FcRn Antagonism Leads to a Decrease of Desmoglein-Specific B Cells: Secondary Analysis of a Phase 2 Study of Efgartigimod in Pemphigus Vulgaris and Pemphigus Foliaceus

Maud Maho-Vaillant, Magdalena Sips, Marie-Laure Golinski, Gestur Vidarsson, Matthias Goebeler, Johanna Stoevesandt, Zsuzsanna Bata-Csörgő, Bianca Balbino, Peter Verheesen, Pascal Joly, Michael Hertl, Sébastien Calbo, Maud Maho-Vaillant, Magdalena Sips, Marie-Laure Golinski, Gestur Vidarsson, Matthias Goebeler, Johanna Stoevesandt, Zsuzsanna Bata-Csörgő, Bianca Balbino, Peter Verheesen, Pascal Joly, Michael Hertl, Sébastien Calbo

Abstract

Background: Immunoglobulin G (IgG) levels are maintained by the IgG-recycling neonatal Fc-receptor (FcRn). Pemphigus vulgaris and pemphigus foliaceus are debilitating autoimmune disorders triggered by IgG autoantibodies against mucosal and epidermal desmogleins. Recently, a phase 2 clinical trial (NCT03334058; https://ichgcp.net/clinical-trials-registry/NCT03334058) was completed in participants with pemphigus using efgartigimod, an FcRn inhibitor, in combination with prednisone. Efgartigimod demonstrated an early effect on diease activity and was well tolerated. In addition to the safety and efficacy assessment, clinical trials present an opportunity to gain more insights into the mechanism of disease, the mode of action of treatment, and potential for corticosteroid-sparing activity.

Objective: The aim of our study was to assess the impact of FcRn antagonism by efgartigimod on immunological parameters known to be directly involved in pemphigus pathology, such as cellular and serological responses.

Methods: We investigated total and antigen-specific IgG subclass level kinetics during and after treatment, assessed antigen-specific B-cell responses, followed T- and B-cell immunophenotypes, and analyzed how different immunophenotypes link to clinical response.

Results: Treatment resulted in reduction of total IgG as well as autoreactive IgG antibody levels. Surprisingly, unlike total IgG and vaccine- or natural-infection-elicited IgG, which returned to baseline levels after stopping efgartigimod treatment, autoreactive antibody levels remained low in several study participants. Efgartigimod showed no effect on total leukocytes, neutrophils, monocytes, or lymphocytes in patients treated with extended efgartigimod therapy. Intriguingly, antigen-specific analyses revealed a loss of desmoglein-specific B cells in several participants responding to efgartigimod, in line with prolonged reduction of pathogenic IgG levels.

Conclusions: Efgartigimod treatment of participants with pemphigus improved their conditions and exerted an immunomodulatory effect beyond the blockade of IgG recycling. Further studies in larger populations with an appropriate placebo control are needed to confirm these potentially important observations to establish long-term clinical responses in autoimmune diseases.

Keywords: B cells; FcRn; efgartigimod; immunoglobulin G; pemphigus foliaceus (PF); pemphigus vulgaris (PV).

Conflict of interest statement

ZB-C reports consulting fees for Sanofi-Genzyme, Novartis, and argenx. MG has served as a consultant and on advisory boards for argenx, Biotest, GSK, Janssen, LEO Pharma, Lilly, Novartis, and UCB. MS, BB, and PV are employed by argenx. PJ has served as a consultant for Amgen, Principia Biopharma, argenx, AstraZeneca, Janssen, Thermo Fisher, Lilly, Sanofi, Akari, Chugai, Novartis, Kezar, Genentech, and Topas. GV is a paid consultant for argenx. SC, MM-V, and M-LG as employees of INSERM U1234, Normandie University have a collaborative research agreement with argenx. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. This study received funding from Argenx. The funder had the following involvement with the study: participated in the study design, research, analysis, data collection, interpretation of data, and review and approval of the publication. Argenx also funded the medical writing support of this manuscript.

Copyright © 2022 Maho-Vaillant, Sips, Golinski, Vidarsson, Goebeler, Stoevesandt, Bata-Csörgő, Balbino, Verheesen, Joly, Hertl and Calbo.

Figures

Figure 1
Figure 1
Sustained clinical responses and transient general reduction of total serum IgG levels following efgartigimod treatment. (A) Clinical responses to efgartigimod are depicted for all participants in cohorts 3 (n = 7) and 4 (n = 15). Participant numbers correspond to those used in Table 1. Blue arrows indicate when efgartigimod was administered, and the blue dotted line indicates every other week administration time period in participants who achieved EoC. (B) Clinical responses of singular representative trial participants with pemphigus foliaceus and pemphigus vulgaris, respectively, are shown. (C) Longitudinal changes in pathogenic (anti-Dsg-3 or anti-Dsg-1) as well as total IgG (tIgG) and PDAI activity scores are represented. The gray shaded area indicates efgartigimod treatment-free follow-up period. Means are compared using two-way ANOVA multiple comparison with Dunnett’s post-test. (D) Pathogenic (anti-Dsg-3 or anti-Dsg-1), total IgG (tIgG), and non-pathogenic antibodies titers (anti-VZV, anti-varicella zoster virus; TT, tetanus toxoid; PCP, pneumococcal capsular polysaccharide) are compared between BL and EoS for all participants. Wilcoxon matched-pairs test was performed. *p ≤ 0.05; **p ≤ 0.01; ***p ≤ 0.001; ****p ≤ 0.0001. BL, baseline; EoS, end of study; DC, disease control; EoC, end of consolidation; CR, complete clinical remission; CRmin, complete clinical remission with minimal treatment; Pt, participant; ns, non significant.
Figure 2
Figure 2
Efgartigimod reduces antigen-specific B cells in the blood. (A) Among the three PV participants with viable PBMC and sustained clinical response, representative flow cytometry plots depicting frequency of Dsg-3+ switched memory B cells (MBC) for one participant with PV at baseline, CR, and EoT were generated. (B) Frequency of circulating Dsg-3+ switched memory B cells in three participants with PV with sustained clinical response during the study in relation to Dsg-3 autoantibody serum titers at baseline and by time point. (C) Among the three PF participants with viable PBMCs, a representative ELISPOT of total IgG-ASCs with 2.5 × 103 to 1 × 104 PBMCs plated per well (left) and Dsg-1 IgG ASCs with 1 × 105 to 4 × 105 PBMCs plated per well (right) detected in PBMCs of a participant with PF. (D) Frequency of peripheral blood Dsg-1-specific ASCs evaluated by ELISPOT assay in 3 participants with PF during study both in relation to Dsg-1 autoantibody serum titers at baseline and by time point. Frequencies are reported as a percentage of total IgG ASC. ASCs, antibody-secreting cells; BL, baseline; CR, complete clinical remission; EoT, end of treatment; EoS, end of study.
Figure 3
Figure 3
Efgartigimod does not affect frequencies of lymphocyte subsets except B cells. (A) Leukocytes (109/L) and frequency (%) of neutrophils, lymphocytes, and monocytes within leukocytes in peripheral blood of cohort 4 participants (n = 10), median, and IQR are plotted. (B) Frequency of CD4+ T cells within lymphocytes and their subsets of cohort 4 participants with viable PBMC (n = 9), median, and IQR are plotted. (C) Frequency of CD19+ B cells within lymphocytes and their subsets of cohort 4 participants with viable PBMC (n = 9), median, and IQR are plotted. Arrows and lines indicate the way the frequency of a parent population was determined. (D) Frequency and counts of CD19+ B cells at baseline, EoT, and EoS of cohort 4 participants with viable PBMC and sustained clinical response (n = 9). Red lines represent medians and dotted lines represent normal limits. A non-parametric one-way ANOVA with Dunn’s post-test was performed. *p ≤ 0.05; **p ≤ 0.01. EoT, end of treatment; EoS, end of study; IQR, interquartile range.
Figure 4
Figure 4
Prednisone exposure in participants with prolonged efgartigimod treatment and observed B-cell immunomodulation (n = 9). Summary profiles of the PV and PF participants achieving sustained clinical response illustrating prednisone dosages, PDAI activity scores, and frequency of total CD19+ B cells in peripheral blood when available. The gray shaded area indicates efgartigimod-free follow-up period. Error bars represent standard error of the mean (SEM). PV, pemphigus vulgaris; PF, pemphigus foliaceus; PDAI, pemphigus disease area index.

