Download as pdf or txt
Download as pdf or txt
You are on page 1of 35

Accepted Manuscript

Dermatomyositis Part 2: Diagnosis and Treatment

Reid Waldman, M.D., Madeline E. DeWane, B.A., Jun Lu, M.D.

PII: S0190-9622(19)32308-4
DOI: https://doi.org/10.1016/j.jaad.2019.05.105
Reference: YMJD 13599

To appear in: Journal of the American Academy of Dermatology

Received Date: 20 March 2019


Revised Date: 9 May 2019
Accepted Date: 11 May 2019

Please cite this article as: Waldman R, DeWane ME, Lu J, Dermatomyositis Part 2: Diagnosis and
Treatment, Journal of the American Academy of Dermatology (2019), doi: https://doi.org/10.1016/
j.jaad.2019.05.105.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
1

1 Dermatomyositis Part 2: Diagnosis and Treatment


2 Reid Waldman, M.D.1 Madeline E. DeWane, B.A.2 Jun Lu, M.D.1
1
3 University of Connecticut Department of Dermatology, Farmington, CT 06032
2
4 University of Connecticut School of Medicine

PT
5 Corresponding Author:
6 Jun Lu, M.D.
7 21 South Road

RI
8 Farmington, CT 06032
9 jlu@uchc.edu

SC
10 Tel: 860-679-4600
11 Word Count: Manuscript – 2997, Abstract – 134

U
12 Figures: 5
AN
13 Tables: 3
14 References: 88
M

15 Funding: This article has no funding source.


16 Conflicts of Interest: None of the authors have relevant conflicts of interest.
D

17 Key Words: Dermatomyositis, Juvenile Dermatomyositis, cancer associated


18 dermatomyositis, anti-MDA5 dermatomyositis
TE

19 Statement on Prior Presentation: This work has not been previously published or presented.
20 Reprint Requests:
EP

21 Jun Lu, M.D.


22 21 South Road
23 Farmington, CT 06032
C

24 jlu@uchc.edu
AC

25 Tel: 860-679-4600
26

27
ACCEPTED MANUSCRIPT
2

28 Abstract

29 Part II of this CME Series reviews the initial evaluation of patients with suspected

30 dermatomyositis (DM), the relevant work-up for malignancy and interstitial lung disease once a

PT
31 diagnosis of DM is made, and treatment recommendations for patients with DM based on disease

32 severity, presence of systemic symptoms, and myositis specific antibody profiles. This review

RI
33 emphasizes the emerging role of myositis specific antibodies (MSAs) in the diagnosis of DM and

34 highlights how MSAs can be used to guide the appropriate work-up for malignancy and

SC
35 interstitial lung disease. The treatment approach proposed by this CME discusses both

U
36 established and novel therapies for DM and highlights the importance of considering lesion type,
AN
37 degree of muscle involvement, presence of systemic symptoms, presence of MSAs, and patient

38 age when determining the best treat approach for a patient with DM.
M

39
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
3

40 Introduction

41 The evaluation and management of patients with suspected dermatomyositis (DM) is

42 evolving. Part II of this CME Series will review the initial evaluation of patients with suspected

PT
43 DM as well as the relevant work-up for systemic manifestations once a diagnosis is made.

44 Recommendations for treatment based on disease severity, the presence of systemic symptoms,

RI
45 the presence of myositis specific antibodies (MSAs), and patient age will be given. The

46 integration of MSAs into the management of DM patients will be emphasized. Treatment

SC
47 recommendations will include a discussion of emerging therapies.

U
48 The Initial Approach: Diagnosing Dermatomyositis
AN
49 The initial evaluation of patients with suspected DM must include a total body skin

50 examination, objective muscle strength examination, and a laboratory work-up.1 In equivocal


M

51 cases, skin biopsy, muscle biopsy, or muscle imaging may clarify the diagnosis.2 Historically, a
D

52 diagnosis of DM was made based on criteria proposed by Bohan and Peter in 1975.3 Many new
TE

53 classification systems have subsequently been proposed (Table 2). Most recently, the European

54 League Against Rheumatism/American College of Rheumatology (EULAR/ACR) developed the


EP

55 first validated classification criteria with a reported sensitivity of 87% and specificity of 82% for

56 a diagnosis of DM. However, the sensitivity of these guidelines is poor for diagnosing
C

57 amyopathic DM because only limited types of cutaneous lesions are included in their scoring
AC

58 system.4,5 Additionally, the only MSA included in the EULAR/ACR criteria is Anti-Jo-1, as

59 other antibodies were not widely available at the time the guidelines were formed.1

60 The authors have modified the EULAR/ACR diagnostic approach to incorporate the use

61 of multiple newly available MSAs (Table 1, Figure 1). Incorporation of MSAs into diagnostic
ACCEPTED MANUSCRIPT
4

62 criteria of DM is beneficial for several reasons: 1) it facilitates diagnosis of DM, especially in

63 cases of CADM; 2) it separates patients with DM into clinically relevant subsets (which helps

64 tailor the additional work-up for systemic manifestations based on an individual’s MSA profile);

65 and, 3) it obfuscates the need for muscle biopsy in many cases. A 2018 study in JAMA

PT
66 Neurology supports the use of MSAs in diagnosis; in this study, patients with idiopathic

RI
67 inflammatory myopathies (IIMs) were appropriately classified as having DM, inclusion body

68 myositis(IBM), immune-mediated necrotizing myositis (IMNM), or anti-synthetase syndrome

SC
69 (ASS) based solely on MSA profile and clinical manifestations.6

U
70 Evaluation for Systemic Manifestations AN
71 Once a diagnosis of DM has been confirmed, patients must undergo additional work-up

72 to identify systemic manifestations. This work up should be directed by the patient’s MSA
M

73 profile. An in-depth review of the clinical manifestations associated with each MSA is provided
D

74 in Part I of this CME Series. Dermatologists using MSAs as part of their clinical decision

75 making must ensure that they are ordering a testing assay that reliably detects and discriminates
TE

76 between relevant MSAs, as some assays perform less reliably than others.7,8
EP

77 Malignancy Work-Up

78 The estimated prevalence of malignancy in adult DM patients is 20%. The risk of


C

79 developing malignancy is highest within a year of diagnosis and remains elevated for up to 5
AC

80 years.9 Malignancy risk is also increased for males and those over 45 at the time of diagnosis.10,9

81 In juvenile dermatomyositis (JDM), malignancy is extremely uncommon with no cases of

82 malignancy-associated JDM identified in the EuroMyositis registry.11 As highlighted in Part I,


ACCEPTED MANUSCRIPT
5

83 malignancy-associated DM primarily occurs in adults who are either anti-TIF1 or anti-NXP2

84 positive.11,12

85 Patients with JDM do not require any work-up for malignancy.11 A suggested algorithm

PT
86 for appropriate malignancy screening in newly diagnosed adult DM patients is detailed in Figure

87 2. Adult DM patients who are both anti-TIF-1 and anti-NXP2 negative (i.e. low-risk of

RI
88 malignancy associated-DM) require history, physical examination, “age-appropriate” cancer

89 screening, and symptom-targeted cancer screening alone as there is not strong evidence to

SC
90 suggest that individuals without these antibodies are at an appreciably elevated risk for

U
91 malignancy compared to the general population.12 Aggressive work-up for malignancy in these
AN
92 patients is likely to be costly and invasive, and available evidence suggests that it is unlikely to

