C. Rouzer and L. Marnett, Cyclooxygenases: structural and functional insights, The Journal of Lipid Research, vol.50, issue.Supplement, pp.29-34, 2009.
DOI : 10.1194/jlr.R800042-JLR200

N. Chandrasekharan, H. Dai, K. Roos, N. Evanson, and J. Tomsik, COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression, Proceedings of the National Academy of Sciences, vol.99, issue.21, pp.13926-13931, 2002.
DOI : 10.1073/pnas.162468699

B. Kis, J. Snipes, T. Isse, K. Nagy, and D. Busija, Putative Cyclooxygenase-3 Expression in Rat Brain Cells, Journal of Cerebral Blood Flow & Metabolism, vol.361, pp.1287-1292, 2003.
DOI : 10.1097/01.WCB.0000090681.07515.81

L. Crofford, COX-1 and COX-2 tissue expression: implications and predictions, J Rheumatol, vol.49, pp.15-19, 1997.

P. Kam and A. See, Cyclo-oxygenase isoenzymes: physiological and pharmacological role, Anaesthesia, vol.93, issue.(Suppl. 9), pp.442-449, 2000.
DOI : 10.1046/j.1365-2044.2000.01271.x

J. Martel-pelletier, J. Pelletier, and H. Fahmi, Cyclooxygenase-2 and prostaglandins in articular tissues, Seminars in Arthritis and Rheumatism, vol.33, issue.3, pp.155-167, 2003.
DOI : 10.1016/S0049-0172(03)00134-3

L. Zhang, A. Bertucci, K. Smith, L. Xu, and S. Datta, Hyperexpression of cyclooxygenase 2 in the lupus immune system and effect of cyclooxygenase 2 inhibitor diet therapy in a murine model of systemic lupus erythematosus, Arthritis & Rheumatism, vol.173, issue.12, pp.4132-4141, 2007.
DOI : 10.1002/art.23054

A. Giulietti, E. Van-etten, L. Overbergh, K. Stoffels, and R. Bouillon, Monocytes from type 2 diabetic patients have a pro-inflammatory profile, Diabetes Research and Clinical Practice, vol.77, issue.1, pp.47-57, 2007.
DOI : 10.1016/j.diabres.2006.10.007

X. Zhao, M. Goswami, N. Pokhriyal, H. Ma, and H. Du, Cyclooxygenase-2 Expression during Immortalization and Breast Cancer Progression, Cancer Research, vol.68, issue.2, pp.467-475, 2008.
DOI : 10.1158/0008-5472.CAN-07-0782

S. Y. Lyu, J. H. Choi, and H. J. Lee, References Korean mistletoe lectin promotes proliferation and invasion of trophoblast cells through regulation of Akt signaling, Reprod. Toxicol, pp.39-72, 2013.

]. Y. Endo, K. Tsurugi, and H. Franz, The site of action of the A-chain of mistletoe lectin I on eukaryotic ribosomes. The RNA N-glycosidase activity of the protein, FEBS. Lett, issue.2, pp.231-378, 1988.

]. F. Stirpe, L. Barbieri, and M. G. Battelli, Ribosome???Inactivating Proteins from Plants: Present Status and Future Prospects, Bio/Technology, vol.18, issue.4, pp.405-412, 1992.
DOI : 10.1038/nbt0492-405

A. Sidler, L. Viviani, and . Rist, Human cancer cells exhibit in vitro individual receptiveness towards different mistletoe extracts, Pharmazie, vol.60, pp.448-454, 2005.

]. H. Hulsen, C. Doser, and F. Mechelke, Differences in the in vitro effectiveness of preparations produced from mistletoes of various host trees, Arzneimittelforschung, vol.36, issue.5, pp.433-436, 1986.

]. J. Kleijnen and P. Knipschild, Mistletoe treatment for cancer review of controlled trials in humans, Phytomedicine, vol.1, issue.3, pp.255-260, 1994.
DOI : 10.1016/S0944-7113(11)80073-5

J. Eggenschwiler, L. Von-balthazar, and B. Stritt, Mistletoe lectin is not the only cytotoxic component in fermented preparations of Viscum album from white fir (Abies pectinata), BMC Complement Taking dendritic cells into medicine, Altern. Med. Nature, vol.7, issue.449, pp.419-426, 2007.

]. P. Pierre, S. J. Turley, and E. Gatti, Developmental regulation of MHC class II transport in mouse dendritic cells, Nature, vol.388, pp.787-792, 1997.

R. M. Steinman, Decisions About Dendritic Cells: Past, Present, and Future, Annual Review of Immunology, vol.30, issue.1, pp.1-22, 2012.
DOI : 10.1146/annurev-immunol-100311-102839

C. Kurts, B. W. Robinson, and P. A. Knolle, Cross-priming in health and disease, Nature Reviews Immunology, vol.68, issue.6, pp.403-414, 2010.
DOI : 10.1038/nri2780

C. Dresch, Y. Leverrier, and J. Marvel, Development of antigen cross-presentation capacity in dendritic cells, Trends in Immunology, vol.33, issue.8, pp.381-388, 2012.
DOI : 10.1016/j.it.2012.04.009

B. Almand, J. R. Resser, and B. Lindman, Clinical significance of defective dendritic cell differentiation in cancer, Clin. Cancer Res, vol.6, pp.1755-1766, 2000.

A. P. Vicari, C. Caux, and G. Trinchieri, Tumour escape from immune surveillance through dendritic cell inactivation, Seminars in Cancer Biology, vol.12, issue.1, pp.33-42, 2002.
DOI : 10.1006/scbi.2001.0400

D. I. Gabrilovich, J. Corak, and I. F. Ciernik, Decreased antigen presentation by dendritic cells in patients with breast cancer, Clin. Cancer Res, vol.3, pp.483-490, 1997.

D. I. Gabrilovich, H. L. Chen, and K. R. Girgis, Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells, Nature Medicine, vol.94, issue.10, pp.1096-1103, 1996.
DOI : 10.1016/0925-5710(95)00412-2

C. Menetrier-caux, G. Montmain, and M. C. Dieu, Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor, Blood, vol.92, pp.4778-4791, 1998.

H. Saito, S. Tsujitani, and M. Ikeguchi, Relationship between the expression of vascular endothelial growth factor and the density of dendritic cells in gastric adenocarcinoma tissue, British Journal of Cancer, vol.78, issue.12, pp.1573-1577, 1998.
DOI : 10.1038/bjc.1998.725

M. Augustin, P. R. Bock, and J. Hanisch, Safety and efficacy of the long-term adjuvant treatment of primary intermediate-to high-risk malignant melanoma (UICC/AJCC stage II and III) with a standardized fermented European mistletoe (Viscum album L.) extract. Results from a multicenter, comparative, epidemiological cohort study in Germany and Switzerland, Arzneimittelforschung, vol.55, pp.38-49, 2005.

J. Melzer, F. Iten, and K. Hostanska, Efficacy and Safety of Mistletoe Preparations (Viscum album) for Patients with Cancer Diseases, Forschende Komplement??rmedizin / Research in Complementary Medicine, vol.16, issue.4, pp.217-226, 2009.
DOI : 10.1159/000226249

S. R. Elluru, J. P. Duong-van-huyen, and S. Delignat, Induction of maturation and activation of human dendritic cells: A mechanism underlying the beneficial effect of Viscum albumas complimentary therapy in cancer, BMC Cancer, vol.210, issue.9, p.161, 2008.
DOI : 10.1016/j.imbio.2005.07.004

G. Ribereau-gayon, M. L. Jung, and D. D. Scala, Comparison of the Effects of Fermented and Unfermented Mistletoe Preparations on Cultured Tumor Cells, Oncology, vol.43, issue.1, pp.35-41, 1986.
DOI : 10.1159/000226418

M. S. Maddur, M. Sharma, and P. Hegde, Human B cells induce dendritic cell maturation and favour Th2 polarization by inducing OX-40 ligand, Nature Communications, vol.141, pp.5-4092, 2014.
DOI : 10.1073/pnas.90.8.3730

J. Zhu and W. E. Paul, CD4 T cells: fates, functions, and faults, Blood, vol.112, issue.5, pp.1557-1569, 2008.
DOI : 10.1182/blood-2008-05-078154

E. Vivier, E. Tomasello, and M. Baratin, Functions of natural killer cells, Nature Immunology, vol.178, issue.5, pp.503-510, 2008.
DOI : 10.1073/pnas.050588297

URL : https://hal.archives-ouvertes.fr/hal-00294184

V. Shankaran, H. Ikeda, and A. T. Bruce, IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity, Nature, vol.410, issue.6832, pp.1107-1111, 2001.
DOI : 10.1038/35074122

S. Hong, J. Qian, and J. Yang, Roles of Idiotype-Specific T Cells in Myeloma Cell Growth and Survival: Th1 and CTL Cells Are Tumoricidal while Th2 Cells Promote Tumor Growth, Cancer Research, vol.68, issue.20, pp.8456-8464, 2008.
DOI : 10.1158/0008-5472.CAN-08-2213

D. H. Kaplan, V. Shankaran, and A. S. Dighe, Demonstration of an interferon ??-dependent tumor surveillance system in immunocompetent mice, Proc. Natl. Acad
DOI : 10.1073/pnas.95.13.7556

S. Y. Lyu and W. B. Park, Effects of Korean Mistletoe Lectin(Viscum album coloratum) on Proliferation and Cytokine Expression in Human Peripheral Blood Mononuclear Cells and T-Lymphocytes, Archives of Pharmacal Research, vol.1551, issue.1, pp.30-1252, 2007.
DOI : 10.1007/BF02980266

K. J. Radford, K. M. Tullett, and M. H. Lahoud, Dendritic cells and cancer immunotherapy, Dendritic cells and cancer immunotherapy, pp.26-32, 2014.
DOI : 10.1016/j.coi.2014.01.005

M. S. Kim, J. Lee, and H. S. So, GAMMA-INTERFERON (IFN-??) AUGMENTS APOPTOTIC RESPONSE TO MISTLETOE LECTIN-II VIA UPREGULATION OF FAS/FAS L EXPRESSION AND CASPASE ACTIVATION IN HUMAN MYELOID U937 CELLS, Immunopharmacology and Immunotoxicology, vol.139, issue.5, pp.55-66, 2001.
DOI : 10.1081/IPH-100102567

H. Walzel, M. Blach, and P. Neels, The B-chain of mistletoe lectin I efficiently stimulates calcium signaling in human Jurkat T-cells, Immunology Letters, vol.78, issue.2, pp.78-57, 2001.
DOI : 10.1016/S0165-2478(01)00238-3

K. Kayser, H. J. Gabius, and S. Gabius, Biotinylated ligands for receptor localization. An alternative for immunohistochemistry], Zentralbl. Pathol, vol.137, pp.473-478, 1991.

O. Podlech, P. N. Harter, and M. Mittelbronn, Fermented mistletoe extract as a multimodal antitumoral agent in gliomas, Evid, Based Complement Alternat. Med, pp.2012-501796, 2012.

A. Engert, F. Burrows, and W. Jung, Evaluation of ricin A chain-containing immunotoxins directed against the CD30 antigen as potential reagents for the treatment of Hodgkin's disease, Cancer Res, pp.50-84, 1990.

A. Stettin, J. L. Schultze, and E. Stechemesser, Anti-mistletoe lectin antibodies are produced in patients during therapy with an aqueous mistletoe extract derived from Viscum album L. and neutralize lectin-induced cytotoxicity in vitro

V. Lavastre, M. Pelletier, and R. Saller, Mechanisms Involved in Spontaneous and Viscum album Agglutinin-I-Induced Human Neutrophil Apoptosis: Viscum album Agglutinin-I Accelerates the Loss of Antiapoptotic Mcl-1 Expression and the Degradation of Cytoskeletal Paxillin and Vimentin Proteins Via Caspases, The Journal of Immunology, vol.168, issue.3, pp.168-1419, 2002.
DOI : 10.4049/jimmunol.168.3.1419

C. Maletzki, M. Linnebacher, and R. Savai, Mistletoe lectin has a shiga toxin-like structure and should be combined with other Toll-like receptor ligands in cancer therapy, Cancer Immunology, Immunotherapy, vol.17, issue.1, pp.62-1283, 2013.
DOI : 10.1007/s00262-013-1455-1

H. J. Park, J. H. Hong, and H. J. Kwon, TLR4-mediated activation of mouse macrophages by Korean mistletoe lectin-C (KML-C), Biochemical and Biophysical Research Communications, vol.396, issue.3
DOI : 10.1016/j.bbrc.2010.04.169

R. A. Abagyan and S. Batalov, Do aligned sequences share the same fold?, Journal of Molecular Biology, vol.273, issue.1, pp.355-368, 1997.
DOI : 10.1006/jmbi.1997.1287

J. P. Duong-van-huyen, J. Bayry, and S. Delignat, Induction of apoptosis of endothelial cells by Viscum album: a role for anti-tumoral properties of mistletoe lectins, Mol. Med, vol.8, pp.600-606, 2002.

J. P. Duong-van-huyen, S. Delignat, and J. Bayry, Interleukin-12 is associated with the in vivo anti-tumor effect of mistletoe extracts in B16 mouse melanoma, Cancer Letters, vol.243, issue.1, pp.32-37, 2006.
DOI : 10.1016/j.canlet.2005.11.016

C. I. Delebinski, M. Twardziok, and S. Kleinsimon, A Natural Combination Extract of Viscum album L. Containing Both Triterpene Acids and Lectins Is Highly Effective against AML In Vivo, PLOS ONE, vol.125, issue.4, p.133892, 2015.
DOI : 10.1371/journal.pone.0133892.s001

R. 1. Maier, T. , J. H. Holda, and H. N. Claman, Natural suppressor cells, Prog Clin Biol Res, vol.288, pp.235-279, 1989.

L. P. Seung, Synergy between T-cell immunity and inhibition of paracrine Pages

D. Galun, Cancer Surgery and Supportive Mistletoe Therapy: From Scepticism to Randomised Clinical Trials, Translational Research in Biomedicine, 2015.

T. Ganz, Defensins: antimicrobial peptides of innate immunity, Nature Reviews Immunology, vol.3, issue.9, pp.710-720, 2003.
DOI : 10.1038/nri1180

J. Gavalchin, R. A. Seder, and S. K. Datta, The NZB X SWR model of lupus nephritis. I. Cross-reactive idiotypes of monoclonal anti-DNA antibodies in relation to antigenic specificity, charge, and allotype. Identification of interconnected idiotype families inherited from the normal SWR and the autoimmune NZB parents, pp.138-128, 1987.

I. Alfen, P. Kastirr, M. D. Gruarin, M. Simone, S. Pagani et al., Plasticity of human CD4 T cell subsets, p.630, 2014.

M. Haynes, M. J. Smyth, G. Kroemer, and L. Zitvogel, Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta- dependent adaptive immunity against tumors, pp.15-1170, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00419823

O. Janssen, A. Scheffler, and D. Kabelitz, In vitro effects of mistletoe extracts and mistletoe lectins. Cytotoxicity towards tumor cells due to the induction of programmed cell death (apoptosis), pp.4-7, 1993.

S. K. Zhang, N. T. Kraeft, T. Seyfried, L. B. Abe, R. S. Chen et al., Expression of cyclooxygenase 2 (COX-2) in human glioma and in vitro inhibition by a specific COX-2 inhibitor, NS-398, Cancer Res60, issue.17, pp.4926-4931, 2000.

T. A. Khwaja, C. B. Dias, and S. Pentecost, Recent Studies on the Anticancer Activities of Mistletoe <i>(Viscum album) </i>and Its Alkaloids, Oncology, vol.43, issue.1, pp.42-50, 1986.
DOI : 10.1159/000226419

G. S. Kienle, A. Glockmann, M. Schink, and H. Kiene, Viscum album L. extracts in breast and gynaecological cancers: a systematic review of clinical and preclinical research, Journal of Experimental & Clinical Cancer Research, vol.28, issue.1, p.79, 2009.
DOI : 10.1186/1756-9966-28-79

L. L. Lanier, NK CELL RECEPTORS, Annual Review of Immunology, vol.16, issue.1, pp.359-393, 1998.
DOI : 10.1146/annurev.immunol.16.1.359

. Masferrer, Cyclooxygenase- 2 inhibition by celecoxib reduces proliferation and induces apoptosis in angiogenic endothelial cells in vivo, Cancer, vol.62, issue.3, pp.625-631, 2002.

K. Kuchroo, Induction and molecular signature of pathogenic TH17 cells, Nat Immunol13, issue.10, pp.991-999, 2012.

X. N. Qian, J. W. Xue, and . Pollard, Macrophages regulate the angiogenic switch in a mouse model of breast cancer, Cancer Res66, issue.23, pp.11238-11246, 2006.

W. W. Lin and M. Karin, A cytokine-mediated link between innate immunity, inflammation, and cancer, Journal of Clinical Investigation, vol.117, issue.5, pp.1175-1183, 2007.
DOI : 10.1172/JCI31537

L. Divekar, J. J. Beaton, S. Hogan, A. Fagarasan, K. G. Liston et al., Foxp3+ follicular regulatory T cells control the germinal center response, pp.17-975, 2011.

B. Van-de-putte and S. B. Abramson, Unresolved issues in the role of cyclooxygenase-2 in normal, 2000.

. Dudler, Naive and memory T cells show distinct pathways of lymphocyte recirculation, J Exp, vol.171, issue.3, pp.801-817, 1990.

. Basse, Molecular cloning, occurrence, and expression of a novel partially secreted protein "psoriasin" that is highly upregulated in psoriatic skin, J Invest, vol.97, issue.4, pp.701-712, 1991.

C. Maletzki, M. Linnebacher, R. Savai, and U. Hobohm, Mistletoe lectin has a shiga toxin-like structure and should be combined with other Toll-like receptor ligands in cancer therapy, Cancer Immunology, Immunotherapy, vol.17, issue.1, pp.1283-1292, 2013.
DOI : 10.1007/s00262-013-1455-1

P. J. Mansky, J. Grem, D. B. Wallerstedt, B. P. Monahan, and M. R. Blackman, Mistletoe and Gemcitabine in Patients with Advanced Cancer: A Model for the Phase I Study of Botanicals and Botanical-Drug Interactions in Cancer Therapy, Integrative Cancer Therapies, vol.2, issue.4, pp.345-352, 2003.
DOI : 10.1177/1534735403259061

A. Mantovani, From phagocyte diversity and activation to probiotics: Back to Metchnikoff, European Journal of Immunology, vol.335, issue.12, pp.3269-3273, 2008.
DOI : 10.1002/eji.200838918

A. Mantovani, P. Allavena, A. Sica, and F. Balkwill, Cancer-related inflammation, Nature, vol.342, issue.7203, pp.436-444, 2008.
DOI : 10.1038/nature07205

L. J. Marnett, The COXIB Experience: A Look in the Rearview Mirror, Annual Review of Pharmacology and Toxicology, vol.49, issue.1, pp.265-290, 2009.
DOI : 10.1146/annurev.pharmtox.011008.145638

C. Murdoch, M. Muthana, S. B. Coffelt, and C. E. Lewis, The role of myeloid cells in the promotion of tumour angiogenesis, Nature Reviews Cancer, vol.13, issue.8, pp.8-618, 2008.
DOI : 10.1038/nrc2444

C. Nathan, Neutrophils and immunity: challenges and opportunities, Nature Reviews Immunology, vol.280, issue.3, pp.173-182, 2006.
DOI : 10.1183/09031936.96.09071482

F. O. Nestle, J. Banchereau, and D. Hart, Dendritic cells: On the move from bench to bedside, pp.7-761, 2001.

H. Kim, Diarylheptanoids and flavonoids from viscum album inhibit LPS-stimulated production of pro-inflammatory cytokines in bone marrow-derived dendritic cells, J Nat, vol.76, issue.4, pp.495-502, 2013.

D. Pardoll, ?, Annual Review of Immunology, vol.21, issue.1, pp.807-839, 2003.
DOI : 10.1146/annurev.immunol.21.120601.141135

D. M. Pardoll, Cancer vaccines, Nat, vol.45, pp.525-531, 1998.

. Dong, A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17, pp.1133-1141, 2005.

K. H. Kim, J. B. Lee, S. K. Kim, and . Song, TLR4-mediated activation of mouse macrophages by Korean mistletoe lectin-C (KML, Biochem Biophys Res Commun, vol.396, issue.3, pp.721-725, 2010.

J. H. Park, C. K. Hyun, and H. K. Shin, Cytotoxicity of heat-treated Korean mistletoe, Cancer Letters, vol.126, issue.1, pp.43-48, 1998.
DOI : 10.1016/S0304-3835(97)00526-0

T. J. Hong and M. J. Yoon, Choi transplantation tolerance, Immunol, vol.182, pp.18-32

F. Salazar-onfray, Interleukin-10: a cytokine used by tumors to escape immunosurveillance, Medical Oncology, vol.182, issue.3, pp.86-94, 1999.
DOI : 10.1007/BF02785841

F. Sallusto, J. Geginat, and A. Lanzavecchia, : Function, Generation, and Maintenance, Annual Review of Immunology, vol.22, issue.1, pp.745-763, 2004.
DOI : 10.1146/annurev.immunol.22.012703.104702

F. Sallusto, D. Lenig, R. Forster, M. Lipp, and A. Lanzavecchia, Two subsets of memory T lymphocytes with distinct homing potentials and effector functions, Nature, vol.9, issue.6754, 1999.
DOI : 10.1038/44385

L. Liotta, E. Cosmi, S. Maggi, F. Romagnani, and . Annunziato, TGF-beta indirectly favors the development of human Th17 cells by inhibiting Th1 cells, Eur J, vol.39, issue.1, pp.207-215, 2009.

G. Schaller, K. Urech, G. Grazi, and M. Giannattasio, Viscotoxin composition of the three European subspecies of Viscum album, Planta, vol.64, issue.7, pp.677-678, 1998.

G. Meier, C. Rassner, and . Garbe, Dermcidin: a novel human antibiotic peptide secreted by sweat glands, Nat Immunol2, issue.12, pp.1133-1137, 2001.

M. Schnare, G. M. Barton, A. C. Holt, K. Takeda, S. Akira et al., Toll-like receptors control activation of adaptive immune responses, pp.2-947, 2001.

H. M. Schramm, The Anti-Cancer Activity of Mistletoe Preparations, as Related to Their Polyphenolic profiles, Translational Research in Biomedicine, 2015.

