Skip to main content

MINI REVIEW article

Front. Immunol., 04 August 2020
Sec. Immunological Tolerance and Regulation
This article is part of the Research Topic Novel Advances in Allergy Diagnosis and Treatment View all 21 articles

Formulations for Allergen Immunotherapy in Human and Veterinary Patients: New Candidates on the Horizon

\nIsabella Pali-Schll,*&#x;Isabella Pali-Schöll1,2*Douglas J. DeBoer&#x;Douglas J. DeBoer3Claudia Alessandri&#x;Claudia Alessandri4Ahmed Adel SeidaAhmed Adel Seida5Ralf S. Mueller&#x;Ralf S. Mueller6Erika Jensen-Jarolim&#x;Erika Jensen-Jarolim2
  • 1University of Veterinary Medicine, Vienna, Austria
  • 2Institute of Pathophysiology and Allergy Research, Center of Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
  • 3Dermatology/Allergy Section, Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
  • 4Associated Centers for Molecular Allergology (CAAM), Rome, Italy
  • 5Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
  • 6Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany

Allergen immunotherapy is currently the only causal treatment for allergic diseases in human beings and animals. It aims to re-direct the immune system into a tolerogenic or desensitized state. Requirements include clinical efficacy, safety, and schedules optimizing patient or owner compliance. To achieve these goals, specific allergens can be formulated with adjuvants that prolong tissue deposition and support uptake by antigen presenting cells, and/or provide a beneficial immunomodulatory action. Here, we depict adjuvant formulations being investigated for human and veterinary allergen immunotherapy.

Introduction

Allergen immunotherapy (AIT) is currently the only causative treatment for allergic diseases of animals and man. Subcutaneous administration of allergen extracts—with or without an aluminum hydroxide adjuvant—historically has proven efficacious for many allergic patients. However, recent studies suggest that desensitizing properties of the allergen potentially can be enhanced by alternate adjuvants or delivery systems, while maintaining freedom from adverse effects.

A number of delivery systems for AIT are currently being investigated (13) and applied in animal models, but rather few human or veterinary clinical studies exist. For nano- (NP) or microparticle (MP) preparations, various particulate compositions are complexed or filled with allergens (3). The particulate materials must be biocompatible (resulting in no adverse reaction) and can either be biodegradable (broken down in the organism) or non-biodegradable. Several non-biodegradable materials tested as delivery systems for allergens in vitro as well as in animal models, such as dendromers/dendrosomes (4), polyethylenimine (5), polypropylene sulfide (6), multiwalled carbon nanotubes (7), gold nanoparticles (8), or fullerenes (9) have been comprehensively reviewed (2). However, their fate in the organism is not absolutely clear and thus must be carefully studied.

We selected here the most promising novel AIT formulations, encompassing both modifications of allergen and inclusion of immunomodulators, and describe their performance in human and veterinary trials.

Formulations With Vehicles That Protect Immunogenicity

For an allergen-specific and prolonged effect of AIT, allergens must reach the immune system in a recognizable form, and be released from any carrier in an optimal, perhaps gradual manner. Thus, one general approach to enhance response to AIT is to protect allergens from degradation, and/or ensure optimal release by packing them into resistant carrier materials.

Methylmethacrylate Coating

Grass pollen allergen was coated with a co-polymer of methacrylic acid and methylmethacrylate, called Eudragit L-100®, to protect against gastric degradation, and administered orally to Guinea pigs (10). The secondary antibody response was greater than with an aqueous solution of the allergen. An encapsulated ragweed allergen extract given to people with hay fever led to an increase of anti-ragweed IgG antibodies, a dampened increase of IgE antibodies, and decreased symptom-medication scores without systemic reactions (11).

Plant Cell-Wall Fusion Proteins

Plant cell-expressed or complexed allergen proteins delivered orally are protected from gastric acid and enzymatic degradation, but are then digested by gut microbes in the colon and release the allergens to the immune system (12). Transgenic rice expressing the major house dust mite (HDM) allergen Der p 1 was developed as an edible AIT product (13). Several other proteins have been used in this fashion to induce tolerance in mice. After oral prophylactic administration of transgenic rice expressing modified Japanese cedar pollen allergens Cry j 1 and Cry j 2 (14, 15) to BALB/c mice or HDM allergen Der p 2 in transgenic tobacco in a murine asthma model (16), a decreased allergic response was uniformly seen. Chemically modified ragweed pollen shells fed to BALB/c mice were incorporated in the subepithelial tissue (17). In addition, bone-marrow derived macrophages and dendritic cells cultured with this pollen increased expression of CD40, CD80, CD86, and MHC class II molecules and secreted proinflammatory cytokines TNF-alpha and IL1-beta. Such studies have not been performed in human or veterinary patients.

Polyanhydrides

Particles made of amphiphilic polyanhydrides are biodegradable and show a favorable safety profile. Poly[methyl vinyl ether-co-maleic anhydride] (Gantrez® AN 119) has been investigated in mouse models for oral immunotherapy against peanut allergy (1820), cashew nut allergy (21), and Lolium perenne pollen allergy (22). Three doses of nanoparticle-coated peanut allergens were able of protecting CD1 mice against severe anaphylaxis induced by a peanut challenge (18). Similarly, in CD1 mice presensitized to peanut, AIT with nanoparticle-encapsulated peanut allergen was associated with significantly lower concentrations of mMCPT-1, and an increased survival rate after challenge, compared to AIT with free peanut extract (20). Similar results were seen with allergens of L. perenne combined with Gantrez nanoparticles and LPS of Brucella ovis (22). Oral administration of cashew nut-loaded nanoparticles to BALB/c mice led to a decrease in splenic Th2 cytokines, and an enhancement of pro-Th1 and regulatory cytokines with an increased expansion of T regulatory cells compared to mice immunized with free allergens (21). Despite promising results in these murine models, no published studies in human or veterinary patients exist.

Evidence thus indicates that formulations protecting the allergens are beneficial, and show Th1- and Treg-inducing capacity.

Allergens Administered With Novel Adjuvants

A different approach incorporates adjuvants along with the allergen, with the goal of enhancing a desirable, non-allergic immune response, optimally counteracting an allergy-immune milieu.

Monophosphoryl Lipid A (MPLA)

Monophosphoryl lipid A (MPLA) is a compound derived from Gram-negative bacteria and effectively applied in human allergic patients since 1975 (23). In vitro studies indicate that it may also induce the secretion of Th1 cytokines from equine cells, thus making it a candidate for the treatment of insect bite hypersensitivity (IBH) (24). Twelve healthy Icelandic horses were immunized with Culicoides nubeculosus allergens adjuvanted with MPLA plus alum, or alum alone (25). When their peripheral blood mononuclear cells (PBMCs) were stimulated, the MPLA/alum-immunized horses produced more IFN-gamma and IL-10, both preferable in allergy.

Gelatin-CpG-ODN

Gelatin particles combined with CpG-ODN (GbpCpG) are among the few preparations already studied in veterinary allergy patients, including canine atopic dermatitis (26, 27) and equine recurrent airway obstruction (RAO, an analog of human asthma) (2830). Uptake of these particles by canine PBMCs could be demonstrated with confocal laser scanning microscopy, and an increase of IL-10 secretion could be shown when cells were incubated with GbpCpG compared to CpG-ODN alone (27). Atopic dogs improved clinically after subcutaneous administration of GbpCpG, while their IL-4 expression decreased (26). Bronchoalveolar lavage cells from RAO horses were incubated with different CpG-ODN sequences; IL-10 and IFN-gamma release was increased, while IL-4 decreased (30). When nebulized with a gelatin nanoparticle-based CpG-ODN formulation, horses with RAO improved clinically and the IL-10 concentration increased in their bronchoalveolar lavage fluid (28). In a subsequent placebo-controlled trial, this treatment caused a persistent decrease of allergic clinical variables in horses treated with nebulized GbpCpG (29). A later study described lyophylisation of GbpCpG facilitating its storage and use (31).

Triacedimannose (TADM)

Incubation of the synthetic trivalent glycocluster TADM with birch-stimulated PBMC of allergic rhinitis patients suppressed the production of all Th2-type cytokines (32). TADM suppressed IgE production and enhanced IFN-gamma production in a mouse model of OVA-induced allergic asthma (32). Intranasal application of TADM and timothy grass pollen extract to sensitized BALB/c mice led to a much greater decrease in lymphocyte and eosinophil counts in blood, BALF, and lung biopsies compared to CpG-ODN and MPLA, and (in contrast to CpG-ODN alone) did not increase neutrophil counts (33).

Polysaccharide Polymers

Carbohydrate-based particles complexed with Phl p 5 grass pollen allergen or cat allergen Fel d 1 were successfully used in several studies of AIT in mice (3437).

The polyaminosaccharide chitosan (poly-D-glucosamine) is approved for use in human wound healing, but is not yet evaluated for AIT. Chitosan particles were used with ovalbumin as a mucoadhesive to promote uptake by oromucosal dendritic cells in vitro (38), and also with allergens from HDM and peanut in mouse models to augment AIT (3941).

Other polysaccharides used for preparation of particulate delivery systems are dextran, alginate, starch, and cellulose derivates. Amylopectin-based microparticles were formulated with Bet v 1 from birch pollen for sublingual treatment of allergic mice (42). Mannan-dextran-maltodextrin covalently attached to OVA and papain were intradermally injected into BALB/c mice, leading to elevated humoral immune responses, and an IgE-to-IgG-shift (43). Another potentially useful polysaccharide is pullulan, a polysaccharide which, coupled to HDM allergen Der p 2, and administered to dogs, effectively reduced clinical signs of atopic dermatitis (44). Carbohydrate-modified ultrafine ceramic-core based nanoparticles, so-called aquasomes, are not biodegradable, and have been applied in the mouse model with ovalbumin (OVA) as model allergen preparation for intradermal application (45).

Heat-Labile Toxin (LT) From E. coli

A patch delivery system for birch pollen allergen rBet v 1 with and without heat-labile toxin (LT) from Escherichia coli was compared to subcutaneus alum-adsorbed rBet v 1 in a guinea pig model (46). Only the rBet v 1-LT was able to induce allergen-specific blocking IgG antibodies comparable to subcutaneous immunization.

Miscellaneous Particulate Formulations

Strontium-doped hydroxyapatite porous spheres (SHAS) have been used with OVA subcutaneously in a mouse model and led to a sustained stimulation of both CD4+ and CD8+ T cells (47). AIT with SHAS-OVA showed a higher efficacy as assessed by symptom scores compared to soluble OVA. This approach was not tested clinically in human or veterinary patients.

Poly(epsilon-caprolactone; PCL) is a biocompatible adjuvant, and in mice sensitized to OVA led to lower IgE, fewer anaphylactic reactions, and higher survival rate compared to alum-adjuvant treated animals (48). Studies in human and veterinary patients are lacking.

Modified difunctional water-soluble PEG dimethacrylate (PEG-acetal-DMA) macromonomers have cleavable acetal units (49), and when those were filled with allergen (OVA, grass pollen allergen, HDM allergen) and encapsulated into liposomes, they could avoid IgE-dependent activation of basophils in vitro, but were taken up by dendritic cells (50).

Poly-glutamic acid particles (PGA) were used with Phleum pretense pollen extract in vitro and increased allergen-specific IL-10 production and proliferation of autologous CD4+ memory T cells (51). Other investigators have shown that PGA per se is an allergen in fermented soybeans, which causes hypersensitivity reactions and even late-onset anaphylaxis (5254). To the authors' knowledge, there are no studies evaluating PGA in animals.

Protamine-based nanoparticles are biodegradable and biocompatible arginine-rich peptides. When complexed with Ara h 2 from peanut and CpG-ODN, they could counteract a Th2-dominated allergen-induced immune response in mice (55). A combination of liposomes with protamine and DNA was also proven effective in combating Chenopodium album allergy in a mouse model (56). At this point, there are no published clinical studies with protamine-based nanoparticles.

Mesoporous silica nanoparticles were successfully used in allergy models (57) with HDM allergen Der f 2 for subcutaneous prophylactic treatment of mice (58). However, when applied epicutaneously with mite extract in the form of agglomerates, they induced AD-like skin lesions and promoted IgE-responses (59). Studies in human and veterinary patients are lacking.

Taken together, many of novel adjuvants have shown Th1-promoting capacity in vitro and in vivo in murine models and even veterinary patient studies for horses and dogs. They were capable of counter-acting IgE, inducing preferentially IFN-γ, and/or IL-10 and also resulting in reduced symptom scores, being more effective than their non-adjuvanted controls.

Allergens Coupled to Immunomodulators

Efforts have also been made to enhance an overall shift in the immune response away from Th2, while at the same time presenting the offending allergen. Some approaches incorporate elements that can redirect the overall immune response from an allergy-prone Th2-IgE-milieu to a more Th1-IgG-dominated reponse.

Modified Adenine Conjugates

Der p 2 allergen bound to 8-OH-modified adenine (nDer p2-Conj) forms an allergen-TLR7 agonist conjugate. When injected subcutaneously, it reduces allergen challenge-induced murine airway inflammation (6062), triggers TLR7, redirects allergen-specific Th2 responses, and promotes a Th1 response as well as an increase in IL-10 with prolonged effects.

Mannan-Modified Allergens and Allergoids

Mannan preparations, alone or allergen-conjugated, appear capable of downregulating IgE responses. Konjac glucomannan (Amorphophallus konjac) fed to BALB/c mice suppressed IgE class switching in B cells and inhibited Th1 and Th2 responses (63). It also suppressed IgE production and clinical signs in a mouse model of allergic rhinitis (64). Administration of neoglycocomplexes of mannan with ovalbumin and papain to sensitized mice led to a class switch from IgE to IgG, and to a decrease in basophil degranulation in vitro (43).

Polymerized allergoids have been coupled to non-oxidized mannan from Saccharomyces cerevisae (PM-allergoids); this is one of the few modified allergen preparations that has been studied in dogs (65). Dendritic cells capture PM-allergoids better than native allergens and enhance Th1/Treg cell responses upon subcutaneous or sublingual administration (66, 67). Interestingly, the addition of alum may impair their tolerogenic properties (68).

DNA Engineered Hybrids With Copolymers

Hybrid allergen molecules are obtained by combining the epitopes of several allergens. Subsequently, their immunogenicity can be enhanced by coupling with copolymers. Engineered hybrids expressing the major allergens from Parietaria pollen allergens Par j 1 and Par j 2 were prepared as nanoaggregated copolymers with poly (hydroxyethyl)-aspartamide (PHEA). They are biodegradable, water-soluble and showed low cytotoxicity, no effect on hemolysis, and no non-specific activation of basophils. Basophil activation properties were, however, maintained in cells from Parietaria-allergic subjects, indicating preserved crosslinking capability of the hybrid allergen (69). No in vivo studies have been reported with this preparation.

Allergen Linked to CpG Oligodeoxynucleotides (CpG-ODN)

CpG-ODN are short, single-stranded synthetic DNA molecules with immunostimulatory properties that induce a Th1-based immune response (70), which prevents Th2-mediated hypersensitivity in mouse models of allergic diseases such as allergic rhinitis (71), asthma (72), conjunctivitis (73), and anaphylactic shock (74). Purified Amb a 1 from Ambrosia artemisiifolia pollen linked to CpG-ODN was successfully tested subcutaneously in humans and resulted in a shift from Th2 to Th1 with an increase of IFN-γ and a decrease in IL-5, proving suitable as an agent for immune redirection in immediate hypersensitivity diseases (75).

Siderophore-Bound Iron or Retinoic Acid as Immunomodulatory Ligands

Bos d 5 cow milk allergen is capable of binding iron via siderophores. The immunomodulatory properties of iron-bound allergen were tested in vitro with human PBMC (76). The empty apo-form of Bos d 5 increased CD4+ cells, IL-13, and IFN-gamma, whereas the complexed holo-form decreased CD4+ cells and induced apopotosis. Similarly, only the apo-form of birch pollen allergen Bet v 1 led to an increase in IL-13, while IFN-gamma was increased with both formulations when incubated with human PBMC (77). Accordingly, spiking of Bet v 1 or Bos d 5 with iron may be an effective approach to improve the efficacy of AIT against birch pollen and cow milk allergy, respectively (76, 77).

The major allergen Bos d 5 was also complexed with the vitamin A metabolite retinoic acid (78). IgE binding was not influenced, but PBMCs from healthy people stimulated with the complex led to a decrease of CD4+ T cells as well as IFN-gamma, IL-13 and IL-10, although induction of CD4+CD25+Foxp3+ regulatory T cells was not seen (79). In contrast to apo-Bos d 5, a highly allergenic molecule, holo-Bos d 5 thus seems to have reduced immunogenicity.

Expression of Allergens by Bacterial Vectors

Streptococcus thermophilus (ST) expressing rBet v 1 was evaluated in a mouse model (80). BALB/c mice were sensitized with rBet v 1 and then treated orally with either ST, ST and rBet v 1, or ST expressing rBet v 1. After aerosol challenge, T regulatory cells, IL-10, and IFN-gamma were increased with the expressed-allergen preparation; bronchial eosinophilia, allergen-induced IL-4, and the rBet v 1-specific IgE/IgG2 ratio were decreased, indicating a shift from Th2 to Th1 and Treg immune responses (80).

