Changes in antigen-specific T-cell number and function during oral desensitization in cow's milk allergy enabled with omalizumab

D Bedoret, A K Singh, V Shaw, E G Hoyte, R Hamilton, R H DeKruyff, L C Schneider, K C Nadeau, D T Umetsu, D Bedoret, A K Singh, V Shaw, E G Hoyte, R Hamilton, R H DeKruyff, L C Schneider, K C Nadeau, D T Umetsu

Abstract

Food allergy is a major public health problem, for which there is no effective treatment. We examined the immunological changes that occurred in a group of children with significant cow's milk allergy undergoing a novel and rapid high-dose oral desensitization protocol enabled by treatment with omalizumab (anti-immunoglobulin (Ig)E monoclonal antibodies). Within a week of treatment, the CD4(+) T-cell response to milk was nearly eliminated, suggesting anergy in, or deletion of, milk-specific CD4(+) T cells. Over the following 3 months while the subjects remained on high doses of daily oral milk, the CD4(+) T-cell response returned, characterized by a shift from interleukin-4 to interferon-γ production. Desensitization was also associated with reduction in milk-specific IgE and a 15-fold increase in milk-specific IgG4. These studies suggest that high-dose oral allergen desensitization may be associated with deletion of allergen-specific T cells, without the apparent development of allergen-specific Foxp3(+) regulatory T cells.

Trial registration: ClinicalTrials.gov NCT00968110.

Conflict of interest statement

Conflict of interest: the authors declare that no conflict of interest exists.

Figures

Figure 1. Milk-induced CD4 + T cell…
Figure 1. Milk-induced CD4+ T cell proliferation is greatly reduced during the rush desensitization phase
(a) Frozen PBMC, isolated from 5 milk allergic patients undergoing rush milk desensitization, were thawed and labeled with CFSE, and cultured in the presence of tetanus toxoid (TT) or milk proteins (MP) for 7 days. Cells were then collected, stained with anti-CD4 mAb, and analyzed by flow cytometry. The number in each panel represents the fraction of total CD4+ cells that are within the CFSElow CD4+ gate (antigen-specific proliferating cells) on day 0. Mean CD4+ milk-specific proliferating cells=7.75%; mean CD4+ tetanus specific cells=24.6%. Mean CD4+ proliferating cells without antigen=0.62%. (b) Each line represents data for all 5 patients combined (solid line, milk protein (MP); dashed line, tetanus toxoid (TT)). Data represent mean % antigen-specific cells (CFSElow CD4+ cells) of total CD4+ cells, normalized to day 0 for each patient ± SEM, over the course of the study. ** p < 0.01, *** p < 0.001 versus baseline and week 8, determined using paired t-test. ° p < 0.05 versus TT response, determined using two-way ANOVA for matched values with Bonferroni’s post hoc test. (c) Each line represents data for an individual patient, of % milk-specific CFSElow CD4+ cells of total CD4+ cells, normalized to day 0 for each patient, over the course of the study.
Figure 2. Milk specific Foxp3 + T…
Figure 2. Milk specific Foxp3+ T cells are not detected during the milk desensitization
(a–c) CFSE-labeled PBMC isolated from 5 milk allergic patients undergoing milk desensitization were cultured in the presence of milk proteins for 7 days. Cells were then collected, fixed and permeabilized, stained with anti-CD4 and anti–Foxp3, and analyzed by flow cytometry. (a–b) Figures represent combined percentages of CD4+ Foxp3+ cells of total cells normalized to day 0. Data represent mean ± SEM. (a) Normalized CD4+ CFSElow cells (proliferating cells, representing milk-specific TReg cells). (b) Normalized CD4+ CFSEhigh cells (nonproliferating cells, representing natural TReg cells). (c) Individual dot plots of flow cytometry analysis are shown. CD4+CFSElow T cells (left panels) represent milk-specific TReg cells, which did not increase over time. CD4+CFSEhigh T cells (right panels) represent natural TReg cells, which did not change over time.
Figure 3. Functional regulatory T cells are…
Figure 3. Functional regulatory T cells are not detected during the milk desensitization
(a–b) CFSE-labeled PBMC isolated from 5 patients undergoing milk desensitization were cultured in the presence of milk (MP) for 7 days. Cells were then collected, stained with anti-CD4 mAb, and analyzed by flow cytometry. (a) Frozen samples from multiple time points were thawed and cultured with or without anti-IL-10 mAb and milk proteins. Percentages of the milk-specific cell populations (CFSElow) within the CD4+ gate are shown. Results with and without anti-IL-10 mAb overlap, indicating an absence of IL-10 mediated suppression. (b) PBMC from week 12 were cultured with or without anti-IL-10, anti-TGF-β mAb or both and compared to baseline PBMC. Figures represent percentages of the milk-specific cell populations (CFSElow) within the CD4+ gate. No increase in proliferation was observed with the mAbs, indicating an absence of IL-10 or TGF-β mediated suppression. (c) CD4+ T cells and regulatory T cells were purified from week 12 and week 14 PBMC, respectively. 1× 104 (+) or 5 × 104 (++) CD4+ CD25+ cells, or 1.5 × 104 (+) or 7.5 × 104 (++) CD4+ cells were then cocultured with 2 × 105 baseline PBMC in the presence of milk proteins. The proliferation was measured as [3H]thymidine incorporation during the last 16 hours of a 6-day culture.
Figure 4. Evidence of milk-specific CD4 +…
Figure 4. Evidence of milk-specific CD4+ T cell anergy
(a). PBMC were isolated from week 12 samples of 5 milk allergic patients undergoing milk desensitization protocol, labeled with CFSE, cultured in the presence of milk proteins (MP) for 7 days and treated with or without IL-2. Cells were then collected and stained with anti-CD4 mAb. Proliferation was analyzed by flow cytometry and compared to PBMC proliferation from baseline samples. Figure shows percentages of the milk-specific cells (CFSElow) within the CD4+ gate as mean ± SEM for 5 patients. ** p < 0.01, *** p < 0.001. Significance was determined using paired t-test. (b–d). Phenotype of the milk specific CD4+ T cells during desensitization. CFSE-labeled PBMC isolated from all 5 of the milk allergic patients undergoing milk desensitization were cultured in the presence of milk proteins for 7 days, then analyzed by flow cytometry. (b) Data represent CD25 expression (mean CD25 MFI (mean fluorescence intensity)) by milk specific (CD4+ CFSElow) cells ± SEM for the 5 patients evaluated, normalized to results on day 0. (c) Solid line represents CD25 expression (mean MFI) for milk specific cells from the 4 patients who were desensitized and who passed the DBPCFC, normalized to results on day 0. Dashed line represents CD25 expression (MFI) by milk specific cells from patient #3, who was only partially desensitized, normalized to results on day 0. * p < 0.05 versus baseline and week 8, using paired t-test. (d) Each line represents CD25 expression (MFI) on milk specific cells from each patient normalized to results on day 0. Patient #3 was only partially desensitized.
Figure 5. Evidence for immune deviation during…
Figure 5. Evidence for immune deviation during milk desensitization
(a–c) PBMC isolated from 5 milk allergic patients undergoing milk desensitization, labeled with CFSE, and cultured in the presence of antigens for 7 days, were restimulated with PMA and ionomycin in the presence of monensin for 5 h. Cytokine production was then assessed using intracellular IL-4, IFN-γ and IL-13 staining. (a) Data for all 5 patients. IFN-γ/IL-4 production ratio for the antigen-specific (CFSElow) CD4+ T cells, normalized to day 0 is shown (mean ± SEM). (b) Solid line represents data from (A), for desensitized patients (patients #1, 2, 4 and 5), and dotted line represents data from (A), for the partially desensitized patient (patient #3). * p < 0.05 versus baseline and week 8, using paired t-test. (c) Each line represents data for an individual patient, representing IFN-γ/IL-4 production ratio by the antigen-specific CD4+ T cells, normalized to day 0. (d) IL-13 production was also assessed in CD4+ T cells from patient #5. Data represents IFN-γ/IL-13 production by milk-specific CD4+ T cells. (e–f) Serum milk-specific IgE (e) and IgG4 (f) levels were determined for the 10 patients who completed the study. Points represent data from different individuals and bars show mean values ± SEM. * p < 0.05, ** p < 0.01 versus baseline, determined using nonparametric Wilcoxon matched-pairs test.
Figure 6. Milk skin test reactivity and…
Figure 6. Milk skin test reactivity and in vitro basophil activation are reduced by oral milk desensitization
(a) Skin prick test wheal with cow’s milk allergen extract (n=10) was reduced at week 52 compared to week 0. ** P < 0.01 determined using paired t-test. ° patient #3, who was slow to become desensitized. (b) Skin prick test wheal with cashew or ovalbumin did not change, comparing week 0 and week 52 time points in 4 patients known to have those allergies. (c) Basophils in whole blood, taken on day 0, but not subsequently, when stimulated ex vivo for 20 minutes with cow’s milk (CM), were activated, as assessed by expression of CD203c, CD63 and histamine release (n=5). Treatment with vehicle/glycerine showed no shift in CD203c or CD63 expression (data not shown). MFI = median fluorescence intensity. (d) Basophil activation with cashew or ovalbumin was blocked by in vivo treatment with omalizumab, as assessed on weeks 8, 9, 10 and 16. Prior to and after omalizumab treatment, basophil activation with cashew or ovalbumin was robust, as assessed by expression of CD203c, CD63 and histamine release.

References

    1. Noon L. Prophylactic inoculation for hay fever. Lancet. 1911;1:1572.
    1. Larche M, Akdis CA, Valenta R. Immunological mechanisms of allergen-specific immunotherapy. Nat Rev Immunol. 2006;6:761–71.
    1. Jutel M, Akdis CA. Immunological mechanisms of allergen-specific immunotherapy. Allergy. 2011 in press.
    1. Secrist H, Chelen CJ, Wen Y, Marshall JD, Umetsu DT. Allergen immunotherapy decreases interleukin 4 production in CD4+ T cells from allergic individuals. J Exp Med. 1993;178:2123–30.
    1. Varney VA, Hamid QA, Gaga M, Ying S, Jacobson M, Frew AJ, et al. Influence of grass pollen immunotherapy on cellular infiltration and cytokine mRNA expression during allergen-induced late-phase cutaneous responses. J Clin Invest. 1993;92:644–51.
    1. Akdis CA, Blesken T, Akdis M, Wuthrich B, Blaser K. Role of interleukin 10 in specific immunotherapy. J Clin Invest. 1998;102:98–106.
    1. 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;205:2887–98.
    1. Weiner HL. Oral tolerance for the treatment of autoimmune diseases. Ann Rev Med. 1997;48:341–51.
    1. de Jong EC, Spanhaak S, Martens BP, Kapsenberg ML, Penninks AH, Wierenga EA. Food allergen (peanut)-specific TH2 clones generated from the peripheral blood of a patient with peanut allergy. J Allergy Clin Immunol. 1996;98:73–81.
    1. Higgins JA, Lamb JR, Lake RA, O’Hehir RE. Polyclonal and clonal analysis of human CD4+ T-lymphocyte responses to nut extracts. Immunology. 1995;84:91–7.
    1. Dorion BJ, Burks AW, Harbeck R, Williams LW, Trumble A, Helm RM, et al. The production of interferon-gamma in response to a major peanut allergy, Ara h II correlates with serum levels of IgE anti-Ara h II. J Allergy Clin Immunol. 1994;93:93–9.
    1. Turcanu V, Maleki SJ, Lack G. Characterization of lymphocyte responses to peanuts in normal children, peanut-allergic children, and allergic children who acquired tolerance to peanuts. J Clin Invest. 2003;111:1065–72.
    1. Shreffler WG, Wanich N, Moloney M, Nowak-Wegrzyn A, Sampson HA. Association of allergen-specific regulatory T cells with the onset of clinical tolerance to milk protein. J Allergy Clin Immunol. 2009;123:43–52. e7.
    1. Pajno GB, Caminiti L, Ruggeri P, De Luca R, Vita D, La Rosa M, et al. Oral immunotherapy for cow’s milk allergy with a weekly up-dosing regimen: a randomized single-blind controlled study. Ann Allergy Asthma Immunol. 2010;105:376–81.
    1. Buchanan AD, Green TD, Jones SM, Scurlock AM, Christie L, Althage KA, et al. Egg oral immunotherapy in nonanaphylactic children with egg allergy. J Allergy Clin Immunol. 2007;119:199–205.
    1. Nash S, Steele P, Kamilaris J, Pons L, Kulis M, Lee L, et al. Oral peanut immunotherapy for children with peanut allergy. J Allergy Clin Immunol. 2008;121:S147.
    1. Hofmann AM, Scurlock AM, Jones SM, Palmer KP, Lokhnygina Y, Steele PH, et al. Safety of a peanut oral immunotherapy protocol in children with peanut allergy. J Allergy Clin Immunol. 2009;124:286–91. 91, e1–6.
    1. Blumchen K, Ulbricht H, Staden U, Dobberstein K, Beschorner J, de Oliveira LC, et al. Oral peanut immunotherapy in children with peanut anaphylaxis. J Allergy Clin Immunol. 2010;126:83–91. e1.
    1. Nadeau K, Schneider L, Hoyte E, Borras I, Umetsu D. Rapid oral desensitization in combination with omalizumab therapy in patients with cow’s milk allergy. J Allergy Clin Immunol. 2011;127:1722–4.
    1. Faria AM, Weiner HL. Oral tolerance. Immunol Rev. 2005;206:232–59.
    1. Hatachi S, Iwai Y, Kawano S, Morinobu S, Kobayashi M, Koshiba M, et al. CD4+ PD-1+ T cells accumulate as unique anergic cells in rheumatoid arthritis synovial fluid. J Rheumatol. 2003;30:1410–9.
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–45.
    1. Sicherer SH, Sampson HA. 9 Food allergy. J Allergy Clin Immunol. 2006;117:S470–5.
    1. Boyce JA, Assa’ad A, Burks AW, Jones SM, Sampson HA, Wood RA, et al. Guidelines for the Diagnosis and Management of Food Allergy in the United States: Summary of the NIAID-Sponsored Expert Panel Report. J Allergy Clin Immunol. 126:1105–18.
    1. Sicherer SH, Sampson HA. Food allergy. J Allergy Clin Immunol. 2010;125:S116–25.
    1. Meglio P, Bartone E, Plantamura M, Arabito E, Giampietro PG. A protocol for oral desensitization in children with IgE-mediated cow’s milk allergy. Allergy. 2004;59:980–7.
    1. Longo G, Barbi E, Berti I, Meneghetti R, Pittalis A, Ronfani L, et al. Specific oral tolerance induction in children with very severe cow’s milk-induced reactions. J Allergy Clin Immunol. 2008;121:343–7.
    1. Staden U, Blumchen K, Blankenstein N, Dannenberg N, Ulbricht H, Dobberstein K, et al. Rush oral immunotherapy in children with persistent cow’s milk allergy. J Allergy Clin Immunol. 2008;122:418–9.
    1. Clark AT, Islam S, King Y, Deighton J, Anagnostou K, Ewan PW. Successful oral tolerance induction in severe peanut allergy. Allergy. 2009;64:1218–20.
    1. Enrique E, Pineda F, Malek T, Bartra J, Basagana M, Tella R, et al. Sublingual immunotherapy for hazelnut food allergy: a randomized, double-blind, placebo-controlled study with a standardized hazelnut extract. J Allergy Clin Immunol. 2005;116:1073–9.
    1. Skripak JM, Matsui EC, Mudd K, Wood RA. The natural history of IgE-mediated cow’s milk allergy. J Allergy Clin Immunol. 2007;120:1172–7.
    1. Francis J, Till S, Durham S. Induction of IL-10+CD4+CD25+ T cells by grass pollen immunotherapy. J Allergy Clin Immunol. 2003;111:1255–61.
    1. Oldfield W, Larche M, Kay A. Effect of T-cell peptides derived from Fel d 1 on allergic reactions and cytokine production in patients sensitive to cats: a randomised controlled trial. Lancet. 2002;360:47–53.
    1. Verhoef A, Alexander C, Kay AB, Larche M. T cell epitope immunotherapy induces a CD4+ T cell population with regulatory activity. PLoS Med. 2005;2:e78.
    1. Aslam A, Chan H, Warrell DA, Misbah S, Ogg GS. Tracking antigen-specific T-cells during clinical tolerance induction in humans. PLoS One. 2010;5:e11028.
    1. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500.
    1. Varshney P, Jones SM, Scurlock AM, Perry TT, Kemper A, Steele P, et al. A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. J Allergy Clin Immunol. 2011;127:654–60.
    1. Jones SM, Pons L, Roberts JL, Scurlock AM, Perry TT, Kulis M, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol. 2009;124:292–300. e1–97.
    1. Mori F, Bianchi L, Pucci N, Azzari C, De Martino M, Novembre E. CD4+CD25+Foxp3+ T regulatory cells are not involved in oral desensitization. Int J Immunopathol Pharmacol. 23:359–61.
    1. Kim EH, Bird JA, Kulis M, Laubach S, Pons L, Shreffler W, et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2011;127:640–6. e1.
    1. Tiemessen MM, Van Hoffen E, Knulst AC, Van Der Zee JA, Knol EF, Taams LS. CD4 CD25 regulatory T cells are not functionally impaired in adult patients with IgE-mediated cow’s milk allergy. J Allergy Clin Immunol. 2002;110:934–6.
    1. Nagata S, McKenzie C, Pender SL, Bajaj-Elliott M, Fairclough PD, Walker-Smith JA, et al. Human Peyer’s patch T cells are sensitized to dietary antigen and display a Th cell type 1 cytokine profile. J Immunol. 2000;165:5315–21.
    1. Stock P, Kallinich T, Akbari O, Quarcoo D, Gerhold K, Wahn U, et al. CD8(+) T cells regulate immune responses in a murine model of allergen-induced sensitization and airway inflammation. Eur J Immunol. 2004;34:1817–27.
    1. Dominguez-Villar M, Baecher-Allan CM, Hafler DA. Identification of T helper type 1-like, Foxp3(+) regulatory T cells in human autoimmune disease. Nat Med. 2011;17:673–5.
    1. Scurlock AM, Vickery BP, Hourihane JO, Burks AW. Pediatric food allergy and mucosal tolerance. Mucosal Immunol. 3:345–54.
    1. van Halteren HK, van der Linden PW, Burgers JA, Bartelink AK. Discontinuation of yellow jacket venom immunotherapy: follow-up of 75 patients by means of deliberate sting challenge. J Allergy Clin Immunol. 1997;100:767–70.
    1. Cox L, Cohn JR. Duration of allergen immunotherapy in respiratory allergy: when is enough, enough? Ann Allergy Asthma Immunol. 2007;98:416–26.
    1. Varshney P, Jones S, Pons L, Kulis M, Steele P, Kemper A, et al. Peanut Oral Immunotherapy (OIT) induces immunologic changes supporting the development of tolerance. J Allergy Clin Immunol. 2010;125:AB59 (Abstract).
    1. Gernez Y, Tirouvanziam R, Yu G, Ghosn EE, Reshamwala N, Nguyen T, et al. Basophil CD203c Levels Are Increased at Baseline and Can Be Used to Monitor Omalizumab Treatment in Subjects with Nut Allergy. Int Arch Allergy Immunol. 2010;154:318–27.

Source: PubMed

3
Prenumerera