Full Text:  <753>

Summary:  <291>

CLC number: 

On-line Access: 2022-06-16

Received: 2022-01-26

Revision Accepted: 2022-05-10

Crosschecked: 2022-09-16

Cited: 0

Clicked: 929

Citations:  Bibtex RefMan EndNote GB/T7714

 ORCID:

Haitao ZHU

https://orcid.org/0000-0003-0867-1354

Zhongwei LIU

https://orcid.org/0000-0002-5548-5450

-   Go to

Article info.
Open peer comments

Journal of Zhejiang University SCIENCE B

Accepted manuscript available online (unedited version)


Biomarkers in oral immunotherapy


Author(s):  Haitao ZHU, Kaifa TANG, Guoqiang CHEN, Zhongwei LIU

Affiliation(s):  Department of Pediatrics (No. 3 Ward), Northwest Womens and Childrens Hospital, Xian 710061, China; more

Corresponding email(s):  medicalman@163.com

Key Words:  Food allergy; Oral immunotherapy; Biomarker; Immune tolerance; Desensitization


Share this article to: More |Next Paper >>>

Haitao ZHU, Kaifa TANG, Guoqiang CHEN, Zhongwei LIU. Biomarkers in oral immunotherapy[J]. Journal of Zhejiang University Science B,in press.Frontiers of Information Technology & Electronic Engineering,in press.https://doi.org/10.1631/jzus.B2200047

@article{title="Biomarkers in oral immunotherapy",
author="Haitao ZHU, Kaifa TANG, Guoqiang CHEN, Zhongwei LIU",
journal="Journal of Zhejiang University Science B",
year="in press",
publisher="Zhejiang University Press & Springer",
doi="https://doi.org/10.1631/jzus.B2200047"
}

%0 Journal Article
%T Biomarkers in oral immunotherapy
%A Haitao ZHU
%A Kaifa TANG
%A Guoqiang CHEN
%A Zhongwei LIU
%J Journal of Zhejiang University SCIENCE B
%P 705-731
%@ 1673-1581
%D in press
%I Zhejiang University Press & Springer
doi="https://doi.org/10.1631/jzus.B2200047"

TY - JOUR
T1 - Biomarkers in oral immunotherapy
A1 - Haitao ZHU
A1 - Kaifa TANG
A1 - Guoqiang CHEN
A1 - Zhongwei LIU
J0 - Journal of Zhejiang University Science B
SP - 705
EP - 731
%@ 1673-1581
Y1 - in press
PB - Zhejiang University Press & Springer
ER -
doi="https://doi.org/10.1631/jzus.B2200047"


Abstract: 
Food allergy (FA) is a global health problem that affects a large population, and thus effective treatment is highly desirable. Oral immunotherapy (OIT) has been showing reasonable efficacy and favorable safety in most FA subjects. Dependable biomarkers are needed for treatment assessment and outcome prediction during OIT. Several immunological indicators have been used as biomarkers in OIT, such as skin prick tests, basophil and mast cell reactivity, T cell and B cell responses, allergen-specific antibody levels, and cytokines. Other novel indicators also could be potential biomarkers. In this review, we discuss and assess the application of various immunological indicators as biomarkers for OIT.

口服免疫治疗中的生物标志物

朱海涛1,唐开发2,陈国强1,刘仲伟3
1西北妇女儿童医院儿内三科,中国西安,710061
2贵州医科大学附属医院泌尿外科,中国贵阳,550004
3陕西省人民医院心血管内科,中国西安,710068
概要:食物过敏(food allergy,FA)是影响大量人口的全球健康问题之一,因此非常需要有效的治疗。口服免疫治疗(oral immunotherapy,OIT)在大多数FA受试者中显示出良好的疗效和安全性。OIT期间需要可靠的生物标志物进行治疗评估和结果预测。多种免疫学指标已被用作OIT的生物标志物,例如皮肤点刺试验、嗜碱性粒细胞和肥大细胞反应性、T细胞和B细胞应答反应、过敏原特异性抗体水平和细胞因子等。其他新的指标也可作为潜在的生物标志物。本文讨论并评估了各种免疫相关指标作为OIT生物标志物的应用。

关键词组:食物过敏;口服免疫治疗;生物标记物;免疫耐受;脱敏

Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article

Reference

[1]AitoroR, PaparoL, AmorosoA, et al., 2017. Gut microbiota as a target for preventive and therapeutic intervention against food allergy. Nutrients, 9(7):672.

[2]AkdisM, AkdisCA, 2014. Mechanisms of allergen-specific immunotherapy: multiple suppressor factors at work in immune tolerance to allergens. J Allergy Clin Immunol, 133(3):621-631.

[3]AkdisM, AabA, AltunbulakliC, et al., 2016. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-‍‍α: receptors,functions, and roles in diseases. J Allergy Clin Immunol, 138(4):984-1010.

[4]AlhamweBA, MeulenbroekLAPM, Veening-GriffioenDH, et al., 2020. Decreased histone acetylation levels at Th1 and regulatory loci after induction of food allergy. Nutrients, 12(10):3193.

[5]AltinJA, GoodnowCC, CookMC, 2012. IL-10+CTLA-4+ Th2 inhibitory cells form in a Foxp3-independent, IL-2-dependent manner from Th2 effectors during chronic inflammation. J Immunol, 188(11):5478-5488.

[6]AmanoT, FurunoT, HirashimaN, et al., 2001. Dynamics of intracellular granules with CD63-GFP in rat basophilic leukemia cells. J Biochem, 129(5):739-744.

[7]AnagnostouK, 2018. Anaphylaxis in children: epidemiology, risk factors and management. Curr Pediatr Rev, 14(3):180-186.

[8]AnagnostouK, IslamS, KingY, et al., 2014. Assessing the efficacy of oral immunotherapy for the desensitisation of peanut allergy in children (STOP II): a phase 2 randomised controlled trial. Lancet, 383(9925):1297-1304.

[9]AngWXG, ChurchAM, KulisM, et al., 2016. Mast cell desensitization inhibits calcium flux and aberrantly remodels actin. J Clin Invest, 126(11):4103-4118.

[10]AnsoteguiIJ, MelioliG, CanonicaGW, et al., 2020. IgE allergy diagnostics and other relevant tests in allergy, a World Allergy Organization position paper. World Allergy Organ J, 13(2):100080.

[11]AtarashiK, TanoueT, ShimaT, et al., 2011. Induction of colonic regulatory T cells by indigenous Clostridium species. Science, 331(6015):337-341.

[12]AtarashiK, TanoueT, OshimaK, et al., 2013. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature, 500(7461):‍‍232-236.

[13]AttiasM, Al-AubodahT, PiccirilloCA, 2019. Mechanisms of human FoxP3+ Treg cell development and function in health and disease. Clin Exp Immunol, 197(1):36-51.

[14]AzadMB, KonyaT, GuttmanDS, et al., 2015. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy, 45(3):632-643.

[15]BaarA, PahrS, ConstantinC, et al., 2014. The high molecular weight glutenin subunit Bx7 allergen from wheat contains repetitive IgE epitopes. Allergy, 69(10):‍1316-1323.

[16]BahriR, CustovicA, KorosecP, et al., 2018. Mast cell activation test in the diagnosis of allergic disease and anaphylaxis. J Allergy Clin Immunol, 142(2):485-496.e16.

[17]BergerA, 2000. Th1 and Th2 responses: what are they? BMJ, 321(7258):424.

[18]BeyerK, Ellman-GruntherL, JarvinenKM, et al., 2003. Measurement of peptide-specific IgE as an additional tool in identifying patients with clinical reactivity to peanuts. J Allergy Clin Immunol, 112(1):202-207.

[19]BockSA, SampsonHA, AtkinsFM, et al., 1988. Double-blind, placebo-controlled food challenge (DBPCFC) as an office procedure: a manual. J Allergy Clin Immunol, 82(6):986-997.

[20]BockSA, Muñoz-FurlongA, SampsonHA, 2007. Further fatalities caused by anaphylactic reactions to food, 2001‒2006. J Allergy Clin Immunol, 119(4):‍1016-1018.

[21]BogunovicM, GinhouxF, HelftJ, et al., 2009. Origin of the lamina propria dendritic cell network. Immunity, 31(3):513-525.

[22]BoonpiyathadT, MeyerN, MoniuszkoM, et al., 2017. High-dose bee venom exposure induces similar tolerogenic B-cell responses in allergic patients and healthy beekeepers. Allergy, 72(3):407-415.

[23]BoonpiyathadT, van de VeenW, WirzO, et al., 2019. Role of Der p 1-specific B cells in immune tolerance during 2 years of house dust mite-specific immunotherapy. J Allergy Clin Immunol, 143(3):‍1077-1086.e10.

[24]BunyavanichS, ShenN, GrishinA, et al., 2016. Early-life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol, 138(4):1122-1130.

[25]BurksAW, JonesSM, WoodRA, et al., 2012. Oral immunotherapy for treatment of egg allergy in children. N Engl J Med, 367(3):233-243.

[26]BurtonOT, RivasMN, ZhouJS, et al., 2014a. Immunoglobulin E signal inhibition during allergen ingestion leads to reversal of established food allergy and induction of regulatory T cells. Immunity, 41(1):141-151.

[27]BurtonOT, LogsdonSL, ZhouJS, et al., 2014b. Oral immunotherapy induces IgG antibodies that act through FcγRIIb to suppress IgE-mediated hypersensitivity. J Allergy Clin Immunol, 134(6):1310-1317.e6.

[28]BurtonOT, TamayoJM, StranksAJ, et al., 2018. Allergen-specific IgG antibody signaling through FcγRIIb promotes food tolerance. J Allergy Clin Immunol, 141(1):189-201.e3.

[29]CalderonTE, FerreroM, MarinoGM, et al., 2010. Meat-specific IgG and IgA antibodies coexist with IgE antibodies in sera from allergic patients: clinical association and modulation by exclusion diet. J Biol Regul Homeost Agents, 24(3):261-271.

[30]CananiRB, PaparoL, NocerinoR, et al., 2019. Gut microbiome as target for innovative strategies against food allergy. Front Immunol, 10:191.

[31]CarrierY, YuanJ, KuchrooVK, et al., 2007. Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-‍βT cell-transgenic mice. J Immunol, 178(1):179-185.

[32]CassardL, JönssonF, ArnaudS, et al., 2012. Fcγ receptors inhibit mouse and human basophil activation. J Immunol, 189(6):2995-3006.

[33]ChenCY, LeeJB, LiuB, et al., 2015. Induction of interleukin-9-producing mucosal mast cells promotes susceptibility to IgE-mediated experimental food allergy. Immunity, 43(4):788-802.

[34]ChinthrajahRS, HernandezJD, BoydSD, et al., 2016. Molecular and cellular mechanisms of food allergy and food tolerance. J Allergy Clin Immunol, 137(4):984-997.

[35]ChinthrajahRS, PuringtonN, AndorfS, et al., 2019. Sustained outcomes in oral immunotherapy for peanut allergy (POISED study): a large, randomised, double-blind, placebo-controlled, phase 2 study. Lancet, 394(10207):1437-1449.

[36]ChinukiY, KanekoS, DekioI, et al., 2012. CD203c expression-based basophil activation test for diagnosis of wheat-dependent exercise-induced anaphylaxis. J Allergy Clin Immunol, 129(5):1404-1406.

[37]ComminsSP, JamesHR, StevensW, et al., 2014. Delayed clinical and ex vivo response to mammalian meat in patients with IgE to galactose-alpha-1,‍3-galactose. J Allergy Clin Immunol, 134(1):108-115.e11.

[38]CostantiniVP, NguyenK, LyskiZ, et al., 2022. Development and validation of an enzyme immunoassay for detection and quantification of SARS-CoV-2 salivary IgA and IgG. J Immunol, 208(6):1500-1508.

[39]CrestaniE, HarbH, CharbonnierLM, et al., 2020. Untargeted metabolomic profiling identifies disease-specific signatures in food allergy and asthma. J Allergy Clin Immunol, 145(3):897-906.

[40]CrivellatoE, TravanL, RibattiD, 2010. Mast cells and basophils: a potential link in promoting angiogenesis during allergic inflammation. Int Arch Allergy Immunol, 151(2):89-97.

[41]CucuT, de MeulenaerB, BridtsC, et al., 2012. Impact of thermal processing and the Maillard reaction on the basophil activation of hazelnut allergic patients. Food Chem Toxicol, 50(5):1722-1728.

[42]CuomoB, IndirliGC, BianchiA, et al., 2017. Specific IgE and skin prick tests to diagnose allergy to fresh and baked cow’s milk according to age: a systematic review. Ital J Pediatr, 43:93.

[43]de QuirosEB, Seoane-ReulaE, Alonso-LebreroE, et al., 2018. The role of regulatory T cells in the acquisition of tolerance to food allergens in children. Allergol Immunopathol (Madr), 46(6):612-618.

[44]di GirolamoF, MuracaM, MazzinaO, et al., 2015. Proteomic applications in food allergy: food allergenomics. Curr Opin Allergy Clin Immunol, 15(3):259-266.

[45]DoAN, WatsonCT, CohainAT, et al., 2020. Dual transcriptomic and epigenomic study of reaction severity in peanut-allergic children. J Allergy Clin Immunol, 145(4):‍1219-1230.

[46]DreskinSC, GerminaroM, ReinholdD, et al., 2019. IgE binding to linear epitopes of Ara h 2 in peanut allergic preschool children undergoing oral immunotherapy. Pediatr Allergy Immunol, 30(8):817-823.

[47]DuboisB, ChapatL, GoubierA, et al., 2003. Innate CD4+CD25+ regulatory T cells are required for oral tolerance and inhibition of CD8+ T cells mediating skin inflammation. Blood, 102(9):3295-3301.

[48]EbrusterH, 1959. The prick test, a recent cutaneous test for the diagnosis of allergic disorders. Wien Klin Wochenschr, 71:551-554.

[49]EigenmannPA, SampsonHA, 1998. Interpreting skin prick tests in the evaluation of food allergy in children. Pediatr Allergy Immunol, 9(4):‍186-191.

[50]EiweggerT, HungL, SanDiego KE, et al., 2019. Recent developments and highlights in food allergy. Allergy, 74(12):2355-2367.

[51]EscuderoC, del RíoPR, Sánchez-GarcíaS, et al., 2015. Early sustained unresponsiveness after short-course egg oral immunotherapy: a randomized controlled study in egg-allergic children. Clin Exp Allergy, 45(12):1833-1843.

[52]EsterházyD, LoschkoJ, LondonM, et al., 2016. Classical dendritic cells are required for dietary antigen-mediated induction of peripheral Treg cells and tolerance. Nat Immunol, 17(5):545-555.

[53]FanDC, WangXD, WangCS, et al., 2016. Suppression of immunotherapy on group 2 innate lymphoid cells in allergic rhinitis. Chin Med J (Engl), 129(23):2824-2828.

[54]FengT, ElsonCO, CongYZ, 2011. Treg cell-IgA axis in maintenance of host immune homeostasis with microbiota. Int Immunopharmacol, 11(5):589-592.

[55]FlintermanAE, KnolEF, LencerDA, et al., 2008. Peanut epitopes for IgE and IgG4 in peanut-sensitized children in relation to severity of peanut allergy. J Allergy Clin Immunol, 121(3):737-743.e10.

[56]FratiF, IncorvaiaC, CavaliereC, et al., 2018. The skin prick test. J Biol Regul Homeost Agents, 32(1 Suppl 1):19-24.

[57]Frischmeyer-GuerrerioPA, GuerrerioAL, ChichesterKL, et al., 2011. Dendritic cell and T cell responses in children with food allergy. Clin Exp Allergy, 41(1):61-71.

[58]Frischmeyer-GuerrerioPA, KeetCA, GuerrerioAL, et al., 2014. Modulation of dendritic cell innate and adaptive immune functions by oral and sublingual immunotherapy. Clin Immunol, 155(1):47-59.

[59]Frischmeyer-GuerrerioPA, MasilamaniM, GuWJ, et al., 2017. Mechanistic correlates of clinical responses to omalizumab in the setting of oral immunotherapy for milk allergy. J Allergy Clin Immunol, 140(4):1043-1053.e8.

[60]FuLL, SongJY, WangC, et al., 2017. Bifidobacterium infantis potentially alleviates shrimp tropomyosin-induced allergy by tolerogenic dendritic cell-dependent induction of regulatory T cells and alterations in gut microbiota. Front Immunol, 8:1536.

[61]GernezY, Nowak-WegrzynA, 2017. Immunotherapy for food allergy: are we there yet? J Allergy Clin Immunol Pract, 5(2):250-272.

[62]GiudiceG, PetsalakiE, 2019. Proteomics and phosphoproteomics in precision medicine: applications and challenges. Brief Bioinform, 20(3):767-777.

[63]GlaumannS, NoppA, JohanssonSGO, et al., 2012. Basophil allergen threshold sensitivity, CD-sens, IgE-sensitization and DBPCFC in peanut-sensitized children. Allergy, 67(2):242-247.

[64]GoberLM, EckmanJA, SterbaPM, et al., 2007. Expression of activation markers on basophils in a controlled model of anaphylaxis. J Allergy Clin Immunol, 119(5):1181-1188.

[65]GorelikM, NarisetySD, GuerrerioAL, et al., 2015. Suppression of the immunologic response to peanut during immunotherapy is often transient. J Allergy Clin Immunol, 135(5):1283-1292.

[66]GreenhawtM, 2015. The learning early about peanut allergy study: the benefits of early peanut introduction, and a new horizon in fighting the food allergy epidemic. Pediatr Clin North Am, 62(6):1509-1521.

[67]GuptaRS, LauCH, HamiltonRG, et al., 2014. Predicting outcomes of oral food challenges by using the allergen-specific IgE-total IgE ratio. J Allergy Clin Immunol Pract, 2(3):300-305.

[68]HadisU, WahlB, SchulzO, et al., 2011. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity, 34(2):237-246.

[69]HaoGD, ZhengYW, WangZX, et al., 2016. High correlation of specific IgE sensitization between birch pollen, soy and apple allergens indicates pollen-food allergy syndrome among birch pollen allergic patients in northern China. J Zhejiang Univ-Sci B (Biomed & Biotechnol), 17(5):‍399-404.

[70]HeZY, VadaliVLG, SzabadyRL, et al., 2021. Increased diversity of gut microbiota during active oral immunotherapy in peanut-allergic adults. Allergy, 76(3):927-930.

[71]HeeringaJJ, McKenzieCI, VareseN, et al., 2020. Induction of IgG2 and IgG4 B-cell memory following sublingual immunotherapy for ryegrass pollen allergy. Allergy, 75(5):1121-1132.

[72]HesselmarB, Hicke-RobertsA, WennergrenG, 2015. Allergy in children in hand versus machine dishwashing. Pediatrics, 135(3):e590-e597.

[73]HohRA, JoshiSA, LiuY, et al., 2016. Single B-cell deconvolution of peanut-specific antibody responses in allergic patients. J Allergy Clin Immunol, 137(1):157-167.

[74]HongHY, LiaoSM, ChenFH, et al., 2020. Role of IL-25, IL-33, and TSLP in triggering united airway diseases toward type 2 inflammation. Allergy, 75(11):2794-2804.

[75]HourihaneJOB, BeyerK, AbbasA, et al., 2020. Efficacy and safety of oral immunotherapy with AR101 in European children with a peanut allergy (ARTEMIS): a multicentre, double-blind, randomised, placebo-controlled phase 3 trial. Lancet Child Adolesc Health, 4(10):728-739.

[76]ImakiireR, FujisawaT, NagaoM, et al., 2020. Basophil activation test based on CD203c expression in the diagnosis of fish allergy. Allergy Asthma Immunol Res, 12(4):‍641-652.

[77]JanziM, KullI, SjöbergR, et al., 2009. Selective IgA deficiency in early life: association to infections and allergic diseases during childhood. Clin Immunol, 133(1):78-85.

[78]JärvinenKM, SichererSH, 2012. Diagnostic oral food challenges: procedures and biomarkers. J Immunol Methods, 383(1-2):30-38.

[79]JiJJ, XuJJ, ZhaoSL, et al., 2016. Myeloid-derived suppressor cells contribute to systemic lupus erythaematosus by regulating differentiation of Th17 cells and Tregs. Clin Sci (Lond), 130(16):1453-1467.

[80]JonesSM, PonsL, RobertsJL, et al., 2009. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol, 124(2):292-300.e97.

[81]JonesSM, BurksAW, KeetC, et al., 2016. Long-term treatment with egg oral immunotherapy enhances sustained unresponsiveness that persists after cessation of therapy. J Allergy Clin Immunol, 137(4):1117-1127.e10.

[82]JutelM, AkdisM, BudakF, et al., 2003. IL-10 and TGF-‍βcooperate in the regulatory T cell response to mucosal allergens in normal immunity and specific immunotherapy. Eur J Immunol, 33(5):1205-1214.

[83]KimEH, PerryTT, WoodRA, et al., 2020. Induction of sustained unresponsiveness after egg oral immunotherapy compared to baked egg therapy in children with egg allergy. J Allergy Clin Immunol, 146(4):851-862.e10.

[84]KimYH, KimYS, ParkY, et al., 2020. Investigation of basophil activation test for diagnosing milk and egg allergy in younger children. J Clin Med, 9(12):3942.

[85]KiyotaniK, MaiTH, YamaguchiR, et al., 2018. Characterization of the B-cell receptor repertoires in peanut allergic subjects undergoing oral immunotherapy. J Hum Genet, 63(2):239-248.

[86]KomataT, SöderströmL, BorresMP, et al., 2007. The predictive relationship of food-specific serum IgE concentrations to challenge outcomes for egg and milk varies by patient age. J Allergy Clin Immunol, 119(5):1272-1274.

[87]KomlósiZI, KovácsN, van de VeenW, et al., 2018. Human CD40 ligand-expressing type 3 innate lymphoid cells induce IL-10-producing immature transitional regulatory B cells. J Allergy Clin Immunol, 142(1):178-194.e11.

[88]KongJ, ChalcraftK, MandurTS, et al., 2015. Comprehensive metabolomics identifies the alarmin uric acid as a critical signal for the induction of peanut allergy. Allergy, 70(5):495-505.

[89]KosoyR, AgasheC, GrishinA, et al., 2016. Transcriptional profiling of egg allergy and relationship to disease phenotype. PLoS ONE, 11(10):e0163831.

[90]KostaraM, ChondrouV, SgourouA, et al., 2020. HLA polymorphisms and food allergy predisposition. J Pediatr Genet, 9(2):77-86.

[91]KrishnaMT, HuissoonAP, 2011. Clinical immunology review series: an approach to desensitization. Clin Exp Immunol, 163(2):131-146.

[92]KrutzNL, WingetJ, RyanCA, et al., 2019. Proteomic and bioinformatic analyses for the identification of proteins with low allergenic potential for hazard assessment. Toxicol Sci, 170(1):210-222.

[93]KuboM, 2018. Mast cells and basophils in allergic inflammation. Curr Opin Immunol, 54:74-79.

[94]KulisM, SabaK, KimEH, et al., 2012. Increased peanut-specific IgA levels in saliva correlate with food challenge outcomes after peanut sublingual immunotherapy. J Allergy Clin Immunol, 129(4):1159-1162.

[95]KulisM, YueXH, GuoRS, et al., 2019. High- and low-dose oral immunotherapy similarly suppress pro-allergic cytokines and basophil activation in young children. Clin Exp Allergy, 49(2):180-189.

[96]KulkarniDH, GustafssonJK, KnoopKA, et al., 2020. Goblet cell associated antigen passages support the induction and maintenance of oral tolerance. Mucosal Immunol, 13(2):271-282.

[97]KurashimaY, KunisawaJ, HiguchiM, et al., 2007. Sphingosine 1-phosphate-mediated trafficking of pathogenic Th2 and mast cells for the control of food allergy. J Immunol, 179(3):1577-1585.

[98]Lao-ArayaM, StevelingE, ScaddingGW, et al., 2014. Seasonal increases in peripheral innate lymphoid type 2 cells are inhibited by subcutaneous grass pollen immunotherapy. J Allergy Clin Immunol, 134(5):1193-1195.e4.

[99]LeungDYM, SampsonHA, YungingerJW, et al., 2003. Effect of anti-IgE therapy in patients with peanut allergy. N Engl J Med, 348(11):986-993.

[100]Leveson-GowerDB, SegaEI, KalesnikoffJ, et al., 2013. Mast cells suppress murine GVHD in a mechanism independent of CD4+CD25+ regulatory T cells. Blood, 122(22):3659-3665.

[101]LiJ, FungI, GlessnerJT, et al., 2015. Copy number variations in CTNNA3 and RBFOX1 associate with pediatric food allergy. J Immunol, 195(4):1599-1607.

[102]LiuEG, YinXY, SwaminathanA, et al., 2021. Antigen-presenting cells in food tolerance and allergy. Front Immunol, 11:616020.

[103]LopesJP, SichererS, 2020. Food allergy: epidemiology, pathogenesis, diagnosis, prevention, and treatment. Curr Opin Immunol, 66:57-64.

[104]LuceS, ChinthrajahS, LyuSC, et al., 2020. Th2A and Th17 cell frequencies and regulatory markers as follow-up biomarker candidates for successful multifood oral immunotherapy. Allergy, 75(6):1513-1516.

[105]LuceS, BatardT, FlochVBL, et al., 2021. Decrease in CD38+ TH2A cell frequencies following immunotherapy with house dust mite tablet correlates with humoral responses. Clin Exp Allergy, 51(8):1057-1068.

[106]LúðvíkssonBR, ArasonGJ, ThorarensenO, et al., 2005. Allergic diseases and asthma in relation to serum immunoglobulins and salivary immunoglobulin A in pre-school children: a follow-up community-based study. Clin Exp Allergy, 35(1):64-69.

[107]MarrsT, FlohrC, PerkinMR, 2015. Assessing the efficacy of oral immunotherapy for the desensitisation of peanut allergy in children (STOP II): a phase 2 randomised controlled trial: a critical appraisal. Br J Dermatol, 173(5):1125-1129.

[108]Martínez-BotasJ, Rodríguez-ÁlvarezM, CerecedoI, et al., 2015. Identification of novel peptide biomarkers to predict safety and efficacy of cow’s milk oral immunotherapy by peptide microarray. Clin Exp Allergy, 45(6):1071-1084.

[109]McDoleJR, WheelerLW, McDonaldKG, et al., 2012. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature, 483(7389):345-349.

[110]McShaneLM, CavenaghMM, LivelyTG, et al., 2013. Criteria for the use of omics-based predictors in clinical trials: explanation and elaboration. BMC Med, 11:220.

[111]Méndez-EnríquezE, SalomonssonM, ErikssonJ, et al., 2022. IgE cross-linking induces activation of human and mouse mast cell progenitors. J Allergy Clin Immunol, 149(4):1458-1463.

[112]MershaTB, AfanadorY, JohanssonE, et al., 2021. Resolving clinical phenotypes into endotypes in allergy: molecular and omics approaches. Clin Rev Allergy Immunol, 60(2):200-219.

[113]MitthamsiriW, PradubpongsaP, SangasapaviliyaA, et al., 2018. Decreased CRTH2 expression and response to allergen re-stimulation on innate lymphoid cells in patients with allergen-specific immunotherapy. Allergy Asthma Immunol Res, 10(6):662-674.

[114]MondouletL, DioszeghyV, BusatoF, et al., 2019. Gata3 hypermethylation and Foxp3 hypomethylation are associated with sustained protection and bystander effect following epicutaneous immunotherapy in peanut-sensitized mice. Allergy, 74(1):152-164.

[115]MonticelliLA, OsborneLC, NotiM, et al., 2015. IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions. Proc Natl Acad Sci USA, 112(34):10762-10767.

[116]MoralyT, de ChambureDP, VerdunS, et al., 2020. Oral immunotherapy for hazelnut allergy: a single-center retrospective study on 100 patients. J Allergy Clin Immunol Pract, 8(2):704-709.e4.

[117]MoritaH, AraeK, UnnoH, et al., 2015. An interleukin-33-mast cell-interleukin-2 axis suppresses papain-induced allergic inflammation by promoting regulatory T cell numbers. Immunity, 43(1):175-186.

[118]MousallemT, BurksAW, 2012. Immunology in the clinic review series; focus on allergies: immunotherapy for food allergy. Clin Exp Immunol, 167(1):26-31.

[119]NachshonL, GoldbergMR, KatzY, et al., 2018. Long-term outcome of peanut oral immunotherapy-real-life experience. Pediatr Allergy Immunol, 29(5):519-526.

[120]NagakuraKI, YanagidaN, SatoS, et al., 2018. Low-dose oral immunotherapy for children with anaphylactic peanut allergy in Japan. Pediatr Allergy Immunol, 29(5):512-518.

[121]NagakuraKI, SatoS, MiuraY, et al., 2021. A randomized trial of oral immunotherapy for pediatric cow’s milk-induced anaphylaxis: heated vs unheated milk. Pediatr Allergy Immunol, 32(1):161-169.

[122]NakajimaS, KabataH, KabashimaK, et al., 2020. Anti-TSLP antibodies: targeting a master regulator of type 2 immune responses. Allergol Int, 69(2):197-203.

[123]NakanoN, KitauraJ, 2022. Mucosal mast cells as key effector cells in food allergies. Cells, 11(3):329.

[124]NiessJH, AdlerG, 2010. Enteric flora expands gut lamina propria CX3CR1+ dendritic cells supporting inflammatory immune responses under normal and inflammatory conditions. J Immunol, 184(4):2026-2037.

[125]Nowak-WęgrzynA, AlbinS, 2015. Oral immunotherapy for food allergy: mechanisms and role in management. Clin Exp Allergy, 45(2):368-383.

[126]Nowak-WegrzynA, BloomKA, SichererSH, et al., 2008. Tolerance to extensively heated milk in children with cow’s milk allergy. J Allergy Clin Immunol, 122(2):‍342-347.e2.

[127]Nowak-WęgrzynA, WoodRA, NadeauKC, et al., 2019. Multicenter, randomized, double-blind, placebo-controlled clinical trial of vital wheat gluten oral immunotherapy. J Allergy Clin Immunol, 143(2):651-661.e9.

[128]NozawaA, OkamotoY, MovérareR, et al., 2014. Monitoring Ara h 1, 2 and 3-sIgE and sIgG4 antibodies in peanut allergic children receiving oral rush immunotherapy. Pediatr Allergy Immunol, 25(4):323-328.

[129]NuceraE, InchingoloR, NicotraR, et al., 2021. Influence of antihistamines on basophil activation test in food allergy to milk and egg. Diagnostics (Basel), 11(1):44.

[130]NurmatovU, DhamiS, ArasiS, et al., 2017. Allergen immunotherapy for IgE-mediated food allergy: a systematic review and meta-analysis. Allergy, 72(8):1133-1147.

[131]OcmantA, MulierS, HanssensL, et al., 2009. Basophil activation tests for the diagnosis of food allergy in children. Clin Exp Allergy, 39(8):1234-1245.

[132]OrgelK, BurkC, SmeekensJ, et al., 2019. Blocking antibodies induced by peanut oral and sublingual immunotherapy suppress basophil activation and are associated with sustained unresponsiveness. Clin Exp Allergy, 49(4):‍461-470.

[133]PajnoGB, BernardiniR, PeroniD, et al., 2017. Clinical practice recommendations for allergen-specific immunotherapy in children: the Italian consensus report. Ital J Pediatr, 43(1):13.

[134]PatelG, SaltounC, 2019. Skin testing in allergy. Allergy Asthma Proc, 40(6):366-368.

[135]PatilSU, OgunniyiAO, CalatroniA, et al., 2015. Peanut oral immunotherapy transiently expands circulating Ara h 2-specific B cells with a homologous repertoire in unrelated subjects. J Allergy Clin Immunol, 136(1):125-134.e12.

[136]PatilSU, SteinbrecherJ, CalatroniA, et al., 2019. Early decrease in basophil sensitivity to Ara h 2 precedes sustained unresponsiveness after peanut oral immunotherapy. J Allergy Clin Immunol, 144(5):1310-1319.e4.

[137]PatilSU, BunyavanichS, BerinMC, 2020. Emerging food allergy biomarkers. J Allergy Clin Immunol Pract, 8(8):2516-2524.

[138]PatriarcaG, SchiavinoD, NuceraE, et al., 1998. Food allergy in children: results of a standardized protocol for oral desensitization. Hepatogastroenterology, 45(19):52-58.

[139]PerezábadL, RecheM, ValbuenaT, et al., 2015. Clinical efficacy and immunological changes subjacent to egg oral immunotherapy. Ann Allergy Asthma Immunol, 114(6):504-509.

[140]PerezábadL, RecheM, ValbuenaT, et al., 2017. Oral food desensitization in children with IgE-mediated cow’s milk allergy: immunological changes underlying desensitization. Allergy Asthma Immunol Res, 9(1):35-42.

[141]PerkinMR, LoganK, TsengA, et al., 2016. Randomized trial of introduction of allergenic foods in breast-fed infants. N Engl J Med, 374(18):1733-1743.

[142]PlanchaisC, MouquetH, 2020. Easy pan-detection of human IgA immunoglobulins. J Immunol Methods, 484-485:112833.

[143]PlumT, WangX, RettelM, et al., 2020. Human mast cell proteome reveals unique lineage, putative functions, and structural basis for cell ablation. Immunity, 52(2):404-416.e5.

[144]RachidR, ChatilaTA, 2016. The role of the gut microbiota in food allergy. Curr Opin Pediatr, 28(6):748-753.

[145]RamananD, SefikE, Galván-PeñaS, et al., 2020. An immunologic mode of multigenerational transmission governs a gut Treg setpoint. Cell, 181(6):1276-1290.e13.

[146]RivasMN, BurtonOT, WiseP, et al., 2015. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity, 42(3):512-523.

[147]Rolinck-WerninghausC, StadenU, MehlA, et al., 2005. Specific oral tolerance induction with food in children: transient or persistent effect on food allergy? Allergy, 60(10):1320-1322.

[148]RubioA, Vivinus-NébotM, BourrierT, et al., 2011. Benefit of the basophil activation test in deciding when to reintroduce cow’s milk in allergic children. Allergy, 66(1):92-100.

[149]Ruinemans-KoertsJ, Schmidt-HieltjesY, JansenA, et al., 2019. The Basophil Activation Test reduces the need for a food challenge test in children suspected of IgE-mediated cow’s milk allergy. Clin Exp Allergy, 49(3):350-356.

[150]RuiterB, ShrefflerWG, 2012. The role of dendritic cells in food allergy. J Allergy Clin Immunol, 129(4):921-928.

[151]RyanJF, HovdeR, GlanvilleJ, et al., 2016. Successful immunotherapy induces previously unidentified allergen-specific CD4+ T-cell subsets. Proc Natl Acad Sci USA, 113(9):E1286-E1295.

[152]SackesenC, Suárez-FariñasM, SilvaR, et al., 2019. A new Luminex-based peptide assay to identify reactivity to baked, fermented, and whole milk. Allergy, 74(2):‍327-336.

[153]SalamonP, SheflerI, MoshkovitsI, et al., 2017. IL-33 and IgE stimulate mast cell production of IL-2 and regulatory T cell expansion in allergic dermatitis. Clin Exp Allergy, 47(11):1409-1416.

[154]SalmivesiS, PaassiltaM, HuhtalaH, et al., 2018. Elevated serum adipsin may predict unsuccessful treatment for cows’ milk allergy but other biomarkers do not. Acta Paediatr, 107(2):328-332.

[155]SantosAF, LackG, 2016. Basophil activation test: food challenge in a test tube or specialist research tool? Clin Transl Allergy, 6:10.

[156]SantosAF, DouiriA, BécaresN, et al., 2014. Basophil activation test discriminates between allergy and tolerance in peanut-sensitized children. J Allergy Clin Immunol, 134(3):645-652.

[157]SantosAF, DuToit G, DouiriA, et al., 2015a. Distinct parameters of the basophil activation test reflect the severity and threshold of allergic reactions to peanut. J Allergy Clin Immunol, 135(1):179-186.

[158]SantosAF, JamesLK, BahnsonHT, et al., 2015b. IgG4 inhibits peanut-induced basophil and mast cell activation in peanut-tolerant children sensitized to peanut major allergens. J Allergy Clin Immunol, 135(5):1249-1256.

[159]SantosAF, Couto-FranciscoN, BécaresN, et al., 2018. A novel human mast cell activation test for peanut allergy. J Allergy Clin Immunol, 142(2):689-691.e9.

[160]SantosAF, DuToit G, O'RourkeC, et al., 2020. Biomarkers of severity and threshold of allergic reactions during oral peanut challenges. J Allergy Clin Immunol, 146(2):‍344-355.

[161]SantosAF, AlpanO, HoffmannHJ, 2021. Basophil activation test: mechanisms and considerations for use in clinical trials and clinical practice. Allergy, 76(8):2420-2432.

[162]SanzML, MaselliJP, GamboaPM, et al., 2002. Flow cytometric basophil activation test: a review. J Investig Allergol Clin Immunol, 12(3):143-154.

[163]SarfatiM, WakaharaK, ChapuyL, et al., 2015. Mutual interaction of basophils and T cells in chronic inflammatory diseases. Front Immunol, 6:399.

[164]SatoguinaJS, WeyandE, LarbiJ, et al., 2005. T regulatory-1 cells induce IgG4 production by B cells: role of IL-10. J Immunol, 174(8):4718-4726.

[165]SavilahtiEM, KuitunenM, SavilahtiE, et al., 2014a. Specific antibodies in oral immunotherapy for cow’s milk allergy: kinetics and prediction of clinical outcome. Int Arch Allergy Immunol, 164(1):32-39.

[166]SavilahtiEM, KuitunenM, ValoriM, et al., 2014b. Use of IgE and IgG4 epitope binding to predict the outcome of oral immunotherapy in cow’s milk allergy. Pediatr Allergy Immunol, 25(3):227-235.

[167]Schmidt-WeberCB, BlaserK, 2002. T-cell tolerance in allergic response. Allergy, 57(9):762-768.

[168]SchwarzA, PanettaV, CappellaA, et al., 2016. IgG and IgG4 to 91 allergenic molecules in early childhood by route of exposure and current and future IgE sensitization: results from the Multicentre Allergy Study birth cohort. J Allergy Clin Immunol, 138(5):1426-1433.e12.

[169]ShakoorZ, al FaifiA, al AmroB, et al., 2016. Prevalence of IgG-mediated food intolerance among patients with allergic symptoms. Ann Saudi Med, 36(6):386-390.

[170]ShamjiMH, KappenJ, Abubakar-WaziriH, et al., 2019. Nasal allergen-neutralizing IgG4 antibodies block IgE-mediated responses: novel biomarker of subcutaneous grass pollen immunotherapy. J Allergy Clin Immunol, 143(3):1067-1076.

[171]ShikD, TomarS, LeeJB, et al., 2017. IL-9-producing cells in the development of IgE-mediated food allergy. Semin Immunopathol, 39(1):69-77.

[172]ShiokawaA, KotakiR, TakanoT, et al., 2017. Mesenteric lymph node CD11b- CD103+ PD-L1high dendritic cells highly induce regulatory T cells. Immunology, 152(1):‍52-64.

[173]ShrefflerWG, LencerDA, BardinaL, et al., 2005. IgE and IgG4 epitope mapping by microarray immunoassay reveals the diversity of immune response to the peanut allergen, Ara h 2. J Allergy Clin Immunol, 116(4):893-899.

[174]ShrefflerWG, WanichN, MoloneyM, et al., 2009. Association of allergen-specific regulatory T cells with the onset of clinical tolerance to milk protein. J Allergy Clin Immunol, 123(1):43-52.e7.

[175]SichererSH, SampsonHA, 2014. Food allergy: epidemiology, pathogenesis, diagnosis, and treatment. J Allergy Clin Immunol, 133(2):291-307.e5.

[176]SmaldiniPL, DelgadoMLO, FossatiCA, et al., 2015. Orally-induced intestinal CD4+ CD25+ FoxP3+ Treg controlled undesired responses towards oral antigens and effectively dampened food allergic reactions. PLoS ONE, 10(10):e0141116.

[177]SmartJM, KempAS, 2002. Increased Th1 and Th2 allergen-induced cytokine responses in children with atopic disease. Clin Exp Allergy, 32(5):796-802.

[178]SmeekensJM, KulisMD, 2020. Evolution of immune responses in food immunotherapy. Immunol Allergy Clin North Am, 40(1):87-95.

[179]SongY, WangJL, LeungN, et al., 2015. Correlations between basophil activation, allergen-specific IgE with outcome and severity of oral food challenges. Ann Allergy Asthma Immunol, 114(4):319-326.

[180]SpertiniF, 2020. Metabolomics and allergy: opening Pandora’s box. J Allergy Clin Immunol, 145(3):782-784.

[181]SporikR, HillDJ, HoskingCS, 2000. Specificity of allergen skin testing in predicting positive open food challenges to milk, egg and peanut in children. Clin Exp Allergy, 30(11):1540-1546.

[182]StanicB, van de VeenW, WirzOF, et al., 2015. IL-10-overexpressing B cells regulate innate and adaptive immune responses. J Allergy Clin Immunol, 135(3):‍771-780.e8.

[183]Suárez-FariñasM, SuprunM, ChangHL, et al., 2019. Predicting development of sustained unresponsiveness to milk oral immunotherapy using epitope-specific antibody binding profiles. J Allergy Clin Immunol, 143(3):1038-1046.

[184]SugimotoM, KamemuraN, NagaoM, et al., 2016. Differential response in allergen-specific IgE, IgGs, and IgA levels for predicting outcome of oral immunotherapy. Pediatr Allergy Immunol, 27(3):276-282.

[185]SuprunM, SichererSH, WoodRA, et al., 2020. Early epitope-specific IgE antibodies are predictive of childhood peanut allergy. J Allergy Clin Immunol, 146(5):1080-1088.

[186]SyedA, GarciaMA, LyuSC, et al., 2014. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol, 133(2):‍500-510.e11.

[187]TakasatoY, KurashimaY, KiuchiM, et al., 2021. Orally desensitized mast cells form a regulatory network with Treg cells for the control of food allergy. Mucosal Immunol, 14(3):640-651.

[188]TangMLK, PonsonbyAL, OrsiniF, et al., 2015. Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol, 135(3):‍737-744.e8.

[189]The Medical Letter, 2020. Peanut allergen powder (PalfoPrzia). JAMA, 324(2):192-193.

[190]The PALISADE Group of Clinical Investigators, 2018. AR101 oral immunotherapy for peanut allergy. N Engl J Med, 379(21):1991-2001.

[191]ThotaLN, PonnusamyT, PhilipS, et al., 2017. Immune regulation by oral tolerance induces alternate activation of macrophages and reduces markers of plaque destabilization in Apobtm2Sgy/Ldlrtm1Her/J mice. Sci Rep, 7:3997.

[192]TordesillasL, BerinMC, 2018. Mechanisms of oral tolerance. Clin Rev Allergy Immunol, 55(2):107-117.

[193]TordesillasL, MondouletL, BlazquezAB, et al., 2017. Epicutaneous immunotherapy induces gastrointestinal LAP+ regulatory T cells and prevents food-induced anaphylaxis. J Allergy Clin Immunol, 139(1):189-201.e4.

[194]TorgersonTR, LinaneA, MoesN, et al., 2007. Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology, 132(5):1705-1717.

[195]TsaiM, MukaiK, ChinthrajahRS, et al., 2020. Sustained successful peanut oral immunotherapy associated with low basophil activation and peanut-specific IgE. J Allergy Clin Immunol, 145(3):885-896.e6.

[196]TsujiM, KomatsuN, KawamotoS, et al., 2009. Preferential generation of follicular B helper T cells from Foxp3+ T cells in gut peyer’s patches. Science, 323(5920):1488-1492.

[197]TurcanuV, BroughHA, DuToit G, et al., 2017. Immune mechanisms of food allergy and its prevention by early intervention. Curr Opin Immunol, 48:92-98.

[198]UranüsS, KronbergerL, NeumayerK, et al., 1990. TA-stapler resection of congenital splenic cyst. Case report. Acta Chir Scand, 156(3):247-249.

[199]van de VeenW, StanicB, YamanG, et al., 2013. IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J Allergy Clin Immunol, 131(4):1204-1212.

[200]van de VeenW, StanicB, WirzOF, et al., 2016. Role of regulatory B cells in immune tolerance to allergens and beyond. J Allergy Clin Immunol, 138(3):654-665.

[201]VarolC, Vallon-EberhardA, ElinavE, et al., 2009. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity, 31(3):502-512.

[202]VarshneyP, JonesSM, ScurlockAM, et al., 2011. A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. J Allergy Clin Immunol, 127(3):654-660.

[203]Vazquez-OrtizM, PascalM, AlsinaL, et al., 2013. Serum sIgG4, but not sIgA, is involved in induced and natural tolerance to egg allergens. Clin Transl Allergy, 3(3):O6.

[204]VickeryBP, PonsL, KulisM, et al., 2010. Individualized IgE-based dosing of egg oral immunotherapy and the development of tolerance. Ann Allergy Asthma Immunol, 105(6):444-450.

[205]VickeryBP, LinJ, KulisM, et al., 2013. Peanut oral immunotherapy modifies IgE and IgG4 responses to major peanut allergens. J Allergy Clin Immunol, 131(1):128-134.e3.

[206]VickeryBP, ScurlockAM, KulisM, et al., 2014. Sustained unresponsiveness to peanut in subjects who have completed peanut oral immunotherapy. J Allergy Clin Immunol, 133(2):468-475.e6.

[207]VickeryBP, BerglundJP, BurkCM, et al., 2017. Early oral immunotherapy in peanut-allergic preschool children is safe and highly effective. J Allergy Clin Immunol, 139(1):173-181.e8.

[208]VivierE, ArtisD, ColonnaM, et al., 2018. Innate lymphoid cells: 10 years on. Cell, 174(5):1054-1066.

[209]VonkMM, DiksMAP, WagenaarL, et al., 2017. Improved efficacy of oral immunotherapy using non-digestible oligosaccharides in a murine cow’s milk allergy model: a potential role for Foxp3+ regulatory T cells. Front Immunol, 8:1230.

[210]VonkMM, BlokhuisBRJ, DiksMAP, et al., 2019. Butyrate enhances desensitization induced by oral immunotherapy in cow’s milk allergic mice. Mediators Inflamm, 2019:9062537.

[211]WaiCYY, LeungNYH, LeungASY, et al., 2021. Cell-based functional IgE assays are superior to conventional allergy tests for shrimp allergy diagnosis. J Allergy Clin Immunol Pract, 9(1):236-244.e9.

[212]WambreE, DelongJH, JamesEA, et al., 2012. Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy. J Allergy Clin Immunol, 129(2):544-551.e7.

[213]WambreE, BajzikV, DelongJH, et al., 2017. A phenotypically and functionally distinct human TH2 cell subpopulation is associated with allergic disorders. Sci Transl Med, 9(401):eaam9171.

[214]WangS, XiaPY, ChenY, et al., 2017. Regulatory innate lymphoid cells control innate intestinal inflammation. Cell, 171(1):201-216.e18.

[215]WangYG, DingYX, GuoNZ, et al., 2019. MDSCs: key criminals of tumor pre-metastatic niche formation. Front Immunol, 10:172.

[216]WeltyNE, StaleyC, GhilardiN, et al., 2013. Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism. J Exp Med, 210(10):2011-2024.

[217]WrightBL, KulisM, OrgelKA, et al., 2016. Component-resolved analysis of IgA, IgE, and IgG4 during egg OIT identifies markers associated with sustained unresponsiveness. Allergy, 71(11):1552-1560.

[218]YamashitaH, MatsuharaH, MiotaniS, et al., 2017. Artificial sweeteners and mixture of food additives cause to break oral tolerance and induce food allergy in murine oral tolerance model for food allergy. Clin Exp Allergy, 47(9):1204-1213.

[219]YeeLM, LivelyTG, McShaneLM, 2018. Biomarkers in early-phase trials: fundamental issues. Bioanalysis, 10(12):‍933-944.

[220]YoneyamaT, NakanoN, HaraM, et al., 2021. Notch signaling contributes to the establishment of sustained unresponsiveness to food allergens by oral immunotherapy. J Allergy Clin Immunol, 147(3):1063-1076.e9.

[221]ZhouL, ChuC, TengF, et al., 2019. Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature, 568(7752):405-409.

Open peer comments: Debate/Discuss/Question/Opinion

<1>

Please provide your name, email address and a comment





Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou 310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn
Copyright © 2000 - 2024 Journal of Zhejiang University-SCIENCE