1
|
Hematianlarki M, Nimmerjahn F. Immunomodulatory and anti-inflammatory properties of immunoglobulin G antibodies. Immunol Rev 2024. [PMID: 39340138 DOI: 10.1111/imr.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Antibodies provide an essential layer of protection from infection and reinfection with microbial pathogens. An impaired ability to produce antibodies results in immunodeficiency and necessitates the constant substitution with pooled serum antibodies from healthy donors. Among the five antibody isotypes in humans and mice, immunoglobulin G (IgG) antibodies are the most potent anti-microbial antibody isotype due to their long half-life, their ability to penetrate almost all tissues and due to their ability to trigger a wide variety of effector functions. Of note, individuals suffering from IgG deficiency frequently produce self-reactive antibodies, suggesting that a normal serum IgG level also may contribute to maintaining self-tolerance. Indeed, the substitution of immunodeficient patients with pooled serum IgG fractions from healthy donors, also referred to as intravenous immunoglobulin G (IVIg) therapy, not only protects the patient from infection but also diminishes autoantibody induced pathology, providing more direct evidence that IgG antibodies play an active role in maintaining tolerance during the steady state and during resolution of inflammation. The aim of this review is to discuss different conceptual models that may explain how serum IgG or IVIg can contribute to maintaining a balanced immune response. We will focus on pathways depending on the IgG fragment crystallizable (Fc) as pre-clinical data in various mouse model systems as well as human clinical data have demonstrated that the IgG Fc-domain recapitulates the ability of intact IVIg with respect to its ability to trigger resolution of inflammation. We will further discuss how the findings already have or are in the process of being translated to novel therapeutic approaches to substitute IVIg in treating autoimmune inflammation.
Collapse
Affiliation(s)
- Marjan Hematianlarki
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Murthy S, Patzelt S, Künstner A, Busch H, Schmidt E, Sadik CD. Intravenous Ig Ameliorates Disease in a Murine Model of Anti-Laminin 332 Mucous Membrane Pemphigoid. J Invest Dermatol 2024:S0022-202X(24)00304-X. [PMID: 38692406 DOI: 10.1016/j.jid.2024.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/09/2023] [Accepted: 02/24/2024] [Indexed: 05/03/2024]
Abstract
Intravenous Ig (IVIg) is used to treat mucous membrane pemphigoid, although its therapeutic effectivity is not sufficiently supported by randomized controlled clinical trials, and its mode of action is only insufficiently understood. We have examined the effect of IVIg in a mouse model of anti-laminin 332 mucous membrane pemphigoid and found that IVIg ameliorates both cutaneous and mucosal inflammatory lesions. Our investigation into the modes of action of IVIg in mucous membrane pemphigoid indicated effective anti-inflammatory mechanisms beyond the enhanced degradation of IgG mediated through inhibition of the FcRn. Our results suggest that IVIg curbs the activation of neutrophils at several levels. This includes a direct, immediate inhibitory effect on neutrophil activation by immune complexes but not C5a, which blunts the release of ROS and leukotriene B4 from neutrophils. IVIg also suppresses the formation of neutrophil extracellular traps in response to calcium ion ionophore. In vivo treatment with IVIg altered the transcriptome of blood leukocytes and bone marrow neutrophils toward less proinflammatory phenotypes. Collectively, our results support the effectivity of IVIg in the treatment of mucous membrane pemphigoid and indicate that effects on neutrophils at multiple levels may significantly contribute to its therapeutic effects.
Collapse
Affiliation(s)
- Sripriya Murthy
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany
| | - Sabrina Patzelt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Axel Künstner
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany; Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Center for Research on Inflammation of the Skin (CRIS), University of Lübeck, Lübeck, Germany
| | - Christian D Sadik
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany; Center for Research on Inflammation of the Skin (CRIS), University of Lübeck, Lübeck, Germany.
| |
Collapse
|
3
|
Pigors M, Patzelt S, Reichhelm N, Dworschak J, Khil'chenko S, Emtenani S, Bieber K, Hofrichter M, Kamaguchi M, Goletz S, Köhl G, Köhl J, Komorowski L, Probst C, Vanderheyden K, Balbino B, Ludwig RJ, Verheesen P, Schmidt E. Bullous pemphigoid induced by IgG targeting type XVII collagen non-NC16A/NC15A extracellular domains is driven by Fc gamma receptor- and complement-mediated effector mechanisms and is ameliorated by neonatal Fc receptor blockade. J Pathol 2024; 262:161-174. [PMID: 37929639 DOI: 10.1002/path.6220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/18/2023] [Accepted: 09/21/2023] [Indexed: 11/07/2023]
Abstract
Bullous pemphigoid (BP) is an autoimmune blistering disease characterized by autoantibodies targeting type XVII collagen (Col17) with the noncollagenous 16A (NC16A) ectodomain representing the immunodominant site. The role of additional extracellular targets of Col17 outside NC16A has not been unequivocally demonstrated. In this study, we showed that Col17 ectodomain-reactive patient sera depleted in NC16A IgG induced dermal-epidermal separation in a cryosection model indicating the pathogenic potential of anti-Col17 non-NC16A extracellular IgG. Moreover, injection of IgG targeting the murine Col17 NC14-1 domains (downstream of NC15A, the murine homologue of human NC16A) into C57BL/6J mice resulted in erythematous skin lesions and erosions. Clinical findings were accompanied by IgG/C3 deposits along the basement membrane and subepidermal blistering with inflammatory infiltrates. Disease development was significantly reduced in either Fc-gamma receptor (FcγR)- or complement-5a receptor-1 (C5aR1)-deficient mice. Inhibition of the neonatal FcR (FcRn), an atypical FcγR regulating IgG homeostasis, with the murine Fc fragment IgG2c-ABDEG, a derivative of efgartigimod, reduced anti-NC14-1 IgG levels, resulting in ameliorated skin inflammation compared with isotype-treated controls. These data demonstrate that the pathogenic effects of IgG targeting the Col17 domain outside human NC16A/murine NC15A are partly attributable to antibody-mediated FcγR- and C5aR1 effector mechanisms while pharmacological inhibition of the FcRn represents a promising treatment for BP. The mouse model of BP will be instrumental in further investigating the role of Col17 non-NC16A/NC15A extracellular epitopes and validating new therapies for this disease. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Manuela Pigors
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Sabrina Patzelt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Niklas Reichhelm
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Jenny Dworschak
- Institute of Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | | | - Shirin Emtenani
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Maxi Hofrichter
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Mayumi Kamaguchi
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Stephanie Goletz
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Gabriele Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lars Komorowski
- Institute of Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | - Christian Probst
- Institute of Experimental Immunology, EUROIMMUN AG, Lübeck, Germany
| | | | | | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany
| | | | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
4
|
Hundt JE, Sadik CD, van Beek N, Busch H, Caux F, Goebeler M, Hammers CM, Hartmann K, Hashimoto T, Ibrahim S, Kasperkiewicz M, Murrell DF, Recke A, Rose C, Schumacher N, Shimanovich I, Sitaru C, Terheyden P, Thaçi D, Ludwig RJ, Schmidt E. A life for autoimmune blistering diseases: in memoriam Detlef Zillikens. Front Immunol 2023; 14:1291590. [PMID: 38175817 PMCID: PMC10621786 DOI: 10.3389/fimmu.2023.1291590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 09/22/2023] [Indexed: 01/06/2024] Open
Abstract
Detlef Zillikens, MD, director and chair of the Department of Dermatology at the University of Lübeck, Lübeck, Germany, died in September 2022, aged only 64. He dedicated his professional life to autoimmune blistering diseases (AIBDs) and built his department into one of the world's leading centers for these diseases. Herein, his professional life and the impact on the field of AIBDs and the research landscape at the University of Lübeck are addressed. With his warm, integrative, open-minded, ever-optimistic attitude, he was a highly reliable colleague, mentor, and friend to many in the field including each of the authors. Combined with his in-depth knowledge of dermatology, interest in many fields of life science, and hard work, Detlef Zillikens initiated the founding of two independent research institutes, the Lübeck Institute of Experimental Dermatology and the Institute and Comprehensive Center for Inflammation Medicine. He was also instrumental in establishing the Center for Research on Inflammation of the Skin, where in a new research building, over 140 scientists pursue research questions related to skin inflammation. By inviting numerous researchers and clinicians to his department and hosting two large international meetings, he brought the field of AIBDs much closer together and inspired multiple national and international research initiatives. His ideas will live on and grow in many of his colleagues and mentees.
