M. Blagosklonny, Cell cycle arrest is not senescence, Aging, vol.3, issue.2, pp.94-101, 2011.
DOI : 10.18632/aging.100281

URL : http://europepmc.org/articles/pmc3082019?pdf=render

A. Young, M. Narita, M. Ferreira, K. Kirschner, M. Sadaie et al., Autophagy mediates the mitotic senescence transition, Genes & Development, vol.23, issue.7, pp.798-803, 2009.
DOI : 10.1101/gad.519709

URL : http://genesdev.cshlp.org/content/23/7/798.full.pdf

M. Narita, S. Nunez, E. Heard, M. Narita, A. Lin et al., Rb-Mediated Heterochromatin Formation and Silencing of E2F Target Genes during Cellular Senescence, Cell, vol.113, issue.6, pp.703-716, 2003.
DOI : 10.1016/S0092-8674(03)00401-X

URL : https://doi.org/10.1016/s0092-8674(03)00401-x

A. Iannello, T. Thompson, M. Ardolino, S. Lowe, and D. Raulet, p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells, The Journal of Experimental Medicine, vol.210, issue.10, pp.2057-2069, 2013.
DOI : 10.1073/pnas.0605752103

URL : http://jem.rupress.org/content/jem/210/10/2057.full.pdf

W. Xue, L. Zender, C. Miething, R. Dickins, E. Hernando et al., Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas, Nature, vol.98, issue.7128, pp.656-660, 2007.
DOI : 10.1073/pnas.211053698

A. Lujambio, L. Akkari, J. Simon, D. Grace, D. Tschaharganeh et al., Non-Cell-Autonomous Tumor Suppression by p53, Cell, vol.153, issue.2, pp.449-460, 2013.
DOI : 10.1016/j.cell.2013.03.020

URL : https://doi.org/10.1016/j.cell.2013.03.020

C. Michaloglou, L. Vredeveld, M. Soengas, C. Denoyelle, T. Kuilman et al., BRAFE600-associated senescence-like cell cycle arrest of human naevi, Nature, vol.436, issue.7051, pp.720-724, 2005.
DOI : 10.1038/nature03890

F. Rodier, J. Coppe, C. Patil, W. Hoeijmakers, D. Munoz et al., Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion, Nature Cell Biology, vol.11, issue.8, pp.973-979, 2009.
DOI : 10.1074/jbc.M408650200

URL : http://europepmc.org/articles/pmc2743561?pdf=render

J. Coppe, C. Patil, F. Rodier, Y. Sun, D. Munoz et al., Senescenceassociated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor, PLoS Biol, vol.6, pp.2853-2868, 2008.

T. Kuilman, C. Michaloglou, L. Vredeveld, S. Douma, R. Van-doorn et al., Oncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory Network, Cell, vol.133, issue.6, pp.1019-1031, 2008.
DOI : 10.1016/j.cell.2008.03.039

URL : https://doi.org/10.1016/j.cell.2008.03.039

J. Acosta, A. Banito, T. Wuestefeld, A. Georgilis, P. Janich et al., A complex secretory program orchestrated by the inflammasome controls paracrine senescence, Nature Cell Biology, vol.122, issue.8, 2013.
DOI : 10.1007/s00401-011-0841-z

URL : http://europepmc.org/articles/pmc3732483?pdf=render

F. Rodier and J. Campisi, Four faces of cellular senescence, The Journal of Cell Biology, vol.62, issue.4, pp.547-556, 2011.
DOI : 10.1038/sj.onc.1201178

URL : http://jcb.rupress.org/content/jcb/192/4/547.full.pdf

J. Coppe, K. Kauser, J. Campisi, and C. Beausejour, Secretion of Vascular Endothelial Growth Factor by Primary Human Fibroblasts at Senescence, Journal of Biological Chemistry, vol.13, issue.40, pp.29568-29574, 2006.
DOI : 10.1101/gad.1243304

S. Parrinello, J. Coppe, A. Krtolica, and J. Campisi, Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation, Journal of Cell Science, vol.118, issue.3, pp.485-496, 2005.
DOI : 10.1242/jcs.01635

URL : http://jcs.biologists.org/content/joces/118/3/485.full.pdf

B. Childs, D. Baker, J. Kirkland, J. Campisi, and J. Van-deursen, Senescence and apoptosis: dueling or complementary cell fates?, EMBO reports, vol.15, issue.11, pp.1139-1153, 2014.
DOI : 10.15252/embr.201439245

URL : http://embor.embopress.org/content/embor/15/11/1139.full.pdf

J. Jackson, V. Pant, Q. Li, L. Chang, A. Quintas-cardama et al., p53-Mediated Senescence Impairs the Apoptotic Response to Chemotherapy and Clinical Outcome in Breast Cancer, Cancer Cell, vol.21, issue.6, pp.793-806, 2012.
DOI : 10.1016/j.ccr.2012.04.027

URL : https://doi.org/10.1016/j.ccr.2012.04.027

S. De-carne-trecesson, Y. Guillemin, A. Belanger, A. Bernard, L. Preisser et al., Escape from p21-mediated Oncogene-induced Senescence Leads to Cell Dedifferentiation and Dependence on Anti-apoptotic Bcl-xL and MCL1 Proteins, Journal of Biological Chemistry, vol.25, issue.15, pp.12825-12838, 2011.
DOI : 10.1016/j.cell.2009.06.034

B. Jonchere, A. Vetillard, B. Toutain, D. Lam, A. Bernard et al., Irinotecan treatment and senescence failure promote the emergence of more transformed and invasive cells that depend on anti-apoptotic Mcl-1, Oncotarget, vol.6, issue.1, pp.409-426, 2015.
DOI : 10.18632/oncotarget.2774

URL : https://hal.archives-ouvertes.fr/hal-01396743

S. Ansieau and C. G. , Senescence versus apoptosis in chemotherapy, Oncotarget, vol.6, issue.7, pp.4551-4553, 2015.
DOI : 10.18632/oncotarget.3114

URL : http://doi.org/10.18632/oncotarget.3114

L. Pfannenstiel and B. Gastman, Mcl-1 and tumor cell persistence, Oncotarget, vol.6, issue.1, 2015.
DOI : 10.18632/oncotarget.3035

URL : https://doi.org/10.18632/oncotarget.3035

B. Chibaudel, C. Tournigand, T. Andre, and A. De-gramont, Therapeutic strategy in unresectable metastatic colorectal cancer, Therapeutic Advances in Medical Oncology, vol.22, issue.2, pp.75-89, 2012.
DOI : 10.1200/JCO.2001.19.21.4097

URL : http://journals.sagepub.com/doi/pdf/10.1177/1758834011431592

S. Staal, O. Connell, M. , and A. Cj, The Marriage of Growth Factor Inhibitors and Chemotherapy: Bliss or Bust?, Journal of Clinical Oncology, vol.27, issue.10, pp.1545-1548, 2009.
DOI : 10.1200/JCO.2008.20.3224

C. Holohan, V. Schaeybroeck, S. Longley, D. Johnston, and P. , Cancer drug resistance: an evolving paradigm, Nature Reviews Cancer, vol.14, issue.10, pp.714-726, 2013.
DOI : 10.1093/neuonc/nos158

A. Prahallad, C. Sun, S. Huang, D. Nicolantonio, F. Salazar et al., Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR, Nature, vol.15, issue.7388, pp.100-103, 2012.
DOI : 10.1016/j.ccr.2009.02.023

URL : https://iris.unito.it/retrieve/handle/2318/126889/19515/Prahallad_AuthorManuscript.pdf

C. Sun and R. Bernards, Feedback and redundancy in receptor tyrosine kinase signaling: relevance to cancer therapies, Trends in Biochemical Sciences, vol.39, issue.10, pp.465-474, 2014.
DOI : 10.1016/j.tibs.2014.08.010

C. Sun, L. Wang, S. Huang, G. Heynen, A. Prahallad et al., Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma, Nature, vol.15, issue.7494, pp.118-122, 2014.
DOI : 10.1016/j.ccr.2009.02.023

URL : https://iris.unito.it/retrieve/handle/2318/152041/24211/POST_2014-Sun%20et%20al_Reversible%20and%20adaptive_4aperto%20NEW.pdf

S. Chandarlapaty, A. Sawai, M. Scaltriti, V. Rodrik-outmezguine, O. Grbovic-huezo et al., AKT Inhibition Relieves Feedback Suppression of Receptor Tyrosine Kinase Expression and Activity, Cancer Cell, vol.19, issue.1, pp.58-71, 2011.
DOI : 10.1016/j.ccr.2010.10.031

URL : https://doi.org/10.1016/j.ccr.2010.10.031

A. Vigneron, E. Gamelin, and O. Coqueret, The EGFR-STAT3 Oncogenic Pathway Up-regulates the Eme1 Endonuclease to Reduce DNA Damage after Topoisomerase I Inhibition, Cancer Research, vol.68, issue.3, pp.815-825, 2008.
DOI : 10.1158/0008-5472.CAN-07-5115

URL : http://cancerres.aacrjournals.org/content/canres/68/3/815.full.pdf

J. Logue and D. Morrison, Complexity in the signaling network: insights from the use of targeted inhibitors in cancer therapy, Genes & Development, vol.26, issue.7, pp.641-650, 2012.
DOI : 10.1101/gad.186965.112

N. Rhodes, D. Heerding, D. Duckett, D. Eberwein, V. Knick et al., Characterization of an Akt Kinase Inhibitor with Potent Pharmacodynamic and Antitumor Activity, Cancer Research, vol.68, issue.7, pp.2366-2374, 2008.
DOI : 10.1158/0008-5472.CAN-07-5783

URL : http://cancerres.aacrjournals.org/content/canres/68/7/2366.full.pdf

S. De-carne-trecesson, Y. Guillemin, A. Belanger, A. Bernard, L. Preisser et al., Escape from p21-mediated oncogene-induced senescence leads to cell dedifferenciation and dependency on anti-apoptotic Bcl-xL and Mcl1 proteins, J Biol Chem, 2011.

B. Zhou, Y. Liao, W. Xia, B. Spohn, M. Lee et al., Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells, Nature Cell Biology, vol.266, issue.3, pp.245-252, 2001.
DOI : 10.1006/abio.1998.2955

L. Rossig, A. Jadidi, C. Urbich, C. Badorff, A. Zeiher et al., Akt-Dependent Phosphorylation of p21Cip1 Regulates PCNA Binding and Proliferation of Endothelial Cells, Molecular and Cellular Biology, vol.21, issue.16, pp.5644-5657, 2001.
DOI : 10.1128/MCB.21.16.5644-5657.2001

URL : http://mcb.asm.org/content/21/16/5644.full.pdf

L. Rossig, C. Badorff, Y. Holzmann, A. Zeiher, and S. Dimmeler, Degradation, Journal of Biological Chemistry, vol.266, issue.12, pp.9684-9689, 2002.
DOI : 10.1038/sj.onc.1204132

F. Rodier, D. Munoz, R. Teachenor, V. Chu, O. Le et al., DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion, Journal of Cell Science, vol.124, issue.1, pp.68-81, 2011.
DOI : 10.1242/jcs.071340

URL : http://jcs.biologists.org/content/joces/124/1/68.full.pdf

O. Leontieva, Z. Demidenko, and M. Blagosklonny, MEK drives cyclin D1 hyperelevation during geroconversion, Cell Death & Differentiation, vol.4, issue.9, pp.1241-1249, 2013.
DOI : 10.4161/cc.21908

URL : http://www.nature.com/cdd/journal/v20/n9/pdf/cdd201386a.pdf

O. Leontieva and M. Blagosklonny, CDK4/6-inhibiting drug substitutes for p21 and p16 in senescence: Duration of cell cycle arrest and MTOR activity determine geroconversion, Cell Cycle, vol.10, issue.18, pp.3063-3069, 2013.
DOI : 10.4161/cc.10.3.14707

V. Dulic, L. Drullinger, E. Lees, S. Reed, and G. Stein, Altered regulation of G1 cyclins in senescent human diploid fibroblasts: accumulation of inactive cyclin E-Cdk2 and cyclin D1-Cdk2 complexes., Proceedings of the National Academy of Sciences, vol.90, issue.23, pp.11034-11038, 1993.
DOI : 10.1073/pnas.90.23.11034

F. Bunz, A. Dutriaux, C. Lengauer, T. Waldman, S. Zhou et al., Requirement for p53 and p21 to Sustain G2 Arrest After DNA Damage, Science, vol.282, issue.5393, pp.1497-1501, 1998.
DOI : 10.1126/science.282.5393.1497

K. Polyak, T. Waldman, T. He, K. Kinzler, and B. Vogelstein, Genetic determinants of p53-induced apoptosis and growth arrest., Genes & Development, vol.10, issue.15, pp.1945-1952, 1996.
DOI : 10.1101/gad.10.15.1945

URL : http://genesdev.cshlp.org/content/10/15/1945.full.pdf

B. Wouters, A. Giaccia, N. Denko, and J. Brown, Loss of p21Waf1/Cip1 sensitizes tumors to radiation by an apoptosis-independent mechanism, Cancer Res, vol.57, pp.4703-4706, 1997.

T. Waldman, Y. Zhang, L. Dillehay, J. Yu, K. Kinzler et al., Cell-cycle arrest versus cell death in cancer therapy, Nature Medicine, vol.20, issue.9, pp.1034-1036, 1997.
DOI : 10.1101/gad.8.21.2540

S. Courapied, H. Sellier, S. De-carne-trecesson, A. Vigneron, A. Bernard et al., The cdk5 Kinase Regulates the STAT3 Transcription Factor to Prevent DNA Damage upon Topoisomerase I Inhibition, Journal of Biological Chemistry, vol.532, issue.35, pp.26765-26778, 2010.
DOI : 10.1200/JCO.2008.21.3264

URL : http://www.jbc.org/content/285/35/26765.full.pdf

E. Bolesta, L. Pfannenstiel, A. Demelash, M. Lesniewski, M. Tobin et al., Inhibition of Mcl-1 Promotes Senescence in Cancer Cells: Implications for Preventing Tumor Growth and Chemotherapy Resistance, Molecular and Cellular Biology, vol.32, issue.10, pp.1879-1892, 2012.
DOI : 10.1128/MCB.06214-11

URL : http://mcb.asm.org/content/32/10/1879.full.pdf

U. Maurer, C. Charvet, A. Wagman, E. Dejardin, and D. Green, Glycogen Synthase Kinase-3 Regulates Mitochondrial Outer Membrane Permeabilization and Apoptosis by Destabilization of MCL-1, Molecular Cell, vol.21, issue.6, pp.749-760, 2006.
DOI : 10.1016/j.molcel.2006.02.009

URL : https://doi.org/10.1016/j.molcel.2006.02.009

R. Hayward, J. Macpherson, J. Cummings, B. Monia, J. Smyth et al., Antisense Bcl-xl down-regulation switches the response to topoisomerase I inhibition from senescence to apoptosis in colorectal cancer cells, enhancing global cytotoxicity, Clin Cancer Res, vol.9, pp.2856-2865, 2003.
DOI : 10.1042/cs103047pb

S. Willis, L. Chen, G. Dewson, A. Wei, E. Naik et al., Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins, Genes & Development, vol.19, issue.11, pp.1294-1305, 2005.
DOI : 10.1101/gad.1304105

URL : http://genesdev.cshlp.org/content/19/11/1294.full.pdf

K. Okumura, S. Huang, and F. Sinicrope, Induction of Noxa Sensitizes Human Colorectal Cancer Cells Expressing Mcl-1 to the Small-Molecule Bcl-2/Bcl-xL Inhibitor, ABT-737, Clinical Cancer Research, vol.14, issue.24, pp.8132-8142, 2008.
DOI : 10.1158/1078-0432.CCR-08-1665

URL : http://clincancerres.aacrjournals.org/content/clincanres/14/24/8132.full.pdf

P. Gomez-bougie, E. Menoret, P. Juin, C. Dousset, C. Pellat-deceunynck et al., Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction, Biochemical and Biophysical Research Communications, vol.413, issue.3, pp.460-464, 2011.
DOI : 10.1016/j.bbrc.2011.08.118

P. Czabotar, E. Lee, M. Van-delft, C. Day, B. Smith et al., Structural insights into the degradation of Mcl-1 induced by BH3 domains, Proceedings of the National Academy of Sciences, vol.98, issue.2, pp.6217-6222, 2007.
DOI : 10.1021/j100058a043

URL : http://www.pnas.org/content/104/15/6217.full.pdf

A. Singh and J. Settleman, EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer, Oncogene, vol.104, issue.34, pp.4741-4751, 2010.
DOI : 10.1016/S0002-9440(10)65297-2

S. Mani, W. Guo, M. Liao, E. Eaton, A. Ayyanan et al., The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells, Cell, vol.133, issue.4, pp.704-715, 2008.
DOI : 10.1016/j.cell.2008.03.027

C. Eyler and J. Rich, Survival of the Fittest: Cancer Stem Cells in Therapeutic Resistance and Angiogenesis, Journal of Clinical Oncology, vol.26, issue.17, pp.2839-2845, 2008.
DOI : 10.1200/JCO.2007.15.1829

C. Scheel and R. Weinberg, Phenotypic plasticity and epithelial-mesenchymal transitions in cancer and normal stem cells?, International Journal of Cancer, vol.18, issue.10, pp.2310-2314, 2011.
DOI : 10.1016/j.ccr.2010.10.012

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.26311/pdf

S. Sahlberg, D. Spiegelberg, B. Glimelius, B. Stenerlow, and M. Nestor, Evaluation of Cancer Stem Cell Markers CD133, CD44, CD24: Association with AKT Isoforms and Radiation Resistance in Colon Cancer Cells, PLoS ONE, vol.77, issue.4, p.94621, 2014.
DOI : 10.1371/journal.pone.0094621.s004

M. Reimann, S. Lee, C. Loddenkemper, J. Dorr, V. Tabor et al., Tumor Stroma-Derived TGF-?? Limits Myc-Driven Lymphomagenesis via Suv39h1-Dependent Senescence, Cancer Cell, vol.17, issue.3, pp.262-272, 2010.
DOI : 10.1016/j.ccr.2009.12.043

URL : https://doi.org/10.1016/j.ccr.2009.12.043

J. Rodon, R. Dienstmann, V. Serra, and J. Tabernero, Development of PI3K inhibitors: lessons learned from early clinical trials, Nature Reviews Clinical Oncology, vol.72, issue.3, pp.143-153, 2013.
DOI : 10.1158/1538-7445.AM2012-CT-02

T. Satoh, H. Yasui, K. Muro, Y. Komatsu, S. Sameshima et al., Pharmacokinetic assessment of irinotecan, SN-38, and SN-38-glucuronide: a substudy of the FIRIS study, Anticancer Res, vol.33, pp.3845-3853, 2013.