1
|
Tard C, Laforet P, de Pouvourville G, Crochard A, Chollet G, Nevoret C, Bouée S, Salort-Campana E. Treatment of myasthenia gravis in france: A retrospective claims database study (STAMINA). J Neurol 2024; 271:7239-7249. [PMID: 39387949 PMCID: PMC11561051 DOI: 10.1007/s00415-024-12714-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND This study aimed to describe treatment patterns in patients with myasthenia gravis (MG) in France. METHODS A retrospective cohort analysis was performed using the French National Health Data System (SNDS) database between 2008 and 2019. MG patients were identified using ICD-10 codes during hospitalization and/or long-term disease. We defined two adult subpopulations: a prevalent MG population of patients alive on 31/12/2019 and an incident population of newly identified patients with MG in 2012 and 2013. RESULTS Among the 22,079 prevalent patients, 53.1% (n = 11,498) received at least one chronic MG treatment in 2019. Among these treated patients, 52.5% received Acetylcholinesterase inhibitors (AChEIs) only, 10.2% were treated with corticosteroids (CS) ± AChEIs, 7.3% with non-steroidal immunosuppressive treatments (NSIST) and CS, 24.2% with NSIST w/o CS, and 5.8% received immunoglobulin and/or plasma exchange. Among the 2,661 incident patients, 84.6% received at least one chronic MG treatment over the 6-year follow-up period, and among them, 79.0% had at least one treatment category change. During the first semester of follow-up, 28.1% of patients were treated with an immunomodulator (CS, NSIST). Among patients starting treatment with immunomodulator, the proportion of those treated with CS decreased from 35.3% at initiation to 10.9% at 6 years. CONCLUSION This study illustrates the complexity of MG management. Significant CS sparing was observed over time. The frequent treatment changes especially in patients with an immunomodulator treatment reflect the high variability of the disease severity. The need for personalised treatment approaches in the management of MG to reduce the burden of disease remains.
Collapse
Affiliation(s)
- C Tard
- Service de Neurologie, U1172, Centre de Référence Des Maladies Neuromusculaires Nord/Est/Ile-de-France, CHU de Lille, Lille, France
| | - P Laforet
- Service de Neurologie, Hôpital Raymond Poincaré, Centre de Référence des Maladies Neuromusculaires Nord-Est-Ile de France, FHU Phenix, Assistance-Publique Hôpitaux de Paris, Paris, Garches, France
- Université Paris-Saclay, UVSQ, INSERM U1179, Versailles, France
| | | | - A Crochard
- UCB Pharma, 420, Rue d'Estienne d'Orves-Immeuble Défense Ouest, La Défense, 92700, Colombes, France.
| | - G Chollet
- UCB Pharma, 420, Rue d'Estienne d'Orves-Immeuble Défense Ouest, La Défense, 92700, Colombes, France
| | | | - S Bouée
- CEMKA, Bourg-La-Reine, France
| | - E Salort-Campana
- Centre de Référence Des Maladies Neuromusculaires PACA Réunion Rhône Alpes, APHM, Service du Pr Attarian, Marseille, France
| |
Collapse
|
2
|
Remijn-Nelissen L, Tannemaat MR, Ruiter AM, Campman YJM, Verschuuren JJGM. Efgartigimod in refractory autoimmune myasthenia gravis. Muscle Nerve 2024; 70:325-332. [PMID: 38899431 DOI: 10.1002/mus.28184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION/AIMS Efgartigimod, a neonatal Fc-receptor inhibitor, has recently been approved as treatment for myasthenia gravis (MG). In this retrospective cohort study, we aimed to systematically assess short- and long-term effectiveness of efgartigimod in patients with refractory MG. METHODS Sixteen patients with refractory autoimmune acetylcholine receptor MG were treated with efgartigimod. Data were collected from January 2021 to March 2023 on Myasthenia Gravis Activities of Daily Living (MG-ADL), Quantitative Myasthenia Gravis score (QMG), Myasthenia Gravis Composite score (MGC) and the 15-item revised version of the Myasthenia Gravis Quality of Life questionnaire (MG-QoL15r). RESULTS A favorable outcome was seen in 56% of patients at the last measurement. Out of 16 patients, 50% were an MG-ADL responder after the first treatment cycle. After 4 weeks, a clinically meaningful improvement compared to baseline was seen on the MG-ADL, QMG, and MGC. There was a statistically significant improvement on the MGQoL15r from baseline to week 4. The improvement was maintained until the last measurement for the MGC and the MGQoL15r. At the last visit, all patients had discontinued 4-weekly dosages, shifting to administration frequencies of 1, 2, or 3 weeks. Drug doses could be decreased for prednisolone (n = 7), azathioprine (n = 2), and intravenous immunoglobulin (n = 9). Frequency of plasma exchange was decreased in nine patients. DISCUSSION In patients with refractory MG, efgartigimod was effective for at least half of all patients. Patients required more frequent dosing compared to the ADAPT phase 3 trial. In 80% of the patients concurrent medication could be reduced or discontinued.
Collapse
Affiliation(s)
| | - Martijn R Tannemaat
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Annabel M Ruiter
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvonne J M Campman
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
3
|
Querol L, De Sèze J, Dysgaard T, Levine T, Rao TH, Rivner M, Hartung HP, Kiessling P, Shimizu S, Marmol D, Bozorg A, Colson AO, Massow U, Eftimov F. Efficacy, safety and tolerability of rozanolixizumab in patients with chronic inflammatory demyelinating polyradiculoneuropathy: a randomised, subject-blind, investigator-blind, placebo-controlled, phase 2a trial and open-label extension study. J Neurol Neurosurg Psychiatry 2024; 95:845-854. [PMID: 38729747 PMCID: PMC11347201 DOI: 10.1136/jnnp-2023-333112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/13/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a peripheral nerve disorder characterised by weakness and sensory loss. We assessed the neonatal Fc receptor inhibitor rozanolixizumab for CIDP management. METHODS CIDP01 (NCT03861481) was a randomised, subject-blind, investigator-blind, placebo-controlled, phase 2a study. Adults with definite or probable CIDP receiving subcutaneous or intravenous immunoglobulin maintenance therapy were randomised 1:1 to 12 once-weekly subcutaneous infusions of rozanolixizumab 10 mg/kg or placebo, stratified according to previous immunoglobulin administration route. Investigators administering treatment and assessing efficacy, and patients, were blinded. The primary outcome was a change from baseline (CFB) to day 85 in inflammatory Rasch-built Overall Disability Scale (iRODS) score. Eligible patients who completed CIDP01 entered the open-label extension CIDP04 (NCT04051944). RESULTS In CIDP01, between 26 March 2019 and 31 March 2021, 34 patients were randomised to rozanolixizumab or placebo (17 (50%) each). No significant difference in CFB to day 85 in iRODS centile score was observed between rozanolixizumab (least squares mean 2.0 (SE 3.2)) and placebo (3.4 (2.6); difference -1.5 (90% CI -7.5 to 4.5)). Overall, 14 (82%) patients receiving rozanolixizumab and 13 (76%) receiving placebo experienced a treatment-emergent adverse event during the treatment period. Across CIDP01 and CIDP04, rozanolixizumab was well tolerated over up to 614 days; no clinically meaningful efficacy results were seen. No deaths occurred. CONCLUSIONS Rozanolixizumab did not show efficacy in patients with CIDP in this study, although this could be due to a relatively high placebo stability rate. Rozanolixizumab was well tolerated over medium-to-long-term weekly use, with an acceptable safety profile.
Collapse
Affiliation(s)
- Luis Querol
- Neuromuscular Diseases Unit - Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Barcelona, Spain
- Center for Network Research in Rare Diseases - CIBERER, Madrid, Spain
| | - Jérôme De Sèze
- Department of Neurology, Clinical Investigation Centre, University Hospital of Strasbourg, Strasbourg, France
| | - Tina Dysgaard
- Department of Neurology, Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Todd Levine
- Honor Health Neurology, Bob Bové Neuroscience Institute, Scottsdale, Arizona, USA
| | - T Hemanth Rao
- The Neurological Institute, PA, Charlotte, North Carolina, USA
| | - Michael Rivner
- Department of Neurology, Augusta University, Augusta, Atlanta, Georgia, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Brain and Mind Center, Medical Faculty, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Palacký University, Olomouc, Czech Republic
| | | | | | | | - Ali Bozorg
- UCB Pharma, Morrisville, North Carolina, USA
| | | | | | - Filip Eftimov
- Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Fodil S, Urbina T, Bredin S, Mayaux J, Lafarge A, Missri L, Maury E, Demoule A, Pene F, Mariotte E, Ait-Oufella H. Bloodstream infections among patients receiving therapeutic plasma exchanges in the intensive care unit: a 10 year multicentric study. Ann Intensive Care 2024; 14:117. [PMID: 39073720 PMCID: PMC11286886 DOI: 10.1186/s13613-024-01346-7] [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: 02/05/2024] [Accepted: 06/29/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Therapeutic plasma exchanges (TPE), which affect the humoral response, are often performed in combination with immunosuppressive drugs. For this reason, TPE may be associated with an increased susceptibility to infections. We aimed to describe blood stream infection (BSI) incidence in ICU patients treated with TPE and to identify associated risk factors. METHODS We retrospectively included patients that had received at least one session of TPE in the ICU of one of the 4 participating centers (all in Paris, France) between January 1st 2010 and December 31th 2019. Patients presenting with a BSI during ICU stay were compared to patients without such an infection. Risk factors for BSI were identified by a multivariate logistic regression model. RESULTS Over 10 years in the 4 ICUs, 387 patients were included, with a median of 5 [2-7] TPE sessions per patient. Most frequent indications for TPE were thrombotic microangiopathy (47%), central nervous system inflammatory disorders (11%), hyperviscosity syndrome (11%) and ANCA associated vasculitis (8.5%). Thirty-one patients (8%) presented with a BSI during their ICU stay, a median of 7 [3-11] days after start of TPE. In a multivariate logistic regression model, diabetes (OR 3.32 [1.21-8.32]) and total number of TPE sessions (OR 1.14 [1.08-1.20]) were independent risk factors for BSI. There was no difference between TPE catheter infection related BSI (n = 11 (35%)) and other sources of BSI (n = 20 (65%)) regarding catheter insertion site (p = 0.458) or rate of TPE catheter related deep vein thrombosis (p = 0.601). ICU course was severe in patients presenting with BSI when compared to patients without BSI, with higher need for mechanical ventilation (45% vs 18%, p = 0.001), renal replacement therapy (42% vs 20%, p = 0.011), vasopressors (32% vs 12%, p = 0.004) and a higher mortality (19% vs 5%, p = 0.010). CONCLUSION Blood stream infections are frequent in patients receiving TPE in the ICU, and are associated with a severe ICU course. Vigilant monitoring is crucial particularly for patients receiving a high number of TPE sessions.
Collapse
Affiliation(s)
- Sofiane Fodil
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France
| | - Tomas Urbina
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France
| | - Swann Bredin
- Service de Médecine Intensive-Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Julien Mayaux
- Service de Médecine Intensive-Réanimation, Groupe Hospitalier Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Lafarge
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France
| | - Louaï Missri
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France
| | - Eric Maury
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France
| | - Alexandre Demoule
- Service de Médecine Intensive-Réanimation, Groupe Hospitalier Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Frederic Pene
- Service de Médecine Intensive-Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric Mariotte
- Service de Médecine Intensive-Réanimation, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hafid Ait-Oufella
- Service de Médecine Intensive-Réanimation, Réanimation Médicale, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 184 rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France.
- Inserm U970, Cardiovascular Research Center, Université Paris-Cité, Paris, France.
| |
Collapse
|
5
|
Yurtsever N, Binns TC, Hendrickson JE, Tormey CA, Lee ES. Therapeutic plasma exchange for hyperviscosity syndrome in IgA multiple myeloma. Lab Med 2024:lmae054. [PMID: 39038224 DOI: 10.1093/labmed/lmae054] [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: 07/24/2024] Open
Abstract
Hyperviscosity syndrome (HVS) is defined as the symptomatic presentation of increased blood thickness due to various clinical conditions such as hypergammaglobulinemia. HVS secondary to immunoglobulin (Ig)A multiple myeloma has been infrequently reported. Although the efficiency of IgM or IgG removal by therapeutic plasma exchange (TPE) is well described, the efficiency of IgA removal by TPE is not as well known. Here, we describe a case of HVS due to IgA myeloma in a patient who received 2 TPE treatments, with subsequent symptomatic improvement as well as decrease in IgA and viscosity levels.
