- Review
- Open access
- Published:
Hemophagocytic lymphohistiocytosis: current treatment advances, emerging targeted therapy and underlying mechanisms
Journal of Hematology & Oncology volume 17, Article number: 106 (2024)
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rapidly progressing, life-threatening syndrome characterized by excessive immune activation, often presenting as a complex cytokine storm. This hyperactive immune response can lead to multi-organ failure and systemic damage, resulting in an extremely short survival period if left untreated. Over the past decades, although HLH has garnered increasing attention from researchers, there have been few advancements in its treatment. The cytokine storm plays a crucial role in the treatment of HLH. Investigating the detailed mechanisms behind cytokine storms offers insights into targeted therapeutic approaches, potentially aiding in early intervention and improving the clinical outcome of HLH patients. To date, there is only one targeted therapy, emapalumab targeting interferon-γ, that has gained approval for primary HLH. This review aims to summarize the current treatment advances, emerging targeted therapeutics and underlying mechanisms of HLH, highlighting its newly discovered targets potentially involved in cytokine storms, which are expected to drive the development of novel treatments and offer fresh perspectives for future studies. Besides, multi-targeted combination therapy may be essential for disease control, but further trials are required to determine the optimal treatment mode for HLH.
Introduction
Hemophagocytic lymphohistiocytosis (HLH) is a fatal disease characterized by pathological immune activation and dysregulated inflammation that cause widespread tissue damage and multi-organ failure [1]. The first case related to HLH under the term “Histyocytic Medullary Reticulosis” was reported in 1939 by Scott and Robb-Smith in their seminal series of articles [2], and the inherited kind of HLH was then recognized in the mid-twentieth century with the name of familial hemophagocytic reticulosis [3] (Fig. 1). With the advancement of genetic technology, the subsequent identification of HLH-related gene mutations has enhanced the understanding of familial HLH and highlighted the importance of distinguishing between inherited (primary) and acquired (secondary) forms of the syndrome.
Timeline of the history of hemophagocytic lymphohistiocytosis (HLH). The figure illustrates the key milestones in the discovery of HLH over the past 80 years. HLH was first described in 1939, and since then, various types of primary and secondary HLH have been defined gradually. Mutations in genes associated with HLH have also been progressively discovered. CHS chédiak-higashi syndrome, GS-2 griscelli syndrome type 2, LPI lysinuric protein intolerance, X-SCID X-linked severe combined immunodeficiency, XLP-1 X-linked lymphoproliferative sisease-1, HPS-2 hermansky–pudlak syndrome 2, XLP-2 X-linked lymphoproliferative disease-2, HIDS hyper-IgD syndrome, MKD mevalonate kinase deficiency, XLA X-linked agammaglobulinemia, ALPS autoimmune lymphoproliferative syndrome
The main clinical manifestations of HLH include fever, hepatosplenomegaly, lymphadenopathy, cytopenia, hyperferritinemia, hypertriglyceridemia, hypofibrinogenemia and multiorgan dysfunction, which may also lead to neurological symptoms [4]. HLH, diagnosed according to the HLH-2004 criteria and HScore, can be classified into primary HLH (pHLH) and secondary HLH (sHLH) based on the presence of underlying genetic defects (Fig. 2). Primary HLH is a rare but severe genetic immune system disorder, primarily caused by a group of genetic mutations associated with immune dysfunction such as LYST, SH2D1A, PRF1, etc [5,6,7,8,9]. Since allogeneic hematopoietic stem cell transplantation (HSCT) can effectively control the development of pHLH, early genetic testing to identify gene abnormalities for pHLH diagnosis is crucial for subsequent treatment and prognosis. On the other hand, patients with sHLH are believed to develop the syndrome as a complication triggered by various diseases, such as infection, malignancy, autoimmune disease, etc. Specifically, HLH secondary to rheumatic or autoinflammatory diseases is also referred to as macrophage activation syndrome (MAS), which is commonly seen in systemic juvenile idiopathic arthritis (sJIA), systemic lupus erythematosus (SLE), Kawasaki disease, and adult Still's disease (AOSD) [10,11,12,13]. Notably, it is a prerequisite for the diagnosis of sHLH to exclude any mutations in these known affected genes.
Diagnostic process for hemophagocytic lymphohistiocytosis (HLH). This figure illustrates the diagnostic strategy for HLH, from initial clinical suspicion to differential diagnosis. One recent pHLH diagnostic guideline recommended incorporating functional testing of NK cells and cytotoxic T cells into the FHL diagnostic criteria, and proposed that the HLH-2004 criteria without testing NK cell function had a higher diagnostic accuracy for FHL at 99.0% (sensitivity 96.2%; specificity 99.5%) [464]. It is noteworthy that HLH can be the initial presentation of an undiagnosed malignancy, and malignancy-associated HLH typically indicates a poor prognosis [465]. The diagnosis of HLH still primarily relies on the HLH-2004 criteria, which often lack specificity in differentiating HLH from other hyperinflammatory disorders, especially in the context of malignancy. Therefore, the importance of imaging studies and biopsies should be emphasized, and all HLH patients may need to undergo tumor screening, and treating the primary malignancy is crucial for improving prognosis. MAS macrophage activation syndrome, LDH lactate dehydrogenase, EBV epstein-barr virus, PET-CT positron emission tomography-computed tomography, ANA Antinuclear Antibody, ENA extractable nuclear antigen, ANCA anti-neutrophil cytoplasmic antibody, FHL familial hemophagocytic lymphohistiocytosis, XLP X-linked lymphoproliferative disease, XIAP X-linked inhibitor of apoptosis protein, CMV cytomegalovirus, HIV human immunodeficiency virus, sJIA systemic juvenile idiopathic arthritis, SLE systemic lupus erythematosus, AOSD adult-onset still's disease, AST aspartate aminotransferase, Hgb hemoglobin, Plt platelet, Abs Neut absolute neutrophil count
The reported 1-month mortality rate was 27.7% and the 1-year survival rate was 50% among HLH patients, underscoring the urgent need for the development of HLH treatment [14, 15]. The HLH-94 regimen remains to be the first-line treatment for controlling acute inflammation in HLH, but the therapeutic resistance and mortality rates are still clinically unacceptable [16, 17]. For pHLH patients with clear HLH-related genetic mutations or those with relapsed and refractory HLH, HSCT can be an option. However, progress in HLH treatment has been limited over the past decades, largely due to the unclear pathogenesis of HLH. For both primary and secondary HLH, the key treatment goal is to control the excessive secretion of inflammatory cytokines, including interleukin-2 (IL-2), IL-6, IL-18, interferon-γ (IFN-γ), etc. Therefore, targeting the inflammatory cytokines to inhibit the cytokine storm is one of the important treatment strategies for HLH. In 2018, the monoclonal antibody targeting IFN-γ, emapalumab, has gained global approval for the treatment of HLH, marking the beginning of targeted therapies for HLH [18].
Hence, considering the delay in exploring HLH diagnosis and treatment, this review aims to provide a comprehensive overview of the preclinical and clinical advances in HLH therapy, highlighting potential innovative strategies targeting inflammatory cytokines and related key molecules. A deep understanding of potential therapeutic targets for HLH can further guide the design of clinical trials and help elucidate their roles in HLH development and disease control.
HLH pathogenesis
Regardless of whether HLH is the primary or secondary subtype, its pathogenesis and development involve a series of proinflammatory cytokines such as IFNγ, IL-1β, IL-6, IL-18, and TNF-α. T cells, NK cells and macrophages are predominantly responsible for the increased secretion of these inflammatory cytokines. However, despite having a hyperinflammatory storm similar to the cytokine release syndrome [19, 20], the specific pathogenic mechanisms of HLH remain not well understood. Nearly all types of HLH patients showed similar clinical manifestations, characterized by a systemic hyperinflammatory syndrome caused by a cytokine storm, leading to widespread tissue damage and multi-organ failure [21].
Under the normal condition, NK cells and cytotoxic T cells (CTLs) recognize target cells upon contact and form an immunological synapse, which then directionally release perforin and granzymes through exocytosis (Fig. 3) [22,23,24,25]. Perforin forms pores in the cell membrane, allowing granzymes to enter the target cell, triggering a series of enzymatic reactions that ultimately lead to apoptosis of the target cell [25]. However, in patients with pHLH, mutations in genes related to granule release function (such as PRF1, UNC13D, STX11, and STXBP2) prevent NK/cytotoxic T cells from effectively eliminating infected or abnormal cells, thereby prolonging the existence of the immunological synapse and leading to excessive production of inflammatory cytokines [26,27,28]. Simultaneously, the immune cells with impaired granule function fail to terminate the activation of antigen-presenting cells (macrophages, monocytes, and dendritic cells), resulting in sustained activation and proliferation of T cells, which further produce pro-inflammatory cytokines like IFNγ, thus forming a cytokine storm [29]. Although patients with sHLH do not usually have lymphocyte dysfunction caused by genetic abnormalities, the cytokine storm is usually triggered by external factors like infections, malignancies or autoimmune diseases that lead to excessive activation of macrophages (Fig. 3).
Schematic diagram of the pathogenesis of hemophagocytic lymphohistiocytosis (HLH). The impaired ability of natural killer cells and cytotoxic T cells to secrete perforin and granzyme results in defective clearance of target cells, leading to sustained immune cell activation and excessive production of cytokines such as interleukin-1 (IL-1), IL-6, IL-18, and tumor necrosis factor-α (TNF-α). The intense pro-inflammatory response mediated by macrophages may also be due to increased production of autoantibodies and immune complexes, resulting in abnormal immune system activation and subsequently persistent inflammatory reactions. Infections, malignancies, and immune checkpoint inhibitors can also lead to excessive immune system activation, causing hyperactivity of macrophages and T cells, which release large amounts of pro-inflammatory cytokines, resulting in a cytokine storm
Currently reported HLH-related genes can be broadly classified into the following categories: Familial HLH genes (PRF1 [5], UNC13D [30], STX11 [31], STXBP2 [32]), X-linked lymphoproliferative disease genes (SH2D1A [33], XIAP [34]), pigment abnormality genes (LYST [35], RAB27A [35], AP3B1 [36]), immune deficiency genes (IL2RG [37], WAS [38], CGD [39], BTK [40], ITK [41], FAS [42], NLRC4 [43], CD27 [44], CDC42 [45], ZNFX1 [46], deletion of 22q11.2 [47]), and some inborn errors of metabolism genes (SLC7A7 [48], PNP [49], MVK [50], ADA [51]). Genes affecting cellular degranulation include Familial HLH genes and pigment abnormality genes. Mechanisms of HLH caused by mutations in genes that do not affect degranulation mainly involve altering the function, proliferation and signal transduction of immune cells, rather than directly inhibiting the release of cytotoxic granules.
HLH therapy
Remission induction
The treatment of HLH is primarily divided into two phases: controlling excessive inflammation and replacing the defective immune system. The standard treatment for the first phase is based on chemotherapy with etoposide (HLH-94 treatment protocol), while the second phase typically involves achieving remission through allogenic HSCT (allo-HSCT) following myeloablative/reductive conditioning [52]. In the early 1990s, researchers from the International Histiocyte Society proposed a treatment regimen consisting of etoposide and corticosteroids (HLH-94), suggesting the use of etoposide (150 mg/m2 intravenous injection, twice weekly during weeks 1–2, then weekly during weeks 3–8) in combination with dexamethasone [53]. This regimen significantly improved the survival rate of HLH patients [54], which has become the standard therapy for all types of HLH/MAS with lymphocytes and macrophages hyperactivated. Glucocorticoids can suppress the activation, differentiation and chemotaxis of inflammatory cytokines, thus controlling HLH characterized by excessive release of inflammatory cytokines. Etoposide is a widely used chemotherapeutic agent that inhibits topoisomerase II, and its mechanism for HLH treatment may involve effectively and selectively eliminating activated T cells and inhibiting the production of inflammatory cytokines [55, 56]. Subsequently, the HLH 2004 trial was conducted, suggesting that adding cyclosporine to the HLH-94 regimen did not help control acute immune activation [57]. The early response to etoposide could quite effectively predict the later mortality rate, but a small fraction of patients did not respond well [58, 59]. A potential drawback of etoposide-based therapy is bone marrow suppression, with some patients experiencing invasive fungal or bacterial infections during treatment.
About 30% of HLH patients did not respond to the standard HLH-94 protocol, and lower than 60% of them achieved disease-free survival through this regimen [60]. Previous studies suggested that the L-DEP regimen (PEG-asparaginase combined with liposomal doxorubicin, etoposide, and methylprednisolone) showed some efficacy as salvage therapy for refractory Epstein-Barr virus (EBV)-related HLH, achieving an overall response rate (ORR) of approximately 80% and a significant reduction in EBV-DNA load [61,62,63]. However, a significant decrease in early EBV-DNA load did not predict better long-term outcomes; therefore, once complete remission is achieved, allo-HSCT should be promptly considered [61].
Allogeneic hematopoietic stem cell transplantation
Although chemotherapy based on the HLH 94/04 protocol can be used for initial treatment, allo-HSCT remains to be the only potentially curative treatment for HLH [64]. However, when transplantation is performed in patients with active disease, allo-HSCT appears to be associated with adverse outcomes. Lai et al. reported in 2018 that the survival rate was higher than 50% when patients underwent allo-HSCT after achieving remission; however, the survival rate was only 33% when patients had active HLH before allo-HSCT [65]. It was reported that a reduced-intensity conditioning (RIC) regimen seemed to be more beneficial [66, 67]. Moreover, utilizing early alemtuzumab before RIC regimen had great tolerability and efficacy [66, 67].
However, it is important to be vigilant that HLH can also occur post allogeneic and autologous HSCT, especially associated with graft-versus-host disease (GVHD) in patients undergoing allo-HSCT [68, 69]. HLH typically occurs in the early phase, within 2–6 weeks post allo-HSCT [70]. Infections, particularly EBV and cytomegalovirus (CMV), can be triggering factors for HLH. Mortality rates were found obviously high in patients with HLH secondary to infections [71]. Multicenter studies reported an estimated incidence of HLH post allo-HSCT at 1.09%, significantly lower after autologous HSCT at 0.15% [72].
Gene therapy and adoptive T cell therapy
Gene therapy, utilizing virus vector-mediated gene transfer into autologous hematopoietic stem cells, has been demonstrated to cure various severe monogenic immunodeficiencies [73, 74]. One preclinical studies in Prf−/− mouse models suggested a significant correction of cytotoxic defects both in vitro and in vivo upon transplantation of PRF1 gene-corrected hematopoietic stem cells and CD8+ T cells [75, 76]. Jinx mice were used as a preclinical mouse model for familial HLH 3 (FHL3). Studies showed that transferring the lentiviral UNC13D gene into Jinx hematopoietic stem cells (HSCs) could restore T cell function in transplanted Jinx mice [77, 78]. Moreover, using lentivirus as a vector restored Munc13-4 expression and degranulation capacity in T cells from FHL3 patients and HSCs from FHL3 disease model mice [78]. The further research demonstrated that effective gene editing of Jinx mouse HSCs resulted in functional T cell responses with a diverse T cell receptor (TCR) repertoire, exhibiting rapid virus clearance and protection against HLH [74]. In X-linked lymphoproliferative disease (XLP)-1 mouse models, HSC gene correction was also able to improve the immunological manifestations of the disease and overcome HSCT-related complications [79].
Adoptive T cell therapy (ATCT) was found to be able to partially restore cellular cytolytic activity in HLH. Kristoffer et al. transferred functional virus-specific T cells into mice models of pHLH (Prf−/− mice and Jinx mice) [80]. The transferred T cells eliminated HLH-inducing viral triggers, silenced disease processes, cured excessive inflammation in Jinx mice and protected HLH mice from fatal HLH progression, without life-threatening side effects. The cured mice were able to avoid HLH recurrence in the long term [80].
Therapy for CNS-HLH
Primary HLH may present with isolated neurological symptoms, which can occur months before the appearance of systemic manifestations of HLH [81]. Therefore, performing a lumbar puncture for screening is necessary to differentiate the central nervous system involvement of HLH (CNS-HLH) from similar conditions such as demyelinating syndromes, chronic infections, malignancies and CNS vasculitis. For CNS-HLH patients, in addition to the systemic treatment, intrathecal injections of methotrexate and dexamethasone can be additionally performed. Receiving HSCT after the systemic treatment of HLH appears to be crucial for improving survival and neurological outcomes [82].
Emerging targeted therapy and mechanisms for HLH
In 20–30% of adult cases, HLH is refractory to first-line treatment or relapses after initial remission [83]. There is still lack of standardized treatment approach for relapsed/refractory pHLH patients, and cure is often achieved only through allo-HSCT, but approximately 20–25% of HLH patients died before transplantation [60, 84, 85]. It is worth mentioning that treatment delay was reported to be an independent poor prognostic factor for HLH, reflecting the importance of early selection of the appropriate treatment to break the cycle of immune dysregulation [86].
Due to the critical role of excessive immune activation and elevated cytokinemia in the pathogenesis of HLH, several targets have been proposed in recent years, including IFN-γ, Janus kinase-signal transducer and activator of transcription (JAK-STAT), IL-6, TNF-α, IL-1, IL-18, CD52, CD20 and programmed cell death protein 1 (PD-1) (Fig. 4). Preclinical studies and clinical investigations, including clinical trials and exploratory clinical studies, concerning the aforementioned targets were also compiled and summarized (Tables 1, 2, 3, 4).
Targeting IFN-γ
IFN-γ, belonging to type II interferons, is a soluble cytokine produced by T lymphocytes, macrophages, NK cells, and other immune cells [87]. The production of IFN-γ is mainly regulated by cytokine stimulation (such as IL-18), antigen stimulation, and other immune stimuli [88, 89]. IFN-γ can bind to the IFN-γ receptor (IFNGR), activate the JAK-STAT pathway and induce the expression of IFN-γ-stimulated genes, playing important roles in tissue homeostasis, immunity, inflammation and tumor immune surveillance [87, 90]. IFN-γ binds to its cell surface receptor IFNGR1 and induces IFNGR1 dimerization, then binding to two IFNGR2 to form a receptor complex. In this receptor complex, IFNGR1 activates the JAK1 kinase, while IFNGR2 activates the JAK2 kinase [91]. Activation of JAK1 and JAK2 can lead to receptor phosphorylation, recruiting and phosphorylating STAT1 [91, 92]. Phosphorylated STAT1 forms dimers and trans-locates to the nucleus, where it can bind to the Gamma-activated sequence (GAS) in the promoter region of target genes, thereby regulating the transcription of downstream genes [93]. Many genes regulated by the IFN-γ/STAT1 signaling pathway are transcription factors, thus the IFN-γ/STAT1 signaling pathway indirectly regulates the expression of more downstream genes [94]. Meanwhile, the IFN-γ/STAT1 signaling pathway can activate MAPK, PI3K-AKT, and NF-κB signaling pathways, enabling IFN-γ/STAT1 to participate in the regulation of the expression of more genes [95].
The loss of cytotoxic function in CD8+ T cells can lead to immune imbalance, promoting abnormal and excessive production of IFN-γ [96]. IFN-γ is a classical activator of macrophages and mediates polarization of macrophages towards the M1 phenotype [87, 96]. M1 macrophages exhibit strong pro-inflammatory properties and release inflammatory mediators such as IL-1β, IL-6, TNF-α, etc [97, 98]. Peripheral levels of IFN-γ were elevated in both primary and secondary HLH patients, with its levels correlating with clinical status, being elevated in active HLH but lower than detection levels in remission patients and healthy controls [99, 100]. Furthermore, one research has shown that IFN-γ was associated with liver function damage and coagulation disorders, and could directly act on macrophages in vivo, altering phagocytic activity and stimulating blood cell uptake, leading to severe anemia [100,101,102].
