M. Malvezzi, G. Carioli, P. Bertuccio, T. Rosso, P. Boffetta et al., European cancer mortality predictions for the year 2016 with focus on leukaemias, Annals of Oncology, vol.27, issue.4, pp.27-725, 2016.
DOI : 10.1093/annonc/mdw022

R. L. Siegel, K. D. Miller, and A. , Cancer statistics, 2016, Cancer statistics, pp.7-30, 2016.
DOI : 10.1111/obr.12321

A. Fusi, G. Procopio, S. Della-torre, R. Ricotta, G. Bianchini et al., Treatment options in hormonerefractory metastatic prostate carcinoma, Tumori, vol.90, pp.535-546, 2004.

D. P. Petrylak, C. M. Tangen, M. H. Hussain, P. N. Lara-jr, J. A. Jones et al., Docetaxel and Estramustine Compared with Mitoxantrone and Prednisone for Advanced Refractory Prostate Cancer, New England Journal of Medicine, vol.351, issue.15, pp.351-1513, 2004.
DOI : 10.1056/NEJMoa041318

I. F. Tannock, R. De-wit, W. R. Berry, J. Horti, A. Pluzanska et al., Docetaxel plus Prednisone or Mitoxantrone plus Prednisone for Advanced Prostate Cancer, New England Journal of Medicine, vol.351, issue.15, pp.351-1502, 2004.
DOI : 10.1056/NEJMoa040720

R. J. Van-soest, E. S. De-morree, L. Shen, I. F. Tannock, M. A. Eisenberger et al., Initial Biopsy Gleason Score as a Predictive Marker for Survival Benefit in Patients with Castration-resistant Prostate Cancer Treated with Docetaxel: Data from the TAX327 Study, European Urology, vol.66, issue.2, pp.66-330, 2014.
DOI : 10.1016/j.eururo.2013.08.007

V. Baylot, M. Katsogiannou, C. Andrieu, D. Taieb, J. Acunzo et al., Targeting TCTP as a New Therapeutic Strategy in Castration-resistant Prostate Cancer, Molecular Therapy, vol.20, issue.12, pp.2244-2256, 2012.
DOI : 10.1038/mt.2012.155

M. Gnanasekar, S. Thirugnanam, G. Zheng, A. Chen, and K. Ramaswamy, Gene silencing of translationally controlled tumor protein (TCTP) by siRNA inhibits cell growth and induces apoptosis of human prostate cancer cells, International Journal of Oncology, pp.34-1241, 2009.
DOI : 10.3892/ijo_00000252

M. Kaarbo, M. L. Storm, S. Qu, H. Waehre, B. Risberg et al., TCTP Is an Androgen-Regulated Gene Implicated in Prostate Cancer, PLoS ONE, vol.105, issue.7, p.69398, 2013.
DOI : 10.1371/journal.pone.0069398.g007

B. J. Rocca, A. Ginori, A. Barone, C. Calandra, F. Crivelli et al., Translationally Controlled Tumor Protein in Prostatic Adenocarcinoma: Correlation with Tumor Grading and Treatment-Related Changes, BioMed Research International, vol.59, pp.2015-985950, 2015.
DOI : 10.1186/1746-1596-7-90

J. Acunzo, V. Baylot, A. So, and P. Rocchi, TCTP as therapeutic target in cancers, Cancer Treatment Reviews, vol.40, issue.6, pp.760-769, 2014.
DOI : 10.1016/j.ctrv.2014.02.007

U. A. Bommer and B. J. Thiele, The translationally controlled tumour protein (TCTP), The International Journal of Biochemistry & Cell Biology, vol.36, issue.3, pp.379-385, 2004.
DOI : 10.1016/S1357-2725(03)00213-9

S. M. Macdonald, Potential role of histamine releasing factor (HRF) as a therapeutic target for treating asthma and allergy, Journal of Asthma and Allergy, vol.5, pp.51-59, 2012.
DOI : 10.2147/JAA.S28868

S. M. Macdonald, T. Rafnar, J. Langdon, and L. M. Lichtenstein, Molecular identification of an IgE-dependent histamine-releasing factor, Science, vol.269, issue.5224, pp.688-690, 1995.
DOI : 10.1126/science.7542803

M. Nagano-ito and S. Ichikawa, Biological Effects of Mammalian Translationally Controlled Tumor Protein (TCTP) on Cell Death, Proliferation, and Tumorigenesis, Biochemistry Research International, vol.112, issue.8, p.2012, 2012.
DOI : 10.1074/jbc.M109.012823

R. Amson, S. Pece, J. C. Marine, P. P. Di-fiore, and A. Telerman, TPT1/ TCTP-regulated pathways in phenotypic reprogramming, Trends in Cell Biology, vol.23, issue.1, pp.37-46, 2013.
DOI : 10.1016/j.tcb.2012.10.002

URL : http://hdl.handle.net/2434/213480

M. Tuynder, G. Fiucci, S. Prieur, A. Lespagnol, A. Geant et al., Translationally controlled tumor protein is a target of tumor reversion, Proceedings of the National Academy of Sciences, vol.16, issue.16, pp.15364-15369, 2004.
DOI : 10.1038/sj.onc.1201729

P. M. Moreno and A. P. Pego, Therapeutic antisense oligonucleotides against cancer: hurdling to the clinic, Frontiers in Chemistry, vol.18, issue.83, 2014.
DOI : 10.1038/gt.2010.133

URL : http://doi.org/10.3389/fchem.2014.00087

J. K. Watts and D. R. Corey, Silencing disease genes in the laboratory and the clinic, The Journal of Pathology, vol.70, issue.2, pp.365-379, 2012.
DOI : 10.1002/path.2993

J. Winkler, Oligonucleotide conjugates for therapeutic applications, Therapeutic Delivery, vol.4, issue.7, pp.791-809, 2013.
DOI : 10.4155/tde.13.47

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3787477

W. R. Epa, P. Rong, P. F. Bartlett, E. J. Coulson, and G. L. Barrett, Enhanced Downregulation of the p75 Nerve Growth Factor Receptor by Cholesteryl and Bis-Cholesteryl Antisense Oligonucleotides, Antisense and Nucleic Acid Drug Development, vol.8, issue.6, pp.489-498, 1998.
DOI : 10.1089/oli.1.1998.8.489

G. Godeau, C. Staedel, and P. Barthélémy, Lipid-Conjugated Oligonucleotides via ???Click Chemistry??? Efficiently Inhibit Hepatitis C Virus Translation, Journal of Medicinal Chemistry, vol.51, issue.15, pp.51-4374, 2008.
DOI : 10.1021/jm800518u

S. Ellipilli, R. Murthy, and K. N. Ganesh, Perfluoroalkylchain conjugation as a new tactic for enhancing cell permeability of peptide nucleic acids (PNAs) via reducing the nanoparticle size, Chem. Commun., vol.3, issue.3, pp.52-521, 2016.
DOI : 10.1039/C5CC05342K

G. Godeau, H. Arnion, C. Brun, C. Staedel, and P. Barthélémy, Fluorocarbon oligonucleotide conjugates for nucleic acids delivery, MedChemComm, vol.31, issue.1, pp.76-78, 2010.
DOI : 10.1039/c0md00054j

A. Aime, N. Beztsinna, A. Patwa, A. Pokolenko, I. Bestel et al., Quantum Dot Lipid Oligonucleotide Bioconjugates: Toward a New Anti-MicroRNA Nanoplatform, Bioconjugate Chemistry, vol.24, issue.8, pp.24-1345, 2013.
DOI : 10.1021/bc400157z

A. Gissot, C. Di-primo, I. Bestel, G. Giannone, H. Chapuis et al., Sensitive liposomes encoded with oligonucleotide amphiphiles: a biocompatible switch, Chemical Communications, vol.27, issue.43, pp.5550-5552, 2008.
DOI : 10.1039/b812398e

A. Pokolenko, A. Gissot, B. Vialet, K. Bathany, T. Alain et al., Lipid oligonucleotide conjugates as responsive nanomaterials for drug delivery, Journal of Materials Chemistry B, vol.40, issue.39, pp.5329-5334, 2013.
DOI : 10.1039/c3tb20357c

F. Li, D. Zhang, and K. Fujise, Characterization of Fortilin, a Novel Antiapoptotic Protein, Journal of Biological Chemistry, vol.276, issue.50, pp.47542-47549, 2001.
DOI : 10.1074/jbc.M108954200

M. Tuynder, L. Susini, S. Prieur, S. Besse, G. Fiucci et al., Biological models and genes of tumor reversion: Cellular reprogramming through tpt1/TCTP and SIAH-1, Proceedings of the National Academy of Sciences, vol.276, issue.50, pp.99-14976, 2002.
DOI : 10.1074/jbc.M108954200

F. Arcuri, S. Papa, A. Carducci, R. Romagnoli, S. Liberatori et al., Translationally controlled tumor protein (TCTP) in the human prostate and prostate cancer cells: Expression, distribution, and calcium binding activity, The Prostate, vol.2, issue.3, pp.130-140, 2004.
DOI : 10.1002/pros.20054

Z. Liu, S. Bengtsson, M. Krogh, M. Marquez, S. Nilsson et al., Somatostatin effects on the proteome of the LNCaP cell-line, International Journal of Oncology, pp.30-1173, 2007.
DOI : 10.3892/ijo.30.5.1173

C. F. Bennett, M. Y. Chiang, H. Chan, J. E. Shoemaker, and C. K. Mirabelli, Cationic lipids enhance cellular uptake and activity of phosphorothioate antisense oligonucleotides, Mol. Pharmacol, pp.41-1023, 1992.

G. D. Gray, S. Basu, and E. Wickstrom, Transformed and immortalized cellular uptake of oligodeoxynucleoside phosphorothioates, 3???-Alkylamino oligodeoxynucleotides, 2???-o-methyl oligoribonucleotides, oligodeoxynucleoside methylphosphonates, and peptide nucleic acids, Biochemical Pharmacology, vol.53, issue.10, pp.53-1465, 1997.
DOI : 10.1016/S0006-2952(97)82440-9

C. A. Stein, J. B. Hansen, J. Lai, S. Wu, A. Voskresenskiy et al., Efficient gene silencing by delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents, Nucleic Acids Research, vol.38, issue.1, p.3, 2010.
DOI : 10.1093/nar/gkp841

S. L. Loke, C. A. Stein, X. H. Zhang, K. Mori, M. Nakanishi et al., Characterization of oligonucleotide transport into living cells., Proceedings of the National Academy of Sciences, vol.86, issue.10, pp.86-3474, 1989.
DOI : 10.1073/pnas.86.10.3474

L. A. Yakubov, E. A. Deeva, V. F. Zarytova, E. M. Ivanova, A. S. Ryte et al., Mechanism of oligonucleotide uptake by cells: involvement of specific receptors?, Proceedings of the National Academy of Sciences, vol.86, issue.17, pp.86-6454, 1989.
DOI : 10.1073/pnas.86.17.6454

E. Koller, T. M. Vincent, A. Chappell, S. De, M. Manoharan et al., Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes, Nucleic Acids Research, vol.39, issue.11, pp.4795-4807, 2011.
DOI : 10.1093/nar/gkr089

C. Wolfrum, S. Shi, K. N. Jayaprakash, M. Jayaraman, G. Wang et al., Mechanisms and optimization of in vivo delivery of lipophilic siRNAs, Nature Biotechnology, vol.409, issue.10, pp.25-1149, 2007.
DOI : 10.1038/nbt1339

A. Wittrup, A. Ai, X. Liu, P. Hamar, R. Trifonova et al., Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown, Nature Biotechnology, vol.33, issue.8, pp.33-870, 2015.
DOI : 10.1172/JCI200215420

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4663660

T. H. Chan, L. Chen, and X. Y. Guan, Role of Translationally Controlled Tumor Protein in Cancer Progression, Biochemistry Research International, vol.2, issue.1, p.2012, 2012.
DOI : 10.1002/1522-2683(20000801)21:14<3048::AID-ELPS3048>3.0.CO;2-W

S. H. Chen, P. S. Wu, C. H. Chou, Y. T. Yan, H. Liu et al., A Knockout Mouse Approach Reveals that TCTP Functions as an Essential Factor for Cell Proliferation and Survival in a Tissue- or Cell Type-specific Manner, Molecular Biology of the Cell, vol.18, issue.7, pp.2525-2532, 2007.
DOI : 10.1091/mbc.E07-02-0188

P. P. Seth, A. Siwkowski, C. R. Allerson, G. Vasquez, S. Lee et al., Short Antisense Oligonucleotides with Novel 2??????4??? Conformationaly Restricted Nucleoside Analogues Show Improved Potency without Increased Toxicity in Animals, Journal of Medicinal Chemistry, vol.52, issue.1, pp.52-62, 2009.
DOI : 10.1021/jm801294h

R. Stanton, S. Sciabola, C. Salatto, Y. Weng, D. Moshinsky et al., Chemical modification study of antisense gapmers, Nucleic Acid Ther, pp.22-344, 2012.

E. E. Swayze, A. M. Siwkowski, E. V. Wancewicz, M. T. Migawa, T. K. Wyrzykiewicz et al., Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals, Nucleic Acids Research, vol.35, issue.2, pp.687-700, 2007.
DOI : 10.1093/nar/gkl1071

URL : http://doi.org/10.1093/nar/gkl1071

S. Karaki, Lipid-oligonucleotide conjugates improve cellular uptake and efficiency of TCTP-antisense in castration-resistant prostate cancer, Journal of Controlled Release, vol.258, pp.1-9, 2017.
DOI : 10.1016/j.jconrel.2017.04.042

URL : https://hal.archives-ouvertes.fr/inserm-01520102