Next Article in Journal
Home Enteral Nutrition in Adults—Nationwide Multicenter Survey
Next Article in Special Issue
Persistent Iron Deficiency Anemia in Patients with Celiac Disease Despite a Gluten-Free Diet
Previous Article in Journal
Current Trends of Essential Trace Elements in Patients with Chronic Liver Diseases
Previous Article in Special Issue
The Gluten Free Diet’s Impact on Growth in Children with Celiac Disease in Two Different Countries
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-IgE-Mediated Gastrointestinal Food Allergies in Children: An Update

by
Roxane Labrosse
1,2,
François Graham
2,3 and
Jean-Christoph Caubet
4,*
1
Division of Hematology-Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
2
Division of Allergy and Immunology, Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, QC H3T 1C5, Canada
3
Division of Allergy and Immunology, Department of Medicine, Centre Hospitalier de l’Universite de Montreal (CHUM), University of Montreal, Montreal, QC H2X 3E4, Canada
4
Pediatric Allergy Unit, Department of Woman, Child and Adolescent, University Hospitals of Geneva, 1205 Geneva, Switzerland
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(7), 2086; https://doi.org/10.3390/nu12072086
Submission received: 11 June 2020 / Revised: 26 June 2020 / Accepted: 2 July 2020 / Published: 14 July 2020
(This article belongs to the Special Issue Nutrition and Pediatric Gastroenterology)

Abstract

:
Non-immunoglobulin E-mediated gastrointestinal food allergic disorders (non-IgE-GI-FA) include food protein-induced enterocolitis syndrome (FPIES), food protein-induced enteropathy (FPE) and food protein-induced allergic proctocolitis (FPIAP), which present with symptoms of variable severity, affecting the gastrointestinal tract in response to specific dietary antigens. The diagnosis of non-IgE-GI-FA is made clinically, and relies on a constellation of typical symptoms that improve upon removal of the culprit food. When possible, food reintroduction should be attempted, with the documentation of symptoms relapse to establish a conclusive diagnosis. Management includes dietary avoidance, nutritional counselling, and supportive measures in the case of accidental exposure. The prognosis is generally favorable, with the majority of cases resolved before school age. Serial follow-up to establish whether the acquisition of tolerance has occurred is therefore essential in order to avoid unnecessary food restriction and potential consequent nutritional deficiencies. The purpose of this review is to delineate the distinctive clinical features of non-IgE-mediated food allergies presenting with gastrointestinal symptomatology, to summarize our current understanding of the pathogenesis driving these diseases, to discuss recent findings, and to address currents gaps in the knowledge, to guide future management opportunities.

1. Introduction

Gastrointestinal (GI) complaints represent a frequent motive for seeking medical attention in the pediatric setting, and diagnosis can be challenging due to the wide variety of potential underlying causes. Particularly in the first years of life, food allergies represent a substantial proportion of disorders involving the GI tract [1]. Food allergies comprise a spectrum of diseases that have in common the immunological reaction to specific dietary proteins, and the reproducibility of symptoms upon re-exposure [2,3]. This is in contrast to food intolerances, in which immunological mechanisms are not involved. Non-IgE-mediated gastrointestinal food allergic diseases (non-IgE-GI-FA), which are being increasingly recognized in children, consist of three main entities: food protein-induced enterocolitis syndrome (FPIES), food protein-induced enteropathy (FPE) and food protein-induced allergic proctocolitis (FPIAP). Despite their potential for serious reactions, both first and second-line health care providers report poor familiarity with these disorders [4,5], perhaps due to the many gaps in knowledge, which include unclear pathophysiology, a paucity of reliable diagnostic tools, and a lack of uniform management protocols. This review will focus on non-IgE-GI-FA in children, summarizing what is currently known, and highlighting the latest advances in the field.

2. Classification and Terminology

Adverse food reactions are divided into immune-mediated reactions (i.e., food allergy) and non-immune mediated reactions (i.e., food intolerances). The term ‘food allergy’ is used to designate an immune-mediated adverse reaction to food proteins. This includes IgE-mediated food allergy (IgE-FA), mixed IgE and non IgE-mediated food allergy, and non-IgE-GI-FA [6]. On the other hand, adverse non-immune mediated reactions that are not classified as food allergy include malabsorption due to enzyme deficiency (ex: lactase deficiency), reaction to toxic contaminants (ex: scombroid poisoning), and pharmacologic food components (ex: caffeine) among others [7], which are beyond the scope of this review.
Gastrointestinal food allergies are generally classified according to their underlying pathogenesis (Figure 1) [2,8]. In IgE-FA, reactions generally occur rapidly after the ingestion of the culprit food. Although isolated gastrointestinal symptoms can occur (termed “gastrointestinal anaphylaxis”), typically with egg, more often, these are accompanied by other features affecting the skin and mucosa (hives, angioedema), respiratory tract (cough, wheezing, nasal congestion) or cardiovascular system (hypotension), and can present as life-threatening anaphylaxis. Reactions in mixed IgE/non-IgE-mediated diseases, such as eosinophilic gastrointestinal diseases, are triggered by complex immunological mechanisms that only partially implicate IgE. Symptoms are dependent upon the affected organs and the extent of eosinophilic infiltration [8,9]. On the other end of the spectrum lie non-IgE-GI-FA, in which circulating food-specific IgE are typically absent. These include FPIES, FPE, and FPIAP. In contrast to IgE-FA, associated GI symptoms are usually delayed after exposure to foods, and can have a chronic presentation [8,10]. Although their underlying pathomechanism is still poorly elucidated, these entities may represent a continuum of disease, where the expression of symptoms and severity is dependent upon the affected segment of the gastrointestinal tract (Figure 2). FPIAP symptomatology is induced by the localized inflammation of the distal colon, causing hematochezia in otherwise well-appearing infants. FPE predominantly affects the small intestine, resulting in lower digestive manifestations such as malabsorption symptoms, potentially accompanied by a failure to thrive (FTT). Finally, FPIES can affect the entire gastrointestinal tract, predominantly causing symptoms of intractable emesis which can be severe enough to cause metabolic disturbances and hypovolemic shock. FPIES can be further classified according to the timing of symptoms (acute vs. chronic FPIES), the severity of clinical manifestations (mild, moderate, severe), the age of onset (early-onset, late-onset, adult FPIES), the type of triggering foods (cow’s milk/soy vs. solid foods), and the presence of food-specific IgE (sIgE) (atypical FPIES) (Figure 3), all of which are described in detail below.

3. Epidemiology

Of the non-IgE-GI-FA, FPIAP is certainly the most frequent, although the exact prevalence is not well established. A large study of an Israeli birth cohort found the overall prevalence of FPIAP to be relatively low, at 0.16% [11]. Conversely, a cumulative incidence of 17% was found in a recent prospective observational study of healthy newborns with pediatrician-diagnosed FPIAP and established evidence of gastrointestinal bleeding in the United States [12]. The absence of diagnosis confirmation by reintroduction and the inclusion of patients with occult blood possibly led to overdiagnosis in the latter study. However, the incidence remained high (7%), despite restricting the analysis to those with gross blood only, thus only partially explaining the 100-fold difference between both studies. In infants with visible rectal bleeding, FPIAP has been found to be causal in up to 60% of cases [13,14].
The prevalence of FPE has not been thoroughly studied, but it is thought to be relatively uncommon, accounting for about a fifth of patients compared to celiac disease in reports from a single-center experience from Finland [15,16]. In another Finnish study, the prevalence of FPE to cow’s milk in older children was rather high, at 2.2% [17]. However, the incidence of this condition appears to be progressively decreasing over time [16,18]. Potential explanations for this decline include the upsurge in breastfeeding practices, which may be protective, and the use of better adapted formulas with a lower protein content [16].
While FPIES is also usually regarded as a rare disease, recent reports challenged this notion, revealing a cumulative incidence of 0.3–0.7% in infancy [19,20], although a lower incidence of 0.015% cases per year was also reported in Australia [21]. However, the latter study may have underestimated the incidence of FPIES, because it relied on a voluntary reporting system from pediatricians, and only included cases of acute FPIES. In cow’s milk-induced FPIES specifically, two independent studies conducted in Israel and Spain have both found comparable cumulative incidences in children during the first 2–3 years of life (0.34% and 0.35%, respectively) [19,20]. Similarly, the latest prevalence studies have estimated that 0.51% of children and 0.22% of adults in the United States report having physician-diagnosed FPIES [22].
Co-atopic disease is highly overrepresented in patients with non-IgE-GI-FA, affecting 40–60% of FPIES patients [23,24,25], and up to 40–50% of FPE and FPIAP patients [12,17] (Table 1). Likewise, a family history of atopy is present in as much as 60% and 80% of first-degree relatives in FPIAP and FPIES, respectively [21,24,26,27].
Interestingly, both FPIES and FPE have been reported in patients with Down syndrome, who may present with a protracted course [15,28,29]. This may be due to intrinsic immune defects seen in these children [30], which include the dysregulated secretion of TNFα and IL-10 [31,32], two key cytokines implicated in FPIES, as described below.

4. Pathophysiology

The pathogenesis of non-IgE-GI-FA is poorly understood, but differs from IgE-FA, in that cellular immunity is thought to be driving the allergic inflammatory response while circulating sIgE are notably absent, although a localized IgE-response to the gut has been described and may be a contributing factor [33].
In FPIES, many groups have shown the presence of specific T cell responses to causative antigens [34,35,36,37,38,39,40,41], with an imbalance between exaggerated TNFα secretion by food-specific T cells, and deficient TGFβ responses being consistently reported [36,39,42,43,44,45,46]. This led to an initial understanding of disease as being the result of local T cell infiltration and inflammation causing an increase in intestinal permeability, with a subsequent fluid shift and increase in antigen influx [39,47,48]. However, this simplistic model has recently been challenged in light of conflicting evidence and a lack of specificity of the findings implicating cellular immunity. Th2 responses, which are typical of IgE-FA, have also been described in FPIES, with increased production of IL-4, IL-5, IL-9, and IL-13 cytokines, a phenomenon which may be explained by the high rates of co-atopy seen in these patients [33,36,37,43,49,50]. Conversely, regulatory T cells (Tregs) and IL-10 secretion may play a role in the acquisition of tolerance [37,49,51]. Taken together, these data support the presence of food-specific T cells in FPIES, although more studies are needed to confirm their distinctive role with regards to disease manifestations. Interestingly, other arms of the immune system have recently been studied, and may also contribute to disease. Recent studies using unbiased approaches, such as CyTOF mass cytometry and transcriptional profiling, revealed the broad, systemic activation of the innate immune system after positive FPIES oral food challenges (OFC) [38,52], with the activation of monocytes, neutrophils, eosinophils, natural killer cells, and eosinophils. In another study, higher levels of IL-8 were present in FPIES patients who reacted to an OFC, suggesting neutrophil involvement in those with active disease [37]. The in vitro production of IL-9, a cytokine implicated in the recruitment of mast cells and intestinal anaphylaxis, was also increased in patients with FPIES compared to those with IgE-FA. Baseline tryptase levels were significantly higher in those patients with positive OFC, further supporting the role of mast cells in FPIES [37]. How these innate cells recognize specific food antigens remains, however, enigmatic. Regarding humoral immunity, low levels of neutralizing specific-IgG4 antibodies have been found [37,41,42], while analyses of specific-IgA levels have yielded contradictory results [41,42,53].
In FPE, the structural damage to the jejunal mucosa appears to result from food-specific (mostly cow’s milk) T cell infiltration, causing malabsorption [16,54]. There is strong evidence to support the predominant role of cytotoxic CD8+ T cells in mediating disease [55,56], although an increased density of intraepithelial γδ-TCR+ cells has also been reported [16].
FPIAP is characterized by the dense eosinophilic infiltration of the rectosigmoid mucosa [57,58]. It is still unknown why the inflammatory response is localized to the distal colon in FPIAP, but the high prevalence of this disease amongst breastfed infants suggests that immunologic components found in breastmilk may play a role. One hypothesis is that immunoglobulins secreted in breastmilk may bind dietary proteins, which are only released after being cleaved off by colonic enzymes. Furthermore, the use of antacids, which has been suggested to increase the risk of FPIAP [12], may also enhance the allergenic potential of proteins by preventing their digestion [59]. However, these data need to be confirmed by further studies.

5. Clinical Manifestations

While non-IgE-GI-FA comprise distinct clinical entities, they may present with overlapping clinical features. However, there is an apparent gradient of symptom magnitude, with FPIAP and FPIES being at opposing ends of the severity spectrum (Figure 2). Furthermore, some distinctive features (highlighted in Table 1) may help discriminate between the three entities.
In acute FPIES, the hallmark feature is profuse and repetitive vomiting (>95% of patients) occurring 1 to 4 h after food ingestion. Diarrhea can also follow 5 to 10 h later, although this is a less common feature (25–50%). Infants often appear septic, presenting with symptoms of lethargy (65–100%), pallor (30–90%) and hypothermia (5%) [24,26,60,61,62,63,64,65,66,67]. Symptoms may be quite severe, with up to 15% experiencing hemodynamic instability [65,68]. Chronic FPIES, on the other hand, presents with chronic watery diarrhea (occasionally with blood or mucus), intermittent emesis, abdominal distension, and/or poor weight gain. In a subgroup of patients, symptoms progressively worsen and can lead to dehydration (15–45%) and metabolic disturbances (5%) [26,62,64]. Typically, chronic FPIES will occur with persistent exposure to cow’s milk or soy-based formula, while intermittent exposure to solid foods such as rice is more likely to cause an acute presentation [24]. A defining feature of chronic FPIES is the recurrence of symptoms presenting acutely when the trigger food is reintroduced after a period of withdrawal (acute-on-chronic phenotype) [24,69]. Rarely, FPIES has been reported in exclusively breastfed infants [70,71], with cases-series reporting an occurrence of this phenomenon in 0–5% of children [21,26,72]. The age of onset can vary, with FPIES to cow’s milk/soy usually presenting earlier within the first weeks or months of life, while FPIES to solids presents somewhat later, at about 4–7 months of life, probably linked to the timing of the introduction of complementary foods into the diet. Although FPIES generally occurs in early infancy, adult-onset FPIES is now also being increasingly recognized, most frequently triggered by seafood [73,74,75,76]. Finally, a rare occurrence of symptomatic fetal FPIES has recently been reported [77], as well as neonatal FPIES due to intrauterine sensitization [78].
In FPE, patients present with a syndrome resembling that of celiac disease. In most cases, symptoms develop in infants shortly after the introduction of cow’s milk in the diet, with chronic diarrhea and features of malabsorption such as steatorrhea and failure to thrive (FTT), which are responsive to cow’s milk elimination. Vomiting is also frequently reported [15,16,28,79,80,81]. Notably, other extra-digestive symptoms seen in celiac disease, such as dermatitis herpetiformis, are absent. Because of the significant mucosal damage, patients with FPE can also suffer from secondary carbohydrate intolerance [82].
In contrast to FPIES and FPE, FPIAP most often occurs in exclusively breasted infants within the first weeks of life because of indirect exposure to maternal dietary protein via breastmilk, although direct feeding can also trigger symptoms [83]. These infants present with bloody, loose stools, sometimes with mucus. Affected children are however well-appearing, have no severe symptoms of emesis and diarrhea and no significant growth failure [27,58,84,85,86,87,88]. A subset of patients can also present with gagging, food refusal and irritability [12], suggesting that perhaps the allergic inflammation is not entirely restricted to the distal colon. Although FPIAP is most often seen in young infants, it has also been reported to occur in older children, representing 18% of the children evaluated for rectal bleeding in one study [89].

6. Reported Food Triggers

Cow’s milk is the most commonly reported food allergy in children [2], of which non-IgE-GI-FA account for up to 40–50% of reactions [90,91,92]. Therefore, unsurprisingly, cow’s milk is responsible for the majority of reactions in FPIES, FPE and FPIAP. However, many other foods have been implicated, as listed in Table 2. In addition to cow’s milk, soy has been frequently reported as a FPIES trigger in countries in which soy formula use is common [93,94], and co-allergy to both foods is seen in up to 40–60% of cases [21,26,95,96]. About a third of patients with cow’s milk/soy FPIES will also react to one or more solid foods [69]. While any solid food can cause FPIES, some overrepresented triggers include grains (rice, oats), vegetables (sweet potato, squash), fruits (banana, avocado), poultry and eggs. Rice is the most commonly reported solid trigger in the United States and Australia, and may be associated with a more severe phenotype [63,97,98,99]. Furthermore, other geographical variations are evident, such as the high prevalence of soy-induced FPIES in the United States [23,25,26,65,95], and of fish and shellfish reactions in Italy and Spain [20,40,60,76,100,101]. These are likely influenced by local dietary practices and may be the result of differences in genetic background [102]. As a whole, common triggers reflect those foods introduced during the first year of life, as index reactions are unlikely to occur in foods introduced thereafter [26,61,93]. While the majority (65–80%) of children react to a single food, most commonly cow’s milk, about 10% of children will react to three foods or more. In contrast, a majority (up to 80%) of those with FPIES to solid food tend to have multiple food triggers [24,26,65]. Frequently, children will react upon their first exposure to the incriminated food [64], indicating that prior sensitization is either not required or occurs through other routes such as the skin.
While there is a paucity of recent studies evaluating FPE food triggers, older studies have consistently reported cow’s milk to be the main culprit [15,18,28,103]. In a case-series of 54 infants with cow’s milk FPE, co-allergy to soy was reported in 4/35 (11%) of those tested, and to wheat in 7/19 (37%) [28]. Other reported triggers included eggs (n = 2), bananas (n = 2), and meat (n = 1).
Finally, FPIAP is most frequently caused by indirect exposure to cow’s milk (and other foods) via breastmilk, occurring in exclusively breasted infants in over one-half of cases [84]. Somewhat less commonly, FPIAP can result from direct ingestion of cow’s milk (44%) or soy-based formula (7%) [83]. Other culprit foods include soy, egg, wheat and corn [12,84,85,89].

7. Diagnosis

The diagnosis of non-IgE-GI-FA remains, for the most part, a clinical one, with the exception of FPE, in which histological confirmation is usually required. Other etiologies presenting with a similar clinical picture should also be excluded. Optimal diagnosis and management may require the expertise of a multidisciplinary team (Figure 4).
The diagnosis of FPIES is established with the presence of a constellation of symptoms concordant with FPIES, and the resolution of symptoms upon the removal of offending foods from the diet. In an effort to standardize the diagnosis of acute FPIES in light of latest available data, recent international consensus guidelines based on expert opinion have defined major and minor criteria (Table 3) [68], although the accuracy of these diagnostic criteria has not yet been prospectively validated. While the OFC is no longer mandatory for diagnosis confirmation based on these criteria, it should be strongly considered when only a single episode has occurred, or when the causative food remains elusive. Tentative diagnostic criteria have also been proposed for chronic FPIES, with pathognomonic features being the rapid resolution of symptoms (within days) after the withdrawal of offending foods, and the acute presentation when the food is later reintroduced after a period of elimination [68]. In contrast to acute FPIES, the OFC is mandatory for chronic FPIES diagnosis, which is intended to decrease the frequent overdiagnosis found with this entity.
There are no accepted diagnostic criteria available for FPE and FPIAP, although some elements routinely used in clinical practice to help support the diagnosis of these diseases are listed in Table 3. FPE presents in young infants (<9 months) with typical symptoms of vomiting and intestinal malabsorption. In addition, a biopsy suggestive of FPE helps to confirm the diagnosis [81]. In FPIAP, the causative association between the rectal bleeding and antigenic food must be established with a resolution of symptoms upon the elimination of the offending foods, and, preferably, with the documented recurrence of symptoms when foods are re-introduced [106]. Furthermore, other causes of hematochezia, such as anal fissures, must be excluded.
Diagnosis of non-IgE-GI-FA may be delayed, because foods such as grains, vegetables and poultry are often perceived as being hypoallergenic, a misconception based on their infrequency as IgE-FA triggers, or because the reactions do not occur immediately after food ingestion, thus making the association more difficult. Furthermore, the lack of awareness of these entities by health care providers may also be a contributing factor [4,5].

8. Oral Food Challenge

OFCs represent the gold standard to confirm the diagnosis of FPIES, and are particularly important to avoid misdiagnosis with other common gastrointestinal diseases of infancy (Table 4). Of importance, OFC performed for suspected FPIES is considered a high-risk procedure. The consensus protocol is to administer 0.3 g/kg (0.06–0.6 g/kg) of food protein (max: 3 g of protein, 10 g of total food or 100 mL of liquid) in three equal doses over 30 min and observe the patient for 4–6 h for potential reactions [68]. In patients with severe reactions, a lower starting dose and a longer observation period may be used. Some centers have opted for performing a 1-dose OFC protocol instead, with success [93]. However, a graded OFC should always be performed in those patients with the presence of food sIgE, because of the risk of an immediate reaction [107].
While OFC protocols may differ slightly between centers, an overriding principle is that OFC should be administered by trained professionals in a facility equipped with intravenous (IV) fluid resuscitation material, as this intervention may be required in up to 50% of patients [68]. Whether a peripheral IV line should systematically be secured prior to undertaking the OFC remains controversial, but should be considered in patients in whom a difficult IV access is anticipated, and in those with a history of severe reactions. Nonetheless, the severity of the index reaction may not reliably predict the severity of the OFC reaction [93]. If symptoms are mild, PO fluids may be sufficient [19]. Ondansetron, a serotonin receptor antagonist used as a potent anti-emetic drug in patients receiving chemotherapy [108], may be helpful in mitigating acute symptoms when used as an adjunctive therapy [109,110]. Ondansetron should not be given to children under 6 months of age due to the lack of safety data, and should be administered with caution in predisposed patients due to the potential for QT interval prolongation. While methylprednisone has been used in an attempt to suppress the presumed cell-mediated inflammation, there are no studies to support its use. Furthermore, the current evidence does not support a role for epinephrine autoinjectors and antihistamines in the management of acute FPIES [68].
Supervised OFC is generally not required for the diagnosis of FPE and FPIAP [111]. However, in FPIAP, an early home challenge has been proposed to confirm the diagnosis after a short period of food elimination, to minimize the risk of misdiagnosis [13,112].

9. Paraclinical Investigations and Biomarkers

9.1. Laboratory Findings

Laboratory studies are somewhat helpful in supporting the diagnosis of non-IgE-GI-FA, although the findings are usually nonspecific. In FPIES, blood testing can uncover anemia, hypoalbuminemia, thrombocytosis, and a high white blood cell count with left shift [63,68]. With increased severity, metabolic acidosis and methemoglobinemia may also develop [113]. In FPE, iron-deficiency, anemia and hypoproteinemia are frequently present [16,114], while these are uncommon in FPIAP (<15%) [27,85,115,116]. Peripheral eosinophilia can be found in FPIES, FPE and FPIAP, although it is a more prominent feature in the two latter entities [28,58,63,83,85,117]. Lymphocyte transformation tests (LTT) remain experimental, and as such, are not recommended [118].

9.2. Allergy Testing

Allergy testing, which includes epicutaneous skin testing and the detection of serum food-specific IgE, is usually negative, and not warranted for the investigation of non-IgE-GI-FA [2,3,8]. However, up to 25% of FPIES patients will develop sIgE to their food trigger [23,26,64]. These patients have what is termed an atypical form of FPIES, which can present with a protracted course and may increase the risk of developing immediate allergic reactions to the culprit food [26,95]. Therefore, although allergy testing is usually not recommended at the first evaluation of FPIES, it can be performed during follow-up to exclude atypical FPIES, particularly before an OFC to adapt the protocol if need be [68]. Atopy patch testing (APT) has shown conflicting efficacy in the diagnosis of FPIES, with one study concluding that APT reliably predicted 85% of OFC outcomes [119], while two other studies found no such benefit [95,120]. These tests are therefore currently not recommended.

9.3. Stool Studies

When conducted in FPIES patients, stool evaluation may reveal nonspecific findings such as neutrophils, eosinophils, Charcot–Leyden crystals, and reducing substances [68]. Several studies have used occult blood testing to aid in the diagnosis of FPIAP [12,121], although occult blood may also be found in FPIES and other diseases. Likewise, while it is not specific to these diseases, fecal calprotectin was found to be elevated in both FPIES and FPIAP, indicative of gut mucosal inflammation [122,123,124,125]. However, this test should not be used before one year of age, due to the wide variability of values reported in infants, and the absence of validated normal ranges [126,127].

9.4. Radiologic Evaluation

Radiologic evaluation is not part of the usual disease workup, unless used to rule-out other disease processes. If abdominal X-rays are performed in children with FPIES, these may reveal nonspecific findings such as air-fluid levels, colon narrowing and thumbprinting, and duodenojejunal thickening of the plicae circulares [128]. Furthermore, intramural gas has also been reported, and may lead to a misdiagnosis of necrotizing enterocolitis (NEC) [129,130]. Recent ultrasonographic studies of 16 patients with non-IgE-GI-FA found that 100% of patients exhibited small intestinal wall thickening and poor peristalsis, which disappeared after culprit food elimination [131]. However, more studies are needed to corroborate the use of intestinal ultrasound before it can be used routinely.

9.5. Endoscopic Evaluation

Endoscopic evaluation is most important when FPE is suspected, as it supports the diagnosis. Histology findings typically phenocopy those seen in celiac disease, although they tend to be less severe [132]. These include varying degrees of jejunal villous atrophy and crypt hyperplasia [28,82], with the villus-to-crypt ratio being a sensitive marker of morphologic change due to jejunal damage [133]. In FPIAP, rectosigmoidoscopy may reveal mucosal congestion with petechial areas [134]. The inflammation is characterized by eosinophilic infiltration and lymphonodular hyperplasia [84,89,134]. The inflammatory cell infiltrate seen in FPIES includes lymphocytes, plasma cells, eosinophils, and mast cells [68].

9.6. Other Biomarkers

Interestingly, C-reactive protein (CRP), an acute phase reactant produced by the liver in response to the pro-inflammatory cytokine IL-6, has been reported to be marginally elevated following positive OFC in patients with FPIES [72,135,136,137]. However, while low elevation can be found, a high CRP (>20 mg/dL) should prompt one towards an alternative diagnosis [63]. Platelets, another acute phase reactant, are often elevated in FPIES, with one study reporting a significantly lower mean platelet volume (MVP) than in bacterial sepsis, which may help discriminate between both conditions [63].

10. Differential Diagnosis

The differential diagnoses of non-IgE-GI-FA are listed in Table 4. Broadly, entities that may mimic acute FPIES include those with acute vomiting in an ill-appearing child, while those resembling chronic FPIES and FPE comprise a variety of diseases leading to chronic GI symptoms with FTT. Alternative sources of hematochezia in infants can also simulate FPIAP, and should be excluded. Infectious causes such as viral and bacterial gastroenteritis are frequent in this age group, and can present with similar features as FPIES. In a moribund infant, the diagnostic workup should include sepsis evaluation. Helpful distinguishing features of acute FPIES include the timing after food ingestion, a history of repeated episodes, and the rapid resolution of symptoms within 24 h, which contrasts with the prolonged symptoms usually seen in infectious causes. Fever is not a common feature of FPIES, although it has been described in Japanese and Chinese studies, a phenotype that appears to be specific to these regions [72,135,138,139]. Monogenetic diseases, such as inborn errors of metabolism, primary immunodeficiencies, and cystic fibrosis, can also present with similar sepsis-like features and failure to thrive, and may not always be detected by newborn screening [140]. Mechanical obstruction caused by pyloric stenosis, intussusception, volvulus or Hirschsprung disease [141] may also present with acute and chronic vomiting, although in contrast to non-IgE-GI-FA, the symptoms will not be food-specific. Finally, IgE-FA, which can be life-threatening, is not always easily discernible from FPIES. However, the immediate occurrence of symptoms (within an hour of food ingestion), and other systemic features such as hives, angioedema, and respiratory symptoms, are uniquely present in IgE-FA.

11. Natural History

The natural history of non-IgE-GI-FA is mostly favorable, with the vast majority being outgrown by school age.
FPIES generally resolves before 3 to 5 years of age, but this may vary by type of food (i.e., solid vs. liquid) and geographical location [61]. An older age at diagnosis and atypical FPIES are associated with a prolonged course [26,75]. Furthermore, those with atypical FPIES are at increased risk of shifting from a non-IgE-mediated disease to an immediate, IgE-mediated phenotype [19,23,62,142,143]. The development of circulating sIgE in these patients with subsequent IgE-FA is perplexing, and may be indicative of some level of fluidity between the pathogenesis of both types of allergy. In line with this hypothesis, some cases of IgE-FA conversely shifting to a non-IgE-FA phenotype have also been described [144,145,146]. An alternative explanation is that the avoidance of the allergenic protein during a window of susceptibility in infancy may in fact increase the risk of developing IgE-FA in susceptible individuals [147,148,149].
In contrast to the permanent course seen in celiac disease, FPE is usually transitory and typically resolves by 1–2 years of age [28,79,150]. However, the disease may persist later into childhood in some cases [151,152,153].
Similarly, FPIAP rarely persists beyond 1–2 years of age [83]. In one large series of children with FPIAP, the mean age at which cow’s milk was successfully reintroduced was 11 months [12]. Notably, in 23 patients (15%), in whom the diet was not restricted at all, the symptoms eventually subsided in all patients, who subsequently tolerated cow’s milk throughout infancy, suggesting a benign course, despite continued exposure in a subset of patients [12]. However, recent studies suggest that patients with FPIAP may be at an increased risk of childhood functional GI disorder, especially in those with a more severe and protracted initial disease [154].

12. Management

12.1. Food Elimination

The cornerstone of the management of non-IgE-GI-FA is the removal of offending foods from the diet. Depending on the severity of the symptoms and on the quantity of triggering foods, two different strategies can be adopted. A “bottom-up approach”, in which causal foods are successively eliminated without broad restrictions of unsuspected triggers, is used in most cases. Indeed, one general principle of the management of food allergies is that the avoidance of causative foods only, without any restriction of foods that are tolerated, should be the norm. However, a “top-down approach” may be warranted in most severe cases where FTT and dehydration are prominent. This approach consists of an initial avoidance of a wide variety of foods, sometimes starting with an elemental diet, with the sequential reintroduction of individual foods, while carefully monitoring for recurrence of symptoms [10,155]. Counselling with a nutritionist is recommended because of the high risk for nutritional deficiencies (discussed below).
Cow’s milk is the most frequent trigger in FPIES, FPE and FPIAP, and should be replaced with an extensively hydrolyzed formula (EHF). While most patients are responsive to EHF, 10–20% of patients may require an amino acid-based formula (AAF) (Table 5) [12,21,24,68,156,157]. Of note, a change in stool consistency is frequent with hypoallergenic formulas, and is not indicative of ongoing colitis. In all cases, partially hydrolyzed formulas should be avoided because of their residual antigenic content, with a concentration of intact cow’s milk protein 1000–100,000-fold greater than that of EHF [158]. When tolerated, a soy-based formula is an acceptable alternative, although it is usually avoided because of the high co-reactivity with cow’s milk, seen in up to 40–60% of patients with FPIES, and in 10–30% of patients with FPE and FPIAP [96,158,159,160]. However, co-sensitization to cow’s milk and soy has mainly been reported in studies from the United States, and may be less frequent in other populations [19,62,104,105].
In contrast to FPIAP, symptomatic FPIES is very rare in the breastfed infant. Therefore, the mother should not avoid trigger foods unless symptoms are clearly documented [68,106]. Although no threshold dose has reliably been established in FPIES, only 0.15 g of protein/kg was sufficient to trigger 13/13 acute FPIES reactions to OFC in one study [119]. A strict avoidance of all offending foods should therefore be adopted. While avoidance of foods with precautionary allergen labeling (“may contain traces of”) is generally not necessary [68], it is usually recommended that extensively baked and processed foods be avoided in FPIES. Nevertheless, studies have shown that a number of patients with FPIES can tolerate these foods in baked form [161,162,163,164]. The long-term outcome of this practice is yet unknown, and OFC should be performed with caution, because some children may still react. Acute FPIES reactions should be treated as per positive OFC (see above). Epinephrine autoinjectors should only be prescribed if there is a concomitant IgE-mediated food allergy. The education of caregivers is central to a comprehensive management plan, and a written emergency plan of action should be given, as well as an explicative letter of the diagnosis and management plan to present to emergency room (ER) physicians who may not be familiar with FPIES. As per IgE-FA, a medical ID bracelet can be useful.
In FPIAP, the vast majority of exclusively breastfed infants will respond to the maternal elimination of all milk products (including butter) [84,165]. Occasionally, the elimination of multiple foods may be required, usually soy and/or egg [84]. Recent European guidelines recommend a 2–4 weeks maternal elimination diet, followed by an attempt to re-introduce foods in order to confirm the diagnosis [106]. In those in whom the maternal restriction diet is unsuccessful or too cumbersome, infants can be placed on an EHF instead. Because episodes of rectal bleeding in infants are often self-resolving, some authors have instead suggested using a “watch and wait” approach during the first month of symptoms, before undertaking an elimination diet [112]. In those with a symptoms duration that is greater than a month, hemoglobin levels should be assessed before undertaking a short cow’s milk elimination trial.

12.2. Nutritional Impact

It is essential to accurately diagnose non-IgE-GI-FA and to offer proper nutritional guidance, as both continued exposure and dietary manipulation can have detrimental effects on health. Efforts should therefore be put in place to limit unnecessary elimination diets, as even single-food avoidance can lead to significant nutritional deficiency [166], although those who avoid a greater number of foods may be at an increased risk [167]. Children with non-IgE-GI-FA can experience poor growth, which may be attributed to increased losses due to intestinal malabsorption and/or to limited food intake due to feeding difficulties, which are found in up to 30% of these patients [64,168]. Consequently, both weight [64,168,169] and height [169] may be affected. Deficient intake of indispensable micronutrients, including vitamin D, calcium, zinc, selenium [170,171,172,173], has also been reported, negatively impacting bone health [173].

12.3. Culprit Food Reintroduction

In the event of a positive OFC in FPIES, follow-up challenges should be performed, to determine whether FPIES has resolved. While there is no international consensus regarding the best time to perform an OFC, a repeat OFC every 12–18 months after the latest reaction is generally proposed in the United States, although this may vary depending on the type of food, the severity of the initial reaction, and distinct geographical characteristics [68].
In FPIAP, cow’s milk can be progressively re-introduced in a stepwise manner by 12 months of age in most patients [84,106,174,175]. If the diagnosis is unclear, re-introduction can be attempted earlier, because some infants with mild rectal bleeding may have a self-limiting course [11,13]. In patients with initially mild symptoms of FPE or FPIAP without any features suggestive of IgE-FA or FPIES, reintroduction can usually be done at home. In the case of failure, the trigger should be removed once again from the diet, with new attempts at reintroduction every six months.

12.4. Introduction of Weaning Foods

While there is no clear guidance regarding the ideal sequence of the introduction of complementary foods in patients with FPIES, current guidelines advocate in favor of introducing low-risk foods first, such as vegetables (broccoli, cauliflower) and fruits (blueberries, strawberries) [68]. Moderate and high-risk foods may then be serially introduced over the following months, when developmentally appropriate. Importantly, foods with different colors, flavors and textures should continuously be offered, in order to minimize the risk of aversive feeding behaviors. Furthermore, the introduction of iron-rich foods should also be encouraged, because the onset of FPIES often correlates with hemoglobin nadir, leaving patients vulnerable to anemia [176].
In FPE and FPIAP, food diversification can follow usual recommendations without any particular restrictions.

13. Quality of Life

While the impact of IgE-mediated food allergies on quality of life (QoL) has long been established, the effect of non-IgE-GI-FA has only been recently studied. Strikingly, one cross-sectional study of 52 children with non-IgE-GI-FA reported lower physical QoL scores compared to children with IgE-FA [177]. Similarly, another prospective observational study of 123 children with non-IgE-GI-FA revealed inferior QoL scores in all evaluated domains compared to children with sickle cell anemia, including spheres of physical functioning, emotional functioning, and worry [178]. Furthermore, QoL scores in those domains were also lower than in patients with intestinal failure. Unsurprisingly, a greater number of foods excluded was associated with lower QoL in both studies.

14. Role of Food Allergens in Other Common Pediatric Gastrointestinal Disorders

Food allergens may play a role in a subset of children with common gastrointestinal disorders, such as gastroesophageal reflux disease (GERD), irritable bowel syndrome (IBS), constipation and colic, although further studies are required to better elucidate the underlying pathomechanisms involved and whether these are immunologic and/or non-immunologic adverse reactions [10,179].
A subset of infants with GERD may have an underlying cow’s milk allergy, especially in patients with severe presentations, atopic dermatitis and FTT [179,180,181]. Cow’s milk feeding can result in gastric dysrhythmia, delayed gastric emptying, and increased episodes of reflux in milk-allergic infants [179,182,183]. However, there is no strong evidence supporting immunological mechanisms for GERD triggered by cow’s milk; the efficacy of hydrolysates in reducing regurgitations in infants with GERD may be due to enhanced gastric emptying compared to native cow’s milk protein [10,184].
IBS is a multifactorial gastrointestinal disorder which involves digestive symptoms that can be triggered by different foods, such as fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPS) [37]. IBS can overlap with non-celiac gluten sensitivity, with controlled studies showing an improvement of patients after the dietary exclusion of wheat [37]. Interestingly, children with atopic disease are at greater risk of developing IBS than non-atopic children [185]. A subset of patients with IBS have positive colonoscopic food allergen provocation with localized mucosal wheal and flare reactions [186] and the improvement of symptoms with cromoglycates [187], suggesting a potential role of food allergens in the pathogenesis of IBS. A recent study found the immediate disruption of the small intestinal barrier by confocal laser endomicroscopy after oral provocation with common allergens (wheat, milk, egg, soy) in skin test/specific IgE negative patients with IBS [188]. Further larger studies and randomized controlled trials are needed to better investigate the exact role and pathophysiology of food allergens as triggers in IBS patients.
The role of food allergens as a cause of constipation is controversial. Transition from breast milk to cow’s milk formula can be associated with mild constipation and increased stool hardness [189]. Prospective trials have found that cow’s milk free diets could improve constipation in a proportion of children, and children responding to this diet were more likely to be atopic [37]. A study in children found increased resting anal pressure and a reduced mucous gel layer, which could be attributed to allergic inflammation triggered by milk [190]. However, the potential effect of cow’s milk could be explained by non-immunologic mechanisms leading to changes in stool consistency [37].
Infantile colic is characterized by paroxysmal inconsolable crying during the early weeks of life [37]. The role of food allergens as triggers of colic is controversial, although food triggers may be relevant in a subset of infants with severe colic and atopic risk factors [37,191]. These infants may potentially benefit from a short trial of extensively hydrolyzed or soy formula [191]. However, a recent meta-analysis found sparse evidence for the effectiveness of dietary modifications for the treatment of infantile colic due to confounding factors (most cases of infantile colic spontaneously improve within a short time frame without elimination diets) [192]. Future larger studies are needed to better define the role of food allergens in infantile colic.

15. Future Perspectives

While tremendous advances have been made in our understanding of non-IgE-GI-FA in the past decade, many questions remain unanswered. For one, the pathogenesis of non-IgE-GI-FA remains poorly elucidated. Multiple factors have hindered the ability to properly study these diseases, including (i) their relatively low prevalence (ii) their occurrence in young infants, where access to blood cells is challenging, (iii) a disease process potentially localized to the GI tract, which requires invasive procedures for tissue sampling and (iv) a lack experimental animal models currently available. Nonetheless, a better understanding of the immunological components driving these diseases is a high priority, since it may help uncover noninvasive biomarkers for efficient diagnosis, which are currently lacking, and provide guidance towards future preventive and therapeutic opportunities.
As in most acquired diseases, the ideal goal in food allergy is prevention. Recent, ground-breaking research has demonstrated that the early introduction of allergenic foods such as peanuts [149] and eggs [193] may reduce the risk of subsequently developing IgE-FA. However, no such preventive measures have been found effective so far in FPIES [19,24]. In FPIAP, the concurrent use of formula while breastfeeding may be protective [12], although more studies are needed to confirm this.
If prevention cannot be achieved, another objective may be to accelerate the acquisition of oral tolerance. Preliminary studies indicate that probiotic supplementation may accelerate the resolution of non-IgE-GI-FA to cow’s milk [124,194,195]. While the introduction of baked egg and milk can facilitate the resolution of IgE-FA [196], it is unknown whether this phenomenon is also present in non-IgE-GI-FA, and if early exposition to baked forms could help prevent a shift to IgE-FA. Interestingly, the early introduction of solid foods (<5.5 months) may accelerate the resolution of FPIAP [85]. Oral immunotherapy (OIT) has not yet been attempted in non-IgE-GI-FA, potentially because spontaneous tolerance is achieved relatively early in most cases, although it may be an interesting therapeutic avenue to study in forms that persist beyond infancy. Finally, the blockade of cytokines implicated in FPIES such as TNFα, IL-1β and IL-6 are exciting potential therapeutic targets [52].

16. Conclusions

In summary, non-IgE-GI-FA are frequently implicated in causing gastrointestinal symptoms in children, and are likely underdiagnosed. Owing to a paucity of available literature, evidence-based protocols to diagnose and treat these diseases are lacking. Currently, diagnosis is made clinically, although diagnostic criteria are evolving to better capture the various phenotypes as our understanding of these diseases progresses. The management relies upon avoidance of dietary triggers, and requires a multidisciplinary approach. Future studies are needed to better understand pathogenesis, to determine additional biomarkers for better diagnosis, and to optimize management practices. Finally, food allergens may also be implicated in a subset of patients with common GI disorders, although the current evidence to support an underlying immunologic pathomechanism is limited.

Author Contributions

R.L. and F.G. performed the review of the literature, and both contributed to the writing of the manuscript. J.-C.C. supervised the writing of the manuscript. All authors discussed the manuscript and contributed to its final version. 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 have no conflicts of interest to declare.

Abbreviations

APTatopy patch test
CRPC reactive protein
FODMAPSfermentable oligosaccharides, disaccharides, monosaccharides and polyols
FPEfood protein-induced enteropathy
FPIAPfood protein-induced allergic proctocolitis
FPIESfood protein-induced enterocolitis syndrome
FTTfailure to thrive
GERDgastroesophageal reflux disease
IBSirritable bowel syndrome
IgEimmunoglobulin E
IgE-GAIgE-mediated food allergy
LTTlymphocyte transformation test
MPVmean platelet volume
NECnecrotizing enterocolitis
non-IgE-GI-FAnon-immunoglobulin E-mediated gastrointestinal food allergic disorders
OFCoral food challenge
QoLquality of life
sIgEserum food-specific IgE

References

  1. Heine, R.G. Gastrointestinal Food Allergies. Chem. Immunol. Allergy 2015, 101, 171–180. [Google Scholar] [CrossRef]
  2. Fenton, M. Guidelines for the diagnosis and management of food allergy in the United States. Clin. Transl. Allergy 2011, 1, S10. [Google Scholar] [CrossRef] [Green Version]
  3. Sampson, H.A.; Aceves, S.; Bock, S.A.; James, J.; Jones, S.; Lang, D.; Nadeau, K.; Nowak-Węgrzyn, A.; Oppenheimer, J.; Perry, T.T.; et al. Food allergy: A practice parameter update—2014. J. Allergy Clin. Immunol. 2014, 134, 1016–1025.e43. [Google Scholar] [CrossRef] [PubMed]
  4. Comberiati, P.; Landi, M.; Martelli, A.; Piacentini, G.; Capristo, C.; Paiola, G.; Peroni, D. Awareness of allergic enterocolitis among primary-care paediatricians: A web-based pilot survey. Allergol. et Immunopathol. 2016, 44, 461–466. [Google Scholar] [CrossRef] [PubMed]
  5. Greenhawt, M.; Bird, J.A.; Nowak-Wegrzyn, A. Trends in Provider Management of Patients with Food Protein–Induced Enterocolitis Syndrome. J. Allergy Clin. Immunol. Pr. 2017, 5, 1319–1324.e12. [Google Scholar] [CrossRef] [PubMed]
  6. Tordesillas, L.; Berin, M.C.; Sampson, H.A. Immunology of Food Allergy. Immunity 2017, 47, 32–50. [Google Scholar] [CrossRef] [Green Version]
  7. Waserman, S.; Bégin, P.; Watson, W. IgE-mediated food allergy. Allergy, Asthma Clin. Immunol. 2018, 14, 55. [Google Scholar] [CrossRef] [Green Version]
  8. Burks, A.W.; 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, 906–920. [Google Scholar] [CrossRef]
  9. Koutri, E.; Papadopoulou, A. Eosinophilic Gastrointestinal Diseases in Childhood. Ann. Nutr. Metab. 2018, 73, 18–28. [Google Scholar] [CrossRef]
  10. Jean-Christoph, C.; Szajewska, H.; Shamir, R.; Nowak-Wegrzyn, A. Non-IgE-mediated gastrointestinal food allergies in children. Pediatr. Allergy Immunol. 2016, 28, 6–17. [Google Scholar] [CrossRef]
  11. Elizur, A.; Cohen, M.; Goldberg, M.R.; Rajuan, N.; Cohen, A.; Leshno, M.; Katz, Y. Cow’s milk associated rectal bleeding: A population based prospective study. Pediatr. Allergy Immunol. 2012, 23, 766–770. [Google Scholar] [CrossRef] [PubMed]
  12. Martin, V.M.; Virkud, Y.V.; Seay, H.; Hickey, A.; Ndahayo, R.; Rosow, R.; Southwick, C.; Elkort, M.; Gupta, B.; Kramer, E.; et al. Prospective Assessment of Pediatrician-Diagnosed Food Protein–Induced Allergic Proctocolitis by Gross or Occult Blood. J. Allergy Clin. Immunol. Pr. 2020, 8, 1692.e1–1699.e1. [Google Scholar] [CrossRef] [PubMed]
  13. Arvola, T.; Galanakis, E.; Bitsori, M.; Dimitriou, H.; Giannakopoulou, C.; Karkavitsas, N.S.; Kalmanti, M. Rectal Bleeding in Infancy: Clinical, Allergological, and Microbiological Examination. Pediatrics 2006, 117, 760–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. A Xanthakos, S.; Schwimmer, J.B.; Melin-Aldana, H.; Rothenberg, M.E.; Witte, D.P.; Cohen, M.B. Prevalence and Outcome of Allergic Colitis in Healthy Infants with Rectal Bleeding: A Prospective Cohort Study. J. Pediatr. Gastroenterol. Nutr. 2005, 41, 16–22. [Google Scholar] [CrossRef]
  15. Verkasalo, M.; Kuitunen, P.; Savilahti, E.; Tiilikainen, A. CHANGING PATTERN OF COW’S MILK INTOLERANCE: An Analysis of the Occurrence and Clinical Course in the 60s and mid-70s. Acta Paediatr. 1981, 70, 289–295. [Google Scholar] [CrossRef]
  16. Savilahti, E. Food-Induced Malabsorption Syndromes. J. Pediatr. Gastroenterol. Nutr. 2000, 30, S61–S66. [Google Scholar] [CrossRef]
  17. Kokkonen, J.; Haapalahti, M.; Tikkanen, S.; Karttunen, R.; Savilahti, E. Gastrointestinal complaints and diagnosis in children: A population-based study. Acta Paediatr. 2004, 93, 880–886. [Google Scholar] [CrossRef]
  18. Vitoria, J.C.; Sojo, A.; Rodriguez-Soriano, J. Changing pattern of cow’s milk protein intolerance. Acta Paediatr. Scand 1990, 79, 566–567. [Google Scholar] [CrossRef]
  19. Katz, Y.; Goldberg, M.R.; Rajuan, N.; Cohen, A.; Leshno, M. The prevalence and natural course of food protein–induced enterocolitis syndrome to cow’s milk: A large-scale, prospective population-based study. J. Allergy Clin. Immunol. 2011, 127, 647–653.e3. [Google Scholar] [CrossRef]
  20. Alonso, S.B.; Ezquiaga, J.G.; Berzal, P.T.; Tardón, S.D.; José, M.M.S.; López, P.A.; Bermejo, T.B.; Teruel, S.Q.; Zudaire, L.; Ángel, E.; et al. Food protein–induced enterocolitis syndrome: Increased prevalence of this great unknown—results of the PREVALE study. J. Allergy Clin. Immunol. 2019, 143, 430–433. [Google Scholar] [CrossRef] [Green Version]
  21. Mehr, S.; Frith, K.; Barnes, E.H.; Campbell, D.E.; Allen, K.J.; Gold, M.; Joshi, P.; Kakakios, A.; Loh, R.; Peake, J.; et al. Food protein–induced enterocolitis syndrome in Australia: A population-based study, 2012–2014. J. Allergy Clin. Immunol. 2017, 140, 1323–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Nowak-Wegrzyn, A.; Warren, C.M.; Brown-Whitehorn, T.; Cianferoni, A.; Schultz-Matney, F.; Gupta, R.S. Food protein-induced enterocolitis syndrome in the US population-based study. J. Allergy Clin. Immunol. 2019, 144, 1128–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Sicherer, S.H.; Eigenmann, P.A.; Sampson, H.A. Clinical features of food protein–induced enterocolitis syndrome. J. Pediatr. 1998, 133, 214–219. [Google Scholar] [CrossRef]
  24. Nowak-Wegrzyn, A.; Sampson, H.A.; Wood, R.A.; Sicherer, S.H. Food protein-induced enterocolitis syndrome caused by solid food proteins. Pediatrics 2003, 111, 829–835. [Google Scholar] [CrossRef] [Green Version]
  25. Maciag, M.C.; Bartnikas, L.M.; Sicherer, S.H.; Herbert, L.J.; Young, M.C.; Matney, F.; Westcott-Chavez, A.A.; Petty, C.R.; Phipatanakul, W.; Bingemann, T.A. A Slice of Food Protein–Induced Enterocolitis Syndrome (FPIES): Insights from 441 Children with FPIES as Provided by Caregivers in the International FPIES Association. J. Allergy Clin. Immunol. Pr. 2020, 8, 1702–1709. [Google Scholar] [CrossRef]
  26. Jean-Christoph, C.; Ford, L.S.; Sickles, L.; Järvinen, K.M.; Sicherer, S.H.; Sampson, H.A.; Nowak-Wegrzyn, A. Clinical features and resolution of food protein–induced enterocolitis syndrome: 10-year experience. J. Allergy Clin. Immunol. 2014, 134, 382.e4–389.e4. [Google Scholar] [CrossRef]
  27. Lucarelli, S.; Di Nardo, G.; Lastrucci, G.; D’Alfonso, Y.; Marcheggiano, A.; Federici, T.; Frediani, S.; Frediani, T.; Cucchiara, S. Allergic proctocolitis refractory to maternal hypoallergenic diet in exclusively breast-fed infants: A clinical observation. BMC Gastroenterol. 2011, 11, 82. [Google Scholar] [CrossRef] [Green Version]
  28. Kuitunen, P.; Visakorpi, J.K.; Savilahti, E.; Pelkonen, P. Malabsorption syndrome with cow’s milk intolerance. Clinical findings and course in 54 cases. Arch. Dis. Child 1975, 50, 351–356. [Google Scholar]
  29. Wakiguchi, H.; Hasegawa, S.; Kaneyasu, H.; Kajimoto, M.; Fujimoto, Y.; Hirano, R.; Katsura, S.; Matsumoto, K.; Ichiyama, T.; Ohga, S. Long-lasting non-IgE-mediated gastrointestinal cow’s milk allergy in infants with Down syndrome. Pediatr. Allergy Immunol. 2015, 26, 821–823. [Google Scholar] [CrossRef]
  30. Kusters, M.A.A.; Verstegen, R.H.; Gemen, E.F.A.; De Vries, E. Intrinsic defect of the immune system in children with Down syndrome: A review. Clin. Exp. Immunol. 2009, 156, 189–193. [Google Scholar] [CrossRef]
  31. Nateghi Rostami, M.; Douraghi, M.; Mohammadi, A.M.; Nikmanesh, B. Altered serum pro-inflammatory cytokines in children with Down’s syndrome. Eur. Cytokine Netw. 2012, 23, 64–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sullivan, K.D.; Evans, N.; Pandey, A.; Hraha, T.H.; Smith, K.P.; Markham, N.; Rachubinski, A.L.; Wolter-Warmerdam, K.; Hickey, F.; Espinosa, J.M.; et al. Trisomy 21 causes changes in the circulating proteome indicative of chronic autoinflammation. Sci. Rep. 2017, 7, 14818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lin, X.P.; Magnusson, J.; Ahlstedt, S.; Dahlman-Höglund, A.; Hanson, L.Å.; Magnusson, O.; Bengtsson, U.; Telemo, E. Local allergic reaction in food-hypersensitive adults despite a lack of systemic food-specific IgE. J. Allergy Clin. Immunol. 2002, 109, 879–887. [Google Scholar] [CrossRef] [PubMed]
  34. Van Sickle, G.J.; Powell, G.K.; McDonald, P.J.; Goldblum, R.M. Milk- and soy protein-induced enterocolitis: Evidence for lymphocyte sensitization to specific food proteins. Gastroenterology 1985, 88, 1915–1921. [Google Scholar] [CrossRef]
  35. Hoffman, K.M.; Ho, D.G.; Sampson, H.A. Evaluation of the usefulness of lymphocyte proliferation assays in the diagnosis of allergy to cow’s milk. J. Allergy Clin. Immunol. 1997, 99, 360–366. [Google Scholar] [CrossRef]
  36. Morita, H.; Nomura, I.; Orihara, K.; Yoshida, K.; Akasawa, A.; Tachimoto, H.; Ohtsuka, Y.; Namai, Y.; Futamura, M.; Shoda, T.; et al. Antigen-specific T-cell responses in patients with non–IgE-mediated gastrointestinal food allergy are predominantly skewed to TH2. J. Allergy Clin. Immunol. 2013, 131, 590.e6–592.e6. [Google Scholar] [CrossRef]
  37. Caubet, J.C.; Bencharitiwong, R.; Ross, A.; Sampson, H.A.; Berin, M.C.; Nowak-Wegrzyn, A. Humoral and cellular responses to casein in patients with food protein-induced enterocolitis to cow’s milk. J. Allergy Clin. Immunol. 2017, 139, 572–583. [Google Scholar] [CrossRef] [Green Version]
  38. Goswami, R.; Blazquez, A.B.; Kosoy, R.; Rahman, A.; Nowak-Wegrzyn, A.; Berin, M.C. Systemic innate immune activation in food protein–induced enterocolitis syndrome. J. Allergy Clin. Immunol. 2017, 139, 1885.e9–1896.e9. [Google Scholar] [CrossRef]
  39. Benlounes, N.; Dupont, C.; Candalh, C.; Blaton, M.; Darmon, N.; Desjeux, J.; Heyman, M. The threshold for immune cell reactivity to milk antigens decreases in cow’s milk allergy with intestinal symptoms. J. Allergy Clin. Immunol. 1996, 98, 781–789. [Google Scholar] [CrossRef]
  40. González-Delgado, P.; Caparrós, E.; Moreno, M.V.; Clemente, F.; Flores, E.; Velásquez, L.; Rubio, G.; Fernandez, J. Clinical and immunological characteristics of a pediatric population with food protein-induced enterocolitis syndrome (FPIES) to fish. Pediatr. Allergy Immunol. 2016, 27, 269–275. [Google Scholar] [CrossRef]
  41. Shek, L.P.; Bardina, L.; Castro, R.; Sampson, H.A.; Beyer, K. Humoral and cellular responses to cow milk proteins in patients with milk-induced IgE-mediated and non-IgE-mediated disorders. Allergy 2005, 60, 912–919. [Google Scholar] [CrossRef] [PubMed]
  42. Konstantinou, G.N.; Bencharitiwong, R.; Grishin, A.; Caubet, J.C.; Bardina, L.; Sicherer, S.H.; Sampson, H.A.; Nowak-Wegrzyn, A. The role of casein-specific IgA and TGF-beta in children with food protein-induced enterocolitis syndrome to milk. Pediatr. Allergy Immunol. 2014, 25, 651–656. [Google Scholar] [CrossRef] [PubMed]
  43. Beyer, K.; Castro, R.; Birnbaum, A.; Benkov, K.; Pittman, N.; Sampson, H.A. Human milk-specific mucosal lymphocytes of the gastrointestinal tract display a TH2 cytokine profile. J. Allergy Clin. Immunol. 2002, 109, 707–713. [Google Scholar] [CrossRef]
  44. Kapel, N.; Matarazzo, P.; Haouchine, D.; Abiola, N.; Guerin, S.; Magne, D.; Gobert, J.G.; Dupont, C. Fecal tumor necrosis factor alpha, eosinophil cationic protein and IgE levels in infants with cow’s milk allergy and gastrointestinal manifestations. Clin. Chem. Lab. Med. 1999, 37, 29–32. [Google Scholar] [CrossRef] [PubMed]
  45. Chung, H.L.; Hwang, J.B.; Park, J.J.; Kim, S.G. Expression of transforming growth factor beta1, transforming growth factor type I and II receptors, and TNF-alpha in the mucosa of the small intestine in infants with food protein-induced enterocolitis syndrome. J. Allergy Clin. Immunol. 2002, 109, 150–154. [Google Scholar] [CrossRef] [PubMed]
  46. Benlounes, N.; Candalh, C.; Matarazzo, P.; Dupont, C.; Heyman, M. The time-course of milk antigen–induced TNF-α secretion differs according to the clinical symptoms in children with cow’s milk allergy. J. Allergy Clin. Immunol. 1999, 104, 863–869. [Google Scholar] [CrossRef]
  47. Heyman, M.; Darmon, N.; Dupont, C.; Dugas, B.; Hirribaren, A.; Blaton, M.A.; Desjeux, J.F. Mononuclear cells from infants allergic to cow’s milk secrete tumor necrosis factor alpha, altering intestinal function. Gastroenterology 1994, 106, 1514–1523. [Google Scholar] [CrossRef]
  48. Jean-Christoph, C.; Nowak-Wegrzyn, A. Current understanding of the immune mechanisms of food protein-induced enterocolitis syndrome. Expert Rev. Clin. Immunol. 2011, 7, 317–327. [Google Scholar] [CrossRef]
  49. Mori, F.; Barni, S.; Cianferoni, A.; Pucci, N.; De Martino, M.; Novembre, E. Cytokine Expression in CD3+ Cells in an Infant with Food Protein-Induced Enterocolitis Syndrome (FPIES): Case Report. Clin. Dev. Immunol. 2009, 2009, 1–4. [Google Scholar] [CrossRef] [Green Version]
  50. Kimura, M.; Ito, Y.; Shimomura, M.; Morishita, H.; Meguro, T.; Adachi, Y.; Seto, S. Cytokine profile after oral food challenge in infants with food protein-induced enterocolitis syndrome. Allergol. Int. 2017, 66, 452–457. [Google Scholar] [CrossRef]
  51. Karlsson, M.R.; Rugtveit, J.; Brandtzaeg, P. Allergen-responsive CD4+CD25+ regulatory T cells in children who have outgrown cow’s milk allergy. J. Exp. Med. 2004, 199, 1679–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Mehr, S.; Lee, E.; Hsu, P.; Anderson, D.; De Jong, E.; Bosco, A.; Campbell, D. Innate immune activation occurs in acute food protein-induced enterocolitis syndrome reactions. J. Allergy Clin. Immunol. 2019, 144, 600–602.e2. [Google Scholar] [CrossRef] [Green Version]
  53. McDonald, P.J.; Goldblum, R.M.; Van Sickle, G.J.; Powell, G.K. Food Protein-induced Enterocolitis: Altered Antibody Response to Ingested Antigen. Pediatr. Res. 1984, 18, 751–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Khoshoo, V.; Bhan, M.K.; Kumar, R.; Arora, N.K.; Stintzing, G. Is cow’s milk protein sensitive enteropathy a cell mediated immunological phenomenon? Acta Paediatr. Scand 1991, 80, 1092–1093. [Google Scholar] [CrossRef] [PubMed]
  55. Nagata, S.; Yamashiro, Y.; Ohtsuka, Y.; Shioya, T.; Oguchi, S.; Shimizu, T.; Maeda, M. Quantitative Analysis and Immunohistochemical Studies on Small Intestinal Mucosa of Food-Sensitive Enteropathy. J. Pediatr. Gastroenterol. Nutr. 1995, 20, 44–48. [Google Scholar] [CrossRef]
  56. Augustin, M.T.; Kokkonen, J.; Karttunen, T.J. Duodenal cytotoxic lymphocytes in cow’s milk protein sensitive enteropathy and coeliac disease. Scand J. Gastroenterol. 2005, 40, 1398–1406. [Google Scholar] [CrossRef]
  57. Goldman, H.; Proujansky, R. Allergic Proctitis and Gastroenteritis in Children. Am. J. Surg. Pathol. 1986, 10, 75–86. [Google Scholar] [CrossRef]
  58. Odze, R.D.; Bines, J.; Leichtner, A.M.; Goldman, H.; Antonioli, D.A. Allergic proctocolitis in infants: A prospective clinicopathologic biopsy study. Hum. Pathol. 1993, 24, 668–674. [Google Scholar] [CrossRef]
  59. Untersmayr, E.; Schöll, I.; Swoboda, I.; Beil, W.J.; Förster-Waldl, E.; Walter, F.; Riemer, A.; Kraml, G.; Kinaciyan, T.; Spitzauer, S.; et al. Antacid medication inhibits digestion of dietary proteins and causes food allergy: A fish allergy model in BALB/c mice. J. Allergy Clin. Immunol. 2003, 112, 616–623. [Google Scholar] [CrossRef]
  60. Díaz, J.J.; Espín, B.; Segarra, O.; Domínguez-Ortega, G.; Blasco-Alonso, J.; Cano, B.; Rayo, A.; Moreno, A. Food Protein-induced Enterocolitis Syndrome. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 232–236. [Google Scholar] [CrossRef]
  61. Caubet, J.; Cianferoni, A.; Groetch, M.; Nowak-Wegrzyn, A. Food protein-induced enterocolitis syndrome. Clin. Exp. Allergy 2019, 49, 1178–1190. [Google Scholar] [CrossRef] [PubMed]
  62. Delahaye, C.; Chauveau, A.; Kiefer, S.; Dumond, P. Food protein-induced enterocolitis syndrome (FPIES) in 14 children. Arch. Pediatr. 2017, 24, 310–316. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, E.; Barnes, E.H.; Mehr, S.; Campbell, D.E. Differentiating Acute Food Protein–Induced Enterocolitis Syndrome From Its Mimics: A Comparison of Clinical Features and Routine Laboratory Biomarkers. J. Allergy Clin. Immunol. Pr. 2019, 7, 471.e3–478.e3. [Google Scholar] [CrossRef]
  64. Su, K.-W.; Patil, S.; Stockbridge, J.; Martin, V.; Virkud, Y.; Huang, J.L.; Shreffler, W.; Yuan, Q. Food aversion and poor weight gain in food protein-induced enterocolitis syndrome: A retrospective study. J. Allergy Clin. Immunol. 2020, 145, AB52. [Google Scholar] [CrossRef]
  65. Blackman, A.C.; Anvari, S.; Davis, C.M.; Anagnostou, A. Emerging triggers of food protein–induced enterocolitis syndrome. Ann. Allergy, Asthma Immunol. 2019, 122, 407–411. [Google Scholar] [CrossRef] [PubMed]
  66. Ludman, S.; Harmon, M.; Whiting, D.; Du Toit, G. Clinical presentation and referral characteristics of food protein-induced enterocolitis syndrome in the United Kingdom. Ann. Allergy, Asthma Immunol. 2014, 113, 290–294. [Google Scholar] [CrossRef]
  67. Sopo, S.M.; Giorgio, V.; Iacono, I.D.; Novembre, E.; Mori, F.; Onesimo, R. A multicentre retrospective study of 66 Italian children with food protein-induced enterocolitis syndrome: Different management for different phenotypes. Clin. Exp. Allergy 2012, 42, 1257–1265. [Google Scholar] [CrossRef]
  68. Nowak-Wegrzyn, A.; Chehade, M.; Groetch, M.E.; Spergel, J.M.; Wood, R.A.; Allen, K.; Atkins, D.; Bahna, S.; Barad, A.V.; Berin, C.; et al. International consensus guidelines for the diagnosis and management of food protein-induced enterocolitis syndrome: Executive summary-Workgroup Report of the Adverse Reactions to Foods Committee, American Academy of Allergy, Asthma & Immunology. J. Allergy Clin. Immunol. 2017, 139, 1111 e4–1126 e4. [Google Scholar]
  69. Sicherer, S.H. Food protein-induced enterocolitis syndrome: Case presentations and management lessons. J. Allergy Clin. Immunol. 2005, 115, 149–156. [Google Scholar] [CrossRef]
  70. Monti, G.; Castagno, E.; Liguori, S.A.; Lupica, M.M.; Tarasco, V.; Viola, S.; Tovo, P.-A. Food protein–induced enterocolitis syndrome by cow’s milk proteins passed through breast milk. J. Allergy Clin. Immunol. 2011, 127, 679–680. [Google Scholar] [CrossRef]
  71. Kaya, A.; Toyran, M.; Civelek, E.; Mısırlıoglu, E.D.; Kırsaçlıoglu, C.T.; Kocabaş, C.N. Food Protein-Induced Enterocolitis Syndrome In Two Exclusively Breastfed Infants. Pediatr. Allergy Immunol. 2016, 27, 749–750. [Google Scholar] [CrossRef]
  72. Nomura, I.; Morita, H.; Hosokawa, S.; Hoshina, H.; Fukuie, T.; Watanabe, M.; Ohtsuka, Y.; Shoda, T.; Terada, A.; Takamasu, T.; et al. Four distinct subtypes of non–IgE-mediated gastrointestinal food allergies in neonates and infants, distinguished by their initial symptoms. J. Allergy Clin. Immunol. 2011, 127, 685.e8–688.e8. [Google Scholar] [CrossRef] [PubMed]
  73. Du, Y.J.; Nowak-Wegrzyn, A.; Vadas, P. FPIES in adults. Ann. Allergy, Asthma Immunol. 2018, 121, 736–738. [Google Scholar] [CrossRef] [PubMed]
  74. Fernandes, B.N.; Boyle, R.J.; Gore, C.; Simpson, A.; Custovic, A. Food protein–induced enterocolitis syndrome can occur in adults. J. Allergy Clin. Immunol. 2012, 130, 1199–1200. [Google Scholar] [CrossRef] [PubMed]
  75. Tan, J.A.; Smith, W.B. Non–IgE-mediated gastrointestinal food hypersensitivity syndrome in adults. J. Allergy Clin. Immunol. Pr. 2014, 2, 355.e1–357.e1. [Google Scholar] [CrossRef]
  76. Gonzalez-Delgado, P.; Caparrós, E.; Moreno, M.V.; Cueva, B.; Fernández, J. Food protein–induced enterocolitis-like syndrome in a population of adolescents and adults caused by seafood. J. Allergy Clin. Immunol. Pr. 2019, 7, 670–672. [Google Scholar] [CrossRef]
  77. Ichimura, S.; Kakita, H.; Asai, S.; Mori, M.; Takeshita, S.; Ueda, H.; Muto, T.; Kondo, T.; Yamada, Y. A Rare Case of Fetal Onset, Food Protein-Induced Enterocolitis Syndrome. Neonatology 2019, 116, 376–379. [Google Scholar] [CrossRef]
  78. Faber, M.R.; Rieu, P.; A Semmekrot, B.; Krieken, J.H.J.M.; Tolboom, J.J.M.; Draaisma, J.M.T. Allergic colitis presenting within the first hours of premature life. Acta Paediatr. 2007, 94, 1514–1515. [Google Scholar] [CrossRef]
  79. Walker-Smith, J. Cow milk-sensitive enteropathy: Predisposing factors and treatment. J. Pediatr. 1992, 121, S111–S115. [Google Scholar] [CrossRef]
  80. Walker-Smith, J.; Harrison, M.; Kilby, A.; Phillips, A.; France, N. Cows’ milk-sensitive enteropathy. Arch. Dis. Child. 1978, 53, 375–380. [Google Scholar] [CrossRef]
  81. Iyngkaran, N.; Robinson, M.J.; Prathap, K.; Sumithran, E.; Yadav, M. Cows’ milk protein-sensitive enteropathy. Combined clinical and histological criteria for diagnosis. Arch. Dis. Child. 1978, 53, 20–26. [Google Scholar] [CrossRef] [Green Version]
  82. Iyngkaran, N.; Abdin, Z.; Davis, K.; Boey, C.; Prathap, K.; Yadav, M.; Lam, S.; Puthucheary, S. Acquired carbohydrate intolerance and cow milk protein-sensitive enteropathy in young infants. J. Pediatr. 1979, 95, 373–378. [Google Scholar] [CrossRef]
  83. Lozinsky, A.C.; Morais, M.B. Eosinophilic colitis in infants. J. Pediatr. (Rio J) 2014, 90, 16–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lake, A.M. Food-Induced Eosinophilic Proctocolitis. J. Pediatr. Gastroenterol. Nutr. 2000, 30, S58–S60. [Google Scholar] [CrossRef] [PubMed]
  85. Cetinkaya, P.G.; Kahveci, M.; Karaatmaca, B.; Esenboga, S.; Sahiner, U.M.; Sekerel, B.E.; Soyer, O. Predictors for late tolerance development in food protein-induced allergic proctocolitis. Allergy Asthma Proc. 2020, 41, e11–e18. [Google Scholar] [CrossRef]
  86. Erdem, S.; Nacaroglu, H.; Karaman, S.; Erdur, C.; Karkıner, C.; Can, D. Tolerance development in food protein-induced allergic proctocolitis: Single centre experience. Allergol. et Immunopathol. 2017, 45, 212–219. [Google Scholar] [CrossRef] [PubMed]
  87. Kaya, A.; Toyran, M.; Civelek, E.; Misirlioğlu, E.; Kirsaclioglu, C.; Kocabaş, C.N.; Kırsaçlıoğlu, C. Characteristics and Prognosis of Allergic Proctocolitis in Infants. J. Pediatr. Gastroenterol. Nutr. 2015, 61, 1. [Google Scholar] [CrossRef] [PubMed]
  88. Sierra Salinas, C.; Blasco Alonso, J.; Olivares Sanchez, L.; Barco Galvez, A.; del Rio Mapelli, L. Dietary protein-induced proctocolitis in childhood. Am. J. Gastroenterol. 2008, 103, 2605–2612. [Google Scholar]
  89. Ravelli, A.; Villanacci, V.; Chiappa, S.; Bolognini, S.; Manenti, S.; Fuoti, M. Dietary protein-induced proctocolitis in childhood. Am. J. Gastroenterol. 2008, 103, 2605–2612. [Google Scholar] [CrossRef] [PubMed]
  90. Høst, A.; Halken, S.; Jacobsen, H.P.; Christensen, A.E.; Herskind, A.M.; Plesner, K.B. Clinical course of cow’s milk protein allergy/intolerance and atopic diseases in childhood. Pediatr. Allergy Immunol. 2002, 13, 23–28. [Google Scholar] [CrossRef]
  91. Vanto, T.; Helppila, 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]
  92. Santos, A.F.; 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]
  93. Wang, K.Y.; Lee, J.; Cianferoni, A.; Ruffner, M.A.; Dean, A.; Molleston, J.M.; Pawlowski, N.A.; Heimall, J.; Saltzman, R.W.; Ram, G.S.; et al. Food Protein–Induced Enterocolitis Syndrome Food Challenges: Experience from a Large Referral Center. J. Allergy Clin. Immunol. Pr. 2019, 7, 444–450. [Google Scholar] [CrossRef] [PubMed]
  94. Hwang, J.-B.; Sohn, S.M.; Kim, A.S. Prospective follow-up oral food challenge in food protein-induced enterocolitis syndrome. Arch. Dis. Child. 2008, 94, 425–428. [Google Scholar] [CrossRef] [PubMed]
  95. Ruffner, M.A.; Ruymann, K.; Barni, S.; Cianferoni, A.; Brown-Whitehorn, T.; Spergel, J.M. Food Protein-induced Enterocolitis Syndrome: Insights from Review of a Large Referral Population. J. Allergy Clin. Immunol. Pr. 2013, 1, 343–349. [Google Scholar] [CrossRef]
  96. Burks, A.W.; Casteel, H.B.; Fiedorek, S.C.; Williams, L.W.; Pumphrey, C.L. Prospective oral food challenge study of two soybean protein isolates in patients with possible milk or soy protein enterocolitis. Pediatr. Allergy Immunol. 1994, 5, 40–45. [Google Scholar] [CrossRef]
  97. Mehr, S.; Kakakios, A.; Frith, K.; Kemp, A.S. Food Protein-Induced Enterocolitis Syndrome: 16-Year Experience. Pediatrics 2009, 123, 459–464. [Google Scholar] [CrossRef]
  98. Mehr, S.; Kakakios, A.M.; Kemp, A.S. Rice: A common and severe cause of food protein-induced enterocolitis syndrome. Arch. Dis. Child. 2009, 94, 220–223. [Google Scholar] [CrossRef]
  99. Hojsak, I.; Kljaić-Turkalj, M.; Mišak, Z.; Kolacek, S. Rice protein-induced enterocolitis syndrome. Clin. Nutr. 2006, 25, 533–536. [Google Scholar] [CrossRef]
  100. Sopo, S.M.; Monaco, S.; Badina, L.; Barni, S.; Longo, G.; Novembre, E.; Viola, S.; Monti, G. Food protein-induced enterocolitis syndrome caused by fish and/or shellfish in Italy. Pediatr. Allergy Immunol. 2015, 26, 731–736. [Google Scholar] [CrossRef]
  101. Vazquez-Ortiz, M.; Machinena, A.; Dominguez, O.; Alvaro, M.; Calvo-Campoverde, K.; Giner, M.T.; Jiménez-Feijoo, R.; Lozano, J.; Piquer, M.; Dias, M.; et al. Food protein–induced enterocolitis syndrome to fish and egg usually resolves by age 5 years in Spanish children. J. Allergy Clin. Immunol. Pr. 2017, 5, 512.e1–515.e1. [Google Scholar] [CrossRef] [PubMed]
  102. Mehr, S.; Frith, K.; Campbell, D.E. Epidemiology of food protein-induced enterocolitis syndrome. Curr. Opin. Allergy Clin. Immunol. 2014, 14, 208–216. [Google Scholar] [CrossRef] [PubMed]
  103. Manuel, P.D.; Walker-Smith, J.A.; Soeparto, P. Cow’s milk sensitive enteropathy in Indonesian infants. Lancet 1980, 2, 1365–1366. [Google Scholar] [CrossRef]
  104. Ajami, R.I.P.; Sari, S.K.C.; Mazas, Y.A.; Calvo, J.B.; Abadía, M.I.G. Experience in food protein-induced enterocolitis syndrome in a paediatric allergy clinic. Anales de Pediatría 2020. [Google Scholar] [CrossRef]
  105. Yilmaz, E.A.; Soyer, O.; Cavkaytar, O.; Karaatmaca, B.; Buyuktiryaki, B.; Sahiner, U.M.; Sekerel, B.E.; Sackesen, C. Characteristics of children with food protein-induced enterocolitis and allergic proctocolitis. Allergy Asthma Proc. 2017, 38, 54–62. [Google Scholar] [CrossRef]
  106. Meyer, R.W.; Lozinsky, A.C.; Fleischer, D.M.; Vieira, M.C.; Du Toit, G.; Vandenplas, Y.; Dupont, C.; Knibb, R.; Uysal, P.; Cavkaytar, O.; et al. Diagnosis and management of Non-IgE gastrointestinal allergies in breastfed infants—An EAACI Position Paper. Allergy 2019, 75, 14–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Nowak-Wegrzyn, A.; Assa’ad, A.H.; Bahna, S.L.; Bock, S.H.; Sicherer, S.A.; Teuber, S.S.; on behalf of the Adverse Reactions to Food Committee of American Academy of Allergy, and Immunology. Work Group report: Oral food challenge testing. J. Allergy Clin. Immunol. 2009, 123, S365–S383. [Google Scholar] [CrossRef] [PubMed]
  108. Culy, C.R.; Bhana, N.; Plosker, G.L. Ondansetron: A review of its use as an antiemetic in children. Pediatr. Drugs 2001, 3, 441–479. [Google Scholar] [CrossRef]
  109. Holbrook, T.; Keet, C.A.; Frischmeyer-Guerrerio, P.A.; Wood, R.A. Use of ondansetron for food protein–induced enterocolitis syndrome. J. Allergy Clin. Immunol. 2013, 132, 1219–1220. [Google Scholar] [CrossRef]
  110. Sopo, S.M.; Battista, A.; Greco, M.; Monaco, S. Ondansetron for Food Protein-Induced Enterocolitis Syndrome. Int. Arch. Allergy Immunol. 2014, 164, 137–139. [Google Scholar] [CrossRef]
  111. Feuille, E.; Nowak-Wegrzyn, A. Food Protein-Induced Enterocolitis Syndrome, Allergic Proctocolitis, and Enteropathy. Curr. Allergy Asthma Rep. 2015, 15, 50. [Google Scholar] [CrossRef] [PubMed]
  112. Sopo, S.M.; Monaco, S.; Bersani, G.; Romano, A.; Fantacci, C. Proposal for management of the infant with suspected food protein-induced allergic proctocolitis. Pediatr. Allergy Immunol. 2018, 29, 215–218. [Google Scholar] [CrossRef]
  113. Murray, K.F.; Christie, D.L. Dietary protein intolerance in infants with transient methemoglobinemia and diarrhea. J. Pediatr. 1993, 122, 90–92. [Google Scholar] [CrossRef]
  114. Baudon, J.J.; Fontaine, J.L.; Mougenot, J.F.; Navarro, J.; Polonovski, C.; Laplane, R. Digestive intolerance to cow’s milk proteins in infants. Biological and histological study. Arch. Fr. Pediatr. 1975, 32, 787–801. [Google Scholar] [PubMed]
  115. Lake, A.M.; Whitington, P.F.; Hamilton, S.R. Dietary protein-induced colitis in breast-fed infants. J. Pediatr. 1982, 101, 906–910. [Google Scholar] [CrossRef]
  116. Molnár, K.; Pintér, P.; Győrffy, H.; Cseh, A.; Muller, K.E.; Arató, A.; Veres, G. Characteristics of allergic colitis in breast-fed infants in the absence of cow’s milk allergy. World J. Gastroenterol. 2013, 19, 3824–3830. [Google Scholar] [CrossRef]
  117. Kimura, M.; Shimomura, M.; Morishita, H.; Meguro, T.; Seto, S. Eosinophilia in infants with food protein-induced enterocolitis syndrome in Japan. Allergol. Int. 2017, 66, 310–316. [Google Scholar] [CrossRef]
  118. Giavi, S.; Megremis, S.; Papadopoulos, N.G. Lymphocyte stimulation test for the diagnosis of non-IgE-mediated cow’s milk allergy: A step closer to a noninvasive diagnostic tool? Int. Arch. Allergy Immunol. 2011, 157, 1–2. [Google Scholar] [CrossRef]
  119. I Fogg, M.; Pawlowski, N.A.; Spergel, J.M.; Brown-Whitehorn, T.A. Atopy patch test for the diagnosis of food protein-induced enterocolitis syndrome. Pediatr. Allergy Immunol. 2006, 17, 351–355. [Google Scholar] [CrossRef]
  120. Järvinen, K.M.; Caubet, J.-C.; Sickles, L.; Ford, L.S.; Sampson, H.A.; Nowak-Węgrzyn, A. Poor utility of atopy patch test in predicting tolerance development in food protein-induced enterocolitis syndrome. Ann. Allergy, Asthma Immunol. 2012, 109, 221–222. [Google Scholar] [CrossRef] [Green Version]
  121. Concha, S.; Cabalin, C.; Iturriaga, C.; Perez-Mateluna, G.; Gomez, C.; Cifuentes, L.; Harris, P.R.; Gana, J.C.; Borzutzky, A. Diagnostic validity of fecal occult blood test in infants with food protein-induced allergic proctocolitis. Rev. Chil. Pediatr. 2018, 89, 630–637. [Google Scholar] [PubMed] [Green Version]
  122. Khan, S. Testing for Fecal Calprotectin in Food Protein–Induced Enterocolitis Syndrome. J. Investig. Allergol. Clin. Immunol. 2018, 28, 287–288. [Google Scholar] [CrossRef] [PubMed]
  123. Beser, O.F.; Sancak, S.; Erkan, T.; Kutlu, T.; Cokugras, H.; Cokugras, F.C. Can Fecal Calprotectin Level Be Used as a Markers of Inflammation in the Diagnosis and Follow-Up of Cow’s Milk Protein Allergy? Allergy Asthma Immunol. Res. 2014, 6, 33–38. [Google Scholar]
  124. Baldassarre, M.E.; Laforgia, N.; Fanelli, M.; Laneve, A.; Grosso, R.; Lifschitz, C. Lactobacillus GG Improves Recovery in Infants with Blood in the Stools and Presumptive Allergic Colitis Compared with Extensively Hydrolyzed Formula Alone. J. Pediatr. 2010, 156, 397–401. [Google Scholar] [CrossRef] [PubMed]
  125. Trillo Belizon, C.; Ortega Paez, E.; Medina Claros, A.F.; Rodriguez Sanchez, I.; Reina Gonzalez, A.; Vera Medialdea, R.; Ramon Salguero, J.M. Faecal calprotectin as an aid to the diagnosis of non-IgE mediated cow’s milk protein allergy. An. Pediatr. (Barc) 2016, 84, 318–323. [Google Scholar] [CrossRef] [PubMed]
  126. Li, F.; Ma, J.; Geng, S.; Wang, J.; Liu, J.; Zhang, J.; Sheng, X. Fecal Calprotectin Concentrations in Healthy Children Aged 1-18 Months. PLoS ONE 2015, 10, e0119574. [Google Scholar] [CrossRef] [Green Version]
  127. Zhu, Q.; Li, F.; Wang, J.; Shen, L.; Sheng, X. Fecal Calprotectin in Healthy Children Aged 1-4 Years. PLoS ONE 2016, 11, e0150725. [Google Scholar] [CrossRef] [Green Version]
  128. Richards, D.G.; Somers, S.; Issenman, R.M.; Stevenson, G.W. Cow’s milk protein/soy protein allergy: Gastrointestinal imaging. Radiology 1988, 167, 721–723. [Google Scholar] [CrossRef]
  129. Powell, G.K. Enterocolitis in low-birth-weight infants associated with milk and soy protein intolerance. J. Pediatr. 1976, 88, 840–844. [Google Scholar] [CrossRef]
  130. Jayasooriya, S.; Fox, A.T.; Murch, S.H. Do Not Laparotomize Food Protein-Induced Enterocolitis Syndrome. Pediatr. Emerg. Care 2007, 23, 173–175. [Google Scholar] [CrossRef]
  131. Jimbo, K.; Ohtsuka, Y.; Kono, T.; Arai, N.; Kyoudo, R.; Hosoi, K.; Aoyagi, Y.; Kudo, T.; Asai, N.; Shimizu, T. Ultrasonographic study of intestinal Doppler blood flow in infantile non-IgE-mediated gastrointestinal food allergy. Allergol. Int. 2019, 68, 199–206. [Google Scholar] [CrossRef]
  132. Variend, S.; Placzek, M.; Raafat, F.; A Walker-Smith, J. Small intestinal mucosal fat in childhood enteropathies. J. Clin. Pathol. 1984, 37, 373–377. [Google Scholar] [CrossRef] [Green Version]
  133. McCalla, R.; Savilahtl, E.; Perkkiö, M.; Kuitunen, P.; Backman, A. Morphology of the Jejunum in Children with Eczema due to Food Allergy. Allergy 1980, 35, 563–571. [Google Scholar] [CrossRef] [PubMed]
  134. Sorea, S.; Dabadie, A.; Bridoux-Henno, L.; Balancon-Morival, M.; Jouan, H.; Le Gall, E. Hemorrhagic colitis in exclusively breast-fed infants. Arch. Pediatr. 2003, 10, 772–775. [Google Scholar] [CrossRef]
  135. Kimura, M.; Ito, Y.; Tokunaga, F.; Meguro, T.; Shimomura, M.; Morishita, H.; Seto, S. Increased C-reactive protein and fever in Japanese infants with food protein-induced enterocolitis syndrome. Pediatr. Int. 2016, 58, 826–830. [Google Scholar] [CrossRef]
  136. Kimura, M.; Shimomura, M.; Morishita, H.; Meguro, T.; Seto, S. Serum C-reactive protein in food protein-induced enterocolitis syndrome versus food protein-induced proctocolitis in Japan. Pediatr. Int. 2016, 58, 836–841. [Google Scholar] [CrossRef] [PubMed]
  137. Pecora, V.; Prencipe, G.; Valluzzi, R.L.; Dahdah, L.; Insalaco, A.; Cianferoni, A.; De Benedetti, F.; Fiocchi, A. Inflammatory events during food protein-induced enterocolitis syndrome reactions. Pediatr. Allergy Immunol. 2017, 28, 464–470. [Google Scholar] [CrossRef] [PubMed]
  138. Yagi, H.; Takizawa, T.; Sato, K.; Inoue, T.; Nishida, Y.; Ishige, T.; Tatsuki, M.; Hatori, R.; Kobayashi, Y.; Yamada, Y.; et al. Severity scales of non-IgE-mediated gastrointestinal food allergies in neonates and infants. Allergol. Int. 2018, 68, 178–184. [Google Scholar] [CrossRef] [PubMed]
  139. Yang, M.; Geng, L.; Xu, Z.; Chen, P.; Friesen, C.A.; Gong, S.; Li, D.-Y. Severe Food Protein-Induced Enterocolitis Syndrome to Cow’s Milk in Infants. Nutrients 2016, 8, 1. [Google Scholar] [CrossRef] [Green Version]
  140. Maines, E.; Di Palma, A.; Burlina, A. Food triggers and inherited metabolic disorders: A challenge to the pediatrician. Ital. J. Pediatr. 2018, 44, 18. [Google Scholar] [CrossRef] [Green Version]
  141. Lee, J.H.; Choe, Y.H.; Lee, S.K.; Seo, J.M.; Kim, J.H.; Suh, Y.L. Allergic proctitis and abdominal distention mimicking Hirschsprung’s disease in infants. Acta Paediatr. 2007, 96, 1784–1789. [Google Scholar] [CrossRef]
  142. Onesimo, R.; Iacono, I.D.; Giorgio, V.; Limongelli, M.G.; Sopo, S.M. Can food protein induced enterocolitis syndrome shift to immediate gastrointestinal hypersensitivity? A report of two cases. Eur. Ann. Allergy Clin. Immunol. 2011, 43, 61–63. [Google Scholar] [PubMed]
  143. Kessel, A.; Dalal, I. The pendulum between food protein-induced enterocolitis syndrome and IgE-mediated milk allergy. Acta Paediatr. 2011, 100, e183–e185. [Google Scholar] [CrossRef] [PubMed]
  144. Banzato, C.; Piacentini, G.L.; Comberiati, P.; Mazzei, F.; Boner, A.L.; Peroni, D.G. Unusual shift from IgE-mediated milk allergy to food protein-induced enterocolitis syndrome. Eur. Ann. Allergy Clin. Immunol. 2013, 45, 209–211. [Google Scholar] [PubMed]
  145. Barni, S.; Mori, F.; Bianchi, A.; Pucci, N.; Novembre, E. Shift from IgE-mediated cow’s milk allergy to food protein-induced enterocolitis syndrome in 2 infants. Pediatr. Allergy Immunol. 2018, 29, 446–447. [Google Scholar] [CrossRef]
  146. Duffey, H.; Egan, M. Development of food protein–induced enterocolitis syndrome (FPIES) to egg after immunoglobulin E–mediated egg allergy. Ann. Allergy, Asthma Immunol. 2018, 121, 379–380. [Google Scholar] [CrossRef]
  147. 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 e1–82 e1. [Google Scholar] [CrossRef]
  148. Flinterman, A.E.; Knulst, A.C.; Meijer, Y.; Pasmans, S.G.M.A.; Bruijnzeel-Koomen, C.A.F.M. Acute allergic reactions in children with AEDS after prolonged cow’s milk elimination diets. Allergy 2006, 61, 370–374. [Google Scholar] [CrossRef]
  149. Du Toit, G.; Roberts, G.; Sayre, P.H.; Bahnson, H.T.; Radulovic, S.; Santos, A.F.; Brough, H.A.; Phippard, D.; Basting, M.; Feeney, M.; et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. New Engl. J. Med. 2015, 372, 803–813. [Google Scholar] [CrossRef] [Green Version]
  150. Walker-Smith, J. Food sensitive enteropathy: Overview and update. Pediatr. Int. 1994, 36, 545–549. [Google Scholar] [CrossRef]
  151. Kokkonen, J.; Haapalahti, M.; Laurila, K.; Karttunen, T.J.; Maki, M. Cow’s milk protein-sensitive enteropathy at school age. J. Pediatr. 2001, 139, 797–803. [Google Scholar] [CrossRef] [PubMed]
  152. Kokkonen, J.; Tikkanen, S.; Savilahti, E. Residual Intestinal Disease After Milk Allergy in Infancy. J. Pediatr. Gastroenterol. Nutr. 2001, 32, 156–161. [Google Scholar] [CrossRef] [PubMed]
  153. Walker, W.A. Cow’s milk protein-sensitive enteropathy at school age: A new entity or a spectrum of mucosal immune responses with age. J. Pediatr. 2001, 139, 765–766. [Google Scholar] [CrossRef] [PubMed]
  154. Di Nardo, G.; Cremon, C.; Frediani, S.; Lucarelli, S.; Villa, M.P.; Stanghellini, V.; La Torre, G.; Martemucci, L.; Barbara, G. Allergic Proctocolitis Is a Risk Factor for Functional Gastrointestinal Disorders in Children. J. Pediatr. 2018, 195, 128–133.e1. [Google Scholar] [CrossRef]
  155. Meyer, R.; Schwarz, C.; Shah, N. A review on the diagnosis and management of food-induced gastrointestinal allergies. Curr. Allergy Clin. Immunol. 2012, 25, 10–17. [Google Scholar]
  156. De Boissieu, D.; Matarazzo, P.; Dupont, C. Allergy to extensively hydrolyzed cow milk proteins in infants: Identification and treatment with an amino acid-based formula. J. Pediatr. 1997, 131, 744–747. [Google Scholar] [CrossRef]
  157. A Vanderhoof, J.; Murray, N.D.; Kaufman, S.S.; Mack, D.R.; Antonson, D.L.; Corkins, M.R.; Perry, D.; Kruger, R. Intolerance to protein hydrolysate infant formulas: An underrecognized cause of gastrointestinal symptoms in infants. J. Pediatr. 1997, 131, 741–744. [Google Scholar] [CrossRef]
  158. American Academy of Pediatrics. Committee on Nutrition. Hypoallergenic infant formulas. Pediatrics 2000, 106, 346–349. [Google Scholar] [CrossRef] [Green Version]
  159. Vandenplas, Y. Prevention and Management of Cow’s Milk Allergy in Non-Exclusively Breastfed Infants. Nutrients 2017, 9, 731. [Google Scholar] [CrossRef]
  160. Vandenplas, Y.; Koletzko, S.; Isolauri, E.; Hill, D.; Oranje, A.P.; Brueton, M.; Staiano, A.; Dupont, C. Guidelines for the diagnosis and management of cow’s milk protein allergy in infants. Arch. Dis. Child. 2007, 92, 902–908. [Google Scholar] [CrossRef] [Green Version]
  161. Sopo, S.M.; Buonsenso, D.; Monaco, S.; Crocco, S.; Longo, G.; Calvani, M. Food protein-induced enterocolitis syndrome (FPIES) and well cooked foods: A working hypothesis. Allergol. et Immunopathol. 2013, 41, 346–348. [Google Scholar] [CrossRef] [PubMed]
  162. Sopo, S.M.; Romano, A.; Bersani, G.; Fantacci, C.; Badina, L.; Longo, G.; Monti, G.; Viola, S.; Tripodi, S.; Barilaro, G.; et al. Cooking influence in tolerance acquisition in egg-induced acute food protein enterocolitis syndrome. Allergol. et Immunopathol. 2019, 47, 221–226. [Google Scholar] [CrossRef]
  163. Uncuoglu, A.; Yologlu, N.; Simsek, I.; Uyan, Z.; 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. et Immunopathol. 2017, 45, 560–566. [Google Scholar] [CrossRef] [PubMed]
  164. Upton, J.E.M.; Nowak-Węgrzyn, A. The Impact of Baked Egg and Baked Milk Diets on IgE- and Non-IgE-Mediated Allergy. Clin. Rev. Allergy Immunol. 2018, 55, 118–138. [Google Scholar] [CrossRef] [PubMed]
  165. Camargo, L.S.; Da Silveira, J.A.C.; Taddei, J.A.; Neto, U.F. ALLERGIC PROCTOCOLITIS IN INFANTS: Analysis of the evolution of the nutritional status. Arq. de Gastroenterol. 2016, 53, 262–266. [Google Scholar] [CrossRef] [Green Version]
  166. Venter, C.; Groetch, M. Nutritional management of food protein-induced enterocolitis syndrome. Curr. Opin. Allergy Clin. Immunol. 2014, 14, 255–262. [Google Scholar] [CrossRef] [Green Version]
  167. Meyer, R.W.; De Koker, C.; Dziubak, R.; Venter, C.; Dominguez-Ortega, G.; Cutts, R.; Yerlett, N.; Skrapak, A.-K.; Fox, A.T.; Shah, N. Malnutrition in children with food allergies in the UK. J. Hum. Nutr. Diet. 2013, 27, 227–235. [Google Scholar] [CrossRef]
  168. Meyer, R.W.; Rommel, N.; Van Oudenhove, L.; Fleming, C.; Dziubak, R.; Shah, N. Feeding difficulties in children with food protein-induced gastrointestinal allergies. J. Gastroenterol. Hepatol. 2014, 29, 1764–1769. [Google Scholar] [CrossRef]
  169. Meyer, R.W.; Wright, K.; Vieira, M.C.; Chong, K.W.; Chatchatee, P.; Vlieg-Boerstra, B.J.; Groetch, M.; Dominguez-Ortega, G.; Heath, S.; Lang, A.; et al. International survey on growth indices and impacting factors in children with food allergies. J. Hum. Nutr. Diet. 2018, 32, 175–184. [Google Scholar] [CrossRef]
  170. Meyer, R.W.; De Koker, C.; Dziubak, R.; Skrapac, A.-K.; Godwin, H.; Reeve, K.; Lozinsky, A.C.; Shah, N. A practical approach to vitamin and mineral supplementation in food allergic children. Clin. Transl. Allergy 2015, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  171. Meyer, R.W. Nutritional disorders resulting from food allergy in children. Pediatr. Allergy Immunol. 2018, 29, 689–704. [Google Scholar] [CrossRef] [PubMed]
  172. Foong, R.-X.M.; Meyer, R.W.; Dziubak, R.; Lozinsky, A.C.; Godwin, H.; Reeve, K.; Hussain, S.T.; Nourzaie, R.; Shah, N. Establishing the prevalence of low vitamin D in non-immunoglobulin-E mediated gastrointestinal food allergic children in a tertiary centre. World Allergy Organ. J. 2017, 10, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Mailhot, G.; Perrone, V.; Alos, N.; Dubois, J.; Delvin, E.; Paradis, L.; Roches, A.D. Cow’s Milk Allergy and Bone Mineral Density in Prepubertal Children. Pediatrics 2016, 137. [Google Scholar] [CrossRef] [Green Version]
  174. Lazare, F.B.; Brand, D.A.; Marciano, T.A.; Daum, F. Rapid Resolution of Milk Protein Intolerance in Infancy. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 215–217. [Google Scholar] [CrossRef] [PubMed]
  175. Luyt, D.; Ball, H.; Makwana, N.; Green, M.R.; Bravin, K.; Nasser, S.M.; Clark, A. BSACI guideline for the diagnosis and management of cow’s milk allergy. Clin. Exp. Allergy 2014, 44, 642–672. [Google Scholar] [CrossRef]
  176. Baker, R.D.; Greer, F.R. Diagnosis and Prevention of Iron Deficiency and Iron-Deficiency Anemia in Infants and Young Children (0-3 Years of Age). Pediatrics 2010, 126, 1040–1050. [Google Scholar] [CrossRef] [Green Version]
  177. Foong, R.X.; Meyer, R.; Godwin, H.; Dziubak, R.; Lozinsky, A.C.; Reeve, K.; Knibb, R.; Shah, N. Parental perception of their child’s quality of life in children with non-immunoglobulin-E-mediated gastrointestinal allergies. Pediatr. Allergy Immunol. 2017, 28, 251–256. [Google Scholar] [CrossRef] [Green Version]
  178. Meyer, R.W.; Godwin, H.; Dziubak, R.; Panepinto, J.A.; Foong, R.-X.M.; Bryon, M.; Lozinsky, A.C.; Reeve, K.; Shah, N. The impact on quality of life on families of children on an elimination diet for Non-immunoglobulin E mediated gastrointestinal food allergies. World Allergy Organ. J. 2017, 10, 8. [Google Scholar] [CrossRef] [Green Version]
  179. Nowak-Wegrzyn, A.; Katz, Y.; Mehr, S.; Koletzko, S. Non–IgE-mediated gastrointestinal food allergy. J. Allergy Clin. Immunol. 2015, 135, 1114–1124. [Google Scholar] [CrossRef]
  180. Iacono, G.; Carroccio, A.; Cavataio, F.; Montalto, G.; Kazmierska, I.; Lorello, D.; Soresi, M.; Notarbartolo, A. Gastroesophageal reflux and cow’s milk allergy in infants: A prospective study. J. Allergy Clin. Immunol. 1996, 97, 822–827. [Google Scholar]
  181. Nielsen, R.G.; Bindslev-Jensen, C.; Kruse-Andersen, S.; Husby, S. Severe Gastroesophageal Reflux Disease and Cow Milk Hypersensitivity in Infants and Children: Disease Association and Evaluation of a New Challenge Procedure. J. Pediatr. Gastroenterol. Nutr. 2004, 39, 383–391. [Google Scholar] [CrossRef] [PubMed]
  182. Ravelli, A.M.; Tobanelli, P.; Volpi, S.; Ugazio, A.G. Vomiting and gastric motility in infants with cow’s milk allergy. J. Pediatr. Gastroenterol. Nutr. 2001, 32, 59–64. [Google Scholar] [CrossRef] [PubMed]
  183. Borrelli, O.; Mancini, V.; Thapar, N.; Giorgio, V.; Elawad, M.; Hill, S.; Shah, N.; Lindley, K.J. Cow’s Milk Challenge Increases Weakly Acidic Reflux in Children with Cow’s Milk Allergy and Gastroesophageal Reflux Disease. J. Pediatr. 2012, 161, 476–481.e1. [Google Scholar] [CrossRef]
  184. Vandenplas, Y. Management of paediatric GERD. Nat. Rev. Gastroenterol. Hepatol. 2013, 11, 147–157. [Google Scholar] [CrossRef] [PubMed]
  185. Loo, E.X.L.; Wang, D.Y.; Siah, K.T.H. Association between Irritable Bowel Syndrome and Allergic Diseases: To Make a Case for Aeroallergen. Int. Arch. Allergy Immunol. 2019, 181, 31–42. [Google Scholar] [CrossRef]
  186. Choung, R.S.; A Murray, J. The Role for Food Allergies in the Pathogenesis of Irritable Bowel Syndrome: Understanding Mechanisms of Intestinal Mucosal Responses Against Food Antigens. Gastroenterology 2019, 157, 15–17. [Google Scholar] [CrossRef] [Green Version]
  187. Stefanini, G.F.; Saggioro, A.; Alvisi, V.; Angelini, G.; Capurso, L.; Di Lorenzo, G.; Dobrilla, G.; Dodero, M.; Galimberti, M.; Gasbarrini, G.; et al. Oral Cromolyn Sodium in Comparison with Elimination Diet in the Irritable Bowel Syndrome, Diarrheic Type Multicenter Study of 428 Patients. Scand. J. Gastroenterol. 1995, 30, 535–541. [Google Scholar] [CrossRef]
  188. Fritscher-Ravens, A.; Pflaum, T.; Mösinger, M.; Ruchay, Z.; Röcken, C.; Milla, P.J.; Das, M.; Böttner, M.; Wedel, T.; Schuppan, D.; et al. Many Patients With Irritable Bowel Syndrome Have Atypical Food Allergies Not Associated With Immunoglobulin E. Gastroenterology 2019, 157, 109.e5–118.e5. [Google Scholar] [CrossRef] [Green Version]
  189. Quinlan, P.T.; Lockton, S.; Irwin, J.; Lucas, A.L. The Relationship between Stool Hardness and Stool Composition in Breast- and Formula-Fed Infants. J. Pediatr. Gastroenterol. Nutr. 1995, 20, 81–90. [Google Scholar] [CrossRef]
  190. Iacono, G.; Bonventre, S.; Scalici, C.; Maresi, E.; Di Prima, L.; Soresi, M.; Di Ges??, G.; Noto, D.; Carroccio, A. Food intolerance and chronic constipation: Manometry and histology study. Eur. J. Gastroenterol. Hepatol. 2006, 18, 143–150. [Google Scholar] [CrossRef] [Green Version]
  191. Nocerino, R.; Pezzella, V.; Cosenza, L.; Amoroso, A.; Di Scala, C.; Amato, F.; Iacono, G.; Canani, R.B. The Controversial Role of Food Allergy in Infantile Colic: Evidence and Clinical Management. Nutrients 2015, 7, 2015–2025. [Google Scholar] [CrossRef] [Green Version]
  192. Gordon, M.; Biagioli, E.; Sorrenti, M.; Lingua, C.; Moja, L.; Banks, S.S.; Ceratto, S.; Savino, F. Dietary modifications for infantile colic. Cochrane Database Syst. Rev. 2018, 10, CD011029. [Google Scholar] [CrossRef] [PubMed]
  193. Natsume, O.; Kabashima, S.; Nakazato, J.; Yamamoto-Hanada, K.; Narita, M.; Kondo, M.; Saito-Abe, M.; Kishino, A.; Takimoto, T.; Inoue, E.; et al. Two-step egg introduction for prevention of egg allergy in high-risk infants with eczema (PETIT): A randomised, double-blind, placebo-controlled trial. Lancet 2017, 389, 276–286. [Google Scholar] [CrossRef] [Green Version]
  194. Qamer, S.; Deshmukh, M.; Patole, S. Probiotics for cow’s milk protein allergy: A systematic review of randomized controlled trials. Eur. J. Nucl. Med. Mol. Imaging 2019, 178, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  195. Canani, R.B.; Nocerino, R.; Terrin, G.; Coruzzo, A.; Cosenza, L.; Leone, L.; Troncone, R. Effect of Lactobacillus GG on tolerance acquisition in infants with cow’s milk allergy: A randomized trial. J. Allergy Clin. Immunol. 2012, 129, 580–582.e5. [Google Scholar] [CrossRef] [PubMed]
  196. Leonard, S.A.; Jean-Christoph, 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. Pr. 2015, 3, 13–23. [Google Scholar] [CrossRef]
Figure 1. Classification of gastrointestinal food allergies. FDEIAn, food-dependent exercise-induced anaphylaxis; FPE, food protein-induced enteropathy; FPIAP, food protein-induced allergic proctocolitis; FPIES, protein-induced enterocolitis syndrome; IgE, immunoglobulin E; OAS, oral allergy syndrome.
Figure 1. Classification of gastrointestinal food allergies. FDEIAn, food-dependent exercise-induced anaphylaxis; FPE, food protein-induced enteropathy; FPIAP, food protein-induced allergic proctocolitis; FPIES, protein-induced enterocolitis syndrome; IgE, immunoglobulin E; OAS, oral allergy syndrome.
Nutrients 12 02086 g001
Figure 2. Gastrointestinal organs affected in the different non-IgE-mediated gastrointestinal food allergies. FPIAP and FPE affect the colon and small intestine, respectively, while in FPIES, the whole gastrointestinal tract can be affected. FPE, food protein-induced enteropathy; FPIAP, food protein-induced allergic proctocolitis; FPIES, protein-induced enterocolitis syndrome.
Figure 2. Gastrointestinal organs affected in the different non-IgE-mediated gastrointestinal food allergies. FPIAP and FPE affect the colon and small intestine, respectively, while in FPIES, the whole gastrointestinal tract can be affected. FPE, food protein-induced enteropathy; FPIAP, food protein-induced allergic proctocolitis; FPIES, protein-induced enterocolitis syndrome.
Nutrients 12 02086 g002
Figure 3. Classification scheme of FPIES. FPIES, food protein-induced enterocolitis syndrome; sIgE, food-specific immunoglobulin E; T21, trisomy 21 (Down syndrome).
Figure 3. Classification scheme of FPIES. FPIES, food protein-induced enterocolitis syndrome; sIgE, food-specific immunoglobulin E; T21, trisomy 21 (Down syndrome).
Nutrients 12 02086 g003
Figure 4. Multidisciplinary approach for diagnosis and management of non-IgE-mediated gastrointestinal food allergies. ER, emergency room; OFC, oral food challenge; PCP, primary care physician.
Figure 4. Multidisciplinary approach for diagnosis and management of non-IgE-mediated gastrointestinal food allergies. ER, emergency room; OFC, oral food challenge; PCP, primary care physician.
Nutrients 12 02086 g004
Table 1. Clinical and laboratory features of non-IgE-mediated gastrointestinal food allergies.
Table 1. Clinical and laboratory features of non-IgE-mediated gastrointestinal food allergies.
.FPIESFPEFPIAP
Age of presentationCow’s milk/soy: First weeks-months of life
Solids: 4–7 months
Can also occur in adults
2–24 months
Can also occur in older children
First weeks-months of life (<6 months)
Can also occur in older children
Top culprit foodsCow’s milk, soy (C > A)
Rice, poultry, fish, fruits, vegetables (A > C)
Cow’s milk, soy
Wheat, egg
Cow’s milk, soy
Egg, corn, wheat
Multiple foodsFrequent
≥3 foods: 5–10%
RareOccasional
Feeding at onsetFormulaFormulaExclusively BF (>50%)
Clinical presentation(A): repeated vomiting, diarrhea, dehydration (shock: 15%), lethargy, pallor, hypothermia
(C): intermittent vomiting, diarrhea, FTT
Diarrhea, intermittent vomiting, FTT, malabsorption (steatorrhea), bloody stools (rare)Blood/mucus streaked stools, mild diarrhea
Otherwise well-appearing
Co-morbid atopy40–60%
Familial: 40–80%
20–40%25–50%
Familial: 30–60%
Laboratory anomaliesAnemia (C)
Eosinophilia (C)
Neutrophilia (A, C)
Thrombocytosis (A)
Methemoglobinemia (A, C)
Metabolic acidosis (A, C)
Anemia
Hypoalbuminemia
Iron deficiency
Mild anemia
Hypoalbuminemia (rare)
Eosinophilia
Stool studiesOccult blood (A, C)
PMN (A, C)
Eosinophils (A, C)
Reducing substances (C)
Fecal fat
Low d-Xylose excretion
Gross/occult blood
Eosinophils
Endoscopy/HistologyFriable mucosa
Ulceration
Villous atrophy
Crypt abscesses
Inflammatory cell infiltrates
Villous atrophy
Crypt hyperplasia
Lymphocytic infiltrate
Mild, focal colitis
Eosinophilic infiltration
Lymphonodular hyperplasia
Allergy evaluationNegative; sIgE+ in 25%Negative (not recommended)Negative (not recommended)
DiagnosisClinical +/− OFCClinical & histologicalClinical +/− OFC
TreatmentAvoidance of offending foodsAvoidance of offending foodsAvoidance of offending foods (maternal exclusion diet if BF)
Time to improvement(A) 4–12 h (<24 h)
(C) 3–10 days
Several weeks
(1–2 weeks)
72 h (up to 2 weeks)
Natural historyResolution < 3–5 y
Later if sIgE+ or solid foods
Resolution < 1–2 yearsResolution < 1–2 years
A: acute; APT, atopy patch test; BF, breastfed; C: chronic; FTT, failure to thrive, OFC, oral food challenge; PMN, polymorphonuclear leukocyte; sIgE, specific immunoglobulin E.
Table 2. Foods commonly implicated in non-IgE-mediated gastrointestinal food allergies in select large case-series studies.
Table 2. Foods commonly implicated in non-IgE-mediated gastrointestinal food allergies in select large case-series studies.
FPIESFPEFPIAP
CountryUSA 1UK 2Spain 3Italy 4Australia 5Turkey 6Finland 7USA 8Turkey 9
N (total)N = 1340N = 54N = 336N = 66N = 265N = 27N = 54N = 95N = 359
%%%%%%%%%
Cow’s milk19–674626–386720–33741006591–100
Soy8–41110–145–34-113 *0–3 *
Rice19–5341–10440–454---
Oat16–3760–1-6–9----
Wheat1–16110–120–3437-0–4
Corn2–820–320–1--6-
Eggs0–231310–2160–12-4187–22
Fish/Shellfish1–151534–54123–515--0–2
Poultry5–1071–433–8---0–3
Meat3–1841-3–4-2-0–10
Sweet potato4–22---3–6----
Potato2–820–1-0–24--0–2
Squash0–12--------
Carrot0–740-0–1---0–1
Banana4–2460–133–444--
Avocado0–16---0–2----
Apple0–1120–1-0–2---0–1
Pear0–9-0–1-0–3----
* Soy allergy likely underrepresented by these studies. 1 From Ruffner et al. [95] (n = 462), Caubet et al. [26] (n = 160), Blackman et al. [65] (n = 74), Maciag et al. [25] (n = 441), Su et al. [64] (n = 203); 2 From Ludman et al. [66] (n = 54); 3 From Vazquez-Ortiz [101] (n = 81), Diaz et al. [60] (n = 120), Pérez Ajami et al. [104] (n = 135); 4 From Miceli Sopo et al. [67] (n = 66); 5 From Mehr et al. [97] (n = 35), Mehr et al. [21] (n = 230); 6 From Arik Yilmaz et al. [105] (n = 27); 7 From Kuitunen et al. [28] (n = 54); 8 From Lake et al. [84] (n = 95); 9 From Kaya et al. [87] (n = 60), Arik Yilmaz et al. [105] (n = 37), Erdem et al. [86] (n = 77), Cetinkaya et al. [85] (n = 185).
Table 3. Diagnostic criteria of non-IgE-mediated gastrointestinal food allergies.
Table 3. Diagnostic criteria of non-IgE-mediated gastrointestinal food allergies.
Acute FPIES 1
Major Criteria, PLUSMinor Criteria (≥3 Occurring with Episode)
1. Vomiting 1–4 h after suspect food ingestion
AND
2. Absence IgE-mediated allergic symptoms
1. ≥2 episodes with same food
2. 1 episode with a different food
3. Lethargy
4. Pallor
5. Need for ER visit
6. Need for IV fluid support
7. Diarrhea within 24 h (usually 5–10 h)
8. Hypotension
9. Hypothermia
Chronic FPIES 2
Symptoms and severityCriteria
Milder (lower doses with intermittent ingestion):
1. Intermittent vomiting and/or diarrhea
2. FTT
3. No dehydration or metabolic acidosis
 
Severe (higher doses with chronic ingestion):
1. Intermittent but progressive vomiting and diarrhea (occasionally with blood)
2. Possible dehydration and metabolic acidosis
1. Resolution of symptoms within days after elimination of offending food(s)
2. Acute recurrence of symptoms (vomiting in 1–4 h, diarrhea in <24 h, usually 5–10 h) when the food is reintroduced
3. Confirmatory OFC, or presumptive diagnosis if OFC not performed
FPE 3
1. Generally <9 months of age at diagnosis, but can also present in older children
2. Repeated exposure to causative foods elicits gastrointestinal symptoms (predominantly vomiting and FTT), without alternative cause
3. Histologic confirmation of the diagnosis in a symptomatic child by biopsy of the small bowel showing villous injury, crypt hyperplasia and inflammation
4. Clinical and histological improvement after removal of offending food(s)
5. Exclusion of alternative causes
FPIAP 4
1. Mild rectal bleeding in an otherwise healthy infant
2. Resolution of symptoms after elimination of offending food(s) (if exclusively breastfed, resolution after a maternal elimination diet)
3. Recurrence of symptoms upon reintroduction of culprit food(s) in the diet (preferable)
4. Exclusion of other causes of rectal bleeding
1 Major criterion must be met (both) plus at least three minor criteria (adapted from the International consensus guidelines [68]); 2 General criteria because of paucity of available data (adapted from the International consensus guidelines [68]); 3 There are no defined criteria in the literature for FPE diagnosis. The elements listed here are generally used for diagnosis in clinical practice (adapted from [81]); 4 There are no defined criteria in the literature for FPIAP diagnosis. The elements listed here are generally used for diagnosis in clinical practice (adapted from EEACI position paper [106]); ER, emergency room; FTT, failure to thrive; IgE, immunoglobulin E; IV, intravenous; OFC, oral food challenge.
Table 4. Differential diagnosis of non-IgE-mediated gastrointestinal food allergies.
Table 4. Differential diagnosis of non-IgE-mediated gastrointestinal food allergies.
Acute FPIESChronic FPIESFPEFPIAP
AllergicAnaphylaxis
Eosinophilic gastroenteropathies
FPIAP
FPE
Eosinophilic gastroenteropathies
Celiac disease
Chronic FPIES
Eosinophilic gastroenteropathies
FPIES
FPE
Eosinophilic gastroenteropathies
InfectiousSepsis
Viral/bacterial/
parasitic gastroenteritis
Viral/bacterial/
parasitic gastroenteritis
Viral/bacterial/
parasitic gastroenteritis
Viral/bacterial/
parasitic gastroenteritis
GastrointestinalHirschsprung
Pyloric stenosis
Intussusception
Volvulus
NEC
GERD
Hirschsprung
Pyloric stenosis
VEOIBD
Cystic fibrosis
VEOIBD
Cystic fibrosis
Anal fissure
Swallowed maternal blood
NEC
Intussusception
Volvulus
Meckel diverticulum
Intestinal duplication kyst
Infantile polyp
VEOIBD
MetabolicInborn errors of metabolism
T1DM
Inborn errors of metabolism
T1DM
Inborn errors of metabolism
Congenital disaccharidase deficiency
T1DM
-
HematologicCongenital methemoglobinemiaCongenital methemoglobinemia-Coagulation defect
Thrombocytopenia
Neuro-
logic
Cyclic vomiting
Intracranial mass
Cyclic vomiting
Intracranial mass
--
CardiovascularCongenital heart defect
Cardiomyopathy
Arrythmia
Congenital heart defect
Cardiomyopathy
-Vascular malformation
Endocri-
nologic
Congenital adrenal hypoplasiaCongenital adrenal hypoplasiaCongenital adrenal hypoplasia-
Immunologic-PID
Autoimmune enteropathy
PID
Autoimmune enteropathy
-
PsychologicFood aversionFood aversionFood aversion
Neglect
-
FPE, food protein-induced enteropathy; FPIAP, food protein-induced allergic proctocolitis; FPIES, protein-induced enterocolitis syndrome; GERD, gastroesophageal reflux disease; NEC, necrotizing enterocolitis; PID, primary immunodeficiency; T1DM, type 1 diabetes mellitus; VEOIBD, very early-onset inflammatory bowel disease.
Table 5. Food-specific dietary advice and food co-reactivity in non-IgE-mediated gastrointestinal food allergies.
Table 5. Food-specific dietary advice and food co-reactivity in non-IgE-mediated gastrointestinal food allergies.
FPIESFPEFPIAP
Cow’s milk1st choice: EHF (10–20% reactivity)
AAF if failure of EHF
Soy formula: 40–60% co-reactivity
Rice formula: co-reactivity unknown
May tolerate baked cow’s milk
First choice: EHF (20% reactivity)
AAF if failure of EHF
Soy formula: 10–30% co-reactivity
If BF:
1st choice: maternal elimination diet
Alternative: EHF
If on formula:
1st choice: EHF (10–20% reactivity)
AAF if failure of EHF
Soy formula: 10–30% co-reactivity
SoyCow’s milk: 40% co-reactivity -May be eliminated if no improvement with cow’s milk exclusion alone
RiceOats: 25–40% co-reactivity
Wheat: 0–5% co-reactivity
Corn: 1%: co-reactivity
--
ChickenAvoid all poultry *: up to 40% co-reactivity--
EggMay tolerate baked eggs-May be eliminated if no improvement with cow’s milk/soy exclusion alone
FishAvoid all fish *: up to 80% co-reactivity between white & red fish
Shellfish: 50% co-reactivity
--
Maternal elimination diet in BFNo, unless symptomaticUnknownYes
May vary in different countries; * Unless already tolerated. In select cases, determination of cross-reactivity can be attempted with an OFC; AAF, amino-acid formula; BF: breastfed; EHF, extensively hydrolyzed formula.

Share and Cite

MDPI and ACS Style

Labrosse, R.; Graham, F.; Caubet, J.-C. Non-IgE-Mediated Gastrointestinal Food Allergies in Children: An Update. Nutrients 2020, 12, 2086. https://doi.org/10.3390/nu12072086

AMA Style

Labrosse R, Graham F, Caubet J-C. Non-IgE-Mediated Gastrointestinal Food Allergies in Children: An Update. Nutrients. 2020; 12(7):2086. https://doi.org/10.3390/nu12072086

Chicago/Turabian Style

Labrosse, Roxane, François Graham, and Jean-Christoph Caubet. 2020. "Non-IgE-Mediated Gastrointestinal Food Allergies in Children: An Update" Nutrients 12, no. 7: 2086. https://doi.org/10.3390/nu12072086

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