1
|
Layhadi JA, Lalioti A, Palmer E, van Zelm MC, Wambre E, Shamji MH. Mechanisms and Predictive Biomarkers of Allergen Immunotherapy in the Clinic. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:59-66. [PMID: 37996041 DOI: 10.1016/j.jaip.2023.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Allergen immunotherapy (AIT) remains to be the only disease-modifying treatment for IgE-mediated allergic diseases such as allergic rhinitis. It can provide long-term clinical benefits when given for 3 years or longer. Mechanisms of immune tolerance induction by AIT are underscored by the modulation of several compartments within the immune system. These include repair of disruption in epithelial barrier integrity, modulation of the innate immune compartment that includes regulatory dendritic cells and innate lymphoid cells, and adaptive immune compartments such as induction of regulatory T and B cells. Altogether, these are also associated with the dampening of allergen-specific TH2 and T follicular helper cell responses and subsequent generation of blocking antibodies. Although AIT is effective in modifying the immune response, there is a lack of validated and clinically relevant biomarkers that can be used to monitor desensitization, efficacy, and the likelihood of response, all of which can contribute to accelerating personalized medication and increasing patient care. Candidate biomarkers comprise humoral, cellular, metabolic, and in vivo biomarkers; however, these are primarily studied in small trials and require further validation. In this review, we evaluate the current candidates of biomarkers of AIT and how we can implement changes in future studies to help us identify clinically relevant biomarkers of safety, compliance, and efficacy.
Collapse
Affiliation(s)
- Janice A Layhadi
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anastasia Lalioti
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Elizabeth Palmer
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Menno C van Zelm
- Department of Immunology, Monash University and Alfred Health, Melbourne, Victoria, Australia; Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Erik Wambre
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mohamed H Shamji
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
2
|
Pitsios C. Allergen Immunotherapy: Biomarkers and Clinical Outcome Measures. J Asthma Allergy 2021; 14:141-148. [PMID: 33633455 PMCID: PMC7901403 DOI: 10.2147/jaa.s267522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Clinical trials for allergen immunotherapy products’ development and approval are conducted, aiming to monitor safety and efficacy of them. Symptom scores and the use of rescue medication are the primary clinical endpoints used in the conducted clinical trials, while Quality of Life scores and symptom-free days are measurements also used as secondary endpoints. Although the use of in vitro biomarkers might have been more practical and objective, there are yet no broadly used reliable ones accurately reflecting the clinical effects of allergen immunotherapy. On the contrary, in vivo biomarkers, such as the nasal allergy provocation test, are reliable and successfully used. The aim of this review is to describe how to adapt and use biomarkers and clinical outcomes in the everyday practice of Allergists who perform allergen immunotherapy.
Collapse
Affiliation(s)
- Constantinos Pitsios
- Allergy Outpatient Clinic, Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
3
|
Sun W, Pan L, Yu Q, Sun Y, Zeng X, Bai X, Li M. The skin prick test response after allergen immunotherapy in different levels of tIgE children with mite sensitive Asthma/Rhinitis in South China. Hum Vaccin Immunother 2018; 14:2510-2515. [PMID: 30047819 DOI: 10.1080/21645515.2018.1482171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND At present, the biomarkers which can predict the clinical efficacy of allergen immunotherapy (AIT) are still much debated. IgE levels are often related to allergic severity. Therefore, this study aimed at relating total IgE (tIgE) levels with the efficacy of AIT assessed by symptoms and drug score and skin prick test (SPT) response. METHODS We evaluated 81 allergic children who had received house-dust mite (HDM) subcutaneous immunotherapy for three years. According to the tIgE levels before treatment, all children were divided into high value, medium value and low value group. Each group according to sIgE/tIgE ratio was divided into subgroups. The efficacy of AIT is assessed by symptoms and drug score. By comparing changes in the grade of SPT in each group, the response of AIT are evaluated. RESULTS The SPT grade changes to determine efficacy had a high degree of consistency with symptoms and drug score judgment (sensitivity 89.7%, specificity 78.3%, Kappa = 0.670, P < 0.001). Compared to ineffective cases, the effective cases had lower tIgE (P < 0.001) and higher ratio of sIgE/tIgE (P < 0.001). The grades of SPT declined the most in the low value group (low value group vs. medium value group, P < 0.05; low value group vs. high value group, P < 0.001; medium value group vs. high value group, P < 0.05). CONCLUSIONS The SPT grade change can be used for efficacy evaluation. Children with lower level of tIgE and higher ratio of sIgE/tIgE that obtain a more satisfactory effect.
Collapse
Affiliation(s)
- Wangming Sun
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Lingzhi Pan
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Qiying Yu
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Yan Sun
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Xiangyan Zeng
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Xiaoli Bai
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| | - Mengrong Li
- a Center for Asthma and Allergy Immunotherapy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , P.R. China
| |
Collapse
|
4
|
Augé J, Vent J, Agache I, Airaksinen L, Campo Mozo P, Chaker A, Cingi C, Durham S, Fokkens W, Gevaert P, Giotakis A, Hellings P, Herknerova M, Hox V, Klimek L, La Melia C, Mullol J, Muluk NB, Muraro A, Naito K, Pfaar O, Riechelmann H, Rondon C, Rudenko M, Samolinski B, Tasca I, Tomazic P, Vogt K, Wagenmann M, Yeryomenko G, Zhang L, Mösges R. EAACI Position paper on the standardization of nasal allergen challenges. Allergy 2018; 73:1597-1608. [PMID: 29377177 DOI: 10.1111/all.13416] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
Nasal allergen challenge (NAC) is an important tool to diagnose allergic rhinitis. In daily clinical routine, experimentally, or when measuring therapeutic success clinically, nasal allergen challenge is fundamental. It is further one of the key diagnostic tools when initiating specific allergen immunotherapy. So far, national recommendations offered guidance on its execution; however, international divergence left many questions unanswered. These differences in the literature caused EAACI to initiate a task force to answer unmet needs and find a consensus in executing nasal allergen challenge. On the basis of a systematic review containing nasal allergen challenges of the past years, task force members reviewed evidence, discussed open issues, and studied variations of several subjective and objective assessment parameters to propose a standardized way of a nasal allergen challenge procedure in clinical practice. Besides an update on indications, contraindications, and preparations for the test procedure, main recommendations are a bilaterally challenge with standardized allergens, with a spray device offering 0.1 mL per nostril. A systematic catalogue for positivity criteria is given for the variety of established subjective and objective assessment methods as well as a schedule for the challenge procedure. The task force recommends a unified protocol for NAC for daily clinical practice, aiming at eliminating the previous difficulty of comparing NAC results due to unmet needs.
Collapse
Affiliation(s)
- J. Augé
- Department of Otorhinolaryngology, Head and Neck Surgery; University of Cologne Medical Center; Cologne Germany
- Institute of Medical Statistics and Computational Biology; Faculty of Medicine; University of Cologne; Cologne Germany
| | - J. Vent
- Department of Otorhinolaryngology, Head and Neck Surgery; University of Cologne Medical Center; Cologne Germany
- Institute of Medical Statistics and Computational Biology; Faculty of Medicine; University of Cologne; Cologne Germany
- Department of Otorhinolaryngology, Head and Neck Surgery; Universitätsmedizin Mannheim, Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
| | - I. Agache
- Transylvania University Brasov; Faculty of Medicine; Department of Allergy and Clinical Immunology; Brasov Romania
| | - L. Airaksinen
- Health and Work Ability; Finnish Institute of Occupational Health; Helsinki Finland
| | - P. Campo Mozo
- Allergy Unit; IBIMA-Regional University Hospital of Málaga, ARADyAL; Málaga Spain
| | - A. Chaker
- Department of Otolaryngology; Center of Allergy and Environment (ZAUM); Klinikum rechts der Isar; Technical University Munich; Munich Germany
| | - C. Cingi
- ENT Department; Faculty of Medicine; Eskisehir Osmangazi University; Eskisehir Turkey
| | - S. Durham
- Allergy and Clinical Immunology; Imperial College; NHLI; London UK
| | - W. Fokkens
- Otorhinolaryngology; Academic Medical Centre; Amsterdam The Netherlands
| | - P. Gevaert
- Otorhinolaryngology; Ghent University; Ghent Belgium
| | - A. Giotakis
- Department of Otorhinolaryngology - Head and Neck Surgery; Medical University of Innsbruck; Medizinische Universitat Innsbruck; Innsbruck Austria
| | - P. Hellings
- Department of Otorhinolaryngology; Cliniques Universitaires Saint-Luc; Brussels Belgium
| | - M. Herknerova
- Alergologie a klinická imunologie; Nemocnice na Homolce; Prague Czech Republic
| | - V. Hox
- Department of Otorhinolaryngology; Cliniques Universitaires Saint-Luc; Brussels Belgium
| | - L. Klimek
- Center for Rhinology and Allergology; Wiesbaden Germany
| | - C. La Melia
- Department of ENT; Azienda Ausl di Imola; Imola Italy
| | - J. Mullol
- Clinical and Experimental Immunoallergy; Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS); Barcelona Spain
- Department of ORL; Hospital Clínic de Barcelona; Universitat de Barcelona; Barcelona Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES); Barcelona Spain
| | - N. B. Muluk
- Department of Otorhinolaryngology; Medical Faculty; Kirikkale University; Kirikkale Turkey
| | - A. Muraro
- Department of Pediatrics; Referral Centre for Food Allergy; Padua General University Hospital; Padua Italy
| | - K. Naito
- Fujita Health University, Otolaryngology; 1-98 Denngakugakubo, Kutukake-cho; Toyoake city Aichi Prefecture Japan
| | - O. Pfaar
- Center for Rhinology and Allergology; Wiesbaden Germany
- Department of Otorhinolaryngology, Head and Neck Surgery; Universitätsmedizin Mannheim, Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
| | - H. Riechelmann
- Department of Otorhinolaryngology - Head and Neck Surgery; Medical University of Innsbruck; Medizinische Universitat Innsbruck; Innsbruck Austria
| | - C. Rondon
- Allergy Unit; IBIMA-Regional University Hospital of Málaga, ARADyAL; Málaga Spain
| | - M. Rudenko
- London Allergy and Immunology Centre; London UK
| | - B. Samolinski
- Department of Prevention of Envinronmental Hazards and Allergology; Medical University of Warsaw; Poland
| | - I. Tasca
- Department of ENT; Azienda Ausl di Imola; Imola Italy
| | - P. Tomazic
- Department of General Otorhinolaryngology, Head and Neck Surgery; Medical University of Graz; Graz Austria
| | - K. Vogt
- Faculty of Medicine; University of Latvia; Riga Latvia
| | - M. Wagenmann
- Department of Otorhinolaryngology; Düsseldorf University Hospital (UKD); Düsseldorf Germany
| | - G. Yeryomenko
- Kharkiv National Medical University; Kharkiv Ukraine
| | - L. Zhang
- Department of Otolaryngology Head and Neck Surgery; Beijing TongRen Hospital; Capital Medical University; Beijing China
| | - R. Mösges
- Institute of Medical Statistics and Computational Biology; Faculty of Medicine; University of Cologne; Cologne Germany
| |
Collapse
|
5
|
Gunawardana NC, Durham SR. New approaches to allergen immunotherapy. Ann Allergy Asthma Immunol 2018; 121:293-305. [PMID: 30025907 DOI: 10.1016/j.anai.2018.07.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE New insights into mechanisms should enable strategic improvement of allergen immunotherapy, aiming to make it safer, faster, more effective, and able to induce long-term tolerance. We review novel approaches with potential to translate into clinical use. DATA SOURCES Database searches were conducted in PubMed, Scopus, and Google Scholar. STUDY SELECTIONS Search terms were based on current and novel approaches in immunotherapy. Literature was selected primarily from recent randomized double-blinded placebo-controlled trials and meta-analyses. RESULTS Alum, microcrystalline tyrosine, and calcium phosphate are adjuvants in current use. Toll-like receptor-4 agonists combined with allergen have potential to shorten duration of treatment. Other novel adjuvants, nanoparticles, and virus-like particles in combination with allergen have shown early promise. Omalizumab lessens systemic side effects but does not improve efficacy. Intralymphatic immunotherapy for aeroallergens, epicutaneous immunotherapy for food allergens, and use of modified allergens (allergoids), recombinant allergens (and hypoallergenic variants), and T- and B-cell peptide approaches have shown evidence of efficacy and permitted shortened courses but have only rarely been compared with conventional extracts. CONCLUSION Novel routes of immunotherapy, use of modified allergens, and combination of allergens with immunostimulatory adjuvants or immune modifiers have been developed to augment downregulation of T-helper cell type 2 immunity and/or induce "protective" blocking antibodies. Although these strategies have permitted shortened courses, confirmatory phase 3 trials are required to confirm efficacy and safety and head-to-head trials are required for comparative efficacy. Currently, subcutaneous and sublingual immunotherapies using in-house standardized crude extracts remain the only approaches proved to induce long-term tolerance.
Collapse
Affiliation(s)
- Natasha C Gunawardana
- Imperial College London, London, United Kingdom; Royal Brompton and Harefield Hospitals, NHS Foundation Trust, London, United Kingdom
| | - Stephen R Durham
- Imperial College London, London, United Kingdom; Royal Brompton and Harefield Hospitals, NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
6
|
Nelson HS. Current and future challenges of subcutaneous and sublingual allergy immunotherapy for allergists in the United States. Ann Allergy Asthma Immunol 2018; 121:278-280. [PMID: 29409924 DOI: 10.1016/j.anai.2017.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Harold S Nelson
- National Jewish Health, University of Colorado Denver School of Medicine, Denver, Colorado.
| |
Collapse
|
7
|
van de Veen W, Wirz OF, Globinska A, Akdis M. Novel mechanisms in immune tolerance to allergens during natural allergen exposure and allergen-specific immunotherapy. Curr Opin Immunol 2017; 48:74-81. [DOI: 10.1016/j.coi.2017.08.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/14/2017] [Accepted: 08/18/2017] [Indexed: 01/22/2023]
|
8
|
Klimek L, Pfaar O, Bousquet J, Senti G, Kündig T. Allergen immunotherapy in allergic rhinitis: current use and future trends. Expert Rev Clin Immunol 2017; 13:897-906. [DOI: 10.1080/1744666x.2017.1333423] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ludger Klimek
- Department of Otorhinolaryngology, Allergy Center, Wiesbaden, Germany
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Allergy Center, Wiesbaden, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jean Bousquet
- MACVIA-France, Contre les MAladies Chroniques pour un VIeillissement Actif en France European Innovation Partnership on Active and Healthy Ageing Reference Site, Montpellier, France
- INSERM U 1168, VIMA: Ageing and chronic diseases Epidemiological and public health approaches, Villejuif, Université Versailles St-Quentin-en-Yvelines, Montigny le Bretonneux, France
| | - Gabriela Senti
- Center for Clinical Trials, Zurich University Hospital, Zurich, Switzerland
| | - Thomas Kündig
- Dept. of Dermatology, Zurich University Hospital, Zurich, Switzerland
| |
Collapse
|
9
|
Schussler E, Cox LS. Efficacy of House Dust Mite Sublingual Immunotherapy in Adults with Allergic Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2017; 5:870-872. [PMID: 36444006 DOI: 10.1016/j.jaip.2016.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/05/2016] [Accepted: 11/09/2016] [Indexed: 06/16/2023]
Affiliation(s)
| | - Linda S Cox
- University of Miami Miller School of Medicine at Holy Cross Hospital, Miami, Fla
| |
Collapse
|
10
|
Calderon MA, Demoly P, Casale T, Akdis CA, Bachert C, Bewick M, Bilò BM, Bohle B, Bonini S, Bush A, Caimmi DP, Canonica GW, Cardona V, Chiriac AM, Cox L, Custovic A, De Blay F, Devillier P, Didier A, Di Lorenzo G, Du Toit G, Durham SR, Eng P, Fiocchi A, Fox AT, van Wijk RG, Gomez RM, Haathela T, Halken S, Hellings PW, Jacobsen L, Just J, Tanno LK, Kleine-Tebbe J, Klimek L, Knol EF, Kuna P, Larenas-Linnemann DE, Linneberg A, Matricardi M, Malling HJ, Moesges R, Mullol J, Muraro A, Papadopoulos N, Passalacqua G, Pastorello E, Pfaar O, Price D, Del Rio PR, Ruëff R, Samolinski B, Scadding GK, Senti G, Shamji MH, Sheikh A, Sisul JC, Sole D, Sturm GJ, Tabar A, Van Ree R, Ventura MT, Vidal C, Varga EM, Worm M, Zuberbier T, Bousquet J. Allergy immunotherapy across the life cycle to promote active and healthy ageing: from research to policies: An AIRWAYS Integrated Care Pathways (ICPs) programme item (Action Plan B3 of the European Innovation Partnership on active and healthy ageing) and the Global Alliance against Chronic Respiratory Diseases (GARD), a World Health Organization GARD research demonstration project. Clin Transl Allergy 2016; 6:41. [PMID: 27895895 PMCID: PMC5120439 DOI: 10.1186/s13601-016-0131-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 11/02/2016] [Indexed: 12/17/2022] Open
Abstract
Allergic diseases often occur early in life and persist throughout life. This life-course perspective should be considered in allergen immunotherapy. In particular it is essential to understand whether this al treatment may be used in old age adults. The current paper was developed by a working group of AIRWAYS integrated care pathways for airways diseases, the model of chronic respiratory diseases of the European Innovation Partnership on active and healthy ageing (DG CONNECT and DG Santé). It considered (1) the political background, (2) the rationale for allergen immunotherapy across the life cycle, (3) the unmet needs for the treatment, in particular in preschool children and old age adults, (4) the strategic framework and the practical approach to synergize current initiatives in allergen immunotherapy, its mechanisms and the concept of active and healthy ageing.
Collapse
Affiliation(s)
- M A Calderon
- National Heart and Lung Institute, Royal Brompton Hospital NHS, Imperial College London, London, UK
| | - P Demoly
- Unité d'allergologie, Département de Pneumologie et AddictologieHôpital Arnaud de Villeneuve, CHRU de Montpellier, Sorbonne Universités, UPMC Paris 06, UMR-S 1136, IPLESP, Equipe EPAR, 75013 Paris, France
| | - T Casale
- University of South Florida Morsani College of Medicine, Tampa, FL USA
| | - C A Akdis
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Swiss Institute of Allergy and Asthma Research (SIAF)University of Zurich, Davos, Switzerland
| | - C Bachert
- Upper Airways Research Laboratory (URL), ENT Department, University Hospital Ghent, Ghent, Belgium
| | - M Bewick
- iQ4U consultants Ltd, London, UK
| | - B M Bilò
- Allergy Unit, Department of Internal Medicine, University Hosp Ospedali Riuniti, Ancona, Italy
| | - B Bohle
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - S Bonini
- Second University of Naples and IFT-CNR, Rome, Italy
| | - A Bush
- National Heart and Lung Institute, Royal Brompton Hospital NHS, Imperial College London, London, UK
| | - D P Caimmi
- Unité d'allergologie, Département de Pneumologie et AddictologieHôpital Arnaud de Villeneuve, CHRU de Montpellier, Sorbonne Universités, UPMC Paris 06, UMR-S 1136, IPLESP, Equipe EPAR, 75013 Paris, France
| | - G W Canonica
- Allergy and Respiratory Diseases Clinic, DIMI, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - V Cardona
- Allergy Section, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - A M Chiriac
- Division of Allergy, Hôpital Arnaud de Villeneuve, Department of Pulmonology, University Hospital of Montpellier, Montpellier - UPMC Univ Paris 06, UMRS 1136, Equipe - EPAR - IPLESP, Sorbonne Universités, Paris, France
| | - L Cox
- Nova Southeastern University, Ft. Lauderdale, FL USA
| | - A Custovic
- National Heart and Lung Institute, Royal Brompton Hospital NHS, Imperial College London, London, UK
| | - F De Blay
- Allergy Division, Chest Disease Department, University Hospital of Strasbourg, Strasbourg, France
| | - P Devillier
- University Versailles Saint-Quentin and Clinical Pharmacology Unit, UPRES EA 220, Department of Airway Diseases, Foch Hospital, Suresnes, France
| | - A Didier
- Respiratory Diseases Department, Rangueil-Larrey Hospital, Toulouse, France
| | - G Di Lorenzo
- Dipartimento BioMedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - G Du Toit
- Guy's and St. Thomas' NHS Trust, Kings College, London, UK
| | - S R Durham
- Allergy and Clinical Immunology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - P Eng
- Department of Pediatric Pulmonology and Allergy, Children's Hospital, Aarau, Switzerland
| | - A Fiocchi
- Division of Allergy, Department of Pediatrics, Bambino Gesù Pediatric Hospital, Vatican City, Rome, Italy
| | - A T Fox
- King's College London Allergy Academy, London, UK
| | - R Gerth van Wijk
- Section of Allergology, Department of Internal Medicine, Erasmus Medical Center, Building Rochussenstraat, Rotterdam, The Netherlands
| | - R M Gomez
- Unidad Alergia and Asma, Hospital San Bernardo, Salta, Argentina
| | - T Haathela
- Skin and Allergy Hospital, Helsinki University Hospital, Helsinki, Finland
| | - S Halken
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - P W Hellings
- Clinical Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, KU Leuven, Louvain, Belgium
| | - L Jacobsen
- Allergy Learning and Consulting, Secretary Immunotherapy Interest Group EAACI, Copenhagen, Denmark
| | - J Just
- Allergology Department, Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand-Trousseau, INSERM, UMR_S 1136, Sorbonne Universités, UPMC Univ Paris, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Equipe EPAR, Paris, France
| | - L K Tanno
- Hospital Sírio Libanês, São Paulo, Brazil ; University Hospital of Montpellier, Montpellier, France ; UPMC Paris 06, UMR-S 1136, IPLESP, Equipe EPAR, Sorbonne Universités, Paris, France
| | - J Kleine-Tebbe
- Allergy and Asthma Center Westend, Outpatient Clinic and Clinical Research Center, Ackermann, Hanf, & Kleine-Tebbe, Berlin, Germany
| | - L Klimek
- Center for Rhinology and Allergology, German Society for Otorhinolaryngology HNS, Wiesbaden, Germany
| | - E F Knol
- Departments of Immunology and Dermatology/Allergology, University Medical Center Utrecht, Heidelberglaan, Utrecht, The Netherlands
| | - P Kuna
- Medical University of Lodz, Lodz, Poland
| | | | - A Linneberg
- Research Centre for Prevention and Health, The Capital Region of Denmark, Copenhagen, Denmark ; Department of Clinical Experimental Research, Rigshospitalet, Copenhagen, Denmark ; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Matricardi
- Pediatric Pneumology and Immunology, Charité Medical University, Berlin, Germany
| | - H J Malling
- Danish Allergy Centre, Allergy Clinic, Gentofte University Hospital, Hellerup, Denmark
| | - R Moesges
- IMSIE, Klinikum der Universität zu Köln A. ö. R., Cologne, Germany
| | - J Mullol
- Unitat de Rinologia i Clínica de l'Olfacte, ENT Department, Hospital Clínic, Clinical and Experimental Respiratory Immunoallergy, IDIBAPS, CIBERES, Barcelona, Catalonia Spain
| | - A Muraro
- Department of Women and Child Health, Food Allergy Referral Centre Veneto Region, Padua General University Hospital, Padua, Italy
| | - N Papadopoulos
- Allergy Unit, 2nd Pediatric Clinic, University of Athens, Athens, Greece
| | - G Passalacqua
- Allergy and Respiratory Diseases, IRCCS San Martino-IST, Univesity of Genoa, Genoa, Italy
| | - E Pastorello
- ASST Grande Ospedale Metropolitano Niguarda, P.zza Ospedale Maggiore, Milan, Italy
| | - O Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Universitätsmedizin Mannheim, Mannheim, Germany ; Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany ; Center for Rhinology and Allergology, Wiesbaden, Germany
| | - D Price
- Division of Applied Health Sciences, Primary Care Respiratory Medicine, Academic Primary Care, University of Aberdeen, Aberdeen, Scotland, UK ; Research in Real Life (RiRL), Oakington, Cambridge, UK ; Optimum Patient Care Ltd, Singapore, Singapore
| | | | - R Ruëff
- Department of Dermatology and Allergology, Ludwig-Maximillian University, Munich, Germany
| | - B Samolinski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - G K Scadding
- Royal National Throat, Nose and Ear Hospital, London, UK ; University College London, London, UK
| | - G Senti
- Clinical Trials Center, University Hospital of Zurich, Zurich, Switzerland
| | - M H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK ; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - A Sheikh
- Asthma UK Centre for Applied Research, Centre for Medical Informatics, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG UK
| | | | - D Sole
- Programa de Pòs-Graduação em Pediatria e Ciências Aplicadas à Pediatria, Departamento de Pediatria EPM, UNIFESP, São Paulo, Brazil
| | - G J Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria ; Allergy Outpatient Clinic Reumannplatz, Vienna, Austria
| | - A Tabar
- Servicio de Alergologia, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - R Van Ree
- Departments of Experimental Immunology and Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - M T Ventura
- Unit of Geriatric Immunoallergology, Interdisciplinary Department of Medicine, University of Bari Medical School, Bari, Italy
| | - C Vidal
- Allergy Department, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - E M Varga
- Respiratory and Allergic Disease Division, Department of Paediatrics, Medical University of Graz, Graz, Austria
| | - M Worm
- Allergie-Centrum-Charité, Klinik für Dermatologie, Venerologie und Allergologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - T Zuberbier
- Allergie-Centrum-Charité, Klinik für Dermatologie, Venerologie und Allergologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - J Bousquet
- University Hospital of Montpellier, Montpellier, France ; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon, European Innovation Partnership on Active and Healthy Ageing Reference Site, Paris, France ; INSERM, VIMA, U1168, Ageing and Chronic Diseases, Epidemiological and Public Health Approaches, Paris, France ; UVSQ, UMR-S 1168, Université Versailles St-Quentin-en-Yvelines, Versailles Cedex, France ; CHRU, 371 Avenue du Doyen Gaston Giraud, 34295 Montpellier Cedex 5, France
| |
Collapse
|
11
|
Nelson HS. Allergen immunotherapy (AIT) for the multiple-pollen sensitive patient. Expert Rev Clin Pharmacol 2016; 9:1443-1451. [PMID: 27687128 DOI: 10.1080/17512433.2016.1237874] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The majority of allergic subjects are polysensitized. In Europe, allergy immunotherapy (AIT) in these patients is usually limited to their single clinically most troublesome allergy while in the U.S. the immunotherapy prescription usually includes all allergen extracts to which the patient has evidence of clinical sensitivity. Areas covered: This article will review the evidence supporting the U.S. practice. It will also review the major new development in the management of polysensitized patients, the introduction of component-resolved diagnosis (CRD). Expert commentary: This allows, in many cases, distinguishing in polysensitized patients between sensitization to the major allergens of several unrelated allergen extracts and to panallergens that cause broad patterns of cross-reactivity.
