Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
J Cachexia Sarcopenia Muscle ; 15(3): 1108-1120, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613252

ABSTRACT

BACKGROUND: Finding sensitive clinical outcome measures has become crucial in natural history studies and therapeutic trials of neuromuscular disorders. Here, we focus on 1-year longitudinal data from quantitative magnetic resonance imaging (MRI) and phosphorus magnetic resonance spectroscopy (31P MRS) in a placebo-controlled study of sirolimus for inclusion body myositis (IBM), also examining their links to functional, strength, and clinical parameters in lower limb muscles. METHODS: Quantitative MRI and 31P MRS data were collected at 3 T from a single site, involving 44 patients (22 on placebo, 22 on sirolimus) at baseline and year-1, and 21 healthy controls. Assessments included fat fraction (FF), contractile cross-sectional area (cCSA), and water T2 in global leg and thigh segments, muscle groups, individual muscles, as well as 31P MRS indices in quadriceps or triceps surae. Analyses covered patient-control comparisons, annual change assessments via standard t-tests and linear mixed models, calculation of standardized response means (SRM), and exploration of correlations between MRI, 31P MRS, functional, strength, and clinical parameters. RESULTS: The quadriceps and gastrocnemius medialis muscles had the highest FF values, displaying notable heterogeneity and asymmetry, particularly in the quadriceps. In the placebo group, the median 1-year FF increase in the quadriceps was 3.2% (P < 0.001), whereas in the sirolimus group, it was 0.7% (P = 0.033). Both groups experienced a significant decrease in cCSA in the quadriceps after 1 year (P < 0.001), with median changes of 12.6% for the placebo group and 5.5% for the sirolimus group. Differences in FF and cCSA changes between the two groups were significant (P < 0.001). SRM values for FF and cCSA were 1.3 and 1.4 in the placebo group and 0.5 and 0.8 in the sirolimus group, respectively. Water T2 values were highest in the quadriceps muscles of both groups, significantly exceeding control values in both groups (P < 0.001) and were higher in the placebo group than in the sirolimus group. After treatment, water T2 increased significantly only in the sirolimus group's quadriceps (P < 0.01). Multiple 31P MRS indices were abnormal in patients compared to controls and remained unchanged after treatment. Significant correlations were identified between baseline water T2 and FF at baseline and the change in FF (P < 0.001). Additionally, significant correlations were observed between FF, cCSA, water T2, and functional and strength outcome measures. CONCLUSIONS: This study has demonstrated that quantitative MRI/31P MRS can discern measurable differences between placebo and sirolimus-treated IBM patients, offering promise for future therapeutic trials in idiopathic inflammatory myopathies such as IBM.


Subject(s)
Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Muscle, Skeletal , Myositis, Inclusion Body , Sirolimus , Humans , Myositis, Inclusion Body/drug therapy , Magnetic Resonance Imaging/methods , Male , Female , Magnetic Resonance Spectroscopy/methods , Muscle, Skeletal/drug effects , Muscle, Skeletal/diagnostic imaging , Sirolimus/therapeutic use , Sirolimus/pharmacology , Middle Aged , Aged , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology
2.
Neuromuscul Disord ; 37: 36-51, 2024 04.
Article in English | MEDLINE | ID: mdl-38522330

ABSTRACT

Since the publication of the 2013 European Neuromuscular Center (ENMC) diagnostic criteria for Inclusion Body Myositis (IBM), several advances have been made regarding IBM epidemiology, pathogenesis, diagnostic tools, and clinical trial readiness. Novel diagnostic tools include muscle imaging techniques such as MRI and ultrasound, and serological testing for cytosolic 5'-nucleotidase-1A antibodies. The 272nd ENMC workshop aimed to develop new diagnostic criteria, discuss clinical outcome measures and clinical trial readiness. The workshop started with patient representatives highlighting several understudied symptoms and the urge for a timely diagnosis. This was followed by presentations from IBM experts highlighting the new developments in the field. This report is composed of two parts, the first part providing new diagnostic criteria on which consensus was achieved. The second part focuses on the use of outcome measures in clinical practice and clinical trials, highlighting current limitations and outlining the goals for future studies.


