M. Dobbelstein and C. S. Sørensen, Exploiting replicative stress to treat cancer, Nat. Rev. Drug Discov, vol.14, pp.405-423, 2015.

A. Ciccia and S. J. Elledge, The DNA damage response: making it safe to play with knives, Mol. Cell, vol.40, pp.179-204, 2010.

S. P. Jackson and J. Bartek, The DNA-damage response in human biology and disease, Nature, vol.461, pp.1071-1078, 2009.

C. J. Lord and A. Ashworth, The DNA damage response and cancer therapy, Nature, vol.481, pp.287-294, 2012.

M. J. O'connor, Targeting the DNA damage response in cancer, Mol. Cell, vol.60, pp.547-560, 2015.

R. Rodriguez and K. M. Miller, Unravelling the genomic targets of small molecules using high-throughput sequencing, Nat. Rev. Genet, vol.15, pp.783-796, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01110554

A. N. Blackford, S. P. Jackson, A. Atm, and D. , the trinity at the heart of the DNA damage response, Mol. Cell, vol.66, pp.801-817, 2017.

A. Bacolla, D. N. Cooper, and K. M. Vasquez, DNA structure matters, Genome Med, vol.5, p.51, 2013.

E. V. Mirkin and S. M. Mirkin, Replication fork stalling at natural impediments. Microbiol, Mol. Biol. Rev, vol.71, pp.13-35, 2007.

G. Wang and K. M. Vasquez, Non-B DNA structure-induced genetic instability, Mut. Res, vol.598, pp.103-119, 2006.

M. Tarsounas and M. Tijsterman, Genomes and G-Quadruplexes: for better or for worse, J. Mol. Biol, vol.425, pp.4782-4789, 2013.

D. Rhodes and H. J. Lipps, G-quadruplexes and their regulatory roles in biology, Nucleic Acids Res, vol.43, pp.8627-8637, 2015.

N. Maizels and L. T. Gray, The G4 genome, PLoS Genet, vol.9, p.1003468, 2013.

B. Lemmens, R. Van-schendel, and M. Tijsterman, Mutagenic consequences of a single G-quadruplex demonstrate mitotic inheritance of DNA replication fork barriers, Nat. Commun, vol.6, p.8909, 2015.

J. Zimmer, E. M. Tacconi, C. Folio, S. Badie, M. Porru et al., ) Targeting BRCA1 and BRCA2 deficiencies with G-quadruplex-interacting compounds, Mol. Cell, vol.61, pp.449-460, 2016.

A. Valton and M. Prioleau, G-quadruplexes in DNA replication: a problem or a necessity?, Trends Genet, vol.32, pp.697-706, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01472913

S. Balasubramanian, L. H. Hurley, and S. Neidle, Targeting G-quadruplexes in gene promoters: a novel anticancer strategy?, Nat. Rev. Drug Discov, vol.10, pp.261-275, 2011.

A. Bugaut and S. Balasubramanian, 5 -UTR RNA G-quadruplexes: translation regulation and targeting, Nucleic Acids Res, vol.40, pp.4727-4741, 2012.

N. Maizels, G4-associated human diseases, EMBO Rep, 2015.

S. Mueller and R. Rodriguez, G-quadruplex interacting small molecules and drugs: from bench toward bedside, Exp. Rev. Clin. Pharmacol, vol.7, pp.663-679, 2014.

S. Neidle, Quadruplex nucleic acids as novel therapeutic targets, J. Med. Chem, vol.59, pp.5987-6011, 2016.

P. Murat, Y. Singh, and E. Defrancq, Methods for investigating G-quadruplex DNA/ligand interactions, Chem. Soc. Rev, vol.40, pp.5293-5307, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01655279

J. Jaumot and R. Gargallo, Experimental methods for studying the interactions between G-quadruplex structures and ligands, Curr. Pharm. Des, vol.18, pp.1900-1916, 2012.

J. M. Nicoludis, S. P. Barrett, J. Mergny, and L. A. Yatsunyk, Interaction of human telomeric DNA with N-methyl mesoporphyrin IX, Nucleic Acids Res, vol.40, pp.5432-5447, 2012.

B. Pagano, C. A. Mattia, and C. Giancola, Applications of isothermal titration calorimetry in biophysical studies of G-quadruplexes, Int. J. Mol. Sci, vol.10, pp.2935-2957, 2009.

I. Haq, B. Z. Chowdhry, and T. C. Jenkins, Calorimetric techniques in the study of high-order DNA-drug interactions, Meth. Enzymol, vol.340, pp.109-149, 2001.

F. Rosu, E. De-pauw, and V. Gabelica, Electrospray mass spectrometry to study drug-nucleic acids interactions, Biochimie, vol.90, pp.1074-1087, 2008.

S. M. Haider, S. Neidle, and G. N. Parkinson, A structural analysis of G-quadruplex/ligand interactions, Biochimie, vol.93, pp.1239-1251, 2011.

D. J. Patel, A. T. Phan, and V. Kuryavyi, Human telomere, oncogenic promoter and 5 -UTR G-quadruplexes: diverse higher order DNA and RNA targets for cancer therapeutics, Nucleic Acids Res, vol.35, pp.7429-7455, 2007.

D. Yang and K. Okamoto, Structural insights into G-quadruplexes: towards new anticancer drugs, Future Med. Chem, vol.2, pp.619-646, 2010.

E. W. White, F. Tanious, M. A. Ismail, A. P. Reszka, S. Neidle et al., Structure-specific recognition of quadruplex DNA by organic cations: Influence of shape, substituents and charge, Biophys. Chem, vol.126, pp.140-153, 2007.

J. S. Ren and J. B. Chaires, Sequence and structural selectivity of nucleic acid binding ligands, Biochemistry, vol.38, pp.16067-16075, 1999.

P. A. Ragazzon, N. C. Garbett, and J. B. Chaires, Competition dialysis: a method for the study of structural selective nucleic acid binding, Methods, vol.42, pp.173-182, 2007.

A. De-cian, L. Guittat, M. Kaiser, B. Sacca, S. Amrane et al., Fluorescence-based melting assays for studying quadruplex ligands, Methods, vol.42, pp.183-195, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00258193

P. A. Rachwal and K. R. Fox, Quadruplex melting, Methods, vol.43, pp.291-301, 2007.

D. Monchaud, C. Allain, H. Bertrand, N. Smargiasso, F. Rosu et al., Ligands playing musical chairs with G-quadruplex DNA: a rapid and simple displacement assay for identifying selective G-quadruplex binders, Biochimie, vol.90, pp.1207-1223, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00311781

P. L. Tran, E. Largy, F. Hamon, M. Teulade-fichou, and J. Mergny, Fluorescence intercalator displacement assay for screening G4 ligands towards a variety of G-quadruplex structures, Biochimie, vol.93, pp.1288-1296, 2011.

E. Largy, F. Hamon, and M. Teulade-fichou, Development of a high-throughput G4-FID assay for screening and evaluation of small molecules binding quadruplex nucleic acid structures, Anal. Bioanal. Chem, vol.400, pp.3419-3427, 2011.

A. Oleksi, A. G. Blanco, R. Boer, I. Usón, J. Aymamí et al., Molecular recognition of a three-way DNA junction by a metallosupramolecular helicate, Angew. Chem, vol.118, pp.1249-1253, 2006.

D. R. Boer, J. M. Kerckhoffs, Y. Parajo, M. Pascu, I. Usón et al., Self-assembly of functionalizable two-component 3D DNA arrays through the induced formation of DNA three-way-junction branch points by supramolecular cylinders, Angew. Chem. Int. Ed, vol.49, pp.2336-2339, 2010.

L. Cerasino, M. J. Hannon, and E. Sletten, DNA three-way junction with a dinuclear iron (II) supramolecular helicate at the center: a NMR structural study, Inorg. Chem, vol.46, pp.6245-6251, 2007.

J. Malina, M. J. Hannon, and V. Brabec, Recognition of DNA three-way junctions by metallosupramolecular cylinders: gel electrophoresis studies, Chem. Eur. J, vol.13, pp.3871-3877, 2007.

Y. Parajó, J. Malina, I. Meistermann, G. J. Clarkson, M. Pascu et al., Effect of bridging ligand structure on the thermal stability and DNA binding properties of iron (II) triple helicates, pp.4868-4874, 2009.

A. C. Hotze, N. J. Hodges, R. E. Hayden, C. Sanchez-cano, C. Paines et al., Supramolecular iron cylinder with unprecedented DNA binding is a potent cytostatic and apoptotic agent without exhibiting genotoxicity, Chem. Biol, vol.15, pp.1258-1267, 2008.

J. Novotna, A. Laguerre, A. Granzhan, M. Pirrotta, M. Teulade-fichou et al., Cationic azacryptands as selective three-way DNA junction binding agents, Org. Biomol. Chem, vol.13, pp.215-222, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02125771

, Nucleic Acids Research, vol.46, issue.3, p.16, 2018.

S. Vuong, L. Stefan, P. Lejault, Y. Rousselin, F. Denat et al., Identifying three-way DNA junction-specific small-molecules, Biochimie, vol.94, pp.442-450, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02125720

L. Stefan, B. Bertrand, P. Richard, P. Le-gendre, F. Denat et al., Assessing the differential affinity of small molecules for noncanonical DNA structures, Chembiochem, vol.13, pp.1905-1912, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02125737

S. A. Barros and D. M. Chenoweth, Recognition of nucleic acid junctions using triptycene-based molecules, Angew. Chem., Int. Ed, vol.53, pp.13746-13750, 2014.

S. A. Barros and D. M. Chenoweth, Triptycene-based small molecules modulate (CAG)·(CTG) repeat junctions, Chem. Sci, vol.6, pp.4752-4755, 2015.

A. Granzhan, E. Largy, N. Saettel, and M. P. Teulade-fichou, Macrocyclic DNA-mismatch-binding ligands: structural determinants of selectivity, Chem. Eur. J, vol.16, pp.878-889, 2010.

A. Granzhan and M. P. Teulade-fichou, A fluorescent bisanthracene macrocycle discriminates between matched and mismatch-containing DNA, Chemistry, vol.15, pp.1314-1318, 2009.

T. Paris, J. Vigneron, J. Lehn, M. Cesario, J. Guilhem et al., Molecular recognition of anionic substrates. Crystal structures of the supramolecular inclusion complexes of terephthalate and isophthalate dianions with a bis-intercaland receptor molecule, J. Incl. Phenom. Macrocyclic Chem, vol.33, pp.191-202, 1999.

A. Granzhan and M. Teulade-fichou, Synthesis of mono-and bibrachial naphthalene-based macrocycles with pyrene or ferrocene units for anion detection, Tetrahedron, vol.65, pp.1349-1360, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00370475

N. P. Barry, F. Edafe, and B. Therrien, Anticancer activity of tetracationic arene ruthenium metalla-cycles, Dalton Trans, vol.40, pp.7172-7180, 2011.

B. Therrien, G. Süss-fink, P. Govindaswamy, A. K. Renfrew, and P. J. Dyson, The "Complex-in-a-Complex" cations [(acac) 2M?Ru 6 (p-iPrC 6 H 4 Me) 6 (tpt) 2 (dhbq) 3 ] 6+ : a trojan horse for cancer cells, Angew. Chem. Int. Ed, vol.47, pp.3773-3776, 2008.

N. P. Barry, O. Zava, P. J. Dyson, and B. Therrien, Excellent correlation between drug release and portal size in metalla-cage drug-delivery systems, Chem. Eur. J, vol.17, pp.9669-9677, 2011.

N. P. Barry, O. Zava, P. J. Dyson, and B. Therrien, Synthesis, characterization and anticancer activity of porphyrin-containing organometallic cubes, Aust. J. Chem, vol.63, pp.1529-1537, 2010.

N. P. Barry, P. Govindaswamy, J. Furrer, G. Süss-fink, and B. Therrien, -pyridyl) porphyrin panels and hydroxyquinonato-bridged diruthenium clips, Inorg. Chem. Commun, vol.10, issue.4, pp.1300-1303, 2008.

R. Haudecoeur, L. Stefan, F. Denat, and D. Monchaud, A model of smart G-quadruplex ligand, J. Am. Chem. Soc, vol.135, pp.550-553, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02125742

A. Granzhan, D. Monchaud, N. Saettel, A. Guédin, J. Mergny et al., One ring to bind them all"--part II: identification of promising G-quadruplex ligands by screening of cyclophane-type macrocycles, J. Nucleic Acids, 2010.

B. Therrien, Arene ruthenium cages: boxes full of surprises, Eur. J. Inorg. Chem, pp.2445-2453, 2009.

R. Haudecoeur, L. Stefan, and D. Monchaud, Multitasking water-soluble synthetic G-quartets: from preferential RNA-quadruplex interaction to biocatalytic activity, Chem. Eur. J, vol.19, pp.12739-12747, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02125745

M. Newman, R. Sfaxi, A. Saha, D. Monchaud, M. Teulade-fichou et al., The G-quadruplex-specific RNA helicase DHX36 regulates p53, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01680286

, Pre-mRNA 3 -end processing following UV-induced DNA damage, J. Mol. Biol, vol.429, pp.3121-3131

S. Phongtongpasuk, S. Paulus, J. Schnabl, R. K. Sigel, B. Spingler et al., Binding of a designed anti-cancer drug to the central cavity of an RNA three-way junction, Angew. Chem. Int. Ed, vol.52, pp.11513-11516, 2013.

Y. Liu and S. C. West, Happy Hollidays: 40th anniversary of the Holliday junction, Nat. Rev. Mol. Cell Biol, vol.5, pp.937-944, 2004.

S. A. Marras, F. R. Kramer, and S. Tyagi, Efficiencies of fluorescence resonance energy transfer and contact-mediated quenching in oligonucleotide probes, Nucleic Acids Res, vol.30, p.122, 2002.

S. A. Marras, Interactive fluorophore and quencher pairs for labelling fluorescent nucleic acid hybridization probes, Mol. Biotechnol, vol.38, pp.247-255, 2008.