search for




 

Review Article
A Recent Update on Histiocytic Disorder in Children: Focus on Diagnosis and Treatment
Clin Pediatr Hematol Oncol 2020;27:32-42.
Published online April 30, 2020
© 2020 Korean Society of Pediatric Hematology-Oncology

Hoi Soo Yoon

Department of Pediatrics, Kyung Hee University College of Medicine, Seoul, Korea
Correspondence to: Hoi Soo Yoon
Department of Pediatrics, Kyung Hee University College of Medicine, 23 Kyungheedae-ro, Dongdaemungu, Seoul 02447, Korea
Tel: +82-2-958-8206
Fax: +82-2-958-8304
E-mail: snoopyi@hanmail.net
ORCID ID: orcid.org/0000-0003-1688-3226
Received April 6, 2020; Revised April 13, 2020; Accepted April 21, 2020.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
The histiocytosis is rare disorder characterized by the accumulation of macrophages, dendritic cells, or monocyte-derived cells in various tissues and organs of children and adults. Classifying histiocytic disorders is difficult and has changed over time as an understanding of the biology of these cells has evolved. The most recently revised 2016 WHO classification of histiocytosis and neoplasms of the macrophage-dendritic cell lineages has proposed grouping this diverse group of over 100 clinical entities into five main groups based on clinical, histologic, and molecular relevance. Comprehensive genomic studies for histiocytosis have been described and our understanding of the pathogenesis and biology has increased over the past decade. These advances will be able to make precision medicine and targeted therapy possible in patients with histiocytosis. Among the histiocytosis, this review mainly focuses on the updated diagnosis and treatment of Langerhans cell histiocytosis (LCH) and hemophagocytic lymphohistiocytosis (HLH) in children.
Keywords: Histiocytic disorder, Langerhans cell histiocytosis, Hemophagocytic lymphohistiocytosis, Children
Introduction

Histiocytosis encompasses a group of diverse proliferative disorders characterized by the accumulation and infiltration of variable numbers of monocytes, macrophages, and dendritic cells in the affected tissues of children and adults. Their clinical behavior ranges from mild to disseminated and, sometimes, life-threatening forms [1-3]. The first classification of histiocytosis, published in 1987 by the Working Group of the Histiocyte Society (HS), consisted of 3 categories: Langerhans cell (LC) or non-LC-related, and malignant histiocytosis (MH) [1]. In 2010, a major breakthrough came with the discovery of recurrent BRAFV600E mutations in Langerhans cell histiocytosis (LCH); this finding paved the way for a revised classification of histiocytic disorders [4]. Histiocytosis is now categorized into five groups with an emphasis on targetable mutations of the mitogen-activated protein kinase (MAPK) pathway: 1) Langerhans-related (L group), 2) cutaneous and mucocutaneous (C group), 3) Rosai-Dorfman disease (R group), 4) malignant histiocytosis (M group), and 5) hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) (H group) (Table 1) [5]. Considerable advances in the understanding of their genetics have led to increased clinical recognition of these conditions and our understanding of these diseases has now evolved from the concept of a primary inflammatory condition to that of a clonal neoplastic disease [6]. This understanding has led to the development of effective mechanism-based therapeutic strategies for patients with histiocytic diseases. This article focuses on the updated diagnosis and treatment for LCH and HLH in children.

Table 1 . Revised classification of histiocytosis and neoplasms of the macrophage-dendritic cell lineage.

GroupDisease
L GroupLangerhans cell histiocytosis (LCH)
Indeterminate cell histiocytosis (ICH)
Erdheim-Chester disease (ECD)
Mixed LCH/ECD
C GroupCutaneous non-LCH
- XG family: JXG, AXG, SRH, BCH, GEH, PNH
- N-XG family: cutaneous RDD, NXG, other NOS
Cutaneous non-LCH with major systemic component
R groupFamilial Rosai-Dorfman Disease (RDD)
Sporadic RDD
- Classical RDD
- Extra-nodal RDD
- RDD with neoplasia or immune disease
- Unclassified
M groupPrimary malignant histiocytosis
Secondary malignant histiocytosis
H groupPrimary hemophagocytic lymphohistiocytosis (HLH)
Secondary HLH
HLH of unknown/uncertain origin

AXG, adult xanthogranuloma; BCH, benign cephalic histiocytosis; GEH, generalized eruptive histiocytosis; JXG, juvenile xanthogranuloma; NXG, necrobiotic xanthogranuloma; PNH, progressive nodular histiocytosis; SRH, solitary reticulohistiocytoma. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages [5]..


Langerhans Cell Histiocytosis (LCH)

For several decades, LCH has been considered to be a reactive clonal proliferation of LCs. However, an ongoing debate over the grouping of LCH was finally settled in favor of neoplasm after the discovery of the BRAFV600E mutation in 2010 [4]. Almost any organ can be affected, and the clinical presentation reflects the tissue-specific inflammatory phenomenon. A definitive diagnosis is made by the combination of clinical presentation, histology, and immunohistochemistry.

1) Pathology and biology

A diagnosis of LCH is made by typical positive staining with CD1a or CD207 [7]. With the development of new technology for accurate detection of cell-free DNA, BRAFV600E mutation analysis has shown to be an effective tool for diagnosis and monitoring of disease activity in patients with LCH [8]. The pathogenic cells are known to originate from a myeloid-derived precursor and are uniformly characterized by activation of the MAPK/ERK (extracellular-signal-regulated kinase) signaling pathway [9,10]. In up to two-thirds of cases, mutations in MAP2K1 or less frequently in other members of the pathway, such as ARAF (A-Raf proto-oncogene, serine/threonine kinase), have been described. About one-quarter of patients have no known genomic mutations.

2) Clinical presentation

The clinical manifestations of LCH depend on the organ involved and the extent of involvement. Bone is the most commonly involved organ; bony involvement is present in 80% of cases, and a painful bony lesion is the most common presentation. Skin is the second most frequently involved organ; the presentation may be a rash (generalized, papular, ulcerative, or vesicular) and/or seborrheic involvement of the scalp. Features of weight loss, diarrhea, edema, dyspnea, jaundice (conjugated hyperbilirubinemia), cytopenias, hepatosplenomegaly, lymphadenopathy, polydipsia, and polyuria indicate specific organ involvement. LCH of the central nervous system (CNS) can present as a neurodegenerative disease (ND-LCH) and/or intracranial tumorous lesions. Patients with ND-LCH may develop clinical symptoms of dysarthria, ataxia, dysmetria, and behavior changes. Due to its heterogenous presentation, from self-limited to disseminated disease, LCH has been clinically classified according to the number of lesions and sites of involvement coupled with the number of risk organs affected [10]. At present, patients of LCH are risk-stratified according to the extent of disease [single system versus (vs.) multisystem] and risk organ (RO) involvement (presence or absence of involvement of the liver, spleen, or bone marrow), in which “risk” refers to higher risk of mortality [11].

3) Treatment

(1) Single-system LCH

① Skin-limited LCH: In most cases, isolated LCH skin lesion regress spontaneously and warrant only observation. Infants, however, require close observation because a large proportion of them are likely to progress to high-risk, disseminated disease [12]. Patients who have symptomatic/refractory skin lesion have been treated with topical steroids, tacrolimus, nitrogen mustard, thalidominde, psoralen and ultraviolet A (PUVA) therapy, surgical excision, oral corticosteroids, or minimal systemic chemotherapy [11,12].

② Single bone lesion: Single bone LCH usually resolves with curettage and/or intralesional corticosteroid injections [13]. Patients with single bone lesion have only a 10% chance of reactivation [14]. Radiation therapy may be effective in older children with single vertebral lesions that have not caused complete collapse of the vertebra or with a lesion in the greater trochanter of the femur at risk for pathologic fracture [15]. A few case reports showed that indomethacin was a useful therapy for LCH involving the bony skeleton and may have a role as first-line treatment in single system bone disease [16,17].

(2) Multisystem LCH

① Multiple bone lesions or bone and other low-risk site: The standard recommended therapy for low-risk multisystem LCH, which is based on the LCH-III trial findings, is 12 months of therapy with vinblastine/prednisone; this has reduced the reactivation rate from the historical 50% to 30% [18]. The LCH-IV trial is currently ongoing with regards to whether further prolongation of this mild treatment may further reduce reactivation. In stratum II, the response to a uniform initial second line therapy (prednisolone, cytarabine, and vincristine) for those patients without risk organ involvement is studied following a randomized comparison of maintenance therapy with either indomethacin or mercaptopurine and methotrexate for studying the inhibitory effect of indomethacin on reactivation. Some reports described that bisphosphonates can significantly improve bone pain and induce remission in active bone LCH and may be an effective treatment for reactivated LCH with bone lesions [19-21].

(3) High-risk LCH

The current standard therapy for high-risk LCH consists of treatment with vinblastine, prednisone, and mercaptopurine for 1 year based on the LCH-III trial [18]. Prolongation of the treatment duration from 6 months in LCH-II to 12 months in LCH-III was beneficial. But, adding etoposide (LCH-II) and methotrexate (LCH-III) did not improve the response or relapse-free survival of high-risk patients [22,23].

(4) CNS lesion

Diabetes insipidus (DI) is the most frequent initial sign of LCH in the CNS [24]. Due to risks of pituitary biopsy, it is reasonable to initiate LCH therapy empirically in such patients and monitor for early response by brain magnetic resonance imaging (MRI). Mass lesions of the brain may respond to vinblastine/prednisone, cladribine, cytarabine, and clofarabine [25,26].

① CNS-risk lesions: The current standard of care for patients with so-called CNS-risk lesions such as the mastoid, sphenoid, orbit, clivus, or temporal bone is 12 months of therapy with vinblastine and corticosteroids [18]. These patients have increased risk of developing DI and/or ND-LCH. The LCH-III study, in which a year of vinblastine/prednisone was used, showed a further reduction of DI incidence to 12%, when compared with surgery alone [26].

② ND-LCH: ND-LCH is a syndrome of progressive, devastating neurodegeneration of unknown etiology.

A brain MRI demonstrates hyperintensity of the dentate nucleus and white matter of the cerebellum on FLAIR and T2-weighted images, or hyperintense lesions of the basal ganglia on T1-weighted images [27]. Intravenous g-globulin (IVIG) and retinoic acid have been reported to stabilize progression of ND-LCH [28,29]. Some studies reported that vincristine/cytarabine was associated with improvement in clinical symptoms and MRI images [30]. In LCH-IV study, additionally the effectiveness of 2-CdA in tumorous CNS-LCH and of IVIG and intravenous cytarabine (Ara-C) in ND-LCH will be prospectively studied.

③ Salvage therapy: In LCH-III study, the probability of a reactivation was 54% in patients without RO involvement who treated with 6 months, but, 37% in those who treated with 12 months [18,23]. Many studies reported that reactivation of patients with LCH tends to occur in the first 2 years [18,31]. Salvage therapy for LCH usually includes agents active against the myeloid cells, such as cytarabine, cladribine, and clofarabine [32-34]. Hematopoietic stem cell transplantation (HSCT) can be another option. However, most data for salvage therapy has small case series or retrospective studies [35,36] (Table 2).

Table 2 . Salvage therapy for relapsed/refractory Langerhans cell histiocytosis.

RegimenStudy (n)ResponseSRRemark
2-CdA (5 mg/m2×5 d) [33]ProspectiveRO+ 22% (RR)2-yr OS2-CdA for RO-multisystem, multifocal bone is effective, RR good Age >2 yr at 2-CdA
LCH-S-2005RO− 62% (RR)RO+ 48%
RO+ (n=46)RO− 97%
RO− (n=37)
2-CdA/Ara-C (9 mg/m2, 1g/m2/d×5 d) [34]ProspectiveRR 92%5yr-OS 85%2-CdA/Ara-C is effective for RO+ multisystem LCH
LCH-S-2005No active: n=2, Better: 23,High toxicity (grade 4 hematologic toxicity, severe infection)
(n=27, RO+)Stable: 2
Clofarabine (25 mg/m2/d×5 d) [35]RetrospectiveCR (61%)1-yr PFS 76%All patients developed grade 4 neutropenia
LCH (n=11),PR (22%)1-yr OS 91%
RO+ (n=3)
RO− (n=8)
HSCT
MAC vs. RIC (CIBMTR & EBMT) [36]Retrospective (after 2000)Relapse rate3-yr OSOS, PFS of MAC and RIC similar.
MAC (n=41) vs. RIC (n=26)MAC (8%) vs. RIC (28%)MAC 77%Relapse rate after RIC marginally higher MAC
RIC 71%
MAC vs. RIC (Japan) [37]RetrospectiveOS
MAC (n=11) vs. RIC (n=19)MAC 63.6%
RIC 56.8%
FFS
MAC 54.6%
RIC 56.8%

CIBMTR, center for international blood and marrow transplant research; CR, complete response; EBMT, european blood and marrow transplant; MAC, myeloablative conditioning; OS, overall survival; PFS, progression free survival; PR, partial response; RIC, reduced intensity conditioning; RR, response rate; RO, risk organ..


(5) Targeted therapy

With the identification of BRAFV600E and other mutations in the MAPK pathway in LCH, early phase trials were begun to find the role of targeted therapy in the clinical field.

BRAF Inhibition: A phase 2 trial in BRAF-mutant ECD/LCH showed disease regression in 86% patients, with no one progressing during Vemurafenib (VMF) therapy [37]. In an international observation study, VMF seemed safe and effective in children with refractory BRAFV600E positive LCH. However, the majority of patients who discontinued VMF experienced LCH reactivation [38]. Moreover, despite the small size of the study, a very high risk of secondary skin tumors (approximately 30%) has been observed in adults with melanoma [39]. A trial of dabrafenib in a pediatric LCH is ongoing, with early patients showing an encouraging response (NCT01677741).

MEK inhibition: The efficacy of MEK inhibitors trametinib and cobimetinib was first reported in non-LCH histiocytosis [40]. An ongoing trial of cobimetinib in adult patients with histiocytic disorders has shown response [41]. In BRAFV600E-mutant melanoma, dual blockade of the MAPK pathway using a BRAF plus MEK inhibitor results in higher efficacy and less toxicity compared with monotherapy with a BRAF inhibitor [42,43]. The efficacy of combined BRAF-MEK inhibition has not been fully explored in BRAF-mutant histiocytosis.

4) Future directions

The ongoing LCH-IV trial has broad goals that aim to determine optimal therapy for all patients, as stratified: high-risk multisystem, low-risk multisystem, low-risk single system, and LCH with special site involvement, particularly focusing on the difficult problem of diffuse CNS disease. The randomized studies in the LCH-IV protocol strive to optimize the outcomes of first line treatments by testing prolonging (12 vs. 24 months) and intensifying (6-mercaptopurine) treatment of high risk patients, and by comparing treatment duration (6 vs. 12-months) for single system disease. The protocol is also testing a randomized study of new combinations as second-line treatment for those individuals with low-risk disease either reactivating or not initially responding. Targeted therapy must be considered not as a replacement but as additional tool that should be used judiciously. The choice of the optimal timing of targeted therapy (as salvage, up-front, or maintenance) and whether it should be treated alone or combination with chemotherapy needs to be studied.

Hemophagocytic Lymphohistiocytosis (HLH)

HLH is a syndrome describing patients with severe systemic hyperinflammation that it is characterized by unremitting fever, cytopenias, hepatosplenomegaly, coagulopathy, and elevations in typical biomarkers including ferritin and soluble interleukin-2 receptor (sIL-2R). The high mortality rate makes prompt recognition and treatment of this hyperinflammatory syndrome essential [44]. HLH is classified into primary and secondary forms. Primary HLH is a hereditary immune disorder, whereas secondary HLH develops as a complication in various conditions such as infection, malignancy, autoimmune disease, and post-HSCT [45,46].

1) Pathogenesis

Patients with active HLH have markedly elevated serum inflammatory cytokines, namely, interferon (IFN)‐g, tumor necrosis factor (TNF)‐α, IL‐1β, IL‐2, IL‐6, IL‐12, IL‐16, and IL‐18 [43]. This cytokine storm arises from the excessive secretion of cytokines by uncontrolled activated cytotoxic T lymphocyte (CTL) and natural killer (NK) cells that in turn hyperactivate macrophages. Of the various cytokines that are elevated in HLH, IFN‐g plays a particularly key role in the development of HLH [47,48].

2) Diagnosis

The HS established a set of clinical and laboratory criteria to help the diagnosis of the syndrome of HLH for its HLH-94 and 2004 clinical trials and this has been modified partly in 2009 [40-42]. However, the diversity of the clinical presentation of HLH has led to confusion. Patients presenting in early disease may not yet show 5 of 8 criteria, and some patients may never meet this criteria, including those with atypical presentations such as isolated CNS disease or acute liver failure [49-52]. Fever above 38.3°C is nearly universal in untreated HLH. While splenomegaly and hepatomegaly are very common in HLH, adenopathy is not [45]. Cytopenias are ubiquitous in HLH. Lack of cytopenias should make one doubt a diagnosis of HLH, except in the special case of isolated, CNS-only disease [53]. Some laboratory tests are used to help in the diagnosis of HLH and are also useful biomarkers for disease activity monitoring. Most patients have much higher levels of ferritin than the threshold suggests (>500 μg/L); however, serum ferritin is also driven by iron overload states and can be elevated in many inflammatory contexts. A serum ferritin level greater than 500 μg/L is over 90% sensitive, but its specificity is only robust at levels greater than 2,000-10,000 μg/L, and in adults, levels greater than 10,000 μg/L are still most commonly associated with malignancy [54,55]. Recently, higher levels of ferritin (e.g.,>3,000 μg/L) have been suggested [56]. One study compared 123 patients with HLH to 320 patients with other hyperferritinemic conditions. At 2,000 μg/L, a trade-off was reached with sensitivity at 70% and specificity at 68% for HLH in that study [57]. Elevated sIL-2R should always be observed in untreated HLH since T-cell activation in central to HLH pathogenesis [58,59]. However, an extremely elevated sIL-2R (>10 to 20-fold above normal) in noninfantile patients suggests undiagnosed lymphoma, especially when ferritin is not similarly elevated [60,61]. Very low or absent NK cell function can indicate a genetic HLH disease, but, other acute illness and various treatments can also temporarily impair NK numbers and function, and a low result has been found to have poor specificity (43%) [62, 63]. A recent study reported that perforin and CD107a tests are more sensitive and no less specific compared with NK cytotoxicity testing for screening for genetic HLH and suggested they be considered for addition to current HLH criteria [62]. New laboratory options such as IL-18 levels, which reflect inflammasome activation, or CXCL9, which indicates pathway activity, and IFN‐g are being more frequently used. A total IL-18 level greater than 24,000 pg/mL distinguished MAS from primary HLH with 83% sensitivity and 94% specificity, and the ratio of IL-18 to CXCL9 has been used to differentiate patients with rheumatologic disease and MAS from patients with HLH. IL-18 binding protein (IL-18BP) was found to be more elevated in FHL and malignancy-associated HLH [64]. Work-up of some other cytokines such as IL-10 and IFN‐g can be helpful in the differential diagnosis of HLH from sepsis [65,66]. The sensitivity and specificity of these biomarkers in the diagnostic criteria should be elucidated and updated through evidence-based international consensus and further study. Fig. 1 displays an updated diagnostic algorithm for consideration of HLH in suspected patients.

Figure 1. Algorithm for diagnostic work-up of HLH.

3) Treatment

Immediate treatment of HLH is generally warranted once a diagnosis is made, while it is important to rule out HLH disease mimics or malignancies before starting therapy in order to avoid inappropriate treatment and/or obscuring the underlying diagnosis [45]. The mainstays of HLH treatment consist of immunosuppressive, chemotherapeutic agents and biologics that aim to control the cytokine storm and eliminate activated T-cells and macrophages [45].

(1) Chemoimmunotherapy

Currently, standard therapy for HLH consists of dexamethasone and etoposide based on the experience of the HS HLH-94 and HLH-2004 studies [67,68]. The 5-year overall survival (OS) rate in children with (n=168) and without (n=201) family history/genetically verified primary HLH was 59% and 64%, respectively [68]. The HS recently published formal recommendations for the use of etoposide-based treatment, and CSA upfront therapy in HLH-2004 is no longer recommended due to toxicity, and the HLH-94 is the standard of care [69]. Though aggressive treatment is needed for most patients, initial therapy with dexamethasone alone with close monitoring may be appropriate before starting etoposide in patients who are not infants and not severely ill [53]. Patients with CNS involvement receive additional intrathecal treatment with methotrexate and steroid, and need treatment of seizures or therapies for specific neurologic deficits [68]. Treatment of HLH should be accompanied by appropriate therapy of the identified underlying trigger. Rituximab containing chemoimmunotherapy can be helpful in the treatment of EBV-HLH [70-72]. IVIG, therapeutic plasma exchange, and/or corticosteroids may be used to temper cytokine storm while the work-up continues. Anakinra (recombinant IL-1 receptor antagonist) can be used for secondary HLH, especially when given early in the disease course [73]. Anakinra is currently being studied in a randomized, double-blind, placebo-controlled trial (ClinicalTrials.gov identifier: NCT02780583) to test its safety and efficacy in the treatment of secondary HLH/MAS in children and adults. Patients with multi-organ failure may require organ-specific therapy.

(2) Treatment of CNS-HLH

The clinical presentation of CNS disease in HLH is highly variable. Furthermore, occurrence of neurological symptoms is not included as a diagnostic criteria of HLH. However, it is important to suspect HLH in a child with unexplained neurologic manifestations, especially patients with fever, cytopenia, and hepatosplenomegaly. To make a diagnosis of CNS-HLH, a lumbar puncture with cerebrospinal fluid (CSF) analysis and MRI should always be done in all cases regardless of the presence or absence of neurological signs or symptom [74]. Treatment options for CNS-HLH include, those commonly used in systemic HLH, including corticosteroids, etoposide, cyclosporine A, anti-thymocyte globulin (ATG), and alemtuzumab. In addition, intrathecal treatment with methotrexate and corticosteroids has become a standard care and is likely to be beneficial [75]. Therapy must be started without delay to prevent late effects in HLH. An ongoing trial (NCT01818492) is an anti-IFN-g antibody (NI-0501), which is currently being tested. Another very promising agent is the Janus kinase (JAK)1/2 inhibitor ruxolitinib shown in a recent study of two murine models of HLH to be effective. In the Rab27a-/-mice, CNS involvement was significantly reduced with ruxolitinib therapy [76]. HSCT also represents an important CNS-HLH treatment [77].

(3) HSCT

HSCT is mandatory in patients with primary HLH, recurrent or progressive HLH despite recommended chemoimmunotherapy, and CNS involvement [56,67,78-80]. The pediatric HLH-94 trial reported a 5-year OS of 66% of those who underwent HCT [81]. Other studies reported similar outcomes with 5-year OS ranging from 49% to 64% with myeloablative conditioning (MAC) approaches, with the vast majority of mortality occurring in the first 6 months after HCT [82,83]. Several studies also note feasibility of umbilical cord transplant for HLH, with retrospective series reporting 65% to 71% long-term OS [84, 85]. The Cincinnati Children’s group showed potential superiority of reduced intensity conditioning (RIC) for HLH in a retrospective review which identified 43% estimated 3-year survival for patients transplanted with MAC (n=14) compared with 92% for patients transplanted with RIC (n=26) in 2010 [86]. RIC regimens are generally recommended as they are associated with better survival, though they can be complicated by high rates of mixed chimerism and graft failure since then [87,88]. Future study with the RIC approach is expected to improve the level and stability of donor engraftment. To avoid delays in proceeding to HSCT, human leukocyte antigen (HLA) typing and initiation of the stem cell donor search should be done as soon as it is suspected that a patient has a genetic form of HLH.

(4) Salvage therapy

It is clear that approximately 25-50% of patients will fail to achieve a complete response to standard therapy and may require additional treatment with the same drugs or alternative “salvage” agents. Relapses may respond to intensification of standard therapy, or may require additional or alternative therapies [89].

① Alemtuzumab: Alemtuzumab is a therapeutic monoclonal antibody directed against the CD52 antigen, a small GPI-anchored protein which is expressed on lymphocytes including T cells, NK cells, and B cells. A larger case series (n=22) of pediatric and young adult patients, who received alemtuzumab for salvage therapy of primary HLH, has been reported [90]. A partial response was achieved in 14 of 22 patients (64%) [90]. The remaining patients failed to respond or had improvement in only one sign or symptom of HLH [90]. Seventy-seven percent of patients survived to undergo allo-HSCT. Patients experienced an acceptable spectrum of complications, including CMV and adenovirus viremia [90]. However, the observation time to determine a response was limited to 2 weeks in this case series given that most patients moved quickly to allo-HSCT [90].

② Ruxolitinib: JAK 1/2 inhibitor, has shown promise in mouse models of primary and secondary HLH [91]. In murine models, ruxolitinib has been shown to both prevent and treat HLH by decreasing cytokine production and inflammation via inhibition of signal transducer and activator of transcription 1 (STAT1) signaling [92]. Two case reports described the use of ruxolitinib for patients with refractory HLH [93,94]. They reported that patients with refractory HLH became afebrile, followed by rapid improvements in respiratory, liver, and hemodynamic function within 24 hours of ruxolitinib treatment [93,94]. The most recent study for ruxolitinib showed that the overall response rate (RR) of the 34 patients was 73.5% (25/34 patients), with 14.7% (5/34 patients) in complete response (CR) and 58.8% (20/34 patients) in PR [95]. All patients experienced an acceptable spectrum of complications [95]. However, the remission depth is not sufficient, thus the authors hoped to combine ruxolitinib with the previous DEP (doxorubicin-etoposide-methylprednisolone) regimen to treat HLH [95].

③ Emapalumab: Emapalumab is a fully human immunoglobulin G1 monoclonal antibody that noncompetitively inhibits IFN‐g [92]. Administration of antibodies to deplete CD8+ T cells, as well as neutralization of IFN‐g, rescued the HLH-like pathology and improved survival in mice [92]. The safety and efficacy of emapalumab was assessed in a phase 2/3 trial (NCT01818492) in pediatric patients with presumed primary HLH [96-99]. The trial included 34 patients with presumed primary HLH and active disease who were treatment or were intolerant to standard treatments [99]. The median age of the study population was 1 year of age. Genetic mutations were present in 79% of patients. The overall RR was 63% (95% confidence interval, 42% to 81%; P=0.0134) and 12-month survival was 73%. Serious adverse reactions occurred in 53% of emapalumab recipients and included infections, gastrointestinal hemorrhage, and multiple organ dysfunction. Emapalumab is now approved for use in patients with adult or pediatric primary HLH that is refractory, recurrent, or progressive or in those who have intolerance to conventional therapy.

4) Future directions

HLH in children remains a challenge, but significant advances have been made in the last 20 years. Advances in rapid screening diagnostics makes it possible to quickly evaluate patients for many inherited diseases, and newer biomarkers are helping to elucidate the physiologic processes in patients with HLH. Novel targeted treatment agents are being developed. Well-designed clinical trials through international cooperation between investigators will bring further improved outcomes for patients with HLH.

Conflict of Interest Statement

The author has no conflict of interest to declare.

References
  1. Histiocytosis syndromes in children. Writing Group of the Histiocyte Society. Lancet 1987;1:208-9.
    Pubmed
  2. Filipovich A, McClain K, Grom A. Histiocytic disorders: recent insights into pathophysiology and practical guidelines. Biol Blood Marrow Transplant 2010;16:S82-9.
    Pubmed CrossRef
  3. Gonzalez CL, Jaffe ES. The histiocytoses: clinical presentation and differential diagnosis. Oncology (Williston Park) 1990;4:47-60; discussion 60, 62.
    Pubmed
  4. Badalian-Very G, Vergilio JA, Degar BA, et al. Recurrent BRAF mutations in Langerhans cell histiocytosis. Blood 2010;116:1919-23.
    Pubmed KoreaMed CrossRef
  5. Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood 2016;127:2672-81.
    Pubmed KoreaMed CrossRef
  6. Julien H, Fleur CA, Barret JR, et al. Histiocytoses: emerging neoplasia behind inflammation. Lancet Oncol 2017;18:e113-25.
    Pubmed CrossRef
  7. Abla O, Egeler RM, Pritchard J, et al. Langerhans cell histiocytosis;current concepts and treatments. Cancer Treat Rev 2010;36:354-9.
    Pubmed CrossRef
  8. Hyman DM, Diamond EL, Vibat CR, et al. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer Discov 2015;5:64-71.
    Pubmed KoreaMed CrossRef
  9. Badalian-Very G, Vergilio JA, Degar BA, Rodriguez-Galindo C, Rollins BJ. Recent advances in the understanding of Langerhans cell histiocytosis. Br J Haematol 2012;156:163-72.
    Pubmed CrossRef
  10. Allen CE, Merad M, McClain KL. Langerhans-cell histiocytosis. N Engl J Med 2018;379:856-68.
    Pubmed KoreaMed CrossRef
  11. Allen CE, Ladisch S, McClain KL. How I treat Langerhans cell histiocytosis. Blood 2015;126:26-35.
    Pubmed KoreaMed CrossRef
  12. Simko SJ, Garmezy B, Abhyankar H, et al. Differentiating skin-limited and multisystem Langerhans cell histiocytosis. J Pediatr 2014;165:990-6.
    Pubmed KoreaMed CrossRef
  13. Nauert C, Zornoza J, Ayala A, Harle TS. Eosinophilic granuloma of bone: diagnosis and management. Skeletal Radiol 1983;10:227-35.
    Pubmed CrossRef
  14. Jubran RF, Marachelian A, Dorey F, Malogolowkin M. Predictors of outcome in children with Langerhans cell histiocytosis. Pediatr Blood Cancer 2005;45:37-42.
    Pubmed CrossRef
  15. Kotecha R, Venkatramani R, Jubran RF, et al. Clinical outcomes of radiation therapy in the management of Langerhans cell histiocytosis. Am J Clin Oncol 2014;37:592-6.
    Pubmed CrossRef
  16. PS Jang, ES Park, KH Yoo, HY Shin, HS Ahn. Treatment of Langerhans cell histiocytosis with indomethacin. Clin Pediatr Hematol Oncol 2001;8:77-81.
  17. Munn SE, Olliver L, Broadbent V, Pritchard J. Use of indomethacin in Langerhans cell histiocytosis. Med Pediatr Oncol 1999;32:247-9.
    Pubmed CrossRef
  18. Gadner H, Minkov M, Grois N, et al. Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis. Blood 2013;121:5006-14.
    Pubmed CrossRef
  19. Farran RP, Zaretski E, Egeler RM. Treatment of Langerhans cell histiocytosis with pamidronate. J Pediatr Hematol Oncol 2001;23:54-6.
    Pubmed CrossRef
  20. Chellapandian D, Makras P, Kaltsas G, et al. Bisphosphonates in Langerhans cell histiocytosis: An international retrospective case series. Mediterr J Hematol Infect Dis 2016;8:e2016033.
    Pubmed KoreaMed CrossRef
  21. Morimoto A, Shioda Y, Imamura T, et al. Nationwide survey of bisphosphonate therapy for children with reactivated Langerhans cell histiocytosis in Japan. Pediatr Blood Cancer 2011;56:110-5.
    Pubmed CrossRef
  22. Gadner H, Grois N, Arico M, et al. A randomized trial of treatment for multisystem Langerhans’ cell histiocytosis. J Pediatr 2001;138:728-34.
    Pubmed CrossRef
  23. Gadner H, Grois N, Pötschger U, et al. Improved outcome in multisystem Langerhans cell histiocytosis is associated with therapy intensification. Blood 2008;111:2556-62.
    Pubmed CrossRef
  24. Dunger DB, Broadbent V, Yeoman E, et al. The frequency and natural history of diabetes insipidus in children with Langerhans-cell histiocytosis. N Engl J Med 1989;321:1157-62.
    Pubmed CrossRef
  25. Dhall G, Finlay JL, Dunkel IJ, et al. Analysis of outcome for patients with mass lesions of the central nervous system due to Langerhans cell histiocytosis treated with 2-chlorodeoxyadenosine. Pediatr Blood Cancer 2008;50:72-9.
    Pubmed CrossRef
  26. Ng Wing Tin S, Martin-Duverneuil N, Idbaih A, et al. Efficacy of vinblastine in central nervous system Langerhans cell histiocytosis:a nationwide retrospective study. Orphanet J Rare Dis 2011;6:83.
    Pubmed KoreaMed CrossRef
  27. Wnorowski M, Prosch H, Prayer D, et al. Pattern and course of neurodegeneration in Langerhans cell histiocytosis. J Pediatr 2008;153:127-32.
    Pubmed CrossRef
  28. Idbaih A, Donadieu J, Barthez MA, et al. Retinoic acid therapy in “degenerative-like” neuro-langerhans cell histiocytosis:a prospective pilot study. Pediatr Blood Cancer 2004;43:55-8.
    Pubmed CrossRef
  29. Imashuku S. High dose immunoglobulin (IVIG) may reduce the incidence of Langerhans cell histiocytosis (LCH)-associated central nervous system involvement. CNS Neurol Disord Drug Targets 2009;8:380-6.
    Pubmed CrossRef
  30. Allen CE, Flores R, Rauch R, et al. Neurodegenerative central nervous system Langerhans cell histiocytosis and coincident hydrocephalus treated with vincristine/cytosine arabinoside. Pediatr Blood Cancer 2010;54:416-23.
    Pubmed KoreaMed CrossRef
  31. Minkov M, Steiner M, Pötschger U, et al. Reactivations in multisystem Langerhans cell histiocytosis: data of the international LCH registry. J Pediatr 2008;153:700-5, 705.e1-2.
    Pubmed CrossRef
  32. Weitzman S, Braier J, Donadieu J, et al. 2’-Chlorodeoxyadenosine (2-CdA) as salvage therapy for Langerhans cell histiocytosis (LCH). Results of the LCH-S-98 protocol of the Histiocyte Society. Pediatr Blood Cancer 2009;53:1271-6.
    Pubmed CrossRef
  33. Donadieu J, Bernard F, van Noesel M, et al. Cladribine and cytarabine in refractory multisystem Langerhans cell histiocytosis:results of an international phase 2 study. Blood 2015;126:1415-23.
    Pubmed KoreaMed CrossRef
  34. Simko SJ, Tran HD, Jones J, et al. Clofarabine salvage therapy in refractory multifocal histiocytic disorders, including Langerhans cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman disease. Pediatr Blood Cancer 2014;61:479-87.
    Pubmed KoreaMed CrossRef
  35. Veys PA, Nanduri V, Baker KS, et al. Haematopoietic stem cell transplantation for refractory Langerhans cell histiocytosis:outcome by intensity of conditioning. Br J Haematol 2015;169:711-8.
    Pubmed KoreaMed CrossRef
  36. Kudo K, Maeda M, Suzuki N, et al. Nationwide retrospective review of hematopoietic stem cell transplantation in children with refractory Langerhans cell histiocytosis. Int J Hematol 2020;111:137-48.
    Pubmed CrossRef
  37. Haroche J, Cohen-Aubart F, Emile JF, et al. Dramatic efficacy of vemurafenib in both multisystemic and refractory Erdheim-Chester disease and Langerhans cell histiocytosis harboring the BRAF V600E mutation. Blood 2013;121:1495-500.
    Pubmed CrossRef
  38. Donadieu J, Larabi IA, Tardieu M, et al. Vemurafenib for refractory multisystem Langerhans cell histiocytosis in children:an international observational study. J Clin Oncol 2019;37:2857-65.
    Pubmed CrossRef
  39. Boussemart L, Routier E, Mateus C, et al. Prospective study of cutaneous side-effects associated with BRAF inhibitor vemurafenib:a study of 42 patients. Ann Oncol 2013;24:1691-7.
    Pubmed CrossRef
  40. Diamond EL, Durham BH, Haroche J, et al. Diverse and targetable kinase alterations drive histiocytic neoplasms. Cancer Discov 2016;6:154-65.
    Pubmed KoreaMed CrossRef
  41. Diamond EL, Durham BH, Dogan A, et al. Phase 2 trial of single-agent cobimetinib for adults with BRAF V600-mutant and wild-type histiocytic disorders. Blood 2017;130(Suppl 1):257.
  42. Long GV, Weber JS, Infante JR, et al. Overall survival and durable responses in patients with BRAF V600-mutant metastatic melanoma receiving dabrafenib combined with trametinib. J Clin Oncol 2016;34:871-8.
    Pubmed CrossRef
  43. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med 2015;372:30-9.
    Pubmed CrossRef
  44. Henter JI, Samuelsson-Horne A, Arico M, et al. Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood 2002;100:2367-73.
    Pubmed CrossRef
  45. Henter JI, Horne A, Arico M, et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2007;48:124-31.
    Pubmed CrossRef
  46. Filipovich AH. Hemophagocytic lymphohistiocytosis (HLH) and related disorders. Hematology Am Soc Hematol Educ Program 2009:127-31.
    Pubmed CrossRef
  47. Fujiwara F, Hibi S, Imashuku S. Hypercytokinemia in hemophagocytic syndrome. Am J Pediatr Hematol Oncol 1993;15:92-8.
    Pubmed CrossRef
  48. Eskander ED, Harvey HA, Givant E, Lipton A. Phase I study combining tumor necrosis factor with interferon-alpha and interleukin-2. Am J Clin Oncol 1997;20:511-4.
    Pubmed CrossRef
  49. Ammann S, Lehmberg K, Zur Stadt U, et al. Effective immunological guidance of genetic analyses including exome sequencing in patients evaluated for hemophagocytic lymphohistiocytosis. J Clin Immunol 2017;37:770-80.
    Pubmed CrossRef
  50. Feldmann J, Menasche G, Callebaut I, et al. Severe and progressive encephalitis as a presenting manifestation of a novel missense perforin mutation and impaired cytolytic activity. Blood 2005;105:2658-63.
    Pubmed CrossRef
  51. Beaty AD, Weller C, Levy B, et al. A teenage boy with late onset hemophagocytic lymphohistiocytosis with predominant neurologic disease and perforin deficiency. Pediatr Blood Cancer 2008;50:1070-2.
    Pubmed CrossRef
  52. Murphy C, Nanthapisal S, Gilmour K, et al. Progressive neurologic disorder: Initial manifestation of hemophagocytic lymphohistiocytosis. Neurology 2016;86:2109-11.
    Pubmed KoreaMed CrossRef
  53. Jordan MB, Allen CE, Greenberg J, et al. Challenges in the diagnosis of hemophagocytic lymphohistiocytosis: recommendations from the North American Consortium for Histiocytosis (NACHO). Pediatr Blood Cancer 2019;66:e27929.
    Pubmed CrossRef
  54. Allen CE, Yu X, Kozinetz CA, McClain KL. Highly elevated ferritin levels and the diagnosis of hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2008;50:1227-35.
    Pubmed CrossRef
  55. Otrock ZK, Hock KG, Riley SB, de Witte T, Eby CS, Scott MG. Elevated serum ferritin is not specific for hemophagocytic lymphohistiocytosis. Ann Hematol 2017;96:1667-72.
    Pubmed CrossRef
  56. Jordan MB, Allen CE, Weitzman S, Filipovich AH, McClain KL. How I treat hemophagocytic lymphohistiocytosis. Blood 2011;118:4041-52.
    Pubmed KoreaMed CrossRef
  57. Lehmberg K, McClain KL, Janka GE, Allen CE. Determination of an appropriate cut-off value for ferritin in the diagnosis of hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2014;61:2101-3.
    Pubmed CrossRef
  58. Hayden A, Lin M, Park S, et al. Soluble interleukin-2 receptor is a sensitive diagnostic test in adult HLH. Blood Adv 2017;1:2529-34.
    Pubmed KoreaMed CrossRef
  59. Lin M, Park S, Hayden A, et al. Clinical utility of soluble interleukin2 receptor in hemophagocytic syndromes: a systematic scoping review. Ann Hematol 2017;96:1241-51.
    Pubmed CrossRef
  60. Gurunathan A, Boucher AA, Mark M, et al. Limitations of HLH-2004 criteria in distinguishing malignancy-associated hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer 2018;65:e27400.
    Pubmed CrossRef
  61. Tsuji T, Hirano T, Yamasaki H, Tsuji M, Tsuda H. A high sIL-2R/ferritin ratio is a useful marker for the diagnosis of lymphoma-associated hemophagocytic syndrome. Ann Hematol 2014;93:821-6.
    Pubmed KoreaMed CrossRef
  62. Rubin TS, Zhang K, Gifford C, et al. Perforin and CD107a testing is superior to NK cell function testing for screening patients for genetic HLH. Blood 2017;129:2993-9.
    Pubmed KoreaMed CrossRef
  63. Chiang SCC, Bleesing JJ, Marsh RA. Current flow cytometric assays for the screening and diagnosis of primary HLH. Front Immunol 2019;10:1740.
    Pubmed KoreaMed CrossRef
  64. Weiss ES, Girard-Guyonvarc'h C, Holzinger D, et al. Interleukin18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood 2018;131:1442-55.
    Pubmed KoreaMed CrossRef
  65. Tang Y, Xu X, Song H, et al. Early diagnostic and prognostic significance of a specific Th1/Th2 cytokine pattern in children with haemophagocytic syndrome. Br J Haematol 2008;143:84-91.
    Pubmed CrossRef
  66. Xu XJ, Tang YM, Song H, et al. Diagnostic accuracy of a specific cytokine pattern in hemophagocytic lymphohistiocytosis in children. J Pediatr 2012;160:984-90.
    Pubmed CrossRef
  67. Henter JI, Samuelsson-Horne A, Arico M, et al. Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood 2002;100:2367-73.
    Pubmed CrossRef
  68. Bergsten E, Horne A, Arico M, et al. Confirmed efficacy of etoposide and dexamethasone in HLH treatment: long-term results of the cooperative HLH-2004 study. Blood 2017;130:2728-38.
    Pubmed KoreaMed CrossRef
  69. Ehl S, Astigarraga I, von Bahr Greenwood T, 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 2018;6:1508-17.
    Pubmed CrossRef
  70. Milone MC, Tsai DE, Hodinka RL, et al. Treatment of primary Epstein-Barr virus infection in patients with X-linked lymphoproliferative disease using B-cell-directed therapy. Blood 2005;105:994-6.
    Pubmed CrossRef
  71. Imashuku S. Treatment of Epstein-Barr virus-related hemophagocytic lymphohistiocytosis (EBVHLH); update 2010. J Pediatr Hematol Oncol 2011;33:35-9.
    Pubmed CrossRef
  72. Chellapandian D, Das R, Zelley K, et al. Treatment of Epstein Barr virus-induced haemophagocytic lymphohistiocytosis with rituximab-containing chemo-immunotherapeutic regimens. Br J Haematol 2013;162:376-82.
    Pubmed KoreaMed CrossRef
  73. Eloseily EM, Weiser P, Crayne CB, et al. Benefit of anakinra in treating pediatric secondary hemophagocytic lymphohistiocytosis. Arthritis Rheumatol 2020;72:326-34.
    Pubmed CrossRef
  74. Horne A, Trottestam H, Arico M, et al. Frequency and spectrum of central nervous system involvement in 193 children with haemophagocytic lymphohistiocytosis. Br J Haematol 2008;140:327-35.
    Pubmed CrossRef
  75. Horne A, Wickstrom R, Jordan MB, et al. How to treat involvement of the central nervous system in hemophagocytic lymphohistiocytosis? Curr Treat Options Neurol 2017;19:3.
    Pubmed KoreaMed CrossRef
  76. Maschalidi S, Sepulveda FE, Garrigue A, Fischer A, de Saint Basile G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood 2016;128:60-71.
    Pubmed CrossRef
  77. Sparber-Sauer M, Honig M, Schulz AS, et al. Patients with early relapse of primary hemophagocytic syndromes or with persistent CNS involvement may benefit from immediate hematopoietic stem cell transplantation. Bone Marrow Transplant 2009;44:333-8.
    Pubmed CrossRef
  78. Jordan MB, Filipovich AH. Hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis: a journey of a thousand miles begins with a single (big) step. Bone Marrow Transplant 2008;42:433-7.
    Pubmed CrossRef
  79. Horne A, Janka G, Maarten Egeler R, et al. Haematopoietic stem cell transplantation in haemophagocytic lymphohistiocytosis. Br J Haematol 2005;129:622-30.
    Pubmed CrossRef
  80. Seo JJ. Hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis: recent advances and controversies. Blood Res 2015;50:131-9.
    Pubmed KoreaMed CrossRef
  81. Trottestam H, Horne A, Aricò M, et al. Chemoimmunotherapy for hemophagocytic lymphohistiocytosis: long-term results of the HLH-94 treatment protocol. Blood 2011;118:4577-84.
    Pubmed KoreaMed CrossRef
  82. Ouachée-Chardin M, Elie C, de Saint Basile G, et al. Hematopoietic stem cell transplantation in hemophagocytic lymphohistiocytosis:a single-center report of 48 patients. Pediatrics 2006;117:e743-e50.
    Pubmed CrossRef
  83. Baker KS, Filipovich AH, Gross TG, et al. Unrelated donor hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis. Bone Marrow Transplant 2008;42:175-80.
    Pubmed CrossRef
  84. Ohga S, Kudo K, Ishii E, et al. Hematopoietic stem cell transplantation for familial hemophagocytic lymphohistiocytosis and Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in Japan. Pediatr Blood Cancer 2010;54:299-306.
    Pubmed CrossRef
  85. Patel SA, Allewelt HA, Troy JD, et al. Durable chimerism and long-term survival after unrelated umbilical cord blood transplantation for pediatric hemophagocytic lymphohistiocytosis:a single-center experience. Biol Blood Marrow Transplant 2017;23:1722-8.
    Pubmed CrossRef
  86. Marsh RA, Vaughn G, Kim MO, et al. Reduced-intensity conditioning significantly improves survival of patients with hemophagocytic lymphohistiocytosis undergoing allogeneic hematopoietic cell transplantation. Blood 2010;116:5824-31.
    Pubmed CrossRef
  87. Marsh RA, Jordan MB, Filipovich AH. Reduced-intensity conditioning haematopoietic cell transplantation for haemophagocytic lymphohistiocytosis: an important step forward. Br J Haematol 2011;154:556-63.
    Pubmed KoreaMed CrossRef
  88. Allen CE, Marsh R, Dawson P, et al. Reduced-intensity conditioning for hematopoietic cell transplant for HLH and primary immune deficiencies. Blood 2018;132:1438-51.
    Pubmed KoreaMed CrossRef
  89. Marsh RA, Jordan MB, Talano JA, et al. Salvage therapy for refractory hemophagocytic lymphohistiocytosis: a review of the published experience. Pediatr Blood Cancer 2017;64:e26308.
    Pubmed CrossRef
  90. Marsh RA, Allen CE, McClain KL, et al. Salvage therapy of refractory hemophagocytic lymphohistiocytosiswith alemtuzumab. Pediatr Blood Cancer 2013;60:101-9.
    Pubmed KoreaMed CrossRef
  91. Das R, Guan P, Sprague L, et al. Janus kinase inhibition lessens inflammation and ameliorates disease in murine models of hemophagocytic lymphohistiocytosis. Blood 2016;127:1666-75.
    Pubmed KoreaMed CrossRef
  92. Maschalidi S, Sepulveda FE, Garrigue A, Fischer A, de Saint Basile G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood 2016;128:60-71.
    Pubmed CrossRef
  93. Broglie L, Pommert L, Rao S, et al. Ruxolitinib for treatment of refractory hemophagocytic lymphohistiocytosis. Blood Adv 2017;1:1533-6.
    Pubmed KoreaMed CrossRef
  94. Wang JS, Wang YN, Wu L, et al. Refractory/relapsed hemophagocytic lymphohistiocytosis treated with ruxolitinib: three cases report and literatures review. Zhonghua Xue Ye Xue Za Zhi 2019;40:73-5.
    Pubmed CrossRef
  95. Wang J, Wang Y, Wu L, et al. Ruxolitinib for refractory/relapsed hemophagocytic lymphohistiocytosis. Haematologica 2019:222471.
    Pubmed CrossRef
  96. Humblet-Baron S, Franckaert D, Dooley J, et al. IFN-γ and CD25 drive distinct pathologic features during hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol 2019;143:2215-26.e7.
    Pubmed CrossRef
  97. Locatelli F, Jordan MB, Allen CE, et al. Safety and efficacy of emapalumab in pediatric patients with primary hemophagocytic lymphohistiocytosis. Blood 2018;132(Suppl 1):LBA-6.
    CrossRef
  98. Al-Salama ZT. Emapalumab: first global approval. Drugs 2019;79:99-103.
    Pubmed CrossRef
  99. Vallurupalli M, Berliner N. Emapalumab for the treatment of relapsed/refractory hemophagocytic lymphohistiocytosis. Blood 2019;134:1783-6.
    Pubmed CrossRef


April 2024, 31 (1)
Full Text PDF
Citation
Twitter
Facebook

Cited By Articles
  • CrossRef (0)

Author ORCID Information
  • Hoi Soo Yoon