Summary

Generation of Discriminative Human Monoclonal Antibodies from Rare Antigen-specific B Cells Circulating in Blood

Published: February 06, 2018
doi:

Summary

We describe a method for the production of human antibodies specific for an antigen of interest, starting from rare B cells circulating in human blood. Generation of these natural antibodies is efficient and rapid, and the antibodies obtained can discriminate between highly related antigens.

Abstract

Monoclonal antibodies (mAbs) are powerful tools useful for both fundamental research and in biomedicine. Their high specificity is indispensable when the antibody needs to distinguish between highly related structures (e.g., a normal protein and a mutated version thereof). The current way of generating such discriminative mAbs involves extensive screening of multiple Ab-producing B cells, which is both costly and time consuming. We propose here a rapid and cost-effective method for the generation of discriminative, fully human mAbs starting from human blood circulating B lymphocytes. The originality of this strategy is due to the selection of specific antigen binding B cells combined with the counter-selection of all other cells, using readily available Peripheral Blood Mononuclear Cells (PBMC). Once specific B cells are isolated, cDNA (complementary deoxyribonucleic acid) sequences coding for the corresponding mAb are obtained using single cell Reverse Transcription-Polymerase Chain Reaction (RT-PCR) technology and subsequently expressed in human cells. Within as little as 1 month, it is possible to produce milligrams of highly discriminative human mAbs directed against virtually any desired antigen naturally detected by the B cell repertoire.

Introduction

The method described here allows the rapid and versatile production of fully human monoclonal antibodies (mAbs) against a desired antigen (Ag). mAbs are essential tools in many fundamental research applications in vitro and in vivo: flow cytometry, histology, western-blotting, and blocking experiments for example. Furthermore, mAbs are being used more and more in medicine to treat autoimmune diseases, cancer, and to control transplantation rejection1. For example, anti-CTLA-4 and anti-PD-1 (or anti-PD-L1) mAbs were recently used as immune checkpoint inhibitors in cancer treatments2.

The first mAbs were produced by immunoglobulin (Ig)-secreting hybridomas obtained from the splenic cells of immunized mice or rats. However, the strong immune response against murine or rat mAbs hampers their therapeutic use in humans, due to their rapid clearance and the probable induction of hypersensitivity reactions3. To tackle this problem, animal protein sequences of mAbs have been partially replaced by human ones to generate so-called chimeric mouse-human or humanized antibodies. However, this strategy only partially decreases immunogenicity, while substantially increasing both the cost and the time-scale of production. A better solution is to generate human mAbs directly from human B cells and several strategies for this are available. One of them is the use of phage or yeast display. This involves displaying variable domains from a combinatorial library of random human Ig heavy and light chains on phages or yeasts, and carrying out a selection step using the specific antigen of interest. A major drawback of this strategy is that heavy and light chains are randomly associated, leading to a very large increase in the diversity of generated antibodies. Antibodies obtained are unlikely to correspond to those that would arise from a natural immune response against a particular Ag. Moreover, human protein folding and post-translational modifications are not systematically reproduced in prokaryotes or even in yeasts. A second human mAb production method is the immortalization of natural human B cells, by Epstein-Barr virus infection or expression of the anti-apoptotic factors BCL-6 and BCL-XL4. However, this method is applicable only to memory B cells and is inefficient, requiring screening of numerous mAb-producing immortalized B cells to identify the few (if any) mAb clones with the desired antigenic specificity. The method is thus both costly and time consuming.

A new protocol has recently been described for production of human mAbs from isolated single B cells5. It relies on an optimized single-cell Reverse Transcription-Polymerase Chain Reaction (RT-PCR) for amplification of both the heavy- and light-chain encoding segments from a single sorted B cell. This is followed by the cloning and expression of these segments in a eukaryotic expression system, thus allowing reconstruction of a fully human mAb. This protocol has been used successfully starting from B cells from vaccinated donors. Cells were harvested several weeks after vaccination to obtain higher frequencies of B cells directed against the desired Ag, and thus limit the time required for screening6. Other fully human mAbs have also been produced from HIV+ (Human Immunodeficiency Virus) infected patients7 and melanoma patients8. Despite these advances, there is still no procedure available that enables the isolation of Ag-specific B cells independent of their memory phenotype or frequency.