Collapse
Affiliation(s)
- Nalan Yurtsever
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, US
| | - Thomas C Binns
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, US
| | - Jeanne E Hendrickson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, US
| | | | - Edward S Lee
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, US
| |
Collapse
|
6
|
Jarius S, Ringelstein M, Schanda K, Ruprecht K, Korporal-Kuhnke M, Viehöver A, Hümmert MW, Schindler P, Endmayr V, Gastaldi M, Trebst C, Franciotta D, Aktas O, Höftberger R, Haas J, Komorowski L, Paul F, Reindl M, Wildemann B. Improving the sensitivity of myelin oligodendrocyte glycoprotein-antibody testing: exclusive or predominant MOG-IgG3 seropositivity-a potential diagnostic pitfall in patients with MOG-EM/MOGAD. J Neurol 2024; 271:4660-4671. [PMID: 38609667 PMCID: PMC11233316 DOI: 10.1007/s00415-024-12285-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Myelin oligodendrocyte glycoprotein antibody-associated encephalomyelitis (MOG-EM; also termed MOG antibody-associated disease, MOGAD) is the most important differential diagnosis of both multiple sclerosis and neuromyelitis optica spectrum disorders. A recent proposal for new diagnostic criteria for MOG-EM/MOGAD explicitly recommends the use of immunoglobulin G subclass 1 (IgG1)- or IgG crystallizable fragment (Fc) region-specific assays and allows the use of heavy-and-light-chain-(H+L) specific assays for detecting MOG-IgG. By contrast, the utility of MOG-IgG3-specific testing has not been systematically evaluated. OBJECTIVE To assess whether the use of MOG-IgG3-specific testing can improve the sensitivity of MOG-IgG testing. METHODS Re-testing of 22 patients with a definite diagnosis of MOG-EM/MOGAD and clearly positive MOG-IgG status initially but negative or equivocal results in H+L- or Fc-specific routine assays later in the disease course (i.e. patients with spontaneous or treatment-driven seroreversion). RESULTS In accordance with previous studies that had used MOG-IgG1-specific assays, IgG subclass-specific testing yielded a higher sensitivity than testing by non-subclass-specific assays. Using subclass-specific secondary antibodies, 26/27 supposedly seroreverted samples were still clearly positive for MOG-IgG, with MOG-IgG1 being the most frequently detected subclass (25/27 [93%] samples). However, also MOG-IgG3 was detected in 14/27 (52%) samples (from 12/22 [55%] patients). Most strikingly, MOG-IgG3 was the predominant subclass in 8/27 (30%) samples (from 7/22 [32%] patients), with no unequivocal MOG-IgG1 signal in 2 and only a very weak concomitant MOG-IgG1 signal in the other six samples. By contrast, no significant MOG-IgG3 reactivity was seen in 60 control samples (from 42 healthy individuals and 18 patients with MS). Of note, MOG-IgG3 was also detected in the only patient in our cohort previously diagnosed with MOG-IgA+/IgG- MOG-EM/MOGAD, a recently described new disease subvariant. MOG-IgA and MOG-IgM were negative in all other patients tested. CONCLUSIONS In some patients with MOG-EM/MOGAD, MOG-IgG is either exclusively or predominantly MOG-IgG3. Thus, the use of IgG1-specific assays might only partly overcome the current limitations of MOG-IgG testing and-just like H+L- and Fcγ-specific testing-might overlook some genuinely seropositive patients. This would have potentially significant consequences for the management of patients with MOG-EM/MOGAD. Given that IgG3 chiefly detects proteins and is a strong activator of complement and other effector mechanisms, MOG-IgG3 may be involved in the immunopathogenesis of MOG-EM/MOGAD. Studies on the frequency and dynamics as well as the clinical and therapeutic significance of MOG-IgG3 seropositivity are warranted.
Collapse
Affiliation(s)
- S Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | - M Ringelstein
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
| | - K Schanda
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - K Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - M Korporal-Kuhnke
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - A Viehöver
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - M W Hümmert
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - P Schindler
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - V Endmayr
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - M Gastaldi
- Neuroimmunology Laboratory and Neuroimmunology Research Unit, IRCCS Mondino Foundation National Neurological Institute, Pavia, Italy
| | - C Trebst
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - D Franciotta
- Neuroimmunology Laboratory and Neuroimmunology Research Unit, IRCCS Mondino Foundation National Neurological Institute, Pavia, Italy
| | - O Aktas
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
| | - R Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - J Haas
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - L Komorowski
- Institute of Experimental Neuroimmunology, affiliated to Euroimmun AG, Lübeck, Germany
| | - F Paul
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - M Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - B Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
7
|
Pambrun E, Loubet P, Fourneron T, Moranne O. Immunogenicity of SARS-CoV-2 vaccines in patients treated with chronic double filtration plasmapheresis. J Clin Apher 2024; 39:e22136. [PMID: 38923591 DOI: 10.1002/jca.22136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND The impact of chronic therapeutic plasmapheresis on humoral response following COVID-19 vaccination is poorly documented, especially among patients treated with double filtration plasmapheresis (DFPP). OBJECTIVES This retrospective single-center study evaluated the humoral response after SARS-CoV-2 vaccination and studied anti-SPIKE seropositivity and antibody dynamics in patients with chronic DFPP at our institution. METHOD All patients undergoing chronic DFPP at a tertiary center in France from December 2020 to November 2022 were included. We defined one patient subgroup as Group 1 to evaluate anti-SPIKE seropositivity after vaccination, with three groups based on their anti-SPIKE titers: (Group 1A) nonresponders (<0.8 UI/mL), (Group 1B) weak responders (0.8 to <250 binding antibody unit [BAU]/mL), and (Group 1C) strong responders (>250 BAU/mL). Group 2 served to evaluate antibody dynamics with anti-SPIKE levels measured 3 months after initial vaccination, Group 2A having a sustained level and Group 2B a declining pattern. RESULTS The 21 patients included had a median age of 63 years, and 13 (56%) were male. The indications for chronic DFPP mainly included dysimmune pathologies (15; 71%) and familial dyslipidemia (6; 29%). For the humoral response to vaccination in Patient Group 1, the only nonresponder was a patient who had undergone kidney transplantation 30 months earlier and was on immunosuppressive medication. For Patient Group 2, the median follow-up of antibody titers was 13 months [12-13]. Two distinct patterns of anti-SPIKE dynamics were observed: a rapid decline in anti-SPIKE antibody titers within 6 months following the initial vaccination or booster dose (n = 10 [71.4%] Group 2A) and stable anti-SPIKE levels above 250 BAU/mL over >6 months (n = 4 [28.6%] Group 2B) with more patients with familial dyslipidemia in the former. CONCLUSIONS Humoral response to SARS-CoV-2 vaccination appears robust in patients undergoing chronic DFPP and may be linked to patients' immune status rather than DFPTP itself. Our results support current recommendations for administering three doses of vaccine with a booster every 6 months.
Collapse
Affiliation(s)
- Emilie Pambrun
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Department of Infectious Disease, Nîmes University Hospital, Nîmes, France
| | - Thomas Fourneron
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
| | - Olivier Moranne
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
- IDESP, UMR-INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
8
|
Chen J, Feng L, Li S, Wang H, Huang X, Shen C, Feng H. Therapeutic Plasma Exchange in AChR-Ab Positive Generalized Myasthenia Gravis: A Real World Study About Its Early Response. J Inflamm Res 2024; 17:2299-2308. [PMID: 38645879 PMCID: PMC11032135 DOI: 10.2147/jir.s455104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/06/2024] [Indexed: 04/23/2024] Open
Abstract
Background Since there is no clear priority or selection principle in the guidelines for myasthenia crisis, therapeutic plasma exchange (TPE) and intravenous immunoglobulin are often administered randomly. However, it should be more prudent in taking TPE due to its higher cost and risk. Studying its early response factors is crucial for managing myasthenia crisis and can improve medical and economic benefits. Methods A prospective observational study was conducted, and patients classified as having "impending myasthenia crisis" or experiencing a myasthenia crisis and treated by TPE were included. The primary endpoint was the response after TPE. Univariate logistic regression analysis and repeated measurement were performed to analyze factors related to TPE efficacy. Results A total of 30 patients who treated with TPE as their fast-acting treatments were enrolled. After TPE, those whose QMGs and/or MGCs decreased by ≥5 points or ≥30% of the baseline were judged as "response group", accounting for 66.67% (20/30). Respiratory symptoms had a response rate of 72.00% (18/25), showing the most remarkable improvement. Meanwhile, extraocular symptoms were the least sensitive, with only 8.00% (2/25) showing efficacy. Thymoma (100.00% vs 50.00%, P=0.002) and a high concentration of AChR-Ab (37.37 nmol/L vs 25.4 nmol/L, P=0.039) were common in the early response group. Repeated measures showed significant changes in AChR-Ab and CD19+ B cells before and after TPE (all with P < 0.05). After treatment, the CD19+ B cells tended to decrease in the response group. Discussion These results indicated that, for AChR-Ab positive generalized MG, TPE can quickly improve respiratory symptoms. Thymoma and a high concentration of AChR-Ab before TPE predict an early better response. Additionally, TPE may work by decreasing AChR-Ab levels and inducing immune regulation. Future prospective and randomized controlled studies are needed.
Collapse
Affiliation(s)
- Jiaxin Chen
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Li Feng
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Shiyin Li
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Haiyan Wang
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Xin Huang
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Cunzhou Shen
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| | - Huiyu Feng
- Department of Neurology and Neurointensive Care Unit, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, People’s Republic of China
| |
Collapse
|
9
|
Mina-Osorio P, Tran MH, Habib AA. Therapeutic Plasma Exchange Versus FcRn Inhibition in Autoimmune Disease. Transfus Med Rev 2024; 38:150767. [PMID: 37867088 DOI: 10.1016/j.tmrv.2023.150767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023]
Abstract
Therapeutic plasma exchange (TPE or PLEX) is used in a broad range of autoimmune diseases, with the goal of removing autoantibodies from the circulation. A newer approach for the selective removal of immunoglobulin G (IgG) antibodies is the use of therapeutic molecules targeting the neonatal Fc receptor (FcRn). FcRn regulates IgG recycling, and its inhibition results in a marked decrease in circulating autoantibodies of the IgG subtype. The difference between FcRn inhibition and PLEX is often questioned. With anti-FcRn monoclonal antibodies (mAbs) and fragments only recently entering this space, limited data are available regarding long-term efficacy and safety. However, the biology of FcRn is well understood, and mounting evidence regarding the efficacy, safety, and potential differences among compounds in development is available, allowing us to compare against nonselective plasma protein depletion methods such as PLEX. FcRn inhibitors may have distinct advantages and disadvantages over PLEX in certain scenarios. Use of PLEX is preferred over FcRn inhibition where removal of antibodies other than IgG or when concomitant repletion of missing plasma proteins is needed for therapeutic benefit. Also, FcRn targeting has not yet been studied for use in acute flares or crisis states of IgG-mediated diseases. Compared with PLEX, FcRn inhibition is associated with less invasive access requirements, more specific removal of IgG versus other immunoglobulins without a broad impact on circulating proteins, and any impacts on other therapeutic drug levels are restricted to other mAbs. In addition, the degree of IgG reduction is similar with FcRn inhibitors compared with that afforded by PLEX. Here we describe the scientific literature regarding the use of PLEX and FcRn inhibitors in autoimmune diseases and provide an expert discussion around the potential benefits of these options in varying clinical conditions and scenarios.
Collapse
Affiliation(s)
| | - Minh-Ha Tran
- Department of Pathology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ali A Habib
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
10
|
Crisafulli S, Boccanegra B, Carollo M, Bottani E, Mantuano P, Trifirò G, De Luca A. Myasthenia Gravis Treatment: From Old Drugs to Innovative Therapies with a Glimpse into the Future. CNS Drugs 2024; 38:15-32. [PMID: 38212553 DOI: 10.1007/s40263-023-01059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Myasthenia gravis (MG) is a rare autoimmune disease that causes debilitating muscle weakness due to impaired neuromuscular transmission. Since most (about 80-90%) MG patients present autoantibodies against the acetylcholine receptor, standard medical therapy consists of symptomatic treatment with acetylcholinesterase inhibitors (e.g., pyridostigmine). In addition, considering the autoimmune basis of MG, standard therapy includes immunomodulating agents, such as corticosteroids, azathioprine, cyclosporine A, and cyclophosphamide. New strategies have been proposed for the treatment of MG and include complement blockade (i.e., eculizumab, ravulizumab, and zilucoplan) and neonatal Fc receptor antagonism (i.e., efgartigimod and rozanolixizumab). The aim of this review is to provide a detailed overview of the pre- and post-marketing evidence on the five pharmacological treatments most recently approved for the treatment of MG, by identifying both preclinical and clinical studies registered in clinicaltrials.gov. A description of the molecules currently under evaluation for the treatment of MG is also provided.
Collapse
Affiliation(s)
| | - Brigida Boccanegra
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Massimo Carollo
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy
| | - Paola Mantuano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluca Trifirò
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy.
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
11
|
Budhram A, Flanagan EP. Optimizing the diagnostic performance of neural antibody testing for paraneoplastic and autoimmune encephalitis in clinical practice. HANDBOOK OF CLINICAL NEUROLOGY 2024; 200:365-382. [PMID: 38494290 DOI: 10.1016/b978-0-12-823912-4.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The detection of neural antibodies in patients with paraneoplastic and autoimmune encephalitis has majorly advanced the diagnosis and management of neural antibody-associated diseases. Although testing for these antibodies has historically been restricted to specialized centers, assay commercialization has made this testing available to clinical chemistry laboratories worldwide. This improved test accessibility has led to reduced turnaround time and expedited diagnosis, which are beneficial to patient care. However, as the utilization of these assays has increased, so too has the need to evaluate how they perform in the clinical setting. In this chapter, we discuss assays for neural antibody detection that are in routine use, draw attention to their limitations and provide strategies to help clinicians and laboratorians overcome them, all with the aim of optimizing neural antibody testing for paraneoplastic and autoimmune encephalitis in clinical practice.
Collapse
Affiliation(s)
- Adrian Budhram
- Department of Clinical Neurological Sciences, Western University, London Health Sciences Centre, London, ON, Canada; Department of Pathology and Laboratory Medicine, Western University, London Health Sciences Centre, London, ON, Canada.
| | - Eoin P Flanagan
- Department of Neurology, Mayo Clinic, Rochester, MN, United States; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
12
|
Tannemaat MR, Huijbers MG, Verschuuren JJGM. Myasthenia gravis-Pathophysiology, diagnosis, and treatment. HANDBOOK OF CLINICAL NEUROLOGY 2024; 200:283-305. [PMID: 38494283 DOI: 10.1016/b978-0-12-823912-4.00026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Myasthenia gravis (MG) is an autoimmune disease characterized by dysfunction of the neuromuscular junction resulting in skeletal muscle weakness. It is equally prevalent in males and females, but debuts at a younger age in females and at an older age in males. Ptosis, diplopia, facial bulbar weakness, and limb weakness are the most common symptoms. MG can be classified based on the presence of serum autoantibodies. Acetylcholine receptor (AChR) antibodies are found in 80%-85% of patients, muscle-specific kinase (MuSK) antibodies in 5%-8%, and <1% may have low-density lipoprotein receptor-related protein 4 (Lrp4) antibodies. Approximately 10% of patients are seronegative for antibodies binding the known disease-related antigens. In patients with AChR MG, 10%-20% have a thymoma, which is usually detected at the onset of the disease. Important differences between clinical presentation, treatment responsiveness, and disease mechanisms have been observed between these different serologic MG classes. Besides the typical clinical features and serologic testing, the diagnosis can be established with additional tests, including repetitive nerve stimulation, single fiber EMG, and the ice pack test. Treatment options for MG consist of symptomatic treatment (such as pyridostigmine), immunosuppressive treatment, or thymectomy. Despite the treatment with symptomatic drugs, steroid-sparing immunosuppressants, intravenous immunoglobulins, plasmapheresis, and thymectomy, a large proportion of patients remain chronically dependent on corticosteroids (CS). In the past decade, the number of treatment options for MG has considerably increased. Advances in the understanding of the pathophysiology have led to new treatment options targeting B or T cells, the complement cascade, the neonatal Fc receptor or cytokines. In the future, these new treatments are likely to reduce the chronic use of CS, diminish side effects, and decrease the number of patients with refractory disease.
Collapse
Affiliation(s)
- Martijn R Tannemaat
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maartje G Huijbers
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
13
|
Sänger F, Dörfelt S, Giani B, Buhmann G, Fischer A, Dörfelt R. Successful Emergency Management of a Dog with Ventilator-Dependent Acquired Myasthenia Gravis with Immunoadsorption. Animals (Basel) 2023; 14:33. [PMID: 38200764 PMCID: PMC10778221 DOI: 10.3390/ani14010033] [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: 10/31/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
A one-year-old, female intact Samoyed, 12.5 kg, was presented with coughing for 2 weeks, progressive appendicular and axial muscle weakness, megaesophagus and labored breathing for 5 days. There was no improvement with standard treatment. Acquired myasthenia gravis was suspected and the dog was referred with increasing dyspnea. At presentation, the dog showed a severely reduced general condition, was non-ambulatory and showed abdominal and severely labored breathing. A marked hypercapnia (PvCO2 = 90.1 mmHg) was present in venous blood gas analysis. The serum anti-acetylcholine receptor antibody test was consistent with acquired myasthenia gravis (2.1 nmol/L). The dog was anesthetized with propofol and mechanically ventilated with a Hamilton C1 ventilator. Immunoadsorption was performed with the COM.TEC® and ADAsorb® platforms and a LIGASORB® adsorber to eliminate anti-acetylcholine receptor antibodies. Local anticoagulation was performed with citrate. Treatment time for immunoadsorption was 1.5 h with a blood flow of 50 mL/min. A total plasma volume of 1.2 L was processed. Further medical treatment included intravenous fluid therapy, maropitant, esomeprazole, antibiotic therapy for aspiration pneumonia and neostigmine 0.04 mg/kg intramuscularly every 6 h for treatment of acquired myasthenia gravis. Mechanical ventilation was stopped after 12 h. A percutaneous gastric feeding tube was inserted under endoscopic control on day 2 for further medical treatment and nutrition. A second treatment with immunoadsorption was performed on day 3. Again, a total plasma volume of 1.2 L was processed. Immediately after this procedure, the dog regained muscle strength and was able to stand and to walk. After 6 days, the dog was discharged from the hospital. This is the first report of immunoadsorption for emergency management of a dog with acute-fulminant acquired myasthenia gravis. Immunoadsorption may be an additional option for emergency treatment in dogs with severe signs of acquired myasthenia gravis.
Collapse
Affiliation(s)
- Florian Sänger
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (F.S.); (B.G.); (G.B.); (A.F.)
| | | | - Bettina Giani
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (F.S.); (B.G.); (G.B.); (A.F.)
| | - Gesine Buhmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (F.S.); (B.G.); (G.B.); (A.F.)
| | - Andrea Fischer
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (F.S.); (B.G.); (G.B.); (A.F.)
| | - René Dörfelt
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (F.S.); (B.G.); (G.B.); (A.F.)
| |
Collapse
|
14
|
Cone Sullivan J, Conklin SE, Conrad S, Horowitz C, Diethelm M, Comenzo R. Therapeutic plasma exchange decreases plasma anti-SARS-CoV-2 spike IgG without increasing the proximate incidence of COVID-19. J Clin Apher 2023; 38:721-726. [PMID: 37706521 DOI: 10.1002/jca.22087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Therapeutic plasma exchange (TPE) removes both pathologic and protective immunoglobulins (Ig). SARS-CoV-2 immunity is partially mediated by anti-SARS-CoV-2 spike antibodies (SAb), which impair viral host-cell invasion. Nonetheless, the systematic effect of TPE on SAb concentration and SARS-CoV-2 immunity is unknown. METHODS Paired plasma waste specimens from the first (first-TPE) and last (last-TPE) TPE treatment were collected from 9 patients between July 21, 2021 and March 1, 2022. The effects of TPE on Ig levels were assessed by quantitatively comparing the SAb, total IgG, and total IgM levels first-/last-TPE treatment. Complementary qualitative assessment for these changes was achieved via protein electrophoresis (PEP) and immunofixation (IFE). A retrospective review was performed to investigate the incidence of new SARS-CoV-2 infections following TPE v. other treatment at the same outpatient apheresis/infusion center during the same time frame. RESULTS Median SAb levels between the first- and last-TPE waste specimens decreased significantly from 424.6 AU/mL to 17.0 AU/mL (P = 0.004). Concordantly, PEP and IFE analysis demonstrated broad Ig decreases. Cumulative incidence of subsequent COVID-19 diagnosis at 30, 90, and 180 days post-procedure did not differ between the TPE v. other treatment groups (n = 709 total patients). CONCLUSIONS TPE significantly reduced SAb levels, a marker of SARS-CoV-2 immunity, but did not appear to provoke increased incidence of COVID-19 infections. Further investigation of the kinetics of TPE-mediated SAb decrease and post-TPE recovery are warranted.
Collapse
Affiliation(s)
- Jensyn Cone Sullivan
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Steven E Conklin
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Anatomic & Clinical Pathology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Stephanie Conrad
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Coby Horowitz
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Mark Diethelm
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Raymond Comenzo
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Yunce M, Katyal N, Monis GF, Muppidi S. Neonatal Fc receptor blockade as emerging therapy in diseases with plasma exchange indications. J Clin Apher 2023; 38:632-640. [PMID: 37183667 DOI: 10.1002/jca.22055] [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: 12/12/2022] [Revised: 02/20/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023]
Abstract
Neonatal Fc receptor (FcRn) blockade may represent a mechanism similar to plasma exchange (PLEX) in reducing immunoglobulin levels and thus have a broad implication for apheresis practitioners. Although only efgartigimod received FDA approval for myasthenia gravis in December 2021, multiple trials are currently underway with different FcRn therapies in a varied group of IgG antibody-mediated neurological and hematological disorders which are outlined in this review. In this review we discuss FcRn's mechanism of action, and its potential use in various neurological and non-neurological diseases. In addition, we further compare the kinetics and adverse events of PLEX and FcRn blockade. We encourage apheresis practitioners to be familiar with this class of drugs in order to better understand how these two therapies can be used either standalone, or in combination with other therapies as both FcRn antagonism and PLEX improve clinical state by reducing IgG levels and pathogenic antibodies.
Collapse
Affiliation(s)
- Muharrem Yunce
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Nakul Katyal
- Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Grace Fortes Monis
- Department of Pathology, University of California Davis, Sacramento, California, USA
| | - Srikanth Muppidi
- Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
16
|
Bhandari V, Bril V. FcRN receptor antagonists in the management of myasthenia gravis. Front Neurol 2023; 14:1229112. [PMID: 37602255 PMCID: PMC10439012 DOI: 10.3389/fneur.2023.1229112] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disorder characterized by autoantibodies specifically directed against proteins located within the postsynaptic membrane of the neuromuscular junction. These pathogenic autoantibodies can be reduced by therapies such as plasma exchange, IVIG infusions and other immunosuppressive agents. However, there are significant side effects associated with most of these therapies. Since there is a better understanding of the molecular structure and the biological properties of the neonatal Fc receptors (FcRn), it possesses an attractive profile in treating myasthenia gravis. FcRn receptors prevent the catabolism of IgG by impeding their lysosomal degradation and facilitating their extracellular release at physiological pH, consequently extending the IgG half-life. Thus, the catabolism of IgG can be enhanced by blocking the FcRn, leading to outcomes similar to those achieved through plasma exchange with no significant safety concerns. The available studies suggest that FcRn holds promise as a versatile therapeutic intervention, capable of delivering beneficial outcomes in patients with distinct characteristics and varying degrees of MG severity. Efgartigimod is already approved for the treatment of generalized MG, rozanolixizumab is under review by health authorities, and phase 3 trials of nipocalimab and batoclimab are underway. Here, we will review the available data on FcRn therapeutic agents in the management of MG.
Collapse
Affiliation(s)
| | - Vera Bril
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Huang EJC, Wu MH, Wang TJ, Huang TJ, Li YR, Lee CY. Myasthenia Gravis: Novel Findings and Perspectives on Traditional to Regenerative Therapeutic Interventions. Aging Dis 2023; 14:1070-1092. [PMID: 37163445 PMCID: PMC10389825 DOI: 10.14336/ad.2022.1215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/15/2022] [Indexed: 05/12/2023] Open
Abstract
The prevalence of myasthenia gravis (MG), an autoimmune disorder, is increasing among all subsets of the population leading to an elevated economic and social burden. The pathogenesis of MG is characterized by the synthesis of autoantibodies against the acetylcholine receptor (AChR), low-density lipoprotein receptor-related protein 4 (LRP4), or muscle-specific kinase at the neuromuscular junction, thereby leading to muscular weakness and fatigue. Based on clinical and laboratory examinations, the research is focused on distinguishing MG from other autoimmune, genetic diseases of neuromuscular transmission. Technological advancements in machine learning, a subset of artificial intelligence (AI) have been assistive in accurate diagnosis and management. Besides, addressing the clinical needs of MG patients is critical to improving quality of life (QoL) and satisfaction. Lifestyle changes including physical exercise and traditional Chinese medicine/herbs have also been shown to exert an ameliorative impact on MG progression. To achieve enhanced therapeutic efficacy, cholinesterase inhibitors, immunosuppressive drugs, and steroids in addition to plasma exchange therapy are widely recommended. Under surgical intervention, thymectomy is the only feasible alternative to removing thymoma to overcome thymoma-associated MG. Although these conventional and current therapeutic approaches are effective, the associated adverse events and surgical complexity limit their wide application. Moreover, Restivo et al. also, to increase survival and QoL, further recent developments revealed that antibody, gene, and regenerative therapies (such as stem cells and exosomes) are currently being investigated as a safer and more efficacious alternative. Considering these above-mentioned points, we have comprehensively reviewed the recent advances in pathological etiologies of MG including COVID-19, and its therapeutic management.
Collapse
Affiliation(s)
- Evelyn Jou-Chen Huang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Meng-Huang Wu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Tsung-Jen Wang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Tsung-Jen Huang
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yan-Rong Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Ching-Yu Lee
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
18
|
Matic A, Alfaidi N, Bril V. An evaluation of rozanolixizumab-noli for the treatment of anti-AChR and anti-MuSK antibody-positive generalized myasthenia gravis. Expert Opin Biol Ther 2023; 23:1163-1171. [PMID: 38099334 DOI: 10.1080/14712598.2023.2296126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION Myasthenia gravis (MG) is an auto-immune disease characterized by fluctuating symptoms of muscle weakness and fatigue. Corticosteroids and corticosteroid-sparing broad-spectrum immunosuppression play a great role in the treatment of myasthenia gravis. However, debilitating side effects and long time to treatment effect highlight the need for development of novel target-specific medications. Rozanolixizumab is a highly specific neonatal Fc receptor (FcRn) inhibitor that acts on immunoglobulin G (IgG) homeostasis. Results from the MycarinG Phase III randomized controlled trial demonstrated significant efficacy of rozanolixizumab in generalized MG in terms of primary outcome and all secondary endpoints, tolerability, and safety compared to placebo. AREAS COVERED We included different trials on myasthenia gravis and rozanolixizumab which include Phase II (NCT03052751) and Phase III MycarinG (NCT03971422) studies. EXPERT OPINION Clinical trials have demonstrated that rozanolixizumab has strong efficacy with a 78% reduction in pathogenic IgG like plasma exchange (PLEX) and has therapeutic benefits comparable with PLEX and IVIG. It has less treatment adverse events and is easily accessible through subcutaneous infusion. The safety and effectiveness of rozanolixizumab need to be assessed further in the real-world context in post-marketing studies. If current trial information holds true, rozanolixizumab may become a medication of choice for MG in succeeding years.
Collapse
Affiliation(s)
- Alexandria Matic
- The Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Nouf Alfaidi
- The Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Vera Bril
- The Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Zhou Y, Jiang S. Roles of FcRn in Antigen-Presenting Cells during Autoimmunity and a Clinical Evaluation of Efgartigimod as an FcRn Blocker. Pathogens 2023; 12:817. [PMID: 37375507 DOI: 10.3390/pathogens12060817] [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: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The immune system is a complex network of multiple cells, tissues, and organs that protects the body against foreign pathogenic invaders. However, the immune system may mistakenly attack healthy cells and tissues due to the cross-reactivity of anti-pathogen immunity, leading to autoimmunity by autoreactive T cells and/or autoantibody-secreting B cells. Autoantibodies can accumulate, resulting in tissue or organ damage. The neonatal crystallizable fragment receptor (FcRn) is an important factor in immune regulation through controlling the trafficking and recycling of immunoglobulin G (IgG) molecules, the most abundant antibody in humoral immunity. In addition to its role in IgG trafficking and recycling, FcRn is also involved in antigen presentation, which is a crucial step in the activation of the adaptive immune response via directing the internalization and trafficking of antigen-bound IgG immune complexes into compartments of degradation and presentation in antigen-presenting cells. Efgartigimod, an FcRn inhibitor, has shown promise in reducing the levels of autoantibodies and alleviating the autoimmune severity of myasthenia gravis, primary immune thrombocytopenia, and pemphigus vulgaris/foliaceus. This article aims to provide an overview of the importance of FcRn in antigen-presenting cells and its potential as a therapeutic target in autoimmune diseases, using efgartigimod as an example.
Collapse
Affiliation(s)
- Yihan Zhou
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Shisong Jiang
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
20
|
Ceglédi A, Dolgos J, Fekete M, Gopcsa L, Várkonyi A, Vilimi B, Mikala G, Bodó I. Delayed spontaneous remission of acquired factor V inhibitor refractory to immunosuppressive therapy with pregnancy-associated improvement. Pathol Oncol Res 2023; 29:1611250. [PMID: 37334173 PMCID: PMC10272408 DOI: 10.3389/pore.2023.1611250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
Introduction: Acquired factor V inhibitor (AFVI) is a rare autoimmune bleeding disorder. The treatment of AFVI is challenging, and patients often require both bleeding control and inhibitor eradication. Methods: We conducted a retrospective analysis of the medical records of a 35-year-old Caucasian woman who presented with severe AFVI-induced bleeding and subsequent immunosuppressive therapy. Results: To provide haemostasis, rFVIIa was given with good efficacy. The patient was treated with various combinations of immunosuppressive regimens over the course of 2.5 years, including plasmapheresis plus immunoglobulins, dexamethasone + rituximab, cyclophosphamide + dexamethasone + rituximab + cyclosporine, cyclosporin + sirolimus + cyclophosphamide + dexamethasone, bortezomib + sirolimus + methylprednisolone, and sirolimus + mycophenolate mofetil. Although these treatment modalities resulted in intermittent partial reversals of AFVI over 2.5 years, eventually the inhibitor became therapy-resistant. However, following the discontinuation of all immunosuppressive therapy, the patient experienced a partial spontaneous remission, which was followed by a pregnancy. During the pregnancy, the FV activity increased to 54% and the coagulation parameters returned to normal levels. The patient underwent Caesarean section without any bleeding complications and delivered a healthy child. Discussion: The use of an activated bypassing agent for bleeding control is effective in patients with severe AFVI. The presented case is unique because the treatment regimens included multiple combinations of immunosuppressive agents. This demonstrates that AFVI patients may undergo spontaneous remission even after multiple courses of ineffective immunosuppressive protocols. Additionally, pregnancy-associated improvement of AFVI is an important finding that warrants further investigation.
Collapse
Affiliation(s)
- Andrea Ceglédi
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - János Dolgos
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Mónika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - László Gopcsa
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Andrea Várkonyi
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Beáta Vilimi
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Gábor Mikala
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Imre Bodó
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| |
Collapse
|
21
|
Hansen M, Neilson L, Parikh M, Katirji B. Greater Number of Plasma Exchanges Does Not Improve Outcome in Myasthenic Crisis. J Clin Neuromuscul Dis 2023; 24:199-206. [PMID: 37219863 DOI: 10.1097/cnd.0000000000000421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
OBJECTIVES To determine the relationship between the number of plasma exchanges and clinical outcome in patients experiencing myasthenic crisis. METHODS We retrospectively reviewed all episodes of myasthenia gravis exacerbation/crisis who received plasmapheresis in patients admitted to a single-center tertiary care referral center from July 2008 to July 2017. We performed statistical analyses to determine whether the increased number of plasma exchanges improves the primary outcome (hospital length of stay) and the secondary outcome (disposition to home, skilled nursing facility, long-term acute care hospital, or death). RESULTS There is neither clinically observable nor statistically significant improvement in length of stay or disposition on discharge in patients who received 6 or greater sessions of plasmapheresis. CONCLUSIONS This study provides class IV evidence that extending the number of plasma exchanges beyond 5 does not correlate with decreased hospital length of stay or improved discharge disposition in patients experiencing myasthenic crisis.
Collapse
Affiliation(s)
- Michael Hansen
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH. Dr. Hansen is now with the Department of Neurology, University of Wisconsin, Madison, WI. Dr. Neilson is now with the Department of Neurology, Oregon Health and Science University, Portland, OR, and the Department of Neurology, Veterans Affairs Medical Center, Portland, OR. Dr. Parikh is now with the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.; and
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Lee Neilson
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH. Dr. Hansen is now with the Department of Neurology, University of Wisconsin, Madison, WI. Dr. Neilson is now with the Department of Neurology, Oregon Health and Science University, Portland, OR, and the Department of Neurology, Veterans Affairs Medical Center, Portland, OR. Dr. Parikh is now with the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.; and
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Melanie Parikh
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH. Dr. Hansen is now with the Department of Neurology, University of Wisconsin, Madison, WI. Dr. Neilson is now with the Department of Neurology, Oregon Health and Science University, Portland, OR, and the Department of Neurology, Veterans Affairs Medical Center, Portland, OR. Dr. Parikh is now with the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.; and
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Bashar Katirji
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH. Dr. Hansen is now with the Department of Neurology, University of Wisconsin, Madison, WI. Dr. Neilson is now with the Department of Neurology, Oregon Health and Science University, Portland, OR, and the Department of Neurology, Veterans Affairs Medical Center, Portland, OR. Dr. Parikh is now with the Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.; and
- Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
22
|
Foettinger F, Pilz G, Wipfler P, Harrer A, Kern JM, Trinka E, Moser T. Immunomodulatory Aspects of Therapeutic Plasma Exchange in Neurological Disorders—A Pilot Study. Int J Mol Sci 2023; 24:ijms24076552. [PMID: 37047524 PMCID: PMC10095570 DOI: 10.3390/ijms24076552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Therapeutic plasma exchange (TPE) is used for drug-resistant neuroimmunological disorders, but its mechanism of action remains poorly understood. We therefore prospectively explored changes in soluble, humoral, and cellular immune components associated with TPE. We included ten patients with neurological autoimmune disorders that underwent TPE and assessed a panel of clinically relevant pathogen-specific antibodies, total serum immunoglobulin (Ig) levels, interleukin-6 (IL-6, pg/mL), C-reactive protein (CRP, mg/dL), procalcitonin (PCT, µg/L) and major lymphocyte subpopulations (cells/µL). Blood was collected prior to TPE (pre-TPE, baseline), immediately after TPE (post-TPE), as well as five weeks (follow-up1) and 130 days (follow-up2) following TPE. Pathogen-specific antibody levels were reduced by −86% (p < 0.05) post-TPE and recovered to 55% (follow-up1) and 101% (follow-up2). Ig subclasses were reduced by −70–89% (p < 0.0001) post-TPE with subsequent complete (IgM/IgA) and incomplete (IgG) recovery throughout the follow-ups. Mean IL-6 and CRP concentrations increased by a factor of 3–4 at post-TPE (p > 0.05) while PCT remained unaffected. We found no alterations in B- and T-cell populations. No adverse events related to TPE occurred. TPE induced a profound but transient reduction in circulating antibodies, while the investigated soluble immune components were not washed out. Future studies should explore the effects of TPE on particular cytokines and assess inflammatory lymphocyte lineages to illuminate the mode of action of TPE beyond autoantibody removal.
Collapse
|
23
|
Wang Y, Zhong X, Wang H, Peng Y, Shi F, Jia D, Yang H, Zeng Q, Quan C, ZhangBao J, Lee M, Qi J, Chen X, Qiu W. Batoclimab as an add-on therapy in neuromyelitis optica spectrum disorder patients with acute attacks. Eur J Neurol 2023; 30:195-203. [PMID: 36087008 DOI: 10.1111/ene.15561] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/13/2022] [Accepted: 09/04/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND PURPOSE Neuromyelitis optica spectrum disorder (NMOSD) is a severe neurological inflammatory disease mainly caused by pathogenic aquaporin-4 antibodies (AQP4-IgG). The safety and efficacy of the neonatal Fc receptor antagonist batoclimab addition to conventional intravenous methylprednisolone pulse (IVMP) therapy in patients with NMOSD acute attacks was assessed. METHODS In an open-label, dose-escalation phase 1b study, NMOSD patients with acute myelitis and/or optic neuritis received four doses of weekly subcutaneous injections of either 340 mg or 680 mg batoclimab with concurrent IVMP and were followed up for 27 weeks. The primary end-points were safety and tolerability. Secondary end-points included pharmacodynamics and efficacy, with key efficacy assessment at week 4. RESULTS In total nine NMOSD patients were enrolled, including two and seven in the 340 and 680 mg groups. Five patients had acute myelitis, while the remaining four had unilateral optic neuritis. Batoclimab add-on therapy had an overall good safety profile without serious adverse events. In the 680 mg group, mean immunoglobulin G (IgG) reached its maximum reduction at the last dose (day 22). In the meantime, AQP4-IgG was undetectable in six of seven subjects whose baseline AQP4-IgG titers ranged from 1:32 to 1:320. Expanded Disability Status Scale score was reduced by 1.3 ± 0.4 at week 4 (2.7 ± 1.3) compared with baseline (4.0 ± 1.0). CONCLUSIONS Batoclimab add-on therapy to IVMP is safe and tolerated in patients with NMOSD. Preliminary evidence suggests a beneficial neurological effect. A randomized controlled trial would be needed to prove the efficacy.
Collapse
Affiliation(s)
- Yuge Wang
- Department of Neurology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaonan Zhong
- Department of Neurology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fudong Shi
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Dongmei Jia
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Quan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingzi ZhangBao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Jun Qi
- Harbour BioMed, Shanghai, China
| | | | - Wei Qiu
- Department of Neurology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | |
Collapse
|
24
|
Meisel A, Baggi F, Behin A, Evoli A, Kostera-Pruszczyk A, Mantegazza R, Morales RJ, Punga AR, Sacconi S, Schroeter M, Verschuuren J, Crathorne L, Holmes K, Leite MI. Role of autoantibody levels as biomarkers in the management of patients with myasthenia gravis: A systematic review and expert appraisal. Eur J Neurol 2023; 30:266-282. [PMID: 36094738 DOI: 10.1111/ene.15565] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/26/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Although myasthenia gravis (MG) is recognized as an immunoglobulin G autoantibody-mediated disease, the relationship between autoantibody levels and disease activity in MG is unclear. We sought to evaluate this landscape through systematically assessing the evidence, testing the impact of predefined variables on any relationship, and augmenting with expert opinion. METHODS In October 2020, a forum of leading clinicians and researchers in neurology from across Europe (Expert Forum for Rare Autoantibodies in Neurology in Myasthenia Gravis) participated in a series of virtual meetings that took place alongside the conduct of a systematic literature review (SLR). RESULTS Forty-two studies were identified meeting inclusion criteria. Of these, 10 reported some correlation between a patient's autoantibody level and disease severity. Generally, decreased autoantibody levels (acetylcholine receptor, muscle-specific kinase, and titin) were positively and significantly correlated with improvements in disease severity (Quantitative Myasthenia Gravis score, Myasthenia Gravis Composite score, Myasthenia Gravis Activities of Daily Living score, Myasthenia Gravis Foundation of America classification). Given the limited evidence, testing the impact of predefined variables was not feasible. CONCLUSIONS This first SLR to assess whether a correlation exists between autoantibody levels and disease activity in patients with MG has indicated a potential positive correlation, which could have clinical implications in guiding treatment decisions. However, in light of the limited and variable evidence, we cannot currently recommend routine clinical use of autoantibody level testing in this context. For now, patient's characteristics, clinical disease course, and laboratory data (e.g., autoantibody status, thymus histology) should inform management, alongside patient-reported outcomes. We highlight the need for future studies to reach more definitive conclusions on this relationship.
Collapse
Affiliation(s)
- Andreas Meisel
- Department of Neurology, Integrated Myasthenia Gravis Center, NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fulvio Baggi
- Neuroimmunology and Neuromuscular Diseases Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Anthony Behin
- Department of Neuromyology, Pitié-Salpêtrière Hospital, Public Hospital Network of Paris, Institute of Myology, Paris, France
| | - Amelia Evoli
- Catholic University of the Sacred Heart and Agostino Gemelli University Polyclinic Foundation, Scientific Institute for Research and Health Care, Rome, Italy
| | | | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Scientific Institute for Research and Health Care Foundation Carlo Besta Neurological Institute, Milan, Italy
| | | | - Anna Rostedt Punga
- Department of Medical Science, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | | | - Michael Schroeter
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Jan Verschuuren
- Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Maria-Isabel Leite
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Guptill JT, Sleasman JW, Steeland S, Sips M, Gelinas D, de Haard H, Azar A, Winthrop KL. Effect of FcRn antagonism on protective antibodies and to vaccines in IgG-mediated autoimmune diseases pemphigus and generalised myasthenia gravis. Autoimmunity 2022; 55:620-631. [PMID: 36036539 DOI: 10.1080/08916934.2022.2104261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Antagonism of the neonatal Fc receptor (FcRn) by efgartigimod has been studied in several autoimmune diseases mediated by immunoglobulin G (IgG) as a therapeutic approach to remove pathogenic IgGs. Whereas reduction of pathogenic titres has demonstrated efficacy in multiple autoimmune diseases, reducing total IgG could potentially increase infection risk in patients receiving FcRn antagonists. The objective of this study was to analyse the effect of FcRn antagonism with efgartigimod on existing protective antibody titres and the ability to mount an immune response after vaccine challenge. Serum levels of total IgG and protective antibodies against tetanus toxoid (TT), varicella zoster virus (VZV), and pneumococcal capsular polysaccharide (PCP) were measured in all patients enrolled in an open-label trial of efgartigimod for the treatment of pemphigus. Vaccine specific-responses were assessed by measuring changes in IgG titres in patients with generalised myasthenia gravis (gMG) who were treated with efgartigimod and who received influenza, pneumococcal, or coronavirus disease 2019 (COVID-19) vaccines during participation in the double-blind trial ADAPT or open-label extension, ADAPT+ (n = 17). FcRn antagonism reduced levels of protective anti-TT, anti-VZV, and anti-PCP antibodies and total IgG to a similar extent; anti-TT and anti-VZV titres remained above minimally protective thresholds for the majority of patients, (10/12) 83% and (14/15) 93% respectively. Protective antibodies returned to baseline values upon treatment cessation. Antigen-specific IgG responses to influenza, pneumococcal, and COVID-19 immunisation were detected in patients with gMG who received these vaccines while undergoing therapy with efgartigimod. In conclusion, FcRn antagonism with efgartigimod did not hamper generation of IgG responses but did transiently reduce IgG titres of all specificities.
Collapse
Affiliation(s)
- Jeffrey T Guptill
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA.,argenx, Ghent, Belgium
| | - John W Sleasman
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | | | | | - Antoine Azar
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin L Winthrop
- Division of Infectious Disease, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
26
|
Tran MH. "Plasma exchange in a bottle": An overview of efgartigimod for apheresis practitioners. J Clin Apher 2022; 37:512-515. [PMID: 35997018 DOI: 10.1002/jca.22002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 11/11/2022]
Abstract
Efgartigimod represents a first-in-class immunomodulatory agent that is comparable to TPE in reducing immunoglobulin levels. This translates to reductions in Myasthenia Gravis symptom scores with maximal effect following the fourth weekly efgartigimod dose. Efgartigimod received FDA approval in December of 2021 and may be an alternative particularly for patients on long-term TPE regimens of weekly or less frequent. Apheresis practitioners, especially those managing long-term apheresis in seropositive individuals, may therefore see some of their patients transitioned from plasma exchange to efgartigimod. Long-term experience with efgartigimod remains lacking and studies are needed to establish the role of efgartigimod in the acute setting.
Collapse
Affiliation(s)
- Minh-Ha Tran
- Department of Pathology and Laboratory Medicine, University of California, Irvine School of Medicine, Orange, California, USA
| |
Collapse
|
27
|
Total Plasma Exchange in Neuromuscular Junction Disorders—A Single-Center, Retrospective Analysis of the Efficacy, Safety and Potential Diagnostic Properties in Doubtful Diagnosis. J Clin Med 2022; 11:jcm11154383. [PMID: 35955999 PMCID: PMC9369332 DOI: 10.3390/jcm11154383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023] Open
Abstract
Neuromuscular junction disorders (NJDs) are a heterogeneous group of diseases including myasthenia gravis (MG). In some cases, patients are present with myasthenic symptoms without evidence of autoimmune antibodies, making diagnosis challenging. Total plasma exchange (TPE) has proven efficacy in NJDs. The objective is to describe the safety and efficacy of TPE in NJD patients with questionable disease activity or uncertain diagnosis in order to assess the diagnostic potential of TPE. We report an observational, retrospective cohort study of clinical routine data. All the data were derived from the electronic medical records of the Department of Neurology at University Hospital Essen. We searched for patients with NJDs between 1 July 2018 and 30 June 2021. Of the 303 patients who presented to the department with NJDs, 20 were treated with TPE; 9 patients did not show a measurable benefit from TPE (45%), 6 of whom were diagnosed with seronegative MG. Of these, 3 (50%) had long-standing ocular symptoms. There were decreases in the mean arterial pressure, hemoglobin, hematocrit and fibrinogen during treatment, which were not considered clinically relevant. In (seronegative) myasthenic patients, TPE may help to verify an uncertain diagnosis or to reveal possible muscle damage, allowing unnecessary therapy to be avoided.
Collapse
|
28
|
Fan L, Yang Y, Zhang F, Huang F. Clinical Efficacy of Immunoglobulin Combined with Glucocorticoids in the Treatment of Oculomotor Myasthenia Gravis in Children and the Effect on Serum Immunity. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1772881. [PMID: 35844442 PMCID: PMC9286931 DOI: 10.1155/2022/1772881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022]
Abstract
To investigate the effects of treatment with immunoglobulin on clinical outcomes and immune function in children with oculomotor myasthenia gravis. The clinical data of 100 pediatric patients with oculomotor myasthenia gravis treated in our hospital from January 2019 to December 2021 were selected as the subjects of this retrospective study and divided into a comparison group and a treatment group according to the different treatment methods. The comparison group was treated with glucocorticoids, and the treatment group was treated with immunoglobulin on the basis of the comparison group. The differences in the serum indexes, the effects of immune function, and the clinical efficacy of the two groups were observed and compared. It was found the comparison of immunoglobulin G (IgG), immunoglobulin A (IgA), and immunoglobulin M (IgM) after treatment was significantly different and lower in the treatment group than in the comparison group; the comparison of CD4+, CD3+, CD4+/CD8+, and NK cells after treatment was significantly different and higher in the treatment group than in the comparison group. The effective rate of 98.00% in the treatment group was significantly higher than that of 76.00% in the comparison group, and the difference was statistically significant. The clinical efficacy of the two groups showed that the fever, cough, sputum, myasthenia gravis crisis, and gastrointestinal reactions in the treatment group were significantly lower than those in the comparison group. The study indicates that comparative study of children with oculomotor myasthenia gravis treated with immunoglobulin combined with glucocorticoids is more effective, effectively improving the immune level of patients and reducing adverse reactions.
Collapse
Affiliation(s)
- Lijun Fan
- Pediatrics, Hubei Suizhou Central Hospital, Suizhou, Hubei 441300, China
| | - Yahui Yang
- Pediatrics, Wuhan Hankou Hospital, Wuhan, Hubei 430012, China
| | - Fan Zhang
- Pediatrics, Hubei Suizhou Maternal and Child Health Hospital, Suizhou, Hubei 441300, China
| | - Fei Huang
- Pediatrics, Hubei Suizhou Central Hospital, Suizhou, Hubei 441300, China
| |
Collapse
|
29
|
Lambert C, Scohy A, Yombi JC, Goffin E, Devresse A. Impact of therapeutic plasma exchange on acquired vaccinal anti-SARS-CoV-2 antibodies. Eur J Intern Med 2022; 100:140-142. [PMID: 35181184 PMCID: PMC8841167 DOI: 10.1016/j.ejim.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Catherine Lambert
- Department of Hematology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| | - Anais Scohy
- Department of Microbiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean Cyr Yombi
- Department of Internal Medicine and Infectious Disease, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Eric Goffin
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arnaud Devresse
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
30
|
Menon D, Bril V. Pharmacotherapy of Generalized Myasthenia Gravis with Special Emphasis on Newer Biologicals. Drugs 2022; 82:865-887. [PMID: 35639288 PMCID: PMC9152838 DOI: 10.1007/s40265-022-01726-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/20/2022]
Abstract
Myasthenia gravis (MG) is a chronic, fluctuating, antibody-mediated autoimmune disorder directed against the post-synaptic neuromuscular junctions of skeletal muscles, resulting in a wide spectrum of manifestations ranging from mild to potentially fatal. Given its unique natural course, designing an ideal trial design for MG has been wrought with difficulties and evidence in favour of several of the conventional agents is weak as per current standards. Despite this, acetylcholinesterases and corticosteroids have remained the cornerstones of treatment for several decades with intravenous immunoglobulins (IVIG) and therapeutic plasma exchange (PLEX) offering rapid treatment response, especially in crises. However, the treatment of MG entails long-term immunosuppression and conventional agents are viable options but take longer to act and have a number of class-specific adverse effects. Advances in immunology, translational medicine and drug development have seen the emergence of several newer biological agents which offer selective, target-specific immunotherapy with fewer side effects and rapid onset of action. Eculizumab is one of the newer agents that belong to the class of complement inhibitors and has been approved for the treatment of refractory general MG. Zilucoplan and ravulizumab are other agents in this group in clinical trials. Neisseria meningitis is a concern with all complement inhibitors, mandating vaccination. Neonatal Fc receptor (FcRn) inhibitors prevent immunoglobulin recycling and cause rapid reduction in antibody levels. Efgartigimod is an FcRn inhibitor recently approved for MG treatment, and rozanolixizumab, nipocalimab and batoclimab are other agents in clinical trial development. Although lacking high quality evidence from randomized clinical trials, clinical experience with the use of anti-CD20 rituximab has led to its use in refractory MG. Among novel targets, interleukin 6 (IL6) inhibitors such as satralizumab are promising and currently undergoing evaluation. Cutting-edge therapies include genetically modifying T cells to recognise chimeric antigen receptors (CAR) and chimeric autoantibody receptors (CAAR). These may offer sustained and long-term remissions, but are still in very early stages of evaluation. Hematopoietic stem cell transplantation (HSCT) allows immune resetting and offers sustained remission, but the induction regimens often involve serious systemic toxicity. While MG treatment is moving beyond conventional agents towards target-specific biologicals, lack of knowledge as to the initiation, maintenance, switching, tapering and long-term safety profile necessitates further research. These concerns and the high financial burden of novel agents may hamper widespread clinical use in the near future.
Collapse
Affiliation(s)
- Deepak Menon
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Vera Bril
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, 5EC-309, Toronto General Hospital, University of Toronto, 200 Elizabeth St, Toronto, M5G 2C4, Canada.
| |
Collapse
|
31
|
Katsumoto TR, Wilson KL, Giri VK, Zhu H, Anand S, Ramchandran KJ, Martin BA, Yunce M, Muppidi S. Plasma Exchange for Severe Immune-Related Adverse Events from Checkpoint Inhibitors: An Early Window of Opportunity? IMMUNOTHERAPY ADVANCES 2022; 2:ltac012. [PMID: 35814850 PMCID: PMC9257781 DOI: 10.1093/immadv/ltac012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of several advanced malignancies leading to durable remission in a subset of patients. Their rapidly expanding use has led to an increased frequency of immune-related adverse events (irAEs). The pathogenesis of irAEs is poorly understood but may involve aberrant activation of T cells leading to inflammatory cytokine release or production of pathogenic antibodies leading to organ damage. Severe irAEs can be extremely debilitating and, in some cases, life threatening. IrAEs may not always be corticosteroid responsive or may require excessively high, often toxic, corticosteroid doses. Therapeutic plasma exchange (PLEX) is a treatment modality that has shown promising results for the management of certain severe irAEs, including irAEs that are not mentioned in current treatment guidelines. PLEX may attenuate ongoing irAEs and prevent delayed irAEs by accelerating clearance of the ICI, or by acutely removing pathogenic antibodies, cytokines, and chemokines. Here, we summarize examples from the literature in which PLEX was successfully used for the treatment of irAEs. We posit that timing may be a critical factor and that earlier utilization of PLEX for life-threatening irAEs may result in more favorable outcomes. In individuals at high risk for irAEs, the availability of PLEX as a potential therapeutic mitigation strategy may encourage life-saving ICI use or rechallenge. Future research will be critical to better define which indications are most amenable to PLEX, particularly to establish the optimal place in the sequence of irAE therapies and to assess the ramifications of ICI removal on cancer outcomes.
Collapse
Affiliation(s)
- Tamiko R Katsumoto
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine , Stanford, CA, USA
| | - Kalin L Wilson
- Department of Medicine, Stanford University School of Medicine, Stanford , CA, USA
| | - Vinay K Giri
- Department of Medicine, Stanford University School of Medicine, Stanford , CA, USA
| | - Han Zhu
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine , Stanford, CA, USA
| | - Shuchi Anand
- Department of Medicine, Division of Nephrology, Stanford University School of Medicine , Stanford, CA, USA
| | - Kavitha J Ramchandran
- Department of Medicine, Division of Oncology, Stanford Cancer Institute , Stanford University School of Medicine, Stanford, CA, USA
| | - Beth A Martin
- Department of Medicine, Division of Hematology, Stanford University School of Medicine , Stanford, CA, USA
| | - Muharrem Yunce
- Department of Pathology, Stanford University School of Medicine, Stanford , CA, USA
| | - Srikanth Muppidi
- Department of Neurology and Neurosciences, Stanford University School of Medicine, Stanford , CA, USA
| |
Collapse
|
32
|
Ouyang S, Yin W, Zeng Q, Li B, Zhang J, Duan W, Li Y, Liang Y, Wang J, Tan H, Yang H. Lymphoplasma Exchange Improves Myasthenia Gravis Exacerbations: A Retrospective Study in a Chinese Center. Front Immunol 2022; 13:757841. [PMID: 35514988 PMCID: PMC9063637 DOI: 10.3389/fimmu.2022.757841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
Background Lymphoplasma exchange (LPE), a technique combining plasma exchange with leukapheresis, is emerging as promising treatment for autoimmune diseases. Data on the efficacy and safety of LPE in myasthenia gravis (MG) therapy are scarce. In this study, we aimed to comprehensively review the clinical efficacy, safety, and immunological characteristics of LPE therapy in MG patients. Study Design and Methods A Chinese cohort of 276 generalized MG patients in state of exacerbation, including impeding crisis, myasthenia crisis, and preparation for thoracic exsection between January 2014 and December 2020, were evaluated in this study. Results A total of 276 patients with a median age of 45.5 ± 16.7 years underwent a total of 635 LPE sessions. Clinical scales of Quantitative Myasthenia Gravis (QMG) scores, Myasthenia Gravis Specific Manual Muscle Testing (MMT) scores, activities of daily living (ADL) scores, and quality of life (QOL) scores were improved during 4 weeks’ follow-up. Adverse effects occurred in 20 out of 276 patients, with 14 patients having one adverse event each. Independent predictive factors for good response to LPE therapy were symptom onset before LPE therapy ≤3 days and age on LPE therapy <50 years of age. LPE decreased the serum levels of antibodies, immunoglobulins, and complements 4 weeks after the first replacement, with decreased levels of interleukin (IL)-17A and interferon (IFN)-γ and increased level of IL-10. Conclusion LPE is an effective treatment for MG patients in state of exacerbation and preparation for thymectomy. Early use of LPE on early-onset MG may have good therapeutic effects. The potential mechanism for LPE is the polarization of cytokines from IL-17A, IFN-γ, into IL-10.
Collapse
Affiliation(s)
- Song Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Medical Center of Neurology, The First Hospital of Changsha City, South China University, Changsha, China
| | - Weifan Yin
- The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Huan Yang, ; Weifan Yin,
| | - Qiuming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bijuan Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Zhang
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Weiwei Duan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Liang
- Medical Center of Neurology, The First Hospital of Changsha City, South China University, Changsha, China
| | - Jiaqi Wang
- Medical Center of Neurology, The First Hospital of Changsha City, South China University, Changsha, China
| | - Hong Tan
- Medical Center of Neurology, The First Hospital of Changsha City, South China University, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Huan Yang, ; Weifan Yin,
| |
Collapse
|
33
|
Therapeutic Effects of Batoclimab in Chinese Patients with Generalized Myasthenia Gravis: A Double-Blinded, Randomized, Placebo-Controlled Phase II Study. Neurol Ther 2022; 11:815-834. [PMID: 35412216 PMCID: PMC9095773 DOI: 10.1007/s40120-022-00345-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction We investigated the safety and explore potential efficacy of batoclimab administered subcutaneously in Chinese patients with generalized myasthenia gravis (gMG). Methods A randomized, double-blinded, placebo-controlled, parallel phase II study was conducted. First, in the double-blinded treatment period, eligible patients received batoclimab (680 mg), batoclimab (340 mg), or placebo on days 1, 8, 15, 22, 29, and 36. In the open-label treatment period, patients received batoclimab (340 mg) on days 50, 64, and 78. In the follow-up period, patients were examined on days 92, 106, and 120. The primary endpoint was Myasthenia Gravis Activities of Daily Living (MG-ADL) score change on day 43 from baseline. Results In total, 30 eligible patients were enrolled, with 11, 10, and 9 patients in the batoclimab 680 mg, batoclimab 340 mg, and placebo groups, respectively. MG-ADL score changes from baseline to day 43 were −2.2 ± 0.9, −4.7 ± 0.6, and −4.4 ± 1.0 in the placebo, batoclimab 340 mg, and 680 mg groups, respectively. Similar changes were observed in Quantitative Myasthenia Gravis, Myasthenia Gravis Composite, and 15-item Myasthenia Gravis Quality of Life scores in the placebo, batoclimab 340 mg, and 680 mg groups, respectively. The proportion of patients with clinically significant improvement on day 43 was higher in the batoclimab groups. On day 120, all four scales in the placebo group had more significant improvement compared with the batoclimab groups, with total serum IgG levels reaching a plateau. No death or treatment-emergent adverse events (TEAEs) led to study discontinuation. Conclusion Batoclimab is effective and safe in Chinese patients with gMG. Trial Registration This study was registered at ClinicalTrials.gov (NCT04346888) on 15 April 2020, with the first patient enrolled on 23 July 2020. Supplementary Information The online version contains supplementary material available at 10.1007/s40120-022-00345-9.
Collapse
|
34
|
Abstract
Neuroimmunological diseases and their treatment compromise the immune system, thereby increasing the risk of infections and serious illness. Consequently, vaccinations to protect against infections are an important part of the clinical management of these diseases. However, the wide variety of immunotherapies that are currently used to treat neuroimmunological disease — particularly multiple sclerosis and neuromyelitis optica spectrum disorders — can also impair immunological responses to vaccinations. In this Review, we discuss what is known about the effects of various immunotherapies on immunological responses to vaccines and what these effects mean for the safe and effective use of vaccines in patients with a neuroimmunological disease. The success of vaccination in patients receiving immunotherapy largely depends on the specific mode of action of the immunotherapy. To minimize the risk of infection when using immunotherapy, assessment of immune status and exclusion of underlying chronic infections before initiation of therapy are essential. Selection of the required vaccinations and leaving appropriate time intervals between vaccination and administration of immunotherapy can help to safeguard patients. We also discuss the rapidly evolving knowledge of how immunotherapies affect responses to SARS-CoV-2 vaccines and how these effects should influence the management of patients on these therapies during the COVID-19 pandemic. In this Review, the authors discuss how various immunotherapies for neuroimmunological diseases interact with vaccination responses, including responses to SARS-CoV-2 vaccinations, and the implications for the safe and effective use of vaccines in patients with these diseases. Vaccination against infection is an essential part of the management of neuroimmunological diseases. All indicated vaccinations should be administered before initiation of immunotherapy whenever possible; appropriate intervals between vaccination and treatment vary with treatment and vaccination. Inactivated vaccines are considered safe in neuroimmunological diseases but live vaccines are generally contraindicated during immunotherapy. Vaccination responses during immunotherapy can be diminished or abrogated, depending on the treatment and vaccination; antibody titre testing to monitor responses can be considered where appropriate. Vaccinations must be avoided during relapses or exacerbations of neuroimmunological diseases. Vaccination against SARS-CoV-2 is recommended for patients with neuroimmunological disease but some immunotherapies limit the immune response; therefore, timing should be considered carefully.
Collapse
|
35
|
Alhaidar MK, Abumurad S, Soliven B, Rezania K. Current Treatment of Myasthenia Gravis. J Clin Med 2022; 11:jcm11061597. [PMID: 35329925 PMCID: PMC8950430 DOI: 10.3390/jcm11061597] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Myasthenia gravis (MG) is the most extensively studied antibody-mediated disease in humans. Substantial progress has been made in the treatment of MG in the last century, resulting in a change of its natural course from a disease with poor prognosis with a high mortality rate in the early 20th century to a treatable condition with a large proportion of patients attaining very good disease control. This review summarizes the current treatment options for MG, including non-immunosuppressive and immunosuppressive treatments, as well as thymectomy and targeted immunomodulatory drugs.
Collapse
|
36
|
Verschuuren JJGM, Palace J, Murai H, Tannemaat MR, Kaminski HJ, Bril V. Advances and ongoing research in the treatment of autoimmune neuromuscular junction disorders. Lancet Neurol 2022; 21:189-202. [DOI: 10.1016/s1474-4422(21)00463-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/16/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
|
37
|
Wolfe GI, Ward ES, de Haard H, Ulrichts P, Mozaffar T, Pasnoor M, Vidarsson G. IgG regulation through FcRn blocking: A novel mechanism for the treatment of myasthenia gravis. J Neurol Sci 2021; 430:118074. [PMID: 34563918 DOI: 10.1016/j.jns.2021.118074] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
The neonatal Fc receptor (FcRn) is an MHC class I-like molecule that is widely distributed in mammalian organs, tissues, and cells. FcRn is critical to maintaining immunoglobulin G (IgG) and albumin levels through rescuing these molecules from lysosomal degradation. IgG autoantibodies are associated with many autoimmune diseases, including myasthenia gravis (MG), a rare neuromuscular autoimmune disease that causes debilitating and, in its generalized form (gMG), potentially life-threatening muscle weakness. IgG autoantibodies are directly pathogenic in MG and target neuromuscular junction proteins, causing neuromuscular transmission failure. Treatment approaches that reduce autoantibody levels, such as therapeutic plasma exchange and intravenous immunoglobulin, have been shown to be effective for gMG patients but are not indicated as ongoing maintenance therapies and can be associated with burdensome side effects. Agents that block FcRn-mediated recycling of IgG represent a rational and promising approach for the treatment of gMG. Blocking FcRn allows targeted reduction of all IgG subtypes without decreasing concentrations of other Ig isotypes; therefore, FcRn blocking could be a safe and effective treatment strategy for a broad population of gMG patients. Several FcRn-blocking antibodies and one antibody Fc fragment have been developed and are currently in various stages of clinical development. This article describes the mechanism of FcRn blockade as a novel approach for IgG-mediated disease therapy and reviews promising clinical data using such FcRn blockers for the treatment of gMG.
Collapse
Affiliation(s)
- Gil I Wolfe
- Department of Neurology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, SUNY, Buffalo, NY, USA.
| | - E Sally Ward
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, SO16 6YD, UK
| | - Hans de Haard
- argenx, Zwijnaarde, Belgium, University of California, Irvine, CA, USA
| | - Peter Ulrichts
- argenx, Zwijnaarde, Belgium, University of California, Irvine, CA, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California, Irvine, CA, USA
| | - Mamatha Pasnoor
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gestur Vidarsson
- Sanquin Research, and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
38
|
Lledo-Garcia R, Dixon K, Shock A, Oliver R. Pharmacokinetic-pharmacodynamic modelling of the anti-FcRn monoclonal antibody rozanolixizumab: Translation from preclinical stages to the clinic. CPT Pharmacometrics Syst Pharmacol 2021; 11:116-128. [PMID: 34735735 PMCID: PMC8752106 DOI: 10.1002/psp4.12739] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/15/2023] Open
Abstract
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.
Collapse
|
39
|
Iijima S. Clinical and pathophysiologic relevance of autoantibodies in neonatal myasthenia gravis. Pediatr Neonatol 2021; 62:581-590. [PMID: 34272198 DOI: 10.1016/j.pedneo.2021.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
Between 10% and 20% of neonates born to mothers with myasthenia gravis (MG) develop neonatal MG due to the transfer of maternal autoantibodies across the placenta. Neonatal MG can occur in infants born not only from mothers with acetylcholine receptor (AChR) or muscle-specific tyrosine kinase (MuSK) antibodies but also from mothers without detectable muscle antibodies. The low incidence rate may be due to specific autoantibody characteristics that differ among individuals, but a genetic predisposition in some infants is possible. The majority of reported neonatal MG cases are anti-AChR antibody-positive (AChR-MG), and a high anti-fetal/anti-adult AChR titer ratio in the mother is predictive of its occurrence. However, patients with anti-MuSK antibody-positive MG (MuSK-MG) are more likely to experience exacerbations during pregnancy and have a higher probability of developing neonatal MG than AChR-MG patients. Moreover, maternal MuSK-MG may be associated with early-onset and more severe manifestations of neonatal MG. Although cholinesterase inhibitors have been effectively used for treating neonatal AChR-MG, neonatal MuSK-MG may be more difficult to treat with this type of medication. Maternal MuSK-MG usually greatly benefits from intravenous immunoglobulin (IVIG) and plasma exchange. In neonatal MG, IVIG is considered for severely affected infants with MuSK-MG, but the efficacy of IVIG remains unclear. Although exchange transfusion may be a management adjunct, its clinical benefits are controversial. As the therapy-induced reduction of autoantibodies may be advantageous for fetal outcomes, maternal MG should be effectively treated during pregnancy. However, caution of drug contraindication during pregnancy and lactation must be exercised to avoid unwanted effects for the fetus and neonate. In the future, MG caused by anti-lipoprotein receptor-related protein 4 or other antibodies might be also identified in pregnant women and neonates. Therefore, the determination of autoantibody specificity is essential for successful management.
Collapse
Affiliation(s)
- Shigeo Iijima
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan.
| |
Collapse
|
40
|
Yeaman MR. Immunosuppression in Glomerular Diseases: Implications for SARS-CoV-2 Vaccines and COVID-19. GLOMERULAR DISEASES 2021; 1:277-293. [PMID: 34935004 PMCID: PMC8678218 DOI: 10.1159/000519182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/19/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Glomerular diseases (GD) are chronic conditions that often involve immune dysfunction and require immunosuppressive therapy (IST) to control underlying pathogenesis. Unfortunately, such diseases appear to heighten risks of severe outcomes in COVID-19 and predispose to other infections that may be life-threatening. Thus, averting preventable infections is imperative in GD patients. SUMMARY The advent of vaccines demonstrated to be safe and efficacious against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has favorably impacted the COVID-19 pandemic epidemiology. However, patients on ISTs were excluded from initial vaccine clinical trials. Thus, only limited and incomplete data are available currently regarding the potential impact of immunosuppression on immune response to or efficacy of the SARS-CoV-2 vaccines. However, new insights are emerging from SARS-CoV-2 vaccine studies, and impacts of ISTs on conventional vaccines are useful to consider. Mechanisms of immunosuppressive agents commonly used in the treatment of GD are reviewed with respect to implications for immune responses induced by SARS-CoV-2 vaccines. ISTs discussed include corticosteroids; alkylating agents; antimetabolites; calcineurin or mammalian target of rapamycin inhibitors; CD38+, CD20+, or CD19+ cell depletion; and complement protein C5 inhibition. KEY MESSAGES Many immunosuppressive therapies may potentially attenuate or impair protective immunity of the SARS-CoV-2 vaccines. However, as vaccines currently in use employ mRNA or nonreplicative viral vectors, they appear to be safe in patients on immunosuppression, further favoring vaccination. Moreover, predominant SARS-CoV-2 vaccines are likely to afford at least partial protective immunity through one or more immune mechanisms even in patients on IST. Guidelines and emerging strategies are also considered to optimize vaccine protection from COVID-19.
Collapse
Affiliation(s)
- Michael R. Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Divisions of Molecular Medicine & Infectious Diseases, Harbor-UCLA Medical Center, Torrance, California, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| |
Collapse
|
41
|
Ipe TS, Davis AR, Raval JS. Therapeutic Plasma Exchange in Myasthenia Gravis: A Systematic Literature Review and Meta-Analysis of Comparative Evidence. Front Neurol 2021; 12:662856. [PMID: 34531809 PMCID: PMC8439193 DOI: 10.3389/fneur.2021.662856] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/12/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Patients with Myasthenia Gravis (MG) can be treated acutely with therapeutic plasma exchange (TPE) or intravenous immune globulin (IVIG). To date, there is no definitive understanding of which of the two treatments is more effective and safer. The purpose of this study was to systematically review the literature on the comparative efficacy and safety of TPE to other available treatments for MG. Methods: A systematic literature search for studies published between 1997 and 2017 was performed per Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines using two database sources, MEDLINE (through the PubMed database) and Cochrane Library. Results: The search strategy resulted in 535 articles whose abstracts were reviewed. Among these, 165 full texts articles were reviewed for eligibility and 101 articles were excluded. Of the 165 articles, 64 articles were included for a systematic literature and 11 articles for a meta-analysis. Conclusions: This systematic literature review and meta-analysis of treatment options showed that there was a higher response rate with TPE than IVIG in acute MG patients and patients undergoing thymectomy. There was no difference in mortality between the two treatment options. Our findings highlight the need for additional randomized clinical trials in these patients with MG.
Collapse
Affiliation(s)
- Tina S. Ipe
- Department of Pathology and Laboratory Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Adeola R. Davis
- Terumo Blood and Cell Technologies, Lakewood, CO, United States
| | - Jay S. Raval
- Department of Pathology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
42
|
Menon D, Urra Pincheira A, Bril V. Emerging drugs for the treatment of myasthenia gravis. Expert Opin Emerg Drugs 2021; 26:259-270. [PMID: 34228579 DOI: 10.1080/14728214.2021.1952982] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Advances in understanding the immune pathomechanisms in myasthenia gravis (MG) allow for the development of novel targeted immune therapies. By working at specific points in the immunopathogenesis, these agents have the potential to provide rapid and efficacious responses compared to conventional immunosuppressive therapy (IST), addressing unmet needs and consequently are a research priority.Areas covered: This paper reviews the advances in MG treatment modalities with their scientific rationale. A search of clinicaltrials.gov and a literature search of PubMed from January 2015 to the end of June 2021 was done using the search terms: MG, treatment, immune targets to obtain information on recent developments of complement inhibitors, FcRn receptor inhibitors, direct and indirect B cell inhibitors, CAR and CAAR- T cell therapy, and hematopoietic stem cell transplantation. Specific agents in various phases of clinical development, evidence from ongoing trials and potential roadblocks are examined.Expert opinion: Despite several promising novel agents, existing data as to the timing of initiation and duration of treatment, long-term safety profile and utility in certain patient subsets are limited and require further research. Despite these considerations, the future of MG treatment is transitioning from broad-spectrum IST toward precise, target-driven and personalized immunotherapy.
Collapse
Affiliation(s)
- Deepak Menon
- Department of Medicine, Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Canada
| | - Alejandra Urra Pincheira
- Department of Medicine, Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Canada
| | - Vera Bril
- Department of Medicine, Ellen & Martin Prosserman Centre for Neuromuscular Diseases, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
43
|
Sheckley H, Malhotra K, Katyal N, Narula N, Govindarajan R. Clinical experience with maintenance therapeutic plasma exchange in refractory generalized myasthenia gravis. J Clin Apher 2021; 36:727-736. [PMID: 34241920 DOI: 10.1002/jca.21923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Despite the availability of several immunomodulatory therapies, about 20% of myasthenia gravis (MG) patients remain refractory to conventional treatments. There is limited evidence to support the use of maintenance therapeutic plasma exchange (TPE) therapy for refractory generalized MG. METHODS Retrospective chart review of 14 patients with refractory generalized MG treated for 12 months with maintenance TPE therapy. Outcome measures were myasthenia gravis composite (MGC) score, myasthenia gravis activities of daily living (MG-ADL), number of acute exacerbations, medication changes, and adverse events. Data were collected at 3 monthly intervals for 12 months before and after initiation of TPE therapy. RESULTS Clinically meaningful reductions in mean MG-ADL (>2 points) (mean MG-ADL score: 9.9 ± 0.5; 12-month pre-TPE to 5.2 ± 0.9; 12-month post-TPE) and MGC (>3 points) (mean MGC score: 25.2 ± 1.6; 12-month pre-TPE to 11.7 ± 1.4; 12-month post-TPE) were observed at 3 months following initiation of TPE and were maintained up to 12 months in all patients. After 12 months of TPE therapy, all patients had a significant reduction in daily prednisone and pyridostigmine use. Patients previously on IVIG or rituximab therapy were successfully weaned off both treatments. There was a significant reduction in acute MG exacerbations; 7.8 ± 1.1 mean exacerbations/patient (12-month pre-TPE) to 2 ± 1.1 mean exacerbations/patient (12-month post-TPE). CONCLUSION Over a period of 12 months, maintenance TPE therapy improved MG-ADL, and MGC with decreased immunosuppressant requirement, while being well-tolerated.
Collapse
Affiliation(s)
- Hunter Sheckley
- Department of Neurology, University of Missouri, Columbia, Missouri, USA
| | - Kunal Malhotra
- Division of Nephrology, Department of Internal Medicine, University of Missouri, Columbia, Missouri, USA
| | - Nakul Katyal
- Department of Neurology, University of Missouri, Columbia, Missouri, USA
| | - Naureen Narula
- Department of Pulmonology and Critical Care Medicine, Staten Island University Hospital, Staten Island, New York, USA
| | | |
Collapse
|
44
|
Ching J, Richards D, Lewis RA, Li Y. Myasthenia gravis exacerbation in association with antibody overshoot following plasmapheresis. Muscle Nerve 2021; 64:483-487. [PMID: 34076268 DOI: 10.1002/mus.27341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/11/2022]
Abstract
INTRODUCTION/AIM Antibody overshoot following therapeutic plasmapheresis (PLEX) is defined by subsequent increase in antibody to levels exceeding those prior to removal. It has been infrequently described in the past, and its influence on the clinical course of myasthenia gravis (MG) remains unclear. METHODS This was a retrospective analysis of five patients with generalized MG treated with PLEX. RESULTS All five patients possessed antibodies against acetylcholine receptor (AChR-Ab). After undergoing 3 to 12 PLEX treatment sessions, AChR-Ab titer increased to a median of 1292% of the baseline level. The median interval from the last PLEX session to peak AChR-Ab detection was 6 wk. In four patients, AChR-Ab overshoot was associated with a clinical deterioration. DISCUSSION The AChR-Ab overshoot may occur following PLEX. In patients who deteriorate following PLEX treatment, the presence of antibody overshoot may serve as additional guidance for treatment adjustment.
Collapse
Affiliation(s)
- Jason Ching
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Danielle Richards
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard A Lewis
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yuebing Li
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Al-Ahmer I, Elshony H. Determinants of quality of life changes with plasmapheresis in patients with myasthenia gravis. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00320-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Immunomodulation, including IVIG and plasma exchange, is useful for a crisis or severe exacerbation. Plasma exchange may be slightly faster and more effective in a myasthenic crisis than IVIG. The aim of the current study was to determine the changes in the quality of life (QOL) after plasmapheresis and factors influencing these changes.
Results
This study was conducted on 98 MG patients diagnosed as moderate to severe myasthenia gravis (according to Myasthenia Gravis Foundation of America classification), 81 patients received alternate day 5 sessions plasmapheresis (TPE group) and 17 patients were on medical treatment only (control group). All patients were subjected to full history, through clinical neurological evaluation and scored with quantitative myasthenia gravis (QMG) score for MG severity at start and after 1 m. Both groups completed the QOL questionnaire at baseline and after 1 month. The MG-QOL-15 scores were computed and we analyzed the change in the QOL scores from baseline to after plasmapheresis groups and compared it with the results for the control group. The scores in QOL scales had significantly decreased after plasmapheresis, and the improvement in QOL scores had a good correlation with the decrease in QMGS. The improvement in QOL and QMG was significantly correlated with younger age, female gender, shorter duration of the illness, presence of AchR antibodies, antibody titer, and thymus hyperplasia.
Conclusion
Plasmapheresis is effective in improving quality of life in myasthenia gravis patients and this improvement influenced by age, gender, duration of illness, presence of AchR antibodies and their titer, and the thymus pathology.
Collapse
|
46
|
Attenuation of age-elevated blood factors by repositioning plasmapheresis: A novel perspective and approach. Transfus Apher Sci 2021; 60:103162. [PMID: 34083162 DOI: 10.1016/j.transci.2021.103162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aging is associated with the impairment of stem cell activation, leading to the functional decline of tissues and increasing the risk for age-associated diseases. The old, damaged or unrepaired tissues disturb distant tissue homeostasis by secreting factors into the circulation, which may not only serve as biomarkers for specific age-associated pathologies but also induce a variety of degenerative phenotypes. In this review, we summarize and discuss systemic determinants that perpetuate age-related tissue dysfunction. We further elaborate on the effects of attenuating these circulating factors by highlighting recent advances which utilize plasmapheresis in a pre-clinical or clinical setting. Overall, we postulate that repositioning therapeutic plasma exchange (TPE) to dilute the systemic factors, which become deleterious at their age-elevated levels, could be a rapidly effective rejuvenation therapy that recalibrates crucial signaling pathways to a youthful state.
Collapse
|
47
|
Ke L, Li Q, Song J, Jiao W, Ji A, Chen T, Pan H, Song Y. The mitochondrial biogenesis signaling pathway is a potential therapeutic target for myasthenia gravis via energy metabolism (Review). Exp Ther Med 2021; 22:702. [PMID: 34007311 PMCID: PMC8120506 DOI: 10.3892/etm.2021.10134] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Myasthenia gravis (MG) is an autoantibody-mediated autoimmune disease that is characterized by muscle weakness and fatigue. Traditional treatments for MG target the neuromuscular junction (NMJ) or the immune system. However, the efficacy of such treatments is limited, and novel therapeutic options for MG are urgently required. In the current review, a new therapeutic strategy is proposed based on the mitochondrial biogenesis and energy metabolism pathway, as stimulating mitochondrial biogenesis and the energy metabolism might alleviate myasthenia gravis. A number of cellular sensors of the energy metabolism were investigated, including AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1). AMPK and SIRT1 are sensors that regulate cellular energy homeostasis and maintain energy metabolism by balancing anabolism and catabolism. Peroxisome proliferator-activated receptor γ coactivator 1α and its downstream transcription factors nuclear respiratory factors 1, nuclear respiratory factors 2, and transcription factor A are key sensors of mitochondrial biogenesis, which can restore mitochondrial DNA and produce new mitochondria. These processes help to control muscle contraction and relieve the symptoms of MG, including muscle weakness caused by dysfunctional NMJ transmission. Therefore, the present review provides evidence for the therapeutic potential of targeting mitochondrial biogenesis for the treatment of MG.
Collapse
Affiliation(s)
- Lingling Ke
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Qing Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Jingwei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Wei Jiao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Aidong Ji
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yafang Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China.,Institute of Pi-Wei, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
48
|
Liu C, Liu P, Ma M, Yang H, Qi G. Efficacy and safety of double-filtration plasmapheresis treatment of myasthenia gravis: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25622. [PMID: 33907116 PMCID: PMC8084046 DOI: 10.1097/md.0000000000025622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 12/09/2020] [Accepted: 04/03/2021] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES To evaluate the efficacy of double-filtration plasmapheresis (DFPP) treatment of myasthenia gravis (MG) through a systematic review and meta-analysis. METHODS PubMed, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), Chinese Scientific Journals Database (VIP), and Wanfang databases were searched for randomized controlled trials (RCTs) and clinical controlled trials (CCTs) on DFPP for MG from database establishment to June 2019. Two researchers independently screened the articles, extracted the data, and cross checked the results. RevMan 5.3 was used for statistical analyses. RESULTS Seven RCTs and 2 CCTs were found comprising 329 patients. The results showed that clinical MG remission rate after DFPP treatment was significantly higher (OR = 4.33; 95% confidence interval [CI], 1.97-9.53; P < .001) and the serum levels of antititin antibody was significantly decreased (standardized mean difference [SMD] = 9.30; 95% CI, 7.51-11.08; P < .001). In addition, the quantitative MG (QMG) score, hospital stay and time to remission of MG symptoms, and acetylcholine receptor antibody (AchRAb) decreased in the DFPP treatment group; however, these outcomes had high heterogeneity among the studies. Only one study has reported on the adverse effects, including hypotension and hematoma. CONCLUSION This meta-analysis suggests that DFPP can be recommended for the short-term mitigation of MG. Because our review was limited by the quantity and quality of the included studies, the above conclusions should be verified by additional high-quality studies.
Collapse
|
49
|
Anesi JA, Lautenbach E, Tamma PD, Thom KA, Blumberg EA, Alby K, Bilker WB, Werzen A, Tolomeo P, Omorogbe J, Pineles L, Han JH. Risk Factors for Extended-Spectrum β-lactamase-Producing Enterobacterales Bloodstream Infection Among Solid-Organ Transplant Recipients. Clin Infect Dis 2021; 72:953-960. [PMID: 32149327 DOI: 10.1093/cid/ciaa190] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Approximately 40% of all Enterobacterales (EB) bloodstream infections (BSIs) among solid organ transplant recipients (SOTRs) are due to extended-spectrum β-lactamase (ESBL)-producing organisms, but risk factors for such infections remain ill defined in this population. We sought to determine the risk factors for ESBL-EB BSIs among SOTRs. METHODS A multicenter case-control study was performed. All SOTRs with an EB BSI at the Hospital of the University of Pennsylvania and University of Maryland Medical Center between 1 January 2007 and 30 June 2018 and at The Johns Hopkins Hospital between 1 January 2005 and 31 December 2015 were included. Cases were those with an ESBL-EB BSI. Controls were those with a non-ESBL-EB BSI. Multivariable logistic regression was performed to determine risk factors for ESBL-EB BSI. RESULTS There were 988 episodes of EB BSI, of which 395 (40%) were due to an ESBL-EB. On multivariable analysis, the independent risk factors for ESBL-EB BSI included: ESBL-EB on prior culture (aOR, 12.75; 95% CI, 3.23-50.33; P < .001), a corticosteroid-containing immunosuppression regimen (aOR 1.30; 95% CI 1.03-1.65; P = .030), acute rejection treated with corticosteroids (aOR 1.18; 95% CI 1.16-1.19; P < .001), and exposure to third-generation cephalosporins (aOR 1.95; 95% CI 1.48-2.57; P < .001), echinocandins (aOR 1.61; 95% CI 1.08-2.40; P = .020), and trimethoprim-sulfamethoxazole (aOR 1.35; 95% CI 1.10-1.64; P = .003). CONCLUSIONS We identified several novel risk factors that are uniquely important to the SOTR population, including exposure to trimethoprim-sulfamethoxazole and corticosteroid-containing immunosuppressive regimens. Further studies exploring these associations and testing interventions aimed at these modifiable risk factors among SOTRs are needed.
Collapse
Affiliation(s)
- Judith A Anesi
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ebbing Lautenbach
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pranita D Tamma
- Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kerri A Thom
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Emily A Blumberg
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kevin Alby
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Warren B Bilker
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alissa Werzen
- Division of Infectious Diseases, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Pam Tolomeo
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacqueline Omorogbe
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lisa Pineles
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
50
|
Doneddu PE, Spina E, Briani C, Fabrizi GM, Manganelli F, Nobile-Orazio E. Acute and chronic inflammatory neuropathies and COVID-19 vaccines: Practical recommendations from the task force of the Italian Peripheral Nervous System Association (ASNP). J Peripher Nerv Syst 2021; 26:148-154. [PMID: 33620123 DOI: 10.1111/jns.12435] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS To develop recommendations for vaccination for coronavirus-19 (COVID-19) in patients with inflammatory neuropathies. METHODS Key questions were formulated in order to perform a literature review on the safety and efficacy of vaccines in patients with inflammatory neuropathies. Based on the best evidence and expert opinion, a list of recommendations was formulated to inform decision on vaccination for COVID-19 in patients with inflammatory neuropathies and increase adherence to vaccination programmes. RESULTS Recommendations addressing safety and efficacy of vaccination in patients with inflammatory neuropathies were formulated. No data are currently available on the safety and efficacy of COVID-19 vaccines in patients with inflammatory neuropathies or other immune-mediated conditions. There is only sparse data on the safety of previous available vaccines in patients with inflammatory neuropathies, but studies on other autoimmune disorders indicate that these are safe and mostly efficacious. Patients with inflammatory neuropathies might be at increased risk for severe illness from COVID-19. INTERPRETATION Patients with inflammatory neuropathies should be encouraged to adhere to the vaccination campaign for COVID-19. These recommendations provide guidance on the management of vaccinations for COVID-19 in patients with inflammatory neuropathies. More research is needed regarding the safety and efficacy of vaccination in patients with inflammatory neuropathies and other immune conditions.
Collapse
Affiliation(s)
- Pietro E Doneddu
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Emanuele Spina
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Chiara Briani
- Neurology Unit, Department of Neuroscience, University of Padova, Padova, Italy
| | - Gian Maria Fabrizi
- Neurology Unit, Department of Neuroscienze, University of Verona, Policlinico Hospital G.B. Rossi, Verona, Italy
| | - Fiore Manganelli
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Eduardo Nobile-Orazio
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, Milan University, Milan, Italy
| | | |
Collapse
|