1,937
Views
50
CrossRef citations to date
0
Altmetric
Special Focus Review

Adjuvants for allergy vaccines

Pages 1492-1498 | Received 08 Jun 2012, Accepted 01 Aug 2012, Published online: 01 Oct 2012

Abstract

Allergen-specific immunotherapy is currently performed via either the subcutaneous or sublingual routes as a treatment for type I (IgE dependent) allergies. Aluminum hydroxide or calcium phosphate are broadly used as adjuvants for subcutaneous allergy vaccines, whereas commercial sublingual vaccines rely upon high doses of aqueous allergen extracts in the absence of any immunopotentiator. Adjuvants to be included in the future in products for allergen specific immunotherapy should ideally enhance Th1 and CD4+ regulatory T cell responses. Imunomodulators impacting dendritic or T cell functions to induce IL10, IL12 and IFNγ production are being investigated in preclinical allergy models. Such candidate adjuvants encompass synthetic or biological immunopotentiators such as glucocorticoids, 1,25-dihydroxy vitamin D3, selected probiotic strains (e.g., Lactobacillus and Bifidobacterium species) as well as TLR2 (Pam3CSK4), TLR4 (monophosphoryl lipid A, synthetic lipid A analogs) or TLR9 (CpGs) ligands. Furthermore, the use of vector systems such as mucoadhesive particules, virus-like particles or liposomes are being considered to enhance allergen uptake by tolerogenic antigen presenting cells present in mucosal tissues.

Introduction

Current trends in allergen specific immunotherapy

Following the pioneer studies by Noon, Freeman et al.Citation1,Citation2 more than a century ago, allergen-specific immunotherapy (AIT) has been progressively established as a reference treatment for type I allergies, most particularly allergic rhinoconjunctivitis with or without moderate asthma induced by common respiratory allergens.Citation3 In contrast to symptomatic treatments, AIT can be curative, due to its capacity to reorient inappropriate allergen-specific humoral and cellular immune responses from a Th2 to a mixed Th1/T reg pattern.Citation4 Allergen-specific immunotherapy is commonly performed via the subcutaneous route as a treatment for allergies to grass and tree pollens, house dust mites, insect venoms, molds, dog and cat dander or even synthetic drugs.Citation3, Citation5 Subcutaneous immunotherapy (SCIT) is performed with soluble allergens in North America, whereas allergen extracts are rather adsorbed on aluminum hydroxide or calcium phosphate as adjuvants in Europe.Citation3,Citation5,Citation6 As will be reviewed below, numerous other candidate adjuvants are being evaluated in preclinical models or in humans, with the aim to reduce the number of injections needed for subcutaneous desensitization.

In the past 20 years, other routes of allergen administration (e.g., intranasal, oral, sublingual, intralymphatic, epicutaneous) have been and are being explored as an alternative to SCIT.Citation8 Most particularly, among non invasive routes, sublingual immunotherapy (SLIT) is now considered as a safe and efficacious alternative to SCIT for respiratory allergies associated with either grass, tree pollens or house dust mites.Citation9Citation11 Commercial sublingual vaccines are based on aqueous allergen extracts presented either as drops or more recently as fast disolving tablets or lyocs. During SLIT, high doses (usually 50 to 100 fold the doses used for SCIT) of the allergen extract are kept under the tongue for 1 to 2 min prior to being swallowed, as per the so called sublingual-swallow procedure. As of today, none of the commercially available sublingual vaccines contain any adjuvant.Citation9

Irrespective of the route of immunization used, current allergy vaccines rely upon either natural or modified allergen extracts. The latter can for example be obtained following treatment with glutaraldehyde to form polymers with altered structure, thus precluding IgE recognition, as is the case for allergoids.Citation3 New allergen presentation platforms are needed to improve the efficacy of such existing subcutaneous and sublingual allergy vaccines.Citation12, Citation13

In addition, in the last few years, second generation treatments based on recombinant allergens (in a natural or hypoallergenic conformation), fusions proteins, mix of peptides, plasmid DNA have raised considerable interest, even if none of those molecular allergens have reached commercialisation.Citation14Citation16 However, in contrast to natural extracts which often possess an intrinsic adjuvant activity (e.g., Th2-inducing activity ascribed to the Der p 1 mite allergen, endotoxin, phytoprostanes),Citation17,Citation18 purified allergens provided under such a well defined molecular form are usually poorly immunogenic. Thus, such second generation vaccines will also require novel immunopotentiators and vector systems (collectively referred to as adjuvants in the present review).

Like for any vaccines, adjuvants to be associated with allergens are expected to allow simplifying immunization regimens, and reaching efficacy faster and for a longer duration. Although allergy vaccines are usually well tolerated, an additional expected benefit of adjuvants in this field is to help lowering the allergen dose, thus improving the safety profile with less local reactions to the site of administration. Importantly, allergy vaccines are therapeutic vaccines, used in patients prone to mounting Th2 responses. Thus, appropriate adjuvants should rather orient the immune response toward Th1 or regulatory mechanisms both known to downregulate Th2 cells.Citation12

Immune mechanisms associated with allergen-specific immunotherapy

Our current understanding of immune mechanisms involved in allergen-specific immunotherapy emphasizes a prominent role for CD4+ T lymphocytes in controlling all effector immune mechanisms linked with allergic inflammation.Citation4, Citation19, Citation20 Specifically, most allergic patients exhibit allergen-specific Th2 responses associated with the secretion of IL-4, IL-5 and IL-13 cytokines by CD4+ T cells. During subcutaneous or sublingual immunotherapy, such allergen-specific CD4+ T cell responses are rather redirected toward both a Th1 type with an increased production of IFNγ (immunodeviation) as well as IL10-producing CD4+ regulatory T cells (immunosuppression).Citation4, Citation19, Citation20 Such changes in the polarization of T cell responses have been documented both in peripheral blood and in respiratory mucosaeCitation21, Citation22

As a consequence of such a change in the cytokine milieu, both SCIT and SLIT are associated with a decrease in seric allergen-specific IgEs, concomitantly with an upregulation of IgG1, IgG4, IgA antibodies,Citation4, Citation20, Citation23 some of which are thought to mediate a “blocking” anti-inflammatory activity.Citation24 Successful immunotherapy also leads to a substantial decrease in the recruitment and activation of proinflammatory cells, including basophils, mast cells and eosinophils in the skin, as well as nasal or bronchial mucosae.Citation25

Based upon the above afore mentioned immune mechanisms thought to be critical for tolerance induction during immunotherapy, adjuvants expected to enhance the efficacy of allergy vaccines include:

(1) Synthetic or biological immunopotentiators capable to reinforce allergen-specific Th1 and/or regulatory T cell responses (). These molecules could act directly on CD4+ T cells, but also on dendritic or even epithelial cells.

Table 1. Immunopotentiators for allergy vaccines

(2) Vector systems facilitating allergen uptake by tolerogenic APCs such as the dendritic cells found in oral tissuesCitation26,Citation27 ().

Table 2. Vector systems for allergy vaccines

Adjuvants for Allergy Vaccines Administered via the Subcutaneous (and Other Parenteral) Route(s)

Immunopotentiators

Mineral adjuvant molecules such as calcium phosphate or aluminum hydroxide are broadly used in humans as adjuvants for subcutaneous allergic vaccinesCitation7, Citation12 (). Aluminum salts are commonly included in vaccines against infectious pathogens with the aim to elicit proinflammatory responses following activation of the inflammasome. In addition however, those compounds also decrease established Th2 responses.Citation7 Beyond such mineral adjuvants, various immunopotentiators were shown in murine models to downregulate Th2 responses when administered via parenteral routes (). For example, TLR ligands (e.g., monophosphoryl lipid A or MPL, imidazoquinolines, CpGs), living or heat-killed bacteria (e.g., Lactobacillus plantarum, Lactococcus lactis, Mycobacterium vaccae), small molecules such as the active 1,25-dihydroxy Vitamin D3 metabolite all contribute in decreasing airway inflammation in murine models of asthma, when used as adjuvants.Citation28Citation33

Several of those immunopotentiators impacting Th1 or Treg responses have been investigated in humans during SCIT. For example, the Th1 adjuvant MPL injected together with a grass pollen extract enhanced allergen-specific IgG1 and IgG4 responses, raising the possibility to reduce the number of preseasonal injections to control allergic symptoms.Citation34, Citation35 Also, a synthetic CpG oligonucleotide acting as a Th1 adjuvant was chemically conjugated with the purified Amb a 1 allergen from short ragweed. This vaccine was shown to improve rhinoconjunctivitis symptoms in patients with allergy to ragweed pollen.Citation36 The parallel use of oral steroids (prednisone) with or without Vitamin D3 (both of which were reported as potential inducers of regulatory T cells) during SCIT did not improve efficacy in asthmatic children allergic to house dust mites.Citation37

Vector systems

Various vector systems have also been used successfully to formulate allergens administered via parenteral routes in animal models (). The latter include liposomes, virosomes, D, L-lactic-co-glycolic acid (PLGA) or immunostimulating complexes (ISCOMS).Citation38, Citation39 Also, plasmid DNA or DNA absorbed on microparticules were shown to enhance systemic and mucosal immune responses.Citation40 Fusion proteins associating the Fel d 1 cat allergen and the Fcγ immunoglobulin fragment were also designed to co-crosslink FcεRI and FcγRII receptors, thus downregulating immunoreactivity to Fel d 1.Citation41 Another fusion protein combining the birch pollen Bet v 1 allergen to the bacterial S layer protein SbpA was shown to form particules facilitating allergen uptake by dendritic cells, with subsequent induction of Th1 and regulatory T cells.Citation42 Fusing the allergen with the TLR5 ligand flagellin was also shown to reduce Th2 responses.Citation43

As of today, only a limited number of vector systems have been evaluated in humans via parental routes. For example, non-replicating virus-like particles (VLPs) obtained following assembly of capsid proteins from the Qβ phage were coupled to peptides derived from the Der p 1 mite allergen. Those recombinant Der p 1 VLPs induce strong IgG responses after a single injection in healthy volonteers.Citation44 Conjugation of Fel d 1 to such VLPs was shown to prevent IgE reactivity while enhancing IgG induction.Citation45 QβG10 VLPs containing a synthetic oligonucleotide as a Th1 adjuvant were also tested in mite allergic patients, leading in a significant reduction of rhinoconjunctivitis and asthma symptoms after six weekly injections.Citation46 More recently, a recombinant fusion protein associating a peptide derived from the Phl p 1 grass pollen allergen with a rhinovirus-derived VP1 protein was also proposed as a candidate vaccine for grass pollen allergies.Citation47 Another fusion protein associating the Fel d 1 allergen to a modular antigen transporter (MAT) was designed to facilitate capture, processing and efficient presentation of the allergen by APCs in association with MHC class II molecules.Citation48 When administered in humans, this vaccine elicited strong IgG4 responses to the allergen.

Adjuvants for Allergy Vaccines Administered via The Sublingual (and Other Mucosal) Route(s)

Immunopotentiators

Various mucosal immunopotentiators have been tested in preclinical tolerance models. The latter include bacterial toxins, such as cholera toxin (CT) or E. coli heat-labile enterotoxin (LT), (as well as genetically detoxified forms or B-subunits without any ADP-ribosyl transferase activity)Citation49, Citation50 (). These toxins were either co-administered, conjugated or fused with allergens.Citation49, Citation50 TLR2 (e.g., Pam3CSK4) or TLR4 ligands (e.g., MPL and OM-294-BA-MP) were shown to strengthen Th1 and T Reg responses when used as adjuvants via the nasal or sublingual routes.Citation51Citation54 Similarly, dexamethasone associated with 1,25-dihydroxy vitamin D3 enhanced the efficacy of SLIT in a murine asthma model following induction of interleukin 10 production by immune cells, including dendritic cells and CD4+ T lymphocytes.Citation55 Another emerging class of potential adjuvants for the sublingual (and other mucosal) routes include probiotics, even if immunomodulatory properties vary considerably depending upon bacterial strains and growth conditions.Citation56, Citation57 Bacteria selected specifically for their capacity to induce a strong production of IL10 and IL12 by mucosal DCs, such as Lactobacillus plantarum or Bifidobacterium bifidum, were shown to enhance tolerance following SLIT in mice with induced asthma to OVA, at least in part by strengthening allergen-specific Th1 and T Reg responses.Citation58, Citation59 Interestingly, in preclinical models of SLIT, none of the pure Th1 adjuvants (ie without any capacity to elicit IL10 production) had any significant impact on tolerance induction.Citation12

Two categories of immunopotentiators have been tested with the aim to increase SLIT efficacy in allergic human patients. High doses of the Th1 adjuvant MPL boosted allergen-specific IgG responses and reduced reactivity to a subsequent nasal allergen challenge in grass pollen allergic patients ().Citation60 BCG administered intradermally to children asthmatic to mite allergens in parallel with SLIT had no impact on clinical outcome,Citation61 possibly because the adjuvant and allergens were administered via distinct routes. In addition, various lactic acid bacterial strains are being considered to prevent or treat allergy in humans.Citation57 For example, oral administration of lactic acid bacteria reduces atopic dermatitis in children with a positive family history of type I allergy.Citation62 Whereas such an observation suggests that selected probiotics could be used as adjuvants for mucosal vaccines, as of today probiotics have not yet been tested in combination with allergens in allergic patients.

Vector systems

The dual interest of vector systems for mucosal vaccination is (1) to enhance the duration of contact of the allergen with the mucosa, for example by using positively charged mucoadhesive polymers binding to epithelial cells, and further (2) to target efficiently phagocytic APCs by presenting the allergen in a particulate form or by interacting with a specific surface receptor expressed by dendritic cellsCitation12, Citation63 (). In this regard, both nanoparticles made from polymerized maltodextrin or chitosan-based microparticles were shown to enhance the uptake of allergens by oral dendritic cells, following sublingual administration to asthmatic mice, thus resulting into a superior priming of allergen-specific Tr1 cells in draining cervical lymph nodes, and consequently in a stronger decrease in airway inflammationCitation23, Citation64 (). Still in murine models, conjugates with the outer membrane protein A from Klebsiella pneumoniae (OmpA) as well as the shiga toxin B subunit have been successfully used to target efficiently antigens/allergens to APCsCitation65Citation67 (). Similarly, the adenylate cyclase protein from Bordetella pertussis fused to OVA enhanced tolerance induction via the sublingual route in mice with OVA-induced asthma, following capture by CD11b+ tolerogenic myeloid DCs and macrophages.Citation68, Citation69

As of today, none of those vector systems have been tested in humans in the context of mucosal allergy vaccines, although they raised the interest of the vaccine industry.Citation70Citation72 Noteworthy, maltodextrin-based mucoadhesive nanoparticles have been used to formulate an experimental flu vaccine, with evidence for the induction of strong mucosal IgA responses when administered intranasally to healthy volonteers.Citation12

Conclusion

New adjuvants and vector systems are needed to improve the efficacy of allergy vaccines, reduce the dose of allergens required and simplify immunization schemes during desensitization. Our current understanding of immune mechanisms supporting the induction and maintenance of antigen-specific immune tolerance suggests an interest of adjuvants eliciting combined Th1 and regulatory CD4+ T cell responses. A variety of candidate adjuvants (ie TLR ligands, bacterial toxins, probiotics) are efficacious in reducing allergic inflammation in murine models, when used in combination with the allergen via either parenteral or mucosal routes. Besides the commonly used mineral adjuvants such as Alum and calcium phosphate, only a limited number of Th1 immunopotentiators (e.g., MPL, CpGs, lactic acid bacteria) have been tested in humans, but their impact on clinical efficacy remains to be fully investigated. Among available vector systems, virus like-particles represent a highly promising platform to present allergens and/or adjuvants to the immune-system. In this regard encouraging clinical efficacy results have been obtained during SCIT of mite allergic patients. Mucoadhesive particulate vectors have not yet been tested in humans, but appear very promising based on preclinical experiments to support the development of safer and more efficient mucosal allergy vaccines. Additional studies evaluating the long-term efficacy of such vectorized allergens in large cohorts of allergic patients are needed to understand the true potential of those technologies in the field of allergen-specific immunotherapy.

References

  • Noon L. Prophylactic inoculation against hay fever. Lancet 1911; 177:1572 - 3; http://dx.doi.org/10.1016/S0140-6736(00)78276-6
  • Freeman J. Further observations on the treatment of hay fever by hypodermic inoculations of pollen vaccine. Lancet 1911; 178:814 - 7; http://dx.doi.org/10.1016/S0140-6736(01)40417-X
  • Bousquet J, Lockey R, Malling HJ. Allergen immunotherapy: therapeutic vaccines for allergic diseases. A WHO position paper. J Allergy Clin Immunol 1998; 102:558 - 62; http://dx.doi.org/10.1016/S0091-6749(98)70271-4; PMID: 9802362
  • Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol 2011; 127:18 - 27, quiz 28-9; http://dx.doi.org/10.1016/j.jaci.2010.11.030; PMID: 21211639
  • Joint Task Force on Practice Parameters, American Academy of Allergy, Asthma and Immunology, American College of Allergy, Asthma and Immunology, Joint Council of Allergy, Asthma and Immunology. Allergen immunotherapy: a practice parameter second update. J Allergy Clin Immunol 2007; 120:Suppl S25 - 85; http://dx.doi.org/10.1016/j.jaci.2007.06.019; PMID: 17765078
  • Bousquet J, Van Cauwenberge P, Khaltaev N, Aria Workshop Group, World Health Organization. Allergic rhinitis and its impact on asthma. J Allergy Clin Immunol 2001; 108:Suppl S147 - 334; http://dx.doi.org/10.1067/mai.2001.118891; PMID: 11707753
  • Wilcock LK, Francis JN, Durham SR. Aluminium hydroxide down-regulates T helper 2 responses by allergen-stimulated human peripheral blood mononuclear cells. Clin Exp Allergy 2004; 34:1373 - 8; http://dx.doi.org/10.1111/j.1365-2222.2004.02052.x; PMID: 15347369
  • Mascarell L, Van Overtvelt L, Moingeon P. Novel ways for immune intervention in immunotherapy: mucosal allergy vaccines. Immunol Allergy Clin North Am 2006; 26:283 - 306, vii-viii; http://dx.doi.org/10.1016/j.iac.2006.02.009; PMID: 16701145
  • Canonica GW, Bousquet J, Casale T, Lockey RF, Baena-Cagnani CE, Pawankar R, et al. Sub-lingual immunotherapy: World Allergy Organization Position Paper 2009. Allergy 2009; 64:Suppl 91 1 - 59; http://dx.doi.org/10.1111/j.1398-9995.2009.02309.x; PMID: 20041860
  • Wilson DR, Torres LI, Durham SR. Sublingual immunotherapy for allergic rhinitis. Cochrane Database Syst Rev 2003; CD002893; PMID: 12804442
  • Passalacqua G, Lombardi C, Troise C, Canonica GW. Sublingual immunotherapy: certainties, unmet needs and future directions. Eur Ann Allergy Clin Immunol 2009; 41:163 - 70; PMID: 20128229
  • Moingeon P, Lombardi V, Saint-Lu N, Tourdot S, Bodo V, Mascarell L. Adjuvants and vector systems for allergy vaccines. Immunol Allergy Clin North Am 2011; 31:407 - 19, xii; http://dx.doi.org/10.1016/j.iac.2011.03.001; PMID: 21530828
  • Wheeler AW, Woroniecki SR. Immunological adjuvants in allergy vaccines: Past, present and future. Allergol Int 2001; 50:295 - 301; http://dx.doi.org/10.1046/j.1440-1592.2001.00230.x
  • Valenta R, Niederberger V. Recombinant allergens for immunotherapy. J Allergy Clin Immunol 2007; 119:826 - 30; http://dx.doi.org/10.1016/j.jaci.2007.01.025; PMID: 17335886
  • Crameri R, Rhyner C. Novel vaccines and adjuvants for allergen-specific immunotherapy. Curr Opin Immunol 2006; 18:761 - 8; http://dx.doi.org/10.1016/j.coi.2006.09.001; PMID: 17010585
  • Vajdy M, Srivastava I, Polo J, Donnelly J, O’Hagan D, Singh M. Mucosal adjuvants and delivery systems for protein-, DNA- and RNA-based vaccines. Immunol Cell Biol 2004; 82:617 - 27; http://dx.doi.org/10.1111/j.1440-1711.2004.01288.x; PMID: 15550120
  • Chapman MD, Wünschmann S, Pomés A. Proteases as Th2 adjuvants. Curr Allergy Asthma Rep 2007; 7:363 - 7; http://dx.doi.org/10.1007/s11882-007-0055-6; PMID: 17697645
  • Trivedi B, Valerio C, Slater JE. Endotoxin content of standardized allergen vaccines. J Allergy Clin Immunol 2003; 111:777 - 83; http://dx.doi.org/10.1067/mai.2003.1338; PMID: 12704357
  • Till SJ, Francis JN, Nouri-Aria K, Durham SR. Mechanisms of immunotherapy. J Allergy Clin Immunol 2004; 113:1025 - 34, quiz 1035; http://dx.doi.org/10.1016/j.jaci.2004.03.024; PMID: 15208578
  • Moingeon P, Batard T, Fadel R, Frati F, Sieber J, Van Overtvelt L. Immune mechanisms of allergen-specific sublingual immunotherapy. Allergy 2006; 61:151 - 65; http://dx.doi.org/10.1111/j.1398-9995.2006.01002.x; PMID: 16409190
  • Francis JN, Till SJ, Durham SR. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. J Allergy Clin Immunol 2003; 111:1255 - 61; http://dx.doi.org/10.1067/mai.2003.1570; PMID: 12789226
  • Durham SR, Ying S, Varney VA, Jacobson MR, Sudderick RM, Mackay IS, et al. Grass pollen immunotherapy inhibits allergen-induced infiltration of CD4+ T lymphocytes and eosinophils in the nasal mucosa and increases the number of cells expressing messenger RNA for interferon-gamma. J Allergy Clin Immunol 1996; 97:1356 - 65; http://dx.doi.org/10.1016/S0091-6749(96)70205-1; PMID: 8648033
  • Razafindratsita A, Saint-Lu N, Mascarell L, Berjont N, Bardon T, Betbeder D, et al. Improvement of sublingual immunotherapy efficacy with a mucoadhesive allergen formulation. J Allergy Clin Immunol 2007; 120:278 - 85; http://dx.doi.org/10.1016/j.jaci.2007.04.009; PMID: 17531296
  • Wachholz PA, Durham SR. Induction of ‘blocking’ IgG antibodies during immunotherapy. Clin Exp Allergy 2003; 33:1171 - 4; http://dx.doi.org/10.1046/j.1365-2222.2003.01765.x; PMID: 12956735
  • Durham SR, Varney VA, Gaga M, Jacobson MR, Varga EM, Frew AJ, et al. Grass pollen immunotherapy decreases the number of mast cells in the skin. Clin Exp Allergy 1999; 29:1490 - 6; http://dx.doi.org/10.1046/j.1365-2222.1999.00678.x; PMID: 10520076
  • Allam JP, Peng WM, Appel T, Wenghoefer M, Niederhagen B, Bieber T, et al. Toll-like receptor 4 ligation enforces tolerogenic properties of oral mucosal Langerhans cells. J Allergy Clin Immunol 2008; 121:368 - 74, e1; http://dx.doi.org/10.1016/j.jaci.2007.09.045; PMID: 18036651
  • Mascarell L, Lombardi V, Louise A, Saint-Lu N, Chabre H, Moussu H, et al. Oral dendritic cells mediate antigen-specific tolerance by stimulating TH1 and regulatory CD4+ T cells. J Allergy Clin Immunol 2008; 122:603 - 9, e5; http://dx.doi.org/10.1016/j.jaci.2008.06.034; PMID: 18774396
  • Zuany-Amorim C, Manlius C, Trifilieff A, Brunet LR, Rook G, Bowen G, et al. Long-term protective and antigen-specific effect of heat-killed Mycobacterium vaccae in a murine model of allergic pulmonary inflammation. J Immunol 2002; 169:1492 - 9; PMID: 12133976
  • Akdis CA, Kussebi F, Pulendran B, Akdis M, Lauener RP, Schmidt-Weber CB, et al. Inhibition of T helper 2-type responses, IgE production and eosinophilia by synthetic lipopeptides. Eur J Immunol 2003; 33:2717 - 26; http://dx.doi.org/10.1002/eji.200323329; PMID: 14515255
  • Trujillo-Vargas CM, Mayer KD, Bickert T, Palmetshofer A, Grunewald S, Ramirez-Pineda JR, et al. Vaccinations with T-helper type 1 directing adjuvants have different suppressive effects on the development of allergen-induced T-helper type 2 responses. Clin Exp Allergy 2005; 35:1003 - 13; http://dx.doi.org/10.1111/j.1365-2222.2005.02287.x; PMID: 16120081
  • Santeliz JV, Van Nest G, Traquina P, Larsen E, Wills-Karp M. Amb a 1-linked CpG oligodeoxynucleotides reverse established airway hyperresponsiveness in a murine model of asthma. J Allergy Clin Immunol 2002; 109:455 - 62; http://dx.doi.org/10.1067/mai.2002.122156; PMID: 11897991
  • Johansen P, Senti G, Martinez Gomez JM, Storni T, von Beust BR, Wüthrich B, et al. Toll-like receptor ligands as adjuvants in allergen-specific immunotherapy. Clin Exp Allergy 2005; 35:1591 - 8; http://dx.doi.org/10.1111/j.1365-2222.2005.02384.x; PMID: 16393325
  • Taher YA, van Esch BC, Hofman GA, Henricks PA, van Oosterhout AJ. 1alpha,25-dihydroxyvitamin D3 potentiates the beneficial effects of allergen immunotherapy in a mouse model of allergic asthma: role for IL-10 and TGF-β. J Immunol 2008; 180:5211 - 21; PMID: 18390702
  • Drachenberg KJ, Wheeler AW, Stuebner P, Horak F. A well-tolerated grass pollen-specific allergy vaccine containing a novel adjuvant, monophosphoryl lipid A, reduces allergic symptoms after only four preseasonal injections. Allergy 2001; 56:498 - 505; http://dx.doi.org/10.1034/j.1398-9995.2001.056006498.x; PMID: 11421893
  • Mothes N, Heinzkill M, Drachenberg KJ, Sperr WR, Krauth MT, Majlesi Y, 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 - 208; http://dx.doi.org/10.1046/j.1365-2222.2003.01699.x; PMID: 12956739
  • Creticos PS, Schroeder JT, Hamilton RG, Balcer-Whaley SL, Khattignavong AP, Lindblad R, et al, Immune Tolerance Network Group. Immunotherapy with a ragweed-toll-like receptor 9 agonist vaccine for allergic rhinitis. N Engl J Med 2006; 355:1445 - 55; http://dx.doi.org/10.1056/NEJMoa052916; PMID: 17021320
  • Majak P, Rychlik B, Stelmach I. The effect of oral steroids with and without vitamin D3 on early efficacy of immunotherapy in asthmatic children. Clin Exp Allergy 2009; 39:1830 - 41; http://dx.doi.org/10.1111/j.1365-2222.2009.03357.x; PMID: 19817753
  • Schöll I, Weissenböck A, Förster-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; http://dx.doi.org/10.1111/j.1365-2222.2004.01884.x; PMID: 14987314
  • Sanders MT, Brown LE, Deliyannis G, Pearse MJ. ISCOM-based vaccines: the second decade. Immunol Cell Biol 2005; 83:119 - 28; http://dx.doi.org/10.1111/j.1440-1711.2005.01319.x; PMID: 15748208
  • Kim N, Kwon SS, Lee J, Kim S, Yoo TJ. Protective effect of the DNA vaccine encoding the major house dust mite allergens on allergic inflammation in the murine model of house dust mite allergy. Clin Mol Allergy 2006; 4:1 - 9; http://dx.doi.org/10.1186/1476-7961-4-4; PMID: 16478539
  • Terada T. New therapeutic strategies: a chimeric human-cat fusion protein inhibits allergic reactivity. Clin Exp Allergy 2006; 6:96 - 100; http://dx.doi.org/10.1111/j.1365-2222.2006.00108.x
  • Gerstmayr M, Ilk N, Jahn-Schmid B, Sleytr UB, Bohle B. Natural self-assembly of allergen-S-layer fusion proteins is no prerequisite for reduced allergenicity and T cell stimulatory capacity. Int Arch Allergy Immunol 2009; 149:231 - 8; http://dx.doi.org/10.1159/000199718; PMID: 19218815
  • Schülke S, Burggraf M, Waibler Z, Wangorsch A, Wolfheimer S, Kalinke U, et al. A fusion protein of flagellin and ovalbumin suppresses the TH2 response and prevents murine intestinal allergy. J Allergy Clin Immunol 2011; 128:1340 - 8, e12; http://dx.doi.org/10.1016/j.jaci.2011.07.036; PMID: 21872305
  • Kündig 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; http://dx.doi.org/10.1016/j.jaci.2006.01.040; PMID: 16751015
  • Schmitz N, Dietmeier K, Bauer M, Maudrich M, Utzinger S, Muntwiler S, et al. Displaying Fel d1 on virus-like particles prevents reactogenicity despite greatly enhanced immunogenicity: a novel therapy for cat allergy. J Exp Med 2009; 206:1941 - 55; http://dx.doi.org/10.1084/jem.20090199; PMID: 19667059
  • Senti G, Johansen P, Haug S, Bull C, Gottschaller C, Müller 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; http://dx.doi.org/10.1111/j.1365-2222.2008.03191.x; PMID: 19226280
  • Edlmayr J, Niespodziana K, Linhart B, Focke-Tejkl M, Westritschnig K, Scheiblhofer S, et al. A combination vaccine for allergy and rhinovirus infections based on rhinovirus-derived surface protein VP1 and a nonallergenic peptide of the major timothy grass pollen allergen Phl p 1. J Immunol 2009; 182:6298 - 306; http://dx.doi.org/10.4049/jimmunol.0713622; PMID: 19414783
  • Crameri R, Kündig TM, Akdis CA. Modular antigen-translocation as a novel vaccine strategy for allergen-specific immunotherapy. Curr Opin Allergy Clin Immunol 2009; 9:568 - 73; http://dx.doi.org/10.1097/ACI.0b013e3283310fdf; PMID: 19680120
  • Freytag LC, Clements JD. Mucosal adjuvants. Vaccine 2005; 23:1804 - 13; http://dx.doi.org/10.1016/j.vaccine.2004.11.010; PMID: 15734046
  • Pizza M, Giuliani MM, Fontana MR, Monaci E, Douce G, Dougan G, et al. Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants. Vaccine 2001; 19:2534 - 41; http://dx.doi.org/10.1016/S0264-410X(00)00553-3; PMID: 11257389
  • Goldman M. Translational mini-review series on Toll-like receptors: Toll-like receptor ligands as novel pharmaceuticals for allergic disorders. Clin Exp Immunol 2007; 147:208 - 16; http://dx.doi.org/10.1111/j.1365-2249.2006.03296.x; PMID: 17223960
  • Patel M, Xu D, Kewin P, Choo-Kang B, McSharry C, Thomson NC, et al. TLR2 agonist ameliorates established allergic airway inflammation by promoting Th1 response and not via regulatory T cells. J Immunol 2005; 174:7558 - 63; PMID: 15944255
  • Lombardi V, Van Overtvelt L, Horiot S, Moussu H, Chabre H, Louise A, et al. Toll-like receptor 2 agonist Pam3CSK4 enhances the induction of antigen-specific tolerance via the sublingual route. Clin Exp Allergy 2008; 38:1819 - 29; PMID: 18644025
  • Mascarell L, Van Overtvelt L, Lombardi L. Razafindratsita, Moussu H, Horiot S, Chabre H, Paccaud J-P, Limal D, Moutel S, Bauer J, Chiavaroli C, Moingeon P. A synthetic triacylated pseudodipeptide molecule promotes Th1/T Reg immune responses and enhances tolerance induction via the sublingual route. Vaccine 2007; 26:108 - 18; http://dx.doi.org/10.1016/j.vaccine.2007.10.050; PMID: 18063445
  • Van Overtvelt L, Lombardi V, Razafindratsita A, Saint-Lu N, Horiot S, Moussu H, et al. IL-10-inducing adjuvants enhance sublingual immunotherapy efficacy in a murine asthma model. Int Arch Allergy Immunol 2008; 145:152 - 62; http://dx.doi.org/10.1159/000108140; PMID: 17848808
  • de Roock S, van Elk M, van Dijk MEA, Timmerman HM, Rijkers GT, Prakken BJ, et al. Lactic acid bacteria differ in their ability to induce functional regulatory T cells in humans. Clin Exp Allergy 2010; 40:103 - 10; PMID: 19817754
  • Vliagoftis H, Kouranos VD, Betsi GI, Falagas ME. Probiotics for the treatment of allergic rhinitis and asthma: systematic review of randomized controlled trials. Ann Allergy Asthma Immunol 2008; 101:570 - 9; http://dx.doi.org/10.1016/S1081-1206(10)60219-0; PMID: 19119700
  • Van Overtvelt L, Moussu H, Horiot S, Samson S, Lombardi V, Mascarell L, et al. Lactic acid bacteria as adjuvants for sublingual allergy vaccines. Vaccine 2010; 28:2986 - 92; http://dx.doi.org/10.1016/j.vaccine.2010.02.009; PMID: 20175969
  • Moussu H, Van Overtvelt L, Horiot S, Tourdot S, Airouche S, Zuercher A, et al. Bifidobacterium bifidum NCC 453 promotes tolerance induction in murine models of sublingual immunotherapy. Int Arch Allergy Immunol 2012; 158:35 - 42; http://dx.doi.org/10.1159/000330101; PMID: 22205338
  • Pfaar O, Barth C, Jaschke C, Hörmann K, Klimek L. Sublingual allergen-specific immunotherapy adjuvanted with monophosphoryl lipid A: a phase I/IIa study. Int Arch Allergy Immunol 2011; 154:336 - 44; http://dx.doi.org/10.1159/000321826; PMID: 20975285
  • Arikan C, Bahceciler NN, Deniz G, Akdis M, Akkoc T, Akdis CA, et al. Bacillus Calmette-Guérin-induced interleukin-12 did not additionally improve clinical and immunologic parameters in asthmatic children treated with sublingual immunotherapy. Clin Exp Allergy 2004; 34:398 - 405; http://dx.doi.org/10.1111/j.1365-2222.2004.01869.x; PMID: 15005733
  • Kalliomäki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 2003; 361:1869 - 71; http://dx.doi.org/10.1016/S0140-6736(03)13490-3; PMID: 12788576
  • De Souza Rebouças 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; http://dx.doi.org/10.1155/2012/474605; PMID: 22496608
  • 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; http://dx.doi.org/10.1111/j.1398-9995.2009.01945.x; PMID: 19220212
  • Jeannin P, Renno T, Goetsch L, Miconnet I, Aubry JP, Delneste Y, et al. OmpA targets dendritic cells, induces their maturation and delivers antigen into the MHC class I presentation pathway. Nat Immunol 2000; 1:502 - 9; http://dx.doi.org/10.1038/82751; PMID: 11101872
  • Goetsch L, Gonzalez A, Plotnicky-Gilquin H, Haeuw JF, Aubry JP, Beck A, et al. Targeting of nasal mucosa-associated antigen-presenting cells in vivo with an outer membrane protein A derived from Klebsiella pneumoniae. Infect Immun 2001; 69:6434 - 44; http://dx.doi.org/10.1128/IAI.69.10.6434-6444.2001; PMID: 11553588
  • Haicheur N, Benchetrit F, Amessou M, Leclerc C, Falguières T, Fayolle C, et al. The B subunit of Shiga toxin coupled to full-size antigenic protein elicits humoral and cell-mediated immune responses associated with a Th1-dominant polarization. Int Immunol 2003; 15:1161 - 71; http://dx.doi.org/10.1093/intimm/dxg118; PMID: 13679386
  • El Azami El Idrissi M, Ladant D, Leclerc C. The adenylate cyclase of Bordetella pertussis: a vector to target antigen presenting cells. Toxicon 2002; 40:1661 - 5; http://dx.doi.org/10.1016/S0041-0101(02)00205-2; PMID: 12457876
  • Mascarell L, Saint-Lu N, Moussu H, Zimmer A, Louise A, Lone Y, et al. Oral macrophage-like cells play a key role in tolerance induction following sublingual immunotherapy of asthmatic mice. Mucosal Immunol 2011; 4:638 - 47; http://dx.doi.org/10.1038/mi.2011.28; PMID: 21775981
  • Hobson P, Barnfield C, Barnes A, Klavinskis LS. Mucosal immunization with DNA vaccines. Methods 2003; 31:217 - 24; http://dx.doi.org/10.1016/S1046-2023(03)00139-7; PMID: 14511954
  • 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; http://dx.doi.org/10.1016/j.vaccine.2010.05.004; PMID: 20478343
  • Hu KF, Lövgren-Bengtsson K, Morein B. Immunostimulating complexes (ISCOMs) for nasal vaccination. Adv Drug Deliv Rev 2001; 51:149 - 59; http://dx.doi.org/10.1016/S0169-409X(01)00165-X; PMID: 11516786

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.