The procedure described here leads to efficient ex vivo isolation of human circulating B cells based on their BCR specificity, followed by the production of fully human antigen-specific mAbs in high yield and with a low screening time. The method is not restricted to memory B cells or antibody-secreting B cells induced after an immune response, but can also be applied to the human naïve B cell repertoire. That it works even starting from Ag-specific B cells present at very low frequencies is a good indication of its efficiency. The principle of the method is as follows: Peripheral Blood Mononuclear Cells (PBMC) are stained with two tetramers presenting the antigen of interest, each labeled with a different fluorochrome (e.g., Phycoerythrin (PE) and Allophycocyanin (APC)), and a third tetramer presenting a closely related antigen conjugated with a third fluorochrome (e.g., Brillant Violet 421 (BV421)). To enrich for antigen-binding cells, cells are then incubated with beads coated with anti-PE and anti-APC Abs, and sorted in cell separation columns. The PE+ APC+ cell fraction is selected, stained with a variety of mAbs specific for different PBMC cell types to permit identification of B cells, and subjected to flow cytometry cell sorting. B cells which are PE+ and APC+, but Brilliant Violet, are isolated. This step counter-selects cells which are not B cells or do not bind to the tetramerized antigen, but do bind to either PE or APC (these cells will be PE+ APCor PE APC+) or to the non-antigen part of the tetramers used (these cells will be BV421+). B cells not highly specific for the epitope of interest are also counter-selected at this step (these cells will also be BV421+). Thus, this method can purify highly specific B cells expressing B-cell Receptors (BCRs) able to discriminate between two very closely related antigens. Single specific B cells are collected in tubes and their PCR-amplified Ig cDNAs (complementary deoxyribonucleic acids) cloned and expressed by a human cell line as secreted IgG mAbs.

As a proof of concept, this study describes the efficient generation of human mAbs, which recognize a peptide presented by a major histocompatibility complex class I (MHC-I) molecule and can discriminate between this peptide and other peptides loaded on the same MHC-I allele. Although the level of complexity of this Ag is important, this method allows (i) high yield recovery of Ag-specific mAbs; (ii) production of mAbs able to discriminate between two structurally close Ags. This approach can be extended to vaccinated or infected patients without any protocol modification, and has also already been successfully implemented in a humanized rat system9. Thus, this study describes a versatile and efficient approach to generate fully human mAbs that can be used in basic research and immunotherapy.

Protocol

All human peripheral blood samples were obtained from anonymous adult donors after informed consent, in accordance with the local ethics committee (Etablissement Français du Sang, EFS, Nantes, procedure PLER NTS-2016-08). 1. Isolation of Human Peripheral Blood Mononuclear Cells NOTE: Starting material can be total human peripheral blood or cytapheresis samples. Samples should not be older than 8 h and supplemented with anticoagulants (e.g., heparin)….

Representative Results

Starting from PBMC from healthy donors, this project presents the generation of human mAbs, which recognize the peptide Pp65495 (Pp65, from human cytomegalovirus) presented by the major histocompatibility complex class I (MHC-I) molecule HLA-A*0201 (HLA-A2). These mAbs can discriminate between this complex and complexes representing other peptides loaded onto the same MHC-I molecule. PBMC were stained with HLA-A2/Pp65…

Discussion

The proposed protocol is a powerful method for the generation of human mAbs directly from Ag-specific B cells circulating in the blood. It combines three important aspects: (i) the use of a tetramer-associated magnetic enrichment, which allows an ex vivo isolation of even rare Ag-binding B cells; (ii) a gating strategy that uses three Ag tetramers (two relevant ones and one irrelevant one) labelled with three different fluorochromes to isolate, by flow cytometry, only the B cells expressing a BCR specific for th…

Disclosures

The authors have nothing to disclose.

Acknowledgements

We thank the Cytometry Facility "CytoCell" (SFR Santé, Biogenouest, Nantes) for expert technical assistance. We thank also all the staff of recombinant protein production (P2R) and of IMPACT platforms (INSERM 1232, SFR Santé, Biogenouest, Nantes) for their technical support. We thank Emmanuel Scotet and Richard Breathnach for constructive comments on the manuscript. This work was financially supported by the IHU-Cesti project funded by the « Investissements d'Avenir » French Government program, managed by the French National Research Agency (ANR) (ANR-10-IBHU-005). The IHU-Cesti project is also supported by Nantes Métropole and Région Pays de la Loire. This work was realized in the context of the LabEX IGO program supported by the National Research Agency via the investment of the future program ANR-11-LABX-0016-01.

Materials

HEK 293A cell line Thermo Fisher scientific R70507
DMEM (1X) Dulbecco's Modified Eagle Medium Gibco by life technologies 21969-035 (+) 4,5g/L D-Glucose
0,11g/L Sodium Pyruvate
(-) L-Glutmine
RPMI medium1640 (1X) Gibco by life technologies 31870-025
Bovine Serum Albumine (BSA) PAA K45-001
Nutridoma-SP Roche 11011375001 100X Conc
PBS-Phosphate Buffered Saline (10X) pH 7,4 Ambion AM9624
EDTA (Ethylenediaminetetraacetic acid) 0,5M pH=8 Invitrogen by Life Technologies 15575-020
Fetal Bovine serum (FBS) Dominique Dutscher S1810-500
Ficoll – lymphocytes separation medium EuroBio CMSMSL01-01 density 1,0777+/-0,001
streptavidin R-phycoerythrin conjugate Invitrogen by Life Technologies S21388 premiun grade 1mg/ml contains 5mM sodium azide
Streptavidin, allophycocyanin conjugate Invitrogen by thermoFisher scientific S32362 1mg/ml
2mM azide premium grade
Brilliant violet 421 streptavidin Biolegend 405225 conc : 0,5mg/ml
Anti-PE conjugated magnetic MicroBeads Miltenyi Biotec 130-048-801
Anti-APC conjugated magnetic MicroBeads Miltenyi Biotec 130-090-855
MidiMACs or QuadroMACS separotor Miltenyi Biotec 130-042-302/130-090-976
LS Columns Miltenyi Biotec 130-042-401
CD3 BV510 BD horizon BD Pharmingen / BD Biosciences 563109 Used dilution 1:20
CD19 FITC BD Pharmingen / BD Biosciences 345788 Used dilution 1:20
CD14 PerCPCy5.5 BD Pharmingen / BD Biosciences 561116 Used dilution 1:50
CD16 PerCPCy5.5 BD Pharmingen / BD Biosciences 338440 Used dilution 1:50
7AAD BD Pharmingen / BD Biosciences 51-68981E (559925) Used dilution 1:1000
FACS ARIA III Cell Sorter Cytometer BD Biosciences
8-strip PCR tubes Axygen 321-10-061
Racks for 96 microtubes Dominique Dutscher 45476
RNAseOUT Ribonuclease Inhibitor (recombinant) Invitrogen by thermoFisher scientific 10777-019 qty:5000U (40U/ul)
Distilled Water Dnase/Rnase Free Gibco 10977-035
Oligod(T)18 mRNA Primer New England BioLabs S1316S 5.0 A260unit
Random hexamers Invitrogen by thermoFisher scientific N8080127 qty : 50uM, 5nmoles
Superscript III Reverse transcriptase Invitrogen by thermoFisher scientific 18080-044 qty : 10000U (200U/ul)
GoTaq G2 Flexi DNA polymerase Promega M7805
dNTP Set, Molecular biology grade Thermo Scientific R0182 4*100umol
5LVH1 Eurofins ACAGGTGCCCACT
CCCAGGTGCAG
First round of PCR – Amplification of heavy chains – Outer primers – Forward Prmers
5LVH3 Eurofins AAGGTGTCCAGTG
TGARGTGCAG
First round of PCR – Amplification of heavy chains – Outer primers – Forward Prmers
5LVL4_6 Eurofins CCCAGATGGGTCC
TGTCCCAGGTGCAG
First round of PCR – Amplification of heavy chains – Outer primers – Forward Prmers
5LVH5 Eurofins CAAGGAGTCTGTT
CCGAGGTGCAG
First round of PCR – Amplification of heavy chains – Outer primers – Forward Prmers
3HuIgG_const_anti Eurofins TCTTGTCCACCTT
GGTGTTGCT
First round of PCR – Amplification of heavy chains – Outer primers -Reverse primers for human Ig- Bacteria PCR screening
3CuCH1 Eurofins GGGAATTCTCACA
GGAGACGA
First round of PCR – Amplification of heavy chains – Outer primers -Reverse primers for human Ig
5AgeIVH1_5_7 Eurofins CTGCAACCGGTGTACATTCC
GAGGTGCAGCTGGTGCAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH3 Eurofins CTGCAACCGGTGTACATTCT
GAGGTGCAGCTGGTGGAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH3_23 Eurofins CTGCAACCGGTGTACATTCT
GAGGTGCAGCTGTTGGAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH4 Eurofins CTGCAACCGGTGTACATTCC
CAGGTGCAGCTGCAGGAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH4_34 Eurofins CTGCAACCGGTGTACATTCC
CAGGTGCAGCTACAGCAGTG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH1_18 Eurofins CTGCAACCGGTGTACATTCC
CAGGTTCAGCTGGTGCAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH1_24 Eurofins CTGCAACCGGTGTACATTCC
CAGGTCCAGCTGGTACAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH3__9_30_33 Eurofins CTGCAACCGGTGTACATTCT
GAAGTGCAGCTGGTGGAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
5AgeIVH6_1 Eurofins CTGCAACCGGTGTACATTCC
CAGGTACAGCTGCAGCAG
Second round of PCR – Amplification of heavy chains – Inner primers -Forward primers
3SalIJH1_2_4_5 Eurofins TGCGAAGTCGACG
CTGAGGAGACGGTGACCAG
Second round of PCR – Amplification of heavy chains – Inner primers -Reverse primers
3SalIJH3 Eurofins TGCGAAGTCGACG
CTGAAGAGACGGTGACCATTG
Second round of PCR – Amplification of heavy chains – Inner primers -Reverse primers
3SalIJH6 Eurofins TGCGAAGTCGACG
CTGAGGAGACGGTGACCGTG
Second round of PCR – Amplification of heavy chains – Inner primers -Reverse primers
5'LVk1_2 Eurofins ATGAGGSTCCCYG
CTCAGCTGCTGG
First round of PCR – Amplification of light chains k – Outer primers -Forward primers
5'LVk3 Eurofins CTCTTCCTCCTGC
TACTCTGGCTCCCAG
First round of PCR – Amplification of light chains k – Outer primers -Forward primers
5'LVk4 Eurofins ATTTCTCTGTTGC
TCTGGATCTCTG
First round of PCR – Amplification of light chains k – Outer primers -Forward primers
3'Ck543_566 Eurofins GTTTCTCGTAGTC
TGCTTTGCTCA
First round of PCR – Amplification of light chains k – Outer primers -Reverse primers- Bacteria PCR screening
5'AgeIVk1 Eurofins CTGCAACCGGTGTACATTCT
GACATCCAGATGACCCAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeIVk1_9_1–13 Eurofins TTGTGCTGCAACCGGTGTAC
ATTCAGACATCCAGTTGACCCAGTCT
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeIVk1D_43_1_8 Eurofins CTGCAACCGGTGTACATTGT
GCCATCCGGATGACCCAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeIVk2 Eurofins CTGCAACCGGTGTACATGGG
GATATTGTGATGACCCAGAC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeIVk2_28_2_30 Eurofins CTGCAACCGGTGTACATGGG
GATATTGTGATGACTCAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeVk3_11_3D_11 Eurofins TTGTGCTGCAACCGGTGTAC
ATTCAGAAATTGTGTTGACACAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeVk3_15_3D_15 Eurofins CTGCAACCGGTGTACATTCA
GAAATAGTGATGACGCAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeVk3_20_3D_20 Eurofins TTGTGCTGCAACCGGTGTAC
ATTCAGAAATTGTGTTGACGCAGTCT
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'AgeVk4_1 Eurofins CTGCAACCGGTGTACATTCG
GACATCGTGATGACCCAGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
3'BsiWIJk1_2_4 Eurofins GCCACCGTACGTT
TGATYTCCACCTTGGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
3'BsiWIJk3 Eurofins GCCACCGTACGTT
TGATATCCACTTTGGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
3'BsiWIJk5 Eurofins GCCACCGTACGTT
TAATCTCCAGTCGTGTC
Second round of PCR – Amplification of light chains k – Inner primers -Forward primers
5'LVl1 Eurofins GGTCCTGGGCCCA
GTCTGTGCTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'LVl2 Eurofins GGTCCTGGGCCCA
GTCTGCCCTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'LVl3 Eurofins GCTCTGTGACCTC
CTATGAGCTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'LVl4_5 Eurofins GGTCTCTCTCSCA
GCYTGTGCTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'LVl6 Eurofins GTTCTTGGGCCAA
TTTTATGCTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'LVl7 Eurofins GGTCCAATTCYCA
GGCTGTGGTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5LVl8 Eurofins GAGTGGATTCTCA
GACTGTGGTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
3'Cl Eurofins CACCAGTGTGGCC
TTGTTGGCTTG
First round of PCR – Amplification of light chains λ – Outer primers -Forward primers
5'AgeIVl1 Eurofins CTGCTACCGGTTCCTGGGCC
CAGTCTGTGCTGACKCAG
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
5'AgeIVl2 Eurofins CTGCTACCGGTTCCTGGGCC
CAGTCTGCCCTGACTCAG
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
5'AgeIVl3 Eurofins CTGCTACCGGTTCTGTGACC
TCCTATGAGCTGACWCAG
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
5'AgeIVl4_5 Eurofins CTGCTACCGGTTCTCTCTCS
CAGCYTGTGCTGACTCA
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
5'AgeIVl6 Eurofins CTGCTACCGGTTCTTGGGCC
AATTTTATGCTGACTCAG
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
5'AgeIVl8 Eurofins CTGCTACCGGTTCCAATTCY
CAGRCTGTGGTGACYCAG
Second round of PCR – Amplification of light chains λ – Inner primers -forward primers
3'XhoICl Eurofins CTCCTCACTCGAG
GGYGGGAACAGAGTG
Second round of PCR – Amplification of light chains λ – Inner primers -Reverse primers – Bacteria PCR screening
Ab-vec-sense Eurofins GCTTCGTTAGAAC
GCGGCTAC
Bacteria PCR screening
QA Agarose-TM, Molecular Biology Grade MP Bio AGAH0500
NucleoFast 96 PCR Plate Macherey Nagel 743.100.100
Enzyme Age I HF New England Biolabs R3552L 20000U/ml
Enzyme SalI HF New England Biolabs R3138L 20000U/ml
Enzyme Xho I New England Biolabs R0146L 20000U/ml
Enzyme BSIWI New England Biolabs R0553L 10000U/ml
HCg1 (Genbank accession number FJ475055)
LCk (Genbank accession number FJ475056 )
LCl (Genbank accession number FJ517647)
T4 DNA ligase Invitrogen by thermoFisher scientific 15224.017 100U (1U/ul)
2X YT medium Sigma Aldrich Y1003-500ML
Ampicillin Sigma Aldrich 10835242001
LB (Luria Bertani) Broth (Lennox) Sigma Aldrich L3022-250G
Nucleospin Plasmid DNA, RNA and protein purification Macherey Nagel 740588.250
Jet PEI DNA transfection reagent PolyPlus 101-40
Flat bottom96-well plate Falcon 353072
V-bottom 96-well plate Nunc/Thermofisher 055142
Nunc easy 175 cm2 flasks Nunc/Thermofisher 12-562-000
ELISA/ELISPOT coating buffer eBiosciences 00-0044-59
Nunc maxisorp flat bottom 96 well ELISA plates Nunc/Thermofisher 44-2404-21 high protein binding
Anti-human IgG Ab conjugated to horseradish peroxidase (HRP) BD Pharmingen / BD Biosciences 55788
TMB substrate BD Biosciences 555214
Streptavidin Sigma S0677
1 mL-HiTrap protein A HP column GE Healthcare 17-0402-01
ÄKTA FPLC GE Healthcare 18190026
Superdex 200 10/300 GL column GE Healthcare 17517501
NGC Quest 10 Plus Chromatography System BioRad 7880003

References

  1. Buss, N. A., Henderson, S. J., McFarlane, M., Shenton, J. M., de Haan, L. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol. 12 (5), 615-622 (2012).
  2. Park, J., Kwon, M., Shin, E. C. Immune checkpoint inhibitors for cancer treatment. Arch Pharm Res. 39 (11), 1577-1587 (2016).
  3. Pendley, C., Schantz, A., Wagner, C. Immunogenicity of therapeutic monoclonal antibodies. Curr Opin Mol Ther. 5 (2), 172-179 (2003).
  4. Kwakkenbos, M. J., et al. Generation of stable monoclonal antibody-producing B cell receptor-positive human memory B cells by genetic programming. Nat Med. 16 (1), 123-128 (2010).
  5. Tiller, T., et al. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J Immunol Methods. 329 (1-2), 112-124 (2008).
  6. Franz, B., May, K. F., Dranoff, G., Wucherpfennig, K. Ex vivo characterization and isolation of rare memory B cells with antigen tetramers. Blood. 118 (2), 348-357 (2011).
  7. Morris, L., et al. Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigen-specific single B cell sorting. PLoS One. 6 (9), e23532 (2011).
  8. Iizuka, A., et al. Identification of cytomegalovirus (CMV)pp65 antigen-specific human monoclonal antibodies using single B cell-based antibody gene cloning from melanoma patients. Immunol Lett. 135 (1-2), 64-73 (2011).
  9. Ouisse, L. H., et al. Antigen-specific single B cell sorting and expression-cloning from immunoglobulin humanized rats: a rapid and versatile method for the generation of high affinity and discriminative human monoclonal antibodies. BMC Biotechnol. 17 (1), 3 (2017).
  10. Kerkman, P. F., et al. Identification and characterisation of citrullinated antigen-specific B cells in peripheral blood of patients with rheumatoid arthritis. Ann Rheum Dis. , (2015).
  11. Hamilton, J. A., et al. General Approach for Tetramer-Based Identification of Autoantigen-Reactive B Cells: Characterization of La- and snRNP-Reactive B Cells in Autoimmune BXD2 Mice. J Immunol. 194 (10), 5022-5034 (2015).
  12. Sanjuan Nandin, I., et al. Novel in vitro booster vaccination to rapidly generate antigen-specific human monoclonal antibodies. J Exp Med. , (2017).
  13. Bowers, P. M., et al. Mammalian cell display for the discovery and optimization of antibody therapeutics. Methods. 65 (1), 44-56 (2014).
check_url/56508?article_type=t

Play Video

Cite This Article
Devilder, M., Moyon, M., Saulquin, X., Gautreau-Rolland, L. Generation of Discriminative Human Monoclonal Antibodies from Rare Antigen-specific B Cells Circulating in Blood. J. Vis. Exp. (132), e56508, doi:10.3791/56508 (2018).

View Video