1
|
Depciuch J, Sieminska I, Jakubczyk P, Klebowski B, Ptak K, Szymońska I, Kwinta P, Siedlar M, Kęsik JJ, Parlinska-Wojtan M, Baran J. Detection of serum composition in pediatric inflammatory multisystem syndrome associated with SARS-CoV-2 and the response for the treatment by FTIR. Sci Rep 2025; 15:4669. [PMID: 39920293 PMCID: PMC11805954 DOI: 10.1038/s41598-025-88976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Pediatric Inflammatory Multisystem Syndrome (PIMS-TS), associated with SARS-CoV-2 infection, is a severe complication after COVID-19 in children. It is caused by the immune reaction to SARS-CoV-2, and usually appears three to six weeks after the infection. Unfortunately, PIMS causes non-specific symptoms, which makes its diagnosis and treatment difficult. In this paper, we propose Fourier Transform InfraRed spectrometry (FTIR) to identify chemical changes in blood serum of children induced by PIMS and caused by subsequent treatment of the syndrome. The results suggest that although the Principal Component Analysis (PCA) of FTIR data did not allow for differentiation of healthy children and children with PIMS before and after the treatment, the implementation of Support Vector Machine (SVM) showed that the accuracy of the FTIR region between 800 cm- 1 and 1800 cm- 1 in PIMS detection is as high as 92% with a sensitivity of 100%. The difference in the chemical compositions of sera from the control group and the children after the treatment was detected in 54%, indicating that the treatment was effective. Indeed, the obtained medical data clearly showed a decrease of C-reactive protein (CRP) and Procalcitonin (PCT) concentration in serum after the treatment. The decision tree showed that peak 1455 cm- 1 could be used as a potential FTIR PIMS marker. Importantly, FTIR data correlates well with medical parameters, however the correlation differs with respect to the groups before and after the treatment.
Collapse
Affiliation(s)
- Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, Kraków, 31-342, Poland.
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodźki 1, Lublin, 20-093, Poland.
| | - Izabela Sieminska
- Institute of Veterinary Sciences, University Center of Veterinary Medicine JU-AU, University of Agriculture in Krakow, Mickiewicza Av. 24/28, Krakow, 30-059, Poland
- Department of Clinical Immunology, Chair of Clinical Immunology and Transplantology, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
| | - Pawel Jakubczyk
- Institute of Physics, University of Rzeszow, al. Rejtana 16c, Rzeszów, 35-959, Poland
| | - Bartosz Klebowski
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, Kraków, 31-342, Poland
| | - Katarzyna Ptak
- Department of Pediatrics, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
| | - Izabela Szymońska
- Department of Pediatrics, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
| | - Przemko Kwinta
- Department of Pediatrics, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
- Department of Pediatrics, University Children's Hospital, Wielicka Str. 265, Krakow, 30-663, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Chair of Clinical Immunology and Transplantology, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
- Department of Clinical Immunology, University Children's Hospital, Wielicka Str. 265, Krakow, 30-663, Poland
| | - Jan Jakub Kęsik
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Magdalena Parlinska-Wojtan
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, Kraków, 31-342, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Chair of Clinical Immunology and Transplantology, Jagiellonian University Medical College, Wielicka Str. 265, Krakow, 30-663, Poland
- Department of Clinical Immunology, University Children's Hospital, Wielicka Str. 265, Krakow, 30-663, Poland
| |
Collapse
|
2
|
Goel AR, Yalcindag A. An Update on Multi-System Inflammatory Syndrome in Children. Curr Rheumatol Rep 2025; 27:16. [PMID: 39883190 DOI: 10.1007/s11926-025-01182-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE To summarize the latest research on the epidemiology, pathogenesis, diagnosis, and treatment of multisystem inflammatory syndrome in children (MIS-C). RECENT FINDINGS The epidemiology of MIS-C has been dynamic since its initial description. The pathogenesis remains poorly understood. Case definitions of MIS-C have evolved over time, and practice patterns for treating MIS-C are variable with generally positive long-term outcomes yet persistent changes noted. MIS-C has become less prevalent and less severe over time, yet racial and ethnic disparities persist, and vaccination against COVID-19 is highly effective in preventing this disease. The link between acute infection and subsequent inflammation is not well understood, with growing evidence describing its immunologic signature. Newer case definitions require excluding other inflammatory conditions, including Kawasaki Disease (KD), before diagnosing MIS-C. Corticosteroid monotherapy may be non-inferior to IVIg alone or combination IVIg plus corticosteroids for initial treatment, distinguishing the approaches to MIS-C and KD. A wide range of biologic therapies have been employed for rescue therapy with general success and no clear benefit of one over another. Despite reports of a high rate of coronary artery abnormality regression and resolution of heart failure, long-term studies suggest persistent changes to cardiac function. The long-term effects of MIS-C continue to be active areas of research.
Collapse
Affiliation(s)
- Anurag Ratan Goel
- Department of Internal Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ali Yalcindag
- Division of Rheumatology, Department of Pediatrics, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI, 02903, USA.
| |
Collapse
|
3
|
Hu C, Yan X, Song H, Dong Q, Yi C, Li J, Lv X. Establishment and validation of a nomogram for coronary artery lesions in children with Kawasaki disease. Front Cardiovasc Med 2025; 11:1522473. [PMID: 39877016 PMCID: PMC11772273 DOI: 10.3389/fcvm.2024.1522473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background The nomogram is a powerful and robust tool in disease risk prediction that summarizes complex variables into a visual model that is interpretable with a quantified risk probability. In the current study, a nomogram was developed to predict the occurrence of coronary artery lesions (CALs) among patients with Kawasaki disease (KD). This is especially valuable in the early identification of the risk of CALs, which will lead to proper diagnosis and treatment to reduce their associated complications. Methods Retrospective clinical data of 677 children diagnosed with KD who were treated in the Children's Hospital Affiliated with Shandong University were analyzed. All the participants were divided into the CAL group and no CAL group according to their coronary echocardiography results. Least absolute shrinkage and selection operator (LASSO) regression was applied for the identification of the most informative predictors of CAL. Based on this, a nomogram was developed for accurate risk estimation. Results The data were divided into a training set and a validation set. Receiver operating characteristic analysis, calibration curves, and decision curve analysis all supported the high accuracy and clinical utility of this model. LASSO regression highlighted five key predictors: sodium, hemoglobin, platelet count, D-dimer, and cystatin C. A nomogram based on these predictors was established and successfully validated in both datasets. In the training set, the AUC was 0.819 and in the validation set it was 0.844. The C-index of the calibration curve in the training set was 0.820, while in the validation set it was 0.844. In the decision curve analysis, the predictive benefit of the model was greater than zero when the threshold probability was below 95% in the training set and below 92% in the validation set. Conclusion The predictive factors identified through the LASSO regression approach and the development of the nomogram are important contributions in this respect. This model had a high predictive accuracy and reliability for identifying high-risk children in the very early stage of disease with remarkable precision, laying the foundation for personalized treatment strategies and targeted treatment and providing a strong scientific basis for precise therapeutic intervention.
Collapse
Affiliation(s)
- Chong Hu
- Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, China
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, China
| | - Xiao Yan
- Department of Hematology, Qingdao Municipal Hospital, Qingdao, China
| | - Henglian Song
- Department of Hematology and Oncology, Jining No. 2 People’s Hospital, Jining, Shandong, China
| | - Qin Dong
- Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, China
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, China
| | - Changying Yi
- Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, China
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, China
| | - Jianzhi Li
- Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, China
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, China
| | - Xin Lv
- Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, China
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, China
| |
Collapse
|
4
|
Atici AE, Noval Rivas M, Arditi M. The Central Role of Interleukin-1 Signalling in the Pathogenesis of Kawasaki Disease Vasculitis: Path to Translation. Can J Cardiol 2024; 40:2305-2320. [PMID: 39084253 PMCID: PMC11646188 DOI: 10.1016/j.cjca.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Kawasaki disease (KD) manifests as an acute febrile condition and systemic vasculitis, the etiology of which remains elusive. Primarily affecting children under 5 years of age, if untreated KD can lead to a significant risk of coronary artery aneurysms and subsequent long-term cardiovascular sequelae, including myocardial ischemia and myocardial infarction. Intravenous immunoglobulin therapy mitigates the risk of aneurysm formation, but a subset of patients exhibit resistance to this treatment, increasing the susceptibility of coronary artery lesions. Furthermore, the absence of a KD-specific diagnostic test or biomarkers complicates early detection and appropriate treatment. Experimental murine models of KD vasculitis have substantially improved our understanding of the disease pathophysiology, revealing the key roles of the NLRP3 inflammasome and interleukin-1 (IL-1) signalling pathway. This review aims to delineate the pathophysiologic findings of KD while summarising the findings for the emerging key role of IL-1β in its pathogenesis, derived from both human data and experimental murine models, and the translational potential of these findings for anti-IL-1 therapies for children with KD.
Collapse
Affiliation(s)
- Asli Ekin Atici
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Magali Noval Rivas
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Moshe Arditi
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, California, USA; Infectious and Immunologic Diseases Research Center, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
5
|
Namin SS, Zhu YP, Croker BA, Tan Z. Turning Neutrophil Cell Death Deadly in the Context of Hypertensive Vascular Disease. Can J Cardiol 2024; 40:2356-2367. [PMID: 39326672 DOI: 10.1016/j.cjca.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/24/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Hypertensive vascular disease (HVD) is a major health burden globally and is a comorbidity commonly associated with other metabolic diseases. Many factors are associated with HVD including obesity, diabetes, smoking, chronic kidney disease, and sterile inflammation. Increasing evidence points to neutrophils as an important component of the chronic inflammatory response in HVD. Neutrophils are abundant in the circulation and can respond rapidly upon stimulation to deploy an armament of antimicrobial effector functions. One of the outcomes of neutrophil activation is the generation of neutrophil extracellular traps (NETs), a regulated extrusion of chromatin and proteases. Although neutrophils and NETs are well described as components of the innate immune response to infection, recent evidence implicates them in HVD. Endothelial cell activation can trigger neutrophil adhesion, activation, and production of NETs promoting vascular dysfunction, vessel remodelling, and loss of resistance. The regulated release of NETs can be controlled by the pore-forming activities of distinct cell death pathways. The best characterized pathways in this context are apoptosis, pyroptosis, and necroptosis. In this review, we discuss how inflammatory cell death signalling and NET formation contribute to hypertensive disease. We also examine novel therapeutic approaches to limit NET production and their future potential as therapeutic drugs for cardiovascular disorders.
Collapse
Affiliation(s)
- Sahand Salari Namin
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Yanfang Peipei Zhu
- Department of Biochemistry and Molecular Biology, Immunology Center of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ben A Croker
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Zhehao Tan
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
6
|
Day-Lewis M, Son MBF, Lo MS. Kawasaki disease: contemporary perspectives. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:781-792. [PMID: 39299749 DOI: 10.1016/s2352-4642(24)00169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 09/22/2024]
Abstract
Kawasaki disease is a paediatric vasculitis that presents with fever, rash, conjunctivitis, mucositis, lymphadenopathy, and extremity changes, and primarily affects children younger than 5 years. Coronary artery aneurysms are observed in approximately 20% of patients without treatment. Giant coronary artery aneurysms are rare but can result in substantial morbidity and mortality due to the risk of thrombosis, stenosis, and myocardial infarction. Infants younger than 6 months and children with coronary artery abnormalities are at highest risk for the development of large or giant coronary artery aneurysms, necessitating swift identification and aggressive treatment. The children at high risk for coronary artery aneurysms warrant primary intensification therapy; however, what the most optimal adjunct therapy might be to reduce their risk is unclear and large-scale international trials are needed. Kawasaki disease is a clinical diagnosis that shares many features with other common febrile illnesses, including multisystem inflammatory syndrome in children. Identifying biomarkers that can distinguish Kawasaki disease from similar conditions and predict coronary artery aneurysm risk are needed to aid timely diagnosis, guide management, and improve patient outcomes.