Collapse
Affiliation(s)
- Jennifer E. Hundt
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | | | - Nina van Beek
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Frédéric Caux
- Department of Dermatology, Groupe Hospitalier Paris Seine-Saint-Denis, AP-HP, Bobigny, France
| | - Matthias Goebeler
- Department of Dermatology, University of Würzburg, Würzburg, Germany
| | - Christoph M. Hammers
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Kiel, Kiel, Germany
| | | | - Takashi Hashimoto
- Department of Dermatology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Saleh Ibrahim
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Khalifa University, Abu Dhabi, United Arab Emirates
| | - Michael Kasperkiewicz
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Dedee F. Murrell
- Department of Dermatology, St George Hospital, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Andreas Recke
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Christian Rose
- Department of Dermatology, University of Lübeck, Lübeck, Germany
- Dermatohistologisches Einsendelabor Lübeck, Lübeck, Germany
| | - Nina Schumacher
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | | | - Cassian Sitaru
- Department of Dermatology, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | | | - Diamant Thaçi
- Institute and Comprehensive Center for Inflammation Medicine (CCIM), University of Lübeck, Lübeck, Germany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Pyzik M, Kozicky LK, Gandhi AK, Blumberg RS. The therapeutic age of the neonatal Fc receptor. Nat Rev Immunol 2023; 23:415-432. [PMID: 36726033 PMCID: PMC9891766 DOI: 10.1038/s41577-022-00821-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
IgGs are essential soluble components of the adaptive immune response that evolved to protect the body from infection. Compared with other immunoglobulins, the role of IgGs is distinguished and enhanced by their high circulating levels, long half-life and ability to transfer from mother to offspring, properties that are conferred by interactions with neonatal Fc receptor (FcRn). FcRn binds to the Fc portion of IgGs in a pH-dependent manner and protects them from intracellular degradation. It also allows their transport across polarized cells that separate tissue compartments, such as the endothelium and epithelium. Further, it is becoming apparent that FcRn functions to potentiate cellular immune responses when IgGs, bound to their antigens, form IgG immune complexes. Besides the protective role of IgG, IgG autoantibodies are associated with numerous pathological conditions. As such, FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases, with several FcRn-blocking strategies on the path to therapeutic use. Here, we describe the current state of knowledge of FcRn-IgG immunobiology, with an emphasis on the functional and pathological aspects, and an overview of FcRn-targeted therapy development.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Diseases Center, Boston, MA, USA.
| |
Collapse
|
6
|
Tešanović Perković D, Bukvić Mokos Z, Marinović B. Epidermolysis Bullosa Acquisita-Current and Emerging Treatments. J Clin Med 2023; 12:jcm12031139. [PMID: 36769788 PMCID: PMC9917799 DOI: 10.3390/jcm12031139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/10/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Epidermolysis bullosa acquisita (EBA) is a rare chronic autoimmune subepidermal blistering disease of the skin and mucous membranes, usually beginning in adulthood. EBA is induced by autoantibodies to type VII collagen, a major component of anchoring fibrils in the dermal-epidermal junction (DEJ). The binding of autoantibodies to type-VII collagen subsequently leads to the detachment of the epidermis and the formation of mucocutaneous blisters. EBA has two major clinical subtypes: the mechanobullous and inflammatory variants. The classic mechanobullous variant presentation consists of skin fragility, bullae with minimal clinical or histological inflammation, erosions in acral distribution that heal with scarring, and milia formation. The inflammatory variant is challenging to differentiate from other autoimmune bullous diseases, most commonly bullous pemphigoid (BP) but also mucous membrane pemphigoid (MMP), Brunsting-Perry pemphigoid, and linear IgA dermatosis. Due to its recalcitrance conventional treatment of epidermolysis bullosa acquisita is shown to be demanding. Here we discuss novel therapeutic strategies that have emerged and which could potentially improve the quality of life in patients with EBA.
Collapse
Affiliation(s)
| | - Zrinka Bukvić Mokos
- Department of Dermatology and Venereology, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia
| | - Branka Marinović
- Department of Dermatology and Venereology, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia
- Correspondence:
| |
Collapse
|
7
|
Ludwig RJ, Schmidt E. Bullous Diseases of the Skin and Mucous Membranes. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
8
|
Haeger SC, Kridin K, Pieper M, Griewahn L, Nimmerjahn F, Zillikens D, König P, Ludwig RJ, Hundt JE. Therapeutic effects of Fc gamma RIV inhibition are mediated by selectively blocking immune complex-induced neutrophil activation in epidermolysis bullosa acquisita. Front Immunol 2022; 13:938306. [PMID: 36311755 PMCID: PMC9606225 DOI: 10.3389/fimmu.2022.938306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022] Open
Abstract
Epidermolysis bullosa acquisita (EBA) is a subepidermal autoimmune bullous disease caused by autoantibodies targeting type VII collagen (COL7). It is characterized by inflammation and subepidermal blistering mainly through immune complex (IC)-mediated activation of neutrophils. In experimental EBA, binding of neutrophils to ICs in the skin and induction of clinical disease depends on the expression of the Fc gamma receptor (FcγR) IV. As activating FcγR mediate both neutrophil extravasation and activation, we used multiphoton imaging to obtain further insights into the mechanistic contribution of FcγRIV in the pathogenesis of EBA. First, we demonstrated that blocking FcγRIV function completely protects LysM-eGFP mice against induction of antibody transfer-induced EBA. To visualize the interactions of anti-COL7 IgG and neutrophils in vivo, fluorescently labeled anti-COL7 IgG was injected into LysM-eGFP mice. Multiphoton microscopy was sequentially performed over a period of 8 days. At all time points, we observed a significantly higher extravasation of neutrophils into the skin of mice treated with anti-FcγRIV antibody compared to controls. However, the percentage of detected neutrophils localized to the target antigen along the dermal-epidermal junction was comparable between both groups. Additionally, reactive oxygen release and migration in vitro assay data demonstrate that FcγRIV antibody treatment inhibits the activation, but not the migration, of neutrophils. Our findings underscore the importance of advanced in vivo imaging techniques to understand the complexity of IC-mediated neutrophil-dependent inflammation, and indicate that the therapeutic utility of FcγRIV blockade is achieved through impairment of IC-mediated neutrophil activation.
Collapse
Affiliation(s)
- Swantje C. Haeger
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
| | - Khalaf Kridin
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Mario Pieper
- Institute of Anatomy, University of Luebeck, Lubeck, Germany
| | - Laura Griewahn
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
| | - Falk Nimmerjahn
- Department of Biology, University of Erlangen-Nuremberg, Erlangen-Nuremberg, Germany
| | - Detlef Zillikens
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
- Department of Dermatology, University of Luebeck, Lubeck, Germany
| | - Peter König
- Institute of Anatomy, University of Luebeck, Lubeck, Germany
| | - Ralf J. Ludwig
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
- Department of Dermatology, University of Luebeck, Lubeck, Germany
| | - Jennifer E. Hundt
- Luebeck Institute of Experimental Dermatology, University of Luebeck, Lubeck, Germany
- *Correspondence: Jennifer E. Hundt,
| |
Collapse
|
9
|
Remlinger J, Madarasz A, Guse K, Hoepner R, Bagnoud M, Meli I, Feil M, Abegg M, Linington C, Shock A, Boroojerdi B, Kiessling P, Smith B, Enzmann V, Chan A, Salmen A. Antineonatal Fc Receptor Antibody Treatment Ameliorates MOG-IgG-Associated Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/2/e1134. [PMID: 35027475 PMCID: PMC8759074 DOI: 10.1212/nxi.0000000000001134] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD) is a rare, autoimmune demyelinating CNS disorder, distinct from multiple sclerosis and neuromyelitis optica spectrum disorder. Characterized by pathogenic immunoglobulin G (IgG) antibodies against MOG, a potential treatment strategy for MOGAD is to reduce circulating IgG levels, e.g., by interference with the IgG recycling pathway mediated by the neonatal Fc receptor (FcRn). Although the optic nerve is often detrimentally involved in MOGAD, the effect of FcRn blockade on the visual pathway has not been assessed. Our objective was to investigate effects of a monoclonal anti-FcRn antibody in murine MOG-IgG-associated experimental autoimmune encephalomyelitis (EAE). METHODS We induced active MOG35-55 EAE in C57Bl/6 mice followed by the application of a monoclonal MOG-IgG (8-18C5) 10 days postimmunization (dpi). Animals were treated with either a specific monoclonal antibody against FcRn (α-FcRn, 4470) or an isotype-matched control IgG on 7, 10, and 13 dpi. Neurologic disability was scored daily on a 10-point scale. Visual acuity was assessed by optomotor reflex. Histopathologic hallmarks of disease were assessed in the spinal cord, optic nerve, and retina. Immune cell infiltration was visualized by immunohistochemistry, demyelination by Luxol fast blue staining and complement deposition and number of retinal ganglion cells by immunofluorescence. RESULTS In MOG-IgG-augmented MOG35-55 EAE, anti-FcRn treatment significantly attenuated neurologic disability over the course of disease (mean area under the curve and 95% confidence intervals (CIs): α-FcRn [n = 27], 46.02 [37.89-54.15]; isotype IgG [n = 24], 66.75 [59.54-73.96], 3 independent experiments), correlating with reduced amounts of demyelination and macrophage infiltration into the spinal cord. T- and B-cell infiltration and complement deposition remained unchanged. Compared with isotype, anti-FcRn treatment prevented reduction of visual acuity over the course of disease (median cycles/degree and interquartile range: α-FcRn [n = 16], 0.50 [0.48-0.55] to 0.50 [0.48-0.58]; isotype IgG [n = 17], 0.50 [0.49-0.54] to 0.45 [0.39-0.51]). DISCUSSION We show preserved optomotor response and ameliorated course of disease after anti-FcRn treatment in an experimental model using a monoclonal MOG-IgG to mimic MOGAD. Selectively targeting FcRn might represent a promising therapeutic approach in MOGAD.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Adrian Madarasz
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Kirsten Guse
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Robert Hoepner
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Maud Bagnoud
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Ivo Meli
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Moritz Feil
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Mathias Abegg
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Christopher Linington
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anthony Shock
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Babak Boroojerdi
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Peter Kiessling
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Bryan Smith
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Volker Enzmann
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Andrew Chan
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany
| | - Anke Salmen
- From the Department of Neurology (J.R., A.M., K.G., R.H., M.B., I.M., A.C., A. Salmen), Inselspital, Bern University Hospital and University of Bern; Department of Biomedical Research (J.R., A.M., K.G., R.H., M.B., I.M., V.E., A.C., A. Salmen), and Graduate School for Cellular and Biomedical Sciences (J.R., A.M., M.B.), University of Bern; Department of Ophthalmology (M.F., M.A., V.E.), Inselspital, Bern University Hospital and University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow; UCB Pharma (A. Shock, B.S.), Slough, United Kingdom; and UCB Pharma (B.B., P.K.), Monheim-am-Rhein, Germany.
| |
Collapse
|
10
|
Wang L, Liu L, Hong X, Liu D, Cheng Z. Delanzomib, a Novel Proteasome Inhibitor, Combined With Adalimumab Drastically Ameliorates Collagen-Induced Arthritis in Rats by Improving and Prolonging the Anti-TNF-α Effect of Adalimumab. Front Pharmacol 2021; 12:782385. [PMID: 34880764 PMCID: PMC8645831 DOI: 10.3389/fphar.2021.782385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Delanzomib is a novel proteasome inhibitor initially developed for treating multiple myeloma. It was found to inhibit the expression of tumor necrosis factor alpha (TNF-α). This study aimed to investigate the ameliorating effect of delanzomib on collagen-induced arthritis (CIA) and to explore the pharmacodynamics and pharmacokinetics (PK) interactions between delanzomib and adalimumab. Rats with CIA were randomly assigned to receive the treatment with delanzomib, adalimumab, delanzomib combined with adalimumab, or placebo. Visual inspection and biochemical examinations including TNF-α, interleukin 6, and C-reactive protein were performed to assess arthritis severity during the treatment. The adalimumab concentration in rats was determined to evaluate the PK interaction between delanzomib and adalimumab. Also, the levels of neonatal Fc receptor (FcRn) and FcRn mRNA were measured to explore the role of FcRn in the PK interaction between delanzomib and adalimumab. As a result, delanzomib combined with adalimumab exhibited stronger anti-arthritis activity than a single drug because both drugs synergistically reduced TNF-α level in vivo. Delanzomib also decreased adalimumab elimination in rats by increasing the level of FcRn. The slower elimination of adalimumab in rats further prolonged the anti-TNF-α effect of adalimumab. Moreover, FcRn level was increased by delanzomib via suppressing FcRn degradation rather than promoting FcRn production. In conclusion, delanzomib combined with adalimumab may be a potential therapeutic approach for treating rheumatoid arthritis. The initial finding that the PK interaction occurred between delanzomib and adalimumab may have clinical relevance for patients who simultaneously take proteasome inhibitors and anti-TNF-α therapeutic proteins.
Collapse
Affiliation(s)
- Lei Wang
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Lixiong Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Zeneng Cheng
- Research Institute of Drug Metabolism and Pharmacokinetics, School of Xiangya Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
11
|
García-Serra A, Radosevic M, Ríos J, Aguilar E, Maudes E, Landa J, Sabater L, Martinez-Hernandez E, Planagumà J, Dalmau J. Blocking Placental Class G Immunoglobulin Transfer Prevents NMDA Receptor Antibody Effects in Newborn Mice. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1061. [PMID: 34580181 PMCID: PMC8477376 DOI: 10.1212/nxi.0000000000001061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/15/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVES To determine in a mouse model whether neonatal Fc receptor (FcRn) blockade prevents the placental transfer of class G immunoglobulin (IgG) derived from patients with anti-NMDA receptor (NMDAR) encephalitis and their pathogenic effects on the fetuses and offspring. METHODS Pregnant C57BL/6J mice were administered via tail vein FcRn antibody (FcRn-ab) or saline solution 6 hours before administration of patients' or controls' IgG on days 14, 15, and 16 of gestation. Three experimental groups were established: mice receiving controls' IgG, patients' IgG, or patients' IgG along with pretreatment with FcRn-ab. Immunohistochemical staining, confocal microscopy, hippocampal long-term potentiation, and standardized developmental and behavioral tasks were used to assess the efficacy of treatment with FcRn-ab. RESULTS In pregnant mice that received patients' IgG, treatment with FcRn-ab prevented the IgG from reaching the fetal brain, abrogating the decrease of NMDAR clusters and the reduction of cortical plate thickness that were observed in fetuses from untreated pregnant mice. Moreover, among the offspring of mothers that received patients' IgG, those whose mothers were treated with FcRn-ab did not develop the alterations that occurred in offspring of untreated mothers, including impairment in hippocampal plasticity, delay in innate reflexes, and visuospatial memory deficits. DISCUSSION FcRn blockade prevents placental transfer of IgG from patients with anti-NMDAR encephalitis and abrogates the synaptic and neurodevelopmental alterations caused by patients' antibodies. This model has potential therapeutic implications for other antibody-mediated diseases of the CNS during pregnancy.
Collapse
Affiliation(s)
- Anna García-Serra
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Marija Radosevic
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - José Ríos
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Esther Aguilar
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Estibaliz Maudes
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Jon Landa
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Lidia Sabater
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Eugenia Martinez-Hernandez
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Jesús Planagumà
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain
| | - Josep Dalmau
- From the Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) (A.G.-S., M.R., E.A., E.M., J.L., L.S., E.M.-H., J.P., J.D.), Hospital Clínic, Universitat de Barcelona; Medical Statistics Core Facility (J.R.), IDIBAPS and Hospital Clínic; Biostatistics Unit (J.R.), School of Medicine, Universitat Autònoma de Barcelona, Spain; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; and Institució Catalana de Recerca i Estudis Avançats (ICREA) (J.D.), Barcelona, Spain.
| |
Collapse
|
12
|
van Beek N, Zillikens D, Schmidt E. Bullous Autoimmune Dermatoses–Clinical Features, Diagnostic Evaluation, and Treatment Options. DEUTSCHES ARZTEBLATT INTERNATIONAL 2021; 118:413-420. [PMID: 34369370 PMCID: PMC8380840 DOI: 10.3238/arztebl.m2021.0136] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/03/2020] [Accepted: 01/27/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bullous autoimmune dermatoses are a clinically and immunopatho - logically heterogeneous group of diseases, characterized clinically by blisters or erosions of the skin and/or mucous membranes. In Germany, their prevalence is approximately 40 000 cases nationwide, and their incidence approximately 20 new cases per million people per year. METHODS This review is based on publications that were retrieved by a selective search of the literature focusing on the current German and European guidelines. RESULTS Recent years have seen the publication of guidelines, controlled prospective clinical trials, and multicenter diagnostic studies improving both diagnosis and therapy. Specific monovalent and multivariate serological test systems and pattern analysis of tissue-bound autoantibodies allow identification of the target antigens in 80-90% of patients. This enables the precise classification of disease entities, with implications for treatment selection and disease outcome. In 2019, the anti-CD20 antibody rituximab was approved by the European Medicines Agency for the treatment of moderate and severe pemphigus vulgaris, with an ensuing marked improvement in the care of the affected patients. To treat mild and moderate bullous pemphigoid, topical clobetasol proprionate is recommended, in severe disease, combined with systemic treatment, i.e. usually (a) prednisolone p.o. at an initial dose of 0.5mg/kg/d , (b) an immunomodulant, e.g. dapsone or doxycycline, or (c) prednisolone plus an immunomodulant. CONCLUSION The early recognition and precise diagnostic evaluation of bullous autoimmune dermatoses now enables improved, often interdisciplinary treatment, in accordance with the available guidelines. Current research projects are focused on new treatment approaches, an improved understanding of the underlying pathophysiology, and further refinements of diagnostic techniques.
Collapse
Affiliation(s)
- Nina van Beek
- Department of Dermatology, Venereology, and Allergology, University of Lübeck, Lübeck, Germany
| | - Detlef Zillikens
- Department of Dermatology, Venereology, and Allergology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Department of Dermatology, Venereology, and Allergology, University of Lübeck, Lübeck, Germany
- Lu¨beck Institute of Experimental Dermatology (LIED), University of Lu¨beck, Lu¨beck, Germany
| |
Collapse
|
13
|
Coutinho E, Jacobson L, Shock A, Smith B, Vernon A, Vincent A. Inhibition of Maternal-to-Fetal Transfer of IgG Antibodies by FcRn Blockade in a Mouse Model of Arthrogryposis Multiplex Congenita. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/4/e1011. [PMID: 34045306 PMCID: PMC8161539 DOI: 10.1212/nxi.0000000000001011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/17/2021] [Indexed: 11/15/2022]
Abstract
Objective To determine whether blocking the neonatal Fc receptor (FcRn) during gestation with an anti-FcRn monoclonal antibody (mAb) reduces transfer of pathogenic maternal antibodies in utero and decreases the likelihood of maternal antibody-mediated neonatal disease in the offspring. Methods Using a previously established maternal-to-fetal transfer mouse model of arthrogryposis multiplex congenita (AMC), we assessed the effect of 4470, an anti-FcRn mAb, on the transfer of total human immunoglobulin G (IgG) and specific acetylcholine receptor (AChR)-antibodies from mother to fetus, as well as its effect on the prevention of neurodevelopmental abnormalities in the offspring. Results Offspring of pregnant dams treated with 4470 during gestation showed a substantial reduction in total human IgG and AChR antibody levels compared with those treated with the isotype mAb control. Treatment with 4470 was also associated with a significant reduction in AMC-IgG–induced deformities (limb or spinal curve malformations) when compared with mAb control–exposed embryos and a nonsignificant increase in the percentage of fetuses showing spontaneous movements. 4470 exposure during pregnancy was not associated with changes in general parameters of maternal well-being or fetal development; indeed, male neonates showed faster weight gain and shorter time to reach developmental milestones. Conclusions FcRn blockade is a promising therapeutic strategy to prevent the occurrence of AMC and other human maternal autoantibody-related diseases in the offspring.
Collapse
Affiliation(s)
- Ester Coutinho
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom
| | - Leslie Jacobson
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom
| | - Anthony Shock
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom
| | - Bryan Smith
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom
| | - Anthony Vernon
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom
| | - Angela Vincent
- From the Department of Basic and Clinical Neuroscience (E.C., A. Vernon), Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute; Medical Research Council Centre for Neurodevelopmental Disorders (E.C., A. Vernon), King's College London; Nuffield Department of Clinical Neurosciences (L.J., A. Vincent), University of Oxford; and UCB Pharma (A.S., B.S.), Slough, United Kingdom.
| |
Collapse
|
14
|
Qi T, Cao Y. In Translation: FcRn across the Therapeutic Spectrum. Int J Mol Sci 2021; 22:3048. [PMID: 33802650 PMCID: PMC8002405 DOI: 10.3390/ijms22063048] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.
Collapse
Affiliation(s)
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA;
| |
Collapse
|
15
|
Patel DD, Bussel JB. Neonatal Fc receptor in human immunity: Function and role in therapeutic intervention. J Allergy Clin Immunol 2021; 146:467-478. [PMID: 32896307 DOI: 10.1016/j.jaci.2020.07.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023]
Abstract
The humoral immune response provides specific, long-lived protection against invading pathogens, via immunoglobulin production and other memory functions. IgG, the most abundant immunoglobulin isotype, has the longest half-life and protects against bacterial and viral infections. The neonatal Fc receptor (FcRn) transports IgG across barriers, for example, the placenta, enhancing fetal humoral immunity to levels similar to their mothers'. Importantly, FcRn, by protecting IgG from intracellular degradation, results in an approximately 21-day circulating IgG half-life and high plasma levels; similarly, FcRn recycles albumin and is the portal of entry for enteric cytopathic human orphan (echo) virus infection. Dysregulated immune responses may lead to antibodies against self-antigens (autoantibodies), resulting in organ-specific or systemic autoimmune diseases. Autoantibody-mediated diseases have been treated by nonspecific immunoglobulin-lowering/modulating therapies, including immunoadsorption, plasma exchange, and high-dose intravenous immunoglobulin. However, targeting FcRn with specific inhibitors results in reduction in only IgG levels. The effectiveness of FcRn inhibitors in autoimmune diseases, including myasthenia gravis and immune thrombocytopenia, provides further evidence that IgG is a primary driver in these autoantibody-mediated diseases. We describe the role of FcRn in human biology, including insights that clinical testing of FcRn inhibitors have provided into FcRn biology and autoimmune disease mechanisms, allowing fact-based speculation on their therapeutic potential.
Collapse
Affiliation(s)
- Dhavalkumar D Patel
- UCB Pharma, Brussels, Belgium; University of North Carolina, Chapel Hill, NC.
| | | |
Collapse
|
16
|
Kasprick A, Hofrichter M, Smith B, Ward P, Bieber K, Shock A, Ludwig RJ, Schmidt E. Treatment with anti-neonatal Fc receptor (FcRn) antibody ameliorates experimental epidermolysis bullosa acquisita in mice. Br J Pharmacol 2020; 177:2381-2392. [PMID: 31975370 PMCID: PMC7174883 DOI: 10.1111/bph.14986] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/04/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023] Open
Abstract
Background and Purpose Pemphigus and pemphigoid diseases are characterized and caused predominantly by IgG autoantibodies targeting structural proteins of the skin. Their current treatment relies on general and prolonged immunosuppression that causes severe adverse events, including death. Hence, novel safe and more effective treatments are urgently needed. Due to its' physiological functions, the neonatal Fc receptor (FcRn) has emerged as a potential therapeutic target for pemphigus and pemphigoid, primarily because IgG is protected from proteolysis after uptake into endothelial cells. Thus, blockade of FcRn would reduce circulating autoantibody concentrations. However, long‐term effects of pharmacological FcRn inhibition in therapeutic settings of autoimmune diseases are unknown. Experimental Approach Therapeutic effects of FcRn blockade were investigated in a murine model of the prototypical autoantibody‐mediated pemphigoid disease, epidermolysis bullosa acquisita (EBA). B6.SJL‐H2s C3c/1CyJ mice with clinically active disease were randomized to receive either an anti‐FcRn monoclonal antibody (4470) or an isotype control over 4 weeks. Key Results While clinical disease continued to worsen in isotype control‐treated mice, overall disease severity continuously decreased in mice injected with 4470, leading to almost complete remission in over 25% of treated mice. These clinical findings were paralleled by a reduction of autoantibody concentrations. Reduction of autoantibody concentrations, rather than modulating neutrophil activation, was responsible for the observed therapeutic effects. Conclusion and Implications The clinical efficacy of anti‐FcRn treatment in this prototypical autoantibody‐mediated disease encourages further development of anti‐FcRn antibodies for clinical use in pemphigoid diseases and potentially in other autoantibody mediated diseases.
Collapse
Affiliation(s)
- Anika Kasprick
- Lübeck Institute of Experimental Dermatology, and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Maxi Hofrichter
- Lübeck Institute of Experimental Dermatology, and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | | | | | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | | | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology, and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| |
Collapse
|