Collapse
Affiliation(s)
- Harold S Nelson
- a Department of Medicine , National Jewish Health , Denver , CO , USA
| |
Collapse
|
12
|
Pfaar O, Twuijver E, Boot JD, Opstelten DJE, Klimek L, Ree R, Diamant Z, Kuna P, Panzner P. A randomized DBPC trial to determine the optimal effective and safe dose of a SLIT-birch pollen extract for the treatment of allergic rhinitis: results of a phase II study. Allergy 2016; 71:99-107. [PMID: 26417901 PMCID: PMC5057302 DOI: 10.1111/all.12760] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) is a potential efficacious and safe treatment option for patients with respiratory, IgE-mediated allergic diseases. A combined tolerability, dose-finding study with a sublingual liquid birch pollen preparation (SB) was conducted. METHODS Two hundred and sixty-nine adults with birch-pollen-induced AR were randomized to placebo, SB: 3333, 10,000, 20,000 or 40,000 AUN/ml. Differences in symptom scores following a titrated nasal provocation test (TNPT) at baseline and after 5 months of treatment were determined. Safety, tolerability, birch-pollen-specific immunoglobulin levels and peak nasal inspiratory flow (PNIF) were also measured (all measures determined outside the birch pollen season). RESULTS In all treatment groups, an improvement in symptom scores after treatment compared to baseline was observed, with an additional stepwise improvement in the active groups compared to placebo, which was significant in high-dose groups (P = 0.008 and P < 0.001, respectively). For this primary endpoint, a significant linear dose-response curve was observed: the higher the dose, the better the improvement observed. Likewise, active treatment resulted in an increase in PNIF and serum IgG levels compared to placebo. The highest improvements were found in the 40,000 AUN/ml group. All active dosages resulted in more adverse reactions than placebo, which were mainly mild and well-controlled. CONCLUSIONS A multicentre trial evaluated the dose-response and tolerability of SB. All active treatment groups showed better responses than placebo for both primary and secondary parameters. The results indicate that, within the studied dose range, SB 40,000 AUN/ml is the most optimal effective and safe dose (ClinicalTrials.gov: NCT01639768).
Collapse
Affiliation(s)
- O. Pfaar
- Center for Rhinology and Allergology Wiesbaden Germany
- Department of Otorhinolaryngology, Head and Neck Surgery Medical Faculty Mannheim Universitätsmedizin Mannheim Heidelberg University Mannheim Germany
| | | | | | | | - L. Klimek
- Center for Rhinology and Allergology Wiesbaden Germany
| | - R. Ree
- Departments of Experimental Immunology and of Otorhinolaryngology Academic Medical Center University of Amsterdam Amsterdam The Netherlands
| | - Z. Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund Sweden
- Departments of Clinical Pharmacy & Pharmacology and General Practice University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - P. Kuna
- Dept. of Internal Medicine Asthma and Allergy Medical University of Lodz Lodz Poland
| | - P. Panzner
- Department of Allergology and Immunology Medical Faculty in Plzen Charles University Prague Prague Czech Republic
| |
Collapse
|
13
|
Prickett SR, Rolland JM, O'Hehir RE. Immunoregulatory T cell epitope peptides: the new frontier in allergy therapy. Clin Exp Allergy 2015; 45:1015-26. [PMID: 25900315 PMCID: PMC4654246 DOI: 10.1111/cea.12554] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2015] [Indexed: 12/12/2022]
Abstract
Allergen immunotherapy (AIT) has been practised since 1911 and remains the only therapy proven to modify the natural history of allergic diseases. Although efficacious in carefully selected individuals, the currently licensed whole allergen extracts retain the risk of IgE-mediated adverse events, including anaphylaxis and occasionally death. This together with the need for prolonged treatment regimens results in poor patient adherence. The central role of the T cell in orchestrating the immune response to allergen informs the choice of T cell targeted therapies for down-regulation of aberrant allergic responses. Carefully mapped short synthetic peptides that contain the dominant T cell epitopes of major allergens and bind to a diverse array of HLA class II alleles, can be delivered intradermally into non-inflamed skin to induce sustained clinical and immunological tolerance. The short peptides from allergenic proteins are unable to cross-link IgE and possess minimal inflammatory potential. Systematic progress has been made from in vitro human models of allergen T cell epitope-based peptide anergy in the early 1990s, through proof-of-concept murine allergy models and early human trials with longer peptides, to the current randomized, double-blind, placebo-controlled clinical trials with the potential new class of synthetic short immune-regulatory T cell epitope peptide therapies. Sustained efficacy with few adverse events is being reported for cat, house dust mite and grass pollen allergy after only a short course of treatment. Underlying immunological mechanisms remain to be fully delineated but anergy, deletion, immune deviation and Treg induction all seem contributory to successful outcomes, with changes in IgG4 apparently less important compared to conventional AIT. T cell epitope peptide therapy is promising a safe and effective new class of specific treatment for allergy, enabling wider application even for more severe allergic diseases.
Collapse
Affiliation(s)
- S R Prickett
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Vic., Australia
- Department of Immunology, Monash University, Melbourne, Vic., Australia
| | - J M Rolland
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Vic., Australia
- Department of Immunology, Monash University, Melbourne, Vic., Australia
| | - R E O'Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Vic., Australia
- Department of Immunology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
14
|
Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ J 2015; 8:17. [PMID: 26023323 PMCID: PMC4430874 DOI: 10.1186/s40413-015-0063-2] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 02/06/2015] [Indexed: 12/25/2022] Open
Abstract
Substantial progress in understanding mechanisms of immune regulation in allergy, asthma, autoimmune diseases, tumors, organ transplantation and chronic infections has led to a variety of targeted therapeutic approaches. Allergen-specific immunotherapy (AIT) has been used for 100 years as a desensitizing therapy for allergic diseases and represents the potentially curative and specific way of treatment. The mechanisms by which allergen-AIT has its mechanisms of action include the very early desensitization effects, modulation of T- and B-cell responses and related antibody isotypes as well as inhibition of migration of eosinophils, basophils and mast cells to tissues and release of their mediators. Regulatory T cells (Treg) have been identified as key regulators of immunological processes in peripheral tolerance to allergens. Skewing of allergen-specific effector T cells to a regulatory phenotype appears as a key event in the development of healthy immune response to allergens and successful outcome in AIT. Naturally occurring FoxP3+ CD4+CD25+ Treg cells and inducible type 1 Treg (Tr1) cells contribute to the control of allergen-specific immune responses in several major ways, which can be summarized as suppression of dendritic cells that support the generation of effector T cells; suppression of effector Th1, Th2 and Th17 cells; suppression of allergen-specific IgE, and induction of IgG4; suppression of mast cells, basophils and eosinophils and suppression of effector T cell migration to tissues. New strategies for immune intervention will likely include targeting of the molecular mechanisms of allergen tolerance and reciprocal regulation of effector and regulatory T cell subsets.
Collapse
Affiliation(s)
- Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Obere Strasse 22, CH7270 Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Obere Strasse 22, CH7270 Davos, Switzerland
| |
Collapse
|
15
|
Klimek L, Sperl A, van Twuijver E, van Ree R, Kleinjans H, Boot JD, Pfaar O. A prospective study comparing the efficacy and safety of two sublingual birch allergen preparations. Clin Transl Allergy 2014; 4:23. [PMID: 25097754 PMCID: PMC4122029 DOI: 10.1186/2045-7022-4-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/09/2014] [Indexed: 11/10/2022] Open
Abstract
Background SUBLIVAC FIX Birch (SUB-B) is a liquid oral preparation of Betula verrucosa pollen extract for the treatment of allergic rhinitis/rhinoconjuctivitis induced by birch pollen. The major allergen content of SUB-B and Staloral Birch (Stal-B) have been shown to be comparable. In order to compare the clinical efficacy and safety of both products, the present study was designed to investigate efficacy of treatment with SUB-B compared to Stal-B by means of reduction in allergy symptoms assessed by a titrated nasal provocation test (TNPT) in subjects suffering from IgE mediated allergy complaints triggered by birch pollen. Methods A prospective, randomized, open, blinded endpoint (PROBE), controlled, single-centre study in 74 birch allergic adults was performed. Treatment consisted of either SUB-B (10,000 AUN/ml) or Stal-B (initial phase 10 I.R./ml and maintenance phase 300 I.R./ml) for 16–20 weeks at maintenance dose. The primary efficacy outcome was defined by the difference in change of the TNPT-threshold dose between the two treatment groups at baseline and after completion of treatment. Secondary outcomes included determination of birch pollen specific IgE and IgG levels, safety lab and ECG. During the first 30 days of treatment, subjects were requested to fill out a diary concerning compliance with study medication, occurrence of AEs and the use of concomitant medication. Results Analysis of the primary efficacy parameter showed that the percentage of subjects showing a beneficial treatment effect was similar in both treatment groups, 33.3% for SUB-B vs. 31.4% for Stal-B in the intention to treat population. Evaluation of the immunologic response, showed that treatment with SUB-B and Stal-B induced similar increases (approximately 2 times) in IgE, IgG and IgG4 specific for Bet v 1. In total, 143 related adverse events (AEs) were reported. The majority of the AEs was of mild intensity. The same pattern of AEs was observed for both products. No clinically relevant changes in other safety parameters, such as safety laboratory parameters, vital signs, physical examination and ECGs were observed. Conclusion Taken together, treatment with both products was effective by means of reduction in allergic symptoms during a TNPT. In addition, safety analysis revealed a good tolerability of both SLIT extracts.
Collapse
Affiliation(s)
- Ludger Klimek
- Center for Rhinology and Allergology Wiesbaden, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Wiesbaden, Germany
| | - Annette Sperl
- Center for Rhinology and Allergology Wiesbaden, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Wiesbaden, Germany
| | | | - Ronald van Ree
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Huub Kleinjans
- HAL Allergy BV, Medical Department, Leiden, The Netherlands
| | | | - Oliver Pfaar
- Center for Rhinology and Allergology Wiesbaden, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Wiesbaden, Germany
| |
Collapse
|
16
|
Pfaar O, Demoly P, Gerth van Wijk R, Bonini S, Bousquet J, Canonica GW, Durham SR, Jacobsen L, Malling HJ, Mösges R, Papadopoulos NG, Rak S, Rodriguez del Rio P, Valovirta E, Wahn U, Calderon MA. Recommendations for the standardization of clinical outcomes used in allergen immunotherapy trials for allergic rhinoconjunctivitis: an EAACI Position Paper. Allergy 2014; 69:854-67. [PMID: 24761804 DOI: 10.1111/all.12383] [Citation(s) in RCA: 311] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2014] [Indexed: 01/17/2023]
Abstract
BACKGROUND Allergen immunotherapy (AIT) has been thoroughly documented in randomized controlled trials (RCTs). It is the only immune-modifying and causal treatment available for patients suffering from IgE-mediated diseases such as allergic rhinoconjunctivitis, allergic asthma and insect sting allergy. However, there is a high degree of clinical and methodological heterogeneity among the endpoints in clinical studies on AIT, for both subcutaneous and sublingual immunotherapy (SCIT and SLIT). At present, there are no commonly accepted standards for defining the optimal outcome parameters to be used for both primary and secondary endpoints. METHODS As elaborated by a Task Force (TF) of the European Academy of Allergy and Clinical Immunology (EAACI) Immunotherapy Interest Group, this Position Paper evaluates the currently used outcome parameters in different RCTs and also aims to provide recommendations for the optimal endpoints in future AIT trials for allergic rhinoconjunctivitis. RESULTS Based on a thorough literature review, the TF members have outlined recommendations for nine domains of clinical outcome measures. As the primary outcome, the TF recommends a homogeneous combined symptom and medication score (CSMS) as a simple and standardized method that balances both symptoms and the need for antiallergic medication in an equally weighted manner. All outcomes, grouped into nine domains, are reviewed. CONCLUSION A standardized and globally harmonized method for analysing the clinical efficacy of AIT products in RCTs is required. The EAACI TF highlights the CSMS as the primary endpoint for future RCTs in AIT for allergic rhinoconjunctivitis.
Collapse
Affiliation(s)
- O. Pfaar
- Center for Rhinology and Allergology Wiesbaden; Department of Otorhinolaryngology, Head and Neck Surgery; University Hospital Mannheim; Mannheim Germany
| | - P. Demoly
- Département de Pneumologie et Addictologie; Hôpital Arnaud de Villeneuve; University Hospital of Montpellier; Montpellier France
- Sorbonne Universités; UPMC Paris 06, UMR-S 1136, IPLESP; Equipe EPAR; Paris France
| | - R. Gerth van Wijk
- Section of Allergology; Department of Internal Medicine; Erasmus MC; Rotterdam the Netherlands
| | - S. Bonini
- Second University of Naples and Institute of Translational Pharmacology; Italian National Research Council (IFT-CNR); Rome Italy
| | - J. Bousquet
- Département de Pneumologie et Addictologie; Hôpital Arnaud de Villeneuve; University Hospital of Montpellier; Montpellier France
- 1018, Respiratory and Environmental Epidemiology Team; INSERM; CESP Centre for research in Epidemiology and Population Health; Villejuif France
| | - G. W. Canonica
- Respiratory Diseases & Allergy Clinic; University of Genova; IRCCS AOU San Martino; Genova Italy
| | - S. R. Durham
- Section of Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College; London UK
| | - L. Jacobsen
- ALC, Allergy Learning and Consulting; Copenhagen Denmark
| | - H. J. Malling
- Allergy Clinic; University Hospital Gentofte; Copenhagen Denmark
| | - R. Mösges
- Institute of Medical Statistics; Informatics and Epidemiology (IMSIE); University of Cologne; Cologne Germany
| | - N. G. Papadopoulos
- Allergy Department; 2nd Pediatric Clinic; University of Athens; Athens Greece
- Centre for Paediatrics and Child Health; Institute of Human Development; University of Manchester; Manchester UK
| | - S. Rak
- Department of Respiratory Medicine and Allergology; Sahlgrenska University Hospital; Goteborg Sweden
| | | | - E. Valovirta
- Department of Clinical Allergology and Pulmonary Diseases; University of Turku; Finland
- Suomen Terveystalo Allergy Clinic; Turku Finland
| | - U. Wahn
- Department for Pediatric Pneumology and Immunology; Charité Medical University; Berlin Germany
| | - M. A. Calderon
- Section of Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College; London UK
| | | |
Collapse
|
17
|
Mechanisms of allergen-specific immunotherapy: multiple suppressor factors at work in immune tolerance to allergens. J Allergy Clin Immunol 2014; 133:621-31. [PMID: 24581429 DOI: 10.1016/j.jaci.2013.12.1088] [Citation(s) in RCA: 391] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 12/18/2013] [Accepted: 12/24/2013] [Indexed: 01/08/2023]
Abstract
Allergen-specific immunotherapy (AIT) has been used for more than 100 years as a desensitizing therapy for IgE-mediated allergic diseases and represents a potentially curative way of treatment. The mechanisms of action of AIT include the induction of very early desensitization of mast cells and basophils; generation of regulatory T and regulatory B (Breg) cell responses; regulation of IgE and IgG4; decreases in numbers and activity of eosinophils and mast cells in mucosal allergic tissues; and decreases in the activity of basophils in circulation. Skewing of allergen-specific effector T and effector B cells to a regulatory phenotype appears as a key event in the course of AIT and normal immune response to allergens. Recently, inducible IL-10-secreting Breg cells were also demonstrated to contribute to allergen tolerance through suppression of effector T cells and selective induction of IgG4 isotype antibodies. Allergen-specific regulatory T and Breg cells orchestrate a general immunoregulatory activity, which can be summarized as suppression of cytokines from inflammatory dendritic cells; suppression of effector TH1, TH2, and TH17 cells; suppression of allergen-specific IgE and induction of IgG4; and suppression of migration of mast cells, basophils, eosinophils, and effector T cells to tissues. A detailed knowledge of the mechanisms of AIT is not only important in designing the prevention and treatment of allergic diseases but might also find applications in the treatment of autoimmune diseases, organ transplantation, chronic infection, and cancer.
Collapse
|
18
|
Burks A, Calderon M, Casale T, Cox L, Demoly P, Jutel M, Nelson H, Akdis C. Mise à jour sur l’immunothérapie allergénique : Rapport de Consensus PRACTALL de l’AAAAI (American Academy of Allergy, Asthma and Immunology) et de l’EAACI (European Academy of Allergy and Clinical Immunology). REVUE FRANCAISE D ALLERGOLOGIE 2014. [DOI: 10.1016/j.reval.2013.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
19
|
Ventura I, Vega A, Chamorro C, Aroca R, Gómez E, Pineda F, Palacios R, Blanca M, Monteseirín J. Allergen immunotherapy decreases LPS-induced NF-κB activation in neutrophils from allergic patients. Pediatr Allergy Immunol 2014; 25:129-35. [PMID: 24118194 DOI: 10.1111/pai.12145] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (IT) is widely used to treat allergic diseases. The molecular mechanisms have not been clarified yet completely. The present work was undertaken to analyze the effect of IT in the activation of NF-κB. METHODS Neutrophils from 15 pollen-allergic IT-treated patients, 10 untreated pollen-allergic patients, and 10 healthy donors were in vitro stimulated with LPS. NF-κB activation (p65/p52) was measured in their nuclear extracts by enzyme-linked immunosorbent assay (ELISA). IκBα phosphorylation, NF-κB-repressing factor (NRF) activation, and thromboxane A2 (TXA2 ) and Interleukin-8 (IL-8) release were measured by ELISA. RESULTS There was a positive correlation between the score of symptoms and NF-κB activation in human neutrophils. IT significantly decreased NF-κB activation levels in neutrophils compared with neutrophils from untreated patients. IκBα phosphorylation and NRF activation levels were, respectively, significantly lower and higher in neutrophils from IT-treated patients than from untreated patients. IL-8 and TXA2 release were significantly lower in neutrophils from IT-treated patients than from untreated patients. CONCLUSIONS IT positive effects are at least in part mediated by the negative regulation of NF-κB activation in human neutrophils. These observations represent a novel view of neutrophils as possible cell target to treat IgE-dependent diseases through NF-κB downmodulation.
Collapse
Affiliation(s)
- Inmaculada Ventura
- Servicio de Inmunología y Alergia, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bokanovic D, Aberer W, Hemmer W, Heinemann A, Komericki P, Scheffel J, Sturm GJ. Determination of sIgE to rPhl p 1 is sufficient to diagnose grass pollen allergy. Allergy 2013; 68:1403-9. [PMID: 24117513 DOI: 10.1111/all.12263] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND New diagnostic tools such as the basophil activation test (BAT) and component-resolved diagnosis (CRD) are promising for Hymenoptera venom or food allergy. A clear benefit for inhalant allergens has not yet been shown. Our aim was to compare new and established tests for grass pollen allergy. METHODS Forty-nine patients with grass pollen allergy and 47 controls were prospectively enrolled in the study. A symptom score was calculated for each patient. Conjunctival provocation tests (CPT), skin prick tests (SPT), BAT, and sIgE determination including CRD were performed. Sensitivity and specificity were compared and results were correlated with the symptom score. RESULTS Single determination of sIgE to rPhl p 1 showed the best balance between sensitivity (98%) and specificity (92%). Use of additional components, such as rPhl p 2 and 5, did not increase sensitivity. Generally, sensitivity of tests was high: SPT 100%, ISAC-112 100%, sIgE to timothy grass 98%, BAT 98%, ISAC-103 84%, and CPT 83%. Specificity ranged from 79% (SPT) to 96% (CPT). All test results and calculated values (e.g. ratio sIgE/tIgE) did not correlate with symptom severity. Asymptomatic sensitization to timothy grass in controls was rare in the CAP (11%) and predominantly due to Phl p 1 sensitization. CONCLUSION rPhl p 1 was sufficient to diagnose grass pollen allergy, and sIgE patterns were the same in symptomatically and asymptomatically sensitized subjects. The testing of multiple components was of minor importance, and no test correlated with symptom severity.
Collapse
Affiliation(s)
- D. Bokanovic
- Department of Dermatology and Venereology; Medical University of Graz; Graz Austria
| | - W. Aberer
- Department of Dermatology and Venereology; Medical University of Graz; Graz Austria
| | - W. Hemmer
- Floridsdorf Allergy Center; Vienna Austria
| | - A. Heinemann
- Institute of Experimental and Clinical Pharmacology; Medical University of Graz; Austria
| | - P. Komericki
- Department of Dermatology and Venereology; Medical University of Graz; Graz Austria
| | - J. Scheffel
- Laboratory of Molecular Immunogenetics; National Institute of Arthritis and Musculoskeletal and Skin Diseases; National Institutes of Health; Bethesda MD USA
| | - G. J. Sturm
- Department of Dermatology and Venereology; Medical University of Graz; Graz Austria
| |
Collapse
|
21
|
Burks AW, Calderon MA, Casale T, Cox L, Demoly P, Jutel M, Nelson H, Akdis CA. Update on allergy immunotherapy: American Academy of Allergy, Asthma & Immunology/European Academy of Allergy and Clinical Immunology/PRACTALL consensus report. J Allergy Clin Immunol 2013; 131:1288-96.e3. [PMID: 23498595 DOI: 10.1016/j.jaci.2013.01.049] [Citation(s) in RCA: 327] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/24/2013] [Accepted: 01/28/2013] [Indexed: 12/16/2022]
Abstract
Allergy immunotherapy (AIT) is an effective treatment for allergic asthma and rhinitis, as well as venom-induced anaphylaxis. In addition to reducing symptoms, AIT can change the course of allergic disease and induce allergen-specific immune tolerance. In current clinical practice immunotherapy is delivered either subcutaneously or sublingually; some allergens, such as grass pollen, can be delivered through either route, whereas others, such as venoms, are only delivered subcutaneously. Both subcutaneous and sublingual immunotherapy appear to have a duration of efficacy of up to 12 years, and both can prevent the development of asthma and new allergen sensitivities. In spite of the advances with AIT, safer and more effective AIT strategies are needed, especially for patients with asthma, atopic dermatitis, or food allergy. Novel approaches to improve AIT include use of adjuvants or recombinant allergens and alternate routes of administration. As part of the PRACTALL initiatives, the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma & Immunology nominated an expert team to develop a comprehensive consensus report on the mechanisms of AIT and its use in clinical practice, as well as unmet needs and ongoing developments in AIT. This resulting report is endorsed by both academies.
Collapse
Affiliation(s)
- A Wesley Burks
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599-7220, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Bousquet J, Yssel H, Demoly P. Prospects for a vaccine in allergic diseases and asthma. BioDrugs 2012; 13:61-75. [PMID: 18034513 DOI: 10.2165/00063030-200013010-00006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Allergen-specific immunotherapy is widely used to treat allergic diseases, and current research is now focusing on the development of therapeutic vaccines acting on the IgE immune response following allergen challenge. The IgE immune response is dependent on genetic and environmental factors; production of IgE results from complex interactions among B cells, T cells, mast cells, basophils,surface and adhesion molecules and various cytokines. New vaccination methods under investigation involve allergen-specific or nonspecific methodology. Allergen-specific methods currently being developed include allergoids, passive saturation of effector cells, plasmid DNA immunisation and antigen-antibody complexes. The mechanisms of immunotherapy using allergen-specific methods differ with the allergens and the route of immunisation used (parenteral, intranasal, sublingual, oral or bronchial). Many vaccines being developed at present comprise synthetic, recombinant or highly purified subunit antigens, which although they have increased safety may also be less immunogenic.It is hoped that the addition of adjuvants will overcome this drawback. Methods of increasing the dose of allergen while reducing the possibility of an anaphylactic reaction include the use of non-anaphylactic isoforms of the allergens, alteration of the tertiary structure of the allergens and construction of minimal allergen-derived T cell peptides. Nonspecific approaches include humanised anti-IgE antibodies,moderation of the T(H)2 cytokine network and antisense oligodeoxynucleotide therapy.
Collapse
Affiliation(s)
- J Bousquet
- Hôpital Arnaud de Villeneuve, INSERM V454, Montpellier, France.
| | | | | |
Collapse
|
23
|
Nelson HS. Some highlights of the first century of immunotherapy. Ann Allergy Asthma Immunol 2011; 107:417-21. [PMID: 22018612 DOI: 10.1016/j.anai.2011.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 05/17/2011] [Indexed: 11/28/2022]
|
24
|
Scichilone N, Minaldi C, Santagata R, Battaglia S, Camarda G, Bellia V. Anti-inflammatory effects of pre-seasonal Th1-adjuvant vaccine to Parietaria judaica in asthmatics. J Asthma Allergy 2011; 4:19-25. [PMID: 21660177 PMCID: PMC3108305 DOI: 10.2147/jaa.s17784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Indexed: 11/26/2022] Open
Abstract
Background: The ultra-short course pre-seasonal allergy vaccine, containing appropriate allergoids with the adjuvant monophosphoryl lipid A (MPL), may be effective in treating allergic symptoms. Objective: To explore the timing of the immunological responses to the pre-seasonal allergy vaccine. Methods: Four subcutaneous injections of the active product (Pollinex Quattro) were administered to 20 Parietaria-sensitive intermittent asthmatics (M/F: 12/8; age: 48 ± 10 years; FEV1% predicted: 108% ± 12%) during the 6 weeks prior to the start of the pollen season. Exhaled breath condensate (EBC) was collected immediately before the first and immediately after the last injections (t1 and t2), during the pollen season (t3) and after (t4) the pollen season. EBC was analyzed to determine the levels of pH and 8-isoprostane. Ten Parietaria-sensitive asthmatics served as the untreated control group at t1 and t2. Results: Measured pH levels were 7.64 ± 0.33 at t1, 7.67 ± 0.23 at t2, 7.72 ± 0.34 at t3, and 7.82 ± 0.34 at t4 (P = 0.049 vs baseline). 8-isoprostane levels were significantly lower than baseline at each visit (mean difference from baseline, for t2: −0.77 pg, P = 0.031; for t3: −0.92 pg, P = 0.010; for t4: −0.70 pg, P = 0.048). In the control group, pH levels were 7.73 ± 0.26 at baseline and did not change after 6 weeks (7.79 ± 0.25, P = 0.33). Similarly, the concentrations of 8-isoprostane in the control group were not different from those of the study group at baseline (P = 0.86), and the levels remained unchanged after 6 weeks (P = 0.58). Conclusion: These findings show that the ultra-short course of vaccine adjuvated with MPL acutely reduces the degree of airway inflammation, as expressed by markers of oxidative stress, and suggest that this reduction is maintained during and after the pollen season.
Collapse
Affiliation(s)
- Nicola Scichilone
- Dipartimento Biomedico di Medicina, Interna e Specialistica (Di.Bi.M.I.S.), Sezione di Pneumologia, University of Palermo, Palermo, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Finegold I, Dockhorn RJ, Ein D, Dolen WK, Oppenheimer J, Potter LH. Immunotherapy throughout the decades: from Noon to now. Ann Allergy Asthma Immunol 2011; 105:328-36; quiz 337, 358. [PMID: 21055658 DOI: 10.1016/j.anai.2010.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 08/08/2010] [Accepted: 08/16/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To review major milestones in the development of subcutaneous allergen immunotherapy in 20-year segments. DATA SOURCES Review of the literature available in textbooks and journals. STUDY SELECTION Articles and books addressing major achievements in the development of subcutaneous allergy immunotherapy were selected for inclusion in this review. RESULTS Immunotherapy administration has improved the lives of possibly millions of patients with hay fever. Asthmatic symptoms have been relieved if not ablated in millions as well. Insect venom hypersensitivity became treatable and highly effective. In the beginning years of immunotherapy, it was clear that immunotherapy worked; in the later years, the mechanisms for this efficacy were discovered. In this case, the therapy preceded its validation. Methods, materials, and safety have vastly improved. Postulated mechanisms explain much but not everything. CONCLUSIONS There is still research to be accomplished, improvements to be made, and, of course, patients to be made well.
Collapse
Affiliation(s)
- Ira Finegold
- Department of Allergy, St. Luke's-Roosevelt Hospital, New York, New York 10022, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol 2011; 127:18-27; quiz 28-9. [PMID: 21211639 DOI: 10.1016/j.jaci.2010.11.030] [Citation(s) in RCA: 309] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 11/22/2010] [Accepted: 11/22/2010] [Indexed: 01/05/2023]
Abstract
Allergen-specific immunotherapy has been used for 100 years as a desensitizing therapy for allergic diseases and represents the potentially curative and specific method of treatment. The mechanisms of action of allergen-specific immunotherapy include the very early desensitization effects, modulation of T-and B-cell responses and related antibody isotypes, and migration of eosinophils, basophils, and mast cells to tissues, as well as release of their mediators. Regulatory T (Treg) cells have been identified as key regulators of immunologic processes in peripheral tolerance to allergens. Skewing of allergen-specific effector T cells to a regulatory phenotype appears as a key event in the development of healthy immune response to allergens and successful outcome in patients undergoing allergen-specific immunotherapy. Naturally occurring forkhead box protein 3-positive CD4(+)CD25(+) Treg cells and inducible T(R)1 cells contribute to the control of allergen-specific immune responses in several major ways, which can be summarized as suppression of dendritic cells that support the generation of effector T cells; suppression of effector T(H)1, T(H)2, and T(H)17 cells; suppression of allergen-specific IgE and induction of IgG4; suppression of mast cells, basophils, and eosinophils; and suppression of effector T-cell migration to tissues. New strategies for immune intervention will likely include targeting of the molecular mechanisms of allergen tolerance and reciprocal regulation of effector and Treg cell subsets.
Collapse
Affiliation(s)
- Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| | | |
Collapse
|
27
|
Cox L, Nelson H, Lockey R, Calabria C, Chacko T, Finegold I, Nelson M, Weber R, Bernstein DI, Blessing-Moore J, Khan DA, Lang DM, Nicklas RA, Oppenheimer J, Portnoy JM, Randolph C, Schuller DE, Spector SL, Tilles S, Wallace D. Allergen immunotherapy: A practice parameter third update. J Allergy Clin Immunol 2011; 127:S1-55. [DOI: 10.1016/j.jaci.2010.09.034] [Citation(s) in RCA: 597] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 09/23/2010] [Indexed: 10/18/2022]
|
28
|
Calderon MA, Alves B, Jacobson M, Hurwitz B, Sheikh A, Durham S. Cochrane review: Allergen injection immunotherapy for seasonal allergic rhinitis. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/ebch.582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
29
|
Calderon M, Mösges R, Hellmich M, Demoly P. Towards evidence-based medicine in specific grass pollen immunotherapy. Allergy 2010; 65:420-34. [PMID: 20028374 DOI: 10.1111/j.1398-9995.2009.02292.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
When initiating grass pollen immunotherapy for seasonal allergic rhinoconjunctivitis, specialist physicians in many European countries must choose between modalities of differing pharmaceutical and regulatory status. We applied an evidence-based medicine (EBM) approach to commercially available subcutaneous and sublingual Gramineae grass pollen immunotherapies (SCIT and SLIT) by evaluating study design, populations, pollen seasons, treatment doses and durations, efficacy, quality of life, safety and compliance. After searching MEDLINE, Embase and the Cochrane Library up until January 2009, we identified 33 randomized, double-blind, placebo-controlled trials (including seven paediatric trials) with a total of 440 specific immunotherapy (SIT)-treated subjects in seven trials (0 paediatric) for SCIT with natural pollen extracts, 168 in three trials (0 paediatric) for SCIT with allergoids, 906 in 16 trials (five paediatric) for natural extract SLIT drops, 41 in two trials (one paediatric) for allergoid SLIT tablets and 1605 in five trials (two paediatric) for natural extract SLIT tablets. Trial design and quality varied significantly within and between SIT modalities. The multinational, rigorous trials of natural extract SLIT tablets correspond to a high level of evidence in adult and paediatric populations. The limited amount of published data on allergoids prevented us from judging the level of evidence for this modality.
Collapse
Affiliation(s)
- M Calderon
- Imperial College-NHLI, Royal Brompton Hospital, London, UK
| | | | | | | |
Collapse
|
30
|
Rosewich M, Schulze J, Eickmeier O, Telles T, Rose MA, Schubert R, Zielen S. Tolerance induction after specific immunotherapy with pollen allergoids adjuvanted by monophosphoryl lipid A in children. Clin Exp Immunol 2010; 160:403-10. [PMID: 20345983 DOI: 10.1111/j.1365-2249.2010.04106.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Specific immunotherapy (SIT) is a well-established and clinically effective treatment for allergic diseases. A pollen allergoid formulated with the T helper type 1 (Th1)-inducing adjuvant monophosphoryl lipid A (MPL) facilitates short-term SIT. Little is known about mechanisms of tolerance induction in this setting. In a prospective study, 34 patients allergic to grass pollen (25 male, nine female, median age 10.2 years) received a total of 44 SIT courses (20 in the first, 24 in the second) with MPL-adjuvanted pollen allergoids. Immunogenicity was measured by levels of specific immunoglobulin G (IgG(grass)) and IgG4(grass) by antibody blocking properties on basophil activation, and by induction of CD4(+), CD25(+) and forkhead box P3 (FoxP3(+)) regulatory T cells (T(reg)). Specific IgG and IgG4 levels increased only slightly in the first year of SIT. In the second year these changes reached significance (P < 0.0001). In keeping with these findings, we were able to show an increase of T(reg) cells and a decreased release of leukotrienes after the second year of treatment. In the first year of treatment we found little evidence for immunological changes. A significant antibody induction was seen only after the second course of SIT. Short-course immunotherapy with pollen allergoids formulated with the Th1-inducing adjuvant MPL needs at least two courses to establish tolerance.
Collapse
Affiliation(s)
- M Rosewich
- Paediatric Pulmonology and Allergology, Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Canonica GW, Bousquet J, Casale T, Lockey RF, Baena-Cagnani CE, Pawankar R, Potter PC, Bousquet PJ, Cox LS, Durham SR, Nelson HS, Passalacqua G, Ryan DP, Brozek JL, Compalati E, Dahl R, Delgado L, van Wijk RG, Gower RG, Ledford DK, Filho NR, Valovirta EJ, Yusuf OM, Zuberbier T. Sub-lingual immunotherapy: world allergy organization position paper 2009. World Allergy Organ J 2009; 2:233-81. [PMID: 23268425 PMCID: PMC3488881 DOI: 10.1097/wox.0b013e3181c6c379] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
32
|
CHAPTER 2: ALLERGEN SPECIFIC IMMUNOTHERAPY. World Allergy Organ J 2009. [DOI: 10.1097/01.wox.0000365044.24907.e9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
33
|
Amar SM, Harbeck RJ, Sills M, Silveira LJ, O'Brien H, Nelson HS. Response to sublingual immunotherapy with grass pollen extract: monotherapy versus combination in a multiallergen extract. J Allergy Clin Immunol 2009; 124:150-156.e1-5. [PMID: 19523672 DOI: 10.1016/j.jaci.2009.04.037] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 04/24/2009] [Accepted: 04/27/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND To date, there have been no randomized, double-blind studies showing the effectiveness of sublingual immunotherapy with multiple allergens. OBJECTIVE The purpose of this study was to examine whether the efficacy of sublingual immunotherapy (SLIT) with standardized timothy extract was reduced by combination with other allergen extracts. METHODS A single-center, randomized, double-blind, placebo-controlled trial with SLIT was conducted. After an observational grass season, SLIT was administered for 10 months to 54 patients randomized to 1 of 3 treatment arms: placebo, timothy extract (19 microg Phl p 5 daily) as monotherapy, or the same dose of timothy extract plus 9 additional pollen extracts. Symptom and medication scores were collected and titrated nasal challenges, titrated skin prick tests, specific IgE, IgG4 and cytokines release by timothy-stimulated lymphocyte proliferation were performed. RESULTS Perhaps because of a very low grass pollen season in 2008, there were no significant differences in medication or symptom scores in either treatment group compared with placebo. Compared with placebo, in the timothy monotherapy group, thresholds for titrated nasal challenge and skin prick tests (P = .03 and P = .001, respectively), and serum-specific IgG4 levels (P = .005) significantly increased, and IFN- gamma levels decreased (P = .02), whereas in the multiallergen group, there was significant improvement only in the titrated skin prick tests (P = .04) which was less than in the monotherapy group. There were no significant differences between the 2 active groups in any outcome measure, and both active groups experienced more adverse events than placebo. There were no systemic reactions. CONCLUSION Improvement in multiple relevant outcomes strongly suggests that SLIT with timothy extract alone was effective; however, the results for symptom and medication scores were not significant. The differences between multiple allergen SLIT and placebo only in skin sensitivity to timothy suggest a reduction in SLIT efficacy in this group. However, further studies are required to confirm these observations.
Collapse
|
34
|
Loewenstein C, Mueller RS. A review of allergen-specific immunotherapy in human and veterinary medicine. Vet Dermatol 2009; 20:84-98. [PMID: 19320877 DOI: 10.1111/j.1365-3164.2008.00727.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This article reviews allergen-specific immunotherapy in human and veterinary medicine. Current hypotheses of possible mechanisms of actions are outlined. Indications, success rates, adverse effects and factors influencing outcome of therapy are discussed in humans, dogs, cats and horses.
Collapse
Affiliation(s)
- Christine Loewenstein
- Tierärztliche Klinik für Kleintiere, Bereich Dermatologie, Im Langgewann 9, 65719 Hofheim, Germany
| | | |
Collapse
|
35
|
Akdis CA, Akdis M. Mechanisms and treatment of allergic disease in the big picture of regulatory T cells. J Allergy Clin Immunol 2009; 123:735-46; quiz 747-8. [PMID: 19348912 DOI: 10.1016/j.jaci.2009.02.030] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 02/25/2009] [Accepted: 02/25/2009] [Indexed: 11/19/2022]
Abstract
Various populations of regulatory T (Treg) cells have been shown to play a central role in the maintenance of peripheral homeostasis and the establishment of controlled immune responses. Their identification as key regulators of immunologic processes in peripheral tolerance to allergens has opened an important era in the prevention and treatment of allergic diseases. Both naturally occurring CD4+CD25+ Treg cells and inducible populations of allergen-specific, IL-10-secreting Treg type 1 (T(R)1) cells inhibit allergen-specific effector cells in experimental models. Skewing of allergen-specific effector T cells to a regulatory phenotype appears to be a key event in the development of healthy immune response to allergens and successful outcome in allergen-specific immunotherapy. Forkhead box protein 3-positive CD4+CD25+ Treg cells and T(R)1 cells contribute to the control of allergen-specific immune responses in several major ways, which can be summarized as suppression of dendritic cells that support the generation of effector T cells; suppression of effector T(H)1, T(H)2, and T(H)17 cells; suppression of allergen-specific IgE and induction of IgG4; suppression of mast cells, basophils, and eosinophils; interaction with resident tissue cells and remodeling; and suppression of effector T-cell migration to tissues. Current strategies for drug development and allergen-specific immunotherapy exploit these observations, with the potential for preventive therapies and cure for allergic diseases.
Collapse
Affiliation(s)
- Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland.
| | | |
Collapse
|
36
|
Rolland JM, Gardner LM, O'Hehir RE. Allergen-related approaches to immunotherapy. Pharmacol Ther 2008; 121:273-84. [PMID: 19111571 DOI: 10.1016/j.pharmthera.2008.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 11/18/2008] [Indexed: 01/20/2023]
Abstract
Allergic diseases, including asthma, rhinitis and eczema, represent a major health burden worldwide. Mainstay treatments are allergen avoidance where feasible and pharmacotherapy for symptom relief. For selected patients, allergen-specific immunotherapy (SIT) offers the prospect of long lasting clinical efficacy. SIT involves the administration of allergen extract using a standardized regimen, usually subcutaneously or increasingly sublingually. However, application of this potentially curative treatment is restricted, largely due to the risk of serious adverse events, especially in asthmatics and for potent allergens such as peanut, seafood and latex. New insights into immunological mechanisms underlying effective SIT and molecular characterization of allergens and their recognition by the immune system suggest strategies for refinement of SIT. Selective targeting of allergen-specific T cells, especially regulatory T cells, is likely to be pivotal for efficacy. Recombinant allergens lacking IgE reactivity and small T cell epitope-based peptides are being trialled clinically with evidence of efficacy without serious IgE-mediated adverse reactions. Adjuvants, either co-administered or incorporated into a recombinant allergen vaccine to target tolerogenic dendritic cells may also increase efficacy. The safer sublingual route of allergen administration is attracting interest and different allergen forms may be optimal for inducing tolerance by this route. Defined allergen-derived molecules or peptides offer ease of standardization and, coupled with appropriate targeting of immunoregulatory mechanisms, will result in more widespread clinical use of SIT. Adjunct therapies such as anti-IgE antibody and corticosteroids may minimize the likelihood of adverse reactions in those with severe allergic disease who would most benefit from this treatment.
Collapse
Affiliation(s)
- Jennifer M Rolland
- Department of Immunology, Monash University, Commercial Road, Melbourne Vic 3004, Australia.
| | | | | |
Collapse
|
37
|
Subiza J, Feliú A, Subiza JL, Uhlig J, Fernández-Caldas E. Cluster immunotherapy with a glutaraldehyde-modified mixture of grasses results in an improvement in specific nasal provocation tests in less than 2.5 months of treatment. Clin Exp Allergy 2008; 38:987-94. [PMID: 18445082 DOI: 10.1111/j.1365-2222.2008.02995.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Cluster immunotherapy is becoming increasingly used. It allows for a rapid build up phase and the administration of higher doses of allergen in a shorter period of time. OBJECTIVES To evaluate the effect of short-term pre-seasonal immunotherapy using a glutaraldeyde-modified allergen vaccine in reducing specific nasal hyperreactivity in nasal challenge tests. MATERIALS AND METHODS Thirty-three patients were selected. All patients had a positive history of allergic rhinitis and skin tests to grass pollen, although most of them (72.7%) were sensitized to other allergens as well. The study was conducted outside of the pollen season and the patients did not receive any pharmacological treatment during this period of time. Two randomized groups of patients were established; Group A: 22 patients (13 females and nine males) and Group B, 11 control patients (seven females and four males). Patients in Group A received immunotherapy with a vaccine containing 50% of the wild grasses Trisetum paniceum and Dactylis glomerata. All patients underwent titrated nasal provocation tests (NPT) before and after completion of the study (2.3 and 2.8 months for Groups A and B, respectively). The administration schedule consisted of 0.1 and 0.2 mL at day 1, followed by 0.3 and 0.5 mL at day 7, 0.5 mL after 2 weeks followed by 0.5 mL monthly. A single vial was used containing an allergen concentration of 10 000 TU/mL (105 microg of total protein and 24.6 microg of Group 1 plus 5 allergens/mL). A mean of 6.5 injections were administered to Group A patients between NPTs. RESULTS There were no significant differences between both groups at the beginning of the study (P=0.48). At the end, only Group A patients needed significant greater threshold concentrations for a positive NPT than at the beginning (P=0.002). CONCLUSIONS A short-term cluster pre-seasonal inmunotherapy with a modified vaccine containing a mixture of grass pollen is effective as determined by an objective measure after only a mean 2.3 months of treatment.
Collapse
Affiliation(s)
- J Subiza
- Centro de Asma y Alergia Subiza, Madrid, Spain.
| | | | | | | | | |
Collapse
|
38
|
Bousquet J, Khaltaev N, Cruz AA, Denburg J, Fokkens WJ, Togias A, Zuberbier T, Baena-Cagnani CE, Canonica GW, van Weel C, Agache I, Aït-Khaled N, Bachert C, Blaiss MS, Bonini S, Boulet LP, Bousquet PJ, Camargos P, Carlsen KH, Chen Y, Custovic A, Dahl R, Demoly P, Douagui H, Durham SR, van Wijk RG, Kalayci O, Kaliner MA, Kim YY, Kowalski ML, Kuna P, Le LTT, Lemiere C, Li J, Lockey RF, Mavale-Manuel S, Meltzer EO, Mohammad Y, Mullol J, Naclerio R, O'Hehir RE, Ohta K, Ouedraogo S, Palkonen S, Papadopoulos N, Passalacqua G, Pawankar R, Popov TA, Rabe KF, Rosado-Pinto J, Scadding GK, Simons FER, Toskala E, Valovirta E, van Cauwenberge P, Wang DY, Wickman M, Yawn BP, Yorgancioglu A, Yusuf OM, Zar H, Annesi-Maesano I, Bateman ED, Ben Kheder A, Boakye DA, Bouchard J, Burney P, Busse WW, Chan-Yeung M, Chavannes NH, Chuchalin A, Dolen WK, Emuzyte R, Grouse L, Humbert M, Jackson C, Johnston SL, Keith PK, Kemp JP, Klossek JM, Larenas-Linnemann D, Lipworth B, Malo JL, Marshall GD, Naspitz C, Nekam K, Niggemann B, Nizankowska-Mogilnicka E, Okamoto Y, Orru MP, Potter P, Price D, Stoloff SW, Vandenplas O, Viegi G, Williams D. Allergic Rhinitis and its Impact on Asthma (ARIA) 2008 update (in collaboration with the World Health Organization, GA(2)LEN and AllerGen). Allergy 2008; 63 Suppl 86:8-160. [PMID: 18331513 DOI: 10.1111/j.1398-9995.2007.01620.x] [Citation(s) in RCA: 3124] [Impact Index Per Article: 183.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
MESH Headings
- Adolescent
- Asthma/epidemiology
- Asthma/etiology
- Asthma/therapy
- Child
- Global Health
- Humans
- Prevalence
- Rhinitis, Allergic, Perennial/complications
- Rhinitis, Allergic, Perennial/diagnosis
- Rhinitis, Allergic, Perennial/epidemiology
- Rhinitis, Allergic, Perennial/therapy
- Rhinitis, Allergic, Seasonal/complications
- Rhinitis, Allergic, Seasonal/diagnosis
- Rhinitis, Allergic, Seasonal/epidemiology
- Rhinitis, Allergic, Seasonal/therapy
- Risk Factors
- World Health Organization
Collapse
Affiliation(s)
- J Bousquet
- University Hospital and INSERM, Hôpital Arnaud de Villeneuve, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wald M, Kahlert H, Weber B, Jankovic M, Keller W, Cromwell O, Nandy A, Fiebig H. Generation of a low immunoglobulin E-binding mutant of the timothy grass pollen major allergen Phl p 5a. Clin Exp Allergy 2007; 37:441-50. [PMID: 17359394 DOI: 10.1111/j.1365-2222.2007.02669.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Immunotherapy of grass pollen allergy is currently based on the administration of pollen extracts containing natural allergens. Specifically designed recombinant allergens with reduced IgE reactivity could be used in safer and more efficacious future therapy concepts. OBJECTIVES This study aimed to generate hypoallergenic variants of the timothy grass major allergen Phl p 5a as candidates for allergen-specific immunotherapy. METHODS Three deletion mutants were produced in Escherichia coli and subsequently purified. The overall IgE-binding capacity of the mutants was compared with the recombinant wild-type allergen by membrane blot and IgE-inhibition assays. The capacity for effector cell activation was determined in basophil activation assays. T cell proliferation assays with allergen-specific T cell lines were performed to confirm the retention of T cell reactivity. Structural properties were characterized by circular dichroism analysis and homogeneity by native isoelectric focusing. The deletion sites were mapped on homology models comprising the N- and C-terminal halves of Phl p 5a, respectively. RESULTS The double-deletion mutant rPhl p 5a Delta(94-113, 175-198) showed strongly diminished IgE binding in membrane blot and IgE-inhibition assays. Both deletions affect predominantly alpha-helical regions located in the N- and C-terminal halves of Phl p 5a, respectively. Whereas deletion of Delta175-198 alone was sufficient to cause a large reduction of the IgE reactivity in a subgroup of allergic sera, only the combination of both deletions was highly effective for all the sera tested. rPhl p 5a Delta(94-113, 175-198) consistently showed at least an 11.5-fold reduced capacity to activate basophils compared with the recombinant wild-type molecule, and the T cell proliferation assays demonstrated retention of T cell reactivity. CONCLUSION The mutant rPhl p 5a Delta(94-113, 175-198) fulfils the basic requirements for a hypoallergenic molecule suitable for a future immunotherapy of grass pollen allergy; it offers substantially reduced IgE binding and maintained T cell reactivity.
Collapse
Affiliation(s)
- M Wald
- Allergopharma J. Ganzer KG, Research & Development, Reinbek, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Radcliffe MJ, Lewith GT, Prescott P, Church MK, Holgate ST. Do skin prick and conjunctival provocation tests predict symptom severity in seasonal allergic rhinoconjunctivitis? Clin Exp Allergy 2007; 36:1488-93. [PMID: 17177671 DOI: 10.1111/j.1365-2222.2006.02594.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND In the investigation of seasonal allergic rhinoconjunctivitis (SAR), quantitative skin and conjunctival allergen challenge tests are used to measure individual allergen sensitivity. These tests are reproducible and relate well to prevalence but their relationship to symptom severity is less well established. OBJECTIVE We wished to determine if quantitative skin prick tests (QSPT) and conjunctival provocation tests (CPTs) using a single grass pollen allergen extract are reproducible and predict symptom severity in SAR. METHODS We retrospectively analysed data from 91 participants in a previously published randomized placebo controlled study of low dosage allergen immunotherapy who were randomized to receive placebo treatment. We examined the relationship between pre-seasonal QSPT, CPT and SAR symptoms. RESULTS We found a high level of reproducibility when repeated measures were compared for both the QSPT (P < 0.001) and the CPT (P < 0.001) and moderate correlation (0.49) between the standard skin prick test (SPT) and the QSPT (P < 0.001). We found weak negative correlation (-0.27) between the QSPT and the CPT (P < 0.001). We found no correlation between seasonal symptom, use of rescue medication or quality of life (QOL) scores and pre-seasonal QSPT or CPT. Conclusion In the assessment of seasonal rhinoconjunctivitis, quantitative skin and conjunctival allergen challenge tests are strongly reproducible, although there is no correlation between these tests and seasonal symptom, use of rescue medication or QOL scores.
Collapse
Affiliation(s)
- Michael J Radcliffe
- Infection Inflammation and Repair Research Division, School of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK.
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol 2007; 119:780-91. [PMID: 17321578 DOI: 10.1016/j.jaci.2007.01.022] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 01/14/2007] [Accepted: 01/16/2007] [Indexed: 10/23/2022]
Abstract
Allergen-specific immunotherapy (SIT) has been used for almost a century as a desensitizing therapy for allergic diseases and represents the only curative and specific method of treatment. Administration of appropriate concentrations of allergen extracts has been shown to be reproducibly effective when patients are carefully selected. The mechanisms by which allergen-SIT has its effects include the modulation of T-cell and B-cell responses and related antibody isotypes as well as effector cells of allergic inflammation, such as eosinophils, basophils, and mast cells. The balance between allergen-specific T-regulatory (Treg) and T(H)2 cells appears to be decisive in the development of allergic and healthy immune responses against allergens. Treg cells consistently represent the dominant subset specific for common environmental allergens in sensitized healthy individuals. In contrast, there is a high frequency of allergen-specific T(H)2 cells in patients with allergy. The induction of a tolerant state in peripheral T cells represents an essential step in allergen-SIT. Peripheral T-cell tolerance is characterized mainly by generation of allergen-specific Treg cells leading to suppressed T-cell proliferation and T(H)1 and T(H)2 cytokine responses against the allergen. This is accompanied by a significant increase in allergen-specific IgG(4), and also IgG(1) and IgA, and a decrease in IgE in the late stage of the disease. In addition, decreased tissue infiltration of mast cells and eosinophils and their mediator release including circulating basophils takes place. Current understanding of mechanisms of allergen-SIT, particularly the role of Treg cells in peripheral tolerance, may enable novel treatment strategies.
Collapse
Affiliation(s)
- Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | | |
Collapse
|
43
|
Calderon MA, Alves B, Jacobson M, Hurwitz B, Sheikh A, Durham S. Allergen injection immunotherapy for seasonal allergic rhinitis. Cochrane Database Syst Rev 2007; 2007:CD001936. [PMID: 17253469 PMCID: PMC7017974 DOI: 10.1002/14651858.cd001936.pub2] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Allergic rhinitis is the most common of the allergic diseases. Despite improved understanding of the pathophysiology of allergic rhinitis and advances in its pharmacological treatment, its prevalence has increased worldwide. For patients whose symptoms remain uncontrolled despite medical treatment, allergen injection immunotherapy is advised. An allergen-based treatment may reduce symptoms, the need for medication and modify the natural course of this disease. OBJECTIVES To evaluate the efficacy and safety of subcutaneous specific allergen immunotherapy, compared with placebo, for reducing symptoms and medication requirements in seasonal allergic rhinitis patients. SEARCH STRATEGY We searched the Cochrane Ear, Nose and Throat Disorders Group Trials Register, the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, Issue 1 2006), MEDLINE (1950 to 2006), EMBASE (1974 to 2006), Pre-MEDLINE, KOREAMED, INDMED, LILACS, PAKMEDINET, Scisearch, mRCT and the National Research Register. The date of the last search was February 2006. SELECTION CRITERIA All studies identified by the searches were assessed to identify randomised controlled trials involving participants with symptoms of seasonal allergic rhinitis and proven allergen sensitivity, treated with subcutaneous allergen specific immunotherapy or corresponding placebo. DATA COLLECTION AND ANALYSIS Two independent authors identified all studies reporting double-blind, placebo controlled randomised trials of specific immunotherapy in patients with seasonal allergic rhinitis due to tree, grass or weed pollens. Two authors independently performed quality assessment of studies. Data from identified studies were abstracted onto a standard extraction sheet and subsequently entered into RevMan 4.2.8. Analysis was performed using the Standardised Mean Difference (SMD) method and a random-effects model; P values < 0.05 were considered statistically significant. The primary outcome measures were symptom scores, medication use, quality of life and adverse events. MAIN RESULTS We retrieved 1111 publications of which 51 satisfied our inclusion criteria. In total there were 2871 participants (1645 active, 1226 placebo), each receiving on average 18 injections. Duration of immunotherapy varied from three days to three years. Symptom score data from 15 trials were suitable for meta-analysis and showed an overall reduction in the immunotherapy group (SMD -0.73 (95% CI -0.97 to -0.50, P < 0.00001)). Medication score data from 13 trials showed an overall reduction in the immunotherapy group (SMD of -0.57 (95% CI -0.82 to -0.33, p<0.00001)). Clinical interpretation of the effect size is difficult. Adrenaline was given in 0.13% (19 of 14085 injections) of those on active treatment and in 0.01% (1 of 8278 injections) of the placebo group for treatment of adverse events. There were no fatalities. AUTHORS' CONCLUSIONS This review has shown that specific allergen injection immunotherapy in suitably selected patients with seasonal allergic rhinitis results in a significant reduction in symptom scores and medication use. Injection immunotherapy has a known and relatively low risk of severe adverse events. We found no long-term consequences from adverse events.
Collapse
Affiliation(s)
- M A Calderon
- Royal Brompton Hospital, Department of Allergy and Respiratory Medicine, Imperial College School of Medicine at the National Heart and Lung Institute, London, UK, SW3 6LY.
| | | | | | | | | | | |
Collapse
|
44
|
Cox L. Accelerated immunotherapy schedules: review of efficacy and safety. Ann Allergy Asthma Immunol 2006; 97:126-37; quiz 137-40, 202. [PMID: 16937741 DOI: 10.1016/s1081-1206(10)60003-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To provide a comprehensive evaluation of accelerated immunotherapy build-up schedules in terms of adverse reactions and clinical efficacy. DATA SOURCES Peer-reviewed studies and review articles were selected from the PubMed database for articles published in the years 1976 to 2006 using the following keywords: rush, cluster immunotherapy in combination with allergic rhinitis, asthma, Hymenoptera, and imported fire ant. STUDY SELECTION Studies were selected if they provided safety and efficacy information on accelerated allergen immunotherapy schedules. Most of the studies reviewed were double-blind, placebo controlled, but some open-observational studies were included if they provided immunotherapy safety or other information the author thought was relevant. RESULTS A wide range of systemic reactions were reported in the literature with inhalant allergens: ranging from 27% to 100% of the patients in rush immunotherapy studies and 0% to 79% of patients in the cluster studies. Predictors of systemic reactions with rush immunotherapy were forced expiratory volume in 1 second less than 80% of predicted and a high degree of skin test reactivity. Premedication clearly reduces the risk of systemic reactions with rush immunotherapy, but the effect on cluster schedules was not as clear. CONCLUSION Accelerated immunotherapy build-up schedules in selected patients may provide a rapid alternative to conventional build-up schedules without a significant increase in risk.
Collapse
Affiliation(s)
- Linda Cox
- Nova Southeastern University School of Osteopathic Medicine, Ft Lauderdale, Florida, USA.
| |
Collapse
|
45
|
Keskin O, Tuncer A, Adalioglu G, Sekerel BE, Saçkesen C, Kalayci O. The effects of grass pollen allergoid immunotherapy on clinical and immunological parameters in children with allergic rhinitis. Pediatr Allergy Immunol 2006; 17:396-407. [PMID: 16925684 DOI: 10.1111/j.1399-3038.2006.00442.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Allergoid immunotherapy is a new form of allergen immunotherapy allowing safe administration of high allergen doses. There is limited information on the effects of allergoid immunotherapy in children with allergic rhinitis. To investigate the immunological and clinical effects of allergoid immunotherapy in children with allergic rhinitis due to grass pollen allergy. Children with allergic rhinitis were assigned to allergoid immunotherapy (n = 27) or control (n = 26, no immunotherapy) groups. Children in the immunotherapy group received seven injections of grass pollen allergoid immunotherapy before grass pollen season and continued to receive maintenance immunotherapy for 27 months. All patients were offered a pharmacotherapy regimen to be used on demand during the pollen seasons. Clinical and laboratory parameters were compared between the immunotherapy and control groups. The rhinoconjunctivitis symptom-medication score and asthma symptom score were lower in the immunotherapy group after 1 yr of maintenance immunotherapy (p < 0.01 for both). Skin test reactivity and nasal reactivity as determined by nasal provocation testing for grass pollen were significantly decreased after 1 yr of immunotherapy (p < 0.001 for both). The seasonal increase in bronchial reactivity and nasal lavage eosinophil cationic protein levels were prevented after the first year of immunotherapy (p < 0.05 for both). The seasonal increase in immunoglobulin (Ig)E decreased (p < 0.05) and grass-specific IgG, IgG(1) and IgG(4) increased significantly already at the end of the seven-injection build-up therapy (p < 0.001, for all). Interleukin (IL)-4 levels in the culture supernatants showed a steady decline from baseline at first and second year of immunotherapy (p < 0.001) but remained unchanged in the control group. Allergoid immunotherapy is an effective method in the treatment of grass pollen-induced allergic rhinitis in children and prevents the seasonal increase in bronchial hyper-reactivity. Changes in specific IgE and IgG levels and decreased IL-4 production in peripheral blood mononuclear cell culture supernatants may account for the observed clinical effects.
Collapse
Affiliation(s)
- Ozlem Keskin
- Pediatric Allergy and Asthma Unit, Hacettepe University School of Medicine, Hacettepe, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
46
|
Jutel M, Akdis M, Blaser K, Akdis CA. Mechanisms of allergen specific immunotherapy--T-cell tolerance and more. Allergy 2006; 61:796-807. [PMID: 16792576 DOI: 10.1111/j.1398-9995.2006.01175.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Specific immune suppression and induction of tolerance are essential processes in the regulation and circumvention of immune defence. The balance between allergen-specific T-regulatory (Treg) cells and T helper 2 cells appears to be decisive in the development of allergic and healthy immune response against allergens. Treg cells consistently represent the dominant subset specific for common environmental allergens in healthy individuals. In contrast, there is a high frequency of allergen-specific T helper 2 cells in allergic individuals. A decrease in interleukin (IL)-4, IL-5 and IL-13 production by allergen-specific CD4+ T cells due to the induction of peripheral T cell tolerance is the most essential step in allergen-specific immunotherapy (SIT). Suppressed proliferative and cytokine responses against the major allergens are induced by multiple suppressor factors, such as cytokines like IL-10 and transforming growth factor (TGF)-beta and cell surface molecules like cytotoxic T lymphocyte antigen-4, programmed death-1 and histamine receptor 2. There is considerable rationale for targeting T cells to increase efficacy of SIT. Such novel approaches include the use of modified allergens produced using recombinant DNA technology and adjuvants or additional drugs, which may increase the generation of allergen-specific peripheral tolerance. By the application of the recent knowledge in Treg cells and related mechanisms of peripheral tolerance, more rational and safer approaches are awaiting for the future of prevention and cure of allergic diseases.
Collapse
Affiliation(s)
- M Jutel
- Department of Internal Medicine and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | | | | | | |
Collapse
|
47
|
Kepley CL. Antigen-Induced Reduction in Mast Cell and Basophil Functional Responses due to Reduced Syk Protein Levels. Int Arch Allergy Immunol 2005; 138:29-39. [PMID: 16088210 DOI: 10.1159/000087355] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Accepted: 04/07/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The high-affinity IgE receptor, FcepsilonRI, is unresponsive on mast cells and basophils from people in several populations through an unknown mechanism. Similarly, FcepsilonRI-positive basophils from 'nonreleasers' are IgE-unresponsive and are deficient in the tyrosine kinase Syk. OBJECTIVE To test the hypothesis that cross-linking FcepsilonRI on mast cells and basophils leads to FcepsilonRI nonresponsiveness through reduction in Syk protein levels. METHODS Human mast cells and basophils were used to determine if FcepsilonRI hyporesponsiveness correlated with reduced Syk levels. RESULTS It is shown that suboptimal antigen challenge, that did not lead to significant mediator release, induced nonresponsiveness and correlated with reduced Syk. Other IgE-associated signaling molecules were unaffected by the same treatment. The ability of IgE-unresponsive mast cells to regain FcepsilonRI responsiveness is paralleled by increased cellular Syk levels in vitro. The reduction of Syk levels with suboptimal antigen concentrations was calcium independent and mediated through a proteasome-dependent mechanism. CONCLUSION These findings confirm and extend our knowledge about a novel regulatory mechanism for maintaining FcepsilonRI in a quiescent state. This mechanism may also explain why low concentrations of allergen given to patients during allergen immunotherapy induce FcepsilonRI nonresponsiveness and therapeutic benefit without inducing systemic anaphylaxis.
Collapse
Affiliation(s)
- Christopher L Kepley
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Virginia Commonwealth University Health Systems, Richmond, VA 23298, USA.
| |
Collapse
|
48
|
Asero R. Lack of de novo sensitization to tropomyosin in a group of mite-allergic patients treated by house dust mite-specific immunotherapy. Int Arch Allergy Immunol 2005; 137:62-5. [PMID: 15832051 DOI: 10.1159/000085105] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Accepted: 12/20/2004] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND There is some evidence that specific immunotherapy may induce de novo sensitization to proteins contained in the allergen extract. This raises the possibility that the novel sensitization to cross-reacting allergens may cause food allergies. OBJECTIVE The aim of this study was to assess whether injection of mite-specific immunotherapy (SIT) may induce sensitization to shrimp tropomyosin. METHODS Seventy mite-allergic subjects who did not show any shrimp sensitization at baseline were followed up for at least 3 years. Thirty-one of them underwent a 3-year SIT course, 39 served as controls. At the follow-up visit, SPTs with both fresh shrimp and commercial extract were performed, and patients underwent an open oral challenge with shrimp. RESULTS Neither patient nor control had become sensitized to shrimp allergens at the end of the study period. Both patients and controls reported the regular intake of crustaceans and/or molluscs. CONCLUSION Injection SIT with house dust mite extracts does not seem to induce de novo tropomyosin sensitization in mite-allergic patients.
Collapse
Affiliation(s)
- Riccardo Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italia.
| |
Collapse
|
49
|
Iglesias-Cadarso A, Hernández-Weigand P, Reaño M, Herrera I, Sánchez-Morillas L, Pérez-Pimiento A. A prospective safety study of allergen immunotherapy in daily clinical practice. Allergol Immunopathol (Madr) 2005; 32:278-83. [PMID: 15456624 DOI: 10.1016/s0301-0546(04)79255-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Despite its clinical effectiveness, allergen immunotherapy (AIT) remains controversial because serious systemic reactions can occur during its administration. Most of the studies on the safety of AIT are retrospective and use different methods, which frequently depart from daily clinical practice. OBJECTIVE The aim of this study was to determine risk factors for adverse reactions, especially systemic adverse reactions, produced during routine AIT administration. METHODS We registered 5,768 consecutive doses of standardized extracts administered to 273 patients in conventional schedules, following the recommendations on safety and data collection of the European Academy of Allergology and Clinical Immunology. Of the 273 patients, 236 were asthmatics, 28 had rhinitis and 9 received immunotherapy due to Hymenoptera anaphylaxis. RESULTS We examined 143 local reactions (2.48 % of the doses) and 145 systemic reactions (78 immediate and 67 delayed). Risk factors for developing an immediate systemic reaction were asthma severity, sensitization to molds, the most concentrated vials and a fall in peak expiratory flow of more than 15 % or an immediate systemic reaction in the previous dose. Late systemic reactions were significantly more frequent with less concentrated vials and in patients with late local reactions in the previous dose. No serious reactions were registered. CONCLUSIONS We believe that AIT is reliable when used with strict safety protocols and administered by specialized staff. Risk factors for adverse reactions to this type of treatment can be identified and reduced by systematic data collection.
Collapse
Affiliation(s)
- A Iglesias-Cadarso
- Servicio de Alergia, Centro de Especialidades de Argüelles, Hospital Puerta de Hierro, C/Quintana 11, 28008 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
50
|
Asero R. Efficacy of injection immunotherapy with ragweed and birch pollen in elderly patients. Int Arch Allergy Immunol 2004; 135:332-5. [PMID: 15564775 DOI: 10.1159/000082328] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2004] [Accepted: 08/27/2004] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The role of specific immunotherapy (SIT) in elderly allergic patients is still debated. OBJECTIVE The aim of this study was to assess whether SIT is associated with a better control of symptoms and a better quality of life also in older allergic patients. METHODS The effect of injection SIT was assessed in 39 patients [age >54 years, median 59; 29 patients (74%) with asthma, otherwise healthy] monosensitized to birch (n = 20) and ragweed (n = 19). Thirty-three younger subjects [age <50 years, median 35; 26 patients (79%) with asthma, otherwise healthy] monosensitized to birch (n = 10) and ragweed (n = 23), and 37 subjects >54 years old (25 males/12 females; age 55-79 years, median 59 years) monosensitized to birch (n = 18) and ragweed (n = 19) who refused to undergo SIT were enrolled as positive and negative controls, respectively. All groups had a disease duration <10 years; the disease was inadequately controlled with standard drug therapies in all cases. SIT was carried out using the same extracts and was administered using a perennial schedule both in patients and controls. Seasonal therapies were the same in all groups and were not changed after SIT was started. After 1-5 years of SIT, patients and controls were asked to quantify symptom reduction on a visual analogue scale: reductions >50% were considered significant. Moreover, the use of cetirizine and/or salbutamol as an adjunct to standard therapies during the last pollen season was assessed as an objective measure of SIT outcome. RESULTS 37/39 (95%) patients versus 32/33 (97%) controls submitted to SIT reported a symptom reduction >50% after 1-5 years of SIT (nonsignificant). The median clinical efficacy of SIT was 80% in both groups (nonsignificant). 27/37 controls not submitted to SIT did not report any change in symptom severity at the follow-up visits, whereas 10/37 reported a more severe disease (4 subjects reported the appearance of seasonal asthma) (p < 0.001 vs. patients submitted to SIT). Patients used less frequently cetirizine (p < 0.001) and/or salbutamol (p < 0.05) than controls not submitted to SIT. CONCLUSION Injection SIT can be considered an effective therapeutic option in otherwise healthy elderly patients with a short disease duration whose symptoms cannot be adequately controlled by drug therapies alone.
Collapse
Affiliation(s)
- Riccardo Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italia.
| |
Collapse
|