• Aucun résultat trouvé

[13] Site internet de l’ANSM : http://ansm.sante.fr/ - Dossier ATU nominatives - Spécialités autorisées entre le 01/10/2017 et le 31/10/2017 (16/11/2017) application/vnd.ms-excel [14] Site internet de l’ANSM. Definition de l’Autorisation Temporaire d’Utilisation (ATU) [15] Site internet de l’Institut National du Cancer (l’INCa): http://www.e-cancer.fr/ Rapport sur l’utilisation des anticancéreux sous ATU.

182

[16] Site internet de l’Organisation Mondial de la Santé – Le Cancer dans le Monde en 2015 http://www.who.int/fr/

[17]Notice du médicament Bevacizumab sur le site de l'Agence Européenne du Médicament (l’EMA)

[18] Rapport de l’HAS - Des progrès thérapeutiques pour certains cancers métastatiques de la prostate - Jevtana®, Xtandi® et Zytiga ®

[19] Classification of anticancer drugs--a new system based on therapeutic targets. Cancer Treat Rev. 2003 Dec ;29(6):515-23 Espinosa E et al.

[20] Classification des anticancéreux selon le Collège National de Pharmacologie Médicale (CNPM) http://pharmacomedicale.org/medicaments/par-specialites/item/anticancereux-les- points-essentiels?

[21] Neal JW et al. Decade in review-targeted therapy: successes, toxicities and challenges in solid tumours. Nat Rev Clin Oncol. 2014 Nov; 11(11):627-8. doi: 10.1038/nrclinonc.2014.171. Epub 2014

[22] Jean-Francois Morère, Francoise Mornex, Denis Soulières, Thérapeutique du Cancer – 2eme edition, Edition Springer

[23] Karen Leroy, Patricia de Cremoux, Anomalie moléculaires des cancers. Ciblage thérapeutique. Edition John Libbey Eurotext

[24] Jean-Charles Soria, Stephane Vignot, Christophe Massard, Olivier Mir Cours de chimiothérapie antitumorale et traitement médical du cancer XXVIIIème cours de Gustave- Roussy

[25] Stéphane Vignot, Jean-Charles Soria Thérapies moléculaires ciblées Edition John Libbey Eurotext. Mai 2008

[26] Gaetan des Guetz et Jean-Yvezs Blay Les therapies ciblées

[27] Cornez N, Breast cancer and Herceptin. Bull Cancer. 2000 Nov;87(11):847-58.

[28] Leyland-Jones B, Trastuzumab: hopes and realities Lancet Oncol. 2002 Mar;3(3):137-44.

[29] Slamon DJ et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987 Jan 9; 235(4785):177-82.

[30] Mitchell MS et al. The role of immunohistochemistry and fluorescence in situ hybridization for HER2/neu in assessing the prognosis of breast cancer. Semin Oncol. 1999 Aug; 26(4 Suppl 12):108-16.

[31] Révillion F. ERBB2 oncogene in human breast cancer and its clinical significance. Eur J Cancer. 1998 May; 34(6) :791-808.

[32] Carlomagno C et al. c-erb B2 overexpression decreases the benefit of adjuvant tamoxifen in early-stage breast cancer without axillary lymph node metastases. J Clin Oncol. 1996 Oct; 14(10):2702-8

[33] Elledge RM et al. HER-2 expression and response to tamoxifen in estrogen receptor- positive breast cancer: a Southwest Oncology Group Study. Clin Cancer Res. 1998 Jan; 4(1):7- 12

183

[34] Houston SJ et al. Overexpression of c-erbB2 is an independent marker of resistance to endocrine therapy in advanced breast cancer. Br J Cancer. 1999 Mar; 79(7-8):1220-6.

[35] Pegram MD et al. The effect of HER-2/neu overexpression on chemotherapeutic drug sensitivity in human breast and ovarian cancer cells Oncogene. 1997 Jul 31; 15(5):537-47 [36] Slamon DJet al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001 Mar 15; 344(11):783- 92.

[37] Von Minckwitz et al. Trastuzumab beyond progression in human epidermal growth factor receptor 2-positive advanced breast cancer: a german breast group 26/breast international group 03-05 study. J Clin Oncol. 2009 Apr 20; 27(12):1999-2006. doi:10.1200/JCO.2008.19.6618. Epub 2009 Mar 16.

[38] Site internet de l’INCa – Prise en charge thérapeutique du cancer su sein http://www.e- cancer.fr/Patients-et-proches/Les-cancers/Cancer-du-sein/Traitements

[39] Eisenberg BL (2003) Imatinib mesylate: a molecularly targeted therapy for gastro intestinal stromal tumors. Oncology 11:1-6

[40] Duensig A et al. (2004) Biology of gasdtrointestinal stromal tumors: KIT mutations and beyond. Cancer Inveszt 22: 106-16

[41] Mahon FX et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010 Nov ;11(11) :1029-35. doi: 10.1016/S1470-2045(10)70233-3. Epub 2010.

[42] Francois Brécheteau et al. How Did We Obtain Complete Remission in Patients Who Had Metastatic Renal Cancer in the Era of Targeted Therapies? Annals of Surgical Oncology [23 Sep 2016, 24(2):369-374]

[43] Amato RJ et al. Chemotherapy for renal cell carcinoma Semin Oncol. 2000 Apr ;27(2):177- 86.

[44] Foss SD et al. Interferon in metastatic renal cell carcinoma. Semin Oncol. 2000 Apr ;27(2):187-93

[45] The Lancet Medical Research Council Renal Cancer Collaborators Interferon-α and survival in metastatic renal carcinoma: early results of a randomised controlled trial Volume 353, No. 9146, p14–17, 2 January 1999

[46] Motzer RJ et al. Effect of cytokine therapy on survival for patients with advanced renal cell carcinoma. J Clin Oncol. 2000 May;18(9):1928-35.

[47] Atzpodien J. et al. Interleukin-2- and interferon alfa-2a-based immune chemotherapy in advanced renal cell carcinoma: A Prospectively Randomized Trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). J Clin Oncol. 2004 Apr 1;22(7):1188- 94. Epub 2004 Feb 23.

[48] Mickisch GH et al. Radical nephrectomy plus interferon-alfa-based immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet. 2001 Sep 22;358(9286):966-70.

184

[49] Flanigan RC et al. Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell cancer. N Engl J Med. 2001 Dec 6;345(23):1655-9. [50] Fujikawa K et al. Serum immunosuppressive acidic protein and natural killer cell activity in patients with metastatic renal cell carcinoma before and after nephrectomy. J Urol. 2000 Sep;164(3 Pt 1):673-5.

[51] Rini BI. The evolving role of surgery for advanced renal cell carcinoma in the era of molecular targeted therapy.

[52] Foster K. Somatic mutations of the von Hippel-Lindau disease tumour suppressor gene in non-familial clear cell renal carcinoma.Hum Mol Genet. 1994 Dec;3(12):2169-73.

[53] Richard S Genetics and angiogenesis: the example of von Hippel-Lindau disease. Bull Cancer. 2007 Jul;94 Spec No: S170-9.

[54] Yang JC et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 2003 Jul 31;349(5):427-34.

[55] Escudier B Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007 Dec 22;370(9605):2103- 11. et al.

[56] Motzer RJ et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007 Jan 11;356(2):115-24.

[57] Site de l’Agence Europpénne du Medicament – AMM du Sunitinib - L’agence europénne du medicament.

[58] Sternberg CN et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010 Feb 20;28(6):1061-8. doi: 10.1200/JCO.2009.23.9764. Epub 2010 Jan 25.

[59] Escudier B et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007 Jan 11;356(2):125-34.

[60] Motzer RJ et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol. 2013 May;14(6):552-62. doi: 10.1016/S1470-2045(13)70093-7. Epub 2013 Apr 16.

[61] George S et al. Safety and Efficacy of Nivolumab in Patients with Metastatic Renal Cell Carcinoma Treated Beyond Progression: A Subgroup Analysis of a Randomized Clinical Trial. JAMA Oncol. 2016 Sep 1;2(9):1179-86. doi: 10.1001/jamaoncol.2016.0775

[62] George S et al. Renal cell carcinoma and antiangiogenic agents: Ongoing controversies are seeking answers for improvement of therapeutic management. Progrès en urologie (2009) 19, 596—605 F. Audenet et al.

[63] Barlesi et al. Biomarqueurs France: résultats de l’analyse en routine de EGFR, HER2, KRAS, BRAF, PI3K, et ALK sur 10 000 patients (pts) atteints de cancer bronchique non à petites cellules (CBNPC) Revue des Maladies Respiratoires Volume 31, numéro S1 F..

185

[64] Site internet de l’HAS - COMMISSION DE LA TRANSPARENCE Avis du 17 mai 2017 concernant le pembrolizumab. https://www.has-sante.fr/portail/upload/docs/evamed/CT- 15941_KEYTRUDA_PIC_EI_poumon_1ere_ligne_Avis2_CT15941.pdf

[65] P. Tomasini Thérapies moléculaires ciblées en cancérologie pulmonaire. EMC - Pneumologie 2017:1-9 [Article 6-002-L-13].

[66] Mok, T. S. et al. Gefitinib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. 361, 947–957 (2009).

[67] Rosell, R. et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 13, 239–246 (2012), ]

[68] Wu, Y.-L. et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, openlabel, ENSURE study. Ann. Oncol. mdv270 (2015). doi:10.1093/annonc/mdv270 ] [69] Mitsudomi, T. et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small- cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 11, 121–128 (2010). ]

[70] Maemondo, M. et al. Gefitinib or Chemotherapy for Non–Small-Cell Lung Cancer with Mutated EGFR. N. Engl. J. Med. 362, 2380–2388 (2010). ]

[71] Han, J.-Y. et al. First-SIGNAL: First-Line Single-Agent Iressa Versus Gemcitabine and Cisplatin Trial in Never-Smokers with Adenocarcinoma of the Lung. J. Clin. Oncol. 30, 1122– 1128 (2012)]

[72] Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol. 15, 213–222 (2014).

[73] Wu, Y.-L. et al. Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations. J. Clin. Oncol. 31, 3327–3334 (2013) ] Sequist, L. V. et al.

[74] Sequist, L. V. et al. Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients with Metastatic Lung Adenocarcinoma with EGFR Mutations. J. Clin. Oncol. 31, 3327–3334 (2013)] [75] K. Wolffe et al. Chapter 124: Cutaneous melanoma,” in Fitzpatrick’s Dermatology In General Medicine, Seventh Edition: Two Volumes, 7th ed., vol. 1, 2 vols., McGraw-Hill Companies,Incorporated, 2007, pp. 1134–1157

[76] R. Mouawad, D. Khayat, and J.-P. Spano, “Les mélanomes : facteurs pronostiques et traitements adjuvants. Traitement médical du mélanome malin métastatique,” in Thérapeutique du cancer, Paris: Springer Paris, 2011, pp. 673–683.

[77] C. Garbe, K. Peris, A. Hauschild, P. Saiag, M. Middleton, A. Spatz, J.-J. Grob, J. Malvehy, J. Newton-Bishop, A. Stratigos, H. Pehamberger, and A. Eggermont, “Diagnosis and treatment of melanoma: European consensusbased interdisciplinary guideline,” European Journal of Cancer, vol. 46, no. 2, pp. 270–283, Jan. 2010.

[78] M. R. Albertini et al. B. J. Longley, P. M. Harari, and D. Reintgen, “Cutaneous Melanoma,” in Oncology, Eds. New York, NY: Springer New York, pp. 1082–1101.

186

[79] L. Finn, S. Markovic, and R. Joseph, “Therapy for metastatic melanoma: the past, present, and future,” BMC Medicine, vol. 10, no. 1, p. 23, Mar. 2012.

[80] E. Vakiani and D. B. Solit, “KRAS and BRAF : drug targets and predictive biomarkers,” The Journal of Pathology, vol. 223, no. 2, pp. 220–230, 2011.

[81] K. T. Flaherty and G. McArthur, “BRAF, a target in melanoma,” Cancer, vol. 116, no. 21, pp. 4902–4913, 2010.

[82] J. A. Sosman, et al. “Survival in BRAF V600–Mutant Advanced Melanoma Treated with Vemurafenib,” New England Journal of Medicine, vol. 366, no. 8, pp. 707–714, 2012.

[83] A de la Fouchardière, “Mélanome et recherche de cibles thérapeutiques,” Annales de pathologie, no. 31, pp. 123–124, Oct. 2011.

[84] E. Espinosa and P. Chillet, “Chapitre 10: Le lymphocyte Th au centre de la réponse immunitaire,” in Immunologie, Ellipses Marketing, 2010, pp. 245–287

[85] Kaplan MG. Ipilimumab plus Dacarbazine in Melanoma,” New England Journal of Medicine, vol. 365, no. 13, pp. 1256–1258, 2011.

[86] Improved Survival with Ipilimumab in Patients with Metastatic Melanoma,” New England Journal of Medicine, vol. 363, no. 8, pp. 711–723, 2010. F. S. Hodi et al.

[87]Robert C et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015 Jan 22; 372(4):320-30. doi: 10.1056/NEJMoa1412082. Epub 2014 Nov 16 [88] Weber JS et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015 Apr; 16(4):375-84. Epub 2015 Mar 18.

[89] Rapport des innovations thérapeutiques dans le mélanome au congrès de l'American Association for Cancer Research (AACR) à la Nouvelle-Orléans de 2016 par le site d’information APMnews - www.apmnews.com/

[90] Critères RECIST version 1.1 Response Evaluation Criteria in Solid Tumors http://www.cepd.fr/CUSTOM/RECIST_1.1.pdf

[91] Daniel Gioeli Targeted therapies: Mechanisms of resistance Molecular and translational medecine. Edition Humana Press

[92] Lackner MR et al. Mechanisms of acquired resistance to targeted cancer therapies Future Oncol. 2012 Aug ;8(8):999-1014. doi: 10.2217/fon.12.86.

[93] Branford S et al. High frequency of point mutations clustered within the adenosine triphosphatebinding region of BCR/ABL in patients with chronic myeloid leukemia or Ph- positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance. Blood. 2002 ; 99(9):3472–5.

[94] Hofmann WK et al. Ph (+) acute lymphoblastic leukemia resistant to the tyrosine kinase inhibitor STI571 has a unique BCR-ABL gene mutation. Blood. 2002 ;99(5) :1860–2

[95] Shyamala Maheswaran et al. Detection of Mutations in EGFR in Circulating Lung-Cancer Cells N Engl J Med. 2008 Jul 24 ; 359(4): 366–377 2008 Jul 2.

[96] Weisberg E et al. Second generation inhibitors of BCR-ABL for the treatment of imatinibresistant

187

chronic myeloid leukaemia. Nat Rev Cancer. 2007 ;7(5):345–56

[97] Mulloy R et al. Epidermal growth factor receptor mutants from human lung cancers exhibit enhanced catalytic activity and increased sensitivity to gefitinib. Cancer Res. 2007 ;67(5): 2325–30

[98] Sequist LV et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl Med.3(75),75ra26 (2011).

[99] Engelman JA et al. Allelic dilution obscures detection of a biologically significant resistance Mutation in EGFR-amplified lung cancer. J Clin Invest. 2006 ;116(10):2695–706.

[100] Liegl B et al. Heterogeneity of kinase inhibitor resistance mechanisms in GIST. J Pathol. 2008 ;216(1):64–74

[101] Balak MN et al. Novel D761Y and common secondary T790M mutations in epidermal growthFactor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res. 2006 ;12(21):6494–501

[102] Roche-Lestienne C, Soenen-Cornu V, Grardel-Duflos N, et al. Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment. Blood 2002 ; 100 : 1014–1018.

[103] Landau DA, Carter SL, Stojanov P, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 2013 ; 152 : 714–726

[104] Klco JM, Spencer DH, Miller CA, et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell 2014 ; 25 : 379–392.

[105] Diaz LA, Jr, Bardelli A. Liquid biopsies : genotyping circulating tumor DNA. J Clin Oncol 2014 ; 32 : 579–586.

[106] amal-Hanjani M, Thanopoulou E, Peggs KS, et al. Tumour heterogeneity and immune- modulation. Curr Opin Pharmacol 2013 ; 13 : 497–503

[107] Sos ML, Koker M, Weir BA et al. PTEN loss contributes to erlotinib resistance in EGFR- mutant lung cancer by activation of Akt and EGFR. Cancer Res.69(8),3256–3261 (2009

[108] Sequist LV, Waltman BA, Dias-Santagata D et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl Med.3(75),75ra26 (2011)

[109] Nagy P, Friedlander E, Tanner M et al. Decreased accessibility and lack of activation of ErbB2 in JIMT-1, a Herceptin-resistant, MUC4-expressing breast cancer cell line. Cancer

Res.65(2),473–482 (2005).

[110] Scaltriti M, Serra V, Normant E et al. Antitumor activity of the Hsp90 inhibitor IPI-504 in HER2-positive trastuzumab-resistant breast cancer. Mol. Cancer Ther.10(5),817–824 (2011).

[111] Modi S, Stopeck AT, Gordon MS et al. Combination of trastuzumab and tanespimycin (17-AAG, KOS-953) is safe and active in trastuzumab-refractory HER-2 overexpressing breast cancer: a Phase I dose-escalation study. J. Clin. Oncol.25(34),5410–5417 (2007).

[112] McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J. Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res.70(4),1625–1634 (2010).

188

[113] Sequist LV, Gettinger S, Senzer NN et al. Activity of IPI-504, a novel heat-shock protein 90 inhibitor, in patients with molecularly defined non-small-cell lung cancer. J. Clin.

Oncol.28(33),4953–4960 (2010).

[114] Wagle N, Emery C, Berger MF et al. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J. Clin. Oncol.29(22),3085–3096 (2011).

[115] Hatzivassiliou G, Liu B, O'Brien C et al. ERK inhibition overcomes acquired resistance to MEK inhibitors. Mol. Cancer Ther.11(5),1143–1154 (2012).

[116] Muellner MK, Uras IZ, Gapp BV et al. A chemical-genetic screen reveals a mechanism of resistance to PI3K inhibitors in cancer. Nat. Chem. Biol.7(11),787–793 (2011).

[117] Peng XX, Tiwari AK, Wu HC, Chen ZS. Overexpression of P-glycoprotein induces acquired resistance to imatinib in chronic myelogenous leukemia cells. Chin. J. Cancer31(2),110–118 (2012).

[118] Mahon FX, Hayette S, Lagarde V et al. Evidence that resistance to nilotinib may be due to BCR-ABL, Pgp, or Src kinase overexpression. Cancer Res.68(23),9809–9816 (2008).

[119] Theou N, Gil S, Devocelle A et al. Multidrug resistance proteins in gastrointestinal stromal tumors: site-dependent expression and initial response to imatinib. Clin. Cancer

Res.11(21),7593–7598 (2005).

[120] Miller TW, Perez-Torres M, Narasanna A et al. Loss of phosphatase and tensin homologue deleted on chromosome 10 engages ErbB3 and insulin-like growth factor-I receptor signaling to promote antiestrogen resistance in breast cancer. Cancer

Res.69(10),4192–4201 (2009).

[121] Rexer BN, Ghosh R, Arteaga CL. Inhibition of PI3K and MEK: it is all about combinations and biomarkers. Clin. Cancer Res.15(14),4518–4520 (2009).

[122] Buonamici S, Williams J, Morrissey M et al. Interfering with resistance to Smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Sci. Transl

Med.2(51),51ra70 (2010).

[123] Sasaki T, Koivunen J, Ogino A et al. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res.71(18),6051–6060 (2011).

[124] Engelman JA, Zejnullahu K, Mitsudomi T et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science316(5827),1039–1043 (2007).

[125] Miller TW, Balko JM, Arteaga CL. Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer. J. Clin. Oncol.29(33),4452–4461 (2011).

[126] Miller TW, Hennessy BT, Gonzalez-Angulo AM et al. Hyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancer. J. Clin. Invest.120(7),2406–2413 (2010).

[127] Chen Y, Sawyers CL, Scher HI. Targeting the androgen receptor pathway in prostate cancer. Curr. Opin Pharmacol.8(4),440–448 (2008).

[128] Carver BS, Chapinski C, Wongvipat J et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell19(5),575–586 (2011).

189

[129] Rexer BN, Ghosh R, Arteaga CL. Inhibition of PI3K and MEK: it is all about combinations and biomarkers. Clin. Cancer Res.15(14),4518–4520 (2009).

[130) Chandarlapaty S, Sawai A, Scaltriti M et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell19(1),58–71 (2011).

[131] Chakrabarty A, Sanchez V, Kuba MG, Rinehart C, Arteaga CL. Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc. Natl

Acad. Sci. USA109(8),2718–2723 (2011

[132] Yauch RL, Januario T, Eberhard DA et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin. Cancer Res.11(24 Pt 1),8686–8698 (2005).

[133] Gupta PB, Onder TT, Jiang G et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell138(4),645–659 (2009).

[134] Chung JH, Rho JK, Xu X et al. Clinical and molecular evidences of epithelial to mesenchymal transition in acquired resistance to EGFR-TKIs. Lung Cancer73(2),176–182 (2011).

[135] Li X, Lewis MT, Huang J et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl Cancer Inst.100(9),672–679 (2008).

[136] Sharma SV, Lee DY, Li B et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell141(1),69–80 (2010).

[137] Scheel C, Weinberg RA. Phenotypic plasticity and epithelial-mesenchymal transitions in cancer and normal stem cells? Int. J. Cancer129(10),2310–2314 (2011).

[138] Guix M, Faber AC, Wang SE et al. Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cells is mediated by loss of IGF-binding proteins. J. Clin. Invest.118(7),2609–2619 (2008).

[139] Morgillo F, Cascone T, D'Aiuto E et al. Antitumour efficacy of MEK inhibitors in human lung cancer cells and their derivatives with acquired resistance to different tyrosine kinase inhibitors. Br. J. Cancer105(3),382–392 (2011).

[140] Corcoran RB, Ebi H, Turke AB et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov.2(3),227–235 (2012).

[141] Nahta R, Yuan LX, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res.65(23),11118–11128 (2005).

[142] Sun Y, Wang BE, Leong KG et al. Androgen deprivation causes epithelial-mesenchymal transition in the prostate : implications for androgen-deprivation therapy. Cancer

Res.72(2),527–536 (2011).

[143] Groupe de travail bas normand THECITOX, Karine Bouhier-Leporrier et al. Prise en charge des effets secondaires des thérapies ciblées Edition Potentiel d’Action

[144] Coquan E et al. Principes de prise en charge des effets indésirables des thérapies ciblées en cancérologie. La revue du praticien, 2012, vol.62

190

[145] Chevreau C. Les effets secondaires des anti-angiogéniques (à l’exception des effets cutanés). Oncologie, 2009, vol 11, pp 285-290

[146] Alliance Pour la Recherche En Cancérologie (APREC), Cellule Information Conseil des Thérapies Ciblées des Cancers, Maxime Japkowicz, Tevy San, Joseph Gligoro Cancers, thromboses veineuses et traitements anti-angiogéniques http://www.jle.com/

[147] Sibaud V, et al. Effets indésirables dermatologiques des thérapies ciblées angio- géniques. Bulletin du cancer, 2011, vol 98, n°10, pp 1221-1229

[148] Lacouture M et al. Clinical practice guidelines for the prevention and treatment of EGFR inhibitor-associated dermotologic toxicities. Support Care Cancer, 2011 vol 19 pp1079-1095 [149] Guilhot Fet al. Indications for imatinib mesylate therapy and clinical managment. The oncologist 2044, vol 9, pp271-28

[150] Bouché O et al. Prise en charge des effets secondaires des thérapeutiques ciblees (hors dermatologiques). Hepato-Gastro, vol 14 pp15-23

[151] Halimi JM, et al. Effets vasculaires et rénaux des médicaments anti-angiogéniques : recommandations francaises pour la pratique. Sang thrombose vaisseaux, 2009, vol.21 pp 151-166.

[152] Izzedine H, et al. Gestion des effets secondaires des thérapies ciblées dans le cancer du rein : Effets secondaires néphrologiques. Bulletin du cancer 2011 vol 98 pp 7-18

[153] Jabbour E, Deininger M et al. Management of adverse events asscioted with tyrosine kinase dans la leucémie myeloide chronique. Leukemia, 2011 pp 2010

[154] Rea D et al. Aspects pratiques des traitements par inhibiteurs de tyrosine kinase dans la leucémie myéloïde chronique. Hématologie 2009 vol15 n°3

[155] Bossert C et al. Les complications thyroïdiennes des inhibiteurs de tyrosine kinase. Médecine thérapeutique, 2011 vol 17 n°4 pp 302-309

[156] Peter Singer, Why We Must Ration Health Care ? New York Times Published: July 15, 2009 http://www.nytimes.com/2009/07/19/magazine/19healthcare-t.html?pagewanted=all