A. Samad, Y. Sultana, and M. Aqil, Liposomal Drug Delivery Systems: An Update Review, Current Drug Delivery, vol.4, issue.4, pp.297-305, 2007.
DOI : 10.2174/156720107782151269

M. C. Woodle, J. W. Park, K. Hong, D. B. Kirpotin, G. Colbern et al., Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle, Nucleic Acids Res Clin Cancer Res, vol.32, issue.8, pp.149-198, 2002.

V. P. 50-torchilin, Targeted pharmaceutical nanocarriers for cancer therapy and imaging, The AAPS Journal, vol.9, issue.2, pp.128-147, 2007.
DOI : 10.1208/aapsj0902015

J. K. Vasir and V. Labhasetwar, Polymeric nanoparticles for gene delivery, Expert Opinion on Drug Delivery, vol.269, issue.3, pp.325-344, 2006.
DOI : 10.1016/j.jconrel.2004.10.024

J. Temsamani and P. Vidal, The use of cell-penetrating peptides for drug delivery, Drug Discovery Today, vol.9, issue.23, pp.1012-1019, 2004.
DOI : 10.1016/S1359-6446(04)03279-9

B. Gupta, T. S. Levchenko, and V. P. Torchilin, Intracellular delivery of large molecules and small particles by cell-penetrating proteins and peptides compound combinatorial libraries and chemical microarrays in signal transduction research, Nonpeptidic alpha(v)beta(3) Integrin Antagonist Libraries: On-Bead Screening and Mass Spectrometric Identification without Tagging, pp.637-651, 2001.

L. Ying and J. Gervay-hague, One-Bead-One-Inhibitor-One-Substrate Screening of Neuraminidase Activity, ChemBioChem, vol.11, issue.10, pp.1857-1865, 2005.
DOI : 10.1002/cbic.200500006

S. 233-dixon, K. T. Ziebart, Z. He, M. Jeddeloh, C. L. Yoo et al., Aminodeoxychorismate Synthase Inhibitors from One-Bead One-Compound Combinatorial Libraries:?? ???Staged??? Inhibitor Design, Journal of Medicinal Chemistry, vol.49, issue.25, pp.7413-7426, 2006.
DOI : 10.1021/jm0609869

F. 234-sebestyén, G. Dibó, A. Kovács, and A. Furkua, Chemical synthesis of peptide libraries, Bioorganic & Medicinal Chemistry Letters, vol.3, issue.3, pp.413-418, 1993.
DOI : 10.1016/S0960-894X(01)80223-7

J. A. 235-kritzer, N. W. Luedtke, E. A. Harker, and A. Schepartz, A Rapid Library Screen for Tailoring ??-Peptide Structure and Function, Journal of the American Chemical Society, vol.127, issue.42, pp.14584-14585, 2005.
DOI : 10.1021/ja055050o

S. Aggarwal, J. L. Harden, and S. R. Denmeade, Synthesis and Screening of a Random Dimeric Peptide Library Using the One-Bead???One-Dimer Combinatorial Approach, Bioconjugate Chemistry, vol.17, issue.2, pp.335-340, 2006.
DOI : 10.1021/bc0502659

O. H. 237-aina, J. Marik, R. Liu, D. H. Lau, and K. S. Lam, Identification of novel targeting peptides for human ovarian cancer cells using "one-bead one-compound" combinatorial libraries, Molecular Cancer Therapeutics, vol.4, issue.5, pp.806-813, 2005.
DOI : 10.1158/1535-7163.MCT-05-0029

K. P. Vollhardt and N. Schore, Acides aminés, peptides, protéines et acides nucléiques, pp.1158-1159, 1999.

D. Hanahan and R. A. Weinberg, The Hallmarks of Cancer, Cell, vol.100, issue.1, pp.57-70, 2000.
DOI : 10.1016/S0092-8674(00)81683-9

J. Ferlay, F. Bray, P. Pisani, D. M. Parkin, and . Globocan, Cancer Incidence, Mortality and Prevalence Worldwide, IARC CancerBase No 5, version 2.0, 2002.

J. Ferlay, P. Autier, M. Boniol, M. Heanue, M. Colombet et al., Estimates of the cancer incidence and mortality in Europe in 2006, Europe, pp.581-592, 2006.
DOI : 10.1093/annonc/mdl498

B. W. Stewart and P. Kleihues, Le cancer, un fardeau mondial. Le cancer dans le monde ed, pp.13-19, 2005.

D. C. Sullivan and G. Kelloff, Seeing into cells, EMBO reports, vol.13, issue.4, pp.292-296, 2005.
DOI : 10.1162/1535350042973526

J. Robert, D. Schrama, R. A. Reisfeld, and J. C. Becker, Pharmacologie des médicaments anticancéreux Antibody targeted drugs as cancer therapeutics, Nat Rev Drug Discov, vol.10, issue.5, pp.403-413, 2005.

. Clinicaltrials, Gov Ranibizumab Injections to Treat Retinal Tumors in Patients With Von Hippel-Lindau Syndrome, J.; Cunningham, D. Panitumumab. Drugs Today, vol.42, pp.711-719, 2006.

G. P. Adams and L. M. Weiner, Monoclonal antibody therapy of cancer, Lippert, J. W. Vascular disrupting agents, pp.1147-1157, 2005.
DOI : 10.1016/j.ccr.2004.12.003

P. Hinnen and F. A. Eskens, Vascular disrupting agents in clinical development, British Journal of Cancer, vol.115, issue.8, pp.1159-1165, 2007.
DOI : 10.1056/NEJMoa021491

M. L. Edelstein, M. R. Abedi, J. Wixon, and R. M. Edelstein, Gene therapy clinical trials worldwide 1989???2004???an overview, The Journal of Gene Medicine, vol.6, issue.6, pp.597-602, 2004.
DOI : 10.1002/jgm.619

URL : http://dx.doi.org/10.1016/j.ymthe.2004.06.954

S. Eckhardt, Molecular targeted therapy: A strategy of disillusions or optimism?, Journal of Laboratory and Clinical Medicine, vol.147, issue.3, pp.108-113, 2006.
DOI : 10.1016/j.lab.2005.11.005

R. Chari, Targeted Cancer Therapy: Conferring Specificity to Cytotoxic Drugs, Accounts of Chemical Research, vol.41, issue.1, pp.98-107, 2008.
DOI : 10.1021/ar700108g

G. Payne, Progress in immunoconjugate cancer therapeutics, Cancer Cell, vol.3, issue.3, pp.207-212, 2003.
DOI : 10.1016/S1535-6108(03)00057-6

P. Carter, Improving the efficacy of antibody-based cancer therapies, Nature Reviews Cancer, vol.1, issue.2, pp.118-129, 2001.
DOI : 10.1038/35101072

A. M. Wu and P. Senter, Arming antibodies: prospects and challenges for immunoconjugates, Nature Biotechnology, vol.90, issue.9, pp.1137-1146, 2005.
DOI : 10.1021/bc050039z

D. Azria, J. Spano, . J. Thérapies-ciblées-en-cancérologie.-ed, and . Libbey-eurotext, VI- 127 p. 23 Perspectives on cancer therapy with radiolabeled monoclonal antibodies, J Nucl Med, vol.46, pp.115-127, 2005.

