Skip to main content
Top
Published in: Current Allergy and Asthma Reports 11/2014

01-11-2014 | IMMUNOTHERAPY AND IMMUNOMODULATORS (L COX, SECTION EDITOR)

Immune Mechanisms of Sublingual Immunotherapy

Authors: David C. Jay, Kari C. Nadeau

Published in: Current Allergy and Asthma Reports | Issue 11/2014

Login to get access

Abstract

Sublingual immunotherapy (SLIT) is a well-established allergen-specific immunotherapy and a safe and effective strategy to reorient inappropriate immune responses in allergic patients. SLIT takes advantage of the tolerogenic environment of the oral mucosa to promote tolerance to the allergen. Several clinical studies have investigated the complex interplay of innate and adaptive immune responses that SLIT exploits. The oral immune system is composed of tolerogenic dendritic cells that, following uptake of allergen during SLIT, support the differentiation of T helper cell type 1 (Th1) and the induction of IL-10-producing regulatory T cells. Following SLIT, allergic disease-promoting T helper cell type 2 (Th2) responses shift to a Th1 inflammatory response, and IL-10 and transforming growth factor (TGF)-β production by regulatory T cells and tolerogenic dendritic cells suppress allergen-specific T cell responses. These immune changes occur both in the sublingual mucosa and in the periphery of a patient following SLIT. SLIT also promotes the synthesis of allergen-specific IgG and IgA antibodies that block allergen-IgE complex formation and binding to inflammatory cells, thus encouraging an anti-inflammatory environment. Several of these revealing findings have also paved the way for the identification of biomarkers of the clinical efficacy of SLIT. This review presents the emerging elucidation of the immune mechanisms mediated by SLIT.
Literature
1.
go back to reference Makatsori M, Scadding GW, Lombardo C, Bisoffi G, Ridolo E, Durham SR, et al. Dropouts in sublingual allergen immunotherapy trials—a systematic review. Allergy. 2014;69:571–80.PubMedCrossRef Makatsori M, Scadding GW, Lombardo C, Bisoffi G, Ridolo E, Durham SR, et al. Dropouts in sublingual allergen immunotherapy trials—a systematic review. Allergy. 2014;69:571–80.PubMedCrossRef
2.
go back to reference Linkov G, Toskala E. Sublingual immunotherapy: what we can learn from the European experience. Curr Opin Otolaryngol Head Neck Surg. 2014;22:208–10.PubMedCrossRef Linkov G, Toskala E. Sublingual immunotherapy: what we can learn from the European experience. Curr Opin Otolaryngol Head Neck Surg. 2014;22:208–10.PubMedCrossRef
4.
go back to reference Nelson HS. Sublingual immunotherapy: the U.S. experience. Curr Opin Allergy Clin Immunol. 2013;13:663–8.PubMedCrossRef Nelson HS. Sublingual immunotherapy: the U.S. experience. Curr Opin Allergy Clin Immunol. 2013;13:663–8.PubMedCrossRef
5.
go back to reference Moran TP, Vickery BP, Burks AW. Oral and sublingual immunotherapy for food allergy: current progress and future directions. Curr Opin Immunol. 2013;25:781–7.PubMedCrossRefPubMedCentral Moran TP, Vickery BP, Burks AW. Oral and sublingual immunotherapy for food allergy: current progress and future directions. Curr Opin Immunol. 2013;25:781–7.PubMedCrossRefPubMedCentral
6.
go back to reference Sato S, Yanagida N, Ogura K, Imai T, Utsunomiya T, Iikura K, et al. Clinical studies in oral allergen-specific immunotherapy: differences among allergens. Int Arch Allergy Immunol. 2014;164:1–9.PubMedCrossRef Sato S, Yanagida N, Ogura K, Imai T, Utsunomiya T, Iikura K, et al. Clinical studies in oral allergen-specific immunotherapy: differences among allergens. Int Arch Allergy Immunol. 2014;164:1–9.PubMedCrossRef
