Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 1.
Published in final edited form as: Ann Allergy Asthma Immunol. 2016 Aug;117(2):138–142. doi: 10.1016/j.anai.2016.06.027

Basic Science for the Clinician: Mechanisms of sublingual and subcutaneous immunotherapy

Monica G Lawrence 1,3, John W Steinke 1,2,3,*, Larry Borish 1,2,3,4,*
PMCID: PMC4978173  NIHMSID: NIHMS801067  PMID: 27499541

Introduction

Subcutaneous immunotherapy (SCIT) has been in use since 1911 for the treatment of respiratory and venom allergies, and current practice involves administering escalating doses of antigen via one or more subcutaneous injections weekly until a maintenance dose is achieved. Maintenance SCIT is generally continued every 4–8 weeks for a period of 3–5 years and has been shown in multiple studies to provide sustained benefit in reducing clinical symptoms to allergens as well as modulate the immune response to allergens (reviewed in1). Recently, newer routes of IT involving the mucosal immune system have emerged and gained popularity including sublingual immunotherapy (SLIT) and oral immunotherapy (OIT) 2. SLIT is administered daily by holding antigen drops or tablets under the tongue, while OIT involves swallowing escalating daily doses of antigen. Characterizing the mechanism of action of IT has been the focus of intense investigation over the past several decades1 and is important for the practicing clinician to understand as he or she considers the different routes of IT available to administer in clinical practice.

Changes in the allergen-specific T cell response in SCIT and SLIT

Mechanisms of T cell modulation following IT

The conventional theory regarding successful IT, be it SCIT or SLIT, has invoked the induction of regulatory T (Treg) cells, the nomenclature of which we will review briefly here3, 4. Several types of Tregs have been described including thymic-derived and peripherally derived Tregs5, 6. Thymus-derived Treg (tTreg; also referred to as natural (nTreg)) cells are characterized as CD4+CD25+ T cells that constitutively express the transcription factor Foxp3 and have a unique, hypomethylated CpG rich Foxp3 locus79. tTreg cells are produced in the thymus in response to expression of self-antigens and are therefore important in the prevention of autoimmunity. While they constitute 5–10% of peripheral CD4+ T cells, they are unlikely to be involved in tolerance to antigens not presented in the thymus such as allergens. An additional Treg subset has been described that develops in the periphery (pTreg; also referred to as inducible (iTreg))6, 10. These pTreg cells can differentiate from preexisting T effector lymphocytes or circulating naïve T cells and may or may not express Foxp3 and CD25 while possessing suppressive activity. These cells are difficult to identify and study based on using CD25 as a surface marker or Foxp3 as an intracellular marker, as both of these markers can be transiently expressed by CD4+ and CD8+ effector T cells following T cell receptor activation and in response to IL-2 via a positive feedback loop11. A sub-group of pTregs that is primarily gut-derived and generates mucosal tolerance has been referred to as Th3. Reflecting their prominent production of transforming growth factor (TGF)-β, in addition to mediating tolerance, Th3 cells are relevant to inducing secretory IgA production and oral tolerance. Suppression by Tregs occurs through contact inhibition, secretion of IL-10, TGF-β and IL-35 that inhibit cellular activity, surface interactions by negative co-stimulatory molecules such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) or by competition for IL-212, 13.

An alternative mechanism for successful immunotherapy has been proposed that involves a shift in allergen-specific T cells from a T helper 2 (Th2) to a Th1 phenotype14. The recent development of major histocompatibility class (MHC) class II tetramers and epitope-guided mapping has allowed researchers to monitor the allergen-specific T cell responses to IT. The frequency of Fel d 1-specific T cells in allergic individuals prior to IT ranges from 1 in 7,000 to 1 in 30,000, with similar frequencies found for other allergen-specific T cells15. Allergen-specific T cells have also been identified in non-allergic subjects, albeit with lower numbers and decreased avidity of the T cell receptor16.

T cell changes during SCIT

A prominent role for IL-10–producing pTreg cells was first described in studies involving bee venom SCIT3. Induction of peripheral tolerance occurred with the use of either whole bee venom extract or short peptides to phospholipase A2, the major bee venom allergen. The peripheral T cell response demonstrated that both Th2 (IL-4, IL-5 and IL-13) and Th1 (IFN-γ) cytokine production decreased, while IL-10 levels were increased. These IL-10 producing T cells suppressed allergen-specific T cell proliferation and activation and are now recognized as pTregs 17.

Use of inhalant allergens in SCIT, including house dust mite and birch pollen, have confirmed the importance of IL-10 (and TGF-β) production by CD4+CD25+ T cells (pTregs) in the suppression of Th2 and Th1 proliferative responses and cytokine production18. Several studies have also demonstrated new or increased IL-10 production by CD4+ cells in the absence of changes in allergen-induced proliferation, but with decreases in Th2 cytokine production including IL-5 within 10 months of treatment4, 1921. Reasons for differences in T cell responses could include dose of allergen used, different adjuvants added to the allergen and differences related to the allergen itself and the ability to induce an immune response.

What is consistent is that each of these studies has found cells capable of making high levels of IL-10 (±TGF-β) consistent with the pTreg cell type. While the appearance of the IL-10 producing pTregs in bee venom and inhalant allergen SCIT is rapid, within 7 days, full tolerance requires 3–5 years of sustained high-dose treatment. The tolerogenic effect of pTreg cells is evident from studies demonstrating that following 4 years of high dose SCIT, subjects remain non-responsive for at least 3 years after SCIT has been stopped, similar to those who had continued SCIT for the full 7 year period. 22

Using the tetramer-guided approach described earlier to monitor the frequency and characteristics of alder pollen specific CD4+ T cells during the course of IT, led to the observation that two populations of allergen-specific memory T cells could be separated based on the expression of CD27. A terminally differentiated CD27 population has characteristics of classical Th2 cells with high levels of CRTh2 and CCR4 on the cell surface and high intracellular levels of IL-4, IL-5 and IL-13. In contrast, the CD27+ population had high CXCR3, CCR7 and CD7 on the cell surface with high intracellular interferon (IFN)-γ levels, characteristics of Th1 cells, as well as suppressive IL-1023. It was the ratio of CD27 (Th2) to CD27+ (Th1) cells that distinguished allergic from non- atopic individuals and suggested that this could be a biomarker for successful immunotherapy. Successful SCIT was associated with preferential loss of the pathogenic CD27 (Th2) population, while the surviving IL-10 secreting CD27+ T cells maintained a suppressed state and limited the development of new allergen-specific Th2 cells24.

T cell changes during SLIT

In an early mechanistic study of SLIT in humans, peripheral blood mononuclear cells (PBMCs) from patients treated with house dust mite (HDM) SLIT for 3 years produced more IL-10 following mitogen stimulation when compared to patients with untreated rhinitis25. Similar to SCIT, this provided indirect evidence for the immunoregulatory role of IL-10 in SLIT. A smaller mechanistic study examining 9 patients undergoing birch SLIT suggested an early and late phase response to therapy26. By 4 weeks of therapy, there was an increase in circulating pTregs accompanied by an increase in IL-10 and Foxp3 mRNA expression levels and non-specific suppression of antigen-driven lymphocyte proliferation. At the end of 52 weeks of therapy, suppression of proliferation was specific to Bet v 1 (birch pollen) and the levels of circulating pTregs as well as IL-10 returned to baseline. Persistent reduction of IL-4 mRNA and an increase in IFN-γ mRNA was maintained. This study suggests that early in therapy, IL-10 producing pTregs act non-specifically to suppress lymphocyte proliferation in response to antigen, but over time this non-specific response is replaced by antigen-specific tolerance on the backdrop of a Th1 skewed cytokine milieu27. Other studies have confirmed increased pTreg numbers, decreased allergen-specific T cell proliferation, and a shift from a Th2-to-Th1 cytokine milieu following SLIT, accompanied by clinical improvement in symptoms28, 29.

Unique local immunomodulatory effects have also been noted with SLIT. In patients treated with grass pollen SLIT for 12–18 months, the number of Foxp3+ cells in the sublingual epithelium significantly increased in the treatment group compared to the placebo group and non-atopic controls30. One caveat is that these cells may have been recently activated effector cells rather than true regulatory T cells as the number of IL-10 and TGF-β mRNA positive cells did not change following SLIT. In contrast, another study of grass pollen SLIT was able to demonstrate an increased number of IL-10 and TGF-β–producing Treg cells following therapy31.

Bonvalet et al. demonstrated a significant (41.8%) improvement in symptom scores after grass pollen challenge in subjects given grass SLIT for 4 months32. However, they were unable to detect a significant decrease in grass allergen (Phl p 1 and Phl p 5)-specific CD4+ T cells numbers during treatment or 4 months after SLIT was stopped. There was a slight decrease in the number of CD27 or “pro-allergic” Th2 cells32. This raises the possibility that while clinical improvement may be seen as soon as 1 month, parallel immunologic changes involving the shift from a Th2 signature and upregulation of pTreg cells require longer treatment protocols (up to 12 months). Another explanation for the potential failure of immunotherapy is that the epigenetic changes noted in pTregs have been shown to be unstable, i.e. the FoxP3 locus is only transiently hypomethylated 33, 34.

Humoral Immunity in SCIT and SLIT

Changes in the Humoral Response During IT

During the course of IT, allergen-specific IgE measured by ImmunoCAP initially rises above baseline and then gradually decreases over months to years of treatment35, 36. However, the normal seasonal increase in IgE due to natural exposure to the allergen is blunted following IT37. In contrast to ImmunoCAP measurements, when Immuno Solid-phase Allergy Chip (ISAC) is used, there is a decrease in grass allergen component-specific IgE seen during the build-up phase of IT, suggesting that ISAC may offer advantages for monitoring the early effects of IT38.

