Abstract
Purpose of review:
Dermatomyositis (DM) is a systemic autoimmune disease affecting multiple organs, including skeletal muscle, skin, and lungs. Although DM disease mechanisms are incompletely understood, accumulating evidence suggests that interferons may play a significant role. Consequently, it is of considerable interest that drugs blocking the activity of interferons by inhibiting the Janus Kinase/Signal Transducer and Activator of Transcription (JAK-STAT) pathway have been approved for use in other autoimmune diseases. This manuscript will examine the IFN pathways and their importance in DM, review the existing literature on the use of JAK-STATs inhibitors in patients with adult or juvenile DM, and discuss the potential utility of JAK-STAT inhibitors to treat this disease.
Recent findings:
Recent reports suggest that muscle and skin involvement in patients with either adult or juvenile DM respond favorably to JAK-STAT inhibitors. Moreover, preliminary data indicates that JAK-STAT inhibitors may be useful to treat clinical manifestations of this disease that are complicated to manage otherwise, such as calcinosis or rapidly progressive interstitial lung disease in DM patients with anti-MDA5 autoantibodies.
Summary:
An increasing number of reports suggest that JAK-STAT inhibitors may be useful to treat the varied manifestations of adult and juvenile DM. However, as most studies were either small or lacked appropriate comparators, further research will be necessary to define the role of these drugs in DM treatment.
Keywords: Autoimmune diseases, muscle disease, myositis, interferon, JAK-STAT
Introduction:
Inflammatory myopathies
The inflammatory myopathies are a heterogeneous group of diseases affecting multiple organs and systems, including the skeletal muscle the skin, the lungs, and/or the joints. Myositis can be subclassified in dermatomyositis (DM), the antisynthetase syndrome (AS), immune-mediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM). [1] Around 70% of the patients with myositis have myositis-specific autoantibodies (MSAs) targeting nuclear or cytoplasmic proteins. These MSAs include anti-Mi2, anti-NXP2, anti-TIF1gamma, anti-MDA5, anti-tRNA synthetases, anti-HMGCR, and anti-SRP.[2]
DM can affect both adults and children (juvenile DM [JDM]). Patients with DM have characteristic skin rashes [3] including purple discoloration on the eyelids (heliotrope rash) and purple discoloration (Gottron’s rash) or purple plaques (Gottron’s sign) on the extensor surfaces of the joints. The presence of perifascicular atrophy is a highly specific finding in the muscle biopsies patients with dermatomyositis and is usually accompanied by other features, such as the presence of endomysial or perivascular inflammation, MHC-I overexpression, or complement deposition in the capillaries and muscle fibers.[4] A subset of patients with DM and autoantibodies recognizing MDA5 frequently suffers from a highly lethal form of rapidly progressive interstitial lung disease.[5]
Most clinical manifestations of DM have been associated with overactivation of the interferon pathway. In the following sections, we will review the interferons and what is known about the interferon pathway in DM.
The interferons
In the 1930s, research conducted by Hoskins demonstrated that rabbits previously infected by the herpes simplex virus were protected against subsequent infections by the same type of virus.[6] Soon later, it was discovered that virus-infected animals were also protected against other types of viruses. But it was not until 1957 when it was observed that cells previously infected by a virus could produce a substance, the “interferon”, that made other cells immune to different viruses. It was later discovered that viral interference was caused by a family of proteins, the interferons, and not by a single molecule.[7, 8]
The interferons represent a widely expressed group of cytokines, including three main classes. Type I is encoded in chromosome 9[9] and includes alpha and beta IFN among others. Type II, or interferon gamma, is also encoded in chromosome 9[7]. Finally, type III IFN or IFNλ,[10, 11] is encoded in chromosome 12.[7]
The various IFNs are encoded by different genes, produced by distinct cells, and their release is triggered by different molecules. Interferon alpha is produced by macrophages, lymphocytes, tumor cells, cells infected by a virus, or affected by mitogens. Interferon beta is produced in the epithelial cells and fibroblasts and its production is triggered by viruses. Finally, interferon gamma is produced in T cells and natural killer cells after stimulation by external mitogens. Interferons have a wide variety of roles including antiviral, antiproliferative, and immunomodulatory functions.[12] These functions are mediated by the binding of these cytokines to receptors in the surface of the cell which activate the Janus Kinase/Signal Transducer and Activator of Transcription (JAK-STAT) signaling pathway, leading to the expression of the so-called interferon-inducible genes.
The JAK-STAT pathway
The role of cytokines in the regulation of the immune system by binding to surface receptors and their different modes of signaling through the JAK-STAT pathway has been long understood. Examples of cytokines using the JAK-STAT pathway include erythropoietin, growth hormone, interleukin (IL) 2, IL 6, IL 7, and interferon (IFN), among others.[13, 14]
The JAK family is composed of four proteins: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYR).[15] The STAT protein family in mammalians is composed of STAT 1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6.[14] Most of the previously described cytokines bind to multiple receptors, whereas JAK3 binds only to one receptor subunit, the common gamma chain or IL2-Rγ chain (γc).[13]
The interferon signaling pathway and its importance in myositis
The best known IFN signaling pathway is the canonical type I IFN signaling pathway. The binding of the IFN I (alpha or beta) or IFN III to its receptors (IFNAR and IFNLR1 or IL10-R2 respectively) on the cell surface activates Janus Kinase 1 (JAK1) and Tyrosine Kinase 2 (TYK2). This leads to the phosphorylation of cytoplasmic transcription factors STAT1 and STAT2 [11, 16] and the creation of a heterodimer that translocates to the nucleus. Once inside the nucleus, the dimer assembles with IFN-regulatory factor 9 (IRF9) creating the IFN-stimulated gene factor 3 (ISGF3). ISGF3 binds to its cognate DNA sequences known as IFN-stimulated response elements (ISREs), leading to the activation of the ISGs.[17] Unlike IFN I and IFN III, when IFN II binds its receptor IFNGR (IFNGR1 and IFNGR2) JAK1 and JAK2 (instead JAK1 and TYK2) are activated, leading to the creation of the homodimer STAT1. This homodimer binds to its cognate IFN gamma activated sequence (GAS) inside the nucleus (instead of the ISREs), leading to the activation of the interferon-stimulated genes (ISGs).[18] (Figure 1)
Figure 1.
