E. Bwp, Production and quality control of monoclonal antibodies and related substances, 2009.

E. /. Chmp, Similar biological medicinal products [Internet], 2014.

C. , N. Probst, A. Watier, H. Thibault, and G. , Recombinant therapeutic monoclonal antibodies: mechanisms of action in relation to structural and functional duality, Crit Rev Oncol Hematol, vol.64, pp.226-259, 2007.

S. Dall-'ozzo, S. Tartas, G. Paintaud, G. Cartron, P. Colombat et al., Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship, Cancer Res, vol.64, pp.4664-4673, 2004.

G. Cartron, L. Dacheux, G. Salles, P. Solal-celigny, P. Bardos et al., Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor Fcgamma RIIIa gene, Blood, vol.99, issue.3, pp.754-762, 2002.
DOI : 10.1182/blood.V99.3.754

URL : http://www.bloodjournal.org/content/bloodjournal/99/3/754.full.pdf

A. Musolino, N. Naldi, B. Bortesi, D. Pezzuolo, M. Capelletti et al., ???Positive Metastatic Breast Cancer, Journal of Clinical Oncology, vol.26, issue.11, pp.1789-96, 2008.
DOI : 10.1200/JCO.2007.14.8957

W. Zhang, M. Gordon, A. Schultheis, D. Yang, F. Nagashima et al., FCGR2A and FCGR3A Polymorphisms Associated With Clinical Outcome of Epidermal Growth Factor Receptor???Expressing Metastatic Colorectal Cancer Patients Treated With Single-Agent Cetuximab, Journal of Clinical Oncology, vol.25, issue.24, pp.3712-3720, 2007.
DOI : 10.1200/JCO.2006.08.8021

T. Junttila, K. Parsons, C. Olsson, Y. Lu, Y. Xin et al., Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer, Cancer Research, vol.70, issue.11, pp.4481-4490, 2010.
DOI : 10.1158/0008-5472.CAN-09-3704

E. Mossner, P. Brunker, S. Moser, U. Puntener, C. Schmidt et al., Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity, Blood, vol.115, issue.22, pp.4393-402, 2009.
DOI : 10.1182/blood-2009-06-225979

L. Sehn, N. Chua, J. Mayer, G. Dueck, M. Trn-eny et al., GADOLIN: Primary results from a phase III study of obinutuzumab plus bendamustine compared with bendamustine alone in patients with rituximab-refractory indolent non-Hodgkin lymphoma., Journal of Clinical Oncology, vol.33, issue.18_suppl, p.8502, 2015.
DOI : 10.1200/jco.2015.33.18_suppl.lba8502

G. Cartron, S. Guibert, . De, M. Dilhuydy, F. Morschhauser et al., Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study, Blood, vol.124, issue.14, pp.2196-202, 2014.
DOI : 10.1182/blood-2014-07-586610

URL : https://hal.archives-ouvertes.fr/hal-01147035

J. Reichert, Antibodies to watch in 2014, mAbs, vol.6, issue.1, pp.5-14, 2014.
DOI : 10.4161/mabs.2.1.10677

URL : http://www.tandfonline.com/doi/pdf/10.4161/mabs.27333?needAccess=true

A. Sanderson, Quantitative titration, kinetic behaviour, and inhibition of cytotoxic mouse isoantisera, Immunology, vol.9, pp.287-3005838200, 1965.

K. Brunner, J. Mauel, J. Cerottini, and B. Chapuis, Quantitative assay of the lytic action of immune lymphoid cells of 51Cr-labelled allogeneic target cells in vitro; inhibition by isoantibody and by drugs, Immunology, vol.14, pp.181-964966657, 1968.

R. Lichtenfels, W. Biddison, H. Schulz, A. Vogt, and R. Martin, CARE-LASS (calcein-release-assay), an improved fluorescence-based test system to measure cytotoxic T lymphocyte activity, Journal of Immunological Methods, vol.172, issue.2, pp.227-390022, 1994.
DOI : 10.1016/0022-1759(94)90110-4

C. Granberg, K. Blomberg, I. Hemmil?-a, and T. L?-ovgren, Determination of cytotoxic T lymphocyte activity by time-resolved fluorometry using europiumlabelled concanavalin A-stimulated cells as targets, J Immunol Methods, vol.11488, pp.191-50022, 1988.
DOI : 10.1016/0022-1759(88)90173-1

S. Somanchi, K. Mcculley, A. Somanchi, L. Chan, and D. Lee, A Novel Method for Assessment of Natural Killer Cell Cytotoxicity Using Image Cytometry, PLOS ONE, vol.3, issue.16, 2015.
DOI : 10.1371/journal.pone.0141074.s002

URL : https://doi.org/10.1371/journal.pone.0141074

D. Flieger, R. Gruber, G. Schlimok, C. Reiter, K. Pantel et al., A novel non-radioactive cellular cytotoxicity test based on the differential assessment of living and killed target and effector cells, Journal of Immunological Methods, vol.180, issue.1, pp.1-137897241, 1995.
DOI : 10.1016/0022-1759(94)00293-6

R. Wilkinson, A. Lee-macary, D. Davies, D. Snary, and E. Ross, Antibody-dependent cell-mediated cytotoxicity: a flow cytometry-based assay using fluorophores, Journal of Immunological Methods, vol.258, issue.1-2, pp.183-91, 2001.
DOI : 10.1016/S0022-1759(01)00474-4

M. Sheehy, A. Mcdermott, S. Furlan, P. Klenerman, and D. Nixon, A novel technique for the fluorometric assessment of T lymphocyte antigen specific lysis, Journal of Immunological Methods, vol.249, issue.1-2, pp.99-110, 2001.
DOI : 10.1016/S0022-1759(00)00329-X

C. Korzeniewski and D. Callewaert, An enzyme-release assay for natural cytotoxicity, Journal of Immunological Methods, vol.64, issue.3, pp.313-200022, 1983.
DOI : 10.1016/0022-1759(83)90438-6

M. Corey, R. Kinders, L. Brown, and R. Vessella, A very sensitive coupled luminescent assay for cytotoxicity and complement-mediated lysis, Journal of Immunological Methods, vol.207, issue.1, pp.43-51, 1997.
DOI : 10.1016/S0022-1759(97)00098-7

I. Cree and P. Andreotti, Measurement of cytotoxicity by ATP-based luminescence assay in primary cell cultures and cell lines, Toxicology in Vitro, vol.11, issue.5, pp.553-559, 1997.
DOI : 10.1016/S0887-2333(97)00060-X

J. Peper, H. Schuster, M. L?-offler, B. Schmid-horch, H. Rammensee et al., An impedance-based cytotoxicity assay for real-time and label-free assessment of T-cell-mediated killing of adherent cells, Journal of Immunological Methods, vol.405, pp.192-200, 2014.
DOI : 10.1016/j.jim.2014.01.012

U. Seidel, F. Vogt, L. Grosse-hovest, G. Jung, R. Handgretinger et al., gd T cell-mediated antibody-dependent cellular cytotoxicity with CD19 antibodies assessed by an impedance-based label-free real-time cytotoxicity assay, Front Immunol, vol.5, p.618, 2014.
DOI : 10.3389/fimmu.2014.00618

URL : http://journal.frontiersin.org/article/10.3389/fimmu.2014.00618/pdf

B. Parekh, E. Berger, S. Sibley, S. Cahya, L. Xiao et al., Development and validation of an antibody-dependent cell-mediated cytotoxicity-reporter gene assay, mAbs, vol.148, issue.3, pp.310-318, 2012.
DOI : 10.1038/nbt1026

URL : http://www.tandfonline.com/doi/pdf/10.4161/mabs.19873?needAccess=true

P. Wolint, M. Betts, R. Koup, and A. Oxenius, T Cells, The Journal of Experimental Medicine, vol.170, issue.7, pp.925-961, 2004.
DOI : 10.4049/jimmunol.170.2.805

C. De-romeuf, C. Dutertre, L. Garff-tavernier, M. Fournier, N. Gaucher et al., Chronic lymphocytic leukaemia cells are efficiently killed by an anti-CD20 monoclonal antibody selected for improved engagement of Fc??RIIIA/CD16, British Journal of Haematology, vol.177, issue.6, pp.635-678, 2008.
DOI : 10.1200/JCO.2003.05.013

R. Repp, C. Kellner, A. Muskulus, M. Staudinger, S. Nodehi et al., Combined Fc-protein- and Fc-glyco-engineering of scFv-Fc fusion proteins synergistically enhances CD16a binding but does not further enhance NK-cell mediated ADCC, Journal of Immunological Methods, vol.373, issue.1-2, pp.67-78, 2011.
DOI : 10.1016/j.jim.2011.08.003

Y. Kawaguchi, K. Kono, Y. Mizukami, K. Mimura, and H. Fujii, Mechanisms of escape from Trastuzumab-mediated ADCC in esophageal squamous cell carcinoma: relation to susceptibility to perforin-granzyme, Anticancer Res, vol.29, pp.2137-4619528474, 2009.

M. Hall, J. Unch, B. Binkowski, M. Valley, B. Butler et al., Engineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone Substrate, ACS Chemical Biology, vol.7, issue.11, pp.1848-57, 2012.
DOI : 10.1021/cb3002478

URL : https://doi.org/10.1021/cb3002478

S. Mamidi, M. Cinci, M. Hasmann, V. Fehring, and M. Kirschfink, Lipoplex mediated silencing of membrane regulators (CD46, CD55 and CD59) enhances complement-dependent anti-tumor activity of trastuzumab and pertuzumab, Molecular Oncology, vol.150, issue.3, pp.580-94, 2013.
DOI : 10.1111/j.1365-2249.2007.03507.x

URL : https://doi.org/10.1016/j.molonc.2013.02.011

B. Cl-emenceau, C. , N. Gallot, G. Vivien, R. Gaschet et al., Antibody-dependent cellular cytotoxicity (ADCC) is mediated by genetically modified antigen-specific human T lymphocytes, Blood, vol.107, issue.12, pp.4669-77, 2006.
DOI : 10.1182/blood-2005-09-3775

H. Sch?-afer, A. Sch?-afer, A. Kiderlen, K. Masihi, and R. Burger, A highly sensitive cytotoxicity assay based on the release of reporter enzymes, from stably transfected cell lines, J Immunol Methods, vol.20497, pp.89-98, 1997.

P. Ho, K. Yue, P. Pandey, L. Breault, F. Harbinski et al., Reporter Enzyme Inhibitor Study To Aid Assembly of Orthogonal Reporter Gene Assays, ACS Chemical Biology, vol.8, issue.5, pp.1009-1026, 2013.
DOI : 10.1021/cb3007264

L. Zhang, G. Song, T. Xu, Q. Wu, X. Shao et al., A novel ultrasensitive bioluminescent receptor-binding assay of INSL3 through chemical conjugation with nanoluciferase, Biochimie, vol.95, issue.12, pp.2454-2463, 2013.
DOI : 10.1016/j.biochi.2013.09.008

A. Stacer, S. Nyati, P. Moudgil, R. Iyengar, K. Luker et al., NanoLuc Reporter for Dual Luciferase Imaging in Living Animals, Molecular Imaging, vol.12, issue.7, pp.1-1324371848, 2013.
DOI : 10.2310/7290.2013.00062

URL : https://doi.org/10.2310/7290.2013.00062

V. Tran, L. Moser, D. Poole, and A. Mehle, Highly Sensitive Real-Time In Vivo Imaging of an Influenza Reporter Virus Reveals Dynamics of Replication and Spread, Journal of Virology, vol.87, issue.24, pp.13321-902381, 2013.
DOI : 10.1128/JVI.02381-13