Subject(s)
Myositis, Inclusion Body , Myositis , Humans , Consensus , Magnetic Resonance Imaging , Myositis/diagnosis , Myositis, Inclusion Body/therapy , Myositis, Inclusion Body/drug therapy , Netherlands , Outcome Assessment, Health Care
4.
Lancet Neurol ; 22(10): 900-911, 2023 10.
Article in English | MEDLINE | ID: mdl-37739573

ABSTRACT

BACKGROUND: Inclusion body myositis is the most common progressive muscle wasting disease in people older than 50 years, with no effective drug treatment. Arimoclomol is an oral co-inducer of the cellular heat shock response that was safe and well-tolerated in a pilot study of inclusion body myositis, reduced key pathological markers of inclusion body myositis in two in-vitro models representing degenerative and inflammatory components of this disease, and improved disease pathology and muscle function in mutant valosin-containing protein mice. In the current study, we aimed to assess the safety, tolerability, and efficacy of arimoclomol in people with inclusion body myositis. METHODS: This multicentre, randomised, double-blind, placebo-controlled study enrolled adults in specialist neuromuscular centres in the USA (11 centres) and UK (one centre). Eligible participants had a diagnosis of inclusion body myositis fulfilling the European Neuromuscular Centre research diagnostic criteria 2011. Participants were randomised (1:1) to receive either oral arimoclomol 400 mg or matching placebo three times daily (1200 mg/day) for 20 months. The randomisation sequence was computer generated centrally using a permuted block algorithm with randomisation numbers masked to participants and trial staff, including those assessing outcomes. The primary endpoint was the change from baseline to month 20 in the Inclusion Body Myositis Functional Rating Scale (IBMFRS) total score, assessed in all randomly assigned participants, except for those who were randomised in error and did not receive any study medication, and those who did not meet inclusion criteria. Safety analyses included all randomly assigned participants who received at least one dose of study medication. This trial is registered with ClinicalTrials.gov, number NCT02753530, and is completed. FINDINGS: Between Aug 16, 2017 and May 22, 2019, 152 participants with inclusion body myositis were randomly assigned to arimoclomol (n=74) or placebo (n=78). One participant was randomised in error (to arimoclomol) but not treated, and another (assigned to placebo) did not meet inclusion criteria. 150 participants (114 [76%] male and 36 [24%] female) were included in the efficacy analyses, 73 in the arimoclomol group and 77 in the placebo group. 126 completed the trial on treatment (56 [77%] and 70 [90%], respectively) and the most common reason for treatment discontinuation was adverse events. At month 20, mean IBMFRS change from baseline was not statistically significantly different between arimoclomol and placebo (-3·26, 95% CI -4·15 to -2·36 in the arimoclomol group vs -2·26, -3·11 to -1·41 in the placebo group; mean difference -0·99 [95% CI -2·23 to 0·24]; p=0·12). Adverse events leading to discontinuation occurred in 13 (18%) of 73 participants in the arimoclomol group and four (5%) of 78 participants in the placebo group. Serious adverse events occurred in 11 (15%) participants in the arimoclomol group and 18 (23%) in the placebo group. Elevated transaminases three times or more of the upper limit of normal occurred in five (7%) participants in the arimoclomol group and one (1%) in the placebo group. Tubulointerstitial nephritis was observed in one (1%) participant in the arimoclomol group and none in the placebo group. INTERPRETATION: Arimoclomol did not improve efficacy outcomes, relative to placebo, but had an acceptable safety profile in individuals with inclusion body myositis. This is one of the largest trials done in people with inclusion body myositis, providing data on disease progression that might be used for subsequent clinical trial design. FUNDING: US Food and Drug Administration Office of Orphan Products Development and Orphazyme.


Subject(s)
Myositis, Inclusion Body , United States , Adult , Humans , Animals , Female , Male , Mice , Myositis, Inclusion Body/drug therapy , Pilot Projects , Double-Blind Method , Disease Progression
5.
Mod Rheumatol Case Rep ; 8(1): 83-85, 2023 Dec 29.
Article in English | MEDLINE | ID: mdl-37210209

ABSTRACT

We present the case of a 75-year-old man diagnosed with myasthenia gravis (MG) based on lower leg weakness and ptosis for the past 2 months before admission to our hospital. The patient was anti-acetylcholine receptor antibody-positive at admission. He was treated with pyridostigmine bromide and prednisolone, which improved the ptosis, but the lower leg muscle weakness remained. An additional lower leg magnetic resonance imaging examination suggested myositis. Inclusion body myositis (IBM) was diagnosed after a subsequent muscle biopsy. Although MG is often associated with inflammatory myopathy, IBM is rare. There is no effective treatment for IBM, but various treatment possibilities have recently been proposed. This case emphasises that myositis complications, including IBM, should be considered when elevated creatine kinase levels are observed and conventional treatments do not address chronic muscle weakness.


Subject(s)
Myasthenia Gravis , Myositis, Inclusion Body , Myositis , Male , Humans , Aged , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/diagnosis , Myositis, Inclusion Body/drug therapy , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Myositis/diagnosis , Muscle Weakness/complications , Treatment Outcome
6.
Neurotherapeutics ; 19(3): 911-921, 2022 04.
Article in English | MEDLINE | ID: mdl-35394612

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.


Subject(s)
Autoimmune Diseases , Myositis, Inclusion Body , Myositis , Polymyositis , Autoantibodies , Humans , Myositis/drug therapy , Myositis, Inclusion Body/drug therapy , Polymyositis/pathology , Polymyositis/therapy
7.
Eur J Neurol ; 29(4): 1284-1287, 2022 04.
Article in English | MEDLINE | ID: mdl-35253967

ABSTRACT

BACKGROUND AND PURPOSE: To provide further evidence for sirolimus, a mammalian target of rapamycin inhibitor, as a treatment strategy for patients with inclusion body myositis (IBM). METHODS: We acquired longitudinal clinical data and immunological assessments of CD8+ T-cell subsets in peripheral blood for evaluation of potential anti-inflammatory treatment effects of sirolimus. RESULTS: Therapy with sirolimus 2 mg/day by mouth led to rapid and sustained clinical improvement of motor symptoms for an observation period of more than 1 year. Treatment was well tolerated, with no occurrence of adverse effects. We did not observe a meaningful alteration of CD8+ T-cell subsets in our patient after 9 and 12 months compared to baseline. CONCLUSIONS: The significant and persistent clinical improvement highlights the use of sirolimus as a potential treatment option in patients with IBM. In light of the lack of immunological treatment effects observed for cytotoxic CD8+ T cells, further studies should investigate the potential myoprotective effects of sirolimus.


Subject(s)
Myositis, Inclusion Body , Sirolimus , CD8-Positive T-Lymphocytes , Humans , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/drug therapy , Sirolimus/adverse effects , Sirolimus/therapeutic use
8.
J Neurol ; 269(8): 4154-4160, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35244767

ABSTRACT

OBJECTIVE: Dysphagia can be troublesome in sporadic inclusion body myositis (sIBM) and oculopharyngeal muscular dystrophy (OPMD), but no established treatment exists. Cricopharyngeal muscle botulinum toxin injection has at case level been reported to be effective. We evaluated safety and efficacy of botulinum toxin injections in the cricopharyngeal muscle in patients with dysphagia due to sIBM or OPMD. METHODS: Participants were included from our outpatient clinic. Cricopharyngeal constriction was confirmed by laryngoscopy. After EMG confirmation of needle placement in the cricopharyngeal muscle, botulinum toxin A was injected in awake patients. An individualized dose of 5-10 units of botulinum toxin A was applied initially and titrated up a maximum of 3 times. Outcome measures were change in dysphagia questionnaire, timed cold-water swallow test and subjective dysphagia status (worse, unchanged, improved). Due to the need for individualized dosing and a limited number of available patients, an uncontrolled, un-blinded design was used. RESULTS: Thirteen patients, 3 with OPMD, received at least 1 injection. In the dysphagia questionnaire, all but 2 subjects, none with subjective worsening, improved (p < 0.001). Subjectively, seven felt an improvement, 4 no change and 2 a worsening. No overall change was seen the timed cold-water swallow test. No serious adverse events were observed. CONCLUSION: Botulinum toxin injection of the cricopharyngeal muscle in patients with OPMD and sIBM had a beneficial effect on dysphagia in most of the treated patients. Two of 13 patients experienced a temporary worsening not reflected in dysphagia score. Limitations are the un-blinded and un-randomized design and subjective assessments methods. PROSPECTIVE TRIAL REGISTRATION: EudraCT-number: 2014-002210-23.


Subject(s)
Botulinum Toxins, Type A , Deglutition Disorders , Muscular Dystrophy, Oculopharyngeal , Myositis, Inclusion Body , Neuromuscular Agents , Botulinum Toxins, Type A/adverse effects , Botulinum Toxins, Type A/therapeutic use , Deglutition Disorders/drug therapy , Deglutition Disorders/etiology , Humans , Muscular Dystrophy, Oculopharyngeal/complications , Muscular Dystrophy, Oculopharyngeal/drug therapy , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/drug therapy , Neuromuscular Agents/adverse effects , Neuromuscular Agents/therapeutic use , Prospective Studies , Water
9.
J Neuropathol Exp Neurol ; 80(11): 1060­1067, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34643702

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.


Subject(s)
Mitochondria/pathology , Myositis, Inclusion Body/diagnosis , Myositis, Inclusion Body/pathology , Polymyositis/pathology , Aged , Biopsy , Diagnosis, Differential , Disease Progression , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Muscle, Skeletal/pathology , Myositis, Inclusion Body/drug therapy , Organ Transplantation , Polymyositis/drug therapy , Retrospective Studies
10.
Neurology ; 96(12): e1595-e1607, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33597289

ABSTRACT

OBJECTIVE: To assess long-term (2 years) effects of bimagrumab in participants with sporadic inclusion body myositis (sIBM). METHODS: Participants (aged 36-85 years) who completed the core study (RESILIENT [Efficacy and Safety of Bimagrumab/BYM338 at 52 Weeks on Physical Function, Muscle Strength, Mobility in sIBM Patients]) were invited to join an extension study. Individuals continued on the same treatment as in the core study (10 mg/kg, 3 mg/kg, 1 mg/kg bimagrumab or matching placebo administered as IV infusions every 4 weeks). The co-primary outcome measures were 6-minute walk distance (6MWD) and safety. RESULTS: Between November 2015 and February 2017, 211 participants entered double-blind placebo-controlled period of the extension study. Mean change in 6MWD from baseline was highly variable across treatment groups, but indicated progressive deterioration from weeks 24-104 in all treatment groups. Overall, 91.0% (n = 142) of participants in the pooled bimagrumab group and 89.1% (n = 49) in the placebo group had ≥1 treatment-emergent adverse event (AE). Falls were slightly higher in the bimagrumab 3 mg/kg group vs 10 mg/kg, 1 mg/kg, and placebo groups (69.2% [n = 36 of 52] vs 56.6% [n = 30 of 53], 58.8% [n = 30 of 51], and 61.8% [n = 34 of 55], respectively). The most frequently reported AEs in the pooled bimagrumab group were diarrhea 14.7% (n = 23), involuntary muscle contractions 9.6% (n = 15), and rash 5.1% (n = 8). Incidence of serious AEs was comparable between the pooled bimagrumab and the placebo group (18.6% [n = 29] vs 14.5% [n = 8], respectively). CONCLUSION: Extended treatment with bimagrumab up to 2 years produced a good safety profile and was well-tolerated, but did not provide clinical benefits in terms of improvement in mobility. The extension study was terminated early due to core study not meeting its primary endpoint. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov identifier NCT02573467. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that for patients with sIBM, long-term treatment with bimagrumab was safe, well-tolerated, and did not provide meaningful functional benefit. The study is rated Class IV because of the open-label design of extension treatment period 2.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Myositis, Inclusion Body/drug therapy , Accidental Falls , Adult , Aged , Aged, 80 and over , Double-Blind Method , Female , Humans , Male , Middle Aged , Muscle Strength/drug effects , Myositis, Inclusion Body/complications , Time , Treatment Outcome , Walk Test
12.
PLoS One ; 15(12): e0231064, 2020.
Article in English | MEDLINE | ID: mdl-33264289

ABSTRACT

Sporadic inclusion body myositis (sIBM) is the most common idiopathic inflammatory myopathy, and several reports have suggested that mitochondrial abnormalities are involved in its etiology. We recruited 9 sIBM patients and found significant histological changes and an elevation of growth differential factor 15 (GDF15), a marker of mitochondrial disease, strongly suggesting the involvement of mitochondrial dysfunction. Bioenergetic analysis of sIBM patient myoblasts revealed impaired mitochondrial function. Decreased ATP production, reduced mitochondrial size and reduced mitochondrial dynamics were also observed in sIBM myoblasts. Cell vulnerability to oxidative stress also suggested the existence of mitochondrial dysfunction. Mitochonic acid-5 (MA-5) increased the cellular ATP level, reduced mitochondrial ROS, and provided protection against sIBM myoblast death. MA-5 also improved the survival of sIBM skin fibroblasts as well as mitochondrial morphology and dynamics in these cells. The reduction in the gene expression levels of Opa1 and Drp1 was also reversed by MA-5, suggesting the modification of the fusion/fission process. These data suggest that MA-5 may provide an alternative therapeutic strategy for treating not only mitochondrial diseases but also sIBM.


Subject(s)
Indoleacetic Acids/therapeutic use , Mitochondria, Muscle/metabolism , Myositis, Inclusion Body/drug therapy , Phenylbutyrates/therapeutic use , Adenosine Triphosphate/biosynthesis , Aged , Aged, 80 and over , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Cells, Cultured , DNA, Mitochondrial/genetics , Drug Evaluation, Preclinical , Dynamins/biosynthesis , Dynamins/genetics , Female , Fibroblast Growth Factors/blood , Fibroblasts/drug effects , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Growth Differentiation Factor 15/biosynthesis , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/genetics , Humans , Indoleacetic Acids/pharmacology , Male , Middle Aged , Mitochondria, Muscle/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/ultrastructure , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Oxygen Consumption , Phenylbutyrates/pharmacology , Reactive Oxygen Species/metabolism , Retrospective Studies
13.
Rheumatology (Oxford) ; 59(11): 3515-3525, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32830270

ABSTRACT

OBJECTIVE: Damage to the vascular endothelium is strongly implicated in the pathogenesis of idiopathic inflammatory myopathies (IIM). Normally, high-density lipoprotein (HDL) protects the vascular endothelium from damage from oxidized phospholipids, which accumulate under conditions of oxidative stress. The current work evaluated the antioxidant function of HDL in IIM patients. METHODS: HDL's antioxidant function was measured in IIM patients using a cell-free assay, which assesses the ability of isolated patient HDL to inhibit oxidation of low-density lipoproteins and is reported as the HDL inflammatory index (HII). Cholesterol profiles were measured for all patients, and subgroup analysis included assessment of oxidized fatty acids in HDL and plasma MPO activity. A subgroup of IIM patients was compared with healthy controls. RESULTS: The antioxidant function of HDL was significantly worse in patients with IIM (n = 95) compared with healthy controls (n = 41) [mean (S.d.) HII 1.12 (0.61) vs 0.82 (0.13), P < 0.0001]. Higher HII associated with higher plasma MPO activity [mean (S.d.) 13.2 (9.1) vs 9.1 (4.6), P = 0.0006] and higher oxidized fatty acids in HDL. Higher 5-hydroxyeicosatetraenoic acid in HDL correlated with worse diffusion capacity in patients with interstitial lung disease (r = -0.58, P = 0.02), and HDL's antioxidant function was most impaired in patients with autoantibodies against melanoma differentiation-associated protein 5 (MDA5) or anti-synthetase antibodies. In multivariate analysis including 182 IIM patients, higher HII was associated with higher disease activity and DM diagnosis. CONCLUSION: The antioxidant function of HDL is abnormal in IIM patients and may warrant further investigation for its role in propagating microvascular inflammation and damage in this patient population.


Subject(s)
Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Lung Diseases, Interstitial/metabolism , Myositis/metabolism , Adult , Aged , Amino Acyl-tRNA Synthetases/immunology , Autoantibodies/immunology , Case-Control Studies , Chromatography, Liquid , Dermatomyositis/drug therapy , Dermatomyositis/immunology , Dermatomyositis/metabolism , Endothelium, Vascular , Fatty Acids/metabolism , Female , Glucocorticoids/therapeutic use , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Immunologic Factors/therapeutic use , Immunosuppressive Agents/therapeutic use , Interferon-Induced Helicase, IFIH1/immunology , Lung Diseases, Interstitial/immunology , Male , Middle Aged , Myositis/drug therapy , Myositis/immunology , Myositis, Inclusion Body/drug therapy , Myositis, Inclusion Body/immunology , Myositis, Inclusion Body/metabolism , Oxidation-Reduction , Peroxidase/metabolism , Polymyositis/drug therapy , Polymyositis/immunology , Polymyositis/metabolism , Pulmonary Diffusing Capacity , Spectrometry, Mass, Electrospray Ionization
14.
Neurology ; 95(14): e1971-e1978, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32690797