7.
go back to reference Dretzke J, Meadows A, Novielli N, Huissoon A, Fry-Smith A, Meads C. Subcutaneous and sublingual immunotherapy for seasonal allergic rhinitis: a systematic review and indirect comparison. J Allergy Clin Immunol. 2013;131:1361–6.PubMedCrossRef Dretzke J, Meadows A, Novielli N, Huissoon A, Fry-Smith A, Meads C. Subcutaneous and sublingual immunotherapy for seasonal allergic rhinitis: a systematic review and indirect comparison. J Allergy Clin Immunol. 2013;131:1361–6.PubMedCrossRef
8.
go back to reference Begin P, Chinthrajah RS, Nadeau KC. Oral immunotherapy for the treatment of food allergy. Hum Vaccines Immunotherapeutics. 2014;10:29–8.CrossRef Begin P, Chinthrajah RS, Nadeau KC. Oral immunotherapy for the treatment of food allergy. Hum Vaccines Immunotherapeutics. 2014;10:29–8.CrossRef
10.
go back to reference Cavkaytar O, Akdis CA, Akdis M. Modulation of immune responses by immunotherapy in allergic diseases. Curr Opin Pharmacol. 2014;17:30–7.CrossRef Cavkaytar O, Akdis CA, Akdis M. Modulation of immune responses by immunotherapy in allergic diseases. Curr Opin Pharmacol. 2014;17:30–7.CrossRef
11.
go back to reference Novak N, Gros E, Bieber T, Allam JP. Human skin and oral mucosal dendritic cells as ‘good guys’ and ‘bad guys’ in allergic immune responses. Clin Exp Immunol. 2010;161:28–33.PubMedPubMedCentral Novak N, Gros E, Bieber T, Allam JP. Human skin and oral mucosal dendritic cells as ‘good guys’ and ‘bad guys’ in allergic immune responses. Clin Exp Immunol. 2010;161:28–33.PubMedPubMedCentral
12.
13.
go back to reference Allam JP, Stojanovski G, Friedrichs N, Peng W, Bieber T, Wenzel J, et al. Distribution of Langerhans cells and mast cells within the human oral mucosa: new application sites of allergens in sublingual immunotherapy? Allergy. 2008;63:720–7.PubMedCrossRef Allam JP, Stojanovski G, Friedrichs N, Peng W, Bieber T, Wenzel J, et al. Distribution of Langerhans cells and mast cells within the human oral mucosa: new application sites of allergens in sublingual immunotherapy? Allergy. 2008;63:720–7.PubMedCrossRef
14.
15.
go back to reference Allam J-P, Würtzen PA, Reinartz M, Winter J, Vrtala S, Chen K-W, et al. Phl p 5 resorption in human oral mucosa leads to dose-dependent and time-dependent allergen binding by oral mucosal Langerhans cells, attenuates their maturation, and enhances their migratory and TGF-β1 and IL-10-producing properties. J Allergy Clin Immunol. 2010;126:638–45.PubMedCrossRef Allam J-P, Würtzen PA, Reinartz M, Winter J, Vrtala S, Chen K-W, et al. Phl p 5 resorption in human oral mucosa leads to dose-dependent and time-dependent allergen binding by oral mucosal Langerhans cells, attenuates their maturation, and enhances their migratory and TGF-β1 and IL-10-producing properties. J Allergy Clin Immunol. 2010;126:638–45.PubMedCrossRef
16.
go back to reference Angelini F, Pacciani V, Corrente S, Silenzi R, Di Pede A, Polito A, et al. Dendritic cells modification during sublingual immunotherapy in children with allergic symptoms to house dust mites. World J Pediatr. 2011;7:24–30.PubMedCrossRef Angelini F, Pacciani V, Corrente S, Silenzi R, Di Pede A, Polito A, et al. Dendritic cells modification during sublingual immunotherapy in children with allergic symptoms to house dust mites. World J Pediatr. 2011;7:24–30.PubMedCrossRef
17.
go back to reference Zimmer A, Bouley J, Le Mignon M, Pliquet E, Horiot S, Turfkruyer M, et al. A regulatory dendritic cell signature correlates with the clinical efficacy of allergen-specific sublingual immunotherapy. J Allergy Clin Immunol. 2012;129:1020–30.PubMedCrossRef Zimmer A, Bouley J, Le Mignon M, Pliquet E, Horiot S, Turfkruyer M, et al. A regulatory dendritic cell signature correlates with the clinical efficacy of allergen-specific sublingual immunotherapy. J Allergy Clin Immunol. 2012;129:1020–30.PubMedCrossRef
18.
go back to reference Fujimura T, Yonekura S, Taniguchi Y, Horiguchi S, Saito A, Yasueda H, et al. The induced regulatory T cell level, defined as the proportion of IL-10(+)Foxp3(+) cells among CD25(+)CD4(+) leukocytes, is a potential therapeutic biomarker for sublingual immunotherapy: a preliminary report. Int Arch Allergy Immunol. 2010;153:378–87.PubMedCrossRef Fujimura T, Yonekura S, Taniguchi Y, Horiguchi S, Saito A, Yasueda H, et al. The induced regulatory T cell level, defined as the proportion of IL-10(+)Foxp3(+) cells among CD25(+)CD4(+) leukocytes, is a potential therapeutic biomarker for sublingual immunotherapy: a preliminary report. Int Arch Allergy Immunol. 2010;153:378–87.PubMedCrossRef
19.
go back to reference Nagai Y, Shiraishi D, Tanaka Y, Nagasawa Y, Ohwada S, Shimauchi H, et al. Transportation of sublingual antigens across sublingual ductal epithelial cells to the ductal antigen-presenting cells in mice. Clin Exp Allergy. 2014. doi:10.1111/cea.12329.PubMed Nagai Y, Shiraishi D, Tanaka Y, Nagasawa Y, Ohwada S, Shimauchi H, et al. Transportation of sublingual antigens across sublingual ductal epithelial cells to the ductal antigen-presenting cells in mice. Clin Exp Allergy. 2014. doi:10.​1111/​cea.​12329.PubMed
20.
go back to reference Durham SR, Yang WH, Pedersen MR, Johansen N, Rak S. Sublingual immunotherapy with once-daily grass allergen tablets: a randomized controlled trial in seasonal allergic rhinoconjunctivitis. J Allergy Clin Immunol. 2006;117:802–9.PubMedCrossRef Durham SR, Yang WH, Pedersen MR, Johansen N, Rak S. Sublingual immunotherapy with once-daily grass allergen tablets: a randomized controlled trial in seasonal allergic rhinoconjunctivitis. J Allergy Clin Immunol. 2006;117:802–9.PubMedCrossRef
21.
22.
go back to reference Guida G, Boita M, Scirelli T, Bommarito L, Heffler E, Badiu I, et al. Innate and lymphocytic response of birch-allergic patients before and after sublingual immunotherapy. Allergy Asthma Proc. 2012;33:411–5.PubMedCrossRef Guida G, Boita M, Scirelli T, Bommarito L, Heffler E, Badiu I, et al. Innate and lymphocytic response of birch-allergic patients before and after sublingual immunotherapy. Allergy Asthma Proc. 2012;33:411–5.PubMedCrossRef
23.
go back to reference Mascarell L, Lombardi V, Zimmer A, Louise A, Tourdot S, Van Overtvelt L, et al. Mapping of the lingual immune system reveals the presence of both regulatory and effector CD4+ T cells. Clin Exp Allergy. 2009;39:1910–9.PubMedCrossRef Mascarell L, Lombardi V, Zimmer A, Louise A, Tourdot S, Van Overtvelt L, et al. Mapping of the lingual immune system reveals the presence of both regulatory and effector CD4+ T cells. Clin Exp Allergy. 2009;39:1910–9.PubMedCrossRef
24.
go back to reference Ciprandi G, Fenoglio D, Cirillo I, Vizzaccaro A, Ferrera A, Tosca MA, et al. Induction of interleukin 10 by sublingual immunotherapy for house dust mites: a preliminary report. Ann Allergy Asthma Immunol. 2005;95:38–44.PubMedCrossRef Ciprandi G, Fenoglio D, Cirillo I, Vizzaccaro A, Ferrera A, Tosca MA, et al. Induction of interleukin 10 by sublingual immunotherapy for house dust mites: a preliminary report. Ann Allergy Asthma Immunol. 2005;95:38–44.PubMedCrossRef