In one pre-clinical study, the anti-IFN-γ antibody significantly improved bone marrow function and survival in perforin-deficient mice after lymphocytic choriomeningitis virus (LCMV) infection [103]. In a mouse model of CpG DNA induced sHLH, the development of HLH was also found to be IFN-γ dependent [104]. However, the efficacy of anti-IFN-γ antibody in secondary HLH murine models is limited, possibly due to the distinct biological mechanisms between pHLH and sHLH [105].
Emapalumab is a fully human IgG1 monoclonal antibody targeting IFN-γ, capable of binding both free-form and receptor-bound IFN-γ (inhibiting receptor dimerization and IFN-γ signal transduction) and neutralizing its biological activity [106]. It is the first targeted therapy approved for HLH treatment, especially beneficial to patients unresponsive to conventional treatment [106, 107]. A phase 2–3, open-label, single-group study demonstrated that 27 relapsed/refractory (r/r) pHLH patients treated with a combination of emapalumab, dexamethasone and others achieved a remission rate of 63%, with a low incidence rate of adverse events [108]. Another potential advantage of IFN-γ blockade therapy for pHLH may be its ability to improve engraftment in allo-HSCT, preventing and treating graft failure [109,110,111]. In a prospective single-arm trial involving 14 patients with treatment-refractory HLH/ MAS who did not respond to high-dose corticosteroids (with or without anakinra), all clinical and laboratory parameters showed rapid improvement after treatment with anti-IFN-γ [112]. By week 8, 13 out of 14 patients achieved remission within a median time of 25 days [112]. Therefore, emapalumab may be an important additional treatment option for HLH, and patients often respond well to it, which is helpful in gradually discontinuing steroids [113]. When used in combination with etoposide, there is rarely a need for etoposide administration more frequently than once a week or every two weeks [113].
Patients with HLH often have concurrent infections. There have been case reports of refractory HLH patients with multiple severe and complicated infections that were treated with emapalumab and supportive antimicrobial therapy [114, 115]. Following this treatment regimen, all clinical symptoms and laboratory parameters gradually became normalized. Additionally, in patients with HLH complicated by severe infection, the inhibition of the inflammatory state by blocking IFN-γ allowed for discontinuation of conventional immunosuppressive therapy, aiding in infection control [115].
In terms of drug safety, emapalumab dosages can be gradually increased from 1 to 10 mg/kg twice weekly based on patient tolerance and clinical progress [116]. Prior to administering emapalumab infusions, latent tuberculosis infection should be excluded through interferon-γ release assays, and EBV and CMV infections should be monitored every two weeks [117]. Additionally, the adjunctive use of acyclovir and trimethoprim-sulfamethoxazole should be considered to prevent herpes zoster and pneumocystis jirovecii infections [117, 118].
However, LCMV infection in IFN-γ−/− and Prf1−/− mice still result in severe HLH-like state, suggesting that the driving cytokines for human HLH are not limited to IFN-γ [119]. Therapies targeting upstream activators of CD8+ T cells, such as interleukin-33/ST2 signaling, can be considered [119]. Additionally, in previous reports, HLH patients often received combination therapy rather than IFN-γ monoclonal antibodies alone. It is conceivable that solely inhibiting IFN-γ may not be sufficient to control the disease in the majority of patients. Targeting multiple cytokines simultaneously may be considered, but further clinical trials are warranted for validation [120].
Targeting JAK-STAT
The classical JAK-STAT pathway transduces extracellular signals activated by cytokines to the nucleus, mediating gene expressions and playing indispensable roles in a range of cellular processes, particularly those with immunomodulatory functions [121, 122]. The JAK family comprises a group of tyrosine kinases associated with cell signal transduction, primarily consisting of four members: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2). JAK1, JAK3 and TYK2 are responsible for immune system development and regulation, while JAK2 primarily participates in hematopoiesis, playing crucial roles in erythrocyte and platelet production [123,124,125]. The enzymatic function of JAK is activated by the binding of cytokines to their receptors. Cytokine-activated JAK phosphorylates tyrosine residues of each other and the intracellular tails of receptor subunits, thereby creating docking sites to recruit downstream signaling molecules [126]. A key subset of substrates binding to phosphorylated cytokine receptors is the STAT family of DNA-binding proteins. Receptor-bound STATs are phosphorylated by JAK, dimerize, and translocate to the nucleus, where they bind to DNA, activating gene transcription [121]. Mammals have seven STATs: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6 [127]. Through selective binding to cytokine receptors, different cytokines have the ability to preferentially recruit different STATs [128, 129].
In HLH patients, the JAK-STAT signaling pathway can be aberrantly activated due to immune dysregulation. Elevated cytokines in HLH, such as IFN-γ, IL-2, IL-6, IL-10, IL-12 and granulocyte–macrophage colony-stimulating factor (GM-CSF), can all signal through the JAK-STAT pathway [129]. By inhibiting downstream signaling of many HLH-related cytokines, such as the JAK-STAT pathway we discussed here, it is possible to effectively alleviate the immune response associated with HLH. Several JAK inhibitors, such as ruxolitinib, tofacitinib, baricitinib and oclacitinib, have been used in the treatment of inflammatory diseases [130]. Preclinical study have indicated that the JAK1/2 inhibitor ruxolitinib was more effective in treating HLH compared to the JAK1 inhibitor itacitinib and the JAK2 inhibitor fedratinib [131].
Ruxolitinib is an orally administered, potent and highly bioavailable JAK1/2 inhibitor, approved by the Food and Drug Administration (FDA) for patients with myeloproliferative neoplasms and steroid-refractory GVHD [132, 133]. Some studies using ruxolitinib to treat Prf1−/− or Rab27a−/− mice infected with LCMV (pHLH model; Table 3), as well as wild-type mice exposed to repeated injections of CpG DNA (sHLH model), have demonstrated that monotherapy with ruxolitinib reversed a series of HLH manifestations and significantly prolonged survival [134,135,136]. Both Ruxolitinib and the anti-IFN-γ antibody improved hemoglobin levels, but only ruxolitinib significantly reduced the number and activation status of immune cells, thus decreasing the frequency and absolute numbers of infiltrating CD8+ cells, monocytes and neutrophils [105]. Using the pHLH mouse model (Prf1−/− mice), the combination of ruxolitinib (4 mg/kg, twice a day) with low-dose anti-IFN-γ antibodies (200 μg per mouse, every 3 days) showed a synergistic effect, effectively alleviating HLH manifestations [137]. However, studies have also shown that higher doses of ruxolitinib (90 mg/kg, twice daily) combined with anti-IFN-γ antibodies (500 µg or 1 mg, administered once every 3–4 days) did not provide superior anti-inflammatory benefits compared to their individua use [138, 139]. Therefore, caution should be exercised when combining these two classes of drugs, especially when higher doses are used [138, 139]. Exploratory studies of combination therapy with dexamethasone and ruxolitinib have found that by blocking cytokine signaling, ruxolitinib can sensitize CD8+ T cells to dexamethasone-induced apoptosis in vitro, effectively overcoming cytokine-induced dexamethasone resistance [136].
For patients with r/r HLH, ruxolitinib has shown promising efficacy in improving the inflammatory state. A study described the use of ruxolitinib in combination with corticosteroids to treat 34 patients with r/r HLH (median age 27.5 years; 1 case of FHL2, 25 cases of EBV-HLH, 2 cases of HLH/MAS, 6 cases unclear) [140]. After two weeks of treatment, ferritin and sCD25 levels significantly decreased, indicating an improvement in the inflammatory state. The ORR was evaluated to be 73.5%, with complete response (CR) in 14.7% and partial response (PR) in 58.8% [140]. However, for patients with EBV-HLH, EBV-DNA levels remained unchanged, suggesting that ruxolitinib reduced inflammation without targeting the underlying cause of HLH, thereby still necessitating the need for allo-HSCT [140, 141]. Another study including 41 r/r HLH patients who had not previously received the DEP or L-DEP regimen showed an ORR of 78.0% with Ru-DEP (ruxolitinib-DEP) treatment [142]. A total of 8 cases (19.5%) achieved CR and 24 cases (58.5%) achieved PR. The CR rate with Ru-DEP was higher than with ruxolitinib monotherapy (14.7%) [142]. Although the response rate with the Ru-DEP regimen (76.2%) was similar to that observed in adult patients with refractory HLH treated with DEP in previous studies, 7 cases still achieved PR (58.3%) among the 12 HLH patients who had failed or relapsed after prior DEP or L-DEP treatment [142]. Some studies have also reported the efficacy of ruxolitinib in suppressing the inflammatory state in pHLH patients, potentially making it a safe bridge therapy for refractory HLH undergoing allo-HSCT [141, 143, 144].
For HLH patients with severe infections, ruxolitinib also demonstrated promising efficacy. Sostad et al. and Zandvakili et al. reported that two cases of sHLH with severe fungal infections showed clinical improvement after receiving ruxolitinib and antimicrobial agents as first-line treatment [145, 146]. Additionally, ruxolitinib also showed favorable outcomes in treating patients with malaria-, tuberculosis-, HIV-, SLE- and lymphoma-associated HLH [147,148,149,150]. There were also case reports of patients with central nervous system-involved r/r HLH achieving remission after receiving emapalumab combined ruxolitinib, followed by transplantation [151].
Overall, ruxolitinib is effective in inflammation control, but cannot eradicate the underlying cause. Nevertheless, allo-HSCT should still be considered the ultimate treatment following ruxolitinib. During drug administration, caution should be exercised regarding the side effects associated with JAK inhibitors, which may be related to off-target effects [152]. The use of JAK inhibitors increases the risk of severe and opportunistic infections, with reactivation of varicella-zoster virus being one of the most common infectious complications [152]. JAK inhibitor therapy further leads to anemia and decreased counts of lymphocytes, NK cells, neutrophils and platelets, possibly due to the inhibition of signaling pathways by cytokines such as JAK2 (e.g., erythropoietin, thrombopoietin) and other hematopoietic growth factors (e.g., IL-6 and IL-11) [139]. During usage, a balance between the therapeutic effects of the disease and the risks of side effects should be considered, ensuring that patients receive optimal treatment outcomes while minimizing adverse reactions.
Targeting IL-6
IL-6 is a core participant involved in acute inflammatory responses, which mediates the acute phase responses during the immune defense and induces the production of inflammation-related biomarkers such as C-reactive protein and procalcitonin [153, 154]. In addition, IL-6 can be used for diagnosis of early inflammation and provide an early warning for the occurrence of sepsis [155,156,157]. Upon encountering pathogen-associated molecular patterns (PAMPs) or damage associated molecular patterns (DAMPs), a variety of innate immune cells such as macrophages and monocytes rapidly initiate the expression and release of IL-6 to eliminate infected cells or damaged tissue [158,159,160]. IL-6 is mainly activated by the signals of IL-1β and TNF-α, with positively regulating by a series of small molecules, including platelet-derived growth factor (PDGF), lipopolysaccharide (LPS), phorbol myristate acetate (PMA), etc. [161,162,163,164,165,166]. However, overproduction of IL-6 can also result in chronic inflammatory diseases, such as SLE, rheumatoid arthritis, etc., as well as fetal cytokine storm-related conditions of receiving chimeric antigen receptor-T cell therapy, suffering severe coronavirus disease 2019 (COVID-19) and HLH [29, 167,168,169,170,171,172]. In sJIA patients, the increased IL-6 levels were associated with disease activity, in accordance with the high risk of HLH/MAS in this population [173, 174]. However, IL-6 is not elevated in HLH as significantly as in sepsis [99], suggesting that other pro-inflammatory cytokines are also critical for HLH development.
IL-6 is typically present as a monomer, and includes one specific binding site for IL-6 receptor (IL-6R) and two gp130 (signal-transducing protein) binding sites, responsible for its complex and extensive functions [154, 175]. IL-6 downstream pathways can be classified into classical signaling, trans-signaling and trans-presentation, all of which require interactions between IL-6 and receptors through cytokine-binding domain, however, leading to distinct biological effects by different ligand-receptor binding modes [154, 175]. Although almost all stromal cells, macrophages, E-selectin mesangial cells, tumor cells, etc. can produce IL-6, IL-6R expression is more restricted and specifically found in immune cells and response-related cells, such as neutrophils, monocytes and hepatocytes, while gp130 is expressed within almost all cells [176,177,178]. Noteworthily, there are two kinds of IL-6R forms, membrane-bound IL-6R (mIL-6R) and the circulating soluble IL-6R (sIL-6R) [179]. In the classical signal pathway, IL-6 binds to its receptor mIL-6R to form a protein complex, which then associates with the membrane protein gp130 to initiate intracellular signal transduction [154, 180]. Cells that express gp130 but do not express IL-6R cannot result in the downstream signals of mIL-6R that are mainly responsible for regeneration and protection. However, the trans-signaling pathway is mediated via binding of IL-6-sIL-6R complex to gp130 in almost all cells that express this signal transduction protein, leading to the formation of protein hexamer that activates JAK for the initiation and development of a series of biological events that include pro-inflammatory responses [181,182,183,184]. Summarily, JAK’s autophosphorylation of tyrosine residues within its intracellular sequence, serving as recruitment sites for transcription factor STAT, feedback regulator SOCS3, adaptor protein and phosphatase SHP2, can activate multiple downstream signals, such as STAT3, MAPK, PKB/Akt and NF-кB pathways that are broadly involved in pathological conditions [181,182,183,184,185,186,187,188,189,190,191,192]. In the trans-presentation pathway, mIL-6R on dendritic cells binds to IL-6, which is then presented to T cells expressing gp130, playing a critical role for Th17 cells [193].
IL-6 is reported to drive the occurrence and development of diseases in human autoimmunity and inflammation [178]. Although IL-6 has been regarded one of molecules involved in the pathogenesis of HLH, its role and associated mechanisms remain unclear and require to be better studied. Current viewpoints suggest that the elevated IL-6 in patients with HLH may be derived from activated macrophages, which initiate its release synchronously with TNF-α and IL-1β during the early stage of inflammation [194,195,196]. One study involving liver tissue biopsies from five patients with MAS found a significant presence of activated macrophages producing IL-6 [195]. However, another study focusing on cytokine release syndrome (CRS) suggested that monocytes are the primary source of IL-1 and IL-6 [197]. In a transgenic mice model with IL-6 overexpression, prolonged exposure to IL-6 in vivo exacerbated the inflammatory responses to toll-like receptor (TLR) ligands, and these mice exhibited clinical manifestations similar to HLH [194]. Another study confirmed that IL-6 reduced the expression of perforin and granzymes by inhibiting the cytotoxic activity of NK cells, which may be one of the mechanisms underlying MAS in sJIA patients [198]. Based on the fact that pHLH is caused by genetic homozygous defects in genes encoding proteins involved in cellular cytotoxicity, including perforin, findings on IL-6’s inhibition of NK cells further support the hypothesis that pHLH and sHLH may share similar pathogenic mechanisms [198,199,200,201]. Besides, sJIA patients who received IL-6 blockade therapy seemed to have a lower incidence of MAS and significantly less severe clinical presentation compared to the untreated patient group [202, 203]. However, inflammatory response-associated biomarkers could be well corrected via the IL-6 blockade therapy in patients with refractory AOSD [204]. Therefore, though these findings may indicate the amplifying effects of IL-6 on inflammatory responses and its relevance to HLH onset and development, the definitive role of IL-6 in HLH still requires more explorations.
IL-6 has been discovered as one of key cytokines involved in the dysregulation of many diseases, and thus targeting the IL-6 pathway has resulted in a series of novel therapeutics for rheumatic diseases, chimeric antigen receptor T (CAR-T) adoptive infusion and immune checkpoint blockade-related CRS, as well as COVID-19 pneumonia and HLH [1, 19, 205,206,207,208,209,210]. Based on the evidences supporting the use of IL-6 pathway inhibition in the treatment of COVID-19 pneumonia and CRS [19, 153, 206, 211], which are similar to HLH in clinical characteristics and surged cytokine profiles, the use of IL-6 antagonists, such as tocilizumab, has shown some efficacy in the treatment of HLH, despite the current limitations of retrospective study and case report [204, 208, 212]. By binding to IL-6R and inhibiting IL-6-mediated signaling, tocilizumab can serve as an alternative therapy for patients with HLH, especially in adults with sHLH or MAS, or as a salvage treatment for those with familial HLH who showed an inadequate response to etoposide and corticosteroid [204, 208, 212]. Still, there is a lack of clinical trials for assessing the safety and efficacy of IL-6 blockade therapy in HLH patients, except one nonrandomized, interventional, parallel phase II trial that focuses on the tocilizumab or alemtuzumab treatment for HLH adults when combining with etoposide and dexamethasone (NCT02385110).
Targeting TNF-α
TNF-α is a pivotal polymorphic cytokine extensively involved in pro-inflammatory responses, and drugs targeting TNF-α for neutralization have emerged as one of highly effective treatments for diseases of the human immune system [213,214,215,216,217]. During the course of HLH occurrence and progression, a significant increase in serum levels of TNF-α has been observed in patients [218, 219] and animal models [96, 218, 220]. However, TNF-α seemed not to be critical in the pathogenesis of HLH as IL-1β, IL-6 and IL-18, but may reflect the activation degree of inflammatory responses [219]. The specific mechanisms underlying the upregulation of TNF-α in HLH are not fully understood, but it may be driven by preliminarily-elevated TLR ligands such as endotoxin or cytokines [218]. During the process, a large amount of TNF-α and chemokines was secreted by various cell types via binding and induction of mature IL-18 molecules [221,222,223] and targeting IL-18 blockade could effectively decrease production of TNF-α and reverse hemophagocytosis-caused outcomes in the preclinical study [218]. In addition, the combination of TNF-α and IFN-γ blocking antibodies has been shown to provide 100% lethal protection in sHLH mice models induced by poly I:C and LPS attack [224, 225]. Thus, we speculate that the increased level of TNF-α tends to be merely one of downstream or intermediate events in the development of HLH, further promoting the activation of inflammatory response and tissue damage through its involvement in the cytokine cascades.
TNF-α exhibits complex regulatory roles of inflammation in both physiological and pathological conditions, particularly in autoimmune diseases [226,227,228,229,230]. TNF-α is synthesized and released by multiple kinds of cells such as macrophages, mononuclear cells, dendritic cells or lymphocytes, especially myeloid cells and activated T cells in response to diverse inflammatory stimuli [222]. Two forms of TNF-α are discovered within humans: a membrane-bound form (mTNF-α) capable of acting as a ligand or receptor, and a soluble form (sTNF-α) that functions as a ligand [231,232,233]. The 26-kDa mTNF-α can be converted into the 17-kDa sTNF-α by TNF-α-converting Enzyme (TACE) [233], which can exert its effects throughout the whole human body after entering the systematic circulation [234,235,236]. Both mTNF-α and sTNF-α play crucial roles in the inflammatory response. However, mTNF-α primarily functions at the local cellular level, whereas sTNF-α exerts systemic effects.
TNF-α can activate numerous downstream signaling pathways upon binding to its two distinct receptors, TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), which share structural similarity but possess divergent biological functions. TNFR1, also called tumor necrosis factor receptor superfamily member 1A (TNFRSF1A) and CD120a, can be responsible for initiating most of TNF-α’s physical activities [237,238,239,240,241]. Upon binding of trimeric TNF-α to TNFR1, multiple intracellular signal cascades are activated by recruiting several death signaling proteins such as adaptor protein TNFR1-associated death domain (TRADD), Fas-associated death domain (FADD) and TNF receptor-associated factor 1 (TRAF1), leading to the activation of key transcription factors, such as NF-κB, to induce inflammation and cell apoptosis [229, 239, 242,243,244,245]. In HLH, the dysregulation of NF-κB signaling pathway contributes to the persistent activation of immune cells and the further production of pro-inflammatory cytokines as TNF-α, IL-1β, IL-6 and etc., promoting the formation of vicious inflammatory cycle that drives the HLH development [26, 209, 246,247,248]. Besides, with the increasing understanding of TNF signaling in recent years, researchers have found that TNF not only directly drives inflammatory responses by inducing the expression of inflammatory genes but also indirectly drives inflammation by inducing cell death, triggering immune responses and promoting disease development [249,250,251,252]. Cell death is one of the driving factors of inflammatory diseases, such as apoptosis, necrosis, and pyroptosis, which lead to the release of DAMPs and activate subsequent inflammatory cascades [253, 254]. In addition to activating the NF-κB signaling pathway to directly promote inflammatory responses, TNF, upon binding to TNFR1, can also indirectly promote inflammatory signaling by inducing cell death [255,256,257,258,259].
However, unlike TNFR1 that widely exists in various cell types, TNFR2 is specifically expressed in thymic T lymphocytes, endothelial cells, microglia, and oligodendrocytes [239, 260,261,262,263]. Only mTNF-α can tightly bind to TNFR2 and fully initiate the following cellular events by recruiting TRAF1 or TRAF2 adaptors to the receptor due to the lack of death domain [229, 242, 244, 245, 264, 265]. The resulting activated signals involve cIAP1/cIAP2 kinases, as well as the canonical and non-canonical NF-κB, JNK, and AKT pathways [229, 242, 244, 245, 264, 265]. In spite of its activations for cell survival and proliferation by upregulation of PI3K/AKT pathway [266], the interaction between mTNF-α and TNFR2 mainly have stimulated effects on regulatory T cells (Treg) [267,268,269] and myeloid-derived suppressive cells (MDSC) [270,271,272] for immune inhibition because of TNFR2’s preferred expressions on their surfaces [265]. During inflammation responses, the excessive expression of mTNF-α is supposed to bind to TNFR2 for activating Treg cell to control the amplification of TNF-α's pro-inflammatory effects [267,268,269, 273, 274]]. However, in HLH, highly activated CD8+ T lymphocytes disrupts IL-2 homeostasis, resulting in a shift away from Treg cell maintenance and toward promotion of a feed-toward inflammation preference [275]. Thus, the dysfunctional Tregs cannot response to the strong mTNF-α-TNFR2 interaction to mitigate the inflammation progression in patients with HLH.
Despite the potent pro-inflammatory effects of TNF-α through the activation of innate and adaptive immunity, it also exerts functional inhibitions on NK cell-like Treg cells [276, 277]. It is speculated that the mechanism involves TNF-α increasing the adhesion of NK cells to endothelial cells or exerting direct cytotoxicity on NK cells [277]. During the occurrence and progression of HLH, the activity of NK cells is suppressed, and impaired or deficient NK cell cytotoxic function can serve as one of the diagnostic criteria for HLH [278,279,280,281]. Therefore, it is hypothesized that high levels of TNF-α may be one of the causes that contribute to the secondary functional defects of NK cells in HLH, leading to sustained activation of inflammatory signals and hindering the disease control.
In summary, as one of the inflammatory effector cytokines, TNF-α can firstly activate a series of immune cells and endothelial cells through binding to TNFR1, leading to the initiation of inflammatory signals and release of numerous inflammatory cytokines. Additionally, TNF-α also serves a function of inflammatory regulation via TNFR2, mainly by activating immunosuppressive cells expressing TNFR2, such as MDSCs and Tregs. In patients with HLH, the crucial regulatory function of Treg cells in controlling inflammation is impaired, thus unable to respond to TNF-α-TNFR2 interaction-mediated inflammatory regulatory signals. This may be one of the mechanisms of amplifying the inflammatory cycle in HLH. Although TNF-α elevation may not be the core mechanism leading to HLH, targeting the TNF-α signaling pathway to alleviate the cytokine storm and reduce tissue damage is worth further researches. TNF-α-blocking antibodies have been used to treat various rheumatic or autoimmune diseases [213,214,215,216,217], and blocking TNF-α seems to have a certain therapeutic effect in MAS [282,283,284,285,286,287]. However, conflicting results have been reported in multiple studies [96, 282,283,284,285,286, 288, 289], which may be associated with the discrepancy of TNF-α levels. For critically-ill patients with a sharp increase in TNF-α, the treatment with TNF-α-neutralizing antibodies may be an option [283]. Furthermore, there were also reports suggesting that TNF-α therapy could indirectly induce HLH or worsen inflammation [290, 291]. Therefore, the application value of targeting TNF-α-related signaling pathways remains to be further studied for treating HLH and requires careful consideration. The intervention timing and inflammation degree may be critical factors, especially for HLH patients secondary to autoimmune diseases.
Targeting IL-1β
The IL-1 family includes several cytokines and receptors, and most of them share similar functions in inflammation and immune regulation as TLR families [292, 293]. IL-1α and IL-1β are two different molecular forms of the IL-1 ligands, both of which belong to immune-stimulated cytokines that mainly initiate innate immune-related inflammatory responses but also have effects on adaptive immune, especially T and B cell activation [293]. A large amount of IL-1 can be released when these cells are activated by foreign antigens or mitogens, which can be regarded as one of innate defense mechanisms [293, 294]. IL-1α and IL-1β signals will exert similar biological functions upon binding to their receptors, IL-1RI and IL-1RII [295, 296].
IL-1β is one of the key pro-inflammatory cytokines involved in the pathogenesis of HLH [209]. Under normal physiological conditions, IL-1β is intracellularly stored as the precursor form known as pro-IL-1β with low biological activity [297, 298]. Upon activation by PAMPs or DAMPs, transcription of pro-IL-1β is obviously upregulated [299,300,301,302,303,304]. In addition, inflammatory cytokines such as IL-18, TNF-α, and IL-1β itself can also promote the production of pro-IL-1β [305,306,307,308,309]. IL-1β acts on the cell surface IL-1RI through autocrine, paracrine, or systemic secretion, mediating inflammation by promoting the release of other pro-inflammatory cytokines such as IL-6 and TNF-α, which plays a critical role in bridging innate and adaptive immunity via interaction with Th1 and Th17 cells [310,311,312]. Therefore, several levels can be regulated to control the inflammatory burst associated with pathologically-elevated IL-1β in the treatment of HLH.
IL-1β is highly associated with sJIA, which is one of the main causes of sHLH [313,314,315,316]. However, the exact role of IL-1β in the development of HLH remains unclear. HLH is characterized by elevated levels of various cytokines, and its clinical manifestations differ from diseases primarily mediated by the increased IL-1β level, such as sJIA, cryopyrin-associated periodic fever syndrome, and familial Mediterranean fever [219, 317]. Generally, these diseases demonstrated great responses to IL-1 blockade therapy, with rapidly reduced IL-1β levels observed after administration [219, 317,318,319]. However, the efficacy of targeting IL-1β in the treatment of HLH remains uncertain.
The competitive inhibitor for IL-1 ligands, Anakinra, is a recombinant soluble receptor antagonist for both IL-1β and IL-1α and has been widely used in the treatment of sJIA. However, previous studies showed its varied efficacy in MAS, and there were reports of Anakinra that might induce the occurrence of MAS [196, 205, 315, 320,321,322,323,324,325,326]. In a re-analysis of data from a phase III multicenter randomized clinical trial evaluating the use of anakinra in severe sepsis, it was found that anakinra reduced the mortality rate by 30% in patients with clinical signs of HLH [327]. Treatments with anakinra significantly alleviated patients’ symptoms and decreased hemophagocytosis scores in HLH patients secondary to severe COVID-19 pneumonia, suggesting its potentials in lowering death risk for cytokine storm-related diseases [328]. Canakinumab is a high-affinity fully human monoclonal antibody against IL-1β that specifically neutralizes IL-1β [329]. Although MAS has been considered an adverse event in clinical trials of canakinumab for the treatment of sJIA, the incidence rate of MAS in these trials does not seem to be higher than those reported in real-world data for sJIA patients [330], suggesting that canakinumab does not affect the occurrence risk of MAS [331, 332]. The efficacy of targeting IL-1β in HLH may be dose-dependent. In most studies reporting successful responses to anakinra in treating MAS, patients were administered at high doses up to 10 mg/kg [205], while lower doses around 1–2 mg/kg might be associated with higher risk of drug-induced MAS [325, 326], although clear conclusions still require further evaluation. The current dosages (< 4 mg/kg) used in canakinumab clinical trials may not be sufficient to neutralize the increased IL-1β levels in MAS [205], thus further exploration of its higher dosages is needed to assess its inflammation control efficacy in MAS.
The aforementioned studies indicate that IL-1β plays a certain role in the occurrence and development of HLH, especially MAS. The therapeutic effect of blocking IL-1β may not only be attributed to the direct reductions in the production and release of IL-1β, but also to the control of the persistently-elevated pro-inflammatory cytokines. However, targeted blockade of IL-1β with canakinumab in sJIA patients were found ineffective both in reducing MAS risk and in treating MAS, also suggesting the limited role of IL-1β in the pathogenesis of MAS. Additionally, the non-selective IL-1α/β inhibitor, anakinra, appears to have better prospects in controlling inflammation in patients suffering MAS. Moreover, the IL-1α/β competitive inhibitor, anakinra, has shown promising results as an adjuvant therapy in twelve pediatric MAS patients [324]. Therefore, further exploration of blocking IL-1α in HLH patients can be worth in the future. Overall, targeting a single blockade of IL-1 signaling may not be the key point for controlling HLH, as other cytokines induced by its resulted cascade responses are supposed to block at the same time.
Targeting IL-18
IL-18 is a pro-inflammatory cytokine that belongs to the IL-1 family, normally existing as an inactive 24 kDa precursor form [333]. Activation of NF-κB following TLR stimulation induces the transcription of Pro-IL-18, which is further cleaved by caspase-1 into one mature and biologically active 18 kDa molecule, then releasing into the extracellular environment [334, 335]. IL-18 is predominantly present in monocytes/macrophages, antigen-presenting cells and epithelial cells in healthy humans and mice [336]. Similar to IL-1, IL-18 induces the production of inflammatory mediators by activating the NF-κB signal [336]. After binding to IL-18 receptor alpha (IL-18Rα) and its following recruitment of IL-18 receptor beta (IL-18Rβ), mature IL-18 initiates TLR/ IL-1R-like pro-inflammatory signaling via the MyD-IRAK1/4-NF-κB axis and p38 MAPK [336,337,338,339].
IL-18 is an important cytokine involved in immune mechanisms of activating macrophages and Th1 cells, which are critical to HLH pathogenesis [340]. A synergistic action of IL-18 and IL-12 stimulates Th1-mediated immune reactions, inducing expressions of chemokines and cell adhesion molecules [341] and promoting the secretion of inflammatory cytokines such as IL-1, IFN-γ and TNF-α [338, 342,343,344,345,346]. Significantly elevated levels of IL-18 can be observed in both primary and secondary HLH patients [347,348,349]. Serum levels of IL-18 were positively correlated with disease activity in HLH [347, 350,351,352,353,354,355]. Specifically, IL-18 was previously referred to as the IFN-γ-inducing factor [356], while IFN-γ rapidly drives the immune activation that promotes HLH occurrence [357]. Moreover, the sustained stimulation of macrophages by IL-18 and their continued activation further promote the release of various inflammatory cytokines, such as IL-1, IL-6, IL-18, and TNF-α, leading to tissue impairment and hemophagocytosis by macrophages [96, 195].
There are other diseases associated with HLH in which IL-18 levels are often significantly elevated, though with distinct underlying mechanisms, such as MAS, X-linked inhibitor of apoptosis protein (XIAP) deficiency and the NLRC4 mutation [205, 358, 359]. MAS is one of the most common secondary form of HLH and usually originates from rheumatic diseases or systemic autoinflammatory diseases (SAID) [360], including sJIA, AOSD, SLE, Kawasaki disease, systemic vasculitis, etc [219, 281, 334]. Actually, MAS can be a potentially life-threatening complication of rheumatic diseases, characterized by excessive activation and expansion of T lymphocytes/macrophages exhibiting hemophagocytic activity [281]. Compared to patients with EBV-HLH, patients with MAS exhibited more elevated levels of serum IL-18 [361], which might partly contribute to the occurrence of liver damage among them by inducing Fas ligands on NK cells [362]. Most patients with XIAP deficiency would experience recurrent HLH and exhibit a high level of serum IL-18 [353]. XIAP deficiency is a rare primary immunodeficiency caused by BIRC4 mutations, also known as XLP-2 [359, 363]. Clinical features of XIAP deficiency include HLH and inflammatory bowel diseases due to defective nucleotide binding oligomerization domain containing 2 (NOD2) responses [79, 359], but the specific mechanism by which mutated XIAP leads to the presentation of HLH remains incompletely understood. Elevated levels of IL-18 may offer crucial insights into the pathogenesis of this disease. On the other hand, the NLRC4 inflammatory is part of the human innate immune system, and its activation can lead to the cleavage of the pro-inflammatory cytokines IL-1β and IL-18, which also promotes inflammation [364]. The gain-of-function mutations in NLRC4 lead to HLH and gastrointestinal pathology [26, 43, 365], resulting in spontaneous activation of NLRC4 inflammasomes that increased IL-18 levels [360, 366]. It was reported that one patient with refractory NLRC4-MAS exhibited a significant response after IL-18 blockade treatment (IL-18 binding protein, IL-18BP) [367].
IL-18BP is a constitutive protein that can bind to IL-18 with high affinity, forming a complex that prevents IL-18 from interacting with its cell surface receptors [339]. Therefore, IL-18BP acts as a natural inhibitor of IL-18, controlling excessive IL-18-mediated inflammatory responses [368,369,370]. Imbalance between IL-18 and IL-18BP may lead to the activation of T lymphocytes and macrophages in HLH [347]. IL-18BP can be induced by IFN-γ, considering that IL-18 signaling has a negative feedback loop [336, 350, 371]. In a pHLH mouse model, the IL-18BP treatment reduced hemophagocytic activity and reversed liver and spleen damage [218]. Meanwhile, it also decreased the production of IFN-γ and TNF-α by CD8+ T cells and NK cells, as well as the expression of Fas ligand on the surface of NK cells. However, this therapeutic did not improve the survival outcome [218]. Some clinical studies reported that recombinant human IL-18BP successfully treated patients with severe inflammatory responses carrying the NLRC4 mutation [367]. Currently, some clinical trials (NCT03512314) are underway for IL-18BP (tadekinig alfa) in patients with NLRC4 or XIAP mutations. Overall, IL18-BP holds great potentials of modulating the inflammatory response triggered by IL-18, thereby exerting a positive impact on HLH. However, further clinical research and assessment are required to determine the efficacy and safety of IL18-BP as a treatment for HLH.
Targeting CD52
CD52 is a glycoprotein consisting of 12 amino acids anchored to glycosylphosphatidylinositol (GPI) [372, 373]. It is a widely distributed antigen found on lymphocytes, monocytes, eosinophils and dendritic cells differentiated from monocytes in the hematopoietic system, with a high density on lymphocytes [374]. Some studies have suggested that CD52 is an important immunomodulatory factor in T cell activation [375]. However, the specific pathways and mechanisms require further elucidation.
Alemtuzumab, also known as Campath-1H (trade name in Europe: MabCampath), is a humanized monoclonal antibody targeting the cell surface CD52 antigen [374]. It has been approved for chronic lymphocytic leukemia, multiple sclerosis (MS), and is also utilized in some autoimmune diseases such as rheumatoid arthritis, solid organ transplantation and GVHD following bone marrow transplantation [376,377,378,379,380,381,382]. Alemtuzumab eliminates T and B lymphocytes through mechanisms such as inducing cell apoptosis, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) [383,384,385]. One study reported the usage of T-cell depletion agents like alemtuzumab as salvage therapy for refractory HLH [386]. The high levels of CD52 expression on T cells and tissue cells make alemtuzumab a rational alternative for disrupting the uncontrolled immune responses like HLH.
One study reported that among 22 patients with refractory HLH treated with alemtuzumab, 14 of them experienced an overall partial response with 77% of them surviving to undergo allo-HSCT [386]. In a case of recurrent atypical HLH refractory to multiple immunosuppressive agents, alemtuzumab induction resulted in remission, enabling successful allo-HSCT [387]. As for HLH/MAS, in a case of SLE-induced HLH, despite refusal of high-intensity immunosuppressive therapy, the patient's condition gradually improved after alemtuzumab treatment [388]. Additionally, RIC regimens typically including alemtuzumab improved survival outcome in HLH patients after allo-HSCT [66]. However, caution is warranted regarding viral reactivation when using alemtuzumab.
For patients with refractory HLH, alemtuzumab may be an effective salvage therapy. However, some previous studies have also indicated that alemtuzumab induced HLH in patients with hematologic malignancies [389]. Therefore, the use of alemtuzumab in HLH should be approached with extreme caution.
Targeting CD20
CD20 remains to be one of most important surface markers expressed on B lymphocytes since the late pre-B cell stage, and is lost in terminally differentiated plasma cells and plasmablasts [390]. CD20 is a 33–37 kDa non-glycosylated protein classified into the membrane-spanning 4-domains subfamily A (MS4A), encoded by MS4A1 [391, 392]. The biological function and physiological ligands of CD20 on B cells are still not fully understood. Some studies suggested that CD20 deficiency lead to the decreased circulating memory B cells, less immunoglobulin isotype switching and lower IgG levels [393]. CD20 is associated with several protein tyrosine kinases, including Lyn, Fyn, Lck, and p75/85 kinases, which can cause activation of phospholipase-C-gamma (PLC-γ) and the subsequent MAPK (JNK, ERK, and p38MAPK) signaling pathways [394]. PLC-γ can also hydrolyze PIP3, generating inositol trisphosphate and diacylglycerol, which are signaling molecules involved in pathways highly similar to B cell receptor (BCR) signaling [395].
Patients with perforin-dependent cytotoxicity defects or genetic predisposition are susceptible to EBV-HLH [396, 397]. Some studies suggested that EBV encoding protein mimicked key signaling pathways within B cells [398,399,400,401]. For instance, LMP1 could simulate active CD40 receptor, and latent membrane protein 2A (LMP2A) could simulate or replace BCR signaling [398,399,400,401]. Furthermore, CD20 indirectly regulated calcium release dependent on the BCR pathway, and CD20+ B cells that lack BCR were unable to initiate calcium-releasing signals [402]. Some studies also demonstrated that CD20 directly functioned as an ion channel, and overexpression and knockout of CD20 might increase or decrease calcium current in B cells, respectively [403].
EBV, also known as human herpesvirus 4, is a double-stranded DNA virus [404], mainly targeting B lymphocytes both in vitro and in vivo, which serves as the location site for virus preservation in healthy carriers [405]. Preferentially, EBV infects B lymphocytes by two strategies: (1) binding to the B-cell surface CD21 through viral envelope glycoprotein gp350; (2) binding to human leukocyte antigen through glycoprotein gp42 [406,407,408]. EBV infection drives the transformation of B lymphocytes [409]. Within these host B cells, EBV may primarily exist as the free form and replicate via host DNA polymerase, but its nucleotide sequences can be integrated into the host genome by the non-random pattern [410]. Within healthy individuals, transformed B lymphocytes will be rapidly eliminated by NK and cytotoxic CD8+T cells, while target cell killing deficiencies in patients with familial or sHLH may trigger the dysregulation of systematic inflammatory responses that contribute to HLH occurrence [279, 411, 412].
Anti-CD20 monoclonal antibodies (mAbs) are targeted drugs against B cells by blocking CD20 molecules [413]. Based on different characteristics, anti-CD20 antibodies can be classified into type I (such as rituximab) and type II (such as obinutuzumab), depending on their ability to induce redistribution of CD20 into lipid rafts on the cell membrane [414]. Type I CD20 mAbs induce the recombination of CD20 molecules into lipid rafts and then effectively activate the classical pathway of complement system. Type II CD20 mAbs exhibit poorer abilities in complement activation, but perform better to induce cell death after directly binding to CD20 without cross-linking through secondary antibodies [393, 414,415,416]. Anti-CD20 mAbs exert their effects through CDC, ADCC and direct cytotoxicity, leading to the destruction of targeted B cells [417, 418]. Moreover, it can also interfere with BCR signaling and downregulate BCR expression [419,420,421]. Rituximab is a chimeric mouse/human mAb that can deplete CD20+ cells within 48 h after administration, decreasing the incidence rate of EBV reactivation [422]. Rituximab is effective in treating various EBV-mediated diseases, such as EBV-induced post-transplant lymphoproliferative disorder (EBV-PTLD) [423,424,425]. EBV usually demonstrates a poor response to anti-viral drugs, and thus its presence within B lymphocytes allows for rapid depletion through the use of targeted mAbs [426, 427].
