Review For reprint orders, please contact [email protected] Recent advances in the diagnosis and therapy of peanut allergy Expert Rev. Clin. Immunol. 9(6), 551–560 (2013) Saira Z Sheikh*1 and A Wesley Burks2 Division of Rheumatology, Allergy and Immunology, Department of Medicine and Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill NC, CB 7280, 3300 Thurston Building, Chapel Hill, NC 27599-7280, USA 2 Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill NC 260 MacNider Hall, CB 7220, Chapel Hill, NC 27599-7220, USA *Author for correspondence: Tel.: +1 866 827 2862 Fax: +1 919 966 1739 [email protected] 1 www.expert-reviews.com Peanut allergy is a life-threatening, IgE-mediated allergic disease. In developed countries, the prevalence rate of peanut allergy in school-aged children is reported to be in excess of 1% and continues to rise, representing a major public health concern. Peanut allergy is diagnosed on the basis of a relevant clinical history combined with results of skin-prick testing and/or peanutspecific IgE levels. A double-blind placebo-controlled oral food challenge is the gold standard for diagnosis. Currently, there is no approved treatment or disease-modifying therapy for peanut allergy. This review discusses recent advances in molecular diagnostic techniques for peanut allergy and highlights advances in peanut allergy therapeutics, discussing allergen-specific and allergen-nonspecific treatments that are currently in Phase I/II clinical trials. Keywords: allergen-nonspecific • allergen-specific • allergy • component resolved • diagnosis • diagnostics • IgE-mediated • immunotherapy • peanut • treatment Peanut allergy is a life-threatening, IgE-mediated allergic disease. The prevalence of peanut allergy in school-aged children in developed countries is estimated to be in excess of 1% and c ontinues to increase [1–12] . Compared with other food allergies, peanut allergy is less likely to be outgrown and is estimated to resolve for only 20% of children by school age [13,14] . All peanutallergic individuals are counseled about strict dietary avoidance and given self-injectable epinephrine. Despite constant parent and patient vigilance, accidental ingestions of peanut are common and are estimated to be 15–40% annually [15–18] . These accidental ingestions in peanut-allergic individuals may result in severe and fatal reactions [19–21] . The first step in taking care of peanut-allergic patients is arriving at an accurate diagnosis, which is critical in order to identify patients at risk of having an allergic reaction. Unfortunately, current diagnostic tests have limitations such as the fact that they are unable to predict the severity of an allergic reaction and that positive tests to tolerated foods can be seen [22] . There has been much discussion and debate about risk factors for development of peanut allergy. The majority of peanut-allergic children react on their first known exposure to peanut [23,24] ; however, the route by which sensitization 10.1586/ECI.13.33 occurs is unclear. Several studies have focused on the role of maternal c onsumption of allergen during pregnancy or lactation; however, interventional studies have failed to demonstrate any benefit of dietary e limination [25,26] . It has been hypothesized that peanut sensitization may occur as a c onsequence of environmental e xposure through cutaneous or inhalational routes rather than from maternal or infant allergen [23] . While peanut allergy appears to have strong heritability, its genetic basis is unknown [27] . Given that loss-of-function mutations within the filaggrin gene are associated with atopic diseases such as atopic dermatitis, it is felt that filaggrin may also be a candidate gene in the etiology of peanut allergy [27,28] . In the past few years, much effort has been dedicated to the development of more sensitive and accurate diagnostic tools for the diagnosis of peanut allergy and this review discusses some of those advances (Box 1) . There is a strong recognition of the unmet need to develop effective treatments for peanut allergy, and much progress has been made in the development of novel therapies, which are showing promise. While there is currently no approved treatment available for peanut allergy beyond allergen avoidance, it is probable that the current work in the field will soon lead to the development of a disease-modifying therapy for peanut allergy. © 2013 Expert Reviews Ltd ISSN 1744-666X 551 Review Sheikh & Burks Box 1. Overview of advances in peanut allergy diagnostics and novel therapeutic approaches. Diagnostics • Component-resolved diagnostics • Protein microarrays • IgE epitope mapping Therapeutics • Allergen-specific approaches – Oral immunotherapy – Sublingual immunotherapy – Epicutaneous therapy – Immunotherapy with modified recombinant food proteins • Allergen-nonspecific approaches – Anti-IgE monoclonal antibodies – Chinese herbal medicine (FAHF-2) FAHF-2: Food Allergy Herbal Formula 2. Diagnosis of peanut allergy Current methods of diagnosis The diagnosis of peanut allergy is typically made on the basis of clinical history combined with evidence of peanut-specific IgE, established by skin-prick testing and/or in vitro testing measuring serum peanut-specific IgE antibodies. While these are good diagnostic tools, they have a number of limitations including the fact that they are unable to predict the severity of an allergic reaction and that positive test results to tolerated foods can be seen (indicating sensitization but not clinical allergy) [22] . Sampson and colleagues have reported 95% predictive decision points of peanut-specific IgE (which can predict clinical reactivity to peanut with greater than 95% certainty). These suggest that with a peanut-specific IgE <2 kU/l (measured in the ImmunoCAP®-FEIA system [Thermo Fisher Scientific, MA, USA), a subject has an approximately 50% chance to pass an open food challenge [10,29] . However, none of these diagnostic tools have been able to achieve sufficiently high predictive values to allow any one test to serve as an accurate modality for the diagnosis of peanut allergy. For patients without a clear clinical history or laboratory evidence that points towards peanut allergy, a physician-supervised food challenge is necessary to help arrive at a definitive diagnosis [30] . A double-blind placebo-controlled oral food challenge (DBPCFC) remains the gold standard for diagnosing peanut allergy. While these are routinely conducted in the research setting, open or single-blind food challenges are appropriate for the routine clinical setting. Currently, such challenges are seldom performed in the clinic due to time and the labor-intensive nature of the procedures as well as the perceived risk for severe allergic reactions. While a DBPCFC is the gold standard for diagnosis of peanut allergy, it is important to recognize that it cannot be used to predict the severity of future allergic reactions, nor does it provide prognostic information regarding development of anaphylaxis. Current focus is on developing techniques to improve the diagnosis of peanut allergy and identifying tools that provide better prognostic information. 552 Advances in peanut allergy diagnostics Allergen component-resolved diagnostics has become the focus of attention, because it offers the potential to become a more accurate diagnostic tool than those already established. Instead of crude allergen extracts, this method utilizes purified allergen proteins, which are produced from natural allergen sources or by recombinant expression of allergen-encoding cDNA [22] . Several studies have suggested that component-resolved diagnostics could improve the specificity of peanut-allergy testing. Koppelman et al. showed that Ara h 2 was the most important allergen in predicting clinical reactivity to peanut as 26 out of 32 peanut-allergic subjects in their cohort recognized it [31] . Nicolaou et al. demonstrated that Ara h 2 was the most important predictor of clinical peanut allergy, showing that Ara h 2-specific IgE levels >0.35 kU/l resulted in 100% sensitivity and 96.1% specificity in identifying subjects with peanut allergy, correctly identifying 97.5% of peanut-allergic subjects in their cohort [32] . Other studies also show that compared with whole peanut-specific IgE levels that are currently used for in vitro testing, Ara h 2-specific IgE levels provide higher accuracy in diagnosing peanut allergy [33,34] . Currently the best method for diagnosis in an individual patient is the peanut-specific IgE; as other studies are completed, it will be interesting to see whether component testing replaces the crude allergen testing. Several studies have shown that the pattern of binding to peanut proteins may vary according to the geographic location [35–39] . Other studies suggest that component testing can help in assessing the severity of an allergic reaction. Binding to Ara h 8, without binding to Ara h 1–3, has been shown to be associated with no reaction or mild reactions [40,41] . In addition, individuals who have isolated Ara h 8 sensitization typically have lower peanut-specific IgE and are sensitized to birch pollen [39–41] . Using either purified or recombinant peanut proteins, Peeters et al. [42] , Palmer et al. [43] and Astier et al. [44] have published studies indicating increased potency of Ara h 2 [39] . However, it is important to point out that studies have not yet specifically correlated clinical severity with Ara h 2 IgE [39] . Hence, at this time, Ara h 2 cannot be used to predict the severity of a clinical reaction in p eanut-allergic individuals. Protein microarrays permit the simultaneous assessment of specific IgE to different peanut protein components. This technique requires small amounts of sera, which is an important consideration in children and may be a cost-efficient approach because it delivers results for multiple components simultaneously [32,45] . Microarray technology can potentially provide additional information, such as assessment of relative IgE antibody affinity [45,46] and the parallel determination of different antibody isotypes [45,47] . IgE-binding epitopes have recently been recognized as important factors in driving allergic reactions to peanut [32,45] . Partially digested and absorbed peanut protein may lead to IgE antibodies that recognize a greater number or a specific pattern of sequential epitopes [45] . This pattern may be indicative of clinical peanut allergy rather than asymptomatic sensitization [48] and s tudies have shown that greater IgE epitope diversity and/or higher affinity may be suggestive of the persistence and severity of peanut allergy [49,50] . Expert Rev. Clin. Immunol. 9(6), (2013) Recent advances in the diagnosis & therapy of peanut allergy In the future, combining component-resolved diagnostics, IgE epitope mapping and high-throughput microarray platforms may allow for an assay that will result in better d iagnostic capability and help identify patients at risk for persistent allergy. Several functional assays such as the basophil activation tests [51] and analysis of peanut-specific T-cell responses [52] are currently being studied. However, currently there is a lack of evidence demonstrating that these tests have diagnostic value in peanut allergy. Advances in treatment of peanut allergy: allergen‑specific approaches The concept of approaching food allergy with strategies beyond allergen avoidance is certainly not a new one and dates back more than 100 years. Despite much effort through the years, there is currently no approved treatment for peanut allergy. Although subcutaneous immunotherapy is used effectively and successfully to treat environmental allergies, this has not been a popular approach for food allergy. A few earlier studies explored the concept of subcutaneous peanut immunotherapy [53,54] . Although there was some evidence that injected peanut allergen could induce desensitization, the high rate of severe adverse reactions with subcutaneous peanut immunotherapy were considered unacceptable, and this approach was abandoned [54,55] . Thus began the pursuit for alternative approaches for treatment of peanut allergy. Peanut oral immunotherapy Oral immunotherapy (OIT) for peanut allergy in young children is one of the most studied research options because of promising results that have been seen with OIT for other foods [56–58] . During peanut OIT, doses of peanut allergen are mixed in a food vehicle and ingested by the subject in gradual incremental doses. Most OIT studies consist of an initial escalation phase that typically occurs in a closely supervised setting such as a research study center, followed by buildup and maintenance phases carried out at home. When it comes to effective outcomes, there are two important concepts to keep in mind while reviewing OIT studies. The first is the concept of desensitization in which daily consumption of food allergen is required in order to maintain protection or a desensitized state. As a result, the threshold of the amount of protein needed to induce a clinical reaction is raised, but only as long as regular ingestion is continued. The next concept is that of clinical tolerance, which can be defined as the ability to ingest food protein without allergic symptoms in the absence of daily OIT, despite prolonged periods of avoidance of food protein [59,60] . While the optimal way to measure clinical tolerance is unknown, development of clinical tolerance in research studies is tested by interruption of OIT dosing for at least 4 weeks or longer followed by a supervised oral food challenge [59,61,62] . In large part, the results of the OIT studies to date are dependent on the dose and length of treatment. In 2009, Jones et al. reported results of an open-label, multicenter uncontrolled trial of peanut OIT [63] . Even though these patients did not undergo an entry challenge, all subjects reacted at www.expert-reviews.com Review less than 50 mg of protein on the initial-day escalation indicating their clinical sensitivity. The study showed that 93% (27 out of 29) of subjects were desensitized, that is, they were able to successfully consume the entire dose of 3900 mg when they were challenged following 4–22 months on a maintenance daily dosing of 300 mg of peanut protein. In terms of adverse effects, 92% of patients experienced adverse symptoms during the initial-day escalation, 46% during the buildup period and 3.5% during home dosing. These allergic side effects were mostly mild and involved the skin and upper airways. Only two subjects received epinephrine after one home dose each. While peanut OIT was shown to be safe in this open-label cohort [64] , it must be mentioned that four subjects withdrew from the study because of allergic reactions to OIT that persisted despite dose reduction. This reinforces the fact that peanut OIT may not be tolerated by all subjects. By 6 months, immunologic changes associated with this desensitization, such as decreased skin-prick tests and basophil hyporesponsiveness, were evident [63] . By 12–18 months, peanut-specific IgE levels decreased and peanut-specific IgG4 antibody levels increased significantly. The epitope-specific nature of the responses appeared to show a shift from IgE to IgG4, so that the IgG4 was directed at the same epitopes that were binding IgE prior to initiation of peanut OIT [60] . At 12 months of OIT, numbers of CD4 +, CD25+, FoxP3 + Tregs were increased, but began to return to baseline by 18 months [60] . It is hypothesized that this transient increase in Tregs is what may drive the suppression of the Th2 response to peanut allergens, and indeed, Th2 cytokines were shown to be decreased after peanut OIT in this study [60] . Subsequent studies have also shown that desensitization can be accomplished by peanut OIT [65,66] . Blumchen et al. reported results of an uncontrolled study of peanut OIT in 23 children, whose peanut allergy was confirmed by means of DBPCFC prior to initiating OIT with roasted peanut [66] . This study showed a highly significant increase in threshold of peanut challenge following OIT in 14 of 23 (60%) subjects who reached a maintenance dose of 500 mg of peanut. Immunologic parameters revealed a significant increase in peanut-specific serum IgG4 and a decrease in peanut-specific IL-5, IL-4 and IL-2 production by peripheral blood mononuclear cells in vitro after OIT [66] . Anagnostou et al. treated 22 peanut-allergic children whose peanut allergy was confirmed with an entry oral peanut challenge, with an open peanut OIT protocol [67] . In contrast to previous OIT studies, this protocol omitted the initial rush dose-escalation day and treatment began with gradual biweekly dose escalation until a targeted maintenance dose of 800 mg of peanut protein was reached. Nineteen subjects tolerated the maintenance dose and when challenged after approximately 30 weeks of maintenance therapy, the mean tolerated dose was increased by 1000-fold compared with baseline challenges. Sixty four percent (14 out of 22) subjects passed a 6600-mg peanut challenge with no symptoms, while four out of 22 subjects experienced mild or moderate symptoms. One patient dropped out during the dosing phase and two patients were unable to reach the 800-mg maintenance dose. In 2011, Varshney et al. published the results of the first double-blind, placebo-controlled study of peanut OIT in which 553 Review Sheikh & Burks 28 children were enrolled [68] . Sixteen of 19 subjects (84%) completed 12 months of peanut OIT treatment (maintenance dose: 4000 mg of peanut protein). This study showed that 100% of subjects reaching maintenance (16 out of 19) were able to successfully pass a 5000-mg peanut oral food challenge. Three out of 19 subjects (16%) were unable to complete the protocol (two out of 19 failed the initial dose-escalation day, and one out of 19 dropped out after the first gradual dose escalation). Overall in this study, peanut OIT was well tolerated by subjects, and no peanut OIT subjects required epinephrine treatment with dose-escalation visits or home OIT doses. Immunologic studies showed a decrease in skin prick testing, a transient increase then decrease in peanutspecific IgE and an increase in peanut-specific IgG4. In addition, peanut OIT was able to result in a decrease in Th2 cytokines (IL-5 and IL-13), with an increase in FoxP3hi :FoxP3intermediate CD4 + CD25 + T cells at the time of oral food challenge. These studies suggest that peanut OIT is a safe and effective therapy that can induce desensitization with ongoing therapy. As outlined earlier, mechanistic data from various studies (Table 1) suggest a shift in allergen-specific cytokine production from a Th2 to a Th1 profile, also pointing towards concurrent immuno modulation. While these results with desensitization are encouraging, little is known about long-term safety, efficacy and most importantly development of clinical tolerance, which would be the ultimate goal of such therapy. It is also important to keep in mind that some subjects are unable to endure the allergic side effects associated with peanut OIT, indicating that this therapy may not be appropriate for all peanut-allergic individuals. The major side effects with OIT are related to reports of approximately 10–20% of subjects across different studies who have been unable to reach the maintenance phase of OIT because of gastrointestinal symptoms, which also highlights the concerns for eosinophilic esophagitis with OIT [69–71] . Based on these concerns, it must be emphasized that peanut OIT is not yet ready for practical everyday use in the clinic setting. 8.6% of placebo doses. As with OIT, peanut SLIT was associated with decreases in skin-prick tests and basophil activation assays (Table 1) . Immunologic studies revealed a transient increase in peanut-specific IgE levels over the first 4 months, which then steadily decreased, and a significant increase in peanut-specific IgG4. Th2 cytokines (IL-5 and IL-13) were decreased in those on active treatment with peanut SLIT, and this was not observed in placebo subjects. Kulis et al. published a study exploring the mechanism of SLIT in which they showed that persistent mucosal exposure of peanut SLIT led to a rise in peanut-specific salivary IgA, further correlating this increase with results of oral food challenges in subjects (p = 0.0011) [75] . Peanut-specific salivary IgA has been thought to block antigen uptake and studies are ongoing to investigate this further. In a recently published randomized, double-blind, placebo-controlled multicenter trial of peanut SLIT with a crossover design in which 40 subjects (age: 12–37 years) were enrolled, Fleischer et al. showed that after receiving 44 weeks of peanut SLIT, 14 out of 20 (70%) subjects were able to consume tenfold more peanut protein than baseline oral food challenge (median successfully consumed dose increased from 3.5 to 496 mg) [76] , compared with three out of 20 (15%) subjects receiving placebo. After 68 weeks of SLIT, the median successfully consumed dose increased to 996 mg compared with 496 mg at the 44-week time point (p = 0.05). This study demonstrated that peanut SLIT was well tolerated, with mainly oropharyngeal symptoms as the notable adverse effects in this study. While encouraging, these results show only a modest level of desensitization with peanut SLIT, compared with results that have been demonstrated with peanut OIT. While there is evidence that peanut SLIT is safe and has a beneficial treatment effect in peanut-allergic subjects, the level of desensitization achieved may not be as robust as seen with peanut OIT. However, it may serve as a viable option in subjects who cannot tolerate peanut OIT, given the low dose