Reviews and feature article
Novel targeted therapies for eosinophilic disorders

https://doi.org/10.1016/j.jaci.2012.07.027Get rights and content

Hypereosinophilic syndromes (HESs) are a diverse group of conditions characterized by clinical manifestations attributable to eosinophilia and eosinophilic infiltration of tissues. HESs are chronic disorders with significant morbidity and mortality. Although the availability of targeted chemotherapeutic agents, including imatinib, has improved quality of life and survival in some patients with HESs, additional agents with increased efficacy and decreased toxicity are sorely needed. The purpose of this review is to provide an overview of eosinophil biology with an emphasis on potential targets of pharmacotherapy and to provide a summary of potential eosinophil-targeting agents, including those in development, in clinical trials, or approved for other disorders.

Section snippets

Eosinophil ontogeny

As is true of all hematopoietic cells, eosinophils differentiate from CD34+ multipotential myeloid progenitors in the bone marrow. These myeloid progenitors give rise to CD34+ cells that also express GATA-1 and IL-5 receptor (IL-5R) α.3, 4 The CD34+IL-5Rα+ eosinophil-committed progenitors undergo further development in response to IL-3, GM-CSF, and IL-5, the most lineage-specific of the cytokines involved in eosinophil hematopoiesis.3, 5 It is because of this lineage specificity that therapies

IL-5–related targets

Targeting IL-5 (or IL-5R) is an appealing approach to the treatment of patients with all types of HESs, given the specificity of this cytokine for the eosinophil lineage and the assumption that tissue damage in patients with HESs is directly related to the presence of activated eosinophils. Anti–IL-5 antibodies target eosinophils by binding to IL-5, interfering with its ligation to IL-5Rα expressed on the eosinophil membrane. Two different humanized anti–IL-5 antibodies, mepolizumab and

Eosinophil survival

There are numerous preclinical pathways that are currently being pursued for the treatment of eosinophil-associated diseases. Interfering with eosinophil-inhibitory receptors involves direct engagement of inhibitory receptors with activating ligands (including antibodies) that result in impairment of eosinophil functional responses or survival signals or directly induce apoptosis. For example, anti–Siglec-8 (or anti–Siglec-F in the mouse) has the capacity to induce direct eosinophil apoptosis

Summary and future directions

Eosinophilic disorders are chronic conditions that require long-term treatment for the prevention of clinical manifestations. Morbidity and mortality for many eosinophilic disorders remain high, and current treatment options are limited by lack of efficacy, significant toxicity, or both. Recent advances in our understanding of eosinophil biology have paved the way for the development of several promising novel therapies. Although clinical trial development in these therapeutic areas has been

References (82)

  • J.K. Garrett et al.

    Anti-interleukin-5 (mepolizumab) therapy for hypereosinophilic syndromes

    J Allergy Clin Immunol

    (2004)
  • S. Kim et al.

    Mepolizumab as a steroid-sparing treatment option in patients with Churg-Strauss syndrome

    J Allergy Clin Immunol

    (2010)
  • A.H. Assa'ad et al.

    An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis

    Gastroenterology

    (2011)
  • M.L. Stein et al.

    Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis

    J Allergy Clin Immunol

    (2006)
  • P. Gevaert et al.

    Nasal IL 5 levels determine the response to anti-IL 5 treatment in patients with nasal polyps

    J Allergy Clin Immunol

    (2006)
  • A.D. Klion et al.

    Safety and efficacy of the monoclonal anti-interleukin-5 antibody SCH55700 in the treatment of patients with hypereosinophilic syndrome

    Blood

    (2004)
  • D. Simon et al.

    Eosinophilic diseases

    J Allergy Clin Immunol

    (2007)
  • L.A. Wagner et al.

    Treatment of FIP1L1/PDGFRA-negative hypereosinophilic syndrome with alemtuzumab, an anti-CD52 antibody

    J Allergy Clin Immunol

    (2009)
  • F.F. Syed et al.

    Response to alemtuzumab in FIP1L1/PDGFRA-negative hypereosinophilic myocarditis on serial cardiac magnetic resonance imaging

    J Am Coll Cardiol

    (2012)
  • D. Simon et al.

    Alefacept (LFA-3/IgG fusion protein) treatment for atopic eczema

    J Allergy Clin Immunol

    (2008)
  • D.K. Moul et al.

    Alefacept for moderate to severe atopic dermatitis: a pilot study in adults

    J Am Acad Dermatol

    (2008)
  • S. Holgate et al.

    The anti-inflammatory effects of omalizumab confirm the central role of IgE in allergic inflammation

    J Allergy Clin Immunol

    (2005)
  • M. Massanari et al.

    Effect of omalizumab on peripheral blood eosinophilia in allergic asthma

    Respir Med

    (2010)
  • S. Wenzel et al.

    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)
  • E. Nutku et al.

    Ligation of Siglec-8: a selective mechanism for induction of human eosinophil apoptosis

    Blood

    (2003)
  • A. Munitz et al.

    The inhibitory receptor IRp60 (CD300a) suppresses the effects of IL-5, GM-CSF, and eotaxin on human peripheral blood eosinophils

    Blood

    (2006)
  • A. Munitz et al.

    A dual activation and inhibition role for the paired immunoglobulin-like receptor B in eosinophils

    Blood

    (2008)
  • M. Komai et al.

    A novel CC-chemokine receptor 3 antagonist, Ki19003, inhibits airway eosinophilia and subepithelial/peribronchial fibrosis induced by repeated antigen challenge in mice

    J Pharmacol Sci

    (2010)
  • H. Okigami et al.

    Inhibition of eosinophilia in vivo by a small molecule inhibitor of very late antigen (VLA)-4

    Eur J Pharmacol

    (2007)
  • P.U. Ogbogu et al.

    Hypereosinophilic syndromes: a multicenter, retrospective analysis of clinical characteristics and response to therapy

    J Allergy Clin Immunol

    (2009)
  • M.E. Rothenberg et al.

    Treatment of patients with the hypereosinophilic syndrome with mepolizumab

    N Engl J Med

    (2008)
  • Y. Mori et al.

    Identification of the human eosinophil lineage-committed progenitor: revision of phenotypic definition of the human common myeloid progenitor

    J Exp Med

    (2009)
  • H. Iwasaki et al.

    Identification of eosinophil lineage-committed progenitors in the murine bone marrow

    J Exp Med

    (2005)
  • M.E. Rothenberg et al.

    The eosinophil

    Ann Rev Immunol

    (2006)
  • M. Milanovic et al.

    IFN consensus sequence binding protein (Icsbp) is critical for eosinophil development

    J Immunol

    (2008)
  • D.E. Zhang et al.

    Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein alpha-deficient mice

    Proc Natl Acad Sci U S A

    (1997)
  • B.S. Bochner

    Verdict in the case of therapies versus eosinophils: the jury is still out

    J Allergy Clin Immunol

    (2004)
  • S. von Gunten et al.

    Appendix A. CD molecules

  • P.F. Weller

    Lipid, peptide and cytokine mediators elaborated by eosinophils

  • S. Feltenmark et al.

    Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells

    Proc Natl Acad Sci U S A

    (2008)
  • R. Moqbel et al.

    Differential secretion of cytokines

    Sci STKE

    (2006)
  • Cited by (0)

    Series editors: Donald Y. M. Leung, MD, PhD, and Dennis K. Ledford, MD

    Supported in part by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases/National Institutes of Health (NIH), and by the Office of Rare Diseases/NIH.

    Disclosure of potential conflict of interest: M. E. Wechsler has received research support from the National Institutes of Health (NIH); has received consulting fees from GlaxoSmithKline, Cephalon, Novartis, Sepracor/Sunovion, Schering-Plough, NKT Therapeutics, Asthmatx/BSCI, Genzyme, MapPharma, Genentech, and Boehringer Ingelheim; and has received honoraria from Merck. B. S. Bochner has received grants from the National Institute of Allergy and Infectious Diseases and the National Heart, Lung, and Blood Institute; has consultant arrangements with Sanofi-Aventis, Genentech, Merck, Roche, GlaxoSmithKline, TEVA, GlycoFi, and Medicis; is on a scientific advisory board for Merck; is a coinventor on existing and pending Siglec-8–related patents; might be entitled to a share of royalties received by his university on the potential sales of Siglec-8–related products but thus far has received no such royalties; receives royalties from UpToDate and Elsevier; is a cofounder of and has purchased stock in Allakos; and holds stock options for Glycomimetics. G. M. Gauvreau has received research support from Topigen Pharma. G. J. Gleich is a board member of the American Partnership for Eosinophilic Disorders (APFED) without compensation; has received consultancy fees from GlaxoSmithKline; has received research support from TRIA Bioscience Corp and ImmViz; has a patent with ImmViz; has received royalties from Teva; and has stock/stock options in Immune Design Corp. T. Henkel has received consultancy fees from Cephalon and is employed by and has received stock/stock options in Ception Therapeutics. R. Kolbeck is employed by and owns stock/stock options in MedImmune. S. K. Mathur has received consultancy fees from Teva Pharmaceuticals, is employed by the University of Wisconsin and William S Middleton VA Hospital, and has received research support from the NIH. H. Ortega is employed by and has received stock/stock options in GlaxoSmithKline. P. Renzi has received research support, consulting fees, and fees for participation in review activities from Pharmaxis and has received royalties and owns a patent as the inventor of TPI-ASM8. M. E. Rothenberg is a member of the board for APFED and the International Eosinophil Society, has received consultancy fees from Immune Pharmaceuticals, is the inventor of patents owned by Cincinnati Children's, and has stock/stock options in Immune Pharmaceuticals and Teva. F. Roufosse has received consulting fees and travel support from GlaxoSmithKline and received royalties from UpToDate Online. D. Simon has provided expert testimony for Basilea Pharmaceutica and Astellas Schweiz, has received research support from OPO Foundation Zurich, and has received lecture fees from Basilea Pharmaceutica. H.-U. Simon has received consultancy fees from Pfizer. P. F. Weller has received consultancy fees from GlaxoSmithKline and receives royalties from UpToDate. A. Wardlaw was a member of the Cephalon-Teva data monitoring board; has received research support from Pfizer and GlaxoSmithKline; and has provided legal consultation/expert witness testimony for Bayer. The rest of the authors declare that they have no relevant conflicts of interest.

    View full text