Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 429
Filtrer
1.
J Clin Immunol ; 45(1): 4, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39264477

RÉSUMÉ

High ferritin is an important and sensitive biomarker for the various forms of hemophagocytic lymphohistiocytosis (HLH), a diverse and deadly group of cytokine storm syndromes. Early action to prevent immunopathology in HLH often includes empiric immunomodulation, which can complicate etiologic work-up and prevent collection of early/pre-treatment research samples. To address this, we instituted an alert system at UPMC Children's Hospital where serum ferritin > 1000 ng/mL triggered real-time chart review, assessment of whether the value reflected "inflammatory hyperferritnemia (IHF)", and biobanking of remnant samples from consenting IHF patients. We extracted relevant clinical data; periodically measured serum total IL-18, IL-18 binding protein (IL-18BP), and CXCL9; retrospectively classified patients by etiology into infectious, rheumatic, or immune dysregulation; and subjected a subgroup of samples to a 96-analyte biomarker screen. 180 patients were identified, 30.5% of which had IHF. Maximum ferritin levels were significantly higher in patients with IHF than with either hemoglobinopathy or transplant, and highly elevated total IL-18 levels were distinctive to patients with Stills Disease and/or Macrophage Activation Syndrome (MAS). Multi-analyte analysis showed elevation in proteins associated with cytotoxic lymphocytes in all IHF samples when compared to healthy controls and depression of proteins such as ANGPT1 and VEGFR2 in samples from hyperferritinemic sepsis patients relative to non-sepsis controls. This real-time IFH screen proved feasible and efficient, validated prior observations about the specificity of IL-18, enabled early sample collection from a complex population, suggested a unique vascular biomarker signature in hyperferritinemic sepsis, and expanded our understanding of IHF heterogeneity.


Sujet(s)
Marqueurs biologiques , Ferritines , Hyperferritinémie , Interleukine-18 , Lymphohistiocytose hémophagocytaire , Humains , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/sang , Lymphohistiocytose hémophagocytaire/immunologie , Marqueurs biologiques/sang , Femelle , Interleukine-18/sang , Mâle , Hyperferritinémie/diagnostic , Hyperferritinémie/sang , Enfant , Ferritines/sang , Enfant d'âge préscolaire , Nourrisson , Adolescent , Diagnostic différentiel , Protéines et peptides de signalisation intercellulaire/sang , Chimiokine CXCL9/sang , Inflammation/diagnostic , Inflammation/sang , Inflammation/immunologie , Études rétrospectives
3.
Adv Exp Med Biol ; 1448: 145-159, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117813

RÉSUMÉ

Natural killer (NK) cells are innate immune lymphocytes that rapidly produce cytokines upon activation and kill target cells. NK cells have been of particular interest in primary hemophagocytic lymphohistiocytosis (pHLH) since all of the genetic defects associated with this disorder cause diminished cytotoxic capacity of NK cells and T lymphocytes, and assays of NK cell killing are used clinically for the diagnosis of HLH. Herein, we review human NK cell biology and the significance of alterations in NK cell function in the diagnosis and pathogenesis of HLH.


Sujet(s)
Syndrome de libération de cytokines , Cellules tueuses naturelles , Lymphohistiocytose hémophagocytaire , Humains , Animaux , Cellules tueuses naturelles/immunologie , Syndrome de libération de cytokines/immunologie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/immunologie , Cytokines/immunologie
4.
Adv Exp Med Biol ; 1448: 75-101, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117809

RÉSUMÉ

Hemophagocytic lymphohistiocytosis (HLH) constitutes a rare, potentially life-threatening hyperinflammatory immune dysregulation syndrome that can present with a variety of clinical signs and symptoms, including fever, hepatosplenomegaly, and abnormal laboratory and immunological findings such as cytopenias, hyperferritinemia, hypofibrinogenemia, hypertriglyceridemia, elevated blood levels of soluble CD25 (interleukin (IL)-2 receptor α-chain), or diminished natural killer (NK)-cell cytotoxicity (reviewed in detail in Chapter 11 of this book). While HLH can be triggered by an inciting event (e.g., infections), certain monogenic causes have been associated with a significantly elevated risk of development of HLH, or recurrence of HLH in patients who have recovered from their disease episode. These monogenic predisposition syndromes are variably referred to as "familial" (FHL) or "primary" HLH (henceforth referred to as "pHLH") and are the focus of this chapter. Conversely, secondary HLH (sHLH) often occurs in the absence of monogenic etiologies that are commonly associated with pHLH and can be triggered by infections, malignancies, or rheumatological diseases; these triggers and the genetics associated with sHLH are discussed in more detail in other chapters in this book.


