1
|
Min YG, Visentin A, Briani C, Rajabally YA. Neuropathy with anti-myelin-associated glycoprotein antibodies: update on diagnosis, pathophysiology and management. J Neurol Neurosurg Psychiatry 2025; 96:340-349. [PMID: 39658134 DOI: 10.1136/jnnp-2024-334678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024]
Abstract
Antimyelin-associated glycoprotein (MAG) neuropathy is a rare autoimmune demyelinating peripheral neuropathy caused by IgM autoantibodies targeting MAG. The typical presentation is that of a slowly progressive, distal, length-dependent, predominantly sensory, sometimes ataxic neuropathy, frequently accompanied by upper limb tremor. Distal motor weakness may subsequently occur. The clinical presentation may vary and rarely be consistent with that of typical chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), as well as have an aggressive and rapidly disabling course. The diagnosis of anti-MAG neuropathy is based on the detection of anti-MAG antibodies through ELISA or western blot analysis, primarily in presence of an IgM monoclonal gammopathy. Anti-MAG neuropathy may occur without or with haematological malignancy. Electrophysiology is characteristic of a predominantly distal demyelinating neuropathy. Intravenous immunoglobulins and plasma exchange have unproven benefits, but may provide short-term effects. Cytotoxic therapies are commonly used, although without an evidence base. Rituximab, an anti-B-cell monoclonal antibody was studied in two randomised controlled trials, neither of which achieved their primary outcome. However, a meta-analysis of these two studies demonstrated improvement of disability at 8-12 months. A recent trial with lenalidomide was interrupted prematurely due to a high rate of venous thromboembolism. There are currently two ongoing trials with Bruton's tyrosine kinase inhibitors. Symptom control is otherwise frequently needed. Outcome measures used for other inflammatory neuropathies present limitations in anti-MAG neuropathy. International registries such as the planned IMAGiNe study may, in future, provide answers to the many remaining questions.
Collapse
Affiliation(s)
- Young Gi Min
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, The Republic of Korea
| | - Andrea Visentin
- Haematology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Chiara Briani
- Department of Neurosciences, Neurology Unit, University of Padova, Padova, Italy
| | | |
Collapse
|
2
|
Kalmi G, Nguyen Y, Amarger S, Aubineau M, Bibes B, Blanchard-Delaunay C, Boccon-Gibod I, Bouillet L, Coppo P, Dalmas MC, Debord-Peguet S, Defendi F, Demoreuil C, Du-Thanh A, Gayet S, Hadjadj J, Jeandel PY, Launay D, Ly KH, Avoy CM, Niault M, Ollivier Y, Pelletier F, Porneuf M, Roos-Weil D, Fain O, Gobert D. Efficacy and Safety of Rituximab-Based Treatments in Angioedema With Acquired C1-Inhibitor Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:212-222. [PMID: 37844846 DOI: 10.1016/j.jaip.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Angioedema (AE) due to acquired C1-inhibitor (C1-INH) deficiency (AAE-C1-INH) is related to excessive consumption of C1-INH or to anti-C1-INH antibodies, and is frequently associated with lymphoproliferative syndromes or monoclonal gammopathies. Standard of care for prophylactic treatment in this condition is not established. Rituximab may be effective to prevent attacks, especially if the lymphoid hemopathy is controlled, but data are scarce. OBJECTIVE To evaluate efficacy of rituximab in AAE-C1-INH. METHODS A retrospective multicenter study was carried out in France, including patients with AAE-C1-INH treated with rituximab between April 2005 and July 2019. RESULTS Fifty-five patients with AAE-C1-INH were included in the study, and 23 of them had an anti-C1-INH antibody. A lymphoid malignancy was identified in 39 patients, and a monoclonal gammopathy in 9. There was no associated condition in 7 cases. Thirty patients received rituximab alone or in association with chemotherapy (n = 25). Among 51 patients with available follow-up, 34 patients were in clinical remission and 17 patients had active AE after a median follow-up of 3.9 years (interquartile range, 1.5-7.7). Three patients died. The presence of anti-C1-INH antibodies was associated with a lower probability of AE remission (hazard ratio, 0.29 [95% CI, 0.12-0.67]; P = .004). Relapse was less frequent in patients with lymphoma (risk ratio, 0.27 [95% CI, 0.09-0.80]; P = .019) and in patients treated with rituximab and chemotherapy (risk ratio, 0.31 [95% CI, 0.12-0.79]; P = .014). CONCLUSIONS Rituximab is an efficient and well-tolerated therapeutic option in AE, especially in lymphoid malignancies and in the absence of detectable anti-C1-INH antibodies.
Collapse
Affiliation(s)
- Galith Kalmi
- Internal Medicine Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France.
| | - Yann Nguyen
- Internal Medicine Department, Nord-Université Paris Cité, AP-HP, Hôpital Beaujon, Clichy-sous-Bois, France
| | - Stephanie Amarger
- Dermatology Department, University Hospital, Clermont-Ferrand, France
| | - Magali Aubineau
- Internal Medicine Department, Hospices Civils de Lyon, Lyon, France
| | - Beatrice Bibes
- Internal Medicine Department, Saint Grégoire Hospital, Rennes, France
| | | | - Isabelle Boccon-Gibod
- Internal Medicine Department, French National Reference Center for Angioedema (CREAK), Grenoble University Hospital, Grenoble, France
| | - Laurence Bouillet
- Internal Medicine Department, French National Reference Center for Angioedema (CREAK), Grenoble University Hospital, Grenoble, France; University Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, Grenoble, France; Internal Medicine Department, University Hospital La Tronche, Grenoble, France
| | - Paul Coppo
- Hematology Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France
| | | | | | - Federica Defendi
- Immunology Department, Grenoble University Hospital, Grenoble, France
| | - Claire Demoreuil
- Internal Medicine Department, La Cavale Blanche University Hospital, Brest, France
| | - Aurélie Du-Thanh
- Dermatology Department, Montpellier University Hospital, Montpellier, France
| | - Stephane Gayet
- Internal Medicine Department, La Timone University Hospital, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Jerôme Hadjadj
- Internal Medicine Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France
| | | | - David Launay
- Internal and Immunological Medicine Department, Lille Hospital, U1286-INFINITE-Institute for Translational Research in Inflammation, Lille University, INSERM F-59000, Lille, France
| | - Kim Heang Ly
- Internal Medicine Department, Dupuytren University Hospital, Limoges, France
| | - Chloé Mc Avoy
- Internal Medicine Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Mathilde Niault
- Hematology Department, Hôpital du Scorff-Lorient, Groupe Hospitalier Bretagne Sud, Lorient, France
| | - Yann Ollivier
- Medicine Department, Cote de Nacre University Hospital, Caen, France
| | - Fabien Pelletier
- Dermatology Department, Allergology Center, Besançon University Hospital, Besançon, France
| | - Marc Porneuf
- Hematology Department, Centre Hospitalier Yves le Foll, Saint-Brieuc, France (x)Hematology Department, Sorbonne Université, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Damien Roos-Weil
- Hematology Department, Centre Hospitalier Yves le Foll, Saint-Brieuc, France (x)Hematology Department, Sorbonne Université, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Olivier Fain
- Internal Medicine Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Delphine Gobert
- Internal Medicine Department, Sorbonne Université, AP-HP, Hôpital Saint Antoine, Paris, France
| |
Collapse
|
3
|
Min YG, Han HJ, Shin HY, Baek JG, Kim JS, Park KS, Baek SH, Yoo I, Huh SY, Kwon YN, Choi SJ, Kim SM, Hong YH, Sung JJ. Therapeutic Outcomes and Electrophysiological Biomarkers in Anti-Myelin-Associated Glycoprotein Neuropathy: A Multicenter Cohort Study in South Korea. J Clin Neurol 2024; 20:50-58. [PMID: 38179632 PMCID: PMC10782088 DOI: 10.3988/jcn.2023.0127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/04/2023] [Accepted: 06/07/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Unlike other immune-mediated neuropathies, anti-myelin-associated glycoprotein (MAG) neuropathy is often refractory to immunotherapy. It is necessary to compare the relative efficacies of various immunotherapies and develop objective biomarkers in order to optimize its clinical management. METHODS This study recruited 91 patients with high anti-MAG antibody titers from 7 tertiary hospitals in South Korea. We analyzed the baseline characteristics, therapeutic outcomes, and nerve conduction study (NCS) findings of 68 patients and excluded 23 false positive cases. RESULTS The rate of positive responses to treatment was highest using zanubrutinib (50%) and rituximab (36.4%), followed by corticosteroids (16.7%), immunosuppressants (9.5%), intravenous immunoglobulin (5%), and plasma exchange (0%). Disability and weakness were significantly associated with multiple NCS parameters at the time of diagnosis, especially distal compound muscle action potential (CMAP) amplitudes. Moreover, the longitudinal trajectory of the average CMAP amplitudes paralleled the clinical courses, with a 16.2 percentile decrease as an optimal cutoff for predicting a clinical exacerbation (area under the receiver operating characteristic curve=0.792). CONCLUSIONS Our study supports the use of NCS as an objective marker for estimating disease burden and tracking clinical changes in patients with anti-MAG neuropathy. We have described the beneficial effects of rituximab and a new drug, zanubrutinib, compared with conventional immunotherapies.
Collapse
Affiliation(s)
- Young Gi Min
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Hee-Jo Han
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Gyu Baek
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jun-Soon Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kyung-Seok Park
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Seol-Hee Baek
- Department of Neurology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Ilhan Yoo
- Department of Neurology, Nowon Eulji Medical Center, Seoul, Korea
| | - So-Young Huh
- Department of Neurology, Kosin Medical University Hospital, Busan, Korea
| | - Young Nam Kwon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Seok-Jin Choi
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Sung-Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Yoon-Ho Hong
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Neurology, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jung-Joon Sung
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
4
|
Stino AM, Elsheikh B, Allen JA. Anti-myelin-associated glycoprotein neuropathy: Where do we stand? Muscle Nerve 2023; 68:823-832. [PMID: 37602932 DOI: 10.1002/mus.27954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 08/22/2023]
Abstract
Myelin-associated glycoprotein (MAG) is a transmembrane glycoprotein concentrated in periaxonal Schwann cell and oligodendroglial membranes of myelin sheaths that serves as an antigen for immunoglobulin M (IgM) monoclonal antibodies. Individuals who harbor anti-MAG antibodies classically develop a progressive autoimmune peripheral neuropathy characterized clinically by ataxia, distal sensory loss, and gait instability, and electrophysiologically by distally accentuated conduction velocity slowing. Although off-label immunotherapy is common, there are currently no proven effective disease-modifying therapeutics, and most patients experience slow accumulation of disability over years and decades. The typically slowly progressive nature of this neuropathy presents unique challenges when trying to find effective anti-MAG therapeutic agents. Drug development has also been hampered by the lack of validated outcome measures that can detect clinically meaningful changes in a reasonable amount of time as well as by the lack of disease activity biomarkers. In this invited review, we provide an update on the state of clinicometric outcome measures and disease activity biomarkers in anti-MAG neuropathy. We highlight the insensitivity of widely used existing clinicometric outcome measures such as the Inflammatory Neuropathy Cause and Treatment (INCAT) disability score as well as the INCAT sensory subscore in anti-MAG neuropathy, referencing the two previous negative randomized controlled clinical trials evaluating rituximab. We then discuss newly emerging candidate therapeutic agents, including tyrosine kinase inhibitors and enhanced B-cell-depleting agents, among others. We conclude with a practical approach to the evaluation and management of anti-MAG neuropathy patients.
Collapse
Affiliation(s)
- Amro Maher Stino
- Department of Neurology, Division of Neuromuscular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bakri Elsheikh
- Department of Neurology, Division of Neuromuscular Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jeffrey A Allen
- Department of Neurology, Division of Neuromuscular Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
5
|
Abstract
The autoimmune peripheral neuropathies with prominent motor manifestations are a diverse collection of unusual peripheral neuropathies that are appreciated in vast clinical settings. This chapter highlights the most common immune-mediated, motor predominant neuropathies excluding acute, and chronic inflammatory demyelinating polyradiculoneuropathy (AIDP and CIDP, respectively). Other acquired demyelinating neuropathies such as distal CIDP and multifocal motor neuropathy will be covered. Additionally, the radiculoplexus neuropathies, resulting from microvasculitis-induced injury to nerve roots, plexuses, and nerves, including diabetic and nondiabetic lumbosacral radiculoplexus neuropathy and neuralgic amyotrophy (i.e., Parsonage-Turner syndrome), will be included. Finally, the motor predominant peripheral neuropathies encountered in association with rheumatological disease, particularly Sjögren's syndrome and rheumatoid arthritis, are covered. Early recognition of these distinct motor predominant autoimmune neuropathies and initiation of immunomodulatory and immunosuppressant treatment likely result in improved outcomes.
Collapse
Affiliation(s)
- Ryan Naum
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States
| | - Kelly Graham Gwathmey
- Neuromuscular Division, Department of Neurology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
6
|
Briani C, Cocito D, Campagnolo M, Doneddu PE, Nobile-Orazio E. Update on therapy of chronic immune-mediated neuropathies. Neurol Sci 2022; 43:605-614. [PMID: 33452933 DOI: 10.1007/s10072-020-04998-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/12/2020] [Indexed: 12/27/2022]
Abstract
Chronic immune-mediated neuropathies, including chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), neuropathies associated with monoclonal gammopathy, and multifocal motor neuropathy (MMN), are a group of disorders deemed to be caused by an immune response against peripheral nerve antigens. Several immune therapies have been reported to be variably effective in these neuropathies including steroids, plasma exchange, and high-dose intravenous (IVIg) or subcutaneous (SCIg) immunoglobulins. These therapies are however far from being invariably effective and may be associated with a number of side effects leading to the use of immunosuppressive agents whose efficacy has not been so far confirmed in randomized trials. More recently, new biological agents, such as rituximab, have proved to be effective in patients with neuropathy associated with IgM monoclonal gammopathy and are currently tested in CIDP.
Collapse
Affiliation(s)
- Chiara Briani
- Neurology Unit, Department of Neuroscience, University of Padova, Via Giustiniani, 5, 35128, Padova, Italy.
| | - Dario Cocito
- Istituti Clinici Scientifici Maugeri, Torino, Italy
| | - Marta Campagnolo
- Neurology Unit, Department of Neuroscience, University of Padova, Via Giustiniani, 5, 35128, Padova, Italy
| | - Pietro Emiliano Doneddu
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Eduardo Nobile-Orazio
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical and Research Institute, Rozzano, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, Milan University, Milan, Italy
| |
Collapse
|
7
|
Parisi M, Dogliotti I, Clerico M, Bertuzzo D, Benevolo G, Orsucci L, Schiavetti I, Cavallo R, Cavallo F, Ragaini S, Di Liberto A, Ferrante M, Bondielli G, Artusi CA, Drandi D, Lopiano L, Ferrero B, Ferrero S. Efficacy of rituximab in anti-myelin-associated glycoprotein demyelinating polyneuropathy: Clinical, hematological and neurophysiological correlations during 2 years of follow-up. Eur J Neurol 2022; 29:3611-3622. [PMID: 36083713 PMCID: PMC9825860 DOI: 10.1111/ene.15553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/31/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND PURPOSE We evaluated the clinical and neurophysiological efficacy of rituximab (RTX) in a neurophysiologically homogeneous group of patients with monoclonal gammopathy and immunoglobulin M (IgM) anti-myelin-associated glycoprotein antibody (anti-MAG) demyelinating polyneuropathy. METHODS Twenty three anti-MAG-positive polyneuropathic patients were prospectively evaluated before and for 2 years after treatment with RTX 375 mg/m2 . The Inflammatory Neuropathy Cause and Treatment (INCAT) disability scale (INCAT-ds), modified INCAT sensory score (mISS), Medical Research Council sum score, Patients' Global Impression of Change scale were used, IgM levels were assessed and extensive electrophysiological examinations were performed before (T0) and 1 year (T1) and 2 years (T2) after RTX treatment. RESULTS At T1 and T2 there was a significant reduction from T0 both in mISS and in INCAT-ds, with a p value < 0.001 in the inferential Friedman's test overall analysis. Ulnar nerve Terminal Latency Index and distal motor latency significantly changed from T0 to T1 and in the overall analysis (p = 0.001 and p = 0.002), and ulnar nerve sensory nerve action potential (SNAP) amplitude was significantly increased at T2 from T1, with a p value < 0.001 in the overall analysis. Analysis of the receiver-operating characteristic curves showed that a 41.8% increase in SNAP amplitude in the ulnar nerve at T2 from T0 was a fair predictor of a mISS reduction of ≥2 points (area under the curve 0.85; p = 0.005; sensitivity: 90.9%, specificity: 83.3%). CONCLUSIONS This study suggests that RTX is effective in patients with clinically active demyelinating anti-MAG neuropathy over 2 years of follow-up, and that some neurophysiological variables might be useful for monitoring this efficacy.
Collapse
Affiliation(s)
- Mattia Parisi
- Department of NeurosciencesUniversity of TurinTurinItaly
| | - Irene Dogliotti
- Stem Cell Transplant UnitUniversity Hospital A.O.U. “Città della Salute e della Scienza di Torino”TurinItaly
| | - Michele Clerico
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly,SSD Myeloma Unit and Clinical Trial, University HaematologyA.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| | | | - Giulia Benevolo
- SSD Myeloma Unit and Clinical Trial, University HaematologyA.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| | - Lorella Orsucci
- Division of Hematology 2A.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| | - Irene Schiavetti
- Section of Biostatistics, Department of Health SciencesUniversity of GenoaGenoaItaly
| | - Roberto Cavallo
- Department of NeurologyOspedale San Giovanni BoscoTurinItaly
| | - Federica Cavallo
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly,SSD Myeloma Unit and Clinical Trial, University HaematologyA.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| | - Simone Ragaini
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly,SSD Myeloma Unit and Clinical Trial, University HaematologyA.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| | | | - Martina Ferrante
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
| | - Giulia Bondielli
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
| | | | - Daniela Drandi
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
| | | | - Bruno Ferrero
- Department of NeurosciencesUniversity of TurinTurinItaly
| | - Simone Ferrero
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly,SSD Myeloma Unit and Clinical Trial, University HaematologyA.O.U. "Città della Salute e della Scienza di Torino"TurinItaly
| |
Collapse
|
8
|
Gonzalez NL, Juel VC, Živković SA. A Case of Probable Multifocal Motor Neuropathy With Clinical Stability for Ten Years After a Single Treatment of Rituximab. J Clin Neuromuscul Dis 2022; 23:136-142. [PMID: 35188910 DOI: 10.1097/cnd.0000000000000358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Multifocal motor neuropathy is a rare, immune-mediated motor neuropathy with asymmetric, often debilitating progressive weakness. The efficacy of intravenous immunoglobulin in this disease is well established; however, the response typically wanes over time. No other agent has shown similar therapeutic efficacy. We describe a case of anti-ganglioside GM1 IgM-positive multifocal motor neuropathy with typical incomplete and diminishing response to intravenous immunoglobulin over time. Sixteen years after symptom onset, rituximab was administered at 2 g/m2 over 2 weeks. No significant progression of disease has occurred over the following 10 years despite no additional treatments, including intravenous immunoglobulin, being given. Only case reports and small, mostly uncontrolled studies have reported the use of rituximab in multifocal motor neuropathy with mixed results. However, given its potential benefits and lack of an established second-line agent, treatment with rituximab may be considered in select patients with refractory multifocal motor neuropathy.
Collapse
Affiliation(s)
| | - Vern C Juel
- Department of Neurology, Duke University Hospital, Durham, NC; and
| | - Saša A Živković
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
9
|
Hänggi P, Aliu B, Martin K, Herrendorff R, Steck AJ. Decrease in Serum Anti-MAG Autoantibodies Is Associated With Therapy Response in Patients With Anti-MAG Neuropathy: Retrospective Study. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/1/e1109. [PMID: 34759022 PMCID: PMC8587733 DOI: 10.1212/nxi.0000000000001109] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 08/30/2021] [Indexed: 11/15/2022]
Abstract
Background and Objectives The objective of the retrospective analysis was to test the hypothesis that changes in serum anti-myelin-associated glycoprotein (MAG) autoantibodies are associated with clinical response to immunotherapy in patients with anti-MAG neuropathy. Methods As of January 29, 2020, we used anti-myelin-associated glycoprotein-related search strings in the Medline database to identify studies that provided information on anti-MAG immunoglobulin M (IgM) autoantibodies and clinical outcomes during immunotherapies. The relative change in anti-MAG IgM titers, paraprotein levels, or total IgM was determined before, during, or posttreatment, and the patients were assigned to “responder,” “nonresponder,”’ or “acute deteriorating” category depending on their clinical response to treatment. The studies were qualified as “supportive” or “not supportive” depending on the percentage of patients exhibiting an association between relative change of anti-MAG antibody titers or levels and change in clinical outcomes. Results Fifty studies with 410 patients with anti-MAG neuropathy were included in the analysis. Forty studies with 303 patients supported the hypothesis that a “responder” patient had a relative reduction of anti-MAG antibody titers or levels that is associated with clinical improvements and “nonresponder” patients exhibited no significant change in anti-MAG IgM antibodies. Six studies with 93 patients partly supported, and 4 studies with 26 patients did not support the hypothesis. Discussion The retrospective analysis confirmed the hypothesis that a relative reduction in serum anti-MAG IgM antibodies is associated with a clinical response to immunotherapies; a sustained reduction of at least 50% compared with pretreatment titers or levels could be a valuable indicator for therapeutic response.
Collapse
Affiliation(s)
- Pascal Hänggi
- From the Polyneuron Pharmaceuticals AG (P.H.,K.M.,R.H.), Basel; Molecular Pharmacy (P.H.,B.A.,R.H.), Pharmacenter, University of Basel; and Clinic of Neurology (A.J.S.), Department of Medicine, University Hospital Basel, University of Basel, Switzerland.
| | - Butrint Aliu
- From the Polyneuron Pharmaceuticals AG (P.H.,K.M.,R.H.), Basel; Molecular Pharmacy (P.H.,B.A.,R.H.), Pharmacenter, University of Basel; and Clinic of Neurology (A.J.S.), Department of Medicine, University Hospital Basel, University of Basel, Switzerland
| | - Kea Martin
- From the Polyneuron Pharmaceuticals AG (P.H.,K.M.,R.H.), Basel; Molecular Pharmacy (P.H.,B.A.,R.H.), Pharmacenter, University of Basel; and Clinic of Neurology (A.J.S.), Department of Medicine, University Hospital Basel, University of Basel, Switzerland
| | - Ruben Herrendorff
- From the Polyneuron Pharmaceuticals AG (P.H.,K.M.,R.H.), Basel; Molecular Pharmacy (P.H.,B.A.,R.H.), Pharmacenter, University of Basel; and Clinic of Neurology (A.J.S.), Department of Medicine, University Hospital Basel, University of Basel, Switzerland
| | - Andreas Johann Steck
- From the Polyneuron Pharmaceuticals AG (P.H.,K.M.,R.H.), Basel; Molecular Pharmacy (P.H.,B.A.,R.H.), Pharmacenter, University of Basel; and Clinic of Neurology (A.J.S.), Department of Medicine, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
10
|
Principles and Guidelines of Immunotherapy in Neuromuscular Disorders. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Pessach I, Dimopoulos MA, Kastritis E. Managing complications secondary to Waldenström's macroglobulinemia. Expert Rev Hematol 2021; 14:621-632. [PMID: 34170207 DOI: 10.1080/17474086.2021.1947236] [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: 10/21/2022]
Abstract
Introduction: Waldenström's macroglobulinemia (WM) is a rare lymphoma characterized by the accumulation of IgM-secreting lymphoplasmacytic cells in the bone marrow and other organs. Clinical sequelae relate to direct tissue infiltration by malignant cells but also to the physicochemical and immunological properties of the monoclonal IgM, resulting in a variety of disease-related complications.Areas covered: This narrative review, following a thorough Pubmed search of pertinent published literature, discusses complications secondary to WM, related to direct tumor infiltration, monoclonal IgM circulation, and deposition, as well as other less common ones. The description and pathophysiology of these complications were described together with their specific management strategies and in the context of available treatment options for WM (anti-CD20 monoclonal antibody-based combinations, proteasome inhibitors, BTK inhibitors, and other emerging ones).Expert opinion: The availability of many novel, active and less toxic regimens for the treatment of WM allows the management of the disease with strategies that depend on clinical presentation and disease-related complications, age, toxicity considerations, and presence of comorbidities.
Collapse
Affiliation(s)
- Ilias Pessach
- Division of Hematology, Athens Medical Center, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Castillo JJ, Callander NS, Baljevic M, Sborov DW, Kumar S. The evaluation and management of monoclonal gammopathy of renal significance and monoclonal gammopathy of neurological significance. Am J Hematol 2021; 96:846-853. [PMID: 33709474 PMCID: PMC8252623 DOI: 10.1002/ajh.26155] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/07/2021] [Indexed: 12/24/2022]
Abstract
Despite the benign nature of monoclonal gammopathy of undetermined significance (MGUS), mounting data are associating MGUS with the development of organ dysfunction, specifically monoclonal gammopathy of renal significance (MGRS) and monoclonal gammopathy of neurological significance (MGNS), which could be associated with substantial morbidity. Emerging evidence suggests that patients with MGRS and MGNS could benefit from treatments used for myeloma, Waldenström macroglobulinemia, or chronic lymphocytic leukemia, depending on the underlying pathology. However, the treatment of MGRS and MGNS is not standardized, and potentially effective therapies might not be reimbursed because these conditions do not formally meet the criteria for malignant processes. The present review aims at establishing standards for the evaluation and management of MGRS and MGNS, which can facilitate the diagnosis of and provide therapeutic options for treating practitioners and patients affected by these conditions. The careful design and execution of clinical trials for patients with MGRS and MGNS are positively encouraged.
Collapse
Affiliation(s)
- Jorge J. Castillo
- Dana‐Farber Cancer Institute Harvard Medical School Boston Massachusetts
| | | | | | | | | |
Collapse
|
13
|
Aliu B, Demeestere D, Seydoux E, Boucraut J, Delmont E, Brodovitch A, Oberholzer T, Attarian S, Théaudin M, Tsouni P, Kuntzer T, Derfuss T, Steck AJ, Ernst B, Herrendorff R, Hänggi P. Selective inhibition of anti-MAG IgM autoantibody binding to myelin by an antigen-specific glycopolymer. J Neurochem 2020; 154:486-501. [PMID: 32270492 PMCID: PMC7497077 DOI: 10.1111/jnc.15021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022]
Abstract
Anti‐myelin‐associated glycoprotein (MAG) neuropathy is a disabling autoimmune peripheral neuropathy that is caused by circulating monoclonal IgM autoantibodies directed against the human natural killer‐1 (HNK‐1) epitope. This carbohydrate epitope is highly expressed on adhesion molecules such as MAG, a glycoprotein present in myelinated nerves. We previously showed the therapeutic potential of the glycopolymer poly(phenyl disodium 3‐O‐sulfo‐β‐d‐glucopyranuronate)‐(1→3)‐β‐d‐galactopyranoside (PPSGG) in selectively neutralizing anti‐MAG IgM antibodies in an immunological mouse model and ex vivo with sera from anti‐MAG neuropathy patients. PPSGG is composed of a biodegradable backbone that multivalently presents a mimetic of the HNK‐1 epitope. In this study, we further explored the pharmacodynamic properties of the glycopolymer and its ability to inhibit the binding of anti‐MAG IgM to peripheral nerves. The polymer selectively bound anti‐MAG IgM autoantibodies and prevented the binding of patients’ anti‐MAG IgM antibodies to myelin of non‐human primate sciatic nerves. Upon PPSGG treatment, neither activation nor inhibition of human and murine peripheral blood mononuclear cells nor alteration of systemic inflammatory markers was observed in mice or ex vivo in human peripheral blood mononuclear cells. Intravenous injections of PPSGG to mice immunized against the HNK‐1 epitope removed anti‐MAG IgM antibodies within less than 1 hr, indicating a fast and efficient mechanism of action as compared to a B‐cell depletion with anti‐CD20. In conclusion, these observations corroborate the therapeutic potential of PPSGG for an antigen‐specific treatment of anti‐MAG neuropathy. ![]()
Read the Editorial Highlight for this article on page 465.
Collapse
Affiliation(s)
- Butrint Aliu
- Institute of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Delphine Demeestere
- Institute of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | | - José Boucraut
- Immunology laboratory, AP-HM, Marseille, France.,INT, UMR CNRS 7289, Aix-Marseille University, Marseille, France
| | - Emilien Delmont
- Center for Neuromuscular Disorders and ALS La Timone Hospital, AP-HM, Marseille, France
| | - Alexandre Brodovitch
- Immunology laboratory, AP-HM, Marseille, France.,Center for Neuromuscular Disorders and ALS La Timone Hospital, AP-HM, Marseille, France
| | | | - Shahram Attarian
- Center for Neuromuscular Disorders and ALS La Timone Hospital, AP-HM, Marseille, France
| | - Marie Théaudin
- Nerve-Muscle Unit, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Pinelopi Tsouni
- Nerve-Muscle Unit, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Thierry Kuntzer
- Nerve-Muscle Unit, Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Tobias Derfuss
- Clinic of Neurology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas J Steck
- Clinic of Neurology, Department of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Beat Ernst
- Institute of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Ruben Herrendorff
- Institute of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.,Polyneuron Pharmaceuticals AG, Basel, Switzerland
| | - Pascal Hänggi
- Institute of Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.,Polyneuron Pharmaceuticals AG, Basel, Switzerland
| |
Collapse
|
14
|
Gao XM, Jia MN, Qian M, Ren HT, Zhang L, Shen KN, Cao XX, Li J. [Anti-myelin-associated glycoprotein antibody positive IgM monoclonal gammopathy related peripheral neuropathy: 11 cases and literature review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:901-905. [PMID: 31856437 PMCID: PMC7342372 DOI: 10.3760/cma.j.issn.0253-2727.2019.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Indexed: 11/08/2022]
Abstract
Objective: To improve the understanding of rare anti-myelin-associated glycoprotein (MAG) positive IgM monoclonal gammopathy related peripheral neuropathy (IgM-PN) . Methods: Eleven cases of IgM paraproteinemia and anti-MAG antibody positive neuropathy diagnosed since 2014 in Peking Medical Union College Hospital were summarized. The medical records including clinical manifestation, lab results, treatment and prognosis were analyzed. Results: Among the 11 patients (8 male and 3 female) , the median onset age is 63 years old (range from 52 to 77 years old) . The peripheral neuropathy of 9 patients were characterized by distal onset of numbness, 6 patients suffered from muscle weakness. The nerve conduction velocity study indicated that all 11 patients had demyelinating peripheral nerve damage, which was sensory predominant and more severe in lower limbs, 6 of them had secondary axonal damage. Monoclonal IgM gammopathy was identified in all 11 patients, among which 6 were IgM κ, 2 IgG κ and IgM κ bi-clonal, 3 IgM λ. Three patients were diagnosed with Waldenström's macroglobulinaemia. The anti-MAG-IgM antibody was positive in all 11 cases. After diagnosis, 9 patients received combination chemotherapy including rituximab or rituximab treatment alone. The monoclonal IgM level declined significantly in 7 patients. The neuropathy was stable or improved. Conclusions: Anti-MAG antibody positive IgM-PN is a rare M protein related disease. In peripheral neuropathy with undetermined etiology, we suggest to screen M protein and anti-MAG antibody. Chemotherapy including rituximab or rituximab alone is recommended as first-line therapy.
Collapse
Affiliation(s)
- X M Gao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - M N Jia
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - M Qian
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - H T Ren
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - L Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - K N Shen
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - X X Cao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| | - J Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences, Beijing 100730, China
| |
Collapse
|
15
|
Abstract
Since the discovery of an acute monophasic paralysis, later coined Guillain-Barré syndrome, almost 100 years ago, and the discovery of chronic, steroid-responsive polyneuropathy 50 years ago, the spectrum of immune-mediated polyneuropathies has broadened, with various subtypes continuing to be identified, including chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and multifocal motor neuropathy (MMN). In general, these disorders are speculated to be caused by autoimmunity to proteins located at the node of Ranvier or components of myelin of peripheral nerves, although disease-associated autoantibodies have not been identified for all disorders. Owing to the numerous subtypes of the immune-mediated neuropathies, making the right diagnosis in daily clinical practice is complicated. Moreover, treating these disorders, particularly their chronic variants, such as CIDP and MMN, poses a challenge. In general, management of these disorders includes immunotherapies, such as corticosteroids, intravenous immunoglobulin or plasma exchange. Improvements in clinical criteria and the emergence of more disease-specific immunotherapies should broaden the therapeutic options for these disabling diseases.
Collapse
|
16
|
Svahn J, Petiot P, Antoine JC, Vial C, Delmont E, Viala K, Steck AJ, Magot A, Cauquil C, Zarea A, Echaniz-Laguna A, Iancu Ferfoglia R, Gueguen A, Magy L, Léger JM, Kuntzer T, Ferraud K, Lacour A, Camdessanché JP. Anti-MAG antibodies in 202 patients: clinicopathological and therapeutic features. J Neurol Neurosurg Psychiatry 2018; 89:499-505. [PMID: 29070644 DOI: 10.1136/jnnp-2017-316715] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To assess the clinicopathological and therapeutic features of patients with low (≥1000 to <10 000 Bühlmann Titre Units) (BTU), medium (10 000-70 000) or high (≥70 000) anti-myelin-associated glycoprotein (anti-MAG) antibody titres. METHODS We retrospectively and prospectively analysed standardised report forms and medical records of 202 patients from 14 neuromuscular centres. RESULTS Mean age at onset and mean time between symptom onset to last follow-up were respectively 62.6 years (25-91.4) and 8.4 years (0.3-33.3). Anti-MAG antibody titres at diagnosis were low, medium or high in 11%, 51% and 38% of patients. Patients presented with monoclonal gammopathy of undetermined significance in 68% of cases. About 17% of patients presented with 'atypical' clinical phenotype independently of anti-MAG titres, including acute or chronic sensorimotor polyradiculoneuropathies (12.4%), and asymmetric or multifocal neuropathy (3%). At the most severe disease stage, 22.4% of patients were significantly disabled. Seventy-eight per cent of patients received immunotherapies. Transient clinical worsening was observed in 12% of patients treated with rituximab (11/92). Stabilisation after rituximab treatment during the 7-12-month follow-up period was observed in 29% of patients. Clinical response to rituximab during the 6-month and/or 7-12-month follow-up period was observed in 31.5% of patients and correlated with anti-MAG titre ≥10 000 BTU. CONCLUSION Our study highlights the extended clinical spectrum of patients with anti-MAG neuropathy, which appears unrelated to antibody titre. Besides, it may also suggest beneficial use of rituximab in the early phase of anti-MAG neuropathy.
Collapse
Affiliation(s)
- Juliette Svahn
- Electroneuromyography and Neuromuscular Department, University Hospital Lyon, Lyon, France
| | - Philippe Petiot
- Electroneuromyography and Neuromuscular Department, University Hospital Lyon, Lyon, France
| | | | - Christophe Vial
- Electroneuromyography and Neuromuscular Department, University Hospital Lyon, Lyon, France
| | - Emilien Delmont
- Department of Neurology, University Hospital Timone, Marseille, France
| | - Karine Viala
- Department of Neurophysiology and Neuropathology, University Hospital Pitié-Salpêtrière, Paris, France
| | - Andreas J Steck
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Armelle Magot
- Neuromuscular Reference Center, University Hospital Nantes, Nantes, France
| | - Cecile Cauquil
- Department of Neurology, University Hospital Bicêtre, Le Kremlin Bicêtre, France
| | - Aline Zarea
- Neuromuscular Competence Center, University Hospital Rouen, Rouen, France
| | | | - Ruxandra Iancu Ferfoglia
- Electroneuromyography and Neuromuscular Disorders Unit, University Hospital Geneva, Geneva, Switzerland
| | - Antoine Gueguen
- Department of Neurology, Fondation Ophtalmologique A. de Rothschild, Paris, France
| | - Laurent Magy
- Department of Neurology, University Hospital Limoges, Limoges, France
| | - Jean-Marc Léger
- Department of Neurology, University Hospital Pitié-Salpêtrière, Paris, France
| | - Thierry Kuntzer
- Department of Clinical Neurosciences, Nerve-Muscle Unit, Neurology Service, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Karine Ferraud
- Department of Neurology, University Hospital Saint-Etienne, Saint-Etienne, France
| | - Arnaud Lacour
- Department of Neurology, University Hospital Saint-Etienne, Saint-Etienne, France
| | | | | |
Collapse
|
17
|
Dalakas MC. Advances in the diagnosis, immunopathogenesis and therapies of IgM-anti-MAG antibody-mediated neuropathies. Ther Adv Neurol Disord 2018; 11:1756285617746640. [PMID: 29403542 PMCID: PMC5791554 DOI: 10.1177/1756285617746640] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/16/2017] [Indexed: 11/17/2022] Open
Abstract
Polyneuropathy with immunoglobulin M (IgM) monoclonal gammopathy is the most common paraproteinemic neuropathy, comprising a clinicopathologically and immunologically distinct entity. The clinical spectrum spans from distal paresthesias and mild gait imbalance to more severe sensory ataxia, with falls and a varying degree of distal sensorimotor deficits. In approximately 75% of patients, the monoclonal IgM immunoreacts with myelin-associated glycoprotein (MAG) and sulfoglucuronyl glycosphingolipid (SGPG), or other peripheral nerve glycolipids that serve as antigens. These antibodies are considered pathogenic because IgM and complement are deposited on the myelin sheath, splitting the myelin lamellae, while adoptive transfer of patients’ IgM into susceptible host animals causes sensory ataxia and reproduces the human pathology. In spite of the apparently convincing pathogenicity of these antibodies, the response to immunotherapies remains suboptimal. Clorambuscil, cladibrine, cyclophospamide and intravenous immunoglobulin may help some patients but the benefits are minimal and transient. Open-label studies in >200 patients indicate that rituximab is helpful in 30–50% of these patients, even with long-term benefits, probably by suppressing IgM anti-MAG antibodies or inducing immunoregulatory T cells. Two controlled studies with rituximab did not however meet the primary endpoint, mostly because of the poor sensitivity of the scales used; they did however show statistical improvement in secondary endpoints and improved clinical functions in several patients. This review provides an overview of the clinical phenotypes and immunoreactivity of IgM to glycolipids or glycoproteins of peripheral nerve myelin, summarizes the progress on treatment with rituximab as a promising therapy, discusses the pitfalls of scales used, identifies possible biomarkers of response to therapy and highlights the promising new anti-B cell or target-specific immunotherapies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Neuroimmunology Unit, Department of Pathophysiology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
18
|
Pruppers MH, Merkies IS, Lunn MP, Notermans NC, van den Bergh P, Blomkwist-Markens P, Cornblath D, D'Sa S, Faber C, Goedee S, Gorson K, Léger JM, Lewis R, Lunn M, Mazawey L, Merkies I, Nobile-Orazio E, Notermans N, Padua L, van der Pol L, Pruppers M, Querol L, Steck A, Willison H. 230th ENMC International Workshop:. Neuromuscul Disord 2017; 27:1065-1072. [DOI: 10.1016/j.nmd.2017.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Indexed: 12/30/2022]
|
19
|
Nobile-Orazio E, Gallia F, Terenghi F, Bianco M. Comparing treatment options for chronic inflammatory neuropathies and choosing the right treatment plan. Expert Rev Neurother 2017; 17:755-765. [DOI: 10.1080/14737175.2017.1340832] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Eduardo Nobile-Orazio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical Institute, Milan, Italy
| | - Francesca Gallia
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical Institute, Milan, Italy
| | - Fabrizia Terenghi
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical Institute, Milan, Italy
| | - Mariangela Bianco
- Neuromuscular and Neuroimmunology Service, IRCCS Humanitas Clinical Institute, Milan, Italy
| |
Collapse
|
20
|
Gazzola S, Delmont E, Franques J, Boucraut J, Salort-Campana E, Verschueren A, Sagui E, Hubert AM, Pouget J, Attarian S. Predictive factors of efficacy of rituximab in patients with anti-MAG neuropathy. J Neurol Sci 2017; 377:144-148. [DOI: 10.1016/j.jns.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/21/2017] [Accepted: 04/10/2017] [Indexed: 01/01/2023]
|
21
|
Dalakas MC. Rituximab in anti-MAG neuropathy: More evidence for efficacy and more predictive factors. J Neurol Sci 2017; 377:224-226. [DOI: 10.1016/j.jns.2017.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
|
22
|
Selective in vivo removal of pathogenic anti-MAG autoantibodies, an antigen-specific treatment option for anti-MAG neuropathy. Proc Natl Acad Sci U S A 2017; 114:E3689-E3698. [PMID: 28416698 DOI: 10.1073/pnas.1619386114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Anti-MAG (myelin-associated glycoprotein) neuropathy is a disabling autoimmune peripheral neuropathy caused by monoclonal IgM autoantibodies that recognize the carbohydrate epitope HNK-1 (human natural killer-1). This glycoepitope is highly expressed on adhesion molecules, such as MAG, present in myelinated nerve fibers. Because the pathogenicity and demyelinating properties of anti-MAG autoantibodies are well established, current treatments are aimed at reducing autoantibody levels. However, current therapies are primarily immunosuppressive and lack selectivity and efficacy. We therefore hypothesized that a significant improvement in the disease condition could be achieved by selectively neutralizing the pathogenic anti-MAG antibodies with carbohydrate-based ligands mimicking the natural HNK-1 glycoepitope 1. In an inhibition assay, a mimetic (2, mimHNK-1) of the natural HNK-1 epitope blocked the interaction of MAG with pathogenic IgM antibodies from patient sera but with only micromolar affinity. Therefore, considering the multivalent nature of the MAG-IgM interaction, polylysine polymers of different sizes were substituted with mimetic 2. With the most promising polylysine glycopolymer PL84(mimHNK-1)45 the inhibitory effect on patient sera could be improved by a factor of up to 230,000 per epitope, consequently leading to a low-nanomolar inhibitory potency. Because clinical studies indicate a correlation between the reduction of anti-MAG IgM levels and clinical improvement, an immunological surrogate mouse model for anti-MAG neuropathy producing high levels of anti-MAG IgM was developed. The observed efficient removal of these antibodies with the glycopolymer PL84(mimHNK-1)45 represents an important step toward an antigen-specific therapy for anti-MAG neuropathy.
Collapse
|
23
|
D'Sa S, Kersten MJ, Castillo JJ, Dimopoulos M, Kastritis E, Laane E, Leblond V, Merlini G, Treon SP, Vos JM, Lunn MP. Investigation and management of IgM and Waldenström-associated peripheral neuropathies: recommendations from the IWWM-8 consensus panel. Br J Haematol 2017; 176:728-742. [PMID: 28198999 DOI: 10.1111/bjh.14492] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Paraproteinaemic neuropathies are a heterogeneous group of disorders most frequently associated with IgM monoclonal gammopathies including Waldenström macroglobulinaemia (WM). Their consequences are significant for affected patients, and their management challenging for their physicians. The variability in clinical presentation and time course hamper classification and management. The indications for invasive investigations such as cerebrospinal fluid analysis, nerve conduction tests and sensory nerve biopsies are unclear, and the optimum way to measure clinical response to treatment unknown. When to intervene and and how to treat, also present challenges to physicians. As part of its latest deliberations at the International Workshops on WM (IWWM) in London, UK (August 2014), the IWWM8 panel have proposed a consensus approach to the diagnosis and management of peripheral neuropathies associated with IgM monoclonal gammopathies, including WM. Importantly, a consensus regarding the use of clinical outcome measures and recommended models of care for this group of patients is discussed, as well as appropriate treatment interventions.
Collapse
Affiliation(s)
- Shirley D'Sa
- Waldenström Clinic, Cancer Division, University College London Hospitals NHS Foundation Trust, London, UK
| | - Marie José Kersten
- Department of Haematology, Academic Medical Centre, Amsterdam, the Netherlands
| | - Jorge J Castillo
- Bing Center for Waldenström Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Meletios Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Edward Laane
- Department of Haematology, North Estonia Medical Centre, Tallinn, Estonia
| | - Véronique Leblond
- AP-HP Hôpital Pitié Salpêtrière, UPMC Univ. Paris 6 GRC-11, Grechy, Paris, France
| | - Giampaolo Merlini
- Centre for Research and Treatment of Systemic Amyloidosis, University of Pavia, Pavia, Italy
| | - Steven P Treon
- Bing Center for Waldenström Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Josephine M Vos
- Department of Haematology, Academic Medical Centre, Amsterdam, the Netherlands.,Cancer Centre, Sint Antonius Ziekenhuis, Nieuwegein, the Netherlands
| | - Michael P Lunn
- Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
24
|
Grimm A, Schubert V, Axer H, Ziemann U. Giant nerves in chronic inflammatory polyradiculoneuropathy. Muscle Nerve 2016; 55:285-289. [PMID: 27463360 DOI: 10.1002/mus.25272] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Nerve enlargement (NE) is described in inflammatory and inherited neuropathies. It is commonly multifocal and moderate in the former and homogeneous and generalized in the latter. We describe 4 cases of massive NE in inflammatory neuropathies. METHODS Patients presented with symptoms of polyneuropathy that progressed over months to years. Nerve conduction studies (NCS), laboratory analysis, nerve MRI, and nerve ultrasound were performed. RESULTS NCS revealed demyelinating neuropathy in all with multifocal conduction blocks or increased terminal latency indices. MRI/ultrasound revealed extensive NE in the roots and nerves. Detailed diagnostics including biopsies, positron emission tomography-computed tomography, and genetic testing revealed no other pathology. Chronic inflammatory demyelinating polyneuropathy variants were diagnosed in all, and immunotherapies were successfully initiated. CONCLUSIONS MRI and ultrasound contributed to diagnosis and therapy. All patients had giant NE in common, which strongly suggested inherited neuropathy. However, the final diagnosis was inflammatory neuropathy. Impressive NE can occur in immune-mediated neuropathies and should be carefully differentiated from inherited neuropathies. Muscle Nerve 55: 285-289, 2017.
Collapse
Affiliation(s)
- Alexander Grimm
- Center of Neurology, Tübingen University Hospital, and Hertie Institute for Clinical Brain Research, Eberhard-Karls University Tübingen, Hoppe-Seyler Str. 3, D-72076, Tübingen, Germany
| | - Victoria Schubert
- Center of Neurology, Tübingen University Hospital, and Hertie Institute for Clinical Brain Research, Eberhard-Karls University Tübingen, Hoppe-Seyler Str. 3, D-72076, Tübingen, Germany
| | - Hubertus Axer
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Ulf Ziemann
- Center of Neurology, Tübingen University Hospital, and Hertie Institute for Clinical Brain Research, Eberhard-Karls University Tübingen, Hoppe-Seyler Str. 3, D-72076, Tübingen, Germany
| |
Collapse
|
25
|
Catroux M, Lauda-Maillen M, Pathe M, De Boisgrollier de Ruolz AC, Cazenave-Roblot F, Roblot P, Souchaud-Debouverie O. [Infectious events during the course of autoimmune diseases treated with rituximab: A retrospective study of 93 cases]. Rev Med Interne 2016; 38:160-166. [PMID: 27836224 DOI: 10.1016/j.revmed.2016.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Describe the occurring infections in patients treated with rituximab for an autoimmune disease. METHODS Retrospective and monocentric study of 93 adult patients treated with rituximab for autoimmune indications over a nine years period. RESULTS Thirty-eight patients suffered from a total of 95 infections. Out of them, 18 patients (19 %) had had at least an infectious episode triggering a hospital admission and/or intravenous treatment. The infections occurred mainly during the first year of the treatment (65 %) and if the courses are repeated (P=0.04). They were mainly pulmonary infections. Severe infections, recorded in 79 % of the cases, were mostly of bacterial origin (43 %) and viral (23 %). Two cases of pneumocystis pneumonia and one case of invasive pulmonary aspergillosis were also recorded. The notion of vaccination was present in less than half of the cases, and 39 % of the patients were already receiving a prophylactic treatment against pneumocystis pneumonia. Patients over the age of 65 years (40 %) had developed less infections (P<0.05). Eight of the initial 93 patients died, half of them because of infectious complications. CONCLUSION Infectious complications are frequent, become early and are potentially severe. Imputability to rituximab is not certain. However, this could lead to better codify rituximab prescriptions and take adapted and associated measures in order to facilitate infection prevention and, if an infection does occur, to treat it at the earliest stage possible. The age doesn't seem to be a risk factor.
Collapse
Affiliation(s)
- M Catroux
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France.
| | - M Lauda-Maillen
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France
| | - M Pathe
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France
| | | | - F Cazenave-Roblot
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France
| | - P Roblot
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France
| | - O Souchaud-Debouverie
- Service de médecine interne et maladies infectieuses, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, BP 577, 86021 Poitiers, France
| |
Collapse
|
26
|
Lunn MPT, Nobile‐Orazio E. Immunotherapy for IgM anti-myelin-associated glycoprotein paraprotein-associated peripheral neuropathies. Cochrane Database Syst Rev 2016; 10:CD002827. [PMID: 27701752 PMCID: PMC6457998 DOI: 10.1002/14651858.cd002827.pub4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Serum monoclonal anti-myelin-associated glycoprotein (anti-MAG) antibodies may be pathogenic in some people with immunoglobulin M (IgM) paraprotein and demyelinating neuropathy. Immunotherapies aimed at reducing the level of these antibodies might be expected to be beneficial. This is an update of a review first published in 2003 and previously updated in 2006 and 2012. OBJECTIVES To assess the effects of immunotherapy for IgM anti-MAG paraprotein-associated demyelinating peripheral neuropathy. SEARCH METHODS On 1 February 2016 we searched the Cochrane Neuromuscular Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, and Embase for randomised controlled trials (RCTs). We also checked trials registers and bibliographies, and contacted authors and experts in the field. SELECTION CRITERIA We included randomised controlled trials (RCTs) or quasi-RCTs involving participants of any age treated with any type of immunotherapy for anti-MAG antibody-associated demyelinating peripheral neuropathy with monoclonal gammopathy of undetermined significance and of any severity.Our primary outcome measures were numbers of participants improved in disability assessed with either or both of the Neuropathy Impairment Scale (NIS) or the modified Rankin Scale (mRS) at six months after randomisation. Secondary outcome measures were: mean improvement in disability, assessed with either the NIS or the mRS, 12 months after randomisation; change in impairment as measured by improvement in the 10-metre walk time, change in a validated linear disability measure such as the Rasch-built Overall Disability Scale (R-ODS) at six and 12 months after randomisation, change in subjective clinical scores and electrophysiological parameters at six and 12 months after randomisation; change in serum IgM paraprotein concentration or anti-MAG antibody titre at six months after randomisation; and adverse effects of treatments. DATA COLLECTION AND ANALYSIS We followed standard methodological procedures expected by Cochrane. MAIN RESULTS We identified eight eligible trials (236 participants), which tested intravenous immunoglobulin (IVIg), interferon alfa-2a, plasma exchange, cyclophosphamide and steroids, and rituximab. Two trials of IVIg (22 and 11 participants, including 20 with antibodies against MAG), had comparable interventions and outcomes, but both were short-term trials. We also included two trials of rituximab with comparable interventions and outcomes.There were very few clinical or statistically significant benefits of the treatments used on the outcomes predefined for this review, but not all the predefined outcomes were used in every included trial and more responsive outcomes are being developed. A well-performed trial of IVIg, which was at low risk of bias, showed a statistical benefit in terms of improvement in mRS at two weeks and 10-metre walk time at four weeks, but these short-term outcomes are of questionable clinical significance. Cyclophosphamide failed to show any benefit in the single trial's primary outcome, and showed a barely significant benefit in the primary outcome specified here, but some toxic adverse events were identified.Two trials of rituximab (80 participants) have been published, one of which (26 participants) was at high risk of bias. In the meta-analysis, although the data are of low quality, rituximab is beneficial in improving disability scales (Inflammatory Neuropathy Cause and Treatment (INCAT) improved at eight to 12 months (risk ratio (RR) 3.51, 95% confidence interval (CI) 1.30 to 9.45; 73 participants)) and significantly more participants improve in the global impression of change score (RR 1.86, 95% CI 1.27 to 2.71; 70 participants). Other measures did not improve significantly, but wide CIs do not preclude some effect. Reported adverse effects of rituximab were few, and mostly minor.There were few serious adverse events in the other trials. AUTHORS' CONCLUSIONS There is inadequate reliable evidence from trials of immunotherapies in anti-MAG paraproteinaemic neuropathy to form an evidence base supporting any particular immunotherapy treatment. IVIg has a statistically but probably not clinically significant benefit in the short term. The meta-analysis of two trials of rituximab provides, however, low-quality evidence of a benefit from this agent. The conclusions of this meta-analysis await confirmation, as one of the two included studies is of very low quality. We require large well-designed randomised trials of at least 12 months' duration to assess existing or novel therapies, preferably employing unified, consistent, well-designed, responsive, and valid outcome measures.
Collapse
Affiliation(s)
- Michael PT Lunn
- National Hospital for Neurology and NeurosurgeryDepartment of Neurology and MRC Centre for Neuromuscular DiseasesQueen SquareLondonUKWC1N 3BG
| | - Eduardo Nobile‐Orazio
- Milan UniversityIRCCS Humanitas Clinical Institute, Neurology 2Istituto Clinico HumanitasVia Manzoni 56, RozzanoMilanItaly20089
| | | |
Collapse
|
27
|
Vallat JM, Magy L, Ciron J, Corcia P, Le Masson G, Mathis S. Therapeutic options and management of polyneuropathy associated with anti-MAG antibodies. Expert Rev Neurother 2016; 16:1111-9. [DOI: 10.1080/14737175.2016.1198257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Iancu Ferfoglia R, Guimarães-Costa R, Viala K, Musset L, Neil J, Marin B, Léger JM. Long-term efficacy of rituximab in IgM anti-myelin-associated glycoprotein neuropathy: RIMAG follow-up study. J Peripher Nerv Syst 2016; 21:10-4. [DOI: 10.1111/jns.12156] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 01/05/2016] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Ruxandra Iancu Ferfoglia
- Department of Neurology, National Referral Center for Rare Neuromuscular Diseases; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
- Department of Neurology; Geneva University Hospitals; Geneva Switzerland
| | - Raquel Guimarães-Costa
- Department of Neurology, National Referral Center for Rare Neuromuscular Diseases; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
| | - Karine Viala
- Department of Neurophysiology; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
| | - Lucile Musset
- Department of Immunology, Laboratory of Immunochemistry & Autoimmunity; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
| | - Jean Neil
- Department of Immunology, Laboratory of Immunochemistry & Autoimmunity; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
| | - Benoit Marin
- Centre d'Epidémiologie de Biostatistique et de Méthodologie de la Recherche; CHU Limoges; Limoges France
- Tropical Neuroepidemiology; INSERM, U1094; Limoges France
- Institute of Neuroepidemiology and Tropical Neurology, CNRS FR 3503 GEIST; University of Limoges, UMR_S 1094, Tropical Neuroepidemiology; Limoges France
| | - Jean-Marc Léger
- Department of Neurology, National Referral Center for Rare Neuromuscular Diseases; University Hospital Pitié-Salpêtrière and University Paris VI; Paris France
| |
Collapse
|
29
|
Uncompacted Myelin Lamellae and Nodal Ion Channel Disruption in POEMS Syndrome. J Neuropathol Exp Neurol 2016; 74:1127-36. [PMID: 26574667 DOI: 10.1097/nen.0000000000000257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
To elucidate the significance of uncompacted myelin lamellae (UML) and ion channel disruption at the nodes of Ranvier in the polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS) syndrome, we evaluated sural nerve biopsy specimens from 33 patients with POEMS syndrome and from 7 control patients. Uncompacted myelin lamellae distribution was assessed by electron microscopy and immunofluorescence microscopy. In the POEMS patient biopsies, UML were seen more frequently in small versus large myelinated fibers. Paranodes and Schmidt-Lanterman incisures, where normal physiologic UM is located, were frequently associated with UM. Widening of the nodes of Ranvier (i.e. segmental demyelination) was not associated with UML. There was axonal hollowing with neurofilament condensation at Schmidt-Lanterman incisures with abnormal UML, suggesting axonal damage at those sites in the POEMS patient biopsies. Myelin sheath irregularity was conspicuous in large myelinated fibers and was associated with abnormally widened bizarrely shaped Schmidt-Lanterman incisures. Indirect immunofluorescent studies revealed abnormalities of sodium (pan sodium) and potassium (KCNQ2) channels, even at nonwidened nodes of Ranvier. Thus, UML was not apparently associated with segmental demyelination but seemed to be associated with axonal damage. These observations suggest that nodal ion channel disruption may be associated with functional deficits in POEMS syndrome patient nerves.
Collapse
|
30
|
Abstract
Immunotherapy has been investigated in a small subset of peripheral neuropathies, including an acute one, Guillain-Barré syndrome, and 3 chronic forms: chronic inflammatory demyelinating polyradiculoneuropathy, multifocal motor neuropathy, and neuropathy associated with IgM anti-myelin-associated glycoprotein. Several experimental studies and clinical data are strongly suggestive of an immune-mediated pathogenesis. Either cell-mediated mechanisms or antibody responses to Schwann cell, compact myelin, or nodal antigens are considered to act together in an aberrant immune response to cause damage to peripheral nerves. Immunomodulatory treatments used in these neuropathies aim to act at various steps of this pathogenic process. However, there are many phenotypic variants and, consequently, there is a significant difference in the response to immunotherapy between these neuropathies, as well as a need to improve our knowledge and long-term management of chronic forms.
Collapse
Affiliation(s)
- Jean-Marc Léger
- National Referral Center for Rare Neuromuscular Diseases, Institut Hospitalo-Universitaire de Neurosciences, University Hospital Pitié-Salpêtrière and University Pierre et Marie Curie (Paris VI), Paris, France.
| | - Raquel Guimarães-Costa
- National Referral Center for Rare Neuromuscular Diseases, Institut Hospitalo-Universitaire de Neurosciences, University Hospital Pitié-Salpêtrière and University Pierre et Marie Curie (Paris VI), Paris, France
| | - Cristina Muntean
- National Referral Center for Rare Neuromuscular Diseases, Institut Hospitalo-Universitaire de Neurosciences, University Hospital Pitié-Salpêtrière and University Pierre et Marie Curie (Paris VI), Paris, France
| |
Collapse
|
31
|
Reynaud Q, Killian M, Robles A, Mounsef F, Camdessanché JP, Mariat C, Cathébras P. Le rituximab dans la vraie vie : revue d’utilisation du rituximab de 2010 à 2013 au CHU de Saint-Étienne. Rev Med Interne 2015; 36:800-12. [DOI: 10.1016/j.revmed.2015.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 05/03/2015] [Accepted: 06/05/2015] [Indexed: 12/13/2022]
|
32
|
Hashimoto R, Koike H, Takahashi M, Ohyama K, Kawagashira Y, Iijima M, Sobue G. Uncompacted Myelin Lamellae and Nodal Ion Channel Disruption in POEMS Syndrome. J Neuropathol Exp Neurol 2015. [DOI: 10.1093/jnen/74.12.1127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Alix JJ, Hadjivassiliou M, Ali R, Slater D, Messenger AG, Rao DG. Sensory ganglionopathy with livedoid vasculopathy controlled by immunotherapy. Muscle Nerve 2014; 51:296-301. [DOI: 10.1002/mus.24452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2014] [Indexed: 12/26/2022]
Affiliation(s)
- James J.P. Alix
- Department of Clinical Neurophysiology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield S10 2JF
| | - Marios Hadjivassiliou
- Department of Neurology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield England
| | - Rokiahmah Ali
- Department of Histopathology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield England
| | - David Slater
- Department of Histopathology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield England
| | - Andrew G. Messenger
- Department of Dermatology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield England
| | - D. Ganesh Rao
- Department of Clinical Neurophysiology; Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust; Glossop Road Sheffield S10 2JF
| |
Collapse
|
34
|
Raheja D, Specht C, Simmons Z. Paraproteinemic neuropathies. Muscle Nerve 2014; 51:1-13. [DOI: 10.1002/mus.24471] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Divisha Raheja
- Department of Neurology; Penn State Hershey Medical Center; EC 037, 30 Hope Drive Hershey Pennsylvania 17033 USA
| | - Charles Specht
- Department of Neurology; Penn State Hershey Medical Center; EC 037, 30 Hope Drive Hershey Pennsylvania 17033 USA
- Department of Pathology; Penn State Hershey Medical Center; Hershey Pennsylvania USA
- Department of Ophthalmology; Penn State Hershey Medical Center; Hershey Pennsylvania USA
- Department of Neurosurgery; Penn State Hershey Medical Center; Hershey Pennsylvania USA
| | - Zachary Simmons
- Department of Neurology; Penn State Hershey Medical Center; EC 037, 30 Hope Drive Hershey Pennsylvania 17033 USA
| |
Collapse
|
35
|
Treatment of chronic inflammatory demyelinating polyneuropathy: from molecular bases to practical considerations. Autoimmune Dis 2014; 2014:201657. [PMID: 24527207 PMCID: PMC3914592 DOI: 10.1155/2014/201657] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disease of the peripheral nervous system, in which both cellular and humoral immune responses are involved. The disease is clinically heterogeneous with some patients displaying pure motor form and others also showing a variable degree of sensory dysfunction; disease evolution may also differ from patient to patient, since monophasic, progressive, and relapsing forms are reported. Underlying such clinical variability there is probably a broad spectrum of molecular dysfunctions that are and will be the target of therapeutic strategies. In this review we first explore the biological bases of current treatments and subsequently we focus on the practical management that must also take into account pharmacoeconomic issues.
Collapse
|
36
|
Zivković SA. Rituximab in the treatment of peripheral neuropathy associated with monoclonal gammopathy. Expert Rev Neurother 2014; 6:1267-74. [PMID: 17009914 DOI: 10.1586/14737175.6.9.1267] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peripheral neuropathy associated with immunoglobulin (Ig)M gammopathy and anti-myelin-associated glycoprotein antibodies is frequently treatment-resistant and different treatment regimens carry substantial toxicity and side effects. More recently, the chimeric anti-CD20 monoclonal antibody rituximab has shown benefits in the treatment of peripheral neuropathy associated with IgM gammopathy with a favorable side-effect profile. There are no published reports of its use in the treatment of neuropathy associated with IgG and IgA gammopathies. Rituximab is usually given at 375 mg/m(2) intravenously with four weekly doses that may be repeated after 6-12 months. Large controlled studies are still pending but rituximab is an exciting and promising treatment offering another option in the treatment of peripheral neuropathy associated with IgM monoclonal gammopathy.
Collapse
Affiliation(s)
- Sasa A Zivković
- Department of Neurology, University of Pittsburgh Medical Center, PUH F875, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
37
|
Souayah N, Noopur R, Tick-Chong PS. Beneficial effects of Rituximab in patients with anti-MAG (myelin-associated glycoprotein) neuropathy: case reports. Immunopharmacol Immunotoxicol 2013; 35:622-4. [DOI: 10.3109/08923973.2013.822507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Mostafa GA, Al-Ayadhi LY. The possible relationship between allergic manifestations and elevated serum levels of brain specific auto-antibodies in autistic children. J Neuroimmunol 2013; 261:77-81. [PMID: 23726766 DOI: 10.1016/j.jneuroim.2013.04.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/25/2013] [Accepted: 04/03/2013] [Indexed: 11/27/2022]
Abstract
Etiology of autism has become an area of a significant controversy. Allergy induced autism is an area of research wherein immune responses to some allergens may play a pathogenic role in autism. Allergy may induce the production of brain specific auto-antibodies in a subgroup of autistic children. We are the first to investigate the possible link between allergic manifestations and serum levels of both anti-myelin basic protein (anti-MBP) and anti-myelin associated glycoprotein (anti-MAG) brain-specific auto-antibodies, which were measured by ELISA method, in 42 autistic children in comparison to 42 healthy-matched children. Allergic manifestations (bronchial asthma, atopic dermatitis and/or allergic rhinitis) were found in 47.6% of autistic patients. Increased serum levels of anti-MBP and anti-MAG auto-antibodies were found in 57.1% and 66.7%, respectively of autistic children. In addition, 78.5% of autistic children had increased serum levels of both anti-MBP and/or anti-MAG auto-antibodies. Autistic patients with allergic manifestations had significantly higher serum levels of anti-MBP and anti-MAG auto-antibodies than those without these manifestations (P<0.001 and P=0.001, respectively). In conclusion, allergy may be a contributing factor to the increased serum levels of anti-MBP and anti-MAG auto-antibodies in some autistic children. Indeed, we need to know more about the links between allergy, immune system and brain in autism for finding new therapeutic modalities in autism.
Collapse
Affiliation(s)
- Gehan Ahmed Mostafa
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | |
Collapse
|
39
|
Léger JM, Viala K, Nicolas G, Créange A, Vallat JM, Pouget J, Clavelou P, Vial C, Steck A, Musset L, Marin B. Placebo-controlled trial of rituximab in IgM anti-myelin-associated glycoprotein neuropathy. Neurology 2013; 80:2217-25. [PMID: 23667063 DOI: 10.1212/wnl.0b013e318296e92b] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether rituximab 375 mg/m(2) was efficacious in patients with immunoglobulin M (IgM) anti-myelin-associated glycoprotein antibody demyelinating neuropathy (IgM anti-MAG demyelinating neuropathy). METHODS Fifty-four patients with IgM anti-MAG demyelinating neuropathy were enrolled in this randomized, double-blind, placebo-controlled trial. The inclusion criteria were inflammatory neuropathy cause and treatment (INCAT) sensory score (ISS) ≥4 and visual analog pain scale >4 or ataxia score ≥2. The primary outcome was mean change in ISS at 12 months. RESULTS Twenty-six patients were randomized to a group receiving 4 weekly infusions of 375 mg/m(2) rituximab, and 28 patients to placebo. Intention-to-treat analysis, with imputation of missing ISS values by the last observation carried forward method, showed a lack of mean change in ISS at 12 months, 1.0 ± 2.7 in the rituximab group, and 1.0 ± 2.8 in the placebo group. However, changes were observed, in per protocol analysis at 12 months, for the number of patients with an improvement of at least 2 points in the INCAT disability scale (p = 0.027), the self-evaluation scale (p = 0.016), and 2 subscores of the Short Form-36 questionnaire. CONCLUSIONS Although primary outcome measures provide no evidence to support the use of rituximab in IgM anti-MAG demyelinating neuropathy, there were improvements in several secondary outcomes in per protocol analysis. LEVEL OF EVIDENCE This study provides Class I evidence that rituximab is ineffective in improving ISS in patients with IgM anti-MAG demyelinating neuropathy.
Collapse
Affiliation(s)
- Jean-Marc Léger
- Department of Neurology, University Hospital Pitié-Salpêtrière, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rojas-García R, Gallardo E, Illa I. Paraproteinemic neuropathies. Presse Med 2013; 42:e225-34. [PMID: 23618626 DOI: 10.1016/j.lpm.2013.02.329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 02/04/2013] [Accepted: 02/12/2013] [Indexed: 12/13/2022] Open
Abstract
The term paraproteinemic neuropathy refers to a heterogeneous group of neuropathies, which share the common feature of a homogeneous immunoglobulin in the serum. The presence of a monoclonal gammopathy indicates an underlying clonal B-cell expansion, which may appear in the context of a lymphoproliferative disorder. If a neoplastic origin of the gammopathy is identified, the treatment should be targeted to the neoplasm. In most patients, however, the monoclonal gammopathy is not associated with malignant haematological disorders, and is defined as monoclonal gammopathy of undetermined significance.
Collapse
Affiliation(s)
- Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
41
|
Kodaira M, Yamamoto K. Rituximab Improves Subclinical Temporal Dispersion of Distal Compound Muscle Action Potential in Anti-MAG/SGPG Neuropathy Associated with Waldenström Macroglobulinemia: A Case Report. Case Rep Neurol 2013; 5:34-9. [PMID: 23525653 PMCID: PMC3604870 DOI: 10.1159/000348395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Patients with anti-myelin-associated glycoprotein (MAG)/sulfated glucuronyl paragloboside (SGPG) neuropathy associated with Waldenström macroglobulinemia show demyelinating neuropathy, but the temporal dispersion of distal compound muscle action potential (CMAP) in motor nerve conduction studies (NCS), which represents heterogeneous demyelination at the motor nerve terminal, is rare. We report on a 70-year-old man with anti-MAG/SGPG neuropathy associated with Waldenström macroglobulinemia; he had a 2-year history of mild dysesthesia of the foot sole without any motor symptoms. He showed marked temporal dispersion of distal CMAP in the tibial nerve with other demyelinating findings in the NCS. The temporal dispersion of distal CMAP in the tibial nerve improved significantly, and motor function was again normal 1 year after rituximab monotherapy. The temporal dispersion of distal CMAP in anti-MAG/SGPG neuropathy is rare, but it could occur from an early stage when the patients show mild or no motor symptoms. Rituximab therapy before secondary axonal degeneration has great potential to reverse the effects of the demyelination including the temporal dispersion of distal CMAP, and to prevent the deterioration of neuropathy in anti-MAG/SGPG neuropathy.
Collapse
Affiliation(s)
- Minori Kodaira
- Department of Neurology, Nagano Municipal Hospital, Nagano, Japan
| | | |
Collapse
|
42
|
Multifocal motor neuropathy, multifocal acquired demyelinating sensory and motor neuropathy, and other chronic acquired demyelinating polyneuropathy variants. Neurol Clin 2013; 31:533-55. [PMID: 23642723 DOI: 10.1016/j.ncl.2013.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic acquired demyelinating neuropathies (CADP) constitute an important group of immune neuromuscular disorders affecting myelin. This article discusses CADP with emphasis on multifocal motor neuropathy, multifocal acquired demyelinating sensory and motor neuropathy, distal acquired demyelinating symmetric neuropathy, and less common variants. Although each of these entities has distinctive laboratory and electrodiagnostic features that aid in their diagnosis, clinical characteristics are of paramount importance in diagnosing specific conditions and determining the most appropriate therapies. Knowledge regarding pathogenesis, diagnosis, and management of these disorders continues to expand, resulting in improved opportunities for identification and treatment.
Collapse
|
43
|
Abstract
The association of neuropathy with monoclonal gammopathy has been known for several years, even if the clinical and pathogenetic relevance of this association is not completely defined. This is not a marginal problem since monoclonal gammopathy is present in 1-3% of the population above 50 years in whom it is often asymptomatic, and in at least 8% of patients is associated with a symptomatic neuropathy, representing one of the leading causes of neuropathy in aged people. Monoclonal gammopathy may result from malignant lymphoproliferative diseases including multiple myeloma or solitary plasmocytoma, Waldenström's macroglobulinemia (WM), other IgM-secreting lymphoma or chronic lymphocytic leukemia, and primary systemic amyloidosis (AL). In most instances it is not associated with any of these disorders and is defined monoclonal gammopathy of undetermined significance (MGUS) for its possible, though infrequent, evolution into malignant forms. Several data support the pathogenetic role of the monoclonal gammopathy in the neuropathy particularly when of IgM isotype where IgM reactivity to several neural antigens has been reported. Increased levels of VEGF have been implicated in POEMS syndrome. However, there are as yet no defined therapies for these neuropathies, as their efficacy has not been confirmed in randomized trials.
Collapse
Affiliation(s)
- Eduardo Nobile-Orazio
- 2nd Neurology, Department of Translational Medicine, Milan University, IRCCS Humanitas Clinical Institute, Rozzano, Milan, Italy.
| |
Collapse
|
44
|
The relationship between the increased frequency of serum antineuronal antibodies and the severity of autism in children. Eur J Paediatr Neurol 2012; 16:464-8. [PMID: 22226851 DOI: 10.1016/j.ejpn.2011.12.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/05/2011] [Accepted: 12/18/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND Autism may involve an autoimmune pathogenesis. Immunotherapy may have a role in autistic children who have brain auto-antibodies. AIM This study aimed to investigate the frequency of serum antineuronal auto-antibodies, as indicators of the presence of autoimmunity to brain, in a group of autistic children. We are the first to measure the relationship between these antibodies and the degree of the severity of autism. METHODS Serum antineuronal antibodies were measured, by indirect immunofluorescence technique, in 80 autistic children, aged between 6 and 12 years, in comparison to 80 healthy-matched children. The severity of autism was assessed by using the Childhood Autism Rating Scale. RESULTS Autistic children had significantly higher percent positivity of serum antineuronal antibodies (62.5%) than healthy controls (5%), P<0.001. The frequency of the positivity of serum antineuronal antibodies was significantly higher in children with severe autism (87.5%) than children with mild to moderate autism (25%), P<0.001. Similarly, the frequency of the positivity of these antibodies was significantly higher in female children with autism (90%) than male autistic children (53.3%), P=0.001. CONCLUSIONS Serum antineuronal antibodies were found in a subgroup of autistic children and they were significantly correlated to the severity of autism. Thus, autism may be, in part, one of the pediatric autoimmune neuropsychiatric disorders. Further wide-scale studies are warranted to shed light on the etiopathogenic role of antineuronal antibodies in autism. The role of immunotherapy in autistic patients, who are seropositive for antineuronal antibodies, should also be studied.
Collapse
|
45
|
Lunn MPT, Nobile-Orazio E. Immunotherapy for IgM anti-myelin-associated glycoprotein paraprotein-associated peripheral neuropathies. Cochrane Database Syst Rev 2012:CD002827. [PMID: 22592686 DOI: 10.1002/14651858.cd002827.pub3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Serum monoclonal anti-myelin-associated glycoprotein antibodies may be pathogenic in some people with immunoglobulin M (IgM) paraprotein and demyelinating neuropathy. Immunotherapies aimed at reducing the level of these antibodies might be expected to be beneficial. This is an update of a review first published in 2003 and previously updated in 2006. OBJECTIVES To assess the effects of immunotherapy for IgM anti-myelin-associated glycoprotein paraprotein-associated demyelinating peripheral neuropathy. SEARCH METHODS We searched the Cochrane Neuromuscular Disease Group Specialized Register 6 June 2011), CENTRAL (2011, Issue 2), MEDLINE (January 1966 to May 2011) and EMBASE (January 1980 to May 2011) for controlled trials. We also checked bibliographies and contacted authors and experts in the field. SELECTION CRITERIA We included randomised or quasi-randomised controlled trials involving participants of any age treated with any type of immunotherapy for anti-myelin-associated glycoprotein antibody-associated demyelinating peripheral neuropathy with monoclonal gammopathy of undetermined significance and of any severity.Our primary outcome measure was change in the Neuropathy Impairment Scale or Modified Rankin Scale at six months after randomisation. Secondary outcome measures were: Neuropathy Impairment Scale or the Modified Rankin Score at 12 months after randomisation; 10-metre walk time, subjective clinical scores and electrophysiological parameters at six and 12 months after randomisation; IgM paraprotein levels and anti-myelin-associated glycoprotein antibody titres at six months after randomisation; and adverse effects of treatments. DATA COLLECTION AND ANALYSIS The two authors independently selected studies. Two authors independently assessed the risk of bias in included studies. MAIN RESULTS We identified seven eligible trials (182 participants), which tested intravenous immunoglobulin, alfa interferon alfa-2a, plasma exchange, cyclophosphamide and steroids, and rituximab. Only two trials, of intravenous immunoglobulin (with 33 participants, including 20 with antibodies against myelin-associated glycoprotein), had comparable interventions and outcomes, but both were short-term trials.There were no clinical or statistically significant benefits of the treatments used on the outcomes predefined for this review, but not all the predefined outcomes were used in every included trial. Intravenous immunoglobulin showed a statistical benefit in terms of improvement in Modified Rankin Scale at two weeks and 10-metre walk time at four weeks. Cyclophosphamide failed to show any benefit in the trial's primary outcome, and showed a barely significant benefit in the primary outcome specified here, but some toxic adverse events were identified. A trial of rituximab was of poor methodological quality with a high risk of bias and a further larger study is awaited. Serious adverse events were few in the other trials. AUTHORS' CONCLUSIONS There is inadequate reliable evidence from trials of immunotherapies in anti-myelin-associated glycoprotein paraproteinaemic neuropathy to form an evidence base supporting any particular immunotherapy treatment. There is very low quality evidence of benefit from rituximab. Large well designed randomised trials of at least six to 12 months duration are required to assess existing or novel therapies, preferably employing unified, consistent, well designed, responsive and valid outcome measures.
Collapse
Affiliation(s)
- Michael P T Lunn
- Department of Neurology and MRC Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK.
| | | |
Collapse
|
46
|
Abstract
Autoimmunity plays a major role in the pathogenesis of many neuromuscular disorders such as chronic inflammatory demyelinating polyneuropathy, Guillain-Barré syndrome, polymyositis, dermatomyositis, myasthenia gravis, Lambert Eaton syndrome, and stiff person syndrome. Although most of these disorders respond favorably to the commonly used immunomodulatory agents such as steroids, intravenous gamma globulin, plasmapheresis, and chemotherapy, some are initially refractory, whereas others gradually lose responsiveness. Therefore, alternative, selective, and novel immunosuppressive agents are used to treat these cases. Among these agents, rituximab has shown promise in some of the neuromuscular disorders with minimal side effects. Rituximab is a genetically engineered antibody that depletes CD20+ B-cells and is Food and Drug Administration- approved for treatment of non-Hodgkin lymphoma, CD20+ CLL, and rheumatoid arthritis. It carries a favorable side effects profile. However, evidence of efficacy is limited to case series and large prospective randomized controlled trials are lacking. In this article, we review and discuss the available literature on rituximab in treatment of various autoimmune neuromuscular diseases.
Collapse
|
47
|
Maurer MA, Rakocevic G, Leung CS, Quast I, Lukačišin M, Goebels N, Münz C, Wardemann H, Dalakas M, Lünemann JD. Rituximab induces sustained reduction of pathogenic B cells in patients with peripheral nervous system autoimmunity. J Clin Invest 2012; 122:1393-402. [PMID: 22426210 DOI: 10.1172/jci58743] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 02/01/2012] [Indexed: 11/17/2022] Open
Abstract
The B cell-depleting IgG1 monoclonal antibody rituximab can persistently suppress disease progression in some patients with autoimmune diseases. However, the mechanism underlying these long-term beneficial effects has remained unclear. Here, we evaluated Ig gene usage in patients with anti-myelin-associated glycoprotein (anti-MAG) neuropathy, an autoimmune disease of the peripheral nervous system that is mediated by IgM autoantibodies binding to MAG antigen. Patients with anti-MAG neuropathy showed substantial clonal expansions of blood IgM memory B cells that recognized MAG antigen. The group of patients showing no clinical improvement after rituximab therapy were distinguished from clinical responders by a higher load of clonal IgM memory B cell expansions before and after therapy, by persistence of clonal expansions despite efficient peripheral B cell depletion, and by a lack of substantial changes in somatic hypermutation frequencies of IgM memory B cells. We infer from these data that the effectiveness of rituximab therapy depends on efficient depletion of noncirculating B cells and is associated with qualitative immunological changes that indicate reconfiguration of B cell memory through sustained reduction of autoreactive clonal expansions. These findings support the continued development of B cell-depleting therapies for autoimmune diseases.
Collapse
Affiliation(s)
- Michael A Maurer
- Institute of Experimental Immunology, Department of Neuroinflammation, University of Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Monoclonal antibodies in inflammatory disease of the muscle and peripheral nervous system. NEUROLOGÍA (ENGLISH EDITION) 2012. [DOI: 10.1016/j.nrleng.2010.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
49
|
Zara G, Zambello R, Ermani M. Neurophysiological and clinical responses to rituximab in patients with anti-MAG polyneuropathy. Clin Neurophysiol 2011; 122:2518-22. [DOI: 10.1016/j.clinph.2011.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 05/19/2011] [Accepted: 05/21/2011] [Indexed: 12/29/2022]
|
50
|
Gruson B, Ghomari K, Beaumont M, Garidi R, Just A, Merle P, Merlusca L, Marolleau JP, Royer B. Long-term response to rituximab and fludarabine combination in IgM anti-myelin-associated glycoprotein neuropathy. J Peripher Nerv Syst 2011; 16:180-5. [DOI: 10.1111/j.1529-8027.2011.00343.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|