.1021/cm071451m

N. Nishiyama, Y. Nakagishi, Y. Morimoto, P. Lai, K. Miyazaki et al., Enhanced photodynamic cancer treatment by supramolecular nanocarriers charged with dendrimer phthalocyanine, Journal of Controlled Release, vol.133, issue.3, pp.245-251, 2009.
DOI : 10.1016/j.jconrel.2008.10.010

H. Lu, W. Syu, N. Nishiyama, K. Kataoka, and P. Lai, Dendrimer phthalocyanine-encapsulated polymeric micelle-mediated photochemical internalization extends the efficacy of photodynamic therapy and overcomes drug-resistance in vivo, Journal of Controlled Release, vol.155, issue.3, pp.458-464, 2011.
DOI : 10.1016/j.jconrel.2011.06.005

P. M. Pereira, S. Silva, J. A. Cavaleiro, C. A. Ribeiro, J. P. Tomé et al., Galactodendritic Phthalocyanine Targets Carbohydrate-Binding Proteins Enhancing Photodynamic Therapy, PLoS ONE, vol.154, issue.4, p.95529, 2014.
DOI : 10.1371/journal.pone.0095529.s002

URL : http://doi.org/10.1371/journal.pone.0095529

S. H. Battah, C. E. Chee, H. Nakanishi, S. Gerscher, A. J. Macrobert et al., Synthesis and Biological Studies of 5-Aminolevulinic Acid-Containing Dendrimers for Photodynamic Therapy, Bioconjugate Chemistry, vol.12, issue.6, pp.980-988, 2001.
DOI : 10.1021/bc010027n

S. Battah, S. Balaratnam, A. Casas, S. O. Neill, C. Edwards et al., Macromolecular delivery of 5-aminolaevulinic acid for photodynamic therapy using dendrimer conjugates, Molecular Cancer Therapeutics, vol.6, issue.3, pp.876-885, 2007.
DOI : 10.1158/1535-7163.MCT-06-0359

A. Casas, S. Battah, G. D. Venosa, P. Dobbin, L. Rodriguez et al., Sustained and efficient porphyrin generation in vivo using dendrimer conjugates of 5-ALA for photodynamic therapy, Journal of Controlled Release, vol.135, issue.2, pp.136-143, 2009.
DOI : 10.1016/j.jconrel.2009.01.002

A. François, S. Battah, A. J. Macrobert, L. Bezdetnaya, F. Guillemin et al., approach using 5-aminolevulinic acid (ALA) dendrimers, BJU International, vol.154, issue.11c, pp.1155-1162, 2012.
DOI : 10.1016/S0002-9440(10)65318-7

S. Hackbarth, V. Horneffer, A. Wiehe, F. Hillenkamp, and B. Röder, Photophysical properties of pheophorbide-a-substituted diaminobutane poly-propylene-imine dendrimer, Chemical Physics, vol.269, issue.1-3, pp.339-346, 2001.
DOI : 10.1016/S0301-0104(01)00337-8

S. Hackbarth, E. A. Ermilov, and B. Röder, Interaction of Pheophorbide a molecules covalently linked to DAB dendrimers, Optics Communications, vol.248, issue.1-3, pp.1-3, 2005.
DOI : 10.1016/j.optcom.2004.11.088

A. Kay, R. Humphry-baker, and M. Graetzel, Artificial Photosynthesis. 2. Investigations on the Mechanism of Photosensitization of Nanocrystalline TiO2 Solar Cells by Chlorophyll Derivatives, The Journal of Physical Chemistry, vol.98, issue.3
DOI : 10.1021/j100054a035

K. I. Salokhiddinov, I. M. Byteva, and G. P. Gurinovich, Lifetime of singlet oxygen in various solvents, Journal of Applied Spectroscopy, vol.8, issue.5, pp.561-564, 1981.
DOI : 10.1007/BF00613067

H. A. Isakau, M. V. Parkhats, V. N. Knyukshto, B. M. Dzhagarov, E. P. Petrov et al., Toward understanding the high PDT efficacy of chlorin e6???polyvinylpyrrolidone formulations: Photophysical and molecular aspects of photosensitizer???polymer interaction in vitro, Journal of Photochemistry and Photobiology B: Biology, vol.92, issue.3
DOI : 10.1016/j.jphotobiol.2008.06.004