Sujet(s)
Lymphohistiocytose hémophagocytaire , Animaux , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/immunologie , Récidive , Facteurs de risque , Prédisposition génétique à une maladie , Sous-unité alpha du récepteur à l'interleukine-2/immunologie , Cellules T tueuses naturelles/immunologie , Cytotoxicité immunologique
5.
Adv Exp Med Biol ; 1448: 9-19, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117804

RÉSUMÉ

Hemophagocytic lymphohistiocytosis (HLH) is a severe cytokine storm syndrome (CSS), which until the turn of the century, was barely known but is now receiving increased attention. The history of HLH dates back to 1939 when it was first described in adults, to be followed in 1952 by the first description of its primary, familial form in children. Secondary forms of HLH are far more frequent and occur with infections, malignancies, metabolic diseases, iatrogenic immune suppression, and autoinflammatory/autoimmune diseases. Identification of the genetic defects leading to the defective function of natural killer (NK) cells and cytotoxic T cells as well as the corresponding mouse models have revolutionized our understanding of HLH and of immune function. Diagnosis relies on clinical and laboratory criteria; functional and genetic tests can help separate primary from secondary forms. Treatment with immunochemotherapy and hematopoietic stem cell transplantation has considerably improved survival in children with primary HLH, a formerly uniformly fatal disease.


Sujet(s)
Lymphohistiocytose hémophagocytaire , Lymphohistiocytose hémophagocytaire/thérapie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/immunologie , Humains , Histoire du 20ème siècle , Animaux , Histoire du 21ème siècle , Cellules tueuses naturelles/immunologie , Transplantation de cellules souches hématopoïétiques
6.
Adv Exp Med Biol ; 1448: 43-58, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117807

RÉSUMÉ

The laboratory diagnosis of cytokine storm syndromes (CSSs), i.e., hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS), is often challenging. The laboratory features using routinely available tests lack specificity, whereas confirmatory testing is available in only few laboratories in the United States. The disease mechanisms are still largely unclear, particularly in adults. In this chapter, the pathogenesis of CSSs, their associated laboratory findings, and recommended diagnostic strategies are reviewed.


Sujet(s)
Syndrome de libération de cytokines , Lymphohistiocytose hémophagocytaire , Syndrome d'activation macrophagique , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/diagnostic , Syndrome de libération de cytokines/anatomopathologie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/anatomopathologie , Syndrome d'activation macrophagique/diagnostic , Syndrome d'activation macrophagique/anatomopathologie , Cytokines/métabolisme
7.
Adv Exp Med Biol ; 1448: 103-119, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117810

RÉSUMÉ

Secondary hemophagocytic lymphohistiocytosis (sHLH) has historically been defined as a cytokine storm syndrome (CSS) occurring in the setting of triggers leading to strong and dysregulated immunological activation, without known genetic predilection. However, recent studies have suggested that existing underlying genetic factors may synergize with particular diseases and/or environmental triggers (including infection, autoimmune/autoinflammatory disorder, certain biologic therapies, or malignant transformation), leading to sHLH. With the recent advances in genetic testing technology, more patients are examined for genetic variations in primary HLH (pHLH)-associated genes, including through whole exome and whole genome sequencing. This expanding genetic and genomic evidence has revealed HLH as a more complex phenomenon, resulting from specific immune challenges in patients with a susceptible genetic background. Rather than a simple, binary definition of pHLH and sHLH, HLH represents a spectrum of diseases, from a severe complication of common infections (EBV, influenza) to early onset familial diseases that can only be cured by transplantation.


