Anti-IgE treatment with oral immunotherapy in multifood allergic participants: a double-blind, randomised, controlled trial

Sandra Andorf, Natasha Purington, Whitney M Block, Andrew J Long, Dana Tupa, Erica Brittain, Amanda Rudman Spergel, Manisha Desai, Stephen J Galli, Kari C Nadeau, R Sharon Chinthrajah, Sandra Andorf, Natasha Purington, Whitney M Block, Andrew J Long, Dana Tupa, Erica Brittain, Amanda Rudman Spergel, Manisha Desai, Stephen J Galli, Kari C Nadeau, R Sharon Chinthrajah

Abstract

Background: Despite progress in single food oral immunotherapy, there is little evidence concerning the safety and efficacy of treating individuals with multiple food (multifood) allergies. We did a pilot study testing whether anti-IgE (omalizumab) combined with multifood oral immunotherapy benefited multifood allergic patients.

Methods: We did a blinded, phase 2 clinical trial at Stanford University. We enrolled participants, aged 4-15 years, with multifood allergies validated by double-blind, placebo-controlled food challenges to their offending foods. Inclusion criteria included a positive skin prick test of 6 mm or more (wheal diameter, above the negative control), a food-specific serum IgE concentration of more than 4 kU/L for each food, or both, and a positive double-blind, placebo-controlled food challenge at 500 mg or less of food protein. Exclusion criteria included eosinophilic oesophagitis and severe asthma. Participants were randomised (3:1) with a block size of four, to receive multifood oral immunotherapy to two to five foods, together with omalizumab (n=36) or placebo (n=12). 12 individuals who fulfilled the same inclusion and exclusion criteria were included as controls. These individuals were not randomised and received neither omalizumab nor oral immunotherapy. Omalizumab or placebo was administered subcutaneously for 16 weeks, with oral immunotherapy starting at week 8, and was stopped 20 weeks before the exit double-blind, placebo-controlled food challenge at week 36. The primary endpoint was the proportion of participants who passed double-blind, placebo-controlled food challenges to at least two of their offending foods. This completed trial is registered with ClinicalTrials.gov, number NCT02643862.

Findings: Between March 25, 2015, and Aug 18, 2016, 165 participants were assessed for eligibility, of whom 84 did not meet the inclusion criteria and 21 declined to participate. We enrolled and randomised 48 eligible participants and the remaining 12 patients were included as nonrandomised, untreated controls. At week 36, a significantly greater proportion of the omalizumab-treated (30 [83%] of 36) versus placebo (four [33%] of 12) participants passed double-blind, placebo-controlled food challenges to 2 g protein for two or more of their offending foods (odds ratio 10·0, 95% CI 1·8-58·3, p=0·0044). All participants completed the study. There were no serious or severe (grade 3 or worse) adverse events. Participants in the omalizumab group had a significantly lower median per-participant percentage of oral immunotherapy doses associated with any adverse events (27% vs 68%; p=0·0082). The most common adverse events in both groups were gastrointestinal events.

Interpretation: In multifood allergic patients, omalizumab improves the efficacy of multifood oral immunotherapy and enables safe and rapid desensitisation.

Funding: US National Institutes of Health (NIH).

Conflict of interest statement

Declaration of interest

We declare no competing interests.

Copyright © 2018 Elsevier Ltd. All rights reserved.

Figures

Figure 1:
Figure 1:
Consort Diagram demonstrating the phase 2 randomized placebo-controlled trial design (Omalizumab vs. Placebo arms).
Figure 2:. Overview of foods in each…
Figure 2:. Overview of foods in each participant’s multifood OIT
Overview of foods in each participant’s multifood OIT (all shown foods had a positive DBPCFC at baseline for that participant), grouped by study arm (red for omalizumab arm, blue for placebo arm) and primary endpoint outcome. The outcome of the 2 g DBPCFC for each food in week 36 is shown in green when negative (i.e., the participant passed the DBPCFC for that food) and in dark red when positive (i.e., the participant failed the DBPCFC for that food). The failures didn’t undergo food challenges in week 36 and the foods in their multifood OIT are marked by gray boxes.
Figure 3:. Percentage of participants per study…
Figure 3:. Percentage of participants per study arm who tolerated 2 g (primary endpoint) or 4 g (secondary endpoint) in DBPCFCs to at least 2 foods at week 36.
Every participant who passed the primary endpoint also passed this secondary endpoint. Significantly more participants in the omalizumab arm (83%) passed either endpoint than in the placebo arm (33%) [P = 0·004, OR: 10, 95% CI: 1·8 – 58·3].
Figure 4:. Time since starting therapy (i.e.,…
Figure 4:. Time since starting therapy (i.e., starting omalizumab or placebo) to reach a 2 g maintenance dose per food
(A) The time since starting therapy (i.e., starting omalizumab or placebo) for participants in each study arm to reach a 2 g maintenance dose per food. Study failures (6 in the omalizumab arm, 8 in the placebo arm) who never reached 2 g maintenance are censored (marked by vertical black tick marks) at week 36. Participants receiving omalizumab reached the 2 g maintenance dose per food faster (P = 0·001) than the participants on placebo. (B) The time since starting therapy for participants in the omalizumab arm to reach a 2 g maintenance dose per food, stratified by the number of foods in that participant’s multifood OIT. Study failures are censored (marked by vertical black tick marks) at week 36. Participants with lower numbers of foods in their OIT show a trend of reaching the 2 g maintenance dose per food faster (P = 0·03).
Figure 5:. Tolerated dose at IDED (initial…
Figure 5:. Tolerated dose at IDED (initial dose escalation day).
(A) Each data point represents the tolerated dose per food (total tolerated dose divided by the number of foods in the multifood OIT) for each participant stratified by study arm (left) or by study arm and primary endpoint outcome (middle and right). (B) The total dose of food protein tolerated at IDED for each participant. The ‘hinges’ represent the first and third quartile. The whiskers are the smallest and largest values after outliers are excluded. Outliers are defined as values greater than the 75th percentile plus 1·5 times the interquartile range (IQR), or less than 25th percentile minus 1·5 times the IQR.

References

    1. 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 2011; 127(6): 1622–4.
    1. Schneider LC, Rachid R, LeBovidge J, Blood E, Mittal M, Umetsu DT. A pilot study of omalizumab to facilitate rapid oral desensitization in high-risk peanut-allergic patients. J Allergy Clin Immunol 2013; 132(6): 1368–74.
    1. Begin P, Dominguez T, Wilson SP, et al. Phase 1 results of safety and tolerability in a rush oral immunotherapy protocol to multiple foods using Omalizumab. Allergy Asthma Clin Immunol 2014; 10(1): 7.
    1. Gupta RS, Springston EE, Warrier MR, et al. The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics 2011; 128(1): e9–17.
    1. Burks A, Jones S, Wood R, et al. Oral immunotherapy for treatment of egg allergy in children. N Engl J Med 2012; 367(3): 233–43.
    1. Jones SM, Pons L, Roberts JL, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. The Journal of allergy and clinical immunology 2009; 124(2): 292–300, e1–97.
    1. Anagnostou K, Islam S, King Y, 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.
    1. Hsiao K-C, Ponsonby A-L, Axelrad C, et al. 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. The Lancet Child & Adolescent Health 2017; 1(2): 97–105.
    1. Navines-Ferrer A, Serrano-Candelas E, Molina-Molina GJ, Martin M. IgE-Related Chronic Diseases and Anti-IgE-Based Treatments. J Immunol Res 2016; 2016: 8163803.
    1. Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol 2016; 16(12): 751–65.
    1. Wood RA, Kim JS, Lindblad R, et al. A randomized, double-blind, placebo-controlled study of omalizumab combined with oral immunotherapy for the treatment of cow’s milk allergy. J Allergy Clin Immunol 2015; 137(4): 1103-10.e1-11.
    1. MacGinnitie AJ, Rachid R, Gragg H, et al. Omalizumab facilitates rapid oral desensitization for peanut allergy. J Allergy Clin Immunol 2016; 139(3): 873–81.e8.
    1. Martorell-Calatayud C, Michavila-Gomez A, Martorell-Aragones A, et al. Anti-IgE-assisted desensitization to egg and cow’s milk in patients refractory to conventional oral immunotherapy. Pediat Allerg Imm-Uk 2016; 27(5): 544–6.
    1. Frischmeyer-Guerrerio PA, Masilamani M, Gu W, et al. Mechanistic correlates of clinical responses to omalizumab in the setting of oral immunotherapy for milk allergy. J Allergy Clin Immunol 2017; 140(4): 1043–53 e8.
    1. Hamilton MA. Choosing the parameter for a 2 x 2 table or a 2 x 2 x 2 table analysis. Am J Epidemiol 1979; 109(3): 362–75.
    1. Fay MP. Confidence intervals that match Fisher’s exact or Blaker’s exact tests. Biostatistics 2010; 11(2): 373–4.
    1. R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2017.
    1. SAS Institute Inc. SAS software version [9.4] of the SAS system. Cary, NC, USA.
    1. Wickham H ggplot2: Elegant Graphics for Data Analysis. New York: Springer; 2009.
    1. Andorf S, Borres M, Block W, et al. Association of clinical reactivity with sensitization to allergen components in multifood-allergic children. J Allergy Clin Immunol Pract 2017; 5(5): 1325–34.e4.
    1. Sampson HA, Ho DG. Relationship between food-specific IgE concentrations and the risk of positive food challenges in children and adolescents. J Allergy Clin Immunol 1997; 100(4): 444–51.
    1. Ta V, Weldon B, Yu G, Humblet O, Neale-May S, Nadeau K. Use of specific IgE and skin prick test to determine clinical reaction severity. Br J Med Med Res 2011; 1(4): 410–29.
    1. Roberts G, Pfaar O, Akdis CA, et al. EAACI Guidelines on Allergen Immunotherapy: Allergic Rhinoconjunctivitis. Allergy 2017; [Epub ahead of print].
    1. Lin C, Lee IT, Sampath V, et al. Combining anti-IgE with oral immunotherapy. Pediatr Allergy Immunol 2017; [Epub ahead of print].
    1. 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; 133(2): 468–75 e6.
    1. Blumchen K, Ulbricht H, Staden U, et al. Oral peanut immunotherapy in children with peanut anaphylaxis. J Allergy Clin Immunol 2010; 126(1): 83–91 e1.
    1. Anagnostou K, Clark A, King Y, Islam S, Deighton J, Ewan P. Efficacy and safety of high-dose peanut oral immunotherapy with factors predicting outcome. Clin Exp Allergy 2011; 41(9): 1273–81.
    1. Vazquez-Ortiz M, Alvaro M, Piquer M, et al. Baseline specific IgE levels are useful to predict safety of oral immunotherapy in egg-allergic children. Clin Exp Allergy 2014; 44(1): 130–41.
    1. Vazquez-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 Allergy 2013; 43(1): 92–102.
    1. Meglio P, Giampietro PG, Carello R, Gabriele I, Avitabile S, Galli E. Oral food desensitization in children with IgE-mediated hen’s egg allergy: a new protocol with raw hen’s egg. Pediatr Allergy Immunol 2013; 24(1): 75–83.
    1. Leonard SA. Debates in allergy medicine: baked milk and egg ingestion accelerates resolution of milk and egg allergy. World Allergy Organ J 2016; 9: 1.
    1. Arasi S, Otani IM, Klingbeil E, et al. Two year effects of food allergen immunotherapy on quality of life in caregivers of children with food allergies. Allergy Asthma Clin Immunol 2014; 10(1): 57.
    1. Nowak-Wegrzyn A, Fiocchi A. Is oral immunotherapy the cure for food allergies? Curr Opin Allergy Clin Immunol 2010; 10(3): 214–9.
    1. Lock RJ, Unsworth DJ. Food allergy: which tests are worth doing and which are not? Ann Clin Biochem 2011; 48(Pt 4): 300–9.
    1. Grabenhenrich L, Lange L, Hartl M, et al. The component-specific to total IgE ratios do not improve peanut and hazelnut allergy diagnoses. J Allergy Clin Immunol 2016; 137(6): 1751–60 e8.
    1. Horimukai K, Hayashi K, Tsumura Y, et al. Total serum IgE level influences oral food challenge tests for IgE-mediated food allergies. Allergy 2015; 70(3): 334–7.

Source: PubMed

3
Abonnere