I. B. Matheson, R. D. Etheridge, N. R. Kratowich, and J. Lee, THE QUENCHING OF SINGLET OXYGEN BY AMINO ACIDS AND PROTEINS, Photochemistry and Photobiology, vol.19, issue.3, pp.165-171, 1975.
DOI : 10.1016/0014-5793(72)80632-X

J. Moan, E. O. Pettersen, and T. Christensen, The mechanism of photodynamic inactivation of human cells in vitro in the presence of haematoporphyrin, British Journal of Cancer, vol.39, issue.4, pp.398-407, 1979.
DOI : 10.1038/bjc.1979.72

J. Terao, Y. Minami, and N. Bando, Singlet molecular oxygen-quenching activity of carotenoids: relevance to protection of the skin from photoaging, Journal of Clinical Biochemistry and Nutrition, vol.48, issue.1, pp.57-62, 2010.
DOI : 10.3164/jcbn.11-008FR

P. Rees, Uptake and Toxicology of Nanoparticles, Frontiers of Nanoscience, pp.123-138, 2013.
DOI : 10.1016/B978-0-08-098338-7.00005-4

K. C. Das and H. P. Misra, Hydroxyl radical scavenging and singlet oxygen quenching properties of polyamines, Molecular and Cellular Biochemistry, vol.262, issue.1/2, pp.127-133, 2004.
DOI : 10.1023/B:MCBI.0000038227.91813.79

H. Yang and S. T. Lopina, Penicillin V-conjugated PEG-PAMAM star polymers, Journal of Biomaterials Science, Polymer Edition, vol.4, issue.10
DOI : 10.1021/bc00019a008

L. M. Kaminskas, B. D. Kelly, V. M. Mcleod, G. Sberna, B. J. Boyd et al., Capping Methotrexate ??-Carboxyl Groups Enhances Systemic Exposure and Retains the Cytotoxicity of Drug Conjugated PEGylated Polylysine Dendrimers, Molecular Pharmaceutics, vol.8, issue.2, pp.338-349, 2011.
DOI : 10.1021/mp1001872

A. Satsangi, S. S. Roy, R. K. Satsangi, R. K. Vadlamudi, and J. L. Ong, Design of a Paclitaxel Prodrug Conjugate for Active Targeting of an Enzyme Upregulated in Breast Cancer Cells, Molecular Pharmaceutics, vol.11, issue.6
DOI : 10.1021/mp500128k

G. M. Di-venosa, A. G. Casas, S. Battah, P. Dobbin, H. Fukuda et al., Investigation of a novel dendritic derivative of 5-aminolaevulinic acid for photodynamic therapy, The International Journal of Biochemistry & Cell Biology, vol.38, issue.1, pp.82-91, 2006.
DOI : 10.1016/j.biocel.2005.08.001

K. Lang, J. Mosinger, and D. M. Wagnerová, Photophysical properties of porphyrinoid sensitizers non-covalently bound to host molecules; models for photodynamic therapy, Coordination Chemistry Reviews, vol.248, issue.3-4, pp.3-4, 2004.
DOI : 10.1016/j.ccr.2004.02.004

A. Parat, C. Bordeianu, H. Dib, A. Garofalo, A. Walter et al., Dendrimer???nanoparticle conjugates in nanomedicine, Nanomedicine, vol.10, issue.6, pp.977-992, 2015.
DOI : 10.2217/nnm.14.196

V. Reshetov, H. Lassalle, A. François, D. Dumas, S. Hupont et al., Photodynamic therapy with conventional and PEGylated liposomal formulations of mTHPC (temoporfin): comparison of treatment efficacy and distribution characteristics in vivo, International Journal of Nanomedicine, p.3817, 2013.
DOI : 10.2147/IJN.S51002

URL : https://hal.archives-ouvertes.fr/hal-00875936

S. Svenson and D. Tomalia, Dendrimers in biomedical applications???reflections on the field, Advanced Drug Delivery Reviews, vol.57, issue.15, pp.2106-2129, 2005.
DOI : 10.1016/j.addr.2005.09.018

E. C. Cho, Q. Zhang, and Y. Xia, The effect of sedimentation and diffusion on cellular uptake of gold nanoparticles, Nature Nanotechnology, vol.97, issue.6, pp.385-391, 2011.
DOI : 10.1016/j.colsurfb.2008.02.013

