Asia Pac Allergy.  2018 Apr;8(2):e14. 10.5415/apallergy.2018.8.e14.

Novel strategies in immunotherapy for allergic diseases

Affiliations
  • 1Khoo Teck Puat – National University Children's Medical Institute, National University Health System, Singapore 119229. mohana_rajakulendran@nuhs.edu.sg
  • 2Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119229.

Abstract

Conventional immunotherapy (IT) for optimal control of respiratory and food allergies has been fraught with concerns of efficacy, safety, and tolerability. The development of adjuvants to conventional IT has potentially increased the effectiveness and safety of allergen IT, which may translate into improved clinical outcomes and sustained unresponsiveness even after cessation of therapy. Novel strategies incorporating the successful use of adjuvants such as allergoids, immunostimulatory DNA sequences, monoclonal antibodies, carriers, recombinant proteins, and probiotics have now been described in clinical and murine studies. Future approaches may include fungal compounds, parasitic molecules, vitamin D, and traditional Chinese herbs. More robust comparative clinical trials are needed to evaluate the safety, clinical efficacy, and cost effectiveness of various adjuvants in order to determine ideal candidates in disease-specific and allergen-specific models. Other suggested approaches to further optimize outcomes of IT include early introduction of IT during an optimal window period. Alternative routes of administration of IT to optimize delivery and yet minimize potential side effects require further evaluation for safety and efficacy before they can be recommended.

Keyword

Immunotherapy; Allergy immunology; Allergoid; Immunologic stimulation; Monoclonal antibody; Probiotics

MeSH Terms

Antibodies, Monoclonal
Asian Continental Ancestry Group
Base Sequence
Cost-Benefit Analysis
Food Hypersensitivity
Humans
Immunization
Immunotherapy*
Probiotics
Recombinant Proteins
Treatment Outcome
Vitamin D
Antibodies, Monoclonal
Recombinant Proteins
Vitamin D

Cited by  1 articles

Spring and allergy
Yoon-Seok Chang
Asia Pac Allergy. 2018;8(2):.    doi: 10.5415/apallergy.2018.8.e22.


Reference

References

1. Larenas-Linnemann D, Pfaar O. Patient-reported outcomes and quality-of-life questionnaires in the assessment of rhinoconjunctivitis in childhood. Curr Opin Allergy Clin Immunol. 2014; 14:192–9.
Article
2. Zuberbier T, Lötvall J, Simoens S, Subramanian SV, Church MK. Economic burden of inadequate management of allergic diseases in the European Union: a GA(2) LEN review. Allergy. 2014; 69:1275–9.
3. Jutel M, Akdis M, Budak F, Aebischer-Casaulta C, Wrzyszcz M, Blaser K, Akdis CA. IL-10 and TGF-beta cooperate in the regulatory T cell response to mucosal allergens in normal immunity and specific immunotherapy. Eur J Immunol. 2003; 33:1205–14.
4. Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy: multiple suppressor factors at work in immune tolerance to allergens. J Allergy Clin Immunol. 2014; 133:621–31.
Article
5. van de Veen W, Stanic B, Yaman G, Wawrzyniak M, Söllner S, Akdis DG, Rückert B, Akdis CA, Akdis M. IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J Allergy Clin Immunol. 2013; 131:1204–12.
Article
6. Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol. 2011; 127:18–27.
Article
7. Tophof MA, Hermanns A, Adelt T, Eberle P, Gronke C, Friedrichs F, Knecht R, Mönter E, Schöpfer H, Schwerk N, Steinbach J, Umpfenbach HU, Weißhaar C, Wilmsmeyer B, Bufe A. Side effects during subcutaneous immunotherapy in children with allergic diseases. Pediatr Allergy Immunol. 2017 Dec 16. [Epub].https://doi.org/10.1111/pai.12847.
Article
8. Pajno GB, Caminiti L, Chiera F, Crisafulli G, Salzano G, Arasi S, Passalacqua G. Safety profile of oral immunotherapy with cow's milk and hen egg: A 10-year experience in controlled trials. Allergy Asthma Proc. 2016; 37:400–3.
Article
9. Kulmala P, Pelkonen AS, Kuitunen M, Paassilta M, Remes S, Schultz R, Dunder T, Turunen S, Mäkelä MJ. Wheat oral immunotherapy was moderately successful but was associated with very frequent adverse events in children aged 6-18 years. Acta Paediatr. 2018 Jan 18. [Epub].https://doi.org/10.1111/apa.14226.
Article
10. Di Costanzo M, Paparo L, Cosenza L, Di Scala C, Nocerino R, Aitoro R, Canani RB. Food allergies: novel mechanisms and therapeutic perspectives. Methods Mol Biol. 2016; 1371:215–21.
Article
11. Jongejan L, van Ree R. Modified allergens and their potential to treat allergic disease. Curr Allergy Asthma Rep. 2014; 14:478.
Article
12. Casanovas M, Gómez MJ, Carnés J, Fernández-Caldas E. Skin tests with native, depigmented and glutaraldehyde polymerized allergen extracts. J Investig Allergol Clin Immunol. 2005; 15:30–6.
13. Gallego MT, Iraola V, Himly M, Robinson DS, Badiola C, García-Robaina JC, Briza P, Carnés J. Depigmented and polymerised house dust mite allergoid: allergen content, induction of IgG4 and clinical response. Int Arch Allergy Immunol. 2010; 153:61–9.
Article
14. Henmar H, Lund G, Lund L, Petersen A, Würtzen PA. Allergenicity, immunogenicity and dose-relationship of three intact allergen vaccines and four allergoid vaccines for subcutaneous grass pollen immunotherapy. Clin Exp Immunol. 2008; 153:316–23.
Article
15. Würtzen PA, Lund L, Lund G, Holm J, Millner A, Henmar H. Chemical modification of birch allergen extract leads to a reduction in allergenicity as well as immunogenicity. Int Arch Allergy Immunol. 2007; 144:287–95.
Article
16. Lund L, Henmar H, Würtzen PA, Lund G, Hjortskov N, Larsen JN. Comparison of allergenicity and immunogenicity of an intact allergen vaccine and commercially available allergoid products for birch pollen immunotherapy. Clin Exp Allergy. 2007; 37:564–71.
Article
17. Heydenreich B, Bellinghausen I, Lorenz S, Henmar H, Strand D, Würtzen PA, Saloga J. Reduced in vitro T-cell responses induced by glutaraldehyde-modified allergen extracts are caused mainly by retarded internalization of dendritic cells. Immunology. 2012; 136:208–17.
Article
18. Reinhold T, Brüggenjürgen B. Cost-effectiveness of grass pollen SCIT compared with SLIT and symptomatic treatment. Allergo J Int. 2017; 26:7–15.
Article
19. Rosewich M, Lee D, Zielen S. Pollinex Quattro: an innovative four injections immunotherapy in allergic rhinitis. Hum Vaccin Immunother. 2013; 9:1523–31.
Article
20. Carnés J, Himly M, Gallego M, Iraola V, Robinson DS, Fernández-Caldas E, Briza P. Detection of allergen composition and in vivo immunogenicity of depigmented allergoids of Betula alba. Clin Exp Allergy. 2009; 39:426–34.
21. Heydenreich B, Bellinghausen I, Lund L, Henmar H, Lund G, Adler Würtzen P, Saloga J. Adjuvant effects of aluminium hydroxide-adsorbed allergens and allergoids – differences in vivo and in vitro. Clin Exp Immunol. 2014; 176:310–9.
Article
22. Higgins D, Marshall JD, Traquina P, Van Nest G, Livingston BD. Immunostimulatory DNA as a vaccine adjuvant. Expert Rev Vaccines. 2007; 6:747–59.
Article
23. Creticos PS, Schroeder JT, Hamilton RG, Balcer-Whaley SL, Khattignavong AP, Lindblad R, Li H, Coffman R, Seyfert V, Eiden JJ. Broide DImmune Tolerance Network Group. Immunotherapy with a ragweed-toll-like receptor 9 agonist vaccine for allergic rhinitis. N Engl J Med. 2006; 355:1445–55.
Article
24. Tulic MK, Fiset PO, Christodoulopoulos P, Vaillancourt P, Desrosiers M, Lavigne F, Eiden J, Hamid Q. Amb a 1-immunostimulatory oligodeoxynucleotide conjugate immunotherapy decreases the nasal inflammatory response. J Allergy Clin Immunol. 2004; 113:235–41.
Article
25. Zhu FG, Kandimalla ER, Yu D, Agrawal S. Oral administration of a synthetic agonist of Toll-like receptor 9 potently modulates peanut-induced allergy in mice. J Allergy Clin Immunol. 2007; 120:631–7.
Article
26. Mantel PY, Kuipers H, Boyman O, Rhyner C, Ouaked N, Rückert B, Karagiannidis C, Lambrecht BN, Hendriks RW, Crameri R, Akdis CA, Blaser K, Schmidt-Weber CB. GATA3-driven Th2 responses inhibit TGF-beta1-induced FOXP3 expression and the formation of regulatory T cells. PLoS Biol. 2007; 5:e329.
27. Rosewich M, Girod K, Zielen S, Schubert R, Schulze J. Induction of bronchial tolerance after 1 cycle of monophosphoryl-a-adjuvanted specific immunotherapy in children with grass pollen allergies. Allergy Asthma Immunol Res. 2016; 8:257–63.
Article
28. Musarra A, Bignardi D, Troise C, Passalacqua G. Long-lasting effect of a monophosphoryl lipid-adjuvanted immunotherapy to parietaria. A controlled field study. Eur Ann Allergy Clin Immunol. 2010; 42:115–9.
29. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, Mitsdoerffer M, Strom TB, Elyaman W, Ho IC, Khoury S, Oukka M, Kuchroo VK. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+IL-10+ Foxp3 (-) effector T cells. Nat Immunol. 2008; 9:1347–55.
30. Chaker AM, Shamji MH, Dumitru FA, Calderon MA, Scadding GW, Makatsori M, Jones I, He QA, Subramanian KK, Arm JP, Durham SR, Schmidt-Weber CB. Short-term subcutaneous grass pollen immunotherapy under the umbrella of anti-IL-4: A randomized controlled trial. J Allergy Clin Immunol. 2016; 137:452–61.
31. Prussin C, Griffith DT, Boesel KM, Lin H, Foster B, Casale TB. Omalizumab treatment downregulates dendritic cell FcepsilonRI expression. J Allergy Clin Immunol. 2003; 112:1147–54.
32. Rolinck-Werninghaus C, Hamelmann E, Keil T, Kulig M, Koetz K, Gerstner B, Kuehr J, Zielen S, Schauer U, Kamin W, Von Berg A, Hammermann J, Weinkauf B, Weidinger G, Stenglein S. Wahn UOmalizumab Rhinitis Study Group. The co-seasonal application of anti-IgE after preseasonal specific immunotherapy decreases ocular and nasal symptom scores and rescue medication use in grass pollen allergic children. Allergy. 2004; 59:973–9.
Article
33. Braido F, Corsico A, Rogkakou A, Ronzoni V, Baiardini I, Canonica GW. The relationship between allergen immunotherapy and omalizumab for treating asthma. Expert Rev Respir Med. 2015; 9:129–34.
Article
34. Lambert N, Guiddir T, Amat F, Just J. Pre-treatment by omalizumab allows allergen immunotherapy in children and young adults with severe allergic asthma. Pediatr Allergy Immunol. 2014; 25:829–32.
Article
35. Boni E, Incorvaia C, Mauro M. Dose-dependence of protection from systemic reactions to venom immunotherapy by omalizumab. Clin Mol Allergy. 2016; 14:14.
Article
36. Ricciardi L. Omalizumab: A useful tool for inducing tolerance to bee venom immunotherapy. Int J Immunopathol Pharmacol. 2016; 29:726–8.
Article
37. Palgan K, Bartuzi Z, Gotz-Zbikowska M. Treatment with a combination of omalizumab and specific immunotherapy for severe anaphylaxis after a wasp sting. Int J Immunopathol Pharmacol. 2014; 27:109–12.
Article
38. Rafi A, Do LT, Katz R, Sheinkopf LE, Simons CW, Klaustermeyer W. Effects of omalizumab in patients with food allergy. Allergy Asthma Proc. 2010; 31:76–83.
Article
39. 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; 132:1368–74.
Article
40. MacGinnitie AJ, Rachid R, Gragg H, Little SV, Lakin P, Cianferoni A, Heimall J, Makhija M, Robison R, Chinthrajah RS, Lee J, Lebovidge J, Dominguez T, Rooney C, Lewis MO, Koss J, Burke-Roberts E, Chin K, Logvinenko T, Pongracic JA, Umetsu DT, Spergel J, Nadeau KC, Schneider LC. Omalizumab facilitates rapid oral desensitization for peanut allergy. J Allergy Clin Immunol. 2017; 139:873–81.
Article
41. Nadeau KC, Schneider LC, Hoyte L, Borras I, Umetsu DT. Rapid oral desensitization in combination with omalizumab therapy in patients with cow's milk allergy. J Allergy Clin Immunol. 2011; 127:1622–4.
Article
42. Sampson HA, Leung DY, Burks AW, Lack G, Bahna SL, Jones SM, Wong DA. A phase II, randomized, double blind, parallel group, placebo controlled oral food challenge trial of Xolair (omalizumab) in peanut allergy. J Allergy Clin Immunol. 2011; 127:1309–10.
43. Wood RA, Sicherer SH, Burks AW, Grishin A, Henning AK, Lindblad R, Stablein D, Sampson HA. A phase 1 study of heat/phenol-killed, E. coli-encapsulated, recombinant modified peanut proteins Ara h 1, Ara h 2, and Ara h 3 (EMP-123) for the treatment of peanut allergy. Allergy. 2013; 68:803–8.
44. Brandström J, Vetander M, Lilja G, Johansson SG, Sundqvist AC, Kalm F, Nilsson C, Nopp A. Individually dosed omalizumab: an effective treatment for severe peanut allergy. Clin Exp Allergy. 2017; 47:540–50.
Article
45. Frischmeyer-Guerrerio PA, Masilamani M, Gu W, Brittain E, Wood R, Kim J, Nadeau K, Jarvinen KM, Grishin A, Lindblad R, Sampson HA. Mechanistic correlates of clinical responses to omalizumab in the setting of oral immunotherapy for milk allergy. J Allergy Clin Immunol. 2017; 140:1043–53.
46. Bégin P, Dominguez T, Wilson SP, Bacal L, Mehrotra A, Kausch B, Trela A, Tavassoli M, Hoyte E, O'Riordan G, Blakemore A, Seki S, Hamilton RG, Nadeau KC. Phase 1 results of safety and tolerability in a rush oral immunotherapy protocol to multiple foods using Omalizumab. Allergy Asthma Clin Immunol. 2014; 10:7.
Article
47. Kramer MF, Heath MD. Aluminium in allergen-specific subcutaneous immunotherapy: a German perspective. Vaccine. 2014; 32:4140–8.
48. Arora N, Gangal SV. Efficacy of liposome entrapped allergen in down regulation of IgE response in mice. Clin Exp Allergy. 1992; 22:35–42.
Article
49. Tasaniyananda N, Chaisri U, Tungtrongchitr A, Chaicumpa W, Sookrung N. Mouse model of cat allergic rhinitis and intranasal liposome-adjuvanted refined fel d 1 vaccine. PLoS One. 2016; 11:e0150463.
Article
50. Meechan P, Tungtrongchitr A, Chaisri U, Maklon K, Indrawattana N, Chaicumpa W, Sookrung N. Intranasal, liposome-adjuvanted cockroach allergy vaccines made of refined major allergen and whole-body extract of Periplaneta americana. Int Arch Allergy Immunol. 2013; 161:351–62.
51. Sehra S, Chugh L, Gangal SV. Polarized TH1 responses by liposome-entrapped allergen and its potential in immunotherapy of allergic disorders. Clin Exp Allergy. 1998; 28:1530–7.
Article
52. Valenta R, Linhart B, Swoboda I, Niederberger V. Recombinant allergens for allergen-specific immunotherapy: 10 years anniversary of immunotherapy with recombinant allergens. Allergy. 2011; 66:775–83.
53. Focke M, Marth K, Flicker S, Valenta R. Heterogeneity of commercial timothy grass pollen extracts. Clin Exp Allergy. 2008; 38:1400–8.
Article
54. Vrtala S, Fohr M, Campana R, Baumgartner C, Valent P, Valenta R. Genetic engineering of trimers of hypoallergenic fragments of the major birch pollen allergen, Bet v 1, for allergy vaccination. Vaccine. 2011; 29:2140–8.
Article
55. Pichler U, Hauser M, Hofer H, Himly M, Hoflehner E, Steiner M, Mutschlechner S, Hufnagl K, Ebner C, Mari A, Briza P, Bohle B, Wiedermann U, Ferreira F, Wallner M. Allergen hybrids – next generation vaccines for Fagales pollen immunotherapy. Clin Exp Allergy. 2014; 44:438–49.
56. Spertini F, Perrin Y, Audran R, Pellaton C, Boudousquié C, Barbier N, Thierry AC, Charlon V, Reymond C. Safety and immunogenicity of immunotherapy with Bet v 1-derived contiguous overlapping peptides. J Allergy Clin Immunol. 2014; 134:239–40.
Article
57. Zieglmayer P, Focke-Tejkl M, Schmutz R, Lemell P, Zieglmayer R, Weber M, Kiss R, Blatt K, Valent P, Stolz F, Huber H, Neubauer A, Knoll A, Horak F, Henning R, Valenta R. Mechanisms, safety and efficacy of a B cell epitope-based vaccine for immunotherapy of grass pollen allergy. EBioMedicine. 2016; 11:43–57.
Article
58. Klimek L, Bachert C, Lukat KF, Pfaar O, Meyer H, Narkus A. Allergy immunotherapy with a hypoallergenic recombinant birch pollen allergen rBet v 1-FV in a randomized controlled trial. Clin Transl Allergy. 2015; 5:28.
Article
59. Wood RA, Kim JS, Lindblad R, Nadeau K, Henning AK, Dawson P, Plaut M, Sampson HA. A randomized, double-blind, placebo-controlled study of omalizumab combined with oral immunotherapy for the treatment of cow's milk allergy. J Allergy Clin Immunol. 2016; 137:1103–10.
Article
60. Wai CY, Leung NY, Leung PS, Chu KH. T cell epitope immunotherapy ameliorates allergic responses in a murine model of shrimp allergy. Clin Exp Allergy. 2016; 46:491–503.
Article
61. Rupa P, Mine Y. Oral immunotherapy with immunodominant T-cell epitope peptides alleviates allergic reactions in a Balb/c mouse model of egg allergy. Allergy. 2012; 67:74–82.
Article
62. Yang M, Yang C, Mine Y. Multiple T cell epitope peptides suppress allergic responses in an egg allergy mouse model by the elicitation of forkhead box transcription factor 3- and transforming growth factor-beta-associated mechanisms. Clin Exp Allergy. 2010; 40:668–78.
63. Liu Y, Sun Y, Chang LJ, Li N, Li H, Yu Y, Bryce PJ, Grammer LC, Schleimer RP, Zhu D. Blockade of peanut allergy with a novel Ara h 2-Fcγ fusion protein in mice. J Allergy Clin Immunol. 2013; 131:213–21.
Article
64. Freidl R, Gstoettner A, Baranyi U, Swoboda I, Stolz F, Focke-Tejkl M, Wekerle T, van Ree R, Valenta R, Linhart B. Blocking antibodies induced by immunization with a hypoallergenic parvalbumin mutant reduce allergic symptoms in a mouse model of fish allergy. J Allergy Clin Immunol. 2017; 139:1897–905.
Article
65. Ueno HM, Kato T, Ohnishi H, Kawamoto N, Kato Z, Kaneko H, Kondo N, Nakano T. T-cell epitope-containing hypoallergenic β-lactoglobulin for oral immunotherapy in milk allergy. Pediatr Allergy Immunol. 2016; 27:818–24.
Article
66. Liu J, Chen FH, Qiu SQ, Yang LT, Zhang HP, Liu JQ, Geng XR, Yang G, Liu ZQ, Li J, Liu ZG, Li HB, Yang PC. Probiotics enhance the effect of allergy immunotherapy on regulating antigen specific B cell activity in asthma patients. Am J Transl Res. 2016; 8:5256–70.
67. Christensen HR, Frøkiaer H, Pestka JJ. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol. 2002; 168:171–8.
Article
68. de Roock S, van Elk M, van Dijk ME, Timmerman HM, Rijkers GT, Prakken BJ, Hoekstra MO, de Kleer IM. Lactic acid bacteria differ in their ability to induce functional regulatory T cells in humans. Clin Exp Allergy. 2010; 40:103–10.
Article
69. Berni Canani R, Nocerino R, Terrin G, Coruzzo A, Cosenza L, Leone L, Troncone R. Effect of Lactobacillus GG on tolerance acquisition in infants with cow's milk allergy: a randomized trial. J Allergy Clin Immunol. 2012; 129:580–2.
70. Hol J, van Leer EH, Elink Schuurman BE, de Ruiter LF, Samsom JN, Hop W, Neijens HJ, de Jongste JC. Nieuwenhuis EECow's Milk Allergy Modified by Elimination and Lactobacilli study group. The acquisition of tolerance toward cow's milk through probiotic supplementation: a randomized, controlled trial. J Allergy Clin Immunol. 2008; 121:1448–54.
Article
71. Jerzynska J, Stelmach W, Balcerak J, Woicka-Kolejwa K, Rychlik B, Blauz A, Wachulec M, Stelmach P, Majak P, Stelmach I. Effect of Lactobacillus rhamnosus GG and vitamin D supplementation on the immunologic effectiveness of grass-specific sublingual immunotherapy in children with allergy. Allergy Asthma Proc. 2016; 37:324–34.
Article
72. Borchers AT, Krishnamurthy A, Keen CL, Meyers FJ, Gershwin ME. The immunobiology of mushrooms. Exp Biol Med (Maywood). 2008; 233:259–76.
Article
73. Hsieh KY, Hsu CI, Lin JY, Tsai CC, Lin RH. Oral administration of an edible-mushroom-derived protein inhibits the development of food-allergic reactions in mice. Clin Exp Allergy. 2003; 33:1595–602.
Article
74. Gomez-Escobar N, van den Biggelaar A, Maizels R. A member of the TGF-beta receptor gene family in the parasitic nematode Brugia pahangi. Gene. 1997; 199:101–9.
75. Gomez-Escobar N, Gregory WF, Maizels RM. Identification of tgh-2, a filarial nematode homolog of Caenorhabditis elegans daf-7 and human transforming growth factor beta, expressed in microfilarial and adult stages of Brugia malayi. Infect Immun. 2000; 68:6402–10.
76. Hirata M, Hirata K, Hara T, Kawabuchi M, Fukuma T. Expression of TGF-beta-like molecules in the life cycle of Schistosoma japonicum. Parasitol Res. 2005; 95:367–73.
77. Urry ZL, Richards DF, Black C, Morales M, Carnés J, Hawrylowicz CM, Robinson DS. Depigmented-polymerised allergoids favour regulatory over effector T cells: enhancement by 1α, 25-dihydroxyvitamin D3. BMC Immunol. 2014; 15:21.
Article
78. Grundström J, Neimert-Andersson T, Kemi C, Nilsson OB, Saarne T, Andersson M, van Hage M, Gafvelin G. 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; 157:136–46.
Article
79. Baris S, Kiykim A, Ozen A, Tulunay A, Karakoc-Aydiner E, Barlan IB. Vitamin D as an adjunct to subcutaneous allergen immunotherapy in asthmatic children sensitized to house dust mite. Allergy. 2014; 69:246–53.
Article
80. Li XM, Zhang TF, Huang CK, Srivastava K, Teper AA, Zhang L, Schofield BH, Sampson HA. Food allergy herbal formula-1 (FAHF-1) blocks peanut-induced anaphylaxis in a murine model. J Allergy Clin Immunol. 2001; 108:639–46.
Article
81. Srivastava KD, Kattan JD, Zou ZM, Li JH, Zhang L, Wallenstein S, Goldfarb J, Sampson HA, Li XM. The Chinese herbal medicine formula FAHF-2 completely blocks anaphylactic reactions in a murine model of peanut allergy. J Allergy Clin Immunol. 2005; 115:171–8.
Article
82. Wang J, Patil SP, Yang N, Ko J, Lee J, Noone S, Sampson HA, Li XM. Safety, tolerability, and immunologic effects of a food allergy herbal formula in food allergic individuals: a randomized, double-blinded, placebo-controlled, dose escalation, phase 1 study. Ann Allergy Asthma Immunol. 2010; 105:75–84.
Article
83. Wang J, Jones SM, Pongracic JA, Song Y, Yang N, Sicherer SH, Makhija MM, Robison RG, Moshier E, Godbold J, Sampson HA, Li XM. Safety, clinical, and immunologic efficacy of a Chinese herbal medicine (food allergy herbal formula-2) for food allergy. J Allergy Clin Immunol. 2015; 136:962–70. e1.
Article
84. Du Toit G, Roberts G, Sayre PH, Bahnson HT, Radulovic S, Santos AF, Brough HA, Phippard D, Basting M, Feeney M, Turcanu V, Sever ML, Gomez Lorenzo M, Plaut M. Lack GLEAP Study Team. Randomized trial of peanut consumption in infants at risk for peanut allergy. N Engl J Med. 2015; 372:803–13.
Article
85. Perkin MR, Logan K, Tseng A, Raji B, Ayis S, Peacock J, Brough H, Marrs T, Radulovic S, Craven J, Flohr C. Lack GEAT Study Team. Randomized trial of introduction of allergenic foods in breast-fed infants. N Engl J Med. 2016; 374:1733–43.
Article
86. Wei-Liang Tan J, Valerio C, Barnes EH, Turner PJ, Van Asperen PA, Kakakios AM. Campbell DEBeating Egg Allergy Trial (BEAT) Study Group. A randomized trial of egg introduction from 4 months of age in infants at risk for egg allergy. J Allergy Clin Immunol. 2017; 139:1621–8. e8.
87. Hamelmann E, Herz U, Holt P, Host A, Lauener RP, Matricardi PM, Wahn U, Wickman M. New visions for basic research and primary prevention of pediatric allergy: an iPAC summary and future trends. Pediatr Allergy Immunol. 2008; 19(Suppl 19):4–16.
Article
88. Senti G, Prinz Vavricka BM, Erdmann I, Diaz MI, Markus R, McCormack SJ, Simard JJ, Wüthrich B, Crameri R, Graf N, Johansen P, Kündig TM. Intralymphatic allergen administration renders specific immunotherapy faster and safer: a randomized controlled trial. Proc Natl Acad Sci U S A. 2008; 105:17908–12.
Article
89. Senti G, Crameri R, Kuster D, Johansen P, Martinez-Gomez JM, Graf N, Steiner M, Hothorn LA, Grönlund H, Tivig C, Zaleska A, Soyer O, van Hage M, Akdis CA, Akdis M, Rose H, Kündig TM. Intralymphatic immunotherapy for cat allergy induces tolerance after only 3 injections. J Allergy Clin Immunol. 2012; 129:1290–6.
Article
90. Witten M, Malling HJ, Blom L, Poulsen BC, Poulsen LK. Is intralymphatic immunotherapy ready for clinical use in patients with grass pollen allergy? J Allergy Clin Immunol. 2013; 132:1248–1252. e5.
Article
91. Lee SP, Choi SJ, Joe E, Lee SM, Lee MW, Shim JW, Kim YJ, Kyung SY, Park JW, Jeong SH, Jung JH. A pilot study of intralymphatic immunotherapy for house dust mite, cat, and dog allergies. Allergy Asthma Immunol Res. 2017; 9:272–7.
Article
92. Lee SP, Jung JH, Lee SM, Joe E, Kang IG, Kim ST, Lee MW, Park SH, Choi SJ. Intralymphatic immunotherapy alleviates allergic symptoms during allergen exposure in daily life. Allergy Asthma Immunol Res. 2018; 10:180–1.
Article
93. Kim ST, Park SH, Lee SM, Lee SP. Allergen-specific intralymphatic immunotherapy in human and animal studies. Asia Pac Allergy. 2017; 7:131–7.
Article
94. Senti G, von Moos S, Tay F, Graf N, Johansen P, Kündig TM. Determinants of efficacy and safety in epicutaneous allergen immunotherapy: summary of three clinical trials. Allergy. 2015; 70:707–10.
Article
95. Senti G, von Moos S, Tay F, Graf N, Sonderegger T, Johansen P, Kündig TM. Epicutaneous allergen-specific immunotherapy ameliorates grass pollen-induced rhinoconjunctivitis: A double-blind, placebo-controlled dose escalation study. J Allergy Clin Immunol. 2012; 129:128–35.
96. Dupont C, Kalach N, Soulaines P, Legoué-Morillon S, Piloquet H, Benhamou PH. Cow's milk epicutaneous immunotherapy in children: a pilot trial of safety, acceptability, and impact on allergic reactivity. J Allergy Clin Immunol. 2010; 125:1165–7.
97. Kitaoka M, Shin Y, Kamiya N, Kawabe Y, Kamihira M, Goto M. Transcutaneous peptide immunotherapy of japanese cedar pollinosis using solid-in-oil nanodispersion technology. AAPS PharmSciTech. 2015; 16:1418–24.
Article
Full Text Links
  • APA
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr