Next Article in Journal
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology
Next Article in Special Issue
Workup and Clinical Assessment for Allergen Immunotherapy Candidates
Previous Article in Journal
TDP-43 Cytoplasmic Translocation in the Skin Fibroblasts of ALS Patients
Previous Article in Special Issue
The Fusion Protein rFlaA:Betv1 Modulates DC Responses by a p38-MAPK and COX2-Dependent Secretion of PGE2 from Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Allergen Immunotherapy: Current and Future Trends

by
Gandhi F. Pavón-Romero
,
Maria Itzel Parra-Vargas
,
Fernando Ramírez-Jiménez
,
Esmeralda Melgoza-Ruiz
,
Nancy H. Serrano-Pérez
and
Luis M. Teran
*
Department of Immunogenetics and Allergy, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
*
Author to whom correspondence should be addressed.
Cells 2022, 11(2), 212; https://doi.org/10.3390/cells11020212
Submission received: 22 October 2021 / Revised: 23 December 2021 / Accepted: 27 December 2021 / Published: 8 January 2022
(This article belongs to the Special Issue Advances in Immunotherapy of Allergic Diseases)

Abstract

:
Allergen immunotherapy (AIT) is the sole disease-modifying treatment for allergic rhinitis; it prevents rhinitis from progressing to asthma and lowers medication use. AIT against mites, insect venom, and certain kinds of pollen is effective. The mechanism of action of AIT is based on inducing immunological tolerance characterized by increased IL-10, TGF-β, and IgG4 levels and Treg cell counts. However, AIT requires prolonged schemes of administration and is sometimes associated with adverse reactions. Over the last decade, novel forms of AIT have been developed, focused on better allergen identification, structural modifications to preserve epitopes for B or T cells, post-traductional alteration through chemical processes, and the addition of adjuvants. These modified allergens induce clinical-immunological effects similar to those mentioned above, increasing the tolerance to other related allergens but with fewer side effects. Clinical studies have shown that molecular AIT is efficient in treating grass and birch allergies. This article reviews the possibility of a new AIT to improve the treatment of allergic illness.

Graphical Abstract

1. Introduction

Allergen immunotherapy (AIT) originated in the early twentieth century [1]. In 1954, the first controlled clinical trial was developed, and AIT improved the symptoms in the group receiving the pollen extract compared with the control group [2]. AIT is recommended for the treatment of allergic rhinitis (AR) and asthma by many medical organizations, based on controlled clinical trials and meta-analyses [3,4,5]. Many of these beneficial effects are due to AIT being the unique therapy able to induce allergen long-term tolerance after discontinuation. The administration of AIT for three years produces persistent clinical-immunological changes for at least two years [6,7].
However, the conventional schemes used in AIT are prolonged. Subcutaneous immunotherapy (SCIT) comprises a build-up phase (in which the allergen concentration increases gradually) and a maintenance phase (in which the projected dose is applied), which must be administered for at least three years (Figure 1a). Additionally, the development of adverse reactions is associated with first dosages, leading to treatment abandonment [8,9]. Likewise, AIT has not shown clinical benefits with all allergens.
New methods and novel molecules have been developed to improve AIT for the last 30 years. Currently, allergoids, recombinant allergens based on specific epitopes or joined to immunological adjuvants—also known as hypoallergenic immunotherapy—even applied by new routes, constitute new variants of this therapeutic. The present review describes the best advances reported concerning each of the AIT areas.

2. Mechanisms of Action of AIT

The purpose of the AIT is to modulate the physiopathology mechanisms of allergy. The allergic response begins with the allergen being endocytosed by the dendritic cells of the airway epithelium. Subsequently, these cells go to local secondary lymphoid organs where the antigen presentation to Th0 happens. Th0 differentiate to Th2 and synthesize its interleukin profile (IL-4, IL-5, and IL-13) to allow the production of IgE-type specific allergen antibodies (Figure 2a).
The mechanism of AIT includes an increase in the number of T regulatory cells (Treg) CD4+CD25+ [10] and IgG4 levels [11]. Treg cells produce TGF-β [12], IFN-γ, and IL-10, which are essential for AIT’s immunomodulatory activities [13,14]. For example, IL-10 inhibits the histamine release by mast cells mediated by its IgE-dependent activation [15] and a decrease in eosinophil cationic protein release [16]. Likewise, the combination with IL-4/IL-13 allows the isotype change to IgG4 instead of IgE in plasma cells [17,18]. IFN-γ inhibits the synthesis of Th2 interleukins as IL-4, IL-5, and IL-13. AIT is also linked to a reduction in antigen-specific T cell clones and IgA2 production. Recently, AIT increased the differentiation of T follicular reg cells-TFH (CXCR5+ Foxp3+) and B regulatory cells—Breg [19]. In the first case, TFH synthesizes the IL-21 and IL-4 necessary for B cell proliferation and antibody synthesis [20]. B cells secrete IL-10/TGF-β, both of which help with immune modulation [21]. These mechanisms are reference points for other new routes, molecules, and adjuvants used in AIT. However, some effects have specific routes (Figure 2b).
SCIT increases the Treg profile and IgG-mediated blockade of the binding of IgE and allergens to plasma cells after twelve months [17]. Interestingly, the titers of the blocking antibodies induced by AIT increase approximately 30 times more than by sublingual immunotherapy (SLIT) in less time (6–10 weeks) [22]. In this context, nasal mucosa biopsies from grass-sensitive patients increased the levels of IL-10 mRNA after two years of SCIT treatment [23].
Although the mechanism of action of SCIT has been studied further, the SLIT mechanism has similarities with the one already explained [24,25]. Langerhans cells in the oral mucosa are essential for the antigen presentation and the activation of T lymphocytes. The Toll-like receptor (TLR) TLR4 activation in these cells increases the synthesis of IL-10, TGF-β, IFN-γ, and IL-2 and Foxp3+ expression in Treg cells from both oral epithelium biopsies and peripheral blood [18,26]. Ihara F. reported that the SLIT applied for 52 weeks in house dust mite (HDM)-sensitive AR patients also decreases the Th2 cells [27], as well as in ryegrass pollen-sensitive patients in six months [12,27]. However, the synthesis of blocking IgG4 antibodies occurs over a longer time (~24 weeks) than that using SCIT (6–10 weeks) [28], maintaining the increase in IgG+ memory B cells 1–3 years after treatment [29,30].
There is evidence that the rupture of the skin barrier facilitates the penetration of allergens into the epidermis, causing Langerhans cells to catch allergens and travel to regional lymph nodes, with the subsequent stimulation and differentiation of T lymphocytes toward a Th2 profile [31]. This mechanism is the basis of percutaneous AIT, which causes a more potent delayed T-cell-mediated immune response than SCIT and SLIT. Unfortunately, the synthesis of IgG4 is less than that using the routes mentioned above [32,33].
Additionally, an inguinal node AIT injection with recombinants (phospholipase A2 and Fel d 1) increased IgG levels 10 times more in a shorter period (two weeks) than SCIT with a dose 100 times lower. These effects are probably due to the allergens reaching the lymph nodes directly compared with SCIT or other routes. Interestingly, this administration route is the only one that produces both IgG2 and IgG4. Similarly to the other routes, intralymphatic immunotherapy induces increased IL-10, IFN-γ, and IL-4 levels but in a shorter time than SCIT [34]. The advantages of this route are that it is painless, and the tolerance against allergens is achieved in less time (four months) and with more durability. However, a disadvantage of this approach is that it should be used under ultrasound guidance [35].

3. Efficacy of Allergen Immunotherapy

The benefits of AIT are not the same for each allergen responsible for sensitization and the different allergic diseases. For example, SCIT or SLIT is strongly recommended for seasonal AR induced by pollen, while SLIT is recommended for HDM in mild asthma but not for all allergens [36,37,38,39], and there is limited evidence for allergic respiratory disease caused by fungal spores [40,41]. The meta-analyses are considered the highest level of evidence The SCIT meta-analyses for seasonal AR showed an improvement in symptoms (SMD, −0.73; p < 0.0001; I2 = 63.21%) and medication scores (SMD, −0.57; p < 0.0001; I2 = 64.02%). Interestingly, this evidence is mainly derived from articles on AIT for grass pollen. However, SCIT against HDM also showed similar results (For the symptoms: SMD, −2.17; p = 0.001; I2 = 96%%/For the medication score: (SMD, −1.17; p = 0.03; I2 = 86%). Concerning SLIT, the most up-to-date version of the Cochrane review reports described a reduction in the outcomes mentioned above, primarily for grasses (For the symptoms: SMD, −0.49; p < 0.00001; I2 = 81%/For the medication score: SMD, −0.32; p = 0.00035; I2 = 50%), and other robust reports concluded the same for HDM (For the symptoms: SMD, −0.95; p < 0.00001; I2 = 92%/For the medication score: SMD, −1.88; p < 0.00001; I2 = 95%) [38,42]. Some reports have found similar levels of efficacy using both routes, even comparing different forms of SLIT (drops and tablets) [43,44]. In relation to SLIT and asthma, a recent meta-analysis could not draw clinically useful conclusions due to the non-validated scores and limited evidence for relevant outcomes such as asthma exacerbations [37]. For other allergens, there is scarce high-quality information. However, evidence supports a clinical improvement in SCIT and SLIT for epitheliums in clinical outcomes such as ocular, nasal, or asthma symptoms, peak expiratory flow rate, and medication scores [45].
Notably, some meta-analyses, particularly those using SLIT, are controversial because of the heterogeneity of the few included trials, different presentations, and doses of the extracts used, and/or of the use of non-validated scales of symptoms and medication scores, limiting the provision of clear clinical conclusions. Additionally, heterogeneity exists in the different clinical trials included in the meta-analyses. Throughout history, an attempt has been made to improve the effectiveness criteria and propose a consensus on the duration of SCIT and SLIT [46]. Furthermore, one of the most interesting properties of AIT is that it provides benefits for many years after the therapy schemes have been concluded. Patients have a reduction in medication and the percentage of eosinophils, as well as an increase in the threshold to the response to methacholine four years after finishing the AIT, according to prospective studies evaluating SLIT regimens administered for at least three years, and even these effects are more prolonged with schemes applied for a longer time [47,48]. In a similar context, the application of a complete AIT scheme for mites avoids the development of new sensitizations in 75% of patients at least three years after its conclusion [49,50].
Regardless of these scores, some previously discussed interleukins (IL-10, TGF-β), antibodies titers (IgG4) [50], IgE [51], specific IgE/total IgE [52], and cell lines (Treg cells, B regs and DC) have been used as biomarkers [53]. Although the modification of other types of lymphocytes and immune cells have also been described. For example, AIT for grasses increase the expression of the transcriptional factor of DCreg (C1QA, FcεRIIIA, FTL) and reduced that of DC2 (C1QA, FcεRIIIA, FTL,); in a similar way, it diminished the expression of CD63/CD203c in basophils, which correlates with the medical score and is considered as a biomarker of efficacy [52,54].
SCIT and SLIT are generally well tolerated. However, as another therapeutic approach, they are not exempt from the development of adverse reactions. The risk of a systemic reaction is more frequent with SCIT than SLIT. A systemic response to AIT injection is documented in approximately 2% of patients, and the mortality related to a SCIT injection is higher in non-controlled asthma patients in the build-up phase and maintenance [8,48]. Low-risk fatal reactions occur in 1 per 2.5 million injections, although this rate has decreased in recent years [55,56]. SLIT is considered the safest route, even in asthma patients [37].
Recently, attempts have been made to improve the adaptability and success of immunotherapy using monoclonal antibodies, particularly with the anti-IgE monoclonal antibody (Omalizumab). Omalizumab in immunotherapy is an off-label treatment. However, clinical trials have shown that the use of Omalizumab in rapid regimens of AIT, such as rush, reduces the adverse reactions attributed to immunotherapy [57]. Another trial showed benefits in the symptom control of seasonal rhinoconjunctivitis and asthma when used before or during immunotherapy schemes [58,59]. However, it will unlikely be approved as a general indication because of its high cost and limited and probably temporary clinical benefits.

4. Allergoids

Allergoids are chemically modified allergens via polymerization with glutaraldehyde or formaldehyde. This modification gives them better immunogenicity features [60] because they react with primary amino groups in the polypeptide chain of the allergen, producing intramolecular and intermolecular cross-linked polymers of high molecular weight allergens [61]. The conformational epitopes of IgE are destroyed, while the linear epitopes of T cells are not affected [62]. This structure allows its administration in high doses during a short-term accumulation phase [63].
Since 1992, diverse allergoids have been used; for example, a mixture of six grass allergens modified with formaldehyde and co-precipitated with aluminum, known as Allergovit®, has been applied in a phase-IIIb study by the subcutaneous route in AR patients [64]. In a phase-III clinical trial, allergoids enhanced the levels of subclass IgG (IgG2 and IgG4) antibodies for grasses [65], decreased basophil activity [57], and relieved allergic symptoms after one year of treatment, doubling this effect in the second year in a phase-III study [65]. However, similar mechanisms have also been shown with allergoids derived exclusively from one grass (Phleum pretense) in a phase-II study. This activity is attributed to the high sequence identity of group 1 and group 5, ~90% and >55%, respectively, among members of the Poaceae family [66]. Instead, Pfaar O. evaluated other glutaraldehyde and formaldehyde (Acaroid®) allergoids for HDM. They identified that doses ≥20,000 UA and 18,000 TU improve the threshold of symptoms in nasal provocation tests and increase blocking antibody levels, respectively [67,68]. This molecular result was also shown with allergoids of birch (Allergovit® Birch phase-II study and PURETHAL® phase-IV study) [69,70].
In the same context, the carbamylation of the lysine groups has been used to develop allergoids of low molecular weight that can be absorbed easily by the mucosa as the monomeric allergoid of group five of Phleum pratense (Grazax® 75,000 standardized quality units) [71]. This protein, applied sublingually, reduced the need for antihistaminic drugs during the pollination season, in addition to the clinical effects mentioned with the other molecule [72]. Additionally, this allergoid maintained its clinical benefits after termination for at least two years [73]. The allergoid LAIS®, a mixture of extracts from group-1 mites, was another carbamylated chemical employed in a phase-II research. LAIS® applied by SLIT at doses of 3000 UA over one year reduced the IL-4 and augmented IFN-γ levels. Additionally, it improved rhinitis severity and reduced drug intake [74]. In the same context, Hüser C. evaluated other similar allergoids but applied them for 12 weeks and noted that 2000 UA/day decreased the symptoms in conjunctival provocation tests [75].
Concerning its safety, the patients treated with Allergovit® for grass allergy in phase-II studies developed mild reactions [76,77], even when applied during the pollination season [64], suggesting that utilizing allergoids in rapid treatment schemes may allow patients to achieve clinical and immunological tolerance more quickly, as well as enhance the adherence to AIT [77].

