Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 616
Filter
1.
Medicine (Baltimore) ; 103(36): e39630, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39252248

ABSTRACT

RATIONALE: Cytokine release syndrome (CRS) is a common adverse event of chimeric antigen receptor T (CAR-T) cell therapy. CRS is generally a systemic inflammatory reaction, but in rare cases, it can occur in specific body areas and is referred to as "local CRS (L-CRS)." A case of laryngeal edema due to L-CRS that required tracheal intubation because of the lack of response to tocilizumab (TCZ) and dexamethasone (DEX) is reported. PATIENT CONCERNS: A 67-year-old woman with relapsed transformed follicular lymphoma was treated with CAR-T cell therapy. Although she had been given TCZ and DEX for CRS, neck swelling appeared on day 4 after infusion. DIAGNOSES: Laryngoscopy showed severe laryngeal edema, which was presumed to be due to L-CRS, since there were no other apparent triggers based on history, physical examination, and computed tomography. INTERVENTIONS: Tracheal intubation was performed because of the risk of upper airway obstruction. Ultimately, 4 doses of tocilizumab (8 mg/kg) and 6 doses of dexamethasone (10 mg/body) were required to improve the L-CRS. OUTCOMES: On day 7, laryngeal edema improved, and the patient could be extubated. LESSONS: The lessons from this case are, first, that CAR-T cell therapy may induce laryngeal edema in L-CRS. Second, TCZ alone may be ineffective in cervical L-CRS. Third, TCZ, as well as DEX, may be inadequate. In such cases, we should recognize L-CRS and manage it early because it may eventually progress to laryngeal edema that requires securing the airway.


Subject(s)
Intubation, Intratracheal , Laryngeal Edema , Lymphoma, Follicular , Humans , Female , Aged , Laryngeal Edema/etiology , Laryngeal Edema/therapy , Intubation, Intratracheal/methods , Intubation, Intratracheal/adverse effects , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/therapy , Lymphoma, Follicular/complications , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cytokine Release Syndrome/etiology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects
2.
Medicine (Baltimore) ; 103(36): e38659, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39252328

ABSTRACT

RATIONALE: Anti-CD19 chimeric antigen receptor T-cell (CAR-T) therapy is a successful treatment for B-cell malignancies associated with cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Cardiovascular toxicities have also been reported in this setting. However, there is scarce data regarding development of autonomic disorders after CAR-T cell therapy. PATIENT CONCERNS: We report a case with a patient with non-Hodgkin B-cell lymphoma, refractory to 2 prior lines of immunochemotherapy, treated with CAR-T therapy. DIAGNOSES: Orthostatic hypotension secondary to autonomic dysfunction was diagnosed as manifestation of ICANS. INTERVENTIONS: The patient received metilprednisolone 1000 mg IV daily for 3 days and anakinra 100 mg IV every 6h. OUTCOMES: The vast majority of autonomic symptoms ceased and 4 months after CAR-T therapy, autonomic dysfunction was resolved. LESSONS: New-onset autonomic dysfunction can occur as manifestation of ICANS in patients who experience persistent neurologic and cardiovascular symptoms after resolution of acute neurotoxicity and should be early recognized. Differences in differential diagnosis, mechanisms and treatment approaches are discussed.


Subject(s)
Autonomic Nervous System Diseases , Humans , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/diagnosis , Immunotherapy, Adoptive/adverse effects , Male , Cytokine Release Syndrome/etiology , Middle Aged , Lymphoma, B-Cell/complications , Lymphoma, B-Cell/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/diagnosis , Hypotension, Orthostatic/etiology , Hypotension, Orthostatic/diagnosis , Methylprednisolone/therapeutic use
3.
Front Immunol ; 15: 1422591, 2024.
Article in English | MEDLINE | ID: mdl-39253080

ABSTRACT

Malignant tumors of the hematologic system have a high degree of malignancy and high mortality rates. Chimeric antigen receptor T cell (CAR-T) therapy has become an important option for patients with relapsed/refractory tumors, showing astonishing therapeutic effects and thus, it has brought new hope to the treatment of malignant tumors of the hematologic system. Despite the significant therapeutic effects of CAR-T, its toxic reactions, such as Cytokine Release Syndrome (CRS) and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS), cannot be ignored since they can cause damage to multiple systems, including the cardiovascular system. We summarize biomarkers related to prediction, diagnosis, therapeutic efficacy, and prognosis, further exploring potential monitoring indicators for toxicity prevention. This review aims to summarize the effects of CAR-T therapy on the cardiovascular, hematologic, and nervous systems, as well as potential biomarkers, and to explore potential monitoring indicators for preventing toxicity, thereby providing references for clinical regulation and assessment of therapeutic effects.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cytokine Release Syndrome/prevention & control , Cytokine Release Syndrome/etiology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Biomarkers , Animals , Receptors, Chimeric Antigen/immunology , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Neoplasms/therapy , Neoplasms/immunology
4.
J Hematol Oncol ; 17(1): 61, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107847

ABSTRACT

Autologous anti-CD19 chimeric antigen receptor (CAR) T cells are now used in routine practice for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Severe (grade ≥ 3) cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS) are still the most concerning acute toxicities leading to frequent intensive care unit (ICU) admission, prolonging hospitalization, and adding significant cost to treatment. We report on the incidence of CRS and ICANS and the outcomes in a large cohort of 925 patients with LBCL treated with axicabtagene ciloleucel (axi-cel) or tisagenlecleucel (tisa-cel) in France based on patient data captured through the DESCAR-T registry. CRS of any grade occurred in 778 patients (84.1%), with 74 patients (8.0%) with grade 3 CRS or higher, while ICANS of any grade occurred in 375 patients (40.5%), with 112 patients (12.1%) with grade ≥ 3 ICANS. Based on the parameters selected by multivariable analyses, two independent prognostic scoring systems (PSS) were derived, one for grade ≥ 3 CRS and one for grade ≥ 3 ICANS. CRS-PSS included bulky disease, a platelet count < 150 G/L, a C-reactive protein (CRP) level > 30 mg/L and no bridging therapy or stable or progressive disease (SD/PD) after bridging. Patients with a CRS-PSS score > 2 had significantly higher risk to develop grade ≥ 3 CRS. ICANS-PSS included female sex, low level of platelets (< 150 G/L), use of axi-cel and no bridging therapy or SD/PD after bridging. Patients with a CRS-PSS score > 2 had significantly higher risk to develop grade ≥ 3 ICANS. Both scores were externally validated in international cohorts of patients treated with tisa-cel or axi-cel.


Subject(s)
Antigens, CD19 , Cytokine Release Syndrome , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Male , Female , Middle Aged , Antigens, CD19/immunology , Prognosis , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Cytokine Release Syndrome/etiology , Aged , Adult , Neurotoxicity Syndromes/etiology , Biological Products/therapeutic use , Biological Products/adverse effects , France , Aged, 80 and over , Receptors, Antigen, T-Cell
5.
Adv Exp Med Biol ; 1448: 103-119, 2024.
Article in English | MEDLINE | ID: mdl-39117810

ABSTRACT

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.


Subject(s)
Cytokine Release Syndrome , Genetic Predisposition to Disease , Lymphohistiocytosis, Hemophagocytic , Humans , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/therapy , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/etiology
6.
Adv Exp Med Biol ; 1448: 59-71, 2024.
Article in English | MEDLINE | ID: mdl-39117808

ABSTRACT

In the past two decades, there has been a great deal of work aimed to devise diagnostic guidelines, classification criteria, and diagnostic scores for cytokine storm syndromes (CSSs). The most notable effort has been the large-scale multinational study that led to the development of the 2016 classification criteria for macrophage activation syndrome (MAS) complicating systemic juvenile idiopathic arthritis (JIA). Future studies should scrutinize the validity of the proposed criteria, particularly in systemic JIA patients treated with biologics, in children with subtle or incomplete forms of MAS, and in patients with MAS complicating other rheumatologic disorders. More generic CSS criteria are also available but often lack sensitivity and specificity in a wide variety of patient populations and CSSs of different etiologies. The coronavirus disease 2019 (COVID-19)-related lung disease led to an evolution of the concept of a "cytokine storm." Emerging and unsolved challenges in the diagnosis of the different forms of CSSs highlight the need for diagnostic tools and well-established classification criteria to enable timely recognition and correct classification of patients.