G. Song, Q. Jiang, T. Xu, Y. Liu, Z. Xu et al., A convenient luminescence assay of ferroportin internalization to study its interaction with hepcidin, FEBS Journal, vol.43, issue.8, pp.1773-81, 2013.
DOI : 10.1007/s00726-011-1164-z

URL : http://onlinelibrary.wiley.com/doi/10.1111/febs.12192/pdf

J. Norisada, Y. Hirata, F. Amaya, K. Kiuchi, and K. Oh-hashi, A sensitive assay for the biosynthesis and secretion of MANF using NanoLuc activity, Biochemical and Biophysical Research Communications, vol.449, issue.4, pp.483-492, 2014.
DOI : 10.1016/j.bbrc.2014.05.031

J. Lopez, O. Susanto, M. Jenkins, N. Lukoyanova, V. Sutton et al., Perforin forms transient pores on the target cell plasma membrane to facilitate rapid access of granzymes during killer cell attack, Blood, vol.121, issue.14, pp.2659-68, 2013.
DOI : 10.1182/blood-2012-07-446146

URL : http://www.bloodjournal.org/content/bloodjournal/121/14/2659.full.pdf

S. Cullen and S. Martin, Mechanisms of granule-dependent killing, Cell Death & Differentiation, vol.1996, issue.2, pp.251-62, 2007.
DOI : 10.1182/blood-2006-03-010348

URL : http://www.nature.com/cdd/journal/v15/n2/pdf/4402244a.pdf

P. A. Saama, unified approach for performing an analytical methods comparability study [Internet], p.15081, 2015.

W. Hauck, R. Capen, J. Callahan, J. Muth, H. Hsu et al., Assessing parallelism prior to determining relative potency, PDA J Pharm Sci Technol, vol.59, pp.127-3715971545, 2005.

I. Naruse, H. Fukumoto, N. Saijo, and K. Nishio, Enhanced Anti-tumor Effect of Trastuzumab in Combination with Cisplatin, Japanese Journal of Cancer Research, vol.36, issue.5, pp.574-81, 2002.
DOI : 10.1016/0090-8258(90)90099-7

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1349-7006.2002.tb01293.x/pdf

T. Wehrman, W. Raab, C. Casipit, R. Doyonnas, J. Pomerantz et al., A system for quantifying dynamic protein interactions defines a role for Herceptin in modulating ErbB2 interactions, Proceedings of the National Academy of Sciences, vol.91, issue.6, pp.19063-19071, 2006.
DOI : 10.1038/sj.bjc.6601676

URL : http://www.pnas.org/content/103/50/19063.full.pdf

S. Kuwada, C. Scaife, J. Kuang, X. Li, R. Wong et al., Effects of trastuzumab on epidermal growth factor receptor-dependent and -independent human colon cancer cells, International Journal of Cancer, vol.105, issue.2, pp.291-301, 2004.
DOI : 10.1002/ijc.11137

URL : http://onlinelibrary.wiley.com/doi/10.1002/ijc.11686/pdf

G. Liminga, P. Nygren, S. Dhar, K. Nilsson, and R. Larsson, Cytotoxic effect of calcein acetoxymethyl ester on human tumor cell lines, Anti-Cancer Drugs, vol.6, issue.4, pp.578-857579562, 1995.
DOI : 10.1097/00001813-199508000-00011

C. Perez, I. Albert, K. Defay, N. Zachariades, L. Gooding et al., A nonsecretable cell surface mutant of tumor necrosis factor (TNF) kills by cell-to-cell contact, Cell, vol.63, issue.2, pp.251-80092, 1990.
DOI : 10.1016/0092-8674(90)90158-B

S. Fath, A. Bauer, M. Liss, A. Spriestersbach, B. Maertens et al., Multiparameter RNA and codon optimization: A standardized tool to assess and enhance autologous mammalian gene expression, PLoS One, vol.6, 2011.
DOI : 10.1371/journal.pone.0017596

URL : https://doi.org/10.1371/journal.pone.0017596