5. Adjuvants

An adjuvant is a molecule that enhances immune responses by interacting with antigens physically or chemically [78] and has been traditionally classified as first-generation (aluminum, microcrystalline tyrosine and calcium phosphate) and second-generation (Toll-like receptors—TLR). Additionally, other adjuvants have been used with promising results, such as liposomes and virus-like particles. (Table 1).

5.1. Aluminum

Aluminum-containing adjuvants (ACDs) have been used widely for AIT [79,80]. ACDs have adsorbent properties, and their administration causes the local deposition of antigens, which are released slowly [81]. The injection of an ACD intramuscularly induces uric acid release in the muscle; both molecules stimulate NOD-like receptors (NLRs), specifically NALP3, an essential component of the inflammasome, along with Pycard and caspase-1, subsequently promoting the synthesis of inflammatory interleukins (IL-1, IL-18, and IL-33) in monocytes [82,83]. IL-1 primarily induces a Th2 response, which is necessary to synthesize IgG subclass antibodies, and this last effect is not fully understood [84]. An increase in the IgG subclass has been shown in AIT, which comprises an ACD and fragments of Bet v 1 (aa 1–74 and aa 75–160) [85]. This adjuvant’s clinical efficacy has been proven: Corrigan C. evaluated ACD-containing Allergovit® for grasses, and this vaccine reduced the symptoms and drug requirements [65]. Furthermore, a phase-IIb study reported that an ACD-containing synthetic contiguous overlapping peptide (COP) vaccine for Bet v1 improves the quality of life of AR patients [86]. ACDs have been integrated into many glutaraldehyde allergoids, and patients who receive these treatments develop mild reactions [87]. However, some reports have indicated that ACDs induce granulomas in the skin and severe reactions inherent to AIT and, in some cases, are associated with neurologic diseases [88,89].

5.2. Microcrystalline Tyrosine (MCT)

L-tyrosine is an amino acid that delays the bioavailability of allergenic materials [90]. Since first reported in the 1980s, the adsorption of allergenic molecules to L-tyrosine has enhanced the induction of IgG [91]. MCT has the property of being rapidly released and metabolized, unlike aluminum, which can be found at the sites of application in murine models of AIT and is associated with the development of granulomas [92]. MCT has not shown toxic properties, although it is contraindicated in tyrosine metabolism disorders [90]. Currently, MCT is patented as an adjuvant for use in immunotherapy [93] and is integrated into glutaraldehyde allergoids for Dermatophagoides to help reduce allergic symptoms and diminish the use of relief drugs [94,95,96,97].

5.3. Calcium Phosphate (CaP)

Calcium salts such as CaP [98] and MCT share physical properties and immunological mechanisms. Allergens are adsorbed by CaP in the shape of microcrystals at the injection site, releasing the antigen slowly in encapsulated particles toward the APC [99]. CaP plus five-grass pollen extract improves nasal symptoms and increases the noise reactivity in nasal challenges as well as the IgG4 levels [100]. CaP has been used as an alternative to ACD [101]. However, patients treated with CaP developed double subcutaneous local reactions [102], but this effect disappears when it is applied intramuscularly [101].

5.4. Toll-like Receptors (TLR)

The main evidence of their potential usefulness in AIT comes from in vitro studies. For example, resquimod, an agonist of TLR8/9, increases the synthesis of IFN-γ (suppressor of the allergic inflammatory response) in PMNC derived from AR patients sensitized to palm pollen [103]. TLR2/6 can be stimulated by a lipopeptide derived from Mycoplasma, known as Macrophage Activating Lipopeptide of 2 kDa, which decreases the Th2 profile and eosinophil counts in bronchoalveolar lavage, but has no effect on T-reg cells in mice sensitized to Phleum pretense [104]. In a mouse allergy model, fusion proteins containing recombinant flagellin A (TLR5 agonist) and Bet v 1 (rFlaA:Betv1) decreased the Th2 responses and avoided allergic sensitization compared to rBet v 1 [105]. Monophosphoryl lipid A (MPL) is a TLR4 agonist, which is a lipopolysaccharide of the cell wall of Salmonella minnesota that stimulates the production of IFN-γ and IL-12, but does not promote IL-5 synthesis [106]. In a phase-I/IIa study, patients treated with MPL and mixed-grass pollen developed an increase in IgG with lower IgE levels and low nasal reactivity following a grass challenge [107]. Similarly, Worm M. evaluated its role with an allergoid of birch and showed that the administration of MPL-birch reduced basophil activation more than 100-fold compared with native allergens [108].
In the same context, MPL integrated into the Pollinex Quattro allergoid vaccine for grass allergy, an alternative ultrashort (four pre-seasonal injections) of SCIT, induces increases in CD4+, CD25+, Foxp3+, and IgG antibodies but not IgE; in addition to improving the allergic symptoms [109,110,111], even after increasing the cumulative dose, its safety is not compromised [112]. Regarding the long-lasting effect of MPL, patients who received MPL-Parietaria (a scheme of four injections before the pollen season for three years) applied using the SCIT route showed clinical improvement up to five years after treatment discontinuation [113]. Similar results were obtained in patients with the cessation of Pollinex Quattro after three years [114]. In the case of TLR intracellular agonists such as cytosine-phosphate-guanosine (CpG) to stimulate TLR9, CpG and a recombinant Chenopodium album increased IL-10 and IFN-γ and reduced IL-4. B cells from patients sensitive to cedar also reduced IL-5 and IL-13 [115,116]. New agonists of TLR continue to be evaluated, such as AZD8848-TLR7, and showed an improved lung function in asthma patients in a phase-II study [117]. These findings suggest that TLR regulation may be a promising therapeutic approach in allergic diseases.

5.5. Liposomes

Liposomes are spheres or vesicles integrated by lipids such as cholesterol and/or phosphatidylcholine that allow the encapsulation of allergens [118]. Liposomes constituted by cationic lipids allow for a better interaction with DCs and are released, processed, and subsequently presented to T cells [119,120]. In a mouse model of food allergy, liposomes containing synthetic neoglycolipids such as mannotriose and dipalmito-ylphosphatidylcholine activate T CD8+, T CD4+, CD25+, and Foxp3+ cells, inhibiting the antibodies and alleviating the allergic symptoms [121]. In the same context, in an AIT for cockroaches, recombinants of arginine kinase Per a 9 encapsulated in this vehicle and applied nasally to cockroach-allergic mice reduced the inflammatory response mediated by the Th2 profile and increased the expression of IL-12, IFN-γ, and IL-10 [120]. Interestingly, when combined with Tregitopes, the liposomes augmented IL-10 and TGF-β but decreased lung inflammation and airway remodeling [118]. Der p 1 coated in liposomes was administered for one year in asthma patients; after treatment, the patients showed reduced symptoms and an increased threshold to methacholine challenges, as well as eosinophilic inflammation, compared with the control group [122,123].

5.6. Virus-like Particles (VLPs)

VLPs are produced from viral capsid proteins and have the potential to activate the immune system through innate mechanisms (PAMPs) that are not dependent on T cells [124]. The main reports of VLP efficacy come from animal models. For example, Fel d 1 incorporated into VLPs derived from a cucumber mosaic virus and applied to an allergic model animal induces a specific IgG response [125]. In humans, CYT003-QbG10 VLPs (TLR9 agonists arranged into VLPs) improve the AR quality of life, asthma symptoms, and lung function [126,127]. Extensive reviews on the role of adjuvants in AIT have already been published [128,129]

6. Peptides and Recombinants

The specificity of the protective antibodies targets the epitopes, not complete antigen molecules. Epitopes are linear segments (~20 aa) of antigen from molecules located in the major histocompatibility complex (MHC) after the APC process during the specific immune response, which induces the synthesis of antibodies or T cell clones [130]. Many products used in hypoallergenic immunotherapy include T cell epitopes. These are classified into short (~20 aa) and overlapping peptides (long peptides), which are a complete allergen sequence with overlaps, ensuring the presence of all possible T-cell epitopes. In both cases, the lack of conformational structures cannot activate IgE-dependent mechanisms [131,132].
In a similar context, recombinant hypoallergens have been designed from native allergens. For example, these molecules show conformational changes in their IgE-binding epitopes, a quality that reduces their immunoallergenicity. The use of recombinant allergens makes it possible to have homogeneous and well-defined standardized products in adequate quantities without undesirable materials that impair the stability of the allergen and their efficacy. The most commonly used technique for recombinant synthesis includes complementary DNA (cDNA), previous identification of allergen (Figure 3A,B). cDNA is a molecule copied from an mRNA molecule by reverse transcriptase and lacks the introns and regulatory sequences present in genomic DNA (Figure 3C). The insertion of a cDNA sequence into the bacterial genome (Escherichia coli) encodes a recombinant protein allergen (Figure 3D). Finally, the recombinant allergen must be validated as another new drug or molecule (Figure 3E). Usually, researchers evaluate the binding to specific IgE, the release of preformed products of basophils or mast cells, the type of interleukin profile induced, the stimulation of cellular clones, and the synthesis of blocking antibodies [133]. Recombinants have been used for synthesized B-cell epitopes for therapeutic uses [134].
However, they can be produced by other methods, such as oligomerization, point mutation, fragmentation, mosaic, and allergen hybrids [135]. Recently, the in silico models have been used for developing AIT for Dermathophagoides. For example, a recombinant allergen of grasses developed by point mutations—the substitution of amino acids located at the calcium-binding site—produces a decrease in the negative charge of the mutant, endowing it with greater flexibility, which is important in the development of the side effects [136]. Another hybrid recombinant of ambrosia pectate lyase (rAmb a 1, residues 1174–397) and mugwort (rArt v 6, residues 173–396), as well as a hybrid of both, have been investigated. The recombinant proteins and chimera did not cause the recognition of IgE in patients sensitive to these weeds or the degranulation of preformed mediators or cytokines of the Th2 profile [137]. Additionally, a fusion protein (rFlaA: Art1hyp), integrated by flagellin A (TLR5 agonist) and hypoallergenic mugwort (Art v1- change in cysteines for serines, altering the epitope for IgE), stimulates the synthesis of subclasses of IgG but not of IgE [138].
Computational vaccine design (in silico design) entails the use of computational tools to map epitopes, select antigens, and develop immunogens [139]; through the design of new molecules and their model in 3D (I-TASSER software), it is validated (ProSA-web) and subsequently synthesized (GenScript). The hybrid proteins synthesized can incorporate many allergen determinants of the same or different species and have been evaluated mainly in allergic murine models for mites. For example, Ferreira F. created two hybrid proteins from Blomia tropicalis fragments (Blot t 5 and Blot t 21). Another hybrid protein is Dpx4, which contains antigenic regions of allergens from Dermathophagoides pteronyssius (Der p 1, Der p 2, Der p 7, and Der p 10). MAVAC-BD-2 is the first molecule to contain epitopes from Dermatophagoides sp. (Blo t 5, Blo t 8, Blo t 10/Der p 1, Der p 2, Der p 7, Der p 8). This protein reduced IgE to Blot 5 and Der p 2 by 20%, approximately, while boosting IgG. Concerning the limits of these products, it is probable that the addition of aggregates could impact protein stability [140,141,142]. The clinical-immune efficacy of some recombinants is better when they are integrated with adjuvants such as MPL or aluminum. For example, MPL coupled to Phleum pratense recombinants (rPhl p 1, rPhl p 2, rPhl p 5) induces a potent humoral response mediated by IgG and IgM and reduces the histamine release from basophils, improving the allergic symptoms [143]. The second adjuvant improved AR patients’ quality of life and increased IgG1 titers in the first year after the therapeutic scheme ended. However, a higher increase in IgG4 was observed until the second year [144]. Peptides and recombinants have been tested in phase-I/II clinical trials to treat cat and pollen allergies.

7. Clinical-Immune Efficacy of Recombinant Allergens

7.1. Cat

Fel d 1 is the most common cat allergen. Fel d 1 hypoallergenicity can be synthesized by introducing duplications of T cell epitopes (DTE). In a murine cat allergy model, a type of recombinant DTE III induced high IgG2 levels. In mice, IgG can reduce skin reactivity and improve airway hyperreactivity by blocking the binding of patients’ IgE to rFel d 1 [145]. AIT for Fel d 1 has been tested in vaccines based on T cell epitope peptides (SPIRES), which are short allergen peptides that make up the allergen’s primary T cell epitopes, and MHC II has been used to construct immune-therapeutic mechanisms [146]. Allervax cats (cat peptide for AIT) showed clinical benefits; however, they had late adverse reactions in clinical phases [147]. Conversely, in phase-II and -III studies, a Cat PAD (also known as ToleroMune Cat) has also shown a reduction in rhinoconjunctivitis symptoms and safety in cat-allergic patients using four intradermal doses of 6 nmol [148,149], decreasing the CRTh2 expression but not altering the number of Fel d 1-TCD4+ cells [150].
In a phase-I research, rFel d 1 was also fused to the HIV-derived translocation peptide (TAT), mediating the cytoplasmic uptake of extracellular proteins and the truncated human invariant chain (MALT-Fel d 1), which was administered intralymphatically in a scheme of three dosages. MALT-Fel d 1 improved the symptoms during the nasal challenge and increased the IgG4 and IL-10 levels [151]; this humoral response was greater than that of another IgG subclass, which increased after the first month of treatment. Interestingly, rIgG4 for cat allergy has been evaluated in cat-allergic patients in a phase-Ib study, demonstrating its ability to increase the IgG/IgE ratio and decrease the clinical symptoms in nasal provocation, with similar results in a scheme of eight days. These data suggest that passive immunization can treat allergies using allergen-specific IgG antibodies [152].

7.2. Birch

rBet v 1 is one of the first molecules evaluated as allergen immunotherapy [138]. Niederberger V. realized in 2004 a phase-II study and administered two fragments of rBet v1 (F1, aa 1–73 without methionine; F2, aa 74–159) and two trimers (comprising three covalently linked copies of Bet v 1) applied in eight doses (maximum dose of 80 µg) before the birch season. These recombinants induced the synthesis of IgG1 and IgG4 after treatment; despite a slight decrease, the antibodies remained present during the pollination season and decreased the release of histamine in serum and IgE levels [153]. Interestingly, an increase in IgG1, IgG2, and IgG4 was identified in the nasal secretion and is associated with reduced nasal sensitivity in the nasal birch challenge [154]. Additionally, the trimer of Bet v 1 decreased the production of the Th2 profile but increased the IL-12 levels, and both recombinant proteins decreased the nasal symptoms and skin reactivity [155].
Allergen-specific T lymphocytes (LT CLA+ and CCR4+, necessary for the migration of T cells from the blood to the skin) were found to increase after an epicutaneous injection of both rBet v1 and two fragments of this protein, in addition to a slight increase in IgG levels and its subclasses but a null humoral IgE response [156].
Other recombinants have been studied. For example, Meyer W. evaluated the response to the rBet v 1-folding variant, which has intact T cell epitopes, in a phase-III study. After exposure to AR patients for eight hours in an environmental exposure chamber with birch, the researcher applied a 10-dose injection scheme (20, 80, 160, and 320 µg) applied weekly, noting that the 80-µg dose of this recombinant induced the greatest synthesis of IgG1, reduced the nasal symptoms, and induced minimal adverse effects [157]. rBet v1 was also tested sublingually in a phase-II study, administering one sublingual tablet per day for five months before the pollination season; this treatment decreased the symptoms and use of rescue medications during the pollination season, with mild effects [158].

7.3. Grasses

From 1999, Gehlhar K. applied two recombinants (5a and 5b) with a homogeneity of approximately 70% with Phl p 5 in pediatric patients. These molecules decreased the AR symptoms and increased the levels of IgG, IgG2, and IgG4 at the end of the study; even the quotient IgG1/IgG4 correlated with the clinical scenario [159].
Recently, a fusion protein based on allergen-derived peptide B cell epitopes of the four major allergens of timothy (Phl p 1, 2, 5, and 6) and PreS protein (an immunogenic carrier that fosters antibody responses [160] from the hepatitis B virus—HBV), adsorbed to aluminum hydroxide, known as BM32, has been proven in patients with AR to grasses [161]. A two-year scheme was used to test BM32 in a phase-IIb study. In the first year, the researchers applied four injections; the initial three dosages were applied three months prior to the European grass pollen season and a booster in the fall (after the season) (Figure 1b). In the second year, they reapplied the first three doses of the scheme mentioned before the next pollination season. With this scheme, an increase in IgG, IgG1, and IgG4 was observed, but this effect declined after five months, particularly for IgG1. However, the booster was sufficient to restore the titers of IgG1 and increase the allergen-specific IgG4 levels. BM32 did not significantly modify the IgE levels compared with the baseline values. In terms of therapeutic advantages, phase-IIb studies showed benefic changes in AR life quality and asthma symptoms during the pollination, and these effects increased in the second year of treatment [162], with the main adverse reactions classified as mild [163].
Allergic mast cell and basophil degranulation may be prevented by the presence of blocking IgG1 and IgG4 antibodies against the IgE binding sites of the major grass pollen allergens. Likewise, as observed in phase II studies, blocking antibodies hinders the IgE-facilitated allergen presentation and the consecutive T cell activation [160]. In the same context, it inhibited the allergen-specific T cell reactivity in both treated patients and in vitro models [134]. Additionally, BM32 induces IL-10 synthesis and low levels of IL-5 and interleukins used as markers of immunological efficacy and tolerance [164].

8. Passive Immunization with IgG Antibodies

Passive immunization using serum from AIT-treated patients was first used by Cooke RA in 1935 [165]. In addition, the administration of IgG antibodies against parvalbumin decreased the allergic reactions in a mouse model of fish allergy [166]. A separate study was undertaken in a model of peanut allergy, where a treatment with allergen-specific IgG antibodies prevented peanut-sensitized mice from suffering anaphylaxis after the intravenous challenge with the whole peanut extract [167]. As previously reported, a clinical research in cat-allergy patients who were passively vaccinated with monoclonal IgG antibodies against the main cat allergen demonstrated an improvement in their symptoms [168].

9. Benefits and Limitations of Novel Immunotherapies

Strong benefits are associated with using hypoallergenic therapy, among which three are notable: preventing the development of allergies in sensitive patients, inducing immunity with allergens related to the primo-sensitizer, and conferring immunity to other entities. In the first case, based on a longitudinal study, the recognition of specific allergen-IgE in early life often precedes allergic symptoms or an allergic disease [169]. Campana R. described the effects of prophylactic AIT in non-allergic patients, but IgE-positive birch using recombinant rBet v1 (aa 1–160) and two hypoallergenic fragments of this protein (F1 aa 1–74 and F2 aa 75–160) in a scheme of three dosages applied monthly before the pollination season, and extra doses one year later, showed that the IgG concentrations increased in patients treated with the recombinants compared with the placebo group. Despite this change, IgG titers decreased at the end of the pollination season. However, they increased after a booster was administered before the following season. Interestingly, the patients who had received both rBetv1 and F1/F2 showed a decreased birch-specific IgE and no reactivity to the skin test, suggesting that IgG antibodies mediated the blockade of IgE. Additionally, the patients who had received active treatment showed no adverse reactions (Figure 1c) [170].
On the other hand, birch sensitivity is not related only to allergic respiratory diseases. Seventy percent of patients with food allergies (FAs) to apple and hazelnut develop sensitivity to this aeroallergen [171], and insufficient evidence exists that convectional AIT with extracts of birch improves the FA symptoms [172]. The administration of 80 µg of the rBet v 1 folding variant elevated IgG4 levels toward Bet v 1 and its related allergens contained in soja, apple, and cherry [173]. Kinaciyan T. described the effects of rMal d 1 and rBet v1 in patients with FA in a phase-II study using one sublingual tablet daily for 16 weeks (25 µg/day). When the patients were challenged through sublingual provocation with apple, the group that had received rMal d 1 showed fewer oral symptoms related to apple compared with rBet v 1 and enhanced IgG4 titers for apple but not rBet v 1. Instead, the patients who had received rBet v 1 induced a protective response to apple [174]. This latter finding has been shown in other allergens from fagal families [175].
Interestingly, after five applications, BM32 can also induce the synthesis of IgG, IgG1, and IgG4 against the N-terminal portion of PreS, whose ligand (sodium taurocholate cotransporter polypeptide located on the surface of hepatocytes) is necessary for the entry of the virus. These findings suggest that some components of AIT with recombinants can induce protective immunity against infectious diseases [176].
Although the clinical benefits of novel forms of AIT show promising results, some limitations should be addressed. For example, the use of hypoallergenic recombinant derivatives designed to reduce IgE reactivity [148] while maintaining T cell epitopes can still cause T-cell-mediated late-phase side effects. In this context, the employment of short non-allergenic peptides containing T cell epitopes that are not IgE-reactive can also cause adverse side effects in treated patients [132]. Additionally, T cell epitopes are too short to induce allergen-specific IgG antibodies and clinical protection [177].
Recently, a DNA vaccine was shown in phase Ia and Ib to treat a Japanese red cedar allergy employing the allergen CryJ2, and the lysosomal-associated membrane protein 1 (LAMP-1) induced humoral protective antibodies against this pollen [178]. This therapy may induce an uncontrolled synthesis of allergens in the body, causing allergic reactions [179]. A passive immunization with monoclonal IgG antibodies against the sensitizing allergen is a good approach. However, the high costs associated with large-scale antibody production remain a significant barrier [168]. The latest generation of hypoallergenic carrier-bound B-cell epitope-containing vaccines appears to be able to overcome the problem of the side effects. This allows for high-dose injections, induces robust allergen-specific IgG responses, does not cause allergic sensitization, and thus holds great promise for revolutionizing AIT and even for prophylactic allergy vaccination [162].

10. Recombinants for Diagnosis

Component-resolved diagnosis (CRD) was established in 1980 as a new concept in allergy diagnosis [180,181]. The CRD identifies a specific IgE toward purified natural or recombinant allergens rather than raw allergen extracts to determine a patient’s sensitization at the molecular level [182]; currently, more than 130 allergen molecules are commercialized [183]. CRD allows for a more precise identification of the allergen, using two types of tests for either one assay per sample (singleplex-ImmunoCAP, ImmuLite, and HyTech) or many allergens per sample depot in microarrays (multiplex platform- ImmunoCAP ISAC-ThermoFisher Scientific/Phadia) [184,185,186,187]. For example, in the case of patients allergic to several types of grass, these tests can help allergists distinguish between a major sensitization agent such as Phl p 1 and a pan allergen implicated in a crossover reaction with other species, such as Phl p 12 or Bet v1. This example can be applied to other proteins (profilins, polcalcins, non-specific lipid transportation proteins, PR-10, or tropomyosins). Recently, Armentia A. demonstrated the cross-reaction between different species, and marijuana and tomato can share lipid-transported proteins [185,188,189].

11. Allergy Proteomics

Over the last few years, proteomics has become critical to identify and structurally characterize allergens. Indeed, proteomics applications include in vitro diagnostics, allergen discovery, and the analysis of biologicals proposed for AIT [190,191]. Immunoproteomics involves a combination of proteomics with bidimensional polyacrylamide gel electrophoresis followed by mass spectrometry (MS) [189]. Briefly, the proteome of a biological specimen (e.g., pollen extract) is separated first by isoelectric focusing (first dimension) to separate proteins depending on their charge, and then by molecular masses (second dimension). Thus, hundreds of proteins are resolved as minute spots on a polyacrylamide gel, followed by western blotting to a membrane using sera from allergic patients. Then, IgE-reactive spots are characterized by MS. Although immunological methods have long been considered the gold standard for allergen identification, MS offers considerable benefits by allowing allergen identification based on MS/MS data from the allergen of interest [192]. For example, we have previously identified novel allergens from Ligustrum lucidum including enolase, pollen-specific polygalacturonases, Fra e 9.01 (β-1,3-glucanase), profilin, alanine aminotransferase, and ATP synthase beta subunit [175]. Using a similar approach, allergens from both red oak and pecan [189,191] have been recognized. The identification of enolase from some kinds of pollen is of particular interest [193]. Currently, proteomic technology is mostly used for research. Proteomic-based miniaturized technologies that allow for a more accurate, faster, and easier diagnosis of allergic sensitization are likely to contribute to the emergence of individualized AIT suited to individual allergic patients in the near future.

12. Conclusions

Despite the development of novel allergen-specific immunotherapy, licensing any vaccine for the clinic has proven difficult. Currently, allergen-specific immunotherapy with natural allergen extracts is the only viable disease-modifying treatment for allergic patients based on long-term symptom relief, and it can also prevent AR from progressing to asthma. However, caution should be taken because allergen injection can be associated with adverse reactions and because of the allergenicity of natural extracts. The side effects are usually harmless and, in rare cases, can cause fatal reactions. Importantly, patients must not show symptoms because of AIT allergenicity, particularly in asthma [194]. Traditional allergen extract-based AIT may be revolutionized in the future by some molecular AIT technologies. The latest generation of carrier-bound B-cell epitope-based allergy vaccines has the potential to transform AIT because it may prevent side effects, allowing the administration of high doses to induce strong allergen-specific IgG responses and providing sensitized patients with lasting effects (Supplementary Table S1).

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cells11020212/s1, Table S1: Summary of allergoids, adjuvants and recombinants used in allergen immunotherapy.

Author Contributions

Conceptualization, G.F.P.-R., F.R.-J. and L.M.T.; methodology, G.F.P.-R. and L.M.T.; software, N.H.S.-P., M.I.P.-V. and E.M.-R.; investigation, G.F.P.-R., M.I.P.-V., N.H.S.-P. and E.M.-R.; resources, L.M.T.; data curation, F.R.-J. and N.H.S.-P.; writing—original draft preparation, G.F.P.-R., M.I.P.-V., E.M.-R. and L.M.T.; writing—review and editing, G.F.P.-R., F.R.-J., M.I.P.-V. and E.M.-R.; visualization, L.M.T. and G.F.P.-R.; supervision, L.M.T. and G.F.P.-R.; project administration, L.M.T.; funding acquisition, L.M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This study was realized with the technical support and assistance of Michelle Alejandra Rodríguez Cervantes and Juan Carlos Vizuet Derueda.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ring, J.; Gutermuth, J. 100 years of hyposensitization: History of allergen-specific immunotherapy (ASIT). Allergy 2011, 66, 713–724. [Google Scholar] [CrossRef] [PubMed]
  2. Frankland, A.W.; Augustin, R. Prophylaxis of summer hay-fever and asthma. Lancet 1954, 263, 1055–1057. [Google Scholar] [CrossRef]
  3. Bousquet, J.; Schünemann, H.J.; Togias, A.; Bachert, C.; Erhola, M.; Hellings, P.W.; Klimek, L.; Pfaar, O.; Wallace, D.; Ansotegui, I.; et al. 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.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Global Initiative of Asthma. Global Strategy for Asthma Management and Prevention. GINA. 2021. Available online: https://ginasthma.org/wp-content/uploads/2021/05/GINA-Main-Report-2021-V2-WMS.pdf (accessed on 15 September 2021).
  5. Pfaar, O.; Bachert, C.; Bufe, A.; Buhl, R.; Ebner, C.; Eng, P.; Friedrichs, F.; Fuchs, T.; Hamelmann, E.; Hartwig-Bade, D.; et al. Guideline on allergen-specific immunotherapy in IgE-mediated allergic diseases. Allergo J. Int. 2014, 23, 282–319. [Google Scholar] [CrossRef]
  6. Durham, S.R.; Walker, S.M.; Varga, E.-M.; Jacobson, M.R.; O’Brien, F.; Noble, W.; Till, S.J.; Hamid, Q.A.; Nouri-Aria, K.T. Long-Term Clinical Efficacy of Grass-Pollen Immunotherapy. N. Engl. J. Med. 1999, 341, 468–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Penagos, M.; Durham, S.R. Duration of allergen immunotherapy for inhalant allergy. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 594–605. [Google Scholar] [CrossRef]
  8. Pavon-Romero, G.F.; Larenas-Linnemann, D.E.; Xochipa Ruiz, K.K.E.; Ramirez-Jimenez, F.; Teran, L.M.; Ramirez-Jimenez, F.; Teran, L. Subcutaneous Allergen-Specific Immunotherapy Is Safe in Pediatric Patients with Allergic Rhinitis. Int. Arch. Allergy Immunol. 2021, 182, 553–561. [Google Scholar] [CrossRef]
  9. Larenas-Linnemann, D.; Luna-Pech, J.; Rodríguez-Pérez, N.; Rodríguez-González, M.; Arias-Cruz, A.; Blandón-Vijil, M.; Costa-Domínguez, M.; Del Río-Navarro, B.; Estrada-Cardona, A.; Navarrete-Rodríguez, E.; et al. GUIMIT 2019, Mexican Guideline on Immunotherapy. Guideline on the diagnosis of IgE-mediated allergic disease and immunotherapy following the ADAPTE approach. Rev. Alerg. Mex. 2019, 66 (Suppl. S1), 1–105. [Google Scholar] [CrossRef]
  10. Gardner, L.; Thien, F.; Douglass, J.; Rolland, J.; O’Hehir, R. Induction of T “regulatory” cells by standardized house dust mite immunotherapy: An increase in CD4+CD25+ interleukin-10+ T cells expressing peripheral tissue trafficking markers. Clin. Exp. Allergy 2004, 34, 1209–1219. [Google Scholar] [CrossRef]
  11. Varona, R.; Ramos, T.; Escribese, M.; Jimeno, L.; Galán, A.; Würtzen, P.; Vega, F.; Marín, A.; Martín, S.; Carrera, A.; et al. 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. [Google Scholar] [CrossRef]
  12. O’Hehir, R.; Gardner, L.; de Leon, M.; Hales, B.; Biondo, M.; Douglass, J.; Rolland, J.; Sandrini, A. House dust mite sublingual immu-notherapy: The role for transforming growth factor-beta and functional regulatory T cells. Am. J. Respir. Crit. Care Med. 2009, 180, 936–947. [Google Scholar] [CrossRef]
  13. Gardner, L.; Rolland, J.M.; O’Hehir, R.E. High Dose Allergen Stimulation of T Cells from House Dust Mite-Allergic Subjects Induces Expansion of IFN-?+ T Cells, Apoptosis of CD4+IL-4+ T Cells and T Cell Anergy. Int. Arch. Allergy Immunol. 2004, 133, 1–13. [Google Scholar] [CrossRef]
  14. Francis, J.N.; Till, S.J.; Durham, S.R. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. J. Allergy Clin. Immunol. 2003, 111, 1255–1261. [Google Scholar] [CrossRef]
  15. Royer, B.; Varadaradjalou, S.; Saas, P.; Guillosson, J.J.; Kantelip, J.P.; Arock, M. Inhibition of IgE-induced activation of human mast cells by IL-10. Clin. Exp. Allergy 2001, 31, 694–704. [Google Scholar] [CrossRef]
  16. Rak, S.; Löwhagen, O.; Venge, P. The effect of immunotherapy on bronchial hyperresponsiveness and eosinophil cationic protein in pollen-allergic patients. J. Allergy Clin. Immunol. 1988, 82, 470–480. [Google Scholar] [CrossRef]
  17. Wachholz, P.A.; Soni, N.K.; Till, S.J.; Durham, S.R. Inhibition of allergen-IgE binding to B cells by IgG antibodies after grass pollen immunotherapy. J. Allergy Clin. Immunol. 2003, 112, 915–922. [Google Scholar] [CrossRef]
  18. Scadding, G.W.; Shamji, M.H.; Jacobson, M.R.; Lee, D.I.; Wilson, D.; Lima, M.T.; Pitkin, L.; Pilette, C.; Nouri-Aria, K.; Durham, S.R. 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. [Google Scholar] [CrossRef] [Green Version]
  19. Linterman, M.A.; Pierson, W.; Lee, S.K.; Kallies, A.; Kawamoto, S.; Rayner, T.F.; Srivastava, M.; Divekar, D.P.; Beaton, L.; Hogan, J.J.; et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 2011, 17, 975–982. [Google Scholar] [CrossRef] [Green Version]
  20. Shulman, Z.; Gitlin, A.D.; Weinstein, J.S.; Lainez, B.; Esplugues, E.; Flavell, R.A.; Craft, J.E.; Nussenzweig, M.C. Dynamic signaling by T follicular helper cells during germinal center B cell selection. Science 2014, 345, 1058–1062. [Google Scholar] [CrossRef] [Green Version]
  21. Matsumoto, M.; Baba, A.; Yokota, T.; Nishikawa, H.; Ohkawa, Y.; Kayama, H.; Kallies, A.; Nutt, S.L.; Sakaguchi, S.; Takeda, K.; et al. Interleukin-10-Producing Plasmablasts Exert Regulatory Function in Autoimmune Inflammation. Immunity 2014, 41, 1040–1051. [Google Scholar] [CrossRef] [Green Version]
  22. 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. 2019, 181, 71–80. [Google Scholar] [CrossRef]
  23. Nouri-Aria, K.T.; Wachholz, P.A.; Francis, J.N.; Jacobson, M.R.; Walker, S.M.; Wilcock, L.K.; Staple, S.Q.; Aalberse, R.C.; Till, S.J.; Durham, S.R. Grass Pollen Immunotherapy Induces Mucosal and Peripheral IL-10 Responses and Blocking IgG Activity. J. Immunol. 2004, 172, 3252–3259. [Google Scholar] [CrossRef] [Green Version]
  24. Suárez-Fueyo, A.; Ramos, T.; Galán, A.; Jimeno, L.; Wurtzen, P.A.; Marin, A.; de Frutos, C.; Blanco, C.; Carrera, A.; Barber, D.; et al. Grass tablet sublingual immunotherapy downregulates the TH2 cytokine response followed by regulatory T-cell generation. J. Allergy Clin. Immunol. 2014, 133, 130–138.e2. [Google Scholar] [CrossRef]
  25. 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]
  26. Allam, J.-P.; Peng, W.-M.; Appel, T.; Wenghoefer, M.; Niederhagen, B.; Bieber, T.; Bergé, S.; Novak, N. Toll-like receptor 4 ligation enforces tolerogenic properties of oral mucosal Langerhans cells. J. Allergy Clin. Immunol. 2008, 121, 368–374.e1. [Google Scholar] [CrossRef]
  27. Ihara, F.; Sakurai, T.; Yonekura, S.; Iinuma, T.; Yagi, R.; Ito, T.; Matsuura, A.; Morimoto, Y.; Arai, T.; Suzuki, S.; et al. Identification of specifically reduced Th2 cell subsets in allergic rhinitis patients after sublingual immunotherapy. Allergy 2018, 73, 1823–1832. [Google Scholar] [CrossRef]
  28. 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]
  29. Heeringa, J.J.; McKenzie, C.I.; Varese, N.; Hew, M.; Bakx, A.T.C.M.; Aui, P.M.; Rolland, J.M.; O’Hehir, R.E.; Van Zelm, M.C. Induction of IgG 2 and IgG 4 B-cell memory following sublingual immunotherapy for ryegrass pollen allergy. Allergy 2020, 75, 1121–1132. [Google Scholar] [CrossRef] [Green Version]
  30. Couroux, P.; Ipsen, H.; Stage, B.S.; Damkjaer, J.T.; Steffensen, M.A.; Salapatek, A.M.; Lund, K.; Würtzen, P.A. A birch sublingual allergy immunotherapy tablet reduces rhinoconjunctivitis symptoms when exposed to birch and oak and induces IgG4 to allergens from all trees in the birch homologous group. Allergy 2019, 74, 361–369. [Google Scholar] [CrossRef] [Green Version]
  31. Senti, G.; Graf, N.; Haug, S.; Rüedi, N.; von Moos, S.; Sonderegger, T.; Johansen, P.; Kündig, T.M. Epicutaneous allergen administration as a novel method of allergen-specific immunotherapy. J. Allergy Clin. Immunol. 2009, 124, 997–1002. [Google Scholar] [CrossRef]
  32. Senti, G.; Von Moos, S.; Tay, F.; Graf, N.; Johansen, P.; Kündig, T.M. Determinants of efficacy and safety in epicutaneous allergen immunotherapy: Summary of three clinical trials. Allergy 2015, 70, 707–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Strid, J.; Hourihane, J.; Kimber, I.; Callard, R.; Strobel, S. Disruption of the stratum corneum allows potent epicutaneous immunization with protein antigens resulting in a dominant systemic Th2 response. Eur. J. Immunol. 2004, 34, 2100–2109. [Google Scholar] [CrossRef] [PubMed]
  34. Martínez-Gómez, J.M.; Johansen, P.; Erdmann, I.; Senti, G.; Crameri, R.; Kündig, T.M. Intralymphatic Injections as a New Administration Route for Allergen-Specific Immunotherapy. Int. Arch. Allergy Immunol. 2009, 150, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Senti, G.; Vavricka, B.M.P.; Erdmann, I.; Diaz, M.I.; Markus, R.; McCormack, S.J.; Simard, J.J.; Wüthrich, 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] [Green Version]
  36. Virchow, J.C.; Backer, V.; Kuna, P.; Prieto, L.; Nolte, H.; Villesen, H.H.; Ljørring, C.; Riis, B.; De Blay, F. Efficacy of a House Dust Mite Sublingual Allergen Immunotherapy Tablet in Adults With Allergic Asthma. JAMA 2016, 315, 1715–1725. [Google Scholar] [CrossRef]
  37. Fortescue, R.; Kew, K.M.; Leung, M.S.T. Sublingual immunotherapy for asthma. Cochrane Database Syst. Rev. 2020, 9, CD011293–CD011293. [Google Scholar] [CrossRef]
  38. Calderón, M.A.; Alves, B.; Jacobson, M.; Hurwitz, B.; Sheikh, A.; Durham, S. Allergen injection immunotherapy for seasonal allergic rhinitis. Cochrane Database Syst. Rev. 2007, 2007, CD001936. [Google Scholar] [CrossRef]
  39. 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] [Green Version]
  40. Coop, C.A. Immunotherapy for Mold Allergy. Clin. Rev. Allergy Immunol. 2013, 47, 289–298. [Google Scholar] [CrossRef]
  41. Bozek, A.; Pyrkosz, K. Immunotherapy of mold allergy: A review. Hum. Vaccines Immunother. 2017, 13, 2397–2401. [Google Scholar] [CrossRef]
  42. Radulovic, S.; Calderón, M.A.; Wilson, D.; Durham, S. Sublingual immunotherapy for allergic rhinitis. Cochrane Database Syst. Rev. 2010, 2010, 002893. [Google Scholar] [CrossRef]
  43. Sieber, J.; Shah-Hosseini, K.; Mösges, R. Specific immunotherapy for allergic rhinitis to grass and tree pollens in daily medical practice—Symptom load with sublingual immunotherapy compared to subcutaneous immunotherapy. Ann. Med. 2011, 43, 418–424. [Google Scholar] [CrossRef]
  44. Nelson, H.; Cartier, S.; Allen-Ramey, F.; Lawton, S.; Calderon, M.A. Network Meta-analysis Shows Commercialized Subcutaneous and Sublingual Grass Products Have Comparable Efficacy. J. Allergy Clin. Immunol. Pract. 2015, 3, 256–266.e3. [Google Scholar] [CrossRef]
  45. Dhami, S.; Agarwal, A. Does evidence support the use of cat allergen immunotherapy? Curr. Opin. Allergy Clin. Immunol. 2018, 18, 350–355. [Google Scholar] [CrossRef]
  46. Calderon, M.A.; Casale, T.B.; Nelson, H.S.; Demoly, P. An evidence-based analysis of house dust mite allergen immunotherapy: A call for more rigorous clinical studies. J. Allergy Clin. Immunol. 2013, 132, 1322–1336. [Google Scholar] [CrossRef]
  47. Marogna, M.; Spadolini, I.; Massolo, A.; Canonica, G.W.; Passalacqua, G. Long-lasting effects of sublingual immunotherapy according to its duration: A 15-year prospective study. J. Allergy Clin. Immunol. 2010, 126, 969–975. [Google Scholar] [CrossRef]
  48. Marogna, M.; Bruno, M.; Massolo, A.; Falagiani, P. Long-Lasting Effects of Sublingual Immunotherapy for House Dust Mites in Allergic Rhinitis with Bronchial Hyperreactivity: A Long-Term (13-Year) Retrospective Study in Real Life. Int. Arch. Allergy Immunol. 2006, 142, 70–78. [Google Scholar] [CrossRef]
  49. Pajno, G.B.; Barberio, G.; De Luca, F.; Morabito, L.; Parmiani, S. Prevention of new sensitizations in asthmatic children monosensitized to house dust mite by specific immunotherapy. A six-year follow-up study. Clin. Exp. Allergy 2001, 31, 1392–1397. [Google Scholar] [CrossRef]
  50. Inal, A.; Altintas, D.; Yilmaz, M.; Karakoc, G.; Kendirli, S.; Sertdemir, Y. Prevention of new sensitizations by specific immunotherapy in children with rhinitis and/or asthma monosensitized to house dust mite. J. Investig. Allergol. Clin. Immunol. 2007, 17, 85–91. [Google Scholar]
  51. Tabar, A.I.; Prieto, L.; Alba, P.; Nieto, A.; Rodríguez, M.; Torrecillas, M.; Huertas, B.; Gómez, E.; Fernández, F.J.; Blanca, M.; et al. Double-blind, randomized, placebo-controlled trial of allergen-specific immunotherapy with the major allergen Alt a 1. J. Allergy Clin. Immunol. 2019, 144, 216–223.e3. [Google Scholar] [CrossRef] [Green Version]
  52. Soyyigit, S.; Guloglu, D.; Ikinciogullari, A.; Secil, D.; Oztuna, D.; Mungan, D.; Misirligil, Z.; Sin, B.A. Immunologic alterations and efficacy of subcutaneous immunotherapy with Dermatophagoides pteronyssinus in monosensitized and polysensitized patients. Ann. Allergy Asthma Immunol. 2016, 116, 244–251.e2. [Google Scholar] [CrossRef]
  53. Kucuksezer, U.C.; Ozdemir, C.; Cevhertas, L.; Ogulur, I.; Akdis, M.; Akdis, C.A. Mechanisms of allergen-specific immunotherapy and allergen tolerance. Allergol. Int. 2020, 69, 549–560. [Google Scholar] [CrossRef]
  54. Özdemir, S.K.; Sin, B.A.; Güloğlu, D.; Ikincioğulları, A.; Gençtürk, Z.; Mısırlıgil, Z. Short-Term Preseasonal Immunotherapy: Is Early Clinical Efficacy Related to the Basophil Response? Int. Arch. Allergy Immunol. 2014, 164, 237–245. [Google Scholar] [CrossRef]
  55. Epstein, T.G.; Liss, G.M.; Murphy-Berendts, K.; Bernstein, D.I. Risk factors for fatal and nonfatal reactions to subcutaneous immunotherapy. Ann. Allergy Asthma Immunol. 2016, 116, 354–359.e2. [Google Scholar] [CrossRef]
  56. Bernstein, D.I.; Wanner, M.; Borish, L.; Liss, G.M.; The Immunotherapy Committee of the American Academy of Allergy, Asthma and Immunology. Twelve-year survey of fatal reactions to allergen injections and skin testing: 1990–2001. J. Allergy Clin. Immunol. 2004, 113, 1129–1136. [Google Scholar] [CrossRef]
  57. Casale, T.B.; Busse, W.W.; Kline, J.; Ballas, Z.; Moss, M.H.; Townley, R.G.; Mokhtarani, M.; Seyfert-Margolis, V.; Asare, A.; Bateman, K. 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]
  58. Kuehr, J.; Brauburger, J.; Zielen, S.; Schauer, U.; Kamin, W.; Von Berg, A.; Leupold, W.; Bergmann, K.-C.; Rolinck-Werninghaus, C.; Gräve, M.; et al. Efficacy of combination treatment with anti-IgE plus specific immunotherapy in polysensitized children and adolescents with seasonal allergic rhinitis. J. Allergy Clin. Immunol. 2002, 109, 274–280. [Google Scholar] [CrossRef]
  59. Kopp, M.V.; Hamelmann, E.; Zielen, S.; Kamin, W.; Bergmann, K.-C.; Sieder, C.; Stenglein, S.; Seyfried, S.; Wahn, U.; for The DUAL Study Group. Combination of omalizumab and specific immunotherapy is superior to immunotherapy in patients with seasonal allergic rhinoconjunctivitis and co-morbid seasonal allergic asthma. Clin. Exp. Allergy 2009, 39, 271–279. [Google Scholar] [CrossRef]
  60. Klimek, L.; Thorn, C.; Pfaar, O. Depigmentierte Allergoide für die allergenspezifische Immuntherapie. HNO 2010, 58, 51–56. [Google Scholar] [CrossRef]
  61. Ibarrola, I.; Sanz, M.L.; Gamboa, P.M.; Mir, A.; Benahmed, D.; Ferrer, A.; Arilla, M.C.; Martinez, A.; Asturias, J. Biological characterization of glutaraldehyde-modified Parietaria judaica pollen extracts. Clin. Exp. Allergy 2004, 34, 303–309. [Google Scholar] [CrossRef]
  62. Würtzen, P.A.; Lund, L.; Lund, G.; Holm, J.; Millner, A.; Henmar, H. Chemical Modification of Birch Allergen Extract Leads to a Reduction in Allergenicity as well as Immunogenicity. Int. Arch. Allergy Immunol. 2007, 144, 287–295. [Google Scholar] [CrossRef] [PubMed]
  63. Gokmen, N.M.; Ersoy, R.; Gulbahar, O.; Ardeniz, O.; Sin, A.; Unsel, M.; Kokuludag, A. Desensitization Effect of Preseasonal Seven-Injection Allergoid Immunotherapy with Olive Pollen on Basophil Activation: The Efficacy of Olive Pollen-Specific Preseasonal Allergoid Immunotherapy on Basophils. Int. Arch. Allergy Immunol. 2012, 159, 75–82. [Google Scholar] [CrossRef] [PubMed]
  64. Mahler, V.; Zielen, S.; Rosewich, M. Year-round treatment initiation for a 6-grasses pollen allergoid in specific immunotherapy of allergic rhinoconjunctivitis and asthma. Immunotherapy 2019, 11, 1569–1582. [Google Scholar] [CrossRef] [PubMed]
  65. Corrigan, C.; Kettner, J.; Doemer, C.; Cromwell, O.; Narkus, A.; the Study Group. Efficacy and safety of preseasonal-specific immunotherapy with an aluminium-adsorbed six-grass pollen allergoid. Allergy 2005, 60, 801–807. [Google Scholar] [CrossRef]
  66. Pfaar, O.; Hohlfeld, J.M.; Al-Kadah, B.; Hauswald, B.; Homey, B.; Hunzelmann, N.; Schliemann, S.; Velling, P.; Worm, M.; Klimek, L. Dose-response relationship of a new Timothy grass pollen allergoid in comparison with a 6-grass pollen allergoid. Clin. Exp. Allergy 2017, 47, 1445–1455. [Google Scholar] [CrossRef] [Green Version]
  67. Pfaar, O.; Nell, M.J.; Boot, J.D.; Versteeg, S.A.; van Ree, R.; Roger, A.; Riechelmann, H.; Sperl, A.; Elberink, J.N.G.O.; Diamant, Z.; et al. A randomized, 5-arm dose finding study with a mite allergoid SCIT in allergic rhinoconjunctivitis patients. Allergy 2016, 71, 967–976. [Google Scholar] [CrossRef]
  68. Jutel, M.; Rudert, M.; Kreimendahl, F.; Kuna, P. Efficacy and tolerability of a house dust mite allergoid in allergic bronchial asthma: A randomized dose-ranging trial. Immunotherapy 2018, 10, 1149–1161. [Google Scholar] [CrossRef]
  69. Zielen, S.; Plückhahn, K.; Akboga, Y.; Rieker-Schwienbacher, J.; Thieme, U.; Rosewich, M. Fast up-dosing with a birch allergoid is safe and well tolerated in allergic rhinitis patients with or without asthma. Immunotherapy 2019, 11, 177–187. [Google Scholar] [CrossRef]
  70. Buczyłko, K.; Boot, D.; Van Ree, R.; Van Der Werf, J.F. Accelerated Up-Dosing of Subcutaneous Immunotherapy with a Registered Allergoid Birch Pollen Preparation. Int. Arch. Allergy Immunol. 2017, 172, 183–186. [Google Scholar] [CrossRef]
  71. Durham, S.R. Sustained effects of grass pollen AIT. Allergy 2011, 66, 50–52. [Google Scholar] [CrossRef]
  72. Mösges, 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® tablets. Acta Dermatovenerol. Alpina Pannonica Adriat. 2010, 19, 1–8. [Google Scholar]
  73. 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] [Green Version]
  74. Di Gioacchino, M.; Cavallucci, E.; Ballone, E.; Cervone, M.; Di Rocco, P.; Piunti, E.; Filardo, G.; Turi, M.; 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]
  75. Hüser, C.; Dieterich, P.; Singh, J.; Shah-Hosseini, K.; Allekotte, S.; Lehmacher, W.; Compalati, E.; Mösges, R. A 12-week DBPC dose-finding study with sublingual monomeric allergoid tablets in house dust mite-allergic patients. Allergy 2016, 72, 77–84. [Google Scholar] [CrossRef] [Green Version]
  76. Kopp, M.V.; Bovermann, X.; Klimek, L. Accelerated Dose Escalation with Three Injections of an Aluminum Hydroxide-Adsorbed Allergoid Preparation of Six Grasses Is Safe for Patients with Moderate to Severe Allergic Rhinitis. Int. Arch. Allergy Immunol. 2019, 181, 94–102. [Google Scholar] [CrossRef]
  77. Chaker, A.M.; Al-Kadah, B.; Luther, U.; Neumann, U.; Wagenmann, M. An accelerated dose escalation with a grass pollen allergoid is safe and well-tolerated: A randomized open label phase II trial. Clin. Transl. Allergy 2015, 6, 4. [Google Scholar] [CrossRef] [Green Version]
  78. Vinay, T.-N.; Park, C.-S.; Kim, H.-Y.; Jung, S.-J. Toxicity and dose determination of quillaja saponin, aluminum hydroxide and squalene in olive flounder (Paralichthys olivaceus). Vet. Immunol. Immunopathol. 2014, 158, 73–85. [Google Scholar] [CrossRef]
  79. He, P.; Zou, Y.; Hu, Z. Advances in aluminum hydroxide-based adjuvant research and its mechanism. Hum. Vaccines Immunother. 2015, 11, 477–488. [Google Scholar] [CrossRef]
  80. Mannhalter, J.; Neychev, H.; Zlabinger, G.; Ahmad, R.; Eibl, M. Modulation of the human immune response by the non-toxic and non-pyrogenic adjuvant aluminium hydroxide: Effect on antigen uptake and antigen presentation. Clin. Exp. Immunol. 1985, 61, 143. [Google Scholar]
  81. Güven, E.; Duus, K.; Laursen, I.; Højrup, P.; Houen, G. Aluminum Hydroxide Adjuvant Differentially Activates the Three Complement Pathways with Major Involvement of the Alternative Pathway. PLoS ONE 2013, 8, e74445. [Google Scholar] [CrossRef]
  82. Eisenbarth, S.; Colegio, O.; O’Connor, W.; Sutterwala, F.S.; Flavell, R.A. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nat. Cell Biol. 2008, 453, 1122–1126. [Google Scholar] [CrossRef]
  83. Lambrecht, B.N.; Kool, M.; Willart, M.A.; Hammad, H. Mechanism of action of clinically approved adjuvants. Curr. Opin. Immunol. 2009, 21, 23–29. [Google Scholar] [CrossRef]
  84. Zaki, S.R.; Austin, G.E.; Chan, W.C.; Conaty, A.L.; Trusler, S.; Trappier, S.; Lindsey, R.B.; Swan, D.C. Chromosomal localization of the human myeloperoxidase gene by in situ hybridization using oligonucleotide probes. Genes Chromosom. Cancer 1990, 2, 266–270. [Google Scholar] [CrossRef]
  85. Gadermaier, E.; Flicker, S.; Aberer, W.; Egger, C.; Reider, N.; Focke, M.; Vrtala, S.; Kundi, M.; Valenta, R. Analysis of the Antibody Responses Induced by Subcutaneous Injection Immunotherapy with Birch and Fagales Pollen Extracts Adsorbed onto Aluminum Hydroxide. Int. Arch. Allergy Immunol. 2010, 151, 17–27. [Google Scholar] [CrossRef]
  86. Spertini, F.; DellaCorte, G.; Kettner, A.; de Blay, F.; Jacobsen, L.; Jutel, M.; Worm, M.; Charlon, V.; Reymond, C. Efficacy of 2 months of allergen-specific immunotherapy with Bet v 1–derived contiguous overlapping peptides in patients with allergic rhinoconjunctivitis: Results of a phase IIb study. J. Allergy Clin. Immunol. 2016, 138, 162–168. [Google Scholar] [CrossRef] [Green Version]
  87. Guzmán-Fulgencio, M.; Caballero, R.; Lara, B.; Mena, M.; Tejera, M.; Sastre, A.; Subiza, J.-L.; Fernández-Caldas, E.; Casanovas, M. Safety of immunotherapy with glutaraldehyde modified allergen extracts in children and adults. Allergol. Immunopathol. 2017, 45, 198–207. [Google Scholar] [CrossRef]
  88. Garcí-Patos, V.; Pujol, R.M.; Alomar, A.; Cisteró, A.; Curell, R.; Fernández-Figueras, M.T.; de Moragas, J.M. Persistent Subcutaneous Nodules in Patients Hyposensitized With Aluminum-Containing Allergen Extracts. Arch. Dermatol. 1995, 131, 1421–1424. [Google Scholar] [CrossRef]
  89. Principi, N.; Esposito, S. Aluminum in vaccines: Does it create a safety problem? Vaccine 2018, 36, 5825–5831. [Google Scholar] [CrossRef]
  90. Baldrick, P.; Richardson, D.; Wheeler, A.W. Review ofL-tyrosine confirming its safe human use as an adjuvant. J. Appl. Toxicol. 2002, 22, 333–344. [Google Scholar] [CrossRef]
  91. Wheeler, A.; Moran, D.; Robins, B.; Driscoll, A. L-Tyrosine as an Immunological Adjuvant. Int. Arch. Allergy Immunol. 1982, 69, 113–119. [Google Scholar] [CrossRef]
  92. McDougall, S.A.; Heath, M.D.; Kramer, M.F.; Skinner, M.A. Analysis of aluminium in rat following administration of allergen immunotherapy using either aluminium or microcrystalline-tyrosine-based adjuvants. Bioanalysis 2016, 8, 547–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bell, A.; Heath, M.; Hewings, S.; Skinner, M. The adsorption of allergoids and 3-O-desacyl-4′-monophosphoryl lipid A (MPL®) to microcrystalline tyrosine (MCT) in formulations for use in allergy immunotherapy. J. Inorg. Biochem. 2015, 152, 147–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Roger, A.; Malet, A.; Moreno, V.; Parra, A.; Gutiérrez, D.; Lleonart, R.; Moreno, F.; Valero, A.; Navarro, B.; Hinojosa, B.; et al. Real-life effect of a microcrystalline tyrosine adjuvanted mite immunotherapy in patients with allergic rhinitis. Immunotherapy 2020, 12, 53–62. [Google Scholar] [CrossRef] [PubMed]
  95. Roger, A.; Depreux, N.; Jurgens, Y.; Serra, A.T.; Heath, M.D.; Garcia, G.; Skinner, M.A. A novel microcrystalline tyrosine-adsorbed, mite-allergoid subcutaneous immunotherapy: 1-year follow-up report. Immunotherapy 2016, 8, 1169–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Roger, A.; Depreux, N.; Jurgens, Y.; Heath, M.D.; Garcia, G.; Skinner, M.A. A novel and well tolerated mite allergoid subcutaneous immunotherapy: Evidence of clinical and immunologic efficacy. Immun. Inflamm. Dis. 2014, 2, 92–98. [Google Scholar] [CrossRef]
  97. Becker, S.; Zieglmayer, P.; Canto, G.; Fassio, F.; Yong, P.; Acikel, C.; Raskopf, E.; Steveling-Klein, E.H.; Allekotte, S.; Mösges, R. A meta-analysis on allergen-specific immunotherapy using MCT ® (MicroCrystalline Tyrosine)-adsorbed allergoids in pollen allergic patients suffering from allergic rhinoconjunctivitis. Clin. Transl. Allergy 2021, 11, e12037. [Google Scholar] [CrossRef]
  98. National Center for Biotechnology Information Calcium Phosphate | Ca3(PO4)2—PubChem. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Calcium-phosphate (accessed on 12 September 2021).
  99. Jones, S.; Asokanathan, C.; Kmiec, D.; Irvine, J.; Fleck, R.; Xing, D.; Moore, B.; Parton, R.; Coote, J. Protein coated microcrystals formulated with model antigens and modified with calcium phosphate exhibit enhanced phagocytosis and immunogenicity. Vaccine 2013, 32, 4234–4242. [Google Scholar] [CrossRef] [Green Version]
  100. Leynadier, F.; Banoun, L.; Dollois, B.; Terrier, P.; Epstein, M.; Guinnepain, M.-T.; Firon, D.; Traube, C.; Fadel, R.; Andre, C. Immunotherapy with a calcium phosphate-adsorbed five-grass-pollen extract in seasonal rhinoconjunctivitis: A double-blind, placebo-controlled study. Clin. Exp. Allergy 2001, 31, 988–996. [Google Scholar] [CrossRef]
  101. Masson, J.-D.; Thibaudon, M.; Bélec, L.; Crépeaux, G. Calcium phosphate: A substitute for aluminum adjuvants? Expert Rev. Vaccines 2016, 16, 289–299. [Google Scholar] [CrossRef]
  102. Kartal, O.; Güleç, M.; Caliskaner, Z.; Musabak, U.H.; Şener, O. Safety of subcutaneous immunotherapy with inhalant allergen extracts: A single-center 30-year experience from Turkey. Immunopharmacol. Immunotoxicol. 2015, 37, 280–286. [Google Scholar] [CrossRef]
  103. Boghdadi, G.; Hammad, N.; Amer, A.; Sammour, S.; Sorour, S. R848, a Toll-Like Receptors 7 and 8 Agonist, a Potential Therapy for Allergic Rhinitis Patients. Inflamm. Allergy—Drug Targets 2014, 13, 144–149. [Google Scholar] [CrossRef]
  104. Fuchs, B.; Knothe, S.; Rochlitzer, S.; Nassimi, M.; Greweling, M.; Lauenstein, H.-D.; Nassenstein, C.; Müller, M.; Ebensen, T.; Dittrich, A.-M.; et al. A Toll-Like Receptor 2/6 Agonist Reduces Allergic Airway Inflammation in Chronic Respiratory Sensitisation to Timothy Grass Pollen Antigens. Int. Arch. Allergy Immunol. 2010, 152, 131–139. [Google Scholar] [CrossRef]
  105. Schülke, S.; Fiedler, A.-H.; Junker, A.-C.; Flaczyk, A.; Wolfheimer, S.; Wangorsch, A.; Heinz, A.; Beckert, H.; Nagl, B.; Bohle, B.; et al. Critical role of mammalian target of rapamycin for IL-10 dendritic cell induction by a flagellin A conjugate in preventing allergic sensitization. J. Allergy Clin. Immunol. 2018, 141, 1786–1798.e11. [Google Scholar] [CrossRef] [Green Version]
  106. Puggioni, F.; Durham, S.R.; Francis, J.N. Monophosphoryl lipid A (MPLR)* promotes allergen-induced immune deviation in favour of Th1 responses. Allergy 2005, 60, 678–684. [Google Scholar] [CrossRef]
  107. Pfaar, O.; Barth, C.; Jaschke, C.; Hormann, K.; Klimek, L. Sublingual Allergen-Specific Immunotherapy Adjuvanted with Monophosphoryl Lipid A: A Phase I/IIa Study. Int. Arch. Allergy Immunol. 2010, 154, 336–344. [Google Scholar] [CrossRef]
  108. Worm, M.; Ernst, D.; Kraller, M.; Babina, M. The Impact on Allergy-Related Cells of a Birch Pollen Allergoid, with and without Monophosphoryl Lipid A, in Comparison with the Native Equivalent. Int. Arch. Allergy Immunol. 2017, 172, 20–26. [Google Scholar] [CrossRef]
  109. Rosewich, M.; Lee, D.; Zielen, S. Pollinex Quattro: An innovative four injections immunotherapy In allergic rhinitis. Hum. Vaccines Immunother. 2013, 9, 1523–1531. [Google Scholar] [CrossRef] [Green Version]
  110. Rosewich, M.; Girod, K.; Zielen, S.; Schubert, R.; Schulze, J. Induction of Bronchial Tolerance After 1 Cycle of Monophosphoryl-A-Adjuvanted Specific Immunotherapy in Children With Grass Pollen Allergies. Allergy Asthma Immunol. Res. 2016, 8, 257–263. [Google Scholar] [CrossRef] [Green Version]
  111. Crivellaro, M.A.; Senna, G.E.; Pappacoda, A.; Vanzelli, R.; Spacal, B.; Marchi, G.; Recchia, G.; Makatsori, M. Safety of ultrashort-term sit with pollen allergoids adjuvanted by monophosphoryl lipid A: A prospective Italian survey. Eur. Ann. Allergy Clin. Immunol. 2011, 43, 58–60. [Google Scholar]
  112. Worm, M.; Higenbottam, T.; Pfaar, O.; Mösges, R.; Aberer, W.; Gunawardena, K.; Wessiepe, D.; Lee, D.; Kramer, M.; 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]
  113. Musarra, A.; Bignardi, D.; Troise, C.; Passalacqua, G. Long-lasting effect of a monophosphoryl lipid-adjuvanted immunotherapy to parietaria. A controlled field study. Eur. Ann. Allergy Clin. Immunol. 2010, 42, 115–119. [Google Scholar]
  114. Zielen, S.; Gabrielpillai, J.; Herrmann, E.; Schulze, J.; Schubert, R.; Rosewich, M. Long-term effect of monophosphoryl lipid A adjuvanted specific immunotherapy in patients with grass pollen allergy. Immunotherapy 2018, 10, 529–536. [Google Scholar] [CrossRef]
  115. Farrokhi, S.; Mousavi, T.; Arshi, S.; Varasteh, A.; Rezaei, N.; Salekmoghadam, A. Co-Administration of Chenopodium Album Allergens and CpG Oligodeoxy-nucleotides Effects on Peripheral Blood Mononuclear Cells of Patients with Allergic Rhinitis Treated with Intranasal Corticosteroids. Iran J. Allergy Asthma Immunol. 2011, 10, 101–110. [Google Scholar]
  116. Kubo, S.; Yamada, T.; Osawa, Y.; Ito, Y.; Narita, N.; Fujieda, S. Cytosine–phosphate–guanosine-DNA induces CD274 expression in human B cells and suppresses T helper type 2 cytokine production in pollen antigen-stimulated CD4-positive cells. Clin. Exp. Immunol. 2012, 169, 1–9. [Google Scholar] [CrossRef]
  117. Leaker, B.R.; Singh, D.; Lindgren, S.; Almqvist, G.; Eriksson, L.; Young, B.; O’Connor, B. Effects of the Toll-like receptor 7 (TLR7) agonist, AZD8848, on allergen-induced responses in patients with mild asthma: A double-blind, randomised, parallel-group study. Respir. Res. 2019, 20, 1–11. [Google Scholar] [CrossRef]
  118. Prangtaworn, P.; Chaisri, U.; Seesuay, W.; Mahasongkram, K.; Onlamoon, N.; Reamtong, O.; Tungtrongchitr, A.; Indrawattana, N.; Chaicumpa, W.; Sookrung, N. Tregitope-linked Refined Allergen Vaccines for Immunotherapy in Cockroach Allergy. Sci. Rep. 2018, 8, 15480. [Google Scholar] [CrossRef]
  119. Foged, C.; Arigita, C.; Sundblad, A.; Jiskoot, W.; Storm, G.; Frokjaer, S. Interaction of dendritic cells with antigen-containing liposomes: Effect of bilayer composition. Vaccine 2004, 22, 1903–1913. [Google Scholar] [CrossRef]
  120. Meechan, P.; Tungtrongchitr, A.; Chaisri, U.; Maklon, K.; Indrawattana, N.; Chaicumpa, W.; Sookrung, N. Intranasal, Liposome-Adjuvanted Cockroach Allergy Vaccines Made of Refined Major Allergen and Whole-Body Extract of Periplaneta americana. Int. Arch. Allergy Immunol. 2013, 161, 351–362. [Google Scholar] [CrossRef]
  121. 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] [PubMed]
  122. Basomba, A.; Tabar, A.I.; de Rojas, D.H.F.; García, B.E.; Alamar, R.; Olaguíbel, J.M.; del Prado, J.M.; Martín, S.; Rico, P. Allergen vaccination with a liposome-encapsulated extract of Dermatophagoides pteronyssinus: A randomized, double-blind, placebo-controlled trial in asthmatic patients. J. Allergy Clin. Immunol. 2002, 109, 943–948. [Google Scholar] [CrossRef] [PubMed]
  123. Alvarez, M.J.; Echechipía, S.; Garcia, B.; Tabar, A.I.; Martin, S.; Rico, P.; Olaguibel, J.M. Liposome-entrappedD. pteronyssinusvaccination in mild asthma patients: Effect of 1-year double-blind, placebo-controlled trial on inflammation, bronchial hyper-responsiveness and immediate and late bronchial responses to the allergen. Clin. Exp. Allergy 2002, 32, 1574–1582. [Google Scholar] [CrossRef] [PubMed]
  124. Klimek, L.; Schmidt-Weber, C.B.; Kramer, M.F.; Skinner, M.A.; Heath, M.D. Clinical use of adjuvants in allergen-immunotherapy. Expert Rev. Clin. Immunol. 2017, 13, 599–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Thoms, F.; Jennings, G.T.; Maudrich, M.; Vogel, M.; Haas, S.; Zeltins, A.; Hofmann-Lehmann, R.; Riond, B.; Grossmann, J.; Hunziker, P.; et al. Immunization of cats to induce neutralizing antibodies against Fel d 1, the major feline allergen in human subjects. J. Allergy Clin. Immunol. 2019, 144, 193–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. 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]
  127. 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]
  128. Jensen-Jarolim, E.; Roth-Walter, F.; Jordakieva, G.; Pali-Schöll, I. Allergens and Adjuvants in Allergen Immunotherapy for Immune Activation, Tolerance, and Resilience. J. Allergy Clin. Immunol. Pract. 2021, 9, 1780–1789. [Google Scholar] [CrossRef]
  129. Feng, Z.; Yi, X.; Hajavi, J. New and old adjuvants in allergen-specific immunotherapy: With a focus on nanoparticles. J. Cell. Physiol. 2021, 236, 863–876. [Google Scholar] [CrossRef]
  130. Zhao, H.; Verma, D.; Li, W.; Choi, Y.; Ndong, C.; Fiering, S.N.; Bailey-Kellogg, C.; Griswold, K.E. Depletion of T Cell Epitopes in Lysostaphin Mitigates Anti-Drug Antibody Response and Enhances Antibacterial Efficacy In Vivo. Chem. Biol. 2015, 22, 629–639. [Google Scholar] [CrossRef] [Green Version]
  131. Norman, P.S.; Ohman, J.L.; Long, A.A.; Creticos, P.S.; Gefter, M.A.; Shaked, Z.; Wood, R.A.; Eggleston, P.A.; Hafner, K.B.; Rao, P.; et al. Treatment of cat allergy with T-cell reactive peptides. Am. J. Respir. Crit. Care Med. 1996, 154, 1623–1628. [Google Scholar] [CrossRef]
  132. Spertini, F.; Perrin, Y.; Audran, R.; Pellaton, C.; Boudousquié, C.; Barbier, N.; Thierry, A.-C.; Charlon, V.; Reymond, C. Safety and immunogenicity of immunotherapy with Bet v 1–derived contiguous overlapping peptides. J. Allergy Clin. Immunol. 2014, 134, 239–240.e13. [Google Scholar] [CrossRef]
  133. Gunawardana, N.C.; Durham, S.R. New approaches to allergen immunotherapy. Ann. Allergy Asthma Immunol. 2018, 121, 293–305. [Google Scholar] [CrossRef] [Green Version]
  134. Eckl-Dorna, J.; Weber, M.; Stanek, V.; Linhart, B.; Ristl, R.; Waltl, E.E.; Merino, S.V.; 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 IgG4 response. EBioMedicine 2019, 50, 421–432. [Google Scholar] [CrossRef] [Green Version]
  135. Tscheppe, A.; Breiteneder, H. Recombinant Allergens in Structural Biology, Diagnosis, and Immunotherapy. Int. Arch. Allergy Immunol. 2017, 172, 187–202. [Google Scholar] [CrossRef]
  136. Raith, M.; Zach, D.; Sonnleitner, L.; Woroszylo, K.; Focke-Tejkl, M.; Wank, H.; Graf, T.; Kuehn, A.; Pascal, M.; Muñoz-Cano, R.M.; et al. Rational design of a hypoallergenic Phl p 7 variant for immunotherapy of polcalcin-sensitized patients. Sci. Rep. 2019, 9, 7802. [Google Scholar] [CrossRef]
  137. Sancho, A.I.; Wallner, M.; Hauser, M.; Nagl, B.; Himly, M.; Asam, C.; Ebner, C.; Jahn-Schmid, B.; Bohle, B.; Ferreira, F. T Cell Epitope-Containing Domains of Ragweed Amb a 1 and Mugwort Art v 6 Modulate Immunologic Responses in Humans and Mice. PLoS ONE 2017, 12, e0169784. [Google Scholar] [CrossRef] [Green Version]
  138. Schülke, S.; Kuttich, K.; Wolfheimer, S.; Duschek, N.; Wangorsch, A.; Reuter, A.; Briza, P.; Pablos, I.; Gadermaier, G.; Ferreira, F.; et al. Conjugation of wildtype and hypoallergenic mugwort allergen Art v 1 to flagellin induces IL-10-DC and suppresses allergen-specific TH2-responses in vivo. Sci. Rep. 2017, 7, 11782. [Google Scholar] [CrossRef] [Green Version]
  139. De Groot, A.S.; Moise, L.; Terry, F.; Gutierrez, A.; Hindocha, P.; Richard, G.; Hoft, D.F.; Ross, T.M.; Noe, A.R.; Takahashi, Y.; et al. Better Epitope Discovery, Precision Immune Engineering, and Accelerated Vaccine Design Using Immunoinformatics Tools. Front. Immunol. 2020, 11, 442. [Google Scholar] [CrossRef]
  140. Martinez, D.; Cantillo, J.F.; Herazo, H.; Wortmann, J.; Keller, W.; Caraballo, L.; Puerta, L. Characterization of a hybrid protein designed with segments of allergens from Blomia tropicalis and Dermatophagoides pteronyssinus. Immunol. Lett. 2018, 196, 103–112. [Google Scholar] [CrossRef]
  141. Da Silva, E.S.; Aglas, L.; Pinheiro, C.S.; Belitardo, E.M.M.D.A.; Silveira, E.F.; Huber, S.; Torres, R.T.; Wallner, M.; Briza, P.; Lackner, P.; et al. A hybrid of two major Blomia tropicalis allergens as an allergy vaccine candidate. Clin. Exp. Allergy 2020, 50, 835–847. [Google Scholar] [CrossRef]
  142. Martínez, D.; Munera, M.; Cantillo, J.F.; Wortmann, J.; Zakzuk, J.; Keller, W.; Caraballo, L.; Puerta, L. An Engineered Hybrid Protein from Dermatophagoides pteronyssinus Allergens Shows Hypoallergenicity. Int. J. Mol. Sci. 2019, 20, 3025. [Google Scholar] [CrossRef] [Green Version]
  143. 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]
  144. 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]
  145. Saarne, T.; Neimert-Andersson, T.; Grönlund, H.; Jutel, M.; Gafvelin, G.; van Hage, M. Treatment with a Fel d 1 hypoallergen reduces allergic responses in a mouse model for cat allergy. Allergy 2010, 66, 255–263. [Google Scholar] [CrossRef]
  146. Klimek, L.; Pfaar, O.; Worm, M. New opportunities for allergen immunotherapy using synthetic peptide immuno-regulatory epitopes (SPIREs). Expert Rev. Clin. Immunol. 2016, 12, 1123–1135. [Google Scholar] [CrossRef]
  147. Maguirea, P.; Nicodemusb, C.; Robinsonb, D.; Aaronson, D.; Umetsu, D.T. The Safety and Efficacy of ALLERVAX CAT in Cat Allergic Patients. Clin. Immunol. 1999, 93, 222–231. [Google Scholar] [CrossRef]
  148. Worm, M.; Lee, H.-H.; Kleine-Tebbe, J.; Hafner, R.P.; Laidler, P.; Healey, D.; Buhot, C.; Verhoef, A.; Maillère, B.; Kay, A.B.; et al. Development and preliminary clinical evaluation of a peptide immunotherapy vaccine for cat allergy. J. Allergy Clin. Immunol. 2011, 127, 89–97.e14. [Google Scholar] [CrossRef]
  149. Couroux, P.; Patel, D.; Hafner, R.P.; Armstrong, K.; Larche, M. Fel d 1-derived synthetic peptide immuno-regulatory epitopes show a long-term treatment effect in cat allergic subjects. Clin. Exp. Allergy 2015, 45, 974–981. [Google Scholar] [CrossRef] [Green Version]
  150. Rudulier, C.D.; Tonti, E.; James, E.; Kwok, W.W.; Larché, M. Modulation of CRTh2 expression on allergen-specific T cells following peptide immunotherapy. Allergy 2019, 74, 2157–2166. [Google Scholar] [CrossRef] [Green Version]
  151. Senti, G.; Crameri, R.; Kuster, D.; Johansen, P.; Martinez-Gomez, J.M.; Graf, N.; Steiner, M.; Hothorn, L.A.; Grönlund, 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]
  152. Grönlund, H.; Gafvelin, G. Recombinant Bet v 1 vaccine for treatment of allergy to birch pollen. Hum. Vaccines 2010, 6, 970–977. [Google Scholar] [CrossRef]
  153. Niederberger, V.; Horak, F.; Vrtala, S.; Spitzauer, S.; Krauth, M.-T.; Valent, P.; Reisinger, J.; Pelzmann, M.; Hayek, B.; Kronqvist, M.; et al. Vaccination with genetically engineered allergens prevents progression of allergic disease. Proc. Natl. Acad. Sci. USA 2004, 101, 14677–14682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Reisinger, J.; Horak, F.; Pauli, G.; van Hage, M.; Cromwell, O.; König, F.; Valenta, R.; Niederberger, V. Allergen-specific nasal IgG antibodies induced by vaccination with genetically modified allergens are associated with reduced nasal allergen sensitivity. J. Allergy Clin. Immunol. 2005, 116, 347–354. [Google Scholar] [CrossRef] [PubMed]
  155. Gafvelin, G.; Thunberg, S.; Kronqvist, M.; Grönlund, H.; Grönneberg, R.; Troye-Blomberg, M.; Akdis, M.; Fiebig, H.; Purohit, A.; Horak, F.; et al. Cytokine and Antibody Responses in Birch-Pollen-Allergic Patients Treated with Genetically Modified Derivatives of the Major Birch Pollen Allergen Bet v 1. Int. Arch. Allergy Immunol. 2005, 138, 59–66. [Google Scholar] [CrossRef] [PubMed]
  156. Campana, R.; Moritz, K.; Neubauer, A.; Huber, H.; Henning, R.; Brodie, T.M.; Kaider, A.; Sallusto, F.; Wöhrl, S.; Valenta, R. Epicutaneous allergen application preferentially boosts specific T cell responses in sensitized patients. Sci. Rep. 2017, 7, 11657. [Google Scholar] [CrossRef] [Green Version]
  157. Meyer, W.; Narkus, A.; Salapatek, A.M.; Hafner, D. Double-blind, placebo-controlled, dose-ranging study of new recombinant hypoallergenic Bet v 1 in an environmental exposure chamber. Allergy 2013, 68, 724–731. [Google Scholar] [CrossRef]
  158. Nony, E.; Bouley, J.; Le Mignon, M.; Lemoine, P.; Jain, K.; Horiot, S.; Mascarell, L.; Pallardy, M.; Vincentelli, R.; Leone, P.; et al. Development and evaluation of a sublingual tablet based on recombinant Bet v 1 in birch pollen-allergic patients. Allergy 2015, 70, 795–804. [Google Scholar] [CrossRef]
  159. Gehlhar, K.; Schlaak, M.; Becker, W.-M.; Bufe, A. Monitoring allergen immunotherapy of pollen-allergic patients: The ratio of allergen-specific IgG4 to IgG1 correlates with clinical outcome. Clin. Exp. Allergy 1999, 29, 497–506. [Google Scholar] [CrossRef]
  160. Cornelius, C.; Schöneweis, K.; Georgi, F.; Weber, M.; Niederberger, V.; Zieglmayer, P.; Niespodziana, K.; Trauner, M.; Hofer, H.; Urban, S.; et al. Immunotherapy With the PreS-based Grass Pollen Allergy Vaccine BM32 Induces Antibody Responses Protecting Against Hepatitis B Infection. EBioMedicine 2016, 11, 58–67. [Google Scholar] [CrossRef] [Green Version]
  161. Valenta, R.; Campana, R.; Niederberger, V. Recombinant allergy vaccines based on allergen-derived B cell epitopes. Immunol. Lett. 2017, 189, 19–26. [Google Scholar] [CrossRef]
  162. Niederberger, V.; Neubauer, A.; Gevaert, P.; Zidarn, M.; Worm, M.; Aberer, W.; Malling, H.J.; Pfaar, O.; Klimek, L.; Pfützner, 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] [Green Version]
  163. Zieglmayer, P.; Focke-Tejkl, M.; Schmutz, R.; Lemell, P.; Zieglmayer, R.; Weber, M.; Kiss, R.; Blatt, K.; Valent, P.; Stolz, F.; et al. Mechanisms, safety and efficacy of a B cell epitope-based vaccine for immunotherapy of grass pollen allergy. EBioMedicine 2016, 11, 43–57. [Google Scholar] [CrossRef] [Green Version]
  164. Rauber, M.M.; Möbs, C.; Campana, R.; Henning, R.; Schulze-Dasbeck, M.; Greene, B.; Focke-Tejkl, M.; Weber, M.; Valenta, R.; Pfützner, 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]
  165. 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. [Google Scholar] [CrossRef] [Green Version]
  166. Freidl, R.; Gstoettner, A.; Baranyi, U.; Swoboda, I.; Stolz, F.; Focke-Tejkl, M.; Wekerle, T.; van Ree, R.; Valenta, R.; Linhart, B. Blocking antibodies induced by immunization with a hypoallergenic parvalbumin mutant reduce allergic symptoms in a mouse model of fish allergy. J. Allergy Clin. Immunol. 2017, 139, 1897–1905.e1. [Google Scholar] [CrossRef] [Green Version]
  167. Storni, F.; Zeltins, A.; Balke, I.; Heath, M.D.; Kramer, M.F.; Skinner, M.A.; Zha, L.; Roesti, E.; Engeroff, P.; Muri, L.; et al. Vaccine against peanut allergy based on engineered virus-like particles displaying single major peanut allergens. J. Allergy Clin. Immunol. 2020, 145, 1240–1253.e3. [Google Scholar] [CrossRef]
  168. 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, 1–15. [Google Scholar] [CrossRef]
  169. Wickman, M.; Lupinek, C.; Andersson, N.; Belgrave, D.; Asarnoj, A.; Benet, M.; Pinart, M.; Wieser, S.; Garcia-Aymerich, J.; Baar, A.; et al. Detection of IgE Reactivity to a Handful of Allergen Molecules in Early Childhood Predicts Respiratory Allergy in Adolescence. EBioMedicine 2017, 26, 91–99. [Google Scholar] [CrossRef]
  170. Campana, R.; Marth, K.; Zieglmayer, P.; Weber, M.; Lupinek, C.; Zhernov, Y.; Elisyutina, O.; Khaitov, M.; Rigler, E.; Westritschnig, K.; et al. Vaccination of nonallergic individuals with recombinant hypoallergenic fragments of birch pollen allergen Bet v 1: Safety, effects, and mechanisms. J. Allergy Clin. Immunol. 2019, 143, 1258–1261. [Google Scholar] [CrossRef] [Green Version]
  171. Geroldinger-Simic, M.; Zelniker, T.; Aberer, W.; Ebner, C.; Egger, C.; Greiderer, A.; Prem, N.; Lidholm, J.; Ballmer-Weber, B.K.; Vieths, S.; et al. Birch pollen–related food allergy: Clinical aspects and the role of allergen-specific IgE and IgG4 antibodies. J. Allergy Clin. Immunol. 2011, 127, 616–622.e1. [Google Scholar] [CrossRef]
  172. De Silva, D.; Geromi, M.; Panesar, S.S.; Muraro, A.; Werfel, T.; Hoffmann-Sommergruber, K.; Roberts, G.; Cardona, V.; Dubois, A.E.J.; Halken, S.; et al. Acute and long-term management of food allergy: Systematic review. Allergy 2013, 69, 159–167. [Google Scholar] [CrossRef] [Green Version]
  173. Treudler, R.; Franke, A.; Schmiedeknecht, A.; Ballmer-Weber, B.; Worm, M.; Werfel, T.; Jappe, U.; Biedermann, T.; Schmitt, J.; Brehler, R.; et al. BASALIT trial: Double-blind placebo-controlled allergen immunotherapy with rBet v 1-FV in birch-related soya allergy. Allergy 2017, 72, 1243–1253. [Google Scholar] [CrossRef] [Green Version]
  174. Kinaciyan, T.; Nagl, B.; Faustmann, S.; Frommlet, F.; Kopp, S.; Wolkersdorfer, M.; Wöhrl, S.; Bastl, K.; Huber, H.; Berger, U.; et al. Efficacy and safety of 4 months of sublingual immunotherapy with recombinant Mal d 1 and Bet v 1 in patients with birch pollen–related apple allergy. J. Allergy Clin. Immunol. 2018, 141, 1002–1008. [Google Scholar] [CrossRef] [Green Version]
  175. Grilo, J.R.; Kitzmüller, C.; Aglas, L.; Acosta, G.S.; Vollmann, U.; Ebner, C.; Horak, F.; Kinaciyan, T.; Radauer, C.; Ferreira, F.; et al. IgE-cross-blocking antibodies to Fagales following sublingual immunotherapy with recombinant Bet v 1. Allergy 2021, 76, 2555–2564. [Google Scholar] [CrossRef]
  176. Tulaeva, I.; Cornelius, C.; Zieglmayer, P.; Zieglmayer, R.; Schmutz, R.; Lemell, P.; Weber, M.; Focke-Tejkl, M.; Karaulov, A.; Henning, R.; et al. Quantification, epitope mapping and genotype cross-reactivity of hepatitis B preS-specific antibodies in subjects vaccinated with different dosage regimens of BM32. EBioMedicine 2020, 59, 102953. [Google Scholar] [CrossRef]
  177. Zhernov, Y.; Curin, M.; Khaitov, M.; Karaulov, A.; Valenta, R. Recombinant allergens for immunotherapy: State of the art. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 402–414. [Google Scholar] [CrossRef]
  178. 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. Vaccines Immunother. 2017, 13, 2804–2813. [Google Scholar] [CrossRef] [Green Version]
  179. Slater, J.E.; Paupore, E.; Zhang, Y.T.; Colberg-Poley, A.M. The latex allergen Hev b 5 transcript is widely distributed after subcutaneous injection in BALB/c mice of its DNA vaccine. J. Allergy Clin. Immunol. 1998, 102, 469–475. [Google Scholar] [CrossRef]
  180. Ibáñez, M.S.; Sastre, J. Molecular allergy diagnosis for the clinical characterization of asthma. Expert Rev. Mol. Diagn. 2015, 15, 789–799. [Google Scholar] [CrossRef]
  181. Thomas, W.R.; Stewart, G.A.; Simpson, R.; Chua, K.Y.; Plozza, T.M.; Dilworth, R.J.; Nisbet, A.; Turner, K.J. Cloning and Expression of DNA Coding for the Major House Dust Mite Allergen Der p 1 in Escherichia coli. Int. Arch. Allergy Immunol. 1988, 85, 127–129. [Google Scholar] [CrossRef]
  182. Sastre, J.; Ibáñez, M.S. Molecular diagnosis and immunotherapy. Curr. Opin. Allergy Clin. Immunol. 2016, 16, 565–570. [Google Scholar] [CrossRef]
  183. Canonica, G.W.; Ansotegui, I.J.; Pawankar, R.; Schmid-Grendelmeier, P.; van Hage, M.; E Baena-Cagnani, C.; Melioli, G.; Nunes, C.; Passalacqua, G.; Rosenwasser, L.; et al. A WAO—ARIA—GA2LEN consensus document on molecular-based allergy diagnostics. World Allergy Organ. J. 2013, 6, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Zuidmeer-Jongejan, L.; Huber, H.; Swoboda, I.; Rigby, N.; Versteeg, S.A.; Jensen, B.M.; Quaak, S.; Akkerdaas, J.H.; Blom, L.; Asturias, J.; et al. Development of a Hypoallergenic Recombinant Parvalbumin for First-in-Man Subcutaneous Immunotherapy of Fish Allergy. Int. Arch. Allergy Immunol. 2015, 166, 41–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Matricardi, P.M.; Kleine-Tebbe, J.; Hoffmann, H.J.; Valenta, R.; Hilger, C.; Hofmaier, S.; Aalberse, R.C.; Agache, I.; Asero, R.; Ballmer-Weber, B.; et al. EAACI Molecular Allergology User’s Guide. Pediatr. Allergy Immunol. 2016, 27 (Suppl. S2), 1–250. [Google Scholar] [CrossRef] [PubMed]
  186. Koch, L.; Laipold, K.; Arzt-Gradwohl, L.; Čerpes, U.; Sturm, E.M.; Aberer, W.; Sturm, G.J. IgE multiplex testing in house dust mite allergy is utile, and sensitivity is comparable to extract-based singleplex testing. Allergy 2020, 75, 2091–2094. [Google Scholar] [CrossRef] [PubMed]
  187. Kleine-Tebbe, J.; Jappe, U. Molecular allergy diagnostic tests: Development and relevance in clinical practice. Allergol. Sel. 2017, 1, 169–189. [Google Scholar] [CrossRef] [PubMed]
  188. Armentia, A.; Herrero, M.; Martín-Armentia, B.; Rihs, H.-P.; Postigo, I.; Martínez-Quesada, J. Molecular diagnosis in cannabis allergy. J. Allergy Clin. Immunol. Pract. 2014, 2, 351–352. [Google Scholar] [CrossRef]
  189. Huerta-Ocampo, J.Á.; Valenzuela-Corral, A.; Robles-Burgueño, M.D.R.; Guzmán-Partida, A.M.; Oñate, M.; Ángel, H.; Vázquez-Moreno, L.; Pavón-Romero, G.F.; Terán, L.M. Proteomic identification of allergenic proteins in red oak (Quercus rubra) pollen. World Allergy Organ. J. 2020, 13, 100111. [Google Scholar] [CrossRef]
  190. Mani, B.M.; Huerta-Ocampo, J.A.; Garcia-Sanchez, J.R.; Barrera-Pacheco, A.; de la Rosa, A.P.B.; Teran, L.M. Identification of Ligustrum lucidum pollen allergens using a proteomics approach. Biochem. Biophys. Res. Commun. 2015, 468, 788–792. [Google Scholar] [CrossRef]
  191. Morales-Amparano, M.B.; Valenzuela-Corral, A.; Montfort, G.R.-C.; Vázquez-Moreno, L.; Escobedo-Moratilla, A.; Pastor-Palacios, G.; Ovando-Vázquez, C.; Teran, L.M.; Huerta-Ocampo, J. Ángel Immunoproteomic identification of allergenic proteins in pecan (Carya illinoinensis) pollen. J. Proteom. 2021, 248, 104348. [Google Scholar] [CrossRef]
  192. Neethirajan, S.; Weng, X.; Tah, A.; Cordero, J.; Ragavan, K. Nano-biosensor platforms for detecting food allergens—New trends. Sens. Bio-Sens. Res. 2018, 18, 13–30. [Google Scholar] [CrossRef]
  193. Morales-Amparano, M.B.; Huerta-Ocampo, J.; Huerta-Ocampo, J.Á.; Pastor-Palacios, G.; Teran, L.M. The Role of Enolases in Allergic Disease. J. Allergy Clin. Immunol. Pract. 2021, 9, 3026–3032. [Google Scholar] [CrossRef]
  194. Martin, J.G.; Panariti, A. Fenotipos del asma, ¿son importantes? Arch. Bronconeumol. Engl. Ed. 2017, 53, 177–179. [Google Scholar] [CrossRef]
Figure 1. Schemes of allergen immunotherapy. (a) Classical subcutaneous immunotherapy scheme consisting of build-up and maintenance phase. (b) Recombinant scheme with grass (c) Recombinant scheme with birch allergen.
Figure 1. Schemes of allergen immunotherapy. (a) Classical subcutaneous immunotherapy scheme consisting of build-up and maintenance phase. (b) Recombinant scheme with grass (c) Recombinant scheme with birch allergen.
Cells 11 00212 g001
Figure 2. Mechanism of allergen-specific immunotherapy. (a) The allergic response begins with the allergen being endocytosed by the dendritic cells of the airway epithelium (DC). Subsequently, DC goes to local secondary lymphoid organs (lymph nodes) where the antigen presentation to Th0 happens. Th0 differentiate to Th2 and synthesize its interleukin profile to allow the production of IgE-type specific allergen antibodies. (b) Subcutaneous, sublingual, and intralymphatic immunotherapies (SCIT, SLIT, IIT) provides the antigen (peptide, recombinant, or protein complex) and induces T naive cells differentiation into different types, which synthesizes their interleukin profiles as Th1 (IFN-γ) or Treg Foxp3+ (IL-10 and TGF-β). TFH CXCR5+ (IL-21 and IL-4) profiles co-helping plasmatic cells to produce IgA1, IgA2, and IgG4 antibodies block against IgE-allergen specific.
Figure 2. Mechanism of allergen-specific immunotherapy. (a) The allergic response begins with the allergen being endocytosed by the dendritic cells of the airway epithelium (DC). Subsequently, DC goes to local secondary lymphoid organs (lymph nodes) where the antigen presentation to Th0 happens. Th0 differentiate to Th2 and synthesize its interleukin profile to allow the production of IgE-type specific allergen antibodies. (b) Subcutaneous, sublingual, and intralymphatic immunotherapies (SCIT, SLIT, IIT) provides the antigen (peptide, recombinant, or protein complex) and induces T naive cells differentiation into different types, which synthesizes their interleukin profiles as Th1 (IFN-γ) or Treg Foxp3+ (IL-10 and TGF-β). TFH CXCR5+ (IL-21 and IL-4) profiles co-helping plasmatic cells to produce IgA1, IgA2, and IgG4 antibodies block against IgE-allergen specific.
Cells 11 00212 g002
Figure 3. Method for synthesizing recombinant allergens. (A) Identification of the amino acid sequence of the proteins associated with allergic symptoms (allergen); (B) Isolation of the messenger RNA through the use of the genetic code and creation of the successive complementary DNA (cDNA) with the reverse transcriptase enzyme constituting the specific gene for this protein; (C) Insertion of the cDNA sequence into the bacterial genetic material (Escherichia coli) and polymerization of the recombinant cDNA, (D) Insertion of the recombinant cDNA into the host microorganism with the subsequent synthesis of hypoallergenic recombinant allergens, (E) Evaluation in clinical-immunology studies.
Figure 3. Method for synthesizing recombinant allergens. (A) Identification of the amino acid sequence of the proteins associated with allergic symptoms (allergen); (B) Isolation of the messenger RNA through the use of the genetic code and creation of the successive complementary DNA (cDNA) with the reverse transcriptase enzyme constituting the specific gene for this protein; (C) Insertion of the cDNA sequence into the bacterial genetic material (Escherichia coli) and polymerization of the recombinant cDNA, (D) Insertion of the recombinant cDNA into the host microorganism with the subsequent synthesis of hypoallergenic recombinant allergens, (E) Evaluation in clinical-immunology studies.
Cells 11 00212 g003
Table 1. Examples of new immunotherapy molecules.
Table 1. Examples of new immunotherapy molecules.
AdjuvantsHybrid ProteinsRecombinants
AluminumBTH2 (Blomia tropicalis)MAT Fel d 1 (Cat)
Microcrystaline TyrosineDPx4 (Dermatophagoides pteronyssinus)CatPAD (Cat)
Calcium PhosphateMAVAC-BD-2 (Blomia tropicalis and Dermatophagoides sp.)REGN1908 (Cat)
Toll-like Receptors rBet v 1 FV (Birch)
Liposomes rBet v 1 (Birch)
Virus Like Particles BM32 (Grass)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pavón-Romero, G.F.; Parra-Vargas, M.I.; Ramírez-Jiménez, F.; Melgoza-Ruiz, E.; Serrano-Pérez, N.H.; Teran, L.M. Allergen Immunotherapy: Current and Future Trends. Cells 2022, 11, 212. https://doi.org/10.3390/cells11020212

AMA Style

Pavón-Romero GF, Parra-Vargas MI, Ramírez-Jiménez F, Melgoza-Ruiz E, Serrano-Pérez NH, Teran LM. Allergen Immunotherapy: Current and Future Trends. Cells. 2022; 11(2):212. https://doi.org/10.3390/cells11020212

Chicago/Turabian Style

Pavón-Romero, Gandhi F., Maria Itzel Parra-Vargas, Fernando Ramírez-Jiménez, Esmeralda Melgoza-Ruiz, Nancy H. Serrano-Pérez, and Luis M. Teran. 2022. "Allergen Immunotherapy: Current and Future Trends" Cells 11, no. 2: 212. https://doi.org/10.3390/cells11020212

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