, Relationship between plasma and tumor SN-38 concentrations in mice treated by bevacizumab SN-38 plasma and tumor mean values of mice treated with irinotecan and bevacizumab were divided by SN- 38 plasma and tumor mean values of mice treated with irinotecan alone

, Mean plasma and tumor concentrations ratios of SN38 As shown on figure 3, higher SN-38 plasma value ratios were observed at two points, 3 and 4 h. Higher SN- 38 tumor value ratios were observed at two points, Figure, vol.3

, REFERENCES

T. Velde, E. Vogten, J. Gebbink, and M. , Enhanced antitumour efficacy by combining conventional chemotherapy with angiostatin or endostatin in a liver metastasis model, British Journal of Surgery, vol.13, issue.10, pp.1302-1309, 2002.
DOI : 10.1007/BF00689637

B. Emmink, W. Van-houdt, and R. Vries, Differentiated Human Colorectal Cancer Cells Protect Tumor-Initiating Cells From Irinotecan, Gastroenterology, vol.141, issue.1, pp.269-278, 2011.
DOI : 10.1053/j.gastro.2011.03.052

C. Chu, C. Abbara, and M. Tandia, Cetuximab increases concentrations of irinotecan and of its active metabolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice, Fundamental & Clinical Pharmacology, vol.31, issue.6, pp.652-660, 2014.
DOI : 10.1124/dmd.31.1.21

F. Wang, Y. Chen, and L. Huang, Cetuximab enhanced the efficacy of chemotherapeutic agent in ABCB1/P-glycoprotein-overexpressing cancer cells, Oncotarget, vol.6, issue.38, pp.40850-40865, 2015.
DOI : 10.18632/oncotarget.5813

L. Li, A. Jiang, and O. Dong, MDR1/P-gp and VEGF Synergistically Enhance the Invasion of Hep-2 Cells with Multidrug Resistance Induced by Taxol, Annals of Surgical Oncology, vol.8, issue.5, pp.1421-1429, 2009.
DOI : 10.4049/jimmunol.171.8.4340

H. Liang, Y. Zhang, and J. Zhang, Effects of arsenic trioxide on expressions of vascular endothelial growth factor and P-glycoprotein in multidrug resistant leukemia cell line K562/A02, Journal of Chinese Integrative Medicine, vol.5, issue.6, pp.647-650, 2007.
DOI : 10.3736/jcim20070609

A. Broggini-tenzer, A. Sharma, and K. Nytko, Combines treatment strategies for microtubule stabilizing agent-resistant tumors
DOI : 10.1093/jnci/dju504

URL : https://academic.oup.com/jnci/article-pdf/107/4/dju504/17313997/dju504.pdf

, JNCI J Natl Cancer Inst, vol.107, issue.4, p.504, 2015.

J. Bénard, D. Silva, J. , D. Blois, and M. , Characterization of a human ovarian adenocarcinoma line, IGROV1, in tissue culture and in nude mice, Cancer Res, vol.45, pp.4970-4979, 1985.

C. Jovelet, J. Bénard, and F. Forestier, Inhibition of P-glycoprotein functionality by vandetanib may reverse cancer cell resistance to doxorubicin, European Journal of Pharmaceutical Sciences, vol.46, issue.5, pp.484-491, 2012.
DOI : 10.1016/j.ejps.2012.03.012

T. Mosman, Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays, Journal of Immunological Methods, vol.65, issue.1-2, p.55, 1983.
DOI : 10.1016/0022-1759(83)90303-4

P. Burtin, F. Mentre, and J. Van-bree, Sparse sampling for assessment of drug exposure in toxicological studies, European Journal of Drug Metabolism and Pharmacokinetics, vol.12, issue.2, pp.105-111, 1996.
DOI : 10.1002/cpt1979263294

A. Bailer, Testing for the equality of area under the curves when using destructive measurement techniques, Journal of Pharmacokinetics and Biopharmaceutics, vol.7, issue.3, pp.303-309, 1988.
DOI : 10.2307/2531827

A. Egron, P. Olivier-abbal, and A. Gouraud, Preventable and potentially preventable serious adverse reactions induced by oral protein kinase inhibitors through a database of adverse drug reaction reports, Targeted Oncology, vol.348, issue.2, pp.229-234, 2015.
DOI : 10.1056/NEJMsa020703

M. Yanagisawa, K. Yorozu, and M. Kurasawa, Bevacizumab improves the delivery and efficacy of paclitaxel, Anti-Cancer Drugs, vol.21, pp.687-694, 2010.
DOI : 10.1097/CAD.0b013e32833b7598

K. Ishizuna, J. Ninomiya, and M. Kojima, Paclitaxel-resistant advanced recurrent breast cancer: a case of partial response due to addition of bevacizumab to paclitaxel therapy: a case report, BMC Research Notes, vol.6, issue.1, p.254, 2013.
DOI : 10.1016/j.ejca.2008.12.016

L. Candeil, I. Gourdier, and D. Peyron, ABCG2 overexpression in colon cancer cells resistant to SN38 and in irinotecan-treated metastases, International Journal of Cancer, vol.63, issue.6, pp.848-854, 2004.
DOI : 10.1002/path.1203

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.20032/pdf

K. Arimori, N. Kuroki, and M. Hidaka, Effect of Pglycoprotein modulator, cyclosporin A, on the gastrointestinal excretion of irinotecan and its metabolite SN-38 in rats, Pharmaceutical Research, vol.20, issue.6, pp.910-917, 2003.
DOI : 10.1023/A:1023847521767

L. Bonhomme-faivre, A. Benyamina, and M. Reynaud, Disposition of Delta tetrahydrocannabinol in CF1 mice deficient in mdr1a P-glycoprotein, Addict. Biol, p.13, 2008.
DOI : 10.1111/j.1369-1600.2008.00096.x

J. Kovarick, D. Beyer, and M. Bizot, Pharmacokinetic interaction between verapamil and everolimus healthy subjets, Br. J. Clin. Pharmacol, p.60, 2005.

S. Linn and G. Giaccone, MDR1/P-glycoprotein expression in colorectal cancer, European Journal of Cancer, vol.31, issue.7-8, pp.31-1291, 1995.
DOI : 10.1016/0959-8049(95)00278-Q

Y. Tokunaga, H. Hosogi, and T. Hoppou, Effects of MDR1/P-glycoprotein expression on prognosis in advanced colorectal cancer after surgery, Oncology Reports, vol.8, pp.815-819, 2001.
DOI : 10.3892/or.8.4.815

L. Goldwirt, K. Beccaria, and A. Carprntier, Preclinical impact of bevacizumab on brain and tumor distribution of irinotecan and temozolomide, Journal of Neuro-Oncology, vol.19, issue.8, pp.273-281, 2015.
DOI : 10.1158/1078-0432.CCR-12-3105

B. Ellingson, T. Cloughesy, and A. Lai, Quantification of edema reduction using differential quantitative T2 (DQT2) relaxometry mapping in recurrent glioblastoma treated with bevacizumab, Journal of Neuro-Oncology, vol.22, issue.9, pp.111-119, 2012.
DOI : 10.1007/s11060-010-0293-7

URL : https://cloudfront.escholarship.org/dist/prd/content/qt4sm994b8/qt4sm994b8.pdf

M. Chamberlain, Bevacizumab for the Treatment of Recurrent Glioblastoma, Clinical Medicine Insights: Oncology, vol.5, pp.117-129, 2011.
DOI : 10.4137/CMO.S7232

URL : http://doi.org/10.4137/cmo.s7232

C. Vangestel, C. Van-de-wiele, and N. Van-damme, 99mTc-(CO)3 His-Annexin A5 Micro-SPECT Demonstrates Increased Cell Death by Irinotecan During the Vascular Normalization Window Caused by Bevacizumab, Journal of Nuclear Medicine, vol.52, issue.11, pp.1786-1794, 2011.
DOI : 10.2967/jnumed.111.092650

URL : http://jnm.snmjournals.org/content/52/11/1786.full.pdf

I. Genentech, Package insert for avastin (bevacizumab), 2007.

J. Gaudreault, V. Shiu, and A. Bricarello, Concomitant Administration of Bevacizumab, Irinotecan, 5-Fluorouracil, and Leucovorin: Nonclinical Safety and Pharmacokinetics, International Journal of Toxicology, vol.37, issue.8, pp.357-363, 2005.
DOI : 10.2165/00003495-199652040-00013

N. Qi and F. Li, Xiaosong Lit al. Combination use of paclitaxel and avastinenhances treatment effect for the NSCLC patients with malignant pleural effusion, Medicine, vol.95, p.47, 2016.

J. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, R. J. Zeuzen et al., Sorafenib improves survival in advanced hepatocellular carcinoma, (HCC): results of a phase III randomized placebo-controlled trial (SHARP trial), J Clin Oncol, vol.18, issue.25, 2007.

, NEXAVAR: EPAR -Scientific Discussion, European Medicines Agency (EMEA), 2007.

, Monograph of sorafenib, 2017.

P. Boudou-rouquetre, S. Ropert, O. Mir, R. Coriat, B. Billemont et al., Variability of Sorafenib Toxicity and Exposure over Time: A Pharmacokinetic/Pharmacodynamic Analysis, The Oncologist, vol.17, issue.9, pp.1204-1212, 2012.
DOI : 10.1634/theoncologist.2011-0439

B. Gao, S. Yeap, A. Clements, B. Balakrishnar, M. Wong et al., Evidence for Therapeutic Drug Monitoring of Targeted Anticancer Therapies, Journal of Clinical Oncology, vol.30, issue.32, pp.4017-4042, 2012.
DOI : 10.1200/JCO.2012.43.5362

D. Marco, V. , D. Vita, F. Koskinas, J. Semela et al., Sorafenib: from literature to clinical practice, Annals of Oncology, vol.57, issue.10, pp.30-37
DOI : 10.1016/j.jhep.2012.02.016

M. Tandia, A. Mhiri, B. Paule, R. Saffroy, V. Cailliez et al., Correlation between clinical response to sorafenib in hepatocellular carcinoma treatment and polymorphisms of Pglycoprotein (ABCB1) and of breast cancer resistance protein (ABCG2): monocentric study, Cancer Chemother Pharmacol, vol.79, issue.4, pp.2017759-766

S. Jurjen, R. A. Lagas, R. W. Van-waterschoot, J. H. Sparidans, A. H. Beijnen et al., Breast Cancer Resistance Protein and P-glycoprotein Limit Sorafenib Brain Accumulation, Mol Cancer Ther, pp.319-326, 20109-02.

J. Dong, B. Zhai, W. Sun, F. Hu, H. Cheng et al., Activation of phosphatidylinositol 3-kinase/AKT/snail signaling pathway contributes to epithelial-mesenchymal transition-induced multi-drug resistance to sorafenib in hepatocellular carcinoma cells, PLOS ONE, vol.10, issue.9, p.185088, 2017.
DOI : 10.1371/journal.pone.0185088.t002

M. Giavazzi-;-carmen-ghilardi, Contribution of tumor endothelial cells to drug resistance: anti-angiogenic tyrosine kinase inhibitors act as p-glycoprotein antagonists, Angiogenesis, vol.20, pp.233-241, 2017.

S. Ambudkar, S. Dey, and C. Hrycyna, BIOCHEMICAL, CELLULAR, AND PHARMACOLOGICAL ASPECTS OF THE MULTIDRUG TRANSPORTER, Annual Review of Pharmacology and Toxicology, vol.39, issue.1, pp.361-398, 1999.
DOI : 10.1146/annurev.pharmtox.39.1.361

B. M. Ellingson, T. F. Cloughesy, and A. Lai, Quantification of edema reduction using differential quantitative T2 (DQT2) relaxometry mapping in recurrent glioblastoma treated with bevacizumab, Journal of Neuro-Oncology, vol.22, issue.9
DOI : 10.1007/s11060-010-0293-7

, J. Neurooncol, vol.106, pp.111-119, 2012.

H. Jang, J. H. Bum-jun-kim, H. S. Kim, and . Kim, The addition of bevacizumab in the firstline treatment for metastatic colorectal cancer: an updated meta-analysis of randomized trials, Oncotarget, vol.8, issue.42, pp.73009-73016, 2017.

J. M. Hubbard, G. Kim, M. J. Borad, E. Johnson, R. Qin et al., Phase I trial of FOLFIRI in combination with sorafenib and bevacizumab in patients with advanced gastrointestinal malignancies, Investigational New Drugs, vol.31, issue.5, pp.96-103, 2016.
DOI : 10.1007/s10637-013-9976-1

V. Philippe, T. Zahel, K. Bärtl, S. Rasch, O. Ebert et al., Influence of Sorafenib and Bevacizumab on pancreatic volume ? A monocentric CT based analysis. Pancreatology, pp.621-624, 2016.

, Alliance for clinical trials in oncology Sorafenib and bevacizumab in treating patients with metastatic colorectal cancer. Clinicaltrials.gouv, 2017.

C. Chu, C. Abbara, and M. Tandia, Cetuximab increases concentrations of irinotecan and of its active metabolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice, Fundamental & Clinical Pharmacology, vol.31, issue.6, pp.652-660, 2014.
DOI : 10.1124/dmd.31.1.21

C. Chu, M. S. Noël-hudson, J. Bénard, T. Ha-duong, F. Allaoui et al., Cetuximab directly inhibits P-glycoprotein function in vitro independently of EGFR binding, European Journal of Pharmaceutical Sciences, vol.76, pp.18-26, 2015.
DOI : 10.1016/j.ejps.2015.04.014

J. Bénard, D. Silva, J. , D. Blois, and M. , Characterization of a human ovarian adenocarcinoma line, IGROV1, in tissue culture and in nude mice, Cancer Res, vol.45, pp.4970-4979, 1985.

C. Jovelet, J. Bénard, and F. Forestier, Inhibition of P-glycoprotein functionality by vandetanib may reverse cancer cell resistance to doxorubicin, European Journal of Pharmaceutical Sciences, vol.46, issue.5, pp.484-491, 2012.
DOI : 10.1016/j.ejps.2012.03.012

B. Blanchet, B. Billemont, J. Cramard, A. Benichou, S. Chhun et al., Validation of an HPLC-UV method for sorafenib determination in human plasma and application to cancer patients in routine clinical practice, Journal of Pharmaceutical and Biomedical Analysis, vol.49, issue.4, pp.1109-1123, 2009.
DOI : 10.1016/j.jpba.2009.02.008

A. Bailer, Testing for the equality of area under the curves when using destructive measurement techniques, Journal of Pharmacokinetics and Biopharmaceutics, vol.7, issue.3, pp.303-309, 1988.
DOI : 10.2307/2531827

A. Schinkel, U. Mayer, E. Wagenaar, C. Mol, L. Van-deemter et al., Normal viability and altered pharmacokinetics in mice lacking mdr1-type (drug-transporting) P-glycoproteins, Proceedings of the National Academy of Sciences, vol.183, issue.4, pp.4028-4061, 1997.
DOI : 10.1084/jem.183.4.1797

URL : http://www.pnas.org/content/94/8/4028.full.pdf

M. Yu, A. Ocana, and I. Tannock, Reversal of ATP-binding cassette drug transporter activity to modulate chemoresistance: why has it failed to provide clinical benefit?, Cancer and Metastasis Reviews, vol.31, issue.1-2, pp.211-238, 2013.
DOI : 10.1016/j.biomaterials.2009.09.048

Y. Huang, Z. Xue, and H. Zhang, Sorafenib reverses resistance of gastric cancer to treatment by cisplatin through down-regulating MDR1 expression, Medical Oncology, vol.47, issue.10, pp.470-480, 2015.
DOI : 10.1016/j.ejca.2011.04.006

L. Wei, N. Huang, and Y. L. , Sorafenib reverses multidrug resistance of hepatoma cells in vitro, pp.1016-1025, 1023.

Q. Ye and H. Chen, Bevacizumab in the treatment of ovarian cancer: a meta-analysis from four phase III randomized controlled trials, Archives of Gynecology and Obstetrics, vol.66, issue.11, pp.655-666, 2013.
DOI : 10.2146/ajhp080455

D. Pawaskar, R. Straubinger, G. Fetterly, B. Hylander, E. Repasky et al., Physiologically based pharmacokinetic models for everolimus and sorafenib in mice, Cancer Chemotherapy and Pharmacology, vol.15, issue.5, 2013.
DOI : 10.1023/A:1011940108365

URL : http://europepmc.org/articles/pmc3755750?pdf=render

H. Saber-mahloogi and D. , Pharmacology review-Sorafenib. Center for Drug Evaluation and Research, NDA, vol.21, pp.923-175, 2005.

, Les cancers en France en 2016 l'essentiel des faits et chiffres, Institut national du cancer

M. Bidart, F. Berger, and L. Pelletier, Les thérapies antiangiogéniques : de la théorie à la pratique, Ann. Biol Clin, vol.71, issue.5, pp.2527-2562, 2013.

, Angiogenesis insights from a systematic overview New York: Nova Science, 2013.

I. Ruiz and C. Féray, Prise en charge du carcinoma hépatocéllulaire

, Cancer/Radiothérapie, issue.19, pp.410-415, 2015.

A. Méjean and T. , Lebret ; la cascade métastatique : angiogenèse et nouveaux concepts, Progrès en Urologie, pp.156-166, 2008.

J. Folkman, Tumor angiogenesis : therapeutic implications, N Engl J Med, vol.285, pp.1182-1188, 197118.

J. Folkman and D. Hanahan, Switch to the angiogenic phenotype during tumorigenesis, Princess Takanatsu Symp, vol.22, pp.339-386, 1991.

M. Jary, C. Borg, O. Bouché, S. Kim, T. André et al., Traitements antiangiogéniques dans le cancer colorectal métastatique : peut-on envisager un blocage continu de l'angiogenèse ? Bulletin du cancer, pp.758-771, 2015.

L. Treps and J. Gavard, L'angiogenèse tumorale quand l'arbre de vie tourne mal. médecine, sciences, vol.31, pp.989-95, 2015.
DOI : 10.1051/medsci/20153111013

URL : http://www.medecinesciences.org/articles/medsci/pdf/2015/12/medsci20153111p989.pdf

J. Holash, P. Maisonpierre, D. Compton, P. Boland, C. Alexander et al., Vessel Cooption, Regression, and Growth in Tumors Mediated by Angiopoietins and VEGF, Science, vol.284, issue.5422, pp.1994-2002, 1999.
DOI : 10.1126/science.284.5422.1994

P. Carmeliet and R. Jain, Angiogenesis in cancer and other diseases, Nature, vol.407, issue.6801, pp.249-57, 2000.
DOI : 10.1038/35025220

S. Eccles and D. Welch, Metastasis: recent discoveries and novel treatment strategies, The Lancet, vol.369, issue.9574, pp.1742-57, 2007.
DOI : 10.1016/S0140-6736(07)60781-8

URL : http://europepmc.org/articles/pmc2214903?pdf=render

D. Fukumura, R. Xavier, T. Sugiura, Y. Chen, E. Park et al., Tumor Induction of VEGF Promoter Activity in Stromal Cells, Cell, vol.94, issue.6, pp.715-740, 1998.
DOI : 10.1016/S0092-8674(00)81731-6

URL : https://doi.org/10.1016/s0092-8674(00)81731-6

. Carmeliet, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.464, issue.7347
DOI : 10.1038/nature08889

URL : http://europepmc.org/articles/pmc4049445?pdf=render

, Nature, issue.7347, pp.473-298, 2011.

A. Byrme, Angiogenic and cell survival functions of Vascular Endothelial Growth Factor (VEGF), Journal of Cellular and Molecular Medicine, vol.8, issue.4, pp.777-94, 2005.
DOI : 10.1073/pnas.2135406100

N. Ferrara, VEGF-A : a critical regulator of blood vessel growth.Eur Cytokine Netw, pp.158-63, 2009.

A. Bikfalvi, Angiogenèse tumorale. Bull Cabcer, pp.94-193, 2007.

R. Coquard and I. , Traitements anti VEGF : un employ universel ? Bull Cancer, pp.191-197, 2007.

M. Terme, Modulation of immunity by Antiangiogenic Molecules in Cancer. Clinical and Development immunology, AZrticle, vol.492920, 2012.

T. Voron, T cells in tumors, The Journal of Experimental Medicine, vol.212, issue.2, pp.139-148
DOI : 10.1002/ijc.26094

E. Tartour, Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy, Cancer and Metastasis Reviews, vol.8, issue.Suppl 10, pp.83-95, 2011.
DOI : 10.1158/1535-7163.MCT-08-1051

V. Th, Control of the immine response by pro-angiogenic factors. Front Oncol, pp.1-9, 2014.

J. Ghiringhelli, Tumor cells convert immature myeloid dendritic cells into TGFbeta-secreting cells inducing CD4+ CD25+ regulatory T cell proliferation, pp.919-929, 2005.

T. Voron, Rôle du VEGF dans l'épuisement des lymphocytes T intratumoraux. m/s. 2015, pp.473-478
DOI : 10.1051/medsci/20153105004

URL : https://www.medecinesciences.org/articles/medsci/pdf/2015/06/medsci20153105p473.pdf

R. Shrimali, Antiangiogenic Agents Can Increase Lymphocyte Infiltration into Tumor and Enhance the Effectiveness of Adoptive Immunotherapy of Cancer, Cancer Research, vol.70, issue.15
DOI : 10.1158/0008-5472.CAN-10-0153

, Cancer Res, vol.70, issue.15, pp.6171-80, 2010.

