1
|
Hogrefe K, Winkler JK, Enk AH. Two Patients with Therapy-Resistant Pemphigus Vulgaris and Severe Underlying Disease Showing Good Response to a New IVIg Preparation. Dermatol Ther (Heidelb) 2025; 15:237-244. [PMID: 39751746 PMCID: PMC11785848 DOI: 10.1007/s13555-024-01326-6] [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: 11/12/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Pemphigus vulgaris is a severe and often therapy-resistant bullous autoimmune disease. Standard therapy with steroids often administered together with another immunosuppressant does not respond in all patients or may not be a good therapeutic option in patients with severe underlying diseases. Intravenous immunoglobulins (IVIgs) represent a treatment alternative, often showing a rapid response which allows one to reduce concomitant immunosuppression. Here, we report on two patients with a complex disease history suffering from severe pemphigus vulgaris who received treatment with a new IVIg preparation (Yimmugo®, 2 g per kg body weight every 4 weeks). IVIg preparations differ regarding manufacturing process and show a varying side effect profile. Both of our patients did not experience any side effects from IVIgs and showed a significant improvement of skin and mucosal erosions. More reports on IVIgs are desirable to help in selecting the optimal preparation and dosing regarding tolerability and effectiveness for individual patients.
Collapse
Affiliation(s)
- Katharina Hogrefe
- Department of Dermatology, University Hospital Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Julia K Winkler
- Department of Dermatology, University Hospital Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Alexander H Enk
- Department of Dermatology, University Hospital Heidelberg, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Schmidt C, Weißmüller S, Heinz CC. Multifaceted Tissue-Protective Functions of Polyvalent Immunoglobulin Preparations in Severe Infections-Interactions with Neutrophils, Complement, and Coagulation Pathways. Biomedicines 2023; 11:3022. [PMID: 38002022 PMCID: PMC10669904 DOI: 10.3390/biomedicines11113022] [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: 09/26/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Severe infections induce immune defense mechanisms and initial tissue damage, which produce an inflammatory neutrophil response. Upon dysregulation of these responses, inflammation, further tissue damage, and systemic spread of the pathogen may occur. Subsequent vascular inflammation and activation of coagulation processes may cause microvascular obstruction at sites distal to the primary site of infection. Low immunoglobulin (Ig) M and IgG levels have been detected in patients with severe infections like sCAP and sepsis, associated with increased severity and mortality. Based on Ig's modes of action, supplementation with polyvalent intravenous Ig preparations (standard IVIg or IgM/IgA-enriched Ig preparations) has long been discussed as a treatment option for severe infections. A prerequisite seems to be the timely administration of Ig preparations before excessive tissue damage has occurred and coagulopathy has developed. This review focuses on nonclinical and clinical studies that evaluated tissue-protective activities resulting from interactions of Igs with neutrophils, complement, and the coagulation system. The data indicate that coagulopathy, organ failure, and even death of patients can possibly be prevented by the timely combined interactions of (natural) IgM, IgA, and IgG with neutrophils and complement.
Collapse
Affiliation(s)
- Carolin Schmidt
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| | | | - Corina C Heinz
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| |
Collapse
|
3
|
Velikova T, Sekulovski M, Bogdanova S, Vasilev G, Peshevska-Sekulovska M, Miteva D, Georgiev T. Intravenous Immunoglobulins as Immunomodulators in Autoimmune Diseases and Reproductive Medicine. Antibodies (Basel) 2023; 12:20. [PMID: 36975367 PMCID: PMC10045256 DOI: 10.3390/antib12010020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Intravenous administration of immunoglobulins has been routinely used for more than 60 years in clinical practice, developed initially as replacement therapy in immunodeficiency disorders. Today, the use of intravenous immunoglobulins (IVIGs) is embedded in the modern algorithms for the management of a few diseases, while in most cases, their application is off-label and thus different from their registered therapeutic indications according to the summary of product characteristics. In this review, we present the state-of-the-art use of IVIGs in various autoimmune conditions and immune-mediated disorders associated with reproductive failure, as approved therapy, based on indications or off-label. IVIGs are often an alternative to other treatments, and the administration of IVIGs continues to expand as data accumulate. Additionally, new insights into the pathophysiology of immune-mediated disorders have been gained. Therefore, the need for immunomodulation has increased, where IVIG therapy represents an option for stimulating, inhibiting and regulating various immune processes.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Metodija Sekulovski
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Anesthesiology and Intensive Care, University Hospital Lozenetz, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Simona Bogdanova
- First Department of Internal Medicine, Medical Faculty, Medical University of Varna, 9000 Varna, Bulgaria
| | - Georgi Vasilev
- Clinic of Neurology, UMHAT “Sv. Georgi”, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Monika Peshevska-Sekulovska
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Gastroenterology, University Hospital Lozenetz, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Dimitrina Miteva
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Tsvetoslav Georgiev
- First Department of Internal Medicine, Medical Faculty, Medical University of Varna, 9000 Varna, Bulgaria
| |
Collapse
|
4
|
Peng X, Luo Z, He S, Zhang L, Li Y. Blood-Brain Barrier Disruption by Lipopolysaccharide and Sepsis-Associated Encephalopathy. Front Cell Infect Microbiol 2021; 11:768108. [PMID: 34804998 PMCID: PMC8599158 DOI: 10.3389/fcimb.2021.768108] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
As a complex multicellular structure of the vascular system at the central nervous system (CNS), the blood-brain barrier (BBB) separates the CNS from the system circulation and regulates the influx and efflux of substances to maintain the steady-state environment of the CNS. Lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, can damage the barrier function of BBB and further promote the occurrence and development of sepsis-associated encephalopathy (SAE). Here, we conduct a literature review of the direct and indirect damage mechanisms of LPS to BBB and the relationship between these processes and SAE. We believe that after LPS destroys BBB, a large number of inflammatory factors and neurotoxins will enter and damage the brain tissue, which will activate brain immune cells to mediate inflammatory response and in turn further destroys BBB. This vicious circle will ultimately lead to the progression of SAE. Finally, we present a succinct overview of the treatment of SAE by restoring the BBB barrier function and summarize novel opportunities in controlling the progression of SAE by targeting the BBB.
Collapse
Affiliation(s)
- Xiaoyao Peng
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Zhixuan Luo
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Shuang He
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Luhua Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ying Li
- Department of Immunology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Chen M, Edwards SR, Reutens DC. Complement in the Development of Post-Traumatic Epilepsy: Prospects for Drug Repurposing. J Neurotrauma 2021; 37:692-705. [PMID: 32000582 DOI: 10.1089/neu.2019.6942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeting neuroinflammation is a novel frontier in the prevention and treatment of epilepsy. A substantial body of evidence supports a key role for neuroinflammation in epileptogenesis, the pathological process that leads to the development and progression of spontaneous recurrent epileptic seizures. It is also well recognized that traumatic brain injury (TBI) induces a vigorous neuroinflammatory response and that a significant proportion of patients with TBI suffer from debilitating post-traumatic epilepsy. The complement system is a potent effector of innate immunity and a significant contributor to secondary tissue damage and to epileptogenesis following central nervous system injury. Several therapeutic agents targeting the complement system are already on the market to treat other central nervous system disorders or are well advanced in their development. The purpose of this review is to summarize findings on complement activation in experimental TBI and epilepsy models, highlighting the potential of drug repurposing in the development of therapeutics to ameliorate post-traumatic epileptogenesis.
Collapse
Affiliation(s)
- Min Chen
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen R Edwards
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - David C Reutens
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
6
|
Schmidt C, Weißmüller S, Bohländer F, Germer M, König M, Staus A, Wartenberg-Demand A, Heinz CC, Schüttrumpf J. The Dual Role of a Polyvalent IgM/IgA-Enriched Immunoglobulin Preparation in Activating and Inhibiting the Complement System. Biomedicines 2021; 9:817. [PMID: 34356880 PMCID: PMC8301464 DOI: 10.3390/biomedicines9070817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Activation of the complement system is important for efficient clearance of a wide variety of pathogens via opsonophagocytosis, or by direct lysis via complement-dependent cytotoxicity (CDC). However, in severe infections dysregulation of the complement system contributes to hyperinflammation. The influence of the novel IgM/IgA-enriched immunoglobulin preparation trimodulin on the complement pathway was investigated in in vitro opsonophagocytosis, binding and CDC assays. Immunoglobulin levels before and after trimodulin treatment were placed in relation to complement assessments in humans. In vitro, trimodulin activates complement and induces opsonophagocytosis, but also interacts with opsonins C3b, C4b and anaphylatoxin C5a in a concentration-dependent manner. This was not observed for standard intravenous IgG preparation (IVIg). Accordingly, trimodulin, but not IVIg, inhibited the downstream CDC pathway and target cell lysis. If applied at a similar concentration range in healthy subjects, trimodulin treatment resulted in C3 and C4 consumption in a concentration-dependent manner, which was extended in patients with severe community-acquired pneumonia. Complement consumption is found to be dependent on underlying immunoglobulin levels, particularly IgM, pinpointing their regulative function in humans. IgM/IgA provide a balancing effect on the complement system. Trimodulin may enhance phagocytosis and opsonophagocytosis in patients with severe infections and prevent excessive pathogen lysis and release of harmful anaphylatoxins.
Collapse
Affiliation(s)
- Carolin Schmidt
- Department of Translational Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (C.S.); (M.K.)
| | - Sabrina Weißmüller
- Department of Translational Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (C.S.); (M.K.)
| | - Fabian Bohländer
- Department of Analytical Development and Validation, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Matthias Germer
- Preclinical Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Martin König
- Department of Translational Research, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany; (C.S.); (M.K.)
| | - Alexander Staus
- Corporate Biostatistics, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Andrea Wartenberg-Demand
- Corporate Clinical Research & Development, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Corina C. Heinz
- Clinical Strategy & Development, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| | - Jörg Schüttrumpf
- Corporate R&D, Biotest AG, Landsteinerstraße 5, 63303 Dreieich, Germany;
| |
Collapse
|
7
|
Farini A, Villa C, Tripodi L, Legato M, Torrente Y. Role of Immunoglobulins in Muscular Dystrophies and Inflammatory Myopathies. Front Immunol 2021; 12:666879. [PMID: 34335568 PMCID: PMC8316973 DOI: 10.3389/fimmu.2021.666879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/25/2021] [Indexed: 01/15/2023] Open
Abstract
Muscular dystrophies and inflammatory myopathies are heterogeneous muscular disorders characterized by progressive muscle weakness and mass loss. Despite the high variability of etiology, inflammation and involvement of both innate and adaptive immune response are shared features. The best understood immune mechanisms involved in these pathologies include complement cascade activation, auto-antibodies directed against muscular proteins or de-novo expressed antigens in myofibers, MHC-I overexpression in myofibers, and lymphocytes-mediated cytotoxicity. Intravenous immunoglobulins (IVIGs) administration could represent a suitable immunomodulator with this respect. Here we focus on mechanisms of action of immunoglobulins in muscular dystrophies and inflammatory myopathies highlighting results of IVIGs from pre-clinical and case reports evidences.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Dino Ferrari Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Dino Ferrari Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
8
|
Corticosteroid actions on dengue immune pathology; A review article. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2020. [DOI: 10.1016/j.cegh.2019.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
9
|
A C5a-Immunoglobulin complex in chronic lymphocytic leukemia patients is associated with decreased complement activity. PLoS One 2019; 14:e0209024. [PMID: 30601845 PMCID: PMC6314568 DOI: 10.1371/journal.pone.0209024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/27/2018] [Indexed: 11/19/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the Western world. The therapeutic approach to CLL includes chemotherapeutic regimens and immunotherapy. Complement-mediated cytotoxicity, which is one of the mechanisms activated by the therapeutic monoclonal antibodies, depends on the availability and activity of the complement (C) system. The aim was to study the structure of circulating C components and evaluate the importance of C5 structural integrity for C activity in CLL patients. Blood samples were collected from 40 naïve CLL patients and 15 normal controls (NC). The Western blot analysis showed abnormal C5 pattern in some CLL patients, while patterns of C3 and C4 were similar in all subjects. Levels of the C activation markers sC5b-9 and C5a were quantified before and after activation via the classical (CP) and alternative (AP) pathways. In patients with abnormal C5, basal levels of sC5b-9 and C5a were increased while activities of the CP and of the CP C5-convertase, the immediate C5-upstream complex, were decreased compared to NC and to patients with normal C5. The data indicate a link between CP activation and apparent C5 alterations in CLL. This provides a potential prognostic tool that may personalize therapy by identifying a sub-group of CLL patients who display an abnormal C5 pattern, high basal levels of sC5b-9 and C5a, and impaired CP activity, and are likely to be less responsive to immunotherapy due to compromised CP activity.
Collapse
|
10
|
Esen F, Ozcan PE, Tuzun E, Boone MD. Mechanisms of action of intravenous immunoglobulin in septic encephalopathy. Rev Neurosci 2018; 29:417-423. [PMID: 29232196 DOI: 10.1515/revneuro-2017-0065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022]
Abstract
Acute brain dysfunction associated with sepsis is a serious complication that results in morbidity and mortality. Intravenous immunoglobulin (IVIg) treatment is known to alleviate behavioral deficits in the experimentally induced model of sepsis. To delineate the mechanisms by which IVIg treatment prevents neuronal dysfunction, an array of immunological and apoptosis markers was investigated. Our results suggest that IVIgG and IgGAM administration ameliorates neuronal dysfunction and behavioral deficits by reducing apoptotic cell death and glial cell proliferation. IgGAM treatment might suppress classical complement pathway by reducing C5a activity and proapoptotic NF-κB and Bax expressions, thereby, inhibiting major inflammation and apoptosis cascades. Future animal model experiments performed with specific C5aR and NF-κB agonists/antagonists or C5aR-deficient mice might more robustly disclose the significance of these pathways. C5a, C5aR, and NF-κB, which were shown to be the key molecules in brain injury pathogenesis in sepsis, might also be utilized as potential targets for future treatment trials of septic encephalopathy.
Collapse
Affiliation(s)
- Figen Esen
- Department of Anesthesiology, Istanbul Faculty of Medicine, Istanbul University, 34393 Istanbul, Turkey
| | - Perihan Ergin Ozcan
- Department of Anesthesiology, Istanbul Faculty of Medicine, Istanbul University, 34393 Istanbul, Turkey
| | - Erdem Tuzun
- Institute of Experimental Medicine, Neuroscience, Istanbul University, 34393 Istanbul, Turkey
| | - M Dustin Boone
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, 02215 MA, USA
| |
Collapse
|
11
|
Liu SY, Yu XL, Zhu J, Liu XM, Zhang Y, Dong QX, Ma S, Liu RT. Intravenous immunoglobulin ameliorates motor and cognitive deficits and neuropathology in R6/2 mouse model of Huntington's disease by decreasing mutant huntingtin protein level and normalizing NF-κB signaling pathway. Brain Res 2018; 1697:21-33. [PMID: 29902468 DOI: 10.1016/j.brainres.2018.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder characterized by progressive movement disorders and cognitive deficits, which is caused by a CAG-repeat expansion encoding an extended polyglutamine (polyQ) tract in the huntingtin protein (HTT). Reduction of mutant HTT levels and inhibition of neuroinflammation has been proposed as a major therapeutic strategy in treating HD. Intravenous immunoglobulin (IVIg) therapy has been firmly established for the treatment of several autoimmune or inflammatory neurological diseases, either as adjunctive treatment or as first-line therapy. However, whether IVIg has therapeutic potential on HD remains unclear. Here we for the first time demonstrated that IVIg treatment remarkably rescued motor and cognitive deficits, prevented synaptic degeneration, attenuated neuroinflammation and oxidative stress in R6/2 mouse model. Further investigation showed that the beneficial effects of IVIg resulted from the reduced levels of mutant HTT and inhibition of NF-κB signalling pathway. These findings suggest that IVIg is a promising therapeutic potential for HD.
Collapse
Affiliation(s)
- Shu-Ying Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiao-Lin Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Zhu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiang-Meng Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yue Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Quan-Xiu Dong
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Shan Ma
- Shandong Institute of Biological Products, Taian 271000, China.
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
12
|
Howard JF. Myasthenia gravis: the role of complement at the neuromuscular junction. Ann N Y Acad Sci 2017; 1412:113-128. [PMID: 29266249 DOI: 10.1111/nyas.13522] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/11/2022]
Abstract
Generalized myasthenia gravis (gMG) is a rare autoimmune disorder characterized by skeletal muscle weakness caused by disrupted neurotransmission at the neuromuscular junction (NMJ). Approximately 74-88% of patients with gMG have acetylcholine receptor (AChR) autoantibodies. Complement plays an important role in innate and antibody-mediated immunity, and activation and amplification of complement results in the formation of membrane attack complexes (MACs), lipophilic proteins that damage cell membranes. The role of complement in gMG has been demonstrated in animal models and patients. Studies in animals lacking specific complement proteins have confirmed that MAC formation is required to induce experimental autoimmune MG (EAMG) and NMJ damage. Complement inhibition in EAMG models can prevent disease induction and reverse its progression. Patients with anti-AChR+ MG have autoantibodies and MACs present at NMJs. Damaged NMJs are associated with more severe disease, fewer AChRs, and MACs in synaptic debris. Current MG therapies do not target complement directly. Eculizumab is a humanized monoclonal antibody that inhibits cleavage of complement protein C5, preventing MAC formation. Eculizumab treatment improved symptoms compared with placebo in a phase II study in patients with refractory gMG. Direct complement inhibition could preserve NMJ physiology and muscle function in patients with anti-AChR+ gMG.
Collapse
Affiliation(s)
- James F Howard
- Department of Neurology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
13
|
Corbí AL, Sánchez-Ramón S, Domínguez-Soto A. The potential of intravenous immunoglobulins for cancer therapy: a road that is worth taking? Immunotherapy 2017; 8:601-12. [PMID: 27140412 DOI: 10.2217/imt.16.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Much has been learned recently about the role of immunoglobulins as effector molecules of the adaptive immunity and as active elements in the maintenance of immune homeostasis. The increasing number of pathologies where intravenous immunoglobulins (IVIg) display a beneficial action illustrates their therapeutic relevance. Considering recent findings on the ability of IVIg to modulate macrophage polarization, herein we review evidences on the antitumoral activity of IVIg. Fragmentary and nonconclusive, available evidences are just suggestive of the potential of IVIg in antitumoral therapy, but encourage for the generation of additional evidences through well-designed clinical trials, and for additional studies to address the molecular effects of IVIg as a means to avoid the extrapolation of data gathered from animal models.
Collapse
Affiliation(s)
- Angel L Corbí
- Centro de Investigaciones Biológicas, CSIC. Ramiro de Maeztu, 9. 28040 Madrid, SPAIN
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology & IdISSC, Hospital Clínico San Carlos, Prof Martín Lagos, S/N, 28040 Madrid, Spain; and, Department of Microbiology I, Complutense University School of Medicine, Madrid, Spain
| | | |
Collapse
|
14
|
Pilch KS, Spaeth PJ, Yuki N, Wakerley BR. Therapeutic complement inhibition: a promising approach for treatment of neuroimmunological diseases. Expert Rev Neurother 2017; 17:579-591. [PMID: 28092989 DOI: 10.1080/14737175.2017.1282821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Autoimmunity is an important cause of disease both in the central and peripheral nervous systems. Aetiologies and clinical manifestations are complex and heterogeneous. Inappropriate control of complement activation at inappropriate sites has been recognized as a major determinant in several neurological conditions, including Guillain-Barré syndrome and neuromyelitis optica. In each case pathogenesis is thought to be associated with generation of autoantibodies which upon binding guide activation of the complement system to self-tissue. Areas covered: Modulation of the complement system activation at such sites may represent a novel therapeutic approach for treatment of immune-mediated inflammatory conditions. In this review we focus on the therapeutic effects of complement inhibitors in Guillain-Barré syndrome and neuromyelitis optica and highlight recent developments within the field. Expert Commentary: Conventional first line treatment strategies in GBS and NMO have the potential disadvantage of causing widespread immunosuppressive effects. A more targeted approach may therefore be more effective and less disruptive to the immune system, especially in the case of NMO, which requires long term immunosuppression. Modulation of the complement system may hold the key and has already been shown to be of clinical benefit in other non-neurological conditions, including paroxysmal nocturnal hemoglobinuria and hereditary angioedema.
Collapse
Affiliation(s)
- Kjara S Pilch
- a Department of Cell and Developmental Biology , University College London , UK
| | - Peter J Spaeth
- b University of Bern, Institute of Pharmacology , Bern , Switzerland
| | - Nobuhiro Yuki
- c Department of Neurology , University of Mishima , Niigata , Japan
| | - Benjamin R Wakerley
- d Department of Neurology , Gloucestershire Hospitals NHS Foundation Trust , Gloucester , United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
15
|
Disease-modifying effect of intravenous immunoglobulin in an experimental model of epilepsy. Sci Rep 2017; 7:40528. [PMID: 28074934 PMCID: PMC5225452 DOI: 10.1038/srep40528] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/07/2016] [Indexed: 12/24/2022] Open
Abstract
Novel therapies that prevent or modify the development of epilepsy following an initiating brain insult could significantly reduce the burden of this disease. In light of evidence that immune mechanisms play an important role in generating and maintaining the epileptic condition, we evaluated the effect of a well-established immunomodulatory treatment, intravenous immunoglobulin (IVIg), on the development of epilepsy in an experimental model of epileptogenesis. In separate experiments, IVIg was administered either before (pre-treatment) or after (post-treatment) the onset of pilocarpine status epilepticus (SE). Our results show that both pre- and post-treatment with IVIg attenuated acute inflammation in the SE model. Specifically, IVIg reduced local activation of glial cells, complement system activation, and blood-brain barrier damage (BBB), which are all thought to play important roles in the development of epilepsy. Importantly, post-treatment with IVIg was also found to reduce the frequency and duration of subsequent spontaneous recurrent seizures as detected by chronic video-electroencephalographic (video-EEG) recordings. This finding supports a novel application for IVIg, specifically its repurposing as a disease-modifying therapy in epilepsy.
Collapse
|
16
|
Abstract
The increasing insight into pathomechanisms of dysregulated host response in several inflammatory diseases led to the implementation of the term “cytokine storm” in the literature more than 20 years ago. Direct toxic effects as well as indirect immunomodulatory mechanisms during cytokine storm have been described and were the basis for the rationale to use several substances and devices in life-threatening infections and hyperinflammatory states. Clinical trials have been performed, most of them in the form of minor, investigator-initiated protocols; major clinical trials focused mostly on sepsis and septic shock. The following review tries to summarize the background, pathophysiology, and results of clinical investigations that had implications for the development of therapeutic strategies and international guidelines for the management of hyperinflammation during syndromes of cytokine storm in adult patients, predominantly in septic shock.
Collapse
Affiliation(s)
- Herwig Gerlach
- Department of Anesthesia, Critical Care Medicine, and Pain Management, Vivantes - Klinikum Neukoelln, Klinik fuer Anaesthesie, operative Intensivmedizin und Schmerztherapie, Berlin, Germany
| |
Collapse
|
17
|
Brennan FH, Kurniawan ND, Vukovic J, Bartlett PF, Käsermann F, Arumugam TV, Basta M, Ruitenberg MJ. IVIg attenuates complement and improves spinal cord injury outcomes in mice. Ann Clin Transl Neurol 2016; 3:495-511. [PMID: 27386499 PMCID: PMC4931715 DOI: 10.1002/acn3.318] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/22/2022] Open
Abstract
Objective Traumatic spinal cord injury (SCI) elicits immediate neural cell death, axonal damage, and disruption of the blood–spinal cord barrier, allowing circulating immune cells and blood proteins into the spinal parenchyma. The inflammatory response to SCI involves robust complement system activation, which contributes to secondary injury and impairs neurological recovery. This study aimed to determine whether intravenous immunoglobulin (IVIg), an FDA‐approved treatment for inflammatory conditions, can scavenge complement activation products and improve recovery from contusive SCI. Methods We used functional testing, noninvasive imaging, and detailed postmortem analysis to assess whether IVIg therapy is effective in a mouse model of severe contusive SCI. Results IVIg therapy at doses of 0.5–2 g/kg improved the functional and histopathological outcomes from SCI, conferring protection against lesion enlargement, demyelination, central canal dilation, and axonal degeneration. The benefits of IVIg were detectable through noninvasive diffusion tensor imaging (DTI), with IVIg treatment counteracting the progressive SCI‐induced increase in radial diffusivity (RD) in white matter. Diffusion indices significantly correlated with the functional performance of individual mice and accurately predicted the degree of myelin preservation. Further experiments revealed that IVIg therapy reduced the presence of complement activation products and phagocytically active macrophages at the lesion site, providing insight as to its mechanisms of action. Interpretation Our findings highlight the potential of using IVIg as an immunomodulatory treatment for SCI, and the value of DTI to assess tissue damage and screen for the efficacy of candidate intervention strategies in preclinical models of SCI, both quantitatively and noninvasively.
Collapse
Affiliation(s)
- Faith H Brennan
- School of Biomedical Sciences The University of Queensland Brisbane 4072 Australia
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging The University of Queensland Brisbane 4072 Australia
| | - Jana Vukovic
- School of Biomedical Sciences The University of Queensland Brisbane 4072 Australia; Queensland Brain Institute The University of Queensland Brisbane 4072 Australia
| | - Perry F Bartlett
- Queensland Brain Institute The University of Queensland Brisbane 4072 Australia
| | | | - Thiruma V Arumugam
- Department of Physiology Yong Loo Lin School of Medicine National University of Singapore 117597 Singapore
| | - Milan Basta
- BioVisions Inc. 9012 Wandering Trail Dr Potomac Maryland 20854 USA
| | - Marc J Ruitenberg
- School of Biomedical Sciences The University of Queensland Brisbane 4072 Australia; Queensland Brain Institute The University of Queensland Brisbane 4072 Australia; Trauma Critical Care and Recovery Brisbane Diamantina Health Partners The University of Queensland Brisbane 4072 Australia
| |
Collapse
|
18
|
Djoumerska-Alexieva I, Roumenina L, Pashov A, Dimitrov J, Hadzhieva M, Lindig S, Voynova E, Dimitrova P, Ivanovska N, Bockmeyer C, Stefanova Z, Fitting C, Bläss M, Claus R, von Gunten S, Kaveri S, Cavaillon JM, Bauer M, Vassilev T. Intravenous Immunoglobulin with Enhanced Polyspecificity Improves Survival in Experimental Sepsis and Aseptic Systemic Inflammatory Response Syndromes. Mol Med 2016; 21:1002-1010. [PMID: 26701312 DOI: 10.2119/molmed.2014.00224] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/14/2015] [Indexed: 11/06/2022] Open
Abstract
Sepsis is a major cause for death worldwide. Numerous interventional trials with agents neutralizing single proinflammatory mediators have failed to improve survival in sepsis and aseptic systemic inflammatory response syndromes. This failure could be explained by the widespread gene expression dysregulation known as "genomic storm" in these patients. A multifunctional polyspecific therapeutic agent might be needed to thwart the effects of this storm. Licensed pooled intravenous immunoglobulin preparations seemed to be a promising candidate, but they have also failed in their present form to prevent sepsis-related death. We report here the protective effect of a single dose of intravenous immunoglobulin preparations with additionally enhanced polyspecificity in three models of sepsis and aseptic systemic inflammation. The modification of the pooled immunoglobulin G molecules by exposure to ferrous ions resulted in their newly acquired ability to bind some proinflammatory molecules, complement components and endogenous "danger" signals. The improved survival in endotoxemia was associated with serum levels of proinflammatory cytokines, diminished complement consumption and normalization of the coagulation time. We suggest that intravenous immunoglobulin preparations with additionally enhanced polyspecificity have a clinical potential in sepsis and related systemic inflammatory syndromes.
Collapse
Affiliation(s)
- Iglika Djoumerska-Alexieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Lubka Roumenina
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Anastas Pashov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Jordan Dimitrov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Maya Hadzhieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Sandro Lindig
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Elisaveta Voynova
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Petya Dimitrova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Nina Ivanovska
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Zvetanka Stefanova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Catherine Fitting
- Cytokines and Inflammation Unit, Institut Pasteur, Paris, France; and
| | - Markus Bläss
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Ralf Claus
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | | | - Srini Kaveri
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | - Michael Bauer
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Tchavdar Vassilev
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
19
|
Pulvirenti F, Granata G, Girelli G, Quinti I. Immunoglobulin-induced hemolysis, splenomegaly and inflammation in patients with antibody deficiencies. Expert Rev Clin Immunol 2016; 12:725-31. [PMID: 26854522 DOI: 10.1586/1744666x.2016.1151787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
IgG replacement for primary antibody deficiencies is a safe treatment administered to prevent recurrent infections and reduce mortality. Recently, several reports described acute hemolytic episodes following IgG administration due to a passive transfer of blood group alloantibodies, including anti-A, anti-B, as well as anti-Rh antibodies. Here, we reviewed and discussed the consequences of passively transferred RBCs antibodies. The chronic passive transfer of alloantibodies might also cause a subclinical condition due to a compensated extravascular chronic hemolysis with poorly understood consequences. This phenomenon might possibly represent an unrecognized cause of splenomegaly and might contribute to inflammation in patients with primary antibody deficiencies.
Collapse
Affiliation(s)
- Federica Pulvirenti
- a Centre for Primary Immune Deficiencies, Department of Molecular Medicine , Sapienza University of Rome , Rome , Italy
| | - Guido Granata
- a Centre for Primary Immune Deficiencies, Department of Molecular Medicine , Sapienza University of Rome , Rome , Italy
| | - Gabriella Girelli
- b Unit of Immunohematology and Transfusion Medicine , Sapienza University of Rome , Rome , Italy
| | - Isabella Quinti
- a Centre for Primary Immune Deficiencies, Department of Molecular Medicine , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
20
|
Živković S. Intravenous immunoglobulin in the treatment of neurologic disorders. Acta Neurol Scand 2016; 133:84-96. [PMID: 25997034 DOI: 10.1111/ane.12444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2015] [Indexed: 12/17/2022]
Abstract
Intravenous immunoglobulins (IVIGs) are often used in the treatment of autoimmune disorders and immunodeficiencies, and it has been estimated that neurologic indications can account for up to 43% of IVIG used in clinical practice. In neurologic clinical practice, IVIG is used for acute therapy of newly diagnosed autoimmune disorders or exacerbations of pre-existing conditions, or as long-term maintenance treatment for chronic disorders. IVIG exerts its effects on humoral and cell-based immunity through multiple pathways, without a single dominant mechanism. Clinical use of IVIG has been supported by guidelines from American Academy of Neurology and European Federation of Neurologic Societies. IVIG is generally recommended for the treatment of Guillain-Barre syndrome and chronic inflammatory demyelinating polyneuropathy in adults, multifocal motor neuropathy and myasthenia gravis, and should be considered as a treatment option for dermatomyositis in adults and Lambert-Eaton myasthenic syndrome. Additional potential indications include stiff person syndrome, multiple sclerosis during pregnancy or while breastfeeding, refractory autoimmune epilepsy, and paraneoplastic disorders. Clinical use of IVIG is mostly safe but few adverse effects may still occur with potentially severe complications, including aseptic meningitis and thromboembolism. In addition to intravenous route (IVIG), subcutaneous immunoglobulins have been used as an alternative treatment option, especially in patients with limited intravenous access. Treatment with IVIG is effective in various autoimmune diseases, but its broader use is constrained by limited supply. This review evaluates the use of immunoglobulins in treatment of neurologic diseases.
Collapse
Affiliation(s)
- S. Živković
- Department of Neurology; University of Pittsburgh Medical Center; Pittsburgh PA USA
| |
Collapse
|
21
|
Ozcan PE, Senturk E, Orhun G, Gumru S, Arican N, Orhan N, Yılmaz CU, Kaya M, Aricioglu F, Esen F. Effects of intravenous immunoglobulin therapy on behavior deficits and functions in sepsis model. Ann Intensive Care 2015; 5:62. [PMID: 26228515 PMCID: PMC4520823 DOI: 10.1186/s13613-015-0062-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/23/2015] [Indexed: 11/18/2022] Open
Abstract
Background We aim to demonstrate behavioral alterations in a sepsis model using intravenous (IV) immunoglobulin G (IgG) and immunoglobulins enriched with IgA and IgM (IgGAM). Methods We divided 48 Wistar albino rats into five groups: control group, sham-operated group (only antibiotic treatment), cecal ligation and puncture (CLP) group (CLP plus antibiotic treatment), IgG group (250 mg/kg IV IgG) and IgGAM group (250 mg/kg IV IgGAM). Intravenous immunoglobulins were given 5 min after the CLP procedure. Experimental animals put into three behavioral tasks 10, 30 and 60 days after the surgery; to evaluate the locomotor activity, an open field test was performed, elevated plus maze test was used to measure anxiety levels, and depressive state was assessed by forced swimming test. The effects of therapy which were acquired from the results of these tests were used to estimate the behavioral changes after CLP. Results The mortality rate of 50% in the septic rats decreased to 30 and 20% with the administration of IgG and IgGAM, respectively. Significant changes on locomotor activity and depressive-like behavior were reported in the sepsis group; on the other hand, the treatment with immunoglobulins reduced the symptoms. Treatment with immunoglobulins attenuated the sepsis-related anxiogenic-like responses. Behavioral alterations returned to normal on day 60 in all groups. Conclusions Sepsis caused deterioration on behavioral parameters. Immunoglobulin treatments alleviated the symptoms of functional disturbances and caused early reversal of behavioral deficits in septic animals.
Collapse
Affiliation(s)
- Perihan Ergin Ozcan
- Department of Anesthesiology, Istanbul Faculty of Medicine, Istanbul University, Capa-Fatih, 34039, Istanbul, Turkey,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Matucci A, Maggi E, Vultaggio A. Mechanisms of action of Ig preparations: immunomodulatory and anti-inflammatory effects. Front Immunol 2015; 5:690. [PMID: 25628625 PMCID: PMC4290674 DOI: 10.3389/fimmu.2014.00690] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/23/2014] [Indexed: 01/16/2023] Open
Abstract
Primary immunodeficiency (PID) disorders that predispose patients to recurrent infections require immunoglobulin (Ig) replacement therapy. Ig replacement therapy has been stated as beneficial, although the optimal IgG trough level to be maintained over time in order to minimize infectious risk has not been established. The most common route of administration of Ig has been intravenously, although there are different options, one of them being the subcutaneous route. Ig replacement therapy has been a life-saving treatment for patients suffering from primary and secondary antibody immunodeficiency. The key role of regular Ig replacement in patients with antibody deficiencies is related to the ability to provide specific antibodies that could not be produced by these patients as demonstrated by the reduction of severe infections such as meningitis and pneumonia. The therapeutic benefits of Ig may also be due to an active role in various anti-inflammatory and immunomodulatory activities, which may complicate the clinical picture of PID. Anti-inflammatory activities are seen more generally when intravenous Ig is administered at high dose. The immunomodulatory and anti-inflammatory activities are important not only in the treatment of autoimmune diseases but also in patients suffering from immunodeficiency.
Collapse
Affiliation(s)
- Andrea Matucci
- Immunoallergology Unit, Department of Biomedicine, Policlinico di Careggi , Florence , Italy
| | - Enrico Maggi
- Immunology and Cellular Therapies Unit, Department of Biomedicine, Centre Denothe, Policlinico di Careggi, University of Florence , Florence , Italy
| | - Alessandra Vultaggio
- Immunoallergology Unit, Department of Biomedicine, Policlinico di Careggi , Florence , Italy
| |
Collapse
|
23
|
High-dose intravenous immunoglobulin exerts neuroprotective effect in the rat model of neonatal asphyxia. Pediatr Res 2014; 75:612-7. [PMID: 24488088 DOI: 10.1038/pr.2014.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/12/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND Neonatal asphyxia is one of the leading causes of death in newborn and permanent neurological disabilities in surviving children. The underlying hypoxic-ischemic (HI) injury triggers an inflammatory response leading to neuronal damage. Here, we tested the hypothesis that high-dose intravenous immunoglobulin (IVIG) could exert immunomodulatory effect in rat pups subjected to HI injury. METHODS HI injury was induced in 7-d-old pups by ligating the common carotid artery followed by exposure to 8% oxygen for 2 h. Brain infarction was evaluated by imaging stained coronal brain sections. Neurological deficits were assessed in weeks 1 through 4 after HI. Western blotting and immunohistochemistry were used to assess complement fragment deposition in the brain tissue. RESULTS Treatment with IVIG at 2 g/kg significantly and in a dose-responsive manner reduced brain infarction size as well as mortality and neurological deficits caused by HI. Anatomical and functional improvements in IVIG-treated pups correlated with decreased deposition of C3b complement fragments in the injured brain hemisphere. CONCLUSION IVIG significantly improved the outcome of HI injury in rat pups and could potentially be used for the treatment of human neonatal asphyxia to target proinflammatory complement fragments.
Collapse
|
24
|
Yadav VR, Nag O, Awasthi V. Biological evaluation of liposome-encapsulated hemoglobin surface-modified with a novel PEGylated nonphospholipid amphiphile. Artif Organs 2014; 38:625-33. [PMID: 24749870 DOI: 10.1111/aor.12304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Traumatic injury is often associated with hemorrhagic shock. Liposome-encapsulated hemoglobin (LEH) is being developed as an artificial oxygen carrier to address post-hemorrhage oxygen and volume deficit. Here, we report a new composition of LEH based on the use of polyethylene glycol (PEG2K ) conjugated with nonphospholipid hexadecylcarbamoylmethylhexadecanoate (HDAS) to modify the surface of LEH particles. LEH was manufactured by the high-pressure homogenization method using dipalmitoylphosphatidylcholine (∼38 mol%), cholesterol (∼38 mol%), HDAS (∼20 mol%), and highly purified stroma-free human hemoglobin. HDAS-PEG2K was postinserted into the resultant LEH to generate HDAS-PEG2K -LEH. We investigated the potential immune response to HDAS-PEG2K -LEH in a mice model. At the same time, the preparation was tested in a rat model to study the effect of repeated HDAS-PEG2K -LEH injection over 4 weeks. We found that HDAS-PEG2K modification substantially reduced the circulating levels of anaphylatoxins C3a and C5a, as well as plasma levels of thromboxane B2, in mice. Repeated injections of HDAS-PEG2K -LEH in rats did not appear to alter its clearance profile after 4 weeks of treatment. No antibody response against human hemoglobin or PEG was detected in rat plasma. Histological observations of lung, liver, spleen, and kidney were not significantly different between saline-treated rats and HDAS-PEG2K -LEH-treated rats. Immunohistochemical staining for rat heme oxygenase-1 (HO-1) did not show induced expression of HO-1 in these organs. These results suggest that the new surface modification of LEH is immune-neutral and does not adversely affect histology even after repeated administration.
Collapse
Affiliation(s)
- Vivek R Yadav
- Department of Pharmaceutical Sciences, Research Imaging Facility, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | | | | |
Collapse
|
25
|
Rosin NL, Gareau AJ, Betsch D, Falkenham A, Sopel MJ, Lee TD, Légaré JF. Antibody therapy can enhance AngiotensinII-induced myocardial fibrosis. FIBROGENESIS & TISSUE REPAIR 2014; 7:6. [PMID: 24721281 PMCID: PMC4021636 DOI: 10.1186/1755-1536-7-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 03/19/2014] [Indexed: 12/12/2022]
Abstract
Background Myocardial fibrosis is a pathological process that is characterized by disrupted regulation of extracellular matrix proteins resulting in permanent scarring of the heart tissue and eventual diastolic heart failure. Pro-fibrotic molecules including transforming growth factor-β and connective tissue growth factor are expressed early in the AngiotensinII (AngII)-induced and other models of myocardial fibrosis. As such, antibody-based therapies against these and other targets are currently under development. Results In the present study, C57Bl/6 mice were subcutaneously implanted with a mini-osmotic pump containing either AngII (2.0 μg/kg/min) or saline control for 3 days in combination with mIgG (1 mg/kg/d) injected through the tail vein. Fibrosis was assessed after picosirius red staining of myocardial cross-sections and was significantly increased after AngII exposure compared to saline control (11.37 ± 1.41%, 4.94 ± 1.15%; P <0.05). Non-specific mIgG treatment (1 mg/kg/d) significantly increased the amount of fibrosis (26.34 ± 3.03%; P <0.01). However, when AngII exposed animals were treated with a Fab fragment of the mIgG or mIgM, this exacerbation of fibrosis was no longer observed (14.49 ± 2.23%; not significantly different from AngII alone). Conclusions These data suggest that myocardial fibrosis was increased by the addition of exogenous non-specific antibodies in an Fc-mediated manner. These findings could have substantial impact on the future experimental design of antibody-based therapeutics.
Collapse
Affiliation(s)
- Nicole L Rosin
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alison J Gareau
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Devin Betsch
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alec Falkenham
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mryanda J Sopel
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Timothy Dg Lee
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada ; Department of Surgery, Dalhousie University, Halifax, Nova Scotia, Canada ; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jean-Francois Légaré
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada ; Department of Surgery, Dalhousie University, Halifax, Nova Scotia, Canada ; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada ; Department of Surgery, New Halifax Infirmary, 1796 Summer St., Room 2269, Halifax, NS B3H 3A7, Canada
| |
Collapse
|
26
|
Widiapradja A, Santro T, Basta M, Sobey CG, Manzanero S, Arumugam TV. Intravenous immunoglobulin (IVIg) provides protection against endothelial cell dysfunction and death in ischemic stroke. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2014; 6:7. [PMID: 24991401 PMCID: PMC4079166 DOI: 10.1186/2040-7378-6-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/06/2014] [Indexed: 04/27/2023]
Abstract
BACKGROUND The brain endothelium is a key component of the blood brain barrier which is compromised following ischemia, allowing infiltration of damaging immune cells and other inflammatory molecules into the brain. Intravenous immunoglobulin (IVIg) is known to reduce infarct size in a mouse model of experimental stroke. FINDINGS Flow cytometry analysis showed that the protective effect of IVIg in ischemia and reperfusion injury in vivo is associated with reduced leukocyte infiltration, suggesting an involvement of the endothelium. In an in vitro model of ischemia, permeability analysis of the mouse brain endothelial cell line bEnd.3 revealed that IVIg prevented the loss of permeability caused by oxygen and glucose deprivation (OGD). In addition, western blot analysis of these brain endothelial cells showed that IVIg prevented the down-regulation of tight junction proteins claudin 5 and occludin and the decline in anti-apoptotic proteins Bcl-2 and Bcl-XL caused by OGD. CONCLUSION IVIg protects endothelial cells from ischemic insult. These studies support the use of IVIg as a pharmacological intervention for stroke therapy.
Collapse
Affiliation(s)
- Alexander Widiapradja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Tomislav Santro
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | | | - Silvia Manzanero
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
27
|
Ramadass M, Ghebrehiwet B, Smith RJ, Kew RR. Generation of multiple fluid-phase C3b:plasma protein complexes during complement activation: possible implications in C3 glomerulopathies. THE JOURNAL OF IMMUNOLOGY 2013; 192:1220-30. [PMID: 24367026 DOI: 10.4049/jimmunol.1302288] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The complement system is tightly regulated to safeguard against tissue damage that results from unwanted activation. The key step of C3 cleavage to C3b is regulated by multiple mechanisms that control the initiation and extent of activation. This study demonstrated that C3b:plasma protein complexes form in the fluid-phase during complement activation. Several different plasma proteins displayed a discrete high molecular SDS-resistant band when any of the three complement activating pathways were triggered in normal human serum or plasma. Serum depleted of individual complement proteins revealed that C3 and factors B and D were essential for complex formation. Inactivation of the thioester bond in C3 also prevented complex formation. In vitro, complexes could be generated using four purified proteins-C3, factor B, factor D, and target protein-and Mg(2+) to allow C3 convertase formation. These studies showed that the complexes consisted of a plasma protein covalently bound to C3b in a 1:1 molar ratio; the C3b portion was rapidly degraded by factors H and I. Analysis of plasma samples from patients with dense deposit disease and C3 glomerulonephritis demonstrated that C3b:protein complexes form spontaneously in the blood of patients with dense deposit disease and, to a lesser extent, in C3 glomerulonephritis patients, but not in healthy controls. This finding supports the underlying hypothesis that these C3 glomerulopathies are diseases of fluid-phase complement dysregulation. These complexes could normally function as a passive mechanism to intercept C3b from depositing on host cells. However, excessive generation and/or defective clearance of fluid-phase C3b:protein complexes may have pathological consequences.
Collapse
Affiliation(s)
- Mahalakshmi Ramadass
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, NY 11794
| | | | | | | |
Collapse
|
28
|
|
29
|
Dengue nonstructural protein-1 status is not associated to circulating levels of interleukin-17, C-reactive protein and complement in children with acute dengue. J Clin Virol 2013. [DOI: 10.1016/j.jcv.2012.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Autoimmune diseases in the intensive care unit. An update. Autoimmun Rev 2013; 12:380-95. [DOI: 10.1016/j.autrev.2012.06.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/12/2012] [Indexed: 12/18/2022]
|
31
|
Nair V, Sawinski D, Akalin E, Friedlander R, Ebcioglu Z, Sehgal V, Dinavahi R, Khaim R, Ames S, Lerner S, Murphy B, Bromberg JS, Heeger PS, Schröppel B. Effect of high-dose intravenous immunoglobulin on anti-HLA antibodies in sensitized kidney transplant candidates. Clin Transplant 2012; 26:E261-8. [PMID: 22686949 DOI: 10.1111/j.1399-0012.2012.01657.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Limited data exist on the effect of intravenous immunoglobulin (IVIg) on anti-HLA antibodies as determined by solid-phase assays. We reviewed our experience treating sensitized wait-listed kidney transplant recipients with IVIg as a method for desensitization and report our results utilizing Luminex single antigen (LSA) bead assay to quantify antibody reactivity (MFI). Fifteen patients with a cPRA > 40% received 2 g/kg IVIg per month for four months or until transplanted. LSA testing was performed before and after IVIg. Median MFI for anti-class I antibodies fell in 11 (73%) and increased in 4 (27%) patients after IVIg. Similar significant changes in MFI for anti-class II antibodies were observed in 10 patients (66%). Administration of IVIg was associated with a modest decrease in reactivity to both class I and II HLA antigens (median MFI change 493 and 1110, respectively; p < 0.0001) but did not significantly alter mean cPRA (85% before IVIg vs. 80% after IVIg; p = 0.1). Our data suggest a smaller effect of IVIg on HLA antibody reactivity than previously described, leading us to question how best to measure the efficacy of a desensitization protocol in current practice.
Collapse
Affiliation(s)
- Vinay Nair
- Division of Nephrology, Mount Sinai School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, UDA.
| |
Collapse
|
33
|
Abstract
For more than 30 years, intravenously administered immunoglobulins (ivIG) have been used to treat primary and secondary syndromes of immune deficiency. Increasing insight into pathomechanisms of severe sepsis and septic shock have led to the implementation of ivIG therapy in the strategies for adjunctive therapy in sepsis in both adults and children. Direct antitoxic effects, as well as indirect immunomodulatory mechanisms of ivIG have been described in the literature and were the basis for the rationale to use these substances in life-threatening infections and hyperinflammatory states. Several clinical trials have been performed, most of them as minor, investigator-initiated protocols. This review summarizes the results of clinical investigations and systematic meta-analyses that have implications for the development of therapeutic strategies, and international guidelines for the management of severe sepsis and septic shock in adult patients.
Collapse
Affiliation(s)
- Susanne Toussaint
- Department of Anesthesia, Critical Care Medicine, and Pain Management, Vivantes - Klinikum Neukoelln, Berlin, Germany
| | | |
Collapse
|
34
|
Ducancel F, Muller BH. Molecular engineering of antibodies for therapeutic and diagnostic purposes. MAbs 2012; 4:445-57. [PMID: 22684311 DOI: 10.4161/mabs.20776] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
During the past ten years, monoclonal antibodies (mAbs) have taken center stage in the field of targeted therapy and diagnosis. This increased interest in mAbs is due to their binding accuracy (affinity and specificity) together with the original molecular and structural rules that govern interactions with their cognate antigen. In addition, the effector properties of antibodies constitute a second major advantage associated with their clinical use. The development of molecular and structural engineering and more recently of in vitro evolution of antibodies has opened up new perspectives in the de novo design of antibodies more adapted to clinical and diagnostic use. Thus, efforts are regularly made by researchers to improve or modulate antibody recognition properties, to adapt their pharmacokinetics, engineer their stability, and control their immunogenicity. This review presents the latest molecular engineering results on mAbs with therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Frédéric Ducancel
- CEA, iBiTec-S, Department of Pharmacology and Immunoanalysis, Laboratory of Antibody Engineering for Health, Gif-sur-Yvette, France.
| | | |
Collapse
|
35
|
Ramos-Medina R, Corbí AL, Sánchez-Ramón S. [Intravenous immunoglobulin: immunomodulatory key of the immune system]. Med Clin (Barc) 2012; 139:112-7. [PMID: 22285062 DOI: 10.1016/j.medcli.2011.11.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/12/2011] [Accepted: 11/17/2011] [Indexed: 12/30/2022]
Abstract
The mechanisms of action of intravenous immunoglobulins (IVIG) are complex and mostly reproduce those of the natural immunoglobulin G (IgG) in our organism. The therapeutic doses used range from substitutive (200-400mg/kg of body weight) in immunodeficiencies to high doses (1-2g/kg of body weight) in autoimmune or inflammatory diseases. The paradoxical pro- or anti-inflammatory effects of IVIG are based on the modulation of the expression of activating versus inhibitory Fc receptors, the type and stage of maturation of the target cell. This huge diversity of actions may explain the extensive and varied range of clinical applications of IVIG nowadays (immunodeficiencies, autoimmune diseases, degenerative diseases such as Alzheimer's, and cancer). On the other hand, biological therapies with monoclonal antibodies mostly consist of IgG molecules with unique antigen specificity, and currently represent a therapeutic field expanding in various pathologies including cancer and diseases of immunological basis. The effects of IgG are added to their specific effects on molecules target.
Collapse
Affiliation(s)
- Rocío Ramos-Medina
- Unidad de Inmunología Clínica, Departamento de Inmunología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | | |
Collapse
|
36
|
Widiapradja A, Vegh V, Lok KZ, Manzanero S, Thundyil J, Gelderblom M, Cheng YL, Pavlovski D, Tang SC, Jo DG, Magnus T, Chan SL, Sobey CG, Reutens D, Basta M, Mattson MP, Arumugam TV. Intravenous immunoglobulin protects neurons against amyloid beta-peptide toxicity and ischemic stroke by attenuating multiple cell death pathways. J Neurochem 2012; 122:321-32. [PMID: 22494053 DOI: 10.1111/j.1471-4159.2012.07754.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intravenous immunoglobulin (IVIg) preparations obtained by fractionating blood plasma, are increasingly being used increasingly as an effective therapeutic agent in treatment of several inflammatory diseases. Its use as a potential therapeutic agent for treatment of stroke and Alzheimer's disease has been proposed, but little is known about the neuroprotective mechanisms of IVIg. In this study, we investigated the effect of IVIg on downstream signaling pathways that are involved in neuronal cell death in experimental models of stroke and Alzheimer's disease. Treatment of cultured neurons with IVIg reduced simulated ischemia- and amyloid βpeptide (Aβ)-induced caspase 3 cleavage, and phosphorylation of the cell death-associated kinases p38MAPK, c-Jun NH2 -terminal kinase and p65, in vitro. Additionally, Aβ-induced accumulation of the lipid peroxidation product 4-hydroxynonenal was attenuated in neurons treated with IVIg. IVIg treatment also up-regulated the anti-apoptotic protein, Bcl2 in cortical neurons under ischemia-like conditions and exposure to Aβ. Treatment of mice with IVIg reduced neuronal cell loss, apoptosis and infarct size, and improved functional outcome in a model of focal ischemic stroke. Together, these results indicate that IVIg acts directly on neurons to protect them against ischemic stroke and Aβ-induced neuronal apoptosis by inhibiting cell death pathways and by elevating levels of the anti-apoptotic protein Bcl2.
Collapse
Affiliation(s)
- Alexander Widiapradja
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Intravenous immunoglobulins prevent the breakdown of the blood-brain barrier in experimentally induced sepsis. Crit Care Med 2012; 40:1214-20. [DOI: 10.1097/ccm.0b013e31823779ca] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
38
|
Padmore RF. Hemolysis upon intravenous immunoglobulin transfusion. Transfus Apher Sci 2011; 46:93-6. [PMID: 22169381 DOI: 10.1016/j.transci.2011.11.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 11/16/2011] [Indexed: 10/14/2022]
Abstract
Intravenous immunoglobulin (IVIG) is a mainstay of therapy in many disorders. An uncommon adverse side effect is IVIG-related hemolysis. Risk factors for IVIG-related hemolysis have been identified, including high dose IVIG given to non-O blood group recipients with an underlying inflammatory state. IVIG-related hemolysis has been linked to anti-A and anti-B hemagglutinins in the IVIG preparations and may involve both IgG and complement mediated hemolysis. A two-hit mechanism with threshold effect is proposed for IVIG-related hemolysis. Strategies exist to minimize or avoid IVIG-related hemolysis.
Collapse
Affiliation(s)
- Ruth F Padmore
- The Ottawa Hospital, General Campus, 501 Smyth Road, Ottawa, ON, Canada K1H 8L6.
| |
Collapse
|
39
|
Jordan SC, Toyoda M, Vo AA. Regulation of immunity and inflammation by intravenous immunoglobulin: relevance to solid organ transplantation. Expert Rev Clin Immunol 2011; 7:341-8. [PMID: 21595600 DOI: 10.1586/eci.11.10] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Intravenous immunoglobulin (IVIg) products are derived from pooled human plasma from thousands of donors and have been used for the treatment of primary immunodeficiency disorders for more than 30 years. IVIg products are also effective in the treatment of autoimmune and inflammatory disorders, however, the precise mechanism(s) of action are not known. Recent data suggest that IVIg has a much broader ability to regulate cellular immunity, including innate and adaptive components. IVIg-induced upregulation of Fcγ receptor IIB on B cells appears to be an important mode of action in suppression of antigen-presenting cell activity and antibody production. IVIg is also a recently recognized modifier of complement activation and injury. Analysis of clinical studies examining the use of IVIg in desensitization protocols and for treatment of antibody-mediated rejection in transplant recipients are supportive. Here, we discuss these important advancements and their relevance to transplant medicine.
Collapse
Affiliation(s)
- Stanley C Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | |
Collapse
|
40
|
Jordan SC, Reinsmoen N, Peng A, Lai CH, Cao K, Villicana R, Toyoda M, Kahwaji J, Vo AA. Advances in diagnosing and managing antibody-mediated rejection. Pediatr Nephrol 2010; 25:2035-45; quiz 2045-8. [PMID: 20077121 PMCID: PMC2923704 DOI: 10.1007/s00467-009-1386-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/05/2009] [Accepted: 11/02/2009] [Indexed: 02/07/2023]
Abstract
Antibody-mediated rejection (AMR) is a unique, significant, and often severe form of allograft rejection that is not amenable to treatment with standard immunosuppressive medications. Significant advances have occurred in our ability to predict patients at risk for, and to diagnose, AMR. These advances include the development of newer anti-human leukocyte antigen (HLA)-antibody detection techniques and assays for non-HLA antibodies associated with AMR. The pathophysiology of AMR suggests a prime role for antibodies, B cells and plasma cells, but other effector molecules, especially the complement system, point to potential targets that could modify the AMR process. An emerging and potentially larger problem is the development of chronic AMR (CAMR) resulting from de novo donor-specific anti-HLA antibodies (DSA) that emerge more than 100 days posttransplantation. Therapeutic options include: (1) High-dose intravenously administered immunoglobulin (IVIG), which has many potential benefits. (2) The use of IVIG+rituximab (anti-CD20, anti-B cell). (3) The combination of plasmapheresis (PP)+low-dose IVIG with or without rituximab. Data support the efficacy of all of the above approaches. Newer approaches to treating AMR include using the proteosome inhibitor (bortezomib), which induces apoptosis in plasma cells, and eculizumab (anti-C5, anticomplement monoclonal antibody).
Collapse
Affiliation(s)
- Stanley C Jordan
- The Transplant Immunotherapy Program, Comprehensive Transplant Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
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: 3.0] [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
|
42
|
Rispens T, Himly M, Heer POD, Bleker THD, Aalberse RC. Traces of pFc’ in IVIG interact with human IgG Fc domains and counteract aggregation. Eur J Pharm Sci 2010; 40:62-8. [DOI: 10.1016/j.ejps.2010.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 02/09/2010] [Accepted: 03/01/2010] [Indexed: 10/19/2022]
|
43
|
Durandy A, Kaveri SV, Kuijpers TW, Basta M, Miescher S, Ravetch JV, Rieben R. Intravenous immunoglobulins--understanding properties and mechanisms. Clin Exp Immunol 2010; 158 Suppl 1:2-13. [PMID: 19883419 DOI: 10.1111/j.1365-2249.2009.04022.x] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
High-dose intravenous immunoglobulin (IVIg) preparations are used currently for the treatment of autoimmune or inflammatory diseases. Despite numerous studies demonstrating efficacy, the precise mode of action of IVIg remains unclear. Paradoxically, IgG can exert both pro- and anti-inflammatory activities, depending on its concentration. The proinflammatory activity of low-dose IVIg requires complement activation or binding of the Fc fragment of IgG to IgG-specific receptors (FcgammaR) on innate immune effector cells. In contrast, when administered in high concentrations, IVIg has anti-inflammatory properties. How this anti-inflammatory effect is mediated has not yet been elucidated fully, and several mutually non-exclusive mechanisms have been proposed. This paper represents the proceedings of a session entitled 'IVIg--Understanding properties and mechanisms' at the 6th International Immunoglobulin Symposium that was held in Interlaken on 26-28 March 2009. The presentations addressed how IgG may affect the cellular compartment, evidence for IVIg-mediated scavenging of complement fragments, the role of the dimeric fraction of IVIg, the anti-inflammatory properties of the minor fraction of sialylated IgG molecules, and the genetic organization and variation in FcgammaRs. These findings demonstrate the considerable progress that has been made in understanding the mechanisms of action of IVIgs, and may influence future perspectives in the field of Ig therapy.
Collapse
Affiliation(s)
- A Durandy
- INSERM U768, Université Paris-Descartes, Hospital Necker, Paris, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Fernández-Cruz E, Alecsandru D, Sánchez Ramón S, Ramón SS. Mechanisms of action of immune globulin. Clin Exp Immunol 2009; 157 Suppl 1:1-2. [PMID: 19630862 DOI: 10.1111/j.1365-2249.2009.03955.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- E Fernández-Cruz
- Hospital General Universitario Gregorio Marañón, Servicio de Inmunología Clínica, Comunidad de Madrid, Madrid, Spain.
| | | | | | | |
Collapse
|
45
|
Abstract
The complement system is an essential component of innate immunity that has been more recently recognized as an unexpected player in various pathological states. These include age-related macular degeneration, atypical haemolytic uraemic syndrome, allergy, foetal loss, and axonal and myelin degradation after trauma. Its importance has also been recognized in physiological processes including haematopoietic stem cell homing to the bone marrow, liver regeneration and modulation of adaptive immune responses. Although the complement system has long been known to be involved in autoimmune and inflammatory diseases, few agents that target the complement system are currently approved for clinical use. However, renewed interest in modulating this system in various pathological conditions has emerged, and several agents are now in development.
Collapse
|
46
|
Nielsen DG. The relationship of interacting immunological components in dengue pathogenesis. Virol J 2009; 6:211. [PMID: 19941667 PMCID: PMC2789730 DOI: 10.1186/1743-422x-6-211] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 11/27/2009] [Indexed: 12/18/2022] Open
Abstract
The World Health Organization (WHO) estimates that there are over 50 million cases of dengue fever reported annually and approximately 2.5 billion people are at risk. Mild dengue fever presents with headache, fever, rash, myalgia, osteogenic pain, and lethargy. Severe disease can manifest as dengue shock syndrome (DSS) or dengue hemorrhagic fever (DHF). Symptoms of DSS/DHF are leukopenia, low blood volume and pressure encephalitis, cold and sweaty skin, gastrointestinal bleeding, and spontaneous bleeding from gums and nose. Currently, there are no therapeutics available beyond supportive care and untreated complicated dengue fever can have a 50% mortality rate. According to WHO DSS/DHF is the leading cause of childhood mortality in some Asian countries. Dendritic cells are professional antigen presenting cells that are primary targets in a dengue infection. Dengue binds to Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin (DC-SIGN). DC-SIGN has a high affinity for ICAM3 which is expressed in activating T-cells. Previous studies have demonstrated an altered T-cell phenotype expressed in dengue infected patients that could be potentially mediated by dengue-infected DCs. Dengue is enhanced by three interacting components of the immune system. Dengue begins by infecting dendritic cells which in immature dendritic cells is mediated by DC-SIGN. In mature dendritic cells, antibodies can enhance dengue infection via Fc receptors. Downstream of dendritic cells T-cells become activated and generate the very cytokines implicated in vascular leak and shock in addition to activating effector cells. Both the virus and the antibodies are involved in release of complement and anaphylatoxins which can cause or exacerbate DHF/DSS. These systems are inextricable and strongly associated with dengue pathogenesis.
Collapse
Affiliation(s)
- David G Nielsen
- Department of Microbiology and Immunology, Tulane University, New Orleans, Louisiana 70112-2699, USA.
| |
Collapse
|
47
|
Spycher M, Matozan K, Minnig K, Zehnder R, Miescher S, Hoefferer L, Rieben R. In vitrocomparison of the complement-scavenging capacity of different intravenous immunoglobulin preparations. Vox Sang 2009; 97:348-54. [DOI: 10.1111/j.1423-0410.2009.01217.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Abstract
Intravenous immunoglobulin (IVIG) products are derived from pooled human plasma and have been used for the treatment of primary immunodeficiency disorders for more than 25 years. IVIG products are also effective in the treatment of autoimmune and inflammatory disorders; however, the precise mechanism(s) of action is not known. Recent investigations suggest that IVIG has a much broader ability to regulate cellular immunity including innate and adaptive components. IVIG is also a recently recognized modifier of complement activation and injury. Here, we discuss these important advancements and how this knowledge applies to desensitization protocols and to the treatment of antibody-mediated rejection.
Collapse
|
49
|
Recombinant antibody therapeutics: the impact of glycosylation on mechanisms of action. Trends Pharmacol Sci 2009; 30:356-62. [DOI: 10.1016/j.tips.2009.04.007] [Citation(s) in RCA: 265] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 04/15/2009] [Accepted: 04/15/2009] [Indexed: 01/20/2023]
|
50
|
Galeotti C, Maddur M, Kazatchkine MD, Mouthon L, Kaveri SV. Immunoglobulines intraveineuses dans les maladies auto-immunes et inflammatoires : au-delà d’une simple substitution. Transfus Clin Biol 2009; 16:75-9. [DOI: 10.1016/j.tracli.2009.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/18/2009] [Indexed: 01/10/2023]
|