Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Aug 1.
Published in final edited form as: Trends Immunol. 2021 Jul 1;42(8):681–705. doi: 10.1016/j.it.2021.06.001

The ‘Cytokine Storm’: molecular mechanisms and therapeutic prospects

Rajendra Karki 1, Thirumala-Devi Kanneganti 1,
PMCID: PMC9310545  NIHMSID: NIHMS1715232  PMID: 34217595

Abstract

Cytokine storm syndrome has generally been described as a collection of clinical manifestations resulting from an overactivated immune system. Cytokine storms (CSs) are associated with various pathologies, as observed in infectious diseases, certain acquired or inherited immunodeficiencies and autoinflammatory diseases, or following therapeutic interventions. Despite the role of CS in tissue damage and multiorgan failure, a systematic understanding of its underlying molecular mechanisms is lacking. Recent studies demonstrate a positive feedback loop between cytokine release and cell death pathways; certain cytokines, PAMPs, and DAMPs, can activate inflammatory cell death, leading to further cytokine secretion. Here, we discuss recent progress in innate immunity and inflammatory cell death, providing insights into the cellular and molecular mechanisms of CSs and therapeutics that might quell ensuing life-threatening effects.

The cytokine storm and cell death

The term ‘cytokine storm’ (CS) has been increasingly used in both the scientific literature and public media to denote an out-of-control inflammatory response, although a specific molecular definition to delineate CS from normal inflammation and to describe the amounts and types of cytokines involved has remained elusive. CS was first used in the early 1990s to describe the effects of graft-versus-host disease (GvHD) [1, 2] and later in the infectious disease setting in the early 2000s in studies of cytomegalovirus (CMV) [3], Epstein–Barr virus (EBV)-associated hemophagocytic lymphohistiocystosis (HLH) [4], group A streptococcus (GAS) [5], influenza virus [6], variola virus [7] and severe acute respiratory syndrome coronavirus (SARS-CoV) [8]. The CS was also described in cases of human H5N1 avian influenza virus infection during the 2005 ‘bird flu’ outbreak [9]. Since then, the term CS has been steadily gaining scientific and public interest, with the ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by the SARS-CoV-2 virus, further fueling attention.

Although the CS has been best studied in infectious diseases and sepsis, it has also been described in non-infectious settings. For example, it is known as macrophage activation syndrome (MAS) when associated with rheumatic diseases such as systemic lupus erythematosus (SLE) and systemic juvenile idiopathic arthritis (sJIA) [10]. The CS also has a role in hyperinflammatory diseases such as inherited and non-inherited forms of HLH [11]. Furthermore, CSs have been reported in response to therapeutic interventions, such as GvHD during hematopoietic stem cell transplantation (HSCT) or hyperinflammation following the administration of immunotherapeutic agents, including antibodies and chimeric antigen receptor (CAR) T cells [1214]. However, despite the clinically important incidence of CSs under diverse conditions, little is known about the mechanisms underlying how they are initiated or perpetuated. Understanding the physiological basis of CSs is crucial to providing new potential insights into fine-tuning immune responses to treating these life-threatening conditions.

Many of the clinical features of the CS, which include fever, hepatosplenomegaly, progressive liver failure with coagulopathy, cytopenias, and hyperferritinemia [15] (Table 1), can be explained by the effects of common pro-inflammatory cytokines, such as IL-1, IL-6, interferon-γ (IFN-γ), tumor necrosis factor (TNF), and IL-18 [16]. Amounts of these cytokines are increased in the circulation of most patients with CS syndrome (CSS; also known as cytokine shock) [15]. Increasing clinical evidence has described links between the pathogenesis of a CS and programmed cell death (PCD) processes. For example, during CSS in patients with sepsis, there is marked cell death of immune cells such as B and T cells in lymphoid organs [17]. Blocking lymphocyte cell death improves survival in a mouse model of sepsis, which suggests that cell death may play a pathogenic role during CSS [18, 19]. Indeed, autopsies of patients with sepsis showed that the death of immune cells was one of the underlying causes of mortality [20]. The cell death involved in CSS was historically characterized as apoptosis, but despite the favorable results of using apoptosis inhibitors in animal models of CSS (such as sepsis), translation of these therapies to the clinic has met with limited success [21]. These limitations suggest that PCD pathways other than apoptosis might also be involved in driving certain pathological features of a CS.

Table 1 |.

Clinical manifestations in cytokine shock syndromes[16]

Constitutional Symptoms Nervous System Gastro-intestinal System Vascular/Lymphatic System Rheumatologic System
• Fever
• Headache
• Fatigue
• Anorexia
• Confusion
• Delirium
• Ataxia
• Seizures
• Anosmia
• Nausea
• Vomiting
• Diarrhea
• Abdominal pain
• Anemia, cytopenia
• Coagulopathy
• Increased ferritin, cytokines, CRP and D-dimer
• Hemorrhage, stroke
• Lymphadenopathy
• Endothelial damage
• Vasculitis
• Arthritis
• Arthralgia
Heart Skin Lungs Liver Kidney
• Hypotension
• Arrhythmias
• Cardiomyopathy
• Ischemia
• Cardiogenic shock
• Urticaria
• Rash
• Edema
• Vesicles
• Pneumonitis
• Pulmonary edema
• Dyspnea
• Hypoxemia
• ARDS
• Hepatomegaly
• Elevated bilirubin
• Liver failure
• Increased AST, ALT and LDH
• Acute kidney injury
• Proteinuria
• Hematuria
• Kidney failure

Recent studies have elucidated the molecular mechanisms underlying synergies between three innate immune-mediated PCD pathways — inflammasome-dependent pyroptosis, apoptosis, and necroptosis; together, these pathways provide a framework for molecularly defining the CS. We refer to the crosstalk between these cytokine-mediated inflammatory PCD pathways as PANoptosis—a PCD activated by pathogens such as influenza A virus (IAV), cytokines, or by sterile triggers such as cancer cells, and regulated by the PANoptosome (a molecular scaffold enabling key molecules from the pyroptosis, apoptosis, and necroptosis pathways to contemporaneously engage) [22]. Studies have shown that cytokines are intricately linked to cell death mechanisms and involved in a positive feedback loop whereby cytokine release causes inflammatory cell death that facilitates further pathogenic cytokine release through membrane pores and cell lysis, culminating in a CS to drive severe, life-threatening damage to host tissues and organs [23]. Indeed, systemic inflammation, tissue damage, multiorgan failure and mortality in CSS are prevented by combined treatments with TNF and IFN-γ neutralizing antibodies, blocking cytokine-mediated inflammatory cell death in murine models of CSs (such as poly(I:C)induced HLH, LPS-induced sepsis, and SARS-CoV-2 infection) [23]. The identification of cell death-associated molecules, primarily caspase-8, Z-DNA-binding protein 1 (ZBP1), transforming growth factor-β-activated kinase (TAK1), and receptor-interacting serine/threonine protein kinase 1 (RIPK1), as master switches of inflammasome activation and of PCD pathways [24-31], has further established the concept of cytokine-mediated inflammatory cell death in infection, inflammatory disease, and cancer [3234]. Understanding this cytokine-cell death positive feedback loop through which cytokines may cause inflammatory cell death to drive further cytokine release, CSs, organ damage, and lethality, is crucial for more effectively treating and possibly preventing CSS during diseases.

In this review, we discuss the mechanisms that initiate and perpetuate the CS. We briefly describe the stimuli that can trigger a CS. Next, we detail a few landmark studies that have elucidated molecular pathways of inflammatory cell death in association with the CS, systemic inflammation, tissue damage, and multiorgan failure. Finally, we discuss therapeutic interventions harboring the potential to mitigate CS-mediated life-threatening inflammation. An improved mechanistic understanding of the CS can inform the development of novel, candidate therapeutic strategies to treat numerous fatal diseases associated with CS such as sepsis, HLH, and the ongoing COVID-19 pandemic.

Stimuli that trigger the CS

Multiple stimuli can result in CSS with largely similar clinical manifestations (Table 2). Based on the source of the stimulus, a CS can be broadly classified into pathogen-induced CS (sepsis), autoinflammatory or monogenic CS, or therapeutic intervention-induced CS. Understanding the underlying mechanisms linking these stimuli to the CS is important to identify the molecular pathways involved.

Table 2 |.

Different forms of CSS: contributing cells and driving cytokines in humans and murine models

Cytokine Storm Syndromes (CSS) Cause(s) Contributing cells Primary driving cytokines References
Pathogen-induced
Bacterial sepsis GAS, Staphylococcus aureus, Francisella tularensis, Klebsiella pneumoniae Yersinia pestis Heterogenous Multiple cytokines, particularly TNF and IFN-γ [37–41]
Viral sepsis IAV, Ebola virus, SARS-CoV, MERS-CoV, SARS-CoV-2 Heterogenous Multiple cytokines, particularly TNF and IFN-γ [45]
Autoinflammatory and monogenic
pHLH Mutations in PRF1, UNC13D, STX11, STXBP2 CD8+ T cells IFN-γ [49–55]
sHLH Epstein-Barr virus, CMV CD8+ T cells, myeloid cells IFN-γ, IL-1 [3, 4]
CAPS NLRP3 mutation Myeloid cells IL-1β [140]
Familial Mediterranean fever MEFV mutation Myeloid cells IL-1β [171]
NLRC4-MAS NLRC4 mutation Myeloid cells IL-18 [142]
Therapeutic intervention-induced
CAR T-cell therapy Infusion of CAR T cells CAR T cells Myeloid cells Tumor cells IL-6, IL-1 [139]
Blinatumomab treatment Infusion of T-cell receptor-engaging antibodies: anti-CD19 and anti-CD3 T cells Myeloid cells Tumor cells IL-6 [13]
Allogeneic stem cell transplantation N/A T cells N/A [172]

CAPS, cryopyrin-associated periodic syndrome; CAR, chimeric antigen receptor; CMV, cytomegalovirus; GAS, group A streptococcus; IAV, influenza A virus; MAS, macrophage activation syndrome; pHLH, primary hemophagocytic lymphohistiocystosis; sHLH, secondary hemophagocytic lymphohistiocystosis.

Pathogen-induced CS.

Infection-associated CS or sepsis is the leading cause of death globally for patients in the intensive care unit (ICU) [35]. According to the World Health Organization, sepsis kills around 11 million people each year [36], and this life-threatening condition has been primarily studied in the context of bacterial and viral respiratory infections (Table 2).

For example, bacterial infections with Streptococcus pyogenes, Staphylococcus aureus, Francisella tularensis, Klebsiella pneumoniae and Yersinia pestis result in a systemic inflammatory response in humans and mice [3741]. These bacteria usually trigger NF-κB signaling and the activation of interferon regulatory factor 3 (IRF3) and IRF7 in mammalian immune cells by binding to pattern recognition receptors (PRRs) [42]. They can also drive inflammasome activation, particularly in innate immune cells [43]. Together, this signaling pathway induces the production of several pro-inflammatory cytokines, including IFN-γ, TNF, IL6, IL-1β and IL-18 in immune cells [43] (FIG. 1). However, only a subset of patients with sepsis develops CSS, while others develop immunoparalysis. Patients who survive the initial CS but subsequently die may be those who do not recover from immunoparalysis [44].

Figure 1 |. Signaling pathways producing pro-inflammatory cytokines.

Figure 1 |

a) The cartoon depicts the recognition of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by pattern recognition receptors (PRRs) such as the Toll-like receptors (TLRs), NOD-like receptors (NLRs), C-type lectin receptors (CLRs), RIG-I–like receptors (RLRs), or the cGAS-STING axis, engaging NF-κB, MAPK, and interferon regulatory factor 3 (IRF3)–IRF7 activation for the transcription of pro-inflammatory cytokine genes [42, 43, 89]. Fungal ligands bind the CLR dectin 1 and signal through the dectin 1–SYK pathway to activate NF-kB signaling. TLR signaling through MYD88 activates the NF-kB pathway, whereas TRIF-dependent TLR signaling primarily involves IRF3 and IRF7 activation. The depiction is based on results from human and murine studies. b) Inflammasome sensors can interact directly with their target ligand or respond to a variety of physiological changes. This leads to the recruitment of apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) and caspase 1 (CASP1) to form an inflammasome complex. CASP1 then undergoes proximity-induced autocatalytic cleavage to take on its active form. Activated CASP1 cleaves gasdermin D (GSDMD) to release the N-terminal domain, which oligomerizes in the plasma membrane and forms pores, thereby inducing cell death through pyroptosis. Activated CASP1 also cleaves pro–interleukin (IL)-1β and pro–IL-18 into their active forms, which are released through the GSDMD pores [43]. The depiction is based on results from human and murine studies. MAVS, mitochondria antiviral signaling; MDA5, melanoma differentiation-associated protein 5.

In the case of viral infections, sepsis has been widely studied in the context of IAV, Ebola virus, and coronaviruses. IAV remains a major threat to public health and has caused four pandemics in the 20th century, most notably in 1918–1919, with the largest known outbreak of any infectious disease to date. An exaggerated immune response contributed to the lethality of the 1918 IAV pandemic. In addition, human-infecting coronaviruses (CoVs), including SARS-CoV, MERS-CoV, and SARS-CoV-2, trigger robust inflammatory responses that can contribute to pathology in humans and mice [45]. The pro-inflammatory genes that are upregulated in innate immune cells in patients with severe or critical COVID-19 are located primarily downstream of the NF-κB and type I IFN signaling pathways [46, 47]. Peripheral blood mononuclear cells (PBMCs) obtained from healthy donors and subsequently infected with SARS-CoV-2 have shown increased production of pro-inflammatory cytokines, including TNF, IL-6, IFN-γ, and IL1β in vitro [23]. Several studies analyzing cytokine profiles from patients with severe COVID-19 have suggested a direct correlation between a CS and lung injury, multiple organ failure, and unfavorable prognosis [48] (Box 1). In particular, TNF and IFΝ-γ are highly upregulated in the serum of patients with severe COVID-19, and as discussed later, TNF- and IFN-γ–induced cell death contribute to systemic inflammation, tissue damage, acute respiratory distress syndrome (ARDS), multiorgan failure, and mortality in COVID-19 [23].

BOX-1: The Cytokine Storm in COVID-19.

Although CSS-inducing agents differ in fundamental respects, the interactions of PAMPs or DAMPs with PRRs evoke similar cytokine profiles. Studies have now compared the cytokine profiles in patients with COVID-19 with those experiencing other types of CSS, leading some to suggest that CSS does not occur in COVID-19 [222224]. A meta-analysis comparing inflammatory cytokine profiles reported lower amounts of pro-inflammatory cytokines, particularly IL-6, in the serum of patients with COVID-19 compared to patients with sepsis or CAR T cell-induced CSS [222]. Also, RNA sequencing analysis of peripheral blood mononuclear cells (PBMCs) from patients with influenza A virus (IAV) infection or with COVID-19 identified lower cytokine expression in PBMCs of patients with COVID-19 [223]. However, a major limitation of these studies is that the authors draw conclusions from comparisons between patients with COVID-19 and other CSS-associated diseases, but not healthy subjects [222, 223]. By contrast, another similar study found increased amounts of pro-inflammatory cytokines, particularly TNF and IL-1, being expressed from PBMCs taken from patients with COVID-19 compared to patients with IAV infection [225]. Additionally, cytokines have not been measured simultaneously on the same platform across diseases with CSS. Moreover, analyzing cytokines at just one time point may not be indicative of the full disease course. There are several other factors that influence the analysis of cytokine measurement, including time and compartment of sample collection, underlying diseases or polymorphisms, as well as previous pathogen exposure and immunosuppression. Indeed, a longitudinal analysis of the host response to SARS-CoV-2 and IAV in ferrets found increased amounts of IL-6, IL-1 and other chemokines in the cells obtained from nasal wash of SARS-CoV-2–infected ferrets compared to those in IAV-infected ferrets 7 days after infection, supporting the idea that a CS does occur in COVID-19 [224]. So far, most studies have focused on direct measurements of cytokines in the peripheral blood compartment and have failed to interrogate the cytokine profile in local tissues such as the lung. All of these points should be taken into consideration when drawing conclusions about the occurrence of a CS in COVID-19 patients.

Autoinflammatory or monogenic CS.

Monogenic abnormalities leading to autoinflammation can also cause CSS. Primary HLH (pHLH) is a classic example of monogenic CSS (Table 2). pHLH, also known as familial HLH, is a rare hereditary disease of immune dysregulation caused by one of several underlying genetic mutations that primarily affect T cell and NK cell activity in infants and young children [4951]. Mutations in the PRF1, UNC13D, STX11 and STXBP2 genes --crucial for granule-dependent cytolytic functions of T and NK cells-- predispose children to pHLH [5255]. Dysregulation of the perforin-dependent cytolytic pathway by which antigen-presenting cells (APCs) are eliminated by cytotoxic lymphocytes (CTLs) results in excessive production of pro-inflammatory cytokines, particularly IFN-γ, and the overactivation of NK cells and CTLs [53]. However, disease onset, severity and incidence in pHLH are highly variable, depending on the genes involved and the types of mutations present [56].

Additionally, HLH can develop without the aforementioned germline mutations as a result of infection, certain autoinflammatory conditions. or malignancies -- referred to as secondary HLH (sHLH), or acquired HLH[4]. Although various infectious agents have been linked to sHLH, viruses --particularly EBV-- remain the most common triggers [4]. However, there is currently no clear distinction between pHLH and sHLH, as children with pHLH often present with coinfections [57]. The crucial role of T cells in the development of HLH is exemplified by a mouse model in which IFN-γ produced by CD8+ T cells contributed to hematological abnormalities in perforin-deficient mice infected with lymphocytic choriomeningitis virus (LCMV) [5860]. However, despite their marked deficiency of T cells and NK cells, patients with severe combined immunodeficiency can still develop HLH, suggesting that the pathogenesis of HLH may not always involve T cells and NK cells [61].

Germline mutations in genes regulating the innate immune system and inflammasome signaling are also associated with CSS. Mutation or dysregulation of PRRs or molecules involved in inflammatory signaling and cell death pathways can lead to the secretion of large quantities of cytokines, contributing to the pathology of several diseases such as Familial Mediterranean fever (Table 2). The specific mechanisms through which genes involved in cell death pathways play roles in driving CSS (e.g. those encoding inflammasome components, ZBP1, caspase-8, FADD, RIPK3 and RIPK1) are discussed below.

Therapeutic intervention-induced CS.

Therapy induced-cytokine release and cell death are crucial to eliminating cancer; however, they can also cause a CS. CAR T cell therapy has been a major innovation in cancer immunotherapy. Although the majority of patients achieve clinical remission following treatment with CAR T cells, a sizable proportion of patients develop CSS, with manifestations ranging from fever and hypotension, to hypoxia and organ failure [12]. It is commonly thought that CSS in these patients occurs due to the activation of bystander cells, particularly myeloid cells, by CAR T cells in the tumor environment [12]. Subsequent inflammatory cell death and tumor lysis induce a CS through the release of inflammatory cytokines [62]. Pro-inflammatory cytokines, including IL-6, IL-1 and IFN-γ, are consistently found to be elevated in the serum of patients treated with CAR T cells who develop a CS [62]. Also, in a mouse model of CAR T cell–induced CS where mice were injected with Raji tumor cells and subsequently treated with 1928z CAR T cells, the overall amounts of pro-inflammatory cytokines (including IL-6, CCL2, G-CSF, IL-3, IFN-γ, GM-CSF and IL-2) strongly correlated with CSS severity and mortality [63]. As not all patients treated with CAR T cells develop a CS, additional factors, such as CAR design, co-morbidity, and underlying genetic polymorphisms, are likely to play a part in the generation of a CS. Additionally, to date, there have been no reported cases of CSS using NK-cell CAR therapy in cancer patients [64].

CSs can also occur in response to other T cell–engaging immunotherapies. One example is the treatment of patients with relapsed or refractory acute lymphoblastic leukemia with blinatumomab-- a bi-specific antibody that binds to CD19+ and CD3+ T cells [13]. Similarly to CAR T cells, this antibody induces T cells and macrophages to produce pro-inflammatory cytokines [13, 14]. Another earlier example of a T cell–engaging immunotherapy is the superagonist anti-CD28 monoclonal antibody TGN1412, which directly activates CD4+ effector memory T cells and which induced rapid CSS within minutes after infusion in six healthy volunteers [65]. TGN1412 stimulation of T cells results specifically in the production of IL-2 and IFN-γ, suggesting that these two cytokines are associated with TGN1412-induced CSS [66]. However, preclinical studies failed to predict the CS in human volunteers. Preclinical studies had been performed in vitro in human PBMCs, as well as in vivo in rodents using CD28SA (surrogate antibody to TG1412 and in cynomolgus macaques using TGN1412 [67]. In rodents, the activation of Tregs by CD28SA prevented the CS: deletion of regulatory T cells (Tregs) in mice resulted in a significant increase in the amount of circulating pro-inflammatory cytokines relative to controls [68]. In macaques with a CS, the failure to respond to TGN1412 was deemed to be due to lack of CD28 expression on CD4+ effector memory T cells in macaques, presumably preventing TGN1412 from activating these cells and inducing cytokine production [66]. Therefore, these preclinical models were flawed in predicting the clinical safety of TGN1412. Other antibody-based therapies have also lead to a CS. For example, infusion of the anti-CD20 monoclonal antibody rituximab has induced a CS in patients with B-cell chronic lymphocytic leukemia [69]. The targeted killing of CD20+ B cells induced by this antibody, including via apoptosis, had led to cytokine release [70]. Collectively, T cell-engaging immunotherapies and antibody-based therapies are known to induce CS in humans, and careful evaluation using optimal preclinical models is imperative in the mechanistic study of these treatments and to be able to assess their safety before putative translation to the clinic.

Molecular mechanisms of a CS

Although many different stimuli can induce CSS, in all cases, the interactions of pathogen-associated or damage-associated molecular patterns (PAMPs or DAMPs) with PRRs of the innate immune system, activate downstream molecular pathways that are conserved across stimuli and mammalian species [43]. The host innate immune response to infection or sterile insults provides the first line of defense against damage and activates major signaling pathways for the production of inflammatory cytokines and chemokines (FIG. 1). These cytokines are crucial for clearing infections and maintaining cellular homeostasis at physiological concentrations, but the dysregulated release of pro-inflammatory cytokines can result in a CS. Thus, characterizing the pathways that can result in dysregulated cytokine release is essential to robustly identifying molecular targets that might lead to novel candidate therapeutic strategies to treat CSS.

Cytokines can induce inflammatory cell death.

Among the numerous pro-inflammatory cytokines that are elevated during a CS, IL-1, IL-6, TNF and IFN-γ are of paramount importance [71]. Of these, TNF and IFN-γ have been widely studied in the context of cell death and independently induce apoptosis or necroptosis in a context-dependent manner [72, 73]. Intraperitoneal injection of TNF and IFN-γ induces mortality in mice [23, 74], mirroring the major symptoms of CSS observed in patients with COVID-19 [23]. TNF and IFN-γ are also known to cause cell death in multiple cell types, contributing to various pathological conditions such as neurological disorders (multiple sclerosis and dementia), liver damage, chronic obstructive pulmonary disease (COPD), fibrosis, and osteoporosis [23, 74]. Mechanistically, the combination of TNF and IFN-γ induces STAT1- and IRF1-dependent nitric oxide (NO) production, which in turn activates caspase-8 and inflammatory cell death, with characteristics of pyroptosis, apoptosis, and necroptosis, in murine macrophages [23] (FIG. 2a). Whereas pyroptosis is executed by gasdermin family members, apoptosis, which can also be inflammatory and lead to cytokine release, is executed by caspase-3 and caspase-7 following the activation of upstream initiator caspases (caspase-8, caspase-10 or caspase-9), and necroptosis is executed by RIPK3–mediated oligomerization of mixed lineage kinase domainlike (MLKL) in multiple cell types [75]. Activation of these cell death pathways results in the release of more cytokines and alarmins, which function as ‗danger signals’ to alert surrounding immune and stromal cells to the presence of cell/tissue damage, and to further fuel the cytokine release, culminating in CS [23, 43]. The mechanistic descriptions of how innate immunemediated inflammatory cell death in response to cytokines, PAMPs, or DAMPs, occurs and induces cytokine release (fueling the cytokine-cell death positive feedback loop), are detailed below.

Figure 2 |. Regulation of inflammatory cell death by cytokines, cell death proteins, and infection.

Figure 2 |

a) Schematic representation of inflammatory cell death induced by the combination of TNF and IFN-γ. Activation of JAK–STAT1 signaling by TNF and IFN-γ induces the upregulation of interferon regulatory factor 1 (IRF1), which in turn triggers upregulation of inducible nitric oxide synthase (iNOS) and the production of nitric oxide (NO). NO then leads to caspase 8 (CASP8) activation. Activated CASP8 induces gasdermin E (GSDME)-mediated pyroptosis, caspase-3 and caspase-7 (CASP3 and CASP7)-driven apoptosis and RIPK3 and mixed lineage kinase domain-like pseudokinase (MLKL)-mediated necroptosis [23]. The depiction is based on results from human and murine studies. b) TNF stimulation induces rapid assembly of a multiprotein signaling complex containing TNF receptor type 1 (TNFR1)-associated DEATH domain protein (TRADD) and receptor interacting protein kinase 1 (RIPK1), which subsequently recruits E3 ubiquitin ligases such as cIAP1/2 and LUBAC [99, 100]. The K63-linked ubiquitin chains generated by cIAP1/2 induce transforming growth factor β-activated kinase 1 (TAK1) recruitment and RIPK1 phosphorylation. LUBAC further conjugates the RIPK1 complex with M1linked linear ubiquitin chains [100]. The deubiquitinase A20 mediates the cleavage of K63-linked ubiquitin chains on RIPK1. Furthermore, A20 can add K48-linked ubiquitin chains to RIPK1, thereby targeting it for proteasomal degradation [101]. The phosphorylation of RIPK1 by IKKα– IKKβ, TBK1–IKKε and TAK1 maintains its prosurvival signaling [88]. Inhibition of RIPK1 phosphorylation dissociates RIPK1 from the TNFR1 complex and triggers its assembly with Fas-associated death domain protein (FADD) and CASP8, which triggers gasdermin D (GSDMD) and GSDME-mediated pyroptosis and CASP3–CASP7–mediated apoptosis [83]. Inhibition of CASP8 induces RIPK1 to phosphorylate RIPK3, thereby triggering MLKL-mediated necroptosis. The depiction is based on results from human and murine studies. c) Influenza A virus (IAV) Z-RNA is recognized by the Zα2 domain of Z-DNA binding protein 1 (ZBP1) [25, 93]. Caspase-6 (CASP6) promotes the interaction between ZBP1 and RIPK3 [92]. ZBP1 triggers NLRP3-dependent GSDMD cleavage, CASP8-dependent CASP3 and CASP7 activation, and MLKL phosphorylation to induce inflammatory cell death during IAV infection [25]. The depiction is based on results from murine studies. ASC, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain; CARD, caspase recruitment domain; DD, death domain; DED, death effector domain; FADD, fas-associated death domain protein; LRR, leucine-rich repeat; RHIM, receptor interacting protein homotypic interaction motif.

The role of RHIM domain proteins in cell death and the pathogenesis of a CS.

RIPK1, RIPK3, ZBP1, and TIR-domain-containing adapter-inducing interferon-β (TRIF), are four mammalian proteins that harbor RIP homotypic interaction motif (RHIM) domains; they are key components of signaling pathways involved in innate immune-mediated cell death in response to cytokines, PAMPs, or DAMPs, as well as in inflammation and subsequent cytokine release in CSS [7680]. The function of these proteins in CSS has been studied in mice and humans: RIPK1 is a master regulator of cytokine signaling as well as of cell survival and death [43]. For example, RIPK1 controls cytokine release in a murine model of human neutrophilic dermatosis, in which a point mutation in the protein tyrosine phosphatase, non-receptor type 6 (Ptpn6spin) causes inflammatory skin disease; irradiated mice transplanted with fetal liver cells from Ptpn6spinRipk1−/− mice are protected from disease, as are Ptpn6spinIl1a−/− mice, highlighting the important role of RIPK1 in mediating IL-1α release, driving this pathology [81]. Additionally, in response to TNF and engagement of related death receptors, RIPK1 undergoes posttranslational modifications such as ubiquitylation and phosphorylation, thereby promoting the formation of a pro-inflammatory and cell survival-mediating protein complex [82] (FIG. 2b). However, in response to defects in NF-κB signaling required for survival, RIPK1 may be subsequently released from the TNF receptor into the cytosol to nucleate other cell death molecules such as FADD, CASP8, or RIPK3, thus activating inflammatory cell death pathways [83] (FIG. 2b). Indeed, the scaffold functions of RIPK1 are known to be crucial for inhibiting systemic inflammation in mice by limiting cell death [76, 77], and loss of RIPK1 in mice leads to postnatal lethality marked by symptoms of CSS, such as systemic inflammation, severe anemia, cytopenia, and neutrophilia [77]. The lethal phenotype caused by RIPK1 deficiency is rescued in mice by co-deletion of the pro-apoptotic caspase-8 and the pro-necroptotic RIPK3, but not by individual deletions of caspase-8, RIPK3 or the downstream necroptosis effector MLKL, suggesting that there is coordination between apoptosis and necroptosis in driving the CS and lethality in the absence of RIPK1 [76, 77]. In addition, loss of TNF signaling in Ripk1−/−Tnfr1−/− mice does not rescue postnatal lethality mediated by RIPK1 deficiency [77]. Whereas Ripk1–/– mice die postnatally, mice expressing a mutant form of RIPK1 that cannot undergo caspasemediated cleavage (Ripk1D325A/D325A mice) die perinatally from systemic inflammation, showing that caspase-mediated RIPK1 cleavage is important to preventing necroptosis and allowing embryogenesis [84]. These mice can still be rescued by the combined loss of caspase-8 and RIPK3, suggesting that apoptosis and necroptosis drive lethality [84]. Moreover, heterozygous missense mutations in RIPK1 -- D324N, D324H, D324V and D324Y, which are resistant to caspase cleavage -- are associated with early-onset periodic fever syndrome and severe intermittent lymphadenopathy in humans [84, 85]. Impaired cleavage of RIPK1 D324 variants by caspase-8 sensitizes patient PBMCs to TNF-induced RIPK1 activation, which drives apoptosis and necroptosis and leads to the overproduction of inflammatory cytokines [85].

Similarly, mutation of the conserved RHIM in RIPK1 (mRHIM) causes perinatal lethality characterized by epidermal hyperplasia, dermatitis, and increased influx of neutrophils in the peritoneum, concomitant with increased cytokine concentrations in the serum of Ripk1RHIM/RHIM mice relative to wildtype mice [78, 79]. Furthermore, mice expressing RIPK1 RHIM only in keratinocytes (Ripk1RHIM/E-KO) develop skin inflammation compared to mice expressing wildtype RIPK1 (Ripk1WT/E-KO) [78]. The perinatal lethality or skin inflammation in mice expressing RIPK1 mRHIM is prevented by loss of RIPK3, MLKL, or ZBP1, which suggests that the RIPK1 RHIM mutation may primarily drive the spontaneous activation of the RIPK3–MLKL-mediated necroptosis pathway in mice [78, 79]. Immunoprecipitation assays in primary mouse embryonic fibroblasts from Ripk1RHIM/RHIM mice show enhanced ZBP1 binding with RIPK3 compared with their interaction in wildtype fibroblasts, suggesting that the RIPK1 RHIM typically prevents ZBP1 from binding and activating RIPK3 [78]. In humans, RIPK1 deficiency caused by rare homozygous mutations is associated with severe lymphopenia, recurrent infections, early-onset inflammatory bowel disease (IBD), and progressive polyarthritis [80]. Relative to healthy patients, IL-1β concentrations are increased in the supernatant of PHA-stimulated whole blood cells, or from LPS-stimulated CD14+ monocytes obtained from patients with homozygous mutations in RIPK1, leading to complete RIPK1 deficiency; these results mirror the increased inflammasome activity that is observed in RIPK1-deficient human monocyte-like THP-1 cells stimulated with LPS [80]. Increased cell death mediated by overt caspase-8 and RIPK3 activation in the absence of RIPK1 may promote NLRP3 inflammasome activation and cytokine release in primary murine macrophages, given that Pam3Cys-induced IL-1β release is prevented in Casp8LysMCre bone marrow derived macrophages (BMDM) treated with nec-1 [86]. Furthermore, LPS-stimulated RIPK1–/– THP-1 cells undergo increased necroptosis compared with RIPK1+/+ THP-1 cells [80], further highlighting the importance of RIPK1 in inflammatory cell death.

In addition to their importance in sterile systemic inflammation, RHIM proteins also play a role in activating cell death pathways in microbial infections associated with CSS. Certain bacteria induce inflammatory cell death through their PAMPs or toxic effectors, such α-hemolysin and Hla, that are secreted into host cells. For example, S. aureus releases α-hemolysin and Hla, which activate necroptosis and pyroptosis via RIPK3 and NLRP3 signaling, resulting in increased inflammatory cytokine release and severe lung pathology in mice [87]. Similarly, Yersinia pestis, a causative agent for plague that has historically led to major pandemics, inactivates TAK1 with its effector protein YopJ, inducing cell death and inflammatory cytokine release in primary murine macrophages [28]. TAK1 is a prosurvival protein kinase that activates NF-κB and MAPK signaling pathways downstream of TLRs, the IL-1 receptor, and the TNF receptor, inducing the expression of a number of pro-inflammatory and pro-survival genes, many of which suppress the activation of caspase-8 and RIPK1 in mammals [88] (FIG. 2b). Upon inhibition of NF-κB signaling, a cytosolic complex containing RIPK1 is formed to drive caspase-8–mediated inflammatory cell death in various cell types, such as mammalian macrophages [83] (FIG. 2b). Moreover, pharmacological inhibition and genetic deletion of TAK1 have led to spontaneous caspase-1 cleavage in an NLRP3-dependent manner and cell death in murine macrophages [26, 27]. Genetic deletion of RIPK1 has enhanced MLKL phosphorylation (necroptosis) but abrogated caspase-1 and GSDMD cleavage (pyroptosis), and activation of caspase-3 and caspase-7 (apoptosis) during infection with Yersinia sp. [28]. However, the combined loss of caspase-8, RIPK3 and caspase-1 has completely protected against Yersinia-induced cell death and cytokine release in murine macrophages [28], highlighting the essential role of these molecules in the cytokine-cell death positive feedback loop.

During IAV infection, the TLR–TRIF axis, RIG-I signaling, and NLRP3 inflammasome, induce the production of type I IFN and pro-inflammatory cytokines, such as IL-1β and IL-18, in immune and non-immune cells [8991]. Type I IFN subsequently upregulates the expression of the Z-nucleic acid sensor ZBP1 via IRF1. The Zα2 domain of ZBP1 senses replicating IAV Z-RNAs to assemble a signaling complex that leads to inflammatory cell death (FIG. 2c), resulting in cytokine release and ultimately, lung damage in mice [25]. Caspase-6 promotes binding of RIPK3 to ZBP1 to promote IAV-induced activation of the NLRP3 inflammasome and cell death in murine macrophages [92]. ZBP1 also drives inflammatory cell death, cytokine release, skin and colon inflammation, as well as perinatal lethality in mice under sterile inflammatory conditions [9396]. Altogether, these findings are relevant in that they highlight the important role of ZBP1 in contributing to a CS under different disease conditions.

Whereas almost all pathogens are capable of inducing similar sets of cytokines, it is still unclear why only some bacteria or viruses induce a CS. One possibility is that certain pathogens possess virulence factors that act on RHIM-containing proteins to inhibit cell death, and thus prevent the positive feedback loop that leads to a CS [97, 98]. Indeed, one of the effectors of enteropathogenic Escherichia coli, EspL, induces proteolytic cleavage of RHIM proteins including RIPK1, RIPK3, ZBP1 and TRIF, possibly limiting necroptosis and inflammation in mice [97]. Similarly, numerous viruses have evolved mechanisms to inhibit cell death and prolong the survival of infected host cells [98].

Collectively, these studies suggest that RHIM-containing proteins can provide a crucial mechanistic link between the occurrence of a CS and cell death. Indeed, RIPK1 is a key molecule in the cytokine-cell death positive feedback loop that limits ZBP1- and RIPK3-mediated inflammatory cell death and cytokine release, thus preventing systemic inflammation and multiorgan damage, as observed in patients with CSS.

The role of TNF in cell death and the pathogenesis of a CS.

In addition to the RHIM-containing proteins, cytokines themselves and their corresponding receptors and signaling pathways are important regulators of cell death and in the CS. TNF receptor signaling is one of the best studied examples of this regulation [15]. TNF signals through two distinct receptors, TNFR1 and TNFR2, and signaling through TNFR1 promotes various biological processes including cell death [99]. Downstream of TNF receptor engagement, components of the linear ubiquitin chain assembly complex (LUBAC) — SHARPIN, HOIP and HOIL-1 — are required for optimal NF-κB signaling and cytokine secretion in mammals [100] (FIG. 2b). In the same pathway, the ubiquitin-editing enzyme TNFAIP3 (also known as A20) is crucial for inhibiting NFκB signaling [101]. However, deficiency in LUBAC components in mice (Hoil1–/–) results in severe inflammation or lethality, mediated by TNF-dependent apoptosis and necroptosis [102]. The severe skin inflammation observed in mice lacking SHARPIN (Sharpincpdm mice) is prevented in SharpincpdmTnf–/– and SharpincpdmCasp8–/+Ripk3–/– mice [99, 100]. MYD88 is required for the production of TNF in Sharpincpdm mice, and SharpincpdmMyD88–/– mice are also protected from systemic inflammation and skin disease [103]. The importance of innate immune-mediated inflammatory cell death mechanisms downstream of cytokine signaling in driving this CS-mediated pathology is further highlighted by the finding that SharpincpdmNlrp3–/–, SharpincpdmCasp1/11–/– or SharpincpdmIl1b–/– mice show delayed onset of inflammation compared with Sharpincpdm mice [104, 105]. By contrast, Hoil1–/–Casp8–/–Ripk3–/– mice experience embryonic lethality [102], possibly owing to impaired fetal hematopoiesis that might be driven by deregulated cytokine production in the presence of RIPK1. Indeed, Hoil1–/–Casp8–/–Ripk3–/– Ripk1–/– mice are protected from embryonic lethality [102]. Similar to these mouse models, deficiency of HOIL-1 and HOIP in humans is associated with autoimmunity, inflammation, and hyperactive cytokine release [106, 107]. Patients with mutations or polymorphisms in HOIL1 or HOIP present with systemic inflammation and immunosuppression and are susceptible to recurrent opportunistic infections [106, 107]. In addition, monocytes derived from HOIL1deficient patients display hyperproduction of IL-6 upon IL-1β stimulation [106]. Also, A20–/– (Tnfaip3–/–) mice exhibit widespread tissue inflammation and perinatal lethality [108]. While most A20–/– mice die within a day of birth, A20–/–Ripk3–/– mice survive 2–3 weeks before succumbing to inflammatory death [109], suggesting the potential involvement of other cell death pathways beyond necroptosis. Indeed, presentation of a spontaneous erosive polyarthritis phenotype characterized by synovial and periarticular inflammation with high numbers of infiltrating mononuclear cells is reduced in A20myel-KONlrp3–/– mice, implying a possible pathogenic role for A20 in pyroptosis [110]. Moreover, polymorphisms in human TNFAIP3 are correlated with increased risk of the autoimmune diseases SLE and psoriasis, as well as with inflammatory pathology associated with cystic fibrosis [111113]. Overall, the components of LUBAC and A20 prevent systemic inflammation by limiting the cell death induced by TNF signaling, and the systemic inflammation and cell death observed in the absence of LUBAC and A20 can be rescued with the loss of PANoptosome components.

The physiological role of TNF signaling in triggering inflammatory cell death and CSS is further supported by in vivo studies in mice that lack caspase-8 or FADD. Mice lacking either of these molecules in intestinal epithelial cells (Casp8ΔIEC and FaddΔIEC) exhibit increased infiltration of immune cells, mostly myeloid cells, into the colonic mucosae and expression of proinflammatory cytokines such as IL-6, IL-1β, IFN-γ and TNF, leading to spontaneous intestinal inflammation [114, 115]. Intestinal epithelial cells of FaddΔIEC mice are characterized by caspase-8 activation that sensitizes them to TNF signaling-induced necroptosis, which potentially drives acute inflammation and lethality in FaddΔIEC mice [114, 115]. Indeed, FaddΔIECTnf–/– mice show mild focal epithelial lesions and immune cell infiltration in the colonic mucosa compared with control mice [114, 115]. The pathogenic role of TNF is further supported by the lethality observed in Casp8ΔIEC mice injected intravenously with TNF, which is not lethal to wildtype mice [114]. Moreover, FaddΔIECRipk3–/– mice or Casp8ΔIEC mice injected with nec-1 show improved intestinal pathology compared with FaddΔIEC or Casp8ΔIEC mice [114, 115]. However, FaddΔIECTnf–/– mice still exhibit inflammation when compared with wildtype mice, suggesting that TNF-independent mechanisms might also contribute to FADD deficiencyinduced pathology in mouse intestinal epithelial cells [115]. Likewise, ablation of RIPK3 in mice lacking FADD in keratinocytes (FaddE-KORipk3–/–) fully prevents the development of skin lesions in FaddE-KO mice [116]. Although Casp8–/–Ripk3–/–, Fadd–/–Ripk3–/–, Casp8–/–Mlkl–/– and Fadd–/– Mlkl–/– mice are protected from embryonic lethality [117, 118], loss of MLKL does not prevent systemic and intestinal inflammation in mice that express catalytically inactive caspase-8 (Casp8C362A/C362AMlkl–/–) [30]. The systemic inflammation observed in Casp8C362A/C362AMlkl–/– mice is characterized by increased concentrations of various pro-inflammatory cytokines, including IL1β and IL-18 [30], which might imply pathological roles of the inflammasome and pyroptosis in these mice. Indeed, Casp8C362A/C362AMlkl–/–Casp1–/– or Casp8C362A/C362AMlkl–/–Asc–/– mice are rescued from lethality and systemic inflammation, indicating that caspase-8 functions as a master regulator of pyroptosis, apoptosis, and necroptosis [29, 30]. Children with early-onset IBD exhibit increased expression of RIPK3 and MLKL, but reduced caspase-8 expression in the inflamed ileum and colon relative to healthy controls [119]. Similarly, patients with inherited caspase-8 (CASP8) deficiency present with early onset intestinal inflammation [120]. These studies provide correlative evidence of a link between inflammatory cell death and IBD in the clinical setting [119, 120]. Overall, inflammatory cytokine signaling pathways are intricately linked with cell death pathways that can contribute to a CS.

The above discussion makes it evident that inflammatory cell death, induced by the modulation of cell death regulators, the sensing of PAMPs, DAMPs, or cytokines, can induce further cytokine secretion leading to physiological disorders such as systemic and local inflammation in the skin and intestine. A further example of the important role of cell death in driving a CS is provided by mutation of the proline-serine-threonine phosphatase-interacting protein 2 gene in mice (Pstpip2cmo), which causes inflammatory lesions in the bones and various degrees of skin and paw inflammation that closely resemble the human disorder known as chronic recurrent multifocal osteomyelitis. Specifically, whereas neutrophils and IL-1β are crucial for the initiation of the disease in Pstpip2cmo mice, joint inflammation and cytokine release are inhibited in Pstpip2cmoRipk3–/–Casp8–/–Casp1/11–/– mice [33, 121]. Overall, these studies may serve to provide a framework and basis for the mechanistic definition of a CS. Although the particular triggers, pathways and molecules involved can differ, each of these distinct responses may initiate a positive feedback loop between cytokine release and cell death pathways that can lead to a CS and thus, potentially to multiple organ failure and unfavorable disease prognosis [16].

Therapeutics to treat a CS

Thus far, treatment of a CS has been largely aimed at maintaining critical organ function by limiting the collateral damage caused by an activated immune system. Although general immunosuppression was initially considered as a potential strategy in CSS, a deeper mechanistic understanding of the pathways involved has highlighted the failures of general immunosuppression and suggested that more targeted therapies may be a superior approach. Corticosteroids, such as glucocorticoids and dexamethasone, act as anti-inflammatory and immunomodulatory agents by inhibiting the synthesis of pro-inflammatory cytokines and driving lymphocyte cell death [122]. However, in IAV-mediated severe pneumonia, a meta-analysis found that patients treated with glucocorticoids might present worse clinical outcomes than those not receiving glucocorticoid therapy -- including an increased risk of secondary bacterial infection and death [123]. In the case of COVID-19, dexamethasone has been shown to be effective only in patients requiring mechanical ventilation or supplemental oxygen in the randomized open-label phase2/3 RECOVERY trial in hospitalized patients with COVID-19 (primary outcome: all-cause mortality based on hazard ratio from Cox regression) (NCT04381936)I [124]; this suggests that patient selection is crucial. Corticosteroids are generally avoided as frontline therapy for the treatment of CSS in patients receiving CAR T cell therapy. However, they are recommended for the treatment of neurologic adverse effects of T cell-engaging therapies, as well as in patients presenting with MAS and HLH [11, 125]. Overall, these findings suggest that while corticosteroids have been widely used across disease indications in the past, more targeted therapies should be considered to treat CSs.

To develop targeted therapeutics, a fundamental understanding of the biological processes and mechanistic details underlying a CS will be crucial. In this review, we discussed the connections between cytokine release and inflammatory cell death, and the cellular and molecular mechanisms of this positive feedback loop in a CS. These mechanistic insights should aid the development of novel therapeutic strategies for the numerous fatal conditions associated with a CS. Based on these insights, we review here existing CS blockers and therapeutics that are under development to target various aspects of the CS loop (Table 3).

Table 3 |.

Targeted therapeutics in cytokine storm-associated diseases in humans

TARGETED PATHWAY TREATMENTS
Therapeutics Anti-IL-1 Anti-IL-18 Anti-IL-6 Anti-TNF Anti-IFN-γ JAK inhibitors Caspase inhibitors
CSS Sepsis [173] [35, 174]
COVID-19 [175, 176] [177, 178] [179181] [182] [157, 183]
sJIA [184, 185] [145] [186] [187]
SLE [188]
Adult Still’s disease [189] [188]
HLH [190] [131, 191] [192]
NLRC4-MAS [145]
Castleman’s disease [193]
GvHD [194, 195] [196]
CAR T-cell therapy [197] [198]
Blinatumomab
Autoinflammatory Rheumatoid arthritis [199] [200] [201] [202]
Giant cell arteritis [203] [149]
Gout [204206]
Crohn’s disease [207, 208] [209] [210]
Ulcerative colitis [211] [212]
Psoriasis [213] [214] [215, 216]
CAPS [217, 218]
FMF [219]
NASH [220]
Refractory asthma [221]
Drugs Anakinra Canakinumab Rilonacept Tadekinig-α Tocilizumab Siltuximab Sarilumab Adalimumab Certolizumab Etanercept Golimumab Infliximab Emapalumab Baricitinib Tofacitinib Upadacitinib Ruxolitinib Emricasan

Green: FDA-approved drugs (for any indication) with efficacy in the given setting

Blue: Mixed responses or ongoing clinical trials

White: N/A

Numbers represent references.

Anti-cytokine therapy.

Cytokines themselves play key roles in driving much of CSS pathology; therefore, treatments aimed at dampening pro-inflammatory signaling could improve clinical outcomes. Suppressing cytokine signaling by using recombinant proteins, inhibiting cytokine production, or by neutralizing cytokines directly with monoclonal antibodies, has shown promising effects in various clinical settings (Table 3).

Anti-TNF Antibodies

One of the central cytokines involved in CSS that has been carefully studied over the years is the pro-inflammatory cytokine TNF. IAV-infected mice treated with a neutralizing antibody against TNF exhibit reduced pulmonary immune cell infiltration and less severe pathology, as well as improved survival relative to controls [126, 127]. The pathological roles of TNF have also been demonstrated in a co-infection model, where the administration of staphylococcal enterotoxin B (SEB) to IAV-infected mice led to enhanced cytokine production and lethality compared to SEB or IAV alone, and the neutralization of TNF by dimeric TNFR Fc fragment antibodies prolonged survival by two days compared with PBS treatment [128]. These findings would be particularly relevant to patients with IAV infection who have a concurrent bacterial infection. Case studies have shown that patients on anti-TNF antibody therapy to treat certain rheumatic diseases, such as rheumatoid arthritis and SLE, who become infected with SARSCoV-2 tend to be less likely to be hospitalized, which suggests that prophylactic anti-cytokine therapy might be beneficial to treat certain cases of COVID-19 [129]. Administration of neutralizing antibodies against TNF has also improved survival in mouse models of poly(I:C)induced HLH and LPS-induced sepsis [23]. Unfortunately, this success has not translated to humans, with most anti-TNF antibody trials showing no clinical benefit for treating sepsis [35]. One of the key reasons behind the failure of anti-TNF antibody therapies in CSS is likely patient heterogeneity, not only in terms of site and source of infection but also with respect to genetic background and comorbidities, but further research is required to robustly address this issue.

Anti–IFN-γ Antibodies

Among several cytokines that are increased in the serum of patients with HLH, IFN-γ seems to be a particularly promising target based on results from mouse models. Neutralization of IFN-γ markedly prevents the development of HLH-like pathology in perforin- and Rab27a-deficient mice (Pfp–/– and Rab27a–/– mice) infected with LCMV-WE [60, 130], although the mechanisms by which excess IFN-γ leads to the characteristic histological and clinical features of HLH in these mice have not been defined. Despite this, recently, the US FDA approved anti–IFN-γ antibody emapalumab to treat patients with relapsed/refractory HLH, based on favorable results in a single arm, open label phase 2/3 trial of 34 pHLH patients treated with emapalumab in combination with dexamethasone and/or other HLH-directed therapies as needed (primary endpoint: overall response) (NCT01818492; NCT02069899)II [131]. Also, neutralizing antibodies against IFN-γ have improved animal survival in a mouse model of LPS-induced sepsis [23]. Furthermore, anti–IFN-γ antibody treatment in LPS-challenged mice overexpressing human IL-6 (IL6-Tg) in an experimental model of MAS associated with sJIA, improved survival and reduced the amounts of several pro-inflammatory cytokines relative to IgG treated IL6-Tg mice [132]. Moreover, administration of IFN-γ has reduced hemophagocytosis in the liver of LCMV-infected Pfp–/– and Rab27a–/– mice compared with IgG treated mice, indicating that HLH can potentially be associated with the phagocytosis-promoting effect of IFN-γ [133]; however, more work is needed to thoroughly investigate this therapy option in patients with HLH.

Anti–IL-1β antibodies and IL-1 receptor antagonists

Cell death through pyroptosis leads to IL-1β release. Monoclonal antibodies against IL-1β, such as canakinumab, or recombinant human IL-1Ra, such as anakinra, are beneficial in individuals with autoinflammatory cryopyrinopathies. Case reports have revealed that patients can be effectively treated with recombinant IL-1Ra (receptor antagonist) in the case of CSS associated with sJIA, adult-onset Still’s disease, and SLE [134136]. Currently, a phase 1 randomized placebo-controlled trial is underway to evaluate the efficacy of anakinra for the treatment of sHLH by comparing the number of acquired infections or deaths in the treatment and placebo groups (primary outcome) (NCT02780583)III [137]. Canakinumab and anakinra are also being studied for treatment of COVID-19-induced ARDS and CSS [138]; for instance, a phase 3 randomized placebo controlled study is evaluating canakinumab in hospitalized patients with COVID-19 with the primary outcome being the number of patients with clinical response (NCT04362813)IV, and a phase 2/3 2-arm study of anakinra vs standard of care in patients with CSS secondary to SARS-CoV-2 infection with a primary outcome of treatment success (NCT04443881)V. In addition, anakinra has abolished CSs and neurotoxicity in humanized mice with high leukemia burden (recapitulating key features of a CS); this approach might potentially offer a therapeutic strategy to tackle neurotoxicity associated with CAR T-cell therapy [139]. Indeed, the long-term safety profile of anakinra has been well-established in patients with rheumatoid arthritis and severe cryopyrin-associated autoinflammatory syndrome (CAPS) [140]. IL-1Ras like anakinra might also be beneficial in treating diseases associated with IL-1α release, such as neutrophilic dermatosis [81], and efficacy has been observed in certain case reports [141].

Anti–IL-18 treatments

Pyroptosis executed by GSDMD pore formation triggers IL-18 release [43]. Patients with a CS caused by MAS have high amounts of IL-18 in their serum [142]. The pro-inflammatory effects of IL-18 are counterbalanced by its high-affinity endogenous antagonist, IL-18 binding protein (IL-18BP) [143]. Repeated challenge of mice deficient in IL-18BP (Il18bp–/–) with a toll-like receptor 9 agonist, mirrors clinical manifestations of MAS, such as pancytopenia and hemophagocytosis in bone marrow smears; blocking IL-18 signaling using a monoclonal antibody against the IL-18 receptor rescued this phenotype [144]. In line with this mouse work, a case study reported that administration of tadekinig-α, a recombinant IL-18BP, reduced MAS flare frequency and severity in a patient with resistant sJIA and recurrent MAS [145]. However, further studies are needed to fully understand the utility of this candidate therapy in the clinical setting, and clinical trials are currently ongoing.

Anti–IL-6 receptor antibodies

Similar to IL-1β and TNF, upregulation of IL-6 involves the NF-κB signaling pathway, and IL-6 binding to its receptor induces robust pro-inflammatory signaling [146]. Tocilizumab, a humanized monoclonal antibody against IL-6R, has been reported to be effective in severe rheumatoid arthritis, sJIA, multicentric Castleman’s disease and CAR T cell-induced CSS [147150]. In humanized mice with high leukemia burden, monocyte-derived IL-1β and IL-6 drive CSS and neurotoxicity induced by CAR T cells, potentially suggesting a role for anti–IL-1 and anti–IL-6 antibody therapy in combating these side effects [63, 139]. Anti–IL-6 antibody therapies have also been tested with mixed clinical responses in COVID-19. Whereas the phase 3 randomized placebo-controlled COVACTA (NCT04320615)VI and Kevzara (NCT04327388)VII trials in patients with severe COVID-19–associated pneumonia using tocilizumab and sarilumab, respectively, (anti-IL-6R antibodies) did not meet their primary or key secondary endpoints of improved clinical status and reduced patient mortality [151], the phase 3 randomized placebo-controlled EMPACTA (NCT04372186)VIII trial of tocilizumab met its primary endpoint of a reduction in the number of patients requiring mechanical ventilation by day 28, but failed to improve mortality. In addition, anti-IL-6R antibody treatment in the phase 4 randomized, embedded, multifactorial adaptive platform REMAP-CAP trial (NCT02735707)IX met its primary endpoint and improved survival in critically ill patients with COVID-19 [152]. The improved results in the REMAP-CAP study, however, might be due to the additional organ support that was provided for patients in the intensive care unit.

Combination therapies

The efficacy of targeting a single cytokine as described may be limited due to the continued production and signaling of other functionally redundant cytokines that contribute to immune activation and end-organ damage in CSS. Indeed, synergy between TNF and IFN-γ occurs upstream of inflammatory cell death in murine and human macrophages, and the consequent signaling cascade and cell death can lead to further release of other cytokines and alarmins [23]. Moreover, the combined administration of neutralizing antibodies against TNF and IFN-γ has improved survival more than single-cytokine blockade of IFN-γ or TNF in mouse models of poly(I:C)-induced HLH and LPS-induced sepsis [23]. The combined neutralization of TNF and IFN-γ has also protected mice from lethality during SARS-CoV-2 infection [23]. Another key example of the potential utility of cytokine combination therapy highlights IL-6 and IL-1 targeting: tocilizumab (anti-IL-6R antibody) fails to protect mice from CAR T cell-mediated neurotoxicity, whereas anakinra (recombinant human IL-1Ra) is uniquely associated with rescue from lethal neurotoxicity [139]; this suggests that anti–IL-6 and anti–IL-1 antibody combination therapies might be effective against CAR T cell-induced CSS and/or neurotoxicity, although this warrants further investigation.

Small molecules.

Several small molecules are available for inhibiting cytokine production and cytokine signaling, scavenging reactive oxygen species (ROS) and preventing cell death. These molecules have the potential to interrupt the CS feedback loop, and some, such as JAK inhibitors, have shown promising effects in both pre-clinical and clinical settings for the treatment of CSS (Table 3). Furthermore, as cytokines exhibit high transcriptional regulation, studies to characterize transcriptional regulators and their mechanisms of action in the production of specific cytokines involved in CSS have the potential to identify whether these molecules might be considered as candidate drugs and targets in therapeutic discovery.

JAK inhibitors

TNF and IFN-γ activate JAK–STAT signaling pathways that can lead to cell death through NO production [23]. JAKs also mediate signaling pathways of multiple other cytokines, including IFNs, IL-2, IL-3, IL-4, IL-6, IL-7, IL-9, IL-12, IL-10, IL-15, IL-21, GM-CSF and G-CSF [153, 154]. JAK–STAT pathways are required for many innate and acquired immune responses, and mutations of JAKs and STATs can predispose affected individuals to certain autoimmune diseases such as rheumatoid arthritis, vascular inflammatory diseases, and insulin resistance [154]. JAK inhibitors are available in an oral form and have good bioavailability, which makes these inhibitors potentially preferable to injectable medications, such as the anti-cytokine antibodies, for chronic and prolonged use. Because JAKs function as common downstream mediators in the signaling pathways of several cytokines, JAK inhibitors offer a broad spectrum of activity and are an appealing strategy to break the CS loop for the treatment of CSS. For example, ruxolitinib, a JAK inhibitor, can reduce tissue immunopathology and disease manifestations in murine models of pHLH and sHLH [155, 156]. Additionally, JAK inhibitors might be beneficial in the fight against COVID-19, as the JAK inhibitor baricitinib, when combined with the broad-spectrum antiviral drug remdesivir, was found to be superior to remdesivir alone in the phase 3 randomized controlled ACTT-2 trial in hospitalized patients with COVID-19 in reducing recovery time, based on a stratified log-rank test (primary outcome) and accelerated improvement in clinical status (NCT04401579)X [157].

Antioxidants

Several studies suggest that ROS and NO contribute to CSS pathology [158]. For example, ROS produced by alveolar phagocytic cells in mice during IAV infection can induce immune cell infiltration into the lungs, thereby driving significant lung immunopathology; depletion of superoxide by injection of superoxide dismutase protects mice against lethal IAV infection, indicating the potential of antioxidant therapy in treating this infection [159]. Edaravone is an US FDA-approved antioxidant for the treatment of patients with amyotrophic lateral sclerosis, a fatal neurodegenerative disease [160], which might be effective in CSS, although to our knowledge, this remains to be tested.

Activated macrophages express high amounts of inducible nitric oxide synthase (iNOS)-- an enzyme that catalyzes the production of NO [23]. Excess NO is harmful to tissues and causes vasodilation and hypotension -- common clinical features of CSS requiring vasopressor administration [161]. Thus, NOS inhibitors reduce hypotension, organ dysfunction, and mortality in murine models of sepsis, including LPS- and TNF-induced shock [162]. However, a randomized, placebo-controlled phase 3 trial of the NOS inhibitor 546C88 in patients with sepsis found that this treatment positively impacted blood pressure and vascular resistance but also increased mortality compared with the control group, leading to premature termination of the study [163]. Although conjectural, one of the possible reasons for this failure might be the nonselective nature of the NOS inhibitor used in the study. NO can be generated by three isoforms of NOS — iNOS, endothelial NOS and neuronal NOS. iNOS can be induced by pathogens and cytokines and contributes to the pathology of various diseases [161]. In animals, most studies using selective NOS inhibitors have shown better protective effects compared with the use of nonselective NOS inhibitors. Indeed, the transgenic overexpression of eNOS in mice (eNOS Tg) improves survival during LPS shock, underscoring the need for NOS specificity in appropriately determining clinical outcomes during sepsis [164]. However, in contrast to the benefits of specific iNOS inhibitors, iNOS-deficient mice (Nos2–/–) are not protected against sepsis [165]. The conflicting results for iNOS inhibitors versus iNOS deficiency might be explained by the concentrations of NO produced in cells and tissues, although this remains to be tested. Reducing iNOS activity might be more beneficial than complete inhibition, with the assumption that residual NO might exert essential protective functions. Overall, these dichotomous roles of NO have proved to be the biggest challenge in the development of potential antioxidant therapies to treat CSS.

Caspase inhibitors

One of the major clinical manifestations of CSS is tissue damage [16]; therefore, protecting organs against the damage triggered by the CS is a relevant therapeutic strategy. TNF and IFN-γ activate caspases-3, 7, 8 and 9 in mouse macrophages, and inhibition of caspase-8 suppresses TNF- and IFN-γ-induced cell death in mouse macrophages and mortality in mice subjected to LPS-induced sepsis [23]. Caspase inhibition can improve organ function and/or survival in various mouse models of ischemia-reperfusion injury, burns, endotoxemia, and sepsis [166]. Administration of the pan-caspase inhibitor zVAD inhibits cell death in thymocytes to improve survival of mice and rats in a cecal ligation and puncture models of sepsis [19, 166]. Emricasan is also an orally available pan-caspase inhibitor which has been tested in a completed randomized, placebo-controlled phase 2 trial in patients with nonalcoholic steatohepatitis cirrhosis (primary outcome: measure of change in alanine aminotransferase from baseline to day 28 (NCT02077374))XI [167], and emricasan might be considered for clinical trials in other diseases associated with CSS. However, while many have attempted to develop caspase inhibitors, caspases are historically difficult to target [168], suggesting that focusing on molecules further upstream in the cell death signaling pathway might be a superior strategy.

Concluding remarks

Innate immune cells such as macrophages, dendritic cells, and NK cells, release pro-inflammatory cytokines in response to infectious and sterile insults. The initial inflammatory response is crucial to clear the infection; however, a dysregulated inflammatory response can cause a CS, potentially leading to tissue and organ damage, and mortality. CS can also occur in response to some therapeutic interventions and underlying conditions during cancer and autoinflammation. Here, we reviewed the molecular mechanisms connecting cytokine release, cytokine signaling, and innate immune-mediated cell death in a positive feedback loop to molecularly define a CS (FIG. 3). An improved understanding of this feedback mechanism, including the specific roles of cytokines involved, how immune cells trigger cell death, and the molecules that constitute the cell death signalosomes, is fundamental to developing candidate therapeutic strategies to ideally and precisely target a CS in a given individual (see Outstanding Questions).

Key Figure, Figure 3 |. Schematic representation of the mechanism of a cytokine storm.

Key Figure, Figure 3 |

During pathogenic infections, autoinflammatory diseases, and other cytokine storm (CS)-inducing conditions, innate immune cells from humans and mice become activated and release multiple pro-inflammatory cytokines, including TNF and IFN-γ [16]. The synergistic response of TNF and IFN-γ can induce inflammatory cell death, which can result in more cytokines being produced and which might ultimately lead to a CS, with potential tissue and organ damage, and mortality in humans and mice [23]. Some components of Figure 3 were derived from BioRender.com.

Outstanding Questions.

  • Currently, a cytokine storm (CS) is often treated in a disease-specific manner that depends on the underlying trigger. However, there may be advantages to finding targets that are independent of the trigger to provide a more global strategy for CS management. What therapeutic targets can be used to prevent CSs regardless of the underlying cause?

  • What is the therapeutic window for anti-cytokine therapies in a CS? How does changing the timing of the treatment impact the course of the disease?

  • TNF and IFN-γ drive cell death, and blocking these cytokines can provide benefits in murine models of CS syndrome, but not a full cure for all cases. What is the contribution that cytokines other than TNF and IFN-γ could have in mediating cell death and a CS?

  • How does cell death affect the release of IL-6? Can neutralization of TNF and IFN-γ inhibit the production of IL-6, and prevent a therapeutic intervention-induced CS? Can neutralization of TNF and IFN-γ be used to treat or prevent global CS-associated diseases?

  • The absence of caspase-8 leads to cell death in vivo, but the caspase-8 node is also essential for inducing various forms of inflammatory cell death in response to PAMPs or DAMPs. How does the same molecule regulate two contrasting phenotypes?

  • Since dexamethasone dampens NF-κB signaling necessary to produce cytokines and JAK inhibitors prevent IFN signaling, can the combination of dexamethasone with a JAK inhibitor mimic conditions similar to neutralization of TNF and IFN-γ to prevent a CS?

Based on the molecular mechanisms linking inflammatory cell death and the CS, it is clear that TNF and IFN-γ are of paramount importance; they are up-regulated in almost all cases of CSs [16], and their synergism induces inflammatory cell death pathways in myeloid cells [23]. Neutralization of these two cytokines can provide superior protection in pre-clinical mouse models of CSS [23], suggesting that combined re-purposing of these therapies might potentially be beneficial in patients with CSS, ideally including those presenting with COVID-19. However, determining the therapeutic window of anti-cytokine therapies is challenging. The paradigm of “hit hard and hit early,” popularized for the treatment of AIDS [169], might also work in the treatment of a CS. Indeed, early release of TNF is a crucial driver of lethality caused by SEB in mice, whereas late release does not play much of a role [170]. Various anti-cytokine therapy results from clinical trials have failed to meet the high expectations raised in preclinical models, most likely owing to the short half-life of most cytokines and the narrow therapeutic windows that have been possible. Further research should also focus on the identification of drugs targeting the upstream innate immune sensors and cell death signaling pathway molecules that can be used across CSS regardless of underlying causes. Because of the interconnection between inflammatory cell death and the CS, such identification might be achieved by looking outside the classical CSS repertoire and considering therapeutic strategies that target cell death.

Highlights.

  • A Cytokine storm (CS) is a clinically relevant condition that has been associated with several life-threatening diseases.

  • Although CS syndrome-inducing agents can fundamentally differ, the interactions of PAMPs or DAMPs with PRRs evoke similar cytokine profiles in mammals.

  • Cytokines are intricately linked to cell death mechanisms in mammals and are involved in a positive feedback loop whereby cytokine signaling or PAMP/DAMP sensing causes inflammatory cell death that facilitates further pathogenic cytokine release, culminating in a CS to drive severe, life-threatening damage to host tissues and organs.

  • Neutralization of TNF and IFN-γ can prevent cell death and inhibit the occurrence of a CS in mouse models of SARS-CoV-2 infection, hemophagocytic lymphohistiocystosis, and sepsis.

  • A Combination of FDA approved drugs might be more beneficial than single treatment approaches for the clinical management of CSs.

Acknowledgments

We appreciate all those who have contributed substantially to the study of cytokine biology to build the foundation that has enabled us to demonstrate and define a CS. We apologize to our colleagues in the field whose work could not be cited due to space limitations. We thank K. Nichols for her insightful comments and feedback, all members of the Kanneganti laboratory for their comments and suggestions and R. Tweedell for scientific editing and writing support. Work from our laboratory is supported by the US National Institutes of Health (AI101935, AI124346, AR056296 and CA253095 to T.-D.K.) and the American Lebanese Syrian Associated Charities (to T.-D.K.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Glossary

Acute respiratory distress syndrome (ARDS)

life-threatening lung injury caused by low oxygen concentrations in the blood as a result of fluid build-up in alveoli.

Alarmins

Damage-associated molecular patterns such as HMGB1 or IL-1α; released by damaged or necrotic cells.

Autoinflammation

Caused by the overactivation of the innate immune system with little or no evidence of a specific adaptive immunity, such as auto-reactive T cells or autoantibodies.

Chimeric antigen receptor (CAR) T cells

T cells that have been engineered with a specific receptor protein to enable targeting of a specific protein. They typically combine antigen-binding and T-cell activating functions into a single receptor.

Chronic obstructive pulmonary disease (COPD)

chronic inflammatory lung disease causing obstructed airflow from the lungs.

Chronic recurrent multifocal osteomyelitis

rare idiopathic autoinflammatory bone disease that mostly affects children and adolescents.

Cryopyrin-associated autoinflammatory syndrome (CAPS)

rare hereditary inflammatory disorder related to a gain-of-function mutation in the protein cryopyrin (NLRP3). CAPS encompasses a continuum of three phenotypes: familial cold autoinflammatory syndrome, Muckle-Wells syndrome, and neonatal-onset multisystem inflammatory disease.

Cryopyrinopathies

group of rare autoinflammatory diseases: includes familial cold autoinflammatory syndrome, Muckle-Wells syndrome and chronic infantile neurologic cutaneous articular syndrome (neonatal-onset multisystemic inflammatory disease).

Early-onset periodic fever syndrome

disease characterized by recurrent spontaneous attacks of multi-systemic inflammation in children.

Gasdermin

conserved family of proteins among vertebrates comprised of six members in humans, gasdermin A, -B, -C, -D, -E (DFNA5) and DFNB59.

Hemophagocytic lymphohistiocystosis

Hemophagocytosis is the process by which activated macrophages or phagocytes engulf erythrocytes, leukocytes, platelets and their precursors. Hemophagocytic lymphohistiocystosis is a life-threatening hyper-inflammatory condition occurring when histiocytes and lymphocytes become overactive and attack the body.

Linear ubiquitin chain assembly complex (LUBAC)

E3 ubiquitin ligase complex composed of HOIL-1, HOIP, and SHARPIN that generates linear polyubiquitin chains and stabilizes TNF-R1 signaling to regulate the NF-κB pathway.

Lymphadenopathy

enlargement or swelling of one or more lymph nodes from several potential causes, including infection, autoimmunity, and malignancy.

Macrophage activation syndromes (MAS)

severe and potentially fatal complication of several rheumatic diseases characterized by uncontrolled activation of macrophages and T lymphocytes.

Multicentric Castleman’s disease

rare disorder involving an overgrowth of cells in multiple lymph nodes throughout the body; associated with human herpes virus type 8 (HHV-8) and HIV-1.

Necroptosis

form of regulated cell death; executed by RIPK3 and MLKL upon caspase inhibition.

PANoptosis

unique, physiologically relevant inflammatory programmed cell death pathway regulated by the PANoptosome; it has key features of pyroptosis, apoptosis, and necroptosis.

PANoptosome

complex assembled under certain conditions to provide a molecular scaffold for contemporaneous engagement of key molecules involved in pyroptosis, apoptosis, and necroptosis.

Pathogen-associated or damage-associated molecular patterns (PAMPs or DAMPs)

PAMPs are small molecular motifs conserved within a class of microbes. DAMPs are produced or released by damaged and dying cells. Both PAMPs and DAMPs are recognized by pattern recognition receptors in host cells to activate innate immune responses.

Pattern recognition receptors (PRRs)

Germline-encoded molecules recognizing conserved molecular structures of microbial origin called PAMPs. They also sense endogenous molecules produced by, or released from damaged and dying cells called DAMPs.

RIP homotypic interaction motif (RHIM)

short non-globular sequence stretch that is necessary for the recruitment of RIPK1 and RIPK3 by ZBP1. This motif is also found in TRIF.

Ripk1D325A/D325A

Mice with this mutation are resistant to the caspase-mediated cleavage of RIPK1 that inhibits necroptosis; mice are hypersusceptible to inflammation and are embryonically lethal.

Severe combined immunodeficiency

group of rare disorders caused by mutations in different genes involved in the development and function of immune cells; patients suffer recurring infections with bacteria, fungi, and viruses.

Systemic juvenile idiopathic arthritis (sJIA)

rare autoinflammatory disease leading to inflammation in the joints and other parts of the body. It affects only 10% to 15% of children with JIA.

Footnotes

Resources

Competing interests

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Ferrara JL et al. (1993) Cytokine storm of graft-versus-host disease: a critical effector role for interleukin-1. Transplant Proc 25 (1 Pt 2), 1216–7. [PubMed] [Google Scholar]
  • 2.Antin JH and Ferrara JL (1992) Cytokine dysregulation and acute graft-versus-host disease. Blood 80 (12), 2964–8. [PubMed] [Google Scholar]
  • 3.Barry S. et al. (2000) Cytopathology or immunopathology? The puzzle of cytomegalovirus pneumonitis revisited. Bone Marrow Transplantation 26 (6), 591–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Imashuku S. (2002) Clinical features and treatment strategies of Epstein-Barr virusassociated hemophagocytic lymphohistiocytosis. Crit Rev Oncol Hematol 44 (3), 259–72. [DOI] [PubMed] [Google Scholar]
  • 5.Bisno AL et al. (2003) Molecular basis of group A streptococcal virulence. Lancet Infect Dis 3 (4), 191–200. [DOI] [PubMed] [Google Scholar]
  • 6.Yokota S. (2003) [Influenza-associated encephalopathy--pathophysiology and disease mechanisms]. Nihon Rinsho 61 (11), 1953–8. [PubMed] [Google Scholar]
  • 7.Jahrling PB et al. (2004) Exploring the potential of variola virus infection of cynomolgus macaques as a model for human smallpox. Proc Natl Acad Sci U S A 101 (42), 15196–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Huang KJ et al. (2005) An interferon-gamma-related cytokine storm in SARS patients. J Med Virol 75 (2), 185–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yuen KY and Wong SS (2005) Human infection by avian influenza A H5N1. Hong Kong Med J 11 (3), 189–99. [PubMed] [Google Scholar]
  • 10.Ravelli A. et al. (2016) 2016 Classification Criteria for Macrophage Activation Syndrome Complicating Systemic Juvenile Idiopathic Arthritis: A European League Against Rheumatism/American College of Rheumatology/Paediatric Rheumatology International Trials Organisation Collaborat. Arthritis & Rheumatology 68 (3), 566–576. [DOI] [PubMed] [Google Scholar]
  • 11.Mahlaoui N. et al. (2007) Immunotherapy of familial hemophagocytic lymphohistiocytosis with antithymocyte globulins: a single-center retrospective report of 38 patients. Pediatrics 120 (3), e622–8. [DOI] [PubMed] [Google Scholar]
  • 12.Lee DW et al. (2014) Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124 (2), 188–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Topp MS et al. (2015) Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. The Lancet Oncology 16 (1), 57–66. [DOI] [PubMed] [Google Scholar]
  • 14.Teachey DT et al. (2013) Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood 121 (26), 5154–5157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tisoncik JR et al. (2012) Into the Eye of the Cytokine Storm. Microbiology and Molecular Biology Reviews 76 (1), 16–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fajgenbaum DC and June CH (2020) Cytokine Storm. New England Journal of Medicine 383 (23), 2255–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hotchkiss RS et al. (2005) Accelerated lymphocyte death in sepsis occurs by both the death receptor and mitochondrial pathways. J Immunol 174 (8), 5110–8. [DOI] [PubMed] [Google Scholar]
  • 18.Hotchkiss RS et al. (1999) Overexpression of Bcl-2 in transgenic mice decreases apoptosis and improves survival in sepsis. J Immunol 162 (7), 4148–56. [PubMed] [Google Scholar]
  • 19.Hotchkiss RS et al. (1999) Prevention of lymphocyte cell death in sepsis improves survival in mice. Proceedings of the National Academy of Sciences 96 (25), 14541–14546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Torgersen C. et al. (2009) Macroscopic postmortem findings in 235 surgical intensive care patients with sepsis. Anesth Analg 108 (6), 1841–7. [DOI] [PubMed] [Google Scholar]
  • 21.Hotchkiss RS and Nicholson DW (2006) Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol 6 (11), 813–22. [DOI] [PubMed] [Google Scholar]
  • 22.Christgen S. et al. (2020) Identification of the PANoptosome: A Molecular Platform Triggering Pyroptosis, Apoptosis, and Necroptosis (PANoptosis). Frontiers in Cellular and Infection Microbiology 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Karki R. et al. (2020) Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gurung P. et al. (2014) FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J Immunol 192 (4), 1835–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kuriakose T. et al. (2016) ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. Science Immunology 1 (2), aag2045-aag2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Malireddi RKS et al. (2018) TAK1 restricts spontaneous NLRP3 activation and cell death to control myeloid proliferation. J Exp Med 215 (4), 1023–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Malireddi RKS et al. (2020) Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity–independent pyroptosis, apoptosis, necroptosis, and inflammatory disease. Journal of Experimental Medicine 217 (3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Malireddi RKS, Kesavardhana S; Karki R; Kancharana B; Burton AB; Kanneganti TD (2020) RIPK1 distinctly regulates Yersinis-induced inflammatory cell death, PANoptosis. ImmunoHorizons. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fritsch M. et al. (2019) Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature 575 (7784), 683–687. [DOI] [PubMed] [Google Scholar]
  • 30.Newton K. et al. (2019) Activity of caspase-8 determines plasticity between cell death pathways. Nature 575 (7784), 679–682. [DOI] [PubMed] [Google Scholar]
  • 31.Schwarzer R. et al. (2020) New insights into the regulation of apoptosis, necroptosis, and pyroptosis by receptor interacting protein kinase 1 and caspase-8. Curr Opin Cell Biol 63, 186193. [DOI] [PubMed] [Google Scholar]
  • 32.Karki R. et al. (2020) Interferon regulatory factor 1 regulates PANoptosis to prevent colorectal cancer. JCI Insight 5 (12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lukens JR et al. (2014) Dietary modulation of the microbiome affects autoinflammatory disease. Nature 516 (7530), 246–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Malireddi RKS et al. (2019) ZBP1 and TAK1: Master Regulators of NLRP3 Inflammasome/Pyroptosis, Apoptosis, and Necroptosis (PAN-optosis). Front Cell Infect Microbiol 9, 406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chousterman BG et al. (2017) Cytokine storm and sepsis disease pathogenesis. Seminars in Immunopathology 39 (5), 517–528. [DOI] [PubMed] [Google Scholar]
  • 36.World Health Organization. (2020) Sepsis Fact Sheet. [Google Scholar]
  • 37.Walker MJ et al. (2014) Disease Manifestations and Pathogenic Mechanisms of Group A Streptococcus. Clinical Microbiology Reviews 27 (2), 264–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Spaulding AR et al. (2013) Staphylococcal and Streptococcal Superantigen Exotoxins. Clinical Microbiology Reviews 26 (3), 422–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sharma J. et al. (2011) Features of sepsis caused by pulmonary infection with Francisella tularensis Type A strain. Microb Pathog 51 (1–2), 39–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mares CA et al. (2008) Initial Delay in the Immune Response to Francisella tularensis Is Followed by Hypercytokinemia Characteristic of Severe Sepsis and Correlating with Upregulation and Release of Damage-Associated Molecular Patterns. Infection and Immunity 76 (7), 3001–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sjostedt A. (2007) Tularemia: history, epidemiology, pathogen physiology, and clinical manifestations. Ann N Y Acad Sci 1105, 1–29. [DOI] [PubMed] [Google Scholar]
  • 42.Ning S. et al. (2011) IRF7: activation, regulation, modification and function. Genes Immun 12 (6), 399–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Man SM et al. (2017) Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev 277 (1), 61–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Monneret G. et al. (2006) Persisting low monocyte human leukocyte antigen-DR expression predicts mortality in septic shock. Intensive Care Med 32 (8), 1175–83. [DOI] [PubMed] [Google Scholar]
  • 45.Lee S. et al. (2020) Coronaviruses: Innate Immunity, Inflammasome Activation, Inflammatory Cell Death, and Cytokines. Trends Immunol 41 (12), 1083–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hadjadj J. et al. (2020) Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369 (6504), 718–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pairo-Castineira E. et al. (2020) Genetic mechanisms of critical illness in Covid-19. Nature. [DOI] [PubMed] [Google Scholar]
  • 48.Lucas C. et al. (2020) Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 584 (7821), 463–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Picard C. et al. (2018) International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on Inborn Errors of Immunity. Journal of Clinical Immunology 38 (1), 96–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Meeths M. et al. (2015) Incidence and clinical presentation of primary hemophagocytic lymphohistiocytosis in Sweden. Pediatr Blood Cancer 62 (2), 346–352. [DOI] [PubMed] [Google Scholar]
  • 51.Ménasché G. et al. (2000) Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nature Genetics 25 (2), 173–176. [DOI] [PubMed] [Google Scholar]
  • 52.Göransdotter Ericson K. et al. (2001) Spectrum of Perforin Gene Mutations in Familial Hemophagocytic Lymphohistiocytosis. 68 (3), 590–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Feldmann J. et al. (2003) Munc13–4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3). Cell 115 (4), 461–73. [DOI] [PubMed] [Google Scholar]
  • 54.zur Stadt U. et al. (2005) Linkage of familial hemophagocytic lymphohistiocytosis (FHL) type-4 to chromosome 6q24 and identification of mutations in syntaxin 11. Hum Mol Genet 14 (6), 827–34. [DOI] [PubMed] [Google Scholar]
  • 55.zur Stadt U. et al. (2009) Familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) is caused by mutations in Munc18–2 and impaired binding to syntaxin 11. Am J Hum Genet 85 (4), 482–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jessen B. et al. (2013) Graded defects in cytotoxicity determine severity of hemophagocytic lymphohistiocytosis in humans and mice. Front Immunol 4, 448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Henter JI et al. (1993) Familial hemophagocytic lymphohistiocytosis and viral infections. Acta Paediatr 82 (4), 369–72. [DOI] [PubMed] [Google Scholar]
  • 58.Humblet-Baron S. et al. (2019) IFN-γ and CD25 drive distinct pathologic features during hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol 143 (6), 2215–2226.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Binder D. et al. (1998) Aplastic anemia rescued by exhaustion of cytokine-secreting CD8+ T cells in persistent infection with lymphocytic choriomeningitis virus. J Exp Med 187 (11), 190320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jordan MB et al. (2004) An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder. Blood 104 (3), 735–743. [DOI] [PubMed] [Google Scholar]
  • 61.Bode SF et al. (2015) The syndrome of hemophagocytic lymphohistiocytosis in primary immunodeficiencies: implications for differential diagnosis and pathogenesis. Haematologica 100 (7), 978–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Liu Y. et al. (2020) Gasdermin E–mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Science Immunology 5 (43), eaax7969. [DOI] [PubMed] [Google Scholar]
  • 63.Giavridis T. et al. (2018) CAR T cell–induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nature Medicine 24 (6), 731–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu E. et al. (2020) Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. New England Journal of Medicine 382 (6), 545–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Suntharalingam G. et al. (2006) Cytokine Storm in a Phase 1 Trial of the Anti-CD28 Monoclonal Antibody TGN1412. New England Journal of Medicine 355 (10), 1018–1028. [DOI] [PubMed] [Google Scholar]
  • 66.Eastwood D. et al. (2010) Monoclonal antibody TGN1412 trial failure explained by species differences in CD28 expression on CD4+ effector memory T-cells. Br J Pharmacol 161 (3), 512–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pallardy M. and Hünig T. (2010) Primate testing of TGN1412: right target, wrong cell. Br J Pharmacol 161 (3), 509–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gogishvili T. et al. (2009) Rapid regulatory T-cell response prevents cytokine storm in CD28 superagonist treated mice. PLoS One 4 (2), e4643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Winkler U. et al. (1999) Cytokine-Release Syndrome in Patients With B-Cell Chronic Lymphocytic Leukemia and High Lymphocyte Counts After Treatment With an Anti-CD20 Monoclonal Antibody (Rituximab, IDEC-C2B8). Blood 94 (7), 2217–2224. [PubMed] [Google Scholar]
  • 70.Shan D. et al. (2000) Signaling events involved in anti-CD20-induced apoptosis of malignant human B cells. Cancer Immunol Immunother 48 (12), 673–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dinarello CA (2000) Proinflammatory Cytokines. Chest 118 (2), 503–508. [DOI] [PubMed] [Google Scholar]
  • 72.Webster JD and Vucic D. (2020) The Balance of TNF Mediated Pathways Regulates Inflammatory Cell Death Signaling in Healthy and Diseased Tissues. Front Cell Dev Biol 8, 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee SH et al. (2017) Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci Rep 7 (1), 10133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Doherty GM et al. (1992) Evidence for IFN-gamma as a mediator of the lethality of endotoxin and tumor necrosis factor-alpha. J Immunol 149 (5), 1666–70. [PubMed] [Google Scholar]
  • 75.Place DE and Kanneganti TD (2020) The innate immune system and cell death in autoinflammatory and autoimmune disease. Curr Opin Immunol 67, 95–105. [DOI] [PubMed] [Google Scholar]
  • 76.Dillon CP et al. (2014) RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell 157 (5), 1189–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rickard JA et al. (2014) RIPK1 regulates RIPK3-MLKL-driven systemic inflammation and emergency hematopoiesis. Cell 157 (5), 1175–88. [DOI] [PubMed] [Google Scholar]
  • 78.Lin J. et al. (2016) RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation. Nature 540 (7631), 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Newton K. et al. (2016) RIPK1 inhibits ZBP1-driven necroptosis during development. Nature 540 (7631), 129–133. [DOI] [PubMed] [Google Scholar]
  • 80.Cuchet-Lourenço D. et al. (2018) Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation. Science 361 (6404), 810–813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lukens JR et al. (2013) RIP1-driven autoinflammation targets IL-1alpha independently of inflammasomes and RIP3. Nature 498 (7453), 224–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mifflin L. et al. (2020) Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat Rev Drug Discov 19 (8), 553–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Delanghe T. et al. (2020) RIPK1 Kinase-Dependent Death: A Symphony of Phosphorylation Events. Trends Cell Biol 30 (3), 189–200. [DOI] [PubMed] [Google Scholar]
  • 84.Lalaoui N. et al. (2020) Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease. Nature 577 (7788), 103–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tao P. et al. (2020) A dominant autoinflammatory disease caused by non-cleavable variants of RIPK1. Nature 577 (7788), 109–114. [DOI] [PubMed] [Google Scholar]
  • 86.Lawlor KE et al. (2015) RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL. Nat Commun 6, 6282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kitur K. et al. (2015) Toxin-induced necroptosis is a major mechanism of Staphylococcus aureus lung damage. PLoS Pathog 11 (4), e1004820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kishida S. et al. (2005) TAK1-binding protein 2 facilitates ubiquitination of TRAF6 and assembly of TRAF6 with IKK in the IL-1 signaling pathway. Genes Cells 10 (5), 447–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Le Goffic R. et al. (2007) Cutting Edge: Influenza A virus activates TLR3-dependent inflammatory and RIG-I-dependent antiviral responses in human lung epithelial cells. J Immunol 178 (6), 3368–72. [DOI] [PubMed] [Google Scholar]
  • 90.Kanneganti TD et al. (2006) Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem 281 (48), 36560–8. [DOI] [PubMed] [Google Scholar]
  • 91.Thomas PG et al. (2009) The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity 30 (4), 566–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zheng M. et al. (2020) Caspase-6 Is a Key Regulator of Innate Immunity, Inflammasome Activation, and Host Defense. Cell 181 (3), 674–687.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kesavardhana S. et al. (2020) The Zα2 domain of ZBP1 is a molecular switch regulating influenza-induced PANoptosis and perinatal lethality during development. Journal of Biological Chemistry 295 (24), 8325–8330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jiao H. et al. (2020) Z-nucleic-acid sensing triggers ZBP1-dependent necroptosis and inflammation. Nature 580 (7803), 391–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang R. et al. (2020) Gut stem cell necroptosis by genome instability triggers bowel inflammation. Nature 580 (7803), 386–390. [DOI] [PubMed] [Google Scholar]
  • 96.Devos M. et al. (2020) Sensing of endogenous nucleic acids by ZBP1 induces keratinocyte necroptosis and skin inflammation. Journal of Experimental Medicine 217 (7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Pearson JS et al. (2017) EspL is a bacterial cysteine protease effector that cleaves RHIM proteins to block necroptosis and inflammation. Nat Microbiol 2, 16258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lamkanfi M. and Dixit VM (2010) Manipulation of host cell death pathways during microbial infections. Cell Host Microbe 8 (1), 44–54. [DOI] [PubMed] [Google Scholar]
  • 99.Rickard JA et al. (2014) TNFR1-dependent cell death drives inflammation in Sharpin-deficient mice. Elife 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Gerlach B. et al. (2011) Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 471 (7340), 591–6. [DOI] [PubMed] [Google Scholar]
  • 101.Vereecke L. et al. (2009) The ubiquitin-editing enzyme A20 (TNFAIP3) is a central regulator of immunopathology. Trends Immunol 30 (8), 383–91. [DOI] [PubMed] [Google Scholar]
  • 102.Peltzer N. et al. (2018) LUBAC is essential for embryogenesis by preventing cell death and enabling haematopoiesis. Nature 557 (7703), 112–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sharma BR et al. (2019) Innate immune adaptor MyD88 deficiency prevents skin inflammation in SHARPIN-deficient mice. Cell Death Differ 26 (4), 741–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Douglas T. et al. (2015) The Inflammatory Caspases-1 and −11 Mediate the Pathogenesis of Dermatitis in Sharpin-Deficient Mice. J Immunol 195 (5), 2365–73. [DOI] [PubMed] [Google Scholar]
  • 105.Gurung P. et al. (2016) Distinct role of IL-1β in instigating disease in Sharpin(cpdm) mice. Sci Rep 6, 36634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Boisson B. et al. (2015) Human HOIP and LUBAC deficiency underlies autoinflammation, immunodeficiency, amylopectinosis, and lymphangiectasia. J Exp Med 212 (6), 939–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Boisson B. et al. (2012) Immunodeficiency, autoinflammation and amylopectinosis in humans with inherited HOIL-1 and LUBAC deficiency. Nat Immunol 13 (12), 1178–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lee EG et al. (2000) Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science 289 (5488), 2350–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Onizawa M. et al. (2015) The ubiquitin-modifying enzyme A20 restricts ubiquitination of the kinase RIPK3 and protects cells from necroptosis. Nat Immunol 16 (6), 618–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Vande Walle L. et al. (2014) Negative regulation of the NLRP3 inflammasome by A20 protects against arthritis. Nature 512 (7512), 69–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Jiang X. et al. (2012) Expression of tumor necrosis factor alpha-induced protein 3 mRNA in peripheral blood mononuclear cells negatively correlates with disease severity in psoriasis vulgaris. Clin Vaccine Immunol 19 (12), 1938–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kelly C. et al. (2013) Expression of the inflammatory regulator A20 correlates with lung function in patients with cystic fibrosis. J Cyst Fibros 12 (4), 411–5. [DOI] [PubMed] [Google Scholar]
  • 113.Musone SL et al. (2008) Multiple polymorphisms in the TNFAIP3 region are independently associated with systemic lupus erythematosus. Nat Genet 40 (9), 1062–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Günther C. et al. (2011) Caspase-8 regulates TNF-α-induced epithelial necroptosis and terminal ileitis. Nature 477 (7364), 335–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Welz PS et al. (2011) FADD prevents RIP3-mediated epithelial cell necrosis and chronic intestinal inflammation. Nature 477 (7364), 330–4. [DOI] [PubMed] [Google Scholar]
  • 116.Bonnet MC et al. (2011) The adaptor protein FADD protects epidermal keratinocytes from necroptosis in vivo and prevents skin inflammation. Immunity 35 (4), 572–82. [DOI] [PubMed] [Google Scholar]
  • 117.Alvarez-Diaz S. et al. (2016) The Pseudokinase MLKL and the Kinase RIPK3 Have Distinct Roles in Autoimmune Disease Caused by Loss of Death-Receptor-Induced Apoptosis. Immunity 45 (3), 513–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kaiser WJ et al. (2011) RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471 (7338), 368–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Pierdomenico M. et al. (2014) Necroptosis is active in children with inflammatory bowel disease and contributes to heighten intestinal inflammation. Am J Gastroenterol 109 (2), 279–87. [DOI] [PubMed] [Google Scholar]
  • 120.Lehle AS et al. (2019) Intestinal Inflammation and Dysregulated Immunity in Patients With Inherited Caspase-8 Deficiency. Gastroenterology 156 (1), 275–278. [DOI] [PubMed] [Google Scholar]
  • 121.Gurung P. et al. (2016) NLRP3 inflammasome plays a redundant role with caspase 8 to promote IL-1β–mediated osteomyelitis. 113 (16), 4452–4457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Rhen T. and Cidlowski JA (2005) Antiinflammatory Action of Glucocorticoids — New Mechanisms for Old Drugs. New England Journal of Medicine 353 (16), 1711–1723. [DOI] [PubMed] [Google Scholar]
  • 123.Rodrigo C. et al. (2016) Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database of Systematic Reviews. [DOI] [PubMed] [Google Scholar]
  • 124.RECOVERY Collaborative Group. (2021) Dexamethasone in Hospitalized Patients with Covid-19. New England Journal of Medicine. 384, 693–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Henter JI et al. (1997) HLH-94: a treatment protocol for hemophagocytic lymphohistiocytosis. HLH study Group of the Histiocyte Society. Med Pediatr Oncol 28 (5), 342–7. [DOI] [PubMed] [Google Scholar]
  • 126.Hussell T. et al. (2001) Inhibition of tumor necrosis factor reduces the severity of virusspecific lung immunopathology. 31 (9), 2566–2573. [DOI] [PubMed] [Google Scholar]
  • 127.Peper RL and Van Campen H. (1995) Tumor necrosis factor as a mediator of inflammation in influenza A viral pneumonia. 19 (3), 175–183. [DOI] [PubMed] [Google Scholar]
  • 128.Zhang WJ et al. (1996) Lethal synergism between influenza infection and staphylococcal enterotoxin B in mice. J Immunol 157 (11), 5049–60. [PubMed] [Google Scholar]
  • 129.Gianfrancesco M. et al. (2020) Characteristics associated with hospitalisation for COVID19 in people with rheumatic disease: data from the COVID-19 Global Rheumatology Alliance physician-reported registry. Annals of the Rheumatic Diseases 79 (7), 859–866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Schmid JP et al. (2009) Neutralization of IFNγ defeats haemophagocytosis in LCMV-infected perforin- and Rab27a-deficient mice. EMBO Molecular Medicine 1 (2), 112–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Locatelli F. et al. (2020) Emapalumab in Children with Primary Hemophagocytic Lymphohistiocytosis. N Engl J Med 382 (19), 1811–1822. [DOI] [PubMed] [Google Scholar]
  • 132.Prencipe G. et al. (2018) Neutralization of IFN-gamma reverts clinical and laboratory features in a mouse model of macrophage activation syndrome. J Allergy Clin Immunol 141 (4), 1439–1449. [DOI] [PubMed] [Google Scholar]
  • 133.Schroder K. et al. (2004) Interferon-γ: an overview of signals, mechanisms and functions. Journal of Leukocyte Biology 75 (2), 163–189. [DOI] [PubMed] [Google Scholar]
  • 134.Kumar A. and Kato H. (2016) Macrophage Activation Syndrome Associated with AdultOnset Still’s Disease Successfully Treated with Anakinra. Case Reports in Rheumatology 2016, 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Bruck N. et al. (2011) Rapid and sustained remission of systemic juvenile idiopathic arthritis-associated macrophage activation syndrome through treatment with anakinra and corticosteroids. J Clin Rheumatol 17 (1), 23–7. [DOI] [PubMed] [Google Scholar]
  • 136.Loh NK et al. (2012) Successful treatment of macrophage activation syndrome complicating adult Still disease with anakinra. Intern Med J 42 (12), 1358–62. [DOI] [PubMed] [Google Scholar]
  • 137.Mehta P. et al. (2020) Silencing the cytokine storm: the use of intravenous anakinra in haemophagocytic lymphohistiocytosis or macrophage activation syndrome. Lancet Rheumatol 2 (6), e358–e367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Generali D. et al. (2021) Canakinumab as treatment for COVID-19-related pneumonia: A prospective case-control study. Int J Infect Dis 104, 433–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Norelli M. et al. (2018) Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nature Medicine 24 (6), 739–748. [DOI] [PubMed] [Google Scholar]
  • 140.Kullenberg T. et al. (2016) Long-term safety profile of anakinra in patients with severe cryopyrin-associated periodic syndromes. Rheumatology 55 (8), 1499–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kluger N. et al. (2011) Efficacy of anti-interleukin-1 receptor antagonist anakinra (Kineret®) in a case of refractory Sweet’s syndrome. Dermatology 222 (2), 123–7. [DOI] [PubMed] [Google Scholar]
  • 142.Weiss ES et al. (2018) Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood 131 (13), 1442–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Mazodier K. (2005) Severe imbalance of IL-18/IL-18BP in patients with secondary hemophagocytic syndrome. Blood 106 (10), 3483–3489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Girard-Guyonvarc’H C. et al. (2018) Unopposed IL-18 signaling leads to severe TLR9induced macrophage activation syndrome in mice. Blood 131 (13), 1430–1441. [DOI] [PubMed] [Google Scholar]
  • 145.Yasin S. et al. (2020) IL-18 as therapeutic target in a patient with resistant systemic juvenile idiopathic arthritis and recurrent macrophage activation syndrome. Rheumatology (Oxford) 59 (2), 442–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Tanaka T. et al. (2014) IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol 6 (10), a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kang S. et al. (2019) Targeting Interleukin-6 Signaling in Clinic. Immunity 50 (4), 1007–1023. [DOI] [PubMed] [Google Scholar]
  • 148.Kotch C. et al. (2019) Tocilizumab for the treatment of chimeric antigen receptor T cellinduced cytokine release syndrome. Expert Rev Clin Immunol 15 (8), 813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Stone JH et al. (2017) Trial of Tocilizumab in Giant-Cell Arteritis. New England Journal of Medicine 377 (4), 317–328. [DOI] [PubMed] [Google Scholar]
  • 150.Casper C. et al. (2015) Analysis of Inflammatory and Anemia-Related Biomarkers in a Randomized, Double-Blind, Placebo-Controlled Study of Siltuximab (Anti-IL6 Monoclonal Antibody) in Patients With Multicentric Castleman Disease. Clinical Cancer Research 21 (19), 4294–4304. [DOI] [PubMed] [Google Scholar]
  • 151.Nasonov E. and Samsonov M. (2020) The role of Interleukin 6 inhibitors in therapy of severe COVID-19. Biomedicine & Pharmacotherapy, 110698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Gordon AC et al. (2021) Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19. New England Journal of Medicine 384 (16), 1491–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Schwartz DM et al. (2017) JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nature Reviews Drug Discovery 16 (12), 843–862. [DOI] [PubMed] [Google Scholar]
  • 154.Stark GR et al. (2018) Responses to Cytokines and Interferons that Depend upon JAKs and STATs. Cold Spring Harbor Perspectives in Biology 10 (1), a028555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Albeituni S. et al. (2019) Mechanisms of action of ruxolitinib in murine models of hemophagocytic lymphohistiocytosis. Blood 134 (2), 147–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Meyer LK et al. (2020) JAK/STAT pathway inhibition sensitizes CD8 T cells to dexamethasone-induced apoptosis in hyperinflammation. Blood 136 (6), 657–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Kalil AC et al. (2021) Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. New England Journal of Medicine 384 (9), 795–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Andrades M. et al. (2011) Bench-to-bedside review: sepsis - from the redox point of view. Crit Care 15 (5), 230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Oda T. et al. (1989) Oxygen radicals in influenza-induced pathogenesis and treatment with pyran polymer-conjugated SOD. Science 244 (4907), 974–976. [DOI] [PubMed] [Google Scholar]
  • 160.Cruz MP (2018) Edaravone (Radicava): A Novel Neuroprotective Agent for the Treatment of Amyotrophic Lateral Sclerosis. P T 43 (1), 25–28. [PMC free article] [PubMed] [Google Scholar]
  • 161.Förstermann U. and Sessa WC (2012) Nitric oxide synthases: regulation and function. Eur Heart J 33 (7), 829–37, 837a-837d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Cauwels A. (2007) Nitric oxide in shock. Kidney International 72 (5), 557–565. [DOI] [PubMed] [Google Scholar]
  • 163.López A. et al. (2004) Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: Effect on survival in patients with septic shock. Critical Care Medicine 32 (1), 21–30. [DOI] [PubMed] [Google Scholar]
  • 164.Yamashita T. et al. (2000) Resistance to Endotoxin Shock in Transgenic Mice Overexpressing Endothelial Nitric Oxide Synthase. 101 (8), 931–937. [DOI] [PubMed] [Google Scholar]
  • 165.Cobb JP et al. (1999) Inducible nitric oxide synthase (iNOS) gene deficiency increases the mortality of sepsis in mice. 126 (2), 438–442. [PubMed] [Google Scholar]
  • 166.Hotchkiss RS et al. (2000) Caspase inhibitors improve survival in sepsis: a critical role of the lymphocyte. Nature Immunology 1 (6), 496–501. [DOI] [PubMed] [Google Scholar]
  • 167.Shiffman M. et al. (2019) Randomised clinical trial: emricasan versus placebo significantly decreases ALT and caspase 3/7 activation in subjects with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 49 (1), 64–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kudelova J. et al. (2015) Pharmacological caspase inhibitors: research towards therapeutic perspectives. J Physiol Pharmacol 66 (4), 473–82. [PubMed] [Google Scholar]
  • 169.Ho DD (1995) Time to hit HIV, early and hard. N Engl J Med 333 (7), 450–1. [DOI] [PubMed] [Google Scholar]
  • 170.Faulkner L. et al. (2005) The Mechanism of Superantigen-Mediated Toxic Shock: Not a Simple Th1 Cytokine Storm. The Journal of Immunology 175 (10), 6870–6877. [DOI] [PubMed] [Google Scholar]
  • 171.Kanneganti A. et al. (2018) GSDMD is critical for autoinflammatory pathology in a mouse model of Familial Mediterranean Fever. J Exp Med 215 (6), 1519–1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Falkenburg JH et al. (2008) T cell therapy in allogeneic stem cell transplantation. Biol Blood Marrow Transplant 14 (1 Suppl 1), 136–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Opal SM et al. (1997) Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. The Interleukin-1 Receptor Antagonist Sepsis Investigator Group. Crit Care Med 25 (7), 1115–24. [DOI] [PubMed] [Google Scholar]
  • 174.Qiu P. et al. (2013) Antitumor Necrosis Factor Therapy Is Associated With Improved Survival in Clinical Sepsis Trials. Critical Care Medicine 41 (10), 2419–2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Huet T. et al. (2020) Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol 2 (7), e393–e400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Clinical Trial of the Use of Anakinra in Cytokine Storm Syndrome Secondary to Covid-19 (ANA-COVID-GEAS) (ANA-COVID-GEAS). https://clinicaltrials.gov/ct2/show/NCT04443881 [Google Scholar]
  • 177.Rosas I. et al. (2021) Tocilizumab in Hospitalized Patients With Severe COVID-19 Pneumonia. New England Journal of Medicine 384, 1503–1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Salama C. et al. (2020) Tocilizumab in nonventilated patients hospitalized with Covid-19 pneumonia. medRxiv, 2020.10.21.20210203. [Google Scholar]
  • 179.Rodriguez-Lago I. et al. (2020) Characteristics and Prognosis of Patients With Inflammatory Bowel Disease During the SARS-CoV-2 Pandemic in the Basque Country (Spain). Gastroenterology 159 (2), 781–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Waggershauser CH et al. (2020) Letter: immunotherapy in IBD patients in a SARS-CoV2 endemic area. Aliment Pharmacol Ther 52 (5), 898–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Immune Modulators for Treating COVID-19 (ACTIV-1 IM). https://clinicaltrials.gov/ct2/show/NCT04593940 [Google Scholar]
  • 182.Efficacy and Safety of Emapalumab and Anakinra in Reducing Hyperinflammation and Respiratory Distress in Patients With COVID-19 Infection. https://clinicaltrials.gov/ct2/show/NCT04324021 [Google Scholar]
  • 183.Cantini F. et al. (2020) Baricitinib therapy in COVID-19: A pilot study on safety and clinical impact. J Infect 81 (2), 318–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Grom AA et al. (2016) Rate and Clinical Presentation of Macrophage Activation Syndrome in Patients With Systemic Juvenile Idiopathic Arthritis Treated With Canakinumab. Arthritis Rheumatol 68 (1), 218–28. [DOI] [PubMed] [Google Scholar]
  • 185.Quartier P. et al. (2011) A multicentre, randomised, double-blind, placebo-controlled trial with the interleukin-1 receptor antagonist anakinra in patients with systemic-onset juvenile idiopathic arthritis (ANAJIS trial). Ann Rheum Dis 70 (5), 747–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Yokota S. et al. (2008) Efficacy and safety of tocilizumab in patients with systemic-onset juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled, withdrawal phase III trial. Lancet 371 (9617), 998–1006. [DOI] [PubMed] [Google Scholar]
  • 187.Efficacy Study Of Tofacitinib In Pediatric JIA Population. https://www.clinicaltrials.gov/ct2/show/NCT02592434 [Google Scholar]
  • 188.Hasni S. et al. (2019) 183 A phase 1B/2A trial of tofacitinib, an oral janus kinase inhibitor, in systemic lupus erythematosus. Lupus Science & Medicine 6 (Suppl 1), A139. [Google Scholar]
  • 189.Nordström D. et al. (2012) Beneficial effect of interleukin 1 inhibition with anakinra in adultonset Still’s disease. An open, randomized, multicenter study. J Rheumatol 39 (10), 2008–2011. [DOI] [PubMed] [Google Scholar]
  • 190.Tocilizumab and Hemophagocytic Lymphohistiocytosis (HLH). https://clinicaltrials.gov/ct2/show/NCT02007239 [Google Scholar]
  • 191.Lounder DT et al. (2019) Treatment of refractory hemophagocytic lymphohistiocytosis with emapalumab despite severe concurrent infections. Blood Adv 3 (1), 47–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.A Study of TQ05105 Tablets in Subjects With Hemophagocytic Lymphohistiocytosis HLH). https://clinicaltrials.gov/ct2/show/NCT04326348 [Google Scholar]
  • 193.Deisseroth A. et al. (2015) FDA approval: siltuximab for the treatment of patients with multicentric Castleman disease. Clin Cancer Res 21 (5), 950–4. [DOI] [PubMed] [Google Scholar]
  • 194.Choi SW et al. (2012) TNF-inhibition with etanercept for graft-versus-host disease prevention in high-risk HCT: lower TNFR1 levels correlate with better outcomes. Biol Blood Marrow Transplant 18 (10) 1525–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Gatza E. et al. (2014) Etanercept plus topical corticosteroids as initial therapy for grade one acute graft-versus-host disease after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 20 (9), 1426–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Jagasia M. et al. (2020) Ruxolitinib for the treatment of steroid-refractory acute GVHD (REACH1): a multicenter, open-label phase 2 trial. Blood 135 (20), 1739–1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Strati P. et al. (2020) Clinical efficacy of anakinra to mitigate CAR T-cell therapyassociated toxicity in large B-cell lymphoma. Blood Adv 4 (13), 3123–3127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Le RQ et al. (2018) FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncologist 23 (8) 943–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Bresnihan B. et al. (1998) Treatment of rheumatoid arthritis with recombinant human interleukin-1 receptor antagonist. Arthritis Rheum 41 (12), 2196–2204. [DOI] [PubMed] [Google Scholar]
  • 200.Smolen JS et al. (2008) Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet 371 (9617), 987–997. [DOI] [PubMed] [Google Scholar]
  • 201.Maini R. et al. (1999) Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. ATTRACT Study Group. Lancet 354 (9194), 1932–1939. [DOI] [PubMed] [Google Scholar]
  • 202.Fleischmann R. et al. (2012) Placebo-Controlled Trial of Tofacitinib Monotherapy in Rheumatoid Arthritis. New England Journal of Medicine 367 (6), 495–507. [DOI] [PubMed] [Google Scholar]
  • 203.Giant Cell Arteritis and Anakinra Trial (GiAnT). https://clinicaltrials.gov/ct2/show/NCT02902731 [Google Scholar]
  • 204.Canakinumab in the Treatment of Acute Gout Flares and Prevention of New Flares in Patients Unable to Use Non-steroidal Anti-inflammatory Drugs (NSAIDs) and/or Colchicine Including a 12 Weeks Extension and an Open-label 48 Weeks Extension Study (β-RELIEVED). https://clinicaltrials.gov/ct2/show/NCT01029652 [Google Scholar]
  • 205.Study of the Safety and Effectiveness of Rilonacept for the Prevention of Gout Flares. https://clinicaltrials.gov/ct2/show/NCT00610363 [Google Scholar]
  • 206.Canakinumab in the Treatment of Acute Gout Flares and Prevention of New Flares in Patients Unable to Use Non-steroidal Anti-inflammatory Drugs (NSAIDs) and/or Colchicines Including a 12 Week Extension and a 1 Year Open-label Extension Study. (β-RELIEVED-II). https://clinicaltrials.gov/ct2/show/NCT01080131 [Google Scholar]
  • 207.Derkx B, et al. (1993) Tumour-necrosis-factor antibody treatment in Crohn’s disease. Lancet 342 (8864), 173–174. [DOI] [PubMed] [Google Scholar]
  • 208.Targan SR et al. (1997) A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s Disease cA2 Study Group. New England Journal of Medicine 337 (15), 1029–1035. [DOI] [PubMed] [Google Scholar]
  • 209.Hommes D. et al. (2004) Fontolizumab (Huzaf), a humanized anti-IFN-gamma antibody, has clinical activity and excellent tolerability in moderate to severe Crohn’s disease. Gastroenterology 127 (1), 332. [Google Scholar]
  • 210.Rogler G. (2020) Efficacy of JAK inhibitors in Crohn’s Disease. Journal of Crohn’s and Colitis 14 (Supplement_2), S746–S754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Rutgeerts P. et al. (2005) Infliximab for induction and maintenance therapy for ulcerative colitis. New England Journal of Medicine 353 (23), 2462–2476. [DOI] [PubMed] [Google Scholar]
  • 212.Sandborn WJ et al. (2017) Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. New England Journal of Medicine 376 (18), 1723–1736. [DOI] [PubMed] [Google Scholar]
  • 213.Gibbs A. et al. (2005) Anakinra (Kineret) in psoriasis and psoriatic arthritis: a single-center, open-label, pilot study. Arthritis Res Ther 7 (Suppl 1), P68. [Google Scholar]
  • 214.Papp KA et al. (2005) A global phase III randomized controlled trial of etanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br J Dermatol 152 (6), 1304–1312. [DOI] [PubMed] [Google Scholar]
  • 215.Merola JF et al. (2017) Efficacy of tofacitinib for the treatment of nail psoriasis: Two 52week, randomized, controlled phase 3 studies in patients with moderate-to-severe plaque psoriasis. J Am Acad Dermatol 77 (1), 79–87.e1. [DOI] [PubMed] [Google Scholar]
  • 216.Papp KA et al. (2015) Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: results from two randomized, placebo-controlled, phase III trials. Br J Dermatol 173 (4), 949–961. [DOI] [PubMed] [Google Scholar]
  • 217.Goldbach-Mansky R. et al. (2006) Neonatal-onset multisystem inflammatory disease responsive to interleukin-1beta inhibition. New England Journal of Medicine 355 (6), 581–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Hawkins P, Lachmann HJ, McDermott MF (2003) Interleukin-1-receptor antagonist in the Muckle-Wells syndrome. New England Journal of Medicine 348 (25), 2583–2584. [DOI] [PubMed] [Google Scholar]
  • 219.De Benedetti F. et al. (2018) Canakinumab for the Treatment of Autoinflammatory Recurrent Fever Syndromes. New England Journal of Medicine 378 (20), 1908–1919. [DOI] [PubMed] [Google Scholar]
  • 220.Harrison SA et al. (2020) A randomized, placebo-controlled trial of emricasan in patients with NASH and F1-F3 fibrosis. J Hepatol 72 (5), 816–827. [DOI] [PubMed] [Google Scholar]
  • 221.Berry MA et al. (2006) Evidence of a role of tumor necrosis factor alpha in refractory asthma. New England Journal of Medicine 354 (7), 697–708. [DOI] [PubMed] [Google Scholar]
  • 222.Leisman DE et al. (2020) Cytokine elevation in severe and critical COVID-19: a rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. The Lancet Respiratory Medicine 8 (12), 1233–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Mudd PA et al. (2020) Distinct inflammatory profiles distinguish COVID-19 from influenza with limited contributions from cytokine storm. Sci Adv. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Blanco-Melo D. et al. (2020) Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 181 (5), 1036–1045.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Lee JS et al. (2020) Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19. Science Immunology 5 (49), eabd1554. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES