P. J. Murray, Macrophage Polarization, Annu Rev Physiol, vol.79, pp.541-566, 2017.

S. Gordon and P. R. Taylor, Monocyte and macrophage heterogeneity, Nat Rev Immunol, vol.5, pp.953-964, 2005.

A. Sica and A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, J Clin Invest, vol.122, pp.787-795, 2012.

E. E. Perez, J. S. Orange, F. Bonilla, J. Chinen, I. K. Chinn et al., Update on the use of immunoglobulin in human disease: A review of evidence, J Allergy Clin Immunol, vol.139, pp.1-46, 2017.

C. Joao, V. S. Negi, M. D. Kazatchkine, J. Bayry, and S. V. Kaveri, Passive serum therapy to immunomodulation by IVIG: A fascinating journey of antibodies, J Immunol, vol.200, pp.1957-1963, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02455445

C. Galeotti, S. V. Kaveri, and J. Bayry, IVIG-mediated effector functions in autoimmune and inflammatory diseases, Int Immunol, vol.29, pp.491-498, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01723555

I. Schwab and F. Nimmerjahn, Intravenous immunoglobulin therapy: how does IgG modulate the immune system?, Nat Rev Immunol, vol.13, pp.176-189, 2013.

J. F. Seite, Y. Shoenfeld, P. Youinou, and S. Hillion, What is the contents of the magic draft IVIg?, Autoimmun Rev, vol.7, pp.435-439, 2008.

A. C. Chan and P. J. Carter, Therapeutic antibodies for autoimmunity and inflammation, Nat Rev Immunol, vol.10, pp.301-316, 2010.

J. Casan, J. Wong, M. J. Northcott, and S. Opat, Anti-CD20 monoclonal antibodies: reviewing a revolution, Hum Vaccin Immunother, vol.14, pp.2820-2841, 2018.

J. Bayry, Repressing immunity in autoimmune disease, N Engl J Med, vol.374, pp.2090-2092, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02455469

A. K. Abbas, E. Trotta, D. R. Simeonov, A. Marson, and J. A. Bluestone, Revisiting IL-2: Biology and therapeutic prospects, Sci Immunol, vol.3, p.1482, 2018.

D. C. Wraith, The future of immunotherapy: A 20-year perspective, Front Immunol, vol.8, p.1668, 2018.

J. Bayry and S. V. Kaveri, Kill 'Em All: Efgartigimod immunotherapy for autoimmune diseases, Trends Pharmacol Sci, vol.39, pp.919-922, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02455435

J. Pozsgay, Z. Szekanecz, and G. Sarmay, Antigen-specific immunotherapies in rheumatic diseases, Nat Rev Rheumatol, vol.13, pp.525-537, 2017.

E. Stephen-victor and J. Bayry, Multimerized IgG1 Fc molecule as an anti-inflammatory agent, Nat Rev Rheumatol, vol.14, pp.390-392, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01835524

L. Cousens, N. Najafian, W. D. Martin, D. Groot, and A. S. , Tregitope: Immunomodulation powerhouse, Hum Immunol, vol.75, pp.1139-1146, 2014.

Y. Lai, S. Suo, R. Wang, X. Kong, Y. Hu et al., Trends involving monoclonal antibody (mAb) research and commercialization: A scientometric analysis of IMS Lifecycle R&D Focus Database (1980-2016), Hum Vaccin Immunother, vol.14, pp.847-855, 2018.

S. Othy, P. Hegde, S. Topçu, 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, J Immunol, vol.190, pp.4535-4541, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02455535

B. M. Fiebiger, J. Maamary, A. Pincetic, and J. V. Ravetch, Protection in antibody-and T cellmediated autoimmune diseases by antiinflammatory IgG Fcs requires type II FcRs, Proc Natl Acad Sci. USA, vol.112, pp.2385-2394, 2015.

M. S. Maddur, E. Stephen-victor, M. Das, P. Prakhar, V. K. Sharma et al., Regulatory T cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to intravenous immunoglobulin therapy, J Neuroinflammation, vol.14, p.58, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01502272

M. Sharma, M. Das, E. Stephen-victor, C. Galeotti, A. Karnam et al., Regulatory T cells induce activation rather than suppression of human basophils, Sci Immunol, vol.3, p.829, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01817714

C. Galeotti, E. Stephen-victor, A. Karnam, M. Das, L. Gilardin et al., Intravenous immunoglobulin induces IL-4 in human basophils by signaling through surface-bound IgE, J Allergy Clin Immunol, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02284256

M. S. Maddur, S. V. Kaveri, and J. Bayry, Circulating normal IgG as stimulator of regulatory T cells: Lessons from intravenous immunoglobulin, Trends Immunol, vol.38, pp.789-792, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01630293

Y. Su, R. Rossi, D. Groot, A. S. Scott, and D. W. , Regulatory T cell epitopes (Tregitopes) in IgG induce tolerance in vivo and lack immunogenicity per se, J Leukoc Biol, vol.94, pp.377-383, 2013.

C. Saha, M. Das, V. Patil, E. Stephen-victor, M. Sharma et al., Monomeric immunoglobulin A from plasma inhibits human Th17 responses in vitro independent of Fc?RI and DC-SIGN, Front Immunol, vol.8, p.275, 2017.

A. H. Massoud, G. N. Kaufman, D. Xue, M. Béland, M. Dembele et al., Peripherally generated Foxp3+ regulatory T cells mediate the immunomodulatory effects of IVIg in allergic airways disease, J Immunol, vol.198, pp.2760-2771, 2017.

S. Bozza, F. Kasermann, S. V. Kaveri, L. Romani, and J. Bayry, Intravenous immunoglobulin protects from experimental allergic bronchopulmonary aspergillosis via a sialylationdependent mechanism, Eur J Immunol, vol.49, pp.195-198, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02455434

V. Ruiz-de-souza, M. P. Carreno, S. V. Kaveri, A. Ledur, H. Sadeghi et al., Selective induction of interleukin-1 receptor antagonist and interleukin-8 in human monocytes by normal polyspecific IgG (intravenous immunoglobulin), Eur J Immunol, vol.25, pp.1267-1273, 1995.

K. H. Park-min, N. V. Serbina, W. Yang, X. Ma, G. Krystal et al., Fc?RIII-dependent inhibition of IFN? responses mediates suppressive effects of intravenous immune globulin, Immunity, vol.26, pp.67-78, 2007.

C. Galeotti, P. Hegde, M. Das, E. Stephen-victor, F. Canale et al., Heme oxygenase-1 is dispensable for the antiinflammatory activity of intravenous immunoglobulin, Sci Rep, vol.6, p.19592, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01274079

L. K. Kozicky, S. C. Menzies, Z. Y. Zhao, T. Vira, K. Harnden et al., IVIg and LPS co-stimulation induces IL-10 production by human monocytes, which is compromised by an Fc?RIIA disease-associated gene variant, Front Immunol, vol.9, p.2676, 2018.

L. K. Kozicky, Z. Y. Zhao, S. C. Menzies, M. Fidanza, G. S. Reid et al., Intravenous immunoglobulin skews macrophages to an anti-inflammatory, IL-10-producing activation state, J Leukoc Biol, vol.98, pp.983-994, 2015.

J. Abe, T. Jibiki, S. Noma, T. Nakajima, H. Saito et al., Gene expression profiling of the effect of high-dose intravenous Ig in patients with Kawasaki disease, J Immunol, vol.174, pp.5837-5845, 2005.

J. K. Damås, L. Gullestad, H. Aass, S. Simonsen, J. G. Fjeld et al., Enhanced gene expression of chemokines and their corresponding receptors in mononuclear blood cells in chronic heart failure--modulatory effect of intravenous immunoglobulin, J Am Coll Cardiol, vol.38, pp.187-193, 2001.

A. Gratchev, J. Kzhyshkowska, K. Köthe, I. Muller-molinet, S. Kannookadan et al., Mphi1 and Mphi2 can be re-polarized by Th2 or Th1 cytokines, respectively, and respond to exogenous danger signals, Immunobiology, vol.211, pp.473-486, 2006.

S. Marino, N. A. Cilfone, J. T. Mattila, J. J. Linderman, J. L. Flynn et al., Macrophage polarization drives granuloma outcome during Mycobacterium tuberculosis infection, Infect Immun, vol.83, pp.324-338, 2015.

A. S. Tjon, R. Van-gent, T. B. Geijtenbeek, and J. Kwekkeboom, Differences in antionflammatory actions of intravenous immunoglobulin between mice and men: more than meets the eye, Front Immunol, vol.6, p.197, 2015.

C. Galeotti, S. V. Kaveri, and J. Bayry, Molecular and immunological biomarkers to predict IVIg response, Trends Mol Med, vol.21, pp.145-147, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01117084

C. Meregalli, I. Marjanovic, C. Scali, L. Monza, N. Spinoni et al., High-dose intravenous immunoglobulins reduce nerve macrophage infiltration and the severity of bortezomib-induced peripheral neurotoxicity in rats, J Neuroinflammation, vol.15, p.232, 2018.

A. Domínguez-soto, M. De-las-casas-engel, R. Bragado, J. Medina-echeverz, L. Aragoneses-fenoll et al., Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization, J Immunol, vol.193, pp.5181-5189, 2014.

S. Von-gunten, Y. Shoenfeld, M. Blank, D. R. Branch, T. Vassilev et al., IVIG pluripotency and the concept of Fc-sialylation: challenges to the scientist, Nat Rev Immunol, vol.14, p.349, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02455513

Y. Kaneko, F. Nimmerjahn, and J. V. Ravetch, Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation, Science, vol.313, pp.670-673, 2006.

R. M. Anthony, F. Wermeling, M. C. Karlsson, and J. V. Ravetch, Identification of a receptor required for the anti-inflammatory activity of IVIG, Proc Natl Acad Sci U S A, vol.105, pp.19571-19578, 2008.

A. H. Massoud, M. Yona, D. Xue, F. Chouiali, H. Alturaihi et al., Dendritic cell immunoreceptor: a novel receptor for intravenous immunoglobulin mediates induction of regulatory T cells, J Allergy Clin Immunol, vol.133, pp.853-863, 2014.

J. F. Séïté, D. Cornec, Y. Renaudineau, P. Youinou, R. A. Mageed et al., IVIg modulates BCR signaling through CD22 and promotes apoptosis in mature human B lymphocytes, Blood, vol.116, pp.1698-1704, 2010.