of peanut allergen required in treatment with peanut SLIT. Peanut sublingual immunotherapy Sublingual immunotherapy (SLIT) involves administration of small drops of allergen extract under the tongue, which are then swallowed (or in some studies spit out). SLIT has been used effectively in Europe for a number of years for the treatment of allergic rhinitis [72,73] . The typical dose of peanut protein used in studies of peanut SLIT is approximately 1000-times less compared with peanut OIT [71] . In 2011, Kim et al. published the results of the first double-blind study of peanut SLIT in which 18 peanut-allergic children were enrolled and underwent dose escalation to a maintenance dose of 2 mg of peanut protein [74] . After 12 months of treatment, subjects on active treatment (n = 11) consumed a median of 1710 mg peanut protein during oral food challenge (although the amount tolerated by those in the active treatment varied significantly), while placebo subjects (n = 7) ingested 85 mg before having an allergic reaction. There were no dropouts from adverse events related to peanut dosing during this study and side effects were mainly oropharyngeal symptoms, observed with 11.5% of active and 554 Peanut epicutaneous therapy Epicutaneous therapy (EPIT) for peanut allergy consists of a small amount of peanut allergen delivered through a patch that is applied to the skin. EPIT has been shown to have some success in treatment of milk allergy based on results from a small pilot study [77] , with common side effects mainly being pruritus and eczema at the site of application of the patch. This type of treatment for peanut allergy has been studied in murine models [78] , and Phase II clinical trials of peanut EPIT are currently ongoing [101,102] . This therapy offers the advantage of an alternative route for delivery of peanut allergen; however, further studies are clearly needed to determine the efficacy and safety of this therapy. Immunotherapy with modified recombinant food proteins Immunotherapy with modified recombinant food proteins involves delivery of allergenic proteins that have undergone point mutations of key amino acid sequences, resulting in a lteration of IgE-binding Expert Rev. Clin. Immunol. 9(6), (2013) Recent advances in the diagnosis & therapy of peanut allergy allergenic epitopes [59] . This technique has been found to decrease/inhibit IgE a ntibodies that bind with major peanut allergens [79] . Bacterial adjuvants have been used to increase the effects of modified recombinant protein immunotherapy and this has been the focus of several studies. Li et al. used subcutaneous heat-killed Listeria monocytogenes and the modified peanut proteins (mAra h 1, 2 and 3) in a peanutsensitized mouse model and showed a marked decrease in incidence and severity of peanut-induced anaphylaxis compared with control mice [80] . This was thought to occur due to a shift from Th2 towards a Th1 profile based on decrease in IL-5 and IL-13 and increased levels of IFN-γ in the treated mice. Next, preliminary testing in mice was initiated to investigate if heat-killed Escherichia coli could serve as an effective adjuvant when combined with modified peanut proteins. Administration of heat-killed E. coli through the subcutaneous route led to skin inflammation [55] ; hence, subsequent studies focused on investigating rectal administration of heatkilled E. coli-mAra h 1, 2 and 3 in peanutsensitized mice [55,81] . Mice challenged with peanut had decreased severity of anaphylaxis, and splenocytes from these mice showed a trend towards deviation from a Th2 to a Th1 cytokine profile. Currently, a vaccine for humans, EMP-123, is in Phase I clinical trials [103] . While the prospect of development of a vaccine for peanut allergy is encouraging, additional studies are needed to investigate this further. Table 1. Immunologic changes seen with peanut oral immunotherapy and peanut sublingual immunotherapy. Study (year) Age of subjects Immunologic changes (years) Ref. Peanut OIT Jones et al. (2009) 1–16 6 months: ↓ skin prick tests [63] 6–12 months: ↑ IL-5, IL-10, IFN-γ and TNF-α 12–18 months: ↓ peanut-specific IgE, ↑ peanut-specific IgG4 12 months: ↑ numbers of CD4 +, CD25 +, FoxP3 + Tregs 18 months: ↓ numbers of CD4 +, CD25 +, FoxP3 + Tregs and return to baseline T-cell microarrays showed downregulation in apoptosis-related gene expression Blumchen et al. (2010) 3–14 ↑ peanut-specific IgG4 [66] ↓ IL-2, IL-4 and IL-5 Varshney et al. (2011) 1–16 ↓ skin prick tests [68] Transient ↑ then ↓ in peanut-specific IgE ↑ peanut-specific IgG4 ↓ IL-5 and IL-13 ↑ FoxP3hi:FoxP3intermediate CD4 + CD25 + T cells Peanut SLIT Kim et al. (2011) 1–11 ↓ skin prick test and basophil activation assays [74] Transient ↑ peanut-specific IgE levels over the first 4 months which then ↓ ↑ peanut-specific IgG4 ↓ IL-5 and IL-13 ↑: Increase; ↓: Decrease; OIT: Oral immunotherapy; SLIT: Sublingual immunotherapy. Advances in treatment of peanut allergy: allergen‑nonspecific approaches Anti-IgE monoclonal antibodies The concept of nonspecific immunomodulation for peanut allergy has been studied with humanized monoclonal murine anti-IgE IgG1 antibodies that bind to IgE with high affinity, hence preventing IgE from binding to FcεRI receptors on mast cells and basophils. Anti-IgE therapy results in decrease in free IgE, which subsequently leads to downregulation of the FcεRI receptors on the surface of mast cells and basophils [82] . In 2003, Leung et al. reported the results of a randomized, double-blind, placebo-controlled study in which 84 challengeconfirmed peanut-allergic subjects were assigned to receive therapy with either placebo or 150, 300 or 450 mg of an experimental anti-IgE drug called TNX-901 (also known as Hu-901) subcutaneously, once a month for four doses [82] . Results showed www.expert-reviews.com Review that only the highest dose of 450 mg of this drug resulted in a statistically significant improvement, increasing the reaction threshold to peanut from 178 to 2805 mg. Approximately 25% of subjects treated with the highest dose of this drug showed no change in their threshold for peanut consumption, hence suggesting that this therapy would not be able to benefit all peanut-allergic subjects. A separate randomized, double-blind, placebo-controlled Phase II trial of a different anti-IgE humanized IgG1 molecule omalizumab (Xolair®, Novartis, NJ, USA) in peanut-allergic subjects was prematurely terminated because of two serious allergic reactions that raised safety concerns during entry peanut challenge (prior to the administration of study drug) [83] . Sampson et al. recently published results from the 14 patients who completed 24 weeks of therapy followed by a second DBPCFC [83] . Preliminary data suggested that subjects receiving Xolair had a 555 Review Sheikh & Burks trend towards greater tolerated dose of peanut protein than in the placebo-treated group (p = 0.054). Given that the study was initially powered to enroll 150 subjects, it is not surprising that the results did not reach statistical significance. Anti-IgE therapy for peanut allergy clearly remains a therapy that warrants further study. If used in combination with peanut OIT, particularly as supplemental therapy during the doseescalation phase of immunotherapy, it could potentially have the advantage of decreasing allergic reactions during peanut dosing. Phase I/II studies of Xolair in combination with peanut OIT are currently ongoing [104] . Chinese herbal medicine Herbs have been used for centuries in traditional Chinese medicine to treat a variety of ailments; however, they have not been used previously for treatment of food allergies. The Food Allergy Herbal Formula (FAHF)-1 was developed using the extracts of 11 herbs with anti-inflammatory properties, which have been used for hundreds of years by practitioners of traditional Chinese medicine to treat conditions such as asthma and gastroenteritis [84] . Li et al. showed that FAHF-1 blocks peanut-induced anaphylaxis in a murine model [85] . A simplified formula called FAHF-2 consisting of nine herbs was subsequently developed and results similar to the previous study were seen regarding reduction of anaphylaxis in peanut-sensitized mice [86] . In addition, mechanistic studies showed significantly decreased levels of plasma histamine, peanut-specific IgE and Th2 cytokines (IL-4, IL-5 and IL-13) in FAHF-2 treated mice, with significantly increased levels of IFN-γ, suggesting a shift from Th2 to Th1 cytokine profile. A Phase I, randomized, double-blind, placebo-controlled, doseescalation study in 19 human subjects with peanut and tree nut allergy demonstrated that FAHF-2 was safe and well tolerated during 7 days of therapy [87] . In terms of adverse effects, one subject in the FAHF-2 group and one subject in the placebo group reported only mild gastrointestinal symptoms. In an extended Phase I study, Patil et al. showed that daily dosing of FAHF-2 was safe and tolerated well by subjects [88] . Srivastava et al. showed that in mice, a butanol-extracted version of FAHF-2 reduced the volume of the dose by approximately fivefold but was able to maintain its efficacy [89] . This would be an important application for human subjects, given that the large tablet load of FAHF-2 (ten tablets three-times a day) is a barrier to compliance and serves as a potential limitation for this therapy. Srivastava et al. recently investigated the use of FAHF-2 in a murine model of multiple food allergies, showing that FAHF-2 was able to block anaphylaxis from three food allergens (peanut, codfish and egg) after sensitized mice were treated with FAHF-2 for 7 weeks [90] . A multicenter, double-blind, placebo-controlled Phase II trial of FAHF-2 is currently ongoing [105] . The concept of using Chinese herbal medicine and the development of FAHF-2 is one of the most exciting advances in the field of food allergy. It has been found to be safe and well tolerated in humans and offers the additional advantage of potentially treating multiple food allergies with one therapy. 556 Expert commentary & five-year view The diagnosis and treatment of peanut allergy is an area that has eluded allergists for years. Millions of individuals suffer from peanut allergy, and while the prevalence of peanut allergy continues to increase, there is currently no approved treatment. The accurate diagnosis of peanut allergy remains a critical first step in order to correctly identify subjects at risk for having an allergic reaction. However, none of the current diagnostic tools have been able to achieve sufficiently high predictive values to allow any one test to serve as an accurate diagnostic modality. While DBPCFC remains the gold standard for diagnosis, these remain difficult for clinicians to conduct routinely in the office setting. Recently, several studies have suggested that component-resolved diagnostics could improve the specificity of peanut allergy testing and have shown that compared with whole peanut-specific IgE levels currently used for in vitro testing, Ara h 2-specific IgE levels provide higher accuracy in diagnosing peanut allergy. While component-resolved diagnostics might be used as a supplement to existing diagnostic tests for peanut allergy, it does not appear to be ready to replace them at this time. The new molecular diagnostic techniques that are being studied are promising, but before they can be implemented in the clinical setting, these assays will need to be studied in larger clinical trials and validated against the current gold standard. While not ready for prime time, combining component-resolved diagnostics, IgE epitope mapping and high-throughput microarray platforms may in the future allow for an assay that will result in better diagnostic capability and help identify patients at risk of persistent allergy. There is a strong recognition of the unmet need to develop effective treatments for peanut allergy and much progress has been made in the development of novel therapies. Several studies have shown the safety and efficacy of peanut OIT and demonstrated its ability to induce desensitization with ongoing therapy, with immunologic changes during peanut OIT showing a shift in allergen-specific cytokine production away from a Th2 profile and also pointing towards concurrent immunomodulation. While desensitization would be able to provide much needed protection against accidental peanut ingestion, the ultimate goal of peanut OIT would be to help subjects achieve long-term clinical and long-lasting immunologic tolerance, which still needs to be investigated. There is evidence that peanut SLIT is safe, efficacious and well tolerated and may be a viable option in peanutallergic subjects who cannot tolerate OIT, given the low dose of peanut allergen required in SLIT. However, the level of desensitization achieved may not be as robust as seen with peanut OIT. Anti-IgE therapy for peanut allergy clearly remains a therapy that warrants further study and might find a role as adjunct or supplemental therapy in combination with peanut OIT, and studies are ongoing to address this further. The application of Chinese herbal medicine and the development of FAHF-2 is perhaps one of the most exciting advances in the field. It has been found to be safe and well tolerated in humans and offers the additional advantage of potentially treating multiple food allergies with one therapy. Expert Rev. Clin. Immunol. 9(6), (2013) Recent advances in the diagnosis & therapy of peanut allergy It is important to note that many of the outlined studies for peanut allergy have limitations and this should be kept in mind when interpreting the literature. First, these studies exclude subjects with a history of previous anaphylactic reactions to peanut due to safety concerns, giving rise to the argument that individuals who possibly need these therapies the most are excluded from the outset. Including young children in whom remission of peanut allergy may occur during the study period may often confound studies. Given that many of the studies are pilot studies, the sample size is small and the lack of a placebo group makes it difficult to establish causality as a result of the therapy. While studies are currently ongoing, the development of prolonged true clinical tolerance to peanut as a result of any of these therapies has not yet been achieved; hence this remains a major clinical and research goal. This question would ideally be answered through conducting a prospective, randomized, double-blind, placebo-controlled trial that is powered to evaluate tolerance as the primary outcome. Therefore, before any of these therapies can be implemented in clinical practice, many issues need to be addressed and large, well designed, randomized, double-blind, placebo-controlled clinical trials are needed to determine risks that may be associated with ongoing therapy, optimal dosing regimens, efficacy and dosing for different age groups, ideal duration of therapy and a ppropriate patient selection for different therapies. In conclusion, several treatment modalities for peanut allergy are showing promise, in the hands of experienced investigators who are able to carry out these clinical trials in the appropriate research setting. While the initial data for these studies appear to be encouraging, it must be kept in mind that there is little Review long-term data available and many questions remain unanswered, regarding both long-term safety and efficacy. It must be emphasized that none of these therapies are ready for use in the clinic setting at this time. While there is currently no approved treatment available for peanut allergy, given the amount of work that is being done in the field and several therapies on the horizon, it is plausible that further investigation in the next 5 years will lead us closer to the development of a disease-modifying therapy for peanut allergy. It is our hope that during this process of discovery, we will gain better understanding of the pathogenesis of peanut allergy as well as immunologic changes induced by different therapies. Financial & competing interests disclosure AW Burks is on the boards of the American Academy of Allergy, Asthma and Immunology and the Food Allergy & Anaphylaxis Network; is on the Medical Advisory Board for the Food Allergy Initiative; is a study section member for the NIH Hypersensitivity, Autoimmunity and Immune-Mediated section; and serves on Merck’s US Allergy Immunotherapy Allergist Advisory Board. He also serves as a consultant for Dow AgroSciences, Dynavax Technologies Corp, ExploraMed Development, LLC, Genalyte, Hycor Biomedical, Merck, Nordic Biotech Advisors ApS and PBN Nutritionals. He is a minority stockholder of Allertein, a speaker for Mylan Specialty and has received royalties from UpToDate. AW Burks holds grants from the NIH and Wallace Research Foundation. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript. Key issues • A double-blind placebo-controlled oral food challenge is the gold standard for diagnosis of peanut allergy. • While not ready for prime time, combining component-resolved diagnostics, IgE epitope mapping and high-throughput microarray platforms may in the future allow for an assay that will result in better diagnostic capability and help identify patients at risk of persistent allergy. • Peanut oral immunotherapy (OIT) has been shown to be relatively safe and efficacious and has demonstrated its ability to induce desensitization with ongoing therapy, with immunologic parameters showing a shift in allergen-specific cytokine production away from a Th2 profile. • Peanut sublingual immunotherapy is safe and may be a viable option in peanut-allergic subjects who cannot tolerate OIT; however, the level of desensitization achieved may not be as robust as seen with peanut OIT. • Anti-IgE therapy for peanut allergy remains a therapy that warrants further study and might find a role as adjunct or supplemental therapy in combination with peanut OIT. • The application of Chinese herbal medicine and the development of Food Allergy Herbal Formula-2 is one of the most exciting advances in the field, offering the advantage of potentially treating multiple food allergies with one therapy. • There is currently no approved treatment available for peanut allergy and none of the above experimental therapies are ready for everyday use in clinical practice. Data are lacking regarding long-term safety and efficacy. References 1 Sicherer SH, Muñoz-Furlong A, Sampson HA. Prevalence of peanut and tree nut allergy in the United States determined by means of a random digit dial telephone survey: a 5-year follow-up study. J. Allergy Clin. Immunol. 112(6), 1203–1207 (2003). www.expert-reviews.com 2 3 Grundy J, Matthews S, Bateman B, Dean T, Arshad SH. Rising prevalence of allergy to peanut in children: data from 2 sequential cohorts. J. Allergy Clin. Immunol. 110(5), 784–789 (2002). Kagan RS, Joseph L, Dufresne C et al. Prevalence of peanut allergy in primary- school children in Montreal, Canada. J. Allergy Clin. Immunol. 112(6), 1223–1228 (2003). 4 Neuman-Sunshine DL, Eckman JA, Keet CA et al. The natural history of persistent peanut allergy. Ann. Allergy Asthma Immunol. 108(5), 326.e3–331.e3 (2012). 557 Review Sheikh & Burks 5 Sicherer SH, Sampson HA. Food allergy. J. Allergy Clin. Immunol. 125(2 Suppl. 2), S116–S125 (2010). 6 Hourihane JO, Aiken R, Briggs R et al. The impact of government advice to pregnant mothers regarding peanut avoidance on the prevalence of peanut allergy in United Kingdom children at school entry. J. Allergy Clin. Immunol. 119(5), 1197–1202 (2007). 7 8 Mullins RJ, Dear KB, Tang ML. Characteristics of childhood peanut allergy in the Australian Capital Territory, 1995 to 2007. J. Allergy Clin. Immunol. 123(3), 689–693 (2009). Venter C, Hasan Arshad S, Grundy J et al. Time trends in the prevalence of peanut allergy: three cohorts of children from the same geographical location in the UK. Allergy 65(1), 103–108 (2010). 17 Ewan PW, Clark AT. Efficacy of a management plan based on severity assessment in longitudinal and case-controlled studies of 747 children with nut allergy: proposal for good practice. Clin. Exp. Allergy 35(6), 751–756 (2005). 18 Fleischer DM, Perry TT, Atkins D et al. Allergic reactions to foods in preschoolaged children in a prospective observational food allergy study. Pediatrics 130(1), e25–e32 (2012). 19 Bock SA, Muñoz-Furlong A, Sampson HA. Further fatalities caused by anaphylactic reactions to food, 2001–2006. J. Allergy Clin. Immunol. 119(4), 1016–1018 (2007). 20 Bock SA, Muñoz-Furlong A, Sampson HA. Fatalities due to anaphylactic reactions to foods. J. Allergy Clin. Immunol. 107(1), 191–193 (2001). 21 Boyce JA, Assa’ad A, Burks AW et al.; NIAID-Sponsored Expert Panel. Guidelines for the Diagnosis and Management of Food Allergy in the United States: summary of the NIAID-Sponsored Expert Panel Report. J. Allergy Clin. Immunol. 126(6), 1105–1118 (2010). 9 Ben-Shoshan M, Kagan RS, Alizadehfar R et al. Is the prevalence of peanut allergy increasing? A 5-year follow-up study in children in Montreal. J. Allergy Clin. Immunol. 123(4), 783–788 (2009). 10 Sicherer SH, Muñoz-Furlong A, Godbold JH, Sampson HA. US prevalence of self-reported peanut, tree nut, and sesame allergy: 11-year follow-up. J. Allergy Clin. Immunol. 125(6), 1322–1326 (2010). 22 Sicherer SH. Epidemiology of food allergy. J. Allergy Clin. Immunol. 127(3), 594–602 (2011). Kattan JD, Wang J. Allergen component testing for food allergy: ready for prime time? Curr. Allergy Asthma Rep. 13(1), 58–63 (2013). 23 Osborne NJ, Koplin JJ, Martin PE et al.; HealthNuts Investigators. Prevalence of challenge-proven IgE-mediated food allergy using population-based sampling and predetermined challenge criteria in infants. J. Allergy Clin. Immunol. 127(3), 668.e1–676.e1 (2011). Fox AT, Sasieni P, du Toit G, Syed H, Lack G. Household peanut consumption as a risk factor for the development of peanut allergy. J. Allergy Clin. Immunol. 123(2), 417–423 (2009). 24 Sicherer SH, Burks AW, Sampson HA. Clinical features of acute allergic reactions to peanut and tree nuts in children. Pediatrics 102(1), e6 (1998). 25 Kramer MS, Kakuma R. Maternal dietary antigen avoidance during pregnancy or lactation, or both, for preventing or treating atopic disease in the child. Cochrane Database Syst. Rev. 9, CD000133 (2012). 11 12 13 14 Hourihane JO, Roberts SA, Warner JO. Resolution of peanut allergy: case-control study. BMJ 316(7140), 1271–1275 (1998). Skolnick HS, Conover-Walker MK, Koerner CB, Sampson HA, Burks W, Wood RA. The natural history of peanut allergy. J. Allergy Clin. Immunol. 107(2), 367–374 (2001). 15 Clark AT, Ewan PW. Good prognosis, clinical features, and circumstances of peanut and tree nut reactions in children treated by a specialist allergy center. J. Allergy Clin. Immunol. 122(2), 286–289 (2008). 16 Yu JW, Kagan R, Verreault N et al. Accidental ingestions in children with peanut allergy. J. Allergy Clin. Immunol. 118(2), 466–472 (2006). 558 26 27 Maslova E, Granström C, Hansen S et al. Peanut and tree nut consumption during pregnancy and allergic disease in children – should mothers decrease their intake? Longitudinal evidence from the Danish National Birth Cohort. J. Allergy Clin. Immunol. 130(3), 724–732 (2012). Miller DS, Brown MP, Howley PM, Hayball JD. Current and emerging immunotherapeutic approaches to treat and prevent peanut allergy. Expert Rev. Vaccines 11(12), 1471–1481 (2012). 28 Brown SJ, Asai Y, Cordell HJ et al. Loss-of-function variants in the filaggrin gene are a significant risk factor for peanut allergy. J. Allergy Clin. Immunol. 127(3), 661–667 (2011). 29 Sampson HA. Utility of food-specific IgE concentrations in predicting symptomatic food allergy. J. Allergy Clin. Immunol. 107(5), 891–896 (2001). 30 Burks AW. Peanut allergy. Lancet 371(9623), 1538–1546 (2008). 31 Koppelman SJ, Wensing M, Ertmann M, Knulst AC, Knol EF. Relevance of Ara h1, Ara h2 and Ara h3 in peanut-allergic patients, as determined by immunoglobulin E Western blotting, basophil-histamine release and intracutaneous testing: Ara h2 is the most important peanut allergen. Clin. Exp. Allergy 34(4), 583–590 (2004). 32 Nicolaou N, Murray C, Belgrave D, Poorafshar M, Simpson A, Custovic A. Quantification of specific IgE to whole peanut extract and peanut components in prediction of peanut allergy. J. Allergy Clin. Immunol. 127(3), 684–685 (2011). 33 Dang TD, Tang M, Choo S et al.; HealthNuts study. Increasing the accuracy of peanut allergy diagnosis by using Ara h 2. J. Allergy Clin. Immunol. 129(4), 1056–1063 (2012). 34 Osborne NJ, Koplin JJ, Martin PE et al.; HealthNuts Study Investigators. The HealthNuts population-based study of paediatric food allergy: validity, safety and acceptability. Clin. Exp. Allergy 40(10), 1516–1522 (2010). 35 Vereda A, van Hage M, Ahlstedt S et al. Peanut allergy: clinical and immunologic differences among patients from 3 different geographic regions. J. Allergy Clin. Immunol. 127(3), 603–607 (2011). 36 Pedrosa M, Boyano-Martínez T, García‑Ara MC, Caballero T, Quirce S. Peanut seed storage proteins are responsible for clinical reactivity in Spanish peanutallergic children. Pediatr. Allergy Immunol. 23(7), 654–659 (2012). 37 Chiang WC, Pons L, Kidon MI, Liew WK, Goh A, Wesley Burks A. Serological and clinical characteristics of children with peanut sensitization in an Asian community. Pediatr. Allergy Immunol. 21(2 Pt 2), e429–e438 (2010). 38 Lin YT, Wu CT, Cheng JH, Huang JL, Yeh KW. Patterns of sensitization to peanut allergen components in Taiwanese Preschool children. J. Microbiol. Immunol. Infect. 45(2), 90–95 (2012). Expert Rev. Clin. Immunol. 9(6), (2013) Recent advances in the diagnosis & therapy of peanut allergy 39 Sicherer S. Advances in diagnosing peanut allergy. J. Allergy Clin. Immunol.: In Practice 1(1), 1–13 (2013). 40 Movérare R, Ahlstedt S, Bengtsson U et al. Evaluation of IgE antibodies to recombinant peanut allergens in patients with reported reactions to peanut. Int. Arch. Allergy Immunol. 156(3), 282–290 (2011). 41 42 43 44 45 46 47 Asarnoj A, Nilsson C, Lidholm J et al. Peanut component Ara h 8 sensitization and tolerance to peanut. J. Allergy Clin. Immunol. 130(2), 468–472 (2012). Peeters KA, Koppelman SJ, van Hoffen E et al. Does skin prick test reactivity to purified allergens correlate with clinical severity of peanut allergy? Clin. Exp. Allergy 37(1), 108–115 (2007). Palmer GW, Dibbern DA Jr, Burks AW et al. Comparative potency of Ara h 1 and Ara h 2 in immunochemical and functional assays of allergenicity. Clin. Immunol. 115(3), 302–312 (2005). The basophil activation test in the diagnosis of allergy: technical issues and critical factors. Allergy 64(9), 1319–1326 (2009). 63 Jones SM, Pons L, Roberts JL et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J. Allergy Clin. Immunol. 124(2), 292–300.e97 (2009). 52 Flinterman AE, Pasmans SG, den Hartog Jager CF et al. T-cell responses to major peanut allergens in children with and without peanut allergy. Clin. Exp. Allergy 40(4), 590–597 (2010). 64 Hofmann AM, Scurlock AM, Jones SM et al. Safety of a peanut oral immuno therapy protocol in children with peanut allergy. J. Allergy Clin. Immunol. 124(2), 286.e1–291.e1 (2009). 53 Oppenheimer JJ, Nelson HS, Bock SA, Christensen F, Leung DY. Treatment of peanut allergy with rush immunotherapy. J. Allergy Clin. Immunol. 90(2), 256–262 (1992). 65 Clark AT, Islam S, King Y, Deighton J, Anagnostou K, Ewan PW. Successful oral tolerance induction in severe peanut allergy. Allergy 64(8), 1218–1220 (2009). 66 54 Nelson HS, Lahr J, Rule R, Bock A, Leung D. Treatment of anaphylactic sensitivity to peanuts by immunotherapy with injections of aqueous peanut extract. J. Allergy Clin. Immunol. 99(6 Pt 1), 744–751 (1997). Blumchen K, Ulbricht H, Staden U et al. Oral peanut immunotherapy in children with peanut anaphylaxis. J. Allergy Clin. Immunol. 126(1), 83.e1–91.e1 (2010). 67 Anagnostou K, Clark A, King Y, Islam S, Deighton J, Ewan P. Efficacy and safety of high-dose peanut oral immunotherapy with factors predicting outcome. Clin. Exp. Allergy 41(9), 1273–1281 (2011). 68 Varshney P, Jones SM, Scurlock AM et al. A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. J. Allergy Clin. Immunol. 127(3), 654–660 (2011). 69 Sánchez-García S, Rodríguez Del Río P, Escudero C, Martínez-Gómez MJ, Ibáñez MD. Possible eosinophilic esophagitis induced by milk oral immunotherapy. J. Allergy Clin. Immunol. 129(4), 1155–1157 (2012). 70 Varshney P, Steele PH, Vickery BP et al. Adverse reactions during peanut oral immunotherapy home dosing. J. Allergy Clin. Immunol. 124(6), 1351–1352 (2009). 71 Kim EH, Burks W. Managing food allergy in childhood. Curr. Opin. Pediatr. 24(5), 615–620 (2012). 72 Canonica GW, Bousquet J, Casale T et al. Sub-lingual immunotherapy: World Allergy Organization Position Paper 2009. Allergy 64(Suppl. 91), 1–59 (2009). 73 Passalacqua G, Lombardi C, Troise C, Canonica GW. Sublingual immuno therapy: certainties, unmet needs and future directions. Eur. Ann. Allergy Clin. Immunol. 41(6), 163–170 (2009). 74 Kim EH, Bird JA, Kulis M et al. Sub lingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J. Allergy Clin. Immunol. 127(3), 640.e1–646.e1 (2011). 75 Kulis M, Saba K, Kim EH et al. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut 55 Stahl MC, Rans TS. Potential therapies for peanut allergy. Ann. Allergy Asthma Immunol. 106(3), 179–187; quiz 188 (2011). 56 Patriarca G, Nucera E, Roncallo C et al. Oral desensitizing treatment in food allergy: clinical and immunological results. Aliment. Pharmacol. Ther. 17(3), 459–465 (2003). 57 Buchanan AD, Green TD, Jones SM et al. Egg oral immunotherapy in nonanaphylactic children with egg allergy. J. Allergy Clin. Immunol. 119(1), 199–205 (2007). Hamilton RG, Saito H. IgE antibody concentration, specific activity, clonality, and affinity measures from future diagnostic confirmatory tests. J. Allergy Clin. Immunol. 122(2), 305–306 (2008). 58 Skripak JM, Nash SD, Rowley H et al. A randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow’s milk allergy. J. Allergy Clin. Immunol. 122(6), 1154–1160 (2008). Renault NK, Gaddipati SR, Wulfert F et al. Multiple protein extract microarray for profiling human food-specific immunoglobulins A, M, G and E. J. Immunol. Methods 364(1–2), 21–32 (2011). 59 Astier C, Morisset M, Roitel O et al. Predictive value of skin prick tests using recombinant allergens for diagnosis of peanut allergy. J. Allergy Clin. Immunol. 118(1), 250–256 (2006). Caubet JC, Sampson HA. Beyond skin testing: state of the art and new horizons in food allergy diagnostic testing. Immunol. Allergy Clin. North Am. 32(1), 97–109 (2012). 60 Nowak-Wegrzyn A, Sampson HA. Future therapies for food allergies. J. Allergy Clin. Immunol. 127(3), 558–573; quiz 574 (2011). Kulis M, Vickery BP, Burks AW. Pioneering immunotherapy for food allergy: clinical outcomes and modulation of the immune response. Immunol. Res. 49(1–3), 216–226 (2011). 48 Sampson HA. Improving in vitro tests for the diagnosis of food hypersensitivity. Curr. Opin. Allergy Clin. Immunol. 2(3), 257–261 (2002). 49 Shreffler WG, Beyer K, Chu TH, Burks AW, Sampson HA. Microarray immunoassay: association of clinical history, in vitro IgE function, and heterogeneity of allergenic peanut epitopes. J. Allergy Clin. Immunol. 113(4), 776–782 (2004). 61 Staden U, Rolinck-Werninghaus C, Brewe F, Wahn U, Niggemann B, Beyer K. Specific oral tolerance induction in food allergy in children: efficacy and clinical patterns of reaction. Allergy 62(11), 1261–1269 (2007). 50 Flinterman AE, Knol EF, Lencer DA et al. Peanut epitopes for IgE and IgG4 in peanut-sensitized children in relation to severity of peanut allergy. J. Allergy Clin. Immunol. 121(3), 737.e10–743.e10 (2008). 62 51 Sturm GJ, Kranzelbinder B, Sturm EM, Heinemann A, Groselj-Strele A, Aberer W. Rolinck-Werninghaus C, Staden U, Mehl A, Hamelmann E, Beyer K, Niggemann B. Specific oral tolerance induction with food in children: transient or persistent effect on food allergy? Allergy 60(10), 1320–1322 (2005). www.expert-reviews.com Review 559 Review Sheikh & Burks sublingual immunotherapy. J. Allergy Clin. Immunol. 129(4), 1159–1162 (2012). 76 77 78 Fleischer DM, Burks AW, Vickery BP et al.; Consortium of Food Allergy Research (CoFAR). Sublingual immunotherapy for peanut allergy: a randomized, double-blind, placebo-controlled multicenter trial. J. Allergy Clin. Immunol. 131(1), 119.e1–127.e1 (2013). Dupont C, Kalach N, Soulaines P, Legoué-Morillon S, Piloquet H, Benhamou PH. Cow’s milk epicutaneous immunotherapy in children: a pilot trial of safety, acceptability, and impact on allergic reactivity. J. Allergy Clin. Immunol. 125(5), 1165–1167 (2010). Mondoulet L, Dioszeghy V, Vanoirbeek JA, Nemery B, Dupont C, Benhamou PH. Epicutaneous immunotherapy using a new epicutaneous delivery system in mice sensitized to peanuts. Int. Arch. Allergy Immunol. 154(4), 299–309 (2011). 79 Scurlock AM, Burks AW. Peanut allergenicity. Ann. Allergy Asthma Immunol. 93(5 Suppl. 3), S12–S18 (2004). 80 Li XM, Srivastava K, Huleatt JW, Bottomly K, Burks AW, Sampson HA. Engineered recombinant peanut protein and heat-killed Listeria monocytogenes coadministration protects against peanut-induced anaphylaxis in a murine model. J. Immunol. 170(6), 3289–3295 (2003). 81 Li XM, Srivastava K, Grishin A et al. Persistent protective effect of heat-killed Escherichia coli producing ‘engineered’, recombinant peanut proteins in a murine model of peanut allergy. J. Allergy Clin. Immunol. 112(1), 159–167 (2003). 560 82 83 84 85 86 87 88 Leung DY, Sampson HA, Yunginger JW et al.; Avon Longitudinal Study of Parents and Children Study Team. Effect of anti-IgE therapy in patients with peanut allergy. N. Engl. J. Med. 348(11), 986–993 (2003). Sampson HA, Leung DY, Burks AW et al. A Phase II, randomized, double-blind, parallel-group, placebo-controlled oral food challenge trial of Xolair (omalizumab) in peanut allergy. J. Allergy Clin. Immunol. 127(5), 1309.e1–1310.e1 (2011). Li XM, Brown L. Efficacy and mechanisms of action of traditional Chinese medicines for treating asthma and allergy. J. Allergy Clin. Immunol. 123(2), 297–306; quiz 307 (2009). Li XM, Zhang TF, Huang CK et al. Food Allergy Herbal Formula-1 (FAHF-1) blocks peanut-induced anaphylaxis in a murine model. J. Allergy Clin. Immunol. 108(4), 639–646 (2001). Srivastava KD, Kattan JD, Zou ZM et al. The Chinese herbal medicine formula FAHF-2 completely blocks anaphylactic reactions in a murine model of peanut allergy. J. Allergy Clin. Immunol. 115(1), 171–178 (2005). Wang J, Patil SP, Yang N et al. Safety, tolerability, and immunologic effects of a food allergy herbal formula in food allergic individuals: a randomized, double-blinded, placebo-controlled, dose escalation, Phase 1 study. Ann. Allergy Asthma Immunol. 105(1), 75–84 (2010). Patil SP, Wang J, Song Y et al. Clinical safety of Food Allergy Herbal Formula-2 (FAHF-2) and inhibitory effect on basophils from patients with food allergy: extended Phase I study. J. Allergy Clin. Immunol. 128(6), 1259.e2–1265.e2 (2011). 89 Srivastava K, Yang N, Chen Y et al. Efficacy, safety and immunological actions of butanol-extracted Food Allergy Herbal Formula-2 on peanut anaphylaxis. Clin. Exp. Allergy 41(4), 582–591 (2011). 90 Srivastava KD, Bardina L, Sampson HA, Li XM. Efficacy and immunological actions of FAHF-2 in a murine model of multiple food allergies. Ann. Allergy Asthma Immunol. 108(5), 351.e1–358.e1 (2012). Websites 101 ClinicalTrials.gov. Epicutaneous immunotherapy in peanut allergy in children (ARACHILD). www.clinicaltrials.gov/ct2/show/ NCT01197053 102 ClinicalTrials.gov. Safety of epicutaneous immunotherapy for the treatment of peanut allergy. www.clinicaltrials.gov/ct2/show/ NCT01170286 103 ClinicalTrials.gov. Peanut allergy vaccine study in healthy and peanut-allergic adults. www.clinicaltrials.gov/ct2/show/ NCT00850668 104 ClinicalTrials.gov. Peanut oral immunotherapy and anti-immunoglobulin E (IgE) for peanut allergy. www.clinicaltrials.gov/ct2/show/ NCT00932282 105 ClinicalTrials.gov. Therapeutic effect of Chinese herbal medicine on food allergy (FAHF-2). www.clinicaltrials.gov/ct2/show/ NCT00602160 Expert Rev. Clin. Immunol. 9(6), (2013)
© Copyright 2024