Profilin (Che a 2), the major allergen of C. album, was expressed in Lactobacillus lactis, and was bound by human anti-profilin IgE (81). However, bacterial survival was greatly reduced with low pH and simluated gastric and intestinal juices. Oral vaccination with recombinant Lactobacillus plantarum expressing the Japanese Cedar pollen allergen Cry j 1 led to a suppressed allergen-specific IgE response and decreased nasal symptoms in a murine model of allergic rhinitis (82).

Allergens Conjugated to Bacterial Products

Bacterial surface S-layer proteins (SLPs) are two-dimensional crystalline arrays of glycoprotein subunits present on the outermost layer of many bacteria, and have strong adjuvant properties. Conjugating recombinant allergens with SLPs leads to strongly reduced IgE-binding activity and promotes the induction of allergen-specific Th0/1 cells and regulatory T cells. This type of allergen modification has been attempted with inhalant allergens (83). Subsequently, bacterial S-layers have been studied as carriers for peanut allergen-derived peptides (84, 85). A fusion protein of an Ara h 2-derived protein and an S-layer protein was recognized by Ara h 2-specific IgE of human patients but was not able to degranulate sensitized rat basophils in vitro (84). The A20, tumor necrosis factor-induced protein 3 (TNFAIP3), is a ubiquitin-modifying protein playing a defensive role in the pathogenesis of allergic diseases. A DNA vaccine coexpressing Der p 2 and ubiquitin A20 encapsulated into nanoparticles used intranasally in a murine model of allergic rhinitis was able to inhibit allergen-specific IgE, IL-4, IL-10, and IL-17 secretion and to increase IgG1, IgG2a, and IFN-γ (86, 87).

A genetically engineered inhalative cholera toxin B subunit/allergen fusion molecule, CTB-Bet v 1, was shown to improve the immunomodulatory capacity of the mucosal delivery system better than chemically coupled products (88).

Overall, the concept of redirecting the immune response from a Th2 to a Th1-bias as part of AIT has promise. However, most immunomodulatory components—except for CpG-ODN—have been tested in murine models only, and need to be further tested in human and veterinary patients.

Combination and Miscellaneous Approaches

Several formulations combine the enhancing and modulating effect on the immune response, in parallel to protecting the antigen from degradation or digestion, and further releasing it in a delayed manner. Different particulate formulations together with immune-cell targeting substances have been used for these attempts.

Liposomes

Liposomes are bilayers of phospholipids, forming vesicles which can transport aqueous substances inside. They are biocompatible, biodegradable, and can be co-formulated with oligomannose coats, a preparation that was tested in human HDM-allergic asthma patients (89). Mouse models were used to study the efficacy of liposomes in treating allergy against Japanese cedar pollen (90), HDM (91), cat (92), OVA (93), or cockroach (94). Lipid nanoparticles together with Parietaria allergen Par j 2 were characterized biochemically and biophysically (95). Liposome complexes with CpG-DNA and individual allergen extracts were used intradermally for treatment of canine atopic dermatitis after failure of conventional AIT (96). Pruritus improved and IL-4 production decreased with treatment (96). Chronic rhinitis in adult cats could be treated with feline IL-2-filled liposomes plus DNA, although a Th2 bias could not be identified in those cats (97). Liposomes with HDM allergens Der p 1 or Der p 2 reduced clinical and medication scores, skin test responses, and bronchial challenge responses in asthmatic patients (89).

Poly-Lactic-Co-Glycolic Acid Particles (PLGA, PLG, PLA)

These polyesters are approved for use in people as absorbable surgical suture. In mouse models for birch allergy, they were successfully administered subcutaneously with Bet v 1 (98, 99). In addition, PLGA-microparticles were used orally with different plant lectins e.g., Aleura aurantia lectin, wheat-germ agglutinin or Ulex europaeus-I, or Vibrio cholerae neuraminidase to target mucosal cells for enhanced uptake (100103). Other allergens used with PLGA-particles in animal models via different routes are the Chenopodium allergen rChe a 3, as sublingual immunotherapy in a mouse model of allergic rhinitis (104, 105), Ole e 1 from olive pollen or T cell epitopes thereof for intranasal prevention (106, 107), bee venom allergen PLA2 (108), pollen-profilin from palm Caryota mitis (109), Der p 2 from HDM (110), peanut extract (111), or beta-lactoglobulin from milk whey (112). PLGA locally induced a regulatory T cell response via the incorporated mediator substances TGF-beta-1, rapamycin, and IL-2 to prevent a subsequent contact dermatitis reaction (113). In addition to complexing PLGA-particles with allergens, PLGA were complexed with immune-modulating substances such as CpG-ODN for allergy and asthma prevention (114) and with Der p 2-A20 DNA in allergic rhinitis (87) in mouse models. There are no studies in companion animals with PLGA.

Virus-Like Particles (VLP)

Virus-like particles are used as carriers for allergens, or without antigen for antigen-independent immunomodulation (115). Particles consisting of bacteriophage coat proteins and a TLR-9 agonist, but without allergen, were injected into HDM-allergic patients and led to lower symptom-medication scores, higher quality of life and better allergen tolerance (116). A second study with A-type CpG-ODN and HDM-extract showed similar results; allergen-specific IgG increased as well (117). Recently, equine IBH was safely treated with IL-5-linked VLP made from cucumber mosaic virus to induce auto-antibodies against IL-5 (118121). Clinical signs of treated horses improved and their eosinophilia was decreased compared to controls. The same principle was used successfully with IL-31-linked VLP for treatment of IBH in horses and for atopic dermatitis in dogs (122, 123). A very interesting approach is the immunization of cats with Fel d 1-VLPs (HypoCatTM) to induce a neutralizing antibody response in the animal against its own Fel d 1-protein for protection of humans against cat allergy (124, 125). In BALB/c mice, adeno-associated VLP were also tested with an OVA-derived B cell epitope (126), with Art v 1 from mugwort (127) and with peanut allergens Ara h 1 and Ara h 2 (128). Fel d 1 displayed on VLPs failed to induce human mast cell activation in vitro (129). The peptide HDM allergen Der p 1 was coupled to a virus-like particle derived from a bacteriophage and injected in healthy volunteers. Significant IgG responses against the allergen were observed and the vaccine was well-tolerated (130).

Aleuria Aurantia Lectin (AAL)

AAL is derived from the edible orange peel mushroom A. aurantia. When birch pollen-sensitized BALB/c mice were fed with birch pollen-AAL-microspheres, the birch pollen-specific IgG2a, but not IgG1 or IgE increased, as well as IFN-gamma, IL-10, and IL-4 (101). Oral administration of birch pollen-AAL-MS led to an IgG2 antibody response in naive BALB/c mice (102). AAL microspheres may have the potential to serve as a vehicle and adjuvant for oral immunotherapy, potentially stimulating specific mucosal immune responses via M-cell targeting (100).

Wheat Germ Agglutinin (WGA)

Birch-pollen allergens were entrapped in poly(D,L-lactic-co-glycolic acid) microspheres, further coated with WGA to target enterocytes used for oral immunotherapy of type I allergy to protect allergens from digestion and to support intestinal uptake (131). The antigenicity of the birch pollen was maintained at ~60% even after 2 h of simulated gastric digestion, and allergen-specific IgG serum concentrations increased in BALB/c mice fed with the WGA-birch pollen-microspheres (131).

With these approaches, VLP, liposomes, and PLGA particles seem to have promise, and are already tested in human, canine, feline, and equine patients.

Summary

Allergen immunotherapy is the only treatment for allergic diseases that is truly causal and modifies the course of the ongoing disease. As this review discusses, many dozens attempts have been made to identify adjuvants, immunomodulators, physical packaging, conjugates, and combinations of the above to modify allergenic proteins, making them safer, and more efficacious in AIT. Many of the formulations have scarcely progressed beyond in vitro studies, though some show great promise in rodent models. Our task is now to select the most promising candidates, and carry them forward into preclinical studies that can more carefully predict which will translate into clinical benefit. Because many human allergic diseases are found nearly identically in animals, veterinary studies could serve as an elegant precursor to the same investigations in human patients.

Author Contributions

IP-S and EJ-J designed and drafted the manuscript, wrote abstract and introduction. IP-S wrote part on particulate delivery systems. CA worked on allergen modifications. AS contributed immune modulators/activators. RM worked on all the parts completing results and references. DD wrote summary and edited, formatted and finalized MS. All authors contributed to the article and approved the submitted version.

Funding

Research work during preparation of this review was supported by the Austrian Science Fund FWF (project SFB F4606-B28 to EJ-J).

Conflict of Interest

EJ-J declares inventorship in patents on allergen immunotherapy formulation with Biomedical International R+D, Vienna, Austria, of which she is shareholder, and is business partner of Bencard Allergie, Germany as well as AllergyTherapeutics, UK. Within the last 5 years, RM has been a consultant, lecturer, or has received financial support for studies from Artuvet, Greer Laboratories, Heska Laboratories, Nextmune, and Synlab.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This paper results from the Working Group of Comparative and Veterinary Allergology of the European Academy of Allergy and Clinical Immunology/EAACI.

Abbreviations

AIT, allergen immunotherapy; FDA, U.S. Food and Drug Administration; HDM, house dust mite; MP, microparticle; MPLA, monophosphoryl lipid A; NP, nanoparticle; ODN, oligodeoxynucelotides; OVA, ovalbumin; PEG, polyethylene glycol; PGA, poly-glutamic acid; PHEA, poly(hydroxyethyl)-aspartamide; PLGA, poly-lactic-co-glycolic acid; TLR, Toll-like receptor; VLP, virus-like particles; RAO, recurrent airway obstruction; IBH, insect bite hypersensitivity; PBMC, peripheral blood mononuclear cells; WGA, wheat germ agglutinin; SLP, S-layer protein; LT, heat-labile toxin; SHAS, Strontium-doped hydroxyapatite porous spheres; TADM, Triacedimannose.

References

1. Scholl I, Boltz-Nitulescu G, Jensen-Jarolim E. Review of novel particulate antigen delivery systems with special focus on treatment of type I allergy. J Control Release. (2005) 104:1–27. doi: 10.1016/j.jconrel.2004.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Pohlit H, Bellinghausen I, Frey H, Saloga J. Recent advances in the use of nanoparticles for allergen-specific immunotherapy. Allergy. (2017) 72:1461–74. doi: 10.1111/all.13199

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Di Felice G, Colombo P. Nanoparticle-allergen complexes for allergen immunotherapy. Int J Nanomedicine. (2017) 12:4493–504. doi: 10.2147/IJN.S134630

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Balenga NA, Zahedifard F, Weiss R, Sarbolouki MN, Thalhamer J, Rafati S. Protective efficiency of dendrosomes as novel nano-sized adjuvants for DNA vaccination against birch pollen allergy. J Biotechnol. (2006) 124:602–14. doi: 10.1016/j.jbiotec.2006.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Garaczi E, Szabo K, Francziszti L, Csiszovszki Z, Lorincz O, Toke ER, et al. DermAll nanomedicine for allergen-specific immunotherapy. Nanomedicine. (2013) 9:1245–54. doi: 10.1016/j.nano.2013.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Ballester M, Jeanbart L, de Titta A, Nembrini C, Marsland BJ, Hubbell JA, et al. Nanoparticle conjugation enhances the immunomodulatory effects of intranasally delivered CpG in house dust mite-allergic mice. Sci Rep. (2015) 5:14274. doi: 10.1038/srep14274

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Ronzani C, Casset A, Pons F. Exposure to multi-walled carbon nanotubes results in aggravation of airway inflammation and remodeling and in increased production of epithelium-derived innate cytokines in a mouse model of asthma. Arch Toxicol. (2014) 88:489–99. doi: 10.1007/s00204-013-1116-3

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Barreto E, Serra MF, Dos Santos RV, Dos Santos CE, Hickmann J, Cotias AC, et al. Local administration of gold nanoparticles prevents pivotal pathological changes in murine models of atopic asthma. J Biomed Nanotechnol. (2015) 11:1038–50. doi: 10.1166/jbn.2015.2024

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Shershakova N, Baraboshkina E, Andreev S, Purgina D, Struchkova I, Kamyshnikov O, et al. Anti-inflammatory effect of fullerene C60 in a mice model of atopic dermatitis. J Nanobiotechnol. (2016) 14:8. doi: 10.1186/s12951-016-0159-z

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wheeler AW, Henderson DC, Youlten LJ, Al J II, Hickman BE, Taylor IH, et al. Immunogenicity in guinea pigs and tolerance in grass pollen-sensitive volunteers of enteric-coated grass pollen allergens. Int Arch Allergy Appl Immunol. (1987) 83:354–8. doi: 10.1159/000234368

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Litwin A, Flanagan M, Entis G, Gottschlich G, Esch R, Gartside P, et al. Oral immunotherapy with short ragweed extract in a novel encapsulated preparation: a double-blind study. J Allergy Clin Immunol. (1997) 100:30–8. doi: 10.1016/S0091-6749(97)70191-X

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Daniell H, Kulis M, Herzog RW. Plant cell-made protein antigens for induction of oral tolerance. Biotechnol Adv. (2019) 37:107413. doi: 10.1016/j.biotechadv.2019.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Yang L, Kajiura H, Suzuki K, Hirose S, Fujiyama K, Takaiwa F. Generation of a transgenic rice seed-based edible vaccine against house dust mite allergy. Biochem Biophys Res Commun. (2008) 365:334–9. doi: 10.1016/j.bbrc.2007.10.186

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Wakasa Y, Takagi H, Hirose S, Yang L, Saeki M, Nishimura T, et al. Oral immunotherapy with transgenic rice seed containing destructed Japanese cedar pollen allergens, Cry j 1 and Cry j 2, against Japanese cedar pollinosis. Plant Biotechnol J. (2013) 11:66–76. doi: 10.1111/pbi.12007

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Fukuda K, Ishida W, Harada Y, Wakasa Y, Takagi H, Takaiwa F, et al. Efficacy of oral immunotherapy with a rice-based edible vaccine containing hypoallergenic Japanese cedar pollen allergens for treatment of established allergic conjunctivitis in mice. Allergol Int. (2018) 67:119–23. doi: 10.1016/j.alit.2017.06.006

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Lee CC, Ho H, Lee KT, Jeng ST, Chiang BL. Construction of a Der p2-transgenic plant for the alleviation of airway inflammation. Cell Mol Immunol. (2011) 8:404–14. doi: 10.1038/cmi.2011.13

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Uddin MJ, Gill HS. Ragweed pollen as an oral vaccine delivery system: mechanistic insights. J Control Release. (2017) 268:416–26. doi: 10.1016/j.jconrel.2017.10.019

PubMed Abstract | CrossRef Full Text | Google Scholar

18. De Souza Reboucas J, Esparza I, Ferrer M, Sanz ML, Irache JM, Gamazo C. Nanoparticulate adjuvants and delivery systems for allergen immunotherapy. J Biomed Biotechnol. (2012) 2012:474605. doi: 10.1155/2012/474605

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Brotons-Canto A, Gamazo C, Martin-Arbella N, Abdulkarim M, Matias J, Gumbleton M, et al. Evaluation of nanoparticles as oral vehicles for immunotherapy against experimental peanut allergy. Int J Biol Macromol. (2018) 110:328–35. doi: 10.1016/j.ijbiomac.2017.09.109

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Gamazo C, Garcia-Azpiroz M, Souza Reboucas J, Gastaminza G, Ferrer M, Irache JM. Oral immunotherapy using polymeric nanoparticles loaded with peanut proteins in a murine model of fatal anaphylaxis. Immunotherapy. (2017) 9:1205–17. doi: 10.2217/imt-2017-0111

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Pereira MA, Reboucas JS, Ferraz-Carvalho RS, de Redin IL, Guerra PV, Gamazo C, et al. Poly(anhydride) nanoparticles containing cashew nut proteins can induce a strong Th1 and treg immune response after oral administration. Eur J Pharm Biopharm. (2018) 127:51–60. doi: 10.1016/j.ejpb.2018.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Gomez S, Gamazo C, San Roman B, Grau A, Espuelas S, Ferrer M, et al. A novel nanoparticulate adjuvant for immunotherapy with Lolium perenne. J Immunol Methods. (2009) 348:1–8. doi: 10.1016/j.jim.2009.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Jensen-Jarolim E, Bachmann M, Bonini S, Jacobsen L, Jutel M, Klimek L, et al. State-of-the-art in marketed adjuvants and formulations in allergen immunotherapy: a position paper of the european academy of allergy and clinical immunology (EAACI). Allergy. (2019) 75:746–60. doi: 10.1111/all.14134

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Ziegler A, Gerber V, Marti E. In vitro effects of the toll-like receptor agonists monophosphoryl lipid A and CpG-rich oligonucleotides on cytokine production by equine cells. Vet J. (2017) 219:6–11. doi: 10.1016/j.tvjl.2016.11.013

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Jonsdottir S, Svansson V, Stefansdottir SB, Schupbach G, Rhyner C, Marti E, et al. A preventive immunization approach against insect bite hypersensitivity: intralymphatic injection with recombinant allergens in alum or alum and monophosphoryl lipid A. Vet Immunol Immunopathol. (2016) 172:14–20. doi: 10.1016/j.vetimm.2016.02.017

CrossRef Full Text | Google Scholar

26. Wagner I, Geh KJ, Hubert M, Winter G, Weber K, Classen J, et al. Preliminary evaluation of cytosine-phosphate-guanine oligodeoxynucleotides bound to gelatine nanoparticles as immunotherapy for canine atopic dermatitis. Vet Rec. (2017) 181:118. doi: 10.1136/vr.104230

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Prelaud AR, Fuchs S, Weber K, Winter G, Coester C, Mueller RS. In vitro effects of CpG oligodeoxynucleotides delivered by gelatin nanoparticles on canine peripheral blood mononuclear cells of atopic and healthy dogs - a pilot study. Vet Dermatol. (2013) 24:494–e117. doi: 10.1111/vde.12056

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Klier J, Fuchs S, May A, Schillinger U, Plank C, Winter G, et al. A nebulized gelatin nanoparticle-based CpG formulation is effective in immunotherapy of allergic horses. Pharm Res. (2012) 29:1650–7. doi: 10.1007/s11095-012-0686-8

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Klier J, Lehmann B, Fuchs S, Reese S, Hirschmann A, Coester C, et al. Nanoparticulate CpG immunotherapy in RAO-affected horses: phase I and IIa study. J Vet Intern Med. (2015) 29:286–93. doi: 10.1111/jvim.12524

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Klier J, May A, Fuchs S, Schillinger U, Plank C, Winter G, et al. Immunostimulation of bronchoalveolar lavage cells from recurrent airway obstruction-affected horses by different CpG-classes bound to gelatin nanoparticles. Vet Immunol Immunopathol. (2011) 144:79–87. doi: 10.1016/j.vetimm.2011.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Geh KJ, Hubert M, Winter G. Progress in formulation development and sterilisation of freeze-dried oligodeoxynucleotide-loaded gelatine nanoparticles. Eur J Pharm Biopharm. (2018) 129:10–20. doi: 10.1016/j.ejpb.2018.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Makinen K, Mukherjee C, Leino M, Panchadhayee R, Lehto M, Wolff H, et al. A novel mannoside-glycocluster adjuvant: compared in vitro to CpG ODN and MPL and tested in vivo in mouse asthma model. Allergol Immunopathol. (2016) 44:9–17. doi: 10.1016/j.aller.2015.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lehto M, Wolff H, Leino R, Alenius H, Savolainen J. A novel glycocluster molecule prevents timothy-induced allergic airway inflammation in mice. Allergy. (2018) 73:1700–6. doi: 10.1111/all.13419

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Thunberg S, Neimert-Andersson T, Cheng Q, Wermeling F, Bergstrom U, Swedin L, et al. Prolonged antigen-exposure with carbohydrate particle based vaccination prevents allergic immune responses in sensitized mice. Allergy. (2009) 64:919–26. doi: 10.1111/j.1398-9995.2008.01905.x

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Neimert-Andersson T, Thunberg S, Swedin L, Wiedermann U, Jacobsson-Ekman G, Dahlen SE, et al. Carbohydrate-based particles reduce allergic inflammation in a mouse model for cat allergy. Allergy. (2008) 63:518–26. doi: 10.1111/j.1398-9995.2008.01644.x

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Andersson TN, Ekman GJ, Gronlund H, Buentke E, Eriksson TL, Scheynius A, et al. A novel adjuvant-allergen complex, CBP-rFel d 1, induces up-regulation of CD86 expression and enhances cytokine release by human dendritic cells in vitro. Immunology. (2004) 113:253–9. doi: 10.1111/j.1365-2567.2004.01943.x

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Gronlund H, Vrtala S, Wiedermann U, Dekan G, Kraft D, Valenta R, et al. Carbohydrate-based particles: a new adjuvant for allergen-specific immunotherapy. Immunology. (2002) 107:523–9. doi: 10.1046/j.1365-2567.2002.01535.x

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Saint-Lu N, Tourdot S, Razafindratsita A, Mascarell L, Berjont N, Chabre H, et al. Targeting the allergen to oral dendritic cells with mucoadhesive chitosan particles enhances tolerance induction. Allergy. (2009) 64:1003–13. doi: 10.1111/j.1398-9995.2009.01945.x

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Liu Z, Guo H, Wu Y, Yu H, Yang H, Li J. Local nasal immunotherapy: efficacy of dermatophagoides farinae-chitosan vaccine in murine asthma. Int Arch Allergy Immunol. (2009) 150:221–8. doi: 10.1159/000222674

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Li J, Liu Z, Wu Y, Wu H, Ran P. Chitosan microparticles loaded with mite group 2 allergen Der f 2 alleviate asthma in mice. J Investig Allergol Clin Immunol. (2008) 18:454–60.

PubMed Abstract | Google Scholar

41. Roy K, Mao HQ, Huang SK, Leong KW. Oral gene delivery with chitosan–DNA nanoparticles generates immunologic protection in a murine model of peanut allergy. Nat Med. (1999) 5:387–91. doi: 10.1038/7385

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Tourdot S, Airouche S, Berjont N, Moussu H, Betbeder D, Nony E, et al. Efficacy of sublingual vectorized recombinant Bet v 1a in a mouse model of birch pollen allergic asthma. Vaccine. (2013) 31:2628–37. doi: 10.1016/j.vaccine.2013.03.041

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Weinberger EE, Himly M, Myschik J, Hauser M, Altmann F, Isakovic A, et al. Generation of hypoallergenic neoglycoconjugates for dendritic cell targeted vaccination: a novel tool for specific immunotherapy. J Control Release. (2013) 165:101–9. doi: 10.1016/j.jconrel.2012.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Kawano K, Mizuno T. A pilot study of the effect of pullulan-conjugated Der f 2 allergen-specific immunotherapy on canine atopic dermatitis. Vet Dermatol. (2017) 28:583–e141. doi: 10.1111/vde.12470

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Pandey RS, Sahu S, Sudheesh MS, Madan J, Kumar M, Dixit VK. Carbohydrate modified ultrafine ceramic nanoparticles for allergen immunotherapy. Int Immunopharmacol. (2011) 11:925–31. doi: 10.1016/j.intimp.2011.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Cabauatan CR, Campana R, Niespodziana K, Reinisch C, Lundberg U, Meinke A, et al. Heat-labile Escherichia coli toxin enhances the induction of allergen-specific IgG antibodies in epicutaneous patch vaccination. Allergy. (2017) 72:164–8. doi: 10.1111/all.13036

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Garbani M, Xia W, Rhyner C, Prati M, Scheynius A, Malissen B, et al. Allergen-loaded strontium-doped hydroxyapatite spheres improve allergen-specific immunotherapy in mice. Allergy. (2017) 72:570–8. doi: 10.1111/all.13041

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Roman BS, Espuelas S, Gomez S, Gamazo C, Sanz ML, Ferrer M, et al. Intradermal immunization with ovalbumin-loaded poly-epsilon-caprolactone microparticles conferred protection in ovalbumin-sensitized allergic mice. Clin Exp Allergy. (2007) 37:287–95. doi: 10.1111/j.1365-2222.2007.02654.x

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Ewald J, Blankenburg J, Worm M, Besch L, Unger RE, Tremel W, et al. Acid-cleavable poly(ethylene glycol) hydrogels displaying protein release at pH 5. Chemistry. (2020) 26:2947–53. doi: 10.1002/chem.201905310

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Pohlit H, Bellinghausen I, Schomer M, Heydenreich B, Saloga J, Frey H. Biodegradable pH-sensitive poly(ethylene glycol) nanocarriers for allergen encapsulation and controlled release. Biomacromolecules. (2015) 16:3103–11. doi: 10.1021/acs.biomac.5b00458

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Broos S, Lundberg K, Akagi T, Kadowaki K, Akashi M, Greiff L, et al. Immunomodulatory nanoparticles as adjuvants and allergen-delivery system to human dendritic cells: implications for specific immunotherapy. Vaccine. (2010) 28:5075–85. doi: 10.1016/j.vaccine.2010.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Inomata N, Miyakawa M, Aihara M. Surfing as a risk factor for sensitization to poly(gamma-glutamic acid) in fermented soybeans, natto, allergy. Allergol Int. (2018) 67:341–6. doi: 10.1016/j.alit.2017.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Inomata N, Nomura Y, Ikezawa Z. Involvement of poly (gamma-glutamic acid) as an allergen in late-onset anaphylaxis due to fermented soybeans (natto). J Dermatol. (2012) 39:409–12. doi: 10.1111/j.1346-8138.2011.01282.x

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Inomata N, Chin K, Nagashima M, Ikezawa Z. Late-onset anaphylaxis due to poly (gamma-glutamic acid) in the soup of commercial cold Chinese noodles in a patient with allergy to fermented soybeans (natto). Allergol Int. (2011) 60:393–6. doi: 10.2332/allergolint.10-CR-0267

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Pali-Scholl I, Szollosi H, Starkl P, Scheicher B, Stremnitzer C, Hofmeister A, et al. Protamine nanoparticles with CpG-oligodeoxynucleotide prevent an allergen-induced Th2-response in BALB/c mice. Eur J Pharm Biopharm. (2013) 85(3 Pt. A):656–64. doi: 10.1016/j.ejpb.2013.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Nouri HR, Varasteh A, Jaafari MR, Davies JM, Sankian M. Induction of a Th1 immune response and suppression of IgE via immunotherapy with a recombinant hybrid molecule encapsulated in liposome-protamine-DNA nanoparticles in a model of experimental allergy. Immunol Res. (2015) 62:280–91. doi: 10.1007/s12026-015-8659-8

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Scheiblhofer S, Machado Y, Feinle A, Thalhamer J, Husing N, Weiss R. Potential of nanoparticles for allergen-specific immunotherapy - use of silica nanoparticles as vaccination platform. Expert Opin Drug Deliv. (2016) 13:1777–88. doi: 10.1080/17425247.2016.1203898

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Peng X, Liang Y, Yin Y, Liao H, Li L. Development of a hollow mesoporous silica nanoparticles vaccine to protect against house dust mite induced allergic inflammation. Int J Pharm. (2018) 549:115–23. doi: 10.1016/j.ijpharm.2018.07.047

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hirai T, Yoshioka Y, Takahashi H, Ichihashi K, Udaka A, Mori T, et al. Cutaneous exposure to agglomerates of silica nanoparticles and allergen results in IgE-biased immune response and increased sensitivity to anaphylaxis in mice. Part Fibre Toxicol. (2015) 12:16. doi: 10.1186/s12989-015-0095-3

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Fili L, Vultaggio A, Cardilicchia E, Manuelli C, Casini A, Nencini F, et al. A novel allergen-adjuvant conjugate suitable for specific immunotherapy of respiratory allergy. J Allergy Clin Immunol. (2013) 132:84–92. doi: 10.1016/j.jaci.2013.01.030

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Nencini F, Pratesi S, Petroni G, Fili L, Cardilicchia E, Casini A, et al. Treatment with 8-OH-modified adenine (TLR7 ligand)-allergen conjugates decreases T helper type 2-oriented murine airway inflammation. Immunology. (2015) 145:570–82. doi: 10.1111/imm.12475

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Pratesi S, Nencini F, Fili L, Occhiato EG, Romagnani S, Parronchi P, et al. Dermatophagoides pteronyssinus group 2 allergen bound to 8-OH modified adenine reduces the Th2-mediated airway inflammation without inducing a Th17 response and autoimmunity. Mol Immunol. (2016) 77:60–70. doi: 10.1016/j.molimm.2016.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Oomizu S, Onishi N, Suzuki H, Ueda K, Mochizuki M, Morimoto K, et al. Oral administration of pulverized konjac glucomannan prevents the increase of plasma immunoglobulin E and immunoglobulin G levels induced by the injection of syngeneic keratinocyte extracts in BALB/c mice. Clin Exp Allergy. (2006) 36:102–10. doi: 10.1111/j.1365-2222.2005.02405.x

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Onishi N, Kawamoto S, Ueda K, Yamanaka Y, Katayama A, Suzuki H, et al. Dietary pulverized konjac glucomannan prevents the development of allergic rhinitis-like symptoms and IgE response in mice. Biosci Biotechnol Biochem. (2007) 71:2551–6. doi: 10.1271/bbb.70378

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Soria I, Alvarez J, Manzano AI, Lopez-Relano J, Cases B, Mas-Fontao A, et al. Mite allergoids coupled to nonoxidized mannan from Saccharomyces cerevisae efficiently target canine dendritic cells for novel allergy immunotherapy in veterinary medicine. Vet Immunol Immunopathol. (2017) 190:65–72. doi: 10.1016/j.vetimm.2017.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Sirvent S, Soria I, Cirauqui C, Cases B, Manzano AI, Diez-Rivero CM, et al. Novel vaccines targeting dendritic cells by coupling allergoids to nonoxidized mannan enhance allergen uptake and induce functional regulatory T cells through programmed death ligand 1. J Allergy Clin Immunol. (2016) 138:558–67 e11. doi: 10.1016/j.jaci.2016.02.029

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Soria I, Lopez-Relano J, Vinuela M, Tudela JI, Angelina A, Benito-Villalvilla C, et al. Oral myeloid cells uptake allergoids coupled to mannan driving Th1/Treg responses upon sublingual delivery in mice. Allergy. (2018) 73:875–84. doi: 10.1111/all.13396

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Benito-Villalvilla C, Soria I, Perez-Diego M, Fernandez-Caldas E, Subiza JL, Palomares O. Alum impairs tolerogenic properties induced by allergoid-mannan conjugates inhibiting mTOR and metabolic reprogramming in human DCs. Allergy. (2020) 75:648–59. doi: 10.1111/all.14036

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Licciardi M, Montana G, Bondi ML, Bonura A, Scialabba C, Melis M, et al. An allergen-polymeric nanoaggregate as a new tool for allergy vaccination. Int J Pharm. (2014) 465:275–83. doi: 10.1016/j.ijpharm.2014.01.031

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. (2002) 20:709–60. doi: 10.1146/annurev.immunol.20.100301.064842

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Mo JH, Park SW, Rhee CS, Takabayashi K, Lee SS, Quan SH, et al. Suppression of allergic response by CpG motif oligodeoxynucleotide-house-dust mite conjugate in animal model of allergic rhinitis. Am J Rhinol. (2006) 20:212–8. doi: 10.1177/194589240602000219

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Li HT, Chen ZG, Liu H, Ye J, Zou XL, Wang YH, et al. Treatment of allergic rhinitis with CpG oligodeoxynucleotides alleviates the lower airway outcomes of combined allergic rhinitis and asthma syndrome via a mechanism that possibly involves in TSLP. Exp Lung Res. (2016) 42:322–33. doi: 10.1080/01902148.2016.1215571

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Magone MT, Chan CC, Beck L, Whitcup SM, Raz E. Systemic or mucosal administration of immunostimulatory DNA inhibits early and late phases of murine allergic conjunctivitis. Eur J Immunol. (2000) 30:1841–50. doi: 10.1002/1521-4141(200007)30:7<1841::AID-IMMU1841>3.0.CO;2-E

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Xu W, Tamura T, Takatsu K. CpG ODN mediated prevention from ovalbumin-induced anaphylaxis in mouse through B cell pathway. Int Immunopharmacol. (2008) 8:351–61. doi: 10.1016/j.intimp.2007.10.019

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Simons FE, Shikishima Y, Van Nest G, Eiden JJ, HayGlass KT. Selective immune redirection in humans with ragweed allergy by injecting Amb a 1 linked to immunostimulatory DNA. J Allergy Clin Immunol. (2004) 113:1144–51. doi: 10.1016/j.jaci.2004.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Roth-Walter F, Pacios LF, Gomez-Casado C, Hofstetter G, Roth GA, Singer J, et al. The major cow milk allergen Bos d 5 manipulates T-helper cells depending on its load with siderophore-bound iron. PLoS ONE. (2014) 9:e104803. doi: 10.1371/journal.pone.0104803

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Roth-Walter F, Gomez-Casado C, Pacios LF, Mothes-Luksch N, Roth GA, Singer J, et al. Bet v 1 from birch pollen is a lipocalin-like protein acting as allergen only when devoid of iron by promoting Th2 lymphocytes. J Biol Chem. (2014) 289:17416–21. doi: 10.1074/jbc.M114.567875

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Hufnagl K, Afify SM, Braun N, Wagner S, Wallner M, Hauser M, et al. Retinoic acid-loading of the major birch pollen allergen Bet v 1 may improve specific allergen immunotherapy: in silico, in vitro and in vivo data in BALB/c mice. Allergy. (2020). doi: 10.1111/all.14259

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Hufnagl K, Ghosh D, Wagner S, Fiocchi A, Dahdah L, Bianchini R, et al. Retinoic acid prevents immunogenicity of milk lipocalin Bos d 5 through binding to its immunodominant T-cell epitope. Sci Rep. (2018) 8:1598. doi: 10.1038/s41598-018-19883-0

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Petrarca C, Clemente E, Toto V, Iezzi M, Rossi C, Zanotta S, et al. rBet v 1 immunotherapy of sensitized mice with Streptococcus thermophilus as vehicle and adjuvant. Hum Vaccin Immunother. (2014) 10:1228–37. doi: 10.4161/hv.28155

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Roozbeh Nasiraie L, Tabatabaie F, Sankian M, Shahidi F, Varasteh A. Construction of a recombinant allergen-producing probiotic bacterial strain: introduction of a new line for a live oral vaccine against Chenopodium album pollen allergy. Rep Biochem Mol Biol. (2013) 2:16–27.

PubMed Abstract | Google Scholar

82. Ohkouchi K, Kawamoto S, Tatsugawa K, Yoshikawa N, Takaoka Y, Miyauchi S, et al. Prophylactic effect of Lactobacillus oral vaccine expressing a Japanese cedar pollen allergen. J Biosci Bioeng. (2012) 113:536–41. doi: 10.1016/j.jbiosc.2011.11.025

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Gerstmayr M, Ilk N, Schabussova I, Jahn-Schmid B, Egelseer EM, Sleytr UB, et al. A novel approach to specific allergy treatment: the recombinant allergen-S-layer fusion protein rSbsC-Bet v 1 matures dendritic cells that prime Th0/Th1 and IL-10-producing regulatory T cells. J Immunol. (2007) 179:7270–5. doi: 10.4049/jimmunol.179.11.7270

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Anzengruber J, Bublin M, Bonisch E, Janesch B, Tscheppe A, Braun ML, et al. Lactobacillus buchneri S-layer as carrier for an Ara h 2-derived peptide for peanut allergen-specific immunotherapy. Mol Immunol. (2017) 85:81–8. doi: 10.1016/j.molimm.2017.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Malamud M, Carasi P, Assandri MH, Freire T, Lepenies B, Serradell MLA. S-Layer Glycoprotein from Lactobacillus kefiri exerts its immunostimulatory activity through glycan recognition by mincle. Front Immunol. (2019) 10:1422. doi: 10.3389/fimmu.2019.01422

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Ou J, Shi W, Xu Y, Tao Z. Intranasal immunization with DNA vaccine coexpressing Der p 1 and ubiquitin in an allergic rhinitis mouse model. Ann Allergy Asthma Immunol. (2014) 113:658–65 e1. doi: 10.1016/j.anai.2014.08.015

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Hu W, Ma L, Yang G, Zeng X, Liu J, Cheng B, et al. Der p2A20 DNA vaccine attenuates allergic inflammation in mice with allergic rhinitis. Mol Med Rep. (2019) 20:4925–32. doi: 10.3892/mmr.2019.10760

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Bublin M, Hoflehner E, Wagner B, Radauer C, Wagner S, Hufnagl K, et al. Use of a genetic cholera toxin B subunit/allergen fusion molecule as mucosal delivery system with immunosuppressive activity against Th2 immune responses. Vaccine. (2007) 25:8395–404. doi: 10.1016/j.vaccine.2007.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Basomba A, Tabar AI, de Rojas DH, Garcia BE, Alamar R, Olaguibel JM, et al. 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) 109:943–8. doi: 10.1067/mai.2002.124465

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Ishii M, Koyama A, Iseki H, Narumi H, Yokoyama N, Kojima N. Anti-allergic potential of oligomannose-coated liposome-entrapped Cry j 1 as immunotherapy for Japanese cedar pollinosis in mice. Int Immunopharmacol. (2010) 10:1041–6. doi: 10.1016/j.intimp.2010.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Chaisri U, Tungtrongchitr A, Indrawattana N, Meechan P, Phurttikul W, Tasaniyananda N, et al. Immunotherapeutic efficacy of liposome-encapsulated refined allergen vaccines against Dermatophagoides pteronyssinus allergy. PLoS ONE. (2017) 12:e0188627. doi: 10.1371/journal.pone.0188627

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Tasaniyananda N, Chaisri U, Tungtrongchitr A, Chaicumpa W, Sookrung N. Mouse model of cat allergic rhinitis and intranasal liposome-adjuvanted refined Fel d 1 vaccine. PLoS ONE. (2016) 11:e0150463. doi: 10.1371/journal.pone.0150463

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Aliu H, Rask C, Brimnes J, Andresen TL. Enhanced efficacy of sublingual immunotherapy by liposome-mediated delivery of allergen. Int J Nanomedicine. (2017) 12:8377–88. doi: 10.2147/IJN.S137033

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Meechan P, Tungtrongchitr A, Chaisri U, Maklon K, Indrawattana N, Chaicumpa W, et al. Intranasal, liposome-adjuvanted cockroach allergy vaccines made of refined major allergen and whole-body extract of Periplaneta americana. Int Arch Allergy Immunol. (2013) 161:351–62. doi: 10.1159/000348314

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bondi ML, Montana G, Craparo EF, Di Gesu R, Giammona G, Bonura A, et al. Lipid nanoparticles as delivery vehicles for the Parietaria judaica major allergen Par j 2. Int J Nanomedicine. (2011) 6:2953–62. doi: 10.2147/IJN.S24264

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Mueller RS, Veir J, Fieseler KV, Dow SW. Use of immunostimulatory liposome-nucleic acid complexes in allergen-specific immunotherapy of dogs with refractory atopic dermatitis - a pilot study. Vet Dermatol. (2005) 16:61–8. doi: 10.1111/j.1365-3164.2005.00426.x

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Veir JK, Lappin MR, Dow SW. Evaluation of a novel immunotherapy for treatment of chronic rhinitis in cats. J Feline Med Surg. (2006) 8:400–11. doi: 10.1016/j.jfms.2006.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Scholl I, Kopp T, Bohle B, Jensen-Jarolim E. Biodegradable PLGA particles for improved systemic and mucosal treatment of Type I allergy. Immunol Allergy Clin North Am. (2006) 26:349–64 xi. doi: 10.1016/j.iac.2006.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Scholl I, Weissenbock A, Forster-Waldl E, Untersmayr E, Walter F, Willheim M, et al. Allergen-loaded biodegradable poly(D,L-lactic-co-glycolic) acid nanoparticles down-regulate an ongoing Th2 response in the BALB/c mouse model. Clin Exp Allergy. (2004) 34:315–21. doi: 10.1111/j.1365-2222.2004.01884.x

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Roth-Walter F, Bohle B, Scholl I, Untersmayr E, Scheiner O, Boltz-Nitulescu G, et al. Targeting antigens to murine and human M-cells with Aleuria aurantia lectin-functionalized microparticles. Immunol Lett. (2005) 100:182–8. doi: 10.1016/j.imlet.2005.03.020

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Roth-Walter F, Scholl I, Untersmayr E, Ellinger A, Boltz-Nitulescu G, Scheiner O, et al. Mucosal targeting of allergen-loaded microspheres by Aleuria aurantia lectin. Vaccine. (2005) 23:2703–10. doi: 10.1016/j.vaccine.2004.11.052

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Roth-Walter F, Scholl I, Untersmayr E, Fuchs R, Boltz-Nitulescu G, Weissenbock A, et al. M cell targeting with Aleuria aurantia lectin as a novel approach for oral allergen immunotherapy. J Allergy Clin Immunol. (2004) 114:1362–8. doi: 10.1016/j.jaci.2004.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Diesner SC, Wang XY, Jensen-Jarolim E, Untersmayr E, Gabor F. Use of lectin-functionalized particles for oral immunotherapy. Ther Deliv. (2012) 3:277–90. doi: 10.4155/tde.11.146

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Hajavi J, Hashemi M, Sankian M. Evaluation of size and dose effects of rChe a 3 allergen loaded PLGA nanoparticles on modulation of Th2 immune responses by sublingual immunotherapy in mouse model of rhinitis allergic. Int J Pharm. (2019) 563:282–92. doi: 10.1016/j.ijpharm.2019.03.040

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Salari F, Varasteh AR, Vahedi F, Hashemi M, Sankian M. Down-regulation of Th2 immune responses by sublingual administration of poly (lactic-co-glycolic) acid (PLGA)-encapsulated allergen in BALB/c mice. Int Immunopharmacol. (2015) 29:672–8. doi: 10.1016/j.intimp.2015.09.011

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Marazuela EG, Prado N, Moro E, Fernandez-Garcia H, Villalba M, Rodriguez R, et al. Intranasal vaccination with poly(lactide-co-glycolide) microparticles containing a peptide T of Ole e 1 prevents mice against sensitization. Clin Exp Allergy. (2008) 38:520–8. doi: 10.1111/j.1365-2222.2007.02922.x

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Batanero E, Barral P, Villalba M, Rodriguez R. Encapsulation of Ole e 1 in biodegradable microparticles induces Th1 response in mice: a potential vaccine for allergy. J Control Release. (2003) 92:395–8. doi: 10.1016/S0168-3659(03)00337-7

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Martinez Gomez JM, Fischer S, Csaba N, Kundig TM, Merkle HP, Gander B, et al. A protective allergy vaccine based on CpG- and protamine-containing PLGA microparticles. Pharm Res. (2007) 24:1927–35. doi: 10.1007/s11095-007-9318-0

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Xiao X, Zeng X, Zhang X, Ma L, Liu X, Yu H, et al. Effects of Caryota mitis profilin-loaded PLGA nanoparticles in a murine model of allergic asthma. Int J Nanomedicine. (2013) 8:4553–62. doi: 10.2147/IJN.S51633

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Joshi VB, Adamcakova-Dodd A, Jing X, Wongrakpanich A, Gibson-Corley KN, Thorne PS, et al. Development of a poly (lactic-co-glycolic acid) particle vaccine to protect against house dust mite induced allergy. AAPS J. (2014) 16:975–85. doi: 10.1208/s12248-014-9624-5

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Srivastava KD, Siefert A, Fahmy TM, Caplan MJ, Li XM, Sampson HA. Investigation of peanut oral immunotherapy with CpG/peanut nanoparticles in a murine model of peanut allergy. J Allergy Clin Immunol. (2016) 138:536–43 e4. doi: 10.1016/j.jaci.2016.01.047

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Kostadinova AI, Middelburg J, Ciulla M, Garssen J, Hennink WE, Knippels LMJ, et al. PLGA nanoparticles loaded with beta-lactoglobulin-derived peptides modulate mucosal immunity and may facilitate cow's milk allergy prevention. Eur J Pharmacol. (2018) 818:211–20. doi: 10.1016/j.ejphar.2017.10.051

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Balmert SC, Donahue C, Vu JR, Erdos G, Falo LD Jr, Little SR. In vivo induction of regulatory T cells promotes allergen tolerance and suppresses allergic contact dermatitis. J Control Release. (2017) 261:223–33. doi: 10.1016/j.jconrel.2017.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Jilek S, Walter E, Merkle HP, Corthesy B. Modulation of allergic responses in mice by using biodegradable poly(lactide-co-glycolide) microspheres. J Allergy Clin Immunol. (2004) 114:943–50. doi: 10.1016/j.jaci.2004.05.065

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Anzaghe M, Schulke S, Scheurer S. Virus-like particles as carrier systems to enhance immunomodulation in allergen immunotherapy. Curr Allergy Asthma Rep. (2018) 18:71. doi: 10.1007/s11882-018-0827-1

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Klimek L, Willers J, Hammann-Haenni A, Pfaar O, Stocker H, Mueller P, et al. Assessment of clinical efficacy of CYT003-QbG10 in patients with allergic rhinoconjunctivitis: a phase IIb study. Clin Exp Allergy. (2011) 41:1305–12. doi: 10.1111/j.1365-2222.2011.03783.x

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Senti G, Johansen P, Haug S, Bull C, Gottschaller C, Muller P, et al. Use of A-type CpG oligodeoxynucleotides as an adjuvant in allergen-specific immunotherapy in humans: a phase I/IIa clinical trial. Clin Exp Allergy. (2009) 39:562–70. doi: 10.1111/j.1365-2222.2008.03191.x

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Fettelschoss-Gabriel A, Fettelschoss V, Thoms F, Giese C, Daniel M, Olomski F, et al. Treating insect-bite hypersensitivity in horses with active vaccination against IL-5. J Allergy Clin Immunol. (2018) 142:1194–205 e3. doi: 10.1016/j.jaci.2018.01.041

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Jonsdottir S, Fettelschoss V, Olomski F, Talker SC, Mirkovitch J, Rhiner T, et al. Safety profile of a virus-like particle-based vaccine targeting self-protein interleukin-5 in horses. Vaccines. (2020) 8:E213. doi: 10.3390/vaccines8020213

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Bachmann MF, El-Turabi A, Fettelschoss-Gabriel A, Vogel M. The prospects of an active vaccine against asthma targeting IL-5. Front Microbiol. (2018) 9:2522. doi: 10.3389/fmicb.2018.02522

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Fettelschoss-Gabriel A, Fettelschoss V, Olomski F, Birkmann K, Thoms F, Buhler M, et al. Active vaccination against interleukin-5 as long-term treatment for insect-bite hypersensitivity in horses. Allergy. (2019) 74:572–82. doi: 10.1111/all.13659

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Olomski F, Fettelschoss V, Jonsdottir S, Birkmann K, Thoms F, Marti E, et al. Interleukin 31 in insect bite hypersensitivity-alleviating clinical symptoms by active vaccination against itch. Allergy. (2020) 75:862–71. doi: 10.1111/all.14145

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Bachmann MF, Zeltins A, Kalnins G, Balke I, Fischer N, Rostaher A, et al. Vaccination against IL-31 for the treatment of atopic dermatitis in dogs. J Allergy Clin Immunol. (2018) 142:279–81 e1. doi: 10.1016/j.jaci.2017.12.994

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Thoms F, Haas S, Erhart A, Nett CS, Rufenacht S, Graf N, et al. Immunization of cats against Fel d 1 results in reduced allergic symptoms of owners. Viruses. (2020) 12:288. doi: 10.3390/v12030288

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Thoms F, Jennings GT, Maudrich M, Vogel M, Haas S, Zeltins A, 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. doi: 10.1016/j.jaci.2019.01.050

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Manzano-Szalai K, Thell K, Willensdorfer A, Weghofer M, Pfanzagl B, Singer J, et al. Adeno-associated virus-like particles as new carriers for B-cell vaccines: testing immunogenicity and safety in BALB/c mice. Viral Immunol. (2014) 27:438–48. doi: 10.1089/vim.2014.0059

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Kratzer B, Kohler C, Hofer S, Smole U, Trapin D, Iturri J, et al. Prevention of allergy by virus-like nanoparticles (VNP) delivering shielded versions of major allergens in a humanized murine allergy model. Allergy. (2019) 74:246–60. doi: 10.1111/all.13573

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Storni F, Zeltins A, Balke I, Heath MD, Kramer MF, Skinner MA, et al. Vaccine against peanut allergy based on engineered virus-like particles displaying single major peanut allergens. J Allergy Clin Immunol. (2020) 145:1240–53 e3. doi: 10.1016/j.jaci.2019.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Engeroff P, Caviezel F, Storni F, Thoms F, Vogel M, Bachmann MF. Allergens displayed on virus-like particles are highly immunogenic but fail to activate human mast cells. Allergy. (2018) 73:341–9. doi: 10.1111/all.13268

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Kundig TM, Senti G, Schnetzler G, Wolf C, Prinz Vavricka BM, Fulurija A, et al. Der p 1 peptide on virus-like particles is safe and highly immunogenic in healthy adults. J Allergy Clin Immunol. (2006) 117:1470–6. doi: 10.1016/j.jaci.2006.01.040

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Walter F, Scholl I, Untersmayr E, Ellinger A, Boltz-Nitulescu G, Scheiner O, et al. Functionalisation of allergen-loaded microspheres with wheat germ agglutinin for targeting enterocytes. Biochem Biophys Res Commun. (2004) 315:281–7. doi: 10.1016/j.bbrc.2004.01.057

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immunotherapy, adjuvant, allergy, allergen, veterinary, human

Citation: Pali-Schöll I, DeBoer DJ, Alessandri C, Seida AA, Mueller RS and Jensen-Jarolim E (2020) Formulations for Allergen Immunotherapy in Human and Veterinary Patients: New Candidates on the Horizon. Front. Immunol. 11:1697. doi: 10.3389/fimmu.2020.01697

Received: 21 May 2020; Accepted: 25 June 2020;
Published: 04 August 2020.

Edited by:

Simon Blank, Helmholtz Zentrum München, Germany

Reviewed by:

Ulrich Matthias Zissler, Technical University of Munich, Germany
Dennis Russkamp, Medigene Immunotherapies GmbH, Germany

Copyright © 2020 Pali-Schöll, DeBoer, Alessandri, Seida, Mueller and Jensen-Jarolim. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Isabella Pali-Schöll, isabella.pali@vetmeduni.ac.at; isabella.pali@meduniwien.ac.at

ORCID: Isabella Pali-Schöll orcid.org/0000-0003-2089-6011
Douglas J. DeBoer orcid.org/0000-0001-7102-7989
Claudia Alessandri orcid.org/0000-0003-2466-6870
Ralf S. Mueller orcid.org/0000-0001-5835-5910
Erika Jensen-Jarolim orcid.org/0000-0003-4019-5765

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.