Review Article

RMMJ Rambam Maimonides Medical Journal Rambam Health Care Campus 2023 April; 14(2): e0008. ISSN: 2076-9172
Published online 2023 April 30. doi: 10.5041/RMMJ.10495

Biological Therapies in Inflammatory Myopathies

Abd El Haleem Natour, M.D.1,2 and Shaye Kivity, M.D.1

1Rheumatology Unit, Meir Medical Center, Kfar Saba, Israel
2Department of Internal Medicine A, Meir Medical Center, Kfar Saba, Israel

Guest Editor Alexandra Balbir-Gurman, M.D.

*To whom correspondence should be addressed. E-mail: kivitys@gmail.com

Abstract

Idiopathic inflammatory myopathies (IIM) are a rare group of disorders that feature progressive immune-mediated skeletal muscle destruction along with skin, lung, and joint involvement. Management of IIMs necessitates glucocorticoid therapy followed by conventional steroid-sparing agents to control disease activity. In the settings of refractory myositis or life-threatening manifestations, e.g. lung involvement or oropharyngeal dysphagia, second-line therapies are needed to minimize disease burden, avoid end-organ damage and steroid toxicity, and decrease mortality. These therapies may include biological disease-modifying antirheumatic drugs (bDMARDs), and to a lesser extent, targeted synthetic disease-modifying antirheumatic drugs (TSD). This article reviews the current use of bDMARDs, e.g. intravenous immunoglobulin and rituximab, and a TSD—Janus kinase inhibitors (JAKI)—along with their indications, efficacy, and safety in managing IIM.

Keywords: Abatacept, bDMARDs, idiopathic inflammatory myopathies, IVIg, JAKI, rituximab

INTRODUCTION

The idiopathic inflammatory myopathies (IIM) represent a rare group of disorders marked by progressive immune-mediated skeletal muscle destruction together with skin, lung, and joint involvement. Organ involvement and disease severity tend to differ in IIM. While in some cases the disease course can be monophasic and short-lived, IIM generally involves persistent disease activity that waxes and wanes. Studies have shown that specific disease phenotypes often have distinctive serologic laboratory findings that may correlate with the disease course and prognosis of patients with IIM. Treatment is based on glucocorticoid therapy followed by steroid-sparing agents such as azathioprine, methotrexate, and mycophenolate mofetil to control disease activity. Refractory myositis can be defined as inadequate response to at least two steroid-sparing agents for a minimum period of 3 months.1 In this setting, to minimize disease burden, avoid end-organ damage and steroid toxicity, and decrease mortality, second-line therapies are needed. These may include biological disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (TSD). This article reviews the current use of bDMARDs and TSD, including their indications, efficacy, and safety, in managing IIM.

INTRAVENOUS IMMUNOGLOBULIN USE IN IIM

Intravenous immunoglobulin (IVIg), a purified liquid IgG concentrated from human plasma, has been approved by the US Food and Drug Administration (FDA) for treating adults with dermatomyositis (DM). In high-risk patients (e.g. dysphagia, severe weakness), IVIg may be used as a first-line treatment. Several mechanisms of action have been proposed for the beneficial effect of IVIg in IIM. These include: (1) inhibition of complement activation and capillary membrane attack complex (MAC) deposition;2,3 (2) downregulation of genes related to inflammation, e.g. IL-2, KAL-1, ICAM-1, C1q;4 (3) upregulation of chemokines CXCL9 and CXCL11-related genes;4 and, finally, (4) blocking the Fc-receptors on autoantibodies that prevent antibody-coated cell phagocytosis.5,6

In 1993 Dalakas et al. reported that IVIg improved muscle strength and reduced neuromuscular symptoms in a randomized controlled trial of 15 patients.7 Later on, in an open-label study from 2002 in which IVIg was used in 35 patients, 50% showed marked improvement in disease activity, with durable efficacy over three years.8 In Aggarwal et al.’s recent randomized controlled trial (ProDERM trial), refractory IIM patients or those receiving concomitant glucocorticoid and immunosuppressive medication saw significant improvement with IVIg, administered at a dose of 2.0 g per kg of body weight. At 16 weeks, 79% of the patients in the IVIg group (37 of 47) and 44% of those in the placebo group (21 of 48) showed at least slight improvement on a composite score of disease activity (P<0.001). Additionally, the median time to at least modest improvement with IVIg was 35 days, while the median time with placebo was 115 days.9 Intravenous Ig was also reported to be highly effective in the setting of myositis-related dysphagia, a life-threatening myositis manifestation. In a retrospective analysis, 12 of 18 patients treated with IVIg for refractory dysphagia had completely recovered by week 52.10 Furthermore, IVIg was reported to be particularly effective in treating cutaneous dermatomyositis; a retrospective study included 42 patients with refractory cutaneous dermatomyositis treated with IVIg and showed that 57% of patients with cutaneous DM responded after one IVIg cycle, and 80% showed improvement after two IVIg cycles, regardless of sex, smoking status, DM subtype, the reason for IVIg initiation, days from DM diagnosis to IVIg initiation, specific cutaneous manifestations, or serological finding. As a result, patients were able to taper off of steroids and discontinue steroid-sparing immunosuppressive agents.11 Finally, a recent meta-analysis by Goswami et al. included 29 studies (a total of 576 patients treated with IVIg). They reported a pooled muscle power improvement with at least a partial response rate of 77.07% with first-line use of IVIg according to the International Myositis Assessment and Clinical Studies (IMACS) measure. The overall mean time to response was 2.9 months, with a significant treatment response on cutaneous disease activity and dysphagia. The steroid and immunomodulating agents sparing effect was reported to be 40.9%.12

In terms of safety, in addition to the well-known adverse effects of IVIg, including nausea, headaches, and fever, venous thromboembolism (VTE) is of particular concern since both IVIg treatment and inflammatory myopathy may increase the risk for VTE.1316 In the ProDERM trial, six patients treated with IVIg experienced eight thromboembolic events (an incidence of 1.54 occurrences per 100 patient-months). This observation led to a protocol change in IVIg infusion rate in order to minimize thromboembolic incidence.9

RITUXIMAB USE IN IIM

Rituximab is a chimeric anti-CD20 monoclonal antibody used to treat various diseases such as rheumatoid arthritis and vasculitis. Juvenile DM (JDM), characterized by substantial increases in type I interferon (IFN) and immature transitional B cells, provides evidence for the involvement of B cells in the etiology of IIM.17 Furthermore, the serum and muscle fibers of IIM have higher levels of B cell activating factor, a member of the tumor necrosis factor (TNF) family.18 These findings support the hypothesis that B-cell depletion may reduce the severity of IIM and have a beneficial effect on its disease burden. A hallmark study regarding rituximab use in IIM, the Rituximab in Myositis (RIM) trial, was a randomized placebo-phase controlled clinical trial of adult patients with refractory IIM. A total of 200 patients were enrolled in the “rituximab early” or “rituximab late” arm of the study 8 weeks later. Even though there were no significant differences between the treatment groups in the time (20 weeks) of achieving the definition of improvement (DOI) based on IMACS, up to 83% of the study patients achieved the DOI at week 44 and were able to taper their glucocorticoid therapy with an excellent general response rate.19 A recent meta-analysis, which included 26 studies and almost 450 patients with IIM, showed that the overall efficacy rate of rituximab was 65%; a complete response rate of 35% was reported in seven trials (121 patients), with improvement in muscle strength, skin involvement burden, and pulmonary function tests/radiographic lung findings in the majority of patients.20

The anti-synthetase syndrome (ASS) is a subcategory of IIM; according to a study of Allenbach et al., only 20% of refractory ASS patients treated with rituximab achieved a primary endpoint of an increase in muscle strength, yet other studies demonstrated a pooled effectiveness of 62% of rituximab in ASS.20,21

Interestingly an observational study demonstrated that patients with IIM treated with rituximab had fewer and milder side effects compared to patients with rheumatoid arthritis and systemic lupus erythematosus.22 The rituximab dosage is crucial because of cost and the raised susceptibility to opportunistic infections and viral infections (e.g. COVID-19). Hence some investigators evaluated the efficacy of low and ultra-low rituximab doses for IIM. One study by Janardana et al. showed that a 0.5 g + 0.5 g rituximab regimen (2 weeks apart) had a similar effect to the 1 g + 1 g regimen.23 Mao et al. used an ultra-low dose of rituximab (100 mg) as an add-on therapy for patients with anti-MDA5-positive interstitial lung disease (ILD), resulting in persistent B-cell depletion that lasted 180 days, and may decrease mortality.24

JANUS KINASE INHIBITOR USE IN IIM

The Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway has a significant role in signaling inflammatory cytokines and immunoregulation. Activation of JAK1/2 induces phosphorylation of STAT1, a key transcription factor that mediates IFN-I signaling. In myositis, upregulation of type I IFN-regulated genes in peripheral blood, muscle, skin, and endothelial cells and elevated serum IFN-a serum correlates with disease activity.25,26 It also seems that IFN-I has a crucial role in the pathogenesis of myopathy, and one study showed that ruxolitinib inhibits the pathogenic effects of IFN-I in both muscle and endothelial cells.27

One of the early indicators for the effectiveness of Janus kinase inhibitors (JAKI) in IIM came from a case report describing a 72-year-old woman with severe muscle and cutaneous DM, diagnosed with a JAK2-V617F mutation-positive myelofibrosis one year after her DM diagnosis. She was treated with ruxolitinib for myelofibrosis, resulting in rapid resolution of her muscle and skin symptoms. Glucocorticoids, mycophenolate mofetil, and IVIg were tapered and eventually discontinued.28 Kim et al. reported four patients with refractory JDM treated with baricitinib 4 mg daily, with significant improvement by week 4 in muscle and skin manifestations, with no serious adverse events reported.29 In addition, in a retrospective study that included 10 patients with either new-onset or refractory JDM, particularly anti-MDA5- or anti-NXP2-positive, treatment with ruxolitinib or baricitinib resulted in clinically inactive disease within 6 months in 50% of patients.30 Evidence for the efficacy of tofacitinib was reported initially in a case series of four patients with refractory DM who responded well to tofacitinib with improvement in their cutaneous, muscle, and joint symptoms.31 Furthermore, in an open-label prospective clinical trial 10 patients with skin-predominant DM and at least moderate skin disease activity by the Cutaneous Dermatomyositis Disease Area and Severity Index (CDASI) were treated with tofacitinib; there was moderate improvement in disease activity in 5 patients according to the IMACS group DOI and a mean reduction in the CDASI score of 18.5.32 Finally, a review of 14 studies including 53 patients with refractory DM showed substantial durable improvement in cutaneous skin signs and muscle strength after using JAKI with the ability to taper down the steroid dose in the majority of patients.33

Although the most reported adverse effect of JAKI was mild-to-moderate herpes zoster infection, VTE is of particular concern, since both IIM and JAKI are associated with increased risk for VTE.2932

ABATACEPT USE IN IIM

The predominance of T cells in the inflammatory infiltrates in DM and polymyositis muscle biopsies is evidence for the role of T cells in its pathogenesis. Furthermore, increased CTLA-4, CD28, CD86, and CD40 expression has been described.34

Abatacept, a fully human fusion protein of CTLA-4 and the Fc portion of human IgG1, is a physiological antagonist of the T cell co-stimulatory molecule CD28.35 Research conducted by Tjärnlund et al. showed that 42% of patients with myositis who were treated early with abatacept had lower disease activity according to the IMACS group DOI, with significant improvement in muscle performance as evidenced by improved manual muscle tests (MMT) 8 score, a validated tool based on assessment of the strength of eight muscle groups.36 Post hoc analysis showed an interesting finding of a positive correlation between the CD4/CD8 ratio in peripheral blood samples at baseline and improved muscle endurance after treatment; however, no significant changes in circulating T and B cell levels were observed.37

Abatacept appears to be a relatively safe biologic. Its most frequently reported adverse event is upper respiratory tract infections, followed by cardiovascular effects, all considered mild or moderate in severity.

TOCILIZUMAB USE IN IIM

Interleukin-6 acts as a mediator of muscle inflammation. Dysregulated IL-6 production has been shown to contribute to the pathogenesis of DM in preclinical studies. Furthermore, tocilizumab, an IL-6 receptor antagonist, had favorable effects on myositis in mouse models.3840 Serum IL-6 levels in adult and juvenile DM have been shown to parallel disease activity, and a small number of patients with refractory polymyositis have responded favorably to treatment with tocilizumab.41,42 Further evidence of tocilizumab tolerability was shown in a prospective phase IIb clinical trial where 36 patients with myositis were randomized 1:1 to receive tocilizumab or a placebo every 4 weeks for 24 weeks. Tocilizumab was well tolerated but not more effective than the placebo.43 Conversely, in a case-control study with 11 patients with refractory immune-mediated necrotizing myopathy, including 3 with anti-3-hydroxy-3-methyl glutaryl-CoA reductase and positive anti-signal recognition particles, 63% achieved clinically significant responses.44 Responders had higher baseline serum IL-6 and muscle IL-6 mRNA levels and higher percentages of CD56-positive muscle fibers than did non-responders. Another study showed the beneficial effect of tocilizumab as a third-line biologic in patients with refractory ASS.45,46 Yet another case-control study using anti-IL-6 suggested that tocilizumab may be used as a salvage therapy for rapidly progressive interstitial lung disease patients refractory to an intensive immunosuppressive regimen.47

MYOSITIS AND ANTI-TUMOR NECROSIS FACTOR

As inhibiting TNF increases type I IFN production, the use of TNF blockers may be a potential trigger for developing or exacerbating inflammatory myopathy.48,49 Although some case reports show a beneficial effect of anti-TNF in myositis, especially in joint or skin predominant disease,50,51 data from retrospective studies reveal an increased risk for ASS and exacerbation of IIM-related ILD.52

LENABASUM AND DERMATOMYOSITIS

Lenabasum, a non-immunosuppressive, non-psychoactive cannabinoid type 2 receptor reverse agonist, is an agent recently investigated in the settings of dermatomyositis.53 Activation of the cannabinoid type 2 receptor has been shown to reduce several vital pro-inflammatory cytokines implicated in DM.53 In a recent double-blind, randomized, placebo-controlled trial, 22 patients were randomized to receive lenabasum or placebo. Lenabasum treatment was associated with more remarkable improvement in the CDASI activity. No serious adverse events were related to lenabasum.54

USE OF BIOLOGIC DMARDS IN MYOSITIS-ASSOCIATED ILD

In addition to the use of bDMARDs in refractory inflammatory myopathy discussed above, interest in the use of these agents in the settings of myositis syndromes with lung involvement is increasing. Although the prevalence, course, histopathology, and severity of myositis-associated ILD vary widely because of many factors, several subtypes, such as amyopathic dermatomyositis, MDA5, and ASS, warrant particular concern.

Intravenous Ig has been reported to be effective in several cases and case series in the settings of progressive ILD associated with myositis. It has also been shown to be effective in cases of rapidly deteriorating MDA5 syndrome.5558

Data are accumulating from case series regarding the use of rituximab in patients with progressive ILD with polymyositis or DM, including ASS.5962 Furthermore, the RECITAL trial showed that rituximab seems to be as effective as cyclophosphamide for myositis-associated ILD, with fewer adverse events.63 Other case reports of patients with MDA5 antibodies described improvement in ILD with rituximab after the failure of other immunosuppressive therapies.64,65

In addition, in a case series of patients with myositis-associated ILD, 26 patients treated with tofacitinib were compared to 35 patients treated with tacrolimus. The 6-month and 1-year mortality rates were significantly lower in the tofacitinib group.66 Furthermore, an open-label study comparing tofacitinib JAKI to a standard regimen of immunomodulating agents among patients with MDA5 antibodies demonstrated a significant (P=0.04) improvement in survival at 6 months in the tofacitinib group (18 patients with 100% survival) compared to 78% survival (25 of 32 patients). Improved diffusing capacity of carbon monoxide and high-resolution computed tomography findings were also seen with tofacitinib.67

In recent years, antifibrotic medications, a group of well-studied agents in treating idiopathic interstitial pulmonary fibrosis,68,69 e.g. nintedanib and pirfenidone, were used to treat autoimmune-associated ILD including myositis-associated ILD. In a retrospective, real-world analysis of IIM-ILD patients, nintedanib, a tyrosine kinase inhibitor, appeared to be protective against the development of rapidly progressive ILD and was associated with improved survival in myositis-associated ILD.70 Similar beneficial effects were shown in a prospective controlled cohort study conducted by Wang et al., in which pirfenidone showed an improvement in pulmonary function tests, imaging findings, and mortality in IIM-ILD patients.71

CONCLUSION

Inflammatory myopathy syndromes are relatively rare disorders. Despite the lack of large cohort randomized controlled trials, in the past several years there has been more solid evidence regarding the use of advanced biologic therapy (e.g. IVIg and rituximab) based on the understanding of the pathogenic mechanisms of these syndromes. Use of JAKI also appears to be promising. While many patients may be managed in the ambulatory setting, acutely ill patients with active myositis or exacerbating lung disease must be managed in hospital and, in some cases, in the medical intensive care unit. To optimize high-quality care, these patients should be managed by a skilled multi-disciplinary team with rheumatologists, chest medicine specialists, and dermatologists.

Nevertheless, despite advances in treating IIM in the past years, much remains unknown and special efforts should be made to offer more precise therapy based on clinical presentation, organ involvement, and biochemical markers.

Abbreviations

bDMARDs biologic disease-modifying antirheumatic drugs
DM dermatomyositis
DOI definition of improvement
DMARDs disease-modifying antirheumatic drugs
IFN interferon
IIMs idiopathic inflammatory myopathies
IMACS International Myositis Assessment and Clinical Studies
IVIg intravenous immunoglobulin
JDM juvenile dermatomyositis
JAK Janus kinase
JAKI Janus kinase inhibitors
STAT signal transducers and activators of transcription
TNF tumor necrosis factor
TSD targeted synthetic disease-modifying antirheumatic drugs
VTE venous thromboembolism.

Footnotes

Conflict of interest: No potential conflict of interest relevant to this article was reported.

REFERENCES
1.
de Souza FHC, Miossi R, Moraes JCB, Bonfá E, Shinjo SK. Favorable rituximab response in patients with refractory idiopathic inflammatory myopathies. Adv Rheumatol. 2018;58:31. https://doi.org/10.1186/s42358-018-0030-z.
2.
Basta M, Dalakas MC. High-dose intravenous immunoglobulin exerts its beneficial effect in patients with dermatomyositis by blocking endomysial deposition of activated complement fragments. J Clin Invest. 1994;94:1729–35. https://doi.org/10.1172/JCI117520.
3.
Kissel JT, Mendell JR, Rammohan KW. Microvascular deposition of complement membrane attack complex in dermatomyositis. N Engl J Med. 1986;314:329–34. https://doi.org/10.1056/NEJM198602063140601.
4.
Raju R, Dalakas MC. Gene expression profile in the muscles of patients with inflammatory myopathies: effect of therapy with IVIg and biologic validation of clinical relevant genes. Brain. 2005;128:1887–96. https://doi.org/10.1093/brain/awh518.
5.
Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science. 2001;291:484–6. https://doi.org/10.1126/science.291.5503.484.
6.
Dalakas MC. Mechanistic effects of IVIG in neuroinflammatory diseases: conclusions based on clinicopathologic correlations. J Clin Immunol. 2014;34(Suppl 1):S120–6. https://doi.org/10.1007/s10875-014-0024-5.
7.
Dalakas MC, Illa I, Dambrosia JM, et al. A controlled trial of high-dose intravenous immune globulin infusions as treatment for dermatomyositis. N Engl J Med. 1993;329:1993–2000. https://doi.org/10.1056/NEJM199312303292704.
8.
Cherin P, Pelletier S, Teixeira A, et al. Results and long-term followup of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients. Arthritis Rheum. 2002;46:467–74. https://doi.org/10.1002/art.10053.
9.
Aggarwal R, Charles-Schoeman C, Schessl J, et al. Trial of intravenous immune globulin in dermatomyositis. N Engl J Med. 2022;387:1264–78. https://doi.org/10.1056/NEJMoa2117912.
10.
Giannini M, Fiorella ML, Tampoia M, et al. Long-term efficacy of adding intravenous immunoglobulins as treatment of refractory dysphagia related to myositis: a retrospective analysis. Rheumatology (Oxford). 2021;60:1234–42. https://doi.org/10.1093/rheumatology/keaa443.
11.
Galimberti F, Kooistra L, Li Y, Chatterjee S, Fernandez AP. Intravenous immunoglobulin is an effective treatment for refractory cutaneous dermatomyositis. Clin Exp Dermatol. 2018;43:906–12. https://doi.org/10.1111/ced.13607.
12.
Goswami RP, Haldar SN, Chatterjee M. Efficacy and safety of intravenous and subcutaneous immunoglobulin therapy in idiopathic inflammatory myopathy: a systematic review and meta-analysis. Autoimmun Rev. 2022;21:102997. https://doi.org/10.1016/j.autrev.2021.102997.
13.
Li Y, Wang P, Li L, Wang F, Liu Y. Increased risk of venous thromboembolism associated with polymyositis and dermatomyositis: a meta-analysis. Ther Clin Risk Manag. 2018;14:157–65. https://doi.org/10.2147/TCRM.S157085.
14.
Carruthers EC, Choi HK, Sayre EC, Aviña-Zubieta JA. Risk of deep venous thrombosis and pulmonary embolism in individuals with polymyositis and dermatomyositis: a general population-based study. Ann Rheum Dis. 2014;75:110–16. https://doi.org/10.1136/annrheumdis-2014-205800.
15.
Paran D, Herishanu Y, Elkayam O, Shopin L, Ben-Ami R. Venous and arterial thrombosis following administration of intravenous immunoglobulins. Blood Coagul Fibrinolysis. 2005;16:313–18. https://doi.org/10.1097/01.mbc.0000172694.85233.a8.
16.
Kapoor M, Hunt I, Spillane J, et al. IVIg-exposure and thromboembolic event risk: findings from the UK Biobank. J Neurol Neurosurg Psychiatry. 2022;93:876–85. https://doi.org/10.1136/jnnp-2022-328881.
17.
Chiu YE, Co DO. Juvenile dermatomyositis: immunopathogenesis, role of myositis-specific autoantibodies, and review of rituximab use. Pediatr Dermatol. 2011;28:357–67. https://doi.org/10.1111/j.1525-1470.2011.01501.x.
18.
Kryštůfková O, Hulejová H, Mann HF, et al. Serum levels of B-cell activating factor of the TNF family (BAFF) correlate with anti-Jo-1 autoantibodies levels and disease activity in patients with anti-Jo-1 positive polymyositis and dermatomyositis. Arthritis Res Ther. 2018;20:158. https://doi.org/10.1186/s13075-018-1650-8.
19.
Oddis CV, Reed AM, Aggarwal R, et al. Rituximab in the treatment of refractory adult and juvenile dermatomyositis and adult polymyositis: a randomized, placebo-phase trial. Arthritis Rheum. 2013;65:314–24. https://doi.org/10.1002/art.37754.
20.
Zhen C, Hou Y, Zhao B, Ma X, Dai T, Yan C. Efficacy and safety of rituximab treatment in patients with idiopathic inflammatory myopathies: a systematic review and meta-analysis. Front Immunol. 2022;13:1051609. https://doi.org/10.3389/fimmu.2022.1051609.
21.
Allenbach Y, Guiguet M, Rigolet A, et al. Efficacy of rituximab in refractory inflammatory myopathies associated with anti-synthetase auto-antibodies: an open-label, phase II trial. PLoS One. 2015;10:e0133702. https://doi.org/10.1371/journal.pone.0133702.
22.
Gilaberte S, Rua J, Isenberg D. Adverse events of treatment with rituximab in patients with myositis. Rheumatology (Oxford). 2023;62:e16–17. https://doi.org/10.1093/rheumatology/keac398.
23.
Janardana R, Amin SN, Rajasekhar L, et al. Low-dose rituximab is efficacious in refractory idiopathic inflammatory myopathies. Rheumatology (Oxford). 2023;62:1243–7. https://doi.org/10.1093/rheumatology/keac438.
24.
Mao M, Xia S, Guo B, et al. Ultra-low dose rituximab as add-on therapy in anti-MDA5-positive patients with polymyositis/dermatomyositis associated ILD. Respir Med. 2020;172:105. https://doi.org/10.1016/j.rmed.2020.105983.
25.
Greenberg SA, Pinkus JL, Pinkus GS, et al. Interferon-α/β-mediated innate immune mechanisms in dermatomyositis. Ann Neurol. 2005;57:664–78. https://doi.org/10.1002/ana.20464.
26.
Melki I, Devilliers H, Gitiaux C. Circulating interferon-α measured with a highly sensitive assay as a biomarker for juvenile inflammatory myositis activity: comment on the article by Mathian et al. Arthritis Rheumatol. 2020;72:195–7. https://doi.org/10.1002/art.41096.
27.
Ladislau L, Suárez-Calvet X, Toquet S, et al. JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain. 2018;141:1609–21. https://doi.org/10.1093/brain/awy105.
28.
Hornung T, Janzen V, Heidgen F-J, Wolf D, Bieber T, Wenzel J. Remission of recalcitrant dermatomyositis treated with ruxolitinib. N Engl J Med. 2014;371:2537–8. https://doi.org/10.1056/NEJMc1412997.
29.
Kim H, Dill S, O’Brien M, et al. Janus kinase (JAK) inhibition with baricitinib in refractory juvenile dermatomyositis. Ann Rheum Diseases. 2020;80:406–8. https://doi.org/10.1136/annrheumdis-2020-218690.
30.
Voyer Le T, Gitiaux C, Authier F-J, et al. JAK inhibitors are effective in a subset of patients with juvenile dermatomyositis: a monocentric retrospective study. Rheumatology (Oxford). 2021;60:5801–8. https://doi.org/10.1093/rheumatology/keab116.
31.
Moghadam-Kia S, Charlton D, Aggarwal R, Oddis CV. Management of refractory cutaneous dermatomyositis: potential role of Janus kinase inhibition with tofacitinib. Rheumatology (Oxford). 2019;58:1011–15. https://doi.org/10.1093/rheumatology/key366.
32.
Paik JJ, Casciola-Rosen L, Shin JY, et al. Study of tofacitinib in refractory dermatomyositis: an open-label pilot study of ten patients. Arthritis Rheumatol. 2021;73:858–65. https://doi.org/10.1002/art.41602.
33.
Paudyal A, Zheng M, Lyu L, et al. JAK-inhibitors for dermatomyositis: a concise literature review. Dermatol Ther. 2021;34:e14939. https://doi.org/10.1111/dth.14939.
34.
Nagaraju K, Raben N, Villalba ML, et al. Costimulatory markers in muscle of patients with idiopathic inflammatory myopathies and in cultured muscle cells. Clin Immunol. 1999;92:161–9. https://doi.org/10.1006/clim.1999.4743.
35.
Genovese MC, Becker J-C, Schiff M, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor α inhibition. N Engl J Med. 2005;353:1114–23. https://doi.org/10.1056/NEJMoa050524.
36.
Tjärnlund A, Tang Q, Wick C, et al. Abatacept in the treatment of adult dermatomyositis and polymyositis: a randomised, phase IIb treatment delayed-start trial. Ann Rheum Dis. 2018;77:55–62. https://doi.org/10.1136/annrheumdis-2017-211751.
37.
Tang Q, Ramsköld D, Krystufkova O, et al. Effect of CTLA4-Ig (abatacept) treatment on T cells and B cells in peripheral blood of patients with polymyositis and dermatomyositis. Scand J Immunol. 2019;89:e12732. https://doi.org/10.1111/sji.12732.
38.
Scuderi F, Mannella F, Marino M, Provenzano C, Bartoccioni E. IL-6-deficient mice show impaired inflammatory response in a model of myosin-induced experimental myositis. J Neuroimmunol. 2006;176:9–15. https://doi.org/10.1016/j.jneuroim.2006.03.026.
39.
Sugihara T, Sekine C, Nakae T, et al. A new murine model to define the critical pathologic and therapeutic mediators of polymyositis. Arthritis Rheum. 2007;56:1304–14. https://doi.org/10.1002/art.22521.
40.
Okiyama N, Sugihara T, Iwakura Y, Yokozeki H, Miyasaka N, Kohsaka H. Therapeutic effects of interleukin-6 blockade in a murine model of polymyositis that does not require interleukin-17A. Arthritis Rheum. 2009;60:2505–12. https://doi.org/10.1002/art.24689.
41.
Bilgic H, Ytterberg SR, Amin S, et al. Interleukin-6 and type I interferon-regulated genes and chemokines mark disease activity in dermatomyositis. Arthritis Rheum. 2009;60:3436–46. https://doi.org/10.1002/art.24936.
42.
Narazaki M, Hagihara K, Shima Y, Ogata A, Kishimoto T, Tanaka T. Therapeutic effect of tocilizumab on two patients with polymyositis. Rheumatology. 2011;50:1344–6. https://doi.org/10.1093/rheumatology/ker152.
43.
Oddis CV, Rockette HE, Zhu L, et al. randomized trial of tocilizumab in the treatment of refractory adult polymyositis and dermatomyositis. ACR Open Rheumatol. 2022;4:983–90. https://doi.org/10.1002/acr2.11493.
44.
Li S, Li W, Jiang W, et al. The efficacy of tocilizumab in the treatment of patients with refractory immune-mediated necrotizing myopathies: an open-label pilot study. Front Pharmacol. 2021;12:635654. https://doi.org/10.3389/fphar.2021.635654.
45.
Murphy SM, Lilleker JB, Chinoy H, Helliwell P. The use of tocilizumab in a case of biologic-refractory antisynthetase syndrome. Rheumatology (Oxford). 2016;55:2277–8. https://doi.org/10.1093/rheumatology/kew296.
46.
Beaumel A, Muis-Pistor O, Tebib J-G, Coury F. Antisynthetase syndrome treated with tocilizumab. Joint Bone Spine. 2016;83:361–2. https://doi.org/10.1016/j.jbspin.2015.03.016.
47.
Zhang X, Zhou S, Wu C, et al. Tocilizumab for refractory rapidly progressive interstitial lung disease related to anti-MDA5-positive dermatomyositis. Rheumatology (Oxford). 2021;60:e227–8. https://doi.org/10.1093/rheumatology/keaa906.
48.
Greenberg SA. Type 1 interferons and myositis. Arthritis Res Ther. 2010;12(Suppl 1):S4. https://doi.org/10.1186/ar2885.
49.
Gaboriau L, Davion J-B, Combret S, et al. Adalimumab and myositis: a case report and review of the French and international Pharmacovigilance Databases. Neuromuscul Disord. 2020;30:915–20. https://doi.org/10.1016/j.nmd.2020.09.026.
50.
Ge Y, Li S, Chen F, et al. The effects of infliximab in treating idiopathic inflammatory myopathies: a review article. Dermatol Ther. 2021;34:e14976. https://doi.org/10.1111/dth.14976.
51.
Campanilho-Marques R, Deakin CT, Simou S, et al. Retrospective analysis of infliximab and adalimumab treatment in a large cohort of juvenile dermatomyositis patients. Arthritis Res Ther. 2020;22:79. https://doi.org/10.1186/s13075-020-02164-5.
52.
Brunasso AMG, Aberer W, Massone C. New onset of dermatomyositis/polymyositis during anti-TNF-α therapies: a systematic literature review. Scientific World Journal. 2014;2014:179180. https://doi.org/10.1155/2014/179180.
53.
Maddukuri S, Patel J, Diaz DA. Cannabinoid type 2 receptor (CB2R) distribution in dermatomyositis skin and peripheral blood mononuclear cells (PBMCs) and in vivo effects of LenabasumTM. Arthritis Res Ther. 2022;24:12. https://doi.org/10.1186/s13075-021-02665-x.
54.
Werth VP, Hejazi E, Pena SM. Safety and efficacy of Lenabasum, a cannabinoid receptor type 2 agonist, in patients with dermatomyositis with refractory skin disease: a randomized clinical trial. J Invest Dermatol. 2022;142:2651–9e1. https://doi.org/10.1016/j.jid.2022.03.029.
55.
Suzuki Y, Hayakawa H, Miwa S, et al. Intravenous immunoglobulin therapy for refractory interstitial lung disease associated with polymyositis/dermatomyositis. Lung. 2009;187:201–6. https://doi.org/10.1007/s00408-009-9146-6.
56.
Bakewell CJ, Raghu G. Polymyositis associated with severe interstitial lung disease. Chest. 2011;139:441–3. https://doi.org/10.1378/chest.10-0360.
57.
Huapaya JA, Hallowell R, Silhan L, et al. Long-term treatment with human immunoglobulin for antisynthetase syndrome-associated interstitial lung disease. Respir Med. 2019;154:6–11. https://doi.org/10.1016/j.rmed.2019.05.012.
58.
Wang L-M, Yang Q-H, Zhang L, et al. Intravenous immunoglobulin for interstitial lung diseases of anti-melanoma differentiation-associated gene 5-positive dermatomyositis. Rheumatology (Oxford). 2022;61:3704–10. https://doi.org/10.1093/rheumatology/keab928.
59.
Andersson H, Sem M, Lund MB, et al. Long-term experience with rituximab in anti-synthetase syndrome-related interstitial lung disease. Rheumatology (Oxford). 2015;54:1420–8. https://doi.org/10.1093/rheumatology/kev004.
60.
Vandenbroucke E, Grutters JC, Altenburg J, Boersma WG, ter Borg EJ, van den Bosch JMM. Rituximab in life threatening antisynthetase syndrome. Rheumatol Int. 2009;29:1499–502. https://doi.org/10.1007/s00296-009-0859-x.
61.
Marie I, Dominique S, Janvresse A, Levesque H, Menard JF. Rituximab therapy for refractory interstitial lung disease related to antisynthetase syndrome. Respir Med. 2012;106:581–7. https://doi.org/10.1016/j.rmed.2012.01.001.
62.
Keir GJ, Maher TM, Ming D, et al. Rituximab in severe, treatment-refractory interstitial lung disease. Respirology. 2013;19:353–9. https://doi.org/10.1111/resp.12214.
63.
Maher TM, Tudor VA, Saunders P, et al. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir Med. 2023;11:45–54. https://doi.org/10.1016/S2213-2600(22)00359-9.
64.
Koichi Y, Aya Y, Megumi U, et al. A case of anti-MDA5-positive rapidly progressive interstitial lung disease in a patient with clinically amyopathic dermatomyositis ameliorated by rituximab, in addition to standard immunosuppressive treatment. Mod Rheumatol. 2015;27:536–40. https://doi.org/10.3109/14397595.2015.1014140.
65.
So H, Wong VTL, Lao VWN, Pang HT, Yip RML. Rituximab for refractory rapidly progressive interstitial lung disease related to anti-MDA5 antibody-positive amyopathic dermatomyositis. Clin Rheumatol. 2018;37:1983–9. https://doi.org/10.1007/s10067-018-4122-2.
66.
Fan L, Lyu W, Liu H. A retrospective analysis of outcome in melanoma differentiation–associated gene 5–related interstitial lung disease treated with tofacitinib or tacrolimus. J Rheumatol. 2022;49:1356–64. https://doi.org/10.3899/jrheum.220367.
67.
Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis–associated interstitial lung disease. N Engl J Med. 2019;381:291–3. https://doi.org/10.1056/NEJMc1900045.
68.
Maher TM, Corte TJ, Fischer A, et al. Pirfenidone in patients with unclassifiable progressive fibrosing interstitial lung disease: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Respir Med. 2020;8:147–57. https://doi.org/10.1016/S2213-2600(19)30341-8.
69.
Kao J-H, Huang H-T, Li KJ. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med. 2020;382:779–81. https://doi.org/10.1056/NEJMc1917224.
70.
Liang J, Cao H, Yang Y. Efficacy and tolerability of nintedanib in idiopathic-inflammatory-myopathy-related interstitial lung disease: a pilot study. Front Med (Lausanne). 2021;8:626953. https://doi.org/10.3389/fmed.2021.626953.
71.
Wang J, Wang X, Qi X. The efficacy and safety of pirfenidone combined with immunosuppressant therapy in connective tissue disease-associated interstitial lung disease: a 24-week prospective controlled cohort study. Front Med (Lausanne). 2022;1861;9:87. https://doi.org/10.3389/fmed.2022.871861.