, Tumeurs (CIT) funded and developed by the Ligue Nationale Contre le Cancer. It was supported by the 'Ligue contre le Cancer, Calvados and Orne Committee), the 'Conseil Regional de Basse-Normandie', and the French Government

E. B. , M. M. , E. V. , C. L. , E. V. et al., were recipients of a doctoral fellowship from the 'Ligue contre le Cancer' (Calvados CommitteeF.) and C.D. was recipient of a post-doctoral fellowship from the 'Conseil Regional de Basse-Normandie'. N.V. was a recipient of a doctoral fellowship from the French Ministry for Higher Education and Research. The present study was supported by a grant from l, Eure Committee: M.M

R. 1. Villedieu, M. Louis, M. Dutoit, S. Brotin, E. Lincet et al., Absence of Bcl-xL down-regulation in response to cisplatin is associated with chemoresistance in ovarian carcinoma cells???, Gynecologic Oncology, vol.105, issue.1, pp.31-44, 2007.
DOI : 10.1016/j.ygyno.2006.12.011

J. Tomasina, A. Malzert-freon, F. Giffard, E. Brotin, M. Louis et al., Sensitization of ovarian carcinoma cells to Bcl-xL-targeting strategies through indirect modulation of Mcl-1 activity by MR22388, a molecule of the tripentone family, Journal of Ovarian Research, vol.6, issue.1, pp.38-2013, 2013.
DOI : 10.1016/j.ccr.2012.02.028

URL : https://hal.archives-ouvertes.fr/hal-00840946

E. Brotin, M. Meryet-figuière, K. Simonin, R. Duval, M. Villedieu et al., and MCL-1 constitute pertinent targets in ovarian carcinoma and their concomitant inhibition is sufficient to induce apoptosis, International Journal of Cancer, vol.119, pp.885-895, 2010.
DOI : 10.3748/wjg.v12.i46.7472

URL : https://hal.archives-ouvertes.fr/hal-01650312

E. Varin, C. Denoyelle, E. Brotin, M. Meryet-figuière, F. Giffard et al., Downregulation of Bcl-x L and Mcl-1 is sufficient to induce cell death in mesothelioma cells highly refractory to conventional chemotherapy, Carcinogenesis, vol.61, issue.4, pp.984-993, 2010.
DOI : 10.1016/j.addr.2009.05.002

K. Simonin, E. Brotin, S. Dufort, S. Dutoit, D. Goux et al., Mcl-1 is an important determinant of the apoptotic response to the BH3-mimetic molecule HA14-1 in cisplatin-resistant ovarian carcinoma cells, Molecular Cancer Therapeutics, vol.8, issue.11, pp.3162-3170, 2009.
DOI : 10.1158/1535-7163.MCT-09-0493

K. Simonin, N. Diaye, M. Lheureux, S. Loussouarn, C. Dutoit et al., Platinum compounds sensitize ovarian carcinoma cells to ABT-737 by modulation of the Mcl-1/Noxa axis, Apoptosis, vol.77, issue.4, pp.492-508, 2013.
DOI : 10.1124/mol.109.060780

URL : https://hal.archives-ouvertes.fr/inserm-00872602

S. Lheureux, N. 'diaye, M. Blanc-fournier, C. Dugue, A. et al., experiment in human serous ovarian carcinoma, International Journal of Cancer, vol.17, issue.106, pp.340-350, 2015.
DOI : 10.1073/pnas.0905056106

C. Gloaguen, A. Voisin-chiret, J. Sopkova-de-oliveira-santos, J. Fogha, F. Gautier et al., -Targeting Strategies, Journal of Medicinal Chemistry, vol.58, issue.4, pp.1644-1668, 2015.
DOI : 10.1021/jm500672y

A. Letai, Diagnosing and exploiting cancer's addiction to blocks in apoptosis, Nature Reviews Cancer, vol.417, issue.2, pp.121-132, 2008.
DOI : 10.1038/bjc.1972.33

M. Certo, D. G. Moore, V. Nishino, M. Wei, G. Korsmeyer et al., Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members, Cancer Cell, vol.9, issue.5, pp.351-365, 2006.
DOI : 10.1016/j.ccr.2006.03.027

