Abstract
Allergic rhinitis is an IgE-mediated inflammation that remains a clinical challenge, affecting 40% of the UK population with a wide range of severity from nasal discomfort to life-threatening anaphylaxis. It can be managed by pharmacotherapeutics and in selected patients by allergen immunotherapy (AIT), which provides long-term clinical efficacy, especially during peak allergy season. However, there are no definitive biomarkers for AIT efficacy. Here, we aim to summarize the key adaptive, innate, humoral, and metabolic advances in biomarker identification in response to AIT. Mechanisms of efficacy consist of an immune deviation towards TH1-secreting IFN-γ, as well as an induction of IL10+ cTFR and TREG have been observed. TH2 cells undergo exhaustion after AIT due to chronic allergen exposure and correlates with the exhaustion markers PD-1, CTLA-4, TIGIT, and LAG3. IL10+ DCREG expressing C1Q and STAB are induced. KLRG1+ IL10+ ILC2 were shown to be induced in AIT in correlation with efficacy. BREG cells secreting IL-10, IL-35, and TGF-β are induced. Blocking antibodies IgG, IgA, and IgG4 are increased during AIT; whereas inflammatory metabolites, such as eicosanoids, are reduced. There are multiple promising biomarkers for AIT currently being evaluated. A panomic approach is essential to better understand cellular, molecular mechanisms and their correlation with clinical outcomes. Identification of predictive biomarkers of AIT efficacy will hugely impact current practice allowing physicians to select eligible patients that are likely to respond to treatment as well as improve patients’ compliance to complete the course of treatment.
Keywords: allergen immunotherapy, biomarkers, T cells, metabolomics
Introduction
Allergic rhinitis (AR) is an IgE-mediated inflammation of the nasal mucosa triggered by aeroallergens. Symptoms involve rhinorrhea, nasal obstruction, and epiphora [1]. Seasonal AR (SAR) is identified due to onset of symptoms in conjunction with seasonal pollen production, peaking during spring, summer, or fall months [2]. SAR remains a major clinical challenge affecting 10 – 30% of the worldwide population, and generally results in a deteriorated quality of life [3, 4]. Apart from pharmacotherapy, allergen immunotherapy (AIT) remains a key strategy for long-term resolution of symptoms, though long-term clinical benefit is only achieved following treatment of 3 years or longer [5]. For this reason, AIT poses a lot of challenges economically due to the high cost and personally to the patients due to long-term the clinical regimen required to achieve sustained response, resulting in low patient compliance. Biomarkers that allow prediction of AIT efficacy is a huge unmet need in clinical practice and will allow physicians to select eligible patients that are likely to respond to treatment as well as improve patients’ compliance to complete the course of treatment. However, there is currently a lack of validated biomarkers to predict efficacy of AIT. The aim of this review is to outline the key developments in the cellular and metabolite biomarkers as indicative models of effective therapy.
Allergic rhinitis
The early phase of allergic responses in sensitized individuals occurs within minutes and lasts for 3 hours. Offending allergens cross-link IgE on the surface of basophils and mast cells, resulting in the degranulation and release of pro-inflammatory mediators (histamines, leukotrienes, and prostaglandins) [6]. In the late allergic phase, typically within 4 – 12 hours, eosinophils, basophils, T cells, and monocytes infiltrate the airway mucosa, resulting in tissue edema and persistent congestion. Cytokines released by mast cells, such as tumor necrosis factor (TNF), promote pro-inflammatory signaling pathways such as nuclear factor-kappa B (NF-kB) activation and endothelial-leukocyte adhesion molecules such as E-selectin, intracellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) [7, 8, 9]. Interleukin (IL)-4 and IL-5, secreted by TH2 cells, induce eosinophil recruitment and differentiation, whilst IL-9, secreted by TH9 cells, promotes mast cell production and localization [9, 10]. The epithelial barrier also produces RANTES (CCL-5), eotaxin, and thymus- and activation-regulated chemokine (TARC/CCL-17), promoting the tissue localization of granulocytes and T cells [11]. These sustained inflammatory reactions are hallmarks of a late-phase response (Figure 1). Avoiding allergens ablates symptoms, but it is not always feasible. Allergen exposure can be avoided by limiting outdoor interaction during peak pollen seasons. However, a multi-pronged approach involving use of non-sedative second generation oral antihistamines and intra-nasal corticosteroids are more reliable for mild to moderate symptoms [12]. Although widely used, pharmacotherapy provides only temporary relief, while training adaptive immune responses provides superior long-term relief for SAR. Moreover, there is a certain population of individuals who are non-responsive to even high doses of pharmacotherapy. AIT aims to shift the inflammatory profile towards a long-lasting tolerance by inducing key regulatory cells within the immune system.
Figure 1. Phases of immune responses after allergen exposure. Early responses in sensitized individuals involve the crosslinking of FcεRI-bound IgE on the surface of mast cells and innate responses, resulting in the release of inflammatory mediators. Late-phase responses involve the infiltration of leukocytes and a general pro-inflammatory state in the local tissue. Created with www.biorender.com.

Allergen immunotherapy
AIT was first performed using grass pollen extracts in patients with SAR before pollen season [13]. It involves the repeated subcutaneous (SCIT) or sublingual (SLIT) administration of high doses of allergens over 3 – 5 years [14]. Both confer long-term clinical benefit and tolerance even after end of treatment. Although AIT is the only disease-modifying treatment for IgE-mediated allergies, there are some limitations, such as associated side effects, poor compliance, and lack of efficacy in non-responders. New approaches to AIT with the aim to enhance safety whilst maintaining or increasing efficacy are currently being researched. A correlation between AIT efficacy and the induction of allergen-neutralizing antibodies as a key biomarker in tolerance induction has been described [15]. The mechanism of action involves the capturing of allergens on mucosal surfaces, inhibiting high-affinity IgE-receptor (FcεRI) activation, preventing low-affinity IgE-receptor (FcεRII/CD23)-mediated antigen presentation, and inhibiting allergic inflammation. On the cellular side, tolerance involves multiple phases and the complex interplay of the innate and adaptive compartments of the immune system, inclusive of effector T cells, dendritic cells (DCs), innate lymphoid cells (ILCs) and B cells.
T-cell biomarkers indicating effective AIT
Immune tolerance following SCIT and SLIT involves the deletion or anergy of effector cells, immune deviation to a TH1 response, and induction of TREG cells (Figure 2). After 2 years of AIT, TH2-cell responses were inhibited and a lower level of IL4 mRNA was detected in the nasal mucosa following allergen challenge, in conjunction with a lower population of CRTH2+CCR4+CD27-CD161+ allergen-specific TH2A cells [16]. Higher levels of nasal IFN-γ resulting in a reduction of the IL-5/IFN-γ ratio upon allergen exposure was observed following SCIT [17]. Another study also demonstrated time course induction of IFN-γ, confirming the immune deviation towards TH1 responses [18]. TH1 responses are generally thought to be induced following apoptosis of TH2 cells. Cells obtained from AIT-treated grass pollen-allergic patients showed an increased number of IL-4+ cells undergoing apoptosis [19]. Additionally, TH1 cells were shown to have increased persistence through Bcl-2, an anti-apoptotic protein [20]. Therefore, a persistent TH1-cell population may be a biomarker of tolerance in AIT.
Figure 2. T-cell biomarkers in response to allergen immunotherapy. Chronic exposure to allergen results in a shift towards to TH1 immune state and deletion of TH2 cells, induction of regulatory T cells and TFR, and the exhaustion of effector T cells. Created with www.biorender.com.

TFH cells expressing CXCR5 and PD-1 have been identified as mediators in inducing tolerance [21]. TFH have now emerged to be an IL-4- and IL-21-producing subset [22]. An impaired TFH and TFR cell ratio was observed in patients with AR. TFH cells in allergic patients have enhanced capacity to induce IgE production compared to healthy controls, whilst TFR have diminished suppressive action [23]. Circulating TFH (cTFH) cells were defined as a distinct subset of T cells from TH2 and TH2A cells lacking BCL-6 expression, efficient in secreting both IL-4 and IL-21 [24]. cTFH cells were elevated in grass pollen-allergic patients compared to non-atopic controls and were lower following both SCIT and SLIT. In contrast, cTFR and IL-10+ cTFH cells were induced after SCIT and SLIT [24].
TREGS are considered essential regulatory cells secreting IL-10 and TGF-β. Both natural (nTREG, FOXP3+) and induced (iTREG, CD49b+ IL-10+) TREG cells have comparable suppressive function. They are characterized by low CD127 and high CD25 expression. Induction of TREG subsets is commonly associated with immune tolerance after immunotherapy in allergic patients [25]. Multiple studies have shown correlation of TREG frequency with SCIT and SLIT compared to a placebo [26, 27]. Increases in FOXP3 and IL-10 expression have been also observed. Additionally, CpG methylation within the FOXP3 regions following SLIT was reduced along with an increase in FOXP3 transcript expression [28]. Regulatory cytokines such as IL-10 and TGF-β promote T-cell exhaustion [29]. The proportion of PD-1+ CD4+ T cells was found to be increased following AIT treatment [30]. The transcriptional profile of CD4+ T cells following AIT demonstrated in another study showed that TREG expressing c-MAF, NFIL3, LAG-3, TIGIT, PD-1, and TIM-3 were key markers of exhaustion [31]. Recent studies stimulating allergen specific CD4+ T cells showed a strong upregulation of PD-1, LAG-3, and CTLA4 upon in vitro stimulation with sensitizing allergen. The blocking of PD-1 re-invigorated T-cell activity by enhancing cytokine production in both allergic and non-allergic individuals [32]. AIT also resulted in the deletion of CD4+CD27- T cells, where chronic exposure to high doses of allergens causes terminal differentiation of TH2 cells resulting in cell exhaustion [33]. Although these findings support T-cell exhaustion as a hallmark of long-term tolerance, more studies are warranted to identify their physiological role. T cells and other cellular biomarkers could be analyzed by isolation from whole blood samples.
Innate-cell biomarkers for effective treatment
Dendritic cells phagocyte allergen fragments and process them for MHC Class II presentation, resulting in induction of both inflammatory and regulatory cells. AIT induces regulatory DCs (DCREG), which have markers such as CQ1 and STAB1 [34, 35] (Figure 3). Moreover, DCREG secrete IL-12, IL-27, and IL-10 whilst downregulating the expression of CD86, a co-stimulatory receptor that enhances inflammation [36]. ILC2s play key roles in SAR, and they mainly produce type 2 cytokines such as IL-5 and IL-13. SCIT treatment has been shown to lower the levels of ILC2 in both grass pollen and house dust mites (HDM) SCIT. Another novel ILC counterpart that produces IL-10 was also shown to be induced in AIT (Figure 3). Surprisingly, both SCIT and SLIT were shown to result in the induction of ILC2s that can produce IL-10 in vitro following stimulation of IL-2, IL-7, IL-33, and retinoic acid (RA) [37]. These observations show that ILCs play key roles in training immunity during AIT within the innate compartment.
Figure 3. Innate responses towards effective allergen immunotherapy involve the induction of DCREG and IL10-secreting ILC2s. Created with www.biorender.com.

B-cell biomarkers indicating effective AIT
BREGS exert negative immunoregulatory roles through the production of different cytokines such as IL-10, IL-35, TGF-β, and through cell contact mechanisms [38] (Figure 4). B-cell activation is required for suppressive action by the activation of toll-like receptors, BCR signaling, and co-stimulation via CD40/CD40L and CD80/CD86. Inflammatory cytokines also activate BREGS via STAT3. Grass pollen and HDM AIT result in elevated levels of IgA- and IgG4-expressing B cells, plasmablasts, and IL-10+ BREGS, which correlated with improvement of clinical symptoms throughout AIT [39].
Figure 4. B-cell and humoral responses to allergen immunotherapy. Chronic allergen exposure results in the induction of BREG that secret IL-10, IL-35, and TGF-β. The development of B cells producing blocking antibodies IgA and IgG are also positive indicators of successful therapy. Created with www.biorender.com.

Ig responses indicative of effective AIT
AIT success requires the immunomodulation of both cellular and humoral responses. In AIT, the induction of allergen-specific IgG, IgG4, and IgA, which have IgE-blocking activity, has been well characterized [40], and their upregulation seems to be in line with clinical symptoms [41]. The main mechanism involves competitively inhibiting IgE for allergen binding, resulting in the reduction of IgE-induced FcεRI activation on mast cells, thus reducing degranulation and release of inflammatory mediators [42]. Additionally, binding of allergen-IgE complexes to low-affinity IgE receptors on B cells is inhibited, resulting in diminished IgE-facilitated presentation to T cells [40] (Figure 4). The IgE-FAB assay showed that blocking antibody activity following SCIT was time- and dose-dependent, peaking between 3 and 6 months [43]. The GRASS trial revealed that SCIT or SLIT is associated with IgG- or IgA-blocking antibodies, respectively [15]. Humoral biomarkers could be measured both systemically or locally in serum and nasal fluids; and they could be useful to predict local responses characteristic of early AIT response [42].
Epigenetics and metabolomics as indicators of effective AIT
Omics sciences have been recently used in an attempt to find biomarkers that could be indicative of an effective response to AIT, and to better determine the underlying mechanisms that lead to AIT success, such as the desensitization of effector cells or the shifting towards a regulatory immune response. This high-throughput approach of working with large datasets with genomics, microbiomics, transcriptomics, proteomics, and metabolomics could significantly facilitate the discover of new biomarkers [44]. Moreover, omics biomarkers can be measured in a wide range of matrices, including serum, nasal fluid, or exhaled breath condensates [45]. Thanks to omics, key factors to a successful AIT treatment, like an appropriate characterization of allergen extracts or a deep profiling of the patients’ IgE reactivity have been discovered [46, 47, 48]. AIT has shown to induce several epigenetic changes in T cells, such as the induction of FOXP3 [28]; and also in monocytes and DCs, including the downregulation of EZH2 gene in DCs [49] and a tolerogenic reprogramming of monocyte and DCs [50].
Regarding metabolomics, there are currently several proposed biomarkers that could predict response to AIT. It has been recently suggested that the sensitization of the patient (either to one or multiple allergens) is relevant to predict their response to AIT [51]. Moreover, a decrease in eicosanoids has been reported after treatment with SCIT for allergy and allergic asthma [52, 53]. Eicosanoids are known to enhance inflammation, with its precursor, the arachidonic acid pathway being increased in severe, uncontrolled allergic asthma [54]. One such evidence includes a study by Xie et al. [55] which demonstrated twelve biomarkers that were found to be different between responsive and unresponsive patients prior to the treatment with SLIT. These included arachidonic acid, sphingosine, or L-phenylalanine, all of which correlated with altered energetic and inflammatory pathways and the arginine and proline metabolism [55]. L-tyrosine was also found to have an opposite trend between patients with a good and a bad response to SCIT treatment (being decreased after successful AIT and increased for patients for whom it was ineffective); and nitric oxide related pathways (such as arginine and proline metabolism, tyrosine metabolism and nitrogen metabolism) were altered during AIT [56] (Figure 5).
Figure 5. Pathways that have been found altered in allergen immunotherapy. Nitric Oxid-related metabolites, along with eicosanoid metabolism, could serve as biomarkers to find responder patients or assess the success of the treatment. Created with www.biorender.com.

Conclusion
Mechanistic studies have allowed a more holistic understanding of the underlying pathways induced by AIT. The efficacy is associated with the inhibition of inflammatory responses and the induction of markers in both adaptive and innate immune systems. Additional studies are required to elucidate the molecular and epigenetic changes and differences in different modes of AIT. Ultimately, a panomic approach is required to uncover full mechanisms which may prove instrumental to develop new targeted therapies.
Funding
None.
Conflict of interest
M.H. Shamji reports grants from Regeneron, Merck, ANGANY Inc, Allergy Therapeutics, and Immune Tolerance Network; reports personal fees from Allergopharma; and reports grants and personal fees from ALK, Allergy Therapeutics, and ANGANY Inc.
References
- 1. Bousquet J Demoly P Michel FB Specific immunotherapy in rhinitis and asthma. Ann Allergy Asthma Immunol. 2001; 87: 38–42. [DOI] [PubMed] [Google Scholar]
- 2. Bousquet J Schünemann HJ Togias A Bachert C Erhola M Hellings PW Klimek L Pfaar O Wallace D Ansotegui I Agache I Bedbrook A Bergmann KC Bewick M Bonniaud P Bosnic-Anticevich S Bossé I Bouchard J Boulet LP Brozek J Next-generation Allergic Rhinitis and Its Impact on Asthma (ARIA) guidelines for allergic rhinitis based on Grading of Recommendations Assessment, Development and Evaluation (GRADE) and real-world evidence. J Allergy Clin Immunol. 2020; 145: 70–80. [DOI] [PubMed] [Google Scholar]
- 3. Passali D Cingi C Staffa P Passali F Muluk NB Bellussi ML The International Study of the Allergic Rhinitis Survey: outcomes from 4 geographical regions. Asia Pac Allergy. 2018; 8: e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Craig TJ McCann JL Gurevich F Davies MJ The correlation between allergic rhinitis and sleep disturbance. J Allergy Clin Immunol. 2004; 114: S139–S145. [DOI] [PubMed] [Google Scholar]
- 5. Varona R Ramos T Escribese MM Jimeno L Galán A Würtzen PA Vega F Marín A Martín S Carrera AC Blanco C Barber D Persistent regulatory T-cell response 2 years after 3 years of grass tablet SLIT: Links to reduced eosinophil counts, sIgE levels, and clinical benefit. Allergy. 2019; 74: 349–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Taylor JA Karas JL Ram MK Green OM Seidel-Dugan C Activation of the high-affinity immunoglobulin E receptor Fc epsilon RI in RBL-2H3 cells is inhibited by Syk SH2 domains. Mol Cell Biol. 1995; 15: 4149–4157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Shi JH Sun SC Tumor Necrosis Factor Receptor-Associated Factor Regulation of Nuclear Factor κB and Mitogen-Activated Protein Kinase Pathways. Front Immunol. 2018; 9: 1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Söderquist B Sundqvist KG Vikerfors T Adhesion molecules (E-selectin, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1)) in sera from patients with Staphylococcus aureus bacteraemia with or without endocarditis. Clin Exp Immunol. 1999; 118: 408–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Fukuda T Fukushima Y Numao T Ando N Arima M Nakajima H Sagara H Adachi T Motojima S Makino S Role of interleukin-4 and vascular cell adhesion molecule-1 in selective eosinophil migration into the airways in allergic asthma. Am J Respir Cell Mol Biol. 1996; 14: 84–94. [DOI] [PubMed] [Google Scholar]
- 10. Sehra S Yao W Nguyen ET Glosson-Byers NL Akhtar N Zhou B Kaplan MH TH9 cells are required for tissue mast cell accumulation during allergic inflammation. J Allergy Clin Immunol. 2015; 136: 433–40.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lukacs NW Role of chemokines in the pathogenesis of asthma. Nat Rev Immunol. 2001; 1: 108–116. [DOI] [PubMed] [Google Scholar]
- 12. Hossenbaccus L Linton S Garvey S Ellis AK Towards definitive management of allergic rhinitis: best use of new and established therapies. Allergy Asthma Clin Immunol. 2020; 16: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Noon L Prophylactic inoculation against hay fever. Int Arch Allergy Appl Immunol. 1953; 4: 285–288. [DOI] [PubMed] [Google Scholar]
- 14. Matsuoka T Shamji MH Durham SR Allergen immunotherapy and tolerance. Allergol Int. 2013; 62: 403–413. [DOI] [PubMed] [Google Scholar]
- 15. Shamji MH Larson D Eifan A Scadding GW Qin T Lawson K Sever ML Macfarlane E Layhadi JA Würtzen PA Parkin RV Sanda S Harris KM Nepom GT Togias A Durham SR Differential induction of allergen-specific IgA responses following timothy grass subcutaneous and sublingual immunotherapy. J Allergy Clin Immunol. 2021; 148: 1061–1071.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Scadding GW Calderon MA Shamji MH Eifan AO Penagos M Dumitru F Sever ML Bahnson HT Lawson K Harris KM Plough AG Panza JL Qin T Lim N Tchao NK Togias A Durham SR Effect of 2 Years of Treatment With Sublingual Grass Pollen Immunotherapy on Nasal Response to Allergen Challenge at 3 Years Among Patients With Moderate to Severe Seasonal Allergic Rhinitis: The GRASS Randomized Clinical Trial. JAMA. 2017; 317: 615–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Shamji MH Durham SR Mechanisms of allergen immunotherapy for inhaled allergens and predictive biomarkers. J Allergy Clin Immunol. 2017; 140: 1485–1498. [DOI] [PubMed] [Google Scholar]
- 18. Ebner C Siemann U Bohle B Willheim M Wiedermann U Schenk S Klotz F Ebner H Kraft D Scheiner O Immunological changes during specific immunotherapy of grass pollen allergy: reduced lymphoproliferative responses to allergen and shift from TH2 to TH1 in T-cell clones specific for Phl p 1, a major grass pollen allergen. Clin Exp Allergy. 1997; 27: 1007–1015. [DOI] [PubMed] [Google Scholar]
- 19. Guerra F Carracedo J Solana-Lara R Sánchez-Guijo P Ramírez R TH2 lymphocytes from atopic patients treated with immunotherapy undergo rapid apoptosis after culture with specific allergens. J Allergy Clin Immunol. 2001; 107: 647–653. [DOI] [PubMed] [Google Scholar]
- 20. Ciepiela O Zawadzka-Krajewska A Kotuła I van Overveld F Kulus M Demkow U Sublingual Immunotherapy for Asthma: Affects T-Cells but Does not Impact Basophil Activation. Pediatr Allergy Immunol Pulmonol. 2014; 27: 17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Yao Y Chen CL Yu D Liu Z Roles of follicular helper and regulatory T cells in allergic diseases and allergen immunotherapy. Allergy. 2021; 76: 456–470. [DOI] [PubMed] [Google Scholar]
- 22. Breitfeld D Ohl L Kremmer E Ellwart J Sallusto F Lipp M Förster R Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. 2000; 192: 1545–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yao Y Wang ZC Wang N Zhou PC Chen CL Song J Pan L Liao B Zhang XH Yang YS Xu XY Zhu RF Yu D Liu Z Allergen immunotherapy improves defective follicular regulatory T cells in patients with allergic rhinitis. J Allergy Clin Immunol. 2019; 144: 118–128. [DOI] [PubMed] [Google Scholar]
- 24. Sharif H Acharya S Dhondalay GKR Varricchi G Krasner-Macleod S Laisuan W Switzer A Lenormand M Kashe E Parkin RV Yi Y Koc M Fedina O Vilà-Nadal G Marone G Eifan A Scadding GW Fear DJ Nadeau KC Durham SR Altered chromatin landscape in circulating T follicular helper and regulatory cells following grass pollen subcutaneous and sublingual immunotherapy. J Allergy Clin Immunol. 2021; 147: 663–676. [DOI] [PubMed] [Google Scholar]
- 25. Shamji MH Layhadi JA Achkova D Kouser L Perera-Webb A Couto-Francisco NC Parkin RV Matsuoka T Scadding G Ashton-Rickardt PG Durham SR Role of IL-35 in sublingual allergen immunotherapy. J Allergy Clin Immunol. 2019; 143: 1131–1142. [DOI] [PubMed] [Google Scholar]
- 26. Radulovic S Jacobson MR Durham SR Nouri-Aria KT Grass pollen immunotherapy induces Foxp3-expressing CD4+ CD25+ cells in the nasal mucosa. J Allergy Clin Immunol. 2008; 121: 1467–1472. [DOI] [PubMed] [Google Scholar]
- 27. Xian M Feng M Dong Y Wei N Su Q Li J Changes in CD4 CD25 FoxP3 Regulatory T Cells and Serum Cytokines in Sublingual and Subcutaneous Immunotherapy in Allergic Rhinitis with or without Asthma. Int Arch Allergy Immunol. 2020; 181: 71–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Swamy RS Reshamwala N Hunter T Vissamsetti S Santos CB Baroody FM Hwang PH Hoyte EG Garcia MA Nadeau KC Epigenetic modifications and improved regulatory T-cell function in subjects undergoing dual sublingual immunotherapy. J Allergy Clin Immunol. 2012; 130: 215–24.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Brooks DG Trifilo MJ Edelmann KH Teyton L McGavern DB Oldstone MBA Interleukin-10 determines viral clearance or persistence in vivo. Nat Med. 2006; 12: 1301–1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Okano M Otsuki N Azuma M Fujiwara T Kariya S Sugata Y Higaki T Kino K Tanimoto Y Okubo K Nishizaki K Allergen-specific immunotherapy alters the expression of B and T lymphocyte attenuator, a co-inhibitory molecule, in allergic rhinitis. Clin Exp Allergy. 2008; 38: 1891–1900. [DOI] [PubMed] [Google Scholar]
- 31. Burton BR Britton GJ Fang H Verhagen J Smithers B Sabatos-Peyton CA Carney LJ Gough J Strobel S Wraith DC Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nat Commun. 2014; 5: 4741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Rosskopf S Jahn-Schmid B Schmetterer KG Zlabinger GJ Steinberger P PD-1 has a unique capacity to inhibit allergen-specific human CD4 T cell responses. Sci Rep. 2018; 8: 13543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wambre E Effect of allergen-specific immunotherapy on CD4+ T cells. Curr Opin Allergy Clin Immunol. 2015; 15: 581–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Zimmer A Bouley J Le Mignon M Pliquet E Horiot S Turfkruyer M Baron-Bodo V Horak F Nony E Louise A Moussu H Mascarell L Moingeon P A regulatory dendritic cell signature correlates with the clinical efficacy of allergen-specific sublingual immunotherapy. J Allergy Clin Immunol. 2012; 129: 1020–1030. [DOI] [PubMed] [Google Scholar]
- 35. Ness S Lin S Gordon JR Regulatory Dendritic Cells, T Cell Tolerance, and Dendritic Cell Therapy for Immunologic Disease. Front Immunol. 2021; 12: 633436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Angelini F Pacciani V Corrente S Silenzi R Di Pede A Polito A Riccardi C Di Cesare S Yammine ML Rossi P Moschese V Chini L Dendritic cells modification during sublingual immunotherapy in children with allergic symptoms to house dust mites. World J Pediatr. 2011; 7: 24–30. [DOI] [PubMed] [Google Scholar]
- 37. Golebski K Layhadi JA Sahiner U Steveling-Klein EH Lenormand MM Li RCY Bal SM Heesters BA Vilà-Nadal G Hunewald O Montamat G He FQ Ollert M Fedina O Lao-Araya M Vijverberg SJH Maitland-van der Zee AH van Drunen CM Fokkens WJ Durham SR Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity. 2021; 54: 291–307. [DOI] [PubMed] [Google Scholar]
- 38. Rosser EC Mauri C Regulatory B cells: origin, phenotype, and function. Immunity. 2015; 42: 607–612. [DOI] [PubMed] [Google Scholar]
- 39. Boonpiyathad T van de Veen W Wirz O Sokolowska M Rückert B Tan G Sangasapaviliya A Pradubpongsa P Fuengthong R Thantiworasit P Sirivichayakul S Ruxrungtham K Akdis CA Akdis M Role of Der p 1-specific B cells in immune tolerance during 2 years of house dust mite-specific immunotherapy. J Allergy Clin Immunol. 2019; 143: 1077–1086.e10. [DOI] [PubMed] [Google Scholar]
- 40. Shamji MH Valenta R Jardetzky T Verhasselt V Durham SR Würtzen PA van Neerven RJJ The role of allergen-specific IgE, IgG and IgA in allergic disease. Allergy. 2021; 76: 3627–3641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Scadding GW Shamji MH Jacobson MR Lee DI Wilson D Lima MT Pitkin L Pilette C Nouri-Aria K Durham SR 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] [PubMed] [Google Scholar]
- 42. López-Sanz C Jiménez-Saiz R Esteban V Delgado-Dolset MI Perales-Chorda C Villaseñor A Barber D Escribese MM Mast Cell Desensitization in Allergen Immunotherapy. Front Allergy. 2022; 3: 398494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Shamji MH Ljørring C Francis JN Calderon MA Larché M Kimber I Frew AJ Ipsen H Lund K Würtzen PA Durham SR Functional rather than immunoreactive levels of IgG4 correlate closely with clinical response to grass pollen immunotherapy. Allergy. 2012; 67: 217–226. [DOI] [PubMed] [Google Scholar]
- 44. Barber D Escribese MM Predictive biomarkers in allergen specific immunotherapy. Allergol Immunopathol (Madr). 2017; 45: 12–14. [DOI] [PubMed] [Google Scholar]
- 45. Villaseñor A Rosace D Obeso D Pérez-Gordo M Chivato T Barbas C Barber D Escribese MM Allergic asthma: an overview of metabolomic strategies leading to the identification of biomarkers in the field. Clin Exp Allergy. 2017; 47: 442–456. [DOI] [PubMed] [Google Scholar]
- 46. Akdis CA Akdis M Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol. 2011; 127: 18–27, quiz 28-29.. [DOI] [PubMed] [Google Scholar]
- 47. Glesner J Filep S Vailes LD Wünschmann S Chapman MD Birrueta G Frazier A Jeong KY Schal C Bacharier L Beigelman A Busse P Schulten V Sette A Pomés A Allergen content in German cockroach extracts and sensitization profiles to a new expanded set of cockroach allergens determine in vitro extract potency for IgE reactivity. J Allergy Clin Immunol. 2019; 143: 1474–1481.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Waldron R McGowan J Gordon N McCarthy C Mitchell EB Fitzpatrick DA Proteome and allergenome of the European house dust mite Dermatophagoides pteronyssinus. PLoS One. 2019; 14: e0216171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Li H Wen Y Wu S Chen D Luo X Xu R Ma R Wen W Epigenetic Modification of Enhancer of Zeste Homolog 2 Modulates the Activation of Dendritic Cells in Allergen Immunotherapy. Int Arch Allergy Immunol. 2019; 180: 120–127. [DOI] [PubMed] [Google Scholar]
- 50. Benito-Villalvilla C Pérez-Diego M Angelina A Kisand K Rebane A Subiza JL Palomares O Allergoid-mannan conjugates reprogram monocytes into tolerogenic dendritic cells via epigenetic and metabolic rewiring. J Allergy Clin Immunol. 2022; 149: 212–222.e9. [DOI] [PubMed] [Google Scholar]
- 51. Barker-Tejeda TC Bazire R Obeso D Mera-Berriatua L Rosace D Vazquez-Cortes S Ramos T Rico MDP Chivato T Barbas C Villaseñor A Escribese MM Fernández-Rivas M Blanco C Barber D Exploring novel systemic biomarker approaches in grass-pollen sublingual immunotherapy using omics. Allergy. 2021; 76: 1199–1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zheng P Bian X Zhai Y Metabolomics reveals a correlation between hydroxyeicosatetraenoic acids and allergic asthma: Evidence from three years’ immunotherapy. Pediatr Allergy Immunol. 2021; 32: 1654–1662. [DOI] [PubMed] [Google Scholar]
- 53. Zheng P Yan G Zhang Y Huang H Luo W Xue M Li N Wu JL Sun B Metabolomics Reveals Process of Allergic Rhinitis Patients with Single- and Double-Species Mite Subcutaneous Immunotherapy. Metabolites. 2021; 11: 613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Delgado-Dolset MI Obeso D Rodríguez-Coira J Tarin C Tan G Cumplido JA Cabrera A Angulo S Barbas C Sokolowska M Barber D Carrillo T Villaseñor A Escribese MM Understanding uncontrolled severe allergic asthma by integration of omic and clinical data. Allergy. 2022; 77: 1772–1785. [DOI] [PubMed] [Google Scholar]
- 55. Xie S Zhang H Liu Y Gao K Zhang J Fan R Xie S Xie Z Wang F Jiang W The Role of Serum Metabolomics in Distinguishing Chronic Rhinosinusitis With Nasal Polyp Phenotypes. Front Mol Biosci. 2021; 7: 305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Shi HY Pan C Ma TT Chen YL Yan WJ Liu JG Cao MD Huang HD Wang DY Wang XY Wei JF Clinical Efficacy Evaluation of 1-Year Subcutaneous Immunotherapy for Artemisia sieversiana Pollen Allergic Rhinitis by Serum Metabolomics. Front Pharmacol. 2020; 11: 305. [DOI] [PMC free article] [PubMed] [Google Scholar]