Rituximab-based chemotherapeutic regimens have been used for EBV-HLH. Chellapandian et al. retrospectively reported a clinical cohort involving 42 patients with EBV-HLH who received a regimen including rituximab with great tolerability, which effectively improved the physical status for 43% of patients with significant decreases in EBV load and serum ferritin levels [428]. There were also one report of two cases of central nervous system involvement in patients with EBV-HLH on which alleviated symptoms were rapidly observed with the use of rituximab as a monotherapy [429, 430]. Monocytes/macrophages play a crucial role in the depletion of B cells, and the activation of macrophages is commonly observed in HLH patients, which may facilitate the ADCC effects of anti-CD20 mAbs [431]. However, in some cases, EBV can also infect other kinds of cells, such as T cells and NK cells [432], which may not be eliminated by giving Rituximab [433, 434]. In a study analyzing EBV-DNA level in lymphocyte subpopulations of 15 HLH patients, it was found that EBV primarily infected T and NK cells in 5 patients, and only infected B cells in the remaining 10 patients [333]. After receiving a regimen including rituximab, the patients who had infected T and NK cells had no obvious changes in EBV viral load, while the other 10 patients showed the significantly decreased EBV levels [333]. In HLH cases whose B cell-depletion have been confirmed, the persistently high EBV-DNA level suggested the EBV infection into T/NK cells [333, 396]. Therefore, combining with etoposide and dexamethasone may help to eliminate infected T and NK cells for promotion of virus clearance [1, 398, 435].
The primary therapeutic principles for EBV-HLH include suppressing excessive inflammation, eliminating EBV and reversing impaired immune system function [333, 436]. Dampening EBV activation and cutting its virus burden were proved to have potentials of controlling clinical symptoms and improving survival outcome [333, 436]. During the active phase, the use of rituximab was able to limit immune responses by getting rid of EBV-infected B cells [428, 437]. However, patients previously treated with rituximab (often in combination with other medications) usually hace varying administration schedules and dosages, potentially leading to reporting biases and confounding factors. Overall, treatment regimens including rituximab have demonstrated a promising outlook in reducing EBV load and alleviating hyperinflammation, which, nevertheless, should be further validated. In patients with EBV-HLH, monitoring of the response to rituximab can be performed using EBV blood polymerase chain reaction assays, which at the same time help reflect the increases in viral load and recovery of B cells after rituximab therapy [396, 428]. However, B cell-targeted therapies lead to a strong immune suppression, thus necessitating precautions such as effective isolation and antifungal prophylaxis. Besides, it is necessary to regularly monitor potential pathogens (CMV, adenovirus or aspergillus) to prevent infection or reactivation.
Anti-CD20 mAbs are expected to be primarily utilized for EBV-HLH. Since EBV tends to infect B cells, targeting EBV-infected B cells using anti-CD20 mAbs may effectively dampen the amplified inflammatory response, but infection monitoring is necessary due to the substantial impact of B cell clearance on the immune function of the body. Currently, the clinical evidence for the use of anti-CD20 mAbs in HLH is limited to case reports or small-sample retrospective studies. The definitive role of CD20 in HLH remains unclear, and further explorations through standardized clinical trials are required.
Targeting PD-1
PD-1, also known as CD279, is a prototypical immune inhibitory checkpoint predominantly found on the surface of T cells [438]. It regulates T cell effector function during various physiological responses, including acute and chronic infections, cancer, autoimmune diseases and immune homeostasis [439]. The cytoplasmic tail of PD-1 contains two tyrosine-based motifs: an Immunoreceptor Tyrosine-based Inhibitory Motif (ITIM) and an Immunoreceptor Tyrosine-based Switch Motif (ITSM) [440, 441]. The PD-1 has two ligands, PD-L1 (CD274) and PD-L2 (CD273) [442]. PD-L1 is broadly expressed across various cell types, found in hematopoietic cells (including T cells, B cells, dendritic cells (DCs), and macrophages) as well as non-hematopoietic cells (including vascular and stromal endothelial cells) [439]. In contrast, PD-L2 expression is more restricted, primarily expressed by DCs, macrophages, and subsets of B cells [439]. Upon binding with its ligands, PD-1 is phosphorylated at these tyrosine residues, leading to the recruitment of protein tyrosine phosphatases (PTPs) such as SHP2 [443]. These PTPs can dephosphorylate kinases and counteract the positive signals generated through T cell receptor (TCR) and CD28, affecting downstream signaling pathways including those involving PI3K-AKT, RAS-ERK, and PLC-γ [439, 444]. The aforementioned interaction between PD-1 and its ligands can suppress T cell proliferation, activation, cytokine production and cytotoxic T lymphocyte killing function, thereby protecting the organism from autoimmune attacks [445]. Many malignant tumors express PD-L1, and thus high PD-L1 expression is associated with poor prognosis in diseases such as malignant melanoma, colon cancer, pancreatic cancer, hepatocellular carcinoma, and ovarian cancer [446]. Therefore, PD-1 inhibitors have been approved for the treatment of various malignant tumors. PD-1 blockade can significantly prolong the survival of patients with such diseases and provide long-term sustained remission. Additionally, some studies have suggested that inhibitors targeting the PD-1 pathway can rescue T cells from exhaustion, reactivate dysfunctional CD8+ T cell populations and restore immune responses [447].
During certain chronic infections, persistent antigen exposure results in sustained PD-1 expression, which limits the clearance of immune-mediated pathogens or tumor cells [439]. It was reported that PD-1 inhibitors had been successfully used to treat the chronic viral infection [448]. In all kinds of HLH cases, infections are a common trigger. Several reports have demonstrated successful treatment of EBV-HLH and chronic active EBV infection (CAEBV) through PD-1 blockade [449,450,451,452,453,454]. A study involving seven r/r EBV-HLH patients treated with nivolumab as a monotherapy showed responses in six patients (85.7%), with five patients (71.4%) achieving clinical CR and a gradual reduction in plasma EBV-DNA copy numbers [452]. Single-cell sequencing revealed positive enrichment of multiple T cell activation pathways and degranulation pathways in CD8+ T cells after nivolumab treatment, suggesting that nivolumab may restore the cytotoxicity function of CD8+ T cells [452].
One study involved 12 EBV-HLH patients in the intensive care unit with sintilimab and ruxolitinib therapy, with six patients (50%) achieving CR within 1 month [455]. With a median follow-up time of 5 (4.4 to 14.7) months, six of them died, resulting in a mortality rate of 50% [455]. For EBV-HLH patients with post-transplant relapse, PD-1 blockade also showed promising effects. An EBV-HLH patient who relapsed after chemotherapy and allo-HSCT might benefit from the addition of sintilimab as salavage therapy, with normalization of fever, cell count, liver enzyme elevation, serum ferritin and sCD25 levels and negative EBV-DNA loads [456]. Therefore, PD-1 blockade therapy may be an option for r/r and critically ill EBV-HLH patients, though further validation is required.
However, stimulating the immune system is a double-edged sword, as sustained immune activation may also trigger HLH or exacerbate HLH symptoms [457]. Some reports have indicated cases of immune checkpoint inhibitor-related HLH in patients with various solid tumors [458,459,460,461]. There have even been studies reporting HLH induction in two CAEBV patients following the treatment with sintilimab [462]. Additionally, one case report described the worsening of symptoms and CRS-related pulmonary injury in a 3-year-old girl with r/r EBV-HLH when treated with nivolumab during the acute phase of HLH disease [463]. Therefore, considering safety concerns, cautions should be exercised when using the PD-1 blockade strategy in HLH during the peak of inflammation.
Conclusions
In summary, HLH is a life-threatening hyperinflammatory syndrome characterized by excessive immune activation. HLH can be hereditary or sporadic, triggered by various events that disrupt immune homeostasis. HLH is typically treated with immunosuppressive therapy to induce remission. For patients with pHLH, allo-HSCT is considered once the high-inflammatory state is controlled. For patients with r/r HLH, cytokine-targeted therapy and immunotherapy can be a treatment option, including the addition of the L-DEP regimen, JAK1/2 inhibitors, anti-CD52 antibodies, anti-CD20 antibodies, and PD-1 blocking agents. The IL-6 antagonists, IL-1 receptor antagonists, TNF-α blocking antibodies and L-18BP may be considered for MAS patients. Besides, anti-the IFN-γ antibody, emapalumab, has been proved to have efficacy for pHLH. All the above-mentioned targeted therapeutics can be combined with the conventional treatments, which is worth looking forward to in the future studies. Specifically, HLH patients planned for allo-HSCT may consider receiving a RIC regimen with anti-CD52 antibodies that should be personalized based on the doctor's expertise and the patient's condition. Overall, further clinical cohort studies are required to explore the efficacy of single-agent and combination therapies with different targeted drugs in HLH.
Availability of data and materials
No datasets were generated or analysed during the current study.
Abbreviations
- HLH:
-
Hemophagocytic lymphohistiocytosis
- HSCT:
-
Hematopoietic stem cell transplantation
- MAS:
-
Macrophage activation syndrome
- sJIA:
-
Systemic juvenile idiopathic arthritis
- SLE:
-
Systemic lupus erythematosus
- AOSD:
-
Adult Still's disease
- IL:
-
Interleukin
- IFN-γ:
-
Interferon-γ
- EBV:
-
Epstein-barr virus
- XLP:
-
X-linked lymphoproliferative disease
- XIAP:
-
X-linked inhibitor of apoptosis protein
- NOD2:
-
Nucleotide binding oligomerization domain containing 2
- CMV:
-
Cytomegalovirus
- CTLs:
-
Cytotoxic T cells
- ORR:
-
Overall response rate
- allo-HSCT:
-
Allogenic HSCT
- RIC:
-
Reduced-intensity conditioning
- GVHD:
-
Graft-versus-host disease
- HSCs:
-
Hematopoietic stem cells
- TCR:
-
T cell receptor
- ATCT:
-
Adoptive T cell therapy
- JAK-STAT:
-
Janus kinase-signal transducer and activator of transcription
- PD-1:
-
Programmed cell death protein 1
- IFNGR:
-
IFN-γ receptor
- GAS:
-
Gamma-activated sequence
- LCMV:
-
Lymphocytic choriomeningitis virus
- CNS:
-
Central nervous system
- TYK2:
-
Tyrosine kinase 2
- GM-CSF:
-
Granulocyte-macrophage colony-stimulating factor
- FDA:
-
Food and Drug Administration
- CR:
-
Complete response
- PR:
-
Partial response
- PAMPs:
-
Pathogen-associated molecular patterns
- DAMPs:
-
Damage associated molecular patterns
- PDGF:
-
Platelet-derived growth factor
- LPS:
-
Lipopolysaccharide
- PMA:
-
Phorbol myristate acetate
- mIL-6R:
-
Membrane-bound IL-6R
- sIL6R:
-
Soluble IL-6R
- TLR:
-
Toll-like receptor
- CAR-T:
-
Chimeric antigen receptor T cell
- CRS:
-
Cytokine release syndrome
- TACE:
-
TNF-α-converting Enzyme
- TNFR1:
-
TNF receptor
- TNFR2:
-
TNF receptor 2
- TNFRSF1A:
-
Tumor necrosis factor receptor superfamily member 1A
- TRADD:
-
Adaptor protein TNFR1-associated death domain
- FADD:
-
Fas-associated death domain
- TRAF1:
-
TNF receptor-associated factor 1
- Treg:
-
Regulatory T cell
- MDSC:
-
Myeloid-derived suppressive cell
- IL-18Rβ:
-
IL-18 receptor beta
- SAID:
-
Systemic autoinflammatory diseases
- IL-18BP:
-
IL-18 binding protein
- GPI:
-
Glycosylphosphatidylinositol
- MS:
-
Multiple sclerosis
- ADCC:
-
Antibody-dependent cellmediated cytotoxicity
- CDC:
-
Complement-dependent cytotoxicity
- PLC-γ:
-
Phospholipase-C-gamma
- BCR:
-
B cell Receptor
- LMP2A:
-
Latent membrane protein 2A
- mAbs:
-
Monoclonal antibodies
- EBV-PTLD:
-
EBV-induced post-transplant lymphoproliferative
- ITIM:
-
Immunoreceptor Tyrosine-based Inhibitory Motif
- ITSM:
-
Immunoreceptor Tyrosine-based Switch Motif
- DCs:
-
Dendritic cells
- PTPs:
-
Protein tyrosine phosphatases
- CAEBV:
-
Chronic active EBV infection
- PBMCs:
-
Peripheral blood mononuclear cells
References
La Rosée P, et al. Recommendations for the management of hemophagocytic lymphohistiocytosis in adults. Blood. 2019;133:2465–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2018894618.
Scott RB, Robb-Smith A. Histiocytic medullary reticulosis. The Lancet. 1939;234:194–8.
Farquhar JW, Claireaux AE. Familial haemophagocytic reticulosis. Arch Dis Child. 1952;27:519–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/adc.27.136.519.
Chinnici A, et al. Approaching hemophagocytic lymphohistiocytosis. Front Immunol. 2023;14:1210041. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1210041.
Stepp SE, et al. Perforin gene defects in familial hemophagocytic lymphohistiocytosis. Science. 1999;286:1957–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.286.5446.1957.
Sayos J, et al. The X-linked lymphoproliferative-disease gene product SAP regulates signals induced through the co-receptor SLAM. Nature. 1998;395:462–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/26683.
Nichols KE, et al. Inactivating mutations in an SH2 domain-encoding gene in X-linked lymphoproliferative syndrome. Proc Natl Acad Sci U S A. 1998;95:13765–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.95.23.13765.
Coffey AJ, et al. Host response to EBV infection in X-linked lymphoproliferative disease results from mutations in an SH2-domain encoding gene. Nat Genet. 1998;20:129–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/2424.
Barbosa MD, et al. Identification of the homologous beige and Chediak-Higashi syndrome genes. Nature. 1996;382:262–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/382262a0.
Borgia RE, Gerstein M, Levy DM, Silverman ED, Hiraki LT. Features, treatment, and outcomes of macrophage activation syndrome in childhood-onset systemic lupus erythematosus. Arthritis Rheumatol. 2018;70:616–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.40417.
García-Pavón S, Yamazaki-Nakashimada MA, Báez M, Borjas-Aguilar KL, Murata C. Kawasaki disease complicated with macrophage activation syndrome: a systematic review. J Pediatr Hematol Oncol. 2017;39:445–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/mph.0000000000000872.
Arlet JB, et al. Reactive haemophagocytic syndrome in adult-onset Still’s disease: a report of six patients and a review of the literature. Ann Rheum Dis. 2006;65:1596–601. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/ard.2005.046904.
Grom AA, Passo M. Macrophage activation syndrome in systemic juvenile rheumatoid arthritis. J Pediatr. 1996;129:630–2. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0022-3476(96)70140-3.
Knauft J, et al. Lymphoma-associated hemophagocytic lymphohistiocytosis (LA-HLH): a scoping review unveils clinical and diagnostic patterns of a lymphoma subgroup with poor prognosis. Leukemia. 2024;38:235–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41375-024-02135-8.
West J, et al. 1-year survival in haemophagocytic lymphohistiocytosis: a nationwide cohort study from England 2003–2018. J Hematol Oncol. 2023;16:56. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-023-01434-4.
Trottestam H, et al. Chemoimmunotherapy for hemophagocytic lymphohistiocytosis: long-term results of the HLH-94 treatment protocol. Blood. 2011;118:4577–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2011-06-356261.
Henter JI, et al. Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood. 2002;100:2367–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2002-01-0172.
Locatelli F, et al. Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 2020;382:1811–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1911326.
Liu D, Zhao J. Cytokine release syndrome: grading, modeling, and new therapy. J Hematol Oncol. 2018;11:121. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-018-0653-x.
Hao Z, Li R, Meng L, Han Z, Hong Z. Macrophage, the potential key mediator in CAR-T related CRS. Exp Hematol Oncol. 2020;9:15. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40164-020-00171-5.
Nguyen TTT, Kim YT, Jeong G, Jin M. Immunopathology of and potential therapeutics for secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome: a translational perspective. Exp Mol Med. 2024;56:559–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-024-01182-6.
de Saint Basile G, Ménasché G, Fischer A. Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules. Nat Rev Immunol. 2010;10:568–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri2803.
Dieckmann NM, Frazer GL, Asano Y, Stinchcombe JC, Griffiths GM. The cytotoxic T lymphocyte immune synapse at a glance. J Cell Sci. 2016;129:2881–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.186205.
Stinchcombe JC, Bossi G, Booth S, Griffiths GM. The immunological synapse of CTL contains a secretory domain and membrane bridges. Immunity. 2001;15:751–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1074-7613(01)00234-5.
Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol. 2006;6:940–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri1983.
Canna SW, Marsh RA. Pediatric hemophagocytic lymphohistiocytosis. Blood. 2020;135:1332–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2019000936.
Jenkins MR, et al. Failed CTL/NK cell killing and cytokine hypersecretion are directly linked through prolonged synapse time. J Exp Med. 2015;212:307–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20140964.
Jessen B, et al. Graded defects in cytotoxicity determine severity of hemophagocytic lymphohistiocytosis in humans and mice. Front Immunol. 2013;4:448. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2013.00448.
Zhang HQ, et al. Cytokine storm and targeted therapy in hemophagocytic lymphohistiocytosis. Immunol Res. 2022;70:566–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12026-022-09285-w.
Feldmann J, et al. Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3). Cell. 2003;115:461–73.
zur Stadt U, et al. Linkage of familial hemophagocytic lymphohistiocytosis (FHL) type-4 to chromosome 6q24 and identification of mutations in syntaxin 11. Hum Mol Genet. 2005;14:827–34.
Zur Stadt U, et al. Familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) is caused by mutations in Munc18–2 and impaired binding to syntaxin 11. Am J Hum Genet. 2009;85:482–92.
Arico M, et al. Hemophagocytic lymphohistiocytosis due to germline mutations in SH2D1A, the X-linked lymphoproliferative disease gene. Blood. 2001;97:1131–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.v97.4.1131.
Rigaud S, et al. XIAP deficiency in humans causes an X-linked lymphoproliferative syndrome. Nature. 2006;444:110–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature05257.
Goldberg J, Nezelof C. Lymphohistiocytosis: a multi-factorial syndrome of macrophagic activation clinico-pathological study of 38 cases. Hematol Oncol. 1986;4:275–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hon.2900040405.
Enders A, et al. Lethal hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type II. Blood. 2006;108:81–7.
Grunebaum E, Zhang J, Dadi H, Roifman CM. Haemophagocytic lymphohistiocytosis in X-linked severe combined immunodeficiency. Br J Haematol. 2000;108:834–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1365-2141.2000.01923.x.
Pasic S, Micic D, Kuzmanovic M. Epstein-Barr virus-associated haemophagocytic lymphohistiocytosis in Wiskott-Aldrich syndrome. Acta Paediatr. 2003;92:859–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/08035250310003631.
Hisano M, et al. Bacteria-associated haemophagocytic syndrome and septic pulmonary embolism caused by Burkholderia cepacia complex in a woman with chronic granulomatous disease. J Med Microbiol. 2007;56:702–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1099/jmm.0.47071-0.
Schultz KA, et al. Familial hemophagocytic lymphohistiocytosis in two brothers with X-linked agammaglobulinemia. Pediatr Blood Cancer. 2008;51:293–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.21573.
Huck K, et al. Girls homozygous for an IL-2-inducible T cell kinase mutation that leads to protein deficiency develop fatal EBV-associated lymphoproliferation. J Clin Invest. 2009;119:1350–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci37901.
Kuijpers TW, et al. Common variable immunodeficiency and hemophagocytic features associated with a FAS gene mutation. J Allergy Clin Immunol. 2011;127:1411-1414.e1412. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2011.01.046.
Canna SW, et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat Genet. 2014;46:1140–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.3089.
Alkhairy OK, et al. Novel mutations in TNFRSF7/CD27: Clinical, immunologic, and genetic characterization of human CD27 deficiency. J Allergy Clin Immunol. 2015;136:703-712.e710. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2015.02.022.
Lam MT, et al. A novel disorder involving dyshematopoiesis, inflammation, and HLH due to aberrant CDC42 function. J Exp Med. 2019;216:2778–99. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20190147.
Vavassori S, et al. Multisystem inflammation and susceptibility to viral infections in human ZNFX1 deficiency. J Allergy Clin Immunol. 2021;148:381–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2021.03.045.
Aricò M, et al. Hemophagocytic lymphohistiocytosis in a patient with deletion of 22q11.2. Am J Med Genet. 1999;87:329–30.
Duval M, et al. Intermittent hemophagocytic lymphohistiocytosis is a regular feature of lysinuric protein intolerance. J Pediatr. 1999;134:236–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0022-3476(99)70423-3.
Grunebaum E, Roifman CM. Gene abnormalities in patients with hemophagocytic lymphohistiocytosis. Isr Med Assoc J. 2002;4:366–9.
Rigante D, et al. First report of macrophage activation syndrome in hyperimmunoglobulinemia D with periodic fever syndrome. Arthritis Rheum. 2007;56:658–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.22409.
Chidambaram AC, Maulik K, Ramamoorthy JG, Parameswaran N. A novel mutation of adenosine deaminase causing SCID presenting as hemophagocytic lymphohistiocytosis with acute kidney injury. Br J Haematol. 2020;191:509–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.17058.
Bode SF, et al. Recent advances in the diagnosis and treatment of hemophagocytic lymphohistiocytosis. Arthritis Res Ther. 2012;14:1–12.
Henter JI, et al. HLH-94: a treatment protocol for hemophagocytic lymphohistiocytosis HLH study Group of the Histiocyte Society. Med Pediatr Oncol. 1997;28:342–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/(sici)1096-911x(199705)28:5%3c342::aid-mpo3%3e3.0.co;2-h.
Trottestam H, et al. Chemoimmunotherapy for hemophagocytic lymphohistiocytosis: long-term results of the HLH-94 treatment protocol. Blood J Am Soc Hematol. 2011;118:4577–84.
Johnson TS, et al. Etoposide selectively ablates activated T cells to control the immunoregulatory disorder hemophagocytic lymphohistiocytosis. J Immunol. 2014;192:84–91.
Zondag TCE, Lika A, van Laar JAM. The role of etoposide in the treatment of adult patients with hemophagocytic lymphohistiocytosis. Exp Hematol Oncol. 2023;12:2. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40164-022-00362-2.
Bergsten E, et al. Confirmed efficacy of etoposide and dexamethasone in HLH treatment: long-term results of the cooperative HLH-2004 study. Blood J Am Soc Hematol. 2017;130:2728–38.
Trottestam H, et al. Risk factors for early death in children with haemophagocytic lymphohistiocytosis. Acta Paediatr. 2012;101:313–8.
Verkamp B, et al. Early response markers predict survival after etoposide-based therapy of hemophagocytic lymphohistiocytosis. Blood Adv. 2023;7:7258–69.
Henter J-I, et al. Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood J Am Soc Hematol. 2002;100:2367–73.
Chen L, Wang J, Wang Z. L-DEP regimen is effective as an initial therapy for adult EBV-HLH. Ann Hematol. 2022;101:2461–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00277-022-04946-0.
Wang Y, et al. Multicenter study of combination DEP regimen as a salvage therapy for adult refractory hemophagocytic lymphohistiocytosis. Blood J Am Soc Hematol. 2015;126:2186–92.
Wang J, et al. PEG-aspargase and DEP regimen combination therapy for refractory Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. J Hematol Oncol. 2016;9:1–10.
Masood A, et al. Efficacy and safety of allogeneic hematopoietic stem cell transplant in adults with hemophagocytic lymphohistiocytosis: a systematic review of literature. Bone Marrow Transplant. 2022;57:866–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41409-022-01668-9.
Lai W, et al. Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in adults and adolescents-a life-threatening disease: analysis of 133 cases from a single center. Hematology. 2018;23:810–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/10245332.2018.1491093.
Gooptu M, et al. Favorable outcomes following allogeneic transplantation in adults with hemophagocytic lymphohistiocytosis. Blood Adv. 2023;7:2309–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/bloodadvances.2022007012.
Marsh RA, et al. Comparison of hematopoietic cell transplant conditioning regimens for hemophagocytic lymphohistiocytosis disorders. J Allergy Clin Immunol. 2022;149:1097-1104.e1092. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2021.07.031.
Sandler RD, et al. Haemophagocytic lymphohistiocytosis (HLH) following allogeneic haematopoietic stem cell transplantation (HSCT)-time to reappraise with modern diagnostic and treatment strategies? Bone Marrow Transplant. 2020;55:307–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41409-019-0637-7.
Asano T, et al. Hemophagocytic lymphohistiocytosis after hematopoietic stem cell transplantation in children: a nationwide survey in Japan. Pediatr Blood Cancer. 2012;59:110–4.
Alblooshi RM, et al. My jamais vu in post allogeneic hematopoietic cell transplant: a review on secondary hemophagocytosis in adults. Bone Marrow Transplant. 2020;55:867–72.
Abdelkefi A, et al. Hemophagocytic syndrome after hematopoietic stem cell transplantation: a prospective observational study. Int J Hematol. 2009;89:368–73.
Sandler RD, et al. Diagnosis and Management of Secondary HLH/MAS Following HSCT and CAR-T Cell Therapy in Adults; A Review of the Literature and a Survey of Practice Within EBMT Centres on Behalf of the Autoimmune Diseases Working Party (ADWP) and Transplant Complications Working Party (TCWP). Front Immunol. 2020;11:524. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.00524.
Li X, et al. Precise CRISPR-Cas9 gene repair in autologous memory T cells to treat familial hemophagocytic lymphohistiocytosis. Sci Immunol. 2024;9:eadi0042. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciimmunol.adi0042.
Dettmer-Monaco V, et al. Gene editing of hematopoietic stem cells restores T-cell response in familial hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol. 2024;153:243-255.e214. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2023.08.003.
Carmo M, et al. Perforin gene transfer into hematopoietic stem cells improves immune dysregulation in murine models of perforin deficiency. Mol Ther. 2015;23:737–45.
Ghosh S, et al. T-cell gene therapy for perforin deficiency corrects cytotoxicity defects and prevents hemophagocytic lymphohistiocytosis manifestations. J Allergy Clin Immunol. 2018;142:904-913.e903. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2017.11.050.
Soheili T, et al. Gene transfer into hematopoietic stem cells reduces HLH manifestations in a murine model of Munc13-4 deficiency. Blood Adv. 2017;1:2781–9.
Takushi SE, et al. Lentiviral gene therapy for familial hemophagocytic lymphohistiocytosis type 3, caused by UNC13D genetic defects. Hum Gene Ther. 2020;31:626–38.
Topal J, et al. Lentiviral gene transfer corrects immune abnormalities in XIAP deficiency. J Clin Immunol. 2023;43:440–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10875-022-01389-0.
Weißert K, et al. Adoptive T cell therapy cures mice from active hemophagocytic lymphohistiocytosis (HLH). EMBO Mol Med. 2022;14: e16085. https://doiorg.publicaciones.saludcastillayleon.es/10.15252/emmm.202216085.
Gupta J, et al. Primary hemophagocytic lymphohistiocytosis with prolonged primary neurologic presentation. Pediatrics. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1542/peds.2022-057848.
Blincoe A, et al. Neuroinflammatory Disease as an Isolated Manifestation of Hemophagocytic Lymphohistiocytosis. J Clin Immunol. 2020;40:901–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10875-020-00814-6.
Yildiz H, Bailly S, Van Den Neste E, Yombi JC. Clinical management of relapsed/refractory hemophagocytic lymphohistiocytosis in adult patients: a review of current strategies and emerging therapies. Ther Clin Risk Manag. 2021;17:293–304. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/tcrm.S195538.
Lee JC, Logan AC. Diagnosis and management of adult malignancy-associated hemophagocytic lymphohistiocytosis. Cancers. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers15061839.
Horne A, et al. Frequency and spectrum of central nervous system involvement in 193 children with haemophagocytic lymphohistiocytosis. Br J Haematol. 2008;140:327–35.
Abdelhay A, et al. Delay in treatment of adult hemophagocytic lymphohistiocytosis is associated with worse in-hospital outcomes. Ann Hematol. 2023;102:2989–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00277-023-05271-w.
Ivashkiv LB. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat Rev Immunol. 2018;18:545–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-018-0029-z.
De Benedetti F, Prencipe G, Bracaglia C, Marasco E, Grom AA. Targeting interferon-γ in hyperinflammation: opportunities and challenges. Nat Rev Rheumatol. 2021;17:678–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41584-021-00694-z.
Hu Y, et al. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J Hematol Oncol. 2016;9:70. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-016-0299-5.
Ding H, Wang G, Yu Z, Sun H, Wang L. Role of interferon-gamma (IFN-γ) and IFN-γ receptor 1/2 (IFNγR1/2) in regulation of immunity, infection, and cancer development: IFN-γ-dependent or independent pathway. Biomed Pharmacother. 2022;155: 113683. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2022.113683.
de Weerd NA, Nguyen T. The interferons and their receptors–distribution and regulation. Immunol Cell Biol. 2012;90:483–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/icb.2012.9.
Zaidi MR, Merlino G. The two faces of interferon-γ in cancer. Clin Cancer Res. 2011;17:6118–24.
Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. 2005;5:375–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri1604.
Han J, Wu M, Liu Z. Dysregulation in IFN-γ signaling and response: the barricade to tumor immunotherapy. Front Immunol. 2023;14:1190333. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1190333.
Hu X, Ivashkiv LB. Cross-regulation of signaling pathways by interferon-gamma: implications for immune responses and autoimmune diseases. Immunity. 2009;31:539–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2009.09.002.
Jordan MB, Hildeman D, Kappler J, Marrack P. An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder. Blood. 2004;104:735–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2003-10-3413.
Shapouri-Moghaddam A, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233:6425–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.26429.
Chen S, et al. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. 2023;8:207. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-023-01452-1.
Xu XJ, et al. Diagnostic accuracy of a specific cytokine pattern in hemophagocytic lymphohistiocytosis in children. J Pediatr. 2012;160:984-990.e981. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jpeds.2011.11.046.
Akashi K, et al. Involvement of interferon-gamma and macrophage colony-stimulating factor in pathogenesis of haemophagocytic lymphohistiocytosis in adults. Br J Haematol. 1994;87:243–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2141.1994.tb04905.x.
Yang SL, et al. Associations between inflammatory cytokines and organ damage in pediatric patients with hemophagocytic lymphohistiocytosis. Cytokine. 2016;85:14–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2016.05.022.
Canna SW, et al. Interferon-γ mediates anemia but is dispensable for fulminant Toll-like receptor 9–induced macrophage activation syndrome and hemophagocytosis in mice. Arthritis Rheum. 2013;65:1764–75.
Schmid JP, et al. Neutralization of IFNγ defeats haemophagocytosis in LCMV-infected perforin-and Rab27a-deficient mice. EMBO Mol Med. 2009;1:112–24.
Buatois V, et al. Use of a mouse model to identify a blood biomarker for IFNγ activity in pediatric secondary hemophagocytic lymphohistiocytosis. Transl Res. 2017;180:37-52.e32. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.trsl.2016.07.023.
Albeituni S, et al. Mechanisms of action of ruxolitinib in murine models of hemophagocytic lymphohistiocytosis. Blood. 2019;134:147–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2019000761.
Al-Salama Z. T. Emapalumab: first global approval. Drugs. 2019;79:99–103. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40265-018-1046-8.
Al-Salama Z. T. Emapalumab: first global approval. Drugs. 2019;79:99–103.
Locatelli F, et al. Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 2020;382:1811–22.
Rottman M, et al. IFN-gamma mediates the rejection of haematopoietic stem cells in IFN-gammaR1-deficient hosts. PLoS Med. 2008;5: e26. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pmed.0050026.
Tucci F, et al. Emapalumab treatment in an ADA-SCID patient with refractory hemophagocytic lymphohistiocytosis-related graft failure and disseminated bacillus Calmette-Guérin infection. Haematologica. 2021;106:641.
Merli P, et al. Role of interferon-γ in immune-mediated graft failure after allogeneic hematopoietic stem cell transplantation. Haematologica. 2019;104:2314.
De Benedetti F, et al. Efficacy and safety of emapalumab in macrophage activation syndrome. Ann Rheum Dis. 2023;82:857–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/ard-2022-223739.
Jordan MB. Hemophagocytic lymphohistiocytosis: A disorder of T cell activation, immune regulation, and distinctive immunopathology. Immunol Rev. 2024;322:339–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/imr.13298.
Tucci F, et al. Emapalumab treatment in an ADA-SCID patient with refractory hemophagocytic lymphohistiocytosis-related graft failure and disseminated bacillus Calmette-Guérin infection. Haematologica. 2021;106:641–6. https://doiorg.publicaciones.saludcastillayleon.es/10.3324/haematol.2020.255620.
Lounder DT, Bin Q, de Min C, Jordan MB. Treatment of refractory hemophagocytic lymphohistiocytosis with emapalumab despite severe concurrent infections. Blood Adv. 2019;3:47–50.
Garonzi C, Chinello M, Cesaro S. Emapalumab for adult and pediatric patients with hemophagocytic lymphohistiocytosis. Expert Rev Clin Pharmacol. 2021;14:527–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/17512433.2021.1901576.
Vallurupalli M, Berliner N. Emapalumab for the treatment of relapsed/refractory hemophagocytic lymphohistiocytosis. Blood. 2019;134:1783–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2019002289.
Merli P, Algeri M, Gaspari S, Locatelli F. Novel therapeutic approaches to familial HLH (emapalumab in FHL). Front Immunol. 2020;11: 608492. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.608492.
Burn TN, et al. Genetic deficiency of interferon-γ Reveals interferon-γ-independent manifestations of murine hemophagocytic lymphohistiocytosis. Arthritis Rheumatol. 2020;72:335–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.41076.
Ehl S, et al. Is neutralization of IFN-γ sufficient to control inflammation in HLH? Pediatr Blood Cancer. 2021;68: e28886. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.28886.
Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther. 2021;6:402. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-021-00791-1.
Awasthi N, Liongue C, Ward AC. STAT proteins: a kaleidoscope of canonical and non-canonical functions in immunity and cancer. J Hematol Oncol. 2021;14:198. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-021-01214-y.
Xue C, et al. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther. 2023;8:204. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-023-01468-7.
Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak–STAT signaling in the immune system. Nat Immunol. 2017;18:374–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.3691.
Argetsinger LS, et al. Identification of JAK2 as a growth hormone receptor-associated tyrosine kinase. Cell. 1993;74:237–44.
Morris R, Kershaw NJ, Babon JJ. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018;27:1984–2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pro.3519.
Wang Y, Levy DE. Comparative evolutionary genomics of the STAT family of transcription factors. Jak-Stat. 2012;1:23–36.
Haura EB, Turkson J, Jove R. Mechanisms of disease: Insights into the emerging role of signal transducers and activators of transcription in cancer. Nat Clin Pract Oncol. 2005;2:315–24.
O’Shea JJ, et al. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–28.
Gadina M, et al. Janus kinases to jakinibs: from basic insights to clinical practice. Rheumatology. 2019;58:i4–16.
Keenan C, et al. Differential effects of JAK1 vs. JAK2 inhibition in mouse models of hemophagocytic lymphohistiocytosis. Blood. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2023021046.
Przepiorka D, et al. FDA approval summary: ruxolitinib for treatment of steroid-refractory acute graft-versus-host disease. Oncologist. 2020;25:e328–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1634/theoncologist.2019-0627.
Mascarenhas J, Hoffman R. Ruxolitinib: the first FDA approved therapy for the treatment of myelofibrosis. Clin Cancer Res. 2012;18:3008–14.
Das R, et al. Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood J Am Soc Hematol. 2016;127:1666–75.
Maschalidi S, Sepulveda FE, Garrigue A, Fischer A, de Saint Basile G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood. 2016;128:60–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2016-02-700013.
Meyer LK, et al. JAK/STAT pathway inhibition sensitizes CD8 T cells to dexamethasone-induced apoptosis in hyperinflammation. Blood J Am Soc Hematol. 2020;136:657–68.
Joly JA, et al. Combined IFN-γ and JAK inhibition to treat hemophagocytic lymphohistiocytosis in mice. J Allergy Clin Immunol. 2023;151:247-259.e247. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2022.07.026.
Albeituni S, et al. Cellular and transcriptional impacts of Janus kinase and/or IFN-gamma inhibition in a mouse model of primary hemophagocytic lymphohistiocytosis. Front Immunol. 2023;14:1137037. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1137037.
Chaturvedi V, Lakes N, Tran M, Castillo N, Jordan MB. JAK inhibition for murine HLH requires complete blockade of IFN-γ signaling and is limited by toxicity of JAK2 inhibition. Blood. 2021;138:1034–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2020007930.
Wang J, et al. Ruxolitinib for refractory/relapsed hemophagocytic lymphohistiocytosis. Haematologica. 2020;105: e210.
Wei A, et al. Short-term effectiveness of ruxolitinib in the treatment of recurrent or refractory hemophagocytic lymphohistiocytosis in children. Int J Hematol. 2020;112:568–76.
Wang J, et al. Ruxolitinib-combined doxorubicin-etoposide-methylprednisolone regimen as a salvage therapy for refractory/relapsed haemophagocytic lymphohistiocytosis: a single-arm, multicentre, phase 2 trial. Br J Haematol. 2021;193:761–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.17331.
Ramanan KM, et al. Successful remission induction in refractory familial hemophagocytic lymphohistiocytosis with ruxolitinib as a bridge to hematopoietic stem cell transplantation. Pediatr Blood Cancer. 2020;67: e28071.
Zhao Y, et al. Salvage therapy with dose-escalating ruxolitinib as a bridge to allogeneic stem cell transplantation for refractory hemophagocytic lymphohistiocytosis. Bone Marrow Transplant. 2020;55:824–6.
Slostad J, et al. Ruxolitinib as first-line treatment in secondary hemophagocytic lymphohistiocytosis: a single patient experience. Am J Hematol. 2018;93:E47–9.
Zandvakili I, Conboy CB, Ayed AO, Cathcart-Rake EJ, Tefferi A. Ruxolitinib as first-line treatment in secondary hemophagocytic lymphohistiocytosis: a second experience. Am J Hematol. 2018;93:E123–5.
Fuchs A, et al. Falciparum malaria-induced secondary hemophagocytic lymphohistiocytosis successfully treated with ruxolitinib. Int J Infect Dis. 2020;100:382–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijid.2020.07.062.
Jung JI, et al. Successful treatment of hemophagocytic lymphohistiocytosis in a patient with systemic lupus erythematosus with ruxolitinib: a case report. J Rheum Dis. 2024;31:125–9. https://doiorg.publicaciones.saludcastillayleon.es/10.4078/jrd.2023.0027.
Hansen S, et al. Ruxolitinib as adjunctive therapy for secondary hemophagocytic lymphohistiocytosis: a case series. Eur J Haematol. 2021;106:654–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ejh.13593.
Liu X, et al. Case report: Ruxolitinib as first-line therapy for secondary hemophagocytic lymphohistiocytosis in patients with AIDS. Front Immunol. 2022;13:1012643. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.1012643.
Triebwasser MP, et al. Combined use of emapalumab and ruxolitinib in a patient with refractory hemophagocytic lymphohistiocytosis was safe and effective. Pediatr Blood Cancer. 2021;68: e29026. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.29026.
Adas MA, et al. The infection risks of JAK inhibition. Expert Rev Clin Immunol. 2022;18:253–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/1744666x.2022.2014323.
Ghosn L, et al. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev. 2023;6:Cd013881. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/14651858.CD013881.pub2.
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-023-00856-y.
Qiu X, et al. Interleukin-6 for early diagnosis of neonatal sepsis with premature rupture of the membranes: a meta-analysis. Medicine. 2018;97:e13146. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/md.0000000000013146.
Copaescu A, Smibert O, Gibson A, Phillips EJ, Trubiano JA. The role of IL-6 and other mediators in the cytokine storm associated with SARS-CoV-2 infection. J Allergy Clin Immunol. 2020;146:518-534.e511. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2020.07.001.
Zeng G, et al. Combination of C-reactive protein, procalcitonin, IL-6, IL-8, and IL-10 for early diagnosis of hyperinflammatory state and organ dysfunction in pediatric sepsis. J Clin Lab Anal. 2022;36: e24505. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcla.24505.
McFadden JP, Puangpet P, Basketter DA, Dearman RJ, Kimber I. Why does allergic contact dermatitis exist? Br J Dermatol. 2013;168:692–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjd.12145.
Serna-Rodríguez MF, Bernal-Vega S, de la Barquera JAO, Camacho-Morales A, Pérez-Maya AA. The role of damage associated molecular pattern molecules (DAMPs) and permeability of the blood-brain barrier in depression and neuroinflammation. J Neuroimmunol. 2022;371: 577951. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jneuroim.2022.577951.
Subramaniam S, Kothari H, Bosmann M. Tissue factor in COVID-19-associated coagulopathy. Thromb Res. 2022;220:35–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.thromres.2022.09.025.
Rougier F, Cornu E, Praloran V, Denizot Y. IL-6 and IL-8 production by human bone marrow stromal cells. Cytokine. 1998;10:93–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1006/cyto.1997.0262.
Sheryanna A, et al. Inhibition of p38 mitogen-activated protein kinase is effective in the treatment of experimental crescentic glomerulonephritis and suppresses monocyte chemoattractant protein-1 but not IL-1beta or IL-6. J Am Soc Nephrol. 2007;18:1167–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1681/asn.2006010050.
Palmqvist P, Lundberg P, Lundgren I, Hänström L, Lerner UH. IL-1beta and TNF-alpha regulate IL-6-type cytokines in gingival fibroblasts. J Dent Res. 2008;87:558–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/154405910808700614.
Virakul S, et al. Basic FGF and PDGF-BB synergistically stimulate hyaluronan and IL-6 production by orbital fibroblasts. Mol Cell Endocrinol. 2016;433:94–104. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mce.2016.05.023.
Hirano S, Zhou Q, Furuyama A, Kanno S. Differential regulation of IL-1β and IL-6 release in murine macrophages. Inflammation. 2017;40:1933–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10753-017-0634-1.
Al-Asmari SS, et al. Pharmacological targeting of STING-dependent IL-6 production in cancer cells. Front Cell Dev Biol. 2021;9: 709618. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2021.709618.
Yao X, et al. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther. 2014;141:125–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2013.09.004.
Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy. 2016;8:959–70. https://doiorg.publicaciones.saludcastillayleon.es/10.2217/imt-2016-0020.
Narazaki M, Tanaka T, Kishimoto T. The role and therapeutic targeting of IL-6 in rheumatoid arthritis. Expert Rev Clin Immunol. 2017;13:535–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/1744666x.2017.1295850.
Kishimoto T, Kang S. IL-6 revisited: from rheumatoid arthritis to CAR T cell therapy and COVID-19. Annu Rev Immunol. 2022;40:323–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-immunol-101220-023458.
Leyfman Y, et al. Cancer and COVID-19: unravelling the immunological interplay with a review of promising therapies against severe SARS-CoV-2 for cancer patients. J Hematol Oncol. 2023;16:39. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-023-01432-6.
Meng Y, et al. Cancer history is an independent risk factor for mortality in hospitalized COVID-19 patients: a propensity score-matched analysis. J Hematol Oncol. 2020;13:75. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-020-00907-0.
de Benedetti F, et al. Correlation of serum interleukin-6 levels with joint involvement and thrombocytosis in systemic juvenile rheumatoid arthritis. Arthritis Rheum. 1991;34:1158–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.1780340912.
Lasigliè D, et al. Role of IL-1 beta in the development of human T(H)17 cells: lesson from NLPR3 mutated patients. PLoS ONE. 2011;6: e20014. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0020014.
Jones SA, Jenkins BJ. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat Rev Immunol. 2018;18:773–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-018-0066-7.
Brocker C, Thompson D, Matsumoto A, Nebert DW, Vasiliou V. Evolutionary divergence and functions of the human interleukin (IL) gene family. Hum Genomics. 2010;5:30–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1479-7364-5-1-30.
Scheller J, Rose-John S. Interleukin-6 and its receptor: from bench to bedside. Med Microbiol Immunol. 2006;195:173–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00430-006-0019-9.
Garbers C, Heink S, Korn T, Rose-John S. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov. 2018;17:395–412. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrd.2018.45.
Feigerlová E, Battaglia-Hsu SF. IL-6 signaling in diabetic nephropathy: from pathophysiology to therapeutic perspectives. Cytokine Growth Factor Rev. 2017;37:57–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2017.03.003.
Mihara M, Hashizume M, Yoshida H, Suzuki M, Shiina M. IL-6/IL-6 receptor system and its role in physiological and pathological conditions. Clin Sci (Lond). 2012;122:143–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/cs20110340.
Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrc2734.
Drucker C, Gewiese J, Malchow S, Scheller J, Rose-John S. Impact of interleukin-6 classic- and trans-signaling on liver damage and regeneration. J Autoimmun. 2010;34:29–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaut.2009.08.003.
Taher MY, Davies DM, Maher J. The role of the interleukin (IL)-6/IL-6 receptor axis in cancer. Biochem Soc Trans. 2018;46:1449–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/bst20180136.
Reeh H, et al. Response to IL-6 trans- and IL-6 classic signalling is determined by the ratio of the IL-6 receptor α to gp130 expression: fusing experimental insights and dynamic modelling. Cell Commun Signal. 2019;17:46. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-019-0356-0.
Podewski EK, et al. Alterations in Janus kinase (JAK)-signal transducers and activators of transcription (STAT) signaling in patients with end-stage dilated cardiomyopathy. Circulation. 2003;107:798–802. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/01.cir.0000057545.82749.ff.
Tron K, et al. Regulation of rat heme oxygenase-1 expression by interleukin-6 via the Jak/STAT pathway in hepatocytes. J Hepatol. 2006;45:72–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jhep.2005.12.019.
Neurath MF, Finotto S. IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev. 2011;22:83–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2011.02.003.
Sansone P, Bromberg J. Targeting the interleukin-6/Jak/stat pathway in human malignancies. J Clin Oncol. 2012;30:1005–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/jco.2010.31.8907.
Calabrese LH, Rose-John S. IL-6 biology: implications for clinical targeting in rheumatic disease. Nat Rev Rheumatol. 2014;10:720–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2014.127.
Sims NA. The JAK1/STAT3/SOCS3 axis in bone development, physiology, and pathology. Exp Mol Med. 2020;52:1185–97. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-020-0445-6.
Giraldez MD, Carneros D, Garbers C, Rose-John S, Bustos M. New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology. Nat Rev Gastroenterol Hepatol. 2021;18:787–803. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41575-021-00473-x.
McElvaney OJ, Curley GF, Rose-John S, McElvaney NG. Interleukin-6: obstacles to targeting a complex cytokine in critical illness. Lancet Respir Med. 2021;9:643–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s2213-2600(21)00103-x.
Heink S, et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic T(H)17 cells. Nat Immunol. 2017;18:74–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.3632.
Strippoli R, et al. Amplification of the response to Toll-like receptor ligands by prolonged exposure to interleukin-6 in mice: implication for the pathogenesis of macrophage activation syndrome. Arthritis Rheum. 2012;64:1680–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.33496.
Billiau AD, Roskams T, Van Damme-Lombaerts R, Matthys P, Wouters C. Macrophage activation syndrome: characteristic findings on liver biopsy illustrating the key role of activated, IFN-gamma-producing lymphocytes and IL-6- and TNF-alpha-producing macrophages. Blood. 2005;105:1648–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2004-08-2997.
Schulert GS, Grom AA. Pathogenesis of macrophage activation syndrome and potential for cytokine- directed therapies. Annu Rev Med. 2015;66:145–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-med-061813-012806.
Norelli M, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24:739–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-018-0036-4.
Cifaldi L, et al. Inhibition of natural killer cell cytotoxicity by interleukin-6: implications for the pathogenesis of macrophage activation syndrome. Arthritis Rheumatol. 2015;67:3037–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.39295.
Hazen MM, et al. Mutations of the hemophagocytic lymphohistiocytosis-associated gene UNC13D in a patient with systemic juvenile idiopathic arthritis. Arthritis Rheum. 2008;58:567–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.23199.
Zhang K, et al. Macrophage activation syndrome in patients with systemic juvenile idiopathic arthritis is associated with MUNC13-4 polymorphisms. Arthritis Rheum. 2008;58:2892–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.23734.
Kaufman KM, et al. Whole-exome sequencing reveals overlap between macrophage activation syndrome in systemic juvenile idiopathic arthritis and familial hemophagocytic lymphohistiocytosis. Arthritis Rheumatol. 2014;66:3486–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.38793.
De Benedetti F, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367:2385–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1112802.
Shimizu M, et al. Tocilizumab masks the clinical symptoms of systemic juvenile idiopathic arthritis-associated macrophage activation syndrome: the diagnostic significance of interleukin-18 and interleukin-6. Cytokine. 2012;58:287–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2012.02.006.
Zhou S, Qiao J, Bai J, Wu Y, Fang H. Biological therapy of traditional therapy-resistant adult-onset Still’s disease: an evidence-based review. Ther Clin Risk Manag. 2018;14:167–71. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/tcrm.S155488.
Grom AA, Horne A, De Benedetti F. Macrophage activation syndrome in the era of biologic therapy. Nat Rev Rheumatol. 2016;12:259–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2015.179.
Freyer CW, Porter DL. Cytokine release syndrome and neurotoxicity following CAR T-cell therapy for hematologic malignancies. J Allergy Clin Immunol. 2020;146:940–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2020.07.025.
Maus MV, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-001511.
Hines MR, et al. Consensus-based guidelines for the recognition, diagnosis, and management of hemophagocytic lymphohistiocytosis in critically Ill children and adults. Crit Care Med. 2022;50:860–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/ccm.0000000000005361.
Setiadi A, Zoref-Lorenz A, Lee CY, Jordan MB, Chen LYC. Malignancy-associated haemophagocytic lymphohistiocytosis. Lancet Haematol. 2022;9:e217–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s2352-3026(21)00366-5.
Liu D. CAR-T “the living drugs”, immune checkpoint inhibitors, and precision medicine: a new era of cancer therapy. J Hematol Oncol. 2019;12:113. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-019-0819-1.
Qudus MS, et al. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol. 2023;95: e28751. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jmv.28751.
Ohmura SI, et al. Successful use of short-term add-on tocilizumab for refractory adult-onset still’s disease with macrophage activation syndrome despite treatment with high-dose glucocorticoids, cyclosporine, and etoposide. Mod Rheumatol Case Rep. 2020;4:202–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/24725625.2020.1741073.
Beelen EMJ, et al. Risk prediction and comparative efficacy of anti-TNF vs thiopurines, for preventing postoperative recurrence in Crohn’s disease: a pooled analysis of 6 trials. Clin Gastroenterol Hepatol. 2022;20:2741-2752.e2746. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cgh.2021.10.021.
D’Haens GR, van Deventer S. 25 years of anti-TNF treatment for inflammatory bowel disease: lessons from the past and a look to the future. Gut. 2021;70:1396–405. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/gutjnl-2019-320022.
Hanzel J, et al. Vedolizumab and extraintestinal manifestations in inflammatory bowel disease. Drugs. 2021;81:333–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40265-020-01460-3.
Jeong DY, et al. Induction and maintenance treatment of inflammatory bowel disease: a comprehensive review. Autoimmun Rev. 2019;18:439–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.autrev.2019.03.002.
Rubbert-Roth A, et al. Failure of anti-TNF treatment in patients with rheumatoid arthritis: the pros and cons of the early use of alternative biological agents. Autoimmun Rev. 2019;18: 102398. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.autrev.2019.102398.
Chiossone L, et al. Protection from inflammatory organ damage in a murine model of hemophagocytic lymphohistiocytosis using treatment with IL-18 binding protein. Front Immunol. 2012;3:239. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2012.00239.
Schulert GS, Grom AA. Macrophage activation syndrome and cytokine-directed therapies. Best Pract Res Clin Rheumatol. 2014;28:277–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.berh.2014.03.002.
Pachlopnik Schmid J, et al. A Griscelli syndrome type 2 murine model of hemophagocytic lymphohistiocytosis (HLH). Eur J Immunol. 2008;38:3219–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.200838488.
Dinarello CA. Interleukin-18 and the pathogenesis of inflammatory diseases. Semin Nephrol. 2007;27:98–114. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semnephrol.2006.09.013.
Kruglov AA, Lampropoulou V, Fillatreau S, Nedospasov SA. Pathogenic and protective functions of TNF in neuroinflammation are defined by its expression in T lymphocytes and myeloid cells. J Immunol. 2011;187:5660–70. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1100663.
Shenoi S, Wallace CA. Tumor necrosis factor inhibitors in the management of juvenile idiopathic arthritis: an evidence-based review. Paediatr Drugs. 2010;12:367–77. https://doiorg.publicaciones.saludcastillayleon.es/10.2165/11532610-000000000-00000.
Karki R, et al. 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. 2021;184:149-168.e117. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2020.11.025.
Wang A, et al. Specific sequences of infectious challenge lead to secondary hemophagocytic lymphohistiocytosis-like disease in mice. Proc Natl Acad Sci U S A. 2019;116:2200–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1820704116.
Aggarwal BB, Gupta SC, Kim JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood. 2012;119:651–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2011-04-325225.
Bechara R, et al. The m(6)A reader IMP2 directs autoimmune inflammation through an IL-17- and TNFα-dependent C/EBP transcription factor axis. Sci Immunol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciimmunol.abd1287.
Davignon JL, et al. Modulation of T-cell responses by anti-tumor necrosis factor treatments in rheumatoid arthritis: a review. Arthritis Res Ther. 2018;20:229. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-018-1725-6.
Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12:49–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2015.169.
O’Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev Immunol. 2002;2:37–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri702.
Horiuchi T, Mitoma H, Harashima S, Tsukamoto H, Shimoda T. Transmembrane TNF-alpha: structure, function and interaction with anti-TNF agents. Rheumatology (Oxford). 2010;49:1215–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keq031.
Qu Y, Zhao G, Li H. Forward and reverse signaling mediated by transmembrane tumor necrosis factor-alpha and TNF receptor 2: potential roles in an immunosuppressive tumor microenvironment. Front Immunol. 2017;8:1675. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2017.01675.
Ramana KV. Tumor necrosis factor-alpha converting enzyme: Implications for ocular inflammatory diseases. Int J Biochem Cell Biol. 2010;42:1076–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biocel.2010.03.011.
Aderka D. The potential biological and clinical significance of the soluble tumor necrosis factor receptors. Cytokine Growth Factor Rev. 1996;7:231–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1359-6101(96)00026-3.
Arroyo E, et al. Tumor necrosis factor-alpha and soluble TNF-alpha receptor responses in young vs. middle-aged males following eccentric exercise. Exp Gerontol. 2017;100:28–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.exger.2017.10.012.
MacEwan DJ. TNF ligands and receptors–a matter of life and death. Br J Pharmacol. 2002;135:855–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.bjp.0704549.
Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002;296:1634–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1071924.
Puimège L, Libert C, Van Hauwermeiren F. Regulation and dysregulation of tumor necrosis factor receptor-1. Cytokine Growth Factor Rev. 2014;25:285–300. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2014.03.004.
Salomon BL. Insights into the biology and therapeutic implications of TNF and regulatory T cells. Nat Rev Rheumatol. 2021;17:487–504. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41584-021-00639-6.
Ting AT, Bertrand MJM. More to Life than NF-κB in TNFR1 Signaling. Trends Immunol. 2016;37:535–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.it.2016.06.002.
Van Hauwermeiren F, Vandenbroucke RE, Libert C. Treatment of TNF mediated diseases by selective inhibition of soluble TNF or TNFR1. Cytokine Growth Factor Rev. 2011;22:311–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2011.09.004.
Atretkhany KN, Gogoleva VS, Drutskaya MS, Nedospasov SA. Distinct modes of TNF signaling through its two receptors in health and disease. J Leukoc Biol. 2020;107:893–905. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jlb.2mr0120-510r.
Chan FK. The pre-ligand binding assembly domain: a potential target of inhibition of tumour necrosis factor receptor function. Ann Rheum Dis. 2000;59(Suppl 1):i50-53. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/ard.59.suppl_1.i50.
Medler J, Wajant H. Tumor necrosis factor receptor-2 (TNFR2): an overview of an emerging drug target. Expert Opin Ther Targets. 2019;23:295–307. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/14728222.2019.1586886.
Naudé PJ, den Boer JA, Luiten PG, Eisel UL. Tumor necrosis factor receptor cross-talk. Febs j. 2011;278:888–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1742-4658.2011.08017.x.
Al-Samkari H, Berliner N. Hemophagocytic Lymphohistiocytosis. Annu Rev Pathol. 2018;13:27–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-pathol-020117-043625.
Brisse E, Wouters CH, Matthys P. Hemophagocytic lymphohistiocytosis (HLH): a heterogeneous spectrum of cytokine-driven immune disorders. Cytokine Growth Factor Rev. 2015;26:263–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2014.10.001.
Ramos-Casals M, Brito-Zerón P, López-Guillermo A, Khamashta MA, Bosch X. Adult haemophagocytic syndrome. Lancet. 2014;383:1503–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0140-6736(13)61048-x.
Duan YW, Chen SX, Li QY, Zang Y. Neuroimmune mechanisms underlying neuropathic pain: the potential role of TNF-α-necroptosis pathway. Int J Mol Sci. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23137191.
Newton K. RIPK1 and RIPK3: critical regulators of inflammation and cell death. Trends Cell Biol. 2015;25:347–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2015.01.001.
Pinci F, et al. Tumor necrosis factor is a necroptosis-associated alarmin. Front Immunol. 2022;13:1074440. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.1074440.
van Loo G, Bertrand MJM. Death by TNF: a road to inflammation. Nat Rev Immunol. 2023;23:289–303. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41577-022-00792-3.
Mázló A, et al. Types of necroinflammation, the effect of cell death modalities on sterile inflammation. Cell Death Dis. 2022;13:423. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-022-04883-w.
Tang R, et al. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 2020;13:110. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-020-00946-7.
Demarco B, et al. Caspase-8-dependent gasdermin D cleavage promotes antimicrobial defense but confers susceptibility to TNF-induced lethality. Sci Adv. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abc3465.
Dondelinger Y, et al. NF-κB-Independent Role of IKKα/IKKβ in Preventing RIPK1 Kinase-Dependent Apoptotic and Necroptotic Cell Death during TNF Signaling. Mol Cell. 2015;60:63–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molcel.2015.07.032.
Lafont E, et al. TBK1 and IKKε prevent TNF-induced cell death by RIPK1 phosphorylation. Nat Cell Biol. 2018;20:1389–99. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41556-018-0229-6.
Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114:181–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0092-8674(03)00521-x.
Wang L, Du F, Wang X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell. 2008;133:693–703. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2008.03.036.
Ahmad S, et al. The key role of TNF-TNFR2 interactions in the modulation of allergic inflammation: a review. Front Immunol. 2018;9:2572. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.02572.
Bradley JR. TNF-mediated inflammatory disease. J Pathol. 2008;214:149–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.2287.
Kouyoumdjian A, Tchervenkov J, Paraskevas S. TFNR2 in ischemia-reperfusion injury, rejection, and tolerance in transplantation. Front Immunol. 2022;13: 903913. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.903913.
Martínez-Reza I, Díaz L, García-Becerra R. Preclinical and clinical aspects of TNF-α and its receptors TNFR1 and TNFR2 in breast cancer. J Biomed Sci. 2017;24:90. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-017-0398-9.
Wajant H, Beilhack A. Targeting regulatory T Cells by addressing tumor necrosis factor and its receptors in allogeneic hematopoietic cell transplantation and cancer. Front Immunol. 2019;10:2040. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.02040.
Yang S, Wang J, Brand DD, Zheng SG. Role of TNF-TNF receptor 2 signal in regulatory T cells and its therapeutic implications. Front Immunol. 2018;9:784. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.00784.
So T, Croft M. Regulation of PI-3-kinase and akt signaling in T lymphocytes and other cells by TNFR family molecules. Front Immunol. 2013;4:139. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2013.00139.
Lamontain V, et al. Stimulation of TNF receptor type 2 expands regulatory T cells and ameliorates established collagen-induced arthritis in mice. Cell Mol Immunol. 2019;16:65–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cmi.2017.138.
Leclerc M, et al. Control of GVHD by regulatory T cells depends on TNF produced by T cells and TNFR2 expressed by regulatory T cells. Blood. 2016;128:1651–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2016-02-700849.
Torrey H, et al. Targeted killing of TNFR2-expressing tumor cells and T(regs) by TNFR2 antagonistic antibodies in advanced Sézary syndrome. Leukemia. 2019;33:1206–18. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41375-018-0292-9.
Shaikh F, He J, Bhadra P, Chen X, Siu SWI. TNF Receptor type II as an emerging drug target for the treatment of cancer, autoimmune diseases, and graft-versus-host disease: current perspectives and in silico search for small molecule binders. Front Immunol. 2018;9:1382. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.01382.
Sheng Y, Li F, Qin Z. TNF receptor 2 makes tumor necrosis factor a friend of tumors. Front Immunol. 2018;9:1170. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.01170.
Zhao X, et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest. 2012;122:4094–104. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci64115.
Mancusi A, Alvarez M, Piccinelli S, Velardi A, Pierini A. TNFR2 signaling modulates immunity after allogeneic hematopoietic cell transplantation. Cytokine Growth Factor Rev. 2019;47:54–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2019.05.001.
Santinon F, et al. Involvement of Tumor necrosis factor receptor type II in FoxP3 stability and as a marker of treg cells specifically expanded by anti-tumor necrosis factor treatments in rheumatoid arthritis. Arthritis Rheumatol. 2020;72:576–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.41134.
Humblet-Baron S, et al. IL-2 consumption by highly activated CD8 T cells induces regulatory T-cell dysfunction in patients with hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol. 2016;138:200-209.e208. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2015.12.1314.
Bo X, Broome U, Remberger M, Sumitran-Holgersson S. Tumour necrosis factor alpha impairs function of liver derived T lymphocytes and natural killer cells in patients with primary sclerosing cholangitis. Gut. 2001;49:131–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/gut.49.1.131.
Kist A, et al. Decrease of natural killer cell activity and monokine production in peripheral blood of patients treated with recombinant tumor necrosis factor. Blood. 1988;72:344–8.
Carvelli J, et al. Functional and genetic testing in adults with HLH reveals an inflammatory profile rather than a cytotoxicity defect. Blood. 2020;136:542–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2019003664.
Cron RQ, Goyal G, Chatham WW. Cytokine storm syndrome. Annu Rev Med. 2023;74:321–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-med-042921-112837.
Henter JI, et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2007;48:124–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.21039.
Janka GE. Familial and acquired hemophagocytic lymphohistiocytosis. Annu Rev Med. 2012;63:233–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-med-041610-134208.
Aeberli D, et al. Inhibition of the TNF-pathway: use of infliximab and etanercept as remission-inducing agents in cases of therapy-resistant chronic inflammatory disorders. Swiss Med Wkly. 2002;132:414–22. https://doiorg.publicaciones.saludcastillayleon.es/10.4414/smw.2002.10031.
Makay B, et al. Etanercept for therapy-resistant macrophage activation syndrome. Pediatr Blood Cancer. 2008;50:419–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.21019.
Prahalad S, Bove KE, Dickens D, Lovell DJ, Grom AA. Etanercept in the treatment of macrophage activation syndrome. J Rheumatol. 2001;28:2120–4.
Ramanan AV, Schneider R. Macrophage activation syndrome following initiation of etanercept in a child with systemic onset juvenile rheumatoid arthritis. J Rheumatol. 2003;30:401–3.
Sandhu C, Chesney A, Piliotis E, Buckstein R, Koren S. Macrophage activation syndrome after etanercept treatment. J Rheumatol. 2007;34:241–2.
Aslani N, et al. TNFAIP3 mutation causing haploinsufficiency of A20 with a hemophagocytic lymphohistiocytosis phenotype: a report of two cases. Pediatr Rheumatol Online J. 2022;20:78. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12969-022-00735-1.
Atteritano M, et al. Haemophagocytic syndrome in rheumatic patients: a systematic review. Eur Rev Med Pharmacol Sci. 2012;16:1414–24.
Stern A, Riley R, Buckley L. Worsening of macrophage activation syndrome in a patient with adult onset Still’s disease after initiation of etanercept therapy. J Clin Rheumatol. 2001;7:252–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/00124743-200108000-00013.
Baker R, Liew JW, Simonson PD, Soma LA, Starkebaum G. Macrophage activation syndrome in a patient with axial spondyloarthritis on adalimumab. Clin Rheumatol. 2019;38:603–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10067-018-4387-5.
Chauveau E, Terrier F, Casassus-Buihle D, Moncoucy X, Oddes B. Macrophage activation syndrome after treatment with infliximab for fistulated Crohn’s disease. Presse Med. 2005;34:583–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0755-4982(05)83984-3.
Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117:3720–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2010-07-273417.
Dinarello CA. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281:8–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/imr.12621.
Ghofrani Nezhad M, Jami G, Kooshkaki O, Chamani S, Naghizadeh A. The role of inflammatory cytokines (interleukin-1 and interleukin-6) as a potential biomarker in the different stages of COVID-19 (mild, severe, and critical). J Interferon Cytokine Res. 2023;43:147–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/jir.2022.0185.
Arend WP. The balance between IL-1 and IL-1Ra in disease. Cytokine Growth Factor Rev. 2002;13:323–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1359-6101(02)00020-5.
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol. 2022;86:280–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcancer.2022.05.002.
Afonina IS, Müller C, Martin SJ, Beyaert R. Proteolytic processing of interleukin-1 family cytokines: variations on a common theme. Immunity. 2015;42:991–1004. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2015.06.003.
Netea MG, van de Veerdonk FL, van der Meer JW, Dinarello CA, Joosten LA. Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol. 2015;33:49–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-immunol-032414-112306.
Aguilera M, Darby T, Melgar S. The complex role of inflammasomes in the pathogenesis of Inflammatory Bowel Diseases: lessons learned from experimental models. Cytokine Growth Factor Rev. 2014;25:715–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2014.04.003.
Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013;59:583–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jhep.2013.03.033.
Di Paolo NC, Shayakhmetov DM. Interleukin 1α and the inflammatory process. Nat Immunol. 2016;17:906–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.3503.
Hammad H, Lambrecht BN. Barrier epithelial cells and the control of type 2 immunity. Immunity. 2015;43:29–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2015.07.007.
Weigt SS, Palchevskiy V, Belperio JA. Inflammasomes and IL-1 biology in the pathogenesis of allograft dysfunction. J Clin Invest. 2017;127:2022–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci93537.
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol. 2023;16:24. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-023-01407-7.
Garlanda C, Mantovani A. Interleukin-1 in tumor progression, therapy, and prevention. Cancer Cell. 2021;39:1023–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ccell.2021.04.011.
Hausmann JS. Targeting cytokines to treat autoinflammatory diseases. Clin Immunol. 2019;206:23–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clim.2018.10.016.
Holzinger D, Kessel C, Omenetti A, Gattorno M. From bench to bedside and back again: translational research in autoinflammation. Nat Rev Rheumatol. 2015;11:573–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2015.79.
Keenan C, Nichols KE, Albeituni S. Use of the JAK inhibitor ruxolitinib in the treatment of hemophagocytic lymphohistiocytosis. Front Immunol. 2021;12: 614704. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.614704.
Sims JE, Smith DE. The IL-1 family: regulators of immunity. Nat Rev Immunol. 2010;10:89–102. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri2691.
Gabay C, Lamacchia C, Palmer G. IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol. 2010;6:232–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2010.4.
van de Veerdonk FL, Netea MG, Dinarello CA, Joosten LA. Inflammasome activation and IL-1β and IL-18 processing during infection. Trends Immunol. 2011;32:110–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.it.2011.01.003.
Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140:1756–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1053/j.gastro.2011.02.016.
Fall N, et al. Gene expression profiling of peripheral blood from patients with untreated new-onset systemic juvenile idiopathic arthritis reveals molecular heterogeneity that may predict macrophage activation syndrome. Arthritis Rheum. 2007;56:3793–804. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.22981.
Federici S, Martini A, Gattorno M. The central role of anti-IL-1 blockade in the treatment of monogenic and multi-factorial autoinflammatory diseases. Front Immunol. 2013;4:351. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2013.00351.
Ling XB, et al. Plasma profiles in active systemic juvenile idiopathic arthritis: biomarkers and biological implications. Proteomics. 2010;10:4415–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pmic.201000298.
Pascual V, Allantaz F, Arce E, Punaro M, Banchereau J. Role of interleukin-1 (IL-1) in the pathogenesis of systemic onset juvenile idiopathic arthritis and clinical response to IL-1 blockade. J Exp Med. 2005;201:1479–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20050473.
Fitzgerald AA, Leclercq SA, Yan A, Homik JE, Dinarello CA. Rapid responses to anakinra in patients with refractory adult-onset Still’s disease. Arthritis Rheum. 2005;52:1794–803. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.21061.
Ozen S, Bilginer Y. A clinical guide to autoinflammatory diseases: familial Mediterranean fever and next-of-kin. Nat Rev Rheumatol. 2014;10:135–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrrheum.2013.174.
Romano M, et al. The 2021 EULAR/American College of Rheumatology points to consider for diagnosis, management and monitoring of the interleukin-1 mediated autoinflammatory diseases: cryopyrin-associated periodic syndromes, tumour necrosis factor receptor-associated periodic syndrome, mevalonate kinase deficiency, and deficiency of the interleukin-1 receptor antagonist. Ann Rheum Dis. 2022;81:907–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/annrheumdis-2021-221801.
Crayne CB, Albeituni S, Nichols KE, Cron RQ. The immunology of macrophage activation syndrome. Front Immunol. 2019;10:119. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.00119.
Dinarello CA, van der Meer JW. Treating inflammation by blocking interleukin-1 in humans. Semin Immunol. 2013;25:469–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.smim.2013.10.008.
Durand M, Troyanov Y, Laflamme P, Gregoire G. Macrophage activation syndrome treated with anakinra. J Rheumatol. 2010;37:879–80. https://doiorg.publicaciones.saludcastillayleon.es/10.3899/jrheum.091046.
Gattorno M, et al. The pattern of response to anti-interleukin-1 treatment distinguishes two subsets of patients with systemic-onset juvenile idiopathic arthritis. Arthritis Rheum. 2008;58:1505–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.23437.
Miettunen PM, Narendran A, Jayanthan A, Behrens EM, Cron RQ. Successful treatment of severe paediatric rheumatic disease-associated macrophage activation syndrome with interleukin-1 inhibition following conventional immunosuppressive therapy: case series with 12 patients. Rheumatology (Oxford). 2011;50:417–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keq218.
Nigrovic PA, et al. Anakinra as first-line disease-modifying therapy in systemic juvenile idiopathic arthritis: report of forty-six patients from an international multicenter series. Arthritis Rheum. 2011;63:545–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.30128.
Zeft A, et al. Anakinra for systemic juvenile arthritis: the Rocky Mountain experience. J Clin Rheumatol. 2009;15:161–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/RHU.0b013e3181a4f459.
Shakoory B, et al. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial. Crit Care Med. 2016;44:275–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/ccm.0000000000001402.
Dimopoulos G, et al. Favorable anakinra responses in severe Covid-19 patients with secondary hemophagocytic lymphohistiocytosis. Cell Host Microbe. 2020;28:117-123.e111. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.chom.2020.05.007.
Hoy SM. Canakinumab: a review of its use in the management of systemic juvenile idiopathic arthritis. BioDrugs. 2015;29:133–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40259-015-0123-8.
Sawhney S, Woo P, Murray KJ. Macrophage activation syndrome: a potentially fatal complication of rheumatic disorders. Arch Dis Child. 2001;85:421–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/adc.85.5.421.
Grom AA, et al. Rate and clinical presentation of macrophage activation syndrome in patients with systemic juvenile idiopathic arthritis treated with canakinumab. Arthritis Rheumatol. 2016;68:218–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/art.39407.
Ruperto N, et al. Two randomized trials of canakinumab in systemic juvenile idiopathic arthritis. N Engl J Med. 2012;367:2396–406. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1205099.
Meng GQ, Wang JS, Wang YN, Wei N, Wang Z. Rituximab-containing immuno-chemotherapy regimens are effective for the elimination of EBV for EBV-HLH with only and mainly B lymphocytes of EBV infection. Int Immunopharmacol. 2021;96: 107606. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2021.107606.
Miyazawa H, Wada T. Immune-mediated inflammatory diseases with chronic excess of serum interleukin-18. Front Immunol. 2022;13: 930141. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.930141.
Dinarello CA, Kaplanski G. Interleukin-18 treatment options for inflammatory diseases. Expert Rev Clin Immunol. 2005;1:619–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1586/1744666X.1.4.619.
Kaplanski G. Interleukin-18: biological properties and role in disease pathogenesis. Immunol Rev. 2018;281:138–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/imr.12616.
Mühl H, Bachmann M. IL-18/IL-18BP and IL-22/IL-22BP: two interrelated couples with therapeutic potential. Cell Signal. 2019;63: 109388. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellsig.2019.109388.
Dinarello CA. IL-18: A TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol. 1999;103:11–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0091-6749(99)70518-x.
Zhou T, et al. IL-18BP is a secreted immune checkpoint and barrier to IL-18 immunotherapy. Nature. 2020;583:609–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-020-2422-6.
Nakanishi K. Unique action of interleukin-18 on T cells and other immune cells. Front Immunol. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.00763.
Hirooka Y, Nozaki Y. Interleukin-18 in inflammatory kidney disease. Front Med. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2021.639103.
Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol. 2001;19:423–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.immunol.19.1.423.
Hyodo Y, et al. IL-18 up-regulates perforin-mediated NK activity without increasing perforin messenger RNA expression by binding to constitutively expressed IL-18 receptor. J Immunol. 1999;162:1662–8.
Tang Y, et al. Excessive IL-10 and IL-18 trigger hemophagocytic lymphohistiocytosis-like hyperinflammation and enhanced myelopoiesis. J Allergy Clin Immunol. 2022;150:1154–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2022.06.017.
Takada H, et al. Oversecretion of IL-18 in haemophagocytic lymphohistiocytosis: a novel marker of disease activity. Br J Haematol. 1999;106:182–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1365-2141.1999.01504.x.
Kohno K, et al. IFN-gamma-inducing factor (IGIF) is a costimulatory factor on the activation of Th1 but not Th2 cells and exerts its effect independently of IL-12. J Immunol. 1997;158:1541–50.
Mazodier K, et al. Severe imbalance of IL-18/IL-18BP in patients with secondary hemophagocytic syndrome. Blood. 2005;106:3483–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2005-05-1980.
Krei JM, Møller HJ, Larsen JB. The role of interleukin-18 in the diagnosis and monitoring of hemophagocytic lymphohistiocytosis/macrophage activation syndrome: a systematic review. Clin Exp Immunol. 2021;203:174–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cei.13543.
Ou W, et al. Serum cytokine pattern in children with hemophagocytic lymphohistiocytosis. Ann Hematol. 2023;102:729–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00277-023-05132-6.
Girard-Guyonvarc’h C, et al. Unopposed IL-18 signaling leads to severe TLR9-induced macrophage activation syndrome in mice. Blood. 2018;131:1430–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2017-06-789552.
Yasin S, et al. IL-18 as a biomarker linking systemic juvenile idiopathic arthritis and macrophage activation syndrome. Rheumatology. 2020;59:361–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/kez282.
Shimizu M, et al. Distinct cytokine profiles of systemic-onset juvenile idiopathic arthritis-associated macrophage activation syndrome with particular emphasis on the role of interleukin-18 in its pathogenesis. Rheumatology. 2010;49:1645–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keq133.
Wada T, et al. Sustained elevation of serum interleukin-18 and its association with hemophagocytic lymphohistiocytosis in XIAP deficiency. Cytokine. 2014;65:74–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2013.09.007.
Takada H, Nomura A, Ohga S, Hara T. Interleukin-18 in hemophagocytic lymphohistiocytosis. Leuk Lymphoma. 2001;42:21–8. https://doiorg.publicaciones.saludcastillayleon.es/10.3109/10428190109097673.
Shimizu M, et al. Distinct cytokine profiles of systemic-onset juvenile idiopathic arthritis-associated macrophage activation syndrome with particular emphasis on the role of interleukin-18 in its pathogenesis. Rheumatology (Oxford). 2010;49:1645–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/rheumatology/keq133.
Dinarello CA, et al. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. 1998;63:658–64.
Griffin G, Shenoi S, Hughes GC. Hemophagocytic lymphohistiocytosis: an update on pathogenesis, diagnosis, and therapy. Best Pract Res Clin Rheumatol. 2020;34: 101515. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.berh.2020.101515.
Risma KA, Marsh RA. Hemophagocytic lymphohistiocytosis: clinical presentations and diagnosis. J Allergy Clin Immunol Pract. 2019;7:824–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaip.2018.11.050.
Latour S, Aguilar C. XIAP deficiency syndrome in humans. Semin Cell Dev Biol. 2015;39:115–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2015.01.015.
Zhao Y, et al. Clinical features and outcomes of patients with hemophagocytic lymphohistiocytosis at onset of systemic autoinflammatory disorder and compare with Epstein-Barr virus (EBV)-related hemophagocytic lymphohistiocytosis. Medicine. 2020;99:e18503. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/md.0000000000018503.
Shimizu M, Inoue N, Mizuta M, Nakagishi Y, Yachie A. Characteristic elevation of soluble TNF receptor II : I ratio in macrophage activation syndrome with systemic juvenile idiopathic arthritis. Clin Exp Immunol. 2018;191:349–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cei.13026.
Tsutsui H, Matsui K, Okamura H, Nakanishi K. Pathophysiological roles of interleukin-18 in inflammatory liver diseases. Immunol Rev. 2000;174:192–209. https://doiorg.publicaciones.saludcastillayleon.es/10.1034/j.1600-0528.2002.017418.x.
Marsh RA, et al. XIAP deficiency: a unique primary immunodeficiency best classified as X-linked familial hemophagocytic lymphohistiocytosis and not as X-linked lymphoproliferative disease. Blood. 2010;116:1079–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2010-01-256099.
Bardet J, et al. NLRC4 GOF mutations, a challenging diagnosis from neonatal age to adulthood. J Clin Med. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm10194369.
Romberg N, et al. Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation. Nat Genet. 2014;46:1135–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.3066.
Wen J, et al. Updating the NLRC4 inflammasome: from bacterial infections to autoimmunity and cancer. Front Immunol. 2021;12: 702527. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2021.702527.
Canna SW, et al. Life-threatening NLRC4-associated hyperinflammation successfully treated with IL-18 inhibition. J Allergy Clin Immunol. 2017;139:1698–701. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2016.10.022.
Dinarello C, Novick D, Kim S, Kaplanski G. Interleukin-18 and IL-18 binding protein. Front Immunol. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2013.00289.
Kim SH, et al. Structural requirements of six naturally occurring isoforms of the IL-18 binding protein to inhibit IL-18. Proc Natl Acad Sci U S A. 2000;97:1190–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.97.3.1190.
Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001;12:53–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1359-6101(00)00015-0.
Mühl H, et al. Interferon-gamma mediates gene expression of IL-18 binding protein in nonleukocytic cells. Biochem Biophys Res Commun. 2000;267:960–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1006/bbrc.1999.2064.
Treumann A, Lifely MR, Schneider P, Ferguson MA. Primary structure of CD52. J Biol Chem. 1995;270:6088–6088.
Domagała A, Kurpisz M. CD52 antigen–a review. Med Sci Monit. 2001;7:RA325–31.
Rao SP, et al. Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS ONE. 2012;7: e39416. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0039416.
Bandala-Sanchez E, et al. T cell regulation mediated by interaction of soluble CD52 with the inhibitory receptor Siglec-10. Nat Immunol. 2013;14:741–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.2610.
Saidu NEB, et al. New approaches for the treatment of chronic graft-versus-host disease: current status and future directions. Front Immunol. 2020;11: 578314. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.578314.
Ruiz-Argüelles GJ, Gil-Beristain J, Magaña M, Ruiz-Delgado GJ. Alemtuzumab-induced resolution of refractory cutaneous chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2008;14:7–9.
Marsh JC, et al. Alemtuzumab with fludarabine and cyclophosphamide reduces chronic graft-versus-host disease after allogeneic stem cell transplantation for acquired aplastic anemia. Blood J Am Soc Hematol. 2011;118:2351–7.
Demko S, Summers J, Keegan P, Pazdur R. FDA drug approval summary: alemtuzumab as single-agent treatment for B-cell chronic lymphocytic leukemia. Oncologist. 2008;13:167–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1634/theoncologist.2007-0218.
Havrdova E, Horakova D, Kovarova I. Alemtuzumab in the treatment of multiple sclerosis: key clinical trial results and considerations for use. Ther Adv Neurol Disord. 2015;8:31–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1756285614563522.
Cooles FA, et al. Immune reconstitution 20 years after treatment with alemtuzumab in a rheumatoid arthritis cohort: implications for lymphocyte depleting therapies. Arthritis Res Ther. 2016;18:302. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-016-1188-6.
Friend PJ. Alemtuzumab induction therapy in solid organ transplantation. Transplant Res. 2013;2:S5. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/2047-1440-2-s1-s5.
Stanglmaier M, Reis S, Hallek M. Rituximab and alemtuzumab induce a nonclassic, caspase-independent apoptotic pathway in B-lymphoid cell lines and in chronic lymphocytic leukemia cells. Ann Hematol. 2004;83:634–45.
Hu Y, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–70.
Crowe J, Hall V, Smith M, Cooper H, Tite J. Humanized monoclonal antibody CAMPATH-1H: myeloma cell expression of genomic constructs, nucleotide sequence of cDNA constructs and comparison of effector mechanisms of myeloma and Chinese hamster ovary cell-derived material. Clin Exp Immunol. 1992;87:105–10.
Marsh RA, et al. Salvage therapy of refractory hemophagocytic lymphohistiocytosis with alemtuzumab. Pediatr Blood Cancer. 2013;60:101–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pbc.24188.
Strout MP, Seropian S, Berliner N. Alemtuzumab as a bridge to allogeneic SCT in atypical hemophagocytic lymphohistiocytosis. Nat Rev Clin Oncol. 2010;7:415–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrclinonc.2010.40.
Keith MP, Pitchford C, Bernstein WB. Treatment of hemophagocytic lymphohistiocytosis with alemtuzumab in systemic lupus erythematosus. J Clin Rheumatol. 2012;18:134–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/RHU.0b013e31824e8d9b.
Ošep AB, Brecl E, Škerget M, Savšek L. An unforeseen reality: Hemophagocytic lymphohistiocytosis following alemtuzumab treatment for a multiple sclerosis. Clin Neurol Neurosurg. 2023;228: 107675. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clineuro.2023.107675.
Pavlasova G, Mraz M. The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy. Haematologica. 2020;105:1494–506. https://doiorg.publicaciones.saludcastillayleon.es/10.3324/haematol.2019.243543.
Eon Kuek L, Leffler M, Mackay GA, Hulett MD. The MS4A family: counting past 1, 2 and 3. Immunol Cell Biol. 2016;94:11–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/icb.2015.48.
Mattiola I, Mantovani A, Locati M. The tetraspan MS4A family in homeostasis, immunity, and disease. Trends Immunol. 2021;42:764–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.it.2021.07.002.
Boross P, Leusen JH. Mechanisms of action of CD20 antibodies. Am J Cancer Res. 2012;2:676–90.
Deans JP, et al. Association of 75/80-kDa phosphoproteins and the tyrosine kinases Lyn, Fyn, and Lck with the B cell molecule CD20: evidence against involvement of the cytoplasmic regions of CD20. J Biol Chem. 1995;270:22632–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.270.38.22632.
Smith MR. Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance. Oncogene. 2003;22:7359–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.onc.1206939.
Marsh RA. Epstein-Barr virus and hemophagocytic lymphohistiocytosis. Front Immunol. 2017;8:1902. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2017.01902.
Dolcetti R. B lymphocytes and Epstein-Barr virus: the lesson of post-transplant lymphoproliferative disorders. Autoimmun Rev. 2007;7:96–101. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.autrev.2007.02.012.
Küppers R. B cells under influence: transformation of B cells by Epstein-Barr virus. Nat Rev Immunol. 2003;3:801–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri1201.
Kilger E, Kieser A, Baumann M, Hammerschmidt W. Epstein-Barr virus-mediated B-cell proliferation is dependent upon latent membrane protein 1, which simulates an activated CD40 receptor. Embo j. 1998;17:1700–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/emboj/17.6.1700.
Anderson LJ, Longnecker R. An auto-regulatory loop for EBV LMP2A involves activation of Notch. Virology. 2008;371:257–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.virol.2007.10.009.
Mancao C, Hammerschmidt W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. 2007;110:3715–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2007-05-090142.
Walshe CA, et al. Induction of cytosolic calcium flux by CD20 is dependent upon B cell antigen receptor signaling*. J Biol Chem. 2008;283:16971–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M708459200.
Bubien JK, Zhou LJ, Bell PD, Frizzell RA, Tedder TF. Transfection of the CD20 cell surface molecule into ectopic cell types generates a Ca2+ conductance found constitutively in B lymphocytes. J Cell Biol. 1993;121:1121–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1083/jcb.121.5.1121.
Zhang N, et al. Epstein-Barr virus and neurological diseases. Front Mol Biosci. 2021;8: 816098. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmolb.2021.816098.
Ni C, et al. In-cell infection: a novel pathway for Epstein-Barr virus infection mediated by cell-in-cell structures. Cell Res. 2015;25:785–800. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cr.2015.50.
Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nat Rev Cancer. 2004;4:757–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrc1452.
Tanner J, Weis J, Fearon D, Whang Y, Kieff E. Epstein-Barr virus gp350/220 binding to the B lymphocyte C3d receptor mediates adsorption, capping, and endocytosis. Cell. 1987;50:203–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0092-8674(87)90216-9.
Fingeroth JD, et al. Epstein-Barr virus receptor of human B lymphocytes is the C3d receptor CR2. Proc Natl Acad Sci U S A. 1984;81:4510–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.81.14.4510.
Mrozek-Gorska P, et al. Epstein-Barr virus reprograms human B lymphocytes immediately in the prelatent phase of infection. Proc Natl Acad Sci U S A. 2019;116:16046–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.1901314116.
Langerak AW, Moreau E, van Gastel-Mol EJ, van der Burg M, van Dongen JJM. Detection of clonal EBV episomes in lymphoproliferations as a diagnostic tool. Leukemia. 2002;16:1572–3. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.leu.2402519.
Grzybowski B, Vishwanath VA. Hemophagocytic lymphohistiocytosis: a diagnostic conundrum. J Pediatr Neurosci. 2017;12:55–60. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/jpn.JPN_140_16.
George MR. Hemophagocytic lymphohistiocytosis: review of etiologies and management. J Blood Med. 2014;5:69–86. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/jbm.S46255.
Guisado-Vasco P, et al. Plitidepsin as a successful rescue treatment for prolonged viral SARS-CoV-2 replication in a patient with previous anti-CD20 monoclonal antibody-mediated B cell depletion and chronic lymphocytic leukemia. J Hematol Oncol. 2022;15:4. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-021-01220-0.
Cragg MS, et al. Complement-mediated lysis by anti-CD20 mAb correlates with segregation into lipid rafts. Blood. 2003;101:1045–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2002-06-1761.
Casan JML, Wong J, Northcott MJ, Opat S. Anti-CD20 monoclonal antibodies: reviewing a revolution. Hum Vaccin Immunother. 2018;14:2820–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/21645515.2018.1508624.
Luo C, et al. Efficacy and safety of new anti-CD20 monoclonal antibodies versus rituximab for induction therapy of CD20+ B-cell non-Hodgkin lymphomas: a systematic review and meta-analysis. Sci Rep. 2021;11:3255. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-82841-w.
Mössner E, et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell–mediated B-cell cytotoxicity. Blood. 2010;115:4393–402. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2009-06-225979.
Pescovitz MD. Rituximab, an anti-cd20 monoclonal antibody: history and mechanism of action. Am J Transplant. 2006;6:859–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1600-6143.2006.01288.x.
Franke A, Niederfellner GJ, Klein C, Burtscher H. Antibodies against CD20 or B-cell receptor induce similar transcription patterns in human lymphoma cell lines. PLoS ONE. 2011;6: e16596. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0016596.
Walshe CA, et al. Induction of cytosolic calcium flux by CD20 is dependent upon B Cell antigen receptor signaling. J Biol Chem. 2008;283:16971–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M708459200.
Kheirallah S, et al. Rituximab inhibits B-cell receptor signaling. Blood. 2010;115:985–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2009-08-237537.
Kroll JL, et al. Reactivation of latent viruses in individuals receiving rituximab for new onset type 1 diabetes. J Clin Virol. 2013;57:115–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jcv.2013.01.016.
Hamilton AJ, et al. Autoimmune haemolytic anaemia associated with Epstein Barr virus infection as a severe late complication after kidney transplantation and successful treatment with rituximab: case report. BMC Nephrol. 2015;16:108. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12882-015-0096-3.
Fitzgerald MP, Armstrong L, Hague R, Russell RK. A case of EBV driven haemophagocytic lymphohistiocytosis complicating a teenage Crohn’s disease patient on azathioprine, successfully treated with rituximab. J Crohns Colitis. 2013;7:314–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.crohns.2012.05.002.
San-Juan R, et al. Current preventive strategies and management of Epstein-Barr virus-related post-transplant lymphoproliferative disease in solid organ transplantation in Europe: Results of the ESGICH Questionnaire-based Cross-sectional Survey. Clin Microbiol Infect. 2015;21:604–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmi.2015.02.002.
Pagano JS, Whitehurst CB, Andrei G. Antiviral drugs for EBV. Cancers. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers10060197.
Stocker N, et al. Pre-emptive rituximab treatment for Epstein-Barr virus reactivation after allogeneic hematopoietic stem cell transplantation is a worthwhile strategy in high-risk recipients: a comparative study for immune recovery and clinical outcomes. Bone Marrow Transplant. 2020;55:586–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41409-019-0699-6.
Chellapandian D, et al. Treatment of Epstein Barr virus-induced haemophagocytic lymphohistiocytosis with rituximab-containing chemo-immunotherapeutic regimens. Br J Haematol. 2013;162:376–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.12386.
Al Asad O, et al. Alternative therapy for epstein-barr virus related hemophagocytic lymphohistiocytosis. Case Rep Oncol Med. 2015;2015: 508387. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2015/508387.
Goudarzipour K, Kajiyazdi M, Mahdaviyani A. Epstein-barr virus-induced hemophagocytic lymphohistiocytosis. Int J Hematol Oncol Stem Cell Res. 2013;7:42–5.
Minard-Colin V, et al. Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcgammaRI, FcgammaRIII, and FcgammaRIV. Blood. 2008;112:1205–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2008-01-135160.
Shannon-Lowe C, Rickinson A. The global landscape of EBV-associated tumors. Front Oncol. 2019;9:713. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2019.00713.
Kimura H. EBV in T-/NK-Cell tumorigenesis. Adv Exp Med Biol. 2018;1045:459–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-981-10-7230-7_21.
Barros MHM, Vera-Lozada G, Segges P, Hassan R, Niedobitek G. Revisiting the tissue microenvironment of infectious mononucleosis: identification of EBV infection in T cells and deep characterization of immune profiles. Front Immunol. 2019;10:146. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2019.00146.
Song Y, Wang Y, Wang Z. Requirement for etoposide in the initial treatment of Epstein-Barr virus–associated haemophagocytic lymphohistiocytosis. Br J Haematol. 2019;186:717–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.15988.
El-Mallawany NK, Curry CV, Allen CE. Haemophagocytic lymphohistiocytosis and Epstein-Barr virus: a complex relationship with diverse origins, expression and outcomes. Br J Haematol. 2022;196:31–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjh.17638.
Schäfer EJ, Jung W, Korsten P. Combination immunosuppressive therapy including rituximab for Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in adult-onset still’s disease. Case Reports in Rheumatology. 2016;2016:8605274. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2016/8605274.
Zheng H, et al. New insights into the important roles of tumor cell-intrinsic PD-1. Int J Biol Sci. 2021;17:2537–47. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijbs.60114.
Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18:153–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nri.2017.108.
Shinohara T, Taniwaki M, Ishida Y, Kawaichi M, Honjo T. Structure and chromosomal localization of the human PD-1 gene (PDCD1). Genomics. 1994;23:704–6.
Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945–54.
Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19:813–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/intimm/dxm057.
Patsoukis N, Wang Q, Strauss L, Boussiotis VA. Revisiting the PD-1 pathway. Sci Adv. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abd2712.
Chi Z, Lu Y, Yang Y, Li B, Lu P. Transcriptional and epigenetic regulation of PD-1 expression. Cell Mol Life Sci. 2021;78:3239–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-020-03737-y.
Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.
Wang X, Teng F, Kong L, Yu J. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016;9:5023–39. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/ott.S105862.
Barber DL, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–7.
Zhou F, et al. PD-1 blockade immunotherapy as a successful rescue treatment for disseminated adenovirus infection after allogeneic hematopoietic stem cell transplantation. J Hematol Oncol. 2024;17:34. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-024-01557-2.
Pi Y, Wang J, Wang Z. Massive pericardial effusion due to chronic active Epstein-Barr virus infection successfully treated with PD-1 blockade: a case report. Medicine. 2022;101:e30298. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/md.0000000000030298.
Ma Y, et al. Outcomes of programmed death protein-1 inhibitors treatment of chronic active Epstein Barr virus infection: a single center retrospective analysis. Front Immunol. 2023;14:1093719. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1093719.
Song Y, et al. PD-1 blockade and lenalidomide combination therapy for chronic active Epstein-Barr virus infection. Clin Microbiol Infect. 2023;29(796):e797-796.e713. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmi.2023.01.017.
Liu P, et al. Nivolumab treatment of relapsed/refractory Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in adults. Blood. 2020;135:826–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2019003886.
Chen R, et al. Sintilimab treatment for chronic active Epstein-Barr virus infection and Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in children. Orphanet J Rare Dis. 2023;18:297. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13023-023-02861-9.
Zhu M, et al. Rescue of HLH with T and B lymphocyte involvement due to Epstein-Barr virus by PD-1 inhibitor/ruxolitinib and rituximab combination regimens: a case report. Iran J Immunol. 2023;20:466–72. https://doiorg.publicaciones.saludcastillayleon.es/10.22034/iji.2023.99254.2629.
Xu Y, Li W, Gan J, He X, Huang X. An analysis of sintilimab combined with ruxolitinib as compassionate therapy for 12 adults with EBV-associated hemophagocytic lymphohistiocytosis. Ann Hematol. 2023;102:3325–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00277-023-05476-z.
Pi Y, Wang J, Wang Z. Successful treatment of relapsed Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis after allo-HSCT with PD-1 Blockade: a case report. Infect Drug Resist. 2022;15:3751–6. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/idr.S372998.
Sadaat M, Jang S. Hemophagocytic lymphohistiocytosis with immunotherapy: brief review and case report. J Immunother Cancer. 2018;6:49. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40425-018-0365-3.
Shah D, Shrestha R, Ramlal R, Hatton J, Saeed H. Pembrolizumab associated hemophagocytic lymphohistiocytosis. Ann Oncol. 2017;28:1403.
Satzger I, et al. Treatment-related hemophagocytic lymphohistiocytosis secondary to checkpoint inhibition with nivolumab plus ipilimumab. Eur J Cancer. 2018;93:150–3.
Takeshita M, Anai S, Mishima S, Inoue K. Coincidence of immunotherapy-associated hemophagocytic syndrome and rapid tumor regression. Ann Oncol. 2017;28:186–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/annonc/mdw537.
Saarela M, et al. Hemophagocytic lymphohistiocytosis in 2 patients with multiple sclerosis treated with alemtuzumab. Neurology. 2018;90:849–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1212/wnl.0000000000005420.
Chen L, Wang J, Wang Z. PD-1 blockade-induced hemophagocytic lymphohistiocytosis, a dilemma therapeutic outcome in 2 patients with CAEBV: a case series. Infect Drug Resist. 2024;17:1545–50. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/idr.S441460.
Xu XJ, Zhao FY, Tang YM. Fulminant cytokine release syndrome in a paediatric patient with refractory Epstein-Barr virus-associated haemophagocytic lymphohistiocytosis receiving nivolumab treatment. Clin Microbiol Infect. 2021;27:1710–2. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmi.2021.08.015.
Henter JI, et al. Diagnostic guidelines for familial hemophagocytic lymphohistiocytosis revisited. Blood. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2024025077.
Yoon SE, et al. A comprehensive analysis of adult patients with secondary hemophagocytic lymphohistiocytosis: a prospective cohort study. Ann Hematol. 2020;99:2095–104. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00277-020-04083-6.
Acknowledgements
Not applicable.
Funding
This work was supported by 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (No. ZYJC21007), 1.3.5 Project of High Altitude Medicine (No. GYYX24003), 1.3.5 Project for Artificial Intelligence (No. ZYAI24054, No. ZYAI24039), West China Hospital, Sichuan University, Key Research and Development Program of Sichuan Province (No. 2023YFS0031, No. 2023YFS0306), National Key Research and Development Program of China (No. 2022YFC2502600, 2022YFC2502603), National Natural Science Foundation of China (No. 82370192, No. 82204490), Postdoctor Research Fund of West China Hospital, Sichuan University (No. 2024HXBH149, No. 2024HXBH006), and Chengdu Science and Technology Program (2024-YF05-00266-SN).
Author information
Authors and Affiliations
Contributions
Y.W. X.S. and K.K: Conceptualization, Visualization, Writing—original draft, Writing—review & editing. Y.Y. and H.L.: Visualization. T.N. and A.Z.: Conceptualization, Supervision, Writing—review & editing. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Wu, Y., Sun, X., Kang, K. et al. Hemophagocytic lymphohistiocytosis: current treatment advances, emerging targeted therapy and underlying mechanisms. J Hematol Oncol 17, 106 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-024-01621-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-024-01621-x