Collapse
Affiliation(s)
- Megan Day-Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Mary Beth F Son
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Mindy S Lo
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Ganapathy T, Villagracia KEM, Kuditini SS, Sosa Hilario S. Multisystem Inflammatory Syndrome in Children: A Case Report. Cureus 2024; 16:e72303. [PMID: 39463906 PMCID: PMC11503855 DOI: 10.7759/cureus.72303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 10/29/2024] Open
Abstract
This case report delves into the case of a patient in the Dominican Republic with multisystem inflammatory syndrome in children (MIS-C). MIS-C is a rare, hyper-inflammatory condition that develops in children as a delayed response to a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, typically appearing with general markers of inflammation such as erythema and, in severe cases, cardiorespiratory symptoms. The three-year-old patient discussed in this report presented with signs of inflammation, such as erythema, rashes, and chapped skin, and reported experiencing diarrhea, vomiting, and fever for multiple days. Notably, a concurrent parasitic presence was found in the patient's fecal sample, and antibiotics were heavily used throughout the course of treatment. We explain the merits and drawbacks behind using antibiotic therapy for MIS-C and suggest steps that clinicians and researchers can take in order to minimize the potential misuse of antibiotics. Specifically, we identify that prioritizing tests for concurrent infections or illnesses is imperative in treating MIS-C patients, and we conclude by stating that using blood cultures and coprological examinations in tandem is an effective strategy for this purpose.
Collapse
|
8
|
Li XQ, Xue P, Zheng YM, Liu S. Kawasaki Disease Presenting with Fever and Jaundice: Case Report. Br J Hosp Med (Lond) 2024; 85:1-12. [PMID: 39347672 DOI: 10.12968/hmed.2024.0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Kawasaki disease (KD), which is also known as cutaneous mucosal lymph node syndrome, is an acute, self-limiting, necrotizing vasculitis with unclear cause that primarily affects small- and medium-sized blood vessels and most commonly affects children aged 6 months to 5 years. Currently, diagnosis is based primarily on typical clinical symptoms. Approximately 15%-20% of patients are highly suspected of having KD; however, they do not match the diagnostic criteria for typical KD, which is referred to as incomplete Kawasaki disease (IKD), and this has become a major challenge in the diagnosis and treatment of KD. We describe a case of a 7-year-old boy who had a fever and jaundice as his initial symptoms. After a series of clinical laboratory and imaging examinations and marked improvement of symptoms after treatment with intravenous immunoglobulin (IVIG), IKD was considered as the diagnosis. When children present with jaundice and fever, physicians should consider KD as a possible diagnosis to ensure early detection and treatment of the disease.
Collapse
Affiliation(s)
- Xiao-Qin Li
- Department of Pediatrics, Taiyuan Children's Hospital & Taiyuan Maternity and Child Health Care Hospital, Taiyuan, Shanxi, China
| | - Ping Xue
- Department of Pediatrics, Taiyuan Children's Hospital & Taiyuan Maternity and Child Health Care Hospital, Taiyuan, Shanxi, China
| | - Yan-Mei Zheng
- Department of Pediatrics, Taiyuan Children's Hospital & Taiyuan Maternity and Child Health Care Hospital, Taiyuan, Shanxi, China
| | - Shuo Liu
- Department of Pediatrics, Taiyuan Children's Hospital & Taiyuan Maternity and Child Health Care Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
9
|
Noval Rivas M, Kocatürk B, Franklin BS, Arditi M. Platelets in Kawasaki disease: mediators of vascular inflammation. Nat Rev Rheumatol 2024; 20:459-472. [PMID: 38886559 DOI: 10.1038/s41584-024-01119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
Kawasaki disease, a systemic vasculitis that affects young children and can result in coronary artery aneurysms, is the leading cause of acquired heart disease among children. A hallmark of Kawasaki disease is increased blood platelet counts and platelet activation, which is associated with an increased risk of developing resistance to intravenous immunoglobulin and coronary artery aneurysms. Platelets and their releasate, including granules, microparticles, microRNAs and transcription factors, can influence innate immunity, enhance inflammation and contribute to vascular remodelling. Growing evidence indicates that platelets also interact with immune and non-immune cells to regulate inflammation. Platelets boost NLRP3 inflammasome activation and IL-1β production by human immune cells by releasing soluble mediators. Activated platelets form aggregates with leukocytes, such as monocytes and neutrophils, enhancing numerous functions of these cells and promoting thrombosis and inflammation. Leukocyte-platelet aggregates are increased in children with Kawasaki disease during the acute phase of the disease and can be used as biomarkers for disease severity. Here we review the role of platelets in Kawasaki disease and discuss progress in understanding the immune-effector role of platelets in amplifying inflammation related to Kawasaki disease vasculitis and therapeutic strategies targeting platelets or platelet-derived molecules.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Begüm Kocatürk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Reiter A, Verweyen EL, Queste E, Fuehner S, Jakob A, Masjosthusmann K, Hinze C, Wittkowski H, Foell D, Meinzer U, Melki I, Kessel C. Proteomic mapping identifies serum marker signatures associated with MIS-C specific hyperinflammation and cardiovascular manifestation. Clin Immunol 2024; 264:110237. [PMID: 38723855 DOI: 10.1016/j.clim.2024.110237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) shares several clinical and immunological features with Kawasaki Disease (KD) and pediatric hyperinflammation, but the immuno-phenotypic overlap among these clinical mimics is still incompletely understood. Here we analyzed serum samples from treatment-naïve patients with MIS-C (n = 31) and KD (n = 11), pediatric hyperinflammation (n = 13) and healthy controls (HC, n = 10) by proximity extension assay (PEA) to profile 184 blood biomarkers. Collectively, immunophenotypic overlap between MIS-C and hyperinflammation exceeds overlap with KD. Overexpression of IL-17A in MIS-C and KD could best separate these conditions from hyperinflammatory conditions, while those were hallmarked by overabundance of adenosin deaminase and IL-18. Depletion in serum TNF-related subfamily member 9 (TNFRSF9) and apoptosis inducing ligand (TRAIL) linked with cardiovascular manifestations and myocarditis in MIS-C. Altogether, our analysis highlights important differences in molecular marker signatures also across different MIS-C and KD cohorts and suggests several previously unidentified molecular associations in context of cardiovascular inflammation.
Collapse
Affiliation(s)
- Andrea Reiter
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Emely L Verweyen
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Emmanuelle Queste
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Université Paris Cité, INSERM, Centre de Recherche sur l'inflammation UMR 1149, Paris, France
| | - Sabrina Fuehner
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - André Jakob
- Division of Pediatric Cardiology and Pediatric Intensive Care, Ludwig-Maximilians University, Munich, Germany
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Claas Hinze
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Helmut Wittkowski
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Ulrich Meinzer
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Université Paris Cité, INSERM, Centre de Recherche sur l'inflammation UMR 1149, Paris, France
| | - Isabelle Melki
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Paediatrics, Rheumatology and Paediatric Internal Medicine, Children's Hospital, F-33000 Bordeaux, France; Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Université Paris Cité, Inserm UMR 1163, F-75015 Paris, France
| | - Christoph Kessel
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany.
| |
Collapse
|
11
|
Lombardi Pereira AP, Aubuchon E, Moreira DP, Lane M, Carvalho TT, Mesquita TRR, Lee Y, Crother TR, Porritt RA, Verri WA, Noval Rivas M, Arditi M. Long-term cardiovascular inflammation and fibrosis in a murine model of vasculitis induced by Lactobacillus casei cell wall extract. Front Immunol 2024; 15:1411979. [PMID: 38989288 PMCID: PMC11234797 DOI: 10.3389/fimmu.2024.1411979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Background Kawasaki disease (KD), an acute febrile illness and systemic vasculitis, is the leading cause of acquired heart disease in children in industrialized countries. KD leads to the development of coronary artery aneurysms (CAA) in affected children, which may persist for months and even years after the acute phase of the disease. There is an unmet need to characterize the immune and pathological mechanisms of the long-term complications of KD. Methods We examined cardiovascular complications in the Lactobacillus casei cell wall extract (LCWE) mouse model of KD-like vasculitis over 4 months. The long-term immune, pathological, and functional changes occurring in cardiovascular lesions were characterized by histological examination, flow cytometric analysis, immunofluorescent staining of cardiovascular tissues, and transthoracic echocardiogram. Results CAA and abdominal aorta dilations were detected up to 16 weeks following LCWE injection and initiation of acute vasculitis. We observed alterations in the composition of circulating immune cell profiles, such as increased monocyte frequencies in the acute phase of the disease and higher counts of neutrophils. We determined a positive correlation between circulating neutrophil and inflammatory monocyte counts and the severity of cardiovascular lesions early after LCWE injection. LCWE-induced KD-like vasculitis was associated with myocarditis and myocardial dysfunction, characterized by diminished ejection fraction and left ventricular remodeling, which worsened over time. We observed extensive fibrosis within the inflamed cardiac tissue early in the disease and myocardial fibrosis in later stages. Conclusion Our findings indicate that increased circulating neutrophil counts in the acute phase are a reliable predictor of cardiovascular inflammation severity in LCWE-injected mice. Furthermore, long-term cardiac complications stemming from inflammatory cell infiltrations in the aortic root and coronary arteries, myocardial dysfunction, and myocardial fibrosis persist over long periods and are still detected up to 16 weeks after LCWE injection.
Collapse
Affiliation(s)
- Ana Paula Lombardi Pereira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Emily Aubuchon
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debbie P. Moreira
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Malcolm Lane
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rebecca A. Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
12
|
Jandus C, Jandus P. Effects of Intravenous Immunoglobulins on Human Innate Immune Cells: Collegium Internationale Allergologicum Update 2024. Int Arch Allergy Immunol 2024; 185:975-996. [PMID: 38852585 DOI: 10.1159/000539069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/22/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Intravenous immunoglobulin (IVIg) has been used for almost 40 years in the treatment of autoimmune and systemic inflammatory diseases. Numerous cells are involved in the innate immune response, including monocytes/macrophages, neutrophils, dendritic cells, mast cells, basophils, eosinophils, natural killer cells, and innate lymphoid cells. Many studies have investigated the mechanisms by which IVIg down-modulates inflammatory and autoimmune processes of innate immune cells. However, questions remain regarding the precise mechanism of action in autoimmune or inflammatory conditions. The aim of this work was to review the immunomodulatory effect of IVIg on only human innate immune cells. A narrative review approach was chosen to summarize key evidence on the immunomodulatory effects of commercially available and unmodified IVIg on human innate immune cells. SUMMARY Numerous different immunomodulatory effects of IVIg have been reported, with some very different effects depending on the immune cell type and disease. Several limitations of the different studies were identified. Of the 77 studies identified and reviewed, 29 (37.7%) dealt with autoimmune or inflammatory diseases. Otherwise, the immunomodulatory effects of IVIg were studied only in healthy donors using an in vitro experimental approach. Some of the documented effects showed disease-specific effects, such as in Kawasaki disease. Various methodological limitations have also been identified that may reduce the validity of some studies. KEY MESSAGE As further insights have been gained into the various inflammatory cascades activated in immunological diseases, interesting insights have also been gained into the mechanism of action of IVIg. We are still far from discovering all the immunomodulatory mechanisms of IVIg.
Collapse
Affiliation(s)
- Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Centre in Onco-Haematology (CRTOH), Geneva, Switzerland
| | - Peter Jandus
- Division of Immunology and Allergology, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
13
|
Abbas Q, Ali H, Amjad F, Hussain MZH, Rahman AR, Khan MH, Padhani ZA, Abbas F, Imam D, Alikhan Z, Belgaumi SM, Mohsin S, Sattar F, Siddiqui A, Lassi ZS, Das JK. Clinical presentation, diagnosis and management of multisystem inflammatory syndrome in children (MIS-C): a systematic review. BMJ Paediatr Open 2024; 8:e002344. [PMID: 38844384 PMCID: PMC11163633 DOI: 10.1136/bmjpo-2023-002344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/16/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Knowledge about multisystem inflammatory syndrome in children (MIS-C) is evolving, and evidence-based standardised diagnostic and management protocols are lacking. Our review aims to summarise the clinical and diagnostic features, management strategies and outcomes of MIS-C and evaluate the variances in disease parameters and outcomes between high-income countries (HIC) and middle-income countries (MIC). METHODS We searched four databases from December 2019 to March 2023. Observational studies with a sample size of 10 or more patients were included. Mean and prevalence ratios for various variables were pooled by random effects model using R. A mixed generalised linear model was employed to account for the heterogeneity, and publication bias was assessed via funnel and Doi plots. The primary outcome was pooled mean mortality among patients with MIS-C. Subgroup analysis was conducted based on the income status of the country of study. RESULTS A total of 120 studies (20 881 cases) were included in the review. The most common clinical presentations were fever (99%; 95% CI 99.6% to 100%), gastrointestinal symptoms (76.7%; 95% CI 73.1% to 79.9%) and dermatological symptoms (63.3%; 95% CI 58.7% to 67.7%). Laboratory investigations suggested raised inflammatory, coagulation and cardiac markers. The most common management strategies were intravenous immunoglobulins (87.5%; 95% CI 82.9% to 91%) and steroids (74.7%; 95% CI 68.7% to 79.9%). Around 53.1% (95% CI 47.3% to 58.9%) required paediatric intensive care unit admissions, and overall mortality was 3.9% (95% CI 2.7% to 5.6%). Patients in MIC were younger, had a higher frequency of respiratory distress and evidence of cardiac dysfunction, with a longer hospital and intensive care unit stay and had a higher mortality rate than patients in HIC. CONCLUSION MIS-C is a severe multisystem disease with better mortality outcomes in HIC as compared with MIC. The findings emphasise the need for standardised protocols and further research to optimise patient care and address disparities between HIC and MIC. PROSPERO REGISTRATION NUMBER CRD42020195823.
Collapse
Affiliation(s)
- Qalab Abbas
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Haider Ali
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Fatima Amjad
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | | | - Abdu R Rahman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Maryam Hameed Khan
- Institute for Global Health and Development, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zahra A Padhani
- School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Fatima Abbas
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Danyal Imam
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zuviya Alikhan
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Sameer M Belgaumi
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Shazia Mohsin
- Department of Pediatric cardiology, Division of cardiothoracic sciences, Sindh institute of Urology and Transplantation (SIUT), Karachi, Sind, Pakistan
| | - Faiza Sattar
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Arsalan Siddiqui
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zohra S Lassi
- School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jai K Das
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
- Institute for Global Health and Development, The Aga Khan University, Karachi, Sind, Pakistan
| |
Collapse
|
14
|
Xu T, Chen T, Fang H, Shen X, Shen X, Tang Z, Zhao J. Human Umbilical Cord Mesenchymal Stem Cells Repair Endothelial Injury and Dysfunction by Regulating NLRP3 to Inhibit Endothelial Cell Pyroptosis in Kawasaki Disease. Inflammation 2024; 47:483-502. [PMID: 37948033 DOI: 10.1007/s10753-023-01921-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Vascular endothelial inflammation and endothelial dysfunction are the main causes of endothelial injury in Kawasaki disease (KD). Human umbilical cord-derived mesenchymal stem cells (Huc-MSCs) have multiple functions in immune regulation. This study examined whether Huc-MSCs inhibited endothelial inflammation and improved endothelial function in KD through constructing cell and in vivo animal KD vasculitis models. The pyroptosis factor NOD-like receptor protein 3 (NLRP3) was involved in the inflammatory process in the acute phase of KD. After tail vein injection of Huc-MSCs, inflammatory cell infiltration and the expression of pyroptosis-related proteins in the LCWE-induced KD mouse vasculitis model were significantly reduced. In vitro, NLRP3-dependent pyroptosis successfully induced human umbilical vein endothelial cell (HUVEC) damage. Huc-MSCs effectively increased the abilities of impaired HUVECs to proliferate, migrate, invade, and form vessel-like tubes, while inhibiting their apoptosis, suggesting that Huc-MSCs can reduce inflammation and improve vascular endothelial function by inhibiting the NLRP3-dependent pyroptosis pathway in KD, providing a possibility and novel target for KD endothelial injury and dysfunction.
Collapse
Affiliation(s)
- Ting Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Tao Chen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Hao Fang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xiwei Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xianjuan Shen
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Jianmei Zhao
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
15
|
Tong T, Jin YH, Wang M, Gong FQ. Treatment of multisystem inflammatory syndrome in children. World J Pediatr 2024; 20:325-339. [PMID: 38509432 DOI: 10.1007/s12519-024-00798-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/29/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Multisystem inflammatory syndrome in children (MIS-C), a relatively uncommon but severe pediatric complication, is associated with coronavirus disease 2019 (COVID-19). A variety of treatment approaches, including intravenous immunoglobulins (IVIGs), glucocorticoids (GCs) and biologic agents, such as anakinra and infliximab, have been described for the management of COVID-19-related MIS-C. Anticoagulant therapy is also important. However, a well-developed treatment system has not been established, and many issues remain controversial. Several recently published articles related to the treatment of MIS-C have been released. Hence, in this review, we identified relevant articles published recently and summarized the treatment of MIS-C more comprehensively and systematically. DATA SOURCES We reviewed the literature on the treatment of MIS-C through 20 September 2023. The PubMed/Medline, Web of Science, EMBASE, and Cochrane Library databases were searched with the combination of the terms "multisystem inflammatory syndrome", "MIS-C", "PIMS-TS", "therapy", "treatment", "drug", "IVIG", "GCs", "intravenous immunoglobulin", "corticosteroids", "biological agent", and "aspirin". RESULTS The severity of MIS-C varies, and different treatment schemes should be used according to the specific condition. Ongoing research and data collection are vital to better understand the pathophysiology and optimal management of MIS-C. CONCLUSIONS MIS-C is a disease involving multiple systems and has great heterogeneity. With the accumulation of additional experience, we have garnered fresh insights into its treatment strategies. However, there remains a critical need for greater standardization in treatment protocols, alongside the pressing necessity for more robust and meticulously conducted studies to deepen our understanding of these protocols. Supplementary file1 (MP4 208044 kb).
Collapse
Affiliation(s)
- Tong Tong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China
| | - Yi-Hua Jin
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China
| | - Min Wang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China
| | - Fang-Qi Gong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China.
| |
Collapse
|
16
|
Lee S, Erdem G, Yasuhara J. Multisystem inflammatory syndrome in children associated with COVID-19: from pathophysiology to clinical management and outcomes. Minerva Pediatr (Torino) 2024; 76:268-280. [PMID: 37284807 DOI: 10.23736/s2724-5276.23.07205-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Multisystem inflammatory syndrome in children (MIS-C), also known as pediatric inflammatory multisystem syndrome (PIMS), is a new postinfectious illness associated with COVID-19, affecting children after SARS-CoV-2 exposure. The hallmarks of this disorder are hyperinflammation and multisystem involvement, with gastrointestinal, cardiac, mucocutaneous, and hematologic disturbances seen most commonly. Cardiovascular involvement includes cardiogenic shock, ventricular dysfunction, coronary artery abnormalities, and myocarditis. Now entering the fourth year of the pandemic, clinicians have gained some familiarity with the clinical presentation, initial diagnosis, cardiac evaluation, and treatment of MIS-C. This has led to an updated definition from the Centers for Disease Control and Prevention in the USA driven by increased experience and clinical expertise. Furthermore, the available evidence established expert consensus treatment recommendations supporting a combination of immunoglobulin and steroids. However, the pathophysiology of the disorder and answers to what causes this remain under investigation. Fortunately, long-term outcomes continue to look promising, although continued follow-up is still needed. Recently, COVID-19 mRNA vaccination is reported to be associated with reduced risk of MIS-C, while further studies are warranted to understand the impact of COVID-19 vaccines on MIS-C. We review the findings and current literature on MIS-C, including pathophysiology, clinical features, evaluation, management, and medium- to long-term follow-up outcomes.
Collapse
Affiliation(s)
- Simon Lee
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Guliz Erdem
- Division of Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA
| | - Jun Yasuhara
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA -
- Center for Cardiovascular Research, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Cardiology, Royal Children's Hospital, Parkville, Australia
| |
Collapse
|
17
|
Hosaka S, Imagawa K, Yano Y, Lin L, Shiono J, Takahashi-Igari M, Hara H, Hayashi D, Imai H, Morita A, Fukushima H, Takada H. The CXCL10-CXCR3 axis plays an important role in Kawasaki disease. Clin Exp Immunol 2024; 216:104-111. [PMID: 37952216 PMCID: PMC10929692 DOI: 10.1093/cei/uxad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/25/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023] Open
Abstract
The precise pathogenesis of Kawasaki disease remains unknown. In an attempt to elucidate the pathogenesis of KD through the analysis of acquired immunity, we comprehensively examined the immunophenotypic changes in immune cells such as lymphocytes and monocytes along with various cytokines, focusing on differences between pre- and post- treatment samples. We found high levels of CXCL9 and CXCL10 chemokines that decreased with treatment, which coincided with a post-treatment expansion of Th1 cells expressing CXCR3. Our results show that the CXCL10-CXCR3 axis plays an important role in the pathogenesis of KD.
Collapse
Affiliation(s)
- Sho Hosaka
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
| | - Kazuo Imagawa
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba City, Japan
| | - Yusuke Yano
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
- Department of Pediatric Cardiology, Ibaraki Children’s Hospital, Mito City, Japan
| | - Lisheng Lin
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba City, Japan
- Department of Pediatric Cardiology, Ibaraki Children’s Hospital, Mito City, Japan
| | - Junko Shiono
- Department of Pediatric Cardiology, Ibaraki Children’s Hospital, Mito City, Japan
| | | | - Hideki Hara
- Department of Pediatrics, Tsukuba Medical Center Hospital, Tsukuba City, Japan
| | - Daisuke Hayashi
- Department of Pediatrics, Tsukuba Medical Center Hospital, Tsukuba City, Japan
| | - Hironori Imai
- Department of Pediatrics, Tsukuba Medical Center Hospital, Tsukuba City, Japan
| | - Atsushi Morita
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
| | - Hiroko Fukushima
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba City, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba City, Japan
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba City, Japan
| |
Collapse
|
18
|
Tsoukas P, Yeung RSM. Kawasaki Disease-Associated Cytokine Storm Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:365-383. [PMID: 39117827 DOI: 10.1007/978-3-031-59815-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Kawasaki disease (KD) is a hyperinflammatory syndrome manifesting as an acute systemic vasculitis characterized by fever, nonsuppurative conjunctival injection, rash, oral mucositis, extremity changes, and cervical lymphadenopathy. KD predominantly affects young children and shares clinical features and immunobiology with other hyperinflammation syndromes including systemic juvenile idiopathic arthritis (sJIA) and multisystem inflammatory syndrome in children (MIS-C). Cytokine storm syndrome (CSS) is an acute complication in ~2% of KD patients; however, the incidence is likely underestimated as many clinical and laboratory features of both diseases overlap. CSS should be entertained when a child with KD is unresponsive to IVIG therapy with recalcitrant fever. Early recognition and prompt institution of immunomodulatory treatment can substantially reduce the mortality and morbidity of CSS in KD. Given the known pathogenetic role of IL-1β in both syndromes, the early use of IL-1 blockers in refractory KD with CSS deserves consideration.
Collapse
Affiliation(s)
- Paul Tsoukas
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rae S M Yeung
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
19
|
Hinze CH, Foell D, Kessel C. Treatment of systemic juvenile idiopathic arthritis. Nat Rev Rheumatol 2023; 19:778-789. [PMID: 37923864 DOI: 10.1038/s41584-023-01042-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 11/06/2023]
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is an inflammatory disease with hallmarks of severe systemic inflammation, which can be accompanied by arthritis. Contemporary scientific insights set this paediatric disorder on a continuum with its counterpart, adult-onset Still disease (AOSD). Patients with sJIA are prone to complications, including life-threatening hyperinflammation (macrophage activation syndrome (sJIA-MAS)) and sJIA-associated lung disease (sJIA-LD). Meanwhile, the treatment arsenal in sJIA has expanded markedly. State-of-the-art therapeutic approaches include biologic agents that target the IL-1 and IL-6 pathways. Beyond these, a range of novel agents are on the horizon, some of them already being used on a compassionate use basis, including JAK inhibitors and biologic agents that target IL-18, IFNγ, or IL-1β and IL-18 simultaneously. However, sJIA, sJIA-MAS and sJIA-LD still pose challenging conundrums to rheumatologists treating paediatric and adult patients worldwide. Although national and international consensus treatment plans exist for the treatment of 'classic' sJIA, the treatment approaches for early sJIA without arthritis, and for refractory or complicated sJIA, are not well defined. Therefore, in this Review we outline current approaches for the treatment of sJIA and provide an outlook on knowledge gaps.
Collapse
Affiliation(s)
- Claas H Hinze
- Department of Paediatric Rheumatology and Immunology, Münster University Hospital, Münster, Germany
| | - Dirk Foell
- Department of Paediatric Rheumatology and Immunology, Münster University Hospital, Münster, Germany.
| | - Christoph Kessel
- Department of Paediatric Rheumatology and Immunology, Münster University Hospital, Münster, Germany
| |
Collapse
|
20
|
Beltran JVB, Lin FP, Chang CL, Ko TM. Single-Cell Meta-Analysis of Neutrophil Activation in Kawasaki Disease and Multisystem Inflammatory Syndrome in Children Reveals Potential Shared Immunological Drivers. Circulation 2023; 148:1778-1796. [PMID: 37905415 DOI: 10.1161/circulationaha.123.064734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/27/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Kawasaki disease (KD) and multisystem inflammatory syndrome in children (MIS-C) share similar clinical manifestations, including cardiovascular complications, suggesting similar underlying immunopathogenic processes. Aberrant neutrophil activation may play a crucial role in the shared pathologies of KD and MIS-C; however, the associated pathogenic mechanisms and molecular drivers remain unknown. METHODS We performed a single-cell meta-analysis of neutrophil activation with 103 pediatric single-cell transcriptomic peripheral blood mononuclear cell data across 9 cohorts, including healthy controls, KD, MIS-C, compared with dengue virus infection, juvenile idiopathic arthritis, and pediatric celiac disease. We used a series of computational analyses to investigate the shared neutrophil transcriptional programs of KD and MIS-C that are linked to systemic damage and cardiac pathologies, and suggested Food and Drug Administration-approved drugs to consider as KD and MIS-C treatment. RESULTS We meta-analyzed 521 950 high-quality cells. We found that blood signatures associated with risks of cardiovascular events are enriched in neutrophils of KD and MIS-C. We revealed the expansion of CD177+ neutrophils harboring hyperactivated effector functions in both KD and MIS-C, but not in healthy controls or in other viral-, inflammatory-, or immune-related pediatric diseases. KD and MIS-C CD177+ neutrophils had highly similar transcriptomes, marked by conserved signatures and pathways related to molecular damage. We found the induction of a shared neutrophil expression program, potentially regulated by SPI1 (Spi-1 proto-oncogene), which confers enhanced effector functions, especially neutrophil degranulation. CD177 and shared neutrophil expression program expressions were associated with acute stages and attenuated during KD intravenous immunoglobulin treatment and MIS-C recovery. Network analysis identified hub genes that correlated with the high activation of CD177+ neutrophils. Disease-gene association analysis revealed that the KD and MIS-C CD177+ neutrophils' shared expression program was associated with the development of coronary and myocardial disorders. Last, we identified and validated TSPO (translocator protein) and S100A12 (S100 calcium-binding protein A12) as main molecular targets, for which the Food and Drug Administration-approved drugs methotrexate, zaleplon, metronidazole, lorazepam, clonazepam, temazepam, and zolpidem, among others, are primary candidates for drug repurposing. CONCLUSIONS Our findings indicate that CD177+ neutrophils may exert systemic pathological damage contributing to the shared morbidities in KD and MIS-C. We uncovered potential regulatory drivers of CD177+ neutrophil hyperactivation and pathogenicity that may be targeted as a single therapeutic strategy for either KD or MIS-C.
Collapse
Affiliation(s)
- Jan Vincent B Beltran
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan (J.V.B.B., T.-M.K.)
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (J.V.B.B., T.-M.K.)
| | - Fang-Ping Lin
- Department of Biological Sciences and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (F.-P.L., C.-L.C., T.-M.K.)
| | - Chaw-Liang Chang
- Department of Biological Sciences and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (F.-P.L., C.-L.C., T.-M.K.)
- Department of Pediatrics, Cathay General Hospital, Hsinchu, Taiwan (C.-L.C.)
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan (C.-L.C.)
| | - Tai-Ming Ko
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan (J.V.B.B., T.-M.K.)
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (J.V.B.B., T.-M.K.)
- Department of Biological Sciences and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (F.-P.L., C.-L.C., T.-M.K.)
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu, Taiwan (T.-M.K.)
- School of Pharmacy, College of Pharmacy, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan (T.-M.K.)
| |
Collapse
|
21
|
Conti F, Moratti M, Leonardi L, Catelli A, Bortolamedi E, Filice E, Fetta A, Fabi M, Facchini E, Cantarini ME, Miniaci A, Cordelli DM, Lanari M, Pession A, Zama D. Anti-Inflammatory and Immunomodulatory Effect of High-Dose Immunoglobulins in Children: From Approved Indications to Off-Label Use. Cells 2023; 12:2417. [PMID: 37830631 PMCID: PMC10572613 DOI: 10.3390/cells12192417] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/23/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND The large-scale utilization of immunoglobulins in patients with inborn errors of immunity (IEIs) since 1952 prompted the discovery of their key role at high doses as immunomodulatory and anti-inflammatory therapy, in the treatment of IEI-related immune dysregulation disorders, according to labelled and off-label indications. Recent years have been dominated by a progressive imbalance between the gradual but constant increase in the use of immunoglobulins and their availability, exacerbated by the SARS-CoV-2 pandemic. OBJECTIVES To provide pragmatic indications for a need-based application of high-dose immunoglobulins in the pediatric context. SOURCES A literature search was performed using PubMed, from inception until 1st August 2023, including the following keywords: anti-inflammatory; children; high dose gammaglobulin; high dose immunoglobulin; immune dysregulation; immunomodulation; immunomodulatory; inflammation; intravenous gammaglobulin; intravenous immunoglobulin; off-label; pediatric; subcutaneous gammaglobulin; subcutaneous immunoglobulin. All article types were considered. IMPLICATIONS In the light of the current imbalance between gammaglobulins' demand and availability, this review advocates the urgency of a more conscious utilization of this medical product, giving indications about benefits, risks, cost-effectiveness, and administration routes of high-dose immunoglobulins in children with hematologic, neurologic, and inflammatory immune dysregulation disorders, prompting further research towards a responsible employment of gammaglobulins and improving the therapeutical decisional process.
Collapse
Affiliation(s)
- Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (A.M.); (A.P.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
| | - Mattia Moratti
- Specialty School of Paediatrics, University of Bologna, 40138 Bologna, Italy; (A.C.); (E.B.)
| | - Lucia Leonardi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| | - Arianna Catelli
- Specialty School of Paediatrics, University of Bologna, 40138 Bologna, Italy; (A.C.); (E.B.)
| | - Elisa Bortolamedi
- Specialty School of Paediatrics, University of Bologna, 40138 Bologna, Italy; (A.C.); (E.B.)
| | - Emanuele Filice
- Department of Pediatrics, Maggiore Hospital, 40133 Bologna, Italy;
| | - Anna Fetta
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria dell’Età Pediatrica, 40139 Bologna, Italy
| | - Marianna Fabi
- Paediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Elena Facchini
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (E.F.); (M.E.C.)
| | - Maria Elena Cantarini
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (E.F.); (M.E.C.)
| | - Angela Miniaci
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (A.M.); (A.P.)
| | - Duccio Maria Cordelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria dell’Età Pediatrica, 40139 Bologna, Italy
| | - Marcello Lanari
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
- Paediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (A.M.); (A.P.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
| | - Daniele Zama
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (A.F.); (D.M.C.); (M.L.); (D.Z.)
- Paediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
22
|
Chang JC, Young CC, Muscal E, Sexson Tejtel SK, Newhams MM, Kucukak S, Crandall H, Maddux AB, Rowan CM, Halasa NB, Harvey HA, Hobbs CV, Hall MW, Kong M, Aguiar CL, Schuster JE, Fitzgerald JC, Singh AR, Wellnitz K, Nofziger RA, Cvijanovich NZ, Mack EH, Schwarz AJ, Heidemann SM, Newburger JW, Zambrano LD, Campbell AP, Patel MM, Randolph AG, Son MBF. Variation in Early Anakinra Use and Short-Term Outcomes in Multisystem Inflammatory Syndrome in Children. Arthritis Rheumatol 2023; 75:1466-1476. [PMID: 36908050 PMCID: PMC10495537 DOI: 10.1002/art.42495] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
OBJECTIVE Evidence regarding effectiveness of interleukin-1 receptor antagonism in multisystem inflammatory syndrome in children (MIS-C) is lacking. We characterized variation in initial treatment with anakinra and evaluated cardiovascular outcomes associated with adding anakinra to standard initial therapy. METHODS We conducted a retrospective cohort study of MIS-C cases in a US surveillance registry from November 2020 to December 2021. Day 0 was the first calendar day of immunomodulatory treatment. Factors associated with initial anakinra use (days 0-1) were identified. We compared cases in patients ages 2-20 years receiving intravenous immunoglobulin (IVIG) and glucocorticoids versus anakinra plus IVIG and/or glucocorticoids on days 0-1, using inverse probability weighting to balance disease severity. Primary outcomes were vasopressor requirement on day 3 and impaired left ventricular ejection fraction on days 3-4. The secondary outcome was 50% reduction in C-reactive protein on day 3. RESULTS Among 1,516 MIS-C cases at 44 sites, 193 (13%) patients received anakinra alone or with other immunomodulators as initial treatment (range 0-74% by site). Site accounted for 59% of residual variance in anakinra use. After balancing disease severity, initial treatment with anakinra plus IVIG and/or glucocorticoids (n = 121) versus IVIG plus glucocorticoids (n = 389) was not associated with significant differences in vasopressor requirement (25.6% versus 20.1%, respectively; risk ratio [RR] 1.27 [95% confidence interval (95% CI) 0.88-1.84]), ventricular dysfunction (33.7% versus 25.7%, respectively; RR 1.31 [95% CI 0.98-1.75]), or C-reactive protein reduction. CONCLUSION We identified substantial variation in initial anakinra use in a real-world population of children with MIS-C, but no average short-term improvement in cardiovascular outcomes associated with early addition of anakinra to IVIG and/or glucocorticoids compared to IVIG and glucocorticoids alone.
Collapse
Affiliation(s)
- Joyce C Chang
- Division of Immunology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Cameron C Young
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Eyal Muscal
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Sara K Sexson Tejtel
- Division of Pediatric Cardiology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Fetal Center, Houston, Texas
| | - Margaret M Newhams
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Suden Kucukak
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Hillary Crandall
- Division of Pediatric Critical Care, Department of Pediatrics, University of Utah and Primary Children's Hospital, Salt Lake City, Utah
| | - Aline B Maddux
- Department of Pediatrics, Section of Critical Care Medicine, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora
| | - Courtney M Rowan
- Division of Pediatric Critical Care Medicine and Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children, Indianapolis
| | - Natasha B Halasa
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Helen A Harvey
- Department of Critical Care Medicine, Rady Children's Hospital-San Diego, San Diego, California
| | - Charlotte V Hobbs
- Division of Infectious Disease, Department of Pediatrics, University of Mississippi Medical Center, Jackson
| | - Mark W Hall
- Division of Critical Care Medicine, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - Michele Kong
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Alabama at Birmingham
| | - Cassyanne L Aguiar
- Department of Pediatric Rheumatology, Children's Hospital of The King's Daughters, Eastern Virginia Medical School, Norfolk
| | - Jennifer E Schuster
- Division of Pediatric Infectious Disease, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, Missouri
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Aalok R Singh
- Pediatric Critical Care Division, Maria Fareri Children's Hospital at Westchester Medical Center and New York Medical College, Valhalla, New York
| | - Kari Wellnitz
- Division of Pediatric Critical Care, Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City
| | - Ryan A Nofziger
- Division of Critical Care Medicine, Akron Children's Hospital, Akron, Ohio
| | - Natalie Z Cvijanovich
- Division of Critical Care Medicine, UCSF Benioff Children's Hospital Oakland, Oakland, California
| | - Elizabeth H Mack
- Division of Pediatric Critical Care Medicine, Medical University of South Carolina, Charleston
| | - Adam J Schwarz
- Division of Critical Care Medicine, Children's Hospital Orange County, Orange, California
| | - Sabrina M Heidemann
- Division of Pediatric Critical Care Medicine, Children's Hospital of Michigan, Central Michigan University, Detroit
| | - Jane W Newburger
- Department of Cardiology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, and Departments of Pediatrics and Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Mary Beth F Son
- Division of Immunology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Basaran O, Batu ED, Kaya Akca U, Atalay E, Kasap Cuceoglu M, Sener S, Balık Z, Karabulut E, Kesici S, Karagoz T, Ozsurekci Y, Bilginer Y, Cengiz AB, Ozen S. The Effect of Biologics in the Treatment of Multisystem Inflammatory Syndrome in Children (Mis-C): A Single-Center Propensity-Score-Matched Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1045. [PMID: 37371276 DOI: 10.3390/children10061045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a serious condition characterized by excessive inflammation that can arise as a complication of SARS-CoV-2 infection in children. While our understanding of COVID-19 and MIS-C has been advancing, there is still uncertainty regarding the optimal treatment for MIS-C. In this study, we aimed to compare the clinical and laboratory outcomes of MIS-C patients treated with IVIG plus corticosteroids (CS) to those treated with IVIG plus CS and an additional biologic drug. We used the propensity score (PS)-matching method to assess the relationships between initial treatment and outcomes. The primary outcome was a left ventricular ejection fraction of less than 55% on day 2 or beyond and/or the requirement of inotrope support on day 2 or beyond. We included 79 MIS-C patients (median age 8.51 years, 33 boys) followed in our center. Among them, 50 children (25 in each group) were allocated to the PS-matched cohort sample. The primary outcome was observed in none of the patients in the IVIG and CS group, while it occurred in eight patients in the IVIG plus CS and biologic group (p = 0.04). MIS-C is a disorder that may progress rapidly and calls for extensive care. For definitive recommendations, further studies, including randomized control trials, are required.
Collapse
Affiliation(s)
- Ozge Basaran
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Ezgi Deniz Batu
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Ummusen Kaya Akca
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Erdal Atalay
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Muserref Kasap Cuceoglu
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Seher Sener
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Zeynep Balık
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Erdem Karabulut
- Department of Biostatistics, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Selman Kesici
- Pediatric Intensive Care Medicine, Life Support Center, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Tevfik Karagoz
- Department of Pediatric Cardiology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Yasemin Ozsurekci
- Department of Pediatric Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Yelda Bilginer
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Ali Bulent Cengiz
- Department of Pediatric Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| |
Collapse
|
24
|
Bayry J, Ahmed EA, Toscano-Rivero D, Vonniessen N, Genest G, Cohen CG, Dembele M, Kaveri SV, Mazer BD. Intravenous Immunoglobulin: Mechanism of Action in Autoimmune and Inflammatory Conditions. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1688-1697. [PMID: 37062358 DOI: 10.1016/j.jaip.2023.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/18/2023]
Abstract
Intravenous immunoglobulin (IVIG) is the mainstay of therapy for humoral immune deficiencies and numerous inflammatory disorders. Although the use of IVIG may be supplanted by several targeted therapies to cytokines, the ability of polyclonal normal IgG to act as an effector molecule as well as a regulatory molecule is a clear example of the polyfunctionality of IVIG. This article will address the mechanism of action of IVIG in a number of important conditions that are otherwise resistant to treatment. In this commentary, we will highlight mechanistic studies that shed light on the action of IVIG. This will be approached by identifying effects that are both common and disease-specific, targeting actions that have been demonstrated on cells and processes that represent both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France; Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, India.
| | - Eisha A Ahmed
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Diana Toscano-Rivero
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Nicholas Vonniessen
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Genevieve Genest
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Casey G Cohen
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Marieme Dembele
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Bruce D Mazer
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Ba H, Zhang L, Peng H, He X, Lin Y, Li X, Li S, Zhu L, Qin Y, Zhang X, Wang Y. Identification of Hub Biomarkers and Immune and Inflammation Pathways Contributing to Kawasaki Disease Progression with RT-qPCR Verification. J Immunol Res 2023; 2023:1774260. [PMID: 39670237 PMCID: PMC11637630 DOI: 10.1155/2023/1774260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/21/2022] [Accepted: 03/18/2023] [Indexed: 05/14/2024] Open
Abstract
Background Kawasaki disease (KD) is characterized by a disordered inflammation response of unknown etiology. Immune cells are closely associated with its onset, although the immune-related genes' expression and possibly involved immune regulatory mechanisms are little known. This study aims to identify KD-implicated significant immune- and inflammation-related biomarkers and pathways and their association with immune cell infiltration. Patients and Methods. Gene microarray data were collected from the Gene Expression Omnibus database. Differential expression analysis, weighted gene coexpression network analysis (WGCNA), least absolute shrinkage and selection operator (LASSO) regression, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were used to find KD hub markers. GSEA was used to assess the infiltration by 28 immune cell types and their connections to essential gene markers. Receiver operating characteristic (ROC) curves were used to examine hub markers' diagnostic effectiveness. Finally, hub genes' expressions were validated in Chinese KD patients by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Results One hundred and fifty-one unique genes were found. Among 10 coexpression modules at WGCNA, one hub module exhibited the strongest association with KD. Thirty-six overlapping genes were identified. Six hub genes were potential biomarkers according to LASSO analysis. Immune infiltration revealed connections among activated and effector memory CD4+ T cells, neutrophils, activated dendritic cells, and macrophages. The six hub genes' diagnostic value was shown by ROC curve analysis. Hub genes were enriched in immunological and inflammatory pathways. RT-qPCR verification results of FCGR1B (P < 0.001), GPR84 (P < 0.001), KREMEN1 (P < 0.001), LRG1 (P < 0.001), and TDRD9 (P < 0.001) upregulated expression in Chinese KD patients are consistent with our database analysis. Conclusion Neutrophils, macrophages, and activated dendritic cells are strongly linked to KD pathophysiology. Through immune-related signaling pathways, hub genes such as FCGR1B, GPR84, KREMEN1, LRG1, and TDRD9 may be implicated in KD advancement.
Collapse
Affiliation(s)
- Hongjun Ba
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Lili Zhang
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Huimin Peng
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Xiufang He
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Yuese Lin
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Xuandi Li
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Shujuan Li
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Ling Zhu
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Youzhen Qin
- Department of Pediatric Cardiology, Heart Center, First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Road 2, Guangzhou 510080, China
| | - Xing Zhang
- Department of Cardiology, Kunming Children's Hospital, 288 Qianxing Road, Xishan District, Kunming 650034, Yunnan, China
| | - Yao Wang
- Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| |
Collapse
|
26
|
Jain S, He F, Brown K, Burns JC, Tremoulet AH. Multisystem Inflammatory Syndrome therapies in children (MISTIC): A randomized trial. Contemp Clin Trials Commun 2023; 32:101060. [PMID: 36694613 PMCID: PMC9852262 DOI: 10.1016/j.conctc.2023.101060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 01/14/2023] [Indexed: 01/22/2023] Open
Abstract
Background Multisystem Inflammatory Syndrome in Children (MIS-C), which occurs 2-6 weeks after initial exposure to SARS-CoV-2, was first identified in early 2020 when patients presented with fever and significant inflammation, often requiring management in the intensive care unit. To date, there has been no clinical trial to determine the most effective treatment. This study compares anti-inflammatory treatments that were selected based on current treatments for Kawasaki disease, a coronary artery vasculitis that shares many clinical features with MIS-C. Methods This randomized, comparative effectiveness trial of children with MIS-C uses the small N Sequential Multiple Assignment Randomized Trial (snSMART) design for rare diseases to compare multiple therapies within an individual. Study participants were treated first with intravenous immunoglobulin (IVIG), and if needed, subjects were then randomized to one of three additional treatments (steroids, anakinra, or infliximab). Participants were re-randomized to remaining treatments if they did not demonstrate clinical improvement. Conclusion This trial continues to enroll eligible participants to determine the most effective therapies in addition to IVIG and best order in which to use them to treat MIS-C. Trial Registration NCT04898231.
Collapse
Affiliation(s)
- Sonia Jain
- Biostatistics Research Center, Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, USA
| | - Feng He
- Biostatistics Research Center, Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, USA
| | - Kiana Brown
- Department of Pediatrics, UCSD School of Medicine/Rady Children's Hospital San Diego, 9500 Gilman Dr, Mail Code 0641, La Jolla, CA, 92093-061, USA
| | - Jane C. Burns
- Department of Pediatrics, UCSD School of Medicine/Rady Children's Hospital San Diego, 9500 Gilman Dr, Mail Code 0641, La Jolla, CA, 92093-061, USA
| | - Adriana H. Tremoulet
- Department of Pediatrics, UCSD School of Medicine/Rady Children's Hospital San Diego, 9500 Gilman Dr, Mail Code 0641, La Jolla, CA, 92093-061, USA,Corresponding author. Department of Pediatrics, UCSD School of Medicine, 9500 Gilman Dr, Mail Code 0641, La Jolla, CA, 92093-0641, USA
| |
Collapse
|
27
|
Channon-Wells S, Vito O, McArdle AJ, Seaby EG, Patel H, Shah P, Pazukhina E, Wilson C, Broderick C, D'Souza G, Keren I, Nijman RG, Tremoulet A, Munblit D, Ulloa-Gutierrez R, Carter MJ, Ramnarayan P, De T, Hoggart C, Whittaker E, Herberg JA, Kaforou M, Cunnington AJ, Blyuss O, Levin M. Immunoglobulin, glucocorticoid, or combination therapy for multisystem inflammatory syndrome in children: a propensity-weighted cohort study. THE LANCET. RHEUMATOLOGY 2023; 5:e184-e199. [PMID: 36855438 PMCID: PMC9949883 DOI: 10.1016/s2665-9913(23)00029-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background Multisystem inflammatory syndrome in children (MIS-C), a hyperinflammatory condition associated with SARS-CoV-2 infection, has emerged as a serious illness in children worldwide. Immunoglobulin or glucocorticoids, or both, are currently recommended treatments. Methods The Best Available Treatment Study evaluated immunomodulatory treatments for MIS-C in an international observational cohort. Analysis of the first 614 patients was previously reported. In this propensity-weighted cohort study, clinical and outcome data from children with suspected or proven MIS-C were collected onto a web-based Research Electronic Data Capture database. After excluding neonates and incomplete or duplicate records, inverse probability weighting was used to compare primary treatments with intravenous immunoglobulin, intravenous immunoglobulin plus glucocorticoids, or glucocorticoids alone, using intravenous immunoglobulin as the reference treatment. Primary outcomes were a composite of inotropic or ventilator support from the second day after treatment initiation, or death, and time to improvement on an ordinal clinical severity scale. Secondary outcomes included treatment escalation, clinical deterioration, fever, and coronary artery aneurysm occurrence and resolution. This study is registered with the ISRCTN registry, ISRCTN69546370. Findings We enrolled 2101 children (aged 0 months to 19 years) with clinically diagnosed MIS-C from 39 countries between June 14, 2020, and April 25, 2022, and, following exclusions, 2009 patients were included for analysis (median age 8·0 years [IQR 4·2-11·4], 1191 [59·3%] male and 818 [40·7%] female, and 825 [41·1%] White). 680 (33·8%) patients received primary treatment with intravenous immunoglobulin, 698 (34·7%) with intravenous immunoglobulin plus glucocorticoids, 487 (24·2%) with glucocorticoids alone; 59 (2·9%) patients received other combinations, including biologicals, and 85 (4·2%) patients received no immunomodulators. There were no significant differences between treatments for primary outcomes for the 1586 patients with complete baseline and outcome data that were considered for primary analysis. Adjusted odds ratios for ventilation, inotropic support, or death were 1·09 (95% CI 0·75-1·58; corrected p value=1·00) for intravenous immunoglobulin plus glucocorticoids and 0·93 (0·58-1·47; corrected p value=1·00) for glucocorticoids alone, versus intravenous immunoglobulin alone. Adjusted average hazard ratios for time to improvement were 1·04 (95% CI 0·91-1·20; corrected p value=1·00) for intravenous immunoglobulin plus glucocorticoids, and 0·84 (0·70-1·00; corrected p value=0·22) for glucocorticoids alone, versus intravenous immunoglobulin alone. Treatment escalation was less frequent for intravenous immunoglobulin plus glucocorticoids (OR 0·15 [95% CI 0·11-0·20]; p<0·0001) and glucocorticoids alone (0·68 [0·50-0·93]; p=0·014) versus intravenous immunoglobulin alone. Persistent fever (from day 2 onward) was less common with intravenous immunoglobulin plus glucocorticoids compared with either intravenous immunoglobulin alone (OR 0·50 [95% CI 0·38-0·67]; p<0·0001) or glucocorticoids alone (0·63 [0·45-0·88]; p=0·0058). Coronary artery aneurysm occurrence and resolution did not differ significantly between treatment groups. Interpretation Recovery rates, including occurrence and resolution of coronary artery aneurysms, were similar for primary treatment with intravenous immunoglobulin when compared to glucocorticoids or intravenous immunoglobulin plus glucocorticoids. Initial treatment with glucocorticoids appears to be a safe alternative to immunoglobulin or combined therapy, and might be advantageous in view of the cost and limited availability of intravenous immunoglobulin in many countries. Funding Imperial College London, the European Union's Horizon 2020, Wellcome Trust, the Medical Research Foundation, UK National Institute for Health and Care Research, and National Institutes of Health.
Collapse
Affiliation(s)
- Samuel Channon-Wells
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Ortensia Vito
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Andrew J McArdle
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Eleanor G Seaby
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Genomic Informatics Group, University of Southampton, Southampton, UK
- Translational Genomics Group, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Harsita Patel
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Priyen Shah
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | | | - Clare Wilson
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Claire Broderick
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Giselle D'Souza
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Ilana Keren
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Ruud G Nijman
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatric Emergency Medicine, Division of Medicine, St Mary's hospital-Imperial College NHS Healthcare Trust, London, London, UK
| | - Adriana Tremoulet
- Department of Paediatrics, University of California San Diego-Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Daniel Munblit
- Inflammation, Repair, and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rolando Ulloa-Gutierrez
- Servicio de Infectologia Pediatrica, Hospital Nacional de Niños "Dr. Carlos Sáenz Herrera", Centro de Ciencias Médicas, Caja Costarricense de Seguro Social (CCSS), San José, Costa Rica
- Instituto de Investigación en Ciencias Médicas UCIMED (IICIMED), San José, Costa Rica
- Cátedra de Pediatría, Facultad de Medicina, Universidad de Ciencias Médicas (UCIMED), San José, Costa Rica
| | - Michael J Carter
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, St Thomas' Hospital, London, UK
| | - Padmanabhan Ramnarayan
- Anaesthetics, Pain Medicine, and Intensive Care (APMIC) Division, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Tisham De
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Clive Hoggart
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elizabeth Whittaker
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatrics, Imperial College Healthcare NHS Trust, London, UK
| | - Jethro A Herberg
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatrics, Imperial College Healthcare NHS Trust, London, UK
| | - Myrsini Kaforou
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Aubrey J Cunnington
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatrics, Imperial College Healthcare NHS Trust, London, UK
| | - Oleg Blyuss
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Michael Levin
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Department of Infectious Disease, Section of Paediatric Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Department of Paediatrics, Imperial College Healthcare NHS Trust, London, UK
| | | |
Collapse
|
28
|
Sujana CS, Wadile S, Srinivas CS, Banpurkar AM, Murthy PR, Kulkarni S. Clinical profile and outcomes of multisystem inflammatory syndrome in children associated with COVID-19 virus after surgery for congenital heart defects. Ann Pediatr Cardiol 2023; 16:87-93. [PMID: 37767163 PMCID: PMC10522151 DOI: 10.4103/apc.apc_111_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/14/2022] [Accepted: 02/13/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction To study the clinical presentation, laboratory profile, echocardiographic details, management, and outcomes of children who were diagnosed to have multisystem inflammatory syndrome in children (MIS-C) in the immediate postoperative period after surgery for congenital heart defects (CHDs). Materials and Methods This is a prospective case-control study that included children diagnosed to have MIS-C in the postoperative period based on clinical signs, rise in inflammatory markers, and echocardiographic features of ventricular dysfunction or coronary involvement. Management included intravenous immunoglobulin (IVIG), steroids, and antiplatelet medications in addition to routine postoperative care. Out of the 461 children who underwent surgery for CHD between April 1st, 2021, and November 30th, 2021, 18 children were diagnosed with MIS-C. After the initial routine postoperative course, all 18 children had sudden worsening in clinical and laboratory parameters. Other causes such as bacterial infection were ruled out. All of these children had features of MIS-C with ventricular dilatation and dysfunction, coronary artery involvement, and reactive COVID-19 immunoglobulin G antibody. There was a significant improvement in coronary artery dimensions after IVIG administration (P = 0.001). The involvement of the left main coronary artery was associated with significantly increased length of intensive care unit (ICU) and hospital stay (P = 0.019). Mean ICU and hospital stay was prolonged in the MIS-C group. There were two deaths in this group due to severe left ventricular dysfunction. Conclusions During the pandemic, a proportion of patients undergoing elective cardiac surgery may develop unexpected worsening in clinical status due to MIS-C. A high index of suspicion and prompt treatment with IVIG and steroids may be helpful in improving outcomes.
Collapse
Affiliation(s)
- Chitturi Sai Sujana
- Department of Pediatric Cardiology, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| | - Santosh Wadile
- Department of Pediatric Cardiology, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| | - Chitturi Subrahmanya Srinivas
- Department of Pediatric Cardiology, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| | - Ashishkumar Moreshwar Banpurkar
- Department of Pediatric Cardiology, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| | - Prabhatha Rashmi Murthy
- Department of Pediatric Cardiac Surgery, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| | - Snehal Kulkarni
- Department of Pediatric Cardiology, Sri Sathya Sai Sanjeevani Centre for Child Heart Care and Training in Pediatric Cardiac Skills, Navi Mumbai, Maharashtra, India
| |
Collapse
|
29
|
Long A, Kleiner A, Looney RJ. Immune dysregulation. J Allergy Clin Immunol 2023; 151:70-80. [PMID: 36608984 DOI: 10.1016/j.jaci.2022.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 01/05/2023]
Abstract
The understanding of immune dysregulation in many different diseases continues to grow. There is increasing evidence that altered microbiome and gut barrier dysfunction contribute to systemic inflammation in patients with primary immunodeficiency and in patients with rheumatic disease. Recent research provides insight into the process of induction and maturation of pathogenic age-associated B cells and highlights the role of age-associated B cells in creating tissue inflammation. T follicular regulatory cells are shown to help maintain B-cell tolerance, and therapeutic approaches to increase or promote T follicular regulatory cells may help prevent or decrease immune dysregulation. Meanwhile, novel studies of systemic-onset juvenile idiopathic arthritis reveal a strong HLA association with interstitial lung disease and identify key aspects of the pathogenesis of macrophage activation syndrome. Studies of hyperinflammatory syndromes, including the recently described multisystem inflammatory syndrome of children, characterize similarities and differences in cytokine profiles and T-cell activation. This review focuses on recent advances in the understanding of immune dysregulation and describes potential key factors that may function as biomarkers for disease or targets for therapeutic interventions. Future trials are necessary to address the many remaining questions with regards to pathogenesis, diagnosis, and treatment of autoimmune, inflammatory, and immunodeficiency syndromes.
Collapse
Affiliation(s)
- Andrew Long
- Allergy Immunology Rheumatology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Anatole Kleiner
- Allergy Immunology Rheumatology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - R John Looney
- Allergy Immunology Rheumatology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
30
|
Cai WJ, Ding SG. Retrospective analysis of clinical characteristics and related influencing factors of Kawasaki disease. Medicine (Baltimore) 2022; 101:e32430. [PMID: 36596080 PMCID: PMC9803503 DOI: 10.1097/md.0000000000032430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To compare the clinical characteristics of complete Kawasaki disease (KD) and incomplete Kawasaki disease (IKD), and analyze the possible risk factors of coronary artery lesion (CAL) in KD. The clinical data of 139 children with KD admitted to the hospital from January 2016 to June 2022 were analyzed retrospectively. The differences of clinical characteristics between children with KD and children with IKD were compared. The risk factors of CAL were analyzed using univariate and multivariate logistic regression. Comparison of clinical characteristics between KD and IKD groups, the results showed there was significant difference in terms of conjunctival congestion, rash, lymph node enlargement, hand and foot redness, intravenous immune globulin non reaction and fever time (P < .05). Comparison of laboratory indicators between 2 groups, the results showed that there was significantly difference in the levels of neutrophils (P < .05). 15 cases (15.31%) in KD group were complicated with CAL, and 17 cases (41.46%) in IKD group were complicated with CAL, and the results showed there was a significant difference between the 2 groups (P < .05). Univariate analysis showed that the age and Hb of children with CAL were lower than those of children with nCAL, while C-reactive protein, NT-proBNP, NEUT, and ESR were higher than those of children with nCAL (P < .05). Multivariate analysis showed that the increase of NT-proBNP and the decrease of Hb may be independent risk factors for the occurrence of CAL in children with KD. The clinical manifestation of children with IKD is not typical. Compared with KD children, the fever time is longer and the incidence of CAL is higher. Under-age, increased NT-proBNP and decreased Hb may be independent risk factors for CAL in KD children.
Collapse
Affiliation(s)
- Wen-Juan Cai
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui China
| | - Sheng-Gang Ding
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui China
- * Correspondence: Sheng-Gang Ding, Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China (e-mail: )
| |
Collapse
|
31
|
Long F, Zhu S, Wang Z, Zhang S, He J, Ge X, Ning J. Update on the treatment of multisystem inflammatory syndrome in children associated with COVID-19. Future Virol 2022:10.2217/fvl-2022-0048. [PMID: 36699562 PMCID: PMC9853872 DOI: 10.2217/fvl-2022-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 12/05/2022] [Indexed: 01/21/2023]
Abstract
In late 2019, SARS-CoV-2 was detected in China and spread worldwide. In rare cases, children who were infected with COVID-19 may develop multisystem inflammatory syndrome (MIS-C), which could have higher mortality than COVID-19 itself. Therefore, diagnosis and management are critical for treatment. Specifically, most of the initial treatment options of MIS-C choose intravenous immunoglobulin (IVIG) and steroids as the first-line treatment for patients. Moreover, antagonists of some cytokines are used as potential future therapeutics. Of note, therapeutic plasmapheresis can be used as a treatment for refractory severe MIS-C. We believe that each patient, especially those with comorbid conditions, should have individualized treatment based on both multidisciplinary consensus approach and expert opinion.
Collapse
Affiliation(s)
- Fangyuan Long
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Shiheng Zhu
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Zeguang Wang
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Shungeng Zhang
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Jinlong He
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Xinbin Ge
- 1Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Jun Ning
- 2Department of Paediatrics, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272029, China,Author for correspondence:
| |
Collapse
|
32
|
Rahmel T, Kraft F, Haberl H, Achtzehn U, Brandenburger T, Neb H, Jarczak D, Dietrich M, Magunia H, Zimmer F, Basten J, Landgraf C, Koch T, Zacharowski K, Weigand MA, Rosenberger P, Ullrich R, Meybohm P, Nierhaus A, Kindgen-Milles D, Timmesfeld N, Adamzik M. Intravenous IgM-enriched immunoglobulins in critical COVID-19: a multicentre propensity-weighted cohort study. Crit Care 2022; 26:204. [PMID: 35799196 PMCID: PMC9260992 DOI: 10.1186/s13054-022-04059-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/13/2022] [Indexed: 12/15/2022] Open
Abstract
Background A profound inflammation-mediated lung injury with long-term acute respiratory distress and high mortality is one of the major complications of critical COVID-19. Immunoglobulin M (IgM)-enriched immunoglobulins seem especially capable of mitigating the inflicted inflammatory harm. However, the efficacy of intravenous IgM-enriched preparations in critically ill patients with COVID-19 is largely unclear. Methods In this retrospective multicentric cohort study, 316 patients with laboratory-confirmed critical COVID-19 were treated in ten German and Austrian ICUs between May 2020 and April 2021. The primary outcome was 30-day mortality. Analysis was performed by Cox regression models. Covariate adjustment was performed by propensity score weighting using machine learning-based SuperLearner to overcome the selection bias due to missing randomization. In addition, a subgroup analysis focusing on different treatment regimens and patient characteristics was performed. Results Of the 316 ICU patients, 146 received IgM-enriched immunoglobulins and 170 cases did not, which served as controls. There was no survival difference between the two groups in terms of mortality at 30 days in the overall cohort (HRadj: 0.83; 95% CI: 0.55 to 1.25; p = 0.374). An improved 30-day survival in patients without mechanical ventilation at the time of the immunoglobulin treatment did not reach statistical significance (HRadj: 0.23; 95% CI: 0.05 to 1.08; p = 0.063). Also, no statistically significant difference was observed in the subgroup when a daily dose of ≥ 15 g and a duration of ≥ 3 days of IgM-enriched immunoglobulins were applied (HRadj: 0.65; 95% CI: 0.41 to 1.03; p = 0.068). Conclusions Although we cannot prove a statistically reliable effect of intravenous IgM-enriched immunoglobulins, the confidence intervals may suggest a clinically relevant effect in certain subgroups. Here, an early administration (i.e. in critically ill but not yet mechanically ventilated COVID-19 patients) and a dose of ≥ 15 g for at least 3 days may confer beneficial effects without concerning safety issues. However, these findings need to be validated in upcoming randomized clinical trials. Trial registrationDRKS00025794, German Clinical Trials Register, https://www.drks.de. Registered 6 July 2021. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-022-04059-0.
Collapse
|
33
|
Aymonnier K, Amsler J, Lamprecht P, Salama A, Witko‐Sarsat V. The neutrophil: A key resourceful agent in immune‐mediated vasculitis. Immunol Rev 2022; 314:326-356. [PMID: 36408947 DOI: 10.1111/imr.13170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The term "vasculitis" refers to a group of rare immune-mediated diseases characterized by the dysregulated immune system attacking blood vessels located in any organ of the body, including the skin, lungs, and kidneys. Vasculitides are classified according to the size of the vessel that is affected. Although this observation is not specific to small-, medium-, or large-vessel vasculitides, patients show a high circulating neutrophil-to-lymphocyte ratio, suggesting the direct or indirect involvement of neutrophils in these diseases. As first responders to infection or inflammation, neutrophils release cytotoxic mediators, including reactive oxygen species, proteases, and neutrophil extracellular traps. If not controlled, this dangerous arsenal can injure the vascular system, which acts as the main transport route for neutrophils, thereby amplifying the initial inflammatory stimulus and the recruitment of immune cells. This review highlights the ability of neutrophils to "set the tone" for immune cells and other cells in the vessel wall. Considering both their long-established and newly described roles, we extend their functions far beyond their direct host-damaging potential. We also review the roles of neutrophils in various types of primary vasculitis, including immune complex vasculitis, anti-neutrophil cytoplasmic antibody-associated vasculitis, polyarteritis nodosa, Kawasaki disease, giant cell arteritis, Takayasu arteritis, and Behçet's disease.
Collapse
Affiliation(s)
- Karen Aymonnier
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Jennifer Amsler
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology University of Lübeck Lübeck Germany
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital University College London London UK
| | | |
Collapse
|
34
|
Spracklen TF, Mendelsohn SC, Butters C, Facey-Thomas H, Stander R, Abrahams D, Erasmus M, Baguma R, Day J, Scott C, Zühlke LJ, Kassiotis G, Scriba TJ, Webb K. IL27 gene expression distinguishes multisystem inflammatory syndrome in children from febrile illness in a South African cohort. Front Immunol 2022; 13:992022. [PMID: 36148243 PMCID: PMC9486543 DOI: 10.3389/fimmu.2022.992022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Multisystem inflammatory syndrome in children (MIS-C) is a severe acute inflammatory reaction to SARS-CoV-2 infection in children. There is a lack of data describing differential expression of immune genes in MIS-C compared to healthy children or those with other inflammatory conditions and how expression changes over time. In this study, we investigated expression of immune-related genes in South African MIS-C patients and controls. Methods The cohort included 30 pre-treatment MIS-C cases and 54 healthy non-inflammatory paediatric controls. Other controls included 34 patients with juvenile systemic lupus erythematosus, Kawasaki disease or other inflammatory conditions. Longitudinal post-treatment MIS-C specimens were available at various timepoints. Expression of 80 immune-related genes was determined by real-time quantitative PCR. Results A total of 29 differentially expressed genes were identified in pre-treatment MIS-C compared to healthy controls. Up-regulated genes were found to be overrepresented in innate immune pathways including interleukin-1 processing and pyroptosis. Post-treatment follow-up data were available for up to 1,200 hours after first treatment. All down-regulated genes and 17/18 up-regulated genes resolved to normal levels in the timeframe, and all patients clinically recovered. When comparing MIS-C to other febrile conditions, only IL27 expression could differentiate these two groups with high sensitivity and specificity. Conclusions These data indicate a unique 29-gene signature of MIS-C in South African children. The up-regulation of interleukin-1 and pyroptosis pathway genes highlights the role of the innate immune system in MIS-C. IL-27 is a potent anti-inflammatory and antiviral cytokine that may distinguish MIS-C from other conditions in our setting.
Collapse
Affiliation(s)
- Timothy F. Spracklen
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Simon C. Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Heidi Facey-Thomas
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Raphaella Stander
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Debbie Abrahams
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Mzwandile Erasmus
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Richard Baguma
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Day
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Christiaan Scott
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Liesl J. Zühlke
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - George Kassiotis
- Retroviral Immunology Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, St Mary’s Hospital, Imperial College, London, United Kingdom
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Kate Webb
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Crick African Network, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
35
|
Meza-Contreras AI, Galdos-Bejar MN, Escalante-Kanashiro R. Multisystem Inflammatory Syndrome in Children Associated with COVID-19 Infection: A Comprehensive Review. J Pediatr Intensive Care 2022. [DOI: 10.1055/s-0042-1748903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
AbstractThe Multisystem Inflammatory Syndrome in Children (MIS-C) is a postinfectious syndrome associated with coronavirus disease 2019 (COVID-19) disease in children. The aim of this study is to conduct a thorough review to assist health care professionals in diagnosis and management of this complication of COVID-19 disease in children. A thorough systematic review was conducted through an on-line search based on MIS-C with the primary focus on epidemiology, clinical characteristics, diagnosis, pathophysiology, management, and long-term follow-up. This syndrome is characterized by an exaggerated and uncontrolled release of proinflammatory cytokines involving dysfunction of both innate and adaptive immunity. In this review, a summary of observational studies and case reports was conducted, in which we found that MIS-C generates multiple-organ failure frequently presenting with hemodynamic instability further characterized by Kawasaki-like symptoms (such as persistent high fever, polymorphic rash, and bilateral conjunctivitis) and predominance of gastrointestinal and cardiovascular signs and symptoms. Keys to effective management involve early diagnosis, timely treatment and re-evaluation following hospital discharge. Diagnosis is marked by significant elevation of inflammatory biomarkers, laboratory evidence of COVID-19 infection or history of recent exposure, and absence of any other plausible explanation for the associated signs, symptoms, and presentation. Management includes hemodynamic stabilization, empiric antibiotic therapy (de-escalation if cultures and polymerase chain reaction studies indicate no bacterial co-infection), immunomodulatory therapy (methylprednisolone, intravenous immunoglobulin, anakinra, tocilizumab, siltuximab, Janus kinase inhibitors, tumor necrosis factor-α inhibitors), antivirals (remdesivir), and anticoagulation (acetylsalicylic acid, unfractionated or low-molecular-weight heparin or new oral anticoagulants). In addition, we identified poor prognostic risk factors to include concurrent comorbidities, blood-component consumption and marrow suppression (lymphopenia, thrombocytopenia), depletion of homeostatic components (hypoalbuminemia), and marked evidence of a hyperinflammatory response to include elevated values of ferritin, C-reactive protein, and D-dimer. MIS-C constitutes a postinfectious syndrome characterized by a marked cytokine storm, characterized by fever, bilateral conjunctivitis, and multiple organ dysfunction. Promoting future research and long-term follow-up will be essential for the development of guidelines and recommendations leading to effective identification and management of MIS-C.
Collapse
Affiliation(s)
| | | | - Raffo Escalante-Kanashiro
- Medical School, Universidad Peruana de Ciencias Aplicadas (UPC), Lima, Perú
- Department of Intensive Care Unit - Instituto Nacional de Salud del Niño, Lima, Perú
| |
Collapse
|
36
|
Hsieh LE, Song J, Tremoulet AH, Burns JC, Franco A. Intravenous immunoglobulin induces IgG internalization by tolerogenic myeloid dendritic cells that secrete IL-10 and expand Fc-specific regulatory T cells. Clin Exp Immunol 2022; 208:361-371. [PMID: 35536993 PMCID: PMC9226148 DOI: 10.1093/cei/uxac046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/28/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is used as an immunomodulatory agent in many inflammatory conditions including Multisystem Inflammatory Syndrome-Children (MIS-C) and Kawasaki disease (KD). However, the exact mechanisms underlying its anti-inflammatory action are incompletely characterized. Here, we show that in KD, a pediatric acute vasculitis that affects the coronary arteries, IVIG induces a repertoire of natural Treg that recognize immunodominant peptides in the Fc heavy chain constant region. To address which antigen-presenting cell (APC) populations present Fc peptides to Treg, we studied the uptake of IgG by innate cells in subacute KD patients 2 weeks after IVIG and in children 1.6–14 years after KD. Healthy adults served as controls. IgG at high concentrations was internalized predominantly by two myeloid dendritic cell (DC) lineages, CD14+ cDC2 and ILT-4+ CD4+ tmDC mostly through Fcγ receptor (R) II and to a lesser extent FcγRIII. Following IgG internalization, these two DC lineages secreted IL-10 and presented processed Fc peptides to Treg. The validation of IVIG function in expanding Fc-specific Treg presented by CD14+ cDC2 and ILT-4+ CD4+ tmDC was addressed in a small cohort of patients with MIS-C. Taken together, these results suggest a novel immune regulatory function of IgG in activating tolerogenic innate cells and expanding Treg, which reveals an important anti-inflammatory mechanism of action of IVIG.
Collapse
Affiliation(s)
- Li-En Hsieh
- University of California San Diego, School of Medicine, Department of Pediatrics, La Jolla, CA 92093-0641, USA
| | - Jaeyoon Song
- University of California San Diego, School of Medicine, Department of Pediatrics, La Jolla, CA 92093-0641, USA
| | - Adriana H Tremoulet
- University of California San Diego, School of Medicine, Department of Pediatrics, La Jolla, CA 92093-0641, USA.,Rady Children's Hospital, San Diego, CA 92123, USA
| | - Jane C Burns
- University of California San Diego, School of Medicine, Department of Pediatrics, La Jolla, CA 92093-0641, USA.,Rady Children's Hospital, San Diego, CA 92123, USA
| | - Alessandra Franco
- University of California San Diego, School of Medicine, Department of Pediatrics, La Jolla, CA 92093-0641, USA
| |
Collapse
|
37
|
An Immunological Axis Involving Interleukin 1β and Leucine-Rich-α2-Glycoprotein Reflects Therapeutic Response of Children with Kawasaki Disease: Implications from the KAWAKINRA Trial. J Clin Immunol 2022; 42:1330-1341. [PMID: 35699824 PMCID: PMC9537216 DOI: 10.1007/s10875-022-01301-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/30/2022] [Indexed: 11/20/2022]
Abstract
Purpose A recent phase II open-label study of the interleukin 1 (IL-1) receptor antagonist (IL-1Ra) anakinra in treating IVIG-resistant Kawasaki disease (KD) patients reported promising results. Here, we aimed to characterize the immunological impact of IL-1 blockade in this unique study population. Methods Patients’ and control sera and supernatants of cells (whole blood, neutrophils, coronary artery endothelial cells) stimulated with recombinant IL-1β were analyzed for single or multiple marker (n = 22) expression by ELISA or multiplexed bead array assay. Data were analyzed using unsupervised hierarchical clustering, multiple correlation, and multi-comparison statistics and were compared to retrospective analyses of KD transcriptomics. Results Inflammation in IVIG-resistant KD (n = 16) is hallmarked by over-expression of innate immune mediators (particularly IL-6 > CXCL10 > S100A12 > IL-1Ra). Those as well as levels of immune or endothelial cell activation markers (sICAM-1, sVCAM-1) declined most significantly in course of anakinra treatment. Prior as well as following IL-1R blockade, over-expression of leucine-rich-α2-glycoprotein 1 (LRG1) associated best with remnant inflammatory activity and the necessity to escalate anakinra dosage and separated inflammatory KD patients from sJIA-MAS (n = 13) and MIS-C (n = 4). Protein as well as retrospective gene expression analyses indicated tight association of LRG1 with IL-1β signaling and neutrophilia, while particularly neutrophil stimulation with recombinant IL-1β resulted in concentration-dependent LRG1 release. Conclusion Our study identifies LRG1 as known trigger of endothelial activation and cardiac re-modeling to associate with IL-1β signaling in KD. Besides a potential patho-mechanistic implication of these findings, our data suggest blood leukocyte and neutrophil counts to best predict response to IL-1Ra treatment in IVIG-resistant KD. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-022-01301-w.
Collapse
|
38
|
IL-1 receptor antagonist, MIS-C, and the peculiar autoimmunity of SARS-CoV-2. THE LANCET RHEUMATOLOGY 2022; 4:e305-e307. [PMID: 35368385 PMCID: PMC8963768 DOI: 10.1016/s2665-9913(22)00090-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Autoantibodies against interleukin-1 receptor antagonist in multisystem inflammatory syndrome in children: a multicentre, retrospective, cohort study. THE LANCET RHEUMATOLOGY 2022; 4:e329-e337. [PMID: 35368387 PMCID: PMC8963770 DOI: 10.1016/s2665-9913(22)00064-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Multisystem inflammatory syndrome in children (MIS-C) is a rare but serious complication of infection with SARS-CoV-2. A possible involvement of pathogenetically relevant autoantibodies has been discussed. Recently, neutralising autoantibodies against inflammatory receptor antagonists progranulin and interleukin-1 receptor antagonist (IL-1Ra) were found in adult patients with critical COVID-19. The aim of this study was to investigate the role of such autoantibodies in MIS-C. Methods In this multicentre, retrospective, cohort study, plasma and serum samples were collected from patients (0–18 years) with MIS-C (as per WHO criteria) treated at five clinical centres in Germany and Spain. As controls, we included plasma or serum samples from children with Kawasaki disease, children with inactive systemic juvenile idiopathic arthritis, and children with suspected growth retardation (non-inflammatory control) across four clinical centres in Germany and Spain (all aged ≤18 years). Serum samples from the CoKiBa trial were used as two further control groups, from healthy children (negative for SARS-CoV-2 antibodies) and children with previous mild or asymptomatic COVID-19 (aged ≤17 years). MIS-C and control samples were analysed for autoantibodies against IL-1Ra and progranulin, and for IL-1Ra concentrations, by ELISA. Biochemical analysis of plasma IL-1Ra was performed with native Western blots and isoelectric focusing. Functional activity of the autoantibodies was examined by an in vitro IL-1β-signalling reporter assay. Findings Serum and plasma samples were collected between March 6, 2011, and June 2, 2021. Autoantibodies against IL-1Ra could be detected in 13 (62%) of 21 patients with MIS-C (11 girls and ten boys), but not in children with Kawasaki disease (n=24; nine girls and 15 boys), asymptomatic or mild COVID-19 (n=146; 72 girls and 74 boys), inactive systemic juvenile idiopathic arthritis (n=10; five girls and five boys), suspected growth retardation (n=33; 13 girls and 20 boys), or in healthy controls (n=462; 230 girls and 232 boys). Anti-IL-1Ra antibodies in patients with MIS-C belonged exclusively to the IgG1 subclass, except in one patient who had additional IL-1Ra-specific IgM antibodies. Autoantibodies against progranulin were only detected in one (5%) patient with MIS-C. In patients with MIS-C who were positive for anti-IL-1Ra antibodies, free plasma IL-1Ra concentrations were reduced, and immune-complexes of IL-1Ra were detected. Notably, an additional, hyperphosphorylated, transiently occurring atypical isoform of IL-1Ra was observed in all patients with MIS-C who were positive for anti-IL-1Ra antibodies. Anti-IL-1Ra antibodies impaired IL-1Ra function in reporter cell assays, resulting in amplified IL-1β signalling. Interpretation Anti-IL-1Ra autoantibodies were observed in a high proportion of patients with MIS-C and were specific to these patients. Generation of these autoantibodies might be triggered by an atypical, hyperphosphorylated isoform of IL-1Ra. These autoantibodies impair IL-1Ra bioactivity and might thus contribute to increased IL-1β-signalling in MIS-C. Funding NanoBioMed fund of the University of Saarland, José Carreras Center for Immuno and Gene Therapy, Dr Rolf M Schwiete Stiftung, Staatskanzlei Saarland, German Heart Foundation, Charity of the Blue Sisters, Bavarian Ministry of Health, the Center for Interdisciplinary Clinical Research at University Hospital Münster, EU Horizon 2020.
Collapse
|
40
|
Cavalcanti A, Islabão A, Magalhães C, Veloso S, Lopes M, do Prado R, Aquilante B, Terrazas AM, Rezende MF, Clemente G, Terreri MT. Paediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2 (PIMS-TS): a Brazilian cohort. Adv Rheumatol 2022; 62:6. [PMID: 35189972 PMCID: PMC8860252 DOI: 10.1186/s42358-022-00237-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/11/2022] [Indexed: 11/17/2022] Open
Abstract
Background Paediatric inflammatory multisystem syndrome (PIMS) associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been described since mid-April 2020 with the first reports coming from Europe. Our objective was to describe the characteristics of patients among the Brazilian population.
Methods A multicenter retrospective study was conducted with the participation of five pediatric rheumatology centers in Brazil during the period from March to November 2020. Children and adolescents with PIMS temporally associated with SARS-CoV-2 (TS) who met the definition criteria for the disease according to the Royal College of Paediatrics and Child Health were included. Demographic, clinical, laboratory, therapeutic characteristics and molecular and serological diagnosis of SARS-CoV-2 infection were described.
Results Fifty-seven children and adolescents with PIMS-TS were evaluated, 54% female, with a median age of 8 (3–11) years. Most (86%) were previously healthy, with asthma being the main comorbidity, present in 10% of the patients. Fever was the main manifestation, present in all patients, followed by mucocutaneous and gastrointestinal features, present in 89% and 81% of the patients, respectively. Myocarditis occurred in 21% of the patients and in 68% of them required intensive care. The Kawasaki disease phenotype occurred in most patients (77%). All patients had elevated inflammatory markers, with elevated CRP being the most found (98%). Anemia and lymphopenia were present in 79% and 72%, respectively. Laboratory evidence of SARS-CoV-2 was found in 77% of the patients, with 39% positive RT-PCR and 84% positive serology for SARS-CoV-2. An immunomodulatory treatment was performed in 91% of the patients, with 67% receiving intravenous immunoglobulin (IVIG) associated with glucocorticoid, 21% receiving IVIG, and 3.5% receiving glucocorticoid. The median length of hospitalization was 10 days. Conclusions This study showed a high morbidity of PIMS-TS in Brazilian children, with a prolonged length of hospitalization and a high rate of admission to pediatric intensive care unit. Multicenter prospective studies are needed to assess the morbidity of the disease in the medium and long term.
Collapse
|
41
|
Morita A, Hosaka S, Imagawa K, Ishiodori T, Nozaki Y, Murakami T, Takada H. Time course of peripheral immunophenotypes of multisystem inflammatory syndrome in children. Clin Immunol 2022; 236:108955. [PMID: 35150919 PMCID: PMC8828386 DOI: 10.1016/j.clim.2022.108955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 01/06/2023]
Abstract
The etiology of multiple inflammatory syndrome in children (MIS-C) remains poorly understood. As clues to elucidate the pathogenic condition, several characteristic peripheral immunophenotypes have been reported in MIS-C. However, no report has demonstrated the time course of the peripheral immunophenotype along with the clinical course in the same patient. Herein, we clarified the immunological characteristics of a Japanese patient with MIS-C. There was an initial cytokine storm followed by T-cell activation, especially of CD8+ T cells, with the expansion of T-cell receptor Vβ 21.3-expressing cells, which suggests superantigen-mediated T-cell activation. In addition, we also found an increase in IgG-producing cells (plasmablasts and switched memory B cells), which were accompanied by elevated serum levels of anti-SARS-CoV-2 spike antigen-specific IgG antibodies. These time course of peripheral immunophenotypes support that immunological activation against SARS-CoV-2 spike protein plays a central role in the etiology of MIS-C.
Collapse
Affiliation(s)
- Atsushi Morita
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan.
| | - Sho Hosaka
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Kazuo Imagawa
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan; Department of Child Health, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takumi Ishiodori
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Yoshihiro Nozaki
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan
| | - Takashi Murakami
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan; Department of Child Health, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576, Japan; Department of Child Health, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
42
|
Multisystem inflammatory syndrome in children and Kawasaki disease: a critical comparison. Nat Rev Rheumatol 2021; 17:731-748. [PMID: 34716418 PMCID: PMC8554518 DOI: 10.1038/s41584-021-00709-9] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 12/12/2022]
Abstract
Children and adolescents infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are predominantly asymptomatic or have mild symptoms compared with the more severe coronavirus disease 2019 (COVID-19) described in adults. However, SARS-CoV-2 is also associated with a widely reported but poorly understood paediatric systemic vasculitis. This multisystem inflammatory syndrome in children (MIS-C) has features that overlap with myocarditis, toxic-shock syndrome and Kawasaki disease. Current evidence indicates that MIS-C is the result of an exaggerated innate and adaptive immune response, characterized by a cytokine storm, and that it is triggered by prior SARS-CoV-2 exposure. Epidemiological, clinical and immunological differences classify MIS-C as being distinct from Kawasaki disease. Differences include the age range, and the geographical and ethnic distribution of patients. MIS-C is associated with prominent gastrointestinal and cardiovascular system involvement, admission to intensive care unit, neutrophilia, lymphopenia, high levels of IFNγ and low counts of naive CD4+ T cells, with a high proportion of activated memory T cells. Further investigation of MIS-C will continue to enhance our understanding of similar conditions associated with a cytokine storm.
Collapse
|
43
|
Ganigara M, Sharma C, Bayry J. Unraveling the mechanisms of IVIG immunotherapy in MIS-C. CELL REPORTS MEDICINE 2021; 2:100431. [PMID: 34608458 PMCID: PMC8481087 DOI: 10.1016/j.xcrm.2021.100431] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In The Journal of Clinical Investigation, Zhu et al.1 report that intravenous immunoglobulin (IVIG) targets IL-1β+ neutrophils to exert anti-inflammatory effects in multisystem inflammatory syndrome in children (MIS-C), a post-infectious inflammatory condition associated with COVID-19.
Collapse
Affiliation(s)
- Madhusudan Ganigara
- Division of Pediatric Cardiology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Chetan Sharma
- Division of Pediatric Cardiology, Children's Hospital of San Antonio/Baylor College of Medicine, San Antonio, TX 78207, USA
| | - Jagadeesh Bayry
- Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, Kerala, India
| |
Collapse
|