Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Modern Rheumatology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/imor20

Biologics in refractory idiopathic inflammatory


myositis (IIM): What experience in juvenile vs
adult myositis tells us about the use of biologics in
pediatric IIM

Anjali Patwardhan & Charles H. Spencer

To cite this article: Anjali Patwardhan & Charles H. Spencer (2021): Biologics in refractory
idiopathic inflammatory myositis (IIM): What experience in juvenile vs adult myositis tells us about
the use of biologics in pediatric IIM, Modern Rheumatology, DOI: 10.1080/14397595.2021.1881027

To link to this article: https://doi.org/10.1080/14397595.2021.1881027

Published online: 06 Apr 2021.

Submit your article to this journal

Article views: 102

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=imor20
MODERN RHEUMATOLOGY
https://doi.org/10.1080/14397595.2021.1881027

REVIEW ARTICLE

Biologics in refractory idiopathic inflammatory myositis (IIM): What experience


in juvenile vs adult myositis tells us about the use of biologics in pediatric IIM
Anjali Patwardhana and Charles H. Spencerb
a
School of Medicine, University of Missouri, Columbia, MO, USA; bUniversity of Mississippi Medical Center, Batson Children’s Hospital,
Jackson, MS, USA

ABSTRACT ARTICLE HISTORY


Juvenile dermatomyositis (JDM) is an extremely heterogeneous orphan disease with limited amount of Received 13 November 2020
dedicated research on the subject matter. Recent research suggests that JDM may not just be the clas- Accepted 21 January 2021
sic antibody driven complements mediated microangiopathy as was thought to be in the past. The
etiopathogenesis of JDM also involves inappropriate stimulation of innate immune system followed by KEYWORDS
Anti-TNF agents;
dysregulation of the adaptive immune response through dendritic cells. Many variable immune factors polymyositis; idiopathic
such as genetics, major histocompatibility complex expressions, immunohistochemical variabilities, and inflammatory myopathy;
diversity in specific and associated autoantibodies may make individual IIM and JDM cases unique. juvenile dermatomyo-
The diversity in IIM and JDM also explains individual variability in response to specific therapies. sitis; rituximab
Classifying and matching the right patients to the right treatment is crucial to the successful treatment
of these patients with better outcomes. Sub-type specific biologic therapy may be the best current
treatment that can match the patient to the best treatment options. A PubMed search was performed
to find all the available cases of refractory myositis patients treated with biologics up to July 2020.
Using this search this article reviews all the current biologic treatment options and experiences for
both adults and children in the context of recent basic science to assist pediatric rheumatologists in
choosing the optimal biologic therapy for a child with recalcitrant JDM.

Background been recognized that inappropriate stimulation of the innate


immune system (interferons and interferon-regulated pro-
IIM, including JDM, is an extremely heterogeneous group
teins) followed by dysregulation of the adaptive immune
of muscle disorders with differences in cytokine expression
response through dendritic cells is also crucial to IIM etio-
profiles, genetics, diseases course, response to treatment,
pathology, especially in DM and JDM [2,3].
and prognosis. Apart from heterogeneity in the IIM There is growing research and published information on
patients, the diagnostic criteria and criteria for improve- serological and genetic biomarkers to classify IIM into dif-
ment, relapse, and remission selected by different research ferent subgroups better. The specific autoantibodies directed
groups in North America and Europe have differences and against some intracellular organelles are becoming an excel-
thereby make the research results harder to compare. There lent tool for confirming the diagnosis of different IIM dis-
is limited dedicated research on biologics in adult and espe- eases and also in identifying the disease subsets, which may
cially pediatric myositis. On most occasions, information run a similar clinical course. As expected, not all IIM
acquired from adult research experience or mixed popula- patients are positive for these autoantibodies, and the role
tion (adult and pediatric population) is used to treat pediat- of these IIM-specific and IIM-associated antibodies
ric myositis populations. is evolving.
Historically, IIM was recognized as antibody driven-com- Despite energetic attempts to unravel new treatment
plements mediated microangiopathy that involved skin, options, the first-line conventional therapy for JDM has not
internal organs, and muscles. It was believed that B cells changed to a great extent over since the 1990s. The initial
play a crucial role. Pieces of evidence supported the auto- treatment still primarily consists of corticosteroids (prednis-
immune etiology of IIM, suggesting the role of B cells. one, intravenous methylprednisolone) as the mainstay of
Evidence was found of immune-mediated muscle damage, induction therapy. The disease-modifying anti-rheumatic
presence of myositis-specific and associated autoantibodies drugs (DMARDs) methotrexate (MTX) or azathioprine
(MAS, MSA), infiltration of muscles with immune cells in (AZA) or mycophenolate (MP) are used with corticosteroids
myofibrils, and the overexpression of major histocompatibil- as the first line steroid-sparing agents with or without IVIg
ity complex (MHC, class I and II) on these myofibrils and infusions. Hydroxychloroquine, corticosteroid cream, and
other muscle components [1]. However, since 2005 it has sunblock are adjunctive therapy. The second line of

CONTACT Anjali Patwardhan patwardhana@health.missouri.edu School of Medicine, University of Missouri, 400 Keene Street, Suite 102 H. South Pavilion,
Columbia, MO, 65201, USA
ß 2021 Japan College of Rheumatology
2 A. PATWARDHAN AND C. H. SPENCER

treatment often consists of mycophenolate mofetil (MMF), published case reports provide limited yet useful informa-
cyclosporine (CSA) or tacrolimus as well as MTX or AZA if tion about the selection of patients for rituximab, the
not used earlier. The tacrolimus and cyclosporine are con- response rate, and likely side effects of rituximab therapy in
sidered better choices if the patient has interstitial lung dis- IIM and JDM [8,10,11].
ease (ILD). Finally, as third-line agents, intravenous
cyclophosphamide and/or plasma exchange might be
reserved for the critically ill patient [4–7]. Experience with rituximab
Based on the response to the conventional therapy or Currently, rituximab is not yet used as initial therapy but
frequent relapses on the conventional treatment, diseases used in recalcitrant adult and pediatric IIM patients with
course is called recalcitrant [8]. We will often use the terms, reasonable success after corticosteroids, methotrexate and
recalcitrant/resistant-to-treat/refractory based on the individ- similar drugs, and IVIg. Before selecting patients for rituxi-
ual study though we prefer recalcitrant. The refractory IIM mab therapy, it is optimal to perform due diligence in deter-
is defined by “The Rituximab for the Treatment of mination of the diagnosis and subtyping of the IIM patients
Refractory Adult and Juvenile Dermatomyositis (DM) and to optimize successful outcomes.
Adult Polymyositis (PM) trial” (ClinicalTrials.gov Identifier: Rituximab is known to have an improvement in muscle
NCT00106184) as ‘Refractory myositis, defined by intoler- enzymes, muscle strength, and steroid-sparing effect with
ance to or inadequate response to corticosteroids plus an good drug tolerability and safety profile in most recalcitrant
adequate regime of at least one other immunosuppressive adult as well as juvenile IIM patients [8–11]. Though some
agent. Intolerance is defined as side effects that require dis- patients require repeat treatment cycles [12,13]. The
continuation of the medication or an underlying condition improvements are mostly reported to continue for a variable
that precludes further use of the medication’ [9]. This period in different studies ranging from 6 months to 3 years
review provides an appraisal of experiences and the treat- [14–16]. All the core set measures of myositis responded to
ment options in the context of specific patient types as well rituximab therapy well. Still, the improvements in muscle
as evolving basic and clinical science to help pediatric diseases are reported to be greater than the skin diseases in
rheumatology clinicians choose biologics in recalcitrant rituximab-treated patients [8,9,17]. The response in DM
JDM patients. sub-groups of patients appears to be better than JDM and
all other sub-groups of IIMs patients [8,9,17]. Plus, there
Methods appears to be a latent period between the first rituximab
therapy and clinical response, which ranged in different
The literature research was conducted on PubMed studies from 2 to 6 months [13,14,18].
(MEDLINE) data source up to July 2020 to look at specific Although in the RIM study (RIM: NCT00106184), the
research question using (PICO) model i.e. people/popula- primary and secondary endpoints were not reached, and the
tion/problem, intervention, comparisons/placebo, outcomes. study could not show any significant differences in the
We used (P) patients (adults as well as Juvenile) with treatment and placebo arm, the overall response in 85% of
Idiopathic Immune Myositis, (I) Biologic therapies, (C) patients were considered positive. The failure to reach end-
compared different studies/experiences, and looked at the points in the RIM study was attributed to the study design
outcomes of individual studies/experiences. A similar search in which the rituximab latent period was not taken into
approach was used to identify reports on individual biologic account. However, the researchers could still show that rit-
use in IIM patients. Only human research was included. In
uximab had a steroid-sparing effect and good tolerability
this review, we are classifying therapies based on their
throughout 44 weeks of the study period as well as improve-
molecular targets of action. As these diseases are rare, it was
ment in 83% of all randomized patients by 20 weeks [11].
not considered necessary to use other search methods such
Rituximab did not consistently show improvement in calci-
as EMBASE, Cochrane Library, or others.
nosis and lipomatosis in JDM patients [15].
In some un-structured studies, on the re-evaluation of
Biologic therapies non-responders to rituximab therapy, new diagnoses (Non-
IIM) were reassigned. These changes in diagnosis re-empha-
B-Cell therapies size the importance of due diligence in the diagnosis and
Rituximab (RTX, anti-CD20 monoclonal antibody) subtyping of patients before instituting biologic therapy
The rationale and mechanism for the therapy. B-Cells [19]. Anecdotal studies in IBM patients have reported a
appear to play a vital role in the initiation and proliferation steroid-sparing effect with improvement in lab abnormalities
of the B-cell-driven immune reactions and be crucial for the after rituximab therapy. These changes included a reduction
pathogenesis of IIM, thereby making rituximab a reasonable in anti-SRP antibodies suggesting the role of B-cell and
therapeutic choice. Rituximab works by depleting CD20 B anti-SRP in etiology [13,20]. Rapid lab test improvements
cells while sparing memory cells and thus has a latent but a poor clinical response to rituximab in many patients
period. There are multiple published reports on varied with anti-SRP positive myopathy may be due to a lack of
responses to rituximab therapy in IIM and JDM patients. In compensatory muscular hypertrophy leading to poor muscle
the absence of standardization and control groups, these strength [21].
Table 1. Summary of clinical trials of biologics in immune inflammatory myositis.
Biologic Main mechanism of action The rationale of use Clinical trial Status Population Country Sponsor/Collaborator
Rituximab Anti-CD20 Myositis is considered an antibody NCT01862926. A Randomized, Double Rec. Adults with IIM and UK Royal Brampton and Harefield
monoclonal antibody mediated vasculitis therefore Blind Controlled Trial Comparing connective NHS Foundation Trust
elimination antibody producing Rituximab Against Intravenous tissue diseases
cells should help controlling Cyclophosphamide in Connective Tissue
the disease. Disease Associated Interstitial Lung
Disease. Phase 2 and Phase 3.
Enrollments; 116. Randomized.
Interventional. Treatment. Parallel
Assignment
Quadruple masking. Study Start Date:
November 2014. Completion Date:
November 2019.
NCT00106184. Rituximab Therapy in Com. Adults DM and PM. JDM USA University of Pittsburgh.
Refractory Adult and Juvenile (age ≥5 years)
Idiopathic Inflammatory Myopathy
(IIM). Phase 2. Randomized,
interventional. Parallel Assignment.
Triple masking. Treatment. 200
participants. Study Start Date: March
2006. Study Completion Date:
August 2010.
NCT01632124. Rituximab-induced Com. Adult RA and IIM. CH University Hospital Zurich
pulmonary function changes - an
observational study in patients with
rheumatoid arthritis and inflammatory
myositis. 80 participants. Observational
model. Study start date: January 2012.
Study completion date:
December 2013.
NCT00774462. Rituximab for the Com. Adults with myositis and Paris, France Assistance Publique - Hôpitaux
Treatment of Refractory Inflammatory generalized de Paris and Roche
Myopathies and Refractory Myasthenia myasthenia gravis. Pharma AG
Gravis (FORCE). Phase 2. Enrollment:
200. Interventional. Total 30
participants. Single group assignment.
Open Label. Study Start Date: January
2008. Study Completion Date:
December 2011.
NCT00106184. Rituximab Therapy in Com. Adults and pediatric IIM. Canada, CZ, University of Pittsburgh.
Refractory Adult and Juvenile Sweden, USA Genentech, Inc. Biogen
Idiopathic Inflammatory Myopathy
(IIM). Phase 2.
RIM trial. Study Start Date: March 2006.
Primary Completion Date: February
2010. Randomized.
Interventional for treatment. Parallel
Assignment
Triple masking.
Belimumab B-cell activating factor The evidence suggests that B- cells NCT02347891. Belimumab for Maintenance Rec. Adults with DM and PM. USA Northwell Health and
(BAFF) or B-lymphocyte play a significant role in the Therapy in Idiopathic Inflammatory GlaxoSmithKline.
stimulator (BLyS) pathogenesis of idiopathic Myositis. Phase 2 and
MODERN RHEUMATOLOGY

inhibitor human inflammatory myositis. High Phase 3. Estimated Enrollment: 60.


monoclonal antibody levels of the B cell survival Randomized.
(continued)
3
4
Table 1. Continued.
Biologic Main mechanism of action The rationale of use Clinical trial Status Population Country Sponsor/Collaborator
cytokine known as B cell Interventional-treatment. Crossover
activating factor (BAFF) are Assignment
found in the serum and muscle Triple masking.
of the IIM patients therefore Study Start Date: January 2015.
BAFF inhibition is considered as Completion Date: December 2019
a therapeutic option for
IIM patients
Alemtuzumab Monoclonal anti- The sensitized, cytotoxic CD8+ T NCT00079768. Effects of a T cell-depleting Com. IBM United States National Institutes of Health
(Campath) CD52 antibody cells clonally expand in situ and monoclonal antibody, alemtuzumab, in clinical center,
raid MHC-I-expressing myofibrils patients with inclusion body myositis: Bethesda, Maryland
to produce IBM. Alemtuzumab phase 2. A pilot clinicopathological
destroys surface marker CD52 study. Interventional-treatment.
bearing lymphocytes therefore Enrollment; 20 participants. Study start
A. PATWARDHAN AND C. H. SPENCER

preventing their clonal date: march 2004. Actual study


proliferation, activation and completion date: march 2007
therefore the attack on
muscle fibrils.
TNF Inhibitors
Abatacept Abatacept is a fusion Abatacept is a human soluble NCT01315938. Abatacept treatment in Study has passed DM, JDM CZ and UK. Karolinska Institute and Institute
protein that modulates recombinant protein consisting of polymyositis and dermatomyositis. its completion of Rheumatology, Prague,
CD28-CD80/86 pathway the cytotoxic t cell lymphocyte Phase 2. Study start date: January date and Czech Republic
mediated T cell co- antigen-4 (CTLA4) fused with Fc 2011, enrollment: 20 participants. status has not King’s College Hospital
stimulatory signal. region of human IgG1. This is Estimated study completion date: June been verified NHS Trust
modified to prevent complement 2013. in more than
fixation and therefore antibody- interventional-treatment. Randomized, 2 years.
dependent cytotoxicity. Antigen- single group assignment. Single
presenting cells (APCs) express masking (outcomes assessor). Delayed-
CD80 (B7-1) and CD86 (B7-2) start trial
molecules to which abatacept NCT03215927. Randomized, controlled Rec. Myositis with interstitial USA Bristol-Myers Squibb and
binds and leads to inhibition of pilot trial to evaluate the efficacy and lung disease (ILD) University of Pittsburgh
signal transduction and T cell safety of subcutaneous abatacept in
activation when CTLA4 binds with treating interstitial lung disease
CD80 or CD86. CTLA4, CD28, CD86, associated with the anti-synthetase
and CD40 have been found to be syndrome. Phase 2. Study start date:
expressed on the inflammatory October 9, 2017. Estimated study
cells in the muscle biopsy completion date: October 1, 2019.
specimens and CTLA4 and CD28 on Enrollment: 20 participants.
muscle cells of patients with Interventional-treatment. Randomized
myositis and supposed to play the parallel assignment.1:1 randomization
role in inflammation. for abatacept 125 mg vs. Placebo
/weekly SQ injection for 24 weeks.
quadruple masking, only the site
pharmacist is unblinded.
NCT02971683. Randomized, double-blind Rec. active IIM USA Bristol-Myers Squibb
clinical trial to evaluate the efficacy
and safety of abatacept SC with
standard treatment compared to
standard treatment alone in improving
disease activity in adults with active
IIM. Phase 3. Study start date: March
2017. Estimated primary completion
date: June 2020. Enrollment: 150
participants. Randomized.
(continued)
Table 1. Continued.
Biologic Main mechanism of action The rationale of use Clinical trial Status Population Country Sponsor/Collaborator
Interventional-treatment. parallel
assignment. Quadruple masking.
NCT02594735. Abatacept for the treatment Rec. JDM USA George Washington University
of refractory juvenile dermatomyositis.
Phase 4, study start date: November
2015, enrollment: 10 participants.
Estimated study completion date:
December 2017. Interventional-
treatment. Single group assignment.
no masking, open label.
NCT02971683. A ‘Phase 3, Randomized,
Double-Blind Clinical Trial to Evaluate
the Efficacy and Safety of Abatacept SC
With Standard Treatment Compared to
Standard Treatment Alone in Improving
Disease Activity in Adults with Active
Idiopathic Inflammatory Myopathy
(IIM)’. (Study Start Date: 13 March
2017. Estimated Study Completion
Date: 18 June 2021.)
Adalimumab Adalimumab a fully human The rationale of use of anti TNF No clinical trials
monoclonal antibody therapy in resistant to treat IIM
(Anti-TNF antibody comes from the knowledge that
group of medication) patients with myositis exhibit
Etanercept Etanercept a fusion protein elevated levels of TNF-α both in NCT00282880. NCT00112385. A Pilot Study Com. DM (18–65 years) USA Brigham and Women’s Hospital
comprising two human p75 the serum as well in the of Etanercept in Dermatomyositis. and Amgen
TNF-α receptors coupled to calcinosis cutis Phase 1. Study Start Date: March 2006.
the Fc portion of a Enrollment; 16 participants. Actual
monoclonal human Primary Completion Date: June 2010.
antibody. Interventional-Treatment. Randomized.
(anti-TNF antibody group Parallel Assignment. Double masking.
of medication)
NCT00802815. Double-blind, randomized, Com. IBM USA Washington University School
placebo-controlled trial of etanercept of Medicine and Amgen.
for 12 months in subjects with
inclusion body myositis. Early phase 1.
Interventional-treatment. Study start
date: April 2005. Enrollment: 20
participants. Actual primary completion
date: May 2014.
NCT00035958. Toward improved Term. JDM (age 4 − 16 years) USA Children’s Hospital Medical
understanding of pathogenesis and Center, Cincinnati
treatment of childhood onset Collaborators and
dermatomyositis. Phase 2and National Institute of Arthritis
phase 3study start date: August 2002. and Musculoskeletal and
Enrollment: 75 participants. Actual Skin Diseases (NIAMS)
primary completion date: august 2002.
Interventional-treatment. Randomized.
Parallel assignment. Open label.
Terminated (incorporating the
recommendations of the NIH-formed
MODERN RHEUMATOLOGY

DSMB in the study procedures would


(continued)
5
6
Table 1. Continued.
Biologic Main mechanism of action The rationale of use Clinical trial Status Population Country Sponsor/Collaborator
make the project budget over the limit
for this funding mechanism)
Pilot Study of Etanercept in Patients with Refractory Juvenile Dermatomyositis in USA. (Rouster-Stevens KA. At el.). Not a registered clinical trial.
Interventional-treatment. Enrollment: 9 participants. Open label.
Single center retrospective analysis of 7 patients (Katherine L Green. At el.). USA. Not a registered clinical trial.
Infliximab Infliximab is a humanized NCT00033891. Randomized, double-blind, Com. Adults with DM and PM USA National Institutes of Health
mouse monoclonal antibody placebo-controlled trial of infliximab in Clinical Center (CC) and
(Anti-TNF antibody group patients with dermatomyositis and National Institute of Arthritis
of medication) polymyositis. Phase 2. and Musculoskeletal and
interventional-treatment. randomized. Skin Diseases (NIAMS)
Crossover assignment. Quadruple
masking. Study start date: 10 April
2002.enrollment: 14 participants. actual
A. PATWARDHAN AND C. H. SPENCER

primary completion date: 20 May 2010.


NCT00443222. Phase 2 and Com. adults with DM, PM, IBM Sweden Schering-Plough and
phase 3. An open trial with TNF Karolinska Institute
blockade with infliximab, in patients
with chronic inflammatory myopathies.
First submitted date: 2 March 2007.
Start and completion date not
provided. Interventional-treatment.
non-randomized. Single group
assignment. Open label.
Interleukin inhibitors
Anakinra Interleukin 1 (IL1) An increased expression of il-1 in NCT01165008. anakinra in patients with Com. Adult PM, DM and IBM Sweden Karolinska Institute
receptor antagonist muscle tissue has been seen in refractory idiopathic inflammatory
patients with inflammatory myopathies phase 2-phase 3. Single
myopathies. Anakinra might favor group assignment
t-cell differentiation into th1 rather interventional-treatment. Study start
than th17 as indicated by more date: September 2003. Study
interferon gamma (IFNγ) and less completion date: September 2008.
IL-17A secretion NCT01150526. Phase 1. Enrollment: 41. Com. Adults with PM, DM USA Metabolic Technologies Inc.
Randomized. Interventional. parallel And Iowa State University
assignment. Masking: quadruple
masking. Primary purpose: basic
science. Study start date: October 2010.
Study completion date: July 2012.
Tocilizumab Anti interlukin-6 antibody IL-6 is involved in the growth and NCT02043548. Tocilizumab in the Rec. Adults with DM/PM USA Chester Oddis, University of
(anti IL-6Ab) differentiation of inflammatory treatment of refractory polymyositis Pittsburgh and
cells. Its role in activating B-cell and dermatomyositis. Phase 2. Start Genentech, Inc.
stimulating factor is recognized. date: October 2014. Estimated study
It is also responsible in growth completion date: January, 2019.
and differentiation of ‘T’ cells Randomized. Interventional-treatment.
therefore playing a crucial role parallel assignment. Triple masking.
in adaptive as well in innate Recruitment number − 40.
immune responses. T cells, B
cells, monocytes and endothelial
cells all can generate IL-6 which
in turn responsible for release
of inflammatory mediators after
binding to its receptor (IL-6R)
and leading to several
intracellular cytokine networks
(continued)
MODERN RHEUMATOLOGY 7

The patients IIM with lower physician’s global assess-


ment of damage and patients treated early (less than 3 years
Sponsor/Collaborator

disease duration) in the disease course are reported to


respond better than others [10,22]. But in the RIM-JDM
patients’ group, this association did not continue beyond 20
weeks post-Rituximab infusion suggesting it may not have
been due to rituximab therapy [22].

The ‘other’ mechanism theory


Country

The response to rituximab therapy in IIM is mostly associ-


ated with adequate B-cell depletion (BCD). Still, relapses are
not necessarily always associated with repopulation/normal-
ization of CD19þ cell count and/or IgG blood levels.
Several researchers have reported unpredictable and variable
correlation between reappearance of CD19þ cells/normal-
Population

ization of IgG and the need for retreatment/relapses, which


may suggest that rituximab may have more than one mech-
anism of actions [16]. The timing of post-rituximab therapy
CD19þ cell repletion and normalization of IgG levels is
considered variable, especially in pediatric patients. The nor-
malization of IgG and B cell numbers at 6 and 12 months
are reported not to be dependent on diseases sub-type or
Status

cycles of RTX therapy received [16,23,24]. Rituximab


induced B cell depletion, and B-cell recovery is not always
associated and proportional to clinical improvement, again
suggesting a possibility of an additional mechanism of
action [17].
The RIM-study results suggested that type-1 IFN-indu-
cible cytokines (IFNCK) and innate cytokine scores at base-
Clinical trial

line can predict the response rate to rituximab therapy in


recalcitrant IIM patients [25].
Most studies including RIM reported good response in
anti-t-RNA synthetase syndrome (ASS) [14,18,23] while
Unger et al. [10] in their series of 19 anti-t-RNA synthetase
syndrome (ASS) patients reported poor response and fre-
quent relapses with rituximab therapy. The autoantibody-
positive patients (MSA/MAA) had better response
correlation between elevated IL-
significantly correlated with the

interferon gene and chemokine


signatures. IL-6 is reported as a
only elevated but the elevation
dermatomyositis (DM) patients,

disease activity. There was also


that both in adult and juvenile

serum levels of IL-6 were not

rate[10,22,25,26].
pathways. There is evidence

inflammation on a mouse
The rationale of use

6 levels and the type I

mediator of muscle

RIM sub-studies
model myositis.

In the RIM sub-study, IFNCK high scores (>30) and posi-


tive autoantibodies were considered the biomarkers for dis-
ease activity [25]. The researchers reported an association
between pretreatment IFN-regulated chemokines levels with
response to rituximab therapy [27]. These biomarkers were
Main mechanism of action

associated with greater clinical responses to rituximab ther-


apy. The clinical response was reported greatest at 16 weeks
of post-rituximab treatment in patients positive for these
biomarkers [25].
Another group of researchers used the RIM study data
retrospectively and reported that at the baseline, the IFNCK
Table 1. Continued.

scores were higher in several MSA positive (anti-synthetase,


TIF1-c, and Mi-2 myositis) patients. They further identified
that after BCD the IFNCK scores improved in anti-synthe-
Biologic

tase, non-myositis associated autoantibodies (MAA), and


Mi-2 positive patients but deteriorated in TIF1-c
8 A. PATWARDHAN AND C. H. SPENCER

autoantibody-positive patients. These results suggested that levels also were higher in patients positive for anti-MDA5
anti-synthetase, non-MAA, and Mi-2 autoantibody-positive autoantibody and their titers were proportionately higher in
patients, as well as those with higher pretreatment IFNCK patients with high titer anti-MDA5 (>200 U). It is known
scores (>30), may have responded well to rituximab ther- that a high anti-MDA5 titer is associated with RP-ILD [35].
apy. Researchers reported an association amongst serum lev- The biologics used to block B-cell stimulation through
els of autoantibodies, IFNCK scores, correlation with disease BAFF/APRIL sites include belimumab as well as tabalumab,
activity, and response to the rituximab treatment. The anti- atacicept, and blisibimod [36–39]. Tabalumab is a human
SRP antibody levels only correlated with muscle enzyme lev- IgG4 monoclonal antibody against both membrane and sol-
els longitudinally and not necessarily with the muscle uble BAFF. Atacicept is a humanized recombinant fusion
strength and clinical response [25]. protein that acts as a ‘decoy receptor’ for BAFF/APRIL,
therefore, preventing the attachment of BAFF/APRIL to
their receptors [40].
Summary
Rituximab appears to be a reasonably safe and effective bio-
Experience with belimumab and similar biologics
logic medicine for IIM and JDM. It has shown excellent tol-
erability in single as well as multiple treatment cycles with The only clinical trial currently recruiting to evaluate the
only a little additional risk of infections when repeated ther- efficacy and safety of belimumab for maintenance therapy
apy cycles were needed. It has been reported to induce in refractory IIM is in progress. Northwell Health sponsors
medium-term remission in many patients, but long-term it, and GlaxoSmithKline was collaborating. The estimated
effects on disease outcomes and well as long-term adverse study enrollment is 60 subjects. It is a randomized, phase 2
events are still not known [22–24,26]. and 3, multicenter (USA) crossover assignment with triple
masking with an estimated completion date was 2020
(NCT02347891).
Belimumab and similar agents that block B-cell
stimulation
Summary of B cell stimulators in IIM and JDM
The rationale for therapy and mechanism
The stimulation of B-Cell occurs mostly due to two cytokine There is not enough current evidence to support the use of
factors: B-cell activating factor (BAFF) and a proliferation- these B-cell stimulation blockers in refractory IIM and espe-
inducing ligand (APRIL). These two cytokine factors are cially JDM, but there may be in the future. More experience
also essential for B-cell survival, B-cell proliferation, B-cell and research with the drug are needed. BAFF/APRIL biolog-
maturation, and B-cell differentiation to plasma cells. The ics are being studied in other autoimmune diseases with
BAFF is a member of the TNF family and appears to be BAFF/APRIL pathways etiologies such as SLE, Sjogren’s,
crucial for the development of autoimmunity [28–30]. and multiple arthritis diseases.
The overexpression of BAFF can lead to prolonged B cell
survival and sometimes B-cell malignancies. BAFF can
increase the expression of a subset of B cells called marginal Alemtuzumab
zone (MZ) B cells [31]. The MZ cells are known stimulators The rationale for therapy
for T-cells. An overexpression of MZ cells is seen in several Alemtuzumab is a recombinant DNA-derived humanized
autoimmune inflammatory conditions. monoclonal, IgG1 glycoprotein receptor CD52 antibody that
The BAFF levels are significantly higher in IIMs patients is specifically targeted against CD52 receptor carrying
than normal controls [32,33]. The BAFF serum levels are immune cells (T-lymphocytes and monocytes).
reported to be higher in IIM (especially JDM) with positive Alemtuzumab’s immunodepleting action is the rationale
autoantibodies but least elevated in anti-SRP positive behind its use in IIM and other autoimmune diseases.
patients [32,34]. The Anti-SRP positive patients are reported
to have significantly elevated IL-17 levels when compared to
other IIM subgroups, suggesting a pathogenic role [32]. Mechanism
Elevated BAFF/APRIL serum levels in IIM patients are Several interesting properties make alemtuzumab different.
also reported to be associated with interstitial lung diseases This glycoprotein receptor CD52 has a varied and patchy
(ILD). This appears to be true especially in rapidly progres- distribution in the human body. It has a higher distribution
sive interstitial lung disease (RP-ILD) in JDM. JDM patients on the B and T lymphocytes, lower on macrophages, and
with RP-ILD have BAFF/APRIL levels that are significantly minimal or none on stem cells, mature NK cells, and neu-
higher than in JDM patients with chronic-ILD. Yet the trophils (the innate immune system) [41–43]. A trend of an
BAFF/APRIL serum levels are significantly higher in JDM increasing number of neutrophils in circulation and lymph-
patients with chronic-ILD than in normal controls [35]. oid organs is seen with the increase in the alemtuzumab
Interestingly, it is reported that BAFF/APRIL serum levels dose and is believed to be due to demarginalization or
in JDM-ILD patients correlated well with blood Krebs von increased recruitment from the bone marrow [41]. In con-
den Lungen-6 (KL-6) and IL-18 levels, which are the bio- trast, the regulatory phenotypic T cells also show relative
markers for lung inflammation. The BAFF/APRIL serum sparing with alemtuzumab therapy [41–44].
MODERN RHEUMATOLOGY 9

Secondly, alemtuzumab’s response is dose-dependent, has been associated with a short latent period and long
and higher doses of the drug are needed to deplete lymph- remissions in the treated patients. On the negative side, it
oid organs completely. Because of the above two factors, may cause significant infusion reactions and induce other
alemtuzumab therapy is associated with a relatively low autoimmune diseases. Despite its use in critically ill patients,
infection rate [41,45–47]. Alemtuzumab infusions are also no mortalities have been reported so far. There are two dif-
associated with non-complement-mediated pulse release of ferent infectious risks with this drug. Alemtuzumab therapy
cytokines (TNF-a, IL-6, and IFN-c) that may lead to severe carries a relatively lower risk for common infections overall.
infusion reactions, especially with the first infusion [41]. It Interestingly, the risk of common infection decreases with
can cause side effects in the patient’s even years after it is subsequent doses of the therapy. It is explained as
stopped. Therefore, more than most biologics, patients ‘consistent with preservation of components of protective
treated with alemtuzumab require ongoing monitoring and immunity’ by Wray S et al. [54]. Though alemtuzumab ther-
long-term follow-up. apy may be a low risk for common infections, unsurpris-
ingly, it has been explicitly associated with a higher risk of
opportunistic infections and cytomegalovirus reactiva-
Experience with alemtuzumab
tion [55].
Alemtuzumab has been mostly used in IBM. In a ‘proof-of-
principle study,’ 0.3 mg/kg/day alemtuzumab was used for
four days in 13 sIBM patients (NCT00079768). The primary Tumor necrosis factor inhibitors
endpoint was disease stabilization at a 6-month endpoint The rationale for therapy
comparative to the natural history of the disease. The study Tumor necrosis factor (TNF), also called cachexin, or
was powered to capture at least 10% differential gain in cachectin, is a cytokine mostly produced by activated mac-
muscle power at six months in therapy. The researchers rophages but also by several other cells in the process of
reported not only depletion of peripheral blood lympho- systemic inflammation. These include natural killer cells,
cytes, but endomysial T cells were also depleted after alem- CD4þ lymphocytes, mast cells, neutrophils, neurons, and
tuzumab therapy. These changes were believed to be eosinophils. It is also responsible for producing acute
altering the natural disease course. The total observation phase responses.
period was 12 months. However, the results showed only a The rationale of the use of anti-TNF therapy in refractory
1.9% overall decline in muscle power, i.e. 13% differential IIM patients arises from multiple research studies.
gain at six months in therapy (p < .002). A 50% mean Along with elevated levels of interleukins (IL2, IL1a,
reduction in CD3 lymphocytes (p < .008) and reduced stres- IL1b), TNFb, and interferon (IFN c), myositis patients have
sor molecule mRNA expression was seen in follow-up biop- shown elevated titers of TNF-a in their serum as well as in
sies. The researchers reported a positive outcome of the the calcinosis cutis suggesting a pathogenic role [56–58].
treatment suggesting alemtuzumab treatment can slow down Significantly higher TNF-a levels, soluble TNF-R55 and
the disease process and change the course of the disease, TNF-R75 have been demonstrated in serum as well as in
reduce muscle inflammation and improve muscle strength calcium deposits in IIM patients vs. healthy controls [59,60].
at least for the short term. Long-term outcomes are not The TNFa-308A allele is associated with increased produc-
reported (NCT00079768) [43,45]. The mRNA expression tion of TNF-a, vascular occlusions, a chronic, polycyclic
levels of the proinflammatory chemokines did not change JDM disease course, as well as ulcerative skin JDM disease
with alemtuzumab treatment [48]. with calcinosis [61,62].
There are a few anecdotal case reports of successful use
of alemtuzumab in adults as well as juvenile recalcitrant PM
patients. Alemtuzumab showed a significant steroid-sparing Mechanisms of action
effect in these recalcitrant patients. The patients stayed in The TNF-a shows polymorphism at 308 promoter regions
moderate to long term remission for few years before one of (A-G polymorphism). The osteopontin promoter and TNF-
them required a successful second therapy cycle of alemtu- 308A allele polymorphism are found to be associated with
zumab [49–52]. A case of drug-induced IIM was reported in elevated levels of a interferon in the untreated myositis
an adult patient seven months of post-alemtuzumab therapy. patients, supporting its etiologic role [63]. TNF-a-308A
This myositis was later successfully treated with ste- allele polymorphism is also associated with a chronic disease
roids [51,53]. course, increased likelihood of calcinosis cutis, increased
occlusion of capillaries, increased expression of TNF-a in
myofibrils, and increased production of TNF-a by periph-
Summary – Alemtuzumab B cell depleting therapy in
eral mononuclear cells in JDM patients reinforcing its role
IIM and JDM
in the etiopathogenesis [56,59]. In an inception cohort of
At this point, alemtuzumab has shown some benefit in IBM. JDM patients, researchers reported that serum IFN-a was
However, there are not enough prospective research trials or significantly higher in female JDM patients who carried the
enough published experience available to recommend its use dual (OPN rs28357094G and TNF-a-308 A) alleles than
of conclusively in any IIM and JDM patients. On the posi- those who did not, suggesting ‘pathogenic synergy’ and
tive side in the few trials performed, alemtuzumab therapy ‘complex gene-gene-sex interaction’ as reported by Niewold
10 A. PATWARDHAN AND C. H. SPENCER

et al. [64]. The OPN rs28357094G gene is identified to be Recently, in a retrospective study in a large cohort of
more prevalent in female JDM patients as well as better JDM patients, successful use of adalimumab is
expressed in the female patients [64]. reported [74].
Isolated myositis had been successfully treated with adali-
mumab, such as Crohn’s diseases related to recurrent orbital
Experience with anti-TNF-a biologics in JDM
myositis, gastrocnemius myositis, and isolated recalcitrant
The current knowledge is based mostly on open-label stud- orbital myositis [75–78].
ies and shared experiences from research studies. One Adalimumab may have a role as a steroid-sparing treat-
exception is a pilot double-blind placebo-controlled RCT ment for inflammatory-stage thyroid eye disease (TED). The
that concluded that infliximab therapy (5mg/kg) was effect- TED occurs due to the upregulation of proinflammatory
ive and safe in a select group of resistant to treatment adult cytokines, mainly tumor necrosis factor-a (TNF-a) [79].
IIM patients [65,66]. There are also anecdotal reports of efficacy and safety of
Etanercept and infliximab are the most commonly adalimumab in the treatment of recalcitrant dermatomyo-
researched anti-TNF agents though one study suggested that sitis-associated interstitial lung disease [80,81].
etanercept may have no benefit in JDM or even exacerbate
JDM [65,67,68]. Other findings of anti-TNF studies in IIM. Anecdotal expe-
Calcinosis is associated with higher initial and prolonged riences, small randomized controlled studies, and open label
elevated muscle enzymes (especially CK levels). Efthimiou studies have shown mixed results ranging from improve-
et al. [66] reported that the patients who had high initial ments of IIM as well as steroid-sparing effects [82,83] on
serum creatine kinase (CK) levels responded best to anti- the one hand, to worsening of the diseases [84,85], develop-
TNF therapy, but others reported the exact opposite [69]. ing new-onset other autoimmune diseases and malignancy
after the treatment with anti-TNF therapy on the other [63].
Paradoxically, anti-TNF therapy itself can induce auto-
Adalimumab immune myositis in adults as well as juvenile patients. The
The rationale for therapy patients who have a genetic background and family history
Adalimumab was the first fully human monoclonal antibody of autoimmunity are more likely to develop autoimmune
against TNF-alpha receptor. The name Humira was derived myositis after anti-TNF therapy [70,86]. The mechanism of
from ‘human monoclonal antibody in rheumatoid arthritis’. this paradox is not entirely clear. Still, it is suggested that
Adalimumab as a fully humanized IgG1 monoclonal anti- after TNF-a blockage, the production of TNY-Y is
body to tumor necrosis factor-alpha after multiple studies increased, which is a known molecule in the etiopathogene-
and long clinical use is considered easy to give, safe, and as sis of DM/PM [87]. Anti-TNF therapy has shown promising
an effective biologic therapy as any anti-TNF drug. It is not results in patients with calcinosis associated myo-
known to produce the neutralizing autoantibodies or chi- sitis [68,74].
meric autoantibodies in the recipients; hence it appears to The incidence of developing chronic lung disease, ILD,
be better tolerated. Unfortunately, there are no formal RCT and drug-induced secondary PM/DM is increased if the
or registered clinical trials on its use in patients with IIM. treated patient is positive for anti-synthetase antibodies plus
Adalimumab historically had an unpredictable effectiveness has a history of ILD, or RA [63].
and side-effects profile when used in IIM patients, which is Anti-TNF therapy carries a specific benefit and risk in
evident from the following examples. the treatment of myositis. TNF–a is known to suppress/
There are several anecdotal reports of patients developing downregulate the INF-a system. When anti-TNF treatment
a myositis reaction after using adalimumab for other unre- is used, the INF-a system may be upregulated, potentially
lated rheumatologic diseases within 2 weeks to 3 years of leading to flareups of IFN-mediated diseases such as DM,
therapy. Most patients were adults with only a few pediatric JDM, or SLE. Fiorentino et al. [88] called this phenomenon
patients who were treated with adalimumab for their pri- ‘the yin and yang of TNF-a inhibition’.
mary diagnosis of [70–72]. The majority of patients who
developed myositis had histologic features of DM, and only
Summary of anti-TNF-a agents in IIM
a few had PM features [71]. The myositis in these patients
did not go away after stopping adalimumab and they Infliximab is the most studied anti-TNF agent in IIM
needed dedicated therapy for it. Most of the patients who patients and may have a definite benefit in some patients. It
developed myositis on adalimumab responded to pulse ster- may be the best of the group of anti-TNF biologics for IIM
oid therapy and only a few required additional DMARDs and JDM in 2020. Other anti-TNF therapy (adalimumab
such as methotrexate and azathioprine. On the other hand, and etanercept) may be effective for a tiny percentage of
a flare in myositis is reported in some overlap syndrome recalcitrant IIM and JDM patients, especially for a short-
patients after treatment with adalimumab for their arthritis term response [74]. Anti-TNF therapy has shown short
[73]. Interestingly, anecdotal reports are suggesting success- term improvement in IIM patients. Repeated cycles of anti-
ful use of adalimumab in patients who developed orbital TNF treatment can lead to the risk of developing anti-chi-
myositis secondary to use of infliximab. meric antibodies and severe infusion reactions.
MODERN RHEUMATOLOGY 11

The anti-TNF therapy in IIM is ‘Not-For-All’ but is Active Idiopathic Inflammatory Myopathy (IIM)’.
effective and safe in the very select group of recalcitrant IIM (Study Start Date: 13 March 2017. Estimated Study
patients where diligent selection of patients is the key to the Completion Date: 18 June 2021. NCT02971683)
success of the treatment [74]. There are anecdotal reports of c. A phase 4 trial, ‘Abatacept for the Treatment of
developing myositis after treatment with anti TNF agents Refractory Juvenile Dermatomyositis’. (Study Start
for other rheumatologic conditions such as arthritis and Date: November 2015. Estimated Study Completion
psoriasis. Some of these patients developed MSA positive Date: October 2020. NCT02594735)
myositis (Anti Jo-1) [89,90]. d. ‘Abatacept in the treatment of adult dermatomyositis
and polymyositis: a randomized, phase IIb treatment
delayed-start trial’ showed significant improvement in
Biologic therapy with CTLA4 anti-costimulatory
fifty percent of patients with IV Abatacept (10mg/
molecules; abatacept
Kg) [98].
The rationale and mechanism for therapy
Co-stimulatory molecules CD28 and CTLA-4 are known to
be upregulated in the muscle of IIM patients [91,92]. Summary on anti-CTLA4 anti-costimulatory molecule
Abatacept (CTLA4-Ig) is a fusion protein engineered to studies in IIM and JDM
modulate the CD28-CD80/86 pathway mediated T-cell co- In contrast to some of the anti-TNF biologics, abatacept
stimulatory signal. For the activation of the cytotoxic-T cells, appears to be a promising therapy for IIM, including JDM
co-stimulation through B7 (CD80/CD86), the binding of the patients. Several ongoing trials may shed light on abatacept’s
receptor CD28, is required. The CD28 co-stimulatory mol- promise. One advantage of using abatacept appears to be its
ecule, CTLA-4, CD86, CD40, and receptor CD80, are also relatively low renal and hepatic toxicity as compared to
expressed on myofibrils and in inflammatory infiltrates in other DMARDs and other biologic therapies.
JDM as well as in adult (DM, PM) patients identified by Further prospective research trials are on the horizon to
researchers through immunohistochemical staining. This evaluate the value of abatacept in IIM.
expression of co-stimulatory molecule CD28 and CTLA-4
appears to be brought about by proinflammatory cyto-
kines [93]. Interleukin inhibitors in myositis
Abatacept is a fusion of immunoglobulin and an extracel- The rationale for therapy. In IIM patients, an increased
lular component of cytotoxic T-lymphocyte antigen-4. In interleukin-1 (IL-1) expression in the muscle fibrils has
JDM, the plasmacytoid dendritic cells, macrophages, and been reported suggesting its role in pathogenesis [99].
CD4þ T and B lymphocytes appear to constitute the main
inflammatory cells scattered in perivascular and perimysium Anakinra. This biologic is an interleukin 1 (IL1) receptor
areas [91]. Macrophages were found sufficiently sensitive to antagonist used for rheumatoid arthritis (RA) and systemic
CTLA4-Ig that on the exposure, they could downregulate juvenile idiopathic arthritis (sJIA). The experience is limited
the production of IL1-b, IL-6, TNF-a, and TGF-b at the cel- and insufficient with IL-1 inhibitors in IIM. Mei Zong et al.
lular level through down-regulating the co-stimulatory sig- reported in their 12-month prospective use of anakinra on
nals [92]. By these mechanisms, abatacept is thought to 15 recalcitrant IIM patients. The clinical and molecular
downregulate the activation of cytotoxic-T cells through co- response in muscle biopsy specimens (cellular infiltrates, IL-
stimulatory signals as well as B cell activation [92,94]. 1a, IL-1b, IL-1Ra, and major histocompatibility complex-
There are some anecdotal reports of successful use of class I by immunohistochemistry) was assessed in all 15
abatacept in recalcitrant IIM patients such as with overlap refractory myositis patients. They were prospectively treated
features [95], severe calcifications and skin ulcerations with Anakinra for 12 months period [100]. The results were
[93,94], anti-signal recognition particle myopathy[96] and inconclusive. There are few other anecdotal reports of an
polymyositis [97]. inadequate response to anakinra therapy in other recalci-
Recently, the interest in abatacept has increased leading trant IIM patients [101–103].
to randomized controlled trials that are underway.
For example:
Tocilizumab
a. ‘Randomized, Controlled Pilot Trial to Evaluate the The rationale for therapy. Tocilizumab is an anti-interleu-
Efficacy and Safety of Subcutaneous Abatacept in kin-6 antibody (anti-IL-6Ab) used for RA and systemic and
Treating Interstitial Lung Disease Associated with the polyarticular JIA. The rationale for the use of interleukin-6
Anti-synthetase Syndrome’ is on-going (Study Start (IL-6) inhibitors in myositis originates from animal studies
Date: 1 June 2017, Estimated Completion Date: 31 and human research that showed the pivotal role of IL-6 in
December 2020. NCT03215927); the pathogenesis of DM, though less so in PM. Several
b. ‘Phase 3, Randomized, Double-Blind Clinical Trial to researchers demonstrated that the serum and the lympho-
Evaluate the Efficacy and Safety of Abatacept SC With cytes infiltrating the muscles in DM had high IL-6 expres-
Standard Treatment Compared to Standard Treatment sion [104–107]. There are a few limited reports available on
Alone in Improving Disease Activity in Adults with the use of tocilizumab in select refractory myositis patients
12 A. PATWARDHAN AND C. H. SPENCER

with anti-synthetase syndrome [108,109], polymyositis [110], alpha-chemokines (CXCL9, CXCL10) showed reasonable
and overlap syndromes [111]. reduction after 12 weeks of tofacitinib treatment but it did
Interestingly, tocilizumab is found to be effective in not reach statistical significance.
Familial Mediterranean Fever (FMF) patients associated There is an open label proof of concept, small (10 sub-
with protracted myalgia, myositis, and fasciitis, and aa amyl- jects) study that is now ongoing titled; Tofacitinib in
oidosis despite FMF being a neutrophil-derived autoinflam- Refractory Dermatomyositis (January 2017 – estimated com-
matory disease [112]. Paradoxically, there had been reports pletion June 2021) (STIR, NCT03002649) [122].
of the development of drug-induced isolated myositis with The Jak inhibitors have also been reported to have a
the use of tocilizumab also [113]. good effect on extensive myositis related calcifications in
A multicenter, double-blind placebo-controlled, parallel JDM [123,124].
assignment, phase two RCT of ‘Tocilizumab in the
Treatment of Refractory Polymyositis and Dermatomyositis
Anti-interferon-a monoclonal antibody: Sifalimumab
(TIM)’ (NCT02043548) is ongoing and pending results.
The rationale for therapy. The type I IFN-alpha is well rec-
Interleukin-17. The role of IL-17A is well recognized in sev- ognized in the etiology of immune inflammatory myositis
eral other chronic autoimmune-inflammatory diseases such syndromes. The treatment is still investigational in IIM and
as RA. An IIM patient with increased IL-17A producing JDM. Sifalimumab is an anti-IFN-a monoclonal antibody.
cells in the muscle has been reported suggesting a patho- The treatment with Sifalimumab is recognized to neutralize
genic role of IL-17A and the Th17 pathway in IIM and reduce type I IFN gene signature (IFNGS) in the blood
[114,115]. Further research and development are pending to as well as muscles of adult dermatomyositis and polymyosi-
know the precise value of therapeutic targeting of IL-17A in tis patients in several clinical trials (NCT00533091,
IIM patients. NCT00979654; Table 1) [121].

Summary of interleukin therapies in IIM and JDM. Limited


Experiences of pediatric rheumatologists in an
research and experience in interleukin inhibitors prohibit
international network
any recommendations for their routine use in IIM and
JDM. Anti-IL-1 therapy has not been effective in IIM One study has published the recent anecdotal experience of
patients. Theoretically, anti-IL-6 treatment is more likely to an international research group of pediatric rheumatologists
be effective in certain subgroups of DM/PM patients than in in the United States and Canada (Childhood Arthritis and
polymyositis patients. The utility of IL-6 inhibitors in DM/ Rheumatism Research alliance-CARRA) with biologics for
JDM is still being evaluated in RCT’s. JDM utilizing three surveys from 2011 to 2017. Nominal
group techniques were used for achieving consensus on the
optimal current choices of biologic drugs 2013–2017. The
Janus kinase inhibitor: Tofacitinib-(included though not
top four biologics for recalcitrant JDM chosen by these
technically a biologic)
techniques were in the order of preference rituximab, abata-
The rationale for therapy. Several cytokines only connect cept, tocilizumab, and infliximab [122].
with Type I and Type II cytokine receptors that employ a
special family of kinases for intracellular signaling and
Conclusion
action, known as janus kinases (JAK). The JAK drugs that
are used to inhibit signaling through JAK receptors are This detailed review of the literature gives a snapshot view
called janus kinase inhibitors (jakinibs). Cytokines can pro- of where we stand today in 2020 in our journey to explore
duce autoimmune diseases through intracellular signaling the best treatment options for patients with difficult to treat
though JAK. There are several types of JAK (JAK1, JAK2, IIM, and for pediatric rheumatologists, JDM.
JAK3 and Tyrosine Kinase 2 or TYK2) and inhibitors (first What does a pediatric rheumatologist do in 2020 in a
generation and second generations). recalcitrant JDM patient when conventional corticosteroids,
Tofacitinib is Food and Drug Administration (FDA) methotrexate, IVIG, and combinations with other immuno-
approved for RA (2016), ulcerative colitis (2018) and psori- suppressive drugs are insufficient, and the patient is doing
atic arthritis (2017). Oral tofacitinib has shown good effi- poorly? From our literature review, it is clear that the best
cacy, safety and steroid sparing effect in a recalcitrant alternative biologic therapy remains rituximab that depletes
classic, and cutaneous dermatomyositis adult patients in the B cells. Similarly, the B cell-depleting drug alemtuzumab
initial studies [116,117]. In several open label reports on also may be useful in IIM and JDM but needs more testing.
refractory JDM as well as adult DM patients, tofacitinib The other B cell therapies that block B cell stimulation may
therapy has appeared to lead to improvement in anti–mela- be available soon but are further behind in development
noma differentiation–associated protein 5 (anti-MDA5) and testing (tabalumab, atacicept, blisibimod).
positive amyopathic dermatomyositis (ADM) associated There is enough experience in the literature and anec-
interstitial lung disease (ILD) as well as with anti-NXP2 and dotally to recommend abatacept as the next biologic alterna-
anti-TIF1c dermatomyositis-specific autoantibodies positive tive. Rituximab and abatacept have the best literature and
patients [118–121]. Interestingly, the muscle expression of anecdotal support by far. There is modest support for
MODERN RHEUMATOLOGY 13

tocilizumab or infliximab as the next alternatives. There is 6. Kotani T, Takeuchi T, Makino S, Hata K, Yoshida S, Nagai K,
insufficient evidence to recommend any of the other com- et al. Combination with corticosteroids and cyclosporin-A
improves pulmonary function test results and chest HRCT
mercially available biologics, but no doubt, individual rheu- findings in dermatomyositis patients with acute/subacute inter-
matologists may utilize them due to necessity. Good stitial pneumonia. Clin Rheumatol. 2011;30(8):1021–8.
examples might be the anti-interferon-a monoclonal anti- 7. Oddis CV, Sciurba FC, Elmagd KA, Starzl TE. Tacrolimus in
body sifalimumab, and the janus kinase inhibitor refractory polymyositis with interstitial lung disease. Lancet.
tofacitibinib. 1999;353(9166):1762–3.
8. Munoz-Beamud F, Isenberg DA. Rituximab as an effective
There are inherent limitations to these studies that are alternative therapy in refractory idiopathic inflammatory myo-
the subject of this review as well as the disease category pathies. Clin Exp Rheumatol. 2013;31(6):896–903.
itself. Most of the published research is found in retrospect- 9. Oddis CV, Reed AM, Aggarwal R, Rider LG, Ascherman DP,
ive observational studies. The results are mostly in adults in Levesque MC, et al., RIM Study Group. Rituximab in the treat-
IIM and not in JDM. Notably, the most significant point is ment of refractory adult and juvenile dermatomyositis and
adult polymyositis: a randomized, placebo-phase trial. Arthritis
the type of IIM studied when evaluation the response to Rheum. 2013;65(2):314–24.
treatment. IIM remains a highly heterogeneous group with 10. Unger L, Kampf S, Luthke K, Aringer M. Rituximab therapy in
different histopathology and etiopathology mechanisms driv- patients with refractory dermatomyositis or polymyositis: differ-
ing their clinical outcomes and responses. These differences ential effects in a real-life population. Rheumatology (Oxford).
may significantly influence the results of these studies. Yet 2014;53(9):1630–8.
11. Barsotti S, Cioffi E, Tripoli A, Tavoni A, D’Ascanio A, Mosca
we must evaluate studies and draw conclusions as best we M, Neri R. The use of rituximab in idiopathic inflammatory
can in 2020 for the benefit of IIM and JDM patients who myopathies: description of a monocentric cohort and review of
are not doing well on conventional therapy and may need the literature. Reumatismo. 2018;70(2):78–84.
biologic treatment to prevent morbidity and mortality. 12. Majmudar S, Hall HA, Zimmermann B. Treatment of adult
These drugs will vary in availability in different countries inflammatory myositis with rituximab: an emerging therapy for
refractory patients. J Clin Rheumatol. 2009;15(7):338–40.
and in countries with commercial insurance, it may depend 13. Luca NJ, Atkinson A, Hawkins C, Feldman BM. Anti-signal
on what is government approved and what the insurances recognition particle-positive juvenile polymyositis successfully
will pay for. treated with rituximab. J Rheumatol. 2012;39(7):1483–5.
We believe that future clinical effectiveness trials may be 14. Basnayake C, Cash K, Blumbergs P, Limaye V. Use of rituxi-
needed comparing the relative worth of these biologics that mab in histologically confirmed idiopathic inflammatory myo-
sitis: a case series. Clin Rheumatol. 2015;34(2):371–7.
are currently available in most countries. These trials will 15. Deligny C, Go€eb V, Dueymes M, Kahn V, Dehlinger V,
help provide better evidence-based recommendations for the Baptiste GJ, et al. Rituximab for patients with myopathy associ-
best biologics for recalcitrant JDM. There may be new bio- ated with anti-signal recognition particle antibodies: comment
logics in the next 5–10 years against other targets that will on the article by Valiyil et al. Arthritis Care Res (Hoboken).
be more effective than the biologics available now. 2011;63(3):460; author reply 461.
16. Mitchell C, Crayne CB, Cron RQ. Patterns of B cell repletion
following rituximab therapy in a pediatric rheumatology cohort.
ACR Open Rheumatol. 2019;1(8):527–32.
Conflict of interest 17. Rider LG, Yip AL, Horkayne-Szakaly I, Volochayev R, Shrader
None. JA, Turner ML, et al. Novel assessment tools to evaluate clinical
and laboratory responses in a subset of patients enrolled in the
Rituximab in Myositis trial. Clin Exp Rheumatol. 2014;32:
ORCID 689–96.
18. Bader-Meunier B, Decaluwe H, Barnerias C, Gherardi R,
Anjali Patwardhan http://orcid.org/0000-0002-1467-1011 Quartier P, Faye A, et al. Safety and efficacy of rituximab in
severe juvenile dermatomyositis: results from 9 patients from
the French Autoimmunity and Rituximab registry. J
References Rheumatol. 2011;38(7):1436–40.
19. Sultan SM, Ng KP, Edwards JC, Isenberg DA, Cambridge G.
1. Dalakas MC. Pathophysiology of inflammatory and auto- Clinical outcome following B cell depletion therapy in eight
immune myopathies. Presse Med. 2011;40(4 Pt 2):e237–e247. patients with refractory idiopathic inflammatory myopathy.
2. Hornung T, Wenzel J. Innate immune-response mechanisms in Clin Exp Rheumatol. 2008;26(5):887–93.
dermatomyositis: an update on pathogenesis, diagnosis and 20. Valiyil R, Casciola-Rosen L, Hong G, Mammen A,
treatment. Drugs. 2014;74(9):981–98. Christopher-Stine L. Rituximab therapy for myopathy associ-
3. Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, ated with anti-signal recognition particle antibodies: a case ser-
Tawil R, et al. Interferon-alpha/beta-mediated innate immune ies. Arthritis Care Res (Hoboken). 2010;62(9):1328–34.
mechanisms in dermatomyositis. Ann Neurol. 2005;57(5): 21. Whelan BR, Isenberg DA. Poor response of anti-SRP-positive
664–78. idiopathic immune myositis to B-cell depletion. Rheumatology
4. Takada K, Nagasaka K, Miyasaka N. Polymyositis/dermatomyo- (Oxford). 2009;48(5):594–5.
sitis and interstitial lung disease: a new therapeutic approach 22. Aggarwal R, Bandos A, Reed AM, Ascherman DP, Barohn RJ,
with T-cell-specific immunosuppressants. Autoimmunity. 2005; Feldman BM, et al., the RIM Study Group. Predictors of clin-
38(5):383–92. ical improvement in rituximab-treated refractory adult and
5. Kotani T, Makino S, Takeuchi T, Kagitani M, Shoda T, Hata A, juvenile dermatomyositis and adult polymyositis. Arthritis
et al. Early intervention with corticosteroids and cyclosporin A Rheumatol. 2014;66(3):740–9.
and 2-hour postdose blood concentration monitoring improves 23. Limaye V, Hissaria P, Liew CL, Koszyka B. Efficacy of rituxi-
the prognosis of acute/subacute interstitial pneumonia in derm- mab in refractory antisynthetase syndrome. Intern Med J. 2012;
atomyositis. J Rheumatol. 2008;35(2):254–9. 42(3):e4–e7.
14 A. PATWARDHAN AND C. H. SPENCER

24. Fasano S, Gordon P, Hajji R, Loyo E, Isenberg DA. Rituximab 41. Isenberg D, Gordon C, Licu D, Copt S, Rossi CP, Wofsy D.
in the treatment of inflammatory myopathies: a review. Efficacy and safety of atacicept for prevention of flares in
Rheumatology (Oxford). 2017;56(1):26–36. patients with moderate-to-severe systemic lupus erythematosus
25. Reed AM, Crowson CS, Hein M, de Padilla CL, Olazagasti JM, (SLE): 52-week data (APRIL-SLE randomised trial). Ann
Aggarwal R, et al., the RIM Study Group. Biologic predictors of Rheum Dis. 2015;74(11):2006–15.
clinical improvement in rituximab-treated refractory myositis. 42. Lenert A, Niewold TB, Lenert P. Spotlight on blisibimod and
BMC Musculoskelet Disord. 2015;16(1):257. its potential in the treatment of systemic lupus erythematosus:
26. Aggarwal R, Oddis CV, Goudeau D, Koontz D, Qi Z, Reed evidence to date. Drug Des Devel Ther. 2017; 11:747–57.
AM, et al. Autoantibody levels in myositis patients correlate 43. Hu Y, Turner MJ, Shields J, Gale MS, Hutto E, Roberts BL,
with clinical response during B cell depletion with rituximab. et al. Investigation of the mechanism of action of alemtuzumab
Rheumatology (Oxford). 2016;55(6):991–9. in a human CD52 transgenic mouse model. Immunology. 2009;
27. Lopez De Padilla CM, Crowson CS, Hein MS, Strausbauch 128(2):260–70.
MA, Aggarwal R, Levesque MC, Reed AM, et al. Interferon- 44. Buggins AG, Mufti GJ, Salisbury J, Codd J, Westwood N, Arno
regulated chemokine score associated with improvement in dis- M, et al. Peripheral blood but not tissue dendritic cells express
ease activity in refractory myositis patients treated with rituxi- CD52 and are depleted by treatment with alemtuzumab. Blood.
mab. Clin Exp Rheumatol. 2015;33(5):655–63. 2002;100(5):1715–20.
28. Craxton A, Magaletti D, Ryan EJ, Clark EA. Macrophage- and 45. Dalakas MC, Rakocevic G, Schmidt J, Salajegheh M, McElroy
dendritic cell-dependent regulation of human B-cell prolifer- B, Harris-Love MO, et al. Effect of Alemtuzumab (CAMPATH
ation requires the TNF family ligand BAFF. Blood 2003; 1-H) in patients with inclusion-body myositis. Brain. 2009;
101(11):4464–71. 132(6):1536–44.
29. Mackay F, Browning JL. BAFF: a fundamental survival factor 46. Brett S, Baxter G, Cooper H, Johnston JM, Tite J, Rapson N.
for B cells. Nat Rev Immunol. 2002;2(7):465–75. Repopulation of blood lymphocyte sub-populations in rheuma-
30. Szodoray P, Alex P, Knowlton N, Centola M, Dozmorov I, toid arthritis patients treated with the depleting humanized
Csipo I, et al. Idiopathic inflammatory myopathies, signified by monoclonal antibody, CAMPATH-1H. Immunology. 1996;
distinctive peripheral cytokines, chemokines and the TNF fam- 88(1):13–9.
ily members B-cell activating factor and a proliferation induc- 47. Cox AL, Thompson SA, Jones JL, Robertson VH, Hale G,
ing ligand. Rheumatology (Oxford). 2010;49(10):1867–77. Waldmann H, Coles AJ, et al. Lymphocyte homeostasis follow-
31. Zouali M, Richard Y. Marginal zone B-cells, a gatekeeper of ing therapeutic lymphocyte depletion in multiple sclerosis. Eur
innate immunity. Front Immunol. 2011;2:63. J Immunol. 2005;35(11):3332–42.
32. Gupta L, Chaurasia S, Srivastava P, Dwivedi S, Lawrence A, 48. Schmidt K, Kleinschnitz K, Rakocevic G, Dalakas MC, Schmidt
Misra R. Serum BAFF in Indian patients with IIM: a retro- J. Molecular treatment effects of alemtuzumab in skeletal
muscles of patients with IBM. BMC Neurol. 2016;16:48.
spective study reveals novel clinico-phenotypic associations in
49. Sa J, Costelha J, Marinho A. Inclusion body myositis treated
children and adults. Clin Rheumatol. 2018;37(5):1265–71.
with alemtuzumab. Eur J Case Rep Intern Med. 2019;6(12):
33. Krystufkova O, Vallerskog T, Helmers SB, Mann H, Putova I,
001368.
Belacek J, et al. Increased serum levels of B cell activating factor
50. Ruck T, Bittner S, Kuhlmann T, Wiendl H, Meuth SG. Long-
(BAFF) in subsets of patients with idiopathic inflammatory
term efficacy of alemtuzumab in polymyositis. Rheumatology
myopathies. Ann Rheum Dis. 2009;68(6):836–43.
(Oxford). 2015;54(3):560–2.
34. Lopez De Padilla CM, McNallan KT, Crowson CS, Bilgic H,
51. Thompson B, Corris P, Miller JAL, Cooper RG, Halsey JP,
Bram RJ, Hein MS, et al. BAFF expression correlates with idio-
Isaacs JD. Alemtuzumab (Campath-1H) for treatment of refrac-
pathic inflammatory myopathy disease activity measures and
tory polymyositis. J Rheumatol. 2008;35(10):2080–2.
autoantibodies. J Rheumatol. 2013;40(3):294–302.
52. Reiff A, Shaham B, Weinberg KI, Crooks GM, Parkman R.
35. Kobayashi N, Kobayashi I, Mori M, Sato S, Iwata N, Shigemura
Anti-CD52 antibody-mediated immune ablation with autolo-
T, et al. Increased serum B cell activating factor and a prolifer- gous immune recovery for the treatment of refractory juvenile
ation-inducing ligand are associated with interstitial lung dis- polymyositis. J Clin Immunol. 2011;31(4):615–22.
ease in patients with juvenile dermatomyositis. J Rheumatol. 53. Aouad P, Yiannikas C, Fernando SL, Parratt J. A case of auto-
2015;42(12):2412–8. immune myositis after treatment with alemtuzumab for mul-
36. Morais SA, Vilas-Boas A, Isenberg DA. B-cell survival factors tiple sclerosis. Mult Scler J Exp Transl Clin. 2018;4(4):
in autoimmune rheumatic disorders. Ther Adv Musculoskelet 2055217318819012.
Dis. 2015;7(4):122–51. 54. Wray S, Havrdova E, Snydman DR, Arnold DL, Cohen JA,
37. Isenberg DA, Petri M, Kalunian K, Tanaka Y, Urowitz MB, Coles AJ, et al. Infection risk with alemtuzumab decreases over
Hoffman RW, et al. Efficacy and safety of subcutaneous tabalu- time: pooled analysis of 6-year data from the CAMMS223,
mab in patients with systemic lupus erythematosus: results CARE-MS I, and CARE-MS II studies and the CAMMS03409
from ILLUMINATE-1, a 52-week, phase III, multicentre, rand- extension study. Mult Scler. 2019;25(12):1605–17.
omised, double-blind, placebo-controlled study. Ann Rheum 55. Martin SI, Marty FM, Fiumara K, Treon SP, Gribben JG,
Dis. 2016;75(2):323–31. Baden LR. Infectious complications associated with alemtuzu-
38. Hoffman RW, Merrill JT, Alarcon-Riquelme MM, Petri M, mab use for lymphoproliferative disorders. Clin Infect Dis.
Dow ER, Nantz E, et al. Gene expression and pharmacody- 2006;43(1):16–24.
namic changes in 1,760 systemic lupus erythematosus patients 56. Pachman LM, Liotta-Davis MR, Hong DK, Kinsella TR,
from two phase III trials of BAFF blockade with tabalumab. Mendez EP, Kinder JM, Chen EH. TNFalpha-308A allele in
Arthritis Rheumatol. 2017;69(3):643–54. juvenile dermatomyositis: association with increased production
39. Merrill JT, Wallace DJ, Wax S, Kao A, Fraser PA, Chang P, of tumor necrosis factor alpha, disease duration, and pathologic
Isenberg D, et al., ADDRESS II Investigators. Efficacy and calcifications. Arthritis Rheum. 2000;43(10):2368–77.
safety of atacicept in patients with systemic lupus erythemato- 57. Salomonsson S, Lundberg IE. Cytokines in idiopathic inflam-
sus: results of a twenty-four-week, multicenter, randomized, matory myopathies. Autoimmunity. 2006;39(3):177–90.
double-blind, placebo-controlled, parallel-arm, phase IIb study. 58. Mielnik P, Chwalinska-Sadowska H, Wiesik-Szewczyk E,
Arthritis Rheumatol. 2018;70(2):266–76. Maslinski W, Olesinska M. Serum concentration of interleukin
40. Kaegi C, Steiner UC, Wuest B, Crowley C, Boyman O. 15, interleukin 2 receptor and TNF receptor in patients with
Systematic review of safety and efficacy of atacicept in treating polymyositis and dermatomyositis: correlation to disease activ-
immune-mediated disorders. Front Immunol. 2020;11:433. ity. Rheumatol Int. 2012;32(3):639–43.
MODERN RHEUMATOLOGY 15

59. Pachman LM, Fedczyna TO, Lechman TS, Lutz J. Juvenile crohn disease successfully treated with adalimumab. Case Rep
dermatomyositis: the association of the TNF alpha-308A allele Gastroenterol. 2016;10(3):661–7.
and disease chronicity. Curr Rheumatol Rep. 2001;3(5):379–86. 77. Verma S, Kroeker KI, Fedorak RN. Adalimumab for orbital
60. De Paepe B, Creus KK, De Bleecker JL. The tumor necrosis fac- myositis in a patient with Crohn’s disease who discontinued
tor superfamily of cytokines in the inflammatory myopathies: infliximab: a case report and review of the literature. BMC
potential targets for therapy. Clin Dev Immunol. 2012;2012: Gastroenterol. 2013;13:59.
369432. 78. Adams AB, Kazim M, Lehman TJ. Treatment of orbital myo-
61. Chinoy H, Salway F, John S, Fertig N, Tait BD, Oddis CV, sitis with adalimumab (Humira). J Rheumatol. 2005;32(7):
et al., UK Adult Onset Myositis Immunogenetic Collaboration 1374–5.
(AOMIC). Tumour necrosis factor-alpha single nucleotide poly- 79. Ayabe R, Rootman DB, Hwang CJ, Ben-Artzi A, Goldberg R.
morphisms are not independent of HLA class I in UK Adalimumab as steroid-sparing treatment of inflammatory-
Caucasians with adult onset idiopathic inflammatory myopa- stage thyroid eye disease. Ophthalmic Plast Reconstr Surg.
thies. Rheumatology (Oxford). 2007;46(9):1411–6. 2014; 30:415–9.
62. Lutz J, Huwiler KG, Fedczyna T, Lechman TS, Crawford S, 80. Park JK, Yoo HG, Ahn DS, Jeon HS, Yoo WH. Successful
Kinsella TR, Pachman LM. Increased plasma thrombospondin- treatment for conventional treatment-resistant dermatomyo-
1 (TSP-1) levels are associated with the TNF alpha-308A allele sitis-associated interstitial lung disease with adalimumab.
in children with juvenile dermatomyositis. Clin Immunol. 2002; Rheumatol Int. 2012;32(11):3587–90.
103(3):260–3. 81. da Silva TC, Zon Pretti F, Shinjo SK. Adalimumab in anti-syn-
63. Brunasso AM, Aberer W, Massone C. New onset of dermato- thetase syndrome. Joint Bone Spine. 2013;80(4):432.
myositis/polymyositis during anti-TNF-a therapies: a systematic 82. Hengstman GJ, van den Hoogen FH, Barrera P, Netea MG,
literature review . Scientificworldjournal. 2014;2014:179180. Pieterse A, van de Putte LB, van Engelen BG. Successful treat-
64. Niewold TB, Kariuki SN, Morgan GA, Shrestha S, Pachman ment of dermatomyositis and polymyositis with anti-tumor-
LM. Gene-gene-sex interaction in cytokine gene polymorphisms necrosis-factor-alpha: preliminary observations. Eur Neurol.
revealed by serum interferon alpha phenotype in juvenile derm- 2003;50(1):10–5.
atomyositis. J Pediatr. 2010;157(4):653–7. 83. Uthman I, El-Sayad J. Refractory polymyositis responding to
65. Schiffenbauer A, Garg M, Castro C, Pokrovnichka A, Joe G, infliximab. Rheumatology (Oxford). 2004;43(9):1198–9.
Shrader J, et al. A randomized, double-blind, placebo-controlled 84. Dastmalchi M, Grundtman C, Alexanderson H, Mavragani CP,
trial of infliximab in refractory polymyositis and dermatomyo- Einarsdottir H, Helmers SB, et al. A high incidence of disease
sitis. Semin Arthritis Rheum. 2018;47(6):858–64. flares in an open pilot study of infliximab in patients with
66. Efthimiou P, Schwartzman S, Kagen LJ. Possible role for refractory inflammatory myopathies. Ann Rheum Dis. 2008;
tumour necrosis factor inhibitors in the treatment of resistant 67(12):1670–7.
dermatomyositis and polymyositis: a retrospective study of 85. Rouster-Stevens KA, Ferguson L, Morgan G, Huang CC,
eight patients. Ann Rheum Dis. 2006;65(9):1233–6. Pachman LM. Pilot study of etanercept in patients with refrac-
67. Muscle Study Group. A randomized, pilot trial of etanercept in tory juvenile dermatomyositis. Arthritis Care Res (Hoboken).
dermatomyositis. Ann Neurol 2011;70:427–36. 2014;66(5):783–7.
68. Tosounidou S, MacDonald H, Situnayake D. Successful treat- 86. Ramos-Casals M, Brito-Zeron P, Munoz S, Soria N, Galiana D,
ment of calcinosis with infliximab in a patient with systemic Bertolaccini L, et al. Autoimmune diseases induced by TNF-tar-
sclerosis/myositis overlap syndrome. Rheumatology (Oxford). geted therapies: analysis of 233 cases. Medicine (Baltimore).
2014;53(5):960–1. 2007;86(4):242–51.
69. Kohsaka H, Mimori T, Kanda T, Shimizu J, Sunada Y, 87. Brunasso AM, Laimer M, Massone C. Paradoxical reactions to
Fujimoto M, et al. Treatment consensus for management of targeted biological treatments: a way to treat and trigger? Acta
polymyositis and dermatomyositis among rheumatologists, neu- Derm Venereol. 2010;90(2):183–5.
rologists and dermatologists. Mod Rheumatol. 2019;29(1):1–19. 88. Fiorentino DF. The Yin and Yang of TNF-{alpha} inhibition.
70. Liu SW, Velez NF, Lam C, Femia A, Granter SR, Townsend Arch Dermatol. 2007;143(2):233–6.
HB, Vleugels RA. Dermatomyositis induced by anti-tumor 89. Zengin O, Onder ME, Alkan S, Kimyon G, Huseynova N,
necrosis factor in a patient with juvenile idiopathic arthritis. Demir ZH, et al. Three cases of anti-TNF induced myositis and
JAMA Dermatol. 2013;149(10):1204–8. literature review. Rev Bras Reumatol Engl Ed. 2016;57:590–5.
71. de Souza FH, Barros TB, Levy-Neto M, Shinjo SK. 90. Ishikawa Y, Yukawa N, Ohmura K, Hosono Y, Imura Y,
Adalimumab induced-inflammatory myopathy in rheumatoid Kawabata D, et al. Etanercept-induced anti-Jo-1-antibody-posi-
arthritis. Acta Reumatol Port. 2012;37(2):180–3. tive polymyositis in a patient with rheumatoid arthritis: a case
72. Nagashima T, Minota S. Dermatomyositis in patients with report and review of the literature. Clin Rheumatol. 2010;29(5):
rheumatoid arthritis during adalimumab therapy. J Rheumatol. 563–6.
2011;38(3):574; author reply 575. 91. Murata K, Dalakas MC. Expression of the costimulatory mol-
73. Liozon E, Ouattara B, Loustaud-Ratti V, Vidal E. Severe poly- ecule BB-1, the ligands CTLA-4 and CD28, and their mRNA in
myositis and flare in autoimmunity following treatment with inflammatory myopathies. Am J Pathol. 1999;155(2):453–60.
adalimumab in a patient with overlapping features of polyar- 92. Nagaraju K, Raben N, Villalba ML, Danning C, Loeffler LA,
thritis and scleroderma. Scand J Rheumatol. 2007;36(6):484–6. Lee E, et al. Costimulatory markers in muscle of patients with
74. Campanilho-Marques R, Deakin CT, Simou S, Papadopoulou idiopathic inflammatory myopathies and in cultured muscle
C, Wedderburn LR, Pilkington CA, for the Juvenile cells. Clin Immunol. 1999;92(2):161–9.
Dermatomyositis Research Group (JDRG). Retrospective ana- 93. Khanna S, Reed AM. Immunopathogenesis of juvenile derm-
lysis of infliximab and adalimumab treatment in a large cohort atomyositis. Muscle Nerve. 2010;41(5):581–92.
of juvenile dermatomyositis patients. Arthritis Res Ther. 2020; 94. Herrero-Beaumont G, Martınez Calatrava MJ, Casta~ neda S.
22(1):79. Abatacept mechanism of action: concordance with its clinical
75. Hernandez-Garfella ML, Gracia-Garcia A, Cervera-Taulet E, profile. Reumatol Clin. 2012;8(2):78–83.
Garcia-Villanueva C, Montero HJ. Adalimumab for recurrent 95. Kerola AM, Kauppi MJ. Abatacept as a successful therapy for
orbital myositis in Crohn’s disease: report of a case with a 3- myositis—a case-based review. Clin Rheumatol. 2015;34(3):
year follow-up. J Crohns Colitis. 2011;5(3):265–6. 609–12.
76. Vadala di Prampero S, Marino M, Toso F, Avellini C, Nguyen 96. Maeshima K, Kiyonaga Y, Imada C, Iwakura M, Hamasaki H,
V, Sorrentino D. Isolated bilateral gastrocnemius myositis in Haranaka M, Ishii K. Successful treatment of refractory anti-
16 A. PATWARDHAN AND C. H. SPENCER

signal recognition particle myopathy using abatacept. treated with tocilizumab: a hopeful treatment strategy for
Rheumatology (Oxford). 2014;53(2):379–80. refractory dermatomyositis? Rheumatology (Oxford). 2014;
97. Musuruana JL, Cavallasca JA. Abatacept for treatment of 53(10):1907–8.
refractory polymyositis. Joint Bone Spine. 2011;78(4):431–2. 112. Umeda M, Aramaki T, Fujikawa K, Iwamoto N, Ichinose K,
98. Tjarnlund A, Tang Q, Wick C, Dastmalchi M, Mann H, Terada K, et al. Tocilizumab is effective in a familial
Tomasova Studynkova J, et al. Abatacept in the treatment of Mediterranean fever patient complicated with histologically
adult dermatomyositis and polymyositis: a randomised, phase proven recurrent fasciitis and myositis. Int J Rheum Dis. 2017;
IIb treatment delayed-start trial. Ann Rheum Dis. 2018;77(1): 20(11):1868–71.
55–62. 113. Raine C, Hamdulay SS, Khanna M, Boyer L, Kinderlerer A. An
99. Grundtman C, Salomonsson S, Dorph C, Bruton J, Andersson unusual complication of tocilizumab therapy: MRI appearances
U, Lundberg IE. Immunolocalization of interleukin-1 receptors of thenar eminence pyomyositis. Joint Bone Spine. 2013;80(2):
in the sarcolemma and nuclei of skeletal muscle in patients
222.
with idiopathic inflammatory myopathies. Arthritis Rheum.
114. Notarnicola A, Lapadula G, Natuzzi D, Lundberg IE, Iannone
2007;56(2):674–87.
F. Correlation between serum levels of IL-15 and IL-17 in
100. Zong M, Dorph C, Dastmalchi M, Alexanderson H, Pieper J,
patients with idiopathic inflammatory myopathies. Scand J
Amoudruz P, et al. Anakinra treatment in patients with refrac-
tory inflammatory myopathies and possible predictive response Rheumatol. 2015;44(3):224–8.
biomarkers: a mechanistic study with 12 months follow-up. 115. Moran EM, Mastaglia FL. The role of interleukin-17 in
Ann Rheum Dis. 2014;73(5):913–20. immune-mediated inflammatory myopathies and possible
101. Kosmidis ML, Alexopoulos H, Tzioufas AG, Dalakas MC. The therapeutic implications. Neuromuscul Disord. 2014;24(11):
effect of anakinra, an IL1 receptor antagonist, in patients with 943–52.
sporadic inclusion body myositis (sIBM): a small pilot study. J 116. Moghadam-Kia S, Charlton D, Aggarwal R, Oddis CV.
Neurol Sci. 2013;334(1–2):123–5. Management of refractory cutaneous dermatomyositis: potential
102. Svensson J, Holmqvist M, Tj€arnlund A, Dastmalchi M, Hanna role of Janus kinase inhibition with tofacitinib. Rheumatology
B, Magnusson Bucher S, Lundberg IE. Use of biologic agents in (Oxford). 2019;58(6):1011–5.
idiopathic inflammatory myopathies in Sweden: a descriptive 117. Kurtzman DJ, Wright NA, Lin J, Femia AN, Merola JF, Patel
study of real life treatment. Clin Exp Rheumatol. 2017;35(3): M, Vleugels RA. Tofacitinib citrate for refractory cutaneous
512–5. dermatomyositis: an alternative treatment. JAMA Dermatol.
103. Furlan A, Botsios C, Ruffatti A, Todesco S, Punzi L. 2016;152(8):944–5.
Antisynthetase syndrome with refractory polyarthritis and fever 118. Navarro-Navarro I, Jimenez-Gallo D, Rodrıguez-Mateos ME,
successfully treated with the IL-1 receptor antagonist, anakinra: Rodrıguez-Hernandez C, Linares-Barrios M. Treatment of
a case report. Joint Bone Spine. 2008;75(3):366–7. refractory anti-NXP2 and anti-TIF1c dermatomyositis with
104. Scuderi F, Mannella F, Marino M, Provenzano C, Bartoccioni tofacitinib. J Dtsch Dermatol Ges. 2020.
E. IL-6-deficient mice show impaired inflammatory response in 119. Sabbagh S, Almeida de Jesus A, Hwang S, Kuehn HS, Kim H,
a model of myosin-induced experimental myositis. J Jung L, et al. Treatment of anti-MDA5 autoantibody-positive
Neuroimmunol. 2006;176(1-2):9–15. juvenile dermatomyositis using tofacitinib. Brain. 2019;142(11):
105. Okiyama N, Sugihara T, Iwakura Y, Yokozeki H, Miyasaka N, e59.
Kohsaka H. Therapeutic effects of interleukin-6 blockade in a 120. Landon-Cardinal O, Benveniste O, Allenbach Y. Reply:
murine model of polymyositis that does not require interleu- Treatment of anti-MDA5 autoantibody-positive juvenile derm-
kin-17A. Arthritis Rheum. 2009;60(8):2505–12. atomyositis using tofacitinib. Brain. 2019;142(11):e60.
106. Yang M, Cen X, Xie Q, Zuo C, Shi G, Yin G. Serum interleu- 121. Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermato-
kin-6 expression level and its clinical significance in patients
myositis-associated interstitial lung disease. N Engl J Med.
with dermatomyositis. Clin Dev Immunol. 2013;2013:717808.
2019;381(3):291–3.
107. Tournadre A, Miossec P. Interleukin-17 in inflammatory myo-
122. John Hopkins University. Study of Tofacitinib in Refractory
pathies. Curr Rheumatol Rep. 2012;14(3):252–6.
Dermatomyositis (STIR). 2017 [cited 2020 Oct 09]. Available
108. Murphy SM, Lilleker JB, Helliwell P, Chinoy H. The successful
use of tocilizumab as third-line biologic therapy in a case of from: https://clinicaltrials.gov/ct2/show/NCT03002649?term=
refractory anti-synthetase syndrome. Rheumatology (Oxford). Tofacitinib&cond=dermatomyositis&draw=2&rank=1
2016;55(12):2277–8. 123. Wendel S, Venhoff N, Frye BC, May AM, Agarwal P, Rizzi M,
109. Beaumel A, Muis-Pistor O, Tebib JG, Coury F. Antisynthetase et al. Successful treatment of extensive calcifications and acute
syndrome treated with tocilizumab. Joint Bone Spine. 2016; pulmonary involvement in dermatomyositis with the Janus-
83(3):361–2. Kinase inhibitor tofacitinib – a report of two cases. J
110. Narazaki M, Hagihara K, Shima Y, Ogata A, Kishimoto T, Autoimmun. 2019;100:131–6.
Tanaka T. Therapeutic effect of tocilizumab on two patients 124. Safina BS, Baker S, Baumgardner M, Blaney PM, Chan BK,
with polymyositis. Rheumatology (Oxford). 2011;50(7):1344–6. Chen YH, et al. Discovery of novel PI3-kinase d specific inhibi-
111. Kondo M, Murakawa Y, Matsumura T, Matsumoto O, Taira tors for the treatment of rheumatoid arthritis: taming CYP3A4
M, Moriyama M, et al. A case of overlap syndrome successfully time-dependent inhibition. J Med Chem. 2012;55(12):5887–900.

You might also like