Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Department Pharmacology Continuing Education

Juvenile Idiopathic
Arthritis: A Focus on
Pharmacologic
Management
Jessica L. Jacobson, PharmD, BCPS, BCPPS, &
Jennifer T. Pham, PharmD, BCPS, BCPPS

ABSTRACT KEY WORDS


Juvenile idiopathic arthritis is a chronic condition that affects Juvenile idiopathic arthritis, pediatrics, pharmacotherapy
many pediatric patients. It is a prevalent disease and has
become the most common rheumatologic disease of child-
hood. The condition encompasses multiple different forms
of chronic arthritides classified based on the location and OBJECTIVES
number of joints affected as well as the presence or lack of 1. Describe the pathophysiology, clinical presentation and
a number of different inflammatory markers. The exact eti- diagnosis of juvenile idiopathic arthritis (JIA).
ology is unknown but is thought to be multifactorial with 2. Discuss the use of nonsteroidal anti-inflammatory drugs,
genetic, humoral, and environmental factors playing a key glucocorticoids, and non-biologic disease modifying anti-
role. Many pharmacologic agents are available for use in the rheumatic drugs in the management of JIA.
treatment of juvenile idiopathic arthritis, with management 3. Explain the role of biologic disease modifying anti-
involving the use of symptom-reducing agents and disease- rheumatic drugs in the management of JIA.
modifying antirheumatic drugs. Treatment is not without 4. Recommend a treatment plan based on classifica-
adverse events, with many of the agents require monitor- tion of JIA and disease activity.
ing regimens and patient education. Without treatment, the 5. Identify monitoring parameters and adverse effects as-
progression and chronicity of the disease can result in sig- sociated with pharmacologic treatment.
nificant morbidity, with the potential for devastating
consequences on the child’s quality of life. J Pediatr Health
Care. (2018) 32, 515-528.
INTRODUCTION
Juvenile idiopathic arthritis (JIA) is the most common
Jessica L. Jacobson, Clinical Pharmacy Specialist-PICU/Pediatrics, rheumatologic disease and has the potential to cause
Department of Pharmacy, Rush University Medical Center, Chicago,
long-term morbidity and physical disability. Older terms,
IL.
including juvenile rheumatoid arthritis, which is used
Jennifer T. Pham, Clinical Pharmacist–Neonatal/Pediatrics and Clini- more commonly in the United States, and juvenile
cal Assistant Professor, Department of Pharmacy Practice, University
of Illinois at Chicago, Chicago, IL.
chronic arthritis, which is preferred in Europe, were
replaced by the term JIA at the meeting of the Inter-
Conflicts of interest: None to report.
national League of Association for Rheumatology (ILAR)
Correspondence: Jennifer T. Pham, PharmD, BCPS, BCPPS, in the late 1990s (European League Against Rheumatism,
Department of Pharmacy Practice, University of Illinois at 1977). The ILAR highlighted the onset of JIA that begins
Chicago, College of Pharmacy, 833 S. Wood St., M/C 886,
during childhood and provided a clear distinction from
Room 164, Chicago, IL 60612; e-mail: Tran@uic.edu
adult-onset rheumatoid arthritis. JIA in itself is not a
0891-5245/$36.00
disease but rather a term that encompasses all forms
Copyright © 2018 by the National Association of Pediatric Nurse of arthritides. It is defined as arthritis of unknown eti-
Practitioners. Published by Elsevier Inc. All rights reserved. ology that begins before the 16th birthday and persists
https://doi.org/10.1016/j.pedhc.2018.02.005 for at least 6 weeks.

www.jpedhc.org September/October 2018 515


JIA is a diagnosis of exclusion, so other conditions siblings having a 15- to 30-fold higher prevalence of
that present with similar clinical manifestations need JIA compared with the normative population (Woo &
to be ruled out, including connective tissue diseases Colbert, 2009). The IL2RA/CD25 and VTCN1 genes have
(e.g., systemic lupus erythematosus), trauma, infec- been proposed to confer susceptibility toward the de-
tion (e.g., septic joints, osteomyelitis, viral illnesses, Lyme velopment of JIA (Hinks et al., 2009). In patients with
disease), and malignancies. Therefore, the diagnosis ERA, test results for human leukocyte antigen (HLA)-
of JIA is usually prolonged, which can delay the ini- B27 are commonly positive, and HLA-B27 has been
tiation of treatment, resulting in devastating consequences correlated with the development of inflammation of
such as permanent joint destruction, joint contractures, the axial skeleton, specifically involving the hip (Woo
leg-length discrepancies, and blindness secondary to & Colbert, 2009). Several environmental factors, in-
chronic uveitis. Recent advances in pharmacologic treat- cluding breastfeeding and vitamin D and sun exposure,
ment have significantly improved the outcome of JIA have been identified as having a protective effect against
in children. the development of JIA. In contrast, environmental
factors such as infection and maternal smoking may
EPIDEMIOLOGY increase the risk of developing or worsening the disease
JIA is the most common pediatric autoimmune mus- (Ellis, Munro, & Ponsonby, 2010). Further studies are
culoskeletal condition. Although the exact incidence needed to comprehensively examine these environ-
of JIA in the United States is unknown, the incidence mental hypotheses.
is estimated to be about 2 to 20 per 100,000 children; Humoral and cell-mediated immunity are involved
the prevalence is about 16 to 150 per 100,000 chil- in the pathogenesis of JIA. Cell-mediated release of
dren in Europe and North America (Giancane et al., proinflammatory cytokines, such as tumor necrosis
2016). Oligoarthritis is the most common subtype of factor–alpha (TNF-α), interleukin (IL)-6, and IL-1 sec-
JIA in North America, with rheumatoid factor–positive ondary to T-cell activation, continues to be the best
(Rf+) polyarthritis being the least common subtype described pathogenesis of JIA and is the focus of many
(Giancane et al., 2016). The disease more commonly newer pharmacologic agents. Radiographic models have
affects females than males (> 2:1); however, sex dis- confirmed this etiology by evidence of a higher per-
tribution varies with disease subtype (Giancane et al., centage of activated T cells in the synovium of patients
2016; Minden et al., 2002). Age at onset of disease also with JIA. In addition, diagnostic tests show that pa-
differs among subtypes, with a median age of onset tients with polyarticular and systemic JIA (SJIA) have
of 4 years in oligoarthritis, 11 years in enthesitis- higher levels of TNF-α, IL-6, and IL-1 compared with
related arthritis (ERA), and 12 years in Rf+ polyarthritis other JIA subtypes (Borchers et al., 2006; Espinosa &
(Minden et al., 2002). In addition, ethnicity can affect Gottlieb, 2012). Activation of the humoral immune re-
the prevalence of disease subtype, highlighting the po- sponse is evident through the production of
tential for involvement of genetic predisposition autoantibodies such as antinuclear antibodies (ANA),
(Schwarz, Simpson, Kerr, & Jarvis, 1997). For example, as well as complement activation and an increase in
African Americans are more likely to develop polyar- serum immunoglobulins (Igs), such as IgM rheuma-
ticular and Rf+ disease (Espinosa & Gottlieb, 2012), toid factor. It is estimated that ANAs are detected in
oligoarthritis is the most common JIA subtype in White 30% to 50% of patients with JIA, and detection has a
children of European descent, and enthesitis-related positive correlation with development of uveitis, a com-
arthritis tends to be more prevalent in children of plication of JIA (Borchers et al., 2006).
Mexican and Asian descent (Woo & Colbert, 2009).
CLINICAL PRESENTATION AND DIAGNOSIS
PATHOPHYSIOLOGY The exact etiology The clinical manifestation and treatment algorithms of
The exact etiology and JIA depend on the subtype of arthritis that is present.
pathogenesis of JIA are
and pathogenesis Arthritis must be present for 6 weeks before the di-
unknown, although of JIA are agnosis of JIA can be made. Morning stiffness or gelling
genetic, environmen- unknown, although phenomenon (i.e., stiffness after long periods of sitting
tal, and autoimmune or inactivity) are common complaints. Arthralgia often
factors are thought to
genetic, occurs during the morning, with improvements through-
play a role in the de- environmental, and out the day. Different classifications of JIA have been
velopment of this autoimmune developed by organizations including the American
disease. A genetic pre- College of Rheumatology (ACR), EULAR, and ILAR
disposition for JIA has
factors are thought (Beukelman et al., 2011; EULAR, 1977; Petty et al., 2004;
been suggested through to play a role in the Ringold et al., 2013). The progression of the disease
concordance rates of development of nomenclature and their differing classifications are sum-
25% to 40% in mono- marized in Table 1. The ILAR 2001 classification was
zygotic twins and
this disease. developed in response to a need for a more consis-

516 Volume 32 • Number 5 Journal of Pediatric Health Care


TABLE 1. Progression of characteristics/classification systems of JIA
Characteristics ACR (1977) EULAR (1978) ILAR (1997, 2001) ACR (2011)
Nomenclature Juvenile rheumatoid arthritis Juvenile chronic arthritis Juvenile idiopathic arthritis Juvenile idiopathic arthritis
Age at onset <16 years <16 years <16 years <16 years
Duration of arthritis ≥6 weeks ≥3 months ≥6 weeks ≥6 weeks
Classifications Systemic Systemic Systemic Arthritis of ≤ 4
Polyarticular Polyarticular Oligoarticular joints
Pauciarticular Juvenile rheumatoid arthritis (persistent/extended) Arthritis of ≥ 5 joints
Pauciarticular Polyarticular (Rf+/Rf–) Active sacroiliac arthritis
Juvenile psoriatic arthritis Psoriatic arthritis Systemic arthritis (without
Juvenile spondylitis ERA active systemic features)
Undifferentiated Systemic arthritis with active
systemic features (without
active arthritis)
Note. Data from EULAR (1977), Beukelman et al. (2011), and Ringold et al. (2013). ACR, American College of Rheumatology; ERA, enthesitis-
related arthritis; EULAR, European League Against Rheumatism; ILAR, International League of Associations for Rheumatology; Rf+, rheumatoid
factor positive; Rf–, Rheumatoid factor negative.

tent classification system than what had been previously tic macular, nonpruritic, salmon-colored rash (linear
defined in the ACR 1977 and EULAR 1978 classifications. macular rash) is most commonly present at time of fever
The ILAR 2001 classification identified six distinct and is typically distributed over the trunk and extremi-
subtypes of JIA based on the pattern and number of ties. Patients with SJIA are more likely to develop
joints involved at onset of illness, specific serologic macrophage activation syndrome (MAS) and im-
markers, and systemic manifestations present during paired growth velocity.
the first 6 months of illness (Petty et al., 2004). The
ILAR classifications of JIA include systemic, oligoarticular, Oligoarthritis
polyarticular, enthesitis-related, psoriatic, and undif- Oligoarthritis is defined as arthritis affecting four or fewer
ferentiated arthritis. Diagnosis of JIA is typically based joints during the first 6 months of disease (Dannecker
on the presence of clinical symptoms and serologic & Quartier, 2009; Petty et al., 2004). This subtype is
markers associated with each classification. Accurate further classified as persistent (affecting four or fewer
classification within a subtype is important because it joints throughout the disease) or extended (affecting
offers insight to prognostic factors, outcomes, and pre- more than 4 joints after the first 6 months of disease).
ferred treatment regimens. Compared with the other subtypes, oligoarthritis appears
to have an earlier onset of illness and is more preva-
Systemic Juvenile Idiopathic Arthritis lent in females (Dannecker & Quartier, 2009). Children
SJIA is described as a triad of arthritis, remittent fever, generally appear well despite ambulating with a limp.
and typical rash (Dannecker & Quartier, 2009; Petty Most patients with oligoarthritis present with unilat-
et al., 2004). The child appears systematically ill with eral pain and swelling of one joint (typically larger,
generalized arthralgia and myalgia. Arthritis is defined lower extremity joints). Elevated erythrocyte sedimen-
as the presence of joint effusion alone or two or more tation rate (ESR) and ANA levels are common diagnostic
of the following symptoms: decreased range of motion, criteria in this subtype of JIA. Compared with other
pain or tenderness during movement, and/or in- subtypes, oligoarthritis is associated with the devel-
creased heat on palpation in one or more joint(s). Fever opment of iridocyclitis. Therefore, these patients should
is typically present for at least 2 weeks’ duration with undergo ophthalmologic examinations more fre-
a daily quotidian pattern for at least 3 days, and ar- quently than what is recommended for other patients
thritis is present in one or more joint(s) accompanied with JIA.
by one or more of the following criteria: evanescent
erythematous rash, generalized lymph node enlarge- Polyarticular JIA (Rheumatoid Factor–Negative
ment, hepatomegaly and/or splenomegaly, or serositis [Rf–]/Positive [Rf+] Polyarthritis)
(Petty et al., 2004). Muscle tenderness to palpation may Polyarticular JIA is defined as arthritis affecting five or
be observed. The classic triad does not need to occur more joints during the first 6 months of disease (Petty
at the same time, with fever and rash oftentimes oc- et al., 2004). This subtype is further classified as Rf+
curring weeks before the onset of arthritis (Dannecker (two positive Rf test results at least 3 months apart during
& Quartier, 2009; Petty et al., 2004). The characteris- the first 6 months of disease) or Rf– (a test result for

www.jpedhc.org September/October 2018 517


Rf is negative), depending on presence of the auto- management. To minimize the number of treatment
antibody within the bloodstream. Rf+ disease is thought algorithms and attempt to simplify recommendations
to have a more aggressive disease course character- for providers, the ACR task force developed their own
ized by a higher incidence of joint deformities because JIA treatment groups based on the number of joints
of the development of erosive joints and rheumatoid affected and classifications of prognosis (features of
nodules (Espinosa & Gottlieb, 2012; Petty et al., 2004). poor prognosis) and disease level activity (low, mod-
The onset of Rf+ disease tends to occur later com- erate, high; Beukelman et al., 2011; Table 2).
pared with other subtypes and predominantly affects The development of risk stratification through the
females in late childhood to early adolescence (Espinosa use of outcomes variables provides a more objective
& Gottlieb, 2012). guide to treatment and an objective method for moni-
toring response to therapy. The six outcomes variables
Enthesitis-Related Arthritis in the ACR guidelines include (a) the physician global
ERA is described as the presence of arthritis and enthesitis assessment of disease activity, which is a subjective
(inflammation of tendons and ligaments where they assessment made by the provider measured on a 10-
attach to bone) or as the presence of arthritis or enthesitis point visual analog scale; (b) parent/patient global
with at least two of the following: (a) presence of or assessment of well-being (mimics the same 10-point
history of sacroiliac joint tenderness and/or inflamma- visual analog scale but is completed by the patient/
tory lumbosacral pain; (b) presence of the HLA-B27 parent); (c) functional assessment; (d) active joint counts;
antigen; (c) onset of arthritis in males older than 6 years; (e) restrictive joint counts; and (f) presence of acute
(d) acute (symptomatic) anterior uveitis; or (e) first- phase reactants such as ESR or C-reactive protein (CRP;
degree relative with a history of ankylosing spondylitis, Beukelman et al., 2011). Recommendations/treatment
ERA, sacroiliitis with inflammatory bowel disease, Reiter’s algorithms for initiation of pharmacologic agents in JIA
syndrome, or acute anterior uveitis (Petty et al., 2004). based on treatment group, prognosis, and disease ac-
The most common symptoms include pain, swelling, tivity can be found in the ACR guidelines and will be
and tenderness with a higher incidence in males older discussed in this review.
than 8 years of age (Espinosa & Gottlieb, 2012). Although this is not part of the ACR guidelines, the
ACR Pedi 30 (Consolaro et al. 2016), which is a tool
Psoriatic Arthritis to assess response to treatment regimens, has been
Psoriatic arthritis in children is usually mild. Patients widely used throughout many clinical trials and clini-
are classified as having psoriatic arthritis if there is pres- cal practice. This tool incorporates the six ACR outcome
ence of arthritis and psoriasis or the presence of arthritis variables as part of the assessment. A response is defined
and at least two additional criteria: dactylitis (diffuse as a minimum of 30% improvement from baseline in
swelling of fingers extending beyond the joint margin), at least three of the six ACR outcome variables and
nail pitting or onycholysis, or psoriasis in a first- no more than one remaining outcome variable wors-
degree relative (Dannecker & Quartier, 2009). Children ening by greater than 30%. Similarly, ACR Pedi 50, 70,
with psoriatic arthritis are often found to have posi- 90, and 100 responses are often reported, and these
tive test results for ANAs and HLA-B27 and therefore definitions require 50%, 70%, 90%, or 100% improve-
are at a higher risk for development of uveitis ment, with no more than one variable worsening by
(Dannecker & Quartier, 2009; Espinosa & Gottlieb, 2012). 30%.
The goal of therapy in JIA is to treat the disease
Undifferentiated Arthritis by decreasing pain and limiting disability, to mini-
Undifferentiated arthritis is the least common type of mize adverse effects, and to prevent disease progression
JIA and is described as the presence of arthritis that (Harris, Kessler, & Verbsky, 2013). The goal is to achieve
does not fit into any of the other five ILAR subtypes clinical remission, defined by the ACR as satisfaction
or fits the descriptions of at least two or more of the of clinical inactive disease for at least 6 continuous
other five subtypes (Petty et al., 2004). months while taking medication or for at least 12 con-
tinuous months while not taking medication (Lovell,
DISEASE MANAGEMENT Ruperto, Giannini, & Martini, 2013; Wallace, Giannini,
Although the ILAR classifications of JIA continued to Huang, Itert, & Ruperto, 2011). The ACR defines clini-
be the most widely used classification system, vali- cal inactive disease as the lack of active arthritis, fever,
dated guidelines providing recommendations for the rash, serositis, splenomegaly, or generalized lymph-
treatment of JIA were still lacking. This deficiency led adenopathy attributable to JIA; no active uveitis; normal
to the development of the 2011 ACR Recommenda- ESR or CRP levels; and no damage progression deter-
tions for the Treatment of JIA, with revision in 2013 mined by radiographic examination.
(Beukelman et al., 2011; Ringold et al., 2013). The ACR A systematic treatment approach begins with a
guidelines do not follow the six distinct ILAR classi- simpler, conservative modality and escalates to more
fications, which can lead to a lack of consistency in complex therapies if treatment fails (Harris et al., 2013).

518 Volume 32 • Number 5 Journal of Pediatric Health Care


www.jpedhc.org

TABLE 2. ACR treatment groups and classifications of prognosis and disease level activity
Features of poor
Treatment group ILAR classifications prognosisa LDAb MDA HDAc
Arthritis of ≤ 4 joints Persistent oligoarthritis Arthritis of hip or cervical AJC ≤ 1 ≥1 features greater than AJC ≥ 2
Psoriatic arthritis spine Normal ESR or CRP level LDA ESR/CRP = 2 × ULN
ERA Arthritis of the ankle or wrist PhysGA: <3 of 10 <3 features of HDA PhysGA: ≥7 of 10
Undifferentiated arthritis and inflammatory marker ParGE: <2 of 10 ParGE: ≥4 of 10
elevation
Radiographic damage
Arthritis of ≥ 5 joints Extended oligoarthritis Arthritis of hip or cervical AJC ≤ 4 ≥1 features greater than AJC ≥ 8
Rf– polyarthritis spine Normal ESR or CRP level LDA ESR/CRP = 2 × ULN
Rf+ polyarthritis Rf+ PhysGA: <4 of 10 <3 features of HDA PhysGA: ≥7 of 10
Psoriatic arthritis Radiographic damage ParGE: <2 of 10 ParGE: ≥5 of 10
ERA
Undifferentiated arthritis
Active sacroiliac arthritis ERA or psoriatic arthritis with Radiographic damage of Normal back flexion ≥1 features greater than ESR/CRP = 2 × ULN
clinical and imaging any joint Normal ESR or CRP level LDA PhysGA: ≥7 of 10
evidence of active PhysGA: <4 of 10 <2 features of HDA ParGE: ≥4 of 10
sacroiliac arthritis ParGE: <2 of 10
Systemic arthritis with active Systemic arthritis (active 6 months of significant Two levels of disease activity:d
systemic features (without fever, SJIA, no arthritis) active systemic Active fever and PhysGA: <7 of 10
active arthritis) disease (fever, elevated Active fever and systemic features of HDA that result in PhysGA: ≥7 of 10
inflammatory markers, or
requirement for treatment
with systemic
glucocorticoids)
Systemic arthritis with active Systemic arthritis (with active Arthritis of hip AJC ≤ 4 ≥1 features greater than AJC ≥ 8
arthritis (without active arthritis) Radiographic damage Normal ESR or CRP level LDA ESR/CRP = 2 × ULN
systemic features) PhysGA: <4 of 10 <3 features of HDA PhysGA: ≥7 of 10
September/October 2018

ParGE: <2 of 10 ParGE: ≥5 of 10

Note. Data from Beukelman et al. (2011).


AJC, active joint count; ACR, American College of Rheumatology; CRP, C-reactive protein; ERA, enthesitis-related arthritis; ESR, erythrocyte sedimentation rate; HDA, high disease activity; ILAR,
International League of Associations for Rheumatology; LDA, low disease activity; MDA, moderate disease activity; ParGE, patient/parent general evaluation of overall well-being; PhysGA, physi-
cian global assessment of disease activity; Rf–, rheumatoid factor negative; Rf+, rheumatoid factor positive; SJIA, systemic juvenile idiopathic arthritis; ULN, upper limit of normal.
a
Must satisfy one criterion.
b
Must satisfy all criteria.
c
Must satisfy three criteria.
d
This criteria applies to all three (LDA, MDA, and HDA).
519
There are four main pharmacologic classes that can 2017). Aspirin is not recommended for treatment of
be used in the management of JIA. These include non- JIA because of its association with Reye syndrome.
steroidal anti-inflammatory drugs (NSAIDs), Cyclooxygenase-2 inhibitors (celecoxib) may be con-
glucocorticoids (intra-articular and systemic), nonbiologic sidered for patients with severe gastrointestinal adverse
disease-modifying antirheumatic drugs (DMARDs), and effects. A 2-month trial of NSAID monotherapy is in-
biologic DMARDs (bDMARDs). This review will provide dicated for patients with low-disease-activity (LDA)
a thorough discussion of each pharmacologic drug class arthritis in fewer than four joints, without joint con-
and its place in therapy, dosing regimens, adverse effects, tracture, and without features of poor prognosis
and monitoring parameters (Tables 3 and 4). (Beukelman et al., 2011). Some experts suggest that
a lack of response to one NSAID does not confer re-
Nonpharmacologic Treatment sponse to all agents in the class; a trial of an alternate
The management of JIA involves a multidisciplinary NSAID may be warranted (Harris et al., 2013).
approach. Nonpharmacologic therapy such as physi- Because NSAIDs provide only symptomatic relief from
cal, occupational, and pain and stiffness associated with arthritis and do not
psychosocial (e.g., The management stop the destructive inflammatory process associated
mental health, psychi- with this illness, most patients inflicted with moder-
atric) therapies have all
of JIA involves a ate to severe JIA receive NSAIDs as adjunctive therapy.
been used to help multidisciplinary Adverse reactions associated with NSAID use include
manage the disease approach. abdominal discomfort, gastritis/peptic ulcer disease, and
process in conjunction thrombocytopenia. Long-term use may result in renal
with pharmacologic toxicity such as renal papillary necrosis (Taketomo et al.,
agents (Giancane et al., 2016; Ostring & Singh-Grewal, 2017). A complete blood count (CBC), liver function
2013). Assistive device (wheelchairs, walkers), aerobic tests (LFTs), and serum creatinine should be com-
conditioning, and splinting are all techniques used to pleted before initiation and at least twice yearly for
help maintain physical functioning and prevent de- chronic daily use and once yearly for routine use (3-4
velopment of disability. Psychiatric counseling has also days per week; Beukelman et al., 2011).
proven beneficial, because many children with JIA suffer
from anxiety and/or depression secondary to chronic
pain and emotional distress due to decreased ability Intra-articular glucocorticoid injections
to perform normal childhood activities. Initiation of phar- Intra-articular glucocorticoid injections are reserved for
macologic therapy and education on the disease should patients with localized disease. Triamcinolone
include the child to ensure optimal medication com- hexacetonide ([TH], available as a 5 mg/ml and 20 mg/
pliance. Providers and parents should work to create ml injection) and triamcinolone acetonide ([TA], available
an environment that minimizes time away from school. as a 10 mg/ml and 40 mg/ml injection) have both been
Surgery may be indicated in patients who are unre- used in the treatment of JIA. TH is the more com-
sponsive to pharmacologic treatment. monly used agent because it has shown superior efficacy
(e.g., longer time to relapse) because of its lower solu-
Pharmacologic Treatment bility, resulting in slower absorption from the injected
joint and a longer duration of action compared with
Nonsteroidal anti-inflammatory drugs TA (Eberhard, Sison, Gottlieb, & Ilowite, 2004). These
NSAIDs remain first-line therapy (approved by the U.S. are considered first-line therapies in patients with mod-
Food and Drug Administration [FDA]) and the most erate disease activity (MDA) or high disease activity
commonly used agents in the treatment of JIA. NSAIDs (HDA) arthritis in fewer than four joints and those who
provide both anti-inflammatory and analgesic proper- failed a trial of NSAIDs (Beukelman et al., 2011). The
ties by blocking the production of prostaglandins through typical dosage of TH is based on the size of the child
cyclooxygenase-1 and cyclooxygenase-2 enzyme in- and the affected joint. Those with larger joints typi-
hibition. Dosing for anti-inflammatory effects is often cally receive TH at 10 to 20 mg/dose, and those with
twice the manufacturer recommended analgesia dose; smaller joints receive 2 to 6 mg/dose every 3 to 4 weeks
however, the lowest possible dose should be used to (Taketomo et al., 2017). Previous studies in children
minimize adverse effects (Giancane et al., 2016). Com- have used a TH dose of 1 mg/kg (maximum = 40 mg/
monly used NSAID regimens include naproxen, 10 to dose or 2 ml using 20 mg/ml injection) and a TA dose
15 mg/kg/day divided twice daily (range = 7-20 mg/ of 2 mg/kg (maximum = 80 mg/dose or 2 ml using
kg/day, maximum = 1,000 mg/day); ibuprofen, 30 to 40 mg/ml solution; Zulian et al., 2004). Clinical im-
40 mg/kg/day divided 3 to 4 times daily (maximum provement should last at least 4 months after joint
= 2,400 mg/day); and indomethacin, 1 to 2 mg/kg/ injection (Beukelman et al., 2011). Improvement for
day divided 2 to 4 times daily (maximum = 4 mg/kg/ less than 4 months may indicate the need for escala-
day or 150-200 mg/day; Taketomo, Hodding, & Kraus, tion of therapy. Common adverse events include skin

520 Volume 32 • Number 5 Journal of Pediatric Health Care


hypopigmentation and subcutaneous (SC) skin atrophy 2016; Taketomo et al., 2017). At this relatively lower
(Harris et al., 2013; Taketomo et al., 2017). dose, MTX functions as an anti-inflammatory agent rather
than a cytotoxic drug. It is typically administered orally
Systemic glucocorticoids initially, but may be given intramuscularly or SC in in-
Systemic glucocorticoids, prednisone and methylpred- stances of non-compliance, intolerance, or poor response
nisolone, are potent anti-inflammatory drugs that were to oral therapy given the lower oral bioavailability of
historically used as a bridge therapy until DMARDs 70%. Higher dosages of 30 mg/m2/week did not show
became effective. Because of the adverse event profile improved therapeutic response in patients with poly-
and the advent of newer therapeutic options, sys- articular JIA for whom 6 months of standard MTX doses
temic glucocorticoids have fallen out of favor. The ACR failed (Giancane et al., 2016). Clinical improvement
guidelines do not include recommendations for use of should be seen 6 to 12 weeks after initiation of therapy,
systemic glucocorticoids (Beukelman et al., 2011). but it could take up to 6 months to see maximal benefit.
However, they remain a therapeutic option for treat- Gastrointestinal (GI) adverse effects are the most
ment of systemic arthritis with active systemic features prominent, with abdominal pain and nausea occur-
(Beukelman et al., 2011). Glucocorticoids can be given ring within the first 24 to 36 hours after administration
as high-dose “pulse” intravenous (IV) methylpredniso- (Taketomo et al., 2017). Other adverse events include
lone at 15 to 30 mg/kg/day up to 1,000 mg/day for 1 oral ulcers, infection, and hematologic and liver tox-
to 3 days (Taketomo et al., 2017). However, because icity. Transient transaminitis usually resolves upon
of the short duration of action, some patients may discontinuation of MTX; no reports of irreversible liver
require chronic oral prednisolone at 1 to 2 mg/kg/ fibrosis have occurred. Glossitis and macrocytic anemia
day in 1 to 2 divided doses (maximum = 60 mg/day; are thought to be due to folate antagonism, and there-
Giancane et al., 2016; Taketomo et al., 2017). Common fore folate supplementation with folic acid or folinic
adverse effects associated with long-term use include acid may be considered. A retrospective, noncontrolled
weight gain, growth suppression, osteoporosis, acne, study of pediatric patients showed a reduction in the
and adrenal suppression (Taketomo et al., 2017). mean number of episodes per patient–year of hepatic
and GI toxicities after folinic acid supplementation
Nonbiologic disease-modifying antirheumatic drugs (Ravelli et al., 1999). Further studies are needed to assess
DMARDs are medications that slow the progression of the efficacy of folic acid/folinic acid and to develop
JIA and prevent long-term morbidity by controlling an appropriate dosing regimen. Monitoring param-
disease before irreversible damage occurs. Treatment eters include a CBC, LFTs, and serum creatinine before
with DMARD therapy may eliminate the need for anti- initiation, 1 month after initiation, 1 to 2 months after
inflammatory drugs. Because DMARDs do not provide dose escalation, and every 3 to 4 months once the
immediate symptom reduction, with benefits typi- patient stable receiving a dosing regimen with prior
cally evident weeks to months after initiation, bridging normal laboratory test values (Beukelman et al., 2011).
therapy may be necessary until the full onset occurs. Methotrexate is contraindicated in pregnant pa-
Although DMARDS are considered first-line disease- tients (because of the teratogenic potential) when used
modifying therapy, long-term outcomes in children with for the management of arthritis, mainly because there
JIA are limited because of the change in the defini- are many other therapies available. Fetal anomalies and
tion of JIA, different patient characteristics, and variations congenital malformations have been reported in preg-
in comparator. Table 3 provides an overview of nant patients exposed to methotrexate. Pregnancy should
DMARDs, including their mechanism of action, ap- be excluded in all patients of childbearing age before
proved indications, detailed dosing information, adverse the initiation of therapy and should be avoided for at
effects, and monitoring parameters. least 3 months after treatment in males and at least
one ovulatory cycle in females (Taketomo et al., 2017).
Methotrexate
Methotrexate (MTX) has an unknown mechanism of Leflunomide
action as an anti-inflammatory agent. MTX affects the Leflunomide is an immunomodulatory drug that in-
immune function and is a folic acid analog that in- hibits the mitochondrial enzyme dihydroorotate
hibits dihydrofolate reductase, interfering with purine dehydrogenase, reversibly inhibiting de novo pyrimi-
synthesis and DNA replication. MTX is recommended dine synthesis (Harris et al., 2013; Taketomo et al., 2017).
as the first-line nonbiologic DMARD for patients with It is recommended as a second-line nonbiologic DMARD
HDA and features of poor prognosis; for patients with in patients mainly because of the greater degree of clini-
joint arthritis involving fewer than four joints; and for cal experience associated with MTX (Beukelman et al.,
patients with HDA, irrespective of prognosis, with in- 2011). In patients with arthritis in five or more joints,
volvement of five or more joints (Beukelman et al., leflunomide may be considered as an approach to initial
2011). MTX dosages in the range of 10 to treatment for patients with HDA and features of poor
15 mg/m2/week are commonly used (Giancane et al., prognosis. Leflunomide is a prodrug that is rapidly

www.jpedhc.org September/October 2018 521


522
Volume 32 • Number 5

TABLE 3. Disease-modifying antirheumatic drugs used in the treatment of JIA


FDA-approved Common adverse Serious adverse Monitoring
Agent Mechanism of action indication Dosing regimen effects effects parameters
Methotrexate Inhibits dihydrofolate Polyarticular JIA 10–15 mg/m2/dose PO/SQ/IM weekly N/V Infection Platelets
reductase, interferes with Maximum = 30 mg/m2/dose Abdominal pain Cirrhosis CBC
purine synthesis and DNA Pulmonary toxicity LFTs
replication SCr
PFT
Leflunomide Inhibits dihydroorotate Off-label use for JIA ≥ 3 Loading dose: 100 mg PO × 1–3 days N/D Hepatic toxicity CBC
dehydrogenase, reversibly years of age (duration based on patient’s weight) Headache LFTs
inhibiting de novo Maintenance: 10–20 mg PO daily Rash
pyrimidine synthesis Maximum = 20 mg/dose Alopecia
Sulfasalazine Inhibits multiple inflammatory JIA ≥ 6 years of age Usual dose = 30–50 mg/kg/day PO in N/V/D Hepatitis CBC
pathways including 2 divided doses Hematologic effects Skin reactions (SJS, LFTs
leukotrienes, Maximum = 2,000 mg/day drug-induced SLE)
prostaglandins, and
possibly TNF-α
Thalidomide Multiple immunomodulatory Off-label use for SJIA ≥ 3 Initial dosage of 2 mg/kg/day PO, Drowsiness Teratogenicity CBC
characteristics, may years of age increased at 2-week intervals to Neutropenia Peripheral neuropathy CPK
suppress TNF-α and IL-6 3–5 mg/kg/day Somnolence
Myalgias
Cyclosporine Inhibits the production and Off-label use for JIA 2–5 mg/kg/day PO BID N/V Renal fibrosis BP
release of IL-2→ inhibition Maximum = 5 mg/kg/day Hypertension Hepatic toxicity SCr
of IL-2–induced Hirsutism Infection LFTs
T-lymphocyte activation Gingival hyperplasia CBC
Hydroxychloroquine Anti-inflammatory effects may Off-label use for JIA 3–5 mg/kg/day (as sulfate) PO in Agranulocytosis Ocular Ophthalmic
be due to the inhibition of 1–2 divided doses examination
migration of neutrophils/ Maximum = 7 mg/kg/day or Leukopenia Dermatologic (alopecia) CBC
eosinophils and 400 mg/day Thrombocytopenia LFT
prostaglandin inhibition Aplastic anemia
Cyclophosphamide Alkylating agent that interferes Not approved 500 mg/m2/dose IV monthly N/V Hemorrhagic cystitis CBC
Journal of Pediatric Health Care

with normal function of Maximum = 1,000 mg/m2/dose Anorexia Sterility LFTs


DNA Bone marrow toxicity SCr
Azathioprine Prevents purine metabolism, Off-label use for JIA and JIA: 2–2.5 mg/kg/dose PO once daily N/D Bone marrow CBC
inhibiting synthesis of DNA, JIA-associated uveitis Uveitis: Initial 2.4 mg/kg/dose PO once suppression
RNA, and proteins daily (range = 1.4–3.2 mg/kg/day) Pancytopenia

Note. Data from Harris et al. (2013), Taketomo, Hodding, and Krause (2017), and Brent (2001).
BID, twice daily; BP, blood pressure; CBC, complete blood count; CPK, creatinine phosphokinase; D, diarrhea; FDA, U.S. Food and Drug Administration; IL, interleukin; IM, intramuscular; IV,
intravenous; JIA, juvenile idiopathic arthritis; LFT, liver function test; N, nausea; PFT, pulmonary function test; PO, by mouth; SCr, serum creatinine; SJS, Stevens–Johnson syndrome; SLE, sys-
temic lupus erythematosus; SQ, subcutaneous; TNF-α, tumor necrosis factor-α; V, vomiting.
converted to its active plasma metabolite, teriflunomide continued sulfasalazine therapy because of drug toxicity.
(Sanofi-Aventis, 2016; Taketomo et al., 2017). Adverse effects include nausea, vomiting, diarrhea, an-
Teriflunomide is highly protein bound, with a pro- orexia, rash, bone marrow suppression, and hepatitis.
longed half-life of approximately 18 days. Given the Because sulfasalazine inhibits dihydrofolate reduc-
prolonged half-life and time to reach steady state, some tase, it may cause folate deficiency and megaloblastic
experts consider the administration of loading doses anemia. Because the data on FA or FLA supplemen-
of 100 mg orally for the first 3 days of therapy. However, tation in those who are taking sulfasalazine are limited,
loading doses may precipitate GI effects. Because of it is not commonly prescribed compared with MTX.
the lower steady state plasma concentration of the active Some clinicians may initiate therapy at half to one third
metabolite of leflunomide in children less than 40 kg, of the maintenance dose with weekly dose titrations
as reported in pharmacokinetic studies, the FDA does to decrease GI effects. Patients with a sulfa allergy should
not recommend its use for the treatment of JIA (Sanofi- avoid sulfasalazine because of the possible develop-
Aventis U.S., 2016; Silverman et al., 2005). ment of Stevens–Johnson syndrome. CBC and liver
Because leflunomide has been shown to be enzyme level should be periodically monitored.
embryotoxic and teratogenic, the FDA considers this
drug to be contraindicated in pregnancy (Sanofi- Other nonbiologic disease-modifying antirheumatic
Aventis U.S., 2016; Taketomo et al., 2017). Women of agents
childbearing potential must have a negative preg- Other nonbiologic DMARDs, including thalidomide,
nancy test result before initiation of leflunomide therapy, cyclosporine, tacrolimus, hydroxychloroquine, cyclo-
and contraceptive practices of patients should be docu- phosphamide, and azathioprine, have been previously
mented. In addition, women who wish to conceive while described in the treatment of JIA (Harris et al., 2013).
taking leflunomide must complete an enhanced drug These agents are not included in the ACR guidelines,
elimination procedure using cholestyramine 8 g three because there are no clear recommendations as to where
times a day for 11 days. This will reduce the half-life they fit within the disease subtypes and treatment
of teriflunomide to about 1 day (Brent, 2001). They groups. These agents may play a role in refractory
should also have two separate documented teriflunomide disease.
drug levels of less than 0.02 µg/ml at least 2 weeks Combination therapy involving multiple nonbiologic
before trying to conceive (Taketomo et al., 2017). The DMARDs has also been studied; however, with com-
most common adverse effects include GI symptoms, bination therapy there is a risk of additive
headache, and dermatologic symptoms (i.e., alope- immunosuppression and adverse effects. Thalido-
cia, skin rash; Taketomo et al., 2017). In addition, mide is one agent that should be used with extreme
patients should be monitored for hepatotoxicity, because caution because of its potent teratogenic potential. All
this drug can cause severe liver injury and is contra- patients, male and female, should receive birth control
indicated in hepatic failure. Liver enzyme level and CBC while receiving thalidomide therapy, and additional
should be obtained every 4 to 12 weeks. Monitoring caution should be taken in adolescents and those of
of serum teriflunomide trough levels may be consid- childbearing potential. Permanent peripheral neuropa-
ered when evaluating therapeutic efficacy; however, thy may occur with prolonged thalidomide exposure,
therapeutic trough concentrations have not been es- and patients should be monitored with regular neu-
tablished for the treatment of JIA (Taketomo et al., 2017). rologic examinations (Taketomo et al., 2017).
Thalidomide use has been described in children with
Sulfasalazine systemic JIA irresponsive to other therapies; however,
Sulfasalazine is a 5-aminosalicyclic acid analog that in- published data are limited to case series and small
terferes with several inflammatory pathways, including sample sizes. (Lehman et al., 2004). Cyclosporine and
leukotriene and prostaglandin production. It is a com- tacrolimus are drugs with a narrow therapeutic index
bination of sulfonamide (an antibiotic) and and, therefore, require therapeutic drug monitoring.
5-aminosalicyclic acid (anti-inflammatory agent). The Hydroxychloroquine can cause ocular abnormality and
ACR guidelines recommend sulfasalazine for treat- bone marrow suppression; ophthalmologic examina-
ment of ERA in patients with a history of arthritis of tions and periodic CBC should be monitored.
fewer than four joints with MDA or HDA after a trial Cyclophosphamide may cause hemorrhagic cystitis; a
of NSAIDs and/or glucocorticoid joint injections urine analysis and chemistries should be monitored.
(Beukelman et al., 2011). Because of conflicting results, There are limited data for azathioprine in the pediat-
the ACR guidelines do not recommend its use in non- ric population for treatment of JIA, and it is considered
ERA patients. Some literature recommends reserving a last-line nonbiologic DMARD therapy.
sulfasalazine as an add-on therapy for patients with
refractory disease receiving combination therapy because Biologic disease-modifying antirheumatic drugs
of conflicting results and adverse effects. Van Rossum The development of biologic agents has greatly changed
et al. (1998) reported that 31% of children with JIA dis- the outcomes and morbidity associated with this disease.

www.jpedhc.org September/October 2018 523


Biologic agents are substances that are made from a In patients with arthritis of fewer than four joints,
living organism or its products and may include mono- TNFi agents are indicated after a 3-month trial of MTX
clonal antibodies, receptor antagonists, or soluble plus glucocorticoid joint injections in patients with MDA
cytokine receptors that target specific proteins in- or HDA with poor prognosis or after a 6-month trial of
volved in the inflammatory cascade. Biologic agents MTX plus glucocorticoid joint injections for patients with
are administered via the IV or SC routes, which may HDA without features of poor prognosis (Beukelman
be more challenging and can affect medication com- et al., 2011). Furthermore, in patients with arthritis of
pliance. The financial implications associated with these five or more joints, a TNFi may be consider after 3
agents are substantial, and pharmacoeconomic months of MTX or leflunomide in patients with MDA
analyses evaluating the cost per treatment among regi- or had, irrespective of poor prognosis, or after 6 months
mens are lacking. Costs associated with these agents of MTX or leflunomide in patients with LDA, irrespec-
must include IV administration and the requirement tive of poor prognosis. Additionally, they can be used
for infusion clinics. in patients with systemic arthritis with active arthritis
Before initiation of these therapies, providers should (without active systemic features) after 3 months of MTX
take into consideration insurance coverage and the fi- in patients with MDA or had, irrespective of poor prog-
nancial impact that will be placed on the patient and nosis. Because response is delayed, a 4-month trial of
family. Because of their relatively new advent in the a TNFi is indicated. If response is not optimal after the
1990s, long-term outcomes and safety data are sparse. initial bDMARD trial, it is reasonable to try a different
Biologic agents also carry the risk of severe immuno- TNFi or switch to a different bDMARD. Infliximab did
suppression, and routine monitoring and close follow- not show a statistically significant benefit compared with
up are extremely important. Table 4 provides a detailed placebo in its primary endpoint (ACR Pedi 30) at week
review of biologic agents that have been used in the 14 of therapy in a Phase 3 study, and therefore it is
treatment of JIA. not FDA approved for JIA treatment (Ruperto et al.,
2007). The ACR guidelines, however, do acknowl-
TNF-α inhibitors edge infliximab as an available TNFi option. Currently,
TNF-α inhibitors (TNFi) remain the most commonly there are no clinical trials comparing TNFi agents head
prescribed biologic agents for the treatment of JIA. El- to head for the treatment of JIA.
evated levels of the proinflammatory cytokine TNF-α
are found in children with JIA, and therefore inhibi- Abatacept
tors of these cytokines play an important role in Abatacept is a soluble fusion protein that is a selec-
modifying the inflammatory disease progression. Avail- tive co-stimulation modulator. It inhibits T-cell activation
able agents in this class include etanercept, adalimumab, by binding to CD80 or CD86 on antigen-presenting cells,
and infliximab. Although all three agents target TNF- preventing interaction of the antigen-presenting cells
α, they differ slightly in the manner in which they with CD28 (Harris et al., 2013). Abatacept is typically
achieve inhibition. Adalimumab and infliximab are reserved for patients for whom therapy with a TNFi
monoclonal antibodies, whereas etanercept is a soluble has failed, which highlights the potential for other in-
receptor antagonist. The monoclonal antibodies differ flammatory mediators and cytokines playing a role in
in terms of their antibody type: adalimumab is a fully the pathogenesis of JIA. Clinical trials showed that of
humanized monoclonal antibody, whereas infliximab patients who discontinued a TNFi, 25% achieved an
is a chimeric monoclonal antibody, thus resulting in ACR Pedi 50 after 4 months of abatacept therapy
a potentially higher immunogenicity. Other differ- (Ruperto et al., 2008). Initial improvement in clinical
ences include their route of administration. Infliximab symptoms is typically seen by the third or fourth dose;
is given via IV infusion, whereas etanercept and however, response may be delayed in some patients.
adalimumab are SC injections. Given the concern for Abatacept is recommended for the following pa-
the risk of infusion-related reactions associated with tients: (a) patients with arthritis in five or more joints
infliximab, etanercept and adalimumab may be the more and who have received a TNFi for 4 months who and
preferable agents. Etanercept was the first biologic agent have HDA, irrespective of poor prognosis, or MDA with
approved in the treatment of moderate to severe poly- features of poor prognosis; (b) patients with arthritis
articular JIA in children 2 years of age and older in in five or more joints who have received more than
1999. In 2008, adalimumab received FDA approval for one TNFi and have MDA or HAD, irrespective of poor
use in children 4 years of age and older with polyar- prognostic features, or LDA with poor prognosis; and
ticular JIA (Taketomo et al., 2017). Currently, clinical (c) patients who have received MTX and a TNFi who
trials on the use of other TNFi agents such as golimumab have HAD, irrespective of poor prognosis, or MDA with
and certolizumab, both approved for adult rheuma- features of poor prognosis (Beukelman et al., 2011).
toid arthritis, for the treatment of JIA are underway. Abatacept is administered as an IV infusion every 2
Golimumab and certolizumab are currently not ad- weeks for the first month and monthly thereafter because
dressed in the ACR guidelines (Beukelman et al., 2011). of its long half-life of 8 to 25 days (Taketomo et al.,

524 Volume 32 • Number 5 Journal of Pediatric Health Care


www.jpedhc.org

TABLE 4. Biologic disease-modifying antirheumatic drugs used in the treatment of JIA


Common adverse Serious adverse Monitoring
Medication Mechanism of action FDA-approved indication Dosing regimen effects effects parameters

TNF-α inhibitors
Etanercept (Enbrel; Amgen, Soluble, dimeric, fusion Moderate to severe PJIA ≥ 2 years of age Weekly: 0.8 mg/kg/dose SC once weekly; maximum = 50 mg/dose Injection site reaction Infection HBsAg
Thousand Oaks, CA) protein that binds to Twice weekly: 0.4 mg/kg/dose given 72–96 hours apart; maximum = 25 mg/ URI Anaphylaxis PPD
TNF-α preventing binding dose twice weekly Urticaria Autoantibodies Dermatologic
to cell-surface receptors Malignancy examination
TB reactivation
Adalimumab (Humira; Abbvie, North Fully humanized Moderate to severe PJIA ≥ 2 years of age PJIA Injection site reaction Infection HBsAg
Chicago, IL) recombinant IgG Off-label use for JIA-associated uveitis 10 to <15 kg: 10 mg SC every other week URI Anaphylaxis PPD
monoclonal Ab that 15 to <30 kg: 20 mg SC every other week Urticaria Autoantibodies
binds to TNF-α ≥30 kg: 40 mg SC every other week Malignancy
Uveitis TB reactivation
<30 kg: 20 mg every other week
≥30 kg: 40 mg every other week
Infliximab (Remicade; Janssen, Monoclonal Ab that binds to Off-label use for JIA and uveitis Age ≥ 4 years: 3 mg/kg/dose IV at 0, 2, and 6 weeks, then 3–6 mg/kg/dose Injection site reaction Infection HBsAg
Raritan, NJ) both soluble and every 8 weeks + MTX Myalgia Anaphylaxis PPD
membrane-bound TNF-α URI Autoantibodies
Urticaria PNA
Malignancy
TB reactivation
T-cell co-stimulation blocker
Abatacept (Orencia; Bristol-Myers T-cell co-stimulator inhibitor Moderate to severe PJIA ≥6 years of age <75 kg: 10 mg/kg/dose Headache Varicella CBC
Squibb, New York, NY) through binding to CD80/ 75–100 kg: 750 mg/dose Nausea Anaphylaxis PPD
CD86 >100 kg: 1,000 mg/dose URI TB reactivation
IV at 0, 2, and 4 weeks, then every 4 weeks thereafter Ovarian cyst
ALL
IL-1 inhibitors
Anakinra (Kineret; Swedish Orphan Human recombinant IL-1 Off-label use for SJIA and PJIA SJIA Injection site reaction Hepatitis SCr
Biovitrum, Stockholm, Sweden) receptor antagonist Initial: 1–2 mg/kg/dose SC daily (maximum = 100 mg/day); may increase to URI Infections ANC
4 mg/kg/day at 2-week intervals (maximum = 200 mg/day) Headache Secondary malignancies CBC
PJIA: Nausea
1 mg/kg/dose SC daily; Max 100 mg/day Diarrhea
Neutropenia
Arthralgia
Rilonacept (Arcalyst; Regeneron, Binds to IL-1 receptor Not approved Age 12–17 years Injection site reaction Serious infections CBC
Tarryton, NY) preventing interaction Loading dose: 4.4 mg/kg/dose SC (maximum = 320 mg/dose) URI
with cell surface May divide into 1–2 injections (maximum injection volume = 2 ml)
receptors Maintenance: 2.2 mg/kg/dose SC weekly (maximum = 160 mg/dose)
Canakinumab (Ilaris; Novartis, East Humanized monoclonal Ab SJIA ≥2 years of age Age ≥ 2 years: 4 mg/kg/dose SC every 4 weeks URI Serious infections CBC
Hanover, NY) that selectively blocks Maximum = 300 mg/dose Abdominal pain
IL-1β
September/October 2018

Injection site reaction


IL-6 inhibitor
Tocilizumab (Actemra; Genetech, Anti–IL-6 receptor SJIA and PJIA ≥2 years SJIA: URI Serious infections BP
South San Francisco, CA) monoclonal Ab <30 kg: 12 mg/kg/dose IV every 2 weeks Headache (PNA, UTI, herpes CBC
≥30 kg: 8 mg/kg/dose IV every 2 weeks HTN zoster) PPD
Maximum = 800 mg/dose Increased ALT GI perforation LFT
PJIA: Anaphylaxis ANC
<30 kg: 10 mg/kg/dose IV every 4 weeks TB Lipid panel
≥30 kg: 8 mg/kg/dose IV every 4 weeks
Maximum = 800 mg/dose
Other drug
Rituximab (Rituxan; Biogen, Anti-CD20 B-cell depleting Off-label use for refractory PJIA and SJIA 375 mg/m2/dose IV infusion once weekly for 2–4 weeks Infusion reactions Neuropathy CBC
Cambridge, MA) monoclonal Ab Maximum = 1,000 mg/dose HTN Tumor lysis syndrome ECG
Increased ALT SJS HBsAg

Ab, antibody; ALL, acute lymphocytic leukemia; ALT, alanine aminotransferase; ANC, absolute neutrophil count; BP, blood pressure; CBC, complete blood count; ECG, electrocardiography; FDA, U.S. Food and Drug Administration; GI, gastrointestinal; HBsAg,
525

hepatitis B surface antigen; HTN, hypertension; Ig, immunoglobulin; IL, interleukin; IV, intravenous; LFT, liver function test; MTX, methotrexate; PJIA, polyarticular juvenile idiopathic arthritis; PNA, pneumonia; PPD, purified protein derivative of tuberculin; SC,
subcutaneous; SJIA, systemic juvenile idiopathic arthritis; SJS, Stevens–Johnson Syndrome; TB, tuberculosis; TNF-α, tumor necrosis factor-α; URI, upper respiratory infection; UTI, urinary tract infection.
2017). Adverse events were common and included head- (Frampton, 2013). Tocilizumab is administered as a
ache, nausea, diarrhea, cough, and upper respiratory 1-hour infusion every 2 weeks for SJIA and every 4
infection (Taketomo et al., 2017). Serious adverse effects weeks for polyarticular JIA. Adverse effects include neu-
such as arthritis flare, varicella, ovarian cyst, and acute tropenia, thrombocytopenia, liver transaminitis,
lymphocytic leukemia were seen in 3% of patients hypertriglyceridemia, hypercholesterolemia, and GI per-
(Ruperto et al., 2008). Because abatacept can alter the foration (Taketomo et al., 2017). Tocilizumab should
immune response against infections and malignan- not be initiated in patients with an active infection, ab-
cies because of T-cell inhibition, consideration should solute neutrophil count less than 2,000/mm3, platelet
be made to hold abatacept doses in patients with sus- count less than 100,000/mm3, or alanine aminotrans-
pected infection. Absence of latent or active tuberculosis ferase or aspartate aminotransferase level more than
should be documented before initiation of therapy, with 1.5 times the upper limit of normal. LFT and CBC should
periodic monitoring of CBC and LFTs every 4 to 12 be monitored before therapy initiation, with the second
weeks while taking abatacept (Taketomo et al., 2017). infusion, and every 2 to 4 weeks (for SJIA) or every 4
Providers should be educated that glucose readings on to 8 weeks (for polyarticular). A lipid panel should also
the day of infusion may be falsely elevated because be obtained before therapy, about 4 to 8 weeks after
the powder for reconstitution contains maltose initiation and every 6 months thereafter while receiv-
(Taketomo et al., 2017). ing therapy (Taketomo et al., 2017).

IL-1 inhibitors Rituximab


IL-1 inhibitors include anakinra, canakinumab, and Rituximab is a monoclonal antibody directed against
rilonacept; anakinra and rilonacept are IL-1 receptor the CD20 antigen on the B-lymphocyte surface, which
antagonists, and canakinumab is an anti-IL-1 mono- thereby inhibits the inflammatory cascade. The ACR
clonal antibody (Espinosa & Gottlieb, 2012). All three guidelines suggest that rituximab may be used in pa-
agents were evaluated for the treatment of JIA by the tients with fewer than five joints involved for whom
current ACR guidelines (Beukelman et al., 2011). IL-1 both an IL-1 inhibitor and tocilizumab sequentially or
inhibitors are indicated for patients with systemic ar- a DMARD plus IL-1 inhibitor or tocilizumab failed. In
thritis with active systemic features who have fever and addition, it can also be used in patients with five or
features of poor prognosis (irrespective of current more joints involved for whom both an IL-1 inhibitor
therapy level) or those with ongoing disease activity and tocilizumab or a DMARD plus IL-1 inhibitor,
while receiving systemic glucocorticoids. It is also in- tocilizumab, TNFi, or abatacept failed (Beukelman et al.,
dicated in patients with systemic arthritis with active 2011). Furthermore, rituximab use has also been re-
arthritis and MDA or HDA, irrespective of features of ported in limited case reports for refractory JIA (Giancane
poor prognosis, after an unsuccessful 3-month trial of et al., 2016). Boxed warnings include severe and oc-
MTX or a 4-month trial of a TNFi (Beukelman et al., casionally fatal infusion-related reactions. Pretreatment
2011). Anakinra should not be used in combination with acetaminophen, an antihistamine, and methyl-
with a TNFi because of the risk of severe neutrope- prednisolone should be considered, and medications
nia. A CBC with differential should be obtained before for the treatment of hypersensitivity reactions (e.g., epi-
initiation, monthly for 3 months and then quarterly there- nephrine, bronchodilators) should be available for
after while receiving therapy (Taketomo et al., 2017). immediate use. Other boxed warnings include severe
IL-1 inhibitors are administered SC, and their fre- mucocutaneous reactions such as Stevens–Johnson syn-
quency is drug dependent. Anakinra has a short half- drome, the development of progressive multifocal
life of 4 to 7 hours and thus requires daily injections. leukoencephalopathy, and the potential for reactiva-
Rilonacept has a half-life of approximately 8 days and tion of hepatitis B (Taketomo et al., 2017). Because
therefore can be administered once a week. of significant adverse events, the use of rituximab in
Canakinumab has a prolonged half-life of 23 to 26 days the management of JIA has decreased significantly. Pa-
and therefore can be administered every 4 weeks tients should be watched closely for infusion-related
(Taketomo et al., 2017). reactions and should receive continuous electrocar-
diographic and vital sign monitoring during and
IL-6 inhibitors immediately after infusion. B-cell levels should be evalu-
Tocilizumab is a recombinant humanized monoclonal ated before and 1 month after infusion, with quantitative
antibody approved for the treatment of JIA and is cur- immunoglobulin levels assessed every 3 months. Clini-
rently the only agent that functions as an IL-6 receptor cal improvement with rituximab is typically seen within
antagonist. Tocilizumab received FDA approval for the 1 month of the initial infusion.
treatment of SJIA and polyarticular JIA in patients 2
years of age or older (Frampton, 2013). It is indicated Adverse effects/monitoring parameters for bDMARDs
in patients with continued disease activity after glu- The risk of infection is one of the most serious adverse
cocorticoid monotherapy, MTX, leflunomide, or anakinra effects associated with bDMARDs. Concerns regard-

526 Volume 32 • Number 5 Journal of Pediatric Health Care


ing serious infection risks including the development MAS is a rare but potentially life-threatening syn-
of opportunistic infections secondary to immunosup- drome that most frequently occurs in SJIA patients. It
pression, hepatitis B and tuberculosis reactivation, and is thought to be secondary to organ infiltration by
a decreased response to vaccination (Taketomo et al., activated macrophages due to a cytokine storm. Pa-
2017). Patients should be screened for hepatitis B surface tients with MAS present with fever, pancytopenia,
antigen, hepatitis B core antibody, and tuberculosis using liver failure, coagulopathy with hemorrhage or throm-
chest radiography and/or purified protein derivative bophilia, encephalopathy, and seizures. Treatment of
testing and/or tuberculosis blood tests (interferon-γ MAS typically requires admission to an intensive care
release assay) before initiation of therapy (Taketomo unit and administration of immunosuppressive agents
et al., 2017). Patients should be monitored for symp- (Brent, 2001).
toms of hepatitis B virus reactivation during treatment
and for several months after therapy. Purified protein SUMMARY
derivative testing should be repeated yearly through- JIA is a chronic, debilitating childhood condition with
out therapy. All efforts should be made to ensure that many long-term consequences. The development of
patients are fully vaccinated before the initiation of the ILAR classification
therapy. As with all forms of immunosuppression, live system and ACR treat- The development
vaccines should be avoided concurrently and for 3 ment guidelines better
months after biologic therapy. Secondary malignan- define this childhood
of the ILAR
cies have also been associated with the use of bDMARDs disease and provide a classification
and are therefore contraindicated in patients with active more objective manner system and ACR
malignancy. Further studies are needed to address this in which to treat and
associated risk, because many of these cases may have monitor patients with
treatment
been compounded by concomitant immunosuppres- JIA. Diagnosis is based guidelines better
sion use and underlying illnesses. mainly on clinical define this
symptoms, and accu-
COMPLICATIONS rate classification of
childhood disease
Untreated or inadequately treated JIA can result in many JIA disease subtype is and provide a more
long-term consequences such as anemia, pericarditis, extremely important. objective manner in
chronic pain, slow rate of growth, and uveitis. Irido- Chronic pain and
cyclitis, a chronic anterior uveitis, is one of the most inflammation are hall-
which to treat and
common complications of JIA occurring in approxi- mark characteristics of monitor patients
mately 15% to 20% of children inflicted with this disease this disorder, and with JIA.
(Espinosa & Gottlieb, 2012). If left untreated, uveitis without treatment many
can result in cataracts, glaucoma, and blindness. Pa- patients will develop
tients with ANA-positive titer results have been found disability. Many pharmacotherapeutic agents are avail-
to be at a higher risk for development of uveitis and able for the management of JIA. DMARDs have been
should undergo monitoring with slit lamp examina- shown to be the most effective in preventing long-
tions by an ophthalmologist more frequently than term consequences of this disease. MTX continues to
children with JIA who have negative ANA titer results. be a preferred agent because of its affordability, proven
Primary treatment of uveitis involves the use of topical efficacy, safety, and tolerability. bDMARDs have revo-
ocular corticosteroid drops; however, some patients may lutionized treatment of JIA, having shown promise in
require systemic or intraocular corticosteroid injec- promoting disease remission, but they are associated
tions (Borchers et al., 2006; Ostring & Singh-Grewal, with high costs and many safety concerns. Clinicians
2013). should evaluate the efficacy and safety of all avail-
Chronic, persistent arthritis in the knee can also lead able treatment options and tailor each medication
to growth disturbances, specifically leg length distur- regimen to the individual patient. The importance of
bances, secondary to accelerated growth in affected routine monitoring and close follow-up should be em-
joints (Borchers et al., 2006; Espinosa & Gottlieb, 2012). phasized, given the complexity of the regimens at hand.
Growth retardation may be seen in patients with per- More studies are needed to assess the long-term safety
sistent arthritis in the wrist and ankles. A higher incidence and efficacy of the bDMARDs for JIA. In addition, the
of osteopenia and osteoporosis have been seen in pa- completion of head-to-head comparative trials of dif-
tients with JIA, possibly secondary to bone loss and ferent treatment options could help identify preferred
reduced bone density (due to corticosteroid treat- drug therapy.
ment), decreased weight-bearing activity, and
malnutrition. In addition, persistent arthritis can result REFERENCES
in limited mobility, decreased quality of life, joint Beukelman, T., Patkar, N. M., Saag, K. G., Tolleson-Rinehart, S., Cron,
damage, and psychosocial issues. R. Q., DeWitt, E. M., … Ruperto, N. (2011). 2011 American

www.jpedhc.org September/October 2018 527


College of Rheumatology recommendations for the treatment Ostring, G. T., & Singh-Grewal, D. (2013). Juvenile idiopathic arthri-
of juvenile idiopathic arthritis: Initiation and safety monitoring of tis in the new world of biologics. Journal of Paediatrics and Child
therapeutic agents for the treatment of arthritis and systemic Health, 49, E405-E412.
features. Arthritis Care & Research, 63, 465-482. Petty, R. E., Southwood, T. R., Manners, P., Baum, J., Glass, D. N.,
Borchers, A. T., Selmi, C., Cheema, G., Keen, C. L., Shoenfeld, Y., Goldenberg, J., … Woo, P. (2004). International League of As-
& Gershwin, M. E. (2006). Juvenile idiopathic arthritis. Autoim- sociations for Rheumatology classification of juvenile idiopathic
munity Reviews, 5, 279-298. arthritis: Second revision, Edmonton, 2001. Journal of Rheu-
Brent, R. L. (2001). Teratogen update: Reproductive risks of leflunomide matology, 31, 390-392.
(Arava); A pyrimidine synthesis inhibitor: Counseling women taking Ravelli, A., Migliavacca, D., Viola, S., Ruperto, N., Pistorio, A., & Martini,
leflunomide before or during pregnancy and men taking A. (1999). Efficacy of folinic acid in reducing methotrexate tox-
leflunomide who are contemplating fathering a child. Teratol- icity in juvenile idiopathic arthritis. Clinical & Experimental
ogy, 63, 106-112. Rheumatology, 17, 625-627.
Consolaro, A., Giancane, G., Schiappapietra, B., Davi, S., Calandra, Ringold, S., Weiss, P. F., Beukelman, T., DeWitt, E. M., Illowite, N. T.,
S., Lanni, S., & Ravelli, A. (2016). Clinical outcome measures Kimura, Y., … Vehe, R. K. (2013). 2013 Update of the 2011
in juvenile idiopathic arthritis. Pediatric Rheumatology, 14, American College of Rheumatology recommendations for the
23. treatment of juvenile idiopathic arthritis. Arthritis & Rheuma-
Dannecker, G. E., & Quartier, P. (2009). Juvenile idiopathic arthritis: tism, 65, 2499-2512.
Classification, clinical presentation and current treatments. Ruperto, N., Lovell, D. J., Cuttica, R., Wilkinson, N., Woo, P., Espada,
Hormone Research, 72(Suppl. 1), 4-12. G., … Giannini, E. H. (2007). A randomized, placebo con-
Eberhard, B. A., Sison, M. C., Gottlieb, B. S., & Ilowite, N. T. (2004). trolled trial of infliximab plus methotrexate for the treatment of
Comparison of the intraarticular effectiveness of triamcinolone polyarticular-course juvenile rheumatoid arthritis. Arthritis & Rheu-
hexacetonide and triamcinolone acetonide in treatment of ju- matism, 56, 3096-3106.
venile rheumatoid arthritis. Journal of Rheumatology, 31, 2507- Ruperto, N., Lovell, D. J., Quartier, P., Paz, E., Rubio-Perez, N., Silva,
2512. C. A., … Giannini, E. H. (2008). Abatacept in children with ju-
Ellis, J. A., Munro, J. E., & Ponsonby, A. L. (2010). Possible envi- venile idiopathic arthritis: A randomized, double-blind, placebo-
ronmental determinants of juvenile idiopathic arthritis. controlled withdrawal trial. Lancet, 372(9636), 383-391.
Rheumatology (Oxford, England), 49, 411-425. Sanofi-Aventis, U. S. (2016). Arava (leflunomide) [package insert].
Espinosa, M., & Gottlieb, B. S. (2012). Juvenile idiopathic arthritis. Bridgewater, NJ. Author.
Pediatrics in Review, 33, 303-313. Schwarz, M. M., Simpson, P., Kerr, K. L., & Jarvis, J. N. (1997). Ju-
European League Against Rheumatism. (1977). EULAR bulletin no. venile rheumatoid arthritis in African Americans. Journal of
4: Nomenclature and classification or arthritis in children. Basel: Rheumatology, 49, 1826-1829.
National Zeitung. Silverman, E., Mouy, R., Spiegel, L., Jung, L. K., Saurenmann, R. K.,
Frampton, J. E. (2013). Tocilizumab: A review of its use in the treat- Lahdenne, P., … Strand, V. (2005). Leflunomide or methotrex-
ment of juvenile idiopathic arthritis. Pediatric Drugs, 15, 515- ate for juvenile rheumatoid arthritis. New England of Journal, 352,
531. 1655-1666.
Giancane, G., Consolaro, A., Lanni, S., Davi, S., Schiappapeitra, B., Taketomo, C. K., Hodding, J. H., & Kraus, D. M. (2017). Pediatric
& Ravelli, A. (2016). Juvenile idiopathic arthritis: Diagnosis and and neonatal dosage handbook (24th ed.). Hudson, OH:
treatment. Rheumatology Therapy, 3, 187-207. Lexi-Comp.
Harris, J. G., Kessler, E. A., & Verbsky, J. W. (2013). Update on the Van Rossum, M. A., Fiselier, T. J., Franssen, M. J., Zwinderman, A. H.,
treatment of juvenile idiopathic arthritis. Current Allergy & Asthma ten Cate, R., van Suijlekom-Smit, L. W., … Diikmans, B. A. (1998).
Reports, 13, 337-346. Sulfasalazine in the treatment of juvenile chronic arthritis: A ran-
Hinks, A., Ke, X., Barton, A., Eyre, S., Bowes, J., Worthington, J., domized, double-blind, placebo-controlled, multicenter study.
… Thomson, N. (2009). Association of the IL2RA/CD25 gene Dutch Juvenile Chronic Arthritis Study Group. Arthritis & Rheu-
with juvenile idiopathic arthritis. Arthritis & Rheumatism, 60, 251- matism, 41, 808-816.
257. Wallace, C. A., Giannini, E. H., Huang, B., Itert, L., & Ruperto, N.
Lehman, T. J., Schechter, S. J., Sundel, R. P., Oliveira, S. K., (2011). American College of Rheumatology provisional criteria
Huttenlocher, A., & Onel, K. B. (2004). Thalidomide for severe for defining clinical inactive disease in select categories of ju-
systemic onset juvenile rheumatoid arthritis: A multicenter study. venile idiopathic arthritis. Arthritis Care & Research (Hoboken),
Journal of Pediatrics, 145, 856-857. 63, 929-936.
Lovell, D. J., Ruperto, N., Giannini, E. H., & Martini, A. (2013). Ad- Woo, P., & Colbert, R. A. (2009). An overview of genetics of paedi-
vances from clinical trials in juvenile idiopathic arthritis. Nature atric rheumatic diseases. Best Practice & Research Clinical
Reviews. Rheumatology, 9, 557-563. Rheumatology, 23, 589-597.
Minden, K., Niewerth, M., Listing, J., Biedermann, T., Bollow, M., Zulian, F., Martini, G., Gobber, D., Plebani, M., Zacchello, F., & Manner,
Schontube, M., & Zink, A. (2002). Long-term outcome in pa- P. (2004). Triamcinolone acetonid and hexacetonide intra-
tients with juvenile idiopathic arthritis. Arthritis & Rheumatism, articular treatment of symmetrical joints in juvenile idiopathic
46, 2392-2401. arthritis: A double-blind trial. Rheumatology, 43, 1288-1291.

528 Volume 32 • Number 5 Journal of Pediatric Health Care

You might also like