Peanut oral immunotherapy transiently expands circulating Ara h 2-specific B cells with a homologous repertoire in unrelated subjects.

BACKGROUND Peanut oral immunotherapy (PNOIT) induces persistent tolerance to peanut in a subset of patients and induces specific antibodies that might play a role in clinical protection. However, the contribution of induced antibody clones to clinical tolerance in PNOIT is unknown. OBJECTIVE We hypothesized that PNOIT induces a clonal, allergen-specific B-cell response that could serve as a surrogate for clinical outcomes. METHODS We used a fluorescent Ara h 2 multimer for affinity selection of Ara h 2-specific B cells and subsequent single-cell immunoglobulin amplification. The diversity of related clones was evaluated by means of next-generation sequencing of immunoglobulin heavy chains from circulating memory B cells with 2x250 paired-end sequencing on the Illumina MiSeq platform. RESULTS Expression of class-switched antibodies from Ara h 2-positive cells confirms enrichment for Ara h 2 specificity. PNOIT induces an early and transient expansion of circulating Ara h 2-specific memory B cells that peaks at week 7. Ara h 2-specific sequences from memory cells have rates of nonsilent mutations consistent with affinity maturation. The repertoire of Ara h 2-specific antibodies is oligoclonal. Next-generation sequencing-based repertoire analysis of circulating memory B cells reveals evidence for convergent selection of related sequences in 3 unrelated subjects, suggesting the presence of similar Ara h 2-specific B-cell clones. CONCLUSIONS Using a novel affinity selection approach to identify antigen-specific B cells, we demonstrate that the early PNOIT-induced Ara h 2-specific B-cell receptor repertoire is oligoclonal and somatically hypermutated and shares similar clonal groups among unrelated subjects consistent with convergent selection.

[1]  H. Sampson,et al.  Peanut epitopes for IgE and IgG4 in peanut-sensitized children in relation to severity of peanut allergy. , 2008, The Journal of allergy and clinical immunology.

[2]  Alan Y. Chiang,et al.  Generalized Additive Models: An Introduction With R , 2007, Technometrics.

[3]  B. Chipps Oral Immunotherapy for Treatment of Egg Allergy in Children , 2013, Pediatrics.

[4]  S. Durham,et al.  Clinical efficacy and immune regulation with peanut oral immunotherapy. , 2009, The Journal of allergy and clinical immunology.

[5]  Véronique Giudicelli,et al.  IMGT/V-QUEST: IMGT standardized analysis of the immunoglobulin (IG) and T cell receptor (TR) nucleotide sequences. , 2011, Cold Spring Harbor protocols.

[6]  G. Ippolito,et al.  Immunoglobulin Analysis Tool: A Novel Tool for the Analysis of Human and Mouse Heavy and Light Chain Transcripts , 2012, Front. Immun..

[7]  Scott D Boyd,et al.  Convergent antibody signatures in human dengue. , 2013, Cell host & microbe.

[8]  A. Lanzavecchia Antigen Uptake and Accumulation in Antigen‐Specific B Cells , 1987, Immunological reviews.

[9]  Jae Ho Lee,et al.  Allergen-specific B cell subset responses in cow's milk allergy of late eczematous reactions in atopic dermatitis. , 2010, Cellular immunology.

[10]  S. Durham,et al.  Allergen specificity of IgG(4)-expressing B cells in patients with grass pollen allergy undergoing immunotherapy. , 2012, The Journal of allergy and clinical immunology.

[11]  S. Wood Generalized Additive Models: An Introduction with R , 2006 .

[12]  B. Diamond,et al.  Identification of an antigen-specific B cell population. , 2003, Journal of immunological methods.

[13]  C. Akdis,et al.  IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. , 2013, The Journal of allergy and clinical immunology.

[14]  Mario Roederer,et al.  Focused Evolution of HIV-1 Neutralizing Antibodies Revealed by Structures and Deep Sequencing , 2011, Science.

[15]  M. Kulis,et al.  Evidence of pathway‐specific basophil anergy induced by peanut oral immunotherapy in peanut‐allergic children , 2012, Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology.

[16]  B. Stollar,et al.  Human immunoglobulin variable region gene analysis by single cell RT-PCR. , 2000, Journal of immunological methods.

[17]  Scott H Sicherer,et al.  Prevalence of peanut and tree nut allergy in the United States determined by means of a random digit dial telephone survey: a 5-year follow-up study. , 2003, The Journal of allergy and clinical immunology.

[18]  Michel C Nussenzweig,et al.  Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. , 2008, Journal of immunological methods.

[19]  S. Barratt-Boyes,et al.  Association between Magnitude of the Virus-Specific Plasmablast Response and Disease Severity in Dengue Patients , 2013, The Journal of Immunology.

[20]  Stacie M Jones,et al.  Sustained Unresponsiveness to Peanut in Subjects Who Have Completed Peanut Oral Immunotherapy , 2014, Pediatrics.

[21]  T. Kepler,et al.  Isolation of a Human Anti-HIV gp41 Membrane Proximal Region Neutralizing Antibody by Antigen-Specific Single B Cell Sorting , 2011, PloS one.

[22]  Jérôme Lane,et al.  IMGT®, the international ImMunoGeneTics information system® , 2004, Nucleic Acids Res..

[23]  R. Cooke,et al.  SEROLOGICAL EVIDENCE OF IMMUNITY WITH COEXISTING SENSITIZATION IN A TYPE OF HUMAN ALLERGY (HAY FEVER) , 1935, The Journal of experimental medicine.

[24]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[25]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[26]  S. Durham,et al.  Functional rather than immunoreactive levels of IgG4 correlate closely with clinical response to grass pollen immunotherapy , 2012, Allergy.

[27]  A. Burks,et al.  Peanut oral immunotherapy modifies IgE and IgG4 responses to major peanut allergens. , 2013, The Journal of allergy and clinical immunology.

[28]  Stephen L. Hauser,et al.  Naive antibody gene-segment frequencies are heritable and unaltered by chronic lymphocyte ablation , 2011, Proceedings of the National Academy of Sciences.

[29]  Steven J. M. Jones,et al.  Circos: an information aesthetic for comparative genomics. , 2009, Genome research.

[30]  A. Knulst,et al.  The diagnostic value of specific IgE to Ara h 2 to predict peanut allergy in children is comparable to a validated and updated diagnostic prediction model. , 2013, The Journal of allergy and clinical immunology.

[31]  Harry B. Greenberg,et al.  Erratum: Lineage structure of the human antibody repertoire in response to influenza vaccination (Science Translational Medicine (2013) 5 (193er8)) , 2013 .

[32]  Mark M. Davis,et al.  Human responses to influenza vaccination show seroconversion signatures and convergent antibody rearrangements. , 2014, Cell host & microbe.

[33]  A. Kemper,et al.  A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. , 2011, The Journal of allergy and clinical immunology.

[34]  A. Burks,et al.  Prevalence of peanut and tree nut allergy in the US determined by a random digit dial telephone survey. , 1999, The Journal of allergy and clinical immunology.

[35]  L. Penland,et al.  Determinism and stochasticity during maturation of the zebrafish antibody repertoire , 2011, Proceedings of the National Academy of Sciences.

[36]  Douglas S Kwon,et al.  Profiling human antibody responses by integrated single-cell analysis. , 2014, Vaccine.

[37]  P. Lipsky,et al.  Secondary Immunization Generates Clonally Related Antigen-Specific Plasma Cells and Memory B Cells , 2010, The Journal of Immunology.

[38]  G. Dranoff,et al.  Ex vivo characterization and isolation of rare memory B cells with antigen tetramers. , 2011, Blood.

[39]  Mark M. Davis,et al.  Lineage Structure of the Human Antibody Repertoire in Response to Influenza Vaccination , 2013, Science Translational Medicine.

[40]  A. Burks,et al.  Evidence of Desensitization by Sublingual Immunotherapy in Peanut-Allergic Children , 2011 .

[41]  S. Durham,et al.  Mechanisms of immunotherapy to aeroallergens , 2011, Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology.

[42]  Marie-Paule Lefranc,et al.  IMGT/V-QUEST: the highly customized and integrated system for IG and TR standardized V-J and V-D-J sequence analysis , 2008, Nucleic Acids Res..

[43]  S. Durham,et al.  Inhibition of allergen-IgE binding to B cells by IgG antibodies after grass pollen immunotherapy. , 2003, The Journal of allergy and clinical immunology.

[44]  H. Sampson,et al.  IgE and IgG4 epitope mapping by microarray immunoassay reveals the diversity of immune response to the peanut allergen, Ara h 2. , 2005, The Journal of allergy and clinical immunology.

[45]  A. Burks,et al.  Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. , 2011, The Journal of allergy and clinical immunology.

[46]  V. Giudicelli,et al.  IMGT(®) tools for the nucleotide analysis of immunoglobulin (IG) and T cell receptor (TR) V-(D)-J repertoires, polymorphisms, and IG mutations: IMGT/V-QUEST and IMGT/HighV-QUEST for NGS. , 2012, Methods in molecular biology.

[47]  A. Ponsonby,et al.  Increasing the accuracy of peanut allergy diagnosis by using Ara h 2. , 2012, The Journal of allergy and clinical immunology.