Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.913
Filter
1.
Front Immunol ; 15: 1404191, 2024.
Article in English | MEDLINE | ID: mdl-38903526

ABSTRACT

Myasthenia Gravis (MG) is a chronic disabling autoimmune disease caused by autoantibodies to the neuromuscular junction (NMJ), characterized clinically by fluctuating weakness and early fatigability of ocular, skeletal and bulbar muscles. Despite being commonly considered a prototypic autoimmune disorder, MG is a complex and heterogeneous condition, presenting with variable clinical phenotypes, likely due to distinct pathophysiological settings related with different immunoreactivities, symptoms' distribution, disease severity, age at onset, thymic histopathology and response to therapies. Current treatment of MG based on international consensus guidelines allows to effectively control symptoms, but most patients do not reach complete stable remission and require life-long immunosuppressive (IS) therapies. Moreover, a proportion of them is refractory to conventional IS treatment, highlighting the need for more specific and tailored strategies. Precision medicine is a new frontier of medicine that promises to greatly increase therapeutic success in several diseases, including autoimmune conditions. In MG, B cell activation, antibody recycling and NMJ damage by the complement system are crucial mechanisms, and their targeting by innovative biological drugs has been proven to be effective and safe in clinical trials. The switch from conventional IS to novel precision medicine approaches based on these drugs could prospectively and significantly improve MG care. In this review, we provide an overview of key immunopathogenetic processes underlying MG, and discuss on emerging biological drugs targeting them. We also discuss on future direction of research to address the need for patients' stratification in endotypes according with genetic and molecular biomarkers for successful clinical decision making within precision medicine workflow.


Subject(s)
Autoantibodies , Myasthenia Gravis , Precision Medicine , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Myasthenia Gravis/drug therapy , Myasthenia Gravis/diagnosis , Autoantibodies/immunology , Autoimmunity , Animals , Immunosuppressive Agents/therapeutic use , Neuromuscular Junction/immunology
3.
J Manag Care Spec Pharm ; 30(6): 517-527, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38824625

ABSTRACT

BACKGROUND: Eculizumab and efgartigimod were approved to treat anti-acetylcholine receptor antibody-positive generalized myasthenia gravis (anti-AChR Ab-positive gMG). These relatively new biological treatments provide a more rapid onset of action and improved efficacy compared with conventional immunosuppressive treatments, but at a higher cost. OBJECTIVE: To assess the cost-effectiveness of eculizumab and, separately, efgartigimod, each added to conventional therapy vs conventional therapy alone, among patients with refractory anti-AChR Ab-positive gMG and those with anti-AChR Ab-positive gMG, respectively. METHODS: A Markov model with 4 health states was developed, evaluating costs and utility with a 4-week cycle length and lifetime time horizon from a health care system perspective and a modified societal perspective including productivity losses from patients and caregiver burden. Model inputs were informed by key clinical trials and relevant publications identified from targeted literature reviews, and drug costs were identified from Micromedex Red Book. Costs and outcomes were discounted at 3% per year. Incremental cost-effectiveness ratios (ICERs; cost per quality-adjusted life-year [QALY] gained) were calculated for each comparison. RESULTS: Among the corresponding populations, lifetime costs and QALYs, respectively, for eculizumab were $5,515,000 and 11.85, and for conventional therapy, $308,000 and 10.29, resulting in an ICER of $3,338,000/QALY gained. For efgartigimod, lifetime costs and QALYs, respectively, were $6,773,000 and 13.22, and for conventional therapy, $322,000 and 9.98, yielding an ICER of $1,987,000/QALY gained. After applying indirect costs in a modified societal perspective, the ICERs were reduced to $3,310,000/QALY gained for eculizumab and $1,959,000/QALY gained for efgartigimod. CONCLUSIONS: Eculizumab and efgartigimod are rapidly acting and effective treatments for myasthenia gravis. However, at their current price, both therapies greatly exceeded common cost-effectiveness thresholds, likely limiting patient access to these therapies.


Subject(s)
Antibodies, Monoclonal, Humanized , Cost-Benefit Analysis , Markov Chains , Myasthenia Gravis , Quality-Adjusted Life Years , Receptors, Cholinergic , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/economics , Myasthenia Gravis/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/economics , Receptors, Cholinergic/immunology , Female , Male , Middle Aged , Drug Costs , Adult , Autoantibodies
4.
CNS Neurosci Ther ; 30(6): e14793, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894580

ABSTRACT

BACKGROUND: We aimed to compare the efficacy of tocilizumab with conventional immunotherapy in refractory patients with acetylcholine receptor antibody-positive (AChR-Ab+) generalized myasthenia gravis (gMG). METHODS: This single-center prospective cohort study was based on patients from an MG registry study in China and conducted from February 10, 2021 to March 31, 2022. Adult refractory patients with AChR-Ab+ gMG were assigned to tocilizumab or conventional immunotherapy groups. The primary efficacy outcome was the mean difference of MG activities of daily living (MG-ADL) change at weeks 4, 8, 12, 16, 20, 24 corresponding to that at the baseline between the two groups. A generalized estimating equation model was used for the primary outcome analysis. Safety was assessed based on adverse events. RESULTS: Of 34 eligible patients, 20 (mean [standard deviation] age, 53.8 [21.9] years; 12 [60.0%] female) received tocilizumab and 14 received conventional immunotherapy (45.8 [18.0] years; 8 [57.1%] female). The tocilizumab group had greater reduction in MG-ADL score at week 4 (adjusted mean difference, -3.4; 95% CI, -4.7 to -2.0; p < 0.001) than the conventional immunotherapy group, with significant differences sustained through week 24 (adjusted mean difference, -4.5; 95% CI, -6.4 to -2.6; p < 0.001). At week 24, the proportion of patients achieving higher levels of MG-ADL (up to 7-point reduction) and QMG (up to 11-point reduction) scores improvement was significantly greater with tocilizumab. Tocilizumab had acceptable safety profiles without severe or unexpected safety issues. CONCLUSION: Tocilizumab is safe and effective in improving the MG-ADL score and reducing prednisone dose in refractory AChR-Ab+ gMG, suggesting tocilizumab has the potential to be a valuable therapeutic option for such patients.


Subject(s)
Antibodies, Monoclonal, Humanized , Myasthenia Gravis , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Female , Middle Aged , Male , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Adult , Aged , Prospective Studies , Treatment Outcome , Cohort Studies , Activities of Daily Living , Immunotherapy/methods , Immunotherapy/adverse effects , Registries
5.
J Neuroinflammation ; 21(1): 126, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734662

ABSTRACT

Myasthenia gravis (MG) is an immune-mediated disease frequently associated with thymic changes. Increased T helper 17 (Th17) cell activity and dysfunctional regulatory T (Treg) cells have been demonstrated in subgroups of MG. On the other hand, hypoxia-inducible factor 1 (HIF-1) has been shown to regulate the Th17/Treg balance by inducing Th17 differentiation while attenuating Treg development. To identify the underlying mechanisms of different thymic pathologies in MG development, we evaluated thymic samples from thymoma-associated myasthenia gravis (TAMG), MG with hyperplasia (TFH-MG) and thymoma without MG (TOMA) patients. Differential gene expression analysis revealed that TAMG and TFH-MG cells are associated with different functional pathways. A higher RORC/FOXP3 ratio provided evidence for Th17/Treg imbalance in TAMG potentially related to increased HIF1A. The hypoxic microenvironment in thymoma may be a driver of TAMG by increasing HIF1A. These findings may lead to new therapeutic approaches targeting HIF1A in the development of TAMG.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Myasthenia Gravis , Th17 Cells , Thymoma , Female , Humans , Male , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Myasthenia Gravis/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/metabolism , Th17 Cells/immunology , Thymoma/complications , Thymoma/genetics , Thymoma/immunology , Thymus Gland/pathology , Thymus Neoplasms/complications , Thymus Neoplasms/genetics
6.
J Neurol Sci ; 461: 123047, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759248

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) with MuSK antibodies (MuSK-MG) represents a distinct subtype with different responses to treatments compared to patients with AChR antibodies, especially in terms of tolerance to acetylcholinesterase inhibitors (AChEI). However, AChEI are often used as first line symptomatic treatment in MuSK-MG, despite reports that they are poorly tolerated, seldom effective or even deleterious. METHODS: We analyzed demographic, clinical and therapeutic responses and side-effects in the large cohort of 202 MuSK-MG patients cared for at the MG Clinic of Azienda Ospedaliero-Universitaria Pisana. RESULTS: 165 patients had received AChEI at first evaluation. Only 7/165 patients (4.2%) reported an initial clinical benefit. Conversely, 76.9% of patients reported at least one side effect, most commonly neuromuscular hyperexcitability (68.4%), gastrointestinal (53.9%) and neurovegetative (35.8%) disturbances. 56 (33.9%) patients reported a concomitant worsening of muscle weakness and twelve patients (7.3%) suffered a cholinergic crisis. According to these patients, the severity of cholinergic side effects was greater at higher doses of AChEI, but side effects occurred regardless of the dose administered and ceased once the drug was discontinued. CONCLUSIONS: This is the largest population of MuSK-MG patients reported for perceived responsiveness and tolerance to AChEI treatment. Our obervations strongly suggest avoiding this treatment in MuSK-MG.


Subject(s)
Autoantibodies , Cholinesterase Inhibitors , Myasthenia Gravis , Receptor Protein-Tyrosine Kinases , Receptors, Cholinergic , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Cholinesterase Inhibitors/therapeutic use , Male , Female , Middle Aged , Receptors, Cholinergic/immunology , Adult , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Aged , Autoantibodies/blood , Young Adult , Adolescent , Aged, 80 and over , Treatment Outcome , Cohort Studies
7.
Int Immunopharmacol ; 134: 112275, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759373

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) is an autoimmune disease mediated by pathogenic antibodies produced by abnormally activated B cells, resulting in neuromuscular junction transmission dysfunction. Interleukin-41 (IL-41) is a novel immunomodulatory cytokine that has been implicated in various metabolic, inflammatory, and autoimmune diseases. The role of IL-41 in MG is still unclear up to now, our study aimed to investigate the level of IL-41 in MG patients and its correlation with clinical features and inflammatory indicators. METHODS: Totally, 60 MG patients and 30 healthy controls (HC) were recruited. Baseline data and laboratory parameters were routinely recorded through electronic medical systems. IL-41 levels were measured by enzyme-linked immunosorbent assay. Proportions of T-cell and B-cell subsets and natural killer cells were analyzed by flow cytometry. The correlation between serum IL-41 and MG related parameters was investigated, and the clinical value of IL-41 in the diagnosis of MG was evaluated by receiver operator characteristic curve (ROC) analysis. RESULTS: Serum IL-41 levels in MG patients were higher than in HC, and were higher in Myasthenia Gravis Foundation of America (MGFA) III + IV group than that in MGFA I + II group. Serum IL-41 was positively correlated with MG-specific activities of daily living scale (MG-ADL), MGFA classification, platelet to lymphocyte ratio (PLR), and proportion of CD19+ B cells, while it was negatively correlated with high-sensitive C-reactive protein (hs-CRP) and circulatory plasma cells in MG patients. Serum IL-41 levels increased in patients who were treated with efgartigimod during the first cycle of therapy. However, compared to disease initiation, serum IL-41 levels decreased when clinical features steadily improved. ROC analysis showed that IL-41 had a diagnostic value for MG. CONCLUSION: The present findings suggested that serum IL-41 was increased in MG patients and was positively associated with the severity of the disease. IL-41 may be essential to the immunopathological mechanism of MG and a potential biomarker for MG.


Subject(s)
Biomarkers , Myasthenia Gravis , Severity of Illness Index , Humans , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Male , Female , Adult , Middle Aged , Biomarkers/blood , Interleukins/blood , Young Adult
8.
Front Immunol ; 15: 1325171, 2024.
Article in English | MEDLINE | ID: mdl-38715598

ABSTRACT

Introduction: Muscle-specific kinase (MuSK)- myasthenia gravis (MG) is caused by pathogenic autoantibodies against MuSK that correlate with disease severity and are predominantly of the IgG4 subclass. The first-line treatment for MuSK-MG is general immunosuppression with corticosteroids, but the effect of treatment on IgG4 and MuSK IgG4 levels has not been studied. Methods: We analyzed the clinical data and sera from 52 MuSK-MG patients (45 female, 7 male, median age 49 (range 17-79) years) from Italy, the Netherlands, Greece and Belgium, and 43 AChR-MG patients (22 female, 21 male, median age 63 (range 2-82) years) from Italy, receiving different types of immunosuppression, and sera from 46 age- and sex-matched non-disease controls (with no diagnosed diseases, 38 female, 8 male, median age 51.5 (range 20-68) years) from the Netherlands. We analyzed the disease severity (assessed by MGFA or QMG score), and measured concentrations of MuSK IgG4, MuSK IgG, total IgG4 and total IgG in the sera by ELISA, RIA and nephelometry. Results: We observed that MuSK-MG patients showed a robust clinical improvement and reduction of MuSK IgG after therapy, and that MuSK IgG4 concentrations, but not total IgG4 concentrations, correlated with clinical severity. MuSK IgG and MuSK IgG4 concentrations were reduced after immunosuppression in 4/5 individuals with before-after data, but data from non-linked patient samples showed no difference. Total serum IgG4 levels were within the normal range, with IgG4 levels above threshold (1.35g/L) in 1/52 MuSK-MG, 2/43 AChR-MG patients and 1/45 non-disease controls. MuSK-MG patients improved within the first four years after disease onset, but no further clinical improvement or reduction of MuSK IgG4 were observed four years later, and only 14/52 (26.92%) patients in total, of which 13 (93.3%) received general immunosuppression, reached clinical remission. Discussion: We conclude that MuSK-MG patients improve clinically with general immunosuppression but may require further treatment to reach remission. Longitudinal testing of individual patients may be clinically more useful than single measurements of MuSK IgG4. No significant differences in the serum IgG4 concentrations and IgG4/IgG ratio between AChR- and MuSK-MG patients were found during follow-up. Further studies with larger patient and control cohorts are necessary to validate the findings.


Subject(s)
Autoantibodies , Immunoglobulin G , Myasthenia Gravis , Receptor Protein-Tyrosine Kinases , Receptors, Cholinergic , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/blood , Myasthenia Gravis/diagnosis , Male , Middle Aged , Female , Adult , Aged , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Retrospective Studies , Young Adult , Adolescent , Autoantibodies/blood , Autoantibodies/immunology , Aged, 80 and over , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Severity of Illness Index , Child
9.
Neuron ; 112(11): 1757-1763.e2, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38697115

ABSTRACT

Myasthenia gravis (MG) and Lambert-Eaton myasthenic syndrome (LEMS) are autoimmune disorders affecting neuromuscular transmission. Their combined occurrence is rare, and treatment remains challenging. Two women diagnosed with concomitant MG/LEMS experienced severe, increasing disease activity despite multiple immunotherapies. Anti-CD19 chimeric antigen receptor (CAR) T cells have shown promise for treating autoimmune diseases. This report details the safe application of anti-CD19 CAR T cells for treating concomitant MG/LEMS. After CAR T cell therapy, both patients experienced rapid clinical recovery and regained full mobility. Deep B cell depletion and normalization of acetylcholine receptor and voltage-gated calcium channel N-type autoantibody levels paralleled major neurological responses. Within 2 months, both patients returned to everyday life, from wheelchair dependency to bicycling and mountain hiking, and remain stable at 6 and 4 months post-CAR T cell infusion, respectively. This report highlights the potential for anti-CD19 CAR T cells to achieve profound clinical effects in the treatment of neuroimmunological diseases.


Subject(s)
Antigens, CD19 , Immunotherapy, Adoptive , Lambert-Eaton Myasthenic Syndrome , Myasthenia Gravis , Humans , Female , Lambert-Eaton Myasthenic Syndrome/immunology , Lambert-Eaton Myasthenic Syndrome/therapy , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Middle Aged , T-Lymphocytes/immunology , Receptors, Chimeric Antigen/immunology , Adult , Treatment Outcome
10.
Autoimmunity ; 57(1): 2347379, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38723105

ABSTRACT

Thymoma is closely associated with myasthenia gravis (MG). However, due to the heterogeneity of thymoma and the intricate pathogenesis of MG, it remains unclear why some patients with thymoma develop MG and others do not. In this study, we conducted a comparative phenotype analysis of thymocytes in type B thymomas in patients with MG (MG (+) thymomas) and without MG (MG (-) thymomas) via fluorescence-activated cell sorting (FACS). Our results show that the developmental stages defined by the expression of CD3, CD4, and CD8 were largely maintained in both MG (+) and MG (-) thymomas, with CD4+CD8+ cells constituting the majority of thymocytes in type B thymoma, and no significant difference between this cell population was observed in MG (+) and MG (-) thymomas.We discovered that CD4+CD8+ thymocytes in MG (+) thymomas expressed low levels of αß TCR and high levels of IL-7 receptor α (IL-7Rα), whereas in MG (-) thymomas, CD4+CD8+ thymocytes exhibited the opposite pattern of αß TCR and IL-7Rα expression. These results suggest that the positive and negative selection processes of CD4+CD8+ thymocytes might differ between MG (+) thymomas and MG (-) thymomas. The expression of the Helios transcription factor is induced during negative selection and marks a group of T cells that have undergone negative selection and are likely to be deleted due to strong TCR binding with self-peptides/MHC ligands. We observed that the percentage of Helios-positive CD4SP T cells was greater in MG (-) than in MG (+) thymomas. Thus, the differentially regulated selection process of CD4+CD8+ thymocytes, which involves TCR and IL-7/IL-7Rα signaling, is associated with the presence of MG in type B thymomas.


Subject(s)
Myasthenia Gravis , Receptors, Antigen, T-Cell, alpha-beta , Thymocytes , Thymoma , Humans , Thymoma/immunology , Thymoma/pathology , Thymoma/metabolism , Myasthenia Gravis/immunology , Myasthenia Gravis/pathology , Myasthenia Gravis/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Male , Thymocytes/immunology , Thymocytes/metabolism , Female , Middle Aged , Receptors, Interleukin-7/metabolism , Receptors, Interleukin-7/immunology , Adult , Aged , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology , Thymus Neoplasms/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunophenotyping
11.
Eur J Neurol ; 31(7): e16306, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38716750

ABSTRACT

BACKGROUND AND PURPOSE: This study was undertaken to highlight neonatal Fc receptor inhibition (efgartigimod) as a valuable therapeutic option for patients with refractory seronegative myasthenia gravis (MG) and to emphasize the concept that seronegative MG is greatly constrained by the limitations of currently available diagnostic methods and therapeutic measures. METHODS: We describe the first refractory, generalized MG (gMG) patient successfully treated with efgartigimod after testing negative on standard autoantibody detection tests. RESULTS: Our patient presented with severe fluctuating bulbar and generalized weakness, resulting in multiple myasthenic crises requiring intubation. After a 28-year medical history of multiple failed lines of treatment, our patient was started on efgartigimod. Over five treatment cycles, a definite improvement in her clinical condition was observed (Myasthenia Gravis Foundation of America class: IIIb to IIb; MG-Activities of Daily Living score: 11 to 0; MG-Quality of Life 15 score: 30 to 0; Quantitative MG score: 28 to 6). Standard autoantibody detection tests failed to detect known pathogenic autoantibodies, but cell-based assay (CBA) identified autoantibodies against clustered adult acetylcholine receptor (AChR). CONCLUSIONS: In light of recent approvals of efgartigimod by the European Medicines Agency and US Food and Drug Administration exclusively for AChR-positive gMG forms, our case highlights evidence suggesting that such an approach might be shortsighted and could limit therapeutic options for patients with refractory seronegative gMG. Additionally, introducing more sensitive analytical techniques, exemplified by CBA, may help bridge the gap between seronegative and seropositive patients. This represents an urgent unmet need for gMG patients, as the antibody profile dramatically influences the therapeutic approach.


Subject(s)
Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Female , Autoantibodies/blood , Receptors, Fc/therapeutic use , Adult , Receptors, Cholinergic/immunology , Middle Aged
12.
Brain Nerve ; 76(5): 613-620, 2024 May.
Article in Japanese | MEDLINE | ID: mdl-38741504

ABSTRACT

Herein, we describe the mechanisms, diagnostic procedures, and treatment options for acetylcholine receptor (AChR) antibody-positive myasthenia gravis (MG). The upstream pathomechanism of this condition involves AChR-sensitized T cell-dependent B cell proliferation and the subsequent production of pathogenic autoantibodies. Downstream molecules include AChR antibodies that activate complement pathways, resulting in the destruction of motor endplates. We further introduce newly-developed molecular targeted drugs for the treatment of MG that aims to secure patients' health-related quality of life.


Subject(s)
Autoantibodies , Myasthenia Gravis , Receptors, Cholinergic , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Myasthenia Gravis/therapy , Humans , Receptors, Cholinergic/immunology , Autoantibodies/immunology
13.
Brain Nerve ; 76(5): 623-629, 2024 May.
Article in Japanese | MEDLINE | ID: mdl-38741505

ABSTRACT

Reportedly, patients with muscle-specific kinase (MuSK) antibody-positive myasthenia gravis (MG) account for approximately 3.0% of all patients with MG in Japan. Compared with patients who have acetylcholine receptor antibody-positive MG, those with MuSK antibody-positive MG show young-onset disease with female predominance, a low rate of ocular involvement (5.9%), and greater severity of dysphagia. The aforementioned types of MG are indistinguishable based on clinical symptoms and electrophysiological tests, and measurement of MuSK antibodies is essential for diagnosis. Thymectomy and complement inhibitors are not indicated for treatment, and acetylcholinesterase inhibitors, steroids, immunosuppressants, plasma exchange, intravenous immunoglobulin therapy, and neonatal Fc receptor inhibitors are used.


Subject(s)
Autoantibodies , Myasthenia Gravis , Receptor Protein-Tyrosine Kinases , Receptors, Cholinergic , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Myasthenia Gravis/therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Autoantibodies/immunology
14.
Front Immunol ; 15: 1382320, 2024.
Article in English | MEDLINE | ID: mdl-38711503

ABSTRACT

Myasthenia gravis (MG), primarily caused by acetylcholine receptor (AChR) autoantibodies, is a chronic autoimmune disorder causing severe muscle weakness and fatigability. In particular, seronegative MG constitutes 10%-15% of MG cases and presents diagnostic challenges especially in early-onset female patients who often show severe disease and resistance to immunosuppressive therapy. Furthermore, the immunopathology of seronegative MG remains unclear. Thus, in this study, we aimed to elucidate the pathogenic mechanism of seronegative MG using scRNA-seq analysis and plasma proteome analysis; in particular, we investigated the relationship between immune dysregulation status and disease severity in refractory seronegative MG. Employing single-cell RNA-sequencing and plasma proteome analyses, we analyzed peripheral blood samples from 30 women divided into three groups: 10 healthy controls, 10 early-onset AChR-positive MG, and 10 refractory early-onset seronegative MG patients, both before and after intravenous immunoglobulin treatment. The disease severity was evaluated using the MG-Activities of Daily Living (ADL), MG composite (MGC), and revised 15-item MG-Quality of Life (QOL) scales. We observed numerical abnormalities in multiple immune cells, particularly B cells, in patients with refractory seronegative MG, correlating with disease activity. Notably, severe MG cases had fewer regulatory T cells without functional abnormalities. Memory B cells were found to be enriched in peripheral blood cells compared with naïve B cells. Moreover, plasma proteome analysis indicated significantly lower plasma protein levels of soluble CD22, expressed in the lineage of B-cell maturation (including mature B cells and memory B cells), in refractory seronegative MG patients than in healthy donors or patients with AChR-positive MG. Soluble CD22 levels were correlated with disease severity, B-cell frequency, and RNA expression levels of CD22. In summary, this study elucidates the immunopathology of refractory seronegative MG, highlighting immune disorders centered on B cells and diminished soluble CD22 levels. These insights pave the way for novel MG treatment strategies focused on B-cell biology.


Subject(s)
B-Lymphocytes , Myasthenia Gravis , Sialic Acid Binding Ig-like Lectin 2 , Humans , Myasthenia Gravis/immunology , Myasthenia Gravis/blood , Female , Adult , B-Lymphocytes/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Middle Aged , Autoantibodies/blood , Autoantibodies/immunology , Immunoglobulins, Intravenous/therapeutic use , Receptors, Cholinergic/immunology , Severity of Illness Index , Young Adult , Proteome
16.
Eur J Neurosci ; 59(12): 3292-3308, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38650308

ABSTRACT

Muscle-specific kinase myasthenia gravis (MuSK MG) is caused by autoantibodies against MuSK in the neuromuscular junction (NMJ). MuSK MG patients have fluctuating, fatigable skeletal muscle weakness, in particular of bulbar muscles. Severity differs greatly between patients, in spite of comparable autoantibody levels. One explanation for inter-patient and inter-muscle variability in sensitivity might be variations in compensatory muscle responses. Previously, we developed a passive transfer mouse model for MuSK MG. In preliminary ex vivo experiments, we observed that muscle contraction of some mice, in particular those with milder myasthenia, had become partially insensitive to inhibition by µ-Conotoxin-GIIIB, a blocker of skeletal muscle NaV1.4 voltage-gated sodium channels. We hypothesised that changes in NaV channel expression profile, possibly co-expression of (µ-Conotoxin-GIIIB insensitive) NaV1.5 type channels, might lower the muscle fibre's firing threshold and facilitate neuromuscular synaptic transmission. To test this hypothesis, we here performed passive transfer in immuno-compromised mice, using 'high', 'intermediate' and 'low' dosing regimens of purified MuSK MG patient IgG4. We compared myasthenia levels, µ-Conotoxin-GIIIB resistance and muscle fibre action potential characteristics and firing thresholds. High- and intermediate-dosed mice showed clear, progressive myasthenia, not seen in low-dosed animals. However, diaphragm NMJ electrophysiology demonstrated almost equal myasthenic severities amongst all regimens. Nonetheless, low-dosed mouse diaphragms showed a much higher degree of µ-Conotoxin-GIIIB resistance. This was not explained by upregulation of Scn5a (the NaV1.5 gene), lowered muscle fibre firing thresholds or histologically detectable upregulated NaV1.5 channels. It remains to be established which factors are responsible for the observed µ-Conotoxin-GIIIB insensitivity and whether the NaV repertoire change is compensatory beneficial or a bystander effect.


Subject(s)
Muscle, Skeletal , Animals , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Humans , Myasthenia Gravis/metabolism , Myasthenia Gravis/physiopathology , Myasthenia Gravis/immunology , Disease Models, Animal , Female , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/immunology , Voltage-Gated Sodium Channels/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/drug effects , Autoantibodies , Male , Conotoxins/pharmacology , Immunization, Passive
17.
J Neuromuscul Dis ; 11(3): 613-623, 2024.
Article in English | MEDLINE | ID: mdl-38578899

ABSTRACT

Background/Objective: Myasthenia Gravis (MG) is an autoimmune disorder characterized by pathogenic autoantibodies (AAbs) targeting nicotinic acetylcholine receptors (AChR), disrupting neuromuscular communication. RadioImmunoPrecipitation Assay (RIPA) is recommended to detect AChR AAbs, but its complexity and radioactive requirements limit widespread use. We compare non-RIPA anti-AChR immunoassays, including Cell-Based Assay (CBA) and two ELISA kits, against the gold standard RIPA. Methods/Results: 145 samples were included with medical indication for anti-AChR testing. By the RIPA method, 63 were negative (RIPA-Neg < 0.02 nmol/L), 18 were classified as Borderline (≥0.02 -1 nmol/L), and 64 were positive (RIPA-Pos > 1 nmol/L). The competitive ELISA showed poor agreement with RIPA (Kappa = 0.216). The indirect ELISA demonstrated substantial agreement with RIPA (Kappa = 0.652), with ∼76% sensitivity and ∼94% specificity for MG diagnostic. The CBA, where fixed cells expressing clustered AChR were used as substrate, exhibited almost perfect agreement with RIPA (Kappa = 0.984), yielding ∼98% sensitivity and 96% specificity for MG. In addition, a semiquantitative analysis showed a strong correlation between CBA titration, indirect ELISA, and RIPA levels (r = 0.793 and r = 0.789, respectively). Conclusions: The CBA displayed excellent analytical performance for MG diagnostic when compared to RIPA, making it a potential replacement for RIPA in clinical laboratories. Some solid-phase assays (such as the indirect ELISA applied here), as well as CBA titration, offer reliable options to estimate anti-AChR AAb levels after confirming positivity by the CBA.∥.


Subject(s)
Autoantibodies , Enzyme-Linked Immunosorbent Assay , Myasthenia Gravis , Radioimmunoprecipitation Assay , Humans , Enzyme-Linked Immunosorbent Assay/methods , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Sensitivity and Specificity , Receptors, Cholinergic/immunology , Female , Male , Middle Aged , Adult , Aged , Young Adult
18.
Rev Med Suisse ; 20(871): 848-851, 2024 Apr 24.
Article in French | MEDLINE | ID: mdl-38665106

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by fluctuating weakness of skeletal muscles. Despite current treatments, a significant percentage of patients remain symptomatic. This review explores new immunosuppressive therapies and ongoing clinical trials in MG, including depletion of B lymphocytes with agents such as rituximab and inebilizumab, as well as the use of eculizumab, efgartigimod, satralizumab, tocilizumab, and CAR-T (Chimeric Antigen Receptor-T) cell therapy. These advancements aim to improve disease control and patients' quality of life.


La myasthénie grave (MG) est une maladie auto-immune caractérisée par une faiblesse fluctuante des muscles squelettiques. Malgré les traitements classiques, un pourcentage significatif de patients reste symptomatique. Cet article explore les nouvelles thérapies immunosuppressives et les essais cliniques en cours pour la MG, notamment la déplétion des lymphocytes B avec des agents tels que le rituximab et l'inébilizumab, ainsi que l'utilisation de l'éculizumab, de l'efgartigimod, du satralizumab, du tocilizumab et de la thérapie par cellules CAR-T (Chimeric Antigen Receptor-T). Ces avancées visent à améliorer le contrôle de la maladie et la qualité de vie des patients.


Subject(s)
Myasthenia Gravis , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Myasthenia Gravis/drug therapy , Humans , Immunosuppressive Agents/therapeutic use , Quality of Life , Immunomodulating Agents/therapeutic use , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , Immunologic Factors/therapeutic use
19.
Rinsho Shinkeigaku ; 64(4): 292-295, 2024 Apr 24.
Article in Japanese | MEDLINE | ID: mdl-38508730

ABSTRACT

A 79-year-old woman who presented ptosis and dysphagia were admitted to our hospital. Anti-acetylcholine receptor antibodies and anti-P/Q-type VGCC antibodies were both positive. Electrophysiological examination showed postsynaptic pattern which supported myasthenia gravis. She did not meet the diagnostic criteria for Lambert-Eaton myasthenic syndrome (LEMS). In cases which these antibodies coexist, careful electrophysiological evaluation is required for the diagnosis. In addition, although anti-P/Q-type VGCC antibodies have been specific to LEMS, patients with these antibodies represent various symptoms other than LEMS. Low and middle titer of the antibodies may be not specific to LEMS.


Subject(s)
Autoantibodies , Myasthenia Gravis , Receptors, Cholinergic , Humans , Female , Myasthenia Gravis/immunology , Myasthenia Gravis/diagnosis , Myasthenia Gravis/complications , Aged , Autoantibodies/blood , Receptors, Cholinergic/immunology , Calcium Channels, Q-Type/immunology , Calcium Channels, P-Type/immunology , Lambert-Eaton Myasthenic Syndrome/immunology , Lambert-Eaton Myasthenic Syndrome/diagnosis , Lambert-Eaton Myasthenic Syndrome/complications
20.
Expert Opin Pharmacother ; 25(4): 395-408, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38523508

ABSTRACT

INTRODUCTION: Myasthenia gravis (MG) is an autoimmune condition targeting the neuromuscular junction, which manifests with neuromuscular symptoms of varying severity and significant morbidity. The mainstay of treatment in MG is mitigation of the immune cascade with steroids and non-steroidal immunosuppressive therapies. The therapeutic strategies in MG are transitioning from broad and indiscriminate immunosuppression to novel agents targeting key steps in MG pathogenesis, including T cell activation, B cell proliferation, complement activation, maintenance of pathogenic antibody production, and proinflammatory cytokine production. AREAS COVERED: In this review, an overview of the pathogenesis of MG and traditional MG therapies is presented, followed by a discussion of the novel MG drugs that have been evaluated in phase 3 clinical trials with an emphasis on those which have received regulatory approval. EXPERT OPINION: Novel MG therapeutics belonging to the classes of complement inhibitors, neonatal Fc receptor (FcRn) inhibitors and B cell depletors, as well as the other emerging MG drugs in the pipeline constitute promising treatment strategies with potentially better efficacy and safety compared to the conventional MG treatments. However, further long-term research is needed in order to optimize the implementation of these new treatment options for the appropriate patient populations.


Subject(s)
Immunosuppressive Agents , Myasthenia Gravis , Humans , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Immunosuppressive Agents/therapeutic use , Animals , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , Complement Inactivating Agents/therapeutic use , Complement Inactivating Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...