Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jun 1.
Published in final edited form as: Ann Rheum Dis. 2019 Sep 27;78(12):1722–1731. doi: 10.1136/annrheumdis-2019-216040

Emergent high fatality lung disease in systemic juvenile arthritis

Vivian E Saper 1, Guangbo Chen 2, Gail H Deutsch 3,4,#, R Paul Guillerman 5,#, Johannes Birgmeier 6,#, Karthik Jagadeesh 6,#, Scott Canna 7,#, Grant Schulert 8,9,#, Robin Deterding 10,11, Jianpeng Xu 1, Ann N Leung 12, Layla Bouzoubaa 13, Khalid Abulaban 14,15, Kevin Baszis 16, Edward M Behrens 17,18, James Birmingham 19,20, Alicia Casey 21,22, Michal Cidon 23,24, Randy Q Cron 25,26, Aliva De 27, Fabrizio De Benedetti 28, Ian Ferguson 29, Martha P Fishman 21,22, Steven I Goodman 30, T Brent Graham 31, Alexei A Grom 8,9, Kathleen Haines 32,33, Melissa Hazen 21,22, Lauren A Henderson 21,22, Assunta Ho 34,35, Maria Ibarra 36,37, Christi J Inman 38, Rita Jerath 39,40, Khulood Khawaja 41, Daniel J Kingsbury 42, Marisa Klein-Gitelman 43, Khanh lai 38, Sivia Lapidus 32,33, Clara Lin 10,11, Jenny Lin 44,45, Deborah R Liptzin 10,11, Diana Milojevic 46, Joy Mombourquette 47, Karen Onel 48,49, Seza Ozen 50, Maria Perez 51, Kathryn Phillippi 52,53, Sampath Prahalad 54,55, Suhas Radhakrishna 56,57, Adam Reinhardt 58, Mona Riskalla 59,60, Natalie Rosenwasser 48,49, Johannes Roth 61, Rayfel Schneider 62,63, Dieneke Schonenberg-Meinema 64,65, Susan Shenoi 4,66, Judith A Smith 67, Hafize Emine Sönmez 50, Matthew L Stoll 25,26, Christopher Towe 8,9, Sara O Vargas 22,68, Richard K Vehe 59,60, Lisa R Young 17,18, Jacqueline Yang 2, Tushar Desai 69, Raymond Balise 13, Ying Lu 70, Lu Tian 70, Gill Bejerano 6, Mark M Davis 71, Purvesh Khatri 2, Elizabeth D Mellins 1, Childhood Arthritis and Rheumatology Research Alliance Registry Investigators
PMCID: PMC7065839  NIHMSID: NIHMS1552540  PMID: 31562126

Abstract

Objective

To investigate the characteristics and risk factors of a novel parenchymal lung disease (LD), increasingly detected in systemic juvenile idiopathic arthritis (sJIA).

Methods

In a multicentre retrospective study, 61 cases were investigated using physician-reported clinical information and centralised analyses of radiological, pathological and genetic data.

Results

LD was associated with distinctive features, including acute erythematous clubbing and a high frequency of anaphylactic reactions to the interleukin (IL)-6 inhibitor, tocilizumab. Serum ferritin elevation and/or significant lymphopaenia preceded LD detection. The most prevalent chest CT pattern was septal thickening, involving the periphery of multiple lobes ± ground-glass opacities. The predominant pathology (23 of 36) was pulmonary alveolar proteinosis and/or endogenous lipoid pneumonia (PAP/ELP), with atypical features including regional involvement and concomitant vascular changes. Apparent severe delayed drug hypersensitivity occurred in some cases. The 5-year survival was 42%. Whole exome sequencing (20 of 61) did not identify a novel monogenic defect or likely causal PAP-related or macrophage activation syndrome (MAS)-related mutations. Trisomy 21 and young sJIA onset increased LD risk. Exposure to IL-1 and IL-6 inhibitors (46 of 61) was associated with multiple LD features. By several indicators, severity of sJIA was comparable in drug-exposed subjects and published sJIA cohorts. MAS at sJIA onset was increased in the drug-exposed, but was not associated with LD features.

Conclusions

A rare, life-threatening lung disease in sJIA is defined by a constellation of unusual clinical characteristics. The pathology, a PAP/ELP variant, suggests macrophage dysfunction. Inhibitor exposure may promote LD, independent of sJIA severity, in a small subset of treated patients. Treatment/prevention strategies are needed.

INTRODUCTION

Systemic juvenile idiopathic arthritis (sJIA) is a chronic, inflammatory disease of childhood, observed worldwide, with an incidence of 0.4–0.9/100 000 in North America and Europe.1 A similar disease occurs in adults (adult-onset Still’s disease (AOSD); incidence: 0.2–0.4/100 000).2 sJIA is characterised by a combination of arthritis, which can be destructive, and systemic inflammation, including daily fever spikes, evanescent macular rash and serositis. A life-threatening complication (mortality rate: 8%–17% in sJIA)3 is overt macrophage activation syndrome (MAS). MAS is a form of secondary haemophagocytic lymphohistiocytosis (HLH) that manifests as a cytokine storm with very high serum ferritin and, in severe cases, organ failure. Therapies that antagonise cytokines interleukin (IL)-1 and IL-6 were introduced into management of sJIA ~15 years ago. This approach is rapidly effective in >65% of patients, implicating these inflammatory mediators as key drivers of sJIA.46

The usual pulmonary complications of sJIA are pleuritis and pleural effusion.1,7 Scattered case reports of other lung diseases (LDs) in sJIA have appeared.812 However, in the last decade, paediatric rheumatologists have increasingly detected cases with types of LDs rarely seen in sJIA previously. Kimura et al13 described 25 cases that occurred before February 2011; diagnoses included pulmonary arterial hypertension (64%), interstitial LD (28%) and pulmonary alveolar proteinosis (PAP) (20%). sJIA course was considered severe, with MAS in 80%.

Here, we performed a multicentre, retrospective study of 61 cases with centralised analyses of radiographic, pathological and genetic data to provide current characterisation of LD in sJIA or sJIA-like disease, search for early indicators and investigate risk factors.

METHODS

Case definition and comparators

We used an operational sJIA case definition, developed by expert consensus as a modification of the International League of Associations for Rheumatology sJIA classification criteria.14 Cases failing to meet the case definition but managed clinically like sJIA were classified as sJIA-like. We identified 61 patients with sJIA or sJIA-like disease and parenchymal LD through the Childhood Arthritis and Rheumatology Research Alliance (CARRA) network and the international paediatric rheumatology listserv (administered by McMaster University, Ontario). Inclusion required verification of sJIA or sJIA-like illness, data at LD diagnosis, and parenchymal LD by chest CT and/or lung tissue, available for expert review. Comparator data were from physician-classified patients with sJIA without known LD enrolled in the CARRA registry (CR)15 between 2015 and 2018, or in the PharmaChild pharmacovigilance registry.16 See online supplementary figures S1AB and S2 and online supplementary information for additional details.

Data collection

Data included demographics, medical history, and clinical features at sJIA onset, at LD diagnosis and at defined time points (visits) before and after LD diagnosis. RegiSCAR score for drug- related eosinophilic systemic syndrome (DReSS) was calculated from clinical data.17 Online supplementary information includes details and definitions (online supplementary table S1).

Data analysis

Centralised analyses were performed for chest CT scans from 58 of 61 subjects, histopathology from 36 of 61 subjects and whole exome sequence (WES) data from 20 subjects. R V.3.5.1 program was used for all statistical analyses. Additional methods are in online supplementary information.

RESULTS

Disease characterisation: features unusual for sJIA

The cohort included 45 cases of sJIA and 16 cases of sJIA-like disease; demographics (table 1) and findings prior to or associated with LD did not differ systematically between these two groups (online supplementary tables S2S4). Demographics of the LD cohort are mostly similar to subjects with sJIA in the CR, with the notable exceptions of significantly lower median age at sJIA onset and significantly higher prevalence of trisomy 21 (T21) (table 1). These findings are discussed further in the Candidate risk factors for parenchymal LD section.

Table 1.

Demographic characteristics

sJIA-lung disease cohort vs CR control
sJIA-lung disease cohort by subgroup
sJIA-lung disease sJIA (CR) P value§ sJIA-like sJIA-ILAR+ OR (95% CI)
Sex (female, %) 66% (40/61) 59% (278/471) 0.41 81% (13/16) 60% (27/45) 0.35 (0.056 to 1.5)
Age, median years (IQR) 2.8 (1.2–6.3) 5.2 (2.8–9.8) 1.7×10−5*** 2.3 (0.9–7.8) 3.1 (1.5–6.2) 0.43 (0.13 to 1.45)
Race 0.53
 White 62% (38/61) 66% (308/466) 50% (8/16) 67% (30/45) 2 (0.53 to 7.4)
 Black 8.2% (5/61) 11% (50/466) 6.2% (1/16) 8.9% (4/45) 1.5 (0.13 to 77)
 Other†† 30% (18/61) 23% (108/466) 44% (7/16) 24% (11/45) 0.42 (0.11 to 1.7)
Region (only USA)‡‡ 0.02*
 Northeast 19% (10/53) 28% (123/442) 27% (4/15) 16% (6/38) 0.5 (0.12 to 2.17)
 Midwest 25% (13/53) 29% (129/442) 20% (3/15) 26% (10/38) 1.43 (0.33 to 6.1)
 South 17% (9/53) 23% (101/442) 20% (3/15) 16% (6/38) 1.25 (0.29 to 5.3)
 West 40% (21/53) 20% (89/442) 33% (5/15) 42% (16/38) 1.45 (0.42 to 5.1)
Genetics
 Trisomy 21 9.8% (6/61) 0.2% (1/471) 0.04* 12% (2/16) 8.9% (4/45) 0.69 (0.087 to 8.4)
 Familial HLH 7.1% (2/28)§§ 22% (2/9) 0% (0/19) 0 (0 to 2.4)
*

P<0.05

**

p<0.01

***

p<0.001.

For OR, 95% CI is shown.

CR, CARRA registry. Diagnosed as sJIA per physician report without specific verification of ILAR features.

sJIA-ILAR+ or sJIA-like classification verified (see the Methods section). sJIA-like had no arthritis (13) or failed modified fever criteria (3).

§

For categorical items, Fisher’s exact tests (including the multicategory form) were performed. For age, a Wilcoxon rank-sum test was performed.

OR showed no significant difference with the exception of trisomy 21 vs CR (OR=50).

††

Other: Hispanic (5), Middle Eastern (1), Asian (2), multiethnic (8) and other (2).

‡‡

Based on US Census Bureau population-balanced regions: www2.census.gov/geo/pdfs/maps-data/maps/reference/us_regdiv.pdf.

§§

Diagnosed by clinical testing; one with 2 UNC13D mutations and one with UNC13D/PRF1 mutations; both sJIA-like.

HLH, haemophagocytic lymphohistiocytosis; ILAR, International League of Associations for Rheumatology; sJIA, systemic juvenile idiopathic arthritis.

Clinical features prior to LD and associated with LD are summarised in online supplementary tables S2 and S3, respectively. At LD diagnosis, respiratory signs and symptoms were typically absent or subtle, although hypoxia was reported in 43% and clinical pulmonary hypertension (PH) in 30%. Strikingly, 61% of patients developed acute clubbing, sometimes as the first indicator of LD. In over half of these, digital erythema occurred (figure 1AC). Other atypical features were pruritic, non-evanescent rashes in 56% (figure 1DF), eosinophilia in 37%, and unexplained, severe abdominal pain in 16% (likely underestimated as this was not directly queried). Anaphylaxis to tocilizumab was unusually common, occurring in 38% of those exposed (14 of 37; see online supplementary table S3, footnote 6), compared with 0.6% (1 of 159) in the CR and 0.9% (1 of 110) in the tocilizumab trial in sJIA.5 Overall, the LD cohort manifested clinical features that are unusual for sJIA18 or pulmonary disease.

Figure 1.

Figure 1

Distinctive clinical features in systemic juvenile idiopathic arthritis (sJIA) with lung disease (LD) and survival outcome. (A) Acute erythematous digital clubbing; (B, C) bulbous deformity with erythematous clubbing of fingers (B) and toes (C); (D) typical salmon-coloured, macular sJIA rash (evanescent); and (E, F) atypical rashes that occur before LD detection: (E) oedematous, urticarial, non-evanescent rash (knee) and (F) serpiginous, eczematous, non-evanescent rash with hyperpigmented borders. (G) Mean±SE blood ferritin values of propensity-matched (online supplementary figure S3) sJIA controls (blue) and LD cases (red) across time points relative to LD diagnosis. n.s., p>0.1, *p<0.05, **p<0.01, ***p<0.001, by Wilcoxon rank-sum test.

Candidate early indicators of LD in sJIA: ferritin and lymphopaenia

The median time to LD diagnosis after sJIA onset was 1.6 years (IQR 0.8–3.3 years), excluding 6 of 61 cases with LD at systemic disease onset. To identify candidate early signs of LD, we analysed laboratory values commonly followed in sJIA. We matched cases to sJIA controls from the CR (1:1) for factors including laboratory test timing relative to sJIA onset, overall drug exposure, sex and sJIA onset age (online supplementary figure S3). We then assessed serum ferritin as an indicator of inflammation. The mean serum ferritin level in cases 1 year prior to LD diagnosis was not distinguishable from that of propensity-matched patients with sJIA in the CR. However, the level rose substantially within the 12 months before LD diagnosis in the cohort (figure 1G, online supplementary figure S4).

Another finding that preceded LD detection was significant lymphopaenia (absolute lymphocyte count <60% of age-adjusted, lower limit of normal).19 This degree of lymphopaenia, without concurrent MAS, was documented between the 6-month and 1-month visit prior to LD diagnosis in 42% of cases (excluding those with LD at sJIA onset; online supplementary table S3). We were unable to compare this with CR controls due to lack of information. However, this degree of lymphopaenia is not a known feature of active sJIA.18 Increased ferritin and lymphopaenia before LD diagnosis suggest a possibly extended incubation phase associated with smouldering inflammation and/or delayed recognition of LD.

Radiological features

As a step towards determining the nature of the LD, chest CT scans from 58 of 61 patients, most obtained at diagnosis, were systematically reviewed (RPG). Most exhibited one or more of five patterns (figure 2AE). Pattern A (septal thickening involving the periphery of multiple lobes, most marked in the lower lung zones, parahilar/paramediastinal and/or anterior upper lobes with or without adjacent ground-glass opacities) was the most frequently observed (60%). Crazy-paving (figure 2B), peripheral consolidation (figure 2C), peribronchovascular consolidation (figure 2D) and predominantly ground-glass opacities (figure 3E) were seen in 21%, 22%, 16% and 12%, respectively. Among those with contrast-enhanced CTs, 11 of 30 (37%) displayed hyperenhancing lymph nodes (figure 2F), a peculiar finding, previously reported in unusual conditions.20,21 Findings like pattern A have been observed with connective tissue disease-associated interstitial LD or interstitial pneumonia with autoimmune features.22 However, unlike these disorders, radiological signs of fibrosis (honeycombing, traction bronchiectasis) were uncommon in our cohort. Overall, the observed CT findings are unexpected in sJIA; the more typical finding of pleural effusion1,7,18 was rare at LD diagnosis (online supplementary table S4).

Figure 2.

Figure 2

Distinctive radiological and pathological features. Panels A–E: representative axial chest CT images: (A) multilobar, predominantly peripheral septal thickening, most marked in the lower lung, parahilar and/or anterior upper lobes with or without adjacent ground-glass opacities; (B) crazy-paving; (C) peripheral consolidations; (D) peribronchovascular consolidations; (e) predominantly ground-glass opacities; and (F) hyperenhancing lymph nodes on contrast-enhanced CT. Panels G–H: histopathological findings (H&E staining) along the pulmonary alveolar proteinosis/endogenous lipoid pneumonia (PAP/ELP) spectrum. Alveolar filling with eosinophilic proteinaceous material (G, left), admixed with a variable degree of ELP, indicated by cholesterol clefts (arrowheads) and foamy (lipid-containing) macrophages (G, middle and right), as described.82,83 Regions of PAP/ELP accompanied by type II alveolar epithelial cell hyperplasia (G, right insert, arrow), mild to moderate interstitial infiltration by inflammatory cells and lobular remodelling (airspace widening with increased interstitial smooth muscle). Typically, PAP/ELP findings were patchy, with involved areas juxtaposed to the normal lung (G, left, arrow). Pulmonary arterial wall thickening; a, artery (G, right). In A–G, the number of cases with pattern/number of assessable cases are indicated. (H) Electron micrograph showing normal lamellar bodies within type II cells (arrows) and macrophage (centre), containing lamellar debris, lipid (*) and cholesterol clefts (arrowhead). Original magnification ×7000. Four PAP/ELP cases (one each: ABCA3 and CSF2RB variants), stained for surfactant proteins (SP-B, proSP-C, SP-D, ABCA3, TTF-1), demonstrated robust immunoreactivity (not shown).

Figure 3.

Figure 3

Annual number of reported cases of LD and PAP/ELP pathology. (A) Annual number of LD cases in this series (total n=61). (B) Percentage of biopsied LD cases (n=36) with PAP/ELP pathology, grouped by year of LD diagnosis. (C) Annual incidence of LD, indicating proportions exposed (black) or not (grey) to anti-IL-1/IL-6 inhibitors. ABCA-3, ATP binding cassette subfamily A member 3; IL, interleukin; LD, lung disease; PAP/ELP, pulmonary alveolar proteinosis/endogenous lipoid pneumonia; proSP-C, prosurfactant D; SP-B, surfactant protein B; SP-D, surfactant protein D; TTF-1, thyroid transcription factor 1.

Histopathology and related genetics

Biopsy or autopsy tissues of 36 of 61 patients were available for centralised analysis (GD). Multicompartment disease (some combination of alveolar, airway, pleural, vascular alterations) was observed in all cases. Using their primary pattern of injury, three subgroups were defined: spectrum of PAP/endogenous lipoid pneumonia (ELP), vasculopathy and other (online supplementary table S4). The pathology typically associated with pattern A on CT is non-specific interstitial pneumonia (NSIP).22,23 Surprisingly, in 21 patients with CT pattern A who had histology, only 1 had NSIP Instead, the predominant pathology (64% of cases reviewed) was PAP/ELP (figure 2G), which was patchy and often accompanied by associated vascular changes (figure 2G, right). PAP/ELP is very rare in rheumatic disease,24 and the CT pattern typically associated with PAP is crazy-paving (pattern B).25

To identify other histological features associated with PAP/ELP-like pathology in our cases, we generated a heat map (online supplementary figure S5A). Not surprisingly, type II alveolar cell hyperplasia was highly associated with PAP/ELP.26,27 The next associated finding was lymphoplasmacytic inflammation (71% of PAP/ELP cases). Third, 55% had mild to moderate pulmonary arterial wall thickening. Hypertensive vascular changes are not typically associated with inherited PAP/ELP, suggesting a secondary disease process.2628 Electron microscopy (EM) (available in nine PAP/ELP cases) demonstrated well-formed lamellar bodies within type II alveolar epithelial cells. Variable accumulation of macrophages containing lamellar debris, lipid and cholesterol clefts was observed (figure 2H). The EM findings are more characteristic of macrophage overloading or dysfunction than of genetic disorders in surfactant metabolism.29,30 When we examined 13 PAP/ELP cases (6 with EM analysis) for genes causing hereditary PAP (SFTPA1, SFTPB, SFTPC, ABCA3, NKX2–1, CSF2RA, CSFR2B, MARS),26,28,3135 6 were heterozygous for protein-changing mutations, but none was de novo in trio analyses (online supplementary table S5A). One (SFPTC p.R167Q) causes PAP with low penetrance31; the others are not known to cause PAP in heterozygotes. While these rare variants (maximum allele frequency <5%) might contribute to LD in these children, they are not likely the full explanation.32,36,37

Vascular abnormalities were the predominant finding in 4 of 36 biopsies (online supplementary figure S5BD, table S4). Consistent with chest CT findings, interstitial fibrosis was generally mild, with advanced fibrosis/remodelling in only 4 of 36 samples, including 2 autopsies (online supplementary figure S5D).

Candidate risk factors for parenchymal LD

Other genetic factors

No evidence for a shared monogenic explanation for LD was found in WES of the 20 cases analysed (not shown). We also assessed the frequency of HLH/MAS-related gene variants (PRF1, LYST, STX11, STXBP2, UNC13D, NRLC4). Rare protein-altering variants, all heterozygous and none de novo, were found (online supplementary table S5B). Concordance between these variants and MAS (at sJIA onset or ever during disease course to data close) was not observed. The frequency of such variants (55%) is higher than reported for sJIA with MAS (36%)38 and could contribute to propensity for inflammation.

Early-onset sJIA

Compared with control subjects with sJIA in the CR, the LD cohort’s median age at sJIA onset was substantially younger (table 1; 2.3 years (1.1–5.0) vs 5.2 years (2.8–9.8), p = 1×10−7; online supplementary figure S6A). The CR cohort showed the full age of onset range of published sJIA cohorts, whereas the LD cohort was similar to a subgroup with younger onset age in sJIA cohorts39,40 (online supplementary figure S6B). Within LD cases, early onset of sJIA/sJIA-like disease was tightly correlated with PAP/ELP-like pathology (Wilcoxon test, p = 2.3e-4; if cut-off for age at sJIA onset <5 years, OR=15, Fisher’s p = 0.001, compared with older children in the cohort). Of cases, 91% (21 of 23) with PAP/ELP had sJIA onset at <5 years (online supplementary figure S6C).

Trisomy 21

In the LD cohort, T21 prevalence (table 1) was strikingly higher (10%) than in sJIA registry cohorts (0.2%; 1 of 471 in the CR and 2 of 914 in PharmaChild, these being similar to the frequency (0.14%) in live births).41 There were suggestions of more aggressive LD in these children, all of whom developed LD on anti-IL-1/IL-6. Four of six children with T21 were hypoxic (OR=7.8, Fisher’s p = 0.08, compared with the proportion of non-T21 with hypoxia). Two of five with T21 showed advanced interstitial fibrosis/remodelling (OR=8.4, Fisher’s p = 0.09), and two of four children with advanced fibrosis had T21. Five of six (83%) had viral or fungal lung infection at LD diagnosis, compared with 16 of 55 (29%) of non-T21 (OR=12, Fisher’s p=0.02)

Pre-exposure to cytokine inhibitors

Compared with previously described patients with sJIA,18 the LD cohort demonstrated rare clinical, radiological and pathological findings. During the time period of the series, the annual number of cases in the LD cohort increased dramatically, although some bias of ascertainment is possible due to increased awareness of this disease (figure 3A). PAP/ELP pathology increased among biopsied cases (figure 3B). The proportion of cases exposed to IL-1/IL-6 inhibitors increased in the cohort (figure 3C) and in all reported LD cases in sJIA (online supplementary figure S7A). These three trends coincided with increased use of IL-1/IL-6 inhibitors for sJIA.42 This prompted us to ask whether, compared with the CR sJIA cohort, the LD cohort was enriched for cases with exposure to IL-1/IL-6 inhibitors prior to LD. We matched the LD cases and CR controls for a number of potential confounders (online supplementary figure S7B). We found no difference in overall exposure to IL-1/IL-6 inhibitors; the exposure level in the CR controls was already high (online supplementary figure 7C; 46 of 53, 87%). Interestingly, among specific drug exposures, we found there was a moderate increase in anakinra exposure (OR=2.2 (0.94–5.5), p = 0.07). We next asked whether exposure to these inhibitors was related to the prevalence of unusual features. The frequencies of acute clubbing, digital erythema, unexplained abdominal pain, peripheral eosinophilia, CT pattern A or D, hyperenhancing lymph nodes and PAP/ELP pathology, but not PH, were substantially higher (p<0.1; false discovery rate <20%) in pre-exposed versus non-pre-exposed subjects (figure 4AB). These associations were not specific to one inhibitor. The median time from IL-1/IL-6 inhibitor to LD diagnosis was 1.2 years (IQR 0.7–2.0 years, n=46).

Figure 4.

Figure 4

Association between unusual features and pre-exposure to anti-IL-1/IL-6 or MAS at sJIA onset. (A) Heat map indicating occurrence of unusual clinical and radiological features (rows) by subjects (columns), grouped by pre-exposure status. (B) Statistical analysis for panel A, indicating p values, FDR and OR with 95% CI. Inf, infinite, #p<0.1, *p<0.05, **p<0.01, ***p<0.001. (C–F) Comparison of severity-related features in pre-exposed LD cases versus published sJIA cohorts. (C) Pre-exposed LD cases compared with Janow et al42 (D) Pre-exposed LD cases with sJIA onset <1.5 years, compared with comparable age group in Russo and Katsicas43; cut-off at <1.5 years was chosen by Russo and Katsicas, based on developmental difference before versus after 18 months. (E) Pre-exposed LD cases with sJIA onset >1.5 years, compared with comparable age group in Russo and Katsicas.43 (F) Pre-exposed LD cases treated with IL-1/IL-6 inhibitors for ≥6 months compared with comparable groups in Pardeo et al84 and Nigrovic et al.80 No bar indicates unavailable data. For details on definitions and published cohorts, see online supplementary table S6. (G) Statistical analysis of associations between MAS at sJIA onset and unusual clinical features of LD in sJIA, indicating p values, FDR and OR with 95% CI. FDR, false discovery rate; HELN, hyperenhancing lymph nodes; IL, interleukin; LD, lung disease; MAS, macrophage activation syndrome; PAP/ELP, pulmonary alveolar proteinosis/endogenous lipoid pneumonia; sJIA, systemic juvenile idiopathic arthritis.

Severe or refractory sJIA

A possible bias in the associations with pre-exposure is increased severity of sJIA and related treatment, that is, channel bias. We assessed five clinical features associated with sJIA severity, as there is no validated sJIA severity index. For increased severity, we assessed MAS at sJIA onset, need for calcineurin inhibitors and persistent arthritis; for reduced severity, we assessed ‘ever off’ steroids and positive treatment response. The small sample of non-exposed subjects prevented us from performing a full analysis within our cohort. We compared the pre-exposed LD subgroup with published cohorts (figure 4CF; online supplementary table S6). In Russo and Katsicas,43 the proportion of children with early-onset sJIA (<1.5 years) with MAS at sJIA onset was 10× higher than the proportion of children with later sJIA onset (>1.5 years) (32% vs 3%). This difference was interpreted to indicate more severe inflammation in early-onset sJIA. Among pre-exposed subjects with early-onset sJIA LD, the proportion with MAS at onset (27%) was comparable with early-onset subjects in Russo and Katsicas.43 For pre-exposed children with later-onset sJIA LD, a significantly higher proportion had MAS at onset versus the comparable group in Russo and Katsicas43 (26% vs 3%; OR=12.75, p = 0.0004) and versus another sJIA cohort, Behrens et al44 (OR=4.83, p = 0.003). These observations raised the possibility that LD was associated with MAS at onset, resulting in increased inhibitor use. However, there was no association in the LD cohort between MAS at onset and any of the unusual features of LD (figure 4G).

The pre-exposed LD cohort was generally similar to published cohorts for frequency of treatment-responsive disease (lack of calcineurin inhibitor, a period of systemic quiescence or substantially reduced steroid treatment; Figure 4CF). One exception was that a lower proportion of the LD subgroup treated with inhibitors for ≥6 months were ever off steroids, perhaps reflecting more severe disease; nonetheless, this proportion was >50% (figure 4F). In addition, at data close, 58% of the pre-exposed (18 of 31) reported inactive sJIA (on medication) despite continuing LD in 94% (17 of 18). These observations argue that refractory sJIA is not required for LD development or persistence.

Survival

The period of follow-up after LD diagnosis was variable (median 1.7 years; IQR 0.75–3 years). Survival was drastically lower in the LD cohort (mortality: 159/1000 person-years; figure 5) than in a UK cohort of patients with sJIA who required biologic agents (mortality: 3.9/1000 person-years).45 The predominant cause of death in our cohort was reported as diffuse LD (12 of 22 deaths), with MAS in 5 of 12 (online supplementary table S7). Among 75 categorical variables (online supplementary table S2S5), male sex, hypoxia at initial LD evaluation, predominantly neutrophilic bronchoalveolar lavage (BAL) (≥40% neutrophils, over 10 times the normal),46,47 but not PH, appeared to associate with shortened survival (online supplementary figure S8AD). These associations were not significant after adjusting for multiple tests. However, BAL neutrophilia (≥50%) has been linked to fatality,48,49 and 100% in our series (12 of 12) with this feature were deceased by data close.

Figure 5.

Figure 5

Survival outcome in systemic juvenile idiopathic arthritis cohort with lung disease (LD). The number of survivors at a given time point after LD diagnosis is shown (strata).

DISCUSSION

LD in sJIA was characterised by young age at sJIA onset and unusual clinical features, including acute erythematous clubbing, atypical rash and anaphylaxis to tocilizumab; severe tocilizumab reaction in sJIA with pulmonary disease was also noted in the data from the PharmaChild registry.50 The most prevalent finding on chest CT was peripheral septal thickening ± ground-glass opacities. Crazy-paving, consolidation and hyperenhancing lymph nodes were also observed. On tissue diagnosis, this group showed primarily PAP/ELP-like pathology. Compared with PAP/ELP in other settings, the LD pathology was distinctive for its patchiness and associated vascular changes.

The proportion of LD cases with PAP/ELP-like tissue diagnosis has increased since 2010, coinciding with increasing use of IL-1/IL-6 inhibitors.40 Pre-exposure to these inhibitors was characteristic of the predominant phenotypic subtype in our series. It is possible that this association is confounded by concomitant reduction in steroids with inhibitor use or by treatment of severe inflammation; our data do not conclusively rule out these possibilities. However, severe disease has been observed since the initial description of sJIA in 1897,51 whereas the LD with associated features described here appears to be new and increasing in frequency. Among biopsied cases, PAP/ELP was found in 80% with pre-exposure (includes one with mostly pleural sample and limited PAP foci) vs 36% not pre-exposed (OR=7 (95% CI 1.45 to 33.7), Fisher’s p = 0.015; online supplementary figure S5E) and was independent of PAP/ELP association with young age (online supplementary figure S9). Thus, IL-1/IL-6 inhibitor exposure may promote development of PAP/ELP-like disease and may qualitatively influence LD-associated features in a subset of patients with sJIA, among the substantially larger group of patients who derive striking benefit from these inhibitors.

Autopsy RNA sequencing data from the Genotype-Tissue Expression project show the lung is a major physiological producer of IL-1 and IL-6 in adults (online supplementary figure S10),52 and cytokine profiling suggests that circulating IL-1RA levels (reflecting the IL-1 activity) in young (<4 years) healthy children are 2× higher than in older healthy children.53 In addition, NFκB, a key transcription factor downstream of IL-1, stimulates angiogenesis and alveolarisation in the postnatal, developing lung.54 These observations raise the possibility of a physiological role of IL-1/IL-6 in the lung, particularly in early childhood. The striking enhancement of LD risk by early age of sJIA onset suggests developmental vulnerabilities that may interact with the inhibitors.

A specific relationship between reduced IL-1 and PAP development is described in mice. IL-1α−/− mice (but not IL-1β−/− mice) challenged with inhaled silicone, an inflammasome (NLRP3) activator,55 develop PAP- like LD.56 In the lung, IL-1 regulates granulocyte-macrophage colony-stimulating factor (GM-CSF) levels57,58 and macrophage function56,57; disruption of either can lead to surfactant accumulation and PAP59,60 These findings imply a link between reduced IL-1 and PAP, but also suggest that additional triggers may be required for disease development, in line with the rarity of severe parenchymal LD among the overall population of patients with sJIA treated with IL-1/IL-6 inhibitors. The association of PAP and paediatric haematological malignancies, especially myeloid leukaemia, can be ascribed in some cases to dysregulation of the GM-CSF/GM-CSF receptor axis and consequent macrophage dysfunction.61

We found an outsized risk of LD in children with T21 and sJIA. T21 carries increased susceptibility to adverse drug reactions62 and to viral pneumonia.63 Another contributing factor may be underlying type 1 interferonopathy, recently described in T21.64 An association of T21 and PAP in the context of haematological malignancy also has been reported.65

Drug hypersensitivity reactions can occur in children treated with biologic drugs for rheumatic disease.66 A subset of LD cases met the criteria for DReSS (online supplementary tables S1 and S8), a delayed form of severe drug-related hypersensitivity with organ involvement that can include lung.17,67,68 DReSS findings included dramatic eosinophilia, often despite concurrent steroids, together with extensive, persistent rash, frequently involving the face, which is uncommon in sJIA.18 Altered drug metabolism in childhood may increase risk of hypersensitivity reactions.69 Another consideration is drug-induced interstitial lung disease (DiILD), previously reported in children with rheumatic disease on biologics.70 DiILD has overlapping chest CT findings with LD in sJIA.71 No pathological findings are pathognomonic of DiILD,71,72 but PAP has been described.24 Drug cessation is indicated when DReSS or DiILD is recognised.68

Infection may exacerbate LD, trigger its detection or be causally linked. Pathogens identified at initial lung evaluation (online supplementary table S3) included rhinovirus (a cause of severe lower respiratory infection in young children73), herpes viruses and pneumocystis, all of which require IL-1 for optimal host defence.55,74,75 Pneumocystis pneumonia (PCP) is a recognised cause of PAP25 and is associated with high mortality in immunocompromised individuals.76 PCP risk is also elevated in DReSS.77 At least four of our cases had PCP; these were diagnosed by PCR of BAL, the preferred test in non-HIV immunosuppressed patients.76 It seems prudent to consider prophylaxis for patients with sJIA with lymphopaenia or steroid use (consistent with recommendations78) or T21.

LD in sJIA has been associated with MAS.13,79 In 23 out of 61 cases in our series, LD detection occurred with concurrent MAS. Out of these 23, 19 had their first MAS episode at or after LD diagnosis, suggesting that LD may trigger systemic inflammation. Consistent with this possibility, 4 of 11 cases with MAS co-occurred with initial detection of LD during three treatment trials of the inhibitors (n=331 subjects in total).46

PH with a range of severity was observed with or without pre-exposure to inhibitors and with or without PAP/ELP pathology. Out of 20 subjects, 2 lacked substantial parenchymal LD at PH detection. Together, these observations argue that PH in this LD cohort has heterogeneous biology.

Persistent arthritis (figure 4F) was less frequent in the pre-exposed LD cohort than in the sJIA cohort in the CARRA legacy registry (2010–2013),42 and of comparable frequency to children treated with IL-1 inhibition as first-line sJIA therapy. Inhibitor therapy may attenuate arthritis when given early or the LD cohort may be enriched for children with systemic inflammation-predominant disease.18,80

Over 50 additional LD cases in sJIA have been reported to us since closing this series. The Food and Drug Administration (FDA) adverse event website (FDA Adverse Event Reporting System) shows 39 adults (rheumatoid arthritis (23), AOSD (11), other (5)) developing alveolar disease or PH on IL-1/IL-6 inhibitors and 4 DReSS cases (second quarter, 2019). An apparent discrepancy between adverse event reports and the frequency of sJIA-LD reported here may reflect its underdiagnosis or under-reporting.81

The association between cytokine inhibition and sJIA-LD and related mechanistic hypotheses demand further investigation. We acknowledge the limitations of retrospective data, use of historical/published data for controls, possible biases as mentioned and false discovery issues associated with multiple hypothesis testing in a large data set. With these limitations, one cannot assign causality to cytokine inhibition in LD in sJIA. Likewise, it is premature to make treatment recommendations solely on the basis of our findings. Therapeutic decision making for patients with sJIA-associated LD is challenging, and currently individualised management is appropriate. However, in children with risk factors, close attention to subtle pulmonary symptoms is advised, and approaches for early detection of altered pulmonary function, guided by a pulmonary specialist, should be considered. In light of high fatality, efforts to determine LD prevalence, uncover molecular mechanism(s), and devise treatment and prevention approaches are urgently needed.

Supplementary Material

annrheumdis_Supplementary File

Key messages.

What is already known about this subject?

  • Pleuritis is common in systemic-onset juvenile idiopathic arthritis (sJIA), but parenchymal lung disease (LD) is very rare, with sporadic reports and one multicentre series of 25 cases (Kimura et al, 2013) in the literature.

Key messages.

What does this study add?

  • In the last decade, increasing numbers of cases of high fatality, parenchymal LD are being detected worldwide, with a majority characterised by unusual clinical and radiological features, in association with a variant of pulmonary alveolar proteinosis/endogenous lipoid pneumonia.

  • Children with trisomy 21 and sJIA or with sJIA onset at <5 years are at increased risk of LD, and an unexplained association with anaphylaxis to tocilizumab is observed.

  • Prodromal features can include lymphopaenia, rising serum ferritin and evidence of drug hypersensitivity (peripheral eosinophilia, extensive atypical rash).

How might this impact on clinical practice or future developments?

  • This study highlights risk factors, prodromal features and clinical characteristics that should raise suspicion of LD, which otherwise can remain subclinical until severe and life-threatening.

  • Clinician awareness of delayed drug hypersensitivity to interleukin (IL)-1 and IL-6 inhibitors could lead to earlier recognition and management, including cessation of the implicated drug.

  • Pneumocystis pneumonia may complicate LD in sJIA; consideration of prophylaxis is warranted.

Acknowledgements

The CARRA registry has been supported by the NIH’s National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and the Arthritis Foundation. We thank all participants and hospital sites that recruited patients for the CARRA registry. The authors thank Drs Joseph A Kovacs, Jay K Kolls and Sergio Vargas for their expert input on pneumocystis pneumonia, Dr James Verbsky for contributing unidentified genetic data, and Dr Xuan Qin for PCP staining of a subset of biopsies. The authors also thank Dr Yuki Kimura for critical reading of the manuscript, Dr Claudia Macaubas for statistical assistance and Ms Melinda Hing for assistance with the manuscript.

Funding This work was supported by the sJIA Foundation (EDM), the Lucile Packard Foundation for Children’s Health (EDM), CARRA-Arthritis Foundation grant (EDM, VES), Life Sciences Research Foundation (GC), Bio-X Stanford Interdisciplinary Graduate Fellowship (JoB), Stanford Graduate Fellowship and the Computational Evolutionary Human Genetics Fellowship (KJ), Bill & Melinda Gates Foundation (PK), and NIH 1U19AI109662 (PK), U19AI057229 (PK) and RO1 AI125197 (PK).

Competing interests VES reports personal fees from Novartis. GD reports personal fees from Novartis. SC reports personal fees from Novartis and grants from AB2 Bio. GS reports personal fees from Novartis. KB reports personal fees from Novartis. RQC is co-PI of an investigator-initiated clinical trial funded by SOBI. RD reports personal fees from Boehringer Ingelheim, other from NowVitals, personal fees and other from Triple Endoscopy, other from Earables, and NowVitals with patents and lung-related device development. AAG reports grants and personal fees from Novartis and grants from NovImmune. SL reports personal fees from Novartis. RS reports personal fees from Novartis, NovImmune and SOBI. SS reports personal fees from Novartis. MLS reports personal fees from Novartis. LRY reports other from Up-To-Date and other from Boehringer Ingelheim, outside the submitted work. EDM reports grants from Novartis.

Footnotes

Patient and public involvement statement Patients were not involved in the research process of this study. Results will be shared via CARRA patient communication mechanisms.

Patient consent for publication Not required.

Ethics approval Ethics approval was obtained through the Stanford University School of Medicine institutional review board. Contributing case reporters obtained approval per local institutional review board requirements. Institutional review board permission was obtained at each institution according to local requirements.

Provenance and peer review Not commissioned; externally peer reviewed.

Data availability statement Data are available upon reasonable request.

REFERENCES

  • 1.Gurion R, Lehman TJA, Moorthy LN. Systemic arthritis in children: a review of clinical presentation and treatment. Int J Inflam 2012;2012:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gerfaud-Valentin M, Cottin V, Jamilloux Y, et al. Parenchymal lung involvement in adult-onset still disease: a STROBE-compliant case series and literature review. Medicine 2016;95:e4258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Minoia F, Davì S, Horne A, et al. Clinical features, treatment, and outcome of macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: a multinational, multicenter study of 362 patients. Arthritis Rheumatol 2014;66:3160–9. [DOI] [PubMed] [Google Scholar]
  • 4.Ruperto N, Brunner HI, Quartier P, et al. Two randomized trials of canakinumab in systemic juvenile idiopathic arthritis. N Engl J Med 2012;367:2396–406. [DOI] [PubMed] [Google Scholar]
  • 5.De Benedetti F, Brunner HI, Ruperto N, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med 2012;367:2385–95. [DOI] [PubMed] [Google Scholar]
  • 6.Ter Haar NM, Dijkhuizen EHP, Swart JF, et al. Treatment to target using recombinant Interleukin-1 receptor antagonist as First-Line monotherapy in New-Onset systemic juvenile idiopathic arthritis: results from a Five-Year Follow-Up study. Arthritis Rheumatol 2019;71:1163–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.García-Peña P, Boixadera H, Barber I, et al. Thoracic findings of systemic diseases at high-resolution CT in children. RadioGraphics 2011;31:465–82. [DOI] [PubMed] [Google Scholar]
  • 8.A threya BH, Doughty RA, Bookspan M, et al. Pulmonary manifestations of juvenile rheumatoid arthritis. A report of eight cases and review. Clin Chest Med 1980;1:361–74. [PubMed] [Google Scholar]
  • 9.Padeh S, Laxer RM, Silver MM, et al. Primary pulmonary hypertension in a patient with systemic-onset juvenile arthritis. Arthritis & Rheumatism 1991;34:1575–9. [DOI] [PubMed] [Google Scholar]
  • 10.S ato K, Takahashi H, Amano H, et al. Diffuse progressive pulmonary interstitial and intra-alveolar cholesterol granulomas in childhood. Eur Respir J 1996;9:2419–22. [DOI] [PubMed] [Google Scholar]
  • 11.S chultz R, Mattila J, Gappa M, et al. Development of progressive pulmonary interstitial and intra-alveolar cholesterol granulomas (PICG) associated with therapy-resistant chronic systemic juvenile arthritis (CJA). Pediatr Pulmonol 2001;32:397–402. [DOI] [PubMed] [Google Scholar]
  • 12.L eber A, Carette S, Chapman KR, et al. A 21-year-old man with systemic-onset juvenile rheumatoid arthritis, cough and progressive dyspnea. Can Respir J 2010;17:e42–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kimura Y, Weiss JE, Haroldson KL, et al. Pulmonary hypertension and other potentially fatal pulmonary complications in systemic juvenile idiopathic arthritis. Arthritis Care Res 2013;65:745–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.DeWitt EM, Kimura Y, Beukelman T, et al. Consensus treatment plans for new-onset systemic juvenile idiopathic arthritis. Arthritis Care Res 2012;64:1001–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Beukelman T, Kimura Y, Ilowite NT, et al. The new Childhood Arthritis and Rheumatology Research Alliance (CARRA) registry: design, rationale, and characteristics of patients enrolled in the first 12 months. Pediatric Rheumatology 2017;15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.S wart J, Giancane G, Horneff G, et al. Pharmacovigilance in juvenile idiopathic arthritis patients treated with biologic or synthetic drugs: combined data of more than 15,000 patients from Pharmachild and national registries. Arthritis Res Ther 2018;20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kardaun SH, Mockenhaupt M, Roujeau J-C. Comments on: DRESS syndrome. J Am Acad Dermatol 2014;71:1000–1000.e2. [DOI] [PubMed] [Google Scholar]
  • 18.De Benedetti F, Schneider R. Systemic Juvenile Idiopathic Arthritis In: Petty R, Lindsley C, Laxer R, et al. , eds. Textbook of pediatric rheumatology. 7th edn. Philadelphia, PA: Elsevier, 2016: 206–16. [Google Scholar]
  • 19.van Gent R, van Tilburg CM, Nibbelke EE, et al. Refined characterization and reference values of the pediatric T- and B-cell compartments. Clin Immunol 2009;133:95–107. [DOI] [PubMed] [Google Scholar]
  • 20.Bonekamp D, Hruban RH, Fishman EK. The great mimickers: Castleman disease. Semin Ultrasound CT MR 2014;35:263–71. [DOI] [PubMed] [Google Scholar]
  • 21.Carucci LR, Halvorsen RA. Abdominal and pelvic CT in the HIV-positive population. Abdom Imaging 2004;29:631–42. [DOI] [PubMed] [Google Scholar]
  • 22.Fischer A, Antoniou KM, Brown KK, et al. An official European Respiratory Society/American Thoracic Society research statement: interstitial pneumonia with autoimmune features. Eur Respir J 2015;46:976–87. [DOI] [PubMed] [Google Scholar]
  • 23.Dell S, Cernelc-Kohan M, Hagood JS. Diffuse and interstitial lung disease and childhood rheumatologic disorders. Curr Opin Rheumatol 2012;24:530–40. [DOI] [PubMed] [Google Scholar]
  • 24.Wardwell NR, Miller R, Ware LB. Pulmonary alveolar proteinosis associated with a disease-modifying antirheumatoid arthritis drug. Respirology 2006;11:663–5. [DOI] [PubMed] [Google Scholar]
  • 25.S eymour JF, Presneill JJ. Pulmonary alveolar proteinosis: progress in the first 44 years. Am J Respir Crit Care Med 2002;166:215–35. [DOI] [PubMed] [Google Scholar]
  • 26.Doan ML, Guillerman RP, Dishop MK, et al. Clinical, radiological and pathological features of ABCA3 mutations in children. Thorax 2008;63:366–73. [DOI] [PubMed] [Google Scholar]
  • 27.Deutsch GH, Young LR, Deterding RR, et al. Diffuse lung disease in young children: application of a novel classification scheme. Am J Respir Crit Care Med 2007;176:1120–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.A bou Taam R, Jaubert F, Emond S, et al. Familial interstitial disease with I73T mutation: a mid- and long-term study. Pediatr Pulmonol 2009;44:167–75. [DOI] [PubMed] [Google Scholar]
  • 29.Golde DW et al. Defective lung macrophages in pulmonary alveolar proteinosis. Ann Intern Med 1976;85:304–9. [DOI] [PubMed] [Google Scholar]
  • 30.S akagami T, Uchida K, Suzuki T, et al. Human GM-CSF autoantibodies and reproduction of pulmonary alveolar proteinosis. N Engl J Med 2009;361:2679–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wert SE, Whitsett JA, Nogee LM. Genetic disorders of surfactant dysfunction. Pediatr Dev Pathol 2009;12:253–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Suzuki T, Maranda B, Sakagami T, et al. Hereditary pulmonary alveolar proteinosis caused by recessive CSF2RB mutations. Eur Respir J 2011;37:201–4. [DOI] [PubMed] [Google Scholar]
  • 33.Suzuki T, Sakagami T, Rubin BK, et al. Familial pulmonary alveolar proteinosis caused by mutations in CSF2RA. J Exp Med 2008;205:2703–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Martinez-Moczygemba M, Doan ML, Elidemir O, et al. Pulmonary alveolar proteinosis caused by deletion of the GM-CSFRα gene in the X chromosome pseudoautosomal region 1. J Exp Med 2008;205:2711–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hadchouel A, Wieland T, Griese M, et al. Biallelic mutations of methionyl-tRNA synthetase cause a specific type of pulmonary alveolar proteinosis prevalent on Réunion island. Am J Hum Genet 2015;96:826–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Turcu S, Ashton E, Jenkins L, et al. Genetic testing in children with surfactant dysfunction. Arch Dis Child 2013;98:490–5. [DOI] [PubMed] [Google Scholar]
  • 37.Wambach JA, Wegner DJ, DePass K, et al. Single ABCA3 mutations increase risk for neonatal respiratory distress syndrome. Pediatrics 2012;130:e1575–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kaufman KM, Linghu B, Szustakowski JD, et al. Whole-exome sequencing reveals overlap between macrophage activation syndrome in systemic juvenile idiopathic arthritis and familial hemophagocytic lymphohistiocytosis. Arthritis Rheumatol 2014;66:3486–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ogilvie EM, Fife MS, Thompson SD, et al. The ?174G allele of the interleukin-6 gene confers susceptibility to systemic arthritis in children: A multicenter study using simplex and multiplex juvenile idiopathic arthritis families. Arthritis Rheum 2003;48:3202–6. [DOI] [PubMed] [Google Scholar]
  • 40.Klotsche J, Raab A, Niewerth M, et al. Outcome and trends in treatment of systemic juvenile idiopathic arthritis in the German national pediatric rheumatologic database, 2000–2013. Arthritis Rheumatol 2016;68:3023–34. [DOI] [PubMed] [Google Scholar]
  • 41.A lexander M, Petri H, Ding Y, et al. Morbidity and medication in a large population of individuals with Down syndrome compared to the general population. Dev Med Child Neurol 2016;58:246–54. [DOI] [PubMed] [Google Scholar]
  • 42.Janow G, Schanberg LE, Setoguchi S, et al. The systemic juvenile idiopathic arthritis cohort of the childhood arthritis and rheumatology research alliance registry: 2010–2013. J Rheumatol 2016;43:1755–62. [DOI] [PubMed] [Google Scholar]
  • 43.Russo RAG, Katsicas MM. Patients with very early-onset systemic juvenile idiopathic arthritis exhibit more inflammatory features and a worse outcome. J Rheumatol 2013;40:329–34. [DOI] [PubMed] [Google Scholar]
  • 44.Behrens EM, Beukelman T, Gallo L, et al. Evaluation of the presentation of systemic onset juvenile rheumatoid arthritis: data from the Pennsylvania systemic onset juvenile arthritis registry (PASOJAR). J Rheumatol 2008;35:343–8. [PubMed] [Google Scholar]
  • 45.Davies R, Southwood T, Kearsley-Fleet L, et al. Mortality rates are increased in patients with systemic juvenile idiopathic arthritis. Arch Dis Child 2017;102:206–7. [DOI] [PubMed] [Google Scholar]
  • 46.Meyer KC, Raghu G, Baughman RP, et al. An official American Thoracic Society clinical practice guideline: the clinical utility of bronchoalveolar lavage cellular analysis in interstitial lung disease. Am J Respir Crit Care Med 2012;185:1004–14. [DOI] [PubMed] [Google Scholar]
  • 47.Midulla F, Villani A, Merolla R, et al. Bronchoalveolar lavage studies in children without parenchymal lung disease: cellular constituents and protein levels. Pediatr Pulmonol 1995;20:112–8. [DOI] [PubMed] [Google Scholar]
  • 48.Radhakrishnan D, Yamashita C, Gillio-Meina C, et al. Translational research in pediatrics III: bronchoalveolar lavage. Pediatrics 2014;134:135–54. [DOI] [PubMed] [Google Scholar]
  • 49.Meyer KC, Raghu G. Bronchoalveolar lavage for the evaluation of interstitial lung disease: is it clinically useful? Eur Respir J 2011;38:761–9. [DOI] [PubMed] [Google Scholar]
  • 50.DeGroot J, Vastert S, Giancane G, et al. Interstitial lung disease in systemic juvenile idiopathic arthritis in the PharmaChild registry. Pediatric Rheumatology Online Journal 2018;16:17.29540190 [Google Scholar]
  • 51.S tills GF. On a form of chronic joint disease in children. Medical Chirurgical Trans (Proceedings of the Royal Medical and Chirurgical Society), 1897:47–60. [PMC free article] [PubMed] [Google Scholar]
  • 52.A rdlie KG, Deluca DS, Segre AV, et al. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 2015;348:648–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.S ack U, Burkhardt U, Borte M, et al. Age-dependent levels of select immunological mediators in sera of healthy children. Clin Diagn Lab Immunol 1998;5:28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.I osef C, Alastalo T-P, Hou Y, et al. Inhibiting NF-κB in the developing lung disrupts angiogenesis and alveolarization. Am J Physiol Lung Cell Mol Physiol 2012;302:L1023–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.S hrivastava G, León-Juárez M, García-Cordero J, et al. Inflammasomes and its importance in viral infections. Immunol Res 2016;64:1101–17. [DOI] [PubMed] [Google Scholar]
  • 56.Huaux F, Lo Re S, Giordano G, et al. IL-1α induces CD11b low alveolar macrophage proliferation and maturation during granuloma formation. J Pathol 2015;235:698–709. [DOI] [PubMed] [Google Scholar]
  • 57.Periasamy S, Harton JA. Interleukin 1α(IL-1α) promotes pathogenic immature myeloid cells and IL-1β favors protective mature myeloid cells during acute lung infection. J Infect Dis 2018;217:1481–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Willart MAM, Deswarte K, Pouliot P, et al. Interleukin-1α controls allergic sensitization to inhaled house dust mite via the epithelial release of GM-CSF and IL-33. J Exp Med 2012;209:1505–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Uchida K, Beck DC, Yamamoto T, et al. Gm-Csf autoantibodies and neutrophil dysfunction in pulmonary alveolar proteinosis. N Engl J Med 2007;356:567–79. [DOI] [PubMed] [Google Scholar]
  • 60.Huaux F, De Gussem V, Lebrun A, et al. New interplay between interstitial and alveolar macrophages explains pulmonary alveolar proteinosis (PAP) induced by indium tin oxide particles. Arch Toxicol 2018;92:1349–61. [DOI] [PubMed] [Google Scholar]
  • 61.Dirksen U, Hattenhorst U, Schneider P, et al. Defective expression of granulocyte-macrophage colony-stimulating factor/interleukin-3/interleukin-5 receptor common beta chain in children with acute myeloid leukemia associated with respiratory failure. Blood 1998;92:1097–103. [PubMed] [Google Scholar]
  • 62.Hefti E, Blanco JG. Pharmacotherapeutic considerations for individuals with Down syndrome. Pharmacotherapy 2017;37:214–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ram G, Chinen J. Infections and immunodeficiency in Down syndrome. Clin Exp Immunol 2011;164:9–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.S ullivan KD, Evans D, Pandey A, et al. Trisomy 21 causes changes in the circulating proteome indicative of chronic autoinflammation. Sci Rep 2017;7:14818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.I naba H, Jenkins JJ, McCarville MB, et al. Pulmonary alveolar proteinosis in pediatric leukemia. Pediatr Blood Cancer 2008;51:66–70. [DOI] [PubMed] [Google Scholar]
  • 66.S oyer O, Demir S, Bilginer Y, et al. Severe hypersensitivity reactions to biological drugs in children with rheumatic diseases. Pediatr Allergy Immunol 2019;11. [DOI] [PubMed] [Google Scholar]
  • 67.Kardaun SH, Sekula P, Valeyrie-A llanore L, et al. Drug reaction with eosinophilia and systemic symptoms (dress): an original multisystem adverse drug reaction. results from the prospective RegiSCAR study. Br J Dermatol 2013;169:1071–80. [DOI] [PubMed] [Google Scholar]
  • 68.Martínez-Cabriales SA, Rodríguez-Bolaños F, Shear NH. Drug reaction with eosinophilia and systemic symptoms (dress): how far have we come? Am J Clin Dermatol 2019;20:236. [DOI] [PubMed] [Google Scholar]
  • 69.Kuyucu S, Caubet J-C. Hypersensitivity reactions to antiepileptic drugs in children: epidemiologic, pathogenetic, clinical, and diagnostic aspects. J Allergy Clin Immunol 2018;6:1879–91. [DOI] [PubMed] [Google Scholar]
  • 70.N isar MK, Östör AJK. Pulmonary complications of biological therapies in children and adults with rheumatic diseases. Paediatr Respir Rev 2013;14:236–41. [DOI] [PubMed] [Google Scholar]
  • 71.S keoch S, Weatherley N, Swift A, et al. Drug-Induced interstitial lung disease: a systematic review. J Clin Med 2018;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Flieder DB, Travis WD. Pathologic characteristics of drug-induced lung disease. Clin Chest Med 2004;25:37–45. [DOI] [PubMed] [Google Scholar]
  • 73.L auinger IL, Bible JM, Halligan EP, et al. Patient characteristics and severity of human rhinovirus infections in children. J Clin Virol 2013;58:216–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Piper SC, Ferguson J, Kay L, et al. The role of interleukin-1 and interleukin-18 in pro-inflammatory and anti-viral responses to rhinovirus in primary bronchial epithelial cells. PLoS One 2013;8:e63365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen W et al. Interleukin 1: an important mediator of host resistance against Pneumocystis carinii. J Exp Med 1992;176:713–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Robert-Gangneux F, Belaz S, Revest M, et al. Diagnosis of Pneumocystis jirovecii pneumonia in immunocompromised patients by real-time PCR: a 4-year prospective study. J Clin Microbiol 2014;52:3370–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kano Y, Ishida T, Hirahara K, et al. Visceral involvements and long-term sequelae in drug-induced hypersensitivity syndrome. Med Clin North Am 2010;94:743–59. [DOI] [PubMed] [Google Scholar]
  • 78.Park JW, Curtis JR, Moon J, et al. Prophylactic effect of trimethoprim-sulfamethoxazole for pneumocystis pneumonia in patients with rheumatic diseases exposed to prolonged high-dose glucocorticoids. Ann Rheum Dis 2018;77:644–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.S chulert GS, Yasin S, Carey B, et al. Systemic juvenile idiopathic Arthritis-Lung disease: characterization and risk factors. Arthritis Rheumatol 2019. doi: 10.1002/art.41073. [Epub ahead of print: 5 Aug 2019]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.N igrovic PA, Mannion M, Prince FHM, et al. Anakinra as first-line disease-modifying therapy in systemic juvenile idiopathic arthritis: report of forty-six patients from an international multicenter series. Arthritis Rheum 2011;63:545–55. [DOI] [PubMed] [Google Scholar]
  • 81.Ransohoff JD, Nikfarjam A, Jones E, et al. Detecting chemotherapeutic skin adverse reactions in social health networks using deep learning. JAMA Oncol 2018;4:581–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.A ntoon JW, Hernandez ML, Roehrs PA, et al. Endogenous lipoid pneumonia preceding diagnosis of pulmonary alveolar proteinosis. Clin Respir J 2016;10:246–9. [DOI] [PubMed] [Google Scholar]
  • 83.Fisher M, Roggli V, Merten D, et al. Coexisting endogenous lipoid pneumonia, cholesterol granulomas, and pulmonary alveolar proteinosis in a pediatric population: a clinical, radiographic, and pathologic correlation. Pediatr Pathol 1992;12:365–83. [DOI] [PubMed] [Google Scholar]
  • 84.Pardeo M, Pires Marafon D, Insalaco A, et al. Anakinra in systemic juvenile idiopathic arthritis: a single-center experience. J Rheumatol 2015;42:1523–7. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

annrheumdis_Supplementary File

RESOURCES