Open-access Inflammatory myopathies: an update for neurologists

Miopatias inflamatórias: uma atualização para neurologistas

ABSTRACT

Idiopathic inflammatory myopathies (IIM) are a heterogenous group of treatable myopathies. Patients present mainly to the rheumatologist and neurologists, complaining of acute or subacute onset of proximal weakness. Extramuscular manifestations may occur, including involvement of the lungs, skin, and joints. Classically, the diagnosis used to be made based on the creatine kinase level increase, abnormalities in electroneuromyography and presence of inflammatory infiltrates in the muscle biopsy. Recently, the importance of autoantibodies has increased, and now they may be identified in more than half of IIM patients. The continuous clinicoseropathological improvement in IIM knowledge has changed the way we see these patients and how we classify them. In the past, only polymyositis, dermatomyositis and inclusion body myopathy were described. Currently, immune-mediated necrotizing myopathy, overlap myositis and antisynthetase syndrome have been considered the most common forms of IIM in clinical practice, increasing the spectrum of classification. Patients previously considered to have polymyositis, in fact have these other forms of seropositive IIM. In this article, we reviewed the new concepts of classification, a practical way to make the diagnosis and how to plan the treatment of patients suffering from IIM.

Keywords: Myositis; Dermatomyositis; Polymyositis; Immune-Mediated Necrotizing Myopathy

RESUMO

As miopatias inflamatórias idiopáticas (MII) são um grupo heterogêneo de miopatias tratáveis. Os pacientes procuram principalmente o reumatologista e o neurologista, queixando-se de início agudo ou subagudo de fraqueza proximal. Manifestações extramusculares podem ocorrer, incluindo envolvimento dos pulmões, pele e articulações. Classicamente, o diagnóstico era feito com base na elevação dos níveis de creatina quinase, anormalidades na eletroneuromiografia e presença de infiltrados inflamatórios na biópsia muscular. Recentemente, a importância dos autoanticorpos aumentou, e agora eles podem ser identificados em mais da metade dos pacientes com MII. A contínua melhora clínico-soropatológica no conhecimento do MII mudou a forma como vemos esses pacientes e como os classificamos. No passado, apenas polimiosite, dermatomiosite e miopatia por corpos de inclusão eram descritas. Atualmente, a miopatia necrosante imunomediada, a miosite de sobreposição e a síndrome antissintetase têm sido consideradas as formas mais comuns de MII na prática clínica, aumentando o espectro de classificação. Pacientes previamente considerados como portadores de polimiosite, na verdade, têm uma dessas outras formas de MII soropositivas. Neste artigo, revisamos os novos conceitos de classificação, uma forma prática de fazer o diagnóstico e como planejar o tratamento de pacientes que sofrem de MII.

Palavras-chave: Miosite; Dermatomiosite; Polimiosite; Miopatia Necrosante Imunomediada

INTRODUCTION

Myopathies are a heterogeneous group of diseases that affect skeletal muscle tissue, classified into hereditary and acquired forms1. In clinical practice, acquired causes are the priority for identification, and must not be missed, because they are potentially treatable. This group includes toxic drug exposure myopathy, infectious conditions, endocrinopathies and, mainly, idiopathic inflammatory myopathies (IIM), also known as myositis, or systemic autoimmune myopathies2.

IIM are characterized by muscle weakness as the main symptom, and muscle inflammation as the central physiopathology3. Extramuscular manifestations may occur, including involvement of the lungs, skin, and joints4. Classically, some subtypes of IIM are described: dermatomyositis, polymyositis, immune-mediated necrotizing myopathy (IMNM), inclusion-body myositis (IBM), and overlap myositis, wherein some authors include antisynthetase syndrome, whereas other authors categorize these as a separate group3 , 5. Here we reviewed the heterogenous group of IIM, regarding advances in its diagnosis, classification, and treatment options, in a practical approach, for general neurologists.

HOW TO PERFORM THE DIAGNOSIS

Patients with IIM will present with acute or subacute onset of proximal weakness, usually over several weeks or a few months, with little or no atrophy, normal tendon reflexes (unless the patient has a severe muscle weakness), and normal sensory tests. The patients suffer from impaired walking, getting up from the chair and climbing stairs as well as lifting their arms. Dysphagia and neck weakness may occur in around one-third of patients, then a dropping head may be a clinical presentation3 , 6 , 7. Although frequently considered as a symptom of IIM, muscle pain or tenderness are not common, but may be present in some forms of IIM with perifascial involvement, such as dermatomyositis8. Isolated muscle pain with normal physical examination is not evidence of IIM. In patients with IBM, a progressive course disease, atrophy in the quadriceps and finger flexors, with hand weakness, are the clinical hallmarks, which is different from other IIM9.

In the clinical history, signs and symptoms of other organs and systems must be examined, especially skin lesions, such as periorbital blue-purple rash, reddish rash on the face, anterior chest (V-sign), and shoulders (shawl sign), and a violaceous eruption on the knuckles (Gottron’s papules), which are characteristics of dermatomyositis (Figure 1)3. Pulmonary complications, due to interstitial lung disease, may occur in 10 to 40% of patients, as well as fever, arthralgia, scleroderma, and Raynaud’s phenomenon, mainly in overlap and antisynthetase syndrome, defined as the presence of myopathy, fever, interstitial lung disease, Raynaud’s phenomenon, arthritis, and “mechanic’s hands”10.

Figure 1.
Skin manifestations of dermatomyositis. A) Reddish purple rash on the face, predominantly in periorbital region. B) Violaceous rash on the dorsal aspect of interphalangeal joints (Gottron’s sign). C) Anterior chest rash (V-sign).

Creatine kinase (CK) is the main biomarker, usually over 10 times the upper limit of normality, but in some rare cases it is normal or just mildly elevated7. Aldolase is another muscle enzyme and may be the only elevated muscle enzyme in perimysial and perifascial inflammation11. Electroneuromyography (EMG) is not essential for the diagnosis, and is usually performed when there are doubts regarding differential diagnosis for myasthenic syndromes. In IIM, the EMG demonstrates motor potential of myopathic characteristics, with small motor unit potentials of short duration, and early recruitment12. Spontaneous activity, such as fibrillation and positive waves, is usually found, because there is active degeneration of muscle fibers, but this is not universal or specific13.

Recently, the importance and use of autoantibodies has increased, and they are identified in more than 50% of IIM patients4. Currently, commercial antibody panels for myositis are available. These autoantibodies may help in the diagnosis, classification of patients into more homogeneous groups, and prediction of additional clinical complications and treatment response14. Approximately 70% of dermatomyositis patients have one of the five dermatomyositis-specific autoantibodies15 , 16. Among them, anti-Mi2 is a well-known antibody related to classical skin lesion, high association with calcinosis and good response to immunosuppressant treatment12 , 14. Other dermatomyositis-specific autoantibodies are anti-TIF1-γ, anti-NXP-2, anti-MDA-5 and anti-SAE (Table 1). In addition, the discovery of autoantibodies related to antisynthetase syndrome, mainly anti-Jo-1, present in up to a quarter of IIM, allowed for the creation of a new subgroup of IIM, based on clinicoserological diagnosis17. Other autoantibodies in this syndrome have been described, such as anti-PL-7 and anti-PL-12 (Table 1)18 , 19. Anti-SRP and anti-HMGCR are present in most patients with IMNM, and have prognostic implications, because the first is more severe and resistant to immunosuppressors (Table 1)7.

Table 1.
Main myositis specific autoantibodies.

Muscle imaging, mainly MRI, is now widely used as a biomarker for diagnosis and treatment follow-up. Magnetic resonance of the muscles demonstrates an increase in heterogeneous signals in the muscles in the T2- and STIR-weighted sequences, as well as an anomalous enhancement by gadolinium, indicating the presence of edema in the muscles (Figure 2A)20. Patients with dermatomyositis frequently present T2 / STIR hyperintensities in the perifascial region of individual muscles, a characteristic seen less often in other subtypes of IIM (Figure 2B-C)21. Although useful for differential diagnosis between hereditary and inflammatory myopathies, MRI was not included in the recent classification criteria for IIM.

Figure 2.
Magnetic resonance imaging in inflammatory myopathies. A) Axial STIR-weighted imaging of a patient with polymyositis with hyperintense areas of muscular edema, mainly in quadriceps (arrows). B) Axial STIR-weighted in a patient with dermatomyositis demonstrating a perifascial hyperintensity (arrows) and well-demarcated fascial hyperintense signal indicating fasciitis (arrowhead). C) Coronal STIR-weighted imaging with peripheral hyperintensity (arrows) in a patient with dermatomyositis.

Muscle biopsy is still the gold standard for diagnosis, and is indicated in most patients, although it can be prevented for those with typical skin manifestation or known autoantibody syndromes (for instance, DM and ASS). Muscle biopsy continues to be essential for myositis-specific antibody-negative IIM and IBM diagnosis. Ideally, the analysis must include immunohistochemistry with antibodies against inflammatory cells, such as T and B lymphocytes (CD4, CD8 and CD20), macrophages (CD68), major histocompatibility complex (MHC) class I antigen and membrane attack complexes (MACs)3 , 7. Two techniques are used to obtain muscle samples: needle biopsy and open biopsy. Both are widely used, usually in the form of an outpatient procedure. We would prefer the open technique, because this has the advantage of obtaining a larger sample, under direct examination, allowing the collection of multiple fragments from different fascicles, increasing the diagnostic yield, especially in IIM, although it is more invasive, requiring a two to four cm incision22.

A proposed diagnostic approach for IIM is presented in Figure 3.

Figure 3.
Flowchart for IIM diagnosis.

HOW TO CLASSIFY THE PATIENTS

For many years, IIM had been classified into three subgroups, including polymyositis and dermatomyositis, based on the Bohan and Peter criteria23 , 24, and inclusion body myositis (IBM) using the Griggs criteria25. However, over the last decade, reviewed classifications, based on expert opinion, have been proposed5 , 12, with advances of imaging methods, better pathological descriptions, and discovery of myositis-specific autoantibodies (MSA)14. Newly identified autoantibodies are reclassifying some patients previously considered as dermatomyositis or polymyositis, now categorized as other forms of IIM5.

IIM are currently classified into several subgroups: dermatomyositis (DM), polymyositis (PM), inclusion body myositis (IBM), immune-mediated necrotizing myopathy (IMNM), antisynthetase syndrome (ASS), and overlap myositis5 , 7 , 12 , 14 , 26. Some authors classify ASS into overlap myositis, whereas others categorize it as a distinct subform of myositis7 , 12 , 27. We prefer to present ASS as a distinct entity. An overview of the classification and the main characteristics of the IIM subgroups are shown here (Table 2).

Table 2.
Classification e key characteristics of the IIM subtypes.

Dermatomyositis and amyopathic dermatomyositis

DM is a condition seen in adults and children, mainly females, characterized by cutaneous manifestations and proximal muscle weakness. Presentation includes generalized erythema, mainly in the upper region of the trunk (shawl or V sign), purplish lesion around the eyes (heliotrope), and violet eruptions on the finger joints (Gottron’s sign), all of which are photosensitive (Figure 1)16. Some patients develop an interstitial lung disease, Raynaud phenomenon and calcifications in muscles or skin, although the trend is to categorize them separately if an ASS-specific autoantibody is recognized5.

There is an increase in CK levels at some stage of the disease in most patients. However, CK may be normal at the onset of the disease, after major muscle atrophies, or in a subgroup, representing around 5-10%, known as hypo myopathic or amyopathic, in which muscle weakness is mild or absent7. In these patients, anti-MDA5 or anti-SAE autoantibodies are more likely to be present5.

The clinical scenario of weakness, typical skin lesions and elevation of CK allows a diagnosis and the beginning of treatment, before performing a muscle biopsy. In cases of diagnostic doubt, a biopsy can be performed. Muscle tissue demonstrates inflammatory infiltration, predominantly CD4 T cells, and macrophages, surrounding blood vessels (perivascular infiltrates) and invading the perimysium (perimysial infiltrates) (Figure 4A-B)28. However, some patients do not have infiltrates but have prominent necrosis, especially in perifascicular regions, which may be indistinguishable from immune-mediated necrotising myopathy7. Perifascicular atrophy and perifascicular increase of MHC class 1 labeling are characteristic but are no longer considered pathognomonic5. Recently, type 1 interferon (IFN1) signature and perifascicular positivity for myxovirus resistance protein A (MxA) by immunohistochemistry have been specifically associated with dermatomyositis and are potential markers on muscle histology for specific diagnosis29.

Figure 4.
Histological characteristics of the main inflammatory myopathies. A and B) HE stains in a patient with dermatomyositis demonstrate perivascular infiltrates (arrows) in A and perifascicular fiber atrophy (arrows) in B. C) HE stain showing endomysial lymphocytic infiltration (arrow) in a patient with polymyositis. D) MHC-I stain in a patient with polymyositis showing a strong diffuse brownish color. E) HE stain in a patient with IBM demonstrating rimmed vacuoles (arrows). F) HE stain showing necrosis (arrows) in a patient with immune-mediated necrotizing myopathy.

Neoplastic investigation is recommended, both in adults and in the juvenile form. The incidence of neoplasms in DM is increased by five to seven times when compared to the general population, with a reported frequency of 9 to 32%30 , 31.

Polymyositis

Polymyositis manifests in adults (rarely in children), with proximal weakness, high CK (10 to 50 times the value of reference), usually without myalgia12. The disease progresses over weeks to months, and therefore is different from rhabdomyolysis, which has an acute and painful onset. Polymyositis, as a distinct entity, is considered uncommon, and its existence has been questioned. Most patients previously classified in this group, following new clinicoseropathological knowledge, are categorized in another IIM27.

There are no skin lesions and therefore diagnosis simply with clinical criteria is not possible. Unlike DM, muscle biopsy in PM is mandatory, making a differential diagnosis with IBM, DM with atypical skin lesions, muscular dystrophies, IMNM, and rare forms of ASS. In the muscle, there is an inflammatory infiltrate between the muscle fibers and an increase in CD8-T-lymphocytes, with invasion of non-necrotic fibers (Figure 4C-D)28. The pathophysiology is different from DM, and it is worth emphasizing that DM is not polymyositis with skin lesions.

Inclusion body myositis (IBM)

IBM is a late-onset form of myopathy, and though classified in the group of IIM, it has a degenerative physiopathology. It is considered the most common form of myopathy in patients over 50 years of age and is more common in men, in a 2:1 proportion32. Clinically, the disease affects predominantly the quadriceps and the gastrocnemius muscles in the lower limbs and the finger flexors in the upper limbs9. The onset is slowly progressive, and the muscle involvement is usually asymmetrical3.

Magnetic resonance imaging shows signal alteration in T1-weighted sequences, mainly in the distal portion of the quadriceps, meaning fat-infiltration of this muscle, and corresponding finds may also be detected by ultrasound33.

The skeletal muscle abnormalities include a T-cell-mediated endomysial inflammatory reaction in association with degenerative changes characterized by the presence of rimmed vacuoles (Figure 4E), intracytoplasmic inclusions formed by the accumulation of abnormal proteins, 𝛽-amyloid deposits, and mitochondrial changes9 , 34.

Autoantibodies against cytosolic 5ʹ-nucleotidase 1A (NT5C1a) has been studied, but its sensitivity (around 30%) and questionable specificity (up to 20-30% of patients with other reumatological conditions may have this autoantibody) do not yet allow its recommendation in clinical practice34. In the absence of a serological marker, muscle biopsy is still mandatory in suspected IBM patients. On the other hand, serologies for chronic viruses, such as HIV, HTLV, and hepatitis C and B must be done, because IBM might develop from these viral infections, resulting in a typical disease course, except for a younger age of onset34.

Recently, it has been shown that most IBM patients present T-cells expressing CD57, a surface marker of T-cell large granular lymphocytic leukemia, and up to one-third of IBM patients in a cohort met criteria for this type of leukemia, with cytopenias35. Although, there is no consensus regarding this point, we think that hematological evaluation may be warranted, and blood cell counts, as well as blood flow cytometry must be considered.

Immune-Mediated Necrotizing Myopathy

IMNM is a myopathy with peculiar clinicopathological features, occurring much more frequently than PM, and presenting in a large range of age14. Onset may be acute or subacute, with high levels of CK, which can be triggered by viral infection, cancer, and statin use36. However, it can develop without any of these factors present. In children, the disease may have a chronic slowly progressive course, and the patients can be initially diagnosed with muscular dystrophy37. Pathologically, muscle necrosis, presence of CD68+ macrophages and little or no lymphocytic inflammation are observed (Figure 4F)36.

The 224th ENMC International Workshop categorized IMNM into three subgroups according to autoantibodies: antisignal recognition particle (SRP) IMNM, anti3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) IMNM, and seronegative IMNM36. This classification is important not only for diagnosis, but also for the prognosis and planning of the treatment. Anti-SRP patients usually have a more severe muscle involvement and a more resistant response to immunosuppressors. Anti-HMGCR were initially found in IMNM patients who were taking statins, but now we know there are many patients with this autoantibody without statin exposure. The association with malignancy is not consistent in anti-SRP and anti-HMGCR subgroups, but the risk is increased in seronegative patients38.

Antisynthetase syndrome

ASS is classically characterized by the presence of inflammatory myopathy, interstitial lung disease and joint involvement, although other findings may be present, such as fever, "mechanic's hands", and Raynaud's phenomenon39 , 40. Serologically, it is characterized by the presence of antibodies against aminoacyl-transfer RNA (tRNA) synthetases. The list of antisynthetase autoantibodies has been evolved over the last decades and three are the most studied and recognized ones: anti-Jo-1 (antihistidyl), anti-PL-7 (antithreonyl), and anti-PL-12 (antialanyl) (Table 1). Now the presence of one of the ASS-specific autoantibodies is considered a necessary condition to classify the patient as having ASS5 , 16 , 41.

Some authors consider ASS as being in a broader group, known as overlap myositis. In fact, patients with ASS often have signs and symptoms that overlap with other connective tissue disorders (CTD), such as Raynaud’s phenomenon, interstitial lung disease, arthritis, gastroesophageal reflux disease, and general systemic symptoms17. Additionally, patients with ASS may present skin rashes and, histologically, the inflammatory involvement resembles that of dermatomyositis. A muscle biopsy demonstrates T-cell and macrophage infiltrations and perifascicular atrophy or perifascicular necrosis7. Without serological evaluation, ASS patients may be misdiagnosed either as PM, IMNM or, if there is skin lesion or perifascicular atrophy on histology, as DM.

Importantly, not all patients with ASS have muscle weakness. For instance, muscle involvement is more common in anti-Jo-1 patients, whereas pulmonary involvement is more common in the presence of anti-PL-12 and anti-PL-7, with some patients presenting no clinical weakness5. Morbidity and mortality are associated with rapidly progressive pulmonary involvement39 , 41.

Overlap myositis

Overlap myositis is the association of inflammatory myopathy with at least one clinical overlap feature of a connective tissue disorder, such as systemic lupus erythematous (SLE), systemic sclerosis (SS), rheumatoid arthritis, or Sjögren’s syndrome14. In these cases, myositis appears as an additional manifestation of a more complex rheumatological disease. As mentioned above, ASS may be considered as an overlap myositis, because of intersect characteristics of myositis, skin lesions, lung involvement, and Raynaud’s phenomenon17.

There are also some autoantibodies associated to this kind of presentation. The most common of these antibodies are anti-PM (polymyositis) /Scl (scleroderma) and anti-U1-RNP (ribonucleoprotein) antibodies. These are observed in up to 20% of all patients with myositis4. Anti-PM/Scl is associated with upper limb weakness predominance and anti-U1-RNP is associated with interstitial lung disease5. Muscle biopsy may present perivascular inflammation, perifascicular necrosis and MHC-I increase.

Interestingly, if we consider ASS as part of a wider overlap myositis group, this will represent approximately 50% of all patients with myositis, followed by dermatomyositis and IMNM, whereas polymyositis is the rarest form of IIM27.

DIFFERENTIAL DIAGNOSIS WITH MUSCULAR DYSTROPHY

Some clinical situations can cause mistakes in the differentiation between IIM and muscular dystrophy. There are several forms of muscular dystrophy that may present an inflammatory infiltrate, such as fascioscapulohumeral dystrophy or girdle dystrophy due to dysferlin or anoctamin deficiency42 , 43. In these cases, the onset can mimic an acquired myopathy, with proximal weakness of subacute installation and very high CK. In cohorts of patients with dysferlinopathies, up to 25% of the patients are initially defined as having inflammatory myopathy44. To differentiate these conditions, in addition to reassessing the clinical phenotype and the response or not to immunosuppressants, some biopsy findings may help. The CD8 lymphocyte infiltrates and diffuse increase in MHC class I labeling are more frequent in inflammatory myopathies, while in dystrophies, lymphocytic infiltrates are focal, and rarely have increased labeling for CD8 or MHC class I. Furthermore, in dystrophies, the finding of fibrosis between the fibers is more frequent and immunohistochemistry may be diagnostic by revealing the absence of muscle proteins (such as dysferlin, sarcoglycans, dystrophin).

