We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Food allergy diagnosis and therapy: where are we now?

    Aleena Syed

    Department of Pediatrics, Division of Allergy, Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA

    Authors contributed equally

    Search for more papers by this author

    ,
    Arunima Kohli

    Department of Pediatrics, Division of Allergy, Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA

    Authors contributed equally

    Search for more papers by this author

    &
    Kari C Nadeau

    * Author for correspondence

    Department of Pediatrics, Division of Allergy, Immunology & Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA.

    Published Online:https://doi.org/10.2217/imt.13.93

    Food allergy is a growing worldwide epidemic that adversely effects up to 10% of the population. Causes and risk factors remain unclear and diagnostic methods are imprecise. There is currently no accepted treatment for food allergy. Therefore, there is an imminent need for greater understanding of food allergies, revised diagnostics and development of safe, effective therapies. Oral immunotherapy provides a particularly promising avenue, but is still highly experimental and not ready for clinical use.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Burks AW, Tang M, Sicherer S et al. ICON: food allergy. J. Allergy Clin. Immunol.129(4),906–920 (2012).
    • Vickery BP, Chin S, Burks AW. Pathophysiology of food allergy. Pediatr. Clin. North Am.58(2),363–376 (2011).
    • Chin S, Vickery BP. Pathogenesis of food allergy in the pediatric patient. Curr. Allergy Asthma Rep.12(6),621–629 (2012).
    • Venter C, Hasan Arshad S, Grundy J et al. Time trends in the prevalence of peanut allergy: three cohorts of children from the same geographical location in the UK. Allergy65(1),103–108 (2010).
    • Osborne NJ, Koplin JJ, Martin PE et al. Prevalence of challenge-proven IgE-mediated food allergy using population-based sampling and predetermined challenge criteria in infants. J. Allergy Clin. Immunol.127(3),668–676.e1–e2 (2011).
    • Hu Y, Chen J, Li H. Comparison of food allergy prevalence among Chinese infants in Chongqing, 2009 versus 1999. Pediatr. Int.52(5),820–824 (2010).
    • Sicherer SH, Sampson HA. Food allergy. J. Allergy Clin. Immunol.125(2 Suppl. 2),S116–S125 (2010).
    • Fleischer DM, Perry TT, Atkins D et al. Allergic reactions to foods in preschool-aged children in a prospective observational food allergy study. Pediatrics130(1),e25–e32 (2012).
    • Gupta RS, Springston EE, Smith B et al. Food allergy knowledge, attitudes, and beliefs of parents with food-allergic children in the United States. Pediatr. Allergy Immunol.21(6),927–934 (2010).
    • 10  Cummings AJ, Knibb RC, King RM, Lucas JS. The psychosocial impact of food allergy and food hypersensitivity in children, adolescents and their families: a review. Allergy65(8),933–945 (2010).
    • 11  Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat. Rev. Immunol.8(3),205–217 (2008).
    • 12  Joseph CL, Ownby DR, Havstad SL et al. Early complementary feeding and risk of food sensitization in a birth cohort. J. Allergy Clin. Immunol.127(5),1203.e5–1210.e5 (2011).
    • 13  Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat. Rev. Immunol.8(6),435–446 (2008).
    • 14  Lin J, Sampson HA. The role of immunoglobulin E-binding epitopes in the characterization of food allergy. Curr. Opin. Allergy Clin. Immunol.9(4),357–363 (2009).
    • 15  Ventura MT, Polimeno L, Amoruso AC et al. Intestinal permeability in patients with adverse reactions to food. Dig. Liver Dis.38(10),732–736 (2006).
    • 16  Levy Y, Davidovits M, Cleper R, Shapiro R. New-onset post-transplantation food allergy in children – is it attributable only to the immunosuppressive protocol? Pediatr. Transplant.13(1),63–69 (2009).
    • 17  Lack G. Update on risk factors for food allergy. J. Allergy Clin. Immunol.129(5),1187–1197 (2012).
    • 18  Shreiner A, Huffnagle GB, Noverr MC. The “microflora hypothesis” of allergic disease. Adv. Exp. Med. Biol.635,113–134 (2008).
    • 19  Jedrychowski W, Perera F, Maugeri U et al. Wheezing and asthma may be enhanced by broad spectrum antibiotics used in early childhood. Concept and results of a pharmacoepidemiology study. J. Physiol. Pharmacol.62(2),189–195 (2011).
    • 20  Sandini U, Kukkonen AK, Poussa T, Sandini L, Savilahti E, Kuitunen M. Protective and risk factors for allergic diseases in high-risk children at the ages of two and five years. Int. Arch. Allergy Immunol.156(3),339–348 (2011).
    • 21  Lewis MC, Inman CF, Patel D et al. Direct experimental evidence that early-life farm environment influences regulation of immune responses. Pediatr. Allergy Immunol.23(3),265–269 (2012).
    • 22  Schaub B, Liu J, Hoppler S et al. Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J. Allergy Clin. Immunol.123(4),774–782 e775 (2009).
    • 23  Azad MB, Konya T, Maughan H et al. Infant gut microbiota and the hygiene hypothesis of allergic disease: impact of household pets and siblings on microbiota composition and diversity. Allergy Asthma Clin. Immunol.9(1),15 (2013) (Epub ahead of print).
    • 24  Sjögren YM, Jenmalm MC, Böttcher MF, Björkstén B, Sverremark-Ekström E. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin. Exp. Allergy39(4),518–526 (2009).
    • 25  Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low diversity of the gut microbiota in infants with atopic eczema. J. Allergy Clin. Immunol.129(2),434–440, 440.e1–440.e2 (2012).
    • 26  Gupta RS, Springston EE, Warrier MR et al. The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics128(1),e9–e17 (2011).▪ Provides extensive detail on prevalence and demographics of pediatric food allergy in the USA in a large and diverse cohort.
    • 27  Hong X, Tsai HJ, Wang X. Genetics of food allergy. Curr. Opin. Pediatr.21(6),770–776 (2009).
    • 28  Hourihane JO, Dean TP, Warner JO. Peanut allergy in relation to heredity, maternal diet, and other atopic diseases: results of a questionnaire survey, skin prick testing, and food challenges. BMJ313(7056),518–521 (1996).
    • 29  Tsai HJ, Kumar R, Pongracic J et al. Familial aggregation of food allergy and sensitization to food allergens: a family-based study. Clin. Exp. Allergy39(1),101–109 (2009).
    • 30  Sicherer SH, Furlong TJ, Maes HH, Desnick RJ, Sampson HA, Gelb BD. Genetics of peanut allergy: a twin study. J. Allergy Clin. Immunol.106(1 Pt 1),53–56 (2000).
    • 31  Kumar R, Tsai HJ, Hong X et al. Race, ancestry, and development of food-allergen sensitization in early childhood. Pediatrics128(4),e821–e829 (2011).
    • 32  Jarvinen KM, Fleischer DM. Can we prevent food allergy by manipulating the timing of food exposure? Immunol. Allergy Clin. North Am.32(1),51–65 (2012).
    • 33  Boyce JA, Assa’ad A, Burks AW et al. Guidelines for the Diagnosis and Management of Food Allergy in the United States: Summary of the NIAID-Sponsored Expert Panel Report. J. Allergy Clin. Immunol.126(6),1105–1118 (2010).
    • 34  Holt PG. Prenatal versus postnatal priming of allergen specific immunologic memory: the debate continues. J. Allergy Clin. Immunol.122(4),717–718 (2008).
    • 35  Koplin JJ, Osborne NJ, Wake M et al. Can early introduction of egg prevent egg allergy in infants?. A population-based study. J. Allergy Clin. Immunol.126(4),807–813 (2010).
    • 36  Katz Y, Rajuan N, Goldberg MR et al. Early exposure to cow’s milk protein is protective against IgE-mediated cow’s milk protein allergy. J. Allergy Clin. Immunol.126(1),77.e1–82.e1 (2010).
    • 37  Nwaru BI, Erkkola M, Ahonen S et al. Age at the introduction of solid foods during the first year and allergic sensitization at age 5 years. Pediatrics125(1),50–59 (2010).
    • 38  Du Toit G, Katz Y, Sasieni P et al. Early consumption of peanuts in infancy is associated with a low prevalence of peanut allergy. J. Allergy Clin. Immunol.122(5),984–991 (2008).▪ One of the first studies to assess the effect of early consumption of allergens in relation to prevalence; well-controlled and provides evidence that early consumption of peanuts may be protective against food allergies.
    • 39  Szeles L, Keresztes G, Torocsik D et al. 1,25-dihydroxyvitamin D3 is an autonomous regulator of the transcriptional changes leading to a tolerogenic dendritic cell phenotype. J. Immunol.182(4),2074–2083 (2009).
    • 40  Dimeloe S, Nanzer A, Ryanna K, Hawrylowicz C. Regulatory T cells, inflammation and the allergic response – the role of glucocorticoids and Vitamin D. J. Steriod Biochem. Mol. Biol.120(2–3),86–95 (2010).
    • 41  Unger WW, Laban S, Kleijwegt FS, van der Slik AR, Roep BO. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1. Eur. J. Immunol.39(11),3147–3159 (2009).
    • 42  Heine G, Niesner U, Chang HD et al. 1,25-dihydroxyvitamin D(3) promotes IL-10 production in human B cells. Eur. J. Immunol.38(8),2210–2218 (2008).
    • 43  Camargo CA Jr, Clark S, Kaplan MS, Lieberman P, Wood RA. Regional differences in EpiPen prescriptions in the United States: the potential role of vitamin D. J. Allergy Clin. Immunol.120(1),131–136 (2007).
    • 44  Mullins RJ, Clark S, Camargo CA Jr. Regional variation in epinephrine autoinjector prescriptions in Australia: more evidence for the vitamin D-anaphylaxis hypothesis. Ann. Allergy Asthma Immunol.103(6),488–495 (2009).
    • 45  Mullins RJ, Clark S, Camargo CA Jr. Regional variation in infant hypoallergenic formula prescriptions in Australia. Pediatr. Allergy Immunol.21(2 Pt 2),e413–e420 (2010).
    • 46  Wjst M, Dharmage S, Andre E et al. Latitude, birth date, and allergy. PLoS Med.2(10),e294 (2005).
    • 47  Rudders SA, Espinola JA, Camargo CA Jr. North-south differences in US emergency department visits for acute allergic reactions. Ann. Allergy Asthma Immunol.104(5),413–416 (2010).
    • 48  Keet CA, Matsui EC, Savage JH et al. Potential mechanisms for the association between fall birth and food allergy. Allergy67(6),775–782 (2012).
    • 49  Wjst M, Hyppönen E. Vitamin D serum levels and allergic rhinitis. Allergy62(9),1085–1086 (2007).
    • 50  Sharief S, Jariwala S, Kumar J, Muntner P, Melamed ML. Vitamin D levels and food and environmental allergies in the United States: results from the National Health and Nutrition Examination Survey 2005–2006. J. Allergy Clin. Immunol.127(5),1195–1202 (2011).
    • 51  Gale CR, Robinson SM, Harvey NC et al. Maternal vitamin D status during pregnancy and child outcomes. Eur. J. Clin. Nutr.62(1),68–77 (2008).
    • 52  Hong X, Wang X. Early life precursors, epigenetics, and the development of food allergy. Semin. Immunopathol.34(5),655–669 (2012).
    • 53  Turcanu V, Maleki SJ, Lack G. Characterization of lymphocyte responses to peanuts in normal children, peanut-allergic children, and allergic children who acquired tolerance to peanuts. J. Clin. Invest.111(7),1065–1072 (2003).
    • 54  Thottingal TB, Stefura BP, Simons FE et al. Human subjects without peanut allergy demonstrate T cell-dependent, Th2-biased, peanut-specific cytokine and chemokine responses independent of Th1 expression. J. Allergy Clin. Immunol.118(4),905–914 (2006).
    • 55  Begin PN, Kari C. Diagnosis of food allergy. Pediatr. Ann.42(6),102–109 (2013).
    • 56  Caubet JC, Sampson HA. Beyond skin testing: state of the art and new horizons in food allergy diagnostic testing. Immunol. Allergy Clin. North Am.32(1),97–109 (2012).▪▪ Comprehensive analysis of available diagnostic techniques; also provides commentary on future diagnostic tools in development.
    • 57  Nowak-Wegrzyn A, Assa’ad AH, Bahna SL et al. Work Group report: oral food challenge testing. J. Allergy Clin. Immunol.123(Suppl. 6),S365–S383 (2009).
    • 58  Wood RA, Sicherer SH, Vickery BP et al. The natural history of milk allergy in an observational cohort. J. Allergy Clin. Immunol.131(3),805–812 (2013).
    • 59  Sampson HA, Gerth van Wijk R, Bindslev-Jensen C et al. Standardizing double-blind, placebo-controlled oral food challenges: American Academy of Allergy, Asthma and Immunology–European Academy of Allergy and Clinical Immunology PRACTALL consensus report. J. Allergy Clin. Immunol.130(6),1260–1274 (2012).
    • 60  Calvani M, Alessandri C, Frediani T et al. Correlation between skin prick test using commercial extract of cow’s milk protein and fresh milk and food challenges. Pediatr. Allergy Immunol.18(7),583–588 (2007).
    • 61  Tripodi S, Businco AD, Alessandri C, Panetta V, Restani P, Matricardi PM. Predicting the outcome of oral food challenges with hen’s egg through skin test end-point titration. Clin. Exp. Allergy39(8),1225–1233 (2009).
    • 62  Peters RL, Gurrin LC, Allen KJ. The predictive value of skin prick testing for challenge-proven food allergy: a systematic review. Pediatr. Allergy Immunol.23(4),347–352 (2012).
    • 63  Turcanu V, Winterbotham M, Kelleher P, Lack G. Peanut-specific B and T cell responses are correlated in peanut-allergic but not in nonallergic individuals. Clin. Exp. Allergy38(7),1132–1139 (2008).
    • 64  Du Toit G, Roberts G, Sayre PH et al. Identifying infants at high risk of peanut allergy: the Learning Early About Peanut Allergy (LEAP) screening study. J. Allergy Clin. Immunol.131(1),135–143.e1–e12 (2013).
    • 65  Ford LS, Bloom KA, Nowak-Wegrzyn AH, Shreffler WG, Masilamani M, Sampson HA. Basophil reactivity, wheal size, and immunoglobulin levels distinguish degrees of cow’s milk tolerance. J. Allergy Clin. Immunol.131(1),180–186.e1–e3 (2013).
    • 66  Mehl A, Niggemann B, Keil T, Wahn U, Beyer K. Skin prick test and specific serum IgE in the diagnostic evaluation of suspected cow’s milk and hen’s egg allergy in children: does one replace the other? Clin. Exp. Allergy42(8),1266–1272 (2012).
    • 67  Siles RI, Hsieh FH. Allergy blood testing: a practical guide for clinicians. Cleve. Clin. J. Med.78(9),585–592 (2011).
    • 68  Wang J, Lin J, Bardina L 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.125(3),695–702, 702.e1–702.e6 (2010).
    • 69  Asarnoj A, Nilsson C, Lidholm J et al. Peanut component Ara h 8 sensitization and tolerance to peanut. J. Allergy Clin. Immunol.130(2),468–472 (2012).
    • 70  Krause S, Reese G, Randow S et al. Lipid transfer protein (Ara h 9) as a new peanut allergen relevant for a Mediterranean allergic population. J. Allergy Clin. Immunol.124(4),771. e5–778.e5 (2009).
    • 71  Sanz ML, Blázquez AB, Garcia BE. Microarray of allergenic component-based diagnosis in food allergy. Curr. Opin. Allergy Clin. Immunol.11(3),204–209 (2011).
    • 72  Ito Y, Moritsugu N, Matsue T et al. An automated multiplex specific IgE assay system using a photoimmobilized microarray. J. Biotechnol.161(4),414–421 (2012).
    • 73  Ott H, Baron JM, Heise R et al. Clinical usefulness of microarray-based IgE detection in children with suspected food allergy. Allergy63(11),1521–1528 (2008).
    • 74  Chan SM, Dumitru C, Turcanu V. Molecular diagnosis of peanut allergy. Expert Rev. Mol. Diagn.12(8),879–891 (2012).
    • 75  Kulis M, Saba K, Kim EH et al. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut sublingual immunotherapy. J. Allergy Clin. Immunol.129(4),1159–1162 (2012).
    • 76  Battaglia M, Gregori S, Bacchetta R, Roncarolo MG. Tr1 cells: from discovery to their clinical application. Semin. Immunol.18(2),120–127 (2006).
    • 77  Gernez Y, Tirouvanziam R, Yu G et al. Basophil CD203c levels are increased at baseline and can be used to monitor omalizumab treatment in subjects with nut allergy. Int. Arch. Allergy Immunol.154(4),318–327 (2011).
    • 78  Wanich N, Nowak-Wegrzyn A, Sampson HA, Shreffler WG. Allergen-specific basophil suppression associated with clinical tolerance in patients with milk allergy. J. Allergy Clin. Immunol.123(4),789. e20–794.e20 (2009).
    • 79  Bedoret D, Singh AK, Shaw V et al. Changes in antigen-specific T-cell number and function during oral desensitization in cow’s milk allergy enabled with omalizumab. Mucosal Immunol.5(3),267–276 (2012).
    • 80  Li XM, Zhang TF, Huang CK et al. Food Allergy Herbal Formula-1 (FAHF-1) blocks peanut-induced anaphylaxis in a murine model. J. Allergy Clin. Immunol.108(4),639–646 (2001).
    • 81  Srivastava KD, Kattan JD, Zou ZM et al. The Chinese herbal medicine formula FAHF-2 completely blocks anaphylactic reactions in a murine model of peanut allergy. J. Allergy Clin. Immunol.115(1),171–178 (2005).
    • 82  Srivastava KD, Bardina L, Sampson HA, Li XM. Efficacy and immunological actions of FAHF-2 in a murine model of multiple food allergies. Ann. Allergy Asthma Immunol.108(5),351.e1–358.e1 (2012).
    • 83  Wang J, Patil SP, Yang N et al. Safety, tolerability, and immunologic effects of a food allergy herbal formula in food allergic individuals: a randomized, double-blinded, placebo-controlled, dose escalation, Phase 1 study. Ann. Allergy Asthma Immunol.105(1),75–84 (2010).
    • 84  Patil SP, Wang J, Song Y et al. Clinical safety of Food Allergy Herbal Formula-2 (FAHF-2) and inhibitory effect on basophils from patients with food allergy: extended Phase I study. J. Allergy Clin. Immunol.128(6),1259. e2–1265.e2 (2011).
    • 85  Nadeau KC, Kohli A, Iyengar S, Dekruyff RH, Umetsu DT. Oral immunotherapy and anti-IgE antibody-adjunctive treatment for food allergy. Immunol. Allergy Clin. North Am.32(1),111–133 (2012).▪▪ Extensive review of oral immunotherapy for food allergy to date. Includes detailed summary tables of protocols and mechanistic diagrams.
    • 86  Sampson HA, Leung DY, Burks AW et al. A Phase II, randomized, doubleblind, parallel group, placebo controlled oral food challenge trial of Xolair (omalizumab) in peanut allergy. J. Allergy Clin. Immunol.127(5),1309. e1–1310.e1 (2011).
    • 87  Nadeau KC, Schneider LC, Hoyte L, Borras I, Umetsu DT. Rapid oral desensitization in combination with omalizumab therapy in patients with cow’s milk allergy. J. Allergy Clin. Immunol.127(6),1622–1624 (2011).
    • 88  Frossard CP, Steidler L, Eigenmann PA. Oral administration of an IL-10-secreting Lactococcus lactis strain prevents food-induced IgE sensitization. J. Allergy Clin. Immunol.119(4),952–959 (2007).
    • 89  Cortes-Perez NG, Ah-Leung S, Bermúdez-Humarán LG et al. Intranasal coadministration of live lactococci producing interleukin-12 and a major cow’s milk allergen inhibits allergic reaction in mice. Clin. Vaccine Immunol.14(3),226–233 (2007).
    • 90  Yao TC, Chang CJ, Hsu YH, Huang JL. Probiotics for allergic diseases: realities and myths. Pediatr. Allergy Immunol.21(6),900–919 (2010).
    • 91  Schnoeller C, Rausch S, Pillai S et al. A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10-producing macrophages. J. Immunol.180(6),4265–4272 (2008).
    • 92  Garud S, Peppercorn MA. Ulcerative colitis: current treatment strategies and future prospects. Therap. Adv. Gastroenterol.2(2),99–108 (2009).
    • 93  Reddy A, Fried B. An update on the use of helminths to treat Crohn’s and other autoimmunune diseases. Parasitol. Res.104(2),217–221 (2009).
    • 94  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.120(3),631–637 (2007).
    • 95  Kulis M, Burks WA. Oral immunotherapy for food allergy: clinical and preclinical studies. Adv. Drug Deliv. Rev.65(6),774–781 (2012).▪ Provides a thorough, yet concise overview of oral immunotherapies for food allergy to date, while also providing commentary on potential mechanisms of disease and treatment.
    • 96  Kim EH, Burks W. Managing food allergy in childhood. Curr. Opin. Pediatr.24(5),615–620 (2012).
    • 97  Sikora JM, Tankersley MS; ACAAI Immunotherapy and Diagnostics Committee. Perception and practice of sublingual immunotherapy among practicing allergists in the United States: a follow-up survey. Ann. Allergy Asthma Immunol.110(3),194.e4–197.e4 (2013).
    • 98  Enrique E, Pineda F, Malek T et al. Sublingual immunotherapy for hazelnut food allergy: a randomized, double-blind, placebo-controlled study with a standardized hazelnut extract. J. Allergy Clin. Immunol.116(5),1073–1079 (2005).
    • 99  de Boissieu D, Dupont C. Sublingual immunotherapy for cow’s milk protein allergy: a preliminary report. Allergy61(10),1238–1239 (2006).
    • 100  Fernández-Rivas M, Garrido Fernández S, Nadal JA et al. Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract. Allergy64(6),876–883 (2009).
    • 101  Kim EH, Bird JA, Kulis M et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J. Allergy Clin. Immunol.127(3),640.e1–646.e1 (2011).
    • 102  Narisety SD, Keet CA. Sublingual vs oral immunotherapy for food allergy: identifying the right approach. Drugs72(15),1977–1989 (2012).
    • 103  García BE, González-Mancebo E, Barber D et al. Sublingual immunotherapy in peach allergy: monitoring molecular sensitizations and reactivity to apple fruit and Platanus pollen. J. Investig. Allergol. Clin. Immunol.20(6),514–520 (2010).
    • 104  Hofmann AM, Scurlock AM, Jones SM et al. Safety of a peanut oral immunotherapy protocol in children with peanut allergy. J. Allergy Clin. Immunol.124(2),286–291, 291. e1–e6 (2009).
    • 105  Beyer K, Eckermann O, Hompes S, Grabenhenrich L, Worm M. Anaphylaxis in an emergency setting – elicitors, therapy and incidence of severe allergic reactions. Allergy67(11),1451–1456 (2012).
    • 106  Burks AW, Jones SM, Wood RA et al. Oral immunotherapy for treatment of egg allergy in children. N. Engl. J. Med.367(3),233–243 (2012).
    • 107  Vázquez-Ortiz M, Alvaro-Lozano M, Alsina L et al. Safety and predictors of adverse events during oral immunotherapy for milk allergy: severity of reaction at oral challenge, specific IgE and prick test. Clin. Exp. Allergy43(1),92–102 (2013).
    • 108  Jones SM, Pons L, Roberts JL et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J. Allergy Clin. Immunol.124(2),292–300, 300.e1–300.e97 (2009).
    • 109  Skripak JM, Nash SD, Rowley H et al. A randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow’s milk allergy. J. Allergy Clin. Immunol.122(6),1154–1160 (2008).
    • 110  Buchanan AD, Green TD, Jones SM et al. Egg oral immunotherapy in nonanaphylactic children with egg allergy. J. Allergy Clin. Immunol.119(1),199–205 (2007).
    • 111  Longo G, Barbi E, Berti I et al. Specific oral tolerance induction in children with very severe cow’s milk-induced reactions. J. Allergy Clin. Immunol.121(2),343–347 (2008).
    • 112  Narisety SD, Skripak JM, Steele P et al. Open-label maintenance after milk oral immunotherapy for IgE-mediated cow’s milk allergy. J. Allergy Clin. Immunol.124(3),610–612 (2009).
    • 113  Blumchen K, Ulbricht H, Staden U et al. Oral peanut immunotherapy in children with peanut anaphylaxis. J. Allergy Clin. Immunol.126(1),83–91 e1 (2010).
    • 114  Ridolo E, De Angelis GL, Dall’aglio P. Eosinophilic esophagitis after specific oral tolerance induction for egg protein. Ann. Allergy Asthma Immunol.106(1),73–74 (2011).
    • 115  Sánchez-García S, Rodríguez Del Río P, Escudero C, Martínez-Gómez MJ, Ibáñez MD. Possible eosinophilic esophagitis induced by milk oral immunotherapy. J. Allergy Clin. Immunol.129(4),1155–1157 (2012).
    • 116  Varshney P, Burks AW. Role of tolerance in the development of eosinophilic gastrointestinal diseases. Immunol. Allergy Clin. North Am.29(1),179–187 (2009).
    • 117  Thyagarajan A, Varshney P, Jones SM et al. Peanut oral immunotherapy is not ready for clinical use. J. Allergy Clin. Immunol.126(1),31–32 (2010).
    • 118  Beyer K, Wahn U. Oral immunotherapy for food allergy in children. Curr. Opin. Allergy Clin. Immunol.8(6),553–556 (2008).
    • 119  Sampson HA. Peanut oral immunotherapy: is it ready for clinical practice? J. Allergy Clin. Immunol.1(1),15–21 (2013).
    • 120  Keet C, Seopaul S, Knorr SD, Narisety S, Skripak JM, Wood RA. Long-term outcomes of milk oral immunotherapy in children. J. Allergy Clin. Immunol. doi:10.1016/j.jaci.2013.05.006 (2013) (Epub ahead of print).
    • 121  Leung DY, Sampson HA, Yunginger JW et al. Effect of anti-IgE therapy in patients with peanut allergy. N. Engl. J. Med.348(11),986–993 (2003).
    • 122  Savage JH, Courneya JP, Sterba PM, Macglashan DW, Saini SS, Wood RA. Kinetics of mast cell, basophil, and oral food challenge responses in omalizumab-treated adults with peanut allergy. J. Allergy Clin. Immunol.130(5),1123.e2–1129.e2 (2012).
    • 123  Burks AW, Vickery BP, Scurlock AM et al. Development of clinical tolerance after peanut OIT. J. Allergy Clin. Immunol.129(Suppl. 2),AB66 (2012).
    • 124  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.127(3),654–660 (2011).
    • 125  Palomares O, Yaman G, Azkur AK, Akkoc T, Akdis M, Akdis CA. Role of Treg in immune regulation of allergic diseases. Eur. J. Immunol.40(5),1232–1240 (2010).
    • 126  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. Allergy42(8),1197–1205 (2012).
    • 127  Pabst O, Mowat AM. Oral tolerance to food protein. Mucosal Immunol.5(3),232–239 (2012).
    • 128  Akdis M, Akdis CA. Therapeutic manipulation of immune tolerance in allergic disease. Nat. Rev. Drug. Discov.8(8),645–660 (2009).▪ Highly scientific review of potential mechanisms of allergy and immune tolerance and therapies. Comments on the association of studied therapies between mechanisms.
    • 129  Rachid R, Umetsu DT. Immunological mechanisms for desensitization and tolerance in food allergy. Semin. Immunopathol.34(5),689–702 (2012).
    • 130  Vickery BP, Scurlock AM, Jones SM, Burks AW. Mechanisms of immune tolerance relevant to food allergy. J. Allergy Clin. Immunol.127(3),576–584; quiz 585–576 (2011).
    • 131  Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Specific immunotherapy and turning off the T cell: how does it work? Ann. Allergy Asthma Immunol.107(5),381–392 (2011).
    • 132  Oppenheimer JJ, Nelson HS, Bock SA, Christensen F, Leung DY. Treatment of peanut allergy with rush immunotherapy. J. Allergy Clin. Immunol.90(2),256–262 (1992).
    • 133  Nelson HS, Lahr J, Rule R, Bock A, Leung D. Treatment of anaphylactic sensitivity to peanuts by immunotherapy with injections of aqueous peanut extract. J. Allergy Clin. Immunol.99(6 Pt 1),744–751 (1997).
    • 134  Katelaris CH. Food allergy and oral allergy or pollen-food syndrome. Curr. Opin. Allergy Clin. Immunol.10(3),246–251 (2010).
    • 135  Yang YH, Chiang BL. Novel approaches to food allergy. Clin. Rev. Allergy Immunol. doi:10.1007/s12016-013-8354-2 (2013) (Epub ahead of print).
    • 136  Järvinen KM, Beyer K, Vila L, Bardina L, Mishoe M, Sampson HA. Specificity of IgE antibodies to sequential epitopes of hen’s egg ovomucoid as a marker for persistence of egg allergy. Allergy62(7),758–765 (2007).
    • 137  Nowak-Wegrzyn A, Sampson HA. Future therapies for food allergies. J. Allergy Clin. Immunol.127(3),558–573; quiz 574–555 (2011).
    • 138  Nowak-Wegrzyn A, Bloom KA, Sicherer SH et al. Tolerance to extensively heated milk in children with cow’s milk allergy. J. Allergy Clin. Immunol.122(2),342–347, e1–e2 (2008).
    • 139  Lemon-Mule H, Sampson HA, Sicherer SH, Shreffler WG, Noone S, Nowak-Wegrzyn A. Immunologic changes in children with egg allergy ingesting extensively heated egg. J. Allergy Clin. Immunol.122(5),977.e1–983.e1 (2008).
    • 140  Kim JS, Nowak-Wegrzyn A, Sicherer SH, Noone S, Moshier EL, Sampson HA. Dietary baked milk accelerates the resolution of cow’s milk allergy in children. J. Allergy Clin. Immunol.128(1),125.e2–131.e2 (2011).
    • 141  Turner PJ, Campbell DE. What’s new in the diagnosis and management of food allergy in children? Asia Pac. Allergy3(2),88–95 (2013).
    • 142  Dupont C, Kalach N, Soulaines P, Legoue-Morillon S, Piloquet H, Benhamou PH. Cow’s milk epicutaneous immunotherapy in children: a pilot trial of safety, acceptability, and impact on allergic reactivity. J. Allergy Clin. Immunol.125(5),1165–1167 (2010).
    • 143  Mondoulet L, Dioszeghy V, Ligouis M, Dhelft V, Dupont C, Benhamou PH. Epicutaneous immunotherapy on intact skin using a new delivery system in a murine model of allergy. Clin. Exp. Allergy40(4),659–667 (2010).
    • 144  Mondoulet L, Dioszeghy V, Vanoirbeek JA, Nemery B, Dupont C, Benhamou PH. Epicutaneous immunotherapy using a new epicutaneous delivery system in mice sensitized to peanuts. Int. Arch. Allergy Immunol.154(4),299–309 (2011).
    • 145  Senti G, Prinz Vavricka BM, Erdmann I et al. Intralymphatic allergen administration renders specific immunotherapy faster and safer: a randomized controlled trial. Proc. Natl Acad. Sci. USA105(46),17908–17912 (2008).
    • 146  Cox L, Compalati E, Kundig T, Larche M. New directions in immunotherapy. Curr. Allergy Asthma Rep.13(2),178–195 (2013).
    • 147  Hylander T, Latif L, Petersson-Westin U, Cardell LO. Intralymphatic allergen-specific immunotherapy: an effective and safe alternative treatment route for pollen-induced allergic rhinitis. J. Allergy Clin. Immunol.131(2),412–420 (2013).
    • 148  Senti G, Crameri R, Kuster D et al. Intralymphatic immunotherapy for cat allergy induces tolerance after only 3 injections. J. Allergy Clin. Immunol.129(5),1290–1296 (2012).
    • 149  Martínez-Gómez JM, Johansen P, Erdmann I, Senti G, Crameri R, Kundig TM. Intralymphatic injections as a new administration route for allergen-specific immunotherapy. Int. Arch. Allergy Immunol.150(1),59–65 (2009).
    • 150  Senti G, Johansen P, Kundig TM. Intralymphatic immunotherapy: from the rationale to human applications. Curr. Top. Microbiol. Immunol.352,71–84 (2011).
    • 151  Moldaver D, Larche M. Immunotherapy with peptides. Allergy66(6),784–791 (2011).
    • 152  Lieberman JA, Nowak-Wegrzyn A. Vaccines and immunomodulatory therapies for food allergy. Curr. Allergy Asthma Rep.12(1),55–63 (2012).▪ Provides a thorough and comprehensive review of nearly all studied immunotherapies for food allergy to date, with information and speculation on the future.
    • 153  Yang M, Yang C, Mine Y. Multiple T cell epitope peptides suppress allergic responses in an egg allergy mouse model by the elicitation of forkhead box transcription factor 3- and transforming growth factor-beta-associated mechanisms. Clin. Exp. Allergy40(4),668–678 (2010).
    • 154  Yang M, Mine Y. Novel T-cell epitopes of ovalbumin in BALB/c mouse: potential for peptide-immunotherapy. Biochem. Biophys. Res. Comm.378(2),203–208 (2009).
    • 155  Prickett SR, Voskamp AL, Dacumos-Hill A, Symons K, Rolland JM, O’Hehir RE. Ara h 2 peptides containing dominant CD4+ T-cell epitopes: candidates for a peanut allergy therapeutic. J. Allergy Clin. Immunol.127(3),608–615.e1–e5 (2011).
    • 156  Delong JH, Simpson KH, Wambre E, James EA, Robinson D, Kwok WW. Ara h 1-reactive T cells in individuals with peanut allergy. J. Allergy Clin. Immunol.127(5),1211.e3–1218.e3 (2011).
    • 157  Sicherer SH, Sampson HA. Food allergy: recent advances in pathophysiology and treatment. Ann. Rev. Med.60,261–277 (2009).
    • 158  Valenta R, Linhart B, Swoboda I, Niederberger V. Recombinant allergens for allergen-specific immunotherapy: 10 years anniversary of immunotherapy with recombinant allergens. Allergy66(6),775–783 (2011).
    • 159  Bannon GA, Cockrell G, Connaughton C et al. Engineering, characterization and in vitro efficacy of the major peanut allergens for use in immunotherapy. Int. Arch. Allergy Immunol.124(1–3),70–72 (2001).
    • 160  Cocco RR, Jarvinen KM, Han N, Beyer K, Sampson HA. Mutational analysis of immunoglobulin E-binding epitopes of beta-casein and beta-lactoglobulin showed a heterogeneous pattern of critical amino acids between individual patients and pooled sera. Clin. Exp. Allergy37(6),831–838 (2007).
    • 161  Cocco RR, Jarvinen KM, Sampson HA, Beyer K. Mutational analysis of major, sequential IgE-binding epitopes in alpha s1-casein, a major cow’s milk allergen. J. Allergy Clin. Immunol.112(2),433–437 (2003).
    • 162  Swoboda I, Bugajska-Schretter A, Linhart B et al. A recombinant hypoallergenic parvalbumin mutant for immunotherapy of IgE-mediated fish allergy. J. Immunol.178(10),6290–6296 (2007).
    • 163  Toda M, Reese G, Gadermaier G et al. Protein unfolding strongly modulates the allergenicity and immunogenicity of Pru p 3, the major peach allergen. J. Allergy Clin. Immunol.128(5),1022–1030.e1–7 (2011).
    • 164  Bolhaar ST, Zuidmeer L, Ma Y et al. A mutant of the major apple allergen, Mal d 1, demonstrating hypo-allergenicity in the target organ by double-blind placebo-controlled food challenge. Clin. Exp. Allergy35(12),1638–1644 (2005).
    • 165  Rupa P, Mine Y. Engineered recombinant ovomucoid third domain can modulate allergenic response in Balb/c mice model. Biochem. Biophys. Res. Comm.342(3),710–717 (2006).
    • 166  Rupa P, Mine Y. Engineered recombinant ovomucoid third domain can desensitize Balb/c mice of egg allergy. Allergy61(7),836–842 (2006).
    • 167  Zhou Y, Kawasaki H, Hsu SC et al. Oral tolerance to food-induced systemic anaphylaxis mediated by the C-type lectin SIGNR1. Nat. Med.16(10),1128–1133 (2010).
    • 168  Pfaar O, Barth C, Jaschke C, Hormann K, Klimek L. Sublingual allergen-specific immunotherapy adjuvanted with monophosphoryl lipid A: a Phase I/IIa study. Int. Arch. Allergy Immunol.154(4),336–344 (2011).
    • 169  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.5(4),387–391 (1999).
    • 170  Li F, Wang L, Jin XM, Yan CH, Jiang S, Shen XM. The immunologic effect of TGF-beta1 chitosan nanoparticle plasmids on ovalbumin-induced allergic BALB/c mice. Immunobiology214(2),87–99 (2009).
    • 171  Li X, Huang CK, Schofield BH et al. Strain-dependent induction of allergic sensitization caused by peanut allergen DNA immunization in mice. J. Immunol.162(5),3045–3052 (1999).
    • 201  Therapeutic Effect of Chinese Herbal Medicine on Food Allergy (FAHF-2). http://clinicaltrials.gov/ct2/show/NCT00602160
    • 202  OIT and Xolair® (Omalizumab) in Cow’s Milk Allergy. http://clinicaltrials.gov/ct2/show/NCT01157117
    • 203  Xolair Treatment for Milk Allergic Children. http://clinicaltrials.gov/ct2/show/NCT00968110
    • 204  Xolair Enhances Oral Desensitization in Peanut Allergic Patients. http://clinicaltrials.gov/ct2/show/NCT01290913
    • 205  Guidelines for the evaluation of probiotics in food. www.who.int/foodsafety/publications/fs_management/probiotics2/en/index.html
    • 206  The Common Fund’s Human Microbiome Project. http://nihroadmap.nih.gov/hmp
    • 207  Trichuris Suis Ova in Peanut and Tree Nut Allergy. http://clinicaltrials.gov/ct2/show/NCT01070498
    • 208  Sublingual Immunotherapy for Peanut Allergy and Induction of Tolerance (SLIT2). http://clinicaltrials.gov/ct2/show/NCT01373242
    • 209  Double Blind Peanut Sublingual Immunotherapy (PN SLIT). http://clinicaltrials.gov/ct2/show/NCT00597727
    • 210  Immunotherapy for Peanut Allergy. http://clinicaltrials.gov/ct2/show/NCT00429429
    • 211  Safety of Epicutaneous Immunotherapy for the Treatment of Peanut Allergy. http://clinicaltrials.gov/ct2/show/NCT01170286
    • 212  Peanut Allergy Vaccine Study in Healthy and Peanut-allergic Adults. http://clinicaltrials.gov/ct2/show/NCT00850668