D. Hanahan and L. Coussens, Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment, Cancer Cell, vol.21, issue.3, pp.309-322, 2012.
DOI : 10.1016/j.ccr.2012.02.022

M. Pein and T. Oskarsson, Microenvironment in metastasis: roadblocks and supportive niches, American Journal of Physiology-Cell Physiology, vol.59, issue.10, pp.627-638, 2015.
DOI : 10.1016/j.ccr.2009.05.017

URL : http://ajpcell.physiology.org/content/ajpcell/309/10/C627.full.pdf

O. Finn, Immuno-oncology: understanding the function and dysfunction of the immune system in cancer, Annals of Oncology, vol.6, issue.3, pp.6-9, 2012.
DOI : 10.1038/nrc1815

A. Johansson, J. Hamzah, and R. Ganss, More than a scaffold: Stromal modulation of tumor immunity, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol.1865, issue.1, pp.3-13, 2016.
DOI : 10.1016/j.bbcan.2015.06.001

R. Offringa, CANCER: Enhanced: Cancer Immunotherapy Is More Than a Numbers Game, Science, vol.314, issue.5796, pp.68-69, 2006.
DOI : 10.1126/science.1133893

A. Schientiger, M. Philip, V. Krisnawan, E. Chiu, J. Delrow et al., Antigen-driven differentiation program initiated early during tumorigenesis, Immunity, vol.45, pp.389-401, 2016.

J. Joyce and D. Fearon, T cell exclusion, immune privilege, and the tumor microenvironment, Science, vol.91, issue.9, pp.74-80, 2015.
DOI : 10.1073/pnas.1211850110

D. Gabrilovich, S. Ostrand-rosenberg, and V. Bronte, Coordinated regulation of myeloid cells by tumours, Nature Reviews Immunology, vol.205, issue.4, pp.253-268, 2012.
DOI : 10.1084/jem.20080108

M. Galdiero, C. Garlanda, S. Jaillon, G. Marone, and A. Mantovani, Tumor associated macrophages and neutrophils in tumor progression, Journal of Cellular Physiology, vol.21, issue.7, pp.1404-1412, 2013.
DOI : 10.1016/j.cytogfr.2009.11.009

A. Chow, B. Brown, and M. Merad, Studying the mononuclear phagocyte system in the molecular age, Nature Reviews Immunology, vol.83, issue.11, pp.788-798, 2011.
DOI : 10.4049/jimmunol.176.4.2465

V. Kumar, P. Cheng, T. Condamine, S. Mony, L. Languino et al., CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation, Immunity, vol.44, issue.2, pp.303-315, 2008.
DOI : 10.1016/j.immuni.2016.01.014

URL : https://doi.org/10.1016/j.immuni.2016.01.014

A. Mantovani, Reflections on immunological nomenclature: in praise of imperfection, Nature Immunology, vol.25, issue.3, pp.215-216, 2016.
DOI : 10.1016/j.it.2004.09.015

F. Ginhoux, J. Schultze, P. Murray, J. Ochando, and S. Biswas, New insights into the multidimensional concept of macrophage ontogeny, activation and function, Nature Immunology, vol.332, issue.1, pp.34-40, 2016.
DOI : 10.1126/science.1198704

D. Roulois, S. Deshayes, M. Guilly, J. Nader, C. Liddell et al., Characterization of preneoplastic and neoplastic rat mesothelial cell lines: the involvement of TETs, DNMTs, and 5-hydroxymethylcytosine, Oncotarget, vol.7, issue.23, pp.34664-346687, 2016.
DOI : 10.18632/oncotarget.8970

URL : https://hal.archives-ouvertes.fr/inserm-01306838

Y. Gao, X. Wang, Z. Sang, Z. Li, F. Liu et al., Quantitative proteomics by SWATH-MS reveals sophisticated metabolic reprogramming in hepatocellular carcinoma tissues. Sci Rep, p.45913, 2017.
DOI : 10.1038/srep45913

URL : http://www.nature.com/articles/srep45913.pdf

S. Anjo, C. Santa, and B. Manadas, SWATH-MS as a tool for biomarker discovery: From basic research to clinical applications, PROTEOMICS, vol.13, issue.3-4, 2017.
DOI : 10.1074/mcp.O114.046102

L. Song, J. Tang, L. Owusu, M. Sun, J. Wu et al., Galectin-3 in cancer, Clinica Chimica Acta, vol.431, pp.185-191, 2014.
DOI : 10.1016/j.cca.2014.01.019

A. Fortuna-costa, A. Gomes, E. Kozlowski, M. Stelling, and M. Pavão, Extracellular Galectin-3 in Tumor Progression and Metastasis, Frontiers in Oncology, vol.945, issue.4, p.138, 2014.
DOI : 10.1007/978-1-62703-125-7_13

URL : https://doi.org/10.3389/fonc.2014.00138

A. Cardoso, L. Andrade, S. Bustos, and R. Chammas, Galectin-3 determines tumor cell adaptative strategies in stressed tumor microenvironments, Front Oncol, vol.6, p.127, 2016.
DOI : 10.3389/fonc.2016.00127

URL : https://www.frontiersin.org/articles/10.3389/fonc.2016.00127/pdf

P. Ruvolo, Galectin 3 as a guardian of the tumor microenvironment, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1863, issue.3, pp.427-437, 2016.
DOI : 10.1016/j.bbamcr.2015.08.008

S. Xu and M. Powers, Nuclear pore proteins and cancer, Seminars in Cell & Developmental Biology, vol.20, issue.5, pp.620-630, 2009.
DOI : 10.1016/j.semcdb.2009.03.003

URL : http://europepmc.org/articles/pmc2706781?pdf=render

V. Khachaturov, G. Xiao, Y. Kinoshita, P. Unger, and D. Burstein, Histone H1.5, a novel prostatic cancer marker: an immunohistochemical study, Human Pathology, vol.45, issue.10, pp.2115-2119, 2014.
DOI : 10.1016/j.humpath.2014.06.015

Y. Cao, H. Liang, F. Zhang, Z. Luan, S. Zhao et al., Prohibitin overexpression predicts poor prognosis and promotes cell proliferation and invasion through ERK pathway activation in gallbladder cancer, Journal of Experimental & Clinical Cancer Research, vol.21, issue.55, p.68, 2016.
DOI : 10.1038/sj.onc.1205944

URL : https://jeccr.biomedcentral.com/track/pdf/10.1186/s13046-016-0346-7?site=jeccr.biomedcentral.com

B. Peng, C. Guo, H. Guan, S. Liu, and M. Sun, Annexin A5 as a potential marker in tumors, Clinica Chimica Acta, vol.427, pp.42-48, 2014.
DOI : 10.1016/j.cca.2013.09.048

A. Bresnick, D. Weber, and D. Zimmer, S100 proteins in cancer, Nature Reviews Cancer, vol.423, issue.2, pp.96-109, 2015.
DOI : 10.1016/j.jmb.2012.07.011

URL : http://europepmc.org/articles/pmc4369764?pdf=render

W. Liu, M. Yan, and A. Chan, A thirty-year quest for a role of R-Ras in cancer: from an oncogene to a multitasking GTPase, Cancer Letters, vol.403, pp.862-865, 2017.
DOI : 10.1016/j.canlet.2017.06.003

S. Zecchini and U. Cavallaro, Neural cell adhesion molecule in cancer: expression and mechanisms Structure and function of the neural cell adhesion molecule NCAM, Advances in experimental medicine and biology, pp.319-333, 2010.

F. Wang, Y. Zheng, M. Orange, C. Yang, Y. B. Liu et al., PTRF suppresses the progression of colorectal cancers, Oncotarget, vol.8, issue.30, pp.48650-48659, 2017.
DOI : 10.18632/oncotarget.9424

J. Glaser, M. Neumann, Q. Mei, B. Betz, N. Seier et al., Macrophage capping protein Cap Gis a putative oncogene involved in migration and invasiveness in ovarian carcinoma, BioMed Res Int, vol.2014, p.379847, 2014.

I. Park, C. Han, J. S. Lee, B. Choi, H. Kwon et al., Myosin regulatory light chains are required to maintain the stability of myosin II and cellular integrity, Biochemical Journal, vol.261, issue.1, pp.171-180, 2011.
DOI : 10.1002/cm.20148

V. Karantza, Keratins in health and cancer: more than mere epithelial cell markers, Oncogene, vol.156, issue.2, pp.127-138, 2011.
DOI : 10.1016/S0002-9440(10)64997-8

URL : http://www.nature.com/onc/journal/v30/n2/pdf/onc2010456a.pdf

I. Stefansson, H. Salvesen, and L. Akslen, Loss of p63 and cytokeratin 5/6 expression is associated with more aggressive tumors in endometrial carcinoma patients, International Journal of Cancer, vol.203, issue.5, pp.1227-1233, 2006.
DOI : 10.1002/path.1559

D. Pouliquen, B. Nawrocki-raby, J. Nader, S. Blandin, M. Robard et al., Evaluation of intracavitary administration of curcumin for the treatment of sarcomatoid mesothelioma, Oncotarget, vol.8, issue.34, pp.57552-57573, 2017.
DOI : 10.18632/oncotarget.15744

URL : https://hal.archives-ouvertes.fr/inserm-01480303

P. Gomez-suaga, S. Paillusson, and C. Miller, ER-mitochondria signaling regulates autophagy, Autophagy, vol.13, issue.7, pp.1250-1251, 2017.
DOI : 10.1080/15548627.2017.1317913

URL : http://europepmc.org/articles/pmc5529068?pdf=render

M. Gorjanacz, Nuclear assembly as a target for anti-cancer therapies, Nucleus, vol.7, issue.1, pp.47-55, 2014.
DOI : 10.1007/s00412-007-0094-8

J. Li, T. Wang, L. Pei, J. Jing, W. Hu et al., Expression of VRK1 and the downstream gene BANF1 in esophageal cancer, Biomedicine & Pharmacotherapy, vol.89, pp.1086-1091, 2017.
DOI : 10.1016/j.biopha.2017.02.095

S. Han, Y. Tang, and R. Smith, Functional diversity of the hnRNPs: past, present and perspectives, Biochemical Journal, vol.16, issue.3, pp.379-392, 2010.
DOI : 10.1101/gad.1934910

P. Thomas, R. Forse, and O. Bajenova, Carcinoembryonic antigen (CEA) and its receptor hnRNP M are mediators of metastasis and the inflammatory response in the liver, Clinical & Experimental Metastasis, vol.13, issue.8, pp.923-932, 2011.
DOI : 10.1158/0008-5472.CAN-10-1073

X. Zhou, W. Liao, J. Liao, P. Liao, and H. Lu, Ribosomal proteins: functions beyond the ribosome, Journal of Molecular Cell Biology, vol.35, issue.(Suppl), pp.92-104, 2015.
DOI : 10.1016/j.molcel.2009.07.014

URL : https://academic.oup.com/jmcb/article-pdf/7/2/92/11735023/mjv014.pdf

M. Ali, U. Rahman, M. Jia, Z. Jiang, and C. , Eukaryotic translation initiation factors and cancer, Tumor Biology, vol.13, issue.6, p.1010428317709805, 2017.
DOI : 10.1016/j.chembiol.2006.10.005

S. Stöcker, K. Van-laer, A. Mijuskovic, and T. Dick, The Conundrum of Hydrogen Peroxide Signaling and the Emerging Role of Peroxiredoxins as Redox Relay Hubs, Antioxidants & Redox Signaling, vol.28, issue.7, 2017.
DOI : 10.1089/ars.2017.7162

A. Nicolussi, D. Inzeo, S. Capalbo, C. Giannini, G. Coppa et al., The role of peroxiredoxins in cancer, Molecular and Clinical Oncology, vol.6, issue.2, pp.139-153, 2017.
DOI : 10.3892/mco.2017.1129

A. Arrigo, Mammalian HspB1 (Hsp27) is a molecular sensor linked to the physiology and environment of the cell, Cell Stress and Chaperones, vol.21, issue.9, pp.517-529, 2017.
DOI : 10.1093/emboj/cdf528

C. Geisler, N. Gaisa, D. Pfister, S. Fuessel, G. Kristiansen et al., Identification and Validation of Potential New Biomarkers for Prostate Cancer Diagnosis and Prognosis Using 2D-DIGE and MS, BioMed Research International, vol.18, issue.4, p.454256, 2015.
DOI : 10.1002/pmic.200800298

I. Nimmrich, S. Erdmann, U. Melchers, S. Chtarbova, U. Finke et al., The novel ependymin related gene UCC1 is highly expressed in colorectal tumor cells, Cancer Letters, vol.165, issue.1, pp.71-79, 2001.
DOI : 10.1016/S0304-3835(01)00390-1

Q. Zhang and Y. Wang, HMG modifications and nuclear function, Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, vol.1799, issue.1-2, p.28, 2010.
DOI : 10.1016/j.bbagrm.2009.11.009

URL : http://europepmc.org/articles/pmc2818492?pdf=render

S. Chew, Y. Okazaki, H. Nagai, N. Misawa, S. Akatsuka et al., Cancer-promoting role of adipocytes in asbestos-induced mesothelial carcinogenesis through dysregulated adipocytokine production, Carcinogenesis, vol.35, issue.1, pp.164-172, 2014.
DOI : 10.1056/NEJMoa051185

Y. Zhang, Y. Sun, F. Yan, Q. Li, Y. Zhang et al., Fatty Acid-Binding Protein E-FABP Restricts Tumor Growth by Promoting IFN-?? Responses in Tumor-Associated Macrophages, Cancer Research, vol.74, issue.11, pp.2986-2998, 2014.
DOI : 10.1158/0008-5472.CAN-13-2689

S. Mishra and B. Nyomba, Prohibitin ??? At the crossroads of obesity-linked diabetes and cancer, Experimental Biology and Medicine, vol.17, issue.11, pp.1170-1177, 2017.
DOI : 10.1126/science.1260825

J. Rho, J. Ladd, C. Li, J. Potter, Y. Zhang et al., Protein and glycomic plasma markers for early detection of adenoma and colon cancer, Gut, vol.67, issue.3, p.312794, 2016.
DOI : 10.1136/gutjnl-2016-312794

M. Mendes, A. Peláez-garcía, M. López-lucendo, R. Bartolomé, E. Calviño et al., Mapping the Spatial Proteome of Metastatic Cells in Colorectal Cancer, PROTEOMICS, vol.10, issue.19, 2017.
DOI : 10.1371/journal.pone.0140870

E. Günther and L. Walter, The major histocompatibility complex of the rat (Rattus norvegicus), Immunogenetics, vol.53, issue.7, pp.520-542, 2001.
DOI : 10.1007/s002510100361

P. Leone, E. Shin, F. Perosa, A. Vacca, F. Dammaco et al., MHC Class I Antigen Processing and Presenting Machinery: Organization, Function, and Defects in Tumor Cells, JNCI Journal of the National Cancer Institute, vol.88, issue.6, pp.1172-1187, 2013.
DOI : 10.1182/blood.V99.10.3661

URL : https://academic.oup.com/jnci/article-pdf/105/16/1172/7675900/djt184.pdf

S. Filén and R. Lahesmaa, GIMAP Proteins in T-Lymphocytes, Journal of Signal Transduction, vol.162, issue.7, p.268589, 2010.
DOI : 10.1038/sj.gene.6364413

Z. Huang, W. Zhang, C. Gao, J. B. , C. X. Zheng et al., Dysregulation of GTPase IMAP family members in hepatocellular cancer, Molecular Medicine Reports, vol.14, issue.5, pp.4119-4123, 2016.
DOI : 10.3892/mmr.2016.5764

E. Kim, H. Kwak, and K. Ahn, Cytosolic Aminopeptidases Influence MHC Class I-Mediated Antigen Presentation in an Allele-Dependent Manner, The Journal of Immunology, vol.183, issue.11, pp.7379-7387, 2009.
DOI : 10.4049/jimmunol.0901489

URL : http://www.jimmunol.org/content/jimmunol/183/11/7379.full.pdf

M. Maeurer, S. Gollin, W. Storkus, W. Swaney, J. Karbach et al., Tumor escape from immune recognition: loss of HLA- A2 melanoma cell surface expression is associated with a complex rearrangement of the short arm of chromosome 6, Clin Cancer Res, vol.2, pp.641-652, 1996.

U. Blank, N. Charles, and M. Benhamou, The high-affinity immunoglobulin E receptor as pharmacological target, European Journal of Pharmacology, vol.778, pp.24-32, 2016.
DOI : 10.1016/j.ejphar.2015.05.070

M. Wieczorek, E. Abualrous, J. Sticht, M. Alvaro-benito, S. Stolzenberg et al., Major Histocompatibility Complex (MHC) Class I and MHC Class II Proteins: Conformational Plasticity in Antigen Presentation, Frontiers in Immunology, vol.110, p.292, 2017.
DOI : 10.1073/pnas.1305688110

Y. Jia, Z. Xiao, W. Jiang, G. Chen, and Z. Wang, Overexpression of IFITM3 predicts poor prognosis in stage IIA esophageal squamous cell carcinoma after Ivor Lewis esophagectomy, Thoracic Cancer, vol.23, issue.6, pp.592-599, 2017.
DOI : 10.1245/s10434-015-4622-0

D. Zhang, H. Wang, H. He, H. Niu, and Y. Li, Interferon induced transmembrane protein 3 regulates the growth and invasion of human lung adenocarcinoma, Thoracic Cancer, vol.31, issue.4, pp.337-343, 2017.
DOI : 10.1089/jir.2010.0134

V. Rolih, G. Barutello, S. Lussich, D. Maria, R. Quaglino et al., CSPG4: a prototype oncoantigen for translational immunotherapy studies, Journal of Translational Medicine, vol.70, issue.48, p.151, 2017.
DOI : 10.1158/0008-5472.CAN-09-2548

URL : https://translational-medicine.biomedcentral.com/track/pdf/10.1186/s12967-017-1250-4?site=translational-medicine.biomedcentral.com

S. Ocak, D. Friedman, H. Chen, J. Ausborn, M. Hassanein et al., Discovery of New Membrane-Associated Proteins Overexpressed in Small-Cell Lung Cancer, Journal of Thoracic Oncology, vol.9, issue.3, pp.324-336, 2014.
DOI : 10.1097/JTO.0000000000000090

K. Wang, B. Seo, C. Fischbach, and D. Gourdon, Fibronectin Mechanobiology Regulates Tumorigenesis, Cellular and Molecular Bioengineering, vol.141, issue.4, pp.1-11, 2016.
DOI : 10.1083/jcb.141.2.539

URL : https://link.springer.com/content/pdf/10.1007%2Fs12195-015-0417-4.pdf

Y. Xie, L. Chen, X. Lv, G. Hou, Y. Wang et al., The levels of serine proteases in colon tissue interstitial fluid and serum serve as an indicator of colorectal cancer progression, Oncotarget, vol.7, issue.22, pp.32592-32606, 2016.
DOI : 10.18632/oncotarget.8693

B. Mondal, V. Patil, S. Shwetha, K. Sravani, A. Hegde et al., Integrative functional genomic analysis identifies epigenetically regulated fibromodulin as an essential gene for glioma cell migration, Oncogene, vol.25, issue.1, pp.71-83, 2017.
DOI : 10.1371/journal.pone.0060335

A. Bergamaschi, E. Tagliabue, T. Sørlie, B. Naume, T. Triulzi et al., Extracellular matrix signature identifies breast cancer subgroups with different clinical outcome, The Journal of Pathology, vol.102, issue.3, pp.357-367, 2008.
DOI : 10.1073/pnas.091062498

G. Lal, S. Hashimi, B. Smith, C. Lynch, L. Zhang et al., Extracellular Matrix 1 (ECM1) Expression Is a Novel Prognostic Marker for Poor Long-Term Survival in Breast Cancer: A Hospital-Based Cohort Study in Iowa, Annals of Surgical Oncology, vol.24, issue.8, pp.2280-2287, 2009.
DOI : 10.1016/S0140-6736(05)17947-1

M. Krisenko, A. Cartagena, A. Raman, and R. Geahlen, Nanomechanical Property Maps of Breast Cancer Cells As Determined by Multiharmonic Atomic Force Microscopy Reveal Syk-Dependent Changes in Microtubule Stability Mediated by MAP1B, Biochemistry, vol.54, issue.1, pp.60-68, 2014.
DOI : 10.1021/bi500325n

D. Hoshino, K. Kirkbride, K. Costello, E. Clark, S. Sinha et al., Exosome Secretion Is Enhanced by Invadopodia and Drives Invasive Behavior, Cell Reports, vol.5, issue.5, pp.1159-1168, 2013.
DOI : 10.1016/j.celrep.2013.10.050