Subject(s)
COVID-19 , Cytokine Release Syndrome , Humans , COVID-19/immunology , COVID-19/diagnosis , COVID-19/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/etiology , Macrophage Activation Syndrome/diagnosis , Macrophage Activation Syndrome/classification , Macrophage Activation Syndrome/immunology , Arthritis, Juvenile/diagnosis , Arthritis, Juvenile/classification , Arthritis, Juvenile/immunology , Arthritis, Juvenile/drug therapy , SARS-CoV-2/immunology , Child , Cytokines/metabolism
7.
Adv Exp Med Biol ; 1448: 161-171, 2024.
Article in English | MEDLINE | ID: mdl-39117814

ABSTRACT

Cytokine storm syndromes (CSSs) are caused by a dysregulated host immune response to an inciting systemic inflammatory trigger. This maladaptive and harmful immune response culminates in collateral damage to host tissues resulting in life-threatening multisystem organ failure. Knowledge of the various immune cells that contribute to CSS pathogenesis has improved dramatically in the past decade. Monocytes, dendritic cells, and macrophages, collective known as monocytic phagocytes, are well-positioned within the immune system hierarchy to make key contributions to the initiation, propagation, and amplification of the hyperinflammatory response in CSS. The plasticity of monocytic phagocytes also makes them prime candidates for mediating immunoregulatory and tissue-healing functions in patients who recover from cytokine storm-mediated immunopathology. Therefore, approaches to manipulate the myriad functions of monocytic phagocytes may improve the clinical outcome of CSS.


Subject(s)
Cytokine Release Syndrome , Monocytes , Phagocytes , Humans , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/etiology , Monocytes/immunology , Phagocytes/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Macrophages/immunology , Dendritic Cells/immunology
8.
Adv Exp Med Biol ; 1448: 269-274, 2024.
Article in English | MEDLINE | ID: mdl-39117820

ABSTRACT

HIV infection triggers an inflammatory response that manifests as acute retroviral syndrome (ARS) in most individuals infected by HIV. While this syndrome is usually self-limited, primary HIV infection sometimes triggers a fulminant inflammatory process consistent with cytokine storm syndrome (CSS). Many of the key findings of CSS including fever, splenomegaly, and cytopenias are routinely observed in ARS, suggesting CSS may be under recognized in the setting of acute HIV infection. Unlike other CSS scenarios, ARS-associated CSS generally responds well to HIV-targeted therapies. Advanced HIV infection is also associated with CSS, although typically this involves additional infectious insults. Occasionally, HIV therapy results in rapid recovery of the immune response that evolves into CSS.


Subject(s)
Cytokine Release Syndrome , HIV Infections , Humans , HIV Infections/complications , HIV Infections/immunology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/etiology , Cytokines
9.
Adv Exp Med Biol ; 1448: 285-291, 2024.
Article in English | MEDLINE | ID: mdl-39117822

ABSTRACT

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.


Subject(s)
Cytokine Release Syndrome , Humans , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/microbiology , Cytokine Release Syndrome/etiology , Animals , Bacterial Zoonoses/microbiology , Lymphohistiocytosis, Hemophagocytic/microbiology , Lymphohistiocytosis, Hemophagocytic/immunology , Zoonoses/microbiology
10.
Adv Exp Med Biol ; 1448: 365-383, 2024.
Article in English | MEDLINE | ID: mdl-39117827

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-1ß in both syndromes, the early use of IL-1 blockers in refractory KD with CSS deserves consideration.


Subject(s)
Cytokine Release Syndrome , Mucocutaneous Lymph Node Syndrome , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/drug therapy , Humans , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/drug therapy , Immunoglobulins, Intravenous/therapeutic use , Child , Cytokines/metabolism
11.
Adv Exp Med Biol ; 1448: 323-353, 2024.
Article in English | MEDLINE | ID: mdl-39117825

ABSTRACT

The cytokine storm syndrome (CSS) associated with systemic juvenile idiopathic arthritis (sJIA) has widely been referred to as macrophage activation syndrome (MAS). In this chapter, we use the term sJIA-associated CSS (sJIA-CSS) when referring to this syndrome and use the term MAS when referencing publications that specifically report on sJIA-associated MAS.


Subject(s)
Arthritis, Juvenile , Cytokine Release Syndrome , Humans , Arthritis, Juvenile/complications , Arthritis, Juvenile/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Macrophage Activation Syndrome/diagnosis , Macrophage Activation Syndrome/etiology , Cytokines/metabolism , Child
12.
Adv Exp Med Biol ; 1448: 355-364, 2024.
Article in English | MEDLINE | ID: mdl-39117826

ABSTRACT

Systemic lupus erythematosus (SLE) is the prototype of autoimmune diseases and can manifest with a plethora of clinical signs and symptoms associated with a myriad of laboratory abnormalities. An infrequent but potentially lethal complication of SLE is macrophage activation syndrome (MAS). The diagnosis of MAS in SLE can be very challenging due to similarities in presentation of both flares and infections, such as fever, lymphadenopathy, splenomegaly, and cytopenias. These aggravating factors contribute to the increased risk of poor outcomes in SLE-associated MAS. Indeed, at the moment MAS remains invariably lethal if untreated and still has a high mortality rate with treatment. In this chapter, we discuss several aspects of MAS in the context of SLE and in particular, the pathogenesis of MAS in SLE, how MAS presents in pediatric versus adult SLE, and, finally, MAS treatment in SLE and future directions.


Subject(s)
Lupus Erythematosus, Systemic , Macrophage Activation Syndrome , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/complications , Humans , Macrophage Activation Syndrome/diagnosis , Macrophage Activation Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/etiology , Cytokines/metabolism
13.
Adv Exp Med Biol ; 1448: 429-440, 2024.
Article in English | MEDLINE | ID: mdl-39117831

ABSTRACT

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.


Subject(s)
Hematologic Neoplasms , Lymphohistiocytosis, Hemophagocytic , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/therapy , Humans , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Neoplasms/immunology , Neoplasms/therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/etiology , Immune Checkpoint Inhibitors/therapeutic use
14.
Adv Exp Med Biol ; 1448: 497-522, 2024.
Article in English | MEDLINE | ID: mdl-39117836

ABSTRACT

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.


Subject(s)
Cytokine Release Syndrome , Disease Models, Animal , Lymphohistiocytosis, Hemophagocytic , Animals , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/pathology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/etiology , Mice , Humans , Cytokines/metabolism
15.
Adv Exp Med Biol ; 1448: 441-457, 2024.
Article in English | MEDLINE | ID: mdl-39117832

ABSTRACT

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.


Subject(s)
Cytokine Release Syndrome , Lymphohistiocytosis, Hemophagocytic , Multiple Organ Failure , Sepsis , Humans , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Sepsis/immunology , Sepsis/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/etiology , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/etiology , Macrophage Activation Syndrome/immunology , Macrophage Activation Syndrome/etiology , Killer Cells, Natural/immunology
16.
Adv Exp Med Biol ; 1448: 525-551, 2024.
Article in English | MEDLINE | ID: mdl-39117837

ABSTRACT

Etoposide has revolutionized the treatment of primary as well as secondary hemophagocytic lymphohistiocytosis (HLH), and it is, together with corticosteroids, the most widely used therapy for HLH. In the early 1980s, long-term survival in primary HLH was <5% but with the etoposide-/dexamethasone-based protocols HLH-94 and HLH-2004, in combination with stem cell transplantation, 5-year survival increased dramatically to around 60% in primary HLH, and based on analyses from the HLH-2004 study, there is likely room for further improvement. Biologically, etoposide administration results in potent selective deletion of activated T cells as well as efficient suppression of inflammatory cytokine production. Moreover, etoposide has also been reported to promote programmed cell death (apoptosis) rather than proinflammatory lytic cell death (pyroptosis), conceivably ameliorating subsequent systemic inflammation, i.e., a treatment very suitable for cytokine storm syndromes (CSS). The combination of etoposide and corticosteroids may also be beneficial in cases of severe or refractory secondary HLH (sHLH) with imminent organ failure, such as infection-associated HLH caused by Epstein-Barr virus (EBV) or malignancy-triggered HLH. In CSS associated with rheumatic diseases (macrophage activation syndrome, MAS or MAS-HLH), etoposide is currently used as second- or third-line therapy. Recent studies suggest that etoposide perhaps should be part of an aggressive therapeutic intervention for patients with severe refractory or relapsing MAS, in particular if there is CNS involvement. Importantly, awareness of sHLH must be further increased since treatment of sHLH is often delayed, thereby missing the window of opportunity for a timely, effective, and potentially life-saving HLH-directed treatment.


Subject(s)
Cytokine Release Syndrome , Etoposide , Lymphohistiocytosis, Hemophagocytic , Humans , Etoposide/therapeutic use , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Cytokines/metabolism , Animals
17.
Adv Exp Med Biol ; 1448: 573-582, 2024.
Article in English | MEDLINE | ID: mdl-39117840

ABSTRACT

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.


Subject(s)
Cytokine Release Syndrome , Immunity, Innate , Interferon-gamma , Lymphohistiocytosis, Hemophagocytic , Humans , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Interferon-gamma/immunology , Animals , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Immunity, Innate/drug effects , Adaptive Immunity/drug effects
18.
Adv Exp Med Biol ; 1448: 611-622, 2024.
Article in English | MEDLINE | ID: mdl-39117843

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) can be considered as a severe cytokine storm syndrome disorder. HLH typically manifests as a life-threatening inflammatory syndrome characterized by fevers, cytopenias, hepatosplenomegaly, and various other accompanying manifestations such as coagulopathy, hepatitis or liver failure, seizures or altered mental status, and even multi-organ failure. Standard up-front treatments do not always bring HLH into remission or maintain adequate response, and salvage or alternative therapies are often needed. For patients with genetic diseases that cause HLH, curative allogeneic hematopoietic cell transplantation is usually offered to prevent future episodes of life-threatening HLH. Here, we will discuss the options and approaches for salvage therapy and hematopoietic cell transplantation for patients with HLH.


Subject(s)
Cytokine Release Syndrome , Hematopoietic Stem Cell Transplantation , Lymphohistiocytosis, Hemophagocytic , Salvage Therapy , Humans , Lymphohistiocytosis, Hemophagocytic/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Cytokine Release Syndrome/therapy , Cytokine Release Syndrome/etiology , Salvage Therapy/methods , Transplantation, Homologous
20.
Intensive Care Med ; 50(9): 1459-1469, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39172238

ABSTRACT

The use of chimeric antigen receptor T (CAR-T) cells is a significant therapeutic improvement increasing the prognosis for patients with a variety of hematological malignancies. However, this therapy has also sometimes life-threatening, complications. Therefore, knowledge of the treatment and management of these complications, especially in treatment centers and intensive care units, respectively, is of outstanding importance. This review provides recommendations for the diagnosis, management, and treatment of CAR-T cell-associated complications such as cytokine release syndrome, immune effector cell associated neurotoxicity syndrome, hematotoxicity, hypogammaglobulinemia, and CAR-T cell-induced pseudo-progression amongst others for physicians treating patients with CAR-T cell-associated complications and intensivists.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Neurotoxicity Syndromes/etiology
SELECTION OF CITATIONS
SEARCH DETAIL