244
Views
0
CrossRef citations to date
0
Altmetric
Review

The role of gluten challenge in the diagnosis of celiac disease: a review

, , &
Pages 691-700 | Received 26 Mar 2023, Accepted 26 May 2023, Published online: 30 May 2023

References

  • Lindfors K, Ciacci C, Kurppa K, et al. Coeliac disease. Nat Rev Dis Primers. 2019;5(1):3. DOI:10.1038/s41572-018-0054-z
  • Husby S, Koletzko S, Ilma Korponay-Szabó I, et al. European society paediatric gastroenterology, hepatology and nutrition guidelines for diagnosing coeliac disease 2020. J Pediatr Gastroenterol Nutr. 2020;70(1):141–156. DOI:10.1097/MPG.0000000000002497
  • Fuchs V, Kurppa K, Huhtala H, et al. Serology-based criteria for adult coeliac disease have excellent accuracy across the range of pre-test probabilities. Aliment Pharmacol Ther. 2019;49(3):277–284. DOI:10.1111/apt.15109
  • Penny H, Raju S, Lau M, et al. Accuracy of a no-biopsy approach for the diagnosis of coeliac disease across different adult cohorts. Gut. 2021;70(5):876–883. DOI:10.1136/gutjnl-2020-320913
  • McNeish A, Harms H, Rey J, et al. The diagnosis of coeliac disease. A commentary on the current practices of members of the European Society for Paediatric Gastroenterology and Nutrition (ESPGAN). Arch Dis Child. 1979;54(10):783–786. DOI:10.1136/adc.54.10.783
  • Revised criteria for diagnosis of coeliac disease. report of working group of European society of paediatric gastroenterology and nutrition. Arch Dis Child. 1990;65(8):909–911. DOI:10.1136/adc.65.8.909
  • Husby S, Koletzko S, Korponay-Szabó IR. European society for pediatric gastroenterology, hepatology, and nutrition guidelines for the diagnosis of coeliac disease. J Pediatr Gastroenterol Nutr. 2012;54:136–160.
  • Popp A, Kivelä L, Fuchs V, et al. Diagnosing celiac disease: towards wide-scale screening and serology-based criteria? Gastroenterol Res Pract. 2019;2019:1–10.
  • Al-Toma A, Volta U, Auricchio R, et al. European Society for the Study of Coeliac Disease (ESsCD) guideline for coeliac disease and other gluten-related disorders. United Eur Gastroenterol J. 2019;7(5):583–613. DOI:10.1177/2050640619844125
  • Kivelä L, Caminero A, Leffler D, et al. Current and emerging therapies for coeliac disease. Nat Rev Gastroenterol Hepatol. 2021;18(3):181–195. DOI:10.1038/s41575-020-00378-1
  • Leinonen H, Kivelä L, Lähdeaho ML, et al. Daily life restrictions are common and associated with health concerns and dietary challenges in adult celiac disease patients diagnosed in childhood. Nutrients. 2019;11(8):1718. DOI:10.3390/nu11081718
  • Kivelä L, Eurén A, Repo M, et al. Coexisting type 1 diabetes, persistent symptoms, and financial issues associate with poorer adherence to a gluten-free diet in celiac disease after transition from pediatrics to adult care. Front Nutr. 2022;9:883220.
  • Green P, Paski S, Ko C, et al. AGA clinical practice update on management of refractory celiac disease: expert review. Gastroenterology. 2022;163(5):1461–1469.
  • Gaesser G, Angadi S. Navigating the gluten-free boom. JAAPA. 2015;28(8):1–7.
  • Lebwohl B, Cao Y, Zong G, et al. Long term gluten consumption in adults without celiac disease and risk of coronary heart disease: prospective cohort study. BMJ. 2017;357:1892.
  • Jones A. The gluten-free diet: fad or necessity? Diabetes Spectr. 2017;30(2):118–123.
  • Hakola L, Miettinen ME, Syrjälä E, et al. Association of cereal, gluten, and dietary fiber intake with islet autoimmunity and type 1 diabetes. JAMA Pediatr. 2019;173(10):953–960. DOI:10.1001/jamapediatrics.2019.2564
  • Corazza G, Villanacci V. Coeliac disease. J Clin Pathol. 2005;58(6):573–574.
