REFERENCES
1. Jensen-Jarolim E, Achatz G, Turner MC, et al. AllergoOncology: the role of IgE-mediated allergy in cancer. Allergy. Oct 2008;63(10):1255-66. doi:10.1111/j.1398-9995.2008.01768.x
2. Jensen-Jarolim E, Bax HJ, Bianchini R, et al. AllergoOncology: Opposite outcomes of immune tolerance in allergy and cancer. Allergy. Feb 2018;73(2):328-340. doi:10.1111/all.13311
3. Untersmayr E, Bax HJ, Bergmann C, et al. AllergoOncology: Microbiota in allergy and cancer-A European Academy for Allergy and Clinical Immunology position paper. Allergy. Jun 2019;74(6):1037-1051. doi:10.1111/all.13718
4. Bergmann C, Poli A, Agache I, et al. AllergoOncology: Danger signals in Allergology and Oncology. A European Academy of Allergy and Clinical Immunology (EAACI) Position Paper. Allergy. Feb 13 2022;doi:10.1111/all.15255
5. Ferastraoaru D, Bax HJ, Bergmann C, et al. AllergoOncology: ultra-low IgE, a potential novel biomarker in cancer-a Position Paper of the European Academy of Allergy and Clinical Immunology (EAACI). Clin Transl Allergy. 2020;10:32. doi:10.1186/s13601-020-00335-w
6. Turner MC. Epidemiology: allergy history, IgE, and cancer. Cancer immunology, immunotherapy : CII. Sep 2012;61(9):1493-510. doi:10.1007/s00262-011-1180-6
7. Amirian ES, Zhou R, Wrensch MR, et al. Approaching a Scientific Consensus on the Association between Allergies and Glioma Risk: A Report from the Glioma International Case-Control Study. Cancer Epidemiol Biomarkers Prev. Feb 2016;25(2):282-90. doi:10.1158/1055-9965.EPI-15-0847
8. McCraw AJ, Chauhan J, Bax HJ, et al. Insights from IgE Immune Surveillance in Allergy and Cancer for Anti-Tumour IgE Treatments. Cancers (Basel). Sep 4 2021;13(17)doi:10.3390/cancers13174460
9. Ogulur I, Pat Y, Ardicli O, et al. Advances and highlights in biomarkers of allergic diseases. Allergy. Dec 2021;76(12):3659-3686. doi:10.1111/all.15089
10. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. Aug 2 2021;23(8):1231-1251. doi:10.1093/neuonc/noab106
11. Capper D, Reifenberger G, French PJ, et al. EANO guideline on rational molecular testing of gliomas, glioneuronal and neuronal tumors in adults for targeted therapy selection. Neuro Oncol. Jan 12 2023;doi:10.1093/neuonc/noad008
12. Radzikowska U, Baerenfaller K, Cornejo-Garcia JA, et al. Omics technologies in allergy and asthma research: An EAACI position paper. Allergy. Oct 2022;77(10):2888-2908. doi:10.1111/all.15412
13. Wanka L, Jappe U. Trained immunity and allergy: State of the art and future perspectives. Allergy. Apr 2021;76(4):1265-1267. doi:10.1111/all.14617
14. Zissler UM, Chaker AM, Effner R, et al. Interleukin-4 and interferon-gamma orchestrate an epithelial polarization in the airways. Mucosal Immunol. Jul 2016;9(4):917-26. doi:10.1038/mi.2015.110
15. Akdis CA, Arkwright PD, Bruggen MC, et al. Type 2 immunity in the skin and lungs. Allergy. Jul 2020;75(7):1582-1605. doi:10.1111/all.14318
16. Romagnani S. Immunologic influences on allergy and the TH1/TH2 balance. J Allergy Clin Immunol. Mar 2004;113(3):395-400. doi:10.1016/j.jaci.2003.11.025
17. Barber D, Diaz-Perales A, Escribese MM, et al. Molecular allergology and its impact in specific allergy diagnosis and therapy. Allergy. Dec 2021;76(12):3642-3658. doi:10.1111/all.14969
18. Duhring L, Petry J, Lilienthal GM, et al. Sialylation of IgE reduces FcepsilonRIalpha interaction and mast cell and basophil activation in vitro and increases IgE half-life in vivo. Allergy. Aug 2023;78(8):2301-2305. doi:10.1111/all.15665
19. Locke A, Hung L, Upton JEM, O'Mahony L, Hoang J, Eiwegger T. An update on recent developments and highlights in food allergy. Allergy. Apr 23 2023;doi:10.1111/all.15749
20. Worm M, Reese I, Ballmer-Weber B, et al. Update of the S2k guideline on the management of IgE-mediated food allergies. Allergol Select. 2021;5:195-243. doi:10.5414/ALX02257E
21. Behrends J, Schwager C, Hein M, Scholzen T, Kull S, Jappe U. Innovative robust basophil activation test using a novel gating strategy reliably diagnosing allergy with full automation. Allergy. Dec 2021;76(12):3776-3788. doi:10.1111/all.14900
22. Mehlich J, Fischer J, Hilger C, et al. The basophil activation test differentiates between patients with alpha-gal syndrome and asymptomatic alpha-gal sensitization. J Allergy Clin Immunol. Jan 2019;143(1):182-189. doi:10.1016/j.jaci.2018.06.049
23. Schwager C, Kull S, Behrends J, et al. Peanut oleosins associated with severe peanut allergy-importance of lipophilic allergens for comprehensive allergy diagnostics. J Allergy Clin Immunol. Nov 2017;140(5):1331-1338 e8. doi:10.1016/j.jaci.2017.02.020
24. Bax HJ, Khiabany A, Stavraka C, et al. Basophil activation test in cancer patient blood evaluating potential hypersensitivity to an anti-tumor IgE therapeutic candidate. Allergy. Aug 2020;75(8):2069-2073. doi:10.1111/all.14245
25. Pascal M, Moreno C, Davila I, et al. Integration of in vitro allergy test results and ratio analysis for the diagnosis and treatment of allergic patients (INTEGRA). Clin Transl Allergy. Aug 2021;11(7):e12052. doi:10.1002/clt2.12052
26. Sala-Cunill A, Cardona V, Labrador-Horrillo M, et al. Usefulness and limitations of sequential serum tryptase for the diagnosis of anaphylaxis in 102 patients. Int Arch Allergy Immunol. 2013;160(2):192-9. doi:10.1159/000339749
27. Muraro A, Worm M, Alviani C, et al. EAACI guidelines: Anaphylaxis (2021 update). Allergy. Feb 2022;77(2):357-377. doi:10.1111/all.15032
28. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. May 4 2012;18(5):716-25. doi:10.1038/nm.2678
29. Trivedi M, Denton E. Asthma in Children and Adults-What Are the Differences and What Can They Tell us About Asthma? Front Pediatr. 2019;7:256. doi:10.3389/fped.2019.00256
30. Gulsen A, Wedi B, Jappe U. Hypersensitivity reactions to biologics (part I): allergy as an important differential diagnosis in complex immune-derived adverse events. Allergo J Int. 2020;29(4):97-125. doi:10.1007/s40629-020-00126-6
31. Wang J, Zhou Y, Zhang H, et al. Pathogenesis of allergic diseases and implications for therapeutic interventions. Signal Transduct Target Ther. Mar 24 2023;8(1):138. doi:10.1038/s41392-023-01344-4
32. Diamant Z, Vijverberg S, Alving K, et al. Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy. Oct 2019;74(10):1835-1851. doi:10.1111/all.13806
33. Hearn AP, Kavanagh J, d'Ancona G, et al. The relationship between Feno and effectiveness of mepolizumab and benralizumab in severe eosinophilic asthma. J Allergy Clin Immunol Pract. May 2021;9(5):2093-2096 e1. doi:10.1016/j.jaip.2021.01.008
34. Palikhe NS, Laratta C, Nahirney D, et al. Elevated levels of circulating CD4(+) CRTh2(+) T cells characterize severe asthma. Clin Exp Allergy. Jun 2016;46(6):825-36. doi:10.1111/cea.12741
35. Huang Z, Chu M, Chen X, et al. Th2A cells: The pathogenic players in allergic diseases. Front Immunol. 2022;13:916778. doi:10.3389/fimmu.2022.916778
36. An J, Lee J-H, Kang Y, et al. Clinical significance of serum MRGPRX2 as a new biomarker in allergic asthma. European Respiratory Journal. 2019;54(suppl 63):PA4263. doi:10.1183/13993003.congress-2019.PA4263
37. Cao TBT, Cha HY, Yang EM, Ye YM. Elevated MRGPRX2 Levels Related to Disease Severity in Patients With Chronic Spontaneous Urticaria. Allergy Asthma Immunol Res. May 2021;13(3):498-506. doi:10.4168/aair.2021.13.3.498
38. Sabato V, Ebo DG, Van Der Poorten MM, et al. Allergenic and Mas-Related G Protein-Coupled Receptor X2-Activating Properties of Drugs: Resolving the Two. J Allergy Clin Immunol Pract. Feb 2023;11(2):395-404. doi:10.1016/j.jaip.2022.12.014
39. Bahri R, Custovic A, Korosec P, et al. Mast cell activation test in the diagnosis of allergic disease and anaphylaxis. J Allergy Clin Immunol. Aug 2018;142(2):485-496 e16. doi:10.1016/j.jaci.2018.01.043
40. Zheng H, Zhang Y, Pan J, et al. The Role of Type 2 Innate Lymphoid Cells in Allergic Diseases. Front Immunol. 2021;12:586078. doi:10.3389/fimmu.2021.586078
41. Jakwerth CA, Chaker AM, Guerth F, et al. Sputum microRNA-screening reveals Prostaglandin EP3 receptor as selective target in allergen-specific immunotherapy. Clin Exp Allergy. Dec 2021;51(12):1577-1591. doi:10.1111/cea.14013
42. Radonjic-Hoesli S, Pavlov N, Simon HU, Simon D. Are blood cytokines reliable biomarkers of allergic disease diagnosis and treatment responses? J Allergy Clin Immunol. Aug 2022;150(2):251-258. doi:10.1016/j.jaci.2022.06.008
43. Breiteneder H, Peng YQ, Agache I, et al. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy. Dec 2020;75(12):3039-3068. doi:10.1111/all.14582
44. Izuhara K, Nunomura S, Nanri Y, Ono J, Takai M, Kawaguchi A. Periostin: An emerging biomarker for allergic diseases. Allergy. Nov 2019;74(11):2116-2128. doi:10.1111/all.13814
45. Roth-Walter F, Schmutz R, Mothes-Luksch N, et al. Clinical efficacy of sublingual immunotherapy is associated with restoration of steady-state serum lipocalin 2 after SLIT: a pilot study. World Allergy Organ J. 2018;11(1):21. doi:10.1186/s40413-018-0201-8
46. Marchica CL, Pinelli V, Borges M, et al. A role for decorin in a murine model of allergen-induced asthma. Am J Physiol Lung Cell Mol Physiol. Jun 2011;300(6):L863-73. doi:10.1152/ajplung.00300.2009
47. Delgado-Dolset MI, Obeso D, Rodriguez-Coira J, et al. Understanding uncontrolled severe allergic asthma by integration of omic and clinical data. Allergy. Jun 2022;77(6):1772-1785. doi:10.1111/all.15192
48. Miller M, Broide DH. Why Is ORMDL3 on Chromosome 17q21 Highly Linked to Asthma? Am J Respir Crit Care Med. Feb 15 2019;199(4):404-406. doi:10.1164/rccm.201810-1941ED
49. Gao W, Gong J, Mu M, et al. The Pathogenesis of Eosinophilic Asthma: A Positive Feedback Mechanism That Promotes Th2 Immune Response via Filaggrin Deficiency. Front Immunol. 2021;12:672312. doi:10.3389/fimmu.2021.672312
50. Zhu AY, Mitra N, Margolis DJ. Longitudinal association of atopic dermatitis progression and keratin 6A. Sci Rep. Aug 10 2022;12(1):13629. doi:10.1038/s41598-022-17946-x
51. Sordillo JE, Lutz SM, Jorgenson E, et al. A polygenic risk score for asthma in a large racially diverse population. Clin Exp Allergy. Nov 2021;51(11):1410-1420. doi:10.1111/cea.14007
52. Young RP, Dekker JW, Wordsworth BP, et al. HLA-DR and HLA-DP genotypes and immunoglobulin E responses to common major allergens. Clin Exp Allergy. May 1994;24(5):431-9. doi:10.1111/j.1365-2222.1994.tb00931.x
53. Zhu Z, Lee PH, Chaffin MD, et al. A genome-wide cross-trait analysis from UK Biobank highlights the shared genetic architecture of asthma and allergic diseases. Nat Genet. Jun 2018;50(6):857-864. doi:10.1038/s41588-018-0121-0
54. Demoly P, Adkinson NF, Brockow K, et al. International Consensus on drug allergy. Allergy. Apr 2014;69(4):420-37. doi:10.1111/all.12350
55. Waage J, Standl M, Curtin JA, et al. Genome-wide association and HLA fine-mapping studies identify risk loci and genetic pathways underlying allergic rhinitis. Nat Genet. Aug 2018;50(8):1072-1080. doi:10.1038/s41588-018-0157-1
56. Weidner J, Bartel S, Kilic A, et al. Spotlight on microRNAs in allergy and asthma. Allergy. Jun 2021;76(6):1661-1678. doi:10.1111/all.14646
57. Wang Z, He Y, Cun Y, Li Q, Zhao Y, Luo Z. Transcriptomic analysis identified SLC40A1 as a key iron metabolism-related gene in airway macrophages in childhood allergic asthma. Front Cell Dev Biol. 2023;11:1164544. doi:10.3389/fcell.2023.1164544
58. Zissler UM, Jakwerth CA, Guerth FM, et al. Early IL-10 producing B-cells and coinciding Th/Tr17 shifts during three year grass-pollen AIT. EBioMedicine. Oct 2018;36:475-488. doi:10.1016/j.ebiom.2018.09.016
59. Bozek A, Cudak A, Walter Canonica G. Long-term efficacy of injected allergen immunotherapy for treatment of grass pollen allergy in elderly patients with allergic rhinitis. Allergy Asthma Proc. Jul 1 2020;41(4):271-277. doi:10.2500/aap.2020.41.200035
60. Rodriguez-Dominguez A, Berings M, Rohrbach A, et al. Molecular profiling of allergen-specific antibody responses may enhance success of specific immunotherapy. J Allergy Clin Immunol. Nov 2020;146(5):1097-1108. doi:10.1016/j.jaci.2020.03.029
61. Gulsen A, Wallis S, Jappe U. Combination of immunotherapies for severe allergic asthma. J Asthma. Jan 2021;58(1):75-78. doi:10.1080/02770903.2019.1658204
62. Pagani M, Bavbek S, Alvarez-Cuesta E, et al. Hypersensitivity reactions to chemotherapy: an EAACI Position Paper. Allergy. Feb 2022;77(2):388-403. doi:10.1111/all.15113
63. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. Jun 2016;131(6):803-20. doi:10.1007/s00401-016-1545-1
64. Wesseling P, Capper D. WHO 2016 Classification of gliomas. Neuropathol Appl Neurobiol. Feb 2018;44(2):139-150. doi:10.1111/nan.12432
65. Brat DJ, Aldape K, Colman H, et al. cIMPACT-NOW update 3: recommended diagnostic criteria for "Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV". Acta Neuropathol. Nov 2018;136(5):805-810. doi:10.1007/s00401-018-1913-0
66. Brat DJ, Aldape K, Colman H, et al. cIMPACT-NOW update 5: recommended grading criteria and terminologies for IDH-mutant astrocytomas. Acta Neuropathol. Mar 2020;139(3):603-608. doi:10.1007/s00401-020-02127-9
67. Weller M, van den Bent M, Preusser M, et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. Mar 2021;18(3):170-186. doi:10.1038/s41571-020-00447-z
68. Wang Q, Hu B, Hu X, et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell. Jul 10 2017;32(1):42-56 e6. doi:10.1016/j.ccell.2017.06.003
69. Neftel C, Laffy J, Filbin MG, et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell. Aug 8 2019;178(4):835-849 e21. doi:10.1016/j.cell.2019.06.024
70. Suva ML, Tirosh I. The Glioma Stem Cell Model in the Era of Single-Cell Genomics. Cancer Cell. May 11 2020;37(5):630-636. doi:10.1016/j.ccell.2020.04.001
71. Barthel FP, Johnson KC, Varn FS, et al. Longitudinal molecular trajectories of diffuse glioma in adults. Nature. Dec 2019;576(7785):112-120. doi:10.1038/s41586-019-1775-1
72. Varn FS, Johnson KC, Martinek J, et al. Glioma progression is shaped by genetic evolution and microenvironment interactions. Cell. Jun 9 2022;185(12):2184-2199 e16. doi:10.1016/j.cell.2022.04.038
73. Eckel-Passow JE, Lachance DH, Decker PA, et al. Inherited genetics of adult diffuse glioma and polygenic risk scores-a review. Neurooncol Pract. Aug 2022;9(4):259-270. doi:10.1093/nop/npac017
74. White K, Connor K, Meylan M, et al. Identification, validation and biological characterisation of novel glioblastoma tumour microenvironment subtypes: implications for precision immunotherapy. Ann Oncol. Mar 2023;34(3):300-314. doi:10.1016/j.annonc.2022.11.008
75. Sharma P, Aaroe A, Liang J, Puduvalli VK. Tumor microenvironment in glioblastoma: Current and emerging concepts. Neurooncol Adv. Jan-Dec 2023;5(1):vdad009. doi:10.1093/noajnl/vdad009
76. Lin C, Wang N, Xu C. Glioma-associated microglia/macrophages (GAMs) in glioblastoma: Immune function in the tumor microenvironment and implications for immunotherapy. Front Immunol. 2023;14:1123853. doi:10.3389/fimmu.2023.1123853
77. Thorsson V, Gibbs DL, Brown SD, et al. The Immune Landscape of Cancer. Immunity. Apr 17 2018;48(4):812-830 e14. doi:10.1016/j.immuni.2018.03.023
78. Berghoff AS, Kiesel B, Widhalm G, et al. Correlation of immune phenotype with IDH mutation in diffuse glioma. Neuro Oncol. Oct 19 2017;19(11):1460-1468. doi:10.1093/neuonc/nox054
79. Bunse L, Pusch S, Bunse T, et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat Med. Aug 2018;24(8):1192-1203. doi:10.1038/s41591-018-0095-6
80. Berghoff AS, Kiesel B, Widhalm G, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. Aug 2015;17(8):1064-75. doi:10.1093/neuonc/nou307
81. Cavalheiro VJ, Campos ACP, Lima L, et al. Unraveling the peripheral and local role of inflammatory cytokines in glioblastoma survival. Cytokine. Jan 2023;161:156059. doi:10.1016/j.cyto.2022.156059
82. Gohar MK, Ammar MG, Alnagar AA, Abd-ElAziz HA. Serum IgE and Allergy Related Genotypes of IL-4R α and IL-13 Genes: Association with Glioma Susceptibility and Glioblastoma Prognosis. Egypt J Immunol. Jan 2018;25(1):19-33.
83. Zhang W, He Y, Kang X, et al. Association between dietary minerals and glioma: A case-control study based on Chinese population. Front Nutr. 2023;10:1118997. doi:10.3389/fnut.2023.1118997
84. Hu X, Deng Q, Ma L, et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. Mar 2020;30(3):229-243. doi:10.1038/s41422-020-0287-8
85. Song E, Mao T, Dong H, et al. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature. Jan 2020;577(7792):689-694. doi:10.1038/s41586-019-1912-x
86. Louveau A, Smirnov I, Keyes TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. Jul 16 2015;523(7560):337-41. doi:10.1038/nature14432
87. Castellani G, Croese T, Peralta Ramos JM, Schwartz M. Transforming the understanding of brain immunity. Science. Apr 7 2023;380(6640):eabo7649. doi:10.1126/science.abo7649
88. Mellinghoff IK, van den Bent MJ, Blumenthal DT, et al. Vorasidenib in IDH1- or IDH2-Mutant Low-Grade Glioma. N Engl J Med. Jun 4 2023;doi:10.1056/NEJMoa2304194
89. Brandes AA, Carpentier AF, Kesari S, et al. A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. Aug 2016;18(8):1146-56. doi:10.1093/neuonc/now009
90. Reardon DA, Brandes AA, Omuro A, et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. Jul 1 2020;6(7):1003-1010. doi:10.1001/jamaoncol.2020.1024
91. Lim M, Weller M, Idbaih A, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. Nov 2 2022;24(11):1935-1949. doi:10.1093/neuonc/noac116
92. Omuro A, Brandes AA, Carpentier AF, et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro Oncol. Jan 5 2023;25(1):123-134. doi:10.1093/neuonc/noac099
93. Cloughesy TF, Mochizuki AY, Orpilla JR, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. Mar 2019;25(3):477-486. doi:10.1038/s41591-018-0337-7
94. Weller M, Butowski N, Tran DD, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. Oct 2017;18(10):1373-1385. doi:10.1016/S1470-2045(17)30517-X
95. Wen PY, Reardon DA, Armstrong TS, et al. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin Cancer Res. Oct 1 2019;25(19):5799-5807. doi:10.1158/1078-0432.CCR-19-0261
96. Liau LM, Ashkan K, Brem S, et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. Jan 1 2023;9(1):112-121. doi:10.1001/jamaoncol.2022.5370
97. Weiss T, Puca E, Silginer M, et al. Immunocytokines are a promising immunotherapeutic approach against glioblastoma. Sci Transl Med. Oct 7 2020;12(564)doi:10.1126/scitranslmed.abb2311
98. Wang G, Xu S, Cao C, et al. Evidence from a large-scale meta-analysis indicates eczema reduces the incidence of glioma. Oncotarget. Sep 20 2016;7(38):62598-62606. doi:10.18632/oncotarget.11545
99. Zhang C, Zhu QX. Allergy is associated with reduced risk of glioma: A meta-analysis. Allergol Immunopathol (Madr). Nov - Dec 2017;45(6):553-559. doi:10.1016/j.aller.2016.12.005
100. Zhu Y, Teng Y, Xu S, et al. Eczema as a protective factor for brain cancer: a meta-analysis. BMC Cancer. Dec 29 2022;22(1):1360. doi:10.1186/s12885-022-10471-0
101. Johansen C, Schuz J, Andreasen AS, Dalton SO. Study designs may influence results: the problems with questionnaire-based case-control studies on the epidemiology of glioma. Br J Cancer. Mar 28 2017;116(7):841-848. doi:10.1038/bjc.2017.46
102. Cahoon EK, Inskip PD, Gridley G, Brenner AV. Immune-related conditions and subsequent risk of brain cancer in a cohort of 4.5 million male US veterans. Br J Cancer. Apr 02 2014;110(7):1825-33. doi:10.1038/bjc.2014.97
103. Mansfield KE, Schmidt SAJ, Darvalics B, et al. Association Between Atopic Eczema and Cancer in England and Denmark. JAMA Dermatol. Oct 1 2020;156(10):1086-1097. doi:10.1001/jamadermatol.2020.1948
104. Liu X, Hemminki K, Forsti A, Sundquist J, Sundquist K, Ji J. Cancer risk and mortality in asthma patients: A Swedish national cohort study. Acta Oncol. 2015;54(8):1120-7. doi:10.3109/0284186X.2014.1001497
105. He MM, Lo CH, Wang K, et al. Immune-Mediated Diseases Associated With Cancer Risks. JAMA Oncol. Feb 1 2022;8(2):209-219. doi:10.1001/jamaoncol.2021.5680
106. Krishnamachari B, Il'yasova D, Scheurer ME, et al. A pooled multisite analysis of the effects of atopic medical conditions in glioma risk in different ethnic groups. Ann Epidemiol. Apr 2015;25(4):270-4. doi:10.1016/j.annepidem.2014.12.007
107. Lupatsch JE, Bailey HD, Lacour B, et al. Childhood brain tumours, early infections and immune stimulation: A pooled analysis of the ESCALE and ESTELLE case-control studies (SFCE, France). Cancer epidemiology. Feb 2018;52:1-9. doi:10.1016/j.canep.2017.10.015
108. Calboli FC, Cox DG, Buring JE, et al. Prediagnostic plasma IgE levels and risk of adult glioma in four prospective cohort studies. Journal of the National Cancer Institute. Nov 2 2011;103(21):1588-95. doi:10.1093/jnci/djr361
109. Schlehofer B, Siegmund B, Linseisen J, et al. Primary brain tumours and specific serum immunoglobulin E: a case-control study nested in the European Prospective Investigation into Cancer and Nutrition cohort. Allergy. Nov 2011;66(11):1434-41. doi:10.1111/j.1398-9995.2011.02670.x
110. Schwartzbaum J, Ding B, Johannesen TB, et al. Association between prediagnostic IgE levels and risk of glioma. Journal of the National Cancer Institute. Aug 22 2012;104(16):1251-9. doi:10.1093/jnci/djs315
111. Amirian ES, Marquez-Do D, Bondy ML, Scheurer ME. Antihistamine use and immunoglobulin E levels in glioma risk and prognosis. Cancer epidemiology. Dec 2013;37(6):908-12. doi:10.1016/j.canep.2013.08.004
112. Dunford PJ, O'Donnell N, Riley JP, Williams KN, Karlsson L, Thurmond RL. The histamine H4 receptor mediates allergic airway inflammation by regulating the activation of CD4+ T cells. J Immunol. Jun 1 2006;176(11):7062-70. doi:10.4049/jimmunol.176.11.7062
113. Morgan RK, McAllister B, Cross L, et al. Histamine 4 receptor activation induces recruitment of FoxP3+ T cells and inhibits allergic asthma in a murine model. J Immunol. Jun 15 2007;178(12):8081-9. doi:10.4049/jimmunol.178.12.8081
114. Schwartzbaum J, Seweryn M, Holloman C, et al. Association between Prediagnostic Allergy-Related Serum Cytokines and Glioma. PLoS One. 2015;10(9):e0137503. doi:10.1371/journal.pone.0137503
115. Scheurer ME, El-Zein R, Thompson PA, et al. Long-term anti-inflammatory and antihistamine medication use and adult glioma risk. Cancer Epidemiol Biomarkers Prev. May 2008;17(5):1277-81. doi:10.1158/1055-9965.EPI-07-2621
116. Saleh M, Wiegmans A, Malone Q, Stylli SS, Kaye AH. Effect of in situ retroviral interleukin-4 transfer on established intracranial tumors. Journal of the National Cancer Institute. Mar 3 1999;91(5):438-45. doi:10.1093/jnci/91.5.438
117. Tepper RI, Coffman RL, Leder P. An eosinophil-dependent mechanism for the antitumor effect of interleukin-4. Science. Jul 24 1992;257(5069):548-51. doi:10.1126/science.1636093
118. Okada H, Villa L, Attanucci J, et al. Cytokine gene therapy of gliomas: effective induction of therapeutic immunity to intracranial tumors by peripheral immunization with interleukin-4 transduced glioma cells. Gene Ther. Aug 2001;8(15):1157-66. doi:10.1038/sj.gt.3301496
119. Ma C, Cao L, Zhao J, et al. Inverse Association between Prediagnostic IgE Levels and the Risk of Brain Tumors: A Systematic Review and Meta-Analysis. Biomed Res Int. 2015;2015:294213. doi:10.1155/2015/294213
120. Zhou M, Bracci PM, McCoy LS, et al. Serum macrophage-derived chemokine/CCL22 levels are associated with glioma risk, CD4 T cell lymphopenia and survival time. Int J Cancer. Aug 15 2015;137(4):826-36. doi:10.1002/ijc.29441
121. Schwartzbaum J, Wang M, Root E, et al. A nested case-control study of 277 prediagnostic serum cytokines and glioma. PLoS One. 2017;12(6):e0178705. doi:10.1371/journal.pone.0178705
122. Chen P, Chen C, Chen K, Xu T, Luo C. Polymorphisms in IL-4/IL-13 pathway genes and glioma risk: an updated meta-analysis. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. Jan 2015;36(1):121-7. doi:10.1007/s13277-014-2895-8
123. Choi SS, Choi H, Baek IC, et al. HLA polymorphisms and risk of glioblastoma in Koreans. PLoS One. 2021;16(12):e0260618. doi:10.1371/journal.pone.0260618
124. Eckel-Passow JE, Drucker KL, Kollmeyer TM, et al. Adult diffuse glioma GWAS by molecular subtype identifies variants in D2HGDH and FAM20C. Neuro Oncol. Nov 26 2020;22(11):1602-1613. doi:10.1093/neuonc/noaa117
125. Disney-Hogg L, Cornish AJ, Sud A, et al. Impact of atopy on risk of glioma: a Mendelian randomisation study. BMC Med. Mar 15 2018;16(1):42. doi:10.1186/s12916-018-1027-5
126. Howell AE, Robinson JW, Wootton RE, et al. Testing for causality between systematically identified risk factors and glioma: a Mendelian randomization study. BMC Cancer. Jun 3 2020;20(1):508. doi:10.1186/s12885-020-06967-2
127. Saunders CN, Cornish AJ, Kinnersley B, et al. Lack of association between modifiable exposures and glioma risk: a Mendelian randomization analysis. Neuro Oncol. Feb 20 2020;22(2):207-215. doi:10.1093/neuonc/noz209
128. Ostrom QT, Edelson J, Byun J, et al. Partitioned glioma heritability shows subtype-specific enrichment in immune cells. Neuro Oncol. Mar 20 2021;doi:10.1093/neuonc/noab072
129. Lehrer S, Rheinstein PH, Rosenzweig KE. Allergy may confer better survival on patients with gliomas. Clin Neurol Neurosurg. Feb 2019;177:63-67. doi:10.1016/j.clineuro.2018.12.021
130. Jaman E, Zhang X, Sandlesh P, et al. History of atopy confers improved outcomes in IDH mutant and wildtype lower grade gliomas. Journal of neuro-oncology. Nov 2021;155(2):133-141. doi:10.1007/s11060-021-03854-z
131. Carr MT, Hochheimer CJ, Rock AK, et al. Comorbid Medical Conditions as Predictors of Overall Survival in Glioblastoma Patients. Sci Rep. Dec 27 2019;9(1):20018. doi:10.1038/s41598-019-56574-w
132. Han S, Huang Y, Wang Z, Li Z, Qin X, Wu A. Increased rate of positive penicillin skin tests among patients with glioma: insights into the association between allergies and glioma risk. J Neurosurg. Nov 2014;121(5):1176-84. doi:10.3171/2014.7.JNS1412
133. Wrensch M, Wiencke JK, Wiemels J, et al. Serum IgE, tumor epidermal growth factor receptor expression, and inherited polymorphisms associated with glioma survival. Cancer research. Apr 15 2006;66(8):4531-41. doi:10.1158/0008-5472.CAN-05-4032
134. Lin Y, Jin Q, Zhang GZ, et al. Increase of plasma IgE during treatment correlates with better outcome of patients with glioblastoma. Chinese medical journal. Oct 2011;124(19):3042-8.
135. Scheurer ME, Amirian E, Cao Y, et al. Polymorphisms in the interleukin-4 receptor gene are associated with better survival in patients with glioblastoma. Clin Cancer Res. Oct 15 2008;14(20):6640-6. doi:10.1158/1078-0432.CCR-07-4681
136. Gousias K, Markou M, Arzoglou V, et al. Frequent abnormalities of the immune system in gliomas and correlation with the WHO grading system of malignancy. J Neuroimmunol. Sep 14 2010;226(1-2):136-42. doi:10.1016/j.jneuroim.2010.05.027
137. De Boeck A, Ahn BY, D'Mello C, et al. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun. Oct 5 2020;11(1):4997. doi:10.1038/s41467-020-18569-4
138. Zhang Z, Huang X, Li J, et al. Interleukin 10 promotes growth and invasion of glioma cells by up-regulating KPNA 2 in vitro. J Cancer Res Ther. 2019;15(4):927-932. doi:10.4103/jcrt.JCRT_284_19
139. Takashima Y, Kawaguchi A, Kanayama T, Hayano A, Yamanaka R. Correlation between lower balance of Th2 helper T-cells and expression of PD-L1/PD-1 axis genes enables prognostic prediction in patients with glioblastoma. Oncotarget. Apr 10 2018;9(27):19065-19078. doi:10.18632/oncotarget.24897
140. Madhugiri VS, Venkatesan S, Dutt A, et al. An Analysis of Eosinophil- and Basophil-Based Indices in Patients with Glioblastoma and their Correlation with Survival. World Neurosurg. Nov 9 2022;doi:10.1016/j.wneu.2022.11.008
141. Zhang X, Li C, Xiao L, et al. Predicting Individual Prognosis and Grade of Patients with Glioma Based on Preoperative Eosinophil and Neutrophil-to-Lymphocyte Ratio. Cancer Manag Res. 2020;12:5793-5802. doi:10.2147/cmar.S260695
142. Zhong QY, Fan EX, Feng GY, et al. A gene expression-based study on immune cell subtypes and glioma prognosis. BMC Cancer. Nov 15 2019;19(1):1116. doi:10.1186/s12885-019-6324-7
143. Vaios EJ, Winter SF, Muzikansky A, Nahed BV, Dietrich J. Eosinophil and lymphocyte counts predict bevacizumab response and survival in recurrent glioblastoma. Neuro-Oncology Advances. 2020;2(1):vdaa031. doi:10.1093/noajnl/vdaa031
144. Madhugiri VS, Moiyadi AV, Shetty P, et al. Analysis of Factors Associated with Long-Term Survival in Patients with Glioblastoma. World Neurosurg. May 2021;149:e758-e765. doi:10.1016/j.wneu.2021.01.103
145. Huang Z, Wu L, Hou Z, Zhang P, Li G, Xie J. Eosinophils and other peripheral blood biomarkers in glioma grading: a preliminary study. BMC Neurol. Dec 5 2019;19(1):313. doi:10.1186/s12883-019-1549-2
146. Zheng L, Yu M, Zhang S. Prognostic value of pretreatment circulating basophils in patients with glioblastoma. Neurosurg Rev. Dec 2021;44(6):3471-3478. doi:10.1007/s10143-021-01524-2
147. Chen R, Wu W, Liu T, et al. Large-scale bulk RNA-seq analysis defines immune evasion mechanism related to mast cell in gliomas. Front Immunol. 2022;13:914001. doi:10.3389/fimmu.2022.914001
148. Põlajeva J, Sjösten AM, Lager N, et al. Mast cell accumulation in glioblastoma with a potential role for stem cell factor and chemokine CXCL12. PLoS One. 2011;6(9):e25222. doi:10.1371/journal.pone.0025222
149. Cernadas J, Vasconcelos MJ, Fernandes AP, Carneiro-Leao L, Gil-da-Costa MJ. Desensitization to carboplatin in low-grade glioma. A revision of 100 treatments in children. Pediatr Allergy Immunol. Aug 2021;32(6):1388-1391. doi:10.1111/pai.13525
150. Chatterjee J, Sanapala S, Cobb O, et al. Asthma reduces glioma formation by T cell decorin-mediated inhibition of microglia. Nat Commun. Dec 8 2021;12(1):7122. doi:10.1038/s41467-021-27455-6
151. Poli A, Oudin A, Muller A, et al. Allergic Airway Inflammation Delays Glioblastoma Progression and Reinvigorates Systemic and Local Immunity in Mice. Allergy. Oct 9 2022;doi:10.1111/all.15545
152. Wu H, Eckhardt CM, Baccarelli AA. Molecular mechanisms of environmental exposures and human disease. Nat Rev Genet. May 2023;24(5):332-344. doi:10.1038/s41576-022-00569-3
153. Jordakieva G, Bianchini R, Reichhold D, et al. IgG4 induces tolerogenic M2-like macrophages and correlates with disease progression in colon cancer. Oncoimmunology. Feb 8 2021;10(1):1880687. doi:10.1080/2162402X.2021.1880687
154. Li H, Xiao Y, Li Q, et al. The allergy mediator histamine confers resistance to immunotherapy in cancer patients via activation of the macrophage histamine receptor H1. Cancer Cell. Jan 10 2022;40(1):36-52 e9. doi:10.1016/j.ccell.2021.11.002
156. Kabata H, Artis D. Neuro-immune crosstalk and allergic inflammation. J Clin Invest. Mar 4 2019;129(4):1475-1482. doi:10.1172/JCI124609
157. Pan Y, Monje M. Neuron-Glial Interactions in Health and Brain Cancer. Adv Biol (Weinh). Sep 2022;6(9):e2200122. doi:10.1002/adbi.202200122
158. Mollgard K, Beinlich FRM, Kusk P, et al. A mesothelium divides the subarachnoid space into functional compartments. Science. Jan 6 2023;379(6627):84-88. doi:10.1126/science.adc8810
159. Mamuladze T, Kipnis J. Type 2 immunity in the brain and brain borders. Cell Mol Immunol. Jul 10 2023;doi:10.1038/s41423-023-01043-8
160. Li S, Olde Heuvel F, Rehman R, et al. Interleukin-13 and its receptor are synaptic proteins involved in plasticity and neuroprotection. Nat Commun. Jan 13 2023;14(1):200. doi:10.1038/s41467-023-35806-8
161. Hanuscheck N, Thalman C, Domingues M, et al. Interleukin-4 receptor signaling modulates neuronal network activity. J Exp Med. Jun 6 2022;219(6)doi:10.1084/jem.20211887
162. Costanza M. Type 2 Inflammatory Responses in Autoimmune Demyelination of the Central Nervous System: Recent Advances. J Immunol Res. 2019;2019:4204512. doi:10.1155/2019/4204512
163. Plum T, Binzberger R, Thiele R, et al. Mast cells link immune sensing to antigen-avoidance behaviour. Nature. Aug 2023;620(7974):634-642. doi:10.1038/s41586-023-06188-0
164. Florsheim EB, Bachtel ND, Cullen JL, et al. Immune sensing of food allergens promotes avoidance behaviour. Nature. Aug 2023;620(7974):643-650. doi:10.1038/s41586-023-06362-4
165. Zhou L, Chen L, Li X, Li T, Dong Z, Wang YT. Food allergy induces alteration in brain inflammatory status and cognitive impairments. Behav Brain Res. May 17 2019;364:374-382. doi:10.1016/j.bbr.2018.01.011
166. Tonelli LH, Katz M, Kovacsics CE, et al. Allergic rhinitis induces anxiety-like behavior and altered social interaction in rodents. Brain Behav Immun. Aug 2009;23(6):784-93. doi:10.1016/j.bbi.2009.02.017
167. Sarlus H, Hoglund CO, Karshikoff B, et al. Allergy influences the inflammatory status of the brain and enhances tau-phosphorylation. J Cell Mol Med. Oct 2012;16(10):2401-12. doi:10.1111/j.1582-4934.2012.01556.x
168. Sarlus H, Eyjolfsdottir H, Eriksdotter M, Oprica M, Schultzberg M. Influence of Allergy on Immunoglobulins and Amyloid-beta in the Cerebrospinal Fluid of Patients with Alzheimer's Disease. J Alzheimers Dis. 2015;48(2):495-505. doi:10.3233/JAD-143147
169. Klein B, Mrowetz H, Thalhamer J, Scheiblhofer S, Weiss R, Aigner L. Allergy Enhances Neurogenesis and Modulates Microglial Activation in the Hippocampus. Front Cell Neurosci. 2016;10:169. doi:10.3389/fncel.2016.00169
170. Yamasaki R, Fujii T, Wang B, et al. Allergic Inflammation Leads to Neuropathic Pain via Glial Cell Activation. J Neurosci. Nov 23 2016;36(47):11929-11945. doi:10.1523/JNEUROSCI.1981-16.2016
171. Peng X, Madany AM, Jang JC, et al. Continuous Inhalation Exposure to Fungal Allergen Particulates Induces Lung Inflammation While Reducing Innate Immune Molecule Expression in the Brainstem. ASN Neuro. Jan-Dec 2018;10:1759091418782304. doi:10.1177/1759091418782304
172. Vogel Ciernia A, Careaga M, LaSalle JM, Ashwood P. Microglia from offspring of dams with allergic asthma exhibit epigenomic alterations in genes dysregulated in autism. Glia. Mar 2018;66(3):505-521. doi:10.1002/glia.23261
173. Gao Z, Chen X, Xiang R, et al. Changes in Resting-State Spontaneous Brain Activity in Patients With Allergic Rhinitis: A Pilot Neuroimaging Study. Front Neurosci. 2021;15:697299. doi:10.3389/fnins.2021.697299
174. Callebaut I, Steelant B, Backaert W, et al. Brain activation after nasal histamine provocation in house dust mite allergic rhinitis patients. Allergy. Jun 2021;76(6):1879-1882. doi:10.1111/all.14677