URL : https://doi.org/10.1016/j.celrep.2013.10.050

C. Yau, L. Esserman, D. Moore, F. Waldman, J. Sninsky et al., A multigene predictor of metastatic outcome in early stage hormone receptor-negative and triple-negative breast cancer, Breast Cancer Research, vol.69, issue.5, p.85, 2010.
DOI : 10.1158/0008-5472.CAN-08-3564

J. Ouderkirk and M. Krendel, Non-muscle myosins in tumor progression, cancer cell invasion, and metastasis, Cytoskeleton, vol.6, issue.6, pp.447-463, 2014.
DOI : 10.1038/ncb1136

URL : http://onlinelibrary.wiley.com/doi/10.1002/cm.21187/pdf

U. Blank, N. Charles, and M. Benhamou, The high-affinity immunoglobulin E receptor as pharmacological target, European Journal of Pharmacology, vol.778, pp.24-32, 2016.
DOI : 10.1016/j.ejphar.2015.05.070

I. Mauldin, N. Wages, A. Stowman, E. Wang, M. Smolkin et al., Intratumoral interferon-gamma increases chemokine production but fails to increase T cell infiltration of human melanoma metastases, Cancer Immunology, Immunotherapy, vol.523, issue.7559, pp.1189-1199, 2016.
DOI : 10.1038/nature14404

D. Wågsäter, S. Löfgren, A. Hugander, O. Dienus, and J. Dimberg, Analysis of single nucleotide polymorphism in the promoter and protein expression of the chemokine Eotaxin-1 in colorectal cancer patients, World Journal of Surgical Oncology, vol.5, issue.1, p.84, 2007.
DOI : 10.1186/1477-7819-5-84

C. Loveridge, E. Mui, R. Patel, E. Tan, I. Ahmad et al., -Deficient Mouse Model of Prostate Carcinogenesis, Cancer Research, vol.77, issue.12, pp.3158-3168, 2017.
DOI : 10.1158/0008-5472.CAN-16-2565

J. Fritz, M. Tennis, D. Orlicky, H. Yin, C. Ju et al., Depletion of tumorassociated macrophages slows the growth of chemically induced mouse lung adenocarcinomas Induction of monocyte chemoattractant proteins in macrophages via the production of granulocyte/macrophage colony-stimulating factor by breast cancer cells, Front Immunol. Front Immunol, vol.5, issue.7 2, 2014.

J. Bourguignon, H. Borghi, R. Sesboüé, M. Diarra-mehrpour, J. Bernaudin et al., Immunohistochemical Distribution of Inter-??-trypsin Inhibitor Chains in Normal and Malignant Human Lung Tissue, Journal of Histochemistry & Cytochemistry, vol.44, issue.12, pp.1625-1632, 1999.
DOI : 10.1002/1097-0142(19890815)64:4<860::AID-CNCR2820640417>3.0.CO;2-7

G. Toledo, A. Nilsson, J. Noborn, F. Sihlbom, C. Larson et al., Positive mode LC-MS/MS analysis of chondroitin sulfate modified glycopeptides derived from light and heavy chains of the human inter-?-trypsin inhibitor complex, Mol Cell Proteomics, vol.1412, pp.3118-3131, 2015.

Z. Yang, D. Ho, C. Lau, C. Lam, C. Lum et al., Allograft inflammatory factor-1 (AIF-1) is crucial for the survival and pro-inflammatory activity of macrophages, International Immunology, vol.17, issue.11, pp.1391-1397, 2005.
DOI : 10.1093/emboj/cdg231

P. Yang, H. An, X. Liu, M. Wen, Y. Zheng et al., The cytosolic nucleic acid sensor LRRFIP1 mediates the production of type I interferon via a ??-catenin-dependent pathway, Nature Immunology, vol.26, issue.6, pp.487-495, 2010.
DOI : 10.1038/labinvest.3700497

D. Madsen, D. Leonard, A. Masedunskas, A. Moyer, H. Jürgensen et al., M2-like macrophages are responsible for collagen degradation through a mannose receptor???mediated pathway, The Journal of Cell Biology, vol.113, issue.6, pp.951-966, 2013.
DOI : 10.1083/jcb.201301081.dv

URL : http://europepmc.org/articles/pmc3776354?pdf=render