L. Huang, C. Perrault, J. Coelho-martins, C. Hu, C. Dulong et al., Induction of acquired drug resistance in endothelial cells and its involvement in anticancer therapy, J Hematol Oncol, vol.6, p.49, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00845842

L. Huang, C. Hu, D. Benedetto, M. Varin, R. Liu et al., Induction of multiple drug resistance in HMEC-1 endothelial cells after long-term exposure to sunitinib, Onco Targets Ther, vol.7, pp.2249-2255, 2014.

L. Huang, C. Hu, D. Benedetto, M. Varin, R. Liu et al., Cross-drug resistance to sunitinib induced by doxorubicin in endothelial cells, Oncol Lett, vol.9, pp.1287-1292, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02378497

H. Naito, T. Wakabayashi, H. Kidoya, F. Muramatsu, K. Takara et al., Endothelial side population cells contribute to tumor angiogenesis and antiangiogenic drug resistance, Cancer Res, vol.76, pp.3200-3210, 2016.

S. M. Weis and D. A. Cheresh, Tumor angiogenesis: Molecular pathways and therapeutic targets, Nat Med, vol.17, pp.1359-1370, 2011.

D. B. Mendel, A. D. Laird, X. Xin, S. G. Louie, J. G. Christensen et al., In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: Determination of a pharmacokinetic/pharmacodynamic relationship, Clin Cancer Res, vol.9, pp.327-337, 2003.

G. C. Jayson, R. Kerbel, L. M. Ellis, and A. L. Harris, Antiangiogenic therapy in oncology: Current status and future directions, Lancet, vol.388, pp.518-529, 2016.

G. Housman, S. Byler, S. Heerboth, K. Lapinska, M. Longacre et al., Drug resistance in cancer: An overview, Cancers (Basel), vol.6, pp.1769-1792, 2014.

E. J. Huijbers, J. R. Van-beijnum, V. L. Thijssen, S. Sabrkhany, P. Nowak-sliwinska et al., Role of the tumor stroma in resistance to anti-angiogenic therapy, Drug Resist Updat, vol.25, pp.26-37, 2016.

Y. Gu, H. Lu, C. Boisson-vidal, H. Li, G. Bousquet et al., Resistance to anti-angiogenic therapy: A clinical and scientific current issue, Med Sci, vol.32, pp.370-377, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02327134

T. Nunes, D. Hamdan, C. Leboeuf, E. Bouchtaoui, M. Gapihan et al., Targeting cancer stem cells to overcome chemoresistance, Int J Mol Sci, vol.19, p.4036, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02124684

A. C. Macintyre and D. J. Cutler, The potential role of lysosomes in tissue distribution of weak bases, Biopharm Drug Dispos, vol.9, pp.513-526, 1988.

M. Duvvuri and J. P. Krise, Intracellular drug sequestration events associated with the emergence of multidrug resistance: A mechanistic review, Front Biosci, vol.10, pp.1499-1509, 2005.

F. Kazmi, T. Hensley, C. Pope, R. S. Funk, G. J. Loewen et al., Lysosomal sequestration (trapping) of lipophilic amine (cationic amphiphilic) drugs in immortalized human hepatocytes (Fa2N-4 cells), Drug Metab Dispos, vol.41, pp.897-905, 2013.

K. J. Gotink, H. J. Broxterman, M. Labots, R. R. De-haas, H. Dekker et al., Lysosomal sequestration of sunitinib: A novel mechanism of drug resistance, Clin Cancer Res, vol.17, pp.7337-7346, 2011.

T. Yamagishi, S. Sahni, D. M. Sharp, A. A. Jansson, P. J. Richardson et al., P-glycoprotein mediates drug resistance via a novel mechanism involving lysosomal sequestration, J Biol Chem, vol.288, pp.31761-31771, 2013.

B. Zhitomirsky and Y. G. Assaraf, Lysosomal sequestration of hydrophobic weak base chemotherapeutics triggers lysosomal biogenesis and lysosome-dependent cancer multidrug resistance, Oncotarget, vol.6, pp.1143-1156, 2015.

B. Chapuy, R. Koch, U. Radunski, S. Corsham, N. Cheong et al., Acquired tumor cell resistance to sunitinib causes resistance in a HT-29 human colon cancer xenograft mouse model without affecting sunitinib biodistribution or the tumor microvasculature, Leukemia, vol.22, pp.844-853, 2008.

M. Santoni, C. Amantini, M. B. Morelli, S. Liberati, V. Farfariello et al., Pazopanib and sunitinib trigger autophagic and non-autophagic death of bladder tumour cells, Br J Cancer, vol.109, pp.1040-1050, 2013.

T. Ikeda, K. A. Ishii, Y. Saito, M. Miura, A. Otagiri et al., Inhibition of autophagy enhances sunitinib-induced cytotoxicity in rat pheochromocytoma PC12 cells, J Pharmacol Sci, vol.121, pp.67-73, 2013.

A. Shouman, S. El-demerdash, E. , E. M. , and A. Ab, Chloroquine synergizes sunitinib cytotoxicity via modulating autophagic, apoptotic and angiogenic machineries, Chem Biol Interact, vol.217, pp.28-40, 2014.

S. Giuliano, Y. Cormerais, M. Dufies, R. Grépin, P. Colosetti et al., Resistance to sunitinib in renal clear cell carcinoma results from sequestration in lysosomes and inhibition of the autophagic flux, Autophagy, vol.11, pp.1891-1904, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02108861

M. C. Maiuri, E. Zalckvar, A. Kimchi, and G. Kroemer, Self-eating and self-killing: Crosstalk between autophagy and apoptosis, Nat Rev Mol Cell Biol, vol.8, pp.741-752, 2007.

E. Turco and S. Martens, Insights into autophagosome biogenesis from in vitro reconstitutions, J Struct Biol, vol.196, pp.29-36, 2016.

M. Antonioli, D. Rienzo, M. , P. , M. Fimia et al., Emerging mechanisms in initiating and terminating autophagy, Trends Biochem Sci, vol.42, pp.28-41, 2017.

D. C. Rubinsztein, P. Codogno, and B. Levine, Autophagy modulation as a potential therapeutic target for diverse diseases, Nat Rev Drug Discov, vol.11, pp.709-730, 2012.

G. J. Yoshida, Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: From pathophysiology to treatment, J Hematol Oncol, vol.10, p.67, 2017.

J. Pu, C. M. Guardia, T. Keren-kaplan, and J. S. Bonifacino, Mechanisms and functions of lysosome positioning, J Cell Sci, vol.129, pp.4329-4339, 2016.

Y. Sr and N. Mizushima, Monitoring and measuring autophagy, Int J Mol Sci, vol.18, p.1865, 2017.

E. L. Eskelinen, Roles of LAMP-1 and LAMP-2 in lysosome biogenesis and autophagy, Mol Aspects Med, vol.27, pp.495-502, 2006.

S. Gagliardi, R. M. Farina, and C. , Chemistry and structure activity relationships of bafilomycin A1, a potent and selective inhibitor of the vacuolar H+-ATPase, Curr Med Chem, vol.6, pp.1197-1212, 1999.

M. Mauthe, I. Orhon, C. Rocchi, X. Zhou, M. Luhr et al., Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion, Autophagy, vol.14, pp.1435-1455, 2018.

J. Heuser, Changes in lysosome shape and distribution correlated with changes in cytoplasmic pH, J Cell Biol, vol.108, pp.855-864, 1989.

L. Ga-l-luzzi, E. H. Baeh-re-cke, A. Ba-l-labio, P. Boya, J. M. Bravo-san-pedro et al., Molecular definitions of autophagy and related processes, EMBO J, vol.36, pp.1811-1836, 2017.

H. Maes, N. Rubio, A. D. Garg, and P. Agostinis, Autophagy: Shaping the tumor microenvironment and therapeutic response, Trends Mol Med, vol.19, pp.428-446, 2013.

H. Maes, A. Kuchnio, A. Peric, S. Moens, K. Nys et al., Tumor vessel normalization by chloroquine independent of autophagy, Cancer Cell, vol.26, pp.190-206, 2014.

D. Huang, Y. Ding, Y. Li, W. M. Luo, Z. F. Zhang et al., Sunitinib acts primarily on tumor endothelium rather than tumor cells to inhibit the growth of renal cell carcinoma, Cancer Res, vol.70, pp.1053-1062, 2010.

J. J. Moslehi, Cardiovascular toxic effects of targeted cancer therapies, N Engl J Med, vol.375, pp.1457-1467, 2016.

M. Bretagne, P. Boudou-rouquette, O. Huillard, A. Thomas-schoemann, A. Chahwakilian et al., Tyrosine kinase inhibiting the VEGF pathway and elderly people: Tolerance, pre-treatment assessment and side effects management, Bull Cancer, vol.103, pp.259-272, 2016.

, This work is licensed under a Creative Commons Attribution-NonCommercial