ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Polyarticular juvenile idiopathic arthritis: Treatment

Polyarticular juvenile idiopathic arthritis: Treatment
Literature review current through: Jan 2024.
This topic last updated: Nov 03, 2023.

INTRODUCTION — Polyarticular juvenile idiopathic arthritis (formerly called polyarticular-onset juvenile rheumatoid arthritis [RA]) is a subset of juvenile idiopathic arthritis (JIA) that is defined by the presence of more than four affected joints during the first six months of illness [1]. In the revised nomenclature, this disease, which comprises 20 to 30 percent of patients with JIA, is included in the group termed "childhood polyarthritis." (See "Classification of juvenile idiopathic arthritis".)

Therapy is directed toward treating the underlying inflammation of JIA and preventing complications associated with JIA (eg, joint damage) or the adverse effects of its treatment [2]. Medications used in the treatment of polyarticular JIA have various mechanisms of action, some of which are unknown, but all suppress inflammation. First-line agents include nonbiologic disease-modifying antirheumatic drugs (DMARDs) such as methotrexate. Second-line drugs referred to as biologic DMARDs (bDMARDs) include anti-tumor necrosis factor (TNF) alpha agents.

Although many parents/caregivers and clinicians voice fear of potential side effects from medications, years of experience make it clear that there is far more to fear from the damage known to be caused by ongoing joint inflammation. The belief statement that children often "grow out of JIA" is incorrect and may lead to delays in appropriate therapy and poor outcomes. It is essential that clinicians make parents/caregivers aware of the fact that children with polyarticular JIA are unlikely to outgrow their disease since this myth may heavily influence their understanding of the risk-to-benefit ratio of medications.

The treatment of polyarticular JIA is reviewed here. The clinical manifestations, diagnosis, complications, and prognosis of polyarticular JIA are discussed separately. (See "Polyarticular juvenile idiopathic arthritis: Clinical manifestations, diagnosis, and complications".)

GOALS OF TREATMENT — Advances in the therapeutic options available to treat polyarticular JIA have made remission a realistic goal for most children. An international task force recommended remission as the treatment goal with emphasis on assuring normal growth and development and avoidance of long-term systemic glucocorticoids [3]. In one single-center study, implementation of structured disease activity monitoring with the clinical Juvenile Arthritis Disease Activity Score (cJADAS-10; score = active joint count [0 to 10] + physician global disease activity score [0 to 10] + caregiver/patient global well-being score [0 to 10]) in the setting of clinical decision support resulted in significant improvements in disease activity in both incident and prevalent cases of disease [4].

MANAGEMENT APPROACH — The approach to the treatment of polyarticular JIA is reviewed here and is consistent with the approach outlined in the 2019 American College of Rheumatology (ACR)/Arthritis Foundation (AF) guideline for therapeutic approaches for JIA [2]. Treatment of polyarticular JIA is individualized based upon disease activity and severity, as well as patient preferences, after reviewing the risks, benefits, and costs of the therapies. The specific agents are discussed in detail separately or in the sections below. (See "NSAIDs (including aspirin): Pharmacology and mechanism of action" and 'Methotrexate' below and 'Tumor necrosis factor inhibitors' below.)

Pretreatment assessment — Patients are assessed for the presence of features associated with poor prognosis, which include [2]:

Cervical or hip arthritis

Rheumatoid factor [RF] positivity

Cyclic citrullinated peptide [CCP] positivity

Radiographic evidence of joint damage (erosions or joint space narrowing)

The updated ACR/AF guidelines do not differentiate initial treatment recommendations for those with low versus moderate/high disease activity (defined as a clinical Juvenile Arthritis Disease Activity Score [cJADAS] of ≤2.5 or >2.5, respectively), with the exception of the option of a limited course of oral glucocorticoids as adjunct therapy for those with moderate/high disease activity and significant impairment [2].

Initial management — Initial drug therapy for children with polyarticular JIA should be aggressive in order to control the inflammatory process and relieve symptoms as quickly as possible while minimizing drug side effects. A disease-modifying antirheumatic drug (DMARD) is recommended for all patients at the time of diagnosis since it is rare for nonsteroidal antiinflammatory drugs (NSAIDs) alone to control the inflammatory process of polyarticular disease, and chronic joint changes may occur if arthritis remains active [5]. NSAIDs are appropriate for children with suspected disease awaiting rheumatology evaluation or as adjunctive therapy for symptom management.

DMARDs used to treat polyarticular JIA include the nonbiologic agents (eg, methotrexate) and the biologic agents (eg, tumor necrosis factor [TNF] inhibitors). Methotrexate is the traditional DMARD of choice. Indications for using a biologic agent in addition to or rather than methotrexate include presence of severe polyarthritis, poor prognostic features, or factors associated with poor response to methotrexate (eg, predominantly axial arthritis).

For children with high or moderate disease activity (cJADAS of >2.5), adjunctive therapy with oral glucocorticoids (total duration of less than three months) is conditionally recommended for those with significant discomfort or mobility impairment. Chronic low-dose glucocorticoid use is not recommended for children, regardless of disease activity or risk factors.

Escalation of therapy is recommended in children with continued disease activity in at least one joint after three months of treatment [2] and may include any of the following:

Intraarticular glucocorticoid injections.

Changing the route of administration (to subcutaneous [SC] if receiving oral formulation) or dose (if not at maximal dose) of methotrexate.

Adding a biologic agent (if only on methotrexate).

Changing to an alternate biologic agent or a Janus kinase (JAK) inhibitor (if already on a biologic agent). If a patient has failed a TNF inhibitor, then switching to a non-TNF inhibitor biologic (eg, tocilizumab, abatacept) or a JAK inhibitor (eg, tofacitinib or possibly baricitinib) is preferred over trying a different TNF inhibitor.

Of the DMARDs, methotrexate has been the standard therapy for children with polyarticular disease [2,5-9]. The usual starting dose of methotrexate is 10 to 15 mg/m2 body surface area (BSA) per week. Methotrexate is effective for many patients, but it does not work quickly, usually taking four to eight weeks before demonstrating its benefits. The trial periods for methotrexate are customary because of the higher risks and costs associated with the biologic agents compared with methotrexate. However, biologic agents have a more rapid onset of action and appear to be more efficacious than methotrexate. Etanercept is the first of the TNF inhibitors approved for clinical use, with a relatively large amount of data regarding long-term safety and efficacy. Standard dosing for etanercept is 0.8 mg/kg once a week or 0.4 mg/kg twice a week. Other TNF inhibitors such as adalimumab and golimumab have since been approved for use in polyarticular JIA. There are no data to indicate that one TNF inhibitor is more efficacious than another for polyarticular JIA. (See 'Tumor necrosis factor inhibitors' below and 'Methotrexate' below.)

The question as to whether TNF inhibitors, with their potentially faster onset of action and greater efficacy, should be used before methotrexate remains unresolved. There is also evidence that the combination of a TNF inhibitor and methotrexate is synergistic for the treatment of arthritis [5,10-15]. These considerations must be balanced against the potential increased risk of opportunistic infections with the use of TNF inhibitors, especially when used as part of multiple drug therapy. Neither TNF inhibitor monotherapy nor TNF inhibitor plus methotrexate combination therapy increased the risk of infections during or resulting in hospitalization compared with methotrexate monotherapy in a large administrative database study of children with JIA [16]. In addition, there is not yet a clear answer to the question of whether benefits of biologic agents justify their significantly higher costs [17,18]. Ongoing studies aimed at identifying markers of response and risk factors for adverse events should improve selection of appropriate candidates for the use of biologic agents and help answer and resolve some of these important issues. (See 'Methotrexate' below.)

Many pediatric rheumatologists start with a biologic agent before or concurrent with methotrexate in children with high disease activity or poor prognostic features (see 'Pretreatment assessment' above) if there are no insurance restrictions with regard to starting a biologic before a trial of methotrexate [2]. Of the biologics, a TNF inhibitor is typically started first, although there is no evidence to guide choice of one biologic over another early in the disease course. In addition, children with longer disease duration, negative antinuclear antibody (ANA) titers, higher level of disability, and arthritis present in both wrists are less likely to respond to methotrexate alone [19], and methotrexate is ineffective in predominantly axial arthritis.

Some centers are using biologic agents at the start of methotrexate therapy, with the goal of preventing additional joint damage during the period necessary for methotrexate to take effect. In a 2013 survey of 138 pediatric rheumatologists, 46 and 9 percent of providers stated that they would use a nonbiologic and a biologic DMARD as initial therapy for children with polyarticular arthritis with and without poor prognostic features, respectively, regardless of disease activity [20]. Less than 1 percent would start a biologic agent alone for polyarticular disease. Labeling indications for the approved anti-TNF therapies (adalimumab, etanercept, golimumab), abatacept, tocilizumab, and the kinase inhibitor tofacitinib do not require failure to respond to methotrexate before their use in children with JIA. All of the biologic agents can be used as monotherapy or in combination with a DMARD such as methotrexate. However, combination with other biologics, kinase inhibitors, or other immunosuppressants such as cyclosporine or azathioprine is not recommended. Ongoing trials should help clarify the optimal approach to new-onset polyarthritis in children [21]. (See 'Biologic agents' below.)

Additional therapies — Additional modes of treatment for polyarticular JIA include NSAIDs, intraarticular glucocorticoids, and physical/occupational therapy.

Nonsteroidal antiinflammatory drugs — An NSAID such as ibuprofen (10 mg/kg every eight hours, maximum dose 3200 mg/day), naproxen (10 to 15 mg/kg/day in two divided doses, maximum dose 1000 mg/day), or indomethacin (1 to 2 mg/kg/day in two to three divided doses, maximum dose 200 mg/day) may help to provide symptomatic relief [2,22,23]. NSAIDs reach full efficacy within two to three months but usually start to relieve symptoms within a few days [22].

Unfortunately, there is no way to predict which NSAID is most likely to benefit an individual patient. A process of trial and error must be used while keeping expectations appropriate. Patients should be aware that NSAIDs are useful for symptomatic relief of pain and stiffness, but they do not delay or prevent joint damage in JIA. Long-term safety data on NSAIDs are needed because potential adverse effects, especially cardiovascular events, may not appear until adulthood [24]. However, an email survey of the pediatric rheumatology community found no recognized cases of children treated with NSAIDs who developed premature myocardial infarctions.

Intraarticular glucocorticoids — Intraarticular injections of glucocorticoids are both safe and effective for children with polyarticular JIA, and their use is recommended as needed for continued disease activity [25-27]. However, the need to repeatedly inject the same joint, or to inject multiple joints simultaneously, suggests that more effective systemic therapy is required. With the availability of anti-TNF agents, non-TNF inhibitor biologics, or kinase inhibitors, the simultaneous injection of multiple joints is rarely necessary.

Injection of intraarticular glucocorticoids into a single inflamed joint, such as the hip or temporomandibular joint, is often dramatically effective [28-30]. The injections are performed under general anesthesia in young children, or local anesthesia in older children, with computed tomography or ultrasound-guided imaging, or without imaging by an experienced clinician, depending upon the joint injected. (See "Joint aspiration or injection in children: Indications, technique, and complications".)

Folic acid — Folic acid supplementation (400 micrograms daily) is recommended for all children with JIA receiving methotrexate, given the beneficial effects noted and the lack of convincing contrary data [5]. If the child experiences methotrexate side effects on 400 micrograms of folic acid daily, the dose of folic acid is typically increased to 1 mg/day. Some side effects such as glossitis and macrocytic anemia are often avoided by the addition of supplemental folic acid, started at the same time as methotrexate. Whether high doses of folic acid also diminish the effectiveness of methotrexate or cause side effects are topics of debate. Published articles report conflicting results regarding potential antagonism of the beneficial effects of methotrexate by folate supplementation [31]. Similarly, studies have shown both an increase [32] and a decrease [33] in the incidence of cancer among adults receiving folic acid supplementation. The effects appear to depend upon a variety of factors, including characteristics of the specific population studied [34]. However, no adverse effects have been demonstrated to date in children receiving supplemental folic acid to minimize methotrexate toxicity. Leucovorin (folinic acid; usual dose 5 mg once a week) is an alternative to folic acid.

Physical and occupational therapy — Physical therapy plays an important role in the rehabilitation of any child with polyarticular disease. However, range of motion and stretching exercises are of limited benefit in the face of ongoing joint inflammation. In addition, the efficacy and availability of the biologic agents have allowed many children to regain full function in a short period of time, limiting the need for rehabilitation. Nonetheless, any child with persistent weakness or flexion contractures should be referred for physical and/or occupational therapy. Physical therapy typically targets the larger joints and lower-extremity joints needed for movement while occupational therapy targets the ability to perform activities of daily living (eg, writing in school).

Ongoing management — Ongoing management includes monitoring for drug side effects, trial off medications at some point after inactive disease is attained, and treatment of refractory or recurrent disease. Inactive disease is defined as meeting the following criteria [35]:

No active arthritis

No fever, rash, serositis, splenomegaly, or lymphadenopathy attributable to JIA

No active uveitis

Normal inflammatory markers

Global assessment of disease activity by clinician indicates no activity

Monitoring on DMARD therapy — Monitoring of liver enzymes, creatinine, and complete blood counts (CBCs) is recommended prior to methotrexate initiation, four to eight weeks after initiation, and every three to four months thereafter [2,5]. Supplemental folic acid or leucovorin (folinic acid) is given to minimize methotrexate toxicity. (See 'Methotrexate' below and 'Folic acid' above.)

A CBC and comprehensive metabolic panel (CMP) are recommended prior to initiation of all biologics and kinase inhibitors. A baseline lipid panel is also recommended for tocilizumab and kinase inhibitors. Labs (CBC and CMP) should be checked again four to eight weeks after therapy starts and then every three months thereafter. Lipids should be rechecked four to eight weeks after starting tocilizumab or a kinase inhibitor and then periodically. Skin testing for tuberculosis prior to initiation of biologics and kinase inhibitor therapy is standard. (See 'Tumor necrosis factor inhibitors' below.)

Duration of therapy — The amount of time that methotrexate [5] and biologics should be continued in children with polyarticular disease is unclear since clinical outcomes following medication withdrawal have not been studied prospectively. Medications are typically tapered off after a period of clinical inactivity between six months and two years while on medication. The decision regarding which medication to stop first is individualized depending upon response to the medication, tolerance of the administration regimen, and other patient factors.

In a large, retrospective study of 171 children with juvenile arthritis, the median time to biologic discontinuation after attainment of inactive disease was six months (range 0 to 69 months) [36]. In this cohort, one-half and two-thirds had relapse of arthritis by 6 and 12 months, respectively. There was also no significant association in this study between the duration of therapy after inactive disease was achieved or total duration of biologic therapy prior to discontinuation and time to disease relapse.

In a 2015 survey of North American pediatric rheumatologists, 75 percent of participants stopped methotrexate or a biologic 6 to 12 months after attainment of inactive disease [37]. Sixty-three percent stop methotrexate before a biologic. RF positivity made clinicians less likely to withdraw therapy.

Refractory disease — Patients are considered to have refractory disease if they have persistent disease activity following four months of treatment with a TNF inhibitor and methotrexate. A trial of an alternate TNF inhibitor agent or another biologic agent (tocilizumab or abatacept) or kinase inhibitor (tofacitinib or possibly baricitinib) is indicated in these patients [2,38,39].

Recurrent disease — Relapse is common within 6 to 12 months of withdrawal of therapy after remission. Patients are restarted on their previous regimen. The regimen is altered if they fail to recapture disease inactivity on medication.

Immunizations — The administration of live-viral vaccines and other standard childhood immunizations in patients with JIA is discussed in detail separately. (See "Oligoarticular juvenile idiopathic arthritis", section on 'Immunizations'.)

Rheumatoid factor-positive patients — Adolescents with RF-positive polyarthritis probably represent the early onset of adult-type disease and are at significant long-term risk for progressive arthritis and the other complications normally associated with adult-onset rheumatoid arthritis (RA) [40,41]. Thus, these children should be treated similarly to adults with moderate-to-severe RA. (See "Initial treatment of rheumatoid arthritis in adults".)

Joint damage in RF-positive polyarticular JIA occurs earlier, leading many investigators to believe that the therapeutic pyramid should be inverted, with early and aggressive treatment of these patients. Studies of early, aggressive therapy in adults with RA confirm better responses and improved joint preservation [42-44]. Ongoing therapeutic trials in children are attempting to determine whether a similar approach involving treatment with disease-modifying agents should be followed in JIA patients at high risk for progressive joint destruction.

DISEASE-MODIFYING ANTIRHEUMATIC DRUGS — The primary disease-modifying antirheumatic drugs (DMARDs) used for polyarticular JIA are methotrexate and the tumor necrosis factor (TNF) inhibitors. The other biologic agents and kinase inhibitors are usually reserved for patients with refractory disease who have failed therapy with a TNF inhibitor. Several of the older agents, such as cyclosporine, azathioprine, and systemic glucocorticoids, are rarely used with the advent of more effective agents.

Methotrexate — Methotrexate is an immunomodulator and acts as an inhibitor of purine synthesis, although other mechanisms of action appear to account for its effects at the doses used to treat JIA [5,45]. At these doses (typically 10 to 15 mg/m2/week, maximum dose 25 mg), methotrexate acts as an antiinflammatory agent rather than as a cytotoxic drug.

Efficacy — The question of whether treatment of early polyarticular JIA with either methotrexate alone or methotrexate in combination with a biologic agent and systemic glucocorticoid is effective was examined in a randomized trial of 85 children aged 2 to 16 years of age [10]. Children were randomly assigned to combination therapy (n = 42) with methotrexate (0.5 mg/kg/week subcutaneously [SC], maximum dose 40 mg); etanercept (0.8 mg/kg/week SC, maximum dose 50 mg); and prednisolone (0.5 mg/kg/day orally, maximum dose 60 mg, tapered off by 17 weeks) or methotrexate alone (n = 43) with etanercept and prednisolone placebos. Both regimens were moderately effective, although there was a trend toward greater efficacy in the initial combination group compared with methotrexate monotherapy that was significant only at the six-month time point. At four months, 30 (71 percent) children in the combination-therapy group had inactive disease compared with 19 (44 percent) in the methotrexate-only group. By six months, only 17 (40 percent) and 9 (21 percent) had inactive disease in the combination and methotrexate groups, respectively. At 12 months, 9 of the 17 children in the combination group had attained clinical remission, and the remainder had inactive disease. In comparison, three children were in clinical remission and four had inactive disease at 12 months in the methotrexate group. Five children in the combination group withdrew over the course of the study: three due to adverse events, one because of an inadequate response, and one due to a disease flare. Eleven children withdrew from the methotrexate group: four due to an adverse event, five because of an inadequate response, one due to a disease flare, and one due to a fear of needles.

Methotrexate is generally ineffective for the treatment of axial arthritis, as may be found in nonradiographic axial spondyloarthritis or ankylosing spondylitis [46]. Thus, it is important to recognize that children with spondyloarthritis are distinct from children with polyarthritis so that they may be advanced to appropriate biologic therapy without the delay and possible toxicity associated with a trial of methotrexate. (See "Spondyloarthritis in children".)

Duration of treatment — Treatment is usually withdrawn 6 to 12 months after attaining inactive disease. However, the relapse rate is high, and there is risk of ongoing joint damage with each relapse. This raises the question of whether the appropriate response is to continue therapy rather than withdraw it.

An open, randomized trial of methotrexate withdrawal after remission found no difference in relapse rates between 12- versus 6-month withdrawal [47]. Both groups experienced a roughly 60 percent rate of relapse, although patients with higher levels of the phagocyte activation marker myeloid-related proteins 8 and 14 heterocomplex (MRP8/14) were at greater risk of relapse after discontinuation of methotrexate. These tests are not routinely available and hence of limited utility. Further studies are needed to identify biomarkers predicative of future relapse.

In a multicenter prospective study of TNF inhibitor withdrawal in children with polyarticular JIA and inactive disease for at least six months, 40 percent of children flared within eight months, with a median time to flare of approximately seven months [48]. Significant predictors of flare included longer duration of disease at enrollment, younger age at diagnosis, and longer disease duration prior to the first episode of inactive disease. In another study evaluating etanercept withdrawal in children with a rheumatoid factor (RF) negative polyarticular course and inactive disease for at least six months, 60 percent of patients flared a median of 4.3 months after the last dose of medication [49]. In this study, only male sex and elevated baseline C-reactive protein (CRP) were significantly associated with flare.

Adverse effects — Potential side effects of methotrexate are many. Any evidence of significant toxicity requires a dose decrease or discontinuation of methotrexate. The more common side effects include leukopenia or thrombocytopenia and elevation of hepatic transaminases (especially if ethanol is consumed). Methotrexate is teratogenic, so safe-sex counseling is recommended for sexually active adolescents. Methotrexate may also cause nausea and vomiting within 24 to 48 hours of administration. Folic acid or leucovorin (folinic acid) supplementation (see 'Folic acid' above), administration of methotrexate on an empty stomach, and/or dose adjustment can help with the gastrointestinal side effects.

Rarer adverse effects include the development of pulmonary hypersensitivity reactions, nodulosis [50], and, possibly, malignancies [51]. In practice, however, dose-limiting side effects are rare [52]. Similarly, although long-term data in children are limited, meta-analyses in adults suggest that the chronic use of methotrexate in rheumatoid arthritis (RA) is associated with a minimally increased risk of infections, no increase in the incidence of opportunistic infections, no clear increase in the risk of malignancies, and an apparent advantage in longevity and decreased cardiovascular mortality [53].

A retrospective study examined the safety and efficacy of methotrexate regimens at two medical centers in the same geographic region, one of which started patients on an initial dose of >0.5 mg/kg/week (similar, but not identical to 15 mg/m2) and the other which began patients on ≤0.5 mg/kg/week (similar to 10 mg/m2) [54]. The high-dose group was more likely to have elevated liver enzymes but was no more likely to have a decreased active joint count.

Genotyping may help predict the risk of methotrexate toxicity. Single-nucleotide polymorphisms (SNPs) in genes within the methotrexate metabolic pathway, such as the C677T allele of the methylenetetrahydrofolate reductase gene, are associated with increased methotrexate-adverse effects [55,56]. However, these tests are not routinely available.

Biologic agents — The most common biologic agents used to treat polyarticular JIA are the TNF inhibitors (eg, etanercept, adalimumab, infliximab, golimumab). Other biologic agents act by blocking the interleukin (IL) 6 receptor, T cell costimulation, or CD20.

Tumor necrosis factor inhibitors — TNF inhibitors, including etanercept, adalimumab, golimumab, and infliximab, often provide rapid control of otherwise disabling arthritis, restoring quality of life and preventing further joint destruction. However, they are associated with an increased risk of infections [57,58]. There were initial concerns about a possible association between treatment with TNF inhibitors and malignancy, but subsequent data have suggested that the risk of malignancy is associated with underlying JIA rather than the medications [59]. These potential side effects are reviewed in greater detail separately. (See "Overview of biologic agents in the rheumatic diseases" and "Tumor necrosis factor-alpha inhibitors: Risk of malignancy" and "Systemic juvenile idiopathic arthritis: Treatment", section on 'Biologic disease-modifying antirheumatic drug (DMARD) therapy' and "Systemic juvenile idiopathic arthritis: Treatment", section on 'Tumor necrosis factor alpha inhibitors'.)

In adults, TNF inhibitors increase the risk of opportunistic infections [60]. In children, there is also an increased risk of fungal infections, especially in endemic areas of histoplasmosis such as the Ohio and Mississippi River valleys, with the use of anti-TNF agents [61]. Clinicians in areas with endemic fungal infections should monitor their patients appropriately. (See "Pathogenesis and clinical manifestations of disseminated histoplasmosis" and "Pathogenesis and clinical features of pulmonary histoplasmosis".)

Etanercept — Etanercept, a fusion protein containing TNF receptor 2 and the Fc domain of immunoglobulin G1 (IgG1), is effective in many children with resistant polyarticular disease, including those under four years of age [11,25,62-64]. Dramatic improvements may be seen within weeks of starting therapy, with benefits persisting during years of use. Most patients respond promptly, although some may have a delayed clinical response [65]. The available data favor the use of biologics that block TNF compared with other therapeutic regimens in children with refractory polyarticular JIA [66-68]. Standard SC dosing is 0.8 mg/kg once a week or 0.4 mg/kg twice a week.

As the first of the TNF inhibitors in clinical use, there is a relatively large amount of data regarding the long-term safety and efficacy of etanercept [67]. In patients who remain on therapy, the clinical benefits are substantial, and significant toxicity is uncommon. Symptoms improve in a sizable proportion of patients with refractory disease [62]. Growth also improves [69]. No cases of malignancy have been reported in these follow-up studies of children treated with etanercept for JIA. There have been case reports of inflammatory bowel disease (IBD) in JIA patients treated with etanercept, although these children might well have had unrecognized IBD at the time they were diagnosed with polyarthritis rather than bowel disease caused by etanercept [70,71].

In one open-label study of 69 children who failed therapy with methotrexate, for example, the administration of etanercept at a dose of 0.4 mg/kg twice a week resulted in a response (defined as >30 percent improvement) in nearly 75 percent of patients within three months [72]. A flare subsequently occurred in 28 percent of those continuing to receive etanercept versus 81 percent in whom placebo was substituted in a randomized and blinded fashion. Thus, in most children, the effects of etanercept cease within a few weeks of its discontinuation.

Subsequent studies reviewed the safety and efficacy of long-term etanercept therapy in the 58 patients who elected to continue therapy after the initial phase of the study [73-75]. At the four-year follow-up, 34 of the 69 original patients remained on therapy, whereas only 20 remained at the eight-year follow-up. Not surprisingly, those who remained on therapy had a significant improvement in symptoms and disease activity. Most patients who still required treatment with glucocorticoids were able to taper to a lower dose. Severe infections were the most common serious adverse event reported in the first four years, while disease flares were the most common in the second four years of treatment.

In another small pilot study, bone mineral status improved in patients who had responded to and continued etanercept treatment for more than one year compared with those who failed to respond [76]. In addition, anti-TNF therapy appears to improve growth velocity in patients with delayed growth [77,78]. Improved linear growth, bone mineralization, and body composition were seen in another small study [79]. These effects are probably due to decreased inflammation resulting from control of arthritis. In general, similar effects to those of etanercept should be expected from other TNF antagonists, such as adalimumab and infliximab.

Adalimumab — Adalimumab is a fully humanized monoclonal anti-TNF antibody that is administered either weekly or every other week as a single SC injection. Standard dosing is 10 mg, 20 mg, or 40 mg every other week for children less than 15 kg, 15 to 30 kg, and 30 kg or greater, respectively. Adalimumab is effective in some children who have responded inadequately to etanercept.

In 2008, adalimumab was approved by the US Food and Drug Administration (FDA) for the treatment of moderate-to-severe JIA in patients ≥4 years of age. Approval was based upon data from a trial of 171 patients who were stratified into two groups: those treated with methotrexate and those not treated with methotrexate [80]. After an open-label lead-in phase of 16 weeks with adalimumab therapy, patients with an American College of Rheumatology Pediatric (ACR Pedi) 30 response were randomly selected to receive adalimumab at a dose of 24 mg/m2 every other week or placebo for an additional 32 weeks. In the second phase of the study, patients who received adalimumab, compared with those who received placebo, experienced fewer disease flares regardless of whether they received methotrexate (37 versus 65 percent) or not (43 versus 71 percent). In this trial, severe adverse effects included neutropenia and infections (herpes simplex and zoster, urinary tract infection, pneumonia, and pharyngitis). No malignancies were reported. In 2014, the indication was extended to include children ages two to four years of age with moderate-to-severe polyarticular JIA [81].

In the 360-week long-term extension study, adalimumab remained effective and well tolerated with or without concomitant methotrexate therapy [82]. Of the 171 patients enrolled into the phase III trial [80], 62 (36 percent) completed the long-term extension study. Thirty-seven percent attained clinical remission for more than six months during the study. Seventy-three percent (observed) achieved a 27-joint Juvenile Arthritis Disease Activity Score (JADAS27) low disease activity assessment (44 percent by nonresponder imputation). Clinical response was typically sustained through week 312. Two-hundred thirty-five adverse events and 3.2 serious adverse events per 100 person-years were reported as possibly related to adalimumab exposure; incidence of adverse events and serious adverse events were similar among those receiving and not receiving concomitant methotrexate. Serious adverse events included herpes zoster, pharyngitis, genital herpes, and pneumonia. No malignancies, tuberculosis, or lupus-like syndromes were reported.

Golimumab — Golimumab is a fully human monoclonal antibody with affinity for TNF-alpha that is available in both intravenous (IV) and SC formulations. The IV formulation (80 mg/m2 dosed at weeks 0 and 4 and then every eight weeks) was approved by the US FDA in 2020 for the treatment of polyarticular JIA based upon extrapolation of efficacy data from adults with RA and a small, open-label study in children with polyarticular JIA who had not responded to methotrexate alone [83]. One-hundred twenty-seven children who had persistently active disease despite at least two months of methotrexate monotherapy were treated with open-label golimumab (80 mg/m2) at weeks 0 and 4, and then every eight weeks thereafter through week 52. At week 28, 47, 70, and 80 percent achieved a JIA ACR 90, 70, or 50, respectively. Serious adverse events include infection (6 percent of patients) and death (one due to septic shock).

SC golimumab has also been studied in children with polyarticular JIA who are refractory to methotrexate [84]. During the 16-week open-label golimumab lead-in period, subjects demonstrated a rapid improvement in disease activity, with 34 percent of subjects attaining inactive disease. At the end of this period, patients were randomly assigned to continue golimumab or start placebo. At week 48, the treatment and placebo groups had comparable rates of disease flares and clinical remission. The reasons for failure to reach the primary endpoint remain unclear but may have been related to low mean baseline CRP in both groups, which, in turn, may have been associated with fewer disease flares regardless of treatment.

Infliximab — Infliximab is a chimeric mouse-human monoclonal antibody with affinity for TNF-alpha. It has been evaluated in combination with methotrexate in clinical trials in patients with recent-onset polyarticular JIA who are refractory to methotrexate monotherapy. It is not approved by the US FDA for the treatment of JIA due to failure to achieve the primary efficacy endpoint at three months in a randomized, placebo-controlled, double-blind study that used standard 3 mg/kg dosing [85]. However, infliximab is still used by some experts in combination with methotrexate because clinical experience suggests that a higher dose (10 mg/kg) is effective [86]. In addition, some patients and their parents/caregivers prefer infusions over injections.

Most centers report an increased incidence of side effects in children treated with infliximab relative to etanercept [85,87,88]. In general, etanercept is better tolerated and more easily administered than infliximab. High doses of infliximab (>5 mg/kg) may be more effective than the standard dose of infliximab (3 mg/kg/dose), but they may also carry increased risks. Further trials are required to characterize the efficacy and safety of various dosing regimens of infliximab in JIA.

In a multicenter, randomized trial, 122 children with polyarticular JIA who had not responded to methotrexate therapy were randomly assigned to one of two treatment regimens [85]. Patients received methotrexate plus infliximab 3 mg/kg at 0, 2, 6, and 14 weeks; placebo at week 16; and then infliximab 3 mg/kg at week 20 and then every 8 weeks through week 44 or methotrexate plus placebo for 14 weeks followed by methotrexate plus infliximab 6 mg/kg every 8 weeks through week 44. The following findings were noted:

During the first 14 weeks, there was a nonsignificant trend towards greater improvement in the ACR Pedi criteria among patients who initially received infliximab 3 mg/kg compared with those who received placebo (64 versus 49 percent).

By week 16, when the placebo-treated patients crossed over to infliximab 6 mg/kg, 73 percent of all patients had achieved a 30 percent improvement in the ACR Pedi criteria.

By week 52, 70 and 52 percent of all patients had achieved a 50 and 70 percent improvement in the ACR Pedi criteria, respectively.

Antibodies to infliximab developed in 25 percent of the 102 patients who were evaluated. A higher proportion of antibody formation was seen in the 3 mg/kg compared with the 6 mg/kg infliximab groups (38 versus 12 percent). Similarly, overall adverse events were more frequent among those who received 3 mg/kg infliximab versus those receiving 6 mg/kg.

Adverse events were greater in patients who received infliximab compared with those who received placebo. They included infection (67 versus 47 percent) and infusion-associated reactions (35 versus 8 percent), such as vomiting, fever, headache, and hypotension. There were two deaths. One patient died during the placebo phase, and one patient receiving the lower dose of infliximab also died.

Seventy-eight patients (64 percent) from this study entered an open-label treatment extension [87]. Eight children discontinued infliximab due to lack of efficacy, and 19 patients stopped the drug due to reasons unrelated to efficacy or safety. By week 204, 40 and 33 percent had a 50 and 70 percent improvement in the ACR Pedi criteria, respectively. Infusion reactions continued to occur at a similar rate (32 percent) as in the randomized phase of the study. These reactions occurred more frequently in those positive for antibodies to infliximab.

In a randomized, open-label study, three regimens were compared for the treatment of recent-onset polyarticular JIA: infliximab (3 to 5 mg/kg) plus methotrexate (n = 19), methotrexate alone (n = 20), and a DMARD combination (methotrexate, sulfasalazine, and hydroxychloroquine; n = 20) [89]. ACR Pedi 75 percent improvement (ACR Pedi 75) was achieved in all patients on infliximab plus methotrexate, 65 percent on the DMARD combination, and 50 percent on methotrexate alone. Inactive disease was attained in 68, 40, and 25 percent of the same treatment groups, respectively.

Other biologic agents — Other biologic agents that are options in patients with refractory disease include tocilizumab, abatacept, rituximab, and intravenous immune globulin.

Tocilizumab — Tocilizumab, a humanized monoclonal antibody against the IL-6 receptor, is approved for the treatment of moderate-to-severe polyarticular JIA in children ages two years and older. It has been used in clinical trials in children who are refractory or intolerant to methotrexate [38,39]. Both IV and SC administration of tocilizumab are US FDA approved for polyarticular JIA.

In part I of a multicenter, randomized withdrawal study, 188 children with polyarticular JIA who had not responded to methotrexate therapy received tocilizumab (<30 kg: 10 mg/kg or >30 kg: 8 mg/kg IV every four weeks) [38]. In part II, the 163 patients who achieved an ACR Pedi 30 response in part I were randomized to receive tocilizumab or placebo. The following findings were noted:

In part I, the open-label lead-in period, 89, 62, and 26 percent of children achieved an ACR Pedi 30, 70, or 90 response, respectively.

In part II, the randomized withdrawal study, there were significantly more flares in the placebo group versus the treatment group (38 percent versus 26 percent, p = 0.002).

Use of methotrexate statistically improved the ACR Pedi 70 and 90 response rates in both the placebo and treatment groups.

There were 16 severe, adverse events in the treatment group, 7 of which led to study withdrawal. Commonly reported adverse events included pneumonia (n = 4), bronchitis (n = 2), and cellulitis (n = 2). There were no deaths and no malignancies. Laboratory abnormalities included transaminitis, neutropenia, and thrombocytopenia. None of the noted laboratory abnormalities were associated with infection or bleeding. Increased low-density lipoprotein (LDL) and total cholesterol levels were noted in 11 and 34 percent of patients, respectively; none required lipid-lowering therapy.

One-hundred sixty of 166 patients in part II entered part III, during which all children received open-label tocilizumab [90]. By week 104, 59.6 and 80.3 percent achieved the ACR 90 and 50, respectively, and 51 percent achieved inactive disease. No new safety signals were revealed.

In an open-label, multicenter study in Japan, 19 children with polyarticular JIA, ages 2 to 19 years, who failed prior therapy or were intolerant to other medications were given three IV doses of tocilizumab (8 mg/kg) every four weeks [39]. The following findings were noted:

Sixty-three, 90, and 95 percent of patients achieved an ACR Pedi 30 at weeks 4, 8, and 12, respectively.

Nine and 58 percent attained an ACR Pedi 50 and 70 at week 12, respectively.

In the 48-week open-label extension period, the ACR Pedi responses were sustained.

In two 52-week phase Ib trials, 52 patients ages 1 to 17 years with polyarticular JIA were given SC tocilizumab every two weeks (≥30 kg) or every three weeks (<30 kg) [91]. Pharmacokinetics, pharmacodynamics, and efficacy assessments were comparable between the SC and IV formulations. By week 52, 31 percent of patients attained clinical remission.

Abatacept — Abatacept blocks T cell costimulation, which interferes with proinflammatory messages being passed to and from T cells. SC abatacept is approved by the US FDA for the treatment of moderate-to-severe polyarticular JIA in children ages two years and older. IV abatacept is approved for use in moderate-to-severe polyarticular JIA in children age six years and older.

The pivotal study demonstrated a decrease in the rate of arthritis flares with abatacept compared with placebo in a trial of 190 patients between 6 and 17 years of age who had a history of active disease in five joints and an inadequate response to at least one DMARD [92]. In this study, 170 patients who completed the open-label lead-in period of four months of abatacept therapy and had a positive response to the drug were randomly selected to continue to receive 10 mg/kg of abatacept every 28 days or placebo for an additional six months or until their arthritis flared. Patients who received abatacept, compared with those who received placebo, experienced fewer disease flares (20 versus 53 percent). There was no difference in the frequency of adverse events between the two groups. Greater improvements were also seen in health-related quality-of-life measures, particularly pain/discomfort and sleep, in the abatacept group compared with the placebo group [93].

In a subsequent phase III, open-label trial, 173 children aged 6 to 17 years and 46 children aged 2 to 5 years with polyarticular JIA who had failed therapy with at least one DMARD were treated with weight-tiered SC abatacept weekly (50 mg if 10 to ≤25 kg, 87.5 mg if 25 to ≤50 kg, and 125 mg if ≥50 kg) for four months and then continued up to 24 months if they had reached at least an ACR Pedi 30 [94]. Approximately 80 percent of the children in both age groups were receiving concomitant methotrexate at baseline. In an intention-to-treat analysis, JIA-ACR30/50/70/90/100/inactive disease responses at four months were 83/73/53/28/14/30 percent in the older children and 89/85/74/59/41/50 percent in the younger children, with responses sustained to month 24. The Juvenile Arthritis Disease Activity Score 71-C-reactive protein (JADAS71-CRP) median scores improved from baseline to month 4 and were maintained to month 24, at which time 16 percent of the older children and 50 percent of the younger children had achieved remission based upon this score. Serious adverse events were reported in 6 to 8 percent of the children.

Rituximab — Rituximab, a chimeric anti-CD20 monoclonal antibody, has been approved for the treatment of adult-onset RA [95,96]. Although most subsets of JIA are not the same disease as adult RA, in limited experience, adolescents with RF-positive and anticyclic citrullinated peptide (CCP) positive disease unresponsive to other agents have benefited from rituximab. The utility of this agent in other settings is unknown. There are anecdotes of its successful use in other subtypes of JIA, but there are no reliable data to date regarding its efficacy in patients with refractory JIA. (See "Rituximab: Principles of use and adverse effects in rheumatoid arthritis".)

Intravenous immune globulin — IV immune globulin has been studied in polyarticular JIA. While it appears to have some efficacy, its effects are short lived and insufficient to justify its cost and potential toxicity [97].

Small molecule inhibitors — Small molecule inhibitors that are options in patients with active disease include Janus kinase (JAK) inhibitors such as tofacitinib and baricitinib.

Tofacitinib — Tofacitinib, a JAK inhibitor, was approved by the US FDA in 2020 for the treatment of polyarticular JIA based upon a small randomized trial [98]. Patients aged 2 to 17 years with polyarticular JIA not responsive or intolerant to methotrexate or a biologic agent were treated with oral tofacitinib (3.2 mg twice daily for body weight 10 kg to <20 kg, 4 mg twice daily for body weight 20 kg to <40 kg, and 5 mg twice daily for body weight ≥40 kg) for 18 weeks. At the end of this run-in phase, patients with at least a 30 percent improvement in the ACR JIA response criteria (ACR30) were randomly assigned to tofacitinib or placebo. In both phases, approximately two-thirds of patients were on concomitant methotrexate, and one-third were on oral glucocorticoids. At week 44, significantly fewer treated patients had a disease flare compared with the placebo group (31 versus 55 percent, respectively; difference in proportion -25 percent [95% CI -39 to -10 percent]).

Baricitinib — The safety and efficacy of baricitinib was studied in a randomized trial of 220 patients aged 2 to 17 years old with JIA (66 percent with polyarticular JIA) who had inadequate response or intolerance to standard therapy [99]. After a 2-week safety period and a 10- to 12-week open-label period during which all patients received baricitinib, 163 patients who had at least an ACR30 were randomly assigned to placebo (n = 81) or baricitinib (n = 82; dosing 2 mg daily for patients 2 to 8 years old and 4 mg daily for patients 9 to 17 years old) for an up to 32-week double-blind withdrawal period. Time to disease flare was shorter in the placebo group than baricitinib group (adjusted hazard ratio 0.241, 95% CI 0.128-0.453), and more patients in the placebo group had a flare during this period (disease flare rate 51 percent compared with 17 percent in the baricitinib group). A greater number of infections were reported in the baricitinib group than placebo during the withdrawal period (38 versus 19 percent, respectively), but rates of serious adverse events were similar in both groups. Pulmonary embolism reported in one patient on baricitinib was felt to be study drug related.

Additional agents — Additional DMARDs that may be useful in patients with polyarticular JIA who have contraindications or intolerance to methotrexate include sulfasalazine and leflunomide. Long-term systemic glucocorticoids are generally not recommended, but short-term, low-dose glucocorticoids and/or intraarticular glucocorticoids may provide rapid symptom relief for arthritis that is particularly disabling in one or more joints. Cyclosporine and azathioprine are rarely used, and gold compounds are no longer used now that more effective drugs (biologic agents) are available.

Sulfasalazine — Sulfasalazine has been shown to be beneficial for many children with polyarticular JIA [100-103].

This utility was illustrated in a randomized 24-week trial of 69 patients with oligoarticular and polyarticular JIA [101]. Sulfasalazine resulted in significant improvement compared with placebo, as determined by overall articular severity score, global assessments, and laboratory parameters. Adverse events, which were found in one-third of patients, were transient or reversible upon cessation of treatment [101,102].

A subsequent study reported the clinical status of this cohort of patients at a median follow-up of nine years [103]. Patients who were in the sulfasalazine treatment group compared with those treated with placebo appeared to require less intensive DMARD treatment based upon a shorter duration of sulfasalazine (2.5 versus 5.2 years) and a trend to less use of methotrexate and other DMARDs. At follow-up assessment, outcome scores were better in the group treated with sulfasalazine compared with the control group. However, more than one-third of patients in both groups reported long periods of noncompliance with therapy, which significantly impacted patient outcomes.

In using sulfasalazine, parents/caregivers must be warned of the possible development of rare, severe reactions seen with sulfa drugs (eg, Stevens-Johnson syndrome). White blood cell (WBC) count, platelet count, and liver enzymes must be periodically monitored as well. Sulfasalazine does not prevent chronic joint changes and, therefore, should not be relied upon in erosive disease. For those with erosive disease, methotrexate or anti-TNF therapies are preferable.

Leflunomide — Leflunomide, an immunomodulator that inhibits pyrimidine synthesis, has been shown to be safe and effective in adults with RA. Although not US FDA approved for pediatric use, efficacy in JIA has been reported but has not been shown to be superior to methotrexate [9,104,105].

In a randomized study of 94 children with JIA, both methotrexate and leflunomide resulted in clinically meaningful improvements [9]. Patients were treated initially with either methotrexate or leflunomide for 16 weeks, with blinded treatment extended for an additional 32 weeks. Both medications resulted in improved articular severity score, global assessments (clinician and patient), and erythrocyte sedimentation rate. Methotrexate appeared to be more effective than leflunomide since a greater percentage of children demonstrated clinical improvement, although the dose of leflunomide used in this study was somewhat low. The most common adverse events for both medications included gastrointestinal symptoms (abdominal pain, nausea, vomiting, or diarrhea), headache, and nasopharyngeal symptoms (eg, pharyngitis). Children on either medication need to be carefully monitored for evidence of hepatic toxicity.

Leflunomide was added to the regimen in one observational study of 32 children with polyarticular JIA who failed methotrexate therapy [106]. At six months, 85 percent achieved an ACR Pedi 30 response. At last follow-up (range: 0.3 to 3 years), 12 of 18 (66 percent) met the ACR Pedi 30 criteria, and 9 of 18 (50 percent) were in clinical remission on medication.

Systemic glucocorticoids — Long-term, high-dose oral glucocorticoids should be avoided in children with polyarticular JIA because of the high incidence of side effects, particularly growth retardation and osteoporosis. Some have advocated pulsed doses of glucocorticoids as being effective in controlling inflammation without causing side effects [25]. Others, however, have noted that over 20 percent of patients receiving pulsed-dose glucocorticoids had one or more adverse reactions [25].

Short-term use of low-dose glucocorticoids (less than 0.25 mg/kg per day of prednisone or its equivalent) may provide substantial benefits for the child who is otherwise incapacitated by arthritis without causing excessive complications. However, most clinicians have found anti-TNF agents to provide an equally rapid clinical response in children with severe JIA. Children with JIA severe enough to warrant consideration of glucocorticoids may ultimately require anti-TNF therapy. Thus, it may be preferable to begin with anti-TNF therapy in such patients.

Cyclosporine and azathioprine — For children with continuing active disease despite appropriate treatment with NSAIDs and methotrexate, azathioprine and cyclosporine have been used with varying success in the past [25]. Cyclosporine is effective in children with dactylitis who have not responded to sulfasalazine. However, these drugs carry significant potential toxicity and appear to be less effective than the newer biologic agents. As a result, most clinicians favor the use of the biologic agents over these drugs.

COURSE AND PROGNOSIS — Children with the early onset of polyarticular JIA, regardless of the presence or absence of rheumatoid factor (RF), have a low likelihood of developing spontaneous remission. Despite the availability of additional medications to treat polyarticular JIA (eg, methotrexate and tumor necrosis factor [TNF] inhibitors), a significant number of patients continue to have some disease activity. The majority of children who "grow out of it" probably represent misclassified cases of reactive arthritis.

The following examples are illustrative:

In a prospective study of 137 children with polyarticular JIA, clinically inactive disease was an unstable state with 18 percent of children experiencing a flare while on TNF inhibition [48]. In that same study, 37 percent of children experienced a major flare within eight months of TNF inhibitor withdrawal. Levels of serum S100 proteins (immunohistochemical markers) were not predictive of maintenance of remission while on TNF inhibition or flare after withdrawal of TNF inhibition [107].

In a large Canadian cohort of children diagnosed with JIA from 2005 to 2010, at one, three, and five years from the time of diagnosis, 34, 78, and 97 percent of children with RF-negative disease achieved inactive disease, respectively [108]. These numbers are slightly lower in children with RF-positive disease (22, 67, and 93, respectively). Only 14 percent of RF-negative and none of the children with RF-positive disease attained remission off medication within five years of diagnosis.

In another observational cohort study from Taiwan, 48 and 13 percent of children with RF-negative disease and 33 and 11 percent of children with RF-positive disease achieved inactive disease and remission off medication over the course of 15 years [109]. Differences in these estimates likely result from slight differences in definitions of inactive disease and remission and follow-up times.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Uveitis" and "Society guideline links: Juvenile idiopathic arthritis".)

SUMMARY AND RECOMMENDATIONS

Goals of treatment – Treatment of polyarticular juvenile idiopathic arthritis (JIA) is directed toward treating the underlying synovitis and associated inflammation. Prompt initiation of therapy that provides relief of arthritic symptoms and preservation of function is critical to an improved outcome. (See 'Introduction' above.)

Considerations in choosing a treatment regimen – Treatment regimens are chosen according to prognostic factors, as well as clinician, parent/caregiver, and patient preferences, and are adjusted based upon clinical response. (See 'Management approach' above.)

Initial therapy – We recommend initial therapy with a disease-modifying antirheumatic drug (DMARD) in all patients with polyarticular JIA (Grade 1B). We use either methotrexate at a dose of 10 mg/m2 body surface area (BSA)/week or a biologic agent that inhibits tumor necrosis factor (TNF) in combination with methotrexate. Indications for using a TNF inhibitor in addition to or rather than methotrexate include presence of severe polyarthritis, poor prognostic features, or factors associated with poor response to methotrexate (eg, predominantly axial arthritis). Nonsteroidal antiinflammatory drugs (NSAIDs) are not appropriate as monotherapy but are suitable as adjunct therapy for symptom management. (See 'Initial management' above and 'Methotrexate' above and 'Tumor necrosis factor inhibitors' above and 'Nonsteroidal antiinflammatory drugs' above.)

Supplemental therapyFolic acid or leucovorin (folinic acid) supplementation is used in all children with JIA receiving methotrexate, given the beneficial effects noted and the lack of convincing contrary data. (See 'Folic acid' above.)

Refractory disease – Individualized therapeutic decisions are necessary in patients who fail to respond to methotrexate and a TNF inhibitor. Options include the use of combination DMARDs, another biologic agent, or a small molecule inhibitor under the supervision of providers with expertise in pediatric rheumatology. (See 'Refractory disease' above and 'Other biologic agents' above and 'Small molecule inhibitors' above and 'Additional agents' above.)

RF- and CCP-positive disease – Adolescents with rheumatoid factor (RF) positive and anticyclic citrullinated peptide (CCP) positive polyarthritis probably represent the early onset of rheumatoid arthritis (RA). As such, these children should be treated similar to adults with moderate-to-severe RA. (See 'Rheumatoid factor-positive patients' above and "Initial treatment of rheumatoid arthritis in adults".)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Thomas JA Lehman, MD, who contributed to earlier versions of this topic review.

  1. Criteria for the classification of juvenile rheumatoid arthritis. Bull Rheum Dis 1972; 23:712.
  2. Ringold S, Angeles-Han ST, Beukelman T, et al. 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Treatment of Juvenile Idiopathic Arthritis: Therapeutic Approaches for Non-Systemic Polyarthritis, Sacroiliitis, and Enthesitis. Arthritis Rheumatol 2019; 71:846.
  3. Ravelli A, Consolaro A, Horneff G, et al. Treating juvenile idiopathic arthritis to target: recommendations of an international task force. Ann Rheum Dis 2018; 77:819.
  4. Buckley L, Ware E, Kreher G, et al. Outcome Monitoring and Clinical Decision Support in Polyarticular Juvenile Idiopathic Arthritis. J Rheumatol 2020; 47:273.
  5. Ferrara G, Mastrangelo G, Barone P, et al. Methotrexate in juvenile idiopathic arthritis: advice and recommendations from the MARAJIA expert consensus meeting. Pediatr Rheumatol Online J 2018; 16:46.
  6. Wallace CA. The use of methotrexate in childhood rheumatic diseases. Arthritis Rheum 1998; 41:381.
  7. Ravelli A, Martini A. Methotrexate in juvenile idiopathic arthritis: answers and questions. J Rheumatol 2000; 27:1830.
  8. Ruperto N, Murray KJ, Gerloni V, et al. A randomized trial of parenteral methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Arthritis Rheum 2004; 50:2191.
  9. Silverman E, Mouy R, Spiegel L, et al. Leflunomide or methotrexate for juvenile rheumatoid arthritis. N Engl J Med 2005; 352:1655.
  10. Wallace CA, Giannini EH, Spalding SJ, et al. Trial of early aggressive therapy in polyarticular juvenile idiopathic arthritis. Arthritis Rheum 2012; 64:2012.
  11. Horneff G, De Bock F, Foeldvari I, et al. Safety and efficacy of combination of etanercept and methotrexate compared to treatment with etanercept only in patients with juvenile idiopathic arthritis (JIA): preliminary data from the German JIA Registry. Ann Rheum Dis 2009; 68:519.
  12. Wallace CA, Ringold S, Bohnsack J, et al. Extension study of participants from the trial of early aggressive therapy in juvenile idiopathic arthritis. J Rheumatol 2014; 41:2459.
  13. Hissink Muller PC, Brinkman DM, Schonenberg D, et al. A comparison of three treatment strategies in recent onset non-systemic Juvenile Idiopathic Arthritis: initial 3-months results of the BeSt for Kids-study. Pediatr Rheumatol Online J 2017; 15:11.
  14. Alexeeva E, Horneff G, Dvoryakovskaya T, et al. Early combination therapy with etanercept and methotrexate in JIA patients shortens the time to reach an inactive disease state and remission: results of a double-blind placebo-controlled trial. Pediatr Rheumatol Online J 2021; 19:5.
  15. Kimura Y, Schanberg LE, Tomlinson GA, et al. Optimizing the Start Time of Biologics in Polyarticular Juvenile Idiopathic Arthritis: A Comparative Effectiveness Study of Childhood Arthritis and Rheumatology Research Alliance Consensus Treatment Plans. Arthritis Rheumatol 2021; 73:1898.
  16. Beukelman T, Xie F, Baddley JW, et al. The risk of hospitalized infection following initiation of biologic agents versus methotrexate in the treatment of juvenile idiopathic arthritis. Arthritis Res Ther 2016; 18:210.
  17. Ungar WJ, Costa V, Hancock-Howard R, et al. Cost-effectiveness of biologics in polyarticular-course juvenile idiopathic arthritis patients unresponsive to disease-modifying antirheumatic drugs. Arthritis Care Res (Hoboken) 2011; 63:111.
  18. Luca NJ, Burnett HF, Ungar WJ, et al. Cost-Effectiveness Analysis of First-Line Treatment With Biologic Agents in Polyarticular Juvenile Idiopathic Arthritis. Arthritis Care Res (Hoboken) 2016; 68:1803.
  19. Vilca I, Munitis PG, Pistorio A, et al. Predictors of poor response to methotrexate in polyarticular-course juvenile idiopathic arthritis: analysis of the PRINTO methotrexate trial. Ann Rheum Dis 2010; 69:1479.
  20. Ringold S, Weiss PF, Colbert RA, et al. Childhood Arthritis and Rheumatology Research Alliance consensus treatment plans for new-onset polyarticular juvenile idiopathic arthritis. Arthritis Care Res (Hoboken) 2014; 66:1063.
  21. Giancane G, Alongi A, Rosina S, et al. Recent therapeutic advances in juvenile idiopathic arthritis. Best Pract Res Clin Rheumatol 2017; 31:476.
  22. Ruperto N, Nikishina I, Pachanov ED, et al. A randomized, double-blind clinical trial of two doses of meloxicam compared with naproxen in children with juvenile idiopathic arthritis: short- and long-term efficacy and safety results. Arthritis Rheum 2005; 52:563.
  23. Foeldvari I, Szer IS, Zemel LS, et al. A prospective study comparing celecoxib with naproxen in children with juvenile rheumatoid arthritis. J Rheumatol 2009; 36:174.
  24. Celebrex gets committee nod for juvenile arthritis, but safety registry urged. "The Pink Sheet" 2006; 68(49):7.
  25. Wallace CA. On beyond methotrexate treatment of severe juvenile rheumatoid arthritis. Clin Exp Rheumatol 1999; 17:499.
  26. Young CM, Shiels WE 2nd, Coley BD, et al. Ultrasound-guided corticosteroid injection therapy for juvenile idiopathic arthritis: 12-year care experience. Pediatr Radiol 2012; 42:1481.
  27. Ringold S, Seidel KD, Koepsell TD, Wallace CA. Inactive disease in polyarticular juvenile idiopathic arthritis: current patterns and associations. Rheumatology (Oxford) 2009; 48:972.
  28. Neidel J, Boehnke M, Küster RM. The efficacy and safety of intraarticular corticosteroid therapy for coxitis in juvenile rheumatoid arthritis. Arthritis Rheum 2002; 46:1620.
  29. Arabshahi B, Dewitt EM, Cahill AM, et al. Utility of corticosteroid injection for temporomandibular arthritis in children with juvenile idiopathic arthritis. Arthritis Rheum 2005; 52:3563.
  30. Ringold S, Torgerson TR, Egbert MA, Wallace CA. Intraarticular corticosteroid injections of the temporomandibular joint in juvenile idiopathic arthritis. J Rheumatol 2008; 35:1157.
  31. Morgan SL, Baggott JE. Folate supplementation during methotrexate therapy for rheumatoid arthritis. Clin Exp Rheumatol 2010; 28:S102.
  32. Ebbing M, Bønaa KH, Nygård O, et al. Cancer incidence and mortality after treatment with folic acid and vitamin B12. JAMA 2009; 302:2119.
  33. Ericson U, Sonestedt E, Gullberg B, et al. High folate intake is associated with lower breast cancer incidence in postmenopausal women in the Malmö Diet and Cancer cohort. Am J Clin Nutr 2007; 86:434.
  34. Ulrich CM. Folate and cancer prevention: a closer look at a complex picture. Am J Clin Nutr 2007; 86:271.
  35. Wallace CA, Ravelli A, Huang B, Giannini EH. Preliminary validation of clinical remission criteria using the OMERACT filter for select categories of juvenile idiopathic arthritis. J Rheumatol 2006; 33:789.
  36. Baszis K, Garbutt J, Toib D, et al. Clinical outcomes after withdrawal of anti-tumor necrosis factor α therapy in patients with juvenile idiopathic arthritis: a twelve-year experience. Arthritis Rheum 2011; 63:3163.
  37. Horton DB, Onel KB, Beukelman T, Ringold S. Attitudes and Approaches for Withdrawing Drugs for Children with Clinically Inactive Nonsystemic JIA: A Survey of the Childhood Arthritis and Rheumatology Research Alliance. J Rheumatol 2017; 44:352.
  38. Brunner HI, Ruperto N, Zuber Z, et al. Efficacy and safety of tocilizumab in patients with polyarticular-course juvenile idiopathic arthritis: results from a phase 3, randomised, double-blind withdrawal trial. Ann Rheum Dis 2015; 74:1110.
  39. Imagawa T, Yokota S, Mori M, et al. Safety and efficacy of tocilizumab, an anti-IL-6-receptor monoclonal antibody, in patients with polyarticular-course juvenile idiopathic arthritis. Mod Rheumatol 2012; 22:109.
  40. Ferucci ED, Majka DS, Parrish LA, et al. Antibodies against cyclic citrullinated peptide are associated with HLA-DR4 in simplex and multiplex polyarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 2005; 52:239.
  41. Dewint P, Hoffman IE, Rogge S, et al. Effect of age on prevalence of anticitrullinated protein/peptide antibodies in polyarticular juvenile idiopathic arthritis. Rheumatology (Oxford) 2006; 45:204.
  42. Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Allaart CF, et al. Clinical and radiographic outcomes of four different treatment strategies in patients with early rheumatoid arthritis (the BeSt study): a randomized, controlled trial. Arthritis Rheum 2005; 52:3381.
  43. Anderson JJ, Wells G, Verhoeven AC, Felson DT. Factors predicting response to treatment in rheumatoid arthritis: the importance of disease duration. Arthritis Rheum 2000; 43:22.
  44. Haraoui B, Pope J. Treatment of early rheumatoid arthritis: concepts in management. Semin Arthritis Rheum 2011; 40:371.
  45. Wessels JA, Huizinga TW, Guchelaar HJ. Recent insights in the pharmacological actions of methotrexate in the treatment of rheumatoid arthritis. Rheumatology (Oxford) 2008; 47:249.
  46. Chen J, Veras MM, Liu C, Lin J. Methotrexate for ankylosing spondylitis. Cochrane Database Syst Rev 2013; :CD004524.
  47. Foell D, Wulffraat N, Wedderburn LR, et al. Methotrexate withdrawal at 6 vs 12 months in juvenile idiopathic arthritis in remission: a randomized clinical trial. JAMA 2010; 303:1266.
  48. Lovell DJ, Johnson AL, Huang B, et al. Risk, Timing, and Predictors of Disease Flare After Discontinuation of Anti-Tumor Necrosis Factor Therapy in Children With Polyarticular Forms of Juvenile Idiopathic Arthritis With Clinically Inactive Disease. Arthritis Rheumatol 2018; 70:1508.
  49. Aquilani A, Marafon DP, Marasco E, et al. Predictors of Flare Following Etanercept Withdrawal in Patients with Rheumatoid Factor-negative Juvenile Idiopathic Arthritis Who Reached Remission while Taking Medication. J Rheumatol 2018; 45:956.
  50. Muzaffer MA, Schneider R, Cameron BJ, et al. Accelerated nodulosis during methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 1996; 128:698.
  51. Padeh S, Sharon N, Schiby G, et al. Hodgkin's lymphoma in systemic onset juvenile rheumatoid arthritis after treatment with low dose methotrexate. J Rheumatol 1997; 24:2035.
  52. Kahn P. Juvenile idiopathic arthritis--current and future therapies. Bull NYU Hosp Jt Dis 2009; 67:291.
  53. Salliot C, van der Heijde D. Long-term safety of methotrexate monotherapy in patients with rheumatoid arthritis: a systematic literature research. Ann Rheum Dis 2009; 68:1100.
  54. Becker ML, Rosé CD, Cron RQ, et al. Effectiveness and toxicity of methotrexate in juvenile idiopathic arthritis: comparison of 2 initial dosing regimens. J Rheumatol 2010; 37:870.
  55. Tuková J, Chládek J, Hroch M, et al. 677TT genotype is associated with elevated risk of methotrexate (MTX) toxicity in juvenile idiopathic arthritis: treatment outcome, erythrocyte concentrations of MTX and folates, and MTHFR polymorphisms. J Rheumatol 2010; 37:2180.
  56. Becker ML, Gaedigk R, van Haandel L, et al. The effect of genotype on methotrexate polyglutamate variability in juvenile idiopathic arthritis and association with drug response. Arthritis Rheum 2011; 63:276.
  57. Diak P, Siegel J, La Grenade L, et al. Tumor necrosis factor alpha blockers and malignancy in children: forty-eight cases reported to the Food and Drug Administration. Arthritis Rheum 2010; 62:2517.
  58. Lehman TJ. Should the Food and Drug Administration warning of malignancy in children receiving tumor necrosis factor alpha blockers change the way we treat children with juvenile idiopathic arthritis? Arthritis Rheum 2010; 62:2183.
  59. Beukelman T, Haynes K, Curtis JR, et al. Rates of malignancy associated with juvenile idiopathic arthritis and its treatment. Arthritis Rheum 2012; 64:1263.
  60. Greenberg JD, Reed G, Kremer JM, et al. Association of methotrexate and tumour necrosis factor antagonists with risk of infectious outcomes including opportunistic infections in the CORRONA registry. Ann Rheum Dis 2010; 69:380.
  61. http://www.fda.gov/bbs/topics/NEWS/2008/NEW01879.html (Accessed on November 23, 2008).
  62. Horneff G, Schmeling H, Biedermann T, et al. The German etanercept registry for treatment of juvenile idiopathic arthritis. Ann Rheum Dis 2004; 63:1638.
  63. Prince FH, Twilt M, ten Cate R, et al. Long-term follow-up on effectiveness and safety of etanercept in juvenile idiopathic arthritis: the Dutch national register. Ann Rheum Dis 2009; 68:635.
  64. Bracaglia C, Buonuomo PS, Tozzi AE, et al. Safety and efficacy of etanercept in a cohort of patients with juvenile idiopathic arthritis under 4 years of age. J Rheumatol 2012; 39:1287.
  65. Otten MH, Prince FH, Twilt M, et al. Delayed clinical response in patients with juvenile idiopathic arthritis treated with etanercept. J Rheumatol 2010; 37:665.
  66. Gartlehner G, Hansen RA, Jonas BL, et al. Biologics for the treatment of juvenile idiopathic arthritis: a systematic review and critical analysis of the evidence. Clin Rheumatol 2008; 27:67.
  67. Giannini EH, Ilowite NT, Lovell DJ, et al. Long-term safety and effectiveness of etanercept in children with selected categories of juvenile idiopathic arthritis. Arthritis Rheum 2009; 60:2794.
  68. Prince FH, de Bekker-Grob EW, Twilt M, et al. An analysis of the costs and treatment success of etanercept in juvenile idiopathic arthritis: results from the Dutch Arthritis and Biologicals in Children register. Rheumatology (Oxford) 2011; 50:1131.
  69. Vojvodich PF, Hansen JB, Andersson U, et al. Etanercept treatment improves longitudinal growth in prepubertal children with juvenile idiopathic arthritis. J Rheumatol 2007; 34:2481.
  70. van Dijken TD, Vastert SJ, Gerloni VM, et al. Development of inflammatory bowel disease in patients with juvenile idiopathic arthritis treated with etanercept. J Rheumatol 2011; 38:1441.
  71. Dallocchio A, Canioni D, Ruemmele F, et al. Occurrence of inflammatory bowel disease during treatment of juvenile idiopathic arthritis with etanercept: a French retrospective study. Rheumatology (Oxford) 2010; 49:1694.
  72. Lovell DJ, Giannini EH, Reiff A, et al. Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 2000; 342:763.
  73. Lovell DJ, Reiff A, Jones OY, et al. Long-term safety and efficacy of etanercept in children with polyarticular-course juvenile rheumatoid arthritis. Arthritis Rheum 2006; 54:1987.
  74. Lovell DJ, Reiff A, Ilowite NT, et al. Safety and efficacy of up to eight years of continuous etanercept therapy in patients with juvenile rheumatoid arthritis. Arthritis Rheum 2008; 58:1496.
  75. Giannini EH, Ruperto N, Ravelli A, et al. Preliminary definition of improvement in juvenile arthritis. Arthritis Rheum 1997; 40:1202.
  76. Simonini G, Giani T, Stagi S, et al. Bone status over 1 yr of etanercept treatment in juvenile idiopathic arthritis. Rheumatology (Oxford) 2005; 44:777.
  77. Tynjälä P, Lahdenne P, Vähäsalo P, et al. Impact of anti-TNF treatment on growth in severe juvenile idiopathic arthritis. Ann Rheum Dis 2006; 65:1044.
  78. Giannini EH, Ilowite NT, Lovell DJ, et al. Effects of long-term etanercept treatment on growth in children with selected categories of juvenile idiopathic arthritis. Arthritis Rheum 2010; 62:3259.
  79. Billiau AD, Loop M, Le PQ, et al. Etanercept improves linear growth and bone mass acquisition in MTX-resistant polyarticular-course juvenile idiopathic arthritis. Rheumatology (Oxford) 2010; 49:1550.
  80. Lovell DJ, Ruperto N, Goodman S, et al. Adalimumab with or without methotrexate in juvenile rheumatoid arthritis. N Engl J Med 2008; 359:810.
  81. http://www.rxabbvie.com/pdf/humira.pdf (Accessed on December 17, 2014).
  82. Lovell DJ, Brunner HI, Reiff AO, et al. Long-term outcomes in patients with polyarticular juvenile idiopathic arthritis receiving adalimumab with or without methotrexate. RMD Open 2020; 6.
  83. Ruperto N, Brunner HI, Pacheco-Tena C, et al. Open-label phase 3 study of intravenous golimumab in patients with polyarticular juvenile idiopathic arthritis. Rheumatology (Oxford) 2021; 60:4495.
  84. Brunner HI, Ruperto N, Tzaribachev N, et al. Subcutaneous golimumab for children with active polyarticular-course juvenile idiopathic arthritis: results of a multicentre, double-blind, randomised-withdrawal trial. Ann Rheum Dis 2018; 77:21.
  85. Ruperto N, Lovell DJ, Cuttica R, et al. A randomized, placebo-controlled trial of infliximab plus methotrexate for the treatment of polyarticular-course juvenile rheumatoid arthritis. Arthritis Rheum 2007; 56:3096.
  86. Weiss PF, Perelman School of Medicine, University of Pennsylvania, 2020, personal communication.
  87. Ruperto N, Lovell DJ, Cuttica R, et al. Long-term efficacy and safety of infliximab plus methotrexate for the treatment of polyarticular-course juvenile rheumatoid arthritis: findings from an open-label treatment extension. Ann Rheum Dis 2010; 69:718.
  88. Lahdenne P, Vähäsalo P, Honkanen V. Infliximab or etanercept in the treatment of children with refractory juvenile idiopathic arthritis: an open label study. Ann Rheum Dis 2003; 62:245.
  89. Tynjälä P, Vähäsalo P, Tarkiainen M, et al. Aggressive combination drug therapy in very early polyarticular juvenile idiopathic arthritis (ACUTE-JIA): a multicentre randomised open-label clinical trial. Ann Rheum Dis 2011; 70:1605.
  90. Brunner HI, Ruperto N, Zuber Z, et al. Efficacy and Safety of Tocilizumab for Polyarticular-Course Juvenile Idiopathic Arthritis in the Open-Label Two-Year Extension of a Phase III Trial. Arthritis Rheumatol 2021; 73:530.
  91. Ruperto N, Brunner HI, Ramanan AV, et al. Subcutaneous dosing regimens of tocilizumab in children with systemic or polyarticular juvenile idiopathic arthritis. Rheumatology (Oxford) 2021; 60:4568.
  92. Ruperto N, Lovell DJ, Quartier P, et al. Abatacept in children with juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled withdrawal trial. Lancet 2008; 372:383.
  93. Ruperto N, Lovell DJ, Li T, et al. Abatacept improves health-related quality of life, pain, sleep quality, and daily participation in subjects with juvenile idiopathic arthritis. Arthritis Care Res (Hoboken) 2010; 62:1542.
  94. Brunner HI, Tzaribachev N, Vega-Cornejo G, et al. Subcutaneous Abatacept in Patients With Polyarticular-Course Juvenile Idiopathic Arthritis: Results From a Phase III Open-Label Study. Arthritis Rheumatol 2018; 70:1144.
  95. Edwards JC, Szczepanski L, Szechinski J, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med 2004; 350:2572.
  96. Kazkaz H, Isenberg D. Anti B cell therapy (rituximab) in the treatment of autoimmune diseases. Curr Opin Pharmacol 2004; 4:398.
  97. Giannini EH, Lovell DJ, Silverman ED, et al. Intravenous immunoglobulin in the treatment of polyarticular juvenile rheumatoid arthritis: a phase I/II study. Pediatric Rheumatology Collaborative Study Group. J Rheumatol 1996; 23:919.
  98. Tofacitinib, tablet. United Sates prescribing information. Revised September 2023. US National Library of Medicine. www.dailymed.nlm.nih.gov/dailymed/index.cfm (Accessed on November 03, 2023).
  99. Ramanan AV, Quartier P, Okamoto N, et al. Baricitinib in juvenile idiopathic arthritis: an international, phase 3, randomised, double-blind, placebo-controlled, withdrawal, efficacy, and safety trial. Lancet 2023; 402:555.
  100. Brooks CD. Sulfasalazine for the management of juvenile rheumatoid arthritis. J Rheumatol 2001; 28:845.
  101. van Rossum MA, Fiselier TJ, Franssen MJ, et al. Sulfasalazine in the treatment of juvenile chronic arthritis: a randomized, double-blind, placebo-controlled, multicenter study. Dutch Juvenile Chronic Arthritis Study Group. Arthritis Rheum 1998; 41:808.
  102. van Rossum MA, Fiselier TJ, Franssen MJ, et al. Effects of sulfasalazine treatment on serum immunoglobulin levels in children with juvenile chronic arthritis. Scand J Rheumatol 2001; 30:25.
  103. van Rossum MA, van Soesbergen RM, Boers M, et al. Long-term outcome of juvenile idiopathic arthritis following a placebo-controlled trial: sustained benefits of early sulfasalazine treatment. Ann Rheum Dis 2007; 66:1518.
  104. Silverman E, Spiegel L, Hawkins D, et al. Long-term open-label preliminary study of the safety and efficacy of leflunomide in patients with polyarticular-course juvenile rheumatoid arthritis. Arthritis Rheum 2005; 52:554.
  105. Foeldvari I, Wierk A. Effectiveness of leflunomide in patients with juvenile idiopathic arthritis in clinical practice. J Rheumatol 2010; 37:1763.
  106. Chickermane PR, Khubchandani RP. Evaluation of the benefits of sequential addition of leflunomide in patients with polyarticular course juvenile idiopathic arthritis failing standard dose methotrexate. Clin Exp Rheumatol 2015; 33:287.
  107. Hinze CH, Foell D, Johnson AL, et al. Serum S100A8/A9 and S100A12 Levels in Children With Polyarticular Forms of Juvenile Idiopathic Arthritis: Relationship to Maintenance of Clinically Inactive Disease During Anti-Tumor Necrosis Factor Therapy and Occurrence of Disease Flare After Discontinuation of Therapy. Arthritis Rheumatol 2019; 71:451.
  108. Guzman J, Oen K, Tucker LB, et al. The outcomes of juvenile idiopathic arthritis in children managed with contemporary treatments: results from the ReACCh-Out cohort. Ann Rheum Dis 2015; 74:1854.
  109. Shen CC, Yeh KW, Ou LS, et al. Clinical features of children with juvenile idiopathic arthritis using the ILAR classification criteria: a community-based cohort study in Taiwan. J Microbiol Immunol Infect 2013; 46:288.
Topic 6428 Version 29.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