25.
go back to reference Burastero SE, Mistrello G, Falagiani P, Paolucci C, Breda D, Roncarolo D, et al. Effect of sublingual immunotherapy with grass monomeric allergoid on allergen-specific T-cell proliferation and interleukin 10 production. Ann Allergy Asthma Immunol. 2008;100:343–50.PubMedCrossRef Burastero SE, Mistrello G, Falagiani P, Paolucci C, Breda D, Roncarolo D, et al. Effect of sublingual immunotherapy with grass monomeric allergoid on allergen-specific T-cell proliferation and interleukin 10 production. Ann Allergy Asthma Immunol. 2008;100:343–50.PubMedCrossRef
26.
go back to reference Bohle B, Kinaciyan T, Gerstmayr M, Radakovics A, Jahn-Schmid B, Ebner C. Sublingual immunotherapy induces IL-10-producing T regulatory cells, allergen-specific T-cell tolerance, and immune deviation. J Allergy Clin Immunol. 2007;120:707–13.PubMedCrossRef Bohle B, Kinaciyan T, Gerstmayr M, Radakovics A, Jahn-Schmid B, Ebner C. Sublingual immunotherapy induces IL-10-producing T regulatory cells, allergen-specific T-cell tolerance, and immune deviation. J Allergy Clin Immunol. 2007;120:707–13.PubMedCrossRef
27.
go back to reference Bahceciler NN, Galip N. Comparing subcutaneous and sublingual immunotherapy: what do we know? Curr Opin Allergy Clin Immunol. 2012;12:640–7.PubMedCrossRef Bahceciler NN, Galip N. Comparing subcutaneous and sublingual immunotherapy: what do we know? Curr Opin Allergy Clin Immunol. 2012;12:640–7.PubMedCrossRef
28.
go back to reference Jones SM, Burks AW, Dupont C. State of the art on food allergen immunotherapy: oral, sublingual, and epicutaneous. J Allergy Clin Immunol. 2014;133:318–23.PubMedCrossRef Jones SM, Burks AW, Dupont C. State of the art on food allergen immunotherapy: oral, sublingual, and epicutaneous. J Allergy Clin Immunol. 2014;133:318–23.PubMedCrossRef
29.
go back to reference Pellerin L, Jenks J, Bégin P, Bacchetta R, Nadeau K. Regulatory T cells and their roles in immune dysregulation and allergy. Immunol Res. 2014;58:358–68.PubMedCrossRefPubMedCentral Pellerin L, Jenks J, Bégin P, Bacchetta R, Nadeau K. Regulatory T cells and their roles in immune dysregulation and allergy. Immunol Res. 2014;58:358–68.PubMedCrossRefPubMedCentral
31.
go back to reference Francis JN, Till SJ, Durham SR. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. J Allergy Clin Immunol. 2003;111:1255–61.PubMedCrossRef Francis JN, Till SJ, Durham SR. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. J Allergy Clin Immunol. 2003;111:1255–61.PubMedCrossRef
32.
go back to reference Kim EH, Bird JA, Kulis M, Laubach S, Pons L, Shreffler W, et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2011;127:640–6.PubMedCrossRefPubMedCentral Kim EH, Bird JA, Kulis M, Laubach S, Pons L, Shreffler W, et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2011;127:640–6.PubMedCrossRefPubMedCentral
33.
go back to reference O’Hehir RE, Gardner LM, de Leon MP, Hales BJ, Biondo M, Douglass JA, et al. House dust mite sublingual immunotherapy. Am J Respir Crit Care Med. 2009;180:936–47.PubMedCrossRef O’Hehir RE, Gardner LM, de Leon MP, Hales BJ, Biondo M, Douglass JA, et al. House dust mite sublingual immunotherapy. Am J Respir Crit Care Med. 2009;180:936–47.PubMedCrossRef
34.
go back to reference Eifan AO, Akkoc T, Yildiz A, Keles S, Ozdemir C, Bahceciler NN, et al. Clinical efficacy and immunological mechanisms of sublingual and subcutaneous immunotherapy in asthmatic/rhinitis children sensitized to house dust mite: an open randomized controlled trial. Clin Exp Allergy. 2010;40:922–32.PubMedCrossRef Eifan AO, Akkoc T, Yildiz A, Keles S, Ozdemir C, Bahceciler NN, et al. Clinical efficacy and immunological mechanisms of sublingual and subcutaneous immunotherapy in asthmatic/rhinitis children sensitized to house dust mite: an open randomized controlled trial. Clin Exp Allergy. 2010;40:922–32.PubMedCrossRef
35.
go back to reference Yamanaka K-I, Yuta A, Kakeda M, Sasaki R, Kitagawa H, Gabazza EC, et al. Induction of IL-10-producing regulatory T cells with TCR diversity by epitope-specific immunotherapy in pollinosis. J Allergy Clin Immunol. 2009;124:842–5.PubMedCrossRef Yamanaka K-I, Yuta A, Kakeda M, Sasaki R, Kitagawa H, Gabazza EC, et al. Induction of IL-10-producing regulatory T cells with TCR diversity by epitope-specific immunotherapy in pollinosis. J Allergy Clin Immunol. 2009;124:842–5.PubMedCrossRef
36.
go back to reference Scadding GW, Shamji MH, Jacobson MR, Lee DI, Wilson D, Lima MT, et al. Sublingual grass pollen immunotherapy is associated with increases in sublingual Foxp3-expressing cells and elevated allergen-specific immunoglobulin G4, immunoglobulin A and serum inhibitory activity for immunoglobulin E-facilitated allergen binding to B cells. Clin Exp Allergy. 2010;40:598–606.PubMed Scadding GW, Shamji MH, Jacobson MR, Lee DI, Wilson D, Lima MT, et al. Sublingual grass pollen immunotherapy is associated with increases in sublingual Foxp3-expressing cells and elevated allergen-specific immunoglobulin G4, immunoglobulin A and serum inhibitory activity for immunoglobulin E-facilitated allergen binding to B cells. Clin Exp Allergy. 2010;40:598–606.PubMed
37.•
go back to reference Bonvalet M, Moussu H, Wambre E, Ricarte C, Horiot S, Rimaniol AC, et al. Allergen-specific CD4+ T cell responses in peripheral blood do not predict the early onset of clinical efficacy during grass pollen sublingual immunotherapy. Clin Exp Allergy. 2012;42:1745–55. By using tetramers to observe allergen-specific T cell responses following SLIT, this study offers a thorough contradiction to previous findings that the changes in the induction of Tregs and the shifting of Th2 to Th1 responses directly correlated with the early clinical efficacy of SLIT.PubMedCrossRef Bonvalet M, Moussu H, Wambre E, Ricarte C, Horiot S, Rimaniol AC, et al. Allergen-specific CD4+ T cell responses in peripheral blood do not predict the early onset of clinical efficacy during grass pollen sublingual immunotherapy. Clin Exp Allergy. 2012;42:1745–55. By using tetramers to observe allergen-specific T cell responses following SLIT, this study offers a thorough contradiction to previous findings that the changes in the induction of Tregs and the shifting of Th2 to Th1 responses directly correlated with the early clinical efficacy of SLIT.PubMedCrossRef
38.•
go back to reference Suárez-Fueyo A, Ramos T, Galán A, Jimeno L, Wurtzen PA, Marin A, et al. Grass tablet sublingual immunotherapy downregulates the TH2 cytokine response followed by regulatory T-cell generation. J Allergy Clin Immunol. 2014;133:130–8. This detailed longitudinal study of the systemic effects of SLIT clearly shows the early and late phases of immune modulations. These changes observed included the early exacerbation of Th2 responses and serum allergen-specific IgE, followed by the later shift of Th2 to Th1 responses and increased blocking allergen-specific IgG4 as well as the induction of Tregs.PubMedCrossRef Suárez-Fueyo A, Ramos T, Galán A, Jimeno L, Wurtzen PA, Marin A, et al. Grass tablet sublingual immunotherapy downregulates the TH2 cytokine response followed by regulatory T-cell generation. J Allergy Clin Immunol. 2014;133:130–8. This detailed longitudinal study of the systemic effects of SLIT clearly shows the early and late phases of immune modulations. These changes observed included the early exacerbation of Th2 responses and serum allergen-specific IgE, followed by the later shift of Th2 to Th1 responses and increased blocking allergen-specific IgG4 as well as the induction of Tregs.PubMedCrossRef
39.
go back to reference Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.PubMedCrossRef Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.PubMedCrossRef
40.
go back to reference Wieczorek G, Asemissen A, Model F, Turbachova I, Floess S, Liebenberg V, et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue. Cancer Res. 2009;69:599–608.PubMedCrossRef Wieczorek G, Asemissen A, Model F, Turbachova I, Floess S, Liebenberg V, et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue. Cancer Res. 2009;69:599–608.PubMedCrossRef
42.•
go back to reference Swamy RS, Reshamwala N, Hunter T, Vissamsetti S, Santos CB, Baroody FM, et al. Epigenetic modifications and improved regulatory T-cell function in subjects undergoing dual sublingual immunotherapy. J Allergy Clin Immunol. 2012;130:215–24. This study clearly shows how following SLIT, epigenetic regulation of the Foxp3 locus through methylation of CpG sites determines the stability and function of induced regulatory T cells, and that this may be a new biomarker for monitoring the clinical efficacy of SLIT.PubMedCrossRefPubMedCentral Swamy RS, Reshamwala N, Hunter T, Vissamsetti S, Santos CB, Baroody FM, et al. Epigenetic modifications and improved regulatory T-cell function in subjects undergoing dual sublingual immunotherapy. J Allergy Clin Immunol. 2012;130:215–24. This study clearly shows how following SLIT, epigenetic regulation of the Foxp3 locus through methylation of CpG sites determines the stability and function of induced regulatory T cells, and that this may be a new biomarker for monitoring the clinical efficacy of SLIT.PubMedCrossRefPubMedCentral
43.
go back to reference Syed A, Garcia MA, Lyu S-C, Bucayu R, Kohli A, Ishida S, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol. 2014;133:500–10.PubMedCrossRef Syed A, Garcia MA, Lyu S-C, Bucayu R, Kohli A, Ishida S, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol. 2014;133:500–10.PubMedCrossRef
44.
go back to reference James LK, Shamji MH, Walker SM, Wilson DR, Wachholz PA, Francis JN, et al. Long-term tolerance after allergen immunotherapy is accompanied by selective persistence of blocking antibodies. J Allergy Clin Immunol. 2011;127:509–16.PubMedCrossRef James LK, Shamji MH, Walker SM, Wilson DR, Wachholz PA, Francis JN, et al. Long-term tolerance after allergen immunotherapy is accompanied by selective persistence of blocking antibodies. J Allergy Clin Immunol. 2011;127:509–16.PubMedCrossRef
45.
go back to reference Queirós MGJ, Silva DAO, Siman IL, Ynoue LH, Araújo NS, Pereira FL, et al. Modulation of mucosal/systemic antibody response after sublingual immunotherapy in mite-allergic children. Pediatr Allergy Immunol. 2013;24:752–61.PubMedCrossRef Queirós MGJ, Silva DAO, Siman IL, Ynoue LH, Araújo NS, Pereira FL, et al. Modulation of mucosal/systemic antibody response after sublingual immunotherapy in mite-allergic children. Pediatr Allergy Immunol. 2013;24:752–61.PubMedCrossRef
46.
go back to reference Ott H, Sieber J, Brehler R, Fölster-Holst R, Kapp A, Klimek L, et al. Efficacy of grass pollen sublingual immunotherapy for three consecutive seasons and after cessation of treatment: the ECRIT study. Allergy. 2009;64:1394–401.PubMedCrossRef Ott H, Sieber J, Brehler R, Fölster-Holst R, Kapp A, Klimek L, et al. Efficacy of grass pollen sublingual immunotherapy for three consecutive seasons and after cessation of treatment: the ECRIT study. Allergy. 2009;64:1394–401.PubMedCrossRef
47.
go back to reference Wahn U, Klimek L, Ploszczuk A, Adelt T, Sandner B, Trebas-Pietras E, et al. High-dose sublingual immunotherapy with single-dose aqueous grass pollen extract in children is effective and safe: a double-blind, placebo-controlled study. J Allergy Clin Immunol. 2012;130:886–93.PubMedCrossRef Wahn U, Klimek L, Ploszczuk A, Adelt T, Sandner B, Trebas-Pietras E, et al. High-dose sublingual immunotherapy with single-dose aqueous grass pollen extract in children is effective and safe: a double-blind, placebo-controlled study. J Allergy Clin Immunol. 2012;130:886–93.PubMedCrossRef
48.
go back to reference Creticos PS, Esch RE, Couroux P, Gentile D, D’Angelo P, Whitlow B, et al. Randomized, double-blind, placebo-controlled trial of standardized ragweed sublingual-liquid immunotherapy for allergic rhinoconjunctivitis. J Allergy Clin Immunol. 2014;133:751–8.PubMedCrossRef Creticos PS, Esch RE, Couroux P, Gentile D, D’Angelo P, Whitlow B, et al. Randomized, double-blind, placebo-controlled trial of standardized ragweed sublingual-liquid immunotherapy for allergic rhinoconjunctivitis. J Allergy Clin Immunol. 2014;133:751–8.PubMedCrossRef
49.
50.
go back to reference Wood RA, Togias A, Wildfire J, Visness CM, Matsui EC, Gruchalla R, et al. Development of cockroach immunotherapy by the inner-city asthma consortium. J Allergy Clin Immunol. 2014;133:846–52.PubMedCrossRef Wood RA, Togias A, Wildfire J, Visness CM, Matsui EC, Gruchalla R, et al. Development of cockroach immunotherapy by the inner-city asthma consortium. J Allergy Clin Immunol. 2014;133:846–52.PubMedCrossRef
51.
go back to reference Radulovic S, Wilson D, Calderon M, Durham S. Systematic reviews of sublingual immunotherapy (SLIT). Allergy. 2011;66:740–52.PubMedCrossRef Radulovic S, Wilson D, Calderon M, Durham S. Systematic reviews of sublingual immunotherapy (SLIT). Allergy. 2011;66:740–52.PubMedCrossRef
52.
go back to reference Fernández-Rivas M, Garrido Fernández S, Nadal JA, Alonso Díaz De Durana MD, García BE, González-Mancebo E, et al. Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract. Allergy. 2009;64:876–83.PubMedCrossRef Fernández-Rivas M, Garrido Fernández S, Nadal JA, Alonso Díaz De Durana MD, García BE, González-Mancebo E, et al. Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract. Allergy. 2009;64:876–83.PubMedCrossRef
53.
go back to reference Keet CA, Frischmeyer-Guerrerio PA, Thyagarajan A, Schroeder JT, Hamilton RG, Boden S, et al. The safety and efficacy of sublingual and oral immunotherapy for milk allergy. J Allergy Clin Immunol. 2012;129:448–55.PubMedCrossRefPubMedCentral Keet CA, Frischmeyer-Guerrerio PA, Thyagarajan A, Schroeder JT, Hamilton RG, Boden S, et al. The safety and efficacy of sublingual and oral immunotherapy for milk allergy. J Allergy Clin Immunol. 2012;129:448–55.PubMedCrossRefPubMedCentral
54.
go back to reference Baron-Bodo V, Horiot S, Lautrette A, Chabre H, Drucbert AS, Danzé PM, et al. Heterogeneity of antibody responses among clinical responders during grass pollen sublingual immunotherapy. Clin Exp Allergy. 2013;43:1362–73.PubMedCrossRef Baron-Bodo V, Horiot S, Lautrette A, Chabre H, Drucbert AS, Danzé PM, et al. Heterogeneity of antibody responses among clinical responders during grass pollen sublingual immunotherapy. Clin Exp Allergy. 2013;43:1362–73.PubMedCrossRef
55.
go back to reference Pereira C, Bartolome B, Asturias JA, Ibarrola I, Tavares B, Loureiro G, et al. Specific sublingual immunotherapy with peach LTP (Pru p 3). One year treatment: a case report. Cases J. 2009;2:6553.PubMedCrossRefPubMedCentral Pereira C, Bartolome B, Asturias JA, Ibarrola I, Tavares B, Loureiro G, et al. Specific sublingual immunotherapy with peach LTP (Pru p 3). One year treatment: a case report. Cases J. 2009;2:6553.PubMedCrossRefPubMedCentral
57.
go back to reference Gloudemans AK, Lambrecht BN, Smits HH. Potential of immunoglobulin a to prevent allergic asthma. Clin Dev Immunol. 2013;2013:12.CrossRef Gloudemans AK, Lambrecht BN, Smits HH. Potential of immunoglobulin a to prevent allergic asthma. Clin Dev Immunol. 2013;2013:12.CrossRef
58.
go back to reference D.O. Miranda, D.A.O. Silva, J.F.C. Fernandes, Queirós, M.G.J., H.F. Chiba, L.H. Ynoue, R.O. Resende, J.D.O. Pena, S.-S.J. Sung, G.R.S. Segundo, E.A. Taketomi (2011) Serum and salivary IgE, IgA, and IgG4 antibodies to Dermatophagoides pteronyssinus and its major allergens, Der p1 and Der p2, in allergic and nonallergic children. Clin Dev Immunol 2011:11. D.O. Miranda, D.A.O. Silva, J.F.C. Fernandes, Queirós, M.G.J., H.F. Chiba, L.H. Ynoue, R.O. Resende, J.D.O. Pena, S.-S.J. Sung, G.R.S. Segundo, E.A. Taketomi (2011) Serum and salivary IgE, IgA, and IgG4 antibodies to Dermatophagoides pteronyssinus and its major allergens, Der p1 and Der p2, in allergic and nonallergic children. Clin Dev Immunol 2011:11.
59.
go back to reference Kulis M, Saba K, Kim EH, Bird JA, Kamilaris N, Vickery BP, et al. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut sublingual immunotherapy. J Allergy Clin Immunol. 2012;129:1159–62.PubMedCrossRefPubMedCentral Kulis M, Saba K, Kim EH, Bird JA, Kamilaris N, Vickery BP, et al. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut sublingual immunotherapy. J Allergy Clin Immunol. 2012;129:1159–62.PubMedCrossRefPubMedCentral
60.
go back to reference Skoner D, Gentile D, Bush R, Fasano MB, McLaughlin A, Esch RE. Sublingual immunotherapy in patients with allergic rhinoconjunctivitis caused by ragweed pollen. J Allergy Clin Immunol. 2010;125:660–6.PubMedCrossRef Skoner D, Gentile D, Bush R, Fasano MB, McLaughlin A, Esch RE. Sublingual immunotherapy in patients with allergic rhinoconjunctivitis caused by ragweed pollen. J Allergy Clin Immunol. 2010;125:660–6.PubMedCrossRef
Metadata
Title
Immune Mechanisms of Sublingual Immunotherapy
Authors
David C. Jay
Kari C. Nadeau
Publication date
01-11-2014
Publisher
Springer US
Published in
Current Allergy and Asthma Reports / Issue 11/2014
Print ISSN: 1529-7322
Electronic ISSN: 1534-6315
DOI
https://doi.org/10.1007/s11882-014-0473-1

Other articles of this Issue 11/2014

Current Allergy and Asthma Reports 11/2014 Go to the issue

ANAPHYLAXIS AND DRUG ALLERGY (DA KHAN AND M CASTELLS, SECTION EDITORS)

Penicillin and Beta-Lactam Allergy: Epidemiology and Diagnosis

OTITIS (DP SKONER, SECTION EDITOR)

Role of Obesity in Otitis Media in Children

Pediatric Allergy and Immunology (JM Portnoy and CE Ciaccio, Section Editors)

Empowering the Child and Caregiver: Yellow Zone Asthma Action Plan

ALLERGIC SKIN DISEASES (L FONACIER, SECTION EDITOR)

Hand Dermatitis: an Allergist’s Nightmare