93 improve outcomes. Possible exceptions to this recommendation are: 1) anti-SAE positive


M

94 individuals, as some studies have demonstrated a risk for malignancy in these patients, although

95 this finding has not been uniformly reproducible and 2) MSA-negative patients, as a recent large,
D

96 retrospective study suggested that these patients have a three-times elevated risk of developing
TE

97 malignancy over matched controls.12


EP

98 Given the paucity of data about cancer risk in these two sub-populations, it may be

99 reasonable to further stratify cancer risk with serum biomarkers, although such an approach has
C

100 never been studied or suggested for this “intermediate-risk” population specifically. Annual
AC

101 soluble Programmed Death Ligand-1 (sPDL-1) measurements can be considered, as extremely

102 elevated levels of sPDL-1 have been associated with malignancy in DM patients.13,14 Use of

103 cancer serum biomarkers (e.g. CEA, CA 19-9) other than prostate specific antigen, a potential

104 component of “age-appropriate” screening, has not been demonstrated to be effective for

105 detecting malignancy in DM patients.15


ACCEPTED MANUSCRIPT
6

106 In addition to “age-appropriate” and “symptom-targeted” malignancy screening,

107 individuals who are anti-TIF1 or anti-NXP2 positive (i.e. high-risk for malignancy-associated

108 DM) should undergo whole body imaging with either CT, MRI, or PET-CT.13,16 These

109 modalities have been proven to detect DM-associated malignancies that would be missed by

PT
110 “age-appropriate” and “symptom-targeted” screening.17 No study has compared superiority of

RI
111 one imaging modality over the others. Studies that have failed to demonstrate benefit with the

112 use of whole-body imaging have not sub-selected for high-risk patients.18

SC
113 The appropriate re-imaging interval in high-risk individuals who had an initially negative

U
114 work-up for malignancy has not been studied. As an increased malignancy risk is present for up
AN
115 to five years after DM onset, some authorities recommend annual imaging until that time point is

116 reached.13 Finally, high-risk individuals who would not receive upper and lower endoscopy as
M

117 part of their “age-appropriate” screening and who do not have malignancy identified on other

118 work-ups should consider undergoing this testing.13


D

119 Interstitial Lung Disease Work-Up


TE

120 Evaluation for interstitial lung disease (ILD) in patients with a new diagnosis of DM
EP

121 involves identifying whether a patient has ILD, and if so, whether a patient has a poor prognosis

122 subset of ILD termed rapidly progressive ILD (RPILD). The early identification of RPILD is
C

123 essential as it is fatal within six months in 50% of cases and the prognosis can be improved if
AC

124 treatment is initiated prior to the development of abnormalities on High-Resolution Computed

125 Tomography (HRCT).19,20

126 Figure 3 provides an algorithm for evaluating DM patients for ILD. All patients require

127 pulmonary function testing (PFTs) and DLCO (diffusion capacity of the lung for carbon
ACCEPTED MANUSCRIPT
7

128 monoxide) at the time of diagnosis.21 In asymptomatic patients with restrictive physiology

129 present on PFTs and decreased DLCO, or in patients with symptoms suggestive of ILD, HRCT is

130 indicated.21 If ILD is not present on an initial work-up, patients can be monitored clinically with

131 a plan to repeat HRCT if new or worsening pulmonary symptoms develop.21 All patients with

PT
132 evidence of ILD require urgent pulmonology evaluation.

RI
133 Anti-MDA5 antibodies are present in at least half of all cases of DM-associated ILD and

134 more than 80% of cases of DM-associated RPILD.20,22,23 In JDM, anti-MDA5 positivity is also

SC
135 predictive of ILD and RPILD with an estimated sensitivity of ~70% for ILD.24 Because RPILD

U
136 can evade imaging during its early stages, testing serum biomarkers that correlate with the
AN
137 presence of ILD and that are elevated prior to imaging changes should be considered.25,26

138 Elevated levels of serum ferritin, Interleukin 18, Krebs von den Lungen-6 (KL-6) and anti-
M

139 MDA5 antibodies themselves have been associated with the presence of ILD, and laboratory

140 testing for these biomarkers should be considered in all patients with anti-MDA5
D

141 dermatomyositis.27–31
TE

142 Other Systemic Work-Up


EP

143 Part I of this CME series discusses the many other potential systemic manifestations of

144 DM and JDM. Given their relative infrequency, screening for these manifestations with a
C

145 targeted review of symptoms is reasonable.


AC

146 General Treatment Approach

147 Management of DM is nuanced; dermatologists frequently diagnose the disease and have

148 primary responsibility for the cutaneous manifestations of the disease, but myopathy and other
ACCEPTED MANUSCRIPT
8

149 systemic manifestations often drive therapy. The appropriate treatment approach is determined

150 by consideration of five factors:

151 1) Lesion type – Is the lesion nonvasculopathic (e.g. shawl sign, heliotrope rash),

PT
152 vasculopathic (digital pulp ulcers, inverse gottron’s papules), or calcinotic?

153 2) Degree of muscle involvement – Is the patient amyopathic/hypomyopathic? Does the

RI
154 patient have persistent cutaneous symptoms despite having controlled myopathy? Is the

155 patient post-myopathic (i.e. have the patient’s muscle symptoms resolved despite being

SC
156 off treatment but residual cutaneous disease is still present)?

U
157 3) Presence of systemic symptoms – Are other organ systems involved? Is there an
AN
158 associated malignancy?

159 4) Presence of MSAs – What clinical subset does the patient have as suggested by the
M

160 presence of MSAs?

161 5) Patient age – Does the patient have adult or juvenile dermatomyositis?
D

162 This section will discuss each of these factors with an emphasis on the use of a multidisciplinary
TE

163 approach in settings where muscle involvement or systemic symptoms are present.
EP

164 Considering Lesion Type

165 Nonvasculopathic Cutaneous Disease


C
AC

166 Three layers of therapy should be used for all patients with nonvasculopathic disease: sun

167 protection, topical therapy with corticosteroids and/or calcineurin inhibitors, and systemic

168 therapy.32,33 This section will focus on systemic therapies. A treatment algorithm for adult DM

169 is presented in Figure 4.

170 Systemic Corticosteroids


ACCEPTED MANUSCRIPT
9

171 Systemic corticosteroids are the gold standard initial treatment for DM-related myopathy.

172 However, they should not be used in CADM and should not be used as a monotherapy as this

173 approach is frequently ineffective and associated with the development of unacceptable long-

174 term adverse effects.34–36 Similarly, in cases where a patient’s myopathy is controlled with

PT
175 corticosteroids but cutaneous symptoms persist, a dose increase in corticosteroids alone is not

RI
176 recommended. A combination of systemic corticosteroids with oral immunosuppressants or

177 biologics should be used at disease onset in patients with myopathy or other systemic symptoms,

SC
178 similar to how combination therapy is employed in bullous disorders to limit systemic

179 corticosteroid use.34,37

U
AN
180 Antimalarials

181 Traditional treatment algorithms have emphasized hydroxychloroquine as the first-line


M

182 systemic agent for cutaneous DM. However, recent evidence suggests that patients treated with
D

183 hydroxychloroquine are more likely to flare their cutaneous disease than they are to achieve

184 satisfactory disease control from hydroxychloroquine monotherapy.35,36,38,39Patient MSA profiles


TE

185 may predict risk of cutaneous flare after hydroxychloroquine initiation with anti-SAE positive
EP

186 patients at the highest risk and anti-MDA5 patients without demonstrable risk. 40 Furthermore,

187 unlike other systemic therapies, none of the antimalarials have an effect on the non-cutaneous
C

188 manifestations of DM (e.g. myopathy, ILD).41,42 Given the favorable side effect profile of
AC

189 antimalarials they can be considered as adjuvants when disease control is inadequate with other

190 systemic agents.35,36,42,43

191 Mycophenolate Mofetil (MMF) and Methotrexate (MTX)


ACCEPTED MANUSCRIPT
10

192 In the absence of vasculopathic or calcinotic lesions the first line systemic therapies for

193 nonvasculopathic DM are mycophenolate mofetil (MMF) and methotrexate (MTX).38,44–48 Both

194 of these medications often require high dosing, with many adults requiring 3 grams of MMF

195 daily or 25 mg of MTX weekly. These medications should be started in conjunction with

PT
196 systemic corticosteroids when myopathy is present. However, systemic corticosteroids do not

RI
197 need to be used in conjunction with these medications when treating CADM or post-myopathic

198 cutaneous disease.

SC
199 There are no head-to-head studies comparing MMF and MTX, but several considerations

U
200 may favor the use of one agent over the other; MTX often has faster onset (~ 4 weeks) and has
AN
201 clinical trial data supporting its use as a steroid-sparing agent. Additionally, MMF is effective for

202 treating ILD .37,49,50 Treatment failure with one agent is not predictive of treatment failure with
M

203 the other.


D

204 Rituximab
TE

205 In cases where a combination of systemic corticosteroids and an oral immunosuppressant

206 fail, rituximab is the appropriate next step in therapy.51–53 In individuals with vasculopathic
EP

207 and/or calcinotic lesions, adults with anti-MDA5 positivity, or children with NXP-2 positivity,

208 rituximab plus systemic corticosteroids can be considered as a first-line treatment.54–56 Support
C

209 for the use of rituximab comes from the largest clinical trial ever conducted for IIMs, the
AC

210 Rituximab in Myositis (RIM) trial.51 This trial demonstrated that 83% of children and adults with

211 DM who previously failed systemic corticosteroids and at least one immunosuppressant

212 improved with rituximab and were tapered off systemic steroids more quickly.51 MSA-positive

213 individuals had a greater chance of responding favorably to rituximab than MSA-negative

214 individuals.51
ACCEPTED MANUSCRIPT
11

215 Additionally, several recent findings suggest rituximab may have disease modifying

216 properties. Rituximab is the only treatment associated with improvement in nail fold capillary

217 abnormalities which may represent prevention of pathogenic vessel damage. In cases where

218 MSAs are presumed to be pathogenic, (e.g. anti-MDA5 DM) rituximab likely works through a

PT
219 similar mechanism as in pemphigus (a condition in which it has been shown to be disease

RI
220 modifying).51,57 The two major limitations to the use of rituximab are that it has a slow onset and

221 a risk of serious infection greater than 6%.51,56

SC
222 The frequency with which individuals receiving rituximab should undergo an additional

U
223 round of treatment is unknown. In the RIM trial, CD19+ numbers rebounded above 5 cells/uL at
AN
224 weeks 32-36 on average.51 Based on the pemphigus literature, it may be reasonable to trend

225 peripheral B cell concentrations to guide therapy.58


M

226 Intravenous Immunoglobulin (IVIG)


D

227 A reasonable treatment option in patients who have failed or who are intolerant of
TE

228 rituximab is intravenous immunoglobulin (IVIG).43,59,60 It is also recommended for patients with

229 controlled myopathy but persistent cutaneous disease.61 In cases with severe disease that is
EP

230 refractory to IVIG, subcutaneous immunoglobulin administration can be considered if

231 available.62
C

232 Calcineurin Inhibitors


AC

233 Calcineurin inhibitors (typically cyclosporine and less frequently tacrolimus) are

234 reasonable third-line options or are useful in cases in which myopathy is controlled but other

235 immunosuppressants are not controlling cutaneous disease.48 Although cyclosporine is as

236 effective as MTX based on the Pediatric Rheumatology International Trials Organization
ACCEPTED MANUSCRIPT
12

237 (PRINTO) data, MTX and MMF are preferred because the PRINTO trial demonstrated that

238 cyclosporine use was associated with a greater risk of serious adverse effects.37 Calcineurin

239 inhibitors are also a reasonable choice in patients with co-morbid interstitial lung disease.63,64

PT
240 Other Traditional Therapies

241 Other therapies that can be considered include infliximab, azathioprine, and

RI
242 cyclophosphamide. However, these should be reserved for refractory cases given the plethora of

SC
243 superior options listed above.65–67

244 Vasculopathic Cutaneous Disease

245
U
Vasculopathic skin lesions include ulceration, inverse Gottron’s papules, and nailfold
AN
246 capillary abnormalities. These lesions are notoriously refractory to immunosuppressive therapy
M

247 and confer significant morbidity even in the absence of other cutaneous disease. The only

248 systemic agent with robust data supporting its use for vasculopathic lesions is rituximab.
D

249 However, rituximab alone is often ineffective for treating ulceration.56,68 Intralesional
TE

250 corticosteroids are frequently used for treating ulcerations and inverse Gottron’s papules and

251 may be effective, but recent evidence supports using vasodilatory agents. Case studies suggest
EP

252 that nifedipine, sildenafil, intravenous prostaglandins, and bosentan should be added as early

253 adjuncts given the otherwise refractory nature of these lesions.69


C
AC

254 Calcinosis Cutis

255 Like vasculopathic lesions, calcinotic lesions are typically refractory to

256 immunosuppressive therapy. Patients with JDM and calcinosis should be preferentially treated

257 with IVIG as it has the best data supporting its use for calcinosis specifically.59,70,71 For a detailed
ACCEPTED MANUSCRIPT
13

258 discussion of calcinosis cutis-directed therapies, the authors encourage readers to review the

259 JAAD CME Series on calcinosis cutis.72

260 Considering Degree of Muscle Involvement

PT
261 Patients with myopathy should be managed in conjunction with a rheumatologist or

262 neurologist. Controlled cutaneous disease is not predictive of controlled myopathy and vice

RI
263 versa. Unlike in patients with CADM (for whom monotherapy with oral immunosuppressants is

SC
264 a reasonable first-line therapy), first line therapy for management of the myopathy component of

265 DM is the simultaneous initiation of corticosteroids and a steroid-sparing immunosuppressant

U
266 (e.g. MMF, MTX).37 Subsequent treatment choices in individuals with recalcitrant myopathy are
AN
267 similar to those highlighted above for the treatment of refractory cutaneous disease.

268 The appropriate treatment for persistent cutaneous involvement in patients with
M

269 controlled myopathy depends on the treatment the patient is currently receiving and the degree of
D

270 severity of the cutaneous involvement.73 Dose escalations of an oral immunosuppressant,


TE

271 initiation of hydroxychloroquine, initiation of IVIG, or initiation of rituximab can all be

272 reasonable next steps depending on the clinical scenario.61Post-myopathic cutaneous disease can
EP

273 be managed similarly to CADM.

274 Considering Systemic Symptoms


C
AC

275 Treatment of the systemic symptoms associated with DM is beyond the scope of this

276 CME series and should be addressed as part of a multidisciplinary approach.

277 Considering MSAs


ACCEPTED MANUSCRIPT
14

278 MSAs will undoubtedly be used to personalize treatment decisions for patients with DM

279 in the future. However, at this time there is a paucity of data supporting such an approach.

280 Several anecdotally supported treatment considerations have been mentioned above.

PT
281 Considering Age

282 Only two clinical trials have specifically evaluated patients with JDM (the PRINTO and

RI
283 RIM trials), but consensus opinions are also available from the Childhood Arthritis and

SC
284 Rheumatology Research Alliance (CARRA) and Single Hub and Access point for pediatric

285 Rheumatology in Europe (SHARE) registries.37,51,56,74,75 Review of these pivotal studies

U
286 supports the approach to JDM proposed in Figure 5. All patients should at least be started on a
AN
287 combination of systemic corticosteroids with either MTX or IVIG to decrease long-term steroid

288 burden.37,75 In patients that fail first-line treatment, have severe disease with ulceration,
M

289 calcinosis, or lipodystrophy at the time of presentation, or have poor prognosis NXP-2 disease,

rituximab plus systemic corticosteroids can be considered.56 Other biologics, cyclophosphamide,


D

290

291 and JAK inhibitors can be considered in patients with refractory disease.65,67,76
TE

292 Emerging Treatments


EP

293 Several novel approaches to DM have recently garnered significant interest. JAK

294 inhibitors (JKIs) will be discussed here as they have the most robust evidence for the treatment
C

295 of cutaneous DM. Other emerging treatments are listed in Table 4.


AC

296 Jak Kinase Inhibitors

297 DM is driven by type I interferons. Both in vivo and in vitro data has shown that JKIs

298 decrease levels of type I interferons in individuals with DM.77 Case series have shown that

299 several JKIs are effective for treating refractory cutaneous disease.78 One series has also shown
ACCEPTED MANUSCRIPT
15

300 that JKIs may be an effective add-on therapy in patients with RP-ILD.79 There is an ongoing

301 clinical trial assessing the efficacy of tofacitinib in refractory DM that is assessing both skin and

302 muscle outcomes.

PT
303 Conclusion

304 A thorough initial assessment of patients with suspected DM is essential to making a

RI
305 diagnosis with as little delay and morbidity as possible. The management of DM is nuanced and

SC
306 requires a constant assessment for the development of systemic symptoms. A multi-disciplinary

307 approach is recommended.

308
U
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
16

309 Figure 1. Diagnostic algorithm for adult and juvenile DM.


310 *As indicated in Part I Table I.
311 **Pts < age 18 at time of symptom onset are considered to have JDM.
312 ***ADM pts should be monitored regularly for development of muscle involvement.

PT
313

314 Figure 2. Malignancy work-up for newly diagnosed DM patients.

RI
315

316 Figure 3. Interstitial lung disease work-up for newly diagnosed DM patients.

SC
317
318 Figure 4. Adult DM treatment algorithm
319
320 Figure 5. JDM treatment algorithm
U
AN
321

322
M

323

324
D

325
TE

326

327
EP

328

329

330
C

331
AC

332

333

334

335

336
ACCEPTED MANUSCRIPT
17

- Total body skin exam


Exam History & physical examination - Manual strength testing
of bilateral extremities
and neck flexors
- CK
Baseline laboratory testing Muscle enzymes - LDH
- AST

PT
- Aldolase
- Mi2
- TIF1

RI
DM-specific autoantibodies - MDA5
- NXP2
- SAE

SC
- Jo-1
Anti-synthetase syndrome - PL-7
Auto-antibody testing autoantibodies - PL-12
- EJ

U
- OJ
IMNM autoantibodies - SRP
AN
(if indicated clinically) - HMGCR

- ANA
Other connective tissue disease- - Ro/La
M

related autoantibodies - dsDNA


(if indicated clinically) - Anti-Sm
- Scl-70
D

If the above testing is equivocal - T2 weighted MRI of area


of weakness
TE

Further testing If diagnosis remains uncertain - Muscle biopsy of


affected area identified
on MRI
337 Table I. Initial workup for suspected DM based on EULAR/ACR criteria
EP

338

339
C

340
AC

341

342

343

344

345

346
ACCEPTED MANUSCRIPT
18

Classification Criteria Included Entities Defined Benefits & Limitations


system
Bohan and Clinical -Definite DM -High sensitivity but low
Peter (1975) -Skin rash (heliotrope rash or -Probable DM specificity82
80,81
Gottron sign) -Possible DM - Outdated
-Symmetric proximal muscle -Definite PM conceptualization of DM
weakness -Probable PM and PM as related entities

PT
-Possible PM on a spectrum of
Laboratory inflammatory myopathy83
-Elevation of skeletal muscle -Does not specify how to

RI
enzymes exclude other forms of
myopathy82
Other
-EMG findings

SC
-Muscle biopsy findings

Tanimoto Clinical

U -DM High sensitivity but low


AN
(1995)84 -Skin rash (heliotrope rash or -PM specificity82
Gottron’s sign or linear extensor
erythema)
-Proximal muscle weakness
M

-Muscle pain on grasping or


spontaneous pain
-Nondestructive arthritis or
D

arthralgia
-Fever
TE

Laboratory
-Elevated CK or aldolase
-Elevated CRP or ESR
EP

-Positive anti-Jo-1 antibodies

Other
-Abnormal EMG findings
C

-Abnormal muscle biopsy


Targoff Clinical -Definite IIM -Sensitivity 93%, specificity
AC

(1997)85 -Skin rash (heliotrope rash or -Probable IIM 89% using EULAR/ACR
Gottron sign) -Possible IIM dataset82
-Symmetric proximal muscle -Subclassifies
weakness DM, IBM, JDM,
amyopathic DC
Laboratory (ADM)
-Elevation of skeletal muscle
enzymes
-Presence of any MSA
ACCEPTED MANUSCRIPT
19

Other
-EMG findings
-Muscle biopsy findings

Dalakas and Clinical -Definite PM -High specificity but low

PT
Hohlfield -Skin rash (or calcinosis) -Probable PM sensitivity using
(2003)86 -Myopathic muscle weakness -Definite DM EULAR/ACR dataset82
-Probable DM

RI
Laboratory -Definite ADM
-Elevated muscle enzymes

Other

SC
-Muscle biopsy findings
-EMG findings
ENMC (2003)87 Clinical -Definite DM -High specificity but low

U
-Age -Probable DM sensitivity using
-Muscle weakness (specifies time -ADM EULAR/ACR dataset82
AN
course and pattern) -DM sine
-Skin rash (heliotrope, periorbital dermatitis
edema, Gottron’s papules/sign, V -Definite PM
sign, shawl sign, holster sign) -Probable PM
M

-Non-specific
Laboratory myositis
-Elevated CK -Immune-
D

-Detection of MSAs mediated


necrotizing
TE

Other myopathy
-EMG findings (IMNM)
-MRI findings
-Muscle biopsy findings
EP

EULAR/ACR Clinical -Algorithm -Large dataset


(2017)82 -Age determines IIM -Sensitivity 93%, specificity
-Muscle weakness probibility 88% (with muscle biopsy
-Skin rash (heliotrope rash, -Subclassifies data)
C

Gottron’s papules, Gottron’s sign) PM, IBM, DM, -Sensitivity 87%, specificity
ADM, JDM 82% (without muscle
AC

Laboratory biopsy data)


-Positive anti-Jo-1 antibody -Subclassification limited by
-Elevated CK, LDH, AST, ALT small sample size for some
entities
Other -Requires additional
-Muscle biopsy findings validation in Asian and
African populations
-Can diagnose DM without
muscle biopsy when typical
skin findings are present
ACCEPTED MANUSCRIPT
20

Mariampillai -47 variables used in multiple -DM


(2018)88 correspondence analysis -IBM
-Included sociodemographic -IMNM
variables, skin lesions, biological -ASS
variables (including CK levels and
MSAs), histological variables,
clinical muscular variables, and

PT
extra-muscular variables
347 Table II. Classification systems for IIMs.

348

RI
349

SC
350

351

352

353
U
AN
354

355
M

356

357
D

358
TE

359

360

361
EP

362

363
C

364
AC

365

366

367

368

369

Drug Name Route of Mechanism of Action Ongoing Clinical Trial (Y/N)


ACCEPTED MANUSCRIPT
21

Administration
Tocilizumab IV IL-6 Inhibitor Y
Aletuzumab IV Anti-CD52 N
Abatacept IV Co-stimulatory Modulator Y
Infliximab IV TNF inhibitor Y
Anakinra SubQ IL-1 Inhibitor N
Eculizumab IV C5-blocking agent N

PT
Apremilast PO PDE4 Inhibitor Y
370 Table III. Emerging treatments for DM

RI
371

372

SC
373

374

U
375
AN
376

377
M

378

379
D

380
TE

381

382
EP

383

384
C

385
AC

386

387

388

389

390
ACCEPTED MANUSCRIPT
22

391 Abbreviations
392 Myositis specific antibodies (MSAs)
393 European League Against Rheumatism/American College of Rheumatology (EULAR/ACR)
394 Idiopathic Inflammatory Myopathy (IIM)

PT
395 Juvenile dermatomyositis (JDM)
396 Inclusion body myositis (IBM)
397 Immune mediated necrotizing myositis (IMNM)

RI
398 Anti-synthetase syndrome (ASS)
399 Soluble Programmed Death Ligand-1 (sPDL-1)

SC
400 Interstitial lung disease (ILD)
401 Rapidly progressive ILD (RPILD)

U
402 High-Resolution Computed Tomography (HRCT)
AN
403 Pulmonary function testing (PFTs)
404 DLCO (diffusion capacity of the lung for carbon monoxide)
M

405 Krebs von den Lungen-6 (KL-6)


406 Mycophenolate mofetil (MMF)
D

407 Methotrexate (MTX)


408 Rituximab in Myositis (RIM)
TE

409 Intravenous Immunoglobulin (IVIG)


410 Pediatric Rheumatology International Trials Organization (PRINTO)
EP

411 Childhood Arthritis and Rheumatology Research Alliance (CARRA)


412 Single Hub and Access point for pediatric Rheumatology in Europe (SHARE)
C

413 JAK Inhibitors (JKIs)


414
AC

415
416
417
418
419
420
ACCEPTED MANUSCRIPT
23

421 References
422 1. Bottai M, Tjärnlund A, Santoni G, et al. EULAR/ACR classification criteria for adult and juvenile
423 idiopathic inflammatory myopathies and their major subgroups: a methodology report. RMD
424 Open. 2017;3(2):e000507. doi:10.1136/rmdopen-2017-000507

425 2. Ran J, Ji S, Morelli JN, Wu G, Li X. T2 mapping in dermatomyositis/polymyositis and correlation

PT
426 with clinical parameters. Clin Radiol. 2018;73(12):1057.e13-1057.e18.
427 doi:10.1016/j.crad.2018.07.106

428 3. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med.

RI
429 1975;292(7):344-347. doi:10.1056/NEJM197502132920706

430 4. Hočevar A, Rotar Z, Krosel M, Čučnik S, Tomšič M. Performance of the 2017 European League

SC
431 Against Rheumatism/American College of Rheumatology classification criteria for adult and
432 juvenile idiopathic inflammatory myopathies in clinical practice. Ann Rheum Dis. 2018;77(12):e90.
433 doi:10.1136/annrheumdis-2017-212774

U
434 5. Patel B, Khan N, Werth VP. Applicability of EULAR/ACR classification criteria for dermatomyositis to
435 amyopathic disease. J Am Acad Dermatol. 2018;79(1):77-83.e1. doi:10.1016/j.jaad.2017.12.055
AN
436 6. Mariampillai K, Granger B, Amelin D, et al. Development of a New Classification System for
437 Idiopathic Inflammatory Myopathies Based on Clinical Manifestations and Myositis-Specific
438 Autoantibodies. JAMA Neurol. 2018;75(12):1528-1537. doi:10.1001/jamaneurol.2018.2598
M

439 7. Ghirardello A, Rampudda M, Ekholm L, et al. Diagnostic performance and validation of


440 autoantibody testing in myositis by a commercial line blot assay. Rheumatol Oxf Engl.
D

441 2010;49(12):2370-2374. doi:10.1093/rheumatology/keq281

442 8. Victor J, Zanardo L, Héron-Mermin D, et al. [Retrospective analysis of anti-TIF1gamma, anti-NXP2


TE

443 and anti-SAE1/2 antibodies carriers at Bordeaux university hospital from November 2014 to
444 February 2017]. Rev Med Interne. 2019;40(2):70-81. doi:10.1016/j.revmed.2018.11.003

445 9. Qiang JK, Kim WB, Baibergenova A, Alhusayen R. Risk of Malignancy in Dermatomyositis and
EP

446 Polymyositis. J Cutan Med Surg. 2017;21(2):131-136. doi:10.1177/1203475416665601

447 10. Yang Z, Lin F, Qin B, Liang Y, Zhong R. Polymyositis/dermatomyositis and malignancy risk: a
C

448 metaanalysis study. J Rheumatol. 2015;42(2):282-291. doi:10.3899/jrheum.140566


AC

449 11. Lilleker JB, Vencovsky J, Wang G, et al. The EuroMyositis registry: an international collaborative
450 tool to facilitate myositis research. Ann Rheum Dis. 2018;77(1):30-39. doi:10.1136/annrheumdis-
451 2017-211868

452 12. Yang H, Peng Q, Yin L, et al. Identification of multiple cancer-associated myositis-specific
453 autoantibodies in idiopathic inflammatory myopathies: a large longitudinal cohort study. Arthritis
454 Res Ther. 2017;19. doi:10.1186/s13075-017-1469-8
ACCEPTED MANUSCRIPT
24

455 13. Selva-O’Callaghan A, Martinez-Gómez X, Trallero-Araguás E, Pinal-Fernández I. The diagnostic


456 work-up of cancer-associated myositis. Curr Opin Rheumatol. 2018;30(6):630-636.
457 doi:10.1097/BOR.0000000000000535

458 14. Chen H, Peng Q, Yang H, et al. Increased Levels of Soluble Programmed Death Ligand 1 Associate
459 with Malignancy in Patients with Dermatomyositis. J Rheumatol. 2018;45(6):835-840.
460 doi:10.3899/jrheum.170544

PT
461 15. Lim CH, Tseng C-W, Lin C-T, et al. The clinical application of tumor markers in the screening of
462 malignancies and interstitial lung disease of dermatomyositis/polymyositis patients: A
463 retrospective study. SAGE Open Med. 2018;6:2050312118781895.

RI
464 doi:10.1177/2050312118781895

465 16. Li Y, Zhou Y, Wang Q. Multiple values of 18F-FDG PET/CT in idiopathic inflammatory myopathy. Clin

SC
466 Rheumatol. 2017;36(10):2297-2305. doi:10.1007/s10067-017-3794-3

467 17. Leatham H, Schadt C, Chisolm S, et al. Evidence supports blind screening for internal malignancy in
468 dermatomyositis: Data from 2 large US dermatology cohorts. Medicine (Baltimore).

U
469 2018;97(2):e9639. doi:10.1097/MD.0000000000009639
AN
470 18. Maliha PG, Hudson M, Abikhzer G, Singerman J, Probst S. 18F-FDG PET/CT versus conventional
471 investigations for cancer screening in autoimmune inflammatory myopathy in the era of novel
472 myopathy classifications. Nucl Med Commun. January 2019.
473 doi:10.1097/MNM.0000000000000981
M

474 19. Kurasawa K, Arai S, Namiki Y, et al. Tofacitinib for refractory interstitial lung diseases in anti-
475 melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatol Oxf
D

476 Engl. 2018;57(12):2114-2119. doi:10.1093/rheumatology/key188


TE

477 20. Chen Z, Cao M, Plana MN, et al. Utility of anti-melanoma differentiation-associated gene 5
478 antibody measurement in identifying patients with dermatomyositis and a high risk for developing
479 rapidly progressive interstitial lung disease: a review of the literature and a meta-analysis. Arthritis
480 Care Res. 2013;65(8):1316-1324. doi:10.1002/acr.21985
EP

481 21. Vij R, Strek ME. Diagnosis and Treatment of Connective Tissue Disease-Associated Interstitial Lung
482 Disease. Chest. 2013;143(3):814-824. doi:10.1378/chest.12-0741
C

483 22. Li L, Wang H, Wang Q, et al. Myositis-specific autoantibodies in dermatomyositis/polymyositis with


484 interstitial lung disease. J Neurol Sci. 2019;397:123-128. doi:10.1016/j.jns.2018.12.040
AC

485 23. Ikeda S, Arita M, Morita M, et al. Interstitial lung disease in clinically amyopathic dermatomyositis
486 with and without anti-MDA-5 antibody: to lump or split? BMC Pulm Med. 2015;15:159.
487 doi:10.1186/s12890-015-0154-4

488 24. Tansley SL, Betteridge ZE, Gunawardena H, et al. Anti-MDA5 autoantibodies in juvenile
489 dermatomyositis identify a distinct clinical phenotype: a prospective cohort study. Arthritis Res
490 Ther. 2014;16(4):R138. doi:10.1186/ar4600
ACCEPTED MANUSCRIPT
25

491 25. Nishioka A, Tsunoda S, Abe T, et al. Serum neopterin as well as ferritin, soluble interleukin-2
492 receptor, KL-6 and anti-MDA5 antibody titer provide markers of the response to therapy in
493 patients with interstitial lung disease complicating anti-MDA5 antibody-positive dermatomyositis.
494 Mod Rheumatol. November 2018:1-7. doi:10.1080/14397595.2018.1548918

495 26. Kurtzman DJB, Vleugels RA. Anti-melanoma differentiation-associated gene 5 (MDA5)
496 dermatomyositis: A concise review with an emphasis on distinctive clinical features. J Am Acad

PT
497 Dermatol. 2018;78(4):776-785. doi:10.1016/j.jaad.2017.12.010

498 27. Hanaoka M, Katsumata Y, Kawasumi H, Kawaguchi Y, Yamanaka H. KL-6 is a long-term disease-
499 activity biomarker for interstitial lung disease associated with polymyositis/dermatomyositis, but is

RI
500 not a short-term disease-activity biomarker. Mod Rheumatol. November 2018:1-20.
501 doi:10.1080/14397595.2018.1553488

SC
502 28. Li L, Wang Q, Wen X, et al. Assessment of anti-MDA5 antibody as a diagnostic biomarker in
503 patients with dermatomyositis-associated interstitial lung disease or rapidly progressive interstitial
504 lung disease. Oncotarget. 2017;8(44):76129-76140. doi:10.18632/oncotarget.19050

U
505 29. Yang Y, Yin G, Hao J, Xie Q, Liu Y. Serum Interleukin-18 Level is Associated With Disease Activity
506 and Interstitial Lung Disease in Patients With Dermatomyositis. Arch Rheumatol. 2017;32(3):181-
AN
507 188. doi:10.5606/ArchRheumatol.2017.6175

508 30. Kobayashi N, Takezaki S, Kobayashi I, et al. Clinical and laboratory features of fatal rapidly
509 progressive interstitial lung disease associated with juvenile dermatomyositis. Rheumatol Oxf Engl.
M

510 2015;54(5):784-791. doi:10.1093/rheumatology/keu385

511 31. Sato S, Uejima Y, Nanbu M, et al. Clinical analysis and outcome of interstitial lung disease
D

512 complicated with juvenile dermatomyositis and juvenile polymyositis. Mod Rheumatol.
513 2017;27(4):652-656. doi:10.1080/14397595.2016.1231102
TE

514 32. Muro Y, Sugiura K, Akiyama M. Cutaneous Manifestations in Dermatomyositis: Key Clinical and
515 Serological Features-a Comprehensive Review. Clin Rev Allergy Immunol. 2016;51(3):293-302.
516 doi:10.1007/s12016-015-8496-5
EP

517 33. Yoshimasu T, Ohtani T, Sakamoto T, Oshima A, Furukawa F. Topical FK506 (tacrolimus) therapy for
518 facial erythematous lesions of cutaneous lupus erythematosus and dermatomyositis. Eur J
519 Dermatol EJD. 2002;12(1):50-52.
C

520 34. Kohsaka H, Mimori T, Kanda T, et al. Treatment consensus for management of polymyositis and
AC

521 dermatomyositis among rheumatologists, neurologists and dermatologists. Mod Rheumatol.


522 2019;29(1):1-19. doi:10.1080/14397595.2018.1521185

523 35. Anyanwu CO, Chansky PB, Feng R, Carr K, Okawa J, Werth VP. The systemic management of
524 cutaneous dermatomyositis: Results of a stepwise strategy. Int J Womens Dermatol.
525 2017;3(4):189-194. doi:10.1016/j.ijwd.2017.05.001

526 36. Pinard J, Femia AN, Roman M, et al. Systemic Treatment for Clinically Amyopathic
527 Dermatomyositis at 4 Tertiary Care Centers. JAMA Dermatol. January 2019.
528 doi:10.1001/jamadermatol.2018.5215
ACCEPTED MANUSCRIPT
26

529 37. Ruperto N, Pistorio A, Oliveira S, et al. Prednisone versus prednisone plus ciclosporin versus
530 prednisone plus methotrexate in new-onset juvenile dermatomyositis: a randomised trial. Lancet
531 Lond Engl. 2016;387(10019):671-678. doi:10.1016/S0140-6736(15)01021-1

532 38. Galimberti F, Li Y, Fernandez AP. Clinically amyopathic dermatomyositis: clinical features, response
533 to medications and malignancy-associated risk factors in a specific tertiary-care-centre cohort. Br J
534 Dermatol. 2016;174(1):158-164. doi:10.1111/bjd.14227

PT
535 39. Pelle MT, Callen JP. Adverse cutaneous reactions to hydroxychloroquine are more common in
536 patients with dermatomyositis than in patients with cutaneous lupus erythematosus. Arch
537 Dermatol. 2002;138(9):1231-1233; discussion 1233.

RI
538 40. Dalakas MC, Illa I, Dambrosia JM, et al. A controlled trial of high-dose intravenous immune globulin
539 infusions as treatment for dermatomyositis. N Engl J Med. 1993;329(27):1993-2000.

SC
540 doi:10.1056/NEJM199312303292704

541 41. Piguet V, Choy E. Dermatomyositis: a slow path towards targeted therapies or will conventional
542 therapies prevail? Br J Dermatol. 2018;179(6):1233-1234. doi:10.1111/bjd.16230

543 42.
U
Woo TY, Callen JP, Voorhees JJ, Bickers DR, Hanno R, Hawkins C. Cutaneous lesions of
AN
544 dermatomyositis are improved by hydroxychloroquine. J Am Acad Dermatol. 1984;10(4):592-600.

545 43. Callander J, Robson Y, Ingram J, Piguet V. Treatment of clinically amyopathic dermatomyositis in
546 adults: a systematic review. Br J Dermatol. 2018;179(6):1248-1255. doi:10.1111/bjd.14726
M

547 44. Olivo Pallo PA, de Souza FHC, Miossi R, Shinjo SK. Mycophenolate mofetil in patients with
548 refractory systemic autoimmune myopathies: case series. Adv Rheumatol Lond Engl.
D

549 2018;58(1):34. doi:10.1186/s42358-018-0035-7


TE

550 45. Edge JC, Outland JD, Dempsey JR, Callen JP. Mycophenolate mofetil as an effective corticosteroid-
551 sparing therapy for recalcitrant dermatomyositis. Arch Dermatol. 2006;142(1):65-69.
552 doi:10.1001/archderm.142.1.65
EP

553 46. Gelber AC, Nousari HC, Wigley FM. Mycophenolate mofetil in the treatment of severe skin
554 manifestations of dermatomyositis: a series of 4 cases. J Rheumatol. 2000;27(6):1542-1545.

555 47. Mainetti C, Terziroli Beretta-Piccoli B, Selmi C. Cutaneous Manifestations of Dermatomyositis: a


C

556 Comprehensive Review. Clin Rev Allergy Immunol. 2017;53(3):337-356. doi:10.1007/s12016-017-


557 8652-1
AC

558 48. Isak V, Jorizzo JL. Recent developments on treatment strategies and the prognosis of
559 dermatomyositis: a review. J Dermatol Treat. 2018;29(5):450-459.
560 doi:10.1080/09546634.2017.1403549

561 49. Waldman R, Strober BE. Laboratory Monitoring Requirements During Mycophenolate Mofetil
562 Therapy for Dermatologic Conditions: A Single Institution Retrospective Chart Review. J Am Acad
563 Dermatol. 2019;0(0). doi:10.1016/j.jaad.2019.02.040
ACCEPTED MANUSCRIPT
27

564 50. Hayashi M, Kikuchi T, Takada T. Mycophenolate mofetil for the patients with interstitial lung
565 diseases in amyopathic dermatomyositis with anti-MDA-5 antibodies. Clin Rheumatol.
566 2017;36(1):239-240. doi:10.1007/s10067-016-3443-2

567 51. Oddis CV, Reed AM, Aggarwal R, et al. Rituximab in the Treatment of Refractory Adult and Juvenile
568 Dermatomyositis and Adult Polymyositis: A Randomized, Placebo-phase Trial. Arthritis Rheum.
569 2013;65(2):314-324. doi:10.1002/art.37754

PT
570 52. Dinh HV, McCormack C, Hall S, Prince HM. Rituximab for the treatment of the skin manifestations
571 of dermatomyositis: a report of 3 cases. J Am Acad Dermatol. 2007;56(1):148-153.
572 doi:10.1016/j.jaad.2006.05.068

RI
573 53. de Souza FHC, Miossi R, de Moraes JCB, Bonfá E, Shinjo SK. Favorable rituximab response in
574 patients with refractory idiopathic inflammatory myopathies. Adv Rheumatol Lond Engl.

SC
575 2018;58(1):31. doi:10.1186/s42358-018-0030-z

576 54. So H, Wong VTL, Lao VWN, Pang HT, Yip RML. Rituximab for refractory rapidly progressive
577 interstitial lung disease related to anti-MDA5 antibody-positive amyopathic dermatomyositis. Clin

U
578 Rheumatol. 2018;37(7):1983-1989. doi:10.1007/s10067-018-4122-2
AN
579 55. Vargas Lebrón C, Ruiz Montesino MD, Moreira Navarrete V, Toyos Sainz de Miera FJ. Treatment
580 With Rituximab in Juvenile Dermatomyositis: Effect on Calcinosis. Reumatol Clin. July 2018.
581 doi:10.1016/j.reuma.2018.06.010
M

582 56. Aggarwal R, Loganathan P, Koontz D, Qi Z, Reed AM, Oddis CV. Cutaneous improvement in
583 refractory adult and juvenile dermatomyositis after treatment with rituximab. Rheumatol Oxf Engl.
584 2017;56(2):247-254. doi:10.1093/rheumatology/kew396
D

585 57. Argobi Y, Smith GP. Tracking changes nailfold capillary abnormalities in Dermatomyositis on
TE

586 treatment. J Am Acad Dermatol. February 2019. doi:10.1016/j.jaad.2019.02.027

587 58. Albers LN, Liu Y, Bo N, Swerlick RA, Feldman RJ. Developing biomarkers for predicting clinical
588 relapse in pemphigus patients treated with rituximab. J Am Acad Dermatol. 2017;77(6):1074-1082.
EP

589 doi:10.1016/j.jaad.2017.07.012

590 59. Galimberti F, Kooistra L, Li Y, Chatterjee S, Fernandez AP. Intravenous immunoglobulin is an


591 effective treatment for refractory cutaneous dermatomyositis. Clin Exp Dermatol. 2018;43(8):906-
C

592 912. doi:10.1111/ced.13607


AC

593 60. Anh-Tu Hoa S, Hudson M. Critical review of the role of intravenous immunoglobulins in idiopathic
594 inflammatory myopathies. Semin Arthritis Rheum. 2017;46(4):488-508.
595 doi:10.1016/j.semarthrit.2016.07.014

596 61. Danieli MG, Calcabrini L, Calabrese V, Marchetti A, Logullo F, Gabrielli A. Intravenous
597 immunoglobulin as add on treatment with mycophenolate mofetil in severe myositis. Autoimmun
598 Rev. 2009;9(2):124-127. doi:10.1016/j.autrev.2009.04.003

599 62. de Inocencio J, Enríquez-Merayo E, Casado R, González-Granado LI. Subcutaneous Immunoglobulin


600 in Refractory Juvenile Dermatomyositis. Pediatrics. 2016;137(4). doi:10.1542/peds.2015-3537
ACCEPTED MANUSCRIPT
28

601 63. Kotani T, Makino S, Takeuchi T, et al. Early intervention with corticosteroids and cyclosporin A and
602 2-hour postdose blood concentration monitoring improves the prognosis of acute/subacute
603 interstitial pneumonia in dermatomyositis. J Rheumatol. 2008;35(2):254-259.

604 64. Shimojima Y, Ishii W, Matsuda M, Kishida D, Ikeda S-I. Effective Use of Calcineurin Inhibitor in
605 Combination Therapy for Interstitial Lung Disease in Patients With Dermatomyositis and
606 Polymyositis. J Clin Rheumatol Pract Rep Rheum Musculoskelet Dis. 2017;23(2):87-93.

PT
607 doi:10.1097/RHU.0000000000000487

608 65. Schiffenbauer A, Garg M, Castro C, et al. A randomized, double-blind, placebo-controlled trial of
609 infliximab in refractory polymyositis and dermatomyositis. Semin Arthritis Rheum. 2018;47(6):858-

RI
610 864. doi:10.1016/j.semarthrit.2017.10.010

611 66. Bunch TW, Worthington JW, Combs JJ, Ilstrup DM, Engel AG. Azathioprine with prednisone for

SC
612 polymyositis. A controlled, clinical trial. Ann Intern Med. 1980;92(3):365-369.

613 67. Deakin CT, Campanilho-Marques R, Simou S, et al. Efficacy and Safety of Cyclophosphamide
614 Treatment in Severe Juvenile Dermatomyositis Shown by Marginal Structural Modeling. Arthritis

U
615 Rheumatol Hoboken NJ. 2018;70(5):785-793. doi:10.1002/art.40418
AN
616 68. Clottu A, Laffitte E, Prins C, Chizzolini C. Response of mucocutaneous lesions to rituximab in a case
617 of melanoma differentiation antigen 5-related dermatomyositis. Dermatol Basel Switz.
618 2012;225(4):376-380. doi:10.1159/000346573
M

619 69. Combalia A, Giavedoni P, Tamez L, Grau-Junyent JM, Mascaró JM. Bosentan for Cutaneous Ulcers
620 in Anti-MDA5 Dermatomyositis. JAMA Dermatol. 2018;154(3):371-373.
621 doi:10.1001/jamadermatol.2017.5462
D

622 70. Schanz S, Ulmer A, Fierlbeck G. Response of dystrophic calcification to intravenous


TE

623 immunoglobulin. Arch Dermatol. 2008;144(5):585-587. doi:10.1001/archderm.144.5.585

624 71. Shahani L. Refractory calcinosis in a patient with dermatomyositis: response to intravenous
625 immune globulin. BMJ Case Rep. 2012;2012. doi:10.1136/bcr-2012-006629
EP

626 72. Reiter N, El-Shabrawi L, Leinweber B, Berghold A, Aberer E. Calcinosis cutis: part II. Treatment
627 options. J Am Acad Dermatol. 2011;65(1):15-22; quiz 23-24. doi:10.1016/j.jaad.2010.08.039
C

628 73. Huber AM, Kim S, Reed AM, et al. Childhood Arthritis and Rheumatology Research Alliance
629 Consensus Clinical Treatment Plans for Juvenile Dermatomyositis with Persistent Skin Rash. J
AC

630 Rheumatol. 2017;44(1):110-116. doi:10.3899/jrheum.160688

631 74. Spencer CH, Rouster-Stevens K, Gewanter H, et al. Biologic therapies for refractory juvenile
632 dermatomyositis: five years of experience of the Childhood Arthritis and Rheumatology Research
633 Alliance in North America. Pediatr Rheumatol Online J. 2017;15(1):50. doi:10.1186/s12969-017-
634 0174-0

635 75. Enders FB, Bader-Meunier B, Baildam E, et al. Consensus-based recommendations for the
636 management of juvenile dermatomyositis. Ann Rheum Dis. 2017;76(2):329-340.
637 doi:10.1136/annrheumdis-2016-209247
ACCEPTED MANUSCRIPT
29

638 76. Papadopoulou C, Hong Y, Omoyinmi E, Brogan PA, Eleftheriou D. Janus kinase 1/2 inhibition with
639 baricitinib in the treatment of juvenile dermatomyositis. Brain J Neurol. February 2019.
640 doi:10.1093/brain/awz005

641 77. Ladislau L, Suárez-Calvet X, Toquet S, et al. JAK inhibitor improves type I interferon induced
642 damage: proof of concept in dermatomyositis. Brain J Neurol. 2018;141(6):1609-1621.
643 doi:10.1093/brain/awy105

PT
644 78. Kurtzman DJB, Wright NA, Lin J, et al. Tofacitinib Citrate for Refractory Cutaneous
645 Dermatomyositis: An Alternative Treatment. JAMA Dermatol. 2016;152(8):944-945.
646 doi:10.1001/jamadermatol.2016.0866

RI
647 79. Kato M, Ikeda K, Kageyama T, et al. Successful Treatment for Refractory Interstitial Lung Disease
648 and Pneumomediastinum With Multidisciplinary Therapy Including Tofacitinib in a Patient With

SC
649 Anti-MDA5 Antibody-Positive Dermatomyositis. J Clin Rheumatol Pract Rep Rheum Musculoskelet
650 Dis. January 2019. doi:10.1097/RHU.0000000000000984

651 80. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med.

U
652 1975;292(7):344-347. doi:10.1056/NEJM197502132920706
AN
653 81. Bohan A, Peter JB. Polymyositis and dermatomyositis (second of two parts). N Engl J Med.
654 1975;292(8):403-407. doi:10.1056/NEJM197502202920807

655 82. Lundberg IE, Tjärnlund A, Bottai M, et al. 2017 European League Against Rheumatism/American
M

656 College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory
657 myopathies and their major subgroups. Ann Rheum Dis. 2017;76(12):1955-1964.
658 doi:10.1136/annrheumdis-2017-211468
D

659 83. Suzuki S, Uruha A, Suzuki N, Nishino I. Integrated Diagnosis Project for Inflammatory Myopathies:
TE

660 An association between autoantibodies and muscle pathology. Autoimmun Rev. 2017;16(7):693-
661 700. doi:10.1016/j.autrev.2017.05.003

662 84. Tanimoto K, Nakano K, Kano S, et al. Classification criteria for polymyositis and dermatomyositis. J
EP

663 Rheumatol. 1995;22(4):668-674.

664 85. Targoff IN, Miller FW, Medsger TA, Oddis CV. Classification criteria for the idiopathic inflammatory
665 myopathies. Curr Opin Rheumatol. 1997;9(6):527-535.
C

666 86. Dalakas MC, Hohlfeld R. Polymyositis and dermatomyositis. Lancet Lond Engl. 2003;362(9388):971-
AC

667 982. doi:10.1016/S0140-6736(03)14368-1

668 87. Hoogendijk JE, Amato AA, Lecky BR, et al. 119th ENMC international workshop: Trial design in
669 adult idiopathic inflammatory myopathies, with the exception of inclusion body myositis, 10–12
670 October 2003, Naarden, The Netherlands. Neuromuscul Disord. 2004;14(5):337-345.
671 doi:10.1016/j.nmd.2004.02.006

672 88. Mariampillai K, Granger B, Amelin D, et al. Development of a New Classification System for
673 Idiopathic Inflammatory Myopathies Based on Clinical Manifestations and Myositis-Specific
674 Autoantibodies. JAMA Neurol. 2018;75(12):1528-1537. doi:10.1001/jamaneurol.2018.2598
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP
C
AC

You might also like