Open Access

Secondary hemophagocytic lymphohistiocytosis in children (Review)

  • This article is part of the special Issue: Hemophagocytic lymphohistiocytosis
  • Authors:
    • Chiara Benevenuta
    • Ilaria Mussinatto
    • Cecilia Orsi
    • Fabio S. Timeus
  • View Affiliations

  • Published online on: July 17, 2023     https://doi.org/10.3892/etm.2023.12122
  • Article Number: 423
  • Copyright: © Benevenuta et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hemophagocytic lymphohistiocytosis (HLH) is a rare, life‑threatening condition characterized by hyperinflammation in an uncontrolled and ineffective immune response. Despite great improvement in diagnosis and treatment, it still represents a challenge in clinical management, with poor prognosis in the absence of an aggressive therapeutic approach. The present literature review focuses on secondary HLH at pediatric age, which represents a heterogeneous group in terms of etiology and therapeutic approach. It summarizes the most recent evidence on epidemiology, pathophysiology, diagnosis, treatment and prognosis, and provides a detailed description and comparison of the major subtypes of secondary HLH. Finally, it addresses the open questions with a focus on diagnosis and new treatment insights.

1. Introduction

Hemophagocytic lymphohistiocytosis (HLH) is a rare, life-threatening condition characterized by hyperinflammation in an uncontrolled and ineffective immune response (1). It was initially described in early infancy (2), but it may develop at any age, from childhood to adulthood. The spectrum of clinical symptoms of the disease is wide (3,4), ranging from fever, cytopenia and splenomegaly to shock, disseminated intravascular coagulation and multiple organ failure (5). Despite improvement in diagnosis and treatment, HLH represents a challenge in clinical management as it still results in a poor prognosis in the absence of an aggressive therapeutic approach (6,7).

The classical description of the disease distinguishes between primary forms due to a known genetic mutation [familial HLH (FHL), and HLH in the context of congenital immunodeficiency] and secondary forms triggered by an external stimulus [secondary HLH (sHLH)] (7). This classification, though widely accepted and used, is not fully effective in clinical practice (8). Often, due to the severity of the clinical picture, it is necessary to start an aggressive treatment before having clarified the primary or secondary nature of the syndrome (9).

The ongoing advances in genetic diagnosis and a deeper understanding of the molecular pathophysiology suggest new strategies in clinical management (10).

The present study focuses on sHLH at pediatric age and explored its pathogenetic and therapeutic peculiarities. To the best of our knowledge, the current review provided for the first time an extensive and exhaustive synopsis of all the known etiologies with an in-depth discussion of the individual aspects, a detailed overview of the most recent evidence and a summary of the remaining open questions.

2. Search/selection criteria

The present study reviewed the literature available on PubMed (https://pubmed-ncbi-nlm-nih-gov.bvsp.idm.oclc.org/) from 1950 to May 2022 to find all published works on sHLH at pediatric age, including case reports, meta-analyses, randomized controlled trials, reviews or systematic reviews and official guidelines. The PubMed search string was: ‘Secondary HLH OR sHLH’. The PubMed age filter ‘child: birth-18 years’ was applied. Reference lists of all articles were manually searched for cross-references and additional articles were included, if relevant. The present study obtained the full text of each article. Exclusion criteria included: A text language other than English, a focus on adult patients and a focus on primary HLH forms. Each article was selected and analyzed for inclusion by two authors in parallel. If the authors disagreed, a third author was consulted, who was responsible for the final decision.

The website ‘Clinical trial.gov’ was searched for new trends in sHLH therapy. The research string was ‘HLH, hemophagocytic lymphohistiocytoses and hemophagocytic syndrome’. The Clinical trial.gov age filter ‘child: birth-17 years’ was applied. Exclusion criteria included a focus on conditioning regimes for hematopoietic stem cell transplantation (HSCT).

3. Results of literature search

The Pubmed search found 283 articles, of which 199 were considered relevant and included. The manual search for cross references included nine more articles for a total of 204 articles. Clinical trial.gov search found 75 studies, 17 of which were included.

4. Epidemiology

HLH is rare and likely underestimated due to its difficult diagnosis. From the institution of the first international HLH registry in 1989 to the HLH-2004 therapeutic study, >700 patients have been formally evaluated worldwide (6). In Europe and Japan an HLH incidence of 1-2 per million was reported in 2005(11), however, there is the possibility that the diagnosis is under-reported, especially in developing countries (12).

5. Clinical presentation

HLH should be suspected in all patients with prolonged high-grade fever associated with splenomegaly and multiple organ involvement (7). The clinical spectrum of the disease is wide, ranging from mild organ dysfunction to multiorgan failure requiring intensive care. Central nervous system (CNS) involvement is frequent and often severe, even though it is not included in the official diagnostic criteria (7). HLH is rare, and a high grade of suspicion is essential for diagnosis. Differentiation between primary and secondary forms is mandatory since they share the same clinical picture but often require different therapeutic approaches (10).

The typical patient is an infant under 1 year of age, ill-appearing (toxic aspect), occasionally with a critical sepsis-like aspect (8). A younger age at onset suggests an underlying genetic basis, as seen in FHL or in HLH forms arising from genetic primary immunodeficiencies; however, familial HLH can present at any age, including adulthood (13).

Progressive splenomegaly is typically observed in patients, and can be associated with hepatic involvement, neurological signs, respiratory and renal failure (14). Skin rashes, erythroderma, edema or petechiae have been reported (14). Lymphadenopathy is uncommon in patients and indicates a potential underlying lymphoma (15).

Typical laboratory findings include cytopenias (at least bilinear), hypertriglyceridemia, hypofibrinogenemia (suggestive of HLH in the context of general inflammation) and hyperferritinemia. Liver function tests are frequently altered (7). Leukocytosis is not typical of HLH [except in HLH-associated with defined rheumatological conditions/macrophage activation syndrome-HLH (MAS-HLH)] (7). Hemophagocytosis is frequently observed but is neither pathognomonic nor mandatory for diagnosis (7,16).

Atypical forms, usually seen in children older than 1 year, include isolated fever of unknown origin (FUOs), isolated CNS involvement (17) or isolated acute liver failure (18,19).

6. Pathophysiology

The pathogenesis of HLH has mostly been studied in FHL. An inherited defect in the perforin/granzyme pathway or in the fusion of cytotoxic lytic granules with the surface of natural killer (NK) cells causes, in the presence of an external trigger, an over-response by cytotoxic CD8+ T lymphocytes. Viral infections are the most common triggers (20).

Cytotoxic CD8+ T cells produce large amounts of INFγ, which in turn activates macrophages. Overstimulated macrophages release large amounts of inflammatory cytokines, such as IL-1β, IL-6, IL-12, IL-18 and TNFα. There is also an increased production of IL-10, with inhibitory activity, but not sufficient to limit the phenomenon. IL-12 and IL-18 produced by macrophages in turn stimulate CD8+ T cells, amplifying the inflammatory response (1). The resulting tissue damage causes a release of IL-33 and IL1-β, which further activates the macrophages. Activated macrophages engulfblood cells and produce large amounts of ferritin. The ‘cytokine storm’ causes all the clinical manifestations of HLH, from endothelial damage to coagulopathy and multi-organ failure (1,13,21-23). The central role of INFgamma in the pathogenesis of FHL has been demonstrated in a perforin-deficient mouse model (24).

A similar pathogenesis of HLH can also be observed in patients with primary immunodeficiencies involving granule trafficking or exocytosis, such as Hermansky-Pudlak syndrome type 2, Griscelli syndrome type 2 and Chediak-Higashi syndrome II, all with reduced cy-totoxic T lymphocytes (CTL) cytotoxicity (3,4). Advances in genetic diagnosis suggest that cell killing by CTLs and NK cells can be affected from mildly to severely, thus explaining the different HLH phenotypes as a continuum (25). The known mutations in genes related to granule-mediated killing account for FHL and primary forms in general (7). Minor alterations of the CTL and NK activity, together with an external trigger, account for sHLH, severe sepsis, multi-organ failure and HLH in the context of rheumatologic diseases (25).

In all cases, a cytokine storm causing a devastating inflammation is the primal agent of the multiorgan failure, regardless of the underlying defect (24-29) and HLH should be considered a clinical syndrome of hyperinflammation with different phenotypes (10).

Moreover, this finding likely explains the numerous similarities and overlaps between HLH and other systemic inflammatory syndromes, such as septic shock, cytokine release syn-drome (CRS) following viral infections (29) and acute liver failure (30).

In 1994, the Histiocyte Society introduced an etoposide-based treatment protocol for HLH, which is still the gold standard in the therapy of the disease (31). Etoposide, dexamethasone and cyclosporine act on the pathogenetic mechanisms of HLH, inhibiting T cells and the release of inflammatory cytokines. In this respect, etoposide has a very high immunosuppressive activity (32).

Another treatment capable of acting on the pathogenesis of HLH is ATG-based immuno-therapy which has shown efficacy as a first-line therapy for FHL (33). Epstein-Barr virus (EBV)-associated HLH can occur in X-linked lymphoproliferative type (XLP)1 and XLP2, where it is caused by signaling lymphocytic activation molecule-associated protein (SAP) or X-linked inhibitor of apoptosis protein (XIAP) deficiency and by the subsequent uncontrolled EBV infection (34), or in individuals without obvious immune defects. In the latter case, activation of cytotoxic CD8+ cells by EBV-infected B cells or a direct activation of CD8+ cells or NK cells by the virus has been hypothesized (35,36). The addition of rituximab to an etoposide-based therapy is effective in the treatment of EBV-HLH, reducing the infected B lymphocyte population and EBV load (37).

The case of malignancy-associated HLH includes two completely heterogeneous mechanisms. Firstly, when HLH is a manifestation of the underlying malignancy, particularly in the case of lymphomas, the overproduction of inflammatory cytokines such as INF-γ and TNF-α is likely caused by neoplastic cells (1,38). Secondly, when HLH develops as the consequence of chemotherapy, the process is more likely to be caused by the association of drug-induced CTL suppression and infection (39).

7. Diagnosis

HLH was first described in the 1950s (2), and the first attempt to provide homogeneous diagnostic criteria by the International Histiocyte Society goes back to 1991(40). The HLH 2004 international trial established a clear and small set of diagnostic criteria that is currently the most used (31). Those diagnostic criteria do not include frequent features of the disease such as CNS involvement, and their specificity remains unknown. Furthermore, they are often inconclusive in differentiating primary and secondary forms of HLH in intensive care settings (41).

In order to accelerate diagnosis, rapid screening by flow cytometry has been used to identify some forms of p-HLH through the assessment of expression of intracellular perforin, SAP, and XIAP protein in peripheral blood cells (42) while waiting for the results of the genetic tests. Moreover, some simplified clinical algorithms have been proposed, for example, the ‘H score’ (43,44) scoring system and the ‘Minimal Parameter set’ (45), but none of these scores are conclusive in differentiating secondary HLH forms that might benefit from a different treatment.

Another notable proposal came from the North American Consortium for Histiocytosis (NACHO) in 2019(8): Patients fulfilling the HLH 2004 criteria should be diagnosed as having a ‘HLH syndrome’, while the subsequent diagnosis of ‘HLH disease’ should be restricted to the patients who additionally show distinctive immune dysregulation (FLH, HLH associated with immune compromise, rheumatological HLH/MAS) and are likely to benefit from deep immune suppression. Disorders leading to the HLH clinical symptoms, but not likely to benefit from immune suppression, should be diagnosed as ‘HLH disease mimics’ (such as infection-related HLH and iatrogenic HLH). This proposal clarifies diagnostic and therapeutic priorities, but some of the suggested categories still overlap (such as infection associated-HLH, malignancy-associated HLH and storage disorders) (41).

The most recent consensus guidelines on recognition, diagnosis and management of HLH in critically ill children (46) stress the importance of a high suspicion rate for HLH in all patients admitted to intensive care units who show a disproportionate inflammatory response and/or rapid clinical deterioration. Those patients should be thoroughly investigated for possible triggers of sHLH and treated aggressively.

New diagnostic parameters

Imaging techniques, such as CT and 18F-fluorodeoxyglucose (FDG) positron emission tomography-computed tomography (PET-CT) have been traditionally used in diagnosis to exclude an underlying malignancy. These imaging techniques can also be useful in raising HLH suspicion in the context of FUOs by showing an inflammatory disease involving bone marrow, lymph nodes and the spleen (47,48). Moreover, some 18F-FDG PET/CT metabolic parameters can help identify the etiology of sHLH in children (for example, malignancy and EBV-HLH), and provide directions for further inspection and information about the prognosis (47-49).

A high sCD25/ferritin ratio has been observed in malignancy-associated HLH (M-HLH) secondary to a lymphoma (50) and in HLH secondary to multisystem Langerhans cell histiocytosis (51), and may represent a novel useful marker for the malignancy-associated subset of sHLH (50,51).

A Chinese group suggested that using the T helper (Th)1/Th2 cytokine profile (dosage of sCD25/IFN-γ signature, TNF, IL-10, IL-6, IL-4 and IL-2 on cytometric bead array technique) for early quick diagnosis of HLH. This has demonstrated that a significant increase of IFN-γ and IL-10 and a slight increase of IL-6 is an early, specific and prognostic cytokine pattern for childhood HLH (52). More recently, to discriminate FHL from sHLH, lower IL-4 and IFN-γ levels have been demonstrated to more likely indicate primary HLH (53). This quick cytokine profile seems also useful in discriminating HLH vs. sepsis (54), and to identify EBV-HLH (55), so that a combined approach HLH2004 and Th1/Th2 profile is currently available.

Other new diagnostic tools are being developed, even though they are currently used in preclinical or research settings. Some relevant examples include studies on IFNγ and its ‘signature’ (CXCL9 and CXCL10) as serum biomarkers of disease activity in both primary HLH (pHLH) and sHLH (56), or as predictors of predominant liver involvement in sHLH (57), or studies on soluble TNF-like weak inducer of apoptosis selective elevation in HLH patients (58) and studies on secretory sphingomyelinase upregulation in HLH, with the C16 ceramide:sphingosine ratio determining a poorer prognosis (59).

Genetic analysis of polymorphisms of the TNF-α promoter (60) or of the interferon regulatory factor 5 gene (61) have identified variants that increase susceptibility to sHLH/MAS-HLH.

Determination of the NK-cell dysfunction type (type 1-4) and of the underlying genetic defect has been used to guide HSCT indication (62) and to estimate the risk of progression to acute leukemia (63).

Finally, targeted sequencing of FHL genes appears to be insufficient to identify pathogenic mechanisms in the majority of patients with HLH, and whole exome sequencing (WES) is increasingly used. WES is rapidly expanding the range of causal mutations, with a double sided effect: It accelerates diagnosis in a subset of patients, but has redefined numerous other cases of secondary HLH as primary HLH (54). Furthermore,a proved mutation in HLH-related gene cannot exclude the coexistence of an underlying malignancy (41).

8. Classification

The most recent international classification of HLH is the one provided in 2016 by the Histiocyte Society in the ‘revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages’ (64). In 2019 the NACHO (8) introduced the concept of ‘HLH syndrome’ and proposed to categorize HLH subtypes by specific etiologic associations instead of classifying HLH as ‘primary’ or ‘secondary’. The two classifications substantially overlap, so the present study referred to the 2016 Histiocyte Society classification because it is the most widely accepted and used to date (64).

Infection-associated HLH

Infection-associated HLH represents a challenge in classification. This is because infectious diseases can trigger HLH in both primary and secondary forms, and because septic shock can either be a presentation of HLH syndrome or may mime it, with great implications in therapeutic approach. A thorough infectious screening is thus highly recommended when facing an HLH syndrome (7,8).

Among the case reports the present study reviewed, the most frequent infectious triggers were viruses, such as the herpetic viruses, dengue virus, Crimean Congo hemorrhagic fever virus, Eastern equine encephalitis virus, hepatitis A virus (HAV), hepatitis B virus (HBV), human immunodeficiency virus (HIV), adenovirus, parvovirus B19, measles, influenza virus and severe acute respiratory syndrome coronavirus 2 (Sars-Cov2).

EBV has been frequently reported from all across the world, and especially from the Asian Countries (37,65-73). The reason for this geographical heterogeneity may involve a higher virulence in EBV viral strains circulating in Asia (74), or a different immune predisposition in the Asian patients, but this issue has not been clarified yet. EBV typically targets B cells, but in a subset of patients with EBV-HLH, of prevalent Asian origin again, it infects T or NK cells leading to oligoclonal or monoclonal proliferation and massive cytokine production (69,75). In extreme cases, EBV-HLH clinical picture can be difficult to differentiate from T cell lymphoproliferative disorder (69). EBV-HLH is usually quite aggressive, with a frequent involvement of CNS. Once EBV is demonstrated by serology tests or molecular biology methods, a targeted approach is recommended (74,76). Clinical scores to differentiate low risk vs. high-risk patients have been proposed (76), but have not achieved conclusive results yet. Targeted treatment is discussed later in this review.

Cytomegalovirus (CMV) is typical of newborns and immunocompromised patients (72,73,77-81) and so is the varicella zoster virus (82). Dengue virus plays a significant role in tropical countries (73,83-90). Tick-borne diseases include the Crimean-Congo hemorrhagic fever nairovirus (91,92) and the Eastern equine encephalitis virus (93). Sporadic case reports include HLH related to HAV (90,94), HBV (95), HIV (89,96), Adenovirus (72,89,97,98), Parvovirus B19(72), Measles (88) and Influenza virus (99-101).

Sars-Cov2 infection, causative of the recent COVID19 pandemic, has been reported to also be a HLH trigger, mostly in adults (102,103). In 2021, a Mexican group reported the unusual case of a 7-years old boy that developed a severe inflammatory syndrome triggered by Sars-Cov-2 and Dengue virus coinfection (104). The authors diagnosed both multisystemic inflammatory syndrome in children (MIS-C) for the presence of coronary involvement and HLH for fulfilling the HLH 2004 criteria. The patient required mechanical ventilation and maximal support treatment for 2 weeks, but completely recovered. The authors agree that it is a very unusual presentation of HLH. In 2022, an Iranian group described the case of an 8-year-old boy who developed MIS-C with heart failure requiring heart transplantation (105). Liver and bone marrow biopsy, together with typical laboratory findings, fulfilled HLH04 diagnostic criteria. He was initially treated with intravenous immunoglobulins (IVIGs) and steroids, followed by heart transplantation and the consequent heavy immunosuppression. Unfortunately, the patient died 20 days after surgery. A third case, regarding a female newborn birthed from a SARS-CoV-2-positive mother via cesarean section at 35 weeks of gestation, was reported from Jordan in 2022(106). The newborn tested positive for SARS-CoV-2 on the first day after birth and progressively developed typical HLH syndrome from the 6th day of life. The patient died on day 51 from severe respiratory failure.

Bacterial infections associated to HLH have frequently been reported in tropical countries and include Brucella (72,107-109), Salmonella enteritidis (73,90,110), Tuberculosis (88,90,111), sepsis by streptococcal infection [group B streptococcus (112) and Streptococcus suis (113)], by Listeria (72) or without identified pathogen (90). Orientia tsutsugamushi, the causing agent of scrub typhus, has been reported in India (73,89,90) and Korea (114-117). Ehrlichia chaffeensis, the agent of human monocytic ehrlichiosis, has been reported from the US (118-122). A case of S. pneumoniae 23A, serotype that is not included in the pneumococcal 13-valent conjugated vaccine (PCV-13) has been reported from Japan (123).

HLH in kidney transplant recipients has been associated with Ehrlichiosis in one case (121), and with Bartonella henselae in one other case (124). A case associated with Serratia marcescens has been reported in a preterm newborn (125).

Fungal infection-associated HLH has been reported in immunocompromised hosts (being treated for aplastic anemia and preB-acute lymphoblastic leukemia, respectively), caused by Trichosporon asahii (126).

Leishmania (71,72,127-135) and Plasmodium (90) [falciparum (136) and vivax (137)] have been reported as parasitic triggers of HLH.

M-HLH

M-HLH represents another challenging category, because it can be the initial manifestation of an underlying malignancy, it may develop during therapy as the consequence of chemotherapy or of treatment-induced mutations [treatment-induced HLH (Ch-HLH)], or it can develop as a consequence of an intercurrent infection (138-141).

HLH as a manifestation of underlying malignancy. The excess of proinflammatory cytokines produced by activated T cells infiltrating or surrounding the tumor or by the neoplastic T-cells in T/NK cell lymphoma is likely to cause M-HLH (1,38). In rare cases, the diagnosis of HLH anticipates that of malignancy by several weeks (138,142).

M-HLH is common in adults, where it accounts for ~50% of HLH cases (15), while it is significantly rarer in children, with a prevalence of 8-11% (138,143).

Lymphomas, though relatively uncommon at pediatric age, are frequently reported in association with HLH (138). Among the cases we reviewed in the present study, HLH has been associated with Hodgkin lymphoma (73,121,140,144), anaplastic large cell lymphoma (73,145,146), peripheral T-cell lymphoma (145,147), post-transplant lymphoproliferative disorder-lymphoma (148), subcutaneous panniculitis-like T-cell lymphoma (71) extranodal NK/T cell lymphoma, hepatosplenic T-cell lymphoma, systemic EBV-positive T-cell lymphoma of childhood (145). In addition, HLH and lymphoma can be alternative diagnoses as well (149), and extreme caution is required in differentiating the two conditions before starting steroid therapy.

Acute leukemia (both myeloblastic and lymphoblastic) presenting as HLH syndrome has also been reported (72,121,138-141,145).

Ch-HLH

Among the cases we reviewed in the present study, HLH developed on therapy or after treatment for juvenile myelomonocytic leukemia (150), acute monocytic leukemia (151), Langherhans cell histiocytosis (152) and solid tumors [Wilms tumor (153), neuroblastoma (72,154), rhabdomyosarcoma (140)]. Langerhans cell histiocytosis has been associated to HLH at presentation, during therapy or as a consequence of viral infection (140,142,155).

The immunosuppression induced by treatment frequently causes viral infections or reactivations, that in turn can trigger HLH. Among the cases reviewed, viral reactivation included EBV, CMV, respiratory syncytial virus, BK virus, human herpes virus 6, adenovirus and parvovirus B19 (141,156).

MAS-HLH

HLH syndrome occurring in the context of rheumatological disorders has been commonly referred to as MAS (157). While traditionally considered separate entities that share common features, it is now clear that they should be viewed as the same disease, regardless of differences in presentation and treatment (64). However, the detailed description of MAS-HLH is beyond the scope of the present work.

HLH associated with iatrogenic immune activation

Various emerging therapies, such as chimeric antigen receptor T cells (CAR-T cells) and Blinatumomab, have been associated with CRS (29). CRS shows major overlaps with HLH, so numerous authors classify it as a form of sHLH (158-160); some other authors, conversely, restrict the diagnosis of HLH following CAR-T infusion (carHLH) to the cases in which a severe CRS is associated to the typical HLH laboratory findings, often including hemophagocitosis (161,162).

CAR-T cells were officially licensed for use in children and young adults by the U.S. Food and Drug Administration in 2017 for the treatment of refractory B-cell acute lymphoblastic leukemia (163). CRS is a common severe adverse reaction following such therapies, especially in the acute phase (29). It probably results from the high amount of IFN-γ and IL-6 produced by the constitutionally activated CAR T cells (164) or by the subsequently activated macrophages (165-167) and by the targeted tumor cell lysis (168). Development of carHLH seems associated with pre-infusion NK lymphopenia (although it is not associated with impaired NK function) that is further amplified by treatment (161).

The St. Jude Children's Research Hospital group recently described a cohort of 27 pediatric patients treated with CD-19 CAR-T cells (162): 12/27 patients developed CRS alone, while four progressed to carHLH despite appropriate therapy. The carHLH subgroup showed higher and more sustained inflammation parameters and a poorer antileukemic response and survival. Blinatumomab, as well, has been associated with CRS and/or HLH in adults (29) and children (169).

Patients treated with T-activating therapies should be closely monitored for HLH, in order to optimize diagnosis and therapeutic approach.

Additional iatrogenic causes of cytokine storm include rituximab (170), gene therapies, immune checkpoint inhibitors, allogenic HSCT and heart transplantation (29).

Transplant-related HLH

HLH has been described in the context o HSCT (171,172) and, more rarely, in kidney (124,165) and liver (173-175) transplant recipients. Post-transplant HLH (PT-HLH) might be triggered by a combination of tissue damage, immunosuppressive therapy (176), alloimmune response, residual malignancies or by infections (171). Viral infections or reactivations, such as EBV and CMV but also gastrointestinal viruses, represent the most frequent trigger, but bacterial and fungal infections have also occasionally been described (124,165,172,177-179).

HLH post-HSCT (post-HSCT HLH) can occur within the first 30 days after transplantation (early onset) or later (late onset) (171). Late onset is usually related to infectious events, while for early onset the causes are not fully understood. In some cases the triggering factor may be the residual disease (171). PT-HLH diagnosis is extremely challenging because of the complex clinical and laboratory picture of the affected patients, for whom specific diagnostic criteria do not exist yet, and a high suspicion rate is essential (180).

In early onset post-HSCT HLH, aspects of differential diagnosis may occur with engraftment syndrome and acute graft-vs.-host disease (171). The treatment of PT-HLH is also particularly complex, since we are dealing with immunosuppressed patients, often with transplant-related toxicity (171). In EBV-related forms, the use of rituximab plays an important role (37,181).

HLH of unknown/uncertain origin

HLH has recently been described in the context of febrile infection related epilepsy syndrome (FIRES) (182), an epileptic encephalopathy characterized by refractory status epilepticus following unspecific febrile illnesses. The pathophysiology of FIRES has not been completely understood, but it likely to depend, once more, on dysfunctional activation of the innate immune system (182). Moreover, though rare, this association stresses the need of screening for patients with HLH presenting with severe CNS symptoms.

A curious case of HLH following spider bite (Loxosceles reclusa) has been reported from the US (183).

Drug reaction with eosinophilia and systemic symptoms induced by vancomycin, carbamazepine and levetiracetam has been described as an occasional trigger of HLH (72).

HLH and metabolic disorders

Among the cases reviewed in the present study, several patients were diagnosed with sHLH/HLH syndrome in the context of an inherited condition associated with metabolic diseases, such as Wolman's disease (184), galactosemia, Gaucher disease (185), Niemann-Pick disease, methylmalonic acidemia and propionic acidemia (186). Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (187), type 1 glycogen storage disease (188), lysosomal acid lipase deficiency (189), mucopolysaccharidosis-plus syndrome (190).

These patients fulfilled the HLH 2004 criteria, but it is possible that some HLH features, such as splenomegaly and cytopenias, were caused by the metabolic disease itself rather than by immune hyperactivation (184-190). On the other hand, it is possible that metabolites accumulation activated macrophages and triggered a proper sHLH. The link between metabolic disorders and HLH needs to be clarified, but an extensive screening for underlying metabolic diseases should be performed in patients presenting with HLH (184,185).

9. Treatment

The aim of HLH treatment is to suppress the life-threatening inflammation that leads to organ damage (23). The first goal invariably is to induce disease remission by controlling the hyper-activated T cells and the cytokine storm they generate (7,191). The second step, instead, may differ according to the underlying condition: in FHL and HLH cases, due to primary immunodeficiency, allogeneic HSCT is the only curative treatment that is currently known, while there is no consensus on sHLH best treatment. The present study agrees that, whenever possible, sHLH should be addressed according to the specific etiology, while the debate on whether treatment of the triggering disease may alone control sHLH or not is still open (7,191,192).

‘Old but gold’-The HLH 94/04

Since a neat distinction between FHL and sHLH is often impossible in the clinical setting, the HLH 94/04 protocol (7,193) suggests a pragmatic approach to HSCT indication, which remains a mainstay of therapy.

HLH 94 induction chemotherapy is based on steroids and etoposide administration for 8 weeks with or without intrathecal methotrexate, followed by maintenance with cyclosporine A (CSA), etoposide and dexamethasone pulses while waiting for HSCT (193). Chemotherapy might be suspended after 8 weeks of treatment once clinical remission is obtained and a clear genetic base is excluded (193). Otherwise, if a clear genetic base is proved, or in case of reactivation, HSCT is recommended (193).

Treatment intensification was proposed in 2004, but failed to improve the outcome (6). The Histiocyte Society thus recommends HLH-94 as the standard of care (7).

New therapeutic frontiers

The heavy mortality rate before HSCT (7) and the significant toxicities of chemotherapy urge the clinicians to find new treatments (6). This is especially true in patients with sHLH, whose prognosis widely varies from a 55% survival at 3 years with the standard protocols (193) to 100% survival in specific conditions (such as Leishmania-triggered HLH when treated appropriately) (135).

The ongoing recognition of the central role played by cytokines suggests a treatment shift towards targeted therapies. Whether these drugs can eradicate HLH alone or require the addition of etoposide remains to be clarified, as well as how they are best used [including doses, drug combinations and use in adults/children (6).

Emapalumab (NI-0501; Novimmune SA), an anti-IFN-γ monoclonal antibody has been used successfully in pediatric patients with refractory or previously untreated pHLH (194,195).

The most recent clinical trials focus on hybrid immunotherapy (ATG + etoposide + dexamethasone; NCT01104025), monoclonal antibodies such as Alemtuzumab (196) (NCT02472054) and tyrosine-kinase inhibitors such as ruxolitinib (197) (NCT04999878; NCT04120090; NCT04551131; NCT03795909; ChiCTR2000029977) and zanubrutinib (NCT05320575). Studies on selectively cytokine-targeting molecules include Tocilizumab (198) (NCT02007239), Anakinra (NCT02780583) and Emapalumab/NI-0501(195) (NCT05001737; NCT03311854; NCT03312751; NCT01818492). In the subset of EBV-HLH patients, the ongoing clinical trials focus on Rituximab (NCT05384743), anti CD20 monoclonal antibody Sintilimab and Lenalidomide (NCT05258136), Tabelecleucel (NCT04554914), programmed cell death protein 1 antibody alone (NCT05039580) or in association with Lenalidomide (NCT04084626).

In intensive care settings, nonspecific physical therapies showed interesting results, at least in the acute phase (199,200). If a massive release of cytokines/a cytokine storm is responsible for organ damage, blood purification techniques can blunt the process with a rapid non-selective effect, potentially translating into survival benefit (199). These techniques include plasma exchange, which has been successfully applied in medium resource contexts (such as Turkey and India) (201,202), and hemofiltration combined with highly immune adsorbent cartridges (119,191,200). HLH recommendations have recognized blood purification as a potential salvage treatment in adults (203). Moreover, these non-specific therapies might be combined with targeted therapies (204), but do not replace appropriate treatment of the underlying trigger of HLH, especially in patients with refractory disease (205).

The most recent consensus guidelines on recognition, diagnosis and management of HLH in critically ill children (46) stress the importance of aggressive treatment. Steroids, in particular, should not be delayed as part of first-line treatment in different protocols (HLH-94 and ATG-based protocols) and are effective in both primary and secondary HLH.

A Canadian group reported nine cases of pediatric liver transplantation in acute liver failure secondary to sHLH, suggesting that HLH might not be an absolute contraindication to transplantation and instead play a role in a very restricted subgroup of patients (19).

Specific treatment considerations for infection-related HLH

Treatment of infection-associated HLH deserves specific considerations according to the underlying agent, since some forms respond to anti-infectious treatment [namely Leishmania (131-134), Ehrlichiosis (122), scrub typhus (89,90) and other tropical diseases-associated forms], while reaction to some other pathogens displays intermediate features and benefits of standard aggressive immunosuppression.

Several studies (131,132,134,206) have reported complete response of Leishmania-associated HLH to amphotericin B therapy +/- steroids, with an excellent prognosis [3 year overall survival rate 24% in pHLH vs. 100% in visceral leishmaniasis-associated HLH; P<0.001; 2021 Iranian cohort of 60 children (133)]. A Chinese retrospective study (135) reported good results with antiparasitic therapy associated with chemotherapy according to the HLH94 protocol in patients with visceral leishmaniosis-HLH; however, the cohort includes children and adults and it is impossible to discriminate whether treatment intensification depends on age.

Human monocytic ehrlichiosis, can also trigger an HLH form that has proved to respond to antimicrobial therapy alone (doxycycline) (122).

Scrub typhus, dengue fever and tropical infections in general respond to steroids alone or to regimens without cytotoxic drugs in cases with mild to moderate disease activity (89,90).

The hepatitis-associated HLH cases reviewed in the present study have experienced complete recovery on antiviral therapy and ruxolitinib in the case of a patient with HBV (95) and after IVIG-infusion in the cases of patients with HAV (94).

A patient developing HLH after HIV diagnosis has required both antiretroviral therapy and chemotherapy according to HLH94 protocol (96).

The Crimean Congo hemorrhagic fever-associated HLH has been resolved using antiviral therapy combined with IVIG therapy and plasma exchange (91).

The case of EBV-HLH is more complex. EBV-HLH standard treatment is by the HLH94 protocol, and early initiation of etoposide-based regimens improves survival in severe EBV-HLH (8,74,207). Chemotherapy can be combined with targeted therapies such as Rituximab (37,181) or Ruxolitinib (208). Treatment intensification with the L-DEP regimen (PEG-asparaginase in combination with liposomal doxorubicin, etoposide and high-dose methylprednisolone) has been reported as salvage therapy and bridge to alloHSCT for children with refractory disease (209). Blood purification techniques (plasma exchange and continuous renal replacement therapy) have proved to be safe and effective in addition to standard chemotherapy in severe cases (199). On the other hand, patients presenting with mild-to-moderate disease activity may only require immunomodulating therapies, such as IVIGs, CSA and steroids (69). A Japanese group (210) described a cohort of 22 patients aged 6 months to 41 years in which >60% recovered after immunotherapy (IVIG, CSA 6 mg/kg/day, prednisone); they suggest that early immunotherapy may modulate T-cell activation and reduce the chance of unnecessary chemotherapy. Sporadic cases of spontaneous resolution have been reported: An American group (68) reported the cases of two adolescents who fulfilled the HLH2004 diagnostic criteria in the context of an acute EBV infection. In both cases, treatment was planned according to HLH94 protocol, but the patients spontaneously improved before the initiation of therapy. The authors recommend caution in starting aggressive treatment, especially when patients experience mild-to-moderate symptoms. These differences in treatment approaches and outcomes underline the need for evidence-based risk stratification criteria in patients with EBV-HLH.

Specific treatment considerations for M-HLH

Malignancy-triggered forms should be distinguished from the forms that occur after chemotherapy (Ch-HLH) to optimize the therapeutic approach.

Ch-HLH. Ch-HLH is often caused by infections related to immunosuppression, and benefits of anti-infectious therapy, steroids and IVIGs.

HLH as a manifestation of underlying malignancy. M-HLH is associated with increased mortality and the early detection of malignancy can favorably influence the prognosis. Treatment is not homogeneous, and can address the underlying malignancy first, be focused on HLH or a combination of the two (41,138,145,211-213). The published series refer to small numbers and suggest an individualized therapeutic approach. Aggressive HLH-directed therapy at the onset of malignancy-associated HLH can delay or complicate anticancer treatment (39,41).

Specific treatment considerations for Rheumatologic HLH (MAS-HLH)

MAS-HLH usually responds to steroid therapy or to anakinra (recombinant human IL-1 receptor antagonist), especially when given in the early course of therapy (214,215). The present study refers to specific international guidelines for more detailed considerations on MAS-HLH (157,216,217).

Specific treatment considerations for HLH associated with iatrogenic immune activation

The cytokine release syndrome following CAR-T or Blinatumomab administration usually responds to Tocilizumab, steroids, Anakinra or to a combination of such molecules (29,165-167,169). Whether carHLH should be differentiated from a severe form of CRS is still an open question, and a more precise definition of both conditions is essential in order to guide therapeutic choices in the most complex cases (162,218).

Specific treatment considerations for transplant-related HLH

Post-transplant HLH requires prompt and aggressive treatment because of the high risk of graft failure, but no international consensus exists on treatment protocol. Reported therapeutic approaches range from corticosteroids, IVIGs and low-dose etoposide (219-221) to standard HLH94 protocol with eventual rescue HSCT (172). Ruxolitinib was recently reported as salvage therapy in 2 children, with alternating results (222). Outcome is unclear, usually compromised by graft failure.

10. Prognosis

The prognosis is related to the cause that induced sHLH, with the worst outcomes in M-HLH. In the 1980s, long-term survival in HLH was <5% (223). The HLH-94 protocol improved life expectancy up to 54% of 5-year survival, with similar results in FHL and sHLH patients. Unfortunately, the subsequent 2004 revision failed to further improve the outcome (6).

An alternative therapeutic approach consisting of steroids, antithymocyte globulin and cyclosporin has obtained satisfactory results, with a 70% remission rate (33). In general, in HLH the highest mortality is in the first weeks of therapy. In patients with pHLH who underwent hematopoietic stem cell transplantation, a 5-year probability of overall survival of ~70% has been observed (224). Overall, HLH still carries a heavy burden of mortality up to the present day.

11. Conclusions

HLH is a rare disease that may develop from childhood to adult life, but HLH subtypes spread with different frequency according to the age of the patient. In early infancy, FLH forms are most typical. They are linked to genetic defects affecting the cytotoxic activity of T lymphocytes and NK cells, and invariably require allogeneic HSCT.

Secondary forms become more frequent with increasing age. They represent a heterogeneous group in terms of etiology and therapeutic approach. The severity of the clinical picture often requires heavy immunosuppression and the use of etoposide while trying to define the etiology. Some significant exceptions exist: i) Less aggressive therapeutic approaches proved to have efficacy in MAS-HLH and in the majority of infection-triggered HLH forms; and ii) anti-tumor therapy, alone or in combination with corticosteroids, can usually control M-HLH when hyperinflammation is triggered by the tumor itself.

Various viruses and EBV can trigger FHL or cause a HLH syndrome in the absence of known genetic alterations. The improvement of genetic investigations will probably clarify the genetic background of several patients with ‘secondary’ forms of HLH.

The future goal in addressing this rare and potentially fatal condition will be to provide a tailored therapy for each patient, based on its genetic and biological characteristics.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

All authors substantially contributed to the present work, both in literature review and in the elaboration of the present article. CB, IM and CO acquired, analyzed and interpreted the literature data. CB, IM and FST wrote and edited the manuscript. FST supervised the study. All authors have read and approved the final manuscript, and agree to be accountable for all aspects of the work. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Authors' information

Dr Fabio S. Timeus, ORCID 0000-0003-0095-283X.

Competing interests

The authors declare that they have no competing interests.

References

1 

Griffin G, Shenoi S and Hughes GC: Hemophagocytic lymphohistiocytosis: An update on pathogenesis, diagnosis, and therapy. Best Pract Res Clin Rheumatol. 34(101515)2020.PubMed/NCBI View Article : Google Scholar

2 

Farquhar JW and Claireaux AE: Familial haemophagocytic reticulosis. Arch Dis Child. 27:519–525. 1952.PubMed/NCBI View Article : Google Scholar

3 

Brisse E, Wouters CH and Matthys P: Advances in the pathogenesis of primary and secondary haemophagocytic lymphohistiocytosis: Differences and similarities. Br J Haematol. 174:203–217. 2016.PubMed/NCBI View Article : Google Scholar

4 

Brisse E, Matthys P and Wouters CH: Understanding the spectrum of haemophagocytic lymphohistiocytosis: Update on diagnostic challenges and therapeutic options. Br J Haematol. 174:175–187. 2016.PubMed/NCBI View Article : Google Scholar

5 

Price B, Lines J, Lewis D and Holland N: Haemophagocytic lymphohistiocytosis: A fulminant syndrome associated with multiorgan failure and high mortality that frequently masquerades as sepsis and shock. S Afr Med J. 104:401–406. 2014.PubMed/NCBI View Article : Google Scholar

6 

Bergsten E, Horne A, Aricó M, Astigarraga I, Egeler RM, Filipovich AH, Ishii E, Janka G, Ladisch S, Lehmberg K, et al: Confirmed efficacy of etoposide and dexamethasone in HLH treatment: Long-term results of the cooperative HLH-2004 study. Blood. 130:2728–2738. 2017.PubMed/NCBI View Article : Google Scholar

7 

Henter JI, Horne A, Aricó M, Egeler RM, Filipovich AH, Imashuku S, Ladisch S, McClain K, Webb D, Winiarski J and Janka G: HLH-2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 48:124–131. 2007.PubMed/NCBI View Article : Google Scholar

8 

Jordan MB, Allen CE, Greenberg J, Henry M, Hermiston ML, Kumar A, Hines M, Eckstein O, Ladisch S, Nichols KE, et al: Challenges in the diagnosis of hemophagocytic lymphohistiocytosis: Recommendations from the North American consortium for histiocytosis (NACHO). Pediatr Blood Cancer. 66(e27929)2019.PubMed/NCBI View Article : Google Scholar

9 

Ehl S, Astigarraga I, von Bahr Greenwood T, Hines M, Horne A, Ishii E, Janka G, Jordan MB, La Rosée P, Lehmberg K, et al: Recommendations for the use of etoposide-based therapy and bone marrow transplantation for the treatment of HLH: Consensus statements by the HLH steering committee of the histiocyte society. J Allergy Clin Immunol Pract. 6:1508–1517. 2018.PubMed/NCBI View Article : Google Scholar

10 

Freeman HR and Ramanan AV: Review of haemophagocytic lymphohistiocytosis. Arch Dis Child. 96:688–693. 2011.PubMed/NCBI View Article : Google Scholar

11 

Ishii E, Ohga S, Imashuku S, Yasukawa M, Tsuda H, Miura I, Yamamoto K, Horiuchi H, Takada K, Ohshima K, et al: Nationwide survey of hemophagocytic lymphohistiocytosis in Japan. Int J Hematol. 86:58–65. 2007.PubMed/NCBI View Article : Google Scholar

12 

Cleves D, Lotero V, Medina D, Perez PM, Patiño JA, Torres-Canchala L and Olaya M: Pediatric hemophagocytic lymphohistiocytosis: A rarely diagnosed entity in a developing country. BMC Pediatr. 21(411)2021.PubMed/NCBI View Article : Google Scholar

13 

Sieni E, Cetica V, Piccin A, Gherlinzoni F, Sasso FC, Rabusin M, Attard L, Bosi A, Pende D, Moretta L and Aricò M: Familial hemophagocytic lymphohistiocytosis may present during adulthood: Clinical and genetic features of a small series. PLoS One. 7(e44649)2012.PubMed/NCBI View Article : Google Scholar

14 

Malinowska I, Machaczka M, Popko K, Siwicka A, Salamonowicz M and Nasiłowska-Adamska B: Hemophagocytic syndrome in children and adults. Arch Immunol Ther Exp (Warsz). 62:385–394. 2014.PubMed/NCBI View Article : Google Scholar

15 

Ramos-Casals M, Brito-Zerón P, López-Guillermo A, Khamashta MA and Bosch X: Adult haemophagocytic syndrome. Lancet. 383:1503–1516. 2014.PubMed/NCBI View Article : Google Scholar

16 

Macheta M, Will AM, Houghton JB and Wynn RF: Prominent dyserythropoiesis in four cases of haemophagocytic lymphohistiocytosis. J Clin Pathol. 54:961–963. 2001.PubMed/NCBI View Article : Google Scholar

17 

Benson LA, Li H, Henderson LA, Solomon IH, Soldatos A, Murphy J, Bielekova B, Kennedy AL, Rivkin MJ, Davies KJ, et al: Pediatric CNS-isolated hemophagocytic lymphohistiocytosis. Neurol Neuroimmunol Neuroinflamm. 6(e560)2019.PubMed/NCBI View Article : Google Scholar

18 

de Kerguenec C, Hillaire S, Molinié V, Gardin C, Degott C, Erlinger S and Valla D: Hepatic manifestations of hemophagocytic syndrome: A study of 30 cases. Am J Gastroenterol. 96:852–857. 2001.PubMed/NCBI View Article : Google Scholar

19 

Amir AZ, Ling SC, Naqvi A, Weitzman S, Fecteau A, Grant D, Ghanekar A, Cattral M, Nalli N, Cutz E, et al: Liver transplantation for children with acute liver failure associated with secondary hemophagocytic lymphohistiocytosis. Liver Transpl. 22:1245–1253. 2016.PubMed/NCBI View Article : Google Scholar

20 

Al-Samkari H and Berliner N: Hemophagocytic lymphohistiocytosis. Annu Rev Pathol. 13:27–49. 2018.PubMed/NCBI View Article : Google Scholar

21 

Ishii E: Hemophagocytic lymphohistiocytosis in children: Pathogenesis and treatment. Front Pediatr. 4(47)2016.PubMed/NCBI View Article : Google Scholar

22 

Skinner J, Yankey B and Shelton BK: Hemophagocytic lymphohistiocytosis. AACN Adv Crit Care. 30:151–164. 2019.PubMed/NCBI View Article : Google Scholar

23 

Morimoto A, Nakazawa Y and Ishii E: Hemophagocytic lymphohistiocytosis: Pathogenesis, diagnosis, and management. Pediatr Int. 58:817–825. 2016.PubMed/NCBI View Article : Google Scholar

24 

Jordan MB, Hildeman D, Kappler J and Marrack P: An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder. Blood. 104:735–743. 2004.PubMed/NCBI View Article : Google Scholar

25 

Castillo L and Carcillo J: Secondary hemophagocytic lymphohistiocytosis and severe sepsis/systemic inflammatory response syndrome/multiorgan dysfunction syndrome/macrophage activation syndrome share common intermediate phenotypes on a spectrum of inflammation. Pediatr Crit Care Med. 10:387–392. 2009.PubMed/NCBI View Article : Google Scholar

26 

Grom AA, Horne A and De Benedetti F: Macrophage activation syndrome in the era of biologic therapy. Nat Rev Rheumatol. 12:259–268. 2016.PubMed/NCBI View Article : Google Scholar

27 

Canna SW and Behrens EM: Making sense of the cytokine storm: A conceptual framework for understanding, diagnosing, and treating hemophagocytic syndromes. Pediatr Clin North Am. 59:329–344. 2012.PubMed/NCBI View Article : Google Scholar

28 

Jordan MB: Emergence of targeted therapy for hemophagocytic lymphohistiocytosis. Hematologist. 15:2018.

29 

Fajgenbaum DC and June CH: Cytokine storm. N Engl J Med. 383:2255–2273. 2020.PubMed/NCBI View Article : Google Scholar

30 

Alonso EM, Horslen SP, Behrens EM and Doo E: Pediatric acute liver failure of undetermined cause: A research workshop. Hepatology. 65:1026–1037. 2017.PubMed/NCBI View Article : Google Scholar

31 

Henter JI, Aricò M, Egeler RM, Elinder G, Favara BE, Filipovich AH, Gadner H, Imashuku S, Janka-Schaub G, Komp D, et al: HLH-94: A treatment protocol for hemophagocytic lymphohistiocytosis. HLH study group of the histiocyte society. Med Pediatr Oncol. 28:342–347. 1997.PubMed/NCBI View Article : Google Scholar

32 

Johnson TS, Terrell CE, Millen SH, Katz JD, Hildeman DA and Jordan MB: Etoposide selectively ablates activated T cells to control the immunoregulatory disorder hemophagocytic lymphohistiocytosis. J Immunol. 192:84–91. 2014.PubMed/NCBI View Article : Google Scholar

33 

Mahlaoui N, Ouachée-Chardin M, de Saint Basile G, Neven B, Picard C, Blanche S and Fischer A: Immunotherapy of familial hemophagocytic lymphohistiocytosis with antithymocyte globulins: A single-center retrospective report of 38 patients. Pediatrics. 120:e622–e628. 2007.PubMed/NCBI View Article : Google Scholar

34 

Latour S and Fischer A: Signaling pathways involved in the T-cell-mediated immunity against Epstein-Barr virus: Lessons from genetic diseases. Immunol Rev. 291:174–189. 2019.PubMed/NCBI View Article : Google Scholar

35 

Moss DJ, Burrows SR, Khanna R, Misko IS and Sculley TB: Immune surveillance against Epstein-Barr virus. Semin Immunol. 4:97–104. 1992.PubMed/NCBI

36 

Kasahara Y and Yachie A: Cell type specific infection of Epstein-Barr virus (EBV) in EBV-associated hemophagocytic lymphohistiocytosis and chronic active EBV infection. Crit Rev Oncol Hematol. 44:283–294. 2002.PubMed/NCBI View Article : Google Scholar

37 

Balamuth NJ, Nichols KE, Paessler M and Teachey DT: Use of rituximab in conjunction with immunosuppressive chemotherapy as a novel therapy for Epstein Barr virus-associated hemophagocytic lymphohistiocytosis. J Pediatr Hematol Oncol. 29:569–573. 2007.PubMed/NCBI View Article : Google Scholar

38 

Daver N, McClain K, Allen CE, Parikh SA, Otrock Z, Rojas-Hernandez C, Blechacz B, Wang S, Minkov M, Jordan MB, et al: A consensus review on malignancy-associated hemophagocytic lymphohistiocytosis in adults. Cancer. 123:3229–3240. 2017.PubMed/NCBI View Article : Google Scholar

39 

Lehmberg K, Nichols KE, Henter JI, Girschikofsky M, Greenwood T, Jordan M, Kumar A, Minkov M, La Rosée P and Weitzman S: Study Group on Hemophagocytic Lymphohistiocytosis Subtypes of the Histiocyte Society. Consensus recommendations for the diagnosis and management of hemophagocytic lymphohistiocytosis associated with malignancies. Haematologica. 100:997–1004. 2015.PubMed/NCBI View Article : Google Scholar

40 

Henter JI, Elinder G and Ost A: Diagnostic guidelines for hemophagocytic lymphohistiocytosis. The FHL study group of the histiocyte society. Semin Oncol. 18:29–33. 1991.PubMed/NCBI

41 

Gurunathan A, Boucher AA, Mark M, Prus KM, O'Brien MM, Breese EH, Mizukawa BE, Absalon MJ, Nelson AS, Jordan MB, et al: Limitations of HLH-2004 criteria in distinguishing malignancy-associated hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 65(e27400)2018.PubMed/NCBI View Article : Google Scholar

42 

Gifford CE, Weingartner E, Villanueva J, Johnson J, Zhang K, Filipovich AH, Bleesing JJ and Marsh RA: Clinical flow cytometric screening of SAP and XIAP expression accurately identifies patients with SH2D1A and XIAP/BIRC4 mutations. Cytometry B Clin Cytom. 86:263–271. 2014.PubMed/NCBI View Article : Google Scholar

43 

Fardet L, Galicier L, Lambotte O, Marzac C, Aumont C, Chahwan D, Coppo P and Hejblum G: Development and validation of the HScore, a score for the diagnosis of reactive hemophagocytic syndrome. Arthritis Rheumatol. 66:2613–2620. 2014.PubMed/NCBI View Article : Google Scholar

44 

Debaugnies F, Mahadeb B, Ferster A, Meuleman N, Rozen L, Demulder A and Corazza F: Performances of the H-score for diagnosis of hemophagocytic lymphohistiocytosis in adult and pediatric patients. Am J Clin Pathol. 145:862–870. 2016.PubMed/NCBI View Article : Google Scholar

45 

Nakashima Y and Koga H: Using a Minimal Parameter Set for Early Diagnosis of Hemophagocytic lymphohistiocytosis in non-European children. Am J Case Rep. 23(e934037)2022.PubMed/NCBI View Article : Google Scholar

46 

Hines MR, von Bahr Greenwood T, Beutel G, Beutel K, Hays JA, Horne A, Janka G, Jordan MB, van Laar JAM, Lachmann G, et al: Consensus-based guidelines for the recognition, diagnosis, and management of hemophagocytic lymphohistiocytosis in critically Ill children and adults. Crit Care Med. 50:860–872. 2022.PubMed/NCBI View Article : Google Scholar

47 

Koç ZP, Akarsu S, Balci T and Unal K: PET/CT images of a patient with haemophagocytic lymphohistiocytosis. BMJ Case Rep. 2012(bcr0320126026)2012.PubMed/NCBI View Article : Google Scholar

48 

Wei A, Lu X, Ma H, Lian H, Yang X, Zhang L, Wang D, Chen S, Zhang Q, Li Z, et al: 18F-FDG PET/CT for identifying the potential primary diseases and predicting prognosis of secondary hemophagocytic lymphohistiocytosis in children. Contrast Media Mol Imaging. 2022(4849081)2022.PubMed/NCBI View Article : Google Scholar

49 

Yang X, Lu X, Liu J, Kan Y, Wang W, Zhang S, Liu L, Li J and Yang J: Development and validation of 18F-FDG PET/CT-based multivariable clinical prediction models for the identification of malignancy-associated hemophagocytic lymphohistiocytosis. Korean J Radiol. 23:466–478. 2022.PubMed/NCBI View Article : Google Scholar

50 

Tabata C and Tabata R: Possible prediction of underlying lymphoma by high sIL-2R/ferritin ratio in hemophagocytic syndrome. Ann Hematol. 91:63–71. 2012.PubMed/NCBI View Article : Google Scholar

51 

Tsuji T, Hirano T, Yamasaki H, Tsuji M and Tsuda H: A high sIL-2R/ferritin ratio is a useful marker for the diagnosis of lymphoma-associated hemophagocytic syndrome. Ann Hematol. 93:821–826. 2014.PubMed/NCBI View Article : Google Scholar

52 

Tang Y, Xu X, Song H, Yang S, Shi S, Wei J, Pan B, Zhao F, Liao C and Luo C: Early diagnostic and prognostic significance of a specific Th1/Th2 cytokine pattern in children with haemophagocytic syndrome. Br J Haematol. 143:84–91. 2008.PubMed/NCBI View Article : Google Scholar

53 

Chen Y, Wang Z, Luo Z, Zhao N, Yang S and Tang Y: Comparison of Th1/Th2 cytokine profiles between primary and secondary haemophagocytic lymphohistiocytosis. Ital J Pediatr. 42(50)2016.PubMed/NCBI View Article : Google Scholar

54 

Xu XJ and Tang YM: Dilemmas in diagnosis and management of hemophagocytic lymphohistiocytosis in children. World J Pediatr. 16:333–340. 2020.PubMed/NCBI View Article : Google Scholar

55 

Han XC, Ye Q, Zhang WY, Tang YM, Xu XJ and Zhang T: Cytokine profiles as novel diagnostic markers of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in children. J Crit Care. 39:72–77. 2017.PubMed/NCBI View Article : Google Scholar

56 

Buatois V, Chatel L, Cons L, Lory S, Richard F, Guilhot F, Johnson Z, Bracaglia C, De Benedetti F, de Min C, et al: Use of a mouse model to identify a blood biomarker for IFNγ activity in pediatric secondary hemophagocytic lymphohistiocytosis. Transl Res. 180:37–52.e2. 2017.PubMed/NCBI View Article : Google Scholar

57 

Prencipe G, Bracaglia C, Caiello I, Pascarella A, Francalanci P, Pardeo M, Meneghel A, Martini G, Rossi MN, Insalaco A, et al: The interferon-gamma pathway is selectively up-regulated in the liver of patients with secondary hemophagocytic lymphohistiocytosis. PLoS One. 14(e0226043)2019.PubMed/NCBI View Article : Google Scholar

58 

Nagasawa M, Yi Z, Imashuku S, Nonoyama S, Ogawa K, Okumura K and Mizutani S: Soluble TWEAK is markedly elevated in hemophagocytic lymphohistiocytosis. Am J Hematol. 83:222–225. 2008.PubMed/NCBI View Article : Google Scholar

59 

Jenkins RW, Clarke CJ, Lucas JT Jr, Shabbir M, Wu BX, Simbari F, Mueller J, Hannun YA, Lazarchick J and Shirai K: Evaluation of the role of secretory sphingomyelinase and bioactive sphingolipids as biomarkers in hemophagocytic lymphohistiocytosis. Am J Hematol. 88:E265–E272. 2013.PubMed/NCBI View Article : Google Scholar

60 

Chang YH, Lee DS, Jo HS, Cho SI, Yoon HJ, Shin S, Yoon JH, Kim HY, Hong YJ, Hong SI and Cho HI: Tumor necrosis factor alpha promoter polymorphism associated with increased susceptibility to secondary hemophagocytic lymphohistiocytosis in the Korean population. Cytokine. 36:45–50. 2006.PubMed/NCBI View Article : Google Scholar

61 

Yanagimachi M, Goto H, Miyamae T, Kadota K, Imagawa T, Mori M, Sato H, Yanagisawa R, Kaneko T, Morita S, et al: Association of IRF5 polymorphisms with susceptibility to hemophagocytic lymphohistiocytosis in children. J Clin Immunol. 31:946–951. 2011.PubMed/NCBI View Article : Google Scholar

62 

Horne A, Zheng C, Lorenz I, Löfstedt M, Montgomery SM, Janka G, Henter JI and Marion Schneider E: Subtyping of natural killer cell cytotoxicity deficiencies in haemophagocytic lymphohistocytosis provides therapeutic guidance. Br J Haematol. 129:658–666. 2005.PubMed/NCBI View Article : Google Scholar

63 

Chang TY, Jaffray J, Woda B, Newburger PE and Usmani GN: Hemophagocytic lymphohistiocytosis with MUNC13-4 gene mutation or reduced natural killer cell function prior to onset of childhood leukemia. Pediatr Blood Cancer. 56:856–858. 2011.PubMed/NCBI View Article : Google Scholar

64 

Emile JF, Abla O, Fraitag S, Horne A, Haroche J, Donadieu J, Requena-Caballero L, Jordan MB, Abdel-Wahab O, Allen CE, et al: Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 127:2672–2681. 2016.PubMed/NCBI View Article : Google Scholar

65 

Mărginean MO, Molnar E and Chinceşan MI: Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in a small child: A case report. Medicine (Baltimore). 99(e18759)2020.PubMed/NCBI View Article : Google Scholar

66 

Fałkowska A, Prądzyńska K and Drabko K: Difficult balance between EBV treatment and posttransplant immunosuppression: A successful transplant in a child with recurrent Epstein-Barr virus-induced hemophagocytic lymphohistiocytosis. Transplant Proc. 53:2035–2039. 2021.PubMed/NCBI View Article : Google Scholar

67 

Pan H, Feng DN, Song L and Sun LR: Acute myeloid leukemia following etoposide therapy for EBV-associated hemophagocytic lymphohistiocytosis: A case report and a brief review of the literature. BMC Pediatr. 16(116)2016.PubMed/NCBI View Article : Google Scholar

68 

Belyea B, Hinson A, Moran C, Hwang E, Heath J and Barfield R: Spontaneous resolution of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 55:754–756. 2010.PubMed/NCBI View Article : Google Scholar

69 

Lin MT, Chang HM, Huang CJ, Chen WL, Lin CY, Lin CY and Chuang SS: Massive expansion of EBV+ monoclonal T cells with CD5 down regulation in EBV-associated haemophagocytic lymphohistiocytosis. J Clin Pathol. 60:101–103. 2007.PubMed/NCBI View Article : Google Scholar

70 

DomInguez-Pinilla N, Baro-Fernández M and González-Granado LI: Hemophagocytic lymphohistiocytosis secondary to Epstein Barr virus and Leishmania co-infection in a toddler. J Postgrad Med. 61:44–45. 2015.PubMed/NCBI View Article : Google Scholar

71 

Gera A, Misra A, Tiwari A, Singh A and Mehndiratta S: A hungry Histiocyte, altered immunity and myriad of problems: Diagnostic challenges for Pediatric HLH. Int J Lab Hematol. 43:1443–1450. 2021.PubMed/NCBI View Article : Google Scholar

72 

Oguz MM, Sahin G, Altinel Acoglu E, Polat E, Yucel H, Oztek Celebi FZ, Unsal H, Akcaboy M, Sari E and Senel S: Secondary hemophagocytic lymphohistiocytosis in pediatric patients: A single center experience and factors that influenced patient prognosis. Pediatr Hematol Oncol. 36:1–16. 2019.PubMed/NCBI View Article : Google Scholar

73 

Paul T, Kalra M, Danewa A, Sachdeva P, Thatikonda KB, Sachdeva D and Sachdeva A: Pediatric hemophagocytic lymphohistiocytosis-a single center study. Indian Pediatr. 59:283–286. 2022.PubMed/NCBI

74 

Imashuku S, Tabata Y, Teramura T and Hibi S: Treatment strategies for Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis (EBV-HLH). Leuk Lymphoma. 39:37–49. 2000.PubMed/NCBI View Article : Google Scholar

75 

Kawaguchi H, Miyashita T, Herbst H, Niedobitek G, Asada M, Tsuchida M, Hanada R, Kinoshita A, Sakurai M, Kobayashi N, et al: Epstein-Barr virus-infected T lymphocytes in Epstein-Barr virus-associated hemophagocytic syndrome. J Clin Invest. 92:1444–1450. 1993.PubMed/NCBI View Article : Google Scholar

76 

Imashuku S: Clinical features and treatment strategies of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Crit Rev Oncol Hematol. 44:259–272. 2002.PubMed/NCBI View Article : Google Scholar

77 

Lohana C, Jhatial MA and Bokhari SWI: A challenging case of haemophagocytic lymphohistiocytosis (HLH) secondary to CMV infection in an adolescent with B-acute lymphoblastic leukemia. J Coll Physicians Surg Pak. 31:1011–1012. 2021.PubMed/NCBI View Article : Google Scholar

78 

Pessoa FS, Gonçalves VC and Lacerda EMDCB: Haemophagocytic lymphohistiocytosis secondary to intrauterine cytomegalovirus infection. Rev Inst Med Trop Sao Paulo. 63(e15)2021.PubMed/NCBI View Article : Google Scholar

79 

Eldem İ, Al-Rahawan MM and Levent F: How to treat cytomegalovirus-induced hemophagocytic lymphohistiocytosis in a child with leukemia. J Pediatr Hematol Oncol. 42:313–315. 2020.PubMed/NCBI View Article : Google Scholar

80 

Silwedel C, Frieauff E, Thomas W, Liese JG and Speer CP: Secondary haemophagocytic lymphohistiocytosis triggered by postnatally acquired cytomegalovirus infection in a late preterm infant. Infection. 45:355–359. 2017.PubMed/NCBI View Article : Google Scholar

81 

Hasan MR, Sundaram MS, Sundararaju S, Tsui KM, Karim MY, Roscoe D, Imam O, Janahi MA, Thomas E, Dobson S, et al: Unusual accumulation of a wide array of antimicrobial resistance mechanisms in a patient with cytomegalovirus-associated hemophagocytic lymphohistiocytosis: A case report. BMC Infect Dis. 20(237)2020.PubMed/NCBI View Article : Google Scholar

82 

Gur G, Cakar N, Uncu N, Ayar G, Basaran O, Taktak A, Koksoy AY, Acar B and Caycı FS: Hemophagocytic lymphohistiocytosis secondary to Varicella zoster infection in a child with Henoch-Schönlein purpura. Pediatr Int. 57:e37–e38. 2015.PubMed/NCBI View Article : Google Scholar

83 

Krishnappa A, Munusamy J, Ray S, Rameshbabu M, Bhatia P, Roy PS, Sundaram V and Kumar P: Neonatal dengue with HLH: Perks of early diagnosis and management. J Pediatr Hematol Oncol. 43:e770–e773. 2021.PubMed/NCBI View Article : Google Scholar

84 

Agrawal G, Wazir S, Sachdeva A and Kumar S: Primary dengue infection triggered haemophagocytic lymphohistiocytosis in a neonate. BMJ Case Rep. 13(e236881)2020.PubMed/NCBI View Article : Google Scholar

85 

Krithika MV, Amboiram P, Latha SM, Ninan B, Suman FR and Scott J: Neonate with haemophagocytic lymphohistiocytosis secondary to dengue infection: A case report. Trop Doct. 47:253–255. 2017.PubMed/NCBI View Article : Google Scholar

86 

Jasmine YSY, Lee SL and Kan FK: Infection associated haemophagocytic syndrome in severe dengue infection-a case series in a district hospital. Med J Malaysia. 72:62–64. 2017.PubMed/NCBI

87 

Phuakpet K, Sanpakit K, Vathana N, Takpradit C, Chokephaibulkit K and Viprakasit V: Hemophagocytic lymphohistiocytosis following dengue hemorrhagic fever in Hb H/Hb Constant Spring patient. Pediatr Int. 57:763–765. 2015.PubMed/NCBI View Article : Google Scholar

88 

Joshi R, Phatarpekar A, Currimbhoy Z and Desai M: Haemophagocytic lymphohistiocytosis: A case series from Mumbai. Ann Trop Paediatr. 31:135–140. 2011.PubMed/NCBI View Article : Google Scholar

89 

Nandhakumar D, Loganatha A, Sivasankaran M, Sivabalan S and Munirathnam D: Hemophagocytic lymphohistiocytosis in children. Indian J Pediatr. 87:526–531. 2020.PubMed/NCBI View Article : Google Scholar

90 

Simon AC, Delhi Kumar CG, Basu D and Ramesh Kumar R: Hemophagocytic lymphohistiocytosis in children: Clinical profile and outcome. J Pediatr Hematol Oncol. 42:e281–e285. 2020.PubMed/NCBI View Article : Google Scholar

91 

Gayretli Aydin ZG, Yesilbas O, Reis GP and Guven B: The first pediatric case of hemophagocytic lymphohistiocytosis secondary to Crimean-Congo haemorrhagic fever successfully treated with therapeutic plasma exchange accompanying ribavirin and intravenous immunoglobulin. J Clin Apher. 36:780–784. 2021.PubMed/NCBI View Article : Google Scholar

92 

Bıçakçı Z, Tavil B, Tezer H and Olcay L: Hemophagocytosis in a case with Crimean-Congo hemorrhagic fever and an overview of possible pathogenesis with current evidence. Turk J Pediatr. 55:344–348. 2013.PubMed/NCBI

93 

Mancao MY, Imran H, Chandra S, Estrada B, Figarola M, Sosnowski J and Vidal R: Eastern equine encephalitis virus infection and hemophagocytic lymphohistiocytosis in a 5-month-old infant. Pediatr Infect Dis J. 28:543–545. 2009.PubMed/NCBI View Article : Google Scholar

94 

Bay A, Bosnak V, Leblebisatan G, Yavuz S, Yilmaz F and Hizli S: Hemophagocytic lymphohistiocytosis in 2 pediatric patients secondary to hepatitis A virus infection. Pediatr Hematol Oncol. 29:211–214. 2012.PubMed/NCBI View Article : Google Scholar

95 

Jianguo L, Zhixuan Z, Rong L and Xiaodong S: Ruxolitinib in alleviating the cytokine storm of hemophagocytic lymphohistiocytosis. Pediatrics. 146(e20191301)2020.PubMed/NCBI View Article : Google Scholar

96 

Chiperi LE, Ionescu AD, Marcu CT, Itu-Mureşan C and Pantelemon C: Hemophagocytic lymphohistiocytosis in a child with human immunodeficiency virus infection-a case report. Rom J Morphol Embryol. 62:279–282. 2021.PubMed/NCBI View Article : Google Scholar

97 

Censoplano N, Gorga S, Waldeck K, Stillwell T, Rabah-Hammad R and Flori H: Neonatal adenovirus infection complicated by hemophagocytic lymphohistiocytosis syndrome. Pediatrics. 141 (Suppl 5):S475–S480. 2018.PubMed/NCBI View Article : Google Scholar

98 

Otto WR, Behrens EM, Teachey DT, Lamson DM, Barrett DM, Bassiri H, Lambert MP, Mount S, Petrosa WL, Romberg N, et al: Human adenovirus 7-associated hemophagocytic lymphohistiocytosis-like illness: Clinical and virological characteristics in a cluster of five pediatric cases. Clin Infect Dis. 73:e1532–e1538. 2021.PubMed/NCBI View Article : Google Scholar

99 

Zhang XY, Ye XW, Feng DX, Han J, Li D and Zhang C: Hemophagocytic lymphohistiocytosis induced by severe pandemic influenza A (H1N1) 2009 virus infection: A case report. Case Rep Med. 2011(951910)2011.PubMed/NCBI View Article : Google Scholar

100 

Shrestha B, Omran A, Rong P and Wang W: Report of a fatal pediatric case of hemophagocytic lymphohistiocytosis associated with pandemic influenza A (H1N1) infection in 2009. Pediatr Neonatol. 56:189–192. 2015.PubMed/NCBI View Article : Google Scholar

101 

Ozdemir H, Çiftçi E, Ince EU, Ertem M, Ince E and Doğru U: Hemophagocytic lymphohistiocytosis associated with 2009 pandemic influenza A (H1N1) virus infection. J Pediatr Hematol Oncol. 33:135–137. 2011.PubMed/NCBI View Article : Google Scholar

102 

Tholin B, Hauge MT, Aukrust P, Fehrle L and Tvedt TH: Hemophagocytic lymphohistiocytosis in a patient with COVID-19 treated with tocilizumab: A case report. J Med Case Rep. 14(187)2020.PubMed/NCBI View Article : Google Scholar

103 

Hakim NN, Chi J, Olazagasti C and Liu JM: Secondary hemophagocytic lymphohistiocytosis versus cytokine release syndrome in severe COVID-19 patients. Exp Biol Med (Maywood). 246:5–9. 2021.PubMed/NCBI View Article : Google Scholar

104 

Jose PMM, Paola ZS, Eduardo DG, Arturo SMMO and Fernando BG: A case of coinfection of a pediatric patient with acute SARS-COV-2 with MIS-C and severe DENV-2 in Mexico: A case report. BMC Infect Dis. 21(1072)2021.PubMed/NCBI View Article : Google Scholar

105 

Mahdavi M, Hejri GM, Pouraliakbar H, Shahzadi H, Hesami M and Houshmand G: Cytokine storm after heart transplantation in COVID-19-related haemophagocytic lymphohistiocytosis (HLH). ESC Heart Fail. 9:219–223. 2022.PubMed/NCBI View Article : Google Scholar

106 

El-Isa HZ, Khader OA, Khader M, Ashour BA, Azzam MI and Badran EF: Secondary hemophagocytic lymphohistiocytosis in a neonate with SARS-CoV-2 infection. Am J Case Rep. 23(e934839)2022.PubMed/NCBI View Article : Google Scholar

107 

Mittal J, Kumar P, Goyal JP and Purohit A: Haemophagocytic lymphohistiocytosis secondary to brucellosis in a young child. BMJ Case Rep. 14(e240759)2021.PubMed/NCBI View Article : Google Scholar

108 

Pekpak E and Sirvan Cetin B: Secondary hemophagocytic lymphohistocytosis in a child with brucellosis. J Pediatr Hematol Oncol. 39:e501–e503. 2017.PubMed/NCBI View Article : Google Scholar

109 

Yaman Y, Gözmen S, Özkaya AK, Oymak Y, Apa H, Vergin C and Devrim İ: Secondary hemophagocytic lymphohistiocytosis in children with brucellosis: Report of three cases. J Infect Dev Ctries. 9:1172–1176. 2015.PubMed/NCBI View Article : Google Scholar

110 

Banday AZ, Mehta R, Vignesh P, Kanaujia R, Durgadevi S, Angrup A, Kumar N, Ray P and Suri D: Case Report: Ceftriaxone-resistant invasive Salmonella enteritidis infection with secondary hemophagocytic lymphohistiocytosis: A contrast with enteric fever. Am J Trop Med Hyg. 103:2515–2517. 2020.PubMed/NCBI View Article : Google Scholar

111 

Maheshwari P, Chhabra R and Yadav P: Perinatal tuberculosis associated hemophagocytic lymphohistiocytosis. Indian J Pediatr. 79:1228–1229. 2012.PubMed/NCBI View Article : Google Scholar

112 

Choi YB and Yi DY: Fatal case of hemophagocytic lymphohistiocytosis associated with group B streptococcus sepsis: A case report. Medicine (Baltimore). 97(e12210)2018.PubMed/NCBI View Article : Google Scholar

113 

Liu SS, Wang Y, Xue L, Ma C and Li CH: Hemophagocytic lymphohistiocytosis due to Streptococcus suis in a 12-year-old girl: A case report. Medicine (Baltimore). 98(e15136)2019.PubMed/NCBI View Article : Google Scholar

114 

Sahu SK, Behera JR and Yadav SK: Scrub typhus with secondary hemophagocytic lymphohistiocytosis in a 3-month-old child from a tertiary care hospital of Odisha. Indian J Public Health. 65:85–86. 2021.PubMed/NCBI View Article : Google Scholar

115 

Han DK, Baek HJ, Shin MG, Kim JW, Kook H and Hwang TJ: Scrub typhus-associated severe hemophagocytic lymphohistiocytosis with encephalomyelitis leading to permanent sequelae: A case report and review of the literature. J Pediatr Hematol Oncol. 34:531–533. 2012.PubMed/NCBI View Article : Google Scholar

116 

Agrwal S, Dabas A, Mantan M and Yadav S: Hemophagocytic lymphohistiocytosis with neurological manifestations in an infant with scrub typhus: A rare fatal occurrence. Trop Doct. 49:52–53. 2019.PubMed/NCBI View Article : Google Scholar

117 

Pazhaniyandi S, Lenin R and Sivathanu S: Hemophagocytic lymphohistiocytosis with a leukemoid reaction in an infant with scrub typhus. J Infect Public Health. 8:626–629. 2015.PubMed/NCBI View Article : Google Scholar

118 

Otrock ZK, Gonzalez MD and Eby CS: Ehrlichia-induced hemophagocytic lymphohistiocytosis: A case series and review of literature. Blood Cells Mol Dis. 55:191–193. 2015.PubMed/NCBI View Article : Google Scholar

119 

Cheng A, Williams F, Fortenberry J, Preissig C, Salinas S and Kamat P: Use of extracorporeal support in hemophagocytic lymphohistiocytosis secondary to ehrlichiosis. Pediatrics. 138(e20154176)2016.PubMed/NCBI View Article : Google Scholar

120 

Burns S, Saylors R and Mian A: Hemophagocytic lymphohistiocytosis secondary to Ehrlichia chaffeensis infection: A case report. J Pediatr Hematol Oncol. 32:e142–143. 2010.PubMed/NCBI View Article : Google Scholar

121 

Singh NS, Pagano AL, Hays AJ, Kats A, Dahl SM, Warady BA, Beins NT and Yin DE: Ehrlichia-induced hemophagocytic lymphohistiocytosis in a pediatric kidney transplant recipient. Pediatr Transplant. 26(e14134)2022.PubMed/NCBI View Article : Google Scholar

122 

Cabler SS, Hogan PG, Fritz SA, Bednarski JJ and Hunstad DA: Incidence and treatment of hemophagocytic lymphohistiocytosis in hospitalized children with Ehrlichia infection. Pediatr Blood Cancer. 67(e28436)2020.PubMed/NCBI View Article : Google Scholar

123 

Tsuge M, Miyamoto M, Miyawaki R, Kondo Y and Tsukahara H: Hemophagocytic lymphohistiocytosis complicating invasive pneumococcal disease: A pediatric case report. BMC Pediatr. 20(15)2020.PubMed/NCBI View Article : Google Scholar

124 

Poudel A, Lew J, Slayton W and Dharnidharka VR: Bartonella henselae infection inducing hemophagocytic lymphohistiocytosis in a kidney transplant recipient. Pediatr Transplant. 18:E83–E87. 2014.PubMed/NCBI View Article : Google Scholar

125 

Edner J, Rudd E, Zheng C, Dahlander A, Eksborg S, Schneider EM, Edner A and Henter JI: Severe bacteria-associated hemophagocytic lymphohistiocytosis in an extremely premature infant. Acta Paediatr. 96:1703–1706. 2007.PubMed/NCBI View Article : Google Scholar

126 

Karapinar DY, Karadaş N, Yazici P, Polat SH and Karapinar B: Trichosporon asahii, sepsis, and secondary hemophagocytic lymphohistiocytosis in children with hematologic malignancy. Pediatr Hematol Oncol. 31:282–284. 2014.PubMed/NCBI View Article : Google Scholar

127 

Guo F, Kang L and Xu M: A case of pediatric visceral leishmaniasis-related hemophagocytic lymphohistiocytosis diagnosed by mNGS. Int J Infect Dis. 97:27–29. 2020.PubMed/NCBI View Article : Google Scholar

128 

Guo X, Chen N, Wang TY, Zhou YC, Li Q and Gao J: Visceral leishmaniasis associated hemophagocytic lymphohistiocytosis: Report of four childhood cases. Zhonghua Er Ke Za Zhi. 49:550–553. 2011.PubMed/NCBI(In Chinese).

129 

Bouguila J, Chabchoub I, Moncef Y, Mlika A, Saghrouni F, Boughamoura L and Essoussi AS: Treatment of severe hemophagocytic syndrome associated with visceral leishmaniasis. Arch Pediatr. 17:1566–1570. 2010.PubMed/NCBI View Article : Google Scholar : (In French).

130 

Castberg FC, Poulsen A, Petersen BL and Lausen B: Visceral leishmaniasis in two children after vacation in Southern Europe. Ugeskr Laeger. 175:2193–2194. 2013.PubMed/NCBI(In Danish).

131 

Rajagopala S, Dutta U, Chandra KSP, Bhatia P, Varma N and Kochhar R: Visceral leishmaniasis associated hemophagocytic lymphohistiocytosis-case report and systematic review. J Infect. 56:381–388. 2008.PubMed/NCBI View Article : Google Scholar

132 

Ay Y, Yildiz B, Unver H, Karapinar DY and Vardar F: Hemophagocytic lymphohistiocytosis associated with H1N1 virus infection and visceral leishmaniasis in a 4.5-month-old infant. Rev Soc Bras Med Trop. 45:407–409. 2012.PubMed/NCBI

133 

Mottaghipisheh H, Kalantar K, Amanati A, Shokripour M, Shahriari M, Zekavat OR, Zareifar S, Karimi M, Haghpanah S and Bordbar M: Comparison of the clinical features and outcome of children with hemophagocytic lymphohistiocytosis (HLH) secondary to visceral leishmaniasis and primary HLH: A single-center study. BMC Infect Dis. 21(732)2021.PubMed/NCBI View Article : Google Scholar

134 

de Carvalho FHG, Lula JF, Teles LF, Caldeira AP and de Carvalho SFG: Hemophagocytic lymphohistiocytosis secondary to visceral leishmaniasis in an endemic area in the north of Minas Gerais, Brazil. Rev Soc Bras Med Trop. 53(e20190491)2020.PubMed/NCBI View Article : Google Scholar

135 

Shi Q, Huang M, Li X, Zheng X, Wang F, Zou Y, Wang L and Jia J: Clinical and laboratory characteristics of hemophagocytic lymphohistiocytosis induced by Leishmania infantum infection. PLoS Negl Trop Dis. 15(e0009944)2021.PubMed/NCBI View Article : Google Scholar

136 

Srivatsav S, Mahalingam S, Ramineni P and Manya S: Dengue and Plasmodium falciparum coinfection with secondary hemophagocytic lymphohistiocytosis in a 3-year-old boy: A clinical conundrum. J Pediatr Hematol Oncol. 44:e253–e254. 2022.PubMed/NCBI View Article : Google Scholar

137 

Padhi S, Varghese RGB, Ramdas A, Phansalkar MD and Sarangi R: Hemophagocytic lymphohistiocytosis: Critical reappraisal of a potentially under-recognized condition. Front Med. 7:492–498. 2013.PubMed/NCBI View Article : Google Scholar

138 

Lehmberg K, Sprekels B, Nichols KE, Woessmann W, Müller I, Suttorp M, Bernig T, Beutel K, Bode SF, Kentouche K, et al: Malignancy-associated haemophagocytic lymphohistiocytosis in children and adolescents. Br J Haematol. 170:539–549. 2015.PubMed/NCBI View Article : Google Scholar

139 

O'Brien MM, Lee-Kim Y, George TI, McClain KL, Twist CJ and Jeng M: Precursor B-cell acute lymphoblastic leukemia presenting with hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 50:381–383. 2008.PubMed/NCBI View Article : Google Scholar

140 

Celkan T, Berrak S, Kazanci E, Ozyürek E, Unal S, Uçar C, Yilmaz S and Gürgey A: Malignancy-associated hemophagocytic lymphohistiocytosis in pediatric cases: A multicenter study from Turkey. Turk J Pediatr. 51:207–213. 2009.PubMed/NCBI

141 

Strenger V, Merth G, Lackner H, Aberle SW, Kessler HH, Seidel MG, Schwinger W, Sperl D, Sovinz P, Karastaneva A, et al: Malignancy and chemotherapy induced haemophagocytic lymphohistiocytosis in children and adolescents-a single centre experience of 20 years. Ann Hematol. 97:989–998. 2018.PubMed/NCBI View Article : Google Scholar

142 

Chellapandian D, Hines MR, Zhang R, Jeng M, van den Bos C, Santa-María López V, Lehmberg K, Sieni E, Wang Y, Nakano T, et al: A multicenter study of patients with multisystem Langerhans cell histiocytosis who develop secondary hemophagocytic lymphohistiocytosis. Cancer. 125:963–971. 2019.PubMed/NCBI View Article : Google Scholar

143 

Chinn IK, Eckstein OS, Peckham-Gregory EC, Goldberg BR, Forbes LR, Nicholas SK, Mace EM, Vogel TP, Abhyankar HA, Diaz MI, et al: Genetic and mechanistic diversity in pediatric hemophagocytic lymphohistiocytosis. Blood. 132:89–100. 2018.PubMed/NCBI View Article : Google Scholar

144 

Cader FZ, Colmenero I and Mussai F: Hemophagocytic lymphohistiocytosis associated with 2 cases of pediatric lymphocyte-depleted classic hodgkin lymphoma. J Pediatr Hematol Oncol. 41:e341–e345. 2019.PubMed/NCBI View Article : Google Scholar

145 

Huang Z, Jia Y, Zuo Y, Wu J, Lu A and Zhang L: Malignancy-associated hemophagocytic lymphohistiocytosis in children: A 10-year experience of a single pediatric hematology center. Hematology. 25:389–399. 2020.PubMed/NCBI View Article : Google Scholar

146 

Sakamoto K, Osumi T, Yoshimura S, Shimizu S, Kato M, Tomizawa D, Fukuda A, Sakamoto S, Nakano N, Yoshioka T, et al: Living-donor liver transplantation providing an adequate chemotherapy for a pediatric patient with anaplastic large cell lymphoma complicated with liver failure due to the aggravation of biliary hepatopathy by secondary hemophagocytic lymphohistiocytosis. Int J Hematol. 112:900–905. 2020.PubMed/NCBI View Article : Google Scholar

147 

Teshigawara S, Katada Y, Maeda Y, Yoshimura M, Kudo-Tanaka E, Tsuji S, Harada Y, Matsushita M, Ohshima S, Watanabe K, et al: Hemophagocytic lymphohistiocytosis with leukoencephalopathy in a patient with dermatomyositis accompanied with peripheral T-cell lymphoma: A case report. J Med Case Rep. 10(212)2016.PubMed/NCBI View Article : Google Scholar

148 

Sinno MG, Rosen D and Wittler R: Concomitant presentation of hemophagocytic lymphohistiocytosis and posttransplant lymphoproliferative disease-like lymphoma in a mildly immunosuppressed leukemia patient: An unusual association. Pediatr Blood Cancer. 63:1474–1476. 2016.PubMed/NCBI View Article : Google Scholar

149 

McCall CM, Mudali S, Arceci RJ, Small D, Fuller S, Gocke CD, Vuica-Ross M, Burns KH, Borowitz MJ and Duffield AS: Flow cytometric findings in hemophagocytic lymphohistiocytosis. Am J Clin Pathol. 137:786–794. 2012.PubMed/NCBI View Article : Google Scholar

150 

Zheng F, Jia Y, Zhang L and Qin J: Hemophagocytic lymphohistiocytosis secondary to juvenile myelomonocytic leukemia: A case report and review of the literature. J Pediatr Hematol Oncol. 44:e580–e584. 2022.PubMed/NCBI View Article : Google Scholar

151 

Farias MG, Freitas PAC, Spagnol F, Souza MV, Alegretti AP, Riegel M, Taniguchi ANR and Daudt LE: Hemophagocytosis by blasts in a child with acute monocytic leukemia after chemotherapy. Rev Paul Pediatr. 39(e2019290)2021.PubMed/NCBI View Article : Google Scholar : (In Portuguese, English).

152 

Dokmanovic L, Krstovski N, Jankovic S and Janic D: Hemophagocytic lymphohistiocytosis arising in a child with Langerhans cell histiocytosis. Turk J Pediatr. 56:452–457. 2014.PubMed/NCBI

153 

Murphy EP, Mo J and Yoon JM: Secondary hemophagocytic lymphohistiocytosis in a patient with favorable histology wilms tumor. J Pediatr Hematol Oncol. 37:e494–e496. 2015.PubMed/NCBI View Article : Google Scholar

154 

Nascimento FA, Nery J, Trennepohl J and Pianovski MAD: Hemophagocytic lymphohistiocytosis after initiation of chemotherapy for bilateral adrenal neuroblastoma. J Pediatr Hematol Oncol. 38:e13–e15. 2016.PubMed/NCBI View Article : Google Scholar

155 

Tang X, Guo X, Gao J, Sun JJ and Wan Z: Hemophagocytic lymphohistiocytosis in langerhans cell histiocytosis: A case series and literature review. J Pediatr Hematol Oncol. 44:e20–e25. 2022.PubMed/NCBI View Article : Google Scholar

156 

Devecioglu O, Anak S, Atay D, Aktan P, Devecioglu E, Ozalp B and Saribeyoglu E: Pediatric acute lymphoblastic leukemia complicated by secondary hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 53:491–492. 2009.PubMed/NCBI View Article : Google Scholar

157 

Henderson LA and Cron RQ: Macrophage activation syndrome and secondary hemophagocytic lymphohistiocytosis in childhood inflammatory disorders: Diagnosis and management. Paediatr Drugs. 22:29–44. 2020.PubMed/NCBI View Article : Google Scholar

158 

Sandler RD, Tattersall RS, Schoemans H, Greco R, Badoglio M, Labopin M, Alexander T, Kirgizov K, Rovira M, Saif M, 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. 11(524)2020.PubMed/NCBI View Article : Google Scholar

159 

Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, Pequignot E, Gonzalez VE, Chen F, Finklestein J, et al: Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6:664–679. 2016.PubMed/NCBI View Article : Google Scholar

160 

Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, et al: Chimeric antigen receptor T-cell therapy-assessment and management of toxicities. Nat Rev Clin Oncol. 15:47–62. 2018.PubMed/NCBI View Article : Google Scholar

161 

Lichtenstein DA, Schischlik F, Shao L, Steinberg SM, Yates B, Wang HW, Wang Y, Inglefield J, Dulau-Florea A, Ceppi F, et al: Characterization of HLH-like manifestations as a CRS variant in patients receiving CD22 CAR T cells. Blood. 138:2469–2484. 2021.PubMed/NCBI View Article : Google Scholar

162 

Hines MR, Keenan C, Maron Alfaro G, Cheng C, Zhou Y, Sharma A, Hurley C, Nichols KE, Gottschalk S, Triplett BM and Talleur AC: Hemophagocytic lymphohistiocytosis-like toxicity (carHLH) after CD19-specific CAR T-cell therapy. Br J Haematol. 194:701–707. 2021.PubMed/NCBI View Article : Google Scholar

163 

Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al: Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 378:439–448. 2018.PubMed/NCBI View Article : Google Scholar

164 

Xu XJ and Tang YM: Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells. Cancer Lett. 343:172–178. 2014.PubMed/NCBI View Article : Google Scholar

165 

Singh N, Hofmann TJ, Gershenson Z, Levine BL, Grupp SA, Teachey DT and Barrett DM: Monocyte lineage-derived IL-6 does not affect chimeric antigen receptor T-cell function. Cytotherapy. 19:867–880. 2017.PubMed/NCBI View Article : Google Scholar

166 

Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A and Sadelain M: CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 24:731–738. 2018.PubMed/NCBI View Article : Google Scholar

167 

Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, Sanvito F, Ponzoni M, Doglioni C, Cristofori P, 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. 24:739–748. 2018.PubMed/NCBI View Article : Google Scholar

168 

Liu Y, Fang Y, Chen X, Wang Z, Liang X, Zhang T, Liu M, Zhou N, Lv J, Tang K, et al: Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 5(eaax7969)2020.PubMed/NCBI View Article : Google Scholar

169 

Teachey DT, Rheingold SR, Maude SL, Zugmaier G, Barrett DM, Seif AE, Nichols KE, Suppa EK, Kalos M, Berg RA, et al: Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood. 121:5154–5157. 2013.PubMed/NCBI View Article : Google Scholar

170 

Winkler U, Jensen M, Manzke O, Schulz H, Diehl V and Engert A: Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood. 94:2217–2224. 1999.PubMed/NCBI

171 

Asano T, Kogawa K, Morimoto A, Ishida Y, Suzuki N, Ohga S, Kudo K, Ohta S, Wakiguchi H, Tabuchi K, et al: Hemophagocytic lymphohistiocytosis after hematopoietic stem cell transplantation in children: A nationwide survey in Japan. Pediatr Blood Cancer. 59:110–114. 2012.PubMed/NCBI View Article : Google Scholar

172 

Noguchi M and Inagaki J: Hemophagocytic lymphohistiocytosis and graft failure following unrelated umbilical cord blood transplantation in children. J Pediatr Hematol Oncol. 42:e440–e444. 2020.PubMed/NCBI View Article : Google Scholar

173 

Jarchin L, Chu J, Januska M, Merola P and Arnon R: Autoimmune hemolytic anemia: An unusual presentation of hemophagocytic lymphohistiocytosis in a pediatric post-liver transplant patient. Pediatr Transplant. 22(e13281)2018.PubMed/NCBI View Article : Google Scholar

174 

Nakajima K, Hiejima E, Nihira H, Kato K, Honda Y, Izawa K, Kawabata N, Kato I, Ogawa E, Sonoda M, et al: Case report: A case of Epstein-Barr virus-associated acute liver failure requiring hematopoietic cell transplantation after emergent liver transplantation. Front Immunol. 13(825806)2022.PubMed/NCBI View Article : Google Scholar

175 

Jha B, Mohan N, Gajendra S, Sachdev R, Goel S, Sahni T, Raina V and Soin A: Prompt diagnosis and management of Epstein-Barr virus-associated post-transplant lymphoproliferative disorder and hemophagocytosis: A dreaded complication in a post-liver transplant child. Pediatr Transplant. 19:E177–E180. 2015.PubMed/NCBI View Article : Google Scholar

176 

Matsuda K, Toyama K, Toya T, Ikemura M, Nakamura F and Kurokawa M: Reactivation of hemophagocytic lymphohistiocytosis triggered by antithymocyte globulin. Intern Med. 57:583–586. 2018.PubMed/NCBI View Article : Google Scholar

177 

Ali S, AlThubaiti S, Renzi S, Krueger J, Chiang KY, Naqvi A, Schechter T, Punnett A and Ali M: Hemophagocytic lymphohistiocytosis is a sign of poor outcome in pediatric Epstein-Barr virus-associated post-transplant lymphoproliferative disease after allogeneic hematopoietic stem cell transplantation. Pediatr Transplant. 23(e13319)2019.PubMed/NCBI View Article : Google Scholar

178 

Park M, Yun YJ, Woo SI, Lee JW, Chung NG and Cho B: Rotavirus-associated hemophagocytic lymphohistiocytosis (HLH) after hematopoietic stem cell transplantation for familial HLH. Pediatr Int. 57:e77–e80. 2015.PubMed/NCBI View Article : Google Scholar

179 

Salvador C, Meister B, Larcher H, Crazzolara R and Kropshofer G: Hemophagocytic lymphohistiocytosis after allogeneic bone marrow transplantation during chronic norovirus infection. Hematol Oncol. 32:102–106. 2014.PubMed/NCBI View Article : Google Scholar

180 

Hattori N, Sato M, Uesugi Y, Nakata A, Sasaki Y, Shimada S, Watanuki M, Fujiwara S, Kawaguchi Y, Arai N, et al: Characteristics and predictors of post-transplant-associated hemophagocytic lymphohistiocytosis in adults. Int J Hematol. 113:693–702. 2021.PubMed/NCBI View Article : Google Scholar

181 

Stefanou C, Tzortzi C, Georgiou F and Timiliotou C: Combining an antiviral with rituximab in EBV-related haemophagocytic lymphohistiocytosis led to rapid viral clearance; and a comprehensive review. BMJ Case Rep. 2016(bcr2016216488)2016.PubMed/NCBI View Article : Google Scholar

182 

Farias-Moeller R, LaFrance-Corey R, Bartolini L, Wells EM, Baker M, Doslea A, Suslovic W, Greenberg J, Carpenter JL and Howe CL: Fueling the FIRES: Hemophagocytic lymphohistiocytosis in febrile infection-related epilepsy syndrome. Epilepsia. 59:1753–1763. 2018.PubMed/NCBI View Article : Google Scholar

183 

Dandoy C and Grimley M: Secondary hemophagocytic lymphohistiocytosis (HLH) from a presumed brown recluse spider bite. J Clin Immunol. 34:544–547. 2014.PubMed/NCBI View Article : Google Scholar

184 

Alabbas F, Elyamany G, Alanzi T, Ali TB, Albatniji F and Alfaraidi H: Wolman's disease presenting with secondary hemophagocytic lymphohistiocytosis: A case report from Saudi Arabia and literature review. BMC Pediatr. 21(72)2021.PubMed/NCBI View Article : Google Scholar

185 

Schüller S, Attarbaschi A, Berger A, Hutter C, Klebermass-Schrehof K and Steiner M: Hemophagocytic lymphohistiocytosis triggered by Gaucher disease in a preterm neonate. Pediatr Hematol Oncol. 33:462–467. 2016.PubMed/NCBI View Article : Google Scholar

186 

Gokce M, Unal O, Hismi B, Gumruk F, Coskun T, Balta G, Unal S, Cetin M, Kalkanoglu-Sivri HS, Dursun A and Tokatlı A: Secondary hemophagocytosis in 3 patients with organic acidemia involving propionate metabolism. Pediatr Hematol Oncol. 29:92–98. 2012.PubMed/NCBI View Article : Google Scholar

187 

Erdol S, Ture M, Baytan B, Yakut T and Saglam H: An unusual case of LCHAD deficiency presenting with a clinical picture of hemophagocytic lymphohistiocytosis: Secondary HLH or coincidence? J Pediatr Hematol Oncol. 38:661–662. 2016.PubMed/NCBI View Article : Google Scholar

188 

Düzenli Kar Y, Özdemir ZC, Kiral E, Kiliç Yildirim G, Dinleyici EÇ and Bör Ö: Hemophagocytic lymphohystiocytosis associated with type Ia glycogen storage disease. J Pediatr Hematol Oncol. 41:e260–e262. 2019.PubMed/NCBI View Article : Google Scholar

189 

Santos Silva E, Klaudel-Dreszler M, Bakuła A, Oliva T, Sousa T, Fernandes PC, Tylki-Szymańska A, Kamenets E, Martins E and Socha P: Early onset lysosomal acid lipase deficiency presenting as secondary hemophagocytic lymphohistiocytosis: Two infants treated with sebelipase alfa. Clin Res Hepatol Gastroenterol. 42:e77–e82. 2018.PubMed/NCBI View Article : Google Scholar

190 

Faraguna MC, Musto F, Crescitelli V, Iascone M, Spaccini L, Tonduti D, Fedeli T, Kullmann G, Canonico F, Cattoni A, et al: Mucopolysaccharidosis-plus syndrome, a rapidly progressive disease: Favorable impact of a very prolonged steroid treatment on the clinical course in a child. Genes (Basel). 13(442)2022.PubMed/NCBI View Article : Google Scholar

191 

Merli P, Quintarelli C, Strocchio L and Locatelli F: The role of interferon-gamma and its signaling pathway in pediatric hematological disorders. Pediatr Blood Cancer. 68(e28900)2021.PubMed/NCBI View Article : Google Scholar

192 

George MR: Hemophagocytic lymphohistiocytosis: Review of etiologies and management. J Blood Med. 5:69–86. 2014.PubMed/NCBI View Article : Google Scholar

193 

Henter JI, Samuelsson-Horne A, Aricò M, Egeler RM, Elinder G, Filipovich AH, Gadner H, Imashuku S, Komp D, Ladisch S, et al: Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood. 100:2367–2373. 2002.PubMed/NCBI View Article : Google Scholar

194 

Jordan M, Prof FL, Allen C, De Benedetti F, Grom AA, Ballabio M, Ferlin WG and De Min C: A novel targeted approach to the treatment of hemophagocytic lymphohistiocytosis (HLH) with an anti-interferon gamma (IFNγ) monoclonal antibody (mAb), NI-0501: First results from a pilot phase 2 study in children with primary HLH. Blood. 126:LBA–3. 2015.

195 

Locatelli F, Jordan MB, Allen C, Cesaro S, Rizzari C, Rao A, Degar B, Garrington TP, Sevilla J, Putti MC, et al: Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 382:1811–1822. 2020.PubMed/NCBI View Article : Google Scholar

196 

Marsh RA, Allen CE, McClain KL, Weinstein JL, Kanter J, Skiles J, Lee ND, Khan SP, Lawrence J, Mo JQ, et al: Salvage therapy of refractory hemophagocytic lymphohistiocytosis with alemtuzumab. Pediatr Blood Cancer. 60:101–109. 2013.PubMed/NCBI View Article : Google Scholar

197 

Das R, Guan P, Sprague L, Verbist K, Tedrick P, An QA, Cheng C, Kurachi M, Levine R, Wherry EJ, et al: Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood. 127:1666–1675. 2016.PubMed/NCBI View Article : Google Scholar

198 

De Benedetti F, Brunner HI, Ruperto N, Kenwright A, Wright S, Calvo I, Cuttica R, Ravelli A, Schneider R, Woo P, et al: Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med. 367:2385–2395. 2012.PubMed/NCBI View Article : Google Scholar

199 

Huang P, Huang C, Xu H, Lu J, Tian R, Wang Z and Chen Y: Early use of blood purification in severe Epstein-Barr virus-associated hemophagocytic syndrome. Pediatrics. 145(e20193197)2020.PubMed/NCBI View Article : Google Scholar

200 

Bottari G, Murciano M, Merli P, Bracaglia C, Guzzo I, Stoppa F, Pardeo M, Nunziata J, Bufalo FD, Genuini L, et al: Hemoperfusion with cytosorb to manage multiorgan dysfunction in the spectrum of hemophagocytic lymphohistiocytosis syndrome in critically Ill children. Blood Purif. 51:417–424. 2022.PubMed/NCBI View Article : Google Scholar

201 

Matsumoto Y, Naniwa D, Banno S and Sugiura Y: The efficacy of therapeutic plasmapheresis for the treatment of fatal hemophagocytic syndrome: Two case reports. Ther Apher. 2:300–304. 1998.PubMed/NCBI View Article : Google Scholar

202 

Demirkol D, Yildizdas D, Bayrakci B, Karapinar B, Kendirli T, Koroglu TF, Dursun O, Erkek N, Gedik H, Citak A, et al: Hyperferritinemia in the critically ill child with secondary hemophagocytic lymphohistiocytosis/sepsis/multiple organ dysfunction syndrome/macrophage activation syndrome: What is the treatment? Crit Care. 16(R52)2012.PubMed/NCBI View Article : Google Scholar

203 

La Rosée P, Horne A, Hines M, von Bahr Greenwood T, Machowicz R, Berliner N, Birndt S, Gil-Herrera J, Girschikofsky M, Jordan MB, et al: Recommendations for the management of hemophagocytic lymphohistiocytosis in adults. Blood. 133:2465–2477. 2019.PubMed/NCBI View Article : Google Scholar

204 

Yan WL, Yang SL, Zhao FY and Xu XJ: Ruxolitinib is an alternative to etoposide for patient with hemophagocytic lymphohistiocytosis complicated by acute renal injury: A case report. J Oncol Pharm Pract. 28:222–227. 2022.PubMed/NCBI View Article : Google Scholar

205 

Broglie L, Pommert L, Rao S, Thakar M, Phelan R, Margolis D and Talano J: Ruxolitinib for treatment of refractory hemophagocytic lymphohistiocytosis. Blood Adv. 1:1533–1536. 2017.PubMed/NCBI View Article : Google Scholar

206 

Cançado GGL, Freitas GG, Faria FHF, de Macedo AV and Nobre V: Hemophagocytic lymphohistiocytosis associated with visceral leishmaniasis in late adulthood. Am J Trop Med Hyg. 88:575–577. 2013.PubMed/NCBI View Article : Google Scholar

207 

Imashuku S, Kuriyama K, Teramura T, Ishii E, Kinugawa N, Kato M, Sako M and Hibi S: Requirement for etoposide in the treatment of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. J Clin Oncol. 19:2665–2673. 2001.PubMed/NCBI View Article : Google Scholar

208 

Huang Z and Xie J: Ruxolitinib in conjunction with the HLH-94 protocol for Epstein-Barr virus-related hemophagocytic lymphohistiocytosis in the intensive care unit: A case report. Medicine (Baltimore). 100(e25188)2021.PubMed/NCBI View Article : Google Scholar

209 

Zhao Y, Li Z, Zhang L, Lian H, Ma H, Wang D, Zhao X, Zhang Q, Wang T and Zhang R: L-DEP regimen salvage therapy for paediatric patients with refractory Epstein-Barr virus-associated haemophagocytic lymphohistiocytosis. Br J Haematol. 191:453–459. 2020.PubMed/NCBI View Article : Google Scholar

210 

Shiraishi A, Ohga S, Doi T, Ishimura M, Takimoto T, Takada H, Miyamoto T, Abe Y and Hara T: Treatment choice of immunotherapy or further chemotherapy for Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 59:265–270. 2012.PubMed/NCBI View Article : Google Scholar

211 

Lackner H, Seidel MG, Strenger V, Sovinz P, Schwinger W, Benesch M, Sperl D and Urban C: Hemophagocytic syndrome in children with acute monoblastic leukemia-another cause of fever of unknown origin. Support Care Cancer. 21:3519–3523. 2013.PubMed/NCBI View Article : Google Scholar

212 

Chi MS, Chang YC and Chi KH: In reply to roos et al. Int J Radiat Oncol Biol Phys. 101(1003)2018.

213 

Han L, Li L, Wu J, Li X, Zhang L, Wang X, Fu X, Ma W, Sun Z, Zhang X, et al: Clinical features and treatment of natural killer/T cell lymphoma associated with hemophagocytic syndrome: comparison with other T cell lymphoma associated with hemophagocytic syndrome. Leuk Lymphoma. 55:2048–2055. 2014.PubMed/NCBI View Article : Google Scholar

214 

Eloseily EM, Weiser P, Crayne CB, Haines H, Mannion ML, Stoll ML, Beukelman T, Atkinson TP and Cron RQ: Benefit of anakinra in treating pediatric secondary hemophagocytic lymphohistiocytosis. Arthritis Rheumatol. 72:326–334. 2020.PubMed/NCBI View Article : Google Scholar

215 

Bami S, Vagrecha A, Soberman D, Badawi M, Cannone D, Lipton JM, Cron RQ and Levy CF: The use of anakinra in the treatment of secondary hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 67(e28581)2020.PubMed/NCBI View Article : Google Scholar

216 

Bagri NK, Gupta L, Sen ES and Ramanan AV: Macrophage activation syndrome in children: Diagnosis and management. Indian Pediatr. 58:1155–1161. 2021.PubMed/NCBI View Article : Google Scholar

217 

Ravelli A, Minoia F, Davì S, Horne A, Bovis F, Pistorio A, Aricò M, Avcin T, Behrens EM, De Benedetti F, et al: 2016 Classification criteria for macrophage activation syndrome complicating systemic juvenile idiopathic arthritis: A European league against rheumatism/American college of rheumatology/paediatric rheumatology international trials organisation collaborative initiative. Arthritis Rheumatol. 68:566–576. 2016.PubMed/NCBI View Article : Google Scholar

218 

Shalabi H, Gust J, Taraseviciute A, Wolters PL, Leahy AB, Sandi C, Laetsch TW, Wiener L, Gardner RA, Nussenblatt V, et al: Beyond the storm-subacute toxicities and late effects in children receiving CAR T cells. Nat Rev Clin Oncol. 18:363–378. 2021.PubMed/NCBI View Article : Google Scholar

219 

Ishida H, Yoshida H, Yoshihara T, Ito M and Morimoto A: Origin of macrophages involved in the development of allogeneic hematopoietic stem cell transplantation-associated hemophagocytic syndrome: Observations on a patient with juvenile myelomonocytic leukemia. Bone Marrow Transplant. 40:701–703. 2007.PubMed/NCBI View Article : Google Scholar

220 

Ostronoff M, Ostronoff F, Coutinho M, Calixto R, Souto Maior AP, Sucupira A, Florencio R and Tagliari C: Hemophagocytic syndrome after autologous peripheral blood stem cell transplantation for multiple myeloma; successful treatment with high-dose intravenous immunoglobulin. Bone Marrow Transplant. 37:797–798. 2006.PubMed/NCBI View Article : Google Scholar

221 

Koyama M, Sawada A, Yasui M, Inoue M and Kawa K: Encouraging results of low-dose etoposide in the treatment of early-onset hemophagocytic syndrome following allogeneic hematopoietic stem cell transplantation. Int J Hematol. 86:466–467. 2007.PubMed/NCBI View Article : Google Scholar

222 

Ono R, Ashiarai M, Hirabayashi S, Mizuki K, Hosoya Y, Yoshihara H, Ohtake J, Mori S, Manabe A and Hasegawa D: Ruxolitinib for hematopoietic cell transplantation-associated hemophagocytic lymphohistiocytosis. Int J Hematol. 113:297–301. 2021.PubMed/NCBI View Article : Google Scholar

223 

Janka GE: Familial hemophagocytic lymphohistiocytosis. Eur J Pediatr. 140:221–230. 1983.PubMed/NCBI View Article : Google Scholar

224 

Messina C, Zecca M, Fagioli F, Rovelli A, Giardino S, Merli P, Porta F, Aricò M, Sieni E, Basso G, et al: Outcomes of children with hemophagocytic lymphohistiocytosis given allogeneic hematopoietic stem cell transplantation in italy. Biol Blood Marrow Transplant. 24:1223–1231. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2023
Volume 26 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Benevenuta C, Mussinatto I, Orsi C and Timeus FS: Secondary hemophagocytic lymphohistiocytosis in children (Review). Exp Ther Med 26: 423, 2023
APA
Benevenuta, C., Mussinatto, I., Orsi, C., & Timeus, F.S. (2023). Secondary hemophagocytic lymphohistiocytosis in children (Review). Experimental and Therapeutic Medicine, 26, 423. https://doi.org/10.3892/etm.2023.12122
MLA
Benevenuta, C., Mussinatto, I., Orsi, C., Timeus, F. S."Secondary hemophagocytic lymphohistiocytosis in children (Review)". Experimental and Therapeutic Medicine 26.3 (2023): 423.
Chicago
Benevenuta, C., Mussinatto, I., Orsi, C., Timeus, F. S."Secondary hemophagocytic lymphohistiocytosis in children (Review)". Experimental and Therapeutic Medicine 26, no. 3 (2023): 423. https://doi.org/10.3892/etm.2023.12122