You are on page 1of 12

C o n n e c t i v e Ti s s u e D i s e a s e

Current Concepts
Anthony P. Fernandez, MD, PhD

KEYWORDS
 Connective tissue disease  Dermatomyositis  Cutaneous lupus erythematosus  Morphea
 Cutaneous vasculitis

KEY POINTS
 Classification criteria for dermatomyositis that includes amyopathic patients has recently been
published and several medications effective for patients with refractory cutaneous dermatomyositis
have been described.
 Progress in objectively defining cutaneous lupus erythematosus, updates in pathogenesis, and
studies optimizing antimalarial use in patients with cutaneous lupus erythematosus have occurred
in recent years.
 There have been updates in clinical features of patients with morphea, in addition to the identifica-
tion of several potential biomarkers of disease activity and novel treatments.
 Clarification of categories applicable to skin vasculitis have recently been described and clinical
studies that aim to better understand pathogenesis and identify effective treatments are ongoing.

INTRODUCTION A number of important updates concerning DM


have occurred in recent years that are particularly
Connective tissue diseases can affect numerous pertinent to dermatologists who treat these pa-
organ systems and often prominently affect the tients. Arguably, the most important is the release
skin. Thus, dermatologists frequently play an of updated idiopathic inflammatory myopathy (IIM)
important role in the diagnosis and management classification guidelines by the European League
of these conditions. Herein is an update of our un- Against Rheumatism/American College of Rheu-
derstanding of various aspects of common con- matology (EULAR/ACR) that help accurately
nective tissue diseases, with many contributions distinguish DM from other IIMs.1 Unlike the classic
resulting from research performed by dermatolo- criteria published by Bohan and Peter,2,3 the new
gists with expertise in these diseases. EULAR/ACR classification criteria included pa-
tients with clinically amyopathic DM (CADM), as
well as comparators with other dermatologic dis-
DERMATOMYOSITIS
ease, in their study population. The EULAR/ACR
Dermatomyositis (DM) is an autoimmune disease classification criteria take into account several
that most commonly affects the skin and muscles. cutaneous manifestations of DM, including

Disclosure Statement: Dr A.P. Fernandez is a Principal Investigator and receives funding from Mallinckrodt (Ef-
ficacy and Safety of H.P. Acthar gel for the treatment of refractory cutaneous manifestations of dermatomy-
ositis; ClinicalTrials.gov identifier NCT02245841) and Pfizer (A Phase 2a, Double-blind, Randomized, Placebo-
controlled Study To Evaluate The Efficacy, Safety, And Tolerability Of Pf-06823859 In Adult Subjects With Der-
matomyositis; ClinicalTrials.gov identifier NCT03181893) for dermatomyositis research and is a consultant for
derm.theclinics.com

Celgene. He also is an investigator for the ARAMIS (ClinicalTrials.gov identifier NCT02939573) and CUTIS
(ClinicalTrials.gov identifier NCT03004326) studies.
Departments of Dermatology and Pathology, Cleveland Clinic Lerner College of Medicine, 9500 Euclid Avenue
A61, Cleveland, OH 44195, USA
E-mail address: fernana6@ccf.org

Dermatol Clin 37 (2019) 37–48


https://doi.org/10.1016/j.det.2018.07.006
0733-8635/19/Ó 2018 Elsevier Inc. All rights reserved.
38 Fernandez

Gottron’s papules, Gottron’s sign, and heliotrope systemic therapy treatment regimens. Thus, a sig-
rash. By using the EULAR/ACR classification nificant percentage of DM adult patients, too, are
criteria, patients with these pathognomonic skin refractory to standard-of-care therapies. Overall,
manifestations of juvenile and adult DM were these data reveal that DM cutaneous disease, in
accurately classified without including muscle general, can often be refractory to treatment and
biopsy data. For DM patients without muscle support there is an unmet need for development
involvement, the new criteria recommend a skin of more effective cutaneous DM treatments.
biopsy is performed to further support a DM Luckily, during the past few years research has
diagnosis. identified several medications with promising effi-
The EULAR/ACR idiopathic inflammatory myop- cacy in treating refractory cutaneous DM that
athy classification criteria represent a tremendous may eventually be welcome additions to routine
step forward for dermatologists who see patients therapy armamentariums. Two Janus kinase inhib-
with DM for several reasons. Not only are specific itors, ruxolitinib and tofacitinib, have recently been
skin manifestations recognized as important fea- reported to effectively treat DM patients, all of
tures of DM that can be used for accurate classifi- whom had refractory cutaneous disease despite
cation, but patients with CADM are now formally exposure to between 4 and 9 systemic medica-
accepted as having DM and can theoretically tions.11,12 Although 3 patients were reported to
qualify for clinical trials that may lead to improved respond well to tofacitinib 5 to 10 mg twice daily
treatment options for this subset of patients. With in one of the reports, the authors now have accu-
that being said, there is still room for improvement mulated a cohort of at least 9 patients with refrac-
in DM classification criteria from a dermatologic tory cutaneous DM who are responding well to
standpoint. In a recently published retrospective tofacitinib (Dr Ruth Ann Vleugals, MD, as pre-
study of a single-center DM cohort, 26.3% of pa- sented at the 4th International Cutaneous Lupus
tients with CADM still would not meet the sug- and Dermatomyositis meeting; Orlando, FL; per-
gested 55% minimum probability cutoff to be sonal communication, 2018).
classified as having DM based on the EULAR/ Adrenocorticotropic hormone gel (repository
ACR criteria.4 The authors of this study propose corticotropin hormone [RCI]) has been approved
inclusion of other cutaneous-specific criteria, by the US Food and Drug Administration to treat
such as skin biopsy findings, may improve classi- DM and other immune-mediated diseases since
fication criteria to include a higher percentage of 1952. However, there has been a paucity of data
CADM patients. concerning efficacy and safety of this treatment
Control of cutaneous DM disease activity is for DM. A recent open-label trial reported RCI
important for numerous reasons. Cutaneous DM treatment was both safe and efficacious for
disease is associated with physical symptoms DM.13 Although the focus was on control of
(pain, pruritus), infection risk, and adverse psycho- myositis, the authors did report 4 of the 5 DM
logical effects, all of which contribute to a signifi- patients (80%) with significant skin disease
cant negative impact on patients’ quality of responded positively to RCI treatment, showing
life.5,6 Furthermore, persistent cutaneous DM can an 88% (83.3%–100%) improvement in cutaneous
lead to permanent damage in the form of scarring, visual analog scale score. Our group is conducting
calcinosis, decreased range of motion, and/or an open-label study focusing on safety and effi-
lipoatrophy. cacy of RCI for refractory cutaneous DM dis-
It has long been suspected by some that cuta- ease.14 Interim results for 9 patients enrolled thus
neous DM manifestations often persist despite far are similar to what was seen by Aggarwal and
aggressive treatment and myositis resolution. colleagues.13 Improvement in skin disease activity
Studies in patients with juvenile DM have found as measured by the Cutaneous Dermatomyositis
up to 60% have persistent cutaneous disease Activity and Severity Index (CDASI) has been
and the percentage with persistent cutaneous dis- seen in 7 of the 9 patients 3 months after initiating
ease is often more than double the percentage RCI, and in all 7 patients who have so far
with persistent myositis.7–9 However, studies in completed 6 months of the study (Fig. 1). Addition-
adults with DM suggesting persistent cutaneous ally, there has been improvement in numerous
disease activity have been lacking. Recently, a patient-reported outcomes, including the Global
cohort of adult DM patients was prospectively Itch Score and Dermatology Life Quality Index
studied to determine the percentage who scores.
achieved clinical remission of their cutaneous dis- Another promising medication for cutaneous DM
ease over a 3-year follow-up period.10 The authors is lenabasum (anabasum), a synthetic, oral, prefer-
found only 28 of 74 patients (38%) achieved clin- ential cannabinoid 2 receptor agonist. Lenabasum
ical remission over 3 years despite aggressive is nonimmunosuppressive and results in reduction
Connective Tissue Disease 39

Fig. 1. (A) 61-year-old woman with classic dermatomyositis and TIF1-g autoantibodies with refractory cutaneous
disease activity after failing 6 systemic treatments, including multidrug regimens. (B) There is significant improve-
ment in her rash 3 months after treatment with repository corticotropin gel (80 U twice weekly) in the setting of
an open-label clinical trial (ClinicalTrials.gov identifier NCT02245841). (From Fernandez A. Efficacy and Safety of
H.P. Acthar Gel for the Treatment of Refractory Cutaneous Manifestations of Dermatomyositis (Acthar Gel) -
ClinicalTrials.gov identifier: NCT02245841. Available at: https://clinicaltrials.gov/ct2/show/NCT02245841; with
permission.)

of peripheral blood mononuclear cell cytokine pro- IFN-a, correlates best with DM cutaneous dis-
duction in DM patients, in addition to inducing res- ease activity.18–20 These findings obviously
olution of innate immune responses.15 In a double- have potential treatment implications. Sifalimu-
blind, randomized, placebo-controlled phase II mab, a monoclonal antibody against IFN-a, has
trial, lenabasum provided statistically significant been studied as a DM treatment, but its develop-
improvement in CDASI activity scores and other ment has been terminated by MedImmune,
secondary efficacy outcomes and displayed excel- LLC.21,22 Thus, it may be that targeting IFN-b is
lent tolerability.16 An open-label extension of this a more effective strategy for controlling DM
trial supports lenabasum has persistent efficacy, than targeting IFN- a. Clinical investigations con-
safety and tolerability in patients with refractory cerning this strategy may be available in the near
skin-predominant DM.17 future.
One of the most potentially critical findings Finally, dermatologists know well that scalp pru-
concerning cutaneous DM relates to the ritus can be particularly disturbing for DM patients
associated inflammatory cytokine profile. DM is and refractory to treatment. A recent study
known to be a type I interferon (IFN)-mediated revealed the mean epidermal nerve fiber density
disease, and theoretically could be primarily was lower in DM patients with refractory scalp pru-
driven by IFN-a, IFN-b, or other type I IFNs. ritus compared with normal control patients in
However, it has recently been shown IFN-b, not posterior parietal and occipital scalp areas,
40 Fernandez

suggesting the possibility of a scalp neuropathy in vacuolar interface dermatitis and further keratino-
the DM patients.23 Additionally, it was just shown cyte death. This process propagates the release
IL-31 levels correlate highly with pruritus and cuta- of more eNA motifs to fuel and sustain chronic
neous disease activity in DM patients, and itch inflammation and development of chronic CLE le-
improvement correlated with significant reduc- sions. eNAs from dying cells seem to induce kera-
tions in IL-31 in DM lesional skin.24 Whether spe- tinocyte production of proinflammatory cytokines,
cifically targeting IL-31 in refractory patients is an in particular, through activation of pathogen
effective strategy for DM remains to be seen, but recognition receptor pathways via nucleic acids.29
appears promising. LL-37 (cathelicidin), which is expressed in CLE le-
sions, may act as a transfectant to introduce eNAs
CUTANEOUS LUPUS ERYTHEMATOSUS into keratinocytes and help to trigger this inflam-
matory cascade.
Cutaneous involvement of lupus erythematosus Another study suggests keratinocytes may be
(LE) can occur as the sole manifestation of dis- inherently primed by IFN-k to produce abundant
ease, but also represents the second most com- amounts of IL-6 in the setting of CLE.31 Thus, a
mon organ involved in systemic LE (SLE).25 predisposition for brisk type I IFN inflammatory re-
Although most dermatologists are familiar with sponses by lupus keratinocytes upon stimulation
the subtypes of cutaneous LE (CLE), there is a may exist owing to an autocrine loop involving
consensus that it remains an ill-defined disease. IFN-k. This may be a mechanism by which envi-
Classification criteria that more objectively defines ronmental factors such as UV exposure trigger
CLE and helps clinicians to distinguish CLE from robust inflammatory responses and release of
other mimics is lacking, and this shortcoming is eNA leading to CLE development.
likely impeding both optimal clinical care and Currently, antimalarials are considered first-line
ability to adequately conduct clinical studies. systemic treatment for CLE. There are 3 antimalar-
Recently, there has been significant activity to ials commonly used to treat CLE: hydroxychloro-
address these inadequacies, first focusing on quine (HCQ), chloroquine (CQ), and quinacrine.
discoid LE. A Delphi consensus exercise per- Quinacrine may be underused owing to its lack
formed by a group of CLE experts determined 12 of availability. However, although quinacrine is
clinical and histologic criteria for potential inclu- currently not manufactured by any company, it
sion into discoid LE classification criteria.26 can be compounded into capsules readily by
Currently, studies to assess these 12 clinical and most compounding pharmacies. Quinacrine has
histologic criteria in discoid LE versus other mim- the advantage of not being associated with retinal
icker diseases is underway. In the future, similar toxicity, unlike HCQ and CQ. Studies have also
studies to develop criteria for other CLE subtypes suggested quinacrine may be a stronger antiin-
may be performed. flammatory agent than HCQ or CQ, and recently
There has also been significant progress in bet- it was shown to more effectively suppress both tu-
ter understanding the pathogenesis of CLE. The mor necrosis factor-a and IFN-a in peripheral
type I IFN inflammatory environment in CLE is blood mononuclear cells from patients with DM
known to be driven by both plasmacytoid den- and CLE.32,33
dritic cells and keratinocytes.27 Additionally, it is Importantly, multiple studies have been per-
thought the accumulation of apoptotic cells is formed recently that can potentially be translated
fundamental to both SLE and CLE pathogen- into safer and more effective use of antimalarials
esis.28 Recent evidence suggests, in a back- for CLE in everyday clinical practice. A major up-
ground of lupus immunologic dysregulation, that date concerns dosing and retinal toxicity
UV irradiation can contribute to pathogenic accu- screening for HCQ. Based on a cohort of 2361 pa-
mulation of apoptotic cells in the skin.29 This tients who had been exposed to antimalarials, it
accumulation of apoptotic material, coupled with was found HCQ doses of 5.0 mg/kg actual
impaired ability to clear cellular debris, leads to body weight were associated with significantly
secondary necrosis of apoptotic cells and the less retinal toxicity risk compared with higher
release of large quantities of endogenous nucleic doses.34 This study triggered new recommenda-
acids (eNA).30 tions from the American Academy of Ophthal-
Strong evidence that eNA may play a patho- mology to limit HCQ dosing to 5 mg/kg actual
genic role in chronic CLE inflammation was also body weight.35 This dosing is similar to previous
demonstrated.29 eNA can strongly induce kerati- recommendations (<6.5 mg/kg ideal body weight)
nocytes to release CXCL10 and IFN-regulated for many patients, but seems to be most important
cytokines. CXCL10 expression then drives recruit- for very thin patients who otherwise may have
ment of CXCR31 cytotoxic immune cells to induce taken doses placing them at high retinal toxicity
Connective Tissue Disease 41

risk. These new recommendations are controver- correlated with response in patients with CLE.40
sial for obese patients, however. This finding raises the obvious question concern-
Additionally, the American Academy of Ophthal- ing whether clinicians should simply dose HCQ
mology recommends, following a baseline exami- to reach adequate whole blood levels on an indi-
nation for retinal toxicity within the first 12 months vidual basis. Chasset and colleagues41 recently
of treatment, that a repeat screening examination explored this possibility. They increased daily
is not required until a patient has been taking HCQ dose (maximum 800 mg/d) in 32 patients
HCQ for 5 years, assuming the patient has normal with refractory CLE until HCQ blood concentra-
retinal toxicity risk.35 This recommendation is tions were greater than 750 ng/mL. With this strat-
based on research supporting cumulative HCQ egy, 26 patients (81%) reached the primary
dose is the most important risk factor for retinal endpoint of significant improvement in Cutaneous
toxicity, and most patients who have developed Lupus Activity and Severity Index activity. Howev-
retinal toxicity have taken cumulative doses of er, the median daily HCQ dose required to achieve
1000 g or more, which would occur if a patient blood levels of greater than 750 ng/mL was 9.8
takes 400 mg/d for 7 years. Although these guide- mg/kg actual body weight (range, 6.8–13.8 mg/
lines are also not without controversy, it is impor- kg actual body weight) and 9.2 mg/kg ideal body
tant for clinicians who prescribe antimalarials to weight (range, 5.7–14.5 mg/kg ideal body weight),
be knowledgeable about them. which is significantly higher than current or past
Despite being well-recognized as first-line recommended safe dosing. Although many
treatment, the first double-blind, randomized trial patients were able to eventually decrease dosing
comparing HCQ with placebo in CLE was only to 400 mg/d, the authors note that this strategy
recently reported.36 The primary endpoint of may be preferable only in patients who
decrease in Cutaneous Lupus Activity and have received a cumulative HCQ dose of less
Severity Index after 16 weeks was met, but did than 1000 g and that it is probably important to
not significantly differ in the HCQ versus placebo limit the time during which increased doses are
groups. However, Physician Global Assessment, used.
pain, and fatigue scores did significantly Despite these useful updates, a percentage of
improve in the HCQ group compared with the patients with CLE will not respond adequately to
placebo group at week 16, supporting the effec- antimalarials. In this case, more traditional immu-
tiveness of HCQ for CLE and its first-line treat- nosuppressive medications should be considered.
ment status. A recent study looking at effective treatments in
Several studies concerning optimizing use of patients with CLE who failed antimalarials found
antimalarials for CLE are worth noting. In 1 study, thalidomide to be particularly effective compared
researchers examined usefulness of switching an- with other options.42 Lenalidamide is a derivative
timalarials after patients either (1) failed to respond of thalidomide that has also been shown to be
or (2) had an adverse reaction to a first antima- effective for CLE.43,44 Lenalidamide attractively
larial.37 They found >50% of patients responded lacks an association with peripheral neuropathy,
to a second antimalarial after failing the first, which occurs in approximately 16% of CLE pa-
although the response seemed to decrease with tients who take thalidomide for extended periods
time. Additionally, 69% of patients exposed to a of time.45
second antimalarial agent after having an adverse There is an abundance of activity regarding a
reaction to the first tolerated it without incident. search for more effective CLE treatments. A novel
Furthermore, 80% of the patients who tolerated thalidomide-like medication has recently been
the second antimalarial responded positively to shown to more effectively decrease Aiolos, a pro-
it. Additional studies suggest adding quinacrine tein implicated in promoting LE disease activity,
to either HCQ or CQ can be a useful strategy for levels than either thalidomide or lenalidamide.46
up to two-thirds of patients who fail either medica- Results of a phase IIa study suggest promising ef-
tion as monotherapy.38,39 Adding quinacrine to ficacy in patients with SLE, and a phase IIb study is
either HCQ or CQ seems not only to be effective, underway in a larger SLE patient population.47,48
but also safe. Thus, switching antimalarials and/ Because skin activity improved in patients enrolled
or adding quinacrine before exposing patients in the phase IIa study, specifically examining effi-
with CLE to more toxic immunosuppressive med- cacy and safety of this medication for CLE should
ications seems to be an important treatment be considered.
consideration. Although a phase I study lacked impressive re-
There continues to be mounting evidence sug- sults, there was improvement in a number of
gesting measurement of whole blood HCQ levels endpoints, including Cutaneous Lupus Activity
can be clinically important, because they are and Severity Index scores, in a phase IIb study
42 Fernandez

evaluating treatment with the anti–IFN-a mono- MORPHEA


clonal antibody, sifalimumab, in patients with
SLE.49,50 However, more promising improve- There are numerous fibrosing skin diseases
ments in CLE were seen in a recent phase IIb treated by dermatologists. Most are challenging
clinical trial assessing efficacy of anifrolumab, to treat and may have devastating negative effects
an anti-type I IFN receptor monoclonal antibody, on patients’ quality of life. Thus, there is a great
in patients with SLE.51 In light of this finding, need for a better understanding of pathogenesis
sifalimumab production has been discontinued and better treatments for this group of conditions.
and immediate future studies will focus on Given the limitations of this review, here we focus
anifrolumab in LE.22 Additional ongoing clinical on updates concerning localized scleroderma, or
trials for novel CLE treatments are listed in morphea.
Table 1. Numerous advancements in the clinical aspects
of morphea have occurred in the past few years.

Table 1
Survey of current clinical trials focused on treatment of cutaneous lupus erythematosus

ClinicalTrials.Gov
Study Title Intervention Identifier
Treatment of Cutaneous Lupus 595 nm flashlamp pulsed dye laser NCT00523588
Erythematosus (CLE) With the 595 nm
Flashlamp Pulsed Dye Laser
Safety and Efficacy of Filgotinib and JAK 1 inhibitor NCT03134222
GS-9876 in Females With Moderately-
to-Severely Active Cutaneous Lupus
Erythematosus (CLE)
To Evaluate the Preliminary Safety, Phosphodiesterase-4 inhibitor NCT01300208
Tolerability, Pharmacokinetics,
Pharmacodynamics and Efficacy of
CC-11050 in Subjects With Discoid
Lupus Erythematosus and Subacute
Cutaneous Lupus Erythematosus
Low-dose UVA1 Radiation in Cutaneous UVA-1 phototherapy 20 J/cm2 3 times NCT01776190
Lupus Patients per week
IVIg Efficacy Study to Treat Lupus 2 g/kg/mo IVIg NCT01841619
Erythematosus
Study to Evaluate BIIB059 in Cutaneous Monoclonal antibody targeting blood NCT02847598
Lupus Erythematosus (CLE) With dendritic cell antigen 2 (BDCA2)
or Without Systemic Lupus
Erythematosus (SLE)
Safety and Efficacy of KRP203 in Sphingosine 1 phosphate receptor NCT01294774
Subacute Cutaneous Lupus agonist
Erythematosus
Safety, Tolerability, Pharmacokinetics, Topical SYK inhibitor NCT02927457
Pharmacodynamics and Clinical Effect
of GSK2646264 in Cutaneous Lupus
Erythematosus Patients
Open-label Study of Tofacitinib for JAK1/2/3 inhibitor NCT03288324
Moderate to Severe Skin Involvement
in Young Adults With Lupus
Safety and Efficacy of Topical R333 in Topical dermatologic JAK/SYK inhibitor NCT01597050
Patients With Discoid Lupus
Erythematosus (DLE) and Systemic
Lupus Erythematosus (SLE) Lesions

Select Clinical Trials listed as Active or Completed on ClinicalTrials.Gov with search terms “cutaneous lupus” on June 27,
2018.
Abbreviations: IVIg, intravenous immunoglobulin; JAK, Janus kinase; SYK, spleen tyrosine kinase.
Connective Tissue Disease 43

Generalized morphea is a relatively common mor- IFN signature in the peripheral blood. This finding
phea subtype and one of the most severe. Howev- differs from that seen in systemic sclerosis (SSc),
er, the criteria for diagnosing generalized morphea suggesting skin fibrosis in these diseases may
differ in various classification schemes, most of result from different processes.
which are not based on well-studied prospective These findings are particularly interesting
cohorts.52,53 This variability can impede clinical because it has been noted some patients with
care and research, akin to CLE described earlier. SSc develop classic morphea lesions.58 Although
Recently, Teske and colleagues54 used computer- the percentage of patients with SSc who devel-
ized lesion mapping to objectively determine the oped morphea lesions was low (12 of 370
cutaneous distribution of lesions in patients with [3.2%]), it is notable some developed morphea
generalized morphea. Their analysis suggests (especially generalized morphea) before SSc diag-
there are 3 groups of generalized morphea pa- nosis. Also of note, Raynaud’s phenomenon and
tients with distinct clinical and demographic fea- nailfold capillary changes were universally present
tures. They propose patients with generalized in these patients with SSc, suggesting clinical fea-
morphea be categorized as having either isomor- tures can help to distinguish between these dis-
phic or symmetric subtypes and that patients eases. Another study examined anatomic
with multiple linear lesions be excluded from distribution of pediatric-onset morphea lesions.59
generalized morphea classification. This classifi- This study revealed morphea disproportionately
cation could allow for more homogenous popula- affects the hands, feet, extensor extremities, pos-
tions to better study the genetics, pathogenesis, terior trunk, and upper face. Because these areas
and treatment of this heterogenous disease. may be more prone to trauma, the findings support
Useful biomarkers and other clues predicting the theory that trauma is an etiologic factor in mor-
morphea clinical activity are also lacking. It has phea pathogenesis.
recently been demonstrated patients with mor- Although classically thought to be a disease iso-
phea have increased myelin basic protein (MBP) lated to skin and occasionally involving the subcu-
serum levels and anti-MBP antibodies, and those tis and/or fascia, a recent study explored the
with anti-MBP antibodies have increased levels of association between morphea and inflammatory
pain and histologic perineural inflammation in arthritis.60 The authors found 11 of 53 pediatric
skin.55 This finding suggests anti-MBP antibodies morphea patients (21%) had polyarthritis, and
to be a potentially useful biomarker for severe involved joints were unrelated to sites of morphea.
morphea and a target for mechanistic studies, This finding is in line with a previous study of 750
but additional research is needed. Another study pediatric morphea patients in which 22% of pa-
systematically evaluated the histologic features tients had extracutaneous manifestations, with
in morphea lesional skin biopsies and their rela- articular disease being the most common.61 The
tionship with clinical features.56 It was found joints most commonly affected were the proximal
bottom-heavy patterns of sclerosis and severe interphalangeal and metacarpophalangeal joints
inflammation both were associated with patient of the fingers (10 of 11 patients).60 Antinuclear anti-
perceived pain and tightness. This finding sug- body positivity was significantly higher in patients
gests value in describing detailed histologic fea- with arthritis (8 of 10) compared with patients
tures in morphea pathology reports and implies without arthritis (8 of 21).
such patients with these histologic features Finally, there have been few studies evaluating
may require more aggressive monitoring and how morphea affects health-related quality of life
treatment. and these have yielded conflicting results. A more
Other potential biomarkers were identified recent study in adults with morphea showed it
recently by evaluating cytokine levels in whole significantly impaired health-related quality of
blood and morphea lesional skin and comparing life, most prominently in terms of emotional
these with healthy control subjects.57 CXCL9 well-being and concerns the disease would prog-
was found to be significantly elevated in morphea ress to internal organs.62 This finding has impor-
serum and levels correlated with disease activity tance for providers who care for morphea
severity. Double-staining immunohistochemistry patients, especially in terms of evaluation and
revealed CXCL9 colocalized with CD681 dermal education.
macrophages, implying from a pathogenesis Treatment of morphea is not well-defined and a
standpoint that skin may be the source of circu- recent survey study suggests there is large varia-
lating cytokines. CXCL10, in contrast, may be a tion in how pediatric dermatologists treat pediatric
marker of overall sclerosis severity. Furthermore, morphea.63 In another survey, dermatologists
morphea patient serum revealed evidence of Th1 generally reported preference for topical treat-
IFN-g imbalances, but absence of a transcriptional ments, whereas rheumatologists reported
44 Fernandez

preference for using systemic immunosuppres- Nomenclature of Vasculitidies (CHCC2012) reflected


sives.64 In this latter survey, however, pediatric updates in the understanding of vasculitis relative to
dermatologists were more closely aligned with pe- the previous conference in 1994.74 However,
diatric rheumatologists in their use of systemic CHCC2012 did not address skin-limited or skin-
medications. Studies have also shown outcomes predominant forms of vasculitis. In light of this defi-
differ depending on timing of morphea onset. ciency, an expert consensus group convened to pro-
Although many patients with pediatric-onset mor- pose an addendum to CHCC2012 that focused on
phea will experience disease activity into adult- cutaneous vasculitidies.75 This addendum, which
hood, patients with morphea onset in adulthood importantly included many dermatologists, helps to
may have increased disease severity.65 clarify CHCC2012 categories applicable to skin
Topical and systemic glucocorticoids with or vasculitis and adds definitions of cutaneous vascu-
without methotrexate represent the mainstay of litis not included in the CHCC2012. This work, similar
treatment for morphea subtypes, but several to what is described in other disease states in this re-
recent reports suggest novel morphea treatments view, can help to better define patients enrolled in
may be emerging. One of these is abatacept, a re- clinical trials and other research studies to optimize
combinant fusion protein composed of an Fc re- chances of better understanding and treating this
gion of IgG1 fused to the extracellular domain of subset of vasculitis patients.
CTLA-4. There have been 2 reports published Of vasculidities predominantly affecting the
that describe a total of 5 patients with deep mor- skin, arguably the most updates have occurred
phea who were treated successfully with abata- in recent years for IgA-mediated cutaneous
cept.66,67 In one of the reports describing 3 vasculitis, also often referred to as Henoch-
patients, the mean Modified Rodnan Skin Score Schonlein purpura (HSP). The initial ACR diag-
improved dramatically from baseline by 37% at nostic criteria for IgA-mediated vasculitis did not
6 months and 74% at 18 months.66 include direct immunofluorescence (DIF) findings
In 2 other reports, a total of 7 children between of IgA deposits in cutaneous vessels.76 However,
6 and 13 years of age with morphea were treated more recent classification criteria such as
with tocilizumab, an anti–IL-6 monoclonal anti- CHCC2012 and EULAR/PRINTO/PRES 2010
body that has shown promise for treating have included this diagnostic criterion.74,77 This
SSc.68 Tocilizumab treatment resulted in signifi- update is important for the dermatology commu-
cantly improved Physician Global Assessment nity, because finding IgA deposits in cutaneous
scores, but not changes in the Modified Local- vessels has classically been a key test used to
ized Scleroderma Skin Activity Index in 1 distinguish IgA-mediated vasculitis from other
report.69 In the other report, 2 children with pan- vasculitidies in the dermatology clinic. In fact,
sclerotic morphea were treated with tocilizumab recent research by our dermatopathology group
and experienced significant decreases in Modi- suggests IgA is the most useful immunoreactant
fied Localized Scleroderma Skin Activity Index to use for DIF studies in patients with suspected
scores.70 cutaneous vasculitis.78
Finally, UVA-1 phototherapy has shown efficacy Two studies found perivascular IgM deposits in
in treating morphea and other fibrosing skin dis- lesional skin DIF specimens to be associated
eases in numerous studies.71,72 A more recent with renal involvement in adults with IgA-mediated
prospective study revealed 60% of patients with vasculitis,79,80 whereas Poterucha and col-
morphea (37 of 62 patients) were responders to leagues81 did not identify this association.
UVA-1, although 46% of responders (17 of 37 pa- Although DIF predictors of renal involvement in
tients) had disease recurrence after UVA-1.73 adult HSP patients would be useful, the discrep-
Given the relative safety of phototherapy ancy in significant findings within these studies im-
compared with most systemic medications, addi- plies no finding is yet ready to trust as a predictor
tional studies aimed at optimizing beneficial ef- in routine clinical practice. Additionally, a recent
fects of UVA-1 in morphea are needed. study suggests the presence of linear purpura
(3 cm in length) is often found in children with
CUTANEOUS VASCULITIS HSP (8 of 31 patients), suggesting this finding
may be an important clinical clue to this diag-
The skin represents one of the most frequent organs nosis82 (Fig. 2). Although interesting, future
affected by vasculitis. In patients with cutaneous studies are needed to determine if this or any
vasculitis, multiple organ systems can be involved clinical finding can reproducibly be useful for
or the vasculitic process may be isolated to predicting diagnosis of IgA-mediated cutaneous
skin (skin-limited vasculitis). The 2012 revised vasculitis/HSP or end-organ involvement other
International Chapel Hill Consensus Conference than the skin.
Connective Tissue Disease 45

Fig. 2. (A) A 7-year-old boy presenting to clinic with purpura on the legs, including several areas on the upper
legs and ankles of linear purpura (>3 cm in length). (B) A biopsy for hematoxylin and eosin staining displayed
leukocytoclastic vasculitis (LCV; not shown), and direct immunofluorescence revealed granular deposition of
IgA within superficial dermal blood vessel walls, compatible with IgA-mediated LCV/Henoch-Schonlein purpura
(original magnification 20).

There are few well-designed clinical studies that ACKNOWLEDGMENTS


have investigated efficacy of treatments for cuta-
neous vasculitis. In fact, the only randomized, The author thanks Janine Sot for her expertise in
controlled study for cutaneous vasculitis was re- preparing the figures for this article.
ported by Sais and colleagues83 in 1995 and
included 41 patients randomized to either oral REFERENCES
colchicine 0.5 mg twice daily or topical emollients.
In this study colchicine had no significant thera- 1. Lundberg IE, Tjarnlund A, Bottai M, et al. 2017 Euro-
peutic effect compared with topical emollients. pean League Against Rheumatism/American Col-
Thus, optimal treatment of cutaneous vasculitis re- lege of Rheumatology classification criteria for
mains unknown. adult and juvenile idiopathic inflammatory myopa-
There are currently 2 ongoing studies that aim to thies and their major subgroups. Arthritis Rheumatol
improve not only our understanding of cutaneous 2017;69:2271–82.
vasculitis, but also identify more optimal treatment 2. Bohan A, Peter JB. Polymyositis and dermatomyosi-
strategies. The Clinical Transcriptomics in Sys- tis (first of two parts). N Engl J Med 1975;292:344–7.
temic Vasculitis (CUTIS) study will analyze molec- 3. Bohan A, Peter JB. Polymyositis and dermatomyosi-
ular characteristics of vasculitic skin lesions tis (second of two parts). N Engl J Med 1975;292:
across a variety of vasculitidies, including sys- 403–7.
temic forms.84 Alternatively, the ARAMIS study (A 4. Patel B, Khan N, Werth VP. Applicability of EULAR/
Randomized Multicenter Study for Isolated Skin ACR classification criteria for dermatomyositis to
Vasculitis) will randomize patients with various amyopathic disease. J Am Acad Dermatol 2018;
skin-isolated, chronic vasculitic diseases to 79:77–83.e1.
receive either colchicine, azathioprine, or dapsone 5. Bailey EE, Fiorentino DF. Amyopathic dermatomyosi-
to determine which of these agents, if any, is more tis: definitions diagnosis and management. Curr
effective for treatment.85 Results of these studies Rheumatol Rep 2014;16:465.
will hopefully be available within the next few years 6. Robinson ES, Feng R, Okawa J, et al. Improvement
and provide better clinical strategies for care of in the cutaneous disease activity of patients with
patients with skin-isolated vasculitis. dermatomyositis is associated with a better quality
of life. Br J Dermatol 2015;172:169–74.
SUMMARY 7. Huber AM, Kim S, Reed AM, et al. Childhood arthritis
and rheumatology research alliance consensus clin-
This review highlights a wealth of activity aimed at ical treatment plans for juvenile dermatomyositis
better understanding and treating cutaneous dis- with persistent skin rash. J Rheumatol 2017;44:
ease associated with various connective tissue 110–6.
disorders. Thanks to the hard work and dedication 8. Sanner H, Sjaastad I, Flatø B. Disease activity and
of a growing group of dermatologist-researchers prognostic factors in juvenile dermatomyositis: a
and other professionals, the future for patients longterm follow-up study applying the Paediatric
who suffer from significant cutaneous manifesta- Rheumatology International Trials Organization
tions of these diseases looks extremely bright. criteria for inactive disease and the myositis disease
46 Fernandez

activity assessment tool. Rheumatology (Oxford) 22. Adis Insight. Sifalimumab. Available at: https://
2014;53:1578–85. adisinsight.springer.com/drugs/800024071.
9. Seshadri R, Feldman BM, Ilowite N, et al. The role of 23. Cirino P, Romiti R, McAdams B, et al. Scalp neurop-
aggressive corticosteroid therapy in patients with ju- athy in dermatomyositis patients with recalcitrant
venile dermatomyositis: a propensity score analysis. scalp pruritus. J Invest Dermatol 2018;138(5,
Arthritis Rheum 2008;59:989–95. Suppl):S74.
10. Wolstencroft PW, Chung L, Li S, et al. Factors asso- 24. Kim HJ, Bonciani D, Zeidi M, et al. Itch in dermato-
ciated with clinical remission of skin disease in der- myositis: the role of increased skin interleukin-31.
matomyositis. JAMA Dermatol 2018;154:44–51. Br J Dermatol 2018. [Epub ahead of print].
11. Hornung T, Janzen V, Heidgen FJ, et al. Remission of 25. Obermoser G, Sontheimer RD, Zelger B. Overview
recalcitrant dermatomyositis treated with ruxolitinib. of common, rare and atypical manifestations of cuta-
N Engl J Med 2014;371:2537–8. neous lupus erythematosus and histopathological
12. Kurtzman DJ, Wright NA, Lin J, et al. Tofacitinib cit- correlates. Lupus 2010;19(9):1050–70.
rate for refractory cutaneous dermatomyositis: an 26. Elman SA, Joyce C, Nyberg F, et al. Development of
alternative treatment. JAMA Dermatol 2016;152: classification criteria for discoid lupus erythemato-
944–5. sus: results of a delphi exercise. J Am Acad Derma-
13. Aggarwal R, Marder G, Koontz DC, et al. Efficacy tol 2017;77:261–7.
and safety of adrenocorticotropic hormone gel in re- 27. Wenzel J, Tuting T. An IFN-associated cytotoxic
fractory dermatomyositis and polymyositis. Ann cellular immune response against viral, self-, or tu-
Rheum Dis 2018;77:720–7. mor antigens is a common pathogenetic feature in
14. Efficacy and Safety of H.P. Acthar Gel for the Treat- “interface dermatitis”. J Invest Dermatol 2008;128:
ment of Refractory Cutaneous Manifestations of 2392e402.
Dermatomyositis (Acthar Gel). Available at: https:// 28. Kuhn A, Wenzel J, Bijl M. Lupus erythematosus re-
clinicaltrials.gov/ct2/show/NCT02245841. Accessed visited. Semin Immunopathol 2016;38:97–112.
June 27, 2018. 29. Scholtissek B, Zahn S, Maier J, et al. Immunostimu-
15. Motwani MP, Bennett F, Norris PC, et al. Potent anti- latory endogenous nucleic acids drive the lesional
inflammatory and pro-resolving effects of anabasum inflammation in cutaneous lupus erythematosus.
in a human model of self-resolving acute inflammation. J Invest Dermatol 2017;137:1484–92.
Clin Pharmacol Ther 2017. [Epub ahead of print]. 30. Mahajan A, Herrmann M, Munoz LE. Clearance defi-
16. Werth VP, Hejazi E, Pena SM, et al. A phase 2 study ciency and cell death pathways: a model for the
of safety and efficacy of anabasum (JBT-101), a pathogenesis of SLE. Front Immunol 2016;7:35.
cannabinoid receptor type 2 agonist, in refractory 31. Stannard JN, Reed TJ, Myers E, et al. Lupus skin is
skin-predominant dermatomyositis [abstract]. primed for IL-6 inflammatory responses through a
Arthritis Rheumatol 2017;69(suppl 10). keratinocytes-mediated autocrine type I interferon
17. Werth VP, Patel B, Concha JS, et al. SAT0512 Safety loop. J Invest Dermatol 2017;137:115–22.
and efficacy of lenabasum in refractory skin- 32. Wallace DJ. The use of quinacrine (Atrabine) in
predominant dermatomyositis subjects treated in rheumatic diseases: a reexamination. Semin Arthritis
an open label extension of trial JBT101-DM-001. Rheum 1989;18:282–97.
Ann Rheum Dis 2018;77(Suppl 2):1111–2. 33. Alves P, Bashir MM, Wysocka M, et al. Quinacrine
18. Huard C, Gulla SV, Bennett DV, et al. Correlation of suppresses tumor necrosis factor-a and IFN-a in
cutaneous disease activity with type 1 interferon dermatomyositis and cutaneous lupus erythemato-
gene signature and interferon B in dermatomyositis. sus. J Investig Dermatol Symp Proc 2017;18:
Br J Dermatol 2017;176:1224–30. S57–63.
19. Wong D, Kea B, Pesich R, et al. Interferon and bio- 34. Melles RB, Marmor MF. The risk of toxic retinopathy
logic signatures in dermatomyositis skin: specificity in patients on long-term hydroxychloroquine ther-
and heterogeneity across diseases. PLoS One apy. JAMA Ophthalmol 2014;132:1453–60.
2012;7:e29161. 35. Marmor MF, Kellner U, Lai TYY, et al. Recommenda-
20. Liao AP, Salajegheh M, Nazareno R, et al. Interferon tions on screening for chloroquine and hydroxychlor-
beta is associated with type 1 interferon-inducible oquine retinopathy (2016 revision). Ophthalmology
gene expression in dermatomyositis. Ann Rheum 2016;123:1386–94.
Dis 2011;70:831–6. 36. Yokogawa N, Eto H, Tanikawa A, et al. Effects of hy-
21. Higgs BW, Zhu W, Morehouse C, et al. A phase 1b droxychloroquine in patients with cutaneous lupus
clinical trial evaluating sifalimumab, an anti-IFN-a erythematosus: a multicenter, double-blind, random-
monoclonal antibody, shows target neutralisation of ized, parallel-group trial. Arthritis Rheumatol 2017;
a type I IFN signature in blood of dermatomyositis 69:791–9.
and polymyositis patients. Ann Rheum Dis 2014; 37. Chasset F, Arnaud L, Jachiet M, et al. Changing anti-
73(1):256–62. malarial agents after inefficacy or intolerance in
Connective Tissue Disease 47

patients with cutaneous lupus erythematosus: a Erythematosus. Available at: https://clinicaltrials.


multicenter observational study. J Am Acad Derma- gov/ct2/show/NCT03161483.
tol 2018;78:107–14. 49. Petri M, Wallace DJ, Spindler A, et al. Sifalimumab, a
38. Chang AY, Piette EW, Foering KP, et al. Response to human anti-interferon-a monoclonal antibody, in sys-
antimalarial agents in cutaneous lupus erythemato- temic lupus erythematosus: a phase I randomized,
sus: a prospective analysis. Arch Dermatol 2011; controlled, dose-escalation study. Arthritis Rheum
147:1261–7. 2013;65:1011–21.
39. Chasset F, Bouaziz JD, Costedoat-Chalumeau N, 50. Khamashta M, Merrill JT, Werth VP, et al. Sifalimu-
et al. Efficacy and comparison of antimalarials in mab, an anti-interferon-a monoclonal antibody, in
cutaneous lupus erythematosus subtypes: a sys- moderate to severe systemic lupus erythematosus:
tematic review and meta-analysis. Br J Dermatol a randomised, double-blind, placebo-controlled
2017;177:188–96. study. Ann Rheum Dis 2016;75:1909–16.
40. Frances C, Cosnes A, Duhaut P, et al. Low blood 51. Furie R, Khamashta M, Merrill JT, et al. CD1013
concentration of hydroxychloroquine in patients study investigators. anifrolumab, an anti-interferon-
with refractory cutaneous lupus erythematosus: a a receptor monoclonal antibody, in moderate-to-
French multicenter prospective study. Arch Derma- severe systemic lupus erythematosus. Arthritis
tol 2012;148:479–84. Rheumatol 2017;69:376–86.
41. Chasset F, Arnaud L, Costedoat-Chalumeau N, et al. 52. Peterson LS, Nelson AM, Su WP. Classification of
The effect of increasing the dose of hydroxychloro- morphea (localized scleroderma). Mayo Clin Proc
quine (HCQ) in patients with refractory cutaneous 1995;70:1068–76.
lupus erythematosus (CLE): an open-label prospec- 53. Laxer RM, Zulian F. Localized scleroderma. Curr
tive pilot study. J Am Acad Dermatol 2016;74:693–9. Opin Rheumatol 2006;18:606–13.
42. Fruchter R, Kurtzman DJB, Patel M, et al. Character- 54. Teske N, Welser J, Jacobe H. Skin mapping for the
istics and alternative treatment outcomes of classification of generalized morphea. J Am Acad
antimalarial-refractory cutaneous lupus erythemato- Dermatol 2018;78:351–7.
sus. JAMA Dermatol 2017;153:937–9. 55. Burger E, Paniagua R, Monson N, et al. Myelin basic
43. Kindle SA, Wetter DA, Davis MDP, et al. Lenalido- protein antibodies as a biomarker in Morphea.
mide treatment of cutaneous lupus erythematosus: J Invest Dermatol 2018;138(5 Suppl):S5.
the Mayo Clinic experience. Int J Dermatol 2016; 56. Walker D, Susa JS, Currimbhoy S, et al. Histopatho-
55:e431–9. logical changes in morphea and their clinical corre-
44. Fennira F, Chasset F, Soubrier M, et al. Lenalidomide lates: results from the Morphea in adults and
for refractory chronic and subacute cutaneous lupus children cohort V. J Am Acad Dermatol 2017;76:
erythematosus: 16 patients. J Am Acad Dermatol 1124–30.
2016;74:1248–51. 57. O’Brien JC, Rainwater YB, Malviya N, et al. Tran-
45. Chasset F, Tounsi T, Cesbron E, et al. Efficacy and scriptional and cytokine profiles identify CXCL9 as
tolerance profile of thalidomide in cutaneous lupus a biomarker of disease activity in Morphea.
erythematosus: a systematic review and meta-anal- J Invest Dermatol 2017;137:1663–70.
ysis. J Am Acad Dermatol 2018;78:342–50. 58. Chen JK, Chung L, Fiorentino DF. Characterization
46. Nakayama Y, Kosek J, Capone L, et al. Aiolos over- of patients with clinical overlap of morphea and sys-
expression in systemic lupus erythematosus B cell temic sclerosis: a case series. J Am Acad Dermatol
subtypes and BAFF-induced memory B cell differen- 2016;74:1272–4.
tiation are reduced by CC-220 modulation of cere- 59. Kiguradze T, Anderson K, Siddalb C, et al. Body site
blon activity. J Immunol 2017;199:2388–407. distribution of pediatric-onset morphea. J Invest
47. Werth VP, Furie R, Gaudy A, et al. CC-220 decreases Dermatol 2018;138(5 Suppl):S57.
B-cell subsets and plasmacytoid dendritic cells in 60. Kashem SW, Correll CK, Vehe RK, et al. Inflamma-
systemic lupus erythematosus (SLE) patients and is tory arthritis in pediatric patients with morphea.
associated with skin improvement: pharmacody- J Am Acad Dermatol 2018;79(1):47–51.e2.
namic results from a phase IIa proof of concept study. 61. Zulian F, Vallongo C, Woo P, et al. Localized sclero-
(abstract) Arthritis Rheumatol 2017;69(suppl 10). derma in childhood is not just a skin disease.
Available at: https://acrabstracts.org/abstract/cc- Arthritis Rheum 2005;52(9):2873–81.
220-decreases-b-cell-subsets-and-plasmacytoid- 62. Klimas NK, Shedd AD, Bernstein IH, et al. Health-
dendritic-cells-in-systemic-lupus-erythematosus-sle- related quality of life in morphoea. Br J Dermatol
patients-and-is-associated-with-skin-improvement- 2015;172:1329–37.
pharmacodynamic-results-from-a-phase-iia-proof/. 63. Tollefson MM, Chiu YE, Brandling-Bennett HA, et al.
Accessed June 29, 2018. Discordance of pediatric morphea treatment by pe-
48. A Study to Evaluate the Efficacy and Safety of diatric dermatologists. Pediatr Dermatol 2018;35:
CC-220 in Subjects With Active Systemic Lupus 47–54.
48 Fernandez

64. Strickland N, Patel G, Strickland Am Jacobe H. Atti- addendum to the 2012 Revised international chapel
tudes and trends in the treatment of morphea: a na- hill consensus conference nomenclature of vasculit-
tional survey. J Am Acad Dermatol 2015;72:727–8. ides. Arthritis Rheumatol 2018;70:171–84.
65. Condie D, Grabell D, Jacobe H. Comparison of out- 76. Mills JA, Michel BA, Bloch DA, et al. The American
comes in adults with pediatric-onset morphea and College of Rheumatology 1990 criteria for the classi-
those with adult-onset Morphea. Arthritis Rheumatol fication of vasculitis. Arthritis Rheum 1990;33:
2014;66:3496–504. 1114–21.
66. Adeeb F, Anjum S, Hodnett P, et al. Early- and late-
77. Ozen S, Pistorio A, Iusan SM, et al. EULAR/PRINTO/
stage morphea subtypes with deep tissue involve-
PRES criteria for Henoch–Schönlein purpura, child-
ment is treatable with Abatacept (Orencia). Semin
hood polyarteritis nodosa, childhood Wegener gran-
Arthritis Rheum 2017;46:775–81.
ulomatosis and childhood Takayasu arteritis: Ankara
67. Stausbøl-Grøn B, Olesen AB, Deleuran B, et al. Aba-
2008. Part II: final classification criteria. Ann Rheum
tacept is a promising treatment for patients with
Dis 2010;69:798–806.
disseminated morphea profunda: presentation of
78. Feasel P, Billings SD, Bergfeld WF, et al. Direct
two cases. Acta Derm Venereol 2011;91:686–8.
immunofluorescence testing in vasculitis—A single
68. Khanna D, Denton CP, Lin CJF, et al. Safety and ef-
institution experience with Henoch-Schönlein pur-
ficacy of subcutaneous tocilizumab in systemic scle-
pura. J Cutan Pathol 2018;45:16–22.
rosis: results from the open-label period of a phase II
randomised controlled trial (faSScinate). Ann Rheum 79. Poterucha TJ, Wetter DA, Gibson LE, et al. Corre-
Dis 2018;77(2):212–20. lates of systemic disease in adult Henoch-Schönlein
69. Lythgoe H, Baildam E, Beresford MW, et al. Tocilizu- purpura: a retrospective study of direct immunofluo-
mab as a potential therapeutic option for children rescence and skin lesion distribution in 87 patients
with severe, refractory juvenile localized sclero- at Mayo Clinic. J Am Acad Dermatol 2012;67:612–6.
derma. Rheumatology 2018;57:398–401. 80. Takeuchi S, Soma Y, Kawakami T. IgM in lesional
70. Martini G, Campus S, Raffeiner B, et al. Tocilizumab skin of adults with Henoch-Schönlein purpura is an
in two children with pansclerotic morphoea: a hope- indication of renal involvement. J Am Acad Dermatol
ful therapy for refractory cases? Clin Exp Rheumatol 2010;63:1026–9.
2017;35(Suppl. 106):S211–3. 81. Belli AA, Dervis E. The correlation between cuta-
71. Kreuter A, Hyun J, Stucker M, et al. A randomized neous IgM deposition and renal involvement in adult
controlled study of low-dose UVA1, medium-dose patients with Henoch-Schönlein purpura. Eur J Der-
UVA1, and narrowband UVB phototherapy in the matol 2014;24:81–4.
treatment of localized scleroderma. J Am Acad Der-
82. Milani GP, Lava SAG, Ramelli V, et al. Prevalence
matol 2006;54:440–7.
and characteristics of nonblanching, palpable skin
72. Andres C, Kollmar A, Mempel M, et al. Successful
lesions with a linear pattern in children with He-
ultraviolet A1 phototherapy in the treatment of local-
noch-Schönlein syndrome. JAMA Dermatol 2017;
ized scleroderma: a retrospective and prospective
153(11):1170–3.
study. Br J Dermatol 2010;162:445–7.
83. Sais G, Vidaller A, Jucglà A, et al. Colchicine in the
73. Vasquez R, Jabbar A, Khan F, et al. Recurrence of
treatment of cutaneous leukocytoclastic vasculitis.
morphea after successful ultraviolet A1 photother-
Results of a prospective, randomized controlled
apy: a cohort study. J Am Acad Dermatol 2014;70:
trial. Arch Dermatol 1995;131(12):1399–402.
481–8.
74. Jennette JC, Falk RJ, Bacon PA, et al. 2012 revised 84. Clinical Transcriptomics in Systemic Vasculitis
International Chapel Hill Consensus Conference (CUTIS). Available at: https://clinicaltrials.gov/ct2/
Nomenclature of Vasculitides. Arthritis Rheum show/NCT03004326.
2013;65(1):1–11. 85. A Randomized Multicenter Study for Isolated Skin
75. Sunderkötter CH, Zelger B, Chen KR, et al. Nomen- Vasculitis (ARAMIS). Available at: https://clinicaltrials.
clature of cutaneous vasculitis: dermatologic gov/ct2/show/NCT02939573.

You might also like