Omalizumab in Food Allergy in Children: Current Evidence and Future Perspectives
Abstract
:1. Introduction
2. Materials and Methods
3. Results
4. Discussion
4.1. Efficacy of OMA in Food Allergy Management
4.2. Impact on Quality of Life
4.3. Economic Considerations and Patient Selection
4.4. Clarifying the Role of OMA in Food Allergy Management
4.5. Regulatory Approval and Future Perspectives
4.6. Considerations for Patients with High IgE Levels
4.7. Comparison with Other Therapeutic Strategies
4.8. Limitations and Future Directions
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Conflicts of Interest
References
- Mahdavinia, M. Food Allergy in Adults: Presentations, Evaluation, and Treatment. Med. Clin. N. Am. 2020, 104, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Anvari, S.; Miller, J.; Yeh, C.Y.; Davis, C.M. IgE-Mediated Food Allergy. Clin. Rev. Allergy Immunol. 2019, 57, 244–260. [Google Scholar] [CrossRef] [PubMed]
- Santos, A.F.; Riggioni, C.; Agache, I.; Akdis, C.A.; Akdis, M.; Alvarez-Perea, A.; Alvaro-Lozano, M.; Ballmer-Weber, B.; Barni, S.; Beyer, K.; et al. EAACI Guidelines on the Diagnosis of IgE-Mediated Food Allergy. Allergy 2023, 78, 3057–3076. [Google Scholar] [CrossRef] [PubMed]
- Foong, R.X.; Dantzer, J.A.; Wood, R.A.; Santos, A.F. Improving Diagnostic Accuracy in Food Allergy. J. Allergy Clin. Immunol. Pract. 2021, 9, 71–80. [Google Scholar] [CrossRef]
- Indolfi, C.; Klain, A.; Dinardo, G.; Grella, C.; Perrotta, A.; Colosimo, S.; Decimo, F.; Miraglia del Giudice, M. Transplant-Acquired Food Allergy in Children. Nutrients 2024, 16, 3201. [Google Scholar] [CrossRef]
- Maris, I.; Dölle-Bierke, S.; Renaudin, J.M.; Lange, L.; Koehli, A.; Spindler, T.; Hourihane, J.; Scherer, K.; Nemat, K.; Kemen, C.; et al. Peanut-Induced Anaphylaxis in Children and Adolescents: Data from the European Anaphylaxis Registry. Allergy 2021, 76, 1517–1527. [Google Scholar] [CrossRef]
- Grabenhenrich, L.B.; Dölle, S.; Moneret-Vautrin, A.; Köhli, A.; Lange, L.; Spindler, T.; Ruëff, F.; Nemat, K.; Maris, I.; Roumpedaki, E.; et al. Anaphylaxis in Children and Adolescents: The European Anaphylaxis Registry. J. Allergy Clin. Immunol. 2016, 137, 1128–1137.e1. [Google Scholar] [CrossRef]
- Westwell-Roper, C.; To, S.; Andjelic, G.; Lu, C.; Lin, B.; Soller, L.; Chan, E.S.; Stewart, S.E. Food-Allergy-Specific Anxiety and Distress in Parents of Children with Food Allergy: A Systematic Review. Pediatr. Allergy Immunol. 2022, 33, e13695. [Google Scholar] [CrossRef]
- Proctor, K.B.; Tison, K.; Estrem, H.H.; Park, J.; Scahill, L.; Vickery, B.P.; Sharp, W.G. A Systematic Review of Parent Report Measures Assessing the Psychosocial Impact of Food Allergy on Patients and Families. Allergy 2022, 77, 1347–1359. [Google Scholar] [CrossRef]
- Pecoraro, L.; Mastrorilli, C.; Arasi, S.; Barni, S.; Caimmi, D.; Chiera, F.; Dinardo, G.; Gracci, S.; Miraglia Del Giudice, M.; Bernardini, R.; et al. Nutritional and Psychosocial Impact of Food Allergy in Pediatric Age. Life 2024, 14, 695. [Google Scholar] [CrossRef]
- Fierro, V.; Di Girolamo, F.; Marzano, V.; Dahdah, L.; Mennini, M. Food Labeling Issues in Patients with Severe Food Allergies: Solving a Hamlet-like Doubt. Curr. Opin. Allergy Clin. Immunol. 2017, 17, 204–211. [Google Scholar] [CrossRef] [PubMed]
- Dinardo, G.; Fierro, V.; Del Giudice, M.M.; Urbani, S.; Fiocchi, A. Food-Labeling Issues for Severe Food-Allergic Consumers. Curr. Opin. Allergy Clin. Immunol. 2023, 23, 233–238. [Google Scholar] [CrossRef] [PubMed]
- Fierro, V.; Mazzuca, C.; Urbani, S.; Dahdah, L.; Calandrelli, V.; Valluzzi, R.L.; Dinardo, G. Perspectives in Precautionary Allergen Labelling of Prepackaged Foods after the FAO/WHO Consultation. Curr. Opin. Allergy Clin. Immunol. 2024, 24, 166–170. [Google Scholar] [CrossRef] [PubMed]
- Klain, A.; Indolfi, C.; Dinardo, G.; Grella, C.; Perrotta, A.; Decimo, F.; Miraglia, M.; Giudice, D. The Dawn of a New Era for Biological Drugs in Allergology. Ital. J. Pediatr. Allergy Immunol. 2024, 38, 14–16. [Google Scholar] [CrossRef]
- Mazzuca, C.; Fiocchi, A.; Dinardo, G.; Mennini, M.; Fierro, V. Small Molecules and Food Allergy. Ref. Modul. Food Sci. 2024, 1, 634–648. [Google Scholar] [CrossRef]
- Dinardo, G.; Cafarotti, A.; Galletta, F.; Fiocchi, A.; Arasi, S. Omalizumab in Severe Asthma and Food Allergies with IgE Levels >1500 KU/L: Two-Year Evaluation. Pediatr. Allergy Immunol. 2023, 34, e14057. [Google Scholar] [CrossRef]
- Arasi, S.; Ebisawa, M.; Eigenmann, P.; Dinardo, G. Editorial Comment on “Oral Immunotherapy as a Curative Treatment for Food-Allergic Preschool Children: Current Evidence and Potential Underlying Mechanisms”. Pediatr. Allergy Immunol. 2024, 35, e14071. [Google Scholar] [CrossRef]
- Wood, R.A.; Togias, A.; Sicherer, S.H.; Shreffler, W.G.; Kim, E.H.; Jones, S.M.; Leung, D.Y.M.; Vickery, B.P.; Bird, J.A.; Spergel, J.M.; et al. Omalizumab for the Treatment of Multiple Food Allergies. N. Engl. J. Med. 2024, 390, 889–899. [Google Scholar] [CrossRef]
- Feuille, E.; Nowak-Wegrzyn, A. Allergen-Specific Immunotherapies for Food Allergy. Allergy. Asthma Immunol. Res. 2018, 10, 189–206. [Google Scholar] [CrossRef]
- Zuberbier, T.; Wood, R.A.; Bindslev-Jensen, C.; Fiocchi, A.; Chinthrajah, R.S.; Worm, M.; Deschildre, A.; Fernandez-Rivas, M.; Santos, A.F.; Jaumont, X.; et al. Omalizumab in IgE-Mediated Food Allergy: A Systematic Review and Meta-Analysis. J. Allergy Clin. Immunol. Pract. 2023, 11, 1134–1146. [Google Scholar] [CrossRef]
- Arasi, S.; Mennini, M.; Cafarotti, A.; Fiocchi, A. Omalizumab as Monotherapy for Food Allergy. Curr. Opin. Allergy Clin. Immunol. 2021, 21, 286–291. [Google Scholar] [CrossRef] [PubMed]
- Zuberbier, T.; Muraro, A.; Nurmatov, U.; Arasi, S.; Stevanovic, K.; Anagnostou, A.; Bonaguro, R.; Chinthrajah, S.; Lack, G.; Fiocchi, A.; et al. GA2LEN ANACARE Consensus Statement: Potential of Omalizumab in Food Allergy Management. Clin. Transl. Allergy 2024, 14, e70002. [Google Scholar] [CrossRef] [PubMed]
- Comberiati, P.; Muraro, A. Omalizumab: A Definitive Cure for Food Allergies? Ital. J. Pediatr. Allergy Immunol. 2024, 38, 36–37. [Google Scholar] [CrossRef]
- Yu, L.; Zhang, H.; Pan, J.; Ye, L. Pediatric Usage of Omalizumab: A Promising One. World Allergy Organ. J. 2021, 14, 100614. [Google Scholar] [CrossRef]
- Nilsson, C.; Nordvall, L.; Johansson, S.G.O.; Nopp, A. Successful Management of Severe Cow’s Milk Allergy with Omalizumab Treatment and CD-Sens Monitoring. Asia Pac. Allergy 2014, 4, 257–260. [Google Scholar] [CrossRef]
- Sakamoto, D.; Hamada, S.; Kobayashi, Y.; Shimono, M.; Shimamura, A.; Kanda, A.; Asako, M.; Iwai, H. Omalizumab Is Effective for a Patient with Pollen-Food Allergy Syndrome Who Experienced Intractable Lip Edema. Auris. Nasus. Larynx 2023, 50, 805–810. [Google Scholar] [CrossRef]
- Crisafulli, G.; Caminiti, L.; Chiera, F.; Arasi, S.; Salzano, G.; Panasiti, I.; Barbalace, A.; Pajno, G.B. Omalizumab in Children with Severe Allergic Disease: A Case Series. Ital. J. Pediatr. 2019, 45, 13. [Google Scholar] [CrossRef]
- Badina, L.; Belluzzi, B.; Contorno, S.; Bossini, B.; Benelli, E.; Barbi, E.; Berti, I. Omalizumab Effectiveness in Patients with a Previously Failed Oral Immunotherapy for Severe Milk Allergy. Immun. Inflamm. Dis. 2022, 10, 117–120. [Google Scholar] [CrossRef]
- Arasi, S.; Cafarotti, A.; Galletta, F.; Panetta, V.; Riccardi, C.; Calandrelli, V.; Fierro, V.; Dahdah, L.; Artesani, M.C.; Valluzzi, R.L.; et al. Omalizumab Reduces Anaphylactic Reactions and Allows Food Introduction in Food-Allergic in Children with Severe Asthma: An Observational Study. Allergy 2024, 80, 1074–1085. [Google Scholar] [CrossRef]
- Crespo, J.B.; Domingo, M.V.; Arauzo, N.H.; Castillo, M.J.; Delavalle, M.B.; Foix, M.P.S.; Sánchez, D.V.; Izquierdo-Domínguez, A. Real Life Study of the Use of Omalizumab for Pediatric Patients with Multiple Food Allergies. Allergol. Immunopathol. 2021, 49, 15–22. [Google Scholar] [CrossRef]
- Steiss, J.O.; Strohner, P.; Zimmer, K.P.; Lindemann, H. Reduction of the Total IgE Level by Omalizumab in Children and Adolescents. J. Asthma 2008, 45, 233–236. [Google Scholar] [CrossRef] [PubMed]
- Sampson, H.A.; Leung, D.Y.M.; Burks, A.W.; Lack, G.; Bahna, S.L.; Jones, S.M.; Wong, D.A. A Phase II, Randomized, Double-blind, Parallel-group, Placebo-controlled Oral Food Challenge Trial of Xolair (Omalizumab) in Peanut Allergy. J. Allergy Clin. Immunol. 2011, 127, 1309–1310.e1. [Google Scholar] [CrossRef] [PubMed]
- Andorf, S.; Purington, N.; Block, W.M.; Long, A.J.; Tupa, D.; Brittain, E.; Rudman Spergel, A.; Desai, M.; Galli, S.J.; Nadeau, K.C.; et al. Anti-IgE Treatment with Oral Immunotherapy in Multifood Allergic Participants: A Double-Blind, Randomised, Controlled Trial. Lancet Gastroenterol. Hepatol. 2018, 3, 85–94. [Google Scholar] [CrossRef] [PubMed]
- Azzano, P.; Paquin, M.; Langlois, A.; Morin, C.; Parizeault, G.; Lacombe-Barrios, J.; Samaan, K.; Graham, F.; Paradis, L.; Des Roches, A.; et al. Determinants of Omalizumab Dose-Related Efficacy in Oral Immunotherapy: Evidence from a Cohort of 181 Patients. J. Allergy Clin. Immunol. 2021, 147, 233–243. [Google Scholar] [CrossRef]
- Mortz, C.G.; Parke, L.; Rasmussen, H.M.; Kjaer, H.F.; Bindslev-Jensen, C. A Randomized, Double-Blind Placebo-Controlled Study on the Efficacy of Omalizumab on Food Allergy Threshold in Children with Severe Food Allergy. Allergy 2024, 79, 964–976. [Google Scholar] [CrossRef]
- Takahashi, M.; Soejima, K.; Taniuchi, S.; Hatano, Y.; Yamanouchi, S.; Ishikawa, H.; Irahara, M.; Sasaki, Y.; Kido, H.; Kaneko, K. Oral Immunotherapy Combined with Omalizumab for High-Risk Cow’s Milk Allergy: A Randomized Controlled Trial. Sci. Rep. 2017, 7, 17453. [Google Scholar] [CrossRef]
- MacGinnitie, A.J.; Rachid, R.; Gragg, H.; Little, S.V.; Lakin, P.; Cianferoni, A.; Heimall, J.; Makhija, M.; Robison, R.; Chinthrajah, R.S.; et al. Omalizumab Facilitates Rapid Oral Desensitization for Peanut Allergy. J. Allergy Clin. Immunol. 2017, 139, 873–881.e8. [Google Scholar] [CrossRef]
- Frischmeyer-Guerrerio, P.A.; Masilamani, M.; Gu, W.; Brittain, E.; Wood, R.; Kim, J.; Nadeau, K.; Jarvinen, K.M.; Grishin, A.; Lindblad, R.; et al. Mechanistic Correlates of Clinical Responses to Omalizumab in the Setting of Oral Immunotherapy for Milk Allergy. J. Allergy Clin. Immunol. 2017, 140, 1043–1053.e8. [Google Scholar] [CrossRef]
- Nadeau, K.C.; Schneider, L.C.; Hoyte, L.; Borras, I.; Umetsu, D.T. Rapid Oral Desensitization in Combination with Omalizumab Therapy in Patients with Cow’s Milk Allergy. J. Allergy Clin. Immunol. 2011, 127, 1622–1624. [Google Scholar] [CrossRef]
- Wood, R.A.; Kim, J.S.; Lindblad, R.; Nadeau, K.; Henning, A.K.; Dawson, P.; Plaut, M.; Sampson, H.A. 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. 2016, 137, 1103–1110.e11. [Google Scholar] [CrossRef]
- Brandström, J.; Vetander, M.; Sundqvist, A.C.; Lilja, G.; Johansson, S.G.O.; Melén, E.; Sverremark-Ekström, E.; Nopp, A.; Nilsson, C. Individually Dosed Omalizumab Facilitates Peanut Oral Immunotherapy in Peanut Allergic Adolescents. Clin. Exp. Allergy 2019, 49, 1328–1341. [Google Scholar] [CrossRef] [PubMed]
- Schneider, L.C.; Rachid, R.; Lebovidge, J.; Blood, E.; Mittal, M.; Umetsu, D.T. A Pilot Study of Omalizumab to Facilitate Rapid Oral Desensitization in High-Risk Peanut-Allergic Patients. J. Allergy Clin. Immunol. 2013, 132, 1368–1374. [Google Scholar] [CrossRef] [PubMed]
- Rafi, A.; Do, L.A.T.; Katz, R.; Sheinkopf, L.E.; Simons, C.W.; Klaustermeyer, W. Effects of Omalizumab in Patients with Food Allergy. Allergy Asthma Proc. 2010, 31, 76–83. [Google Scholar] [CrossRef]
- Brandström, J.; Vetander, M.; Lilja, G.; Johansson, S.G.O.; Sundqvist, A.C.; Kalm, F.; Nilsson, C.; Nopp, A. Individually Dosed Omalizumab: An Effective Treatment for Severe Peanut Allergy. Clin. Exp. Allergy 2017, 47, 540–550. [Google Scholar] [CrossRef]
- Warren, C.; Herbert, L.; Lombard, L.; Bilaver, L.; Ibrahim, K.; Pappalardo, A.; Newmark, P.; Sharma, H.; Tobin, M.; Assa’ad, A.; et al. Validation of the Parent-Proxy Reported Food Allergy-Related Quality of Life Questionnaire (FAQLQ-PF-10) Short Form in a Large, Multisite Longitudinal Cohort of Food Allergic Child-Parent Dyads. J. Allergy Clin. Immunol. 2022, 149, AB123. [Google Scholar] [CrossRef]
- Flokstra-De Blok, B.M.J.; Van Der Meulen, G.N.; Dunngalvin, A.; Vlieg-Boerstra, B.J.; Oude Elberink, J.N.G.; Duiverman, E.J.; Hourihane, J.O.B.; Dubois, A.E.J. Development and Validation of the Food Allergy Quality of Life Questionnaire—Adult Form. Allergy 2009, 64, 1209–1217. [Google Scholar] [CrossRef]
- Fiocchi, A.; Artesani, M.C.; Riccardi, C.; Mennini, M.; Pecora, V.; Fierro, V.; Calandrelli, V.; Dahdah, L.; Valluzzi, R.L. Impact of Omalizumab on Food Allergy in Patients Treated for Asthma: A Real-Life Study. J. Allergy Clin. Immunol. Pract. 2019, 7, 1901–1909.e5. [Google Scholar] [CrossRef]
- Arasi, S.; Nurmatov, U.; Dunn-Galvin, A.; Roberts, G.; Turner, P.J.; Shinder, S.B.; Gupta, R.; Eigenmann, P.; Nowak-Wegrzyn, A.; Ansotegui, I.J.; et al. WAO Consensus on DEfinition of Food Allergy SEverity (DEFASE). World Allergy Organ. J. 2023, 16, 100753. [Google Scholar] [CrossRef]
- Sampson, H.A.; Bird, J.A.; Fleischer, D.; Shreffler, W.G.; Spergel, J.M. Who Are the Potential Patients for Omalizumab for Food Allergy? Ann. Allergy Asthma Immunol. 2024, 132, 569–571. [Google Scholar] [CrossRef]
- Sindher, S.B.; Fiocchi, A.; Zuberbier, T.; Arasi, S.; Wood, R.A.; Chinthrajah, R.S. The Role of Biologics in the Treatment of Food Allergy. J. Allergy Clin. Immunol. Pract. 2023, 12, 562–568. [Google Scholar] [CrossRef]
- Dinardo, G.; Cafarotti, A.; Fierro, V.; Artesani, M.C.; Indolfi, C.; Miraglia Del Giudice, M.; Fiocchi, A. Role of Biologics in Severe Food Allergy. Curr. Opin. Allergy Clin. Immunol. 2024, 24, 138–143. [Google Scholar] [CrossRef] [PubMed]
- FDA Approves First Medication to Help Reduce Allergic Reactions to Multiple Foods After Accidental Exposure|FDA. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-medication-help-reduce-allergic-reactions-multiple-foods-after-accidental (accessed on 1 March 2024).
- Menzella, F.; Just, J.; Sauerbeck, I.S.; Mailaender, C.; Saccheri, F.; Thonnelier, C.; Jaumont, X.; Mala, L. Omalizumab for the Treatment of Patients with Severe Allergic Asthma with Immunoglobulin E Levels above >1500 IU/ML. World Allergy Organ. J. 2023, 16, 100787. [Google Scholar] [CrossRef] [PubMed]
- Johansson, S.G.O.; Haahtela, T.; O’Byrne, P.M. Omalizumab and the Immune System: An Overview of Preclinical and Clinical Data. Ann. Allergy. Asthma Immunol. 2002, 89, 132–138. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.Y.; Sindher, S.B.; Stinson, R.; DaVeiga, S.P. Efficacy and Safety of Omalizumab in Pediatric Patients with High Immunoglobulin E Levels: A Case Series. Allergy Asthma Proc. 2018, 39, 289–291. [Google Scholar] [CrossRef]
- Morales-Cabeza, C.; Infante, S.; Cabrera-Freitag, P.; Fuentes-Aparicio, V.; Zubeldia, J.M.; Álvarez-Perea, A. Oral Immunotherapy and Risk of Eosinophilic Esophagitis in Children: 15 Years’ Experience. J. Pediatr. Gastroenterol. Nutr. 2023, 76, 53–58. [Google Scholar] [CrossRef]
- Calvani, M.; Leonardi, L.; Remotti, D.; Paparella, R.; Bianchi, A. Is a Strict Cow’s Milk-Free Diet Necessary to Treat Milk Oral Immunotherapy-Related Eosinophilic Esophagitis? Acta Biomed. 2021, 92, e2021033. [Google Scholar] [CrossRef]
Reference | Type of Study | Number of Participants | Age (Range) | Intervention | OMA Dosage | Endpoints | Foods Involved | Duration | Results | Adverse Events |
---|---|---|---|---|---|---|---|---|---|---|
Wood et al., 2024 [18] | Double-blind, randomized, placebo-controlled trial | 180 participants, of which 177 aged 1 to 17 years | 1–55 years | OMA monotherapy | 300 mg every 2 weeks or 225 mg every 4 weeks based on weight and IgE levels | Primary endpoint: To assess whether participants could tolerate at least 600 mg of peanut protein in a single dose without experiencing dose-limiting symptoms after 16–20 weeks of treatment. Secondary endpoints: To determine whether participants could tolerate at least 1000 mg of cashew, milk, and egg (each) without dose-limiting symptoms; to evaluate tolerance to increasing cumulative doses of one, two, or all three foods, up to a total of 6044 mg | Peanut, cashew, milk, egg, walnut, wheat, hazelnut | 52 weeks | Increased threshold for peanut and other allergens; 67% achieved tolerance to ≥600 mg peanut protein vs. 7% placebo. At week 16, tolerance to a single dose of ≥1000 mg of food protein was achieved in the omalizumab group, by 41% for cashew (28/68 vs. 3%, 1/31), 67% for egg (34/51 vs. 0%, 0/20), and 66% for milk (27/41 vs. 10%, 2/21). No improvement in QoL was detected during the blinded phase | The safety profile was similar between the OMA and placebo groups, except for injection-site reactions, which were more common in the OMA group. One serious adverse event occurred in a 1-year-old participant who experienced elevated liver enzymes. No serious adverse events were reported during the open-label extension phase |
Mortz et al., 2024 [35] | Randomized, double-blind, placebo-controlled | 20 | 6–17 years | OMA monotherapy | Asthma-based dosing adjusted to total IgE and weight; doses every 2–4 weeks per protocol | Primary endpoint: To evaluate whether omalizumab increases the allergen threshold in food-allergic children after 3 months of treatment, assessed using a double-blind placebo-controlled food challenge (DBPCFC). Secondary endpoints: Changes in SPT size; Levels of specific IgE, specific IgG4 (s-IgG4), and total IgE (t-IgE); threshold changes at 6 months; QoL and atopic comorbidities | Peanut, hazelnut, cashew, walnut, egg | 3–6 months | Significant increase in food allergy thresholds; all treated participants increased tolerance levels ≥2 challenge steps (p = 0.003) At 6 months, many children tolerated very high doses (up to 44,000 mg of food protein). Improvements were observed in SPT: significant reduction in wheal size (p < 0.02); s-IgG4: significantly increased (p < 0.006); s-IgE and t-IgE: increased as expected during anti-IgE therapy. No significant change in QoL | No significant differences in adverse events between the two groups. OMA group (n = 14): 5 children had mild adverse events within 3 months (viral infections, cystitis, constipation, pneumonia). From 3–6 months, 3 additional mild events occurred. No events occurred from 3–6 months |
Andorf et al., (2018) [33] | Randomized, double-blind, placebo-controlled, phase 2 | 48 | 4–15 years | OMA combined with multifood OIT | Dosage based on weight and IgE levels, stopped by week 36 | Primary endpoint: The efficacy of OMA combined with multifood OIT in passing DBPCFCs at week 36 from at least 2 of their offending foods. Secondary endpoints: Tolerance to higher doses; tolerance to more foods; successful completion of OIT with minimal symptoms; time to reach maintenance dosing: the speed at which participants were able to reach the 2 g maintenance dose per food | Cashew, walnut, hazelnut, almond, sesame, milk, egg, peanut, soy, wheat | 36 weeks | 83% of participants in the OMA group tolerated 2 g of protein for ≥ 2 foods vs. 33% in placebo (p = 0.004). All participants who passed the 2 g challenge (primary endpoint) also passed the 4 g challenge for at least 2 foods | No serious or severe adverse event were reported. AE incidence (weeks 8–16) was lower in the OMA group: 27% OMA vs. 68% (placebo) p = 0.008. No increase in AEs after OMA was discontinued at week 16 |
MacGinnitie et al. (2017) [37] | Randomized, double-blind, placebo-controlled, multicenter trial | 37 | 7–19 years | OMA treatment followed by OIT for peanut allergy | Based on weight and IgE levels | Primary endpoint: Tolerance to peanut protein six weeks after discontinuation of OMA or placebo. Secondary endpoint: Tolerance to a greater dose of peanut protein during an oral food challenge 12 weeks after stopping treatment | Peanut | 32 weeks |
| Adverse events were mostly mild and manageable, with allergic reactions occurring after 7.8% of OIT doses in the OMA group versus 16.8% in placebo. Moderate and severe reactions were more common in the placebo group, though not statistically significant. Three cases of suspected eosinophilic esophagitis resolved after stopping peanut intake |
Brandström et al. (2017) [44] | Open-label, phase 2 study | 23 | 12–19 years | Individually dosed OMA based on CD-sens monitoring | Adjusted based on IgE and weight. Final dose: up to 600 mg every 2 weeks | Primary endpoint: The suppression of peanut allergy symptoms, measured by a negative or favorable CD-sens test (basophil allergen threshold sensitivity) and the ability to undergo an open peanut challenge without severe allergic reactions. Secondary endpoints: The peanut protein dose tolerated during the challenge; the reduction in symptom severity and the dose adjustment strategy guided by CD-sens and IgE biomarkers, to optimize individual OMA dosing | Peanut | 8–24 weeks | Significant reduction in allergen sensitivity:
| After OMA treatment, 78% of patients had no allergic symptoms during the peanut challenge, and the remaining 22% experienced only mild symptoms. No severe reactions occurred. The CD-sens-guided dosing strategy was both safe and effective, with no significant differences in adverse event rates between dosing groups |
Crisafulli et al. (2019) [27] | Case series | 8 | 7–15 years | OMA therapy combined with OIT | 150–450 mg every 2–4 weeks based on IgE levels and weight | - | Cow’s milk | 6–26 months |
| Adverse events included local reactions at the site of injection and asthenia fever, headache, and generalized malaise after drug administration. Anaphylaxis occurred in 3 patients, leading to therapy discontinuation. Other adverse effects included nausea, urticaria, rhinitis, and asthma |
Badina et al. (2022) [28] | Single-center, prospective, observational | 4 | 8–24 years | OMA treatment followed by OIT | Based on s-IgE and weight; maximum 600 mg as per asthma guidelines | Primary endpoint: Facilitation of milk reintroduction by OMA Secondary endpoint: Increase in tolerated milk protein dose after 8 weeks of OMA; changes in immunological markers; improvement in asthma control | Cow’s milk | 12 months | Improved tolerance to cow’s milk protein, no severe reactions during OIT, increased IgG4 levels, improved quality of life. Thresholds maintained post-OMA. Food Allergy Quality of Life Questionnaire (FAQLQ) scores improved in all four patients by the end of the study | No adverse effects related to omalizumab were recorded. One patient chose to reduce their milk intake post-OMA discontinuation due to anxiety |
Sampson et al. (2011) [32] | Phase 2, randomized, double-blind, placebo-controlled | 14 patients of which 8 were pediatric | 6–75 years | OMA treatment followed by oral food challenge (OFC) | Minimum 0.016 mg/kg/IgE every 4 weeks, adjusted by IgE levels | Primary endpoint: Assess OMA’s efficacy in increasing peanut tolerance. Secondary endpoints: IgE reduction and safety | Peanut flour | 24 weeks | 44.4% of OMA-treated subjects tolerated ≥1000 mg of peanut flour after 24 weeks vs. 20% in placebo. Trend toward greater tolerance shift with OMA (p = 0.054). A significant decrease in free IgE was observed | OMA group: 76.5% experienced mild-to-moderate AEs; 2 cases of anaphylaxis occurred during pre-treatment oral food challenges (OFCs) Placebo group: 88.9% experienced mild-to-moderate AEs. No severe AEs reported during treatment |
Nilsson et al. (2014) [25] | Case series | 5 | 6–16 years | OMA treatment with CD-sens monitoring (basophil allergen threshold sensitivity) | Based on serum IgE and body weight; maximum 600 mg/2 weeks | Primary endpoint: Assess efficacy of OMA in achieving tolerance to cow’s milk in severe allergy. Secondary endpoints: Immunological changes and clinical improvements | Cow’s milk | 16 weeks of OMA, continued for several years at lowest effective dose (75 mg/4 weeks) to sustain tolerance | All the patients achieved negative CD-sens and tolerated milk in challenges. 1 patient required doubled OMA dose to reach negative CD-sens. Reduction in IgE antibodies to milk proteins. CD-sens values dropped to 0 in all patients post-treatment. Asthma/rhino-conjunctivitis/eczema symptoms improved in most patients | No severe AEs attributed to OMA. The anaphylaxis risk was avoided during challenges due to CD-sens monitoring |
Steiss et al. (2008) [31] | Observational study | 9 | 8–17 years | OMA therapy for severe allergic asthma | Dosage based on body weight and IgE level: −150 to 750 mg every 2–4 weeks, depending on patient-specific factors (e.g., weight, IgE baseline) | Primary endpoint: Reduction of total IgE levels during OMA therapy in children/adolescents with severe allergic asthma. Secondary endpoints: Asthma exacerbations; rescue medication use and comorbid allergy management | Peanut, others not specifically mentioned | 6 months up to 24 months |
Significant decrease in total IgE after 6 months of therapy (p < 0.01)
| No severe AEs reported. No systemic reactions to subcutaneous injections |
Frischmeyer-Guerrerio et al. (2017) [38] | Randomized, placebo-controlled study | 57 patients of which 40 aged 7–17 years old | 7–35 years | OMA in combination with OIT for milk allergy | 0.016 mg/kg/IgE IU (max dose ≤750 mg) | Primary endpoint: Investigate mechanistic effects of OMA combined with milk OIT and identify baseline biomarkers predictive of clinical benefit. Secondary endpoints: Safety and immunological changes | Cow’s milk (casein) | OMA administered for 16 months (combined with OIT from month 4–28). OIT continued for 24–32 months | OMA significantly suppressed milk-induced basophil CD63 expression at month 4 (before starting OIT), with p values of 0.0074 and 0.0135 for 0.1 and 10 mg/mL milk, respectively. Compared to placebo, CD63 expression was also significantly lower at various concentrations (p = 0.0040 to 0.0014). Histamine release was significantly increased in OMA-treated subjects upon stimulation with milk and anti-IgE (p values ranging from 0.0035 to 0.0302), suggesting enhanced basophil sensitivity in washed cell preparations | In the OMA group, reduced moderate/severe reactions vs. placebo (p < 0.01) were observed. No severe AEs attributed to OMA. In the placebo group higher rates of gastrointestinal and systemic symptoms during OIT were noted |
Azzano et al. (2021) [34] | Cohort study | 181 | 5.2–13.9 | OMA pre-treatment (≥2 months) to OIT | Weight (and IgE)-based dosage | Primary endpoint: Identify determinants of OMA dose-related efficacy in OIT for food allergy. Secondary endpoint: Pharmacokinetics/pharmacodynamics | Up to 6 allergens simultaneously (e.g., peanuts, egg, milk, tree nuts) | OMA started ≥2 months pre-OIT. The total duration varied by patients |
| 118/181 (65%) had reactions; only 8 (4.4%) were moderate/severe. No severe AEs attributed to OMA |
Takahashi et al. (2017) [36] | Randomized controlled trial | 16 | 6–14 years | OIT combined with OMA (OMA-OIT) | Dose calculated based on total IgE and body weight. Typically administered every 2–4 weeks for 24 weeks | Primary endpoint: Induction of desensitization to cow’s milk at 32 weeks after study entry. Secondary endpoints: Change in successfully consumed dose; immunologic responses and safety | Cow’s milk | 32 weeks (24 weeks of OMA + OIT, followed by 8 weeks of OIT alone) | All 10 children in OMA-OIT group desensitized; none in control group desensitized. Wheal size reduced in OMA-OIT group. Significant increase in tolerance for CM protein (median 2080 mg vs. 0 mg in controls; p < 0.001) and fresh CM (200 mL vs. 0 mL; p = 0.006) | No severe AEs reported |
Wood et al. (2016) [40] | Randomized, double-blind, placebo-controlled trial | 57 | 7–32 years. Not specified how many patients were pediatric | OMA combined with milk oral immunotherapy (MOIT) | Based on weight and IgE level; max 750 mg every 2–4 weeks | Primary endpoint: Passing a 10 g milk protein oral food challenge (OFC) after 8 weeks off OIT. Secondary endpoints: Desensitization at month 28 and safety/adverse events | Cow’s milk | 32 months | Significant safety improvements: fewer dose-related reactions. No significant difference in desensitization or sustained unresponsiveness (SU) between the two groups | OMA group had fewer and milder reactions. No severe reactions requiring discontinuation. Epinephrine use: 2 doses (OMA) vs. 18 doses (placebo) |
Nadeau et al. (2011) [39] | Pilot phase 1 study | 11 | 7–17 years | OMA combined with OIT | Dosed every 2–4 weeks; based on weight and IgE level (up to 300 mg) | Primary endpoint: Desensitization to 2000 mg/day of milk protein within 7–11 weeks of starting OIT. Secondary endpoints: Safety and long-term tolerability | Cow’s milk | 52 weeks | 9 of 11 participants achieved desensitization to 2000 mg/day in 7–11 weeks. All 9 patients could tolerate ≥8000 mg/day after discontinuation of OMA | Low reaction rate (1.6%), mostly mild. Epinephrine needed in 3/11 subjects |
Schneider et al. (2013) [42] | Pilot study | 13 | 7–16 years | Oral peanut desensitization with OMA pre-treatment | Administered every 2–4 weeks based on European guidelines. Doses ranged from 150–600 mg depending on IgE levels and weight | Primary endpoint: First-day desensitization to 500 mg peanut flour. Secondary endpoints: Achieving maintenance dose (4000 mg/day); long-term tolerance (8000 mg challenge at Week 32) and safety | Peanut | 48–156 weeks of OMA treatment continued for median of 4 years | 92% reached a maintenance dose of 4000 mg peanut flour/day. Tolerance to 8000 mg (approx. 20 peanuts) achieved in 85% of participants after OMA discontinuation | Reaction rate: 2% of doses (72 reactions total): mild 1.8% (no treatment needed), moderate 0.2% (antihistamines used), severe 0.06% (epinephrine used in 2 cases) |
Brandström et al. (2019) [41] | Phase 2 study | 23, 13/23 (57%) were ≤17 years old | 12–19 years | Peanut oral immunotherapy (pOIT) with OMA pre-treatment | Individualized, adjusted every 2–4 weeks, based on IgE levels and clinical response | Primary endpoint: Tolerating pOIT for 12 weeks after stopping OMA, followed by a negative open peanut challenge (2800 mg). Secondary endpoint: Safety, immunological markers, clinical outcomes | Peanut | 8-week up-dosing phase, with an individualized tapering schedule afterward | 48% achieved the primary endpoint of tolerating 2800 mg peanut protein 12 weeks after stopping OMA. Adverse reactions increased when OMA was reduced or stopped | Systemic reactions: 43 reactions in 16,095 doses (0.3%): mild: 20 (0.1%). Moderate: 22 (0.1%). Severe: 1 (0.006%). Common symptoms: oropharyngeal and abdominal. Adrenaline use: 8/23 (35%) required adrenaline |
Rafi et al. (2010) [43] | Prospective pilot study | 22 patients, of which 5 were pediatric | 14–66 years | Treatment of IgE-mediated food allergies in asthma patients using OMA | 150–300 mg every 2–4 weeks | Primary endpoint: Reduction or elimination of allergic symptoms upon accidental or intentional reexposure to sensitized foods. Secondary endpoints: Symptom-specific improvements and safety | Fish, shellfish, peanut, tree nuts, egg, soybean, wheat | At least 1 year | 100% of patients (22/22) showed improvement by the 6th dose. Food-induced asthma exacerbations improved in 13/22 (59%) patients. Atopic dermatitis: improved in 8/22 (36%) patients. Rhinosinusitis: Improved in 6/22 (27%) patients. Urticaria: improved in 3/22 (14%) patients. Angioedema/anaphylaxis: improved in 9/22 (41%) patients | Common AEs: injection site reactions, viral infections, upper respiratory infections, sinusitis, headaches. Serious AEs (SAEs): anaphylaxis risk (required 2 h post-injection monitoring) |
Arasi et al. (2024) [29] | Observational study | 69 | 6–18 years | OMA therapy for food allergies in asthmatic children | Dosage adjusted per baseline IgE and weight, capped at t-IgE of 1500 IU/L before June 2021, later ≤2500 IU/L | Primary endpoint: Increase in threshold of reactivity (NOAEL) to food allergens after 4 months of OMA treatment, measured via oral food challenges (OFCs). Secondary endpoints: Proportion of negative OFCs at T1 (tolerance without reaction). NOAEL increase at T2 and T3 in patients who failed T1 challenges. Reduction in anaphylactic episodes (pre-treatment vs. post-treatment). Reduction in non-anaphylactic food reactions | Peanut, tree nuts, fish, egg, milk, wheat | 1 year | Increased tolerance to foods: 66.4% of foods tolerated at T1; quality of life (FA-QoL): children ≤12 years: improved from 4.63 ± 0.74 to 2.02 ± 1.13; adolescents: improved from 4.68 ± 0.92 to 1.90 ± 1.50. Significant improvements noted across physical, emotional, and social domains | None reported related to omalizumab administration |
Sakamoto et al. (2023) [26] | Case report | 1 | 12 years | Sublingual immunotherapy (SLIT) combined with OMA | 600 mg/month | Improvement/resolution of intractable lip edema caused by pollen–food allergy syndrome (PFAS) | Oranges, apples, peaches, tomatoes, strawberries, tomato sauce | 4 years | Marked improvement in lip edema and nasal symptoms; no relapse of lip edema for ~2 years | No adverse reactions reported from OMA during the study period |
Crespo et al. (2021) [30] | Observational study | 5 | 4–8 years | OMA treatment with OFC | 150–300 mg every 4 weeks; adjusted per patient | Primary endpoint: Effectiveness of OMA in enabling tolerance to previously allergenic foods in children with multiple food allergies (MFAs), evaluated through oral food challenges (OFCs). Secondary endpoints: Changes in total and specific IgE levels. Introduction of foods previously excluded due to anaphylaxis. Ability to reduce OMA dose or extend administration interval | Various foods including nuts, fish, vegetables, fruits, and cereals | 2.3 to 6.2 years | Significant increase in food tolerance, reduction in allergic reactions, improved quality of life | No local or systemic adverse events related to OMA reported |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Indolfi, C.; Perrotta, A.; Dinardo, G.; Klain, A.; Grella, C.; Palumbo, P.; Miraglia del Giudice, M. Omalizumab in Food Allergy in Children: Current Evidence and Future Perspectives. Life 2025, 15, 681. https://doi.org/10.3390/life15050681
Indolfi C, Perrotta A, Dinardo G, Klain A, Grella C, Palumbo P, Miraglia del Giudice M. Omalizumab in Food Allergy in Children: Current Evidence and Future Perspectives. Life. 2025; 15(5):681. https://doi.org/10.3390/life15050681
Chicago/Turabian StyleIndolfi, Cristiana, Alessandra Perrotta, Giulio Dinardo, Angela Klain, Carolina Grella, Paola Palumbo, and Michele Miraglia del Giudice. 2025. "Omalizumab in Food Allergy in Children: Current Evidence and Future Perspectives" Life 15, no. 5: 681. https://doi.org/10.3390/life15050681
APA StyleIndolfi, C., Perrotta, A., Dinardo, G., Klain, A., Grella, C., Palumbo, P., & Miraglia del Giudice, M. (2025). Omalizumab in Food Allergy in Children: Current Evidence and Future Perspectives. Life, 15(5), 681. https://doi.org/10.3390/life15050681