H. Xu, K. Baidoo, A. J. Gunn, C. A. Boswell, D. E. Milenic et al., Design, Synthesis, and Characterization of a Dual Modality Positron Emission Tomography and Fluorescence Imaging Agent for Monoclonal Antibody Tumor-Targeted Imaging, Journal of Medicinal Chemistry, vol.50, issue.19, pp.4759-4765, 2007.
DOI : 10.1021/jm070657w

M. Gutowski, M. Carcenac, D. Pourquier, C. Larroque, B. Saint-aubert et al., Intraoperative immunophotodetection for radical resection of cancers: evaluation in an experimental model, Clin Cancer Res, vol.7, pp.1142-1148, 2001.

P. Holliger and P. J. Hudson, Engineered antibody fragments and the rise of single domains, Nature Biotechnology, vol.10, issue.9, pp.1126-1136, 2005.
DOI : 10.1182/blood-2005-03-1153

P. J. Carter, Potent antibody therapeutics by design, Nature Reviews Immunology, vol.421, issue.5, pp.343-357, 2006.
DOI : 10.1038/nri1837

S. S. Sidhu, F. Fellouse, S. P. Vyas, and V. Sihorkar, Synthetic therapeutic antibodies Endogenous carriers and ligands in non-immunogenic site-specific drug delivery, Nat Chem Biol Adv Drug Deliv Rev, vol.2, issue.43, pp.682-688, 2000.

T. R. Daniels, T. Delgado, G. Helguera, and M. L. Penichet, The transferrin receptor part II: Targeted delivery of therapeutic agents into cancer cells, Clinical Immunology, vol.121, issue.2, pp.159-176, 2006.
DOI : 10.1016/j.clim.2006.06.006

S. Jaracz, J. Chen, L. V. Kuznetsova, and I. Ojima, Recent advances in tumor-targeting anticancer drug conjugates, Bioorganic & Medicinal Chemistry, vol.13, issue.17, pp.5043-5054, 2005.
DOI : 10.1016/j.bmc.2005.04.084

E. Ruoslahti, Targeting tumor vasculature with homing peptides from phage display, Seminars in Cancer Biology, vol.10, issue.6, pp.435-442, 2000.
DOI : 10.1006/scbi.2000.0334

O. H. Aina, T. C. Sroka, M. L. Chen, and K. Lam, Therapeutic cancer targeting peptides, Therapeutic cancer targeting peptides, pp.184-199, 2002.
DOI : 10.1002/bip.10257

M. Mammen, S. Chio, and G. M. Whitesides, Polyvalent interactions in biological systems: implications for design and use of multivalent ligands and inhibitors, Angew Chem Int Ed Engl, vol.37, pp.2755-2794, 1998.

J. E. Gestwicki, C. W. Cairo, L. E. Strong, K. A. Oetjen, and L. L. Kiessling, Influencing Receptor???Ligand Binding Mechanisms with Multivalent Ligand Architecture, Journal of the American Chemical Society, vol.124, issue.50, pp.14922-14933, 2002.
DOI : 10.1021/ja027184x

A. J. Schraa, R. J. Kok, A. D. Berendsen, H. E. Moorlag, E. J. Bos et al., Endothelial cells internalize and degrade RGD-modified proteins developed for tumor vasculature targeting, Journal of Controlled Release, vol.83, issue.2, pp.241-251, 2002.
DOI : 10.1016/S0168-3659(02)00206-7

L. L. Kiessling, J. E. Gestwicki, and L. Strong, Synthetic Multivalent Ligands as Probes of Signal Transduction, Angewandte Chemie International Edition, vol.76, issue.15, pp.2348-2368, 2006.
DOI : 10.1002/anie.200502794

C. B. Carlson, P. Mowery, R. M. Owen, E. C. Dykhuizen, and L. L. Kiessling, Selective Tumor Cell Targeting Using Low-Affinity, Multivalent Interactions, ACS Chemical Biology, vol.2, issue.2, pp.119-127, 2007.
DOI : 10.1021/cb6003788

A. Rawat, B. Vaidya, K. Khatri, A. K. Goyal, P. N. Gupta et al., Targeted intracellular delivery of therapeutics: an overview, Pharmazie, vol.62, pp.643-658, 2007.

T. C. Liu, E. Galanis, and D. Kirn, Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress, Nature Clinical Practice Oncology, vol.1, issue.2, pp.101-117, 2007.
DOI : 10.1038/ncponc0736

R. M. Owen, C. B. Carlson, J. Xu, P. Mowery, E. Fasella et al., Bifunctional Ligands that Target Cells Displaying the ??v??3 Integrin, Bifunctional ligands that target cells displaying the alpha v beta3 integrin, pp.68-82, 2007.
DOI : 10.1002/cbic.200600339

H. Wu, T. Seki, I. Dmitriev, T. Uil, E. Kashentseva et al., Double modification of adenovirus fiber with RGD and polylysine motifs improves coxsackievirus-adenovirus receptorindependent gene transfer efficiency, pp.1647-1653, 2002.

P. Couvreur and C. Vauthier, Nanotechnology: Intelligent Design to Treat Complex Disease, Pharmaceutical Research, vol.4, issue.421, pp.1417-1450, 2006.
DOI : 10.1007/s11095-006-0284-8

I. Cheong, X. Huang, K. Thornton, L. A. Diaz, . Jr et al., Targeting Cancer with Bugs and Liposomes: Ready, Aim, Fire, Cancer Research, vol.67, issue.20, pp.9605-9608, 2007.
DOI : 10.1158/0008-5472.CAN-07-1565

V. Russ and E. Wagner, Cell and Tissue Targeting of Nucleic Acids for Cancer Gene Therapy, Pharmaceutical Research, vol.17, issue.Suppl. 1, pp.1047-1057, 2007.
DOI : 10.1007/s11095-006-9233-9

A. Samad, Y. Sultana, and M. Aqil, Liposomal Drug Delivery Systems: An Update Review, Current Drug Delivery, vol.4, issue.4, pp.297-305, 2007.
DOI : 10.2174/156720107782151269

R. M. Schiffelers, A. Ansari, J. Xu, Q. Zhou, Q. Tang et al., Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle, Nucleic Acids Research, vol.32, issue.19, pp.149-1172, 2002.
DOI : 10.1093/nar/gnh140

V. P. Torchilin, Targeted pharmaceutical nanocarriers for cancer therapy and imaging 51. Clinicaltrials.Gov Safety Study of Infusion of SGT-53 to Treat Solid Tumors, NCT00470613. 52. Torchilin, V. P. Multifunctional nanocarriers, pp.128-147, 2006.

J. K. Vasir and V. Labhasetwar, Polymeric nanoparticles for gene delivery, Expert Opinion on Drug Delivery, vol.269, issue.3, pp.325-344, 2006.
DOI : 10.1016/j.jconrel.2004.10.024

J. Temsamani and P. Vidal, The use of cell-penetrating peptides for drug delivery, Drug Discovery Today, vol.9, issue.23, pp.1012-1019, 2004.
DOI : 10.1016/S1359-6446(04)03279-9

B. Gupta, T. S. Levchenko, and V. P. Torchilin, Intracellular delivery of large molecules and small particles by cell-penetrating proteins and peptides, Advanced Drug Delivery Reviews, vol.57, issue.4, pp.637-651, 2005.
DOI : 10.1016/j.addr.2004.10.007

L. N. Patel, J. L. Zaro, and W. Shen, Cell Penetrating Peptides: Intracellular Pathways and Pharmaceutical Perspectives, Pharmaceutical Research, vol.1712, issue.(Pt 8), pp.1977-1992, 2007.
DOI : 10.1007/s11095-007-9303-7

S. Deshayes, M. Morris, F. Heitz, and G. Divita, Delivery of proteins and nucleic acids using a non-covalent peptide-based strategy, Advanced Drug Delivery Reviews, vol.60, issue.4-5, pp.537-547, 2008.
DOI : 10.1016/j.addr.2007.09.005

URL : https://hal.archives-ouvertes.fr/hal-00350872

F. Maurel, F. Debart, F. Cavelier, A. R. Thierry, B. Lebleu et al., Toward high yield synthesis of peptide???oligonucleotide chimera through a disulfide bridge: A simplified method for oligonucleotide activation, Bioorganic & Medicinal Chemistry Letters, vol.15, issue.22, pp.5084-5087, 2005.
DOI : 10.1016/j.bmcl.2005.07.086

URL : https://hal.archives-ouvertes.fr/hal-00117471

A. Dirksen, S. Langereis, B. F. De-waal, M. H. Van-genderen, T. M. Hackeng et al., A supramolecular approach to multivalent target-specific MRI contrast agents for angiogenesis, Chemical Communications, vol.307, issue.22, pp.2811-2813, 2005.
DOI : 10.1039/b502347e

Y. Liu, A. C. Deisseroth, and P. Choong, Tumor vascular targeting therapy with viral vectors Targeting of small molecule anticancer drugs to the tumour and its vasculature using cationic liposomes: lessons from gene therapy, Cancer Cell Int, vol.107, issue.6, pp.3027-3033, 2006.

F. Sonvico, S. Mornet, S. Vasseur, C. Dubernet, D. Jaillard et al., Folate-Conjugated Iron Oxide Nanoparticles for Solid Tumor Targeting as Potential Specific Magnetic Hyperthermia Mediators: Synthesis, Physicochemical Characterization, and in Vitro Experiments, Bioconjugate Chemistry, vol.16, issue.5, pp.1181-1188, 2005.
DOI : 10.1021/bc050050z

URL : https://hal.archives-ouvertes.fr/hal-00102165

Y. Liu, H. Miyoshi, and M. Nakamura, Nanomedicine for drug delivery and imaging: A promising avenue for cancer therapy and diagnosis using targeted functional nanoparticles, International Journal of Cancer, vol.311, issue.12, pp.2527-2537, 2007.
DOI : 10.1002/ijc.22709

H. D. Maynard, S. Y. Okada, and R. H. Grubbs, Inhibition of Cell Adhesion to Fibronectin by Oligopeptide-Substituted Polynorbornenes, Journal of the American Chemical Society, vol.123, issue.7, pp.1275-1279, 2001.
DOI : 10.1021/ja003305m

Y. Cheng, Y. Gao, T. Rao, Y. Li, and T. Xu, Dendrimer-Based Prodrugs: Design, Synthesis, Screening and Biological Evaluation, Combinatorial Chemistry & High Throughput Screening, vol.10, issue.5, pp.336-349, 2007.
DOI : 10.2174/138620707781662808

T. Darbre and J. L. Reymond, Peptide Dendrimers as Artificial Enzymes, Receptors, and Drug-Delivery Agents, Accounts of Chemical Research, vol.39, issue.12, pp.925-934, 2006.
DOI : 10.1021/ar050203y

L. Baldini, A. Casnati, F. Sansone, and R. Ungaro, Calixarene-based multivalent ligands, Chem. Soc. Rev., vol.126, issue.2, pp.254-266, 2007.
DOI : 10.1016/j.ica.2006.07.023

K. J. Jensen and J. Brask, Carbohydrates in peptide and protein design, Biopolymers, vol.125, issue.6, pp.747-761, 2005.
DOI : 10.1002/bip.20300

R. Tong, J. Cheng, K. M. Fichter, L. Zhang, K. L. Kiick et al., Anticancer Polymeric Nanomedicines Polymer Reviews Peptide-Functionalized Poly(ethylene glycol) Star Polymers: DNA Delivery Vehicles with Multivalent Molecular Architecture, Bioconjug Chem, vol.47, issue.19, pp.345-381, 2007.

J. P. Tam, Synthetic peptide vaccine design: synthesis and properties of a high-density multiple antigenic peptide system., Proceedings of the National Academy of Sciences, vol.85, issue.15, pp.5409-5413, 1988.
DOI : 10.1073/pnas.85.15.5409

J. Ni, R. Powell, I. V. Baskakov, A. Devico, G. K. Lewis et al., Synthesis, conformation, and immunogenicity of monosaccharide-centered multivalent HIV-1 gp41 peptides containing the sequence of DP178, Bioorganic & Medicinal Chemistry, vol.12, issue.12, pp.3141-3148, 2004.
DOI : 10.1016/j.bmc.2004.04.008

H. Li and L. Wang, Cholic acid as template for multivalent peptide assembly, Organic & Biomolecular Chemistry, vol.1, issue.20, pp.3507-3513, 2003.
DOI : 10.1039/b307995c

C. Guarise, L. J. Prins, and P. Scrimin, Fully symmetrical functionalization of multivalent scaffold molecules on solid support Recent Carbohydrate-Based Chemoselective Ligation Applications, Tetrahedron Curr Org Synth, vol.62, issue.2, pp.11670-11674, 2005.

S. Kent, Total chemical synthesis of enzymes, Journal of Peptide Science, vol.38, issue.9, pp.574-593, 2003.
DOI : 10.1002/psc.475

S. Papot, I. Tranoy, F. Tillequin, J. C. Florent, and J. P. Gesson, Design of Selectively Activated Anticancer Prodrugs: Elimination and Cyclization Strategies, Current Medicinal Chemistry-Anti-Cancer Agents, vol.2, issue.2, pp.155-185, 2002.
DOI : 10.2174/1568011023354173

F. Kratz, I. A. Muller, C. Ryppa, and A. Warnecke, Prodrug Strategies in Anticancer Chemotherapy, ChemMedChem, vol.9, issue.1, pp.20-53, 2007.
DOI : 10.1002/cmdc.200700159

F. Kratz, K. Abu-ajaj, and A. Warnecke, Anticancer carrier-linked prodrugs in clinical trials, Expert Opinion on Investigational Drugs, vol.24, issue.7, pp.1037-1058, 2007.
DOI : 10.1080/07357900500359935

E. Sausville, P. Lorusso, M. Quinn, K. Forman, C. Leamon et al., A phase I study of EC145 administered weeks 1 and 3 of a 4-week cycle in patients with refractory solid tumors, ASCO Annual Meeting Proceedings 2007, 25, 2577. 83. Herbst, R. S. Role of novel targeted therapies in the clinic, pp.21-27, 2005.

P. Carmeliet, Angiogenesis in health and disease, Nature Medicine, vol.9, issue.6, pp.653-660, 2003.
DOI : 10.1038/nm0603-653

E. Goldman, The growth of malignant disease in man and the lower animals with special reference to the vascular system, Lancet, vol.2, pp.1236-1240, 1907.

G. H. Algire and H. W. Chalkley, Vascular reactions of normal and malignant tissues in vivo, J Natl Cancer Inst, vol.41, pp.111-124, 1945.

J. Folkman, Tumor angiogenesis: therapeutic implications Angiogenesis and tumor metastasis, N Engl J Med Annu Rev Med, vol.285, issue.49, pp.1182-1186, 1971.

P. Auguste, S. Lemiere, F. Larrieu-lahargue, and A. Bikfalvi, Molecular mechanisms of tumor vascularization, Critical Reviews in Oncology/Hematology, vol.54, issue.1, pp.53-61, 2003.
DOI : 10.1016/j.critrevonc.2004.11.006

D. Hanahan, J. Folkman, P. Carmeliet, and R. K. Jain, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis Angiogenesis in cancer and other diseases Angiogenesis as a therapeutic target Angiogenesis in life, disease and medicine, Cell Nature Nature Nature, vol.86, issue.438, pp.353-364, 1996.

A. Bikfalvi, Tumor angiogenesis, Bull Cancer, vol.90, pp.449-458, 2003.

P. Alessi, C. Ebbinghaus, and D. Neri, Molecular targeting of angiogenesis, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol.1654, issue.1, pp.39-49, 2004.
DOI : 10.1016/j.bbcan.2003.08.001

R. K. Jain, Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy, Science, vol.307, issue.5706, pp.58-62, 2005.
DOI : 10.1126/science.1104819

B. H. Luo and T. A. Springer, Integrin structures and conformational signaling, Current Opinion in Cell Biology, vol.18, issue.5, pp.579-586, 2006.
DOI : 10.1016/j.ceb.2006.08.005

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1618925

J. P. Xiong, T. Stehle, B. Diefenbach, R. Zhang, R. Dunker et al., Crystal Structure of the Extracellular Segment of Integrin alpha Vbeta 3, Science, vol.294, issue.5541, pp.339-345, 2001.
DOI : 10.1126/science.1064535

J. Takagi, B. M. Petre, T. Walz, and T. A. Springer, Global Conformational Rearrangements in Integrin Extracellular Domains in Outside-In and Inside-Out Signaling, Cell, vol.110, issue.5, pp.599-511, 2002.
DOI : 10.1016/S0092-8674(02)00935-2

M. Kim, C. V. Carman, and T. A. Springer, Bidirectional Transmembrane Signaling by Cytoplasmic Domain Separation in Integrins, Science, vol.301, issue.5640, pp.1720-1725, 2003.
DOI : 10.1126/science.1084174

B. H. Luo, C. V. Carman, and T. A. Springer, Structural Basis of Integrin Regulation and Signaling, Annual Review of Immunology, vol.25, issue.1, pp.619-647, 2007.
DOI : 10.1146/annurev.immunol.25.022106.141618

C. Buensuceso, M. De-virgilio, and S. J. Shattil, Detection of Integrin ??IIb??3Clustering in Living Cells, Journal of Biological Chemistry, vol.278, issue.17, pp.15217-15224, 2003.
DOI : 10.1074/jbc.M213234200

R. O. Hynes, L. E. Reynolds, L. Wyder, J. C. Lively, D. Taverna et al., A reevaluation of integrins as regulators of angiogenesis, Nature Medicine, vol.4, issue.9, pp.918-921, 2002.
DOI : 10.1038/nm0302-193

R. Max, R. R. Gerritsen, P. T. Nooijen, S. L. Goodman, A. Sutter et al., Immunohistochemical analysis of integrin ??v??3 expression on tumor-associated vessels of human carcinomas, International Journal of Cancer, vol.133, issue.3, pp.320-324, 1994.
DOI : 10.1002/(SICI)1097-0215(19970502)71:3<320::AID-IJC2>3.0.CO;2-#

E. H. Lim, N. Danthi, M. Bednarski, and K. C. Li, A review: Integrin ??v??3-targeted molecular imaging and therapy in angiogenesis, Nanomedicine: Nanotechnology, Biology and Medicine, vol.1, issue.2, pp.110-114, 2005.
DOI : 10.1016/j.nano.2005.03.008

J. A. Posey, M. B. Khazaeli, A. Delgrosso, M. N. Saleh, C. Y. Lin et al., ) Antibody in Patients with Metastatic Cancer, Cancer Biotherapy & Radiopharmaceuticals, vol.16, issue.2, pp.125-132, 2001.
DOI : 10.1089/108497801300189218

A. R. Hsu, A. Veeravagu, W. Cai, L. C. Hou, V. Tse et al., Integrin alphav beta3 Antagonists for Anti-Angiogenic Cancer Treatment. Recent Pat on Anti-Cancer Drug Discovery, pp.143-158, 2007.

P. Silva, P. Yalamanchili, S. Robinson, J. Lazewatsky, J. Barrett et al., Design, synthesis, and evaluation of radiolabeled integrin alphav beta3 receptor antagonists for tumor imaging and radiotherapy, Cancer Biother Radiopharm, vol.18, pp.627-641, 2003.

J. L. Sutcliffe-goulden, M. J. O-'doherty, P. K. Marsden, I. R. Hart, J. F. Marshall et al., Rapid solid phase synthesis and biodistribution of 18F-labelled linear peptides, European Journal of Nuclear Medicine and Molecular Imaging, vol.29, issue.6, pp.754-759, 2002.
DOI : 10.1007/s00259-001-0756-3

M. Gurrath, G. Muller, H. Kessler, M. Aumailley, and R. Timpl, Conformation/activity studies of rationally designed potent anti-adhesive RGD peptides, European Journal of Biochemistry, vol.31, issue.3, pp.911-921, 1992.
DOI : 10.1063/1.438208

M. Pfaff, K. Tangemann, B. Muller, M. Gurrath, G. Muller et al., Selective recognition of cyclic RGD peptides of NMR defined conformation by alphaIIb beta3, alphav beta3, and alpha5 beta1 integrins, J Biol Chem, vol.269, pp.20233-20238, 1994.

H. Kessler, Methylated cyclic RGD peptides as highly active and selective alphav beta3 integrin antagonists, J Med Chem, vol.42, pp.3033-3040, 1999.

S. L. Goodman, G. Holzemann, G. A. Sulyok, H. R. Kessler, M. D. Pierschbacher et al., Nanomolar small molecule inhibitors for alphav beta6, alphav beta5, and alphav beta3 integrins Incorporation of thioether building blocks into an alphav beta3-specific RGD peptide: synthesis and biological activity, J Med Chem Clinicaltrials.Gov Cilengitide Biopolymers, vol.45, issue.71, pp.1045-1051, 2002.

E. Koivunen, B. Wang, and E. Ruoslahti, Phage Libraries Displaying Cyclic Peptides with Different Ring Sizes: Ligand Specificities of the RGD-Directed Integrins, Bio/Technology, vol.268, issue.3, pp.265-270, 1995.
DOI : 10.1016/0076-6879(82)82103-4

P. Holig, M. Bach, T. Volkel, T. Nahde, S. Hoffmann et al., Novel RGD lipopeptides for the targeting of liposomes to integrin-expressing endothelial and melanoma cells, Protein Engineering Design and Selection, vol.17, issue.5, pp.433-441, 2004.
DOI : 10.1093/protein/gzh055

K. Temming, R. M. Schiffelers, G. Molema, and R. J. Kok, RGD-based strategies for selective delivery of therapeutics and imaging agents to the tumour vasculature, Drug Resistance Updates, vol.8, issue.6, pp.381-402, 2005.
DOI : 10.1016/j.drup.2005.10.002

J. P. Xiong, T. Stehle, R. Zhang, A. Joachimiak, M. Frech et al., Crystal Structure of the Extracellular Segment of Integrin alpha Vbeta 3 in Complex with an Arg-Gly-Asp Ligand, Science, vol.296, issue.5565, pp.151-155, 2002.
DOI : 10.1126/science.1069040

F. Mitjans, T. Meyer, C. Fittschen, S. Goodman, A. Jonczyk et al., In vivo therapy of malignant melanoma by means of antagonists of ??v integrins, International Journal of Cancer, vol.59, issue.5, pp.716-723, 2000.
DOI : 10.1002/1097-0215(20000901)87:5<716::AID-IJC14>3.0.CO;2-R

P. A. Burke, S. J. Denardo, L. A. Miers, K. R. Lamborn, S. Matzku et al., Cilengitide targeting of alphav beta3 integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts, Cancer Res, vol.62, pp.4263-4272, 2002.

R. Haubner, H. J. Wester, U. Reuning, R. Senekowitsch-schmidtke, B. Diefenbach et al., Radiolabeled alphav beta3 integrin antagonists: a new class of tracers for tumor targeting, J Nucl Med, vol.40, pp.1061-1071, 1999.

H. Kessler and M. Schwaiger, Glycosylated RGD-containing peptides: tracer for tumor targeting and angiogenesis imaging with improved biokinetics, J Nucl Med, vol.42, pp.326-336, 2001.

R. Haubner, H. J. Wester, W. A. Weber, C. Mang, S. I. Ziegler et al., Noninvasive imaging of alphav beta3 integrin expression using 18F-labeled RGD-containing glycopeptide and positron emission tomography, Cancer Res, vol.61, pp.1781-1785, 2001.

X. Chen, M. Tohme, R. Park, Y. Hou, J. R. Bading et al., Micro-PET Imaging of ??<SUB>v</SUB>??<SUB>3</SUB>-Integrin Expression with <SUP>18</SUP>F-Labeled Dimeric RGD Peptide, Molecular Imaging, vol.3, issue.2, pp.96-104, 2004.
DOI : 10.1162/1535350041464892

X. Chen, Y. Hou, M. Tohme, R. Park, V. Khankaldyyan et al., Pegylated Arg-Gly-Asp peptide: 64Cu labeling and PET imaging of brain tumor alphav beta3-integrin expression, J Nucl Med, vol.45, pp.1776-1783, 2004.

B. Jia, J. Shi, Z. Yang, B. Xu, Z. Liu et al., Expression, Bioconjugate Chemistry, vol.17, issue.4, pp.1069-1076, 2006.
DOI : 10.1021/bc060055b

URL : https://hal.archives-ouvertes.fr/hal-00123203

E. Garanger, D. Boturyn, and P. Dumy, Tumor Targeting with RGD Peptide Ligands-Design of New Molecular Conjugates for Imaging and Therapy of Cancers, Anti-Cancer Agents in Medicinal Chemistry, vol.7, issue.5, pp.552-558, 2007.
DOI : 10.2174/187152007781668706

S. Liu, Targeted Radiotracers for Tumor Imaging, Molecular Pharmaceutics, vol.3, issue.5, pp.472-487, 2006.
DOI : 10.1021/mp060049x

P. Dumy, M. Favrot, D. Boturyn, J. Coll, K. Altmann et al., Synthesis and characterization of novel systems for guidance and vectorization of compounds having a therapeutic activity Vuilleumier, S. Strategies for the de novo design of proteins Non-native architectures in protein design and mimicry, Brevet PCT Tetrahedron Cell Mol Life Sci, vol.135, issue.53, pp.771-785, 1988.

P. Dumy, I. M. Eggleston, S. Cervigni, U. Sila, X. Sun et al., A convenient synthesis of cyclic peptides as regioselectively addressable functionalized templates (RAFT), Tetrahedron Letters, vol.36, issue.8, pp.1255-1258, 1995.
DOI : 10.1016/0040-4039(94)02481-P

E. Garanger, D. Boturyn, O. Renaudet, E. Defrancq, and P. Dumy, Chemoselectively Addressable Template:?? A Valuable Tool for the Engineering of Molecular Conjugates, The Journal of Organic Chemistry, vol.71, issue.6, pp.2402-2410, 2006.
DOI : 10.1021/jo0525480

D. Boturyn, E. Defrancq, G. T. Dolphin, J. Garcia, P. Labbe et al., RAFT Nano-constructs: surfing to biological applications, Journal of Peptide Science, vol.22, issue.2, pp.224-240, 2008.
DOI : 10.1002/psc.964

S. Futaki, Peptide ion channels: Design and creation of function, Biopolymers, vol.47, issue.1, pp.75-81, 1998.
DOI : 10.1002/(SICI)1097-0282(1998)47:1<75::AID-BIP8>3.3.CO;2-1

G. Tuchscherer, C. Servis, G. Corradin, U. Blum, J. Rivier et al., Total chemical synthesis, characterization, and immunological properties of an MHC class I model using the TASP concept for protein de novo design Control of amyloid beta-peptide protofibril formation by a designed template assembly, Protein Sci Dolphin, G. T Angew Chem Int Ed Engl, vol.1, issue.45, pp.1377-1386, 1992.

S. Grigalevicius, S. Chierici, O. Renaudet, R. Lo-man, E. Deriaud et al., Chemoselective Assembly and Immunological Evaluation of Multiepitopic Glycoconjugates Bearing Clustered Tn Antigen as Synthetic Anticancer Vaccines, Bioconjugate Chemistry, vol.16, issue.5, pp.1149-1159, 2005.
DOI : 10.1021/bc050010v

P. Dumy, I. M. Eggleston, G. Esposito, S. Nicula, and M. Mutter, Solution structure of regioselectively addressable functionalized templates: An NMR and restrained molecular dynamics investigation, Biopolymers, vol.37, issue.12, pp.297-308, 1996.
DOI : 10.1002/(SICI)1097-0282(199609)39:3<297::AID-BIP3>3.0.CO;2-J

Z. H. Peng, Solid phase synthesis and NMR conformational studies on cyclic decapeptide template molecule, Biopolymers, vol.49, issue.7, pp.565-574, 1999.
DOI : 10.1002/(SICI)1097-0282(199906)49:7<565::AID-BIP3>3.3.CO;2-E

S. Peluso, T. Ruckle, C. Lehmann, M. Mutter, C. Peggion et al., Crystal structure of a synthetic cyclodecapeptide for template-assembled synthetic protein design, ChemBioChem 148. Katsara, M.; Tselios, T.; Deraos, S.; Deraos, G.; Matsoukas, M. T J, vol.2, pp.432-437, 2001.

V. Apostolopoulos, Round and round we go: cyclic peptides in disease, Curr Med Chem, vol.13, pp.2221-2232, 2006.

D. Boturyn and P. Dumy, A convenient access to ??V??3/??V??5 integrin ligand conjugates: regioselective solid-phase functionalisation of an RGD based peptide, Tetrahedron Letters, vol.42, issue.15, pp.2787-2790, 2001.
DOI : 10.1016/S0040-4039(01)00293-3

D. Boturyn, J. L. Coll, E. Garanger, M. C. Favrot, and P. Dumy, Template Assembled Cyclopeptides as Multimeric System for Integrin Targeting and Endocytosis, Journal of the American Chemical Society, vol.126, issue.18, pp.5730-5739, 2004.
DOI : 10.1021/ja049926n

E. Garanger and . Conception, synthèse et caractérisation de nouveaux systèmes de guidage et de vectorisation pour la cancérologie. 280 p. Thèse: Chimie-Biologie Université Joseph Fourier Multivalent RGD synthetic peptides as potent alphav beta3 integrin ligands, Org Biomol Chem, vol.4, 1958.

E. Garanger, D. Boturyn, Z. Jin, P. Dumy, M. C. Favrot et al., New Multifunctional Molecular Conjugate Vector for Targeting, Imaging, and Therapy of Tumors, Molecular Therapy, vol.12, issue.6, pp.1168-1175, 2005.
DOI : 10.1016/j.ymthe.2005.06.095

L. Sancey, V. Ardisson, L. M. Riou, M. Ahmadi, D. Marti-batlle et al., In vivo imaging of tumour angiogenesis in mice with the ??v??3 integrin-targeted tracer 99mTc-RAFT-RGD, European Journal of Nuclear Medicine and Molecular Imaging, vol.18, issue.Pt 19, pp.2037-2047, 2007.
DOI : 10.1007/s00259-007-0497-z

URL : https://hal.archives-ouvertes.fr/inserm-00176669

I. Texier, J. Coll, P. Dumy, D. Boturyn, and M. Favrot, Target vector with activable imaging function Activatable fluorescent probes for tumour-targeting imaging in live mice, Brevet PCT J. L. ChemMedChem, vol.156, issue.1, pp.1069-1072, 1749.

S. Su, D. E. Acquilano, J. Arumugasamy, A. B. Beeler, E. L. Eastwood et al., Convergent Synthesis of a Complex Oxime Library Using Chemical Domain Shuffling, Organic Letters, vol.7, issue.13, pp.2751-2754, 2005.
DOI : 10.1021/ol051023r

K. D. Roberts, J. N. Lambert, N. J. Ede, and A. M. Bray, Preparation of cyclic peptide libraries using intramolecular oxime formation, Journal of Peptide Science, vol.22, issue.11, pp.659-665, 2004.
DOI : 10.1002/psc.575

O. P. Edupuganti, E. Defrancq, and P. Dumy, Head-to-Tail Oxime Cyclization of Oligodeoxynucleotides for the Efficient Synthesis of Circular DNA Analogues, The Journal of Organic Chemistry, vol.68, issue.22, pp.8708-8710, 2003.
DOI : 10.1021/jo035064h

H. C. Hang and C. R. Bertozzi, Chemoselective Approaches to Glycoprotein Assembly, Accounts of Chemical Research, vol.34, issue.9, pp.727-736, 2001.
DOI : 10.1021/ar9901570

X. Chen, G. S. Lee, A. Zettl, and C. R. Bertozzi, Biomimetic Engineering of Carbon Nanotubes by Using Cell Surface Mucin Mimics, Angewandte Chemie International Edition, vol.191, issue.45, pp.6111-6116, 2004.
DOI : 10.1002/anie.200460620

G. G. Kochendoerfer and S. B. Kent, Chemical protein synthesis, Current Opinion in Chemical Biology, vol.3, issue.6, pp.665-671, 1999.
DOI : 10.1016/S1367-5931(99)00024-1

K. F. Geoghegan and J. G. Stroh, Site-directed conjugation of nonpeptide groups to peptides and proteins via periodate oxidation of a 2-amino alcohol. Application to modification at N-terminal serine, Bioconjugate Chemistry, vol.3, issue.2, pp.138-146, 1992.
DOI : 10.1021/bc00014a008

P. Lloyd-williams, F. Albericio, E. Giralt, S. Sakakibara, Y. Shimonishi et al., Chemical Approaches to the Synthesis of Peptides and Proteins Use of anhydrous hydrogen fluoride in peptide synthesis. I. Behavior of various protective groups in anhydrous hydrogen fluoride Amide protection and amide supports in solid-phase peptide synthesis, Bull Chem Soc Jpn Pietta, P. G. Journal of the Chemical Society, vol.40, issue.11, pp.278-169, 1967.

G. R. Matsueda and J. M. Stewart, A p-methylbenzhydrylamine resin for improved solid-phase synthesis of peptide amides, Peptides, vol.2, issue.1, pp.45-50, 1981.
DOI : 10.1016/S0196-9781(81)80010-1

S. S. Wang, -Alkoxybenzyloxycarbonylhydrazide Resin for Solid Phase Synthesis of Protected Peptide Fragments, Journal of the American Chemical Society, vol.95, issue.4, pp.1328-1333, 1973.
DOI : 10.1021/ja00785a602

URL : https://hal.archives-ouvertes.fr/jpa-00209906

E. Atherton, D. L. Clive, and R. C. Sheppard, Polyamide supports for polypeptide synthesis, Journal of the American Chemical Society, vol.97, issue.22, pp.6584-6585, 1975.
DOI : 10.1021/ja00855a053

J. Rivier, R. Wolbers, and R. Burgus, Application of high pressure liquid chromatography to peptides. Pept, Proc. Am. Pept. Symp., 5th, pp.52-55, 1977.

G. Barany and R. B. Merrifield, A new amino protecting group removable by reduction. Chemistry of the dithiasuccinoyl (Dts) function, Journal of the American Chemical Society, vol.99, issue.22, pp.7363-7365, 1977.
DOI : 10.1021/ja00464a050

C. D. Chang and J. Meienhofer, SOLID-PHASE PEPTIDE SYNTHESIS USING MILD BASE CLEAVAGE OF N??FLUORENYLMETHYLOXYCARBONYLAMINO ACIDS, EXEMPLIFIED BY A SYNTHESIS OF DIHYDROSOMATOSTATIN, International Journal of Peptide and Protein Research, vol.7, issue.3, pp.246-249, 1978.
DOI : 10.1111/j.1399-3011.1978.tb02845.x

H. M. Geysen, R. H. Meloen, and S. J. Barteling, Use of peptide synthesis to probe viral antigens for epitopes to a resolution of a single amino acid., Proceedings of the National Academy of Sciences, vol.81, issue.13, pp.3998-4002, 1984.
DOI : 10.1073/pnas.81.13.3998

R. A. Houghten, General method for the rapid solid-phase synthesis of large numbers of peptides: specificity of antigen-antibody interaction at the level of individual amino acids., Proceedings of the National Academy of Sciences, vol.82, issue.15, pp.5131-5135, 1985.
DOI : 10.1073/pnas.82.15.5131

I. Sucholeiki, New developments in solid phase synthesis supports Annual Reports in Combinatorial Chemistry and Molecular Diversity, pp.9-14, 1999.

F. Albericio and G. Barany, An acid-labile anchoring linkage for solid-phase synthesis of C-terminal peptide amides under mild conditions*, International Journal of Peptide and Protein Research, vol.34, issue.7, pp.206-216, 1987.
DOI : 10.1111/j.1399-3011.1987.tb03328.x

W. Stuber, J. Knolle, and G. Breipohl, Synthesis of peptide amides by Fmoc-solid-phase peptide synthesis and acid labile anchor groups, International Journal of Peptide and Protein Research, vol.111, issue.3, pp.215-221, 1989.
DOI : 10.1111/j.1399-3011.1989.tb00233.x

P. Sieber, An improved method for anchoring of 9-fluorenylmethoxycarbonyl-amino acids to 4-alkoxybenzyl alcohol resins., Tetrahedron Letters, vol.28, issue.49, pp.6147-6150, 1987.
DOI : 10.1016/S0040-4039(00)61832-4

M. Mergler, R. Tanner, J. Gosteli, P. Grogg, W. Schaefer et al., Peptide synthesis by a combination of solid-phase and solution methods. I. A new very acid-labile anchor group for the solid phase synthesis of fully protected fragments Esterification of partially protected peptide fragments with resins. Synthesis of Leu15-gastrin I using 2-chlorotrityl chloride resin, Tetrahedron Lett Barlos, K.; Gatos, D.; Kapolos, S.; Papaphotiu, G Tetrahedron Lett, vol.29, issue.30, pp.4005-4008, 1988.

G. Jung, Combinatorial chemistry : synthesis, chemistry, screening Rapid microwave-assisted solid phase peptide synthesis, Synthesis, vol.11, pp.1592-1596, 1999.
DOI : 10.1002/9783527613502

P. White, J. W. Keyte, K. Bailey, and G. Bloomberg, Expediting the Fmoc solid phase synthesis of long peptides through the application of dimethyloxazolidine dipeptides, Journal of Peptide Science, vol.10, issue.1, pp.18-26, 2004.
DOI : 10.1002/psc.484

O. Melnyk, J. S. Fruchart, C. Grandjean, and H. Gras-masse, Tartric Acid-Based Linker for the Solid-Phase Synthesis of C-Terminal Peptide ??-Oxo Aldehydes, The Journal of Organic Chemistry, vol.66, issue.12, pp.4153-4160, 2001.
DOI : 10.1021/jo001509f

J. C. Spetzler and T. Hoeg-jensen, Masked side-chain aldehyde amino acids for solid-phase synthesis and ligation, Tetrahedron Letters, vol.43, issue.12, pp.2303-2306, 2002.
DOI : 10.1016/S0040-4039(02)00230-7

J. M. Garcia, S. Far, E. Diesis, and O. Melnyk, Determination of glyoxylyl-peptide concentration using oxime chemistry and RP-HPLC analysis, Journal of Peptide Science, vol.2, issue.11, pp.734-738, 2006.
DOI : 10.1002/psc.791

H. F. Gaertner, K. Rose, R. Cotton, D. Timms, R. Camble et al., Construction of protein analogues by site-specific condensation of unprotected fragments, Bioconjug Chem, vol.3, pp.262-268, 1992.
DOI : 10.1007/978-94-011-1470-7_95

I. Lang, N. Donze, P. Garrouste, P. Dumy, and M. Mutter, Chemoselectively addressable HCan building blocks in peptide synthesis:L-homocanaline derivatives, Journal of Peptide Science, vol.39, issue.1, pp.72-80, 1998.
DOI : 10.1002/(SICI)1099-1387(199802)4:1<72::AID-PSC130>3.0.CO;2-G

J. C. Spetzler and T. Hoeg-jensen, Preparation and application ofO-amino-serine, Ams, a new building block in chemoselective ligation chemistry, Journal of Peptide Science, vol.54, issue.12, pp.582-592, 1999.
DOI : 10.1002/(SICI)1099-1387(199912)5:12<582::AID-PSC228>3.0.CO;2-Z

A. R. Khomutov, J. Vepsalainen, A. S. Shvetsov, T. Hyvonen, T. Keinanen et al., Synthesis of hydroxylamine analogues of polyamines, Tetrahedron, vol.52, issue.43, pp.13751-13766, 1996.
DOI : 10.1016/0040-4020(96)00836-8

J. Brask and K. J. Jensen, Carbopeptides: chemoselective ligation of peptide aldehydes to an aminooxy-functionalized D-galactose template, Journal of Peptide Science, vol.6, issue.6, pp.290-299, 2000.
DOI : 10.1002/1099-1387(200006)6:6<290::AID-PSC257>3.0.CO;2-L

J. Coste, D. Le-nguyen, and B. Castro, PyBOP??: A new peptide coupling reagent devoid of toxic by-product, Tetrahedron Letters, vol.31, issue.2, pp.205-208, 1990.
DOI : 10.1016/S0040-4039(00)94371-5

E. Kaiser, R. L. Colescott, C. D. Bossinger, and P. Cook, Color test for detection of free terminal amino groups in the solid-phase synthesis of peptides, Analytical Biochemistry, vol.34, issue.2, pp.595-598, 1970.
DOI : 10.1016/0003-2697(70)90146-6

W. S. Hancock and J. Battersby, A new micro-test for the detection of incomplete coupling reactions in solid-phase peptide synthesis using 2,4,6-trinitrobenzene-sulphonic acid, Analytical Biochemistry, vol.71, issue.1, pp.260-264, 1976.
DOI : 10.1016/0003-2697(76)90034-8

S. Foillard, M. Ohsten-rasmussen, J. Razkin, D. Boturyn, P. Dumy et al., 1-Ethoxyethylidene, a New Compatible Protecting Group for the Stepwise SPPS of Aminooxy Peptides Solid-phase functionalization of peptides by an alpha-hydrazinoacetyl group, J Org Chem Bonnet, D Bourel-Bonnet, L J Org Chem, vol.73, issue.68, pp.983-991, 2003.

N. Thieret, J. Alsina, E. Giralt, F. Guibe, and F. Albericio, Use of Alloc-amino acids in solid-phase peptide synthesis. Tandem deprotection-coupling reactions using neutral conditions, Tetrahedron Letters, vol.38, issue.41, pp.7275-7278, 1997.
DOI : 10.1016/S0040-4039(97)01690-0

R. Haubner, D. Finsinger, and H. Kessler, Stereoisomeric Peptide Libraries and Peptidomimetics for Designing Selective Inhibitors of the??v??3 Integrin for a New Cancer Therapy, Angewandte Chemie International Edition in English, vol.36, issue.1314, pp.1374-1389, 1997.
DOI : 10.1002/anie.199713741

S. Houdier, M. Legrand, D. Boturyn, S. Croze, E. Defrancq et al., A new fluorescent probe for sensitive detection of carbonyl compounds Molecular Expression Endosomes and Endocytosis Quantification of the efficiency of cargo delivery by peptidic and pseudo-peptidic Trojan carriers using MALDI-TOF mass spectrometry, Chassaing, G. Quantification of the cellular uptake of cellpenetrating peptides by MALDI-TOF mass spectrometry, pp.253-263, 1999.

S. Girault, G. Chassaing, J. C. Blais, A. Brunot, and G. Bolbach, Coupling of MALDI-TOF Mass Analysis to the Separation of Biotinylated Peptides by Magnetic Streptavidin Beads, Analytical Chemistry, vol.68, issue.13, pp.2122-2126, 1996.
DOI : 10.1021/ac960043r

J. H. Flanagan, . Jr, S. H. Khan, S. Menchen, S. A. Soper et al., Functionalized Tricarbocyanine Dyes as Near-Infrared Fluorescent Probes for Biomolecules, Bioconjugate Chemistry, vol.8, issue.5, pp.751-756, 1997.
DOI : 10.1021/bc970113g

W. Leung and R. P. Haugland, Alexa Dyes, a Series of New Fluorescent Dyes that Yield Exceptionally Bright, Photostable Conjugates, J Histochem Cytochem, vol.47, pp.1179-1188, 1999.

S. Freitag, I. Le-trong, L. Klumb, P. S. Stayton, and R. E. Stenkamp, Structural studies of the streptavidin binding loop, Protein Sci, vol.6, pp.1157-1166, 1997.

G. Saito, J. A. Swanson, and K. D. Lee, Drug delivery strategy utilizing conjugation via reversible disulfide linkages: role and site of cellular reducing activities, Advanced Drug Delivery Reviews, vol.55, issue.2, pp.199-215, 2000.
DOI : 10.1016/S0169-409X(02)00179-5

C. R. Lombardo, R. Rao, E. Ruoslahti, D. E. Bredesen, and R. Pasqualini, Anti-cancer activity of targeted pro-apoptotic peptides, Nat Med, vol.5, pp.1032-1038, 1999.

M. M. Javadpour, M. M. Juban, W. C. Lo, S. M. Bishop, J. B. Alberty et al., De Novo Antimicrobial Peptides with Low Mammalian Cell Toxicity, Journal of Medicinal Chemistry, vol.39, issue.16, pp.3107-3113, 1996.
DOI : 10.1021/jm9509410

D. Rio, G. Castro-obregon, S. Rao, R. Ellerby, H. M. Bredesen et al., APAP, a sequence-pattern recognition approach identifies substance P as a potential apoptotic peptide Peptide nucleic acid (PNA): its medical and biotechnical applications and promise for the future, FEBS Lett Ray, A. Faseb J, vol.494, issue.14, pp.213-219, 2000.

P. E. Nielsen, M. Egholm, R. H. Berg, O. B. Buchardt, and E. E. Swayze, Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide, 223. Clinicaltrials.Gov G3139, Oblimersen, pp.1497-1500, 1991.
DOI : 10.1126/science.1962210

R. H. Griffey, C. F. Bennett, and M. A. Maier, Structure-activity relationship study on a simple cationic peptide motif for cellular delivery of antisense peptide nucleic acid, J Med Chem, vol.48, pp.6741-6749, 2005.

Z. N. Oltvai, C. L. Milliman, and S. J. Korsmeyer, Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programed cell death, Cell, vol.74, issue.4, pp.609-619, 1993.
DOI : 10.1016/0092-8674(93)90509-O

F. E. Cotter, P. Johnson, P. Hall, C. Pocock, N. Mahdi et al., Antisense oligonucleotides suppress B-cell lymphoma growth in a SCID-hu mouse model, Oncogene, vol.9, pp.3049-3055, 1994.

L. Mologni, P. E. Nielsen, and C. Gambacorti-passerini, In Vitro Transcriptional and Translational Block of the bcl-2 Gene Operated by Peptide Nucleic Acid, Biochemical and Biophysical Research Communications, vol.264, issue.2, pp.537-543, 1999.
DOI : 10.1006/bbrc.1999.1548

S. I. Pai, Y. Y. Lin, B. Macaes, A. Meneshian, C. F. Hung et al., Prospects of RNA interference therapy for cancer, Gene Therapy, vol.33, issue.6, pp.464-477, 2006.
DOI : 10.1038/sj.gt.3302694

J. Yang, H. Chen, I. R. Vlahov, J. X. Cheng, and P. S. Low, Evaluation of disulfide reduction during receptor-mediated endocytosis by using FRET imaging, Proceedings of the National Academy of Sciences, vol.103, issue.37, pp.13872-13877, 2006.
DOI : 10.1073/pnas.0601455103

K. S. Lam, R. Liu, S. Miyamoto, A. L. Lehman, and J. M. Tuscano, Applications of One-Bead One-Compound Combinatorial Libraries and Chemical Microarrays in Signal Transduction Research, Accounts of Chemical Research, vol.36, issue.6, pp.370-377, 2003.
DOI : 10.1021/ar0201299

C. Gibson, G. A. Sulyok, D. Hahn, S. L. Goodman, G. Holzemann et al., Nonpeptidic ??v??3 Integrin Antagonist Libraries: On-Bead Screening and Mass Spectrometric Identification without Tagging, Angewandte Chemie International Edition, vol.3, issue.1, pp.165-169, 2001.
DOI : 10.1002/1521-3773(20010105)40:1<165::AID-ANIE165>3.0.CO;2-8

L. Ying and J. Gervay-hague, One-Bead-One-Inhibitor-One-Substrate Screening of Neuraminidase Activity, ChemBioChem, vol.11, issue.10, pp.1857-1865, 2005.
DOI : 10.1002/cbic.200500006

S. Dixon, K. T. Ziebart, Z. He, M. Jeddeloh, C. L. Yoo et al., Aminodeoxychorismate Synthase Inhibitors from One-Bead One-Compound Combinatorial Libraries:?? ???Staged??? Inhibitor Design, Journal of Medicinal Chemistry, vol.49, issue.25, pp.7413-7426, 2006.
DOI : 10.1021/jm0609869

F. Sebestyén, G. Dibó, A. Kovács, and A. Furkua, Chemical synthesis of peptide libraries, Bioorganic & Medicinal Chemistry Letters, vol.3, issue.3, pp.413-418, 1993.
DOI : 10.1016/S0960-894X(01)80223-7

J. A. Kritzer, N. W. Luedtke, E. A. Harker, and A. Schepartz, A Rapid Library Screen for Tailoring ??-Peptide Structure and Function, Journal of the American Chemical Society, vol.127, issue.42, pp.14584-14585, 2005.
DOI : 10.1021/ja055050o

S. Aggarwal, J. L. Harden, and S. R. Denmeade, Synthesis and Screening of a Random Dimeric Peptide Library Using the One-Bead???One-Dimer Combinatorial Approach, Bioconjugate Chemistry, vol.17, issue.2, pp.335-340, 2006.
DOI : 10.1021/bc0502659

O. H. Aina, J. Marik, R. Liu, D. H. Lau, and K. S. Lam, Identification of novel targeting peptides for human ovarian cancer cells using "one-bead one-compound" combinatorial libraries, Molecular Cancer Therapeutics, vol.4, issue.5, pp.806-813, 2005.
DOI : 10.1158/1535-7163.MCT-05-0029

E. C. Johnson, T. Durek, and S. B. Kent, Total Chemical Synthesis, Folding, and Assay of a Small Protein on a Water-Compatible Solid Support, Angewandte Chemie International Edition, vol.8, issue.20, pp.3283-3287, 2006.
DOI : 10.1002/anie.200600381

F. Albericio, M. Cases, J. Alsina, S. A. Triolo, L. A. Carpino et al., On the use of PyAOP, a phosphonium salt derived from HOAt, in solid-phase peptide synthesis, Tetrahedron Letters, vol.38, issue.27, pp.4853-4856, 1997.
DOI : 10.1016/S0040-4039(97)01011-3

C. A. Montalbetti and V. Falque, Amide bond formation and peptide coupling, Tetrahedron, pp.61-10827, 2005.

S. Aggarwal, S. Janssen, R. M. Wadkins, J. L. Harden, and S. R. Denmeade, A combinatorial approach to the selective capture of circulating malignant epithelial cells by peptide ligands, Biomaterials, vol.26, issue.30, pp.6077-6086, 2005.
DOI : 10.1016/j.biomaterials.2005.03.040

D. H. Lau, L. Guo, R. Liu, A. Song, C. Shao et al., Identifying peptide ligands for cell surface receptors using cell-growth-on-bead assay and one-bead one-compound combinatorial library, Biotechnology Letters, vol.24, issue.6, pp.497-500, 2002.
DOI : 10.1023/A:1014538322185