Y. Huang, Vascular Normalization as an Emerging Strategy to Enhance Cancer Immunotherapy, Cancer Research, vol.73, issue.10, pp.2943-2991
DOI : 10.1158/0008-5472.CAN-12-4354

. Bouzin, Effects of Vascular Endothelial Growth Factor on the Lymphocyte-Endothelium Interactions: Identification of Caveolin-1 and Nitric Oxide as Control Points of Endothelial Cell Anergy, The Journal of Immunology, vol.178, issue.3, pp.1505-1511, 2007.
DOI : 10.4049/jimmunol.178.3.1505

M. Bidart, F. Berger, and L. Pelletier, Les thérapies antiangiogéniques : de la théorie à la pratique, Ann. Biol Clin, vol.71, issue.5, 2013.

, Page, vol.178

P. Marquet, P. Longeray, and F. Barlesi, Recherche translationnelle : médecine personnalisée, médecine de précision, thérapies ciblées : marketing ou science ? Thérapie 2015 janvier-février, pp.1-10

R. Jain, Normalisation of tumor vasculature: an emerging concept in antiangiogenic therapy, Science, vol.3, pp.391-400, 2005.

Y. Gu, H. Lu, C. Boisson-vidal, H. Li, G. Bousquet et al., La résistance aux traitements antiangiogéniques Une actualité clinique et scientifique. médecine, pp.370-377, 2016.

S. Oudard, J. Médioni, and J. Ayllon, Surveillance d'un traitement antiangiogénique, Rev Prat Med Gen, vol.23, pp.685-692, 2009.

S. Giuliano and G. Pages, Mechanisms of resistance to anti-angiogenesis therapies, Biochimie, vol.95, issue.6, pp.1110-1119, 2013.
DOI : 10.1016/j.biochi.2013.03.002

L. Huang, C. Perrault, and J. Coelho-martins, Induction of acquired drug resistance in endothelial cells and its involvement in anticancer therapy, Journal of Hematology & Oncology, vol.6, issue.1, p.49, 2013.
DOI : 10.1158/1078-0432.CCR-09-0902

URL : https://hal.archives-ouvertes.fr/inserm-00845842

L. Huang, C. Hu, D. Benedetto, and M. , Cross-drug resistance to sunitinib induced by doxorubicin in endothelial cells, Oncology Letters, vol.9, issue.3, pp.1287-1292, 2015.
DOI : 10.3892/ol.2014.2819

L. Huang, L. Huang, and C. Hu, 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.

K. Hida, K. Akiyama, and N. Ohga, Tumour endothelial cells acquire drug resistance in a tumour microenvironment, Journal of Biochemistry, vol.307, issue.5706, pp.243-249, 2013.
DOI : 10.1126/science.1104819

URL : https://academic.oup.com/jb/article-pdf/153/3/243/2437236/mvs152.pdf

B. Schneider, M. Wang, and M. Radovich, Association of Vascular Endothelial Growth Factor and Vascular Endothelial Growth Factor Receptor-2 Genetic Polymorphisms With Outcome in a Trial of Paclitaxel Compared With Paclitaxel Plus Bevacizumab in Advanced Breast Cancer: ECOG 2100, Journal of Clinical Oncology, vol.26, issue.28, pp.4672-4678, 2008.
DOI : 10.1200/JCO.2008.16.1612

L. Benet and B. Hoener, Changes in plasma protein binding have little clinical relevance, Clinical Pharmacology & Therapeutics, vol.71, issue.3, pp.115-121, 2002.
DOI : 10.1067/mcp.2002.121829

J. Lin, assessment, Expert Opinion on Drug Metabolism & Toxicology, vol.50, issue.1, pp.81-92, 2007.
DOI : 10.1074/jbc.M007794200

URL : https://hal.archives-ouvertes.fr/hal-01648954

P. Beaune and M. Loriot, Bases mol??culaires de la susceptibilit?? aux x??nobiotiques : aspects m??taboliques., m??decine/sciences, vol.16, issue.10, pp.1051-1057, 2000.
DOI : 10.4267/10608/1524

V. , A. Aegerter, and V. Von-gunten, Institut Central (ICHV)

. Glycoprotéine, une pompe d'efflux : attention aux interactions Médicamenteuses

, Caduceus Express. Sept, vol.13, issue.6, 2011.

U. Meyer, Genotype or phenotype: the definition of a pharmacogenetic polymorphism, Pharmacogenetics, vol.1, issue.2, pp.66-73, 1991.
DOI : 10.1097/00008571-199111000-00002

, Page, vol.180

M. Ingelman-sundberg, M. Oscarson, and R. Mclellan, Polymorphic human cytochrome P450 enzymes: an opportunity for individualized drug treatment, Trends in Pharmacological Sciences, vol.20, issue.8, pp.342-351, 1999.
DOI : 10.1016/S0165-6147(99)01363-2

J. Blanc, J. Barbare, V. Boige, K. Boudjema, G. Créhange et al., , 2015.

. Invs, Projection de l'incidence et de la mortalité par cancer en France métropolitaine en 2015

M. Schwartz, S. Roayaie, and M. Konstadoulakis, Strategies for the management of hepatocellular carcinoma, Nature Clinical Practice Oncology, vol.15, issue.7, pp.424-432, 2007.
DOI : 10.3748/wjg.v12.i19.3114

J. Bruix, M. Sherman, and J. Llovet, Clinical Management of Hepatocellular Carcinoma. Conclusions of the Barcelona-2000 EASL Conference, Journal of Hepatology, vol.35, issue.3, pp.421-430, 2001.
DOI : 10.1016/S0168-8278(01)00130-1

J. Bruix and M. Sherman, Management of hepatocellular carcinoma, Hepatology, vol.35, issue.5, pp.1208-1236, 2005.
DOI : 10.1016/0016-5085(88)90436-2

C. Lo, H. Ngan, and W. Tso, Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma, Hepatology, vol.12, issue.5, pp.1164-1171, 2002.
DOI : 10.1002/hep.1840120422

P. Hilgard, M. Hamami, and A. Fouly, Radioembolization with yttrium-90 glass microspheres in hepatocellular carcinoma: European experience on safety and long-term survival, Hepatology, vol.74, issue.5, pp.1741-1749, 2010.
DOI : 10.1016/j.ijrobp.2008.10.005

E. Thompson, D. Newgreen, and D. Tarin, Carcinoma Invasion and Metastasis: A Role for Epithelial-Mesenchymal Transition?, Cancer Research, vol.65, issue.14, pp.5991-95, 2005.
DOI : 10.1158/0008-5472.CAN-05-0616

J. Xu, R. Wang, and Z. Xie, Prostate cancer metastasis: Role of the host microenvironment in promoting epithelial to mesenchymal transition and increased bone and adrenal gland metastasis, The Prostate, vol.357, issue.15, pp.1664-73, 2006.
DOI : 10.1002/pros.20488

L. Larue and A. Bellacosa, Epithelial???mesenchymal transition in development and cancer: role of phosphatidylinositol 3??? kinase/AKT pathways, Oncogene, vol.24, issue.50, pp.7443-54, 2005.
DOI : 10.1074/jbc.M308168200

J. Thiery and J. Sleeman, Complex networks orchestrate epithelial???mesenchymal transitions, Nature Reviews Molecular Cell Biology, vol.117, issue.2, pp.131-173, 2006.
DOI : 10.1016/j.cell.2004.06.006

, Carcinome hépatocellulaire : dépistage, diagnostic et traitement

O. Digestive, Hépato-Gastro & Oncologie Digestive, pp.46-64, 2016.

I. Ruiz and C. Feray, Prise ne charge du carcinome hépatocellulaire

, Cancer/Radiothérapie, vol.19, pp.410-415, 2015.

, ©2017 VIDAL. Version : 2017.05.0. Données du 12, 2017.

J. Phelip, O. Bouché, T. Conroy, P. Michel, C. Penna et al., , 2016.

V. Heinemann, V. Weikersthal, L. Decker, and T. , Randomized comparison of FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment Page 182

, KRAS-wild type metastatic colorectal cancer: German AIO study KRK-0306 (FIRE-3), Lancet Oncol, vol.15, pp.1065-75, 2014.

A. Venook, D. Niedzwiecki, D. , H. Lenz, and H. , Phase III trial of irinotecan/5-FU/leucovorin (FOLFIRI) or

. Fu-/-leucovorin, with bevacizumab (BV) or cetuximab (CET) for patients (pts) with KRAS wild-type (wt) untreated metastatic adenocarcinoma of the colon or rectum (MCRC), J Clin Oncol, vol.32, issue.mFOLFOX6, pp.5-2014

L. Ye, T. Liu, L. Ren, Y. Wei, D. Zhu et al., Wild-Type Unresectable Colorectal Liver-Limited Metastases, Journal of Clinical Oncology, vol.31, issue.16, pp.1931-1939, 2013.
DOI : 10.1200/JCO.2012.44.8308

T. Gruenberger, J. Bridgewater, and I. Chau, Randomized, phase II study of bevacizumab with mFOLFOX6 or FOLFOXIRI in patients with initially unresectable liver metastases from colorectal cancer: resectability and safety in OLIVIA, J Clin Oncol, vol.31, issue.3619, p.2013

M. Seymour, T. Maughan, J. Ledermann, C. Topham, R. James et al., Different strategies of sequential and combination chemotherapy for patients with poor prognosis advanced colorectal cancer (MRC FOCUS): a randomised controlled trial, The Lancet, vol.370, issue.9582, pp.143-52, 2007.
DOI : 10.1016/S0140-6736(07)61087-3

M. Ducreux, D. Malka, J. Mendiboure, P. Etienne, P. Texereau et al., Sequential versus combination chemotherapy for the treatment of advanced colorectal cancer (FFCD 2000???05): an open-label, randomised, phase 3 trial, The Lancet Oncology, vol.12, issue.11
DOI : 10.1016/S1470-2045(11)70199-1

, Lancet Oncol, vol.12, pp.1032-1044, 2011.

M. Koopman, N. Antonini, J. Douma, J. Wals, A. Honkoop et al., Sequential versus combination chemotherapy with capecitabine, p.183

, oxaliplatin in advanced colorectal cancer (CAIRO): a phase III randomised controlled trial, Lancet, vol.370, pp.135-177, 2007.

. Cunningham, Two different first-line 5-fluorouracil regimens with or without oxaliplatin in patients with metastatic colorectal cancer, Annals of Oncology, vol.17, issue.3, pp.244-249, 2009.
DOI : 10.1093/annonc/mdj104

T. Aparicio, S. Lavau-denes, and J. Phelip, Randomized phase III trial in elderly patients comparing LV5FU2 with or without irinotecan for first-line treatment of metastatic colorectal cancer (FFCD 2001???02), Annals of Oncology, vol.15, issue.1, pp.121-128, 2001.
DOI : 10.1056/NEJMoa032691

URL : https://hal.archives-ouvertes.fr/hal-01910082

J. Tabernero, P. Pfeiffer, and A. Cervantes, Administration of Cetuximab Every 2 Weeks in the Treatment of Metastatic Colorectal Cancer: An Effective, More Convenient Alternative to Weekly Administration?, The Oncologist, vol.13, issue.2, pp.113-122, 2008.
DOI : 10.1634/theoncologist.2007-0201

P. Pfeiffer, D. Nielsen, J. Bjerregaard, C. Qvortrup, M. Yilmaz et al., Biweekly cetuximab and irinotecan as third-line therapy in patients with advanced colorectal cancer after failure to irinotecan, oxaliplatin and 5-fluorouracil, Annals of Oncology, vol.19, issue.6, pp.1141-1146, 2008.
DOI : 10.1093/annonc/mdn020

M. Bouchahda, T. Macarulla, G. Liedo, F. Lévi, M. Elez et al., Feasibility of cetuximab given with a simplified schedule every 2??weeks in advanced colorectal cancer: a multicenter, retrospective analysis, Medical Oncology, vol.68, issue.2, 2010.
DOI : 10.1159/000086776

T. Ciuleanu, V. Nikolic, E. Shmueli, D. Vrbanec, S. Plate et al., Cetuximab weekly (q1w) versus every two weeks (q2w) plus FOLFOX4 as first-line therapy in patients (pts) with KRAS wild-type (wt) metastatic colorectal cancer (mCRC), J Clin Oncol, vol.29, issue.3580, p.2011

O. Mir, J. Alexandre, and R. Coriat, Safety of bevacizumab 7.5 mg/kg infusion over 10 minutes in NSCLC patients, Investigational New Drugs, vol.30, issue.5 Suppl 16, pp.1756-60, 2012.
DOI : 10.1053/j.seminoncol.2003.08.013

T. Mahfoud, R. Tanz, and M. Mesmoudi, Bevacizumab 5 or 7.5??mg/kg in Metastatic Colorectal Cancer Can Be Infused Safely Over 10??Minutes, Journal of Gastrointestinal Cancer, vol.11, issue.2, pp.244-252, 2012.
DOI : 10.1023/A:1008309405678

D. Reidy, K. Chung, and J. Timoney, Bevacizumab 5 mg/kg Can Be Infused Safely Over 10 Minutes, Journal of Clinical Oncology, vol.25, issue.19, pp.2691-2696, 2007.
DOI : 10.1200/JCO.2006.09.3351

K. Mcglynn, J. Petrick, and W. London, Global Epidemiology of Hepatocellular Carcinoma, Clinics in Liver Disease, vol.19, issue.2, pp.223-238, 2015.
DOI : 10.1016/j.cld.2015.01.001

E. Maillard and . Epidémiologie, histoire naturelle et pathogenèse du carcinome hépatocellulaire. cancer/radiothérapie, Feb, vol.15, issue.1, pp.3-6, 2011.

H. Yang, S. Yeh, P. Chen, U. Iloeje, C. Jen et al., Associations Between Hepatitis B Virus Genotype and Mutants and the Risk of Hepatocellular Carcinoma, JNCI Journal of the National Cancer Institute, vol.25, issue.9
DOI : 10.1093/carcin/bgh172

URL : https://academic.oup.com/jnci/article-pdf/100/16/1134/7682333/djn243.pdf

, J Natl cancer Inst, vol.100, pp.1134-1143, 2008.

S. Bruno, A. Crasignani, P. Maisonneuve, S. Rossi, E. Sitini et al., Hepatitis C virus genotype 1b as a major risk factor associated with hepatocellular carcinoma in patients with cirrhosis: A seventeen-year prospective cohort study, Hepatology, vol.58, issue.5
DOI : 10.1136/gut.47.1.131

, Hepatology, vol.46, pp.1350-1356, 2007.

G. Fattovich, T. Stroffolini, I. Zagni, and F. Danata, Hepatocellular carcinoma in cirrhosis: Incidence and risk factors, Gastroenterology, vol.127, issue.5, pp.35-50, 2004.
DOI : 10.1053/j.gastro.2004.09.014

A. Haouala, N. Widmer, M. Duchosal, M. Montemurro, T. Buclin et al., Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib, Blood, vol.117, issue.8, pp.75-87, 2011.
DOI : 10.1182/blood-2010-07-294330

J. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, R. J. Zeuzen et al., Sorafénib improves survival in advanced hepatocellular carcinoma, p.185

, phase III randomized placebo-controlled trial (SHARP trial), J Clin Oncol, 2007.

, 18S, vol.25

, NEXAVAR: EPAR -Scientific Discussion, European Medicines Agency (EMEA), 2007.

, 86 Monograph of sorafénib, 2014.

I. Ichiro, Functional Significance of Genetic Polymorphisms in P-glycoprotein (MDR1, ABCB1) and Breast Cancer Resistance Protein (BCRP, ABCG2), Drug Metab Pharmacokinet, vol.27, issue.1, pp.85-105, 2012.

B. Blanchet, B. Billemont, J. Cramard, A. Benichou, S. Chhun et al., Validation of an HPLC-UV method for sorafenib determination in human plasma and application to cancer patients in routine clinical practice, Journal of Pharmaceutical and Biomedical Analysis, vol.49, issue.4, pp.1109-1114, 2009.
DOI : 10.1016/j.jpba.2009.02.008

C. Fucile, S. Marenco, M. Bazzica, M. Zuccoli, F. Lantieri et al., Measurement of sorafenib plasma concentration by high-performance liquid chromatography in patients with advanced hepatocellular carcinoma: is it useful the application in clinical practice? A pilot study, Medical Oncology, vol.38, issue.1, pp.1-7
DOI : 10.1111/j.1346-8138.2010.01059.x

J. Arrondeau, O. Mir, P. Boudou-rouquette, R. Coriat, S. Ropert et al., Sorafenib exposure decreases over time in patients with hepatocellular carcinoma, Investigational New Drugs, vol.3, issue.2, pp.2046-2055, 2012.
DOI : 10.1177/1758834010396117

P. Boudou-rouquette, S. Ropert, O. Mir, R. Coriat, B. Billemont et al., Variability of Sorafenib Toxicity and Exposure over Time: A Pharmacokinetic/Pharmacodynamic Analysis, The Oncologist, vol.17, issue.9, pp.1204-1212, 2012.
DOI : 10.1634/theoncologist.2011-0439

P. Boudou-rouquette, C. Narjoz, J. Golmard, A. Thomas-schoemann, O. Mir et al., Early Sorafenib-Induced Toxicity Is Associated with Drug Exposure and UGTIA9 Genetic Polymorphism in Patients with Solid Tumors: A Preliminary Study, PLoS ONE, vol.7, issue.8, p.42875, 2012.
DOI : 10.1371/journal.pone.0042875.t005

D. Strumberg, J. Clark, A. Awada, M. Moore, H. Richly et al., Safety, Pharmacokinetics, and Preliminary Antitumor Activity of Sorafenib: A Review of Four Phase I Trials in Patients with Advanced Refractory Solid Tumors, The Oncologist, vol.12, issue.4, pp.426-437, 2007.
DOI : 10.1634/theoncologist.12-4-426

L. Jain, S. Woo, E. Gardner, W. Dahut, E. Kohn et al., Population pharmacokinetic analysis of sorafenib in patients with solid tumours, British Journal of Clinical Pharmacology, vol.23, issue.2, pp.294-305, 2011.
DOI : 10.1007/BF02353466

N. Van-erp, H. Gelderblom, and H. Guchelaar, Clinical pharmacokinetics of tyrosine kinase inhibitors, Cancer Treatment Reviews, vol.35, issue.8, pp.692-706, 2009.
DOI : 10.1016/j.ctrv.2009.08.004

V. Baldissera, A. De-mattos, G. Coral, F. De-araujo, C. Marroni et al., Evaluation of the C3435T polymorphism in the MDR1 gene in patients with hepatocellular carcinoma, Ann Hepatol, vol.11, issue.6, pp.899-906, 2012.

M. Fukuda, Y. Kawahara, T. Hirota, S. Akizuki, S. Shigeto et al., Genetic Polymorphisms of Hepatic ABC-Transporter in Patients with Hepatocellular Carcinoma, Journal of Cancer Therapy, vol.01, issue.03, pp.114-123, 2010.
DOI : 10.4236/jct.2010.13019

URL : http://www.scirp.org/journal/PaperDownload.aspx?paperID=2603

J. Gao and D. People, Association of MDR1 gene polymorphisms with the risk of hepatocellular carcinoma in the Chinese Han population, Brazilian Journal of Medical and Biological Research, vol.40, issue.182, pp.311-318, 2013.
DOI : 10.1007/s11033-012-2251-2

M. Timucin, H. Algozlu, S. Ozdemir, and O. Ozdemir, Association Between ABCB1 (MDR1) Gene Polymorphism and Unresponsiveness Combined Therapy in Chronic Hepatitis C virus, Hepatitis Monthly, vol.13, issue.4, pp.7522-187, 2013.
DOI : 10.5812/hepatmon.7522

URL : http://cdn.neoscriber.org/cdn/serve/313ea/a9381a6ba9543fb5ce3932ad4ef3d63a91f6beff/15209-pdf.pdf

S. Bins, A. Lenting, and S. Bouazzaoui, are associated with sorafenib-induced toxicity, Pharmacogenomics, vol.17, issue.14, pp.1483-90, 2016.
DOI : 10.1007/s11095-006-9159-2

T. Velde, E. Vogten, J. Gebbink, and M. , Enhanced antitumour efficacy by combining conventional chemotherapy with angiostatin or endostatin in a liver metastasis model, British Journal of Surgery, vol.13, issue.10, pp.1302-1309, 2002.
DOI : 10.1007/BF00689637

B. Emmink, W. Van-houdt, and R. Vries, Differentiated Human Colorectal Cancer Cells Protect Tumor-Initiating Cells From Irinotecan, Gastroenterology, vol.141, issue.1, pp.269-278, 2011.
DOI : 10.1053/j.gastro.2011.03.052

C. Chu, C. Abbara, and M. Tandia, Cetuximab increases concentrations of irinotecan and of its active metabolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice, Fundamental & Clinical Pharmacology, vol.31, issue.6, pp.652-660, 2014.
DOI : 10.1124/dmd.31.1.21

F. Wang, Y. Chen, and L. Huang, Cetuximab enhanced the efficacy of chemotherapeutic agent in ABCB1/P-glycoprotein-overexpressing cancer cells, Oncotarget, vol.6, issue.38
DOI : 10.18632/oncotarget.5813

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=5813&path%5B%5D=17745

, Oncotarget, vol.638, pp.40850-40865, 2015.

L. Li, A. Jiang, and O. Dong, MDR1/P-gp and VEGF Synergistically Enhance the Invasion of Hep-2 Cells with Multidrug Resistance Induced by Taxol, Annals of Surgical Oncology, vol.8, issue.5, pp.1421-1429, 2009.
DOI : 10.4049/jimmunol.171.8.4340

H. Liang, Y. Zhang, and J. Zhang, Effects of arsenic trioxide on expressions of vascular endothelial growth factor and P-glycoprotein in multidrug resistant leukemia cell line K562/A02, Journal of Chinese Integrative Medicine, vol.5, issue.6, pp.647-650, 2007.
DOI : 10.3736/jcim20070609

, Page, vol.188

A. Broggini-tenzer, A. Sharma, and K. Nytko, Combines treatment strategies for microtubule stabilizing agent-resistant tumors, JNCI J Natl Cancer Inst, vol.107, issue.4, p.504, 2015.

A. Egron, P. Olivier-abbal, and A. Gouraud, Preventable and potentially preventable serious adverse reactions induced by oral protein kinase inhibitors through a database of adverse drug reaction reports, Targeted Oncology, vol.348, issue.2, p.10, 2015.
DOI : 10.1056/NEJMsa020703

M. Yanagisawa, K. Yorozu, and M. Kurasawa, Bevacizumab improves the delivery and efficacy of paclitaxel, Anti-Cancer Drugs, vol.21, pp.687-694, 2010.
DOI : 10.1097/CAD.0b013e32833b7598

K. Ishizuna, J. Ninomiya, and M. Kojima, Paclitaxel-resistant advanced recurrent breast cancer: a case of partial response due to addition of bevacizumab to paclitaxel therapy: a case report, BMC Research Notes, vol.6, issue.1, p.254, 2013.
DOI : 10.1016/j.ejca.2008.12.016

L. Candeil, I. Gourdier, and D. Peyron, ABCG2 overexpression in colon cancer cells resistant to SN38 and in irinotecan-treated metastases, International Journal of Cancer, vol.63, issue.6, pp.848-854, 2004.
DOI : 10.1002/path.1203

K. Arimori, N. Kuroki, and M. Hidaka, Effect of Pglycoprotein modulator, cyclosporin A, on the gastrointestinal excretion of irinotecan and its metabolite SN-38 in rats, Pharmaceutical Research, vol.20, issue.6, pp.910-917, 2003.
DOI : 10.1023/A:1023847521767

L. Bonhomme-faivre, A. Benyamina, and M. Reynaud, Disposition of Delta tetrahydrocannabinol in CF1 mice deficient in mdr1a P-glycoprotein, Addict. Biol, pp.13-189, 2008.
DOI : 10.1111/j.1369-1600.2008.00096.x

J. Kovarick, D. Beyer, and M. Bizot, Pharmacokinetic interaction between verapamil and everolimus healthy subjets, Br. J. Clin. Pharmacol, vol.60, 2005.

S. Linn and G. Giaccone, MDR1/P-glycoprotein expression in colorectal cancer, European Journal of Cancer, vol.31, issue.7-8
DOI : 10.1016/0959-8049(95)00278-Q

, Eur J Cancer, pp.31-1291, 1995.

Y. Tokunaga, H. Hosogi, and T. Hoppou, Effects of MDR1/P-glycoprotein expression on prognosis in advanced colorectal cancer after surgery, Oncology Reports, vol.8, pp.815-819, 2001.
DOI : 10.3892/or.8.4.815

L. Goldwirt, K. Beccaria, and A. Carprntier, Preclinical impact of bevacizumab on brain and tumor distribution of irinotecan and temozolomide, Journal of Neuro-Oncology, vol.19, issue.8, pp.273-281, 2015.
DOI : 10.1158/1078-0432.CCR-12-3105

B. Ellingson, T. Cloughesy, and A. Lai, Quantification of edema reduction using differential quantitative T2 (DQT2) relaxometry mapping in recurrent glioblastoma treated with bevacizumab, Journal of Neuro-Oncology, vol.22, issue.9, pp.111-119, 2012.
DOI : 10.1007/s11060-010-0293-7

M. Chamberlain, Bevacizumab for the Treatment of Recurrent Glioblastoma, Clinical Medicine Insights: Oncology, vol.5
DOI : 10.4137/CMO.S7232

, Clin Med Insights Oncol, vol.5, pp.117-129, 2011.

C. Vangestel, C. Van-de-wiele, and N. Van-damme, 99mTc-(CO)3 His-Annexin A5 Micro-SPECT Demonstrates Increased Cell Death by Irinotecan During the Vascular Normalization Window Caused by Bevacizumab, Journal of Nuclear Medicine, vol.52, issue.11, pp.1786-1794, 2011.
DOI : 10.2967/jnumed.111.092650

URL : http://jnm.snmjournals.org/content/52/11/1786.full.pdf

I. Genentech, Package insert for avastin (bevacizumab), 2007.

J. Gaudreault, V. Shiu, and A. Bricarello, Concomitant Administration of Bevacizumab, Irinotecan, 5-Fluorouracil, and Leucovorin: Nonclinical Safety and Pharmacokinetics, International Journal of Toxicology, vol.37, issue.8, pp.357-363, 2005.
DOI : 10.2165/00003495-199652040-00013

N. Qi and F. Li, Xiaosong Lit al. Combination use of paclitaxel and avastinenhances treatment effect for the NSCLC patients with malignant pleural effusion, Medicine, vol.95, p.47, 2016.

B. Gao, S. Yeap, A. Clements, B. Balakrishnar, M. Wong et al., Evidence for Therapeutic Drug Monitoring of Targeted Anticancer Therapies, Journal of Clinical Oncology, vol.30, issue.32, 2012.
DOI : 10.1200/JCO.2012.43.5362

, Nov, vol.10, issue.3032, pp.4017-4042

D. Marco, V. , D. Vita, F. Koskinas, J. Semela et al.,

, Sorafénib: from literature to clinical practice, Ann Oncol. 2013, vol.24, issue.2, pp.30-37

M. Tandia, A. Mhiri, B. Paule, R. Saffroy, V. Cailliez et al.,

. Bonhomme-faivre, Correlation between clinical response to sorafénib in hepatocellular carcinoma treatment and polymorphisms of Pglycoprotein (ABCB1) and of breast cancer resistance protein (ABCG2): monocentric study, Cancer Chemother Pharmacol. 2017, vol.79, issue.4, pp.759-766

S. Jurjen, R. A. Lagas, R. W. Van-waterschoot, J. H. Sparidans, A. H. Beijnen et al., Breast Cancer Resistance Protein and Pglycoprotein Limit Sorafénib Brain Accumulation. Mol Cancer Ther, 2010.

J. Dong, B. Zhai, W. Sun, F. Hu, H. Cheng et al., Activation of phosphatidylinositol 3-kinase/AKT/snail signaling pathway contributes to epithelial-mesenchymal Page 191
DOI : 10.1371/journal.pone.0185088

URL : http://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0185088&type=printable

, transition-induced multi-drug resistance to sorafénib in hepatocellular carcinoma cells, PLoS ONE, vol.12, issue.9, p.185088, 2017.

M. Giavazzi-;-carmen-ghilardi, Contribution of tumor endothelial cells to drug resistance: anti-angiogenic tyrosine kinase inhibitors act as p-glycoprotein antagonists, Angiogenesis, vol.20, pp.233-241, 2017.

S. Ambudkar, S. Dey, and C. Hrycyna, BIOCHEMICAL, CELLULAR, AND PHARMACOLOGICAL ASPECTS OF THE MULTIDRUG TRANSPORTER, Annual Review of Pharmacology and Toxicology, vol.39, issue.1, pp.361-398, 1999.
DOI : 10.1146/annurev.pharmtox.39.1.361

B. M. Ellingson, T. F. Cloughesy, and A. Lai, Quantification of edema reduction using differential quantitative T2 (DQT2) relaxometry mapping in recurrent glioblastoma treated with bevacizumab, Journal of Neuro-Oncology, vol.22, issue.9, pp.111-119, 2012.
DOI : 10.1007/s11060-010-0293-7

H. Jang, J. H. Bum-jun-kim, H. S. Kim, and . Kim, The addition of bevacizumab in the first-line treatment for metastatic colorectal cancer: an updated meta-analysis of randomized trials, Oncotarget, vol.8, issue.42, pp.73009-73016, 2017.
DOI : 10.18632/oncotarget.20314

J. M. Hubbard, G. Kim, M. J. Borad, E. Johnson, R. Qin et al., Phase I trial of FOLFIRI in combination with sorafenib and bevacizumab in patients with advanced gastrointestinal malignancies, Investigational New Drugs, vol.31, issue.5, pp.96-103, 2016.
DOI : 10.1007/s10637-013-9976-1

V. Philippe, T. Zahel, K. Bärtl, S. Rasch, O. Ebert et al., Influence of Sorafénib and Bevacizumab on pancreatic volume ? A monocentric CT based analysis. Pancreatology, pp.621-624, 2016.

, Alliance for clinical trials in oncology Sorafénib and bevacizumab in treating patients with metastatic colorectal cancer. Clinicaltrials.gouv, 2017.

C. Chu, M. S. Noël-hudson, J. Bénard, T. Ha-duong, F. Allaoui et al.,

. Bonhomme-faivre, Cetuximab directly inhibits P-glycoprotein function in vitro independently of EGFR binding, European Journal of Pharmaceutical Sciences, vol.76, pp.18-26, 2015.

A. Schinkel, U. Mayer, E. Wagenaar, C. Mol, L. Van-deemter et al., Normal viability and altered pharmacokinetics in mice lacking mdr1-type (drug-transporting) P-glycoproteins, Proceedings of the National Academy of Sciences, vol.183, issue.4, pp.4028-4061, 1997.
DOI : 10.1084/jem.183.4.1797

URL : http://www.pnas.org/content/94/8/4028.full.pdf

M. Yu, A. Ocana, and I. Tannock, Reversal of ATP-binding cassette drug transporter activity to modulate chemoresistance: why has it failed to provide clinical benefit?, Cancer and Metastasis Reviews, vol.31, issue.1-2, pp.211-238, 2013.
DOI : 10.1016/j.biomaterials.2009.09.048

Y. Huang, Z. Xue, and H. Zhang, Sorafenib reverses resistance of gastric cancer to treatment by cisplatin through down-regulating MDR1 expression, Medical Oncology, vol.47, issue.10, pp.470-480, 2015.
DOI : 10.1016/j.ejca.2011.04.006

L. Wei, N. Huang, and Y. L. , Sorafénib reverses multidrug resistance of hepatoma cells in vitro, pp.1016-1025, 1023.

Q. Ye and H. Chen, Bevacizumab in the treatment of ovarian cancer: a meta-analysis from four phase III randomized controlled trials, Archives of Gynecology and Obstetrics, vol.66, issue.11, pp.655-666, 2013.
DOI : 10.2146/ajhp080455

D. Pawaskar, R. Straubinger, G. Fetterly, B. Hylander, E. Repasky et al., Physiologically based pharmacokinetic models for everolimus and sorafenib in mice, Cancer Chemotherapy and Pharmacology, vol.15, issue.5, pp.1219-1248, 2013.
DOI : 10.1023/A:1011940108365

URL : http://europepmc.org/articles/pmc3755750?pdf=render

H. Saber-mahloogi and D. , Pharmacology review-Sorafénib. Center for Drug Evaluation and Research, NDA, vol.21923, pp.194-195, 2005.

P. C. Chu, C. Abbara, and M. Tandia, Cetuximab increases concentrations of irinotecan and of its active metabolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice, Fundamental & Clinical Pharmacology, vol.31, issue.6, pp.652-660, 2014.
DOI : 10.1124/dmd.31.1.21

M. Tandia, A. Mhiri, B. Paule, R. Saffroy, V. Cailliez et al.,

. Bonhomme-faivre, Correlation between clinical response to sorafenib in hepatocellular carcinoma treatment and polymorphisms of Pglycoprotein (ABCB1) and of breast cancer resistance protein (ABCG2): monocentric study, Cancer Chemother Pharmacol, vol.79, issue.4, pp.759-766, 2017.

M. Tandia, J. M. Alili, C. Abbara, M. S. Noel-hudson, F. Ayadi et al.,

. Bonhomme-faivre, Bevacizumab modulates P-glycoprotein function in vitro and increases concentrations of irinotecan and of its active metabolite SN-38 in plasma of human colorectal carcinoma-bearing mice, Research & Reviews in Pharmacy &Pharmaceutical Sciences Journal RRJPPS |, vol.6, issue.4, 2017.

M. Tandia, C. Abbara, M. S. Noel-hudson, D. Amor, M. Polrot et al.,

. Bonhomme-faivre, Bevacizumab and Sorafenib modulate P-glycoprotein function in vitro and Bevacizumab increases in vivo Sorafenib concentrations in plasma of human colorectal carcinoma-bearing mice, p.196, 2017.

, POSTERS

M. Tandia, C. Abbara, M. S. Noel-hudson, D. Amor, M. Polrot et al.,

. Bonhomme-faivre, Bevacizumab inhibits P-glycoprotein function in vitro and increases in vivo sorafenib concentrations in plasma of human colorectal carcinomabearing mice. Congrès Frontiers Immunology Research Network, pp.1-4

M. Tandia, A. Mhiri, B. Paule, R. Saffroy, V. Delvart et al.,

. Bonhomme-faivre,

, Influence of P-glycoprotein (ABCB1) and of Breast Cancer Resistance Protein (ABCG2) polymorphism on therapeutic monitoring of sorafenib in hepatocellular carcinoma treatment Journée de l'école doctorale Innovation Thérapeutique du fondamental à l'appliqué, p.2015