J. Belmar and S. Fesik, Small molecule Mcl-1 inhibitors for the treatment of cancer, Pharmacology & Therapeutics, vol.145, pp.76-84, 2015.
DOI : 10.1016/j.pharmthera.2014.08.003

S. Varadarajan, M. Vogler, M. Butterworth, D. Dinsdale, L. Walensky et al., Evaluation and critical assessment of putative MCL-1 inhibitors, Cell Death & Differentiation, vol.15, issue.11, pp.1475-1484, 2013.
DOI : 10.1593/neo.13230

J. Leverson, H. Zhang, J. Chen, S. Tahir, D. Phillips et al., Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax), Cell Death & Disease, vol.15, issue.1, p.1590, 2015.
DOI : 10.1172/JCI39964

G. Deleavey and M. Damha, Designing Chemically Modified Oligonucleotides for Targeted Gene Silencing, Chemistry & Biology, vol.19, issue.8, pp.937-954, 2012.
DOI : 10.1016/j.chembiol.2012.07.011

S. Colombo, X. Zeng, H. Ragelle, and C. Foged, Complexity in the therapeutic delivery of RNAi medicines: an analytical challenge, Expert Opinion on Drug Delivery, vol.14, issue.9, pp.1481-1495, 2014.
DOI : 10.1038/nature04303

A. Miller, Delivery of RNAi therapeutics: work in progress, Expert Review of Medical Devices, vol.7, issue.3, pp.781-811, 2013.
DOI : 10.1038/sj.embor.7400637

F. Takeshita, Y. Minakuchi, S. Nagahara, K. Honma, H. Sasaki et al., Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo, Proceedings of the National Academy of Sciences, vol.3, issue.8, pp.12177-12182, 2005.
DOI : 10.4161/cbt.3.8.994

Y. Takei, K. Kadomatsu, Y. Yuzawa, S. Matsuo, and T. Muramatsu, A Small Interfering RNA Targeting Vascular Endothelial Growth Factor as Cancer Therapeutics, Cancer Research, vol.64, issue.10, pp.3365-3370, 2004.
DOI : 10.1158/0008-5472.CAN-03-2682

I. Fujimoto and Y. Takei, Atelocollagen-mediated siRNA delivery: future promise for therapeutic application, Therapeutic Delivery, vol.5, issue.2012, pp.369-371, 2014.
DOI : 10.2131/jts.36.751

D. Svintradze and G. Mrevlishvili, Fiber molecular model of atelocollagen???small interfering RNA (siRNA) complex, International Journal of Biological Macromolecules, vol.37, issue.5, pp.283-286, 2005.
DOI : 10.1016/j.ijbiomac.2005.11.008

T. Ochiya, S. Nagahara, A. Sano, H. Itoh, and M. Terada, Biomaterials for Gene Delivery Atelocollagen-mediated Controlled Release of Molecular Medicines, Current Gene Therapy, vol.1, issue.1, pp.31-52, 2001.
DOI : 10.2174/1566523013348887

S. Forootan, . Bao-zz, F. Forootan, L. Kamalian, Y. Zhang et al., Atelocollagen-delivered siRNA targeting the FABP5 gene as an experimental therapy for prostate cancer in mouse xenografts, Int J Oncol, vol.36, pp.69-76, 2010.

E. Kawata, E. Ashihara, S. Kimura, K. Takenaka, K. Sato et al., Administration of PLK-1 small interfering RNA with atelocollagen prevents the growth of liver metastases of lung cancer, Molecular Cancer Therapeutics, vol.7, issue.9, pp.2904-2912, 2008.
DOI : 10.1158/1535-7163.MCT-08-0473

P. Mu, S. Nagahara, N. Makita, Y. Tarumi, K. Kadomatsu et al., Systemic delivery of siRNA specific to tumor mediated by atelocollagen: Combined therapy using siRNA targeting Bcl-xL and cisplatin against prostate cancer, International Journal of Cancer, vol.58, issue.12, pp.2978-2990, 2009.
DOI : 10.1016/S0962-8924(01)02124-9

Y. Minakuchi, F. Takeshita, N. Kosaka, H. Sasaki, Y. Yamamoto et al., Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing in vitro and in vivo, Nucleic Acids Research, vol.32, issue.13, pp.109-2004, 2004.
DOI : 10.1093/nar/gnh093

H. Nozawa, T. Tadakuma, T. Ono, M. Sato, S. Hiroi et al., Small interfering RNA targeting epidermal growth factor receptor enhances chemosensitivity to cisplatin, 5-fluorouracil and docetaxel in head and neck squamous cell carcinoma, Cancer Science, vol.61, issue.10, pp.1115-1124, 2006.
DOI : 10.1073/pnas.0501753102

K. Iwaki, K. Shibata, M. Ohta, Y. Endo, H. Uchida et al., A small interfering RNA targeting proteinase-activated receptor-2 is effective in suppression of tumor growth in a Panc1 xenograft model, International Journal of Cancer, vol.32, issue.3, pp.658-663, 2008.
DOI : 10.1177/002215549804600204

Y. Takei, K. Kadomatsu, T. Goto, and T. Muramatsu, Combinational antitumor effect of siRNA against midkine and paclitaxel on growth of human prostate cancer xenografts, Cancer, vol.57, issue.4, pp.864-873, 2006.
DOI : 10.1074/jbc.M005911200

I. Takigami, T. Ohno, Y. Kitade, A. Hara, A. Nagano et al., Synthetic siRNA targeting the breakpoint of EWS/Fli-1 inhibits growth of Ewing sarcoma xenografts in a mouse model, International Journal of Cancer, vol.20, issue.1, pp.216-226, 2011.
DOI : 10.1128/MCB.14.5.3230

E. Ashihara, E. Kawata, Y. Nakagawa, C. Shimazaski, J. Kuroda et al., ??-Catenin Small Interfering RNA Successfully Suppressed Progression of Multiple Myeloma in a Mouse Model, Clinical Cancer Research, vol.15, issue.8, pp.2731-2738, 2009.
DOI : 10.1158/1078-0432.CCR-08-1350

H. Sudo, A. Tsuji, A. Sugyo, M. Kohda, C. Sogawa et al., Knockdown of COPA, Identified by Loss-of-Function Screen, Induces Apoptosis and Suppresses Tumor Growth in Mesothelioma Mouse Model, Genomics, vol.95, issue.4, pp.210-216, 2010.
DOI : 10.1016/j.ygeno.2010.02.002

T. Koyanagi, Y. Suzuki, Y. Saga, S. Machida, Y. Takei et al., delivery of siRNA targeting vasohibin-2 decreases tumor angiogenesis and suppresses tumor growth in ovarian cancer, Cancer Science, vol.102, issue.12, pp.1705-1710, 2013.
DOI : 10.1111/j.1349-7006.2010.01812.x

M. Tasaki, K. Shimada, H. Kimura, K. Tsujikawa, and N. Konishi, ALKBH3, a human AlkB homologue, contributes to cell survival in human non-small-cell lung cancer, British Journal of Cancer, vol.13, issue.4, pp.700-706, 2011.
DOI : 10.1073/pnas.161288698

T. Fujita, K. Yanagihara, F. Takeshita, K. Aoyagi, T. Nishimura et al., prolongs the survival of scirrhous gastric cancer model mice, Cancer Science, vol.26, issue.2, pp.214-222, 2013.
DOI : 10.1038/sj.onc.1210475

K. Azuma, K. Nakashiro, T. Sasaki, H. Goda, J. Onodera et al., Anti-tumor effect of small interfering RNA targeting the androgen receptor in human androgen-independent prostate cancer cells, Biochemical and Biophysical Research Communications, vol.391, issue.1, pp.1075-1079, 2010.
DOI : 10.1016/j.bbrc.2009.12.024

T. Sasaki, K. Nakashiro, H. Tanaka, K. Azuma, H. Goda et al., Knockdown of Akt isoforms by RNA silencing suppresses the growth of human prostate cancer cells in vitro and in vivo, Biochemical and Biophysical Research Communications, vol.399, issue.1, pp.79-83, 2010.
DOI : 10.1016/j.bbrc.2010.07.045

S. Woo, K. Min, B. Seo, J. Nam, K. Choi et al., Cafestol overcomes ABT-737 resistance in Mcl-1-overexpressed renal carcinoma Caki cells through downregulation of Mcl-1 expression and upregulation of Bim expression, Cell Death & Disease, vol.298, issue.11, p.1514, 2014.
DOI : 10.1128/MCB.01266-09

K. Sadahira, M. Sagawa, T. Nakazato, H. Uchida, Y. Ikeda et al., Gossypol induces apoptosis in multiple myeloma cells by inhibition of interleukin-6 signaling and Bcl-2/Mcl-1 pathway, International Journal of Oncology, vol.45, issue.6, pp.2278-2286, 2014.
DOI : 10.3892/ijo.2014.2652

X. Cao, J. Yap, M. Newell-rogers, C. Peddaboina, W. Jiang et al., The novel BH3 ??-helix mimetic JY-1-106 induces apoptosis in a subset of cancer cells (lung cancer, colon cancer and mesothelioma) by disrupting Bcl-xL and Mcl-1 protein???protein interactions with Bak, Molecular Cancer, vol.12, issue.1, pp.42-2013, 2013.
DOI : 10.1371/journal.pone.0048817

G. Ozcan, B. Ozpolat, R. Coleman, A. Sood, and G. Lopez-berestein, Preclinical and clinical development of siRNA-based therapeutics, Advanced Drug Delivery Reviews, vol.87, pp.108-119, 2015.
DOI : 10.1016/j.addr.2015.01.007

S. Wu, G. Lopez-berestein, G. Calin, and A. Sood, RNAi Therapies: Drugging the Undruggable, Science Translational Medicine, vol.7, issue.6, pp.240-247, 2014.
DOI : 10.1038/nnano.2012.73

J. Guo, K. Fisher, R. Darcy, J. Cryan, O. Driscoll et al., Therapeutic targeting in the silent era: advances in non-viral siRNA delivery, Molecular BioSystems, vol.6, pp.1143-1161, 2010.
DOI : 10.1039/c001050m

R. Kanasty, J. Dorkin, A. Vegas, and D. Anderson, Delivery materials for siRNA therapeutics, Nature Materials, vol.134, issue.11, pp.967-977, 2013.
DOI : 10.1021/ja301621z

K. Nakazawa, T. Nemoto, T. Hata, Y. Seyama, S. Nagahara et al., Single-injection ornithine decarboxylase-directed antisense therapy using atelocollagen to suppress human cancer growth, Cancer, vol.33, issue.5, pp.993-1002, 2007.
DOI : 10.4161/cbt.209

G. Bosma, R. Custer, and M. Bosma, A severe combined immunodeficiency mutation in the mouse, Nature, vol.17, issue.5900, pp.527-530, 1983.
DOI : 10.1038/301527a0

E. Larsson, C. Sander, and D. Marks, mRNA turnover rate limits siRNA and microRNA efficacy, Mol Syst Biol, vol.6, p.433, 2010.
DOI : 10.1038/msb.2010.113

URL : http://msb.embopress.org/content/6/1/454.full.pdf

L. Wilsbacher, S. Yamazaki, E. Herzog, E. Song, L. Radcliffe et al., Photic and circadian expression of luciferase in mPeriod1-luc transgenic mice in vivo, Proceedings of the National Academy of Sciences, vol.288, issue.5465, pp.489-494, 2002.
DOI : 10.1126/science.288.5465.483

J. Reed, Y. Tsujimoto, S. Epstein, M. Cuddy, T. Slabiak et al., Regulation of bcl-2 gene expression in lymphoid cell lines containing normal #18 or t(14;18) chromosomes, Oncogene Res, vol.4, pp.271-282, 1989.

G. Wei, A. Margolin, L. Haery, E. Brown, L. Cucolo et al., Chemical Genomics Identifies Small-Molecule MCL1 Repressors and BCL-xL as a Predictor of MCL1 Dependency, Cancer Cell, vol.21, issue.4, pp.547-562, 2012.
DOI : 10.1016/j.ccr.2012.02.028

R. Rooswinkel, B. Van-de-kooij, E. De-vries, M. Paauwe, R. Braster et al., Antiapoptotic potency of Bcl-2 proteins primarily relies on their stability, not binding selectivity, Blood, vol.123, issue.18, pp.2806-2815, 2014.
DOI : 10.1182/blood-2013-08-519470