H. Hatakeyama, H. Akita, and H. Harashima, A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: A strategy for overcoming the PEG dilemma, Advanced Drug Delivery Reviews, vol.63, issue.3, pp.152-160, 2011.
DOI : 10.1016/j.addr.2010.09.001

U. Schmidt-erfurth, H. Diddens, R. Birngruber, and T. Hasan, Photodynamic targeting of human retinoblastoma cells using covalent low-density lipoprotein conjugates, British Journal of Cancer, vol.75, issue.1, pp.54-61, 1997.
DOI : 10.1038/bjc.1997.9

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2222700/pdf

N. S. Soukos, M. R. Hamblin, and T. Hasan, Conjugates, Photochemistry and Photobiology, vol.48, issue.4, pp.723-729, 1997.
DOI : 10.1080/09553009014551501

J. D. Spikes, New trends in photobiology, Journal of Photochemistry and Photobiology B: Biology, vol.6, issue.3, pp.259-274, 1990.
DOI : 10.1016/1011-1344(90)85096-F

D. Shcharbin, J. Mazur, M. Szwedzka, M. Wasiak, B. Palecz et al., Interaction between PAMAM 4.5 dendrimer, cadmium and bovine serum albumin: A study using equilibrium dialysis, isothermal titration calorimetry, zeta-potential and fluorescence, Colloids and Surfaces B: Biointerfaces, vol.58, issue.2, pp.286-289, 2007.
DOI : 10.1016/j.colsurfb.2007.04.003

J. L. Perry, K. G. Reuter, M. P. Kai, K. P. Herlihy, S. W. 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. 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, Journal of the American Chemical Society, vol.134, issue.4, pp.2139-2147, 2012.
DOI : 10.1021/ja2084338

O. P. Perumal, R. Inapagolla, S. Kannan, and R. M. Kannan, The effect of surface functionality on cellular trafficking of dendrimers, Biomaterials, vol.29, issue.24-25, pp.24-25, 2008.
DOI : 10.1016/j.biomaterials.2008.04.038

G. Sahay, J. O. Kim, A. V. Kabanov, and T. K. Bronich, The exploitation of differential endocytic pathways in normal and tumor cells in the selective targeting of nanoparticulate chemotherapeutic agents, Biomaterials, vol.31, issue.5, pp.923-933, 2010.
DOI : 10.1016/j.biomaterials.2009.09.101

M. Teiten, L. Bezdetnaya, P. Morlière, R. Santus, and F. Guillemin, Endoplasmic reticulum and Golgi apparatus are the preferential sites of Foscan?? localisation in cultured tumour cells, British Journal of Cancer, vol.88, issue.1
DOI : 10.1038/sj.bjc.6600664

J. Moan, K. Berg, E. Kvam, A. Western, Z. Malik et al., Intracellular Localization of Photosensitizers, Ciba Found. Symp, vol.847, pp.95-107, 1989.
DOI : 10.1042/bj1980067

S. J. Holt, Some Observations on the Occurrence and Nature of Esterases in Lysosomes in Ciba Foundation Symposium -Anterior Pituitary Secretion (Book I of Colloquia on Endocrinology, pp.114-125, 1963.

W. Luo, R. Liu, J. Zhu, Y. Li, and H. Liu, Subcellular location and photodynamic therapeutic effect of chlorin e6 in the human tongue squamous cell cancer Tca8113 cell line, Oncology Letters, 2014.
DOI : 10.3892/ol.2014.2720

S. Zhu, M. Hong, L. Zhang, G. Tang, Y. Jiang et al., PEGylated PAMAM Dendrimer-Doxorubicin Conjugates: In Vitro Evaluation and In Vivo Tumor Accumulation, Pharmaceutical Research, vol.76, issue.1, pp.161-174, 2010.
DOI : 10.7326/0003-4819-80-2-249

H. Xu, Y. Deng, D. Chen, W. Hong, Y. Lu et al., Esterase-catalyzed dePEGylation of pH-sensitive vesicles modified with cleavable PEG-lipid derivatives, Journal of Controlled Release, vol.130, issue.3, pp.238-245, 2008.
DOI : 10.1016/j.jconrel.2008.05.009

M. Najlah, S. Freeman, D. Attwood, and A. D. Emanuele, In vitro evaluation of dendrimer prodrugs for oral drug delivery, International Journal of Pharmaceutics, vol.336, issue.1, pp.183-190, 2007.
DOI : 10.1016/j.ijpharm.2006.11.047