1
|
Allameen NA, Salam S, Reddy V, Machado PM. Inclusion body myositis and immunosenescence: current evidence and future perspectives. Rheumatology (Oxford) 2025; 64:952-961. [PMID: 39504446 PMCID: PMC11879327 DOI: 10.1093/rheumatology/keae614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024] Open
Abstract
IBM remains an enigmatic and complex muscle disorder where a deeper understanding of disease pathomechanisms and the identification of potential genetic contributors represent an unmet need. The absence of effective treatments has spurred endeavours to reassess the interplay between degeneration, including autophagy, mitochondrial dysfunction and proteasomal dysregulation, and autoimmunity. IBM is unique among the other idiopathic inflammatory myopathies owing to its molecular signature involving highly differentiated cytotoxic T cells that evade immune regulation. This has led to a resurgence of interest in the development of immunomodulatory therapy. This review discusses the potential role of cellular immunosenescence in sustaining inflammation and/or fibrotic remodelling observed in IBM and appraises the rationale for some potential therapeutic approaches to mitigate disease progression.
Collapse
Affiliation(s)
- Nur Azizah Allameen
- Division of Rheumatology, Department of Medicine, Woodlands Health, Woodlands, Singapore
- Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sharfaraz Salam
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Venkat Reddy
- Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Pedro M Machado
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Rheumatology, Division of Medicine, University College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| |
Collapse
|
2
|
Neves A, Viveiros L, Venturelli V, Isenberg DA. Where are we now in biologic drugs for myositis? Rheumatology (Oxford) 2024; 63:2938-2947. [PMID: 38321569 DOI: 10.1093/rheumatology/keae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/08/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a rare and heterogeneous group of chronic autoimmune disorders. Up to 40% of IIM patients have long-term sequelae and significant functional disability. Its management can be challenging and new therapies are badly needed. The small number of cases with diverse presentations and different diagnostic criteria significantly affect clinical trial results. Only IVIG has been internationally approved for IIM patients. Most clinical trials of new biologic therapies have failed to meet their primary endpoints in IIM, with only one biologic drug recommended for refractory IIM treatment (rituximab), although not approved. We review several new emerging biologic drugs, including B cell depletion therapies, abatacept, Janus kinase inhibitors, and aldesleukin. Encouragingly, some phase II randomized controlled trials have evaluated the efficacy and safety of new biologics in IIM, demonstrating an improvement in clinical and laboratory measures.
Collapse
Affiliation(s)
- Ana Neves
- Internal Medicine Department, Centro Hospitalar Universitário de São João, Oporto, Portugal
| | - Luísa Viveiros
- Internal Medicine Department, Centro Hospitalar Universitário de Santo António, Oporto, Portugal
| | - Veronica Venturelli
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - David A Isenberg
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| |
Collapse
|
3
|
Veldkamp SR, van Wijk F, van Royen-Kerkhof A, Jansen MH. Personalised medicine in juvenile dermatomyositis: From novel insights in disease mechanisms to changes in clinical practice. Best Pract Res Clin Rheumatol 2024; 38:101976. [PMID: 39174374 DOI: 10.1016/j.berh.2024.101976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
Juvenile dermatomyositis is characterized by childhood-onset chronic inflammation of the muscles and skin, with potential involvement of other organs. Patients are at risk for long-term morbidity due to insufficient disease control and steroid-related toxicity. Personalised treatment is challenged by a lack of validated tools that can reliably predict treatment response and monitor ongoing (subclinical) inflammation, and by a lack of evidence regarding the best choice of medication for individual patients. A better understanding of the involved disease mechanisms could reveal potential biomarkers and novel therapeutic targets. In this review, we highlight the most relevant immune and non-immune mechanisms, elucidating the effects of interferon overexpression on tissue alongside the interplay between the interferon signature, mitochondrial function, and immune cells. We review mechanism-based biomarkers that are promising for clinical implementation, and the latest advances in targeted therapy development. Finally, we discuss key steps needed for translating these discoveries into clinical practice.
Collapse
Affiliation(s)
- Saskia R Veldkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Annet van Royen-Kerkhof
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marc Ha Jansen
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Jensen KY, Nielsen JL, Aagaard P, Jacobsen M, Jørgensen AN, Bech RD, Frandsen U, Diederichsen LP, Schrøder HD. Effects of sporadic inclusion body myositis on skeletal muscle fibre type specific morphology and markers of regeneration and inflammation. Rheumatol Int 2024; 44:1077-1087. [PMID: 38581449 PMCID: PMC11108868 DOI: 10.1007/s00296-024-05567-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/23/2024] [Indexed: 04/08/2024]
Abstract
Sporadic inclusion body myositis (sIBM) is a subgroup of idiopathic inflammatory myopathies characterised by progressive muscle weakness and skeletal muscle inflammation. Quantitative data on the myofibre morphology in sIBM remains scarce. Further, no previous study has examined fibre type association of satellite cells (SC), myonuclei number, macrophages, capillaries, and myonuclear domain (MD) in sIBM patients. Muscle biopsies from sIBM patients (n = 18) obtained previously (NCT02317094) were included in the analysis for fibre type-specific myofibre cross-sectional area (mCSA), SCs, myonuclei and macrophages, myonuclear domain, and capillarisation. mCSA (p < 0.001), peripheral myonuclei (p < 0.001) and MD (p = 0.005) were higher in association with type 1 (slow-twitch) than type 2 (fast-twitch) fibres. Conversely, quiescent SCs (p < 0.001), centrally placed myonuclei (p = 0.03), M1 macrophages (p < 0.002), M2 macrophages (p = 0.013) and capillaries (p < 0.001) were higher at type 2 fibres compared to type 1 fibres. In contrast, proliferating (Pax7+/Ki67+) SCs (p = 0.68) were similarly associated with each fibre type. Type 2 myofibres of late-phase sIBM patients showed marked signs of muscle atrophy (i.e. reduced mCSA) accompanied by higher numbers of associated quiescent SCs, centrally placed myonuclei, macrophages and capillaries compared to type 1 fibres. In contrast, type 1 fibres were suffering from pathological enlargement with larger MDs as well as fewer nuclei and capillaries per area when compared with type 2 fibres. More research is needed to examine to which extent different therapeutic interventions including targeted exercise might alleviate these fibre type-specific characteristics and countermeasure their consequences in impaired functional performance.
Collapse
Affiliation(s)
- Kasper Yde Jensen
- Copenhagen Research Center for Autoimmune Connective Tissue Diseases (COPEACT), Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Juliane Maries Vej 10, 2100, Copenhagen, Denmark.
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark.
- Department of Pathology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark.
| | - Jakob Lindberg Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Per Aagaard
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Mikkel Jacobsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Anders Nørkær Jørgensen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Rune Dueholm Bech
- Department of Orthopaedics and Traumatology, Zealand University Hospital, Koege, Denmark
| | - Ulrik Frandsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Louise Pyndt Diederichsen
- Copenhagen Research Center for Autoimmune Connective Tissue Diseases (COPEACT), Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Juliane Maries Vej 10, 2100, Copenhagen, Denmark
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Henrik Daa Schrøder
- Department of Pathology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| |
Collapse
|
5
|
Guglielmi V, Cheli M, Tonin P, Vattemi G. Sporadic Inclusion Body Myositis at the Crossroads between Muscle Degeneration, Inflammation, and Aging. Int J Mol Sci 2024; 25:2742. [PMID: 38473988 PMCID: PMC10932328 DOI: 10.3390/ijms25052742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Sporadic inclusion body myositis (sIBM) is the most common muscle disease of older people and is clinically characterized by slowly progressive asymmetrical muscle weakness, predominantly affecting the quadriceps, deep finger flexors, and foot extensors. At present, there are no enduring treatments for this relentless disease that eventually leads to severe disability and wheelchair dependency. Although sIBM is considered a rare muscle disorder, its prevalence is certainly higher as the disease is often undiagnosed or misdiagnosed. The histopathological phenotype of sIBM muscle biopsy includes muscle fiber degeneration and endomysial lymphocytic infiltrates that mainly consist of cytotoxic CD8+ T cells surrounding nonnecrotic muscle fibers expressing MHCI. Muscle fiber degeneration is characterized by vacuolization and the accumulation of congophilic misfolded multi-protein aggregates, mainly in their non-vacuolated cytoplasm. Many players have been identified in sIBM pathogenesis, including environmental factors, autoimmunity, abnormalities of protein transcription and processing, the accumulation of several toxic proteins, the impairment of autophagy and the ubiquitin-proteasome system, oxidative and nitrative stress, endoplasmic reticulum stress, myonuclear degeneration, and mitochondrial dysfunction. Aging has also been proposed as a contributor to the disease. However, the interplay between these processes and the primary event that leads to the coexistence of autoimmune and degenerative changes is still under debate. Here, we outline our current understanding of disease pathogenesis, focusing on degenerative mechanisms, and discuss the possible involvement of aging.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Cellular and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marta Cheli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Gaetano Vattemi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| |
Collapse
|
6
|
Quinn C, Moulton K, Farwell M, Le W, Wilson I, Goel N, McConathy J, Greenberg SA. Imaging With PET/CT of Diffuse CD8 T-Cell Infiltration of Skeletal Muscle in Patients With Inclusion Body Myositis. Neurology 2023; 101:e1158-e1166. [PMID: 37487752 PMCID: PMC10513879 DOI: 10.1212/wnl.0000000000207596] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/12/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Inclusion body myositis (IBM) is a progressive autoimmune skeletal muscle disease in which cytotoxic CD8+ T cells infiltrate muscle and destroy myofibers. IBM has required a muscle biopsy for diagnosis. Here, we administered to patients with IBM a novel investigational PET tracer 89Zr-Df-crefmirlimab for in vivo imaging of whole body skeletal muscle CD8 T cells. This technology has not previously been applied to patients with autoimmune disease. METHODS Four patients with IBM received 89Zr-Df-crefmirlimab followed by PET/CT imaging 24 hours later, and the results were compared with similar imaging of age-matched patients with cancer. Mean standardized uptake value (SUVmean) was measured for reference tissues using spherical regions of interest (ROIs). RESULTS 89Zr-Df-crefmirlimab was safe and well-tolerated. PET imaging demonstrated diffusely increased uptake qualitatively and quantitatively in IBM limb musculature. Quantitation of 89Zr-Df-crefmirlimab intensity in ROIs demonstrated particularly increased CD8 T-cell infiltration in patients with IBM compared with patients with cancer in quadriceps (SUVmean 0.55 vs 0.20, p < 0.0001), biceps brachii (0.62 vs 0.26, p < 0.0001), triceps (0.61 vs 0.25, p = 0.0005), and forearm finger flexors (0.71 vs 0.23, p = 0.008). DISCUSSION 89Zr-Df-crefmirlimab uptake in muscles of patients with IBM was present at an intensity greater than the comparator population. The ability to visualize whole body in vivo cytotoxic T-cell tissue infiltration in the autoimmune disease IBM may hold utility as a biomarker for diagnosis, disease activity, and therapeutic development and potentially be applicable to other diseases with cytotoxic T-cell autoimmunity.
Collapse
Affiliation(s)
- Colin Quinn
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA.
| | - Kelsey Moulton
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - Michael Farwell
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - William Le
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - Ian Wilson
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - Niti Goel
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - Jonathan McConathy
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| | - Steven A Greenberg
- From the Departments of Neurology (C.Q., K.M.), and Radiology (M.F.), University of Pennsylvania, Perelman School of Medicine, Philadelphia; ImaginAb (W.L., I.W.), Inc., Inglewood, CA; Department of Medicine (N.G.), Duke University School of Medicine, Durham, NC; Abcuro, Inc., Newton, MA; Department of Radiology (J.M.), University of Alabama at Birmingham, Heersink School of Medicine; and Department of Neurology (S.A.G.), Brigham and Women's Hospital and Boston Childrens Hospital, Harvard Medical School, MA
| |
Collapse
|
7
|
Michelle EH, Pinal-Fernandez I, Casal-Dominguez M, Albayda J, Paik JJ, Tiniakou E, Adler B, Mecoli CA, Danoff SK, Christopher-Stine L, Mammen AL, Lloyd TE. Clinical Subgroups and Factors Associated With Progression in Patients With Inclusion Body Myositis. Neurology 2023; 100:e1406-e1417. [PMID: 36690456 PMCID: PMC10065210 DOI: 10.1212/wnl.0000000000206777] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 11/18/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Sporadic inclusion body myositis (IBM) is the most common acquired myopathy in individuals older than 50 years. The disorder is slowly progressive, and although many therapies have been investigated, response has generally been poor. Clinical heterogeneity may influence treatment responsiveness; however, data regarding heterogeneity in IBM are limited and often conflicting. We aim to identify clinically distinct subgroups within a large IBM cohort and prognostic factors for disease progression. METHODS Clinical, histologic, radiologic, and electrophysiologic data were analyzed for all patients with IBM and other forms of myositis enrolled in a longitudinal cohort from The Johns Hopkins Myositis Center from 2003 to 2018. Patients with IBM were included if they met at least one of the following criteria: Griggs possible, European Neuromuscular Centre 2011 probable, or Lloyd-Greenberg data-derived criteria for IBM. Univariate, multivariate, and graphical analyses were used to identify prognostic factors in patients with IBM. Thus, linear and logistic regressions were used to adjust for potential confounding variables. The evolution of creatine kinase and muscle strength was studied using multilevel linear regression models. Nonmodifiable risk factors (sex, race, disease duration, and age at the onset of first symptoms) were used as adjusting covariates for the regression analyses. RESULTS Among the 335 patients meeting the inclusion criteria for IBM, 64% were male with an average age of disease onset of 58.7 years and delay to diagnosis of 5.2 years. Initial misdiagnosis (52%) and immunosuppressant treatment (42%) were common. Less than half (43%) of muscle biopsies demonstrated all 3 pathologic hallmarks: endomysial inflammation, mononuclear cell invasion, and rimmed vacuoles. Black patients had significantly weaker arm abductors, hip flexors, and knee flexors compared with non-Black patients. Female patients had stronger finger flexors and knee extensors compared with their male counterparts. Younger age (<50 years) at onset was not associated with increased weakness. DISCUSSION Our study demonstrates that female and Black patients have distinct clinical phenotypes and trajectories within the overarching IBM clinical phenotype. These subgroups may have different responses to therapies, which may influence the design of future clinical trials in IBM.
Collapse
Affiliation(s)
- Elizabeth Harlan Michelle
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Iago Pinal-Fernandez
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Maria Casal-Dominguez
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Jemima Albayda
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Julie J Paik
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Eleni Tiniakou
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Brittany Adler
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Christopher A Mecoli
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Sonye K Danoff
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Lisa Christopher-Stine
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Andrew L Mammen
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Thomas E Lloyd
- From the Departments of Neurology (E.H.M., I.P.-F., M.C.-D., A.L.M., T.E.L.), and Medicine (J.A., J.J.P., E.T., B.A., C.A.M., S.K.D., L.C.-S.), Johns Hopkins University School of Medicine, Baltimore, MD; Muscle Disease Unit (I.P.-F., M.C.-D., A.L.M.), Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; and Faculty of Health Sciences and Faculty of Computer Science (I.P.-F.), Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain.
| |
Collapse
|
8
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Treatment of autoimmunity: The impact of disease-modifying therapies in multiple sclerosis and comorbid autoimmune disorders. Autoimmun Rev 2023; 22:103312. [PMID: 36924922 DOI: 10.1016/j.autrev.2023.103312] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
More than 10 disease-modifying therapies (DMT) are approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) and new therapeutic options are on the horizon. Due to different underlying therapeutic mechanisms, a more individualized selection of DMTs in MS is possible, taking into account the patient's current situation. Therefore, concomitant treatment of various comorbid conditions, including autoimmune mediated disorders such as rheumatoid arthritis, should be considered in MS patients. Because the pathomechanisms of autoimmunity partially overlap, DMT could also treat concomitant inflammatory diseases and simplify the patient's treatment. In contrast, the exacerbation and even new occurrence of several autoimmune diseases have been reported as a result of immunomodulatory treatment of MS. To simplify treatment and avoid disease exacerbation, knowledge of the beneficial and adverse effects of DMT in other autoimmune disorders is critical. Therefore, we conducted a literature search and described the beneficial and adverse effects of approved and currently studied DMT in a large number of comorbid autoimmune diseases, including rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel diseases, cutaneous disorders including psoriasis, Sjögren´s syndrome, systemic lupus erythematosus, systemic vasculitis, autoimmune hepatitis, and ocular autoimmune disorders. Our review aims to facilitate the selection of an appropriate DMT in patients with MS and comorbid autoimmune diseases.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany..
| | - Matthias Schefzyk
- Department of Dermatology, Allergology and Venerology, Hannover Medical School, 30625 Hannover, Germany..
| | - Miriam Wiestler
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| |
Collapse
|
9
|
Nagy S, Khan A, Machado PM, Houlden H. Inclusion body myositis: from genetics to clinical trials. J Neurol 2023; 270:1787-1797. [PMID: 36399165 PMCID: PMC9971047 DOI: 10.1007/s00415-022-11459-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022]
Abstract
Inclusion body myositis (IBM) belongs to the group of idiopathic inflammatory myopathies and is characterized by a slowly progressive disease course with asymmetric muscle weakness of predominantly the finger flexors and knee extensors. The disease leads to severe disability and most patients lose ambulation due to lack of curative or disease-modifying treatment options. Despite some genes reported to be associated with hereditary IBM (a distinct group of conditions), data on the genetic susceptibility of sporadic IBM are very limited. This review gives an overview of the disease and focuses on the current genetic knowledge and potential therapeutic implications.
Collapse
Affiliation(s)
- Sara Nagy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK.
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Alaa Khan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- Molecular Diagnostic Unit, Clinical Laboratory Department, King Abdullah Medical City in Makkah, Mecca, Saudi Arabia
| | - Pedro M Machado
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
- Division of Medicine, Centre for Rheumatology, University College London, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
10
|
Abstract
The autoimmune inflammatory myopathies constitute a heterogeneous group of acquired myopathies that have in common the presence of endomysial inflammation and moderate to severe muscle weakness. Based on currently evolved distinct clinical, histologic, immunopathologic, and autoantibody features, these disorders can be best classified as dermatomyositis, necrotizing autoimmune myositis, antisynthetase syndrome-overlap myositis, and inclusion body myositis. Although polymyositis is no longer considered a distinct subset but rather an extinct entity, it is herein described because its clinicopathologic information has provided over many years fundamental information on T-cell-mediated myocytotoxicity, especially in reference to inclusion body myositis. Each inflammatory myopathy subset has distinct immunopathogenesis, prognosis, and response to immunotherapies, necessitating the need to correctly diagnose each subtype from the outset and avoid disease mimics. The paper describes the main clinical characteristics that aid in the diagnosis of each myositis subtype, highlights the distinct features on muscle morphology and immunopathology, elaborates on the potential role of autoantibodies in pathogenesis or diagnosis , and clarifies common uncertainties in reference to putative triggering factors such as statins and viruses including the 2019-coronavirus-2 pandemic. It extensively describes the main autoimmune markers related to autoinvasive myocytotoxic T-cells, activated B-cells, complement, cytokines, and the possible role of innate immunity. The concomitant myodegenerative features seen in inclusion body myositis along with their interrelationship between inflammation and degeneration are specifically emphasized. Finally, practical guidelines on the best therapeutic approaches are summarized based on up-to-date knowledge and controlled studies, highlighting the prospects of future immunotherapies and ongoing controversies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States; Neuroimmunology Unit National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
11
|
Cellular senescence in neuroinflammatory disease: new therapies for old cells? Trends Mol Med 2022; 28:850-863. [DOI: 10.1016/j.molmed.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/08/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022]
|
12
|
Connolly CM, Plomp L, Paik JJ, Allenbach Y. Possible future avenues for myositis therapeutics: DM, IMNM and IBM. Best Pract Res Clin Rheumatol 2022; 36:101762. [PMID: 35778272 DOI: 10.1016/j.berh.2022.101762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Idiopathic inflammatory myopathies (IIMs) represent a heterogeneous group of systemic autoimmune diseases characterized by immune-mediated muscle injury. As insights into pathogenesis of IIM evolve, novel therapeutic strategies have become available to optimize outcomes. Herein, we summarize novel and emerging strategies in the management of dermatomyositis (DM), immunemediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM).
Collapse
Affiliation(s)
- Caoilfhionn M Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lotta Plomp
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Julie J Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France.
| |
Collapse
|
13
|
Clinical implication of denervation in sporadic inclusion body myositis. J Neurol Sci 2022; 439:120317. [PMID: 35709642 DOI: 10.1016/j.jns.2022.120317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/08/2022] [Accepted: 06/03/2022] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Sporadic inclusion body myositis (sIBM) is often accompanied by signs suggestive of denervation on electromyography (EMG), which mimics neurogenic disorders. Hence, the current study aimed to assess reinnervation after denervation in sIBM and its clinical impllcation. METHODS We retrospectively examined consecutive muscle biopsy specimens collected from 109 sIBM patients who were referred to our institution for diagnostic muscle biopsy from 2001 to 2018. Reinnervation after denervation in sIBM patients was assessed via muscle biopsy and EMG. The levels of acetylcholine receptor subunit γ (Chrng) and muscle-specific kinase (MuSK) mRNA, which are markers of denervation, were examined using real-time polymerase chain reaction. Response to treatment was defined as an increase of grade 1 or higher in two or more muscle groups as assessed using the Medical Research Council scale. RESULTS In total, 93 (85.3%) of 109 sIBM patients had reinnervation after denervation on histological examination and/or EMG. The mean disease duration before biopsy was significantly longer in patients with reinnervation after denervation than in those without (p < 0.00001). Patients with denervation had significantly higher levels of Chrng and MuSK mRNA than those without. The proportion of patients who responded to immunosuppressive therapies was smaller in the patients with denervation than those without (p < 0.05). However, there was no significant difference regarding time from onset to using a walking aid between the two groups. DISCUSSION Reinnervation after denervation is associated with disease duration and short-term response to therapy in individuals with sIBM.
Collapse
|
14
|
Zschüntzsch J, Meyer S, Shahriyari M, Kummer K, Schmidt M, Kummer S, Tiburcy M. The Evolution of Complex Muscle Cell In Vitro Models to Study Pathomechanisms and Drug Development of Neuromuscular Disease. Cells 2022; 11:1233. [PMID: 35406795 PMCID: PMC8997482 DOI: 10.3390/cells11071233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Many neuromuscular disease entities possess a significant disease burden and therapeutic options remain limited. Innovative human preclinical models may help to uncover relevant disease mechanisms and enhance the translation of therapeutic findings to strengthen neuromuscular disease precision medicine. By concentrating on idiopathic inflammatory muscle disorders, we summarize the recent evolution of the novel in vitro models to study disease mechanisms and therapeutic strategies. A particular focus is laid on the integration and simulation of multicellular interactions of muscle tissue in disease phenotypes in vitro. Finally, the requirements of a neuromuscular disease drug development workflow are discussed with a particular emphasis on cell sources, co-culture systems (including organoids), functionality, and throughput.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Matthias Schmidt
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Susann Kummer
- Risk Group 4 Pathogens–Stability and Persistence, Biosafety Level-4 Laboratory, Center for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| |
Collapse
|
15
|
Zeng R, Glaubitz S, Schmidt J. Antibody Therapies in Autoimmune Inflammatory Myopathies: Promising Treatment Options. Neurotherapeutics 2022; 19:911-921. [PMID: 35394612 PMCID: PMC9294121 DOI: 10.1007/s13311-022-01220-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2022] [Indexed: 12/29/2022] Open
Abstract
Inflammatory myopathies, including polymyositis (PM), dermatomyositis (DM), inclusion body myositis (IBM), necrotizing myopathy (NM), antisynthetase syndrome (ASS) and overlap myositis (OM), in short myositis, are rare diseases. All forms of myositis have progressive muscle weakness in common, with each subtype characterized by different autoantibody profiles, histological findings and extramuscular manifestations. Due to better understanding of the pathogenesis of the muscle inflammation in myositis, new molecular pathways for targeted therapy have been discovered. Current therapies aim at different components of the innate or the adaptive immune response. Additionally, non-inflammatory mechanisms in myositis have come into focus as possible treatment targets. The use of therapeutical antibodies in myositis has been examined in various clinical studies, several of them randomized controlled ones: Depletion of B-cells by rituximab has been established as treatment of refractory myositis. IVIG, an antibody therapy in the wider sense, has now been licensed for DM following a recent positive clinical trial. Negative study results were reported in randomized trials with infliximab, sifalimumab and bimagrumab. Studies on basiliximab and eculizumab are currently underway, and are expected to yield results in a couple of years. Despite some promising results of clinical studies with antibody therapy in myositis, further research is crucial to optimize the treatment for this debilitating disease and to find treatment alternatives for treatment-refractory patients.
Collapse
Affiliation(s)
- Rachel Zeng
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefanie Glaubitz
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Department of Neurology and Pain Treatment, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany.
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany.
| |
Collapse
|
16
|
Dalakas MC. Complement in autoimmune inflammatory myopathies, the role of myositis-associated antibodies, COVID-19 associations, and muscle amyloid deposits. Expert Rev Clin Immunol 2022; 18:413-423. [PMID: 35323101 DOI: 10.1080/1744666x.2022.2054803] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The inflammatory myopathies (IM) have now evolved into distinct subsets requiring clarification about their immunopathogenesis to guide applications of targeted therapies. AREAS COVERED Immunohistopathologic criteria of IM with a focus on complement, anti-complement therapeutics, and other biologic immunotherapies. The COVID19-triggered muscle autoimmunity along with the correct interpretation of muscle amyloid deposits is discussed. EXPERT OPINION The IM, unjustifiably referred as idiopathic, comprise Dermatomyositis (DM), Necrotizing Autoimmune Myositis (NAM), Anti-synthetase syndrome-overlap myositis (Anti-SS-OM), and Inclusion-Body-Myositis (IBM). In DM, complement activation with MAC-mediated endomysial microvascular destruction and perifascicular atrophy is the fundamental process, while innate immunity activation factors, INF1 and MxA, sense and secondarily enhance inflammation. Complement participates in muscle fiber necrosis from any cause and may facilitate muscle-fiber necrosis in NAM but seems unlikely that myositis-associated antibodies participate in complement-fixing. Accordingly, anti-complement therapeutics should be prioritized for DM. SARS-CoV-2 can potentially trigger muscle autoimmunity, but systematic studies are needed as the reported autopsy findings are not clinically relevant. In IBM, tiny amyloid deposits within muscle fibers are enhanced by inflammatory mediators contributing to myodegeneration; in contrast, spotty amyloid deposits in the endomysial connective tissue do not represent 'amyloid myopathy' but only have diagnostic value for amyloidosis due to any cause.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,University of Athens Medical School, Neuroimmunology Unit, National and Kapodistrian University, Athens, Greece
| |
Collapse
|
17
|
Gutschmidt K, Schoser B. Erworbene Myopathien und ihre neuen Therapien. DIE NEUROLOGIE & PSYCHIATRIE 2022; 23. [PMCID: PMC9713102 DOI: 10.1007/s15202-022-5546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
- Kristina Gutschmidt
- Friedrich-Baur-Institut, Neurologische Klinik, Ludwig-Maximilians-Universität München, Ziemssenstr. 1a, 80336 München, Germany
| | - Benedikt Schoser
- Klinikum der Universität München, Friedrich Baur-Institut / Neurologische Klinik und Poliklinik, Ziemssenstraße 1a, 80336 München, Germany
| |
Collapse
|
18
|
Treatment and Management of Autoimmune Myopathies. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00022-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Shi J, Tang M, Zhou S, Xu D, Zhao J, Wu C, Wang Q, Tian X, Li M, Zeng X. Programmed Cell Death Pathways in the Pathogenesis of Idiopathic Inflammatory Myopathies. Front Immunol 2021; 12:783616. [PMID: 34899749 PMCID: PMC8651702 DOI: 10.3389/fimmu.2021.783616] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
Idiopathic inflammatory myopathy (IIM) is a heterogeneous group of acquired, autoimmune muscle diseases characterized by muscle inflammation and extramuscular involvements. Present literatures have revealed that dysregulated cell death in combination with impaired elimination of dead cells contribute to the release of autoantigens, damage-associated molecular patterns (DAMPs) and inflammatory cytokines, and result in immune responses and tissue damages in autoimmune diseases, including IIMs. This review summarizes the roles of various forms of programmed cell death pathways in the pathogenesis of IIMs and provides evidence for potential therapeutic targets.
Collapse
Affiliation(s)
- Jia Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Mingwei Tang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Shuang Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Dong Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Chanyuan Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology & Clinical Immunology, Ministry of Education, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| |
Collapse
|
20
|
Piazzi M, Bavelloni A, Cenni V, Faenza I, Blalock WL. Revisiting the Role of GSK3, A Modulator of Innate Immunity, in Idiopathic Inclusion Body Myositis. Cells 2021; 10:cells10113255. [PMID: 34831477 PMCID: PMC8625526 DOI: 10.3390/cells10113255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Idiopathic or sporadic inclusion body myositis (IBM) is the leading age-related (onset >50 years of age) autoimmune muscular pathology, resulting in significant debilitation in affected individuals. Once viewed as primarily a degenerative disorder, it is now evident that much like several other neuro-muscular degenerative disorders, IBM has a major autoinflammatory component resulting in chronic inflammation-induced muscle destruction. Thus, IBM is now considered primarily an inflammatory pathology. To date, there is no effective treatment for sporadic inclusion body myositis, and little is understood about the pathology at the molecular level, which would offer the best hopes of at least slowing down the degenerative process. Among the previously examined potential molecular players in IBM is glycogen synthase kinase (GSK)-3, whose role in promoting TAU phosphorylation and inclusion bodies in Alzheimer’s disease is well known. This review looks to re-examine the role of GSK3 in IBM, not strictly as a promoter of TAU and Abeta inclusions, but as a novel player in the innate immune system, discussing some of the recent roles discovered for this well-studied kinase in inflammatory-mediated pathology.
Collapse
Affiliation(s)
- Manuela Piazzi
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alberto Bavelloni
- Laboratorio di Oncologia Sperimentale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Vittoria Cenni
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Irene Faenza
- Dipartimento di Scienze Biomediche and Neuromotorie, Università di Bologna, 40136 Bologna, Italy;
| | - William L. Blalock
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence:
| |
Collapse
|
21
|
Zeng R, Glaubitz S, Schmidt J. Inflammatory myopathies: shedding light on promising agents and combination therapies in clinical trials. Expert Opin Investig Drugs 2021; 30:1125-1140. [PMID: 34779311 DOI: 10.1080/13543784.2021.2003776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Due to new insights into the pathogenesis of inflammatory myopathies - in short myositis - and the urgent need for new treatment options in patients who are refractory to standard therapy, multiple novel drugs have been developed and studied in clinical trials. In light of this exciting development, a critical evaluation of the present data is necessary in order to identify the best pathway to future treatment of inflammatory myopathies. AREAS COVERED This review focuses on the current evidence from clinical trials in myositis and encompasses dermatomyositis, polymyositis, necrotizing myopathy, antisynthetase-syndrome, overlap myositis, and inclusion body myositis. The results of studies on new therapeutic agents are summarized, in particular larger cohort studies and randomized trials from recent years. When such data were not available, earlier and smaller representative studies were included instead. EXPERT OPINION Current studies in most myositis subtypes have shown positive effects of novel biologicals such as abatacept, sifalimumab, JAK-Inhibitors as well as known agents such as rituximab, but further studies are needed to confirm these observations. In inclusion body myositis, the eagerly awaited recent therapeutic trials have missed their primary endpoints, except for the phase 2 study with rapamycin, which has demonstrated significant improvements in secondary endpoints. Future trials will also need to focus on combination therapies of multiple immunomodulatory agents.
Collapse
Affiliation(s)
- Rachel Zeng
- Muscle Immunobiology Group, Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - Stefanie Glaubitz
- Muscle Immunobiology Group, Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Muscle Immunobiology Group, Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany.,Department of Neurology and Pain Treatment, University Hospital of the Medical School Brandenburg, Immanuel Klinik Rüdersdorf, Rüdersdorf bei Berlin, Germany.,Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
| |
Collapse
|
22
|
Glaubitz S, Zeng R, Rakocevic G, Schmidt J. Update on Myositis Therapy: from Today's Standards to Tomorrow's Possibilities. Curr Pharm Des 2021; 28:863-880. [PMID: 34781868 DOI: 10.2174/1381612827666211115165353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022]
Abstract
Inflammatory myopathies, in short, myositis, are heterogeneous disorders that are characterized by inflammation of skeletal muscle and weakness of arms and legs. Research over the past few years has led to a new understanding regarding the pathogenesis of myositis. The new insights include different pathways of the innate and adaptive immune response during the pathogenesis of myositis. The importance of non-inflammatory mechanisms such as cell stress and impaired autophagy has been recently described. New target-specific drugs for myositis have been developed and are currently being tested in clinical trials. In this review, we discuss the mechanisms of action of pharmacological standards in myositis and provide an outlook of future treatment approaches.
Collapse
Affiliation(s)
- Stefanie Glaubitz
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen. Germany
| | - Rachel Zeng
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen. Germany
| | - Goran Rakocevic
- Department of Neurology, Neuromuscular Division, University of Virginia, Charlottesville. United States
| | - Jens Schmidt
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen. Germany
| |
Collapse
|
23
|
Winkler M, von Landenberg C, Kappes-Horn K, Neudecker S, Kornblum C, Reimann J. Diagnosis and Clinical Development of Sporadic Inclusion Body Myositis and Polymyositis With Mitochondrial Pathology: A Single-Center Retrospective Analysis. J Neuropathol Exp Neurol 2021; 80:1060–1067. [PMID: 34643702 DOI: 10.1093/jnen/nlab101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To review our diagnostic and treatment approaches concerning sporadic inclusion body myositis (sIBM) and polymyositis with mitochondrial pathology (PM-Mito), we conducted a retrospective analysis of clinical and histological data of 32 patients diagnosed as sIBM and 7 patients diagnosed as PM-Mito by muscle biopsy. Of 32 patients identified histologically as sIBM, 19 fulfilled the 2011 European Neuromuscular Center (ENMC) diagnostic criteria for "clinico-pathologically defined sIBM" at the time of biopsy. Among these, 2 patients developed sIBM after years of immunosuppressive treatment for organ transplantation. Of 11 patients fulfilling the histological but not the clinical criteria, including 3 cases with duration <12 months, 8 later fulfilled the criteria for clinico-pathologically defined sIBM. Of 7 PM-Mito patients, 4 received immunosuppression with clinical improvement in 3. One of these later developed clinico-pathologically defined sIBM; 1 untreated patient progressed to clinically defined sIBM. Thus, muscle histology remains important for this differential diagnosis to identify sIBM patients not matching the ENMC criteria and the PM-Mito group. In the latter, we report at least 50% positive, if occasionally transient, response to immunosuppressive treatments and progression to sIBM in a minority. The mitochondrial abnormalities defining PM-Mito do not seem to define the threshold to immunosuppression unresponsiveness.
Collapse
Affiliation(s)
- Maren Winkler
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| | - Christina von Landenberg
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| | - Karin Kappes-Horn
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| | - Stephan Neudecker
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| | - Cornelia Kornblum
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| | - Jens Reimann
- From the Section of Neuromuscular Diseases, Department of Neurology, University Hospital of Bonn, Bonn, Germany (MW, CvL, KK-H, CK, JR); Group Practice for Neurology, Bonn, Germany (SN); Center for Rare Diseases, University Hospital of Bonn, Bonn, Germany (CK)
| |
Collapse
|
24
|
Pinto MV, Laughlin RS, Klein CJ, Mandrekar J, Naddaf E. Inclusion body myositis: correlation of clinical outcomes with histopathology, electromyography and laboratory findings. Rheumatology (Oxford) 2021; 61:2504-2511. [PMID: 34617994 DOI: 10.1093/rheumatology/keab754] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To determine whether histopathological, electromyographic and laboratory markers correlate with clinical measures in Inclusion Body Myositis (IBM). METHODS We reviewed our electronic medical records to identify patients with IBM according to ENMC 2011 criteria, seen between 2015 and 2020. We only included patients who had a muscle biopsy and needle electromyography (EMG) performed on the same muscle (opposite or same side). We used a detailed grading system (0- normal to 4- severe) to score histopathological and EMG findings. Clinical severity was assessed by the modified Rankin scale (mRS), muscle strength sum score (SSS), quadriceps strength and severity of dysphagia on swallow evaluation. Serum markers of interest were creatine kinase level, and cN-1A antibodies. RESULTS We included 50 IBM patients, with a median age of 69 years; 64% were males. Median disease duration at diagnosis was 51 months. On muscle biopsy, endomysial inflammation mainly correlated with dysphagia, and inversely correlated with mRS. Vacuoles and congophilic inclusions did not correlate with any of the clinical measures. On EMG, the shortness of motor unit potential (MUP) duration correlated with all clinical measures. Myotonic discharges, and not fibrillation potentials, correlated with the severity of inflammation. Serum markers did not have a statistically-significant correlation with any of the clinical measures. CONCLUSIONS Dysphagia was the main clinical feature of IBM correlating with endomysial inflammation. Otherwise, inclusion body myositis clinical measures had limited correlation with histopathological features in this study. The shortness of MUP duration correlated with all clinical measures.
Collapse
Affiliation(s)
- Marcus V Pinto
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Jay Mandrekar
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Elie Naddaf
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Unger L. Therapieoptionen und outcome bei idiopathischen entzündlichen Muskelerkrankungen. AKTUEL RHEUMATOL 2021. [DOI: 10.1055/a-1423-7579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
ZusammenfassungDie idiopathischen entzündlichen Muskelerkrankungen (IIM) sind eine sehr heterogene Gruppe, die sich immer besser differenzieren lässt. Damit eröffnen sich mehr Möglichkeiten für gezieltere Therapien, die zum einen auf die Veränderung pathogenetischer Faktoren gerichtet sind. Zum anderen sollen sie Krankheitsaktivität vermindern, Muskelaufbau fördern, Organschäden verhindern und Lebensqualität verbessern.Die folgende Übersichtsarbeit fasst die vorhandenen Daten zu bereits angewandten Behandlungen in der Praxis zusammen und gibt einen Ausblick auf zukünftige Alternativen.Für die Polymyositis (PM) und Dermatomyositis (DM) sind Glukokortikoide weiter unverzichtbarer Therapiebestandteil. Eine frühe Kombination mit konventionellen DMARDs hat sich durchgesetzt. Die ProDerm- Studie stellt für die Immunglobulintherapie in der Praxis eine gute Basis dar. Rituximab (RTX) löst Cyclophosphamid (CYC) bei schweren Verlaufsformen immer mehr ab.Für Abatacept, Jak-Kinase-Hemmer, Apremilast, Sifalimumab und Lenabasum müssen vielversprechende erste Ergebnisse durch weiter Studien untermauert werden. Anspruchsvoll ist die Behandlung bei extramuskulärer Beteiligung. Von großem Interesse ist der zukünftige Stellenwert von Nintedanib bei der interstitiellen Lungenerkrankung im Rahmen einer Myositis (IIM-ILD).Die Therapie der Einschlusskörperchen- Myositis (IBM) ist immer noch eine große Herausforderung. Zahlreiche Studien haben bisher nicht überzeugend zu einer Besserung der Prognose führen können. Spätestens bei therapierefraktärem Verlauf sollte an eine Malignom- assoziierte Myositis gedacht werden. Gelegentlich verbirgt sich auch eine hereditäre Myopathie hinter einer, zum Beispiel durch einen Infekt oder Überlastung getriggerten, Myositis.Komplikationen im Verlauf, wie Dysphagie, Infektionen, Myokardbeteiligung stellen keine Seltenheit dar. Häufig besteht Multimorbidität. Eine interdisziplinäre Zusammenarbeit in einem kompetenten Team, in dem erfahrene Physio-, Ergo- und Psychotherapeuten fester Bestandteil sind, ist unabdingbar für eine erfolgreiche Begleitung dieser Patienten.
Collapse
Affiliation(s)
- Leonore Unger
- Städtisches Klinikum Dresden, I. Medizinische Klinik, Dresden, Deutschland
| |
Collapse
|
26
|
Abstract
Autoimmune neurogenic dysphagia refers to manifestation of dysphagia due to autoimmune diseases affecting muscle, neuromuscular junction, nerves, roots, brainstem, or cortex. Dysphagia is either part of the evolving clinical symptomatology of an underlying neurological autoimmunity or occurs as a sole manifestation, acutely or insidiously. This opinion article reviews the autoimmune neurological causes of dysphagia, highlights clinical clues and laboratory testing that facilitate early diagnosis, especially when dysphagia is the presenting symptom, and outlines the most effective immunotherapeutic approaches. Dysphagia is common in inflammatory myopathies, most prominently in inclusion body myositis, and is frequent in myasthenia gravis, occurring early in bulbar-onset disease or during the course of progressive, generalized disease. Acute-onset dysphagia is often seen in Guillain–Barre syndrome variants and slowly progressive dysphagia in paraneoplastic neuropathies highlighted by the presence of specific autoantibodies. The most common causes of CNS autoimmune dysphagia are demyelinating and inflammatory lesions in the brainstem, occurring in patients with multiple sclerosis and neuromyelitis optica spectrum disorders. Less common, but often overlooked, is dysphagia in stiff-person syndrome especially in conjunction with cerebellar ataxia and high anti-GAD autoantibodies, and in gastrointestinal dysmotility syndromes associated with autoantibodies against the ganglionic acetyl-choline receptor. In the setting of many neurological autoimmunities, acute-onset or progressive dysphagia is a potentially treatable condition, requiring increased awareness for prompt diagnosis and early immunotherapy initiation.
Collapse
|
27
|
Clinical Trials in Myositis: Where Do We Stand? CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021. [DOI: 10.1007/s40674-021-00180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Abstract
PURPOSE OF REVIEW To review the pathogenesis of inclusion body myositis (IBM). RECENT FINDINGS IBM is an autoimmune disease. Multiple arms of the immune system are activated, but a direct attack on muscle fibers by highly differentiated T cells drives muscle destruction. SUMMARY Further understanding of the pathogenesis of IBM guides rational approaches to developing therapeutic strategies.
Collapse
|
29
|
Shelly S, Mielke MM, Mandrekar J, Milone M, Ernste FC, Naddaf E, Liewluck T. Epidemiology and Natural History of Inclusion Body Myositis: A 40-Year Population-Based Study. Neurology 2021; 96:e2653-e2661. [PMID: 33879596 DOI: 10.1212/wnl.0000000000012004] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/24/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES To determine the prevalence and natural history of sporadic inclusion body myositis (sIBM) and to test the hypothesis that patients with sIBM have higher cancer or mortality rates than the general population. METHODS We sought patients with sIBM defined by the 2011 European Neuromuscular Centre (ENMC) diagnostic criteria among Olmsted County, Minnesota, residents in 40-year time period. RESULTS We identified 20 patients (10 clinicopathologically defined, 9 clinically defined, and 1 probable) according to the ENMC criteria and 1 patient with all features of clinicopathologically defined sIBM except for symptom onset at <45 years of age. The prevalence of sIBM in 2010 was 18.20 per 100,000 people ≥50 years old. Ten patients developed cancers. The incidence of cancers in sIBM did not differ from that observed in the general population (odds ratio 1.89, 95% confidence interval [CI] 0.639-5.613, p = 0.24). Two-thirds of patients developed dysphagia, and half required a feeding tube. Nine patients required a wheelchair. The median time from symptom onset to wheelchair dependence was 10.5 (range 1-29) years. Overall life expectancy was shorter in the sIBM group compared to the general population (84.1 [95% CI 78-88.4] vs 87.5 [95% CI 85.2-89.5] years, p = 0.03). Thirteen patients died; 9 deaths were sIBM related (7 respiratory and 2 unspecified sIBM complications). Female sex (p = 0.03) and dysphagia (p = 0.05) were independent predictors of death. CONCLUSION Olmsted County has the highest prevalence of sIBM reported to date. Patients with sIBM have similar risk of cancer, but slightly shorter life expectancy compared to matched patients without sIBM. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that patients with sIBM have similar risks of cancers and slightly shorter life expectancy compared to controls.
Collapse
Affiliation(s)
- Shahar Shelly
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN
| | - Michelle M Mielke
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN
| | - Jay Mandrekar
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN
| | - Margherita Milone
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN
| | - Floranne C Ernste
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN
| | - Elie Naddaf
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN.
| | - Teerin Liewluck
- From the Department of Neurology (S.S., M.M.M., J.M., M.M., E.N., T.L.), Department of Health Sciences Research (M.M.M., J.M.), and Division of Rheumatology (F.C.E.), Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
30
|
Patwardhan A, Spencer CH. Biologics in refractory idiopathic inflammatory myositis (IIM): What experience in juvenile vs adult myositis tells us about the use of biologics in pediatric IIM. Mod Rheumatol 2021; 31:933-948. [PMID: 33499694 DOI: 10.1080/14397595.2021.1881027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Juvenile dermatomyositis (JDM) is an extremely heterogeneous orphan disease with limited amount of dedicated research on the subject matter. Recent research suggests that JDM may not just be the classic antibody driven complements mediated microangiopathy as was thought to be in the past. The etiopathogenesis of JDM also involves inappropriate stimulation of innate immune system followed by dysregulation of the adaptive immune response through dendritic cells. Many variable immune factors such as genetics, major histocompatibility complex expressions, immunohistochemical variabilities, and diversity in specific and associated autoantibodies may make individual IIM and JDM cases unique. The diversity in IIM and JDM also explains individual variability in response to specific therapies. Classifying and matching the right patients to the right treatment is crucial to the successful treatment of these patients with better outcomes. Sub-type specific biologic therapy may be the best current treatment that can match the patient to the best treatment options. A PubMed search was performed to find all the available cases of refractory myositis patients treated with biologics up to July 2020. Using this search this article reviews all the current biologic treatment options and experiences for both adults and children in the context of recent basic science to assist pediatric rheumatologists in choosing the optimal biologic therapy for a child with recalcitrant JDM.
Collapse
Affiliation(s)
| | - Charles H Spencer
- University of Mississippi Medical Center, Batson Children's Hospital, Jackson, MS, USA
| |
Collapse
|
31
|
In Pursuit of an Effective Treatment: the Past, Present and Future of Clinical Trials in Inclusion Body Myositis. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021. [DOI: 10.1007/s40674-020-00169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Abstract
Purpose of review
No clinical trial in sporadic inclusion body myositis (IBM) thus far has shown a clear and sustained therapeutic effect. We review previous trial methodology, explore why results have not translated into clinical practice, and suggest improvements for future IBM trials.
Recent findings
Early trials primarily assessed immunosuppressive medications, with no significant clinical responses observed. Many of these studies had methodological issues, including small participant numbers, nonspecific diagnostic criteria, short treatment and/or assessment periods and insensitive outcome measures. Most recent IBM trials have instead focused on nonimmunosuppressive therapies, but there is mounting evidence supporting a primary autoimmune aetiology, including the discovery of immunosuppression-resistant clones of cytotoxic T cells and anti-CN-1A autoantibodies which could potentially be used to stratify patients into different cohorts. The latest trials have had mixed results. For example, bimagrumab, a myostatin blocker, did not affect the 6-min timed walk distance, whereas sirolimus, a promotor of autophagy, did. Larger studies are planned to evaluate the efficacy of sirolimus and arimoclomol.
Summary
Thus far, no treatment for IBM has demonstrated a definite therapeutic effect, and effective treatment options in clinical practice are lacking. Trial design and ineffective therapies are likely to have contributed to these failures. Identification of potential therapeutic targets should be followed by future studies using a stratified approach and sensitive and relevant outcome measures.
Collapse
|
32
|
Gklinos P, Papadopoulou M, Stanulovic V, Mitsikostas DD, Papadopoulos D. Monoclonal Antibodies as Neurological Therapeutics. Pharmaceuticals (Basel) 2021; 14:ph14020092. [PMID: 33530460 PMCID: PMC7912592 DOI: 10.3390/ph14020092] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
Over the last 30 years the role of monoclonal antibodies in therapeutics has increased enormously, revolutionizing treatment in most medical specialties, including neurology. Monoclonal antibodies are key therapeutic agents for several neurological conditions with diverse pathophysiological mechanisms, including multiple sclerosis, migraines and neuromuscular disease. In addition, a great number of monoclonal antibodies against several targets are being investigated for many more neurological diseases, which reflects our advances in understanding the pathogenesis of these diseases. Untangling the molecular mechanisms of disease allows monoclonal antibodies to block disease pathways accurately and efficiently with exceptional target specificity, minimizing non-specific effects. On the other hand, accumulating experience shows that monoclonal antibodies may carry class-specific and target-associated risks. This article provides an overview of different types of monoclonal antibodies and their characteristics and reviews monoclonal antibodies currently in use or under development for neurological disease.
Collapse
Affiliation(s)
- Panagiotis Gklinos
- Department of Neurology, KAT General Hospital of Attica, 14561 Athens, Greece;
| | - Miranta Papadopoulou
- Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Vid Stanulovic
- Global Pharmacovigilance, R&D Sanofi, 91385 Chilly-Mazarin, France;
| | - Dimos D. Mitsikostas
- 1st Neurology Department, Aeginition Hospital, National and Kapodistrian University of Athens, 11521 Athens, Greece;
| | - Dimitrios Papadopoulos
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 129 Vasilissis Sophias Avenue, 11521 Athens, Greece
- Salpetriere Neuropsychiatric Clinic, 149 Papandreou Street, Metamorphosi, 14452 Athens, Greece
- Correspondence:
| |
Collapse
|
33
|
Dalakas MC. Inflammatory myopathies: update on diagnosis, pathogenesis and therapies, and COVID-19-related implications. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2020; 39:289-301. [PMID: 33458584 PMCID: PMC7783437 DOI: 10.36185/2532-1900-032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
The inflammatory myopathies constitute a heterogeneous group of acquired myopathies that have in common the presence of endomysial inflammation. Based on steadily evolved clinical, histological and immunopathological features and some autoantibody associations, these disorders can now be classified in five characteristic subsets: Dermatomyositis (DM) Polymyositis (PM), Necrotizing Autoimmune Myositis (NAM), Anti-synthetase syndrome-overlap myositis (Anti-SS-OM), and Inclusion-Body-Myositis (IBM). Each inflammatory myopathy subset has distinct immunopathogenesis, prognosis and response to immunotherapies, necessitating the need to correctly identify each subtype from the outset to avoid disease mimics and proceed to early therapy initiation. The review presents the main clinicopathologic characteristics of each subset highlighting the importance of combining expertise in clinical neurological examination with muscle morphology and immunopathology to avoid erroneous diagnoses and therapeutic schemes. The main autoimmune markers related to autoreactive T cells, B cells, autoantibodies and cytokines are presented and the concomitant myodegenerative features seen in IBM muscles are pointed out. Most importantly, unsettled issues related to a role of autoantibodies and controversies with reference to possible triggering factors related to statins are clarified. The emerging effect SARS-CoV-2 as the cause of hyperCKemia and potentially NAM is addressed and practical guidelines on the best therapeutic approaches and concerns regarding immunotherapies during COVID-19 pandemic are summarized.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA USA and the Neuroimmunology Unit, National and Kapodistrian University University of Athens Medical School, Athens, Greece
| |
Collapse
|
34
|
Biliciler S, Kwan J. Inflammatory Myopathies: Utility of Antibody Testing. Neurol Clin 2020; 38:661-678. [PMID: 32703475 DOI: 10.1016/j.ncl.2020.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Inflammatory myopathies are a group of immune-mediated muscle disorders comprising dermatomyositis; polymyositis; overlap myositis, including antisynthetase syndromes and nonspecific myositis, immune-mediated necrotizing myopathies, and sporadic inclusion body myositis. They are now much more eloquently classified both pathologically and clinically because of the discovery of several myositis-specific and myositis-associated antibodies. These antibodies also aid in choosing the best treatment options in each case. Based on the initial classifications of inflammatory myopathies, inclusion body myositis, overlap myositis, and necrotizing myositis were all included in the polymyositis group. This article discusses cases, diagnostic tools, associated antibodies, and pathology.
Collapse
Affiliation(s)
- Suur Biliciler
- Department of Neurology, UT Health Science Center in Houston, McGovern Medical School, 6341 Fannin Street, MSC #466, Houston, TX 77030, USA.
| | - Justin Kwan
- Department of Neurology, Temple University, Lewis Katz School of Medicine, 3401 North Broad Street Street, Suite C525, Philadelphia, PA 19410, USA
| |
Collapse
|
35
|
Khadilkar SV, Dhamne MC. What is New in Idiopathic Inflammatory Myopathies: Mechanisms and Therapies. Ann Indian Acad Neurol 2020; 23:458-467. [PMID: 33223661 PMCID: PMC7657284 DOI: 10.4103/aian.aian_400_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 11/18/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of disorders that cause muscle weakness and also have extramuscular manifestations involving various organ systems; namely the lung, skin, heart, and joints. Previously classified broadly as dermatomyositis (DM) and polymyositis now the spectrum of the disease has evolved into more clinical subtypes. There are now five clinicoserological subtypes recognized worldwide DM, antisynthetase syndrome (AS), overlap myositis (OM), immune mediated necrotizing myopathy (IMNM), and inclusion body myositis. Each of these subtypes has a unique phenotype and specific antibodies associated. With the evolving treatment options from the use of immunosuppressive medications to the use of targeted therapy with biologic agents, and further understanding of the pathogenesis of inflammatory myositis, we may have more effective treatment options. We discuss in this review, various myositis-associated antibodies associated with each clinicoserological subtype of IIM and their role. We also describe the evolving therapies and the evidence for the newer biologic therapies in the treatment of IIMs.
Collapse
Affiliation(s)
- Satish V Khadilkar
- Department of Neurology, Bombay Hospital Institute of Medical Sciences, Mumbai, Maharashtra, India
| | - Megha C Dhamne
- Department of Neurology, Dr. L H Hiranandani Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
36
|
Oldroyd AGS, Lilleker JB, Williams J, Chinoy H, Miller JAL. Long‐term strength and functional status in inclusion body myositis and identification of trajectory subgroups. Muscle Nerve 2020; 62:76-82. [PMID: 32134516 PMCID: PMC8629114 DOI: 10.1002/mus.26859] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/26/2020] [Accepted: 03/01/2020] [Indexed: 11/08/2022]
Abstract
Introduction Methods Results Discussion See editorial on pages 7–9 in this issue.
Collapse
Affiliation(s)
- Alexander G. S. Oldroyd
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre Manchester United Kingdom
- Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Science Centre Manchester United Kingdom
- Centre for Epidemiology Versus Arthritis University of Manchester Manchester United Kingdom
- Department of Rheumatology Salford Royal NHS Foundation Trust Salford United Kingdom
| | - James B. Lilleker
- Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Science Centre Manchester United Kingdom
- Manchester Centre for Clinical Neurosciences Salford Royal NHS Foundation Trust, Manchester Academic Health Sciences Centre United Kingdom
| | - Jacob Williams
- Manchester Medical School University of Manchester Manchester United Kingdom
| | - Hector Chinoy
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre Manchester United Kingdom
- Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Science Centre Manchester United Kingdom
- Department of Rheumatology Salford Royal NHS Foundation Trust Salford United Kingdom
- Manchester Centre for Clinical Neurosciences Salford Royal NHS Foundation Trust, Manchester Academic Health Sciences Centre United Kingdom
| | - James A. L. Miller
- Department of Neurology Royal Victoria Hospitals, The Newcastle upon Tyne Hospitals NHS Foundation Trust Queen Victoria Road, Newcastle United Kingdom
| |
Collapse
|
37
|
Greenberg SA. Inclusion body myositis: clinical features and pathogenesis. Nat Rev Rheumatol 2020; 15:257-272. [PMID: 30837708 DOI: 10.1038/s41584-019-0186-x] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Inclusion body myositis (IBM) is often viewed as an enigmatic disease with uncertain pathogenic mechanisms and confusion around diagnosis, classification and prospects for treatment. Its clinical features (finger flexor and quadriceps weakness) and pathological features (invasion of myofibres by cytotoxic T cells) are unique among muscle diseases. Although IBM T cell autoimmunity has long been recognized, enormous attention has been focused for decades on several biomarkers of myofibre protein aggregates, which are present in <1% of myofibres in patients with IBM. This focus has given rise, together with the relative treatment refractoriness of IBM, to a competing view that IBM is not an autoimmune disease. Findings from the past decade that implicate autoimmunity in IBM include the identification of a circulating autoantibody (anti-cN1A); the absence of any statistically significant genetic risk factor other than the common autoimmune disease 8.1 MHC haplotype in whole-genome sequencing studies; the presence of a marked cytotoxic T cell signature in gene expression studies; and the identification in muscle and blood of large populations of clonal highly differentiated cytotoxic CD8+ T cells that are resistant to many immunotherapies. Mounting evidence that IBM is an autoimmune T cell-mediated disease provides hope that future therapies directed towards depleting these cells could be effective.
Collapse
Affiliation(s)
- Steven A Greenberg
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA. .,Children's Hospital Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Allenbach Y, Benveniste O. Miopatie infiammatorie. Neurologia 2020. [DOI: 10.1016/s1634-7072(20)43301-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
39
|
Glaubitz S, Zeng R, Schmidt J. New insights into the treatment of myositis. Ther Adv Musculoskelet Dis 2020; 12:1759720X19886494. [PMID: 31949477 PMCID: PMC6950531 DOI: 10.1177/1759720x19886494] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022] Open
Abstract
The myositis syndromes include polymyositis, dermatomyositis (DM), necrotizing myopathy, inclusion body myositis (IBM), antisynthetase syndrome and overlap syndromes with myositis. These syndromes mostly occur in middle-aged patients, while juvenile DM occurs in children and adolescents. Patients mostly show a subacute weakness and myalgia in the upper and lower limbs, the diagnosis is based upon these clinical findings in combination with muscle biopsy results and specific serum autoantibodies. In recent years, research achieved a better understanding about the molecular mechanism underlying the myositis syndromes, as well as disease progress and extramuscular organ manifestations, such as interstitial lung disease and association with neoplasias. Treatment mainly consists of glucocorticosteroids and immunosuppressants. IBM is usually refractory to treatments. This review provides an overview of the current standards of treatment and new treatment options like monoclonal antibodies and new molecular therapies and their first results from clinical trials.
Collapse
Affiliation(s)
- Stefanie Glaubitz
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - Rachel Zeng
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Robert-Koch-Sr. 40, 37075 Göttingen, Germany
| |
Collapse
|
40
|
Khoo T, Limaye V. Biologic therapy in the idiopathic inflammatory myopathies. Rheumatol Int 2019; 40:191-205. [PMID: 31680207 DOI: 10.1007/s00296-019-04467-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/24/2019] [Indexed: 11/29/2022]
Abstract
The idiopathic inflammatory myopathies (IIM) are a group of autoimmune diseases resulting from inflammation of muscle and manifesting as weakness, though a range of extra-muscular manifestations are observed. These are often correlated closely with disease subtype and the presence of myositis-specific/myositis-associated antibodies. IIM are notoriously difficult to treat and often refractory to glucocorticoid therapy and synthetic immunosuppressants. Both the innate and adaptive immune systems are implicated in the pathogenesis of IIM. A growing understanding of the key cytokines as well as the cell-mediated and antibody effectors of disease has identified multiple potential targets for biologic therapy. The most widely used of these is B-cell depletion via rituximab though the tumour necrosis factor inhibitors and other biologic therapies used in diseases such as rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis have also been trialled. This review summarises the literature thus far on biologic therapy in IIM, highlighting both the significant trials that influence current treatment regimens and also the continuing need for further research to inform more effective therapies.
Collapse
Affiliation(s)
- Thomas Khoo
- Central Adelaide Local Health Network, Adelaide, Australia
| | - Vidya Limaye
- Rheumatology Unit, Royal Adelaide Hospital, Adelaide, Australia. .,Discipline of Medicine, School of Medicine, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
41
|
Greenberg SA, Pinkus JL, Kong SW, Baecher-Allan C, Amato AA, Dorfman DM. Highly differentiated cytotoxic T cells in inclusion body myositis. Brain 2019; 142:2590-2604. [DOI: 10.1093/brain/awz207] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/21/2019] [Accepted: 05/16/2019] [Indexed: 01/12/2023] Open
Abstract
Abstract
Inclusion body myositis is a late onset treatment-refractory autoimmune disease of skeletal muscle associated with a blood autoantibody (anti-cN1A), an HLA autoimmune haplotype, and muscle pathology characterized by cytotoxic CD8+ T cell destruction of myofibres. Here, we report on translational studies of inclusion body myositis patient muscle compared with a diverse set of other muscle disease samples. Using available microarray data on 411 muscle samples from patients with inclusion body myositis (n = 40), other muscle diseases (n = 265), and without neuromuscular disease (normal, n = 106), we identified a signature of T-cell cytotoxicity in inclusion body myositis muscle coupled with a signature of highly differentiated CD8 T-cell effector memory and terminally differentiated effector cells. Further, we examined killer cell lectin-like receptor G1 (KLRG1) as a marker of this population of cells, demonstrated the correlation of KLRG1 gene expression with lymphocyte cytotoxicity across 28 870 human tissue samples, and identified the presence of KLRG1 on pathogenic inclusion body myositis muscle invading T cells and an increase in KLRG1 expressing T cells in inclusion body myositis blood. We examined inclusion body myositis muscle T-cell proliferation by Ki67 immunohistochemistry demonstrating that diseased muscle-invading T cells are minimally or non-proliferative, in accordance with known properties of highly differentiated or terminally differentiated T cells. We found low expression of KLRG1 on infection-protective human lymphoid tissue central memory T cells and autoimmune-protective human blood regulatory T cells. Targeting highly differentiated cytotoxic T cells could be a favourable approach to treatment of inclusion body myositis.
Collapse
Affiliation(s)
- Steven A Greenberg
- Brigham and Women’s Hospital Department of Neurology, Division of Neuromuscular Disease, and Harvard Medical School, Boston, MA, USA
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA and Harvard Medical School, Boston, MA, USA
| | - Jack L Pinkus
- Brigham and Women’s Hospital Department of Neurology, Division of Neuromuscular Disease, and Harvard Medical School, Boston, MA, USA
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA and Harvard Medical School, Boston, MA, USA
| | - Clare Baecher-Allan
- Brigham and Women’s Hospital Department of Neurology, Division of Neuromuscular Disease, and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Anthony A Amato
- Brigham and Women’s Hospital Department of Neurology, Division of Neuromuscular Disease, and Harvard Medical School, Boston, MA, USA
| | - David M Dorfman
- Brigham and Women’s Hospital Department of Neurology, Division of Neuromuscular Disease, and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Ioannis M, Foivos P, Dimitrios K. A review on the treatment of sporadic inclusion body myositis with Bimagrumab and Alemtuzumab. Int J Neurosci 2018; 129:297-302. [DOI: 10.1080/00207454.2018.1527329] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mavroudis Ioannis
- Department of Neurology, Leeds General Infirmary, LTHT, Leeds, UK
- Laboratory of Neuropathology and Electron Microscopy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Petridis Foivos
- Laboratory of Neuropathology and Electron Microscopy, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Third Department of Neurology, G. Papanikolaou General Hospital, Thessaloniki, Greece
| | - Kazis Dimitrios
- Third Department of Neurology, G. Papanikolaou General Hospital, Thessaloniki, Greece
| |
Collapse
|
43
|
Naddaf E, Barohn RJ, Dimachkie MM. Inclusion Body Myositis: Update on Pathogenesis and Treatment. Neurotherapeutics 2018; 15:995-1005. [PMID: 30136253 PMCID: PMC6277289 DOI: 10.1007/s13311-018-0658-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inclusion body myositis is the most common acquired myopathy after the age of 50. It is characterized by progressive asymmetric weakness predominantly affecting the quadriceps and/or finger flexors. Loss of ambulation and dysphagia are major complications of the disease. Inclusion body myositis can be associated with cytosolic 5'-nucleotidase 1A antibodies. Muscle biopsy usually shows inflammatory cells surrounding and invading non-necrotic muscle fibers, rimmed vacuoles, congophilic inclusions, and protein aggregates. Disease pathogenesis remains poorly understood and consists of an interplay between inflammatory and degenerative pathways. Antigen-driven, clonally restricted, cytotoxic T cells represent a main feature of the inflammatory component, whereas abnormal protein homeostasis with protein misfolding, aggregation, and dysfunctional protein disposal is the hallmark of the degenerative component. Inclusion body myositis remains refractory to treatment. Better understanding of the disease pathogenesis led to the identification of novel therapeutic targets, addressing both the inflammatory and degenerative pathways.
Collapse
Affiliation(s)
- Elie Naddaf
- Neuromuscular Medicine Division, Department of Neurology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Richard J Barohn
- Neuromuscular Medicine Division, Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, 66103, USA
| | - Mazen M Dimachkie
- Neuromuscular Medicine Division, Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, 66103, USA.
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to give an update on treatment modalities for patients with idiopathic inflammatory myopathies, or shortly myositis, excluding the subgroup inclusion body myositis, based on a literature survey on therapies used in myositis. Few controlled trials have been performed in patients with myositis; therefore, we also included a summary of open-label trials, case series, and case reports. RECENT FINDINGS Glucocorticoid (GC) in high doses is still the first-line treatment of patients with myositis. There is a general recommendation to combine GCs with another immunosuppressive agent in the early phase of disease to better control disease activity and possibly to reduce the risk for GC-related side effects. Furthermore, combining pharmacological treatment with individualized and supervised exercise can be recommended based on evidence. There is some evidence for the effect of rituximab in patients with certain myositis-specific autoantibodies, whereas other biologic agents are currently being tested in clinical trials. SUMMARY Immunosuppressive treatment in combination with exercise is recommended for patients with myositis to reduce disease activity and improve muscle performance. Subgrouping of patients into clinical and serological subtypes may be a way to identify biomarkers for response to specific immunosuppressive and biological agents and should be considered in future trials.
Collapse
Affiliation(s)
- Simone Barsotti
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Ingrid E. Lundberg
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Solna, SE-171 76 Stockholm, Sweden
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW To describe recent advancements in diagnostic and therapeutic approaches to inclusion body myositis (IBM). RECENT FINDINGS Our understanding of the implications of anti-cytosolic 5'-nucleotidase 1A autoantibody status in IBM and other diseases is increasing. Muscle imaging using magnetic resonance techniques and ultrasound is increasingly being performed and characteristic patterns of muscle involvement may help with diagnosis. Longitudinal imaging studies are likely to help with monitoring and as an outcome measure in clinical trials. Recent small-scale studies of Arimoclomol and Rapamycin have shown promising results and further investigation of these medications is ongoing. Exercise is likely to form an increasingly important facet of management of patients with IBM, but the optimal type of exercise programme to enrol patients in is not yet determined. SUMMARY Antibody testing and muscle imaging results may improve our ability to diagnose IBM and the availability of effective disease modifying treatments targeting novel non-inflammatory pathways could soon become a reality. It remains the duty of those involved in the management of patients with IBM to facilitate involvement in clinical trials and other research studies.
Collapse
|
46
|
Novel Therapeutic Options in Treatment of Idiopathic Inflammatory Myopathies. Curr Treat Options Neurol 2018; 20:37. [DOI: 10.1007/s11940-018-0521-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
47
|
Abstract
INTRODUCTION The idiopathic inflammatory myopathies (IIM) dermatomyositis (DM) and polymyositis (PM) are chronic diseases affecting the striated muscles with variable involvement of other organs. Glucocorticoids are considered the cornerstone of treatment, but some patients require adjunctive immunosuppressive agents because of insufficient response to glucocorticoids, flares upon glucocorticoid tapering, or glucocorticoid-related adverse events. Areas covered: The aim of this article was to review (PubMed search until February 2018) the evidence on established and new therapies derived from randomized controlled trials (RCTs) on adult DM and PM. In addition, key data from open-label trials, case reports, and abstracts were included where data from RCT were lacking. Expert commentary: Numerous synthetic and biological immunosuppressive agents are currently available to treat the IIM, sometimes in combination. The choice of the specific medication in the individual patient depends upon the disease phenotype and patient's characteristics. Exercise improves muscle performance without causing disease flares and should be an integral part of the treatment of the IIM. Prompt diagnosis and treatment can lead to better outcome.
Collapse
Affiliation(s)
- Nicolò Pipitone
- a SC di Reumatologia, Dipartimento Medicina Interna e Specialità Mediche, Azienda Unità Sanitaria Locale di Reggio Emilia - Istituto di Ricerca e Cura a Carattere Scientifico , Reggio , Emilia-Romagna , Italy
| | - Carlo Salvarani
- a SC di Reumatologia, Dipartimento Medicina Interna e Specialità Mediche, Azienda Unità Sanitaria Locale di Reggio Emilia - Istituto di Ricerca e Cura a Carattere Scientifico , Reggio , Emilia-Romagna , Italy.,b Rheumatology Department , University of Modena and Reggio Emilia , Italy
| |
Collapse
|
48
|
Abstract
Inflammatory disorders of the skeletal muscle include polymyositis (PM), dermatomyositis (DM), (immune mediated) necrotizing myopathy (NM), overlap syndrome with myositis (overlap myositis, OM) including anti-synthetase syndrome (ASS), and inclusion body myositis (IBM). Whereas DM occurs in children and adults, all other forms of myositis mostly develop in middle aged individuals. Apart from a slowly progressive, chronic disease course in IBM, patients with myositis typically present with a subacute onset of weakness of arms and legs, often associated with pain and clearly elevated creatine kinase in the serum. PM, DM and most patients with NM and OM usually respond to immunosuppressive therapy, whereas IBM is largely refractory to treatment. The diagnosis of myositis requires careful and combinatorial assessment of (1) clinical symptoms including pattern of weakness and paraclinical tests such as MRI of the muscle and electromyography (EMG), (2) broad analysis of auto-antibodies associated with myositis, and (3) detailed histopathological work-up of a skeletal muscle biopsy. This review provides a comprehensive overview of the current classification, diagnostic pathway, treatment regimen and pathomechanistic understanding of myositis.
Collapse
Affiliation(s)
- Jens Schmidt
- Department of Neurology, Muscle Immunobiology Group, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany,Correspondence to: Prof. Dr. Jens Schmidt, MD, FEAN, FAAN, Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany. Tel.: +49 551 39 22355; Fax: +49 551 39 8405; E-mail:
| |
Collapse
|
49
|
Jørgensen AN, Aagaard P, Frandsen U, Boyle E, Diederichsen LP. Blood-flow restricted resistance training in patients with sporadic inclusion body myositis: a randomized controlled trial. Scand J Rheumatol 2018; 47:400-409. [PMID: 29775118 DOI: 10.1080/03009742.2017.1423109] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To investigate the effect of 12 weeks of low-load blood-flow restricted resistance (BFR) training on self-reported and objective physical function, and maximal muscle strength in patients with sporadic inclusion body myositis (sIBM). METHOD Twenty-two patients with sIBM were randomized into a training group (BFR group) or a non-exercising control group, according to CONsolidated Standards Of Reporting Trials (CONSORT) guidelines. The BFR group performed 12 weeks of BFR training twice per week. The primary outcome was the physical function domain of the 36-item Short Form Health Survey (pf-SF-36), which was used to measure self-reported physical function. All patients performed physical function tests (2-Minute Walk Test, Timed Up and Go, and 30-Second Chair Stand), completed the Inclusion Body Myositis Functional Rating Scale (IBMFRS), and were tested for isolated knee extensor muscle strength. RESULTS No effects of the training intervention were observed for pf-SF-36 or the objective physical function tests. Leg muscle strength decreased in controls (-9.2%, p = 0.02), but was unaltered in the BFR group (+0.9%, p = 0.87), resulting in a between-group difference in the per-protocol analysis (p = 0.026). Between-group differences in baseline to follow-up changes emerged for IBMFRS, in favour of the BFR group (p = 0.018). CONCLUSION Twelve weeks of BFR training did not improve self-reported or objective physical function in these sIBM patients. However, the training protocol had a preventive (retaining) effect on the disease-related decline in leg muscle strength, which may aid the long-term preservation of physical function and postpone the need for healthcare assistance.
Collapse
Affiliation(s)
- A N Jørgensen
- a Department of Sports Science and Clinical Biomechanics, SDU Muscle Research Cluster (SMRC) , University of Southern Denmark , Odense , Denmark.,b Department of Clinical Research , University of Southern Denmark , Odense , Denmark
| | - P Aagaard
- a Department of Sports Science and Clinical Biomechanics, SDU Muscle Research Cluster (SMRC) , University of Southern Denmark , Odense , Denmark
| | - U Frandsen
- a Department of Sports Science and Clinical Biomechanics, SDU Muscle Research Cluster (SMRC) , University of Southern Denmark , Odense , Denmark
| | - E Boyle
- a Department of Sports Science and Clinical Biomechanics, SDU Muscle Research Cluster (SMRC) , University of Southern Denmark , Odense , Denmark.,c Division of Epidemiology, Dalla Lana School of Public Health , University of Toronto , Toronto , ON , Canada
| | - L P Diederichsen
- b Department of Clinical Research , University of Southern Denmark , Odense , Denmark.,d Department of Rheumatology , Odense University Hospital , Odense , Denmark
| |
Collapse
|
50
|
Abstract
Idiopathic inflammatory myopathies (IIMs) are heterogeneous disorders that affect the skeletal muscles. Polymyositis, dermatomyositis, and inclusion body myositis are major IIM subsets. Immune-mediated necrotizing myopathy became recognized as a potentially new IIM subset. Since the new classification criteria published by the International Myositis Classification Criteria Project have higher sensitivity and specificity for IIM classification and subclassification than the previous criteria, they should help precise diagnosis. It should be noted that several tests available in current clinical practice, such as electromyography, magnetic resonance imaging, and other myositis-specific autoantibodies than anti-Jo-1 antibodies, were not included in the new criteria. As for treatment, glucocorticoids are used empirically as the first-line treatment despite their various adverse effects. Concomitant treatment with steroid-sparing immunosuppressive agents, including methotrexate, azathioprine, calcineurin inhibitors, mycophenolate mofetil, and cyclophosphamide, reduces successfully initial glucocorticoid doses for the remission induction, the relapse risk during glucocorticoid tapering, and adverse effects of glucocorticoids. Treatment with biologics, including rituximab and abatacept, seems promising in some IIM patients. Multi-target treatment with glucocorticoids and several steroid-sparing immunosuppressive agents is effective in refractory IIM patients. Considering proven steroid-sparing efficacy and tolerability of multi-target treatment in patients with other autoimmune diseases, it should be a good therapeutic option for IIMs.
Collapse
Affiliation(s)
- Hirokazu Sasaki
- a Department of Rheumatology, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University (TMDU) , Tokyo , Japan
| | - Hitoshi Kohsaka
- a Department of Rheumatology, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University (TMDU) , Tokyo , Japan
| |
Collapse
|