Driven by IL-10 production, there is an accompanying rise in allergen-specific IgG antibodies, particularly IgG4 antibodies, which are proposed to have a “blocking” effect by competing for allergen and thus inhibiting allergen binding to membrane-bound IgE. This would prevent IgE crosslinking and mast cell and basophil degranulation; however, since mast cells are located on mucosal surfaces that directly contact allergen, it is unclear how an IgG antibody could access the allergen prior to its binding to the mast cell39. Another potential effect of IgG4 is to limit activation of T cells via inhibition of IgE-facilitated antigen presentation (FAP) by FcεRI+ and CD23+ antigen-presenting cells (APC). Lastly, IgG4 may bind to the low affinity inhibitory FcγRIIB, suppressing release of pro-inflammatory mediators40. The primary source of IL-10 is not only Tregs, but also so-called regulatory B cells (Bregs). In a classic paper using beekeepers with frequent venom exposure as a model of bee venom IT, the number of Bregs increased over time with higher IL-10 and IgG4 production by the Bregs. Additionally, IL-10+ Bregs were found at higher frequency in non-allergic beekeepers41.

An increase in TGF-β driven allergen-specific IgA and IgG1 production has been reported in some but not all studies of IT4245. IgA is believed to play a protective role in mucosal immunity, so this mechanism would be more relevant in OIT and SLIT rather than SCIT. As a whole, studies have consistently failed to demonstrate a correlation between the timing or magnitude of the decrease in specific IgE levels and the rise in IgG4 and IgA levels with clinical benefit. Thus, the changes in the humoral compartment are best interpreted as a marker of changes in the overall immune response in systems described elsewhere in this review.

Differences in humoral immunity induced by SCIT versus SLIT

Studies directly comparing SLIT versus SCIT that include measurements not just of clinical efficacy but also of well-defined immunologic markers, are lacking in the literature. A recent randomized placebo-controlled trial comparing SCIT to SLIT for grass pollen allergy found that the initial increase in grass specific IgE was quicker (1 month compared to 1–2 months) and threefold higher in the SLIT group compared with the SCIT group37. SCIT-treated patients demonstrated a higher level of IgE blocking activity than those treated with SLIT (50% versus 27%) as well as increased inhibition of FAP (22.5% versus 17.2%). This effect was also more rapid with SCIT (3 months versus 15 months)37. Dramatic increases in specific IgG4 antibody levels have been reported in multiple studies of patients receiving maintenance therapy with SCIT (30–40 fold) compared to SLIT (3–4 fold)37, 4648.

One possible explanation for the increased humoral immune response noted in SCIT versus SLIT pertains to the level of allergen exposure49. While the total cumulative dose of allergen given sublingually in SLIT is significantly higher than that given subcutaneously in SCIT (anywhere from 5- to 100-fold lower according to various estimates37, 50, the effective dose that is absorbed by the oral mucosa and available for the immune response is postulated to be much lower in SLIT due to the neutralizing effects of saliva. In addition, SCIT protocols often rely upon use of an adjuvant such as aluminum hydroxide (alum) or calcium phosphate, while such adjuvants are not used in sublingual preparations. The use of adjuvants boosts the immune response, a feature frequently taken advantage of in vaccine development.

Antigen Processing and Presentation: Dendritic Cells in SCIT and SLIT

Antigen Processing and Presentation in IT

In large part, the phenotype of the immune response generated following IT administration depends upon which APC is responsible for processing and presenting the allergen. As the most important “professional” APC, it is the dendritic cell (DC) that has the most important role in driving this component of the subsequent immune response and amongst the DC, it is which class of DC, its degree of maturation, and its inherent state of Th1/Th2/Th17 immune deviating “bias” that determines the response. Dendritic cells include myeloid, plasmacytoid, conventional, Langerhans and many others: each having different anatomical locations and unique inherent tendencies to drive distinct effector and tolerogenic immune responses that are likely to be differently engaged in SCIT and SLIT (reviewed in detail elsewhere51).

Antigen Processing and Presentation in SCIT

The location of the immune response ensuing after a maintenance allergy injection, whether it is a primary (spleen), secondary (axillary lymph node) or tertiary (local) lymphoid organ, is certain to influence which APC is responsible for initiating the immune response, the nature of the subsequently engaged T effector cells and, ultimately, the phenotype of the ensuing immune response. Surprisingly little is known regarding the natural history of allergen after it is injected, typically in the subcutaneous tissue in the deltoid region. Some of the allergen drains via local lymphatics into secondary lymphoid organs (e.g., axillary lymph nodes). It is estimated, however, that only a very small fraction of the injected allergen reaches these secondary lymphatics52, 53. Subcutaneously injected allergen is also clearly capable of accessing the circulation either directly through local vasculature or indirectly via the lymphatics and thoracic duct as evinced by the potential for systemic anaphylaxis and the ability to measure allergen in circulation after a subcutaneous injection50, 54. This systemic access suggests that subsequent immune responses could also develop in virtually any lymphoid organ including primary lymphoid organs such as the spleen. Lastly, given the presence of local reactions, including what are often severe, painful reactions that can persist for many days or even weeks after an injection, it is also plausible that T effector immune responses are evolving in tertiary lymphoid organs or developing de novo in the subcutaneous tissue itself. Indeed, it is this surprising lack of understanding of the events occurring following subcutaneous injection of allergen that has led to a desire to develop newer techniques with more predictable responses. These include SLIT, but also the consideration of direct intralymphatic injection of allergen, a mechanism that produces much more predictable tolerogenic processes than SCIT and, shares with SLIT the ability to greatly reduce the risk for anaphylaxis 55.

Antigen Processing and Presentation in SLIT

In contrast to SCIT, somewhat more (or rather, “something”) is known regarding the role of antigen processing pathways in SLIT. The administration of a SL tablet and its being held in place for ~5 minutes before being swallowed largely focuses the allergen to the sublingual tissue. Not being injected and, as such, being less able to directly access vascular structures, sublingually administered allergen is less capable of being measured in the circulation50, 54. This largely eliminates the risk for anaphylaxis, but also focuses the arena of investigation of the immune response to the sublingual mucosa. The DCs involved in SLIT processing are primarily myeloid (mDC) and Langerhans cells (LCs)51, 56, 57. The capacity of these DCs to rapidly capture sublingually administered antigen can be demonstrated by the take-up of biotinylated ovalbumin40, 58. Both sublingual LCs and mDCs are characterized by inherent production of IL-10, IL-12, and TGF-β. As such, the tendency of both of these DC classes is to promote Th1 (via IL-12) and Th3 immunity (via TGF-β and IL-10), but not Th2 responses, along with also promoting IgA-mediated humoral immune responses (via TGF-β) 31. These DCs are also characterized by expression of high affinity IgE receptors (FcεRI) 5860. This expression of FcεRI promotes beneficial immune responses via IgE-mediated FAP.

While reducing the potential for anaphylaxis and arguably focusing the potential phenotypes of developing immune responses to those that can be mediated by mDCs and LCs (i.e. a Th1/Treg response) there is one potential downside to the sublingual administration of allergen. While the systemic availability of SCIT raises the risk for anaphylaxis, it also makes available the entire immune organ as an arena for antigen processing and presentation. In contrast, the immune response to SLIT is limited to the fixed number of DCs present in the sublingual tongue mucosa. One of the surprising observations of SLIT is the absence of compelling dose responses. That is, in numerous studies once an effective dose is achieved no further benefit is observed with higher doses6163. And in at least one study, the addition of numerous other allergens to a fixed dose of timothy grass appeared to reduce its beneficial effects for all allergens in the mixture64. Neither of these limitations, if confirmed, is generally associated with SCIT and may reflect the unlimited capacity for antigen processing and presentation after subcutaneous administration. In contrast, the sublingual surface provides only a relatively fixed number of DCs to process and present allergen, suggesting the possibility of saturation and competitive inhibition for available DCs.

Innate Lymphoid Type 2 cells in SCIT and SLIT

A novel subset of immune cells termed the innate lymphoid type 2 cells (ILC2) has been described and recognized as having central importance in pathogenesis of allergic diseases secondary to their capacity to rapidly produce large amounts of IL-5 and IL-13 in response to “danger” and “alarm” signals (reviewed in 6566). The impact of IT on ILC2 function has only been minimally investigated. However, a recent study demonstrated that ILC2 numbers increased in seasonal allergic rhinitics during the pollen season and that that increase was attenuated among subjects67. The increased numbers of ILC2s were characterized by IL-13 expression 67.

Conclusion

Both SCIT and SLIT are clinically effective in meta-analyses and both induce similar, but not identical changes in the immune response (summarized Table I). What remains to be seen is if the subtle differences that have been noted have relevance to factors such as the persistence of the protective immune response following discontinuation of treatment, or the ideal allergens targeted by each route. It is clear that for the clinician, the last decade represents a true sea change in the field of allergy with the emergence of SLIT and OIT as viable alternatives to SCIT, and that keeping up to date with these changes will be critical to the practice of allergy in the future.

Table I.

