J. Ferlay, H. Shin, F. Bray, D. Forman, C. Mathers et al., Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008, International Journal of Cancer, vol.8, issue.19, p.2893917, 2010.
DOI : 10.1002/ijc.25516

J. Ferlay, E. Steliarova-foucher, J. Lortet-tieulent, S. Rosso, J. Coebergh et al., Cancer incidence and mortality patterns in Europe: estimates for 40 countries, 2012.

W. Organization, WHO Classification of Tumours of the Digestive System -4ième édition, 2010.

B. Morson, J. Whiteway, E. Jones, and F. Macrae, Williams : CB. Histopathology and prognosis of malignant colorectal polyps treated by endoscopic polypectomy, Gut. mai, vol.25, issue.5, p.43744, 1984.

P. Galiatsatos and W. Foulkes, Familial Adenomatous Polyposis, The American Journal of Gastroenterology, vol.112, issue.2, p.38598, 2006.
DOI : 10.1007/BF02234304

H. Vasen, G. Moslein, A. Alonso, S. Aretz, I. Bernstein et al., Guidelines for the clinical management of familial adenomatous polyposis (FAP), Gut, vol.57, issue.5, p.70413, 2008.
DOI : 10.1136/gut.2007.136127

N. Al-tassan, N. Chmiel, J. Maynard, N. Fleming, A. Livingston et al., Inherited variants of MYH associated with somatic G:C???T:A mutations in colorectal tumors, Nature Genetics, vol.30, issue.2, p.22732, 2002.
DOI : 10.1038/ng828

R. Riddell, H. Goldman, D. Ransohoff, H. Appelman, C. Fenoglio et al., Dysplasia in inflammatory bowel disease: Standardized classification with provisional clinical applications, Human Pathology, vol.14, issue.11, p.93168, 1983.
DOI : 10.1016/S0046-8177(83)80175-0

W. Kühnel, Atlas de poche d'Histologie. Flammarion, éditeur, 1997.

R. Bird, Observation and quantification of aberrant crypts in the murine colon treated with a colon carcinogen: Preliminary findings, Cancer Letters, vol.37, issue.2, p.14751, 1987.
DOI : 10.1016/0304-3835(87)90157-1

G. Caderni, F. Bianchini, A. Mancina, M. Spagnesi, and P. Dolara, Effect of dietary carbohydrates on the growth of dysplastic crypt foci in the colon of rats treated with 1,2- dimethylhydrazine, Cancer Res. 15 juill, vol.51, issue.14, p.37215, 1991.

T. Pretlow, B. Barrow, W. Ashton, O. Riordan, M. Pretlow et al., Aberrant crypts: putative preneoplastic foci in human colonic mucosa, Cancer Res. 1 mars, vol.51, issue.5, p.15647, 1991.
DOI : 10.1002/jcb.240501111

K. Otori, K. Sugiyama, T. Hasebe, S. Fukushima, and H. Esumi, Emergence of adenomatous aberrant crypt foci (ACF) from hyperplastic ACF with concomitant increase in cell proliferation, Cancer Res. 1 nov, vol.55, issue.21, p.47436, 1995.

M. Nucci, C. Robinson, P. Longo, P. Campbell, and S. Hamilton, Phenotypic and genotypic characteristics of aberrant crypt foci in human colorectal mucosa, Human Pathology, vol.28, issue.12, p.13961407, 1997.
DOI : 10.1016/S0046-8177(97)90230-6

S. Kudo, Endoscopic Mucosal Resection of Flat and Depressed Types of Early Colorectal Cancer, Endoscopy, vol.25, issue.07, p.45561, 1993.
DOI : 10.1055/s-2007-1010367

K. Polyak, S. Hamilton, B. Vogelstein, and K. Kinzler, Early alteration of cell-cycleregulated gene expression in colorectal neoplasia, Am J Pathol. août, vol.149, issue.2, p.381387, 1996.

R. Schlemper, R. Riddell, Y. Kato, F. Borchard, H. Cooper et al., The Vienna classification of gastrointestinal epithelial neoplasia, Gut, vol.47, issue.2, p.2515, 2000.
DOI : 10.1136/gut.47.2.251

H. Yoon, A. Martin, R. Benamouzig, E. Longchampt, J. Deyra et al., Interobserver agreement on histological diagnosis of colorectal polyps: the APACC study], Gastroentérologie Clin Biol. mars, vol.26, issue.3, p.220224, 2002.

M. Terry, A. Neugut, R. Bostick, J. Potter, R. Haile et al., Reliability in the classification of advanced colorectal adenomas, Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol. juill, vol.11, issue.7, p.660663, 2002.

C. Lengauer, K. Kinzler, and B. Vogelstein, Genetic instabilities in human cancers, Nature. 17 déc, vol.396, issue.6712, p.6439, 1998.

B. Vogelstein, E. Fearon, S. Hamilton, S. Kern, A. Preisinger et al., Genetic Alterations during Colorectal-Tumor Development, New England Journal of Medicine, vol.319, issue.9, p.52532, 1988.
DOI : 10.1056/NEJM198809013190901

W. Samowitz, H. Albertsen, J. Herrick, T. Levin, C. Sweeney et al., Evaluation of a Large, Population-Based Sample Supports a CpG Island Methylator Phenotype in Colon Cancer, Gastroenterology, vol.129, issue.3, p.83745, 2005.
DOI : 10.1053/j.gastro.2005.06.020

M. Kane, M. Loda, G. Gaida, . J. Lipman, R. Mishra et al., Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines, Cancer Res. 1 mars, vol.57, issue.5, p.808811, 1997.

H. Lynch and A. De-la-chapelle, Hereditary colorectal cancer, N Engl J Med. 6 mars, vol.348, issue.10, p.919932, 2003.

P. Peltomäki and A. De-la-chapelle, Mutations Predisposing to Hereditary Nonpolyposis Colorectal Cancer, Adv Cancer Res, vol.71, p.93119, 1997.
DOI : 10.1016/S0065-230X(08)60097-4

N. Papadopoulos and A. Lindblom, Molecular basis of HNPCC: mutations of MMR genes, Hum Mutat, vol.10, issue.2, p.8999, 1997.

S. Fewell, K. Travers, J. Weissman, and J. Brodsky, The Action of Molecular Chaperones in the Early Secretory Pathway, Annual Review of Genetics, vol.35, issue.1, p.149191, 2001.
DOI : 10.1146/annurev.genet.35.102401.090313

C. Dobson, Principles of protein folding, misfolding and aggregation, Seminars in Cell & Developmental Biology, vol.15, issue.1, p.316, 2004.
DOI : 10.1016/j.semcdb.2003.12.008

A. Zavialov and M. Ehrenberg, Peptidyl-tRNA Regulates the GTPase Activity of Translation Factors, Cell, vol.114, issue.1, p.113122, 2003.
DOI : 10.1016/S0092-8674(03)00478-1

S. Blanchard, R. Gonzalez, H. Kim, S. Chu, and J. Puglisi, tRNA selection and kinetic proofreading in translation, Nature Structural & Molecular Biology, vol.7, issue.10, p.10081014, 2004.
DOI : 10.1126/science.289.5481.905

T. Dever, L. Feng, R. Wek, A. Cigan, T. Donahue et al., Phosphorylation of initiation factor 2 alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 in yeast, Cell. 7 févr, vol.68, issue.3, p.585596, 1992.

P. Hochachka, L. Buck, C. Doll, and S. Land, Unifying theory of hypoxia tolerance: molecular/metabolic defense and rescue mechanisms for surviving oxygen lack., Proceedings of the National Academy of Sciences, vol.93, issue.18, p.94939498, 1996.
DOI : 10.1073/pnas.93.18.9493

C. Koumenis, C. Naczki, M. Koritzinsky, S. Rastani, A. Diehl et al., Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2 alpha, Mol Cell Biol. nov, vol.22, issue.21, p.74057416, 2002.

M. Bi, C. Naczki, M. Koritzinsky, D. Fels, J. Blais et al., ER stress-regulated translation increases tolerance to extreme hypoxia and, Embo J. 5 oct, vol.24, p.347081, 2005.

D. Feldman, V. Chauhan, and A. Koong, The Unfolded Protein Response: A Novel Component of the Hypoxic Stress Response in Tumors, Molecular Cancer Research, vol.3, issue.11, p.597605, 2005.
DOI : 10.1158/1541-7786.MCR-05-0221

Y. Kozutsumi, M. Segal, K. Normington, M. Gething, and J. Sambrook, The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-related proteins, Nature. 31 mars, vol.332, issue.6163, p.462464, 1988.

H. Harding, Y. Zhang, and R. D. , Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase, Nature. 21 janv, vol.397, issue.6716, p.271274, 1999.

K. Travers, C. Patil, L. Wodicka, D. Lockhart, J. Weissman et al., Functional and Genomic Analyses Reveal an Essential Coordination between the Unfolded Protein Response and ER-Associated Degradation, Cell, vol.101, issue.3, p.249258, 2000.
DOI : 10.1016/S0092-8674(00)80835-1

O. Larsson, M. Carlberg, and A. Zetterberg, Selective killing induced by an inhibitor of Nlinked glycosylation, J Cell Sci. sept, vol.106, p.299307, 1993.

T. Nakagawa, H. Zhu, N. Morishima, E. Li, J. Xu et al., Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta, Nature. 6 janv, vol.403, issue.6765, p.98103, 2000.

L. Scorrano, S. Oakes, J. Opferman, E. Cheng, M. Sorcinelli et al., BAX and BAK Regulation of Endoplasmic Reticulum Ca2+: A Control Point for Apoptosis, Science, vol.300, issue.5616, p.135139, 2003.
DOI : 10.1126/science.1081208

S. Marciniak, C. Yun, S. Oyadomari, I. Novoa, Y. Zhang et al., CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum, Genes & Development, vol.18, issue.24, p.306677, 2004.
DOI : 10.1101/gad.1250704

J. Lin, H. Li, D. Yasumura, H. Cohen, C. Zhang et al., IRE1 Signaling Affects Cell Fate During the Unfolded Protein Response, Science, vol.318, issue.5852, p.944949, 2007.
DOI : 10.1126/science.1146361

A. Bertolotti, Y. Zhang, L. Hendershot, H. Harding, and R. D. , Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response, Nat Cell Biol. juin, vol.2, issue.6, p.326332, 2000.

J. Ye, R. Rawson, R. Komuro, X. Chen, U. Dave et al., ER Stress Induces Cleavage of Membrane-Bound ATF6 by the Same Proteases that Process SREBPs, Molecular Cell, vol.6, issue.6, p.13551364, 2000.
DOI : 10.1016/S1097-2765(00)00133-7

K. Okamura, Y. Kimata, H. Higashio, A. Tsuru, and K. Kohno, Dissociation of Kar2p/BiP from an ER Sensory Molecule, Ire1p, Triggers the Unfolded Protein Response in Yeast, Biochemical and Biophysical Research Communications, vol.279, issue.2, p.445450, 2000.
DOI : 10.1006/bbrc.2000.3987

C. Liu, Z. Xu, and R. Kaufman, Structure and intermolecular interactions of the luminal dimerization domain of human IRE1 alpha, J Biol Chem. 16 mai, vol.278, issue.20, p.1768017687, 2003.

J. Credle, J. Finer-moore, F. Papa, R. Stroud, and P. Walter, On the mechanism of sensing unfolded protein in the endoplasmic reticulum, Proceedings of the National Academy of Sciences, vol.102, issue.52, p.1877318784, 2005.
DOI : 10.1073/pnas.0509487102

C. Shamu and P. Walter, Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus, Embo J. 17 juin, vol.15, issue.12, p.30283039, 1996.

C. Sidrauski and P. Walter, The Transmembrane Kinase Ire1p Is a Site-Specific Endonuclease That Initiates mRNA Splicing in the Unfolded Protein Response, Cell, vol.90, issue.6, p.10311039, 1997.
DOI : 10.1016/S0092-8674(00)80369-4

H. Yoshida, T. Matsui, A. Yamamoto, T. Okada, and K. Mori, XBP1 mRNA Is Induced by ATF6 and Spliced by IRE1 in Response to ER Stress to Produce a Highly Active Transcription Factor, Cell, vol.107, issue.7, p.881891, 2001.
DOI : 10.1016/S0092-8674(01)00611-0

H. Yoshida, M. Oku, M. Suzuki, and K. Mori, pXBP1(U) encoded in XBP1 pre-mRNA negatively regulates unfolded protein response activator pXBP1(S) in mammalian ER stress response, The Journal of Cell Biology, vol.113, issue.4, p.565575, 2006.
DOI : 10.1016/S1534-5807(03)00022-4

J. Hollien, J. Lin, H. Li, N. Stevens, P. Walter et al., Regulated Ire1-dependent decay of messenger RNAs in mammalian cells, The Journal of Cell Biology, vol.186, issue.3, p.323331, 2009.
DOI : 10.1128/MCB.22.11.3864-3874.2002

S. Marciniak, L. Garcia-bonilla, J. Hu, H. Harding, and R. D. , Activation-dependent substrate recruitment by the eukaryotic translation initiation factor 2 kinase PERK, The Journal of Cell Biology, vol.9, issue.2, p.201209, 2006.
DOI : 10.1128/MCB.22.11.3864-3874.2002

Y. Shi, K. Vattem, R. Sood, A. J. Liang, J. Stramm et al., Identification and characterization of pancreatic eukaryotic initiation factor 2 alpha-subunit kinase, PEK, involved in translational control, Mol Cell Biol. déc, vol.18, issue.12, p.74997509, 1998.

A. Hinnebusch, Molecular Mechanism of Scanning and Start Codon Selection in Eukaryotes, Microbiology and Molecular Biology Reviews, vol.75, issue.3, p.434467, 2011.
DOI : 10.1128/MMBR.00008-11

G. Pavitt, K. Ramaiah, S. Kimball, and A. Hinnebusch, eIF2 independently binds two distinct eIF2B subcomplexes that catalyze and regulate guanine-nucleotide??exchange, Genes & Development, vol.12, issue.4, p.514526, 1998.
DOI : 10.1101/gad.12.4.514

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC316533

H. Harding, I. Novoa, Y. Zhang, H. Zeng, R. Wek et al., Regulated Translation Initiation Controls Stress-Induced Gene Expression in Mammalian Cells, Molecular Cell, vol.6, issue.5, p.10991108, 2000.
DOI : 10.1016/S1097-2765(00)00108-8

URL : http://doi.org/10.1016/s1097-2765(00)00108-8

D. Scheuner, B. Song, E. Mcewen, C. Liu, R. Laybutt et al., Translational Control Is Required for the Unfolded Protein Response and In Vivo Glucose Homeostasis, Molecular Cell, vol.7, issue.6, p.11651176, 2001.
DOI : 10.1016/S1097-2765(01)00265-9

K. Vattem and R. Wek, Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells, Proceedings of the National Academy of Sciences, vol.101, issue.31, p.1126911274, 2004.
DOI : 10.1073/pnas.0400541101

P. Lu, H. Harding, and R. D. , Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response, The Journal of Cell Biology, vol.7, issue.1, p.2733, 2004.
DOI : 10.1128/MCB.22.19.6681-6688.2002

H. Harding, Y. Zhang, H. Zeng, I. Novoa, P. Lu et al., An Integrated Stress Response Regulates Amino Acid Metabolism and Resistance to Oxidative Stress, Molecular Cell, vol.11, issue.3, p.619633, 2003.
DOI : 10.1016/S1097-2765(03)00105-9

Y. Ma, J. Brewer, J. Diehl, and L. Hendershot, Two Distinct Stress Signaling Pathways Converge Upon the CHOP Promoter During the Mammalian Unfolded Protein Response, Journal of Molecular Biology, vol.318, issue.5, p.13511365, 2002.
DOI : 10.1016/S0022-2836(02)00234-6

I. Novoa, H. Zeng, H. Harding, and R. D. , Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2 alpha, J Cell Biol. 28 mai, vol.153, issue.5, p.10111021, 2001.

Y. Ma and L. Hendershot, Delineation of a Negative Feedback Regulatory Loop That Controls Protein Translation during Endoplasmic Reticulum Stress, Journal of Biological Chemistry, vol.278, issue.37, p.3486434873, 2003.
DOI : 10.1074/jbc.M301107200

H. Zinszner, M. Kuroda, X. Wang, N. Batchvarova, R. Lightfoot et al., CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic??reticulum, Genes & Development, vol.12, issue.7, p.982995, 1998.
DOI : 10.1101/gad.12.7.982

K. Mccullough, J. Martindale, L. Klotz, T. Aw, and N. Holbrook, Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2, Mol Cell Biol. févr, vol.21, issue.4, p.124959, 2001.

S. Oyadomari, K. Takeda, M. Takiguchi, T. Gotoh, M. Matsumoto et al., Nitric oxide-induced apoptosis in pancreatic beta cells is mediated by the, Proc Natl Acad Sci U A. 11 sept, vol.98, p.1084550, 2001.

K. Haze, H. Yoshida, H. Yanagi, T. Yura, and K. Mori, Mammalian Transcription Factor ATF6 Is Synthesized as a Transmembrane Protein and Activated by Proteolysis in Response to Endoplasmic Reticulum Stress, Molecular Biology of the Cell, vol.10, issue.11, p.37873799, 1999.
DOI : 10.1091/mbc.10.11.3787

Y. Wang, J. Shen, N. Arenzana, W. Tirasophon, R. Kaufman et al., Activation of ATF6 and an ATF6 DNA binding site by the endoplasmic reticulum stress response, J Biol Chem. 1 sept, vol.275, issue.35, p.2701327020, 2000.

J. Shen, X. Chen, L. Hendershot, and R. Prywes, ER Stress Regulation of ATF6 Localization by Dissociation of BiP/GRP78 Binding and Unmasking of Golgi Localization Signals, Developmental Cell, vol.3, issue.1, p.99111, 2002.
DOI : 10.1016/S1534-5807(02)00203-4

J. Wu, D. Rutkowski, M. Dubois, J. Swathirajan, T. Saunders et al., ATF6 alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress, Dev Cell. sept, vol.13, issue.3, p.351364, 2007.
DOI : 10.1016/j.devcel.2007.07.005

URL : http://doi.org/10.1016/j.devcel.2007.07.005

G. Gazit, J. Lu, and A. Lee, De???regulation of GRP stress protein expression in human breast cancer cell lines, Breast Cancer Research and Treatment, vol.90, issue.2, p.13546, 1999.
DOI : 10.1023/A:1006102411439

P. Fernandez, S. Tabbara, L. Jacobs, F. Manning, T. Tsangaris et al., Overexpression of the glucose-regulated stress gene GRP78 in malignant but not benign human breast lesions, Breast Cancer Research and Treatment, vol.58, issue.Suppl.740, p.1526, 2000.
DOI : 10.1023/A:1006332011207

K. Ozawa, Y. Tsukamoto, O. Hori, Y. Kitao, H. Yanagi et al., Regulation of tumor angiogenesis by oxygen-regulated protein 150, an inducible endoplasmic reticulum chaperone, Cancer Res. 15 mai, vol.61, issue.10, p.420613, 2001.

M. Song, Y. Park, J. Lee, and K. Park, Induction of glucose-regulated protein 78 by chronic hypoxia in human gastric tumor cells through a protein kinase C-epsilon/ERK/AP-1 signaling cascade, Cancer Res. 15 nov, vol.61, issue.22, p.832230, 2001.

M. Shuda, N. Kondoh, N. Imazeki, K. Tanaka, T. Okada et al., Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis, Journal of Hepatology, vol.38, issue.5, p.60514, 2003.
DOI : 10.1016/S0168-8278(03)00029-1

X. Chen, Y. Ding, C. Liu, S. Mikhail, and C. Yang, Overexpression of glucose-regulated protein 94 (Grp94) in esophageal adenocarcinomas of a rat surgical model and humans, Carcinogenesis, vol.23, issue.1, p.12330, 2002.
DOI : 10.1093/carcin/23.1.123

X. Xing, M. Lai, Y. Wang, E. Xu, and Q. Huang, Overexpression of glucose-regulated protein 78 in colon cancer, Clinica Chimica Acta, vol.364, issue.1-2, p.308315, 2006.
DOI : 10.1016/j.cca.2005.07.016

C. Koumenis and B. Wouters, "Translating" Tumor Hypoxia: Unfolded Protein Response (UPR)-Dependent and UPR-Independent Pathways, Molecular Cancer Research, vol.4, issue.7, p.423436, 2006.
DOI : 10.1158/1541-7786.MCR-06-0150

E. Jorgensen, A. Stinson, L. Shan, J. Yang, D. Gietl et al., Cigarette smoke induces endoplasmic reticulum stress and the unfolded protein response in normal and malignant human lung cells, BMC Cancer, vol.7, issue.4, p.000259072000001, 2008.
DOI : 10.1186/1471-2407-8-229

E. Lee, P. Nichols, D. Spicer, S. Groshen, M. Yu et al., GRP78 as a Novel Predictor of Responsiveness to Chemotherapy in Breast Cancer, Cancer Research, vol.66, issue.16, p.78497853, 2006.
DOI : 10.1158/0008-5472.CAN-06-1660

S. Daneshmand, M. Quek, E. Lin, C. Lee, R. Cote et al., Glucose-regulated protein GRP78 is up-regulated in prostate cancer and correlates with recurrence and survival, Human Pathology, vol.38, issue.10, p.15471552, 2007.
DOI : 10.1016/j.humpath.2007.03.014

J. Zhang, Y. Jiang, Z. Jia, Q. Li, W. Gong et al., Association of elevated GRP78 expression with increased lymph node metastasis and poor prognosis in patients with gastric cancer, Clinical & Experimental Metastasis, vol.51, issue.7-8, pp.7-8401410, 2006.
DOI : 10.1007/s10585-006-9051-9

Y. Fu, S. Wey, M. Wang, R. Ye, C. Liao et al., Pten null prostate tumorigenesis and AKT activation are blocked by targeted knockout of ER chaperone GRP78/BiP in prostate epithelium, Proceedings of the National Academy of Sciences, vol.105, issue.49, p.1944419449, 2008.
DOI : 10.1073/pnas.0807691105

L. Romero-ramirez, H. Cao, D. Nelson, E. Hammond, A. Lee et al., XBP1 Is Essential for Survival under Hypoxic Conditions and Is Required for Tumor Growth, Cancer Research, vol.64, issue.17, p.59437, 2004.
DOI : 10.1158/0008-5472.CAN-04-1606

J. Blais, C. Addison, R. Edge, T. Falls, H. Zhao et al., Perk-Dependent Translational Regulation Promotes Tumor Cell Adaptation and Angiogenesis in Response to Hypoxic Stress, Molecular and Cellular Biology, vol.26, issue.24, p.951732, 2006.
DOI : 10.1128/MCB.01145-06

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1698539

J. Ye, M. Kumanova, L. Hart, K. Sloane, H. Zhang et al., The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation, The EMBO Journal, vol.15, issue.12, p.208296, 2010.
DOI : 10.1002/jcb.21888

Z. Su, L. Emdad, D. Sarkar, A. Randolph, K. Valerie et al., Potential molecular mechanism for rodent tumorigenesis: mutational generation of Progression Elevated Gene-3 (PEG-3), Oncogene, vol.24, issue.13, p.224755, 2005.
DOI : 10.1038/sj.onc.1208420

L. Dalton, H. Clarke, J. Knight, M. Lawson, J. Wason et al., The endoplasmic reticulum stress marker CHOP predicts survival in malignant mesothelioma, British Journal of Cancer, vol.16, issue.6
DOI : 10.1101/gad.12.7.982

M. Spiotto, A. Banh, I. Papandreou, H. Cao, M. Galvez et al., Imaging the Unfolded Protein Response in Primary Tumors Reveals Microenvironments with Metabolic Variations that Predict Tumor Growth, Cancer Research, vol.70, issue.1, p.7888, 2010.
DOI : 10.1158/0008-5472.CAN-09-2747

G. Wang, Z. Yang, and K. Zhang, Endoplasmic reticulum stress response in cancer: molecular mechanism and, Am J Transl Res, vol.2, issue.1, p.6574, 2010.

I. Papandreou, N. Denko, M. Olson, H. Van-melckebeke, S. Lust et al., Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma, Blood, vol.117, issue.4, p.13114, 2011.
DOI : 10.1182/blood-2010-08-303099

N. Mimura, M. Fulciniti, G. Gorgun, Y. Tai, D. Cirstea et al., Blockade of XBP1 splicing by inhibition of IRE1alpha is a promising therapeutic option in multiple myeloma, Blood. United States, p.577281, 2012.

V. Eroschenko and M. Fiore, Di Fiore's atlas of histology with functional correlations, 2005.

M. Farquhar, G. Palade, . Junctional, . In, and . Epithelia, JUNCTIONAL COMPLEXES IN VARIOUS EPITHELIA, The Journal of Cell Biology, vol.17, issue.2, p.375, 1963.
DOI : 10.1083/jcb.17.2.375

J. Ivaska, H. Pallari, J. Nevo, and J. Eriksson, Novel functions of vimentin in cell adhesion, migration, and signaling, Experimental Cell Research, vol.313, issue.10, p.20502062, 2007.
DOI : 10.1016/j.yexcr.2007.03.040

E. Hay, The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it, Developmental Dynamics, vol.82, issue.3, p.70620, 2005.
DOI : 10.1002/dvdy.20345

G. Edelman, W. Gallin, A. Delouvee, B. Cunningham, and J. Thiery, Early epochal maps of two different cell adhesion molecules., Proceedings of the National Academy of Sciences, vol.80, issue.14, p.43844388, 1983.
DOI : 10.1073/pnas.80.14.4384

J. Veltmaat, C. Orelio, D. Oostwaard, M. Van-rooijen, C. Mummery et al., Snail is an immediate early target gene of parathyroid hormone related peptide signaling in parietal endoderm formation, Int J Dev Biol. avr, vol.44, issue.3, p.297307, 2000.

Y. Nakajima, T. Yamagishi, S. Hokari, and H. Nakamura, Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: Roles of transforming growth factor (TGF)-?? and bone morphogenetic protein (BMP), The Anatomical Record, vol.122, issue.2, p.119127, 2000.
DOI : 10.1002/(SICI)1097-0185(20000201)258:2<119::AID-AR1>3.0.CO;2-U

J. Rivera-feliciano, K. Lee, S. Kong, S. Rajagopal, Q. Ma et al., Development of heart valves requires Gata4 expression in endothelial-derived cells, Development, vol.133, issue.18, p.36073618, 2006.
DOI : 10.1242/dev.02519

A. Fischer, C. Steidl, T. Wagner, E. Lang, P. Jakob et al., Combined Loss of Hey1 and HeyL Causes Congenital Heart Defects Because of Impaired Epithelial to Mesenchymal Transition, Circulation Research, vol.100, issue.6, p.856863, 2007.
DOI : 10.1161/01.RES.0000260913.95642.3b

P. Savagner, D. Kusewitt, E. Carver, F. Magnino, C. Choi et al., Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes, Journal of Cellular Physiology, vol.154, issue.3, p.85866, 2005.
DOI : 10.1002/jcp.20188

URL : https://hal.archives-ouvertes.fr/inserm-00148036

M. Blanco, G. Moreno-bueno, D. Sarrio, A. Locascio, A. Cano et al., Correlation of Snail expression with histological grade and lymph node status in breast carcinomas, Oncogene, vol.21, issue.20, p.32413246, 2002.
DOI : 10.1038/sj.onc.1205416

T. Graham, H. Zhau, V. Odero-marah, A. Osunkoya, K. Kimbro et al., Insulin-like growth factor-l-dependent up-regulation of ZEB1 drives epithelial-tomesenchymal transition in human prostate cancer cells, Cancer Res. 1 avr, vol.68, issue.7, p.24792488, 2008.

J. Gotzmann, A. Fischer, M. Zojer, M. Mikula, V. Proell et al., A crucial function of PDGF in TGF-beta-mediated cancer progression of hepatocytes, Oncogene. 25 mai, vol.25, issue.22, p.31703185, 2006.

V. Vasko, A. Espinosa, W. Scouten, H. He, H. Auer et al., Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion, Proceedings of the National Academy of Sciences, vol.104, issue.8, p.28032808, 2007.
DOI : 10.1073/pnas.0610733104

T. Brabletz, A. Jung, S. Reu, M. Porzner, F. Hlubek et al., Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment, Proc Natl Acad Sci, vol.98, issue.18, p.1035610361, 2001.

C. Chung, J. Parker, K. Ely, J. Carter, Y. Yi et al., Gene expression profiles identify epithelial-to-mesenchymal transition and activation of nuclear factorkappa B signaling as characteristics of a high-risk head and neck squamous cell carcinoma, Cancer Res. 15 août, vol.66, issue.16, p.82108218, 2006.

S. Alonso, L. Tracey, P. Ortiz, B. Perez-gomez, J. Palacios et al., A highthroughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis, Cancer Res. 1 avr, vol.67, issue.7, p.34503460, 2007.

T. Hagemann, T. Bozanovic, S. Hooper, A. Ljubic, V. Slettenaar et al., Molecular profiling of cervical cancer progression, British Journal of Cancer, vol.9, issue.2, p.321328, 2007.
DOI : 10.1038/nrc798

K. Gravdal, O. Halvorsen, S. Haukaas, and L. Akslen, A switch from E-cadherin to Ncadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer, Clin Cancer Res. 1 déc, vol.13, issue.23, p.70037011, 2007.

. Ble, K. Chschmidt, S. Sassen, B. Schmalfeldt, T. Schuster et al., The E-cadherin repressor Snail is associated with lower overall survival of ovarian cancer patients, Br J Cancer. 22 janv, vol.98, issue.2, p.489495, 2008.

M. Yang, C. Chen, G. Chau, S. Chiou, C. Su et al., Comprehensive analysis of the independent effect of twist and snail in promoting metastasis of hepatocellular carcinoma, Hepatology, vol.65, issue.Pt 3, p.14641474, 2009.
DOI : 10.1002/hep.23221

M. Shioiri, T. Shida, K. Koda, K. Oda, K. Seike et al., Slug expression is an independent prognostic parameter for poor survival in colorectal carcinoma patients, British Journal of Cancer, vol.11, issue.12
DOI : 10.1016/S1368-8375(00)00059-2

Y. Kato, T. Hirano, K. Yoshida, K. Yashima, S. Akimoto et al., Frequent loss of E-cadherin and/or catenins in intrabronchial lesions during carcinogenesis of the bronchial epithelium, Lung Cancer, vol.48, issue.3, p.323330, 2005.
DOI : 10.1016/j.lungcan.2004.11.012

J. Shen, C. Behrens, I. Wistuba, L. Feng, J. Lee et al., Identification and Validation of Differences in Protein Levels in Normal, Premalignant, and Malignant Lung Cells and Tissues Using High-Throughput Western Array and Immunohistochemistry, Cancer Research, vol.66, issue.23, p.11194206, 2006.
DOI : 10.1158/0008-5472.CAN-04-1444

L. Prudkin, D. Liu, N. Ozburn, M. Sun, C. Behrens et al., Epithelial-tomesenchymal transition in the development and progression of adenocarcinoma and squamous cell carcinoma of the lung, Mod Pathol. mai, vol.22, issue.5, p.668678, 2009.

C. Tellez, D. Juri, K. Do, A. Bernauer, C. Thomas et al., EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic, Cancer Res. 15 avr, vol.71, issue.8, p.308797, 2011.
DOI : 10.1158/0008-5472.can-10-3035

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3078195

L. Ulianich, C. Garbi, A. Treglia, D. Punzi, C. Miele et al., ER stress is associated with dedifferentiation and an epithelial-to-mesenchymal transition-like phenotype in PC Cl3 thyroid cells, J Cell Sci. févr, vol.121, p.47786, 2008.

Q. Zhong, B. Zhou, D. Ann, P. Minoo, Y. Liu et al., Role of endoplasmic reticulum stress in epithelial-mesenchymal transition of, Am J Respir Cell Mol Biol. sept, vol.45, issue.3, p.498509, 2011.

J. Kononen, L. Bubendorf, A. Kallioniemi, M. Barlund, P. Schraml et al., Tissue microarrays for high-throughput molecular profiling of tumor specimens, Nature Medicine, vol.54, issue.7, p.844847, 1998.
DOI : 10.1023/A:1005769925670

L. Sobin, M. Gospodarowicz, and C. Wittekind, TNM classification of malignant tumours Disponible sur: Publisher description http, 2010.

N. Piton, Endoplasmic reticulum stress and the development of bronchial squamous cell carcinoma. [Cambridge, Royaume-Uni]: University of Cambridge, 2013.

D. Allred, J. Harvey, M. Berardo, and G. Clark, Prognostic and predictive factors in breast cancer by immunohistochemical analysis, Mod Pathol. févr, vol.11, issue.2, p.155168, 1998.

R. Mclean, W. Sanders, and W. Stroup, A unified approach to mixed linear models, Am Stat. févr, vol.45, issue.1, p.5464, 1991.

U. S. Institut-pierre-louis-d-'epidémiologie-et-de-santé-publique, M. Pierre, and . Curie, BiostaTGV -Statistiques en ligne [Internet]. [cité 15 juill 2014]. Disponible sur: http://marne.u707.jussieu.fr/biostatgv/?module=tests/spearman 148. Porter AM. Misuse of correlation and regression in three medical journals, J R Soc Med. mars, vol.92, issue.3, p.123128, 1999.

D. Hinkle, W. Wiersma, and S. Jurs, Applied Statistics for the Behavioral Sciences, Boston: Cengage Learning, 2002.

J. Gerdes, U. Schwab, H. Lemke, and H. Stein, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation, International Journal of Cancer, vol.18, issue.1, p.1320, 1983.
DOI : 10.1002/ijc.2910310104

M. Weigel and M. Dowsett, Current and emerging biomarkers in breast cancer: prognosis and prediction, Endocrine Related Cancer, vol.17, issue.4, p.24562, 2010.
DOI : 10.1677/ERC-10-0136

F. Lopez, F. Belloc, F. Lacombe, P. Dumain, J. Reiffers et al., Modalities of synthesis of Ki67 antigen during the stimulation of lymphocytes, Cytometry, vol.92, issue.1, p.429, 1991.
DOI : 10.1002/cyto.990120107

R. Jones, J. Salter, A. Hern, R. Nerurkar, A. Parton et al., The prognostic significance of Ki67 before and after neoadjuvant chemotherapy in breast cancer, Breast Cancer Research and Treatment, vol.100, issue.1, p.5368, 2009.
DOI : 10.1007/s10549-008-0081-7

URL : https://hal.archives-ouvertes.fr/hal-00478239

R. Yerushalmi, R. Woods, P. Ravdin, M. Hayes, and K. Gelmon, Ki67 in breast cancer: prognostic and predictive potential, The Lancet Oncology, vol.11, issue.2, p.174183, 2010.
DOI : 10.1016/S1470-2045(09)70262-1

M. Wheelock and K. Johnson, Cadherins as Modulators of Cellular Phenotype, Annual Review of Cell and Developmental Biology, vol.19, issue.1, p.20735, 2003.
DOI : 10.1146/annurev.cellbio.19.011102.111135

G. Christofori, Changing neig: HBours, changing behaviour: cell adhesion moleculemediated signalling during tumour progression, Embo J. 15 mai, vol.22, issue.10, p.231823, 2003.
DOI : 10.1093/emboj/cdg228

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC155986

R. Hazan, R. Qiao, R. Keren, I. Badano, and K. Suyama, Cadherin Switch in Tumor Progression, Annals of the New York Academy of Sciences, vol.59, issue.Pt 6, p.15563, 2004.
DOI : 10.1196/annals.1294.016

H. Peinado, D. Olmeda, and C. A. Snail, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype?, Nature Reviews Cancer, vol.176, issue.6, p.41528, 2007.
DOI : 10.1038/nrc2131

N. Roy, P. Bommi, U. Bhat, S. Bhattacharjee, I. Elangovan et al., DDB2 Suppresses Epithelial-to-Mesenchymal Transition in Colon Cancer, Cancer Research, vol.73, issue.12, p.377182, 2013.
DOI : 10.1158/0008-5472.CAN-12-4069

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3686976

F. Sharp and M. Bernaudin, HIF1 and oxygen sensing in the brain, Nature Reviews Neuroscience, vol.19, issue.6, p.43748, 2004.
DOI : 10.1128/MCB.23.1.359-369.2003

G. Wang, B. Jiang, E. Rue, and G. Semenza, Hypoxia-inducible factor 1 is a basichelix-loop-helix-PAS heterodimer regulated by cellular O2 tension, Proc Natl Acad Sci U A. 6 juin, vol.92, issue.12, p.55104, 1995.
DOI : 10.1073/pnas.92.12.5510

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC41725/pdf

R. Fukuda, K. Hirota, F. Fan, Y. Jung, L. Ellis et al., Insulin-like Growth Factor 1 Induces Hypoxia-inducible Factor 1-mediated Vascular Endothelial Growth Factor Expression, Which is Dependent on MAP Kinase and Phosphatidylinositol 3-Kinase Signaling in Colon Cancer Cells, Journal of Biological Chemistry, vol.277, issue.41, p.3820511, 2002.
DOI : 10.1074/jbc.M203781200

B. Jiang, G. Jiang, J. Zheng, Z. Lu, T. Hunter et al., Phosphatidylinositol 3-kinase signaling controls levels of hypoxia-inducible factor 1, Cell Growth Differ. juill, vol.12, issue.7, p.3639, 2001.

T. Okuda, T. Azuma, M. Ohtani, R. Masaki, Y. Ito et al., Hypoxia-inducible factor 1 alpha and vascular endothelial growth factor overexpression in ischemic colitis, World Journal of Gastroenterology, vol.11, issue.10
DOI : 10.3748/wjg.v11.i10.1535

M. Yang, M. Wu, S. Chiou, P. Chen, S. Chang et al., Direct regulation of TWIST by HIF-1alpha promotes metastasis, Nat Cell Biol. mars, vol.10, issue.3, p.295305, 2008.

P. Mak, I. Leav, B. Pursell, D. Bae, X. Yang et al., ERbeta impedes prostate cancer EMT by destabilizing HIF-1alpha and inhibiting VEGF-mediated snail nuclear localization: implications for Gleason grading, Cancer Cell. 13 avr, vol.17, issue.4, p.31932, 2010.

T. Imai, A. Horiuchi, C. Wang, K. Oka, S. Ohira et al., Hypoxia Attenuates the Expression of E-Cadherin via Up-Regulation of SNAIL in Ovarian Carcinoma Cells, The American Journal of Pathology, vol.163, issue.4, p.143747, 2003.
DOI : 10.1016/S0002-9440(10)63501-8

M. Hollander, Q. Zhan, I. Bae, and A. Fornace, Mammalian GADD34, an apoptosisand DNA damage-inducible gene, J Biol Chem. 23 mai, vol.272, issue.21, p.1373113737, 1997.
DOI : 10.1074/jbc.272.21.13731

H. Adler, R. Chinery, D. Wu, S. Kussick, J. Payne et al., Leukemic HRX Fusion Proteins Inhibit GADD34-Induced Apoptosis and Associate with the GADD34 and hSNF5/INI1 Proteins, Molecular and Cellular Biology, vol.19, issue.10, p.70507060, 1999.
DOI : 10.1128/MCB.19.10.7050

K. Minami, H. Inoue, T. Terashita, T. Kawakami, R. Watanabe et al., GADD34 induces p21 expression and cellular senescence, Oncology Reports, vol.17, issue.6, p.14811485, 2007.
DOI : 10.3892/or.17.6.1481

C. Tanaka, S. Ito, N. Nishio, Y. Kodera, H. Sakurai et al., GADD34 suppresses wound healing by upregulating expression of myosin IIA, Transgenic Research, vol.61, issue.4, p.637645, 2010.
DOI : 10.1007/s11248-009-9347-z

A. Knudson, . Mutation, . Cancer-statistical, . Study, and . Retinoblastoma, Mutation and Cancer: Statistical Study of Retinoblastoma, Proceedings of the National Academy of Sciences, vol.68, issue.4, p.820, 1971.
DOI : 10.1073/pnas.68.4.820

M. Fero, E. Randel, K. Gurley, J. Roberts, and C. Kemp, The murine gene p27(Kip1) is haplo-insufficient for tumour suppression, Nature. 12 nov, vol.396, issue.6707, p.177180, 1998.

A. Willis, E. Jung, T. Wakefield, and X. Chen, Mutant p53 exerts a dominant negative effect by preventing wild-type p53 from binding to the promoter of its target genes, Oncogene, vol.23, issue.13, p.23302338, 2004.
DOI : 10.1038/sj.onc.1207396

J. Issa, Opinion: CpG island methylator phenotype in cancer, Nature Reviews Cancer, vol.10, issue.12, p.988993, 2004.
DOI : 10.1016/S0092-8674(00)81333-1

S. Baylin and P. Jones, A decade of exploring the cancer epigenome ??? biological and translational implications, Nature Reviews Cancer, vol.9, issue.16, p.726734, 2011.
DOI : 10.1038/nrc3130

D. Wang, C. Qiu, H. Zhang, J. Wang, Q. Cui et al., Human MicroRNA Oncogenes and Tumor Suppressors Show Significantly Different Biological Patterns: From Functions to Targets, PLoS ONE, vol.5, issue.9, 2010.
DOI : 10.1371/journal.pone.0013067.s008

URL : http://doi.org/10.1371/journal.pone.0013067

W. Grady, L. Myeroff, S. Swinler, A. Rajput, S. Thiagalingam et al., Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers, Cancer Res. 15 janv, vol.59, issue.2, p.320324, 1999.

T. He, A. Sparks, C. Rago, H. Hermeking, L. Zawel et al., Identification of c-MYC as a Target of the APC Pathway, Science, vol.281, issue.5382, p.15091512, 1998.
DOI : 10.1126/science.281.5382.1509

A. Dulak, S. Schumacher, J. Van-lieshout, Y. Imamura, C. Fox et al., Gastrointestinal Adenocarcinomas of the Esophagus, Stomach, and Colon Exhibit Distinct Patterns of Genome Instability and Oncogenesis, Cancer Research, vol.72, issue.17, p.43834393, 2012.
DOI : 10.1158/0008-5472.CAN-11-3893

M. Ilyas, H. Grabsch, I. Ellis, C. Womack, R. Brown et al., Guidelines and considerations for conducting experiments using tissue microarrays, Histopathology, vol.10, issue.6, p.82739, 2013.
DOI : 10.1111/his.12118

B. Glimelius, Neo-adjuvant radiotherapy in rectal cancer, World Journal of Gastroenterology, vol.19, issue.46, p.8489501, 2013.
DOI : 10.3748/wjg.v19.i46.8489

R. Panganiban, O. Mungunsukh, and R. Day, X-irradiation induces ER stress, apoptosis, and senescence in pulmonary artery endothelial cells, International Journal of Radiation Biology, vol.86, issue.8, p.65667, 2013.
DOI : 10.4161/cbt.8.9.8143

B. Zhang, Y. Wang, X. Pang, Y. Su, A. G. Wang et al., ER stress induced by ionising radiation in IEC-6 cells, International Journal of Radiation Biology, vol.49, issue.6, p.42935, 2010.
DOI : 10.1128/MCB.21.4.1249-1259.2001

K. Graham, A. De-las-morenas, A. Tripathi, C. King, M. Kavanah et al., Gene expression in histologically normal epithelium from breast cancer patients and from cancer-free prophylactic mastectomy patients shares a similar profile, British Journal of Cancer, vol.97, issue.8, p.128493, 2010.
DOI : 10.1002/cncr.24129

L. Schmidt, S. Biesterfeld, A. Kummel, A. Faldum, M. Sebastian et al., Tissue microarrays are reliable tools for the clinicopathological characterization of lung cancer tissue, Anticancer Res. janv, vol.29, issue.1, p.2019, 2009.

D. Gregorio, C. Losi, L. Fante, R. Modica, S. Ghidoni et al., Histology of aberrant crypt foci in the human colon, Histopathology, vol.30, issue.4, p.32834, 1997.
DOI : 10.1046/j.1365-2559.1997.d01-626.x

E. Luporsi, F. André, F. Spyratos, P. Martin, J. Jacquemier et al., Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review, Breast Cancer Research and Treatment, vol.63, issue.Suppl, p.895915, 2012.
DOI : 10.1007/s10549-011-1837-z

R. Stuart-harris, C. Caldas, S. Pinder, and P. Pharoah, Proliferation markers and survival in early breast cancer: A systematic review and meta-analysis of 85 studies in 32,825 patients, The Breast, vol.17, issue.4, p.323334, 2008.
DOI : 10.1016/j.breast.2008.02.002

N. Mucci, G. Akdas, S. Manely, and M. Rubin, Neuroendocrine expression in metastatic prostate cancer: Evaluation of high throughput tissue microarrays to detect heterogeneous protein expression, Hum Pathol. avr, vol.31, issue.4, p.406414, 2000.

R. Camp, L. Charette, and D. Rimm, Validation of Tissue Microarray Technology in Breast Carcinoma, Laboratory Investigation, vol.59, issue.12, 2000.
DOI : 10.1093/jnci/92.11.937

A. Nocito, L. Bubendorf, E. Tinner, K. Suess, U. Wagner et al., Microarrays of bladder cancer tissue are highly representative of proliferation index and histological grade, The Journal of Pathology, vol.5, issue.3, p.349357, 2001.
DOI : 10.1002/1096-9896(200107)194:3<349::AID-PATH887>3.0.CO;2-D

N. Goldstein, M. Ferkowicz, E. Odish, A. Mani, and F. Hastah, Minimum Formalin Fixation Time for Consistent Estrogen Receptor Immunohistochemical Staining of Invasive Breast Carcinoma, American Journal of Clinical Pathology, vol.120, issue.1, p.8692, 2003.
DOI : 10.1309/QPHDRB00QXGMUQ9N

D. Marzo, A. Fedor, H. Gage, W. Rubin, and M. , Inadequate formalin fixation decreases reliability of p27(Kip1) immunohistochemical staining: Probing optimal fixation time using high-density tissue microarrays, Hum Pathol. juill, vol.33, issue.7, p.756760, 2002.

T. Khoury, S. Sait, H. Hwang, R. Chandrasekhar, G. Wilding et al., Delay to formalin fixation effect on breast biomarkers, Modern Pathology, vol.12, issue.11, p.14571467, 2009.
DOI : 10.1097/00000478-200007000-00014

W. Fadhil and M. Ilyas, Immunostaining for mismatch repair (MMR) protein expression in colorectal cancer is better and easier to interpret when performed on diagnostic biopsies, Histopathology, vol.56, issue.4, p.653655, 2012.
DOI : 10.1111/j.1365-2559.2011.04021.x

G. Mann, V. Fahey, F. Feleppa, and M. Buchanan, Reliance on Hormone Receptor Assays of Surgical Specimens May Compromise Outcome in Patients With Breast Cancer, Journal of Clinical Oncology, vol.23, issue.22, p.51485154, 2005.
DOI : 10.1200/JCO.2005.02.076

J. Huang, R. Qi, J. Quackenbush, E. Dauway, E. Lazaridis et al., Effects of Ischemia on Gene Expression, Journal of Surgical Research, vol.99, issue.2, p.222227, 2001.
DOI : 10.1006/jsre.2001.6195

E. Obeng, L. Carlson, D. Gutman, W. Harrington, . Jr et al., Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells, Blood, vol.107, issue.12, p.490716, 2006.
DOI : 10.1182/blood-2005-08-3531

T. Pitts, M. Morrow, S. Kaufman, J. Tentler, and S. Eckhardt, Vorinostat and bortezomib exert synergistic antiproliferative and proapoptotic effects in colon cancer cell models, Molecular Cancer Therapeutics, vol.8, issue.2, p.3429, 2009.
DOI : 10.1158/1535-7163.MCT-08-0534

I. Voutsadakis, A. Patrikidou, K. Tsapakidis, A. Karagiannaki, E. Hatzidaki et al., Additive inhibition of colorectal cancer cell lines by aspirin and bortezomib, International Journal of Colorectal Disease, vol.11, issue.7, p.795804, 2010.
DOI : 10.1007/s00384-010-0939-0

H. Mackay, D. Hedley, P. Major, C. Townsley, M. Mackenzie et al., A Phase II Trial with Pharmacodynamic Endpoints of the Proteasome Inhibitor Bortezomib in Patients with Metastatic Colorectal Cancer, Clinical Cancer Research, vol.11, issue.15, p.552633, 2005.
DOI : 10.1158/1078-0432.CCR-05-0081

D. Schewe and J. Aguirre-ghiso, Inhibition of eIF2alpha dephosphorylation maximizes bortezomib efficiency and eliminates quiescent multiple myeloma cells surviving proteasome inhibitor therapy, Cancer Res. 15 févr, vol.69, issue.4, p.154552, 2009.