S. S. Verburg, W. W. Yu, G. S. Zhao, and . Geis, Anti- inflammatory and upper gastrointestinal effects of celecoxib in rheumatoid arthritis: a randomized controlled trial, pp.282-1921, 1999.

W. L. Smith, D. L. Dewitt, and R. M. Garavito, Cyclooxygenases: Structural, Cellular, and Molecular Biology, Annual Review of Biochemistry, vol.69, issue.1, pp.145-182, 2000.
DOI : 10.1146/annurev.biochem.69.1.145

M. J. Smyth, G. P. Dunn, and R. D. Schreiber, Cancer Immunosurveillance and Immunoediting: The Roles of Immunity in Suppressing Tumor Development and Shaping Tumor Immunogenicity, Adv, vol.90, pp.1-50, 2006.
DOI : 10.1016/S0065-2776(06)90001-7

T. Taniguchi, S. B. Kawano, N. Y. Pelikan, D. I. Crowe, and . Godfrey, Differential tumor surveillance by natural killer (NK) and NKT cells, J Exp, vol.191, issue.4, pp.661-668, 2000.

. Allavena, Tumorconditioned macrophages secrete migration-stimulating factor: a new marker for M2-polarization, influencing tumor cell motility, J Immunol185, issue.1, pp.642-652, 2010.

C. M. Ulrich, J. Bigler, and J. D. Potter, Non-steroidal anti-inflammatory drugs for cancer prevention: promise, perils and pharmacogenetics, Nature Reviews Cancer, vol.97, issue.2, pp.130-140, 2006.
DOI : 10.1038/nrc1801

D. Unutmaz, RORC2: The master of human Th17 cell programming, European Journal of Immunology, vol.106, issue.6, 2009.
DOI : 10.1002/eji.200939540

K. Urech, Chemical constituents of Viscum album L.: Implications for the pharmaceutical Preparation of Mistletoe, 2015.

A. G. Golenbock, M. Tielens, and . Yazdanbakhsh, A novel host-parasite lipid cross-talk, 2002.

J. R. Vane, Inhibition of Prostaglandin Synthesis as a Mechanism of Action for Aspirin-like Drugs, Nature New Biology, vol.231, issue.25, pp.232-235, 1971.
DOI : 10.1038/newbio231232a0

D. B. Vendramini-costa and J. E. Carvalho, Molecular Link Mechanisms between Inflammation and Cancer, Current Pharmaceutical Design, vol.18, issue.26, pp.3831-3852, 2012.
DOI : 10.2174/138161212802083707

V. Low, E. C. , F. G. Perabo, R. Siener, and S. C. Muller, Review. Facts and fiction of phytotherapy for prostate cancer: a critical assessment of preclinical and clinical data, pp.21-189, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01318883

H. Liang and . Han, Notch signaling determines the M1 versus M2 polarization of macrophages in antitumor immune responses, Cancer Res70, issue.12, pp.4840-4849, 2010.

A. J. Wardlaw, R. Moqbel, and A. B. Kay, Eosinophils: Biology and Role in Disease, Adv, vol.60, pp.151-266, 1995.
DOI : 10.1016/S0065-2776(08)60586-6

J. A. Bukasa, J. R. Mitchell, and . Vane, Nonsteroid drug selectivities for cyclo-oxygenase-1, 1999.

. Braun, Synthetic mycoplasma-derived lipopeptide MALP-2 induces maturation and function of dendritic cells, 2003.

P. Rigley, A filarial nematode-secreted product signals dendritic cells to acquire a phenotype that drives development of Th2 cells, J Immunol164, issue.12, pp.6453-6460, 2000.

K. Wode, T. Schneider, I. Lundberg, and G. S. Kienle, Mistletoe treatment in cancer-related fatigue: a case report, Cases Journal, vol.2, issue.1, p.77, 2009.
DOI : 10.1186/1757-1626-2-77

. Ristimaki, Expression of cyclooxygenase-2 in human lung carcinoma, Cancer Res58, issue.22, pp.4997-5001, 1998.

B. Zafirova, F. M. Wensveen, M. Gulin, and B. Polic, Regulation of immune cell function and differentiation by the NKG2D receptor, Cellular and Molecular Life Sciences, vol.31, issue.21, pp.3519-3529, 2011.
DOI : 10.1007/s00018-011-0797-0

X. Zhang, L. X. Chen, L. Ouyang, Y. Cheng, and B. Liu, Plant natural compounds: targeting pathways of autophagy as anti-cancer, 2012.

C. Saha, P. Hegde, A. Friboulet, J. Bayry, and S. Kaveri, Viscum album-Mediated COX-2 Inhibition Implicates Destabilization of COX-2 mRNA, PLOS ONE, vol.70, issue.2, p.114965, 2015.
DOI : 10.1371/journal.pone.0114965.g004

URL : https://hal.archives-ouvertes.fr/hal-01344611

E. Stephen-victor, C. Saha, M. Sharma, S. Holla, K. Balaji et al., Inhibition of Programmed Death 1 Ligand 1 on Dendritic Cells Enhances Mycobacterium-Mediated Interferon ?? (IFN-??) Production Without Modulating the Frequencies of IFN-??-Producing CD4+ T Cells, Journal of Infectious Diseases, vol.211, issue.6, pp.1027-1029, 2015.
DOI : 10.1093/infdis/jiu532

S. Elluru, C. Saha, P. Hegde, A. Friboulet, J. Bayry et al., Dissecting the anti-inflammatory effects of Viscum album: Inhibition of cytokine-induced expression of cyclooxygenase-2 and secretion of prostaglandin E2, Mistletoe: From Mythology to Evidence-Based-Medicine (Edited by Zaenker KS, and Kaveri SV), 2015.
URL : https://hal.archives-ouvertes.fr/hal-01344595

S. Kaveri, M. Lecerf, C. Saha, M. Kazatchkine, S. Lacroix-desmazes et al., Intravenous immunoglobulin and immune response, Clinical & Experimental Immunology, vol.122, issue.1, pp.94-96, 2014.
DOI : 10.1111/cei.12526

M. Sharma, Y. Schoindre, P. Hegde, C. Saha, M. Maddur et al., Intravenous immunoglobulin-induced IL-33 is insufficient to mediate basophil expansion in autoimmune patients, Scientific Reports, vol.18, p.5672
DOI : 10.1038/srep05672

URL : https://hal.archives-ouvertes.fr/hal-01358914

M. Sharma, C. Saha, Y. Schoindre, L. Gilardin, O. Benveniste et al., Interferon-alpha inhibition by intravenous immunoglobulin is independent of modulation of the plasmacytoid dendritic cell population in the circulation: Arthritis Rheumatol, pp.2308-2309, 2014.

S. Othy, S. Topcu, C. Saha, P. Kothapalli, S. Lacroix-desmazes et al., Sialylation may be dispensable for reciprocal modulation of helper T cells by intravenous immunoglobulin, European Journal of Immunology, vol.127, issue.7, pp.2059-2063, 2014.
DOI : 10.1002/eji.201344230

C. Saha, A. Friboulet, J. Bayry, and S. Kaveri, Differential effect of Viscum album preparations on maturation and activation of human dendritic cells and T cell response

S. Shafiani, G. Tucker-heard, A. Kariyone, K. Takatsu, and K. Urdahl, Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis, The Journal of Experimental Medicine, vol.162, issue.7, pp.1409-1429, 2010.
DOI : 10.1038/ni1059

S. Holla, M. Kurowska-stolarska, J. Bayry, and K. Balaji, -responsive WNT5A and SHH signaling, Autophagy, vol.123, issue.2, pp.311-341, 2014.
DOI : 10.1073/pnas.1019536108

URL : https://hal.archives-ouvertes.fr/hal-01304323

A. Singh, A. Mohan, A. Dey, and D. Mitra, Inhibiting the Programmed Death 1 Pathway Rescues Mycobacterium tuberculosis-Specific Interferon ??-Producing T Cells From Apoptosis in Patients With Pulmonary Tuberculosis, Journal of Infectious Diseases, vol.208, issue.4, pp.603-618, 2013.
DOI : 10.1093/infdis/jit206

M. Butte, M. Keir, T. Phamduy, A. Sharpe, and G. Freeman, Programmed Death-1 Ligand 1 Interacts Specifically with the B7-1 Costimulatory Molecule to Inhibit T Cell Responses, Immunity, vol.27, issue.1, pp.111-133, 2007.
DOI : 10.1016/j.immuni.2007.05.016

M. Maddur, M. Sharma, and P. Hegde, Human B cells induce dendritic cell maturation and favour Th2 polarization by inducing OX-40 ligand, Nature Communications, vol.141, 2014.
DOI : 10.1073/pnas.90.8.3730

S. Periasamy, R. Dhiman, and P. Barnes, Programmed Death 1 and Cytokine Inducible SH2-Containing Protein Dependent Expansion of Regulatory T Cells Upon Stimulation With Mycobacterium tuberculosis, Journal of Infectious Diseases, vol.203, issue.9, pp.1256-63, 2011.
DOI : 10.1093/infdis/jir011

J. Trinath, M. Maddur, S. Kaveri, K. Balaji, and J. Bayry, Mycobacterium tuberculosis Promotes Regulatory T-Cell Expansion via Induction of Programmed Death-1 Ligand 1 (PD-L1, CD274) on Dendritic Cells, Journal of Infectious Diseases, vol.205, issue.4, pp.694-700, 2012.
DOI : 10.1093/infdis/jir820

S. Ha, S. Mueller, and E. Wherry, Enhancing therapeutic vaccination by blocking PD-1???mediated inhibitory signals during chronic infection, The Journal of Experimental Medicine, vol.88, issue.3, pp.543-55, 2008.
DOI : 10.4049/jimmunol.169.7.3760

S. Sable, Programmed Death 1 Lives Up to Its Reputation in Active Tuberculosis, Journal of Infectious Diseases, vol.208, issue.4, pp.541-544, 2013.
DOI : 10.1093/infdis/jit211

I. , F. , I. ). Alves, C. Booty, M. Carpenter et al., National Institute of Immunohaematology, Mumbai, India; and References 1 In search of a new paradigm for protective immunity to TB, Nat Rev Microbiol, vol.12, pp.289-99, 2014.

M. Debre, M. Bonnet, and W. Fridman, Infusion of Fc?? fragments for treatment of children with acute immune thrombocytopenic purpura, The Lancet, vol.342, issue.8877, pp.945-954, 1993.
DOI : 10.1016/0140-6736(93)92000-J

. Quantification-of-cytokines, IL-33 in the plasma samples of the patients and in cellfree culture supernatants was quantified by ELISA (R&D systems, France). IL-4 in the plasma was also measured by ELISA

M. C. Dalakas, Intravenous Immunoglobulin in Autoimmune Neuromuscular Diseases, JAMA, vol.291, issue.19, pp.2367-2375, 2004.
DOI : 10.1001/jama.291.19.2367

M. D. Kazatchkine and S. Kaveri, Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin, N. Engl. J. Med, vol.345, pp.747-755, 2001.

Y. Arnson, Y. Shoenfeld, and H. Amital, Intravenous immunoglobulin therapy for autoimmune diseases, Autoimmunity, vol.13, issue.6, pp.553-560, 2009.
DOI : 10.1002/ana.1088

J. Bayry, V. S. Negi, and S. Kaveri, Intravenous immunoglobulin therapy in rheumatic diseases, Nature Reviews Rheumatology, vol.124, issue.6, pp.349-359, 2011.
DOI : 10.1038/nrrheum.2011.61

I. Schwab and F. Nimmerjahn, Intravenous immunoglobulin therapy: how does IgG modulate the immune system?, Nature Reviews Immunology, vol.113, issue.3, pp.176-189, 2013.
DOI : 10.1038/nri3401

J. F. Seite, Y. Shoenfeld, P. Youinou, and S. Hillion, What is the contents of the magic draft IVIg?, Autoimmunity Reviews, vol.7, issue.6, pp.435-439, 2008.
DOI : 10.1016/j.autrev.2008.04.012

T. Tha-in, J. Bayry, H. J. Metselaar, S. V. Kaveri, and J. Kwekkeboom, Modulation of the cellular immune system by intravenous immunoglobulin, Trends in Immunology, vol.29, issue.12, pp.608-615, 2008.
DOI : 10.1016/j.it.2008.08.004

M. Ballow, The IgG molecule as a biological immune response modifier: Mechanisms of action of intravenous immune serum globulin in autoimmune and inflammatory disorders, Journal of Allergy and Clinical Immunology, vol.127, issue.2, pp.315-323, 2011.
DOI : 10.1016/j.jaci.2010.10.030

S. C. Jordan, M. Toyoda, and A. A. Vo, Intravenous Immunoglobulin a Natural Regulator of Immunity and Inflammation, Transplantation, vol.88, issue.1, pp.1-6, 2009.
DOI : 10.1097/TP.0b013e3181a9e89a

J. W. Semple, Intravenous Immunoglobulin Prevents Murine Antibody-Mediated Acute Lung Injury at the Level of Neutrophil Reactive Oxygen Species (ROS) Production, PLoS ONE, vol.125, issue.Suppl 1, p.31357, 2012.
DOI : 10.1371/journal.pone.0031357.g008

J. Bayry, Intravenous immunoglobulin abrogates dendritic cell differentiation induced by interferon-? present in serum from patients with systemic lupus erythematosus, Arthritis & Rheumatism, vol.18, issue.12, pp.3497-3502, 2003.
DOI : 10.1002/art.11346

E. Aubin, R. Lemieux, and R. Bazin, Indirect inhibition of in vivo and in vitro T-cell responses by intravenous immunoglobulins due to impaired antigen presentation, Blood, vol.115, issue.9, pp.1727-1734, 2010.
DOI : 10.1182/blood-2009-06-225417

M. S. Maddur, Inhibition of differentiation, amplification, and function of human TH17 cells by intravenous immunoglobulin, Journal of Allergy and Clinical Immunology, vol.127, issue.3, pp.823-830, 2011.
DOI : 10.1016/j.jaci.2010.12.1102

M. S. Maddur, S. V. Kaveri, and J. Bayry, Comparison of different IVIg preparations on IL-17 production by human Th17 cells, Autoimmunity Reviews, vol.10, issue.12, pp.809-810, 2011.
DOI : 10.1016/j.autrev.2011.02.007

M. S. Maddur, Inhibitory Effect of IVIG on IL-17 Production by Th17 Cells is Independent of Anti-IL-17 Antibodies in the Immunoglobulin Preparations, Journal of Clinical Immunology, vol.129, issue.Suppl 1, pp.62-66, 2013.
DOI : 10.1007/s10875-012-9752-6

A. Kessel, Intravenous Immunoglobulin Therapy Affects T Regulatory Cells by Increasing Their Suppressive Function, The Journal of Immunology, vol.179, issue.8, pp.5571-5575, 2007.
DOI : 10.4049/jimmunol.179.8.5571

D. Groot and A. S. , Activation of natural regulatory T cells by IgG Fc-derived peptide "Tregitopes", Blood, vol.112, issue.8, pp.3303-3311, 2008.
DOI : 10.1182/blood-2008-02-138073

A. H. Massoud, Intravenous immunoglobulin attenuates airway inflammation through induction of forkhead box protein 3???positive regulatory T cells, Journal of Allergy and Clinical Immunology, vol.129, issue.6, pp.1656-1665, 2012.
DOI : 10.1016/j.jaci.2012.02.050

J. Trinath, Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells, Blood, vol.122, issue.8, pp.1419-1427, 2013.
DOI : 10.1182/blood-2012-11-468264

J. F. Seite, C. Goutsmedt, P. Youinou, J. O. Pers, and S. Hillion, Intravenous immunoglobulin induces a functional silencing program similar to anergy in human B cells, Journal of Allergy and Clinical Immunology, vol.133, issue.1, pp.181-188, 2014.
DOI : 10.1016/j.jaci.2013.08.042

URL : https://hal.archives-ouvertes.fr/hal-00926676

G. References-1-zandman-goddard, A. Krauthammer, Y. Levy, P. Langevitz, and Y. Shoenfeld, Long-Term Therapy with Intravenous Immunoglobulin is Beneficial in Patients with Autoimmune Diseases, Clinical Reviews in Allergy & Immunology, vol.7, issue.Suppl 1, pp.247-255, 2012.
DOI : 10.1007/s12016-011-8278-7

S. V. Kaveri, Intravenous immunoglobulin: Exploiting the potential of natural antibodies, Autoimmunity Reviews, vol.11, issue.11, pp.792-794, 2012.
DOI : 10.1016/j.autrev.2012.02.006

M. D. Kazatchkine and S. V. Kaveri, Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin, N. Engl

T. Tha-in, J. Bayry, H. J. Metselaar, S. V. Kaveri, and J. Kwekkeboom, Modulation of the cellular immune system by intravenous immunoglobulin, Trends in Immunology, vol.29, issue.12, pp.608-615, 2008.
DOI : 10.1016/j.it.2008.08.004

J. F. Seite, Y. Shoenfeld, P. Youinou, and S. Hillion, What is the contents of the magic draft IVIg?, Autoimmunity Reviews, vol.7, issue.6, pp.435-439, 2008.
DOI : 10.1016/j.autrev.2008.04.012

W. , D. Groot, and A. S. , Tregitope update: mechanism of action parallels IVIg, Autoimmun. Rev, vol.12, pp.436-443, 2013.

S. Casulli, S. Topcu, L. Fattoum, S. Von-gunten, H. U. Simon et al., A Differential Concentration-Dependent Effect of IVIg on Neutrophil Functions: Relevance for Anti-Microbial and Anti-Inflammatory Mechanisms, PLoS ONE, vol.6, issue.10, p.26469, 2011.
DOI : 10.1371/journal.pone.0026469.g004

L. M. Araujo, A. Chauvineau, R. Zhu, S. Diem, E. A. Bourgeois et al., Cutting Edge: Intravenous Ig Inhibits Invariant NKT Cell-Mediated Allergic Airway Inflammation through Fc??RIIIA-Dependent Mechanisms, The Journal of Immunology, vol.186, issue.6, pp.3289-3293, 2011.
DOI : 10.4049/jimmunol.1003076

R. M. Anthony, T. Kobayashi, F. Wermeling, and J. V. Ravetch, Intravenous gammaglobulin suppresses inflammation through a novel

I. Schwab, M. Biburger, G. Kro¨nkekro¨nke, G. Schett, and F. Nimmerjahn, IVIg-mediated amelioration of ITP in mice is dependent on sialic acid and SIGNR1, European Journal of Immunology, vol.310, issue.4, pp.826-830, 2012.
DOI : 10.1002/eji.201142260

I. Schwab, S. Mihai, M. Seeling, M. Kasperkiewicz, R. Ludwig et al., Broad requirement for terminal sialic acid residues and Fc??RIIB for the preventive and therapeutic activity of intravenous immunoglobulins in vivo, European Journal of Immunology, vol.16, issue.Suppl 1
DOI : 10.1002/eji.201344230

I. Schwab, M. Seeling, M. Biburger, S. Aschermann, L. Nitschke et al., B cells and CD22 are dispensable for the immediate antiinflammatory activity of intravenous immunoglobulins in vivo, European Journal of Immunology, vol.310, issue.Suppl 1
DOI : 10.1002/eji.201242710

T. Guhr, J. Bloem, N. I. Derksen, M. Wuhrer, A. H. Koenderman et al., Enrichment of Sialylated IgG by Lectin Fractionation Does Not Enhance the Efficacy of Immunoglobulin G in a Murine Model of Immune Thrombocytopenia, PLoS ONE, vol.73, issue.6, p.21246, 2011.
DOI : 10.1371/journal.pone.0021246.s003

F. Ka¨sermannka¨sermann, D. J. Boerema, M. Ruegsegger, A. Hofmann, S. Wymann et al., Analysis and Functional Consequences of Increased Fab-Sialylation of Intravenous Immunoglobulin (IVIG) after Lectin Fractionation, PLoS ONE, vol.18, issue.6, p.37243, 2012.
DOI : 10.1371/journal.pone.0037243.s005

D. Leontyev, Y. Katsman, X. Z. Ma, S. Miescher, F. Ka¨sermannka¨sermann et al., Sialylation-independent mechanism involved in the amelioration of murine immune thrombocytopenia using intravenous gammaglobulin . Transfusion, pp.1799-1805, 2012.

D. Leontyev, Y. Katsman, and D. R. Branch, Mouse background and IVIG dosage are critical in establishing the role of inhibitory Fc?? receptor for the amelioration of experimental ITP, Blood, vol.119, issue.22, pp.5261-5264, 2012.
DOI : 10.1182/blood-2012-03-415695

J. Bayry, K. Bansal, M. D. Kazatchkine, and S. V. Kaveri, DC-SIGN and alpha2,6-sialylated IgG Fc interaction is dispensable for the antiinflammatory activity of IVIg on human dendritic cells, Proc. Natl. Acad

M. Magy and L. , Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells, Blood, vol.122, pp.1419-1427, 2013.

A. E. Wiedeman, D. M. Santer, W. Yan, S. Miescher, F. Ka¨sermannka¨sermann et al., Contrasting mechanisms of interferon-alpha inhibition by intravenous immunoglobulin after induction by immune complexes versus Toll-like receptor agonists. Arthritis Rheum, pp.2713-2723, 2013.

T. Korn, E. Bettelli, M. Oukka, and V. Kuchroo, IL-17 and Th17 Cells, IL-17 and Th17 Cells, pp.485-517, 2009.
DOI : 10.1146/annurev.immunol.021908.132710

R. Gold, C. Linington, and H. Lassmann, Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research, Brain, vol.129, issue.8, pp.1953-1971, 2006.
DOI : 10.1093/brain/awl075

S. Othy, P. Hegde, S. Topcu, M. Sharma, M. S. Maddur et al., Intravenous Gammaglobulin Inhibits Encephalitogenic Potential of Pathogenic T Cells and Interferes with their Trafficking to the Central Nervous System, Implicating Sphingosine-1 Phosphate Receptor 1-Mammalian Target of Rapamycin Axis, The Journal of Immunology, vol.190, issue.9, pp.4535-4541, 2013.
DOI : 10.4049/jimmunol.1201965

M. S. Maddur, J. Vani, P. Hegde, S. Lacroix-desmazes, S. V. Kaveri et al., Inhibition of differentiation, amplification, and function of human TH17 cells by intravenous immunoglobulin, Journal of Allergy and Clinical Immunology, vol.127, issue.3, pp.823-830, 2011.
DOI : 10.1016/j.jaci.2010.12.1102