You are on page 1of 16

REVIEWS

I­nclusion body myositis: clinical


features and pathogenesis
Steven A. Greenberg1,2,3
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.

Inclusion body myositis (IBM) is a slowly progressive and macrophagic myofasciitis). Further subdivision of
skeletal muscle disease with unique clinical and patho­ dermatomyositis into distinct syndromes associated
logical features (including finger flexor and quadriceps with specific autoantibodies is currently an active area
weakness and the presence of infiltrating cytotoxic of research11.
T cells in muscle). In patients with IBM, the onset of Many excellent general IBM reviews have been
symptoms of muscle weakness typically occurs between ­published over the past 5 years12–19, and some have
45 and 70 years of age, and progression is insidious. The focused specifically on the pathogenesis20,21. This Review
combination of slow progression towards disability and discusses IBM generally, with a focus on some of the
the lack of any reliable treatment to reverse the disease more confusing areas around nomenclature and clini­
course results in a substantial unmet medical need. cal features, and addresses key questions regarding
IBM lies within the category of inflammatory myop­ its pathogenesis.
athies. This category started with the single member,
polymyositis, in 1887 (refs1,2) and has since undergone History of IBM
repeated subdivision (Fig. 1). Dermatomyositis was A historical perspective provides some insight into what
­recognized as a separate entity early in 1891 (refs3,4), has shaped our current understanding of IBM (Fig. 1). It
1
Department of Neurology,
Brigham and Women’s
but it took almost a century before the next refine­ was first viewed as a form of polymyositis resulting from
Hospital, Boston, MA, USA. ment, IBM, was adequately recognized in 1978 (ref.5). chronic viral infection of muscle in 1967 (ref.22), leading
2
Children’s Hospital Immune-​mediated necrotizing myopathy (IMNM) to an ongoing line of research directed towards the iden­
Computational Health was distinguished in 1991 (ref.6). Advances continue to tification of a persistent muscle viral infection23–26. The
Informatics Program, Boston reduce the remaining category of polymyositis7, splitting name inclusion body myositis is derived from a 1971
Children’s Hospital, Boston,
off non-​specific myositis8 (or unclassified myositis9) and single case report entitled ‘Inclusion body myositis’27 that
MA, USA.
virtually eliminating the term polymyositis from what described a 26-year-​old patient with a limb-​girdle pat­
3
Harvard Medical School,
Boston, MA, USA.
once constituted the entire category of inflammatory tern of weakness and quadriceps sparing with onset at
myopathies. The generally accepted scheme currently age 18 years (which is not the disease we now call IBM).
e-​mail: sagreenberg@
bwh.harvard.edu consists of five members (polymyositis, dermato­ The first IBM clinical case series was published in
https://doi.org/10.1038/ myositis, IBM, IMNM and non-​specific myositis)8,10 1978 and described patients with a distinct form of
s41584-019-0186-x plus other rare entities (such as granulomatous myositis inflammatory myopathy characterized by prominent

Nature Reviews | Rheumatology


Reviews

Key points mitochondrial abnormalities in muscle biopsy samples


of patients with IBM82, both of which seem to be second­
• Inclusion body myositis (IBM) progresses slowly and is commonly misdiagnosed ary to autoimmunity83–85; the recognition of the genetic
initially as arthritis or polymyositis; IBM is associated with cardiovascular linkage of IBM to the HLA region (like other auto­
complications and other autoimmune diseases and has a high economic cost. immune diseases)86, which was confirmed by many sub­
• IBM has unique physical examination features (such as finger flexor and knee sequent studies42,87–93 including whole-​genome studies94;
extensor weakness) that distinguish it from most other muscle diseases. the first blinded placebo-​controlled IBM therapeutic
• IBM has a greater range of autoimmune T cell abnormalities than any other muscle clinical trial95; studies of evidence that rimmed vacuoles
disease; treatment refractoriness has paradoxically given rise to the view that IBM is are derived from myonuclei96 and their lack of necessity
not an autoimmune disease.
as biomarkers in the diagnosis of IBM9,97; and findings
• Degenerative abnormalities that can occur in IBM include numerous myofibre protein demonstrating the apparent ability of some sera from
aggregates associated with endoplasmic reticulum stress.
patients with IBM to induce muscle protein aggregation
• Degenerative abnormalities might occur following autoimmunity in cell culture and in passive transfer in vitro and in vivo animal models98.
mouse models and following immune cell dysfunction in patients infected with HIV
or human T cell lymphotropic virus type 1.
Nomenclature
• Treatment refractoriness probably reflects the inability of current therapies to inhibit
Several similar abbreviations of other diseases have cre­
or deplete the highly differentiated population of effector memory and terminally
differentiated effector T cells present in IBM.
ated confusion regarding IBM. IBM is an abbreviation
for ‘inclusion body myositis’ not ‘inclusion body myop­
athy’. The abbreviation IBM refers unambiguously to
quadriceps weakness and distal weakness (not specified the single disease IBM, not to a collection of disparate
as the wrist and finger flexor involvement now associ­ diseases that share the microscopic histological feature
ated with the disease)5; in hindsight, the authors mis­ of rimmed vacuoles. The rimmed vacuole myopathies
takenly used the term IBM for these patients, despite include IBM but also a set of genetic non-​immune dis­
the differences in phenotype between these patients orders that have been arbitrarily split off into heredi­
and the one in the original 1971 case report. Numerous tary inclusion body myopathies (hIBMs) (for example,
retrospective case series studies followed in the 1980s disorders due to mutations in GNE, VCP or MYH2),
and 1990s, emphasizing the unique distribution of other inherited disorders that are not viewed as forms
IBM muscle weakness particularly in finger flexors, of hIBM (such as oculopharyngeal muscular dystrophy
quadriceps and ankle dorsiflexors28–37. The past two due to PABPN1 mutations) and yet others that have
decades have seen the publication of large amounts of been classified as hIBM by some authors but not others
cross-​sectional data38–40 addressing the clinical features (for example, myofibrillar myopathy due to desmin
of IBM, including detailed views of regional muscle mutations, formerly called IBM1 in OMIM99). The
involvement38,41,42. Some longitudinal data are currently hIBM diseases lack the two main characteristic features
available, such as paired measurements of outcome of IBM: its distinctive pattern of muscle weakness and
measures in 51 patients 1 year apart and in 13 patients the presence of muscle histological immune cell infil­
4 years apart41,43–47, but more data are needed. tration. Many publications have used the abbreviation
A crucial discovery regarding IBM pathogenesis came IBM to refer to both IBM and hIBM; the interpretation
around 1984–1988 with the identification of cytotoxic of laboratory models of hIBM as relevant to patients with
CD8+ T cells invading myofibres of patients with IBM IBM is a mistake arising from this practice100.
through the application of a novel technology (mono­ The term sporadic IBM (sIBM) was introduced to
clonal antibody immunohistochemistry)48–51. These try and avoid confusion with hIBM by emphasizing its
observations launched an ongoing line of research into sporadic, or non-​inherited, occurrence. However, the use
IBM T cell clonality52–61, T cell differentiation phenotype of this term might give the mistaken impression that the
(including the relationship to large granular lymphocytic diseases sIBM and hIBM have similar clinical and patho­
leukaemia (LGLL)62–65) and therapies targeting T cells66–68. logical features, representing the same pathophysiological
Although the identification of unique circulating process that can either occur sporadically or be inherited.
autoantibodies in polymyositis and dermatomyositis69 This misconception could impede IBM research.
began in 1976 (refs70,71) and is still ongoing11,72, the rec­ Familial IBM (fIBM) refers to the occurrence of typi­
ognition that autoantibodies were also present in IBM cal IBM within families, almost always within a single
was greatly delayed until 2011. The identification of generation of siblings101–103. This pattern is analogous
­molecular signatures associated with B cells and plasma to the familial occurrence of many other autoimmune
cells in IBM muscle73 and the identification and char­ diseases, such as myasthenia gravis and multiple scle­
acterization of intramuscular plasma cells74, including rosis, and probably results from the associated genetic
clonally related plasma cells (the presence of which risk from HLA variants present in IBM and common to
implicates an underlying antigen-​driven process)75–77, in many autoimmune diseases.
IBM then led to searches for circulating blood autoanti­
bodies78 and the discovery of the target antigen of these Clinical features of IBM
antibodies, cytosolic 5′-nucleotidase 1A (cN1A; encoded Epidemiology and economic costs
by NT5C1A)79,80. Calculations of the prevalence of IBM are probably
Further key events in IBM history, which are dis­ underestimates because of the high rate of misdiag­
cussed in more detail later, include the following: the nosis. In a rigorous meta-​analysis published in 2017, the
identification of myofibre protein aggregates81 and pooled prevalence of IBM was 46 patients per million104.

www.nature.com/nrrheum
Reviews

Identification of the molecular target (cN1A;


NT5C1A) of the IBM circulating autoantibody
Muscle protein aggregation is secondary to
immune system MHC I upregulation in
murine models

First clinical and Recognition of CD28+ (CD244–) highly


pathological case mitochondrial differentiated cytotoxic
series description abnormalities CD8+ T cell identified
Genome-wide studies
Identification of Congo Identification of B cell confirm linkage to
First description red protein aggregates role using microarray common autoimmune
of IBM pathology in 0.5% of myofibres technology disease haplotype

1887 1891 1967 1971 1978 1984 1991 1992 1993 1997 2002 2008 2010 2011 2013 2016 2017 2018

Identification of Recognition that rimmed Recognition of highly


cytotoxic CD8+ T cells vacuoles are commonly differentiated effector
using immunohisto- absent in IBM muscle and effector memory
chemistry CD8+ T cells in IBM
muscle and blood
Name Origin of First blinded overlapping with T-LGLL
‘IBM’ used β-amyloid placebo
in case theory controlled Identification of a
report trial (IVIG) IBM-associated
circulating autoantibody

1887 1891 1978 1991 2018

DM DM DM DM
PM
IBM IBM IBM
PM
PM IMNM IMNM
PM Non-specific
PM

Fig. 1 | evolution of inflammatory myopathy classification and history of IBm. The upper timeline shows the key events
in the history of inclusion body myositis (IBM). The lower timeline shows the progressive subdivision of the category
polymyositis (PM) over time and current ongoing subcategorization of dermatomyositis (DM). IMNM, immune-​mediated
necrotizing myopathy; IVIG, intravenous immunoglobulin; T-​LGLL, T cell large granular lymphocytic leukaemia.

The results of this study suggest a sharp increase in to estimate US health-​care costs for patients with IBM and
IBM prevalence over the past decade, probably owing Medicare coverage suggests annual costs of 35,000 US dol­
to improved ascertainment105, with the first published lars per year in excess of the costs of matched controls and
estimate of IBM in 2000 (from a nationwide study in a prevalence of 84 per million in people >65 years of age114.
the Netherlands) being 5 per million106 and the most
recent estimate in 2017 (from a population-​based study Clinical features
in Ireland) being 112 per million107. IBM typically presents in middle or late age with slowly
Typical estimates of the mean age of IBM symp­ progressive, painless difficulty walking or using the
tom onset range from 61 to 68 years39,105,108–110. IBM is hands. Walking difficulties typically result from knee
often viewed as affecting people >50 years old; however, buckling, owing to knee extensor weakness, or tripping
a substantial proportion (20%38) of patients develop owing to ankle dorsiflexion weakness. Grip impairment
symptoms in their forties. The reported male-​to-female results from finger flexor weakness. Symptoms are often
gender ratio varies widely from 0.5 (ref.111) to 6.5 (ref.35) initially attributed to age or arthritis; when a neuromus­
(mean of 1.6). IBM is initially misdiagnosed as another cular disease is recognized, a diagnosis of polymyositis
condition in 40–53% of patients42,110,112, and the mean or, less often, motor neuron disease is more typical than
duration from symptom onset to correct diagnosis is an immediate recognition of IBM. Muscle pain is very
4.6–5.8 years42,109,110,112,113. uncommon in IBM42,97.
Before 2010, no International Classification of Dysphagia (difficult swallowing) is an underestimated
Diseases, Ninth Revision, Clinical Modification (ICD- component of IBM115 and is under-​reported as a present­
9CM) code existed for IBM, making estimates of the ing symptom42,116 but is commonly present if sought by
prevalence and health-​care costs from US medical pay­ history or radiological studies116,117. Dysphagia becomes
ment databases impossible. In 2018, the use of this code more evident as the disease progresses and might

Nature Reviews | Rheumatology


Reviews

a b

c d e Normal

IBM

Fig. 2 | IBm physical examination and imaging features. a,b | Weakness of finger flexion in patients attempting hand
grip with partial involvement (part a) and end-​stage (part b) inclusion body myositis (IBM). Relative preservation of
lumbricals enables flexion at metacarpophalangeal but not interphalangeal joints. Subtle finger flexion weakness is often
present at earlier stages of disease. c | Ventral forearm flexor muscle atrophy (arrows). d | Medial thigh atrophy (arrows).
e | Thigh axial proton density fast spin echo MRI, with abnormal muscle signal due to fibrosis, highlighted in vastus medialis
(arrows). Panels a–d reprinted with permission from the Inclusion Body Myositis Foundation.

result in nutritional deficiency, weight loss and aspira­ two (index finger)36; preservation of the adductor ­pollicis
tion pneumonia, which is a major cause of mortality in (the muscle that controls opposition of the thumb to
IBM40,43,118. Cricopharyngeal sphincter dysfunction can index finger in the plane of the hand); and preservation
be present116, and cricopharyngeal muscle biopsy sam­ of hip abduction42 and adduction even in patients with
ples might show similar histological features to those complete loss of knee extensor muscles. Complete paraly­
of limb muscle biopsy samples119,120. Asymptomatic sis of all deep and superficial finger flexors with preser­
respiratory function impairment and sleep-​disordered vation of the adductor pollicis and lumbricals (muscles
breathing are common121. of the hands that flex the metacarpophalangeal joints)38
IBM has unique physical examination features whose results in the common end-​stage IBM hand appear­
lack of recognition largely contributes to the high initial ance, allowing patients to grasp objects through thumb
misdiagnosis rate (Fig. 2a–e). The value of these p ­ hysical adduction and finger flexion at the metacarpophalan­
examination features is sufficiently high that, when prop­ geal joints (Fig. 2b). Ventral forearm atrophy (Fig. 2c),
erly appreciated, the additional value of muscle histo­ although striking in later disease stages, is often hard to
pathology for a diagnosis of IBM is questioned122. There recognize during the early stages of disease. Knee exten­
is preferential involvement of muscles controlling finger sor weakness often requires functional testing, such as
flexion, knee extension and ankle dorsiflexion with less deep knee bends, hopping on one leg or climbing stairs,
involvement of the muscles that are typically affected in to detect. Among the quadriceps, the vastus medialis
other acquired myopathies, such as those controlling (and vastus lateralis) is affected considerably sooner
proximal shoulder abduction, hip flexion and hip abduc­ than the rectus femoris (Fig. 2d); thus, direct palpation
tion. This pattern of IBM is often referred to as distal, but of the medial thigh during attempted knee extension
this term might be confusing because truly distal hand against resistance is an extremely helpful technique to
muscles, such as the intrinsic muscles mediating finger detect its involvement.
abduction and adduction, are relatively spared in IBM. Beyond physical examination, MRI can reveal charac­
A number of helpful physical examination findings teristic regional muscle involvement37,123–128. For example,
greatly facilitate the diagnosis of IBM (Fig. 3). These find­ in patients with remarkable quadriceps muscle atrophy
ings include the following: orbicularis oculi weakness but only mild knee extensor weakness owing largely to
(41% in one series)38; greater weakness of elbow flex­ the relative preservation of the rectus femoris in the early
ion (controlled by the biceps) than shoulder abduction stages of disease, muscle loss is still detectable by MRI
(controlled by the deltoids); greater weakness of flexor (Fig. 2e). Ultrasonography might show similar regional
digitorum profundi than flexor digitorum superficialis muscle involvement129–132. Videofluoroscopy and real-​
(the muscles of the forearm that control finger flexing) time MRI frequently show abnormalities related to
and greater weakness of digit five (little finger) than digit dysphagia115,116,118,133.

www.nature.com/nrrheum
Reviews

Front view Back view probably the result of the varying methods of testing that
have been used. Thus, a negative test should not be used
to diminish confidence in the diagnosis of IBM. More
controversial is the prevalence of anti-​cN1A antibodies
Shoulder in non-​neuromuscular autoimmune diseases, which will
abduction
affect the diagnostic specificity of such assays for IBM.
Elbow Elbow Different laboratories have found a widely varying preva­
flexion extension lence of these antibodies in systemic lupus erythematosus
Wrist (0–20%) and Sjögren syndrome (0–36%)98,137,138,140–142.
flexion
Finger
flexion Hip abduction Microscopic pathology
and extension The standard microscopic pathological findings in IBM
are endomysial lymphocytic infiltration (Fig. 4a), with
Hip lymphocytes surrounding and occasionally invading
Knee
flexion
extension myofibres (Fig. 4a,b). When phenotype is determined
by immunohistochemistry, most lymphocytes are
Ankle CD8+ T cells (Fig. 4c). A small number of myofibres
dorsiflexion contain rimmed vacuoles (0.4–6.4%) 81,96,143 (Fig. 4d)
and mitochondrial abnormalities, such as the pres­
ence of COX−SDH+ and ragged red myofibres (Fig. 4e).
Haematoxylin and eosin staining and trichrome stain­
ing show variation in fibre size, centralized nuclei and
type 2 fibre atrophy and, occasionally, cytoplasmic
Muscles preferentially affected in IBM inclusions. Electron microscopy or light microscopic
Muscles preferentially affected in other inflammatory myopathies examination of glutaraldehyde-​fixed, semithin sections
Muscles commonly affected in both
often shows cytomembranous whorls and, less com­
monly, tubulofilaments, within 2–6% of myonuclei144.
Fig. 3 | Pattern of muscle involvement in IBm and other inflammatory myopathies.
Weakness of finger flexion, knee extension and ankle dorsiflexion is characteristic of
Immunohistochemical stains for MHC class I and MHC
inclusion body myositis (IBM) with less involvement of muscles typically affected in other class II show diffuse myofibre surface staining and,
acquired myopathies, such as proximal shoulder abduction, hip flexion and hip abduction. sometimes, punctate cytoplasmic staining, but MHC
class I staining has been reported to be highly non-​
specific (present in 93% of biopsy samples from patients
Abnormal laboratory test results typically include with non-​inflammatory myopathies), with MHC class II
modest increases in serum levels of creatine phospho­ staining being more specific to inflammatory myopa­
kinase in most patients and a monoclonal immuno­ thies145. Other special stains can demonstrate protein
globulin population (by serum immunofixation) in aggregates (such as TDP43, Fig. 4 f).
~15% of patients97,134. Antinuclear antibodies, anti-​Ro A pathological diagnosis of IBM is often not made
or anti-​La antibodies, and rheumatoid factor positiv­ without the presence of rimmed vacuoles, although
ity might also be evident97. Testing for anti-​cN1A (also they are absent in 20% of patients with typical clinical
called anti-​NT5C1A) autoantibodies is helpful. Testing features97,146. The clinical course for patients with and
for HIV and human T cell lymphotropic virus type 1 without rimmed vacuoles seems to be similar97, but one
(HTLV-1) might be considered in particular circum­ study has suggested a more aggressive course in patients
stances, such as young age of IBM onset or the pres­ with rimmed vacuoles147. Some authors have preferred
ence of factors that suggest an underlying increased to split IBM into two categories, classifying patients
risk of being infected with HIV or HTLV-1. Additional without rimmed vacuoles as having polymyositis with
blood b­ iomarkers of highly differentiated CD8+ T cells mitochondrial pathology (PM-​Mito), and view IBM and
(CD8 +CD57 + T cells by flow cytometry, a reduced PM-​Mito as part of a broader category of inflammatory
CD4:CD8 blood ratio and lymphocytosis evident myopathy with vacuoles, aggregates and mitochondrial
on complete blood count differential)64 are the most pathology (IM-​VAMP)148. The observation that patients
­common of all blood abnormalities in IBM but await with typical treatment-​responsive polymyositis might
further studies of their clinical utility. also have rimmed vacuoles has been little emphasized149.

Anti-​cN1A autoantibody Diagnostic criteria


Serum anti-​cN1A antibodies are highly specific to IBM Various diagnostic criteria for IBM were proposed
among the neuromuscular diseases and are seen in from 1987 to 2002 through individual author opin­
90–95% of patients with IBM compared with 5–10% ion31,150–152 and from 1995 to 2013 by consensus expert
of patients with polymyositis, dermatomyositis or non-​ opinion106,153–157. Criteria based on expert opinion have
immune neuromuscular diseases79,80,135–138. Accordingly, been standard in the field, although studies evaluating
a positive test result in a patient with a muscle disease is their performance were lacking before 2013. Subsequent
highly predictive of IBM. However, the diagnostic sen­ studies have disclosed major limitations in the diagnos­
sitivity of anti-​cN1A antibody assays for IBM has varied tic sensitivity of these criteria, with many patients with
widely79,80,135–140, ranging from 37%140 to 76%135, and is IBM failing to meet them122,158. In particular, the criteria

Nature Reviews | Rheumatology


Reviews

a b c

20 µm 20 µm 20 µm

d e f

1
2

20 µm 20 µm 10 µm

Fig. 4 | IBm muscle pathology. a | Infiltration of muscle endomysium by immune cells (arrow) visualized by haematoxylin
and eosin staining. b | Deep invasion of non-​necrotic myofibre by immune cells (arrow) visualized by haematoxylin and eosin
staining. c | Many invading immune cells are CD8+ cytotoxic T cells (arrow), as visualized by staining for CD8. d | Rimmed
vacuoles (arrows) in trichrome stained sections. e | Ragged red fibre (a mitochondrial abnormality) (arrow) visualized by
haematoxylin and eosin staining. f | TDP43 sarcoplasmic aggregates (red and arrow) visualized by immunofluorescence.
The myofibre labelled 1 contains numerous sarcoplasmic aggregates; the myofibre labelled 2 has normal TDP43 localization
to the myonuclei (blue). IBM, inclusion body myositis.

for the detection of rimmed vacuoles by microscopy, Medicare database study reported a higher preva­
often referred to as biopsy-​proven IBM, can be restric­ lence of comorbidities in patients with IBM than in
tive9,122,158. Criteria with sensitivity of 90% and speci­ matched patients without IBM114, with increased rates
ficity of 96% were derived through machine learning of hypertension (66% versus 22%), hyperlipidaemia
approaches; these criteria included the presence of the (47% versus 18%), diabetes mellitus (34% versus 10%),
following: finger flexor or quadriceps weakness, biopsy myocardial infarction (13% versus 2%), congestive
sample demonstrating endomysial inflammation and heart failure (17% versus 5%), pneumonia (11% ver­
biopsy sample demonstrating invasion by T cells of non-​ sus 2%; aspiration pneumonia, 6% versus 0.3%) and
necrotic muscle fibres or rimmed vacuoles158. The use of anaemia (32% versus 7%). These IBM comorbidities
blood testing for anti-​cN1A autoantibodies has not yet do not seem to be a consequence of complications
been incorporated into diagnostic criteria. of ­immunosuppressant therapy, such as corticoste­
roids114, and are probably instead related to the systemic
Associated disorders and comorbidities inflammatory environment resulting from autoimmun­
IBM is associated with another autoimmune disease ity, as in other forms of inflammatory myopathy168–173
in 13–24% of patients97,109,134, most commonly Sjögren and other autoimmune diseases174,175.
syndrome (10–12%)91,109,159–161. A surprising number of
patients with IBM (58% in one series) meet the diagno­ Progression
stic criteria for LGLL, and a small number have overt IBM is a progressive disease, with mean or median loss
leukaemia with cytopenias64. of device-​free ambulation from symptom onset esti­
IBM might develop from HIV162–165 and HTLV-1 mated at 7.5–10 years for a cane42,44 and 13–15 years
(refs 164,166) infection, resulting in a typical disease for a wheelchair38,39,44. Hand impairment and dyspha­
course except for a younger age of onset. HIV-​associated gia ­progression are less well quantified. Published
­myositis has been categorized as either polymyositis longitudinal data regarding IBM progression are insuf­
or IBM. Patients with HIV-​associated myositis might ficient41,43–47, and non-​uniform methods have been used
show improved strength of proximal muscles as a result to measure it. Furthermore, published annualized rates
of treatment162,163, but in a study of 11 patients with of progression of 4–28% per year41,43–47,53 are specific to
HIV-​associated myositis, all subsequently developed the outcome measure used, such as quantitative muscle
treatment-​refractory IBM162. testing, and assume linearity over time. Some patients
The prevalence of some comorbidities, such as experience very little or no disease progression over
cardiovascular disease and diabetes mellitus, in IBM periods of time ranging from 4 years45 to 12 years43.
seemed to be high in a number of population-​based Although IBM has been characterized as not fatal
cohort and case series studies that lacked comparator because statistical analyses of IBM populations have not
groups (for example, 65% of patients had hypertension detected a reduction in life expectancy39, it is a cause
and 24% had diabetes mellitus in one study population of premature mortality in some patients, most directly
of patients with IBM)109,167,168. A matched case–control from aspiration pneumonia40,43,118.

www.nature.com/nrrheum
Reviews

IBM therapeutics one showed directional trends towards minor benefit in


Standard of care for most patients with IBM involves limb and swallowing muscles, with some statistical effi­
strictly nonpharmacological management, includ­ cacy95, and the other two (one that included prednisone,
ing emotional support, physical therapy, education which might have been counterproductive) showed no
on fall precautions and exercise176–178, and dyspha­ directional trend or statistical benefit in limb or swallow­
gia evaluation. Dysphagia can be treated transiently ing muscles185,186. For such a slowly progressive disease,
with ­pharyngoesophageal dilatation or cricopharyn­ 3–6 months of therapy seems insufficient for a clinical
geal myotomy118. Few physicians routinely prescribe trial efficacy end point. Preliminary results of a trial of
immunosuppressant therapies, with the belief that such rapamycin have been reported in an abstract68.
­therapies result in transient responses at best14,38,97. Trials of two muscle-​building therapies (oxandro­
lone and bimagrumab), neither of which addresses the
Expert opinion and open-​label trials specific mechanism of myofibre injury, have shown
The use of corticosteroids in IBM derives from expert some statistical benefits. Oxandrolone190 showed statis­
opinion. Corticosteroids decrease serum levels of cre­ tical efficacy for multiple end points. In a small study
atine phosphokinase, which is sometimes mistaken for (n = 14)191, bimagrumab showed improvement in skeletal
a clinical response; however, the disease continues to muscle mass. In a registration trial (n = 240)192 presented
progress in almost all patients32. Some patients with in abstract, bimagrumab again showed improvement in
substantial proximal weakness, an atypical phenotype, whole-​b ody skeletal muscle mass and in a patient-​
do experience improvement in proximal strength32. reported secondary outcome measure, but no statis­
A view that corticosteroids might even worsen IBM tical efficacy in the primary outcome measure (the
has been suggested on the basis of retrospective cross-​ 6-minute walk distance). A trial of arimoclomol did not
sectional data in which there were more patients with achieve proof of concept of the hypothesized mechanis­
severe disease in a treated cohort than there were in tic effect (increase in muscle levels of HSP70) or show
an untreated cohort. However, this view is compli­ clinical efficacy100.
cated by an uncontrolled confounder — the treated
cohort also had substantially longer duration of disease Pathogenesis of IBM
diagnosis than the untreated cohort (50 months versus No feature of IBM has generated more debate and
18 months)39. The marked type 2 muscle fibre atrophy ­speculation than the nature of its pathogenesis. IBM is
present in IBM179 is expected to worsen with cortico­ often viewed as both a degenerative and an autoimmune
steroid treatment and is another theoretical argument disease. A number of perspectives have shaped these
against its use. views and are addressed here.
Broad lymphocyte depletion strategies achieved
­statistical efficacy for strength measurements in two Degenerative biomarkers and mechanisms
open-​label comparator group studies: antithymocyte Various IBM muscle histochemical abnormalities have
globulin (ATG) with methotrexate compared with collectively been called degenerative (Fig. 5). However,
methotrexate alone (P = 0.02)66 and alemtuzumab67 referring to IBM as a degenerative disease does not pro­
compared with baseline (pretreatment; P < 0.002). vide information on the mechanisms by which the mus­
Intravenous immunoglobulin (IVIG) or subcuta­ cle is injured. The degenerative features of IBM include
neous immunoglobulin (SCIG) has been reported to rimmed vacuoles and the related myonuclear degener­
be helpful specifically for patients with IBM-​associated ation5,193, mitochondrial pathology82,194,195 and myofibre
dysphagia180–182. Results of an open-​label trial of threecytoplasmic protein aggregates.
combined interventions (follistatin gene therapy, pred­ The concept of protein aggregation has dominated
nisone and exercise) were published as an interim mechanistic thought since the early 1990s, with inter­
analysis; somewhat c­ ontroversially, the efficacy was est initially focused on four muscle biomarkers (amy­
attributed to follistatin gene therapy alone despite beingloid detected by Congo red81, ubiquitin196, β-​amyloid197
tested only in combination183,184. An open-​label trial of and tau198). The number of distinct molecules reported
exercise alone has demonstrated statistical efficacy for as aggregated grew to >80 by 2010 (ref. 199) . A the­
strength measurements178. ory of β-​amyloid accumulation and myotoxicity has
­dominated144,200–202 and continues to be featured in con­
Blinded placebo-​controlled trials ceptual models of IBM10. In hindsight, a published belief
Although the literature is filled with the view that IBM in this theory now seems to be out of proportion to the
is treatment refractory, high-​quality data on therapy for data supporting it. Indeed, citation distortions in its cita­
IBM are very limited, with published blinded placebo-​ tion network might have contributed to overvaluing the
controlled clinical trials for only five FDA-​approved role of β-​amyloid deposition in IBM pathogenesis203,204.
therapeutics (IVIG with and without prednisone95,185,186, The supporting data were based on methods (immuno­
methotrexate187, IFNβ188,189 and oxandrolone190) and two histochemistry) and reagents (anti-​β-amyloid antibodies)
investigational therapeutic candidates (bimagrumab191 that do not distinguish β-​amyloid from its precursor pro­
and arimoclomol100) (Table 1). tein203, the latter reported as non-​specifically expressed in
Oral methotrexate, 5–20 mg daily for 1 year, showed regenerating myofibres in all muscle diseases205. Similar
a directional trend towards benefit but no statistical interest in tau protein deposits (using SMI-31 immuno­
efficacy187. Dysphagia was not assessed in this trial. reactivity) also occurred206 and has similar limitations207.
Of three trials of IVIG of 3–6 months treatment duration, Focus on these biomarkers proceeded for decades despite

