Skip to main content
Log in

Microbial Adjuncts for Food Allergen Immunotherapy

  • Immunotherapy and Immunomodulators (B Vickery, Section Editor)
  • Published:
Current Allergy and Asthma Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Food allergen immunotherapy may benefit from adjunct therapies to enhance safety and efficacy. We review preclinical studies investigating the effects of probiotics and other microbial-based interventions on oral tolerance, describe the human clinical trial evidence thus far for microbial adjuncts, and discuss steps for translating research findings in this area to clinical therapy.

Recent Findings

Murine studies support that microbial-based interventions confer protection against sensitization and may augment treatment efficacy for food allergy. Microbial adjunct therapies can promote regulatory T cells and modulate Th1 vs. Th2 responses. There is a wide array of novel modalities utilizing microbial components. Ongoing efforts are focused on translating preclinical data into potential treatments.

Summary

Probiotics, prebiotics, and microbial components have all been examined as microbial adjunct therapies in murine models of food allergy. The effects of probiotics appear to be strain-specific. Prebiotics and bacterial components are innovative modalities to modulate oral tolerance. Better characterization of dysbiosis in human cohorts with food allergy, deeper mechanistic understanding of microbial adjunct therapies, safety evaluation, and careful clinical trial design will be crucial for the development of microbial adjuncts for food allergen immunotherapy. Microbial adjunct therapies have the potential to enhance the efficacy, safety, and durability of food allergen immunotherapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Sicherer SH, Sampson HA. Food allergy: a review and update on epidemiology, pathogenesis, diagnosis, prevention, and management. J Allergy Clin Immunol. 2018;141(1):41–58. https://doi.org/10.1016/j.jaci.2017.11.003.

    Article  CAS  PubMed  Google Scholar 

  2. Osborne NJ, Koplin JJ, Martin PE, Gurrin LC, Lowe AJ, Matheson MC, et al. Prevalence of challenge-proven IgE-mediated food allergy using population-based sampling and predetermined challenge criteria in infants. J Allergy Clin Immunol. 2011;127(3):668–76 e1-2. https://doi.org/10.1016/j.jaci.2011.01.039.

    Article  CAS  PubMed  Google Scholar 

  3. Nowak-Wegrzyn A, Wood RA, Nadeau KC, Pongracic JA, Henning AK, Lindblad RW, et al. Multicenter, randomized, double-blind, placebo-controlled clinical trial of vital wheat gluten oral immunotherapy. J Allergy Clin Immunol. 2018.

  4. •• Investigators PGoC, Vickery BP, Vereda A, Casale TB, Beyer K, du Toit G, et al. AR101 oral immunotherapy for peanut allergy. N Engl J Med. 2018;379(21):1991–2001. This study is the first large scale phase 3 randomized, controlled trial for peanut OIT. In the age 4 to 17 years cohort, 67.2% of active treated group, as compared to 4.0% of the placebo group, were able to ingest 600 mg or more of peanut protein without symptoms. Efficacy was not shown in participants age 18 years or older. Adverse events during treatment were observed in more than 95% of the participants age 4 to 17 years. The study highlights the efficacy and current limitations of OIT. https://doi.org/10.1056/NEJMoa1812856.

    Article  Google Scholar 

  5. Nurmatov U, Dhami S, Arasi S, Pajno GB, Fernandez-Rivas M, Muraro A, et al. Allergen immunotherapy for IgE-mediated food allergy: a systematic review and meta-analysis. Allergy. 2017;72(8):1133–47. https://doi.org/10.1111/all.13124.

    Article  CAS  Google Scholar 

  6. Anagnostou K, Islam S, King Y, Foley L, Pasea L, Bond S, et al. Assessing the efficacy of oral immunotherapy for the desensitisation of peanut allergy in children (STOP II): a phase 2 randomised controlled trial. Lancet. 2014;383(9925):1297–304. https://doi.org/10.1016/S0140-6736(13)62301-6.

    Article  CAS  Google Scholar 

  7. Jones SM, Pons L, Roberts JL, Scurlock AM, Perry TT, Kulis M, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol. 2009;124(2):292–300, e1-97. https://doi.org/10.1016/j.jaci.2009.05.022.

    Article  Google Scholar 

  8. Clark AT, Islam S, King Y, Deighton J, Anagnostou K, Ewan PW. Successful oral tolerance induction in severe peanut allergy. Allergy. 2009;64(8):1218–20. https://doi.org/10.1111/j.1398-9995.2009.01982.x.

    Article  CAS  PubMed  Google Scholar 

  9. Perkin MR. Oral desensitization to peanuts. N Engl J Med. 2018;379(21):2074–5. https://doi.org/10.1056/NEJMe1813314.

    Article  PubMed  Google Scholar 

  10. Zhao W, Ho HE, Bunyavanich S. The gut microbiome in food allergy. Ann Allergy Asthma Immunol. 2018.

  11. Ho HE, Bunyavanich S. Role of the microbiome in food allergy. Curr Allergy Asthma Rep. 2018;18(4):27.

    Article  Google Scholar 

  12. Sampson HA, O'Mahony L, Burks AW, Plaut M, Lack G, Akdis CA. Mechanisms of food allergy. J Allergy Clin Immunol. 2018;141(1):11–9. https://doi.org/10.1016/j.jaci.2017.11.005.

    Article  CAS  PubMed  Google Scholar 

  13. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775–87. https://doi.org/10.1016/j.cell.2008.05.009.

    Article  CAS  PubMed  Google Scholar 

  14. Hadis U, Wahl B, Schulz O, Hardtke-Wolenski M, Schippers A, Wagner N, et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 2011;34(2):237–46. https://doi.org/10.1016/j.immuni.2011.01.016.

    Article  CAS  Google Scholar 

  15. Cassani B, Villablanca EJ, Quintana FJ, Love PE, Lacy-Hulbert A, Blaner WS, et al. Gut-tropic T cells that express integrin alpha4beta7 and CCR9 are required for induction of oral immune tolerance in mice. Gastroenterology. 2011;141(6):2109–18. https://doi.org/10.1053/j.gastro.2011.09.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Noval Rivas M, Burton OT, Wise P, Charbonnier LM, Georgiev P, Oettgen HC, et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42(3):512–23. https://doi.org/10.1016/j.immuni.2015.02.004.

    Article  Google Scholar 

  17. Lee JB, Chen CY, Liu B, Mugge L, Angkasekwinai P, Facchinetti V, et al. IL-25 and CD4(+) TH2 cells enhance type 2 innate lymphoid cell-derived IL-13 production, which promotes IgE-mediated experimental food allergy. J Allergy Clin Immunol. 2016;137(4):1216–25 e5. https://doi.org/10.1016/j.jaci.2015.09.019.

    Article  Google Scholar 

  18. Gernez Y, Nowak-Wegrzyn A. Immunotherapy for food allergy: are we there yet? J Allergy Clin Immunol Pract. 2017;5(2):250–72. https://doi.org/10.1016/j.jaip.2016.12.004.

    Article  PubMed  Google Scholar 

  19. Tordesillas L, Berin MC. Mechanisms of oral tolerance. Clin Rev Allergy Immunol. 2018;55(2):107–17. https://doi.org/10.1007/s12016-018-8680-5.

    Article  CAS  PubMed  Google Scholar 

  20. Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol. 2016;16(12):751–65. https://doi.org/10.1038/nri.2016.111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thyagarajan A, Jones SM, Calatroni A, Pons L, Kulis M, Woo CS, et al. Evidence of pathway-specific basophil anergy induced by peanut oral immunotherapy in peanut-allergic children. Clin Exp Allergy. 2012;42(8):1197–205. https://doi.org/10.1111/j.1365-2222.2012.04028.x.

    Article  CAS  Google Scholar 

  22. Kulis M, Saba K, Kim EH, Bird JA, Kamilaris N, Vickery BP, et al. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut sublingual immunotherapy. J Allergy Clin Immunol. 2012;129(4):1159–62. https://doi.org/10.1016/j.jaci.2011.11.045.

    Article  CAS  Google Scholar 

  23. Vickery BP, Lin J, Kulis M, Fu Z, Steele PH, Jones SM, et al. Peanut oral immunotherapy modifies IgE and IgG4 responses to major peanut allergens. The J Allergy Clin Immunol. 2013;131(1):128–34 12e1–3.

    Article  Google Scholar 

  24. Varshney P, Jones SM, Scurlock AM, Perry TT, Kemper A, Steele P, et al. A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. J Allergy Clin Immunol. 2011;127(3):654–60. https://doi.org/10.1016/j.jaci.2010.12.1111.

    Article  CAS  Google Scholar 

  25. Vickery BP, Scurlock AM, Jones SM, Burks AW. Mechanisms of immune tolerance relevant to food allergy. J Allergy Clin Immunol. 2011;127(3):576–84; quiz 85-6. https://doi.org/10.1016/j.jaci.2010.12.1116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. • Syed A, Garcia MA, Lyu SC, Bucayu R, Kohli A, Ishida S. et al.Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol. 2014;133(2):500–10. This study evaluated the immune profile of subjects who underwent peanut OIT. After completion of peanut OIT and 3 months of therapy withdrawl, T-cell function and demethylation of FOXP3 CpG sites in antigen-induced Treg cells were signficantly different between peanut-tolerant versus nontolerant subjects. The results implicated Treg cells as a part of the mechanisms of actions of OIT. https://doi.org/10.1016/j.jaci.2013.12.1037.

    Article  Google Scholar 

  27. Jones SM, Sicherer SH, Burks AW, Leung DY, Lindblad RW, Dawson P, et al. Epicutaneous immunotherapy for the treatment of peanut allergy in children and young adults. J Allergy Clin Immunol. 2017;139(4):1242–52 e9. https://doi.org/10.1016/j.jaci.2016.08.017.

    Article  Google Scholar 

  28. Hofmann AM, Scurlock AM, Jones SM, Palmer KP, Lokhnygina Y, Steele PH, et al. Safety of a peanut oral immunotherapy protocol in children with peanut allergy. J Allergy Clin Immunol. 2009;124(2):286–91, 91 e1–6. https://doi.org/10.1016/j.jaci.2009.03.045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Virkud YV, Burks AW, Steele PH, Edwards LJ, Berglund JP, Jones SM, et al. Novel baseline predictors of adverse events during oral immunotherapy in children with peanut allergy. J Allergy Clin Immunol. 2017;139(3):882–8 e5. https://doi.org/10.1016/j.jaci.2016.07.030.

    Article  Google Scholar 

  30. Wasserman RL, Factor JM, Baker JW, Mansfield LE, Katz Y, Hague AR, et al. Oral immunotherapy for peanut allergy: multipractice experience with epinephrine-treated reactions. J Allergy Clin Immunol Pract. 2014;2(1):91–6. https://doi.org/10.1016/j.jaip.2013.10.001.

    Google Scholar 

  31. Varshney P, Steele PH, Vickery BP, Bird JA, Thyagarajan A, Scurlock AM, et al. Adverse reactions during peanut oral immunotherapy home dosing. J Allergy Clin Immunol. 2009;124(6):1351–2. https://doi.org/10.1016/j.jaci.2009.09.042.

    Article  Google Scholar 

  32. McCutcheon JP, Moran NA. Extreme genome reduction in symbiotic bacteria. Nat Rev Microbiol. 2011;10(1):13–26. https://doi.org/10.1038/nrmicro2670.

    Article  CAS  PubMed  Google Scholar 

  33. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16(6):341–52. https://doi.org/10.1038/nri.2016.42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al. Host-gut microbiota metabolic interactions. Science. 2012;336(6086):1262–7. https://doi.org/10.1126/science.1223813.

    Article  CAS  Google Scholar 

  35. Shreiner AB, Kao JY, Young VB. The gut microbiome in health and in disease. Curr Opin Gastroenterol. 2015;31(1):69–75. https://doi.org/10.1097/MOG.0000000000000139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Huang YJ, Marsland BJ, Bunyavanich S, O'Mahony L, Leung DY, Muraro A, et al. The microbiome in allergic disease: current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J Allergy Clin Immunol. 2017;139(4):1099–110. https://doi.org/10.1016/j.jaci.2017.02.007.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Fazlollahi M, Chun Y, Grishin A, Wood RA, Burks AW, Dawson P, et al. Early-life gut microbiome and egg allergy. Allergy. 2018;73(7):1515–24. https://doi.org/10.1111/all.13389.

    Article  CAS  Google Scholar 

  38. Bunyavanich S, Shen N, Grishin A, Wood R, Burks W, Dawson P, et al. Early-life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol. 2016;138(4):1122–30. https://doi.org/10.1016/j.jaci.2016.03.041.

    Article  CAS  Google Scholar 

  39. Rodriguez B, Prioult G, Hacini-Rachinel F, Moine D, Bruttin A, Ngom-Bru C, et al. Infant gut microbiota is protective against cow's milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol. 2012;79(1):192–202. https://doi.org/10.1111/j.1574-6941.2011.01207.x.

    Article  Google Scholar 

  40. Feehley T, Plunkett CH, Bao R, Choi Hong SM, Culleen E, Belda-Ferre P, et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat Med. 2019. https://doi.org/10.1038/s41591-018-0324-z.

    Article  Google Scholar 

  41. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331(6015):337–41. https://doi.org/10.1126/science.1198469.

    Article  Google Scholar 

  42. •• Narushima S, Sugiura Y, Oshima K, Atarashi K, Hattori M, Suematsu M. et al.Characterization of the 17 strains of regulatory T cell-inducing human-derived Clostridia. Gut Microbes. 2014;5(3):333–9. In line with the concept of probiotics, this group isolated 17 strains of Clostridia from heathy human fecal samples that are capable of attenuating the symptoms of OVA-induced allergic diarrhea when introduced in mice. These 17 Clostridia strains significantly increased the number and function of colonic Treg cells in colonized mice. https://doi.org/10.4161/gmic.28572.

    Article  Google Scholar 

  43. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A. 2014;111(36):13145–50.

    Article  CAS  Google Scholar 

  44. Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol. 1997;159(4):1739–45.

    CAS  PubMed  Google Scholar 

  45. Aitoro R, Simeoli R, Amoroso A, Paparo L, Nocerino R, Pirozzi C, et al. Extensively hydrolyzed casein formula alone or with L. rhamnosus GG reduces beta-lactoglobulin sensitization in mice. Pediatr Allergy Immunol. 2017;28(3):230–7. https://doi.org/10.1111/pai.12687.

    Article  Google Scholar 

  46. •• Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, et al. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 2016;15(12):2809–24. In line with the concept of prebiotics, the group demonstrated that a high-fiber diet in mice was protective for food allergy in a peanut sensitization model. High-fiber feeding was shown to reshape the gut microbiota and increase the release of select short-chain fatty acids, leading to enhanced tolerogenic CD103+ DC activities in a vitamin A-dependent manner. https://doi.org/10.1016/j.celrep.2016.05.047.

    Article  CAS  PubMed  Google Scholar 

  47. Cait A, Hughes MR, Antignano F, Cait J, Dimitriu PA, Maas KR, et al. Microbiome-driven allergic lung inflammation is ameliorated by short-chain fatty acids. Mucosal Immunol. 2018;11(3):785–95.

    Article  Google Scholar 

  48. Zhu FG, Kandimalla ER, Yu D, Agrawal S. Oral administration of a synthetic agonist of Toll-like receptor 9 potently modulates peanut-induced allergy in mice. J Allergy Clin Immunol. 2007;120(3):631–7. https://doi.org/10.1016/j.jaci.2007.05.015.

    Article  CAS  PubMed  Google Scholar 

  49. Kulis M, Gorentla B, Burks AW, Zhong XP. Type B CpG oligodeoxynucleotides induce Th1 responses to peanut antigens: modulation of sensitization and utility in a truncated immunotherapy regimen in mice. Mol Nutr Food Res. 2013;57(5):906–15. https://doi.org/10.1002/mnfr.201200410.

    Article  CAS  PubMed  Google Scholar 

  50. Soos TJ, Li L, Graver K, Schiding J, Xenos A, Chen D, et al. Glucopyranosyl lipid A (GLA) a Toll-like receptor 4 (TLR4) agonist for use as an adjuvant in combination with peanut allergen immunotherapy. J Allergy Clin Immunol. 2016;137(2):AB129. https://doi.org/10.1016/j.jaci.2015.12.555.

    Article  Google Scholar 

  51. • Kim JH, Jeun EJ, Hong CP, Kim SH, Jang MS, Lee EJ, et al. Extracellular vesicle-derived protein from Bifidobacterium longum alleviates food allergy through mast cell suppression. J Allergy Clin Immunol. 2016;137(2):507–16 e8. This group identified an extracellular vesicle-derived protein from Bifidobacterium longum that is capable of suppressing mast cell response through mast cell-specific apoptosis. Injection of this bacterial protein reduced allergic diarrhea symptoms in an egg OVA sensitization murine model. https://doi.org/10.1016/j.jaci.2015.08.016.

    PubMed  Google Scholar 

  52. Matteoli G, Mazzini E, Iliev ID, Mileti E, Fallarino F, Puccetti P, et al. Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut. 2010;59(5):595–604. https://doi.org/10.1136/gut.2009.185108.

    Article  CAS  Google Scholar 

  53. Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity. 2008;28(4):546–58. https://doi.org/10.1016/j.immuni.2008.02.017.

    Article  CAS  Google Scholar 

  54. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–6. https://doi.org/10.1038/nature12331.

    Article  CAS  Google Scholar 

  55. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, de Roos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5. https://doi.org/10.1038/nature12726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–73. https://doi.org/10.1126/science.1241165.

    Article  CAS  PubMed  Google Scholar 

  57. Berni Canani R, Sangwan N, Stefka AT, Nocerino R, Paparo L, Aitoro R, et al. Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. The ISME Journal. 2016;10(3):742–50. https://doi.org/10.1038/ismej.2015.151.

    Article  Google Scholar 

  58. Dabbagh K, Lewis DB. Toll-like receptors and T-helper-1/T-helper-2 responses. Curr Opin Infect Dis. 2003;16(3):199–204. https://doi.org/10.1097/00001432-200306000-00003.

    Article  PubMed  Google Scholar 

  59. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000;408(6813):740–5. https://doi.org/10.1038/35047123.

    Article  CAS  Google Scholar 

  60. Kim EH, Bird JA, Kulis M, Laubach S, Pons L, Shreffler W, et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2011;127(3):640–6 e1. https://doi.org/10.1016/j.jaci.2010.12.1083.

    Article  Google Scholar 

  61. Patel P, Holdich T, Fischer von Weikersthal-Drachenberg KJ, Huber B. Efficacy of a short course of specific immunotherapy in patients with allergic rhinoconjunctivitis to ragweed pollen. J Allergy Clin Immunol. 2014;133(1):121–9 e1–2. https://doi.org/10.1016/j.jaci.2013.05.032.

    Article  PubMed  Google Scholar 

  62. Shen Y, Giardino Torchia ML, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe. 2012;12(4):509–20. https://doi.org/10.1016/j.chom.2012.08.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. • Tang ML, Ponsonby AL, Orsini F, Tey D, Robinson M, Su EL, et al. Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol. 2015;135(3):737–44 e8. This was the first placebo-controlled randomized peanut OIT trial utilizing probiotic (Lactobacillus rhamnosus GG) as adjunct therapy. A high prevalence of sustained unresponsiveness to peanut (82.1%) was reported in the treated children; however, the specific contribution of probiotics was unclear due to the lack of an OIT-only treatment arm. https://doi.org/10.1016/j.jaci.2014.11.034.

    Article  Google Scholar 

  64. Hsiao KC, Ponsonby AL, Axelrad C, Pitkin S, Tang MLK, Team PS. Long-term clinical and immunological effects of probiotic and peanut oral immunotherapy after treatment cessation: 4-year follow-up of a randomised, double-blind, placebo-controlled trial. Lancet Child Adolesc Health. 2017;1(2):97–105. https://doi.org/10.1016/S2352-4642(17)30041-X.

    Article  PubMed  Google Scholar 

  65. Tang MLK, Hsiao KC, Ponsonby AL, Donath S, Orsini F, Axelrad C, et al. Probiotics and oral immunotherapy for peanut allergy—authors' reply. Lancet Child Adolesc Health. 2017;1(3):e1–2. https://doi.org/10.1016/S2352-4642(17)30101-3.

    Article  Google Scholar 

  66. Mileti E, Matteoli G, Iliev ID, Rescigno M. Comparison of the immunomodulatory properties of three probiotic strains of Lactobacilli using complex culture systems: prediction for in vivo efficacy. PLoS One. 2009;4(9):e7056. https://doi.org/10.1371/journal.pone.0007056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol. 2013;14(7):676–84. https://doi.org/10.1038/ni.2640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Dardalhon V, Korn T, Kuchroo VK, Anderson AC. Role of Th1 and Th17 cells in organ-specific autoimmunity. J Autoimmun. 2008;31(3):252–6. https://doi.org/10.1016/j.jaut.2008.04.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Levy MB, Elizur A, Goldberg MR, Nachshon L, Katz Y. Clinical predictors for favorable outcomes in an oral immunotherapy program for IgE-mediated cow's milk allergy. Ann Allergy Asthma Immunol. 2014;112(1):58–63 e1. https://doi.org/10.1016/j.anai.2013.10.001.

    Article  CAS  PubMed  Google Scholar 

  70. Wright BL, Kulis M, Orgel KA, Burks AW, Dawson P, Henning AK, et al. Component-resolved analysis of IgA, IgE, and IgG4 during egg OIT identifies markers associated with sustained unresponsiveness. Allergy. 2016;71(11):1552–60. https://doi.org/10.1111/all.12895.

    Article  CAS  Google Scholar 

  71. Vazquez-Ortiz M, Alvaro M, Piquer M, Giner MT, Dominguez O, Lozano J, et al. Life-threatening anaphylaxis to egg and milk oral immunotherapy in asthmatic teenagers. Ann Allergy Asthma Immunol. 2014;113(4):482–4. https://doi.org/10.1016/j.anai.2014.07.010.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Supinda Bunyavanich.

Ethics declarations

Conflict of Interest

Dr. Bunyavanich reports grants from the National Institutes of Health, during the conduct of the study. Dr. Hsi-en Ho declares no conflicts of interest relevant to this manuscript.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Immunotherapy and Immunomodulators

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ho, He., Bunyavanich, S. Microbial Adjuncts for Food Allergen Immunotherapy. Curr Allergy Asthma Rep 19, 25 (2019). https://doi.org/10.1007/s11882-019-0859-1

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11882-019-0859-1

Keywords

Navigation