151
|
Baerenwaldt A, Biburger M, Nimmerjahn F. Mechanisms of action of intravenous immunoglobulins. Expert Rev Clin Immunol 2010; 6:425-34. [PMID: 20441428 DOI: 10.1586/eci.10.9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Intravenous immunoglobulin (IVIg) has been used for nearly three decades as an efficient anti-inflammatory therapeutic regimen in a growing number of autoimmune diseases. Despite this their success in clinical application, the mechanism of action of IVIg therapy remains elusive. During the last few years, several mechanisms dependent on either the IgG variable or constant fragment have been proposed to explain the potent immunomodulatory activity of IVIg. This review will discuss which molecular and cellular pathways might be involved in the anti-inflammatory activity of IVIg and for which types of autoimmune diseases they might be relevant.
Collapse
Affiliation(s)
- Anne Baerenwaldt
- Department of Biology, Institute of Genetics, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | | | | |
Collapse
|
152
|
Sesarman A, Vidarsson G, Sitaru C. The neonatal Fc receptor as therapeutic target in IgG-mediated autoimmune diseases. Cell Mol Life Sci 2010; 67:2533-50. [PMID: 20217455 PMCID: PMC11115620 DOI: 10.1007/s00018-010-0318-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 01/29/2010] [Accepted: 02/12/2010] [Indexed: 01/01/2023]
Abstract
Therapy approaches based on lowering levels of pathogenic autoantibodies represent rational, effective, and safe treatment modalities of autoimmune diseases. The neonatal Fc receptor (FcRn) is a major factor regulating the serum levels of IgG antibodies. While FcRn-mediated half-life extension is beneficial for IgG antibody responses against pathogens, it also prolongs the serum half-life of IgG autoantibodies and thus promotes tissue damage in autoimmune diseases. In the present review article, we examine current evidence on the relevance of FcRn in maintaining high autoantibody levels and discuss FcRn-targeted therapeutic approaches. Further investigation of the FcRn-IgG interaction will not only provide mechanistic insights into the receptor function, but should also greatly facilitate the design of therapeutics combining optimal pharmacokinetic properties with the appropriate antibody effector functions in autoimmune diseases.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Cassian Sitaru
- Department of Dermatology, University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
- Centre for Biological Signalling Studies (bioss), University of Freiburg, Freiburg, Germany
| |
Collapse
|
153
|
The Thomsen-Friedenreich antigen-binding lectin jacalin interacts with desmoglein-1 and abrogates the pathogenicity of pemphigus foliaceus autoantibodies in vivo. J Invest Dermatol 2010; 130:2773-80. [PMID: 20631728 DOI: 10.1038/jid.2010.209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pemphigus foliaceus (PF) is an autoimmune skin blistering disease mediated by pathogenic autoantibodies against the desmosomal core glycoprotein desmoglein-1 (Dsg1). This study demonstrated that the O-glycan-specific plant lectin jacalin binds Dsg1 and inhibits the interaction of Dsg1/PF IgG. N-glycosylation is not involved in the interaction of Dsg1/jacalin or Dsg1/PF IgG. Subcutaneous injection of jacalin into neonatal mice drastically reduced PF IgG deposition at the epidermal cell surface and blocked PF IgG-induced skin blisters, both clinically and histologically. Interestingly, another plant lectin, peanut agglutinin, which shares the same carbohydrate specificity toward the O-linked carbohydrate structure known as Thomsen-Friedenreich antigen (TF antigen, Galβ1-3GalNAcα-O-Ser/Thr), also bound Dsg1 and blocked the skin blistering. In contrast, the plant lectin vicia villosa-B4 (VVL-B4), which shares the carbohydrate specificity toward the O-linked monosaccharide known as Thomsen-nouveau antigen (GalNAc-α1-O-Ser/Thr), did not bind Dsg1 and did not show a protective effect against the disease induced by the autoantibodies. Collectively, these results suggest that the binding of jacalin to O-linked TF carbohydrate motifs on Dsg1 impairs the Dsg1/PF autoantibody interactions and abrogates its pathogenicity in vivo. TF-specific binding ligands may have a potential therapeutic value for PF.
Collapse
|
154
|
High-dose intravenous immunoglobulin (IVIG) therapy in autoimmune skin blistering diseases. Clin Rev Allergy Immunol 2010; 38:186-95. [PMID: 19557317 DOI: 10.1007/s12016-009-8153-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Treatment of autoimmune bullous skin diseases can often be challenging and primarily consists of systemic corticosteroids and a variety of immunosuppressants. Current treatment strategies are effective in most cases but hampered by the side effects of long-term immunosuppressive treatment. Intravenous immunoglobulin (IVIG) is one potential promising therapy for patients with autoimmune bullous skin diseases, and evidence of its effectiveness and safety is increasing. A number of autoimmune bullous skin diseases have been identified in which IVIG treatment may be beneficial. However, experience with IVIG in patients with autoimmune skin blistering disease is limited, where it is recommended for patients not responding to conventional therapy. The mode of action of IVIG in autoimmune diseases, including bullous diseases is far from being completely understood. We here summarize the clinical evidence supporting the notion, that IVIG is a promising therapeutic agent for the treatment of patients with autoimmune bullous skin disease. In addition, we review the proposed modes of action. In the future, randomized controlled trials are necessary to better determine the efficacy and adverse effects of IVIG in the treatment of autoimmune bullous skin diseases. In addition, insights into IVIG's mode of action might enable us to develop novel therapeutics to overcome the current shortage of IVIG.
Collapse
|
155
|
|
156
|
Friedman DM, Llanos C, Izmirly PM, Brock B, Byron J, Copel J, Cummiskey K, Dooley MA, Foley J, Graves C, Hendershott C, Kates R, Komissarova EV, Miller M, Paré E, Phoon CKL, Prosen T, Reisner D, Ruderman E, Samuels P, Yu JK, Kim MY, Buyon JP. Evaluation of fetuses in a study of intravenous immunoglobulin as preventive therapy for congenital heart block: Results of a multicenter, prospective, open-label clinical trial. ACTA ACUST UNITED AC 2010; 62:1138-46. [PMID: 20391423 DOI: 10.1002/art.27308] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE The recurrence rate of anti-SSA/Ro-associated congenital heart block (CHB) is 17%. Sustained reversal of third-degree block has never been achieved. Based on potential reduction of maternal autoantibody titers as well as fetal inflammatory responses, intravenous immunoglobulin (IVIG) was evaluated as preventive therapy for CHB. METHODS A multicenter, prospective, open-label study based on Simon's 2-stage optimal design was initiated. Enrollment criteria included the presence of anti-SSA/Ro antibodies in the mother, birth of a previous child with CHB/neonatal lupus rash, current treatment with < or = 20 mg/day of prednisone, and <12 weeks pregnant. IVIG (400 mg/kg) was given every 3 weeks from week 12 to week 24 of gestation. The primary outcome was the development of second-degree or third-degree CHB. RESULTS Twenty mothers completed the IVIG protocol before the predetermined stopping rule of 3 cases of advanced CHB in the study was reached. CHB was detected at 19, 20, and 25 weeks; none of the cases occurred following the finding of an abnormal PR interval on fetal Doppler monitoring. One of these mothers had 2 previous children with CHB. One child without CHB developed a transient rash consistent with neonatal lupus. Sixteen children had no manifestations of neonatal lupus at birth. No significant changes in maternal titers of antibody to SSA/Ro, SSB/La, or Ro 52 kd were detected over the course of therapy or at delivery. There were no safety issues. CONCLUSION This study establishes the safety of IVIG and the feasibility of recruiting pregnant women who have previously had a child with CHB. However, IVIG at low doses consistent with replacement does not prevent the recurrence of CHB or reduce maternal antibody titers.
Collapse
|
157
|
Mutasim DF. Autoimmune bullous dermatoses in the elderly: an update on pathophysiology, diagnosis and management. Drugs Aging 2010; 27:1-19. [PMID: 20030429 DOI: 10.2165/11318600-000000000-00000] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Elderly individuals are susceptible to autoimmune bullous dermatoses (ABDs), which may be associated with high morbidity and mortality. ABDs result from an autoimmune response to components of the basement membrane zone at the dermal-epidermal junction or desmosomes. Bullous pemphigoid results from autoimmunity to hemidesmosomal proteins present in the basement membrane of stratified squamous epithelia. Patients present with tense blisters in flexural areas of the skin. Mild disease may be treated with potent topical corticosteroids, while extensive disease usually requires systemic corticosteroids or systemic immunosuppressive agents such as azathioprine. Mucosal pemphigoid affects one or more mucous membranes that are lined by stratified squamous epithelia. The two most commonly involved sites are the eye and the oral cavity. Lesions frequently result in scar formation that may cause blindness. Patients with severe disease or ocular involvement require aggressive therapy with corticosteroids and cyclophosphamide. Epidermolysis bullosa acquisita results from autoimmunity to type VII collagen in the anchoring fibrils of the basement membrane. Lesions may either arise on an inflammatory base or be non-inflammatory and result primarily from trauma. Treatment options include corticosteroids, dapsone, ciclosporin, methotrexate and plasmapheresis/immunoapheresis. Paraneoplastic pemphigus results from autoimmunity to multiple desmosomal antigens. The disorder is associated with neoplasms, especially leukaemia, lymphoma and thymoma. Patients present with stomatitis and polymorphous skin eruption. The disease may respond to successful treatment of the underlying neoplasm or may require immunosuppressive therapy.
Collapse
Affiliation(s)
- Diya F Mutasim
- Department of Dermatology, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, P.O. Box 670592, Cincinnati, OH 45267-0592, USA.
| |
Collapse
|
158
|
McDonnell KA, Low SC, Hoehn T, Donnelly R, Palmieri H, Fraley C, Sakorafas P, Mezo AR. Synthesis and structure-activity relationships of dimeric peptide antagonists of the human immunoglobulin G-human neonatal Fc receptor (IgG-FcRn) interaction. J Med Chem 2010; 53:1587-96. [PMID: 20092334 DOI: 10.1021/jm901128z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor, FcRn, regulates the half-life of IgG in vivo and may be a target in the treatment of autoimmune disease. Monomeric peptide antagonists of the human IgG-human FcRn interaction were dimerized using three different synthetic methodologies: thiol/alkyl halide coupling of unprotected peptides, reductive alkylation of unprotected peptides, and on-resin amide bond formation with protected peptides. It was found that dimerization of monomeric peptides increased the in vitro activity of the peptide monomers more than 200-fold. Human IgG catabolism experiments in human FcRn transgenic mice were used to assess the in vivo activity of peptide dimers that possessed different linkers, cyclizations, and affinities for FcRn. Overall, it was found that the linker joining two monomeric peptides had only a minor effect on the in vitro potency but that in vitro potency was predictive of in vivo activity.
Collapse
Affiliation(s)
- Kevin A McDonnell
- Syntonix Pharmaceuticals, Inc., a Subsidiary of Biogen Idec, 9 Fourth Avenue, Waltham, Massachusetts 02451, USA
| | | | | | | | | | | | | | | |
Collapse
|
159
|
|
160
|
AOYAMA Y. What’s new in i.v. immunoglobulin therapy and pemphigus: High-dose i.v. immunoglobulin therapy and its mode of action for treatment of pemphigus. J Dermatol 2010; 37:239-45. [DOI: 10.1111/j.1346-8138.2009.00796.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
161
|
Bieber K, Sun S, Ishii N, Kasperkiewicz M, Schmidt E, Hirose M, Westermann JÃ, Yu X, Zillikens D, Ludwig RJ. Animal models for autoimmune bullous dermatoses. Exp Dermatol 2010; 19:2-11. [DOI: 10.1111/j.1600-0625.2009.00948.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
162
|
Aschermann S, Lux A, Baerenwaldt A, Biburger M, Nimmerjahn F. The other side of immunoglobulin G: suppressor of inflammation. Clin Exp Immunol 2009; 160:161-7. [PMID: 20041883 DOI: 10.1111/j.1365-2249.2009.04081.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Immunoglobulin G (IgG) molecules can have two completely opposite functions. On one hand, they induce proinflammatory responses and recruit innate immune effector cells during infection with pathogenic microorganisms or autoimmune disease. On the other hand, intravenous infusion of high doses of pooled IgG molecules from thousands of donors [intravenous IG (IVIG) therapy] represents an efficient anti-inflammatory treatment for many autoimmune diseases. Whereas our understanding of the mechanism of the proinflammatory activity of IgG is quite advanced, we are only at the very beginning to comprehend how the anti-inflammatory activity comes about and what cellular and molecular players are involved in this activity. This review will summarize our current knowledge and focus upon the two major models of either IVIG-mediated competition for IgG-triggered effector functions or IVIG-mediated adjustment of cellular activation thresholds used to explain the mechanism of the anti-inflammatory activity.
Collapse
Affiliation(s)
- S Aschermann
- Laboratory of Experimental Immunology and Immunotherapy, Nikolaus-Fiebiger-Centre for Molecular Medicine, Medical Department III, University of Erlangen-Nuernberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
163
|
Mouthon L, Bussone G, Kaveri S. Indications et mécanismes d’action des immunoglobulines intraveineuses dans les pathologies auto-immunes et inflammatoires systémiques. Rev Med Interne 2009; 30:H14-20. [DOI: 10.1016/s0248-8663(09)73168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
164
|
Masuda A, Yoshida M, Shiomi H, Morita Y, Kutsumi H, Inokuchi H, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Role of Fc Receptors as a therapeutic target. ACTA ACUST UNITED AC 2009; 8:80-6. [PMID: 19275696 DOI: 10.2174/187152809787582525] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcgammaR), high-affinity IgE receptor (FcepsilonRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcgammaRs have been well known to play an important role in many biologic processes including those associated with the response to infection and cancer as well as in the pathogenesis of immune-mediated diseases. Both positive and negative regulatory function has ascribed to Fc receptors and FcgammaRs in particular which serve to establish a threshold for immune cell activation. In other cases, Fc receptors such as FcRn possess a novel structure and function by playing a major role in the transport of IgG across polarized epithelial barriers at mucosal surfaces and in the regulation of IgG half-life. These diverse functions highlight the potential effectiveness of targeting Fc receptors for therapeutic purposes. This review summarizes new information available in the therapeutic applications of this biology.
Collapse
Affiliation(s)
- Atsuhiro Masuda
- Department of Gastroenterology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Schmidt E. [Optimizing therapy in patients with severe autoimmune blistering skin diseases]. Hautarzt 2009; 60:633-40. [PMID: 19536513 DOI: 10.1007/s00105-008-1680-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Autoimmune bullous diseases are a heterogeneous group of disorders that can be subdivided according to the level of split formation in the intraepidermal blistering pemphigus diseases and subepidermal bullous disorders, latter including pemphigoid diseases, epidermolysis bullosa acquisita (EBA), and dermatitis herpetiformis. In the majority of autoimmune bullous disorders, disease activity can be sufficiently controlled by systemic corticosteroids in combination with further immunsuppressants/-modulants such as dapsone, doxycycline, azathioprine, mycophenolate mofetil, or methotrexate. In contrast, in pemphigus, mucous membrane pemphigoid, and EBA, treatment is challenging and conventional immunosuppressive therapy induces clinical remission only in a minority of patients. Until recently, only cyclosphosphamide and high-dose intravenous immunoglobulin (IVIG) were available as potent second-line therapies. Meanwhile, immunoadsorption and the monoclonal anti-CD20 antibody rituximab have been established as further therapeutic options. The present review focuses on efficacy, adverse events, treatment protocols, and mechanisms of action of IVIG, immunoadsorption, and rituximab in the treatment of severe and/or refractory patients with bullous autoimmune diseases.
Collapse
Affiliation(s)
- E Schmidt
- Klinik für Dermatologie, Allergologie und Venerologie, Universität zu Lübeck, Ratzeburger Allee 160, 23538 Lübeck.
| |
Collapse
|
166
|
Imbach P, Lazarus AH, Kühne T. Intravenous immunoglobulins induce potentially synergistic immunomodulations in autoimmune disorders. Vox Sang 2009; 98:385-94. [PMID: 19821958 DOI: 10.1111/j.1423-0410.2009.01264.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The increase in platelets in patients with immune thrombocytopenia (ITP) by intravenous administration of human immunoglobulin concentrates (IVIG) reflects a therapeutic immunomodulatory intervention targeted at the disturbed immune response in many inflammatory and autoimmune disorders. These immunoglobulin concentrates contain large numbers of antibodies as well as trace levels of various other immunologically active molecules. Clinical and laboratory studies have documented various mechanisms of action of IVIG. The complex network of immunological reactions resulting from the infusion of IVIG includes changes in several cytokines, interactions with dendritic cells, T- and B- lymphocyte effects, macrophage effects, mediated by distinct Fc-gamma receptors. In addition, effects on complement components and apoptosis have also been observed. Synergism between the different elements of the immune response characterizes the beneficial effects of IVIG in inflammatory and autoimmune disorders. They have immunopathogeneses and clinical manifestations which are difficult to define and therefore IVIG treatment indications remain heterogeneous. Dose finding studies are missing for most of the indications of the drug. In future research, defining the appropriate subgroups of patients should be undertaken. This may be accomplished by prospective registries collecting data on large numbers of patients with long-term follow-up. Controlled clinical and laboratory studies may follow based on new, validated patient selection criteria and focused on mechanisms of action, leading to more evidence-based indications.
Collapse
Affiliation(s)
- P Imbach
- Pediatric Hematology-Oncology, University Children's Hospital, Medical Faculty of University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
167
|
Bystryn JC, Jiao D. IVIg selectively and rapidly decreases circulating pathogenic autoantibodies in pemphigus vulgaris. Autoimmunity 2009; 39:601-7. [PMID: 17101504 DOI: 10.1080/08916930600972016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Intraveneous immunoglobulin (IVIg) is increasingly used to treat pemphigus vulgaris (PV). The mechanism by which it does so is not known. The following study was conducted to confirm the effectiveness of IVIg for the acute control of active PV and to elucidate the mechanism by which it does. METHODS Twelve patients with active and severe PV unresponsive to conventional therapy with high doses of systemic steroids together with or without a cytotoxic drug were treated with a single dose of IVIg (400 mg/kg/day for 5 days). All patients were concurrently given cyclophosphamide or azathioprine of not already on one of these two drugs. The primary end-points were healing of skin lesions, changes in serum levels of intercelular (IC) autoantibodies and in steroid doses one to 3 weeks after initiation of IVIg. RESULTS Within 1 week of initiating IVIg the activity of PV was controlled in most cases. Within 3 weeks the average baseline dose of systemic steroid was reduced by 40%. Serum levels of IC antibodies rapidly declined by an average of 59% within 1 week of initiating IVIg and by 70% within 2 weeks. The decrease was selective, as the average serum levels of antibody to varicella-herpes zoster did not decrease in the 4 patients in whom they were measured. The decrease in IC antibodies was inversely related to serum levels of total inmmunoglobulin (IgG). The decrease in IC antibodies was not due to blocking factors in the IVIg preparation and was too rapid to be due to suppression of IgG synthesis, suggesting that it resulted from increased catabolism. CONCLUSIONS IVIg can rapidly control active PV unresponsive to conventional therapy by causing a selective and very rapid decline in the autoantibodies that mediate the disease. We believe it does so by increasing the catabolism of all serum IgG antibodies, and that this results in a selective decrease in only abnormal autoantibodies as catabolized normal anti bodies are replaced by those present in the IVIg preparation. IVIg is the first treatment that achieves the ideal therapeutic goal in auto-antibody diseases, the selective removal of the pathogenic antibodies without affecting the level of normal antibodies.
Collapse
Affiliation(s)
- Jean-Claude Bystryn
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA.
| | | |
Collapse
|
168
|
Low SC, Mezo AR. Inhibitors of the FcRn:IgG protein-protein interaction. AAPS JOURNAL 2009; 11:432-4. [PMID: 19499344 DOI: 10.1208/s12248-009-9120-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 05/20/2009] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor, FcRn, is responsible for controlling the half-life of IgG antibodies. As a result, inhibitors of FcRn have been investigated as a possible way to modulate IgG half-lives. Such inhibitors could have possible applications in reducing autoantibody levels in autoimmune disease states. To date, monoclonal antibodies, engineered Fc domains, and short peptides have been reported to inhibit FcRn function and modulate IgG half-lives in vivo.
Collapse
Affiliation(s)
- Susan C Low
- Syntonix Pharmaceuticals Inc, A subsidiary of Biogen Idec, Waltham, MA 02451, USA
| | | |
Collapse
|
169
|
Abstract
The basic understanding of inflammatory dermatoses and autoimmune-mediated skin disorders has greatly advanced and broadened our understanding of underlying immune mechanisms that shape the complex network of chronic inflammation and autoimmunity. The new treatment options for psoriasis exemplify how new insights into (auto)immune responses, especially the role and function of various immune cells and proinflammatory cytokines, may lead to new therapeutic strategies. The concept of targeting B cells in autoimmune-mediated disorders is closely related to the discovery of autoantibodies and their cellular origin. However, the appreciation of B cells in autoimmunity has significantly changed and is not limited to their role as progenitors of autoantibody secreting plasma cells. Recent investigations of various inflammatory skin diseases, that is, autoimmune blistering disorders, collagen vascular diseases, and atopic dermatitis, actually support the concept that B cells might be as important as T cells in the etiopathogenesis of these disorders. The striking clinical improvement seen in patients with rheumatoid arthritis following B-cell depletion with the anti-CD20 mAb rituximab has tremendously catalyzed the interest in B-cell-targeted therapies in different autoimmune diseases. Future translational and clinical investigations are mandatory to precisely define the role and the contribution of impaired B-cell function in (auto)immune-mediated skin diseases.
Collapse
|
170
|
Li N, Zhao M, Wang J, Liu Z, Diaz LA. Involvement of the apoptotic mechanism in pemphigus foliaceus autoimmune injury of the skin. THE JOURNAL OF IMMUNOLOGY 2009; 182:711-7. [PMID: 19109205 DOI: 10.4049/jimmunol.182.1.711] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pemphigus foliaceus (PF) is an organ-specific autoimmune skin disease characterized by subcorneal epidermal cell detachment (acantholysis) and pathogenic autoantibodies against desmoglein 1. The mechanism responsible for pemphigus autoantibody-induced epidermal injury is not fully understood. In this study, we used the IgG passive transfer mouse model of PF to investigate the relevance of the apoptotic mechanism in pemphigus pathogenesis. TUNEL-positive epidermal cells and increased oligonucleosomes in the epidermal cytosolic fractions were detected in the diseased mice. Time course study reveals that TUNEL-positive epidermal cells appear before intraepidermal blisters. Moreover, the proapoptotic factor Bax was up-regulated at the earlier time points (2 and 4 h), whereas the antiapoptotic factor Bcl-x(L) was down-regulated at the later time points (6, 8, and 20 h) post-PF IgG injection by Western blot analysis. The active forms of caspase-3 and -6 were detected at the later time period (6, 8, and 20 h). Administration of Ac-DEVD-cmk, a peptide-based caspase-3/7 inhibitor, protected mice from developing intraepidermal blisters and clinical disease induced by PF IgG. The same protective effect was also observed using a broad-spectrum caspase inhibitor, Bok-D-fmk. Collectively, these findings show that biochemical events of apoptosis are provoked in the epidermis of mice injected with PF autoantibodies. Caspase activation may contribute to acantholytic blister formation in PF.
Collapse
Affiliation(s)
- Ning Li
- Department of Dermatology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | |
Collapse
|
171
|
Andersen JT, Sandlie I. The Versatile MHC Class I-related FcRn Protects IgG and Albumin from Degradation: Implications for Development of New Diagnostics and Therapeutics. Drug Metab Pharmacokinet 2009; 24:318-32. [DOI: 10.2133/dmpk.24.318] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
172
|
Nakamura A, Kubo T, Takai T. Fc receptor targeting in the treatment of allergy, autoimmune diseases and cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:220-33. [PMID: 19065795 DOI: 10.1007/978-0-387-09789-3_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fc receptors (FcRs) play an important role in the maintenance of an adequate activation threshold of various cells in antibody-mediated immune responses. Analyses of murine models show that the inhibitory FcR, FcyRIIB plays a pivotal role in the suppression of antibody-mediated allergy and autoimmunity. On the other hand, the activating-type FcRs are essential for the development of these diseases, suggesting that regulation of inhibitory or activating FcR is an ideal target for a therapeutic agent. Recent experimental or clinical studies also indicate that FcRs function as key receptors in the treatment with monoclonal antibodies (mAbs) therapy. This review summarizes FcR functions and highlights possible FcR-targeting therapies including mAb therapies for allergy, autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Akira Nakamura
- Department of Experimental Immunology and CREST program of Japan Science and Technology Agency, Institute of Development, Aging and Cancer, Tohoku University, Seiryo 4-1, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
173
|
Culton DA, Qian Y, Li N, Rubenstein D, Aoki V, Filhio GH, Rivitti EA, Diaz LA. Advances in pemphigus and its endemic pemphigus foliaceus (Fogo Selvagem) phenotype: a paradigm of human autoimmunity. J Autoimmun 2008; 31:311-24. [PMID: 18838249 PMCID: PMC2704386 DOI: 10.1016/j.jaut.2008.08.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 08/08/2008] [Accepted: 08/10/2008] [Indexed: 11/22/2022]
Abstract
Pemphigus encompasses a group of organ specific, antibody mediated autoimmune diseases of the skin characterized by keratinocyte detachment that leads to the development of blisters and erosions, which can become life-threatening. The pathogenic autoantibodies recognize desmogleins, which are members of the desmosomal cadherin family of cell adhesion molecules. Desmoglein 3 is targeted in pemphigus vulgaris while desmoglein 1 is targeted in pemphigus foliaceus and its endemic form, Fogo Selvagem. This review will briefly define the salient features of pemphigus and the proposed steps in pathogenesis. We will then summarize the most recent advances in three important areas of investigation: (i) epidemiologic, genetic, and immunologic features of Fogo Selvagem, (ii) molecular mechanisms of injury to the epidermis, and (iii) novel therapeutic strategies targeting specific steps in disease pathogenesis. The advances in each of these three seemingly separate areas contribute to the overall understanding of the pemphigus disease model. These recent advancements also underscore the dynamic interplay between the treatment of patients in a clinical setting and basic science research and have led to an integrative understanding of disease pathogenesis and treatment, allowing pemphigus to serve as a paradigm of human autoimmunity.
Collapse
Affiliation(s)
- Donna A. Culton
- Department of Dermatology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Ye Qian
- Department of Dermatology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Ning Li
- Department of Dermatology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - David Rubenstein
- Department of Dermatology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Valeria Aoki
- Departamento de Dermatologia, Universidade de Sao Paulo, Brazil
| | - Gunter Hans Filhio
- Departamento de Dermatologia, Universidade Federal de Mato Grosso do Sul, Brazil
| | | | - Luis A. Diaz
- Department of Dermatology, University of North Carolina at Chapel Hill, NC 27599, USA
| |
Collapse
|
174
|
Tayer-Shifman OE, Ilan Y. Immune modulation by antibodies and antibody receptors. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543770802547196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
175
|
Neonatal Fc receptor deficiency protects from tissue injury in experimental epidermolysis bullosa acquisita. J Mol Med (Berl) 2008; 86:951-9. [PMID: 18542899 DOI: 10.1007/s00109-008-0366-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 04/11/2008] [Accepted: 04/25/2008] [Indexed: 01/18/2023]
|
176
|
|
177
|
Mezo AR, McDonnell KA, Castro A, Fraley C. Structure–activity relationships of a peptide inhibitor of the human FcRn:human IgG interaction. Bioorg Med Chem 2008; 16:6394-405. [DOI: 10.1016/j.bmc.2008.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 05/01/2008] [Accepted: 05/02/2008] [Indexed: 10/22/2022]
|
178
|
Mao X, Payne AS. Seeking approval: present and future therapies for pemphigus vulgaris. CURRENT OPINION IN INVESTIGATIONAL DRUGS (LONDON, ENGLAND : 2000) 2008; 9:497-504. [PMID: 18465660 PMCID: PMC3933995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Pemphigus vulgaris is an autoimmune blistering disease of the skin and mucous membranes. Despite the potentially fatal prognosis, there are currently no FDA-approved treatments specifically for pemphigus. In 2006, the FDA designated orphan drug status to mycophenolate mofetil for the treatment of pemphigus vulgaris, indicating both federal and commercial interest in developing therapies for this devastating disease. This review focuses on pemphigus therapies that are currently in preclinical or clinical trials, as well as potential novel therapies based on recent advances in the understanding of the pathophysiology of this disease.
Collapse
|
179
|
Czernik A, Beutner EH, Bystryn JC. Intravenous immunoglobulin selectively decreases circulating autoantibodies in pemphigus. J Am Acad Dermatol 2008; 58:796-801. [DOI: 10.1016/j.jaad.2008.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 12/27/2007] [Accepted: 01/10/2008] [Indexed: 10/22/2022]
|
180
|
The Mechanisms of Action of Intravenous Immunoglobulin and Polyclonal Anti-D Immunoglobulin in the Amelioration of Immune Thrombocytopenic Purpura: What Do We Really Know? Transfus Med Rev 2008; 22:103-16. [DOI: 10.1016/j.tmrv.2007.12.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
181
|
Abstract
The remarkable success story of the therapeutic application of pooled immunoglobulin G (IgG) preparations from thousands of donors, the so-called intravenous IgG (IVIG) therapy, to patients with a variety of hematological and immunological disorders began more than half a century ago. Since then, the use of this primary blood product has increased constantly, resulting in the serious danger of shortages in supply. Despite its widespread use and therapeutic success, the mechanisms of action, especially of the anti-inflammatory activity, are only beginning to be understood. In this review, we summarize the clinical use of IVIG for different diseases and discuss recent data on the molecular mechanisms that might explain how this potent drug mediates its activity in vivo.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Experimental Immunology and Immunotherapy, Nikolaus-Fiebiger-Center for Molecular Medicine, University of Erlangen-Nuremberg, 91054 Erlangen, Germany.
| | | |
Collapse
|
182
|
Lanza A, Cirillo N, Rossiello R, Rienzo M, Cutillo L, Casamassimi A, de Nigris F, Schiano C, Rossiello L, Femiano F, Gombos F, Napoli C. Evidence of key role of Cdk2 overexpression in pemphigus vulgaris. J Biol Chem 2008; 283:8736-45. [PMID: 18199752 DOI: 10.1074/jbc.m702186200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The pathogenesis of pemphigus vulgaris (PV) is still poorly understood. Autoantibodies present in PV patients can promote detrimental effects by triggering altered transduction of signals, which results in a final acantholysis. To investigate mechanisms involved in PV, cultured keratinocytes were treated with PV serum. PV sera were able to promote the cell cycle progression, inducing the accumulation of cyclin-dependent kinase 2 (Cdk2). Microarray analysis on keratinocytes detected that PV serum induced important changes in genes coding for one and the same proteins with known biological functions involved in PV disease (560 differentially expressed genes were identified). Then, we used two different approaches to investigate the role of Cdk2. First, small interfering RNA depletion of Cdk2 prevented cell-cell detachment induced by PV sera. Second, pharmacological inhibition of Cdk2 activity through roscovitine prevented blister formation and acantholysis in the mouse model of the disease. In vivo PV serum was found to alter multiple different pathways by microarray analysis (1463 differentially expressed genes were identified). Major changes in gene expression induced by roscovitine were studied through comparison of effects of PV serum alone and in association with roscovitine. The most significantly enriched pathways were cell communication, gap junction, focal adhesion, adherens junction, and tight junction. Our data indicate that major Cdk2-dependent multiple gene regulatory events are present in PV. This alteration may influence the evolution of PV and its therapy.
Collapse
Affiliation(s)
- Alessandro Lanza
- Regional Center on Craniofacial Malformations, Clinical Odontostomatology, and Human Pathology, 1st School of Medicine and Surgery, II University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Reduction of IgG in nonhuman primates by a peptide antagonist of the neonatal Fc receptor FcRn. Proc Natl Acad Sci U S A 2008; 105:2337-42. [PMID: 18272495 DOI: 10.1073/pnas.0708960105] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The neonatal Fc receptor FcRn provides IgG molecules with their characteristically long half-lives in vivo by protecting them from intracellular catabolism and then returning them to the extracellular space. Other investigators have demonstrated that mice lacking FcRn are protected from induction of various autoimmune diseases, presumably because of the accelerated catabolism of pathogenic IgGs in the animals. Therefore, targeting FcRn with a specific inhibitor may represent a unique approach for the treatment of autoimmune disease or other diseases where the reduction of pathogenic IgG will have a therapeutic benefit. Using phage display peptide libraries, we screened for ligands that bound to human FcRn (hFcRn) and discovered a consensus peptide sequence that binds to hFcRn and inhibits the binding of human IgG (hIgG) in vitro. Chemical optimization of the phage-identified sequences yielded the 26-amino acid peptide dimer SYN1436, which is capable of potent in vitro inhibition of the hIgG-hFcRn interaction. Administration of SYN1436 to mice transgenic for hFcRn induced an increase in the rate of catabolism of hIgG in a dose-dependent manner. Treatment of cynomolgus monkeys with SYN1436 led to a reduction of IgG by up to 80% without reducing serum albumin levels that also binds to FcRn. SYN1436 and related peptides thus represent a previously uncharacterized family of potential therapeutic agents for the treatment of humorally mediated autoimmune and other diseases.
Collapse
|
184
|
Ballow M. Immunoglobulin therapy: replacement and immunomodulation. Clin Immunol 2008. [DOI: 10.1016/b978-0-323-04404-2.10085-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
185
|
Shimanovich I, Nitschke M, Rose C, Grabbe J, Zillikens D. Treatment of severe pemphigus with protein A immunoadsorption, rituximab and intravenous immunoglobulins. Br J Dermatol 2007; 158:382-8. [DOI: 10.1111/j.1365-2133.2007.08358.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
186
|
Clynes R. Protective mechanisms of IVIG. Curr Opin Immunol 2007; 19:646-51. [DOI: 10.1016/j.coi.2007.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 09/02/2007] [Indexed: 11/17/2022]
|
187
|
Liu X, Ye L, Christianson GJ, Yang JQ, Roopenian DC, Zhu X. NF-kappaB signaling regulates functional expression of the MHC class I-related neonatal Fc receptor for IgG via intronic binding sequences. THE JOURNAL OF IMMUNOLOGY 2007; 179:2999-3011. [PMID: 17709515 PMCID: PMC2667116 DOI: 10.4049/jimmunol.179.5.2999] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The neonatal Fc receptor for IgG (FcRn) functions to transport maternal IgG to a fetus or newborn and to protect IgG from degradation. Although FcRn is expressed in a variety of tissues and cell types, the extent to which FcRn expression is regulated by immunological and inflammatory events remains unknown. Stimulation of intestinal epithelial cell lines, macrophage-like THP-1, and freshly isolated human monocytes with the cytokine TNF-alpha rapidly up-regulated FcRn gene expression. In addition, the TLR ligands LPS and CpG oligodeoxynucleotide enhanced the level of FcRn expression in THP-1 and monocytes. Treatment of TNF-stimulated THP-1 cells with the NF-kappaB-specific inhibitor or overexpression of a dominant negative mutant inhibitory NF-kappaB (IkappaBalpha; S32A/S36A) resulted in down-regulation of FcRn expression. By using chromatin immunoprecipitation we identified three NF-kappaB binding sequences within introns 2 and 4 of the human FcRn gene. An EMSA confirmed the p50/p50 and/or p65/p50 complex (s) bound to intron 2- or 4-derived oligonucleotides containing putative NF-kappaB binding sequences, respectively. The intronic NF-kappaB sequences in combination with the promoter or alone regulated the expression of a luciferase reporter gene in response to TNF-alpha stimulation or overexpression of NF-kappaB p65 and p50. DNA looping interactions potentially occurred after the stimulation between intronic NF-kappaB sequences and the FcRn promoter as shown by a chromosome conformation capture assay. Finally, TNF-alpha stimulations enhanced IgG transport across an intestinal Caco-2 epithelial monolayer. Together, these data provide the first evidence that NF-kappaB signaling via intronic sequences regulates FcRn expression and function.
Collapse
Affiliation(s)
- Xindong Liu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Lilin Ye
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | | | - Jun-Qi Yang
- Department of Genome Science, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | | | - Xiaoping Zhu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
- Address correspondence and reprint requests to Dr. Xiaoping Zhu, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, MD 20742. E-mail address:
| |
Collapse
|
188
|
Abstract
The neonatal Fc receptor for IgG (FcRn) has been well characterized in the transfer of passive humoral immunity from a mother to her fetus. In addition, throughout life, FcRn protects IgG from degradation, thereby explaining the long half-life of this class of antibody in the serum. In recent years, it has become clear that FcRn is expressed in various sites in adults, where its potential function is now beginning to emerge. In addition, recent studies have examined the interaction between FcRn and the Fc portion of IgG with the aim of either improving the serum half-life of therapeutic monoclonal antibodies or reducing the half-life of pathogenic antibodies. This Review summarizes these two areas of FcRn biology.
Collapse
Affiliation(s)
- Derry C Roopenian
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | |
Collapse
|
189
|
Desensitization protocols for crossing human leukocyte antigen and ABO incompatible barriers. Curr Opin Organ Transplant 2007. [DOI: 10.1097/mot.0b013e3282703903] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
190
|
Zachary AA, Montgomery RA, Jordan SC, Reinsmoen NL, Claas FHJ, Reed EF. 14th International HLA and Immunogenetics Workshop: Report on understanding antibodies in transplantation. ACTA ACUST UNITED AC 2007; 69 Suppl 1:160-73. [PMID: 17445194 DOI: 10.1111/j.1399-0039.2006.00764.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A session of the 14 International Histocompatibility Workshop brought together experts representing the major clinical protocols, clinical research, and basic research dealing with overcoming the barrier of alloantibody in transplantation and in understanding the mechanisms by which those antibodies exert their effect on a transplanted organ. This report is an integration of the presentations of those scientists.
Collapse
Affiliation(s)
- A A Zachary
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2222, USA.
| | | | | | | | | | | |
Collapse
|
191
|
Segura S, Iranzo P, Martínez-de Pablo I, Mascaró JM, Alsina M, Herrero J, Herrero C. High-dose intravenous immunoglobulins for the treatment of autoimmune mucocutaneous blistering diseases: evaluation of its use in 19 cases. J Am Acad Dermatol 2007; 56:960-7. [PMID: 17368865 DOI: 10.1016/j.jaad.2006.06.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2006] [Revised: 05/05/2006] [Accepted: 06/14/2006] [Indexed: 11/28/2022]
Abstract
BACKGROUND The mainstay of therapy of autoimmune mucocutaneous blistering diseases has been prolonged high-dose systemic corticosteroids and immunosuppressive agents. Recently, high-dose intravenous immunoglobulin (IVIg) has been employed in selected cases, with excellent results in most of them. OBJECTIVE We sought to evaluate the outcome of the use of IVIg in patients with autoimmune mucocutaneous blistering diseases refractory to conventional therapy or with contraindications for it. METHODS We performed a retrospective analysis of clinical response to monthly cycles of IVIg in 19 patients affected with autoimmune mucocutaneous blistering diseases: 10 patients with pemphigus vulgaris (PV), 2 with pemphigus foliaceus (PF), 4 with mucous membrane pemphigoid (MMP), 2 with epidermolysis bullosa acquisita, and one with linear IgA bullous dermatosis. RESULTS Four (21%) of 19 cases presented a complete response (2 PV, 1 MMP and 1 epidermolysis bullosa acquisita). Five (26%) patients did not respond to the treatment (3 PV, 1 PF, 1 MMP). Ten patients (53%) had a partial response. LIMITATIONS This was a retrospective noncontrolled study with a heterogeneous group of patients. CONCLUSION The effectiveness of IVIg was inferior to that previously reported. This difference could be attributed to the preparations employed, the different severity of the disease, or individual responses in each patient dependent on Fc receptor gamma polymorphisms.
Collapse
Affiliation(s)
- Sonia Segura
- Department of Dermatology, Hospital Clinic, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
192
|
Negi VS, Elluru S, Sibéril S, Graff-Dubois S, Mouthon L, Kazatchkine MD, Lacroix-Desmazes S, Bayry J, Kaveri SV. Intravenous immunoglobulin: an update on the clinical use and mechanisms of action. J Clin Immunol 2007; 27:233-45. [PMID: 17351760 DOI: 10.1007/s10875-007-9088-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 02/21/2007] [Indexed: 01/27/2023]
Abstract
Initially used as a replacement therapy for immunodeficiency diseases, intravenous immunoglobulin (IVIg) is now widely used for a number of autoimmune and inflammatory diseases. Considerable progress has been made in understanding the mechanisms by which IVIg exerts immunomodulatory effects in autoimmune and inflammatory disorders. The mechanisms of action of IVIg are complex, involving modulation of expression and function of Fc receptors, interference with activation of complement and the cytokine network and of idiotype network, regulation of cell growth, and effects on the activation, differentiation, and effector functions of dendritic cells, and T and B cells.
Collapse
Affiliation(s)
- Vir-Singh Negi
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Nimmerjahn F, Ravetch JV. The antiinflammatory activity of IgG: the intravenous IgG paradox. ACTA ACUST UNITED AC 2007; 204:11-5. [PMID: 17227911 PMCID: PMC2118416 DOI: 10.1084/jem.20061788] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
How high doses of intravenous IgG (IVIG) suppress autoimmune diseases remains unresolved. We have recently shown that the antiinflammatory activity of IVIG can be attributed to a minor species of IgGs that is modified with terminal sialic acids on their Fc-linked glycans. Here we propose that these Fc-sialylated IgGs engage a unique receptor on macrophages that, in turn, leads to the upregulation of an inhibitory Fcgamma receptor (FcgammaR), thereby protecting against autoantibody-mediated pathology.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
194
|
Mignogna MD, Fortuna G, Ruoppo E, Adamo D, Leuci S, Fedele S. Variations in serum hemoglobin, albumin, and electrolytes in patients receiving intravenous immunoglobulin therapy: a real clinical threat? Am J Clin Dermatol 2007; 8:291-9. [PMID: 17902731 DOI: 10.2165/00128071-200708050-00004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Intravenous immunoglobulin (IVIg) is a solution of globulins containing antibodies derived from pooled human plasma of donors and used in the treatment of a number of immune deficiencies and autoimmune diseases. However, several investigators have reported biochemical alterations with use of IVIg. The objective of this study was to evaluate the effects of IVIg therapy on selected biochemical and hematologic parameters in patients with autoimmune mucocutaneous blistering diseases (AMBDs). METHODS In this preliminary clinical study, ten patients with AMBDs (seven with pemphigus vulgaris and three with mucous membrane pemphigoid) received 133 cycles of IVIg for a total of 399 infusions. We evaluated the effects of IVIg therapy on serum hemoglobin (Hb), albumin, and electrolyte levels, including sodium (Na+), potassium (K+), chloride (Cl-) and calcium (Ca2+). Values of these parameters were measured 24 hours before, during, and 24 hours and 4 weeks after the 3-day infusion period. RESULTS The observed variations in serum electrolyte levels were physiologically and clinically negligible. Furthermore, 24 hours after the last infusion, mean electrolyte values had spontaneously returned to normal levels without the need for additional supplementation: Na+ 137.59+/-1.42 mmol/L (p=0.6091 vs baseline); K+ 3.97+/-0.5 mmol/L (p=0.2689); Cl- 103.4+/-2.69 mmol/L (p=0.0388); and Ca2+ 9.07+/-0.44 mg/dL (p=0.5332). Conversely, significant variations in mean Hb and albumin levels were observed. When measured 24 hours after the last infusion, mild/moderate decreases in Hb (11.62+/-2.12 g/dL; p=0.009 vs baseline) and/or albumin (mean 3.14+/-0.24 g/dL; p=0.0016 vs baseline) were evident. Such changes may, albeit very rarely, be of sufficient clinical significance in individual patients as to necessitate additional treatment. CONCLUSION In patients receiving intravenous IVIg for AMBDs, electrolyte values should be monitored but do not represent a real clinical threat. Hemoglobin and albumin values may be altered sufficiently to require additional treatment but this is a very rare occurrence. These findings confirm and extend previous reports of the safety of IVIg therapy.
Collapse
Affiliation(s)
- Michele D Mignogna
- Section of Oral Medicine, Department of Odontostomatological and Maxillofacial Sciences, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
195
|
Zhao M, Trimbeger ME, Li N, Diaz LA, Shapiro SD, Liu Z. Role of FcRs in animal model of autoimmune bullous pemphigoid. THE JOURNAL OF IMMUNOLOGY 2006; 177:3398-405. [PMID: 16920981 DOI: 10.4049/jimmunol.177.5.3398] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bullous pemphigoid (BP) is a bullous dermatosis associated with autoantibodies directed against the hemidesmosomal Ags BP180 and BP230. Lesional skin is characterized by detachment of the epidermis from the dermis with an intense inflammatory cell infiltrate in the upper dermis. In experimental BP, subepidermal blistering is triggered by rabbit anti-murine BP180 (mBP180) IgG and depends upon complement activation, mast cell degranulation, and neutrophil infiltration. In this study, we determined the role of Fc gammaRs on neutrophils in experimental BP. Mice deficient in Fc gammaRIII (Fc gammaRIII-/-) and those deficient in both Fc gammaRI and Fc gammaRIII (Fc gammaRI&III-/-) but not in Fc gammaRII (Fc gammaRII-/-) were resistant to BP. Pathogenic IgG activated wild-type neutrophils, but not Fc gammaRIII-deficient neutrophils, to secrete proteolytic enzymes. The function of anti-mBP180 IgG depended entirely on its Fc domain; F(ab')2 of IgG had no pathogenic activities. In wild-type mice injected with pathogenic IgG, an Fc gammaR blocker abolished the BP phenotype and inhibited activation of wild-type neutrophils stimulated by pathogenic IgG. Results from this study establish that Fc gammaRIII plays a critical role in the activation of infiltrating neutrophils and the subsequent blistering in experimental BP.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Glycocalyx
- Immunoglobulin G/immunology
- Mast Cells/cytology
- Mast Cells/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Infiltration/immunology
- Neutrophils/immunology
- Pemphigoid, Bullous/genetics
- Pemphigoid, Bullous/immunology
- Pemphigoid, Bullous/metabolism
- Pemphigoid, Bullous/pathology
- Receptors, Fc/deficiency
- Receptors, Fc/genetics
- Receptors, Fc/immunology
- Receptors, Fc/metabolism
Collapse
Affiliation(s)
- Minglang Zhao
- Department of Dermatology, University of North Carolina School of Medicine, Chapel Hill 27599, USA
| | | | | | | | | | | |
Collapse
|
196
|
Yoshida M, Kobayashi K, Kuo TT, Bry L, Glickman JN, Claypool SM, Kaser A, Nagaishi T, Higgins DE, Mizoguchi E, Wakatsuki Y, Roopenian DC, Mizoguchi A, Lencer WI, Blumberg RS. Neonatal Fc receptor for IgG regulates mucosal immune responses to luminal bacteria. J Clin Invest 2006; 116:2142-2151. [PMID: 16841095 PMCID: PMC1501111 DOI: 10.1172/jci27821] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/16/2006] [Indexed: 01/22/2023] Open
Abstract
The neonatal Fc receptor for IgG (FcRn) plays a major role in regulating host IgG levels and transporting IgG and associated antigens across polarized epithelial barriers. Selective expression of FcRn in the epithelium is shown here to be associated with secretion of IgG into the lumen that allows for defense against an epithelium-associated pathogen (Citrobacter rodentium). This pathway of host resistance to a bacterial pathogen as mediated by FcRn involves retrieval of bacterial antigens from the lumen and initiation of adaptive immune responses in regional lymphoid structures. Epithelial-associated FcRn, through its ability to secrete and absorb IgG, may thus integrate luminal antigen encounters with systemic immune compartments and as such provide essential host defense and immunoregulatory functions at the mucosal surfaces.
Collapse
Affiliation(s)
- Masaru Yoshida
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Timothy T. Kuo
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Lynn Bry
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Jonathan N. Glickman
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Steven M. Claypool
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Arthur Kaser
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Takashi Nagaishi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Darren E. Higgins
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Emiko Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Yoshio Wakatsuki
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Derry C. Roopenian
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Atsushi Mizoguchi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Wayne I. Lencer
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital,
Department of Pathology, Brigham and Women’s Hospital, and
Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA.
Division of Clinical Bioregulatory Science, Kyoto University, Kyoto, Japan.
The Jackson Laboratory, Bar Harbor, Maine, USA.
Gastrointestinal Cell Biology, Department of Medicine, Children’s Hospital, Boston, Massachusetts, USA.
Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
197
|
Aukrust P, Yndestad A, Ueland T, Damås JK, Frøland SS, Gullestad L. The role of intravenous immunoglobulin in the treatment of chronic heart failure. Int J Cardiol 2006; 112:40-5. [PMID: 16893578 DOI: 10.1016/j.ijcard.2006.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 05/10/2006] [Indexed: 11/16/2022]
Abstract
Chronic heart failure (HF), including both ischemic and idiopathic dilated cardiomyopathies, is accompanied by a dysregulated cytokine network characterized not only by a rise in inflammatory cytokines, but also by an inadequate elevation of anti-inflammatory mediators. This dysregulation has been implicated in the development and progression of chronic HF, and in the last decade, attempts have been made to modulate this persistent inflammation. Failure of anti-tumor necrosis factor therapy in HF has led to further interest in a more general immunomodulatory approach, directed against the inflammatory imbalance rather than one particular cytokine. Treatment with intravenous immunoglobulin (IVIg) may represent such a broad-based approach trying to restore the dysregulated cytokine network through various mechanisms such as Fc receptor blockade, neutralization of microbial antigens and superantigens and more direct anti-inflammatory effects on the cytokine network. However, although one randomized placebo-controlled study in patients with chronic HF showed that IVIg improved left ventricular ejection fraction, accompanied by anti-inflammatory net effects, IVIg had no effect in another placebo-controlled study examining the effect of this medication in recent-onset cardiomyopathy. So far, few patients have been included in clinical trials, and there is clearly a need for larger placebo-controlled mortality studies involving a diverse group of patients with regard to cause and severity of HF.
Collapse
Affiliation(s)
- Pål Aukrust
- Research Institute for Internal Medicine, Rikshospitalet University Hospital, University of Oslo, N-0027 Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
198
|
Transplantation of the highly human leukocyte antigen–sensitized patient: long-term outcomes and future directions. Transplant Rev (Orlando) 2006. [DOI: 10.1016/j.trre.2006.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|