You are on page 1of 13

Clinic Rev Allerg Immunol

DOI 10.1007/s12016-015-8513-8

The Clinical Features of Myositis-Associated


Autoantibodies: a Review
Harsha Gunawardena 1

# Springer Science+Business Media New York 2015

Abstract The idiopathic inflammatory myopathies (IIM) are a range of clinical manifestations including interstitial pneu-
a group of autoimmune diseases traditionally defined by clin- monia, cutaneous disease, cancer-associated myositis and
ical manifestations including skeletal muscle weakness, skin autoimmune-mediated necrotising myopathy. This review de-
rashes, elevated skeletal muscle enzymes, and neurophysio- scribes their diagnostic utility, potential role in disease moni-
logical and/or histological evidence of muscle inflammation. toring and response to treatment. In the future, routine use of
Patients with myositis overlap can develop other features in- these autoAb will allow a stratified approach to managing this
cluding parenchymal lung disease, inflammatory arthritis, complex set of conditions.
gastrointestinal manifestations and marked constitutional
symptoms. Although patients may be diagnosed as having Keywords Myopathy . Polymyositis . Dermatomyositis .
polymyositis (PM) or dermatomyositis (DM) under the IIM Systemic sclerosis . Interstitial lung disease . Autoantibodies
spectrum, it is quite clear that disease course between sub-
groups of patients is different. For example, interstitial lung
disease may predominate in some, whereas cutaneous compli- Introduction
cations, cancer risk, or severe refractory myopathy may be a
significant feature in others. Therefore, tools that facilitate Historically, patients presenting with symmetrical proximal
diagnosis and indicate which patients require more detailed muscle weakness, elevated skeletal muscle enzymes, skin le-
investigation for disease complications are invaluable in clin- sions, neurophysiological and/or histological evidence of
ical practice. The expanding field of autoantibodies (autoAbs) muscle inflammation were simply diagnosed as having poly-
associated with connective tissue disease (CTD)-myositis myositis (PM) or dermatomyositis (DM). This was based on
overlap has generated considerable interest over the last few hallmark clinical and pathological classification criteria de-
years. Using an immunological diagnostic approach, this scribed some 40 years ago [1, 2], which we now recognise,
group of heterogeneous conditions can be separated into a and does not highlight the heterogeneous nature of these com-
number of distinct clinical phenotypes. Rather than diagnose plex diseases. Further criteria published in 1995 did describe
a patient as simply having PM, DM or overlap CTD, we can additional clinical features and were the first to include the
define syndromes to differentiate disease subsets that empha- concept of myositis autoAbs; however, the number of sero-
sise clinical outcomes and guide management. There are now logical markers identified at the time was limited [3].
over 15 CTD-myositis overlap autoAbs found in patients with The diagnosis of CTD-myositis overlap remains primarily
a clinical one. Patients have shared clinical features including
muscle inflammation and weakness, skin rashes, fever and
fatigue. However, in some adult and juvenile cases, presenta-
* Harsha Gunawardena tion and disease course is variable with different manifesta-
harsha.gunawardena@nbt.nhs.uk tions. Some have severe muscle disease at onset, whereas
some are amyopathic. Some are more at risk of lung disease
1
Clinical and Academic Rheumatology, North Bristol NHS Trust, or inflammatory arthritis, in contrast to those who have more
Southmead Hospital, Bristol BS10 5NB, UK skin disease with complications including ulceration or
Clinic Rev Allerg Immunol

calcinosis or cancer-associated myositis (CAM). Therefore, [6–9]. A number of research groups are now producing
we should no longer purely diagnose a patient with PM or ELISA and line-blot testing for some of the novel autoAb
DM but concentrate on defining autoAbs syndromes. In some including the rarer non-Jo1 anti-synthetases, anti-MDA5, an-
cases, this may avoid the need for a muscle biopsy where ti-TIF1g, anti-NXP2 and anti-HMGCR [6, 8, 10–18]. Cross
findings can be non-specific and thus lead to diagnostic delay. collaboration is required to ensure methods are reproducible
Moreover, adopting this approach can determine which pa- and validated in large patient cohorts, so that in the near future,
tients may require more detailed investigation for cancer or we can routinely test for all CTD-myositis overlap antibodies
lung disease, as well as potentially guide treatment. The aim of in a clinical setting.
this review is to highlight to clinicians looking these patients
that now well-defined autoAbs within this disease spectrum
should routinely be part of the diagnostic assessment and used Terminology
as prognostic tools [4, 5].
We should move away from the concept of separating ‘myo-
sitis-specific and myositis-associated’ antibodies (MSAs /
Methodology MAAs), primarily because some patients within this spectrum
who are found to be positive for a so-called MSAs may never
Anti-nuclear and anti-cytoplasmic autoantibodies (ANA/ actually develop myositis. It is clear that some of these
ACA) are the serological hallmark of systemic autoimmune autoAbs are also found in dermato-pulmonary syndromes
CTD that are used in conjunction with clinical features to and patients with overlap systemic sclerosis (SSc) [11,
confirm an early diagnosis. However, in patients with CTD- 19–21]. Therefore, to highlight to the clinician that these pa-
myositis overlap, standard ANA testing with standard extract- tients can present in different ways to a number of different
able nuclear antigen (ENA) panels may be uninformative, specialties, we should consider alternative nomenclature such
unless the patient is for example positive for anti-Jo-1 or as ‘CTD-myositis overlap’ and/or ILD autoAbs.
anti-U1RNP autoAbs. A ‘negative’ ANA should not deter
further serological testing in myositis patients or those with
interstitial lung disease (ILD) with other features such as Clinical Manifestations and AutoAbs Groups (Fig. 1)
Raynaud’s or arthropathy. Therefore, identifying specific
autoAbs requires a diagnostic two-step strategy. Screening Interstitial Lung Disease and Anti-Cytoplasmic AutoAbs
using indirect immunofluorescence (IIF) on human epithelial
cell lines (HEp-2) remains the best means of ANA subtyping. Diffuse parenchymal lung disease, in particular ILD, now
A key question to the clinician when approaching this disease termed as interstitial pneumonia is a major clinical manifesta-
group should be is the ANA really negative or is it just low tion in CTD-myositis patients. A number of radiological and
titre (e.g. 1/40 or 1/80), and what is the staining pattern. In histopathological subtypes are recognised, with non-specific
general, a nucleolar or cytoplasmic speckle coupled alongside interstitial pneumonia (NSIP) the most frequently observed,
specific clinical manifestations can provide insight into the but also organising pneumonia (OP), usual interstitial pneu-
target autoantigen, for example the aminoacyl tRNA synthe- monia (UIP) and rarely acute interstitial pneumonia (AIP/dif-
tase group, signal recognition particle (SRP), hydroxy-3- fuse alveolar damage) [6, 22, 23]. Traditional risk factors in-
methylglutaryl-coenzyme A reductase (HMGCR) (all of clude ethnicity, those with amyopathic disease, systemic fea-
which are cytoplasmic autoantigens), or PM-Scl 100/75 com- tures with fever, and/or a marked acute phase response at
plex and U3-ribonucleoprotein (U3RNP) (the latter both nu- presentation. Moreover, it is apparent in this group of patients
cleolar autoantigens). IIF of other DM-associated autoAbs is that lung disease can pre-date the development of myositis and
usually non-specific and low titre low (and may be reported as be the predominant manifestation [23]. Sub-dividing patients
negative), but this should not preclude further serological in- by autoAb syndrome will aid identification further with anti-
terrogation, as it is clear that these are not seronegative condi- synthetase syndrome (ASS), overlap SSc-myositis syndromes
tions [4, 5]. Therefore, more specific confirmatory methods and the novel anti-MDA5 sub-group being at greatest risk.
are required, such as immunoprecipitation (IPP), enzyme-
linked immunosorbent assay (ELISA) or immunoblotting. Anti-Synthetase Syndrome
Although IPP is an optimal research method to detect CTD-
myositis autoAbs [4, 6], in routine clinical practice, it is not There are currently eight identified autoAb that target the
practical. There is now considerable focus to develop com- amino-acyl tRNA synthetase enzymes (ARS), anti- Jo-1,
mercially available tests and ELISAs, immunoblotting and PL12, PL7, EJ, OJ, KS, Ha and Zo, which define ASS that
line or bead immunoassays that can test for the majority of was first described in 1990 [24]. Classic clinical features in-
anti-synthetase group, anti-PM-Scl, anti-SRP and anti-Mi2 clude myositis, non-erosive inflammatory arthritis, cutaneous
Clinic Rev Allerg Immunol

Fig. 1 CTD-myositis overlap—myositis-associated autoantigens. *Aminoacyl tRNA synthetases—Jo1, PL7, PL12, OJ, KS, EJ, Zo, Ha. ILD interstitial
lung disease, RP-ILD rapidly progressive interstitial lung disease

lesions (including mechanic’s hands), fever, Raynaud’s phe- pneumonia and arthropathy to identify patients and thus not
nomenon and interstitial pneumonia. In comparison to adult delay diagnosis. This is particularly important in lung disease
patients, it is important to note that ASS is a relatively uncom- patients, where early intervention with immunomodulatory
mon subtype within the juvenile myositis spectrum. It is in- therapy may well confer a better prognosis in comparison
creasingly evident that ASS is a spectrum disorder that not just idiopathic forms of the disease.
includes myositis. However, sub-phenotyping the syndrome is
difficult due to the relative rarity of non-Jo1 anti-ARS com- Anti-MDA5 AutoAb—a Distinct Cutaneous-Pulmonary
pared to anti-Jo1, although this may be more to do with lim- Syndrome
itations in diagnostic testing [25, 26]. It is possible that the
prevalence of the non-Jo1 ARS group is higher in different Recent literature has highlighted the clinical significance of
cohorts such as patients labelled as ‘idiopathic’ IP, particularly anti-MDA5 autoAb, not just in the field of myositis but also
in those with UIP and AIP (DAD) subtypes [27]. Data from perhaps more importantly in respiratory and dermatology
several studies suggest that UIP and AIP are commonly seen clinics. AutoAb targeting a cytoplasmic protein, first termed
in non-Jo1 ASS, in particular anti-EJ, anti-PL7 and anti-PL12 CADM-140 and subsequently identified as melanoma differ-
patients, where lung disease presents early in disease course entiation gene 5, was first described in Asian cohorts and more
and is the predominant manifestation [22, 27–32]. In a large recently in US and European adult and juvenile studies [11,
series of Japanese patients positive for anti-Jo-1, anti-EJ, anti- 35–39]. Anti-MDA5 autoAb defines a specific dermato-
PL-7, anti-PL-12, anti-KS or anti-OJ highlighted the degree of pulmonary syndrome, but within this, it is evident that Asian
heterogeneity within ASS in terms of distribution and onset of and Caucasian patients differ implying genetic and environ-
myositis, lung disease and skin lesions [33]. Furthermore, mental factors influence the phenotype. Asian patients typi-
non-erosive inflammatory arthritis may be the presenting clin- cally present with clinically amyopathic DM (CADM), sys-
ical feature in around one third of ASS patients, with or temic features including fever and most importantly rapidly
followed by ILD [34]. It now seems clear that as we will progressive interstitial lung disease (RP-ILD), which is fre-
evaluate the degree of myositis and dermatological findings, quently fatal [11, 35]. Markers of severe lung damage in this
we need to concentrate on different presentations of interstitial group of patients include hyper-ferritinaemia, serum IL-18
Clinic Rev Allerg Immunol

concentrations and anti-MDA5 autoAb titres [40, 41]. In US digital ulceration appears less common. Skeletal muscle
cohorts, ILD again is a predominant feature, but RP-ILD, a weakness has been a consistent observed association but can
less prevalent pattern with other subtypes of interstitial pneu- be subclinical with features on electromyography or MRI im-
monia, is also seen [36]. In addition, anti-MDA5 patients who aging. Other subcutaneous ± cutaneous features include ‘puffy
present with co-existent myositis, as opposed to CADM, may swollen fingers’, nailfold capillary distortion, calcinosis and
also have less severe lung fibrosis, typically NSIP disease that hyperkeratosis with fissuring of the skin on the digits, charac-
responds well to treatment (personal data). This group of pa- teristic of mechanic’s hands. The frequency of ILD is similar
tients has been described as having a novel dermatological to that seen in ASS [49], although OP and milder forms of
phenotype representative of severe vasculopathy, including NSIP may predominate in anti-PM-Scl phenotype that re-
(muco)-cutaneous ulceration with oral mucosal pain, tender sponds well to immunomodulatory treatment (personal obser-
palmar papules around the nailfolds and Gottron’s lesions vation). In addition, forced vital capacity (FVC%) at presen-
[36, 42]. Another US study has widened the ‘MDA5’ spec- tation and improvement during disease course tends to be
trum with phenotypic similarities to ASS, including symmet- higher in this sub-group. This may partly explain why several
rical inflammatory arthritis, some with overt clinical myopa- studies have shown higher progression-free survival in this
thy, Raynaud’s, fever and mechanic’s hands, as well as ILD group of patients [21, 47, 48, 50, 51].
[43]. This observation is intriguing as both syndromes are
associated with cytoplasmic autoantigens with similar biolog- Anti-U3RNP
ical functions, where anti-Ro52 autoAb is also often detected.
It is significant to note that anti-MDA5 is a major autoAb in Antibodies against U3RNP complex, of which fibrillarin is the
JDM, found in over one third of Japanese children and around autoantigenic subunit, are more commonly seen in diffuse
7 % of a UK cohort. Clinical findings are similar to adult cutaneous SSc overlap patients of African-American race
counterparts with characteristic cutaneous lesions including [21, 46]. Patients may present younger and disease course
ulcerations, arthritis, and milder muscle or amyopathic dis- more progressive. Myositis is a consistent feature with also a
ease. Both chronic ILD and RP-ILD are prevalent especially risk of other internal organ involvement including renal, car-
the latter in the Asian population, making anti-MDA5 an im- diac, gastro-intestinal disease and parenchymal lung disease
portant serological marker in children, as parenchymal lung [21, 46, 52, 53]. A recent study has shown that ILD occurs in
disease is generally uncommon in JDM [37, 44, 45]. around one third of anti-U3RNP patients, with a similar fre-
quency of intrinsic pulmonary hypertension (i.e. primary vas-
culopathy unrelated to lung disease), the latter being the major
Systemic Sclerosis-Myositis Overlap poor prognostic determinant [46].
and Anti-Nucleolar/Anti-Nuclear AutoAb
Anti-RuvBL1/Anti-RuvBL2
Anti-nucleolar autoAb (AnuAb) are found in 10–15 % of SSc
patients, particularly those with overlap syndromes [21, 46]. A novel autoantigen target RuvBL1/RuvBL2 (a nuclear/
To date, four specificities, anti-RNAPol3, anti-Th/To, anti- nucleolar matrix complex) has been recently identified in
P M - S c l a n d a n t i - U 3 R N P, h a v e b e e n i d e n t i f i e d . SSc patients with myositis in a combined Japanese and US
Characterisation of AnuAb can guide the clinician and help cohort. Anti-RuvBL1/anti-RuvBL2 autoAb was detected
define specific clinical manifestations and in some cases prog- ∼2 % of SSc patients, where the frequency of ILD appears
nosis, for example anti-RNAPol3 is a strong predictor of SSc similar to anti-PM-Scl patients (just over 50 % of patients in
renal crisis. In the context of CTD-myositis overlap, autoAb both autoAb subgroups). However, anti-RuvBL1/2 patients
against PM-Scl protein complex and U3RNP is the most rel- appear unique in that they are older at disease onset, more
evant, with a higher frequency of parenchymal or vascular likely to be male and have diffuse skin thickening [54]. It will
lung involvement [21, 46–48]. be interesting to see whether this finding is replicated in other
cohorts and whether pooled data can then identify specific
PM-Scl Overlap Syndrome subtypes of lung disease, which may the determine prognosis.

Antibodies directed against the PM-Scl complex in the human


exosome identify a distinct subset of patients with SSc and/or Anti-Nuclear AutoAb in Cancer-Associated DM,
myositis overlap [21, 46–48]. Studies highlight how anti-PM- Skin and Subcutaneous Disease
Scl patients tend to be younger at disease onset and have
milder cutaneous SSc features with limited skin disease and Four specific nuclear proteins, namely transcription interme-
in some inflammatory arthropathy [46–48]. Although diary factor 1 complex (TIF1), nuclear matrix protein 2
Raynaud’s is a common feature, peripheral vasculopathy with (NXP2), small ubiquitin activating enzyme (SAE) and Mi2,
Clinic Rev Allerg Immunol

have been identified as important autoantigens in the adult and seen in other clinico-serological subgroups such as
juvenile DM spectrum [4, 5]. It is of interest that these nuclear Raynaud’s, arthritis and ILD were uncommon [63].
proteins have analogous cellular functions including gene tran- AutoAb recognising NXP2 has now been shown to be
scription, DNA synthesis and cellular repair suggesting a shared another frequent and important group in adult DM. A
pathogenic mechanism, despite variations in clinical presentation. combined US cohort study detected anti-NXP2 in 17 %
and anti-TIF1 in 38 % patients, respectively, with a higher
Anti-TIF1 and Anti-NXP2 AutoAb frequency of the latter compared to older studies possibly
explained by their own higher specificity IPP assay using
The potential relationship between cancer and autoimmu- cell lysates transfected with TIF1-γ [64]. Interestingly,
nity has been interrogated for decades. The idea that anti- compared to previous work, the risk of cancer with anti-
tumour immune responses may attempt to regulate the TIF1-γ was not as strong, and the authors suggest that
emergence of cancer, leading to the over-expression of this might be due to the higher specificity assay identify-
autoantigens, autoAb production and bystander tissue in- ing patients with lower positive titres combined with the
jury is an intriguing theory. The increased relative risk of overall cohort having a low frequency of cancer (one third
malignancy and dermatomyositis compared to age- and of patients included had follow-up of <3 years). This
sex-matched controls, alongside a clear temporal relation- study did demonstrate a cancer risk with anti-NXP2 pos-
ship between cancer and CTD onset, emphasises the con- itivity, particularly amongst male patients (not explained
nection [55]. However, because of the previously small by just male-associated tumours). This observation was
number of myositis-associated autoAb described, histori- previously suggested in a smaller Japanese series [65]. It
cally cancer-associated DM was felt to be a ‘seronegative’ is noteworthy that Fiorentino and colleagues conclude in
subset. Indeed, if one screens myositis-overlap patients by DM patients under the age of 60 who are antiTIF1-γ or
‘routine’ standard hospital laboratory assays, autoAb- anti-NXP2 negative have a very low risk of cancer [64].
positive patients have a lower risk of cancer compared Apart from cancer, both anti-TIF1 and anti-NXP2 define
to those who are autoAb negative [56]. Over the last their own characteristic phenotypes, highlighted by the
few years, it is now evident that cancer-associated DM finding that they form the main serological groups in
is an antibody-rich rheumatic phenotype with the discov- JDM. Collectively, both autoAb are detected in ∼50 %
ery of anti-TIF1 and more recently anti-NXP2 autoAb. of JDM cases, which is important because previously,
Anti-TIF1 has now been described in around 15–25 % these patients were classified as seronegative. In children,
of DM cohort studies [57–60]. The identity of the 155/ anti-TIF1 positivity has been shown to be associated with
140 kDa protein was subsequently identified as the TIF1 more extensive skin disease (similar to adult counter-
(α, β, γ subunits) family proteins, with TIF1-γ the main parts), cutaneous oedema, vasculopathy with ulcers,
target [60]. The incidence of cancer in anti-TIF1 adult lipoatrophy and limb contracture [66–68]. In contrast,
DM patients is high in comparison to all other myositis- anti-NXP in JDM appears to be a clear serological marker
associated autoAb groups. A meta-analysis of six studies for calcinosis, associated with a greater degree of muscle
using the same methodology to detect anti-TIF1 has weakness, as well as younger age of disease onset [69,
shown a high negative predictive value and significant 70]. Other disease associations include muscle contrac-
diagnostic odds ratio of 27 for cancer-associated DM tures, atrophy and worse function status [68]. Data sug-
[61]. In clinical practice, once routine testing for anti- gests that anti-NXP2 is also associated with calcinosis in
TIF1 autoAb becomes available, we will be able to iden- adult DM (personal observation) [71–73]. In a recent
tify those DM patients at particular risk of occult malig- study, 11 % had calcinosis overall, associated indepen-
nancy and focus on early detection with imaging tech- dently with disease duration, fingertip ulcers, as well as
niques such as FDG positron emission tomography-CT anti-NXP2 [71].
[62]. A recent US study has highlighted that adult anti- Returning to the potential link between autoimmunity and
TIF1 patients have another unique skin phenotype, which cancer, the observation that both anti-TIF1 and antiNXP2 de-
is of significant interest because this identifies additional fine similar cutaneous and subcutaneous subtypes in adult and
risk factors for cancer-associated DM when autoAb test- juvenile DM, but only cancer in adults (and in general older
ing is not available. Patients were shown to have more adults) is tantalising. This perhaps strengthens the hypothesis
severe skin disease in terms of distribution (scalp, face, that abnormal cancer cellular responses initiate disease in this
V-sign and shawl-sign) with palmar hyperkeratotic group of patients regardless of age. A subsequent anti-cancer
Gottron’s papules, ‘psoriatic’ looking lesions and immune response leads to over-expression of particular anti-
hypopigmented patches with punctate telangiectasia or er- gens and then tissue damage [74]. Perhaps in children and
ythematous macules (termed by the authors as ‘red on younger adults with a more efficient immune system, the can-
white lesions’). Common myositis-overlap manifestations cer is abolished, whereas in some, it still emerges alongside
Clinic Rev Allerg Immunol

autoAb production (namely anti-TIF1 and NXP2) leading to into the endoplasmic reticulum [85, 86]. Despite the relative
the emergence of DM. absence of inflammatory infiltrate, the predominance of
myofibre necrosis and in the case of anti-HMG-CoA where
Anti-SAE discontinuation of statins does not result in clinical improve-
ment, AIMN responds to immunotherapy again highlighting
AutoAb against small ubiquitin-like modifier activating en- that this is an immune-mediated phenotype.
zyme (SAE) has now been described in a number of adult
DM cohorts with varying frequency of 1.5–8 % [75–79]. Anti-SRP AINM
Patients tend to present with hallmark skin disease first
(amyopathic DM) and then progress to muscle weakness with Patients with anti-SRP AINM typically present with acute/
systemic features including in some marked gastro-intestinal subacute onset proximal weakness, associated with high CK
involvement with dysphagia [75, 78]. It is notable that the levels (often over ten times the upper limit of normal) that
frequency of anti-SAE DM patients in Japanese studies in may progress to severe disability. In addition, dysphagia
comparison to European cohorts is low. Although numbers secondary to pharyngeal muscle weakness combined with
are small, certain clinical differences are described including respiratory muscle weakness increases the risk of aspiration
more ILD, which may reflect ethnic and environmental differ- in these patients. A number of studies have described certain
ences [79]. When combining studies, anti-SAE does not ap- extra-muscular manifestations including parenchymal lung
pear to be a significant clinico-serological phenotype in JDM, disease and cardiac involvement [87, 88], but this does not
seen in <1 %. appear to be a consistent feature [89, 90]. Subclinical myo-
carditis may explain why some patients develop palpitations,
Anti-Mi-2 paroxysmal arrhythmias and raised cardiac-specific Troponin
levels, regardless of cardiac MRI findings (personal obser-
Anti-Mi-2 was the first specific serological marker to be de- vation). The key in anti-SRP patients is to intervene early,
scribed in DM [80]. AutoAb recognising this nucleosome recognise that some patients may be refractory to standard
helicase protein is commonly found in up to 20 % of adult treatments and adopt more intensive management strategies
DM patients and up to 10 % of JDM. Patients present with [4, 9, 91, 92].
classic skin eruptions particularly in sun-exposed areas along-
side myositis, and it has been shown that surface ultra-violet Anti-HMGCR Statin-Associated AINM
(UV) irradiation intensity correlates with phenotype inci-
dence, particularly in women [81, 82]. However, a recent The classification of the statin-induced myopathy spec-
study suggests other factors other than UV influence the de- trum includes statin-induced myalgia (usually self-
velopment of anti-Mi-2 DM, where autoAb frequency was limiting muscle symptoms without CK rise), statin-
significantly different in two Mexican cohorts, with similar induced myositis and statin-induced rhabdomyolysis
UV exposures [83]. Both adults and children with anti-Mi-2 [93]. Many patients report self-limiting symptoms on
DM tend to have mild to moderate myositis and have less discontinuation of statin therapy, and some in the case
systemic disease with a low frequency of lung and joint dis- of statin rhabdomyolysis may have an underlying meta-
ease [84]. In addition, patients respond well to corticosteroid bolic myopathy. However, a small number of patients
therapy and often run a monophasic disease course, with good develop progressive weakness and persistently elevated
outcomes. CK. A number of studies have now shown that this
latter phenotype forms a distinct entity outside the ‘clas-
sic’ toxic stain myopathy spectrum characterised by
Autoimmune Necrotising Myopathy autoAb against the enzyme target of statins HMGCR.
Muscle biopsy changes are characteristic of an AINM
Autoimmune necrotising myopathy (AINM) describes a sub- i.e. myonecrosis with little inflammation with also MHC
group of IIM with similar clinical, biochemical and histopath- class 1 up-regulation and MAC deposition on non-
ological features. It is clear that seropositive AIMN is a distinct necrotic fibres. Expression of HMGCR is also upregu-
entity to what we would have previously termed classic PM. lated in regenerating muscle fibres [16, 94, 95]. In ad-
Anti-SRP and anti-HMGCR autoAbs are found in patients who dition, the highest risk for anti-HMGCR statin-induced
present predominately with acute severe muscle disease, mir- AINM has been shown in HLA-DR11 carriers (HLA
rored by CK levels, and unlike other myositis autoAb, subtypes DRB1*11:01 allele) [95, 96], in comparison to the sin-
have few overlap features. In this context, it is noteworthy that gle nucleotide polymorphism in SLCO1B1, a genetic
both target autoantigens are found in the cytoplasm and studies risk factor seen in ‘standard’ statin toxic myositis [97].
have shown that SRP is required for the insertion of HMGCR It is notable that anti-HMGCR AINM is also seen in
Clinic Rev Allerg Immunol

patients with no prior history of statin exposure [18, ASS, this may be due to delays in diagnosis with patients
95]. Therefore, the finding of anti-HMGCR autoAb-as- mislabelled as idiopathic pulmonary fibrosis and/or more
sociated myositis should signify to the clinician that extensive parenchymal lung involvement with coarse
patients require treatment with steroids and at least fixed fibrosis at presentation (personal observation) [26,
one other immunomodulatory agent with most showing 27]. In anti-MDA5 syndrome, we know that in specific
either partial or full response [94, 98, 99]. A number of cohorts, the occurrence of RP-ILD influences outcome
questions remain; are anti-HMGCR autoAb detected in [11, 35, 40, 41, 104], with around 50 % mortality rate
‘normal’ subjects, if they are will the introduction of a within 6 months due to respiratory failure [104]. In the
statins the trigger AINM, and can treated patients be case of other subsets, progressive muscle weakness in
rechallenged with an alternative statin. refractory seropositive AINM and cutaneous complica-
tions in certain DM phenotypes influences morbidity and
therefore management. However, due to heterogeneity of
Autoantibody Titres and Disease Activity CTD-myositis overlap, there are no standardised treatment
approaches. Therapeutic strategies are essentially based
With the appreciation that myositis-CTD overlap autoAbs on ‘best’ clinical practice and a limited number of case
are useful diagnostically and define clinical phenotypes, series, open-label studies or randomised controlled trials
there is a real focus to develop serological techniques that (RCTs) [105]. Alongside corticosteroids, a number of
can be used in routine clinical practice. Commercially treatment options are used in ASS and anti-MDA5 lung
available ELISA kits are available for a number of disease including induction cyclophosphamide, mycophe-
autoAb including most anti-ARS, nucleolar SSc-myositis nolate mofetil (MMF) and calcineurin inhibitors. Use of
ANAs, anti-Mi2 and anti-SRP. In addition, a number of the former two agents is essentially based on clinical best
research groups have developed ELISAs using highly pu- practice using experience adopted in other conditions such
rified recombinant proteins to detect the more novel as systemic sclerosis, as well as retrospective observation-
MAAs, namely the rarer anti-ARS, anti-MDA5, anti- al studies [106–108]. A number of studies have suggested
TIF1, anti-NXP2 and anti-HMGCR [9–18]. The advan- a potential role for calcineurin inhibitors, especially tacro-
tage of ELISA detection is higher sensitivity and efficien- limus as a first-line and rescue treatment in ILD-
cy with rapid turnover and results. Moreover, quantitative associated ASS [109–112]. Similar regimes have been de-
techniques where autoAb titres can be measured may be scribed in amyopathic DM RP-ILD, including efficacy of
useful in monitoring disease activity. A number of recent combination therapy with cyclosporine and cyclophospha-
studies have highlighted how myositis-CTD overlap mide [112–115]. Additional second-line treatments for
autoAb titres may be useful in this setting. Anti-Jo1, muscle, skin and subcutaneous disease in other pheno-
anti-SRP and anti-HMGCR titres have all been shown to types include methotrexate, azathioprine and MMF, and
correlate with CK levels and myositis scores [9, 99, 100]. in refractory cases intravenous immunoglobulin. The use
In the case of cancer-associated DM, higher anti-TIF1 of rituximab in myositis overlap syndromes has generated
levels at baseline are associated with the presence of can- considerable interest over the last few years, particularly
cer [12]. In terms of lung and skin disease, the diagnostic in those with refractory lung and muscle disease. Prior to
utility of anti-MDA5 titres has been highlighted by sever- the RIM study (rituximab in refractory adult and juvenile
al studies, where levels at baseline and in response to myositis), a number of retrospective series have suggested
treatment predict outcomes of rapidly progressive ILD a potential benefit [116–120]. The RIM study did not
and severity of skin ulceration. Importantly, high titres demonstrate significant differences in primary or second-
despite treatment indicate refractory lung disease with in- ary end-points between treatment groups, but the majority
creased risk of mortality [101, 102]. with refractory disease met the study definition of im-
provement [121]. Moreover, a subsequent subgroup anal-
ysis has highlighted that response to rituximab may be
Does Clinico-Serological Phenotype Influence influenced by serological phenotype with ASS and anti-
Outcome and Treatment Approaches? Mi2 patients more likely to gain benefit [122]. A thera-
peutic role for rituximab within the different clinico-
Over the last few years, a number of studies have serological phenotypes has been further emphasised in a
highlighted that different treatment responses and thus number of series, including refractory patients with ASS-
prognosis are seen depending on serological profile. For ILD, anti-MDA5 and anti-SRP disease [117, 123–129].
example, non-Jo1 ASS and anti-MDA5 patients have re- Therefore, it seems that stratification based on autoAbs
duced survival compared to other serological subtypes can act as predictive markers of clinical response to treat-
[11, 29–32, 40, 41, 103, 104]. In the case of non-Jo1 ment, which again highlights their diagnostic utility.
Table 1 CTD-myositis overlap autoantibodies and clinical associations
ANA IIF Cytoplasmic Nucleolar Nuclear

Autoantibody An-SRP An-HMGCR An-ARSa An-MDA5 An-PM-Scl An-U3RNP An-RuvBL1/2 An-Mi2 An-SAE An-TIF1 An-NXP2

Myopathy-pulmonary spectrum
Syndrome Autoimmune Dermatomyosis
necrozing myopathy ASS Dermatopulmonary Overlap CTD-SSc-myopathy

Manifestationss
Subacute severe Subacute ILD: NSIP +/- OP or UIP or AIP (RP-ILD) ILD Classic DM CADM Adults:
myopathy myopathy
+/- Stans CADM Diffuse skin disease Less systemic Systemic Severe skin and subcutaneous disease
disease Red on white lesions
Mechanic’s hands PAH Dysphagia Palmar Goron’s lesions

Goron’s lesions Gastro-intesnal GI associated Cancer


disease
Raynaud’s phenomenon Children:

Inflammatory arthris Ulceraon


Oedema
Mucocutaneous ulcers Lipoatrophy
Contractures
Palmar papules
Calcinosis

Disease activity An-SRP tres An-HMGCR An-Jo1 tres An-MDA5 tres An-TIF1 tres
markers tres
Ferrin

IL18

Prognosis Oen refractory Related to subtype of ILD (non-an-Jo1 ASS and an-MDA5 RP-ILD) – worse Good Related to significant skin +/-
subcutaneous disease and malignancy in
Cellular NSIP and OP – beer adults

ASS anti-synthetase syndrome, MDA5 melanoma differentiation gene 5, SRP signal recognition particle, HMGCR hydroxy-3-methylglutaryl-coenzyme A reductase, SAE small-ubiquitin like modifier
activating enzyme, TIF1 transcriptional intermediary factor 1, NXP2 nuclear matrix protein 2, ILD interstitial lung disease, NSIP non-specific organising pneumonia, OP organising pneumonia, UIP usual
interstitial pneumonia, AIP acute interstitial pneumonia, RP-ILD rapidly progressive ILD, CADM clinically-amyopathic dermatomyositis
a
Anti-ARS (acetyl-tRNA synthetases): anti-Jo1, anti-PL12, anti-PL7, anti-OJ, anti-EJ, anti-KS, anti-Ha, anti-Zo
Clinic Rev Allerg Immunol
Clinic Rev Allerg Immunol

Summary 12. Aggarwal R, Oddis CV, Goudeau D, Fertig N, Metes I, Stephens C


et al (2014) Anti-transcription intermediary factor 1-gamma auto-
antibody ELISA development and validation. Rheumatology
Over the last 10 years, it has become evident that CTD- 53(3):433–437
myositis overlap is an autoAb-rich condition, where serology 13. Labrador-Horrillo M, Martinez MA, Selva-O’Callaghan A et al
defines sub-syndromes within the disease spectrum. It is clear (2012) Anti-TIF1 antibodies (antip155) in adult patients with der-
that this approach helps diagnosis and indicates which patients matomyositis: comparison of different diagnostic assays. Ann
Rheum Dis 71(6):993–996
are at risk of more severe organ-based disease. In addition, 14. Ishikawa A, Muro Y, Sugiura K, Akiyama M (2012) Development
identification of specific autoAb and quantification of titres of an ELISA for detection of autoantibodies to nuclear matrix
may be useful disease activity markers, help guide treatment protein 2. Rheumatology 51(7):1181–1187
strategies and indicate prognosis. The goal must now be to 15. Muro Y, Sugiura K, Akiyama M (2013) A new ELISA for derma-
tomyositis autoantibodies: rapid introduction of autoantigen
make testing for this group of autoAb readily available in cDNA to recombinant assays for autoantibody measurement.
clinical practice, so we can routinely adopt a clinico- Clin Dev Immunol 2013:856815
serological approach that in turn will improve management 16. Mammen AL, Chung T, Christopher-Stine L (2011)
of this difficult and diverse set of conditions (Table 1). Autoantibodies against 3-hydroxy-3-methylglutaryl-coenzyme
A reductase in patients with statin associated autoimmune myop-
athy. Arthritis Rheum 63(3):713–721
17. Musset L, Miyara M, Benveniste O, Charuel J-L, Shikhman A,
Boyer O et al (2014) Analysis of Autoantibodies to 3-Hydroxy-3-
References methylglutaryl-coenzyme A Reductase Using Different
Technologies. Int J Immunol Res 2014:405956
18. Drouot L, Allenbach Y, Jouen F, Charuel J-L, Martinet JRM,
1. Bohan A, Peter JB (1975) Polymyositis and dermatomyositis (first
Meyer A et al (2014) Exploring necrotizing autoimmune myopa-
of two parts). N Engl J Med 292(7):344–347
thies with a novel immunoassay for anti-3-hydroxy-3- methyl-
2. Bohan A, Peter JB (1975) Polymyositis and dermatomyositis (sec-
glutaryl-CoA reductase autoantibodies. Arthritis Res Ther 16(1):
ond of two parts). N Engl J Med 292(8):403–407
1–11
3. Tanimoto K, Nakano K, Kano S, Mori S, Ueki H, Nishitani H,
19. Friedman AW, Targoff IN, Arnett FC (1996) Interstitial lung dis-
Sato T, Kiuchi T, Ohashi Y (1995) Classification criteria for poly-
ease with autoantibodies against aminoacyl-tRNA synthetases in
myositis and dermatomyositis. J Rheumatol 22(4):668–674
the absence of clinically apparent myositis. Semin Arthritis
4. Gunawardena H, Betteridge ZE, McHugh NJ (2009) Myositis
Rheum 26(1):459–467
specific autoantibodies: their clinical and pathogenic significance
20. Kalluri M, Sahn SA, Oddis CV, Gharib SL, Christopher-Stine L,
in disease expression. Rheumatology 48(6):607–612
Danoff SK et al (2009) Clinical profile of anti-PL-12 autoanti-
5. Tansley S, Gunawardena H (2014) The evolving spectrum of
body: cohort study and review of the literature. Chest 135(6):
polymyositis and dermatomyositis–moving towards
1550–1556
clinicoserological syndromes: a critical review. Clin Rev Allergy
21. Steen VD (2005) Autoantibodies in systemic sclerosis. Semin
Immunol 47(3):264–273
Arthritis Rheum 35(1):35–42
6. Kang EH, Kuwana M, Okazaki Y, Lee EY, Lee YJ, Lee EB et al
(2014) Comparison of radioimmunoprecipitation versus antigen- 22. Debray MP, Borie R, Revel MP, Naccache JM, Khalil A, Toper C
specific assays for identification of myositis-specific autoantibodies et al (2015) Interstitial lung disease in anti-synthetase syndrome:
in dermatomyositis patients. Mod Rheumatol 24(6):945–948 initial and follow up CT findings. Eur J Radiol 84(3):516–523
7. Ronnelid J, Barbasso Helmers S, Storfors H, Grip K, Ronnblom 23. Fischer A, Brown KK (2015) Interstitial lung disease in undiffer-
L, Franck-Larsson K et al (2009) Use of a commercial line blot entiated forms of connective tissue disease. Arthritis Care Res
assay as a screening test for autoantibodies in inflammatory my- 67(1):4–11
opathies. Autoimmun Rev 9(1):58–61 24. Marguerie C, Bunn CC, Beynon HL, Bernstein RM, Hughes JM,
8. Nakashima R, Imura Y, Hosono Y, Seto M, Murakami A, So AK, Walport MJ (1990) Polymyositis, pulmonary fibrosis and
Watanabe K et al (2014) The multicenter study of a New assay autoantibodies to aminoacyl-tRNA synthetase enzymes. Q J Med
for simultaneous detection of multiple anti-aminoacyl-tRNA syn- 77(282):1019–1038
thetases in myositis and interstitial pneumonia. PLoS ONE 9(1), 25. Hallowell RW, Ascherman DP, Danoff SK (2014) Pulmonary
e85062 manifestations of polymyositis/dermatomyositis. Semin Respir
9. Aggarwal R, Oddis CV, Goudeau D, Fertig N, Metes I, Stephens C Crit Care Med 35(2):239–248
et al. (2014) Anti-signal recognition particle autoantibody ELISA 26. Hallowell RW, Danoff SK (2014) Interstitial lung disease associ-
validation and clinical associations. Rheumatology Dec 17 pii: ated with the idiopathic inflammatory myopathies and the
keu436 [Epub ahead of print] antisynthetase syndrome: recent advances. Curr Opin Rheumatol
10. Hane H, Muro Y, Watanabe K, Ogawa Y, Sugiura K, Akiyama M 26(6):684–689
(2014) Establishment of an ELISA to detect anti-glycyl-tRNA 27. Watanabe K, Handa T, Tanizawa K, Hosono Y, Taguchi Y, Noma
synthetase antibody (anti-EJ), a serological marker of S et al (2011) Detection of antisynthetase syndrome in patients
dermatomyositis/polymyositis and interstitial lung disease. Clin with idiopathic interstitial pneumonias. Respir Med 105(8):
Chim Acta 431:9–14 1238–1472
11. Sato S, Hoshino K, Satoh T, Fujita T, Kawakami Y, Fujita T, 28. Schneider F, Yousem SA, Bi D, Gibson KF, Oddis CV, Aggarwal
Kuwana M (2009) RNA helicase encoded by melanoma R (2014) Pulmonary pathologic manifestations of anti-glycl-
differentiation-associated gene 5 is a major autoantigen in patients tRNA synthetase (anti-EJ)- related inflammatory myopathy. J
with clinically amyopathic dermatomyositis: association with rap- Clin Pathol 67(8):678–683
idly progressive interstitial lung disease. Arthritis Rheum 60(7): 29. Marie I, Josse S, Decaux O, Diot E, Landron C, Roblot P et al
2193–2200 (2013) Clinical manifestations and outcome of anti-PL7 positive
Clinic Rev Allerg Immunol

patients with anti-synthetase syndrome. Eur J Intern Med 24(5): diseases associated with juvenile dermatomyositis. J Pediatr
474–479 158(4):675–677
30. Johnson C, Connors GR, Oaks J, Han S, Truong A, Richardson B 45. Kobayashi N, Takezaki S, Kobayashi I, Iwata N, Mori M, Nagai K
et al (2014) Clinical and pathologic differences in interstitial lung et al (2014) Clinical and laboratory features of fatal rapidly pro-
disease based on antisynthetase antibody type. Respir Med gressive interstitial lung disease associated with juvenile dermato-
108(10):1542–1548 myositis. Rheumatology
31. Marie I, Josse S, Decaux O, Dominique S, Landron C, Roblot P 46. Aggarwal R, Lucas M, Fertig N, Oddis CV, Medsger TA Jr (2009)
et al (2013) Outcome of anti-PL12 positive patients with Anti-U3 RNP autoantibodies in systemic sclerosis. Arthritis
antisynthetase syndrome. Presse Med 42(6):e153–e158 Rheum 60(4):1112–1118
32. Marie I, Josse S, Decaux O, Dominique S, Diot E, Landron C et al 47. Koschik RW 2nd, Fertig N, Lucas MR, Domsic RT, Medsger TA
(2012) Comparison of long-term outcome between anti-Jo1- and Jr (2012) Anti-PM-Scl antibody in patients with systemic sclero-
anti-PL7/PL12 positive patients with antisynthetase syndrome. sis. Clin Exp Rheumatol 30(2):S12–S16
Autoimmun Rev 11(10):739–745 48. D’Aoust J, Hudson M, Tatibouet S, Wick J, Canadian
33. Hamaguchi Y, Fujimoto M, Matsushita T, Kaji K, Komura K, Scleroderma Research Group, Mahler M, Baron M, Fritzler MJ
Hasegawa M et al (2013) Common and distinct clinical features (2014) Clinical and serologic correlates of anti-PM/Scl antibodies
in adult patients with anti-aminoacyl-tRNA synthetase antibodies: in systemic sclerosis: a multicenter study of 763 patients. Arthritis
heterogeneity within the syndrome. PLoS One 8(4), e60442 Rheum 66(6):1608–1615
34. Lefevre G, Meyer A, Launay D, Machelart I, DeBandt M, 49. Lega JC, Cottin V, Fabien N, Thivolet-Béjui F, Cordier JF (2010)
Michaud J et al (2014) Seronegative polyarthritis revealing Interstitial lung disease associated with anti-PM/Scl or anti-
antisynthetase syndrome: a multicentre study of 40 patients. aminoacyl-tRNA synthetase autoantibodies: a similar condition?
Rheumatology 27 J Rheumatol 37(5):1000–1009
35. Sato S, Hirakata M, Kuwana M, Suwa A, Inada S, Mimori T et al 50. Hanke K, Brückner CS, Dähnrich C, Huscher D, Komorowski L,
(2005) Autoantibodies to a 140-kd polypeptide, CADM-140, in Meyer W et al (2009) Antibodies against PM/Scl-75 and PM/Scl-
Japanese patients with clinically amyopathic dermatomyositis. 100 are independent markers for different subsets of systemic
Arthritis Rheum 52(5):1571–1576 sclerosis patients. Arthritis Res Ther 11(1):R22
51. Guillen-Del Castillo A, Pilar Simeón-Aznar C, Fonollosa-Pla V,
36. Fiorentino D, Chung L, Zwerner J, Rosen A, Casciola-
Alonso-Vila S, Reverte-Vinaixa MM, Muñoz X et al (2014) Good
Rosen L (2011) The mucocutaneous and systemic pheno-
outcome of interstitial lung disease in patients with scleroderma
type of dermatomyositis patients with antibodies to MDA5
associated to anti-PM/Scl antibody. Semin Arthritis Rheum 44(3):
(CADM-140): a retrospective study. J Am Acad Dermatol
331–337
65(1):25–34
52. Arnett FC, Reveille JD, Goldstein R, Pollard KM, Leaird K, Smith
37. Tansley SL, Betteridge ZE, Gunawardena H, Jacques TS, Owens
EA, Leroy EC, Fritzler MJ (1996) Autoantibodies to fibrillarin in
CM, Pilkington C et al (2014) Anti-MDA5 autoantibodies in ju-
systemic sclerosis (scleroderma). An immunogenetic, serologic,
venile dermatomyositis identify a distinct clinical phenotype: a
and clinical analysis. Arthritis Rheum 39(7):1151–1160
prospective cohort study. Arthritis Res Ther 16(4):R138
53. Tormey VJ, Bunn CC, Denton CP, Black CM (2001) Anti-
38. Ceribelli A, Fredi M, Taraborelli M, Cavazzana I, Tincani A, fibrillarin antibodies in systemic sclerosis. Rheumatology
Selmi C et al (2014) Prevalence and clinical significance of anti- 40(10):1157–1162
MDA5 antibodies in European patients with polymyositis/derma- 54. Kaji K, Fertig N, Medsger TA Jr, Satoh T, Hoshino K, Hamaguchi
tomyositis. Clin Exp Rheumatol 32(6):891–897 Y et al (2014) Autoantibodies to RuvBL1 and RuvBL2: a novel
39. Labrador-Horrillo M, Martinez MA, Selva-O’Callaghan A, systemic sclerosis-related antibody associated with diffuse cutane-
Trallero-Araguas E, Balada E, Vilardell-Tarres M, Juárez C ous and skeletal muscle involvement. Arthritis Care Res 66(4):
(2014) Anti-MDA5 antibodies in a large Mediterranean popula- 575–584
tion of adults with dermatomyositis. Int J Immunol Res 2014: 55. Olazagasti JM, Baez PJ, Wetter DA, Ernste FC (2015) Cancer risk
290797 in dermatomyositis: a meta-analysis of cohort studies. Am J Clin
40. Gono T, Kawaguchi Y, Satoh T, Kuwana M, Katsumata Y, Dermatol 16(2):89–98
Takagi K et al (2010) Clinical manifestation and prognostic 56. Chinoy H, Fertig N, Oddis CV, Ollier WE, Cooper RG (2007) The
factor in anti-melanoma differentiation-associated gene 5 diagnostic utility of myositis autoantibody testing for predicting
antibody-associated interstitial lung disease as a complication the risk of cancer-associated myositis. Ann Rheum Dis 66(10):
of dermatomyositis. Rheumatology 49(9):1713–1719 1345–1349
41. Gono T, Sato S, Kawaguchi Y, Kuwana M, Hanaoka M, 57. Targoff IN, Mamyrova G, Trieu EP, Perurena O, Koneru B,
Katsumata Y et al (2012) Anti-MDA5 antibody, ferritin and IL- O’Hanlon TP, Miller FW, Rider LG, Childhood Myositis
18 are useful for the evaluation of response to treatment in inter- Heterogeneity Study Group; International Myositis
stitial lung disease with anti-MDA5 antibody-positive dermato- Collaborative Study Group (2006) A novel autoantibody to a
myositis. Rheumatology 51(9):1563–1567 155-kd protein is associated with dermatomyositis. Arthritis
42. Chaisson NF, Paik J, Orbai AM, Casciola-Rosen L, Fiorentino D, Rheum 54(11):3682–3689
Danoff S, Rosen A (2012) A novel dermato-pulmonary syndrome 58. Kaji K, Fujimoto M, Hasegawa M, Kondo M, Saito Y et al (2007)
associated with MDA-5 antibodies: report of 2 cases and review of Identification of a novel autoantibody reactive with 155 and
the literature. Medicine (Baltimore) 91(4):220–228 140 kDa nuclear proteins in patients with dermatomyositis: an
43. Hall JC, Casciola-Rosen L, Samedy LA, Werner J, Owoyemi K, association with malignancy. Rheumatology 46(1):25–28
Danoff SK, Christopher-Stine L (2013) Anti-melanoma differen- 59. Trallero-Araguás E, Labrador-Horrillo M, Selva-O’Callaghan A,
tiation-associated protein 5-associated dermatomyositis: Martínez MA, Martínez-Gómez X, Palou E, Rodriguez-Sanchez
expanding the clinical spectrum. Arthritis Care Res 65(8):1307– JL, Vilardell-Tarrés M (2010) Cancer-associated myositis and
1315 anti-p155 autoantibody in a series of 85 patients with idiopathic
44. Kobayashi I, Okura Y, Yamada M, Kawamura N, Kuwana M, inflammatory myopathy. Medicine (Baltimore) 89(1):47–52
Ariga T (2011) Anti-melanoma differentiation -associated gene 5 60. Fujimoto M, Hamaguchi Y, Kaji K, Matsushita T, Ichimura Y,
antibody is a diagnostic and predictive marker for interstitial lung Kodera M et al (2012) Myositis-specific anti-155/140
Clinic Rev Allerg Immunol

autoantibodies target transcription intermediary factor 1 family autoantigen target, in UK Caucasian adult-onset myositis. Ann
proteins. Arthritis Rheum 64(2):513–522 Rheum Dis 68(10):1621–1625
61. Trallero-Araguás E, Rodrigo-Pendás JÁ, Selva-O’Callaghan A, 76. Tarricone E, Ghirardello A, Rampudda M, Bassi N, Punzi L,
Martínez-Gómez X, Bosch X, Labrador-Horrillo M, Grau- Doria A (2012) Anti-SAE antibodies in autoimmune myositis:
Junyent JM, Vilardell-Tarrés M (2012) Usefulness of anti-p155 identification by unlabelled protein immunoprecipitation in an
autoantibody for diagnosing cancer-associated dermatomyositis: Italian patient cohort. J Immunol Methods 384(1–2):128–134
a systematic review and meta-analysis. Arthritis Rheum 64(2): 77. Muro Y, Sugiura K, Akiyama M (2013) Low prevalence of anti-
523–532 small ubiquitin-like modifier activating enzyme antibodies in der-
62. Selva-O’Callaghan A, Trallero-Araguás E, Grau-Junyent JM, matomyositis patients. Autoimmunity 46(4):279–284
Labrador-Horrillo M (2010) Malignancy and myositis: novel au- 78. Bodoki L, Nagy-Vincze M, Griger Z, Betteridge Z, Szöllősi L,
toantibodies and new insights. Curr Opin Rheumatol 22(6):627– Dankó K (2014) Four dermatomyositis-specific autoantibodies-
632 anti-TIF1γ, anti-NXP2, anti-SAE and anti-MDA5-in adult and
63. Fiorentino DF, Kuo K, Chung L, Zaba L, Li S, Casciola-Rosen L juvenile patients with idiopathic inflammatory myopathies in a
(2015) Distinctive cutaneous and systemic features associated Hungarian cohort. Autoimmun Rev 13(12):1211–1219
with antitranscriptional intermediary factor-1γ antibodies in adults 79. Fujimoto M, Matsushita T, Hamaguchi Y, Kaji K, Asano Y,
with dermatomyositis. J Am Acad Dermatol 72(3):449–455 Ogawa F et al (2013) Autoantibodies to small ubiquitin-like mod-
64. Fiorentino DF, Chung LS, Christopher-Stine L, Zaba L, Li S, ifier activating enzymes in Japanese patients with dermatomyosi-
Mammen AL, Rosen A, Casciola-Rosen L (2013) Most patients tis: comparison with a UK Caucasian cohort. Ann Rheum Dis
with cancer-associated dermatomyositis have antibodies to nucle- 72(1):151–153
ar matrix protein NXP-2 or transcription intermediary factor 1γ. 80. Targoff IN, Reichlin M (1985) The association between Mi-2
Arthritis Rheum 65(11):2954–2962 antibodies and dermatomyositis. Arthritis Rheum 28(7):796–803
65. Ichimura Y, Matsushita T, Hamaguchi Y, Kaji K, Hasegawa M, 81. Okada S, Weatherhead E, Targoff IN, Wesley R, Miller FW,
Tanino Y et al (2012) Anti-NXP2 autoantibodies in adult patients International Myositis Collaborative Study Group (2003) Global
with idiopathic inflammatory myopathies: possible association surface ultraviolet radiation intensity may modulate the clinical
with malignancy. Ann Rheum Dis 71(5):710–713 and immunologic expression of autoimmune muscle disease.
66. Gunawardena H, Wedderburn LR, North J, Betteridge Z, Dunphy Arthritis Rheum 48(8):2285–2293
J, Chinoy H et al (2008) Clinical associations of autoantibodies to 82. Love LA, Weinberg CR, McConnaughey DR, Oddis CV, Medsger
a p155/140 kDa doublet protein in juvenile dermatomyositis. TA Jr, Reveille JD, Arnett FC, Targoff IN, Miller FW (2009)
Rheumatology 47(3):324–328 Ultraviolet radiation intensity predicts the relative distribution of
67. Bingham A, Mamyrova G, Rother KI, Oral E, Cochran E, dermatomyositis and anti-Mi-2 autoantibodies in women. Arthritis
Premkumar A et al (2008) Predictors of acquired lipodystrophy Rheum 60(8):2499–2504
in juvenile-onset dermatomyositis and a gradient of severity. 83. Petri MH, Satoh M, Martin-Marquez BT, Vargas-Ramírez R, Jara
Medicine (Baltimore) 87(2):70–86 LJ, Saavedra MA et al (2013) Implications in the difference of
68. Espada G, Maldonado Cocco JA, Fertig N, Oddis CV (2009) anti-Mi-2 and -p155/140 autoantibody prevalence in two derma-
Clinical and serologic characterization of an Argentine pediatric tomyositis cohorts from Mexico City and Guadalajara. Arthritis
myositis cohort: identification of a novel autoantibody (anti-MJ) Res Ther 4;15(2):R48
to a 142-kDa protein. J Rheumatol 36(11):2547–2551 84. Rider LG, Shah M, Mamyrova G, Huber AM, Rice MM, Targoff
69. Gunawardena H, Wedderburn LR, Chinoy H, Betteridge ZE, IN, Miller FW, Childhood Myositis Heterogeneity Collaborative
North J, Ollier WE et al (2009) Autoantibodies to a 140-kd protein Study Group (2013) The myositis autoantibody phenotypes of the
in juvenile dermatomyositis are associated with calcinosis. juvenile idiopathic inflammatory myopathies. Medicine
Arthritis Rheum 60(6):1807–1814 (Baltimore) 92(4):223–243
70. Tansley SL, Betteridge ZE, Shaddick G, Gunawardena H, Arnold 85. Brown DA, Simoni RD (1984) Biogenesis of 3-hydroxy-3-
K, Wedderburn LR, McHugh NJ, Juvenile Dermatomyositis methylglutaryl-coenzyme A reductase, an integral glycoprotein
Research Group (2014) Calcinosis in juvenile dermatomyositis of the endoplasmic reticulum. Proc Natl Acad Sci U S A 81(6):
is influenced by both anti-NXP2 autoantibody status and age at 1674–1678
disease onset. Rheumatology 53(12):2204–2208 86. Olender EH, Simon RD (1992) The intracellular targeting and
71. Valenzuela A, Chung L, Casciola-Rosen L, Fiorentino D (2014) membrane topology of 3-hydroxy-3-methylglutaryl-CoA reduc-
Identification of clinical features and autoantibodies associated tase. J Biol Chem 25;267(6):4223–4235
with calcinosis in dermatomyositis. JAMA Dermatol 150(7): 87. Hengstman GJ, ter Laak HJ, Vree Egberts WT, Lundberg IE,
724–729 Moutsopoulos HM, Vencovsky J et al (2006) Anti-signal recogni-
72. Ceribelli A, Fredi M, Taraborelli M, Cavazzana I, Franceschini F, tion particle autoantibodies: marker of a necrotising myopathy.
Quinzanini M et al (2012) Anti-MJ/NXP-2 autoantibody specific- Ann Rheum Dis 65(12):1635–1638
ity in a cohort of adult Italian patients with polymyositis/dermato- 88. Targoff IN, Johnson AE, Miller FW (1990) Antibody to signal
myositis. Arthritis Res Ther 14(2):R97 recognition particle in polymyositis (1990). Arthritis Rheum
73. Sugiura K, Muro Y, Akiyama M (2012) Autoantibodies to nuclear 33(9):1361–1370
matrix protein 2/MJ in adult-onset dermatomyositis with severe 89. Hengstman GJ, Brouwer R, Egberts WT, Seelig HP, Jongen
calcinosis. J Am Acad Dermatol 67(4):e167–e168 PJ, van Venrooij WJ, van Engelen BG (2002) Clinical and
74. Shah AA, Casciola-Rosen L, Rosen A (2015) Review: cancer- serological characteristics of 125 Dutch myositis patients.
induced autoimmunity in the rheumatic diseases. Arthritis Myositis specific autoantibodies aid in the differential diag-
Rheum 67(2):317–326 nosis of the idiopathic inflammatory myopathies. J Neurol
75. Betteridge ZE, Gunawardena H, Chinoy H, North J, Ollier WE, 249(1):69–75
Cooper RG, McHugh NJ, UK Adult Onset Myositis 90. Basnayake SK, Blumbergs P, Tan JA, Roberts-Thompson PJ,
Immunogenetic Collaboration (2009) Clinical and human leuco- Limaye V (2015) Inflammatory myopathy with anti-SRP antibod-
cyte antigen class II haplotype associations of autoantibodies to ies: case series of a South Australian cohort. Clin Rheumatol
small ubiquitin-like modifier enzyme, a dermatomyositis-specific 34(3):603–608
Clinic Rev Allerg Immunol

91. Miller T, Al-Lozi MT, Lopate G, Pestronk A (2002) Myopathy 107. Morganroth PA, Kreider ME, Werth VP (2010) Mycophenolate
with antibodies to the signal recognition particle: clinical and path- mofetil for interstitial lung disease in dermatomyositis. Arthritis
ological features. J Neurol Neurosurg Psychiatry 73(4):420–428 Care Res 62(10):1496–1501
92. Suzuki S, Hayashi YK, KuwanaM TR, Suzuki N, Nishino I 108. Fischer A, Brown KK, Du Bois RM, Frankel SK, Cosgrove GP,
(2012) Myopathy associated with antibodies to signal recognition Fernandez-Perez ER et al (2013) Mycophenolate mofetil improves
particle: disease progression and neurological outcome. Arch lung function in connective tissue disease-associated interstitial
Neurol 69(6):728–732 lung disease. J Rheumatol 40(5):640–646
93. Pasternak RC, Smith SC Jr, Bairey-Merz CN, Grundy SM, 109. Wilkes MR, Sereika SM, Fertig N, Lucas MR, Oddis CV (2005)
Cleeman JI, Lenfant C, American College of Cardiology; Treatment of antisynthetase-associated interstitial lung disease
American Heart Association; National Heart, Lung and Blood with tacrolimus. Arthritis Rheum 52(8):2439–2446
Institute (2002) ACC/AHA/NHLBI clinical advisory on the use 110. Labirua-Iturburu A, Selva-O’Callaghan A, Martínez-Gómez X,
and safety of statins. J Am Coll Cardiol 40(3):567–572 Trallero-Araguás E, Labrador-Horrillo M, Vilardell-Tarrés M
94. Christopher-Stine L, Casciola-Rosen LA, Hong G, Chung T, Corse (2013) Calcineurin inhibitors in a cohort of patients with
AM, Mammen AL (2010) A novel autoantibody recognizing 200-kd antisynthetase-associated interstitial lung disease. Clin Exp
and 100-kd proteins is associated with an immune-mediated necro- Rheumatol 31(3):436–439
tizing myopathy. Arthritis Rheum 62(9):2757–2766 111. Cavagna L, Caporali R, Abdì-Alì L, Dore R, Meloni F,
95. Mammen AL, Gaudet D, Brisson D, Christopher-Stine L, Lloyd Montecucco C (2013) Cyclosporine in anti-Jo1-positive patients
TE, Leffell MS, Zachary AA (2012) Increased frequency of with corticosteroid-refractory interstitial lung disease. J
DRB1*11:01 in anti-hydroxymethylglutaryl-coenzyme A Rheumatol 40(4):484–492
reductase-associated autoimmune myopathy. Arthritis Care Res 112. Kurita T, Yasuda S, Amengual O, Atsumi T (2015) The efficacy of
64(8):1233–1237 calcineurin inhibitors for the treatment of interstitial lung disease
96. Limaye V, Bundell C, Hollingsworth P, Rojana-Udomsart A, associated with polymyositis/dermatomyositis. Lupus 24(1):3–9
Mastaglia F, Blumbergs P, Lester S (2014) Clinical and genetic 113. Muro Y, Sugiura K, Akiyama M (2013) Limitations of a single-
associations of autoantibodies to 3-hydroxy-3-methyl-glutaryl-co- point evaluation of anti-MDA5 antibody, ferritin, and IL-18 in
enzyme a reductase in patients with immune-mediated myositis predicting the prognosis of interstitial lung disease with anti-
and necrotizing myopathy. Muscle Nerve. doi:10.1002/mus.24541 MDA5 antibody-positive dermatomyositis. Clin Rheumatol
97. SEARCH Collaborative Group, Link E, Parish S, Armitage J, 32(3):395–398
Bowman L, Heath S, Matsuda F, Gut I, Lathrop M, Collins R
114. Tanaka F, Origuchi T, Migita K, Tominaga M, Kawakami A,
(2008) SLCO1B1 variants and statin-induced myopathy–a
Kawabe Y, Eguchi K (2000) Successful combined therapy of cy-
genomewide study. N Engl J Med 21;359(8):789–799
clophosphamide and cyclosporine for acute exacerbated intersti-
98. Grable-Esposito P, Katzberg HD, Greenberg SA, Srinivasan J,
tial pneumonia associated with dermatomyositis. Intern Med
Katz J, Amato AA (2010) Immune-mediated necrotizing myopa-
39(5):428–430
thy associated with statins. Muscle Nerve 41(2):185–190
115. Kameda H, Nagasawa H, Ogawa H, Sekiguchi N, Takei H,
99. Werner JL, Christopher-Stine L, Ghazarian SR, Pak KS, Kus JE,
Tokuhira M, Amano K, Takeuchi T (2005) Combination therapy
Daya NR, Lloyd TE, Mammen AL (2012) Antibody levels corre-
with corticosteroids, cyclosporin A, and intravenous pulse cyclo-
late with creatine kinase levels and strength in anti-3-hydroxy-3-
phosphamide for acute/subacute interstitial pneumonia in patients
methylglutaryl-coenzyme A reductase-associated autoimmune
with dermatomyositis. J Rheumatol 32(9):1719–1726
myopathy. Arthritis Rheum 64(12):4087–4093
116. Levine TD (2005) Rituximab in the treatment of dermatomyositis:
100. Stone KB, Oddis CV, Fertig N, Katsumata Y, Lucas M, Vogt M,
an open-label pilot study. Arthritis Rheum 52(2):601–607
Domsic R, Ascherman DP (2007) Anti-Jo-1 antibody levels cor-
relate with disease activity in idiopathic inflammatory myopathy. 117. Valiyil R, Casciola-Rosen L, Hong G, Mammen A, Christopher-
Arthritis Rheum 56(9):3125–3131 Stine L (2010) Rituximab therapy for myopathy associated with
101. Sato S, Kuwana M, Fujita T, Suzuki Y (2013) Anti-CADM-140/ anti-signal recognition particle antibodies: a case series. Arthritis
MDA5 autoantibody titer correlates with disease activity and pre- Care Res 62(9):1328–1334
dicts disease outcome in patients with dermatomyositis and rapidly 118. Couderc M, Gottenberg JE, Mariette X, Hachulla E, Sibilia J, Fain
progressive interstitial lung disease. Mod Rheumatol 23(3):496–502 O et al (2011) Efficacy and safety of rituximab in the treatment of
102. Muro Y, Sugiura K, Hoshino K, Akiyama M (2012) refractory inflammatory myopathies in adults: results from the
Disappearance of anti-MDA-5 autoantibodies in clinically AIR registry. Rheumatology 50(12):2283–2289
amyopathic DM/interstitial lung disease during disease remis- 119. Bader-Meunier B, Decaluwe H, Barnerias C, Gherardi R, Quartier P,
sion. Rheumatology 51:800–804 Faye A et al (2011) Safety and efficacy of rituximab in severe juvenile
103. Aggarwal R, Cassidy E, Fertig N, Koontz DC, Lucas M, dermatomyositis: results from 9 patients from the French
Ascherman DP, Oddis CV (2014) Patients with non-Jo-1 anti- Autoimmunity and Rituximab registry. J Rheumatol 38(7):1436–1440
tRNA-synthetase autoantibodies have worse survival than Jo-1 120. Mahler EA, Blom M, Voermans NC, van Engelen BG, van Riel
positive patients. Ann Rheum Dis 73(1):227–232 PL, Vonk MC (2011) Rituximab treatment in patients with refrac-
104. Nakashima R, Imura Y, Kobayashi S, Yukawa N, Yoshifuji H, tory inflammatory myopathies. Rheumatology 50(12):2206–2213
Nojima T et al (2010) The RIG-I-like receptor IFIH1/MDA5 is a 121. Oddis CV, Reed AM, Aggarwal R, Rider LG, Ascherman DP,
dermatomyositis-specific autoantigen identified by the anti- Levesque MC et al (2013) Rituximab in the treatment of refractory
CADM-140 antibody. Rheumatology (Oxford) 49(3):433–440 adult and juvenile dermatomyositis and adult polymyositis: a ran-
105. Gordon PA, Winer JB, Hoogendijk JE, Choy EH (2012) domized, placebo-phase trial. Arthritis Rheum 65(2):314–324
Immunosuppressant and immunomodulatory treatment for der- 122. Aggarwal R, Bandos A, Reed AM, Ascherman DP, Barohn RJ,
matomyositis and polymyositis. Cochrane Database Syst Rev Feldman BM (2014) Predictors of clinical improvement in
15(8):CD003643 rituximab-treated refractory adult and juvenile dermatomyositis
106. Yamasaki Y, Yamada H, Yamasaki M, Ohkubo M, Azuma K, and adult polymyositis. Arthritis Rheum 66(3):740–749
Matsuoka S et al (2007) Intravenous cyclophosphamide therapy 123. Marie I, Dominique S, Janvresse A, Levesque H, Menard JF
for progressive interstitial pneumonia in patients with polymyosi- (2012) Rituximab therapy for refractory interstitial lung disease
tis/dermatomyositis. Rheumatology 46(1):124–130 related to antisynthetase syndrome. Respir Med 106(4):581–587
Clinic Rev Allerg Immunol

124. Clottu A, Laffitte E, Prins C, Chizzolini C (2012) Response of mu- differential effects in a real-life population. Rheumatology 53(9):
cocutaneous lesions to rituximab in a case of melanoma differentiation 1630–1638
antigen 5-related dermatomyositis. Dermatology 225(4):376–380 128. Andersson H, Sem M, Lund MB, Aaløkken TM, Günther A,
125. Nalotto L, Iaccarino L, Zen M, Gatto M, Borella E, Domenighetti M, Walle-Hansen R, Garen T (2015) Long-term experience with ri-
Punzi L, Doria A (2013) Rituximab in refractory idiopathic inflam- tuximab in anti-synthetase syndrome-related interstitial lung dis-
matory myopathies and antisynthetase syndrome: personal experience ease. Rheumatology
and review of the literature. Immunol Res 56(2–3):362–370 129. Koichi Y, Aya Y, Megumi U, Shunichi K, Masafumi S,
126. Muñoz-Beamud F, Isenberg DA (2014) Rituximab as an effective Hiroaki M et al (2015) A case of anti-MDA5-positive rapidly
alternative therapy in refractory idiopathic inflammatory myopa- progressive interstitial lung disease in a patient with clinical-
thies. Clin Exp Rheumatol 31(6):896–903 ly amyopathic dermatomyositis ameliorated by rituximab, in
127. Unger L, Kampf S, Lüthke K, Aringer M (2014) Rituximab ther- addition to standard immunosuppressive treatment. Mod
apy in patients with refractory dermatomyositis or polymyositis: Rheumatol 12:1–5

You might also like