The safety and efficacy of sublingual and oral immunotherapy for milk allergy

Corinne A Keet, Pamela A Frischmeyer-Guerrerio, Ananth Thyagarajan, John T Schroeder, Robert G Hamilton, Stephen Boden, Pamela Steele, Sarah Driggers, A Wesley Burks, Robert A Wood, Corinne A Keet, Pamela A Frischmeyer-Guerrerio, Ananth Thyagarajan, John T Schroeder, Robert G Hamilton, Stephen Boden, Pamela Steele, Sarah Driggers, A Wesley Burks, Robert A Wood

Abstract

Background: Oral immunotherapy (OIT) and sublingual immunotherapy (SLIT) are potential therapies for food allergy, but the optimal method of administration, mechanism of action, and duration of response remain unknown.

Objective: We sought to explore the safety and efficacy of OIT and SLIT for the treatment of cow's milk (CM) allergy.

Methods: We randomized children with CM allergy to SLIT alone or SLIT followed by OIT. After screening double-blind, placebo-controlled food challenges and initial SLIT escalation, subjects either continued SLIT escalation to 7 mg daily or began OIT to either 1000 mg (the OITB group) or 2000 mg (the OITA group) of milk protein. They were challenged with 8 g of milk protein after 12 and 60 weeks of maintenance. If they passed the 60-week challenge, therapy was withdrawn, with challenges repeated 1 and 6 weeks later. Mechanistic correlates included end point titration skin prick testing and measurement of CM-specific IgE and IgG(4) levels, basophil histamine release, constitutive CD63 expression, CD203c expression, and intracellular spleen tyrosine kinase levels.

Results: Thirty subjects with CM allergy aged 6 to 17 years were enrolled. After therapy, 1 of 10 subjects in the SLIT group, 6 of 10 subjects in the SLIT/OITB group, and 8 of 10 subjects in the OITA group passed the 8-g challenge (P = .002, SLIT vs OIT). After avoidance, 6 of 15 subjects (3 of 6 subjects in the OITB group and 3 of 8 subjects in the OITA group) regained reactivity, 2 after only 1 week. Although the overall reaction rate was similar, systemic reactions were more common during OIT than during SLIT. By the end of therapy, titrated CM skin prick test results and CD63 and CD203c expression decreased and CM-specific IgG(4) levels increased in all groups, whereas CM-specific IgE and spontaneous histamine release values decreased in only the OIT group.

Conclusion: OIT was more efficacious for desensitization to CM than SLIT alone but was accompanied by more systemic side effects. Clinical desensitization was lost in some cases within 1 week off therapy.

Copyright © 2011 American Academy of Allergy, Asthma & Immunology. Published by Mosby, Inc. All rights reserved.

Figures

FIG 1
FIG 1
Study timeline with key features highlighted. Subjects were randomized at T2 to either SLIT (goal dose, 7 mg), OITB (goal dose, 1000 mg), or OITA (goal dose, 2000 mg). CM-specific IgE, CM-specific IgG4, and CM-titrated skin prick test (SPT) results were obtained in all subjects at T1, T3 to T5, and T7. Basophil studies were done at T1, T2 to T5, and T7.
FIG 2
FIG 2
Food challenge outcome. The CM protein threshold is shown for baseline (T1), after 12 weeks of maintenance (T4), after 60 weeks of maintenance (T5), and 1 week (T6) and 6 weeks (T7) off therapy. A, SLIT/SLIT group. B, SLIT/OITB group. C, SLIT/OITC groups. Bars represent medians. *P < .05 and **P < .01 compared with T1.
FIG 3
FIG 3
Mechanistic changes. Individual line plots showing changes with treatment for the 3 randomization groups (A, SLIT/SLIT group; B, SLIT/OITB group; and C, SLIT/OITA group) in CM-specific IgE level (1), CM-specific IgG4 levels (2), and end point titration skin prick test responses. Shown is the average wheal size over 5 CM dilution. Bars represent medians. The shaded blue area represents the period of withdrawal of therapy. *P < .05 and **P < .01 compared with T1.
FIG 4
FIG 4
Basophil activity. A, HR in media alone (SHR) or after stimulation with CM extract, goat anti-human IgE antibody (IgE), peanut extract (PN), or N-formylmethionine (f-met) at T1 to T5 and T7. B, Constitutive surface expression of CD63 and CD203c and intracellular Syk by basophils at T1 to T5 and T7. A box-and-whisker plot is shown; outliers are not shown. *P < .05 and **P < .01 compared with T1.

References

    1. 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. 1997;99:744–51.
    1. Allam JP, Stojanovski G, Friedrichs N, Peng W, Bieber T, Wenzel J, et al. Distribution of Langerhans cells and mast cells within the human oral mucosa: new application sites of allergens in sublingual immunotherapy? Allergy. 2008;63:720–7.
    1. Untersmayr E, Jensen-Jarolim E. The role of protein digestibility and antacids on food allergy outcomes. J Allergy Clin Immunol. 2008;121:1301–8.
    1. Enrique E, Pineda F, Malek T, Bartra J, Basagana M, Tella R, et al. Sublingual immunotherapy for hazelnut food allergy: a randomized, double-blind, placebo-controlled study with a standardized hazelnut extract. J Allergy Clin Immunol. 2005;116:1073–9.
    1. de Boissieu D, Dupont C. Sublingual immunotherapy for cow’s milk protein allergy: a preliminary report. Allergy. 2006;61:1238–9.
    1. Patriarca G, Nucera E, Pollastrini E, Roncallo C, De Pasquale T, Lombardo C, et al. Oral specific desensitization in food-allergic children. Dig Dis Sci. 2007;52:1662–72.
    1. 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;127:640–6. e1.
    1. Mempel M, Rakoski J, Ring J, Ollert M. Severe anaphylaxis to kiwi fruit: immunologic changes related to successful sublingual allergen immunotherapy. J Allergy Clin Immunol. 2003;111:1406–9.
    1. Fernandez-Rivas M, Fernandez S Garrido, Nadal JA, Garcia BE, Gonzalez-Mancebo E, et al. Randomized double-blind, placebo-controlled trial of sublingual immunotherapy with a Pru p 3 quantified peach extract. Allergy. 2009;64:876–83.
    1. 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;124:292–300. e1–97.
    1. Clark AT, Islam S, King Y, Deighton J, Anagnostou K, Ewan PW. Successful oral tolerance induction in severe peanut allergy. Allergy. 2009;64:1218–20.
    1. 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;127:654–60.
    1. Buchanan AD, Green TD, Jones SM, Scurlock AM, Christie L, Althage KA, et al. Egg oral immunotherapy in nonanaphylactic children with egg allergy. J Allergy Clin Immunol. 2007;119:199–205.
    1. 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;105:444–50.
    1. 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;122:1154–60.
    1. Pajno GB, Caminiti L, Ruggeri P, De Luca R, Vita D, La Rosa M, et al. Oral immunotherapy for cow’s milk allergy with a weekly up-dosing regimen: a randomized single-blind controlled study. Ann Allergy Asthma Immunol. 2010;105:376–81.
    1. Caminiti L, Passalacqua G, Barberi S, Vita D, Barberio G, De Luca R, et al. A new protocol for specific oral tolerance induction in children with IgE-mediated cow’s milk allergy. Allergy Asthma Proc. 2009;30:443–8.
    1. Fleischer DM, Conover-Walker MK, Christie L, Burks AW, Wood RA. Peanut allergy: recurrence and its management. J Allergy Clin Immunol. 2004;114:1195–201.
    1. Durham SR, Emminger W, Kapp A, Colombo G, de Monchy JG, Rak S, et al. Long-term clinical efficacy in grass pollen-induced rhinoconjunctivitis after treatment with SQ-standardized grass allergy immunotherapy tablet. J Allergy Clin Immunol. 2010;125:131–8. e1–7.
    1. Dahl R, Kapp A, Colombo G, de Monchy JG, Rak S, Emminger W, et al. Sublingual grass allergen tablet immunotherapy provides sustained clinical benefit with progressive immunologic changes over 2 years. J Allergy Clin Immunol. 2008;121:512–8. e2.
    1. Bordignon V, Burastero SE. Multiple daily administrations of low-dose sublingual immunotherapy in allergic rhinoconjunctivitis. Ann Allergy Asthma Immunol. 2006;97:158–63.
    1. 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;124:1351–2.
    1. Staden U, Blumchen K, Blankenstein N, Dannenberg N, Ulbricht H, Dobberstein K, et al. Rush oral immunotherapy in children with persistent cow’s milk allergy. J Allergy Clin Immunol. 2008;122:418–9.
    1. Uermosi C, Beerli RR, Bauer M, Manolova V, Dietmeier K, Buser RB, et al. Mechanisms of allergen-specific desensitization. J Allergy Clin Immunol. 2010;126:375–83.
    1. Knol EF, Mul FP, Jansen H, Calafat J, Roos D. Monitoring human basophil activation via CD63 monoclonal antibody 435. J Allergy Clin Immunol. 1991;88:328–38.
    1. Amano T, Furuno T, Hirashima N, Ohyama N, Nakanishi M. Dynamics of intracellular granules with CD63-GFP in rat basophilic leukemia cells. J Biochem. 2001;129:739–44.
    1. Kepley CL. Antigen-induced reduction in mast cell and basophil functional responses due to reduced Syk protein levels. Int Arch Allergy Immunol. 2005;138:29–39.
    1. 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. 2009;123:789–94. e20.
    1. Ono E, Taniguchi M, Higashi N, Mita H, Kajiwara K, Yamaguchi H, et al. CD203c expression on human basophils is associated with asthma exacerbation. J Allergy Clin Immunol. 2010;125:483–9. e3.
    1. Narisety SD, Skripak JM, Steele P, Hamilton RG, Matsui EC, Burks AW, et al. Open-label maintenance after milk oral immunotherapy for IgE-mediated cow’s milk allergy. J Allergy Clin Immunol. 2009;124:610–2.
    1. 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;124:286–91. e1–6.
    1. 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;122:1154–60.
    1. Schroeder JT, Saini SS. In: Manual of Molecular and Clinical Laboratory Immunology. 7th ed Detrick B, Hamilton RG, Folds JD, editors. ASM Press; Washington (DC): 2006.
    1. Bieneman AP, Chichester KL, Chen YH, Schroeder JT. Toll-like receptor 2 ligands activate human basophils for both IgE-dependent and IgE-independent secretion. J Allergy Clin Immunol. 2005;115:295–301.
    1. MacGlashan DW, Jr, Ishmael S, MacDonald SM, Langdon JM, Arm JP, Sloane DE. Induced loss of Syk in human basophils by non-IgE-dependent stimuli. J Immunol. 2008;180:4208–17.
    1. Ishmael SS, MacGlashan DW., Jr Syk expression in peripheral blood leukocytes, CD341 progenitors, and CD34-derived basophils. J Leukoc Biol. 2010;87:291–300.

Source: PubMed

3
Abonnere