Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 28.
Published in final edited form as: Annu Rev Pharmacol Toxicol. 2014 Oct 17;55:633–656. doi: 10.1146/annurev-pharmtox-010814-124407

Novel Targeted Therapies for Eosinophil-Associated Diseases and Allergy

Susanne Radonjic-Hoesli 1, Peter Valent 2, Amy D Klion 3, Michael E Wechsler 4, Hans-Uwe Simon 1
PMCID: PMC4924608  EMSID: EMS68619  PMID: 25340931

Abstract

Eosinophil-associated diseases often present with life-threatening manifestations and/or chronic organ damage. Currently available therapeutic options are limited to a few drugs that often have to be prescribed on a life-long basis to keep eosinophil counts under control. In the last 10 years, treatment options and outcomes in patients with clonal eosinophilic and other eosinophilic disorders have improved substantially. Several new targeted therapies have emerged, addressing different aspects of eosinophil expansion and inflammation. In this review, we discuss available and currently tested agents, as well as new strategies and drug targets relevant to both primary and secondary eosinophilic diseases, including allergic disorders.

Keywords: tyrosine kinase inhibitor, biologicals, asthma, chronic eosinophilic leukemia, hypereosinophilic syndrome

Introduction

Eosinophil-associated diseases include a variety of neoplastic and reactive conditions of different etiologies, including allergic and autoimmune disorders (1), where an elevation of eosinophil levels is the common denominator. The eosinophil is a granulocytic cell that stores highly cytotoxic proteins within its granules. Although the cytotoxic effects of eosinophils and their toxins were first described in the context of parasitic helminth infections (2), they can also be key mediators of tissue damage in inflammation. Normally, eosinophils reside in diverse tissues, such as the gastrointestinal tract (GIT), female genital tract, mammary glands, and thymus (3). Eosinophil infiltration in mucosal tissues is believed to contribute to the maintenance of a barrier against invasive microbes (4). More recently, immunomodulatory functions of eosinophils have increasingly come into focus (5), with murine models demonstrating a role for eosinophils in plasma cell homeostasis in bone marrow (6) and regulation of glucose metabolism in adipose tissue (7).

Eosinophils are generated in the bone marrow from multipotent and lineage-restricted progenitor cells which are themselves constantly replenished from a small pool of self-renewing hematopoietic stem cells (HSC) (8). Essential growth and differentiation factors for eosinophils include interleukin (IL)-5 (9), granulocyte/macrophage colony-stimulating factor (GM-CSF) (10), and IL-3. These cytokines are collectively called eosinopoietins. Whereas GM-CSF and IL-3 are well-described growth factors for other leukocytes, the effects of IL-5 are largely restricted to the eosinophil lineage. IL-5 regulates the proliferation of early progenitor cells, the mobilization of eosinophils from the bone marrow, as well as eosinophil maturation, activation, tissue recruitment, priming, and survival. The receptor for IL-5 shares a common β-chain with the receptors for GM-CSF and IL-3, but contains a specific α-chain (IL-5Rα) (11), expressed almost exclusively on eosinophils, basophils, mast cells, and their progenitors (12).

Eosinophilia is the result of an imbalance between eosinopoiesis, eosinophil clearance, and eosinophil death (13, 14). This may be due to intrinsic, eosinophil-related causes (e.g. by mutations in eosinophil precursor cells) or extrinsic causes (e.g. by enhanced production of eosinopoietins) (1). If blood eosinophil levels exceed 1.5 × 106/l, the condition is referred to as hypereosinophilia (HE) (1517). In the absence of any of the common triggers of eosinophilia (allergens, parasites, drugs), the term hypereosinophilic syndrome (HES) is appropriate if HE is documented over a period of at least 4 weeks and is accompanied by eosinophil-mediated organ damage (16).

A number of different HES classifications have been proposed, attempting to identify subgroups of patients who may respond similarly to particular therapies (1, 1517). It is useful to distinguish between patients with an underlying primary or clonal process and those with secondary eosinophilia. Patients with primary or clonal HE suffer from a myeloid or stem cell-derived neoplasm, i.e. eosinophils belong to the malignant clone. The FIP1-like 1 (FIP1L1) - platelet-derived growth factor receptor alpha (PDGFRA) fusion gene is the most frequent recurrent aberration in clonal HE and is detected in 30-50% of all cases (18). However, HES may also occur in the setting of other myeloid neoplasms accompanied by clonal HE (1, 1517).

Secondary HES variants are mediated by production of one or several eosinopoietins, e.g. by normal/reactive (activated) T cells, clonal T cells, or other tumor cells (1517). Both CD4+ and CD8+ T cells have been identified as eosinopoietin-producers (19). When eosinopoietin-producing T cells drive HE, the term lymphocytic HES (LHES) is appropriate (1, 1517). In many patients with LHES, expansion of a T cell clone can be identified (1, 1517, 20). In a subset of these patients, overt Non-Hodgkin’s lymphoma (NHL) may eventually develop (21).

The eosinophilia or HE observed in the setting of eosinophilic allergic disorders is typically mediated by eosinopoietin-producing T cells (1). Furthermore, the clinical manifestations of these disorders overlap with those of HESs. Although therapeutic approaches to HESs and eosinophilic allergic disorders have historically differed, the availability of novel targeted therapies and a better understanding of the pathogenesis of HE and HES variants now allow a more structured approach (1, 1517). In this review, we discuss targeted therapeutic options currently being investigated for primary and secondary eosinophilic diseases, including allergic disorders.

Clonal Eosinophilic Disorders

Somatic mutations of certain genes involved in proliferation and survival of eosinophil progenitor cells can result in clonal HE and/or a primary (clonal) HES. In recent years, a number of molecular defects have been identified in patients with clonal eosinophilic disorders, the most common being the FIP1L1-PDGFRA gene fusion (22). The FIP1L1-PDGFRA fusion results in constitutive, ligand-independent PDGFRA tyrosine kinase activity (22). Interestingly, the oncogenic potential of the FIP1L1-PDGFRA mutant can be enhanced by escape of the fusion product from normal protein degradation processes, leading to its accumulation (23).

Other, fusion genes involving PDGFRα or PDGFRβ can also cause clonal HE or HES (22). Most result in a constitutively active tyrosine kinase receptor that acts as oncogenic driver. Rarely, clonal HE or HES is caused by a chromosomal translocation involving the fibroblast growth factor receptor 1 (FGFR1) gene on chromosome 8p11-12, the so-called “8p11 syndrome” (24). This syndrome typically has an aggressive course with primary multilineage involvement and acute leukemia of mostly myeloid or mixed lineage in the terminal phase. As these patients are usually treatment-resistant, their prognosis is poor (24). Finally, clonal eosinophilia has been described in D816V KIT positive systemic mastocytosis (25) and in association with cytogenetic abnormalities, including PCM1-JAK2 (26). From a therapeutic standpoint, this is important to recognize since these genetic abnormalities do not respond to imatinib and require alternative approaches.

Tyrosine Kinase-Targeting Drugs

Imatinib

Patients with clonal eosinophilia typically do not have a sustained response to glucocorticosteroid therapies. Imatinib was originally designed to target the fusion oncogene, BCR/ABL, in chronic myeloid leukemia (CML) (27). The FIP1L1-PDGFRA kinase is 200-fold more sensitive to imatinib than BCR/ABL (28) and imatinib (100-400 mg/d) is first-line therapy for patients with PDGFR-associated disease (17). Clinical and hematological responses are rapid and dramatic (29) with molecular remission (no detectable FIP1L1-PDGFRA) typically observed within 2-3 months (30). Although imatinib is generally well-tolerated, myocardial necrosis has been reported in patients with eosinophilic cardiac involvement. Thus, in patients with elevated serum troponin levels or echocardiographic evidence of endomyocardial fibrosis, concomitant glucocorticosteroid therapy is recommended with imatinib initially to reduce this risk. Imatinib is not curative in the majority of cases (30, 31) and life-long therapy is recommended.

Though rare, if no hematological response is observed within 4 weeks, primary resistance should be considered (32). On a molecular basis, primary resistance to imatinib has been linked to the occurrence of a S601P mutation in PDGFRA, which leads to destabilization of the inactive conformation of the kinase domain binding imatinib (33). Acquired resistance to imatinib also appears to be uncommon. Most cases have been in association with the T674I mutation of FIP1L1-PDGFRA, a single base substitution in the imatinib-binding sequence analogous to the T315I mutation in BCR/ABL, which also promotes resistance to imatinib and related TKIs (28).

Second- and third-generation TKIs

Second-generation TKIs were developed to address the unfavorable outcome due to imatinib intolerance or resistance observed in approximately 30% of CML patients (34, 35). Indeed, higher response rates and less resistance were found in CML patients receiving nilotinib, dasatinib, or bosutinib in clinical trials (3638). However, although sorafenib and nilotinib show in vitro efficacy against T315I BCR/ABL and T674I FIP1L1-PDGFRA, their clinical success with the T674I FIP1L1-PDGFRA resistance mutation has been disappointing (39, 40). Bosutinib is largely ineffective as this BCR/ABL blocker does not recognize PDGFRs (41). Therefore, the treatment of choice for such patients remains allogeneic stem cell transplantation (40).

Newer TKIs, such as ponatinib (42) and DCC-2036 (43), third-generation BCR/ABL blockers that are effective in vitro against the T674I-mutation in FIP1L1-PDGFRA are in development. Third-generation TKIs, including gefitinib (44), midostaurin (45), and ponatinib (46) have also shown in vitro efficacy against FGFR1 fusion genes, and may prove effective in PDGFR negative clonal eosinophilic disorders, such as 8p11 syndrome. In one patient with a ZNF198-FGFR1 gene fusion, a stable 6-month remission was obtained upon administration of midostaurin; increased maturation of myeloid cells and normalization of the myeloid-to-erythroid ratio in the bone marrow was achieved within 12 weeks (45).

It is important to recognize that, unlike imatinib, which is extremely well-tolerated, these novel TKIs can induce clinically relevant side effects. For instance, in CML patients treated with nilotinib and ponatinib, an increased rate of severe peripheral and central arterial occlusive diseases, leading to infarction, amputations, and disability, have been described (47, 48). In 2013, ponatinib was even taken off the market because of these side effects.

Interestingly, most of the currently available TKI, including imatinib, midostaurin, nilotinib, and ponatinib, but not bosutinib, recognize and block wild-type KIT tyrosine kinase activity (49). This finding is important, because leukemic stem cells (LSC) utilize this stem cell kinase as a critical mediator of survival and proliferation, suggesting that combination therapy using more toxic TKIs with enhanced activity against LSC to attack the vascular stem cell niche in an early (induction) phase, followed by maintenance therapy with imatinib, might be used in the future for patients with aggressive disease. It should be noted that midostaurin and ponatinib (50), but not imatinib and nilotinib (51), are effective against D816V, the most common KIT mutation in patients with systemic mastocytosis.

Clearly, the development of resistance, the requirement for life-long administration of imatinib, and the toxicity of the second- and third-generation TKIs remain clinical problems. In the following sections, we discuss new strategies that may help overcome these limitations in treatment of clonal eosinophilic disorders (Figure 1).

Figure 1. Potential novel targeted-treatment strategies for clonal eosinophilic disorders.

Figure 1

The development of clonal or primary HES can often be linked to the presence of a constitutively active stem cell kinase, such as the FIP1L1-PDGFRA oncoprotein. Imatinib is a highly effective inhibitor of FIP1L1-PDGFRA and remains first-line treatment in patients with this fusion gene. However, resistance may occur, often in association with secondary mutations in FIP1L1-PDGFRA, and discontinuation of imatinib in remission leads to reoccurrence of the disease, probably because of persisting LSC. Besides new, more effective TKIs, other promising novel therapeutic strategies for patients with clonal or primary HES are presented below:
  1. Antibodies targeting eosinophil-specific and LSC surface receptors. Although IL-5 is the most important eosinopoietin, clonal eosinophils are known to grow independently of IL-5. Consequently, anti-IL-5 antibodies are not likely to be effective in most cases. Whether targeting the IL-5Rα will elicit better responses is unknown. The immunophenotype of the clonal eosinophil LSC is currently unknown, but based on other leukemic models, it seems unlikely that these cells express IL-5 receptors. Specific antibodies to LSC surface molecules, alone or in combination with imatinib, may be a promising strategy for eliminating LSC and thus achieving cures in patients with clonal eosinophilic disorders. Please note that imatinib as well as the second-generation TKIs are also potent inhibitors of the wild-type stem cell kinase KIT.
  2. Restoration of protein degradation. Oncogenic enhancement is further supported by an escape of the fusion protein from protein degradation. Therefore, promoting ubiquitination, thus targeting the fusion protein for proteasomal degradation, might augment TKI effects.
  3. Inhibition of autophagy. Autophagy is a survival mechanism and is known to be highly active in adult stem cells. Targeting autophagy in cell lines and primary CML cells significantly increased cell death. In addition, TKI have been identified as activators of autophagy, a mechanism, which can contribute to drug resistance.
  4. Induction of apoptosis. Aberrantly activated anti-apoptotic BCL-2 family members can contribute to inappropriate survival and accumulation of eosinophils in clonal eosinophilia. Different strategies to inhibit anti-apoptotic members of the BCL-2 family are currently being tested in vitro and in clinical trials with the goal of tipping the balance in favor of cell death. Both sabutoclax and obatoclax target MCL-1 and other BCL-2 family members. Omacetaxine, on the other hand, causes a decrease of the short-lived MCL-1 through inhibition of protein synthesis.
  5. Inhibition of altered transcription. Altered transcription pathways have been linked to self-renewal maintenance and blocking of expression of tumor suppressors. As examples, drugs which inhibit histone deacetylation or β-catenin expression are indicated. Increased levels of β-catenin enhance its interaction with the transcription factors of the TCF-LEF family, its translocation to the nucleus, and modulation of transcription of downstream genes. Indomethacin was shown to attenuate transcription of β-catenin-TCF responsive genes.
  6. Mobilization of clonal eosinophil LSC. Plerixafor is an antagonist of the stem cell homing receptor CXCR4 that is also expressed on eosinophils. Correspondingly, the CXCR4 ligand SDF-1 is a strong chemoattractant for both stem cells and eosinophils, pointing to a major role of SDF-1/CXCR4 interactions in clonal eosinophilic disorders. Plerixafor disrupts this interaction and thus leads to rapid mobilization of normal and neoplastic stem- and progenitor cells. Following mobilization from the bone marrow niche, these cells are more likely to respond to anti-leukemic therapies, e.g. imatinib.
  7. Targeting the LSC niche. It is hypothesized that hypoxic areas are increased in the leukemic bone marrow. PR-104 is a small-molecule pre-drug activated by reduction to a DNA cross-linker. It inhibits DNA repair and synthesis, causing cell-cycle arrest, and apoptosis in LSC and progenitor cells. There are many other drugs that can attack the vascular bone marrow niche. Some of these drugs can directly inhibit the growth of endothelial cells and angiogenesis. Among these drugs are lenalidomide and some of the second- and third-generation BCR/ABL TKIs (nilotinib, ponatinib).

New Targeted Treatment Strategies

Targeting eosinophil-specific surface receptors

Although neutralization of IL-5 by antibody treatment can be beneficial for patients with FIP1L1-PDGFRA positive clonal eosinophilic disorders (52), this strategy has limited potential in this group of patients since it fails to address the underlying LSC. Another approach is to target eosinophils directly using afucosylated antibodies that bind to eosinophil-specific surface receptors, thus marking the cell for enhanced natural killer (NK) cell – mediated killing (Figure 1A). Of the antibodies currently in clinical development, benralizumab, an afucosylated antibody that binds to the surface receptor for IL-5 (IL-5Rα) (53), is likely the most promising since IL-5Rα is expressed on both eosinophils and eosinophil precursors in the bone marrow, peripheral blood, and tissues (12). Benralizumab has been tested in phase I and phase II trials in normal subjects and patients with asthma. In a recent study, bone marrow biopsies performed following a single dose of benralizumab demonstrated depletion of eosinophils and precursors in all 4 subjects (54). The fact that IL-5Rα is also expressed on mast cells and basophils (12) is an additional potential advantage in treatment of clonal HESs, since these lineages are often involved.

Several additional antibodies targeting eosinophil surface receptors are in preclinical development. The epidermal growth factor-like modul-containing, mucin-like hormone receptor 1 (EMR1) is an eosinophil-specific receptor (55) expressed on mature eosinophils. A novel, humanized, afucosylated anti-EMR1 IgG1 antibody was recently shown to enhance NK cell-mediated killing of eosinophils in vitro and in non-human primates (55).

Siglec-8 is an inhibitory receptor expressed on eosinophils, basophils, and mast cells (56). Crosslinking of Siglec-8 induces eosinophil cell death and inhibition of mast cell degranulation in vitro (57). More importantly, administration of anti-Siglec-F antibody in a mouse model (mouse Siglec-F is the closest functional paralog of human Siglec-8) reduced eosinophil levels in the blood and tissues (58). Of note, human intravenous immunoglobulin preparations contain natural anti-Siglec-8 autoantibodies (59). Unfortunately, since neither EMR1 nor Siglec-8 is expressed in early eosinophil differentiation stages, antibodies targeting these receptors are unlikely to deplete the causative cells in clonal eosinophilic disorders.

Enhancing protein degradation

The FIP1L1-PDGFRA gene fusion product is able to escape protein degradation, resulting in enhanced oncogenic potential (23). In non-leukemic states, receptor tyrosine kinase activation leads to Cbl-mediated mono-ubiquitination of lysines, and subsequent degradation. Since chimeric and patient-derived FIP1L1-PDGFRA have more stable expression compared with wild-type PDGFRA (23), specifically targeting the mutant proteins for degradation could be a promising therapeutic strategy (Figure 1B).

Targeting regulators of autophagy

Pharmacological targeting of different steps in the autophagy process in imatinib-treated CML cells significantly increased cell death (60). Furthermore, siRNA-mediated down-regulation of the autophagy-related protein 5 (ATG5) or ATG7 resulted in significant reductions in colony formation by imatinib-treated CML cells (60). Autophagic activity has not yet been investigated in clonal HESs. However, since autophagy regulates both self-renewal and differentiation of adult stem cells (61), it is possible that inhibitors of autophagy could both overcome TKI resistance and induce death in quiescent LSC in the bone marrow (Figure 1C).

Targeting regulators of apoptosis

Cell death is largely regulated by members of the BCL-2 family. Therefore, targeting anti-apoptotic members of this family, such as BCL-2 or myeloid cell leukemia-1 (MCL-1), is an emerging strategy in cancer therapy (Figure 1D). Targeting aberrantly activated BCL-2 family proteins is not only a promising approach to overcome TKI resistance, but it may also sensitize LSC for tyrosine kinase inhibition (62).

Omacetaxine is an inhibitor of protein synthesis (63). Inhibition leads to the down-regulation of short-lived anti-apoptotic proteins, such as MCL-1, with subsequent induction of apoptosis. The lack of target specificity (i.e. independence from BCR/ABL binding) makes it also independent of the BCR/ABL mutation status. The clinical effectiveness of omacetaxine in imatinib-resistant CML is as high as 77% (64), but remains unknown in FIP1L1-PDGFRA positive HES.

Sabutoclax is a novel inhibitor of anti-apoptotic members of the BCL-2 family (including MCL-1), currently being tested in various in vitro leukemia models. For instance, sabutoclax rendered CML blast crisis bone marrow stem cells sensitive to the tyrosine kinase inhibitor dasatinib, but had no effect on normal progenitors (62). Obatoclax, another small molecule targeting MCL-1 and other anti-apoptotic members of the BCL-2 family (65), is currently being tested in phase II clinical trials for treatment of leukemia, lymphoma, myelofibrosis, and mastocytosis.

Finally, antisense oligonucleotides, such as PNT2258, can be used to target anti-apoptotic BCL-2 family members. In a phase I study in patients with solid tumors, PNT2258 was both safe and well tolerated (66). A phase II study in patients with Non-Hodgkin’s lymphoma is currently underway.

Targeting signaling pathways and transcription

Investigation of the role of self-renewal signaling pathways in CML-LSC revealed an important role of β-catenin, a key effector in the Wnt pathway (67). Targeting β-catenin with the COX2 inhibitor indomethacin increased the therapeutic effect of imatinib (Figure 1E) (67, 68). Similarly, pan-histone deacetylase inhibitors sensitized CML-LSC to imatinib treatment (69), resulting, however, in parallel toxicity to normal HSC (70). Sirtulin 1 (SIRT1), which belongs to a subgroup of histone-deacetylases not targeted by the pan-histone-deacetylase inhibitor, has been linked to self-renewal maintenance as well as to the regulation of tumor suppressors like p53 and FoxOs (71). In CML-LSC, SIRT1 overexpression has been documented and pharmacological inhibition or knock-down of its expression resulted in increased activation of p53 and increased apoptosis (70). Additionally, SIRT1 inhibition was able to potentiate the effect of imatinib (70).

Targeting surface receptors on LSC

It is generally appreciated that LSC represent an important target cell population, especially for any curative treatment approaches (72, 73). Although the exact immunophenotype of LSC in clonal eosinophilic disorders is unknown, based on similar disease models, it is unlikely that the LSC in clonal eosinophilic disorders reside within a CD34+ fraction. In CML, these cells have been defined and their target expression profiles have been characterized in some detail (73). Major targets expressed on LSC in CML include CD25 (IL-2RA), CD33 (Siglec-3), CD44 (Pgp-1), CD52 (Campath-1), CD123 (IL-3RA), and CD184 (CXCR4). Targeted antibodies against these surface structures have the advantage that slowly cycling, or even quiescent cells, could be depleted, which might be an achievable goal in both CML and CEL (Figure 1A).

Enhancing LSC release from the stem cell niche

A pivotal factor in promoting bone marrow homing of both hematological and leukemic progenitor cells is CXCR4 (CD184) (74, 75). In diverse leukemias, CXCR4 is expressed on LSC and progenitor cells. Plerixafor is a CXCR4 antagonist, currently FDA-approved for use in mobilization of HSC from the bone marrow for autologous stem cell transplantation (75). Recent data suggest that plerixafor can also mobilize neoplastic stem cells in various types of leukemias and may thereby enhance the responsiveness of these cells to various anti-leukemic drugs (Figure 1F) (74). As CXCR4 is an important homing factor for diverse types of HSCs, this stem cell receptor is also likely to play an important role in clonal eosinophil progenitors homing to shelter sites, and, thus, might be a target for enhancing TKI efficacy in patients with clonal eosinophilic disorders in the same way as in other leukemia models. The limitation of this strategy is certainly the lack of distinction between leukemic and normal hematopoietic progenitors in the mobilization from the bone marrow. On the other hand, the effects of concomitant TKI administration might lead to a more specific destruction of LSC.

Targeting the stem cell niche

Similar to HSC, LSC are believed to depend critically on their niches in the bone marrow (72). In the currently emerging scenario, LSC are thought to remodel the bone marrow niche to achieve a survival advantage over non-leukemic stem cells (76). One hypothesis is that hypoxic areas are enhanced in the leukemic bone marrow. Consequent stabilization of hypoxia-inducible factor α (HIF-α) in LSC alters their metabolism to glycolysis enabling survival in a quiescent state (Figure 1G) (77). In addition, hypoxia promotes VEGF production and, thus, bone marrow angiogenesis together with the related vascular niche. Based on these assumptions, targeting of hypoxic niches may be a promising new approach for the destruction of LSC. Experimental therapy with PR-104, a dinitrobenzamide mustard, activated by reduction to the cytotoxic hydroxylamine metabolite, caused a reduction of LSC and increased host survival in immune-deficient mice injected with primary, acute lymphoblastic leukemia cells (78), providing a proof of principle.

Recent data suggest that LSC evolution is regulated by the endosteal and vascular stem cell niches (72, 79). Therefore, several strategies target LSC-related angiogenesis with the final goal of destroying both the niche and niche-dependent LSC. Interestingly, some of the new TKIs, such as nilotinib, act on endothelial cells to inhibit their growth and, hence, angiogenesis. These TKIs and other anti-angiogenic drugs, such as lenalidomide (80), may therefore be interesting drugs to target the vascular LSC niche in patients with drug-resistant leukemia.

Lymphocytic Hypereosinophilic Syndromes

The hallmark of LHESs is increased production of eosinopoietins, especially IL-5, by activated polyclonal T cells or T cell clones (1, 1517, 20, 81). Several different T cell immune-phenotypic abnormalities have been described, including double negative T cells (CD3+CD4-CD8-) (81) and T cells lacking CD3 expression (CD3-CD4+) (82), although the surface phenotype may be normal even in the setting of a T cell clone. Abnormal expression of other lineage-associated markers, such as CD2, CD5, CD6, and CD7, are often present (20).

Although glucocorticosteroid therapy is effective in reducing eosinophilia and symptoms in the majority of patients with LHES, moderate to high doses are often necessary (20, 29, 30). Furthermore, glucocorticosteroid treatment does not eliminate cytokine-producing T cells. In this regard, IFN-α, the most common second-line agent for LHESs, has the theoretical advantage of potentially depleting the cytokine-producing T cell clone (21). In severe asthma, IFN-α has been shown to support Th1 differentiation and IL-10 production (83), suggesting additional mechanisms of anti-eosinophil activity. It should be noted that IFN-α has also been successfully used in the treatment of HESs (29, 30). In the following section, we will address a few selected, novel therapeutic approaches for the treatment of LHESs (Supplemental Table 1 and Figure 2).

Figure 2. Current targeted therapeutic approaches for allergy are presented below.

Figure 2

Compounds not yet in clinical trials are not included.
  1. Schematic overview of the targeted cells and effector molecules involved in the pathogenesis of allergy. Briefly, mast cell and basophil activation is initiated upon crosslinking of surface-bound IgE. The mediators released following activation include chemotactic factors for T cells and eosinophils (PGD2), as well as IL-4 and IL-13. IL-4 and TSLP, largely produced by epithelial cells, which promote the differentiation of Th0 to Th2 cells. IL-4 and IL-13 enhance IgE production in B cells. IL-5 and eotaxin, released by epithelial cells, enhance eosinophil infiltration. Targeted therapeutic strategies are directed against both effector molecules and surface receptors. The expression of critical receptors that can be targeted by currently available drugs is indicated for each cell with a dotted line.
  2. Omalizumab, a humanized antibody, binds to free IgE and, as a consequence, decreases FcεR-bound IgE. This also leads to a down-regulation of the high-affinity FcεR on mast cells and basophils.
  3. Different strategies for blocking the IL-4/IL-13 axis are currently under investigation. Both receptors contain an IL-4Rα chain. Antagonism of both IL-4 and IL-13 signaling pathways may thus be achieved by blocking IL-4Rα either through targeted antibodies (dupilumab) or receptor antagonism (pitrakinra). Furthermore, IL-13 blocking antibodies have shown promising results.
  4. Blocking the effects of PGD2 is currently being addressed by several receptor antagonists.
  5. The human monoclonal antibody AMG 157 binds to TSLP, thereby interfering with the interaction of TSLP with its receptor on mast cells, basophils, and eosinophils, as well as dendritic cells (not shown).
  6. Eotaxins are critical chemotactic factors for eosinophils. The currently investigated antibody targeting these chemokines is bertilimumab, directed against eotaxin-1 (CCL11). IL-5 is a critical pro-survival and activating factor of eosinophils. Its effect can be blocked by either targeting the cytokine with mepolizumab or reslizumab, or by targeting IL-5Rα. Receptor antagonism of CCR3 (CD193) is an attempt to directly block eosinophil chemotaxis. Decreased expression of both CCR3 and the common β-chain of the IL-5R is the aim of treatment with TP ASM8, an antisense oligonucleotide.

Eosinophil Targets

IL-5

Both mepolizumab and reslizumab, humanized antibodies to IL-5 in development for treatment of asthma, have demonstrated efficacy in patients with non-clonal HES (Supplemental Table 1). Based on the results of two small studies, which demonstrated that mepolizumab could safely reduce eosinophilia and improve symptoms in patients with non-clonal HES (84, 85), 85 FIP1L1-PDGFRA negative HES patients were enrolled in a double-blind, placebo-controlled study of monthly mepolizumab as a steroid-sparing agent (86). Overall, 84% of the subjects who received mepolizumab met the primary endpoint of a glucocorticosteroid dose of ≤ 10 mg/d for ≥ 8 weeks (86). Subgroup analysis revealed a comparable steroid-sparing effect in the 13 patients with LHES caused by an IL-5-producing T cell clone compared to the group as a whole, although eosinophil counts were suppressed to a lesser degree (87). Among 78 patients participating in a long-term open-label extension of the placebo-controlled study, 62% were prednisone-free for ≥12 consecutive weeks, confirming the long-term efficacy of mepolizumab in suppressing eosinophilia and symptoms in patients with HES (88). Ex vivo experiments suggested that the high efficacy resulted both from a decrease in total eosinophil numbers and from reduced activation of the remaining eosinophils (89). Although data is limited to a single open-label study in 4 patients with treatment-refractory disease, reslizumab is likely to show similar efficacy to mepolizumab in reducing eosinophilia in HESs, including LHESs (52).

Both mepolizumab and reslizumab have been well-tolerated in HES patients in clinical trials to date, and no safety concerns were identified in the long-term mepolizumab study (88). Furthermore, the physiological eosinophil infiltration of the gastrointestinal tract was unaltered following anti-IL-5 antibody treatment in patients with eosinophilic esophagitis (EoE) (90). Nevertheless, maturational arrest in the bone marrow, increased production of intracellular IL-5 in CD4+ and CD8+ T cells and increased serum levels of IL-5 after mepolizumab treatment (89) have raised concern for potentially uncontrolled IL-5 - mediated inflammation upon interruption of therapy (91). In fact, an increase in blood eosinophilia above baseline, accompanied by an exacerbation of symptoms, has been observed after a single dose of reslizumab as drug levels declined several weeks after drug discontinuation (52). It is possible that increased eotaxin levels following IL-5 neutralization (92) contribute, at least partially, to such unwanted events following drug withdrawal.

IL-5Rα

Targeting IL-5Rα by benralizumab is thought to exert an anti-eosinophil effect both by interfering with IL-5 binding and by promoting antibody-dependent cell-mediated cytotoxicity (ADCC) with consequent enhanced apoptosis (53). When cultured in the presence of benralizumab, eosinophils and basophils undergo NK cell-mediated apoptosis. Importantly, such apoptosis was not associated with eosinophil degranulation under these conditions (53). In a phase I dose-escalation trial in patients with mild atopic asthma, benralizumab treatment caused a dose-dependent decrease in peripheral blood eosinophil counts within 24 h that persisted for up to 12 weeks after a single 3 mg/kg dose (93). A subsequent phase II trial in 27 subjects with eosinophilic asthma confirmed safety and efficacy of benralizumab in reducing blood, bone marrow, and tissue eosinophilia (54). A phase III trial in patients with asthma and a phase II trial in patients with HES are ongoing.

Siglec-8 and EMR1

As discussed above, monoclonal antibodies to the eosinophil surface receptors, Siglec-8 and EMR1, have been developed (55, 56) and are in pre-clinical development for the treatment of eosinophilic disorders. Both receptors are highly expressed on eosinophils from normal and eosinophilic individuals (55, 94) and are promising therapeutic targets in HESs, including LHESs.

T Cell Targets

CD52

CD52, also known as Campath-1 antigen, is a surface glycoprotein expressed on T cells, B cells, NK cells, most monocytes, macrophages, and eosinophils (91). Alemtuzumab is a humanized anti-CD52 monoclonal antibody that was initially approved for the treatment of B-cell chronic lymphocytic leukemia (CLL). Two small case series and several additional case reports demonstrated the potential of alemtuzumab to induce hematological remission in HES patients (95, 96). Since both eosinophils and T cells express CD52, potential mechanisms of action include a direct effect on eosinophils and/or destruction of eosinopoietin-producing T cells.

Despite initial benefit in a majority of patients in the two published case series, 70% and 83% of patients relapsed upon cessation of therapy (95, 96). In addition, alemtuzumab can lead to severe side effects, including severe hematological toxicity with resulting opportunistic infections (96). In one case series in HES, almost 50% of the patients died during the follow-up period and in the majority, the cause of death was attributed to the treatment rather than to the underlying eosinophilia (95).

CD2

Alefacept is a fusion protein of the human IgG1 Fc domain and lymphocyte function-associated antigen (LFA)-3 (CD58) (97). LFA-3 binding to CD2 prevents co-stimulation, which is required for T cell activation. Furthermore, binding of IgG1 to Fcγ receptors, particularly CD16, is thought to mediate interaction between T and NK cells, with consequent memory T cell apoptosis (91). Alefacept has been shown to be effective in the treatment of patients with atopic dermatitis (98) and psoriasis (99) and has demonstrated anti-eosinophil activity in patients with atopic dermatitis (98), perhaps, due to the depletion of cytokine-producing T cells (91). However, the manufacturer discontinued the development of alefacept in 2011 (www.amevive.com).

Allergic Diseases

Allergic diseases are characterized by Th2-driven inflammatory responses, often resulting in increased IgE levels and eosinophilia (Figure 2A). They comprise a wide spectrum of conditions affecting many different organ systems, each of which may require treatment with different drugs. Glucocorticosteroids are widely used to suppress the underlying chronic inflammation in allergic diseases, including those of the nose, eye, lung, skin, and gastrointestinal tract. However, glucocorticosteroids have potentially debilitating, unwanted effects when used incorrectly or inappropriately. Moreover, these drugs are not effective initially in all patients and many patients develop glucocorticosteroid resistance over time. Clearly, the development of novel, more specific and targeted therapies is needed. Below, we discuss new developments in therapies that target different components of the immune system (Supplemental Table 1, Figure 2B-F), with the exception of so-called immunotherapy, which has been reviewed elsewhere (100). It should be noted that, since allergy-associated eosinophilia is driven by eosinopoietin-producing T cells, all anti-eosinophil strategies described for LHESs also apply to the treatment of allergic diseases.

Targeting IgE

Recombinant humanized anti-IgE monoclonal antibody treatment with omalizumab was shown to be beneficial as a steroid-sparing agent in asthma patients, reducing asthma exacerbations, improving symptom severity, and enhancing the quality of life in these patients (101). Of note, the effectiveness of omalizumab for reduction of asthma exacerbations was shown to be best in allergic asthmatics with high blood eosinophilia, high periostin levels (a marker for IL-4 and IL-13 activity and a potential predictor of airway eosinophilia) and increased fractional exhaled nitric oxide (a marker of airway inflammation) (102). Serum levels of IgE, on the other hand, were not predictive of responsiveness to omalizumab (103). In a cohort of patients with nasal polyposis and comorbid asthma, treatment with omalizumab led to a significant reduction in airway symptoms and endoscopic polyp scores, whether or not associated with allergy (104). Recently, treatment of standard, therapy-resistant urticaria has evolved as a novel, currently still off-label, indication for omalizumab (105). Omalizumab is generally well tolerated (106).

Omalizumab binds free IgE precluding its binding to IgE receptors on mast cells and basophils (Figure 2B) (107). This leads to decreased expression of IgE receptors on mast cells, basophils, and dendritic cells (108), and a decrease in the functional responses of these cells to IgE-dependent stimuli. Downregulation of IL-2 and IL-13 production by T cells (109), as well as reduced bronchial eosinophil infiltration and sputum eosinophil counts, were also observed in patients treated with omalizumab (110). Moreover, IgE synthesis in B cells was decreased, potentially due to induction of a state of anergy in IgE-bearing B cells (111). Omalizumab treatment also reduced expression of TSLP and increased expression of IL-10 in the skin of children with severe atopic dermatitis (112).

Recently, an IgE-specific DARPin (designed ankyrin repeat protein) has been reported to be able to bind IgE with very high affinity causing IgE molecules to be easily dissociated from high-affinity IgE receptors (113). Clearly, such compounds may pave the way for the next generation of IgE-blocking molecules with the goal of blocking allergic reactions.

Eosinophil Targets

IL-5 and IL-5Rα

Despite the failure of early studies to show efficacy of anti-IL-5 antibody therapy in the treatment of asthma (114, 115), recently multiple studies have demonstrated improvement of peripheral eosinophilia, a significant reduction of asthma exacerbations, and/or improvements in airway function among asthmatic patients with blood and/or sputum eosinophilia receiving mepolizumab or reslizumab (116119).

Eosinophilic granulomatosis with polyangiitis (EGPA), previously known as Churg-Strauss syndrome, is characterized by asthma, sinusitis with nasal polyposis, eosinophilic vasculitis with extravascular granulomas, and peripheral blood eosinophilia (120). Although the pathogenesis of EGPA is believed to be multifunctional, elevated IL-5 levels and the expression of eosinophil activation markers are common, especially in ANCA-negative patients. The safety and efficacy of mepolizumab as a steroid-sparing agent in EGPA has been demonstrated in a small placebo-controlled trial (121). In a second small open-label trial, mepolizumab treatment decreased the relapse rate in patients with relapsing or refractory EGPA, although the effect was not sustained when the drug was discontinued (122, 123). A multicenter, phase III trial of mepolizumab as a steroid-sparing agent in patients with relapsing or refractory EGPA is currently underway. Reslizumab has not been studied in EGPA, but has been reported to have an impact on nasal polyposis in patients with elevated IL-5 in nasal secretions (124).

Results of clinical trials of anti-IL-5 therapy for other allergic eosinophilic disorders have been disappointing. In pediatric and adult patients with EoE, significant clinical improvement was not observed despite reductions in the numbers of blood and tissue eosinophils (92, 125, 126). A similar lack of clinical efficacy was seen in a placebo-controlled trial of mepolizumab in patients with atopic dermatitis (127).

In a multicenter, double-blind, placebo-controlled phase II study in asthmatic patients with sputum eosinophilia, benralizumab reduced airway eosinophil counts by 95.8%, sputum eosinophil counts by 89.9% and blood eosinophilia by 100% after 4 weeks of subcutaneous administration (54). Moreover, benralizumab suppressed eosinophil counts in the airway mucosa by 61.9% following intravenous application. No serious adverse events related to benralizumab were reported (54). A phase III study of subcutaneous benralizumab in asthmatic patients is currently underway (Supplemental Table 1).

Eotaxin – CCR3

Eotaxins are a family of three chemokines, CCL11 (eotaxin-1), CCL24 (eotaxin-2) and CCL26 (eotaxin-3), which evoke chemotactic activity in cells expressing the eotaxin receptor CCR3, primarily eosinophils and basophils (Figure 2F) (128). Eotaxin-1/2 knockout mice fail to recruit eosinophils to the lung upon allergen challenge. IL-4 and IL-13 production were significantly reduced in these mice, as well as in eosinophil-depleted mice (128). These data suggest that targeting the eotaxin-CCR3 axis would strongly reduce eosinophil recruitment to allergic sites.

Bertilimumab, a human anti-eotaxin-1 antibody, is currently in a phase II clinical trial for ulcerative colitis, an autoimmune inflammatory disease known to be associated with elevated CCL11 levels (129). Ongoing studies with GW766944 and other investigational CCR3 antagonists have been reviewed recently (130).

TPI ASM8 is a novel agent consisting of antisense oligonucleotides to human CCR3 and common β-chain transcripts. Its application leads to decreased expression of CCR3 and the common β-chain of the IL-5R, IL-3R, and GM-CSFR. In mild atopic asthmatics, inhaled TPI ASM8 significantly reduced allergen-induced sputum eosinophilia (131). In a second study, early and late asthmatic responses were significantly attenuated (132).

T Cell Targets

TSLP

Thymic stromal lymphopoietin (TSLP) has been linked to the initial phases of allergic disorders and is a critical driver of Th2 differentiation through its effect on dendritic cells (133). Overexpression of TSLP in the skin of mice causes a disease that resembles atopic dermatitis (134) and lung-specific TSLP-expressing transgenic mice develop a chronic disease with characteristics of bronchial asthma (135). Furthermore, increased TSLP expression has been observed in skin biopsies of patients with atopic dermatitis and genetic alterations in the TSLP gene have been associated with EoE (136), consistent with the hypothesis that TSLP is critically involved in the evolution of various allergic diseases. Recent data demonstrating that the TSLP receptor is expressed on eosinophils and that eosinophil extracellular DNA traps are formed upon TSLP stimulation (137), suggest that eosinophils play a key role in TSLP-mediated pro-inflammatory effects. Basophils may also be important as evidenced by studies linking TSLP gain-of-function polymorphisms in EoE patients to higher basophil activity and a dependency on TSLP and basophils in a mouse model of EoE-like disease (138). Thus, therapy targeting TSLP could result in both anti-basophil and anti-eosinophil effects (Figure 2D). Anti-TSLP antibody is currently being investigated in several phase I clinical trials (Supplemental Table 1).

IL-4/IL-13 and their receptors

Targeting both arms of the IL-4 and IL-13 axis is a promising strategy for the treatment of asthma (139). Such an approach has been taken with pitrakinra, a recombinant protein derived from human IL-4 that acts as a competitive antagonist of both IL-4 and IL-13 (Figure 2C) (139). Pitrakinra significantly reduced allergen-induced airway hyperreactivity in an experimental animal model of asthma (140) and prevented asthma exacerbations in a subgroup of asthma patients (139), although clinical efficacy was dependent on the presence of a specific IL-4 receptor polymorphism (141). Dupilumab, a fully human antibody directed against the IL-4Rα shared by IL-4 and IL-13 receptors (Figure 2C), has been evaluated in persistent, moderate-to-severe asthma with elevated blood eosinophil counts (142). Dupilumab treatment, as compared with placebo, brought fewer asthma exacerbations and improved lung function following the withdrawal of inhaled glucocorticosteroid and long-acting beta-agonist therapies (142). Levels of TARC, eotaxin-3, and IgE were significantly reduced following dupilumab treatment, confirming that the IL-4-driven pathway was blocked (142). Of note, neither blood eosinophil counts or sputum eosinophils were significantly decreased by pitrakinra or dupilumab treatment (139, 142). A human IgG4 bispecific antibody able to neutralize both cytokines is currently in clinical development (143).

Neutralization of IL-13 alone has also been investigated (Figure 2C). Two anti-IL-13 antibodies have been developed and used in clinical trials (Supplemental Table 1). Lebrikizumab is a humanized monoclonal anti-IL-13 antibody and was evaluated in a randomized, double-blind, placebo-controlled study in 219 adults with inadequately controlled asthma despite inhaled glucocorticosteroid therapy (144). Interestingly, the improvement in lung function correlated with serum periostin levels, an indicator of IL-13 activity (144). These data provided further evidence of the heterogeneous nature of asthma and confirmed the utility of careful patient selection before applying targeted treatment approaches. More recently, lebrikizumab was also shown to reduce the late asthmatic response in patients with mild asthma (145). Tralokinumab is a fully human anti-IL-13 monoclonal IgG4 antibody. In a large randomized, placebo-controlled study enrolling 194 patients with moderate-to-severe uncontrolled asthma, tralokinumab safely improved lung function (146).

CRTh2

In recent years, evidence has accumulated for the critical role of prostaglandin D2 (PGD2), a prostanoid produced mainly by mast cells, in allergic inflammation. PGD2 has been shown to mediate activation and recruitment of eosinophils, basophils, and Th2 cells, as well as promote enhanced Th2 cytokine production through the PGD2 receptor chemoattractant receptor-homologous molecule on Th2 cells (CRTh2) (147). Moreover, CRTh2 agonists increased allergic inflammation in murine models of atopic dermatitis and allergic asthma (148). In agreement with these findings, CRTh2 deficient mice mounted a less pronounced inflammatory response to hapten-specific IgE injections, and this was associated with decreased tissue infiltration of eosinophils, basophils, and lymphocytes (149). CRTh2 has thus evolved as a promising new therapeutic target for treatment of allergic disease (Figure 2D; Supplemental Table 1). The CRTh2 antagonist OC000459 significantly reduced eosinophil-inflammation in asthma (150) and in EoE (151), leading to clinical improvement in both patient groups as compared to patients receiving placebo. OC000459 has also shown efficacy in reduction of grass pollen-induced conjunctivitis and rhinitis symptoms (152).

GATA-3 specific DNAzyme

The importance of the transcription factor GATA-3 in Th2 cell activation and regulation of mast cells, eosinophils, and epithelial cells has led to investigationof GATA-3 as an emerging new therapeutic target. Topical application of so-called DNAzyme, single-stranded DNA molecules targeting and cleaving specific sequences in target mRNA molecules (i.e., gd21 and hgd40), was shown to be highly effective in the prevention of allergic inflammation and airway hyperresponsiveness in murine models of acute allergy airway inflammation (153, 154). No off-target effects were observed with gd21 (153) and aerosol inhalation of hgd40 in rats and dogs was well-tolerated with only mild systemic effects (154). A dose-dependent increase in IL-10 and IFN-γ was seen in the hgd40 study, but at very low levels (154).

Targeting B Cells

Rituximab is a chimeric monoclonal anti-CD20 antibody, originally developed for the therapy of B cell malignancies. CD20 is expressed on pre-B cells and mature B cells, but not on plasma cells. In an investigator-driven study, six patients with severe atopic dermatitis were treated with rituximab (155). All patients showed improvement in skin symptoms within 4 to 8 weeks. Of note, expression of IL-5 and IL-13 was reduced after therapy, whereas allergen-specific IgE levels were not altered (155). These data suggest that depletion of B cells may decrease T cell activation, resulting in reduced Th2 cytokine expression.

Concluding Remarks

During the past few years, our understanding of HE and HES has increased substantially, and several new treatment approaches have been translated from preclinical models into clinical practice. Today, further development of new targeted therapies for eosinophilic disorders, including allergic diseases, is closely linked to an understanding of mechanisms for evolution of disease progression and drug resistance. In line with this notion, any targeted therapy requires optimal patient selection confirmed by adequate biomarkers. Ongoing research aimed at defining the physiological and pathological roles of eosinophils will provide additional targets and drugs that can be applied to eosinophilic diseases and allergic disorders. In eosinophil neoplasms, the ultimate goal will be to eradicate LSC in order to achieve long-term cures.

Supplementary Material

Supplemental Table 1

Acknowledgments

Work in the authors’ laboratories is funded by grants from the Swiss National Science Foundation (310030-146181) and Swiss Cancer League (3099-02-2013) to H.U.S., from Allergy Foundation Ulrich Müller-Gierok, Bern, to S.R.H. and H.U.S., from the Austrian Science Fund, FWF (SFB F4704-B20) to P.V., and from the National Institutes of Health (U01 AI097073) to M.E.W. This work is funded in part by the Division of Intramural Research, NIAID, NIH. S.R.H. received a fellowship from the Swiss National Science Foundation. We thank Dr. L. Scapozza (Dept. of Pharmaceutical Sciences, University of Geneva, Switzerland) for valuable comments on second- and third-generation TKIs.

Footnotes

Disclosure Statement

M.E.W. has received consulting honoraria from Merck, Novartis, GlaxoSmithKline, Medimmune, Boston Scientific, Regeneron, Cytos, and Teva. The other authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

Literature Cited

  • 1.Simon D, Simon HU. Eosinophilic disorders. J Allergy Clin Immunol. 2007;119:1291–1300. doi: 10.1016/j.jaci.2007.02.010. [DOI] [PubMed] [Google Scholar]
  • 2.Linch SN, Gold JA. The role of eosinophils in non-parasitic infections. Endocr Metab Immune Disord Drug Targets. 2011;11:165–72. doi: 10.2174/187153011795564188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Blanchard C, Rothenberg ME. Biology of the eosinophil. Adv Immunol. 2009;101:81–121. doi: 10.1016/S0065-2776(08)01003-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Straumann A, Simon HU. The physiological and pathophysiological roles of eosinophils in the gastrointestinal tract. Allergy. 2004;59:15–25. doi: 10.1046/j.1398-9995.2003.00382.x. [DOI] [PubMed] [Google Scholar]
  • 5.Jacobsen EA, Helmers RA, Lee JJ, Lee NA. The expanding role(s) of eosinophils in health and disease. Blood. 2012;120:3882–90. doi: 10.1182/blood-2012-06-330845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chu VT, Fröhlich A, Steinhauser G, Scheel T, Roch T, et al. Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat Immunol. 2011;12:151–9. doi: 10.1038/ni.1981. [DOI] [PubMed] [Google Scholar]
  • 7.Wu D, Molofsky AB, Liang HE, Ricardo-Gonzalez RR, Jouihan HA, et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332:243–7. doi: 10.1126/science.1201475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ackerman SJ. To be, or not to be, an eosinophil: that is the ??? Blood. 2013;122:621–3. doi: 10.1182/blood-2013-06-508507. [DOI] [PubMed] [Google Scholar]
  • 9.Lopez AF, Begley CG, Williamson DJ, Warren DJ, Vadas MA, Sanderson CJ. Murine eosinophil differentiation factor. An eosinophil-specific colony-stimulating factor with activity for human cells. J Exp Med. 1986;163:1085–99. doi: 10.1084/jem.163.5.1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Metcalf D. The molecular biology and functions of the granulocyte-macrophage colony-stimulating factors. Blood. 1986;67:257–67. [PubMed] [Google Scholar]
  • 11.Martinez-Moczygemba M, Huston DP. Biology of common beta receptor-signaling cytokines: IL-3, IL-5, and GM-CSF. J Allergy Clin Immunol. 2003;112:653–65. doi: 10.1016/S0091. [DOI] [PubMed] [Google Scholar]
  • 12.Wilson TM, Maric I, Shukla J, Brown M, Santos C, et al. IL-5 receptor alpha levels in patients with marked eosinophilia or mastocytosis. J Allergy Clin Immunol. 2011;128:1086–92. doi: 10.1016/j.jaci.2011.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Simon HU, Blaser K. Inhibition of programmed eosinophil death – a key pathogenic event for eosinophilia? Immunol Today. 1995;16:53–5. doi: 10.1016/0167-5699(95)80086-7. [DOI] [PubMed] [Google Scholar]
  • 14.Geering B, Stoeckle C, Conus S, Simon HU. Living and dying for inflammation: neutrophils, eosinophils, basophils. Trends Immunol. 2013;34:398–409. doi: 10.1016/j.it.2013.04.002. [DOI] [PubMed] [Google Scholar]
  • 15.Simon HU, Rothenberg ME, Bochner BS, Weller PF, Wardlaw AJ, 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]
  • 16.Valent P, Klion AD, Horny HP, Roufosse F, Gotlib J, et al. Contemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromes. J Allergy Clin Immunol. 2012;130:607–12. doi: 10.1016/j.jaci.2012.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Valent P, Klion AD, Rosenwasser LJ, Arock M, Bochner BS, et al. ICON: eosinophil disorders. WAO Journal. 2012;5:174–81. doi: 10.1097/WOX.0b013e31827f4192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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. doi: 10.1038/leu.2008.287. [DOI] [PubMed] [Google Scholar]
  • 19.Stoeckle C, Simon HU. CD8(+) T cells producing IL-3 and IL-5 in non-IgE-mediated eosinophilic diseases. Allergy. 2013;68:1622–5. doi: 10.1111/all.12311. [DOI] [PubMed] [Google Scholar]
  • 20.Simon HU, Plötz SG, Dummer R, Blaser K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med. 1999;341:1112–20. doi: 10.1056/NEJM199910073411503. [DOI] [PubMed] [Google Scholar]
  • 21.Simon HU, Plötz SG, Simon D, Dummer R, Blaser K. Clinical and immunological features of patients with interleukin-5 producing T cell clones and eosinophilia. Int Arch Allergy Clin Immunol. 2001;124:242–5. doi: 10.1159/000053723. [DOI] [PubMed] [Google Scholar]
  • 22.Cross NC, Reiter A. Fibroblast growth factor receptor and platelet-derived growth factor receptor abnormalities in eosinophilic myeloproliferative disorders. Acta Haematol. 2008;119:199–206. doi: 10.1159/000140631. [DOI] [PubMed] [Google Scholar]
  • 23.Toffalini F, Kallin A, Vandenberghe P, Pierre P, Michaux L, et al. The fusion proteins TEL-PDGFRβ and FIP1L1-PDGFRα escape ubiquitination and degradation. Haematol. 2009;94:1085–93. doi: 10.3324/haematol.2008.001149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jackson CC, Medeiros LJ, Miranda RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76. doi: 10.1016/j.humpath.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 25.Pardanani A, Reeder T, Li CY, Tefferi A. Eosinophils are derived from the neoplastic clone in patients with systemic mastocytosis and eosinophilia. Leuk Res. 2003;27:883–5. doi: 10.1016/s0145-2126(03)00065-1. [DOI] [PubMed] [Google Scholar]
  • 26.Bacher U, Reiter A, Haferlach T, Mueller L, Schnittger S, et al. A combination of cytomorphology, cytogenetic analysis, fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction for establishing clonality in cases of persisting hypereosinophilia. Haematologica. 2006;91:817–20. [PubMed] [Google Scholar]
  • 27.Druker BJ. Translation of the Philadelphia chromosome into therapy for CML. Blood. 2008;112:4808–17. doi: 10.1182/blood-2008-07-077958. [DOI] [PubMed] [Google Scholar]
  • 28.Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, 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;348:1201–14. doi: 10.1056/NEJMoa025217. [DOI] [PubMed] [Google Scholar]
  • 29.Simon HU, Klion A. Therapeutic approaches to patients with hypereosinophilic syndromes. Semin Hematol. 2012;49:160–70. doi: 10.1053/j.seminhematol.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gotlib J. World Health Organization-defined eosinophilic disorders: 2012 update of diagnosis, risk stratification, and management. Am J Hematol. 2012;87:904–14. doi: 10.1002/ajh.23293. [DOI] [PubMed] [Google Scholar]
  • 31.Klion AD, Robyn J, Maric I, Fu W, Schmid L, et al. Relapse following discontinuation of imatinib mesylate therapy for FIP1L1/PGGFRA-positive chronic eosinophilic leukemia: implications for optimal dosing. Blood. 2007;110:3552–6. doi: 10.1182/blood-2007-07-100164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Simon D, Salemi S, Yousefi S, Simon HU. Primary resistance to imatinib in Fip1-like 1-platlet-derived growth factor receptor α-positive eosinophilic leukemia. J Allergy Clin Immunol. 2008;121:1054–6. doi: 10.1016/j.jaci.2007.11.027. [DOI] [PubMed] [Google Scholar]
  • 33.Salemi S, Yousefi S, Simon D, Schmid I, Moretti L, et al. A novel FIP1L1-PDGFRA mutant destabilizing the inactive conformation of the kinase domain in chronic eosinophilic leukemia/hypereosinophilic syndrome. Allergy. 2009;64:913–8. doi: 10.1111/j.1398-9995.2009.01943.x. [DOI] [PubMed] [Google Scholar]
  • 34.Frankfurt O, Licht JD. Ponatinib--a step forward in overcoming resistance in chronic myeloid leukemia. Clin Cancer Res. 2013;19:5828–34. doi: 10.1158/1078-0432.CCR-13-0258. [DOI] [PubMed] [Google Scholar]
  • 35.Lau A, Seiter K. Second-line therapy for patients with chronic myeloid leukemia resistant to first-line imatinib. Clin Lymphoma Myeloma Leuk. 2013 doi: 10.1016/j.clml.2013.11.002. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 36.Giles FJ, Abruzzese E, Rosti G, Kim DW, Bhatia R, et al. Nilotinib is active in chronic and accelerated phase chronic myeloid leukemia following failure of imatinib and dasatinib therapy. Leukemia. 2010;24:1299–1301. doi: 10.1038/leu.2010.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Latagliata R, Breccia M, Castagnetti F, Stagno F, Luciano L, et al. Dasatinib is safe and effective in unselected chronic myeloid leukaemia elderly patients resistant/intolerant to imatinib. Leuk Res. 2011;35:1164–9. doi: 10.1016/j.leukres.2011.05.015. [DOI] [PubMed] [Google Scholar]
  • 38.Syed YY, McCormack PL, Plosker GL. Bosutinib: a review of its use in patients with Philadelphia chromosome-positive chronic myelogenous leukemia. BioDrugs. 2014;28:107–20. doi: 10.1007/s40259-013-0082-x. [DOI] [PubMed] [Google Scholar]
  • 39.Lierman E, Michaux L, Beullens E, Pierre P, Marynen P, et al. FIP1L1-PDGFRalpha D842V, a novel panresistant mutant, emerging after treatment of FIP1L1-PDGFRalpha T674I eosinophilic leukemia with single agent sorafenib. Leukemia. 2009;23:845–51. doi: 10.1038/leu.2009.2. [DOI] [PubMed] [Google Scholar]
  • 40.Metzgeroth G, Erben P, Martin H, Mousset S, Teichmann M, et al. Limited clinical activity of nilotinib and sorafenib in FIP1L1-PDGFRA positive chronic eosinophilic leukemia with imatinib-resistant T674I mutation. Leukemia. 2012;26:162–4. doi: 10.1038/leu.2011.181. [DOI] [PubMed] [Google Scholar]
  • 41.Remsing Rix LL, Rix U, Colinge J, Hantschel O, Bennett KL, et al. Global target profile of the kinase inhibitor bosutinib in primary chronic myeloid leukemia cells. Leukemia. 23:477–85. doi: 10.1038/leu.2008.334. [DOI] [PubMed] [Google Scholar]
  • 42.Jin Y, Ding K, Li H, Xue M, Shi X, et al. Ponatinib efficiently kills imatinib-resistant chronic eosinophilic leukemia cells harboring gatekeeper mutant T674I FIP1L1-PDGFRα: roles of Mcl-1 and β-catenin. Mol Cancer. 2014;13:17. doi: 10.1186/1476-4598-13-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shen Y, Shi X, Pan J. The conformational control inhibitor of tyrosine kinases DCC-2036 is effective for imatinib-resistant cells expressing T674I FIP1L1-PDGFRalpha. PloS One. 2013;8:e73059. doi: 10.1371/journal.pone.0073059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chase A, Grand FH, Cross NCP. Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome. Blood. 2007;110:3729–34. doi: 10.1182/blood-2007-02-074286. [DOI] [PubMed] [Google Scholar]
  • 45.Chen J, DeAngelo DJ, Kutok JL, Williams IR, Lee BH, et al. PKC412 inhibits the zinc finger 198-fibroblast growth factor receptor 1 fusion tyrosine kinase and is active in treatment of stem cell myeloproliferative disorder. PNAS. 2004;101:14479–84. doi: 10.1073/pnas.0404438101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chase A, Bryant C, Score J, Cross NCP. Ponatinib as targeted therapy for FGFR1 fusions associated with the 8p11 myeloproliferative syndrome. Haematol. 2013;98:103–6. doi: 10.3324/haematol.2012.066407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Aichberger KJ, Herndlhofer S, Schernthaner GH, Schillinger M, Mitterbauer-Hohendanner G, et al. Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML. Am J Hematol. 2011;86:533–9. doi: 10.1002/ajh.22037. [DOI] [PubMed] [Google Scholar]
  • 48.Kim TD, Rea D, Schwarz M, Grille P, Nicolini FE, et al. Peripheral artery occlusive disease in chronic phase chronic myeloid leukemia patients treated with nilotinib or imatinib. Leukemia. 2013;27:1316–1321. doi: 10.1038/leu.2013.70. [DOI] [PubMed] [Google Scholar]
  • 49.Ustin C, DeRemer DL, Akin C. Tyrosine kinase inhibitors in the treatment of systemic mastocytosis. Leuk Res. 2011;35:1143–52. doi: 10.1016/j.leukres.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 50.Gleixner KV, Peter B, Blatt K, Suppan V, Reiter A, et al. Synergistic growth-inhibitory effects of ponatinib and midostaurin (PKC412) on neoplastic mast cells carrying KIT D816V. Haematologica. 2013;98:1450–7. doi: 10.3324/haematol.2012.079202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Verstovsek S, Akin C, Manshouri T, Quintas-Cardama A, Huynh L, et al. Effects of AMN107, a novel aminopyrimidine tyrosine kinase inhibitor, on human mast cells bearing wild-type or mutated codon 816 c-kit. Leuk Res. 2006;30:1365–70. doi: 10.1016/j.leukres.2006.04.005. [DOI] [PubMed] [Google Scholar]
  • 52.Klion A, 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;103:2939–41. doi: 10.1182/blood-2003-10-3620. [DOI] [PubMed] [Google Scholar]
  • 53.Kolbeck R, Kozhich A, Koike M, Peng L, Andersson CK, 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;125:1344–53. doi: 10.1016/j.jaci.2010.04.004. [DOI] [PubMed] [Google Scholar]
  • 54.Laviolette M, Gossage DL, Gauvreau G, Leigh R, Olivenstein R, et al. Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol. 2013;132:1086–96. doi: 10.1016/j.jaci.2013.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Legrand F, Tomasevic N, Simakova O, Lee CCR, Wang Z, et al. The eosinophil surface receptor epidermal growth factor-like module containing mucin-like hormone receptor 1 (EMR1): A novel therapeutic target for eosinophilic disorders. J Allergy Clin Immunol. 2014 doi: 10.1016/j.jaci.2013.11.041. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kiwamoto T, Kawasaki N, Paulson JC, Bochner BS. Siglec-8 as a drugable target to treat eosinophil and mast cell-associated conditions. Pharmacol Ther. 2012;135:327–36. doi: 10.1016/j.pharmthera.2012.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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]
  • 58.Zimmermann N, McBride ML, Yamada Y, Hudson SA, Jones C, et al. Siglec-F antibody administration to mice selectively reduces blood and tissue eosinophils. Allergy. 2008;63:1156–1163. doi: 10.1111/j.1398-9995.2008.01709.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.von Gunten S, Vogel M, Schaub A, Stadler BM, Miescher S, et al. Intravenous immunoglobulin preparations contain anti-Siglec-8 autoantibodies. J Allergy Clin Immunol. 2007;119:1005–11. doi: 10.1016/j.jaci.2007.01.023. [DOI] [PubMed] [Google Scholar]
  • 60.Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest. 2009;119:1109–23. doi: 10.1172/JCI35660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Salemi S, Yousefi S, Constantinescu MA, Fey MF, Simon HU. Autophagy is required for self-renewal and differentiation of adult human stem cells. Cell Res. 2012;22:432–5. doi: 10.1038/cr.2011.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Goff DJ, Recart AC, Sadarangani A, Chun HJ, Barrett CL, et al. A pan-Bcl2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell. 2013;12:316–28. doi: 10.1016/j.stem.2012.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gandhi V, Plunkett W, Cortes JE. Omacetaxine: a protein translation inhibitor for treatment of chronic myelogenous leukemia. Clin Cancer Res. 2014 doi: 10.1158/1078-0432.CCR-13-1283. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bose P, Park H, Al-Khafaji J, Grant S. Strategies to circumvent the T315I gatekeeper mutation in the Bcr-Abl tyrosine kinase. Leuk Res Rep. 2013;2:18–20. doi: 10.1016/j.lrr.2013.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. PNAS. 2007;104:19512–7. doi: 10.1073/pnas.0709443104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tolcher AW, Rodrigueza WV, Rasco DW, Patnaik A, Papadopoulos KP, et al. A phase 1 study of the BCL2-targeted deoxyribonucleic acid inhibitor (DNAi) PNT2258 in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2014;73:363–71. doi: 10.1007/s00280-013-2361-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Heidel FH, Bullinger L, Feng Z, Wang Z, Neff TA, et al. Genetic and pharmacologic inhibition of β-catenin targets imatinib-resistant leukemia stem cells in CML. Cell Stem Cell. 2012;10:412–24. doi: 10.1016/j.stem.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dihlmann S, Siermann A, von Knebel Doeberitz M. The nonsteroidal anti-inflammatory drugs aspirin and indomethacin attenuate beta-catenin/TCF-4 signaling. Oncogene. 2001;20:645–53. doi: 10.1038/sj.onc.1204123. [DOI] [PubMed] [Google Scholar]
  • 69.Zhang B, Strauss AC, Chu S, Li M, Ho Y, et al. Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate. Cancer Cell. 2010;17:427–42. doi: 10.1016/j.ccr.2010.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li L, Wang L, Li L, Wang Z, Ho Y, et al. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell. 2012;21:266–81. doi: 10.1016/j.ccr.2011.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Brooks CL, Gu W. How does SIRT1 affect metabolism, senescence and cancer? Nat Rev Cancer. 2009;9:123–8. doi: 10.1038/nrc2562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Valent P, Bonnet D, De Maria R, Lapidot T, Copland M, et al. Cancer stem cell definitions and terminology: the devil is in the details. Nat Rev Cancer. 2012;12:767–75. doi: 10.1038/nrc3368. [DOI] [PubMed] [Google Scholar]
  • 73.Florian S, Sonneck K, Hauswirth AW, Krauth MT, Schernthaner GH, et al. Detection of molecular targets on the surface of CD34+/CD38-- stem cells in various myeloid malignancies. Leuk Lymphoma. 2006;47:207–22. doi: 10.1080/10428190500272507. [DOI] [PubMed] [Google Scholar]
  • 74.Weisberg E, Azab AK, Manley PW, Kung AL, Christie AL, et al. Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib. Leukemia. 2012;26:985–90. doi: 10.1038/leu.2011.360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fruehauf S. Current clinical indications for plerixafor. Transfus Med Hemother. 2013;40:246–50. doi: 10.1159/000354229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Huang Mm, Zhu J. The regulation of normal and leukemic hematopoietic stem cells by niches. Cancer Microenvironment. 2012;5:295–305. doi: 10.1007/s12307-012-0114-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nwajei F, Konopleva M. The bone marrow microenvironment as niche retreats for hematopoietic and leukemic stem cells. Adv Hematol. 2013 doi: 10.1155/2013/953982. ID 953982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Benito J, Shi Y, Szymanska B, Carol H, Boehm I, et al. Pronounced hypoxia in models of murine and human leukemia : high efficacy of hypoxia-activated pro-drug PR-104. PLoS One. 2011 doi: 10.1371/journal.pone.0023108. ID e23108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502:637–43. doi: 10.1038/nature12612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bartlett JB, Dredge K, Dalgleish AG. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat Rev Cancer. 2004;4:314–22. doi: 10.1038/nrc1323. [DOI] [PubMed] [Google Scholar]
  • 81.Simon HU, Yousefi S, Dommann-Scherrer CC, Zimmermann DR, Bauer S, et al. Expansion of cytokine-producing CD4−CD8− T cells associated with abnormal Fas expression and hypereosinophilia. J Exp Med. 1996;183:1071–82. doi: 10.1084/jem.183.3.1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Roufosse F, Schandene L, Sibille C, Willard-Gallo K, Kennes B, et al. Clonal Th2 lymphocytes in patients with the idiopathic hypereosinophilic syndrome. Br J Haematol. 2000;109:540–8. doi: 10.1046/j.1365-2141.2000.02097.x. [DOI] [PubMed] [Google Scholar]
  • 83.Simon HU, Seelbach H, Ehmann R, Schmitz M. Clinical and immunological effects of low-dose IFN-alpha treatment in patients with corticosteroid-resistant asthma. Allergy. 2003;58:1250–5. doi: 10.1046/j.1398-9995.2003.00424.x. [DOI] [PubMed] [Google Scholar]
  • 84.Plötz SG, Simon HU, Darsow U, Simon D, Vassina E, 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]
  • 85.Garrett JK, Jameson SC, Thomson B, Collins MH, Wagoner LE, 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]
  • 86.Rothenberg ME, Klion AD, Roufosse FE, Kahn JE, Weller PF, 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]
  • 87.Roufosse F, de Lavareille A, Schandené L, Cogan E, Georgelas A, et al. Mepolizumab as a corticosteroid-sparing agent in lymphocytic variant hypereosinophilic syndrome. J Allergy Clin Immunol. 2010;126:828–35. doi: 10.1016/j.jaci.2010.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Roufosse F, Kahn JE, Gleich GJ, Schwartz LB, Singh AD, et al. Long-term safety of mepolizumab for the treatment of hypereosinophilic syndromes. J Allergy Clin Immunol. 2013;131:461–7. doi: 10.1016/j.jaci.2012.07.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Stein LM, Villanueva JM, Buckmeier BK, Yamada Y, Filipovich AH, et al. Anti-IL-5 (mepolizumab) therapy reduces eosinophil activation ex vivo and increases IL-5 and IL-5 receptor levels. J Allergy Clin Immunol. 2008;121:1473–83. doi: 10.1016/j.jaci.2008.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Conus S, Straumann A, Bettler E, Simon HU. Mepolizumab does not alter levels of eosinophils, T cells, and mast cells in the duodenal mucosa in eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126:175–7. doi: 10.1016/j.jaci.2010.04.029. [DOI] [PubMed] [Google Scholar]
  • 91.Wechsler ME, Fulkerson PC, Bochner BS, Gauvreau GM, Gleich GJ, et al. Novel targeted therapies for eosinophilic disorders. J Allergy Clin Immunol. 2012;130:563–71. doi: 10.1016/j.jaci.2012.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Straumann A, Conus S, Grzonka P, Kita H, Kephart G, 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]
  • 93.Busse WW, Katial R, Gossage D, Sari S, Wang B, et al. Safety profile, pharmacokinetics, and biologic activity of MEDI-563, an anti-IL-5 receptor alpha antibody, in a phase I study of subjects with mild asthma. J Allergy Clin Immunol. 2010;125:1237–44. doi: 10.1016/j.jaci.2010.04.005. [DOI] [PubMed] [Google Scholar]
  • 94.Na HJ, Hamilton RG, Klion AD, Bochner BS. Biomarkers of eosinophil involvement in allergic and eosinophilic diseases: review of phenotypic and serum markers including a novel assay to quantify levels of soluble Siglec-8. J Immunol Meth. 2012;383:39–46. doi: 10.1016/j.jim.2012.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Verstovsek S, Tefferi A, Kantarjian H, Mashouri T, Luthra R, 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]
  • 96.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]
  • 97.Miller GT, Hochman PS, Meier W, Tozard R, Bixler S, et al. Specific interaction of lymphocyte function-associated antigen 3 with CD2 can inhibit T cell responses. J Exp Med. 1993;178:211–22. doi: 10.1084/jem.178.1.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Simon D, Wittwer J, Kostylina G, Büttiker U, Simon HU, Yawalkar N. Alefacept (LFA-3/IgG fusion protein) treatment for atopic eczema? J Allergy Clin Immunol. 2008;122:423–4. doi: 10.1016/j.jaci.2008.06.010. [DOI] [PubMed] [Google Scholar]
  • 99.Ellis CN, Mordin MM, Adler EY. Effects of alefacept on health-related quality of life in patients with psoriasis: results from a randomized, placebo-controlled phase II trial. Am J Clin Dermatol. 2003;4:131–9. doi: 10.2165/00128071-200304020-00005. [DOI] [PubMed] [Google Scholar]
  • 100.Burks AW, Calderon MA, Casale T, Cox L, Demoly P, et al. Update on allergen immunotherapy: Academy of Allergy Asthma and Immunology/European Academy of Allergy and Clinical Immunology/PRACTALL consensus report. J Allergy Clin Immunol. 2013;131:1288–96. doi: 10.1016/j.jaci.2013.01.049. [DOI] [PubMed] [Google Scholar]
  • 101.Barnes N, Menzies-Gow A, Mansur AH, Spencer D, Percival F, et al. Effectiveness of omalizumab in severe allergic asthma: a retrospective UK real-world study. J Asthma. 2013;50:529–36. doi: 10.3109/02770903.2013.790419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hanania NA, Wenzel S, Rosén K, Hsieh HJ, Mosesova S, et al. Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am J Respir Crit Care Med. 2013;187:804–11. doi: 10.1164/rccm.201208-1414OC. [DOI] [PubMed] [Google Scholar]
  • 103.Bousquet J, Wenzel S, Holgate S, Lumry W, Freeman P, Fox H. Predicting response to omalizumab an anti-IgE antibody in patients with allergic asthma. Chest. 2004;125:1378–86. doi: 10.1378/chest.125.4.1378. [DOI] [PubMed] [Google Scholar]
  • 104.Gevaert P, Calus L, Van Zele T, Blomme K, De Ruyck N, et al. Omalizumab is effective in allergic and nonallergic patients with nasal polyps and asthma. J Allergy Clin Immunol. 2013;131:110–6. doi: 10.1016/j.jaci.2012.07.047. [DOI] [PubMed] [Google Scholar]
  • 105.Kaplan A, Ledford D, Ashby M, Canvin J, Zazzali JL, et al. Omalizumab in patients with symptomatic chronic idiopathic/spontaneous urticaria despite standard combination therapy. J Allergy Clin Immunol. 2013;132:101–9. doi: 10.1016/j.jaci.2013.05.013. [DOI] [PubMed] [Google Scholar]
  • 106.Cruz AA, Lima F, Sarinho E, Ayre G, Martin C, Fox H, Cooper PJ. Safety of anti-immunoglobulin E therapy with omalizumab in allergic patients at risk of geohelminth infection. Clin Exp Allergy. 2007;37:197–207. doi: 10.1111/j.1365-2222.2007.02650.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Presta LG, Lahr SJ, Shields RL, Porter JP, Gorman CM, et al. Humanization of an antibody directed against IgE. J Immunol. 1993;151:2623–32. [PubMed] [Google Scholar]
  • 108.MacGlashan DW, Jr, Bochner BS, Adelman DC, Jardieu PM, Togias A, et al. Down-regulation of Fc(epsilon)RI expression on human basophils during in vivo treatment of atopic patients with anti-IgE antibody. J Immunol. 1997;158:1438–45. [PubMed] [Google Scholar]
  • 109.Noga O, Hanf G, Brachmann I, Klucken AC, Kleine-Tebbe J, et al. Effect of omalizumab treatment on peripheral eosinophil and T-lymphocyte function in patients with allergic asthma. J Allergy Clin Immunol. 2006;117:1493–9. doi: 10.1016/j.jaci.2006.02.028. [DOI] [PubMed] [Google Scholar]
  • 110.Plewako H, Arvidsson M, Petruson K, Oancea I, Holmberg K, et al. The effect of omalizumab on nasal allergic inflammation. J Allergy Clin Immunol. 2002;110:68–71. doi: 10.1067/mai.2002.125488. [DOI] [PubMed] [Google Scholar]
  • 111.Chan MA, Gigliotti NM, Dotson AL, Rosenwasser LJ. Omalizumab may decrease IgE synthesis by targeting membrane IgE+ human B cells. Clin Transl Allergy. 2013;3:29. doi: 10.1186/2045-7022-3-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Iyengar SR, Hoyte EG, Loza A, Bonaccorso S, Chiang D, et al. Immunologic effects of omalizumab in children with severe refractory atopic dermatitis: a randomized, placebo-controlled clinical trial. Int Arch Allergy Immunol. 2013;162:89–93. doi: 10.1159/000350486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kim B, Eggel A, Tarchevskaya SS, Vogel M, Prinz H, et al. Accelerated disassembly of IgE-receptor complexes by a disruptive macromolecular inhibitor. Nature. 2012;491:613–7. doi: 10.1038/nature11546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Leckie MJ, ten Brinke A, Khan J, Diamant Z BJ, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356:2144–8. doi: 10.1016/s0140-6736(00)03496-6. [DOI] [PubMed] [Google Scholar]
  • 115.Kips JC, O’Connor BJ, Langley SJ, Woodcock A, Kerstjens HA, et al. Effect of SCH55700, a humanized anti-human interleukin-5 antibody, in severe persistent asthma: a pilot study. Am J Respir Crit Care Med. 2003;167:1655–9. doi: 10.1164/rccm.200206-525OC. [DOI] [PubMed] [Google Scholar]
  • 116.Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360:973–84. doi: 10.1056/NEJMoa0808991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Nair P, Pizzichini MM, Kjarsgaard M, Inman MD, Efthimiadis A, et al. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N Engl J Med. 2009;360:985–93. doi: 10.1056/NEJMoa0805435. [DOI] [PubMed] [Google Scholar]
  • 118.Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet. 2012;380:651–9. doi: 10.1016/S0140-6736(12)60988-X. [DOI] [PubMed] [Google Scholar]
  • 119.Castro M, Mathur S, Hargreave F, Boulet LP, Xie F, et al. Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study. Am J Respir Crit Care Med. 2011;184:1125–32. doi: 10.1164/rccm.201103-0396OC. [DOI] [PubMed] [Google Scholar]
  • 120.Vaglio A, Buzio C, Zwerina J. Eosinophilic granulomatosis with polyangiitis (Churg-Strauss): state of the art. Allergy. 2013;68:261–73. doi: 10.1111/all.12088. [DOI] [PubMed] [Google Scholar]
  • 121.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]
  • 122.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]
  • 123.Herrmann K, Gross WL, Moosig F. Extended follow-up after stopping mepolizumab in relapsing/refractory Churg-Strauss syndrome. Clin Exp Rheumatol. 2012;30(1 Suppl 70):S62–5. [PubMed] [Google Scholar]
  • 124.Gevaert P, Lang-Loidolt D, Lackner A, Stammberger H, Staudinger H, et al. Nasal IL-5 levels determine the response to anti-IL-5 treatment in patients with nasal polyps. J Allergy Clin Immunol. 2006;118:1133–41. doi: 10.1016/j.jaci.2006.05.031. [DOI] [PubMed] [Google Scholar]
  • 125.Assa’ad AH, Gupta SK, Collins MH, Thomson M, Heath AT, 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]
  • 126.Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, 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. doi: 10.1016/j.jaci.2011.11.044. [DOI] [PubMed] [Google Scholar]
  • 127.Oldhoff JM, Darsow U, Werfel T, Katzer K, Wulf A, et al. Anti-IL-5 recombinant humanized monoclonal antibody (mepolizumab) for the treatment of atopic dermatitis. Allergy. 2005;60:693–6. doi: 10.1111/j.1398-9995.2005.00791.x. [DOI] [PubMed] [Google Scholar]
  • 128.Fulkerson PC, Fischetti CA, McBride M, Hassman LM, Hogan SP, Rothenberg ME. A central regulatory role for eosinophils and the eotaxin/CCR3 axis in chronic experimental allergic airway inflammation. PNAS. 2006;103:16418–23. doi: 10.1073/pnas.0607863103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lampinen M, Waddell A, Ahrens R, Carlson M, Hogan SP. CD14+CD33+ myeloid cell-CCL11-eosinophil signature in ulcerative colitis. J Leukoc Biol. 2013;94:1061–70. doi: 10.1189/jlb.1212640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Bahl A, Springthorpe B, Riley R. Chemokine CCR3 antagonists. In: Hansel TT, Barnes PJ, editors. New drugs and targets for asthma and COPD. Basel: Karger; 2010. pp. 153–9. [Google Scholar]
  • 131.Imaoka H, Campbell H, Babirad I, Watson RM, Mistry M, et al. TPI ASM8 reduces eosinophil progenitors in sputum after allergen challenge. Clin Exp Allergy. 2011;41:1740–6. doi: 10.1111/j.1365-2222.2011.03816.x. [DOI] [PubMed] [Google Scholar]
  • 132.Gauvreau GM, Pageau R, Séguin R, Carballo D, Gauthier J, et al. Dose-response effects of TPI ASM8 in asthmatics after allergen. Allergy. 2011;66:1242–8. doi: 10.1111/j.1398-9995.2011.02638.x. [DOI] [PubMed] [Google Scholar]
  • 133.Ziegler SF. Thymic stromal lymphopoietin (TSLP) and allergic disease. J Allergy Clin Immunol. 2012;130:845–52. doi: 10.1016/j.jaci.2012.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3:673–80. doi: 10.1038/ni805. [DOI] [PubMed] [Google Scholar]
  • 135.Headley MB, Zhou B, Shih WX, Aye T, Comeau MR, Ziegler SF. TSLP conditions the lung immune environment for the generation of pathogenic innate and antigen-specific adaptive immune responses. J Immunol. 2009;182:1641–7. doi: 10.4049/jimmunol.182.3.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Straumann A, Aceves SS, Blanchard C, Collins MH, Furuta GT, et al. Pediatric and adult eosinophilic esophagitis: similarities and differences. Allergy. 2012;67:477–90. doi: 10.1111/j.1398-9995.2012.02787.x. [DOI] [PubMed] [Google Scholar]
  • 137.Morshed M, Yousefi S, Stöckle C, Simon HU, Simon D. Thymic stromal lymphopoietin stimulates the formation of eosinophil extracellular traps. Allergy. 2012;67:1127–37. doi: 10.1111/j.1398-9995.2012.02868.x. [DOI] [PubMed] [Google Scholar]
  • 138.Noti M, Wojno ED, Kim BS, Siracusa MC, Giacomin PR, et al. Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat Med. 2013;19:1005–13. doi: 10.1038/nm.3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M. Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet. 2007;370:1422–31. doi: 10.1016/S0140-6736(07)61600-6. [DOI] [PubMed] [Google Scholar]
  • 140.Tomkinson A, Tepper J, Morton M, Bowden A, Stevens L, et al. Inhaled vs subcutaneous effects of a dual IL-4/IL-13 antagonist in a monkey model of asthma. Allergy. 2010;65:69–77. doi: 10.1111/j.1398-9995.2009.02156.x. [DOI] [PubMed] [Google Scholar]
  • 141.Slager RE, Hawkins GA, Ampleford EJ, Bowden A, Stevens LE, et al. IL-4 receptor α polymorphisms are predictors of a pharmacogenetic response to a novel IL-4/IL-13 antagonist. J Allergy Clin Immunol. 2010;126:875–8. doi: 10.1016/j.jaci.2010.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Wenzel S, Ford L, Pearlman D, Spector S, Sher L, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368:2455–66. doi: 10.1056/NEJMoa1304048. [DOI] [PubMed] [Google Scholar]
  • 143.Spiess C, Bevers J, 3rd, Jackman J, Chiang N, Nakamura G, et al. Development of a human IgG4 bispecific antibody for dual targeting of interleukin-4 (IL-4) and interleukin-13 (IL-13) cytokines. J Biol Chem. 2013;288:26583–93. doi: 10.1074/jbc.M113.480483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Agrawal S, Townley RG. Role of periostin, FENO, IL-13, lebrikizumab, other IL-13 antagonist and dual IL-4/IL-13 antagonist in asthma. Exp Opin Biol Ther. 2014;14:165–81. doi: 10.1517/14712598.2014.859673. [DOI] [PubMed] [Google Scholar]
  • 145.Scheerens H, Arron JR, Zheng Y, Putnam WS, Erickson RW, et al. The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Clin Exp Allergy. 2014;44:38–46. doi: 10.1111/cea.12220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Piper E, Brightling C, Niven R, Oh C, Faggioni R, et al. A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. EurRespir J. 2012;41:330–8. doi: 10.1183/09031936.00223411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Arima M, Fukuda T. Prostaglandin D₂ and T(H)2 inflammation in the pathogenesis of bronchial asthma. Korean J Intern Med. 2011;26:8–18. doi: 10.3904/kjim.2011.26.1.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Spik I, Brénuchon C, Angéli V, Staumont D, Fleury S, et al. Activation of the prostaglandin D2 receptor DP2/CRTH2 increases allergic inflammation in mouse. J Immunol. 2005;174:3703–8. doi: 10.4049/jimmunol.174.6.3703. [DOI] [PubMed] [Google Scholar]
  • 149.Satoh T, Moroi R, Aritake K, Urade Y, Kanai Y, et al. Prostaglandin D2 plays an essential role in chronic allergic inflammation of the skin via CRTH2 receptor. J Immunol. 2006;177:2621–9. doi: 10.4049/jimmunol.177.4.2621. [DOI] [PubMed] [Google Scholar]
  • 150.Barnes N, Pavord I, Chuchalin A, Bell J, Hunter M, et al. A randomized, double-blind, placebo-controlled study of the CRTH2 antagonist OC000459 in moderate persistent asthma. Clin Exp Allergy. 2012;42:38–48. doi: 10.1111/j.1365-2222.2011.03813.x. [DOI] [PubMed] [Google Scholar]
  • 151.Straumann A, Hoesli S, Bussmann Ch, Stuck M, Perkins M, et al. Anti-eosinophil activity and clinical efficacy of the CRTH2 antagonist OC000459 in eosinophilic esophagitis. Allergy. 2013;68:375–85. doi: 10.1111/all.12096. [DOI] [PubMed] [Google Scholar]
  • 152.Horak F, Zieglmayer P, Zieglmayer R, Lemell P, Collins LP, et al. The CRTH2 antagonist OC000459 reduces nasal and ocular symptoms in allergic subjects exposed to grass pollen, a randomised, placebo-controlled, double-blind trial. Allergy. 2012;67:1572–9. doi: 10.1111/all.12042. [DOI] [PubMed] [Google Scholar]
  • 153.Fuhst R, Runge F, Buschmann J, Ernst H, Praechter C, et al. Toxicity profile of the GATA-3-specific DNAzyme hgd40 after inhalation exposure. Pulm Pharmacol Ther. 2013;26:281–9. doi: 10.1016/j.pupt.2012.12.005. [DOI] [PubMed] [Google Scholar]
  • 154.Sel S, Wegmann M, Dicke T, Sel S, Henke W, et al. Effective prevention and therapy of experimental allergic asthma using a GATA-3-specific DNAzyme. J Allergy Clin Immunol. 2008;121:910–6. doi: 10.1016/j.jaci.2007.12.1175. [DOI] [PubMed] [Google Scholar]
  • 155.Simon D, Hösli S, Kostylina G, Yawalkar N, Simon HU. Anti-CD20 (rituximab) treatment improves atopic eczema. J Allergy Clin Immunol. 2008;121:122–8. doi: 10.1016/j.jaci.2007.11.016. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Table 1

RESOURCES