H. Lodish, A. Berk, S. L. Zipursky, P. Matsudaira, D. Baltimore et al., Molecular Cell Biology, 2000.

A. M. Belikova, V. F. Zarytova, and N. I. Grineva, Synthesis of Ribonucleosides and Diribonucleoside Phosphates Containing 2-Chloroethylamine and Nitrogen Mustard Residues, Tetrahedron Lett, vol.37, pp.3557-3562, 1967.

J. Summerton and P. Bartlett, Sequence-Specific Crosslinking Agents for Nucleic Acids, J. Mol. Biol, vol.122, issue.2, pp.90032-90035, 1978.

P. C. Zamecnik and M. L. Stephenson, Inhibition of Rous Sarcoma Virus Replication and Cell Transformation by a Specific Oligodeoxynucleotide, Proc. Natl. Acad. Sci. U. S. A, vol.75, issue.1, pp.280-284, 1978.

M. L. Stephenson and P. C. Zamecnik, Inhibition of Rous Sarcoma Viral RNA Translation by a Specific Oligodeoxyribonucleotide, Proc. Natl. Acad. Sci. U. S. A, vol.75, issue.1, pp.285-288, 1978.

, Antisense Oligodeoxynucleotides and Antisense RNANovel Pharmacological and Therapeutic Agents, 1997.

C. Napoli, C. Lemieux, and R. Jorgensen, Introduction of a Chimeric Chalcone Synthase Gene into Petunia Results in Reversible Co-Suppression of Homologous Genes in Trans, Plant Cell, vol.2, issue.4, pp.279-289, 1990.

A. Fire, S. Xu, M. K. Montgomery, S. A. Kostas, S. E. Driver et al., Potent and Specific Genetic Interference by Double-Stranded RNA in Caenorhabditis Elegans, Nature, vol.391, issue.6669, pp.806-811, 1998.

B. F. Baker, S. S. Lot, T. P. Condon, S. Cheng-flournoy, E. A. Lesnik et al., -Methoxy)Ethyl-Modified Anti-Intercellular Adhesion Molecule 1 (ICAM-1) Oligonucleotides Selectively Increase the ICAM-1 MRNA Level and Inhibit Formation of the ICAM-1 Translation Initiation Complex in Human Umbilical Vein Endothelial Cells, J. Biol. Chem, issue.2, pp.11994-12000, 1997.

P. Lengyel, J. F. Speyer, and S. Ochoa, Synthetic Polynucleotides and the Amino Acid Code, Proc. Natl. Acad. Sci. U. S. A, vol.47, pp.1936-1942, 1961.

C. Cazenave, C. A. Stein, N. Loreau, N. T. Thuong, L. M. Neckers et al., Comparative Inhibition of Rabbit Globin MRNA Translation by Modified Antisense Oligodeoxynucleotides, Nucleic Acids Res, vol.17, issue.11, pp.4255-4273, 1989.

J. H. Chan, S. Lim, and W. S. Wong, Antisense Oligonucleotides: From Design to Therapeutic Application, Clin. Exp. Pharmacol. Physiol, vol.33, issue.5-6, pp.533-540, 2006.

T. A. Vickers, J. R. Wyatt, T. Burckin, C. F. Bennett, and S. M. Freier, Fully Modified 2' MOE Oligonucleotides Redirect Polyadenylation, Nucleic Acids Res, vol.29, issue.6, pp.1293-1299, 2001.

L. He, X. He, L. P. Lim, E. De-stanchina, Z. Xuan et al., A MicroRNA Component of the P53 Tumour Suppressor Network, Nature, vol.447, issue.7148, pp.1130-1134, 2007.

M. S. Kumar, S. J. Erkeland, R. E. Pester, C. Y. Chen, M. S. Ebert et al., Suppression of Non-Small Cell Lung Tumor Development by the Let-7 MicroRNA Family, Proc. Natl. Acad. Sci. U. S. A, issue.10, pp.3903-3908, 2008.

,

S. Davis, S. Propp, S. M. Freier, L. E. Jones, M. J. Serra et al., Potent Inhibition of MicroRNA in Vivo without Degradation, Nucleic Acids Res, vol.37, issue.1, pp.70-77, 2009.

C. Esau, X. Kang, E. Peralta, E. Hanson, E. G. Marcusson et al., Mi-croRNA-143 Regulates Adipocyte Differentiation, J. Biol. Chem, issue.50, pp.52361-52365, 2004.

,

J. C. Norton, M. A. Piatyszek, W. E. Wright, J. W. Shay, and D. R. Corey, Inhibition of Human Telomerase Activity by Peptide Nucleic Acids, Nat. Biotechnol, vol.14, issue.5, pp.615-619, 1996.

,

S. Agrawal, J. Temsamani, W. Galbraith, and J. Tang, Pharmacokinetics of Antisense Oligonucleotides, Clin. Pharmacokinet, vol.28, issue.1, pp.7-16, 1995.

P. P. Seth, C. R. Allerson, A. Berdeja, A. Siwkowski, P. S. Pallan et al., An Exocyclic Methylene Group Acts as a Bioisostere of the 2'-Oxygen Atom in LNA, J. Am. Chem. Soc, issue.42, pp.14942-14950, 2010.

A. Khvorova and J. K. Watts, The Chemical Evolution of Oligonucleotide Therapies of Clinical Utility, Nat. Biotechnol, vol.2017, issue.3, pp.238-248

M. Manoharan, RNA Interference and Chemically Modified Small Interfering RNAs, Curr. Opin. Chem. Biol, vol.8, issue.6, pp.570-579, 2004.

E. Zamaratski, P. I. Pradeepkumar, and J. Chattopadhyaya, A Critical Survey of the Structure-Function of the Antisense Oligo/RNA Heteroduplex as Substrate for RNase H, J. Biochem. Biophys. Methods, vol.48, issue.3, pp.189-208, 2001.

C. A. Stein, L. Benimetskaya, and S. Mani, Antisense Strategies for Oncogene Inactivation. Semin. Oncol, vol.32, issue.6, pp.563-572, 2005.

M. S. Kupryushkin, D. V. Pyshnyi, and D. A. Stetsenko, Phosphoryl Guanidines: A New Type of Nucleic Acid Analogues, Acta Naturae, vol.6, issue.4, pp.116-118, 2014.

D. A. Stetsenko, M. S. Kupryushkin, and D. V. Pyshnyi, Modified Oligonucleotides and Methods for Their Synthesis, 2016.

J. Summerton and D. Weller, Morpholino Antisense Oligomers: Design, Preparation, and Properties, Antisense Nucleic Acid Drug Dev, vol.7, issue.3, pp.187-195, 1997.

C. Tros-de-ilarduya, Y. Sun, and N. Düzgüne?, Gene Delivery by Lipoplexes and Polyplexes, Eur. J. Pharm. Sci, vol.2010, issue.3, pp.159-170

M. A. Mintzer and E. E. Simanek, Nonviral Vectors for Gene Delivery, Chem. Rev, vol.109, issue.2, pp.259-302, 2009.

,

H. Yin, R. L. Kanasty, A. A. Eltoukhy, A. J. Vegas, J. R. Dorkin et al., Non-Viral Vectors for Gene-Based Therapy, Nat. Rev. Genet, vol.15, issue.8, pp.541-555, 2014.

L. Wasungu, D. C. Hoekstra, and . Lipids, Lipoplexes and Intracellular Delivery of Genes, J. Control. Release Off. J. Control. Release Soc, vol.116, issue.2, pp.255-264, 2006.

C. E. Nelson, J. R. Kintzing, A. Hanna, J. M. Shannon, M. K. Gupta et al., Balancing Cationic and Hydrophobic Content of PEGylated SiRNA Polyplexes Enhances Endosome Escape, Stability, Blood Circulation Time, and Bioactivity In Vivo, ACS Nano, issue.7, 2013.

F. J. Verbaan, C. Oussoren, I. M. Van-dam, Y. Takakura, M. Hashida et al., The Fate of Poly(2-Dimethyl Amino Ethyl)Methacrylate-Based Polyplexes after Intravenous Administration, Int. J. Pharm, vol.214, issue.1, pp.642-648, 2001.

F. Sakurai, T. Terada, K. Yasuda, F. Yamashita, Y. Takakura et al., The Role of Tissue Macrophages in the Induction of Proinflammatory Cytokine Production Following Intravenous Injection of Lipoplexes, Gene Ther, vol.9, issue.16, pp.1120-1126, 2002.

,

P. Neuberg and A. Kichler, Recent Developments in Nucleic Acid Delivery with Polyethylenimines, Adv. Genet, vol.88, pp.263-288, 2014.

M. Chiper, N. Tounsi, R. Kole, A. Kichler, and G. Zuber, Self-Aggregating 1.8kDa Polyethylenimines with Dissolution Switch at Endosomal Acidic PH Are Delivery Carriers for Plasmid DNA, MRNA, SiRNA and Exon-Skipping Oligonucleotides, J. Controlled Release, vol.246, pp.60-70, 2017.

Y. Zhu, G. Liang, B. Sun, T. Tian, F. Hu et al., A Novel Type of Self-Assembled Nanoparticles as Targeted Gene Carriers: An Application for Plasmid DNA and AntimicroRNA Oligonucleotide Delivery, Int. J. Nanomedicine, vol.11, pp.399-411, 2016.

,

S. Agarwal, Y. Zhang, S. Maji, A. Greiner, and . Based, Gene Delivery Materials. Mater. Today, vol.2012, issue.9, pp.70165-70172

Y. Jiang, H. Lu, Y. Y. Khine, A. Dag, and M. H. Stenzel, Polyion Complex Micelle Based on Albumin-Polymer Conjugates: Multifunctional Oligonucleotide Transfection Vectors for Anticancer Chemotherapeutics, Biomacromolecules, vol.15, issue.11, pp.4195-4205, 2014.

,

V. A. Svyatchenko, M. V. Tarasova, S. V. Netesov, and P. M. Chumakov, Oncolytic Adenoviruses in Anticancer Therapy: Current Status and Prospects, Mol. Biol, vol.2012, issue.4, pp.496-507

,

A. N. Lukashev and A. A. Zamyatnin, Viral Vectors for Gene Therapy: Current State and Clinical Perspectives, Biochem. Biokhimiia, vol.81, issue.7, pp.700-708, 2016.

O. Merten and B. Gaillet, Viral Vectors for Gene Therapy and Gene Modification Approaches, Biochem. Eng. J, vol.108, pp.98-115, 2016.

Y. Zhao and L. Huang, Lipid Nanoparticles for Gene Delivery, Adv. Genet, vol.88, pp.13-36, 2014.

A. Okamoto, T. Asai, Y. Hirai, K. Shimizu, H. Koide et al., Systemic Administration of SiRNA with Anti-HB-EGF Antibody-Modified Lipid Nanoparticles for the Treatment of Triple-Negative Breast Cancer, Mol. Pharm, 2018.

J. Han, B. J. Zern, V. V. Shuvaev, P. F. Davies, S. Muro et al., Acute and Chronic Shear Stress Differently Regulate Endothelial Internalization of Nanocarriers Targeted to Platelet-Endothelial Cell Adhesion Molecule-1, ACS Nano, vol.6, issue.10, pp.8824-8836, 2012.

J. Han, V. V. Shuvaev, P. F. Davies, D. M. Eckmann, S. Muro et al., Flow Shear Stress Differentially Regulates Endothelial Uptake of Nanocarriers Targeted to Distinct Epitopes of PECAM-1, J. Control. Release Off. J. Control. Release Soc, vol.210, pp.39-47, 2015.

H. Parhiz, V. V. Shuvaev, N. Pardi, M. Khoshnejad, R. Y. Kiseleva et al., PECAM-1 Directed Re-Targeting of Exogenous MRNA Providing Two Orders of Magnitude Enhancement of Vascular Delivery and Expression in Lungs Independent of Apolipoprotein E-Mediated Uptake, J. Control. Release Off. J. Control. Release Soc, vol.291, pp.106-115, 2018.

J. Baillet, V. Desvergnes, A. Hamoud, L. Latxague, and P. Barthélémy, Lipid and Nucleic Acid Chemistries: Combining the Best of Both Worlds to Construct Advanced Biomaterials, Adv. Mater, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02374786

V. Allain, C. Bourgaux, and P. Couvreur, Self-Assembled Nucleolipids: From Supramolecular Structure to Soft Nucleic Acid and Drug Delivery Devices, Nucleic Acids Res, vol.40, issue.5, pp.1891-1903, 2012.

S. Khiati, D. Luvino, K. Oumzil, B. Chauffert, M. Camplo et al., Nucleoside-Lipid-Based Nanoparticles for Cisplatin Delivery, ACS Nano, vol.5, issue.11, pp.8649-8655, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00696838

,

P. Chabaud, M. Camplo, D. Payet, G. Serin, L. Moreau et al., Cationic Nucleoside Lipids for Gene Delivery, Bioconjug. Chem, vol.17, issue.2, pp.466-472, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00165573

,

A. Gissot, M. Camplo, M. W. Grinstaff, P. Barthélémy, and . Nucleoside, Nucleotide and Oligonucleotide Based Amphiphiles: A Successful Marriage of Nucleic Acids with Lipids, Org. Biomol. Chem, vol.6, issue.8, pp.1324-1333, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02484364

D. Luvino, S. Khiati, K. Oumzil, P. Rocchi, M. Camplo et al., Efficient Delivery of Therapeutic Small Nucleic Acids to Prostate Cancer Cells Using Ketal Nucleoside Lipid Nanoparticles, J. Control. Release Off. J. Control. Release Soc, vol.2013, issue.3, pp.954-961
URL : https://hal.archives-ouvertes.fr/hal-00975040

C. Ceballos, C. A. Prata, S. Giorgio, F. Garzino, D. Payet et al.,

M. R. Alam, X. Ming, M. Fisher, J. G. Lackey, K. G. Rajeev et al., Multivalent Cyclic RGD Conjugates for Targeted Delivery of Small Interfering RNA, Bioconjug. Chem, issue.8, pp.1673-1681, 2011.

R. L. Juliano, X. Ming, O. Nakagawa, R. Xu, and H. Yoo, Integrin Targeted Delivery of Gene Therapeutics, Theranostics, vol.1, pp.211-219, 2011.

M. Gooding, M. Malhotra, J. C. Evans, R. Darcy, and C. M. O'driscoll, Oligonucleotide Conjugates -Candidates for Gene Silencing Therapeutics, Eur. J. Pharm. Biopharm. Off. J. Arbeitsgemeinschaft Pharm. Verfahrenstechnik, vol.107, pp.321-340, 2016.

,

X. Ming, M. R. Alam, M. Fisher, Y. Yan, X. Chen et al., Intracellular Delivery of an Antisense Oligonucleotide via Endocytosis of a G Protein-Coupled Receptor, Nucleic Acids Res, issue.19, pp.6567-6576, 2010.

O. Nakagawa, X. Ming, K. Carver, and R. Juliano, Conjugation with Receptor-Targeted Histidine-Rich Peptides Enhances the Pharmacological Effectiveness of Antisense Oligonucleotides, Bioconjug. Chem, vol.25, issue.1, pp.165-170, 2014.

D. Lobeek, G. M. Franssen, M. T. Ma, H. Wester, C. Decristoforo et al., In Vivo Characterization of Four 68Ga-Labeled Multimeric RGD Peptides to Image ?v?3 Integrin Expression in Two Human Tumor Xenograft Mouse Models, J. Nucl. Med. Off. Publ. Soc. Nucl. Med, 2018.

S. K. Shaw, C. L. Schreiber, F. M. Roland, P. M. Battles, S. P. Brennan et al., High Expression of Integrin ?v?3 Enables Uptake of Targeted Fluorescent Probes into Ovarian Cancer Cells and Tumors, Bioorg. Med. Chem, vol.26, issue.8, pp.2085-2091, 2018.

J. Zhang, F. Mao, G. Niu, L. Peng, L. Lang et al., 68Ga-BBN-RGD PET/CT for GRPR and Integrin ?v?3 Imaging in Patients with, Breast Cancer, vol.8, issue.4, pp.1121-1130, 2018.

Y. Asami, K. Yoshioka, K. Nishina, T. Nagata, and T. Yokota, Drug Delivery System of Therapeutic Oligonucleotides, Drug Discov. Ther, vol.10, issue.5, pp.256-262, 2016.

K. Nishina, T. Unno, Y. Uno, T. Kubodera, T. Kanouchi et al., Efficient in Vivo Delivery of SiRNA to the Liver by Conjugation of Alpha-Tocopherol, Mol. Ther. J. Am. Soc

, Gene Ther, vol.16, issue.4, pp.734-740, 2008.

T. Nishina, J. Numata, K. Nishina, K. Yoshida-tanaka, K. Nitta et al., Chimeric Antisense Oligonucleotide Conjugated to ?-Tocopherol, Mol. Ther. Nucleic Acids, vol.4, issue.1, 2015.

J. O. Mcnamara, E. R. Andrechek, Y. Wang, K. D. Viles, R. E. Rempel et al., Cell Type-Specific Delivery of SiRNAs with Aptamer-SiRNA Chimeras, Nat. Biotechnol, vol.24, issue.8, pp.1005-1015, 2006.

C. P. Neff, J. Zhou, L. Remling, J. Kuruvilla, J. Zhang et al., An Aptamer-SiRNA Chimera Suppresses HIV-1 Viral Loads and Protects from Helper CD4+ T Cell Decline in Humanized Mice, Sci. Transl. Med, vol.3, issue.66, 2011.

J. Zhou and J. Rossi, Cell-Type Specific Aptamer and Aptamer-SiRNA Conjugates for Targeted HIV-1 Therapy, J. Investig. Med. Off. Publ. Am. Fed. Clin. Res, vol.62, issue.7, pp.914-919, 2014.

,

J. Zhou, H. Li, S. Li, J. Zaia, and J. J. Rossi, Novel Dual Inhibitory Function Aptamer-SiRNA Delivery System for HIV-1 Therapy, Mol. Ther. J. Am. Soc. Gene Ther, vol.16, issue.8, pp.1481-1489, 2008.

S. Kruspe and P. H. Giangrande, Aptamer-SiRNA Chimeras: Discovery, Progress, and Future Prospects, Biomedicines, vol.2017, issue.3, p.45

S. Ni, H. Yao, L. Wang, J. Lu, F. Jiang et al., Chemical Modifications of Nucleic Acid Aptamers for Therapeutic Purposes, Int. J. Mol. Sci, vol.2017, issue.8, p.1683

E. Song, P. Zhu, S. Lee, D. Chowdhury, S. Kussman et al., Antibody Mediated in Vivo Delivery of Small Interfering RNAs via Cell-Surface Receptors, Nat. Biotechnol, vol.23, issue.6, pp.709-717, 2005.

N. Satake, C. Duong, S. Yoshida, M. Oestergaard, C. Chen et al., Novel Targeted Therapy for Precursor B-Cell Acute Lymphoblastic Leukemia: Anti-CD22 Antibody-MXD3 Antisense Oligonucleotide Conjugate

. Med, , vol.22, pp.632-642, 2016.

T. L. Cuellar, D. Barnes, C. Nelson, J. Tanguay, S. Yu et al., Systematic Evaluation of Antibody-Mediated SiRNA Delivery Using an Industrial Platform of THIOMAB-SiRNA Conjugates, Nucleic Acids Res, vol.43, issue.2, pp.1189-1203, 2015.

G. Godeau, C. Staedel, and P. Barthélémy, Lipid-Conjugated Oligonucleotides via "Click Chemistry, J. Med. Chem, issue.15, pp.4374-4376, 2008.

N. S. Petrova, I. V. Chernikov, M. I. Meschaninova, . Dovydenko, .. S. Ii et al., Carrier-Free Cellular Uptake and the Gene-Silencing Activity of the Lipophilic SiRNAs Is Strongly Affected by the Length of the Linker between SiRNA and Lipophilic Group, Nucleic Acids Res, vol.40, issue.5, pp.2330-2344, 2012.

I. V. Chernikov, D. V. Gladkikh, M. I. Meschaninova, A. G. Ven'yaminova, M. A. Zenkova et al., Cholesterol-Containing Nuclease-Resistant SiRNA Accumulates in Tumors in a Carrier-Free Mode and Silences MDR1 Gene, Mol. Ther. Nucleic Acids, vol.6, pp.209-220, 2017.

,

J. Yang, C. Chen, and X. Tang, Cholesterol-Modified Caged SiRNAs for Photoregulating Exogenous and Endogenous Gene Expression. Bioconjug. Chem, 2018.

J. Soutschek, A. Akinc, B. Bramlage, K. Charisse, R. Constien et al., Therapeutic Silencing of an Endogenous Gene by Systemic Administration of Modified SiRNAs, Nature, vol.432, issue.7014, pp.173-178, 2004.

M. Difiglia, M. Sena-esteves, K. Chase, E. Sapp, E. Pfister et al., Therapeutic Silencing of Mutant Huntingtin with SiRNA Attenuates Striatal and Cortical Neuropathology and Behavioral Deficits, Proc. Natl. Acad. Sci. U. S. A, vol.104, issue.43, pp.17204-17209, 2007.

J. Yang, C. Chen, and X. Tang, Cholesterol-Modified Caged SiRNAs for Photoregulating Exogenous and Endogenous Gene Expression. Bioconjug. Chem, 2018.

M. Nakajima, T. Kasuya, S. Yokota, R. Onishi, T. Ikehara et al., Gene Silencing Activity and Hepatic Accumulation of Antisense Oligonucleotides Bearing Cholesterol-Conjugated Thiono Triester at the Gap Region. Nucleic Acid Ther, vol.27, pp.232-237, 2017.

S. A. Moschos, S. W. Jones, M. M. Perry, A. E. Williams, J. S. Erjefalt et al., Lung Delivery Studies Using SiRNA Conjugated to TAT(48?60) and Penetratin Reveal Peptide Induced Reduction in Gene Expression and Induction of Innate Immunity, Bioconjug. Chem, vol.18, issue.5, pp.1450-1459, 2007.

M. Raouane, D. Desmaële, G. Urbinati, L. Massaad-massade, and P. Couvreur, Lipid Conjugated Oligonucleotides: A Useful Strategy for Delivery, Bioconjug. Chem, vol.2012, issue.6, pp.1091-1104
URL : https://hal.archives-ouvertes.fr/hal-02330037

M. Raouane, D. Desmaele, M. Gilbert-sirieix, C. Gueutin, F. Zouhiri et al., Synthesis, Characterization, and in Vivo Delivery of SiRNA-Squalene Nanoparticles Targeting Fusion Oncogene in Papillary Thyroid Carcinoma, J. Med. Chem, vol.54, issue.12, pp.4067-4076, 2011.

B. Herbert, G. C. Gellert, A. Hochreiter, K. Pongracz, W. E. Wright et al., Lipid Modification of GRN163, an N3? ? P5? Thio-Phosphoramidate Oligonucleotide, Enhances the Potency of Telomerase Inhibition, Oncogene, vol.24, issue.33, pp.5262-5268, 2005.

,

K. M. Burchett, Y. Yan, and M. M. Ouellette, Telomerase Inhibitor Imetelstat (GRN163L) Limits the Lifespan of Human Pancreatic Cancer Cells, PLoS ONE, vol.2014, issue.1

E. M. Goldblatt, P. A. Erickson, E. R. Gentry, S. M. Gryaznov, and B. Herbert, Lipid-Conjugated Telomerase Template Antagonists Sensitize Resistant HER2-Positive Breast Cancer Cells to Trastuzumab, Breast Cancer Res. Treat, vol.118, issue.1, pp.21-32, 2009.

A. Gissot, C. D. Primo, I. Bestel, G. Giannone, H. Chapuis et al., Sensitive Liposomes Encoded with Oligonucleotide Amphiphiles: A Biocompatible Switch, Chem. Commun, issue.43, pp.5550-5552, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02484276

O. Pokholenko, A. Gissot, B. Vialet, K. Bathany, A. Thiéry et al., Lipid Oligonucleotide Conjugates as Responsive Nanomaterials for Drug Delivery, J. Mater. Chem. B, vol.2013, issue.39, p.5329
URL : https://hal.archives-ouvertes.fr/hal-02484550

S. Karaki, S. Benizri, R. Mejías, V. Baylot, N. Branger et al., Lipid-Oligonucleotide Conjugates Improve Cellular Uptake and Efficiency of TCTP-Antisense in Castration-Resistant Prostate Cancer, J. Controlled Re, vol.258, pp.1-9, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01520102

J. Acunzo, V. Baylot, A. So, and P. Rocchi, TCTP as Therapeutic Target in Cancers, Cancer Treat. Rev, vol.40, issue.6, pp.760-769, 2014.

V. Baylot, M. Katsogiannou, C. Andrieu, D. Taieb, J. Acunzo et al., Targeting TCTP as a New Therapeutic Strategy in Castration-Resistant Prostate Cancer, Mol. Ther, vol.2012, issue.12, pp.2244-2256

,

M. Kirby, C. Hirst, and E. D. Crawford, Characterising the Castration-Resistant Prostate Cancer Population: A Systematic Review, Int. J. Clin. Pract, vol.65, issue.11, pp.1180-1192, 2011.

B. Tajik-ahmadabad, A. Polyzos, F. Separovic, and F. Shabanpoor, Amphiphilic Lipopeptide Significantly Enhances Uptake of Charge-Neutral Splice Switching Morpholino Oligonucleotide in Spinal Muscular Atrophy Patient-Derived Fibroblasts, Int. J. Pharm, vol.2017, issue.1, pp.21-28

F. Wada, T. Yamamoto, T. Ueda, M. Sawamura, S. Wada et al., Cholesterol-GalNAc Dual Conjugation Strategy for Reducing Renal Distribution of Antisense Oligonucleotides, Nucleic Acid Ther, vol.28, issue.1, pp.50-57, 2018.

,

T. Yamamoto, M. Sawamura, F. Wada, M. Harada-shiba, and S. Obika, Serial Incorporation of a Monovalent GalNAc Phosphoramidite Unit into Hepatocyte-Targeting Antisense Oligonucleotides, Bioorg. Med. Chem, vol.24, issue.1, pp.26-32, 2016.

J. I. Cutler, E. Auyeung, and C. A. Mirkin, Spherical Nucleic Acids. J. Am. Chem. Soc, vol.2012, issue.3, pp.1376-1391

,

K. Zhang, L. Hao, S. J. Hurst, and C. A. Mirkin, Antibody-Linked Spherical Nucleic Acids for Cellular Targeting, J. Am. Chem. Soc, vol.2012, issue.40, pp.16488-16491

A. F. Radovic-moreno, N. Chernyak, C. C. Mader, S. Nallagatla, R. S. Kang et al., Immunomodulatory Spherical Nucleic Acids. Proc. Natl. Acad. Sci, vol.2015, issue.13, pp.3892-3897

,

K. Skakuj, S. Wang, L. Qin, A. Lee, B. Zhang et al., Conjugation Chemistry-Dependent T-Cell Activation with Spherical Nucleic Acids, J. Am. Chem. Soc, vol.140, issue.4, pp.1227-1230, 2018.

S. N. Barnaby, G. A. Perelman, K. L. Kohlstedt, A. B. Chinen, G. C. Schatz et al., Design Considerations for RNA Spherical Nucleic Acids (SNAs), Bioconjug. Chem, vol.27, issue.9, pp.2124-2131, 2016.

D. S. Seferos, D. A. Giljohann, N. L. Rosi, and C. A. Mirkin, Locked Nucleic Acid-Nanoparticle Conjugates, ChemBioChem, vol.8, issue.11, pp.1230-1232, 2007.

C. H. Choi, L. Hao, S. P. Narayan, E. Auyeung, and C. A. Mirkin, Mechanism for the Endocytosis of Spherical Nucleic Acid Nanoparticle Conjugates, Proc. Natl. Acad. Sci, vol.110, pp.7625-7630, 2013.

R. J. Banga, N. Chernyak, S. P. Narayan, S. T. Nguyen, and C. A. Mirkin, Liposomal Spherical Nucleic Acids, J. Am. Chem. Soc, vol.136, issue.28, pp.9866-9869, 2014.

D. Zheng, D. A. Giljohann, D. L. Chen, M. D. Massich, X. Wang et al., Topical Delivery of SiRNA-Based Spherical Nucleic Acid Nanoparticle Conjugates for Gene Regulation, Proc. Natl. Acad. Sci, vol.109, pp.11975-11980, 2012.

K. T. Lewandowski, R. Thiede, N. Guido, W. L. Daniel, R. Kang et al., Topically Delivered Tumor Necrosis Factor-?-Targeted Gene Regulation for Psoriasis, J. Invest. Dermatol, vol.137, issue.9, pp.2027-2030, 2017.

P. Piascik, Fomiversen Sodium Approved to Treat CMV Retinitis, J. Am. Pharm. Assoc. WashingtonDC, vol.39, issue.1, pp.84-85, 19961999.

I. Kozak, J. A. Mccutchan, and W. R. Freeman, Chapter 81 -HIV-Associated Infections, Retina

S. J. Ryan, S. R. Sadda, D. R. Hinton, A. P. Schachat, S. R. Sadda et al., , pp.1441-1472, 2013.

C. F. Bennett and E. E. Swayze, RNA Targeting Therapeutics: Molecular Mechanisms of Antisense Oligonucleotides as a Therapeutic Platform, Annu. Rev. Pharmacol. Toxicol, vol.50, issue.1, pp.259-293, 2010.

K. M. Schoch and T. M. Miller, Antisense Oligonucleotides: Translation from Mouse Models to Human Neurodegenerative Diseases, Neuron, vol.2017, issue.6, pp.1056-1070

M. M. Evers, L. J. Toonen, and W. M. Van-roon-mom, Antisense Oligonucleotides in Therapy for Neurodegenerative Disorders, Adv. Drug Deliv. Rev, vol.87, pp.90-103, 2015.

V. L. Herrera, A. H. Colby, N. Ruiz-opazo, D. G. Coleman, and M. W. Grinstaff, Nucleic Acid Nanomedicines in Phase II/III Clinical Trials: Translation of Nucleic Acid Therapies for Reprogramming Cells. Nanomed, 2018.

R. L. Juliano, The Delivery of Therapeutic Oligonucleotides, Nucleic Acids Res, vol.44, issue.14, pp.6518-6548, 2016.

,

S. T. Crooke, J. L. Witztum, C. F. Bennett, and B. F. Baker, RNA-Targeted Therapeutics, Cell Metab, vol.27, issue.4, pp.714-739, 2018.

G. Corporation, Geron Announces Fast Track Designation Granted to Imetelstat for Lower Risk Myelodysplastic Syndromes, 2018.

N. Machin and M. V. Ragni, An Investigational RNAi Therapeutic Targeting Antithrombin for the Treatment of Hemophilia A and B, J. Blood Med, vol.9, pp.135-140, 2018.

,

S. L. Fine, D. F. Martin, P. P. Kirkpatrick, and . Sodium, Nat. Rev. Drug Discov, vol.4, issue.3, pp.187-188, 2005.

,

E. W. Ng, D. T. Shima, P. Calias, E. T. Cunningham, D. R. Guyer et al., Aptamer for Ocular Vascular Disease, Nat. Rev. Drug Discov, vol.5, issue.2, pp.123-132, 2006.

A. O. Garba and S. A. Mousa, Bevasiranib for the Treatment of Wet, Age-Related Macular Degeneration. Ophthalmol. Eye Dis, vol.2, pp.75-83, 2010.

X. Shen, D. R. Corey, and . Chemistry, Mechanism and Clinical Status of Antisense Oligonucleotides and Duplex RNAs, Nucleic Acids Res, vol.46, issue.4, pp.1584-1600, 2018.

C. J. Niemietz, V. Sauer, J. Stella, L. Fleischhauer, G. Chandhok et al., Evaluation of Therapeutic Oligonucleotides for Familial Amyloid Polyneuropathy in Patient-Derived Hepatocyte-Like Cells, PLoS ONE, vol.2016, issue.9, p.11

E. J. Ackermann, S. Guo, M. D. Benson, S. Booten, S. Freier et al., Suppressing Transthyretin Production in Mice, Monkeys and Humans Using 2nd-Generation Antisense Oligonucleotides, Amyloid Int. J. Exp. Clin. Investig. Off. J. Int. Soc. Amyloidosis, vol.23, issue.3, pp.148-157, 2016.

M. D. Benson, N. R. Dasgupta, S. M. Rissing, J. Smith, and H. Feigenbaum, Safety and Efficacy of a TTR Specific Antisense Oligonucleotide in Patients with Transthyretin Amyloid Cardiomyopathy, Amyloid, vol.2017, issue.4, pp.217-223

,

M. Rizk and ?. Tüzmen, Update on the Clinical Utility of an RNA Interference-Based Treatment: Focus on Patisiran. Pharmacogenomics Pers, vol.10, pp.267-278, 2017.

K. E. Lundin, O. Gissberg, and C. I. Smith, Oligonucleotide Therapies: The Past and the Present, Hum. Gene Ther, vol.26, issue.8, pp.475-485, 2015.

H. L. Janssen, H. W. Reesink, E. J. Lawitz, S. Zeuzem, M. Rodriguez-torres et al., Treatment of HCV Infection by Targeting Mi-croRNA, N. Engl. J. Med, vol.368, issue.18, pp.1685-1694, 2013.

M. H. Van-der-ree, A. J. Van-der-meer, J. De-bruijne, R. Maan, A. Van-vliet et al., Long-Term Safety and Efficacy of MicroRNA-Targeted Therapy in Chronic Hepatitis C Patients, Antiviral Res, vol.111, pp.53-59, 2014.

M. S. Beg, A. J. Brenner, J. Sachdev, M. Borad, Y. Kang et al., Phase I Study of MRX34, a Liposomal MiR-34a Mimic, Administered Twice Weekly in Patients with Advanced Solid Tumors, Invest. New Drugs, vol.35, issue.2, pp.180-188, 2017.

I. Zlatev, A. Castoreno, C. R. Brown, J. Qin, S. Waldron et al., Reversal of SiRNA-Mediated Gene Silencing in Vivo, Nat. Biotechnol, vol.36, issue.6, pp.509-511, 2018.

I. Zlatev, A. Castoreno, C. R. Brown, J. Qin, S. Waldron et al., Reversal of SiRNA-Mediated Gene Silencing in Vivo, Nat. Biotechnol, vol.36, issue.6, pp.509-511, 2018.

I. Ekladious,

Y. L. Colson and M. W. Grinstaff, Polymer-drug Conjugates for Cancer Treatment: Realities, Insights, and Prospects Nat. Rev. Drug Disc, 2019.

M. W. Grinstaff, H. M. Kaplan, and J. Kohn, Predoctoral and Postdoctoral Training Pipeline in Translational Biomaterials Research and Regenerative Medicine, ACS Biomater. Sci. Eng, 2017.