Immune Mechanisms of Immunotherapy

Immune Mechanism Demonstrated for SCIT Demonstrated for SLIT
Cellular Immunity Induction of Allergen-Specific pTregs X X
Th2-to-Th1 immune deviation X X
Selective apoptosis of CD27 allergen-specific T cells X
Induction of IL-10, TGF-β X X
Induction of TGF-β+ allergen-specific Th3 cells X
Reduction of ILC2s X
Humoral Immunity Transient rise and long-term inhibition of allergen-specific IgE X X
Rapid decrease in allergen component IgE X
Increase in allergen-specific IgA and IgG4 X X
Inhibition of facilitated antigen presentation (FAP) X

Acknowledgments

Supported by NIH grants R01-AI057438, R56-AI120055, U01-AI100799, and UO1-AI123337

Footnotes

Trial registration: Not applicable

All authors report no conflicts of interest

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Bibliography

  • 1.Finegold I, Dockhorn RJ, Ein D, Dolen WK, Oppenheimer J, Potter LH. Immunotherapy throughout the decades: from Noon to now. Ann Allergy Asthma Immunol. 2010;105:328–336. doi: 10.1016/j.anai.2010.08.008. quiz 337, 358. [DOI] [PubMed] [Google Scholar]
  • 2.Jay DC, Nadeau KC. Immune mechanisms of sublingual immunotherapy. Curr Allergy Asthma Rep. 2014;14:473. doi: 10.1007/s11882-014-0473-1. [DOI] [PubMed] [Google Scholar]
  • 3.Akdis CA, Blesken T, Akdis M, Wuthrich B, Blaser K. The role of IL-10 in specific immunotherapy. Journal of Clinical Investigation. 1998;102:98–106. doi: 10.1172/JCI2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Francis JN, Till SJ, Durham SR. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. The Journal of allergy and clinical immunology. 2003;111:1255–1261. doi: 10.1067/mai.2003.1570. [DOI] [PubMed] [Google Scholar]
  • 5.Sakaguchi S. Regulatory T cells: key controllers of immunologic self-tolerance. Cell. 2000;101:455–458. doi: 10.1016/s0092-8674(00)80856-9. [DOI] [PubMed] [Google Scholar]
  • 6.Abbas AK, Benoist C, Bluestone JA, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol. 2013;14:307–308. doi: 10.1038/ni.2554. [DOI] [PubMed] [Google Scholar]
  • 7.Hori S, Sakaguchi S. Foxp3: a critical regulator of the development and function of regulatory T cells. Microbes Infect. 2004;6:745–751. doi: 10.1016/j.micinf.2004.02.020. [DOI] [PubMed] [Google Scholar]
  • 8.Attridge K, Walker LS. Homeostasis and function of regulatory T cells (Tregs) in vivo: lessons from TCR-transgenic Tregs. Immunol Rev. 2014;259:23–39. doi: 10.1111/imr.12165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Swamy RS, Reshamwala N, Hunter T, et al. Epigenetic modifications and improved regulatory T-cell function in subjects undergoing dual sublingual immunotherapy. The Journal of allergy and clinical immunology. 2012;130:215–224. e217. doi: 10.1016/j.jaci.2012.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Akdis CA, Blaser K. IL-10-induced anergy in peripheral T cell and reactivation by microenviromental cytokines: two key steps in specific immunotherapy. FASEB Journal. 1999;13:603–609. doi: 10.1096/fasebj.13.6.603. [DOI] [PubMed] [Google Scholar]
  • 11.Kmieciak M, Gowda M, Graham L, et al. Human T cells express CD25 and Foxp3 upon activation and exhibit effector/memory phenotypes without any regulatory/suppressor function. J Transl Med. 2009;7:89. doi: 10.1186/1479-5876-7-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8:523–532. doi: 10.1038/nri2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang YM, Ghali J, Zhang GY, et al. Development and function of Foxp3(+) regulatory T cells. Nephrology (Carlton) 2016;21:81–85. doi: 10.1111/nep.12652. [DOI] [PubMed] [Google Scholar]
  • 14.Nepom GT. MHC class II tetramers. J Immunol. 2012;188:2477–2482. doi: 10.4049/jimmunol.1102398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kwok WW, Roti M, Delong JH, et al. Direct ex vivo analysis of allergen-specific CD4+ T cells. The Journal of allergy and clinical immunology. 2010;125:1407–1409. e1401. doi: 10.1016/j.jaci.2010.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kinnunen T, Nieminen A, Kwok WW, et al. Allergen-specific naive and memory CD4+ T cells exhibit functional and phenotypic differences between individuals with or without allergy. Eur J Immunol. 2010;40:2460–2469. doi: 10.1002/eji.201040328. [DOI] [PubMed] [Google Scholar]
  • 17.Muller U, Akdis CA, Fricker M, et al. Successful immunotherapy with T-cell epitope peptides of bee venom phospholipase A2 induces specific T-cell anergy in patients allergic to bee venom. The Journal of allergy and clinical immunology. 1998;101:747–754. doi: 10.1016/S0091-6749(98)70402-6. [DOI] [PubMed] [Google Scholar]
  • 18.Jutel M, Akdis M, Budak F, et al. IL-10 and TGF-b cooperate in the regulatory T cell response to mucosal allergens in normal immunity and specific immunotherapy. European Journal of Immunolgy. 2003;33:1205–1214. doi: 10.1002/eji.200322919. [DOI] [PubMed] [Google Scholar]
  • 19.Schulten V, Tripple V, Aasbjerg K, et al. Distinct modulation of allergic T cell responses by subcutaneous vs. sublingual allergen-specific immunotherapy. Clin Exp Allergy. 2016;46:439–448. doi: 10.1111/cea.12653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tian M, Wang Y, Lu Y, Jiang YH, Zhao DY. Effects of sublingual immunotherapy for Dermatophagoides farinae on Th17 cells and CD4(+) CD25(+) regulatory T cells in peripheral blood of children with allergic asthma. Int Forum Allergy Rhinol. 2014;4:371–375. doi: 10.1002/alr.21305. [DOI] [PubMed] [Google Scholar]
  • 21.Li CW, Lu HG, Chen de H, Lin ZB, Wang de Y, Li TY. In vivo and in vitro studies of Th17 response to specific immunotherapy in house dust mite-induced allergic rhinitis patients. PLoS One. 2014;9:e91950. doi: 10.1371/journal.pone.0091950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Durham SR, Walker SM, Varga EM, et al. Long-term clinical efficacy of grass-pollen immunotherapy. N Engl J Med. 1999;341:468–475. doi: 10.1056/NEJM199908123410702. [DOI] [PubMed] [Google Scholar]
  • 23.Wambre E, DeLong JH, James EA, LaFond RE, Robinson D, Kwok WW. Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy. The Journal of allergy and clinical immunology. 2012;129:544–551. 551 e541–547. doi: 10.1016/j.jaci.2011.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wambre E. Effect of allergen-specific immunotherapy on CD4+ T cells. Curr Opin Allergy Clin Immunol. 2015;15:581–587. doi: 10.1097/ACI.0000000000000216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ciprandi G, Fenoglio D, Cirillo I, et al. Induction of interleukin 10 by sublingual immunotherapy for house dust mites: a preliminary report. Ann Allergy Asthma Immunol. 2005;95:38–44. doi: 10.1016/S1081-1206(10)61186-6. [DOI] [PubMed] [Google Scholar]
  • 26.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. The Journal of allergy and clinical immunology. 2007;120:707–713. doi: 10.1016/j.jaci.2007.06.013. [DOI] [PubMed] [Google Scholar]
  • 27.Frew AJ. How does sublingual immunotherapy work? The Journal of allergy and clinical immunology. 2007;120:533–536. doi: 10.1016/j.jaci.2007.08.003. [DOI] [PubMed] [Google Scholar]
  • 28.O’Hehir RE, Gardner LM, de Leon MP, et al. House dust mite sublingual immunotherapy: the role for transforming growth factor-beta and functional regulatory T cells. American journal of respiratory and critical care medicine. 2009;180:936–947. doi: 10.1164/rccm.200905-0686OC. [DOI] [PubMed] [Google Scholar]
  • 29.Potter PC, Baker S, Fenemore B, Nurse B. Clinical and cytokine responses to house dust mite sublingual immunotherapy. Ann Allergy Asthma Immunol. 2015;114:327–334. doi: 10.1016/j.anai.2014.12.015. [DOI] [PubMed] [Google Scholar]
  • 30.Scadding GW, Shamji MH, Jacobson MR, 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. doi: 10.1111/j.1365-2222.2010.03462.x. [DOI] [PubMed] [Google Scholar]
  • 31.Allam JP, Wurtzen PA, Reinartz M, 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. The Journal of allergy and clinical immunology. 2010;126:638–645. e631. doi: 10.1016/j.jaci.2010.04.039. [DOI] [PubMed] [Google Scholar]
  • 32.Bonvalet M, Moussu H, Wambre E, 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;42:1745–1755. doi: 10.1111/cea.12015. [DOI] [PubMed] [Google Scholar]
  • 33.Okada M, Hibino S, Someya K, Yoshmura A. Regulation of regulatory T cells: epigenetics and plasticity. Adv Immunol. 2014;124:249–273. doi: 10.1016/B978-0-12-800147-9.00008-X. [DOI] [PubMed] [Google Scholar]
  • 34.Syed A, Garcia MA, Lyu SC, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3) The Journal of allergy and clinical immunology. 2014;133:500–510. doi: 10.1016/j.jaci.2013.12.1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Van Ree R, Van Leeuwen WA, Dieges PH, et al. Measurement of IgE antibodies against purified grass pollen allergens (Lol p 1, 2, 3 and 5) during immunotherapy. Clin Exp Allergy. 1997;27:68–74. doi: 10.1046/j.1365-2222.1997.d01-416.x. [DOI] [PubMed] [Google Scholar]
  • 36.Creticos PS, Van Metre TE, Mardiney MR, Rosenberg GL, Norman PS, Adkinson NF., Jr Dose response of IgE and IgG antibodies during ragweed immunotherapy. The Journal of allergy and clinical immunology. 1984;73:94–104. doi: 10.1016/0091-6749(84)90490-1. [DOI] [PubMed] [Google Scholar]
  • 37.Aasbjerg K, Backer V, Lund G, et al. Immunological comparison of allergen immunotherapy tablet treatment and subcutaneous immunotherapy against grass allergy. Clin Exp Allergy. 2014;44:417–428. doi: 10.1111/cea.12241. [DOI] [PubMed] [Google Scholar]
  • 38.Schmid JM, Wurtzen PA, Dahl R, Hoffmann HJ. Pretreatment IgE sensitization patterns determine the molecular profile of the IgG4 response during updosing of subcutaneous immunotherapy with timothy grass pollen extract. The Journal of allergy and clinical immunology. 2016;137:562–570. doi: 10.1016/j.jaci.2015.05.023. [DOI] [PubMed] [Google Scholar]
  • 39.Frew AJ. Allergen immunotherapy. The Journal of allergy and clinical immunology. 2010;125:S306–313. doi: 10.1016/j.jaci.2009.10.064. [DOI] [PubMed] [Google Scholar]
  • 40.Moingeon P, Batard T, Fadel R, Frati F, Sieber J, Van Overtvelt L. Immune mechanisms of allergen-specific sublingual immunotherapy. Allergy. 2006;61:151–165. doi: 10.1111/j.1398-9995.2006.01002.x. [DOI] [PubMed] [Google Scholar]
  • 41.van de Veen W, Stanic B, Yaman G, et al. IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. The Journal of allergy and clinical immunology. 2013;131:1204–1212. doi: 10.1016/j.jaci.2013.01.014. [DOI] [PubMed] [Google Scholar]
  • 42.Taudorf E, Laursen L, Lanner A, Bjorksten B, Dreborg S, Weeke B. Specific IgE, IgG, and IgA antibody response to oral immunotherapy in birch pollinosis. The Journal of allergy and clinical immunology. 1989;83:589–594. doi: 10.1016/0091-6749(89)90070-5. [DOI] [PubMed] [Google Scholar]
  • 43.Leonard SA, Martos G, Wang W, Nowak-Wegrzyn A, Berin MC. Oral immunotherapy induces local protective mechanisms in the gastrointestinal mucosa. The Journal of allergy and clinical immunology. 2012;129:1579–1587. e1571. doi: 10.1016/j.jaci.2012.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Akdis CA, Barlan IB, Bahceciler N, Akdis M. Immunological mechanisms of sublingual immunotherapy. Allergy. 2006;61(Suppl 81):11–14. doi: 10.1111/j.1398-9995.2006.01159.x. [DOI] [PubMed] [Google Scholar]
  • 45.Taudorf E, Moller C, Russell MW. Secretory IgA response in oral immunotherapy. Investigation in birch pollinosis. Allergy. 1994;49:760–765. doi: 10.1111/j.1398-9995.1994.tb02099.x. [DOI] [PubMed] [Google Scholar]
  • 46.Lima MT, Wilson D, Pitkin L, et al. Grass pollen sublingual immunotherapy for seasonal rhinoconjunctivitis: a randomized controlled trial. Clin Exp Allergy. 2002;32:507–514. doi: 10.1046/j.0954-7894.2002.01327.x. [DOI] [PubMed] [Google Scholar]
  • 47.Till S. Mechanisms of immunotherapy and surrogate markers. Allergy. 2011;66(Suppl 95):25–27. doi: 10.1111/j.1398-9995.2011.02627.x. [DOI] [PubMed] [Google Scholar]
  • 48.James LK, Shamji MH, Walker SM, et al. Long-term tolerance after allergen immunotherapy is accompanied by selective persistence of blocking antibodies. The Journal of allergy and clinical immunology. 2011;127:509–516. e501–505. doi: 10.1016/j.jaci.2010.12.1080. [DOI] [PubMed] [Google Scholar]
  • 49.Bohle B. Immune mechanisms of SCIT and SLIT: facing possible differences? Clin Exp Allergy. 2014;44:304–306. doi: 10.1111/cea.12263. [DOI] [PubMed] [Google Scholar]
  • 50.Moingeon P. Update on immune mechanisms associated with sublingual immunotherapy: practical implications for the clinician. J Allergy Clin Immunol Pract. 2013;1:228–241. doi: 10.1016/j.jaip.2013.03.013. [DOI] [PubMed] [Google Scholar]
  • 51.Liu K, Nussenzweig MC. Origin and development of dendritic cells. Immunol Rev. 2010;234:45–54. doi: 10.1111/j.0105-2896.2009.00879.x. [DOI] [PubMed] [Google Scholar]
  • 52.Senti G, Johansen P, Kundig TM. Intralymphatic immunotherapy. Curr Opin Allergy Clin Immunol. 2009;9:537–543. doi: 10.1097/ACI.0b013e3283310ff7. [DOI] [PubMed] [Google Scholar]
  • 53.Martinez-Gomez JM, Johansen P, Erdmann I, Senti G, Crameri R, Kundig TM. Intralymphatic injections as a new administration route for allergen-specific immunotherapy. Int Arch Allergy Immunol. 2009;150:59–65. doi: 10.1159/000210381. [DOI] [PubMed] [Google Scholar]
  • 54.Calderon MA, Simons FE, Malling HJ, Lockey RF, Moingeon P, Demoly P. Sublingual allergen immunotherapy: mode of action and its relationship with the safety profile. Allergy. 2012;67:302–311. doi: 10.1111/j.1398-9995.2011.02761.x. [DOI] [PubMed] [Google Scholar]
  • 55.Hylander T, Latif L, Petersson-Westin U, Cardell LO. Intralymphatic allergen-specific immunotherapy: an effective and safe alternative treatment route for pollen-induced allergic rhinitis. The Journal of allergy and clinical immunology. 2013;131:412–420. doi: 10.1016/j.jaci.2012.10.056. [DOI] [PubMed] [Google Scholar]
  • 56.Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Smarr CB, Bryce PJ, Miller SD. Antigen-specific tolerance in immunotherapy of th2-associated allergic diseases. Critical reviews in immunology. 2013;33:389–414. doi: 10.1615/critrevimmunol.2013007046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mascarell L, Lombardi V, Louise A, et al. Oral dendritic cells mediate antigen-specific tolerance by stimulating TH1 and regulatory CD4+ T cells. The Journal of allergy and clinical immunology. 2008;122:603–609. e605. doi: 10.1016/j.jaci.2008.06.034. [DOI] [PubMed] [Google Scholar]
  • 59.Mascarell L, Saint-Lu N, Moussu H, et al. Oral macrophage-like cells play a key role in tolerance induction following sublingual immunotherapy of asthmatic mice. Mucosal Immunol. 2011;4:638–647. doi: 10.1038/mi.2011.28. [DOI] [PubMed] [Google Scholar]
  • 60.Allam JP, Novak N, Fuchs C, et al. Characterization of dendritic cells from human oral mucosa: a new Langerhans’ cell type with high constitutive FcepsilonRI expression. The Journal of allergy and clinical immunology. 2003;112:141–148. doi: 10.1067/mai.2003.1607. [DOI] [PubMed] [Google Scholar]
  • 61.Didier A, Malling HJ, Worm M, et al. Optimal dose, efficacy, and safety of once-daily sublingual immunotherapy with a 5-grass pollen tablet for seasonal allergic rhinitis. The Journal of allergy and clinical immunology. 2007;120:1338–1345. doi: 10.1016/j.jaci.2007.07.046. [DOI] [PubMed] [Google Scholar]
  • 62.Nolte H, Hebert J, Berman G, et al. Randomized controlled trial of ragweed allergy immunotherapy tablet efficacy and safety in North American adults. Ann Allergy Asthma Immunol. 2013;110:450–456. e454. doi: 10.1016/j.anai.2013.03.013. [DOI] [PubMed] [Google Scholar]
  • 63.Bergmann KC, Demoly P, Worm M, et al. Efficacy and safety of sublingual tablets of house dust mite allergen extracts in adults with allergic rhinitis. The Journal of allergy and clinical immunology. 2014;133:1608–1614. e1606. doi: 10.1016/j.jaci.2013.11.012. [DOI] [PubMed] [Google Scholar]
  • 64.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. The Journal of allergy and clinical immunology. 2009;124:150–156. e151–155. doi: 10.1016/j.jaci.2009.04.037. [DOI] [PubMed] [Google Scholar]
  • 65.Neill DR, Wong SH, Bellosi A, et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 2010;464:1367–1370. doi: 10.1038/nature08900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chang JE, Doherty TA, Baum R, Brodie D. Prostglandin D2 regulates human type 2 innate lymphoid cell chemotaxis. The Journal of allergy and clinical immunology. 2014;133:899–901. e3. doi: 10.1016/j.jaci.2013.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lao-Araya M, Steveling E, Scadding GW, Durham SR, Shamji MH. Seasonal inceases in peripheral innate lymphoid type 2 cells are inhibited by subcutaneous grass pollen immunotherapy. The Journal of allergy and clinical immunology. 2014;134:1193–1195. e4. doi: 10.1016/j.jaci.2014.07.029. [DOI] [PubMed] [Google Scholar]
  • 68.Doherty TA, Scott D, Walford HH, et al. Allergen challenge in allergic rhinitis rapidly induces peripheral blood type 2 innate lymphoid cells that express CD84. The Journal of allergy and clinical immunology. 2014;123:1203–1205. e7. doi: 10.1016/j.jaci.2013.12.1086. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES