Skip to main content

Advertisement

Log in

Biomarkers in Food Allergy Immunotherapy

  • Food Allergy (E Kim, Section Editor)
  • Published:
Current Allergy and Asthma Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Investigational allergen immunotherapies (AITs) including oral immunotherapy (OIT), sublingual immunotherapy (SLIT), and epicutaneous immunotherapy (EPIT) have proven to increase allergen thresholds required to elicit an allergic reaction in a majority of subjects. However, these studies lack consistent biomarkers to predict therapy outcomes. Here, we will review biomarkers that are currently being investigated for AIT.

Recent Findings

The mechanisms underlying the therapeutic benefit of AIT involve various cell types, including mast cells, basophils, T cells, and B cells. Skin prick and basophil activation tests assess effector cell sensitivity to allergen and are decreased in subjects on AIT. Allergen-specific IgE increases initially and decreases with continued therapy, while allergen-specific IgG and IgA increase throughout therapy. Allergen-induced regulatory T cells (Tregs) increase throughout therapy and were found to be associated with sustained unresponsiveness after OIT. Subjects on OIT and SLIT have decreased Th2 cytokine production during therapy.

Summary

Although trends have been reported, a common limitation of these biomarkers is that none are able to reproducibly predict prognosis during AIT. Further studies are needed to expand the currently available biomarker repertoire to provide personalized approaches to AIT.

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.

Similar content being viewed by others

Abbreviations

AIT:

Allergen immunotherapy

BAT:

Basophil activation test

DBPCFC:

Double-blind, placebo-controlled food challenge

EPIT:

Epicutaneous immunotherapy

MAT:

Mast cell activation test

OIT:

Oral immunotherapy

SLIT:

Sublingual immunotherapy

SPT:

Skin prick test

References

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

  1. Branum AM, Lukacs SL (2008) Food allergy among U.S. children: trends in prevalence and hospitalizations. NCHS Data Brief

  2. Gupta RS, Springston EE, Warrier MR, Smith B, Kumar R, Pongracic J, et al. The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics. 2011. https://doi.org/10.1542/peds.2011-0204.

  3. Gupta RS, Warren CM, Smith BM, Jiang J, Blumenstock JA, Davis MM, et al. Prevalence and Severity of Food Allergies Among US Adults. JAMA Netw Open. 2019. https://doi.org/10.1001/jamanetworkopen.2018.5630.

  4. Avery NJ, King RM, Knight S, Hourihane JOB. Assessment of quality of life in children with peanut allergy. Pediatr Allergy Immunol. 2003. https://doi.org/10.1034/j.1399-3038.2003.00072.x.

  5. Longo G, Berti I, Burks AW, Krauss B, Barbi E. IgE-mediated food allergy in children. Lancet. 2013. https://doi.org/10.1016/S0140-6736(13)60309-8.

  6. Sampson HA, O’Mahony L, Burks AW, Plaut M, Lack G, Akdis CA. Mechanisms of food allergy. J Allergy Clin Immunol. 2018. https://doi.org/10.1016/j.jaci.2017.11.005.

  7. Tordesillas L, Berin MC, Sampson HA. Immunology of Food Allergy. Immunity. 2017. https://doi.org/10.1016/j.immuni.2017.07.004.

  8. Vickery BP, Scurlock AM, Kulis M, et al. Sustained unresponsiveness to peanut in subjects who have completed peanut oral immunotherapy. J Allergy Clin Immunol. 2014. https://doi.org/10.1016/j.jaci.2013.11.007.

  9. Vickery BP, Berglund JP, Burk CM, et al. Early oral immunotherapy in peanut-allergic preschool children is safe and highly effective. J Allergy Clin Immunol. 2017. https://doi.org/10.1016/j.jaci.2016.05.027.

  10. Jones SM, Burks AW, Keet C, et al. Long-term treatment with egg oral immunotherapy enhances sustained unresponsiveness that persists after cessation of therapy. J Allergy Clin Immunol. 2016. https://doi.org/10.1016/j.jaci.2015.12.1316.

  11. • Vickery BP, Vereda A, Casale TB, et al. AR101 oral immunotherapy for peanut allergy. N Engl J Med. 2018. https://doi.org/10.1056/NEJMoa1812856. This recent, phase 3 OIT study demonstrates changes in various biomarkers.

  12. Keet CA, Frischmeyer-Guerrerio PA, Thyagarajan A, et al. The safety and efficacy of sublingual and oral immunotherapy for milk allergy. J Allergy Clin Immunol. 2012. https://doi.org/10.1016/j.jaci.2011.10.023.

  13. 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. https://doi.org/10.1016/j.jaci.2010.12.1083.

  14. Kim EH, Yang L, Ye P, Guo R, Li Q, Kulis MD, et al. Long-term sublingual immunotherapy for peanut allergy in children: Clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2019. https://doi.org/10.1016/j.jaci.2019.07.030.

  15. Fleischer DM, Burks AW, Vickery BP, et al. Sublingual immunotherapy for peanut allergy: A randomized, double-blind, placebo-controlled multicenter trial. J Allergy Clin Immunol. 2013. https://doi.org/10.1016/j.jaci.2012.11.011.

  16. Narisety SD, Frischmeyer-Guerrerio PA, Keet CA, Gorelik M, Schroeder J, Hamilton RG, et al. A randomized, double-blind, placebo-controlled pilot study of sublingual versus oral immunotherapy for the treatment of peanut allergy. J Allergy Clin Immunol. 2015. https://doi.org/10.1016/j.jaci.2014.11.005.

  17. Fleischer DM, Greenhawt M, Sussman G, et al. Effect of Epicutaneous Immunotherapy vs Placebo on Reaction to Peanut Protein Ingestion among Children with Peanut Allergy: The PEPITES Randomized Clinical Trial. JAMA - J Am Med Assoc. 2019. https://doi.org/10.1001/jama.2019.1113.

  18. Allan Bock S, Sampson HA, Atkins FM, Zeiger RS, Lehrer S, Sachs M, et al. Double-blind, placebo-controlled food challenge (DBPCFC) as an office procedure: A manual. J Allergy Clin Immunol. 1988. https://doi.org/10.1016/0091-6749(88)90135-2.

  19. Sicherer SH, Morrow EH, Sampson HA. Dose-response in double-blind, placebo-controlled oral food challenges in children with atopic dermatitis. J Allergy Clin Immunol. 2000. https://doi.org/10.1067/mai.2000.104941.

  20. Hill DJ, Heine RG, Hosking CS. The diagnostic value of skin prick testing in children with food allergy. Pediatr Allergy Immunol. 2004. https://doi.org/10.1111/j.1399-3038.2004.00188.x.

  21. Eigenmann PA, Sampson HA. Interpreting skin prick tests in the evaluation of food allergy in children. Pediatr Allergy Immunol. 1998. https://doi.org/10.1111/j.1399-3038.1998.tb00371.x.

  22. Sporik R, Hill DJ, Hosking CS. Specificity of allergen skin testing in predicting positive open food challenges to milk, egg and peanut in children. Clin Exp Allergy. 2000. https://doi.org/10.1046/j.1365-2222.2000.00928.x.

  23. Varshney P, Jones SM, Scurlock AM, et al. A randomized controlled study of peanut oral immunotherapy: Clinical desensitization and modulation of the allergic response. J Allergy Clin Immunol. 2011. https://doi.org/10.1016/j.jaci.2010.12.1111.

  24. Vickery BP, Pons L, Kulis M, Steele P, Jones SM, Burks AW. Individualized IgE-based dosing of egg oral immunotherapy and the development of tolerance. Ann Allergy, Asthma Immunol. 2010. https://doi.org/10.1016/j.anai.2010.09.030.

  25. Fernández-Rivas M, Garrido Fernández S, Nadal JA, De Durana MDAD, García BE, González-Mancebo E, et al. Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract. Allergy Eur J Allergy Clin Immunol. 2009. https://doi.org/10.1111/j.1398-9995.2008.01921.x.

  26. Burks AW, Jones SM, Wood RA, et al. Oral immunotherapy for treatment of egg allergy in children. N Engl J Med. 2012. https://doi.org/10.1056/NEJMoa1200435.

  27. Skripak JM, Nash SD, Rowley H, Brereton NH, Oh S, Hamilton RG, et al. A randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow’s milk allergy. J Allergy Clin Immunol. 2008. https://doi.org/10.1016/j.jaci.2008.09.030.

  28. •• Jones SM, Sicherer SH, Burks AW, et al. Epicutaneous immunotherapy for the treatment of peanut allergy in children and young adults. J Allergy Clin Immunol. 2017. https://doi.org/10.1016/j.jaci.2016.08.017. This study reported food allergen EPIT induced changes in biomarkers.

  29. Kabashima K, Nakashima C, Nonomura Y, Otsuka A, Cardamone C, Parente R, et al. Biomarkers for evaluation of mast cell and basophil activation. Immunol Rev. 2018. https://doi.org/10.1111/imr.12639.

  30. MacGlashan D. Expression of CD203c and CD63 in human basophils: Relationship to differential regulation of piecemeal and anaphylactic degranulation processes. Clin Exp Allergy. 2010. https://doi.org/10.1111/j.1365-2222.2010.03572.x.

  31. Knol EF, Mul FPJ, Jansen H, Calafat J, Roos D. Monitoring human basophil activation via CD63 monoclonal antibody 435. J Allergy Clin Immunol. 1991. https://doi.org/10.1016/0091-6749(91)90094-5.

  32. Bühring HJ, Giesert C, Seiffert M, Marxer A, Valent P, Kanz L. The basophil activation marker defined by antibody 97a6 is identical with the ectoenzyme npp3/e-npp3 (cd203c). Blood. 2000.

  33. Hennersdorf F, Florian S, Jakob A, Baumgärtner K, Sonneck K, Nordheim A, et al. Identification of CD13, CD107a, and CD164 as novel basophil-activation markers and dissection of two response patterns in time kinetics of IgE-dependent upregulation. Cell Res. 2005. https://doi.org/10.1038/sj.cr.7290301.

  34. Kouser L, Kappen J, Walton RP, Shamji MH. Update on Biomarkers to Monitor Clinical Efficacy Response During and Post Treatment in Allergen Immunotherapy. Curr Treat Options Allergy. 2017. https://doi.org/10.1007/s40521-017-0117-5.

  35. Thyagarajan A, Jones SM, Calatroni A, et al. Evidence of pathway-specific basophil anergy induced by peanut oral immunotherapy in peanut-allergic children. Clin Exp Allergy. 2012. https://doi.org/10.1111/j.1365-2222.2012.04028.x.

  36. •• Patil SU, Steinbrecher J, Calatroni A, Smith N, Ma A, Ruiter B, et al. Early decrease in basophil sensitivity to Ara h 2 precedes sustained unresponsiveness after peanut oral immunotherapy. J Allergy Clin Immunol. 2019. https://doi.org/10.1016/j.jaci.2019.07.028. This OIT study correlates basophil activation to sustained unresponsiveness.

  37. Wright BL, Kulis M, Orgel KA, et al. Component-resolved analysis of IgA, IgE, and IgG4 during egg OIT identifies markers associated with sustained unresponsiveness. Allergy Eur J Allergy Clin Immunol. 2016. https://doi.org/10.1111/all.12895.

  38. Sicherer SH, Wood RA, Vickery BP, et al. The natural history of egg allergy in an observational cohort. J Allergy Clin Immunol. 2014. https://doi.org/10.1016/j.jaci.2013.12.1041.

  39. Peters RL, Dharmage SC, Gurrin LC, et al. The natural history and clinical predictors of egg allergy in the first 2 years of life: A prospective, population-based cohort study. J Allergy Clin Immunol. 2014. https://doi.org/10.1016/j.jaci.2013.11.032.

  40. Dang TD, Peters RL, Allen KJ. Debates in allergy medicine: Baked egg and milk do not accelerate tolerance to egg and milk Debates in Allergy Medicine. World Allergy Organ J. 2016. https://doi.org/10.1186/s40413-015-0090-z.

  41. Dang TD, Peters RL, Koplin JJ, Dharmage SC, Gurrin LC, Ponsonby AL, et al. Egg allergen specific IgE diversity predicts resolution of egg allergy in the population cohort HealthNuts. Allergy Eur J Allergy Clin Immunol. 2019. https://doi.org/10.1111/all.13572.

  42. Beyer K, Ellman-Grunther L, Järvinen KM, Wood RA, Hourihane J, Sampson HA. Measurement of peptide-specific IgE as an additional tool in identifying patients with clinical reactivity to peanuts. J Allergy Clin Immunol. 2003. https://doi.org/10.1067/mai.2003.1621.

  43. Ayuso R, Sánchez-Garcia S, Pascal M, Lin J, Grishina G, Fu Z, et al. Is epitope recognition of shrimp allergens useful to predict clinical reactivity? Clin Exp Allergy. 2012. https://doi.org/10.1111/j.1365-2222.2011.03920.x.

  44. Wang J, Lin J, Bardina L, Goldis M, Nowak-Wegrzyn A, Shreffler WG, et al. Correlation of IgE/IgG4 milk epitopes and affinity of milk-specific IgE antibodies with different phenotypes of clinical milk allergy. J Allergy Clin Immunol. 2010. https://doi.org/10.1016/j.jaci.2009.12.017.

  45. Hamilton RG. Clinical laboratory assessment of immediate-type hypersensitivity. J Allergy Clin Immunol. 2010. https://doi.org/10.1016/j.jaci.2009.09.055.

  46. 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. https://doi.org/10.1016/S0140-6736(13)62301-6.

  47. Koppelman S, Peillon A, Sampson H, Martin L. Epicutaneous immunotherapy (EPIT) for peanut allergy modifies IgG4 responses to major peanut allergens. Allergy Eur J Allergy Clin Immunol. 2017. https://doi.org/10.1111/all.13252.

  48. Jones SM, Pons L, Roberts JL, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol. 2009. https://doi.org/10.1016/j.jaci.2009.05.022.

  49. Vickery BP, Lin J, Kulis M, et al. Peanut oral immunotherapy modifies IgE and IgG4 responses to major peanut allergens. J Allergy Clin Immunol. 2013. https://doi.org/10.1016/j.jaci.2012.10.048.

  50. Burk CM, Kulis M, Leung N, Kim EH, Burks AW, Vickery BP. Utility of component analyses in subjects undergoing sublingual immunotherapy for peanut allergy. Clin Exp Allergy. 2016. https://doi.org/10.1111/cea.12635.

  51. Suárez-Fariñas M, Suprun M, Chang HL, Gimenez G, Grishina G, Getts R, et al. Predicting development of sustained unresponsiveness to milk oral immunotherapy using epitope-specific antibody binding profiles. J Allergy Clin Immunol. 2019. https://doi.org/10.1016/j.jaci.2018.10.028.

  52. Gupta RS, Lau CH, Hamilton RG, Donnell A, Newhall KK. Predicting Outcomes of Oral Food Challenges by Using the Allergen-Specific IgE-Total IgE Ratio. J Allergy Clin Immunol Pract. 2014. https://doi.org/10.1016/j.jaip.2013.12.006.

  53. Shamji MH, Layhadi JA, Scadding GW, Cheung DKM, Calderon MA, Turka LA, et al. Basophil expression of diamine oxidase: A novel biomarker of allergen immunotherapy response. J Allergy Clin Immunol. 2015. https://doi.org/10.1016/j.jaci.2014.09.049.

  54. Shamji MH, Ljørring C, Francis JN, et al. Functional rather than immunoreactive levels of IgG4 correlate closely with clinical response to grass pollen immunotherapy. Allergy Eur J Allergy Clin Immunol. 2012. https://doi.org/10.1111/j.1398-9995.2011.02745.x.

  55. •• Orgel K, Burk C, Smeekens J, Suber J, Hardy L, Guo R, et al. Blocking antibodies induced by peanut oral and sublingual immunotherapy suppress basophil activation and are associated with sustained unresponsiveness. Clin Exp Allergy. 2018. https://doi.org/10.1111/cea.13305. BATs were used to assess functional changes in IgG blocking ability from desensitized and SU subjects undergoing OIT and SLIT.

  56. Burton OT, Logsdon SL, Zhou JS, et al. Oral immunotherapy induces IgG antibodies that act through FcγRIIb to suppress IgE-mediated hypersensitivity. J Allergy Clin Immunol. 2014. https://doi.org/10.1016/j.jaci.2014.05.042.

  57. Santos AF, James LK, Bahnson HT, et al. IgG4 inhibits peanut-induced basophil and mast cell activation in peanut-tolerant children sensitized to peanut major allergens. J Allergy Clin Immunol. 2015. https://doi.org/10.1016/j.jaci.2015.01.012.

  58. Patil SU, Ogunniyi AO, Calatroni A, Tadigotla VR, Ruiter B, Ma A, et al. Peanut oral immunotherapy transiently expands circulating Ara h 2-specific B cells with a homologous repertoire in unrelated subjects. J Allergy Clin Immunol. 2015. https://doi.org/10.1016/j.jaci.2015.03.026.

  59. 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. https://doi.org/10.1016/j.jaci.2011.11.045.

  60. Flinterman AE, Pasmans SGMA, Den Hartog Jager CF, Hoekstra MO, Bruijnzeel-Koomen CAFM, Knol EF, et al. T cell responses to major peanut allergens in children with and without peanut allergy. Clin Exp Allergy. 2010. https://doi.org/10.1111/j.1365-2222.2009.03431.x.

  61. Dang TD, Allen KJ, Martino JD, Koplin JJ, Licciardi PV, MLK T. Food-allergic infants have impaired regulatory T-cell responses following in vivo allergen exposure. Pediatr Allergy Immunol. 2016. https://doi.org/10.1111/pai.12498.

  62. Berin MC, Shreffler WG. Mechanisms Underlying Induction of Tolerance to Foods. Immunol Allergy Clin North Am. 2016. https://doi.org/10.1016/j.iac.2015.08.002.

  63. Wambre E, James EA, Kwok WW. Characterization of CD4+ T cell subsets in allergy. Curr Opin Immunol. 2012. https://doi.org/10.1016/j.coi.2012.07.009.

  64. Kulis M, Yue X, Guo R, et al. High- and low-dose oral immunotherapy similarly suppress pro-allergic cytokines and basophil activation in young children. Clin Exp Allergy. 2019. https://doi.org/10.1111/cea.13256.

  65. Wisniewski JA, Commins SP, Agrawal R, et al. Analysis of cytokine production by peanut-reactive T cells identifies residual Th2 effectors in highly allergic children who received peanut oral immunotherapy. Clin Exp Allergy. 2015. https://doi.org/10.1111/cea.12537.

  66. Gorelik M, Narisety SD, Guerrerio AL, Chichester KL, Keet CA, Bieneman AP, et al. Suppression of the immunologic response to peanut during immunotherapy is often transient. J Allergy Clin Immunol. 2015. https://doi.org/10.1016/j.jaci.2014.11.010.

  67. Syed A, Garcia MA, Lyu SC, 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. https://doi.org/10.1016/j.jaci.2013.12.1037.

  68. • Wambre E, Bajzik V, DeLong JH, et al (2017) A phenotypically and functionally distinct human TH2 cell subpopulation is associated with allergic disorders. Sci Transl Med. https://doi.org/10.1126/scitranslmed.aam9171. This was the first study to identify a Th2a subset that is associated with food allergic subjects.

  69. Wambre E, Delong JH, James EA, Lafond RE, Robinson D, Kwok WW. Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy. J Allergy Clin Immunol. 2012. https://doi.org/10.1016/j.jaci.2011.08.034.

  70. Dhondalay GK, Rael E, Acharya S, et al. Food allergy and omics. J Allergy Clin Immunol. 2018. https://doi.org/10.1016/j.jaci.2017.11.007.

  71. Watson CT, Cohain AT, Griffin RS, et al. Integrative transcriptomic analysis reveals key drivers of acute peanut allergic reactions. Nat Commun. 2017. https://doi.org/10.1038/s41467-017-02188-7.

  72. Crestani E, Leirer J, Motsinger-Reif A, Phipatanakul W, Kaddurah-Daouk R, Chatila TA. Untargeted Metabolomic Profiling Identifies Disease-Specific Pathways in Food Allergy and Asthma. J Allergy Clin Immunol. 2019. https://doi.org/10.1016/j.jaci.2018.12.778.

  73. Kong J, Chalcraft K, Mandur TS, et al. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy Eur J Allergy Clin Immunol. 2015. https://doi.org/10.1111/all.12579.

  74. Noval Rivas M, Burton OT, Wise P, et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. 2013. https://doi.org/10.1016/j.jaci.2012.10.026.

  75. Rodriguez B, Prioult G, Hacini-Rachinel F, et al. Infant gut microbiota is protective against cow’s milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol. 2012. https://doi.org/10.1111/j.1574-6941.2011.01207.x.

  76. Feehley T, Plunkett CH, Bao R, et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat Med. 2019. https://doi.org/10.1038/s41591-018-0324-z.

  77. Santos AF, Couto-Francisco N, Bécares N, Kwok M, Bahnson HT, Lack G. A novel human mast cell activation test for peanut allergy. J Allergy Clin Immunol. 2018. https://doi.org/10.1016/j.jaci.2018.03.011.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to LaKeya C. Hardy.

Ethics declarations

Conflict of Interest

The authors declare 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 Food Allergy

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hardy, L.C., Smeekens, J.M. & Kulis, M.D. Biomarkers in Food Allergy Immunotherapy. Curr Allergy Asthma Rep 19, 61 (2019). https://doi.org/10.1007/s11882-019-0894-y

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11882-019-0894-y

Keywords

Navigation