Immunology
Mechanisms of Sublingual Immunotherapy

https://doi.org/10.1016/j.iac.2011.02.005Get rights and content

Section snippets

Current proposed mechanism of SLIT

Fig. 1 illustrates a hypothetical model of the pathway of allergen and downstream effects of SLIT. In this model, allergen is taken up by submucosal dendritic cells (perhaps oral Langerhans cells), which then migrate to local regional lymphoid tissues, including submandibular and cervical lymph nodes. There, dendritic cells present peptide fragments to allergen-specific T cells in a protolerogenic manner, resulting in inhibition of activation and proli,feration of Th2 cells and stimulation of

Allergen uptake and presentation in SLIT

Some years ago, a series of pharmacodynamic studies by Bagnasco and colleagues15, 16 explored the local and systemic distribution of allergen in vivo after application to the human sublingual region. Radiolabeled Parietaria allergen was tracked, by both local scintillography and plasma radioactivity, to identify the distribution of allergen after sublingual application, compared with oral (swallowed) allergen. Not only was allergen retained within the sublingual space for several hours, with

Downstream immunologic effects of SLIT

In considering the immunologic effects of effective SLIT, it seems prudent to first briefly consider the better-established effects of SCIT.33, 34 SCIT has been shown to have rapid desensitizing effects on mast cells and basophils, providing some protection from anaphylactic responses in days to weeks. Within weeks there is evidence of T-cell tolerance and the induction of regulatory T cells, including cells expressing IL-10 and TGF-β, as well as the prototypic regulatory cell transcription

Antibody effects of SLIT

Allowing for a break with the proposed chronology of events, as outlined earlier for SCIT, most clinical studies have investigated the effect of SLIT on allergen-specific antibody levels. The observed changes do seem to reflect those of SCIT, albeit at a lower magnitude.

In large, randomized, placebo-controlled trials of grass pollen SLIT significant increases in IgG,36 IgG4,14, 37 or IgE-blocking antibody38 have been recorded, becoming significant versus placebo from about 8 weeks of treatment.

T-cell and cytokine responses during SLIT

Data concerning the effects of SLIT on T-cell proliferation, cytokine secretion, and on the induction of putative regulatory T cells have been varied and at times conflicting. As with data concerning antibodies, this situation may be the result of different allergens, doses, and treatment regimens, but in addition, may also reflect the increased complexity and variability of the assays used by different researchers.

Local mucosal effects of SLIT

SCIT has been shown to reduce allergic inflammation at mucosal sites such as the nose and lung, including reductions in mast cell and eosinophil numbers.61, 62 More recently, phenotypic regulatory T cells have been identified in the nasal mucosa of SCIT-treated patients during natural seasonal pollen exposure.63 These regulatory cells likely have direct antiinflammatory or tolerogenic effects within the target organ. Somewhat less is known regarding local mucosal effects of SLIT.

A

Time course and long-term effects of SLIT

The clinical time course of onset of specific immunotherapy can be difficult to measure, especially when aeroallergen exposure can differ from season to season, day to day, and between individuals. A recent study made use of an artificial allergen exposure chamber to expose both grass pollen SLIT and placebo-treated patients to standardized levels of pollen before and then at regular intervals during the course of treatment.69 Moreover, because the study was entirely conducted outside the

Areas of uncertainty and future study

Although the oral Langerhans cell is an attractive candidate for the role of key antigen-presenting cell in SLIT, it is clear, at least from murine studies, that alternative antigen-presenting cells are also likely involved, including other monocyte-derived dendritic cells, macrophages, and potentially B cells.19, 25 Furthermore, the mechanism of allergen uptake by these cells is as yet uncertain. Further clinical studies using adjuvants are needed, particularly in view of their apparent

Summary

On a systemic level, current evidence suggests SLIT shares many mechanistic properties in common with SCIT. Evidence points to similar, although less pronounced, effects on specific IgG, IgE, and IgA. Furthermore, IgG antibodies at least seem to be functional. Effects on the T-cell compartment are less certain, but a picture of regulatory and Th1 cell induction and Th2 inhibition is emerging.

Understanding of the local mucosal mechanisms is rapidly progressing, with evidence accumulating for a

Acknowledgments

The authors are grateful to Dr Pablo Rodríguez del Río for the construction of Fig. 4.

First page preview

First page preview
Click to open first page preview

References (79)

  • A. Didier et al.

    Optimal dose, efficacy, and safety of once-daily sublingual immunotherapy with a 5-grass pollen tablet for seasonal allergic rhinitis

    J Allergy Clin Immunol

    (2007)
  • B. Bohle et al.

    Sublingual immunotherapy induces IL-10-producing T regulatory cells, allergen-specific T-cell tolerance, and immune deviation

    J Allergy Clin Immunol

    (2007)
  • G. Ciprandi et al.

    Induction of interleukin 10 by sublingual immunotherapy for house dust mites: a preliminary report

    Ann Allergy Asthma Immunol

    (2005)
  • G. Ciprandi et al.

    Sublingual immunotherapy induces spirometric improvement associated with IL-10 production: preliminary reports

    Int Immunopharmacol

    (2006)
  • S.R. Durham et al.

    Grass pollen immunotherapy inhibits allergen-induced infiltration of CD4+ T lymphocytes and eosinophils in the nasal mucosa and increases the number of cells expressing messenger RNA for interferon-gamma

    J Allergy Clin Immunol

    (1996)
  • G. Passalacqua et al.

    Randomised controlled trial of local allergoid immunotherapy on allergic inflammation in mite-induced rhinoconjunctivitis

    Lancet

    (1998)
  • G. Passalacqua et al.

    Clinical and immunologic effects of a rush sublingual immunotherapy to Parietaria species: a double-blind, placebo-controlled trial

    J Allergy Clin Immunol

    (1999)
  • M. Marogna et al.

    Clinical, functional, and immunologic effects of sublingual immunotherapy in birch pollinosis: a 3-year randomized controlled study

    J Allergy Clin Immunol

    (2005)
  • R. Dahl et al.

    Efficacy and safety of sublingual immunotherapy with grass allergen tablets for seasonal allergic rhinoconjunctivitis

    J Allergy Clin Immunol

    (2006)
  • H. Smith et al.

    Randomized controlled trial of high-dose sublingual immunotherapy to treat seasonal allergic rhinitis

    J Allergy Clin Immunol

    (2004)
  • M. Marogna et al.

    Long-lasting effects of sublingual immunotherapy according to its duration: a 15-year prospective study

    J Allergy Clin Immunol

    (2010)
  • E. Novembre et al.

    Coseasonal sublingual immunotherapy reduces the development of asthma in children with allergic rhinoconjunctivitis

    J Allergy Clin Immunol

    (2004)
  • D.R. Wilson et al.

    Sublingual immunotherapy for allergic rhinitis: systematic review and meta-analysis

    Allergy

    (2005)
  • S. Radulovic et al.

    Sublingual immunotherapy for allergic rhinitis

    Cochrane Database Syst Rev

    (2010)
  • M. Fernández-Rivas et al.

    Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract

    Allergy

    (2009)
  • E. Nettis et al.

    Double-blind, placebo-controlled study of sublingual immunotherapy in patients with latex-induced urticaria: a 12-month study

    Br J Dermatol

    (2007)
  • G. Passalacqua et al.

    The safety of allergen specific sublingual immunotherapy

    Curr Drug Saf

    (2007)
  • S.R. Durham et al.

    Long-term clinical efficacy in grass pollen-induced rhinoconjunctivitis after treatment with SQ-standardized grass allergy immunotherapy tablet

    J Allergy Clin Immunol

    (2010)
  • H. Ott et al.

    Efficacy of grass pollen sublingual immunotherapy for three consecutive seasons and after cessation of treatment: the ECRIT study

    Allergy

    (2009)
  • L. Van Overtvelt et al.

    IL-10-inducing adjuvants enhance sublingual immunotherapy efficacy in a murine asthma model

    Int Arch Allergy Immunol

    (2008)
  • N. Saint-Lu et al.

    Targeting the allergen to oral dendritic cells with mucoadhesive chitosan particles enhances tolerance induction

    Allergy

    (2009)
  • J.P. Allam et al.

    Distribution of Langerhans cells and mast cells within the human oral mucosa: new application sites of allergens in sublingual immunotherapy?

    Allergy

    (2008)
  • L. Mascarell et al.

    Mapping of the lingual immune system reveals the presence of both regulatory and effector CD4+ T cells

    Clin Exp Allergy

    (2009)
  • G.W. Scadding 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)
  • M. Bagnasco et al.

    Pharmacokinetics of an allergen and a monomeric allergoid for oromucosal immunotherapy in allergic volunteers

    Clin Exp Allergy

    (2001)
  • J.P. Allam et al.

    Toll-like receptor 4 ligation enforces tolerogenic properties of oral mucosal Langerhans cells

    J Allergy Clin Immunol

    (2008)
  • J.P. Allam 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-beta1 and IL-10-producing properties

    J Allergy Clin Immunol

    (2010)
  • L. Mascarell et al.

    Oral dendritic cells mediate antigen-specific tolerance by stimulating TH1 and regulatory CD4+ T cells

    J Allergy Clin Immunol

    (2008)
  • C. Rask et al.

    Shorter dosing intervals of sublingual immunotherapy lead to more efficacious treatment in a mouse model of allergic inflammation

    Scand J Immunol

    (2010)
  • Cited by (22)

    • Immunotherapy of Allergic Disease

      2019, Clinical Immunology: Principles and Practice
    • Nasal delivery of Japanese cedar pollen Cryj1 by using self-gelling immunostimulatory DNA for effective induction of immune responses in mice

      2015, Journal of Controlled Release
      Citation Excerpt :

      The first is reduced Th2 responses manifested as reduced activation of basophils and eosinophils, and reduced class-switching of B-cells to produce IgE [4,5]. The second is CpG DNA-induced humoral immunity such as competition of allergen-specific IgG and IgA with IgE for allergen binding [6,7]. Previous clinical studies have shown that subcutaneous injection of CpG DNA conjugated with ragweed pollen antigen was effective for preventing ragweed seasonal allergic rhinitis in humans [8,9].

    • Sublingual immunotherapy: World Allergy Organization position paper 2013 update

      2014, World Allergy Organization Journal
      Citation Excerpt :

      These studies provide evidence for long-term disease remission and disease modification consistent with the induction of antigen-specific tolerance. In parallel with these novel clinical data, there have been advances in our understanding of the underlying mechanisms of SLIT that may give rise to putative biomarkers to predict the clinical response [5–8]. The oral cavity is a naturally tolerogenic environment, remaining noninflamed despite being exposed continuously to multiple foreign proteins.

    • Sublingual Immunotherapy for Inhalant Allergens

      2014, Middleton's Allergy: Principles and Practice: Eighth Edition
    • Author response

      2013, Annals of Allergy, Asthma and Immunology
    • Pediatric sublingual immunotherapy efficacy: Evidence analysis, 2009-2012

      2013, Annals of Allergy, Asthma and Immunology
      Citation Excerpt :

      Several studies have reported on the preventive effect of immunotherapy in children with allergic rhinitis (AR) because it appears to reduce the development of new allergic sensitizations and/or new-onset asthma.1,2 Today, clear humoral, cellular, and tissue level changes have been documented with AIT,3–5 and its clinical efficacy leads to economic savings after 6 months of treatment.6 In 2011, the centenary of subcutaneous immunotherapy (SCIT) was celebrated7,8; concurrently, it was 25 years ago that the first double-blind, placebo-controlled (DBPC) trial with SLIT was published.9

    View all citing articles on Scopus

    No conflicts of interest to disclose.

    Stephen Durham has received lecture fees and payments for consultancies from ALK Abello, a manufacturer of allergy vaccines, and research grants from ALK Abello via Imperial College London.

    View full text