References

    1. Cooper GS, Stroehla BC. The Epidemiology of Autoimmune Diseases. Autoimmun Rev (2003) 2(3):119–25. doi: 10.1016/s1568-9972(03)00006-5
    1. Eaton WW, Rose NR, Kalaydjian A, Pedersen MG, Mortensen PB. Epidemiology of Autoimmune Diseases in Denmark. J Autoimmun (2007) 29(1):1–9. doi: 10.1016/j.jaut.2007.05.002
    1. Hofmann K, Clauder AK, Manz RA. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front Immunol (2018) 9:835. doi: 10.3389/fimmu.2018.00835
    1. Joly P, Maho-Vaillant M, Prost-Squarcioni C, Hebert V, Houivet E, Calbo S, et al. . French Study Group on Autoimmune Bullous Skin D. First-Line Rituximab Combined With Short-Term Prednisone Versus Prednisone Alone for the Treatment of Pemphigus (Ritux 3): A Prospective, Multicentre, Parallel-Group, Open-Label Randomised Trial. Lancet (2017) 389(10083):2031–40. doi: 10.1016/S0140-6736(17)30070-3
    1. Werth VP, Joly P, Mimouni D, Maverakis E, Caux F, Lehane P, et al. . Rituximab Versus Mycophenolate Mofetil in Patients With Pemphigus Vulgaris. N Engl J Med (2021) 384(24):2295–305. doi: 10.1056/NEJMoa2028564
    1. Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant Autoantibody Production by Early Human B Cell Precursors. Science (2003) 301(5638):1374–7. doi: 10.1126/science.1086907
    1. Holstein J, Solimani F, Baum C, Meier K, Pollmann R, Didona D, et al. . Immunophenotyping in Pemphigus Reveals a TH17/TFH17 Cell-Dominated Immune Response Promoting Desmoglein1/3-Specific Autoantibody Production. J Allergy Clin Immunol (2020)147(6):2358–69. doi: 10.1016/j.jaci.2020.11.008
    1. Hertl M, Eming R, Veldman C. T Cell Control in Autoimmune Bullous Skin Disorders. J Clin Invest (2006) 116(5):1159–66. doi: 10.1172/JCI28547
    1. Eming R, Nagel A, Wolff-Franke S, Podstawa E, Debus D, Hertl M. Rituximab Exerts a Dual Effect in Pemphigus Vulgaris. J Invest Dermatol (2008) 128(12):2850–8. doi: 10.1038/jid.2008.172
    1. Nagel A, Podstawa E, Eickmann M, Muller HH, Hertl M, Eming R. Rituximab Mediates a Strong Elevation of B-Cell-Activating Factor Associated With Increased Pathogen-Specific IgG But Not Autoantibodies in Pemphigus Vulgaris. J Invest Dermatol (2009) 129(9):2202–10. doi: 10.1038/jid.2009.27
    1. Hampe CS. B Cell in Autoimmune Diseases. Sci (Cairo) (2012) 2012: 1–18. doi: 10.6064/2012/215308
    1. Stefanski AL, Dorner T. Immune Checkpoints and the Multiple Faces of B Cells in Systemic Lupus Erythematosus. Curr Opin Rheumatol (2021) 33(6):592–7. doi: 10.1097/BOR.0000000000000825
    1. Pollmann R, Walter E, Schmidt T, Waschke J, Hertl M, Mobs C, et al. . Identification of Autoreactive B Cell Subpopulations in Peripheral Blood of Autoimmune Patients With Pemphigus Vulgaris. Front Immunol (2019) 10:1375. doi: 10.3389/fimmu.2019.01375
    1. Hale M, Rawlings DJ, Jackson SW. The Long and the Short of it: Insights Into the Cellular Source of Autoantibodies as Revealed by B Cell Depletion Therapy. Curr Opin Immunol (2018) 55:81–8. doi: 10.1016/j.coi.2018.10.008
    1. Ludwig RJ, Vanhoorelbeke K, Leypoldt F, Kaya Z, Bieber K, McLachlan SM, et al. . Mechanisms of Autoantibody-Induced Pathology. Front Immunol (2017) 8:603. doi: 10.3389/fimmu.2017.00603
    1. Berlyn KA, Schultes B, Leveugle B, Noujaim AA, Alexander RB, Mann DL. Generation of CD4(+) and CD8(+) T Lymphocyte Responses by Dendritic Cells Armed With PSA/anti-PSA (Antigen/Antibody) Complexes. Clin Immunol (2001) 101(3):276–83. doi: 10.1006/clim.2001.5115
    1. Junker F, Gordon J, Qureshi O. Fc Gamma Receptors and Their Role in Antigen Uptake, Presentation, and T Cell Activation. Front Immunol (2020) 11:1393. doi: 10.3389/fimmu.2020.01393
    1. Baker K, Rath T, Pyzik M, Blumberg RS. The Role of FcRn in Antigen Presentation. Front Immunol (2014) 5:408. doi: 10.3389/fimmu.2014.00408
    1. Erdei A, Kovacs KG, Nagy-Balo Z, Lukacsi S, Macsik-Valent B, Kurucz I, et al. . New Aspects in the Regulation of Human B Cell Functions by Complement Receptors CR1, CR2, CR3 and CR4. Immunol Lett (2021)237:42–57. doi: 10.1016/j.imlet.2021.06.006
    1. Altman EM. Novel Therapies for Pemphigus Vulgaris. Am J Clin Dermatol (2020) 21(6):765–82. doi: 10.1007/s40257-020-00544-w
    1. Schmidt E, Kasperkiewicz M, Joly P. Pemphigus. Lancet (2019) 394(10201):882–94. doi: 10.1016/S0140-6736(19)31778-7
    1. Liu D, Ahmet A, Ward L, Krishnamoorthy P, Mandelcorn ED, Leigh R, et al. . A Practical Guide to the Monitoring and Management of the Complications of Systemic Corticosteroid Therapy. Allergy Asthma Clin Immunol (2013) 9(1):30. doi: 10.1186/1710-1492-9-30
    1. Min DI, Monaco AP. Complications Associated With Immunosuppressive Therapy and Their Management. Pharmacotherapy (1991) 11(5):119S–25S.
    1. Vaccaro C, Zhou J, Ober RJ, Ward ES. Engineering the Fc Region of Immunoglobulin G to Modulate In Vivo Antibody Levels. Nat Biotechnol (2005) 23(10):1283–8. doi: 10.1038/nbt1143
    1. Ulrichts P, Guglietta A, Dreier T, van Bragt T, Hanssens V, Hofman E, et al. . Neonatal Fc Receptor Antagonist Efgartigimod Safely and Sustainably Reduces IgGs in Humans. J Clin Invest (2018) 128(10):4372–86. doi: 10.1172/JCI97911
    1. Howard JF, Jr., Bril V, Burns TM, Mantegazza R, Bilinska M, Szczudlik A, et al. . Randomized Phase 2 Study of FcRn Antagonist Efgartigimod in Generalized Myasthenia Gravis. Neurology (2019) 92(23):e2661–73. doi: 10.1212/WNL.0000000000007600
    1. Newland AC, Sanchez-Gonzalez B, Rejto L, Egyed M, Romanyuk N, Godar M, et al. . Phase 2 Study of Efgartigimod, A Novel FcRn Antagonist, in Adult Patients With Primary Immune Thrombocytopenia. Am J Hematol (2020) 95(2):178–87. doi: 10.1002/ajh.25680
    1. Goebeler M, Bata-Csorgo Z, De Simone C, Didona B, Remenyik E, Reznichenko N, et al. . Treatment of Pemphigus Vulgaris and Foliaceus With Efgartigimod, A Neonatal Fc Receptor Inhibitor: A Phase 2 Multicentre, Open-Label Feasibility Trial. Br J Dermatol (2021)186(3):429–39. doi: 10.1111/bjd.20782
    1. Hebert V, Boulard C, Houivet E, Duvert Lehembre S, Borradori L, Della Torre R, et al. . Large International Validation of ABSIS and PDAI Pemphigus Severity Scores. J Invest Dermatol (2019) 139(1):31–7. doi: 10.1016/j.jid.2018.04.042
    1. Drouot L, Hantz S, Jouen F, Velay A, Lamia B, Veber B, et al. . Evaluation of Humoral Immunity to SARS-CoV-2: Diagnostic Value of a New Multiplex Addressable Laser Bead Immunoassay. Front Microbiol (2020) 11:603931. doi: 10.3389/fmicb.2020.603931
    1. Muller R, Svoboda V, Wenzel E, Gebert S, Hunzelmann N, Muller HH, et al. . IgG Reactivity Against non-Conformational NH-Terminal Epitopes of the Desmoglein 3 Ectodomain Relates to Clinical Activity and Phenotype of Pemphigus Vulgaris. Exp Dermatol (2006) 15(8):606–14. doi: 10.1111/j.1600-0625.2006.00451.x
    1. Maho-Vaillant M, Perals C, Golinski ML, Hebert V, Caillot F, Mignard C, et al. . Rituximab and Corticosteroid Effect on Desmoglein-Specific B Cells and Desmoglein-Specific T Follicular Helper Cells in Pemphigus. J Invest Dermatol (2021)141(9):2132–40. doi: 10.1016/j.jid.2021.01.031
    1. Murrell DF, Dick S, Ahmed AR, Amagai M, Barnadas MA, Borradori L, et al. . Consensus Statement on Definitions of Disease, End Points, and Therapeutic Response for Pemphigus. J Am Acad Dermatol (2008) 58(6):1043–6. doi: 10.1016/j.jaad.2008.01.012
    1. Futei Y, Amagai M, Ishii K, Kuroda-Kinoshita K, Ohya K, Nishikawa T. Predominant IgG4 Subclass in Autoantibodies of Pemphigus Vulgaris and Foliaceus. J Dermatol Sci (2001) 26(1):55–61. doi: 10.1016/s0923-1811(00)00158-4
    1. Blumberg LJ, Humphries JE, Jones SD, Pearce LB, Holgate R, Hearn A, et al. . Blocking FcRn in Humans Reduces Circulating IgG Levels and Inhibits IgG Immune Complex-Mediated Immune Responses. Sci Adv (2019) 5(12):eaax9586. doi: 10.1126/sciadv.aax9586
    1. Granger V, Peyneau M, Chollet-Martin S, de Chaisemartin L. Neutrophil Extracellular Traps in Autoimmunity and Allergy: Immune Complexes at Work. Front Immunol (2019) 10:2824. doi: 10.3389/fimmu.2019.02824
    1. Cho A, Caldara AL, Ran NA, Menne Z, Kauffman RC, Affer M, et al. . Single-Cell Analysis Suggests That Ongoing Affinity Maturation Drives the Emergence of Pemphigus Vulgaris Autoimmune Disease. Cell Rep (2019) 28(4):909–22.e6. doi: 10.1016/j.celrep.2019.06.066
    1. Koutsakos M, Wheatley AK, Loh L, Clemens EB, Sant S, Nussing S, et al. . Circulating TFH Cells, Serological Memory, and Tissue Compartmentalization Shape Human Influenza-Specific B Cell Immunity. Sci Transl Med (2018) 10(428):eaan8405. doi: 10.1126/scitranslmed.aan8405
    1. Yuan H, Zhou S, Liu Z, Cong W, Fei X, Zeng W, et al. . Pivotal Role of Lesional and Perilesional T/B Lymphocytes in Pemphigus Pathogenesis. J Invest Dermatol (2017) 137(11):2362–70. doi: 10.1016/j.jid.2017.05.032
    1. Pollmann R, Schmidt T, Eming R, Hertl M. Pemphigus: A Comprehensive Review on Pathogenesis, Clinical Presentation and Novel Therapeutic Approaches. Clin Rev Allergy Immunol (2018) 54(1):1–25. doi: 10.1007/s12016-017-8662-z
    1. Werth VP, Culton DA, Concha JSS, Graydon JS, Blumberg LJ, Okawa J, et al. . 3rd. Safety, Tolerability, and Activity of ALXN1830 Targeting the Neonatal Fc Receptor in Chronic Pemphigus. J Invest Dermatol (2021) 141(12):2858–2865.e4. doi: 10.1016/j.jid.2021.04.031
    1. Colliou N, Picard D, Caillot F, Calbo S, Le Corre S, Lim A, et al. . Long-Term Remissions of Severe Pemphigus After Rituximab Therapy are Associated With Prolonged Failure of Desmoglein B Cell Response. Sci Transl Med (2013) 5(175):175ra30. doi: 10.1126/scitranslmed.3005166
    1. Abasq C, Mouquet H, Gilbert D, Tron F, Grassi V, Musette P, et al. . ELISA Testing of Anti-Desmoglein 1 and 3 Antibodies in the Management of Pemphigus. Arch Dermatol (2009) 145(5):529–35. doi: 10.1001/archdermatol.2009.9
    1. Mignard C, Maho-Vaillant M, Golinski ML, Balaye P, Prost-Squarcioni C, Houivet E, et al. . French Study Group on Autoimmune Bullous Skin D. Factors Associated With Short-Term Relapse in Patients With Pemphigus Who Receive Rituximab as First-Line Therapy: A Post Hoc Analysis of a Randomized Clinical Trial. JAMA Dermatol (2020) 156(5):545–52. doi: 10.1001/jamadermatol.2020.0290
    1. Payne AS, Ishii K, Kacir S, Lin C, Li H, Hanakawa Y, et al. . Genetic and Functional Characterization of Human Pemphigus Vulgaris Monoclonal Autoantibodies Isolated by Phage Display. J Clin Invest (2005) 115(4):888–99. doi: 10.1172/JCI24185
    1. Edwards G, Diercks GFH, Seelen MAJ, Horvath B, van Doorn MBA, Damman J. Complement Activation in Autoimmune Bullous Dermatoses: A Comprehensive Review. Front Immunol (2019) 10:1477. doi: 10.3389/fimmu.2019.01477
    1. Jordon RE, McDuffie FC. Serum and Blister Fluid Anticomplementary Activity in Pemphigus and Bullous Pemphigoid. Sucrose Density Gradient Studies. Proc Soc Exp Biol Med (1976) 151(3):594–8. doi: 10.3181/00379727-151-39267
    1. Boesch AW, Kappel JH, Mahan AE, Chu TH, Crowley AR, Osei-Owusu NY, et al. . Enrichment of High Affinity Subclasses and Glycoforms From Serum-Derived IgG Using FcgammaRs as Affinity Ligands. Biotechnol Bioeng (2018) 115(5):1265–78. doi: 10.1002/bit.26545
    1. Bruggeman CW, Dekkers G, Bentlage AEH, Treffers LW, Nagelkerke SQ, Lissenberg-Thunnissen S, et al. . Enhanced Effector Functions Due to Antibody Defucosylation Depend on the Effector Cell Fcgamma Receptor Profile. J Immunol (2017) 199(1):204–11. doi: 10.4049/jimmunol.1700116
    1. Hogarth PM, Pietersz GA. Fc Receptor-Targeted Therapies for the Treatment of Inflammation, Cancer and Beyond. Nat Rev Drug Discov (2012) 11(4):311–31. doi: 10.1038/nrd2909
    1. Tuijnman WB, Van Wichen DF, Schuurman HJ. Tissue Distribution of Human IgG Fc Receptors CD16, CD32 and CD64: An Immunohistochemical Study. APMIS (1993) 101(4):319–29. doi: 10.1111/j.1699-0463.1993.tb00117.x
    1. Mouquet H, Musette P, Gougeon ML, Jacquot S, Lemercier B, Lim A, et al. . B-Cell Depletion Immunotherapy in Pemphigus: Effects on Cellular and Humoral Immune Responses. J Invest Dermatol (2008) 128(12):2859–69. doi: 10.1038/jid.2008.178
    1. Hebert V, Petit M, Maho-Vaillant M, Golinski ML, Riou G, Derambure C, et al. . Modifications of the Transcriptomic Profile of Autoreactive B Cells From Pemphigus Patients After Treatment With Rituximab or a Standard Corticosteroid Regimen. Front Immunol (2019) 10:1794. doi: 10.3389/fimmu.2019.01794
    1. Yanis R, Bergua C, Christelle B, Maillot F, Bigot A, Beurier P, et al. . Neonatal Fc Receptor Expression in Lymphoid and Myeloid Cells in Systemic Lupus Erythematosus. Lupus (2021) 12:9612033211045049. doi: 10.1177/09612033211045049
    1. Mi W, Wanjie S, Lo ST, Gan Z, Pickl-Herk B, Ober RJ, et al. . Targeting the Neonatal Fc Receptor for Antigen Delivery Using Engineered Fc Fragments. J Immunol (2008) 181(11):7550–61. doi: 10.4049/jimmunol.181.11.7550
    1. Yan SX, Deng XM, Wang QT, Sun XJ, Wei W. Prednisone Treatment Inhibits the Differentiation of B Lymphocytes Into Plasma Cells in MRL/MpSlac-Lpr Mice. Acta Pharmacol Sin (2015) 36(11):1367–76. doi: 10.1038/aps.2015.76
    1. Nicholas MW, Dooley MA, Hogan SL, Anolik J, Looney J, Sanz I, et al. . A Novel Subset of Memory B Cells is Enriched in Autoreactivity and Correlates With Adverse Outcomes in SLE. Clin Immunol (2008) 126(2):189–201. doi: 10.1016/j.clim.2007.10.004
    1. Warnatz K, Wehr C, Drager R, Schmidt S, Eibel H, Schlesier M, et al. . Expansion of CD19(hi)CD21(lo/neg) B Cells in Common Variable Immunodeficiency (CVID) Patients With Autoimmune Cytopenia. Immunobiology (2002) 206(5):502–13. doi: 10.1078/0171-2985-00198
    1. Isnardi I, Ng YS, Menard L, Meyers G, Saadoun D, Srdanovic I, et al. . Complement Receptor 2/CD21- Human Naive B Cells Contain Mostly Autoreactive Unresponsive Clones. Blood (2010) 115(24):5026–36. doi: 10.1182/blood-2009-09-243071
    1. Liu Z, Zeng W, Huang X, Wang S, Zheng J, Pan M, et al. . Peripheral CD19(hi) B Cells Exhibit Activated Phenotype and Functionality in Promoting IgG and IgM Production in Human Autoimmune Diseases. Sci Rep (2017) 7(1):13921. doi: 10.1038/s41598-017-14089-2
    1. Golinski ML, Demeules M, Derambure C, Riou G, Maho-Vaillant M, Boyer O, et al. . CD11c(+) B Cells Are Mainly Memory Cells, Precursors of Antibody Secreting Cells in Healthy Donors. Front Immunol (2020) 11:32. doi: 10.3389/fimmu.2020.00032
    1. Murrell DF, Patsatsi A, Stavropoulos P, Baum S, Zeeli T, Kern JS, et al. . Investigators Bt. Proof of Concept for the Clinical Effects of Oral Rilzabrutinib, the First Bruton Tyrosine Kinase Inhibitor for Pemphigus Vulgaris: The Phase II BELIEVE Study. Br J Dermatol (2021)185(4):745–55. doi: 10.1111/bjd.20431
    1. Sanofi . Sanofi Provides Update on Phase 3 Study Evaluating Rilzabrutinib for the Treatment of Pemphigus. (2021).

Source: PubMed

3
Předplatit