1
|
Vogel M, Engeroff P. A Comparison of Natural and Therapeutic Anti-IgE Antibodies. Antibodies (Basel) 2024; 13:58. [PMID: 39051334 PMCID: PMC11270207 DOI: 10.3390/antib13030058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/17/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Immunoglobulin E (IgE) plays a critical role for the immune system, fighting against parasites, toxins, and cancer. However, when it reacts to allergens without proper regulation, it can cause allergic reactions, including anaphylaxis, through a process initiated by effector cells such as basophils and mast cells. These cells display IgE on their surface, bound to the high-affinity IgE receptor FcεRI. A cross-linking antigen then triggers degranulation and the release of inflammatory mediators from the cells. Therapeutic monoclonal anti-IgE antibodies such as omalizumab, disrupt this process and are used to manage IgE-related conditions such as severe allergic asthma and chronic spontaneous urticaria. Interestingly, naturally occurring anti-IgE autoantibodies circulate at surprisingly high levels in healthy humans and mice and may thus be instrumental in regulating IgE activity. Although many open questions remain, recent studies have shed new light on their role as IgE regulators and their mechanism of action. Here, we summarize the latest insights on natural anti-IgE autoantibodies, and we compare their functional features to therapeutic monoclonal anti-IgE autoantibodies.
Collapse
Affiliation(s)
- Monique Vogel
- Department of Rheumatology and Immunology, University Hosptial of Bern, 3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
| | - Paul Engeroff
- Department of Rheumatology and Immunology, University Hosptial of Bern, 3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
2
|
Cohen JR, Brych SR, Prabhu S, Bi V, Elbaradei A, Tokuda JM, Xiang C, Hokom M, Cui X, Ly C, Amos N, Sun J, Calamba D, Herskovitz J, Capili A, Nourbakhsh K, Merlo A, Carreon J, Wypych J, Narhi LO, Jawa V, Joubert MK. A High Threshold of Biotherapeutic Aggregate Numbers is Needed to Induce an Immunogenic Response In Vitro, In Vivo, and in the Clinic. Pharm Res 2024; 41:651-672. [PMID: 38519817 DOI: 10.1007/s11095-024-03678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/15/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND PURPOSE There is concern that subvisible aggregates in biotherapeutic drug products pose a risk to patient safety. We investigated the threshold of biotherapeutic aggregates needed to induce immunogenic responses. METHODS AND RESULTS Highly aggregated samples were tested in cell-based assays and induced cellular responses in a manner that depended on the number of particles. The threshold of immune activation varied by disease state (cancer, rheumatoid arthritis, allergy), concomitant therapies, and particle number. Compared to healthy donors, disease state patients showed an equal or lower response at the late phase (7 days), suggesting they may not have a higher risk of responding to aggregates. Xeno-het mice were used to assess the threshold of immune activation in vivo. Although highly aggregated samples (~ 1,600,000 particles/mL) induced a weak and transient immunogenic response in mice, a 100-fold dilution of this sample (~ 16,000 particles/mL) did not induce immunogenicity. To confirm this result, subvisible particles (up to ~ 18,000 particles/mL, containing aggregates and silicone oil droplets) produced under representative administration practices (created upon infusion of a drug product through an IV catheter) did not induce a response in cell-based assays or appear to increase the rate of adverse events or immunogenicity during phase 3 clinical trials. CONCLUSION The ability of biotherapeutic aggregates to elicit an immune response in vitro, in vivo, and in the clinic depends on high numbers of particles. This suggests that there is a high threshold for aggregates to induce an immunogenic response which is well beyond that seen in standard biotherapeutic drug products.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| | - Stephen R Brych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vivian Bi
- The Department of Biosimilars, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Ahmed Elbaradei
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Joshua M Tokuda
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Martha Hokom
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of BioAnalytical Sciences, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Xiaohong Cui
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Claudia Ly
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Nathan Amos
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jilin Sun
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Dominador Calamba
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Jonathan Herskovitz
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Allyson Capili
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Kimya Nourbakhsh
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Julia Carreon
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jette Wypych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Linda O Narhi
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vibha Jawa
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
3
|
Tiligada E, Gafarov D, Zaimi M, Vitte J, Levi-Schaffer F. Novel Immunopharmacological Drugs for the Treatment of Allergic Diseases. Annu Rev Pharmacol Toxicol 2024; 64:481-506. [PMID: 37722722 DOI: 10.1146/annurev-pharmtox-051623-091038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
The exponential rise in the prevalence of allergic diseases since the mid-twentieth century has led to a genuine public health emergency and has also fostered major progress in research on the underlying mechanisms and potential treatments. The management of allergic diseases benefits from the biological revolution, with an array of novel immunomodulatory therapeutic and investigational tools targeting players of allergic inflammation at distinct pathophysiological steps. Prominent examples include therapeutic monoclonal antibodies against cytokines, alarmins, and their receptors, as well as small-molecule modifiers of signal transduction mainly mediated by Janus kinases and Bruton's tyrosine kinases. However, the first-line therapeutic options have yet to switch from symptomatic to disease-modifying interventions. Here we present an overview of available drugs in the context of our current understanding of allergy pathophysiology, identify potential therapeutic targets, and conclude by providing a selection of candidate immunopharmacological molecules under investigation for potential future use in allergic diseases.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Daria Gafarov
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| | - Maria Zaimi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Joana Vitte
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
- Desbrest Institute of Epidemiology and Public Health, University of Montpellier, INSERM
- Montpellier, France
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel;
| |
Collapse
|
4
|
Storni F, Vogel M, Bachmann MF, Engeroff P. IgG in the control of FcεRI activation: a battle on multiple fronts. Front Immunol 2024; 14:1339171. [PMID: 38274816 PMCID: PMC10808611 DOI: 10.3389/fimmu.2023.1339171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
The rising global incidence of IgE-mediated allergic reactions poses a significant challenge to the quality of life of affected individuals and to healthcare systems, with current treatments being limited in effectiveness, safety, and disease-modifying capabilities. IgE acts by sensitizing the high-affinity IgE receptor FcεRI expressed by mast cells and basophils, tuning these cells for inflammatory degranulation in response to future allergen encounters. In recent years, IgG has emerged as an essential negative regulator of IgE-dependent allergic inflammation. Mechanistically, studies have proposed different pathways by which IgG can interfere with the activation of IgE-mediated inflammation. Here, we briefly summarize the major proposed mechanisms of action by which IgG controls the IgE-FcεRI inflammatory axis and how those mechanisms are currently applied as therapeutic interventions for IgE-mediated inflammation.
Collapse
Affiliation(s)
- Federico Storni
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Monique Vogel
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| | - Paul Engeroff
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
5
|
Furci F, Luxi N, Senna G, Trifirò G. Anaphylaxis due to antiallergic and antiasthmatic biologics. Curr Opin Allergy Clin Immunol 2023; 23:364-369. [PMID: 37555938 DOI: 10.1097/aci.0000000000000937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
PURPOSE OF REVIEW To provide a better understanding of the risk of anaphylaxis due to antiallergic and antiasthmatic biologics through an analysis of data reported in literature and in clinical trials, and by conducting a retrospective descriptive analysis of individual case safety reports on VigiBase, the WHO International Pharmacovigilance database. RECENT FINDINGS Analysis of the data, as described, demonstrated safety of the antiallergic and antiasthmatic biologics with a low incidence of anaphylaxis. SUMMARY Biologic therapies have revolutionized the treatment of many diseases, such as atopic dermatitis, nasal polyps, spontaneous chronic urticarial and severe asthma with a precise immunological action, in the sphere of precision medicine.Albeit these drugs are generally well tolerated, generating real-world evidence is crucial to re-evaluate clinically relevant adverse events, such as anaphylaxis, allowing to confirm their safety profile in particular in special populations such as paediatric patients.
Collapse
Affiliation(s)
- Fabiana Furci
- Provincial Healthcare Unit, Section of Allergy, Vibo Valentia
| | | | - Gianenrico Senna
- Department of Medicine, University of Verona
- Allergy Unit and Asthma Center, Verona University Hospital
| | - Gianluca Trifirò
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
6
|
Terán MG, García-Ramírez B, Mares-Mejía I, Ortega E, O’Malley A, Chruszcz M, Rodríguez-Romero A. Molecular Basis of Plant Profilins' Cross-Reactivity. Biomolecules 2023; 13:608. [PMID: 37189355 PMCID: PMC10135586 DOI: 10.3390/biom13040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Profilins are ubiquitous allergens with conserved structural elements. Exposure to profilins from different sources leads to IgE-cross-reactivity and the pollen-latex-food syndrome. Monoclonal antibodies (mAbs) that cross-react with plant profilins and block IgE-profilin interactions are relevant for diagnosis, epitope mapping, and specific immunotherapy. We generated IgGs mAbs, 1B4, and 2D10, against latex profilin (anti-rHev b 8) that inhibit the interaction of IgE and IgG4 antibodies from sera of latex- and maize-allergic patients by 90% and 40%, respectively. In this study, we evaluated 1B4 and 2D10 recognition towards different plant profilins, and mAbs recognition of rZea m 12 mutants by ELISAs. Interestingly, 2D10 highly recognized rArt v 4.0101 and rAmb a 8.0101, and to a lesser extent rBet v 2.0101, and rFra e 2.2, while 1B4 showed recognition for rPhl p 12.0101 and rAmb a 8.0101. We demonstrated that residue D130 at the α-helix 3 in profilins, which is part of the Hev b 8 IgE epitope, is essential for the 2D10 recognition. The structural analysis suggests that the profilins containing E130 (rPhl p 12.0101, rFra e 2.2, and rZea m 12.0105) show less binding with 2D10. The distribution of negative charges on the profilins' surfaces at the α-helices 1 and 3 is relevant for the 2D10 recognition, and that may be relevant to explain profilins' IgE cross-reactivity.
Collapse
Affiliation(s)
- María G. Terán
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.G.T.); (B.G.-R.); (I.M.-M.)
| | - Benjamín García-Ramírez
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.G.T.); (B.G.-R.); (I.M.-M.)
| | - Israel Mares-Mejía
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.G.T.); (B.G.-R.); (I.M.-M.)
| | - Enrique Ortega
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad. Universitaria, Coyoacán, Mexico City 04510, Mexico;
| | - Andrea O’Malley
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29209, USA; (A.O.); (M.C.)
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29209, USA; (A.O.); (M.C.)
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Adela Rodríguez-Romero
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.G.T.); (B.G.-R.); (I.M.-M.)
| |
Collapse
|
7
|
Duchesne M, Okoye I, Lacy P. Epithelial cell alarmin cytokines: Frontline mediators of the asthma inflammatory response. Front Immunol 2022; 13:975914. [PMID: 36311787 PMCID: PMC9616080 DOI: 10.3389/fimmu.2022.975914] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
The exposure of the airway epithelium to external stimuli such as allergens, microbes, and air pollution triggers the release of the alarmin cytokines IL-25, IL-33 and thymic stromal lymphopoietin (TSLP). IL-25, IL-33 and TSLP interact with their ligands, IL-17RA, IL1RL1 and TSLPR respectively, expressed by hematopoietic and non-hematopoietic cells including dendritic cells, ILC2 cells, endothelial cells, and fibroblasts. Alarmins play key roles in driving type 2-high, and to a lesser extent type 2-low responses, in asthma. In addition, studies in which each of these three alarmins were targeted in allergen-challenged mice showed decreased chronicity of type-2 driven disease. Consequently, ascertaining the mechanism of activity of these upstream mediators has implications for understanding the outcome of targeted therapies designed to counteract their activity and alleviate downstream type 2-high and low effector responses. Furthermore, identifying the factors which shift the balance between the elicitation of type 2-high, eosinophilic asthma and type-2 low, neutrophilic-positive/negative asthma by alarmins is essential. In support of these efforts, observations from the NAVIGATOR trial imply that targeting TSLP in patients with tezepelumab results in reduced asthma exacerbations, improved lung function and control of the disease. In this review, we will discuss the mechanisms surrounding the secretion of IL-25, IL-33, and TSLP from the airway epithelium and how this influences the allergic airway cascade. We also review in detail how alarmin-receptor/co-receptor interactions modulate downstream allergic inflammation. Current strategies which target alarmins, their efficacy and inflammatory phenotype will be discussed.
Collapse
|
8
|
Odales J, Guzman Valle J, Martínez-Cortés F, Manoutcharian K. Immunogenic properties of immunoglobulin superfamily members within complex biological networks. Cell Immunol 2020; 358:104235. [PMID: 33137645 PMCID: PMC7548077 DOI: 10.1016/j.cellimm.2020.104235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/04/2020] [Accepted: 10/04/2020] [Indexed: 12/01/2022]
Abstract
Antibody-based therapies induce CDR-specific T and B cell responses. Idiotype-anti-idiotype network alters immune system memory compartment. Antigenized antibodies are efficient vaccine immunogen.
Antibodies, T cell receptors and major histocompatibility complex molecules are members of the immunoglobulin superfamily and have pivotal roles in the immune system. The fine interrelation between them regulates several immune functions. Here, we describe lesser-known functions ascribed to these molecules in generating and maintaining immune response. Particularly, we outline the contribution of antibody- and T cell receptor-derived complementarity-determining region neoantigens, antigenized antibodies, as well as major histocompatibility complex class I molecules-derived epitopes to the induction of protective/therapeutic immune responses against pathogens and cancer. We discuss findings of our own and other studies describing protective mechanisms, based on immunogenic properties of immunoglobulin superfamily members, and evaluate the perspectives of application of this class of immunogens in molecular vaccines design.
Collapse
Affiliation(s)
- Josué Odales
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Jesus Guzman Valle
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Fernando Martínez-Cortés
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Karen Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico.
| |
Collapse
|
9
|
Mares-Mejía I, García-Ramírez B, Torres-Larios A, Rodríguez-Hernández A, Osornio-Hernández AI, Terán-Olvera G, Ortega E, Rodríguez-Romero A. Novel murine mAbs define specific and cross-reactive epitopes on the latex profilin panallergen Hev b 8. Mol Immunol 2020; 128:10-21. [PMID: 33045539 DOI: 10.1016/j.molimm.2020.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/10/2020] [Accepted: 09/25/2020] [Indexed: 01/06/2023]
Abstract
The production of specific antibodies able to recognize allergens from different sources or block interactions between allergens and antibodies mediating allergic reactions is crucial for developing successful tools for diagnostics and therapeutics. Panallergens are highly conserved proteins present in widely different species, implicated in relevant cross-reactions. The panallergen latex profilin (Hev b 8) has been associated with the latex-food-pollen syndrome. We generated five monoclonal IgGs and one IgE from murine hybridomas against recombinant Hev b 8 and evaluated their interaction with this allergen using ELISA and biolayer interferometry (BLI). Affinity purified mAbs exhibited high binding affinities towards rHev b 8, with KD1 values ranging from 10-10 M to 10-11 M. Some of these antibodies also recognized the recombinant profilins from maize and tomato (Zea m 12 and Sola l 1), and the ash tree pollen (Fra e 2). Competition ELISA demonstrated that some mAb pairs could bind simultaneously to rHev b 8. Using BLI, we detected competitive, non-competitive, and partial-competition interactions between pairs of mAbs with rHev b 8, suggesting the existence of at least two non-overlapping epitopes on the surface of this allergen. Three-dimensional models of the Fv of 1B4 and 2D10 IgGs and docking simulations of these Fvs with rHev b 8 revealed these epitopes. Furthermore, these two mAbs inhibited the interaction of polyclonal IgE and IgG4 antibodies from profilin-allergic patients with rHev b 8, indicating that the mAbs and the antibodies present in sera from allergic patients bind to overlapping epitopes on the allergen. These mAbs can be useful tools for immune-localization studies, immunoassay development, or standardization of allergenic products.
Collapse
Affiliation(s)
- Israel Mares-Mejía
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Benjamín García-Ramírez
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Alfredo Torres-Larios
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Annia Rodríguez-Hernández
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Ana Isabel Osornio-Hernández
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Gabriela Terán-Olvera
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico
| | - Enrique Ortega
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico.
| | - Adela Rodríguez-Romero
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Cd. Universitaria, Coyoacán, Ciudad de México, 04510, Mexico.
| |
Collapse
|
10
|
Lázaro-Gorines R, López-Rodríguez JC, Benedé S, González M, Mayorga C, Vogel L, Martínez-Del-Pozo Á, Lacadena J, Villalba M. Der p 1-based immunotoxin as potential tool for the treatment of dust mite respiratory allergy. Sci Rep 2020; 10:12255. [PMID: 32703972 PMCID: PMC7378242 DOI: 10.1038/s41598-020-69166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 11/25/2022] Open
Abstract
Immunotoxins appear as promising therapeutic molecules, alternative to allergen-specific-immunotherapy. In this work, we achieved the development of a protein chimera able to promote specific cell death on effector cells involved in the allergic reaction. Der p 1 allergen was chosen as cell-targeting domain and the powerful ribotoxin α-sarcin as the toxic moiety. The resultant construction, named proDerp1αS, was produced and purified from the yeast Pichia pastoris. Der p 1-protease activity and α-sarcin ribonucleolytic action were effectively conserved in proDerp1αS. Immunotoxin impact was assayed by using effector cells sensitized with house dust mite-allergic sera. Cell degranulation and death, triggered by proDerp1αS, was exclusively observed on Der p 1 sera sensitized-humRBL-2H3 cells, but not when treated with non-allergic sera. Most notably, equivalent IgE-binding and degranulation were observed with both proDerp1αS construct and native Der p 1 when using purified basophils from sensitized patients. However, proDerp1αS did not cause any cytotoxic effect on these cells, apparently due to its lack of internalization after their surface IgE-binding, showing the complex in vivo panorama governing allergic reactions. In conclusion, herein we present proDerp1αS as a proof of concept for a potential and alternative new designs of therapeutic tools for allergies. Development of new, and more specific, second-generation of immunotoxins following proDerp1αS, is further discussed.
Collapse
Affiliation(s)
- Rodrigo Lázaro-Gorines
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain
| | - Juan Carlos López-Rodríguez
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain
| | - Sara Benedé
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain
| | - Miguel González
- Allergy Research Laboratory, IBIMA, Hospital Regional Universitario de Málaga, UMA, Málaga, Spain
| | - Cristobalina Mayorga
- Allergy Research Laboratory, IBIMA, Hospital Regional Universitario de Málaga, UMA, Málaga, Spain.,U.G.C. Allergy, IBIMA, Hospital Regional Universitario de Málaga, UMA, Málaga, Spain
| | - Lothar Vogel
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Álvaro Martínez-Del-Pozo
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain
| | - Javier Lacadena
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain.
| | - Mayte Villalba
- Biochemistry and Molecular Biology Department, Chemical Sciences Faculty, Complutense University of Madrid, Av. Complutense w/n, 28040, Madrid, Spain.
| |
Collapse
|
11
|
Harding SD, Faccenda E, Southan C, Pawson AJ, Maffia P, Alexander SPH, Davenport AP, Fabbro D, Levi‐Schaffer F, Spedding M, Davies JA. The IUPHAR Guide to Immunopharmacology: connecting immunology and pharmacology. Immunology 2020; 160:10-23. [PMID: 32020584 PMCID: PMC7160657 DOI: 10.1111/imm.13175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Given the critical role that the immune system plays in a multitude of diseases, having a clear understanding of the pharmacology of the immune system is crucial to new drug discovery and development. Here we describe the International Union of Basic and Clinical Pharmacology (IUPHAR) Guide to Immunopharmacology (GtoImmuPdb), which connects expert-curated pharmacology with key immunological concepts and aims to put pharmacological data into the hands of immunologists. In the pursuit of new therapeutics, pharmacological databases are a vital resource to researchers through providing accurate information on the fundamental science underlying drug action. This extension to the existing IUPHAR/British Pharmacological Society Guide to Pharmacology supports research into the development of drugs targeted at modulating immune, inflammatory or infectious components of disease. To provide a deeper context for how the resource can support research we show data in GtoImmuPdb relating to a case study on the targeting of vascular inflammation.
Collapse
Affiliation(s)
- Simon D. Harding
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Elena Faccenda
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Christopher Southan
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
- Present address:
TW2Informatics LtdGöteborg42166Sweden
| | - Adam J. Pawson
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Pasquale Maffia
- Centre for ImmunobiologyInstitute of Infection, Immunity and InflammationCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Institute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | | | | | - Doriano Fabbro
- Cellestia Biotech SABaselSwitzerland
- TargImmune Therapeutics AGBaselSwitzerland
| | | | | | - Jamie A. Davies
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
12
|
Structural and clinical impact of anti-allergy agents: An overview. Bioorg Chem 2019; 94:103351. [PMID: 31668464 DOI: 10.1016/j.bioorg.2019.103351] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023]
Abstract
Allergic disorders are markedly rising in industrialized countries. The identification of compounds that trigger the immunoglobulin E (IgE)-dependent allergic reaction remain the means to improve the quality of life by limiting patient's exposure to critical allergens. Information concerning the treatment and onset of allergic disorders including atopic dermatitis, allergic rhinitis, and bronchial asthma has been provided by the research over the past decade. Recent studies also indicated that allergic inflammation is associated closely with their exacerbation and progression and indeed is the basic pathophysiology of allergic diseases. As a result of immunological and molecular biological studies our understanding of the mechanism of allergic inflammation with regard to therapeutic agents has improved. While much effort has been paid to developing a new anti-allergic agent, the allergic disease has yet to be completely conquered. The more extensive research will allow the development of new therapeutics to combat allergic diseases. Currently, with respect to mechanism of action anti-allergy drugs are classified into five types including histamine H1 antagonists, leukotriene antagonists, Th2 cytokine inhibitors, thromboxane A2 inhibitors and mediator-release inhibitors. The use of two or more anti-allergy agents together is not acknowledged at present, but this will be the subject of research in the future because with different mechanisms of action anti-allergy agents used at the same time will theoretically increase their effects. This review article focuses on anti-allergy agents highlighting their applications, clinical trials and recent advancement on drugs.
Collapse
|
13
|
Legrand F, Landolina N, Zaffran I, Emeh RO, Chen E, Klion AD, Levi-Schaffer F. Siglec-7 on peripheral blood eosinophils: Surface expression and function. Allergy 2019; 74:1257-1265. [PMID: 30690753 DOI: 10.1111/all.13730] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/03/2018] [Accepted: 12/17/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Siglec-7 is an inhibitory receptor (IR) expressed on human blood eosinophils. Whereas activation of other IRs, including Siglec-8 and CD300a, has been shown to downregulate eosinophil function, little is known about the role of Siglec-7 on human eosinophils. OBJECTIVE To examine Siglec-7 expression and function in eosinophils from normal (ND) and eosinophilic (EO) donors. METHODS Eosinophil expression of Siglec-7 was quantified by flow cytometry and quantitative PCR. Soluble Siglec-7 (sSiglec-7) levels were measured by ELISA in serum. The effect of Siglec-7 on eosinophil viability and degranulation was assessed in vitro by AnnexinV-FITC/7-AAD staining and by measuring GM-CSF-induced mediator release in culture supernatants. Signal transduction was studied by Western blot. RESULTS Siglec-7 was expressed ex vivo on blood eosinophils from all eosinophilic and normal individuals studied. Siglec-7 surface, but not SIGLEC-7mRNA expression, was correlated with absolute eosinophil count (AEC). Siglec-7 was upregulated on purified eosinophils after in vitro stimulation with GM-CSF or IL-5. Serum sSiglec-7 was detectable in 133/144 subjects tested and correlated with AEC. Siglec-7 cross-linking inhibited GM-CSF-induced release of eosinophil peroxidase, TNF-α, and IL-8 (n = 7-8) but did not promote eosinophil apoptosis (n = 5). Finally, Siglec-7 cross-linking on GM-CSF-activated eosinophils induced phosphorylation of SHP-1 and de-phosphorylation of ERK1/2 and p38. CONCLUSIONS Siglec-7 is constitutively expressed on human eosinophils and downmodulates eosinophil activation. Targeting of Siglec-7 on eosinophils might enhance treatment efficacy in eosinophil-driven disorders. Conversely, therapeutic interventions that inhibit Siglec-7 could have unanticipated consequences and promote eosinophilic inflammation.
Collapse
Affiliation(s)
- Fanny Legrand
- Human Eosinophil Section; Laboratory of Parasitic Diseases; National Institute of Allergy and Infectious Diseases; Bethesda Maryland
| | - Nadine Landolina
- Pharmacology and Experimental Therapeutics Unit; School of Pharmacy; Institute for Drug Research; Faculty of Medicine; Hebrew University of Jerusalem; Jerusalem Israel
| | - Ilan Zaffran
- Pharmacology and Experimental Therapeutics Unit; School of Pharmacy; Institute for Drug Research; Faculty of Medicine; Hebrew University of Jerusalem; Jerusalem Israel
| | - Robert O. Emeh
- Human Eosinophil Section; Laboratory of Parasitic Diseases; National Institute of Allergy and Infectious Diseases; Bethesda Maryland
| | - Elizabeth Chen
- Human Eosinophil Section; Laboratory of Parasitic Diseases; National Institute of Allergy and Infectious Diseases; Bethesda Maryland
- University of Maryland; College Park Maryland
| | - Amy D. Klion
- Human Eosinophil Section; Laboratory of Parasitic Diseases; National Institute of Allergy and Infectious Diseases; Bethesda Maryland
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit; School of Pharmacy; Institute for Drug Research; Faculty of Medicine; Hebrew University of Jerusalem; Jerusalem Israel
| |
Collapse
|
14
|
Eichenfield LF, Bieber T, Beck LA, Simpson EL, Thaçi D, de Bruin-Weller M, Deleuran M, Silverberg JI, Ferrandiz C, Fölster-Holst R, Chen Z, Graham NMH, Pirozzi G, Akinlade B, Yancopoulos GD, Ardeleanu M. Infections in Dupilumab Clinical Trials in Atopic Dermatitis: A Comprehensive Pooled Analysis. Am J Clin Dermatol 2019; 20:443-456. [PMID: 31066001 PMCID: PMC6533236 DOI: 10.1007/s40257-019-00445-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Patients with moderate-to-severe atopic dermatitis (AD) have increased infection risk, including skin infections and systemic infections. Immunomodulators (e.g., anti-tumor necrosis factors, anti-interleukin [anti-IL]-23, anti-IL-17, Janus kinase inhibitors) increase risk of infections. Dupilumab (a monoclonal antibody blocking the shared receptor component for IL-4 and IL-13) is approved for inadequately controlled moderate-to-severe AD and for moderate-to-severe eosinophilic or oral corticosteroid-dependent asthma. OBJECTIVE The aim was to determine the impact of dupilumab on infection rates in patients with moderate-to-severe AD. METHODS This analysis pooled data from seven randomized, placebo-controlled dupilumab trials in adults with moderate-to-severe AD. Exposure-adjusted analyses assessed infection rates. RESULTS Of 2932 patients, 1091 received placebo, 1095 dupilumab 300 mg weekly, and 746 dupilumab 300 mg every 2 weeks. Treatment groups had similar infection rates overall per 100 patient-years (placebo, 155; dupilumab weekly, 150; dupilumab every 2 weeks, 156; dupilumab combined, 152), and similar non-skin infection rates. Serious/severe infections were reduced with dupilumab (risk ratio 0.43; p < 0.05), as were bacterial and other non-herpetic skin infections (risk ratio 0.44; p < 0.001). Although herpesviral infection rates overall were slightly higher with dupilumab than placebo, clinically important herpesviral infections (eczema herpeticum, herpes zoster) were less common with dupilumab (risk ratio 0.31; p < 0.01). Systemic anti-infective medication use was lower with dupilumab. CONCLUSIONS Dupilumab is associated with reduced risk of serious/severe infections and non-herpetic skin infections and does not increase overall infection rates versus placebo in patients with moderate-to-severe AD. CLINICALTRIALS. GOV IDENTIFIERS NCT01548404, NCT02210780, NCT01859988, NCT02277743, NCT02277769, NCT02260986, and NCT02755649.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal, Humanized
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Dermatitis, Atopic/complications
- Dermatitis, Atopic/diagnosis
- Dermatitis, Atopic/drug therapy
- Double-Blind Method
- Humans
- Incidence
- Injections, Subcutaneous
- Placebos/administration & dosage
- Placebos/adverse effects
- Randomized Controlled Trials as Topic
- Severity of Illness Index
- Skin Diseases, Infectious/epidemiology
- Skin Diseases, Infectious/etiology
- Skin Diseases, Infectious/prevention & control
- Treatment Outcome
Collapse
Affiliation(s)
- Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California, San Diego, San Diego, CA, 92123, USA.
- Division of Pediatric and Adolescent Dermatology, Rady Children's Hospital, 3020 Children's Way, Mail Code 5092, San Diego, CA, 92123, USA.
| | - Thomas Bieber
- Department of Dermatology and Allergy, Christine Kühne-Center for Allergy Research and Education, University of Bonn, Bonn, Germany
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| | - Diamant Thaçi
- Comprehensive Center for Inflammation Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Mette Deleuran
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Carlos Ferrandiz
- Servicio de Dermatología, Hospital Universitario Germans Trias i Pujol, Badalona Universidad Autónoma de Barcelona, Barcelona, Spain
| | | | - Zhen Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | |
Collapse
|
15
|
Sharman JL, Harding SD, Southan C, Faccenda E, Pawson AJ, Davies JA. Accessing Expert-Curated Pharmacological Data in the IUPHAR/BPS Guide to PHARMACOLOGY. ACTA ACUST UNITED AC 2019; 61:1.34.1-1.34.46. [PMID: 30040201 DOI: 10.1002/cpbi.46] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The IUPHAR/BPS Guide to PHARMACOLOGY is an expert-curated, open-access database of information on drug targets and the substances that act on them. This unit describes the procedures for searching and downloading ligand-target binding data and for finding detailed annotations and the most relevant literature. The database includes concise overviews of the properties of 1,700 data-supported human drug targets and related proteins, divided into families, and 9,000 small molecule and peptide experimental ligands and approved drugs that bind to those targets. More detailed descriptions of pharmacology, function, and pathophysiology are provided for a subset of important targets. The information is reviewed regularly by expert subcommittees of the IUPHAR Committee on Receptor Nomenclature and Drug Classification. A new immunopharmacology portal has recently been added, drawing together data on immunological targets, ligands, cell types, processes and diseases. The data are available for download and can be accessed computationally via Web services. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Joanna L Sharman
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon D Harding
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher Southan
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Faccenda
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Adam J Pawson
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | -
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Hoggard M, Waldvogel-Thurlow S, Zoing M, Chang K, Radcliff FJ, Wagner Mackenzie B, Biswas K, Douglas RG, Taylor MW. Inflammatory Endotypes and Microbial Associations in Chronic Rhinosinusitis. Front Immunol 2018; 9:2065. [PMID: 30283438 PMCID: PMC6157407 DOI: 10.3389/fimmu.2018.02065] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/21/2018] [Indexed: 11/13/2022] Open
Abstract
A complex mix of inflammatory and microbial associations underscores the chronic inflammatory condition chronic rhinosinusitis (CRS), and the etiology remains poorly understood. Recent work has begun to delineate between variants (endotypes) of CRS on the basis of inflammatory biomarkers. This study aimed to assess inflammatory patterns in CRS phenotypes, identify putative endotypes of CRS, and to assess inflammatory associations with the sinonasal microbiota. Ten cytokines and six inflammatory cell types were assessed in mucosal biopsies from 93 CRS subjects and 17 controls via cytometric bead array and immunohistochemical techniques. Putative endotypes were identified via cluster analysis of subjects on the basis of inflammatory markers and comorbidities including polyposis, asthma, and aspirin sensitivity. Finally, previously published bacterial data for this cohort were reanalyzed to evaluate associations with inflammatory markers and CRS subtypes. Inflammatory patterns were highly variable within standard CRS phenotypes. Cluster analysis identified eight subject clusters, with strong delineation on the basis of polyposis and asthma, but also subtle distinctions in inflammatory markers. An association was also identified between depletion of several "health-associated" bacterial taxa, reduced bacterial diversity and increased overall bacterial load, with markers of inflammation and clinical severity. This study contributes to ongoing efforts to define distinct endotypes of CRS on the basis of underlying inflammatory processes, and also offers compelling evidence of a link between bacterial community dysbiosis and inflammation in CRS. Further resolving the heterogeneity of CRS is vital to inform clinical management and personalized treatment approaches.
Collapse
Affiliation(s)
- Michael Hoggard
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Melissa Zoing
- School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Kevin Chang
- Department of Statistics, Statistical Consulting Centre, The University of Auckland, Auckland, New Zealand
| | - Fiona J Radcliff
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Kristi Biswas
- School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Michael W Taylor
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
17
|
The effect of omalizumab treatment on severe allergic asthma and allergic comorbidities: real-life experience from the Czech Anti-IgE Registry. Postepy Dermatol Alergol 2018; 35:510-515. [PMID: 30429711 PMCID: PMC6232552 DOI: 10.5114/ada.2018.77243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/11/2018] [Indexed: 01/23/2023] Open
Abstract
Introduction Omalizumab is indicated for the treatment of severe allergic asthma (SAA) and chronic spontaneous urticaria, although a number of studies have confirmed the effectiveness of this therapy also for other IgE-mediated diseases. Aim To assess the impact of anti-IgE therapy on SAA and comorbid IgE-mediated allergic diseases in patients treated with omalizumab for SAA enrolled in the CAR (Czech Anti-IgE Registry). Material and methods Three hundred and ten patients with SAA treated with omalizumab were enrolled in the CAR. Two hundred and twenty-nine individuals were evaluated after 12 months of omalizumab treatment for asthma control test (ACT), examination of fractional exhaled nitric oxide (FENO), forced expiratory volume in 1 s (FEV1), the use of systemic corticosteroids, side effects of treatment and clinical effect of omalizumab on allergic comorbidities (allergic rhinitis, chronic urticaria, atopic dermatitis and food allergy). Results After 12 months of treatment with omalizumab, patients experienced a significant improvement of ACT and FEV1, reduction of FENO, use of systemic corticosteroids for asthma exacerbations and dose of maintenance oral corticosteroid therapy. The positive effect of treatment with omalizumab was observed in 82.2% of patients with allergic rhinitis, in 85.7% of patients with chronic urticaria, in 82.1% of patients with atopic dermatitis, and in 67.3% of patients with food allergy. Conclusions In the CAR registry, patients with SAA treated with omalizumab showed a significant positive effect of anti-IgE therapy not only on the asthma control, but also on allergic comorbidities.
Collapse
|
18
|
Manoutcharian K, Perez-Garmendia R, Gevorkian G. Recombinant Antibody Fragments for Neurodegenerative Diseases. Curr Neuropharmacol 2018; 15:779-788. [PMID: 27697033 PMCID: PMC5771054 DOI: 10.2174/1570159x01666160930121647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/04/2016] [Accepted: 09/28/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recombinant antibody fragments are promising alternatives to full-length immunoglobulins and offer important advantages compared with conventional monoclonal antibodies: extreme specificity, higher affinity, superior stability and solubility, reduced immunogenicity as well as easy and inexpensive large-scale production. OBJECTIVE In this article we will review and discuss recombinant antibodies that are being evaluated for neurodegenerative diseases in pre-clinical models and in clinical studies and will summarize new strategies that are being developed to optimize their stability, specificity and potency for advancing their use. METHODS Articles describing recombinant antibody fragments used for neurological diseases were selected (PubMed) and evaluated for their significance. RESULTS Different antibody formats such as single-chain fragment variable (scFv), single-domain antibody fragments (VHHs or sdAbs), bispecific antibodies (bsAbs), intrabodies and nanobodies, are currently being studied in pre-clinical models of cancer as well as infectious and autoimmune diseases and many of them are being tested as therapeutics in clinical trials. Immunotherapy approaches have shown therapeutic efficacy in several animal models of Alzheimer´s disease (AD), Parkinson disease (PD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), Huntington disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). It has been demonstrated that recombinant antibody fragments may neutralize toxic extra- and intracellular misfolded proteins involved in the pathogenesis of AD, PD, DLB, FTD, HD or TSEs and may target toxic immune cells participating in the pathogenesis of MS. CONCLUSION Recombinant antibody fragments represent a promising tool for the development of antibody-based immunotherapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Roxanna Perez-Garmendia
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP 04510, Mexico. 0
| |
Collapse
|
19
|
Scott‐Taylor TH, Axinia S, Amin S, Pettengell R. Immunoglobulin G; structure and functional implications of different subclass modifications in initiation and resolution of allergy. Immun Inflamm Dis 2018; 6:13-33. [PMID: 29164823 PMCID: PMC5818455 DOI: 10.1002/iid3.192] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/14/2017] [Accepted: 07/23/2017] [Indexed: 12/24/2022] Open
Abstract
IgE and not IgG is usually associated with allergy. IgE lodged on mast cells in skin or gut and basophils in the blood allows for the prolonged duration of allergy through the persistent expression of high affinity IgE receptors. However, many allergic reactions are not dependent on IgE and are generated in the absence of allergen specific and even total IgE. Instead, IgG plasma cells are involved in induction of, and for much of the pathogenesis of, allergic diseases. The pattern of IgG producing plasma cells in atopic children and the tendency for direct or further class switching to IgE are the principle factors responsible for long-lasting sensitization of mast cells in allergic children. Indirect class switching from IgG producing plasma cells has been shown to be the predominant pathway for production of IgE while a Th2 microenvironment, genetic predisposition, and the concentration and nature of allergens together act on IgG plasma cells in the atopic tendency to undergo further immunoglobulin gene recombination. The seminal involvement of IgG in allergy is further indicated by the principal role of IgG4 in the natural resolution of allergy and as the favourable immunological response to immunotherapy. This paper will look at allergy through the role of different antibodies than IgE and give current knowledge of the nature and role of IgG antibodies in the start, maintenance and resolution of allergy.
Collapse
Affiliation(s)
| | - Stefan‐Claudiu Axinia
- School of Life SciencesLondon Metropolitan University166‐220 Holloway RoadLondon, N7 8DB
| | - Sumeya Amin
- School of Life SciencesLondon Metropolitan University166‐220 Holloway RoadLondon, N7 8DB
| | - Ruth Pettengell
- Department of HaematologySt George's University of LondonCranmer TerraceLondon SW17 0RE
| |
Collapse
|
20
|
Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S, Gray AJG, Bruce L, Alexander SPH, Anderton S, Bryant C, Davenport AP, Doerig C, Fabbro D, Levi-Schaffer F, Spedding M, Davies JA. The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res 2018; 46:D1091-D1106. [PMID: 29149325 PMCID: PMC5753190 DOI: 10.1093/nar/gkx1121] [Citation(s) in RCA: 1501] [Impact Index Per Article: 214.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023] Open
Abstract
The IUPHAR/BPS Guide to PHARMACOLOGY (GtoPdb, www.guidetopharmacology.org) and its precursor IUPHAR-DB, have captured expert-curated interactions between targets and ligands from selected papers in pharmacology and drug discovery since 2003. This resource continues to be developed in conjunction with the International Union of Basic and Clinical Pharmacology (IUPHAR) and the British Pharmacological Society (BPS). As previously described, our unique model of content selection and quality control is based on 96 target-class subcommittees comprising 512 scientists collaborating with in-house curators. This update describes content expansion, new features and interoperability improvements introduced in the 10 releases since August 2015. Our relationship matrix now describes ∼9000 ligands, ∼15 000 binding constants, ∼6000 papers and ∼1700 human proteins. As an important addition, we also introduce our newly funded project for the Guide to IMMUNOPHARMACOLOGY (GtoImmuPdb, www.guidetoimmunopharmacology.org). This has been 'forked' from the well-established GtoPdb data model and expanded into new types of data related to the immune system and inflammatory processes. This includes new ligands, targets, pathways, cell types and diseases for which we are recruiting new IUPHAR expert committees. Designed as an immunopharmacological gateway, it also has an emphasis on potential therapeutic interventions.
Collapse
Affiliation(s)
- Simon D Harding
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Joanna L Sharman
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Elena Faccenda
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Chris Southan
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Adam J Pawson
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sam Ireland
- Department of Structural & Molecular Biology, University College London, London WC1E 6BT, UK
| | - Alasdair J G Gray
- School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Liam Bruce
- School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, UK
| | - Stephen Anderton
- MRC Centre for inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Clare Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Christian Doerig
- Department of Microbiology, Monash University, Clayton 3800, Australia
| | | | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | | | - Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- To whom correspondence should be addressed. Tel: +44 131 650 2999;
| | | |
Collapse
|
21
|
|
22
|
Chronic Rhinosinusitis and the Evolving Understanding of Microbial Ecology in Chronic Inflammatory Mucosal Disease. Clin Microbiol Rev 2017; 30:321-348. [PMID: 27903594 DOI: 10.1128/cmr.00060-16] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chronic rhinosinusitis (CRS) encompasses a heterogeneous group of debilitating chronic inflammatory sinonasal diseases. Despite considerable research, the etiology of CRS remains poorly understood, and debate on potential roles of microbial communities is unresolved. Modern culture-independent (molecular) techniques have vastly improved our understanding of the microbiology of the human body. Recent studies that better capture the full complexity of the microbial communities associated with CRS reintroduce the possible importance of the microbiota either as a direct driver of disease or as being potentially involved in its exacerbation. This review presents a comprehensive discussion of the current understanding of bacterial, fungal, and viral associations with CRS, with a specific focus on the transition to the new perspective offered in recent years by modern technology in microbiological research. Clinical implications of this new perspective, including the role of antimicrobials, are discussed in depth. While principally framed within the context of CRS, this discussion also provides an analogue for reframing our understanding of many similarly complex and poorly understood chronic inflammatory diseases for which roles of microbes have been suggested but specific mechanisms of disease remain unclear. Finally, further technological advancements on the horizon, and current pressing questions for CRS microbiological research, are considered.
Collapse
|
23
|
Bell DC, Brown SJ. Atopic eczema treatment now and in the future: Targeting the skin barrier and key immune mechanisms in human skin. World J Dermatol 2017; 6:42-51. [DOI: 10.5314/wjd.v6.i3.42] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/14/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023] Open
Abstract
The skin facilitates a number of key roles but its functioning can be impaired by disease. Atopic eczema is a chronic inflammatory disease where the skin barrier has become leaky, and inflammation occurs. It affects up to 20% of children and 3% of adults worldwide, manifesting as red itchy patches of skin with varying severity. This review aims to investigate the leaky skin barrier and immune mechanisms from the perspective of potential novel treatments. The complexity of atopic eczema as a disease is what makes it difficult to treat. Genome-wide association studies have highlighted possible genetic variations associated with atopic eczema, however in some cases, individuals develop the disease without these genetic risk factors. Loss of function mutations in the filaggrin gene are one of these associations and this is plausible due to its key role in barrier function. The Th2 immune response is the link with regards to the immune mechanisms as atopic inflammation often occurs through increased levels of interleukin (IL)-4 and IL-13. Eczematous inflammation also creates susceptibility to colonisation and damage by bacteria such as Staphylococcus aureus. Potential novel treatments are becoming ever more specific, offering the hope of fewer side effects and better disease control. The best new treatments highlighted in this review target the immune response with human beta defensin 2, phosphodiesterase-4 inhibitors and monoclonal antibodies all showing promise.
Collapse
|
24
|
Lemaire M, Oppliger A, Hotz P, Renauld JC, Braun J, Maggi M, Barresi F, Schmid-Grendelmeier P, Huaux F, Dressel H. Can serum cytokine profile discriminate irritant-induced and allergen-induced symptoms? A cross-sectional study in workers mostly exposed to laboratory animals. Occup Environ Med 2017; 74:592-600. [PMID: 28416643 DOI: 10.1136/oemed-2016-104137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/02/2017] [Accepted: 03/19/2017] [Indexed: 11/03/2022]
Abstract
BACKGROUND In workers exposed mostly to laboratory animals (LA), symptoms may be due to irritants or allergens. Correct aetiological diagnosis is important for health surveillance. OBJECTIVES This study aims to test whether work-related (WR) allergen-induced symptoms are associated with a cytokine profile distinct from that due to irritants. METHODS In a cross-sectional study (n=114), WR respiratory and/or skin symptoms were assessed through a standardised clinical examination and sensitisation to rat and/or mouse allergen determined by serum immunoglobulin E. Serum cytokine concentrations were measured by multiplex assays. The predefined cytokine profiles 'sensitiser' (interleukin (IL)-4, IL-5, IL-13, eotaxin-1) and 'irritation' (IL-8, IL-17A, IL-17F, IL-22) were considered positive, when ≥3 concentrations exceeded the 95th percentile of the asymptomatic non-sensitised group. Results were examined by hierarchical clustering analyses (HCA) and multiple linear regression. Explorative analyses were carried out for nine additional cytokines. Exposure to allergens and endotoxin was assessed in a subpopulation. RESULTS The prevalence of the profile 'irritation' was comparable in 28 symptomatic non-sensitised workers and 71 asymptomatic non-sensitised workers. HCA showed that nearly all symptomatic non-sensitised workers were gathered in two subclusters, characterised by high IL-17A levels, but different IL-8 levels. Multiple linear regression identified drug consumption and current complaints as confounders. Sensitised subjects were too few (n=14) for testing the profile 'sensitiser'. CONCLUSIONS In this unselected population of LA workers, the profile 'irritation' did not prove to be a valuable health surveillance tool. Low power precluded assessment of the profile 'sensitiser'. The increased IL-17A concentration may originate from irritative constituents of organic dust.
Collapse
Affiliation(s)
- Muriel Lemaire
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
| | - Anne Oppliger
- Institute for Work and Health, University of Lausanne, Lausanne, Switzerland
| | - Philipp Hotz
- Division of Occupational and Environmental Medicine, EBPI, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jean-Christophe Renauld
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Ludwig Institute for Cancer Research, Brussels Branch, Brussels, Belgium
| | - Julia Braun
- Department of Biostatistics, EBPI, University of Zurich, Zurich, Switzerland.,Department of Epidemiology, EBPI, University of Zurich, Zurich, Switzerland
| | - Marion Maggi
- Division of Occupational and Environmental Medicine, EBPI, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Fabio Barresi
- Division of Occupational and Environmental Medicine, EBPI, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Holger Dressel
- Division of Occupational and Environmental Medicine, EBPI, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Hong JY, Bae JH, Lee KE, Kim M, Kim MH, Kang HJ, Park EH, Yoo KS, Jeong SK, Kim KW, Kim KE, Sohn MH. Antibody to FcεRIα Suppresses Immunoglobulin E Binding to High-Affinity Receptor I in Allergic Inflammation. Yonsei Med J 2016; 57:1412-9. [PMID: 27593869 PMCID: PMC5011273 DOI: 10.3349/ymj.2016.57.6.1412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/21/2016] [Accepted: 03/24/2016] [Indexed: 11/27/2022] Open
Abstract
PURPOSE High-affinity receptor I (FcεRI) on mast cells and basophils plays a key role in the immunoglobulin E (IgE)-mediated type I hypersensitivity mediated by allergen cross-linking of the specific IgE-FcεRI complex. Thus, prevention of IgE binding to FcεRI on these cells is an effective therapy for allergic disease. We have developed a strategy to disrupt IgE binding to FcεRI using an antibody targeting FcεRIα. MATERIALS AND METHODS Fab fragment antibodies, which lack the Fc domain, with high affinity and specificity for FcεRIα and effective inhibitory activity against IgE-FcεRI binding were screened. IgE-induced histamine, β-hexosaminidase and Ca²⁺ release in basophils were determined by ELISA. A B6.Cg-Fcer1a(tm1Knt) Tg(FCER1A)1Bhk/J mouse model of passive cutaneous anaphylaxis (PCA) was used to examine the inhibitory effect of NPB311 on allergic skin inflammation. RESULTS NPB311 exhibited high affinity to human FcεRIα (KD=4 nM) and inhibited histamine, β-hexosaminidase and Ca²⁺ release in a concentration-dependent manner in hFcεRI-expressing cells. In hFcεRIα-expressing mice, dye leakage was higher in the PCA group than in controls, but decreased after NPB311 treatment. NPB311 could form a complex with FcεRIα and inhibit the release of inflammation mediators. CONCLUSION Our approach for producing anti-FcεRIα Fab fragment antibody NPB311 may enable clinical application to a therapeutic pathway in IgE/FcεRI-mediated diseases.
Collapse
Affiliation(s)
- Jung Yeon Hong
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | - Kyung Eun Lee
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Mina Kim
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Min Hee Kim
- CRID Center, NeoPharm Co., Ltd., Daejeon, Korea
| | | | | | | | | | - Kyung Won Kim
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Kyu Earn Kim
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Hyun Sohn
- Department of Pediatrics and Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
26
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
27
|
Landolina N, Levi-Schaffer F. Monoclonal antibodies: the new magic bullets for allergy: IUPHAR Review 17. Br J Pharmacol 2016; 173:793-803. [PMID: 26620589 DOI: 10.1111/bph.13396] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/08/2015] [Accepted: 11/22/2015] [Indexed: 12/18/2022] Open
Abstract
Allergic diseases and conditions are widespread and their incidence is on the increase. They are characterized by the activation of mast cells resident in tissues and the consequent infiltration and stimulation of several inflammatory cells, predominantly eosinophils. Cell-cell cross-talk and the release of mediators are responsible for the symptoms and for the modulation of the response. The gold standard of therapeutic intervention is still glucocorticosteroids, although they are not effective in all patients and may cause numerous side effects. Symptomatic medications are also widespread. As research has led to deeper insights into the mechanisms governing the diseases, new avenues have been opened resulting in recent years in the development of monoclonal antibodies (mAbs) such as anti-IgE mAbs (omalizumab) and others still undergoing clinical trials aimed to specifically target molecules involved in the migration and stimulation of inflammatory cells. In this review, we summarize new developments in the field of anti-allergic mAbs with special emphasis on the treatment of asthma, particularly severe forms of this condition, and atopic dermatitis, which are two unmet clinical needs.
Collapse
Affiliation(s)
- N Landolina
- Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - F Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|