ACTUAL CONCEPTION OF ALLERGEN-SPECIFIC IMMUNOTHERAPY MECHANISMS, POTENTIAL BIOMARKERS OF EFFICACY AND WAYS OF ENHANCEMENT



Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

The article analyzes international position papers on the allergen-specific immunotherapy mechanisms, discusses potential biomarkers for evaluation of ASIT efficacy, as well as the perspectives for ASIT enhancement.

Full Text

Restricted Access

About the authors

K S Pavlova

NRC Institute of Immunology FMBA of Russia

24, Kashirskoe Shosse, Moscow, 115478, Russia

O M Kurbacheva

NRC Institute of Immunology FMBA of Russia

Email: kurbacheva@gmail.com
24, Kashirskoe Shosse, Moscow, 115478, Russia

M A Galitskaya

NRC Institute of Immunology FMBA of Russia

24, Kashirskoe Shosse, Moscow, 115478, Russia

D S Smirnov

NRC Institute of Immunology FMBA of Russia

24, Kashirskoe Shosse, Moscow, 115478, Russia

References

  1. Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol. 2007;(119):780-791. doi: 10.1016/j.jaci.2007.01.022.
  2. Akdis M. Healthy immune response to allergens: T-regulatory cells and more. Curr Opin Immunol. 2006;(18):738-744. doi: 10.1016/j.coi.2006.06.003.
  3. Larche M, Akdis CA, Valenta R. Immunological mechanisms of allergen-specific immunotherapy. Nat Rev Immunol. 2006;(6):761-771. doi: 10.1038/nri1934.
  4. Durham SR, Walker SM, Varga EV, Jacobson MR, O’Brien F, Noble W et al. Long-term clinical efficacy of grass-pollen immunotherapy. N Engl J Med. 1999;(341):468-475. doi: 10.1056/NEJM199908123410702.
  5. Vickery BP, Scurlock AM, Kulis M, Steele PH, Kamilaris J, Berglund JP et al. Sustained unresponsiveness to peanut in subjects who have completed peanut oral immunotherapy. J Allergy Clin Immunol. 2014;(133):468-475. DOI: 10.1016/j. jaci.2013.11.007.
  6. Pajno GB, Barberio G, De Luca F, Morabito L, Parmiani S. Prevention of new sensitizations in asthmatic children monosensitized to house dust mite by specific immunotherapy. A six-year follow-up study. Clin Exp Allergy. 2001;(31):1392-1397. doi: 10.1046/j.1365-2222.2001.01161.x.
  7. Moller C, Dreborg S, Ferdousi HA, Halken S, Host A, Jacobsen L et al. Pollen immunotherapy reduces the development of asthma in children with seasonal rhinoconjunctivitis (the PAT-study) J Allergy Clin Immunol. 2002;(109):251-256. doi: 10.1067/mai.2002.121317.
  8. Wang M, Takeda K, Shiraishi Y, Okamoto M, Dakhama A, Joetham A et al. Peanut-induced intestinal allergy is mediated through a mast cell-IgE-Fc RI-IL-13 pathway. J Allergy Clin Immunol. 2010;(126):306-316. DOI: 10.1016/j. jaci.2010.05.017.
  9. Berin MC, Shreffler WG. Th2 adjuvants: implications for food allergy. J Allergy Clin Immunol. 2008;(121):1311-1320. doi: 10.1016/j.jaci.2008.04.023.
  10. Chatila TA, Li N, Garcia-Lloret M, Kim HJ, Nel AE. T-cell effector pathways in allergic diseases: transcriptional mechanisms and therapeutic targets. J Allergy Clin Immunol. 2008;(121):812-823. doi: 10.1016/j.jaci.2008.02.025.
  11. Kang CM, Jang AS, Ahn MH, Shin JA, Kim JH, Choi YS et al. Interleukin-25 and interleukin-13 production by alveolar macrophages in response to particles. Am J Respir Cell Mol Biol. 2005;(33):290-296. doi: 10.1165/rcmb.2005-0003OC.
  12. Wang YH, Angkasekwinai P, Lu N, Voo KS, Arima K, Hanabuchi S et al. IL-25 augments type 2 immune responses by enhancing the expansion and functions of TSLP-DC-activated Th2 memory cells. J Exp Med. 2007;(204):1837-1847. doi: 10.1084/jem.20070406.
  13. Dillon SR, Sprecher C, Hammond A, Bilsborough J, Rosenfeld-Franklin M, Presnell SR et al. Interleukin-31, a cytokine produced by activated T-cells, induces dermatitis in mice. Nat Immunol. 2004;(5):752-760. doi: 10.1038/ni1084.
  14. Bilsborough J, Leung DY, Maurer M, Howell M, Boguniewicz M, Yao L et al. IL-31 is associated with cutaneous lymphocyte antigen-positive skin homing T-cells in patients with atopic dermatitis. J Allergy Clin Immunol. 2006;(117):418-425. doi: 10.1016/j.jaci.2005.10.046.
  15. Kakkar R, Lee RT The IL-33/ST2 pathway: therapeutic target and novel biomarker. Nat Rev Drug Discov. 2008;(7):827-840. doi: 10.1038/nrd2660.
  16. Prefontaine D, Nadigel J, Chouiali F, Audusseau S, Semlali A, Chakir J et al. Increased IL-33 expression by epithelial cells in bronchial asthma. J Allergy Clin Immunol. 2010;(125):752-754. doi: 10.1016/j.jaci.2009.12.935.
  17. Rank MA, Kobayashi T, Kozaki H, Bartemes KR, Squillace DL, Kita H. IL-33-activated dendritic cells induce an atypical TH2-type response. J Allergy Clin Immunol. 2009;(123):1047-1054. doi: 10.1016/j.jaci.2009.02.026.
  18. Romagnani S. Immunologic influences on allergy and the TH1/TH2 balance. J Allergy Clin Immunol. 2004;(113):395-400. doi: 10.1016/j.jaci.2003.11.025.
  19. Finotto S, Neurath MF, Glickman JN, Qin S, Lehr HA, Green FH et al. Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet. Science. 2002;(295):336-338. doi: 10.1126/science.1065544.
  20. Akkoc T, de Koning PJ, Ruckert B, Barlan I, Akdis M, Akdis CA. Increased activation-induced cell death of high IFN-y-producing Th1 cells as a mechanism of Th2 predominance in atopic diseases. J Allergy Clin Immunol. 2008;(121):652-658. doi: 10.1016/j.jaci.2007.12.1171.
  21. Trautmann A, Akdis M, Kleemann D, Altznauer F, Simon HU, Graeve T et al. T-cell-mediated Fas-induced keratinocyte apoptosis plays a key pathogenetic role in eczematous dermatitis. J Clin Invest. 2000;(106):25-35. DOI: 10.1172/ JCI9199.
  22. Burgler S, Ouaked N, Bassin C, Basinski TM, Mantel PY, Siegmund K et al. Differentiation and functional analysis of human T(H)17 cells. J Allergy Clin Immunol. 2009;(123):588-595. doi: 10.1016/j.jaci.2008.12.017.
  23. Zheng Y, Danilenko DM, Valdez P, Kasman I, Eastham-Anderson J, Wu J et al. Interleukin-22, a Th17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature. 2007;(445):648-651. doi: 10.1038/nature05505.
  24. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH et al. A distinct lineage of CD4 T-cells regulates tissue inflammation by producing interleukin-17. Nat Immunol. 2005;(6):1133-1141. doi: 10.1038/ni1261.
  25. Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO et al. Transforming growth factor-beta induces development of the Th17 lineage. Nature. 2006;(441):231-234. doi: 10.1038/nature04754.
  26. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM et al. Interleukin 17-producing CD4+ effector T-cells develop via a lineage distinct from the T-helper type 1 and 2 lineages. Nat Immunol. 2005;(6):1123-1132. doi: 10.1038/ni1254.
  27. Makihara S, Okano M, Fujiwara T, Kariya S, Noda Y Higaki T et al. Regulation and characterization of IL-17A expression in patients with chronic rhinosinusitis and its relationship with eosinophilic inflammation. J Allergy Clin Immunol. 2010;(126):397-400. doi: 10.1016/j.jaci.2010.05.014.
  28. Martin P, Gomez M, Lamana A, Matesanz Marin A, Cortés JR, Ramirez-Huesca M et al. The leukocyte activation antigen CD69 limits allergic asthma and skin contact hypersensitivity. J Allergy Clin Immunol. 2010;(126):355-365. doi: 10.1016/j.jaci.2010.05.010.
  29. Sergejeva S, Ivanov S, Lotvall J, Linden A. Interleukin-17 as a recruitment and survival factor for airway macrophages in allergic airway inflammation. Am J Respir Cell Mol Biol. 2005;(33):248-253. doi: 10.1165/rcmb.2004-0213OC.
  30. Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, Overbergh L et al. Interleukin-17 orchestrates the granulocyte influx into airways after allergen inhalation in a mouse model of allergic asthma. Am J Respir Cell Mol Biol. 2003;(28):42-50. doi: 10.1165/rcmb.4832.
  31. Bush KA, Farmer KM, Walker JS, Kirkham BW Reduction of joint inflammation and bone erosion in rat adjuvant arthritis by treatment with interleukin-17 receptor IgG1 Fc fusion protein. Arthritis Rheum. 2002;(46):802-805. doi: 10.1002/art.10173.
  32. Rangachari M, Mauermann N, Marty RR, Dirnhofer S, Kurrer MO, Komnenovic V et al. T-bet negatively regulates autoimmune myocarditis by suppressing local production of interleukin 17. J Exp Med. 2006;(203):2009-2019. DOI: 10.1084/ jem.20052222.
  33. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, So-bel RA et al. IL-4 inhibits TGF-β-induced Foxp3+ T-cells and, together with TGF-β, generates IL-9+ IL-10+ Foxp3- effector T-cells. Nat Immunol. 2008;(9):1347-1355. DOI: 10.1038/ ni.1677.
  34. Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J et al. Transforming growth factor-beta ‘reprograms’ the differentiation of T-helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol. 2008;(9):1341-1346. doi: 10.1038/ni.1659.
  35. Nograles KE, Zaba LC, Shemer A, Fuentes-Duculan J, Cardinale I, Kikuchi T et al. IL-22-producing «T22» T-cells account for upregulated IL-22 in atopic dermatitis despite reduced IL-17-producing TH17 T-cells. J Allergy Clin Immunol. 2009;(123):1244-1252. doi: 10.1016/j.jaci.2009.03.041.
  36. Akdis CA, Akdis M. Mechanisms and treatment of allergic disease in the big picture of regulatory T-cells. J Allergy Clin Immunol. 2009;(123):735-746. doi: 10.1016/j.jaci.2009.02.030.
  37. Sugita K, Hanakawa S, Honda T, Kondoh G, Miyachi Y, Kabashima K et al. Generation of Helios reporter mice and an evaluation of the suppressive capacity of Helios+ regulatory T-cells in vitro. Exp Dermatol. 2015;(24):554-556. doi: 10.1111/exd.12711.
