J. Ferlay, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012, Int J Cancer, vol.136, pp.359-386, 2015.

P. Saraon, A. P. Drabovich, K. A. Jarvi, and E. P. Diamandis, Mechanisms of Androgen-Independent Prostate Cancer, EJIFCC, vol.25, pp.42-54, 2014.

T. Chandrasekar, J. C. Yang, A. C. Gao, and C. P. Evans, Mechanisms of resistance in castration-resistant prostate cancer (CRPC), Transl Androl Urol, vol.4, pp.365-380, 2015.

N. Nussbaum, Patient experience in the treatment of metastatic castration-resistant prostate cancer: state of the science, Prostate Cancer Prostatic Dis, vol.19, pp.111-121, 2016.

H. I. Scher, Increased survival with enzalutamide in prostate cancer after chemotherapy, N Engl J Med, vol.367, pp.1187-1197, 2012.

P. W. Kantoff, Sipuleucel-T immunotherapy for castration-resistant prostate cancer, N Engl J Med, vol.363, pp.411-422, 2010.

C. Parker, Alpha emitter radium-223 and survival in metastatic prostate cancer, N Engl J Med, vol.369, pp.213-223, 2013.

C. Tomasetti, L. Li, and B. Vogelstein, Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention, Science, vol.355, pp.1330-1334, 2017.

A. D. Keefe, S. Pai, and A. Ellington, Aptamers as therapeutics, Nat Rev Drug Discov, vol.9, pp.537-550, 2010.

E. Magbanua and U. Hahn, Chemical Biology of Nucleic Acids. RNA Technologies, 2014.

G. Mayer, The Chemical Biology of Aptamers, Angewandte Chemie International Edition, vol.48, pp.2672-2689, 2009.

M. Alibolandi, Smart AS1411-aptamer conjugated pegylated PAMAM dendrimer for the superior delivery of camptothecin to colon adenocarcinoma in vitro and in vivo, Int J Pharm, vol.519, pp.352-364, 2017.

G. A. Clawson, A Cholecystokinin B Receptor-Specific DNA Aptamer for Targeting Pancreatic Ductal Adenocarcinoma, Nucleic Acid Ther, vol.27, pp.23-35, 2017.

S. Yoon, Targeted Delivery of C/EBPalpha -saRNA by Pancreatic Ductal Adenocarcinoma-specific RNA Aptamers Inhibits Tumor Growth In Vivo, Mol Ther, vol.24, pp.1106-1116, 2016.

J. P. Dassie, Targeted inhibition of prostate cancer metastases with an RNA aptamer to prostate-specific membrane antigen, Mol Ther, vol.22, pp.1910-1922, 2014.

W. H. Thiel, Smooth Muscle Cell-targeted RNA Aptamer Inhibits Neointimal Formation, Mol Ther, vol.24, pp.779-787, 2016.

A. Cibiel, From ugly duckling to swan: unexpected identification from cell-SELEX of an anti-Annexin A2 aptamer targeting tumors, PLoS One, vol.9, p.87002, 2014.

G. Zhu, Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics, Proc Natl Acad Sci, vol.110, pp.7998-8003, 2013.

P. Gray and B. , Tunable cytotoxic aptamer-drug conjugates for the treatment of prostate cancer, Proceedings of the National Academy of Sciences, vol.115, pp.4761-4766, 2018.

M. Avci-adali, H. Steinle, T. Michel, C. Schlensak, and H. P. Wendel, Potential capacity of aptamers to trigger immune activation in human blood, PLoS One, vol.8, p.68810, 2013.

M. Kimoto, R. Yamashige, K. Matsunaga, S. Yokoyama, and I. Hirao, Generation of high-affinity DNA aptamers using an expanded genetic alphabet, Nature Biotechnology, vol.31, p.453, 2013.

A. Taylor, S. Arangundy-franklin, and P. Holliger, Towards applications of synthetic genetic polymers in diagnosis and therapy, vol.22, 2014.

L. Zhang, Aptamers against Cells Overexpressing Glypican 3 from Expanded Genetic Systems Combined with Cell Engineering and Laboratory Evolution, Angew Chem Int Ed Engl, vol.55, pp.12372-12375, 2016.

F. Pfeiffer, Identification and characterization of nucleobase-modified aptamers by click-SELEX, Nature Protocols, vol.13, p.1153, 2018.

D. Cunningham and Z. You, In vitro and in vivo model systems used in prostate cancer research, J Biol Methods, vol.2, 2015.

K. Fizazi, High efficacy of docetaxel with and without androgen deprivation and estramustine in preclinical models of advanced prostate cancer, Anticancer Res, vol.24, pp.2897-2903, 2004.

J. M. Healy, Pharmacokinetics and biodistribution of novel aptamer compositions, Pharm Res, vol.21, pp.2234-2246, 2004.

D. W. Drolet, L. S. Green, L. Gold, and N. Janjic, Fit for the Eye: Aptamers in Ocular Disorders, Nucleic Acid Ther, vol.26, pp.127-146, 2016.

M. Blank, Next-Generation Analysis of Deep Sequencing Data: Bringing Light into the Black Box of SELEX Experiments, Methods Mol Biol, vol.1380, pp.85-95, 2016.

J. Zhou and J. Rossi, Aptamers as targeted therapeutics: current potential and challenges, Nat Rev Drug Discov, vol.16, pp.181-202, 2017.

S. Burge, G. N. Parkinson, P. Hazel, A. K. Todd, S. Neidle et al., DNA: sequence, topology and structure, Nucleic Acids Res, vol.34, pp.5402-5415, 2006.

D. Sen and W. Gilbert, A sodium-potassium switch in the formation of four-stranded G4-DNA, Nature, vol.344, p.410, 1990.

J. Kypr, I. Kejnovska, D. Renciuk, and M. Vorlickova, Circular dichroism and conformational polymorphism of DNA, Nucleic Acids Res, vol.37, pp.1713-1725, 2009.

F. Opazo, Modular Assembly of Cell-targeting Devices Based on an Uncommon G-quadruplex Aptamer, Mol Ther Nucleic Acids, vol.4, p.251, 2015.

K. P. Leister, Two High Throughput Screen Assays for Measurement of TNF-alpha in THP-1 Cells, Curr Chem Genomics, vol.5, pp.21-29, 2011.

S. Bauer, S. Pigisch, D. Hangel, A. Kaufmann, and S. Hamm, Recognition of nucleic acid and nucleic acid analogs by Toll-like receptors 7, 8 and 9, Immunobiology, vol.213, pp.315-328, 2008.

J. Mi, In vivo selection of tumor-targeting RNA motifs, Nature Chemical Biology, vol.6, pp.22-24, 2010.

C. Cheng, Y. H. Chen, K. A. Lennox, M. A. Behlke, and B. L. Davidson, In vivo SELEX for Identification of Brain-penetrating Aptamers, Molecular Therapy -Nucleic Acids, vol.2, p.67, 2013.

J. Mi, In Vivo Selection Against Human Colorectal Cancer Xenografts Identifies an Aptamer That Targets RNA Helicase Protein DHX9, Molecular Therapy -Nucleic Acids, vol.5, p.315, 2016.

H. Liu, A Novel DNA Aptamer for Dual Targeting of Polymorphonuclear Myeloid-derived Suppressor Cells and Tumor Cells, Theranostics, vol.8, pp.31-44, 2018.

E. W. Ng, Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease, Nat Rev Drug Discov, vol.5, pp.123-132, 2006.

J. P. Vavalle and M. G. Cohen, The REG1 anticoagulation system: a novel actively controlled factor IX inhibitor using RNA aptamer technology for treatment of acute coronary syndrome, Future Cardiol, vol.8, pp.371-382, 2012.

J. O. Mcnamara, Multivalent 4-1BB binding aptamers costimulate CD8+T cells and inhibit tumor growth in mice, J Clin Invest, vol.118, pp.376-386, 2008.

F. Mongelard and P. Bouvet, AS-1411, a guanosine-rich oligonucleotide aptamer targeting nucleolin for the potential treatment of cancer, including acute myeloid leukemia, Curr Opin Mol Ther, vol.12, pp.107-114, 2010.

L. C. Bock, L. C. Griffin, J. A. Latham, E. H. Vermaas, and J. J. Toole, Selection of single-stranded DNA molecules that bind and inhibit human thrombin, Nature, vol.355, pp.564-566, 1992.

A. Gomez-outes, New parenteral anticoagulants in development, Ther Adv. Cardiovasc Dis, vol.5, pp.33-59, 2011.

M. Takahashi, High throughput sequencing analysis of RNA libraries reveals the influences of initial library and PCR methods on SELEX efficiency, Sci Rep, vol.6, p.33697, 2016.

J. M. Harris and R. B. Chess, Effect of pegylation on pharmaceuticals, Nat Rev Drug Discov, vol.2, pp.214-221, 2003.

D. Gachoka, Polyethylene Glycol (PEG)-Induced Anaphylactic Reaction During Bowel Preparation, ACG Case Rep J, vol.2, pp.216-217, 2015.
DOI : 10.14309/crj.2015.63

URL : https://doi.org/10.14309/crj.2015.63

E. C. Wenande, P. S. Skov, H. Mosbech, L. K. Poulsen, and L. H. Garvey, Inhibition of polyethylene glycol-induced histamine release by monomeric ethylene and diethylene glycol: a case of probable polyethylene glycol allergy, J Allergy Clin Immunol, vol.131, pp.1425-1427, 2013.

A. Girones, M. Roan-roan, J. De-la-hoz, B. Sanchez-cano, and M. , Immediate allergic reactions by polyethylene glycol 4000: two cases, Allergol Immunopathol (Madr), vol.36, pp.110-112, 2008.

B. Yang, S. I. Lim, J. C. Kim, G. Tae, and I. Kwon, Site-Specific Albumination as an Alternative to PEGylation for the Enhanced Serum Half-Life in Vivo, Biomacromolecules, vol.17, pp.1811-1817, 2016.

Y. Qi and A. Chilkoti, Protein-polymer conjugation-moving beyond PEGylation, Curr Opin Chem Biol, vol.28, pp.181-193, 2015.
DOI : 10.1016/j.cbpa.2015.08.009

URL : http://europepmc.org/articles/pmc4624571?pdf=render

A. Berezhnoy, Isolation and optimization of murine IL-10 receptor blocking oligonucleotide aptamers using high-throughput sequencing, Mol Ther, vol.20, pp.1242-1250, 2012.

T. Schutze, Probing the SELEX process with next-generation sequencing, PLoS One, vol.6, p.29604, 2011.

V. Domenyuk, Plasma Exosome Profiling of Cancer Patients by a Next Generation Systems Biology Approach, Sci Rep, vol.7, p.42741, 2017.

. Domenyuk, Poly-ligand profiling differentiates trastuzumab-treated breast cancer patients according to their outcomes, Nature Communicationsvolume, vol.9, p.1219, 2018.

N. Nguyen-quang, G. Perret, and F. Duconge, Applications of High-Throughput Sequencing for In Vitro Selection and Characterization of Aptamers, Pharmaceuticals (Basel), p.9, 2016.

K. K. Alam, J. L. Chang, and D. H. Burke, FASTAptamer: A Bioinformatic Toolkit for High-throughput Sequence Analysis of Combinatorial Selections, Mol Ther Nucleic Acids, vol.4, p.230, 2015.

P. J. Thul, A subcellular map of the human proteome, 2017.

R. E. Martell, J. R. Nevins, and B. A. Sullenger, Optimizing aptamer activity for gene therapy applications using expression cassette SELEX, Mol Ther, vol.6, pp.30-34, 2002.

J. P. Dassie, Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors, Nat Biotechnol, vol.27, pp.839-849, 2009.

J. L. Diener, Inhibition of von Willebrand factor-mediated platelet activation and thrombosis by the anti-von Willebrand factor A1-domain aptamer ARC1779, J Thromb Haemost, vol.7, pp.1155-1162, 2009.

C. Da-pieve, P. Williams, D. M. Haddleton, R. M. Palmer, and S. Missailidis, Modification of thiol functionalized aptamers by conjugation of synthetic polymers, Bioconjug Chem, vol.21, pp.169-174, 2010.

S. E. Lupold, B. J. Hicke, Y. Lin, and D. S. Coffey, Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigen, Cancer Res, vol.62, pp.4029-4033, 2002.

H. Y. Liu, X. Yu, H. Liu, D. Wu, and J. X. She, Co-targeting EGFR and survivin with a bivalent aptamer-dual siRNA chimera effectively suppresses prostate cancer, Sci Rep, vol.6, p.30346, 2016.

J. O. Mcnamara, Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras, Nat Biotechnol, vol.24, pp.1005-1015, 2006.

F. Pastor, D. Kolonias, P. H. Giangrande, and E. Gilboa, Induction of tumour immunity by targeted inhibition of nonsense-mediated mRNA decay, Nature, vol.465, pp.227-230, 2010.

O. C. Farokhzad, Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo, Proc Natl Acad Sci, vol.103, pp.6315-6320, 2006.

W. Qiu and G. H. Su, Development of orthotopic pancreatic tumor mouse models, Methods Mol Biol, vol.980, pp.215-223, 2013.

M. Saxena and G. Christofori, Rebuilding cancer metastasis in the mouse, Mol Oncol, vol.7, pp.283-296, 2013.

R. E. Mcintyre, S. J. Buczacki, M. J. Arends, and D. J. Adams, Mouse models of colorectal cancer as preclinical models, Bioessays, vol.37, pp.909-920, 2015.

P. Mordant, Bioluminescent orthotopic mouse models of human localized non-small cell lung cancer: feasibility and identification of circulating tumour cells, PLoS One, vol.6, p.26073, 2011.

S. Gupta, Chemically modified DNA aptamers bind interleukin-6 with high affinity and inhibit signaling by blocking its interaction with interleukin-6 receptor, J Biol Chem, vol.289, pp.8706-8719, 2014.

M. Hirota, Chemically Modified Interleukin-6 Aptamer Inhibits Development of Collagen-Induced Arthritis in Cynomolgus Monkeys, Nucleic Acid Ther, vol.26, pp.10-19, 2016.

N. Mor-vaknin, DEK-targeting DNA aptamers as therapeutics for inflammatory arthritis, Nat Commun, vol.8, p.14252, 2017.

F. Tolle and G. Mayer, Nucleic Acid Aptamers: Selection, Characterization, and Application, pp.77-84, 2016.

I. Theodorou, In Vitro and In Vivo Imaging of Fluorescent Aptamers, Methods Mol Biol, vol.1380, pp.135-150, 2016.

, Andreas Lingnau, vol.10, issue.2

, Chemical Biology and Chemical Genetics

. Str, , vol.1, p.53121

D. Cea, Molecular Imaging Research Center (MIRCen), vol.18, p.92260

. Fontenay-aux-roses, France 3 KTB Tumorforschungsgesellschaft mbH, Research Division ProQinase, Breisacher Str, vol.117, p.79106

. Freiburg,

A. Gmbh, Germany 7 Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Sigmund-Freud-Str. 27; 53127 Bonn, Germany DNA libraries from selection cycles C1 to C10 of the in vivo selection using D3P (c), Life and Medical Sciences, vol.31