Next Article in Journal
A Comparison of Sugar Intake between Individuals with High and Low Trait Anxiety: Results from the NutriNet-Santé Study
Next Article in Special Issue
The Use of an Amino Acid Formula Containing Synbiotics in Infants with Cow’s Milk Protein Allergy—Effect on Clinical Outcomes
Previous Article in Journal
An Intermittent Fasting Mimicking Nutrition Bar Extends Physiologic Ketosis in Time Restricted Eating: A Randomized, Controlled, Parallel-Arm Study
Previous Article in Special Issue
Amino Acid Formula Containing Synbiotics in Infants with Cow’s Milk Protein Allergy: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cow’s Milk Protein Allergy as a Model of Food Allergies

1
Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
2
Specialty School of Paediatrics, Alma Mater Studiorum, University of Bologna, via G.Massarenti n 11, 40138 Bologna, Italy
3
Department of Medical and Surgical Sciences (DIMEC), University of Bologna, via G. Massarenti n.9, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Submission received: 30 March 2021 / Revised: 18 April 2021 / Accepted: 26 April 2021 / Published: 30 April 2021
(This article belongs to the Special Issue New Insights into Cow's Milk and Allergy)

Abstract

:
Cow’s milk allergy (CMA) is one of the most common food allergies in infants, and its prevalence has increased over recent years. In the present paper, we focus on CMA as a model of food allergies in children. Understanding the diagnostic features of CMA is essential in order to manage patients with this disorder, guide the use of an elimination diet, and find the best moment to start an oral food challenge (OFC) and liberalize the diet. To date, no shared tolerance markers for the diagnosis of food allergy have been identified, and OFC remains the gold standard. Recently, oral immunotherapy (OIT) has emerged as a new therapeutic strategy and has changed the natural history of CMA. Before this, patients had to strictly avoid the food allergen, resulting in a decline in quality of life and subsequent nutritional, social, and psychological impairments. Thanks to the introduction of OIT, the passive approach involving rigid exclusion has changed to a proactive one. Both the heterogeneity in the diagnostic process among the studies and the variability of OIT data limit the comprehension of the real epidemiology of CMA, and, consequentially, its natural history. Therefore, well-planned randomized controlled trials are needed to standardize CMA diagnosis, prevention, and treatment strategies.

1. Introduction

The prevalence of food allergies (FAs) and especially cow’s milk allergy (CMA, currently one of the most common FAs among children [1,2]) has increased in recent decades.
CMA is defined as a reproducible adverse reaction to one or more cow’s milk (CM) protein (usually casein or serum β-lactoglobulin) [3]. The underlying immunological mechanism, presentation times, and organs involved differentiate CMA from other adverse reactions to CM such as lactose intolerance [4].
CMA, like all FAs, can be divided into two main categories according to the type of immunological mechanism underlying it: immunoglobulin (Ig)E-mediated or non-IgE mediated [5] (Figure 1). IgE-mediated reactions are the most common. On the other hand, there are non-IgE mediated reactions that can arise from other cellular processes involving eosinophils or T-cells.
CMA usually occurs in the first 2 years of life and especially within the first year, unlike other allergies, such as peanut, tree nuts, fish and shellfish allergies which may develop later in childhood or adulthood. Most allergies (including CM allergies) resolve spontaneously during childhood or adolescence, whereas peanut and tree nut allergies are more likely to persist into adulthood [6,7].

2. Cow’s Milk Composition

CM contains from 30 to 35 g of protein per liter and many proteins, which are all potential allergens [8].
Through the acidification of raw skim milk to pH 4.6 at 20 °C it is possible to obtain two different fractions: the coagulum, containing the casein proteins which account for 80% of the fraction, and the lactoserum (whey proteins), representing 20% of the total milk proteins [9]. These proteins can also be divided into soluble and insoluble proteins [10].
The major milk allergens are soluble proteins, named whey proteins, which represent approximately 20% of total proteins [11]. Allergens present in the serum fraction include α-lactalbumin (Bos d 4) and β-lactoglobulin (Bos d 5), which are the most abundant, and immunoglobulins (Bos d 7), serum albumin (BSA, Bos d 6), and traces of lactoferrin, lysozyme, proteose-peptone, and transferrin [9]. In particular lactoferrin, lactoperoxidase, and lysozyme are important antimicrobial agents, while lactoferrin, β-lactoglobulin, and α-lactalbumin are important tumor suppressors [12].
The remaining 80% is represented by insoluble proteins known as caseins (known as Bos d 8). Total caseins can be divided into four proteins, representing different percentages of the whole fraction: αS1-casein, which is the most important (Bos d 9, 32%), as well as αS2-casein (Bos d 10, 10%), β-casein (Bos d 11, 28%), and κ-casein (Bos d 12, 10%). The main role of caseins relates to their mineral binding and carrier capacity, specifically for calcium and phosphorus [12].
CMAs are most frequently caused by whey proteins, but they can also be promoted by caseins [13]. As a matter of fact, most patients are sensitized to casein (Bos d 8), β-lactoglobulin (Bos d 5), and α-lactalbumin (Bos d 4), which are the major milk allergens. There are only a few studies that describe allergies to minor serum proteins such as immunoglobulin, bovine serum albumin, or lactoferrin [14].
CM contains at least 20 potentially allergenic proteins [15]. Most children with milk-allergy are sensitized to more than one allergen, with a greater variability of symptoms.
Table 1 provides a short synthesis of cow’s milk allergens and their characteristics.

3. Subtypes of Immune-Mediated Reactions to CM

CMAs have a range of clinical manifestations, with variable intensity. Moreover, clinical features can differ from “immediate” to “delayed” reactions, and this reflects the different pathogenesis (Figure 1).
Approximately 60% of CMA are IgE-mediated, although estimates change according to the study population and age [1].
The remaining 40% is divided into non IgE-mediated and mixed forms. The latter have different underlying mechanisms, presentations, and implications, which complicate the attempts to estimate the prevalence of CMA.
Non IgE-mediated CMAs are caused by less clear immune mechanisms. In this case, clinical findings are deferred and may occur 48 h or days after CM ingestion. Moreover, there are no specific symptoms or biomarkers that can guide the diagnosis, making it difficult to reach a conclusion. Typical non-IgE-mediated forms of CMA include CM enteropathy, food protein-induced proctitis/proctocolitis (FPIAP), food protein-induced enterocolitis syndrome (FPIES), and Heiner syndrome (pulmonary hemosiderosis) [1].
There are also mixed forms of CMA (both IgE- and non-IgE-mediated) that may have either humoral and/or cell-mediated mechanisms and may present with acute and/or chronic symptoms. These include atopic dermatitis, allergic eosinophilic esophagitis, and eosinophilic gastritis [1].

4. Prevalence of CMA

Before 1950, CMA was rarely diagnosed. Since 1970, significantly varying estimates of the incidence have been reported (ranging from 1.8% to 7.5%), reflecting the differences in diagnostic criteria and study design [16].
As a matter of fact, the prevalence estimates of CMA are affected by many factors. There is a marked heterogeneity in the prevalence of FA in the majority of papers. This could be the result of misleading differences in study design or methodology (including use of different definitions of CMA), or differences between populations and geographic areas [17].
Alongside the IgE, non-IgE, and mixed forms, there are also non-immune mediated reactions (i.e., intolerance), which are sometimes misclassified.
Another confounding factor while assessing prevalence is that many studies come from self-reports, with the consequent limitations linked to the subjective nature of the data [17]. Actually, the majority of studies on the epidemiology or natural history of FA have limitations. A precise evaluation requires a prospective ascertainment with a confirmatory oral food challenge (OFC) at predetermined intervals over time. For these reasons, studies such as these are rarely conducted due to their intrinsically reduced feasibility and ethical issues [17].
Therefore, determining an accurate diagnosis of CMA is fundamental. The process begins with an allergy-focused history that will guide all further investigations. In case that personal history is suggestive of allergy, a skin prick test (SPT) or specific IgE (sIgE) blood assay should be performed.
Evidence of sensitization (positive SPT or sIgE) with a suggestive history is usually sufficient to confirm the diagnosis, although OFC remains the gold standard [18].
It is important to emphasize that sensitization, i.e., raised sIgE directed against a specific antigen or positive SPT, in the absence of a supporting clinical history, is common in the general population but insufficient for a diagnosis of CMA. If diagnostic uncertainty remains even after a focused history and SPT/sIgE, an OFC is recommended to confirm diagnosis [18].
Other issues that may affect the estimates are the study design (prospective cohort vs cross-sectional), study population (demographic factors, geography, genetic/environmental factors), and natural history (incomplete identification of resolved cases) [1].
Despite these limitations during the assessment, a large number of studies in the United States and worldwide have attempted to estimate the prevalence of CMA [1].
An important contribution to prevalence studies was made by a meta-analysis performed by Rona et al., who analyzed publications from 1990 to 2005 and included only original studies (a total of 51 papers were considered appropriate for inclusion). The prevalence of self-reported FA was very high compared to that obtained using objective measures. Self-reported prevalence of CMA varied from 1.2% to 17%. The prevalence of CMA using SPT alone and sIgE alone, instead, was from 0.2% to 2.5% and from 2% to 9%, respectively. Studies using symptoms and sensitization (SPT ≥ 3 mm or sIgE > 0.35 kU/L) ranged from 0% to 2% prevalence, and those relying on OFC from 0% to 3% [19].
Another important meta-analysis and systematic review of CMA prevalence in Europe was performed by Nwaru et al., who analyzed publications from 2000 to 2012 (including 42 papers) [20]. The prevalence of self-reported CMA was 2.3% (95% CI 2.1–2.5), greater than that using SPT alone (0.3%, 95% CI 0.03–0.6) and sIgE alone (4.7%, 95% CI 4.2–5.1). The prevalence of CMA diagnosed by OFC was 0.6% (95% CI 0.5–0.8) and that using OFC or reported history of CMA was 1.6% (95% CI 1.2–1.9).
These meta-analyses show that prevalence estimates can be influenced by many factors such as geographic region, source population (high risk of referral bias vs general population), age and participation rates, and limitations of diagnosis [6].
Another important contribution was the EuroPrevall birth cohort study, published in 2015. In this study 12,049 children from nine different countries were enrolled, and 9336 (77.5%) were followed up until the age of 2 years. The authors calculated an overall incidence of challenge-proven CMA of 0.54% (95% CI 0.41–0.70) and showed differences in national incidences ranging from 1% (in the Netherlands and United Kingdom) to <0.3% (in Lithuania, Germany, and Greece). In this unique cohort study, they also showed that affected infants, without detectable specific antibodies to CM, were very likely to tolerate CM 1 year after diagnosis, whereas only half of those with specific antibodies in serum overcame their disease in the same period of time [21].

5. Diagnosis

Establishing an early and certain diagnosis of CMA is important to initiate the elimination diet in the case the diagnosis is confirmed or to avoid unnecessary dietary restrictions when it is not [22].
The diagnosis of CMA begins from the onset of signs and symptoms of CMA. Diagnosis is based on the combination of clinical history and physical examination, allergy tests such as sIgE and SPT, and, when indicated, OFC.
The first step is to evaluate the family and personal history in order to analyze every sign and symptom of the patient. Successively, it is fundamental to perform a differential diagnosis through laboratory evaluation and physical examination [23].
In addition to physical examination, there are objective assays used routinely in both epidemiological studies and clinical practice to investigate the condition, including sIgE and SPT. Despite the usefulness of sIgE antibodies (for tissue-bound and circulating IgE antibodies), these tests cannot differentiate between sensitization alone and clinical allergy [22]. The union of an evident history of allergic symptoms after CM exposure, associated with evidence of sensitization, certainly helps to make a definite diagnosis.
It has been demonstrated that the greater the food sIgE levels are and the SPT wheal size is, the higher the chances that the patient will manifest adverse reactions during an OFC. Numerous papers have analyzed the possibility of establishing a cutoff for sIgEs and SPTs for CM and its proteins that could predict whether a patient would react to an OFC [24]. Actually, several studies showed that cutoffs can vary with age [25], and many researchers are attempting to recommend diagnostic cutoffs for children [26,27]. However, cutoffs may change among different studies because of the type of allergen used to perform SPTs (commercial extract vs raw milk) or because of the degree of cooking [24].
Methods using sIgE measured [28] with in vitro immunoassays are still commonly referred to as IgE radioallergosorbent tests (RAST), and identify the level of IgE binding to specific proteins. Many studies have proposed a range of predictive cutoff values for the diagnosis of CMA, demonstrating a lack of agreement among different centers (Table 2).
Predictive cutoff values are found to be lower in younger children and increase with age [32], with diagnostic cutoff values remaining valid independently of the total serum IgE [37].
Therefore, it is difficult to assess standardized cutoffs for CM sIgE above which an OFC would not be required. Each patient would consequently need to be evaluated individually.
Traditionally sensitization is defined as the observation of a detectable sIgE level, (often sIgE > 0.35 kU/L but sometimes >0.10 kU/L) [1], although an OFC would be required if the sIgE level is positive but low.
SPTs have been used for decades to demonstrate or to exclude sensitization to allergens, as they are easy to perform, inexpensive, well tolerated, and provide immediately available results [22]. Traditionally, measured sensitization is often defined as a wheal at least 3 mm larger than the negative control [1]. Therefore, many studies aimed to avoid OFC by finding a cutoff of SPT able to predict a positive outcome of OFC [38] (Table 3).
A recent review by Cuomo et al. [24] reported that none of the cutoffs proposed in the literature could be used to definitively diagnose CMA. However, they found that in children aged <2 years, CMA diagnosis seemed to be highly likely when the sIgE reaction to CM extract was ≥5 kU/L, or when SPTs reactions with a commercial extract were above 6 mm, or when prick-by-prick reactions with fresh CM were above 8 mm [24].
However, if the clinical history is uncertain, SPT wheals measuring between 3 and 5 mm may be clinically irrelevant, and low levels of sIgE may be found in children without clinical CMA [33].
The negative predictive value of SPTs and sIgE is excellent (>95%) for immediate reactions [33]. Therefore, despite negative IgE tests, if there is a strong suspicion of CMA, an OFC is necessary to confirm the absence of a clinical allergy [13].
Despite this, even if OFC still remains the gold standard for CMA diagnosis (particularly the DBPCFC) [1], it is rarely required in clinical practice.
The OFC in CMA is performed by using baked or fresh milk. As baked milk is less allergenic in a context where a positive challenge is unexpected, it may be used initially because reactions are less likely to be severe [22].
OFCs should be performed under medical supervision in a hospital setting with an emergency kit available, especially in case of positive SPT or sIgE to CM and in infants at risk of an immediate reaction [13].
There is a lot of interest among clinicians in identifying markers that can predict the chance of developing tolerance and hence overcoming the allergy. Many studies have showed that IgE levels, expressed either as SPT wheal size or serum sIgE level, could be useful in discriminating between children who remained hypersensitive and those who became tolerant [44,45].The aim of most of these studies was to determine whether the monitoring of food sIgE levels over time could aid in the prediction of when patients would develop clinical tolerance. The likelihood estimates founded in these studies could help clinicians in providing prognostic information and in timing subsequent food challenges, thereby decreasing the number of premature and unnecessary DBPCFC [46].
Even if there is a lack of studies in this regard, some markers that may predict a persistent CMA have been identified over time. This explains the great interest of clinicians in finding a marker.
As a general rule, higher maximum IgE levels are associated with a reduced likelihood of developing tolerance [47].
Expressly regarding CMA, the association between increasing levels and persistence of allergy has been demonstrated, whereas decreasing levels indicate a faster recovery [48]. Low levels of IgG4 to β-lactoglobulin, instead, were found in children who required a longer elimination diet [36].
Vanto et al. [44] found that SPT wheal size <5 mm at diagnosis could be used to correctly identify 83% of individuals who developed tolerance at 4 years, whilst a wheal size ≥5 mm correctly identified 74% with persistent CMA.
These cutoff levels vary from study to study, possibly because of differences between the studied groups.
Garcia-Ara et al. [32] showed that sIgE levels predictive of clinical reactivity increased with age.
Shek and colleagues reported that the rate of reduction in food sIgE levels over time was predictive of the likelihood of developing tolerance in milk allergy. They were able to elaborate estimates of developing tolerance based on the reduction in sIgE levels, with a probability of tolerance of 31% in the case of a decrease by 50% in IgE levels, a probability of 45% in the case of a decrease by 70%, a probability of 66% when the decrease was 90%, and a probability of 94% for a decrease by 95% [46].
The value of sIgE has been analyzed with the aim of providing individuate cutoff levels related to the development of tolerance. Sampson et al. reported a positive predictive value of 95% in children with a median age of 3.8 years and CM-sIgE ≥ 15 kU/L [31]. Yavuz et al. examined sIgE levels which could predict a negative OFC in infants at different ages: by sIgE < 2.8 kU/L for children under 1 year, by 11.1 kU/L for children aged <2 years, and by <13.7 KU/l for children aged <6 years [49].

6. Risk Factors for CMA

The development of all FAs is influenced by genetics, environment, and genome–environment interactions, including epigenetic effects. Numerous risk factors for CMA have been identified or proposed that can contribute to allergy or sensitization.
There are unchangeable risk factors associated with a higher risk of FA, such as sex (male sex in children), race/ethnicity (increased among Asian and black children compared to white children), and family history of atopy [6]. It is generally presumed that atopy in parents increases atopic risk for the developing infant. This latter is one of the strongest risk factors, as occurs in other atopic diseases.
Koplin et al. discovered in a population of one-year-old infants with FA that the risk of FA increased to 40% in patients with one immediate family member with any allergic disease and to 80% in patients with two immediate family members with any allergic disease as compared to children no family history of allergy [50].
Controversy still exists as to whether a family history of atopy is also associated with a higher risk of CMA in infants. Goldberg et al., in a population-based study in 2013 [51], compared the parental atopic status of children with IgE-CMA (n = 66) with a group of healthy infants (n = 156). They reported no significant differences between the two groups and concluded that parental history of atopy alone cannot be used to anticipate which infants are at greater risk of developing IgE-mediated CMA [51].
On the contrary, in a recent paper by Sardecka et al. [52] on 138 infants with CMA and 101 healthy infants without allergy (with CMA confirmed by an elimination test and OFC), it was reported that the incidence of CMA was three times higher in infants with a positive family history for allergy. In this study it was also found that mothers of children with CMA were four times more likely to have a university-level education as compared to mothers of children without allergy [52].
As with all FAs, it is well known that atopic disease in general and particularly atopic dermatitis is an important risk factor for IgE-mediated CMA. This explain why the suspicion of CMA should be stronger in moderate-to-severe atopic dermatitis that starts in the first 6 months of life [53].
Atopic comorbidities such as asthma, especially when inadequately handled, are associated with frequent and severe reactions to milk [54]. Actually, it is still not known whether this is caused by a more severe allergic phenotype or by a barrier function.
Another factor that may have a protective role with respect to food sensitization and allergy later in childhood is increased food diversity in infancy [55]. In fact, Roduit et al. reported that an increased diversity of food within the first year of life might have a protective effect with respect to asthma, FA, and food sensitization [55].
Another risk factor for FA that has been identified is parents’ country of origin. The NHANES study, conducted between 2005 and 2006, compared the risk of food sensitization between US-born children and foreign-born children.
Compared to those born outside the United States, US-born children and adolescents had higher risk of sensitization to any food. Among the foreign-born, those who arrived before 2 years of age had higher odds of food sensitization than those who arrived later [56].
Other potential risk factors that can be examined to reduce/prevent FAs, are: increased hygiene, the influence of the microbiome [57], dietary fat (reduced consumption of omega-3-polyunsaturated fatty acids), reduced consumption of antioxidants, increased use of antacids (reducing digestion of allergens), obesity (being an inflammatory state), and the timing of food introduction in diet (increased risk of delaying oral ingestion of allergens, with environmental exposure, in the absence of oral exposure, leads to sensitization and allergy) [6].
A National Academies of Sciences report (2016) examined many factors and theories suggested to influence the risk of FA [58]. This group examined the “dual allergen exposure hypothesis”, attributed to Gideon Lack, and assessed that there is limited but consistent evidence that an impaired skin barrier plays a role in sensitization as a first step toward FA [6]. This theory suggest that low-dose cutaneous exposure is sensitizing and facilitated by an impaired skin barrier and inflammation. Meanwhile, oral exposure could cause the development of tolerance [6].
Numerous perinatal factors can influence the development of CMA and FAs, but the relationship between them is still controversial [59,60]. Sardecka et al. reported an increased risk of CMA in premature newborns, as previously noted [61], which may result from increased intestinal permeability [52]. The mode of delivery may also influence the development of FAs. The incidence of CMA may be higher in infants born by cesarean section because of the influence on the microbiota and, consequentially, on the immune system. Actually, no relationship between CMA and the type of delivery has ever been observed in studies [52,62].

6.1. The Role of Vitamin D

During the last decades, interest in the role of vitamin D in allergic disease has progressively increased. Numerous papers have suggested its possible role as an immune modulator in allergies, especially with respect to lymphocyte activation, antigen receptor function, and signaling pathways [63]. Still, the precise molecular mechanisms involved in vitamin D’s genomic and non-genomic actions remain incompletely defined [64] and not fully understood.
A lack of vitamin D has been associated with an increased risk of FA in many papers [64,65]. Actually, these associations are controversial and need further exploration, as vitamin D sufficiency has also been associated with an increased risk of allergic sensitization.
It is known that populations with lower levels of vitamin D are more susceptible to developing food allergies [66].
An Italian cross-sectional study performed by Lombardi et al. showed that the association between vitamin D levels and allergies was weak, and reported that it was necessary to implement studies involving larger samples to better assess this association [67].
Ecological studies have shown that there is an association between lower sunlight exposure and FA [68].
On the other hand, other papers reported that higher levels of vitamin D could increase the risk of allergic sensitization and FA.
For this reason, the relationship between vitamin D and development of FA is still controversial. Further studies are required to assess the role of vitamin D in the prevention of allergic diseases [68].

6.2. The Role of Breastfeeding

It is known that breastfeeding has an important role in the establishment of gut microbiota, nutritional status (with a role in preventing obesity and other nutritional disorders), the immunological system, and neuro-psychomotor development.
Human milk is composed of many molecules with potential immune-modulating roles: antibodies, predominantly secretory immunoglobulin A (s-IgA), cytokines (TGF-β, IL-10, IL-12, thymic stromal lymphopoietin) and chemokines, hormones and growth factors, polyunsaturated fatty acids (PUFAs), glutamine and dietary nucleotides, glycoproteins, oligosaccharides, and microRNA [69,70]. During infancy, it also represents the principal source of protein, fat, calcium, phosphorus, and vitamin B12. For this reason, breastfeeding should not be easily eliminated but encouraged instead [71].
According to the World Health Organization (WHO), breastfeeding is the perfect method of infant feeding; it is recommended exclusively in the first 6 months of life and partially until 2 years of age [72]. Exclusive breastfeeding means that infants should be fed only with breastmilk in the first 6 months of life and receive no other liquid or solids except for vitamins, mineral supplements, or medicines.
The main allergens in CM are casein, α-lactalbumin, and β-lactoglobulin. It is known that casein and α-lactalbumin are natural elements of human milk, while β-lactoglobulin is not present in it [15]. Therefore, the presence of β-lactoglobulin in human milk is caused by maternal ingestion of CM. Actually, it is still not understood whether there is a transfer of proteins such as β-lactoglobulin into breast milk. In any case, it seems that a small fraction of dietary proteins can resist digestion and become eventually allergenic [73].
Current guidelines expressly affirm that in infants with CMA, mothers should be encouraged to continue breastfeeding [69].
As suggested by American Academy of Pediatrics (AAP) and ESPGHAN recommendations, breastfeeding should be continued while solid foods are introduced into the diet [74]. From an allergological standpoint, continuing breastfeeding during solid food introduction and delaying this introduction until at least 17 weeks of age were associated with fewer FAs [75]. Thus, the early introduction of allergenic foods while breastfeeding might be a protective factor against FAs.
As recommended by the AAP, when breastfeeding is not possible or not sufficient, the introduction of CM proteins can be done in the first days or weeks of life through a CM formula [76].
However, the role of early exposure to CM proteins as a risk factor for the development of CMA and FA in general is still not clear. Data regarding a direct relationship between breastfeeding and FA are insufficient [77].
Exclusive breastfeeding for at least 4–6 months was first recommended by the European Section of Pediatrics and the AAP in the early 2000s to prevent FA and CMA in early childhood [78].
In 2008, the AAP affirmed that there was still no convincing evidence that the delayed introduction of allergenic foods beyond 4–6 months had a significant protective effect against the development of allergic disease [78].
A randomized trial study by Saarinen et al. [79] reported that feeding with CM formula in maternity hospitals increased the risk of CMA when compared with feeding with other supplements. However exclusive breastfeeding does not totally eliminate the risk.
Another prospective study on CMA in exclusively breastfed infants by Host et al. [80] underlined that early accidental and occasional exposure to CM proteins may initiate sensitization in predisposed newborns. Subsequent exposure to small amounts of CM proteins in human milk may act as booster doses by eliciting allergic reactions [80].
In a 2018 study, Sardecka et al. [52] found that the risk of CMA in children during the first year of life decreased as a result of a longer period of breastfeeding.
On the contrary, in a prospective large birth cohort study of 13,019 infants, Katz et al. reported that only 0.05% of the newborns who received a regular introduction of CM formula within the first 14 days developed CM-IgE, whereas 1.75% of the infants who started the CM formula between the ages of 105 and 194 days developed CM-IgE [81]. Thus, the authors concluded that early exposure to CM, in addition to breastfeeding, might stimulate tolerance.
Furthermore, a recent case—control study by Onizawa et al. [82] supported the hypothesis that early, regular, and continuous exposure to CM formula, started within the first month of life, can prevent CMA.
Further studies are required in order to confirm the possibility of preventing CMA with regular and early administration of CM formula.

7. Natural History of CMA

This paper focuses on CMA as a model of FAs in children.
The rate of resolution of FAs was reviewed by Savage et al. in 2016 [7]; it was found that some allergies have a high rate of resolution in childhood, such as milk (>50% by age 5–10 years), egg (approximately 50% by age 2–9 years), wheat (50% by age 7 years) and soy (45% by age 6 years) allergies, with complete resolution in adolescence. Others that usually persist include peanut (approximately 20% by age 4 years), tree nut (approximately 10%), fish, and shellfish allergies (further studies are necessary).
The natural history of CMA is unique. Resolution is usually common [1] and this contributes to the complication of prevalence estimates.
Actually, there is some heterogeneity in the estimated rate of resolution: studies show different resolution rates (Table 4). Whereas until now there has been a clearly notable heterogeneity among studies, in a combined analysis of patients with CMA in infancy, by the age of 5, 50% developed tolerance and by early adolescence, 75% developed tolerance [1].
A large population cohort study in Israel showed that only 57% of children with CMA resolved their allergy by the age of 4–5, and the majority of these allergies were resolved by age 2 [83].
In contrast, it is important to underline that clinically based studies suggest a poorer prognosis, since they include children who are at higher risk of allergy [17].
Santos et al., in a recent prospective study analyzing Portuguese children with CMA between 1997 and 2006, found that only 41% developed tolerance at the age of 10 [47].
Another important retrospective study of milk allergy by Skripak et al. [84] evidenced that the median age of outgrowing CMA was 10 years. In this case the definition of the development of allergy was having sIgE for milk <3 kUA/L or passing an OFC. Despite important heterogeneity among the studies (CMA diagnosis, study population, clinical features of CMA), it seems that more recent studies have shown less optimistic results, with lower rates of resolution.
It is accepted that 87% of children outgrow CMA by age of 3 [85], but the percentage of patients with persistent allergy has increased, although it was found to vary among publications. This may suggest that the natural history of CMA is changing over time.
Understanding these overall trends and the reasons for the variation has important implications for management and treatment.
Table 4. Natural history of CMA in different populations and settings, adapted from Mousan et al. [2].
Table 4. Natural history of CMA in different populations and settings, adapted from Mousan et al. [2].
Authors/Year of PublicationNumber of SubjectsPopulation/Study DesignTolerance RateAge of Tolerance(year)
Høst et al., 2002 [86]39 (24 IgE-mediated)General prospective birth cohort56%1
77%2
87%3
92%5
97%15
Vanto et al., 2004 [44]162 (95 IgE-mediated)Referral retrospective44%2
69%3
77%4
Garcia-Ara et al., 2004 [32]66 IgE-mediatedReferral retrospective68%4
Saarinen et al., 2005 [85]118 (75 IgE-mediated)General prospective birth cohort51%2
74%5
85%8.6
Skripak et al., 2007 [84]807 IgE-mediatedReferral retrospective19%4
42%8
64%12
79%16
Fiocchi et al., 2008 [87]112 IgE-mediatedReferral retrospective52.7%5
Martorell et al., 2008 [34]170 IgE-mediatedReferral retrospective82%4
Santos et al., 2010 [47]139 (66 IgE-mediated)Referral retrospective41%2
Ahrens et al., 2012 [88]52 IgE-mediatedReferral retrospective61.5%12
Elizur et al., 2012 [83]54 IgE-mediatedGeneral prospective birth cohort57.4%2
65%4
Wood et al., 2013 [89]293 IgE-mediatedProspective53%5.5
Yavuz et al., 2013 [49]148 IgE-mediatedReferral retrospective20%2
34%4
39%6
Schoemaker et al., 2015 [21]55EuroPrevall, European population-based57%2
Prospective

8. Factors Associated with the Natural History

Many factors have been associated with the natural history of FA with respect to both the development of tolerance and its persistence. Most of these markers have been studied for CMA.
Early identification of patients in which CMA is likely to persist will provide the clinician with useful insights about when and how to start CM reintroduction [22].
As a general rule, non-IgE-mediated allergy resolves more rapidly than IgE-mediated [85].
First of all, it is known that persistent allergy has been connected with more severe clinical features at presentation. Secondly, persistent FA has been linked with an earlier age at diagnosis [83], the presence of other atopic diseases and their severity (allergic rhinitis, asthma, eczema) [49], the presence of other FAs (most commonly egg allergy [49,59]), and a lower threshold dose to trigger a reaction [7]. In addition, reactivity to baked milk (BM) on first exposure is also associated with the persistence of CMA [90].
The natural history of FA varies substantially, and it is known that the variation of food-sIgE or SPT wheal size in time can predict the probability of resolution and the development of tolerance [48]. Generally speaking, larger wheal size on SPT or higher sIgE levels are associated with persistence of FA [89,91]. These associations are consistent with findings from Kim et al. showing that higher sIgE levels at first reaction were the most significant predictors of persistent CMA [92].
Moreover, a strong relation between the food allergen and the persistence of FA has been demonstrated. Some FAs, such as those to milk and egg, have a high likelihood of resolution, whereas sufferers of other allergies such as those to peanut and tree nuts have lower probability of developing tolerance. To sum up, when approaching FA, the clinician should take into account the type of allergen involved, together with SPT results and sIgE levels. Yearly follow-up with repetition of sIgE should be advisable until complete stabilization, as this has a higher prognostic value in predicting tolerance acquisition and the likelihood of passing an OFC [7,93].

The Role of Baked Milk

The baking process alters protein epitopes which are no longer recognizable by the epitope-specific IgE, leading to decreased allergenicity [1].
Regarding CM, whey proteins such as α-lactalbumin and β-lactoglobulin include conformational epitopes that are heat-labile, whereas casein contains mostly heat-resistant epitopes [94].
As a consequence, BM-driven reactions are indicative of more persistent CMA.
To date, only few papers have compared the development of tolerance to BM as compared to fresh milk, although many retrospective studies have supposed a quicker CMA resolution upon regular ingestion of BM [95].
In a recent study conducted in infants under the age of 2 years, Uncuoglu et al. found that 81% of children with IgE-mediated CMA were baked-milk tolerant [96].
Nowak-Wegrzyn et al., in their study based on 100 children with documented CMA, showed that 75% tolerated BM and were able to include BM products into their diet. After 3 months of BM ingestion, children had significantly smaller SPT wheals as compared to baseline [97].
Kim et al. [92] reported that tolerance to BM products was an advantageous prognostic factor for the development of tolerance to unheated milk. Moreover, the ingestion of BM products seemed to considerably accelerate the development of tolerance as opposed to a strict avoidance diet.
A controlled randomized clinical trial by Esmaeilzadeh et al. further confirmed that regular assumption of BM products accelerated the tolerance to fresh milk [98].
In conclusion, tolerance to BM usually precedes tolerance to fresh milk and represents a reliable predictor for the less severe and persistent CMA phenotype [94].

9. Treatment and Oral Immunotherapy

Since the 1930s, scientific literature on CMA has continuously increased. In recent decades, new diagnostic methods and therapies have been developed [99].
From the beginning, the cornerstone of treatment was represented by an elimination diet with strict avoidance of the offending food and its substitution, when possible, with hydrolyzed formulas [100]. However, despite rigid adherence to diet, CM can be inadvertently ingested through processed foods.
In support of this statement, Alonso et al. followed 80 patients with CMA [101] until the achievement of tolerance or up to the age of 18 years (in the case of other allergic diseases), finding accidental ingestion of milk in at least a third of them [102]. This can lead to a feeling of insecurity in children and their parents, significantly worsening their QoL [103].
Given that the avoidance diet as the preferred approach towards CMA has demonstrated limited efficacy, finding a new targeted therapy is pivotal. In response to this need, a novel treatment known as oral immunotherapy (OIT) has emerged through the last decade, changing the history of CMA and FA in general.
Oral tolerance induction testing was first attempted in laboratory animals in 1909 by Besredka, who demonstrated that oral or rectal administration of CM could protect from clinical manifestations such as anaphylaxis [104].
Before this new treatment, patients had to strictly avoid the food allergen, with the consequent reduction in QoL as well as nutritional, social, and psychologic impairment [105].
Nowadays, OIT is the most promising approach for the management of FA [106]. It consists of repeated administration of increasing amounts of the food allergen until reaching a target dose, in order to provide protection against the clinical features and inflammation [107]. Once the target is achieved, the patient must maintain a regular intake of the allergen to preserve the state of desensitization [108]. In fact, “desensitization” refers to a reversible state in which patients can eat higher doses of the food allergen without symptoms as compared with pre-treatment doses [109]. On the other hand, “tolerance” is the ability to introduce the allergen without any adverse reaction once therapy is completed.
Thus, the aim of OIT is to introduce allergenic food into the normal diet, or, in high-risk patients, to prevent life-threatening conditions after inadvertent ingestion [107]. In those patients, OIT is given at a lower dose in order to avoid severe reactions after accidental exposure [107].
OIT is potentially indicated for children with evidence of IgE-mediated CMA and in whom avoidance diet is ineffective, undesirable, or decreases QoL [110]. According to EAACI Guidelines [110], OIT is recommended for persistent CMA for children from around 4 to 5 years of age in order to increase the threshold for clinical manifestations of allergy. Restricting OIT in this age group can be explained by the fact that achievement of tolerance occurs before school age.
OIT has an immunological role in the modulation of humoral and cellular immunity. In particular, humoral changes such as a decrease in IgE levels and a rise in IgG levels, mainly IgG4, have been described. IgG4 could have an antigen-neutralizing effect and decrease basophil and mast cell activation, with the suppression of IgE production [107]. Moreover, OIT drives a reduction in Th2 cell line and Th2 cytokine expression [95].
OIT can be divided into two different phases. The first is the so-called “induction phase” or “dose escalation phase” where the main target is to achieve the tolerated dose [111]. It starts with a small dose, usually in micrograms of allergenic proteins which do not cause a reaction, and continues until the achievement of a target dose or until symptoms preclude further increments [112]. The allergen dose is augmented once or twice a week until reaching a maintenance dose. There are many protocols differing in the amounts of time required: from flash protocols (one week) to slow protocols (>6 months) [107].
Furthermore, the initial dose needs to be adapted to the patient following a personalized medicine approach. For example, low doses should be used in high-risk patients, while a more rapid introduction of the allergenic food could be performed in low-risk patients [113].
The second phase is a “maintenance phase” characterized by repeated intake of maximum tolerated doses of CM [114].
Whether the induced tolerance by OIT is permanent or transient, the long-term effects are still unclear. In agreement with the most recent available studies, not all the children subjected to OIT are able to introduce normal amounts of CM in their diet. Thus, OIT substantially increases the threshold dose necessary to elicit clinical manifestations, resulting in clinical tolerance in a large number of patients [115]. Permanent oral tolerance is reasonably achievable only in a part of the treated patients [116].
Table 5 summarizes the data from various studies on the efficacy of OIT milk.
To date, only few randomized studies have compared the efficacy of immunotherapy to elimination diet, underlining the need for further research with more homogeneous and wider populations.
In this regard, Staden et al. [112] described a population of 47 children with DBPCFC-confirmed CMA, randomly assigned either to OIT or elimination diet. The patients were clinically evaluated at baseline and after a period of 21 months, with repetition of an OFC to assess the persistence of tolerance. At the follow-up OFC, 36% and 35% of the patients in the OIT group and in the control group, respectively, achieved complete tolerance. Although similar results were observed, if we include children who achieved partial tolerance (patients who needed a regular intake of the allergen to maintain tolerance), the efficacy rate of OIT increased to 64% [112]. OIT resulted superior to the elimination diet, with an increased threshold dose for allergic reactions and a reduced burden of severe reactions following accidental ingestion of CM. In a randomized study by Morisset et al. [135], 60 infants with CMA were randomized into an OIT group and a control group treated with elimination diet group. OIT was proposed for those who did not react to the OFC (60 mL of milk). In this study, the rate of spontaneous recovery after 6 months in the control group (60%) resulted considerably lower as compared with that of the OIT group (88.9%). In addition, patients treated with OIT showed lower reactivity in terms of SPT size and lower sIgE levels [135], in accordance with the results of other studies [136].
A recent review with a meta-analysis on OIT in CMA by Martorell Calatayud et al. reported that rates of desensitization after OIT ranged from 36% to 77%, with an estimated tolerance close to 30% [28]. They concluded that this new strategy was an effective and reasonably safe alternative to the avoidance diet. Moreover, their study showed that significantly more patients achieved tolerance with OIT than with the elimination diet [28].
Although many studies highlighted the benefits of OIT, there is still a lack of well-conducted studies concerning the risk of side effects of this novel therapeutic approach.
Since several studies have reported side effect rates of 50–60% [117,136], increasing with exercise and the pollen season, continuous medical supervision during OIT is still mandatory. During OIT mild, localized, and self-limiting side effects usually occur, including oral itching and rhino-conjunctivitis. Symptoms that can lead to the discontinuation of OIT occur in just a small percentage of cases, and include abdominal pain (the most common), wheezing, laryngeal spasms, vomiting, and urticaria [137]. In fact, OIT is usually associated with a modest increase in risks of systemic side effects and a substantial increase in minor local adverse reaction [138]. Among various studies, anaphylactic reactions and the resulting intramuscular administration of epinephrine have been reported in 6.7% to 30.8% of all patients subjected to OIT [28].
The risk–benefit ratio of OIT is still debated. While the efficacy of this approach has been well studied, the evaluation of an effective improvement in the QoL of the patient remains limited to small or uncontrolled studies [139]. In fact, only few studies evaluate children’s perceptions of improved of their QoL as compared to perceptions by parents. A recent paper [139] described how OIT could improve the QoL of (both partially and totally desensitized) food-allergic children and their parents, through the Food Allergy Quality of Life Questionnaire—Child Form (FAQLQ-CF) and the FAQLQ—Parent Form (FAQLQ-PF), respectively. These results were concordant with those of Carraro et al., who gave the same questionnaire to parents of patients with CMA and found a substantial increase in all the investigated areas of QoL (emotional impact, food anxiety, and social and dietary limitations) in children with CMA [140].
Conversely, in 2021 Kauppila et al. reported that the Health-Related Quality of Life (HRQoL) among OIT patients did not differ significantly from that of the age- and gender-standardized general population [141].
In conclusion, OIT is an effective strategy for treating CMA which needs to be performed under the cautious supervision of an experienced specialist. The children who can obtain the greatest benefit from this therapy are those with high sIgE levels and high risk of life-threatening conditions such as anaphylaxis. For these patients, OIT should be considered as a possibility to avoid severe reactions and improve quality of life. However, OIT should be considered as an individualized treatment, and each step needs to be adapted to the specific patient [107].

10. Conclusions

CMA represents a model of FAs, since it is the most common and most studied FA in early life. For this reason, clearly understanding its epidemiology, diagnostic criteria, and appropriate treatment can guide the clinician and provide useful insights to better comprehend all other allergies. Ensuring proper diagnosis and prognosis and identifying the possibility of allergy resolution are therefore key components of management.
Thus far there are still no shared tolerance markers for the diagnosis of CMA and FAs in general. Although the negative predictive value of SPT and sIgE is excellent, the OFC, particularly DBPCFC, remains the gold standard for diagnosis. The heterogeneity of diagnostic tools used in literature further limits a reliable estimate of CMA epidemiology and consequentially of its natural history. Appropriately diagnosing CMA is therefore pivotal in understanding its natural history and avoiding unnecessary strict diets that may lead to nutritional deficiencies. In fact, milk is the most important element in children’s diets, providing the necessary intake of fats, proteins, calcium, phosphorus, and vitamin B12. Furthermore, the elimination diet is known to be possibly linked to an increased risk of severe reaction after the inadvertent ingestion of the allergen.
In the last decade there have been many changes in the approach to CMA, which has become proactive. OIT can indeed lead to a change in the natural history of this disease, accelerating tolerance acquisition and the likelihood of passing an OFC.
Therefore, further larger, well-designed, randomized, placebo-controlled trials are necessary to find new diagnosis, prevention, and treatment strategies.

Author Contributions

Conceptualization: A.G., G.T.V. and G.R.; Resources: A.G., G.T.V., A.M. and E.d.P.; Methodology: A.G., G.R. and A.P.; Writing—Original Draft Preparation: A.G., G.T.V., A.M., and E.d.P.; Writing—Review and Editing: A.G., G.T.V., G.R., and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Flom, J.D.; Sicherer, S.H. Epidemiology of cow’s milk allergy. Nutrients 2019, 11, 1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mousan, G.; Kamat, D. Cow’s milk protein allergy. Clin. Pediatr. 2016, 55, 1054–1063. [Google Scholar] [CrossRef] [PubMed]
  3. Hill, D.J.; Firer, M.A.; Shelton, M.J.; Hosking, C.S. Manifestations of milk allergy in infancy: Clinical and immunologic findings. J. Pediatr. 1986, 109, 270–276. [Google Scholar] [CrossRef]
  4. Lomer, M.C.; Parkes, G.C.; Sanderson, J.D. Review article: Lactose intolerance in clinical practice—Myths and realities. Aliment. Pharmacol. Ther. 2008, 27, 93–103. [Google Scholar] [CrossRef]
  5. Burks, A.; Tang, M.; Sicherer, S.; Muraro, A.; Eigenmann, P.A.; Ebisawa, M.; Fiocchi, A.; Chiang, W.; Beyer, K.; Wood, R.; et al. ICON: Food allergy. J. Allergy Clin. Immunol. 2012, 129, 129906. [Google Scholar] [CrossRef]
  6. Sicherer, S.H.; Sampson, H.A. Food allergy: A review and update on epidemiology, pathogenesis, diagnosis, prevention, and management. J. Allergy Clin. Immunol. 2018, 141, 41–58. [Google Scholar] [CrossRef] [Green Version]
  7. Savage, J.; Sicherer, S.; Wood, R. The natural history of food allergy. J. Allergy Clin. Immunol. Pract. 2016, 4, 196–203. [Google Scholar] [CrossRef]
  8. Hochwallner, H.; Schulmeister, U.; Swoboda, I.; Spitzauer, S.; Valenta, R. Cow’s milk allergy: From allergens to new forms of diagnosis, therapy and prevention. Methods 2014, 66, 22–33. [Google Scholar] [CrossRef]
  9. Wal, J.M. Bovine milk allergenicity. Ann. Allergy Asthma Immunol. 2004, 93, S2–S11. [Google Scholar] [CrossRef]
  10. Séverin, S.; Wenshui, X. Milk biologically active components as nutraceuticals: 546 review. Crit. Rev. food Sci. Nutr. 2005, 45, 645–656. [Google Scholar] [CrossRef]
  11. Haug, A.; Høstmark, A.T.; Harstad, O.M. Bovine milk in human nutrition—A review. Lipids Health Dis. 2007, 6, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pereira, P.C. Milk nutritional composition and its role in human health. Nutrition 2014, 30, 619–627. [Google Scholar] [CrossRef]
  13. Caffarelli, C.; Baldi, F.; Bendandi, B.; Calzone, L.; Marani, M.; Pasquinelli, P. Cow’s milk protein allergy in children: A practical guide. Ital. J. Pediatr. 2010, 36, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Restani, P.; Ballabio, C.; Di Lorenzo, C.; Tripodi, S.; Fiocchi, A. Molecular aspects of milk allergens and their role in clinical events. Anal. Bioanal. Chem. 2009, 395, 47–56. [Google Scholar] [CrossRef] [PubMed]
  15. Fiocchi, A.; Schünemann, H.J.; Brozek, J.; Restani, P.; Beyer, K.; Troncone, R.; Martelli, A.; Terracciano, L.; Bahna, S.L.; Rancé, F.; et al. Diagnosis and rationale for action against cow’s milk allergy (DRACMA): A summary report. J. Allergy Clin. Immunol. 2010, 126, 1119–1128.e12. [Google Scholar] [CrossRef]
  16. Host, A. Cow’s milk protein allergy and intolerance in infancy. Some clinical, epidemiological and immunological aspects. Pediatr. Allergy Immunol. 1994, 5, 1–36. [Google Scholar] [CrossRef]
  17. Savage, J.; Johns, C.B. Food allergy epidemiology and natural history food allergy epidemiology natural history peanut milk egg. Immunol. Allergy Clin. NA 2015, 35, 45–59. [Google Scholar] [CrossRef] [Green Version]
  18. Helyeh, S.; David, L.; Gary, S. Advances in the management of food allergy in children. Curr. Pediatr. Rev. 2018, 14, 150–155. [Google Scholar] [CrossRef]
  19. Rona, R.J.; Keil, T.; Summers, C.; Gislason, D.; Zuidmeer, L.; Sodergren, E.; Sigurdardottir, S.T.; Lindner, T.; Goldhahn, K.; Dahlstrom, J.; et al. The prevalence of food allergy: A meta-analysis. J. Allergy Clin. Immunol. 2007, 120, 638–646. [Google Scholar] [CrossRef]
  20. Nwaru, B.I.; Hickstein, L.; Panesar, S.S.; Roberts, G.; Muraro, A.; Sheikh, A. Prevalence of common food allergies in Europe: A systematic review and meta-analysis. Allergy Eur. J. Allergy Clin. Immunol. 2014, 69, 992–1007. [Google Scholar] [CrossRef]
  21. Schoemaker, A.A.; Sprikkelman, A.B.; Grimshaw, K.E.; Roberts, G.; Grabenhenrich, L.; Rosenfeld, L.; Siegert, S.; Dubakiene, R.; Rudzeviciene, O.; Reche, M.; et al. Incidence and natural history of challenge-proven cow’s milk allergy in European children—EuroPrevall birth cohort. Allergy Eur. J. Allergy Clin. Immunol. 2015, 70, 963–972. [Google Scholar] [CrossRef] [PubMed]
  22. Luyt, D.; Ball, H.; Makwana, N.; Green, M.R.; Bravin, K.; Nasser, S.M.; Clark, A.T. BSACI guideline for the diagnosis and management of cow’s milk allergy. Clin. Exp. Allergy 2014, 44, 642–672. [Google Scholar] [CrossRef]
  23. Kansu, A.; Yüce, A.; Dalgıç, B.; Şekerel, B.E.; Çullu-Çokuğraş, F.; Çokuğraş, H. Consensus statement on diagnosis, treatment and follow-up of cow’s milk protein allergy among infants and children in Turkey. Turk. J. Pediatr. 2016, 58, 1–11. [Google Scholar] [CrossRef] [Green Version]
  24. Cuomo, B.; Indirli, G.C.; Bianchi, A.; Arasi, S.; Caimmi, D.; Dondi, A.; La Grutta, S.; Panetta, V.; Verga, M.C.; Calvani, M. Specific IgE and skin prick tests to diagnose allergy to fresh and baked cow’s milk according to age: A systematic review. Ital. J. Pediatr. 2017, 43, 1–10. [Google Scholar] [CrossRef] [Green Version]
  25. Komata, T.; Söderström, L.; Borres, M.P.; Tachimoto, H.; Ebisawa, M. The predictive relationship of food-specific serum IgE concentrations to challenge outcomes for egg and milk varies by patient age. J. Allergy Clin. Immunol. 2007, 119, 1272. [Google Scholar] [CrossRef]
  26. Sampson, H.A.; Aceves, S.; Bock, S.A.; James, J.; Jones, S.; Lang, D.; Nadeau, K.; Nowak-Wegrzyn, A.; Oppenheimer, J.; Perry, T.T.; et al. Food allergy: A practice parameter update-2014. J. Allergy Clin. Immunol. 2014, 134, 1016–1025. [Google Scholar] [CrossRef]
  27. Nowak-Wegrzyn, A.; Assa’ad, A.H.; Bahna, S.L.; Bock, S.A.; Sicherer, S.H.; Teuber, S.S. Work group report: Oral food challenge testing. Adverse reactions to food committee of American academy of allergy, asthma & immunology. J. Allergy Clin. Immunol. 2009, 123 (Suppl. 6), S365–S383. [Google Scholar]
  28. Calatayud, C.M.; García, A.M.; Aragonés, A.M.; Caballer, B.D. Safety and efficacy profile and immunological changes associated with oral immunotherapy for IgE-mediated cow’s milk allergy in children: Systematic review and meta-analysis. J. Investig. Allergol. Clin. Immunol. 2014, 24, 298–307. [Google Scholar]
  29. Sampson, H.A.; Ho, G. Clinical aspects of allergic disease Relationship between food-specific IgE concentrations and the risk of positive food challenges in children and adolescents. J. Allergy Clin. Immunol. 1997, 100, 444–451. [Google Scholar] [CrossRef]
  30. García-Ara, C.; Boyano-Martínez, T.; Díaz-Pena, J.M.; Martín-Muñoz, F.; Reche-Frutos, M.; Martín-Esteban, M. Food and drug reactions and anaphylaxis Specific IgE levels in the diagnosis of immediate hypersensitivity to cow’s milk protein in the infant. J. Allergy Clin. Immunol. 2001, 107, 185–190. [Google Scholar] [CrossRef]
  31. Sampson, H.A. Utility of food-specific IgE concentrations in predicting symptomatic food allergy. J. Allergy Clin. Immunol 2001, 107, 891–896. [Google Scholar] [CrossRef]
  32. García-Ara, M.C.; Boyano-Martínez, M.T.; Díaz-Pena, J.M.; Martín-Muñoz, M.F.; Martín-Esteban, M. Cow’s milk-specific immunoglobulin E levels as predictors of clinical reactivity in the follow-up of the cow’s milk allergy infants. Clin. Exp. Allergy 2004, 34, 866–870. [Google Scholar] [CrossRef]
  33. Celik-Bilgili, S.; Mehl, A.; Verstege, A.; Staden, U.; Nocon, M.; Beyer, K.; Niggemann, B. The predictive value of specific immunoglobulin E levels in serum for the outcome of oral food challenges. Clin. Exp. Allergy 2005, 35, 268–273. [Google Scholar] [CrossRef]
  34. Martorell, A.; Plaza, A.M.; Nevot, S.; Echeverria, L.; Alonso, E.; Garde, J. The predictive value of specific immunoglobulin E levels in serum for the outcome of the development of tolerance in cow’s milk allergy. Allergol. Immunopathol. 2008, 36, 325–330. [Google Scholar] [CrossRef]
  35. Van der Gugten, A.C.; den Otter, M.; Meijer, Y.; Pasmans, S.G.A.M.; Knulst, A.C.; Hoekstra, M.O. Usefulness of specific IgE levels in predicting cow’s milk allergy. J. Allergy Clin. Immunol. 2008, 121, 531–533. [Google Scholar] [CrossRef]
  36. Ott, H.; Baron, J.M.; Heise, R.; Ocklenburg, C.; Stanzel, S.; Merk, H.F.; Niggemann, B.; Beyer, K. Clinical usefulness of microarray-based IgE detection in children with suspected food allergy. Allergy 2008, 63, 1521–1528. [Google Scholar] [CrossRef]
  37. Mehl, A.; Verstege, A.; Staden, U.; Kulig, M.; Nocon, M.; Beyer, K.; Niggemann, B. Utility of the ratio of food-specific IgE/total IgE in predicting symptomatic food allergy in children. Allergy 2005, 60, 1034–1039. [Google Scholar] [CrossRef] [PubMed]
  38. Onesimo, R.; Monaco, S.; Greco, M.; Caffarelli, C.; Calvani, M.; Tripodi, S.; Sopo, S.M. Predictive value ofMP4 (Milk prick four), a panel of skin prick test for the diagnosis of pediatric immediate cow’s milk allergy. Eur. Ann. Allergy Clin. Immunol. 2013, 45, 201–208. [Google Scholar] [PubMed]
  39. Eigenmann, P.A.; Sampson, H.A. Interpreting skin prick tests in the evaluation of food allergy in children. Pediatr Allergy Immunol. 1998, 9, 186–191. [Google Scholar] [CrossRef]
  40. Sporik, R.; Hill, D.J.; Hosking, C.S. Specificity of allergen skin testing in predicting positive open food challenges to milk, egg and peanut in children. Clin. Exp. Allergy 2000, 30, 1540–1546. [Google Scholar] [CrossRef]
  41. Calvani, M.; Alessandri, C.; Frediani, T.; Lucarelli, S.; Miceli Sopo, S.; Panetta, V.; Zappalã, D.; Zicari, A.M. Correlation between skin prick test using commercial extract of cow’s milk protein and fresh milk and food challenges. Pediatr. Allergy Immunol. 2007, 18, 583–588. [Google Scholar]
  42. Calvani, M.; Berti, I.; Fiocchi, A.; Galli, E.; Giorgio, V.; Martelli, A.; Miceli Sopo, S.; Panetta, V. Oral food challenge: Safety, adherence to guidelines and predictive value of skin prick testing. Pediatr Allergy Immunol. 2012, 23, 755–761. [Google Scholar] [CrossRef]
  43. Kido, J.; Hirata, M.; Ueno, H.; Nishi, N.; Mochinaga, M.; Ueno, Y.; Yanai, M.; Johno, M.M. Evaluation of the skin-prick test for predicting the outgrowth of cow’s milk allergy. Allergy Rhinol. 2016, 7, 39–143. [Google Scholar] [CrossRef]
  44. Vanto, T.; Helppilä, S.; Juntunen-Backman, K.; Kalimo, K.; Klemola, T.; Korpela, R.; Koskinen, P. Prediction of the development of tolerance to milk in children with cow’s milk hypersensitivity. J. Pediatr. 2004, 144, 218–222. [Google Scholar] [CrossRef] [PubMed]
  45. Sicherer, S.H.; Sampson, H.A. Cow’s milk protein-specific IgE concentrations in two age groups of milk-allergic children and in children achieving clinical tolerance. Clin. Exp. Allergy 1999, 29, 507–512. [Google Scholar] [CrossRef]
  46. Shek, L.P.C.; Soderstrom, L.; Ahlstedt, S.; Beyer, K.; Sampson, H.A. Determination of food specific IgE levels over time can predict the development of tolerance in cow’s milk and hen’s egg allergy. J. Allergy Clin. Immunol. 2004, 114, 387–391. [Google Scholar] [CrossRef]
  47. Santos, A.; Dias, A.; Pinheiro, J.A. Predictive factors for the persistence of cow’s milk allergy. Pediatr. Allergy Immunol. 2010, 21, 1127–1134. [Google Scholar] [CrossRef] [PubMed]
  48. Dupont, C. How to reintroduce cow’s milk? Pediatr. Allergy Immunol. 2013, 24, 627–632. [Google Scholar] [CrossRef]
  49. Yavuz, S.T.; Buyuktiryaki, B.; Sahiner, U.M.; Birben, E.; Tuncer, A.; Yakarisik, S.; Karabulut, E.; Kalayci, O.; Sackesen, C. Factors that predict the clinical reactivity and tolerance in children with cow’s milk allergy. Ann. Allergy Asthma Immunol. 2013, 110, 284–289. [Google Scholar] [CrossRef] [PubMed]
  50. Koplin, J.J.; Allen, K.J.; Gurrin, L.C.; Peters, R.L.; Healthnuts, T.; Team, S.; Ponsonby, A.; Hill, D.; Matheson, M.; Wake, M.; et al. The impact of family history of allergy on risk of food allergy: A population-based study of infants. Int. J. Environ. Res. Public Health 2013, 10, 5364–5377. [Google Scholar] [CrossRef]
  51. Goldberg, M.; Eisenberg, E.; Elizur, A.; Rajuan, N.; Rachmiel, M.; Cohen, A.; Zadik-Mnuhin, G.; Katz, Y. Role of parental atopy in cow’s milk allergy: A population-based study. Ann. Allergy Asthma Immunol. 2013, 110, 279–283. [Google Scholar] [CrossRef]
  52. Sardecka, I.; Los-Rycharska, E.; Ludwig, H.; Gawryjołek, J.; Krogulska, A. Early risk factors for cow’s milk allergy in children in the first year of life. Allergy Asthma Proc. 2018, 39, e44–e54. [Google Scholar] [CrossRef] [PubMed]
  53. Hill, D.J.; Hosking, C.S. Food allergy and atopic dermatitis in infancy: An epidemiologic study. Pediatr. Allergy Immunol. 2004, 15, 421–427. [Google Scholar] [CrossRef] [PubMed]
  54. Boyano-Martinez, T.; Garcia-Ara, C.; Pedrosa, M.; Diaz-Pena, J.M.; Quirce, S. Accidental allergic reactions in children allergic to cow’s milk proteins. J. Allergy Clin. Immunol. 2009, 123, 883–888. [Google Scholar] [CrossRef]
  55. Roduit, C.; Frei, R.; Depner, M.; Schaub, B.; Loss, G.; Genuneit, J.; Pfefferle, P.; Hyvärinen, A.; Karvonen, A.M.; Riedler, J.; et al. Increased food diversity in the first year of life is inversely associated with allergic diseases. J. Allergy Clin. Immunol. 2014, 133, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
  56. Keet, C.A.; Wood, R.A.; Matsui, E.C. Personal and parental nativity as risk factors for food sensitization. J. Allergy Clin. Immunol. 2012, 129, 169–175.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Savage, J.H.; Lee-Sarwar, K.A.; Sordillo, J.; Bunyavanich, S.; Zhou, Y.; O’Connor, G.; Sandel, M.; Bacharier, L.B.; Zeiger, R.; Sodergren, E.; et al. A prospective microbiome-wide association study of food sensitization and food allergy in early childhood. Allergy Eur. J. Allergy Clin. Immunol. 2018, 73, 145–152. [Google Scholar] [CrossRef] [PubMed]
  58. National Academies of Sciences, Engineering and Medicine. Finding a Path to Safety in Food Allergy: Assessment of Global Burden, Causes, Prevention, Management, and Public Policy; National Academies of Sciences: Washington, DC, USA, 2016. [Google Scholar]
  59. Miyazawa, T.; Itabashi, K.; Imai, T. Retrospective multicenter survey on food-related symptoms suggestive of cow’s milk allergy in NICU neonates. Allergol Int. 2013, 62, 85–90. [Google Scholar] [CrossRef] [Green Version]
  60. Morita, Y.; Iwakura, H.; Ohtsuka, H.; Kohno, Y.; Shimojo, N. Milk allergy in the neonatal intensive care unit: Comparison between premature and full-term neonates. Asia Pac. Allergy. 2013, 3, 335–341. [Google Scholar] [CrossRef]
  61. Edwards, M.O.; Kotecha, S.J.; Lowe, J.; Richards, L.; Watkins, W.J.; Kotecha, S. Early-term birth is a risk factor for wheezing in childhood: A cross- sectional population study. J. Allergy Clin. Immunol. 2015, 136, 581–587.e2. [Google Scholar] [CrossRef] [PubMed]
  62. Kvenshagen, B.; Halvorsen, R.; Jacobsen, M. Is there an increased frequency of food allergy in children delivered by caesarean section compared to those delivered vaginally? Acta Paediatr. 2009, 98, 324–327. [Google Scholar] [CrossRef] [PubMed]
  63. von Essen, M.R.; Kongsbak, M.; Schjerling, P.; Olgaard, K.; Odum, N.; Geisler, C. Vitamin D controls T cell antigen receptor signaling and activation of human T cells. Nat. Immunol. 2010, 11, 344–349. [Google Scholar] [CrossRef]
  64. Rochoutsou, A.I.; Kloukina, V.; Samitas, K.; Xanthou, G. Vitamin-D in the immune system: Genomic and non-genomic actions. Mini Rev. Med. Chem. 2015, 15, 953–963. [Google Scholar] [CrossRef]
  65. Poole, A.; Song, Y.; Brown, H.; Hart, P.H.; Zhang, G. (Brad) Cellular and molecular mechanisms of vitamin D in food allergy. J. Cell. Mol. Med. 2018, 22, 3270–3277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sharief, S.; Jariwala, S.; Kumar, J.; Muntner, P.; Melamed, M.L. Vitamin D levels and food and environmental allergies in the United States: Results from NHANES 2005–2006. J. Allergy Clin. Immunol. 2011, 127, 1195–1202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lombardi, C.; Passalacqua, G. Vitamin D levels and allergic diseases. An Italian cross-sectional multicenter survey. Eur. Ann. Allergy Clin. Immunol. 2017, 49, 75–79. [Google Scholar]
  68. Giannetti, A.; Bernardini, L.; Cangemi, J.; Gallucci, M.; Masetti, R.; Ricci, G. Role of Vitamin D in prevention of food allergy in infants. Front. Pediatr. 2020, 8, 1–9. [Google Scholar] [CrossRef]
  69. Fiocchi, A.; Dahda, L.; Dupont, C.; Campoy, C.; Fierro, V.; Nieto, A. Cow’s milk allergy: Towards an update of DRACMA guidelines. World Allergy Organ. J. 2016, 9, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Minniti, F.; Comberiati, P.; Munblit, D.; Piacentini, G.L.; Antoniazzi, E.; Zanoni, L.; Boner, A.L.; Peroni, D.G. Breast-milk characteristics protecting against allergy. Endocr Metab Immune Disord Drug Targets 2014, 14, 9–15. [Google Scholar] [CrossRef]
  71. Verduci, E.; D’elios, S.; Cerrato, L.; Comberiati, P.; Calvani, M.; Palazzo, S.; Martelli, A.; Landi, M.; Trikamjee, T.; Peroni, D.G. Cow’s milk substitutes for children: Nutritional aspects of milk from different mammalian species, special formula and plant-based beverages. Nutrients 2019, 11, 1739. [Google Scholar] [CrossRef] [Green Version]
  72. Department of Child Adolescent Health Development. The optimal duration of exclusive breastfeeding. In Report of an Expert Consultation. Department of Nutrition for Health and Development; World Health Organization: Geneva, Switzerland, 2001. [Google Scholar]
  73. Matangkasombut, P.; Padungpak, S.; Thaloengsok, S.; Kamchaisatian, W.; Sasisakulporn, C.; Jotikasthira, W.; Manuyakorn, W. Paediatrics and International child health detection of β-lactoglobulin in human breast-milk 7 days after cow milk ingestion. Paediatr. Int. Child. Health 2017, 37, 199–203. [Google Scholar] [CrossRef]
  74. Agostoni, C.; Decsi, T.; Fewtrell, M.; Goulet, O.; Kolacek, S.; Koletzko, B.; Michaelsen, K.F.; Moreno, L.; Puntis, J.; Rigo, J.; et al. Complementary feeding: A commentary by the ESPGHAN Committee on Nutrition. J. Pediatr. Gastroenterol. Nutr. 2008, 46, 99–110. [Google Scholar] [CrossRef] [Green Version]
  75. Grimshaw, K.E.; Maskell, J.; Oliver, E.M.; Morris, R.C.; Foote, K.D.; Mills, E.N.; Roberts, G.M. Introduction of complementary foods and the relationship to food allergy. Pediatrics 2013, 132, e1529–e1538. [Google Scholar] [CrossRef] [Green Version]
  76. Institute of Medicine (US); Committee on the Evaluation of the Addition of Ingredients New to Infant Formula. Comparing Infant Formulas with Human Milk; National Academies Press: Cambridge, MA, USA, 2004. [Google Scholar]
  77. Greer, F.R.; Sicherer, S.H.; Burks, A.W.; Committee on Nutrition, & Section on Allergy and Immunology. The effects of early nutritional interventions on the development of atopic disease in infants and children: The Role of maternal dietary restriction, breastfeeding, hydrolyzed formulas, and timing of introduction of allergenic complementary foods. Pediatrics 2019, 143, e20190281. [Google Scholar] [CrossRef] [Green Version]
  78. Greer, F.R.; Sicherer, S.H.; Burks, A.W.; American Academy of Pediatrics Committee on Nutrition, & American Academy of Pediatrics Section on Allergy and Immunology. Effects of early nutritional interventions on the development of atopic disease in infants and children: The role of maternal dietary restriction, breastfeeding, timing of introduction of complementary foods, and hydrolyzed formulas. Pediatrics 2008, 121, 183–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Saarinen, K.M.; Juntunen-Backman, K.; Järvenpää, A.L.; Kuitunen, P.; Lope, L.; Renlund, M.; Siivola, M.; Savilahti, E. Supplementary feeding in maternity hospitals and the risk of cow’s milk allergy: A prospective study of 6209 infants. J. Allergy Clin. Immunol. 1999, 104, 457–461. [Google Scholar] [CrossRef]
  80. Høst, A.; Husby, S.; Osterballe, O. A prospective study of cow s milk allergy in exclusively breast-fed infants. Acta Paediatr. Scand. 1988, 77, 663–670. [Google Scholar] [CrossRef] [PubMed]
  81. Katz, Y.; Rajuan, N.; Goldberg, M.R.; Eisenberg, E.; Heyman, E.; Cohen, A.; Leshno, M. Early exposure to cow’s milk protein is protective against IgE-mediated cow’s milk protein allergy. J. Allergy Clin. Immunol. 2010, 126, 77–82.e1. [Google Scholar] [CrossRef] [PubMed]
  82. Onizawa, Y.; Noguchi, E.; Okada, M.; Sumazaki, R. The Association of the delayed introduction of cow’s milk with IgE-mediated cow’s milk allergies. J. Allergy Clin. Immunol. Pract. 2021, 4, 481–488.e2. [Google Scholar] [CrossRef] [PubMed]
  83. Elizur, A.; Rajuan, N.; Goldberg, M.R.; Leshno, M.; Cohen, A.; Katz, Y. Natural course and risk factors for persistence of IgE-mediated cow’s milk allergy. YMPD 2012, 161, 482–487.e1. [Google Scholar] [CrossRef]
  84. Skripak, J.M.; Matsui, E.C.; Mudd, K.; Wood, R.A. The natural history of IgE-mediated cow’s milk allergy. J. Allergy Clin. Immunol. 2007, 120, 1172–1177. [Google Scholar] [CrossRef]
  85. Saarinen, K.M.; Pelkonen, A.S.; Mäkelä, M.J.; Savilahti, E. Clinical course and prognosis of cow’s milk allergy are dependent on milk-specific IgE status. J. Allergy Clin. Immunol. 2005, 116, 869–875. [Google Scholar] [CrossRef]
  86. Høst, A.; Halken, S.; Jacobsen, H.P.; Christensen, A.E.; Herskind, A.M.; Plesner, K. Clinical course of cow’s milk protein allergy/intolerance and atopic diseases in childhood. Pediatr. Allergy Immunol. 2002, 13, 23–28. [Google Scholar] [CrossRef] [PubMed]
  87. Fiocchi, A.; Terracciano, L.; Bouygue, G.R.; Veglia, F.; Sarratud, T.; Martelli, A.; Restani, P. Incremental prognostic factors associated with cow’s milk allergy outcomes in infant and child referrals: the Milan Cow’s Milk Allergy Cohort study. Annals of allergy, asthma & immunology: Official publication of the American College of Allergy. Asthma Immunol. 2008, 101, 166–173. [Google Scholar]
  88. Ahrens, B.; Lopes de Oliveira, L.C.; Grabenhenrich, L.; Schulz, G.; Niggemann, B.; Wahn, U.; Beyer, K. Individual cow’s milk allergens as prognostic markers for tolerance development? Clin. Exp. Allergy 2012, 42, 1630–1637. [Google Scholar] [CrossRef]
  89. Wood, R.A.; Sicherer, S.H.; Vickery, B.P.; Jones, S.M.; Liu, A.H.; Fleischer, D.M.; Henning, A.K.; Mayer, L.; Burks, A.W.; Grishin, A.; et al. The natural history of milk allergy in an observational cohort. J. Allergy Clin. Immunol. 2013, 131, 805–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Kim, J.S.; Nowak-Wegrzyn, A.; Sicherer, S.H.; Noone, S.; Moshier, E.L.; Sampson, H.A. Dietary baked milk accelerates the resolution of cow’s milk allergy in children. J. Allergy Clin. Immunol. 2011, 128, 125–131.e2. [Google Scholar] [CrossRef] [Green Version]
  91. Sicherer, S.H.; Wood, R.A.; Vickery, B.P.; Jones, S.M.; Liu, A.H.; Fleischer, D.M.; Dawson, P.; Mayer, L.; Burks, A.W.; Grishin, A.; et al. The natural history of egg allergy in an observational cohort. J. Allergy Clin. Immunol. 2015, 133, 492–499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kim, M.; Lee, Y.; Yang, H.; Won, J. The natural course of immediate-type cow’s milk and egg allergies in children. Int. Arch. Allergy Immunol. 2020, 181, 103–110. [Google Scholar] [CrossRef]
  93. Perry, T.T.; Matsui, E.C.; Conover-Walker, M.K.; Wood, R.A. The relationship of allergen-specific IgE levels and oral food challenge outcome. J. Allergy Clin. Immunol. 2004, 114, 144–149. [Google Scholar] [CrossRef]
  94. Leonard, S.A.; Caubet, J.C.; Kim, J.S.; Groetch, M.; Nowak-Wegrzyn, A. Baked milk-and egg-containing diet in the management of milk and egg allergy. J. Allergy Clin. Immunol. Pract. 2015, 3, 13–23. [Google Scholar] [CrossRef] [PubMed]
  95. D’Auria, E.; Salvatore, S.; Pozzi, E.; Mantegazza, C.; Sartorio, M.U.A.; Pensabene, L.; Baldassarre, M.E.; Agosti, M.; Vandenplas, Y.; Zuccotti, G. Cow’s milk allergy: Immunomodulation by dietary intervention. Nutrients 2019, 11, 1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Uncuoglu, A.; Yologlu, N.; Simsek, I.E.; Uyan, Z.S.; Aydogan, M. Tolerance to baked and fermented cow’s milk in children with IgE-mediated and non-IgE-mediated cow’s milk allergy in patients under two years of age. Allergol. Immunopathol. 2017, 45, 560–566. [Google Scholar] [CrossRef] [PubMed]
  97. Nowak-Wegrzyn, A.; Bloom, K.A.; Sicherer, S.H.; Shreffler, W.G.; Noone, S.; Wanich, N.; Sampson, H.A. Tolerance to extensively heated milk in children with cow’s milk allergy. J. Allergy Clin. Immunol. 2008, 122, 342–347.e2. [Google Scholar] [CrossRef]
  98. Esmaeilzadeh, H.; Alyasin, S.; Haghighat, M.; Nabavizadeh, H.; Esmaeilzadeh, E.; Mosavat, F. The effect of baked milk on accelerating unheated cow’s milk tolerance: A control randomized clinical trial. Pediatr. Allergy Immunol. 2018, 29, 747–753. [Google Scholar] [CrossRef] [PubMed]
  99. Host, A.; Halken, S. Cow’s milk allergy: Where have we come from and where are we going? Endocrine Metab. Immune Disord. Targets 2014, 14, 2–8. [Google Scholar] [CrossRef] [PubMed]
  100. Brotons-Canto, A.; Martín-Arbella, N.; Gamazo, C.; Irache, J.M. New pharmaceutical approaches for the treatment of food allergies. Expert Opin. Drug Deliv. 2018, 15, 675–686. [Google Scholar] [CrossRef] [PubMed]
  101. Alonso Lebrero, E.; Fernández Moya, L.; Somoza Álvarez, M.L. Sesión de actualización alergia a alimentos en niños. Alergol. Inmunol. Clin. 2001, 16, 96–115. [Google Scholar]
  102. Alonso Lebrero, E.; Fernández, L.S. Alergia a leche y huevo en niños. Alergol. Inmunol. Clin. 2001, 6, 96–110. [Google Scholar]
  103. Zapatero, L.; Alonso, E.; Fuentes, V.; Martínez, M.I. Oral desensitization in children with cow’s milk allergy. J. Investig. Allergol. Clin. Immunol. 2008, 18, 389–396. [Google Scholar]
  104. Mestecky, J.; McGhee, J.R.; Bienenstock, J.; Lamm, M.E.; Strober, W.; Cebra, J.J.; Mayer, L.; Ogra, P.L.; Russell, M.W. Historical aspects of mucosal immunology. In Mucosal Immunology, 4th ed.; Nature: Oberhaching, Germany, 1983; pp. 37–51. [Google Scholar]
  105. Noimark, L.; Cox, H.E. Nutritional problems related to food allergy in childhood. Pediatr. Allergy Immunol. 2008, 19, 188–195. [Google Scholar] [CrossRef] [PubMed]
  106. Tang, M.L.K.; Martino, D.J. Oral immunotherapy and tolerance induction in childhood. Pediatr. Allergy Immunol. 2013, 24, 512–520. [Google Scholar] [CrossRef] [PubMed]
  107. Mori, F.; Barni, S.; Liccioli, G.; Novembre, E. Oral immunotherapy (OIT): A personalized medicine. Medicina 2019, 55, 684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Sanchez-Garcıa, S.; Rodríguez del Río, P.; Escudero, C.; Garcıa-Fernandez, C.; Ramirez, A.; Ibáñez, M.D. Efficacy of oral immunotherapy protocol for specific oral tolerance induction in children with cow’s milk allergy. ISR Med. Assoc. J. 2012, 14, 43–47. [Google Scholar] [PubMed]
  109. Chinthrajah, R.S.; Hernandez, J.D.; Boyd, S.D.; Galli, S.J.; Nadeau, K.C. Molecular and cellular mechanisms of food allergy and food tolerance. J. Allergy Clin. Immunol. 2016, 137, 984–997. [Google Scholar] [CrossRef] [Green Version]
  110. Pajno, G.B.; Fernandez-Rivas, M.; Arasi, S.; Roberts, G.; Akdis, C.A.; Alvaro-Lozano, M.; Beyer, K.; Bindslev-Jensen, C.; Burks, W.; Ebisawa, M.; et al. EAACI guidelines on allergen immunotherapy: IgE-mediated food allergy. Allergy Eur. J. Allergy Clin. Immunol. 2018, 73, 799–815. [Google Scholar] [CrossRef] [Green Version]
  111. Sánchez-García, S.; Cipriani, F.; Ricci, G. Food allergy in childhood: Phenotypes, prevention and treatment. Pediatr. Allergy Immunol. 2015, 26, 711–720. [Google Scholar] [CrossRef]
  112. Staden, U.; Rolinck-Werninghaus, C.; Brewe, F.; Wahn, U.; Niggemann, B.; Beyer, K. Specific oral tolerance induction in food allergy in children: Efficacy and clinical patterns of reaction. Allergy 2007, 62, 1261–1269. [Google Scholar] [CrossRef]
  113. Barni, S.; Mori, F.; Piccorossi, A.; Sarti, L.; Pucci, N.; Maresca, M.; Giovannini, M.; Liccioli, G.; Novembre, E. Low-Dose oral food challenge with hazelnut: E_cacy and tolerability in children. Int. Arch. Allergy Immunol. 2019, 178, 97–100. [Google Scholar] [CrossRef] [PubMed]
  114. Pajno, G.B.; Caminiti, L.; Salzano, G.; Crisafulli, G.; Aversa, T.; Messina, M.F.; Wasniewska, M.; Passalacqua, G. Comparison between two maintenance feeding reimens after successful cow’s milk oral desensitization. Pediatr. Allergy Immunol. 2013, 24, 376–381. [Google Scholar] [CrossRef]
  115. Caminiti, L.; Passalacqua, G.; Barberi, S.; Vita, D.; Barberio, G.; De Luca, R.; Pajno, G.B. A new protocol for specific oral tolerance induction in children with IgE-mediated cow’s milk allergy. Allergy Asthma Proc. 2009, 30, 443–448. [Google Scholar] [CrossRef] [PubMed]
  116. Nowak-Wegrzyn, A.; Sato, S.; Fiocchi, A.; Ebisawa, M. Oral and sublingual immunotherapy for food allergy. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 606–613. [Google Scholar] [CrossRef] [PubMed]
  117. Meglio, P.; Bartone, E.; Plantamura, M.; Arabito, E.; Giampietro, P.G. A protocol for oral desensitization in children with IgE-mediated cow’s milk allergy. Allergy 2004, 59, 980–987. [Google Scholar] [CrossRef] [PubMed]
  118. Narisety, S.D.; Skripak, J.M.; Steele, P.; Hamilton, R.G.; Matsui, E.C.; Burks, A.W.; Wood, R.A. Open-label maintenance after milk oral immunotherapy for IgE-mediated cow’s milk allergy. J. Allergy Clin. Immunol. 2009, 124, 610–612. [Google Scholar] [CrossRef] [Green Version]
  119. Goldberg, M.R.; Nachshon, L.; Appel, M.Y.; Elizur, A.; Levy, M.B.; Eisenberg, E.; Sampson, H.A.; Katz, Y. Efficacy of baked milk oral immunotherapy in baked milk-reactive allergic patients. J. Allergy Clin. Immunol. 2015, 136, 1601–1606. [Google Scholar] [CrossRef]
  120. Takahashi, M.; Taniuchi, S.; Soejima, K.; Hatano, Y.; Yamanouchi, S.; Kaneko, K. Two-weeks-sustained unresponsiveness by oral immunotherapy using microwave heated cow’s milk for children with cow’s milk allergy. Allergy Asthma Clin. Immunol. 2016, 12, 44. [Google Scholar] [CrossRef] [Green Version]
  121. Ebrahimi, M.; Gharagozlou, M.; Mohebbi, A.; Hafezi, N.; Azizi, G.; Brahimi, M.; Gharagozlou, M.; Mohebbi, A.; Hafezi, N.; Azizi, G.; et al. The efficacy of oral immunotherapy in patients with cow’s milk allergy. Iran. J. Allergy Asthma Immunol. 2017, 16, 183–192. [Google Scholar]
  122. Skripak, J.M.; Nash, S.D.; Rowley, H.; Brereton, N.H.; Oh, S.; Hamilton, R.G.; Matsui, E.C.; Burks, A.W.; Wood, R.A. A randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow’s milk allergy. J. Allergy Clin. Immunol. 2008, 122, 1154–1160. [Google Scholar] [CrossRef] [Green Version]
  123. Longo, G.; Barbi, E.; Berti, I.; Meneghetti, R.; Pittalis, A.; Ronfani, L.; Ventura, A. Specific oral tolerance induction in children with very severe cow’s milk-induced reactions. J. Allergy Clin. Immunol. 2008, 121, 343–347. [Google Scholar] [CrossRef]
  124. Pajno, G.B.; Caminiti, L.; Ruggeri, P.; De Luca, R.; Vita, D.; La Rosa, M.; Passalacqua, G. Oral immunotherapy for cow’s milk allergy with a weekly up-dosing regimen: A randomized single-blind controlled study. Ann. Allergy Asthma Immunol. 2010, 105, 376–381. [Google Scholar] [CrossRef]
  125. Martorell, A.; De la Hoz, B.; Ibáñez, M.D.; Bone, J.; Terrados, M.S.; Michavila, A.; Plaza, A.M.; Alonso, E.; Garde, J.; Nevot, S.; et al. Oral desensitization as a useful treatment in 2-year-old children with cow’s milk allergy. Clin. Exp. Allergy. 2011, 41, 1297–1304. [Google Scholar] [CrossRef]
  126. Amat, F.; Kouche, C.; Gaspard, W.; Lemoine, A.; Guiddir, T.; Lambert, N.; Zakariya, M.; Ridray, C.; Nemni, A.; Saint-Pierre, P.; et al. Is a slow-progression baked milk protocol of oral immunotherapy always a safe option for children with cow’s milk allergy? A randomized controlled trial. Clin. Exp. Allergy. 2017, 47, 1491–1496. [Google Scholar] [CrossRef]
  127. Maeda, M.; Imai, T.; Ishikawa, R.; Nakamura, T.; Kamiya, T.; Kimura, A.; Fujita, S.; Akashi, K.; Tada, H.; Morita, H.; et al. Effect of oral immunotherapy in children with milk allergy: The ORIMA study. Allergol Int. 2021, 70, 223–228. [Google Scholar] [CrossRef]
  128. Mota, I.; Piedade, S.; Gaspar, Â.; Benito-Garcia, F.; Sampaio, G.; Borrego, L.M.; Morais-Almeida, M. Cow’s milk oral immunotherapy in real life: 8-year long-term follow-up study. Asia Pac. Allergy 2018, 8, e28. [Google Scholar] [CrossRef]
  129. Berti, I.; Badina, L.; Cozzi, G.; Giangreco, M.; Bibalo, C.; Ronfani, L.; Barbi, E.; Ventura, A.; Longo, G. Early oral immunotherapy in infants with cow’s milk protein allergy. Pediatr. Allergy Immunol. 2019, 30, 572–574. [Google Scholar] [CrossRef] [PubMed]
  130. De Schryver, S.; Mazer, B.; Clarke, A.E.; Pierre, Y.S.; Lejtenyi, D. Adverse events in oral immunotherapy for the desensitization of cow’s milk allergy in children: A randomized controlled trial. J. Allergy Clin. Immunol. Pract. 2019, 7, 1912–1919. [Google Scholar] [CrossRef] [PubMed]
  131. Efron, A.; Zeldin, Y.; Gotesdyner, L.; Stauber, T.; Maoz Segal, R.; Binson, I.; Dinkin, M.; Dinkowitz, L.; Shahar, D.; Deutch, M.; et al. A structured gradual exposure protocol to baked and heated milk in the treatment of milk allergy. J. Pediatr. 2018, 203, 204–209. [Google Scholar] [CrossRef]
  132. Kauppila, T.K.; Paassilta, M.; Kukkonen, A.K.; Kuitunen, M.; Pelkonen, A.S.; Makela, M.J. Outcome of oral immunotherapy for persistent cow’s milk allergy from 11 years of experience in Finland. Pediatr. Allergy Immunol. 2019, 30, 356–362. [Google Scholar] [CrossRef]
  133. Demir, E.; Ciğerci Günaydın, N.; Gülen, F.; Tanaç, R. Oral Immunotherapy for cow’s milk allergy: five years’ experience from a single center in Turkey. Balk. Med. J. 2020, 37, 316–323. [Google Scholar] [CrossRef] [PubMed]
  134. Gruzelle, V.; Juchet, A.; Martin-Blondel, A.; Michelet, M.; Chabbert-Broue, A.; Didier, A. Benefits of baked milk oral immunotherapy in French children with cow’s milk allergy. Pediatr. Allergy Immunol. 2020, 31, 364–370. [Google Scholar] [CrossRef]
  135. Morisset, M.; Moneret-Vautrin, D.A.; Guenard, L.; Cuny, J.M.; Frentz, P.; Hatahet, R.; Hanss, C.H.; Beaudouin, E.; Petit, N.; Kanny, G. Oral desensitization in children with milk and egg allergies obtains recovery in a significant proportion of cases. A randomized study in 60 children with cow’s milk allergy and 90 children with egg allergy. Eur. Ann. Allergy Clin. Immunol. 2007, 39, 12–19. [Google Scholar] [PubMed]
  136. Patriarca, G.; Nucera, E.; Roncallo, C.; Pollastrini, E.; Bartolozzi, F.; De Pasquale, T.; Buonomo, A.; Gasbarrini, G.; Di Campli, C.; Schiavino, D. Oral desensitizing treatment in food allergy: Clinical and immunological results. Aliment. Pharmacol. Ther. 2003, 17, 459–465. [Google Scholar] [CrossRef] [PubMed]
  137. Wood, R.A. Oral immunotherapy for food allergy. J. Investig. Allergol. Clin. Immunol. 2017, 27, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Nurmatov, U.; Dhami, S.; Arasi, S.; Pajno, G.B.; Fernandez-Rivas, M.; Muraro, A.; Roberts, G.; Akdis, C.; Alvaro-Lozano, M.; Beyer, K.; et al. Allergen immunotherapy for IgE-mediated food allergy: a systematic review and meta-analysis. Allergy 2017, 72, 1133–1147. [Google Scholar] [CrossRef]
  139. Epstein-Rigbi, N.; Goldberg, M.R.; Levy, M.B.; Nachshon, L.; Elizur, A. Quality of life of children aged 8-12 years undergoing food allergy oral immunotherapy: Child and parent perspective. Allergy Eur. J. Allergy Clin. Immunol. 2020, 75, 2623–2632. [Google Scholar] [CrossRef]
  140. Carraro, S.; Frigo, A.C.; Perin, M.; Stefani, S.; Cardarelli, C.; Bozzetto, S.; Baraldi, E.; Zanconato, S. Impact of oral immunotherapy on quality of life in children with cow milk allergy: A pilot study. Int. J. Immunopathol. Pharmacol. 2012, 25, 793–798. [Google Scholar] [CrossRef] [Green Version]
  141. Kauppila, T.K.; Pelkonen, A.S.; Roine, R.P.; Paassilta, M.; Kukkonen, K.; Sintonen, H.; Mäkelä, M. Health-related quality of life in patients who had partaken in milk oral immunotherapy and comparison to the general population. Allergy Eur. J. Allergy Clin. Immunol. 2021, 76, 387–390. [Google Scholar] [CrossRef]
Figure 1. Clinical presentation of IgE and non IgE CMA.
Figure 1. Clinical presentation of IgE and non IgE CMA.
Nutrients 13 01525 g001
Table 1. Main characteristics of CM allergens, adapted from Hochwallner [8].
Table 1. Main characteristics of CM allergens, adapted from Hochwallner [8].
Allergen NameProteinConcentration (g/L)Size (kDa)Prevalence (% of Patients)Allergenic Activity (% of Patients)
Whey (20%)
(5 g/L)
Bos d 4α–Lactalbumin1–1.514.20–6712
Bos d 5β–Lactoglobulin3–418.313–6219
Bos d 6Bovine serum albumin0.1–0.466.30–761
Bos d 7Immunoglobulins0.6–116012–36
Lactoferrin0.09800–353
Whole casein (80%) (30 g/L)Bos d 9αS1–casein12–1523.665–10026
Bos d 10αS2–casein3–425.2
Bos d 11β–casein9–112435–4435
Bos d 12k–casein3–41935–4126
Table 2. Positive predictive values of 90% and 95% of sIgE levels for a positive challenge.
Table 2. Positive predictive values of 90% and 95% of sIgE levels for a positive challenge.
Author, YearAge90%95%Method
Sampson and Ho, 1997 [29]5.2 years23 kU/L32 kU/LCAP system FEIA
Garcia–Ara et al., 2001 [30]4.8 months2.5 kU/L5 kU/LCAP system FEIA
Sampson, 2001 [31]3.8 years15 kU/L32 kU/LCAP system FEIA
Garcia–Ara et al., 2004 [32]13–18 months1.5 kU/L2.7 kU/LCAP system FEIA
19–24 months6 kU/L9 kU/L
25–36 months14 kU/L24 kU/L
Celik–Bilgili et al., 2005 [33]<1 year25.8 kU/L CAP system FEIA
Komata et al., 2007 [25]<1 year 5.8 kU/LCAP system FEIA
1 year38.6 kU/L
2 years57.3 kU/L
Martorell et al., 2008 [34]12 months 5.8 kU/LCAP system FEIA
18 months9.8 kU/L
24 months27.5 kU/L
36 months7.4 kU/L
48 months5 kU/L
Van der Gutgen et al., 2008 [35]<2.5 years5 kU/L7.5 kU/LCAP system FEIA
Ott. et al., 2008 [36] 52.7 kU/L66.9 kU/LCAP system FEIA
Table 3. Positive predictive value of 90% and 95% of SPT for a positive challenge.
Table 3. Positive predictive value of 90% and 95% of SPT for a positive challenge.
Author, YearAgeØ SPT 90%Ø SPT 95%Type of Allergen
Eigenmann and Sampson, 1998 [39]4.6 years >5 mmGlycerinate extract
Sporik et al., 2000 [40]3 years >8 mmGlycerinate extract
<2 years>6 mm
Calvani et al., 2007 [41]3.6 years 15 mmFresh milk
12 mmα-Lactalbumin
8 mmCasein
10 mmβ-lactoglobulin
Calvani et al., 2012 [42]3.7 years 20 mmFresh milk
10 mmα-Lactalbumin
7 mmCasein
8 mmβ-Lactoglobulin
Onesimo et al., 2013 [38]2.7 years 4.9 mmα-Lactalbumin
4.3 mmCasein
5.6 mmβ-Lactaglobulin
Kido et al., 2016 [43]1.4 years15 mm Glycerinate extract
Table 5. Efficacy of milk OIT.
Table 5. Efficacy of milk OIT.
Author, YearType of StudyType of MilkPopulation (n)Age (years)Partial ToleranceComplete Tolerance
Meglio P. et al., 2004 [117]Open-labelFresh CM216–1014.3% (40–80 mL of CM)71.4% (200 mL of CM)
Narisety SD. et al., 2009 [118]OpenFresh CM156–16 33% (16 g of CM proteins)
Goldberg M. et al., 2015 [119]OpenBaked CM146.5–12.7 21% (1.3 g of CM proteins)
Takahashi M. et al. 2016 [120]OpenMicrowave heated CM315–17 45.2% (200 mL of CM)
Ebrahimi M. et al.2017 [121]OpenFresh CM143.5–7 92.9% (200 to 250 mL of CM)
Skripak et al., 2008 [122]Randomized, double-blind, placebo-controlledFresh CM136–17 30.8% (500 mg of CM proteins)
Longo G. et al., 2008 [123] Randomized open-labelFresh CM305–1754% (5–150 mL of CM)36% (> 150 mL of CM)
Pajno GB. et al., 2010 [124]Randomized, placebo controlledFresh CM154–10 67% (200 mL ofCM)
Martorell A. et al., 2011 [125]Randomized, placebo controlledFresh CM302–3 90% (200 mL of CM)
Amat F. et al.2017 [126]RandomizedBaked CMFresh CM433–1026.8% (0.27–2.5 g of CM proteins)36.6% (2.72 g of CM proteins)
Maeda M, et al., 2020 [127]Randomized controlledFresh CM283–12 50% (100 mL of CM)
Mota I. et al., 2018 [128] ProspectiveFresh CM422–18 92% (200 mL of CM)
Berti I. et al., 2019 [129] ProspectiveFresh CM733–11 97% (150 mL of CM)
De Schryver S. et al., 2019 [130]Prospective, randomized-controlledFresh CM416–18 73.2% (200 mL of CM)
Efron A. et al.,2018 [131]Retrospective, case-controlFresh CM431–4 86% (250 mL of CM)
Kauppila T.K. et al., 2019 [132]RetrospectiveFresh CM2965–17 56% (200 mL of CM)
Demir E. et al., 2020 [133]Retrospective, cohort studyFresh CM473–13 89.3% (200 mL of CM)
Gruzelle V. et al., 2020 [134]RetrospectiveBaked CM642–16 42.2% (254 mL of CM)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giannetti, A.; Toschi Vespasiani, G.; Ricci, G.; Miniaci, A.; di Palmo, E.; Pession, A. Cow’s Milk Protein Allergy as a Model of Food Allergies. Nutrients 2021, 13, 1525. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051525

AMA Style

Giannetti A, Toschi Vespasiani G, Ricci G, Miniaci A, di Palmo E, Pession A. Cow’s Milk Protein Allergy as a Model of Food Allergies. Nutrients. 2021; 13(5):1525. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051525

Chicago/Turabian Style

Giannetti, Arianna, Gaia Toschi Vespasiani, Giampaolo Ricci, Angela Miniaci, Emanuela di Palmo, and Andrea Pession. 2021. "Cow’s Milk Protein Allergy as a Model of Food Allergies" Nutrients 13, no. 5: 1525. https://0-doi-org.brum.beds.ac.uk/10.3390/nu13051525

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop