Hypereosinophilic syndromes (HESs) are a heterogeneous group of rare disorders characterized by peripheral eosinophilia and eosinophilic end organ complications. Conventional therapies, including glucocorticoids and cytotoxic and immunomodulatory agents, have variable efficacy and significant toxicity. Although the recent development of agents that target eosinophils, including tyrosine kinase inhibitors and monoclonal antibodies, provides the possibility of more effective, less toxic approaches to treatment of HES, there are little available data to guide their use in these conditions. In the following review, the controversies regarding the definition and classification of HES will be discussed, and a pragmatic approach to treatment based on clinically defined HES variants will be presented. An illustrative case will be used to highlight the complexities of treatment selection in HES patients.

Learning Objectives
  • Compare and contrast the most common agents used to treat hypereosinophilic syndrome

  • Discuss recent advances in targeted therapies for eosinophilic disorders

A 50-year-old woman with a history of urticaria, sinusitis with nasal polyps, and eosinophilia presented with dyspnea and palpitations. Absolute eosinophil count (AEC) was 23 400/μL. Echocardiogram showed severe mitral regurgitation, marked bilateral atrial enlargement, and severe pulmonary hypertension. A bone marrow biopsy performed several years earlier was normocellular with increased eosinophils, but there was no evidence of myeloid neoplasia.

Since the first description of eosinophilic endomyocardial fibrosis by Loeffler in 1934,1  the definition and classification of hypereosinophilic syndromes (HESs) have evolved considerably. In 1975, Chusid et al2  described 14 cases of “idiopathic” HES defined by the presence of an AEC ≥1500/μL for 6 months (or death before that time), the absence of a known secondary cause, and evidence of eosinophilic end organ manifestations. The patients in that study included individuals with “Churg’s eosinophilic granulomatosis” (or eosinophilic granulomatosis with polyangiitis [EGPA]), idiopathic eosinophilia with gastrointestinal involvement (eosinophilic gastrointestinal disease), and eosinophilic leukemia, clearly showing the heterogeneity of HES. In the 40 years since their report, more effective therapies have become available, making the 6-month requirement to make a diagnosis increasingly problematic. Moreover, the identification of recurrent molecular abnormalities and secondary causes driving the eosinophilic manifestations in some patients with HES, as defined by Chusid et al,2  has led to controversy regarding the definition and classification of this rare syndrome.

For the purposes of this review, HES will be defined according to a consensus definition developed by a multispecialty group of experts as (1) AEC ≥ 1500/μL and clinical manifestations attributable to eosinophilia or (2) tissue hypereosinophilia with blood eosinophilia (AEC above the upper limit of normal for the reference laboratory).3-5  In contrast to the World Health Organization definition of HES,6  which excludes myeloid neoplasms associated with recurrent molecular abnormalities, the consensus definition includes PDGFRA-associated myeloid neoplasms, eosinophilic endomyocardial fibrosis in the setting of helminth infection (Loeffler’s endocarditis), and other HESs of known cause, because the clinical manifestations of these syndromes can be indistinguishable from those of idiopathic HES.

The same consensus group proposed dividing this heterogeneous group of patients with HES into clinically defined categories in an attempt to capture differences that might reflect the underlying etiology of the eosinophilia and/or predict treatment responses.4  The 6 proposed clinical variants are as follows: (1) myeloid hypereosinophilic syndrome (MHES)—HES with presumed or proven clonal eosinophilia, (2) lymphocytic variant hypereosinophilic syndrome (LHES)—HES with an aberrant or clonal lymphocyte population driving the eosinophilia, (3) overlap HES—eosinophilic disease restricted to a single organ system or defined eosinophilic syndromes that overlap in presentation with idiopathic HES (eg, EGPA), (4) associated HES—HES in the setting of a known secondary cause that requires treatment directed at the cause rather than the eosinophilia itself (eg, helminth infection, drug hypersensitivity, and immunodeficiency) (Table 1), (5) familial HES, and (6) idiopathic HES—HES that does not fall into any of the other categories. Although these categories continue to be refined as technical advances in diagnostic testing and the availability of novel targeted therapies augment our understanding of the underlying pathogenesis of HES, they have proven useful in predicting response to conventional and novel targeted therapies.7-9 

Table 1.

Secondary causes of hypereosinophilia

CategoryExamples (not inclusive)
Allergic disorders* Asthma, atopic dermatitis 
Drug hypersensitivity Varied 
Infection  
 Helminthic Varied, including strongyloidiasis, trichinellosis, filariasis, schistosomiasis, hookworm 
 Ectoparasite Scabies, myiasis 
 Protozoan Isosporiasis, sarcocystis myositis 
 Fungal Coccidiomycosis, allergic bronchopulmonary aspergillosis, histoplasmosis 
 Viral HIV 
Neoplasms Leukemia, lymphoma, adenocarcinoma 
Immunologic disorders  
 Immunodeficiency DOCK8 deficiency, hyper-IgE syndrome, Omenn’s syndrome 
 Autoimmune and idiopathic Sarcoidosis, inflammatory bowel disease, IgG4 disease, and other connective tissue disorders 
Miscellaneous Radiation exposure, cholesterol emboli, hypoadrenalism, IL-2 therapy 
CategoryExamples (not inclusive)
Allergic disorders* Asthma, atopic dermatitis 
Drug hypersensitivity Varied 
Infection  
 Helminthic Varied, including strongyloidiasis, trichinellosis, filariasis, schistosomiasis, hookworm 
 Ectoparasite Scabies, myiasis 
 Protozoan Isosporiasis, sarcocystis myositis 
 Fungal Coccidiomycosis, allergic bronchopulmonary aspergillosis, histoplasmosis 
 Viral HIV 
Neoplasms Leukemia, lymphoma, adenocarcinoma 
Immunologic disorders  
 Immunodeficiency DOCK8 deficiency, hyper-IgE syndrome, Omenn’s syndrome 
 Autoimmune and idiopathic Sarcoidosis, inflammatory bowel disease, IgG4 disease, and other connective tissue disorders 
Miscellaneous Radiation exposure, cholesterol emboli, hypoadrenalism, IL-2 therapy 

IgE, immunoglobulin E; IgG4, immunoglobulin G4; IL-2, interleukin-2. Adapted from Klion4  with permission.

*

Allergic disorders, including asthma and atopic dermatitis, can be associated with AEC ≥ 1500/μL, especially in children, although extremely high eosinophil counts (AEC ≥ 5000/μL) should prompt consideration of another cause. Because allergic manifestations are common in patients with idiopathic HES and LHES, the distinction between allergic disease with marked eosinophilia and HES with concomitant allergic disease may be impossible in some cases.

Drug hypersensitivity can occur in response to any prescription or nonprescription drug or supplement. Although drug-associated eosinophilia can be asymptomatic, well-described syndromes include eosinophilia-myalgia syndrome, drug reaction with eosinophilia and systemic symptoms, interstitial nephritis, and eosinophilic hepatitis.

Hypereosinophilia can occur in the setting of a wide variety of immunologic disorders, particularly those characterized by lymphocyte dysregulation. Clinical sequelae of eosinophilia may or may not be present and can be difficult to distinguish from manifestations of the underlying disorder.

The patient underwent emergency mitral valve replacement with a bovine bioprosthesis and tricuspid repair. The excised valve showed evidence of eosinophilic infiltration and fibrosis. Prednisone (1 mg/kg) was initiated for presumed idiopathic HES.

Both the acuity and nature of the clinical presentation are key in guiding the choice of initial therapy in a patient presenting with an AEC ≥ 1500/μL (Figure 1). Because systemic glucocorticoids (GCs) are the first-line agent for the treatment of most forms of HES, the identification of patients who require alternative therapies is of paramount importance in the initial evaluation. These include patients with (1) HES due to a secondary cause for which specific therapy is available (associated HES) (Table 1); (2) myeloid neoplasms associated with imatinib-sensitive mutations, including FIP1L1-PDGFRA, or known to be GC resistant; and (3) overlap syndromes that can be managed with topical agents, such as eosinophilic esophagitis. Equally important is the decision to treat or observe asymptomatic patients with AEC ≥ 1500/μL and no features of MHES or evidence of end organ manifestations (ie, hypereosinophilia of unknown significance), because biomarkers that predict disease progression are lacking.10,11 

Figure 1.

Initial approach to the treatment of a patient presenting with hypereosinophilia. GC, glucocorticoid; HEUS, hypereosinophilia of unknown significance; N, no; Y, yes.

Figure 1.

Initial approach to the treatment of a patient presenting with hypereosinophilia. GC, glucocorticoid; HEUS, hypereosinophilia of unknown significance; N, no; Y, yes.

Close modal

High-dose GCs (eg, prednisone at 1 mg/kg per day) remain the initial therapy of choice for most patients who present with life-threatening or potentially disabling manifestations of HES. In patients with potential exposure to Strongyloides at any time during their life, empiric ivermectin therapy (200 μg/kg orally daily for 2 days) should be given concomitantly to prevent potentially fatal GC-associated hyperinfection syndrome.12  Intravenous administration of GC should be considered for patients who are acutely ill and/or have gastrointestinal involvement that may impair absorption. Cyclophosphamide should be added if EGPA is suspected based on clinical features, including but not restricted to asthma, sinus disease, and pulmonary infiltrates, and criteria for poor prognosis are met.13 

Patients with known imatinib-sensitive mutations, including FIP1L1-PDGFRA and translocations involving PDGFRB, should receive imatinib mesylate (100-400 mg daily) as first-line therapy. These patients are commonly GC resistant and have a poor prognosis without effective therapy. The response to imatinib therapy in PDGFR-associated myeloid neoplasms is typically rapid (within days) and complete. Because molecular testing can take several days, not all PDGFR mutations are detected using currently available testing, and false negative results can occur, imatinib should also be considered as first-line therapy for patients who present with clinical features of this diagnosis (eg, male gender, splenomegaly, elevated serum B12 and/or tryptase levels, and a hypercellular marrow with dysplastic eosinophils and mast cells) and unknown or negative PDGFR status.7  High-dose GCs should be added for the first few days if there is evidence of cardiac involvement (elevated serum troponin or echocardiographic abnormalities) to prevent myocardial necrosis, a rare complication of imatinib in this patient population.14 

Although every attempt should be made to perform a limited diagnostic evaluation before the administration of GC, therapy should not be delayed for this purpose in the face of worsening signs and symptoms. More stable patients with presumed HES should undergo comprehensive evaluation before initiation of treatment. Initial diagnostic testing should focus on identification of the etiology of the eosinophilia, classification by clinical variant, and delineation of end organ involvement (Table 2). In an acutely ill patient, tests that are affected by GC therapy should be prioritized.

Table 2.

Initial diagnostic testing in the patient with presumed HES

TestComment
All patients with presumed HES  
 Complete blood count*  
 Routine chemistries, including liver function tests*  
 Quantitative serum immunoglobulin levels, including IgE IgE levels are typically elevated in LHES, EGPA and some immunodeficiencies associated with eosinophilia (eg, DOCK8, hyper-IgE syndrome) 
 Serum troponin,* echocardiogram, electrocardiogram If abnormal, cardiac MRI should be considered, because this may show characteristic features of eosinophilic involvement; tissue involvement may be patchy, limiting the utility of biopsy 
 Pulmonary function tests*  
 Chest/abdomen/pelvis CT* To assess for splenomegaly, lymphadenopathy, and occult neoplasms 
 Bone marrow biopsy, including cytogenetics* Recommended in all patients with AEC > 5000/μL and if the clinical picture is suggestive of MHES or LHES; should be considered in other patients 
 Biopsies of affected tissues (if possible) * Eosinophilic infiltration in endomyocardial fibrosis tends to be patchy and occur early in disease, limiting the value of biopsy 
 Other testing for secondary causes As indicated by history and clinical manifestations; may include serology or stool examination for diagnosis of parasitic infection, HIV testing, antineutrophil antibodies (ANCA) 
 Serum tryptase and B12 levels Elevated serum B12 levels are common in all forms of MHES; serum tryptase is typically elevated in the setting of mutations in PDGFRA and KIT 
 FIP1L1/PDGFRA analysis by FISH or RT-PCR Testing of peripheral blood has comparable sensitivity to bone marrow; false negative results can occur, especially with FISH testing 
 T- and B-cell receptor rearrangement studies Clonal patterns are typical (but not diagnostic) of LHES 
 Lymphocyte phenotyping by flow cytometry* At a minimum, CD3, CD4, CD8, and CD19 or CD20 staining should be performed to assess for aberrant CD3−CD4+, CD3+CD4+CD8+, and CD3+CD4−CD8− populations and B-cell lymphoproliferative disorders 
Patients with features of MHES  
 Additional testing for BCR-ABL1, PDGFRB, JAK2, FGFR1, and KIT mutations by PCR, FISH, or other methods as appropriate Testing should be guided by bone marrow findings 
 Myeloid mutation panel testing Consider if BCR-ABL1, PDGFRB, JAK2, FGFR1, and KIT testing is negative 
Patients with evidence of LHES  
 Consider PET scan,* lymph node biopsy* For exclusion of lymphoma 
 EBV viral load  
TestComment
All patients with presumed HES  
 Complete blood count*  
 Routine chemistries, including liver function tests*  
 Quantitative serum immunoglobulin levels, including IgE IgE levels are typically elevated in LHES, EGPA and some immunodeficiencies associated with eosinophilia (eg, DOCK8, hyper-IgE syndrome) 
 Serum troponin,* echocardiogram, electrocardiogram If abnormal, cardiac MRI should be considered, because this may show characteristic features of eosinophilic involvement; tissue involvement may be patchy, limiting the utility of biopsy 
 Pulmonary function tests*  
 Chest/abdomen/pelvis CT* To assess for splenomegaly, lymphadenopathy, and occult neoplasms 
 Bone marrow biopsy, including cytogenetics* Recommended in all patients with AEC > 5000/μL and if the clinical picture is suggestive of MHES or LHES; should be considered in other patients 
 Biopsies of affected tissues (if possible) * Eosinophilic infiltration in endomyocardial fibrosis tends to be patchy and occur early in disease, limiting the value of biopsy 
 Other testing for secondary causes As indicated by history and clinical manifestations; may include serology or stool examination for diagnosis of parasitic infection, HIV testing, antineutrophil antibodies (ANCA) 
 Serum tryptase and B12 levels Elevated serum B12 levels are common in all forms of MHES; serum tryptase is typically elevated in the setting of mutations in PDGFRA and KIT 
 FIP1L1/PDGFRA analysis by FISH or RT-PCR Testing of peripheral blood has comparable sensitivity to bone marrow; false negative results can occur, especially with FISH testing 
 T- and B-cell receptor rearrangement studies Clonal patterns are typical (but not diagnostic) of LHES 
 Lymphocyte phenotyping by flow cytometry* At a minimum, CD3, CD4, CD8, and CD19 or CD20 staining should be performed to assess for aberrant CD3−CD4+, CD3+CD4+CD8+, and CD3+CD4−CD8− populations and B-cell lymphoproliferative disorders 
Patients with features of MHES  
 Additional testing for BCR-ABL1, PDGFRB, JAK2, FGFR1, and KIT mutations by PCR, FISH, or other methods as appropriate Testing should be guided by bone marrow findings 
 Myeloid mutation panel testing Consider if BCR-ABL1, PDGFRB, JAK2, FGFR1, and KIT testing is negative 
Patients with evidence of LHES  
 Consider PET scan,* lymph node biopsy* For exclusion of lymphoma 
 EBV viral load  

ANCA, anti-nuclear cytoplasmic antibody; CT, computed tomography; DOCK8, dedicator of cytokinesis 8; EBV, Epstein-Barr virus; FISH, fluorescence in situ hybridization; IgE, immunoglobulin E; MRI, magnetic resonance imaging; PCR, polymerase chain reaction; PET, positron emission tomography; RT-PCR, reverse transcription polymerase chain reaction; TARC, thymus and activation regulated chemokine.

*

Substantially affected by glucocorticoid therapy.

Not all patients with LHES will have an aberrant T-cell population detectable by flow cytometry or a clonal population identified by PCR. Clinical and laboratory findings suggestive, but not diagnostic, of LHES include skin and soft tissue manifestations, elevated serum IgE, and TARC levels.

The patient’s eosinophilia continued to rise, peaking at 33 700/μL, despite high-dose prednisone therapy (200 mg by mouth daily), and she was hospitalized for congestive heart failure. Echocardiogram revealed stenosis of the bioprosthetic mitral valve. Hydroxyurea (1 g daily) and ultimately, cyclophosphamide (750 mg/m2 monthly) were added. She improved after the addition of cyclophosphamide, with normalization of her eosinophil count. Hydroxyurea was discontinued, and prednisone was tapered. After her second dose of cyclophosphamide, she presented with atrial fibrillation and a left atrial clot and underwent mitral valve annuloplasty <4 months after her initial cardiac surgery. Pathology of the excised valve tissue again showed eosinophilic infiltration. After the second surgery, she was discharged on interferon-α (1 mU subcutaneously daily). One month later, her AEC rose above 1000/μL, and cyclophosphamide was restarted at 70 mg/m2 monthly. Despite this, her AEC remained uncontrolled (1000-6000/μL).

The most common second-line agents for the treatment of GC-resistant, PDGFR mutation–negative HES are imatinib, hydroxyurea, interferon-α, methotrexate, and cyclosporine.15,16  Although the recent Food and Drug Administration approval of 2 anti–interleukin-5 (anti–IL-5) antibodies (mepolizumab and reslizumab) and 1 antibody to IL-5 receptor-α (benralizumab) for eosinophilic asthma is beginning to change this paradigm, only mepolizumab is approved for a variant of HES (EGPA), and cost remains an important factor.

Which second-line agent to use in an individual patient is a complex decision that depends on a number of factors, including anticipated efficacy, side effect profile, and patient and physician preference (Table 3). A brief overview of the most commonly used second-line agents follows. Mepolizumab will be discussed in the next section.

Table 3.

HES therapy

DrugUsual dosingSide effects*Comments
Imatinib 100-400 mg orally Cytopenias, hepatitis, diarrhea, edema, necrotizing myocarditis First line for PDGFR-associated myeloid neoplasms, second line for other forms of MHES 
Prednisone Varied, oral, swallowed, or intravenous Weight gain, osteopenia, diabetes, mood disturbance First line for most PDGFR-negative HES; adjunct for PDGFRA positive with cardiac involvement 
Hydroxyurea 1-2 g/d, oral Cytopenias, diarrhea Second line for idiopathic HES and PDGFRA-negative MHES; low dose may potentiate activity of interferon-α 
Interferon-α 1-3 mU subcutaneously daily or 3 times per week; varied (pegylated) Flu-like symptoms, depression, cytopenias, hypothyroidism, neuropathy, liver toxicity Second line for all forms of HES; preferred second line for LHES 
Methotrexate 7.5-20 mg weekly, orally or subcutaneously Cytopenias, liver toxicity, pneumonitis, desquamative skin rash, encephalopathy, secondary malignancy Alternative second-line agent for EGPA, HES with pulmonary involvement 
Cyclosporine 150 mg daily orally Nephrotoxicity, hypertension, neurotoxicity, secondary malignancy Little data to support use in HES, although anecdotal reports of efficacy in LHES 
DrugUsual dosingSide effects*Comments
Imatinib 100-400 mg orally Cytopenias, hepatitis, diarrhea, edema, necrotizing myocarditis First line for PDGFR-associated myeloid neoplasms, second line for other forms of MHES 
Prednisone Varied, oral, swallowed, or intravenous Weight gain, osteopenia, diabetes, mood disturbance First line for most PDGFR-negative HES; adjunct for PDGFRA positive with cardiac involvement 
Hydroxyurea 1-2 g/d, oral Cytopenias, diarrhea Second line for idiopathic HES and PDGFRA-negative MHES; low dose may potentiate activity of interferon-α 
Interferon-α 1-3 mU subcutaneously daily or 3 times per week; varied (pegylated) Flu-like symptoms, depression, cytopenias, hypothyroidism, neuropathy, liver toxicity Second line for all forms of HES; preferred second line for LHES 
Methotrexate 7.5-20 mg weekly, orally or subcutaneously Cytopenias, liver toxicity, pneumonitis, desquamative skin rash, encephalopathy, secondary malignancy Alternative second-line agent for EGPA, HES with pulmonary involvement 
Cyclosporine 150 mg daily orally Nephrotoxicity, hypertension, neurotoxicity, secondary malignancy Little data to support use in HES, although anecdotal reports of efficacy in LHES 
*

Not inclusive.

Imatinib

Although clearly the drug of choice for PDGFR-associated myeloid neoplasms, the utility of imatinib in patients without a known imatinib-sensitive mutation remains controversial, with efficacy rates ranging from 9% to 60% depending on the series.17-19  This is likely due in part to the heterogeneity of the patients included in these studies. In a recent prospective study of imatinib therapy in FIP1L1-PDGFRA–negative patients with GC-refractory HES, 50% of those with MHES (as defined by ≥4 features suggestive of a primary myeloid disorder) responded to imatinib in contrast to 0% of those without MHES.7  Some PDGFR-negative patients seem to require higher doses of imatinib and respond more slowly.18  Consequently, imatinib (400 mg daily for at least 4 weeks) is recommended. Patients experiencing a suboptimal or partial response should undergo repeat bone marrow examination, because unmasking of pre–B-cell acute lymphocytic leukemia has been reported. Although second generation tyrosine kinase inhibitors, including nilotinib,20  dasatinib, and sorafenib, have shown efficacy in small numbers of patients with imatinib-resistant or -intolerant PDGFRA-associated disease, their effects in mutation-negative HES are unknown.

Hydroxyurea

Hydroxyurea has been used for the treatment of GC-resistant HES since 197821  and is effective in reducing eosinophil counts in a majority of patients, although durable and complete responses are relatively uncommon with monotherapy. Dose-related cytopenias are a major limiting factor.15  Low-dose hydroxyurea (500 mg daily) has been reported to potentiate the effects of interferon-α without increasing toxicity.22 

Interferon-α

First used for the treatment of HES resistant to GC and hydroxyurea, interferon-α has been used as a second-line agent for the treatment of HES for decades.16  In addition to its direct effect on eosinophils, interferon-α downregulates Th2 cytokine production, providing a theoretical advantage for the treatment of LHES. Cytogenetic remissions have also been reported in MHES. Unfortunately, HES is a chronic disease, and drug discontinuation due to the debilitating side effects of interferon-α is common.15  Pegylated interferon, at doses similar to those used for treatment of viral hepatitis, seems to be equally effective and slightly better tolerated.23 

Cyclosporine

Cyclosporine has effects on T-cell activation and cytokine production, and case reports have documented clinical response in patients with LHES, despite persistence of the aberrant clonal T-cell population. In a retrospective series of 188 patients, 11 (6%) were treated with cyclosporine (150-500 mg daily). Although 3 of 5 subjects who received cyclosporine monotherapy had a partial or complete response, the majority of patients (82%) discontinued cyclosporine due to toxicity.

Methotrexate

Although there are little published data directly supporting the use of methotrexate as a GC-sparing agent in HES, it is commonly used in EGPA at relatively low doses (7.5-20 mg weekly) and seems to be as effective as cyclophosphamide in maintaining remission with markedly less toxicity.24  Consequently, methotrexate has been considered an alternative second-line therapy in HES, particularly in patients with pulmonary symptoms or findings suggestive of EGPA. In view of the toxicity profile, which includes black box warnings for rare but potentially fatal toxicities and secondary lymphomas, it is likely that methotrexate use will soon be replaced by mepolizumab (and other targeted therapies) with efficacy in this subgroup of HES patients.

Her eosinophil count remained significantly elevated despite the addition of interferon-α. Approval was obtained for compassionate use mepolizumab (antibody to IL-5; 750 mg intravenously monthly). She resumed monthly cyclophosphamide with only partial suppression of her hypereosinophilia and debilitating side effects, including fatigue, malaise, nausea, and vomiting lasting for 3 to 5 days after each dose. She was subsequently enrolled on a placebo-controlled, double-blind clinical trial of benralizumab (afucosylated antibody to IL-5 receptor-α; 30 mg subcutaneously monthly). At the time of enrollment, her AEC was 6650/μL, despite monthly cyclophosphamide infusions. She showed a dramatic response with AEC 0/μL and no progression of cardiac disease on benralizumab monotherapy for >3 years.

Of the novel biologics recently approved for asthma, the humanized anti–IL-5 antibody mepolizumab (Nucala; GlaxoSmithKline) is the best studied in HES. A double-blind, placebo-controlled phase 2 trial in 85 PDGFRA-negative patients showed that monthly mepolizumab (750 mg intravenously) was safe and effective as a steroid-sparing agent in HES, including LHES.25,26  Long-term safety and efficacy of this dose were confirmed in a second study.27  More recently, mepolizumab (300 mg subcutaneously monthly) was approved for the treatment of EGPA after a double-blind, placebo-controlled phase 3 trial showed a significant reduction in flares with mepolizumab therapy.28  A phase 3 study of mepolizumab (300 mg subcutaneously monthly) to prevent flares in HES is ongoing (NCT02836496). It should be noted that all of the above studies enrolled GC-responsive subjects and used a dose of mepolizumab that is higher than the dose approved for eosinophilic asthma (100 mg subcutaneously monthly). A recent study examining mepolizumab response in a cohort of patients with HES who received the drug as part of a multicenter compassionate use trial suggests that patients with treatment-resistant HES may be less likely to respond to mepolizumab than GC-responsive patients (43% vs 83% complete response, respectively).9  In the same analysis, clinical variant was a useful predictor of response to mepolizumab, with the highest and lowest response rates in patients with overlap HES and MHES, respectively. Although some trial participants were able to reduce mepolizumab dosing in this retrospective trial, 3 of 11 who attempted dose reduction were unsuccessful. Although the monoclonal anti–IL-5 antibody, reslizumab (Cinqair; Teva Pharmaceuticals), is likely to have a similar safety and efficacy profile as mepolizumab, data in HES are limited to a small pilot study in 4 patients.29  Of note, reslizumab is dosed by weight and may provide a theoretical advantage in obese patients.

Benralizumab is an afucosylated antibody to IL-5 receptor-α that depletes eosinophils in the blood, bone marrow, and tissue through enhanced antibody-dependent cell-mediated cytotoxicity. Approved for use in eosinophilic asthma, a recently completed phase 2 trial in HES showed promising results,30  and several additional trials in HES clinical variants are ongoing or planned (NCT03473977 and NCT03010436).

Other agents in clinical development include dexpramipexole (Knopp Inc.) and AK002 (Allakos Inc.). Dexpramipexole is an oral agent with an excellent safety profile that was developed for the treatment of amyotrophic lateral sclerosis and found serendipitously to lower peripheral eosinophil counts.31  In a recent pilot study, dexpramipexole (150 mg orally twice daily) was found to deplete eosinophils in the blood, bone marrow, and tissues of 4 of 10 patients with HES through induction of lineage-specific maturational arrest.32  The drug was well tolerated, and additional studies are planned. AK002 is an afucosylated monoclonal antibody to Siglec-8, an inhibitory receptor expressed on eosinophils, basophils, and mast cells, that is expected to deplete eosinophils in a manner similar to benralizumab. A clinical trial of AK002 for eosinophilic gastritis is currently underway (NCT03496571). Other targeted agents, including JAK inhibitors33  and alemtuzumab,34  have been used in small numbers of patients with LHES with some efficacy, although side effects can be significant and long-term outcomes are lacking. A clinical trial of ruxolitinib for the treatment of GC-refractory HES is ongoing (NCT00044304).

HES is a heterogeneous group of disorders with varied etiologies, clinical manifestations, and prognoses. Although the recent explosion in targeted therapies with improved toxicity profiles has dramatically improved outcomes in some patients with HES, with the exception of imatinib-sensitive mutations, the factors predicting response to a given therapy are still largely unknown. Given the high cost of these new therapies and the challenges in designing and implementing the clinical trials necessary for regulatory approval (and insurance reimbursement) in rare diseases, selection of the right drug for an individual patient is becoming increasingly important. Recent data suggest that clinical variant may be useful in this regard.

This work was funded by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health.

Amy Klion, Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 4 Memorial Dr, Bethesda, MD 20892; e-mail: aklion@niaid.nih.gov.

1.
Hoffman
FG
,
Rosenbaum
D
,
Genovese
PD
.
Fibroplastic endocarditis with eosinophilia (Löffler’s endocarditis parietalis fibroplastica): case report and review of literature
.
Ann Intern Med
.
1955
;
42
(
3
):
668
-
680
.
2.
Chusid
MJ
,
Dale
DC
,
West
BC
,
Wolff
SM
.
The hypereosinophilic syndrome: analysis of fourteen cases with review of the literature
.
Medicine (Baltimore)
.
1975
;
54
(
1
):
1
-
27
.
3.
Valent
P
,
Klion
AD
,
Horny
H-P
, et al
.
Contemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromes
.
J Allergy Clin Immunol
.
2012
;
130
(
3
):
607
-
612.e9
.
4.
Klion
AD
.
How I treat hypereosinophilic syndromes
.
Blood
.
2015
;
126
(
9
):
1069
-
1077
.
5.
Simon
H-U
,
Rothenberg
ME
,
Bochner
BS
, et al
.
Refining the definition of hypereosinophilic syndrome
.
J Allergy Clin Immunol
.
2010
;
126
(
1
):
45
-
49
.
6.
Gotlib
J
.
World Health Organization-defined eosinophilic disorders: 2017 update on diagnosis, risk stratification, and management
.
Am J Hematol
.
2017
;
92
(
11
):
1243
-
1259
.
7.
Khoury
P
,
Desmond
R
,
Pabon
A
, et al
.
Clinical features predict responsiveness to imatinib in platelet-derived growth factor receptor-alpha-negative hypereosinophilic syndrome
.
Allergy
.
2016
;
71
(
6
):
803
-
810
.
8.
Khoury
P
,
Abiodun
AO
,
Holland-Thomas
N
,
Fay
MP
,
Klion
AD
.
Hypereosinophilic syndrome subtype predicts responsiveness to glucocorticoids
.
J Allergy Clin Immunol Pract
.
2018
;
6
(
1
):
190
-
195
.
9.
Kuang
FL
,
Fay
MP
,
Ware
J
, et al
.
Long-term clinical outcomes of high-dose mepolizumab treatment for hypereosinophilic syndrome
.
J Allergy Clin Immunol Pract
.
2018
;
6
(
5
):
1518
-
1527.e5
.
10.
Chen
Y-YK
,
Khoury
P
,
Ware
JM
, et al
.
Marked and persistent eosinophilia in the absence of clinical manifestations
.
J Allergy Clin Immunol
.
2014
;
133
(
4
):
1195
-
1202
.
11.
Helbig
G
,
Hus
M
,
Francuz
T
,
Dziaczkowska-Suszek
J
,
Soja
A
,
Kyrcz-Krzemień
S
.
Characteristics and clinical outcome of patients with hypereosinophilia of undetermined significance
.
Med Oncol
.
2014
;
31
(
1
):
815
.
12.
Mejia
R
,
Nutman
TB
.
Screening, prevention, and treatment for hyperinfection syndrome and disseminated infections caused by Strongyloides stercoralis
.
Curr Opin Infect Dis
.
2012
;
25
(
4
):
458
-
463
.
13.
Mouthon
L
,
Dunogue
B
,
Guillevin
L
.
Diagnosis and classification of eosinophilic granulomatosis with polyangiitis (formerly named Churg-Strauss syndrome) [published correction appears in J Autoimmun. 2014;55:94]
.
J Autoimmun
.
2014
;
48-49
:
99
-
103
.
14.
Pitini
V
,
Arrigo
C
,
Azzarello
D
, et al
.
Serum concentration of cardiac Troponin T in patients with hypereosinophilic syndrome treated with imatinib is predictive of adverse outcomes
.
Blood
.
2003
;
102
(
9
):
3456
-
3457
.
15.
Ogbogu
PU
,
Bochner
BS
,
Butterfield
JH
, et al
.
Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy
.
J Allergy Clin Immunol
.
2009
;
124
(
6
):
1319
-
25.e3
.
16.
Butterfield
JH
,
Weiler
CR
.
Treatment of hypereosinophilic syndromes–the first 100 years
.
Semin Hematol
.
2012
;
49
(
2
):
182
-
191
.
17.
Ogbogu
PU
,
Klion
AD
.
Hypereosinophilic disorders
.
J Allergy Clin Immunol Pract
.
2015
;
3
(
2
):
304
-
305, quiz 306
.
18.
Metzgeroth
G
,
Walz
C
,
Erben
P
, et al
.
Safety and efficacy of imatinib in chronic eosinophilic leukaemia and hypereosinophilic syndrome: a phase-II study
.
Br J Haematol
.
2008
;
143
(
5
):
707
-
715
.
19.
Pardanani
A
,
Brockman
SR
,
Paternoster
SF
, et al
.
FIP1L1-PDGFRA fusion: prevalence and clinicopathologic correlates in 89 consecutive patients with moderate to severe eosinophilia
.
Blood
.
2004
;
104
(
10
):
3038
-
3045
.
20.
Tabouret
E
,
Charbonnier
A
,
Mozziconacci
M-J
,
Ivanov
V
.
Low-dose Nilotinib can maintain complete molecular remissions in FIP1L1/PDGFRA-positive hypereosinophilic syndrome
.
Leuk Res
.
2011
;
35
(
1
):
136
.
21.
Parrillo
JE
,
Fauci
AS
,
Wolff
SM
.
Therapy of the hypereosinophilic syndrome
.
Ann Intern Med
.
1978
;
89
(
2
):
167
-
172
.
22.
Demiroglu
H
,
Dündar
S
.
Combination of interferon-alpha and hydroxyurea in the treatment of idiopathic hypereosinophilic syndrome
.
Br J Haematol
.
1997
;
97
(
4
):
928
-
930
.
23.
Butterfield
JH
,
Weiler
CR
.
Use of pegylated interferon in hypereosinophilic syndrome
.
Leuk Res
.
2012
;
36
(
2
):
192
-
197
.
24.
Maritati
F
,
Alberici
F
,
Oliva
E
, et al
.
Methotrexate versus cyclophosphamide for remission maintenance in ANCA-associated vasculitis: a randomised trial
.
PLoS One
.
2017
;
12
(
10
):
e0185880
.
25.
Rothenberg
ME
,
Klion
AD
,
Roufosse
FE
, et al
;
Mepolizumab HES Study Group
.
Treatment of patients with the hypereosinophilic syndrome with mepolizumab
.
N Engl J Med
.
2008
;
358
(
12
):
1215
-
1228
.
26.
Roufosse
F
,
de Lavareille
A
,
Schandené
L
, et al
.
Mepolizumab as a corticosteroid-sparing agent in lymphocytic variant hypereosinophilic syndrome
.
J Allergy Clin Immunol
.
2010
;
126
(
4
):
828
-
835.e3
.
27.
Roufosse
FE
,
Kahn
J-E
,
Gleich
GJ
, et al
.
Long-term safety of mepolizumab for the treatment of hypereosinophilic syndromes
.
J Allergy Clin Immunol
.
2013
;
131
(
2
):
461
-
467.e1-5
.
28.
Wechsler
ME
,
Akuthota
P
,
Jayne
D
, et al
;
EGPA Mepolizumab Study Team
.
Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis
.
N Engl J Med
.
2017
;
376
(
20
):
1921
-
1932
.
29.
Klion
AD
,
Law
MA
,
Noel
P
,
Kim
YJ
,
Haverty
TP
,
Nutman
TB
.
Safety and efficacy of the monoclonal anti-interleukin-5 antibody SCH55700 in the treatment of patients with hypereosinophilic syndrome
.
Blood
.
2004
;
103
(
8
):
2939
-
2941
.
30.
Kuang
FL
,
Alao
H
,
Kumar
S
, et al
.
Benralizumab (anti-IL5Rα) depletes gut tissue eosinophilia and improves symptoms in hypereosinophilic syndrome with gastrointestinal involvement
.
J Allergy Clin Immunol
.
2018
;
141
(
2
).
Abstract 196
.
31.
Dworetzky
SI
,
Hebrank
GT
,
Archibald
DG
,
Reynolds
IJ
,
Farwell
W
,
Bozik
ME
.
The targeted eosinophil-lowering effects of dexpramipexole in clinical studies
.
Blood Cells Mol Dis
.
2017
;
63
:
62
-
65
.
32.
Panch
SR
,
Bozik
ME
,
Brown
T
, et al
.
Dexpramipexole as an oral steroid-sparing agent in hypereosinophilic syndromes
.
Blood
.
2018
;
132
(
5
):
501
-
509
.
33.
King
B
,
Lee
AI
,
Choi
J
.
Treatment of hypereosinophilic syndrome with cutaneous involvement with the jak inhibitors tofacitinib and ruxolitinib
.
J Invest Dermatol
.
2017
;
137
(
4
):
951
-
954
.
34.
Verstovsek
S
,
Tefferi
A
,
Kantarjian
H
, et al
.
Alemtuzumab therapy for hypereosinophilic syndrome and chronic eosinophilic leukemia
.
Clin Cancer Res
.
2009
;
15
(
1
):
368
-
373
.

Competing Interests

Conflict-of-interest disclosure: The author declares no competing financial interest.

Author notes

Off-label drug use: None disclosed.