You are on page 1of 11

FOCUS ON MYOSITIS

REVIEWS
Treatment in myositis
Chester V. Oddis* and Rohit Aggarwal
Abstract | As with the treatment of many immune-mediated diseases, managing myositis
encompasses diverse factors, which present a challenge to the physician caring for these
patients. The idiopathic inflammatory myopathies (IIMs, also known as myositis), are
fundamentally heterogeneous; many contributory immunological perturbations are involved in
the pathogenesis of myositis, leading to varying clinical phenotypic presentations. Targeting any
one or several of these deleterious pathways with a therapeutic agent might seem reasonable,
but the desired response is not uniformly predictable. The presence of many serious
extramuscular manifestations, such as severe skin rash, interstitial lung disease and arthritis,
complicates the management of myositis. Myositis is rare, and very few large treatment trial
results are available to guide clinicians. Outcome measures to effectively gauge treatment
responses have been available for only a few years, and response criteria that incorporate critical
core set measures continue to evolve. Nevertheless, a multitude of immunosuppressive and
immunomodulatory agents are available to clinicians managing myositis, and the emergence of
biologic agents targeting potential pathogenic pathways offers hope for mitigating or curing this
enigmatic group of diseases. Paradigm shifts in the nonpharmacological approach to treat
myositis have also occurred as more aggressive exercise regimens have shown benefit in patients,
even those with active disease.

The idiopathic inflammatory myopathies (IIMs), also responses in myositis are discussed in this Issue in a
referred to as myositis, are fundamentally hetero- Review by Rider and colleagues1. The recognition of the
geneous, as discussed in this Review. Despite the simple protean clinical features seen with myositis, advances
name, myositis encompasses much more than simply in the understanding of myositis immunopathogenesis
autoimmune inflammation in muscle tissue, and the and the availability of targeted therapy coupled with
presence of extramuscular disease manifestations might more sophisticated outcome measures and robust
limit the use of specific immunosuppressive agents. For response criteria all validate the necessity of a thoughtful
example, lung involvement is one of the most common treatment approach to myositis.
features in myositis, the presence of which requires a In this Review, we examine the many immuno-
thoughtful approach that might narrow the range of suppressive and immunomodulatory agents available to
general treatment options available. Advances in identi- clinicians managing patients with myositis, as well as the
fying the many immunological perturbations involved increasing number of biologic agents available that target
in the pathogenesis of myositis have certainly informed potential pathogenic pathways (FIG. 1). We also discuss
research into myositis therapies, resulting in clinical exercise, an often-neglected area in the management of
trials employing interesting biologic therapies as phar- myositis, including the molecular basis that such ther-
maceutical companies move in the direction of treating apy is indeed anti-inflammatory. As alluded to above,
patients with rare autoimmune diseases. Furthermore, skin manifestations might dominate the clinical course
novel therapeutic approaches are addressing the treat- of dermatomyositis, and ILD remains the major mor-
ment of the many serious or stubborn extramuscular bid complication of this disease. Hence, we specifically
Division of Rheumatology and
manifestations of myositis, such as severe skin rashes consider the management of myositis beyond treating
Clinical Immunology,
University of Pittsburgh in dermatomyositis, interstitial lung disease (ILD) and inflammation of the muscle. Finally, we briefly discuss
School of Medicine, even inflammatory arthritis and dysphagia. However, future prospects in the management of this enigmatic
Pittsburgh, PA, USA. myositis is rare, and there are very few large treatment subset of autoimmune disease. In this Review, we do not
*e-mail: cvo5@pitt.edu trials available to guide clinicians managing patients address the treatment of inclusion body myositis, a sub-
doi:10.1038/nrrheum.2018.42 with myositis. Newly developed outcome measures set of myositis notoriously refractory to pharmacological
Published online 29 Mar 2018 and response criteria for accurately assessing treatment intervention.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 1

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
REVIEWS

Key points early, as opposed to established, polymyositis and der-


matomyositis, providing evidence that these cytokines
• Managing myositis and its systemic complications represents a challenge to the might contribute to disease pathogenesis9. The linkage
clinicians providing care as patients often have severe muscle weakness, notable skin of activated T cells to IL-17 is further supported by
rashes and life-threatening organ involvement. the demonstration of this cytokine in the lymphocytic
• Conventional therapies include glucocorticoids usually in combination with another infiltrates of muscle tissue.
or multiple immunosuppressive agents, but biologic therapies targeting Toll-like receptors (TLRs) serve as links between
immunopathogenic pathways are being increasingly utilized.
the innate and adaptive immune systems. Patients with
• Interstitial lung disease is a major cause of morbidity and mortality in myositis polymyositis or dermatomyositis overexpress TLR3 and
requiring combinations of glucocorticoids, immunosuppressive drugs and agents that
TLR7, which are primarily detected in the inflammatory
modulate T cell function and deplete B cells.
infiltrates within the muscle tissue of these patients10. In
• Exercise, once considered controversial in the management of myositis, has emerged
myositis, the TLR pathway is supposedly activated by
as an important adjunct in treating patients with myositis; molecular evidence
suggests that exercise regimens are both safe and anti-inflammatory.
mediators released from necrotic muscle cells.
The emergence of biologic and small-molecule ther-
apies over the past several years that specifically attack
Pathogenic pathways inform treatment the aforementioned immunological targets that cause
Insights from immunopathogenic pathways or are associated with myositis provides the opportu-
The finding of T and B cell inflammatory infiltrates nity for mitigating disease in ways that have not been
in the skin and muscles of patients with polymyositis previously feasible.
or dermatomyositis, along with the identification of
myositis-associated autoantibodies, clearly point towards Insights from non-immune pathways
a disease process that is responsive to glucocorticoid and Muscle weakness in myositis might persist even when
immunosuppressive therapy. Treatment with gluco- there is no evidence of muscle damage and/or no
corticoids and conventional immunosuppressive drugs demonstrable inflammatory cells within the muscle tis-
is a fairly nonspecific approach. However, B cells and sue. There is increasing evidence that noninflammatory
T cells, as well as the specific cytokines and receptors or non-immune mechanisms might contribute to the
implicated in myositis pathogenesis, serve as poten- pathogenesis of myositis and muscle weakness. In
tially attractive therapeutic targets. In polymyositis, fact, in a murine model of myositis, muscle weakness
myeloid dendritic cells activate B cells and contribute to precedes the infiltration of inflammatory cells11, and
the formation of autoantibodies2, whereas in dermato- muscle weakness persists in patients with polymyositis
myositis and to a lesser extent in polymyositis, plasma- or dermatomyositis even when treatment has cleared
cytoid dendritic cells (professional producers of type I the myositis-associated inflammatory infiltrates12. The
interferon), are implicated in disease pathogenesis. In precise mechanisms underlying the noninflammatory
dermatomyositis, genes induced by type I interferon component of muscle weakness are not well understood,
(known as a type I interferon signature) are highly but investigators have shown that the endoplasmic retic-
overexpressed in the muscle tissue; similarly, immuno- ulum (ER) stress pathways are activated in myositis11–16.
histochemical analysis has demonstrated the deposition Activation of these pathways is directly associated with
of an interferon-inducible protein, interferon-induced muscle fibre degeneration and muscle dysfunction in
GTP-binding protein MX1, in muscle tissue3. In patients myositis14. In non-muscle cells, ER stress also leads to
with dermatomyositis, disease activity correlates with altered redox homeostasis and possible oxidative dam-
both the expression of a type I interferon gene signa- age14. In 2015, investigators proposed that the genera-
ture and serum levels of IL-6. Furthermore, IL-6 levels tion of modified reactive oxygen species (ROS) might
correlate with the expression of a type I interferon gene be involved in non-immune cell-mediated muscle
signature and the type I interferon chemokine signature weakness in myositis through a mechanism involving
in such patients, suggesting that the co-regulation of ER stress induction and that therapies that target ROS
interferon-driven chemokines and IL-6 is linked to the generation could have a role in myositis14.
pathogenesis of dermatomyositis4.
Whereas CD4+ T cells are important in dermato- Treatment strategies in myositis
myositis pathogenesis, CD8+ T cells are more often Immunosuppressive agents
implicated in polymyositis pathogenesis. T cells have Glucocorticoids. The management of myositis is not
also been implicated in the pathogenesis of myositis- guideline-based; however, despite the lack of controlled
associated ILD as activated CD8+ T cells have been noted clinical trials, it is generally agreed that glucocorti-
in both the lung tissue and bronchoalveolar lavage fluid coids should remain the anchor drug of initial myosi-
of patients with myositis5,6, whereas patients with inter- tis treatment. The starting dose depends on various
stitial pneumonitis associated with various autoimmune factors (such as age and indication), but in adults with
diseases (including myositis) have decreased levels of prominent muscle weakness the dose approximates
regulatory T cells7. IL-17 is also emerging as a poten- to 1 mg per kg daily, with an average dose of 60 mg
tial mediator of the inflammatory process in myositis daily for a patient with myositis, generally not exceed-
as levels of this cytokine correlate with disease duration ing 80 mg per day. If a patient has severe myositis or
and levels of IL-15, particularly in patients with der- myositis with other prominent extramuscular com-
matomyositis8. IL-17 and IL-23 levels are increased in plications, such as ILD, pulse therapy with 1,000 mg of

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
F O C U S O N MRYEO
V ISEI W
TIS

First-line
Glucocorticoids and Methotrexate or azathioprine and/or IVIG By contrast, another open-label, randomized, placebo-
therapy:
controlled, multicentre trial in Europe that assessed
MMF, tacrolimus or ciclosporin
the efficacy and safety of combined methotrexate and
Second-line
Glucocorticoids and or combination therapy of and/or IVIG glucocorticoid therapy versus glucocorticoids alone (the
therapy:
methotrexate and azathioprine Prometheus trial) in adults with dermatomyositis or
polymyositis failed to complete recruitment (62% target)
Third-line
Glucocorticoids and
Rituximab, cyclophosphamide,
and/or IVIG
and the primary end point was not reached22.
therapy: RCI or other biologic agents
Azathioprine. Azathioprine (2–3 mg per kg daily, orally),
Figure 1 | Overview of pharmacological therapy in idiopathic inflammatory a prodrug that is converted into the active metabolite
myopathies. The management of myositis is not guideline-based but includes
Nature Reviews many
| Rheumatology 6-mercaptopurine, is preferred by some clinicians as the
immunosuppressive, immunomodulatory and biologic agents that are noted in this
initial immunosuppressive agent of choice (generally
figure. The first-line therapy nearly always includes glucocorticoids in combination with
either methotrexate or azathioprine. The second-line therapy includes mycophenolate in combination with glucocorticoids) for patients with
mofetil (MMF), tacrolimus or ciclosporin or combination therapy such as azathioprine myositis, and comparative studies have suggested that
and methotrexate. The third-line therapy includes rituximab, cyclophosphamide, methotrexate and azathioprine have similar efficacies23.
repository corticotropin injection (RCI) or other experimental biologic agents. Azathioprine therapy might be preferable to methotrexate
Intravenous immunoglobulin (IVIG) can be used alone as a first-line, second-line or for patients with liver disease, patients unwilling to abstain
third-line therapy or as a concomitant therapy with any drug on the basis of the clinical from alcohol or patients with severe ILD. In a study in the
manifestation or refractoriness of the disease. early 1980s of patients with polymyositis, patients being
treated with azathioprine in combination with prednisone
methylprednisolone for 3 consecutive days can often be failed to show considerable differences in muscle strength
administered. Tapering guidelines vary, but a reasonable or creatine kinase levels (a marker of muscle damage),
approach is to drop the dose by 20–25% of the existing compared with patients being treated with prednisone
dose monthly with the goal of achieving a low daily dose alone, at 3 months24,25, but a long-term follow-up of this
of prednisone of approximately 5–10 mg daily within study showed that the combined prednisone plus aza-
6 months. Adrenocorticotropic hormone (ACTH) gel, thioprine therapy was superior in improving functional
also known as repository corticotropin injection (RCI), status and reducing prednisone dose24,25. In a randomized
was approved for polymyositis and dermatomyositis in crossover study of 30 patients with an initial inadequate
1952 and approval was later retained in 2010. This long- response to methotrexate or azathioprine alone, a combi-
lasting agent contains full-sequence ACTH and other nation of oral methotrexate and azathioprine was better
pro-opiomelanocortin-derived peptides. Melanocortin than intravenous methotrexate26.
receptors are expressed by many immune-mediated
cells, and activation of these receptors (for example, by Mycophenolate mofetil. Mycophenolate mofetil (MMF)
ACTH) leads to anti-inflammatory and immunomodula- is a prodrug of mycophenolic acid that inhibits purine
tory effects15. The efficacy of RCI was previously demon- synthesis, leading to immunosuppression by impairing
strated in retrospective case series16,17, but favourable B cell and T cell proliferation. Investigations of the use
results from an open-label clinical trial are now available, of MMF in myositis have mainly been limited to case
showing that treatment with RCI is safe and effective in series (generally involving doses of 2,000 − 3,000 mg
patients with refractory myositis and leads to a reduction daily, orally)27–30, but in an open-label study of seven
in concomitant steroid dosing 18. patients with either refractory polymyositis or dermato-
myositis, all the patients achieved complete remission fol-
Methotrexate. Glucocorticoids are rarely used alone lowing treatment with MMF combined with intravenous
owing to their associated adverse effects and high relapse immunoglobulin (IVIG) therapy 31.
rates. Methotrexate is often used in combination with
glucocorticoids as an initial therapy in myositis and in Calcineurin inhibitors. There is a rationale for targeting
the treatment of patients experiencing disease flares fol- T cells in both the treatment of myositis and the treat-
lowing the tapering of glucocorticoids, despite the lack ment of ILD associated with myositis. Ciclosporin is a cal-
of clinical trials supporting the use of methotrexate in cineurin inhibitor that blocks the production and release
adults with myositis. Older, retrospective studies support of IL-2 and IL-2-induced activation of T cells, whereas
the use of methotrexate as a first-line therapy in myosi- tacrolimus is a second-generation calcineurin inhibitor
tis, and this therapy can be administered either orally or that binds to an intracellular protein, peptidyl-prolyl cis-
subcutaneously with a dose of up to 25 mg weekly 19,20. In trans isomerase FKBP12, leading to inhibition of T cell
2016, results from an open-label, randomized, placebo- activation. In a case series of eight patients with refractory
controlled, multicentre trial of treatment-naive patients myositis (six patients with anti-histidyl-transfer RNA
with juvenile dermatomyositis demonstrated that treat- synthetase (Jo1) autoantibodies and two patients with
ment with methotrexate in combination with prednisone anti-signal recognition particle (SRP) autoantibodies),
resulted in a better response than prednisone alone21. A treatment with tacrolimus led to an improvement in
combination therapy of methotrexate or ciclosporin with muscle strength and a decrease in serum creatine kinase
prednisone also resulted in a shorter median time to levels32. In a Japanese observational study of 16 patients
clinical remission and prednisone discontinuation and a with polymyositis and 15 patients with dermatomyositis,
longer time to treatment failure than prednisone alone21. treatment with tacrolimus led to a substantial decrease

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 3

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
REVIEWS

in serum creatine kinase levels 2–4 months after treat- following rituximab treatment 38, whereas the results of
ment initiation and a similar improvement in muscle another open-label trial of eight patients with dermato-
strength scores33. However, tacrolimus therapy is gen- myositis were less encouraging, with only three patients
erally reserved for patients with refractory myositis showing a modest improvement in muscle strength and no
owing to toxicity concerns associated with its use and patients showing substantial improvements in skin disease
the necessity for checking tacrolimus blood levels. by 24 weeks39. In the largest randomized, double-blind,
controlled clinical trial ever completed in myositis, the
Cyclophosphamide. Cyclophosphamide, an alkylating Rituximab in Myositis (RIM) trial, 195 individuals (75
agent that interferes with the growth of cells, is generally with polymyositis, 72 with dermatomyositis and 48 with
reserved for the treatment of patients with severe myosi- juvenile dermatomyositis) were randomly assigned to
tis, patients with rapidly progressive ILD or overlapping receive two 1 g rituximab infusions either at baseline or
systemic vasculitis or patients refractory to several other 8 weeks later 40. The patients included were refractory to
second-line or third-line agents. Cyclophosphamide can glucocorticoids and at least one immunosuppressive agent
be administered orally or intravenously, but its use is and had prominent muscle weakness on entry. The pri-
limited owing to its toxic effects and the increased risk mary end point, which compared the time to achieve the
of malignancy associated with this therapy. International Myositis Assessment and Clinical Studies
(IMACS) definition of improvement (DOI; which incor-
Biologic agents porates the IMACS’s six core set measures of disease
Whereas standard immunosuppressive therapies are activity)41 in patients receiving rituximab at baseline and
generally indirect and nonspecific, biologic therapies can patients receiving rituximab treatment 8 weeks later, was
directly target the immune cells or cytokines implicated not met. However, 83% of the patients included in the pri-
in disease pathogenesis. A number of biologic therapies mary outcome analysis met the DOI by the end of the trial
have been investigated in the treatment of myositis, (that is, by 44 weeks), with a median time to achieve the
including in some large clinical trials (TABLE 1). DOI of 20 weeks. Rituximab use also had a considerable
steroid-sparing effect and was well tolerated, with a lack of
Rituximab. Rituximab depletes CD20+ B cells, and several substantial adverse events. The presence of anti-aminoacyl
case reports and case series of patients with myositis have transfer RNA synthetase antibodies (in particular anti-Jo1
reported rituximab as an effective therapy 34–36, including autoantibodies), commonly referred to as antisynthetase
in the treatment of patients with the severe and refrac- autoantibodies, as well as anti-Mi2 autoantibodies and a
tory subset of immune-mediated necrotizing myopathy lower disease damage score at trial entry were strong pre-
(IMNM) associated with the anti-SRP autoantibody 37. In dictors of a beneficial response to rituximab in the RIM
an open-label trial of four patients with refractory poly- trial42. Patients with juvenile dermatomyositis also fared
myositis, all patients had a marked drop in serum creatine better following B cell depletion in the RIM trial than
kinase levels and regained full muscle strength at 28 weeks patients with other myositis subsets.

Table 1 | Summary of biologic agents used in the treatment of myositis: selected trials
Biologic Therapeutic Study population Study design Primary end points Results Refs
agent target
Rituximab B cells Adult polymyositis and Randomized, IMACS DOI Primary end point was 39
dermatomyositis and double-blind and not met, but 83% of study
JDM (n = 200) placebo-phase participants met the DOI
Anti-SRP-positive Open-label MMT and creatine kinase Six of eight patients 38
individuals (n = 8) decline showed improvements
Infliximab TNF Adult polymyositis and Randomized, double- ≥15% MMT improvement <33% response rate 50
dermatomyositis (n = 12) blind, placebo-controlled
and crossover
Etanercept TNF Adult dermatomyositis Randomized, Adverse events; time Five of 11 etanercept- 43
(n = 16) double-blind and from randomization treated patients were
placebo-controlled to treatment failure; weaned off prednisone;
prednisone wean no adverse events
Tocilizumab IL-6 Adult polymyositis, Randomized, Myositis Total Study in progress 58
dermatomyositis and double-blind and Improvement Score
IMNM (n = 40) placebo-controlled
Abatacepta T cells Adult polymyositis and Randomized, open-label IMACS DOI Treatment resulted in 63
dermatomyositis (n = 20) and ‘delayed-start’ lower disease activity in
nearly 50% of patients
Anakinra IL-1 receptor Adult polymyositis, Open-label IMACS DOI and functional Seven of the 15 patients 64
dermatomyositis and index responded to treatment
IBM (n = 15)
DOI, definition of improvement; IBM, inclusion body myositis; IMACS, International Myositis Assessment and Clinical Studies; IMNM, immune-mediated
necrotizing myopathy; JDM, juvenile dermatomyositis; MMT, manual muscle testing; SRP, signal recognition particle. aLarger, international study in progress65.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
F O C U S O N MRYEO
V ISEI W
TIS

Anti-TNF therapies. Although one might expect ben- open-label, ‘delayed-start’ treatment trial were released;
eficial results with anti-TNF therapies in patients with this trial investigated abatacept treatment in 20 patients
myositis, the use of such therapies has had variable with either refractory dermatomyositis (n = 11) or refrac-
results. In a case series of five patients with dermato- tory polymyositis (n = 9), assessing both disease activity
myositis, treatment with etanercept resulted in worsen- and changes in muscle biopsy samples64. Patients received
ing of muscle weakness with persistently elevated creatine active treatment (monthly intravenous abatacept) either
kinase levels and no improvements in skin rash43. By con- immediately or after a 3-month delayed start, and the
trast, in a randomized, double-blind, placebo-controlled primary end point of the trial was the number of patients
trial of patients with dermatomyositis, treatment with achieving the IMACS DOI after 6 months of active treat-
etanercept was associated with a longer time to treatment ment. In this pilot trial, nearly half (42%) of the patients
failure and a greater prednisone-sparing effect than pla- achieved this primary end point, and in those patients in
cebo after 24 weeks44. Although a number of case reports whom a repeat muscle biopsy was performed, an increase
and case series have reported beneficial responses to in regulatory T cell markers was observed in the muscle
infliximab therapy in some patients with myositis45–48, in tissue at 6 months compared with levels before treatment,
a follow-up report of two such patients, their symptoms supporting a beneficial effect of this T cell-targeting ther-
worsened, and one of the patients developed anaphy- apy. There were no unusual safety signals in this trial, and
laxis on a second infusion of infliximab49. Furthermore, the therapy was well tolerated. These encouraging results
in an open-label pilot trial of 13 patients with refractory have led to an ongoing phase III clinical trial assessing
myositis, infliximab was ineffective50, and a subsequent abatacept therapy in patients with myositis65.
randomized double-blind placebo-controlled cross-
over clinical trial in 2017 investigating infliximab in 12 Anakinra. Anakinra, a recombinant IL-1 receptor antag-
patients with either polymyositis or dermatomyositis also onist (IL-1Ra), was investigated in a 2014 study of 15
revealed discouraging results, with the patients receiving patients with refractory myositis; in this study, the patients
infliximab treatment showing a response rate of <33% received anakinra (100 mg daily, subcutaneously) for
after 14 weeks51. However, infliximab was well tolerated 12 months66. Seven of the patients had a clinical response
in these studies, and whether a higher dose or longer according to the IMACS DOI, whereas four patients
follow-up might lead to better response rates is yet to be showed an improvement in function as measured by the
determined. Currently, the use of anti-TNF therapy can- functional index score. Post-treatment muscle biopsy
not be fully endorsed for patients with myositis, especially samples from all patients who met the response criteria
given that some studies report that these agents might demonstrated the expression of IL-1Ra whereas samples
cause myositis52–54. Nevertheless, in some patients with from only three of the eight non-responders demon-
a prominent and refractory inflammatory arthropathy, strated IL-1Ra expression. The investigators concluded
anti-TNF therapy should be considered. Further, there that patients with refractory myositis might respond
might be a role for anti-TNF inhibitors in the treatment to anakinra, noting that IL-1α expression might be
of calcinosis associated with juvenile dermatomyositis55. associated with a beneficial treatment response.

Tocilizumab. Following the approval of tocilizumab, an The role of immunomodulation


IL-6 receptor antagonist, for the treatment of rheumatoid IVIG suppresses immune-mediated processes, although
arthritis (RA), interest has grown in testing this drug in the precise mechanism of action of this immunomodula-
other autoimmune diseases. In two patients with refrac- tory agent is unknown. IVIG is usually administered as
tory polymyositis, treatment with tocilizumab resulted in a monthly infusion (2 g per kg), but the dose or interval
a decrease in creatine kinase levels along with the disap- of administration can vary depending on the disease
pearance of inflammatory changes in the thigh muscles severity and subsequent therapeutic response of the
as assessed by MRI56, whereas in a patient with an over- patient. IVIG therapy is beneficial when used in com-
lap syndrome involving dermatomyositis and systemic bination with other immunosuppressive agents and in
sclerosis who was refractory to multiple therapies, tocili- the setting of infection or malignancy, but this therapy
zumab treatment resulted in the resolution of skin symp- is also expensive and has a long administration time,
toms, a gradual decrease in serum creatine kinase levels dissuading its routine use.
and an improvement in muscle strength57. An investi- IVIG therapy demonstrated efficacy in treating
gator-initiated, multicentre, randomized, double-blind, myositis almost 25 years ago in a double-blind, cross-
controlled trial is ongoing to assess the efficacy of tocili- over, placebo-controlled trial of 15 patients with refrac-
zumab in treating adults with refractory polymyositis or tory dermatomyositis67 and was associated with sustained
dermatomyositis58 and uses the newly revised DOI59. clinical improvement in 70% of patients with poly-
myositis in an open-label trial68. Efficacy was maintained
Abatacept. Abatacept is a fully human fusion protein of in half of the patients up to 3 years after stopping IVIG
cytotoxic T lymphocyte protein 4 (CTLA4) and the Fc therapy 68. However, in a randomized, double-blind,
portion of human IgG1 that inhibits T cell co-stimulation. placebo-controlled trial in Japan involving 26 individuals
Although there have been a number of case reports (16 patients with polymyositis and 10 patients with der-
demonstrating the effectiveness of abatacept in treating matomyositis, who were refractory to glucocorticoids),
patients with myositis60–63, randomized controlled tri- patients receiving IVIG therapy failed to show improve-
als were lacking. In 2017, the results of a randomized, ments in muscle strength (as assessed by manual muscle

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 5

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
REVIEWS

testing) compared with those receiving placebo, although changes in gene expression, proteomics and capillary
other secondary end points did improve69. A pivotal density on repeat muscle biopsies. Taken together, these
double-blind, randomized, placebo-controlled, phase III clinical and experimental studies support the benefits
study is ongoing to confirm the efficacy and safety of of exercise as being anti-inflammatory in patients with
IVIG in patients with refractory dermatomyositis70. both active and stable myositis.
Administration of subcutaneous gamma-globulin by
a programmable pump in seven patients (four patients Special treatment considerations
with dermatomyositis and three patients with poly- As alluded to above, patients with myositis can present
myositis) led to a decrease in creatine kinase levels and an with various manifestations in clinical practice, includ-
improvement in muscle strength and quality of life, along ing necrotizing myopathy and extramuscular manifes-
with a beneficial steroid-sparing effect and a reduction in tations such as ILD, skin rash and dysphagia, which
concomitant immunosuppressive medication71. represent distinct scenarios. Such manifestations are
treated using the same agents already discussed in this
Exercise in the management of myositis Review, but a separate discussion is warranted given the
Historically, an active exercise programme would have severity and difficulty in managing these complications.
been discouraged in the management of myositis owing
to the rationale that exercise would damage the mus- Immune-mediated necrotizing myopathy
cles of a patient with myositis, perhaps inducing a form IMNM is often a more severe subset of myositis and
of secondary inflammation. However, there has been a is frequently associated with the presence of anti-
clear shift in this paradigm as emerging data support 3-hydroxy-3-methylglutaryl-coA reductase (HMGCR)77
the safety and effectiveness of exercise in adults with or anti-SRP autoantibodies. Patients with IMNM have
myositis. Physical exercise as a treatment for adult and prominent symmetrical proximal upper and lower
juvenile myositis has been comprehensively reviewed extremity weakness and very high levels of serum cre-
elsewhere72, and in this section, we highlight selected atine kinase but generally lack other extramuscular
aspects of exercise intervention. autoimmune features77. The initial treatment of patients
Exercise was originally reported to be safe in a 1998 with IMNM should involve an aggressive therapy that
study of patients with myositis with chronic and stable includes high-dose glucocorticoids in combination
disease activity 73; these patients showed improvements with another immunosuppressive agent. Case series of
in muscle strength and function after a 6-week exer- patients with this myositis subset have demonstrated
cise programme in which effects were assessed for up that IVIG and/or rituximab are effective and should
to 6 months73. The safety of exercise therapy was con- be considered early in the treatment of IMNM at the
firmed in patients with inactive myositis in another open same dosage recommended for the standard treatment
study in which a resistive home exercise programme of myositis37,78.
was implemented for 12 weeks74. A later study demon-
strated that a 12-week resistive programme, in combi- Interstitial lung disease
nation with conventional immunosuppressive therapy, ILD is a major cause of morbidity and mortality in
was safe in 11 patients with polymyositis or dermato- patients with myositis, and several drugs have been
myositis who had active and new-onset myositis75. For studied in the treatment of this complication. An
5 days each week, patients performed resistive exercise algorithmic approach to the treatment of myositis-
for 15 minutes followed by a 15-minute walk. No signs associated ILD is provided in FIG. 2. Treatment with
of increased inflammation were noted in these patients, MMF showed encouraging results in two small case
as assessed by creatine kinase levels, muscle MRI find- series of patients with myositis-associated ILD79,80 and
ings and on a repeat muscle biopsy compared with their was later investigated in a large cohort of 125 patients
original immunohistochemical results. Furthermore, with autoimmune ILD (32 with polymyositis or der-
muscle function and quality of life improved during matomyositis) who were treated for a mean period of
the 12-week exercise programme. Although the relative 897 days81. In this cohort, MMF treatment was associ-
effects of exercise and pharmacological therapy could ated with a trend towards improvement in the forced
not be distinguished, it was important to note that active vital capacity (FVC) and diffusing capacity of the lungs
exercise was well tolerated, with no adverse effects, in for carbon monoxide (DLCO) at 52 and 104 weeks. In
this small cohort of patients with active and recently a separate case study, MMF treatment was also benefi-
diagnosed myositis. cial in a patient with amyopathic dermatomyositis with
The molecular effects of exercise in polymyositis and rapidly progressive ILD82. Azathioprine treatment led
dermatomyositis were also studied in a controlled pilot to some improvement in lung function in a retrospec-
study; this study investigated the effects of a 12-week tive case series of patients with myositis-associated ILD
endurance training programme on skeletal muscle com- as well as in a cohort of 70 such patients (in which 25
pared with a stable level of physical activity 76. The exer- patients showed improvements)83,84.
cise programme activated an aerobic phenotype (that Cyclophosphamide use in myositis-associated ILD
is, the upregulation of molecular pathways involved in has primarily been reported retrospectively in case
aerobic capacity, capillary growth and muscle remodel- reports and case series85–87. In one case series, 11 of the 17
ling) while concomitantly mitigating the inflammatory patients given monthly intravenous cyclophosphamide
response in the patients’ muscles, as demonstrated by (300–800 mg/m2) for a minimum of 6 months showed

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
F O C U S O N MRYEO
V ISEI W
TIS

Mild to moderate disease Severe disease

Induction therapy

Oral glucocorticoids (such as prednisone, 1 mg per kg daily) Intravenous glucocorticoids (pulse therapy)
or and
Intravenous glucocorticoids (pulse therapy) Cyclophosphamide (intravenous) or rituximab

Maintenance therapy
Glucocorticoid-sparing drugs:
First-line Oral glucocorticoids and MMF or azathioprine
therapy:
Second-line Oral glucocorticoids and Tacrolimus or ciclosporin
therapy:

Refractory cases

Rituximab or cyclophosphamide or combination Combination of rituximab and cyclophosphamide


therapy of MMF and tacrolimus

Disease remains progressive

Abatacept or agents being tested in clinical trials

Figure 2 | Proposed approach to treating myositis-associated interstitial lung disease. The treatment of interstitial
lung disease (ILD) can be broken down into treating either mild to moderate disease or severe disease119. Treatment of
mild to moderate disease includes an aggressive approach involving high-dose glucocorticoids (either oral or intravenous
administration) as an induction therapy followed by maintenance therapy with slowly tapering Nature Reviews | Rheumatology
glucocorticoids and the
addition of any one of several immunosuppressive agents such as mycophenolate mofetil (MMF), azathioprine, tacrolimus
or ciclosporin. If mild to moderate disease is refractory to this induction or maintenance therapy, a more aggressive
approach with either rituximab or cyclophosphamide or a combination of MMF and tacrolimus is used. The management
of severe myositis-associated ILD includes induction therapy with pulse glucocorticoids along with more potent
immunosuppressive drugs, such as cyclophosphamide or rituximab, early on in the course of treatment followed by
maintenance therapy with MMF, tacrolimus or ciclosporin (or sometimes azathioprine). If severe disease is refractory to
induction or maintenance therapy, a more aggressive approach is used with a combination of rituximab and
cyclophosphamide. If disease is still progressive, then other biologic agents being investigated in clinical trials for myositis
are used, such as abatacept. Maintenance therapy of all forms of myositis-associated ILD depends on the response to
initial treatment and subsequent progression. In cases of relapse, high doses of steroids (oral, intravenous or pulse therapy,
depending on the disease severity) are given along with alternative immunosuppressive agents (not shown). Adapted from
REF. 119, Future Medicine.

improvements in their dyspnoea, and 6 of the 7 patients treatment with a combination of prednisolone and ciclo-
who had previously required oxygen treatment discontin- sporin (4 mg per kg daily within 12 days of diagnosis)
ued their supplemental oxygen87. Twelve of these patients improved both pulmonary function testing (PFT) and
had >10% improvement in their FVC and showed lung HRCT findings91. Similarly, in 48 Asian patients with
improvements as assessed by high-resolution CT (HRCT) dermatomyositis-associated ILD, early ciclosporin
scanning of the lung parenchyma. In addition, in a pro- treatment resulted in improved survival compared with
spective open-label study, cyclophosphamide treatment patients who received delayed ciclosporin treatment 92.
resulted in stabilization and functional improvement in In 13 patients with ILD who were positive for anti-
patients with progressive ILD, with a median follow-up synthetase autoantibodies, tacrolimus treatment for an
of 35 months88. IVIG use has also been described in case average of 51 months also resulted in improvements in
reports of patients with myositis-associated ILD89,90, with all PFT parameters93. In a retrospective study of 49 pre-
some patients, including one patient with amyopathic viously untreated patients with myositis-associated ILD,
dermatomyositis-associated ILD (who was refractory to treatment with tacrolimus plus conventional therapy led
high-dose glucocorticoids and ciclosporin A), showing a to longer event-free survival than treatment with con-
response to IVIG therapy 89,90. ventional therapy alone94. Furthermore, in three small
The notion that T cells are potential targets in the series of patients with myositis-associated ILD, in whom
management of ILD5–7 has led to the consideration ciclosporin was previously ineffective, tacrolimus treat-
of ciclosporin and tacrolimus for use in myositis- ment improved their lung disease93,95,96. With tacrolimus
associated ILD. In a retrospective study of 14 patients therapy, serum trough levels should be monitored to
with dermatomyositis and interstitial pneumonia, early limit toxicity.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 7

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
REVIEWS

Rituximab has been increasingly studied in patients ointments and creams, as well as topical calcineurin
with myositis-associated ILD, but no randomized con- inhibitors (tacrolimus and pimecrolimus), are consid-
trolled trials have been performed to date. In a retrospec- ered if the rash is mild and more limited. Antimalarials
tive study of 50 patients with ILD associated with various are occasionally beneficial and used in combination
autoimmune diseases, rituximab therapy was most effec- with other immunosuppressive agents, but hydroxy-
tive in patients with myositis-associated ILD out of all the chloroquine use can lead to a diffuse erythroderma in
patients, with 5 of the 10 such patients demonstrating some patients104,105. In most patients, treatment of the
an improvement in FVC (>10%) and/or an increase in rash of dermatomyositis often requires a therapy that is
DLCO (>15%)97. In another retrospective report of 24 just as aggressive as the therapy required for targeting
antisynthetase autoantibody-positive patients with severe muscle weakness in myositis; hence, the use of metho-
ILD, who were followed for >12 months, after rituximab trexate, azathioprine, MMF, IVIG and tacrolimus is rec-
therapy, the patients demonstrated a median 24% increase ommended in some instances. Two uncontrolled case
in FVC, a 22% increase in forced expiratory volume in series suggest that MMF is effective in treating refrac-
1 second (FEV1) and a 17% increase in DLCO98. The best tory cutaneous dermatomyositis106,107. In the RIM trial,
outcomes were noted in patients with a disease duration rituximab use was associated with notable improvements
of <12 months and/or in patients who had an acute onset in cutaneous disease activity in adult and juvenile der-
or exacerbation of their ILD. A limitation of this study matomyositis subsets, including notable improvements
was the concomitant administration of another immuno- in erythroderma, erythematous rashes (without second-
suppressive agent; 10 of the 12 patients also received cyclo- ary changes of ulceration or necrosis), heliotrope rash,
phosphamide, preventing the attribution of improvement Gottron sign and Gottron papules108. In 2016, tofacitinib
to rituximab alone. In a multicentre, open-label trial of was reported to be effective in treating three patients with
ten antisynthetase autoantibody-positive patients with dermatomyositis who had multidrug-resistant cutaneous
refractory ILD, treatment with rituximab on day 0, day manifestations109, and tofacitinib treatment was simi-
15 and then 6 months later resulted in a substantial ster- larly associated with improvements in skin, joint and
oid-sparing effect along with reductions in the serum muscle features in another patient with refractory
creatine kinase levels and improvements in manual dermatomyositis in a subsequent case report110.
muscle testing. ILD improvement (that is, increases in Calcinosis is probably the most challenging and diffi-
FVC and/or DLCO) was noted in five of these patients, cult cutaneous feature to treat in myositis. The presence
and FVC stabilized in four of the patients99. In another of anti-nuclear matrix protein 2 (NXP2; also known as
retrospective study, 16 of the 17 anti-Jo1-positive MORC3) autoantibodies is associated with calcinosis in
patients who received rituximab therapy demonstrated both adult and juvenile dermatomyositis111. Many med-
a more rapid and marked response (as assessed by ications have been tried, but none is uniformly effective,
myositis and ILD outcomes) than the 30 patients who and surgical excision might be the only option to con-
were treated with conventional immunosuppressive sider in severe cases. In a case series, bisphosphonates in
agents100. Rituximab is usually administered as two 1 g combination with immunosuppressive therapy and/or
doses 2 weeks apart, but the subsequent dosing inter- IVIG reduced calcinosis in four of the six patients with
val might vary, and no regimen has been standardized juvenile dermatomyositis112. Sodium thiosulfate ther-
for the treatment of myositis with or without ILD. apy, administered either intravenously or directly into
A randomized pilot trial assessing the effect of aba- the calcinotic lesions, has been associated with some
tacept is under way in the treatment of antisynthetase improvement in skin outcomes in patients with adult or
autoantibody-positive patients101. Methotrexate was juvenile dermatomyositis113,114.
often considered to be contraindicated in patients posi-
tive for the anti-Jo1 autoantibody or other antisynthetase Dysphagia in myositis
autoantibodies owing to its potential for inducing pul- The proximal dysphagia seen in many patients with
monary toxicity. However, there has been a shift in recent severe myositis is a stubborn and severe manifestation.
years with the finding that methotrexate is highly effec- Although many patients respond to glucocorticoids and
tive in treating muscle and articular features of patients other immunosuppressive agents, additional IVIG should
with antisynthetase syndrome. Moreover, the reported also always be considered for dysphagia. In a retrospective
small increase in risk of methotrexate-induced pneu- study of 73 patients with either polymyositis or dermato-
monitis is primarily seen in patients with RA102,103. Thus, myositis who had steroid-refractory life-threatening
it is reasonable to administer methotrexate as long as oesophageal dysphagia, a first-line therapy of combined
myositis-associated ILD is not the primary manifestation high-dose glucocorticoids and IVIG was beneficial115.
being treated. As with the standard treatment of myositis, IVIG can
be administered by monthly infusions of 2 g per kg over
Skin rash in dermatomyositis 2 consecutive days or over a 5-day period if necessary.
The cutaneous features of dermatomyositis can be quite
difficult to treat and, in some patients, might represent Future prospects
the dominant disease manifestation. Simple measures of There are a number of potential and investigational
treating dermatomyositis include wearing protective sun- agents to be considered in the treatment of myositis
screen and avoiding the use of medications that increase (FIG. 3). The prominent expression of a type I interferon
photosensitivity. Occasionally, topical glucocorticoid signature in the serum and tissues of patients with

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
F O C U S O N MRYEO
V ISEI W
TIS

Genetic factors Environmental factors


• HLA • Injury • Infection
• Non-HLA • Toxins • UV light

Innate immunity
IMO-8400 • Toll-like receptors • Dendritic cells
• Macrophages • Chemokines

Abatacept
APC

CD40L CD80/
Rituximab TFH MHC II MHC I
CD40 CD86
CD28 TCR

B cell CD4 CD8

BAFF APRIL
TH2 TH17

TH1
Belimumab
IL-4 IL-23 IL-17

IFNα
Plasma cell Secukinumab

• Sifalimumab
• Anifrolumab
Autoantibodies
Cytotoxic
M1/M2 macrophage T cell

• Canakinumab
TNF IL-6 IL-1β • Anakinra

Anti-TNF Tocilizumab
Immune
complexes
Cytokines
and
chemokines
?
Immune complex Capillaries and myocytes Cell-mediated
IVIG deposition damage

Figure 3 | Immune-related potential therapeutic targets in myositis. There are several factors involved in the
pathogenesis of myositis that lead to the involvement or activation of both the innate and adaptive arms of the immune
system as well as non-immune mechanisms (not shown). Many of the current immunosuppressive agents studied in
myositis affect these immune pathways, whereas the biologic agents in this figure function in a more targeted fashion
to neutralize selected areas of these immune-mediated pathways. Noted in the figure are agents previously and
currently being studied in the treatment of myositis as well as those proposed in future clinical trials. APC,
antigen-presenting cell; APRIL, a proliferation-inducing ligand (also known as TNFSF13); BAFF, B cell activating
factor; CD40L, CD40 ligand; IVIG, intravenous immunoglobulin; TCR, T cell receptor; TFH, T follicular helper cell;
TH, T helper cell.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 9

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
REVIEWS

dermatomyositis or polymyositis led to the study of Conclusions


sifalimumab, an anti-IFNα monoclonal autoantibody, in The future of myositis treatment includes many poten-
treating these patients. In a phase Ib randomized, double- tial therapies. The immunosuppressive agents discussed
blind, controlled trial, investigators noted a suppression in this Review will certainly continue to have a major
of the interferon signature in both the serum and mus- role in myositis therapy, and biologic therapies will
cle of individuals with dermatomyositis or polymyositis probably have an increasing role given that these ther-
following sifalimumab treatment 116. Future studies of apies target pathogenic pathways implicated in myosi-
anti-interferon agents are being considered. Similarly, tis. Investigators must use the technologies available,
the blockade of interferon-mediated activation of Janus including advanced immunochemistry, microarray and
kinase (JAK)–signal transducer and activator of tran- RNA sequencing analysis, cytokine and chemokine pan-
scription (STAT) pathways with oral JAK inhibitors is els and sophisticated flow cytometry-based techniques,
worthy of further study, particularly regarding dermat- to determine logical therapeutic targets. Although we
omyositis and the refractory cutaneous features117. The live in an exciting time in terms of myositis treatment
effect of blocking TLRs is currently being assessed in interventions, we must be cautious of the potential
dermatomyositis in a multicentre international study 118. short-term and long-term effects of these novel agents.

1. Rider, L. G. et al. Update on outcome assessment in 18. Aggarwal, R. et al. Efficacy and safety of 37. Valiyil, R. et al. Rituximab therapy for myopathy
myositis. Nat. Rev. Rheumatol. https://doi. adrenocorticotropic hormone gel in refractory associated with anti-signal recognition particle
org/10.1038/nrrheum.2018.33 (2018). dermatomyositis and polymyositis. Ann. Rheum. Dis. antibodies: a case series. Arthritis Care Res. 62,
2. Greenberg, S. A. et al. Myeloid dendritic cells in https://doi.org/10.1136/annrheumdis-2017-212047 1328–1334 (2010).
inclusion-body myositis and polymyositis. Muscle (2017). 38. Mok, C. C., Ho, L. Y. & To, C. H. Rituximab for
Nerve 35, 17–23 (2007). 19. Joffe, M. M. et al. Drug therapy of the idiopathic refractory polymyositis: an open-label prospective
3. Greenberg, S. A. et al. Interferon-alpha/beta-mediated inflammatory myopathies: predictors of response to study. J. Rheumatol. 34, 1864–1868 (2007).
innate immune mechanisms in dermatomyositis. prednisone, azathioprine, and methotrexate and a 39. Chung, L., Genovese, M. C. & Fiorentino, D. F. A pilot
Ann. Neurol. 57, 664–678 (2005). comparison of their efficacy. Am. J. Med. 94, 379–387 trial of rituximab in the treatment of patients with
4. Bilgic, H. et al. Interleukin-6 and type I interferon- (1993). dermatomyositis. Arch. Dermatol. 143, 763–767
regulated genes and chemokines mark disease activity 20. Newman, E. D. & Scott, D. W. The use of low-dose (2007).
in dermatomyositis. Arthritis Rheum. 60, 3436–3446 methotrexate in the treatment of pollymyositis and 40. Oddis, C. V. et al. Rituximab in the treatment of
(2009). dermatomyositis. J. Clin. Rheumatol 1, 99–102 (1995). refractory adult and juvenile dermatomyositis and
5. Kurasawa, K. et al. Activation of pulmonary T cells in 21. Ruperto, N. et al. Prednisone versus prednisone plus adult polymyositis: a randomized, placebo-phase trial.
corticosteroid-resistant and -sensitive interstitial ciclosporin versus prednisone plus methotrexate in Arthritis Rheum. 65, 314–324 (2013).
pneumonitis in dermatomyositis/polymyositis. new-onset juvenile dermatomyositis: a randomised 41. Rider, L. G. et al. International consensus on
Clin. Exp. Immunol. 129, 541–548 (2002). trial. Lancet 387, 671–678 (2016). preliminary definitions of improvement in adult and
6. Yamadori, I. et al. Lymphocyte subsets in lung tissues 22. US National Library of Medicine. ClinicalTrials.gov juvenile myositis. Arthritis Rheum. 50, 2281–2290
of interstitial pneumonia associated with untreated https://clinicaltrials.gov/ct2/show/NCT00651040 (2004).
polymyositis/dermatomyositis. Rheumatol. Int. 21, (2016). 42. Aggarwal, R. et al. Predictors of clinical improvement
89–93 (2001). 23. Joffe, M. M. et al. Drug therapy of the idiopathic in rituximab-treated refractory adult and juvenile
7. Katagiri, A. et al. Decrease in CD4+CD25+ and inflammatory myopathies: predictors of response to dermatomyositis and adult polymyositis. Arthritis
CD8+CD28+ T cells in interstitial pneumonitis prednisone, azaathioprine, and methotrexate and a Rheumatol. 66, 740–749 (2014).
associated with rheumatic disease. Mod. Rheumatol. comparison of their efficacy. Am. J. Med. 94, 379–387 43. Iannone, F. et al. Use of etanercept in the treatment of
18, 562–569 (2008). (1993). dermatomyositis: a case series. J. Rheumatol. 33,
8. Notarnicola, A. et al. Possible interplay between 24. Bunch, T. W. Prednisone and azathioprine for 1802–1804 (2006).
interleukin-15 and interleukin-17 into the polymyositis: long-term followup. Arthritis Rheum. 24, 44. Muscle Study, G. A randomized, pilot trial of
pathogenesis of idiopathic inflammatory myopathies. 45–48 (1981). etanercept in dermatomyositis. Ann. Neurol. 70,
Reumatismo 66, 215–223 (2014). 25. Bunch, T. W. et al. Azathioprine with prednisone for 427–436 (2011).
9. Shen, H. et al. Interleukin-17 and interleukin-23 in polymyositis. A controlled, clinical trial. Ann. Intern. 45. Anandacoomarasamy, A., Howe, G. & Manolios, N.
patients with polymyositis and dermatomyositis. Med. 92, 365–369 (1980). Advanced refractory polymyositis responding to
Scand. J. Rheumatol. 40, 217–220 (2011). 26. Villalba, L. et al. Treatment of refractory myositis: a infliximab. Rheumatology 44, 562–563 (2005).
10. Tournadre, A., Lenief, V. & Miossec, P. Expression of randomized crossover study of two new cytotoxic 46. Efthimiou, P., Schwartzman, S. & Kagen, L. J. Possible
Toll-like receptor 3 and Toll-like receptor 7 in muscle is regimens. Arthritis Rheum. 41, 392–399 (1998). role for tumour necrosis factor inhibitors in the
characteristic of inflammatory myopathy and is 27. Majithia, V. & Harisdangkul, V. Mycophenolate mofetil treatment of resistant dermatomyositis and
differentially regulated by Th1 and Th17 cytokines. (CellCept): an alternative therapy for autoimmune polymyositis: a retrospective study of eight patients.
Arthritis Rheum. 62, 2144–2151 (2010). inflammatory myopathy. Rheumatology 44, 386–389 Ann. Rheum. Dis. 65, 1233–1236 (2006).
11. Nagaraju, K. Update on immunopathogenesis in (2005). 47. Hengstman, G. J. et al. Successful treatment of
inflammatory myopathies. Curr. Opin. Rheumatol. 13, 28. Pisoni, C. N. et al. Mycophenolate mofetil treatment dermatomyositis and polymyositis with anti-tumor-
461–468 (2001). in resistant myositis. Rheumatology 46, 516–518 necrosis-factor-alpha: preliminary observations.
12. Englund, P. et al. Skeletal muscle fibers express (2007). Eur. Neurol. 50, 10–15 (2003).
major histocompatibility complex class II antigens 29. Rowin, J. et al. Mycophenolate mofetil in 48. Selva-O’Callaghan, A. et al. Refractory adult
independently of inflammatory infiltrates in dermatomyositis: is it safe? Neurology 66, 1245–1247 dermatomyositis with pneumatosis cystoides
inflammatory myopathies. Am. J. Pathol. 159, (2006). intestinalis treated with infliximab. Rheumatology 43,
1263–1273 (2001). 30. Schneider, C. et al. Mycophenolate mofetil in the therapy 1196–1197 (2004).
13. Nagaraju, K. et al. Activation of the endoplasmic of polymyositis associated with a polyautoimmune 49. Hengstman, G. J., F. H. van den Hoogen & van
reticulum stress response in autoimmune myositis: syndrome. Muscle Nerve 25, 286–288 (2002). Engelen, B. G. Treatment of dermatomyositis and
potential role in muscle fiber damage and dysfunction. 31. Danieli, M. G. et al. Intravenous immunoglobulin as polymyositis with anti-tumor necrosis factor-alpha:
Arthritis Rheum. 52, 1824–1835 (2005). add on treatment with mycophenolate mofetil in long-term follow-up. Eur. Neurol. 52, 61–63
14. Lightfoot, A. P. et al. In the idiopathic inflammatory severe myositis. Autoimmun. Rev. 9, 124–127 (2009). (2004).
myopathies (IIM), do reactive oxygen species (ROS) 32. Oddis, C. V. et al. Tacrolimus in refractory polymyositis 50. Dastmalchi, M. et al. A high incidence of disease flares
contribute to muscle weakness? Ann. Rheum. Dis. 74, with interstitial lung disease. Lancet 353, 1762–1763 in an open pilot study of infliximab in patients with
1340–1346 (2015). (1999). refractory inflammatory myopathies. Ann. Rheum. Dis.
15. Catania, A. et al. The melanocortin system in control of 33. Mitsui, T. et al. The effects of FK506 on refractory 67, 1670–1677 (2008).
inflammation. ScientificWorldJournal 10, 1840–1853 inflammatory myopathies. Acta Neurol. Belg. 111, 51. Schiffenbauer, A. et al. A randomized, double-blind,
(2010). 188–194 (2011). placebo-controlled trial of infliximab in refractory
16. Levine, T. Treating refractory dermatomyositis or 34. Lambotte, O. et al. Efficacy of rituximab in refractory polymyositis and dermatomyositis. Semin. Arthritis
polymyositis with adrenocorticotropic hormone gel: polymyositis. J. Rheumatol 32, 1369–1370 (2005). Rheum. https://doi.org/10.1016/j.
a retrospective case series. Drug Des. Devel. Ther. 6, 35. Levine, T. D. Rituximab in the treatment of semarthrit.2017.10.010 (2017).
133–139 (2012). dermatomyositis: an open-label pilot study. Arthritis 52. Ishikawa, Y. et al. Etanercept-induced
17. Patel, A., Seely, G. & Aggarwal, R. Repository Rheum. 52, 601–607 (2005). anti-Jo-1-antibody-positive polymyositis in a patient
corticotropin injection for treatment of idiopathic 36. Mahler, E. A. et al. Rituximab treatment in patients with rheumatoid arthritis: a case report and review
inflammatory myopathies. Case Rep. Rheumatol. with refractory inflammatory myopathies. of the literature. Clin. Rheumatol. 29, 563–566
2016, 9068061 (2016). Rheumatology 50, 2206–2213 (2011). (2010).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ
F O C U S O N MRYEO
V ISEI W
TIS

53. Klein, R. et al. Tumor necrosis factor inhibitor- 77. Mammen, A. L. et al. Autoantibodies against 99. Allenbach, Y. et al. Efficacy of rituximab in refractory
associated dermatomyositis. Arch. Dermatol. 146, 3-hydroxy-3-methylglutaryl-coenzyme A reductase in inflammatory myopathies associated with anti-
780–784 (2010). patients with statin-associated autoimmune synthetase auto-antibodies: an open-label, phase II
54. Riolo, G. & Towheed, T. E. Anti-tumor necrosis factor myopathy. Arthritis Rheum. 63, 713–721 trial. PLoS ONE 10, e0133702 (2015).
inhibitor therapy-induced dermatomyositis and (2011). 100. Bauhammer, J. et al. Rituximab in the treatment of
fasciitis. J. Rheumatol. 39, 192–194 (2012). 78. Mammen, A. L. & Tiniakou, E. Intravenous immune Jo1 antibody-associated antisynthetase syndrome:
55. Riley, P. et al. Effectiveness of infliximab in the globulin for statin-triggered autoimmune Anti-Ro52 positivity as a marker for severity and
treatment of refractory juvenile dermatomyositis with myopathy. N. Engl. J. Med. 373, 1680–1682 treatment response. J. Rheumatol. 43, 1566–1574
calcinosis. Rheumatology 47, 877–880 (2008). (2015). (2016).
56. Narazaki, M. et al. Therapeutic effect of tocilizumab 79. Morganroth, P. A., Kreider, M. E. & Werth, V. P. 101. US National Library of Medicine. ClinicalTrials.gov
on two patients with polymyositis. Rheumatology 50, Mycophenolate mofetil for interstitial lung disease in https://clinicaltrials.gov/ct2/show/NCT03215927
1344–1346 (2011). dermatomyositis. Arthritis Care Res. 62, 1496–1501 (2017).
57. Kondo, M. et al. A case of overlap syndrome (2010). 102. Conway, R. et al. Methotrexate and lung disease in
successfully treated with tocilizumab: a hopeful 80. Mira-Avendano, I. C. et al. A retrospective review of rheumatoid arthritis: a meta-analysis of randomized
treatment strategy for refractory dermatomyositis? clinical features and treatment outcomes in steroid- controlled trials. Arthritis Rheumatol. 66, 803–812
Rheumatology 53, 1907–1908 (2014). resistant interstitial lung disease from polymyositis/ (2014).
58. US National Library of Medicine. ClinicalTrials.gov dermatomyositis. Respir. Med. 107, 890–896 103. Conway, R. et al. Methotrexate use and risk of lung
https://clinicaltrials.gov/ct2/show/NCT02043548 (2013). disease in psoriasis, psoriatic arthritis, and
(2017). 81. Fischer, A. et al. Mycophenolate mofetil improves lung inflammatory bowel disease: systematic literature
59. Aggarwal, R. et al. 2016 American College of function in connective tissue disease-associated review and meta-analysis of randomised controlled
Rheumatology/European League Against Rheumatism interstitial lung disease. J. Rheumatol. 40, 640–646 trials. BMJ 350, h1269 (2015).
criteria for minimal, moderate, and major clinical (2013). 104. Pai, S. B. et al. Hydroxychloroquine-induced
response in adult dermatomyositis and polymyositis: 82. Tsuchiya, H. et al. Mycophenolate mofetil therapy for erythroderma. Indian J. Pharmacol. 49, 132–134
an International Myositis Assessment and Clinical rapidly progressive interstitial lung disease in a (2017).
Studies Group/Paediatric Rheumatology International patient with clinically amyopathic dermatomyositis. 105. Slagel, G. A. & James, W. D. Plaquenil-induced
Trials Organisation Collaborative initiative. Arthritis Mod. Rheumatol. 24, 694–696 (2014). erythroderma. J. Am. Acad. Dermatol. 12, 857–862
Rheumatol. 69, 898–910 (2017). 83. Douglas, W. W. et al. Polymyositis-dermatomyositis- (1985).
60. Arabshahi, B. et al. Abatacept and sodium thiosulfate associated interstitial lung disease. Ann. J. Respir. 106. Edge, J. C. et al. Mycophenolate mofetil as an effective
for treatment of recalcitrant juvenile dermatomyositis Crit. Care Med. 164, 1182–1185 (2001). corticosteroid-sparing therapy for recalcitrant
complicated by ulceration and calcinosis. J. Pediatr. 84. Marie, I. et al. Interstitial lung disease in polymyositis dermatomyositis. Arch. Dermatol. 142, 65–69
160, 520–522 (2012). and dermatomyositis. Arthritis Rheum. 47, 614–622 (2006).
61. Kerola, A. M. & Kauppi, M. J. Abatacept as a (2002). 107. Gelber, A. C., Nousari, H. C. & Wigley, F. M.
successful therapy for myositis-a case-based review. 85. Kameda, H. et al. Combination therapy with Mycophenolate mofetil in the treatment of severe
Clin. Rheumatol. 34, 609–612 (2015). corticosteroids, cyclosporin A, and intravenous pulse skin manifestations of dermatomyositis: a series of 4
62. Maeshima, K. et al. Successful treatment of refractory cyclophosphamide for acute/subacute interstitial cases. Rheumatol. J. 27, 1542–1545 (2000).
anti-signal recognition particle myopathy using pneumonia in patients with dermatomyositis. 108. Aggarwal, R. et al. Cutaneous improvement in
abatacept. Rheumatology 53, 379–380 (2014). J. Rheumatol. 32, 1719–1726 (2005). refractory adult and juvenile dermatomyositis after
63. Musuruana, J. L. & Cavallasca, J. A. Abatacept for 86. Mok, C. C., To, C. H. & Szeto, M. L. Successful treatment with rituximab. Rheumatology 56,
treatment of refractory polymyositis. Joint Bone Spine treatment of dermatomyositis-related rapidly 247–254 (2017).
78, 431–432 (2011). progressive interstitial pneumonitis with sequential 109. Kurtzman, D. J. et al. Tofacitinib citrate for refractory
64. Tjarnlund, A. et al. Abatacept in the treatment of oral cyclophosphamide and azathioprine. Scand. cutaneous dermatomyositis: an alternative treatment.
adult dermatomyositis and polymyositis: a J. Rheumatol. 32, 181–183 (2003). JAMA Dermatol. 152, 944–945 (2016).
randomised, phase IIb treatment delayed-start trial. 87. Yamasaki, Y. et al. Intravenous cyclophosphamide 110. Paik, J. J. & Christopher-Stine, L. A case of refractory
Ann. Rheum. Dis. 77, 55–62 (2018). therapy for progressive interstitial pneumonia in dermatomyositis responsive to tofacitinib. Semin.
65. US National Library of Medicine. ClinicalTrials.gov patients with polymyositis/dermatomyositis. Arthritis Rheum. 46, e19 (2017).
https://clinicaltrials.gov/ct2/show/NCT02971683 Rheumatology 46, 124–130 (2007). 111. Rogers, A. et al. Cutaneous and systemic findings
(2018). 88. Schnabel, A. et al. Interstitial lung disease in associated with nuclear matrix protein 2 antibodies in
66. Zong, M. et al. Anakinra treatment in patients with polymyositis and dermatomyositis: clinical course and adult dermatomyositis patients. Arthritis Care Res.
refractory inflammatory myopathies and possible response to treatment. Semin. Arthritis Rheum. 32, 69, 1909–1914 (2017).
predictive response biomarkers: a mechanistic study 273–284 (2003). 112. Tayfur, A. C. et al. Bisphosphonates in juvenile
with 12 months follow-up. Ann. Rheum. Dis. 73, 89. Bakewell, C. J. & Raghu, G. Polymyositis associated with dermatomyositis with dystrophic calcinosis. Mod.
913–920 (2014). severe interstitial lung disease: remission after three Rheumatol. 25, 615–620 (2015).
67. Dalakas, M. C. et al. A controlled trial of high-dose doses of IV immunoglobulin. Chest 139, 441–443 113. Pagnini, I. et al. Sodium thiosulfate for the treatment
intravenous immune globulin infusions as treatment (2011). of calcinosis secondary to juvenile dermatomyositis.
for dermatomyositis. N. Engl. J. Med. 329, 90. Suzuki, Y. et al. Intravenous immunoglobulin Clin. Exp. Rheumatol. 32, 408–409 (2014).
1993–2000 (1993). therapy for refractory interstitial lung disease 114. Smith, G. P., Intradermal sodium thiosulfate for
68. Cherin, P. et al. Results and long-term followup of associated with polymyositis/dermatomyositis. exophytic calcinosis cutis of connective tissue disease.
intravenous immunoglobulin infusions in chronic, Lung 187, 201–206 (2009). J. Am. Acad. Dermatol. 69, e146–147 (2013).
refractory polymyositis: an open study with thirty-five 91. Kotani, T. et al. Combination with corticosteroids 115. Marie, I. et al. Intravenous immunoglobulins for
adult patients. Arthritis Rheum. 46, 467–474 and cyclosporin-A improves pulmonary function steroid-refractory esophageal involvement related to
(2002). test results and chest HRCT findings in polymyositis and dermatomyositis: a series of 73
69. Miyasaka, N. et al. Effects of intravenous dermatomyositis patients with acute/subacute patients. Arthritis Care Res. 62, 1748–1755
immunoglobulin therapy in Japanese patients with interstitial pneumonia. Clin. Rheumatol. 30, (2010).
polymyositis and dermatomyositis resistant to 1021–1028 (2011). 116. Higgs, B. W. et al. A phase 1b clinical trial evaluating
corticosteroids: a randomized double-blind placebo- 92. Go, D. J. et al. Survival benefit associated with early sifalimumab, an anti-IFN-alpha monoclonal antibody,
controlled trial. Mod. Rheumatol. 22, 382–393 (2012). cyclosporine treatment for dermatomyositis- shows target neutralisation of a type I IFN signature in
70. US National Library of Medicine. ClinicalTrials.gov associated interstitial lung disease. Rheumatol. Int. blood of dermatomyositis and polymyositis patients.
https://clinicaltrials.gov/ct2/show/NCT02728752 36, 125–131 (2016). Ann. Rheum. Dis. 73, 256–262 (2014).
(2018). 93. Wilkes, M. R. et al. Treatment of antisynthetase- 117. US National Library of Medicine. ClinicalTrials.gov
71. Danieli, M. G. et al. Subcutaneous immunoglobulin in associated interstitial lung disease with tacrolimus. https://clinicaltrials.gov/ct2/show/NCT03002649
polymyositis and dermatomyositis: a novel Arthritis Rheum. 52, 2439–2446 (2005). (2018).
application. Autoimmun. Rev. 10, 144–149 (2011). 94. Kurita, T. et al. The efficacy of tacrolimus in patients 118. US National Library of Medicine. ClinicalTrials.gov
72. Alexanderson, H. Physical exercise as a treatment for with interstitial lung diseases complicated with https://clinicaltrials.gov/ct2/show/NCT02612857
adult and juvenile myositis. J. Intern. Med. 280, polymyositis or dermatomyositis. Rheumatology 54, (2017).
75–96 (2016). 39–44 (2015). 119. Moghadam-Kia, S., Oddis, C. V. & Aggarwal, R.
73. Wiesinger, G. F. et al. Improvement of physical fitness 95. Ochi, S. et al. Favorable outcomes with tacrolimus in Update on the treatment of myositis. Int. J. Clin.
and muscle strength in polymyositis/dermatomyositis two patients with refractory interstitial lung disease Rheumatol. 9, 505–518 (2014).
patients by a training programme. Br. J. Rheumatol. associated with polymyositis/dermatomyositis. Clin.
37, 196–200 (1998). Exp. Rheumatol. 23, 707–710 (2005). Author contributions
74. Alexanderson, H., Stenstrom, C. H. & Lundberg, I. 96. Takada, K., Nagasaka, K. & Miyasaka, N. Both authors wrote the article, provided substantial contri-
Safety of a home exercise programme in patients with Polymyositis/dermatomyositis and interstitial lung butions to discussions of its content and undertook review
polymyositis and dermatomyositis: a pilot study. disease: a new therapeutic approach with T-cell- and/or editing of the manuscript before submission.
Rheumatology 38, 608–611 (1999). specific immunosuppressants. Autoimmunity 38,
75. Alexanderson, H. et al. The safety of a resistive home 383–392 (2005). Competing interests
exercise program in patients with recent onset active 97. Keir, G. J. et al. Rituximab in severe, treatment- Both C.V.O. and R.A. receive clinical trial support from
polymyositis or dermatomyositis. Scand. refractory interstitial lung disease. Respirology 19, Genentech, Idera Pharmaceuticals, Bristol-Myers Squibb and
J. Rheumatol. 29, 295–301 (2000). 353–359 (2014). Mallinckrodt.
76. Munters, L. A. et al. Endurance exercise improves 98. Andersson, H. et al. Long-term experience with
molecular pathways of aerobic metabolism in patients rituximab in anti-synthetase syndrome-related Publisher’s note
with myositis. Arthritis Rheumatol. 68, 1738–1750 interstitial lung disease. Rheumatology 54, Springer Nature remains neutral with regard to jurisdictional
(2016). 1420–1428 (2015). claims in published maps and institutional affiliations.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 11

ǟ ƐƎƏƘ !,(++- 4 +(2'#12 (,(3#"Ʀ /13 .$ /1(-%#1 341#ƥ ++ 1(%'32 1#2#15#"ƥ
ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ ɥ

You might also like