  38. Yamada T, Tongu M, Goda K, Aoi N, Morikura I, Fuchiwaki T et al. Sublingual immunotherapy induces regulatory function of IL-10-expressing CD4+CD25+FoxP3+ T-cells of cervical lymph nodes in murine allergic rhinitis model. J. Allergy. 2012;(2012):1-11. doi: 10.1155/2012/490905.
  39. Bacchetta R, Gambineri E, Roncarolo MG. Role of regulatory T-cells and FoxP3 in human diseases, Allergy Clin. Immunol. 2007;(120):227-235. doi: 10.1016/j.jaci.2007.06.023.
  40. Lai CS, Fisher SE, Hurst JA, Vargha-Khadem F, Monaco AP. A forkhead-domain gene is mutated in a severe speech and language disorder. Nature. 2001;(413):519-523. doi: 10.1038/35097076.
  41. Akdis M, Verhagen J, Taylor A, Karamloo F, Karagiannidis C, Crameri R et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T-regulatory 1 and T-helper 2 cells. J Exp Med. 2004;(199):1567-1575. doi: 10.1084/jem.20032058.
  42. Girtsman T, Jaffar Z, Ferrini M, Shaw P, Roberts K. Natural FoxP3+ regulatory T-cells inhibit Th2 polarization but are biased toward suppression of Th17-driven lung inflammation. J Leukoc Biol. 2010;(88):537-546. doi: 10.1189/jlb.0110044.
  43. Jaffar Z, Ferrini ME, Girtsman TA, Roberts K. Antigen-specific Treg regulate Th17-mediated lung neutrophilic inflammation, B-cell recruitment and polymeric IgA and IgM levels in the airways. Eur J Immunol. 2009;(39):3307-3314. doi: 10.1002/eji.200939498.
  44. Yang L, Ma QL, Yao W, Zhang Q, Chen H, Wang G et al. Relationship between the anti-inflammatory properties of salmeterol/fluticasone and the expression of CD4+CD25+FoxP3+ regulatory T-cells in COPD. Respir Res. 2011;(12):142. doi: 10.1186/1465-9921-12-142.
  45. Hori S, Nomura T, Sakaguchi S. Control of regulatory T-cell development by the transcription factor Foxp3. Science. 2003;(299):1057-1061. doi: 10.1126/science.1079490.
  46. Smith EL, Finney HM, Nesbitt AM, Ramsdell F, Robinson MK. Splice variants of human FOX P3 are functional inhibitors of human CD4+ T-cell activation. Immunology. 2006;(119):203-211. doi: 10.1111/j.1365-2567.2006.02425.x.
  47. Kaur G, Goodall JC, Jarvis LB, Gaston JSH. Characterisation of Foxp3 splice variants in human CD4+ and CD8+ T-cells - identification of Foxp357 in human regulatory T-cells. Mol Immunol. 2010;(48):321-332. DOI: 10.1016/j. molimm.2010.07.008.
  48. Mailer RKW, Kirsten Falk K, Rotzschke O. Absence of leucine zipper in the natural FOX P3D2D7 isoform does not affect dimerization but abrogates suppressive capacity. PLoS ONE. 2009;(4):1-17. doi: 10.1371/journal.pone.0006104.
  49. Du J, Huang C, Zhou B, Ziegler SF. Isoform-specific inhibition of RORa-mediated transcriptional activation by human FOX P3. J Immunol. 2008;(180):4785-4792. DOI: 10.4049/ jimmunol.180.7.4785.
  50. Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K et al. Foxp3 inhibits RORyt-mediated IL-17A mRNA transcription through direct interaction with RORyt. J Biol Chem. 2008;(283):17003-17008. doi: 10.1074/jbc.m801286200.
  51. Magg TH, Mannert J, Ellwart JW, Schmid I, Albert MH. Subcellular localization of FOX P3 in human regulatory and nonregulatory T-cells. Eur. J Immunol. 2012;(42):1627-1638. doi: 10.1002/eji.201141838.
  52. Митин АН, Литвина ММ, Митина ТА, Голенков АК, Ярилин АА. Анализ экспрессии молекулы foxp3 и ее изоформ CD4+ Т-клетками периферической крови при различных формах течения множественной миеломы методом проточной цитометрии Иммунология. 2014;(№4):215-219
  53. Kucuksezer UC, Palomares O, Ruckert B, Jartti T, Puhakka T, Nandy A et al. Triggering of specific Toll-like receptors and proinflammatory cytokines breaks allergen-specific T-cell tolerance in human tonsils and peripheral blood. J Allergy Clin Immunol. 2013;(131):875-885. DOI: 10.1016/j. jaci.2012.10.051.
  54. Palomares O, Rückert B, Jartti T, Kücüksezer UC, Puhaka T, Gomez E et al. Induction and maintenance of allergen-specific FOXP3+ Treg cells in human tonsils as potential first-line organs of oral tolerance. J Allergy Clin Immunol. 2012;(129):510-520. doi: 10.1016/j.jaci.2011.09.031.
  55. Jartti T, Palomares O, Waris M, Tastan O, Nieminen R, Puhakka T et al. Distinct regulation of tonsillar immune response in virus infection. Allergy. 2014;(69):658-667. DOI: 10.1111/ all.12396.
  56. Mehta AK, Duan W, Doerner AM, Traves SL, Broide DH, Proud D et al. Rhino- virus infection interferes with induction of tolerance to aeroantigens through OX40 ligand, thymic stromal lymphopoietin, and IL-33. J Allergy Clin Immunol. 2016;(137):278-288. doi: 10.1016/j.jaci.2015.05.007.
  57. Van de Veen W, Stanic B, Yaman G, Wawrzyniak M, Söllner S, Akdis DG et al. IgG4 production is confined to human IL-10-producing regulatory B-cells that suppress antigen-specific immune responses. J Allergy Clin Immunol. 2013;(131):1204-1212. doi: 10.1016/j.jaci.2013.01.014.
  58. Stanic B, van de Veen W, Wirz OF, Rückert B, Morita H, Söllner S et al. IL-10-overexpressing B-cells regulate innate and adaptive immune responses. J Allergy Clin Immunol. 2014;(135):771-780. doi: 10.1016/j.jaci.2014.07.041.
  59. Bartemes KR, Kephart GM, Fox SJ, Kita H. Enhanced innate type 2 immune response in peripheral blood from patients with asthma. J Allergy Clin Immunol. 2014;(1340):671-678. doi: 10.1016/j.jaci.2014.06.024.
  60. Annunziato F, Romagnani C, Romagnani S. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol. 2015;(135):626-635. DOI: 10.1016/j. jaci.2014.11.001.
  61. Jutel M, Agache I, Bonini S, Burks AW, Calderon M, Canonica W International Consensus On (ICON) Allergy Immunotherapy (AIT) II: mechanisms, standardization and pharmacoeconomics. J Allergy Clin Immunol. 2016;(137):358-368. doi: 10.1016/j.jaci.2015.12.1300.
  62. Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. WAO Journal. 2015;(8):17. doi: 10.1186/s40413-015-0063-2.
  63. Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy: multiple suppressor factors at work in immune tolerance to allergens. J Allergy Clin Immunol. 2014;(133):621-631. doi: 10.1016/j.jaci.2013.12.1088.
  64. Burks AW, Calderon MA, Casale T, Cox L, Demoly P, Jutel M et al. Update on allergy immunotherapy: American Academy of Allergy, Asthma & Immunology/European Academy of Allergy and Clinical Immunology/PRACTALL consensus report. J Allergy Clin Immunol. 2013;(131): 1288-1296. doi: 10.1016/j.jaci.2013.01.049.
  65. Agache I, Sugita K, Morita H, Akdis M, Akdis CA. The complex type 2 endotype in allergy and asthma: from laboratory to bedside. Curr Allergy Asthma Rep. 2015;(15):29. doi: 10.1007/s11882-015-0529-x.
  66. Tarzi M, Klunker S, Texier C, Verhoef A, Stapel SO, Akdis CA et al. Induction of interleukin-10 and suppressor of cytokine signalling-3 gene expression following peptide immunotherapy. Clin Exp Allergy. 2006;(36):465-474. doi: 10.1111/j.1365-2222.2006.02469.x.
  67. Alexander C, Ying S, Kay A, Larche M. Fel d 1-derived T-cell peptide therapy induces recruitment of CD4+CD25+; CD4+ interferon-gamma + T-helper type 1 cells to sites of allergen-induced late-phase skin reactions in cat-allergic subjects. Clin Exp Allergy. 2005;(35):52-58. doi: 10.1111/j.1365-2222.2005.02143.x.
  68. Romano A, Torres MJ, Castells M, Sanz ML, Blanca M. Diagnosis and management of drug hypersensitivity reactions. J Allergy Clin Immunol. 2011;(127):S67-73. DOI: 10.1016/j. jaci.2010.11.047.
  69. Jutel M, Akdis M, Akdis CA. Histamine, histamine receptors and their role in immune pathology. Clin Exp Allergy. 2009;(39):1786-1800. doi: 10.1111/j.1365-2222.2009.03374.x.
  70. Meiler F, Zumkehr J, Klunker S, Ruckert B, Akdis CA, Akdis M. In vivo switch to IL-10-secreting T-regulatory cells in high dose allergen exposure. J Exp Med. 2008;(205):2887-2898. doi: 10.1084/jem.20080193.
  71. Novak N, Mete N, Bussmann C, Maintz L, Bieber T, Akdis M et al. Early suppression of basophil activation during allergen-specific immunotherapy by histamine receptor 2. J Allergy Clin Immunol. 2012;(130):1153-1158. DOI: 10.1016/j. jaci.2012.04.039.
  72. Ferstl R, Frei R, Schiavi E, Konieczna P, Barcik W, Ziegler M et al. Histamine receptor 2 is a key influence in immune responses to intestinal histamine- secreting microbes. J Allergy Clin Immunol. 2014;(134):744-746. DOI: 10.1016/j. jaci.2014.04.034.
  73. Shamji MH, Layhadi JA, Scadding GW, Cheung DK, Calderon MA, Turka LA et al. Basophil expression of diamine oxidase: A novel biomarker of allergen immunotherapy response. J Allergy Clin Immunol. 2015;(135):913-921. DOI: 10.1016/j. jaci.2014.09.049.
  74. Harvima IT, Levi-Schaffer F, Draber P, Friedman S, Polakovicova I, Gibbs BF et al. Molecular targets on mast cells and basophils for novel therapies. J Allergy Clin Immunol. 2014;(134):530-544. doi: 10.1016/j.jaci.2014.03.007.
  75. Kaczorowski M, Jutel M. Human T-regulatory cells: on the way to cognition. Arch Immunol Ther Exp. 2013;(61):229-236. doi: 10.1007/s00005-013-0217-2.
  76. Jutel M, Akdis CA. Immunological mechanisms of allergen-specific immunotherapy. Allergy. 2011;(66):725-732. doi: 10.1111/j.1398-9995.2011.02589.x.
  77. Suarez-Fueyo A, Ramos T, Galan A, Jimeno L, Wurtzen PA, Marin A et al. Grass tablet sublingual immunotherapy down-regulates the TH2 cytokine response followed by regulatory T-cell generation. J Allergy Clin Immunol. 2014;(133):130-138. doi: 10.1016/j.jaci.2013.09.043.
  78. Lou W, Wang C, Wang Y, Han D, Zhang L. Responses of CD4+CD25+ Foxp3+ and IL-10-secreting type I T-regulatory cells to cluster-specific immunotherapy for allergic rhinitis in children. Pediatr Allergy Immunol. 2011;(23):141-150. doi: 10.1111/j.1399-3038.2011.01249.x.
  79. Mobs C, Ipsen H, Mayer L, Slotosch C, Petersen A, Wurtzen PA et al. Birch pollen immunotherapy results in long-term loss of Bet v 1-specific TH2 responses, transient TR1 activation, and synthesis of IgE-blocking antibodies. J Allergy Clin Immunol. 2012;(130):1108-1116. DOI: 10.1016/j. jaci.2012.07.056.
  80. Radulovic S, Jacobson MR, Durham SR, Nouri-Aria KT. Grass pollen immunotherapy induces Foxp3-expressing CD4+CD25+ cells in the nasal mucosa. J Allergy Clin Immunol. 2008;(121):1467-1472. doi: 10.1016/j.jaci.2008.03.013.
  81. Meiler F, Klunker S, Zimmermann M, Akdis CA, Akdis M. Distinct regulation of IgE, IgG4 and IgA by T-regulatory cells and toll-like receptors. Allergy. 2008;(63):1455-1463. doi: 10.1111/j.1398-9995.2008.01774.x.
  82. De Wit J, Jorritsma T, Makuch M, Remmerswaal EB, Klaasse Bos H, Souwer Y et al. Human B-cells promote T-cell plasticity to optimize antibody response by inducing coexpression of T(H)1/T(FH) signatures. J Allergy Clin Immunol. 2015;(135):1053-1060. doi: 10.1016/j.jaci.2014.08.012.
  83. Wachholz PA, Durham SR. Mechanisms of immunotherapy: IgG revisited. Curr Opin Allergy Clin Immunol. 2004;(4):313-318. doi: 10.1097/01.all.0000136753.35948.c0.
  84. Van der Neut Kolfschoten M, Schuurman J, Losen M, Bleeker WK, Martinez-Martinez P, Vermeulen E et al. Anti-inflammatory activity of human IgG4 antibodies by dynamic Fab arm exchange. Science. 2007;(317):1554-1557. DOI: 10.1126/ science.1144603.
  85. Aalberse RC, Schuurman J. IgG4 breaking the rules. Immunology. 2002;(105):9-19. doi: 10.1046/j.0019-2805.2001.01341.x.
  86. Gold MJ, Antignano F, Halim TY, Hirota JA, Blanchet MR, Zaph C et al. Group 2 innate lymphoid cells facilitate sensitization to local, but not systemic, TH2-inducing allergen exposures. J Allergy Clin Immunol. 2014;(133):1142-1148. doi: 10.1016/j.jaci.2014.02.033.
  87. Nagakumar P, Denney L, Fleming L, Bush A, Lloyd CM, Saglani S. Type 2 innate lymphoid cells in induced sputum from children with severe asthma. J Allergy Clin Immunol. 2016;(137):624-626. doi: 10.1016/j.jaci.2015.06.038.
  88. Lao-Araya M, Steveling E, Scadding GW, Durham SR, Shamji MH. Seasonal increases in peripheral innate lymphoid type 2 cells are inhibited by subcutaneous grass pollen immunotherapy. J Allergy Clin Immunol. 2014;(134):1193-1195. doi: 10.1016/j.jaci.2014.07.029.
  89. Shamji MH, Kappen JH, Akdia M, Jensen-Jarolim E, Knol EF, Kleine-Tebbe J et al. Biomarkers for monitoring clinical efficacy of allergen immunotherapy for allergic rhinoconjunctivitis and allergic asthma: an EAACI Position Paper. Allergy. 2017;(72):1156-1173. doi: 10.1111/all.13138.
  90. Pfaar O, Demoly P, Gerth van Wijk R, Bonini S, Bousquet J, Canonica GW et al. EAACI. Recommendations for the standardization of clinical outcomes used in allergen immunotherapy trials for allergic rhinoconjunctivitis: an EAACI Position Paper. Allergy. 2014;(69):854-867. DOI: 10.1111/ all.12383.
  91. Casale TB, Stokes JR. Immunotherapy: what lies beyond. J Allergy Clin Immunol. 2014;(133):612-619. DOI: 10.1016/j. jaci.2014.01.007.
  92. Jutel M, Akdis CA. Novel immunotherapy vaccine development. Curr Opin Allergy Clin Immunol. 2014;(14):557-563. doi: 10.1097/aci.0000000000000121.
  93. Sharma P, Gaur SN, Arora N. Immunotherapy with B-cell epitopes ameliorates inflammatory responses in Balb/c mice. Clin Exp Immunol. 2014;(179):128-136. DOI: 10.1111/ cei.12442.
  94. Jones SM, Burks AW, Dupont C. State of the art on food allergen immunotherapy: oral, sublingual, and epicutaneous. J Allergy Clin Immunol. 2014;(133):318-323. DOI: 10.1016/j. jaci.2013.12.1040.
  95. Jutel M, Solarewicz-Madejek K, Smolinska S. Recombinant allergens: the present and the future. Hum Vaccin Immunother. 2012;(8):1534-1543. doi: 10.4161/hv.22064.
  96. Linhart B, Focke-Tejkl M, Weber M, Narayanan M, Neubauer A, Mayrhofer H et al. Molecular evolution of hypoallergenic hybrid proteins for vaccination against grass pollen allergy. J Immunol. 2015;(194):4008-4018. DOI: 10.4049/ jimmunol.1400402.
  97. Fili L, Cardilicchia E, Maggi E, Parronchi P. Perspectives in vaccine adjuvants for allergen-specific immunotherapy. Immunol Lett. 2014;(161):207-210. doi: 10.1016/j.imlet.2013.12.011.
  98. Grundstrom J, Neimert-Andersson T, Kemi C, Nilsson OB, Saarne T, Andersson M et al. Covalent coupling of vitamin D3 to the major cat allergen Fel d 1 improves the effects of allergen-specific immunotherapy in a mouse model for cat allergy. Int Arch Allergy Immunol. 2012;(157):136-146. doi: 10.1159/000327546.
  99. Linhart B, Narayanan M, Focke-Tejkl M, Wrba F, Vrtala S, Valenta R. Prophylactic and therapeutic vaccination with carrier-bound Bet v 1 peptides lacking allergen-specific T-cell epitopes reduces Bet v 1-specific T-cell responses via blocking antibodies in a murine model for birch pollen allergy. Clin Exp Allergy. 2014;(44):278-287. doi: 10.1111/cea.12216.
  100. Schneider LC, Rachid R, LeBovidge J, Blood E, Mittal M, Umetsu DT A pilot study of omalizumab to facilitate rapid oral desensitization in high-risk peanut- allergic patients. J Allergy Clin Immunol. 2013;(132):1368-1374. DOI: 10.1016/j. jaci.2013.09.046.
  101. Larenas-Linnemann D, Wahn U, Kopp M. Use of omalizumab to improve desensitization safety in allergen immunotherapy. J Allergy Clin Immunol. 2014;(133):937-937. doi: 10.1016/j.jaci.2013.12.1089.
  102. Kundig TM, Johansen P, Bachmann MF, Cardell LO, Senti G. Intralymphatic immunotherapy: time interval between injections is essential. J Allergy Clin Immunol. 2014;(133):930-931. doi: 10.1016/j.jaci.2013.11.036.
  103. Creticos PS, Esch RE, Couroux P, Gentile D, D’Angelo P, Whitlow B et al. Randomized, double-blind, placebo-controlled trial of standardized ragweed sublingual-liquid immunotherapy for allergic rhinoconjunctivitis. J Allergy Clin Immunol. 2014;(133):751-758. doi: 10.1016/j.jaci.2013.10.041.
  104. Patel P, Holdich T, Fischer von Weikersthal-Drachenberg KJ, Huber B. Efficacy of a short course of specific immunotherapy in patients with allergic rhinoconjunctivitis to ragweed pollen. J Allergy Clin Immunol. 2014;(133):121-129. DOI: 10.1016/j. jaci.2013.05.032.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright © Pharmarus Print Media, 2017



This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies