Next Article in Journal
Innovative Epicardial Bigels Containing Amiodarone Hydrochloride: Pharmacotechnical and Analytical Characterization
Next Article in Special Issue
Adjuvant Effect of Lactobacillus paracasei in Sublingual Immunotherapy of Asthmatic Mice
Previous Article in Journal
Identification of Estrogen-Responsive Proteins in Mouse Seminal Vesicles Through Mass Spectrometry-Based Proteomics
Previous Article in Special Issue
Therapies for Chronic Spontaneous Urticaria: Present and Future Developments
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Approaches to Allergen Immunotherapy for Respiratory Allergies

by
Mongkol Lao-Araya
Division of Allergy and Clinical Immunology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
Pharmaceuticals 2024, 17(11), 1510; https://doi.org/10.3390/ph17111510
Submission received: 15 October 2024 / Revised: 1 November 2024 / Accepted: 6 November 2024 / Published: 9 November 2024
(This article belongs to the Special Issue Drug Candidates for Allergic Diseases)

Abstract

:
Allergen immunotherapy (AIT) remains the cornerstone for managing respiratory allergies, offering long-term symptom relief, disease modification, and prevention of disease progression. While novel approaches like intralymphatic and epicutaneous immunotherapy and the combination of allergens with adjuvants show promise, traditional methods remain effective and safe. Hypoallergenic T-cell peptide vaccines and recombinant allergens require further research to confirm their clinical benefits. Passive immunotherapy, while demonstrating effectiveness in specific cases, needs exploration of its long-term efficacy and broader applicability. Combining AIT with biologics may enhance safety and treatment outcomes. Despite emerging innovations, allergen-specific immunotherapy with natural allergen extracts remains the primary disease-modifying treatment, offering long-term symptom relief and prevention of disease progression. Continued research is essential to refine and optimize allergen immunotherapy strategies, providing patients with more effective and personalized treatment options.

1. Introduction

Allergen immunotherapy (AIT), a therapeutic approach rooted in the concept of immunization, has evolved significantly since its inception. Its origins can be traced back to Edward Jenner’s pioneering smallpox vaccine in 1796, demonstrating the therapeutic potential of inducing immunity. The modern understanding of allergic disease emerged in the late 19th century with Blackley’s observations linking pollen exposure to hay fever symptoms [1,2]. The therapeutic application of AIT for hay fever was first introduced by Noon and Freeman in 1911, marking the beginning of a century-long journey of research and development [3]. While early studies laid the groundwork, rigorous clinical trials with well-characterized allergen extracts did not emerge until the 1980s, establishing the dose-dependent therapeutic effect of AIT [2,4,5,6,7].
Over the past century, AIT has undergone significant advancements. Despite its initial discovery over 110 years ago, AIT remains the standard therapy for allergic rhinitis (AR) and asthma [1,2]. Its proven efficacy in reducing symptoms and improving quality of life has earned the recommendation of numerous medical organizations [8,9,10]. AIT offers a unique disease-modifying approach, inducing long-term allergen tolerance and reducing allergic inflammation [11]. This is achieved by administering increasing doses of allergens via subcutaneous (SCIT) or sublingual (SLIT) routes.
While AIT offers substantial benefits, traditional treatment regimens can be lengthy and associated with adverse reactions. The need for prolonged administration, often spanning three years or more, can deter patients. Additionally, the risk of adverse reactions, particularly during dose escalation, can lead to treatment discontinuation [12]. Furthermore, AIT’s effectiveness varies across different allergens and patient populations [13].
To address these limitations, ongoing research focuses on optimizing AIT delivery methods, treatment procedures, and patient adherence, particularly in pediatric populations. Novel approaches, such as allergen component therapy, hypoallergenic immunotherapy, new delivery routes, and the combination of allergens with adjuvants or biologics, hold promise in enhancing the efficacy and safety of AIT [2,14,15,16,17,18]. These innovations are expanding the therapeutic landscape for AIT, offering hope for individuals with respiratory allergies (Figure 1).
This review will explore the latest advancements in AIT, encompassing improvements in allergen extracts, delivery methods, and treatment strategies. By highlighting these innovations, we aim to provide insights into the future of AIT and its potential to enhance the lives of individuals with respiratory allergies.

2. Mechanisms of AIT (Table 1)

The allergic response is a multifaceted process involving the interaction of various immune cells, mediators, and cytokines. It begins when antigen-presenting cells (APCs), such as dendritic cells (DCs), recognize an allergen. These APCs, activated by epithelial-derived cytokines and those from type 2 innate lymphoid cells (ILC2) [19] and basophils, present the allergen to naive T cells, guiding their differentiation into T-helper 2 (TH2) cells. TH2 and follicular T-helper (Tfh) cells play a pivotal role in driving the allergic response by producing IL-4 and IL-13. These cytokines promote IgE production by B cells, enhancing the allergic inflammatory environment. In individuals with allergies, repeated exposure to low-dose allergens through a compromised epithelial barrier can lead to increased sIgE levels and subsequent allergic reactions [20,21,22].
While research on AIT mechanisms has primarily focused on aeroallergens, studies suggest that high-dose allergen exposure can play a pivotal role in restoring the epithelial barrier and inducing a shift from allergic TH2 inflammation to a more tolerant TH1 response [23]. This shift involves the generation of suppressive regulatory immune cells and a decrease in pro-allergic cytokines (IL-4, IL-5, IL-9, and IL-13). The reduction in allergic inflammation is accompanied by a decrease in mast cells, basophils, and eosinophils, key cells involved in allergic reactions. Additionally, there is an increase in allergen-specific T regulatory (Treg) cells [24,25], T follicular regulatory (Tfr) cells [26], and B regulatory (Breg) cells, which can help suppress the allergic immune response [22,23].
Furthermore, AIT is effective in inducing dendritic cell-derived regulatory cells (DCregs) [27] and innate lymphoid cell-derived regulatory cells (ILCregs) [28,29,30]. These regulatory cells produce cytokines like TGF-β, IL-12, IL-27, and IL-10, which can suppress allergic responses [24,31]. AIT is also associated with the generation of Treg-cell subsets, which can further suppress TH2 and Tfh-cell responses, leading to a shift towards TH1 cells [32].
Initial immunotherapy may lead to a temporary increase in sIgE levels. However, over time, the immune response shifts towards a tolerogenic state. Breg cells, stimulated by cytokines like IL-10 and IL-35, play a crucial role in this process. These cells promote the production of blocking antibodies, including IgG1, IgG4, IgA1, and IgA2, which can bind to allergens and prevent them from triggering allergic responses [33]. These blocking antibodies are found both in the bloodstream and in mucosal secretions. For SCIT, IgG4 is the primary blocking antibody, while for SLIT, IgA1 and IgA2 are more prominent in both nasal and systemic compartments [23,34,35,36].
In summary, the mechanisms of immunotherapy involve a complex interplay of innate and adaptive immune cells, cytokines, and antibodies, ultimately leading to a shift towards a more tolerant immune response and reduced allergic symptoms [23,37].
Table 1. A summary of the main mechanisms and proven clinical benefits of allergen immunotherapy.
Table 1. A summary of the main mechanisms and proven clinical benefits of allergen immunotherapy.
Mechanism of AIT
  • Restoration of epithelial cell integrity
  • Decrease in allergen-dependent mast cell/basophil degranulation
  • Reduction in type 2 immune responses
  • Regulation of T-cell responses: suppression of TH2 and immune deviation toward a TH1 response, reduction of circulating Tfh cells
  • Modulation of ILCs: reduction of circulating ILC2s, induction of IL-10+ regulatory ILC2s
  • Induction of regulatory response: induction of Treg and Breg cells
  • Stimulation of allergen-specific blocking antibody production, including IgG1, IgG2, IgG4, and IgA, in both systemic and mucosal immune responses
  • Induction of tolerogenic cytokines: IL-10, IL-12, IL-27, IL-35 and TGF-β
Proven clinical benefits of AIT in allergic rhinoconjunctivitis +/− asthma
  • Reduced symptoms and medication use
  • Improved quality of life
  • The benefits can last for many years, even after treatment is stopped.
  • Reduced risk of developing asthma in children with AR
  • Prevent subsequent allergic sensitization

3. Conventional AIT: SCIT and SLIT

SCIT and SLIT have been used as standard treatments for respiratory allergies for decades [10]. SCIT involves administering allergen extracts in gradually increasing doses over several weeks or months, followed by a maintenance phase of monthly injections for 3 to 5 years. While SCIT is generally safe, it carries a risk of systemic allergic reactions (up to 22%), including anaphylaxis [8,10,38,39,40]. However, the risk can be minimized with appropriate patient selection, adequate facilities, well-trained staff, and availability of emergency treatment [8,10]. The efficacy of SCIT varies depending on the allergen and the specific product used. Meta-analyses have demonstrated that SCIT is approximately 30% more effective than placebo in treating seasonal and perennial allergic rhinitis, [41] exceeding the World Allergy Organization’s defined minimally clinically important difference of 20% [42].
While SCIT has been shown to be as effective as or more effective than pharmacotherapy in extrapolated pooled analyses, there are few direct comparisons between the two [43,44]. In practice, adherence and persistence with SCIT can be challenging, with studies showing that less than 50% of users completed the recommended 3 years of treatment [12,45].
In recent years, SLIT has emerged as a well-validated alternative to SCIT. Several large randomized controlled trials have confirmed the efficacy of SLIT tablets for patients with house dust mite (HDM), [46,47,48] grass pollen, [49] ragweed, [50] and Japanese cedar [51,52] pollen allergic rhinitis, including those with mild to moderately severe controlled HDM-induced allergic asthma [53,54].
SLIT involves taking tablets or drops sublingually daily for 3 years or starting 2 to 4 months before the allergy season in patients with seasonal AR [55]. After initial supervision and a 30-min observation period, SLIT can be self-administered, making it convenient for patients. Home administration and scheduled follow-up visits contribute to improved adherence with SLIT. Compared to SCIT, SLIT is generally safer. While SLIT is associated with local side effects like oropharyngeal itching and swelling, these are typically self-limiting and resolve within 1 to 2 weeks. Systemic side effects are rare, making SLIT a safer alternative to SCIT [38,56,57].
Both SCIT and SLIT have been shown to be effective for both seasonal and perennial allergies [41]. Indirect meta-analyses suggest that SLIT is at least as effective as current pharmacotherapy, although head-to-head controlled studies are needed for definitive confirmation [44,58]. A recent direct comparison using nasal allergen challenge found that SCIT was more effective than SLIT in the first year of treatment. However, the two treatments were equally effective in the second year [36]. Due to the heterogeneity between studies, treatment decisions should be based on the evidence available for specific products and individual patient factors [57,59,60].

4. Alternative Routes (Table 2)

4.1. Intralymphatic Immunotherapy

Intralymphatic immunotherapy (ILIT) is a targeted immunotherapy approach that involves injecting allergen extracts directly into lymph nodes, usually in the groin, under ultrasound guidance [61]. An ILIT injection of recombinant allergens (phospholipase A2 and Fel d 1) has demonstrated a significant increase in IgG levels compared to SCIT, achieving a 10-fold increase in just two weeks at a 100-fold lower dose [62,63]. This direct approach targets the immune system, potentially enhancing allergen presentation to T cells and avoiding direct mast cell activation [18,64].
A randomized open-label trial compared a 3-dose ILIT regimen administered over 2 months to a 3-year SCIT regimen in patients with grass pollen allergy, finding that ILIT achieved a persistent effect over 3 years with fewer side effects and significantly improved compliance [61]. While ILIT resulted in slightly less medication use and similar symptom score improvements, the low compliance rate in the SCIT group and the open-label nature of the study make it challenging to draw definitive conclusions.
Several studies have shown the efficacy of ILIT for grass and tree pollen allergy, although not all studies have confirmed its effectiveness [61,65,66]. A small-scale study showed that three intralymphatic injections of a recombinant cat Fel d 1 allergen, fused with a translocation sequence and a human invariant chain fragment, were effective in protecting against nasal challenge with whole cat allergen extract [62].
Overall, ILIT offers potential advantages in terms of reduced side effects, improved compliance, and shorter treatment duration [61,67]. However, it requires specialist skills, experience, and ultrasound guidance for injections, making it more technically demanding than traditional routes. Individual products, doses, and timing of injections still need to be optimized [64]. Further research is needed to confirm its efficacy and safety for various allergens and patient populations.
Table 2. Routes of allergen immunotherapy administration.
Table 2. Routes of allergen immunotherapy administration.
Routes of AITAdvantagesDrawbacks
Subcutaneous (SCIT)
  • Proven efficacy and safety
  • Standard applications in respiratory and venom AIT
  • Repeated injections
  • Healthcare unit dependency
  • Long duration of treatment
  • Risk of severe hypersensitivity reactions
Sublingual (SLIT)
  • Proven efficacy and safety
  • Standard applications in respiratory AIT
  • No injections
  • Less healthcare unit dependency
  • Home application
  • Lower risk of severe hypersensitivity reactions
  • Need for daily self-applications which may affect the adherence
  • Higher allergen dose
Intralymphatic (ILIT)
  • Applicability in respiratory and venom allergens
  • Reduced number of injections
  • Reduced treatment duration
  • Reduced allergen dosage use
  • Early clinical studies revealed promising results
  • Requirement of experienced HCP for injections under ultrasound guidance
  • Risk for side effects due to allergens injected into lymph nodes
  • Further clinical study needed
Epicutaneous (EPIT)
  • Applicability in food and respiratory allergens
  • Good safety profile
  • Early clinical studies revealed promising results
  • Risk of local adverse reactions
  • Further clinical study needed
Intradermal (IDIT)
  • May reduce allergen dosage use
  • No clinical efficacy proven

4.2. Epicutaneous Immunotherapy

Epicutaneous immunotherapy (EPIT) involves applying allergen patches to the skin for several hours, aiming to increase local antigen presentation while preventing systemic allergen absorption. By removing the top layer of skin with adhesive tape, keratinocytes can be activated, increasing allergen exposure and potentially stimulating DC responses. This needle-free approach can improve patient compliance, especially in children [68,69].
A placebo-controlled randomized trial applied grass pollen extract in petroleum to the skin, stripped with adhesive tape, weekly for 6 months pre-seasonally in 48 participants. The study observed a 48% improvement in seasonal symptom scores in the first year compared to a 10% improvement in the placebo group. However, there were no significant differences in combined treatment and medication scores. Two further randomized controlled trials achieved similar results [70]. While higher doses were effective, they also led to high rates of local irritation, eczema, and occasional systemic allergic side effects, limiting their clinical utility compared to currently available SCIT [17,71].
A placebo-controlled randomized trial applied grass pollen extract to the skin, prepared with adhesive tape, weekly for 6 months pre-seasonally in 48 participants. The study observed a significant improvement in seasonal symptom scores in the treatment group compared to the placebo group [70]. However, while higher doses were effective, they also led to increased local skin reactions and systemic side effects, limiting their clinical utility. Two further randomized controlled trials yielded similar results. While EPIT has shown promise, its efficacy and safety profile still need to be optimized to compete with established SCIT [17,71].

4.3. Intradermal Immunotherapy

Intradermal allergen administration may increase the immune response and decrease the required allergen dose due to the presence of DCs in the intradermal area [17,71]. Early studies suggested that repeated low-dose intradermal injections could suppress late allergic responses and induce allergen-specific IgG antibodies [17,72]. However, a phase 2b trial of pre-seasonal low-dose intradermal grass pollen allergen failed to demonstrate significant clinical improvement. In fact, nasal symptoms worsened, and a heightened TH2 response was observed at the injection site [73]. These findings suggest that intradermal allergen administration may have paradoxical effects, potentially inducing both sensitization and tolerance. Therefore, this approach is not currently recommended for allergen immunotherapy.

5. Adjuvants (Table 3)

An adjuvant is a substance that enhances the immune response to a vaccine. By physically or chemically interacting with antigens, adjuvants can modify the pharmacological and immunological effects of allergen vaccines. They can modulate allergen delivery, act as a depot, stimulate immune responses, and steer the immune response towards either tolerance or a TH1-biased response. Additionally, adjuvants can help reduce the risk of anaphylactic reactions and unwanted side effects [14,17,18,74].
Traditionally, adjuvants have been classified into first-generation (aluminum hydroxide, microcrystalline tyrosine [MCT], and calcium phosphate) and second-generation (Monophosphoryl Lipid A [MPLA], Toll-like receptor [TLR] agonists) categories. Other promising adjuvants include liposomes and virus-like particles (VLP) [14]. The adjuvants currently approved for use in humans (aluminum hydroxide, calcium phosphate, MCT, and MPLA) [18] offer several advantages for improving AIT. These include prolonging antigen exposure at the injection site and stimulating the production of allergen-specific IgG antibodies.
Table 3. Adjuvants in allergen immunotherapy.
Table 3. Adjuvants in allergen immunotherapy.
AdjuvantsApproved for Clinical Use [18]Mechanism of Action
Aluminum hydroxide
(alum)
EU (1937)
  • Allergen depot
  • Inhibits TH2 and enhances TH1 responses
  • Low biodegradability, may lead to adverse reactions
Calcium Phosphate
(CaP)
EU (1980)
  • Allergen depot
  • Natural component
  • High biodegradability
Microcrystalline
tyrosine (MCT)
EU (1970)
  • Biodegradable depot adjuvant
  • Inhibits TH2 and enhances TH1 responses
Monophosphoryl Lipid
A (MPLA)
EU (1999)
  • TLR-4 agonist
  • Stimulates TH1 response
  • Works synergistically with MCT
CpG-Oligodesoxy-
nucleotides
No
  • TLR-9 agonist
  • Stimulates TH1 response
  • Combined in VLP as ‘allergen-independent TH1 simulant’
  • Conflicting results in Phase 3 clinical trial
Nanoparticles: lipophilic liposomes, virus-like particles (VLPs) and other particles from
synthetic and
natural polymers
No
  • Encapsulation of allergen
  • Allowing uptake into APCs without IgE binding
  • Activation of innate immunity without T cell help
  • Limited data in clinical trials

5.1. Alum and Calcium Phosphate

Aluminum salts (alum) have been used as a vaccine adjuvant since 1926 [75] and remain one of the most widely used adjuvants in human vaccines [74]. While alum-based allergen extracts are licensed for SCIT in Europe, they are not approved for use in the United States. Alum functions by adsorbing the allergen and triggering both innate and adaptive immune responses, including inflammasome activation and T-cell activation, which can enhance antibody responses. Although alum can increase TH2 responses in mouse models during sensitization, in humans, it has been shown to inhibit established allergic TH2 responses and promote TH1 responses, both in vitro and in vivo [14,74].
Alum-based allergy extracts have a long history of safe and effective use in Europe. While alum is well-tolerated, it can induce acute and chronic inflammation at the injection site due to its low biodegradability. Although there is a theoretical risk of aluminum accumulation and systemic side effects, this has not been observed in humans [74,76].
Calcium phosphate, a natural component of the body, is another depot adjuvant with better biodegradability and biocompatibility than alum [14]. Although calcium phosphate-adjuvanted AIT products were once available in the European market, they are no longer available for reasons that remain unclear [74].

5.2. Microcrystalline Tyrosine (MCT)

MCT, the crystalline form of the non-essential amino acid L-tyrosine, is a promising adjuvant for AIT [14,74]. While less commonly used than alum, MCT has demonstrated safety and efficacy in humans. It is a biodegradable depot adjuvant with a short half-life of 48 h. Since its initial report in the 1980s, MCT has been shown to enhance the induction of IgG antibodies when used with allergenic molecules. Unlike aluminum, which can accumulate at injection sites in murine models of AIT and is associated with granuloma formation, MCT is rapidly released and metabolized, reducing the risk of long-term accumulation [77,78].
MCT, a non-toxic adjuvant except in individuals with tyrosine metabolism disorders, [14] is currently patented for immunotherapy. It is integrated into glutaraldehyde allergoids to alleviate allergic symptoms and reduce reliance on relief medications [79].

5.3. Toll-like Receptors (TLRs)

Toll-like receptors (TLRs), a class of pattern-recognition receptors, are primarily expressed on APCs. Upon recognition of specific pathogen-associated molecular patterns (PAMPs), they initiate both innate and adaptive immune responses [14,17,74].
Monophosphoryl lipid A (MPL), a TLR4 agonist derived from the lipopolysaccharide of Salmonella minnesota, stimulates the production of IFN-γ and IL-12 but does not promote IL-5 synthesis [74]. MPL has shown promise as an adjuvant for AIT. In a study of grass pollen allergy, pre-seasonal injections with MPL-containing allergoids led to a significant reduction in symptoms compared to placebo, with effects lasting up to 5 years after discontinuation [80,81].
MCT-adjuvanted allergen vaccines, which may include native or modified (allergoid) allergens, are often combined with MPL, as exemplified by Pollinex®Quattro, an allergen therapeutic available for the treatment of seasonal allergic rhinoconjunctivitis. Combining allergens with MPL and MCT may address some of the major drawbacks of traditional AIT, including long treatment durations, poor patient adherence, and adverse effects [82,83,84,85]. While Pollinex®Quattro is currently available in Germany, sufficient clinical human data supporting marketing authorization in other European states have not been reported [74].
Cytosine-phosphodiester-guanine (CpG) motifs, conserved PAMPs-bacterial DNA sequences, are recognized by TLR-9, a receptor primarily expressed on B cells and plasmacytoid dendritic cells (pDCs). Activation of TLR-9 stimulates the innate immune response in a TH1-type fashion [18,74]. A phase 2 trial demonstrated that combining CpG motifs with the ragweed allergen Amb a 1 suppressed seasonal symptoms in ragweed allergy patients [86]. However, these results were not replicated in a larger phase 3 trial [17]. While TLR-9 agonists show potential as adjuvants for AIT, additional research is needed to fully elucidate the mechanisms of action of these adjuvants and optimize their clinical application.

5.4. Liposomes and Virus-like Particles (VLPs)

Polymeric biodegradable nanoparticles, including polyesters, polysaccharides, polyamides, liposomes, and virus-like particles (VLPs), offer promising strategies for encapsulating allergens or proteins and delivering them to APCs without IgE binding. While many of these delivery systems are still under development, they have shown potential for inducing TH1-biased immune responses in preclinical studies [14,17,74].
Liposomes, spherical or vesicular structures composed of lipids, can effectively package and deliver water-soluble antigens to target cells. Studies have demonstrated that liposome-encapsulated allergens can induce higher levels of allergen-specific IgG while reducing IgE production in mice [87]. Liposomes may also enhance lymph node allergen delivery, potentially improving the efficacy of AIT [64].
VLPs, derived from viral capsids, can activate the immune system through innate pathways, bypassing the need for T-cell help. VLPs are efficiently taken up by APCs, resulting in the activation of cytotoxic T cells and the complement system. VLPs can be used in various ways for AIT, including as standalone VLPs with or without embedded adjuvants, as solutions mixed with allergens, or with allergens encapsulated within the VLPs. These different approaches offer flexibility in designing AIT regimens and may have varying effects on the immune response. Conjugating allergens with VLPs can enhance their uptake into the lymphatic system, potentially minimizing the risk of mast cell degranulation [2,16,74].
VLPs have demonstrated promising results in preclinical studies. A phase 2 trial involving patients with HDM allergy found that combining VLPs with a cytosine-phosphodiester-guanine TLR-9 agonist (CβG10) and allergen encapsulation (CYT003) resulted in similar symptom improvements compared to the adjuvant alone [88,89]. Encouraged by these findings, larger placebo-controlled trials were conducted. A double-blind, randomized trial involving 299 HDM allergy patients found that QβG10, a VLP-based therapy administered without additional allergen, reduced symptom scores in a dose-dependent manner [90]. Additionally, a higher dose of QβG10 was associated with a greater number of patients achieving increased allergen tolerance [90]. These results suggest that VLP-based therapies, even without the addition of allergens, may have therapeutic benefits in allergic diseases [16,74].
However, subsequent trials yielded mixed results. A study in asthmatic patients undergoing steroid withdrawal found that QβG10 reduced asthma-related symptom scores and increased the number of patients with well-controlled asthma [88]. However, a larger phase IIb trial in patients with persistent moderate-to-severe allergic asthma did not demonstrate significant differences between QβG10 and placebo [91].
Nanoparticle-based delivery systems offer potential advantages for AIT by improving allergen delivery, modulating immune responses, and potentially reducing side effects. However, further research is necessary to evaluate their safety and efficacy in clinical trials.

6. Modified Allergens (Table 4)

Modifying allergens to alter their tertiary protein structure or target non-IgE-reactive epitopes can help preserve or enhance their ability to induce immune responses while minimizing allergic reactions, promoting tolerogenic outcomes.
Table 4. Modified allergens and novel approaches in allergen immunotherapy.
Table 4. Modified allergens and novel approaches in allergen immunotherapy.
Modified AllergensAdvantagesDrawbacks
  • Allergoids
Reduced allergenicity, improved safety profiles, shorter up-dosing phaseLimited efficacy data compared to conventional allergen extracts
  • Recombinant allergens
Personalized AIT, almost unlimited supply, may improve safety and efficacyLimited efficacy and safety advantages compared to conventional extracts, reported late-phase adverse allergic reactions
  • T-cell peptides
Retained T-cell epitope stimulation, reduced IgE binding, may improve safety and efficacyPotential for late-phase adverse responses, similar side effects to SCIT, failure to demonstrate clinical efficacy in phase 3 trials
  • B-cell peptides
Induce protective humoral antibody responses without stimulating IgE production, demonstrated increases in blocking IgG1 and IgG4 antibodies in phase 2 trialLimited clinical data, did not reach statistical significance in primary analysis of combined seasonal symptom medication scores
  • DNA-based vaccine
Induce TH1 and Treg cell responses while downregulating TH2 cell responses in preclinical studiesConcern of theoretical risks of plasmid DNA integration into the human genome and development of anti-DNA antibodies, failure to demonstrate clinical efficacy in phase 3 trials
Passive immunotherapy (monoclonal antibodies)Protection against nasal allergen challenge for almost 3 months after 1 dose injectionNo long-term clinical efficacy shown
AIT combined with biologicsEnhanced efficacy and safety, potential for long-term allergen tolerance High cost, further research needed to evaluate long-term benefits

6.1. Allergoids

Allergoids are chemically modified allergens produced through polymerization with glutaraldehyde or formaldehyde or monomerization with carbamylation. These modifications aim to alter the tertiary protein structure, reducing the ability of allergens to cross-link IgE while preserving shorter linear T-cell epitopes, thereby retaining immunogenicity. This approach allows for higher doses to be administered during a shorter-term accumulation phase, potentially improving treatment efficiency and reducing the risk of side effects [14,18,92].
Several allergoids have demonstrated efficacy in placebo-controlled trials for various allergens, including ragweed [92,93], grass [94,95,96], tree pollen [97,98,99], and mite [100,101,102,103,104] allergy. A phase 3 trial of a formaldehyde-treated, alum-adsorbed 6-grass pollen mix showed a significant 26.6% reduction in combined seasonal symptom medication scores after one year compared to placebo. Although some participants experienced mild to moderate late systemic reactions, no serious adverse events, such as anaphylaxis, were reported [105].
Carbamylation, a chemical modification of the lysine groups, has been used to develop low molecular weight allergoids that can be easily absorbed by the mucosa. A sublingual allergoid produced using this method reduced the need for antihistamines during the pollination season and maintained its clinical benefits for at least two years [106,107]. Another example is the HDM-monomeric allergoid SLIT, which has shown promise in improving rhinitis severity and reducing drug intake in phase II research [102,104]. These findings suggest that allergoids may offer a viable treatment option for respiratory allergies, with potential advantages over traditional allergen extracts.
Head-to-head comparisons between allergoids and conventional allergen extracts are limited, making it difficult to directly assess their relative efficacy. Quantification of modified allergens can be complex, leading to batch-to-batch variation and hindering comparisons of hypoallergenic effects between allergoids and whole allergens, both in vitro and in vivo [17,108].

6.2. Recombinant Allergens

Molecular diagnostics and therapeutics have significantly advanced the field of allergy medicine, enabling the identification of individual allergic components. This has paved the way for more precise allergy diagnosis and treatment, including the potential for personalized “tailor-made” allergen immunotherapy [109,110,111].
Molecular diagnosis also helps assess the clinical relevance of allergen sensitization profiles, particularly in cases of cross-reactivity between pollens and certain foods or different pollen types. By differentiating between true significant sensitization and pan-allergen sensitization, clinicians can select the most appropriate allergens for inclusion in immunotherapy, optimizing treatment outcomes for patients with complex sensitization profiles [109,110,111].
The potential of recombinant allergens to improve the safety and efficacy of AIT has been explored. The first cloning of allergen cDNAs was attempted in the late 1980s, and the first clinical trial using recombinant hypoallergens was conducted in the early 2000s [16,112,113].
A phase 2 trial showed that a mixture of six-grass pollen recombinant allergens was effective in treating seasonal allergic rhinitis. This treatment was associated with increases in allergen-specific IgG but not specific IgE, unlike whole allergen extract immunotherapy [114]. A phase 3 randomized controlled trial of recombinant birch allergen Bet v 1 demonstrated a 50% improvement in medication and symptom scores in patients with seasonal allergic rhinitis. While effective, it did not show significant differences in efficacy or side effects compared to natural protein or whole Bet v 1 extract [115].
Recombinant allergens offer several advantages, including standardization, genetic modification to deliver specific IgE-binding and T-cell epitopes, and an almost unlimited supply of purified allergen [14,16]. However, there are also potential disadvantages. One limitation of recombinant proteins is that they expose patients to only one or a few allergen molecules, unlike natural extracts that contain multiple allergens. While targeting a single allergen component may be effective for some patients, those sensitized to multiple components may benefit less from this approach. Additionally, recombinant allergens were initially thought to be safer due to their potential to stimulate only T or B cells, avoiding mast cell activation and related adverse events. However, clinical trials have reported adverse allergic reactions, including anaphylaxis, even hours after administration [16,112].
Despite showing promise in phase 2 trials, recombinant vaccines and hypoallergenic variants have not yet demonstrated significant improvements in efficacy or safety compared to current standardized allergen extracts. Further research is necessary to fully assess their potential benefits and address their limitations. With increasing knowledge of individual sensitization profiles and the role of major allergen epitopes, recombinant approaches may become more significant in the future of AIT [16].

6.3. T-Cell Peptides

T-cell epitopes are short, linear sequences of amino acids recognized by T-cell receptors. Unlike whole allergens, which can trigger IgE-mediated allergic reactions, T-cell epitopes do not induce IgE-mediated responses. When presented without co-stimulation, peptides can induce T-cell unresponsiveness (anergy) to the whole allergen. Additionally, peptides may induce tolerance by eliminating pathogenic allergen-specific T cells or altering the dominant T-cell phenotype towards Treg cells [116,117,118].
While peptide-based vaccines may have limited impact on humoral antibody responses compared to whole allergen immunotherapy, they offer potential advantages in terms of safety and the ability to modulate T-cell responses. However, it is important to note that while minimizing the risk of IgE-mediated anaphylaxis, peptide-based therapies may still induce T-cell-dependent late-phase asthmatic responses [118].
Several phase 2 studies have evaluated the efficacy of T-cell peptide immunotherapy. Initial studies on T-cell peptide immunotherapy showed promise, with a clinical trial demonstrating reduced rhinoconjunctivitis symptoms in cat-allergic patients treated with a mixture of seven Fel d 1 peptides [119]. However, larger phase 3 field trials involving Fel d 1-derived T-cell peptides (ClinicalTrials.gov, NCT01620762) failed to demonstrate efficacy, leading to the discontinuation of further development for these peptides. These setbacks may be attributed to factors such as a high placebo response in the control group and the inclusion of cat owners who might already have some degree of tolerance due to ongoing exposure to the allergen [2].
Contiguous overlapping peptides are longer sequences of amino acids that encompass a broader range of T-cell epitopes while disrupting IgE epitopes. This approach aims to target T-cell responses without triggering allergic reactions. A phase 2b dose-finding study of birch pollen contiguous overlapping peptides demonstrated a modest but statistically significant treatment effect (7%) over placebo at the highest dose [120,121].
Hydrolyzing whole allergens into medium-chain length peptides can reduce their allergenicity while preserving their ability to induce T- and B-cell responses. This approach may offer a broader range of epitopes compared to synthetic allergen sources [118].
A large clinical trial found that hydrolyzed rye grass allergen peptides administered in a short-term pre-seasonal regimen resulted in a significant reduction in allergy symptoms compared to placebo. The side effects associated with this approach were similar to those observed with conventional SCIT, suggesting that the primary benefit lies in the shortened treatment course [122,123].

6.4. B-Cell Peptides

B-cell peptide immunotherapy aims to induce protective humoral antibody responses without stimulating IgE production. One approach involves developing non-IgE-reactive peptides and conjugating them with a carrier protein unrelated to the allergen. This strategy exploits alternative T-helper responses, facilitated by the carrier protein, to induce protective allergen-specific IgG responses while preventing IgE stimulation [17,124].
A placebo-controlled field study involving 181 participants evaluated a mixture of recombinant non-IgE-reactive linear peptides (BM32) derived from grass pollen allergens, fused to a carrier protein (Pre-S protein). The study showed increased levels of allergen-specific IgG1 and IgG4 antibodies with minimal changes in IgE levels. While the primary analysis of combined seasonal symptom medication scores did not reach statistical significance, improvements were observed in asthma symptom scores and quality of life [125,126,127]. The results of phase 3 trials are eagerly awaited to further evaluate the efficacy and safety of this B-cell peptide immunotherapy approach.

6.5. DNA-Based Vaccine

DNA-based vaccines have shown promise in mouse models of allergy, inducing preferential TH1 and Treg cell responses while downregulating TH2 cell responses. The repeated use of DNA-based vaccines in humans has raised concerns, including the theoretical risk of plasmid DNA integration into the human genome, the development of anti-DNA antibodies, and the potential for long-term allergen persistence, which could lead to severe allergic reactions [2,16].
A clinical study evaluated a DNA-based vaccine targeting Cry j 2, a major allergen of Japanese cedar pollen allergy. The vaccine incorporated lysosomal-associated membrane protein 1 (LAMP1), a protein that directs the plasmid to the lysosomal compartment, reducing the risk of allergen release and anaphylaxis. After four intramuscular injections, 10 out of 12 participants showed a reduced immediate skin test response to Cry j 2 at 4 months. However, the study did not assess other clinical outcomes [128].
Another approach has involved combining allergens with bacterial DNA sequences containing CpG motifs, which are recognized by TLR-9, a receptor predominantly expressed on human B cells and dendritic cells. A phase 2 trial using a ragweed allergen (Amb a 1) covalently linked to a B-type CpG-containing oligodeoxynucleotide (ODN) showed promising results in ragweed pollen-induced hay fever [86]. However, these findings were not confirmed in a larger phase 3 trial, leading to the discontinuation of this approach [2].
Given their remarkable effectiveness against SARS-CoV-2 during the COVID-19 pandemic, mRNA-based vaccines have garnered significant interest for their potential applications in various medical fields, including allergic diseases [2,16]. Preclinical studies have demonstrated their ability to induce type 1 immune deviation and suppress allergic inflammation in mouse models of allergy [129].

7. Passive Immunotherapy

In 1935, Cooke et al. demonstrated that passive transfer of serum from ragweed immunotherapy patients could confer localized protection against ragweed skin prick test reactions in passively sensitized individuals [130]. This passive transfer of immunity was attributed to the transfer of blocking antibodies.
Subsequent studies in cat allergy patients have further validated this concept [131]. A single subcutaneous injection of a mixture of two recombinant anti-Fel d 1 antibodies conferred protection against nasal challenge with whole cat allergen extract for nearly 3 months, accompanied by a reduction in nasal fluid TH2-type cytokines and an increase in serum and nasal IgE-blocking activity [132].
A similar approach has been used to treat seasonal birch pollen allergy, where a cocktail of three monoclonal antibodies targeting the major birch allergen Bet v 1 effectively inhibited the clinical response to birch pollen nasal challenge for at least 2 months [133].
These findings suggest that passive immunotherapy, utilizing monoclonal antibodies targeting specific allergens, may offer a potential treatment option for allergic diseases, particularly for short-term protection or as a bridge therapy during periods of allergen exposure.

8. AIT Combination with Biologics

The combination of AIT with biologics, such as monoclonal antibodies targeting specific immune pathways, has emerged as a promising strategy for improving the management of allergic diseases. By combining the disease-modifying effects of AIT with the targeted therapeutic actions of biologics, this approach aims to enhance efficacy and safety while addressing specific challenges associated with AIT [134].
Omalizumab, a humanized monoclonal antibody, targets IgE, preventing it from binding to its receptors and triggering allergic reactions. It is currently approved for the treatment of moderate to severe asthma, chronic rhinosinusitis with nasal polyps, and chronic spontaneous urticaria. When used concurrently with AIT, omalizumab can lessen AIT-associated side effects, increasing tolerability. This allows patients to receive higher doses of allergen more quickly, making AIT suitable for higher-risk patients with asthma and enabling the use of rush protocols [135]. The combination of AIT with omalizumab has demonstrated efficacy in various allergens, including birch, grass, ragweed, perennial allergens, cat, dog, and HDM. Studies have shown a significant reduction in symptom scores, up to 48% less than SCIT alone, with a decrease in rescue medication use during seasonal exposure [136,137].
While the combination of grass pollen subcutaneous immunotherapy and dupilumab, an anti-IL-4 receptor antibody, reduced circulating IL-4-expressing TH2 cells, it did not significantly alter the magnitude or duration of allergen-induced late skin responses compared to allergen immunotherapy alone [138]. A clinical trial (NCT04502966) is currently evaluating the combination of grass pollen allergen with dupilumab, targeting both IL-4-dependent and IL-13-dependent pathways, in the context of inhalant immunotherapy [134].
Tezepelumab, a humanized monoclonal antibody targeting thymic stromal lymphopoietin (TSLP), has demonstrated promise in reducing eosinophilic inflammation, airway hyperresponsiveness, and asthma exacerbations. Additionally, it decreases serum IL-5, IL-13, and total IgE, suggesting potential synergy with AIT. A recent clinical trial investigated the combination of cat dander SCIT with tezepelumab. The study found that tezepelumab combined with SCIT was more effective than SCIT alone at reducing nasal response to cat allergen challenge after 52 weeks. This suppression persisted even one year after stopping treatment, suggesting a potential long-term benefit [139]. These findings suggest that tezepelumab may enhance the effectiveness of AIT by promoting long-term allergen tolerance [134].
Combining AIT with biologics has shown promise in improving short-term efficacy, enhancing safety, and improving patient tolerance during dose escalation. However, further research is needed to assess the long-term benefits and cost-effectiveness of these combination therapies.

9. Conclusions

AIT, including SLIT and SCIT, remains a safe and effective treatment for respiratory allergies. While novel approaches, such as the use of adjuvants, recombinant allergens, and biologics, show promise, their clinical efficacy and safety profiles still require further investigation.
Key findings and future directions include:
Traditional AIT: SCIT and SLIT continue to be the primary options for AIT, offering long-term symptom relief and the potential to prevent progression to asthma;
Novel Approaches: While modified allergens combined with novel adjuvants have shown promise in preclinical and early phase clinical trials, their superiority over traditional AIT has not been definitively established;
Personalized Immunotherapy: The development of personalized allergen immunotherapy based on individual sensitization profiles and the identification of specific epitopes, recombinant allergen, offers potential for improved efficacy and safety;
Combination Therapies: Combining AIT with biologics may enhance safety, treatment outcomes and address specific challenges associated with AIT;
Passive Immunotherapy: Recent studies suggest that passive immunotherapy using monoclonal antibodies may offer a viable option for short-term protection against allergic symptoms.
Overall, while AIT remains a valuable treatment option for respiratory allergies, ongoing research is essential to further refine existing approaches and develop novel strategies that can improve efficacy, safety, and patient outcomes.

Funding

This research received no external funding.

Data Availability Statement

The datasets generated during the current study are available from the corresponding author upon reasonable request.

Conflicts of Interest

M.L. has received honoraria for scientific lectures from Abbott, A. Menarini, AstraZeneca, GSK, Takeda, Viatris, Organon, Sanofi, Novartis, and research supports from Amgen.

Abbreviations

AITallergen immunotherapy
APCantigen-presenting cell
ARallergic rhinitis
BregB regulatory cell
CpGCytosine-phosphodiester-guanine
DCdendritic cell
DCregdendritic cell-derived regulatory cell
EPITepicutaneous immunotherapy
HCPhealth care personnel
HDMhouse dust mite
IgImmunoglobulin
IDITIntradermal Immunotherapy
ILinterleukin
ILC2type 2 innate lymphoid cell
ILCreginnate lymphoid cell-derived regulatory cell
ILITIntralymphatic immunotherapy
LAMP1lysosomal-associated membrane protein 1
MCTmicrocrystalline tyrosine
MPLAMonophosphoryl lipid A
ODNoligodeoxynucleotide
pDCplasmacytoid dendritic cell
SCITsubcutaneous immunotherapy
SLITsublingual immunotherapy
TfhT-helper cell
TfrT follicular regulatory cell
TGFtransforming growth factor
TH1T-helper 1 cell
TH2T-helper 2 cell
TLRToll-like receptor
TregT regulatory cells
TSLPthymic stromal lymphopoietin
VLPvirus-like particle

References

  1. Gutermuth, J.; Grosber, M.; Pfaar, O.; Bergmann, K.C.; Ring, J. 111 years of allergen-immunotherapy: A long and successful history of the only available disease-modifier in allergic diseases. Allergol. Select 2022, 6, 248–258. [Google Scholar] [CrossRef] [PubMed]
  2. Durham, S.R.; Shamji, M.H. Allergen immunotherapy: Past, present and future. Nat. Rev. Immunol. 2023, 23, 317–328. [Google Scholar] [CrossRef] [PubMed]
  3. Noon, L. Prophylactic inoculation against hay fever (historical document). Ann. Allergy 1955, 13, 713–716. [Google Scholar] [PubMed]
  4. Frankland, A.W.; Augustin, R. Prophylaxis of summer hay-fever and asthma: A controlled trial comparing crude grass-pollen extracts with the isolated main protein component. Lancet 1954, 266, 1055–1057. [Google Scholar] [CrossRef] [PubMed]
  5. Lowell, F.C.; Franklin, W. A double-blind study of the effectiveness and specificity of injection therapy in ragweed hay fever. N. Engl. J. Med. 1965, 273, 675–679. [Google Scholar] [CrossRef]
  6. Norman, P.S.; Lichtenstein, L.M. The clinical and immunologic specificity of immunotherapy. J. Allergy Clin. Immunol. 1978, 61, 370–377. [Google Scholar] [CrossRef]
  7. Hunt, K.J.; Valentine, M.D.; Sobotka, A.K.; Benton, A.W.; Amodio, F.J.; Lichtenstein, L.M. A controlled trial of immunotherapy in insect hypersensitivity. N. Engl. J. Med. 1978, 299, 157–161. [Google Scholar] [CrossRef]
  8. Cox, L.; Nelson, H.; Lockey, R.; Calabria, C.; Chacko, T.; Finegold, I.; Nelson, M.; Weber, R.; Bernstein, D.I.; Blessing-Moore, J.; et al. Allergen immunotherapy: A practice parameter third update. J. Allergy Clin. Immunol. 2011, 127, S1–S55. [Google Scholar] [CrossRef]
  9. Agache, I.; Lau, S.; Akdis, C.A.; Smolinska, S.; Bonini, M.; Cavkaytar, O.; Flood, B.; Gajdanowicz, P.; Izuhara, K.; Kalayci, O.; et al. EAACI Guidelines on Allergen Immunotherapy: House dust mite-driven allergic asthma. Allergy 2019, 74, 855–873. [Google Scholar] [CrossRef]
  10. Roberts, G.; Pfaar, O.; Akdis, C.A.; Ansotegui, I.J.; Durham, S.R.; Gerth van Wijk, R.; Halken, S.; Larenas-Linnemann, D.; Pawankar, R.; Pitsios, C.; et al. EAACI Guidelines on Allergen Immunotherapy: Allergic rhinoconjunctivitis. Allergy 2018, 73, 765–798. [Google Scholar] [CrossRef]
  11. Arshad, H.; Lack, G.; Durham, S.R.; Penagos, M.; Larenas-Linnemann, D.; Halken, S. Prevention Is Better than Cure: Impact of Allergen Immunotherapy on the Progression of Airway Disease. J. Allergy Clin. Immunol. Pract. 2024, 12, 45–56. [Google Scholar] [CrossRef] [PubMed]
  12. Larenas-Linnemann, D. Long-term adherence strategies for allergen immunotherapy. Allergy Asthma Proc. 2022, 43, 299–304. [Google Scholar] [CrossRef] [PubMed]
  13. Bonertz, A.; Roberts, G.C.; Hoefnagel, M.; Timon, M.; Slater, J.E.; Rabin, R.L.; Bridgewater, J.; Pini, C.; Pfaar, O.; Akdis, C.; et al. Challenges in the implementation of EAACI guidelines on allergen immunotherapy: A global perspective on the regulation of allergen products. Allergy 2018, 73, 64–76. [Google Scholar] [CrossRef] [PubMed]
  14. Pavon-Romero, G.F.; Parra-Vargas, M.I.; Ramirez-Jimenez, F.; Melgoza-Ruiz, E.; Serrano-Perez, N.H.; Teran, L.M. Allergen Immunotherapy: Current and Future Trends. Cells 2022, 11, 212. [Google Scholar] [CrossRef] [PubMed]
  15. Zemelka-Wiacek, M.; Agache, I.; Akdis, C.A.; Akdis, M.; Casale, T.B.; Dramburg, S.; Jahnz-Rozyk, K.; Kosowska, A.; Matricardi, P.M.; Pfaar, O.; et al. Hot topics in allergen immunotherapy, 2023: Current status and future perspective. Allergy 2024, 79, 823–842. [Google Scholar] [CrossRef]
  16. Pfaar, O.; Portnoy, J.; Nolte, H.; Chaker, A.M.; Luna-Pech, J.A.; Patterson, A.; Pandya, A.; Larenas-Linnemann, D. Future Directions of Allergen Immunotherapy for Allergic Rhinitis: Experts’ Perspective. J. Allergy Clin. Immunol. Pract. 2024, 12, 32–44. [Google Scholar] [CrossRef]
  17. Gunawardana, N.C.; Durham, S.R. New approaches to allergen immunotherapy. Ann. Allergy Asthma Immunol. 2018, 121, 293–305. [Google Scholar] [CrossRef]
  18. Sosic, L.; Paolucci, M.; Flory, S.; Jebbawi, F.; Kundig, T.M.; Johansen, P. Allergen immunotherapy: Progress and future outlook. Expert. Rev. Clin. Immunol. 2023, 19, 745–769. [Google Scholar] [CrossRef]
  19. Licona-Limon, P.; Kim, L.K.; Palm, N.W.; Flavell, R.A. TH2, allergy and group 2 innate lymphoid cells. Nat. Immunol. 2013, 14, 536–542. [Google Scholar] [CrossRef]
  20. Rondon, C.; Campo, P.; Togias, A.; Fokkens, W.J.; Durham, S.R.; Powe, D.G.; Mullol, J.; Blanca, M. Local allergic rhinitis: Concept, pathophysiology, and management. J. Allergy Clin. Immunol. 2012, 129, 1460–1467. [Google Scholar] [CrossRef]
  21. Rosenwasser, L.J. Current understanding of the pathophysiology of allergic rhinitis. Immunol. Allergy Clin. N. Am. 2011, 31, 433–439. [Google Scholar] [CrossRef] [PubMed]
  22. Shamji, M.H.; Durham, S.R. Mechanisms of allergen immunotherapy for inhaled allergens and predictive biomarkers. J. Allergy Clin. Immunol. 2017, 140, 1485–1498. [Google Scholar] [CrossRef] [PubMed]
  23. Layhadi, J.A.; Lalioti, A.; Palmer, E.; van Zelm, M.C.; Wambre, E.; Shamji, M.H. Mechanisms and Predictive Biomarkers of Allergen Immunotherapy in the Clinic. J. Allergy Clin. Immunol. Pract. 2024, 12, 59–66. [Google Scholar] [CrossRef] [PubMed]
  24. Ling, E.M.; Smith, T.; Nguyen, X.D.; Pridgeon, C.; Dallman, M.; Arbery, J.; Carr, V.A.; Robinson, D.S. Relation of CD4+CD25+ regulatory T-cell suppression of allergen-driven T-cell activation to atopic status and expression of allergic disease. Lancet 2004, 363, 608–615. [Google Scholar] [CrossRef] [PubMed]
  25. Boonpiyathad, T.; Sozener, Z.C.; Akdis, M.; Akdis, C.A. The role of Treg cell subsets in allergic disease. Asian Pac. J. Allergy Immunol. 2020, 38, 139–149. [Google Scholar] [CrossRef]
  26. Sharif, H.; Acharya, S.; Dhondalay, G.K.R.; Varricchi, G.; Krasner-Macleod, S.; Laisuan, W.; Switzer, A.; Lenormand, M.; Kashe, E.; Parkin, R.V.; et al. 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. [Google Scholar] [CrossRef]
  27. Zimmer, A.; Bouley, J.; Le Mignon, M.; Pliquet, E.; Horiot, S.; Turfkruyer, M.; Baron-Bodo, V.; Horak, F.; Nony, E.; Louise, A.; et al. A regulatory dendritic cell signature correlates with the clinical efficacy of allergen-specific sublingual immunotherapy. J. Allergy Clin. Immunol. 2012, 129, 1020–1030. [Google Scholar] [CrossRef]
  28. Golebski, K.; Layhadi, J.A.; Sahiner, U.; Steveling-Klein, E.H.; Lenormand, M.M.; Li, R.C.Y.; Bal, S.M.; Heesters, B.A.; Vila-Nadal, G.; Hunewald, O.; et al. Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity 2021, 54, 291–307.e7. [Google Scholar] [CrossRef]
  29. Lao-Araya, M.; Steveling, E.; Scadding, G.W.; Durham, S.R.; Shamji, M.H. Seasonal increases in peripheral innate lymphoid type 2 cells are inhibited by subcutaneous grass pollen immunotherapy. J. Allergy Clin. Immunol. 2014, 134, 1193–1195.e4. [Google Scholar] [CrossRef]
  30. Boonpiyathad, T.; Tantilipikorn, P.; Ruxrungtham, K.; Pradubpongsa, P.; Mitthamsiri, W.; Piedvache, A.; Thantiworasit, P.; Sirivichayakul, S.; Jacquet, A.; Suratannon, N.; et al. IL-10-producing innate lymphoid cells increased in patients with house dust mite allergic rhinitis following immunotherapy. J. Allergy Clin. Immunol. 2021, 147, 1507–1510.e8. [Google Scholar] [CrossRef]
  31. 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, 707–713. [Google Scholar] [CrossRef] [PubMed]
  32. Gueguen, C.; Bouley, J.; Moussu, H.; Luce, S.; Duchateau, M.; Chamot-Rooke, J.; Pallardy, M.; Lombardi, V.; Nony, E.; Baron-Bodo, V.; et al. Changes in markers associated with dendritic cells driving the differentiation of either TH2 cells or regulatory T cells correlate with clinical benefit during allergen immunotherapy. J. Allergy Clin. Immunol. 2016, 137, 545–558. [Google Scholar] [CrossRef] [PubMed]
  33. Boonpiyathad, T.; van de Veen, W.; Wirz, O.; Sokolowska, M.; Ruckert, B.; Tan, G.; Sangasapaviliya, A.; Pradubpongsa, P.; Fuengthong, R.; Thantiworasit, P.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  34. Shamji, M.H.; Larson, D.; Eifan, A.; Scadding, G.W.; Qin, T.; Lawson, K.; Sever, M.L.; Macfarlane, E.; Layhadi, J.A.; Wurtzen, P.A.; et al. Differential induction of allergen-specific IgA responses following timothy grass subcutaneous and sublingual immunotherapy. J. Allergy Clin. Immunol. 2021, 148, 1061–1071.e11. [Google Scholar] [CrossRef] [PubMed]
  35. Shamji, M.H.; Kappen, J.; Abubakar-Waziri, H.; Zhang, J.; Steveling, E.; Watchman, S.; Kouser, L.; Eifan, A.; Switzer, A.; Varricchi, G.; et al. Nasal allergen-neutralizing IgG(4) antibodies block IgE-mediated responses: Novel biomarker of subcutaneous grass pollen immunotherapy. J. Allergy Clin. Immunol. 2019, 143, 1067–1076. [Google Scholar] [CrossRef]
  36. Scadding, G.W.; Calderon, M.A.; Shamji, M.H.; Eifan, A.O.; Penagos, M.; Dumitru, F.; Sever, M.L.; Bahnson, H.T.; Lawson, K.; Harris, K.M.; et al. 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. [Google Scholar] [CrossRef]
  37. Boonpiyathad, T.; Lao-Araya, M.; Chiewchalermsri, C.; Sangkanjanavanich, S.; Morita, H. Allergic Rhinitis: What Do We Know About Allergen-Specific Immunotherapy? Front. Allergy 2021, 2, 747323. [Google Scholar] [CrossRef]
  38. Calderon, M.A.; Vidal, C.; Rodriguez Del Rio, P.; Just, J.; Pfaar, O.; Tabar, A.I.; Sanchez-Machin, I.; Bubel, P.; Borja, J.; Eberle, P.; et al. European Survey on Adverse Systemic Reactions in Allergen Immunotherapy (EASSI): A real-life clinical assessment. Allergy 2017, 72, 462–472. [Google Scholar] [CrossRef]
  39. Asllani, J.; Mitsias, D.; Konstantinou, G.; Priftanji, A.; Hoxha, M.; Sinani, G.; Christoff, G.; Zlatko, D.; Makris, M.; Aggelidis, X.; et al. The Allergen Immunotherapy Adverse Events Registry: Setup & methodology of a European Academy of Allergy and Clinical Immunology taskforce project. Clin. Transl. Allergy 2023, 13, e12266. [Google Scholar] [CrossRef]
  40. Bernstein, D.I.; Epstein, T.E.G. Safety of allergen immunotherapy in North America from 2008-2017: Lessons learned from the ACAAI/AAAAI National Surveillance Study of adverse reactions to allergen immunotherapy. Allergy Asthma Proc. 2020, 41, 108–111. [Google Scholar] [CrossRef]
  41. Dhami, S.; Nurmatov, U.; Arasi, S.; Khan, T.; Asaria, M.; Zaman, H.; Agarwal, A.; Netuveli, G.; Roberts, G.; Pfaar, O.; et al. Allergen immunotherapy for allergic rhinoconjunctivitis: A systematic review and meta-analysis. Allergy 2017, 72, 1597–1631. [Google Scholar] [CrossRef] [PubMed]
  42. Canonica, G.W.; Baena-Cagnani, C.E.; Bousquet, J.; Bousquet, P.J.; Lockey, R.F.; Malling, H.J.; Passalacqua, G.; Potter, P.; Valovirta, E. Recommendations for standardization of clinical trials with Allergen Specific Immunotherapy for respiratory allergy. A statement of a World Allergy Organization (WAO) taskforce. Allergy 2007, 62, 317–324. [Google Scholar] [CrossRef] [PubMed]
  43. Durham, S.R.; Penagos, M. Sublingual or subcutaneous immunotherapy for allergic rhinitis? J. Allergy Clin. Immunol. 2016, 137, 339–349.e0. [Google Scholar] [CrossRef] [PubMed]
  44. Penagos, M.; Durham, S.R. Allergen immunotherapy for long-term tolerance and prevention. J. Allergy Clin. Immunol. 2022, 149, 802–811. [Google Scholar] [CrossRef]
  45. Conroy, E.R.; Banzon, T.M.; Simoneau, T.; Phipatanakul, W.; van Boven, J.F.M.; Larenas-Linnemann, D. An Overview of Adherence-What It Is and Why It Is Important. J. Allergy Clin. Immunol. Pract. 2024; in press. [Google Scholar] [CrossRef]
  46. Kulalert, P.; Phinyo, P.; Lao-Araya, M. Efficacy and safety of house dust mite sublingual immunotherapy tablets in allergic rhinitis: A systematic review and meta-analysis. World Allergy Organ. J. 2022, 15, 100691. [Google Scholar] [CrossRef]
  47. Bergmann, K.C.; Demoly, P.; Worm, M.; Fokkens, W.J.; Carrillo, T.; Tabar, A.I.; Nguyen, H.; Montagut, A.; Zeldin, R.K. Efficacy and safety of sublingual tablets of house dust mite allergen extracts in adults with allergic rhinitis. J. Allergy Clin. Immunol. 2014, 133, 1608–1614.e6. [Google Scholar] [CrossRef]
  48. Demoly, P.; Corren, J.; Creticos, P.; De Blay, F.; Gevaert, P.; Hellings, P.; Kowal, K.; Le Gall, M.; Nenasheva, N.; Passalacqua, G.; et al. A 300 IR sublingual tablet is an effective, safe treatment for house dust mite-induced allergic rhinitis: An international, double-blind, placebo-controlled, randomized phase III clinical trial. J. Allergy Clin. Immunol. 2021, 147, 1020–1030.e10. [Google Scholar] [CrossRef]
  49. Durham, S.R.; Emminger, W.; Kapp, A.; de Monchy, J.G.; Rak, S.; Scadding, G.K.; Wurtzen, P.A.; Andersen, J.S.; Tholstrup, B.; Riis, B.; et al. SQ-standardized sublingual grass immunotherapy: Confirmation of disease modification 2 years after 3 years of treatment in a randomized trial. J. Allergy Clin. Immunol. 2012, 129, 717–725.e5. [Google Scholar] [CrossRef]
  50. Creticos, P.S.; Maloney, J.; Bernstein, D.I.; Casale, T.; Kaur, A.; Fisher, R.; Liu, N.; Murphy, K.; Nekam, K.; Nolte, H. Randomized controlled trial of a ragweed allergy immunotherapy tablet in North American and European adults. J. Allergy Clin. Immunol. 2013, 131, 1342–1349e6. [Google Scholar] [CrossRef]
  51. Yonekura, S.; Gotoh, M.; Kaneko, S.; Maekawa, Y.; Okubo, K.; Okamoto, Y. Disease-Modifying Effect of Japanese Cedar Pollen Sublingual Immunotherapy Tablets. J. Allergy Clin. Immunol. Pract. 2021, 9, 4103–4116.e4. [Google Scholar] [CrossRef] [PubMed]
  52. Okamoto, Y.; Okubo, K.; Yonekura, S.; Hashiguchi, K.; Goto, M.; Otsuka, T.; Murata, T.; Nakao, Y.; Kanazawa, C.; Nagakura, H.; et al. Efficacy and safety of sublingual immunotherapy for two seasons in patients with Japanese cedar pollinosis. Int. Arch. Allergy Immunol. 2015, 166, 177–188. [Google Scholar] [CrossRef] [PubMed]
  53. Virchow, J.C.; Backer, V.; Kuna, P.; Prieto, L.; Nolte, H.; Villesen, H.H.; Ljorring, C.; Riis, B.; de Blay, F. Efficacy of a House Dust Mite Sublingual Allergen Immunotherapy Tablet in Adults With Allergic Asthma: A Randomized Clinical Trial. JAMA 2016, 315, 1715–1725. [Google Scholar] [CrossRef] [PubMed]
  54. Mosbech, H.; Deckelmann, R.; de Blay, F.; Pastorello, E.A.; Trebas-Pietras, E.; Andres, L.P.; Malcus, I.; Ljorring, C.; Canonica, G.W. Standardized quality (SQ) house dust mite sublingual immunotherapy tablet (ALK) reduces inhaled corticosteroid use while maintaining asthma control: A randomized, double-blind, placebo-controlled trial. J. Allergy Clin. Immunol. 2014, 134, 568–575.e7. [Google Scholar] [CrossRef] [PubMed]
  55. Horak, F.; Jaeger, S.; Worm, M.; Melac, M.; Didier, A. Implementation of pre-seasonal sublingual immunotherapy with a five-grass pollen tablet during optimal dosage assessment. Clin. Exp. Allergy 2009, 39, 394–400. [Google Scholar] [CrossRef]
  56. Greenhawt, M.; Oppenheimer, J.; Nelson, M.; Nelson, H.; Lockey, R.; Lieberman, P.; Nowak-Wegrzyn, A.; Peters, A.; Collins, C.; Bernstein, D.I.; et al. Sublingual immunotherapy: A focused allergen immunotherapy practice parameter update. Ann. Allergy Asthma Immunol. 2017, 118, 276–282.e2. [Google Scholar] [CrossRef]
  57. Lao-Araya, M.; Sompornrattanaphan, M.; Kanjanawasee, D.; Tantilipikorn, P.; Allergy Asthma and Immunology Association of Thailand (AAIAT) interesting group on immunotherapy. Allergen immunotherapy for respiratory allergies in clinical practice: A comprehensive review. Asian Pac. J. Allergy Immunol. 2022, 40, 283–294. [Google Scholar] [CrossRef]
  58. Durham, S.R.; Creticos, P.S.; Nelson, H.S.; Li, Z.; Kaur, A.; Meltzer, E.O.; Nolte, H. Treatment effect of sublingual immunotherapy tablets and pharmacotherapies for seasonal and perennial allergic rhinitis: Pooled analyses. J. Allergy Clin. Immunol. 2016, 138, 1081–1088.e4. [Google Scholar] [CrossRef]
  59. Kennedy, J.L.; Robinson, D.; Christophel, J.; Borish, L.; Payne, S. Decision-making analysis for allergen immunotherapy versus nasal steroids in the treatment of nasal steroid-responsive allergic rhinitis. Am. J. Rhinol. Allergy 2014, 28, 59–64. [Google Scholar] [CrossRef]
  60. Settipane, R.A.; Bukstein, D.A. Allergen immunotherapy and shared decision-making. Allergy Asthma Proc. 2022, 43, 350–355. [Google Scholar] [CrossRef]
  61. Senti, G.; Prinz Vavricka, B.M.; Erdmann, I.; Diaz, M.I.; Markus, R.; McCormack, S.J.; Simard, J.J.; Wuthrich, B.; Crameri, R.; Graf, N.; et al. Intralymphatic allergen administration renders specific immunotherapy faster and safer: A randomized controlled trial. Proc. Natl. Acad. Sci. USA 2008, 105, 17908–17912. [Google Scholar] [CrossRef]
  62. Senti, G.; Crameri, R.; Kuster, D.; Johansen, P.; Martinez-Gomez, J.M.; Graf, N.; Steiner, M.; Hothorn, L.A.; Gronlund, H.; Tivig, C.; et al. Intralymphatic immunotherapy for cat allergy induces tolerance after only 3 injections. J. Allergy Clin. Immunol. 2012, 129, 1290–1296. [Google Scholar] [CrossRef] [PubMed]
  63. Chabot, A.; Senti, G.; Erdmann, I.; Prinz, B.M.; Wuthrich, B.; Sosic, L.; Kundig, T.M.; Johansen, P. Intralymphatic Immunotherapy (ILIT) With Bee Venom Allergens: A Clinical Proof-of-Concept Study and the Very First ILIT in Humans. Front. Allergy 2022, 3, 832010. [Google Scholar] [CrossRef] [PubMed]
  64. Senti, G.; Freiburghaus, A.U.; Larenas-Linnemann, D.; Hoffmann, H.J.; Patterson, A.M.; Klimek, L.; Di Bona, D.; Pfaar, O.; Ahlbeck, L.; Akdis, M.; et al. Intralymphatic Immunotherapy: Update and Unmet Needs. Int. Arch. Allergy Immunol. 2019, 178, 141–149. [Google Scholar] [CrossRef] [PubMed]
  65. Konradsen, J.R.; Grundstrom, J.; Hellkvist, L.; Tran, T.A.T.; Andersson, N.; Gafvelin, G.; Kiewiet, M.B.G.; Hamsten, C.; Tang, J.; Parkin, R.V.; et al. Intralymphatic immunotherapy in pollen-allergic young adults with rhinoconjunctivitis and mild asthma: A randomized trial. J. Allergy Clin. Immunol. 2020, 145, 1005–1007.e7. [Google Scholar] [CrossRef] [PubMed]
  66. Skaarup, S.H.; Schmid, J.M.; Skjold, T.; Graumann, O.; Hoffmann, H.J. Intralymphatic immunotherapy improves grass pollen allergic rhinoconjunctivitis: A 3-year randomized placebo-controlled trial. J. Allergy Clin. Immunol. 2021, 147, 1011–1019. [Google Scholar] [CrossRef]
  67. Hoang, M.P.; Seresirikachorn, K.; Chitsuthipakorn, W.; Snidvongs, K. Intralymphatic immunotherapy for allergic rhinoconjunctivitis: A systematic review and meta-analysis. Rhinology 2021, 59, 236–244. [Google Scholar] [CrossRef]
  68. Senti, G.; von Moos, S.; Kundig, T.M. Epicutaneous allergen administration: Is this the future of allergen-specific immunotherapy? Allergy 2011, 66, 798–809. [Google Scholar] [CrossRef]
  69. Senti, G.; Graf, N.; Haug, S.; Ruedi, N.; von Moos, S.; Sonderegger, T.; Johansen, P.; Kundig, T.M. Epicutaneous allergen administration as a novel method of allergen-specific immunotherapy. J. Allergy Clin. Immunol. 2009, 124, 997–1002. [Google Scholar] [CrossRef]
  70. Senti, G.; von Moos, S.; Tay, F.; Graf, N.; Johansen, P.; Kundig, T.M. Determinants of efficacy and safety in epicutaneous allergen immunotherapy: Summary of three clinical trials. Allergy 2015, 70, 707–710. [Google Scholar] [CrossRef]
  71. Senti, G.; Kundig, T.M. Novel Delivery Routes for Allergy Immunotherapy: Intralymphatic, Epicutaneous, and Intradermal. Immunol. Allergy Clin. N. Am. 2016, 36, 25–37. [Google Scholar] [CrossRef] [PubMed]
  72. Rotiroti, G.; Shamji, M.; Durham, S.R.; Till, S.J. Repeated low-dose intradermal allergen injection suppresses allergen-induced cutaneous late responses. J. Allergy Clin. Immunol. 2012, 130, 918–924.e1. [Google Scholar] [CrossRef] [PubMed]
  73. Slovick, A.; Douiri, A.; Muir, R.; Guerra, A.; Tsioulos, K.; Hay, E.; Lam, E.P.S.; Kelly, J.; Peacock, J.L.; Ying, S.; et al. Intradermal grass pollen immunotherapy increases T(H)2 and IgE responses and worsens respiratory allergic symptoms. J. Allergy Clin. Immunol. 2017, 139, 1830–1839.e13. [Google Scholar] [CrossRef] [PubMed]
  74. Lin, Y.J.; Zimmermann, J.; Schulke, S. Novel adjuvants in allergen-specific immunotherapy: Where do we stand? Front. Immunol. 2024, 15, 1348305. [Google Scholar] [CrossRef] [PubMed]
  75. Norman, P.S.; Lichtenstein, L.M. Comparisons of alum-precipitated and unprecipitated aqueous ragweed pollen extracts in the treatment of hay fever. J. Allergy Clin. Immunol. 1978, 61, 384–389. [Google Scholar] [CrossRef]
  76. Principi, N.; Esposito, S. Aluminum in vaccines: Does it create a safety problem? Vaccine 2018, 36, 5825–5831. [Google Scholar] [CrossRef]
  77. Baldrick, P.; Richardson, D.; Wheeler, A.W. Review of L-tyrosine confirming its safe human use as an adjuvant. J. Appl. Toxicol. 2002, 22, 333–344. [Google Scholar] [CrossRef]
  78. Wheeler, A.W.; Moran, D.M.; Robins, B.E.; Driscoll, A. l-Tyrosine as an immunological adjuvant. Int. Arch. Allergy Appl. Immunol. 1982, 69, 113–119. [Google Scholar] [CrossRef]
  79. Becker, S.; Zieglmayer, P.; Canto, G.; Fassio, F.; Yong, P.; Acikel, C.; Raskopf, E.; Steveling-Klein, E.H.; Allekotte, S.; Mosges, R. A meta-analysis on allergen-specific immunotherapy using MCT((R)) (MicroCrystalline Tyrosine)-adsorbed allergoids in pollen allergic patients suffering from allergic rhinoconjunctivitis. Clin. Transl. Allergy 2021, 11, e12037. [Google Scholar] [CrossRef]
  80. Mothes, N.; Heinzkill, M.; Drachenberg, K.J.; Sperr, W.R.; Krauth, M.T.; Majlesi, Y.; Semper, H.; Valent, P.; Niederberger, V.; Kraft, D.; et al. Allergen-specific immunotherapy with a monophosphoryl lipid A-adjuvanted vaccine: Reduced seasonally boosted immunoglobulin E production and inhibition of basophil histamine release by therapy-induced blocking antibodies. Clin. Exp. Allergy 2003, 33, 1198–1208. [Google Scholar] [CrossRef]
  81. Drachenberg, K.J.; Wheeler, A.W.; Stuebner, P.; Horak, F. A well-tolerated grass pollen-specific allergy vaccine containing a novel adjuvant, monophosphoryl lipid A, reduces allergic symptoms after only four preseasonal injections. Allergy 2001, 56, 498–505. [Google Scholar] [CrossRef] [PubMed]
  82. Bell, A.J.; Heath, M.D.; Hewings, S.J.; Skinner, M.A. The adsorption of allergoids and 3-O-desacyl-4′-monophosphoryl lipid A (MPL(R)) to microcrystalline tyrosine (MCT) in formulations for use in allergy immunotherapy. J. Inorg. Biochem. 2015, 152, 147–153. [Google Scholar] [CrossRef] [PubMed]
  83. Jutek, M.; Bachert, C. Ultra-short-course seasonal allergy vaccine (Pollinex Quattro): Viewpoints. Drugs 2006, 66, 939–940. [Google Scholar] [CrossRef] [PubMed]
  84. Rosewich, M.; Lee, D.; Zielen, S. Pollinex Quattro: An innovative four injections immunotherapy in allergic rhinitis. Hum. Vaccin. Immunother. 2013, 9, 1523–1531. [Google Scholar] [CrossRef]
  85. Worm, M.; Higenbottam, T.; Pfaar, O.; Mosges, R.; Aberer, W.; Gunawardena, K.; Wessiepe, D.; Lee, D.; Kramer, M.F.; Skinner, M.; et al. Randomized controlled trials define shape of dose response for Pollinex Quattro Birch allergoid immunotherapy. Allergy 2018, 73, 1812–1822. [Google Scholar] [CrossRef]
  86. Creticos, P.S.; Schroeder, J.T.; Hamilton, R.G.; Balcer-Whaley, S.L.; Khattignavong, A.P.; Lindblad, R.; Li, H.; Coffman, R.; Seyfert, V.; Eiden, J.J.; et al. Immunotherapy with a ragweed-toll-like receptor 9 agonist vaccine for allergic rhinitis. N. Engl. J. Med. 2006, 355, 1445–1455. [Google Scholar] [CrossRef]
  87. Kawakita, A.; Shirasaki, H.; Yasutomi, M.; Tokuriki, S.; Mayumi, M.; Naiki, H.; Ohshima, Y. Immunotherapy with oligomannose-coated liposomes ameliorates allergic symptoms in a murine food allergy model. Allergy 2012, 67, 371–379. [Google Scholar] [CrossRef]
  88. Beeh, K.M.; Kanniess, F.; Wagner, F.; Schilder, C.; Naudts, I.; Hammann-Haenni, A.; Willers, J.; Stocker, H.; Mueller, P.; Bachmann, M.F.; et al. The novel TLR-9 agonist QbG10 shows clinical efficacy in persistent allergic asthma. J. Allergy Clin. Immunol. 2013, 131, 866–874. [Google Scholar] [CrossRef]
  89. Senti, G.; Johansen, P.; Haug, S.; Bull, C.; Gottschaller, C.; Muller, P.; Pfister, T.; Maurer, P.; Bachmann, M.F.; Graf, N.; et al. Use of A-type CpG oligodeoxynucleotides as an adjuvant in allergen-specific immunotherapy in humans: A phase I/IIa clinical trial. Clin. Exp. Allergy 2009, 39, 562–570. [Google Scholar] [CrossRef]
  90. Klimek, L.; Willers, J.; Hammann-Haenni, A.; Pfaar, O.; Stocker, H.; Mueller, P.; Renner, W.A.; Bachmann, M.F. Assessment of clinical efficacy of CYT003-QbG10 in patients with allergic rhinoconjunctivitis: A phase IIb study. Clin. Exp. Allergy 2011, 41, 1305–1312. [Google Scholar] [CrossRef]
  91. Casale, T.B.; Cole, J.; Beck, E.; Vogelmeier, C.F.; Willers, J.; Lassen, C.; Hammann-Haenni, A.; Trokan, L.; Saudan, P.; Wechsler, M.E. CYT003, a TLR9 agonist, in persistent allergic asthma—a randomized placebo-controlled Phase 2b study. Allergy 2015, 70, 1160–1168. [Google Scholar] [CrossRef] [PubMed]
  92. Marsh, D.G.; Lichtenstein, L.M.; Campbell, D.H. Studies on ”allergoids” prepared from naturally occurring allergens. I. Assay of allergenicity and antigenicity of formalinized rye group I component. Immunology 1970, 18, 705–722. [Google Scholar] [PubMed]
  93. Norman, P.S.; Lichtenstein, L.M.; Kagey-Sobotka, A.; Marsh, D.G. Controlled evaluation of allergoid in the immunotherapy of ragweed hay fever. J. Allergy Clin. Immunol. 1982, 70, 248–260. [Google Scholar] [CrossRef] [PubMed]
  94. Bousquet, J.; Braquemond, P.; Feinberg, J.; Guerin, B.; Maasch, H.; Michel, F.B. Specific IgE response before and after rush immunotherapy with a standardized allergen or allergoid in grass pollen allergy. Ann. Allergy 1986, 56, 456–459. [Google Scholar]
  95. Bozek, A.; Cudak, A.; Walter Canonica, G. Long-term efficacy of injected allergen immunotherapy for treatment of grass pollen allergy in elderly patients with allergic rhinitis. Allergy Asthma Proc. 2020, 41, 271–277. [Google Scholar] [CrossRef]
  96. Bousquet, J.; Hejjaoui, A.; Soussana, M.; Michel, F.B. Double-blind, placebo-controlled immunotherapy with mixed grass-pollen allergoids. IV. Comparison of the safety and efficacy of two dosages of a high-molecular-weight allergoid. J. Allergy Clin. Immunol. 1990, 85, 490–497. [Google Scholar] [CrossRef]
  97. Worm, M.; Rak, S.; Samolinski, B.; Antila, J.; Hoiby, A.S.; Kruse, B.; Lipiec, A.; Rudert, M.; Valovirta, E. Efficacy and safety of birch pollen allergoid subcutaneous immunotherapy: A 2-year double-blind, placebo-controlled, randomized trial plus 1-year open-label extension. Clin. Exp. Allergy 2019, 49, 516–525. [Google Scholar] [CrossRef]
  98. Lund, L.; Henmar, H.; Wurtzen, P.A.; Lund, G.; Hjortskov, N.; Larsen, J.N. Comparison of allergenicity and immunogenicity of an intact allergen vaccine and commercially available allergoid products for birch pollen immunotherapy. Clin. Exp. Allergy 2007, 37, 564–571. [Google Scholar] [CrossRef]
  99. Klimek, L.; Dormann, D.; Jarman, E.R.; Cromwell, O.; Riechelmann, H.; Reske-Kunz, A.B. Short-term preseasonal birch pollen allergoid immunotherapy influences symptoms, specific nasal provocation and cytokine levels in nasal secretions, but not peripheral T-cell responses, in patients with allergic rhinitis. Clin. Exp. Allergy 1999, 29, 1326–1335. [Google Scholar] [CrossRef]
  100. Klimek, L.; Fox, G.C.; Thum-Oltmer, S. SCIT with a high-dose house dust mite allergoid is well tolerated: Safety data from pooled clinical trials and more than 10 years of daily practice analyzed in different subgroups. Allergo J. Int. 2018, 27, 131–139. [Google Scholar] [CrossRef]
  101. Jutel, M.; Bruggenjurgen, B.; Richter, H.; Vogelberg, C. Real-world evidence of subcutaneous allergoid immunotherapy in house dust mite-induced allergic rhinitis and asthma. Allergy 2020, 75, 2050–2058. [Google Scholar] [CrossRef] [PubMed]
  102. Passalacqua, G.; Albano, M.; Fregonese, L.; Riccio, A.; Pronzato, C.; Mela, G.S.; Canonica, G.W. Randomised controlled trial of local allergoid immunotherapy on allergic inflammation in mite-induced rhinoconjunctivitis. Lancet 1998, 351, 629–632. [Google Scholar] [CrossRef] [PubMed]
  103. Gallego, M.T.; Iraola, V.; Himly, M.; Robinson, D.S.; Badiola, C.; Garcia-Robaina, J.C.; Briza, P.; Carnes, J. Depigmented and polymerised house dust mite allergoid: Allergen content, induction of IgG4 and clinical response. Int. Arch. Allergy Immunol. 2010, 153, 61–69. [Google Scholar] [CrossRef] [PubMed]
  104. Di Gioacchino, M.; Cavallucci, E.; Ballone, E.; Cervone, M.; Di Rocco, P.; Piunti, E.; Filardo, G.S.; Turi, M.C.; Mangifesta, R.; Quecchia, C.; et al. Dose-dependent clinical and immunological efficacy of sublingual immunotherapy with mite monomeric allergoid. Int. J. Immunopathol. Pharmacol. 2012, 25, 671–679. [Google Scholar] [CrossRef] [PubMed]
  105. Corrigan, C.J.; Kettner, J.; Doemer, C.; Cromwell, O.; Narkus, A.; Study, G. Efficacy and safety of preseasonal-specific immunotherapy with an aluminium-adsorbed six-grass pollen allergoid. Allergy 2005, 60, 801–807. [Google Scholar] [CrossRef] [PubMed]
  106. Agostinis, F.; Foglia, C.; Bruno, M.E.; Falagiani, P. Efficacy, safety and tolerability of sublingual monomeric allergoid in tablets given without up-dosing to pediatric patients with allergic rhinitis and/or asthma due to grass pollen. Eur. Ann. Allergy Clin. Immunol. 2009, 41, 177–180. [Google Scholar]
  107. Mosges, R.; Ritter, B.; Kayoko, G.; Allekotte, S. Carbamylated monomeric allergoids as a therapeutic option for sublingual immunotherapy of dust mite- and grass pollen-induced allergic rhinoconjunctivitis: A systematic review of published trials with a meta-analysis of treatment using Lais(R) tablets. Acta Dermatovenerol. Alp. Pannonica Adriat. 2010, 19, 3–10. [Google Scholar]
  108. Klimek, L.; Pfaar, O.; Bousquet, J.; Senti, G.; Kundig, T. Allergen immunotherapy in allergic rhinitis: Current use and future trends. Expert. Rev. Clin. Immunol. 2017, 13, 897–906. [Google Scholar] [CrossRef]
  109. Barber, D.; Diaz-Perales, A.; Escribese, M.M.; Kleine-Tebbe, J.; Matricardi, P.M.; Ollert, M.; Santos, A.F.; Sastre, J. Molecular allergology and its impact in specific allergy diagnosis and therapy. Allergy 2021, 76, 3642–3658. [Google Scholar] [CrossRef]
  110. Dramburg, S.; Hilger, C.; Santos, A.F.; de Las Vecillas, L.; Aalberse, R.C.; Acevedo, N.; Aglas, L.; Altmann, F.; Arruda, K.L.; Asero, R.; et al. EAACI Molecular Allergology User’s Guide 2.0. Pediatr. Allergy Immunol. 2023, 34 (Suppl. S28), e13854. [Google Scholar] [CrossRef]
  111. Gonzalez-Perez, R.; Poza-Guedes, P.; Pineda, F.; Sanchez-Machin, I. Advocacy of Precision Allergy Molecular Diagnosis in Decision Making for the Eligibility of Customized Allergen Immunotherapy. Curr. Issues Mol. Biol. 2023, 45, 9976–9984. [Google Scholar] [CrossRef] [PubMed]
  112. Valenta, R.; Campana, R.; Focke-Tejkl, M.; Niederberger, V. Vaccine development for allergen-specific immunotherapy based on recombinant allergens and synthetic allergen peptides: Lessons from the past and novel mechanisms of action for the future. J. Allergy Clin. Immunol. 2016, 137, 351–357. [Google Scholar] [CrossRef] [PubMed]
  113. Valenta, R.; Linhart, B.; Swoboda, I.; Niederberger, V. Recombinant allergens for allergen-specific immunotherapy: 10 years anniversary of immunotherapy with recombinant allergens. Allergy 2011, 66, 775–783. [Google Scholar] [CrossRef] [PubMed]
  114. Jutel, M.; Jaeger, L.; Suck, R.; Meyer, H.; Fiebig, H.; Cromwell, O. Allergen-specific immunotherapy with recombinant grass pollen allergens. J. Allergy Clin. Immunol. 2005, 116, 608–613. [Google Scholar] [CrossRef] [PubMed]
  115. Pauli, G.; Larsen, T.H.; Rak, S.; Horak, F.; Pastorello, E.; Valenta, R.; Purohit, A.; Arvidsson, M.; Kavina, A.; Schroeder, J.W.; et al. Efficacy of recombinant birch pollen vaccine for the treatment of birch-allergic rhinoconjunctivitis. J. Allergy Clin. Immunol. 2008, 122, 951–960. [Google Scholar] [CrossRef]
  116. Larche, M. Immunotherapy with allergen peptides. Allergy Asthma Clin. Immunol. 2007, 3, 53–59. [Google Scholar] [CrossRef]
  117. Moldaver, D.; Larche, M. Immunotherapy with peptides. Allergy 2011, 66, 784–791. [Google Scholar] [CrossRef]
  118. Wraith, D.C.; Krishna, M.T. Peptide allergen-specific immunotherapy for allergic airway diseases-State of the art. Clin. Exp. Allergy 2021, 51, 751–769. [Google Scholar] [CrossRef]
  119. Patel, D.; Couroux, P.; Hickey, P.; Salapatek, A.M.; Laidler, P.; Larche, M.; Hafner, R.P. Fel d 1-derived peptide antigen desensitization shows a persistent treatment effect 1 year after the start of dosing: A randomized, placebo-controlled study. J. Allergy Clin. Immunol. 2013, 131, 103–109.e7. [Google Scholar] [CrossRef]
  120. Oldfield, W.L.; Larche, M.; Kay, A.B. Effect of T-cell peptides derived from Fel d 1 on allergic reactions and cytokine production in patients sensitive to cats: A randomised controlled trial. Lancet 2002, 360, 47–53. [Google Scholar] [CrossRef]
  121. Kettner, A.; DellaCorte, G.; de Blay, F.; Jacobsen, L.; Jutel, M.; Worm, M.; Charlon, V.; Simonsen, K.; Reymond, C.; Spertini, F. Benefit of Bet v 1 contiguous overlapping peptide immunotherapy persists during first follow-up season. J. Allergy Clin. Immunol. 2018, 142, 678–680.e7. [Google Scholar] [CrossRef] [PubMed]
  122. Sharif, H.; Singh, I.; Kouser, L.; Mosges, R.; Bonny, M.A.; Karamani, A.; Parkin, R.V.; Bovy, N.; Kishore, U.; Robb, A.; et al. Immunologic mechanisms of a short-course of Lolium perenne peptide immunotherapy: A randomized, double-blind, placebo-controlled trial. J. Allergy Clin. Immunol. 2019, 144, 738–749. [Google Scholar] [CrossRef] [PubMed]
  123. Mosges, R.; Kasche, E.M.; Raskopf, E.; Singh, J.; Sohlich, L.; Astvatsatourov, A.; Shah-Hosseini, K.; Pirotton, S.; Haazen, L.; Durham, S.R.; et al. A randomized, double-blind, placebo-controlled, dose-finding trial with Lolium perenne peptide immunotherapy. Allergy 2018, 73, 896–904. [Google Scholar] [CrossRef] [PubMed]
  124. Narayanan, M.; Freidl, R.; Focke-Tejkl, M.; Baranyi, U.; Wekerle, T.; Valenta, R.; Linhart, B. A B Cell Epitope Peptide Derived from the Major Grass Pollen Allergen Phl p 1 Boosts Allergen-Specific Secondary Antibody Responses without Allergen-Specific T Cell Help. J. Immunol. 2017, 198, 1685–1695. [Google Scholar] [CrossRef]
  125. Niederberger, V.; Neubauer, A.; Gevaert, P.; Zidarn, M.; Worm, M.; Aberer, W.; Malling, H.J.; Pfaar, O.; Klimek, L.; Pfutzner, W.; et al. Safety and efficacy of immunotherapy with the recombinant B-cell epitope-based grass pollen vaccine BM32. J. Allergy Clin. Immunol. 2018, 142, 497–509.e9. [Google Scholar] [CrossRef]
  126. Rauber, M.M.; Mobs, C.; Campana, R.; Henning, R.; Schulze-Dasbeck, M.; Greene, B.; Focke-Tejkl, M.; Weber, M.; Valenta, R.; Pfutzner, W. Allergen immunotherapy with the hypoallergenic B-cell epitope-based vaccine BM32 modifies IL-10- and IL-5-secreting T cells. Allergy 2020, 75, 450–453. [Google Scholar] [CrossRef]
  127. Eckl-Dorna, J.; Weber, M.; Stanek, V.; Linhart, B.; Ristl, R.; Waltl, E.E.; Villazala-Merino, S.; Hummel, A.; Focke-Tejkl, M.; Froeschel, R.; et al. Two years of treatment with the recombinant grass pollen allergy vaccine BM32 induces a continuously increasing allergen-specific IgG(4) response. EBioMedicine 2019, 50, 421–432. [Google Scholar] [CrossRef]
  128. Su, Y.; Romeu-Bonilla, E.; Anagnostou, A.; Fitz-Patrick, D.; Hearl, W.; Heiland, T. Safety and long-term immunological effects of CryJ2-LAMP plasmid vaccine in Japanese red cedar atopic subjects: A phase I study. Hum. Vaccin. Immunother. 2017, 13, 2804–2813. [Google Scholar] [CrossRef]
  129. Roesler, E.; Weiss, R.; Weinberger, E.E.; Fruehwirth, A.; Stoecklinger, A.; Mostbock, S.; Ferreira, F.; Thalhamer, J.; Scheiblhofer, S. Immunize and disappear-safety-optimized mRNA vaccination with a panel of 29 allergens. J. Allergy Clin. Immunol. 2009, 124, 1070–1077.e11. [Google Scholar] [CrossRef]
  130. Cooke, R.A.; Barnard, J.H.; Hebald, S.; Stull, A. Serological Evidence of Immunity with Coexisting Sensitization in a Type of Human Allergy (Hay Fever). J. Exp. Med. 1935, 62, 733–750. [Google Scholar] [CrossRef]
  131. Orengo, J.M.; Radin, A.R.; Kamat, V.; Badithe, A.; Ben, L.H.; Bennett, B.L.; Zhong, S.; Birchard, D.; Limnander, A.; Rafique, A.; et al. Treating cat allergy with monoclonal IgG antibodies that bind allergen and prevent IgE engagement. Nat. Commun. 2018, 9, 1421. [Google Scholar] [CrossRef] [PubMed]
  132. Shamji, M.H.; Singh, I.; Layhadi, J.A.; Ito, C.; Karamani, A.; Kouser, L.; Sharif, H.; Tang, J.; Handijiev, S.; Parkin, R.V.; et al. Passive Prophylactic Administration with a Single Dose of Anti-Fel d 1 Monoclonal Antibodies REGN1908-1909 in Cat Allergen-induced Allergic Rhinitis: A Randomized, Double-Blind, Placebo-controlled Clinical Trial. Am. J. Respir. Crit. Care Med. 2021, 204, 23–33. [Google Scholar] [CrossRef] [PubMed]
  133. Gevaert, P.; De Craemer, J.; De Ruyck, N.; Rottey, S.; de Hoon, J.; Hellings, P.W.; Volckaert, B.; Lesneuck, K.; Orengo, J.M.; Atanasio, A.; et al. Novel antibody cocktail targeting Bet v 1 rapidly and sustainably treats birch allergy symptoms in a phase 1 study. J. Allergy Clin. Immunol. 2022, 149, 189–199. [Google Scholar] [CrossRef] [PubMed]
  134. Olivieri, B.; Gunaydin, F.E.; Corren, J.; Senna, G.; Durham, S.R. The combination of allergen immunotherapy and biologics for inhalant allergies: Exploring the synergy. Ann. Allergy Asthma Immunol. 2024; in press. [Google Scholar] [CrossRef] [PubMed]
  135. Casale, T.B.; Busse, W.W.; Kline, J.N.; Ballas, Z.K.; Moss, M.H.; Townley, R.G.; Mokhtarani, M.; Seyfert-Margolis, V.; Asare, A.; Bateman, K.; et al. Omalizumab pretreatment decreases acute reactions after rush immunotherapy for ragweed-induced seasonal allergic rhinitis. J. Allergy Clin. Immunol. 2006, 117, 134–140. [Google Scholar] [CrossRef]
  136. Dantzer, J.A.; Wood, R.A. The use of omalizumab in allergen immunotherapy. Clin. Exp. Allergy 2018, 48, 232–240. [Google Scholar] [CrossRef]
  137. De Filippo, M.; Votto, M.; Caminiti, L.; Carella, F.; Castro, G.; Landi, M.; Olcese, R.; Panasiti, I.; Vernich, M.; Barberi, S.; et al. Omalizumab and allergen immunotherapy for respiratory allergies: A mini-review from the Allergen-Immunotherapy Committee of the Italian Society of Pediatric Allergy and Immunology (SIAIP). Allergol. Immunopathol. 2022, 50, 47–52. [Google Scholar] [CrossRef]
  138. Corren, J.; Saini, S.S.; Gagnon, R.; Moss, M.H.; Sussman, G.; Jacobs, J.; Laws, E.; Chung, E.S.; Constant, T.; Sun, Y.; et al. Short-Term Subcutaneous Allergy Immunotherapy and Dupilumab are Well Tolerated in Allergic Rhinitis: A Randomized Trial. J. Asthma Allergy 2021, 14, 1045–1063. [Google Scholar] [CrossRef]
  139. Corren, J.; Larson, D.; Altman, M.C.; Segnitz, R.M.; Avila, P.C.; Greenberger, P.A.; Baroody, F.; Moss, M.H.; Nelson, H.; Burbank, A.J.; et al. Effects of combination treatment with tezepelumab and allergen immunotherapy on nasal responses to allergen: A randomized controlled trial. J. Allergy Clin. Immunol. 2023, 151, 192–201. [Google Scholar] [CrossRef]
Figure 1. Approaches of allergen immunotherapy for respiratory allergies.
Figure 1. Approaches of allergen immunotherapy for respiratory allergies.
Pharmaceuticals 17 01510 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lao-Araya, M. Novel Approaches to Allergen Immunotherapy for Respiratory Allergies. Pharmaceuticals 2024, 17, 1510. https://doi.org/10.3390/ph17111510

AMA Style

Lao-Araya M. Novel Approaches to Allergen Immunotherapy for Respiratory Allergies. Pharmaceuticals. 2024; 17(11):1510. https://doi.org/10.3390/ph17111510

Chicago/Turabian Style

Lao-Araya, Mongkol. 2024. "Novel Approaches to Allergen Immunotherapy for Respiratory Allergies" Pharmaceuticals 17, no. 11: 1510. https://doi.org/10.3390/ph17111510

APA Style

Lao-Araya, M. (2024). Novel Approaches to Allergen Immunotherapy for Respiratory Allergies. Pharmaceuticals, 17(11), 1510. https://doi.org/10.3390/ph17111510

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop