Preprint
Review

This version is not peer-reviewed.

Engineering Allergenicity: A Review of Non-Thermal Processing Approaches to Reduce Immune Reactivity in Foods

Submitted:

11 August 2025

Posted:

12 August 2025

You are already at the latest version

Abstract
Food allergy rates are escalating globally, driven by the interplay of inherited traits, environmental triggers, diet, and gut microbiome composition. Sensitization typically occurs through dermal, respiratory, or gastrointestinal routes, with epithelial barrier dysfunction and Th2-skewed immune responses playing key roles. Understanding the mechanisms and pathways of sensitization is critical to developing effective mitigation strategies. Efforts are underway to evaluate how both traditional heating and innovative non-thermal techniques can modify allergenic proteins in commonly allergenic foods. Thermal methods can denature allergenic proteins but may also compromise nutritional and sensory quality. Unlike heat-based processes, innovative non-thermal methods—including pressure-based, light-based, plasma, and sound technologies—can restructure proteins and diminish IgE reactivity without compromising food integrity. However, outcomes are highly context-dependent, influenced by food matrix, processing parameters, and assessment methods. Limitations in current in vitro and in vivo models, lack of standardized allergenicity testing protocols, and insufficient clinical validation remain key barriers. This review synthesizes current knowledge of allergy mechanisms with technological approaches for allergen reduction, highlighting promising interventions, existing constraints, and the need for integrated research to develop safe, hypoallergenic foods.
Keywords: 
;  ;  ;  ;  ;  ;  

1. Food Allergy and Sensitization: Mechanisms, Pathways, and Risk Factors

An atypical immune reaction to dietary proteins defines food allergy, which is reported in roughly 6% of pediatric and 3–4% of adult populations. Symptoms most often affect the skin, gut, or respiratory tract. These conditions are typically categorized into those involving IgE antibodies and those that are cell-mediated [1,2].
Typically, the immune system develops tolerance to food-derived antigens through a mechanism called oral tolerance; however, in cases of food allergy, this regulatory process breaks down. Although a wide range of foods can elicit allergic reactions, a limited number of foods account for most clinically significant cases. Common allergenic foods include dairy products, eggs, legumes like peanuts, various nuts, and seafood such as fish and shellfish [2,3,4].
A reliable diagnosis of food allergy necessitates a methodical approach grounded in clinical evidence. This usually begins with an in-depth medical history, supplemented by specific tests such as serum IgE assays, cutaneous testing, dietary interventions, and oral food challenges when warranted. Recent breakthroughs in molecular allergology have facilitated the identification of key allergenic components, expanding both diagnostic accuracy and therapeutic potential. Nevertheless, the current mainstay of management remains strict avoidance of known allergens, comprehensive patient education, and readiness to manage accidental exposures—primarily through the use of emergency epinephrine [2].
Food allergies can develop through various routes of sensitization. Proteins implicated in Class 1 food allergies are acquired through ingestion and often exhibit resistance to heat, low pH, and enzymatic breakdown during digestion [5,6,7]. These allergens are often introduced during infancy or early childhood, a period when the immune system is particularly susceptible to dysregulation. By contrast, class 2 food allergies typically stem from initial immune sensitization to airborne particles like pollen, which share structural features with certain proteins found in uncooked fruits and vegetables. This molecular resemblance can provoke cross-reactive immune responses, often manifesting as symptoms of pollen–food syndrome [5,6].
Emerging evidence indicates that exposure to specific class 1 food allergens, including peanuts and eggs, has the potential to trigger sensitization through transdermal routes, especially among those who have compromised skin barrier integrity [8,9]. A large proportion of class 1 food allergens are glycoproteins that are soluble and stable under various processing conditions have molecular masses within the range of 10–70 kilodaltons, and they typically resist both enzymatic degradation and heat treatment. Representative examples include milk caseins, egg-derived ovomucoid, peanut vicilins, and lipid transport proteins lacking substrate specificity present in fruits like apples (Mal d 3) and grains such as corn (Zea m 14) [5,6]. Bet v 1, the principal allergen in birch pollen, serves as a model for class 2 allergens. It can trigger respiratory sensitization and lead to oral allergic symptoms following ingestion of structurally similar proteins like Mal d 1 from apples or Dau c 1 from carrots. A large proportion of known food allergens are sourced from a relatively small group belonging to protein families, including those in the members of the Cupin and Prolamin superfamilies, along with various pathogenesis-related protein groups [5,6].
The clinical significance of particular sensitizations differs. For example, the peanut proteins Ara h 1, Ara h 2, and Ara h 3 are commonly involved in more severe IgE-mediated allergic reactions, whereas Ara h 8—a protein structurally related to Bet v 1—is typically connected to milder allergic manifestations, such as oral allergy syndrome [10]. Despite structural similarities between proteins, true clinical cross-reactivity is often less common than laboratory tests suggest [1,6].
Food allergies can result in a range of clinical disorders affecting one or more organ systems. Many gastrointestinal disorders attributed to food allergies exhibit overlapping symptoms, but they can often be distinguished through careful clinical evaluation and diagnostic testing [11,12,13]. Additionally, some pediatric gastrointestinal conditions—such as colic, constipation, and reflux—are occasionally suspected to have allergic etiologies (Figure 1).
Anaphylaxis represents a severe, potentially fatal reaction triggered by certain foods, often leading to severe complications and constituting a major reason for emergency interventions in outpatient care [14]. Life-threatening events are most frequently linked as a result of tree nut or peanut contact in youths and young adults who have both diagnosed food allergies and asthma, especially when epinephrine is not administered in a timely manner [12,15].
Adverse food reactions more broadly, termed food intolerances, arise from a variety of mechanisms. Unfavorable food reactions are typically classified into toxic and non-toxic categories. Among the non-toxic types, non–immune-based intolerances, including those resulting from enzyme deficiencies (like lactose intolerance) or from compounds such as biogenic amines, occur more frequently than true immune-driven food allergies. Nonetheless, immune-mediated food allergies, though less prevalent, affect millions globally and pose significant medical and economic burdens. In their most severe form, these allergies can cause life-threatening anaphylaxis [2,16,17]. According to the National Institute of Allergy and Infectious Diseases (NIAID), a food allergy is characterized as “a targeted immune response triggered by food, leading to repeatable adverse health outcomes” [18,19], encompassing both adaptive food allergens are sourced from a relatively small group of protein families, such as and innate immune pathways.
The term “allergy” was originally used to describe hypersensitivity reactions, including serum sickness observed in children treated with antiserum [20,21]. Later, hypersensitivity reactions were classified into four immunological types [22]. The predominant mechanism behind food allergies involves Type I hypersensitivity, where IgE antibodies target specific food proteins. These reactions involve complex immune cascades, including T cell activation and eosinophil recruitment [16]. Diagnosis often includes detection of specific IgE antibodies or IgE-related cellular responses [2].
Although there has been speculation regarding the involvement of food-specific IgG antibodies in Type II or Type III hypersensitivity reactions, current scientific evidence does not support a pathogenic role. Accordingly, professional societies discourage the use of IgG-based food allergy testing [23,24,25]. Type IV hypersensitivity reactions, mediated by T cells, contribute to conditions such as celiac disease, in which gluten proteins trigger autoimmune inflammation in the gut [26]. Food protein-induced enterocolitis syndrome (FPIES) is another condition driven by T cells, primarily impacting infants and occurring independently of IgE-mediated responses [27].
Recent studies also implicate the innate immune system in food-induced inflammation. Molecules such as wheat-derived amylase-trypsin inhibitors alongside specific milk oligosaccharides activate Toll-like receptor 4 (TLR4), initiating intestinal inflammation [4,28]. Such mechanisms may underlie disorders like non-celiac gluten sensitivity [4,29,30].
The occurrence of IgE-mediated food allergies is estimated to affect approximately 3% to 8% of children and 1% to 3% of adults in developed countries [2,17,31]. These conditions impose more than merely physical health threats but also emotional and social burdens due to dietary limitations and lifestyle adjustments. Major food allergenic substances encompass dairy, wheat, eggs, tree nuts, peanuts, sesame seeds, fish, as well as various fruits and vegetables [24,31]. While sensitivities to milk, eggs, and wheat frequently diminish by adolescence, allergies to tree nuts, peanuts, and seafood are more likely to continue into adulthood [2].
Estimating the real occurrence of food allergies remains challenging due to variability in diagnostic methodologies and reliance on self-reported versus clinically confirmed cases [32]. However, research indicates a rising trend in both the frequency and intensity of food allergies, influenced by multifaceted interactions among genetic predispositions and environmental elements such as dietary habits and lifestyle choices [9,33,34,35].
Studies in epigenetics have identified unique DNA methylation signatures in CD4+ T cells of individuals, particularly children, suffering from IgE-driven food allergies, highlighting the influence of gene-environment interplay throughout the progression of these allergies [36]. The “hygiene hypothesis,” first proposed by Strachan [37], posits that reduced microbial exposure—due to smaller family sizes and improved sanitation—impairs immune system development, increasing susceptibility to allergic disease. Supporting this, children raised in anthroposophic environments—with minimal antibiotic or vaccine exposure and adherence to organic diets—show lower rates of allergies [38,39].
Reduced exposure to diverse diets and microbes during infancy may hinder the development of immune tolerance, potentially leading to a rise in immune-related disorders in industrialized populations [40]. Allergic sensitization refers to the immune system’s initial response upon first exposure to an allergen [4,41]. Two primary routes of sensitization have been described. Primary food allergens—commonly referred to as Class 1 allergens associated with egg, milk, and peanut—induce sensitization through the gastrointestinal tract [42]. Secondary allergens like Bet v 1, derived from birch pollen, sensitize via the respiratory tract and can cross-react with homologous nutritional allergens like Mal d 1 present in apples, resulting in oral allergy syndrome [4,43,44,45,46].
Epicutaneous sensitization—where allergens enter through inflamed or damaged skin—has also been proposed as a sensitization route. In murine models, such exposure leads to intestinal mast cell expansion and systemic anaphylaxis [47,48,49].
Multiple factors influence the likelihood of sensitization:
  • Allergen structure and stability: Resistance to gastrointestinal degradation enhances allergenicity [5,6,50].
  • Epithelial barrier integrity: Genetic mutations (such as those in the filaggrin gene) along with environmental influences—like alcohol consumption, pathogen exposure, and use of NSAIDs—can impair mucosal barrier integrity, thereby promoting allergen entry [51,52,53].
  • Adjuvant effects of food matrices: Nonallergenic components and microbial products in foods may act as immunological adjuvants, promoting sensitization [6].
Conversely, secretory immunoglobulins—particularly SIgA and SIgM—strengthen mucosal defenses and promote oral tolerance. The heightened intestinal permeability and susceptibility to allergen-induced anaphylaxis observed in mice lacking SIgA transport can be mitigated through the induction of regulatory T cells [54,55].

2. Mitigation Strategies for Food Allergens

2.1. Allergen Elimination and Control Strategies

Managing allergens in the food supply chain necessitates a comprehensive, preventive strategy aimed at their elimination or reduction. One approach involves genetic modification or selective breeding to eliminate or suppress genes encoding allergenic proteins, such as the targeted elimination of Ara h 2 and Ara h 6 proteins in peanuts through gene editing [56]. Enzymatic hydrolysis is another promising approach, in which proteolytic enzymes cleave allergenic epitopes to decrease immunoreactivity. This method has proven effective in hydrolyzed milk and wheat products [57,58]. However, proteolysis may not fully eliminate allergenic potential. For example, Sen et al. [59] observed that Ara h 2 allergen present in peanut retained IgE-binding regions of allergenic proteins post-hydrolysis, especially when disulfide bonds were intact. This highlights the importance of linear epitopes in allergen stability, and suggests that reducing disulfide bonds alone may not sufficiently degrade allergenic regions.
In addition to ingredient-level interventions, allergen control measures begin with careful raw material management. This includes supplier verification and traceability systems to ensure allergen-free inputs [60]. In manufacturing, dedicated lines, validated sanitation, and equipment separation are necessary to avoid cross-contact [61,62,63]. Hazard Analysis and Critical Control Points (HACCP), ingredient monitoring, and accurate food labeling are central to allergen risk management. Finally, workforce training and allergen awareness programs are vital for maintaining safety throughout the supply chain [64].

2.2. Influence of Processing Methods on the Mitigation of Food Allergens

Food processing is essential for altering allergenic characteristics, simultaneously improving safety, extending shelf life, and preserving nutrient quality. Physical, chemical, and biochemical processing methods can influence allergenicity based on food type and processing conditions [65,66,67]. The structural integrity and allergenic potential of proteins are significantly influenced by processing. Changes in physicochemical properties and protein stability can alter immunogenicity [57,68,69]. The food matrix can modulate these effects, at times enhancing allergenic responses [70,71]. These treatments could decrease, enhance, or leave allergenicity unchanged. Importantly, certain processing methods may generate neoallergens—novel allergenic molecules formed during food treatment—that have the potential to exacerbate allergic responses [72,73,74].
Both heat-based and non-heat-based processing methods can modify the structure of allergenic epitopes, potentially decreasing allergenicity by breaking down key immune-reactive sites or, conversely, generating new allergenic compounds. The phenomenon of neoallergen formation has been recognized since the 1970s, when it was reported that processed foods could elicit allergic responses even when raw forms did not [75]. Subsequent findings identified neoallergens in processed pecans [76] and wheat flour [77]. Processing typically reduces allergenicity in many foods. Fermentation boosts nutrient quality and food preservation, in addition to impacting allergenicity. Lactic acid bacteria, for example, degrade IgE-binding epitopes in milk proteins, such as β-lactoglobulin, thus reducing allergenicity [78]. However, fermentation’s effects are product-specific. Soy sauce, a fermented product, retains allergenicity despite microbial activity [79], while fermented dairy products such as yogurt often show reduced immunogenicity due to acid denaturation and proteolysis [80,81].

2.3. Thermal Processing and Its Effects on Allergenicity

Thermal processing is a commonly employed technique throughout the food manufacturing industry that can substantially modify the molecular structure and allergenic characteristics of proteins. Heat-induced denaturation leads to unfolding of proteins, disruption of disulfide bonds, and potential destruction of IgE-binding epitopes. For example, heating egg proteins such as ovomucoid or ovalbumin at 100°C for extended periods reduces their IgE-binding capacity [81]. Similarly, heat-based processes like roasting or boiling peanuts can alter allergenic components, including Ara h 1 and Ara h 2, potentially decreasing or, in specific situations, increasing their allergenic effects (Table 1) [82,83].
Baking and extrusion may reduce the allergenicity of wheat and soy by Maillard reactions and protein aggregation, although new allergenic determinants can sometimes be formed [84]. In specific instances, heat processing can raise resistance of certain allergens to digestive enzymes, potentially enhancing their capacity to provoke allergic responses. Thus, heat treatments must be carefully optimized based on food type and target allergen.
Application of humid thermal methods has been found to lessen the immune-reactive characteristics of various food substances. For instance, sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) revealed a marked loss of protein bands in canned tuna and salmon extracts compared to raw or conventionally cooked samples. Immunoblot analyses revealed very limited IgE-binding activity in canned fish extracts. Results from ELISA inhibition assays and oral food challenges involving subjects allergic to canned salmon demonstrated a reduction in allergenic reactivity, suggesting that some fish allergens may be more heat-sensitive than previously believed [60,85].
Similarly, kiwi fruit allergy provides a compelling case of heat-mediated allergenicity reduction. Differences in individual allergic responses often correlate with protein variability and cross-reactivity with airborne allergens such as pollen. However, steam-treating kiwi at 100°C for five minutes and then mechanically processing it markedly diminished allergic symptoms in certain pediatric cases [86,87,88].
For milk allergens, β-lactoglobulin's ability to bind IgE antibodies was evaluated both in its purified form and as part of whole milk using the FEIA-CAP inhibition method. While mild heating at 74°C caused only slight reductions, significant decreases in IgE binding were observed at 90°C. Although binding decreased, elevated levels of β-lactoglobulin were still capable of neutralizing comparable quantities of IgE antibodies regardless of the heat treatment applied [80].
Hypersensitivity responses arise when IgE antibodies recognize particular regions on allergenic proteins. The interaction causes clustering of IgE receptors on mast cells or basophils, prompting the secretion of histamine and various inflammatory agents [89]. Thermal sensitivity varies by epitope type: conformational epitopes are more easily denatured, while linear epitopes are more heat-resistant [57,90]. As a result, heating may reduce allergenicity in some foods like lentils, enhance it in others like peanuts, or have no effect, as observed in boiled peas [91,92].
Pollen-related allergens in fruits and vegetables often become less immunogenic after heating. However, allergens in foods like shrimp can remain heat-stable and retain immunogenicity post-cooking. The type of epitope also affects stability: conformational epitopes, reliant on three-dimensional protein folding, are more susceptible to denaturation during processing, whereas linear epitopes may resist heat but are subject to enzymatic or chemical alteration. Genetic engineering allows precise modification of such epitopes to reduce allergenicity.
Protein structures respond differently to thermal conditions: mild heat (70–80°C) disrupts secondary structures, moderate heat (80–90°C) affects disulfide bonding, and intense heat (90–100°C) induces aggregation [90,93]. Heat may also initiate Maillard reactions, where lysine residues form covalent bonds with sugars, generating advanced glycation end products that could increase allergenicity [90].
Heating can enhance digestibility and reduce allergenicity in legumes such as chickpeas, lentils, lupins, and black gram [94]. In contrast, it can diminish the digestive breakdown comprising peanut allergens, Ara h 1 and Ara h 2 [83,95]. Thermal processing may also produce neoantigens, which elicit stronger immune responses [69,83]. The Maillard reaction is a key mechanism behind this neoantigen formation, where heat-induced protein-sugar interactions yield glycation products with heightened immunogenic potential [96]. According to a study conducted by [97], blood sera collected from 57 individuals analyzed with suspected meat allergy exhibited enhanced IgE affinity was higher for raw meat extracts than for those heated at 140°C for 20 minutes, suggesting thermal degradation of most allergens. However, chicken was an exception, as six out of 24 sera exhibited a stronger reaction to heated chicken meat. Common allergenic proteins in raw and cooked chicken were identified at 17–66 kDa, while heat-labile proteins (45 and 150 kDa) disappeared and neoallergens emerged in the 14–90 kDa range.
Elevated temperatures and prolonged heating have been demonstrated to markedly diminish the IgE-binding ability of wheat proteins. When wheat flour and dough (flour mixed with 50% water) were subjected to heat treatments at 80°C, 100°C, and 120°C for 10, 20, and 60 minutes, both neutral and acidic protein extracts showed reduced IgE reactivity in skin prick tests (SPT) and radioallergosorbent tests (RAST) using sera from cereal-sensitive adults. No further reduction in allergenicity was observed beyond 10 minutes at 120°C, indicating a thermal stability threshold. Pooled sera RAST scores dropped shifting from class 4, which corresponds to untreated flour, down to class 2 following heat treatment. Notably, heated dough demonstrated lower allergenic potential than heated flour at 80°C and 100°C, though this difference was no longer evident at 120°C [98].
Heat treatment by microwave heating of kiwi fruit to 40, 60, 80, and 90°C resulted in a progressive reduction regarding allergenic potential evaluated through skin prick tests (SPT) among three individuals allergic to kiwi. At 90°C, SPT responses were negative in two patients and markedly reduced in the third, indicating significant thermal degradation of allergenic proteins [99].
Protein solubility in minced beef decreases with extended heat treatment. Following heating at 85°C for up to 2 hours, SDS–PAGE revealed faint bands at 17.8, 19, 14, 20, 45, and >60 kDa, which were still present in thoroughly cooked samples (80°C, 20 min) via immunoblotting. Among these, the 17.8 kDa protein showed the highest IgE-binding activity among individuals diagnosed with an allergy to cooked beef (positive DBPCFC). In minced beef heated at 80°C, bovine serum albumin and gamma-globulin became undetectable after for durations of 10 and 3 minutes, consequently. Conversely, purified serum albumin maintained its stability when heated to 95°C for 15 minutes, while purified gamma-globulin was completely denatured after 15 minutes at 65°C [100].
Dry heat processing can influence the capacity of foods to cause allergic reactions through complex reactions including the Maillard reaction and enzymatic browning. These processes can irreversibly alter or destroy conformational epitopes. For example, Gruber et al. [101] demonstrated that cherry allergen Pru av 1 loses its IgE-binding capacity due to the Maillard reaction and oxidation catalyzed by polyphenol oxidase. In a similar manner, roasting hazelnuts decreased their allergenic potential, especially in people sensitized to birch pollen–associated allergens like Cor a 1.04 and Cor a 2 [102]. However, five out of seventeen birch pollen–allergic individuals still reacted to roasted hazelnuts in food challenge tests conducted under double-blind placebo-controlled conditions, indicating that roasting does not eliminate allergenicity for all patients. These findings suggest that individual sensitivity to conformational versus linear epitopes is a critical consideration in evaluating the capacity to cause allergic reactions in thermally processed foods. Conversely, Maleki et sl. [103] found that thermal processing actually enhanced the IgE reactivity of key peanut allergens Ara h 1 and Ara h 2 indicating that heating may enhance peanut allergenicity in some cases.
Because processing alone may not eliminate all allergens, integrating multiple strategies is more effective [104]. Thermal techniques such as blanching, pasteurization, canning, and roasting offer accessible and cost-effective allergen mitigation and microbial control [105]. Freezing inhibits microbial growth below −9.5°C (15°F), contributing to food stability [60,79]. Drying enhances shelf life and lowers storage costs, though its allergen-modifying effects vary. Edible crickets with three major allergens identified: actin, hexamerin-like protein 2, and tropomyosin, particularly Gryllus bimaculatus (two-spotted cricket), are increasingly recognized for their high protein content; nevertheless, knowledge about how processing affects their allergenicity is limited. Evidence showed that tray drying caused a greater decrease in the allergenic potential of heat-sensitive proteins [106].
Fiocchi and colleagues [107] assessed immune reactions to beef processed by freeze-drying and homogenization in a group of ten children diagnosed with sensitivity to beef proteins. Each participant showed positive responses in skin prick testing for both uncooked and heated beef, as well as positive placebo-controlled, double-blind food challenge test outcomes after consuming 180 grams of beef subjected to heat treatment at 100°C for 5 minutes. Untreated bovine serum albumin elicited positive responses in all children in skin testing, with half also responding during the challenge. In contrast, only one child showed a positive skin test to freeze-dried beef, and none exhibited reactions in the challenge test. Beef prepared by homogenization did not provoke any positive test responses. In a related study, the same team [106] reported minimal immune reactivity to freeze-dried and homogenized beef and freeze-dried lamb meat, with just a small fraction (2 of 12) of children with confirmed beef protein sensitivity affected [88].

2.4. Non-Thermal Processing for Allergen Reduction

Non-heat-based processing approaches have attracted significant interest as effective approaches to lower food protein allergenic potential without compromising nutritional value, taste, and functional characteristics. Unlike traditional thermal treatments, non-thermal methods such as high-pressure processing (HPP), cold plasma, pulsed electric fields (PEF), ultraviolet (UV), and ultrasound (US), treatments provide more targeted approaches for modifying allergenic proteins. These techniques can disrupt protein structures, mask or cleave IgE-binding epitopes, and trigger biochemical changes that reduce immunoreactivity. With the global increase in food allergy prevalence, non-thermal interventions provide a compelling alternative for developing hypoallergenic food products while maintaining consumer acceptability and regulatory compliance (Table 2).

2.4.1. High Hydrostatic Pressure and Allergenicity in Foods

High hydrostatic pressure (HHP) treatment is a preservation technique which exerts pressures between 100 and 600 MPa to food items, usually conducted at room or chilled temperatures without using heat. This treatment is particularly impactful on protein molecules, as it disrupts non-covalent interactions for example, hydrogen bonding, ionic bonds, hydrophobic forces, in addition to van der Waals interactions [108,109]. These interactions are fundamental to maintaining protein secondary, tertiary, and quaternary conformations. However, unlike thermal processing, HHP generally does not cleave covalent bonds such as peptide bonds or disulfide bridges unless extremely high pressures or extended treatment durations are applied [110]. As a result, while the primary amino acid sequence typically remains intact, HHP can lead to significant unfolding and denaturation of proteins, modifying their structural and functional characteristics [111].
One of the most critical implications of HHP-induced protein denaturation is its effect on allergenicity. Food allergens are proteins or glycoproteins that possess immunologically active regions—referred to as epitopes, which are identified by Immunoglobulin E (IgE) antibodies in sensitized individuals [112]. Epitopes can either be conformational, shaped by the protein’s three-dimensional folding, or linear, constituted by a continuous sequence of amino acids. HHP has the potential to disrupt these epitopes, especially conformational ones, by unfolding the protein structure and eliminating the spatial arrangements necessary for epitope recognition. Consequently, the IgE-binding capacity of many food allergens is reduced after HHP treatment [84].
In addition to epitope disruption, HHP can also cause protein aggregation. During unfolding, reactive groups become exposed, leading to intermolecular interactions and protein cluster formation This aggregation can result in the concealment of linear epitopes or the creation of neoepitopes—new epitopic structures that may be more or less allergenic depending on the protein and processing conditions [113]. Furthermore, HHP-treated proteins often become more susceptible to proteolysis. The unfolding of the protein enhances accessibility for digestive enzymes such as pepsin and trypsin, promoting more effective breakdown during gastrointestinal digestion. This reduced stability and increased enzymatic degradation decrease the likelihood of allergenic epitopes surviving digestion and triggering immune responses [112].
The efficacy of HHP in reducing allergenicity is not uniform across all food proteins and is dependent on multiple variables such as pressure magnitude, duration of treatment, and temperature, pH, and the composition of the surrounding food matrix. For instance, pressures exceeding 400 MPa are generally required to significantly reduce allergenicity in robust proteins like β-lactoglobulin (milk originated), tropomyosin (shellfish originated), or ovomucoid (egg originated) [114]. Additionally, the existence of additional food constituents, including lipids and carbohydrates, or salts—can stabilize protein structures or shield epitopes, thereby diminishing the effectiveness of HHP [115,116]. These matrix effects must be considered when designing HHP-based allergen control strategies.
HHP may induce modifications in the structure of allergenic proteins, which can lower their immunoreactivity by disrupting conformational epitopes. However, in some cases, HHP can expose hidden epitopes or modify existing ones, resulting in unchanged or even increased allergenicity. The result varies according to the type of protein, the intensity of the pressure exerted, and the inclusion of additional treatments such as heat alongside HHP. For β-lactoglobulin (BLG), one of the principal milk allergens, exposure to 600 MPa resulted in lasting changes in its secondary and tertiary structures, accompanied by the liberation of free thiol groups and an increase in surface hydrophobicity [117]. These structural changes enhanced the protein’s IgE-binding capacity, indicating increased allergenicity post-treatment.
Kato et al. [118] demonstrated that HHP, when combined with 8 M urea, significantly reduced rice allergen levels. The proposed mechanism involved structural damage to allergens caused by pressure, followed by enhanced extraction with urea to remove these proteins. Egg allergenicity was shown to decrease when pressure was applied in combination with heat. Furthermore, the joint application of pressure and heat markedly decreased the allergenic response to hen’s egg, compared to either treatment alone [119]. HHP applied to soybean sprouts at treatment at 400 MPa produced considerable reductions in the levels of Gly m 1, a key soybean allergen [120]. The treatment also reduced antigenicity of soybean protein isolates, with modifications in protein structure likely responsible for the lowered immunoreactivity. Overall, HHP is a beneficial strategy for allergen mitigation in food systems. However, the effects are protein-specific and process-dependent. When combined with thermal treatments or other chemical agents, HHP may significantly reduce allergenicity, making it an important strategy in the development of hypoallergenic foods (Table 3).

2.4.2. Pulsed Electric Field (PEF) Technology and the Influence It Has on Food Allergenicity

Pulsed electric field (PEF) processing is an advanced, non-heat-based method in which brief, high-intensity electrical pulses—generally ranging from 1 to 50 kV/cm—are delivered to biological materials, including tissues and liquids. This approach results in electroporation, a temporary increase in cell membrane permeability caused by dielectric breakdown, facilitating improved mass transfer and promoting the release of internal constituents like proteins, lipids, and bioactive molecules [121,122,123,124]. PEF has gained broad acceptance across multiple domains within the food industry. Its applications range from microbial inactivation and juice extraction to drying pre-treatment, seed enhancement, detoxification of mycotoxins, and starch transformation [125,126,127,128]. Fundamentally, PEF’s mechanism involves inducing electroporation in cell membranes through high-intensity, short-duration pulses. These pulses lead to reversible or irreversible structural modifications embedded in the membrane, thereby enhancing the transfer of substances and aiding in the recovery of intracellular compounds [123,124].
The performance of PEF systems is closely tied to the electrical behavior of cell membranes, which are largely composed of lipid bilayers acting as insulators. These membranes isolate the conductive environments inside and outside the cell, making each cell resemble a microscopic capacitor [122]. When immersed in a conductive medium and underwent an applied electric field, the cell alters the field’s distribution, causing the membrane regions aligned with the field to experience intensified polarization at their poles [124].
A standard PEF system consists of a pulse generator capable of delivering voltages between 10 and 80 kV/cm, a treatment chamber equipped with insulated electrodes, a cooling unit, and a flow mechanism to ensure uniform treatment [122,125]. The system design is tailored through parameters like pulse duration, waveform, frequency, and electrode configuration. Pulse modes can be exponential decay, sinusoidal, monopolar, or bipolar. For instance, monopolar pulses have a constant polarity, while bipolar ones alternate it, minimizing polarization effects on electrodes. Different waveforms—such as square, decaying exponential, logarithmic, or oscillatory—offer varying benefits. Square waves, for example, ensure consistent membrane disruption, whereas decaying pulses can reduce energy use and thermal damage. Selecting the appropriate pulse configuration is essential to attain the targeted outcomes—such as microbial inactivation, compound extraction, or texture alteration—while maintaining the food’s nutritional value and sensory attributes. Advanced control and safety systems are incorporated into industrial PEF equipment to ensure continuous, hygienic, and reliable processing [122,125,129,130]. Following exposure to the electric field, the cell—functioning as a dielectric sphere in a conductive environment—undergoes enhanced localized electric field intensity at its poles. These intensified regions are key sites for electroporation, often experiencing a field strength far exceeding the average applied to the bulk medium [124].
Although investigations into how PEF affects protein structures are still relatively scarce, multiple studies have documented significant structural and functional modifications that vary with the protein’s nature [131], and evidence indicates that PEF may alter proteins’ secondary and tertiary conformations. These structural changes are probably a result of the ionization of particular chemical groups and the disturbance of electrostatic forces [132,133]. Initially, protein molecules undergo polarization, which causes hydrophobic amino acid residues to be revealed to the surrounding solvent. This, in turn, promotes protein unfolding and aggregation when subjected to high-intensity electric fields [134]. In the food sector, PEF treatment has attracted significant attention due to its energy efficiency, capacity to retain nutritional value, and suitability for processing liquid-based food systems [135]. For instance, treatment of soybean protein isolates with PEF has demonstrated alterations in physicochemical properties, including denaturation and aggregation behaviors [136]. Similarly, studies on purified enzymes, including horseradish peroxidase and pectin esterase, showed reduced enzymatic activity after treatment, mainly as a consequence of alterations in the proteins’ structural conformation [137]. However, it is important to consider that these changes may not result solely from the electric field itself; thermal effects due to associated Ohmic heating could also contribute to the observed outcomes [137]. Johnson et al. [138] examined how PEF treatment influences the structural integrity of specific food allergens, namely the peanut allergens Ara h 2 and Ara h 6, as well as the apple allergens Mal d 3 and Mal d 1b, all of which were produced through heterologous expression. Structural changes were assessed using circular dichroism spectroscopy and gel-filtration chromatography. The results indicated that PEF application caused no substantial changes in either the secondary structure or the degree of allergen aggregation. This outcome implies that PEF functions as a structurally gentle food processing technique, exerting minimal influence on the conformation of purified food allergens.
An investigation assessing PEF application at 25 kV/cm and 50 °C on Pru p 3, the primary allergenic protein in peach, showed that the treatment caused structural denaturation, as identified through ELISA with rabbit IgG. Nonetheless, this process did not alter the IgE-binding ability of Pru p 3, as demonstrated by a competitive fluorescent immunoassay using sera from individuals allergic to peach. Additionally, skin prick test results varied among individuals; more than 50% of participants exhibited increased skin reactivity following PEF treatment, indicating that patient-specific sensitization patterns may persist despite structural alterations in the allergen [139].
Experimental studies have demonstrated that PEF applications at high field strengths, particularly at 25–35 kV/cm for lengths spanning 60 to 180 µs, significantly impact the structural conformation of ovalbumin, including the α-helix content, thereby altering its immunogenic properties. The most substantial reduction in immunoreactivity was observed at 35 kV/cm with 180 µs treatment time [140], demonstrating the capability of this technology to lower the egg proteins’ allergenic capacity.
In a molecular dynamics simulation study, Vanga et al. [141] explored the effects of dynamic and static-field conditions (2450 MHz frequency and 0.05 V/nm energy) at various temperature levels (300, 380, and 425 K) examining the structural characteristics of Ara h 6, one of the major allergenic proteins in principal peanut. The simulations revealed significant conformational modifications under all tested conditions, implying potential impacts on the allergen’s functional properties. Subsequently, Vanga et al. [142] experimentally treated peanut flour with varying electric field applications of 10, 15, and 20 kV for 60–180 minutes. It was reported that time-dependent increases in α-helix conformational changes, probably resulting from the formation of novel random coil conformations and protein aggregates generated in the course of processing.
However, not all findings confirm a consistent reduction in allergenicity. For instance, Johnson et al. [138] reported no significant structural changes in allergenic peanut proteins of Ara h 2 and Ara h 6 (peanut 2S albumins), Mal d 1, and Mal d 3 (apple allergens) following electric fields application in the range of 0 to 35 kV/cm, with 2 Hz frequency and up to 130 kJ/kg energy input. Similarly, Paschke [143] found no notable reduction in celery allergen content when treated with a 10 kV PEF at 50 Hz.
A recent study investigated the modulation of ovalbumin (OVA) allergenicity using PEF treatment, highlighting structural changes linked to reduced immunoreactivity. PEF treatment at 6 kHz inhibited IgE and IgG1 binding by 30.41%, accompanied by visible microstructural surface cracks and loss of secondary structural elements in the protein. Spectroscopic analyses demonstrated a blue shift of the amide I spectral band, reductions in α-helical and β-sheet structural composition, and conformational changes in disulfide bonds. Increased fluorescence intensity suggested exposure of hydrophobic residues such as tryptophan and tyrosine. Molecular dynamics simulations further confirmed reduced structural stability and hydrogen bonding. These findings suggest that PEF disrupts allergenic epitopes by altering protein conformation, supporting its potential application in developing hypoallergenic egg products [144]. Compared with both thermal and alternative non-thermal methods, PEF seems to have a relatively modest effect on altering the structure and immunoreactivity of food allergens. These inconsistent outcomes highlight the need for further optimization of PEF parameters to achieve reliable and reproducible allergen mitigation.

2.4.3. Pulsed Ultraviolet (PUV) Light and Its Impact on Food Allergenicity

Pulsed light (PL) technology utilizes a series of brief, high-intensity bursts of broad-spectrum white light, predominantly containing 54% ultraviolet (UV) wavelengths followed by 26% light in the visible light spectrum along with 20% radiation in the infrared spectrum [145]. The PUV light spans wavelengths within the 200–1000 nm range, allowing it to deliver intense light energy within a very short duration [146]. As a result, PUV light can achieve energy intensities up to a thousand times greater than those of traditional continuous UV systems (Shriver and Yang 2011). The technology utilizes xenon flash lamps to generate short-duration (typically <1 ms), high-energy light bursts at a frequency ranging from 1 to 20 Hz. These pulses result in rapid microbial inactivation via photochemical, photothermal, and photophysical mechanisms, primarily targeting nucleic acids and protein structures. In addition to microbial control, PUV has been investigated for its potential to alter food allergens, degrade toxins, and induce structural and functional changes in biomolecules such as proteins and lipids, without significantly elevating product temperature or compromising sensory and nutritional qualities [147,148,149,150].
In food applications, PUV light has been investigated for its capacity to alter the structural configuration of allergenic proteins. This includes altering conformational epitopes and promoting protein aggregation, which may reduce allergenicity [146,150]. However, post-treatment re-association of peptide fragments might cause the development of novel epitopes—new structures that could increase the propensity of the food to cause allergies [151].
The functional mechanisms of PUV light inactivation can be grouped into light-driven photochemical, photothermal, and photophysical effects. These impacts play a role in chemical transformations and modifications in protein structure, influenced by rapid heating and the intermittent delivery of high-energy pulses [146,152].
When applied to peanut samples—including raw, roasted, and peanut butter extracts—PUV light at distances of 10.8, 14.6, and 18.2 cm for 1–6 minutes caused a marked lowering of the abundance of Ara h 1, Ara h 2, and Ara h 3 allergenic proteins, as evidenced by SDS-PAGE analysis. Increased energy doses (111.6 to 223.2 J/cm2) and longer exposure times further enhanced this reduction, while greater distance from the light source diminished the effect. ELISA results showed that IgE binding decreased approximately threefold in raw peanut extracts and sevenfold in peanut butter slurry compared to untreated controls. These effects are attributed to changes regarding protein solubility and the generation of insoluble aggregates as a result of PUV processing [153].
PUV light treatment of soy extracts for 2, 4, and 6 minutes led to a time-dependent decline in major allergens such as glycinin and β-conglycinin. Indirect ELISA using sera from soy-allergic individuals showing decreases in IgE binding of 20%, 44%, and 50%, respectively. The detected decrease is likely associated alongside the creation of aggregated structures of allergenic proteins when subjected to PUV [146]. PUV application also reduced the immune response potential of the soy allergens Gly m5 and Gly m6 when applied at a distance of 8–10 cm for up to 6 minutes. Gel bands corresponding to Gly m5 disappeared after just 2 minutes of treatment, while Gly m6 bands remained visible even after 6 minutes, indicating differential susceptibility to degradation or precipitation [154].
Similarly, ultraviolet-C (UV-C) treatment has shown promising effects in reducing the capacity of milk proteins to cause allergic reactions. Significant reductions in IgE binding were also observed in milk allergens including α-lactalbumin, α-casein, and β-lactoglobulin after 15 “minutes of UV-C exposure. The decrease in immunoreactivity is likely due to alterations in discontinuous epitope structures. The enhanced effect in whey solutions has been linked to the greater pulse intensity and energy associated with PUV treatment [155]. The enhanced allergen reduction observed in whey proteins subjected to PUV treatment could be linked to the higher energy and pulse rate involved in the process [156].
For egg white proteins, ultraviolet exposure caused both aggregation and backbone cleavage; however, no substantial changes in immunoreactivity were observed compared to untreated controls. This suggests that the structural changes induced by UV light may not significantly affect allergenic epitopes in egg proteins [157].
Exposure of raw and shrimp extracts prepared by boiling, 5 mg/mL, to PUV light at a pulse duration of 360 µs, at a frequency of three pulses per second, and a distance of 10 cm led to a notable and irreversible reduction in allergenic reactivity. This reduction has been attributed resulting in the creation of high molecular weight protein complexes, likely due to cross-linking between tropomyosin and other heat-sensitive proteins [145,146]. This effect was correlated with the generation of high–molecular weight compounds through cross-linking between tropomyosin and other heat-sensitive proteins during the treatment [145].
PUV light has shown the capacity to lower allergenic potential in soy, peanut, milk, and shrimp products, though it appears ineffective for modifying egg allergens. Although these findings are encouraging, specifically aimed at producing hypoallergenic foods, additional research—such as clinical trials and in vivo experiments—is required to validate its wider use in food processing [142]. However, not all studies report a reduction in allergenicity with UV processing. For instance, Manzocco et al. [157] found that although ultraviolet exposure led to aggregation and backbone cleavage in egg white proteins, these structural changes did not significantly affect their immunoreactivity compared to untreated samples, suggesting that the epitope structures remained largely unchanged.
Overall, the evidence suggests that PUV light may be capable of lowering allergenic potential in foods like soy, milk, shrimp, and peanuts, while it appears to have limited impact on egg allergens (Table 4). Although the technology shows potential for developing hypoallergenic food items, additional research —particularly clinical and in vivo studies—is essential before broader adoption across the food processing industry [104].

2.4.4. Gamma Irradiation and Its Impact on Food Allergenicity

Radiation refers to the transfer or release of energy across space or a medium as waves or particles. It is broadly classified into two types: Nonionizing radiation possesses lower energy compared to ionizing radiation. Nonionizing radiation does not have sufficient energy to dislodge electrons from atoms or molecules and is typically considered non-harmful, causing little to no chemical changes. This category includes ultraviolet light, visible-spectrum radiation, infrared energy, microwaves, and radio waves—each possessing lower energy levels and typically not employed in food processing. In contrast, ionizing radiation carries sufficient energy to dislodge electrons, resulting in the formation of ions. This form includes energetic electromagnetic waves and subatomic particles capable of altering molecular structures. Among ionizing methods, gamma irradiation is considered one of the most straightforward techniques [164,165]. The reduction in immunoreactivity was linked to protein denaturation, assessed through parameters such as turbidity, surface hydrophobicity, and chromogenic reactivity. These findings suggest that clumping of allergenic proteins could have a crucial role in minimizing allergenicity post-irradiation [166]. Exposure of wheat germ agglutinin (WGA) to irradiation was found to initially cause polypeptide chain fragmentation, followed by the creation of large, insoluble, non-crystalline aggregates, ultimately leading to a decrease in allergenicity [167].
Radiation has been established as a practical approach for preserving food while maintaining its nutritional value and sensory qualities. It induces structural modifications in food proteins—including fragmentation, aggregation, cross-linking, and alterations to amino acids—that can influence their immunogenic properties [168]. These changes are largely mediated by reactive oxygen species formed during the radiolysis of water when proteins are irradiated in aqueous environments.
Exposure to ionizing radiation at doses of 10 and 50 kGy (administered at 10 °C with a dosage rate of 10 kGy per hour) altered the conformational epitopes in peanuts, leading to a significant reduction in IL-4 cytokine production by splenocytes from sensitized mice [169]. Similarly, Luo et al. [168] demonstrated that irradiation of purified peanut allergen proteins and whole peanut extract (at 1, 3, 5, and 10 kGy, 10 °C) resulted in a notable decline in IgG binding, as detected by ELISA, with higher radiation doses enhancing the effect. Interestingly, up to 5 kGy, the IgG response for whole peanut extract exceeded that for Ara h 6 alone, potentially due to the presence of other components in the extract that shielded Ara h 6 epitopes.
A comparable pattern was reported by Zhenxing et al. [170] in shrimp, where protein extracts irradiated at doses between 3 and 15 kGy (10 °C, 1 kGy/h) showed reduced IgE binding. However, intact shrimp muscle initially exhibited increased IgE reactivity up to 5 kGy, which then declined at higher doses. A dose-dependent response to irradiation was observed in a study on cow milk allergy by Lee et al. [156], where IgE binding to isolated β-lactoglobulin (β-lg) increased up to a dose of 5 kGy. Beyond this level, protein agglomeration occurred, likely masking specific epitopes and causing a decline in allergenic potential. In contrast, Kaddouri et al. [171] reported enhanced recognition of anti-β-lg IgG antibodies following irradiation with 3–10 kGy at 13 Gy/min rate of both liquid-form and freeze-dried bovine milk and whey. These differing results may be attributed to the sample form—whether β-lg was isolated or present in a milk-based matrix—and the class of antibodies applied for detection. These findings suggest that the allergenic response of purified proteins and whole food extracts to irradiation can vary, which holds relevance for food industry applications. Furthermore, differences in dose rate among studies indicate that the impact of varying dose rates at a constant radiation dose on protein allergenicity remains unclear.
In other research, irradiation of whole almonds, cashews, and walnuts did not cause modifications in the structure of allergenic proteins or affect their capacity to trigger allergic reactions [172]. Conversely, investigations have demonstrated that irradiation might elevate immunological responsiveness of gliadin and wheat flour [173]. These opposing results may stem from the physical state of the samples—solid versus solution—which can influence how effectively irradiation reduces allergenicity. Overall, applying irradiation to food allergens in liquid form appears to be the most promising approach for minimizing their immunoreactivity.
Ovalbumin, an egg allergen, was reduced after gamma irradiation with cobalt-60 at 100 kGy, whereas treatment at 10 kGy produced no notable effect [174]. This reduction was believed to result from alterations in the protein’s molecular weight. Gamma irradiation is known to promote protein crosslinking—such as disulfide bond formation—and enhance hydrophobic interactions, both of which can lead to protein aggregation [175]. However, a 100 kGy dose exceeds levels typically considered safe for food processing, as doses up to around 10 kGy are generally recognized as safe for consumption [176]. In a separate study, Seo et al. [177] also reported reduced ovalbumin immunoreactivity in white cake samples irradiated at 10–20 kGy.
In studies involving crustacean allergens, such as shrimp tropomyosin, gamma irradiation at doses of 7 kGy and higher resulted in the disappearance of the characteristic 36 kDa tropomyosin band on SDS–PAGE, indicating structural degradation. Additionally, IgE binding was significantly diminished at the highest doses tested [178]. Multiple studies have demonstrated the effectiveness of gamma irradiation in modifying allergenic proteins. To illustrate, Seo et al. [174] reported that ovalbumin, a principal allergen. Gamma irradiation may promote protein cross-link formation through the creation of disulfide linkages and intensified nonpolar molecular interactions, which promote aggregation and possible loss of immunoreactive epitopes (Table 5) [166].

2.4.5. High-Intensity Ultrasound and Its Impact on Food Allergenicity

Ultrasound technology has garnered considerable recognition in the area of food processing due to its non-thermal and energy-efficient nature. It involves the application of acoustic waves typically at frequencies beyond human auditory perception between 20 kHz and 100 kHz in food systems [180]. During the use of ultrasound at high intensity, it induces an occurrence described as acoustic cavitation—the process in which microbubbles form, expand, and collapse violently in a liquid environment [181]. The core mechanism involves cavitation, wherein alternating compression and rarefaction cycles form microbubbles that collapse violently, generating localized temperatures approaching 5000 K and pressures attaining 1000 atm [182]. These extreme conditions facilitate protein denaturation, potentially altering allergenic structures and epitopes. This collapse generates localized hot spots with extremely extreme heat conditions (up to 5000 K) and pressures (up to 1000 atm), although these effects are confined to the microscopic scale and last for only a few microseconds [183]. Alongside thermal effects, cavitation leads to intense shear forces, microjetting, turbulence, and formation of reactive species like hydroxyl radicals from water sonolysis), all of which can contribute to physical and chemical transformations in food matrices [184].
High-intensity ultrasound (HIU) has been explored for various food processing operations, including peeling of fruits and vegetables [185], reduction of oil content and shelf-life extension of fried products [186], and shortening of parboiling time in rice processing [187]. More recently, its potential to alter protein structure and reduce food allergenicity has attracted scientific interest. The primary mechanism by which HIU affects allergens lies in its capacity to alter the spatial structure of allergenic proteins. During sonication, proteins can undergo unfolding, aggregation, or fragmentation due to mechanical stress and localized heating, leading to changes in epitope exposure and antigenicity. Additionally, free radicals generated during cavitation may oxidize specific amino acid residues, contributing further to conformational changes [58,166].
Processing of shrimp samples to high-intensity ultrasound were conducted at a frequency of 30 kHz for durations ranging from 130 to 180 minutes and this treatment significantly reduced IgE-binding capacity in both isolated tropomyosin (the major shrimp allergen) and within unrefined shrimp protein extracts. In particular, IgE binding to the isolated tropomyosin was reduced by 81.3–88.5%, whereas binding in the shrimp extract decreased by approximately 68.9%, based on ELISA assays. Immunoblotting also revealed that prolonged ultrasound exposure brought about the formation of new, lower-molecular-weight protein bands, suggesting fragmentation of the allergenic protein. This structural disruption likely contributed to the observed decrease in allergenic reactivity. Interestingly, the treatment duration was relatively long, yet no significant temperature rise or quality deterioration of the shrimp product was reported [187]. This highlights one of the advantages of HIU as a non-thermal method for allergen mitigation. Zhenxing et al. [170] further confirmed that shrimp allergenicity decreased more significantly at elevated temperatures (50 °C) during ultrasound treatment. In another study, Zhang et al. [188] subjected shrimp tropomyosin (TM) to 15 minutes of ultrasound (100–800 W), reporting a substantial degradation of TM and associated reduction in allergenicity, as evidenced through ELISA and immunoblot analyses with sera obtained from allergic individuals. However, some shellfish allergens appear more resistant to ultrasound. Chen et al. [189] found minimal degradation and unchanged IgE-binding activity of arginine kinase (AK) in crayfish even after ultrasound treatment at 200 W and 30 °C for up to 180 minutes. Dairy proteins also showed limited response; Tammineedi et al. [155] found no meaningful alterations in SDS-PAGE profiles of α-casein, β-lactoglobulin, and α-lactalbumin after treatment with 500 W ultrasound at 20 kHz for up to 30 minutes. Similar findings were reported in other milk allergen studies [155,190].
In soy protein, Tammineedi et al. [155] demonstrated a 24% decrease in immunoreactivity following treatment using high-power ultrasound at 37 kHz for a duration of 10 minutes. The observed reduction is linked to changes in structural arrangements at the secondary and tertiary levels. Similarly, significant allergen reduction has been observed in roasted peanuts; Li et al. [187] reported decreases of 84.8% in Ara h 1 and 4.88% in Ara h 2 after exposure to 50 Hz ultrasound for two hours. Alterations concerning the architecture of the α-helical IgE-binding regions of these proteins are believed to account for this change [191,192]. Zhenxing et al. [170] further confirmed that shrimp allergenicity decreased more significantly at elevated temperatures (50 °C) during ultrasound treatment. In another study, Zhang, He et al. (2018b) subjected shrimp tropomyosin (TM) to 15 minutes of ultrasound (100–800 W), reporting a substantial degradation of TM and associated reduction in allergenicity, as evidenced by ELISA and immunoblotting employing sera obtained from individuals with allergies.
The efficiency of ultrasound appears to increase when combined with thermal treatment, suggesting a synergistic effect. Ultrasound processing offers several benefits, including reduced energy consumption, minimal chemical usage, enhanced mass transfer, and better retention of sensory qualities [193]. Given these advantages and its proven potential in reducing immunoreactivity in soy, peanuts, and shellfish, ultrasound is a compelling alternative to conventional processing methods. Nevertheless, optimization of key parameters—including frequency, length of treatment, and temperature—is essential to maximize its efficacy in allergen reduction across various food types. Overall, high-intensity ultrasound offers a promising approach to reduce the potential to trigger allergies in food proteins through an integration of physical and chemical effects (Table 6). While more data is needed to understand optimal parameters and food matrix-specific responses, current evidence supports its utility as part of an integrated strategy for allergen control in food systems [165].

2.4.6. Cold Plasma and Its Impact on Food Allergenicity

Cold plasma is primarily used for the purpose of microbial reduction and food decontamination owing to its lower energy expenditure and reduced required temperature ranges compared to traditional methods [199]. Plasma, a form of matter composed of ionized particles, is frequently described as the fourth state of matter encompasses a blend of reactive entities, for example ultraviolet photons, charged ions, electrons, and reactive radicals, molecules, and excited atoms. These components are formed through the energy absorption and ion production of gases and are capable of interacting with proteins, resulting in conformational changes. Since most food allergens are proteins, it is hypothesized that cold plasma treatment could similarly alter their structures through these reactive agents [165,199]. Various research efforts have revealed the potential of plasma treatment in reducing food allergenicity. For example, Venkataratnam et al. [200] applied cold atmospheric plasma (80 kV) for different durations (0 to 60 minutes) to dry, fat-extracted peanut flour and intact peanuts. Competitive ELISA using bovine serum albumin showed up to a 43% reduction in antigenicity, accompanied by secondary structure changes detected through circular dichroism. In another investigation, direct-application dielectric discharge-generated cold plasma treatment for 5 minutes at 30 kV and 60 Hz led to a 76% decrease in shrimp allergen tropomyosin’s immunoreactivity [166].
In one study, the impact of both direct and remote effects of cold atmospheric pressure plasma (CAPP) treatment on soy protein isolate immunoreactivity was examined [165]. Researchers measured factors such as mass of the sample, surface temperature, pH level, and hydrogen peroxide concentration. SDS-PAGE analysis revealed a noticeable decline of protein band density associated with to the primary allergenic components of soy, β-conglycinin (Gly m5) and glycinin (Gly m6). The most substantial decrease in immunoreactivity—ranging from 91% to 100%—was observed in the fraction of soluble proteins following direct CAPP treatment. Remote CAPP exposure also reduced immunoreactivity, though to a slightly lesser extent, with reductions of up to 89%. These reductions in immunoreactivity are linked to the disturbance or concealment of conformational epitopes by free radicals produced during plasma exposure, which hinders IgE antibody recognition [201]. Meinlschmidt et al. [154] reported that CAPP treatment of soy protein extracts for up to 10 minutes at 9, 10, and 11 kVpp (3.0 kHz) nearly eliminated Gly m5 allergen reactivity. This was likely due to alterations in conformational and linear epitopes or destruction of antibody binding sites, as shown through a sandwich ELISA employing human sera and monoclonal antibodies.
Alternatively, application of cold atmospheric plasma to milk α-casein at 13.56 MHz radiofrequency and 30.7 L/min argon gas flow for 5, 10, and 15 minutes. They found no significant reduction in immunoreactivity, possibly due to limited plasma exposure or insufficient treatment intensity [155]. These varied findings across food types suggest that cold plasma’s effectiveness in allergen reduction is not uniform and is influenced by factors including sample composition along with processing parameters. To date, reductions in allergen levels have been observed in soy, peanuts, shrimp, and wheat after cold plasma treatment, while no notable changes have been reported in milk proteins. Furthermore, concerns remain regarding the quality degradation associated with this method. Several studies have linked cold plasma processing to undesirable effects, including accelerated lipid oxidation, nutrient loss (such as vitamins), and compromised sensory qualities [202‒204]. Therefore, while cold plasma holds potential, its limitations highlight the need for continued research and the development of alternative food processing technologies.

2.4.7. Genetic Modification and Its Role in Reducing Allergenicity

Genetic modification (GM) involves intentionally changing an organism’s DNA through biotechnological techniques, enabling the introduction, deletion, or modification of specific genes to achieve desired traits, allowing for the insertion, deletion, or silencing of specific genes to achieve desired traits. In the context of food production, GM is employed to increase crop productivity, boost resistance against pests and diseases, and elevate nutritional or functional qualities. Over recent years, its potential to reduce food allergenicity has become a growing area of research, although concerns regarding safety, stability, and consumer perception remain significant. One strategy to reduce allergenicity in foods through GM involves silencing the expression of specific allergenic proteins by employing post-transcriptional gene silencing or co-suppression mechanisms. This approach aims to prevent the production of allergenic proteins during gene expression. However, uncertainties persist with respect to the long-term stability of these modifications. Incomplete inhibition or loss of gene silencing could reintroduce allergens, posing potential health risks to sensitized individuals. Additionally, removing key proteins might impact the structural and functional integrity of food products, as many allergenic proteins serve essential functions in metabolism and development of the source organism [79].
Public skepticism also surrounds the possibility that modifying protein structures may inadvertently result in the emergence of new allergenic determinants or epitopes that the immune system may fail to recognize as harmless. Therefore, a more targeted strategy—such as altering specific IgE-binding epitopes while maintaining the general configuration and function of the protein—could provide a safer and more efficient approach [205].
Experimental studies provide insights into these strategies. For instance, Rupa et al. [206] modified the IgE-binding sites of ovomucoid, a major egg allergen, by introducing glycosylation at known epitope locations. This post-translational modification significantly reduced IgE-mediated immune responses in a mouse model, demonstrating the feasibility of epitope-focused interventions. However, since the modified proteins were expressed in yeast cells rather than in the natural egg matrix, it remains unclear how such changes would impact the final food product. In a study targeting peanut allergens, Chu et al. [207] applied RNA interference to suppress the expression of Ara h 2 and Ara h 6, two major peanut allergens. Although this intervention significantly decreased the binding of IgE antibodies to these allergens, the whole peanut extract still retained immunoreactivity, likely due to the presence of other allergenic proteins. Interestingly, the silencing of these genes did not lead to any noticeable morphological changes in the peanut plants. Similarly, Dodo et al. [56] reported reduced IgE binding following genetic silencing of Ara h 2. In another example, Herman et al. [208] established that silencing Gly m Bd 30, a key soybean allergen, nearly eliminated IgE binding. Detailed protein analyses and microscopic evaluations confirmed that the modified soybeans were structurally and compositionally similar to their non-GM counterparts and did not produce any new allergens. Although these findings highlight the potential of GM approaches to reduce allergenicity (Table 7), broader questions concerning consumer acceptance, regulatory approval, and long-term safety must still be addressed.

3. Conclusions

The rising prevalence of food allergies necessitates the creation of efficient mitigation approaches that guarantee both consumer safety and food quality. A mounting body of evidence highlights the complex interplay between the molecular characteristics of food allergens, processing conditions, and the surrounding food matrix in determining allergenic potential. Multiple processing approaches, including thermal and non-thermal—induce distinct physicochemical modifications in food proteins, thereby influencing their digestibility, bioavailability, and IgE-binding capacity.
Thermal treatments such as moist heat generally reduce allergenicity by disrupting protein conformation and enhancing proteolytic susceptibility, while dry heat methods (e.g., roasting and baking) may increase allergenicity through the formation of neo-epitopes via Maillard reactions. Non-thermal technologies, including HPP, PEF, cold plasma, PUV, US, and gamma irradiation, offer promising alternatives for allergen mitigation by inducing conformational changes without compromising nutritional and sensory attributes. Additionally, microbial fermentation and enzymatic hydrolysis can effectively target linear epitopes, further reducing allergenic potential.
Despite these advances, translating reductions in IgE reactivity observed in vitro to clinically meaningful outcomes remain a significant challenge. The variability in digestive stability, structural resilience of allergenic proteins, and individual immune responses necessitates the use of complementary in vivo assessments including skin prick testing and oral food challenge procedures, and mediator release assays to validate hypoallergenic claims. Furthermore, a comprehensive understanding of both conformational and linear epitope modifications is essential for the design of processing strategies that minimize elicitation thresholds without compromising safety.
Future research should focus on standardizing allergenicity assessment protocols, integrating multi-hurdle approaches for simultaneous allergen and microbial risk reduction, and expanding studies to encompass emerging alternative proteins. Ultimately, a multidisciplinary approach that bridges food processing technology, immunology, and clinical validation is crucial for developing safe, effective, and industry-relevant solutions to mitigate food allergenicity and improve public health outcomes.

Author Contributions

Gulsun Akdemir Evrendilek: Literature review, writing original manuscript; Alper Güven: writing manuscript, formatting.

Declaration

The authors report no competing or conflicting interests.

References

  1. Sicherer, S.H. Clinical Implications of Cross-Reactive Food Allergens. Journal of Allergy and Clinical Immunology 2001, 108, 881–890. [Google Scholar] [CrossRef]
  2. Sicherer, S.H.; Sampson, H.A. Food Allergy: Epidemiology, Pathogenesis, Diagnosis, and Treatment. Journal of Allergy and Clinical Immunology 2014, 133, 291–307.e5. [Google Scholar] [CrossRef] [PubMed]
  3. Renz, H.; Allen, K.J.; Sicherer, S.H.; Sampson, H.A.; Lack, G.; Beyer, K.; Oettgen, H.C. Food Allergy. Nat Rev Dis Primers 2018, 4, 17098. [Google Scholar] [CrossRef]
  4. Valenta, R.; Hochwallner, H.; Linhart, B.; Pahr, S. Food Allergies: The Basics. Gastroenterology 2015, 148, 1120–1131.e4. [Google Scholar] [CrossRef] [PubMed]
  5. Breiteneder, H.; Ebner, C. Molecular and Biochemical Classification of Plant-Derived Food Allergens. Journal of Allergy and Clinical Immunology 2000, 106, 27–36. [Google Scholar] [CrossRef]
  6. Breiteneder, H.; Mills, E.N.C. Molecular Properties of Food Allergens. Journal of Allergy and Clinical Immunology 2005, 115, 14–23. [Google Scholar] [CrossRef] [PubMed]
  7. Chehade, M.; Mayer, L. Oral Tolerance and Its Relation to Food Hypersensitivities. Journal of Allergy and Clinical Immunology 2005, 115, 3–12. [Google Scholar] [CrossRef]
  8. Hsieh, K.-Y.; Tsai, C.-C.; Herbert Wu, C.H.; Lin, R.-H. Epicutaneous Exposure to Protein Antigen and Food Allergy. Clinical & Experimental Allergy 2003, 33, 1067–1075. [Google Scholar] [CrossRef]
  9. Lack, G. Update on Risk Factors for Food Allergy. Journal of Allergy and Clinical Immunology 2012, 129, 1187–1197. [Google Scholar] [CrossRef]
  10. Mittag, D.; Akkerdaas, J.; Ballmer-Weber, B.K.; Vogel, L.; Wensing, M.; Becker, W.-M.; Koppelman, S.J.; Knulst, A.C.; Helbling, A.; Hefle, S.L.; et al. Ara h 8, a Bet v 1–Homologous Allergen from Peanut, Is a Major Allergen in Patients with Combined Birch Pollen and Peanut Allergy. Journal of Allergy and Clinical Immunology 2004, 114, 1410–1417. [Google Scholar] [CrossRef]
  11. Bischoff, S.; Crowe, S.E. Gastrointestinal Food Allergy: New Insights into Pathophysiology and Clinical Perspectives. Gastroenterology 2005, 128, 1089–1113. [Google Scholar] [CrossRef]
  12. Sampson, H.A.; Sicherer, S.H.; Birnbaum, A.H. AGA Technical Review on the Evaluation of Food Allergy in Gastrointestinal Disorders. Gastroenterology 2001, 120, 1026–1040. [Google Scholar] [CrossRef]
  13. Sampson, H.A.; Anderson, J.A. Summary and Recommendations: Classification of Gastrointestinal Manifestations Due to Immunologic Reactions to Foods in Infants and Young Children. J Pediatr Gastroenterol Nutr 2000, 30 Suppl, S87–94. [Google Scholar] [CrossRef] [PubMed]
  14. Yocum, M.W.; Butterfield, J.H.; Klein, J.S.; Volcheck, G.W.; Schroeder, D.R.; Silverstein, M.D. Epidemiology of Anaphylaxis in Olmsted County: A Population-Based Study. Journal of Allergy and Clinical Immunology 1999, 104, 452–456. [Google Scholar] [CrossRef]
  15. Bock, S.A.; Muñoz-Furlong, A.; Sampson, H.A. Fatalities Due to Anaphylactic Reactions to Foods. Journal of Allergy and Clinical Immunology 2001, 107, 191–193. [Google Scholar] [CrossRef] [PubMed]
  16. Badina, L.; Barbi, E.; Berti, I.; Radillo, O.; Matarazzo, L.; Ventura, A.; Longo, G. The Dietary Paradox in Food Allergy: Yesterday’s Mistakes, Today’s Evidence and Lessons for Tomorrow. Current Pharmaceutical Design 2012, 18, 5782–5787. [Google Scholar] [CrossRef] [PubMed]
  17. de Silva, R.; Dasanayake, W.M.D.K.; Wickramasinhe, G.D.; Karunatilake, C.; Weerasinghe, N.; Gunasekera, P.; Malavige, G.N. Sensitization to Bovine Serum Albumin as a Possible Cause of Allergic Reactions to Vaccines. Vaccine 2017, 35, 1494–1500. [Google Scholar] [CrossRef] [PubMed]
  18. Boyce, J.A.; Assa’ad, A.; Burks, A.W.; Jones, S.M.; Sampson, H.A.; Wood, R.A.; Plaut, M.; Cooper, S.F.; Fenton, M.J.; Arshad, S.H.; et al. Guidelines for the Diagnosis and Management of Food Allergy in the United States: Summary of the NIAID-Sponsored Expert Panel Report. Journal of Pediatric Nursing 2011, 26, e2–e17. [Google Scholar] [CrossRef]
  19. Finkelman, F.D.; Boyce, J.A.; Vercelli, D.; Rothenberg, M.E. Key Advances in Mechanisms of Asthma, Allergy, and Immunology in 2009. Journal of Allergy and Clinical Immunology 2010, 125, 312–318. [Google Scholar] [CrossRef]
  20. Silverstein, A.M. Clemens Freiherr von Pirquet: Explaining Immune Complex Disease in 1906. Nat Immunol 2000, 1, 453–455. [Google Scholar] [CrossRef]
  21. Turk, J.L. Von Pirquet, Allergy and Infectious Diseases: A Review. J R Soc Med 1987, 80, 31–33. [Google Scholar] [CrossRef]
  22. Palmiere, C.; Comment, L.; Mangin, P. Allergic Reactions Following Contrast Material Administration: Nomenclature, Classification, and Mechanisms. Int J Legal Med 2014, 128, 95–103. [Google Scholar] [CrossRef]
  23. Kagan, R.S. Food Allergy: An Overview. Environmental Health Perspectives 2003, 111, 223–225. [Google Scholar] [CrossRef]
  24. Molina, M.A.F.; Kram, Y.E.; Lanser, B.J. The Global Burden of Food Allergy. Immunology and Allergy Clinics 2025, 45, 325–337. [Google Scholar] [CrossRef]
  25. Stapel, S.O.; Kleine-Tebbe, J. Allergy Testing in the Laboratory. In Allergy and Allergic Diseases; John Wiley & Sons, Ltd., 2008; pp. 1346–1367. ISBN 978-1-4443-0091-8. [Google Scholar]
  26. Schuppan, D.; Junker, Y.; Barisani, D. Celiac Disease: From Pathogenesis to Novel Therapies. Gastroenterology 2009, 137, 1912–1933. [Google Scholar] [CrossRef]
  27. Caubet, J.-C.; Ponvert, C. Vaccine Allergy. Immunology and Allergy Clinics 2014, 34, 597–613. [Google Scholar] [CrossRef] [PubMed]
  28. Junker, Y.; Zeissig, S.; Kim, S.-J.; Barisani, D.; Wieser, H.; Leffler, D.A.; Zevallos, V.; Libermann, T.A.; Dillon, S.; Freitag, T.L.; et al. Wheat Amylase Trypsin Inhibitors Drive Intestinal Inflammation via Activation of Toll-like Receptor 4. J Exp Med 2012, 209, 2395–2408. [Google Scholar] [CrossRef] [PubMed]
  29. Catassi, C.; Bai, J.C.; Bonaz, B.; Bouma, G.; Calabrò, A.; Carroccio, A.; Castillejo, G.; Ciacci, C.; Cristofori, F.; Dolinsek, J.; et al. Non-Celiac Gluten Sensitivity: The New Frontier of Gluten Related Disorders. Nutrients 2013, 5, 3839–3853. [Google Scholar] [CrossRef]
  30. Ruiter, B.; Shreffler, W.G. The Role of Dendritic Cells in Food Allergy. Journal of Allergy and Clinical Immunology 2012, 129, 921–928. [Google Scholar] [CrossRef]
  31. Badina, L.; Barbi, E.; Berti, I.; Radillo, O.; Matarazzo, L.; Ventura, A.; Longo, G. The Dietary Paradox in Food Allergy: Yesterday’s Mistakes, Today’s Evidence and Lessons for Tomorrow. Current Pharmaceutical Design 2012, 18, 5782–5787. [Google Scholar] [CrossRef]
  32. Burney, P.; Jarvis, D.; Perez-Padilla, R. The Global Burden of Chronic Respiratory Disease in Adults. The International Journal of Tuberculosis and Lung Disease 2015, 19, 10–20. [Google Scholar] [CrossRef]
  33. Baumann, S.; Lorentz, A. Obesity - A Promoter of Allergy? Int Arch Allergy Immunol 2013, 162, 205–213. [Google Scholar] [CrossRef] [PubMed]
  34. Gray, C.L.; Levin, M.E. Epidemiology of Food Allergy : Review Article. Current Allergy & Clinical Immunology 2014, 27, 170–176. [Google Scholar]
  35. Marrs, T.; Bruce, K.D.; Logan, K.; Rivett, D.W.; Perkin, M.R.; Lack, G.; Flohr, C. Is There an Association between Microbial Exposure and Food Allergy? A Systematic Review. Pediatric Allergy and Immunology 2013, 24, 311–320.e8. [Google Scholar] [CrossRef]
  36. Martino, D.; Neeland, M.; Dang, T.; Cobb, J.; Ellis, J.; Barnett, A.; Tang, M.; Vuillermin, P.; Allen, K.; Saffery, R. Epigenetic Dysregulation of Naive CD4+ T-Cell Activation Genes in Childhood Food Allergy. Nat Commun 2018, 9, 3308. [Google Scholar] [CrossRef]
  37. Strachan, D.P. Hay Fever, Hygiene, and Household Size. BMJ 1989, 299, 1259–1260. [Google Scholar] [CrossRef]
  38. Alm, B.; Åberg, N.; Erdes, L.; Möllborg, P.; Pettersson, R.; Norvenius, S.G.; Goksör, E.; Wennergren, G. Early Introduction of Fish Decreases the Risk of Eczema in Infants. Archives of Disease in Childhood 2009, 94, 11–15. [Google Scholar] [CrossRef]
  39. McGeady, S.J. Immunocompetence and Allergy. Pediatrics 2004, 113, 1107–1113. [Google Scholar] [CrossRef]
  40. Thorburn, A.N.; Macia, L.; Mackay, C.R. Diet, Metabolites, and “Western-Lifestyle” Inflammatory Diseases. Immunity 2014, 40, 833–842. [Google Scholar] [CrossRef] [PubMed]
  41. Valenta, R. The Future of Antigen-Specific Immunotherapy of Allergy. Nat Rev Immunol 2002, 2, 446–453. [Google Scholar] [CrossRef] [PubMed]
  42. Han, Y.; Kim, J.; Ahn, K. Food Allergy. Korean J Pediatr 2012, 55, 153–158. [Google Scholar] [CrossRef]
  43. Ebner, C. Allergene. In Allergologie; Heppt, W., Renz, H., Röcken, M., Eds.; Springer: Berlin, Heidelberg, 1998; pp. 89–99. ISBN 978-3-662-05660-8. [Google Scholar]
  44. Ebner, C.; Hoffmann-Sommergruber, K.; Breiteneder, H. Plant Food Allergens Homologous to Pathogenesis-Related Proteins. Allergy 2001, 56, 43–44. [Google Scholar] [CrossRef]
  45. Ortolani, C.; Ispano, M.; Ansaloni, R.; Rotondo, F.; Incorvaia, C.; Pastorello, E.A. Diagnostic Problems Due to Cross-reactions in Food Allergy. Allergy 1998, 53, 58–61. [Google Scholar] [CrossRef]
  46. Ortolani, C.; Pastorello, E.A. Food Allergies and Food Intolerances. Best Practice & Research Clinical Gastroenterology 2006, 20, 467–483. [Google Scholar] [CrossRef]
  47. Asero, R.; Antonicelli, L. Does Sensitization to Foods in Adults Occur Always in the Gut? Int Arch Allergy Immunol 2011, 154, 6–14. [Google Scholar] [CrossRef] [PubMed]
  48. Bartnikas, L.M.; Phipatanakul, W. Turning Up the Heat on Skin Testing for Baked Egg Allergy. Clin Exp Allergy 2013, 43, 1095–1096. [Google Scholar] [CrossRef] [PubMed]
  49. Noti, M.; Kim, B.S.; Siracusa, M.C.; Rak, G.D.; Kubo, M.; Moghaddam, A.E.; Sattentau, Q.A.; Comeau, M.R.; Spergel, J.M.; Artis, D. Exposure to Food Allergens through Inflamed Skin Promotes Intestinal Food Allergy through the Thymic Stromal Lymphopoietin–Basophil Axis. Journal of Allergy and Clinical Immunology 2014, 133, 1390–1399.e6. [Google Scholar] [CrossRef]
  50. Astwood, J.D.; Leach, J.N.; Fuchs, R.L. Stability of Food Allergens to Digestion in Vitro. Nat Biotechnol 1996, 14, 1269–1273. [Google Scholar] [CrossRef] [PubMed]
  51. Groschwitz, K.R.; Hogan, S.P. Intestinal Barrier Function: Molecular Regulation and Disease Pathogenesis. Journal of Allergy and Clinical Immunology 2009, 124, 3–20. [Google Scholar] [CrossRef]
  52. Irvine, A.D.; McLean, W.H.I.; Leung, D.Y.M. Filaggrin Mutations Associated with Skin and Allergic Diseases. New England Journal of Medicine 2011, 365, 1315–1327. [Google Scholar] [CrossRef]
  53. Perrier, C.; Corthésy, B. Gut Permeability and Food Allergies. Clinical & Experimental Allergy 2011, 41, 20–28. [Google Scholar] [CrossRef]
  54. Karlsson, M.R.; Johansen, F.-E.; Kahu, H.; Macpherson, A.; Brandtzaeg, P. Hypersensitivity and Oral Tolerance in the Absence of a Secretory Immune System. Allergy 2010, 65, 561–570. [Google Scholar] [CrossRef]
  55. Thyssen, J.P.; Linneberg, A.; Menné, T.; Johansen, J.D. The Epidemiology of Contact Allergy in the General Population – Prevalence and Main Findings. Contact Dermatitis 2007, 57, 287–299. [Google Scholar] [CrossRef]
  56. Dodo, H.W.; Konan, K.N.; Chen, F.C.; Egnin, M.; Viquez, O.M. Alleviating Peanut Allergy Using Genetic Engineering: The Silencing of the Immunodominant Allergen Ara h 2 Leads to Its Significant Reduction and a Decrease in Peanut Allergenicity. Plant Biotechnology Journal 2008, 6, 135–145. [Google Scholar] [CrossRef]
  57. Mills, E.N.C.; Sancho, A.I.; Rigby, N.M.; Jenkins, J.A.; Mackie, A.R. Impact of Food Processing on the Structural and Allergenic Properties of Food Allergens. Molecular Nutrition & Food Research 2009, 53, 963–969. [Google Scholar] [CrossRef]
  58. Rahaman, T.; Vasiljevic, T.; Ramchandran, L. Effect of Processing on Conformational Changes of Food Proteins Related to Allergenicity. Trends in Food Science & Technology 2016, 49, 24–34. [Google Scholar] [CrossRef]
  59. Sen, M.; Kopper, R.; Pons, L.; Abraham, E.C.; Burks, A.W.; Bannon, G.A. Protein Structure Plays a Critical Role in Peanut Allergen Stability and May Determine Immunodominant IgE-Binding Epitopes. J Immunol 2002, 169, 882–887. [Google Scholar] [CrossRef] [PubMed]
  60. Taylor, S.L. Food Allergies and Other Food Sensitivities. 2001, 55.
  61. Crevel, R.W.R. 3 - Food Allergen Risk Assessment and Management. In Handbook of Food Allergen Detection and Control; Flanagan, S., Ed.; Woodhead Publishing Series in Food Science, Technology and Nutrition; Woodhead Publishing, 2015; pp. 41–66. ISBN 978-1-78242-012-5. [Google Scholar]
  62. Crevel, R.W.R.; Baumert, J.L.; Baka, A.; Houben, G.F.; Knulst, A.C.; Kruizinga, A.G.; Luccioli, S.; Taylor, S.L.; Madsen, C.B. Development and Evolution of Risk Assessment for Food Allergens. Food and Chemical Toxicology 2014, 67, 262–276. [Google Scholar] [CrossRef]
  63. Crevel, R.W.R.; Baumert, J.L.; Luccioli, S.; Baka, A.; Hattersley, S.; Hourihane, J.O.; Ronsmans, S.; Timmermans, F.; Ward, R.; Chung, Y. Translating Reference Doses into Allergen Management Practice: Challenges for Stakeholders. Food and Chemical Toxicology 2014, 67, 277–287. [Google Scholar] [CrossRef] [PubMed]
  64. Petersen, K.D.; Kronborg, C.; Larsen, J.N.; Dahl, R.; Gyrd-Hansen, D. Patient Related Outcomes in a Real Life Prospective Follow up Study: Allergen Immunotherapy Increase Quality of Life and Reduce Sick Days. World Allergy Organ J 2013, 6, 1–9. [Google Scholar] [CrossRef]
  65. Arora, R.; Kumar, A.; Singh, I.K.; Singh, A. Pathogenesis Related Proteins: A Defensin for Plants but an Allergen for Humans. International Journal of Biological Macromolecules 2020, 157, 659–672. [Google Scholar] [CrossRef]
  66. Basant, N.; Jain, K.; Mittal, N.; Pandey, S.; Srivastava, V.; Pandey, K.; Ayushi; Sharma, S.; Tiwari, N. Food Allergies and Their Management. In Traditional Foods; CRC Press, 2025; ISBN 978-1-00-351424-4. [Google Scholar]
  67. Cabanillas, B.; Novak, N. Effects of Daily Food Processing on Allergenicity. Critical Reviews in Food Science and Nutrition 2019, 59, 31–42. [Google Scholar] [CrossRef]
  68. Sathe, S.K.; Teuber, S.S.; Roux, K.H. Effects of Food Processing on the Stability of Food Allergens. Biotechnology Advances 2005, 23, 423–429. [Google Scholar] [CrossRef]
  69. Sathe, S.K.; Sharma, G.M. Effects of Food Processing on Food Allergens. Molecular Nutrition & Food Research 2009, 53, 970–978. [Google Scholar] [CrossRef]
  70. Kroghsbo, S.; Bøgh, K.L.; Rigby, N.M.; Mills, E.N.C.; Rogers, A.; Madsen, C.B. Sensitization with 7S Globulins from Peanut, Hazelnut, Soy or Pea Induces IgE with Different Biological Activities Which Are Modified by Soy Tolerance. Int Arch Allergy Immunol 2011, 155, 212–224. [Google Scholar] [CrossRef] [PubMed]
  71. Madsen, J.L.; Kroghsbo, S.; Madsen, C.B.; Pozdnyakova, I.; Barkholt, V.; Bøgh, K.L. The Impact of Structural Integrity and Route of Administration on the Antibody Specificity against Three Cow’s Milk Allergens - a Study in Brown Norway Rats. Clin Transl Allergy 2014, 4, 25. [Google Scholar] [CrossRef] [PubMed]
  72. An, S.B.; Yang, B.-G.; Jang, G.; Kim, D.-Y.; Kim, J.; Oh, S.-M.; Oh, N.; Lee, S.; Moon, J.-Y.; Kim, J.-A.; et al. Combined IgE Neutralization and Bifidobacterium Longum Supplementation Reduces the Allergic Response in Models of Food Allergy. Nat Commun 2022, 13, 5669. [Google Scholar] [CrossRef]
  73. Brown, C.N.; Dupre, R.A.; Ebmeier, C.C.; Patil, S.; Smith, B.; Mattison, C.P. Heating Differentiates Pecan Allergen Stability: Car i 4 Is More Heat Labile Than Car i 1 and Car i 2. Food Science & Nutrition 2025, 13, e4747. [Google Scholar] [CrossRef] [PubMed]
  74. Garcia-Calvo, E.; García-García, A.; Madrid, R.; Martin, R.; García, T. From Polyclonal Sera to Recombinant Antibodies: A Review of Immunological Detection of Gluten in Foodstuff. Foods 2021, 10, 66. [Google Scholar] [CrossRef]
  75. Spies, J.R. Allergens. Available online: https://pubs.acs.org/doi/pdf/10.1021/jf60191a005 (accessed on 1 August 2025).
  76. Malanin, K.; Lundberg, M.; Johansson, S.G.O. Anaphylactic Reaction Caused by Neoallergens in Heated Pecan Nut. Allergy 1995, 50, 988–991. [Google Scholar] [CrossRef]
  77. Gayraud, J.; Mairesse, M.; Fontaine, J.F.; Thillay, A.; Leduc, V.; Rancé, F.; Parisot, L.; Moneret-Vautrin, D.A. The Prevalence of Sensitization to Lupin Flour in France and Belgium: A Prospective Study in 5,366 Patients, by the Allergy Vigilance Network.
  78. Abbring, S.; Xiong, L.; P. Diks, M.A.; Baars, T.; Garssen, J.; Hettinga, K.; van Esch, B.C.A.M. Loss of Allergy-Protective Capacity of Raw Cow’s Milk after Heat Treatment Coincides with Loss of Immunologically Active Whey Proteins. Food & Function 2020, 11, 4982–4993. [Google Scholar] [CrossRef]
  79. Hefle, S.L.; Taylor, S.L. Revealing and Diagnosing Food Allergies and Intolerances. Clinical Nutrition Insight 2005, 31, 1. [Google Scholar]
  80. Ehn, B.-M.; Ekstrand, B.; Bengtsson, U.; Ahlstedt, S. Modification of IgE Binding during Heat Processing of the Cow’s Milk Allergen β-Lactoglobulin. J. Agric. Food Chem. 2004, 52, 1398–1403. [Google Scholar] [CrossRef]
  81. Mine, Y.; Yang, M. Recent Advances in the Understanding of Egg Allergens: Basic, Industrial, and Clinical Perspectives. J. Agric. Food Chem. 2008, 56, 4874–4900. [Google Scholar] [CrossRef]
  82. Maleki, S.J.; Schmitt, D.A.; Galeano, M.; Hurlburt, B.K. Comparison of the Digestibility of the Major Peanut Allergens in Thermally Processed Peanuts and in Pure Form. Foods 2014, 3, 290–303. [Google Scholar] [CrossRef]
  83. Maleki, S.J.; Sathe, S.K. The Effects of Processing Methods on Allergenic Properties of Food Proteins. In Food Allergy; John Wiley & Sons, Ltd., 2006; pp. 309–322. ISBN 978-1-68367-175-6. [Google Scholar]
  84. Verhoeckx, K.C.M.; Vissers, Y.M.; Baumert, J.L.; Faludi, R.; Feys, M.; Flanagan, S.; Herouet-Guicheney, C.; Holzhauser, T.; Shimojo, R.; van der Bolt, N.; et al. Food Processing and Allergenicity. Food and Chemical Toxicology 2015, 80, 223–240. [Google Scholar] [CrossRef]
  85. Bernhisel-Broadbent, J.; Scanlon, S.M.; Sampson, H.A. Fish Hypersensitivity: I. In Vitro and Oral Challenge Results in Fish-Allergic Patients. Journal of Allergy and Clinical Immunology 1992, 89, 730–737. [Google Scholar] [CrossRef]
  86. Alemán, A.; Sastre, J.; Quirce, S.; de las Heras, M.; Carnés, J.; Fernández-Caldas, E.; Pastor, C.; Blázquez, A.B.; Vivanco, F.; Cuesta-Herranz, J. Allergy to Kiwi: A Double-Blind, Placebo-Controlled Food Challenge Study in Patients from a Birch-Free Area. Journal of Allergy and Clinical Immunology 2004, 113, 543–550. [Google Scholar] [CrossRef]
  87. Fiocchi, A.; Bouygue, G.R.; Albarini, M.; Restani, P. Molecular Diagnosis of Cow’s Milk Allergy. Current Opinion in Allergy and Clinical Immunology 2011, 11, 216. [Google Scholar] [CrossRef]
  88. Fiocchi, A.; Pecora, V.; Petersson, C.J.; Dahdah, L.; Borres, M.P.; Amengual, M.J.; Huss-Marp, J.; Mazzina, O.; Di Girolamo, F. Sensitization Pattern to Inhalant and Food Allergens in Symptomatic Children at First Evaluation. Ital J Pediatr 2015, 41, 96. [Google Scholar] [CrossRef]
  89. Aalberse, R.C. Structural Biology of Allergens. Journal of Allergy and Clinical Immunology 2000, 106, 228–238. [Google Scholar] [CrossRef]
  90. Thomas, K.; Herouet-Guicheney, C.; Ladics, G.; Bannon, G.; Cockburn, A.; Crevel, R.; Fitzpatrick, J.; Mills, C.; Privalle, L.; Vieths, S. Evaluating the Effect of Food Processing on the Potential Human Allergenicity of Novel Proteins: International Workshop Report. Food and Chemical Toxicology 2007, 45, 1116–1122. [Google Scholar] [CrossRef]
  91. Iwan, M.; Vissers, Y.M.; Fiedorowicz, E.; Kostyra, H.; Kostyra, E.; Savelkoul, H.F.J.; Wichers, H.J. Impact of Maillard Reaction on Immunoreactivity and Allergenicity of the Hazelnut Allergen Cor a 11. J. Agric. Food Chem. 2011, 59, 7163–7171. [Google Scholar] [CrossRef]
  92. Vissers, Y.M.; Blanc, F.; Skov, P.S.; Johnson, P.E.; Rigby, N.M.; Przybylski-Nicaise, L.; Bernard, H.; Wal, J.-M.; Ballmer-Weber, B.; Zuidmeer-Jongejan, L.; et al. Effect of Heating and Glycation on the Allergenicity of 2S Albumins (Ara h 2/6) from Peanut. PLoS ONE 2011, 6, e23998. [Google Scholar] [CrossRef]
  93. Prabakaran, S.; Damodaran, S. Thermal Unfolding of β-Lactoglobulin:  Characterization of Initial Unfolding Events Responsible for Heat-Induced Aggregation. J. Agric. Food Chem. 1997, 45, 4303–4308. [Google Scholar] [CrossRef]
  94. Bindslev-Jensen, C.; Skov, P.S.; Roggen, E.L.; Hvass, P.; Brinch, D.S. Investigation on Possible Allergenicity of 19 Different Commercial Enzymes Used in the Food Industry. Food and Chemical Toxicology 2006, 44, 1909–1915. [Google Scholar] [CrossRef]
  95. Kopper, R.A.; Odum, N.J.; Sen, M.; Helm, R.M.; Stanley, J.S.; Burks, A.W. Peanut Protein Allergens: The Effect of Roasting on Solubility and Allergenicity. Int Arch Allergy Immunol 2005, 136, 16–22. [Google Scholar] [CrossRef]
  96. Matsuo, K.; Hamajima, N.; Hirai, T.; Kato, T.; Koike, K.; Inoue, M.; Takezaki, T.; Tajima, K. Aldehyde Dehydrogenase 2(ALDH2)Genotype Affects Rectal Cancer Susceptibility Due to Alcohol Consumption. Journal of Epidemiology 2002, 12, 70–76. [Google Scholar] [CrossRef]
  97. Ayuso, R.; Lehrer, S.B.; Tanaka, L.; Ibanez, M.D.; Pascual, C.; Burks, A.W.; Sussman, G.L.; Goldberg, B.; Lopez, M.; Reese, G. IgE Antibody Response to Vertebrate Meat Proteins Including Tropomyosin. Annals of Allergy, Asthma & Immunology 1999, 83, 399–405. [Google Scholar] [CrossRef]
  98. Varjonen, E.; Björkstén, F.; Savolainen, J. Stability of Cereal Allergens. Clinical & Experimental Allergy 1996, 26, 436–443. [Google Scholar] [CrossRef]
  99. Posch, A.; Chen, Z.; Dunn, M.J.; Wheeler, C.H.; Petersen, A.; Leubner-Metzger, G.; Baur, X. Latex Allergen Database. Electrophoresis 1997, 18, 2803–2810. [Google Scholar] [CrossRef]
  100. Werfel, S.J.; Cooke, S.K.; Sampson, H.A. Clinical Reactivity to Beef in Children Allergic to Cow’s Milk. J Allergy Clin Immunol 1997, 99, 293–300. [Google Scholar] [CrossRef]
  101. Gruber, P.; Vieths, S.; Wangorsch, A.; Nerkamp, J.; Hofmann, T. Maillard Reaction and Enzymatic Browning Affect the Allergenicity of Pru Av 1, the Major Allergen from Cherry (Prunus Avium). J. Agric. Food Chem. 2004, 52, 4002–4007. [Google Scholar] [CrossRef]
  102. Hansen, K.S.; Ballmer-Weber, B.K.; Lüttkopf, D.; Skov, P.S.; Wüthrich, B.; Bindslev-Jensen, C.; Vieths, S.; Poulsen, L.K. Roasted Hazelnuts – Allergenic Activity Evaluated by Double-Blind, Placebo-Controlled Food Challenge. Allergy 2003, 58, 132–138. [Google Scholar] [CrossRef]
  103. Maleki, S.J.; Viquez, O.; Jacks, T.; Dodo, H.; Champagne, E.T.; Chung, S.-Y.; Landry, S.J. The Major Peanut Allergen, Ara h 2, Functions as a Trypsin Inhibitor, and Roasting Enhances This Function. Journal of Allergy and Clinical Immunology 2003, 112, 190–195. [Google Scholar] [CrossRef]
  104. Vanga, S.K.; Singh, A.; Raghavan, V. Review of Conventional and Novel Food Processing Methods on Food Allergens. Critical Reviews in Food Science and Nutrition 2017, 57, 2077–2094. [Google Scholar] [CrossRef]
  105. Pi, X.; Zhu, L.; Liu, J.; Zhang, B. Effect of Thermal Processing on Food Allergenicity: Mechanisms, Application, Influence Factor, and Future Perspective. J. Agric. Food Chem. 2024, 72, 20225–20240. [Google Scholar] [CrossRef]
  106. Khammeethong, T.; Phiriyangkul, P.; Inson, C.; Sinthuvanich, C. Effect of Microwave Vacuum Drying and Tray Drying on the Allergenicity of Protein Allergens in Edible Cricket, Gryllus Bimaculatus. Food Control 2024, 160, 110328. [Google Scholar] [CrossRef]
  107. Fiocchi, A.; Restani, P.; Riva, E.; Mirri, G.P.; Santini, I.; Bernardo, L.; Galli, C.L. Heat Treatment Modifies the Allergenicity of Beef and Bovine Serum Albumin. Allergy 1998, 53, 798–802. [Google Scholar] [CrossRef]
  108. Campus, M. High Pressure Processing of Meat, Meat Products and Seafood. Food Eng. Rev. 2010, 2, 256–273. [Google Scholar] [CrossRef]
  109. Messens, W.; Van Camp, J.; Huyghebaert, A. The Use of High Pressure to Modify the Functionality of Food Proteins. Trends in Food Science & Technology 1997, 8, 107–112. [Google Scholar] [CrossRef]
  110. Tauscher, B. Pasteurization of Food by Hydrostatic High Pressure: Chemical Aspects. Z Lebensm Unters Forch 1995, 200, 3–13. [Google Scholar] [CrossRef]
  111. Considine, K.M.; Kelly, A.L.; Fitzgerald, G.F.; Hill, C.; Sleator, R.D. High-Pressure Processing – Effects on Microbial Food Safety and Food Quality. FEMS Microbiol Lett 2008, 281, 1–9. [Google Scholar] [CrossRef]
  112. Dhakal, S.; Liu, C.; Zhang, Y.; Roux, K.H.; Sathe, S.K.; Balasubramaniam, V.M. Effect of High Pressure Processing on the Immunoreactivity of Almond Milk. Food Research International 2014, 62, 215–222. [Google Scholar] [CrossRef]
  113. Barba, F.J.; Esteve, M.J.; Frígola, A. High Pressure Treatment Effect on Physicochemical and Nutritional Properties of Fluid Foods During Storage: A Review. Comprehensive Reviews in Food Science and Food Safety 2012, 11, 307–322. [Google Scholar] [CrossRef]
  114. Chicón, R.; Belloque, J.; Alonso, E.; López-Fandiño, R. Antibody Binding and Functional Properties of Whey Protein Hydrolysates Obtained under High Pressure. Food Hydrocolloids 2009, 23, 593–599. [Google Scholar] [CrossRef]
  115. Heremans, K. The Effects of High Pressure on Biomaterials. In Ultra High Pressure Treatments of Foods; Hendrickx, M.E.G., Knorr, D., Ludikhuyze, L., Van Loey, A., Heinz, V., Eds.; Springer US: Boston, MA, 2001; pp. 23–51. ISBN 978-1-4615-0723-9. [Google Scholar]
  116. Peñas, E.; Préstamo, G.; Luisa Baeza, M.; Martínez-Molero, M.I.; Gomez, R. Effects of Combined High Pressure and Enzymatic Treatments on the Hydrolysis and Immunoreactivity of Dairy Whey Proteins. International Dairy Journal 2006, 16, 831–839. [Google Scholar] [CrossRef]
  117. Kleber, N.; Maier, S.; Hinrichs, J. Antigenic Response of Bovine β-Lactoglobulin Influenced by Ultra-High Pressure Treatment and Temperature. Innovative Food Science & Emerging Technologies 2007, 8, 39–45. [Google Scholar] [CrossRef]
  118. Kato, T.; Katayama, E.; Matsubara, S.; Omi, Y.; Matsuda, T. Release of Allergenic Proteins from Rice Grains Induced by High Hydrostatic Pressure. J. Agric. Food Chem. 2000, 48, 3124–3129. [Google Scholar] [CrossRef]
  119. Odani, S.; Kanda, Y.; Hara, T.; Matuno, M.; Suzuki, A. Effects of High Hydrostatic Pressure Treatment on the Allergenicity and Structure of Chicken Egg White Ovomucoid. High Pressure Bioscience and Biotechnology, 2007, 1, 252–258. [Google Scholar] [CrossRef]
  120. Wang, X.-S.; Tang, C.-H.; Li, B.-S.; Yang, X.-Q.; Li, L.; Ma, C.-Y. Effects of High-Pressure Treatment on Some Physicochemical and Functional Properties of Soy Protein Isolates. Food Hydrocolloids 2008, 22, 560–567. [Google Scholar] [CrossRef]
  121. Akdemir Evrendilek, G. Chapter 14 - Pulsed Electric Field Processing: Food Pasteurization, Tissue Treatment, and Seed Disinfection. In Food Packaging and Preservation; Jaiswal, A.K., Shankar, S., Eds.; Academic Press, 2024; pp. 259–273. ISBN 978-0-323-90044-7. [Google Scholar]
  122. Barba, F.J.; Parniakov, O.; Pereira, S.A.; Wiktor, A.; Grimi, N.; Boussetta, N.; Saraiva, J.A.; Raso, J.; Martin-Belloso, O.; Witrowa-Rajchert, D.; et al. Current Applications and New Opportunities for the Use of Pulsed Electric Fields in Food Science and Industry. Food Research International 2015, 77, 773–798. [Google Scholar] [CrossRef]
  123. Chatzimitakos, T.; Athanasiadis, V.; Kalompatsios, D.; Mantiniotou, M.; Bozinou, E.; Lalas, S.I. Pulsed Electric Field Applications for the Extraction of Bioactive Compounds from Food Waste and By-Products: A Critical Review. Biomass 2023, 3, 367–401. [Google Scholar] [CrossRef]
  124. Yarmush, M.L.; Golberg, A.; Serša, G.; Kotnik, T.; Miklavčič, D. Electroporation-Based Technologies for Medicine: Principles, Applications, and Challenges. Annual Review of Biomedical Engineering 2014, 16, 295–320. [Google Scholar] [CrossRef]
  125. Evrendilek, G.A. Pulsed Electric Field Treatment for Beverage Production and Preservation; Springer International Publishing, 2017; ISBN 978-3-319-32886-7. [Google Scholar]
  126. Evrendilek, G.A. Influence of High Pressure Processing on Food Bioactives. In Retention of Bioactives in Food Processing; Jafari, S.M., Capanoglu, E., Eds.; Food Bioactive Ingredients; Springer International Publishing: Cham, 2022; pp. 467–485. ISBN 978-3-030-96885-4. [Google Scholar]
  127. Lebovka, N.; Vorobiev, E.; Chemat, F. Enhancing Extraction Processes in the Food Industry; CRC Press, 2016; ISBN 978-1-4398-4595-0. [Google Scholar]
  128. Toepfl, S.; Heinz, V.; Knorr, D. High Intensity Pulsed Electric Fields Applied for Food Preservation. Chemical Engineering and Processing: Process Intensification 2007, 46, 537–546. [Google Scholar] [CrossRef]
  129. Akdemir Evrendilek, G.; Tanriverdi, H.; Demir, I.; Uzuner, S. Shelf-Life Extension of Traditional Licorice Root “Sherbet” with a Novel Pulsed Electric Field Processing. Frontiers in Food Science and Technology 2023, 3. [Google Scholar] [CrossRef]
  130. Akdemir Evrendilek, G.; Hitit Özkan, B. Pulsed Electric Field Processing of Fruit Juices with Inactivation of Enzymes with New Inactivation Kinetic Model and Determination of Changes in Quality Parameters. Innovative Food Science & Emerging Technologies 2024, 94, 103678. [Google Scholar] [CrossRef]
  131. Van Loey, A.; Verachtert, B.; Hendrickx, M. Effects of High Electric Field Pulses on Enzymes. Trends in Food Science & Technology 2001, 12, 94–102. [Google Scholar] [CrossRef]
  132. Wei, J.-N.; Zeng, X.-A.; Tang, T.; Jiang, Z.; Liu, Y.-Y. Unfolding and Nanotube Formation of Ovalbumin Induced by Pulsed Electric Field. Innovative Food Science & Emerging Technologies 2018, 45, 249–254. [Google Scholar] [CrossRef]
  133. Zhao, W.; Yang, R. Comparative Study of Inactivation and Conformational Change of Lysozyme Induced by Pulsed Electric Fields and Heat. Eur Food Res Technol 2008, 228, 47–54. [Google Scholar] [CrossRef]
  134. Zhao, W.; Yang, R. Effect of High-Intensity Pulsed Electric Fields on the Activity, Conformation and Self-Aggregation of Pepsin. Food Chemistry 2009, 114, 777–781. [Google Scholar] [CrossRef]
  135. Han, Z.; Cai, M.; Cheng, J.-H.; Sun, D.-W. Effects of Electric Fields and Electromagnetic Wave on Food Protein Structure and Functionality: A Review. Trends in Food Science & Technology 2018, 75, 1–9. [Google Scholar] [CrossRef]
  136. Li, Y.; Chen, Z.; Mo, H. Effects of Pulsed Electric Fields on Physicochemical Properties of Soybean Protein Isolates. LWT - Food Science and Technology 2007, 40, 1167–1175. [Google Scholar] [CrossRef]
  137. Zhang, R.; Cheng, L.; Wang, L.; Guan, Z. Inactivation Effects of PEF on Horseradish Peroxidase (HRP) and Pectinesterase (PE). IEEE Transactions on Plasma Science 2006, 34, 2630–2636. [Google Scholar] [CrossRef]
  138. Johnson, P.E.; Van der Plancken, I.; Balasa, A.; Husband, F.A.; Grauwet, T.; Hendrickx, M.; Knorr, D.; Mills, E.N.C.; Mackie, A.R. High Pressure, Thermal and Pulsed Electric-Field-Induced Structural Changes in Selected Food Allergens. Molecular Nutrition & Food Research 2010, 54, 1701–1710. [Google Scholar] [CrossRef]
  139. Tobajas, A.P.; Agulló-García, A.; Cubero, J.L.; Colás, C.; Segura-Gil, I.; Sánchez, L.; Calvo, M.; Pérez, M.D. Effect of High Pressure and Pulsed Electric Field on Denaturation and Allergenicity of Pru p 3 Protein from Peach. Food Chemistry 2020, 321, 126745. [Google Scholar] [CrossRef]
  140. Yang, W.; Tu, Z.; Wang, H.; Zhang, L.; Gao, Y.; Li, X.; Tian, M. Immunogenic and Structural Properties of Ovalbumin Treated by Pulsed Electric Fields. International Journal of Food Properties 2017, 20, S3164–S3176. [Google Scholar] [CrossRef]
  141. Vanga, S.K.; Singh, A.; Raghavan, V. Effect of Thermal and Electric Field Treatment on the Conformation of Ara h 6 Peanut Protein Allergen. Innovative Food Science & Emerging Technologies 2015, 30, 79–88. [Google Scholar] [CrossRef]
  142. Vanga, S.K.; Singh, A.; Kalkan, F.; Gariepy, Y.; Orsat, V.; Raghavan, V. Effect of Thermal and High Electric Fields on Secondary Structure of Peanut Protein. International Journal of Food Properties 2016, 19, 1259–1271. [Google Scholar] [CrossRef]
  143. Paschke, A. Aspects of Food Processing and Its Effect on Allergen Structure. Molecular Nutrition & Food Research 2009, 53, 959–962. [Google Scholar] [CrossRef] [PubMed]
  144. Dong, L.; Lu, X.; Zeng, X.; Lin, S. Regulation of Ovalbumin Allergenicity and Structure-Activity Relationship Analysis Based on Pulsed Electric Field Technology. International Journal of Biological Macromolecules 2024, 261, 129695. [Google Scholar] [CrossRef] [PubMed]
  145. Shriver, S.K.; Yang, W.W. Thermal and Nonthermal Methods for Food Allergen Control. Food Eng. Rev. 2011, 3, 26–43. [Google Scholar] [CrossRef]
  146. Chung, S.-Y.; Yang, W.; Krishnamurthy, K. Effects of Pulsed UV-Light on Peanut Allergens in Extracts and Liquid Peanut Butter. Journal of Food Science 2008, 73, C400–C404. [Google Scholar] [CrossRef]
  147. Garvey, M.; Rowan, N. Pulsed UV as a Potential Surface Sanitizer in Food Production Processes to Ensure Consumer Safety. Current Opinion in Food Science 2019, 26, 65–70. [Google Scholar] [CrossRef]
  148. Gómez-lópez, V.M. Continuous UV-C Light. In Decontamination of Fresh and Minimally Processed Produce; John Wiley & Sons, Ltd., 2012; pp. 365–378. ISBN 978-1-118-22918-7. [Google Scholar]
  149. Gómez-López, V.M.; Koutchma, T.; Linden, K. Chapter 8 - Ultraviolet and Pulsed Light Processing of Fluid Foods. In Novel Thermal and Non-Thermal Technologies for Fluid Foods; Cullen, P.J., Tiwari, B.K., Valdramidis, V.P., Eds.; Academic Press: San Diego, 2012; pp. 185–223. ISBN 978-0-12-381470-8. [Google Scholar]
  150. Krishnamurthy, K.; Tewari, J.C.; Irudayaraj, J.; Demirci, A. Microscopic and Spectroscopic Evaluation of Inactivation of Staphylococcus Aureus by Pulsed UV Light and Infrared Heating. Food Bioprocess Technol 2010, 3, 93–104. [Google Scholar] [CrossRef]
  151. Gorman, S.; McGlade, J.P.; Lambert, M.J.M.; Strickland, D.H.; Thomas, J.A.; Hart, P.H. UV Exposure and Protection against Allergic Airways Disease. Photochem Photobiol Sci 2010, 9, 571–577. [Google Scholar] [CrossRef]
  152. Keklik, N.M.; Krishnamurthy, K.; Demirci, A. 12 - Microbial Decontamination of Food by Ultraviolet (UV) and Pulsed UV Light. In Microbial Decontamination in the Food Industry; Demirci, A., Ngadi, M.O., Eds.; Woodhead Publishing Series in Food Science, Technology and Nutrition; Woodhead Publishing, 2012; pp. 344–369. ISBN 978-0-85709-085-0. [Google Scholar]
  153. Yang, W.W.; Mwakatage, N.R.; Goodrich-Schneider, R.; Krishnamurthy, K.; Rababah, T.M. Mitigation of Major Peanut Allergens by Pulsed Ultraviolet Light. Food Bioprocess Technol 2012, 5, 2728–2738. [Google Scholar] [CrossRef]
  154. Meinlschmidt, P.; Ueberham, E.; Lehmann, J.; Reineke, K.; Schlüter, O.; Schweiggert-Weisz, U.; Eisner, P. The Effects of Pulsed Ultraviolet Light, Cold Atmospheric Pressure Plasma, and Gamma-Irradiation on the Immunoreactivity of Soy Protein Isolate. Innovative Food Science & Emerging Technologies 2016, 38, 374–383. [Google Scholar] [CrossRef]
  155. Tammineedi, C.V.R.K.; Choudhary, R.; Perez-Alvarado, G.C.; Watson, D.G. Determining the Effect of UV-C, High Intensity Ultrasound and Nonthermal Atmospheric Plasma Treatments on Reducing the Allergenicity of α-Casein and Whey Proteins. LWT - Food Science and Technology 2013, 54, 35–41. [Google Scholar] [CrossRef]
  156. Lee, J.-W.; Kim, J.-H.; Yook, H.-S.; Kang, K.-O.; Lee, S.-Y.; Hwang, H.-J.; Byun, M.-W. Effects of Gamma Radiation on the Allergenic and Antigenic Properties of Milk Proteins. Journal of Food Protection 2001, 64, 272–276. [Google Scholar] [CrossRef]
  157. Manzocco, L.; Panozzo, A.; Nicoli, M.C. Effect of Ultraviolet Processing on Selected Properties of Egg White. Food Chemistry 2012, 135, 522–527. [Google Scholar] [CrossRef] [PubMed]
  158. Surowsky, B.; Schlüter, O.; Knorr, D. Interactions of Non-Thermal Atmospheric Pressure Plasma with Solid and Liquid Food Systems: A Review. Food Eng Rev 2015, 7, 82–108. [Google Scholar] [CrossRef]
  159. Meza-Morales, W.; Estévez-Carmona, M.M.; Alvarez-Ricardo, Y.; Obregón-Mendoza, M.A.; Cassani, J.; Ramírez-Apan, M.T.; Escobedo-Martínez, C.; Soriano-García, M.; Reynolds, W.F.; Enríquez, R.G. Full Structural Characterization of Homoleptic Complexes of Diacetylcurcumin with Mg, Zn, Cu, and Mn: Cisplatin-Level Cytotoxicity in Vitro with Minimal Acute Toxicity in Vivo. Molecules 2019, 24, 1598. [Google Scholar] [CrossRef]
  160. Shriver, S.; Yang, W.; Chung, S.-Y.; Percival, S. Pulsed Ultraviolet Light Reduces Immunoglobulin E Binding to Atlantic White Shrimp (Litopenaeus Setiferus) Extract. International Journal of Environmental Research and Public Health 2011, 8, 2569–2583. [Google Scholar] [CrossRef]
  161. Jiménez-Saiz, R.; Benedé, S.; Molina, E.; López-Expósito, I. Effect of Processing Technologies on the Allergenicity of Food Products. Critical Reviews in Food Science and Nutrition 2015, 55, 1902–1917. [Google Scholar] [CrossRef]
  162. Uesugi, A.R.; Danyluk, M.D.; Harris, L.J. Survival of Salmonella Enteritidis Phage Type 30 on Inoculated Almonds Stored at −20, 4, 23, and 35°C. Journal of Food Protection 2006, 69, 1851–1857. [Google Scholar] [CrossRef]
  163. Guerrero-Beltrán, J.; Welti-Chanes, J.; Barbosa-Cánovas, G.V. Ultraviolet-C light processing of grape, cranberry and grapefruit juices to inactivate Saccharomyces cerevisiae. Journal of Food Process Engineering 2009, 32, 916–916. [Google Scholar] [CrossRef]
  164. Pathak, B.; Omre, P.K.; Bisht, B.; Saini, D. Effect of thermal and non-thermal processing methods on food allergens.
  165. Tolouie, H.; Mohammadifar, M.A.; Ghomi, H.; Hashemi, M. Cold Atmospheric Plasma Manipulation of Proteins in Food Systems. Critical Reviews in Food Science and Nutrition 2018, 58, 2583–2597. [Google Scholar] [CrossRef] [PubMed]
  166. Shriver, S.K.; Yang, W.W. Thermal and Nonthermal Methods for Food Allergen Control. Food Eng. Rev. 2011, 3, 26–43. [Google Scholar] [CrossRef]
  167. Vaz, A.F.M.; Souza, M.P.; Carneiro-da-Cunha, M.G.; Medeiros, P.L.; Melo, A.M.M.A.; Aguiar, J.S.; Silva, T.G.; Silva-Lucca, R.A.; Oliva, M.L.V.; Correia, M.T.S. Molecular Fragmentation of Wheat-Germ Agglutinin Induced by Food Irradiation Reduces Its Allergenicity in Sensitised Mice. Food Chemistry 2012, 132, 1033–1039. [Google Scholar] [CrossRef]
  168. Luo, C.; Hu, C.; Gao, J.; Li, X.; Wu, Z.; Yang, A.; Chen, H. A Potential Practical Approach to Reduce Ara h 6 Allergenicity by Gamma Irradiation. Food Chemistry 2013, 136, 1141–1147. [Google Scholar] [CrossRef] [PubMed]
  169. Oh, S.; Jang, D.-I.; Lee, J.-W.; Kim, J.-H.; Byun, M.-W.; Lee, S.-Y. Evaluation of Reduced Allergenicity of Irradiated Peanut Extract Using Splenocytes from Peanut-Sensitized Mice. Radiation Physics and Chemistry 2009, 78, 615–617. [Google Scholar] [CrossRef]
  170. Zhenxing, L.; Hong, L.; Limin, C.; Jamil, K. The Influence of Gamma Irradiation on the Allergenicity of Shrimp (Penaeus Vannamei). Journal of Food Engineering 2007, 79, 945–949. [Google Scholar] [CrossRef]
  171. Kaddouri, H.; Mimoun, S.; El-Mecherfi, K.E.; Chekroun, A.; Kheroua, O.; Saidi, D. Impact of γ-Radiation on Antigenic Properties of Cow’s Milk β-Lactoglobulin. Journal of Food Protection 2008, 71, 1270–1272. [Google Scholar] [CrossRef] [PubMed]
  172. Su, M.; Venkatachalam, M.; Teuber, S.S.; Roux, K.H.; Sathe, S.K. Impact of γ-Irradiation and Thermal Processing on the Antigenicity of Almond, Cashew Nut and Walnut Proteins. Journal of the Science of Food and Agriculture 2004, 84, 1119–1125. [Google Scholar] [CrossRef]
  173. Leszczynska, J.; Łącka, A.; Szemraj, J.; Lukamowicz, J.; Zegota, H. The Influence of Gamma Irradiation on the Immunoreactivity of Gliadin and Wheat Flour. Eur Food Res Technol 2003, 217, 143–147. [Google Scholar] [CrossRef]
  174. Seo, J.-H.; Kim, J.-H.; Lee, J.-W.; Yoo, Y.-C.; Kim, M.R.; Park, K.-S.; Byun, M.-W. Ovalbumin Modified by Gamma Irradiation Alters Its Immunological Functions and Allergic Responses. International Immunopharmacology 2007, 7, 464–472. [Google Scholar] [CrossRef]
  175. Davies, K.J.; Delsignore, M.E. Protein Damage and Degradation by Oxygen Radicals. III. Modification of Secondary and Tertiary Structure. Journal of Biological Chemistry 1987, 262, 9908–9913. [Google Scholar] [CrossRef]
  176. Byun, M.-W.; Kim, J.-H.; Lee, J.-W.; Park, J.-W.; Hong, C.-S.; Kang, I.-J. Effects of Gamma Radiation on the Conformational and Antigenic Properties of a Heat-Stable Major Allergen in Brown Shrimp. Journal of Food Protection 2000, 63, 940–944. [Google Scholar] [CrossRef]
  177. Seo, J.-H.; Lee, J.-W.; Lee, Y.-S.; Lee, S.-Y.; Kim, M.-R.; Yook, H.-S.; Byun, M.-W. Change of an Egg Allergen in a White Layer Cake Containing Gamma-Irradiated Egg White. Journal of Food Protection 2004, 67, 1725–1730. [Google Scholar] [CrossRef]
  178. Zhao, J.; Zhu, W.; Zeng, J.; Liu, Y.; Li, H.; Wang, H.; Zhang, Z.; Lin, H.; Li, Z. Insight into the Mechanism of Allergenicity Decreasing in Recombinant Sarcoplasmic Calcium-Binding Protein from Shrimp (Litopenaeus Vannamei) with Thermal Processing via Spectroscopy and Molecular Dynamics Simulation Techniques. Food Research International 2022, 157, 111427. [Google Scholar] [CrossRef] [PubMed]
  179. Kume, T.; Furuta, M.; Todoriki, S.; Uenoyama, N.; Kobayashi, Y. Status of Food Irradiation in the World. Radiation Physics and Chemistry 2009, 78, 222–226. [Google Scholar] [CrossRef]
  180. Piyasena, P.; Mohareb, E.; McKellar, R.C. Inactivation of Microbes Using Ultrasound: A Review. International Journal of Food Microbiology 2003, 87, 207–216. [Google Scholar] [CrossRef] [PubMed]
  181. Chandrapala, J.; Oliver, C.; Kentish, S.; Ashokkumar, M. Ultrasonics in Food Processing. Ultrasonics Sonochemistry 2012, 19, 975–983. [Google Scholar] [CrossRef]
  182. Soria, A.C.; Villamiel, M. Effect of Ultrasound on the Technological Properties and Bioactivity of Food: A Review. Trends in Food Science & Technology 2010, 21, 323–331. [Google Scholar] [CrossRef]
  183. Chemat, F.; Rombaut, N.; Sicaire, A.-G.; Meullemiestre, A.; Fabiano-Tixier, A.-S.; Abert-Vian, M. Ultrasound Assisted Extraction of Food and Natural Products. Mechanisms, Techniques, Combinations, Protocols and Applications. A Review. Ultrasonics Sonochemistry 2017, 34, 540–560. [Google Scholar] [CrossRef]
  184. Knorr, D.; Froehling, A.; Jaeger, H.; Reineke, K.; Schlueter, O.; Schoessler, K. Emerging Technologies in Food Processing. Annual Review of Food Science and Technology 2011, 2, 203–235. [Google Scholar] [CrossRef]
  185. Mason, T.J. Ultrasonic Cleaning: An Historical Perspective. Ultrasonics Sonochemistry 2016, 29, 519–523. [Google Scholar] [CrossRef]
  186. Zhou, C.; Sun, D.; Sun, X.; Zhu, C.; Wang, Q. Combining Ultrasound and Microwave to Improve the Yield and Quality of Single-Cell Oil from Mortierella Isabellina NTG1−121. Journal of the American Oil Chemists’ Society 2018, 95, 1535–1547. [Google Scholar] [CrossRef]
  187. Liu, X.; Guo, Y.; Wang, S.; Wang, X.; Wang, Z.; Wang, Z. Effects of Ultrasound Combined with Hydrothermal Treatment on the Quality of Brown Rice. LWT 2024, 196, 115874. [Google Scholar] [CrossRef]
  188. Zhang, Z.; Zhang, X.; Chen, W.; Zhou, P. Conformation Stability, in Vitro Digestibility and Allergenicity of Tropomyosin from Shrimp (Exopalaemon Modestus) as Affected by High Intensity Ultrasound. Food Chemistry 2018, 245, 997–1009. [Google Scholar] [CrossRef]
  189. Chen, H.-L.; Mao, H.-Y.; Cao, M.-J.; Cai, Q.-F.; Su, W.-J.; Zhang, Y.-X.; Liu, G.-M. Purification, Physicochemical and Immunological Characterization of Arginine Kinase, an Allergen of Crayfish (Procambarus Clarkii). Food and Chemical Toxicology 2013, 62, 475–484. [Google Scholar] [CrossRef]
  190. Stanic-Vucinic, D.; Stojadinovic, M.; Atanaskovic-Markovic, M.; Ognjenovic, J.; Grönlund, H.; van Hage, M.; Lantto, R.; Sancho, A.I.; Velickovic, T.C. Structural Changes and Allergenic Properties of β-Lactoglobulin upon Exposure to High-Intensity Ultrasound. Molecular Nutrition & Food Research 2012, 56, 1894–1905. [Google Scholar] [CrossRef]
  191. Barre, A.; Jacquet, G.; Sordet, C.; Culerrier, R.; Rougé, P. Homology Modelling and Conformational Analysis of IgE-Binding Epitopes of Ara h 3 and Other Legumin Allergens with a Cupin Fold from Tree Nuts. Molecular Immunology 2007, 44, 3243–3255. [Google Scholar] [CrossRef]
  192. Wu, J.; Lin, L.; Chau, F. Ultrasound-Assisted Extraction of Ginseng Saponins from Ginseng Roots and Cultured Ginseng Cells. Ultrasonics Sonochemistry 2001, 8, 347–352. [Google Scholar] [CrossRef]
  193. Wang, J.; Vanga, S.K.; McCusker, C.; Raghavan, V. A Comprehensive Review on Kiwifruit Allergy: Pathogenesis, Diagnosis, Management, and Potential Modification of Allergens Through Processing. Compr Rev Food Sci Food Saf 2019, 18, 500–513. [Google Scholar] [CrossRef] [PubMed]
  194. Dong, X.; Wang, J.; Raghavan, V. Critical Reviews and Recent Advances of Novel Non-Thermal Processing Techniques on the Modification of Food Allergens. Critical Reviews in Food Science and Nutrition 2021, 61, 196–210. [Google Scholar] [CrossRef]
  195. Jadhav, H.B.; Annapure, U.S.; Deshmukh, R.R. Non-Thermal Technologies for Food Processing. Front. Nutr. 2021, 8. [Google Scholar] [CrossRef]
  196. Li, Y.-Q.; Chen, Q.; Liu, X.-H.; Chen, Z.-X. Inactivation of Soybean Lipoxygenase in Soymilk by Pulsed Electric Fields. Food Chemistry 2008, 109, 408–414. [Google Scholar] [CrossRef] [PubMed]
  197. Li, Y.; Wang, J.; Zeng, Q.-H.; Wang, L.; Wang, J.J.; Li, S.; Zhu, J.; Zeng, X.-A. Novel Thawing Method of Ultrasound-Assisted Slightly Basic Electrolyzed Water Improves the Processing Quality of Frozen Shrimp Compared with Traditional Thawing Approaches. Ultrasonics Sonochemistry 2024, 107, 106931. [Google Scholar] [CrossRef] [PubMed]
  198. Sun, Y.; Zhang, M.; Bhandari, B.; Yang, C. Ultrasound Treatment of Frozen Crayfish with Chitosan Nano-Composite Water-Retaining Agent: Influence on Cryopreservation and Storage Qualities. Food Research International 2019, 126, 108670. [Google Scholar] [CrossRef]
  199. Ekezie, F.-G.C.; Cheng, J.-H.; Sun, D.-W. Effects of Atmospheric Pressure Plasma Jet on the Conformation and Physicochemical Properties of Myofibrillar Proteins from King Prawn (Litopenaeus Vannamei). Food Chemistry 2019, 276, 147–156. [Google Scholar] [CrossRef]
  200. Venkataratnam, H.; Cahill, O.; Sarangapani, C.; Cullen, P.J.; Barry-Ryan, C. Impact of Cold Plasma Processing on Major Peanut Allergens. Sci Rep 2020, 10, 17038. [Google Scholar] [CrossRef] [PubMed]
  201. Nooji, J.K. 2011. Reduction of wheat allergen potency by pulsed ultraviolet light, high hydrostatic pressure, and non-thermal plasma (Doctoral dissertation, University of Florida).
  202. Dasan, B.G.; Boyaci, I.H. Effect of Cold Atmospheric Plasma on Inactivation of Escherichia Coli and Physicochemical Properties of Apple, Orange, Tomato Juices, and Sour Cherry Nectar. Food Bioprocess Technol 2018, 11, 334–343. [Google Scholar] [CrossRef]
  203. Pankaj, S.K.; Wan, Z.; Keener, K.M. Effects of Cold Plasma on Food Quality: A Review. Foods 2018, 7, 4. [Google Scholar] [CrossRef] [PubMed]
  204. Pankaj, S.K.; Keener, K.M. Cold Plasma: Background, Applications and Current Trends. Current Opinion in Food Science 2017, 16, 49–52. [Google Scholar] [CrossRef]
  205. Taylor, S.L.; Hefle, S.L. Will Genetically Modified Foods Be Allergenic? Journal of Allergy and Clinical Immunology 2001, 107, 765–771. [Google Scholar] [CrossRef]
  206. Rupa, P.; Nakamura, S.; Katayama, S.; Mine, Y. Attenuation of Allergic Immune Response Phenotype by Mannosylated Egg White in Orally Induced Allergy in Balb/c Mice. J. Agric. Food Chem. 2014, 62, 9479–9487. [Google Scholar] [CrossRef]
  207. Chu, Y.; Faustinelli, P.; Ramos, M.L.; Hajduch, M.; Stevenson, S.; Thelen, J.J.; Maleki, S.J.; Cheng, H.; Ozias-Akins, P. Reduction of IgE Binding and Nonpromotion of Aspergillus Flavus Fungal Growth by Simultaneously Silencing Ara h 2 and Ara h 6 in Peanut. J. Agric. Food Chem. 2008, 56, 11225–11233. [Google Scholar] [CrossRef]
  208. Herman, E.M.; Helm, R.M.; Jung, R.; Kinney, A.J. Genetic Modification Removes an Immunodominant Allergen from Soybean. Plant Physiol 2003, 132, 36–43. [Google Scholar] [CrossRef]
Figure 1. Grouping of food allergies (adapted from [169]).
Figure 1. Grouping of food allergies (adapted from [169]).
Preprints 171975 g001
Table 1. Influence of thermal processing on various food allergen types (adapted from [57]).
Table 1. Influence of thermal processing on various food allergen types (adapted from [57]).
Allergen Type Response to Thermal Processing
Proteins homologous to Bet v 1, such as Mal d 1 in apples and Pru av 1 in cherries Highly sensitive to heat; prone to unfolding. Susceptible to chemical changes such as Maillard reaction products in high-sugar foods and interactions with polyphenols, leading to decreased allergenic potential.
Proteins belonging to the Prolamin superfamily, including non-specific lipid transfer proteins (nsLTPs), 2S albumins such as Mal d 3, as well as tropomyosin and parvalbumin Moderately heat stable; proteins unfold to a limited extent but tend to regain their structure upon cooling. Maillard reactions may still enhance allergenic potential.
Proteins from the Cupin family, such as Ara h 1 found in peanuts, and lipocalins including β-lactoglobulin and α-lactalbumin present in milk Partially resistant to denaturation; undergo partial unfolding and tend to aggregate. Can form structural networks (e.g., in emulsions or gels). Heat can also lead to Maillard reactions, which may increase allergenicity.
Flexible proteins (e.g., caseins in milk, gluten storage proteins in wheat, ovomucoid in egg) Structurally dynamic and heat-resistant; maintain mobility and do not exhibit classic denaturation behavior under thermal conditions. Their allergenicity remains largely unchanged.
Table 2. Overview of nonthermal methods for allergen reduction (adapted from [90,95,104,105]).
Table 2. Overview of nonthermal methods for allergen reduction (adapted from [90,95,104,105]).
Technology Mechanism of Action Limitations
High-Pressure Processing Disrupts non-covalent bonds within proteins; alters protein conformation through aggregation and gelation, affecting epitope exposure; enhances enzymatic hydrolysis. Dual effects of pressure require accurate control; combining with other treatments may be necessary to optimize results.
Pulsed Electric Fields Alters the secondary structure of allergenic proteins. Often used alongside heat treatments to improve efficacy.
Pulsed UV Light Delivers high-intensity UV pulses that cause photochemical modifications in proteins, including structural changes and epitope destruction Limited penetration depth; potential degradation of sensitive nutrients; treatment uniformity can be challenging depending on food surface characteristics.
Cold Plasma Reactive oxygen and nitrogen molecules (ROS and RNS) engage with antigens, leading to changes in protein structures. High equipment costs; limited understanding of how cold plasma mitigates food allergens; potential cytotoxic effects of treated liquids warrant further investigation.
Ultrasound Cavitation generated by ultrasonic waves breaks peptide bonds, leading to irreversible unfolding and structural disruption of allergenic proteins. Can negatively impact product color, flavor, and nutritional quality.
Gamma Irradiation Allergenic proteins absorb radiation, altering their 3D structures; free radical formation leads to water radiolysis and alteration of amino acid side groups. Effective dose levels are not clearly established; not all protein-rich foods are currently approved for irradiation in the EU; concerns remain regarding food irradiation safety.
Table 3. Impact concerning how high hydrostatic pressure influences the allergenicity of different food products.
Table 3. Impact concerning how high hydrostatic pressure influences the allergenicity of different food products.
Food Treatment parameters Major Observations Sources
Soybean 300 MPa at 40°C for 15 min HHP notably reduced allergenicity in soybean sprouts, with only an 18% decrease in essential amino acids and overall nutritional value. HHP may offer a viable method for producing low-allergen soybean sprouts. [2]
Soybean 400 MPa Treatment improved protein solubility and hydrophobicity, while decreasing β-sheet content. [3]
Soy protein isolate (infant formula) 300 MPa for 15 min Allergenicity reduced by 46.8% due to structural and interaction changes in SPI, suggesting enhanced safety in allergic individuals. [4]
Peanut 150–800 MPa at 20–80°C for 10 min; 60–180 MPa No major changes observed in allergen secondary structure under HHP. However, high-pressure microfluidization reduced Ara h 2 allergenicity by modifying its structure and increasing UV absorption and hydrophobicity. [5,6]
Tofu 300 MPa at 40°C for 15 min HHP did not change tofu protein composition but lowered intensity of some protein bands. Allergenicity remained unchanged. [2]
Rice 100–400 MPa for 10–120 min; 300 MPa for 30 min + Protease N Pressure facilitated allergen release into surrounding solution; with protease, allergens were nearly eliminated from rice grains. [7]
Almond 600 MPa at 4–70°C for 5–30 min No significant change in allergen concentration or IgE-binding capacity as per SDS-PAGE, WB, and ELISA. [8]
Whey protein isolate 200–600 MPa at 30–68°C for 10–30 min The antigenic response of β-lactoglobulin increased with rising pressure, temperature, and duration of treatment. [9]
Skim milk 200–600 MPa at 30–68°C for 10–30 min β-lactoglobulin levels rose post-treatment; thermal addition mitigated allergenicity, but HHP-treated samples retained higher β-lg than controls. [9]
Sweet whey 200–600 MPa at 30–68°C for 10–30 min β-lg antigenicity rose at moderate temperatures and pressures; higher temperatures reduced but did not eliminate antigenicity. [9]
Milk >100 MPa + chymotrypsin/trypsin for 20 min; Proteolysis of β-lg was accelerated under HHP, suggesting potential for hypoallergenic food production. [10]
Apple 400–800 MPa at 80°C for 10 min Mal d 3 allergen lost α-helical structure, becoming a random coil; immunoreactivity declined at ≥400 MPa. [6]
Apple 700 MPa at 115°C for 10 min Mal d 1 showed minimal changes at 20°C, but more pronounced at 80°C; Mal d 3 IgE reactivity dropped by ~30%. [11]
Apple 600 MPa for 5 min Mal d 1 immunoreactivity decreased by >50% with combined high pressure and heat. [11]
Apple 600 MPa for 5 min; repeated consumption Daily intake of HHP-treated apple gel for 3 weeks led to desensitization in highly allergic individuals; 90% negative skin tests post-treatment. [12]
Carrot 500 MPa at 50°C for 10 min Slight increase in β-sheet structure of Dau c 1; no change in immunoreactivity observed. [13]
Carrot juice 400–550 MPa for 3–10 min; 500 MPa at 30–50°C for 10 min HHP had no observable effect on the allergenic potential of carrot juice. [13]
Celeriac 700 MPa at 118°C for 10 min Allergenicity of Api g 1 was significantly reduced through combined pressure and thermal processing. [11]
Celery 500 MP at 50°C for 10 min Api g 1 structural changes were pressure-dependent but did not impact allergenicity. [14]
Table 4. Outcome of UVC processing on allergenity of various foods (adapted from [169]).
Table 4. Outcome of UVC processing on allergenity of various foods (adapted from [169]).
Food Allergen of interest Treatment Impact on immune reactivity Sources
Milk α-casein, α-lactalbumin UVC treatment for 15 min 25% α-casein reduction [166]
β-lactoglobulin UVC treatment for 15 min 27.7% whey fractions reduction [166]
Egg Ovalbumin UV processing with (0.61 kJ/m2 energy No effect [168]
Ovomucoid UV processing with 63.7 kJ/m2 energy No effect [168]
Shrimp Tropomyosin PUV sterilization for 4 min Reduced [170]
Peanut Ara h 1, Ara h 2, Ara h 3 PUV treatment on butter slurry for 1–3 min; peanuts in raw and roasted form for 2–6 min 67% reduction IgE binding of peanut butter slurry; 12.5 folds reduction, 100% reduction total extracts [164]
Soy Gly m5 PUV treatment (1–6 min) 100% reduction Gly m5 reduced [165]
Gly m6 PUV treatment (1–6 min) Gly m6 retained [165]
Soy extracts (e.g., glycinin, β-conglycinin) PUV treatment 2 min 20% reduction [164]
PUV treatment 4 min 40% reduction [164]
PUV treatment 6 min 50% reduction [164]
Table 5. Mechanisms by which PUV affects protein structure and allergenicity.
Table 5. Mechanisms by which PUV affects protein structure and allergenicity.
Mechanism Details Impact on Allergenicity Sources
Photo-oxidation PUV emits intense UV-C light (200–280 nm) that causes oxidative modifications, particularly in amino acids like tryptophan, tyrosine, and methionine. Oxidative damage to amino acid residues alters the structure of allergenic epitopes. [158]
Disruption of Disulfide Bonds High-energy UV pulses can break disulfide bridges that maintain protein conformation. Destabilization of tertiary structure reduces IgE-binding ability. [160]
Epitope Modification PUV alters conformational and linear epitopes through structural denaturation. IgE-binding is reduced due to loss of native allergenic regions. [68,101]
Protein Aggregation / Fragmentation UV treatment may lead to cross-linking or fragmentation, depending on exposure time and intensity. Aggregation can mask epitopes; fragmentation may eliminate allergenic potential. [101,160,161]
Surface Effects PUV has limited penetration depth (~microns); primarily impacts food surface proteins. Effective for surface allergens; limited impact on allergens embedded within the food matrix. [162,163]
Dose- and Matrix-Dependent Effects Allergen reduction is influenced by pulse energy, duration, distance, and the optical properties (color, opacity) of the food. Matrix composition may shield proteins or influence energy absorption, thus modulating allergenicity outcomes. [164]
Table 6. Effects of gamma radiation treatment on food allergens.
Table 6. Effects of gamma radiation treatment on food allergens.
Food system Radiation Dose (kGy) Impact on allergen Mechanism/Observation Sources
Egg (Ovalbumin) 10–100 Reduced allergenicity Change in molecular weight; protein aggregation and cross-linking (disulfide bonds) [174,177]
White Cake (with egg) 10–20 Reduced ovalbumin reactivity Reduction in IgE binding [174]
Shrimp (Tropomyosin) 7–10 Undetectable tropomyosin band; reduced IgE binding Protein denaturation, aggregation; turbidity and surface hydrophobicity changes [145]
Shrimp (Tropomyosin) 1–15 + heat (100°C) 5–30-fold reduction in IgE binding Synergistic effect with heat treatment [170]
Egg (Ovomucoid) 10 + heat Almost undetectable levels of ovomucoid Irradiation more effective than heat alone due to ovomucoid heat stability [156]
Tree Nuts (Almonds, Cashews, Walnuts) 1–25 ± heat Minimal change in allergenicity Allergens stable under irradiation and heat [179]
Table 7. A review of recent studies examining how ultrasound technology affects allergenicity during food processing (adapted from [194]).
Table 7. A review of recent studies examining how ultrasound technology affects allergenicity during food processing (adapted from [194]).
Food Target Allergen Treatment Immunoreactivity Sources
Soy Proteins 37 kHz, 10 min Reduced by 24% [195]
Milk α-casein 20 kHz and 500 W (10–30 min) No noticeable impact [155]
β-lactoglobulin 20 kHz and 500 W (10–30 min) No noticeable impact [195]
α-lactalbumin 20 kHz and 500 W (10–30 min) No noticeable impact [190]
Peanut Ara h1 50 Hz applied for 5 hours Reduced by 84.8% [196]
Ara h2 50 Hz applied for 5 hours Reduced by 4.88% [197]
Shrimp (boiled) Proteins 30 kHz with 800 W at 0–50 °C range for 0–30 min Reduction up to 40%–50% [197]
Shrimp (raw) Proteins 800 W with 30 kHz at 0–50 °C for 0–30 min Reduction up to 8% [197]
Crayfish Tropomyosin 100–800 W for 15 min Reduced [198]
Shrimp and allergens Multiple (including tropomyosin) 800 W and 30 Hz for 30–180 min Reduction up to 75% [170]
Crayfish Arginine kinase 200 W at 30 °C for 10–180 min No noticeable impact [189]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated