C. H. Topham and S. S. Taylor, Mitosis and apoptosis: how is the balance set?, Curr. Opin. Cell Biol, vol.25, pp.780-785, 2013.

K. E. Gascoigne and S. S. Taylor, Cancer cells display profound intra-and interline variation following prolonged exposure to anti-mitotic drugs, Cancer Cell, vol.14, pp.111-122, 2008.

S. Barillé-nion, N. Bah, E. Véquaud, and P. Juin, Regulation of cancer cell survival by BCL2 family members upon prolonged mitotic arrest: opportunities for anticancer therapy, Anticancer Res, vol.32, pp.4225-4233, 2012.

P. Juin, O. Geneste, F. Gautier, S. Depil, and M. Campone, Decoding and unlocking the BCL-2 dependency of cancer cells, Nat. Rev. Cancer, vol.13, pp.455-465, 2013.

J. Montero, Drug-induced death signaling strategy rapidly predicts cancer response to chemotherapy, Cell, vol.160, pp.977-989, 2015.

O. Kutuk and A. Letai, Displacement of Bim by Bmf and Puma rather than increase in Bim level mediates paclitaxel-induced apoptosis in breast cancer cells, Cell Death Differ, vol.17, pp.1624-1635, 2010.

N. Bah, Bcl-xL controls a switch between cell death modes during mitotic arrest, Cell Death Dis, vol.5, p.1291, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02446956

P. Wang, Phosphorylation of the pro-apoptotic BH3-only protein bid primes mitochondria for apoptosis during mitotic arrest, Cell Rep, vol.7, pp.661-671, 2014.

O. J. Shah, Bcl-XL represents a druggable molecular vulnerability during aurora B inhibitor-mediated polyploidization, Proc. Natl Acad. Sci. USA, vol.107, pp.12634-12639, 2010.

M. D. Haschka, The NOXA-MCL1-BIM axis defines lifespan on extended mitotic arrest, Nat. Commun, vol.6, pp.6891-6904, 2015.

T. Ni-chonghaile, Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy, Science, vol.334, pp.1129-1133, 2011.

D. R. Green, A BH3 mimetic for killing cancer cells, Cell, vol.165, p.1560, 2016.

S. R. Oakes, Sensitization of BCL-2-expressing breast tumors to chemotherapy by the BH3 mimetic ABT-737, Proc. Natl Acad. Sci. USA, vol.109, pp.2766-2771, 2012.

J. D. Leverson, Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy, Sci. Transl. Med, vol.7, pp.279-319, 2015.

V. Nangia, Exploiting MCL1 dependency with combination MEK + MCL1 inhibitors leads to induction of apoptosis and tumor regression in KRAS-mutant non-small cell, Lung Cancer Cancer Discov, vol.8, pp.1598-1613, 2018.

K. J. Mackenzie, cGAS surveillance of micronuclei links genome instability to innate immunity, Nature, vol.548, pp.461-465, 2017.

S. M. Harding, Mitotic progression following DNA damage enables pattern recognition within micronuclei, Nature, vol.548, pp.466-470, 2017.

A. Rongvaux, Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA, Cell, vol.159, pp.1563-1577, 2014.

M. J. White, Apoptotic caspases suppress mtDNA-induced STINGmediated type I IFN production, Cell, vol.159, pp.1549-1562, 2014.

C. Séveno, D. Loussouarn, M. Campone, P. Juin, and S. Barillé-nion, Notch inhibition triggers cell death in breast cancer cells and breast tumors through Noxa activation, Breast Cancer Res, vol.14, p.96, 2012.

J. M. Ramanjulu, Design of amidobenzimidazole STING receptor agonists with systemic activity, Nature, vol.564, pp.439-443, 2018.

Z. Wang, Complex regulation pattern of IRF3 activation revealed by a novel dimerization reporter system, J. Immunol, vol.196, pp.4322-4430, 2016.

E. M. Hatch, A. H. Fischer, T. J. Deerinck, and M. W. Hetzer, Catastrophic nuclear envelope collapse in cancer cell micronuclei, Cell, vol.154, pp.47-60, 2013.

L. He, STING signaling in tumorigenesis and cancer therapy: a friend or foe?, Cancer Lett, vol.402, pp.203-212, 2017.

G. N. Barber, STING: infection, inflammation and cancer, Nat. Rev. Immunol, vol.15, pp.760-770, 2015.

Y. Kitai, DNA-containing exosomes derived from cancer cells treated with topotecan activate a STING-dependent pathway and reinforce antitumor immunity, J. Immunol, vol.198, pp.1649-1659, 2017.

T. Xia, H. Konno, J. Ahn, and G. N. Barber, Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis, Cell Rep, vol.14, pp.282-297, 2016.

C. Pantelidou, PARP inhibitor efficacy depends on CD8+ T-cell recruitment via Intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast cancer, Cancer Discov, vol.9, pp.722-737, 2019.

M. E. Legrier, Activation of IFN/STAT1 signalling predicts response to chemotherapy in oestrogen receptor-negative breast cancer, Br. J. Cancer, vol.114, pp.177-187, 2016.

J. A. Sprowl, Alterations in tumor necrosis factor signaling pathways are associated with cytotoxicity and resistance to taxanes: a study in isogenic resistant tumor cells, Breast Cancer Res, vol.14, p.2, 2012.

M. Budhwani, R. Mazzieri, and R. Dolcetti, Plasticity of Type I interferonmediated responses in cancer therapy: from anti-tumor immunity to resistance, Front. Oncol, vol.8, p.322, 2018.

F. Balkwill, Tumour necrosis factor and cancer, Nat. Rev. Cancer, vol.9, pp.361-371, 2009.

A. Sistigu, Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy, Nat. Med, vol.20, pp.1301-1309, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02047408

Y. Okubo, T. Mera, L. Wang, and D. L. Faustman, Homogeneous expansion of human T-regulatory cells via tumor necrosis factor receptor 2, Sci. Rep, vol.3, pp.3153-3164, 2013.

M. Brault, T. M. Olsen, J. Martinez, D. B. Stetson, and A. Oberst, Intracellular nucleic acid sensing triggers necroptosis through synergistic type I IFN and TNF signaling, J. Immunol, vol.200, pp.2748-2756, 2018.

Y. Sun and D. W. Leaman, Involvement of Noxa in cellular apoptotic responses to interferon, double-stranded RNA, and virus infection, J. Biol. Chem, vol.280, pp.15561-15568, 2005.

G. Wei, Chemical genomics identifies small-molecule MCL1 repressors and BCL-xL as a predictor of MCL1 dependency, Cancer Cell, vol.21, pp.547-562, 2012.

R. S. Soderquist, Systematic mapping of BCL-2 gene dependencies in cancer reveals molecular determinants of BH3 mimetic sensitivity, Nat. Commun, vol.9, p.3513, 2018.

J. M. Balko, Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets, Cancer Discov, vol.4, pp.232-245, 2014.

S. E. Logue, Inhibition of IRE1 RNase activity modulates the tumor cell secretome and enhances response to chemotherapy, Nat. Commun, vol.9, p.3267, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01879955

A. S. Qadir, CD95/Fas increases stemness in cancer cells by inducing a STAT1-dependent type I interferon response, Cell Rep, vol.18, pp.2373-2386, 2017.

R. R. Weichselbaum, An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer, Proc. Natl Acad. Sci. USA, vol.105, pp.18490-18495, 2008.

H. Cheon, IFN?-dependent increases in STAT1, STAT2, and IRF9 mediate resistance to viruses and DNA damage, EMBO J, vol.32, pp.2751-2763, 2013.

S. Terawaki, IFN-? directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity, J. Immunol, vol.186, pp.2772-2779, 2011.

F. Bertrand, TNF? blockade overcomes resistance to anti-PD-1 in experimental melanoma, Nat. Commun, vol.8, p.2256, 2017.

J. Shi, Y. Zhou, H. C. Huang, T. J. Mitchison, and . Navitoclax, ABT-263) accelerates apoptosis during drug-induced mitotic arrest by antagonizing Bcl-xL, Cancer Res, vol.71, pp.4518-4526, 2011.

M. Wong, ABT-263) reduces Bcl-x(L)-mediated chemoresistance in ovarian cancer models, Mol. Cancer Ther, vol.11, pp.1026-1035, 2012.

E. G. Panayotopoulou, Targeting of apoptotic pathways by SMAC or BH3 mimetics distinctly sensitizes paclitaxel-resistant triple negative breast cancer cells, Oncotarget, vol.8, pp.45088-45104, 2017.

G. Vlahovic, A phase I safety and pharmacokinetic study of ABT-263 in combination with carboplatin/paclitaxel in the treatment of patients with solid tumors, Invest. N. Drugs, vol.32, pp.976-984, 2014.

L. Gandhi, Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors, J. Clin. Oncol, vol.29, pp.909-916, 2011.

J. Montero and A. Letai, Why do BCL-2 inhibitors work and where should we use them in the clinic?, Cell Death Differ, vol.25, pp.56-64, 2018.

L. A. Díaz-martínez, Genome-wide siRNA screen reveals coupling between mitotic apoptosis and adaptation, EMBO J, vol.33, pp.1960-1976, 2014.

M. F. Gulen, Signalling strength determines pro-apoptotic functions of STING, Nat. Commun, vol.8, p.427, 2017.

N. Yatim, RIPK1 and NF-?B signaling in dying cells determines crosspriming of CD8 + T cells, Science, vol.350, pp.328-334, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01380732

E. Giampazolias, Mitochondrial permeabilization engages NF-?Bdependent anti-tumour activity under caspase deficiency, Nat. Cell Biol, vol.19, pp.1116-1129, 2017.

A. P. West, Mitochondrial DNA stress primes the antiviral innate immune response, Nature, vol.520, pp.553-557, 2015.

N. Yatim, S. Cullen, and M. L. Albert, Dying cells actively regulate adaptive immune responses, Nat. Rev. Immunol, vol.17, pp.262-275, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01491773

P. D. Bhola, Functionally identifiable apoptosis-insensitive subpopulations determine chemoresistance in acute myeloid leukemia, J. Clin. Invest, vol.126, pp.3827-3836, 2016.

S. F. Bakhoum, Chromosomal instability drives metastasis through a cytosolic DNA response, Nature, vol.553, pp.467-472, 2018.

K. Jeganathan, L. Malureanu, D. J. Baker, S. C. Abraham, and J. Van-deursen, Bub1 mediates cell death in response to chromosome missegregation and acts to suppress spontaneous tumorigenesis, J. Cell Biol, vol.179, pp.255-267, 2007.

N. E. Sanjana, O. Shalem, and F. Zhang, Improved vectors and genome-wide libraries for CRISPR screening, Nat. Methods, vol.11, pp.783-784, 2014.

N. Sachs, A living captures disease biobank of breast cancer organoids heterogeneity, Cell, vol.172, pp.373-386, 2018.

Y. S. Derose, Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes, Nat. Med, vol.17, pp.1514-1520, 2011.

P. Jézéquel, Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response, Breast Cancer Res, vol.17, p.43, 2015.

K. Yoshihara, The cytoplasmic DNA sensor cGAS promotes mitotic cell death, 2017.

S. Kilens, Parallel derivation of isogenic human primed and naive induced pluripotent stem cells, Nat. Commun, vol.9, p.360, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-01758726

M. V. Kuleshov, Enrichr: a comprehensive gene set enrichment analysis web server 2016 update, Nucleic Acids Res, vol.44, pp.90-97, 2016.