On the other hand, IMNM caused by anti-SRP and anti-HMGCR antibodies may present with an insidious course, clinically confusing with muscular dystrophy, even with muscle biopsy showing some degree of connective tissue increase, and with little response to corticosteroids42. In these cases, the presence of serum autoantibodies and the finding of necrosis as the main histological characteristic, associated with an increase in immunohistochemical staining for macrophages (CD68) and MHC class I, are the elements that favor the diagnosis of IMNM.

HOW TO PLAN THE TREATMENT

Due to the rarity and heterogeneity of IIM, evidence guiding how to plan patients’ treatment is limited, and essentially based on case reports, series, expert opinions, and a few non-high-quality randomized trials (Figure 5)45. Treatment in clinical practice consists of corticosteroid therapy, and prednisone 0.5 to 1 mg/kg is usually prescribed in mild cases, over a period of four to eight weeks with serum CK levels monitoring and muscle strength evaluation, before tapering or adding another immunosuppressor7 , 46. Intravenous pulses of methylprednisolone (MP) 1,000 mg daily for three to five days is recommended for more severe cases associated with functional disability, dysphagia, interstitial lung disease, ulcerated skin lesions or a rapidly progressive course46. In even more dramatic cases with significant weakness (for example, patients unable to walk unassisted), respiratory involvement, severe dysphagia or refractory to initial corticosteroid treatment, intravenous human immunoglobulin (IVIg), at a dose of 2 g/kg, divided over two to five days may be used46. Both pulse therapies (MP and IVIg) may be repeated monthly until there is a good clinical response, and in some case, to maintain remission3.The total duration of treatment is not consensual, but most experts start gradual reduction of corticosteroids after disease remission, followed by progressive reduction of other immunosupressors, in a period over one to two years following disease control46.

Figure 5.
Schematic flowchart for IIM treatment.

Immunosuppressants are indicated in practically all cases for glucocorticoid sparing and to avoid relapses during corticosteroid tapering3 , 47. The most used drugs are azathioprine (2 to 3 mg/kg/day), methotrexate (10 to 25mg weekly), mycophenolate mofetil (2 to 3 g/day) and cyclosporine (3 to 5 mg/kg/day)7 , 46. The choice is based mainly upon the experience of the prescriber and adverse effects profile of each patient.

Immunobiologic agents that have been approved for the treatment of other immune diseases may be considered in refractory cases. The most used is the rituximab (anti-CD20 antibody) at a dose of 2g46. It seems effective in some patients with dermatomyositis, polymyositis, or necrotizing autoimmune myositis3. A multicenter placebo-controlled study involving 200 patients did not demonstrate a response in the eighth week (the primary endpoint), but at week 44, when all had received rituximab, 83% presented the definition of improvement in treatment48. Another option proposed for refractory patients is to maintain chronically subcutaneous immunoglobulin49.

For IBM patients there is no defined treatment. Available studies have failed to demonstrate a real benefit in using immunosuppressants34. Currently, intravenous human immunoglobulin may be used for the treatment of dysphagia3.

Physical exercise programs under supervision are safe in all types of IIM50. The patient must be encouraged to start a routine of exercises to increase strength, reduce disability, so helping a faster improvement51.

In conclusion, advances have been seen in IIM over the last decade. There are new concepts for classification, especially based on the development in serodiagnosis, with many autoantibodies currently known. Probably, two-thirds of IIM patients have an autoantibody, which may make the subgroups more homogenous, improving not only the diagnosis, but prognosis and treatment planning. Knowledge in this field is continuing and rapidly evolving, so neurologists must be updated to better assist their patients with these treatable myopathies.

References

  • 1. Dubowitz V, Sewry CA, Oldfors A. Muscle biopsy: a practical approach. 4th ed. Oxford (UK): Saunders Elsevier; 2013.
  • 2. Barohn RJ, Dimachkie MM, Jackson CE. A pattern recognition approach to patients with a suspected myopathy. Neurol Clin. 2014 Aug;32(3):569-33. https://doi.org/10.1016/j.ncl.2014.04.008
    » https://doi.org/10.1016/j.ncl.2014.04.008
  • 3. Dalakas MC. Inflammatory mscle diseases. N Engl J Med. 2015 Apr 15;372(4):1734-47. https://doi.org/10.1056/NEJMra1402225
    » https://doi.org/10.1056/NEJMra1402225
  • 4. Findlay AR, Goyal NA, Mozaffar T. An overview of polymyositis and dermatomyositis. Muscle Nerve. 2015 May;51(5):638-56. https://doi.org/10.1002/mus.24566
    » https://doi.org/10.1002/mus.24566
  • 5. Tanboon J, Uruha A, Stenzel W, Nishino I. Where are we moving in the classification of idiopathic inflammatory myopathies? Curr Opin Neurol. 2020 Oct;33(5):590-603. https://doi.org/10.1097/WCO.0000000000000855
    » https://doi.org/10.1097/WCO.0000000000000855
  • 6. Oh TH, Brumfield KA, Hoskin TL, Stolp KA, Murray JA, Bassford JR. Dysphagia in inflammatory myopathy: clinical characteristics, treatment strategies, and outcome in 62 patients. Mayo Clin Proc. 2007 Apr 1;82(4):441-7. https://doi.org/10.4065/82.4.441
    » https://doi.org/10.4065/82.4.441
  • 7. Selva-O'Callaghan A, Pinal-Fernandez I, Trallero-Araguás E, Milisenda JC, Grau-Junyent JM, Mammen AL. Classification and management of adult inflammatory myopathies. Lancet Neurol. 2018 Sep;17(9):816-28. https://doi.org/10.1016/S1474-4422(18)30254-0
    » https://doi.org/10.1016/S1474-4422(18)30254-0
  • 8. Ernste FC, Reed AM. Idiopathic inflammatory myopathies: current trends in pathogenesis, clinical features, and up-to-date treatment recommendations. Mayo Clin Proc. 2013 Jan;88(1):83-105. https://doi.org/10.1016/j.mayocp.2012.10.017
    » https://doi.org/10.1016/j.mayocp.2012.10.017
  • 9. de Camargo LV, de Carvalho MS, Shinjo SK, de Oliveira ASB, Zanoteli E. Clinical, histological, and immunohistochemical findings in inclusion body myositis. Biomed Res Int. 2018 Jan 29;2018:5069042. https://doi.org/10.1155/2018/5069042
    » https://doi.org/10.1155/2018/5069042
  • 10. Kiely PDW, Chua F. Interstitial lung disease in inflammatory myopathies: clinical phenotypes and prognosis. Curr Rheumatol Rep. 2013 Sep;15(9):359. https://doi.org/10.1007/s11926-013-0359-6
    » https://doi.org/10.1007/s11926-013-0359-6
  • 11. Nozaki K, Pestronk A. High aldolase with normal creatine kinase in serum predicts a myopathy with perimysial pathology. J Neurol Neurosurg Psychiatry. 2009 Aug;80(8):904-8. https://doi.org/10.1136/jnnp.2008.161448
    » https://doi.org/10.1136/jnnp.2008.161448
  • 12. Schmidt J. Current classification and management of inflammatory myopathies. J Neuromuscul Dis. 2018;5(2):109-29. https://doi.org/10.3233/JND-180308
    » https://doi.org/10.3233/JND-180308
  • 13. Mills KR. The basics of electromyography. J Neurol Neurosurg Psychiatry. 2005 Jun 16;76(2 Suppl 2):ii32-5. https://doi.org/10.1136/jnnp.2005.069211
    » https://doi.org/10.1136/jnnp.2005.069211
  • 14. Tanboon J, Nishino I. Classification of idiopathic inflammatory myopathies: pathology perspectives. Curr Opin Neurol. 2019 Oct;32(5):704-14. https://doi.org/10.1097/WCO.0000000000000740
    » https://doi.org/10.1097/WCO.0000000000000740
  • 15. Betteridge Z, McHugh N. Myositis-specific autoantibodies: an important tool to support diagnosis of myositis. J Intern Med. 2016; 280(1):8-23. https://doi.org/10.1111/joim.12451
    » https://doi.org/10.1111/joim.12451
  • 16. Mammen AL, Allenbach Y, Stenzel W, Benveniste O; ENMC 239th Workshop Study Group. 239th ENMC International Workshop: classification of dermatomyositis, Amsterdam, the Netherlands, 14-16 December 2018. Neuromuscul Disord. 2020 Jan 1;30(1):P70-92. https://doi.org/10.1016/j.nmd.2019.10.005
    » https://doi.org/10.1016/j.nmd.2019.10.005
  • 17. Lega JC, Fabien N, Reynaud Q, Durieu I, Durupt S, Dutertre M, et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun Rev. 2014 Sep;13(9):883-91. https://doi.org/10.1016/j.autrev.2014.03.004
    » https://doi.org/10.1016/j.autrev.2014.03.004
  • 18. Souza FHC, Cruellas MG, Levy-Neto M, Shinjo SK. Anti-synthetase syndrome: anti-PL-7, anti-PL-12 and anti-EJ. Rev Bras Reumatol. 2013 Jul-Aug;53(4):352-7. https://doi.org/10.1590/S0482-50042013000400007
    » https://doi.org/10.1590/S0482-50042013000400007
  • 19. Cruellas MGP, Viana VST, Levy-Neto M, Souza FHC, Shinjo SK. Myositis-specific and myositis-associated autoantibody profiles and their clinical associations in a large series of patients with polymyositis and dermatomyositis. Clinics (São Paulo). 2013 Jul;68(7):909-14. https://doi.org/10.6061/clinics/2013(07)04
    » https://doi.org/10.6061/clinics/2013(07)04
  • 20. Barsotti S, Zampa V, Talarico R, Minichilli F, Ortori S, Iacopetti V, et al. Thigh magnetic resonance imaging for the evaluation of disease activity in patients with idiopathic inflammatory myopathies followed in a single center. Muscle Nerve. 2016 Oct;54(4):666-72. https://doi.org/10.1002/mus.25099
    » https://doi.org/10.1002/mus.25099
  • 21. Pinal-Fernandez I, Casal-Dominguez M, Carrino JA, Lahouti AH, Basharat P, Albayda J, et al. Thigh muscle MRI in immune-mediated necrotising myopathy: extensive oedema, early muscle damage and role of anti-SRP autoantibodies as a marker of severity. Ann Rheum Dis. 2017 Apr;76(4):681-7. https://doi.org/10.1136/annrheumdis-2016-210198
    » https://doi.org/10.1136/annrheumdis-2016-210198
  • 22. Silva AMS, Zanoteli E. Instrumento diagnóstico em miopatias: biópsias musculares. Rev Paul Reumatol. 2017 Oct-Dec;16(4):27-30. https://doi.org/10.46833/reumatologiasp.2017.16.4.27-30
    » https://doi.org/10.46833/reumatologiasp.2017.16.4.27-30
  • 23. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975 Feb 13;292(7):344-7. https://doi.org/10.1056/NEJM197502132920706
    » https://doi.org/10.1056/NEJM197502132920706
  • 24. Bohan A, Peter JB. Polymyositis and dermatomyositis (second of two parts). N Engl J Med. 1975 Feb 20;292(8):403-7. https://doi.org/10.1056/NEJM197502202920807
    » https://doi.org/10.1056/NEJM197502202920807
  • 25. Griggs RC, Askanas V, DiMauro S, Engel A, Karpati G, Mendell JR, et al. Inclusion body myositis and myopathies. Ann Neurol. 1995 Nov;38(5):705-13. https://doi.org/10.1002/ana.410380504
    » https://doi.org/10.1002/ana.410380504
  • 26. Lundberg IE, Tjärnlund A, Bottai M, Werth VP, Pilkington C, Visser M, et al. 2017 European League Against Rheumatism/American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major sub- groups. Ann Rheum Dis. 2017 Dec;76(12):1955-64. https://doi.org/10.1136/annrheumdis-2017-211468
    » https://doi.org/10.1136/annrheumdis-2017-211468
  • 27. Senécal J-L, Raynauld J-P, Troyanov Y. Editorial: a new classification of adult autoimmune myositis. Arthritis Rheumatol. 2017 May;69(5):878-84. https://doi.org/10.1002/art.40063
    » https://doi.org/10.1002/art.40063
  • 28. Dalakas MC. Muscle biopsy findings in inflammatory myopathies. Rheum Dis Clin North Am. 2002 Nov;28(4):779-98. https://doi.org/10.1016/s0889-857x(02)00030-3
    » https://doi.org/10.1016/s0889-857x(02)00030-3
  • 29. Pinal-Fernandez I, Casal-Dominguez M, Derfoul A, Pak K, Plotz P, Miller FW, et al. Identification of distinctive interferon gene signatures in different types of myositis. Neurology. 2019 Sep 17;93(12):e1193-204. https://doi.org/10.1212/WNL.0000000000008128
    » https://doi.org/10.1212/WNL.0000000000008128
  • 30. Hill CL, Zhang Y, Sigurgeirsson B, Pukkala E, Mellemkjaer L, Airio A, et al. Frequency of specific cancer types in dermatomyositis and polymyositis: a population-based study. Lancet. 2001 Jan 13;357(9250):96-100. https://doi.org/10.1016/S0140-6736(00)03540-6
    » https://doi.org/10.1016/S0140-6736(00)03540-6
  • 31. Chen Y-J, Wu C-Y, Huang Y-L, Wang C-B, Shen J-L, Chang Y-T. Cancer risks of dermatomyositis and polymyositis: a nationwide cohort study in Taiwan. Arthritis Res Ther. 2010 Apr 16;12(2):R70. https://doi.org/10.1186/ar2987
    » https://doi.org/10.1186/ar2987
  • 32. Mastaglia FL, Phillips BA. Idiopathic inflammatory myopathies: epidemiology, classification, and diagnostic criteria. Rheum Dis Clin North Am. 2002 Nov;28(4):723-41. https://doi.org/10.1016/s0889-857x(02)00021-2
    » https://doi.org/10.1016/s0889-857x(02)00021-2
  • 33. Guimaraes JB, Cavalcante WCP, Cruz IAN, Nico MA, Ormond Filho AG, Silva AMS, et al. Musculoskeletal ultrasound in inclusion body myositis: a comparative study with magnetic resonance imaging. Ultrasound Med Biol. 2021 Aug 1;47(8):P2186-92. https://doi.org/10.1016/j.ultrasmedbio.2021.04.019
    » https://doi.org/10.1016/j.ultrasmedbio.2021.04.019
  • 34. Greenberg SA. Inclusion body myositis: clinical features and pathogenesis. Nat Rev Rheumatol. 2019 May;15(5):257-72. https://doi.org/10.1038/s41584-019-0186-x
    » https://doi.org/10.1038/s41584-019-0186-x
  • 35. Greenberg SA, Pinkus JL, Amato AA, Kristensen T, Dorfman DM. Association of inclusion body myositis with T cell large granular lymphocytic leukaemia. Brain. 2016 May;139(5):1348-60. https://doi.org/10.1093/brain/aww024
    » https://doi.org/10.1093/brain/aww024
  • 36. Allenbach Y, Mammen AL, Benveniste O, Stenzel W; Immune-Mediated Necrotizing Myopathies Working Group. 224th ENMC International Work-shop: clinico-sero-pathological classification of immune-mediated necrotizing myopathies Zandvoort, The Netherlands, 14-16 October 2016. Neuromuscul Disord. 2018 Jan 1;28(1):P87-99. https://doi.org/10.1016/j.nmd.2017.09.016
    » https://doi.org/10.1016/j.nmd.2017.09.016
  • 37. Liang W-C, Uruha A, Suzuki S, Murakami N, Takeshita E, Chen W-Z, et al. Pediatric necrotizing myopathy associated with anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase antibodies. Rheumatology (Oxford). 2017 Feb;56(2):287-93. https://doi.org/10.1093/rheumatology/kew386
    » https://doi.org/10.1093/rheumatology/kew386
  • 38. Lim J, Rietveld A, De Bleecker JL, Badrising UA, Saris CGJ, van der Kooi AJ, et al. Seronegative patients form a distinctive subgroup of immune-mediated necrotizing myopathy. Neurol Neuroimmunol Neuroinflamm. 2018 Oct 16;6(1):e513. https://doi.org/10.1212/NXI.0000000000000513
    » https://doi.org/10.1212/NXI.0000000000000513
  • 39. Witt LJ, Curran JJ, Strek ME. The diagnosis and treatment of antisynthetase syndrome. Clin Pulm Med. 2016 Sep;23(5):218-26. https://doi.org/10.1097/CPM.0000000000000171
    » https://doi.org/10.1097/CPM.0000000000000171
  • 40. Katzap E, Barilla-LaBarca M-L, Marder G. Antisynthetase syndrome. Curr Rheumatol Rep. 2011 Apr 1;13(3):175-81. https://doi.org/10.1007/s11926-011-0176-8
    » https://doi.org/10.1007/s11926-011-0176-8
  • 41. Connors GR, Christopher-Stine L, Oddis CV, Danoff SK. Interstitial lung disease associated with the idiopathic inflammatory myopathies: what progress has been made in the past 35 years? Chest. 2010 Dec 1;138(6):P1464-74. https://doi.org/10.1378/chest.10-0180
    » https://doi.org/10.1378/chest.10-0180
  • 42. Benveniste O, Romero NB. Myositis or dystrophy? Traps and pitfalls. Presse Med. 2011 Apr;40(4):e249-55. https://doi.org/10.1016/j.lpm.2010.11.023
    » https://doi.org/10.1016/j.lpm.2010.11.023
  • 43. Silva AMS, Coimbra-Neto AR, Souza PVS, Winckler PB, Gonçalves MVM, Cavalcanti EBU, et al. Clinical and molecular findings in a cohort of ANO5-related myopathy. Ann Clin Transl Neurol. 2019 Jul;6(7):1225-38. https://doi.org/10.1002/acn3.50801
    » https://doi.org/10.1002/acn3.50801
  • 44. Nguyen K, Bassez G, Krahn M, Bernard R, Laforêt P, Labelle V, et al. Phenotypic study in 40 patients with dysferlin gene mutations: high frequency of atypical phenotypes. Arch Neurol. 2007 Aug;64(8):1176-82. https://doi.org/10.1001/archneur.64.8.1176
    » https://doi.org/10.1001/archneur.64.8.1176
  • 45. Gordon PA, Winer JB, Hoogendijk JE, Choy EHS. Immunosuppressant and immunomodulatory treatment for dermatomyositis and polymyositis. Cochrane Database Syst Rev. 2012 Aug 15;2012(8):CD003643. https://doi.org/10.1002/14651858.CD003643.pub4
    » https://doi.org/10.1002/14651858.CD003643.pub4
  • 46. de Souza FHC, de Araújo DB, Vilela VS, Bezerra MC, Simões RS, Bernardo WM, et al. Guidelines of the Brazilian Society of Rheumatology for the treatment of systemic autoimmune myopathies. Adv Rheumatol. 2019 Jan 22;59(1):6. https://doi.org/10.1186/s42358-019-0048-x
    » https://doi.org/10.1186/s42358-019-0048-x
  • 47. Joffe MM, Love LA, Leff RL, Fraser DD, Targoff IN, Hicks JE, et al. Drug therapy of the idiopathic inflammatory myopathies: predictors of response to prednisone, azathioprine, and methotrexate and a comparison of their efficacy. Am J Med. 1993 Apr 1;94(4):P379-87. https://doi.org/10.1016/0002-9343(93)90148-i
    » https://doi.org/10.1016/0002-9343(93)90148-i
  • 48. Oddis CV, Reed AM, Aggarwal R, Rider LG, Ascherman DP, Levesque MC, et al. Rituximab in the treatment of refractory adult and juvenile dermatomyositis and adult polymyositis: a randomized, placebo- phase trial. Arthritis Rheum. 2013 Feb;65(2):314-24. https://doi.org/10.1002/art.37754
    » https://doi.org/10.1002/art.37754
  • 49. Cherin P, Belizna C, Cartry O, Lascu-Dubos G, de Jaeger C, Delain JC, et al. Long-term subcutaneous immunoglobulin use in inflammatory myopathies: a retrospective review of 19 cases. Autoimmun Rev. 2016 Mar;15(3):281-6. https://doi.org/10.1016/j.autrev.2015.12.003
    » https://doi.org/10.1016/j.autrev.2015.12.003
  • 50. Alexanderson H, Lundberg IE. Exercise as a therapeutic modality in patients with idiopathic inflammatory myopathies. Curr Opin Rheumatol. 2012 Mar;24(2):201-7. https://doi.org/10.1097/BOR.0b013e32834f19f5
    » https://doi.org/10.1097/BOR.0b013e32834f19f5
  • 51. Alexanderson H, Munters LA, Dastmalchi M, Loell I, Heimbürger M, Opava CH, et al. Resistive home exercise in patients with recent-onset polymyositis and dermatomyositis - a randomized controlled single-blinded study with a 2-year followup. J Rheumatol. 2014 Jun;41(6):1124-32. https://doi.org/10.3899/jrheum.131145
    » https://doi.org/10.3899/jrheum.131145

Publication Dates

  • Publication in this collection
    12 Aug 2022
  • Date of issue
    May 2022

History

  • Received
    29 Mar 2022
  • Accepted
    29 Apr 2022
location_on
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org
rss_feed Acompanhe os números deste periódico no seu leitor de RSS
Acessibilidade / Reportar erro