Next Article in Journal
Genetics and Genomics of SOST: Functional Analysis of Variants and Genomic Regulation in Osteoblasts
Next Article in Special Issue
Inflammatory Microenvironment and Specific T Cells in Myeloproliferative Neoplasms: Immunopathogenesis and Novel Immunotherapies
Previous Article in Journal
Steroidogenesis in Peripheral and Transition Zones of Human Prostate Cancer Tissue
Previous Article in Special Issue
Atypical Chronic Myeloid Leukemia: Where Are We Now?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Pathogenesis and Treatment Perspectives for Hypereosinophilia and Hypereosinophilic Syndromes

1
Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
2
Center of Experimental Oncology and Hematology, A.O.U. Policlinico “G.Rodolico-San Marco”, 95123 Catania, Italy
3
Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(2), 486; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020486
Submission received: 18 November 2020 / Revised: 29 December 2020 / Accepted: 30 December 2020 / Published: 6 January 2021
(This article belongs to the Special Issue BCR-ABL1 Negative Myeloproliferative Neoplasms)

Abstract

:
Hypereosinophilia (HE) is a heterogeneous condition with a persistent elevated eosinophil count of >350/mm3, which is reported in various (inflammatory, allergic, infectious, or neoplastic) diseases with distinct pathophysiological pathways. HE may be associated with tissue or organ damage and, in this case, the disorder is classified as hypereosinophilic syndrome (HES). Different studies have allowed for the discovery of two major pathogenetic variants known as myeloid or lymphocytic HES. With the advent of molecular genetic analyses, such as T-cell receptor gene rearrangement assays and Next Generation Sequencing, it is possible to better characterize these syndromes and establish which patients will benefit from pharmacological targeted therapy. In this review, we highlight the molecular alterations that are involved in the pathogenesis of eosinophil disorders and revise possible therapeutic approaches, either implemented in clinical practice or currently under investigation in clinical trials.

1. Introduction

1.1. Eosinophil Development

Eosinophils are white blood cells of the granulocytic lineage that play an important role in innate immune functions [1] and develop in bone marrow from pluripotent stem cells expressing CD34+CD125+ antigens. These cells represent about 5% of the circulating blood leukocytes with an absolute eosinophil count (AEC) in healthy adults that is usually between 350 and 500/mm3, which increases during inflammatory processes, such as allergic diseases, parasitic, bacteria, and virus infection [2,3].
Structurally, they possess segmented bi-lobed nuclei and specific primary and secondary granules. Primary granules exhibit lysophospholipase activity that is involved in eosinophilic-dependent tissues inflammation [4]. Secondary granules contain many mediators, such as major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil peroxidase (EPO), and eosinophil-derived neurotoxin (EDN), which are all able to induce both inflammation and tissue damage [5].
Furthermore, eosinophils are equipped of lipid bodies that play a critical role in asthma, as they cause eicosanoidis production [6]. Finally, they are potent productors of both reactive oxygen species and nitric oxide, which promote the anti-bacterial activity, while the ability to internalize the respiratory syncytial (RSV) and influenza viruses document the role of eosinophils in the viral response [7,8].

1.2. Eosinophil Contents, Biology and Homeostatic Immune Role

Detecting tissue-resident eosinophils showed that they are distributed in heart, skin, lung, and kidneys [9]. Despite this observation, under the homeostasis condition, eosinophils are particularly abundant in the gastrointestinal tract (GI), where they are involved in different biological processes. Both a beneficial and nonbeneficial role of eosinophils in GI tract have been postulated.
The first role is based on the ability of gastrointestinal eosinophils to mediate anti-parasitic response and promote with them a symbiotic association that is aimed at the maintenance of tissue homeostasis. Moreover, in the GI tract, a high number of eosinophils trapping bacteria represents an effective mechanism to protect this tissue from bacterial invasion.
The nonbeneficial role of gastrointestinal eosinophils is reported in Eosinophil Gastrointestinal Disorders (EGIDs) and Inflammation Bowel Disease (IBD). The pathogenesis of both diseases is dependent on tissue infiltration of eosinophils, followed by the accumulation of activated immune cells, such as B and T cells, as well as the production of pro-inflammatory cytokines [8].
An additional particular function of eosinophil is their role in tissue regeneration and remodeling. By the secretion of IL-4, eosinophils are able to facilitate liver and muscle regeneration [10,11], while the increased presence of these cells in the endometrium prompted speculation that they may play a role in tissue remodeling during ovulation and menstruation [12]. The absolute number and biology of eosinophils are both usually controlled by type-2 cytokines, such as interleukin-5 (IL-5), granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukin-3 (IL-3), produced by T-lymphocytes, mast cells, and stromal cells. IL-5, IL-3, and GM-CSF induce eosinophils maturation, survival, and apoptosis inhibition by PI3K, ERK, and STATs pathway activation. However, in this biological system, IL-5 shows a more prominent role than IL-3 and GM-CSF dependent on its high specificity for this leukocyte subset. In particular, the IL-5 receptor (IL-5R) plays a central role of intracellular signals regulation by its α and β-chains. The α-chain contains the ligand-binding subunit while the β-chain is defined as non-ligand-binding subunit and it mediates the intracellular signal transduction. The β-chain, in turn, is shared with IL-3 and GM-CSF receptors (IL-3R) (GM-CSFR), thus supporting the intriguing role of IL-5 as central mediator of type-2 cytokines-dependent eosinophil survival. An interestingly involvement of IL-5 concerns its production by group 2 innate lymphoid cells (ILC2s) after interleukin-33 (IL-33) stimulation. IL-33 promotes the IL-5 production from ILC2s, which, in turn, releasing IL-5 improves eosinophils expansion and survival. Hence, ILC2s are a new important mediator of IL-33-driven eosinophil disorder development [13,14].
Hence, IL-5, to date, is an attractive therapeutic target for the treatment of eosinophil-mediated disorders [15]. Nevertheless, despite these results, the published data report that IL-5 over-expression or down-regulation, alone, fail to induce eosinophil-mediated damage or eosinophils maturation [16], making its role unclear. However, all together, these observations implicate that the deregulation of IL-3, IL-5, and GM-CSF signaling may cause HES [15,17,18].
Finally, an important role in eosinophils biology concerns the fibrogenic activity of transforming growth factor beta-1 (TGF-B1). Eosinophils are a strong productor of TGF-B1, which is involved in airway remodeling or in disease state in different tissue, such as skin (atopy), nose (nasal polyposis), and blood (idiopathic hypereosinophilic syndrome) [19].
Eosinophils biology is also regulated by the cell surface glycan-binding protein, named siglecs (sialic acid immunoglobulin-like lectins). The most important glycan in eosinophils biology is Siglec-8 (originally named sialoadhesin family 2—SAF-2), as it is expressed selectively in these leukocyte types. Siglec-8 is stimulated by sialic acids, which induce the activation of two tyrosine-based motifs, defined immunoreceptor tyrosine-based inhibitor motif (ITIM) and a membrane-distal immunoreceptor tyrosine-based switch motif (ITSM), thus initiating the downstream receptor functions. Siglec-8 is involved in reactive oxygen species (ROS) production, in the loss of mitochondrial membrane potential, ERK1/2 activation, and caspase cleavage modulating apoptosis and cell survival [20]. For these regions, Siglec-8 was studied as a therapeutic target in patients with eosinophilic disorders using chimeric antibodies [21].

1.3. Eosinophil Recruitment into Blood and Tissue, Survival and Death

The term eosinophilia is employed for a small increase of the AEC in the blood (up to 1500/mm3), while hypereosinophilia (HE) indicates an AEC greater than 1500/mm3 on two consecutive blood samples drawn at a one-month interval. This persistent eosinophilia is usually linked to helminth infections, allergies, atopy, drugs, neoplastic disorders, or autoimmune diseases [22,23].
A second type of hypereosinophilia is tissue HE, which is defined as a percentage of eosinophils in the bone marrow (BM) that exceeds 20% of all nucleated cells, followed by extensive tissue infiltration, such as skin (in 69% of subjects), lung, and the gastrointestinal tract (44% and 38%, respectively) [22,23,24].
Upon activation, eosinophils infiltrate tissues, degranulate, and release proinflammatory cytokines that cause organ damage and dysfunction, defined as the Hypereosinophilic Syndrome (HES). HES represents a group of heterogeneous disorders characterized by persistent and unexplained HE in the blood or peripheral tissues usually associated with multiple organ damage or dysfunction. This damage may be due to direct cytotoxic effects of the eosinophil granulate contents or may occur because of the secondary involvement of other cell types [24,25]. Once eosinophils leave the blood circulation and migrate into tissue sites, they do not recirculate. Into the tissue, the survival is dependent on local production of cytokines that also prevent eosinophil apoptosis for several days. In fact, the patterns of cytokines regulate the recruitment of eosinophils to the specific tissue sites, activate endothelial cells, and induce tissue-resident cells to produce eosinophil-active chemokines to facilitate their preferential migration [26].
The cardiovascular system is often involved in HES [27,28].
The presenting symptoms of HE and HES are variable and they may include weakness, fatigue, cough, dyspnea and rhinitis, myalgias or angioedema, rash or fever, as well as severe tissue damage or end-organ failure [23,29]. Leukocytosis, anemia, abnormal platelet counts, increased vitamin B12 (>1000 pg/mL), and tryptase (>12 ng/mL) levels represent additional alterations that are associated with the disease [23,24].
Because HES is a rare neoplasm, its epidemiology has not been accurately investigated. Hence, the disease true incidence is unknown, which is mainly due to the lack of specific coding for the different HES variants. Although HES is mainly diagnosed in adults, 20 to 50 years old, it can also affect children and the elderly [30].
The aim of this review is to outline the molecular alterations that are involved in the pathogenesis of eosinophil disorders and provide an update on the therapeutic approaches that are available for the treatment of these disorders.

2. Classification of Hypereosinophilia and Hypereosinophilic Syndrome

HE and HES can usually be divided into multiple subgroups based upon clinical, laboratory, and molecular features.
HE is classified in three groups: primary (neoplastic or clonal), secondary (reactive), and idiopathic [24]. Primary HE is caused by a clonal stem cell disorder affecting the myeloid or lymphoid lineage of the malignant clone, while secondary HE can be associated with both pathological phenomena (e.g., parasitic infections, allergies, autoimmune disorders) and/or non-myeloid or solid tumors, in which eosinophilia results from the production of cytokines by malignant cells, such as in peripheral T-cell lymphoma and classical Hodgkin lymphoma. If the etiology is not primary or secondary and the HE persists for six or more months, then the disease is defined as Idiopathic HE (HEUS) [30]. Moreover, a rare familiar form of HE was recently identified that is characterized by autosomal dominant inheritance with a benign clinical course and seldom characterized by organ dysfunction [23,24,31].
Over the years, the Working Conference on Eosinophil Disorder and Syndromes proposed various terminologies for eosinophilic syndromes. The HE subtypes were then divided into a hereditary (familial) variant (HEFA), HE of undetermined significance (HEUS), primary (clonal/neoplastic) HE produced by clonal/neoplastic eosinophils (HEN), and secondary (reactive) HE (HER), with the latter group including the lymphocyte variant as a subtype. The HEUS acronym was introduced as a novel term instead of idiopathic HE [30].
Similar to the HE classification, HES can be grouped in primary (neoplastic), secondary (reactive), and idiopathic (undetermined significance), respectively, named HESN, HESR, and HESUS [23].
The main two pathogenic forms of HES are the myeloproliferative (M-HES) and lymphocytic (L-HES) forms of the disease, respectively, classified as Myeloid Hypereosinophilic Syndrome and Lymphocytic Hypereosinophilic Syndrome. Each group includes several clinically distinct HES disorders [32]. Patients that do not display the M- or L-HES diagnosis can be classified as being affected by a Idiopathic Hypereosinophilic Syndrome or a Chronic Eosinophilic Leukemia non otherwise specified (CEL-NOS) (Figure 1).

2.1. Myeloid Hypereosinophilic Syndrome

Patients with M-HES are characterized by hepatomegaly, splenomegaly, circulating myeloid precursors, increased serum vitamin B12 and/or tryptase levels, anemia, thrombocytopenia, hematologic diseases (myeloid fibrosis, left shift in maturation of myeloid precursors), and/or cytogenetic abnormalities [27,31,33,34]. The primary molecular defect that is responsible for this distinct phenotype is a gene fusion between FIP1-like 1 (FIPL1) and platelet-derived growth factor receptor alpha (PDGFRα), known as FIP1L1–PDGFRα fusion. Several other fusions involve genes encoding for the PDGFRα, PDGFRβ, fibroblast growth factor receptor 1 (FGFR1), breakpoint cluster region (BCR), janus kinase 2 (JAK2), fms-like tyrosine kinase 3 (FLT3), and Abelson tyrosine kinase 1 (ABL1) genes. Recently, the WHO added the periocentriolar material 1 (PCM1)-JAK2 fusion in the classification of this group [32,35]. Rarely, PDGFRβ rearrangements are cryptic, even if patients with this anomaly (involving over 30 gene fusion partners) can present a disease without eosinophilia [23]. M-HES related to gene fusions involving the FGFR1 gene are rare, although several groups reported up to 14 different FGFR1 gene partners [30,35].
In the last decade, cytogenetics and DNA sequencing have allowed for investigating the molecular alterations found in HES, demonstrating that somatic mutations are usually uncommon in patients harboring PDGFRα, PDGFRβ, or PCM1-JAK2 rearrangements, but significantly more frequent in FGFR1 rearranged cases [24].

2.2. Lymphocytic Hypereosinophilic Syndrome

The lymphocytic variant of HES is a less clearly defined disease entity that is characterized by the overproduction of eosinophilopoietic cytokines (IL-5 and/or IL-3) causing the recruitment of clonal activated T-lymphocytes (T-cells). IL-5 overproduction by T-cells is responsible for blood and tissue HE, which leads to clinical manifestations, while the expansion of the abnormal T-cell subset is usually asymptomatic, with the exception of a few cases that evolve to T-cell lymphoma [27]. Immunophenotypically, these abnormal T cells include double-negative cells, immature T-cells (e.g., CD3+CD4CD8), or cells without CD3 expression (e.g., CD3CD4+). Furthermore, additional immunophenotypic abnormalities include high CD5 expression on CD3CD4+ cells and the loss of the CD7 surface marker and/or expression of CD27 [23,36,37].
L-HES is found in 17–27% of subjects with unexplained eosinophilia or HES. Primary disease manifestations are superficial adenopathy (62%) with rheumatologic (29%), gastrointestinal (24%), pulmonary (19%), neurologic (10%), and cardiovascular (5%) organ involvement, but no significant lymphocytosis [27].
Moreover, elevated IgE and thymus and activation-regulated chemokine (TARC) in serum are common in patients with L-HES. Particularly, the detection of serum TARC levels, in addition to the increased production of cytokines, may provide additional support for a correct diagnosis [23,37,38].

2.3. Idiopathic Hypereosinophilic Syndrome and Chronic Eosinophilic Leukemia non Otherwise Specified

Idiopathic HES was defined as persistent HES with tissue/organ damage of unknown cause, whereas CEL-NOS presents clonal cytogenetic or molecular genetic abnormalities. In both syndromes, no rearrangements of PDGFRα, PDGFRβ, FGFR1 or PCM1-JAK2, ETS Variant Transcription Factor 1 (ETV6)-JAK2, and BCR-JAK2 fusion genes are present. In these cases, the cytogenetic and molecular alterations of chronic myeloid leukemia (CML), myelodysplastic/myeloproliferative neoplasms (MDS/MPN), chronic neutrophilic leukemia (CNL), and chronic myelomonocytic leukemia (CMML) should be excluded [39,40,41,42,43,44].

3. Molecular Pathogenesis in Hypereosinophilic Syndrome

The laboratory screening performed to formulate a HES diagnosis allow for us to understand molecular events that cause gene driver alterations in myeloid and lymphoid disorders that are associated with eosinophilia (summarized in Table 1).
PDGFRα and PDGFRβ fusions: PDGFRα and PDGFRβ are a class of receptors with TK activity, which are characterized by an extracellular ligand-binding region and two intracellular TK domains [45,46]. PDGFRα and β are monomeric transmembrane proteins that dimerize after binding PDGF, leading to TK domain activation. The activated catalytic domain promotes a cascade of signaling events via downstream pro-survival and anti-apoptotic effectors, such as SRC, STAT5, and the PI3K/RAS/MAP kinase pathway [47,48,49,50]. The most common PDGFR gene alterations reported in the eosinophilic syndrome are rearrangements with several partner genes, such as FIP1L1, BCR, and ETV6 (Figure 2) [34].
FIP1L1-PDGFRα fusions: the FIP1L1-PDGFRα rearrangement represents the most frequently recurrent aberration in eosinophilia detected in different hematopoietic cells, including eosinophils, neutrophils, T-, or B-cells [51]. Although it is generally expressed in chronic myeloid neoplasms that are associated with eosinophilia, patients with lymphoblastic leukemia/lymphoma (T-ALL/LBL) or, less frequently, B-cell acute leukemia display this fusion transcript [23]. The FIP1L1-PDGFRα fusion protein is expressed in 10–20% of patients that are affected by HEN/HESN, with a higher prevalence in males [24].
The FIP1L1-PDGFRα transcript is generated by juxtaposition of the 5′ and 3′ regions of FIP1L1 and PDGFRα, respectively. The fusion between these two genes is caused by an internal cryptic deletion, which is not detectable by cytogenetic banding techniques, resulting in an apparently normal karyotype. This deletion disrupts the juxtamembrane PDGFRα negative regulator domain, thus deregulating its TK activity [52,53,54,55]. The 5′-3′ juxtaposition involves exons 9–13 of FIP1L1 and exon 12 of PDGFRα, which can generate different FIP1L1-PDGFRα splicing isoforms (not shown) (Figure 2a). In addition, an insertion of an additional FIP1L1 intron sequence was identified in different patients [52,55,56].
Different researchers conducted several studies in human cell and in clonal HES mouse model in order to investigate the role of FIP1L1-PDGFRα in the pathogenesis of the disease. Several data were reported and, although the constitutive FIP1L1-PDGFRα kinase activation constrains a murine eosinophil-lineage commitment, in human hematopoietic progenitor cells promotes cytokine-independent colony formation without favoring eosinophil lineage by STAT5 and nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) activation [57,58,59]. In contrast, on the contrary of native PDGFRα, FIP1L1-PDGFRα does not activate the MAPK pathway, which suggests that its transforming properties do not require extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) [49,52,60]. This effect is dependent on different subcellular localization of native or fusion proteins. Hence, while FIP1L1- PDGFRα has cytosolic location, PDGFRα is a transmembrane receptor that may access RAS, an upstream mediator of MAPK signaling [56]. However, other authors showed that FIP1L1- PDGFRα may activate the eosinophilic linage-specific transcription factors through RAS/MEK/p38 cascade [61].
BCR-PDGFRα fusions: several authors reported that, unlike the common BCR-ABL1 chimeric gene found in CML [62,63], the BCR-PDGFRα fusion detected in HES associates with different hematological diseases, such as atypical CML and pre-B ALL [64,65,66]. Molecular analysis revealed that the rearrangement process causes an in-frame mRNA fusion between exons 1, 7, or 12 of BCR and 12 or 13 of PDGFRα (Figure 2b). This molecular event disrupts the negative regulator domain of PDGFRα and catalyzes aberrant TK activity that is mediated by BCR-dependent oligomerization [56].
ETV6-PDGFRβ fusions: over 32 fusion genes involving the PDGFRβ have been discovered and ETV6-PDGFRβ represents the most frequent translocation t(5;12)(q33;p13) [30,35] found in eosinophilia associated with CML [67]. In the most common variant, the ETV6-PDGFRβ fusion transcript is generated by exons 4 and 11, respectively (Figure 2c) [68].
The extracellular PDGFRβ region contains the ligand binding site that is replaced by ETV6, which promotes an oligomerization process resulting in the activation of the PDGFRβ tyrosine kinase [69]. Studies that were conducted in murine models indicate that ETV6-PDGFRβ causes growth factor-independent proliferation of Ba/F3 cells and—after mouse transplantation—leads to a myeloproliferative disease that is not associated with eosinophilia [70]. Moreover, human CD34+ cells lentivirally transduced with the chimeric protein displayed increased proliferation and showed an eosinophil differentiation when stimulated by eosinopoietic cytokines in a NF-kB-dependent manner [59,71].
FGFR1 fusions: the fibroblast growth factor receptor 1 (FGFR1) is a monomeric protein that dimerizes upon ligand binding. Dimerization drives TK activity, which promotes cell proliferation [72]. FGFR1 abnormalities involve at least 14 gene partners generating gene fusions with different incidences. FGFR1 rearrangements are not cryptic; hence, they can be diagnosed by conventional cytogenetic analyses [73]. The most common fusions are represented by Zinc finger MYM-type protein 2 (ZMYM2)-FGFR1 (ZMYM2 exon 17–FGFR1 exon 9: 40%), BCR-FGFR1 (BCR exon 4–FGFR1 exon 9: 18%), and Centriolin-FGFR1 [CNTRL (CEP110) exon 15–FGFR1 exon 9: 15%) [30,35,74,75] (Figure 3). In all of these pathologic fusions, the improper constitutive activation of the FGFR1 catalytic domain drives disease initiation and progression. In turn, the FGFR1 kinase phosphorylates specific tyrosine residues, promoting proliferation and pro-survival mediators, such as RAS/MAPK, PI3K/AKT, and STATs [24,72]. Hematological neoplasias that are associated with eosinophilia and expressing these chimeric proteins are MPN, ALL, and AML [24].
JAK2 fusions: JAK2 is a component of the intracellular JAK/STAT pathway. The signals that are mediated by this signaling axis play a critical role in modulating the immune system through multiple cytokine receptors [76,77,78,79]. Chromosome abnormalities that are located in 9p24 involve JAK2 and different gene partners (Figure 4) [80]. The PCM1-JAK2 fusion (PCM1 exon 25-JAK2 exon 9) (Figure 4a) [57] may be expressed in MPN, MDS B-ALL, or AML, and has been associated with different levels of eosinophilia in both blood and BM [57,58,81]. Additional patients have been described exhibiting rare JAK2 rearrangements that are identified as BCR-JAK2 (BCR exon 1-JAK2 exons 15, 17, 18 or 19) (Figure 4b) and ETV6-JAK2 (ETV6 exon 5-JAK2 exon 12) (Figure 4c) [82,83,84,85,86,87,88].
PCM1, BCR, and ETV6 encode for proteins containing a coiled-coil region that mediates an oligomerization process of the ensuing chimeric proteins. This event causes constitutive activation of the JAK2 kinase increasing cell proliferation, survival, and differentiation by STAT signaling [89,90,91]. The JAK2-rearranged eosinophilia displays an unfavorable clinical course with a rapid progression from chronic to acute leukemia [85].
Other gene fusions: eosinophilia-associated neoplasm can be characterized by rare rearrangements involving FLT3 and ABL1. The most common include ETV6-FLT3 and ETV6-ABL1, which are typically identified in chronic myeloid diseases, such as eosinophilic leukemia and/or in T-ALL leukemia/lymphoma [92,93]. For both rearrangements, the ETV6 coiled-coil region triggers an oligomerization process, which causes constitutive activation of the catalytic domains of either FLT3 or ABL1. This induces the activation of the RAS/MAPK pathway, which promotes cell survival and proliferation [24].
T-cell receptor gene rearrangements: IL-5 overproduction by activated mature T-cells, leading to the polyclonal expansion of eosinophils, has been reported in several studies [23,32,94,95]. Therefore, the detection of an aberrant T-cell immunophenotype by clonal TCR gene rearrangement is required for the diagnosis of most L-HES patients [95,96]. T-cell clonality is not detected in all patients with demonstrated aberrant lymphocyte cells [36]. However, a negative analysis may not reflect the true absence of clonality, as the clonal nature of the disease may go undetected, due to a lack of sensibility when the aberrant cells represent a small proportion of the total lymphocyte population. In these patients, Roufosse and colleagues suggest repeating clonality testing after the purification of aberrant T-cells and analyzing T-cell cytokine secretion profiles by measuring the concentrations in supernatants of cultured peripheral blood mononuclear cells [36]. It should be noted that a high portion of HESUS patients (18/42 patients, 43%) exhibit a clonal TCR gene rearrangement by PCR, although it is unclear whether such clonal T-cell population is always the cause of the disease [96].

Next Generations Sequencing Approaches to Investigate DNA Mutations in Patients with Eosinophilic Disorders

In the recent years, the next generation sequencing (NGS) [77] has been used in the identification of numerous mutations in a large proportion of myeloproliferative disorders and/or AML and CML patients [97,98,99,100,101]. Moreover, different studies investigated the use of NGS-based mutation panels to study HE patients. Baer and colleagues reported that somatic mutations are more frequent in patients with FGFR1 rearrangements when compared to those with PDGFRα, PDGFRβ, or PCM1-JAK2 alterations. For example, 83% of FGFR1-rearranged individuals harbored RUNX1 mutations [102]. Two additional studies reported a wide range of mutation frequencies (11–28%) in different cohorts of 98 and 51 patients with HEus and/or HESus [103,104]. By performing an NGS panel that was designed for myeloid neoplasias, Wand et al. found that, in 51 idiopathic HES individuals, the most frequently mutated genes are ASXL transcriptional regulator 1 (ASXL1) (43%), Tet methylcytosine dioxygenase 2 (TET2) (36%), Enhancer of zeste homolog 2(EZH2) (29%), SET binding protein 1 (SETBP1) (22%), Casitas B-lineage Lymphoma (CBL) (14%), and Notch homolog 1, translocation-associated (NOTCH1) (14%) [104]. The authors provided evidence of clonality for subjects with clinical or morphologic features that are suggestive of neoplasia and/or contributed to the diagnosis of CEL, NOS [104]. A Korean study investigated T-cell clonality and the impact of the mutations in 30 individuals that were diagnosed with HEus/HESus by performing NGS, TCR gene rearrangement assays, and a pathway network analysis [105]. They found that a 53.3% mutation frequency with the most frequently altered genes NOTCH1 (26.7%), Scribble Planar Cell Polarity Protein (SCRIB), and Stromal Antigen 2 (STAG2) (16.7%) and SH2B adapter protein 3 (SH2B3) (13.3%). They also identified 5 (MAPK1, RUNX1, GATA2, NOTCH1, and TP53) out of 21 candidate genes functionally linked to the eosinophilopoietic pathways and observed that 13.3% of patients had a clonal TCR rearrangement. The study suggested that mutations affecting eosinophilopoiesis highlighted a special subgroup of IHE/IHES and that these mutations were more likely to be associated with a clonal eosinophil proliferation [105]. Finally, Cross and colleagues found an activating STAT5B N642H driver mutation in 1.6% of patients with lymphoproliferative disorders that were referred for eosinophilia. The authors demonstrated that individuals with additional mutated genes, other than Splicing factor 3B subunit 1 (SF3B1), had an inferior overall survival (OS) when compared to those with the STAT5B mutation alone [106].

4. Therapeutic Option for Patients with Eosinophilic Disorders

The best clinical treatment of HES depends on disease etiology and subtypes. However, even in the absence of a known cause, HES must be promptly treated in order to reduce potential morbidity that can result from organ damage. In this regard, an AEC of 350–500/mm3 has been recommended as a threshold for starting treatment [23]. Multiple therapeutic approaches are currently employed (Table 2) with further compounds under investigation or in ongoing trials (Table 3). The main therapeutic options for HES patients can be divided in five groups: corticosteroids, cytotoxic agents, tyrosine kinase inhibitors, monoclonal antibodies (mAb), and chemotherapy (Figure 5, Table 2 and Table 3).

4.1. Corticosteroids

This group of drugs is the current mainstay for slowing and/or preventing organ damage and it can be used as first-line therapy in patients with strictly defined HES. Because steroid therapy can be complicated by side effects in patients requiring long-term treatment, different studies have been conducted in order to better define the right steroid dose [107,108]. An ongoing study (NCT 01524536) is trying to determine whether a single prednisone dose can be used to predict which subjects with hypereosinophilia will respond to treatment with individuals developing symptom recurrence or an increase in their AEC requiring the addition of a second drug, such as hydroxyurea (HU) (Table 3).

4.2. Cytotoxic Agents

The main drugs that are employed for the treatment of HES are hydroxyurea (as a first-line agent or in combination with corticosteroids in non-responders patients) [29,109] and Interferona (IFNα), with the latter drug used as a second-line agent after steroid-failure (Figure 5 and Table 2). IFNα can also be used in conjunction with corticosteroids or as a steroid-sparing agent for patients requiring higher doses of prednisone or presenting contraindications to steroid therapy [110,111,112]. In addition to HU and IFNα, dexpramipexole is an orally bioavailable synthetic aminobenzothiazole, which, in a non-randomized, proof-of-principle study, reduced blood and tissue eosinophils and enabled corticosteroid reduction or cessation in HES patients (Figure 5 and Table 3) [113]. Currently, a trial (NCT02101138) is evaluating whether dexpramipexole can reduce the steroid dose that is needed to control eosinophilia and HES symptoms (Table 3).

4.3. Tyrosine Kinase Inhibitors

Imatinib and Nilotinib: currently used as ABL1-directed inhibitors for CML patients [114,115,116], they were considered to be possible therapeutic agents in HES for their ability to inhibit PDGFR kinase activity. However, to date, only imatinib has been approved as a first-line treatment for patients with myeloid disease, with eosinophilia expressing FIP1L1-PDGFRα or carrying other PDGFRα or PDGFRβ fusions [107,117] (Table 2). The Food and Drug Administration (FDA)-recommended starting dose for patients with the FIP1L1-PDGFRα rearrangement is 100 mg daily, which is sufficient to achieve complete hematologic and molecular remissions. For patients with myeloid neoplasms and eosinophilia expressing PDGFRβ fusions, the recommended starting dose is 400 mg, being lowered to 100 mg during maintenance [117]. A phase 2-trial is evaluating the safety and efficacy of a combination of imatinib and ruxolitinib in reducing peripheral blood eosinophilia in patients with the myeloid form of HES (NCT00044304) (Table 3 and Figure 5). Furthermore, the experimental data obtained while using rat or mouse models demonstrated that ABL inhibition by imatinib reduces the TGF-B1 profibrogenic activity in renal and lung tissues interested from eosinophil disorders [118,119].
An ongoing study designed as a managed access program presently allows access to nilotinib for eligible patients that were diagnosed with HES (NCT04498871) (Table 3 and Figure 5).
Pemigatinib: INCB054828 is an oral FGFR1, 2 and 3 inhibitor that is currently under evaluation in FGFR1-rearranged myeloid/lymphoid neoplasms [120].
Ruxolitinib and Sorafenib: these two multi-kinase inhibitors should be considered to be a bridge to HSCT for patients displaying the JAK2 or FLT3 tyrosine kinase fusions, respectively (Table 2 and Figure 5) [92,121,122,123]. Ruxolitinib is also under investigation in HES patients expressing the BCR-JAK2 fusion, in order to determine their overall hematologic response to this drug (NCT03801434) (Table 3). In addition, Lierman and colleagues reported that Sorafenib seems to be an in vitro potent inhibitor in FIP1L1-PDGFRα rearranged patients with T674I mutation [124].
Monoclonal antibodies: several FDA-approved antibodies have shown benefit in reducing circulating eosinophils, either by targeting eosinophilopoietic cytokines, or by depleting eosinophils via antibody-dependent cellular cytotoxicity. The antibodies that are presently available or undergoing clinical trials are directed against IL-5, the IL-5 receptor, IgE, or the CD52 antigen.
Mepolizumab: a fully humanized monoclonal IgG antibody that inhibits the binding of IL-5 to the chain of the IL-5 receptor expressed on eosinophils, reducing their survival and TGF-B production [125,126,127]. The FDA has currently approved this compound for severe asthma and eosinophilic granulomatosis with polyangiitis, but not for HES. It is available on a compassionate use-bases for individuals with life-threatening HES who have failed at least three standard lines of treatment (NCT00244686) (Table 3 and Figure 5).
Reslizumab: a humanized anti-IL5 IgG4 monoclonal antibody approved by the FDA for severe eosinophilic asthma that has not yet been studied in HES [128] (Table 3 and Figure 5).
Benralizumab: an anti-IL5 receptor antibody that is employed in patients with severe, uncontrolled asthma [129], with initially unsatisfactory results in HES [130]. After binding to IL-5R, eosinophils become a target for destruction by NK cells via antibody-dependent cell-mediated cytotoxicity [131]. Currently, a phase 3 study is evaluating the efficacy and safety of benralizumab for HES patients (NCT04191304). A second trial (NCT02130882) is testing the ability of this drug to safely decrease eosinophils in individuals that are diagnosed with HES (Table 3 and Figure 5).
Omalizumab: an anti-IgE monoclonal antibody approved by the FDA for the treatment of asthma and chronic spontaneous urticaria. It showed promising activity in some eosinophilic disorders, although results were not as consistent as those that were seen with anti-IL-5 or anti-IL-5R antibodies (Table 2 and Figure 5) [132]. The drug may lead to the inhibition of the release of cytochines, such as IL-4, IL-5, and IL-13, as these are responsible for eosinophils recruitment and activation [132].
Alemtuzumab: an anti-CD52 monoclonal antibody that has been evaluated in HESus based on the expression of the CD52 antigen on the eosinophil surface (Table 2 and Figure 5) [133,134]. Verstovsek and colleagues found that alemtuzumab achieved a complete hematologic remission in 10/12 (83%) patients with refractory HES and a partial remission in the remaining two subjects [135].
Finally, chemotherapy, as well as autologous stem cell transplant, are usually employed for patients with eosinophilic leukemia, T-cell lymphomas, or other types of primary HES that are refractory to alternative treatments [136,137]. As an alternative to chemotherapy, an ongoing phase I/II trial (NCT03862157) is studying the association of venetoclax, azacitidine, and pevonedistat in patients with newly diagnosed acute myeloid leukemia and other hematological disorders, including CEL-NOS (Table 3 and Figure 5).

5. Conclusions

Eosinophilic disorders represent a group of highly heterogeneous diseases that are characterized by various degrees of persistent blood and/or tissue hypereosinophilia with potential for end-organ dysfunction [2,23]. Hence, a timely diagnosis is essential and it requires a combination of histopathologic, immunophenotypic, cytogenic, and molecular analyses. The identification of specific and recurrent genetic alterations in HES suggests the possible use of molecularly targeted therapies that have proven to be successful in many tumor types, as this approach selectively kills neoplastic cells that harbor a specific molecular hallmark [138,139,140,141,142,143,144].
In this context, the distinction of each HES variant is critical for the appropriate management of the disease. The development of non-invasive sampling methods, coupled with an extensive NGS-based molecular characterization, will be important in distinguishing the different disease variants and discriminating an eosinophil myeloid neoplasia from HEUS. Moreover, this approach will enable an accurate disease monitoring, promptly identifying patients with a rapidly progressing hematological malignancy.
The recent focus of HES is based on the increasing of availability compounds targeting different mediators and the cells involved in the mechanism of the disease.
Different ongoing clinical trials (Table 3) based on different drugs, used alone or in combination, will allow for a better understanding of the best initial therapy for any single patients with HES while taking the individual pathogenesis into consideration. Indeed, more targeted approach to treatment need an implementation of significative changes in the way that patients are managed through a more personalized approach to prognostication, the prediction of treatment responses.
Finally, with increasing use of anti-IL-5 or anti-IL-5R antibodies for hypereosinophilic disease in clinical practice, in the near future the focus should be on optimizing doses and regimens. A better combination of different active molecules will be investigated in order to design efficacious and minimally toxic tailored treatment regiments for patients with these complex disorders.

Author Contributions

Conceptualization, S.S.; writing-original draft preparation, S.S., M.M., L.M., M.S.P. and E.T.; data curation, S.S.; formal analysis, C.R., S.R.V., A.P. and C.T.; writing-review and editing, S.S., M.M., L.M., M.S.P., E.T., C.R., S.R.V., A.P., C.T. and S.D.G.; G.A.P. and P.V. critically reviewed the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Giuseppe Alberto Palumbo: homoraria from Novartis, Bristol Myers Squibb, Celgen, Amgen and Abbvie Paolo Vigneri: research funding from Novartis and Pfizer; honoraria from Astra-Zeneca, Celgene, Italfarmaco, Incyte, Novartis, Pfizer, Tesaro, and Teva. The other authors declare no conflict of interest.

Abbreviations

ABL1Abelson murine leukemia
AECAbsolute Eosinophil Count
ALLAcute Lymphoblastic Leukemia
BCRBreakpoint cluster region
CEL-NOSChronic Eosinophilic Leukemia non otherwise specified
CMLChronic Myeloid Leukemia
CMMLChronic Myelomonocytic Leukemia
CNLChronic Neutrophilic Leukemia
CNTRLCentriolin
FGFR1Fibroblast growth factor receptor 1
FIPL1FIP1-like 1
FLT3fms-like tyrosine kinase 3
GM-CSFGranulocyte-Macrophage Colony-Stimulating Factor
GUSββ-glucuronidase
HEHypereosinophilia
HESHypereosinophilia syndrome
IL-5Interleukin 5
JAK2janus kinase 2
MDSmyelodysplastic sindromes
MPNmyeloproliferative neoplasms
PDGFRαPlatelet-Derived Growth Factor Receptor alpha
PCM1Periocentriolar material 1
TARCThymus and activation-regulated chemokine
TKTyrosine kinase
TKITyrosine kinase inhibitor
TFRTreatment-free remission
ZMYM2Zinc finger MYM-type protein 2

References

  1. Rothenberg, M.E. Eosinophilia. N. Engl. J. Med. 1998, 338, 1592–1600. [Google Scholar] [CrossRef] [PubMed]
  2. Kovalszki, A.; Weller, P.F. Eosinophilia. Prim. Care 2016, 43, 607–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Acharya, K.R.; Ackerman, S.J. Eosinophil granule proteins: Form and function. J. Biol. Chem. 2014, 289, 17406–17415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dvorak, A.M.; Letourneau, L.; Login, G.R.; Weller, P.F.; Ackerman, S.J. Ultrastructural localization of the Charcot-Leyden crystal protein (lysophospholipase) to a distinct crystalloid-free granule population in mature human eosinophils. Blood 1988, 72, 150–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Giembycz, M.A.; Lindsay, M.A. Pharmacology of the eosinophil. Pharmacol. Rev. 1999, 51, 213–340. [Google Scholar]
  6. Shamri, R.; Xenakis, J.J.; Spencer, L.A. Eosinophils in innate immunity: An evolving story. Cell Tissue Res. 2011, 343, 57–83. [Google Scholar] [CrossRef] [Green Version]
  7. Persson, T.; Andersson, P.; Bodelsson, M.; Laurell, M.; Malm, J.; Egesten, A. Bactericidal activity of human eosinophilic granulocytes against Escherichia coli. Infect. Immun. 2001, 69, 3591–3596. [Google Scholar] [CrossRef] [Green Version]
  8. Sabogal Pineros, Y.S.; Bal, S.M.; Dijkhuis, A.; Majoor, C.J.; Dierdorp, B.S.; Dekker, T.; Hoefsmit, E.P.; Bonta, P.I.; Picavet, D.; van der Wel, N.N.; et al. Eosinophils capture viruses, a capacity that is defective in asthma. Allergy 2019, 74, 1898–1909. [Google Scholar] [CrossRef]
  9. Weller, P.F.; Spencer, L.A. Functions of tissue-resident eosinophils. Nat. Rev. Immunol. 2017, 17, 746–760. [Google Scholar] [CrossRef]
  10. Goh, Y.P.; Henderson, N.C.; Heredia, J.E.; Red Eagle, A.; Odegaard, J.I.; Lehwald, N.; Nguyen, K.D.; Sheppard, D.; Mukundan, L.; Locksley, R.M.; et al. Eosinophils secrete IL-4 to facilitate liver regeneration. Proc. Natl. Acad. Sci. USA 2013, 110, 9914–9919. [Google Scholar] [CrossRef] [Green Version]
  11. Heredia, J.E.; Mukundan, L.; Chen, F.M.; Mueller, A.A.; Deo, R.C.; Locksley, R.M.; Rando, T.A.; Chawla, A. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 2013, 153, 376–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jeziorska, M.; Salamonsen, L.A.; Woolley, D.E. Mast cell and eosinophil distribution and activation in human endometrium throughout the menstrual cycle. Biol. Reprod. 1995, 53, 312–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Choi, H.S.; Won, T.; Hou, X.; Chen, G.; Bracamonte-Baran, W.; Talor, M.V.; Jurcova, I.; Szarszoi, O.; Curnova, L.; Striz, I.; et al. Innate Lymphoid Cells Play a Pathogenic Role in Pericarditis. Cell Rep. 2020, 30, 2989–3003.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Johansson, K.; Malmhall, C.; Ramos-Ramirez, P.; Radinger, M. Bone marrow type 2 innate lymphoid cells: A local source of interleukin-5 in interleukin-33-driven eosinophilia. Immunology 2018, 153, 268–278. [Google Scholar] [CrossRef]
  15. Roufosse, F. Targeting the Interleukin-5 Pathway for Treatment of Eosinophilic Conditions Other than Asthma. Front. Med. 2018, 5, 49. [Google Scholar] [CrossRef] [Green Version]
  16. Dent, L.A.; Strath, M.; Mellor, A.L.; Sanderson, C.J. Eosinophilia in transgenic mice expressing interleukin 5. J. Exp. Med. 1990, 172, 1425–1431. [Google Scholar] [CrossRef] [Green Version]
  17. Esnault, S.; Kelly, E.A. Essential Mechanisms of Differential Activation of Eosinophils by IL-3 Compared to GM-CSF and IL-5. Crit. Rev. Immunol. 2016, 36, 429–444. [Google Scholar] [CrossRef] [Green Version]
  18. Dougan, M.; Dranoff, G.; Dougan, S.K. GM-CSF, IL-3, and IL-5 Family of Cytokines: Regulators of Inflammation. Immunity 2019, 50, 796–811. [Google Scholar] [CrossRef]
  19. McBrien, C.N.; Menzies-Gow, A. The Biology of Eosinophils and Their Role in Asthma. Front. Med. 2017, 4, 93. [Google Scholar] [CrossRef]
  20. O’Sullivan, J.A.; Carroll, D.J.; Bochner, B.S. Glycobiology of Eosinophilic Inflammation: Contributions of Siglecs, Glycans, and Other Glycan-Binding Proteins. Front. Med. 2017, 4, 116. [Google Scholar] [CrossRef] [Green Version]
  21. Legrand, F.; Cao, Y.; Wechsler, J.B.; Zhu, X.; Zimmermann, N.; Rampertaap, S.; Monsale, J.; Romito, K.; Youngblood, B.A.; Brock, E.C.; et al. Sialic acid-binding immunoglobulin-like lectin (Siglec) 8 in patients with eosinophilic disorders: Receptor expression and targeting using chimeric antibodies. J. Allergy Clin. Immunol. 2019, 143, 2227–2237. [Google Scholar] [CrossRef] [PubMed]
  22. Valent, P.; Klion, A.D.; Horny, H.P.; Roufosse, F.; Gotlib, J.; Weller, P.F.; Hellmann, A.; Metzgeroth, G.; Leiferman, K.M.; Arock, M.; et al. Contemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromes. J. Allergy Clin. Immunol. 2012, 130, 607–612. [Google Scholar] [CrossRef] [PubMed]
  23. Shomali, W.; Gotlib, J. World Health Organization-defined eosinophilic disorders: 2019 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2019, 94, 1149–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mattis, D.M.; Wang, S.A.; Lu, C.M. Contemporary Classification and Diagnostic Evaluation of Hypereosinophilia. Am. J. Clin. Pathol. 2020. [Google Scholar] [CrossRef]
  25. Schwaab, J.; Jawhar, M.; Naumann, N.; Schmitt-Graeff, A.; Fabarius, A.; Horny, H.P.; Cross, N.C.; Hofmann, W.K.; Reiter, A.; Metzgeroth, G. Diagnostic challenges in the work up of hypereosinophilia: Pitfalls in bone marrow core biopsy interpretation. Ann. Hematol. 2016, 95, 557–562. [Google Scholar] [CrossRef]
  26. Ackerman, S.J.; Bochner, B.S. Mechanisms of eosinophilia in the pathogenesis of hypereosinophilic disorders. Immunol. Allergy Clin. N. Am. 2007, 27, 357–375. [Google Scholar] [CrossRef] [Green Version]
  27. Lefevre, G.; Copin, M.C.; Staumont-Salle, D.; Avenel-Audran, M.; Aubert, H.; Taieb, A.; Salles, G.; Maisonneuve, H.; Ghomari, K.; Ackerman, F.; et al. The lymphoid variant of hypereosinophilic syndrome: Study of 21 patients with CD3-CD4+ aberrant T-cell phenotype. Medicine 2014, 93, 255–266. [Google Scholar] [CrossRef]
  28. Leru, P.M. Eosinophilic disorders: Evaluation of current classification and diagnostic criteria, proposal of a practical diagnostic algorithm. Clin. Transl. Allergy 2019, 9, 36. [Google Scholar] [CrossRef] [Green Version]
  29. Weller, P.F.; Bubley, G.J. The idiopathic hypereosinophilic syndrome. Blood 1994, 83, 2759–2779. [Google Scholar] [CrossRef] [Green Version]
  30. Swerdlow, S.H.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. In WHO Classification of Tumours, Revised, 4th ed.; IARC Pubblication: Lyon, France, 2017; Volume 2. [Google Scholar]
  31. Dispenza, M.C.; Bochner, B.S. Diagnosis and Novel Approaches to the Treatment of Hypereosinophilic Syndromes. Curr. Hematol. Malig. Rep. 2018, 13, 191–201. [Google Scholar] [CrossRef]
  32. Simon, H.U.; Rothenberg, M.E.; Bochner, B.S.; Weller, P.F.; Wardlaw, A.J.; Wechsler, M.E.; Rosenwasser, L.J.; Roufosse, F.; Gleich, G.J.; Klion, A.D. Refining the definition of hypereosinophilic syndrome. J. Allergy Clin. Immunol. 2010, 126, 45–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Klion, A.D. Eosinophilic myeloproliferative disorders. Hematol. Am. Soc. Hematol. Educ. Program. 2011, 2011, 257–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Havelange, V.; Demoulin, J.B. Review of current classification, molecular alterations, and tyrosine kinase inhibitor therapies in myeloproliferative disorders with hypereosinophilia. J. Blood Med. 2013, 4, 111–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Reiter, A.; Gotlib, J. Myeloid neoplasms with eosinophilia. Blood 2017, 129, 704–714. [Google Scholar] [CrossRef] [Green Version]
  36. Roufosse, F.; Cogan, E.; Goldman, M. Recent advances in pathogenesis and management of hypereosinophilic syndromes. Allergy 2004, 59, 673–689. [Google Scholar] [CrossRef]
  37. Roufosse, F. Hypereosinophilic syndrome variants: Diagnostic and therapeutic considerations. Haematologica 2009, 94, 1188–1193. [Google Scholar] [CrossRef] [Green Version]
  38. de Lavareille, A.; Roufosse, F.; Schmid-Grendelmeier, P.; Roumier, A.S.; Schandene, L.; Cogan, E.; Simon, H.U.; Goldman, M. High serum thymus and activation-regulated chemokine levels in the lymphocytic variant of the hypereosinophilic syndrome. J. Allergy Clin. Immunol. 2002, 110, 476–479. [Google Scholar] [CrossRef]
  39. Szuber, N.; Elliott, M.; Tefferi, A. Chronic neutrophilic leukemia: 2020 update on diagnosis, molecular genetics, prognosis, and management. Am. J. Hematol. 2020, 95, 212–224. [Google Scholar] [CrossRef]
  40. Solary, E.; Itzykson, R. How I treat chronic myelomonocytic leukemia. Blood 2017, 130, 126–136. [Google Scholar] [CrossRef] [Green Version]
  41. Stagno, F.; Vigneri, P.; Del Fabro, V.; Stella, S.; Cupri, A.; Massimino, M.; Consoli, C.; Tambe, L.; Consoli, M.L.; Antolino, A.; et al. Influence of complex variant chromosomal translocations in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors. Acta Oncol. 2010, 49, 506–508. [Google Scholar] [CrossRef]
  42. Vigneri, P.; Stagno, F.; Stella, S.; Cupri, A.; Forte, S.; Massimino, M.; Antolino, A.; Siragusa, S.; Mannina, D.; Impera, S.S.; et al. High BCR-ABL/GUS(IS) Levels at Diagnosis of Chronic Phase CML Are Associated with Unfavorable Responses to Standard-Dose Imatinib. Clin. Cancer Res. 2017, 23, 7189–7198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wang, S.A. The Diagnostic Work-Up of Hypereosinophilia. Pathobiology 2019, 86, 39–52. [Google Scholar] [CrossRef] [PubMed]
  44. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef] [PubMed]
  45. Reilly, J.T. Cytogenetic and molecular genetic aspects of idiopathic myelofibrosis. Acta Haematol. 2002, 108, 113–119. [Google Scholar] [CrossRef]
  46. Yarden, Y.; Escobedo, J.A.; Kuang, W.J.; Yang-Feng, T.L.; Daniel, T.O.; Tremble, P.M.; Chen, E.Y.; Ando, M.E.; Harkins, R.N.; Francke, U.; et al. Structure of the receptor for platelet-derived growth factor helps define a family of closely related growth factor receptors. Nature 1986, 323, 226–232. [Google Scholar] [CrossRef]
  47. Claesson-Welsh, L. Platelet-derived growth factor receptor signals. J. Biol. Chem. 1994, 269, 32023–32026. [Google Scholar] [CrossRef]
  48. Heldin, C.H.; Ostman, A.; Ronnstrand, L. Signal transduction via platelet-derived growth factor receptors. Biochim. Biophys. Acta 1998, 1378, F79–F113. [Google Scholar] [CrossRef]
  49. Valgeirsdottir, S.; Paukku, K.; Silvennoinen, O.; Heldin, C.H.; Claesson-Welsh, L. Activation of Stat5 by platelet-derived growth factor (PDGF) is dependent on phosphorylation sites in PDGF beta-receptor juxtamembrane and kinase insert domains. Oncogene 1998, 16, 505–515. [Google Scholar] [CrossRef] [Green Version]
  50. Sachsenmaier, C.; Sadowski, H.B.; Cooper, J.A. STAT activation by the PDGF receptor requires juxtamembrane phosphorylation sites but not Src tyrosine kinase activation. Oncogene 1999, 18, 3583–3592. [Google Scholar] [CrossRef] [Green Version]
  51. Robyn, J.; Lemery, S.; McCoy, J.P.; Kubofcik, J.; Kim, Y.J.; Pack, S.; Nutman, T.B.; Dunbar, C.; Klion, A.D. Multilineage involvement of the fusion gene in patients with FIP1L1/PDGFRA-positive hypereosinophilic syndrome. Br. J. Haematol. 2006, 132, 286–292. [Google Scholar] [CrossRef]
  52. Cools, J.; DeAngelo, D.J.; Gotlib, J.; Stover, E.H.; Legare, R.D.; Cortes, J.; Kutok, J.; Clark, J.; Galinsky, I.; Griffin, J.D.; et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N. Engl. J. Med. 2003, 348, 1201–1214. [Google Scholar] [CrossRef] [PubMed]
  53. Elling, C.; Erben, P.; Walz, C.; Frickenhaus, M.; Schemionek, M.; Stehling, M.; Serve, H.; Cross, N.C.; Hochhaus, A.; Hofmann, W.K.; et al. Novel imatinib-sensitive PDGFRA-activating point mutations in hypereosinophilic syndrome induce growth factor independence and leukemia-like disease. Blood 2011, 117, 2935–2943. [Google Scholar] [CrossRef] [PubMed]
  54. Maric, I.; Robyn, J.; Metcalfe, D.D.; Fay, M.P.; Carter, M.; Wilson, T.; Fu, W.; Stoddard, J.; Scott, L.; Hartsell, M.; et al. KIT D816V-associated systemic mastocytosis with eosinophilia and FIP1L1/PDGFRA-associated chronic eosinophilic leukemia are distinct entities. J. Allergy Clin. Immunol. 2007, 120, 680–687. [Google Scholar] [CrossRef] [PubMed]
  55. Gotlib, J.; Cools, J. Five years since the discovery of FIP1L1-PDGFRA: What we have learned about the fusion and other molecularly defined eosinophilias. Leukemia 2008, 22, 1999–2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Gotlib, J.; Cross, N.C.; Gilliland, D.G. Eosinophilic disorders: Molecular pathogenesis, new classification, and modern therapy. Best Pract. Res. Clin. Haematol. 2006, 19, 535–569. [Google Scholar] [CrossRef]
  57. Reiter, A.; Walz, C.; Watmore, A.; Schoch, C.; Blau, I.; Schlegelberger, B.; Berger, U.; Telford, N.; Aruliah, S.; Yin, J.A.; et al. The t(8;9)(p22;p24) is a recurrent abnormality in chronic and acute leukemia that fuses PCM1 to JAK2. Cancer Res. 2005, 65, 2662–2667. [Google Scholar] [CrossRef] [Green Version]
  58. Tang, G.; Sydney Sir Philip, J.K.; Weinberg, O.; Tam, W.; Sadigh, S.; Lake, J.I.; Margolskee, E.M.; Rogers, H.J.; Miranda, R.N.; Bueso-Ramos, C.C.; et al. Hematopoietic neoplasms with 9p24/JAK2 rearrangement: A multicenter study. Mod. Pathol. 2019, 32, 490–498. [Google Scholar] [CrossRef]
  59. Thakral, B.; Muzzafar, T.; Wang, S.A.; Medeiros, L.J. Myeloid neoplasm with eosinophilia and BCR-JAK2/t(9;22)(p24;q11.2) morphologically mimicking chronic myeloid leukemia. Ann. Diagn. Pathol. 2020, 44, 151405. [Google Scholar] [CrossRef]
  60. Yu, J.; Deuel, T.F.; Kim, H.R. Platelet-derived growth factor (PDGF) receptor-alpha activates c-Jun NH2-terminal kinase-1 and antagonizes PDGF receptor-beta -induced phenotypic transformation. J. Biol. Chem. 2000, 275, 19076–19082. [Google Scholar] [CrossRef] [Green Version]
  61. Fukushima, K.; Matsumura, I.; Ezoe, S.; Tokunaga, M.; Yasumi, M.; Satoh, Y.; Shibayama, H.; Tanaka, H.; Iwama, A.; Kanakura, Y. FIP1L1-PDGFRalpha imposes eosinophil lineage commitment on hematopoietic stem/progenitor cells. J. Biol. Chem. 2009, 284, 7719–7732. [Google Scholar] [CrossRef] [Green Version]
  62. Buffa, P.; Romano, C.; Pandini, A.; Massimino, M.; Tirro, E.; Di Raimondo, F.; Manzella, L.; Fraternali, F.; Vigneri, P.G. BCR-ABL residues interacting with ponatinib are critical to preserve the tumorigenic potential of the oncoprotein. FASEB J. 2014, 28, 1221–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Jabbour, E.; Kantarjian, H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am. J. Hematol. 2020, 95, 691–709. [Google Scholar] [CrossRef] [PubMed]
  64. Baxter, E.J.; Hochhaus, A.; Bolufer, P.; Reiter, A.; Fernandez, J.M.; Senent, L.; Cervera, J.; Moscardo, F.; Sanz, M.A.; Cross, N.C. The t(4;22)(q12;q11) in atypical chronic myeloid leukaemia fuses BCR to PDGFRA. Hum. Mol. Genet. 2002, 11, 1391–1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Trempat, P.; Villalva, C.; Laurent, G.; Armstrong, F.; Delsol, G.; Dastugue, N.; Brousset, P. Chronic myeloproliferative disorders with rearrangement of the platelet-derived growth factor alpha receptor: A new clinical target for STI571/Glivec. Oncogene 2003, 22, 5702–5706. [Google Scholar] [CrossRef] [Green Version]
  66. Safley, A.M.; Sebastian, S.; Collins, T.S.; Tirado, C.A.; Stenzel, T.T.; Gong, J.Z.; Goodman, B.K. Molecular and cytogenetic characterization of a novel translocation t(4;22) involving the breakpoint cluster region and platelet-derived growth factor receptor-alpha genes in a patient with atypical chronic myeloid leukemia. Genes Chromosomes Cancer 2004, 40, 44–50. [Google Scholar] [CrossRef]
  67. Keene, P.; Mendelow, B.; Pinto, M.R.; Bezwoda, W.; MacDougall, L.; Falkson, G.; Ruff, P.; Bernstein, R. Abnormalities of chromosome 12p13 and malignant proliferation of eosinophils: A nonrandom association. Br. J. Haematol. 1987, 67, 25–31. [Google Scholar] [CrossRef]
  68. Curtis, C.E.; Grand, F.H.; Waghorn, K.; Sahoo, T.P.; George, J.; Cross, N.C. A novel ETV6-PDGFRB fusion transcript missed by standard screening in a patient with an imatinib responsive chronic myeloproliferative disease. Leukemia 2007, 21, 1839–1841. [Google Scholar] [CrossRef]
  69. Toffalini, F.; Hellberg, C.; Demoulin, J.B. Critical role of the platelet-derived growth factor receptor (PDGFR) beta transmembrane domain in the TEL-PDGFRbeta cytosolic oncoprotein. J. Biol. Chem. 2010, 285, 12268–12278. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, S.A.; Hasserjian, R.P.; Tam, W.; Tsai, A.G.; Geyer, J.T.; George, T.I.; Foucar, K.; Rogers, H.J.; Hsi, E.D.; Rea, B.A.; et al. Bone marrow morphology is a strong discriminator between chronic eosinophilic leukemia, not otherwise specified and reactive idiopathic hypereosinophilic syndrome. Haematologica 2017, 102, 1352–1360. [Google Scholar] [CrossRef] [Green Version]
  71. Montano-Almendras, C.P.; Essaghir, A.; Schoemans, H.; Varis, I.; Noel, L.A.; Velghe, A.I.; Latinne, D.; Knoops, L.; Demoulin, J.B. ETV6-PDGFRB and FIP1L1-PDGFRA stimulate human hematopoietic progenitor cell proliferation and differentiation into eosinophils: The role of nuclear factor-kappaB. Haematologica 2012, 97, 1064–1072. [Google Scholar] [CrossRef] [Green Version]
  72. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Iurlo, A.; Cattaneo, D.; Gianelli, U. Hypereosinophilic syndromes in the precision medicine era: Clinical, molecular aspects and therapeutic approaches (targeted therapies). Expert Rev. Hematol. 2019, 12, 1077–1088. [Google Scholar] [CrossRef] [PubMed]
  74. Peiris, M.N.; Meyer, A.N.; Nelson, K.N.; Bisom-Rapp, E.W.; Donoghue, D.J. Oncogenic fusion protein BCR-FGFR1 requires the breakpoint cluster region-mediated oligomerization and chaperonin Hsp90 for activation. Haematologica 2020, 105, 1262–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kulkarni, S.; Reiter, A.; Smedley, D.; Goldman, J.M.; Cross, N.C. The genomic structure of ZNF198 and location of breakpoints in the t(8;13) myeloproliferative syndrome. Genomics 1999, 55, 118–121. [Google Scholar] [CrossRef]
  76. Seif, F.; Khoshmirsafa, M.; Aazami, H.; Mohsenzadegan, M.; Sedighi, G.; Bahar, M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun. Signal. 2017, 15, 23. [Google Scholar] [CrossRef] [Green Version]
  77. Rawlings, J.S.; Rosler, K.M.; Harrison, D.A. The JAK/STAT signaling pathway. J. Cell Sci. 2004, 117, 1281–1283. [Google Scholar] [CrossRef] [Green Version]
  78. Harrison, D.A. The Jak/STAT pathway. Cold Spring Harb. Perspect. Biol. 2012, 4. [Google Scholar] [CrossRef] [Green Version]
  79. Elli, E.M.; Barate, C.; Mendicino, F.; Palandri, F.; Palumbo, G.A. Mechanisms Underlying the Anti-inflammatory and Immunosuppressive Activity of Ruxolitinib. Front. Oncol. 2019, 9, 1186. [Google Scholar] [CrossRef] [Green Version]
  80. Bain, B.J.; Fletcher, S.H. Chronic eosinophilic leukemias and the myeloproliferative variant of the hypereosinophilic syndrome. Immunol. Allergy Clin. N. Am. 2007, 27, 377–388. [Google Scholar] [CrossRef]
  81. Stewart, K.; Carstairs, K.C.; Dube, I.D.; Keating, A. Neutrophilic myelofibrosis presenting as Philadelphia chromosome negative BCR non-rearranged chronic myeloid leukemia. Am. J. Hematol. 1990, 34, 59–63. [Google Scholar] [CrossRef]
  82. Elnaggar, M.M.; Agersborg, S.; Sahoo, T.; Girgin, A.; Ma, W.; Rakkhit, R.; Zorrilla, I.; Leal, A. BCR-JAK2 fusion as a result of a translocation (9;22)(p24;q11.2) in a patient with CML-like myeloproliferative disease. Mol. Cytogenet. 2012, 5, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cook, J.R.; Rogers, H.J.; Chandra, P.K.; Prescott, J.L.; Mukherjee, S. Myeloid neoplasm with eosinophilia and ETV6-JAK2 fusion. Leuk. Lymphoma 2020, 61, 213–216. [Google Scholar] [CrossRef] [PubMed]
  84. Griesinger, F.; Hennig, H.; Hillmer, F.; Podleschny, M.; Steffens, R.; Pies, A.; Wormann, B.; Haase, D.; Bohlander, S.K. A BCR-JAK2 fusion gene as the result of a t(9;22)(p24;q11.2) translocation in a patient with a clinically typical chronic myeloid leukemia. Genes Chromosomes Cancer 2005, 44, 329–333. [Google Scholar] [CrossRef] [PubMed]
  85. He, R.; Greipp, P.T.; Rangan, A.; Mai, M.; Chen, D.; Reichard, K.K.; Nelsen, L.L.; Pardanani, A.; Hanson, C.A.; Viswanatha, D.S. BCR-JAK2 fusion in a myeloproliferative neoplasm with associated eosinophilia. Cancer Genet. 2016, 209, 223–228. [Google Scholar] [CrossRef] [PubMed]
  86. Peeters, P.; Raynaud, S.D.; Cools, J.; Wlodarska, I.; Grosgeorge, J.; Philip, P.; Monpoux, F.; Van Rompaey, L.; Baens, M.; Van den Berghe, H.; et al. Fusion of TEL, the ETS-variant gene 6 (ETV6), to the receptor-associated kinase JAK2 as a result of t(9;12) in a lymphoid and t(9;15;12) in a myeloid leukemia. Blood 1997, 90, 2535–2540. [Google Scholar] [CrossRef] [Green Version]
  87. Bain, B.J.; Ahmad, S. Should myeloid and lymphoid neoplasms with PCM1-JAK2 and other rearrangements of JAK2 be recognized as specific entities? Br. J. Haematol. 2014, 166, 809–817. [Google Scholar] [CrossRef]
  88. Cirmena, G.; Aliano, S.; Fugazza, G.; Bruzzone, R.; Garuti, A.; Bocciardi, R.; Bacigalupo, A.; Ravazzolo, R.; Ballestrero, A.; Sessarego, M. A BCR-JAK2 fusion gene as the result of a t(9;22)(p24;q11) in a patient with acute myeloid leukemia. Cancer Genet. Cytogenet. 2008, 183, 105–108. [Google Scholar] [CrossRef]
  89. Patterer, V.; Schnittger, S.; Kern, W.; Haferlach, T.; Haferlach, C. Hematologic malignancies with PCM1-JAK2 gene fusion share characteristics with myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, and FGFR1. Ann. Hematol. 2013, 92, 759–769. [Google Scholar] [CrossRef]
  90. Schwaller, J. Modeling ETV6-JAK2-induced leukemia: Insights from the zebrafish. Haematologica 2012, 97, 1783–1785. [Google Scholar] [CrossRef]
  91. Cuesta-Dominguez, A.; Ortega, M.; Ormazabal, C.; Santos-Roncero, M.; Galan-Diez, M.; Steegmann, J.L.; Figuera, A.; Arranz, E.; Vizmanos, J.L.; Bueren, J.A.; et al. Transforming and tumorigenic activity of JAK2 by fusion to BCR: Molecular mechanisms of action of a novel BCR-JAK2 tyrosine-kinase. PLoS ONE 2012, 7, e32451. [Google Scholar] [CrossRef] [Green Version]
  92. Falchi, L.; Mehrotra, M.; Newberry, K.J.; Lyle, L.M.; Lu, G.; Patel, K.P.; Luthra, R.; Popat, U.; Verstovsek, S. ETV6-FLT3 fusion gene-positive, eosinophilia-associated myeloproliferative neoplasm successfully treated with sorafenib and allogeneic stem cell transplant. Leukemia 2014, 28, 2090–2092. [Google Scholar] [CrossRef] [Green Version]
  93. Chonabayashi, K.; Hishizawa, M.; Matsui, M.; Kondo, T.; Ohno, T.; Ishikawa, T.; Takaori-Kondo, A. Successful allogeneic stem cell transplantation with long-term remission of ETV6/FLT3-positive myeloid/lymphoid neoplasm with eosinophilia. Ann. Hematol. 2014, 93, 535–537. [Google Scholar] [CrossRef] [PubMed]
  94. Kahn, J.E.; Groh, M.; Lefevre, G. (A Critical Appraisal of) Classification of Hypereosinophilic Disorders. Front. Med. 2017, 4, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Maric, I.; Sun, X. Advances in diagnosis of mastocytosis and hypereosinophilic syndrome. Semin. Hematol. 2019, 56, 22–29. [Google Scholar] [CrossRef] [PubMed]
  96. Helbig, G.; Wieczorkiewicz, A.; Dziaczkowska-Suszek, J.; Majewski, M.; Kyrcz-Krzemien, S. T-cell abnormalities are present at high frequencies in patients with hypereosinophilic syndrome. Haematologica 2009, 94, 1236–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Song, J.; Hussaini, M.; Zhang, H.; Shao, H.; Qin, D.; Zhang, X.; Ma, Z.; Hussnain Naqvi, S.M.; Zhang, L.; Moscinski, L.C. Comparison of the Mutational Profiles of Primary Myelofibrosis, Polycythemia Vera, and Essential Thrombocytosis. Am. J. Clin. Pathol. 2017, 147, 444–452. [Google Scholar] [CrossRef] [Green Version]
  98. Hussaini, M.O.; Mirza, A.S.; Komrokji, R.; Lancet, J.; Padron, E.; Song, J. Genetic Landscape of Acute Myeloid Leukemia Interrogated by Next-generation Sequencing: A Large Cancer Center Experience. Cancer Genom. Proteom. 2018, 15, 121–126. [Google Scholar] [CrossRef] [Green Version]
  99. Palumbo, G.A.; Stella, S.; Pennisi, M.S.; Pirosa, C.; Fermo, E.; Fabris, S.; Cattaneo, D.; Iurlo, A. The Role of New Technologies in Myeloproliferative Neoplasms. Front. Oncol. 2019, 9, 321. [Google Scholar] [CrossRef] [Green Version]
  100. Soverini, S.; Bavaro, L.; De Benedittis, C.; Martelli, M.; Iurlo, A.; Orofino, N.; Sica, S.; Sora, F.; Lunghi, F.; Ciceri, F.; et al. Prospective assessment of NGS-detectable mutations in CML patients with nonoptimal response: The NEXT-in-CML study. Blood 2020, 135, 534–541. [Google Scholar] [CrossRef]
  101. Soverini, S.; Abruzzese, E.; Bocchia, M.; Bonifacio, M.; Galimberti, S.; Gozzini, A.; Iurlo, A.; Luciano, L.; Pregno, P.; Rosti, G.; et al. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: A position paper. J. Hematol. Oncol. 2019, 12, 131. [Google Scholar] [CrossRef] [Green Version]
  102. Baer, C.; Muehlbacher, V.; Kern, W.; Haferlach, C.; Haferlach, T. Molecular genetic characterization of myeloid/lymphoid neoplasms associated with eosinophilia and rearrangement of PDGFRA, PDGFRB, FGFR1 or PCM1-JAK2. Haematologica 2018, 103, e348–e350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Pardanani, A.; Lasho, T.; Wassie, E.; Finke, C.; Zblewski, D.; Hanson, C.A.; Ketterling, R.P.; Gangat, N.; Tefferi, A. Predictors of survival in WHO-defined hypereosinophilic syndrome and idiopathic hypereosinophilia and the role of next-generation sequencing. Leukemia 2016, 30, 1924–1926. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, S.A.; Tam, W.; Tsai, A.G.; Arber, D.A.; Hasserjian, R.P.; Geyer, J.T.; George, T.I.; Czuchlewski, D.R.; Foucar, K.; Rogers, H.J.; et al. Targeted next-generation sequencing identifies a subset of idiopathic hypereosinophilic syndrome with features similar to chronic eosinophilic leukemia, not otherwise specified. Mod. Pathol. 2016, 29, 854–864. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, J.S.; Seo, H.; Im, K.; Park, S.N.; Kim, S.M.; Lee, E.K.; Kim, J.A.; Lee, J.H.; Kwon, S.; Kim, M.; et al. Idiopathic hypereosinophilia is clonal disorder? Clonality identified by targeted sequencing. PLoS ONE 2017, 12, e0185602. [Google Scholar] [CrossRef] [Green Version]
  106. Cross, N.C.P.; Hoade, Y.; Tapper, W.J.; Carreno-Tarragona, G.; Fanelli, T.; Jawhar, M.; Naumann, N.; Pieniak, I.; Lubke, J.; Ali, S.; et al. Recurrent activating STAT5B N642H mutation in myeloid neoplasms with eosinophilia. Leukemia 2019, 33, 415–425. [Google Scholar] [CrossRef] [Green Version]
  107. Ogbogu, P.U.; Bochner, B.S.; Butterfield, J.H.; Gleich, G.J.; Huss-Marp, J.; Kahn, J.E.; Leiferman, K.M.; Nutman, T.B.; Pfab, F.; Ring, J.; et al. Hypereosinophilic syndrome: A multicenter, retrospective analysis of clinical characteristics and response to therapy. J. Allergy Clin. Immunol. 2009, 124, 1319–1325. [Google Scholar] [CrossRef] [Green Version]
  108. Helbig, G.; Wisniewska-Piaty, K.; Francuz, T.; Dziaczkowska-Suszek, J.; Kyrcz-Krzemien, S. Diversity of clinical manifestations and response to corticosteroids for idiopathic hypereosinophilic syndrome: Retrospective study in 33 patients. Leuk. Lymphoma 2013, 54, 807–811. [Google Scholar] [CrossRef]
  109. Fauci, A.S.; Harley, J.B.; Roberts, W.C.; Ferrans, V.J.; Gralnick, H.R.; Bjornson, B.H. NIH conference. The idiopathic hypereosinophilic syndrome. Clinical, pathophysiologic, and therapeutic considerations. Ann. Intern. Med. 1982, 97, 78–92. [Google Scholar] [CrossRef]
  110. Ceretelli, S.; Capochiani, E.; Petrini, M. Interferon-alpha in the idiopathic hypereosinophilic syndrome: Consideration of five cases. Ann. Hematol. 1998, 77, 161–164. [Google Scholar] [CrossRef]
  111. Butterfield, J.H. Interferon treatment for hypereosinophilic syndromes and systemic mastocytosis. Acta Haematol. 2005, 114, 26–40. [Google Scholar] [CrossRef]
  112. Jabbour, E.; Kantarjian, H.; Cortes, J.; Thomas, D.; Garcia-Manero, G.; Ferrajoli, A.; Faderl, S.; Richie, M.A.; Beran, M.; Giles, F.; et al. PEG-IFN-alpha-2b therapy in BCR-ABL-negative myeloproliferative disorders: Final result of a phase 2 study. Cancer 2007, 110, 2012–2018. [Google Scholar] [CrossRef] [PubMed]
  113. Panch, S.R.; Bozik, M.E.; Brown, T.; Makiya, M.; Prussin, C.; Archibald, D.G.; Hebrank, G.T.; Sullivan, M.; Sun, X.; Wetzler, L.; et al. Dexpramipexole as an oral steroid-sparing agent in hypereosinophilic syndromes. Blood 2018, 132, 501–509. [Google Scholar] [CrossRef] [PubMed]
  114. Massimino, M.; Stella, S.; Tirro, E.; Romano, C.; Pennisi, M.S.; Puma, A.; Manzella, L.; Zanghi, A.; Stagno, F.; Di Raimondo, F.; et al. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol. Cancer 2018, 17, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Westerweel, P.E.; Te Boekhorst, P.A.W.; Levin, M.D.; Cornelissen, J.J. New Approaches and Treatment Combinations for the Management of Chronic Myeloid Leukemia. Front. Oncol. 2019, 9, 665. [Google Scholar] [CrossRef]
  116. Stagno, F.; Stella, S.; Spitaleri, A.; Pennisi, M.S.; Di Raimondo, F.; Vigneri, P. Imatinib mesylate in chronic myeloid leukemia: Frontline treatment and long-term outcomes. Expert Rev. Anticancer Ther. 2016, 16, 273–278. [Google Scholar] [CrossRef]
  117. Helbig, G. Imatinib for the treatment of hypereosinophilic syndromes. Expert Rev. Clin. Immunol. 2018, 14, 163–170. [Google Scholar] [CrossRef]
  118. Daniels, C.E.; Wilkes, M.C.; Edens, M.; Kottom, T.J.; Murphy, S.J.; Limper, A.H.; Leof, E.B. Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycin-mediated lung fibrosis. J. Clin. Investig. 2004, 114, 1308–1316. [Google Scholar] [CrossRef]
  119. Wang, S.; Wilkes, M.C.; Leof, E.B.; Hirschberg, R. Imatinib mesylate blocks a non-Smad TGF-beta pathway and reduces renal fibrogenesis in vivo. FASEB J. 2005, 19, 1–11. [Google Scholar] [CrossRef] [Green Version]
  120. Bose, P.; Masarova, L.; Verstovsek, S. Novel treatment strategies for myeloproliferative neoplasms. Rinsho Ketsueki 2019, 60, 1176–1185. [Google Scholar] [CrossRef]
  121. Rumi, E.; Milosevic, J.D.; Selleslag, D.; Casetti, I.; Lierman, E.; Pietra, D.; Cavalloni, C.; Bellini, M.; Milanesi, C.; Dambruoso, I.; et al. Efficacy of ruxolitinib in myeloid neoplasms with PCM1-JAK2 fusion gene. Ann. Hematol. 2015, 94, 1927–1928. [Google Scholar] [CrossRef]
  122. Schwaab, J.; Knut, M.; Haferlach, C.; Metzgeroth, G.; Horny, H.P.; Chase, A.; Tapper, W.; Score, J.; Waghorn, K.; Naumann, N.; et al. Limited duration of complete remission on ruxolitinib in myeloid neoplasms with PCM1-JAK2 and BCR-JAK2 fusion genes. Ann. Hematol. 2015, 94, 233–238. [Google Scholar] [CrossRef] [PubMed]
  123. Walz, C.; Erben, P.; Ritter, M.; Bloor, A.; Metzgeroth, G.; Telford, N.; Haferlach, C.; Haferlach, T.; Gesk, S.; Score, J.; et al. Response of ETV6-FLT3-positive myeloid/lymphoid neoplasm with eosinophilia to inhibitors of FMS-like tyrosine kinase 3. Blood 2011, 118, 2239–2242. [Google Scholar] [CrossRef] [PubMed]
  124. Lierman, E.; Folens, C.; Stover, E.H.; Mentens, N.; Van Miegroet, H.; Scheers, W.; Boogaerts, M.; Vandenberghe, P.; Marynen, P.; Cools, J. Sorafenib is a potent inhibitor of FIP1L1-PDGFRalpha and the imatinib-resistant FIP1L1-PDGFRalpha T674I mutant. Blood 2006, 108, 1374–1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hart, T.K.; Cook, R.M.; Zia-Amirhosseini, P.; Minthorn, E.; Sellers, T.S.; Maleeff, B.E.; Eustis, S.; Schwartz, L.W.; Tsui, P.; Appelbaum, E.R.; et al. Preclinical efficacy and safety of mepolizumab (SB-240563), a humanized monoclonal antibody to IL-5, in cynomolgus monkeys. J. Allergy Clin. Immunol. 2001, 108, 250–257. [Google Scholar] [CrossRef] [PubMed]
  126. Kelly, E.A.; Esnault, S.; Liu, L.Y.; Evans, M.D.; Johansson, M.W.; Mathur, S.; Mosher, D.F.; Denlinger, L.C.; Jarjour, N.N. Mepolizumab Attenuates Airway Eosinophil Numbers, but Not Their Functional Phenotype, in Asthma. Am. J. Respir. Crit. Care Med. 2017, 196, 1385–1395. [Google Scholar] [CrossRef]
  127. Busse, W.W.; Ring, J.; Huss-Marp, J.; Kahn, J.E. A review of treatment with mepolizumab, an anti-IL-5 mAb, in hypereosinophilic syndromes and asthma. J. Allergy Clin. Immunol. 2010, 125, 803–813. [Google Scholar] [CrossRef]
  128. Walsh, G.M. Reslizumab, a humanized anti-IL-5 mAb for the treatment of eosinophil-mediated inflammatory conditions. Curr. Opin. Mol. Ther. 2009, 11, 329–336. [Google Scholar]
  129. Bleecker, E.R.; FitzGerald, J.M.; Chanez, P.; Papi, A.; Weinstein, S.F.; Barker, P.; Sproule, S.; Gilmartin, G.; Aurivillius, M.; Werkstrom, V.; et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta2-agonists (SIROCCO): A randomised, multicentre, placebo-controlled phase 3 trial. Lancet 2016, 388, 2115–2127. [Google Scholar] [CrossRef]
  130. Kuang, F.L.; Legrand, F.; Makiya, M.; Ware, J.; Wetzler, L.; Brown, T.; Magee, T.; Piligian, B.; Yoon, P.; Ellis, J.H.; et al. Benralizumab for PDGFRA-Negative Hypereosinophilic Syndrome. N. Engl. J. Med. 2019, 380, 1336–1346. [Google Scholar] [CrossRef]
  131. Kolbeck, R.; Kozhich, A.; Koike, M.; Peng, L.; Andersson, C.K.; Damschroder, M.M.; Reed, J.L.; Woods, R.; Dall’acqua, W.W.; Stephens, G.L.; et al. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J. Allergy Clin. Immunol. 2010, 125, 1344–1353. [Google Scholar] [CrossRef]
  132. Massanari, M.; Holgate, S.T.; Busse, W.W.; Jimenez, P.; Kianifard, F.; Zeldin, R. Effect of omalizumab on peripheral blood eosinophilia in allergic asthma. Respir. Med. 2010, 104, 188–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Pitini, V.; Teti, D.; Arrigo, C.; Righi, M. Alemtuzumab therapy for refractory idiopathic hypereosinophilic syndrome with abnormal T cells: A case report. Br. J. Haematol. 2004, 127, 477. [Google Scholar] [CrossRef] [PubMed]
  134. Sefcick, A.; Sowter, D.; DasGupta, E.; Russell, N.H.; Byrne, J.L. Alemtuzumab therapy for refractory idiopathic hypereosinophilic syndrome. Br. J. Haematol. 2004, 124, 558–559. [Google Scholar] [CrossRef] [PubMed]
  135. Verstovsek, S.; Tefferi, A.; Kantarjian, H.; Manshouri, T.; Luthra, R.; Pardanani, A.; Quintas-Cardama, A.; Ravandi, F.; Ault, P.; Bueso-Ramos, C.; et al. Alemtuzumab therapy for hypereosinophilic syndrome and chronic eosinophilic leukemia. Clin. Cancer Res. 2009, 15, 368–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Roufosse, F. Management of Hypereosinophilic Syndromes. Immunol. Allergy Clin. N. Am. 2015, 35, 561–575. [Google Scholar] [CrossRef]
  137. Gotlib, J. World Health Organization-defined eosinophilic disorders: 2017 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2017, 92, 1243–1259. [Google Scholar] [CrossRef] [Green Version]
  138. Mehnert, J.M.; Varga, A.; Brose, M.S.; Aggarwal, R.R.; Lin, C.C.; Prawira, A.; de Braud, F.; Tamura, K.; Doi, T.; Piha-Paul, S.A.; et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced, PD-L1-positive papillary or follicular thyroid cancer. BMC Cancer 2019, 19, 196. [Google Scholar] [CrossRef]
  139. Manzella, L.; Massimino, M.; Stella, S.; Tirro, E.; Pennisi, M.S.; Martorana, F.; Motta, G.; Vitale, S.R.; Puma, A.; Romano, C.; et al. Activation of the IGF Axis in Thyroid Cancer: Implications for Tumorigenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 3258. [Google Scholar] [CrossRef] [Green Version]
  140. Brose, M.S.; Bible, K.C.; Chow, L.Q.M.; Gilbert, J.; Grande, C.; Worden, F.; Haddad, R. Management of treatment-related toxicities in advanced medullary thyroid cancer. Cancer Treat. Rev. 2018, 66, 64–73. [Google Scholar] [CrossRef]
  141. Massimino, M.; Tirro, E.; Stella, S.; Pennisi, M.S.; Vitale, S.R.; Puma, A.; Romano, C.; Romeo, M.A.; Gregorio, S.D.; Romeo, M.A.; et al. Targeting BCL-2 as a Therapeutic Strategy for Primary (p210)BCR-ABL1-positive B-ALL Cells. In Vivo 2020, 34, 511–516. [Google Scholar] [CrossRef] [Green Version]
  142. Tirro, E.; Massimino, M.; Romano, C.; Pennisi, M.S.; Stella, S.; Vitale, S.R.; Fidilio, A.; Manzella, L.; Parrinello, N.L.; Stagno, F.; et al. Chk1 Inhibition Restores Inotuzumab Ozogamicin Citotoxicity in CD22-Positive Cells Expressing Mutant p53. Front. Oncol. 2019, 9, 57. [Google Scholar] [CrossRef] [PubMed]
  143. Zanardi, E.; Bregni, G.; de Braud, F.; Di Cosimo, S. Better Together: Targeted Combination Therapies in Breast Cancer. Semin. Oncol. 2015, 42, 887–895. [Google Scholar] [CrossRef] [PubMed]
  144. Lopez, J.S.; Banerji, U. Combine and conquer: Challenges for targeted therapy combinations in early phase trials. Nat. Rev. Clin. Oncol. 2017, 14, 57–66. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Hypereosinophilic syndromes (HES) classification.
Figure 1. Hypereosinophilic syndromes (HES) classification.
Ijms 22 00486 g001
Figure 2. Schematic representation of platelet-derived growth factor receptor alpha (PDGFRα) and PDGFRβ fusion rearrangements. (a) FIP1L1-PDGFRα; (b) breakpoint cluster region-PDGFRα (BCR-PDGFRα); and, (c) ETV6-PDGFRβ. The arrows indicate the position of breakpoints of PDGFRs. BCR = breakpoint cluster region; CC = Coiled-Coil; ETS = (Erythroblast Transformation Specific) DNA-binding domain; ETV = ETS Variant Transcription Factor 1; FIP1L1 = Fip1-like1; GEF = Guanine Nucleotide Exchange Factor; HLH = Helix-Loop-Helix oligomerization domain; NLS = Nuclear Localization Signal; PDGFR = Platelet-Derived Growth Factor Receptor; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; TM = Transmembrane; TK = Tyrosine Kinase.
Figure 2. Schematic representation of platelet-derived growth factor receptor alpha (PDGFRα) and PDGFRβ fusion rearrangements. (a) FIP1L1-PDGFRα; (b) breakpoint cluster region-PDGFRα (BCR-PDGFRα); and, (c) ETV6-PDGFRβ. The arrows indicate the position of breakpoints of PDGFRs. BCR = breakpoint cluster region; CC = Coiled-Coil; ETS = (Erythroblast Transformation Specific) DNA-binding domain; ETV = ETS Variant Transcription Factor 1; FIP1L1 = Fip1-like1; GEF = Guanine Nucleotide Exchange Factor; HLH = Helix-Loop-Helix oligomerization domain; NLS = Nuclear Localization Signal; PDGFR = Platelet-Derived Growth Factor Receptor; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; TM = Transmembrane; TK = Tyrosine Kinase.
Ijms 22 00486 g002
Figure 3. Schematic representation of FGFR1 fusion rearrangements. (a) ZMYM2-FGFR1; (b) BCR-FGFR1; (c) CNTRL-FGFR1. The arrows indicate the position of breakpoints of FGFR1. BCR = breakpoint cluster region; CC = Coiled–Coil; CNTRL = Centriolin; FGFR1 = fibroblast growth factor receptor 1; GEF = Guanine Nucleotide Exchange Factor; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; TK = Tyrosine Kinase; LRR = Leucine-Rich Repeat; ZNF = Zing Finger; ZMYM2 = Zinc finger MYM-type protein 2.
Figure 3. Schematic representation of FGFR1 fusion rearrangements. (a) ZMYM2-FGFR1; (b) BCR-FGFR1; (c) CNTRL-FGFR1. The arrows indicate the position of breakpoints of FGFR1. BCR = breakpoint cluster region; CC = Coiled–Coil; CNTRL = Centriolin; FGFR1 = fibroblast growth factor receptor 1; GEF = Guanine Nucleotide Exchange Factor; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; TK = Tyrosine Kinase; LRR = Leucine-Rich Repeat; ZNF = Zing Finger; ZMYM2 = Zinc finger MYM-type protein 2.
Ijms 22 00486 g003
Figure 4. Schematic representation of JAK2 fusion rearrangements. (a) PCM1-JAK2; (b) BCR-JAK2; and, (c) ETV6-JAK2. The arrows indicate the position of breakpoints of JAK2. BCR = breakpoint cluster region; CC = Coiled Coil; GEF = Guanine Nucleotide Exchange Factor; ETS = (Erythroblast Transformation Specific) DNA-binding domain; HLH = Helix-Loop-Helix oligomerization domain; FERM = 4.1 ezrin, radixin and moesin domain; JAK2 = janus kinase 2; PCM1 = Pericentriolar Material 1; PK = Pseudo kinase domain; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; SH2 = Src-homology-2 domain; TK = Tyrosine Kinase.
Figure 4. Schematic representation of JAK2 fusion rearrangements. (a) PCM1-JAK2; (b) BCR-JAK2; and, (c) ETV6-JAK2. The arrows indicate the position of breakpoints of JAK2. BCR = breakpoint cluster region; CC = Coiled Coil; GEF = Guanine Nucleotide Exchange Factor; ETS = (Erythroblast Transformation Specific) DNA-binding domain; HLH = Helix-Loop-Helix oligomerization domain; FERM = 4.1 ezrin, radixin and moesin domain; JAK2 = janus kinase 2; PCM1 = Pericentriolar Material 1; PK = Pseudo kinase domain; RacGAP = COOH-terminal GTPase Activating Protein (GAP) domain; SH2 = Src-homology-2 domain; TK = Tyrosine Kinase.
Ijms 22 00486 g004
Figure 5. Working model depicting the mechanism of action of pharmacological agents used in hypereosinophilia disorders.Alemtuzumab = anti-CD52 monoclonal antibody; Benralizumab = anti-IL5-R monoclonal antibody; Dexpramipexole, Hydroxyurea and IFNα = cytotoxic agents; Imatinib and Nilotinib = PDGFRs inhibitors; Pegatinib = FGFR1 inhibitor; Mepolizumab, Reslizumab and 610 = recombinant anti-IL5 humanized monoclonal antibody; Omalizumab = anti-IgE monoclonal antibody; Prednisolone = corticosteroid; Ruxolitinib = Jak2 inhibitor; Sorafenib = PDGFRα and FLT3 inhibitor; Venetoclax in combination with Azacitidine and Pevonedistat = chemotherapy regimens.
Figure 5. Working model depicting the mechanism of action of pharmacological agents used in hypereosinophilia disorders.Alemtuzumab = anti-CD52 monoclonal antibody; Benralizumab = anti-IL5-R monoclonal antibody; Dexpramipexole, Hydroxyurea and IFNα = cytotoxic agents; Imatinib and Nilotinib = PDGFRs inhibitors; Pegatinib = FGFR1 inhibitor; Mepolizumab, Reslizumab and 610 = recombinant anti-IL5 humanized monoclonal antibody; Omalizumab = anti-IgE monoclonal antibody; Prednisolone = corticosteroid; Ruxolitinib = Jak2 inhibitor; Sorafenib = PDGFRα and FLT3 inhibitor; Venetoclax in combination with Azacitidine and Pevonedistat = chemotherapy regimens.
Ijms 22 00486 g005
Table 1. Molecular pathogenesis in Hypereosinophilic Syndrome.
Table 1. Molecular pathogenesis in Hypereosinophilic Syndrome.
Fusion Genes
GeneTranslocationGeneTranslocation
PDGFRα PDGFRβ
FIP1L1-PDGFRαdel(4)(q12;q12)ETV6-PDGFRβt(5;12)(q33;p13)
BCR-PDGFRαt(4;22)(q12;q11)
FGFR1 JAK2
ZMYM2-FGFR1t(8;13)(p11.2;q12.1)PCM1-JAK2t(8;9)(p22;p24)
CNTRL-FGFR1t(8;9)(p11.2;q33.2)BCR-JAK2t(9;22)(p24;q11.2)
BCR-FGFR1t(8;22)(p11.2;q11.2)ETV6-JAK2t(9;12)(p24;p13)
Other Genes
ETV6-FLT3t(12;13)(p13;q12)
ETV6-ABL1t(9;12)(q34;p13)
Receptor Rearrangements
T Cell receptor rearrangement
Mutated Genes
GenesPercentage of mutationGenesPercentage of mutation
RUNX183%SETBP122%
ASXL143%CBL14%
TET236%NOTCH114%
EZH229%
Table 2. Therapeutic options for eosinophilic disorder.
Table 2. Therapeutic options for eosinophilic disorder.
Drug Mechanism of ActionDoseTarget Neoplasm
Corticosteroids
PrednisoneSlow and prevent end-organ damage1 mg/kg dailyHES
Cytotoxic agents
Hydroxyurea Inhibit DNA synthesis 500–1000 mg/dailyHES (+ corticosteroids);
Steroid non-responders.
IFNαInhibit cell growth and induct apoptosisInitiation: 1 million units tiw *
Escalation: 3–4 million units tiw *
HES (+ corticosteroids);
HES & CEL, NOS refractory to other therapies; Lymphocyte-variant
hypereosinophilia.
Targeted therapies
TKIs
ImatinibInhibit both TGFb and PDGF-R pathway100–400 mg/dailyPDGFRα rearranged;
PDGRFβ rearranged;
Alternate PDGRFβ fusions;
Selected cases HES and CEL, NOS.
RuxolitinibInhibit dysregulated JAK/STAT signalling pathway20 mg PO BID Eosinophilic leukemia with the PCM1-JAK2 fusion [t(8;9)(p22;p24)].
SorafenibInhibit several kinases involved in both tumour cell proliferation and angiogenesis400 mg/twice dailyFIP1L1-PDGFRα rearranged pts with T674I mutation; FLT3-rearranged cases.
Monoclonal antibodies
Anti-IL-5
MepolizumabInhibit binding of IL-5 to the α chain of the IL-5R100–300 mg every 4 weeksEosinophilic asthma and eosinophilic granulomatosis with polyangiitis.
ReslizumabInhibit the proliferation of eosinophils by binding to the α chain of the IL-5R1 mg/kg Eosinophilic asthma and eosinophilic esophagitis.
Anti-IL-5R
BenralizumabInhibit hetero-oligomerization of α and β subunits of IL-5R 30 mg by subcutaneous injection every 4 weeksSevere asthma.
Anti-IgE
OmalizumabInhibit release of cytokines such as IL-4, IL-5, and IL-13; block unbound IgE.dose/frequency calculated bases on weight per serum IgEEosinophilic disorders, in particular
asthma/nasal polyps
Anti-CD52
AlemtuzumabMediate the lysis of CD52+ cells 5–30 mg 1 to 3 times weeklyRefractory HES pts.
* subcutaneous injection three times a week. IFNα: Interferon alfa; HES: Hypereosinophilic Syndrome; CEL, NOS: Chronic Eosinophilic Leukemia, Not Otherwise Specified; pts: patients; IL5: Interleukin5; IL-5R: Interleukin5 Receptor; PO BID: Orally twice day; PDGF-R: Platelet-Derived Growth Factor Receptor; TGFb: Transforming Growth Factor-b; TKIs: Tyrosine kinases inhibitors.
Table 3. Ongoing clinical trials for eosinophilic disorder.
Table 3. Ongoing clinical trials for eosinophilic disorder.
Drug CombinationTargetDesignPatientsIdentifierPhaseStatus
Corticosteroids
Prednisone--Single Group Assignment; Open Label100NCT01524536Phase IVRecruiting
Dexpramipexole--Non-Randomized; Single Group Assignment; Open Label15NCT02101138 Phase IIUnknown
Targeted therapies
TKIs
ImatinibRuxolitinibFIP1L1-PDGFRα & PDGFRβNon-Randomized; Sequential Assignment; Open Label60NCT00044304Phase IIRecruiting
Nilotinib-FIP1L1-PDGFRα & PDGFRβNPNPNCT04498871NPAvailable
Ruxolitinib-BCR-JAK2 Fusion Protein ExpressionSingle Group Assignment; Open Label25NCT03801434Phase IINot yet recruiting
Monoclonal antibodies
Mepolizumab IL-5NPNPNCT00244686NPAvailable
610 *PlaceboIL-5Randomized; Parallel Assignment52NCT04445038Phase IRecruiting
Benralizumab-IL-5RMulticentre; randomised; double-blind; parallel Assignment120NCT04191304Phase IIINot yet recruiting
PlaceboRandomized; Parallel Assignment22NCT02130882Phase II/IIIActive, not recruiting
Chemotherapy
VenetoclaxAzacitidine, Pevonedistat-Single Group Assignment; Open Label40NCT03862157Phase I/IIRecruiting
IL5: Interleukin5; IL-5R: Interleukin5 Receptor; NP: Not provided. * Recombinant anti-IL5 humanized monoclonal antibodies.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Stella, S.; Massimino, M.; Manzella, L.; Pennisi, M.S.; Tirrò, E.; Romano, C.; Vitale, S.R.; Puma, A.; Tomarchio, C.; Gregorio, S.D.; et al. Molecular Pathogenesis and Treatment Perspectives for Hypereosinophilia and Hypereosinophilic Syndromes. Int. J. Mol. Sci. 2021, 22, 486. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020486

AMA Style

Stella S, Massimino M, Manzella L, Pennisi MS, Tirrò E, Romano C, Vitale SR, Puma A, Tomarchio C, Gregorio SD, et al. Molecular Pathogenesis and Treatment Perspectives for Hypereosinophilia and Hypereosinophilic Syndromes. International Journal of Molecular Sciences. 2021; 22(2):486. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020486

Chicago/Turabian Style

Stella, Stefania, Michele Massimino, Livia Manzella, Maria Stella Pennisi, Elena Tirrò, Chiara Romano, Silvia Rita Vitale, Adriana Puma, Cristina Tomarchio, Sandra Di Gregorio, and et al. 2021. "Molecular Pathogenesis and Treatment Perspectives for Hypereosinophilia and Hypereosinophilic Syndromes" International Journal of Molecular Sciences 22, no. 2: 486. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020486

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop