Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Neurotherapeutics ; 19(3): 729-740, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34997443

RESUMEN

Autoantibodies are increasingly recognized for their pathogenic potential in a growing number of neurological diseases. While myasthenia gravis represents the prototypic antibody (Ab)-mediated neurological disease, many more disorders characterized by Abs targeting neuronal or glial antigens have been identified over the past two decades. Depletion of humoral immune components including immunoglobulin G (IgG) through plasma exchange or immunoadsorption is a successful therapeutic strategy in most of these disease conditions. The neonatal Fc receptor (FcRn), primarily expressed by endothelial and myeloid cells, facilitates IgG recycling and extends the half-life of IgG molecules. FcRn blockade prevents binding of endogenous IgG to FcRn, which forces these antibodies into lysosomal degradation, leading to IgG depletion. Enhancing the degradation of endogenous IgG by FcRn-targeted therapies proved to be a powerful therapeutic approach in patients with generalized MG and is currently being tested in clinical trials for several other neurological diseases including autoimmune encephalopathies, neuromyelitis optica spectrum disorders, and inflammatory neuropathies. This review illustrates mechanisms of FcRn-targeted therapies and appraises their potential to treat neurological diseases.


Asunto(s)
Enfermedades del Sistema Nervioso , Receptores Fc , Humanos , Inmunoglobulina G , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Neurología , Receptores Fc/antagonistas & inhibidores , Receptores Fc/química
2.
CPT Pharmacometrics Syst Pharmacol ; 11(1): 116-128, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34735735

RESUMEN

Rozanolixizumab is a fully humanized high-affinity anti-human neonatal Fc receptor (FcRn) monoclonal antibody (mAb) that accelerates the removal of circulating immunoglobulin G (IgG), including pathogenic IgG autoantibodies, via the natural lysosomal degradation pathway. The aim of this study was to develop a pharmacokinetic/pharmacodynamic (PK/PD) model characterizing the effect of rozanolixizumab on IgG levels in cynomolgus monkeys, translate it into humans to support the first-in-human (FIH) rozanolixizumab clinical trial study design, and, ultimately, develop a PK/PD model in humans. Simulations from the preclinical model were performed to predict IgG responses in humans and select clinically relevant doses in the FIH study. Good alignment was observed between predicted and observed reductions in IgG, which increased with increasing dose in the FIH study. The model successfully described the PK of the 4 and 7 mg/kg intravenous (i.v.) dose groups, although the PKs were underpredicted for the 1 mg/kg i.v. dose group. Updating the model with subsequent human data identified parameters that deviated from preclinical assumptions. The updated PK/PD model was able to effectively characterize the PK FcRn-IgG nonlinear system in response to rozanolixizumab in the FIH data.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Inmunoglobulina G/efectos de los fármacos , Receptores Fc/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Simulación por Computador , Relación Dosis-Respuesta a Droga , Antígenos de Histocompatibilidad Clase I , Humanos , Macaca fascicularis , Modelos Biológicos
3.
Pharmacol Res ; 173: 105873, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34500060

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease targeting the synovium. Previous studies have found that IgD may be a potential target for the treatment of RA. We designed a new type of fusion protein, hIgDFc-Ig (DG), to block the binding of IgD to IgD receptor (IgDR). In this study, we found that DG has a significant therapeutic effect in mice with collagen-induced arthritis (CIA). DG improved the claw of irritation symptoms in these mice, inhibited the pathological changes in spleen and joint tissues, and had a moderating effect on B cell subsets at different inflammatory stages. Moreover, DG could also decrease the levels of IgA, IgD, IgM and IgG subtypes of immunoglobulin in the serum of mice with CIA. In vitro, B cell antigen receptor (BCR) knockout Ramos cells were established using the CRISPR/Cas9 technology to further study the activation of BCR signalling by IgD and the effect of DG. We found that the therapeutic effect of DG in mice with CIA may be achieved by inhibiting the activation of BCR signalling by IgD, which may be related to the activation of Igß. In summary, DG may be a potential biological agent for the treatment of RA and it has broad application prospects in the future.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Inmunoglobulinas/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/farmacología , Ratones , Ratones Endogámicos DBA , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores Fc/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Quinasa Syk/metabolismo , Timo/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
4.
Front Immunol ; 12: 728322, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512662

RESUMEN

Novel molecules that directly target the neonatal Fc receptor (FcRn) and/or Fc gamma receptors (FcγRs) are emerging as promising treatments for immunoglobulin G (IgG)-dependent autoimmune pathologies. Mutated Fc regions and monoclonal antibodies that target FcRn are currently in clinical development and hold promise for reducing the levels of circulating IgG. Additionally, engineered structures containing multimeric Fc regions allow the dual targeting of FcRn and FcγRs; however, their tolerance needs to first be validated in phase I clinical studies. Here, for the first time, we have developed a modified monomeric recombinant Fc optimized for binding to all FcRns and FcγRs without the drawback of possible tolerance associated with FcγR cross-linking. A rational approach using Fc engineering allowed the selection of LFBD192, an Fc with a combination of six mutations that exhibits improved binding to human FcRn and FcγR as well as mouse FcRn and FcγRIV. The potency of LFBD192 was compared with that of intravenous immunoglobulin (IVIg), an FcRn blocker (Fc-MST-HN), and a trimeric Fc that blocks FcRn and/or immune complex-mediated cell activation through FcγR without triggering an immune reaction in several in vitro tests and validated in three mouse models of autoimmune disease.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/prevención & control , Autoinmunidad/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Receptores Fc/antagonistas & inhibidores , Receptores de IgG/antagonistas & inhibidores , Animales , Antirreumáticos/metabolismo , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Unión Competitiva , Complemento C5a/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Interleucina-2/metabolismo , Células Jurkat , Cinética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fagocitosis/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Unión Proteica , Ingeniería de Proteínas , Receptores Fc/genética , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de IgG/genética , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Vías Secretoras , Transducción de Señal , Células THP-1
5.
Blood Coagul Fibrinolysis ; 32(7): 427-433, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34224465

RESUMEN

Immune thrombocytopenia is a haematological, autoimmune disorder characterized by elevated platelet demolition due to the presence of antiplatelet autoantibodies derived from B cells and to an irregular, deficient process of platelets production in bone marrow. In this review, after a brief presentation of 'old' strategies used nowadays yet, we focused on new drugs used in the treatment of immune thrombocytopenia and their mechanism of action and posology, basing on the last scientific literature. The observation that CoViD-19 can be associated with immune thrombocytopenia is also put in evidence. Particular attention will be dedicated on the concept that the ideal treatment should represent a solution not only for the failure of normal processes of production and survival of platelets, but also it should improve quality of life of patients, with minimum adverse events. Anyway, despite enormous advances of the last years, further investigations are necessary in order to define scrupulously long-term efficacy of new molecules proposed.


Asunto(s)
Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Aminopiridinas/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , COVID-19/complicaciones , COVID-19/inmunología , Antígenos de Histocompatibilidad Clase I , Humanos , Inmunosupresores/uso terapéutico , Morfolinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/inmunología , Pirimidinas/uso terapéutico , Receptores Fc/antagonistas & inhibidores , Receptores de Trombopoyetina/agonistas , SARS-CoV-2/inmunología , Quinasa Syk/antagonistas & inhibidores , Tiazoles/uso terapéutico , Tiofenos/uso terapéutico
6.
J Invest Dermatol ; 141(12): 2820-2828, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34246620

RESUMEN

Pathomechanisms in IgA pemphigus are assumed to rely on Fc-dependent cellular activation by antigen-specific IgA autoantibodies; however, models for the disease and more detailed pathophysiologic data are lacking. In this study, we aimed to establish in vitro models of disease for IgA pemphigus, allowing us to study the effects of the interaction of anti-keratinocyte IgA with cell surface FcαRs. Employing multiple in vitro assays, such as a skin cryosection assay and a human skin organ culture model, in this study, we present mechanistic data for the pathogenesis of IgA pemphigus, mediated by anti-desmoglein 3 IgA autoantibodies. Our results reveal that this disease is dependent on FcαR-mediated activation of leukocytes in the epidermis. Importantly, this cell-dependent pathology can be dose-dependently abrogated by peptide-mediated inhibition of FcαR:IgA-Fc interaction, as confirmed in an additional model for IgA-dependent disease, that is, IgA vasculitis. These data suggest that IgA pemphigus can be modeled in vitro and that IgA pemphigus and IgA vasculitis are FcαR-dependent disease entities that can be specifically targeted in these experimental systems.


Asunto(s)
Inmunoglobulina A/inmunología , Neutrófilos/fisiología , Pénfigo/etiología , Receptores Fc/antagonistas & inhibidores , Antígenos CD/fisiología , Desmogleína 3/inmunología , Proteínas del Ojo/farmacología , Humanos , Pénfigo/inmunología , Fragmentos de Péptidos/farmacología , Receptores Fc/fisiología
7.
J Invest Dermatol ; 141(12): 2858-2865.e4, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34126109

RESUMEN

Pemphigus is a debilitating IgG-mediated autoimmune disease requiring better tolerated, more targeted, and rapid onset therapies. ALXN1830 is a humanized IgG4 antibody that blocks neonatal Fc receptor interactions with IgG. A multicenter, open-label safety and tolerability phase 1b/2 trial (NCT03075904) was conducted in North America from July 2017 to January 2019 and included patients aged ≥18 years with a confirmed diagnosis of pemphigus (vulgaris or foliaceus) and active disease. Dosing included five weekly intravenous doses of ALXN1830 (10 mg/kg) and follow-up through day 112 (study termination). Pharmacokinetics, pharmacodynamics, safety, and efficacy, as evaluated by determining the change in the median pemphigus disease area index, were determined. In this pilot study of eight patients, five weekly infusions of ALXN1830 produced a rapid improvement in the pemphigus disease area index score within 14 days of the first dose. Pemphigus disease area index improvement increased further together with reductions in IgG, circulating immune complexes of IgG, and anti-desmoglein antibodies without affecting albumin, IgM, IgA, or C-reactive protein levels. ALXN1830 was well-tolerated, with headache as the most common adverse event. This study reveals the importance of neonatal Fc receptor in the biology of pemphigus and the potential for use of ALXN1830 in pemphigus treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inmunoglobulina G/uso terapéutico , Pénfigo/tratamiento farmacológico , Receptores Fc/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Autoanticuerpos/sangre , Enfermedad Crónica , Femenino , Antígenos de Histocompatibilidad Clase I , Humanos , Inmunoglobulina G/efectos adversos , Masculino , Persona de Mediana Edad , Pénfigo/inmunología , Adulto Joven
8.
Am J Obstet Gynecol ; 225(2): 120-127, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33839095

RESUMEN

Fetal and neonatal alloimmune thrombocytopenia, the platelet equivalent of hemolytic disease of the fetus and newborn, can have devastating effects on both the fetus and neonate. Current management of fetal and neonatal alloimmune thrombocytopenia in a subsequent affected pregnancy involves antenatal administration of intravenous immune globulin and prednisone to the pregnant woman to prevent the development of severe fetal thrombocytopenia and secondary intracranial hemorrhage in utero. That therapy has proven to be highly effective but is associated with maternal side effects and is expensive. This commentary describes 4 advances that could substantially change the current approach to detecting and managing fetal and neonatal alloimmune thrombocytopenia in the near future. The first would be an introduction of a program to screen all antepartum patients in this country for pregnancies at risk of developing fetal and neonatal alloimmune thrombocytopenia. Strategies to implement this complex process have been described. A second advance is testing of cell-free fetal DNA obtained from maternal blood to noninvasively determine the fetal human platelet antigen 1 genotype. A third, in preliminary development, is creation of a prophylactic product that would be the platelet equivalent of Rh immune globulin (RhoGAM). Finally, a fourth major potential advance is the development of neonatal Fc receptor inhibitors to replace the current medical therapy administered to pregnant women with an affected fetus. Neonatal Fc receptor recycles plasma immunoglobulin G to increase its half-life and is the means by which immunoglobulin G crosses the placenta from the maternal to the fetal circulation. Blocking the neonatal Fc receptor is an ideal way to prevent maternal immunoglobulin G antibody from causing fetal and neonatal alloimmune thrombocytopenia in a fetus at risk of developing that disorder. The pertinent pathophysiology and rationale for each of these developments will be presented in addition to our thoughts relating to steps that must be taken and difficulties that each approach would face for them to be successfully implemented.


Asunto(s)
Antígenos de Plaqueta Humana/inmunología , Factores Inmunológicos/uso terapéutico , Receptores Fc/antagonistas & inhibidores , Trombocitopenia Neonatal Aloinmune/diagnóstico , Trombocitopenia Neonatal Aloinmune/prevención & control , Antígenos de Plaqueta Humana/genética , Ácidos Nucleicos Libres de Células/genética , Desarrollo de Medicamentos , Femenino , Genotipo , Glucocorticoides/uso terapéutico , Antígenos de Histocompatibilidad Clase I , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Integrina beta3/genética , Integrina beta3/inmunología , Intercambio Materno-Fetal/inmunología , Pruebas Prenatales no Invasivas/métodos , Prednisona/uso terapéutico , Embarazo , Diagnóstico Prenatal , Medición de Riesgo , Trombocitopenia Neonatal Aloinmune/inmunología , Trombocitopenia Neonatal Aloinmune/terapia
9.
Transfusion ; 61(5): 1350-1354, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33650699

RESUMEN

FcRn, a non-classical Fc gamma (γ) receptor (FcγR) with near ubiquitous expression, plays key roles in disease pathogenesis and progression though immunoglobulin G (IgG) transport, IgG recycling, and IgG-immune complex clearance. FcRn function can be inhibited using IgG-based and non-IgG-based antagonists, by exploiting the pH-dependent binding affinity of FcRn for the IgG Fc region. FcRn therapeutics have shown promise in murine models and human clinical trials for autoimmune diseases and maternal-fetal immune cytopenias; they appear safe, well-tolerated, and reduce circulating IgG levels. Compared to traditional therapeutics, inhibiting FcRn has fewer adverse side effects and represents a new approach that is less invasive, time-consuming, and costly.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Receptores Fc/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/inmunología , Autoinmunidad/efectos de los fármacos , Desarrollo de Medicamentos , Femenino , Feto/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoglobulina G/inmunología , Terapia Molecular Dirigida , Embarazo , Púrpura Trombocitopénica Idiopática/inmunología , Receptores Fc/inmunología
10.
Clin Transl Sci ; 14(5): 1769-1779, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33742786

RESUMEN

Blockade of the binding between neonatal Fc receptor and IgG-Fc reduces circulating IgG, and thus emerges as a potential therapy for IgG-mediated autoimmune conditions. This was a double blind, randomized, single ascending dose study to evaluate the safety, pharmacokinetics, and pharmacodynamics of HBM9161 (a fully humanized Fc receptor monoclonal antibody) in healthy Chinese volunteers. Subjects were randomized to receive a single s.c. dose of HBM9161 or placebo in a 3:1 ratio in 3 dosing cohorts (340 mg, 510 mg, or 680 mg, respectively), and then followed up for 85 days. Study end points included incidence of adverse event (AE), serum drug concentration, IgG and its subclasses, and anti-drug antibodies (ADAs). Twenty-four subjects were randomized. Dose-dependent reduction of total IgG occurred rapidly from baseline to reach nadir at day 11, then recovered steadily from day 11 to day 85. The mean maximum percentage reductions from baseline total IgG were 21.0 ± 9.3%, 39.8 ± 5.13%, and 41.2 ± 10.4% for subjects receiving HBM9161 340 mg, 510 mg, and 680 mg, respectively. The exposure of HBM9161 (areas under the curve [AUCs] and peak plasma concentration [Cmax ]) increased in a more than dose-proportional manner at the dose examined. All reported AEs were mild in severity. The most reported AEs in the HBM9161 groups were influenza-like illness and rash. Two subjects developed ADA during the study period. A single s.c. dose of HBM9161 results in sustained and dose-dependent IgG reduction, and was well-tolerated at a dose up to 680 mg in Chinese subjects. The data warrant further investigation of its effects in IgG-mediated autoimmune disorders.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Receptores Fc , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , China , Relación Dosis-Respuesta a Droga , Voluntarios Sanos , Antígenos de Histocompatibilidad Clase I , Receptores Fc/antagonistas & inhibidores
11.
Biochem Pharmacol ; 183: 114360, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33301760

RESUMEN

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial membrane hyperplasia, infiltration of inflammatory cells and bone tissue destruction. Although there have been many measures taken for RA therapy in recent years, they are not sufficiently safe or effective. Thus, it is very important to develop new drugs and slow down damage to other healthy organs in the case of RA. Lately, immunoglobulin Fc receptors (FcRs), such as the IgG Fc receptor (FcγR), IgA Fc receptor (FcαR), and IgD Fc receptor (FcδR), have been found to be involved in inducing or suppressing arthritis. FcRs interacting with immune complexes (ICs) are a key factor in the etiopathogenesis of RA. Therefore, an increasing number of methodsfor the targeted treatment of RA with FcRs are emerging, such as recombinant soluble FcγRs, recombinant multimeric Fc fragments and monoclonal antibodies, and have been demonstrated to significantly improve RA symptoms. Simultaneously, certain kinases involved in the downstream signaling of FcRs can also be a target for the treatment of RA, such as Syk and Btk inhibitors. An overview of these FcRs is provided in this review, including a description of FcR-related functions, signaling pathways, and potential FcR-targeting molecules for RA therapy. To date, the initial results of those developed FcR-targeting molecules have been promising. With this, FcRs might offer a better alternative to RA medication. Additionally, further pharmacological characterization and a better understanding of the unique mechanisms of FcR-targeting molecules are necessary.


Asunto(s)
Antirreumáticos/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Receptores Fc/inmunología , Receptores Fc/metabolismo , Animales , Antirreumáticos/administración & dosificación , Artritis Reumatoide/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Receptores Fc/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
12.
Curr Opin Hematol ; 27(6): 392-398, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868670

RESUMEN

PURPOSE OF REVIEW: Intravenous immunoglobulin (IVIg) is an effective treatment for an increasing number of autoimmune and inflammatory conditions. However, IVIg continues to be limited by problems of potential shortages and cost. A number of mechanisms have been described for IVIg, which have been captured in newly emergent IVIg mimetic and IVIg alternative therapies. This review discusses the recent developments in IVIg mimetics and alternatives. RECENT FINDINGS: Newly emergent IVIg mimetics and alternatives capture major proposed mechanisms of IVIg, including FcγR blockade, FcRn inhibition, complement inhibition, immune complex mimetics and sialylated IgG. Many of these emergent therapies have promising preclinical and clinical trial results. SUMMARY: Significant research has been undertaken into the mechanism of IVIg in the treatment of autoimmune and inflammatory disease. Understanding the major IVIg mechanisms has allowed for rational development of IVIg mimetics and alternatives for several IVIg-treatable diseases.


Asunto(s)
Enfermedades Autoinmunes/terapia , Inmunoglobulinas Intravenosas/farmacología , Inmunoglobulinas Intravenosas/uso terapéutico , Inflamación/terapia , Animales , Enfermedades Autoinmunes/inmunología , Activación de Complemento/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inflamación/inmunología , Receptores Fc/antagonistas & inhibidores , Receptores Fc/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
13.
J Allergy Clin Immunol ; 146(3): 467-478, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32896307

RESUMEN

The humoral immune response provides specific, long-lived protection against invading pathogens, via immunoglobulin production and other memory functions. IgG, the most abundant immunoglobulin isotype, has the longest half-life and protects against bacterial and viral infections. The neonatal Fc receptor (FcRn) transports IgG across barriers, for example, the placenta, enhancing fetal humoral immunity to levels similar to their mothers'. Importantly, FcRn, by protecting IgG from intracellular degradation, results in an approximately 21-day circulating IgG half-life and high plasma levels; similarly, FcRn recycles albumin and is the portal of entry for enteric cytopathic human orphan (echo) virus infection. Dysregulated immune responses may lead to antibodies against self-antigens (autoantibodies), resulting in organ-specific or systemic autoimmune diseases. Autoantibody-mediated diseases have been treated by nonspecific immunoglobulin-lowering/modulating therapies, including immunoadsorption, plasma exchange, and high-dose intravenous immunoglobulin. However, targeting FcRn with specific inhibitors results in reduction in only IgG levels. The effectiveness of FcRn inhibitors in autoimmune diseases, including myasthenia gravis and immune thrombocytopenia, provides further evidence that IgG is a primary driver in these autoantibody-mediated diseases. We describe the role of FcRn in human biology, including insights that clinical testing of FcRn inhibitors have provided into FcRn biology and autoimmune disease mechanisms, allowing fact-based speculation on their therapeutic potential.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes/terapia , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulinas Intravenosas/uso terapéutico , Receptores Fc/metabolismo , Animales , Presentación de Antígeno , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad , Embarazo , Receptores Fc/antagonistas & inhibidores , Receptores Fc/inmunología
15.
Am J Clin Dermatol ; 21(6): 765-782, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32860200

RESUMEN

Pemphigus vulgaris (PV) is a severe chronic autoimmune blistering disease that affects the skin and mucous membranes. It is characterized by suprabasal acantholysis due to disruption of desmosomal connections between keratinocytes. Autoantibodies against desmosomal cadherins, desmoglein 3 and 1, have been shown to induce disease. Certain human leukocyte antigen (HLA) types and non-HLA foci confer genetic susceptibility. Until the discovery of corticosteroids in the 1950s, PV was 75% fatal. Since then, multiple PV treatments, such as systemic corticosteroids and adjunctive therapy with immunosuppressive medications (mycophenolate mofetil, azathioprine, cyclophosphamide, cyclosporine, methotrexate, gold, and others) have been introduced; however, none have led to long-term remissions and many have undesired adverse effects. Our growing understanding of the pathophysiologic mechanisms in PV is leading to development of new targeted therapies, such as intravenous immunoglobulin, anti-CD20 monoclonal antibodies, inhibitors of Bruton tyrosine kinase and neonatal Fc receptors, and adoptive cellular transfer, that may result in lasting control of this life-threatening disease.


Asunto(s)
Inmunosupresores/uso terapéutico , Inmunoterapia Adoptiva/métodos , Pénfigo/terapia , Plasmaféresis , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/inmunología , Antígenos CD20/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Terapia Combinada/métodos , Quimioterapia Combinada/métodos , Predisposición Genética a la Enfermedad , Antígenos HLA/genética , Antígenos HLA/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoglobulinas Intravenosas/farmacología , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/farmacología , Terapia Molecular Dirigida/métodos , Pénfigo/genética , Pénfigo/inmunología , Receptores Fc/antagonistas & inhibidores , Receptores Fc/metabolismo , Inducción de Remisión/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Resultado del Tratamiento
16.
Pharmacology ; 105(11-12): 618-629, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32570239

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). SUMMARY: Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. Key Message: The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.


Asunto(s)
Artritis Experimental/etiología , Artritis Experimental/inmunología , Productos Biológicos/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Receptores Fc/inmunología , Receptores Fc/metabolismo , Animales , Productos Biológicos/farmacología , Humanos , Fragmentos Fc de Inmunoglobulinas/farmacología , Inmunoglobulinas Intravenosas/farmacología , Inmunoglobulinas Intravenosas/uso terapéutico , Ratones , Receptores Fc/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
17.
Am J Hematol ; 95(2): 178-187, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31821591

RESUMEN

Primary immune thrombocytopenia (ITP) is an acquired autoimmune bleeding disorder, characterized by a low platelet count (<100 × 109 /L) in the absence of other causes associated with thrombocytopenia. In most patients, IgG autoantibodies directed against platelet receptors can be detected. They accelerate platelet clearance and destruction, inhibit platelet production, and impair platelet function, resulting in increased risk of bleeding and impaired quality of life. Efgartigimod is a human IgG1 antibody Fc-fragment, a natural ligand of the neonatal Fc receptor (FcRn), engineered for increased affinity to FcRn, while preserving its characteristic pH-dependent binding. Efgartigimod blocks FcRn, preventing IgG recycling, and causing targeted IgG degradation. In this Phase 2 study, 38 patients were randomized 1:1:1 to receive four weekly intravenous infusions of either placebo (N = 12) or efgartigimod at a dose of 5 mg/kg (N = 13) or 10 mg/kg (N = 13). This short treatment cycle of efgartigimod in patients with ITP, predominantly refractory to previous lines of therapy, was shown to be well tolerated, and demonstrated a favorable safety profile consistent with Phase 1 data. Efgartigimod induced a rapid reduction of total IgG levels (up to 63.7% mean change from baseline), which was associated with clinically relevant increases in platelet counts (46% patients on efgartigimod vs 25% on placebo achieved a platelet count of ≥50 × 109 /L on at least two occasions, and 38% vs 0% achieved ≥50 × 109 /L for at least 10 cumulative days), and a reduced proportion of patients with bleeding. Taken together, these data warrant further evaluation of FcRn antagonism as a novel therapeutic approach in ITP.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoglobulina G/uso terapéutico , Púrpura Trombocitopénica Idiopática , Receptores Fc/antagonistas & inhibidores , Adulto , Anciano , Método Doble Ciego , Femenino , Estudios de Seguimiento , Antígenos de Histocompatibilidad Clase I/sangre , Humanos , Masculino , Persona de Mediana Edad , Recuento de Plaquetas , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Receptores Fc/sangre
18.
Autoimmun Rev ; 18(10): 102366, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31404703

RESUMEN

In recent years, there has been a surge in the research and development of novel molecules as potential therapeutic alternatives to traditional treatments (such as intravenous immunoglobulins) for autoimmune disorders. The aim of this review is to describe different drug development strategies and evaluate how various molecules have performed in clinical trials to date. Broadly, three main approaches have been pursued. Recombinant fragment crystallisable (rFc) multimers primarily target Fcγ receptors (FcγRs) but may also affect the complement system. These include PF-06755347 (GL-2045), CSL730 (M230), CSL777 and Pan Fc Receptor Interacting Molecule (PRIM). Neonatal Fc receptor (FcRn)-targeting therapeutics block the FcRn receptor and are represented by candidate drugs such as the Fc fragment efgartigimod and the monoclonal antibodies rozanolixizumab (UCB7665), M281 and SYNT001. Finally, Fc and FcγR-targeting therapeutics, comprise molecules that target the Fc of IgG, such as the recombinant soluble FcγIIb receptor valziflocept (SM101/SHP652) and various monoclonal antibodies directed against the receptors. The developmental status of these three classes of molecules ranges from preclinical to ongoing phase 3 clinical studies. Efgartigimod and rozanolixizumab are the most advanced and have demonstrated encouraging results from phase 2 trials in immune thrombocytopenia and myasthenia gravis. Although initial results are promising, further long-term data and a better understanding of the unique mechanisms of action of the different molecules are needed. The efficacy, safety, convenience of administration, duration of effects, and cost will all contribute to determining which of the molecules will be successful in the clinic.


Asunto(s)
Enfermedades Autoinmunes/terapia , Productos Biológicos/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/inmunología , Terapia Molecular Dirigida , Receptores Fc/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Receptores Fc/inmunología
19.
Neurology ; 92(23): e2661-e2673, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31118245

RESUMEN

OBJECTIVE: To investigate safety and explore efficacy of efgartigimod (ARGX-113), an anti-neonatal Fc receptor immunoglobulin G1 Fc fragment, in patients with generalized myasthenia gravis (gMG) with a history of anti-acetylcholine receptor (AChR) autoantibodies, who were on stable standard-of-care myasthenia gravis (MG) treatment. METHODS: A phase 2, exploratory, randomized, double-blind, placebo-controlled, 15-center study is described. Eligible patients were randomly assigned (1:1) to receive 4 doses over a 3-week period of either 10 mg/kg IV efgartigimod or matched placebo combined with their standard-of-care therapy. Primary endpoints were safety and tolerability. Secondary endpoints included efficacy (change from baseline to week 11 of Myasthenia Gravis Activities of Daily Living, Quantitative Myasthenia Gravis, and Myasthenia Gravis Composite disease severity scores, and of the revised 15-item Myasthenia Gravis Quality of Life scale), pharmacokinetics, pharmacodynamics, and immunogenicity. RESULTS: Of the 35 screened patients, 24 were enrolled and randomized: 12 received efgartigimod and 12 placebo. Efgartigimod was well-tolerated in all patients, with no serious or severe adverse events reported, no relevant changes in vital signs or ECG findings observed, and no difference in adverse events between efgartigimod and placebo treatment. All patients treated with efgartigimod showed a rapid decrease in total immunoglobulin G (IgG) and anti-AChR autoantibody levels, and assessment using all 4 efficacy scales consistently demonstrated that 75% showed a rapid and long-lasting disease improvement. CONCLUSIONS: Efgartigimod was safe and well-tolerated. The correlation between reduction of levels of pathogenic IgG autoantibodies and disease improvement suggests that reducing pathogenic autoantibodies with efgartigimod may offer an innovative approach to treat MG. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that efgartigimod is safe and well-tolerated in patients with gMG.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Miastenia Gravis/tratamiento farmacológico , Receptores Fc/antagonistas & inhibidores , Actividades Cotidianas , Corticoesteroides/uso terapéutico , Adulto , Anciano , Autoanticuerpos/inmunología , Inhibidores de la Colinesterasa/uso terapéutico , Método Doble Ciego , Femenino , Antígenos de Histocompatibilidad Clase I , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología , Resultado del Tratamiento , Adulto Joven
20.
Exp Neurol ; 317: 133-143, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30851266

RESUMEN

Myasthenia gravis is hallmarked by fatigable muscle weakness resulting from neuromuscular synapse dysfunction caused by IgG autoantibodies. The variant with muscle-specific kinase (MuSK) autoantibodies is characterized by prominent cranial and bulbar weakness and a high frequency of respiratory crises. The majority of MuSK MG patients requires long-term immunosuppressive treatment, but the result of these treatments is considered less satisfactory than in MG with acetylcholine receptor antibodies. Emergency treatments are more frequently needed, and many patients develop permanent facial weakness and nasal speech. Therefore, new treatment options would be welcome. The neonatal Fc receptor protects IgG from lysosomal breakdown, thus prolonging IgG serum half-life. Neonatal Fc receptor antagonism lowers serum IgG levels and thus may act therapeutically in autoantibody-mediated disorders. In MuSK MG, IgG4 anti-MuSK titres closely correlate with disease severity. We therefore tested efgartigimod (ARGX-113), a new neonatal Fc receptor blocker, in a mouse model for MuSK myasthenia gravis. This model involves 11 daily injections of purified IgG4 from MuSK myasthenia gravis patients, resulting in overt myasthenic muscle weakness and, consequently, body weight loss. Daily treatment with 0.5 mg efgartigimod, starting at the fifth passive transfer day, reduced the human IgG4 titres about 8-fold, despite continued daily injection. In muscle strength and fatigability tests, efgartigimod-treated myasthenic mice outperformed control myasthenic mice. Electromyography in calf muscles at endpoint demonstrated less myasthenic decrement of compound muscle action potentials in efgartigimod-treated mice. These substantial in vivo improvements of efgartigimod-treated MuSK MG mice following a limited drug exposure period were paralleled by a tendency of recovery at neuromuscular synaptic level (in various muscles), as demonstrated by ex vivo functional studies. These synaptic improvements may well become more explicit upon longer drug exposure. In conclusion, our study shows that efgartigimod has clear therapeutic potential in MuSK myasthenia gravis and forms an exciting candidate drug for many autoantibody-mediated neurological and other disorders.


Asunto(s)
Debilidad Muscular/tratamiento farmacológico , Debilidad Muscular/genética , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/genética , Proteínas Tirosina Quinasas Receptoras/genética , Potenciales de Acción , Animales , Electromiografía , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/sangre , Técnicas In Vitro , Ratones , Ratones Endogámicos NOD , Ratones SCID , Placa Motora/efectos de los fármacos , Contracción Muscular , Debilidad Muscular/etiología , Miastenia Gravis Autoinmune Experimental/complicaciones , Receptores Fc/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...