  • Ravelli A, Villanacci V, Monfredini C, et al. How patchy is patchy villous atrophy? Distribution pattern of histological lesions in the duodenum of children with celiac disease. Am J Gastroenterol. 2010;105(9):2103–2110. DOI:10.1038/ajg.2010.153
  • Taavela J, Koskinen O, Huhtala H, et al. Validation of morphometric analyses of small-intestinal biopsy readouts in celiac disease. PLoS ONE. 2013;8(10):e76163. DOI:10.1371/journal.pone.0076163
  • Werkstetter K, Korponay-Szabó IR, Popp A, et al. Accuracy in diagnosis of celiac disease without biopsies in clinical practice. Gastroenterology. 2017;153(4):924–935. DOI:10.1053/j.gastro.2017.06.002
  • Gustafsson I, Repo M, Popp A, et al. Prevalence and diagnostic outcomes of children with duodenal lesions and negative celiac serology. Dig Liver Dis. 2020;52(3):289–295. DOI:10.1016/j.dld.2019.11.011
  • Aziz I, Peerally M, Barnes JH, et al. The clinical and phenotypical assessment of seronegative villous atrophy; a prospective UK centre experience evaluating 200 adult cases over a 15-year period (2000-2015). Gut. 2017;66(9):1563–1572. DOI:10.1136/gutjnl-2016-312271
  • Ylönen V, Lindfors K, Repo M, et al. Non-biopsy serology-based diagnosis of celiac disease in adults is accurate with different commercial kits and pre-test probabilities. Nutrients. 2020;12(9):2736. DOI:10.3390/nu12092736
  • Schiepatti A, Rej A, Maimaris S, et al. Clinical classification and long-term outcomes of seronegative coeliac disease: a 20-year multicentre follow-up study. Aliment Pharmacol Ther. 2021;54(10):1278–1289. DOI:10.1111/apt.16599
  • Husby S, Murray J, Katzka D. AGA clinical practice update on diagnosis and monitoring of celiac disease: changing utility of serology and histologic measures: expert review. Gastroenterology. 2019;156(4):885–889.
  • Kaukinen K, Partanen J, Mäki M, et al. HLA-DQ typing in the diagnosis of celiac disease. Am J Gastroenterol. 2002;97(3):695–699.
  • Rubio-Tapia A, Hill I, Kelly C, et al. ACG clinical guidelines: diagnosis and management of celiac disease. Am J Gastroenterol. 2013;108(5):656–676. DOI:10.1038/ajg.2013.79
  • Gatti S, Rossi M, Alfonsi S, et al. Beyond the intestinal celiac mucosa: diagnostic role of anti-TG2 deposits, a systematic review. Front Med. 2014;1:9.
  • Koskinen O, Collin P, Lindfors K, et al. Usefulness of small-bowel mucosal transglutaminase-2 specific autoantibody deposits in the diagnosis and follow-up of celiac disease. J Clin Gastroenterol. 2010;44(7):483–488. DOI:10.1097/MCG.0b013e3181b64557
  • Salmi T, Collin P, Korponay-Szabó IR, et al. Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut. 2006;55(12):1746–1753. DOI:10.1136/gut.2005.071514
  • Mearin L, Agardh D, Antunes H, et al. ESPGHAN position paper on management and follow-up of children and adolescents with celiac disease. J Pediatr Gastroenterol Nutr. 2022;75(3):369–386. DOI:10.1097/MPG.0000000000003540
  • van Overbeek F, Uil-Dieterman I, Mol I, et al. The daily gluten intake in relatives of patients with coeliac disease compared with that of the general Dutch population. Eur J Gastroenterol Hepatol. 1997;9(11):1097–1099. DOI:10.1097/00042737-199711000-00013
  • Holm K, Mäki M, Vuolteenaho N, et al. Oats in the treatment of childhood coeliac disease: a 2-year controlled trial and a long-term clinical follow-up study. Aliment Pharmacol Ther. 2006;23(10):1463–1472. DOI:10.1111/j.1365-2036.2006.02908.x
  • Mäki M, Lähdeaho ML, Hällström O, et al. Postpubertal gluten challenge in coeliac disease. Arch Dis Child. 1989;64(11):1604–1607.
  • Korponay-Szabó IR, Kovács JM, Lörincz M, et al. Prospective significance of antiendomysium antibody positivity in subsequently verified celiac disease. J Pediatr Gastroenterol Nutr. 1997;25(1):56–63. DOI:10.1097/00005176-199707000-00009
  • Koskinen O, Villanen M, Korponay-Szabo IR, et al. Oats do not induce systemic or mucosal autoantibody response in children with coeliac disease. J Pediatr Gastroenterol Nutr. 2009;48(5):559–565. DOI:10.1097/MPG.0b013e3181668635
  • Kurppa K, Koskinen O, Collin P, et al. Changing phenotype of celiac disease after long-term gluten exposure. J Pediatr Gastroenterol Nutr. 2008;47(4):500–503. DOI:10.1097/MPG.0b013e31817d8120
  • Hardy M, Girardin A, Pizzey C, et al. Consistency in polyclonal T-cell responses to gluten between children and adults with celiac disease. Gastroenterology. 2015;149(6):1541–1552. DOI:10.1053/j.gastro.2015.07.013
  • Wahab P, Crusius J, Meijer J, et al. Gluten challenge in borderline gluten-sensitive enteropathy. Am J Gastroenterol. 2001;96(5):1464–1469.
  • Cornell H, Macrae F, Melny J, et al. Enzyme therapy for management of coeliac disease. Scand J Gastroenterol. 2005;40(11):1304–1312. DOI:10.1080/00365520510023855
  • Tye-Din J, Stewart J, Dromey J, et al. Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease. Sci Transl Med. 2010;2(41):41ra51. DOI:10.1126/scitranslmed.3001012
  • Brottveit M, Ráki M, Bergseng E, et al. Assessing possible celiac disease by an HLA-DQ2-gliadin tetramer test. Am J Gastroenterol. 2011;106(7):1318–1324. DOI:10.1038/ajg.2011.23
  • Daveson J, Jones D, Gaze S, et al. Effect of hookworm infection on wheat challenge in celiac disease–a randomised double-blinded placebo controlled trial. PLoS ONE. 2011;6(3):e17366. DOI:10.1371/journal.pone.0017366
  • Beitnes AC, Ráki M, Brottveit M, et al. Rapid accumulation of CD14+CD11c+ dendritic cells in gut mucosa of celiac disease after in vivo gluten challenge. PLoS ONE. 2012;7(3):e33556. DOI:10.1371/journal.pone.0033556
  • Camarca A, Radano G, Di Mase R, et al. Short wheat challenge is a reproducible in-vivo assay to detect immune response to gluten. Clin Exp Immunol. 2012;169(2):129–136. DOI:10.1111/j.1365-2249.2012.04597.x
  • Leffler D, Kelly C, Abdallah H, et al. A randomized, double-blind study of larazotide acetate to prevent the activation of celiac disease during gluten challenge. Am J Gastroenterol. 2012;107(10):1554–1562. DOI:10.1038/ajg.2012.211
  • Brottveit M, Beitnes A, Tollefsen S, et al. Mucosal cytokine response after short-term gluten challenge in celiac disease and non-celiac gluten sensitivity. Am J Gastroenterol. 2013;108(5):842–850. DOI:10.1038/ajg.2013.91
  • Leffler D, Schuppan D, Pallav K, et al. Kinetics of the histological, serological and symptomatic responses to gluten challenge in adults with coeliac disease. Gut. 2013;62(7):996–1004. DOI:10.1136/gutjnl-2012-302196
  • Tack G, van de Water J, Bruins M, et al. Consumption of gluten with gluten-degrading enzyme by celiac patients: a pilot-study. World J Gastroenterol. 2013;19(35):5837–5847. DOI:10.3748/wjg.v19.i35.5837
  • López-Palacios N, Pascual V, Castaño M, et al. Evaluation of T cells in blood after a short gluten challenge for coeliac disease diagnosis. Dig Liver Dis. 2018;50(11):1183–1188. DOI:10.1016/j.dld.2018.04.014
  • Sarna V, Skodje G, Reims H, et al. HLA-DQ: gluten tetramer test in blood gives better detection of coeliac patients than biopsy after 14-day gluten challenge. Gut. 2018;67(9):1606–1613. DOI:10.1136/gutjnl-2017-314461
  • Tye-Din J, Daveson J, Ee H, 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. DOI:10.1111/apt.15477
  • Daveson J, Tye-Din J, Goel G, et al. 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. DOI:10.1111/apt.15551
  • Goel G, Daveson J, Hooi C, et al. Serum cytokines elevated during gluten-mediated cytokine release in coeliac disease. Clin Exp Immunol. 2020;199(1):68–78. DOI:10.1111/cei.13369
  • Yohannes D, de Kauwe A, Kaukinen K, et al. Effects of in vivo gluten challenge on PBMC gene expression profiles in diet treated celiac disease. Front Immunol. 2020;11:594243.
  • Leonard M, Silvester J, Leffler D, et al. Evaluating responses to gluten challenge: a randomized, double-blind, 2-dose gluten challenge trial. Gastroenterology. 2021;160(3):720–733. DOI:10.1053/j.gastro.2020.10.040
  • Stamnaes J, Stray D, Stensland M, et al. In well-treated celiac patients low-level mucosal inflammation predicts response to 14-day gluten challenge. Adv Sci. 2021;8(4):2003526. DOI:10.1002/advs.202003526
  • Pyle G, Paaso B, Anderson B, et al. Low-dose gluten challenge in celiac sprue: malabsorptive and antibody responses. Clin Gastroenterol Hepatol. 2005;3(7):679–686. DOI:10.1016/S1542-3565(05)00365-4
  • Lähdeaho ML, Mäki M, Laurila K, et al. Small-bowel mucosal changes and antibody responses after low- and moderate-dose gluten challenge in celiac disease. BMC Gastroenterol. 2011;11:129.
  • Kelly C, Green PH, Murray JA, et al. Larazotide acetate in patients with coeliac disease undergoing a gluten challenge: a randomised placebo-controlled study. Aliment Pharmacol Ther. 2013;37(2):252–262. DOI:10.1111/apt.12147
  • Lähdeaho ML, Kaukinen K, Laurila K, et al. Glutenase ALV003 attenuates gluten-induced mucosal injury in patients with celiac disease. Gastroenterology. 2014;146(7):1649–1658. DOI:10.1053/j.gastro.2014.02.031
  • Lähdeaho ML, Scheinin M, Vuotikka P, et al. Safety and efficacy of AMG 714 in adults with coeliac disease exposed to gluten challenge: a phase 2a, randomised, double-blind, placebo-controlled study. Lancet Gastroenterol Hepatol. 2019;4(12):948–959. DOI:10.1016/S2468-1253(19)30264-X
  • Taavela J, Viiri K, Popp A, et al. Histological, immunohistochemical and mRNA gene expression responses in coeliac disease patients challenged with gluten using PAXgene fixed paraffin-embedded duodenal biopsies. BMC Gastroenterol. 2019;19(1):189. DOI:10.1186/s12876-019-1089-7
  • Dotsenko V, Oittinen M, Taavela J, et al. Genome-wide transcriptomic analysis of intestinal mucosa in celiac disease patients on a gluten-free diet and postgluten challenge. Cell Mol Gastroenterol Hepatol. 2021;11(1):13–32. DOI:10.1016/j.jcmgh.2020.07.010
  • Schuppan D, Mäki M, Lundin K, et al. A randomized trial of a transglutaminase 2 inhibitor for celiac disease. N Engl J Med. 2021;385(1):35–45. DOI:10.1056/NEJMoa2032441
  • Murray JA, Syage J, Wu T, et al. Latiglutenase protects the mucosa and attenuates symptom severity in patients with celiac disease exposed to a gluten challenge. Gastroenterology. 2022;163(6):1510–1521. DOI:10.1053/j.gastro.2022.07.071
  • Silvester J, Kurada S, Szwajcer A, et al. Tests for serum transglutaminase and endomysial antibodies do not detect most patients with celiac disease and persistent villous atrophy on gluten-free diets: a meta-analysis. Gastroenterology. 2017;153(3):689–701. DOI:10.1053/j.gastro.2017.05.015
  • Rostami-Nejad M, Asri N, Olfatifar M, et al. Systematic review and dose-response meta-analysis on the Relationship between different gluten doses and risk of coeliac disease relapse. Nutrients. 2023;15(6):1390. DOI:10.3390/nu15061390
  • Goel G, Tye-Din J, Qiao SW. Cytokine release and gastrointestinal symptoms after gluten challenge in celiac disease. Sci Adv. 2019;5(8):eaaw7756.
  • Costantino A, Aversano G, Lasagni G, et al. Diagnostic management of patients reporting symptoms after wheat ingestion. Front Nutr. 2022;9:1007007.
  • Mumolo M, Rettura F, Melissari S, et al. Is gluten the only culprit for non-celiac gluten/wheat sensitivity? Nutrients. 2020;12(12):3785. DOI:10.3390/nu12123785
  • van Megen F, Skodje G, Lergenmuller S, et al. A low FODMAP diet reduces symptoms in treated celiac patients with ongoing symptoms–a randomized controlled trial. Clin Gastroenterol Hepatol. 2022;20(10):2258–2266. DOI:10.1016/j.cgh.2022.01.011
  • Ajamian M, Rosella G, Newnham E, et al. Effect of gluten ingestion and FODMAP restriction on intestinal epithelial integrity in patients with irritable bowel syndrome and self-reported non-coeliac gluten sensitivity. Mol Nutr Food Res. 2021;65(5):e1901275. DOI:10.1002/mnfr.201901275
  • Saadati S, Sadeghi A, Mohaghegh-Shalmani H, et al. Effects of a gluten challenge in patients with irritable bowel syndrome: a randomized single-blind controlled clinical trial. Sci Rep. 2022;12(1):4960. DOI:10.1038/s41598-022-09055-6
  • Sainsbury A, Sanders D, Ford A. Prevalence of irritable bowel syndrome-type symptoms in patients with celiac disease: a meta-analysis. Clin Gastroenterol Hepatol. 2013;11(4):359–365.
  • Zanchi C, Ventura A, Martelossi S, et al. Rapid anti-transglutaminase assay and patient interview for monitoring dietary compliance in celiac disease. Scand J Gastroenterol. 2013;48(6):764–766. DOI:10.3109/00365521.2013.786129
  • Mehta P, Pan Z, Riley M, et al. Adherence to a gluten-free diet: assessment by dietician interview and serology. J Pediatr Gastroenterol Nutr. 2018;66(3):e67–e70.
  • Bannister E, Cameron D, Ng J, et al. Can celiac serology alone be used as a marker of duodenal mucosal recovery in children with celiac disease on a gluten-free diet? Am J Gastroenterol. 2014;109(9):1478–1483. DOI:10.1038/ajg.2014.200
  • Adelman D, Murray J, Wu T, et al. Measuring change in small intestinal histology in patients with celiac disease. Am J Gastroenterol. 2018;113(3):339–347. DOI:10.1038/ajg.2017.480
  • Arguelles-Grande C, Tennyson Lewis S, Green PH, et al. Variability in small bowel histopathology reporting between different pathology practice settings: impact on the diagnosis of coeliac disease. J Clin Pathol. 2012;65(3):242–247.
  • Corazza G, Villanacci V, Zambelli C, et al. Comparison of the interobserver reproducibility with different histologic criteria used in celiac disease. Clin Gastroenterol Hepatol. 2007;5(7):838–843. DOI:10.1016/j.cgh.2007.03.019
  • Mubarak A, Nikkels P, Houwen R, et al. Reproducibility of the histological diagnosis of celiac disease. Scand J Gastroenterol. 2011;46(9):1065–1073.
  • Popp A, Arvola T, Taavela J, et al. Nonbiopsy approach for celiac disease is accurate when using exact duodenal histomorphometry: prospective study in 2 countries. J Clin Gastroenterol. 2021;55(3):227–232. DOI:10.1097/MCG.0000000000001349
  • Kurppa K, Taavela J, Saavalainen P, et al. Novel diagnostic techniques for celiac disease. Expert Rev Gastroenterol Hepatol. 2016;10(7):795–805.
  • Virta J, Hannula M, Tamminen I, et al. X-ray microtomography is a novel method for accurate evaluation of small-bowel mucosal morphology and surface area. Sci Rep. 2020;10(1):13164. DOI:10.1038/s41598-020-69487-w
  • Virta J, Hannula M, Lindfors K, et al. Validation of the X-ray microtomography in the assessment of duodenal morphometry and surface area in celiac disease. Front Immunol. 2022;13:945197.
  • Risnes L, Christophersen A, Dahal-Koirala S, et al. Disease-driving CD4+ T cell clonotypes persist for decades in celiac disease. J Clin Invest. 2018;128(6):2642–2650. DOI:10.1172/JCI98819
  • Kurki A, Kemppainen E, Laurikka P, et al. The use of peripheral blood mononuclear cells in celiac disease diagnosis and treatment. Expert Rev Gastroenterol Hepatol. 2021;15(3):305–316.
  • Anderson R, van Heel D, Tye-Din J, et al. T cells in peripheral blood after gluten challenge in coeliac disease. Gut. 2005;54(9):1217–1223. DOI:10.1136/gut.2004.059998
  • Anderson R, Goel G, Hardy M, et al. Whole blood interleukin-2 release test to detect and characterize rare circulating gluten-specific T cell responses in coeliac disease. Clin Exp Immunol. 2021;204(3):321–334. DOI:10.1111/cei.13578
  • Christophersen A, Ráki M, Bergseng E, et al. Tetramer-visualized gluten-specific CD4+ T cells in blood as a potential diagnostic marker for coeliac disease without oral gluten challenge. United Eur Gastroenterol J. 2014;2(4):268–278. DOI:10.1177/2050640614540154
  • Sarna V, Lundin K, Mørkrid L, et al. HLA-DQ-Gluten tetramer blood test accurately identifies patients with and without celiac disease in absence of gluten consumption. Gastroenterology. 2018;154(4):886–896. DOI:10.1053/j.gastro.2017.11.006

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.