Nature Reviews | Rheumatology


Reviews

studies demonstrating their rarity or non-​existence in immunoreactivity. For example, one comparative study
IBM muscle (for example, the presence of Congo red found TDP43 aggregates in 23% of IBM myofibres
amyloid in 0.4% of myofibres208, ubiquitin in 0.7% of compared with Congo red amyloid in 0.6%, SMI-31 in
myofibres209 and β-​amyloid in 0% of myofibres210). 0.8% and β-​amyloid in 0%218. Another study found
A second generation of muscle histochemical degen­ both p62 and TDP43 aggregates in 12% of IBM myo­
erative biomarkers (p62, LC3 and TDP43 (refs211,212)) has fibres compared with ubiquitin in 1.1% and β-​amyloid
evolved around the unfolded protein response and endo­ in 0.1%211. p62 and TPD43 aggregates are rarely seen in
plasmic reticulum (ER) stress213, altered autophagy214,215 non-​IBM autoimmune muscle disease, suggesting that
and mislocalized myonuclear heterogeneous nuclear these second-​generation biomarkers have some value in
ribonucleoproteins (hnRNPs)216–218. Some of these the study of IBM.
studies have combined immunohistochemical results
with western blots (TDP43 (refs217,218) and p62 (ref.219)). Autoimmune biomarkers and mechanisms
Furthermore, results have been reproduced by multiple T cells. T cells, myeloid dendritic cells, macrophages and
independent laboratories, with quantitative data show­ plasma cells all invade IBM muscle (Fig. 5), but it is the
ing superior biomarker performance compared with infiltration of muscle by T cells that is the most obvi­
Congo red amyloid, ubiquitin, β-​amyloid or SMI-31 ous histological feature of IBM muscle. The invasion of

Table 1 | IBm clinical trials


therapeutic year of year of Number Duration of outcome measures: Refs
clinical trial publication of treatment primary (secondary)
registration patients (months)
Open-​label studies
Azathioprine + methotrexate + NA 1993 11 3–6 MMT 296

intravenous methotrexate
IVIG NA 1993 4 2 Muscle strength 297

1994 9 1–3 MMT 298

Prednisone NA 1995 8 6–12 Muscle strength 299

Antithymocyte NA 2003 11 12 QMT 66

globulin + methotrexate versus


methotrexate alone
Etanercept NA 2006 9 17 QMT 300

Anakinra NA 2013 4 5–12 MMT (Grip) 301

Alemtuzumab 2004 2009 13 12 QMT (MMT) 67,302

Lithium 2008 NA 20 6 QMT (MMT) 303

Follistatin gene 2012 2017 6 24 Safety (6MWD) 183,304

therapy + prednisone + exercise


Natalizumab 2013 NA 6 6 Safety (Biopsy) 305

Bimagrumab 2014 NA 14 40 Safety (DEXA) 306

Blinded placebo-​controlled studies


IVIG NA 1997 19 3 QMT (MMT) 95

2000 22 6 MMT (NSS) 185

IVIG + prednisone NA 2001 37 3 QMT (MMT) 186

Low-​dose IFNβ1a NA 2001 30 6 QMT (MMT) 188

High-​dose IFNβ1a NA 2004 30 6 QMT (MMT) 189

Oxandrolone NA 2002 19 6 QMT (MMT) 190

Methotrexate NA 2002 44 11 QMT (MMT) 187

Etanercept 2005 NA 20 12 QMT (MMT) 307

Arimoclomol 2008 2016 24 4 Safety (QMT) 100,308

Bimagrumab 2011 2014 14 6 MRI (QMT) 191,309

2013 NA 240 12 6MWD (sIFA) 310

Rapamycin 2015 NA 44 12 QMT (grip QMT) of 311

quadriceps
Arimoclomol 2016 NA 150 20 IBMFRS (MMT) 312

6MWD, 6-minute walking distance; DEXA , dual-​energy X-​ray absorptiometry ; IBMFRS, inclusion body myositis functional rating
scale; IVIG, intravenous immunoglobulin; MMT, manual muscle testing; NA , not applicable; NSS, neuromuscular symptom score;
QMT, quantitative muscle testing; sIFA , sIBM functional assessment.

www.nature.com/nrrheum
Reviews

of the effector memory T (TEM) cell and terminally differ­


Perimysium entiated effector memory T (TEMRA) cell populations and
Endomysium
Muscle fibre
are sometimes characterized as senescent or of having
Muscle
fascicle a senescence-​associated secretory phenotype221, terms
that obscure their aggressive cytokine secreting and cyto­
toxic capacities222–225. CD8+CD28− T cells in IBM blood
are major producers of IFNγ63. Highly d ­ ifferentiated
CD4+ T cells that have lost CD28 expression are no
longer helper T cells but instead have become cytotoxic
Infiltrating
T cells killer cells226,227, and levels of these cells are known to be
increased in other autoimmune diseases228–236. Highly dif­
Healthy IBM
ferentiated CD8+ T cells express high levels of cytotoxic
molecules (such as perforin and granzymes) and overlap
phenotypically and functionally with natural killer cells.
Protein
aggregate CD8+ T cell loss of CD5 expression and evolution into
Capillary natural-​killer-like T cells that express CD16 or CD56 are
common in IBM blood64 and suggest that, in IBM muscle,
Vacuole T cell cytotoxic injury to myofibres occurs independently
of antigen recognition through CD3 or costimulation
through CD28. These T cell phenotypic changes are also
seen in T cell LGLL (T-​LGLL), a treatment-​refractory
expansion of clonal highly differentiated effector
CD8+ T cells237.
Autoimmune features Degenerative features
Infiltrating cells • Rimmed vacuoles Plasma cells and autoantibodies. Circulating autoanti­
• T cells (mainly CD8+ cytotoxic T cells) • Myonuclear degeneration
• Plasma cells • Mitochondrial pathology bodies have been known to be present in polymyosi­
• Macrophages • Protein aggregates tis and dermatomyositis for >40 years69–71. However,
• Myeloid dendritic cells
an autoantibody in IBM was recognized only in 2011
(ref.78). This delay was probably related to the early

Fig. 5 | IBm muscle inflammation. Inclusion body myositis (IBM) shows features of an observations that, in IBM muscle, only sparse numbers
autoimmune disease with protein aggregates. Immune cells present in the endomysial of CD20+ endomysial B cells (0.5%) were present, com­
region surround and invade myofibres; infrequent myofibres contain rimmed vacuoles pared with the large numbers of T cells (75%)48, com­
(~1%) or p62 and TDP43 protein aggregates (~10%). bined with the popularity of the degenerative model of
IBM. The application of newly invented gene expression
microarray profiling to IBM muscle in 2002 provided
myofibres by cytotoxic T cells is easily visible by micros­ an opportunity for non-​hypothesis-driven research —
copy. IBM might be the only muscle disease involving to examine IBM muscle globally without bias. These
cytotoxic T cell-​mediated myofibre injury; although studies unexpectedly demonstrated robust production
polymyositis has been said to share this feature, con­ of immunoglobulin transcripts73, which could only be
siderable scepticism exists regarding this view9,220, in coming from intramuscular plasma cells74. The pau­
part because of the historical misdiagnosis of IBM as city of CD20+ B cells in IBM muscle seems to be the
polymyositis or the evolution of patients with partially result of an intense autoimmune environment causing
responsive polymyositis into treatment-​refractory IBM antigen-​driven transformation of CD20+ B cells into
over time. In IBM, there is an estimated fivefold greater CD19+ plasmablasts and clonal CD138+ plasma cells75–77.
preponderance of endomysial CD8+ T cells than CD4+ Recognition of this pathway facilitated the identifica­
T cells in muscle48–51. Numerous studies have reported tion of circulating blood ­autoantibodies78 and the target
clonal expansion of these T cells in the muscle and blood antigen of these autoantibodies, cN1A (NT5C1A)79,80.
of patients with IBM52–61, indirectly providing evidence
that T cells are targeting some unknown antigens, and Genetics. Strong genetic linkage of IBM has been estab­
studies of T cell phenotype have found that T cells in lished exclusively to immune gene variants, specifi­
both the muscle and blood of patients with IBM have cally to HLA-​DRB1*03:01 and HLA-​B*08:01, both of
expanded repeatedly and have been driven into a highly which are part of the common autoimmune disease 8.1
differentiated state62–65. ancestral MHC haplotype42,86–93, and to the chemokine
In IBM, CD4+ and CD8+ T cells are driven into highly receptor CCR5 gene. A variant in TOMM40, encoding a
differentiated effector cells in muscle62,64 and in blood62–64 mitochondrial protein, has been found to influence age
(Fig. 6). These highly differentiated populations of T cells of onset238,239 and perhaps disease risk238.
have been reported in IBM using various overlapping
markers, such as the loss of CD28 (CD28−; CD28 null) Chemokines and cytokines. IBM muscle is highly
on CD4+ and CD8+ T cells62,63, the loss of CD62L on enriched with many secreted immune chemokines and
CD45RA+ T cells63 and the gain of CD244 (ref.62), CD57 cytokines that are difficult to measure at the protein
(ref.64) and KLRG1 (S.A.G., unpublished observations) on level240,241 but whose transcripts can be detected through
CD8+ T cells. These subpopulations of cells are composed microarray experiments 73. The marked elevation

Nature Reviews | Rheumatology


Reviews

Blood Chronic stimulation of levels of CXCL9 and CCL5 (ref.73) in the muscle
strongly suggests upstream T cell activation and IFNγ
production. The expression of genes encoding members
of the IFNγ signalling cascade (the IFNγ signature) is
TCR CD28 higher in myofibres invaded by CD8+ T cells than in
Naive or non-​invaded myofibres242.
memory
T cells Is IBM an autoimmune disease?
IBM has sometimes been viewed as not being an auto­
CD4+ T cell CD8+ T cell
immune disease202, or the autoimmunity is viewed as
• Treatment refractory being a secondary phenomenon. Some reviews have
• Leukaemia-like phenotype classified it as a non-​immune myopathy or excluded
• Loss of CD28 expression
• Gain of expression of it from discussions of autoimmune myopathies243–245.
CD57, CD244 and KLRG1 Yet the magnitude of expression of numerous biomark­
ers of T cell autoimmunity is far higher in IBM muscle
than in any other muscle disease, including other forms
Clonal highly of myositis. These markers include an abundance of
differentiated T cells (particularly CD8+ cytotoxic T cells), the cytotoxic
cytotoxic
T cells molecules granzymes and perforin and an a­ bundance of
T cell-​related chemokines and cytokines.
The invasion of non-​necrotic muscle fibres by cyto­
Circulating T cells
toxic T cells has long been a diagnostic feature of IBM;
might lead to other invaded fibres were found to be approximately eight
autoimmune features times more common than myofibres containing amyloid
Endothelium
(detected by Congo red)208. By contrast, the harmful effect
of protein aggregates on myofibres is speculative. Over the
past 5 years, IBM has joined the ranks of other forms of
myositis with autoantibody biomarkers through the iden­
tification of a circulating IBM autoantibody (anti-​cN1A).
As we are now in the era of whole-​genome sequencing,
the lack of any causative gene mutation in patients with
IBM has virtually eliminated the possibility that it is an
IFNγ
unrecognized Mendelian genetic disorder and further­
more has disclosed strong linkage to the HLA region,
viewed by some as providing the strongest evidence to
date that autoimmunity is causative in IBM246.
↑ MHC
MHC class I
class I Evidence for causality in IBM
Although IBM degenerative and autoimmune biomark­
ers are associated, establishing causality will ultimately
require human clinical trials of mechanistically unam­
Myofibre
Cytotoxic biguously specific interventions. Currently, the evidence
granules suggests that causality flows from autoimmunity to
degeneration, not the reverse (Fig. 6).
• ER stress First, genetic diseases with biomarkers of degener­
• Protein aggregation Myofibre damage
ation do not result in autoimmunity. Although most of
Fig. 6 | Proposed pathogenesis of IBM: highly differentiated cytotoxic T cells drive the >80 proteins that aggregate in IBM muscle have also
IBm myofibre injury. Chronic antigenic stimulation of T cells results over time in been reported to aggregate in hIBM, these diseases do
transformation of naive or early effector memory T cells (TEM1 and TEM2 cells) into a highly not manifest autoimmunity. Specifically, TDP43, p62 and
differentiated population of clonal cytotoxic CD8+ T cells (TEM3, TEM4 and terminally ubiquitylated protein aggregates are prominent in biopsy
differentiated (TEMRA) cells), identified by CD8+CD28−, CD8+CD244+, CD8+CD57+ or samples from patients with genetic myotilinopathies and
CD8+KLRG1+) and CD4+ T cells (CD4+CD28−) present in inclusion body myositis (IBM) desminopathies247,248 and VCP-​mutation hIBM217 with­
muscle and blood. These late stage T cells have the following features: they are out autoimmunity. Vacuolar myopathy produced by
highly cytotoxic and have escaped requirements for costimulation (for example, loss colchicine or hydrochloroquine in patients is associated
of CD28); they invade myofibres and injure them through release of cytotoxic granules with ER stress and with p62 and LC3 aggregates249, but
(such as perforin and granzymes); and they produce abundant IFNγ, which results in
not autoimmunity. Some genetic muscular dystrophies,
upregulation of MHC class I on myofibres and causes endoplasmic reticulum (ER) stress
and aggregation of proteins (including p62 and TDP43). These highly differentiated particularly facioscapulohumeral muscular dystrophy
CD8+ T cells have similar features to those present in large granular lymphocytic and dysferlinopathies, have inflammation, but only
leukaemia, including resistance to corticosteroids. Furthermore, some patients with necrotic myofibres are invaded and only by macrophages
IBM have features of disease other than just muscle manifestations, such as (which are CD4+) and perhaps CD4+ T cells250,251. Many
autoimmune features in bone marrow (cytopenias) or epithelial secretory tissue other genetic disorders have mouse models demon­
(Sjögren syndrome). TCR, T cell receptor. strating impaired autophagy, with p62 and LC3 mouse

www.nature.com/nrrheum
Reviews

muscle aggregates without autoimmunity252. Similarly, criteria that classify all patients with IBM as having poly­
the strong correlation between IBM muscle mitochon­ myositis269, corticosteroids did not result in a reduction
drial patho­logy (number of COX-​deficient myofibres) of invading CD28− T cells in the muscle266. In retrospec­
and auto­immunity (number of invading T cells)85 sug­ tive comparisons, patients with IBM receiving long-​term
gests that the mitochondrial pathology results from immunosuppressive treatment (a mean of 35 mg daily of
autoimmunity, as muscle genetic mitochondrial myopa­ prednisone for a mean of 5.7 years) had the same number
thies do not produce autoimmunity. Dermatomyositis, of invaded myofibres as patients with IBM not receiving
which is also autoimmune, is similarly accompanied by treatment208. In prospective intervention trials, patients
mitochondrial abnormalities253,254. with IBM receiving corticosteroids or IVIG did not expe­
Second, intervention studies show that manipulat­ rience substantial reduction in the total muscle-​invading
ing the immune system can produce skeletal muscle T cell population54,186. Because of its close mechanistic
and other cellular protein aggregate biomarkers. Thus, pathogenesis, experience with T-​LGLL is informative
in vitro treatment of cultured human myoblasts with for IBM. The T-​LGLL population of highly differenti­
cytokines (IL-1β and IFNγ) produces protein aggre­ ated clonal T cells is resistant to apoptosis237,270 and is
gates83, treatment of various mouse cells with IFNγ not reduced following treatment with corticosteroids271
causes ubiquitylated inclusions255,256, and forced overex­ or alemtuzumab 272, the latter potentially owing to
pression of MHC class I in mouse muscle cells causes variable expression of CD52, its target, on these cells273.
ER stress257. Inflammatory stimuli ­(lipopolysaccharide) Two other immune therapies have undergone open-​
cause in vitro cytoplasmic mislocalization of TDP43 in label IBM clinical trials and have shown trends towards
microglia and astrocytes258 and p62 accumulation in efficacy: ATG66 and alemtuzumab67. ATG274 and alemtu­
macrophages259. In vivo upregulation of muscle MHC zumab275 rapidly deplete nearly 100% of blood CD4+ and
class I (which is highly induced by IFNγ) alone is suffi­ CD8+ T cells, but the subsequent immune reconstitution
cient in mouse models to result in ER stress and a severe results in paradoxical expansion above pretreatment
myopathy with rimmed vacuoles84. Third, a single study levels (known as homeostatic proliferation276) of highly
found that purified serum blood immunoglobulin frac­ differentiated pathogenic TEM and TEMRA cells, identified
tions from patients positive for anti-​cN1A with IBM as CD45RA−CD62L− T cells267, CD28−CD57+ T cells277
cause muscle degenerative p62 protein aggregation in and CD45RO−CD62L− T cells278. Furthermore, T regu­
cell culture and mouse models, although only in a small latory cells, which are potentially beneficial to con­
proportion of muscle cells or fibres98. trolling autoimmune disease, are decreased in number
Finally, proof that causality flows from autoimmun­ compared with pretreatment levels after alemtuzumab
ity to degenerative muscle pathology in people comes immune reconstitution278.
from the existence of the clinical syndromes of HIV-​ Finally, other autoimmune diseases are known to be
associated IBM and HTLV-1-associated IBM162–164,166,260. treatment refractory, including Sjögren syndrome and
HIV and HTLV-1 infect T cells; their antigens are primary biliary cholangitis (PBC)279–282. Like IBM, both
present in IBM endomysial T cells and macrophages diseases involve CD8+ T cell-​mediated damage (in exo­
but not myofibres164,166,260. These syndromes demon­ crine epithelial cells in Sjögren syndrome283 and bile duct
strate that an external event that alters the function of cholangiocytes in PBC284–286), progress slowly and have
the immune system can produce the full clinical and cytotoxic T cell genomic signatures and expansions of
pathological picture of IBM including the degenera­ the highly differentiated CD8+ TEMRA population287,288.
tive biomarkers of rimmed vacuoles and p62, LC3 and
TDP43 cytoplasmic aggregates163. Protein aggregates in autoimmune diseases
A role for ER stress and autophagy has been identified
Why is IBM treatment refractory? in other autoimmune diseases289–291. Two main histolog­
The refractoriness of IBM might suggest that it is a ical markers of IBM that are argued to indicate degen­
non-​autoimmune degenerative disease. However, treat­ eration, p62 (refs211,212) and LC3 (ref.212), are aggregated
ment refractoriness might instead suggest an autoim­ in cholangiocytes in PBC292. Other markers of ER stress
mune process in which the immune system has escaped and autophagy are also aggregated in PBC cholangio­
from typical checkpoints designed to restrain it, such cytes293,294 and Sjögren syndrome salivary gland epithe­
as lymphocyte apoptosis and lymphocyte activation-​ lial cells295. Why then has the field of IBM attracted so
induced cell death261. The highly differentiated effec­ much more interest in protein aggregates than other
tor memory and effector cytotoxic CD4+ and CD8+ autoimmune diseases? Perhaps it is simply easier to see
T cells (loss of CD28 and gain of CD244, CD57 and these cytoplasmic aggregates under the microscope in
KLRG1 expression, as discussed above) present in skeletal muscle than in other tissues. Skeletal muscle
IBM are resistant to apoptosis and activation-​induced fibres are unique syncytial structures that are densely
cell death229,262–264. packed with large proteins undergoing constant turno­
Unlike naive T cells, highly differentiated blood-​ ver; they have larger proportions of cytoplasm and larger
derived human T cells are resistant to corticosteroid cross-​sectional areas (10,000–20,000 μm2; approximately
inhibition ex vivo265–267. Individuals treated with cortico­ 50–100 times larger) than most other cells. There simply
steroids have reduced numbers of naive CD8+ T cells in is not sufficient room in cell types other than myofibres
their blood but increased numbers of highly differen­ for large cytoplasmic aggregates, such as p62 aggregates
tiated effector memory CD8+ T cells268. In a cohort of with sizes of 5–50 μm2, to accumulate and become
patients with a diagnosis of polymyositis, defined using visible by microscopy.

Nature Reviews | Rheumatology


Reviews

Conclusions as treatment refractory, only very limited therapeutic


Tremendous progress has been made in the clinical and approaches have been carefully studied to date, and none
pathogenic understanding of IBM over the past decade. of these has been rationally directed towards targeting
Major advances in biomarker identification, including the this population of T cells; some treatments (such as cor­
notable whole-​genome linkage to an HLA autoimmune ticosteroids and alemtuzumab) have had predictably
haplotype, the identification of a specific autoantibody counterproductive liabilities. Complete understanding
and T cell phenotypical abnormalities, have advanced of the pathogenesis of IBM, like most acquired diseases
IBM diagnosis and pathogenic understanding. IBM is in humans, awaits successful therapeutic responses from
unique among muscle diseases owing to its molecular mechanistically targeted therapies.
signature involving highly differentiated cytotoxic T cells
that have escaped immune regulation. Although viewed Published online xx xx xxxx

1. Unverricht, H. Polymyositis acuta progressiva. Z. Klin. 22. Chou, S. M. Myxovirus-​like structures in a case 44. Cortese, A. et al. Longitudinal observational study
Med. 12, 533–549 (1887). of human chronic polymyositis. Science 158, of sporadic inclusion body myositis: implications
2. Bohan, A. History and classification of polymyositis 1453–1455 (1967). for clinical trials. Neuromuscul. Disord. 23, 404–412
and dermatomyositis. Clin. Dermatol. 6, 3–8 23. Nishino, H., Engel, A. G. & Rima, B. K. Inclusion body (2013).
(1988). myositis: the mumps virus hypothesis. Ann. Neurol. 45. Hogrel, J. Y. et al. Four-​year longitudinal study of
3. Uverricht, H. Dermatomyositis acuta. Dtsch. Med. 25, 260–264 (1989). clinical and functional endpoints in sporadic inclusion
Wochenschr. 17, 41–44 (1891). 24. Kallajoki, M. et al. Inclusion body myositis and body myositis: implications for therapeutic trials.
4. Levine, T. D. History of dermatomyositis. Arch. Neurol. paramyxoviruses. Hum. Pathol. 22, 29–32 (1991). Neuromuscul. Disord. 24, 604–610 (2014).
60, 780–782 (2003). 25. Fox, S. A., Ward, B. K., Robbins, P. D., Mastaglia, F. L. 46. Alfano, L. N. et al. Modeling functional decline over
5. Carpenter, S., Karpati, G., Heller, I. & Eisen, A. & Swanson, N. R. Inclusion body myositis: investigation time in sporadic inclusion body myositis.
Inclusion body myositis: a distinct variety of of the mumps virus hypothesis by polymerase chain Muscle Nerve 55, 526–531 (2016).
idiopathic inflammatory myopathy. Neurology 28, reaction. Muscle Nerve 19, 23–28 (1996). 47. Rose, M. R. et al. A prospective natural history study
8–17 (1978). 26. Uruha, A. et al. Hepatitis C virus infection in inclusion of inclusion body myositis: implications for clinical
6. Emslie-​Smith, A. M. & Engel, A. G. Necrotizing body myositis: a case-​control study. Neurology 86, trials. Neurology 57, 548–550 (2001).
myopathy with pipestem capillaries, microvascular 211–217 (2016). 48. Arahata, K. & Engel, A. G. Monoclonal antibody
deposition of the complement membrane attack 27. Yunis, E. J. & Samaha, F. J. Inclusion body myositis. analysis of mononuclear cells in myopathies. I:
complex (MAC), and minimal cellular infiltration. Lab. Invest. 25, 240–248 (1971). Quantitation of subsets according to diagnosis and
Neurology 41, 936–939 (1991). 28. Danon, M. J., Reyes, M. G., Perurena, O. H., sites of accumulation and demonstration and counts
7. van der Meulen, M. F. et al. Polymyositis: Masdeu, J. C. & Manaligod, J. R. Inclusion body of muscle fibers invaded by T cells. Ann. Neurol. 16,
an overdiagnosed entity. Neurology 61, 316–321 myositis. A corticosteroid-​resistant idiopathic 193–208 (1984).
(2003). inflammatory myopathy. Arch. Neurol. 39, 760–764 49. Engel, A. G. & Arahata, K. Monoclonal antibody
8. Hoogendijk, J. E. et al. 119th ENMC international (1982). analysis of mononuclear cells in myopathies. II:
workshop: trial design in adult idiopathic inflammatory 29. Eisen, A., Berry, K. & Gibson, G. Inclusion body Phenotypes of autoinvasive cells in polymyositis and
myopathies, with the exception of inclusion body myositis (IBM): myopathy or neuropathy? Neurology inclusion body myositis. Ann. Neurol. 16, 209–215
myositis, 10-12 October 2003, Naarden, The 33, 1109–1114 (1983). (1984).
Netherlands. Neuromuscul. Disord. 14, 337–345 30. Ringel, S. P., Kenny, C. E., Neville, H. E., Giorno, R. 50. Arahata, K. & Engel, A. G. Monoclonal antibody
(2004). & Carry, M. R. Spectrum of inclusion body myositis. analysis of mononuclear cells in myopathies. III:
9. van de Vlekkert, J., Hoogendijk, J. E. & de Visser, M. Arch. Neurol. 44, 1154–1157 (1987). Immunoelectron microscopy aspects of cell-​mediated
Myositis with endomysial cell invasion indicates 31. Calabrese, L. H., Mitsumoto, H. & Chou, S. M. Inclusion muscle fiber injury. Ann. Neurol. 19, 112–125 (1986).
inclusion body myositis even if other criteria are not body myositis presenting as treatment-​resistant 51. Arahata, K. & Engel, A. G. Monoclonal antibody
fulfilled. Neuromuscul. Disord. 25, 451–456 polymyositis. Arthritis Rheum. 30, 397–403 (1987). analysis of mononuclear cells in myopathies. IV:
(2015). 32. Lotz, B. P., Engel, A. G., Nishino, H., Stevens, J. C. & Cell-​mediated cytotoxicity and muscle fiber necrosis.
10. Dalakas, M. C. Inflammatory muscle diseases. N. Engl. Litchy, W. J. Inclusion body myositis. Observations in Ann. Neurol. 23, 168–173 (1988).
J. Med. 372, 1734–1747 (2015). 40 patients. Brain 112, 727–747 (1989). 52. O’Hanlon, T. P., Dalakas, M. C., Plotz, P. H. & Miller, F. W.
11. Wolstencroft, P. W. & Fiorentino, D. F. 33. Sayers, M. E., Chou, S. M. & Calabrese, L. H. Inclusion The αβT cell receptor repertoire in inclusion body
Dermatomyositis clinical and pathological phenotypes body myositis: analysis of 32 cases. J. Rheumatol. 19, myositis: diverse patterns of gene expression by muscle-​
associated with myositis-​specific autoantibodies. 1385–1389 (1992). infiltrating lymphocytes. J. Autoimmun. 7, 321–333
Curr. Rheumatol. Rep. 20, 28 (2018). 34. Lindberg, C., Persson, L. I., Bjorkander, J. & (1994).
12. Schmidt, J. & Dalakas, M. C. Inclusion body myositis: Oldfors, A. Inclusion body myositis: clinical, 53. Lindberg, C., Oldfors, A. & Tarkowski, A. Restricted
from immunopathology and degenerative mechanisms morphological, physiological and laboratory findings use of T cell receptor V genes in endomysial infiltrates
to treatment perspectives. Expert Rev. Clin. Immunol. in 18 cases. Acta Neurol. Scand. 89, 123–131 of patients with inflammatory myopathies. Eur. J.
9, 1125–1133 (2013). (1994). Immunol. 24, 2659–2663 (1994).
13. Machado, P. M. et al. Ongoing developments in 35. Amato, A. A. et al. Inclusion body myositis: clinical and 54. Lindberg, C., Oldfors, A. & Tarkowski, A. Local T cell
sporadic inclusion body myositis. Curr. Rheumatol. pathological boundaries. Ann. Neurol. 40, 581–586 proliferation and differentiation in inflammatory
Rep. 16, 477 (2014). (1996). myopathies. Scand. J. Immunol. 41, 421–426 (1995).
14. Dimachkie, M. M. & Barohn, R. J. Inclusion body 36. Felice, K. J., Relva, G. M. & Conway, S. R. Further 55. Fyhr, I. M., Moslemi, A. R., Tarkowski, A., Lindberg, C.
myositis. Neurol. Clin. 32, 629–646 (2014). observations on forearm flexor weakness in inclusion & Oldfors, A. Limited T cell receptor V gene usage in
15. Mastaglia, F. L. & Needham, M. Inclusion body body myositis. Muscle Nerve 21, 659–661 (1998). inclusion body myositis. Scand. J. Immunol. 43,
myositis: a review of clinical and genetic aspects, 37. Phillips, B. A. et al. Patterns of muscle involvement 109–114 (1996).
diagnostic criteria and therapeutic approaches. in inclusion body myositis: clinical and magnetic 56. Fyhr, I. M. et al. Oligoclonal expansion of muscle
J. Clin. Neurosci. 22, 6–13 (2015). resonance imaging study. Muscle Nerve 24, infiltrating T cells in inclusion body myositis.
16. Greenberg, S. A. Inclusion body myositis. Continuum 1526–1534 (2001). J. Neuroimmunol. 79, 185–189 (1997).
22, 1871–1888 (2016). 38. Badrising, U. A. et al. Inclusion body myositis. 57. Bender, A., Behrens, L., Engel, A. G. & Hohlfeld, R.
17. Gallay, L. & Petiot, P. Sporadic inclusion-​body Clinical features and clinical course of the disease T cell heterogeneity in muscle lesions of inclusion body
myositis: recent advances and the state of the art in in 64 patients. J. Neurol. 252, 1448–1454 (2005). myositis. J. Neuroimmunol. 84, 86–91 (1998).
2016. Rev. Neurol. 172, 581–586 (2016). 39. Benveniste, O. et al. Long-​term observational study 58. Amemiya, K., Granger, R. P. & Dalakas, M. C. Clonal
18. Needham, M. & Mastaglia, F. L. Sporadic inclusion of sporadic inclusion body myositis. Brain 134, restriction of T cell receptor expression by infiltrating
body myositis: a review of recent clinical advances 3176–3184 (2011). lymphocytes in inclusion body myositis persists over
and current approaches to diagnosis and treatment. 40. Price, M. A. et al. Mortality and causes of death in time. Studies in repeated muscle biopsies. Brain 123,
Clin. Neurophysiol. 127, 1764–1773 (2016). patients with sporadic inclusion body myositis: survey 2030–2039 (2000).
19. Schmidt, K. & Schmidt, J. Inclusion body myositis: study based on the clinical experience of specialists in 59. Muntzing, K., Lindberg, C., Moslemi, A. R. & Oldfors, A.
advancements in diagnosis, pathomechanisms, and Australia, Europe and the USA. J. Neuromuscul. Dis. Inclusion body myositis: clonal expansions of muscle-​
treatment. Curr. Opin. Rheumatol. 29, 632–638 3, 67–75 (2016). infiltrating T cells persist over time. Scand. J. Immunol.
(2017). 41. Felice, K. J. & North, W. A. Inclusion body myositis in 58, 195–200 (2003).
20. Benveniste, O. et al. Amyloid deposits and Connecticut: observations in 35 patients during an 60. Dimitri, D. et al. Shared blood and muscle CD8+T cell
inflammatory infiltrates in sporadic inclusion body 8-year period. Medicine 80, 320–327 (2001). expansions in inclusion body myositis. Brain 129,
myositis: the inflammatory egg comes before the 42. Needham, M. et al. Sporadic inclusion body myositis: 986–995 (2006).
degenerative chicken. Acta Neuropathol. 129, phenotypic variability and influence of HLA-​DR3 in a 61. Salajegheh, M. et al. T cell receptor profiling in muscle
611–624 (2015). cohort of 57 Australian cases. J. Neurol. Neurosurg. and blood lymphocytes in sporadic inclusion body
21. Keller, C. W., Schmidt, J. & Lunemann, J. D. Immune Psychiatry 79, 1056–1060 (2008). myositis. Neurology 69, 1672–1679 (2007).
and myodegenerative pathomechanisms in inclusion 43. Cox, F. M. et al. A 12-year follow-​up in sporadic 62. Pandya, J. M. et al. Expanded T cell receptor
body myositis. Ann. Clin. Transl Neurol. 4, 422–445 inclusion body myositis: an end stage with major Vβ-​restricted T cells from patients with sporadic
(2017). disabilities. Brain 134, 3167–3175 (2011). inclusion body myositis are proinflammatory and

www.nature.com/nrrheum
Reviews

cytotoxic CD28null T cells. Arthritis Rheum. 62, 89. Price, P. et al. Two major histocompatibility complex 113. Paltiel, A. D. et al. Demographic and clinical features
3457–3466 (2010). haplotypes influence susceptibility to sporadic of inclusion body myositis in North America.
63. Allenbach, Y. et al. Th1 response and systemic treg inclusion body myositis: critical evaluation of an Muscle Nerve 52, 527–533 (2015).
deficiency in inclusion body myositis. PLOS ONE 9, association with HLA-​DR3. Tissue Antigens 64, 114. Keshishian, A., Greenberg, S. A., Agashivala, N.,
e88788 (2014). 575–580 (2004). Baser, O. & Johnson, K. Health care costs and
64. Greenberg, S. A., Pinkus, J. L., Amato, A. A., 90. Scott, A. P. et al. Sporadic inclusion body myositis comorbidities for patients with inclusion body
Kristensen, T. & Dorfman, D. M. Association of in Japanese is associated with the MHC ancestral myositis. Curr. Med. Res. Opin. 34, 1679–1685
inclusion body myositis with T cell large granular haplotype 52.1. Neuromuscul. Disord. 16, 311–315 (2018).
lymphocytic leukaemia. Brain 139, 1348–1360 (2006). 115. Ko, E. H. & Rubin, A. D. Dysphagia due to inclusion
(2016). 91. Rojana-​udomsart, A. et al. The association of sporadic body myositis: case presentation and review of the
65. Hohlfeld, R. & Schulze-​Koops, H. Cytotoxic T cells inclusion body myositis and Sjögren’s syndrome in literature. Ann. Otol. Rhinol. Laryngol. 123, 605–608
go awry in inclusion body myositis. Brain 139, carriers of HLA-​DR3 and the 8.1 MHC ancestral (2014).
1312–1314 (2016). haplotype. Clin. Neurol. Neurosurg. 113, 559–563 116. Cox, F. M. et al. Detecting dysphagia in inclusion body
66. Lindberg, C., Trysberg, E., Tarkowski, A. & Oldfors, A. (2011). myositis. J. Neurol. 256, 2009–2013 (2009).
Anti-​T-lymphocyte globulin treatment in inclusion body 92. Rojana-​udomsart, A. et al. High-​resolution HLA-​DRB1 117. Houser, S. M., Calabrese, L. H. & Strome, M.
myositis: a randomized pilot study. Neurology 61, genotyping in an Australian inclusion body myositis Dysphagia in patients with inclusion body myositis.
260–262 (2003). (s-​IBM) cohort: an analysis of disease-​associated Laryngoscope 108, 1001–1005 (1998).
67. Dalakas, M. C. et al. Effect of Alemtuzumab (CAMPATH alleles and diplotypes. J. Neuroimmunol. 250, 77–82 118. Oh, T. H., Brumfield, K. A., Hoskin, T. L.,
1-H) in patients with inclusion-​body myositis. Brain (2012). Kasperbauer, J. L. & Basford, J. R. Dysphagia in
132, 1536–1544 (2009). 93. Rojana-​udomsart, A. et al. Analysis of HLA-​DRB3 inclusion body myositis: clinical features, management,
68. Hogrel, J. Y. et al. Rapamycin vs. placebo for the alleles and supertypical genotypes in the MHC and clinical outcome. Am. J. Phys. Med. Rehabil. 87,
treatment of inclusion body myositis: improvement of class II region in sporadic inclusion body myositis. 883–889 (2008).
the 6 min walking distance, a functional scale, the FVC J. Neuroimmunol. 254, 174–177 (2013). 119. Riminton, D. S., Chambers, S. T., Parkin, P. J., Pollock, M.
and muscle quantitative MRI. Arthritis Rheumatol. 69, 94. Rothwell, S. et al. Immune-​array analysis in sporadic & Donaldson, I. M. Inclusion body myositis presenting
5L (2017). inclusion body myositis reveals HLA-​DRB1 amino acid solely as dysphagia. Neurology 43, 1241–1243 (1993).
69. Targoff, I. N. Autoantibodies and their significance in heterogeneity across the myositis spectrum. 120. Verma, A., Bradley, W. G., Adesina, A. M., Sofferman, R.
myositis. Curr. Rheumatol. Rep. 10, 333–340 (2008). Arthritis Rheumatol. 69, 1090–1099 (2017). & Pendlebury, W. W. Inclusion body myositis with
70. Nishikai, M. & Reichlin, M. Heterogeneity of 95. Dalakas, M. C. et al. Treatment of inclusion-​body cricopharyngeus muscle involvement and severe
precipitating antibodies in polymyositis and myositis with IVIg: a double-​blind, placebo-​controlled dysphagia. Muscle Nerve 14, 470–473 (1991).
dermatomyositis. Characterization of the Jo-1 study. Neurology 48, 712–716 (1997). 121. Rodriguez Cruz, P. M., Needham, M., Hollingsworth, P.,
antibody system. Arthritis Rheum. 23, 881–888 96. Greenberg, S. A., Pinkus, J. L. & Amato, A. A. Mastaglia, F. L. & Hillman, D. R. Sleep disordered
(1980). Nuclear membrane proteins are present within breathing and subclinical impairment of respiratory
71. Reichlin, M. & Mattioli, M. Description of a serological rimmed vacuoles in inclusion-​body myositis. Muscle function are common in sporadic inclusion body
reaction characteristic of polymyositis. Clin. Immunol. Nerve 34, 406–416 (2006). myositis. Neuromuscul. Disord. 24, 1036–1041
Immunopathol. 5, 12–20 (1976). 97. Chahin, N. & Engel, A. G. Correlation of muscle biopsy, (2014).
72. McHugh, N. J. & Tansley, S. L. Autoantibodies in clinical course, and outcome in PM and sporadic IBM. 122. Brady, S., Squier, W. & Hilton-​Jones, D. Clinical
myositis. Nat. Rev. Rheumatol. 14, 290–302 (2018). Neurology 70, 418–424 (2008). assessment determines the diagnosis of inclusion
73. Greenberg, S. A. et al. Molecular profiles of 98. Tawara, N. et al. Pathomechanisms of anti-​cytosolic body myositis independently of pathological features.
inflammatory myopathies. Neurology 59, 1170–1182 5′-nucleotidase 1 A autoantibodies in sporadic J. Neurol. Neurosurg. Psychiatry 84, 1240–1246
(2002). inclusion body myositis. Ann. Neurol. 81, 512–525 (2013).
74. Greenberg, S. A. et al. Plasma cells in muscle in (2017). 123. Dion, E. et al. Magnetic resonance imaging criteria
inclusion body myositis and polymyositis. Neurology 99. Johns Hopkins University. Myopathy, myofibrillar, 1; for distinguishing between inclusion body myositis
65, 1782–1787 (2005). MFM1. OMIM https://www.omim.org/entry/601419 and polymyositis. J. Rheumatol. 29, 1897–1906
75. Bradshaw, E. M. et al. A local antigen-​driven humoral (2014). (2002).
response is present in the inflammatory myopathies. 100. Ahmed, M. et al. Targeting protein homeostasis in 124. Cox, F. M. et al. Magnetic resonance imaging of
J. Immunol. 178, 547–556 (2007). sporadic inclusion body myositis. Sci. Transl Med. 8, skeletal muscles in sporadic inclusion body myositis.
76. Salajegheh, M. et al. Permissive environment for 331ra41 (2016). Rheumatology 50, 1153–1161 (2011).
B cell maturation in myositis muscle in the absence 101. Sivakumar, K., Semino-​Mora, C. & Dalakas, M. C. 125. Inaishi, Y. et al. MRI for evaluation of flexor digitorum
of B cell follicles. Muscle Nerve 42, 576–583 (2010). An inflammatory, familial, inclusion body myositis with profundus muscle involvement in inclusion body
77. Ray, A. et al. Autoantibodies produced at the autoimmune features and a phenotype identical to myositis. Can. J. Neurol. Sci. 41, 780–781 (2014).
site of tissue damage provide evidence of humoral sporadic inclusion body myositis. Studies in three 126. Tasca, G. et al. Magnetic resonance imaging pattern
autoimmunity in inclusion body myositis. PLOS ONE families. Brain 120, 653–661 (1997). recognition in sporadic inclusion-​body myositis.
7, e46709 (2012). 102. Ranque-​Francois, B. et al. Familial inflammatory Muscle Nerve 52, 956–962 (2015).
78. Salajegheh, M., Lam, T. & Greenberg, S. A. inclusion body myositis. Ann. Rheum. Dis. 64, 127. Guimaraes, J. B. et al. Sporadic inclusion body myositis:
Autoantibodies against a 43kDa muscle protein in 634–637 (2005). MRI findings and correlation with clinical and functional
inclusion body myositis. PLOS ONE 6, e20266 (2011). 103. Tateyama, M. et al. Familial inclusion body myositis: parameters. AJR Am. J. Roentgenol. 209, 1340–1347
79. Larman, H. B. et al. Cytosolic 5′-nucleotidase 1A a report on two Japanese sisters. Intern. Med. 42, (2017).
autoimmunity in sporadic inclusion body myositis. 1035–1038 (2003). 128. Tsukita, K., Yagita, K., Sakamaki-​Tsukita, H. & Suenaga, T.
Ann. Neurol. 73, 408–418 (2013). 104. Callan, A., Capkun, G., Vasanthaprasad, V., Freitas, R. Sporadic inclusion body myositis: magnetic resonance
80. Pluk, H. et al. Autoantibodies to cytosolic & Needham, M. A. Systematic review and meta-​ imaging and ultrasound characteristics. QJM 111,
5′-nucleotidase IA in inclusion body myositis. analysis of prevalence studies of sporadic inclusion 667–668 (2018).
Ann. Neurol. 73, 397–407 (2013). body myositis. J. Neuromuscul. Dis. 4, 127–137 129. Noto, Y. et al. Contrasting echogenicity in flexor
81. Mendell, J. R., Sahenk, Z., Gales, T. & Paul, L. Amyloid (2017). digitorum profundus-​flexor carpi ulnaris: a diagnostic
filaments in inclusion body myositis. Novel findings 105. Tan, J. A. et al. Incidence and prevalence of ultrasound pattern in sporadic inclusion body
provide insight into nature of filaments. Arch. Neurol. idiopathic inflammatory myopathies in South myositis. Muscle Nerve 49, 745–748 (2014).
48, 1229–1234 (1991). Australia: a 30-year epidemiologic study of 130. Nodera, H. et al. Intramuscular dissociation of
82. Oldfors, A., Larsson, N. G., Lindberg, C. & Holme, E. histology-​proven cases. Int. J. Rheum. Dis. 16, echogenicity in the triceps surae characterizes
Mitochondrial DNA deletions in inclusion body 331–338 (2013). sporadic inclusion body myositis. Eur. J. Neurol. 23,
myositis. Brain 116, 325–336 (1993). 106. Badrising, U. A. et al. Epidemiology of inclusion body 588–596 (2016).
83. Schmidt, J. et al. Interrelation of inflammation myositis in the Netherlands: a nationwide study. 131. Albayda, J. et al. Pattern of muscle involvement in
and APP in sIBM: IL-1β induces accumulation of Neurology 55, 1385–1387 (2000). inclusion body myositis: a sonographic study. Clin. Exp.
β-​amyloid in skeletal muscle. Brain 131, 1228–1240 107. Lefter, S., Hardiman, O. & Ryan, A. M. A population-​ Rheumatol. 36, 996–1002 (2018).
(2008). based epidemiologic study of adult neuromuscular 132. Bachasson, D., Dubois, G. J. R., Allenbach, Y.,
84. Freret, M. et al. Overexpression of MHC class I in disease in the Republic of Ireland. Neurology 88, Benveniste, O. & Hogrel, J. Y. Muscle shear wave
muscle of lymphocyte-​deficient mice causes a severe 304–313 (2017). elastography in inclusion body myositis: feasibility,
myopathy with induction of the unfolded protein 108. Suzuki, N. et al. Increase in number of sporadic reliability and relationships with muscle
response. Am. J. Pathol. 183, 893–904 (2013). inclusion body myositis (sIBM) in Japan. J. Neurol. impairments. Ultrasound Med. Biol. 44,
85. Rygiel, K. A. et al. Mitochondrial and inflammatory 259, 554–556 (2012). 1423–1432 (2018).
changes in sporadic inclusion body myositis. 109. Dobloug, G. C. et al. High prevalence of inclusion body 133. Olthoff, A. et al. Evaluation of dysphagia by novel
Neuropathol. Appl. Neurobiol. 41, 288–303 (2015). myositis in Norway; a population-​based clinical real-​time MRI. Neurology 87, 2132–2138 (2016).
86. Garlepp, M. J., Laing, B., Zilko, P. J., Ollier, W. epidemiology study. Eur. J. Neurol. 22, 672 (2015). 134. Koffman, B. M., Rugiero, M. & Dalakas, M. C.
& Mastaglia, F. L. HLA associations with inclusion 110. Suzuki, N. et al. Multicenter questionnaire survey for Immune-​mediated conditions and antibodies
body myositis. Clin. Exp. Immunol. 98, 40–45 sporadic inclusion body myositis in Japan. Orphanet associated with sporadic inclusion body myositis.
(1994). J. Rare Dis. 11, 146 (2016). Muscle Nerve 21, 115–117 (1998).
87. Koffman, B. M., Sivakumar, K., Simonis, T., Stroncek, D. 111. Wilson, F. C., Ytterberg, S. R., St Sauver, J. L. 135. Greenberg, S. A. Cytoplasmic 5′-nucleotidase
& Dalakas, M. C. HLA allele distribution distinguishes & Reed, A. M. Epidemiology of sporadic inclusion autoantibodies in inclusion body myositis:
sporadic inclusion body myositis from hereditary body myositis and polymyositis in Olmsted County, Isotypes and diagnostic utility. Muscle Nerve 50,
inclusion body myopathies. J. Neuroimmunol. 84, Minnesota. J. Rheumatol. 35, 445–447 (2008). 488–492 (2014).
139–142 (1998). 112. Chilingaryan, A., Rison, R. A. & Beydoun, S. R. 136. Goyal, N. A. et al. Seropositivity for NT5c1A antibody
88. Lampe, J. B. et al. Analysis of HLA class I and II alleles Misdiagnosis of inclusion body myositis: two case in sporadic inclusion body myositis predicts more severe
in sporadic inclusion-​body myositis. J. Neurol. 250, reports and a retrospective chart review. J. Med. Case motor, bulbar and respiratory involvement. J. Neurol.
1313–1317 (2003). Rep. 9, 169 (2015). Neurosurg. Psychiatry 87, 373–378 (2016).

Nature Reviews | Rheumatology


Reviews

137. Lloyd, T. E. et al. Cytosolic 5′-nucleotidase 1A as a 162. Lloyd, T. E. et al. Overlapping features of polymyositis 186. Dalakas, M. C. et al. A controlled study of
target of circulating autoantibodies in autoimmune and inclusion body myositis in HIV-​infected patients. intravenous immunoglobulin combined with
diseases. Arthritis Care Res. 68, 66–71 (2016). Neurology 88, 1454–1460 (2017). prednisone in the treatment of IBM. Neurology 56,
138. Kramp, S. L. et al. Development and evaluation 163. Hiniker, A., Daniels, B. H. & Margeta, M. T-​cell- 323–327 (2001).
of a standardized ELISA for the determination of mediated inflammatory myopathies in HIV-​positive 187. Badrising, U. A. et al. Comparison of weakness
autoantibodies against cN-1A (Mup44, NT5C1A) individuals: a histologic study of 19 cases. progression in inclusion body myositis during
in sporadic inclusion body myositis. Auto Immun. J. Neuropathol. Exp. Neurol. 75, 239–245 (2016). treatment with methotrexate or placebo. Ann. Neurol.
Highlights 7, 16 (2016). 164. Cupler, E. J. et al. Inclusion body myositis in HIV-1 and 51, 369–372 (2002).
139. Felice, K. J. et al. Sensitivity and clinical utility of the HTLV-1 infected patients. Brain 119, 1887–1893 188. Muscle Study, G. Randomized pilot trial of βINF1a
anti-​cytosolic 5’-nucleotidase 1 A (cN1A) antibody test (1996). (Avonex) in patients with inclusion body myositis.
in sporadic inclusion body myositis: Report of 40 165. Couture, P. et al. Inclusion body myositis and human Neurology 57, 1566–1570 (2001).
patients from a single neuromuscular center. immunodeficiency virus type 1: a new case report and 189. Muscle Study, G. Randomized pilot trial of high-​dose
Neuromuscul. Disord. 28, 600–664 (2018). literature review. Neuromuscul. Disord. 28, 334–338 βINF-1a in patients with inclusion body myositis.
140. Herbert, M. K. et al. Disease specificity of (2018). Neurology 63, 718–720 (­20­04­).
autoantibodies to cytosolic 5′-nucleotidase 1A 166. Matsuura, E. et al. Inclusion body myositis 190. Rutkove, S. B. et al. A pilot randomized trial of
in sporadic inclusion body myositis versus known associated with human T-​lymphotropic virus-​type I oxandrolone in inclusion body myositis. Neurology 58,
autoimmune diseases. Ann. Rheum. Dis. 75, 696–701 infection: eleven patients from an endemic area in 1081–1087 (2002).
(2016). Japan. J. Neuropathol. Exp. Neurol. 67, 41–49 191. Amato, A. A. et al. Treatment of sporadic inclusion
141. Muro, Y., Nakanishi, H., Katsuno, M., Kono, M. (2008). body myositis with bimagrumab. Neurology 83,
& Akiyama, M. Prevalence of anti-​NT5C1A antibodies 167. Cox, F. M. et al. The heart in sporadic inclusion body 2239–2246 (2014).
in Japanese patients with autoimmune rheumatic myositis: a study in 51 patients. J. Neurol. 257, 192. Amato, A. A. et al. A randomized, double-​blind,
diseases in comparison with other patient cohorts. 447–451 (2010). placebo-​controlled study of bimagrumab in patients
Clin. Chim. Acta 472, 1–4 (2017). 168. Limaye, V. S., Lester, S., Blumbergs, P. with sporadic inclusion body myositis [abstract 8L].
142. Rietveld, A. et al. Autoantibodies to cytosolic & Roberts-​Thomson, P. J. Idiopathic inflammatory Arthritis Rheumatol. 68, 4367–4369 (2016).
5′-nucleotidase 1A in primary Sjögren’s syndrome myositis is associated with a high incidence of 193. Chou, S. M. Myxovirus-​like structures and
and systemic lupus erythematosus. Front. Immunol. 9, hypertension and diabetes mellitus. Int. J. Rheum. accompanying nuclear changes in chronic
1200 (2018). Dis. 13, 132–137 (2010). polymyositis. Arch. Pathol. 86, 649–658 (1968).
143. Mhiri, C. & Gherardi, R. Inclusion body myositis in 169. Lai, Y. T. et al. Dermatomyositis is associated with an 194. Rifai, Z., Welle, S., Kamp, C. & Thornton, C. A.
French patients. A clinicopathological evaluation. increased risk of cardiovascular and cerebrovascular Ragged red fibers in normal aging and inflammatory
Neuropathol. Appl. Neurobiol. 16, 333–344 (1990). events: a Taiwanese population-​based longitudinal myopathy. Ann. Neurol. 37, 24–29 (1995).
144. Askanas, V. & Engel, W. K. Molecular pathology and follow-​up study. Br. J. Dermatol. 168, 1054–1059 195. Oldfors, A. et al. Mitochondrial abnormalities in
pathogenesis of inclusion-​body myositis. Microsc. Res. (2013). inclusion-​body myositis. Neurology 66, S49–S55
Tech. 67, 114–120 (2005). 170. Wang, H., Tang, J., Chen, X., Li, F. & Luo, J. Lipid profiles (2006).
145. Rodriguez Cruz, P. M. et al. An analysis of the in untreated patients with dermatomyositis. J. Eur. 196. Askanas, V., Serdaroglu, P., Engel, W. K. &
sensitivity and specificity of MHC-​I and MHC-​II Acad. Dermatol. Venereol. 27, 175–179 (2013). Alvarez, R. B. Immunolocalization of ubiquitin in muscle
immunohistochemical staining in muscle biopsies 171. Wang, H. et al. Altered lipid levels in untreated biopsies of patients with inclusion body myositis and
for the diagnosis of inflammatory myopathies. patients with early polymyositis. PLOS ONE 9, oculopharyngeal muscular dystrophy. Neurosci. Lett.
Neuromuscul. Disord. 24, 1025–1035 (2014). e89827 (2014). 130, 73–76 (1991).
146. Ikenaga, C. et al. Clinicopathologic features of myositis 172. Diederichsen, L. P. et al. Traditional cardiovascular risk 197. Askanas, V., Engel, W. K. & Alvarez, R. B. Light and
patients with CD8-MHC-1 complex pathology. factors and coronary artery calcification in adults with electron microscopic localization of β-​amyloid protein
Neurology 89, 1060–1068 (2017). polymyositis and dermatomyositis: a Danish multicenter in muscle biopsies of patients with inclusion-​body
147. Temiz, P., Weihl, C. C. & Pestronk, A. Inflammatory study. Arthritis Care Res. 67, 848–854 (2015). myositis. Am. J. Pathol. 141, 31–36 (1992).
myopathies with mitochondrial pathology and protein 173. Rai, S. K., Choi, H. K., Sayre, E. C. & Avina-​Zubieta, J. A. 198. Askanas, V., Engel, W. K., Bilak, M., Alvarez, R. B.
aggregates. J. Neurol. Sci. 278, 25–29 (2009). Risk of myocardial infarction and ischaemic stroke & Selkoe, D. J. Twisted tubulofilaments of inclusion
148. Pestronk, A. Acquired immune and inflammatory in adults with polymyositis and dermatomyositis: body myositis muscle resemble paired helical
myopathies: pathologic classification. Curr. Opin. a general population-​based study. Rheumatology 55, filaments of Alzheimer brain and contain
Rheumatol. 23, 595–604 (2011). 461–469 (2016). hyperphosphorylated tau. Am. J. Pathol. 144,
149. van der Meulen, M. F. et al. Rimmed vacuoles and the 174. Sherer, Y. & Shoenfeld, Y. Mechanisms of disease: 177–187 (1994).
added value of SMI-31 staining in diagnosing sporadic atherosclerosis in autoimmune diseases. Nat. Clin. 199. Greenberg, S. A. Theories of the pathogenesis of
inclusion body myositis. Neuromuscul. Disord. 11, Pract. Rheumatol. 2, 99–106 (2006). inclusion body myositis. Curr. Rheumatol. Rep. 12,
447–451 (2001). 175. Ahearn, J., Shields, K. J., Liu, C. C. & Manzi, S. 221–228 (2010).
150. Dalakas, M. C. Polymyositis, dermatomyositis and Cardiovascular disease biomarkers across autoimmune 200. Askanas, V. & Engel, W. K. Proposed pathogenetic
inclusion-​body myositis. N. Engl. J. Med. 325, diseases. Clin. Immunol. 161, 59–63 (2015). cascade of inclusion-​body myositis: importance of
1487–1498 (1991). 176. Alexanderson, H. Exercise in inflammatory amyloid-​β, misfolded proteins, predisposing genes,
151. Mastaglia, F. L. & Phillips, B. A. Idiopathic myopathies, including inclusion body myositis. and aging. Curr. Opin. Rheumatol. 15, 737–744
inflammatory myopathies: epidemiology, classification, Curr. Rheumatol. Rep. 14, 244–251 (2012). (2003).
and diagnostic criteria. Rheum. Dis. Clin. North Am. 177. Arnardottir, S., Alexanderson, H., Lundberg, I. E. 201. Askanas, V. & Engel, W. K. Inclusion-​body myositis:
28, 723–741 (2002). & Borg, K. Sporadic inclusion body myositis: pilot a myodegenerative conformational disorder associated
152. Tawil, R. & Griggs, R. C. Inclusion body myositis. study on the effects of a home exercise program on with Aβ, protein misfolding, and proteasome inhibition.
Curr. Opin. Rheumatol. 14, 653–657 (2002). muscle function, histopathology and inflammatory Neurology 66, S39–S48 (2006).
153. Verschuuren, J. J., van Engelen, B. G. M., van der reaction. J. Rehabil. Med. 35, 31–35 (2003). 202. Askanas, V., Engel, W. K. & Nogalska, A. Sporadic
Hoeven, H. & Hoogendijk, J. Inclusion body myositis 178. Johnson, L. G., Edwards, D. J., Walters, S. E., inclusion-​body myositis: a degenerative muscle
diagnostic criteria. Inclusion Body Myositis. http:// Thickbroom, G. W. & Mastaglia, F. L. The effectiveness disease associated with aging, impaired muscle
ibmmyositis.com/emery81.pdf (1997). of an individualized, home-​based functional exercise protein homeostasis and abnormal mitophagy.
154. Griggs, R. C. et al. Inclusion body myositis and program for patients with sporadic inclusion body Biochim. Biophys. Acta 1852, 633–643 (2015).
myopathies. Ann. Neurol. 38, 705–713 (1995). myositis. J. Clin. Neuromuscul. Dis. 8, 187–194 203. Greenberg, S. A. How citation distortions create
155. Hilton-​Jones, D. et al. Inclusion body myositis: (2007). unfounded authority: analysis of a citation network.
MRC Centre for Neuromuscular Diseases, IBM 179. Parker, K. C. et al. Fast-​twitch sarcomeric and BMJ 339, b2680 (2009).
workshop, London, 13 June 2008. Neuromuscul Disord. glycolytic enzyme protein loss in inclusion body 204. Fergusson, D. Inappropriate referencing in research.
20, 142–147 (2010). myositis. Muscle Nerve 39, 739–753 (2009). BMJ 339, b2049 (2009).
156. Benveniste, O. & Hilton-​Jones, D. International 180. Cherin, P. et al. Intravenous immunoglobulin for 205. Sarkozi, E., Askanas, V., Johnson, S. A., McFerrin, J.
Workshop on Inclusion Body Myositis held at the dysphagia of inclusion body myositis. Neurology 58, & Engel, W. K. Expression of β-​amyloid precursor
Institute of Myology, Paris, on 29 May 2009. 326 (2002). protein gene is developmentally regulated in human
Neuromuscul. Disord. 20, 414–421 (2010). 181. Pars, K. et al. Subcutaneous immunoglobulin treatment muscle fibers in vivo and in vitro. Exp. Neurol. 128,
157. Rose, M. R. 188th ENMC International Workshop: of inclusion-​body myositis stabilizes dysphagia. 27–33 (1994).
Inclusion Body Myositis, 2–4 December 2011, Muscle Nerve 48, 838–839 (2013). 206. Askanas, V. & Engel, W. K. Sporadic inclusion-​body
Naarden, The Netherlands. Neuromuscul. Disord. 23, 182. Cherin, P., Delain, J. C., de Jaeger, C. & Crave, J. C. myositis: conformational multifactorial ageing-​related
1044–1055 (2013). Subcutaneous immunoglobulin use in inclusion body degenerative muscle disease associated with
158. Lloyd, T. E. et al. Evaluation and construction of myositis: a review of 6 cases. Case Rep. Neurol. 7, proteasomal and lysosomal inhibition, endoplasmic
diagnostic criteria for inclusion body myositis. 227–232 (2015). reticulum stress, and accumulation of amyloid-​β42
Neurology 83, 426–433 (2014). 183. Mendell, J. R. et al. Follistatin gene therapy for oligomers and phosphorylated tau. Presse Med. 40,
159. Kanellopoulos, P., Baltoyiannis, C. & Tzioufas, A. G. sporadic inclusion body myositis improves e219–e235 (2011).
Primary Sjögren’s syndrome associated with functional outcomes. Mol. Ther. 25, 870–879 207. Salajegheh, M. et al. Nature of “Tau” immunoreactivity
inclusion body myositis. Rheumatology 41, (2017). in normal myonuclei and inclusion body myositis.
440–444 (2002). 184. Greenberg, S. A. Unfounded claims of improved Muscle Nerve 40, 520–528 (2009).
160. Misterska-​Skora, M., Sebastian, A., Dziegiel, P., functional outcomes attributed to follistatin gene 208. Pruitt, J. N. 2nd, Showalter, C. J. & Engel, A. G.
Sebastian, M. & Wiland, P. Inclusion body myositis therapy in inclusion body myositis. Mol. Ther. 25, Sporadic inclusion body myositis: counts of different
associated with Sjögren’s syndrome. Rheumatol. Int. 2235–2237 (2017). types of abnormal fibers. Ann. Neurol. 39, 139–143
33, 3083–3086 (2013). 185. Walter, M. C. et al. High-​dose immunoglobulin therapy (1996).
161. Colafrancesco, S. et al. Myositis in primary Sjögren’s in sporadic inclusion body myositis: a double-​blind, 209. Banwell, B. L. & Engel, A. G. αB-​Crystallin
syndrome: data from a multicentre cohort. Clin. Exp. placebo-​controlled study. J. Neurol. 247, 22–28 immunolocalization yields new insights into inclusion
Rheumatol. 33, 457–464 (2015). (2000). body myositis. Neurology 54, 1033–1041 (2000).

www.nature.com/nrrheum
Reviews

210. Sherriff, F. E., Joachim, C. L., Squier, M. V. & Esiri, M. M. 234. Duftner, C. et al. Prevalence, clinical relevance and 259. Zhong, Z. et al. NF-​κB restricts inflammasome
Ubiquitinated inclusions in inclusion-​body myositis characterization of circulating cytotoxic CD4+CD28- activation via elimination of damaged mitochondria.
patients are immunoreactive for cathepsin D but not T cells in ankylosing spondylitis. Arthritis Res. Ther. 5, Cell 164, 896–910 (2016).
β-​amyloid. Neurosci. Lett. 194, 37–40 (1995). R292–R300 (2003). 260. Ozden, S. et al. Direct evidence for a chronic CD8+-
211. Dubourg, O. et al. Diagnostic value of markers of 235. Pinto-​Medel, M. J. et al. The CD4+T cell subset lacking T cell-​mediated immune reaction to tax within the
muscle degeneration in sporadic inclusion body expression of the CD28 costimulatory molecule is muscle of a human T cell leukemia/lymphoma virus
myositis. Acta Myol 30, 103–108 (2011). expanded and shows a higher activation state in type 1-infected patient with sporadic inclusion body
212. Hiniker, A., Daniels, B. H., Lee, H. S. & Margeta, M. multiple sclerosis. J. Neuroimmunol. 243, 1–11 myositis. J. Virol. 78, 10320–10327 (2004).
Comparative utility of LC3, p62 and TDP-43 (2012). 261. Green, D. R., Droin, N. & Pinkoski, M. Activation-​
immunohistochemistry in differentiation of inclusion 236. Garcia de Tena, J. et al. Active Crohn’s disease patients induced cell death in T cells. Immunol. Rev. 193,
body myositis from polymyositis and related show a distinctive expansion of circulating memory 70–81 (2003).
inflammatory myopathies. Acta Neuropathol. CD4+CD45RO+CD28null T cells. J. Clin. Immunol. 24, 262. Vallejo, A. N., Schirmer, M., Weyand, C. M.
Commun. 1, 29 (2013). 185–196 (2004). & Goronzy, J. J. Clonality and longevity of
213. Vattemi, G., Engel, W. K., McFerrin, J. & Askanas, V. 237. Leblanc, F., Zhang, D., Liu, X. & Loughran, T. P. Large CD4+CD28null T cells are associated with defects in
Endoplasmic reticulum stress and unfolded protein granular lymphocyte leukemia: from dysregulated apoptotic pathways. J. Immunol. 165, 6301–6307
response in inclusion body myositis muscle. Am. J. pathways to therapeutic targets. Future Oncol. 8, (2000).
Pathol. 164, 1–7 (2004). 787–801 (2012). 263. Spaulding, C., Guo, W. & Effros, R. B. Resistance to
214. Lunemann, J. D. et al. β-​amyloid is a substrate of 238. Mastaglia, F. L. et al. Polymorphism in the TOMM40 apoptosis in human CD8+T cells that reach replicative
autophagy in sporadic inclusion body myositis. gene modifies the risk of developing sporadic inclusion senescence after multiple rounds of antigen-​specific
Ann. Neurol. 61, 476–483 (2007). body myositis and the age of onset of symptoms. proliferation. Exp. Gerontol. 34, 633–644 (1999).
215. Nakano, S., Oki, M. & Kusaka, H. The role of p62/ Neuromuscul. Disord. 23, 969–974 (2013). 264. Posnett, D. N., Edinger, J. W., Manavalan, J. S., Irwin, C.
SQSTM1 in sporadic inclusion body myositis. 239. Gang, Q. et al. The effects of an intronic polymorphism & Marodon, G. Differentiation of human CD8 T cells:
Neuromuscul. Disord. 27, 363–369 (2017). in TOMM40 and APOE genotypes in sporadic implications for in vivo persistence of CD8+CD28-
216. Pinkus, J. L., Amato, A. A., Taylor, J. P. inclusion body myositis. Neurobiol. Aging 36, 1766. cytotoxic effector clones. Int. Immunol. 11, 229–241
& Greenberg, S. A. Abnormal distribution of e1–1766.e3 (2015). (1999).
heterogeneous nuclear ribonucleoproteins in sporadic 240. De Paepe, B. & De Bleecker, J. L. The nonnecrotic 265. Hodge, G. & Hodge, S. Steroid resistant CD8+CD28null
inclusion body myositis. Neuromuscul. Disord. 24, invaded muscle fibers of polymyositis and sporadic NKT-​like pro-​inflammatory cytotoxic cells in chronic
611–616 (2014). inclusion body myositis: on the interplay of chemokines obstructive pulmonary disease. Front. Immunol. 7,
217. Weihl, C. C. et al. TDP-43 accumulation in inclusion and stress proteins. Neurosci. Lett. 535, 18–23 617 (2016).
body myopathy muscle suggests a common pathogenic (2013). 266. Pandya, J. M. et al. Effects of conventional
mechanism with frontotemporal dementia. J. Neurol. 241. De Paepe, B., Creus, K. K. & De Bleecker, J. L. immunosuppressive treatment on CD244+(CD28null)
Neurosurg. Psychiatry 79, 1186–1189 (2008). Chemokines in idiopathic inflammatory myopathies. and FOXP3+T cells in the inflamed muscle of patients
218. Salajegheh, M. et al. Sarcoplasmic redistribution Front. Biosci. 13, 2548–2577 (2008). with polymyositis and dermatomyositis. Arthritis Res.
of nuclear TDP-43 in inclusion body myositis. 242. Ivanidze, J. et al. Inclusion body myositis: Ther. 18, 80 (2016).
Muscle Nerve 40, 19–31 (2009). laser microdissection reveals differential up-​regulation 267. Pearl, J. P. et al. Immunocompetent T cells with a
219. Nogalska, A., Terracciano, C., D’Agostino, C., of IFN-​γ signaling cascade in attacked versus memory-​like phenotype are the dominant cell type
King Engel, W. & Askanas, V. p62/SQSTM1 is nonattacked myofibers. Am. J. Pathol. 179, following antibody-​mediated T cell depletion. Am. J.
overexpressed and prominently accumulated in 1347–1359 (2011). Transplant. 5, 465–474 (2005).
inclusions of sporadic inclusion-​body myositis muscle 243. Mammen, A. L. Autoimmune myopathies. Continuum 268. Olnes, M. J. et al. Effects of systemically administered
fibers, and can help differentiating it from polymyositis 22, 1852–1870 (2016). hydrocortisone on the human immunome. Sci. Rep. 6,
and dermatomyositis. Acta Neuropathol. 118, 244. Mammen, A. L. Autoimmune myopathies: 23002 (2016).
407–413 (2009). autoantibodies, phenotypes and pathogenesis. 269. Bohan, A. & Peter, J. B. Polymyositis and
220. Hengstman, G. J. & van Engelen, B. G. Polymyositis Nat. Rev. Neurol. 7, 343–354 (2011). dermatomyositis (first of two parts). N. Engl. J. Med.
invasion of non-​necrotic muscle fibres, and the art of 245. Mammen, A. L. Which nonautoimmune myopathies 292, 344–347 (1975).
repetition. BMJ 329, 1464–1467 (2004). are most frequently misdiagnosed as myositis? 270. Shah, M. V. et al. Molecular profiling of LGL leukemia
221. Callender, L. A. et al. Human CD8+ EMRA T cells Curr. Opin. Rheumatol. 29, 618–622 (2017). reveals role of sphingolipid signaling in survival of
display a senescence-​associated secretory phenotype 246. Britson, K. A., Yang, S. Y. & Lloyd, T. E. New cytotoxic lymphocytes. Blood 112, 770–781 (2008).
regulated by p38 MAPK. Aging Cell 17, e12675 developments in the genetics of inclusion body 271. Bareau, B. et al. Analysis of a French cohort of
(2018). myositis. Curr. Rheumatol. Rep. 20, 26 (2018). patients with large granular lymphocyte leukemia:
222. Chong, L. K. et al. Proliferation and interleukin 5 247. Olive, M. et al. Expression of mutant ubiquitin (UBB+1) a report on 229 cases. Haematologica 95,
production by CD8hi CD57+ T cells. Eur. J. Immunol. and p62 in myotilinopathies and desminopathies. 1534–1541 (2010).
38, 995–1000 (2008). Neuropathol. Appl. Neurobiol. 34, 76–87 (2008). 272. Dumitriu, B. et al. Alemtuzumab in T cell large granular
223. Henson, S. M. & Akbar, A. N. KLRG1—more than a 248. Olive, M. et al. TAR DNA-​binding protein 43 lymphocytic leukaemia: interim results from a single-​
marker for T cell senescence. Age 31, 285–291 (2009). accumulation in protein aggregate myopathies. arm, open-​label, phase 2 study. Lancet Haematol. 3,
224. Akbar, A. N. & Henson, S. M. Are senescence and J. Neuropathol. Exp. Neurol. 68, 262–273 (2009). e22–e29 (2016).
exhaustion intertwined or unrelated processes that 249. Duleh, S., Wang, X., Komirenko, A. & Margeta, M. 273. Mohan, S. R. et al. Therapeutic implications of
compromise immunity? Nat. Rev. Immunol. 11, Activation of the Keap1/Nrf2 stress response pathway variable expression of CD52 on clonal cytotoxic
289–295 (2011). in autophagic vacuolar myopathies. Acta Neuropathol. T cells in CD8+large granular lymphocyte leukemia.
225. Melis, L., Van Praet, L., Pircher, H., Venken, K. Commun. 4, 115 (2016). Haematologica 94, 1407–1414 (2009).
& Elewaut, D. Senescence marker killer cell lectin-​like 250. Arahata, K. et al. Inflammatory response in 274. Gitelman, S. E. et al. Antithymocyte globulin therapy
receptor G1 (KLRG1) contributes to TNF-​α production facioscapulohumeral muscular dystrophy (FSHD): for patients with recent-​onset type 1 diabetes: 2 year
by interaction with its soluble E-​cadherin ligand in immunocytochemical and genetic analyses. results of a randomised trial. Diabetologia 59,
chronically inflamed joints. Ann. Rheum. Dis. 73, Muscle Nerve Suppl. 2, S56–S66 (1995). 1153–1161 (2016).
1223–1231 (2014). 251. Gallardo, E. et al. Inflammation in dysferlin myopathy: 275. Scarsi, M. et al. The number of circulating recent
226. Dumitriu, I. E. The life (and death) of CD4+CD28null immunohistochemical characterization of 13 patients. thymic emigrants is severely reduced 1 year after
T cells in inflammatory diseases. Immunology 146, Neurology 57, 2136–2138 (2001). a single dose of alemtuzumab in renal transplant
185–193 (2015). 252. Castets, P., Frank, S., Sinnreich, M. & Ruegg, M. A. recipients. Transpl. Int. 23, 786–795 (2010).
227. Maly, K. & Schirmer, M. The story of CD4+CD28- “Get the balance right”: pathological significance of 276. Neujahr, D. C. et al. Accelerated memory cell
T cells revisited: solved or still ongoing? J. Immunol. autophagy perturbation in neuromuscular disorders. homeostasis during T cell depletion and approaches
Res. 2015, 348746 (2015). J. Neuromuscul. Dis. 3, 127–155 (2016). to overcome it. J. Immunol. 176, 4632–4639
228. Lima, X. T. et al. Frequency and characteristics of 253. Varadhachary, A. S., Weihl, C. C. & Pestronk, A. (2006).
circulating CD4+ CD28null T cells in patients with Mitochondrial pathology in immune and inflammatory 277. Crepin, T. et al. ATG-​induced accelerated immune
psoriasis. Br. J. Dermatol. 173, 998–1005 (2015). myopathies. Curr. Opin. Rheumatol. 22, 651–657 senescence: clinical implications in renal transplant
229. Schirmer, M., Vallejo, A. N., Weyand, C. M. (2010). recipients. Am. J. Transplant. 15, 1028–1038 (2015).
& Goronzy, J. J. Resistance to apoptosis and elevated 254. Meyer, A. et al. IFN-​β-induced reactive oxygen species 278. Macedo, C. et al. Long-​term effects of alemtuzumab
expression of Bcl-2 in clonally expanded CD4+CD28- and mitochondrial damage contribute to muscle on regulatory and memory T cell subsets in kidney
T cells from rheumatoid arthritis patients. J. Immunol. impairment and inflammation maintenance in transplantation. Transplantation 93, 813–821
161, 1018–1025 (1998). dermatomyositis. Acta Neuropathol. 134, 655–666 (2012).
230. Schirmer, M. et al. Circulating cytotoxic CD8+ CD28- (2017). 279. Ramos-​Casals, M. & Brito-​Zeron, P. Emerging
T cells in ankylosing spondylitis. Arthritis Res. 4, 255. Nathan, J. A. et al. Immuno- and constitutive biological therapies in primary Sjögren’s syndrome.
71–76 (2002). proteasomes do not differ in their abilities to degrade Rheumatology 46, 1389–1396 (2007).
231. Liaskou, E. et al. Loss of CD28 expression by liver-​ ubiquitinated proteins. Cell 152, 1184–1194 (2013). 280. Lombard, M. et al. Cyclosporin A treatment in
infiltrating T cells contributes to pathogenesis of 256. Seifert, U. et al. Immunoproteasomes preserve protein primary biliary cirrhosis: results of a long-​term
primary sclerosing cholangitis. Gastroenterology 147, homeostasis upon interferon-​induced oxidative stress. placebo controlled trial. Gastroenterology 104,
221–232 (2014). Cell 142, 613–624 (2010). 519–526 (1993).
232. Dejaco, C. et al. NKG2D stimulated T cell autoreactivity 257. Nagaraju, K. et al. Activation of the endoplasmic 281. Mitchison, H. C. et al. A pilot, double-​blind, controlled
in giant cell arteritis and polymyalgia rheumatica. reticulum stress response in autoimmune myositis: 1-year trial of prednisolone treatment in primary
Ann. Rheum. Dis. 72, 1852–1859 (2013). potential role in muscle fiber damage and dysfunction. biliary cirrhosis: hepatic improvement but greater
233. Dejaco, C., Duftner, C., Klauser, A. & Schirmer, M. Arthritis Rheum. 52, 1824–1835 (2005). bone loss. Hepatology 10, 420–429 (1989).
Altered T cell subtypes in spondyloarthritis, 258. Correia, A. S., Patel, P., Dutta, K. & Julien, J. P. 282. Wiesner, R. H. et al. A controlled trial of cyclosporine
rheumatoid arthritis and polymyalgia rheumatica. Inflammation induces TDP-43 mislocalization and in the treatment of primary biliary cirrhosis. N. Engl.
Rheumatol. Int. 30, 297–303 (2010). aggregation. PLOS ONE 10, e0140248 (2015). J. Med. 322, 1419–1424 (1990).

Nature Reviews | Rheumatology


Reviews

283. Fujihara, T. et al. Preferential localization of 293. Sasaki, M., Miyakoshi, M., Sato, Y. & Nakanuma, Y. 303. US National Library of Medicine. ClinicalTrials.gov
CD8+αEβ7+T cells around acinar epithelial cells with Autophagy mediates the process of cellular senescence http://www.clinicaltrials.gov/ct2/show/NCT00917956
apoptosis in patients with Sjögren’s syndrome. characterizing bile duct damages in primary biliary (2010).
J. Immunol. 163, 2226–2235 (1999). cirrhosis. Lab. Invest. 90, 835–843 (2010). 304. US National Library of Medicine. ClinicalTrials.gov
284. Kita, H. Autoreactive CD8-specific T cell response in 294. Sasaki, M., Yoshimura-​Miyakoshi, M., Sato, Y. http://www.clinicaltrials.gov/ct2/show/NCT01519349
primary biliary cirrhosis. Hepatol. Res. 37 (Suppl. 3), & Nakanuma, Y. A possible involvement of (2017).
402–405 (2007). endoplasmic reticulum stress in biliary epithelial 305. US National Library of Medicine. ClinicalTrials.gov
285. Si, L., Whiteside, T. L., Schade, R. R., Starzl, T. E. autophagy and senescence in primary biliary cirrhosis. http://www.clinicaltrials.gov/ct2/show/NCT02483845
& Van Thiel, D. H. T-​Lymphocyte subsets in liver J. Gastroenterol. 50, 984–995 (2015). (2017).
tissues of patients with primary biliary cirrhosis (PBC), 295. Katsiougiannis, S., Tenta, R. & Skopouli, F. N. 306. US National Library of Medicine. ClinicalTrials.gov
patients with primary sclerosing cholangitis (PSC), and Endoplasmic reticulum stress causes autophagy http://www.clinicaltrials.gov/ct2/show/NCT02250443
normal controls. J. Clin. Immunol. 4, 262–272 (1984). and apoptosis leading to cellular redistribution (2018).
286. Bjorkland, A. et al. Blood and liver-​infiltrating of the autoantigens Ro/Sjögren’s syndrome-​related 307. US National Library of Medicine. ClinicalTrials.gov
lymphocytes in primary biliary cirrhosis: increase in antigen A (SSA) and La/SSB in salivary gland epithelial http://www.clinicaltrials.gov/ct2/show/NCT00802815
activated T and natural killer cells and recruitment of cells. Clin. Exp. Immunol. 181, 244–252 (2015). (2014).
primed memory T cells. Hepatology 13, 1106–1111 296. Leff, R. L., Miller, F. W., Hicks, J., Fraser, D. D. & 308. US National Library of Medicine. ClinicalTrials.gov
(1991). Plotz, P. H. The treatment of inclusion body myositis: http://www.clinicaltrials.gov/ct2/show/NCT00769860
287. Tasaki, S. et al. Multiomic disease signatures converge a retrospective review and a randomized, prospective (2017).
to cytotoxic CD8 T cells in primary Sjögren’s syndrome. trial of immunosuppressive therapy. Medicine 72, 309. US National Library of Medicine. ClinicalTrials.gov
Ann. Rheum. Dis. 76, 1458–1466 (2017). 225–235 (1993). http://www.clinicaltrials.gov/ct2/show/NCT01423110
288. Tsuda, M. et al. Fine phenotypic and functional 297. Soueidan, S. A. & Dalakas, M. C. Treatment of (2017).
characterization of effector cluster of differentiation inclusion-​body myositis with high-​dose intravenous 310. US National Library of Medicine. ClinicalTrials.gov
8 positive T cells in human patients with primary immunoglobulin. Neurology 43, 876–879 (1993). http://www.clinicaltrials.gov/ct2/show/NCT01925209
biliary cirrhosis. Hepatology 54, 1293–1302 (2011). 298. Amato, A. A. et al. Inclusion body myositis: treatment (2017).
289. Yang, Z., Goronzy, J. J. & Weyand, C. M. Autophagy with intravenous immunoglobulin. Neurology 44, 311. US National Library of Medicine. ClinicalTrials.gov
in autoimmune disease. J. Mol. Med. 93, 707–717 1516–1518 (1994). http://www.clinicaltrials.gov/ct2/show/NCT02481453
(2015). 299. Barohn, R. J., Amato, A. A., Sahenk, Z., Kissel, J. T. (2019).
290. Hosomi, S., Kaser, A. & Blumberg, R. S. Role of & Mendell, J. R. Inclusion body myositis: explanation 312. US National Library of Medicine. ClinicalTrials.gov
endoplasmic reticulum stress and autophagy as for poor response to immunosuppressive therapy. http://www.clinicaltrials.gov/ct2/show/NCT02753530
interlinking pathways in the pathogenesis of Neurology 45, 1302–1304 (1995). (2018).
inflammatory bowel disease. Curr. Opin. 300. Barohn, R. J. et al. Pilot trial of etanercept in the
Gastroenterol. 31, 81–88 (2015). treatment of inclusion-​body myositis. Neurology 66,
Competing interests
291. Grootjans, J., Kaser, A., Kaufman, R. J. & Blumberg, R. S. S123–S124 (2006).
S.A.G. is an inventor of intellectual property related to myosi-
The unfolded protein response in immunity and 301. Kosmidis, M. L., Alexopoulos, H., Tzioufas, A. G. &
tis diagnostics and therapeutics, owned and managed by
inflammation. Nat. Rev. Immunol. 16, 469–484 Dalakas, M. C. The effect of anakinra, an IL1 receptor
Brigham and Women’s Hospital; he receives sponsored
(2016). antagonist, in patients with sporadic inclusion body
research from Pfizer, Inc. and is a founder of Abcuro, Inc.
292. Sasaki, M., Miyakoshi, M., Sato, Y. & Nakanuma, Y. A myositis (sIBM): a small pilot study. J. Neurol. Sci. 334,
possible involvement of p62/sequestosome-1 in the 123–125 (2013).
process of biliary epithelial autophagy and senescence 302. US National Library of Medicine. ClinicalTrials.gov Publisher’s note
in primary biliary cirrhosis. Liver Int. 32, 487–499 http://www.clinicaltrials.gov/ct2/show/NCT00079768 Springer Nature remains neutral with regard to jurisdictional
(2012). (2010). claims in published maps and institutional affiliations.

www.nature.com/nrrheum

You might also like