ABSTRACT

OBJECTIVE: To assess the long-term safety and tolerability and to monitor benefits of extended use of bimagrumab in individuals with sporadic inclusion body myositis (sIBM) who completed a single-dose core study. METHODS: In this multicenter, open-label extension study, 10 adults received bimagrumab 10 mg/kg IV every 4 weeks up to 2 years (104 weeks). Safety (primary endpoint) was assessed by recording adverse events (AEs). Clinical benefits were assessed by changes from baseline in thigh muscle volume (TMV), lean body mass (LBM), 6-minute walk distance (6MWD), handgrip, and quadriceps strength. RESULTS: Participants had a mean age of 70.1 (SD 10.4) years. All participants (n = 10) discontinued the treatment due to early termination of the study (n = 7) or AEs (n = 3; myocardial infarction, esophageal carcinoma, and dementia, none of which were treatment related). The most common AEs were muscle spasms and falls (both 9 of 10, 90%), followed by diarrhea (6 of 10, 60%) and acne and skin eruption (both 5 of 10, 50%). At weeks 8 and 16, mean TMV increased from baseline by 4.1% (SD 4.3%) and 4.5% (SD 6.3%). Mean LBM increased from baseline and was sustained at 6.9% (SD 3.9%) at week 76. Means of 6MWD showed a progressive decline from baseline to week 76, during which there was a modest numerical increase in handgrip strength and no significant changes in quadriceps strength. CONCLUSIONS: Long-term treatment up to 2 years with bimagrumab had a good safety profile and was well tolerated in individuals with sIBM. An increase in muscle mass was noted on a group level; however, there was no evidence of clinical improvement. CLINICALTRIALSGOV IDENTIFIER: NCT02250443. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that for patients with sIBM, long-term bimagrumab treatment was safe and well tolerated and did not lead to functional improvement.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Myositis, Inclusion Body/drug therapy , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Muscle Strength/drug effects , Time , Treatment Outcome
15.
Semin Neurol ; 40(3): 342-348, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32252099

ABSTRACT

The inflammatory myopathies comprise disorders of immune-mediated muscle injury. The histopathology and clinical features help distinguish them. Juvenile dermatomyositis (JDM) is the most common form of myositis in children and adolescents. Children with JDM present with proximal muscle weakness and characteristic rashes. The presentation is similar in children and adults, but JDM is a primary disorder and the adult form often is concerning for a paraneoplastic syndrome. Proximal muscle weakness occurs with dermatomyositis, polymyositis, and immune-mediated necrotizing myopathy, but the latter two conditions have no dermatologic findings or distinct tissue changes which set them apart from dermatomyositis. Inclusion body myositis, also included in the inflammatory myopathies, presents with more distal involvement, and microscopically exhibits identifiable rimmed vacuoles. We review key features of these disorders, focusing in more detail on JDM because it is more often encountered by the child neurologist.


Subject(s)
Autoimmune Diseases of the Nervous System , Dermatomyositis , Inflammation , Myositis, Inclusion Body , Adolescent , Adult , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/physiopathology , Child , Dermatomyositis/diagnosis , Dermatomyositis/drug therapy , Dermatomyositis/immunology , Dermatomyositis/physiopathology , Humans , Inflammation/diagnosis , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Myositis, Inclusion Body/diagnosis , Myositis, Inclusion Body/drug therapy , Myositis, Inclusion Body/immunology , Myositis, Inclusion Body/physiopathology
16.
Rheumatol Int ; 40(2): 191-205, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31680207

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.


Subject(s)
Antirheumatic Agents/therapeutic use , Biological Products/therapeutic use , Immunologic Factors/therapeutic use , Myositis/drug therapy , Tumor Necrosis Factor Inhibitors/therapeutic use , Abatacept/therapeutic use , Alemtuzumab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Basiliximab/therapeutic use , Dermatomyositis/drug therapy , Dermatomyositis/immunology , Humans , Immunosuppressive Agents/therapeutic use , Myositis/immunology , Myositis, Inclusion Body/drug therapy , Myositis, Inclusion Body/immunology , Polymyositis/drug therapy , Polymyositis/immunology , Rituximab/therapeutic use
17.
Semin Arthritis Rheum ; 50(3): 498-502, 2020 06.
Article in English | MEDLINE | ID: mdl-31806155

ABSTRACT

OBJECTIVES: To describe two cases of anti-PM-Scl antibody-associated systemic sclerosis with evidence of inclusion body myositis on muscle biopsy. METHODS: Two female patients with anti-PM-Scl antibody-associated systemic sclerosis developed progressive proximal myopathy. Both patients had profound muscle weakness that was refractory to treatment with glucocorticoids with or without other oral immunosuppressive agents. Quadriceps muscle biopsy in both cases indicated inclusion body myositis (IBM). Monthly intravenous immunoglobulin (IVIG) infusions were added to the treatment regimen. RESULTS: One patient's myopathy appears to have temporarily stabilized with IVIG infusions (2 g/Kg) every four weeks, though severe residual muscle weakness has persisted. The other patient's myopathy continues to progress slowly, despite being on 4-weekly IVIG infusions, along with weekly oral methotrexate and 6-monthly rituximab infusions. In this report, we compare these two patients with two similar cases that were previously reported and hypothesize a possible pathomechanism of this association. CONCLUSION: This extremely rare association of IBM and anti-PM-Scl antibody-associated scleromyositis sheds new light on the possible pathogenesis of IBM. It strengthens the hypothesis that autoimmune muscle disease can potentially trigger myodegeneration. Whether early intervention with aggressive immunosuppressive therapy can prevent progression to treatment-refractory IBM, should be a subject of future research.


Subject(s)
Muscle Weakness/etiology , Myositis, Inclusion Body/diagnosis , Scleroderma, Systemic/diagnosis , Disease Progression , Female , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunosuppressive Agents/therapeutic use , Middle Aged , Myositis, Inclusion Body/drug therapy , Raynaud Disease/etiology , Scleroderma, Systemic/drug therapy
18.
Rinsho Shinkeigaku ; 59(12): 806-813, 2019 Dec 25.
Article in Japanese | MEDLINE | ID: mdl-31761834

ABSTRACT

A global, randomized, double-blind placebo-controlled study was conducted to confirm that BYM338 (bimagrumab), an anti-activin type II receptor antibody, improves motor function in patients with sporadic inclusion body myositis after 52 weeks' treatment consisting of intravenous administration every 4 weeks at doses of 10, 3, and 1 mg/kg. In a Japanese sub-population (20 patients in total, 5 per dose group), no significant differences in the change from baseline of the 6-minute walking distance at Week 52 (primary endpoint) were observed between the placebo group and each BYM338 dose group. Furthermore, the lean body mass as an indicator of skeletal muscle mass increased in all BYM338 groups compared with the placebo group and the effects were dose-dependent. Overall, the Japanese sub-population showed similar trends as observed in the entire population (251 patients in total).


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Myositis, Inclusion Body/drug therapy , Adult , Aged , Aged, 80 and over , Asian People , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Motor Activity , Myositis, Inclusion Body/physiopathology , Treatment Outcome
19.
Lancet Neurol ; 18(9): 834-844, 2019 09.
Article in English | MEDLINE | ID: mdl-31397289

ABSTRACT

BACKGROUND: Inclusion body myositis is an idiopathic inflammatory myopathy and the most common myopathy affecting people older than 50 years. To date, there are no effective drug treatments. We aimed to assess the safety, efficacy, and tolerability of bimagrumab-a fully human monoclonal antibody-in individuals with inclusion body myositis. METHODS: We did a multicentre, double-blind, placebo-controlled study (RESILIENT) at 38 academic clinical sites in Australia, Europe, Japan, and the USA. Individuals (aged 36-85 years) were eligible for the study if they met modified 2010 Medical Research Council criteria for inclusion body myositis. We randomly assigned participants (1:1:1:1) using a blocked randomisation schedule (block size of four) to either bimagrumab (10 mg/kg, 3 mg/kg, or 1 mg/kg) or placebo matched in appearance to bimagrumab, administered as intravenous infusions every 4 weeks for at least 48 weeks. All study participants, the funder, investigators, site personnel, and people doing assessments were masked to treatment assignment. The primary outcome measure was 6-min walking distance (6MWD), which was assessed at week 52 in the primary analysis population and analysed by intention-to-treat principles. We used a multivariate normal repeated measures model to analyse data for 6MWD. Safety was assessed by recording adverse events and by electrocardiography, echocardiography, haematological testing, urinalysis, and blood chemistry. This trial is registered with ClinicalTrials.gov, number NCT01925209; this report represents the final analysis. FINDINGS: Between Sept 26, 2013, and Jan 6, 2016, 251 participants were enrolled to the study, of whom 63 were assigned to each bimagrumab group and 62 were allocated to the placebo group. At week 52, 6MWD change from baseline did not differ between any bimagrumab dose and placebo (least squares mean treatment difference for bimagrumab 10 mg/kg group, 17·6 m, SE 14·3, 99% CI -19·6 to 54·8; p=0·22; for 3 mg/kg group, 18·6 m, 14·2, -18·2 to 55·4; p=0·19; and for 1 mg/kg group, -1·3 m, 14·1, -38·0 to 35·4; p=0·93). 63 (100%) participants in each bimagrumab group and 61 (98%) of 62 in the placebo group had at least one adverse event. Falls were the most frequent adverse event (48 [76%] in the bimagrumab 10 mg/kg group, 55 [87%] in the 3 mg/kg group, 54 [86%] in the 1 mg/kg group, and 52 [84%] in the placebo group). The most frequently reported adverse events with bimagrumab were muscle spasms (32 [51%] in the bimagrumab 10 mg/kg group, 43 [68%] in the 3 mg/kg group, 25 [40%] in the 1 mg/kg group, and 13 [21%] in the placebo group) and diarrhoea (33 [52%], 28 [44%], 20 [32%], and 11 [18%], respectively). Adverse events leading to discontinuation were reported in four (6%) participants in each bimagrumab group compared with one (2%) participant in the placebo group. At least one serious adverse event was reported by 21 (33%) participants in the 10 mg/kg group, 11 (17%) in the 3 mg/kg group, 20 (32%) in the 1 mg/kg group, and 20 (32%) in the placebo group. No significant adverse cardiac effects were recorded on electrocardiography or echocardiography. Two deaths were reported during the study, one attributable to subendocardial myocardial infarction (secondary to gastrointestinal bleeding after an intentional overdose of concomitant sedatives and antidepressants) and one attributable to lung adenocarcinoma. Neither death was considered by the investigator to be related to bimagrumab. INTERPRETATION: Bimagrumab showed a good safety profile, relative to placebo, in individuals with inclusion body myositis but did not improve 6MWD. The strengths of our study are that, to the best of our knowledge, it is the largest randomised controlled trial done in people with inclusion body myositis, and it provides important natural history data over 12 months. FUNDING: Novartis Pharma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Myositis, Inclusion Body/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Treatment Outcome
20.
Neurol Neuroimmunol Neuroinflamm ; 6(4): e581, 2019 07.
Article in English | MEDLINE | ID: mdl-31355317

ABSTRACT

Objective: To assess whether canakinumab, a monoclonal antibody against IL-1ß approved for autoinflammatory diseases, is effective as target-specific therapy in patients with sporadic inclusion body myositis (sIBM). Methods: Because in sIBM IL-1ß colocalizes with amyloid precursor protein and upregulates amyloid aggregates enhancing degeneration, targeting IL-1ß with canakinumab may arrest disease progression. On this basis, 5 ambulatory patients with sIBM participated in an institutional review board--approved open-labeled study with 150 mg canakinumab [4 bimonthly, then monthly subcutaneous injections] for a mean period of 15.8 months. Patients were assessed bimonthly with a manual dynamometer in 12 proximal and distal muscles and with grip force (GF) in both hands. Total muscle strength (TMS) was expressed in kilograms. Efficacy was defined as >15% increased strength after 12 months. Results: Patient 1 stopped at month 5 because of 23% loss in TMS and 32.35% in GF; patient 2 showed 37.1% increase in TMS and 13% in GF by month 9; patient 3 exhibited 26.7% reduction in TMS and 10% in GF at month 33; patient 4 showed 6.5% reduction in TMS and 1.6% in GF after 15 months, denoting relative stability; and patient 5 showed 30.4% loss in TMS and 20.8% in GF after 18 months. In patients 2 and 4, in whom 3-year longitudinal data were available, no effect on disease progression was noted. Conclusions: In this long-term, open-label study, canakinumab showed small, but not clinically appreciable, stabilizing benefits in 2 of 5 patients with sIBM over 1 year, was ineffective in 2 others, and might have worsened one. No patient improved. Classification of evidence: This study provides Class IV evidence that canakinumab was ineffective for patients with sIBM.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Interleukin-1beta/antagonists & inhibitors , Myositis, Inclusion Body/drug therapy , Aged , Aged, 80 and over , Disease Progression , Female , Hand Strength , Humans , Injections, Subcutaneous , Male , Middle Aged , Muscle Strength/drug effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...