Sujet(s)
Syndrome de libération de cytokines , Prédisposition génétique à une maladie , Lymphohistiocytose hémophagocytaire , Humains , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/thérapie , Syndrome de libération de cytokines/génétique , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/étiologie
8.
Adv Exp Med Biol ; 1448: 21-31, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117805

RÉSUMÉ

In 1979, it became recognized in the literature that what we call hemophagocytic lymphohistiocytosis (HLH) was a nonmalignant disease of histiocytes. Subsequently a familial form and a secondary form of HLH were differentiated. When HLH is secondary to an autoimmune disease, rheumatologists refer to this entity as macrophage activation syndrome (MAS) to differentiate it from HLH itself. Although the first cases of MAS likely appeared in the literature in the 1970s, it was not until 1985 that the term activated macrophages was used to describe patients with systemic juvenile idiopathic arthritis (sJIA) complicated by MAS and the term macrophage activation syndrome first appeared in the title of a paper in 1993.MAS is one of the many types of secondary HLH and should not be confused with primary HLH. Experience has taught that MAS secondary to different autoimmune diseases is not equal. In the 30 years since initial description in patients with sJIA, the clinical spectrum, diseases associated with MAS, therapy, and understanding the pathogenesis have all made significant gains. The diagnostic/classification criteria for MAS secondary to sJIA, SLE, RA, and KD differ based on the different laboratory abnormalities associated with each (Ahn et al., J Rheumatol 44:996-1003, 2017; Han et al., Ann Rheum Dis 75:e44, 2016; Ravelli et al., Ann Rheum Dis 75:481-489, 2016; Borgia et al., Arthritis Rheumatol 70:616-624, 2018). These examples include the thrombocytosis associated with sJIA, a chronic generalized activation of the immune system, leading to elevations of fibrinogen and sIL-2R, low platelet count associated with SLE, and more acute inflammation associated with KD. Therefore, individual diagnostic criteria are required, and they all differ from the diagnostic criteria for HLH, which are based on a previously non-activated immune system (Ahn et al., J Rheumatol 44:996-1003, 2017; Han et al., Ann Rheum Dis 75:e44, 2016; Ravelli et al., Ann Rheum Dis 75:481-489, 2016; Borgia et al., Arthritis Rheumatol 70:616-624, 2018; Henter et al., Pediatr Blood Cancer 48:124-131, 2007). This helps to explain why the HLH diagnostic criteria do not perform well in MAS.The initial treatment remains high-dose steroids and IVIG followed by the use of a calcineurin inhibitor for resistant cases. IVIG can be used if there is a concern about malignancy to wait for appropriate investigations or with steroids. Interluekin-1 inhibition is now the next therapy if there is a failure to respond to steroids and calcineurin inhibitors. Advances in understanding the mechanisms leading to MAS, which has been greatly aided by the use of mouse models of MAS and advances in genome sequencing, offer a bright future for more specific therapies. More recent therapies are directed to specific cytokines involved in the pathogenesis of MAS and can lead to decreases in the morbidity and mortality associated with MAS. These include therapies directed to inhibiting the JAK/STAT pathway and/or specific cytokines, interleukin-18 and gamma interferon, which are currently being studied in MAS. These more specific therapies may obviate the need for nonspecific immunosuppressive therapies including high-dose prolonged steroids, calcineurin inhibitors, and etoposide.


Sujet(s)
Maladies auto-immunes , Syndrome d'activation macrophagique , Humains , Syndrome d'activation macrophagique/diagnostic , Syndrome d'activation macrophagique/immunologie , Maladies auto-immunes/immunologie , Histoire du 20ème siècle , Histoire du 21ème siècle , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/thérapie
9.
Adv Exp Med Biol ; 1448: 121-126, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117811

RÉSUMÉ

Macrophage activation syndrome (MAS) is a life-threatening episode of hyperinflammation driven by excessive activation and expansion of T cells (mainly CD8) and hemophagocytic macrophages producing proinflammatory cytokines. MAS has been reported in association with almost every rheumatic disease, but it is by far most common in systemic juvenile idiopathic arthritis (SJIA). Clinically, MAS is similar to familial or primary hemophagocytic lymphohistiocytosis (pHLH), a group of rare autosomal recessive disorders linked to various genetic defects all affecting the perforin-mediated cytolytic pathway employed by NK cells and cytotoxic CD8 T lymphocytes. Decreased cytolytic activity in pHLH patients leads to prolonged survival of target cells associated with increased production of proinflammatory cytokines that overstimulate macrophages. The resulting cytokine storm is believed to be responsible for the frequently fatal multiorgan system failure seen in MAS. Whole exome sequencing as well as targeted sequencing of pHLH-associated genes in patients with SJIA-associated MAS demonstrated increased "burden" of rare protein-altering variants affecting the cytolytic pathway compared to healthy controls, suggesting that as in pHLH, genetic variability in the cytolytic pathway contributes to MAS predisposition. Functional studies of some of the novel variants have shown that even in a heterozygous state, their presence partially reduces cytolytic activity that may lead to increased cytokine production.


Sujet(s)
Arthrite juvénile , Syndrome d'activation macrophagique , Humains , Syndrome d'activation macrophagique/génétique , Syndrome d'activation macrophagique/immunologie , Arthrite juvénile/génétique , Arthrite juvénile/immunologie , Arthrite juvénile/complications , Prédisposition génétique à une maladie , Cellules tueuses naturelles/immunologie , Cytokines/génétique , Cytokines/métabolisme , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/immunologie , Macrophages/immunologie , Macrophages/métabolisme
10.
Adv Exp Med Biol ; 1448: 227-248, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117818

RÉSUMÉ

Epstein-Barr virus (EBV) is a ubiquitous and predominantly B cell tropic virus. One of the most common viruses to infect humans, EBV, is best known as the causative agent of infectious mononucleosis (IM). Although most people experience asymptomatic infection, EBV is a potent immune stimulus and as such it elicits robust proliferation and activation of the B-lymphocytes it infects as well as the immune cells that respond to infection. In certain individuals, such as those with inherited or acquired defects affecting the immune system, failure to properly control EBV leads to the accumulation of EBV-infected B cells and EBV-reactive immune cells, which together contribute to the development of often life-threatening cytokine storm syndromes (CSS). Here, we review the normal immune response to EBV and discuss several CSS associated with EBV, such as chronic active EBV infection, hemophagocytic lymphohistiocytosis, and post-transplant lymphoproliferative disorder. Given the critical role for cytokines in driving inflammation and contributing to disease pathogenesis, we also discuss how targeting specific cytokines provides a rational and potentially less toxic treatment for EBV-driven CSS.


Sujet(s)
Syndrome de libération de cytokines , Cytokines , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/virologie , Herpèsvirus humain de type 4/immunologie , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/complications , Cytokines/immunologie , Cytokines/métabolisme , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/virologie , Lymphocytes B/immunologie , Lymphocytes B/virologie , Syndromes lymphoprolifératifs/immunologie , Syndromes lymphoprolifératifs/virologie , Animaux
11.
Adv Exp Med Biol ; 1448: 249-267, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117819

RÉSUMÉ

A wide variety of infections can trigger cytokine storm syndromes including those caused by bacteria, viruses, fungi and parasites. The most frequent viral trigger is Epstein-.Barr virus which is covered in Chapter 16. CSS associated with COVID-19 is also discussed separately (Chapter 22). This chapter will focus on other viruses including the hemorrhagic fever viruses, influenza, parainfluenza, adenovirus, parvovirus, hepatitis viruses, measles, mumps, rubella, enterovirus, parechovirus, rotavirus, human metapneumovirus and human T-lymphotropic virus. The published literature consists of many single case reports and moderate-sized case series reporting CSS, in most circumstances meeting the 2004 diagnostic criteria for hemophagocytic lymphohistiocytosis (HLH). There is no published clinical trial evidence specifically for management of HLH associated with these viruses. In some situations, patients received supportive therapy and blood product transfusions only but in most cases, they were treated with one or more of intravenous corticosteroids, intravenous immunoglobulin and/or etoposide. These were successful in many patients although in significant numbers progression of infection to CSS was associated with mortality.


Sujet(s)
COVID-19 , Syndrome de libération de cytokines , Humains , Syndrome de libération de cytokines/immunologie , COVID-19/complications , COVID-19/immunologie , COVID-19/thérapie , COVID-19/virologie , Lymphohistiocytose hémophagocytaire/thérapie , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/virologie , SARS-CoV-2 , Fièvres hémorragiques virales/virologie
12.
Adv Exp Med Biol ; 1448: 285-291, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117822

RÉSUMÉ

Zoonotic infections can result in life-threatening complications that can manifest with hemophagocytic lymphohistiocytosis (HLH)/cytokine storm syndrome (CSS). Bacteria constitute the largest group of zoonotic infection-related HLH cases. The growing list of zoonotic bacterial infections associated with HLH/CSS include Brucella spp., Rickettsia spp., Ehrlichia, Coxiella burnetii, Mycobacterium spp., and Bartonella spp. Patients most commonly present with fever, cytopenias, hepatosplenomegaly, myalgias, and less frequently with rash, jaundice, and lymphadenopathy.


Sujet(s)
Syndrome de libération de cytokines , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/microbiologie , Syndrome de libération de cytokines/étiologie , Animaux , Zoonoses bactériennes/microbiologie , Lymphohistiocytose hémophagocytaire/microbiologie , Lymphohistiocytose hémophagocytaire/immunologie , Zoonoses/microbiologie
13.
Adv Exp Med Biol ; 1448: 429-440, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117831

RÉSUMÉ

Hemophagocytic lymphohistiocytosis (HLH) has been described for decades in association with malignancies (M-HLH). While its mechanism is unknown, M-HLH has a poor prognosis, ranging from 10% to 30% overall survival. Mature T-cell lymphomas, diffuse large B-cell lymphoma, and Hodgkin lymphoma, with or without viral co-triggers such as Epstein-Barr virus, are among the most frequent underlying entities. Most M-HLH cases occur at the presentation of malignancy, but they may also occur during therapy as a result of immune compromise from chemotherapy (HLH in the context of immune compromise, IC-HLH) and (typically) disordered response to infection or after immune-activating therapies (Rx-HLH, also known as cytokine release syndrome, CRS). IC-HLH typically occurs months after diagnosis in the context of fungal, bacterial, or viral infection, though it may occur without an apparent trigger. Rx-HLH can be associated with checkpoint blockade, chimeric antigen receptor T-cell therapy, or bispecific T-cell engaging therapy. Until recently, M-HLH diagnosis and treatment strategies were extrapolated from familial HLH (F-HLH), though optimized diagnostic and therapeutic treatment strategies are emerging.


Sujet(s)
Tumeurs hématologiques , Lymphohistiocytose hémophagocytaire , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/diagnostic , Lymphohistiocytose hémophagocytaire/étiologie , Lymphohistiocytose hémophagocytaire/thérapie , Humains , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/thérapie , Tumeurs/immunologie , Tumeurs/thérapie , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/étiologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
14.
Adv Exp Med Biol ; 1448: 481-496, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117835

RÉSUMÉ

Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory disease caused by mutations in effectors and regulators of cytotoxicity in cytotoxic T cells (CTL) and natural killer (NK) cells. The complexity of the immune system means that in vivo models are needed to efficiently study diseases like HLH. Mice with defects in the genes known to cause primary HLH (pHLH) are available. However, these mice only develop the characteristic features of HLH after the induction of an immune response (typically through infection with lymphocytic choriomeningitis virus). Nevertheless, murine models have been invaluable for understanding the mechanisms that lead to HLH. For example, the cytotoxic machinery (e.g., the transport of cytotoxic vesicles and the release of granzymes and perforin after membrane fusion) was first characterized in the mouse. Experiments in murine models of pHLH have emphasized the importance of cytotoxic cells, antigen-presenting cells (APC), and cytokines in hyperinflammatory positive feedback loops (e.g., cytokine storms). This knowledge has facilitated the development of treatments for human HLH, some of which are now being tested in the clinic.


Sujet(s)
Syndrome de libération de cytokines , Modèles animaux de maladie humaine , Lymphohistiocytose hémophagocytaire , Animaux , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/génétique , Souris , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/génétique , Syndrome de libération de cytokines/anatomopathologie , Cytokines/métabolisme , Cytokines/génétique , Lymphocytes T cytotoxiques/immunologie , Cellules tueuses naturelles/immunologie
15.
Adv Exp Med Biol ; 1448: 497-522, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117836

RÉSUMÉ

Hemophagocytic lymphohistiocytosis (HLH) comprises a broad spectrum of life-threatening cytokine storm syndromes, classified into primary (genetic) or secondary (acquired) HLH. The latter occurs in a variety of medical conditions, including infections, malignancies, autoimmune and autoinflammatory diseases, acquired immunodeficiency, and metabolic disorders. Despite recent advances in the field, the pathogenesis of secondary HLH remains incompletely understood. Considering the heterogeneity of triggering factors and underlying diseases in secondary HLH, a large diversity of animal models has been developed to explore pivotal disease mechanisms. To date, over 20 animal models have been described that each recapitulates certain aspects of secondary HLH. This review provides a comprehensive overview of the existing models, highlighting relevant findings, discussing the involvement of different cell types and cytokines in disease development and progression, and considering points of interest toward future therapeutic strategies.


Sujet(s)
Syndrome de libération de cytokines , Modèles animaux de maladie humaine , Lymphohistiocytose hémophagocytaire , Animaux , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/anatomopathologie , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/anatomopathologie , Syndrome de libération de cytokines/étiologie , Souris , Humains , Cytokines/métabolisme
16.
Adv Exp Med Biol ; 1448: 441-457, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117832

RÉSUMÉ

There is extensive overlap of clinical features among familial or primary HLH (pHLH), reactive or secondary hemophagocytic lymphohistiocytosis (sHLH) [including macrophage activation syndrome (MAS) related to rheumatic diseases], and hyperferritinemic sepsis-induced multiple organ dysfunction syndrome (MODS); however, the distinctive pathobiology that causes hyperinflammatory process in each condition requires careful considerations for therapeutic decision-making. pHLH is defined by five or more of eight HLH-2004 criteria [1], where genetic impairment of natural killer (NK) cells or CD8+ cytolytic T cells results in interferon gamma (IFN-γ)-induced hyperinflammation regardless of triggering factors. Cytolytic treatments (e.g., etoposide) or anti-IFN-γ monoclonal antibody (emapalumab) has been effectively used to bridge the affected patients to hematopoietic stem cell transplant. Secondary forms of HLH also have normal NK cell number with decreased cytolytic function of varying degrees depending on the underlying and triggering factors. Although etoposide was uniformly used in sHLH/MAS in the past, the treatment strategy in different types of sHLH/MAS is increasingly streamlined to reflect the triggering/predisposing conditions, severity/progression, and comorbidities. Accordingly, in hyperferritinemic sepsis, the combination of hepatobiliary dysfunction (HBD) and disseminated intravascular coagulation (DIC) reflects reticuloendothelial system dysfunction and defines sepsis-associated MAS. It is demonstrated that as the innate immune response to infectious organism prolongs, it results in reduction in T cells and NK cells with subsequent lymphopenia even though normal cytolytic activity continues (Figs. 30.1, 30.2, 30.3, and 30.4). These changes allow free hemoglobin and pathogens to stimulate inflammasome activation in the absence of interferon-γ (IFN-γ) production that often responds to source control, intravenous immunoglobulin (IVIg), plasma exchange, and interleukin 1 receptor antagonist (IL-1Ra), similar to non-EBV, infection-induced HLH.


Sujet(s)
Syndrome de libération de cytokines , Lymphohistiocytose hémophagocytaire , Défaillance multiviscérale , Sepsie , Humains , Défaillance multiviscérale/étiologie , Défaillance multiviscérale/immunologie , Sepsie/immunologie , Sepsie/complications , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/étiologie , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/étiologie , Syndrome d'activation macrophagique/immunologie , Syndrome d'activation macrophagique/étiologie , Cellules tueuses naturelles/immunologie
17.
Adv Exp Med Biol ; 1448: 601-609, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117842

RÉSUMÉ

Cytokine storm syndromes (CSS) include different entities such as macrophage activation syndrome, primary and secondary hemophagocytic lymphohistiocytosis (HLH), and multisystem inflammatory syndrome in children (MIS-C) associated with COVID-19. An effective management strategy is critical in CSS. While biologics have become an essential part of CSS treatment, hematopoietic stem cell transplantation (HSCT) has changed the fate of primary HLH patients. This chapter will focus on the available alternative immunomodulatory therapies in CSS, which include corticosteroids, cyclosporine A, intravenous immunoglobulin, interleukin 18 binding protein, therapeutic plasmapheresis, HSCT, and mesenchymal stromal cell-based therapies.


Sujet(s)
COVID-19 , Syndrome de libération de cytokines , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/thérapie , COVID-19/immunologie , COVID-19/thérapie , COVID-19/complications , Transplantation de cellules souches hématopoïétiques , SARS-CoV-2/immunologie , Lymphohistiocytose hémophagocytaire/thérapie , Lymphohistiocytose hémophagocytaire/immunologie , Plasmaphérèse/méthodes , Agents immunomodulateurs/usage thérapeutique , Immunoglobulines par voie veineuse/usage thérapeutique , Transplantation de cellules souches mésenchymateuses/méthodes , Ciclosporine/usage thérapeutique , Syndrome de réponse inflammatoire généralisée/immunologie , Syndrome de réponse inflammatoire généralisée/thérapie
18.
Adv Exp Med Biol ; 1448: 553-563, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117838

RÉSUMÉ

Interleukin-1 is a prototypic proinflammatory cytokine that is elevated in cytokine storm syndromes (CSSs), such as secondary hemophagocytic lymphohistiocytosis (sHLH) and macrophage activation syndrome (MAS). IL-1 has many pleotropic and redundant roles in both innate and adaptive immune responses. Blockade of IL-1 with recombinant human interleukin-1 receptor antagonist has shown efficacy in treating CSS. Recently, an IL-1 family member, IL-18, has been demonstrated to be contributory to CSS in autoinflammatory conditions, such as in inflammasomopathies (e.g., NLRC4 mutations). Anecdotally, recombinant IL-18 binding protein can be of benefit in treating IL-18-driven CSS. Lastly, another IL-1 family member, IL-33, has been postulated to contribute to CSS in an animal model of disease. Targeting of IL-1 and related cytokines holds promise in treating a variety of CSS.


Sujet(s)
Syndrome de libération de cytokines , Interleukine-1 , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/traitement médicamenteux , Interleukine-1/antagonistes et inhibiteurs , Interleukine-1/immunologie , Interleukine-1/génétique , Interleukine-1/métabolisme , Animaux , Antagoniste du récepteur à l'interleukine-1/usage thérapeutique , Antagoniste du récepteur à l'interleukine-1/génétique , Protéines de liaison au calcium/génétique , Interleukine-18/génétique , Interleukine-18/immunologie , Syndrome d'activation macrophagique/immunologie , Syndrome d'activation macrophagique/traitement médicamenteux , Syndrome d'activation macrophagique/génétique , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/traitement médicamenteux , Protéines adaptatrices de signalisation CARD
19.
Adv Exp Med Biol ; 1448: 573-582, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117840

RÉSUMÉ

A vast body of evidence provides support to a central role of exaggerated production of interferon-γ (IFN-γ) in causing hypercytokinemia and signs and symptoms of hemophagocytic lymphohistiocytosis (HLH). In this chapter, we will describe briefly the roles of IFN-γ in innate and adaptive immunity and in host defense, summarize results from animal models of primary HLH and secondary HLH with particular emphasis on targeted therapeutic approaches, review data on biomarkers associated with activation of the IFN-γ pathway, and discuss initial efficacy and safety results of IFN-γ neutralization in humans.


Sujet(s)
Syndrome de libération de cytokines , Immunité innée , Interféron gamma , Lymphohistiocytose hémophagocytaire , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/étiologie , Interféron gamma/immunologie , Animaux , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/traitement médicamenteux , Immunité innée/effets des médicaments et des substances chimiques , Immunité acquise/effets des médicaments et des substances chimiques
20.
Adv Exp Med Biol ; 1448: 565-572, 2024.
Article de Anglais | MEDLINE | ID: mdl-39117839

RÉSUMÉ

Interleukin-6 (IL-6) is a pro-inflammatory cytokine elevated in cytokine storm syndromes, including hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS). It is also elevated in cytokine release syndrome (CRS) after immune activating cancer therapies such as chimeric antigen receptor (CAR) T-cells or bispecific T-cell engagers (BITEs) and in some patients after infection with SARS-CoV-2. The interaction of IL-6 with its receptor complex can happen in several forms, making effectively blocking this cytokine's effects clinically challenging. Fortunately, effective clinical agents targeting the IL-6 receptor (tocilizumab) and IL-6 directly (siltuximab) have been developed and are approved for use in humans. IL-6 blockade has now been used to safely and effectively treat several cytokine storm syndromes (CSS). Other methods of investigation in effective IL-6 blockade are underway.


Sujet(s)
Anticorps monoclonaux humanisés , COVID-19 , Syndrome de libération de cytokines , Interleukine-6 , Récepteurs à l'interleukine-6 , Humains , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/traitement médicamenteux , Interleukine-6/antagonistes et inhibiteurs , Interleukine-6/immunologie , Interleukine-6/métabolisme , Anticorps monoclonaux humanisés/usage thérapeutique , COVID-19/immunologie , Récepteurs à l'interleukine-6/antagonistes et inhibiteurs , Récepteurs à l'interleukine-6/immunologie , SARS-CoV-2/immunologie , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/traitement médicamenteux , Anticorps monoclonaux/usage thérapeutique , Syndrome d'activation macrophagique/immunologie , Syndrome d'activation macrophagique/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE