Abstract
Hypereosinophilic syndromes (HES) are a heterogeneous group of rare disorders defined by the presence of marked peripheral or tissue eosinophilia resulting in end organ damage. Although conventional therapies, including glucocorticoids, hydroxyurea, and interferon-α, are initially effective in reducing eosinophilia and symptoms in a majority of patients with platelet-derived growth factor (PDGFR) mutation-negative HES, development of resistance and treatment-related toxicity are common. In contrast, targeted therapy with the tyrosine kinase inhibitor, imatinib, is well-tolerated but effective only in the subset of HES patients with a primary myeloid disorder. Eosinophil-targeted biotherapeutics offer the potential of improved efficacy with few, if any, adverse effects. The aims of this review are to provide an overview of current approaches to the use of conventional HES therapies and a discussion of existing biotherapeutics that target eosinophils and their potential use in the treatment of HES. With the continuing expansion of eosinophil-targeted biotherapeutics, the future for patients with eosinophilic disorders is promising.
Keywords: Hypereosinophilic Syndrome, Monoclonal Antibody, Interleukin 5
Introduction
Hypereosinophilic syndromes (HES) are rare disorders defined by peripheral eosinophilia greater than 1.5 × 109/L or excessive tissue eosinophilia in the setting of clinical manifestations attributable to the eosinophilia1. Conventional therapies include glucocorticoids (GC), hydroxyurea, interferon-α, imatinib and other commercially available immunomodulatory and suppressive therapies. Although these agents are effective in reducing eosinophilia and controlling symptoms in many patients, resistance and drug-related toxicities lead to discontinuation of therapy in a significant proportion of cases2. Eosinophil-targeted biotherapeutics offer the potential of improved efficacy with few, if any, adverse effects. The past decade has seen an increase in the number of eosinophil-targeted therapies in pre-clinical and clinical trials for eosinophil-associated disorders, including eosinophilic asthma and HES. Although no biologic agents have been approved for use in the treatment of HES to date, two monoclonal antibodies, mepolizumab (Nucala; GlaxoSmithKline) and reslizumab (Cinqair; Teva Pharmaceuticals), that target interleukin-5 (IL-5), the cytokine critical for eosinophil maturation and activation, recently received FDA approval for treatment of eosinophilic asthma. A third monoclonal antibody, benralizumab (AstraZeneca/MedImmune), that depletes eosinophils by binding to IL-5 receptor and targeting the cell for enhanced antibody-dependent cell cytotoxicity, has completed phase 3 trials for eosinophilic asthma and is awaiting FDA approval.
As the number of potential therapies for HES continues to expand, it will become increasingly important to identify factors that may impact treatment choice for individual patients. In this clinical commentary, the overall approach to the diagnosis and treatment of HES will be reviewed with emphasis on the influence of clinical HES subtype on treatment responses. This will be followed by a discussion of the available clinical data regarding the efficacy and safety of existing biotherapeutics for which clinical data is available and the theoretical efficacy of those that are in clinical or pre-clinical trials. For the purposes of this commentary, the scope of biotherapeutics under discussion will be limited to monoclonal antibodies.
Classification of HES
HES is a heterogeneous group of disorders unified by the presence of marked blood or tissue eosinophilia (HE) and clinical manifestations. A detailed description of the diagnostic evaluation of HE and HES is beyond the scope of this review, but has been recently outlined (Table 1;3). The eosinophilia in HES can be primary (due to abnormalities in the myeloid lineage) or secondary (driven by cytokines or other mediators produced by cells other than eosinophils and their precursors). Although the clinical manifestations can be similar irrespective of the cause of the eosinophilia, evidence is mounting to support the classification of patients with HES into clinical subtypes in large part because of the implications for treatment3. For the purposes of this review, HES will be divided into the 6 major clinical variants proposed at a series of multidisciplinary consensus conferences held between 2005 and 20121,4,5: myeloid (M-HES), lymphocytic (L-HES), overlap, associated, familial and idiopathic.
Table 1.
Diagnostic Evaluation of Hypereosinophilia
| Test | Comment |
|---|---|
| All Patients with HE | |
| Compete blood count* | |
| Routine chemistries, including liver function tests* | |
| Quantitative serum immunoglobulin levels, including IgE | |
| Serum troponin*, echocardiogram | If abnormal, consider cardiac MRI as this may show characteristic features of eosinophilic involvement |
| Pulmonary function tests* | |
| Chest/abdomen/pelvis CT* | To assess for splenomegaly, lymphadenopathy, occult neoplasms, as well as pulmonary infiltrates |
| Bone marrow biopsy, including cytogenetics* | Recommended in all patients with AEC > 5.0 × 109/L and features of M-HES or L-HES. |
| Biopsies of affected tissues (if possible)* | |
| Other testing as indicated by history, signs and symptoms | May include parasitic serologies, anti-neutrophil cytoplasmic antibodies, and HIV |
| Serum tryptase and B12 levels | Markedly elevated in myeloid HES. Vitamin B12 levels are elevated due to aberrant B12 binding proteins on granulocytes. |
| FIP1L1/PDGFRA analysis by FISH or RT-PCR | Peripheral blood testing is sufficient |
| T- and B-cell receptor rearrangement studies | |
| Lymphocyte immunophenotyping by flow cytometry* | Minimum of CD3, CD4, CD8 and CD19/20 staining to assess for aberrant lymphocyte populations and B-cell lymphoproliferative disorders |
| Patients with features of M-HES | |
| Additional testing for BCR-ABL1, PDGFRB, JAK2, FGFR1, and KIT mutations by FISH, PCR or other methods as appropriate | Testing should be guided by clinical and/or bone marrow findings |
| Patients with evidence of L-HES | |
| Consider PET scan*, lymph node biopsy* | |
| EBV viral load |
Results substantially affected by GC therapy. Consider obtaining prior to treatment if clinically stable.
Adapted from Klion A “How I treat hypereosinophilic syndromes” in Blood (2015) 126(9) 1069–77.
Myeloid variant HES
M-HES includes patients with definite and presumed primary myeloid disorders presenting as HES. The most common known cause of this variant is an interstitial deletion in chromosome 4q12 leading to the creation of the imatinib-sensitive fusion gene FIP1L1-PDGFRA. Other fusions and point mutations in PDGFRA have also been described. Defined mutations driving HES can also occur in other tyrosine kinases, including PDGFRB, FGFR1, KIT,, and JAK2. Some patients have a clinical phenotype with myeloid features similar to what was described in PDGFRA-positive patients prior to the discovery of FIP1L1-PDGFRA (see Table 2). These patients have idiopathic M-HES. Finally, chronic eosinophilic leukemia-not otherwise specified (CEL-NOS) is also considered a form of M-HES, but will not be discussed since CEL-NOS is typically treatment refractory and most-often requires bone marrow transplantation.
Table 2.
Conventional Treatment Approaches based on HES Clinical Subtypes
| HES Clinical Subtype* | Secondary Classification | First-line therapy | Second-line Agents |
|---|---|---|---|
|
| |||
| Myeloid-variant (11%) | PDGFR-associated MN | Imatinib | Other tyrosine kinase inhibitors with activity against PDGFR |
| IHES with myeloid features (MHES)* | Glucocorticoids | Imatinib | |
| Other myeloid neoplasms presenting as HES | Varied | Varied | |
|
| |||
| Lymphocytic-variant (11%) | GC | Interferon-α Alternatives: other T cell active therapies | |
|
| |||
| Single Organ Overlap (32%) | EGID | Topical GC, diet | Systemic GC |
| EGPA | Systemic GC ± cyclophosphamide | Methotrexate, azathioprine, rituximab | |
| Other | Varied | ||
|
| |||
| Associated (10%) | Treat underlying disease | ||
|
| |||
| Familial (1%) | Observation | ||
|
| |||
| Idiopathic (35%) | Systemic GC | Hydroxyurea or interferon-α | |
Percentages taken from NIH HES cohort (n=415) as of January 2017.
Lymphocytic variant HES
Lymphocytic variant HES (L-HES) is defined by the presence of a clonal or phenotypically aberrant lymphocyte population (most commonly CD3-CD4+) secreting IL-5 or other eosinophil-promoting cytokines. Although intracellular flow cytometry is rarely performed to confirm cytokine secretion, a presumptive diagnosis can be made in the setting of a clonal or aberrant population and features suggestive of this clinical variant, including skin and soft tissue manifestations and an elevated serum IgE level. Occult T cell leukemia/lymphoma can present as L-HES6,7. Conversely, some patients with L-HES progress to lymphoma after many years of stable disease.
Overlap HES
Overlap variants include eosinophilic disease restricted to a single organ system, such as EGID, eosinophilic fasciitis and chronic eosinophilic pneumonia, and multisystem eosinophilic disorders with a distinct clinical phenotype, such as EGPA. Although the etiologies of these disorders are incompletely understood, they can be difficult to distinguish from idiopathic HES (especially in the setting of dramatic peripheral eosinophilia).
Associated HES
Marked peripheral eosinophilia and eosinophilic end organ manifestations (HES) can accompany a wide variety of conditions, including helminth infections, neoplasms, and hypersensitivity disorders. Although the clinical manifestations of these associated HES can be indistinguishable from other forms of HES, treatment is focused on the underlying disorder rather than the eosinophilia itself.
Familial HE/HES
Familial HE/HES is an extremely rare autosomal dominant condition, likely due to dysregulation of IL-5 expression8,9. Since most affected family members remain asymptomatic despite lifelong HE, treatment is generally not indicated. In the event of progression to HES, these patients would need to be classified with respect to clinical phenotype and treated accordingly.
Idiopathic HES
Idiopathic HES is the term used for patients with HES who do not fit into any of the above categories.
General approach to treatment
The overall goals of therapy in all patients with HES are to reduce the absolute eosinophil count (AEC), improve signs and symptoms and prevent progression of disease. Although these three outcomes are linked in most patients, the target AEC can vary depending on the severity of the clinical manifestations and the toxicity of the therapies required to suppress the eosinophilia. Patients with HE and no evidence of clinical manifestations (HEUS and familial HES) typically do not require therapy, but should be followed closely for evidence of progression to HES.
Systemic glucocorticoid (GC) therapy is considered first-line in all patients with HES except: 1) patients with associated HES due to an identified treatable secondary cause (such as a helminth infection, drug hypersensitivity or neoplasm), 2) patients with a known mutation in PDGFRA or PDGFRB, who should be treated with imatinib mesylate (see below), and 3) patients with overlap syndromes, such as eosinophilic gastrointestinal disease (EGID), who may respond to topical GC therapy. In a multicenter retrospective study of 188 patients with HES, GC response rates approached 85% at 1 month2. However, >40% of subjects ultimately discontinued GC therapy due to lack of efficacy, intolerance or other reasons, and the dose required to suppress eosinophilia varied widely. A more recent retrospective study of GC-responsiveness in PDGFR mutation-negative HES demonstrated that clinical subtype was the single best predictor of the minimum GC dose required to suppress eosinophilia and symptoms (Khoury et al. J Allergy Clin Immunol In Pract 2017, in press). Although preliminary, these data suggest that alternative and/or GC-sparing therapies should be considered early in patients with M-HES or L-HES.
Imatinib mesylate is the only FDA-approved drug for the treatment of HES and has radically changed the prognosis of patients with PDGFR-associated disease, especially those with the FIP1L1-PDGFRA fusion. Response rates approach 100% in most series, and recent data suggest that imatinib therapy may be curative in some patients10. Although uncommon, imatinib resistance and severe imatinib toxicity leading to discontinuation of therapy do occur. In this setting, second line tyrosine kinase inhibitors that target PDFGR should be initiated.
As a general rule, addition of a second-line agent, most commonly hydroxyurea, interferon-α or imatinib, should be considered for PDGFR-mutation negative patients with HES who require >10 mg of prednisone daily (or equivalent). Unfortunately, there is little published data to guide selection of a particular agent in this setting. Consequently, the choice of second-line therapy has traditionally been based on a combination of factors, including side effect profiles, ease of administration and patient/physician preferences (Table 2). A recent study of imatinib treatment in PDGFR-mutation negative HES found a 50% response rate in patients with M-HES compared to 0% in those with GC-resistant HES without myeloid features, suggesting that clinical subtype is a useful predictor of imatinib response10. Although data is lacking for other second-line therapies, it would seem reasonable to preferentially select agents that have an effect on T cells, such as interferon-α or cyclosporine, for the treatment for patients with L-HES. A general approach to the use of conventional therapies for HES by clinical subtype is given in Table 2.
Biotherapeutics for HES
Despite the wide variety of commercially available immunomodulatory and cytotoxic agents, a significant proportion of patients with HES are treatment-refractory or experience treatment-related toxicity2. Monoclonal antibodies that target eosinophils, either directly or indirectly by blocking mediators involved in eosinophil production, migration or apoptosis, provide a promising alternative (see recent review)11. Although none of these agents is FDA-approved for HES, several, including alemtuzumab, omalizumab, mepolizumab and reslizumab, have been studied in patients with HES and are approved for other indications.
Alemtuzumab
After two promising case reports of remission with alemtuzumab (anti-CD52) treatment in patients with treatment-refractory HES12,13, alemtuzumab was used to treat 11 subjects with treatment-refractory idiopathic HES or CEL-NOS14. Although 10 subjects achieved complete hematologic remission, 7 subjects experienced relapse with discontinuation of therapy and 3 developed significant complications related to immunosuppression. Reinstitution of therapy was successful in some cases, but sustained remission required long-term administration15. Given the significant side effect profile associated with long-term use of this agent, alemtuzumab is rarely used for HES.
Omalizumab
Omalizumab is FDA-approved for use in asthma and chronic idiopathic urticaria and is well-tolerated in most patients. Consistent with prior studies, pooled analysis of data from 5 placebo controlled studies of omalizumab treatment for moderate to severe asthma (n=2236) demonstrated a significant decrease in AEC in patients who received omalizumab and improved clinical outcomes with reduced AEC regardless of the therapy received16. An open-label study of 9 patients with EGID also showed a significant decrease in AEC and symptomatic improvement with omalizumab therapy, although the reduction in tissue eosinophilia failed to reach statistical significance17. Finally, despite promising results from a small open-label trial of omalizumab treatment for eosinophilic esophagitis (EoE) in which 5/15 subjects demonstrated complete histopathologic remission and improved symptoms18, these findings were not replicated in a placebo-controlled trial19. Of note, response in the open-label trial was associated with a normal AEC. These data suggest that omalizumab may be useful in selected patients with allergen-driven eosinophilic disease, but is unlikely to be useful in the most patients with HES.
Anti-IL-5 (Mepolizumab and Reslizumab)
Mepolizumab and reslizumab are humanized monoclonal antibodies to IL-5, a growth factor crucial for eosinophil maturation and activation, initially developed for use in asthma. After several small clinical trials demonstrated the efficacy of mepolizumab and reslizumab in selected patients with HES20–22, a multicenter, placebo-controlled trial of mepolizumab (750 mg IV monthly) was conducted in 84 GC-responsive patients with PDGFRA-negative HES. Mepolizumab therapy was well-tolerated and had a significant GC-sparing effect with 84% of patients achieving the primary endpoint of ≤10mg prednisone daily for ≥8 consecutive weeks23,24. Of note, 3/7 (86%) patients with L-HES met the primary endpoint compared to 23/24 (96%) of those with a normal T cell profile (p=NS) and a significantly lower proportion of L-HES patients achieved an AEC<600/µL for the duration of the study25, suggesting that high levels of IL-5 production by the T cell clone may decrease drug efficacy. Although baseline IL-5 levels were not predictive of response in the trial, subjects entered the trial with AEC< 1000/µL on prednisone and the majority had undetectable serum IL-5 levels. The GC-sparing effect of mepolizumab was sustained for up to 302 weeks (mean exposure 251 weeks) in a subsequent open-label extension (14). Although both mepolizumab (Nucala®; 100mg sc monthly) and reslizumab (Cinqair®; 3 mg/kg iv monthly) are FDA-approved as add-on maintenance therapy of severe eosinophilic asthma, the approved dosing is lower than that used in any of the trials in HES to date, and treatment of HES is specifically excluded in the package inserts for both drugs.
To date, more than two hundred patients with HES, including prior participants in mepolizumab trials and patients with life-threatening, treatment-refractory HES, have been enrolled in a compassionate use observational study of mepolizumab at doses ranging from 300mg sc to 700mg IV monthly (Duncan et al, ATS 2015 poster; NCT00244686). This larger cohort should provide an opportunity to explore the effects of mepolizumab in GC-responsive and -refractory HES, including dosing and predictors of response. A multi-center placebo-controlled trial of mepolizumab 300mg sc in HES has recently begun enrollment and will also begin to address some of these questions (https://clinicaltrials.gov: NCT02836496).
Mepolizumab has also been used in two clinical trials in patients with EGPA (including probable EGPA/overlap HES). In a pilot open-label study of monthly mepolizumab at 750 mg IV, a GC-sparing effect was observed in all 7 subjects (overall decrease in mean GC dose of 61% after 4 months)26. This prompted a multicenter placebo-controlled trial of mepolizumab (300mg sc monthly) as a GC-sparing agent in 136 patients with EGPA27. Both primary endpoints, total accrued weeks in remission and the proportion of patients in remission at weeks 36 and 48, were met, although 47% of mepolizumab treated EGPA patients never achieved disease remission. The characteristics of these patients have yet to be fully characterized, although possible explanations include contributions of non-eosinophil driven manifestations, incomplete tissue depletion of eosinophils and inadequate dosing.
Both mepolizumab and reslizumab have been used in clinical trials in EGID. In a pilot study in 4 adult patients with treatment-refractory eosinophilic gastritis/enteritis and peripheral eosinophilia, single dose reslizumab (1 mg/kg IV) led to reduction in the AEC and symptom improvement in all 4 subjects28. In a subsequent double-blind placebo controlled trial in 226 children with EoE, reslizumab significantly reduced esophageal eosinophilia compared to placebo at all doses tested (1, 2 and 3 mg/kg IV monthly for 4 months), but did not significantly improve symptoms over placebo29. Similar results were observed with mepolizumab (750 mg or 0.55–10 mg/kg IV monthly) in 3 separate studies in children and adults with EoE30–32.
In summary, high dose mepolizumab (and likely reslizumab) appear to be effective in GC-sensitive HES, including idiopathic HES, L-HES and EGPA/HES Overlap. Data on M-HES is unavailable at this time, although the compassionate use mepolizumab program likely includes patients in this subgroup. The data from studies in EGID suggest that anti-IL-5 therapy may be less effective in EoE than in other forms of HES. Potential reasons for this include incomplete tissue depletion of eosinophils, involvement of cells other than eosinophils in disease pathogenesis and the contribution of structural abnormalities to symptoms.
Other FDA-approved monoclonal antibodies
The utility of other biologic therapies recently approved for the treatment of allergic and inflammatory conditions, including dupilumab (anti-IL4Rα), brodalumab (anti-IL17R), and secukinumab (anti-IL17), in treating HES remains to be seen.
Biotherapeutics on the horizon
Anti-IL-5Rα (Benralizumab)
IL-5 receptor (IL-5R) is expressed on mature eosinophils, mast cells and basophils, and their precursors. It is a heterodimer composed of an α chain (unique to IL-5) and a β chain (common to IL-3R, and GM-CSFR). Benralizumab (anti-IL-5Rα) is an afucosylated monoclonal antibody that binds the IL5Rα chain, recruits natural killer (NK) cells via its Fc chain and causes eosinophil cell death by antibody mediated cytotoxic killing. Like anti-IL-5, it was developed to treat eosinophilic asthma and has been shown in two multi-center placebo-controlled trials to be safe and effective in reducing asthma exacerbations and improving FEV133,34.
There are a number of theoretical advantages and disadvantages of benralizumab therapy over anti-cytokine (IL-5) therapy. Theoretical advantages include lack of interference from high IL-5 levels, targeting eosinophil precursors and complete depletion of tissue-resident eosinophils. Moreover, benralizumab may be effective in M-HES where eosinophils may be IL-5-independent, but retain IL-5R on their surface. Potential disadvantages include the observation that in untreated patients, as AEC increases, IL-5Rα expression decreases and soluble IL-5Rα levels increase35. This could lead to decreased efficacy in patients with the highest AECs (i.e. HES patients). Factors that affect cytotoxic cell function, including CD16 polymorphisms, could also affect drug efficacy. Finally, although not observed in the asthma trials to date, eosinophil lysis in the setting of enhanced killing could complicate therapy in patients with high-grade eosinophilia. A double-blind, placebo-controlled trial of benralizumab for treatment-refractory HES has just completed enrollment (NCT02130882) and should help address some of these questions.
Anti-Siglec-8
Sialic acid-binding immunoglobulin-like lectin 8 (Siglec-8), an inhibitory receptor highly expressed on mature eosinophils, mast cells and basophils, is another promising target36. Crosslinking of Siglec- 8 with anti-Siglec antibodies induces apoptosis of human eosinophils in vitro that is enhanced in the presence of cytokines that normally promote eosinophil survival (including IL-5), providing a theoretical advantage to the use of such antibodies in the treatment of patients with HES36. Siglec-8 antibody also inhibits degranulation of human mast cells37, a potential advantage in the treatment of EGID and other HES subtypes, where tissue mast cells may be pathogenic. Two anti-Siglec-8 antibodies are currently in clinical trials: AK001, an IgG4 antibody expected to induce eosinophil apoptosis, is in Phase 2 trial for moderate to severe nasal polyposis (Clinicaltrials.gov: NCT02734849) and AK002, an afucosylated antibody that both induces eosinophil apoptosis and targets cells expressing Siglec-8 for enhanced ADCC, is in Phase 1 trial for indolent systemic mastocytosis (Clinicaltrials.gov: NCT02808793).
Conclusions
As our understanding of the mechanisms driving eosinophilia in HES increases, the approach to therapy has become increasingly complex. In this regard, novel anti-eosinophil biologics have the potential to be effective across HES subgroups without off-target effects. Conversely, in eosinophilic disorders in which other cells or processes play a role, clinical improvement may be incomplete but provide insight into disease pathogenesis. Although the heterogeneous nature of the clinical presentations of HES has made the development of a uniform assessment tool challenging, complicating the endpoints for clinical trials, the future for patients with eosinophilic-associated diseases is promising as new therapeutics become available to test hypotheses suggested by clinical experience.
Table 3.
Clinical Trials of Biologics for the Treatment of HES*
| HES subtype |
Drug | Design** | Patient population | Dose | Outcome | Reference |
|---|---|---|---|---|---|---|
| PDGFRA-negative HES | mepolizumab | Ph 2 DBPC | Adults with GC-sensitive HES controlled on 20–50 mg prednisone (n=85) | 750 mg iv monthly | Met primary endpoint: proportion of subjects achieving prednisone dose ≤ 10 mg/ day × 8 weeks | 23,25 |
| Ph 2 OL | Adults with GC-sensitive HES controlled on 20–50 mg prednisone (n=78) | 750 mg iv; variable frequency | N/A | 24 | ||
| CUP | Prior participation in clinical trial of mepolizumab for HES OR Refractory, life-threatening HES | 300 mg sc–700 mg iv; variable frequency | Ongoing | NCT00244686 | ||
| Ph 3 DBPC | GC-responsive adults and children ≥12 years old with AEC ≥1000 on stable therapy (estimated n=120) | 300 mg sc monthly | Ongoing | NCT02836496 | ||
| reslizumab | Ph 2 OL | Adults with HES refractory to or intolerant of conventional therapy (n=4) | 1 mg/kg single dose | 2 of 4 subjects had reduction in eosinophilia | 20 | |
| benralizumab | Ph 2 DBPC/OL | Adults with HES and AEC ≥1000 despite background therapy (n=20) | 30mg sc monthly | Recently completed | NCT02130882 | |
| Overlap (EGPA) | mepolizumab | Ph 2 OL | Adults with active disease and on stable prednisolone dose > 12.5 mg for at least one week (n=7) | 750mg IV monthly | Met primary endpoint: percentage of subjects attaining remission | 38 |
| Ph 2 OL | Adults with EGPA requiring ≥10 mg/day prednisone (n=7) | 750mg IV monthly | Met primary endpoint: decreased prednisone dose at the end of treatment phase | 26 | ||
| Ph 3 DBPC | Adults with relapsing probable or definite EGPA requiring ≥ 7.5mg/day prednisone (n=136) | 300mg sc monthly | Met co-primary endpoints: accrued duration of remission and proportion of subjects in remission at week 36 and 48 | 27 | ||
| Reslizumab | Ph 2 OL | Adults with probable or definite EGPA on ≥5 mg/day of prednisone (estimated n=10) | 3 mg/kg monthly | Ongoing | NCT02947945 | |
| Benralizumab | Ph 2 OL | Adults with probable or definite EGPA on ≥5 mg/day of prednisone (estimated n=10) | 30mg sc monthly | Ongoing | NCT03010436 | |
| Rituximab | Ph3 DBPC | Adults with newly diagnosed or relapsing EGPA (estimated n=108) | No details | Ongoing | NCT02807103 | |
| Overlap (EGID) | Mepolizumab | Ph 2/3 OL | Adults with active EoE (peak eos > 20/hpf) (n=4) | 750mg IV monthly × 3 doses | Reduced blood and tissue eosinophils, improved symptoms (n=4) | 32 |
| Ph 2 DBPC | Adults with active EoE (peak eos > 20/hpf) (n=11) | 750mg IV weekly × 2 doses, then monthly × 2 if no response | Reduced blood and tissue eosinophils, no change in symptoms | 31 | ||
| Ph 2 DB | Children with active EoE (peak eos > 20/hpf) (n=59) | 0.55 mg/kg, 2.5 mg/kg, 10 mg/kg IV monthly × 3 doses | Reduction of tissue eosinophils, no significant effect on symptoms | 30 | ||
| Reslizumab | Ph 2 DBPC | Children with active EoE (peak eos> 24/hpf) (n=226) | 1 to 3 mg/kg monthly × 4 doses | Met one co-primary endpoint (reduction of tissue eosinophils), but symptoms improved in both drug and placebo groups | 29 | |
| Omalizumab | Ph 2 OL | Adults with active EoE (peak eos > 15 /hpf) and serum IgE 30–700 IU/mL who failed steroid therapy (n=30) | 0.016 mg/kg/IgE [IU/mL] every 2–4 weeks × 16 weeks | Did not meet primary endpoints: reduction in tissue eosinophilia and dysphagia score | 19 | |
| Dectrecumab (QAX576, anti-IL-13) | Ph 2 DBPC | Symptomatic adult EoE subjects who failed elimination diets (n=23) | 6 mg/kg IV every month × 3 months | Did not meet primary endpoint: proportion with ≥75% reduction in tissue eosinophils | 39 | |
| RPC4046 (anti-IL-13) | Ph 2 DBPC | Adult EoE subjects with symptoms (estimated n=100) | 1st dose IV, followed by low and high dose sc injections weekly × 16 weeks, then open label extension | Recently completed | NCT02098473 | |
| Dupilumab (anti-IL4Rα antagonist) | Ph 2 DBPC/OL | Adult EoE subjects with symptoms (n=47) | No details | Ongoing; enrollment complete | NCT02379052 |
Note: this list is based on a search of ClinicalTrials.gov and EudraCT on August 4, 2017 using the search terms “hypereosinophilic syndrome” or “eosinophilic disorders” and includes only phase 2 and 3 clinical trials.
DB – double-blind, PC - placebo-controlled; OL – open-label, CUP – compassionate use program
Acknowledgments
This work was supported by the Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health
Abbreviations
- AEC
Absolute eosinophil count
- CEL-NOS
chronic eosinophilic leukemia-not otherwise specified
- CT
computer-assisted tomography
- EBV
Epstein-Barr virus
- EGID
Eosinophilic gastrointestinal disorders
- EGPA
Eosinophilic granulomatosis with polyangiitis
- FDA
Food and Drug Administration
- FISH
fluorescence in situ hybridization
- GC
glucocorticoid
- GM-CSF
granulocyte-macrophage stimulating factor
- HE
hypereosinophilia
- HEUS
hypereosinophilia of unknown significance
- HES
hypereosinophilic syndrome
- HIV
human immunodeficiency virus
- IL
interleukin
- IL-5R
IL-5 receptor
- L-HES
lymphocytic variant HES
- M-HES
myeloid HES
- NK
natural killer cell
- PDGFR
platelet-derived growth factor receptor
- PET
positron emission tomography
- RT-PCR
reverse transcriptase polymerase chain reaction
- Siglec
Sialic acid-binding immunoglobulin-like lectin
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Valent P, Klion AD, Horny H-P, Roufosse F, Gotlib J, Weller PF, et al. Contemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromes. J Allergy Clin Immunol. 2012;130:607–12.e9. doi: 10.1016/j.jaci.2012.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ogbogu PU, Bochner BS, Butterfield JH, Gleich GJ, Huss-Marp J, Kahn JE, et al. Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy. J Allergy Clin Immunol. 2009;124:1319–25.e3. doi: 10.1016/j.jaci.2009.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Klion AD. How I treat hypereosinophilic syndromes. Blood. 2015;126:1069–77. doi: 10.1182/blood-2014-11-551614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Klion AD, Bochner BS, Gleich GJ, Nutman TB, Rothenberg ME, Simon H-U, et al. Approaches to the treatment of hypereosinophilic syndromes: a workshop summary report. J Allergy Clin Immunol. 2006;117:1292–302. doi: 10.1016/j.jaci.2006.02.042. [DOI] [PubMed] [Google Scholar]
- 5.Simon H-U, Rothenberg ME, Bochner BS, Weller PF, Wardlaw AJ, Wechsler ME, et al. Refining the definition of hypereosinophilic syndrome. J Allergy Clin Immunol. 2010;126:45–9. doi: 10.1016/j.jaci.2010.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Roufosse F, Garaud S, de Leval L. Lymphoproliferative disorders associated with hypereosinophilia. Semin Hematol. 2012;49:138–48. doi: 10.1053/j.seminhematol.2012.01.003. [DOI] [PubMed] [Google Scholar]
- 7.Jin JJ, Butterfield JH, Weiler CR. Hematologic Malignancies Identified in Patients with Hypereosinophilia and Hypereosinophilic Syndromes. J Allergy Clin Immunol Pract. 2015;3:920–5. doi: 10.1016/j.jaip.2015.06.009. [DOI] [PubMed] [Google Scholar]
- 8.Prakash Babu S, Chen YYK, Bonne-Annee S, Yang J, Maric I, Myers TG, et al. Dysregulation of interleukin 5 expression in familial eosinophilia. Allergy. 2017 doi: 10.1111/all.13146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Klion AD, Law MA, Riemenschneider W, McMaster ML, Brown MR, Horne M, et al. Familial eosinophilia: a benign disorder? Blood. 2004;103:4050–5. doi: 10.1182/blood-2003-11-3850. [DOI] [PubMed] [Google Scholar]
- 10.Khoury P, Desmond R, Pabon A, Holland-Thomas N, Ware JM, Arthur DC, et al. Clinical features predict responsiveness to imatinib in platelet-derived growth factor receptor-alpha-negative hypereosinophilic syndrome. Allergy. 2016;71:803–10. doi: 10.1111/all.12843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Legrand F, Klion AD. Biologic therapies targeting eosinophils: current status and future prospects. J Allergy Clin Immunol Pract. 2015;3:167–74. doi: 10.1016/j.jaip.2015.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.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. doi: 10.1111/j.1365-2141.2004.05206.x. [DOI] [PubMed] [Google Scholar]
- 13.Wagner LA, Speckart S, Cutter B, Gleich GJ. Treatment of FIP1L1/PDGFRA-negative hypereosinophilic syndrome with alemtuzumab, an anti-CD52 antibody. J Allergy Clin Immunol. 2009;123:1407–8. doi: 10.1016/j.jaci.2009.01.069. [DOI] [PubMed] [Google Scholar]
- 14.Verstovsek S, Tefferi A, Kantarjian H, Manshouri T, Luthra R, Pardanani A, et al. Alemtuzumab therapy for hypereosinophilic syndrome and chronic eosinophilic leukemia. Clin Cancer Res. 2009;15:368–73. doi: 10.1158/1078-0432.CCR-08-1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Strati P, Cortes J, Faderl S, Kantarjian H, Verstovsek S. Long-term follow-up of patients with hypereosinophilic syndrome treated with Alemtuzumab, an anti-CD52 antibody. Clin Lymphoma Myeloma Leuk. 2013;13:287–91. doi: 10.1016/j.clml.2012.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Massanari M, Holgate ST, Busse WW, Jimenez P, Kianifard F, Zeldin R. Effect of omalizumab on peripheral blood eosinophilia in allergic asthma. Respir Med. 2010;104:188–96. doi: 10.1016/j.rmed.2009.09.011. [DOI] [PubMed] [Google Scholar]
- 17.Foroughi S, Foster B, Kim N, Bernardino LB, Scott LM, Hamilton RG, et al. Anti-IgE treatment of eosinophil-associated gastrointestinal disorders. J Allergy Clin Immunol. 2007;120:594–601. doi: 10.1016/j.jaci.2007.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Loizou D, Enav B, Komlodi-Pasztor E, Hider P, Kim-Chang J, Noonan L, et al. A pilot study of omalizumab in eosinophilic esophagitis. PLoS ONE. 2015;10:e0113483. doi: 10.1371/journal.pone.0113483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Clayton F, Fang JC, Gleich GJ, Lucendo AJ, Olalla JM, Vinson LA, et al. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology. 2014;147:602–9. doi: 10.1053/j.gastro.2014.05.036. [DOI] [PubMed] [Google Scholar]
- 20.Klion AD, Law MA, Noel P, Kim Y-J, 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;103:2939–41. doi: 10.1182/blood-2003-10-3620. [DOI] [PubMed] [Google Scholar]
- 21.Garrett JK, Jameson SC, Thomson B, Collins MH, Wagoner LE, Freese DK, et al. Anti-interleukin-5 (mepolizumab) therapy for hypereosinophilic syndromes. J Allergy Clin Immunol. 2004;113:115–9. doi: 10.1016/j.jaci.2003.10.049. [DOI] [PubMed] [Google Scholar]
- 22.Plötz S-G, Simon H-U, Darsow U, Simon D, Vassina E, Yousefi S, et al. Use of an anti-interleukin-5 antibody in the hypereosinophilic syndrome with eosinophilic dermatitis. N Engl J Med. 2003;349:2334–9. doi: 10.1056/NEJMoa031261. [DOI] [PubMed] [Google Scholar]
- 23.Rothenberg ME, Klion AD, Roufosse FE, Kahn JE, Weller PF, Simon H-U, et al. Treatment of patients with the hypereosinophilic syndrome with mepolizumab. N Engl J Med. 2008;358:1215–28. doi: 10.1056/NEJMoa070812. [DOI] [PubMed] [Google Scholar]
- 24.Roufosse FE, Kahn J-E, Gleich GJ, Schwartz LB, Singh AD, Rosenwasser LJ, et al. Long-term safety of mepolizumab for the treatment of hypereosinophilic syndromes. J Allergy Clin Immunol. 2013;131:461–7.e1. doi: 10.1016/j.jaci.2012.07.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Roufosse F, de Lavareille A, Schandene L, Cogan E, Georgelas A, Wagner L, et al. Mepolizumab as a corticosteroid-sparing agent in lymphocytic variant hypereosinophilic syndrome. J Allergy Clin Immunol. 2010;126:828–35.e3. doi: 10.1016/j.jaci.2010.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kim S, Marigowda G, Oren E, Israel E, Wechsler ME. Mepolizumab as a steroid-sparing treatment option in patients with Churg-Strauss syndrome. J Allergy Clin Immunol. 2010;125:1336–43. doi: 10.1016/j.jaci.2010.03.028. [DOI] [PubMed] [Google Scholar]
- 27.Wechsler ME, Akuthota P, Jayne D, Khoury P, Klion A, Langford CA, et al. Mepolizumab or Placebo for Eosinophilic Granulomatosis with Polyangiitis. N Engl J Med. 2017;376:1921–32. doi: 10.1056/NEJMoa1702079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kim Y-J, Prussin C, Martin B, Law MA, Haverty TP, Nutman TB, et al. Rebound eosinophilia after treatment of hypereosinophilic syndrome and eosinophilic gastroenteritis with monoclonal anti-IL-5 antibody SCH55700. J Allergy Clin Immunol. 2004;114:1449–55. doi: 10.1016/j.jaci.2004.08.027. [DOI] [PubMed] [Google Scholar]
- 29.Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, Fuchs G, et al. Reslizumab in children and adolescents with eosinophilic esophagitis: results of a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2012;129:456–63. 463.e1. doi: 10.1016/j.jaci.2011.11.044. [DOI] [PubMed] [Google Scholar]
- 30.Assa’ad AH, Gupta SK, Collins MH, Thomson M, Heath AT, Smith DA, et al. An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis. Gastroenterology. 2011;141:1593–604. doi: 10.1053/j.gastro.2011.07.044. [DOI] [PubMed] [Google Scholar]
- 31.Straumann A, Conus S, Grzonka P, Kita H, Kephart G, Bussmann C, et al. Anti-interleukin-5 antibody treatment (mepolizumab) in active eosinophilic oesophagitis: a randomised, placebo-controlled, double-blind trial. Gut. 2010;59:21–30. doi: 10.1136/gut.2009.178558. [DOI] [PubMed] [Google Scholar]
- 32.Stein ML, Collins MH, Villanueva JM, Kushner JP, Putnam PE, Buckmeier BK, et al. Anti- IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol. 2006;118:1312–9. doi: 10.1016/j.jaci.2006.09.007. [DOI] [PubMed] [Google Scholar]
- 33.FitzGerald JM, Bleecker ER, Nair P, Korn S, Ohta K, Lommatzsch M, et al. Benralizumab, an anti-interleukin-5 receptor α monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. The Lancet. 2016;388:2128–41. doi: 10.1016/S0140-6736(16)31322-8. [DOI] [PubMed] [Google Scholar]
- 34.Bleecker ER, FitzGerald JM, Chanez P, Papi A, Weinstein SF, Barker P, et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. The Lancet. 2016;388:2115–27. doi: 10.1016/S0140-6736(16)31324-1. [DOI] [PubMed] [Google Scholar]
- 35.Wilson TM, Maric I, Shukla J, Brown M, Santos C, Simakova O, et al. IL-5 receptor α levels in patients with marked eosinophilia or mastocytosis. J Allergy Clin Immunol. 2011;128:1086–92.e1. doi: 10.1016/j.jaci.2011.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nutku E, Aizawa H, Hudson SA, Bochner BS. Ligation of Siglec-8: a selective mechanism for induction of human eosinophil apoptosis. Blood. 2003;101:5014–20. doi: 10.1182/blood-2002-10-3058. [DOI] [PubMed] [Google Scholar]
- 37.Yokoi H, Choi OH, Hubbard W, Lee H-S, Canning BJ, Lee HH, et al. Inhibition of FcepsilonRI-dependent mediator release and calcium flux from human mast cells by sialic acid-binding immunoglobulin-like lectin 8 engagement. J Allergy Clin Immunol. 2008;121:499–505.e1. doi: 10.1016/j.jaci.2007.10.004. [DOI] [PubMed] [Google Scholar]
- 38.Moosig F, Gross WL, Herrmann K, Bremer JP, Hellmich B. Targeting interleukin-5 in refractory and relapsing Churg-Strauss syndrome. Ann Intern Med. 2011;155:341–3. doi: 10.7326/0003-4819-155-5-201109060-00026. [DOI] [PubMed] [Google Scholar]
- 39.Rothenberg ME, Wen T, Greenberg A, Alpan O, Enav B, Hirano I, et al. Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2015;135:500–7. doi: 10.1016/j.jaci.2014.07.049. [DOI] [PubMed] [Google Scholar]
