Patient factors influencing acute gluten reactions and cytokine release in treated coeliac disease

Jason A Tye-Din, A James M Daveson, Kaela E Goldstein, Holly L Hand, Kristin M Neff, Gautam Goel, Leslie J Williams, Kenneth E Truitt, Robert P Anderson, RESET CeD Study Group, A Adams, J Andrews, C Behrend, G Brown, S Chen Yi Mei, A Coates, A J Daveson, A DiMarino, H Ee, D Elliott, R Epstein, B Feyen, R Fogel, K Friedenberg, R Gearry, M Gerdis, M Goldstein, V Gupta, R Holmes, G Holtmann, S Idarraga, G James, T King, T Klein, S Kupfer, B Lebwohl, J Lowe, J Murray, E Newton, D Quinn, D Radin, T Ritter, H Stacey, C Strout, R Stubbs, S Thackwray, V Trivedi, J A Tye-Din, J Weber, S Wilson, Jason A Tye-Din, A James M Daveson, Kaela E Goldstein, Holly L Hand, Kristin M Neff, Gautam Goel, Leslie J Williams, Kenneth E Truitt, Robert P Anderson, RESET CeD Study Group, A Adams, J Andrews, C Behrend, G Brown, S Chen Yi Mei, A Coates, A J Daveson, A DiMarino, H Ee, D Elliott, R Epstein, B Feyen, R Fogel, K Friedenberg, R Gearry, M Gerdis, M Goldstein, V Gupta, R Holmes, G Holtmann, S Idarraga, G James, T King, T Klein, S Kupfer, B Lebwohl, J Lowe, J Murray, E Newton, D Quinn, D Radin, T Ritter, H Stacey, C Strout, R Stubbs, S Thackwray, V Trivedi, J A Tye-Din, J Weber, S Wilson

Abstract

Background: Patients with coeliac disease (CD) commonly report a variety of adverse symptoms to gluten, but descriptions of the symptomatic response in the literature may have been confounded by the presence of food components such as fermentable carbohydrates (FODMAPs) causing symptoms of irritable bowel syndrome independent of gluten. In recent unmasked and masked low FODMAP gluten challenge studies in small groups of treated CD patients, nausea and vomiting were shown to be the key symptoms associated with serum interleukin (IL)-2 release. Our objective was to utilise a large and diverse cohort of people with CD undertaking a standardised gluten food challenge to characterise the demographic, genetic and clinical factors influencing the severity and timing of acute gluten reactions and IL-2 production.

Methods: A total of 295 adults treated for CD were observed for 6 h after an unmasked food challenge consisting of 10 g vital wheat gluten (low in FODMAPs) in 100 ml water. Assessments included patient-reported outcomes, serum IL-2 and adverse events. Responses were analysed according to patient characteristics, HLA-DQ genotype, duodenal histology and response to a second gluten challenge.

Results: Peak symptom severity was at 3 h (median severity 5/10). Peak IL-2 was at 4 h (median 4 pg/ml, range undetectable to 1028 pg/ml). Older age, older age at diagnosis, HLA-DQ2.5 positivity and homozygosity for HLA-DQB1*02 were each significantly associated with IL-2 elevations after gluten. Patients positive for HLA-DQ2.5, DQ8, DQ2.2 or DQ7 showed elevated IL-2 after gluten. Patient factors were not significantly associated with severity of digestive symptoms, but symptoms were correlated to one another and serum IL-2. Gluten challenge after 5 months caused more vomiting and higher IL-2 levels, but responses correlated with the first.

Conclusions: Gluten-induced symptoms and cytokine release is common in adults with treated CD. Age, genetics and previous response to gluten predict these acute reactions to gluten challenge. Structured symptom assessment and serum IL-2 after standardised gluten challenge may inform on patient diagnosis, the role of gluten in symptomatology and the need for adjunctive treatment.

Trial registration: ClinicalTrials.gov , NCT03644069 Registered 21 May 2018.

Keywords: Coeliac disease; Cytokine; Food challenge; Gluten; Patient-reported outcome.

Conflict of interest statement

GG, KEG, HLH, KMN, KET, LJW and RPA were former employees of ImmusanT, Inc. JAT-D and AJMD served as advisors to ImmusanT, Inc. RPA is the inventor of patents, owned or licenced by ImmusanT, Inc., relating to the diagnostic application of gluten challenge.

Figures

Fig. 1
Fig. 1
Study outline
Fig. 2
Fig. 2
Six-hour time series after gluten for patient-reported outcome scores (modified CeD PRO and Global Symptom Survey). Profiles are for all patients (ac) and for patients whose worst global gastrointestinal score over 6 h was “no symptoms” (df), “mild symptoms” (gi), “very mild symptoms” (jl), “moderate symptoms” (mo) and “severe symptoms” (pr), “very severe symptoms” (su). Points indicate mean ± standard error of the mean for scores rated 0 to 10
Fig. 3
Fig. 3
Serum IL-2 elevation in 295 patients over 6 h after gluten challenge. a Median and interquartile range of serum IL-2 concentrations at 2-h intervals after gluten; significance tested by Wilcoxon matched-pairs signed rank test. b The frequency distribution of peak serum IL-2 concentrations in patients after gluten. c Median and interquartile range of serum IL-2 concentrations at 4 h and peak hourly numerical rating for global severity of digestive symptoms (GloSS). d Median and interquartile range of serum IL-2 concentrations 4 h and peak hourly severity of nausea. e Median and interquartile range of serum IL-2 concentrations at 4 h and whether patients experienced vomiting after gluten challenge. f Mean and standard error of the mean of serum IL-2 concentrations after gluten according to gluten reaction severity (peak severity descriptor of digestive symptoms rated each hour in the GloSS). g Peak serum IL-2 concentrations after gluten according to villous height to crypt depth ratio (VH:CrD) in the second part duodenal biopsies indicating the presence of villous atrophy (VH:CrD ≤ 2) (n = 39 patients) or absence (n = 13). h Paired serum IL-2 concentrations at 4 h after two separate food challenges with 6-g gluten protein. Data for 36 patients who received placebo treatment during the RESET CeD Study and had both a screening (unmasked) and a second (masked) challenge 20 to 22 weeks later. The gluten challenges used the same format administering the equivalent of 6-g gluten protein in 10-g vital wheat gluten mixed in water

References

    1. Ludvigsson JF, Leffler DA, Bai JC, Biagi F, Fasano A, Green PH, Hadjivassiliou M, Kaukinen K, Kelly CP, Leonard JN, et al. The Oslo definitions for coeliac disease and related terms. Gut. 2013;62(1):43–52. doi: 10.1136/gutjnl-2011-301346.
    1. Caio G, Volta U, Sapone A, Leffler DA, De Giorgio R, Catassi C, Fasano A. Celiac disease: a comprehensive current review. BMC Med. 2019;17(1):142. doi: 10.1186/s12916-019-1380-z.
    1. Silvester JA, Graff LA, Rigaux L, Walker JR, Duerksen DR. Symptomatic suspected gluten exposure is common among patients with coeliac disease on a gluten-free diet. Aliment Pharmacol Ther. 2016;44(6):612–619.
    1. Skodje GI, Sarna VK, Minelle IH, Rolfsen KL, Muir JG, Gibson PR, Veierod MB, Henriksen C, Lundin KEA. Fructan, rather than gluten, induces symptoms in patients with self-reported non-celiac gluten sensitivity. Gastroenterology. 2018;154(3):529–539.
    1. Colloca L, Barsky AJ. Placebo and nocebo effects. N Engl J Med. 2020;382(6):554–561.
    1. Daveson AJM, Tye-Din JA, Goel G, Goldstein KE, Hand HL, Neff KM, Williams LJ, Truitt KE, Anderson RP, Group RCS Masked bolus gluten challenge low in FODMAPs implicates nausea and vomiting as key symptoms associated with immune activation in treated coeliac disease. Aliment Pharmacol Ther. 2020;51(2):244–252.
    1. Tye-Din JA, Daveson AJM, Ee HC, Goel G, MacDougall J, Acaster S, Goldstein KE, Dzuris JL, Neff KM, Truitt KE, et al. Elevated serum interleukin-2 after gluten correlates with symptoms and is a potential diagnostic biomarker for coeliac disease. Aliment Pharmacol Ther. 2019;50(8):901–910.
    1. Goel G, Tye-Din JA, Qiao SW, Russell AK, Mayassi T, Ciszewski C, Sarna VK, Wang S, Goldstein KE, Dzuris JL, et al. Cytokine release and gastrointestinal symptoms after gluten challenge in celiac disease. Sci Adv. 2019;5(8):eaaw7756.
    1. Tye-Din JA, Skodje GI, Sarna VK, Dzuris JL, Russell AK, Goel G, Wang S, Goldstein KE, Williams LJ, Sollid LM, et al. Cytokine release after gluten ingestion differentiates coeliac disease from self-reported gluten sensitivity. United European Gastroenterol J. 2020;8(1):108–118.
    1. Goel G, Daveson AJM, Hooi CE, Tye-Din JA, Wang S, Szymczak E, Williams LJ, Dzuris JL, Neff KM, Truitt KE, et al. Serum cytokines elevated during gluten-mediated cytokine release in coeliac disease. Clin Exp Immunol. 2020;199(1):68–78.
    1. Goel G, King T, Daveson AJ, Andrews JM, Krishnarajah J, Krause R, Brown GJE, Fogel R, Barish CF, Epstein R, et al. Epitope-specific immunotherapy targeting CD4-positive T cells in coeliac disease: two randomised, double-blind, placebo-controlled phase 1 studies. Lancet Gastroenterol Hepatol. 2017;2(7):479–493.
    1. Varney J, Barrett J, Scarlata K, Catsos P, Gibson PR, Muir JG. FODMAPs: food composition, defining cutoff values and international application. J Gastroenterol Hepatol. 2017;32(Suppl 1):53–61.
    1. Leffler DA, Dennis M, Edwards George JB, Jamma S, Magge S, Cook EF, Schuppan D, Kelly CP. A simple validated gluten-free diet adherence survey for adults with celiac disease. Clin Gastroenterol Hepatol. 2009;7(5):530–536.
    1. Daveson AJM, Popp A, Taavela J, Goldstein KE, Isola J, Truitt KE, Maki M, Anderson RP, Group RCS Baseline quantitative histology in therapeutics trials reveals villus atrophy in most patients with coeliac disease who appear well controlled on gluten-free diet. GastroHep. 2020;2(1):22–30.
    1. Tinto N, Cola A, Piscopo C, Capuano M, Galatola M, Greco L, Sacchetti L. High frequency of haplotype HLA-DQ7 in celiac disease patients from South Italy: retrospective evaluation of 5,535 subjects at risk of celiac disease. PLoS One. 2015;10(9):e0138324.
    1. Tye-Din JA, Cameron DJ, Daveson AJ, Day AS, Dellsperger P, Hogan C, Newnham ED, Shepherd SJ, Steele RH, Wienholt L, et al. Appropriate clinical use of human leukocyte antigen typing for coeliac disease: an Australasian perspective. Intern Med J. 2015;45(4):441–450.
    1. Lee DW, Santomasso BD, Locke FL, Ghobadi A, Turtle CJ, Brudno JN, Maus MV, Park JH, Mead E, Pavletic S, et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol Blood Marrow Transplant. 2019;25(4):625–638.
    1. Baumgart DC, Lowder JN, Targan SR, Sandborn WJ, Frankel MB. Transient cytokine-induced liver injury following administration of the humanized anti-CD3 antibody visilizumab (HuM291) in Crohn’s disease. Am J Gastroenterol. 2009;104(4):868–876.
    1. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra138.
    1. Al-Toma A, Goerres MS, Meijer JW, Pena AS, Crusius JB, Mulder CJ. Human leukocyte antigen-DQ2 homozygosity and the development of refractory celiac disease and enteropathy-associated T-cell lymphoma. Clin Gastroenterol Hepatol. 2006;4(3):315–319.
    1. Karell K, Louka AS, Moodie SJ, Ascher H, Clot F, Greco L, Ciclitira PJ, Sollid LM, Partanen J. Hla types in celiac disease patients not carrying the DQA1*05-DQB1*02 (DQ2) heterodimer: results from the European Genetics Cluster on Celiac Disease. Hum Immunol. 2003;64(4):469–477.

Source: PubMed

3
구독하다