Skip to main content

MINI REVIEW article

Front. Nutr., 02 December 2022
Sec. Nutritional Immunology
Volume 9 - 2022 | https://doi.org/10.3389/fnut.2022.1050554

The potential role of preventive and therapeutic immunonutrition strategies for pediatric food allergy: A mini-review

Serena Coppola1,2 Laura Carucci1,2 Roberta De Michele3 Roberto Berni Canani1,2*
  • 1Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
  • 2ImmunoNutritionLAB at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
  • 3Department of Community Medicine and Primary Care, University of Naples Federico II, Naples, Italy

Food allergy (FA) represents one of the main chronic conditions of the pediatric population. The gut microbiome (GM)-immune system axis is a milestone in affecting FA susceptibility. The dynamic and bidirectional crosstalk between the GM and immune system starts early in life, and it is deeply modulated during the first 1,000 days of life. Nutritional factors during this crucial period mainly influence the proper GM-immune system development and function across the lifespan, with potential beneficial or detrimental effects on health status. Immunonutrition strategies, applied from conception, could represent an innovative target for prevention and treatment of pediatric FA. Here we described the potential role of preventive and therapeutic immunonutrition strategies for pediatric FA, highlighting putative future perspectives in this field.

Introduction

Food allergy (FA) is one of the major chronic diseases of the pediatric population, affecting up to 10% of children in industrialized countries, mainly in the first years of life (1). Pediatric FA derives from a failure of the immune tolerance network in early life. Alterations of the proper development and functioning of the immune system, leading to the immune tolerance network failure, are mainly modulated by the gut microbiome (GM). This evidence suggests the importance of the “GM-immune system axis” (2, 3). Indeed, negative factors affecting GM composition in the pre-natal or early life (e.g., cesarean delivery, proton pump inhibitor, anti-septic and antibiotic use etc.) leads to a reduction in bacterial biodiversity, to an increased intestinal permeability, and to an impairment of GM function with a decreased production of immunomodulatory compounds such as short chain fatty acids (SCFAs), deeply involved in the regulation of immune tolerance mechanisms and T regulatory (T reg) induction (4, 5). Indeed, FA children present specific GM signatures and growing evidence shows that GM alterations, leading to an impairment of the immune system, could also facilitate the occurrence of other chronic non-communicable diseases (NCDs), such as obesity and autoimmune and inflammatory disorders (57). By contrast, exposure to factors that positively affect the structure and function of the GM (e.g., breastfeeding, high fiber diet, assumption of pre-, pro, and synbiotics, etc.) leads to a positive modulation of the axis and potential subsequent protection against the occurrence of NCDs in early life as well in adulthood (810). The GM of healthy breastfed infants is mainly characterized by an abundance of Bifidobacteria, which play a pivotal role in optimal immune system development. Indeed, Bifidobacterium species through the production of some immunomodulating factors such as SCFAs are able to exert a positive influence on innate immunity, secretory IgA production and Th1/Th2 balance, favoring a Th1 response with a protective effect against allergic diseases. The underlying mechanism is not completely defined, but it involves an increase of regulatory T cells, needed for maintaining the intestinal immune homeostasis (5, 1114). Figure 1 graphically represents the mechanisms involved in GM-immune system axis regulation. The dynamic and bidirectional interaction between the GM and immune system starts early in life, from the conception and has lifelong effects. However, the crucial time frame to establish the health status for the baby and for his adult life are the “first 1,000 days” (15), a critical window of opportunity in which both GM colonization and immune system development occur (16).

FIGURE 1
www.frontiersin.org

Figure 1. The GM-immune system axis is responsible for pivotal regulation of long term health status, mainly through epigenetic mechanisms. For instance, healthy dietary and lifestyle habits such as Mediterranen diet (full of fibers, fermented foods, antioxidants, omega-3), breastfeeding, exposure to pets and/or rural environment, vaginal delivery, and use of probiotics are responsible for a positive influence on the axis. Conversely, cesarean section, prenatal, and early-life exposure to antibiotics/gastric acidity inhibitors/antiseptic agents, unhealthy diet (low fibers/high saturated fats and junk foods) influence negatively the axis. These environmental variables are becoming important targets for active prevention of FA.

Starting from this evidence, an early integrated approach could prevent and counteract gut dysbiosis by modulating the interaction among dietary factors, the GM, and the immune system and it open the way for the “immunonutrition” concept. This strategy refers to the potential of specific nutrients in the modulation of immune system activity (17). From this innovative point of view, the immunonutrition approach, applied early in life, could represent a new strategy to prevent and to potentially treat GM-immune system derived diseases such as FA.

In this Mini-Review we described the preventive and potential therapeutic immunonutrition strategies for pediatric FA highlighting future perspectives in this field.

The main immunonutrition strategies to prevent FA

During the critical period of the first 1,000 days of life, nutritional factors are the main environmental factors influencing the immune system function and the future disease susceptibility (18). In this paragraph we report the main immunonutrition strategies to prevent FA in the first 1,000 days.

The first nutritional exposure for the fetus derives from the maternal diet during pregnancy, representing one of the main in utero programming factors able to modulate the risk of later life allergy (19). The consumption, during pregnancy, of a holistic diet based on healthy foodstuffs typical of the Mediterranean diet and low in saturated fats and refined foods, may protect the offspring against allergies (2025). The Mediterranean diet is characterized by high consumption of unprocessed cereals, vegetables, legumes, fruits, nuts, and fish, providing large amounts of poly-unsaturated (eicosapentaenoic and docosahexaenoic acids) and mono-unsaturated fatty acids, vitamins, trace elements, polyphenols, and antioxidants, known for their anti-inflammatory and anti-allergic properties (26). Notably, the Mediterranean diet's beneficial effects are due to the interactive and synergistic nutrient combinations, and the epigenetic changes that affect the gene expression is one of the mechanisms by which this dietary pattern could lead to regulatory effects in the offspring immune system (27).

After delivery, maternal diet remains one of the main factors able to influence the allergy risk in the newborn through breastfeeding. A high adherence to the Mediterranean diet during lactation positively influences the composition of human milk (HM), potentially protecting the child against FA (28). Although the European Academy of Allergy and Clinical Immunology (EAACI) guidelines for the prevention of FA reports no recommendation for the use of breastfeeding to prevent FA, healthcare professionals should encourage breastfeeding wherever possible for its overall positive benefits for infants and mothers (29). HM is considered the “gold standard” for infant nutrition, and it is recommended to be the exclusive source of nourishment for the first 4–6 months of life and, after the start of complementary feeding at 4–6 months, up to 2 years of age or beyond (30). Several reasons explain the potential protective effects of HM on allergy susceptibility in children: breastfeeding extends the relationship with the maternal immune system, it could impact the immune tolerance to dietary antigens, and it is a source of a large amount of biologically active compounds that positively influence the infant's GM and immune system (31). Among the bioactive factors, breast milk contains oligosaccharides (HMOs), which have prebiotic activity, pass undigested to the colon, and serve as fermentation substrates for beneficial commensal bacteria, such as Bifidobacterium and Lactobacillus, to support the proper development of the GM (32). The main microbial fermentation products of HMOs are SCFAs, which exert immunomodulatory functions, beneficially modulating the immune system development and function (33). Among SCFAs, butyrate is able to modulate several tolerogenic mechanisms, exerting a pivotal role in the FA protection. In addition, considerable amounts of butyrate were detected in HM, confirming again the protective role of breastfeeding against pediatric FA (34). Furthermore, HM is a source of several non-nutritive protective factors, which provide passive immunity as well as stimulation for the maturation of the infant's GM and immune system through the interaction with the digestive and respiratory tracts mucous membranes, such as immunoglobulins (secretory IgA, IgM), cytokines, enzymes (lactoferrin, lysozyme, etc.), nucleotides, complement system components, leukocytes, microRNA and hormones (35).

The timing of the introduction of allergenic foods into the infant's diet is currently one of the most promising strategies to prevent FA. The current guidelines do not recommend avoiding or delaying the introduction of food allergens in the infant's diet to prevent allergy, suggesting the protective effect of early introduction of complementary foods against FA (29). The introduction of complementary solid foods and liquids, other than HM or infant formula, should not be started before 4 months and not be delayed beyond 6 months of life (36). At weaning, the complementary diet composition and the quality of foods are the major determinants of the GM development and function (37). The consumption in the first year of life of healthy foodstuffs such as fruit, vegetables, yogurt, and fish, has been associated with high levels of fecal butyrate and a reduced risk of FA (38). On the contrary, the increased consumption of ultra-processed foods typical of a Western diet, rich in dietary advanced glycation end-products (e.g., fast foods, roasted/barbecued meats, sweets, and beverages), could result in the development of allergic phenotypes (39).

Starting from all of this evidence, early immunonutrition strategies are the key to prevent the onset of FA and to protect against the future disease susceptibility.

The potential therapeutic immunonutrition strategies for pediatric FA: The active diet therapy

In this paragraph, we focus on the potential therapeutic effects of immunonutrition against pediatric FA. Once FA have already arisen, the cornerstone of immunonutrition is moving from a passive line of action, based on the elimination from the diet of food allergens to relieve symptoms, to a “pro-active” one, underlining the possibility of positively modulating the immune system toward the immune tolerance acquisition. Different active dietary strategies could be adopted, based on a positive modulation of the GM-immune system axis. These could be summarized in the infant formula choice for infants who are not breastfed and are affected by one of the most common FA, the cow's milk protein allergy (CMA), and the use of pro-, pre-, and synbiotics, the dietary intake of baked foodstuffs containing the food allergens, and oral immunotherapy (17).

In CMA babies, if breastfeeding is not available, a substitutive infant formula adapted to CMA dietary management is required (40). The commercially available infant formulas for CMA management are extensively hydrolyzed whey formula, extensively hydrolyzed casein formula, soy formula, partially hydrolyzed rice formula, and amino acid-based formula (41). Substitutive formulas represent the main cost for the management of CMA, thus, options to speed up the immune tolerance acquisition are important for families of affected infants and for health care systems. Our Research Team have recently demonstrated an in vitro different modulation exerted by the protein fraction of the above-mentioned infant formulas used for CMA management on tolerogenic mechanisms (42). In particular, we showed that only the protein fraction deriving from the extensively hydrolyzed casein formula stimulated tolerogenic effects, at least by an epigenetic modulation of the gene expression of FoxP3, which plays a central role in maintaining the homeostasis and tolerance of the immune system (42). Similarly, several randomized-controlled clinical trials have demonstrated that the consumption of extensively hydrolyzed casein formula (alone or added with the probiotic Lactobacillus rhamnosus GG), compared to other substitutive formulas, could accelerate the acquirement of the immune tolerance and could protect CMA children against the onset of Atopic March (4345).

Furthermore, factors that positively modulate the composition and function of the GM, e.g., pro-, pre, and synbiotics, could drive the immune system of FA children toward a tolerogenic pathway. The administration of probiotics (live microorganisms that, when administered in adequate amounts, confer health benefits) could be a useful strategy to improve gastrointestinal symptoms of CMA infants, as shown in a randomized study evaluating the efficacy of 1 × 109 CFU daily dose of Bifidobacterium lactis BB-12 and of 1 × 108 CFU daily dose of Streptococcus thermophilus TH-4 (46). Another double blind, randomized, placebo-controlled study, performed on 100 infants affected by CMA, indicated the efficacy of the oral administration of Lactobacillus rhamnosus GG in association with a cow's milk protein free-diet for 4 weeks in ameliorating symptoms such as vomiting, diarrhea, restiveness, bloody and mucous stool, and abdominal distension, compared to the placebo (47). Our group have also demonstrated that the consumption of an extensively hydrolyzed casein formula with added Lactobacillus rhamnosus GG determined a higher rate of immune tolerance acquirement and a lower incidence of other atopic manifestations onset at 6 months and 1 and 3 years later than the extensively hydrolyzed casein formula alone and the other commercially available substitutive formula for CMA management (43, 44, 48). Furthermore, our group have also shown that the management of CMA infants with an extensively hydrolyzed casein formula with added Lactobacillus rhamnosus GG resulted in an increase of the gut bacterial strains butyrate producing the main tolerogenic metabolite (49).

In addition, the role of prebiotics and synbiotics in the GM-immune system axis modulation has also been suggested. Prebiotics are the main substrate for the growth and/or functionality of GM beneficial microbes (50). The addition of the prebiotic lactose to an extensively hydrolyzed whey formula was able to increase the total fecal amounts of Lactobacillus/Bifidobacteria in CMA infants, resulting in an increase of the beneficial metabolite butyrate (51). Instead, synbiotics are defined as a mixture of live microorganisms and substrate(s) selectively used by host microorganisms that confer health benefits (52). A recent multicenter trial in which infants affected by non-IgE-mediated CMA were treated with amino acid-based formula containing a symbiotic, consisting of Bifidobacterium breve M-16V and fructo-oligosaccharides, elicited beneficial effects on the GM composition of CMA patients, bringing it close to a healthy breastfed GM (53, 54).

In weaned FA children, other active dietary strategies could be adopted, such as the possibility to introduce baked products containing the food allergens. In fact, the allergenic properties of food allergen proteins could be modified during food handling such as lactic fermentation and heat treatment (55), and the immunoglobulins-binding capability could be affected by these proteins' structural changes (glycation, aggregation, unfolding, etc…), with a potential decrease of allergenicity (56, 57). Furthermore, the exposure of food allergen proteins to elevated temperatures in culinary recipes in association with a matrix (e.g., wheat, sugar, and oil, for the creation of a muffin) resulted in a final reduced immunoreactivity if compared with just heated food containing allergens (58). It has been reported that there is a generally good prognosis of cow's milk and egg protein allergies with the introduction into the diet of particularly baked forms of milk and egg (55, 5963); conversely, the prognoses of wheat and soy allergies have not been well describrd. Identifying children who tolerate baked products could be extremely important, both to increase the diet variety and the quality of life, but also to accelerate the immune tolerance acquisition (64). In this direction, due to the large number of patients tolerant to baked products, a step-by-step approach has been proposed by the British Society for Allergy and Clinical Immunology. They have suggested “milk ladder” and “egg ladder” strategies, based on the gradual increase of quantity and allergenicity consumption of food allergens, to speed up the immune tolerance acquisition to fresh milk and egg (65). Unfortunately, to date, there are no diagnostic screening tests to detect patients that could tolerate baked products, and a need for standardization is required to use ladders safely and successfully in the clinical practice (66).

In the case of persistent FA, one of the potential therapeutic immunonutrition strategies is the oral immunotherapy (OIT). This kind of strategy consists of a regular administration of incremental doses of the culprit food, mainly adopted for peanuts, cow's milk, and eggs allergies (6769). There are no standardized OIT protocols, but regular step-up consumption of the allergen has been shown to increase the reactivity threshold of a specific antigen, resulting in a protection against allergic reactions due to accidental ingestion of the culprit food (70). One of the weaknesses of OIT is related to the risk of the development of severe allergic reactions during the protocol as well as the lack of data regarding a long-term effect of this strategy (71). Indeed, OIT is able to transiently modulate different humoral and cellular pathways, but the ability to induce a long-lasting effect in terms of immune tolerance has not yet been established (17). OIT surely represents an emerging area of research, but due the limitations regarding safety and long-term efficacy, is not yet used in the clinical treatment routine (72). The development of biological therapies could change the fate of OIT. In fact different biological drugs administered as adjunctive OIT treatment have shown promising results in terms of OIT efficacy and safety (73).

Discussion

The incidence and prevalence of pediatric FA has increased significantly in recent decades. Preventive and therapeutic strategies are advocated to limit the disease burden.

Through the immunonutrition approach, i.e., the ability of specific dietary factors to modulate the development and function of the immune system, pediatric FA could be prevented and, when already arisen, could be managed through an integrated proactive approach able to speed up the oral tolerance acquisition and to prevent the Atopic March onset.

This narrative Mini-Review provided an overview of the more solid and scientific-based preventive and therapeutic immunonutrition strategies for pediatric FA. Figure 2 graphically summarizes the key messages of these immunonutrition strategies.

FIGURE 2
www.frontiersin.org

Figure 2. Through a positive modulation of GM-Immune System axis, nutritional factors are able to prevent and potentially treat FA.

Author contributions

SC, LC, RDM, and RBC analyzed the literature and wrote and read the manuscript. All authors have substantially contributed to the development, conception, and design of the present Mini-Review and listed have made a direct and intellectual contribution to the manuscript and approved it for publication.

Acknowledgments

We thank all researchers of our Center at the ImmunoNutritionLAB dedicated to the clinical and basic research activities of new preventive and therapeutic strategies for pediatric food allergy.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Loh W, Tang MLK. The epidemiology of food allergy in the global context. Int J Environ Res Public Health. (2018) 15:2043. doi: 10.3390/ijerph15092043

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. (2020) 30:492–506. doi: 10.1038/s41422-020-0332-7

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Carucci L, Coppola S, Luzzetti A, Giglio V, Vanderhoof J, Berni Canani R. The role of probiotics and postbiotics in modulating the gut microbiome-immune system axis in the pediatric age. Minerva Pediatr. (2021) 73:115–27. doi: 10.23736/S2724-5276.21.06188-0

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Di Costanzo M, Carucci L, Berni Canani R, Biasucci G. Gut microbiome modulation for preventing and treating pediatric food allergies. Int J Mol Sci. (2020) 21:5275. doi: 10.3390/ijms21155275

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Berni Canani R, Paparo L, Nocerino R, Di Scala C, Della Gatta G, Maddalena Y, et al. Gut microbiome as target for innovative strategies against food allergy. Front Immunol. (2019) 10:191. doi: 10.3389/fimmu.2019.00191

PubMed Abstract | CrossRef Full Text | Google Scholar

6. De Filippis F, Paparo L, Nocerino R, Della Gatta G, Carucci L, Russo R, et al. Specific gut microbiome signatures and the associated pro-inflammatory functions are linked to pediatric allergy and acquisition of immune tolerance. Nat Commun. (2021) 12:5958. doi: 10.1038/s41467-021-26266-z

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Noce A, Marrone G, Di Daniele F, Ottaviani E, Wilson Jones G, Bernini R, et al. Impact of gut microbiota composition on onset and progression of chronic non-communicable diseases. Nutrients. (2019) 11:1073. doi: 10.3390/nu11051073

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Feehley T, Plunkett CH, Bao R, Choi Hong SM, Culleen E, Belda-Ferre P, et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat Med. (2019) 25:448–53. doi: 10.1038/s41591-018-0324-z

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Zhang X, Zhivaki D, Lo-Man R. Unique aspects of the perinatal immune system. Nat Rev Immunol. (2017) 17:495–507. doi: 10.1038/nri.2017.54

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. (2014) 158:705–21. doi: 10.1016/j.cell.2014.05.052

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Arroyo Hornero R, Hamad I, Côrte-Real B, Kleinewietfeld, M. The impact of dietary components on regulatory T cells and disease. Front Immunol. (2020) 11:253. doi: 10.3389/fimmu.2020.00253

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Harmsen HJ, Wildeboer–Veloo AC, Raangs GC, Wagendorp AA, Klijn N, Bindels JG, et al. Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. J Pediatr Gastroenterol Nutr. (2000) 30:61–7. doi: 10.1097/00005176-200001000-00019

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Rivière A, Selak M, Lantin D, Leroy F, De Vuyst L. Bifidobacteria and butyrate-producing colon bacteria: importance and strategies for their stimulation in the human gut. Front Microbiol. (2016) 7:979. doi: 10.3389/fmicb.2016.00979

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hinz D, Bauer M, Röder S, Olek S, Huehn J, Sack U, et al. Cord blood tregs with stable FOXP3 expression are influenced by prenatal environment and associated with atopic dermatitis at the age of one year. Allergy. (2012) 67:380–9. doi: 10.1111/j.1398-9995.2011.02767.x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Torow N, Hornef MW. The neonatal window of opportunity: setting the stage for life-long host-microbial interaction and immune homeostasis. J Immunol. (2017) 198:557–63. doi: 10.4049/jimmunol.1601253

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Selma-Royo M, Tarrazó M, García-Mantrana I, GómezGallego C, Salminen S, Collado MC. Shaping microbiota during the first 1000 days of life. Adv Exp Med Biol. (2019). 1125:3–24. doi: 10.1007/5584_2018_312

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Carucci L, Coppola S, Luzzetti A, Voto L, Giglio V, Paparo L, et al. Immunonutrition for pediatric patients with cow's milk allergy: how early interventions could impact long-term outcomes. Front Allergy. (2021) 2:676200. doi: 10.3389/falgy.2021.676200

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Moreno Villares JM, Collado MC, Larqué E, Leis Trabazo R, Saenz De Pipaón M, Moreno Aznar LA. Los primeros 1000 días: una oportunidad para reducir la carga de las enfermedades no transmisibles [The first 1000 days: an opportunity to reduce the burden of noncommunicable diseases]. Nutr Hosp. (2019) 36:218–32. doi: 10.20960/nh.02453

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Grieger JA, Clifton VL, Tuck AR, Wooldridge AL, Robertson SA, Gatford KL. In utero programming of allergic susceptibility. Int Arch Allergy Immunol. (2016) 169:80–92. doi: 10.1159/000443961

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Chatzi L, Garcia R, Roumeliotaki T, Basterrechea M, Begiristain H, Iniguez C, et al. Mediterranean diet adherence during pregnancy and risk of wheeze and eczema in the first year of life: INMA (Spain) and RHEA (Greece) mother-child cohort studies. Br J Nutr. (2013) 110:2058–68. doi: 10.1017/S0007114513001426

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chatzi L, Torrent M, Romieu I, Garcia-Esteban R, Ferrer C, Vioque J, et al. Mediterranean diet in pregnancy is protective for wheeze and atopy in childhood. Thorax. (2008) 63:507–13. doi: 10.1136/thx.2007.081745

PubMed Abstract | CrossRef Full Text | Google Scholar

22. de Batlle J, Garcia-Aymerich J, Barraza-Villarreal A, Anto JM, Romieu I. Mediterranean diet is associated with reduced asthma and rhinitis in Mexican children. Allergy. (2008) 63:1310–6. doi: 10.1111/j.1398-9995.2008.01722.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Erkkola M, Nwaru BI, Kaila M, Kronberg-Kippila C, Ilonen J, Simmel O, et al. Risk of asthma and allergic outcomes in the offspring in relation to maternal food consumption during pregnancy: a Finnish birth cohort study. Pediatr Allergy Immunol. (2012) 23:186–94. doi: 10.1111/j.1399-3038.2012.01272.x

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Nwaru BI, Erkkola M, Lumia M, Kronberg-Kippila C, Ahonen S, Kaila M, et al. Maternal intake of fatty acids during pregnancy and allergies in the offspring. Br J Nutr. (2012) 108:720–32. doi: 10.1017/S0007114511005940

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Sausenthaler S, Koletzko S, Schaaf B, Lehmann I, Borte M, Herbarth O, et al. Maternal diet during pregnancy in relation to eczema and allergic sensitization in the offspring at 2 years of age. Am J Clin Nutr. (2007) 85:530–7. doi: 10.1093/ajcn/85.2.530

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Hogenkamp A, Ehlers A, Garssen J, Willemsen LEM. Allergy modulation by N-3 long chain polyunsaturated fatty acids and fat soluble nutrients of the mediterranean. Diet Front Pharmacol. (2020) 11:1244. doi: 10.3389/fphar.2020.01244

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Del Chierico F, Vernocchi P, Dallapiccola B, Putignani L. Mediterranean diet and health: food effects on gut microbiota and disease control. Int J Mol Sci. (2014) 15:11678–99. doi: 10.3390/ijms150711678

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Vassilopoulou E, Feketea G, Konstantinou GN, Zekakos Xypolias D, Valianatou M, Petrodimopoulou M, et al. Food protein-induced allergic proctocolitis: the effect of maternal diet during pregnancy and breastfeeding in a mediterranean population. Front Nutr. (2022) 9:843437. doi: 10.3389/fnut.2022.843437

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Halken S, Muraro A, de Silva D, Khaleva E, Angier E, Arasi S, et al. EAACI guideline: preventing the development of food allergy in infants and young children (2020 update). Pediatr Allergy Immunol. (2021) 32:843–58. doi: 10.1111/pai.13496

PubMed Abstract | CrossRef Full Text | Google Scholar

30. World Health Organization. Infant and Young Child Feeding. (2021). Available online at: https://www.who.int (accessed October 29, 2022).

Google Scholar

31. Ferrante G, Carta M, Montante C, Notarbartolo V, Corsello G, Giuffrè M. Current insights on early life nutrition and prevention of allergy. Front Pediatr. (2020) 8:448. doi: 10.3389/fped.2020.00448

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Lyons KE, Ryan CA, Dempsey EM, Ross RP, Stanton C. Breast milk, a source of beneficial microbes and associated benefits for infant health. Nutrients. (2020) 12:1039. doi: 10.3390/nu12041039

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. (2013) 431:569–73. doi: 10.1126/science.1241165

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Paparo L, Nocerino R, Ciaglia E, Di Scala C, De Caro C, Russo R, et al. Butyrate as a bioactive human milk protective component against food allergy. Allergy. (2021) 76:1398–415. doi: 10.1111/all.14625

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Cacho NT, Lawrence RM. Innate immunity and breast milk. Front Immunol. (2017) 8:584. doi: 10.3389/fimmu.2017.00584

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Fewtrell M, Bronsky J, Campoy C, Domellöf M, Embleton N, Fidler Mis N, et al. Complementary feeding: a position paper by the European society for paediatric gastroenterology, hepatology, and nutrition (ESPGHAN) committee on nutrition. J Pediatr Gastroenterol Nutr. (2017) 64:119–32. doi: 10.1097/MPG.0000000000001454

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Laursen MF, Andersen LB, Michaelsen KF, Mølgaard C, Trolle E, Bahl MI, et al. Infant gut microbiota development is driven by transition to family foods independent of maternal obesity. MSphere. (2016) 1:e00069–15. doi: 10.1128/mSphere.00069-15

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Roduit C, Frei R, Ferstl R, Loeliger S, Westermann P, Rhyner C, et al. High levels of butyrate and propionate in early life are associated with protection against atopy. Allergy. (2019) 74:799–809. doi: 10.1111/all.13660

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Smith PK, Masilamani M, Li XM, Sampson HA. The false alarm hypothesis: food allergy is associated with high dietary advanced glycation end-products and proglycating dietary sugars that mimic alarmins. J Allergy Clin Immunol. (2017) 139:429-437. doi: 10.1016/j.jaci.2016.05.040

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Koletzko S, Niggemann B, Arato A, Dias JA, Heuschkel R, Husby S, et al. Diagnostic approach and management of cow's-milk protein allergy in infants and children: ESPGHAN GI committee practical guidelines. J Pediatr Gastroenterol Nutr. (2012) 55:221–9. doi: 10.1097/MPG.0b013e31825c9482

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Fiocchi A, Schunemann H, Ansotegui I, Assa'ad A, Bahna S, Berni Canani R, et al. The global impact of the DRACMA guidelines cow's milk allergy clinical practice. World Allergy Organ J. (2018) 11:2. doi: 10.1186/s40413-017-0179-7

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Paparo L, Picariello G, Bruno C, Pisapia L, Canale V, Sarracino A, et al. Tolerogenic effect elicited by protein fraction derived from different formulas for dietary treatment of cow's milk allergy in human cells. Front Immunol. (2021) 1:3910. doi: 10.3389/fimmu.2020.604075

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Berni Canani R, Nocerino R, Terrin G, Frediani T, Lucarelli S, Cosenza L, et al. Formula selection for management of children with cow's milk allergy influences the rate of acquisition of tolerance: a prospective multicenter study. J Pediatr. (2013) 163:771–7. doi: 10.1016/j.jpeds.2013.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Berni Canani R, Di Costanzo M, Bedogni G, Amoroso A, Cosenza L, Di Scala C, et al. Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestations in children with cow's milk allergy: 3-year randomized controlled trial. J Allergy Clin Immunol. (2017) 139:1906–13.e4. doi: 10.1016/j.jaci.2016.10.050

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Nocerino R, Bedogni G, Carucci L, Cosenza L, Cozzolino T, Paparo L, et al. The impact of formula choice for the management of pediatric cow's milk allergy on the occurrence of other allergic manifestations: the atopic march cohort study. J Pediatr. (2021) 232:183–91.e3. doi: 10.1016/j.jpeds.2021.01.059

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Ivakhnenko ES, Nian'kovskii SL. Effect of probiotics on the dynamics of gastrointestinal symptoms of food allergy to cow's milk protein in infants. Georgian Med News. (2013) 219:46–52.

PubMed Abstract | Google Scholar

47. Basturk A, Isik I, Atalay A, Yilmaz A. Investigation of the efficacy of Lactobacillus rhamnosus gG in infants with cow's milk protein allergy: a randomised double-blind placebo-controlled trial. Probiotics Antimicrob Proteins. (2020) 12:138–43. doi: 10.1007/s12602-019-9516-1

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Berni Canani R, Nocerino R, Terrin G, Coruzzo A, Cosenza L, Leone L, et al. Effect of Lactobacillus GG on tolerance acquisition in infants with cow's milk allergy: a randomized trial. J Allergy Clin Immunol. (2012) 129:580–2:2.e1–5. doi: 10.1016/j.jaci.2011.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Berni Canani R, Sangwan N, Stefka AT, Nocerino R, Paparo L, Aitoro R, et al. Lactobacillus rhamnosus GG supplemented formula expands butyrate producing bacterial strains in food allergic infants. ISME J. (2016) 10:742–50. doi: 10.1038/ismej.2015.151

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, et al. Expert consensus document: the international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. (2017) 14:491–502. doi: 10.1038/nrgastro.2017.75

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Francavilla R, Calasso M, Calace L, Siragusa S, Ndagijimana M, Vernocchi P, et al. Effect of lactose on gut microbiota and metabolome of infants with cow's milk allergy. Pediatr Allergy Immunol. (2012) 23:420–7. doi: 10.1111/j.1399-3038.2012.01286.x

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Swanson KS, Gibson GR, Hutkins R, Reimer RA, Reid G, Verbeke K, et al. The international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. (2020) 17:687–701. doi: 10.1038/s41575-020-0344-2

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Fox AT, Wopereis H, Van Ampting MTJ, Oude Nijhuis MM, Butt AM, Peroni DG, et al. A specific synbiotic-containing amino acid-based formula in dietary management of cow's milk allergy: a randomized controlled trial. Clin Transl Allergy. (2019) 9:5. doi: 10.1186/s13601-019-0241-3

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Wopereis H, van Ampting MTJ, Cetinyurek-Yavuz A, Slump R, Candy DCA, Butt AM, et al. A specific synbiotic-containing amino acid based formula restores gut microbiota in non-IgE mediated cow's milk allergic infants: a randomized controlled trial. Clin Transl Allergy. (2019) 9:27. doi: 10.1186/s13601-019-0267-6

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Küçükosmanoglu E, Özen E, Eltan SB, Özkars MY, Keskin Ö. Most children who are allergic to cow's milk tolerate yogurt. J Int Med Res. (2018) 46:5099–106. doi: 10.1177/0300060518790430

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Bavaro SL, De Angelis E, Barni S, Pilolli R, Mori F, Novembre EM, et al. Modulation of milk allergenicity by baking milk in foods: a proteomic investigation. Nutrients. (2019) 11:1536. doi: 10.3390/nu11071536

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Villa C, Costa J, Oliveira MBPP, Mafra I. Bovine milk allergens: a comprehensive review. Compr Rev Food Sci Food Saf. (2018) 17:137–64. doi: 10.1111/1541-4337.12318

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Vilar LK, Araújo FA, Santos TP, Menezes TT, Cheik MF, Segundo GRS. Baked tolerance in Cow's milk allergy: quite frequent, hard to predict! Int Arch Allergy Immunol. (2021) 182:319–23. doi: 10.1159/000511148

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Monaco S, Russo G, Romano A, Liotti L, Verga MC, Miceli Sopo S. Yogurt is tolerated by the majority of children with IgE-mediated cow's milk allergy. Allergol Immunopathol. (2019) 47:322–7. doi: 10.1016/j.aller.2018.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Weinbrand-Goichberg J, Benor S, Rottem M, Shacham N, Mandelberg A, Levine A, et al. Long-term outcomes following baked milk-containing diet for IgE-mediated milk allergy. J Allergy Clin Immunol Pract. (2017) 5:1776–78.e1. doi: 10.1016/j.jaip.2017.04.018

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Sackesen C, Suárez-Fariñas M, Silva R, Lin J, Schmidt S, Getts R, et al. A new luminex-based peptide assay to identify reactivity to baked, fermented, and whole milk. Allergy. (2019) 74:327–36. doi: 10.1111/all.13581

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Sirin Kose S, Asilsoy S, Uzuner N, Karaman O, Anal O. Outcomes of baked milk and egg challenge in cow's milk and hen's egg allergy: can tolerance be predicted with allergen-specific IgE and prick-to-prick test? Int Arch Allergy Immunol. (2019) 180:264–73. doi: 10.1159/000502957

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Alessandri C, Sforza S, Palazzo P, Lambertini F, Paolella S, Zennaro D, et al. Tolerability of a fully maturated cheese in cow's milk allergic children: biochemical, immunochemical, and clinical aspects. PLoS ONE. (2012) 7:e40945. doi: 10.1371/journal.pone.0040945

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Luyt D, Ball H, Makwana N, Green MR, Bravin K, Nasser SM, et al. BSACI guideline for the diagnosis and management of cow's milk allergy. Clin Exp Allergy. (2014) 44:642–72. doi: 10.1111/cea.12302

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Leech SC, Ewan PW, Skypala IJ, Brathwaite N, Erlewyn-Lajeunesse M, Heath S, et al. BSACI 2021 guideline for the management of egg allergy. Clin Exp Allergy. (2021) 51:1262–78. doi: 10.1111/cea.14009

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Venter C, Meyer R, Ebisawa M, Athanasopoulou P, Mack DP. Food allergen ladders: a need for standardization. Pediatr Allergy Immunol. (2022) 33:e13714. doi: 10.1111/pai.13714

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Vickery BP, Berglund JP, Burk CM, Fine JP, Kim EH, Kim JI, et al. Early oral immunotherapy in peanut-allergic preschool children is safe and highly effective. J Allergy Clin Immunol. (2017) 173–81.e8. doi: 10.1016/j.jaci.2016.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

68. 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) 24:75–83. doi: 10.1111/j.1399-3038.2012.01341.x

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Pajno GB, Caminiti L, Salzano G, Crisafulli G, Aversa T, Messina MF, et al. Comparison between two maintenance feeding regimens after successful cow's milk oral desensitization. Pediatr Allergy Immunol. (2013) 32:376–81. doi: 10.1111/pai.12077

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Wood RA. Oral immunotherapy for food allergy. J Investig Allergol Clin Immunol. (2017) 27:151–9. doi: 10.18176/jiaci.0143

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Kim EH, Burks AW. Food allergy immunotherapy: oral immunotherapy and epicutaneous immunotherapy. Allergy. (2020) 75:1337–46. doi: 10.1111/all.14220

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Anagnostou K, Stiefel G, Brough H, du Toit G, Lack G, Fox AT. Active management of food allergy: an emerging concept. Arch Dis Child. (2015) 100:386–90. doi: 10.1136/archdischild-2014-306278

PubMed Abstract | CrossRef Full Text | Google Scholar

73. De Martinis M, Sirufo MM, Suppa M, Ginaldi L. New perspectives in food allergy. Int J Mol Sci. (2020) 21:1474. doi: 10.3390/ijms21041474

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: pediatric food allergy, allergic diseases, gut microbiome, active diet, immunonutrition, allergy

Citation: Coppola S, Carucci L, De Michele R and Berni Canani R (2022) The potential role of preventive and therapeutic immunonutrition strategies for pediatric food allergy: A mini-review. Front. Nutr. 9:1050554. doi: 10.3389/fnut.2022.1050554

Received: 21 September 2022; Accepted: 01 November 2022;
Published: 02 December 2022.

Edited by:

Elvira Verduci, San Paolo Hospital, Italy

Reviewed by:

Mohanned Alhussien, Technical University of Munich, Germany

Copyright © 2022 Coppola, Carucci, De Michele and Berni Canani. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Roberto Berni Canani, berni@unina.it

Download