Mechanism of action of JAK-STAT inhibitors used in myositis
The pathogenesis of DM is not completely understood. However, the upregulation of interferon-alpha/beta inducible genes and proteins has been widely reported both in DM[19–23] and JDM.[24–26] This overexpression occurs in the muscle,[19, 23, 25] skin,[27] and blood of these patients[21, 28] More recently, our group and others have shown that also IFN 2 inducible genes are also overexpressed in DM.[23, 29] Moreover, the expression of IFN inducible genes correlates with disease activity in DM,[22, 23, 28] and JDM.[25]
JAK-STAT inhibitors: history, types, and uses
Autoimmune diseases have been classically treated with non-specific immunosuppressive therapies like corticosteroids and corticosteroid-sparing agents. More recently, these diseases have been treated with pathway-specific immunosuppressant treatments like monoclonal antibodies and JAK-STAT inhibitors.[30] (Table 1). The development of JAK inhibitors has its origin in the 1993 work of Riedy et al. which reported the sequence and chromosomal localization of JAK3.[31]
Table 1.
JAK-STAT inhibitors used in myositis.
REFERENCES | DRUG | DOSE | TYPE OF DERMATOMYOSITIS | PATIENTS | CONCOMITANT TREATMENT |
---|---|---|---|---|---|
Adult dermatomyositis | |||||
Hornung, 2014[48] | Ruxolitinib | 5mg/d up to 15 mg/bid | Treatment of myelofibrosis in a patient with refractory DM | 1 | Prednisone, mycophenolate, IVIg |
Kurtzman DJ, 2016[49] | Tofacitinib | 10 mg/bid (1 patient), 5 mg/bid (2 patients) | 3 patient with refractory DM | 3 | HCQ (1 patient) |
Paik JJ, 2016[51] | Tofacitinib | 5 mg/bid | Refractory DM | 1 | Prednisolone |
Paik JJ, 2018[53] | Tofacitinib | 11 mg/d | Refractory DM (6 patients anti-TIF1g) | 10 | Washed out of any steroid sparing agent and not allowed more than 20 mg of prednisone/d prior to study entry |
Ladislau L, 2018[54] | Ruxolitinib | Up to 40 mg/d | 3 refractory anti-TIF1g DM and 1 refractory anti-SAE DM | 4 | Prednisone +/− IVIG |
Kurasawa K, 2018[59] | Tofacitinib | 10 mg/d | Refractory anti-MDA5 JDM with ILD | 5 | Glucocorticoid pulse therapy, prednisolone, CYC, CP |
Kato M, 2019[60] | Tofacitinib | 5 mg/bid | Refractory MDA5 refractory JDM with ILD | 1 | Methilprednisolone, CP |
Chen Z, 2019[61] | Tofacitinib | 5 mg/bid | 18 early-stage anti-MDA5 DM with ILD and 32 historical comparators with anti-MDA5 DM with ILD | 18 | Prednisone |
Wendel S, 2019[62] | Tofacitinib | 5 mg/bid | 1 anti-Mi2 DM with calcinosis and 1 anti MDA5 DM with ILD | 2 | MTX and prednisone(1 patient)/ Monotherapy with Tofacitinib (1 patient) |
Ishikawa Y, 2020[63] | Tofacitinib | 10 mg/d | Refractory anti-MDA5 DM with ILD | 1 | Prednisolone |
Juvenile dermatomyositis | |||||
Aeschlimann FA, 2018[64] | Ruxolitinib | 10 mg/bid | Refractory anti-NXP2 JDM | 1 | Rituximab, MYC |
Papadopoulou C, 2019[65] | Baricitinib | 6 mg/bid | Refractory anti-TIF1g JDM with calcinosis | 1 | Prednisolone |
Sabbagh S, 2019[66] | Tofacitinib | 5 mg/bid up to 10 mg/bid (1 patient)/ 5 mg/bid (1 patient) | Refractory anti-MDA5 JDM with and calcinosis | 2 | Pulse methylprednisolone, prednisone, MMF (1)/ CYC, rituximab, sildenafil (1) |
Kim H, 2020[67] | Baricitinib | 4–8 mg/d divided in 2 times per day | 2 Anti-TIF1g and 2 anti-NXP2 refractory JDM | 4 | Biologics other than IVIg were washed out and other medications continued: MYC (4), MTX (1), CYC (1), TAC (1) |
Yu Z, 2020[68] | Tofacitinib | 5 mg/bid | Refractory JDM | 3 | Prednisone + HCQ (2 patients) / Prednisone (1 patient) |
DM: dermatomyositis, JDM: juvenile DM, ILD: interstitial lung disease
IVIG: intravenous immunoglobulin, MYC: mycophenolate, HCQ: hydroxychloroquine, MMF: mycophenolate, CP: cyclophosphamide, CYC: ciclosporin, MTX: methotrexate; d: day, bid: two times a day
Currently, there are five JAK-STAT inhibitors approved by the FDA: tofacitinib, ruxolitinib, baricitinib, upadacitinib, and fedratinib. These have been approved to treat myeloproliferative neoplasms and autoimmune diseases such as ulcerative colitis, rheumatoid arthritis(RA), and psoriatic arthritis.[30]
Tofacitinib was the first JAK-STAT inhibitor to be developed. This drug interferes with the IFN pathway by inhibiting JAK 1 and JAK3 signaling pathways and was shown to impair Th1 and Th2 differentiation as well as the production of Th17 cells.[32] Tofacitinib was approved by the FDA in 2012 for the treatment of RA patients with poor response to methotrexate[33] and ulcerative colitis.[34, 35] Ruxolitinib, which blocks the IFN pathway inhibiting the JAK1 and JAK2 signaling, was the first JAK inhibitor treatment approved by the FDA for myeloproliferative diseases.[36–38] This drug has also been approved to treat acute graft-versus-host disease[39] and is also effective to treat RA.[40, 41] Baricitinib inhibits the JAK1/JAK2 and, which has demonstrated efficacy in RA [42], has been approved by the FDA for the treatment of this disease since 2018.[30] Upadacitinib inhibits the JAK1 and was recently approved to be used for the treatment of RA.[30] Finally, fedratinib, a JAK2 inhibitor, has been approved to treat myelofibrosis.[30]
Several other JAK-STAT inhibitors are being tested for the treatment of autoimmune diseases, myeloproliferative diseases, or other neoplasms.[36] For example, Filgotinib, a JAK1 inhibitor[43, 44] has been studied as a treatment for RA in combination with methotrexate in patients who don’t respond to monotherapy. This revealed that filgotinib has a rapid onset of action and improves the symptoms of active RA within 24 weeks.[45–47]
Application of JAK-STAT inhibitors in dermatomyositis
As discussed in the previous section, the JAK-STAT inhibitors are only FDA-approved for a limited number of autoimmune diseases. Nevertheless, these drugs have been used in patients with DM that were not responsive to the usual therapy. In the following sections, we will discuss the use of JAK-STAT inhibitors in DM and JDM.
Adult dermatomyositis:
The first case of a DM patient treated with a drug that inhibits the JAK-STAT cascade was published in 2014. A patient with long-standing and poorly controlled DM developed myelofibrosis and, for this latter reason, was treated with Ruxolitinib. Surprisingly, not only the myelofibrosis, but also the signs and symptoms of DM, improved after treatment with this drug.[48] As previously mentioned, IFN activates JAK-STATs leading to the expression of genes that may have a role in the pathogenesis of DM. Thus, with the premise that JAK inhibitors, by blocking the activation of JAK-STAT may cause a downregulation in the inflammatory cascade, other researchers reported treating patients with DM with this type of medications. First, in 2016, Kurtzman et al reported a series of 3 with refractory cutaneous DM that were treated with the tofacitinib for an average of 9.3 months.[49] An improvement in skin disease was documented using the validated Cutaneous Dermatomyositis Disease Area and Severity Index (CDASI) activity score.[50] The authors also reported the adverse effects, the need for other therapies, and the tolerability of the drug. The patients had been previously refractory to prednisone, methotrexate, hydroxychloroquine, mycophenolate, or intravenous immunoglobulins (IVIGs), among others. An average improvement of 12 points in the CDASI was noted and muscle weakness was subjectively improved in two of the three patients. No adverse event was reported, and two of the three patients continued using tofacitinib as monotherapy. Also, in 2016, Paik et al. reported a refractory DM patient with persistent involvement of the muscle, skin, and joint even after treatment with methotrexate, mycophenolate, azathioprine, and intravenous immunoglobulin (each administered for at least for 6 months (as well as two cycles of rituximab). After receiving tofacitinib, the patient had marked improvements in muscle strength, an improved rash, and less severe arthritis.[51]
Following their case report, Paik and colleagues initiated an open-label clinical trial that included 10 DM patients, including 6 with anti-TIF1 gamma autoantibodies (ClinicalTrials.gov NCT03002649). The preliminary results of this study have been reported as a communication to the American College of Rheumatology meeting in 2018. Their first objective was to assess the proportion of patients achieving the definition of improvement defined by the International Myositis Assessment and Clinical Studies (IMACS)[52] at 12 weeks of treatment. Other objectives of this clinical trial were to assess the mean change in the CDASI, the safety and tolerability of tofacitinib, and the steroid-sparing effect of the drug. Patients were washed out of any other steroid-sparing agent and treated for 12 weeks with 11mg/day of tofacitinib. They were allowed to maintain up to 20 mg of prednisone a day when entering the study. All of the participants showed an improvement of the disease (half with “minimal” and half with “moderate” improvement), decreased CDASI scores, and a trend towards a decrease in levels of the IFN-induced chemokines CXCL-9/10.[53]
An interesting study including not only the treatment of refractory DM patients with JAK inhibitors but also assessing the in vitro effects of manipulating the IFN 1 pathway in muscle cells and endothelial cells was performed by Ladislau et al. They aimed to determine the pathogenicity of type 1 IFN and to assess the role of type 1 IFN pathway blockade as a treatment for DM.[54] For this purpose, they used human primary skeletal muscle cells and human clonal microvascular endothelial cells (HMEC-I cells) treated with IFN stimulators and IFN suppressors. In the muscle cells, it was shown that the activation of the IFN 1 pathway impaired the formation of myotubes, diminished the expression of myogenin, and reduced the area of the myotubes. Moreover, it upregulated some genes previously linked to atrophy. On the contrary, when the myoblast and myotubes were preincubated with IFN 1 pathway suppressors before being incubated with the IFN 1 activators, the abovementioned pathogenic effects were not observed. Type I IFN was also shown to impair the formation of capillaries, having a deleterious effect on the segment length, the number of meshes and the number of endothelial cell junctions compared with those that were not treated with IFN 1 activators. Moreover, pretreatment of both the muscle and the endothelial cells with ruxolitinib prevented damage to the muscle and endothelial cells induced by the activation of the IFN 1 pathway. Next, these investigators aimed to confirm their findings in vivo by analyzing six frozen muscle biopsies of patients with DM. For the immunohistochemical analysis, they used anti-FBXO32, anti-MXA, anti-VEGF, and anti-ISG15. They showed that the atrophic muscle fibers located in the perifascicular area that were positive for MxA expressed also FBXO32/atrogin and that there was a co-localization of VEGF and MX1/MxA in the capillaries.
Based on the abovementioned results, the authors initiated a three-month treatment trial of ruxolitinib in four refractory DM patients while maintaining concommitant treatment with low-dose prednisone with or without IVIG. The refractoriness of the disease was based on continued disease activity after treatment with two different immunosuppressive drugs in combination with IVIG or corticosteroids. Apart from the typical DM skin rash, two patients had skeletal muscle weakness, and one of them had high creatine kinase (CK) levels. The authors reported an improvement in the skin features as well as in the strength and CK levels after the treatment. Moreover, the IFN levels in the serum and the ISG score in the PBMCs decreased. Despite the limited sample size, both the in vitro and in-vivo findings of this study are very promising.
As mentioned earlier, DM patients often have MSAs that define the phenotype of the disease. Among these autoantibodies, those directed against the melanoma differentiation-associated protein 5 (MDA5) are associated with severe DM-skin involvement, minimal or no clinical muscle involvement[5, 55], and a rapidly progressive interstitial lung disease.[56–58] Given the high mortality of anti-MDA5 patients with rapidly progressive interstitial lung disease (RP-ILD), finding effective treatments to prevent or revert the lung involvement is an important area of research.
There are published several cases and case series as well as an open-label clinical trial of anti-MDA5-positive DM patients with refractory ILD treated with JAK-STAT inhibitors. Kurasawa et al. published a series of 5 such patients who received tofacitinib in combination with other immunosuppressant drugs. They showed that tofacitinib combined with the other therapies controlled refractory ILD in anti-MDA5-positive DM patients. However, these patients did experience an increased risk of infection.[59] Similarly, Kato et al. reported a patient with anti-MDA5-positive DM and refractory ILD that improved after being treated with tofacitinib along with other immunosuppressant drugs.[60] Importantly, in 2019, 18 patients with anti-MDA5-positive amyopathic DM were treated with tofacitinib and glucocorticoids and compared with a historical cohort of 32 patients. Six months after ILD onset all the patients treated with tofacitinib were alive compared with 78% in the historical control comparator group. Moreover, the ferritin levels, FVC, DLCO, and high-resolution CT findings in the patients treated with tofacitinib improved over time. These patients experienced minimal side effects.[61] Finally, Wendel[62] and Ishikawa[63] also reported a good response to tofacitinib in a small number of anti-MDA5-positive DM patients with ILD.
Juvenile dermatomyositis
There is also accumulating evidence that JAK-STAT inhibitors may be useful in JDM. In 2018, Aeschlimann et al. described a 13 years-old female with severe refractory anti-NXP2-positive JDM complicated by vasculopathy.[64] She did not improve with high-dose corticosteroids and methotrexate and had to be admitted to the ICU with dysphagia, tetraparesis, dysphonia, abdominal pain, bloody diarrhea, and worsening of her cutaneous features. She seemed to improve under an intensive treatment regimen consisting of prednisone (1.6 mg/kg/day), IVIG ((2 g/kg for three consecutive days), plasma exchange, and rituximab (three weekly doses at 375 mg/m2). These treatments were tapered but soon after she started getting weak and developing skin lesions. Despite starting mycophenolate mofetil, increasing the dose of prednisone, and administering additional cycles of IVIG, plasma exchange, and rituximab, she continued having flares of the disease. The authors found high levels of IFN protein in serum and that serum ISGs were overexpressed. They also reported that the phosphorylation of STAT1 and STAT3 in T lymphocytes and monocytes was higher than in the controls. Thus, ruxolitinib was started, and soon after the clinical manifestations of the patient improved. Moreover, compared with the controls, STAT1 phosphorylation decreased in CD3 + lymphocytes as well as in monocytes; 2 months after the ruxolitinib was started the STAT 1 phosphorylation was only present in monocytes.
Similar cases of refractory JDM patients treated with JAK-STAT inhibitors have been reported in the literature. Papadopoulou et al. described the case of an 11-year-old Caucasian male with anti-TIF 1 gamma-positive JDM and anti-Ro52 autoantibodies since 2.5 years-old.[65] Later in life, his disease worsened and he began to experience dysphagia, elevated CK and LDH levels, as well as subcutaneous calcinosis. Over subsequent years, he was treated with corticosteroids, methotrexate, cyclophosphamide, and IVIg. At 11.5 years-old, after the patient was started on baricitinib due to the progression of his skin lesions, the skin involvement, muscle weakness, and CK levels improved. The patient experienced a relapse after stopping the JAK-STAT inhibitor voluntarily. Strikingly, only15 days after baricitinib was reintroduced, the patient experienced improvement. Furthermore, he was able to taper the corticosteroids after 18 months of treatment. As in the case described by Aeschilmann’s, following treatment with baricitinib, STAT1 phosphorylation in lymphocytes decreased to levels similar to that seen in controls.
In 2019, Sabbagh et al. described cases of a 12-year-old male and a 15-year-old female with refractory JDM and anti-MDA5 autoantibodies with calcinosis who were treated with tofacitinib. Both patients achieved clinical improvement in their muscle weakness, skin involvement, and ILD.[66] One of the patients also experienced an improvement in calcinosis. Moreover, a 28-gene IFN score improved in these patients following with the JAK-STAT inhibitor.
Recently, four patients with refractory JDM were treated with baricinib in a compassionate use study at the NIH (ClinicalTrials.gov NCT03002649). Other immunosuppressant medications were continued during the treatment with the JAK-STAT inhibitor. Biologics were washed out before starting the baricitinib and patients received 4–8mg of the drug divided two times per day. The activity of the disease as well as the CDASI showed an improvement. Moreover, two of the patients with baseline weakness experienced an improvement in their strength.[67]
Most recently, Zhongxun Yu et al. reported that treating three cases of JDM with tofacitinib resulted in improvement of the muscle strength, resolution of cutaneous lesions, increased daily quality of life, and successful tapering of the corticosteroids.[68]
Future of JAK-STAT inhibitors in myositis
As described here, preliminary experiences with JAK-STAT inhibitors in DM have been encouraging. However, these studies were either too small to draw definite conclusions or lacked the appropriate controls to verify the efficacy of the drug. Thus, well-designed, placebo-controlled, randomized clinical trials will be key to demonstrate the efficacy of JAK-STAT inhibitors to treat key features of the disease.
One of the most relevant clinical problems in patients with DM is the presence of muscle weakness. Considerable evidence now suggests that over-activation of the interferon pathway plays an important role in the development of muscle dysfunction. Thus, it was suspected that inhibiting this pathway would have a therapeutic effect. However, given the high efficacy of conventional immunosuppressant treatments in most patients with DM, the challenge will be to demonstrate the efficacy of JAK-STAT inhibitors compared to standard therapies.
Most of these preliminary studies have focused on demonstrating the improvement of skin involvement with JAK-STAT inhibitors. Given that skin involvement is often more refractory to treatment than muscle weakness, JAK-STAT inhibitors may have a therapeutic role here. The main difficulty in demonstrating the efficacy of these drugs in a randomized clinical trial will be to use tools to evaluate the skin involvement that is quantitative and sensitive enough. Current evaluation systems, like the CDASI, may be insensitive to small differences between study arms using relatively small sample sizes.
Finally, some manifestations of DM and JDM, such as calcinosis and/or ILD are often difficult to treat with current immunosuppressant treatments. For these manifestations of DM, the JAK-STAT inhibitors may be useful therapeutic modalities in selected patients. Although the pathogenesis of calcinosis is still not well understood, IL 6 has been found in the calcium-laden fluid collections of patients with JDM[69] and it is known that this cytokine signals through the JAK-STAT pathway. Thus, it would be reasonable to hypothesize, that, as the abovementioned preliminary evidence suggests,[65, 66] JAK-STAT inhibitors could be effective, if not to improve, at least to stabilize the spread of calcinosis. Also, in certain clinical groups with a high risk of developing recalcitrant calcinosis, such as those patients with anti-NXP2-positive DM,[70] it would be interesting to know if early treatment with JAK-STAT inhibitors could prevent the initiation of calcinosis.
Alternatively, in patients with ILD, particularly those with anti-MDA5 autoantibodies, the JAK-STAT inhibitors may be helpful not only to treat but to prevent the development of this highly lethal manifestation of the disease. Given the recently published preliminary evidence of their efficacy,[61] it may be ethically questionable to randomize patients to receive placebo in anti-MDA5-positive DM patients with rapidly progressive ILD. Alternatively, larger series of patients treated with JAK-STAT inhibitors may be helpful to verify the efficacy of these drugs in preventing the progression of the ILD anti-MDA5 DM.
In conclusion, over the past two decades, the pathogenic relevance of the interferon pathway has been established in DM. Now, preliminary evidence discussed in this review suggests that JAK-STAT inhibitors may be helpful to treat patients with DM. Notwithstanding these encouraging findings, more studies with a higher level of evidence will be necessary to define the therapeutic role of the JAK-STAT inhibitors in DM.
FUNDING:
This research was supported by the Intramural Research Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health (AR-041203).
Footnotes
Conflict of Interest
Maria Casal-Dominguez, IngoPinal-Fernandez, and Andrew L. Mammen declare that they have no conflict of interest
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
- 1.Selva-O’Callaghan A, Pinal-Fernandez I, Trallero-Araguas E, Milisenda JC, Grau-Junyent JM, Mammen AL. Classification and management of adult inflammatory myopathies. Lancet Neurol. 2018;17(9):816–28. doi: 10.1016/S1474-4422(18)30254-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ghirardello A, Borella E, Beggio M, Franceschini F, Fredi M, Doria A. Myositis autoantibodies and clinical phenotypes. Auto Immun Highlights. 2014;5(3):69–75. doi: 10.1007/s13317-014-0060-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zong M, Lundberg IE. Pathogenesis, classification and treatment of inflammatory myopathies. Nat Rev Rheumatol. 2011;7(5):297–306. doi: 10.1038/nrrheum.2011.39. [DOI] [PubMed] [Google Scholar]
- 4.Lahoria R, Selcen D, Engel AG. Microvascular alterations and the role of complement in dermatomyositis. Brain. 2016;139(Pt 7):1891–903. doi: 10.1093/brain/aww122. [DOI] [PubMed] [Google Scholar]
- 5.Sato S, Hoshino K, Satoh T, Fujita T, Kawakami Y, Fujita T, et al. RNA helicase encoded by melanoma differentiation-associated gene 5 is a major autoantigen in patients with clinically amyopathic dermatomyositis: Association with rapidly progressive interstitial lung disease. Arthritis Rheum. 2009;60(7):2193–200. doi: 10.1002/art.24621. [DOI] [PubMed] [Google Scholar]
- 6.Hoskins M: A protective action of neutropic against viscerotropic yellow fever virus in Macacus rhesus. Am J Trop Med Hyg. [Google Scholar]
- 7.De Andrea M, Ravera R, Gioia D, Gariglio M, Landolfo S. The interferon system: an overview. Eur J Paediatr Neurol. 2002;6 Suppl A:A41–6; discussion A55–8. doi: 10.1053/ejpn.2002.0573. [DOI] [PubMed] [Google Scholar]
- 8.Vilcek J Fifty years of interferon research: aiming at a moving target. Immunity. 2006;25(3):343–8. doi: 10.1016/j.immuni.2006.08.008. [DOI] [PubMed] [Google Scholar]
- 9.Kalliolias GD, Ivashkiv LB. Overview of the biology of type I interferons. Arthritis Res Ther. 2010;12 Suppl 1:S1. doi: 10.1186/ar2881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li M, Liu X, Zhou Y, Su SB. Interferon-lambdas: the modulators of antivirus, antitumor, and immune responses. J Leukoc Biol. 2009;86(1):23–32. doi: 10.1189/jlb.1208761. [DOI] [PubMed] [Google Scholar]
- 11.Gr Stark, Kerr M, Williams BR, Silverman RH, RD S. How cells respond to interferons. Annu Rev Biochem. 1998;67:227–64. [DOI] [PubMed] [Google Scholar]
- 12.Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. 2005;5(5):375–86. doi: 10.1038/nri1604. [DOI] [PubMed] [Google Scholar]
- 13.O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med. 2013;368(2):161–70. doi: 10.1056/NEJMra1202117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
- 15.Ghoreschi K, Laurence A, O’Shea JJ. Janus kinases in immune cell signaling. Immunol Rev. 2009;228(1):273–87. doi: 10.1111/j.1600-065X.2008.00754.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Levy DE, Darnell JE Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3(9):651–62. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
- 17.Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14(1):36–49. doi: 10.1038/nri3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nallar SC, Kalvakolanu DV. Interferons, signal transduction pathways, and the central nervous system. J Interferon Cytokine Res. 2014;34(8):559–76. doi: 10.1089/jir.2014.0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70(11):2029–36. doi: 10.1136/ard.2011.150326. [DOI] [PubMed] [Google Scholar]
- 20.Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, Tawil R, et al. Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol. 2005;57(5):664–78. doi: 10.1002/ana.20464. [DOI] [PubMed] [Google Scholar]
- 21.Baechler EC, Bauer JW, Slattery CA, Ortmann WA, Espe KJ, Novitzke J, et al. An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity. Mol Med. 2007;13(1–2):59–68. doi: 10.2119/2006-00085.Baechler. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Walsh RJ, Kong SW, Yao Y, Jallal B, Kiener PA, Pinkus JL, et al. Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum. 2007;56(11):3784–92. doi: 10.1002/art.22928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pinal-Fernandez I, Casal-Dominguez M, Derfoul A, Pak K, Plotz P, Miller FW, et al. Identification of distinctive interferon gene signatures in different types of myositis. Neurology. 2019;93(12):e1193–e204. doi: 10.1212/WNL.0000000000008128. [DOI] [PMC free article] [PubMed] [Google Scholar]; * Study describing the activation of the type I and type II interferon pathways in different types in myositis.
- 24.Niewold TB, Kariuki SN, Morgan GA, Shrestha S, Pachman LM. Elevated serum interferon-alpha activity in juvenile dermatomyositis: associations with disease activity at diagnosis and after thirty-six months of therapy. Arthritis Rheum. 2009;60(6):1815–24. doi: 10.1002/art.24555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.O’Connor KA, Abbott KA, Sabin B, Kuroda M, Pachman LM. MxA gene expression in juvenile dermatomyositis peripheral blood mononuclear cells: association with muscle involvement. Clin Immunol. 2006;120(3):319–25. doi: 10.1016/j.clim.2006.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tezak Z, Hoffman EP, Lutz JL, Fedczyna TO, Stephan D, Bremer EG, et al. Gene expression profiling in DQA1*0501+ children with untreated dermatomyositis: a novel model of pathogenesis. J Immunol. 2002;168(8):4154–63. doi: 10.4049/jimmunol.168.8.4154. [DOI] [PubMed] [Google Scholar]
- 27.Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, et al. Interferon and biologic signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PLoS One. 2012;7(1):e29161. doi: 10.1371/journal.pone.0029161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Greenberg SA, Higgs BW, Morehouse C, Walsh RJ, Kong SW, Brohawn P, et al. Relationship between disease activity and type 1 interferon- and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun. 2012;13(3):207–13. doi: 10.1038/gene.2011.61. [DOI] [PubMed] [Google Scholar]
- 29.Rigolet M, Hou C, Baba Amer Y, Aouizerate J, Periou B, Gherardi RK, et al. Distinct interferon signatures stratify inflammatory and dysimmune myopathies. RMD Open. 2019;5(1):e000811. doi: 10.1136/rmdopen-2018-000811. [DOI] [PMC free article] [PubMed] [Google Scholar]; * Study describing the activation of the type I and type II interferon pathways in different types in myositis.
- 30.Damsky WPD, Ramseier J, Al-Bawardy B, Chun H, Proctor D SV, Flavell RA, King B, The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseases, Journal of Allergy and Clinical Immunology (2020), doi: 10.1016/j.jaci.2020.10.022. [DOI] [PubMed] [Google Scholar]
- 31.Riedy MC, Dutra AS, Blake TB, Modi W, Lal BK, Davis J, et al. Genomic sequence, organization, and chromosomal localization of human JAK3. Genomics. 1996;37(1):57–61. doi: 10.1006/geno.1996.0520. [DOI] [PubMed] [Google Scholar]
- 32.Ghoreschi K, Jesson MI, Li X, Lee JL, Ghosh S, Alsup JW, et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol. 2011;186(7):4234–43. doi: 10.4049/jimmunol.1003668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Berekmeri A, Mahmood F, Wittmann M, Helliwell P. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(9):719–30. doi: 10.1080/1744666X.2018.1512404. [DOI] [PubMed] [Google Scholar]
- 34.Sandborn WJ, Ghosh S, Panes J, Vranic I, Wang W, Niezychowski W, et al. A phase 2 study of tofacitinib, an oral Janus kinase inhibitor, in patients with Crohn’s disease. Clin Gastroenterol Hepatol. 2014;12(9):1485–93 e2. doi: 10.1016/j.cgh.2014.01.029. [DOI] [PubMed] [Google Scholar]
- 35.Xeljanz [package insert]. Pfizer Inc. [Google Scholar]
- 36.O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–28. doi: 10.1146/annurev-med-051113-024537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363(12):1117–27. doi: 10.1056/NEJMoa1002028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799–807. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Abedin SM, Hamadani M. Ruxolitinib: a potential treatment for corticosteroid refractory acute graft-versus-host disease. Expert Opin Investig Drugs. 2020;29(5):423–7. doi: 10.1080/13543784.2020.1757069. [DOI] [PubMed] [Google Scholar]
- 40.Fridman JS, Scherle PA, Collins R, Burn TC, Li Y, Li J, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol. 2010;184(9):5298–307. doi: 10.4049/jimmunol.0902819. [DOI] [PubMed] [Google Scholar]
- 41.Clark JD, Flanagan ME, Telliez JB. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57(12):5023–38. doi: 10.1021/jm401490p. [DOI] [PubMed] [Google Scholar]
- 42.Russell SM, Tayebi N, Nakajima H, Riedy MC, Roberts JL, Aman MJ, et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science. 1995;270(5237):797–800. doi: 10.1126/science.270.5237.797. [DOI] [PubMed] [Google Scholar]
- 43.Namour F, Desrivot J, Van der Aa A, Harrison P, Tasset C, van’t Klooster G. Clinical Confirmation that the Selective JAK1 Inhibitor Filgotinib (GLPG0634) has a Low Liability for Drug-drug Interactions. Drug Metab Lett. 2016;10(1):38–48. doi: 10.2174/1872312810666151223103353. [DOI] [PubMed] [Google Scholar]
- 44.Van Rompaey L, Galien R, van der Aar EM, Clement-Lacroix P, Nelles L, Smets B, et al. Preclinical characterization of GLPG0634, a selective inhibitor of JAK1, for the treatment of inflammatory diseases. J Immunol. 2013;191(7):3568–77. doi: 10.4049/jimmunol.1201348. [DOI] [PubMed] [Google Scholar]
- 45.Vermeire S, Schreiber S, Petryka R, Kuehbacher T, Hebuterne X, Roblin X, et al. Clinical remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet. 2017;389(10066):266–75. doi: 10.1016/S0140-6736(16)32537-5. [DOI] [PubMed] [Google Scholar]
- 46.NCT02065700 L-tF-uSoGiARAPDCgI.
- 47.Westhovens R, Taylor PC, Alten R, Pavlova D, Enriquez-Sosa F, Mazur M, et al. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis. 2017;76(6):998–1008. doi: 10.1136/annrheumdis-2016-210104. [DOI] [PubMed] [Google Scholar]
- 48.Hornung T, Janzen V, Heidgen FJ, Wolf D, Bieber T, Wenzel J. Remission of recalcitrant dermatomyositis treated with ruxolitinib. N Engl J Med. 2014;371(26):2537–8. doi: 10.1056/NEJMc1412997. [DOI] [PubMed] [Google Scholar]
- 49.Kurtzman DJ, Wright NA, Lin J, Femia AN, Merola JF, Patel M, et al. Tofacitinib Citrate for Refractory Cutaneous Dermatomyositis: An Alternative Treatment. JAMA Dermatol. 2016;152(8):944–5. doi: 10.1001/jamadermatol.2016.0866. [DOI] [PubMed] [Google Scholar]
- 50.Anyanwu CO, Fiorentino DF, Chung L, Dzuong C, Wang Y, Okawa J, et al. Validation of the Cutaneous Dermatomyositis Disease Area and Severity Index: characterizing disease severity and assessing responsiveness to clinical change. Br J Dermatol. 2015;173(4):969–74. doi: 10.1111/bjd.13915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Paik JJ, Christopher-Stine L. A case of refractory dermatomyositis responsive to tofacitinib. Semin Arthritis Rheum. 2017;46(4):e19. doi: 10.1016/j.semarthrit.2016.08.009. [DOI] [PubMed] [Google Scholar]
- 52.Rider LG, Giannini EH, Brunner HI, Ruperto N, James-Newton L, Reed AM, et al. International consensus on preliminary definitions of improvement in adult and juvenile myositis. Arthritis Rheum. 2004;50(7):2281–90. doi: 10.1002/art.20349. [DOI] [PubMed] [Google Scholar]
- 53.Paik JJ, Albayda J, Tiniakou E, Koenig A, Christopher-Stine A. Study of Tofacitinib in Refractory Dermatomyositis (STIR): An Open Label Pilot Study in Refractory Dermatomyositis. 2018 ACR/ARHP Annual Meeting. 2018. [Google Scholar]; ** First clinical trial using tofacitinib in refractory dermatomyositis.
- 54.Ladislau L, Suarez-Calvet X, Toquet S, Landon-Cardinal O, Amelin D, Depp M, et al. JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain. 2018;141(6):1609–21. doi: 10.1093/brain/awy105. [DOI] [PubMed] [Google Scholar]; ** Comprehensive study describing the the efficacy of ruxolitinib in dermatomyositis.
- 55.Sato S, Hirakata M, Kuwana M, Suwa A, Inada S, Mimori T, et al. Autoantibodies to a 140-kd polypeptide, CADM-140, in Japanese patients with clinically amyopathic dermatomyositis. Arthritis Rheum. 2005;52(5):1571–6. doi: 10.1002/art.21023. [DOI] [PubMed] [Google Scholar]
- 56.Kameda H, Nagasawa H, Ogawa H, Sekiguchi N, Takei H, Tokuhira M, et al. Combination therapy with corticosteroids, cyclosporin A, and intravenous pulse cyclophosphamide for acute/subacute interstitial pneumonia in patients with dermatomyositis. J Rheumatol. 2005;32(9):1719–26. [PubMed] [Google Scholar]
- 57.Kawachi Y, Maruyama H, Furuta J, Fujisawa Y, Nakamura Y, Takahashi T, et al. Cutaneous deep necrosis with dermatomyositis: correlation with interstitial pneumonia. Eur J Dermatol. 2007;17(4):345–6. doi: 10.1684/ejd.2007.0220. [DOI] [PubMed] [Google Scholar]
- 58.Chaisson NF, Paik J, Orbai AM, Casciola-Rosen L, Fiorentino D, Danoff S, et al. A novel dermato-pulmonary syndrome associated with MDA-5 antibodies: report of 2 cases and review of the literature. Medicine (Baltimore). 2012;91(4):220–8. doi: 10.1097/MD.0b013e3182606f0b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kurasawa K, Arai S, Namiki Y, Tanaka A, Takamura Y, Owada T, et al. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford). 2018;57(12):2114–9. doi: 10.1093/rheumatology/key188. [DOI] [PubMed] [Google Scholar]
- 60.Kato M, Ikeda K, Kageyama T, Kasuya T, Kumagai T, Furuya H, et al. Successful Treatment for Refractory Interstitial Lung Disease and Pneumomediastinum With Multidisciplinary Therapy Including Tofacitinib in a Patient With Anti-MDA5 Antibody-Positive Dermatomyositis. J Clin Rheumatol. 2019. doi: 10.1097/RHU.0000000000000984. [DOI] [PubMed] [Google Scholar]
- 61.Chen Z, Wang X, Ye S. Tofacitinib in Amyopathic Dermatomyositis-Associated Interstitial Lung Disease. N Engl J Med. 2019;381(3):291–3. doi: 10.1056/NEJMc1900045. [DOI] [PubMed] [Google Scholar]; ** Study showing evidence that tofacitinib may prevent the progression of the interstitial lung disease in patients anti-MDA5 dermatomyositis.
- 62.Wendel S, Venhoff N, Frye BC, May AM, Agarwal P, Rizzi M, et al. Successful treatment of extensive calcifications and acute pulmonary involvement in dermatomyositis with the Janus-Kinase inhibitor tofacitinib - A report of two cases. J Autoimmun. 2019;100:131–6. doi: 10.1016/j.jaut.2019.03.003. [DOI] [PubMed] [Google Scholar]
- 63.Ishikawa Y, Kasuya T, Fujiwara M, Kita Y. Tofacitinib for recurrence of antimelanoma differentiation-associated gene 5 antibody-positive clinically amyopathic dermatomyositis after remission: A case report. Medicine (Baltimore). 2020;99(37):e21943. doi: 10.1097/MD.0000000000021943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Aeschlimann FA, Fremond ML, Duffy D, Rice GI, Charuel JL, Bondet V, et al. A child with severe juvenile dermatomyositis treated with ruxolitinib. Brain. 2018;141(11):e80. doi: 10.1093/brain/awy255. [DOI] [PubMed] [Google Scholar]
- 65.Papadopoulou C, Hong Y, Omoyinmi E, Brogan PA, Eleftheriou D. Janus kinase 1/2 inhibition with baricitinib in the treatment of juvenile dermatomyositis. Brain. 2019;142(3):e8. doi: 10.1093/brain/awz005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sabbagh S, Almeida de Jesus A, Hwang S, Kuehn HS, Kim H, Jung L, et al. Treatment of anti-MDA5 autoantibody-positive juvenile dermatomyositis using tofacitinib. Brain. 2019;142(11):e59. doi: 10.1093/brain/awz293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kim H, Dill S, O’Brien M, Vian L, Li X, Manukyan M, et al. Janus kinase (JAK) inhibition with baricitinib in refractory juvenile dermatomyositis. Ann Rheum Dis. 2020. doi: 10.1136/annrheumdis-2020-218690. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** First clinical trial showing evidence that baricitinib may be effective in patients with refractory juvenile dermatomyositis.
- 68.Yu Z, Wang L, Quan M, Zhang T, Song H. Successful management with Janus kinase inhibitor tofacitinib in refractory juvenile dermatomyositis: a pilot study and literature review. Rheumatology (Oxford). 2020. doi: 10.1093/rheumatology/keaa558. [DOI] [PubMed] [Google Scholar]
- 69.Mukamel M, Horev G, Mimouni M. New insight into calcinosis of juvenile dermatomyositis: a study of composition and treatment. J Pediatr. 2001;138(5):763–6. doi: 10.1067/mpd.2001.112473. [DOI] [PubMed] [Google Scholar]
- 70.Albayda J, Pinal-Fernandez I, Huang W, Parks C, Paik J, Casciola-Rosen L, et al. Antinuclear Matrix Protein 2 Autoantibodies and Edema, Muscle Disease, and Malignancy Risk in Dermatomyositis Patients. Arthritis Care Res (Hoboken). 2017;69(11):1771–6. doi: 10.1002/acr.23188. [DOI] [PMC free article] [PubMed] [Google Scholar]