Next Article in Journal
The Impact of Polychlorinated Biphenyls on the Development of Zebrafish (Danio rerio)
Previous Article in Journal
Prognostic Protein Biomarker Screening for Thyroid Carcinoma Based on Cancer Proteomics Profiles
Previous Article in Special Issue
Leveraging Hypotension Prediction Index to Forecast LPS-Induced Acute Lung Injury and Inflammation in a Porcine Model: Exploring the Role of Hypoxia-Inducible Factor in Circulatory Shock
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Adult-Onset Still’s Disease (AOSD)—On the Basis of Own Cases

by
Małgorzata Wisłowska
Rheumatology Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, 1 Spartanska Street, 02-637 Warsaw, Poland
Biomedicines 2024, 12(9), 2067; https://doi.org/10.3390/biomedicines12092067
Submission received: 19 June 2024 / Revised: 22 July 2024 / Accepted: 26 August 2024 / Published: 10 September 2024

Abstract

:
Introduction: Adult-onset Still’s disease (AOSD) is a rare chronic autoinflammatory condition characterized by a spiking fever, arthritis, a rash, hepatosplenomegaly, lymphadenopathy, leucocytosis, and hyperferritinemia. It is sometimes accompanied by life-threatening complications like macrophage activation syndrome/hemophagocytic lymphohistiocytosis (MAS/HLH). Treatment options for AOSD include glucocorticoids (GCs), immunosuppressive drugs, biological medications, and Janus kinase (JAK) inhibitors. The features that differentiate MAS/HLH from AOSD are: in MAS/HLH, a different type of fever, which is persistent, a sharp decrease in the number of leukocytes and thrombocytes, a further increase in the level of transaminases and ferritin, significant hepatosplenomegaly, lymphadenopathy, symptoms of the central nervous system (CNS), disseminated intravascular coagulation (DIC) and hemophagocytosis in the bone marrow. This study aimed to evaluate the course of AOSD, which results in MAS/HLD. Patients and methods: Nine AOSD patients, four of whom developed MAS/HLH, were treated at the Rheumatology Clinic in the Central Clinical Hospital of the Ministry of Interior Affairs from 1 January 2015 to 15 March 2020 and at the Rheumatology Clinic in the National Institute of Geriatric, Rheumatology and Rehabilitation from 1 September 2021 to 1 March 2024. Medical history, clinical data, demographic data, laboratory data, imaging data, Hscore, and treatment data were collected. Results: All the patients with MAS and an Hscore above 150 recovered. Discussion: MAS/HLH requires rapid diagnosis as well as treatment with methylprednisolone pulses, cyclosporine A, and etoposide. When comparing patients who developed MAS/HLH with those who did not, possible risk factors were identified: the presence of pregnancy (two cases) and an aggressive course of AOSD. The Hscore is a useful tool for identifying patients with MAS/HLH.

1. Introduction

Adult-onset Still’s disease (AOSD) was described by Eric Bywaters in 1970 as an inflammatory disease in young adults [1]. It is part of the spectrum of systemic-onset juvenile idiopathic arthritis (SJIA), which was described one hundred years ago by Sir John Still [2]. AOSD is a rare, difficult-to-diagnose systemic autoimmune inflammatory disease of unknown etiology, and is influenced by both genetic and environmental factors [3]. In Poland, the annual incidence rate of AOSD is 0.32 per 100,000, and the prevalence is 2.7 per 100,000 [4].
Still’s disease is characterized by a daily recurrent spiking fever that does not respond to treatment, and is accompanied by either two major criteria OR one major criterion and two minor criteria.
The major criteria include the following:
  • Transient (non-persistent) erythematous rash in the form of salmon-colored spots, most often located on the trunk and limbs; the rash is macular or slightly urticarial, and sometimes persistent red-brownish papules and plaques may be seen.
  • Arthritis (initially it may be very discreet, but then it causes significant damage to the musculoskeletal system).
The minor criteria include the following:
  • Generalized lymphadenopathy and/or hepatomegaly and/or splenomegaly.
  • Serositis.
  • Arthralgia lasting 2 weeks or more (in the absence of arthritis).
  • Leukocytosis (≥15,000/μL) with neutrophilia [5].
In Still’s disease, the course may vary in following forms:
  • A monocyclic course, characterized by a single episode followed by persistent remission.
  • A polycyclic course is characterized by recurrent flares and remissions.
  • A chronic course [6].
There are two subtypes of Still’s disease in adulthood:
  • Systemic AOSD is characterized by an increased concentration of the interleukin (IL)-1β, IL-18, interferon (INF)α/β, INFγ, IL-4, IL-10, hyperferritinemia, thrombocytopenia, and Natural-killer (NK) cell dysfunction. Clinical symptoms include fever, serositis, hepatitis, and MAS [6]. Monoarthritis is more prevalent in systemic AOSD, which correlates with the lower frequency of MAS among patients with polyarthritis, the chronic articular disease course, and the lower inflammatory markers [6]. MAS can present with wide range of hepatic dysfunction, from mild elevation of the transaminases to liver failure. [6]. Treatment should include IL-1, IL-18, or INFγ inhibitors [7].
  • Articular AOSD is associated with increased concentrations of IL-17, IL-23, tumor necrosis factor (TNF)α, and IL-6, as well as thrombocytosis. The clinical symptoms include arthritis with joint destruction. The recommended treatment includes IL-6 or TNFα or IL-17 inhibitors [7].
Table 1 presents different classification criteria for AOSD, e.g., Cush [8], Yamagushi [9], and Fautrel [10], and ILAR criteria for SJIA [11]. These criteria are very similar, but the most commonly used are Yamaguchi criteria [9], which have over 90% sensitivity. Notably, these criteria do not include increased ferritin levels, which have a high diagnostic value [9]. The Fautrel criteria include glycosylated ferritin, which further improves the diagnosis [10].
Table 2 shows the frequency of symptoms [12], typical joint involvement [8], and laboratory test abnormalities [13,14] in AOSD. Often, inflammation of the wrists and metacarpophalangeal (MCP) joints can lead to ankylosis.
The diagnosis of Still’s disease requires a broad differential diagnosis, including the following:
  • Severe viral infections (rubella, hepatitis, parvoviruses, coxsackie viruses, Epstein–Barr viruses [EBVs], cytomegaloviruses [CMVs], human immunodeficiency virus [HIV], severe acute respiratory syndrome coronavirus-2 [SARS-CoV-2], pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2 [PIMS-TS]), or bacterial infections (bacterial endocarditis, meningococcal infection, brucellosis, yersiniosis, tuberculosis, Lyme disease, syphilis, as well as sepsis.
  • Malignancies, including leukemia, lymphoma, neuroblastoma, and paraneoplastic syndromes.
  • Rheumatic fever.
  • Kawasaki disease.
  • Other systemic connective tissue diseases, like systemic lupus erythematosus, idiopathic myositis, mixed connective tissue disease, rheumatoid arthritis, and vasculitis.
  • Other autoinflammatory diseases related to IL-1, like Mediterranean fever, cryopyrin-associated periodic syndromes (CAPSs), TNF receptor-associated periodic fever syndrome (TRAPS), and mevalonate kinase deficiency (MKD) [5].
The differential diagnosis should also include the following: sarcoidosis, reactive arthritis, Schnitzler syndrome, Sweet syndrome, and hypersensitivity syndrome on a drug [5].
Complications of Still’s disease may include the following: pleurisy, pericarditis, cardiac tamponade, myocarditis, endocarditis, disseminated intravascular coagulation (DIC), pulmonary fibrosis, pulmonary hypertension, vesicular bleeding, kidney insufficiency, neuropathy, aseptic meningitis, cataracts, fulminant hepatitis, thrombotic thrombocytopenic purpura, and MAS/HLH [5,8,14].
Symptoms of MAS include persistent high fever, hepatosplenomegaly, generalized lymphadenopathy, CNS symptoms (headaches, disturbances of consciousness, coma), DIC, respiratory, circulatory and urinary insufficiency, and a rash [15]. Laboratory tests show relative cytopenia, which might be in the normal range, involving ≥ 2 cell lines, as well as leucopenia, lymphopenia, anemia, thrombocytopenia, increased AST activity, hypertriglyceridemia, hypofibrinogenemia, hyperferritinemia, decreased albumin levels, increased lactate dehydrogenase (LDH) activity > 900 U/L, decreased or absent NKs activity, and hemophagocytosis in the bone marrow [15]. According to Ravelli et al. [15], the following values confirming MAS were observed: ferritin > 684 ng/mL, platelet count ≤ 181 × 109/L, AST > 48 U/L, triglycerides > 156 mg/dL, and fibrinogen ≤ 360 mg/dL.
Symptoms of MAS include persistent high fever, hepatosplenomegaly, generalized lymphadenopathy, CNS symptoms (headaches, disturbances of consciousness, coma), DIC, respiratory, circulatory and urinary insufficiency, and a rash [15]. Laboratory tests show relative cytopenia, which might be in the normal range, involving ≥ 2 cell lines, as well as leucopenia, lymphopenia, anemia, thrombocytopenia, increased AST activity, hypertriglyceridemia, hypofibrinogenemia, hyperferritinemia, decreased albumin levels, increased lactate dehydrogenase (LDH) activity > 900 U/L, decreased or absent NKs activity, and hemophagocytosis in the bone marrow [15]. According to Ravelli et al. [15], the following values confirming MAS were observed: ferritin > 684 ng/mL, platelet count ≤ 181 × 109/L, AST > 48 U/L, triglycerides > 156 mg/dL, and fibrinogen ≤ 360 mg/dL.
Symptoms of MAS include persistent high fever, hepatosplenomegaly, generalized lymphadenopathy, CNS symptoms (headaches, disturbances of consciousness, coma), DIC, respiratory, circulatory and urinary insufficiency, and a rash [15]. Laboratory tests show relative cytopenia, which might be in the normal range, involving ≥ 2 cell lines, as well as leucopenia, lymphopenia, anemia, thrombocytopenia, increased AST activity, hypertriglyceridemia, hypofibrinogenemia, hyperferritinemia, decreased albumin levels, increased lactate dehydrogenase (LDH) activity > 900 U/L, decreased or absent NKs activity, and hemophagocytosis in the bone marrow [15]. According to Ravelli et al. [15], the following values confirming MAS were observed: ferritin > 684 ng/mL, platelet count ≤ 181 × 109/L, AST > 48 U/L, triglycerides > 156 mg/dL, and fibrinogen ≤ 360 mg/dL.
Both HLH/MAS and SJIA/AOSD present with a high fever, hepatosplenomegaly, lymphadenopathy, liver dysfunction, hyperferritinemia, and coagulopathy.
The primary distinction is the level of neutrophilia, which increases in AOSD/SJIA and decreases in HLH [5,8]. MAS indicators include an increase in ferritin levels, a sharp decrease in the number of leukocytes and an increase in the level of transaminases.
IL-18 affects NKs and the cluster of differentiation (CD)8+ cells, leading to excessive secretion of INFy and hemophagocytic transformation of macrophages [16]. IL-1, the inflammasome receptor, and the Nod-like receptor (NLR) 3 play a similar role [17].
Hematologists use the “Hscore” to diagnose HLH. This score assesses the likelihood of HLH in a patient. An Hscore of ≤90 HLH may indicate that HLH can be excluded, while a value ≥250 suggests a greater than 99% probability of HLH [18]. Table 3 presents the Hscore criteria for the diagnosis of HLH.
The purpose of this study was to evaluate the progression of AOSD, which can result in MAS/HLD, a severe complication of AOSD. An intriguing open question remains regarding cases that are more susceptible to this serious complication, and this paper attempts to answer it and to offer the patient a successful treatment.

2. Materials and Methods

Nine patients with severe AOSD were treated at the Rheumatology Clinic in the Central Clinical Hospital of the Ministry of Interior Affairs Warsaw, Woloska 137 street, Poland between 1 January 2015 and 15 March 2020 (Head of Clinic prof. M. Wislowska MD, PhD) and at the Rheumatology Clinic in the National of Institute of Geriatric, Rheumatology and Rehabilitation Warszaw, Spartanska 1 street, Poland between 1 September 2021 and 1 February 2024 (Head of Clinic prof. M. Wislowska MD, PhD). The demographic data, medical history, clinical data, laboratory data, and imaging data were collected. The medical treatments, side effects, and outcomes were recorded chronologically. All data were collected at the time of the AOSD diagnosis and at the time of the MAS diagnosis. Hscores were retrospectively calculated [18]. Table 4 presents nine cases of AOSD, detailing their age, sex, symptoms, and laboratory data, which confirmed the AOSD diagnosis and the medical treatment pursued. During hospitalization, four cases developed MAS/HLH syndrome; among these were two pregnant patients.

3. Ethics

Ethical approval was obtained from the institutional review board [N. KBT-4/3/2024]. All the patients signed informed consent forms.

4. Results

In four cases, complications like MAS/HLH were observed. The symptoms, laboratory data confirming MAS/HLH syndrome, and the treatment of these patients are presented in Table 5. The differences between patients with or without MAS are the following: hepatomegaly, a rapid decrease in the level of thrombocytes in one case, and a rapid increase in the level of ferritin. In all patients who developed MAS, a change in the fever pattern from spiking to a persistent high fever was observed. All patients had hepatomegaly and elevated transaminases, with one case exceeding 2000 IU/L. A decrease in erythrocytes was observed in two patients, leukopenia was observed in one patient, and thrombocytopenia was observed in three patients. Furthermore, all patients presented with elevated ferritin levels. The Hscores for all patients exceeded 150 points and are presented in Table 6. As treatment, all patients received glucocorticosteroids in pulses; additionally, three patients received cyclosporine A and IVIG, two received tocilizumab, and one underwent plasmapheresis. All the patients were treated immediately and therefore, they were successfully cured.

5. Discussion

A dangerous complication of SJIA and AOSD is MAS/HLH, a potentially life-threatening complication characterized by a systemic inflammatory reaction due to an uncontrolled and dysfunctional immune response, which results in a massive hypersecretion of pro-inflammatory cytokines [15]. The basis of this syndrome is the uncontrolled expansion of T cells and macrophages with a dysfunction of NK and cytotoxic CD8+ cells, as well as a reduced expression of perforin and granzyme H, which results in an escalation of the production of the INFy and pro-inflammatory cytokines as well as macrophage hemophagocytosis [15].
SJIA and AOSD are categorized as polygenic autoinflammatory diseases. There are distinct mechanisms that differentiate autoimmune diseases (“autoimmunities”) from autoinflammatory diseases (“autoinflammation”). “Autoimmunity” involves a dysregulation of adaptive immunity, resulting in an aberrant response of the dendritic cells, B cells, and T cells to their own tissues, which leads to a breakdown of tolerance and immunological responses to self-antigens, accompanied by the production of autoantibodies. In contrast, “autoinflammation” refers to the dysregulation of an innate immunity that triggers excessive activation of phagocytes, including macrophages and neutrophils, in response to stress factors such as PAMSs (pathogen-associated molecular patterns) and DAMPs (danger-associated molecular patterns), resulting in tissue damage [19]. While autoimmune diseases are easily diagnosed by the presence of antibodies, there are no specific biomarkers for diagnosing systemic autoinflammatory disorders (SAIDs) [20]. Still’s disease is a polygenic non-familial SAID that responds to treatment with IL-1 blockers. Infectious agents such as bacteria and viruses, including Yersinia enterocolitica, Mycoplasma pneumoniae, rubella, and parvovirus B19, can act as triggers [21]. Examining the liver enzymes and cytokines as biomarkers will contribute to the differential diagnosis of infection-associated diseases, e.g., viral hepatitis or COVID-19, but the specificity issues of these biomarkers should be carefully considered [22,23].
Toll-like receptors (TLRs) play crucial roles in detecting viral nucleic acids, a key type of viral PAMP and DAMP, and initiate signaling pathways that lead to the activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB) [24]. The tripartite-motif protein-56 (TRIM56) enhances the induction of the type I interferon (INF) response through the TLR3 pathway by boosting IRF3 activation [25]. The depletion of chaperone proteins, like the glucose-regulated protein 78 kDa (GRP78), hampers the establishment of an antiviral state mediated by TLR3 [26]. Upon binding the PAMP molecules to the TLRs in macrophages and neutrophils, the activated Nod-like receptor (NLR) 3 forms a complex called an inflammasome that activates caspase 1 or 11. The caspases in the inflammasome lead to an overproduction of IL-1β, TNF, IL-6, IL-8, IL-17, and IL-18, triggering a “cytokine storm”. Additionally, alarmins, such as the S100 and S100A12 proteins, activate the macrophages and neutrophils. A disruption of the anti-inflammatory mechanisms and a deficiency in regulatory T cells, NKs, cells and IL-10 occur alongside these processes. The genetic background is also a significant factor [5]. Genetic factors in SJIA include the presence of the human leukocyte antigen (HLA)—Bw35, DR2, DR4, DR5, and Dw7— as well as the presence of macrophage migration inhibitory factors (MIFs), IL6, IL18, TNFα polymorphisms, and, in AOSD, HLA- Bw35, DR2, DR4, DRB1, Dw7, and MIFs, which are IL18 polymorphisms [5].
In AOSD, the adaptive immune system plays a minor role. Markers such as macrophage colony-stimulating factor (M-CSF) and INF-γ, calprotectin, MIFs, and intercellular adhesion molecule (ICAM)-1, which activates monocytes, correlate with disease activity [27]. In SJIA, there is a disturbed regulation of innate immune receptors such as NLR, TLR5, and TLR4. TLR4 recognizes S100 proteins. High concentrations of circulating S100 proteins occur in the active phase of the disease, and the S100-A8/S100-A9 complex, through TLR4, triggers the secretion of IL-1β [27]. IL-1β likely initiates AOSD by causing systemic inflammation and stimulating damage to cartilage and bone [27]. High levels of IL-18 are found in the serum, synovium, lymph nodes, and liver of patients with SJIA and AOSD. IL-18 induces a T helper (Th) 1 response and affects the secretion of INF-γ by cytotoxic CD8+ and NKs and it has been suggested that it plays a major role in the pathogenesis of reactive hemophagocytic lymphohistiocytosis [28]. IL-6 and TNFα also contribute to the pathogenesis of AOSD, although TNFα levels do not correlate with disease activity [21]. IL-17 increases inflammation, stimulates the production of chemokines that recruit neutrophils, enhances granulopoiesis and myelopoiesis, and plays an important role in the development of arthritis in AOSD [29]. In SJIA and AOSD, there are not only pro-inflammatory mechanisms, but also defects in immunoregulation [30].
High serum ferritin levels indicate macrophage activation and correlate with the disease activity in AOSD, serving as a sensitive marker [13]. Besides ferritin, glycosylated ferritin (GF) is crucial for diagnosis. Normally, it accounts for over half the ferritin level. However, its level drops to 20–50% in inflammatory diseases and falls below 20% in AOSD [31,32].
Procalcitonin, typically a marker of severe systemic infection, may rise in patients with active AOSD, thus complicating the differentiation between acute infections and AOSD flare [5]. Additionally, increases in IL-1, IL-6, IL-18, TNF, and INFγ have been observed in AOSD [27].
These processes are still not fully understood, especially in AOSD patients who develop HLH/MAS. According to a study by Javaux et al. [33], which analyzed 206 patients with Still’s disease across nine regional French centers from January 2001 to March 2021, 20 of the patients developed MAS. Patients with MAS were more likely to exhibit hepatosplenomegaly and neurological symptoms. Similarly, in my small cohort, I observed hepatosplenomegaly in my patients. Javaux et al. [33] also noted that extreme hyperferritinemia at the onset of Still’s disease is a prognostic factor for the development of MAS. Not all of my MAS patients exhibited extreme hyperferritinemia.
An important paper discussed the clinical and laboratory features associated with macrophage activation syndrome in Still’s disease based on data from the international Autoinflammatory Disease Alliance (AIDA) Still’s Disease Registry [34]. Of the 414 patients with Still’s disease included in the study, 39 of them developed MAS. A multivariate analysis showed direct associations between MAS, hepatomegaly, and monoarthritis, while a normal platelet count or thrombocytosis appeared to be protective [34].
Nonsteroidal anti-inflammatory drugs (NSAIDs) are used as the first-line drugs in the treatment of AOSD before diagnosis is established. After diagnosis, GCs should be administered. The best response is observed with an initial dose of 0.8–1 mg/kg of prednisone, to which approximately 60% of patients respond, or after pulsatile administration of methylprednisolone at a dose of 500–1000 mg/day for 2–3 days. Synthetic disease modifying anti-rheumatic drugs (DMARDs), such as methotrexate in a dosage of up to 25 mg/week (approximately 70% effectiveness), along with other options like azathioprine, leflunomide, cyclosporine A, and tacrolimus, are considered. Patients with AOSD should not be treated with sulfasalazine [5,14,35].
Treatment of AOSD depends on the clinical picture. In the case of arthralgia or arthritis predominance, TNF inhibitors are preferred as second-line medications, but they are ineffective for HLH [36]. In cases of fever, rash, pharyngitis, and serositis, leukocytosis IL-1β inhibitors (anakinra, canakinumab, rilonacept) are the treatment of choice [37,38,39]. Anakinra, an interleukin 1 receptor antagonist, is administered at a dose of 2 mg/kg to 4 mg/kg of body weight, with 100–200 mg given subcutaneously (SC) daily [37]; canakinumab, an anti-interleukin 1 antibody, is given at a dose of 4 mg/kg of body weight every 4 weeks, with a maximum of 300 mg given SC every 4 weeks [38]; rilonacept, a fusion protein composed of the human extracellular domain of the IL-1 receptor and the Fc fragment of the IgG1 immunoglobulin, is administered SC at a dose of 100–320 mg every week [39]. In clinical studies, complete remission was observed in 73% of cases after anakinra treatment, in 55% after rilonacept treatment, and 75% after canakinumab treatment [37,38,39]. Tadekinig alfa, an IL-18 binding protein inhibitor, is given subcutaneously at a dose of 80–160 mg every week [16]. In cases of HLH/MAS, hyperferritinemia, and liver involvement, the IL-1 inhibitors or the IL-6 receptor inhibitor (Tocilizumab) [40] as well as the JAK inhibitors (Tofacitinib, Baricitinib) [41,42] are very useful. Approximately 20–25% of patients with AOSD are treated with biological DMARDs [36,37,38,39,40].
Tocilizumab, an interleukin 6 receptor antagonist, is administered IV and SC at a dose of 162 mg every week or 8 mg/kg of body weight every 2 weeks. It is effective in treating arthritis and systemic symptoms [40].
The risk of MAS in patients with AOSD treated with IL-1 and IL-6 inhibitors shows that the incidence of MAS is significantly higher in patients treated with tocilizumab (p = 0.01) [43].
Some AOSD patients may be treated with rituximab (1 g given IV on days 1 and 15, and subsequent cycles every 6 months) [44] or cyclophosphamide [45]. Intravenous immunoglobulins (IVIG) administered at 0.4g/kg to 2 g/kg of body weight for 2–5 days a month have shown potential efficiency [46]. Abatacept has been used in the treatment of AOSD in some patients [47]. The inhibitor IL-17 is under preliminary observations [48].
MAS can lead to progressive multi-organ failure and, eventually, a fatal outcome [48]. According to the 2022 EULAR/ACR guidelines for the early stages of diagnosis and the management of suspected hemophagocytic lymphohistiocytosis/macrophage activation syndrome (HLH/MAS), HLH and MAS are life-threatening systemic hyperinflammatory syndromes characterized by a fever, elevated ferritin levels, low blood cell counts, DIC, hepatitis, CNS inflammation, and a high risk of progression to multiple organ dysfunction, shock, and even death [49]. The term HLH originally described a pathological condition in young children as a “primary” genetic defect. MAS, on the other hand, is a complication seen in rheumatic diseases such as SJIA or systemic lupus erythematosus (SLE). The task force agreed to refer to the entire spectrum of primary or secondary HLH as “HLH/MAS”. This condition can occur in any age group and develops in the context of infectious, malignant or rheumatological diseases, or due to the congenital errors of immunity that predispose people to hyperinflammation. However, early recognition of MAS/HLH is often challenging yet crucial [49].
MAS/HLH requires quick diagnosis and immediate intensive treatment with methylprednisolone pulses, cyclosporine A, and etoposide. The neutralization of the IL-18/INFγ axis is a promising new therapeutic target in MAS/HLH [16]. Emapalumab, an INFy blocker, is a recombinant human IgG1 monoclonal antibody to gamma interferon, which inhibits its binding to the cell surface interferon receptors and the subsequent activation of the intracellular pro-inflammatory signaling pathways [50]. Gamma interferon levels are elevated in HLH patients. Emapalumab was approved for use in HLH in the United States in 2018. It is available as a solution in single-dose vials of 10 mg in 2 mL or 50 mg in 10 mL (5 mg/mL). The recommended starting dose is 1 mg/kg as an intravenous infusion twice per week. Doses can be adjusted based on clinical and laboratory results and therapy can be continued until hematopoietic cell transplantation is performed, unacceptable toxicity occurs, or it is no longer deemed necessary.
The most critical factor for the effective treatment of MAS/HLH is the immediate diagnosis of this syndrome. Comparing patients who developed MAS/HLH with those without these complications, an additional factor that determined a more severe disease course was the concomitant pregnancy in patients N.1 and N.8. Patient N.7 had a very long and aggressive course of the disease and had previously suffered one incident of MAS/HLH. However, patient N.9 had a very aggressive onset of the disease and developed MAS, which resolved after a treatment of high doses of GCs. In the cases I presented, MAS/HLH syndrome was diagnosed immediately, supporting the effectiveness of aggressive treatment. A limitation of this study is the small number of patients; however, the disease is very rare. The Polish population is about 38 million people, but the annual incidence rate of AOSD in Poland is 0.32 per 100,000 people, and the incidence of MAS in these patients is even rarer [4].

6. Conclusions

AOSD is a rare systemic autoinflammatory disease that can have a severe course and is accompanied by a wide spectrum of symptoms and complications, including MAS/HLH. Activation of the innate immune response and overproduction of several inflammatory cytokines, such as IL-1, IL-6, and IL-18, play key role in its pathogenesis. A high ferritin concentration is most commonly observed in the cases of AOSD or HLH. An immediate diagnosis and early, aggressive treatment of MAS/HLH during the course of AOSD can ensure effective management of this life-threatening complication.

Funding

This research received no external funding.

Institutional Review Board Statement

Ethical approval was obtained from the institutional review board [N. KBT-4/3/2024].

Informed Consent Statement

All the patients signed informed consent forms.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author.

Acknowledgments

Thank you to patient N. 8, presented by Krzysztof Bonek.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Bywaters, E.G. Still’s disease in the adult. Ann. Rheum. Dis. 1971, 30, 121–133. [Google Scholar] [CrossRef] [PubMed]
  2. Still, G.F. On a form of chronic joint disease in children. Med. Chir. Trans. 1897, 80, 9. [Google Scholar] [CrossRef]
  3. Martini, A.; Ravelli, A.; Avcin, T.; Beresford, M.W.; Burgos-Vargas, R.; Cuttica, R.; Ilowite, N.T.; Khubchandani, R.; Laxer, R.M.; Lovell, D.J.; et al. Toward New Classification Criteria for Juvenile Idiopathic Arthritis: First Steps, Pediatric Rheumatology International Trials Organization International Consensus. J. Rheumatol. 2018, 46, 190–197. [Google Scholar] [CrossRef] [PubMed]
  4. Bogdan, M.; Osuch, A.; Samel-Kowalik, P.; Gorynski, P.; Tyszko, P.; Kanecki, K. Adult-onset Still’s disease in Poland—A nationwide population-based study. Ann. Agric. Environ. Med. 2021, 28, 250–254. [Google Scholar] [CrossRef]
  5. Feist, E.; Mitrovic, S.; Fautrel, B. Mechanisms, biomarkers and targets for adult-onset Still’s disease. Nat. Rev. Rheumatol. 2018, 14, 603–618. [Google Scholar] [CrossRef]
  6. Shumizu, M.; Nakagishi, Y.; Yachie, A. Distinct subsets of patients with systemic juvenile idiopathic arthritis based on their cytokine profiles. Cytokine 2013, 61, 345–348. [Google Scholar] [CrossRef]
  7. Ichida, H.; Kawaguchi, Y.; Sugiura, T.; Takagi, K.; Katsumata, Y.; Gono, T.; Ota, Y.; Kataoka, S.; Kawasumi, H.; Yamanaka, H. Clinical Manifestations of Adult-Onset Still’s Disease Presenting with Erosive Arthritis: Association with Low Levels of Ferritin and Interleukin-18. Arthritis Care Res. 2013, 66, 642–646. [Google Scholar] [CrossRef]
  8. Cush, J.J.; Medsger, T.A., Jr.; Christy, W.C.; Herbert, D.C.; Cooperstein, L.A. Adult-onset Still’s disease: Clinical course and outcome. Arthritis Rheum. 1987, 30, 186–194. [Google Scholar] [CrossRef]
  9. Yamaguchi, M.; Ohta, A.; Tsunematsu, T.; Kasukawa, R.; Mizushima, Y.; Kashiwagi, H.; Kashiwazaki, S.; Tanimoto, K.; Matsumoto, Y.; Ota, T. Preliminary criteria for classification of adult Still’s disease. J. Rheumatol. 1992, 19, 424–430. [Google Scholar]
  10. Fautrel, B.; Zing, E.; Golmard, J.-L.; Moel, G.L.E.; Bissery, A.; Rioux, C.; Rozenberg, S.; Piette, J.-C.; Bourgeois, P. Proposal for a New Set of Classification Criteria for Adult-Onset Still Disease. Medicine 2002, 81, 194–200. [Google Scholar] [CrossRef]
  11. Petty, R.E.; Southwood, T.R.; Manners, P.; Baum, J.; Glass, D.N.; Goldenberg, J.; He, X.; Maldonado-Cocco, J.; Orozco-Alcala, J.; Prieur, A.-M.; et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second revision, Edmonton, 2001. J. Rheumatol. 2004, 31, 390–392. [Google Scholar] [PubMed]
  12. Tomaras, S.; Goetzke, C.C.; Kallinich, T.; Feist, E. Adult-Onset Still’s Disease: Clinical Aspects and Therapeutic Approach. J. Clin. Med. 2021, 10, 733. [Google Scholar] [CrossRef]
  13. Fautrel, B. Adult-onset Still disease. Best Pract. Res. Clin. Rheumato. 2008, 22, 779–792. [Google Scholar] [CrossRef]
  14. Pouchot, J.; Sampalis, J.S.; Beaudet, F.; Carette, S.; Décary, F.; Salusinsky-Sternbach, M.; Hill, R.; Gutkowski, A.; Harth, M.; Myhal, D.; et al. Adult Still’s disease: Manifestations, disease course and outcome in 62 patients. Medicine 1991, 70, 118–136. [Google Scholar] [CrossRef] [PubMed]
  15. Ravelli, A.; Minoia, F.; Davì, S.; Horne, A.; Bovis, F.; Pistorio, A.; Aricò, M.; Avcin, T.; Behrens, E.M.; De Benedetti, F.; et al. 2016 classification criteria for macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: A European League against Rheumatism/American College of Rheumatology/Pediatric Rheumatology International Trials Organisation Collaborative Initiative. Arthritis Rheumatol. 2016, 68, 566–576. [Google Scholar]
  16. Gabay, C.; Fautrel, B.; Rech, J.; Spertini, F.; Feist, E.; Kötter, I.; Hachulla, E.; Morel, J.; Schaeverbeke, T.; Hamidou, M.A.; et al. Open-label, multicentre, dose-escalating phase II clinical trial on the safety and efficacy of tadekining alfa (IL-18BP) in adult-onset Still’s disease. Ann. Rheum. Dis. 2018, 77, 840–847. [Google Scholar]
  17. Hayem, F.; Hayem, G. Still’s disease and the mitochondrion: The other face of an old friend? Med. Hypothese 2012, 79, 136–137. [Google Scholar] [CrossRef] [PubMed]
  18. Fardet, L.; Galicier, L.; Lambotte, O.; Marzac, C.; Aumont, C.; Chahwan, D.; Coppo, P.; Hejblum, G. Development and Validation of the HScore, a Score for the Diagnosis of Reactive Hemophagocytic Syndrome. Arthritis Rheumatol. 2014, 66, 2613–2620. [Google Scholar] [CrossRef] [PubMed]
  19. Jamilloux, Y.; Gerfaud-Valentin, M.; Martinon, F.; Belot, A.; Henry, T.; Sève, P. Pathogenesis of adult-onset Still’s disease: New insights from the juvenile counterpart. Immunol. Res. 2014, 61, 53–62. [Google Scholar] [CrossRef]
  20. McGonagle, D.; McDermott, M.R. A proposed classification of the immunological diseases. PLoS Med. 2006, 3, e297. [Google Scholar] [CrossRef]
  21. Choi, J.-H.; Suh, C.-H.; Lee, Y.-M.; Suh, Y.-J.; Lee, S.-K.; Kim, S.-S.; Nahm, D.-H.; Park, H.-S. Serum cytokine profiles in patients with adult onset Still’s disease. J. Rheumatol. 2003, 30, 2422–2427. [Google Scholar] [PubMed]
  22. Liu, B.; Yang, J.X.; Yan, L.; Zhuang, H.; Li, T. Novel HBV recombinants between genotypes B and C in 3′-terminal reverse transcriptase (RT) sequences are associated with enhanced viral DNA load, higher RT point mutation rates and place of birth among Chinese patients. Infect. Genet. Evol. 2018, 57, 26–35. [Google Scholar] [CrossRef]
  23. Peng, Y.; Liu, B.; Hou, J.; Sun, J.; Hao, R.; Xiang, K.; Yan, L.; Zhang, J.; Zhuang, H.; Li, T. Naturally occurring deletions/insertions in HBV core promoter tend to decrease in hepatitis B e antigen-positive chronic hepatitis B patients during antiviral therapy. Antivir. Ther. 2015, 20, 623–632. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, B.M.; Li, N.L.; Wang, R.; Li, X.; Li, Z.A.; Marion, T.N.; Li, K. Key roles for phosphorylation and the coiled-coil domain in TRIM56-madiated positive regulation of TLR3-TRIF-dependent innate immunity. J. Biol. Chem. 2024, 300, 107249. [Google Scholar] [CrossRef]
  25. Shen, Y.; Li, N.L.; Wang, J.; Liu, B.; Lester, S.; Li, K. TRIM56 Is an Essential Component of the TLR3 Antiviral Signaling Pathway. J. Biol. Chem. 2012, 287, 36404–36413. [Google Scholar] [CrossRef]
  26. Wei, D.; Li, N.L.; Zeng, Y.; Liu, B.; Kumthip, K.; Wang, T.T.; Huo, D.; Ingels, J.F.; Lu, L.; Shang, J.; et al. The molecular chaperone GRP78 contributes to Toll-like receptor 3-mediated innate immune ewsponse to hepatitis C virus in hepatocytes. J. Biol. Chem. 2016, 291, 12294–12309. [Google Scholar] [CrossRef]
  27. Park, H.; Bourla, A.B.; Kastner, D.L.; Colbert, R.A.; Siegel, R.M. Lighting the fires within: The cell biology of autoinflammatory diseases. Nat. Rev. Immunol. 2012, 12, 570–580. [Google Scholar] [CrossRef]
  28. Conigliaro, P.; Priori, R.; Bombardieri, M.; Alessandri, C.; Barone, F.; Pitzalis, C.; McInnes, I.B.; Valesini, G. Lymph node IL-18 expression in adult-onset Still’s fdisease. Ann. Rheum. Dis. 2009, 68, 442–443. [Google Scholar] [CrossRef] [PubMed]
  29. Meng, G.; Zhang, F.; Fuss, I.; Kitani, A.; Strober, W. A mutation in the NLRP3 gene causing inflammasone hyperactivation potentiates Th17 cell-dominant immune responses. Immunity 2009, 30, 860–874. [Google Scholar] [CrossRef]
  30. Lee, S.; Cho, Y.; Kim, T.; Park, S.; Park, D.; Jin, H.; Lee, S.; Kee, S.; Kim, N.; Yoo, D.; et al. Natural killer T cell deficiency in active adult-onset Still’s disease: Correlation of deficiency of natural killer T cells with dysfunction of natural killer cells. Arthritis Rheum. 2012, 64, 2868–2877. [Google Scholar] [CrossRef]
  31. Zandman-Goddart, G.; Shoenfeld, Y. Ferritin in autoimmune diseases. Autoimmun. Rev. 2007, 6, 457–463. [Google Scholar] [CrossRef] [PubMed]
  32. Mitrivic, S.; Fautrel, B. New markers for adult-onset Still’s disease. Jt. Bone Spine 2018, 85, 285–293. [Google Scholar] [CrossRef] [PubMed]
  33. Javaux, C.; El-Jammal, T.; Neau, P.-A.; Fournier, N.; Gerfaud-Valentin, M.; Perard, L.; Fouillet-Desjonqueres, M.; Le Scanff, J.; Vignot, E.; Durupt, S.; et al. Detection and Prediction of Macrophage Activation Syndrome in Still’s Disease. J. Clin. Med. 2021, 11, 206. [Google Scholar] [CrossRef] [PubMed]
  34. Triggianese, P.; Vitale, A.; Lopalco, G.; Giardini, H.A.M.; Ciccia, F.; Al-Maghlouth, I.; Ruscitti, P.; Sfikakis, P.P.; Iannone, F.; Antonelli, I.P.d.B.; et al. Clinical and laboratory features associated with macrophage activation syndrome in Still’s disease: Data from the international AIDA Network Still’s Disease Registry. Intern. Emerg. Med. 2023, 18, 2231–2243. [Google Scholar] [CrossRef] [PubMed]
  35. Sola, D.; Smirne, C.; Bruggi, F.; Sbaratta, C.B.; Njata, A.C.T.; Walente, G.; Pavanelli, M.C.; Vitetta, R.; Bellan, M.; De Paoli, L.; et al. Unveiling the Mystery of Adult-Onset Still’s Disease:A Compelling Case Report. Life 2024, 14, 195. [Google Scholar] [CrossRef]
  36. Fautrel, B.; Sibilia, J.; Mariette, X.; Combe, B. Tumor necrosis factor alpha blocking agents in refractory adult Still’s disease: An observational study of 20 cases. Ann. Rheum. Dis. 2005, 64, 262–266. [Google Scholar] [CrossRef]
  37. Ortiz-Sanjuán, F.; Blanco, R.; Riancho-Zarrabeitia, L.; Castaneda, S.; Olivé, A.; Riveros, A.; Velloso-Feijoo, M.L.; Narváez, J.; Jiménez-Moleón, I.; Maiz-Alonso, O.; et al. Efficacy of Anakinra in refractory adult-onset Still’s disease: Multicenter study of 41 patients and literature review. Medicine 2015, 94, e1554. [Google Scholar] [CrossRef]
  38. Kedor, C.; Listing, J.; Zernicke, J.; Weiß, A.; Behrens, F.; Blank, N.; Henes, J.C.; Kekow, J.; Rubbert-Roth, A.; Schulze-Koops, H.; et al. Canakinumab for treatment of adult-onset Still’s disease to achieve reduction of arthritic manifestation (CONSIDER): Phase II, randomised, double-blind, placebo-controlled, multicentre, investigator-initiated trial. Ann. Rheum. Dis. 2020, 79, 1090–1097. [Google Scholar] [CrossRef]
  39. Ilowite, N.T.; Prather, K.; Lokhnygina, Y.; Schanberg, L.E.; Elder, M.; Milojevic, D.; Verbsky, J.W.; Spalding, S.J.; Kimura, Y.; Imundo, L.F.; et al. Randomized, double-blind, placebo-controlled trial of the efficacy and safety of rilonacept in the treatment of systemic juvenile idiopathic arthritis. Arthritis Rheumatol. 2014, 66, 2570–2579. [Google Scholar] [CrossRef]
  40. Ma, Y.; Wu, M.; Zhang, X.; Xia, Q.; Yang, J.; Xu, S.; Pan, F. Efficacy and safety of tocilizumab with inhibition of interleukin-6 in adult-onset Still’s disease: A meta-analysis. Mod. Rheumatol. 2018, 28, 849–857. [Google Scholar] [CrossRef]
  41. Honda, M.; Moriyama, M.; Kondo, M.; Kumakura, S.; Murakawa, Y. Tofacitinib-induced remission in refractory adult-onset Still’s complicated by macrophage activation syndrome. Scand. J. Rheumatol. 2020, 49, 336–338. [Google Scholar] [CrossRef] [PubMed]
  42. Kacar, M.; Fitton, J.; Gough, A.K.; Buch, M.H.; McGonagle, D.G.; Savic, S. Mixed results with baricitinib in biological-resistant adult-onset Still’s disease and undifferentiated systemic autoinflammatory disease. RMD Open 2020, 6, e001246. [Google Scholar] [CrossRef] [PubMed]
  43. Adachi, S.; Takase-Minegishi, K.; Maeda, A.; Nagai, H.; Horita, N.; Yoshimi, R.; Kirino, Y.; Nakajima, H. Risk of macrophage activation syndrome in patients with adult-onset Still’s disease treated with IL-1 and IL-6 inhibitors: A meta-analysis and single center experience. Rheumatol. Ther. 2023, 10, 1623–1636. [Google Scholar] [CrossRef]
  44. Ahmadi-Simab, K.; Lamprecht, P.; Jankowiak, C.; Gross, W.L. Succesful treatment of refractory adult onset Still’s disease with rituximab. Ann. Rheum. Dis. 2006, 65, 1117–1118. [Google Scholar] [CrossRef]
  45. Przybyszewska, W.; Geisler, P.; Kisiel, B.; Raczkiewicz, A.; Elert-Kopeć, S.; Malczuk, E.; Choroś, D.; Tłustochowicz, W. The use of cyclophosphamide in the treatment of Still’s disease—A case report. Pol. Merkur. Lek. 2021, 49, 317–373. [Google Scholar]
  46. Vignes, S.; Wechsler, B.; Amoura, Z.; Papo, T.; Frances, C.; Huong, D.L.; Veyssier, P.; Godeau, P.; Piette, J.C. Intravenous immunoglobulin in adult Still’s disease refractory to non-steroidal anti-inflammatory drugs. Clin. Exp. Rheumatol. 1998, 16, 295–298. [Google Scholar]
  47. Quartuccio, L.; Maset, M.; de Vita, S. Efficacy of abatacept in a refractory case of adult-onset Still’s disease. Clin. Exp. Rheumatol. 2010, 28, 265–267. [Google Scholar]
  48. Kessel, C.; Lippitz, K.; Weinhage, T.; Hinze, C.; Wittkowski, H.; Holzinger, D.; Fall, N.; Grom, A.A.; Gruen, N.; Foell, D. Proinflammatory Cytokine Environments Can Drive Interleukin-17 Overexpression by γ/δ T Cells in Systemic Juvenile Idiopathic Arthritis. Arthritis Rheumatol. 2017, 69, 1480–1494. [Google Scholar] [CrossRef]
  49. Shakoory, B.; Geerlinks, A.; Wilejto, M.; Kernan, K.; Hines, M.; Romano, M.; Piskin, D.; Ravelli, A.; Sinha, R.; Aletaha, D.; et al. The 2022 EULAR/ACR points to consider at the early stages of diagnosis and management of suspected haemophagocytic lymphohistiocytosis/macrophage activation syndrome (HLH/MAS). Ann. Rheum. Dis. 2023, 82, 1271–1285. [Google Scholar] [CrossRef]
  50. De Benedetti, F.; Grom, A.A.; Brogan, P.A.; Bracaglia, C.; Pardeo, M.; Marucci, G.; Eleftheriou, D.; Papadopoulou, C.; Schulert, G.S.; Quartier, P.; et al. Efficacy and safety of emapalumab in macrophage activation syndrome. Ann. Rheum. Dis. 2023, 82, 857–865. [Google Scholar] [CrossRef]
Table 1. Classification criteria for AOSD and the revised definition of the International League of Associations for Rheumatology (ILAR) diagnostic criteria for SJIA.
Table 1. Classification criteria for AOSD and the revised definition of the International League of Associations for Rheumatology (ILAR) diagnostic criteria for SJIA.
1987 Cush Criteria [8]1992 Yamaguchi Criteria [9]2002 Fautrel Criteria [10]2004 ILAR Criteria [11]
2 points each:
Quotidian fever > 39 °C
Evanescent rash
White blood cells (WBC) > 12,000/μL and erythrocytes sedimentation rate (ESR) > 40 mm/1h
Negative antinuclear antibody (ANA) and rheumatoid factor (RF)
Carpal ankylosis
Major criteria:
Fever > 39 °C, intermittent, one week or longer
Arthralgia or arthritis ≥ 2 weeks
Characteristic rash
WBC > 10,000/μL with > 80% granulocytes
Major criteria:
Spiking fever ≥ 39 °C
Arthralgia
Transient erythema
Pharyngitis
≥80% granulocytes
Glycosylated ferritin ≤ 20%
Major criteria:
Arthritis in at least one joint
Fever > 2 weeks, daily for at least 3 days
1 point each:
Onset age > 35 years
Arthritis
Sore throat
Reticuloendothelial system involvement or liver function test (LFT) abnormality
Serositis
Cervical or tarsal ankylosis
Minor criteria:
Sore throat
Lymphadenopathy
Hepatomegaly or splenomegaly
Abnormal liver function tests
Negative ANA and RF
Minor criteria:
Maculopapular rash
Leucocytes ≥ 10,000/μL
Minor criteria:
Evanescent erythematous rash
Generalized lymph node enlargement
Hepatomegaly
Splenomegaly
Serositis
Exclusion criteria:
None
Exclusion criteria:
Infections, malignancy, other rheumatic diseases that mimic AOSD
Exclusion criteria:
None
Exclusion criteria:
Other forms of JIA must be excluded
Algorithm:
Probable AOSD:
10 points during 12 weeks of observation
Definite AOSD:
10 points with 6 months observation
Algorithm:
5 criteria: at least 2 major ones AND no exclusion criteria
Algorithm:
4 major criteria OR 3 major with 2 minor ones
Algorithm:
All major criteria AND at least 1 minor criterion
Sensitivity:
Not applicable
Specificity:
Not applicable
Sensitivity:
96.2%
Specificity:
92.1%
Sensitivity:
80.6%
Specificity:
98.5%
Sensitivity:
Not applicable
Specificity:
Not applicable
Table 2. Frequency of symptoms, joint involvement, and laboratory test abnormalities in AOSD.
Table 2. Frequency of symptoms, joint involvement, and laboratory test abnormalities in AOSD.
SymptomsFrequency in %Involved JointsFrequency in %Laboratory Test AbnormalitiesFrequency in %
Fever
Arthralgia/arthritis
Skin rashes
Myalgia
Sore throat
Lymphadenopathy
Hepatomegaly
Splenomegaly
Abdominal pain
Pleuritis or pericarditis
85–100
73–95
68–81
44–65
53–62
31–61
7–22
5–67
14–18
20–25
Knee
Wrist
Ankle
Proximal interphalangeal (PIP)
Elbow
Shoulder
MCP
Metatarsophalangeal (MTP)
Hip
Distal interphalangeal (DIP)
Temporomandi-bular
84
74
57
47

50
43
34
19

14
18

8
Increased ESR and CRP
Leukocytosis > 10,000/mm3
Neutrophil polymorphonuc-lear count ≥ 80%
Anemia ≤ 10g/100 mL
Thrombocytosis > 400,000/mm3
Abnormal liver function results
Increased ferritin concentration
Reduced glycosylated ferritin concentration
ANA absence
RF absence
99
89–94
88–93

50–75
62

43–76

69–93
80–90

92
93
Table 3. Hscore for the diagnosis of HLH [18].
Table 3. Hscore for the diagnosis of HLH [18].
VariableVariableNumber of Points
Temperature<38.4 °C
38.4–39.4 °C
>39.4 °C
0
33
49
OrganomegalyNone
Hepatomegaly or splenomegaly
Hepatomegaly and splenomegaly
0
23
38
CytopeniaOne lineage
Two lineages
Three lineages
0
24
34
Triglycerides (mmol/L)<1.5
1.5–4.0
>4.0
0
44
64
Fibrinogen (g/L)>2.5
≤2.5
0
30
Ferritin (ng/mL)<2000
2000–6000
>6000
0
35
50
Serum aspartate aminotransferase (IU/L)<30
≥30
0
19
Hemophagocytosis on bone marrow aspirateNo
Yes
0
35
Known immunosuppressionNo
Yes
0
18
Table 4. Cases of Still’s disease.
Table 4. Cases of Still’s disease.
No. of CaseSex/AgeCriteria of DiagnosisComplicationsTreatment
1F/34
18-week HBD
Intermittent fever > 39 °C, arthralgia, typical rash, leucocytes > 28,000 μg/L, granulocytes 84.5%, sore throat, ferritin 2217 ng/mLMAS/HLH
Th5 compressive fracture
GCs, IVIG, cyclosporin A (CsA), methotrexate (MTX), Etanercept, Tocilizumab
2M/24Arthritis (wrists, knee), sore throat, maculopapular rash, leucocytes > 18,300 μg/L, 84.4% granulocytes, ferritin 2006 ng/mL GCs, MTX
3F/30Intermittent fever > 39 °C, arthralgia, typical rash, leucocytes > 20,100 μg/L, granulocytes 83%, sore throat, ferritin 2067 ng/mL GCs, MTX
4F/31Intermittent fever > 39 °C, arthralgia, typical rash, leucocytes > 15,700 μg/L, granulocytes 82% sore throat, ferritin 4405 ng/mL GCs, MTX
5M/41Intermittent fever > 39 °C, arthralgia, typical rash, leucocytes > 13,870 μg/L, granulocytes 83%, ferritin 16,016 ng/mL GCs
6F/28Intermittent fever > 39 °C, arthritis (wrists), typical rash, lymphadenopathy, leucocytes > 14,720 μg/L, granulocytes 83.3% sore throat, ferritin 799 ng/mL GCs, MTX
7M/19Intermittent fever > 39 °C, arthritis (wrists), typical rash, leucocytes > 20,980 μg/L, granulocytes 89.4% sore throat, ferritin 4405 ng/mLMAS/HLHGCs, CyC, intravenous immunoglobulin (IVIG), Tocilizumab
8F/26,
22-week HBD
Intermittent fever > 39 °C, arthritis (wrists, knees), typical rash, sore throat, pericarditis, pleuritis, leucocytes > 21,030 μg/L, granulocytes 88.6%, ferritin 21,140 ng/mL, hepatomegaly.
Previously, at age 10, the patient suffered one incident of MAS/HLH
MAS/HLHGCs, CsA, IVIG, etanercept, MTX
9F/30Intermittent fever > 39 °C, arthritis (wrists, feet), slight urticarial rash, lymphadenopathy, hepatosplenomegaly, leucocytes > 8670 μg/L, ferritin 885 ng/mLMAS/HLHGCs
Table 5. Cases with AOSD who developed MAS/HLH syndrome.
Table 5. Cases with AOSD who developed MAS/HLH syndrome.
Clinical and Laboratory FeatureCase 1
Female/34 Years
18 Week HBD
Case 7
Male/19 Years
Case 8
Female/26 Years
22 Week HBD
Case 9
Female/30 Years
Persistent fever>39.5 °C>39.5 °C>39.5 °C>39.5 °C
Hemorrhagic rashAbsentPresentAbsentAbsent
HepatosplenomegalyPresentPresentOnly hepatomegalyPresent
Erythrocytes (μg/L)26,000 Within normal range28,500Within normal range
Lymphocytes (μg/L)Within normal rangeWithin normal rangeWithin normal range2.46
Thrombocytes (μg/L)106,000 55,000159,000Within normal range
Ferritin (ng/mL)6217 79,18736,9851062
AspAT (IU/L)2423 13845366
Fibrinogen (g/L)1.980.884.01.67
Triglycerides (mg/dL)215.2335.4275138.5
Table 6. The value of Hscore patients with MAS/HLH syndrome.
Table 6. The value of Hscore patients with MAS/HLH syndrome.
Parameters of HscoreCase 1
Female/34 Years
18 Week HBD
Case 7
Male/19 Years
Case 8
Female/26 Years
22 Week HBD
Case 9
Female/30 Years
Immunosuppression18181818
Temperature49494949
Organomegaly38382338
Cytopenias240240
Ferritin5050500
Fibrinogen3030030
Triglycerides4464440
Transaminase19191919
Total score272268227154
Treatment GCs, CyS, IVIG, tocilizumabGCs, CyS, IVIG, tocilizumabGCs, CyS, IVIG, plasmapheresisGCs
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wisłowska, M. Adult-Onset Still’s Disease (AOSD)—On the Basis of Own Cases. Biomedicines 2024, 12, 2067. https://doi.org/10.3390/biomedicines12092067

AMA Style

Wisłowska M. Adult-Onset Still’s Disease (AOSD)—On the Basis of Own Cases. Biomedicines. 2024; 12(9):2067. https://doi.org/10.3390/biomedicines12092067

Chicago/Turabian Style

Wisłowska, Małgorzata. 2024. "Adult-Onset Still’s Disease (AOSD)—On the Basis of Own Cases" Biomedicines 12, no. 9: 2067. https://doi.org/10.3390/biomedicines12092067

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop