Z Gastroenterol 2022; 60(05): 790-856
DOI: 10.1055/a-1741-5946
Leitlinie

Aktualisierte S2k-Leitlinie Zöliakie der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS)

Dezember 2021 – AWMF-Registernummer: 021-021Collaborators
Jörg Felber
1   Medizinische Klinik II – Gastroenterologie, Hepatologie, Endokrinologie, Hämatologie und Onkologie, RoMed Klinikum Rosenheim, Rosenheim, Deutschland
,
Hendrik Bläker
2   Institut für Pathologie, Universitätsklinikum Leipzig AöR, Leipzig, Deutschland
,
Wolfgang Fischbach
3   Innere Medizin und Gastroenterologie Aschaffenburg, Deutschland
,
Sibylle Koletzko
4   Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU-Klinikum München, München, Deutschland
5   Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, 10-719 Olsztyn, Polen
,
Martin Laaß
6   Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Deutschland
,
Nils Lachmann
7   Institut für Transfusionsmedizin, Charité – Universitätsmedizin Berlin, Berlin, Deutschland
,
Pia Lorenz
8   Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
,
Petra Lynen
8   Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
,
Imke Reese
9   Ernährungsberatung und -therapie Allergologie, München, Deutschland
,
Katharina Scherf
10   Institute of Applied Biosciences Department of Bioactive and Functional Food Chemistry, Karlsruhe Institute of Technology (KIT), Karlsruhe, Deutschland
,
Detlef Schuppan
11   Institut für Translationale Immunologie, Johannes Gutenberg-Universität Mainz, Mainz, Deutschland
12   Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
,
Michael Schumann
13   Medizinische Klinik I für Gastroenterologie, Infektiologie und Rheumatologie, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Deutschland
,
D. Aust
,
S. Baas
,
S. Beisel
,
J. de Laffolie
,
E. Duba
,
W. Holtmeier
,
L. Lange
,
C. Loddenkemper
,
G. Moog
,
T. Rath
,
E. Roeb
,
D. Rubin
,
J. Stein
,
H. Török
,
Y. Zopf
› Institutsangaben

Abkürzungsverzeichnis

ASHI: American Society for Histocompatibility and Immunogenetics
ATIs: Amylase-Trypsin-Inhibitoren
BAT: Basophile-Aktivierungstest
BDP: Bundesverband Deutscher Pathologen e. V.
CHOP-Regime: Kombinationstherapie aus den 4 folgenden Medikamenten (C: Cyclophosphamid; H: Hydroxydaunorubicin aka Doxorubicin; Vincristin; P: Prednison, Prednisolon)
CLE: Konfokale Laserendomikroskopie
CT: Computertomografie
DGAKI: Deutsche Gesellschaft für Allergologie und Klinische Immunologie e. V.
DGE: Deutsche Gesellschaft für Ernährung e. V.
DGEM: Deutsche Gesellschaft für Ernährungsmedizin e. V.
DGIM: Deutsche Gesellschaft für Innere Medizin e. V.
DGKJ: Deutsche Gesellschaft für Kinder- und Jugendmedizin e. V.
DGI: Deutsche Gesellschaft für Immungenetik
DGP: Deutsche Gesellschaft für Pathologie e. V.
dGP-IgA: deaminierte Gliadinpeptid-IgA-Antikörper
dGP-IgG: deaminierte Gliadinpeptid-IgG-Antikörper
EATL: Enteropathie-assoziiertes T-Zelllymphom
EFI: European Federation for Immunogenetics
EMA-IgA: Endomysium-IgA-Antikörper
ESPGHAN: European Society for Paediatric Gastroenterology Hepatology and Nutrition
18F-FDG PET-CT: 18F-Fluordesoxyglukose Positronenemissionstomografie-Computertomografie
FICE: Fuji Intelligent Chromo Endoscopy
FocusINCD: Focus in Celiac Disease
FODMAP: Fermentierbare Oligo-, Di-, Monosaccharide und Polyole
FPIES: Food protein-induced enterocolitis syndrome
GFD: glutenfreie Diät
GIP: Immunogene Glutenpeptide
GPA: Gesellschaft für Pädiatrische Allergologie und Umweltmedizin e. V.
GPGE: Gesellschaft für Pädiatrische Gastroenterologie und Ernährung e. V.
HD-WLE: High definition-Weißlichtendoskopie
HLA: Humane Leukozytenantigen-System
HRT: Histaminfreisetzungstest
IBS: Reizdarmsyndrom
IEL: Intraepitheliale Lymphozyten
IPI: Internationaler Prognoseindex
MRT: Magnetresonanztomografie
NBI: Narrow band imaging
NCWS: Nicht-Zöliakie-Weizensensitivität
NET: Neuroendokrine Tumoren
ÖGD: Ösophago-Gastro-Duodenoskopie
PPV: Positiver Vorhersagewert
QUETHEB: Deutsche Gesellschaft der qualifizierten Ernährungstherapeuten und Ernährungsberater e. V.
RCD: Refraktäre Zöliakie
SCT: Stammzelltransplantation
SDS: Standard Deviation Score
STIKO: Ständige Impfkommission
TLR4: Toll-like Rezeptor 4
TSH: Thyroidea-stimulierendes Hormon
tTG-IgA: Gewebstransglutaminase-IgA-Antikörper
tTG-IgG: Gewebstransglutaminase-IgG-Antikörper
tTG: Gewebstransglutaminase, synonym: Transglutaminase-2
UJ: Ulzerative Jejunitis
VAS: Visuelle Analogskala
VCE: Video-Kapselendoskopie
VDD: Verband der Diätassistenten – Deutscher Bundesverband
VFED: Verband für Ernährung und Diätetik e. V.
VdOe: BerufsVerband Oecotrophologie e. V.
WDEIA: Wheat Dependent Exercise Induced Anaphylaxis
WHO: World Health Organization



Publikationsverlauf

Artikel online veröffentlicht:
11. Mai 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • Literatur

  • 1 Academy of Nutrition and Dietetics. International Dietetics and Nutrition Terminology (IDNT) Reference Manual: Standardized Language for the Nutrition Care Process: Academy of Nutrition and Dietetics. 2013
  • 2 Verband der Diätassistenten- Deutscher Bundesverband e. V.. VDD-Leitlinie für die Ernährungstherapie und das prozessgeleitete Handeln in der Diätetik Band 1. Manual für den German-Nutrition Care Process (G-NCP): Pabst Science Publishers. 2015
  • 3 Lammert F, Jansen PL, Lerch MM. Weissbuch Gastroenterologie 2020/2021. Frank L, Petra Lynen J, Markus ML. , editors De Gruyter; 2019
  • 4 Hujoel IA, Reilly NR, Rubio-Tapia A. Celiac Disease: Clinical Features and Diagnosis. Gastroenterology clinics of North America 2019; 48 (01) 19-37
  • 5 Green PH, Krishnareddy S, Lebwohl B. Clinical manifestations of celiac disease. Digestive diseases (Basel, Switzerland) 2015; 33 (02) 137-140
  • 6 Lebwohl B, Rubio-Tapia A. Epidemiology, Presentation, and Diagnosis of Celiac Disease. Gastroenterology 2021; 160 (01) 63-75
  • 7 Smalley W, Falck-Ytter C, Carrasco-Labra A. et al. AGA Clinical Practice Guidelines on the Laboratory Evaluation of Functional Diarrhea and Diarrhea-Predominant Irritable Bowel Syndrome in Adults (IBS-D). Gastroenterology 2019; 157 (03) 851-854
  • 8 Cash BD, Rubenstein JH, Young PE. et al. The prevalence of celiac disease among patients with nonconstipated irritable bowel syndrome is similar to controls. Gastroenterology 2011; 141 (04) 1187-1193
  • 9 Ford AC, Chey WD, Talley NJ. et al. Yield of diagnostic tests for celiac disease in individuals with symptoms suggestive of irritable bowel syndrome: systematic review and meta-analysis. Archives of internal medicine 2009; 169 (07) 651-658
  • 10 Card TR, Siffledeen J, West J. et al. An excess of prior irritable bowel syndrome diagnoses or treatments in Celiac disease: evidence of diagnostic delay. Scandinavian journal of gastroenterology 2013; 48 (07) 801-807
  • 11 Mohseninejad L, Feenstra T, van der Horst HE. et al. Targeted screening for Coeliac Disease among irritable bowel syndrome patients: analysis of cost-effectiveness and value of information. The European journal of health economics: HEPAC: health economics in prevention and care 2013; 14 (06) 947-957
  • 12 Thörn M, Sjöberg D, Ekbom A. et al. Microscopic colitis in Uppsala health region, a population-based prospective study 2005-2009. Scandinavian journal of gastroenterology 2013; 48 (07) 825-830
  • 13 Ukkola A, Mäki M, Kurppa K. et al. Changes in body mass index on a gluten-free diet in coeliac disease: a nationwide study. European journal of internal medicine 2012; 23 (04) 384-388
  • 14 Jericho H, Sansotta N, Guandalini S. Extraintestinal Manifestations of Celiac Disease: Effectiveness of the Gluten-Free Diet. Journal of pediatric gastroenterology and nutrition 2017; 65 (01) 75-79
  • 15 Therrien A, Kelly CP, Silvester JA. Celiac Disease: Extraintestinal Manifestations and Associated Conditions. Journal of clinical gastroenterology 2020; 54 (01) 8-21
  • 16 Choi JM, Lebwohl B, Wang J. et al. Increased prevalence of celiac disease in patients with unexplained infertility in the United States. The Journal of reproductive medicine 2011; 56 (05) 199-203
  • 17 Castaño M, Gómez-Gordo R, Cuevas D. et al. Systematic Review and Meta-Analysis of Prevalence of Coeliac Disease in Women with Infertility. Nutrients 2019; 11 (08) 1950 DOI: 10.3390/nu11081950.
  • 18 Singh P, Arora S, Lal S. et al. Celiac Disease in Women With Infertility: A Meta-Analysis. Journal of clinical gastroenterology 2016; 50 (01) 33-39
  • 19 Tata LJ, Card TR, Logan RF. et al. Fertility and pregnancy-related events in women with celiac disease: a population-based cohort study. Gastroenterology 2005; 128 (04) 849-855
  • 20 Dhalwani NN, West J, Sultan AA. et al Women with celiac disease present with fertility problems no more often than women in the general population. Gastroenterology 2014; 147 (06) 1267-1274 e1 ; quiz e13-4.
  • 21 Abdul Sultan A, Tata LJ, Fleming KM. et al. Pregnancy complications and adverse birth outcomes among women with celiac disease: a population-based study from England. The American journal of gastroenterology 2014; 109 (10) 1653-1661
  • 22 Saccone G, Berghella V, Sarno L. et al. Celiac disease and obstetric complications: a systematic review and metaanalysis. American journal of obstetrics and gynecology 2016; 214 (02) 225-234
  • 23 Tersigni C, Castellani R, de Waure C. et al. Celiac disease and reproductive disorders: meta-analysis of epidemiologic associations and potential pathogenic mechanisms. Human reproduction update 2014; 20 (04) 582-593
  • 24 Ludvigsson JF, Montgomery SM, Ekbom A. Celiac disease and risk of adverse fetal outcome: a population-based cohort study. Gastroenterology 2005; 129 (02) 454-463
  • 25 Zugna D, Richiardi L, Akre O. et al. A nationwide population-based study to determine whether coeliac disease is associated with infertility. Gut 2010; 59 (11) 1471-1475
  • 26 Grode L, Bech BH, Plana-Ripoll O. et al. Reproductive life in women with celiac disease; a nationwide, population-based matched cohort study. Human reproduction (Oxford, England) 2018; 33 (08) 1538-1547
  • 27 Schiepatti A, Sprio E, Sanders DS. et al. Coeliac disease and obstetric and gynaecological disorders: where are we now?. European journal of gastroenterology & hepatology 2019; 31 (04) 425-433
  • 28 Ludvigsson JF, Montgomery SM, Ekbom A. Coeliac disease in the father and risk of adverse pregnancy outcome: a population-based cohort study. Scandinavian journal of gastroenterology 2006; 41 (02) 178-185
  • 29 Zugna D, Richiardi L, Akre O. et al. Celiac disease is not a risk factor for infertility in men. Fertility and sterility 2011; 95 (05) 1709-1713 e1-3
  • 30 Liu E, Wolter-Warmerdam K, Marmolejo J. et al. Routine Screening for Celiac Disease in Children With Down Syndrome Improves Case Finding. Journal of pediatric gastroenterology and nutrition 2020; 71 (02) 252-256
  • 31 Bonamico M, Pasquino AM, Mariani P. et al. Prevalence and clinical picture of celiac disease in Turner syndrome. The Journal of clinical endocrinology and metabolism 2002; 87 (12) 5495-5498
  • 32 Gale L, Wimalaratna H, Brotodiharjo A. et al. Down's syndrome is strongly associated with coeliac disease. Gut 1997; 40 (04) 492-496
  • 33 Denham JM, Hill ID. Celiac disease and autoimmunity: review and controversies. Current allergy and asthma reports 2013; 13 (04) 347-353
  • 34 Bibbò S, Pes GM, Usai-Satta P. et al. Chronic autoimmune disorders are increased in coeliac disease: A case-control study. Medicine 2017; 96 (47) e8562
  • 35 Iqbal T, Zaidi MA, Wells GA. et al. Celiac disease arthropathy and autoimmunity study. Journal of gastroenterology and hepatology 2013; 28 (01) 99-105
  • 36 Pham-Short A, Donaghue KC, Ambler G. et al. Screening for Celiac Disease in Type 1 Diabetes: A Systematic Review. Pediatrics 2015; 136 (01) e170-e176
  • 37 Goodwin G. Type 1 Diabetes Mellitus and Celiac Disease: Distinct Autoimmune Disorders That Share Common Pathogenic Mechanisms. Hormone research in paediatrics 2019; 92 (05) 285-292
  • 38 Casella G, D'Incà R, Oliva L. et al. Prevalence of celiac disease in inflammatory bowel diseases: An IG-IBD multicentre study. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2010; 42 (03) 175-178
  • 39 Pereyra L, Gonzalez R, Mohaidle A. et al. Risk of colorectal neoplasia in patients with celiac disease: a multicenter study. Journal of Crohn's & colitis 2013; 7 (12) e672-e677
  • 40 Zipser RD, Patel S, Yahya KZ. et al. Presentations of adult celiac disease in a nationwide patient support group. Digestive diseases and sciences 2003; 48 (04) 761-764
  • 41 Kabbani TA, Kelly CP, Betensky RA. et al. Patients with celiac disease have a lower prevalence of non-insulin-dependent diabetes mellitus and metabolic syndrome. Gastroenterology 2013; 144 (05) 912-917 e1
  • 42 Ludvigsson JF, Hemminki K, Wahlström J. et al. Celiac disease confers a 1.6-fold increased risk of asthma: a nationwide population-based cohort study. The Journal of allergy and clinical immunology 2011; 127 (04) 1071-1073
  • 43 Yaqoob Z, Al-Kindi SG, Zein J. Association Between Celiac Disease and Asthma. Digestive diseases and sciences 2016; 61 (12) 3636-3637
  • 44 Canova C, Pitter G, Ludvigsson JF. et al. Coeliac disease and asthma association in children: the role of antibiotic consumption. The European respiratory journal 2015; 46 (01) 115-122
  • 45 Rodrigo L, Beteta-Gorriti V, Alvarez N. et al. Cutaneous and Mucosal Manifestations Associated with Celiac Disease. Nutrients 2018; 10 (07) 800 DOI: 10.3390/nu10070800.
  • 46 Potter MDE, Walker MM, Hancock S. et al. A Serological Diagnosis of Coeliac Disease Is Associated with Osteoporosis in Older Australian Adults. Nutrients 2018; 10 (07) 849 DOI: 10.3390/nu10070849.
  • 47 Zanchetta MB, Longobardi V, Bai JC. Bone and Celiac Disease. Current osteoporosis reports 2016; 14 (02) 43-48
  • 48 Dickey W. Low serum vitamin B12 is common in coeliac disease and is not due to autoimmune gastritis. European journal of gastroenterology & hepatology 2002; 14 (04) 425-427
  • 49 Wierdsma NJ, van Bokhorst-de van der Schueren MA, Berkenpas M. et al. Vitamin and mineral deficiencies are highly prevalent in newly diagnosed celiac disease patients. Nutrients 2013; 5 (10) 3975-3992
  • 50 Cooke WT, Smith WT. Neurological disorders associated with adult coeliac disease. Brain: a journal of neurology 1966; 89 (04) 683-722
  • 51 Häuser W, Janke KH, Klump B. et al. Anxiety and depression in adult patients with celiac disease on a gluten-free diet. World journal of gastroenterology 2010; 16 (22) 2780-2787
  • 52 Smith DF, Gerdes LU. Meta-analysis on anxiety and depression in adult celiac disease. Acta psychiatrica Scandinavica 2012; 125 (03) 189-193
  • 53 Lebwohl B, Haggård L, Emilsson L. et al. Psychiatric Disorders in Patients With a Diagnosis of Celiac Disease During Childhood From 1973 to 2016. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2020; DOI: 10.1016/j.cgh.2020.08.018.
  • 54 Quan J, Panaccione N, Jeong J. et al. Association Between Celiac Disease and Autism Spectrum Disorder: A Systematic Review. Journal of pediatric gastroenterology and nutrition 2021; 72 (05) 704-711
  • 55 Dimitrova AK, Ungaro RC, Lebwohl B. et al. Prevalence of migraine in patients with celiac disease and inflammatory bowel disease. Headache 2013; 53 (02) 344-355
  • 56 Zis P, Julian T, Hadjivassiliou M. Headache Associated with Coeliac Disease: A Systematic Review and Meta-Analysis. Nutrients 2018; 10 (10) 1445 DOI: 10.3390/nu10101445.
  • 57 Ludvigsson JF, Zingone F, Tomson T. et al. Increased risk of epilepsy in biopsy-verified celiac disease: a population-based cohort study. Neurology 2012; 78 (18) 1401-1407
  • 58 Mearns ES, Taylor A, Thomas Craig KJ. et al. Neurological Manifestations of Neuropathy and Ataxia in Celiac Disease: A Systematic Review. Nutrients 2019; 11 (02) 380 DOI: 10.3390/nu11020380.
  • 59 Hadjivassiliou M, Croall ID, Zis P. et al. Neurologic Deficits in Patients With Newly Diagnosed Celiac Disease Are Frequent and Linked With Autoimmunity to Transglutaminase 6. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2019; 17 (13) 2678-2686 e2
  • 60 Mones RL, Atienza KV, Youssef NN. et al. Celiac crisis in the modern era. Journal of pediatric gastroenterology and nutrition 2007; 45 (04) 480-483
  • 61 Jamma S, Rubio-Tapia A, Kelly CP. et al. Celiac crisis is a rare but serious complication of celiac disease in adults. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2010; 8 (07) 587-590
  • 62 Forrest EA, Wong M, Nama S. et al. Celiac crisis, a rare and profound presentation of celiac disease: a case report. BMC gastroenterology 2018; 18 (01) 59
  • 63 Catassi C. Celiac crisis/refeeding syndrome combination: new mechanism for an old complication. Journal of pediatric gastroenterology and nutrition 2012; 54 (04) 442-443
  • 64 Rubio-Tapia A, Murray JA. The Liver and Celiac Disease. Clinics in liver disease 2019; 23 (02) 167-176
  • 65 Kaukinen K, Halme L, Collin P. et al. Celiac disease in patients with severe liver disease: gluten-free diet may reverse hepatic failure. Gastroenterology 2002; 122 (04) 881-888
  • 66 Ludvigsson JF, Elfström P, Broomé U. et al. Celiac disease and risk of liver disease: a general population-based study. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2007; 5 (01) 63-69 e1
  • 67 Castillo NE, Vanga RR, Theethira TG. et al. Prevalence of abnormal liver function tests in celiac disease and the effect of a gluten-free diet in the US population. The American journal of gastroenterology 2015; 110 (08) 1216-1222
  • 68 McGowan KE, Lyon ME, Butzner JD. Celiac disease and IgA deficiency: complications of serological testing approaches encountered in the clinic. Clinical chemistry 2008; 54 (07) 1203-1209
  • 69 Lougaris V, Sorlini A, Monfredini C. et al. Clinical and Laboratory Features of 184 Italian Pediatric Patients Affected with Selective IgA Deficiency (SIgAD): a Longitudinal Single-Center Study. Journal of clinical immunology 2019; 39 (05) 470-475
  • 70 Swain S, Selmi C, Gershwin ME. et al. The clinical implications of selective IgA deficiency. Journal of translational autoimmunity 2019; 2: 100025
  • 71 Catassi C, Fabiani E, Corrao G. et al. Risk of non-Hodgkin lymphoma in celiac disease. Jama 2002; 287 (11) 1413-1419
  • 72 Askling J, Linet M, Gridley G. et al. Cancer incidence in a population-based cohort of individuals hospitalized with celiac disease or dermatitis herpetiformis. Gastroenterology 2002; 123 (05) 1428-1435
  • 73 Smedby KE, Akerman M, Hildebrand H. et al. Malignant lymphomas in coeliac disease: evidence of increased risks for lymphoma types other than enteropathy-type T cell lymphoma. Gut 2005; 54 (01) 54-59
  • 74 Elfström P, Granath F, Ekström Smedby K. et al. Risk of lymphoproliferative malignancy in relation to small intestinal histopathology among patients with celiac disease. Journal of the National Cancer Institute 2011; 103 (05) 436-444
  • 75 Green PH, Fleischauer AT, Bhagat G. et al. Risk of malignancy in patients with celiac disease. The American journal of medicine 2003; 115 (03) 191-195
  • 76 Grainge MJ, West J, Solaymani-Dodaran M. et al. The long-term risk of malignancy following a diagnosis of coeliac disease or dermatitis herpetiformis: a cohort study. Alimentary pharmacology & therapeutics 2012; 35 (06) 730-739
  • 77 van Gils T, Nijeboer P, Overbeek LI. et al. Risks for lymphoma and gastrointestinal carcinoma in patients with newly diagnosed adult-onset celiac disease: Consequences for follow-up: Celiac disease, lymphoma and GI carcinoma. United European gastroenterology journal 2018; 6 (10) 1485-1495
  • 78 Lebwohl B, Granath F, Ekbom A. et al. Mucosal healing and risk for lymphoproliferative malignancy in celiac disease: a population-based cohort study. Annals of internal medicine 2013; 159 (03) 169-175
  • 79 Catassi C, Kryszak D, Louis-Jacques O. et al. Detection of Celiac disease in primary care: a multicenter case-finding study in North America. The American journal of gastroenterology 2007; 102 (07) 1454-1460
  • 80 Hujoel IA, Van Dyke CT, Brantner T. et al. Natural history and clinical detection of undiagnosed coeliac disease in a North American community. Alimentary pharmacology & therapeutics 2018; 47 (10) 1358-1366
  • 81 Ludvigsson JF, Leffler DA, Bai JC. et al. The Oslo definitions for coeliac disease and related terms. Gut 2013; 62 (01) 43-52
  • 82 Bajor J, Szakács Z, Farkas N. et al. Classical celiac disease is more frequent with a double dose of HLA-DQB1*02: A systematic review with meta-analysis. PloS one 2019; 14 (02) e0212329
  • 83 Kurppa K, Paavola A, Collin P. et al. Benefits of a gluten-free diet for asymptomatic patients with serologic markers of celiac disease. Gastroenterology 2014; 147 (03) 610-617 e1
  • 84 Rosén A, Ivarsson A, Nordyke K. et al. Balancing health benefits and social sacrifices: a qualitative study of how screening-detected celiac disease impacts adolescents' quality of life. BMC pediatrics 2011; 11: 32
  • 85 Nordyke K, Norström F, Lindholm L. et al. Health-related quality-of-life in children with coeliac disease, measured prior to receiving their diagnosis through screening. Journal of medical screening 2011; 18 (04) 187-192
  • 86 Paavola A, Kurppa K, Ukkola A. et al. Gastrointestinal symptoms and quality of life in screen-detected celiac disease. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2012; 44 (10) 814-818
  • 87 Aziz I, Sanders DS. Are we diagnosing too many people with coeliac disease?. The Proceedings of the Nutrition Society 2012; 71 (04) 538-544
  • 88 Vilppula A, Kaukinen K, Luostarinen L. et al. Clinical benefit of gluten-free diet in screen-detected older celiac disease patients. BMC gastroenterology 2011; 11: 136
  • 89 Auricchio R, Tosco A, Piccolo E. et al. Potenzial celiac children: 9-year follow-up on a gluten-containing diet. The American journal of gastroenterology 2014; 109 (06) 913-921
  • 90 Auricchio R, Mandile R, Del Vecchio MR. et al. Progression of Celiac Disease in Children With Antibodies Against Tissue Transglutaminase and Normal Duodenal Architecture. Gastroenterology 2019; 157 (02) 413-420.e3
  • 91 Malamut G, Cellier C. Refractory Celiac Disease. Gastroenterology clinics of North America 2019; 48 (01) 137-144
  • 92 Malamut G, Cording S, Cerf-Bensussan N. Recent advances in celiac disease and refractory celiac disease. F1000Research 2019; 8 DOI: 10.12688/f1000research.18701.1.
  • 93 Antiga E, Maglie R, Quintarelli L. et al. Dermatitis Herpetiformis: Novel Perspectives. Frontiers in immunology 2019; 10: 1290
  • 94 Sankari H, Hietikko M, Kurppa K. et al. Intestinal TG3- and TG2-Specific Plasma Cell Responses in Dermatitis Herpetiformis Patients Undergoing a Gluten Challenge. Nutrients 2020; 12 (02) 467 DOI: 10.3390/nu12020467.
  • 95 Sárdy M, Kárpáti S, Merkl B. et al. Epidermal transglutaminase (TGase 3) is the autoantigen of dermatitis herpetiformis. The Journal of experimental medicine 2002; 195 (06) 747-757
  • 96 Rose C, Armbruster FP, Ruppert J. et al. Autoantibodies against epidermal transglutaminase are a sensitive diagnostic marker in patients with dermatitis herpetiformis on a normal or gluten-free diet. Journal of the American Academy of Dermatology 2009; 61 (01) 39-43
  • 97 Rose C, Bröcker EB, Zillikens D. Clinical, histological and immunpathological findings in 32 patients with dermatitis herpetiformis Duhring. Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology: JDDG 2010; 8 (04) 265-270 , 271
  • 98 Görög A, Antiga E, Caproni M. et al. S2k guidelines (consensus statement) for diagnosis and therapy of dermatitis herpetiformis initiated by the European Academy of Dermatology and Venereology (EADV). Journal of the European Academy of Dermatology and Venereology: JEADV 2021; 35 (06) 1251-1277
  • 99 Ludvigsson JF, Lindelöf B, Zingone F. et al. Psoriasis in a nationwide cohort study of patients with celiac disease. The Journal of investigative dermatology 2011; 131 (10) 2010-2016
  • 100 Ungprasert P, Wijarnpreecha K, Kittanamongkolchai W. Psoriasis and Risk of Celiac Disease: A Systematic Review and Meta-analysis. Indian journal of dermatology 2017; 62 (01) 41-46
  • 101 Acharya P, Mathur M. Association between psoriasis and celiac disease: A systematic review and meta-analysis. Journal of the American Academy of Dermatology 2020; 82 (06) 1376-1385
  • 102 Husby S, Koletzko S, Korponay-Szabó I. et al. European Society Paediatric Gastroenterology, Hepatology and Nutrition Guidelines for Diagnosing Coeliac Disease 2020. Journal of pediatric gastroenterology and nutrition 2020; 70 (01) 141-156
  • 103 Bruins MJ. The clinical response to gluten challenge: a review of the literature. Nutrients 2013; 5 (11) 4614-4641
  • 104 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 (07) 996-1004
  • 105 Lähdeaho ML, Kaukinen K, Laurila K. et al. Glutenase ALV003 attenuates gluten-induced mucosal injury in patients with celiac disease. Gastroenterology 2014; 146 (07) 1649-1658
  • 106 Andrén Aronsson C, Lee HS, Hård Af Segerstad EM. et al. Association of Gluten Intake During the First 5 Years of Life With Incidence of Celiac Disease Autoimmunity and Celiac Disease Among Children at Increased Risk. Jama 2019; 322 (06) 514-523
  • 107 Vriezinga SL, Auricchio R, Bravi E. et al. Randomized feeding intervention in infants at high risk for celiac disease. The New England journal of medicine 2014; 371 (14) 1304-1315
  • 108 Agardh D, Lee HS, Kurppa K. et al. Clinical features of celiac disease: a prospective birth cohort. Pediatrics 2015; 135 (04) 627-634
  • 109 Smith LB, Lynch KF, Kurppa K. et al. Psychological Manifestations of Celiac Disease Autoimmunity in Young Children. Pediatrics 2017; 139 (03) DOI: 10.1542/peds.2016-2848.
  • 110 Jansen MA, Kiefte-de Jong JC, Gaillard R. et al. Growth trajectories and bone mineral density in anti-tissue transglutaminase antibody-positive children: the Generation R Study. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2015; 13 (05) 913-920 e5
  • 111 Bjorck S, Lynch K, Brundin C. et al. Repeated Screening Can Be Restricted to At-Genetic-Risk Birth Cohorts. Journal of pediatric gastroenterology and nutrition 2016; 62 (02) 271-275
  • 112 Uenishi RH, Gandolfi L, Almeida LM. et al. Screening for celiac disease in 1st degree relatives: a 10-year follow-up study. BMC gastroenterology 2014; 14: 36
  • 113 Khan MR, Silvester JA, Sparks B. et al. The Utility of IgA-Based Serologic Markers in Diagnosing Celiac Disease in Children 24 Months of Age or Younger. J Pediatr 2020; 224: 158-161 e2
  • 114 Giersiepen K, Lelgemann M, Stuhldreher N. et al. Accuracy of diagnostic antibody tests for coeliac disease in children: summary of an evidence report. Journal of pediatric gastroenterology and nutrition 2012; 54 (02) 229-241
  • 115 Candon S, Mauvais FX, Garnier-Lengline H. et al. Monitoring of anti-transglutaminase autoantibodies in pediatric celiac disease using a sensitive radiobinding assay. Journal of pediatric gastroenterology and nutrition 2012; 54 (03) 392-396
  • 116 Werkstetter KJ, Korponay-Szabo IR, Popp A. et al. Accuracy in Diagnosis of Celiac Disease Without Biopsies in Clinical Practice. Gastroenterology 2017; 153 (04) 924-935
  • 117 Nellikkal SS, Hafed Y, Larson JJ. et al. High Prevalence of Celiac Disease Among Screened First-Degree Relatives. Mayo Clin Proc 2019; 94 (09) 1807-1813
  • 118 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 European gastroenterology journal 2019; 7 (05) 583-613
  • 119 Wolf J, Petroff D, Richter T. et al. Validation of Antibody-Based Strategies for Diagnosis of Pediatric Celiac Disease Without Biopsy. Gastroenterology 2017; 153 (02) 410-419 e17
  • 120 Horwitz A, Skaaby T, Karhus LL. et al. Screening for celiac disease in Danish adults. Scandinavian journal of gastroenterology 2015; 50 (07) 824-831
  • 121 Absah I, Rishi AR, Gebrail R. et al. Lack of Utility of Anti-tTG IgG to Diagnose Celiac Disease When Anti-tTG IgA Is Negative. Journal of pediatric gastroenterology and nutrition 2017; 64 (05) 726-729
  • 122 Ludvigsson JF, Neovius M, Hammarstrom L. Association between IgA deficiency & other autoimmune conditions: a population-based matched cohort study. Journal of clinical immunology 2014; 34 (04) 444-451
  • 123 Husby S, Koletzko S, Korponay-Szabó IR. et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of coeliac disease. Journal of pediatric gastroenterology and nutrition 2012; 54 (01) 136-160
  • 124 Falth-Magnusson K, Magnusson KE. Elevated levels of serum antibodies to the lectin wheat germ agglutinin in celiac children lend support to the gluten-lectin theory of celiac disease. Pediatr Allergy Immunol 1995; 6 (02) 98-102
  • 125 Kappler M, Krauss-Etschmann S, Diehl V. et al. Detection of secretory IgA antibodies against gliadin and human tissue transglutaminase in stool to screen for coeliac disease in children: validation study. BMJ 2006; 332: 213-214
  • 126 Leffler DA, Schuppan D. Update on serologic testing in celiac disease. The American journal of gastroenterology 2010; 105 (12) 2520-2524
  • 127 Felber J, Aust D, Baas S. et al. [Results of a S2k-Consensus Conference of the German Society of Gastroenterolgy, Digestive- and Metabolic Diseases (DGVS) in conjunction with the German Coeliac Society (DZG) regarding coeliac disease, wheat allergy and wheat sensitivity]. Zeitschrift fur Gastroenterologie 2014; 52 (07) 711-743
  • 128 Volta U, Caio G, Giancola F. et al. Features and Progression of Potenzial Celiac Disease in Adults. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2016; 14 (05) 686-693 e1
  • 129 Volta U, Caio G, Boschetti E. et al. Seronegative celiac disease: Shedding light on an obscure clinical entity. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2016; 48 (09) 1018-1022
  • 130 Ylonen 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 (09) DOI: 10.3390/nu12092736.
  • 131 Gidrewicz D, Potter K, Trevenen CL. et al. Evaluation of the ESPGHAN Celiac Guidelines in a North American Pediatric Population. The American journal of gastroenterology 2015; 110 (05) 760-767
  • 132 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. Journal of clinical gastroenterology 2021; 55 (03) 227-232
  • 133 Penny HA, Raju SA, Lau MS. et al. Accuracy of a no-biopsy approach for the diagnosis of coeliac disease across different adult cohorts. Gut 2021; 70 (05) 876-883
  • 134 Simell S, Hoppu S, Hekkala A. et al. Fate of five celiac disease-associated antibodies during normal diet in genetically at-risk children observed from birth in a natural history study. The American journal of gastroenterology 2007; 102 (09) 2026-2035
  • 135 Lionetti E, Castellaneta S, Pulvirenti A. et al. Prevalence and natural history of potential celiac disease in at-family-risk infants prospectively investigated from birth. J Pediatr 2012; 161 (05) 908-914
  • 136 Leonard MM, Silvester JA, Leffler D. et al. Evaluating Responses to Gluten Challenge: A Randomized, Double-Blind, 2-Dose Gluten Challenge Trial. Gastroenterology 2021; 160 (03) 720-733 e8
  • 137 Hunt KA, Zhernakova A, Turner G. et al. Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet 2008; 40 (04) 395-402
  • 138 Poddighe D, Rebuffi C, De Silvestri A. et al. Carrier frequency of HLA-DQB1*02 allele in patients affected with celiac disease: A systematic review assessing the potential rationale of a targeted allelic genotyping as a first-line screening. World journal of gastroenterology 2020; 26 (12) 1365-1381
  • 139 Liu E, Lee HS, Aronsson CA. et al. Risk of pediatric celiac disease according to HLA haplotype and country. The New England journal of medicine 2014; 371 (01) 42-49
  • 140 Sciurti M, Fornaroli F, Gaiani F. et al. Genetic susceptibilty and celiac disease: what role do HLA haplotypes play?. Acta Biomed 2018; 89 (09) 17-21
  • 141 Bourgey M, Calcagno G, Tinto N. et al. HLA related genetic risk for coeliac disease. Gut 2007; 56 (08) 1054-1059
  • 142 Karell K, Louka AS, Moodie SJ. et al. 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 (04) 469-477
  • 143 Lebwohl B, Sanders DS, Green PHR. Coeliac disease. Lancet 2018; 391: 70-81
  • 144 Harper JW, Holleran SF, Ramakrishnan R. et al. Anemia in celiac disease is multifactorial in etiology. American journal of hematology 2007; 82 (11) 996-1000
  • 145 Garcia-Manzanares A, Lucendo AJ. Nutritional and dietary aspects of celiac disease. Nutrition in clinical practice: official publication of the American Society for Parenteral and Enteral Nutrition 2011; 26 (02) 163-173
  • 146 Mahadev S, Laszkowska M, Sundstrom J. et al. Prevalence of Celiac Disease in Patients With Iron Deficiency Anemia-A Systematic Review With Meta-analysis. Gastroenterology 2018; 155 (02) 374-382 e1
  • 147 Casella S, Zanini B, Lanzarotto F. et al. Celiac disease in elderly adults: clinical, serological, and histological characteristics and the effect of a gluten-free diet. Journal of the American Geriatrics Society 2012; 60 (06) 1064-1069
  • 148 Garcia-Manzanares A, Tenias JM, Lucendo AJ. Bone mineral density directly correlates with duodenal Marsh stage in newly diagnosed adult celiac patients. Scandinavian journal of gastroenterology 2012; 47 (08) 927-936
  • 149 Cassio A, Ricci G, Baronio F. et al. Long-term clinical significance of thyroid autoimmunity in children with celiac disease. J Pediatr 2010; 156 (02) 292-295
  • 150 Diamanti A, Ferretti F, Guglielmi R. et al. Thyroid autoimmunity in children with coeliac disease: a prospective survey. Archives of disease in childhood 2011; 96 (11) 1038-1041
  • 151 Sadeghi A, Rad N, Ashtari S. et al. The value of a biopsy in celiac disease follow up: assessment of the small bowel after 6 and 24 months treatment with a gluten free diet. Rev Esp Enferm Dig 2020; 112 (02) 101-108
  • 152 Gidrewicz D, Trevenen CL, Lyon M. et al. Normalization Time of Celiac Serology in Children on a Gluten-free Diet. Journal of pediatric gastroenterology and nutrition 2017; 64 (03) 362-367
  • 153 Leonard MM, Weir DC, DeGroote M. et al. Value of IgA tTG in Predicting Mucosal Recovery in Children With Celiac Disease on a Gluten-Free Diet. Journal of pediatric gastroenterology and nutrition 2017; 64 (02) 286-291
  • 154 Koletzko S, Auricchio R, Dolinsek J. et al. No Need for Routine Endoscopy in Children With Celiac Disease on a Gluten-free Diet. Journal of pediatric gastroenterology and nutrition 2017; 65 (03) 267-269
  • 155 Pekki H, Kurppa K, Maki M. et al. Performing routine follow-up biopsy 1 year after diagnosis does not affect long-term outcomes in coeliac disease. Alimentary pharmacology & therapeutics 2017; 45 (11) 1459-1468
  • 156 Petroff D, Wolf J, Richter T. et al. Antibody Concentrations Decrease 14-Fold in Children With Celiac Disease on a Gluten-Free Diet but Remain High at 3 Months. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2018; 16 (09) 1442-1449 e5
  • 157 Silvester JA, 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 (03) 689-701.e1
  • 158 Penny HA, Mooney PD, Burden M. et al. High definition endoscopy with or without I-Scan increases the detection of celiac disease during routine endoscopy. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2016; 48 (06) 644-649
  • 159 Banerjee R, Reddy DN. High-resolution narrow-band imaging can identify patchy atrophy in celiac disease: targeted biopsy can increase diagnostic yield. Gastrointestinal endoscopy 2009; 69 (04) 984-985
  • 160 Kurien M, Evans KE, Hopper AD. et al. Duodenal bulb biopsies for diagnosing adult celiac disease: is there an optimal biopsy site?. Gastrointestinal endoscopy 2012; 75 (06) 1190-1196
  • 161 Cammarota G, Cesaro P, Cazzato A. et al. Optimal band imaging system: a new tool for enhancing the duodenal villous pattern in celiac disease. Gastrointestinal endoscopy 2008; 68 (02) 352-357
  • 162 Cammarota G, Ianiro G, Sparano L. et al. Image-enhanced endoscopy with I-scan technology for the evaluation of duodenal villous patterns. Digestive diseases and sciences 2013; 58 (05) 1287-1292
  • 163 Iacucci M, Poon T, Gui XS. et al. High definition i-SCAN endoscopy with water immersion technique accurately reflects histological severity of celiac disease. Endoscopy international open 2016; 4 (05) E540-E546
  • 164 Tabibian JH, Perrault JF, Murray JA. et al. Narrow band imaging evaluation of duodenal villi in patients with and without celiac disease: A prospective study. World journal of gastrointestinal endoscopy 2019; 11 (02) 145-154
  • 165 Valitutti F, Oliva S, Iorfida D. et al. Narrow band imaging combined with water immersion technique in the diagnosis of celiac disease. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2014; 46 (12) 1099-1102
  • 166 Badreldin R, Barrett P, Wooff DA. et al. How good is zoom endoscopy for assessment of villous atrophy in coeliac disease?. Endoscopy 2005; 37 (10) 994-998
  • 167 De Luca L, Ricciardiello L, Rocchi MB. et al. Narrow band imaging with magnification endoscopy for celiac disease: results from a prospective, single-center study. Diagnostic and therapeutic endoscopy 2013; 2013: 580526
  • 168 Iovino P, Pascariello A, Russo I. et al. Difficult diagnosis of celiac disease: diagnostic accuracy and utility of chromo-zoom endoscopy. Gastrointestinal endoscopy 2013; 77 (02) 233-240
  • 169 Raju SA, White WL, Lau MS. et al. A comparison study between Magniview and high definition white light endoscopy in detecting villous atrophy and coeliac disease: A single centre pilot study. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2018; 50 (09) 920-924
  • 170 Singh R, Mei SL, Jayanna M. et al. Education and imaging: Gastrointestinal: Patchy distribution of coeliac disease diagnosed with narrow band imaging and optical magnification. Journal of gastroenterology and hepatology 2013; 28 (03) 584
  • 171 Stoven SA, Choung RS, Rubio-Tapia A. et al. Analysis of Biopsies From Duodenal Bulbs of All Endoscopy Patients Increases Detection of Abnormalities but has a Minimal Effect on Diagnosis of Celiac Disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2016; 14 (11) 1582-1588
  • 172 Barret M, Malamut G, Rahmi G. et al. Diagnostic yield of capsule endoscopy in refractory celiac disease. The American journal of gastroenterology 2012; 107 (10) 1546-1553
  • 173 Kurien M, Evans KE, Aziz I. et al. Capsule endoscopy in adult celiac disease: a potential role in equivocal cases of celiac disease?. Gastrointestinal endoscopy 2013; 77 (02) 227-232
  • 174 Rokkas T, Niv Y. The role of video capsule endoscopy in the diagnosis of celiac disease: a meta-analysis. European journal of gastroenterology & hepatology 2012; 24 (03) 303-308
  • 175 Delabie J, Holte H, Vose JM. et al. Enteropathy-associated T-cell lymphoma: clinical and histological findings from the international peripheral T-cell lymphoma project. Blood 2011; 118 (01) 148-155
  • 176 Murray JA, Rubio-Tapia A. Diarrhoea due to small bowel diseases. Best practice & research Clinical gastroenterology 2012; 26 (05) 581-600
  • 177 Lidums I, Teo E, Field J. et al. Capsule endoscopy: a valuable tool in the follow-up of people with celiac disease on a gluten-free diet. Clinical and translational gastroenterology 2011; 2 (08) e4
  • 178 Atlas DS, Rubio-Tapia A, Van Dyke CT. et al. Capsule endoscopy in nonresponsive celiac disease. Gastrointestinal endoscopy 2011; 74 (06) 1315-1322
  • 179 Culliford A, Daly J, Diamond B. et al. The value of wireless capsule endoscopy in patients with complicated celiac disease. Gastrointestinal endoscopy 2005; 62 (01) 55-61
  • 180 Daum S, Wahnschaffe U, Glasenapp R. et al. Capsule endoscopy in refractory celiac disease. Endoscopy 2007; 39 (05) 455-458
  • 181 Perez-Cuadrado-Robles E, Lujan-Sanchis M, Elli L. et al. Role of capsule endoscopy in alarm features and non-responsive celiac disease: A European multicenter study. Digestive endoscopy: official journal of the Japan Gastroenterological Endoscopy Society 2018; 30 (04) 461-466
  • 182 Efthymakis K, Milano A, Laterza F. et al. Iron deficiency anemia despite effective gluten-free diet in celiac disease: Diagnostic role of small bowel capsule endoscopy. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2017; 49 (04) 412-416
  • 183 Ress K, Luts K, Rägo T. et al. Nationwide study of childhood celiac disease incidence over a 35-year period in Estonia. European Journal of Pediatrics 2012; 171 (12) 1823-1828
  • 184 Luján-Sanchis M, Pérez-Cuadrado-Robles E, García-Lledó J. et al. Role of capsule endoscopy in suspected celiac disease: A European multi-centre study. World journal of gastroenterology 2017; 23 (04) 703-711
  • 185 Valitutti F, Di Nardo G, Barbato M. et al. Mapping histologic patchiness of celiac disease by push enteroscopy. Gastrointestinal endoscopy 2014; 79 (01) 95-100
  • 186 Hadithi M, Al-toma A, Oudejans J. et al. The value of double-balloon enteroscopy in patients with refractory celiac disease. The American journal of gastroenterology 2007; 102 (05) 987-996
  • 187 Tomba C, Elli L, Bardella MT. et al. Enteroscopy for the early detection of small bowel tumours in at-risk celiac patients. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2014; 46 (05) 400-404
  • 188 Elli L, Casazza G, Locatelli M. et al. Use of enteroscopy for the detection of malignant and premalignant lesions of the small bowel in complicated celiac disease: a meta-analysis. Gastrointestinal endoscopy 2017; 86 (02) 264-273
  • 189 Husby S, Koletzko S, Korponay-Szabo IR. et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of coeliac disease. Journal of pediatric gastroenterology and nutrition 2012; 54 (01) 136-160
  • 190 Hill ID, Dirks MH, Liptak GS. et al. Guideline for the diagnosis and treatment of celiac disease in children: recommendations of the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition. Journal of pediatric gastroenterology and nutrition 2005; 40 (01) 1-19
  • 191 Branski D, Faber J, Freier S. et al. Histologic evaluation of endoscopic versus suction biopsies of small intestinal mucosae in children with and without celiac disease. Journal of pediatric gastroenterology and nutrition 1998; 27 (01) 6-11
  • 192 Oderda G, Forni M, Morra I. et al. Endoscopic and histologic findings in the upper gastrointestinal tract of children with coeliac disease. Journal of pediatric gastroenterology and nutrition 1993; 16 (02) 172-177
  • 193 Abdulkarim AS, Burgart LJ, See J. et al. Etiology of nonresponsive celiac disease: results of a systematic approach. The American journal of gastroenterology 2002; 97 (08) 2016-2021
  • 194 Leffler DA, Dennis M, Hyett B. et al. Etiologies and predictors of diagnosis in nonresponsive celiac disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2007; 5 (04) 445-450
  • 195 Mooney P, Evans K, Singh S. et al. Treatment Failure in Coeliac Disease: A practical guide to investigation and treatment of non-responsive and refractory coeliac disease. Journal of gastrointestinal and liver diseases: JGLD 2012; 21: 197-203
  • 196 Nijeboer P, van Wanrooij RLJ, Tack GJ. et al. Update on the diagnosis and management of refractory coeliac disease. Gastroenterol Res Pract 2013; 2013: 518483
  • 197 Rubio-Tapia A, Murray JA. Liver involvement in celiac disease. Minerva medica 2008; 99 (06) 595-604
  • 198 Rubio-Tapia A, Hill ID, Kelly CP. et al ACG clinical guidelines: diagnosis and management of celiac disease. The American journal of gastroenterology 2013; 108 (05) 656-676 ; quiz 677
  • 199 Rashid M, MacDonald A. Importance of duodenal bulb biopsies in children for diagnosis of celiac disease in clinical practice. BMC gastroenterology 2009; 9: 78
  • 200 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. The American journal of gastroenterology 2010; 105 (09) 2103-2110
  • 201 Bonamico M, Thanasi E, Mariani P. et al. Duodenal bulb biopsies in celiac disease: a multicenter study. Journal of pediatric gastroenterology and nutrition 2008; 47 (05) 618-622
  • 202 Green PH, Cellier C. Celiac disease. The New England journal of medicine 2007; 357 (17) 1731-1743
  • 203 Lebwohl B, Kapel RC, Neugut AI. et al. Adherence to biopsy guidelines increases celiac disease diagnosis. Gastrointestinal endoscopy 2011; 74 (01) 103-109
  • 204 Rostom A, Murray JA, Kagnoff MF. American Gastroenterological Association (AGA) Institute technical review on the diagnosis and management of celiac disease. Gastroenterology 2006; 131 (06) 1981-2002
  • 205 Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. European journal of gastroenterology & hepatology 1999; 11 (10) 1185-1194
  • 206 Villanacci V, Ceppa P, Tavani E. et al. Coeliac disease: the histology report. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2011; 43 (Suppl. 04) S385-S395
  • 207 Dickson BC, Streutker CJ, Chetty R. Coeliac disease: an update for pathologists. Journal of clinical pathology 2006; 59 (10) 1008-1016
  • 208 Pellegrino S, Villanacci V, Sansotta N. et al. Redefining the intraepithelial lymphocytes threshold to diagnose gluten sensitivity in patients with architecturally normal duodenal histology. Alimentary pharmacology & therapeutics 2011; 33 (06) 697-706
  • 209 Walker MM, Murray JA, Ronkainen J. et al. Detection of celiac disease and lymphocytic enteropathy by parallel serology and histopathology in a population-based study. Gastroenterology 2010; 139 (01) 112-119
  • 210 Cooper R, Papworth NJ, Harris C. et al. Counting Intraepithelial Lymphocytes: A Comparison Between Routine Staining and CD3 Immunohistochemistry. International journal of surgical pathology 2020; 28 (04) 367-370
  • 211 Hudacko R, Kathy Zhou X, Yantiss RK. Immunohistochemical stains for CD3 and CD8 do not improve detection of gluten-sensitive enteropathy in duodenal biopsies. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc 2013; 26 (09) 1241-1245
  • 212 Malamut G, Afchain P, Verkarre V. et al. Presentation and long-term follow-up of refractory celiac disease: comparison of type I with type II. Gastroenterology 2009; 136 (01) 81-90
  • 213 Roshan B, Leffler DA, Jamma S. et al. The incidence and clinical spectrum of refractory celiac disease in a north american referral center. Am J Gastroenterol 2011; 106 (05) 923-928
  • 214 Rubio-Tapia A, Kelly DG, Lahr BD. et al Clinical staging and survival in refractory celiac disease: a single center experience. Gastroenterology 2009; 136 (01) 99-107 ; quiz 352-353
  • 215 Ashton-Key M, Diss TC, Pan L. et al. Molecular analysis of T-cell clonality in ulcerative jejunitis and enteropathy-associated T-cell lymphoma. The American journal of pathology 1997; 151 (02) 493-498
  • 216 Cellier C, Delabesse E, Helmer C. et al. Refractory sprue, coeliac disease, and enteropathy-associated T-cell lymphoma. French Coeliac Disease Study Group. Lancet 2000; 356: 203-208
  • 217 Patey-Mariaud De Serre N, Cellier C, Jabri B. et al. Distinction between coeliac disease and refractory sprue: a simple immunohistochemical method. Histopathology 2000; 37 (01) 70-77
  • 218 Rubio-Tapia A, Murray JA. Classification and management of refractory coeliac disease. Gut 2010; 59 (04) 547-557
  • 219 de Mascarel A, Belleannée G, Stanislas S. et al. Mucosal intraepithelial T-lymphocytes in refractory celiac disease: a neoplastic population with a variable CD8 phenotype. The American journal of surgical pathology 2008; 32 (05) 744-751
  • 220 Cheminant M, Bruneau J, Malamut G. et al. NKp46 is a diagnostic biomarker and may be a therapeutic target in gastrointestinal T-cell lymphoproliferative diseases: a CELAC study. Gut 2019; 68 (08) 1396-1405
  • 221 Arguelles-Grande C, Tennyson CA, Lewis SK. et al. Variability in small bowel histopathology reporting between different pathology practice settings: impact on the diagnosis of coeliac disease. Journal of clinical pathology 2012; 65 (03) 242-247
  • 222 Caruso R, Pallone F, Stasi E. et al. Appropriate nutrient supplementation in celiac disease. Annals of Medicine 2013; 45 (08) 522-531
  • 223 Kårhus LL, Skaaby T, Petersen J. et al. Long-term Consequences of Undiagnosed Celiac Seropositivity. The American journal of gastroenterology 2020; DOI: 10.14309/ajg.0000000000000737.
  • 224 Tuire I, Marja-Leena L, Teea S. et al. Persistent Duodenal Intraepithelial Lymphocytosis Despite a Long-Term Strict Gluten-Free Diet in Celiac Disease. American Journal of Gastroenterology 2012; 107 (10) 1563-1569
  • 225 Yachha SK, Srivastava A, Mohindra S. et al. Effect of a gluten-free diet on growth and small-bowel histology in children with celiac disease in India. Journal of gastroenterology and hepatology 2007; 22 (08) 1300-1305
  • 226 Aydogdu S, Midyat L, Cakir M. et al. Long-Term Effect of Gluten-Free Diet on Growth Velocity in Turkish Children with Celiac Disease. Digestive diseases and sciences 2008; 54 (10) 2183-2187
  • 227 Artz E, Warren-Ulanch J, Becker D. et al. Seropositivity to celiac antigens in asymptomatic children with type 1 diabetes mellitus: association with weight, height, and bone mineralization. Pediatric Diabetes 2008; 9 (04) 277-284
  • 228 Clinical utility of serologic testing for celiac disease in asymptomatic patients: an evidence-based analysis. Ontario health technology assessment series 2011; 11 (03) 1-63
  • 229 Bettendorf M, Doerr HG, Hauffa BP. et al. Prevalence of Autoantibodies Associated with Thyroid and Celiac Disease in Ullrich-Turner Syndrome in Relation to Adult Height After Growth Hormone Treatment. Journal of Pediatric Endocrinology and Metabolism 2006; 19 (02) DOI: 10.1515/jpem.2006.19.2.149.
  • 230 Khashan AS, Henriksen TB, Mortensen PB. et al. The impact of maternal celiac disease on birthweight and preterm birth: a Danish population-based cohort study. Human Reproduction 2009; 25 (02) 528-534
  • 231 Kumar A, Meena M, Begum N. et al. Latent celiac disease in reproductive performance of women. Fertility and sterility 2011; 95 (03) 922-927
  • 232 Tata LJ, Card TR, Logan RFA. et al. Fertility and pregnancy-related events in women with celiac disease: A population-based cohort study. Gastroenterology 2005; 128 (04) 849-855
  • 233 Özgör B, Selimoğlu MA. Coeliac disease and reproductive disorders. Scandinavian journal of gastroenterology 2009; 45 (04) 395-402
  • 234 Matysiak-Budnik T, Malamut G, de Serre NPM. et al. Long-term follow-up of 61 coeliac patients diagnosed in childhood: evolution toward latency is possible on a normal diet. Gut 2007; 56 (10) 1379-1386
  • 235 Kurppa K, Paavola A, Collin P. et al. Benefits of a Gluten-Free Diet for Asymptomatic Patients With Serologic Markers of Celiac Disease. Gastroenterology 2014; 147 (03) 610-617-e1
  • 236 Laurikka P, Nurminen S, Kivelä L. et al. Extraintestinal Manifestations of Celiac Disease: Early Detection for Better Long-Term Outcomes. Nutrients 2018; 10 (08) 1015
  • 237 Kivelä L, Popp A, Arvola T. et al. Long‐term health and treatment outcomes in adult coeliac disease patients diagnosed by screening in childhood. United European gastroenterology journal 2018; 6 (07) 1022-1031
  • 238 Itzlinger A, Branchi F, Elli L. et al. Gluten-Free Diet in Celiac Disease – Forever and for All?. Nutrients 2018; 10 (11) 1796
  • 239 Catassi C, Fabiani E, Iacono G. et al. A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. The American Journal of Clinical Nutrition 2007; 85 (01) 160-166
  • 240 Akobeng AK, Thomas AG. Systematic review: tolerable amount of gluten for people with coeliac disease. Alimentary pharmacology & therapeutics 2008; 27 (11) 1044-1052
  • 241 Stemmer E. Grundlagen der glutenfreien Ernährung. Ernährung & Medizin 2015; 30 (01) 45-48
  • 242 Kuderer J. Ernährungstherapie bei Zöliakie. Ernährung & Medizin 2017; 32 (01) 43-48
  • 243 A. E.. Zöliakie Grundlagen, Diagnostik, Ernährungstherapie. ErnährungsUmschau. 2011 4. 202-210
  • 244 Schäfer C. Glutenfreie Diät – Welche Konsequenzen sollen bedacht werden?. Allergologie 2018; 41 (06) 244-251
  • 245 Mozer-Glassberg Y, Zevit N, Rosenbach Y. et al. Follow-Up of Children with Celiac Disease – Lost in Translation?. Digestion 2011; 83 (04) 283-287
  • 246 Husby S, Koletzko S, Korponay-Szabó I. et al. European Society Paediatric Gastroenterology, Hepatology and Nutrition Guidelines for Diagnosing Coeliac Disease 2020. Journal of Pediatric Gastroenterology & Nutrition 2020; 70 (01) 141-156
  • 247 Haboubi NY, Taylor S, Jones S. Coeliac disease and oats: a systematic review. Postgraduate Medical Journal 2006; 82 (972) 672-678
  • 248 Pulido OM, Gillespie Z, Zarkadas M. et al. Chapter 6 Introduction of Oats in the Diet of Individuals with Celiac Disease. Advances in Food and Nutrition Research: Elsevier 2009; 235-285 DOI: 10.1016/S1043-4526(09)57006-4.
  • 249 Garsed K, Scott BB. Can oats be taken in a gluten-free diet? A systematic review. Scandinavian journal of gastroenterology 2007; 42 (02) 171-178
  • 250 Verma A, Gatti S, Galeazzi T. et al. Gluten Contamination in Naturally or Labeled Gluten-Free Products Marketed in Italy. Nutrients 2017; 9 (02) 115
  • 251 Wierdsma N, van Bokhorst-de van der Schueren M, Berkenpas M. et al. Vitamin and Mineral Deficiencies Are Highly Prevalent in Newly Diagnosed Celiac Disease Patients. Nutrients 2013; 5 (10) 3975-3992
  • 252 Annibale B, Severi C, Chistolini A. et al. Efficacy of gluten-free diet alone on recovery from iron deficiency anemia in adult celiac patients. The American journal of gastroenterology 2001; 96 (01) 132-137
  • 253 Lucendo AJ, García-Manzanares A. Bone mineral density in adult coeliac disease: An updated review. Revista Española de Enfermedades Digestivas 2013; 105 (03) 154-162
  • 254 Shepherd SJ, Gibson PR. Nutritional inadequacies of the gluten-free diet in both recently-diagnosed and long-term patients with coeliac disease. Journal of Human Nutrition and Dietetics 2012; 26 (04) 349-358
  • 255 Martin J, Geisel T, Maresch C. et al. Inadequate Nutrient Intake in Patients with Celiac Disease: Results from a German Dietary Survey. Digestion 2013; 87 (04) 240-246
  • 256 Wild D, Robins GG, Burley VJ. et al. Evidence of high sugar intake, and low fibre and mineral intake, in the gluten-free diet. Alimentary pharmacology & therapeutics 2010; 32 (04) 573-581
  • 257 Öhlund K, Olsson C, Hernell O. et al. Dietary shortcomings in children on a gluten-free diet. Journal of Human Nutrition and Dietetics 2010; 23 (03) 294-300
  • 258 Weisbrod VM, Silvester JA, Raber C. et al. Preparation of Gluten-Free Foods Alongside Gluten-Containing Food May Not Always Be as Risky for Celiac Patients as Diet Guides Suggest. Gastroenterology 2020; 158 (01) 273-275
  • 259 Pietzak MM. Follow-up of patients with celiac disease: Achieving compliance with treatment. Gastroenterology 2005; 128 (04) S135-S141
  • 260 Hughey JJ, Ray BK, Lee AR. et al. Self-reported dietary adherence, disease-specific symptoms, and quality of life are associated with healthcare provider follow-up in celiac disease. BMC gastroenterology 2017; 17 (01) DOI: 10.1186/s12876-017-0713-7.
  • 261 Pekki H, Kaukinen K, Ilus T. et al. Long-term follow-up in adults with coeliac disease: Predictors and effect on health outcomes. Digestive and Liver Disease 2018; 50 (11) 1189-1194
  • 262 Kurppa K, Lauronen O, Collin P. et al. Factors Associated with Dietary Adherence in Celiac Disease: A Nationwide Study. Digestion 2012; 86 (04) 309-314
  • 263 Zanini B, Lanzarotto F, Mora A. et al. Five year time course of celiac disease serology during gluten free diet: results of a community based “CD-Watch” program. Digestive and Liver Disease 2010; 42 (12) 865-870
  • 264 Murray JA, Watson T, Clearman B. et al. Effect of a gluten-free diet on gastrointestinal symptoms in celiac disease. The American Journal of Clinical Nutrition 2004; 79 (04) 669-673
  • 265 Ukkola A, Mäki M, Kurppa K. et al. Changes in body mass index on a gluten-free diet in coeliac disease: A nationwide study. European Journal of Internal Medicine 2012; 23 (04) 384-388
  • 266 Silvester JA, 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 (03) 689-701.e1
  • 267 Lebwohl B, Granath F, Ekbom A. et al. Mucosal healing and mortality in coeliac disease. Alimentary pharmacology & therapeutics 2012; 37 (03) 332-339
  • 268 Szakács Z, Gede N, Gyöngyi Z. et al. A Call for Research on the Prognostic Role of Follow-Up Histology in Celiac Disease: A Systematic Review. Frontiers in Physiology 2019; 10 DOI: 10.3389/fphys.2019.01408.
  • 269 Moreno ML, Cebolla Á, Muñoz-Suano A. et al. Detection of gluten immunogenic peptides in the urine of patients with coeliac disease reveals transgressions in the gluten-free diet and incomplete mucosal healing. Gut 2017; 66 (02) 250-257
  • 270 Ruiz-Carnicer Á, Garzón-Benavides M, Fombuena B. et al. Negative predictive value of the repeated absence of gluten immunogenic peptides in the urine of treated celiac patients in predicting mucosal healing: new proposals for follow-up in celiac disease. The American Journal of Clinical Nutrition 2020; 112 (05) 1240-1251
  • 271 Coleman SH, Rej A, Penny H. et al. P267 Redefining the diagnosis of type 1 refractory coeliac disease using urine gluten immunogenic peptides. Gut 2021; 70 (Suppl. 01) A179
  • 272 Martín M, Nestares MT, Diaz C. et al. Multifactorial Etiology of Anemia in Celiac Disease and Effect of Gluten-Free Diet: A Comprehensive Review. Nutrients 2019; 11 (11) 2557
  • 273 Stefanelli G, Viscido A, Longo S. et al. Persistent Iron Deficiency Anemia in Patients with Celiac Disease Despite a Gluten-Free Diet. Nutrients 2020; 12 (08) 2176
  • 274 De Falco L, Tortora R, Imperatore N. et al. The role of TMPRSS6 and HFE variants in iron deficiency anemia in celiac disease. American journal of hematology 2017; 93 (03) 383-393
  • 275 Burger JPW, Van der Laan JJH, Jansen TA. et al. Low Yield for Routine Laboratory Checks in Follow-up of Coeliac Disease. Journal of Gastrointestinal and Liver Diseases 2018; 27 (03) 233-239
  • 276 Wessels MMS, van Veen II, Vriezinga SL. et al. Complementary Serologic Investigations in Children with Celiac Disease Is Unnecessary during Follow-Up. The Journal of Pediatrics 2016; 169: 55-60
  • 277 Hallert C, Svensson M, Tholstrup J. et al. Clinical trial: B vitamins improve health in patients with coeliac disease living on a gluten-free diet. Alimentary pharmacology & therapeutics 2009; 29 (08) 811-816
  • 278 Ahlawat R, Weinstein T, Markowitz J. et al. Should We Assess Vitamin D Status in Pediatric Patients With Celiac Disease?. Journal of Pediatric Gastroenterology & Nutrition 2019; 69 (04) 449-454
  • 279 Lionetti E, Galeazzi T, Dominijanni V. et al. Lower Level of Plasma 25-Hydroxyvitamin D in Children at Diagnosis of Celiac Disease Compared with Healthy Subjects: A Case-Control Study. The Journal of Pediatrics 2021; 228: 132-137.e1
  • 280 Sun X, Lu L, Yang R. et al. Increased Incidence of Thyroid Disease in Patients with Celiac Disease: A Systematic Review and Meta-Analysis. PloS one 2016; 11 (12) e0168708
  • 281 Ciccone A, Gabrieli D, Cardinale R. et al. Metabolic Alterations in Celiac Disease Occurring after Following a Gluten-Free Diet. Digestion 2018; 100 (04) 262-268
  • 282 Koskinen I, Virta LJ, Huhtala H. et al. Overall and Cause-Specific Mortality in Adult Celiac Disease and Dermatitis Herpetiformis Diagnosed in the 21st Century. The American journal of gastroenterology 2020; 115 (07) 1117-1124
  • 283 Heikkilä K, Pearce J, Mäki M. et al. Celiac disease and bone fractures: a systematic review and meta-analysis. The Journal of clinical endocrinology and metabolism 2015; 100 (01) 25-34
  • 284 Pfeil A, Lehmann G, Lange U. Update DVO-Leitlinie 2017 „Prophylaxe, Diagnostik und Therapie der Osteoporose bei postmenopausalen Frauen und Männern“. Zeitschrift für Rheumatologie 2018; 77 (09) 759-763
  • 285 Ganji R, Moghbeli M, Sadeghi R. et al. Prevalence of osteoporosis and osteopenia in men and premenopausal women with celiac disease: a systematic review. Nutrition Journal 2019; 18 (01) 9
  • 286 Walker MD, Williams J, Lewis SK. et al. Measurement of Forearm Bone Density by Dual Energy X-Ray Absorptiometry Increases the Prevalence of Osteoporosis in Men With Celiac Disease. Clinical Gastroenterology and Hepatology 2020; 18 (01) 99-106
  • 287 Pritchard L, Wilson S, Griffin J. et al. Prevalence of reduced bone mineral density in adults with coeliac disease – are we missing opportunities for detection in patients below 50 years of age?. Scandinavian journal of gastroenterology 2018; 53 (12) 1433-1436
  • 288 Canova C, Pitter G, Zanier L. et al. Risk of Fractures in Youths with Celiac Disease—A Population-Based Study. The Journal of Pediatrics 2018; 198: 117-120
  • 289 Micic D, Rao VL, Semrad CE. Celiac Disease and Its Role in the Development of Metabolic Bone Disease. Journal of Clinical Densitometry 2020; 23 (02) 190-199
  • 290 Passanisi S, Dipasquale V, Romano C. Vaccinations and Immune Response in Celiac Disease. Vaccines 2020; 8 (02) 278
  • 291 Simons M, Scott-Sheldon LAJ, Risech-Neyman Y. et al. Celiac Disease and Increased Risk of Pneumococcal Infection: A Systematic Review and Meta-Analysis. The American journal of medicine 2018; 131 (01) 83-89
  • 292 Kreutz JM, Adriaanse MPM, van der Ploeg EMC. et al. Narrative Review: Nutrient Deficiencies in Adults and Children with Treated and Untreated Celiac Disease. Nutrients 2020; 12 (02) 500
  • 293 Sue A, Dehlsen K, Ooi CY. Paediatric Patients with Coeliac Disease on a Gluten-Free Diet: Nutritional Adequacy and Macro- and Micronutrient Imbalances. Current Gastroenterology Reports 2018; 20 (01) DOI: 10.1007/s11894-018-0606-0.
  • 294 Zuccotti G, Fabiano V, Dilillo D. et al. Intakes of nutrients in Italian children with celiac disease and the role of commercially available gluten-free products. Journal of Human Nutrition and Dietetics 2012; 26 (05) 436-444
  • 295 Hopman EGD, le Cessie S, von Blomberg BME. et al. Nutritional Management of the Gluten-free Diet in Young People with Celiac Disease in The Netherlands. Journal of Pediatric Gastroenterology & Nutrition 2006; 43 (01) 102-108
  • 296 Anderson JW, Baird P, Davis Jr RH. et al. Health benefits of dietary fiber. Nutrition Reviews 2009; 67 (04) 188-205
  • 297 Tortora R, Capone P, De Stefano G. et al. Metabolic syndrome in patients with coeliac disease on a gluten-free diet. Alimentary pharmacology & therapeutics 2015; 41 (04) 352-359
  • 298 Kabbani TA, Goldberg A, Kelly CP. et al. Body mass index and the risk of obesity in coeliac disease treated with the gluten-free diet. Alimentary pharmacology & therapeutics 2012; 35 (06) 723-729
  • 299 Zeng F, Wei W, Li M. et al. Heavy Metal Contamination in Rice-Producing Soils of Hunan Province, China and Potenzial Health Risks. International Journal of Environmental Research and Public Health 2015; 12 (12) 15584-15593
  • 300 Raehsler SL, Choung RS, Marietta EV. et al. Accumulation of Heavy Metals in People on a Gluten-Free Diet. Clinical Gastroenterology and Hepatology 2018; 16 (02) 244-251
  • 301 Fajardo V, González MP, Martínez M. et al. Updated Food Composition Database for Cereal-Based Gluten Free Products in Spain: Is Reformulation Moving on?. Nutrients 2020; 12 (08) 2369
  • 302 Theethira TG, Dennis M. Celiac Disease and the Gluten-Free Diet: Consequences and Recommendations for Improvement. Digestive Diseases 2015; 33 (02) 175-182
  • 303 Ludvigsson JF, Leffler DA, Bai JC. et al. The Oslo definitions for coeliac disease and related terms. Gut 2012; 62 (01) 43-52
  • 304 Felber J, Aust D, Baas S. et al. Ergebnisse einer S2k-Konsensuskonferenz der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselerkrankungen (DGVS) gemeinsam mit der Deutschen Zöliakie-Gesellschaft (DZG) zur Zöliakie, Weizenallergie und Weizensensitivität. Zeitschrift fur Gastroenterologie 2014; 52 (07) 711-743
  • 305 McCarty TR, O’Brien CR, Gremida A. et al. Efficacy of duodenal bulb biopsy for diagnosis of celiac disease: a systematic review and meta-analysis. Endoscopy international open 2018; 06 (11) E1369-E1378
  • 306 Biagi F. Intraepithelial lymphocytes in the villous tip: do they indicate potential coeliac disease?. Journal of Clinical Pathology 2004; 57 (08) 835-839
  • 307 Auricchio R, Tosco A, Piccolo E. et al. Potenzial Celiac Children: 9-Year Follow-Up on a Gluten-Containing Diet. American Journal of Gastroenterology 2014; 109 (06) 913-921
  • 308 Volta U, Caio G, Giancola F. et al. Features and Progression of Potenzial Celiac Disease in Adults. Clinical Gastroenterology and Hepatology 2016; 14 (05) 686-693
  • 309 Ludvigsson JF, Bai JC, Biagi F. et al. Diagnosis and management of adult coeliac disease: guidelines from the British Society of Gastroenterology. Gut 2014; 63 (08) 1210-1228
  • 310 Caio G, Volta U, Sapone A. et al. Celiac disease: a comprehensive current review. BMC Medicine 2019; 17 (01) DOI: 10.1186/s12916-019-1380-z.
  • 311 Popp A, Mäki M. Gluten-Induced Extra-Intestinal Manifestations in Potenzial Celiac Disease-Celiac Trait. Nutrients 2019; 11 (02) 320
  • 312 Guarino M, Gambuti E, Alfano F. et al. Life-threatening onset of coeliac disease: a case report and literature review. BMJ Open Gastroenterology 2020; 7 (01) e000406
  • 313 Aziz I, Evans KE, Papageorgiou V. et al. Are patients with coeliac disease seeking alternative therapies to a gluten-free diet?. Journal of gastrointestinal and liver diseases: JGLD 2011; 20 (01) 27-31
  • 314 Alhassan E, Yadav A, Kelly CP. et al. Novel Nondietary Therapies for Celiac Disease. Cellular and Molecular Gastroenterology and Hepatology 2019; 8 (03) 335-345
  • 315 Yoosuf S, Makharia GK. Evolving Therapy for Celiac Disease. Frontiers in Pediatrics 2019; 7 DOI: 10.3389/fped.2019.00193.
  • 316 Plugis NM, Khosla C. Therapeutic approaches for celiac disease. Best Practice & Research Clinical Gastroenterology 2015; 29 (03) 503-521
  • 317 Ivarsson A, Persson L, Nyström L. et al. Epidemic of coeliac disease in Swedish children. Acta Paediatrica 2000; 89 (02) 165-171
  • 318 Ivarsson A. The Swedish epidemic of coeliac disease explored using an epidemiological approach—some lessons to be learnt. Best Practice & Research Clinical Gastroenterology 2005; 19 (03) 425-440
  • 319 Akobeng AK. Effect of breast feeding on risk of coeliac disease: a systematic review and meta-analysis of observational studies. Archives of disease in childhood 2005; 91 (01) 39-43
  • 320 Vriezinga SL, Auricchio R, Bravi E. et al. Randomized Feeding Intervention in Infants at High Risk for Celiac Disease. New England Journal of Medicine 2014; 371 (14) 1304-1315
  • 321 Lionetti E, Castellaneta S, Francavilla R. et al. Introduction of Gluten, HLA Status, and the Risk of Celiac Disease in Children. New England Journal of Medicine 2014; 371 (14) 1295-1303
  • 322 Jansen MAE, Tromp IIM, Kiefte-de Jong JC. et al. Infant feeding and anti-tissue transglutaminase antibody concentrations in the Generation R Study. The American Journal of Clinical Nutrition 2014; 100 (04) 1095-1101
  • 323 Silano M, Agostoni C, Sanz Y. et al. Infant feeding and risk of developing celiac disease: a systematic review. BMJ Open 2016; 6 (01) e009163
  • 324 Ivarsson A, Hernell O, Stenlund H. et al. Breast-feeding protects against celiac disease. The American Journal of Clinical Nutrition 2002; 75 (05) 914-921
  • 325 Andrén Aronsson C, Lee H-S, Koletzko S. et al. Effects of Gluten Intake on Risk of Celiac Disease: A Case-Control Study on a Swedish Birth Cohort. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2016; 14 (03) 403-409.e3
  • 326 Andrén Aronsson C, Lee H-S, Hård af Segerstad EM. et al. Association of Gluten Intake During the First 5 Years of Life With Incidence of Celiac Disease Autoimmunity and Celiac Disease Among Children at Increased Risk. Jama 2019; 322 (06) 514
  • 327 Crespo-Escobar P, Mearin ML, Hervás D. et al. The role of gluten consumption at an early age in celiac disease development: a further analysis of the prospective PreventCD cohort study. The American Journal of Clinical Nutrition 2017; 105 (04) 890-896
  • 328 Kemppainen KM, Lynch KF, Liu E. et al. Factors That Increase Risk of Celiac Disease Autoimmunity After a Gastrointestinal Infection in Early Life. Clinical Gastroenterology and Hepatology 2017; 15 (05) 694-702.e5
  • 329 Myléus A, Hernell O, Gothefors L. et al. Early infections are associated with increased risk for celiac disease: an incident case-referent study. BMC pediatrics 2012; 12 (01) DOI: 10.1186/1471-2431-12-194.
  • 330 Canova C, Zabeo V, Pitter G. et al. Association of Maternal Education, Early Infections, and Antibiotic Use With Celiac Disease: A Population-Based Birth Cohort Study in Northeastern Italy. American Journal of Epidemiology 2014; 180 (01) 76-85
  • 331 Dydensborg Sander S, Hansen AV, Størdal K. et al. Mode of delivery is not associated with celiac disease. Clinical Epidemiology 2018; 10: 323-332
  • 332 Pozo-Rubio T, Capilla A, Mujico JR. et al. Influence of breastfeeding versus formula feeding on lymphocyte subsets in infants at risk of coeliac disease: the PROFICEL study. European Journal of Nutrition 2012; 52 (02) 637-646
  • 333 van Gils T, Nijeboer P, van Wanrooij RL. et al. Mechanisms and management of refractory coeliac disease. Nat Rev Gastroenterol Hepatol 2015; 12 (10) 572-579
  • 334 Al-Toma A, Verbeek WH, Hadithi M. et al. Survival in refractory coeliac disease and enteropathy-associated T-cell lymphoma: retrospective evaluation of single-centre experience. Gut 2007; 56 (10) 1373-1378
  • 335 Cellier C, Patey N, Mauvieux L. et al. Abnormal intestinal intraepithelial lymphocytes in refractory sprue. Gastroenterology 1998; 114 (03) 471-481
  • 336 Ettersperger J, Montcuquet N, Malamut G. et al. Interleukin-15-Dependent T-Cell-like Innate Intraepithelial Lymphocytes Develop in the Intestine and Transform into Lymphomas in Celiac Disease. Immunity 2016; 45 (03) 610-625
  • 337 Cording S, Lhermitte L, Malamut G. et al. Oncogenetic landscape of lymphomagenesis in coeliac disease. Gut 2021; DOI: 10.1136/gutjnl-2020-322935.
  • 338 Holmes GK, Stokes PL, Sorahan TM. et al. Coeliac disease, gluten-free diet, and malignancy. Gut 1976; 17 (08) 612-619
  • 339 Rubio-Tapia A, Malamut G, Verbeek WH. et al. Creation of a model to predict survival in patients with refractory coeliac disease using a multinational registry. Alimentary pharmacology & therapeutics 2016; 44 (07) 704-714
  • 340 Schumann M, Daum S, Preiss JC. et al. [Rare diseases on the trail--the registry for refractory sprue]. Zeitschrift fur Gastroenterologie 2015; 53 (02) 136
  • 341 Al-toma A, Verbeek WH, Mulder CJ. The management of complicated celiac disease. Digestive diseases (Basel, Switzerland) 2007; 25 (03) 230-236
  • 342 Daum S, Ipczynski R, Schumann M. et al. High rates of complications and substantial mortality in both types of refractory sprue. European journal of gastroenterology & hepatology 2009; 21 (01) 66-70
  • 343 Malamut G, Meresse B, Cellier C. et al. Refractory celiac disease: from bench to bedside. Semin Immunopathol 2012; 34 (04) 601-613
  • 344 Bardella MT, Trovato C, Quatrini M. et al. Mesenteric lymph node cavitation: a rare hallmark of celiac disease. Scandinavian journal of gastroenterology 1999; 34 (12) 1257-1259
  • 345 Sanson E, Gassler N, Trautwein C. et al. Cavitating mesenteric lymph node syndrome: a rare complication of refractory celiac disease. Zeitschrift fur Gastroenterologie 2010; 48 (09) 1133-1137
  • 346 Schmitz F, Herzig KH, Stuber E. et al. On the pathogenesis and clinical course of mesenteric lymph node cavitation and hyposplenism in coeliac disease. Int J Colorectal Dis 2002; 17 (03) 192-198
  • 347 Daum S, Ullrich R, Heise W. et al. Intestinal non-Hodgkin's lymphoma: a multicenter prospective clinical study from the German Study Group on Intestinal non-Hodgkin's Lymphoma. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 2003; 21 (14) 2740-2746
  • 348 Daum S, Cellier C, Mulder CJ. Refractory coeliac disease. Best practice & research Clinical gastroenterology 2005; 19 (03) 413-424
  • 349 Murray JA, Watson T, Clearman B. et al. Effect of a gluten-free diet on gastrointestinal symptoms in celiac disease. Am J Clin Nutr 2004; 79 (04) 669-673
  • 350 Dipper CR, Maitra S, Thomas R. et al. Anti-tissue transglutaminase antibodies in the follow-up of adult coeliac disease. Alimentary pharmacology & therapeutics 2009; 30 (03) 236-244
  • 351 Kaukinen K, Sulkanen S, Maki M. et al. IgA-class transglutaminase antibodies in evaluating the efficacy of gluten-free diet in coeliac disease. European journal of gastroenterology & hepatology 2002; 14 (03) 311-315
  • 352 Ruiz-Carnicer A, Garzon-Benavides M, Fombuena B. et al. Negative predictive value of the repeated absence of gluten immunogenic peptides in the urine of treated celiac patients in predicting mucosal healing: new proposals for follow-up in celiac disease. Am J Clin Nutr 2020; DOI: 10.1093/ajcn/nqaa188.
  • 353 Moreno ML, Cebolla A, Munoz-Suano A. et al. Detection of gluten immunogenic peptides in the urine of patients with coeliac disease reveals transgressions in the gluten-free diet and incomplete mucosal healing. Gut 2017; 66 (02) 250-257
  • 354 Al-Toma A, Goerres MS, Meijer JW. et al. Human leukocyte antigen-DQ2 homozygosity and the development of refractory celiac disease and enteropathy-associated T-cell lymphoma. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2006; 4 (03) 315-319
  • 355 Vader W, Stepniak D, Kooy Y. et al. The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses. Proc Natl Acad Sci USA 2003; 100 (21) 12390-12395
  • 356 Daum S, Weiss D, Hummel M. et al. Frequency of clonal intraepithelial T lymphocyte proliferations in enteropathy-type intestinal T cell lymphoma, coeliac disease, and refractory sprue. Gut 2001; 49 (06) 804-812
  • 357 O'Shea U, Abuzakouk M, O'Morain C. et al. Investigation of molecular markers in the diagnosis of refractory coeliac disease in a large patient cohort. J Clin Pathol 2008; 61 (11) 1200-1202
  • 358 Vahedi K, Mascart F, Mary JY. et al. Reliability of antitransglutaminase antibodies as predictors of gluten-free diet compliance in adult celiac disease. The American journal of gastroenterology 2003; 98 (05) 1079-1087
  • 359 Ritter J, Zimmermann K, Johrens K. et al. T-cell repertoires in refractory coeliac disease. Gut 2018; 67 (04) 644-653
  • 360 Wahnschaffe U, Ignatius R, Loddenkemper C. et al. Diagnostic value of endoscopy for the diagnosis of giardiasis and other intestinal diseases in patients with persistent diarrhea from tropical or subtropical areas. Scandinavian journal of gastroenterology 2007; 42 (03) 391-396
  • 361 Moos V, Schneider T. [Whipple's disease]. Dtsch Med Wochenschr 2014; 139 (49) 2507-2509
  • 362 Weber M, Schumann M, Felber J. et al. Jejunal ulcerations – a diagnostic challenge in a patient with coeliac disease. Zeitschrift fur Gastroenterologie 2015; 53 (11) 1261-1266
  • 363 Malamut G, Verkarre V, Suarez F. et al. The enteropathy associated with common variable immunodeficiency: the delineated frontiers with celiac disease. The American journal of gastroenterology 2010; 105 (10) 2262-2275
  • 364 Corazza GR, Biagi F, Volta U. et al. Autoimmune enteropathy and villous atrophy in adults. Lancet 1997; 350: 106-109
  • 365 Gentile NM, Murray JA, Pardi DS. Autoimmune enteropathy: a review and update of clinical management. Curr Gastroenterol Rep 2012; 14 (05) 380-385
  • 366 Akram S, Murray JA, Pardi DS. et al Adult autoimmune enteropathy: Mayo Clinic Rochester experience. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2007; 5 (11) 1282-1290 ; quiz 1245
  • 367 Bismar MM, Sinicrope FA. Radiation enteritis. Curr Gastroenterol Rep 2002; 4 (05) 361-365
  • 368 Rubio-Tapia A, Herman ML, Ludvigsson JF. et al. Severe spruelike enteropathy associated with olmesartan. Mayo Clin Proc 2012; 87 (08) 732-738
  • 369 McCarthy AJ, Sheahan K. Classification of eosinophilic disorders of the small and large intestine. Virchows Arch 2018; 472 (01) 15-28
  • 370 Shah VH, Rotterdam H, Kotler DP. et al. All that scallops is not celiac disease. Gastrointestinal endoscopy 2000; 51 (06) 717-720
  • 371 Berthoux E, Fabien N, Chayvialle JA. et al. Adult celiac disease with thrombosis: a case series of seven patients. Role of thrombophilic factors. Rev Med Interne 2011; 32 (10) 600-604
  • 372 Malamut G, Chandesris O, Verkarre V. et al. Enteropathy associated T cell lymphoma in celiac disease: a large retrospective study. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2013; 45 (05) 377-384
  • 373 Wahab PJ, Meijer JW, Mulder CJ. Histologic follow-up of people with celiac disease on a gluten-free diet: slow and incomplete recovery. American journal of clinical pathology 2002; 118 (03) 459-463
  • 374 Verbeek WH, Goerres MS, von Blomberg BM. et al. Flow cytometric determination of aberrant intra-epithelial lymphocytes predicts T-cell lymphoma development more accurately than T-cell clonality analysis in Refractory Celiac Disease. Clinical immunology (Orlando, Fla) 2008; 126 (01) 48-56
  • 375 Verbeek WH, von Blomberg BM, Scholten PE. et al. The presence of small intestinal intraepithelial gamma/delta T-lymphocytes is inversely correlated with lymphoma development in refractory celiac disease. The American journal of gastroenterology 2008; 103 (12) 3152-3158
  • 376 Cellier C, Bouma G, van Gils T. et al. Safety and efficacy of AMG 714 in patients with type 2 refractory coeliac disease: a phase 2a, randomised, double-blind, placebo-controlled, parallel-group study. Lancet Gastroenterol Hepatol 2019; 4 (12) 960-970
  • 377 Daum S, Hummel M, Weiss D. et al. Refractory sprue syndrome with clonal intraepithelial lymphocytes evolving into overt enteropathy-type intestinal T-cell lymphoma. Digestion 2000; 62 (01) 60-65
  • 378 Liu H, Brais R, Lavergne-Slove A. et al. Continual monitoring of intraepithelial lymphocyte immunophenotype and clonality is more important than snapshot analysis in the surveillance of refractory coeliac disease. Gut 2010; 59 (04) 452-460
  • 379 Celli R, Hui P, Triscott H. et al. Clinical Insignficance of Monoclonal T-Cell Populations and Duodenal Intraepithelial T-Cell Phenotypes in Celiac and Nonceliac Patients. Am J Surg Pathol 2019; 43 (02) 151-160
  • 380 Hussein S, Gindin T, Lagana SM. et al. Clonal T cell receptor gene rearrangements in coeliac disease: implications for diagnosing refractory coeliac disease. J Clin Pathol 2018; 71 (09) 825-831
  • 381 Mallant M, Hadithi M, Al-Toma AB. et al. Abdominal computed tomography in refractory coeliac disease and enteropathy associated T-cell lymphoma. World journal of gastroenterology 2007; 13 (11) 1696-1700
  • 382 Van Weyenberg SJ, Meijerink MR, Jacobs MA. et al. MR enteroclysis in refractory celiac disease: proposal and validation of a severity scoring system. Radiology 2011; 259 (01) 151-161
  • 383 Van Weyenberg SJ, Bouman K, Jacobs MA. et al. Comparison of MR enteroclysis with video capsule endoscopy in the investigation of small-intestinal disease. Abdominal imaging 2013; 38 (01) 42-51
  • 384 Van Weyenberg SJ, Smits F, Jacobs MA. et al. Video capsule endoscopy in patients with nonresponsive celiac disease. Journal of clinical gastroenterology 2013; 47 (05) 393-399
  • 385 Olaussen RW, Lovik A, Tollefsen S. et al. Effect of elemental diet on mucosal immunopathology and clinical symptoms in type 1 refractory celiac disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2005; 3 (09) 875-885
  • 386 Brar P, Lee S, Lewis S. et al. Budesonide in the treatment of refractory celiac disease. The American journal of gastroenterology 2007; 102 (10) 2265-2269
  • 387 Daum S, Ipczynski R, Heine B. et al. Therapy with budesonide in patients with refractory sprue. Digestion 2006; 73 (01) 60-68
  • 388 Mukewar SS, Sharma A, Rubio-Tapia A. et al. Open-Capsule Budesonide for Refractory Celiac Disease. The American journal of gastroenterology 2017; 112 (06) 959-967
  • 389 Therrien A, Silvester JA, Leonard MM. et al. Enteric-Release Budesonide May Be Useful in the Management of Non-Responsive Celiac Disease. Digestive diseases and sciences 2020; DOI: 10.1007/s10620-020-06454-5.
  • 390 Wahab PJ, Crusius JB, Meijer JW. et al. Cyclosporin in the treatment of adults with refractory coeliac disease--an open pilot study. Alimentary pharmacology & therapeutics 2000; 14 (06) 767-774
  • 391 Goerres MS, Meijer JW, Wahab PJ. et al. Azathioprine and prednisone combination therapy in refractory coeliac disease. Alimentary pharmacology & therapeutics 2003; 18 (05) 487-494
  • 392 Tack GJ, van Asseldonk DP, van Wanrooij RL. et al. Tioguanine in the treatment of refractory coeliac disease--a single centre experience. Alimentary pharmacology & therapeutics 2012; 36 (03) 274-281
  • 393 Daum S, Sahin E, Jansen A. et al. Adult autoimmune enteropathy treated successfully with tacrolimus. Digestion 2003; 68 (02) 86-90
  • 394 Gillett HR, Arnott ID, McIntyre M. et al. Successful infliximab treatment for steroid-refractory celiac disease: a case report. Gastroenterology 2002; 122 (03) 800-805
  • 395 Turner SM, Moorghen M, Probert CS. Refractory coeliac disease: remission with infliximab and immunomodulators. European journal of gastroenterology & hepatology 2005; 17 (06) 667-669
  • 396 Jamma S, Leffler DA, Dennis M. et al. Small intestinal release mesalamine for the treatment of refractory celiac disease type I. Journal of clinical gastroenterology 2011; 45 (01) 30-33
  • 397 Al-Toma A, Goerres MS, Meijer JW. et al Cladribine therapy in refractory celiac disease with aberrant T cells. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2006; 4 (11) 1322-1327 ; quiz 00
  • 398 Tack GJ, Verbeek WH, Al-Toma A. et al. Evaluation of Cladribine treatment in refractory celiac disease type II. World journal of gastroenterology 2011; 17 (04) 506-513
  • 399 Hadjibabaie M, Iravani M, Taghizadeh M. et al. Evaluation of nutritional status in patients undergoing hematopoietic SCT. Bone marrow transplantation 2008; 42 (07) 469-473
  • 400 Verbeek WH, Mulder CJ, Zweegman S. Alemtuzumab for refractory celiac disease. The New England journal of medicine 2006; 355 (13) 1396-1397 ; author reply 7
  • 401 Vivas S, Ruiz de Morales JM, Ramos F. et al. Alemtuzumab for refractory celiac disease in a patient at risk for enteropathy-associated T-cell lymphoma. The New England journal of medicine 2006; 354 (23) 2514-2515
  • 402 Al-toma A, Visser OJ, van Roessel HM. et al. Autologous hematopoietic stem cell transplantation in refractory celiac disease with aberrant T cells. Blood 2007; 109 (05) 2243-2249
  • 403 Tack GJ, Wondergem MJ, Al-Toma A. et al. Auto-SCT in refractory celiac disease type II patients unresponsive to cladribine therapy. Bone marrow transplantation 2011; 46 (06) 840-846
  • 404 Chander U, Leeman-Neill RJ, Bhagat G. Pathogenesis of Enteropathy-Associated T Cell Lymphoma. Curr Hematol Malig Rep 2018; 13 (04) 308-317
  • 405 Nijeboer P, Malamut G, Mulder CJ. et al. Enteropathy-associated T-cell lymphoma: improving treatment strategies. Digestive diseases (Basel, Switzerland) 2015; 33 (02) 231-235
  • 406 Chott A, Haedicke W, Mosberger I. et al. Most CD56+ intestinal lymphomas are CD8+CD5-T-cell lymphomas of monomorphic small to medium size histology. The American journal of pathology 1998; 153 (05) 1483-1490
  • 407 Chan JK, Chan AC, Cheuk W. et al. Type II enteropathy-associated T-cell lymphoma: a distinct aggressive lymphoma with frequent gammadelta T-cell receptor expression. Am J Surg Pathol 2011; 35 (10) 1557-1569
  • 408 Zing NPC, Fischer T, Zain J. et al. Peripheral T-Cell Lymphomas: Incorporating New Developments in Diagnostics, Prognostication, and Treatment Into Clinical Practice-PART 2: ENKTL, EATL, Indolent T-Cell LDP of the GI Tract, ATLL, and Hepatosplenic T-Cell Lymphoma. Oncology (Williston Park) 2018; 32 (08) e83-e89
  • 409 Müller-Hermelink K, Delabie J, Ko Y. et al. Enteropathy-associated T-cell lymphoma. In: Bosman FT, Carneiro F, Hruban RH. et al. , editor WHO classification of tumours of the digestive system: IARC. 2010: 112-114
  • 410 Silano M, Volta U, Vincenzi AD. et al. Effect of a gluten-free diet on the risk of enteropathy-associated T-cell lymphoma in celiac disease. Digestive diseases and sciences 2008; 53 (04) 972-976
  • 411 Swerdlow SH, Campo E, Pileri SA. et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016; 127 (20) 2375-2390
  • 412 Ondrejka S, Jagadeesh D. Enteropathy-Associated T-Cell Lymphoma. Curr Hematol Malig Rep 2016; 11 (06) 504-513
  • 413 Moffitt AB, Ondrejka SL, McKinney M. et al. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. The Journal of experimental medicine 2017; 214 (05) 1371-1386
  • 414 Herrera AF, Crosby-Thompson A, Friedberg JW. et al. Comparison of referring and final pathology for patients with T-cell lymphoma in the National Comprehensive Cancer Network. Cancer 2014; 120 (13) 1993-1999
  • 415 Foss FM, Horwitz SM, Civallero M. et al. Incidence and outcomes of rare T cell lymphomas from the T Cell Project: hepatosplenic, enteropathy associated and peripheral gamma delta T cell lymphomas. American journal of hematology 2020; 95 (02) 151-155
  • 416 d'Amore F, Relander T, Lauritzsen GF. et al. Up-front autologous stem-cell transplantation in peripheral T-cell lymphoma: NLG-T-01. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 2012; 30 (25) 3093-3099
  • 417 Wilhelm M, Smetak M, Reimer P. et al. First-line therapy of peripheral T-cell lymphoma: extension and long-term follow-up of a study investigating the role of autologous stem cell transplantation. Blood Cancer J 2016; 6 (07) e452
  • 418 Al-Toma A, Verbeek WH, Visser OJ. et al. Disappointing outcome of autologous stem cell transplantation for enteropathy-associated T-cell lymphoma. Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 2007; 39 (07) 634-641
  • 419 Hadithi M, Mallant M, Oudejans J. et al. 18F-FDG PET versus CT for the detection of enteropathy-associated T-cell lymphoma in refractory celiac disease. Journal of nuclear medicine: official publication, Society of Nuclear Medicine 2006; 47 (10) 1622-1627
  • 420 Feeney J, Horwitz S, Gonen M. et al. Characterization of T-cell lymphomas by FDG PET/CT. Am J Roentgenol 2010; 195 (02) 333-340
  • 421 Casulo C, Schoder H, Feeney J. et al. 18F-fluorodeoxyglucose positron emission tomography in the staging and prognosis of T cell lymphoma. Leuk Lymphoma 2013; 54 (10) 2163-2197
  • 422 Ruskone-Fourmestraux A, Dragosics B, Morgner A. et al. Paris staging system for primary gastrointestinal lymphomas. Gut 2003; 52 (06) 912-913
  • 423 Musshoff K, Schmidt-Vollmer H. Proceedings: Prognosis of non-Hodgkin's lymphomas with special emphasis on the staging classification. Z Krebsforsch Klin Onkol Cancer Res Clin Oncol 1975; 83 (04) 323-341
  • 424 Fischbach W. Gastric MALT lymphoma – update on diagnosis and treatment. Best practice & research Clinical gastroenterology 2014; 28 (06) 1069-1077
  • 425 Moskowitz AJ, Lunning MA, Horwitz SM. How I treat the peripheral T-cell lymphomas. Blood 2014; 123 (17) 2636-2644
  • 426 Lunning MA, Horwitz S. Treatment of peripheral T-cell lymphoma: are we data driven or driving the data?. Curr Treat Options Oncol 2013; 14 (02) 212-223
  • 427 Leventaki V, Manning Jr JT, Luthra R. et al. Indolent peripheral T-cell lymphoma involving the gastrointestinal tract. Hum Pathol 2014; 45 (02) 421-426
  • 428 Sharaiha RZ, Lebwohl B, Reimers L. et al. Increasing incidence of enteropathy-associated T-cell lymphoma in the United States, 1973-2008. Cancer 2012; 118 (15) 3786-3792
  • 429 Ansell SM, Armitage J. Non-Hodgkin lymphoma: diagnosis and treatment. Mayo Clin Proc 2005; 80 (08) 1087-1097
  • 430 Gallamini A, Zaja F, Patti C. et al. Alemtuzumab (Campath-1H) and CHOP chemotherapy as first-line treatment of peripheral T-cell lymphoma: results of a GITIL (Gruppo Italiano Terapie Innovative nei Linfomi) prospective multicenter trial. Blood 2007; 110 (07) 2316-2323
  • 431 Jantunen E, Boumendil A, Finel H. et al. Autologous stem cell transplantation for enteropathy-associated T-cell lymphoma: a retrospective study by the EBMT. Blood 2013; 121 (13) 2529-2532
  • 432 Han X, Zhang W, Zhou D. et al. Autologous stem cell transplantation as frontline strategy for peripheral T-cell lymphoma: A single-centre experience. J Int Med Res 2017; 45 (01) 290-302
  • 433 Sieniawski M, Angamuthu N, Boyd K. et al. Evaluation of enteropathy-associated T-cell lymphoma comparing standard therapies with a novel regimen including autologous stem cell transplantation. Blood 2010; 115 (18) 3664-3670
  • 434 Corradini P, Tarella C, Zallio F. et al. Long-term follow-up of patients with peripheral T-cell lymphomas treated up-front with high-dose chemotherapy followed by autologous stem cell transplantation. Leukemia 2006; 20 (09) 1533-1538
  • 435 Rodriguez J, Conde E, Gutierrez A. et al. Frontline autologous stem cell transplantation in high-risk peripheral T-cell lymphoma: a prospective study from The Gel-Tamo Study Group. Eur J Haematol 2007; 79 (01) 32-38
  • 436 Mercadal S, Briones J, Xicoy B. et al. Intensive chemotherapy (high-dose CHOP/ESHAP regimen) followed by autologous stem-cell transplantation in previously untreated patients with peripheral T-cell lymphoma. Ann Oncol 2008; 19 (05) 958-963
  • 437 Beitinjaneh A, Saliba RM, Medeiros LJ. et al. Comparison of survival in patients with T cell lymphoma after autologous and allogeneic stem cell transplantation as a frontline strategy or in relapsed disease. Biol Blood Marrow Transplant 2015; 21 (05) 855-859
  • 438 Corradini P, Dodero A, Zallio F. et al. Graft-versus-lymphoma effect in relapsed peripheral T-cell non-Hodgkin's lymphomas after reduced-intensity conditioning followed by allogeneic transplantation of hematopoietic cells. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 2004; 22 (11) 2172-2176
  • 439 Schitz N, Nickelsen M, Altmann B. Allogeneic or autologous transplantation as first-line therapy for younger patients with peripheral T cell lymphoma – Results of the interim analysis of the AATT trial. Hematol Oncol, Abstract 0332015.
  • 440 Kharfan-Dabaja MA, Kumar A, Ayala E. et al. Clinical Practice Recommendations on Indication and Timing of Hematopoietic Cell Transplantation in Mature T Cell and NK/T Cell Lymphomas: An International Collaborative Effort on Behalf of the Guidelines Committee of the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2017; 23 (11) 1826-1838
  • 441 Rolinck-Werninghaus C, Niggemann B, Grabenhenrich L. et al. Outcome of oral food challenges in children in relation to symptom-eliciting allergen dose and allergen-specific IgE. Allergy 2012; 67 (07) 951-957
  • 442 Worm M, Reese I, Ballmer-Weber B. et al. S2 Leitlinie Management IgE-vermittelter Nahrungsmittelallergien. Allergo Journal International 2015; 24 (07) 256-293
  • 443 Wassmann A, Werfel T. Atopic eczema and food allergy. Chemical immunology and allergy 2015; 101: 181-190
  • 444 Nowak-Wegrzyn A, Chehade M, Groetch ME. et al. International consensus guidelines for the diagnosis and management of food protein-induced enterocolitis syndrome: Executive summary-Workgroup Report of the Adverse Reactions to Foods Committee, American Academy of Allergy, Asthma & Immunology. The Journal of allergy and clinical immunology 2017; 139 (04) 1111-1126.e4
  • 445 Christensen MJ, Eller E, Mortz CG. et al. Exercise Lowers Threshold and Increases Severity, but Wheat-Dependent, Exercise-Induced Anaphylaxis Can Be Elicited at Rest. The journal of allergy and clinical immunology In practice 2018; 6 (02) 514-520
  • 446 Varjonen E, Vainio E, Kalimo K. Life-threatening, recurrent anaphylaxis caused by allergy to gliadin and exercise. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology 1997; 27 (02) 162-166
  • 447 Roberts G, Ollert M, Aalberse R. et al. A new framework for the interpretation of IgE sensitization tests. Allergy 2016; 71 (11) 1540-1551
  • 448 Matricardi PM, Bockelbrink A, Beyer K. et al. Primary versus secondary immunoglobulin E sensitization to soy and wheat in the Multi-Centre Allergy Study cohort. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology 2008; 38 (03) 493-500
  • 449 Sievers S, Rawel HM, Ringel KP. et al. Wheat protein recognition pattern in tolerant and allergic children. Pediatr Allergy Immunol 2016; 27 (02) 147-155
  • 450 Cianferoni A. Wheat allergy: diagnosis and management. Journal of asthma and allergy 2016; 9: 13-25
  • 451 Sievers S, Rohrbach A, Beyer K. Wheat-induced food allergy in childhood: ancient grains seem no way out. Eur J Nutr 2020; 59 (06) 2693-2707
  • 452 Brockow K, Kneissl D, Valentini L. et al. Using a gluten oral food challenge protocol to improve diagnosis of wheat-dependent exercise-induced anaphylaxis. The Journal of allergy and clinical immunology 2015; 135 (04) 977-984.e4
  • 453 Christensen MJ, Eller E, Mortz CG. et al. Wheat-Dependent Cofactor-Augmented Anaphylaxis: A Prospective Study of Exercise, Aspirin, and Alcohol Efficacy as Cofactors. The journal of allergy and clinical immunology In practice 2019; 7 (01) 114-121
  • 454 Catassi C, Bai JC, Bonaz B. et al. Non-Celiac Gluten sensitivity: the new frontier of gluten related disorders. Nutrients 2013; 5 (10) 3839-3853
  • 455 Sapone A, Bai JC, Ciacci C. et al. Spectrum of gluten-related disorders: consensus on new nomenclature and classification. BMC Med 2012; 10: 13
  • 456 Biesiekierski JR, Newnham ED, Irving PM. et al Gluten causes gastrointestinal symptoms in subjects without celiac disease: a double-blind randomized placebo-controlled trial. The American journal of gastroenterology 2011; 106 (03) 508-514 ; quiz 15
  • 457 Uhde M, Ajamian M, Caio G. et al. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut 2016; DOI: 10.1136/gutjnl-2016-311964.
  • 458 Lebwohl B, Leffler DA. Exploring the Strange New World of Non-Celiac Gluten Sensitivity. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2015; 13 (09) 1613-1615
  • 459 Reese I, Schafer C, Kleine-Tebbe J. et al. Non-celiac gluten/wheat sensitivity (NCGS)-a currently undefined disorder without validated diagnostic criteria and of unknown prevalence: Position statement of the task force on food allergy of the German Society of Allergology and Clinical Immunology (DGAKI). Allergo J Int 2018; 27 (05) 147-151
  • 460 Lerner BA, Green PHR, Lebwohl B. Going Against the Grains: Gluten-Free Diets in Patients Without Celiac Disease – Worthwhile or Not?. Digestive diseases and sciences 2019; DOI: 10.1007/s10620-019-05663-x.
  • 461 Barone M, Gemello E, Viggiani MT. et al. Evaluation of Non-Celiac Gluten Sensitivity in Patients with Previous Diagnosis of Irritable Bowel Syndrome: A Randomized Double-Blind Placebo-Controlled Crossover Trial. Nutrients 2020; 12 (03) DOI: 10.3390/nu12030705.
  • 462 Biesiekierski JR, Peters SL, Newnham ED. et al. No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poorly absorbed, short-chain carbohydrates. Gastroenterology 2013; 145 (02) 320-328.e1-3
  • 463 Dieterich W, Schuppan D, Schink M. et al. Influence of low FODMAP and gluten-free diets on disease activity and intestinal microbiota in patients with non-celiac gluten sensitivity. Clinical nutrition (Edinburgh, Scotland) 2018; DOI: 10.1016/j.clnu.2018.03.017.
  • 464 Skodje GI, Sarna VK, Minelle IH. et al. Fructan, Rather Than Gluten, Induces Symptoms in Patients With Self-reported Non-celiac Gluten Sensitivity. Gastroenterology 2017; DOI: 10.1053/j.gastro.2017.10.040.
  • 465 Catassi C, Elli L, Bonaz B. et al. Diagnosis of Non-Celiac Gluten Sensitivity (NCGS): The Salerno Experts' Criteria. Nutrients 2015; 7 (06) 4966-4977
  • 466 Zevallos VF, Raker V, Tenzer S. et al. Nutritional Wheat Amylase-Trypsin Inhibitors Promote Intestinal Inflammation via Activation of Myeloid Cells. Gastroenterology 2017; 152 (05) 1100-1113.e12
  • 467 Junker Y, Zeissig S, Kim SJ. et al. Wheat amylase trypsin inhibitors drive intestinal inflammation via activation of toll-like receptor 4. The Journal of experimental medicine 2012; 209 (13) 2395-2408
  • 468 Dos Santos Guilherme M, Zevallos VF, Pesi A. et al. Dietary Wheat Amylase Trypsin Inhibitors Impact Alzheimer's Disease Pathology in 5xFAD Model Mice. International journal of molecular sciences 2020; 21 (17) DOI: 10.3390/ijms21176288.
  • 469 Zevallos VF, Raker VK, Maxeiner J. et al. Dietary wheat amylase trypsin inhibitors exacerbate murine allergic airway inflammation. Eur J Nutr 2019; 58 (04) 1507-1514
  • 470 Caminero A, McCarville JL, Zevallos VF. et al. Lactobacilli Degrade Wheat Amylase Trypsin Inhibitors to Reduce Intestinal Dysfunction Induced by Immunogenic Wheat Proteins. Gastroenterology 2019; 156 (08) 2266-2280
  • 471 Bellinghausen I, Weigmann B, Zevallos V. et al. Wheat amylase-trypsin inhibitors exacerbate intestinal and airway allergic immune responses in humanized mice. The Journal of allergy and clinical immunology 2019; 143 (01) 201-212.e4
  • 472 Pickert G, Wirtz S, Matzner J. et al. Wheat Consumption Aggravates Colitis in Mice via Amylase Trypsin Inhibitor-mediated Dysbiosis. Gastroenterology 2020; 159 (01) 257-272.e17
  • 473 Ashfaq-Khan M, Aslam M, Qureshi MA. et al. Dietary wheat amylase trypsin inhibitors promote features of murine non-alcoholic fatty liver disease. Scientific reports 2019; 9 (01) 17463
  • 474 Fritscher-Ravens A, Schuppan D, Ellrichmann M. et al. Confocal endomicroscopy shows food-associated changes in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology 2014; 147 (05) 1012-1020.e4
  • 475 Fritscher-Ravens A, Pflaum T, Mösinger M. et al. Many Patients With Irritable Bowel Syndrome Have Atypical Food Allergies Not Associated With Immunoglobulin E. Gastroenterology 2019; 157 (01) 109-118.e5
  • 476 Zanini B, Basche R, Ferraresi A. et al. Randomised clinical study: gluten challenge induces symptom recurrence in only a minority of patients who meet clinical criteria for non-coeliac gluten sensitivity. Alimentary pharmacology & therapeutics 2015; 42 (08) 968-976
  • 477 Dale HF, Hatlebakk JG, Hovdenak N. et al. The effect of a controlled gluten challenge in a group of patients with suspected non-coeliac gluten sensitivity: A randomized, double-blind placebo-controlled challenge. Neurogastroenterology and motility: the official journal of the European Gastrointestinal Motility Society 2018; DOI: 10.1111/nmo.13332.
  • 478 Huaman JW, Mego M, Manichanh C. et al. Effects of Prebiotics vs a Diet Low in FODMAPs in Patients With Functional Gut Disorders. Gastroenterology 2018; 155 (04) 1004-1007
  • 479 Schumann D, Klose P, Lauche R. et al. Low fermentable, oligo-, di-, mono-saccharides and polyol diet in the treatment of irritable bowel syndrome: A systematic review and meta-analysis. Nutrition (Burbank, Los Angeles County, Calif) 2018; 45: 24-31
  • 480 Geisslitz S, Longin CFH, Koehler P. et al. Comparative quantitative LC-MS/MS analysis of 13 amylase/trypsin inhibitors in ancient and modern Triticum species. Scientific reports 2020; 10 (01) 14570
  • 481 Geisslitz S, Ludwig C, Scherf KA. et al. Targeted LC-MS/MS Reveals Similar Contents of α-Amylase/Trypsin-Inhibitors as Putative Triggers of Nonceliac Gluten Sensitivity in All Wheat Species except Einkorn. Journal of agricultural and food chemistry 2018; 66 (46) 12395-12403
  • 482 Carroccio A, Mansueto P, Soresi M. et al. Wheat Consumption Leads to Immune Activation and Symptom Worsening in Patients with Familial Mediterranean Fever: A Pilot Randomized Trial. Nutrients 2020; 12 (04) DOI: 10.3390/nu12041127.
  • 483 Carrera-Bastos P, Fontes-Villalba M, O'Keefe JH. et al. The western diet and lifetyle and diseases of civilization. Research Reports in Clinical Cardiology 2011; 2: 15-35
  • 484 Spisni E, Imbesi V, Giovanardi E. et al. Differential Physiological Responses Elicited by Ancient and Heritage Wheat Cultivars Compared to Modern Ones. Nutrients 2019; 11 (12) DOI: 10.3390/nu11122879.
  • 485 Laatikainen R, Koskenpato J, Hongisto SM. et al. Pilot Study: Comparison of Sourdough Wheat Bread and Yeast-Fermented Wheat Bread in Individuals with Wheat Sensitivity and Irritable Bowel Syndrome. Nutrients 2017; 9 (11) DOI: 10.3390/nu9111215.
  • 486 Schiepatti A, Savioli J, Vernero M. et al. Pitfalls in the Diagnosis of Coeliac Disease and Gluten-Related Disorders. Nutrients 2020; 12 (06) DOI: 10.3390/nu12061711.
  • 487 Marild K, Stordal K, Bulik CM. et al. Celiac Disease and Anorexia Nervosa: A Nationwide Study. Pediatrics 2017; 139 (05) DOI: 10.1542/peds.2016-4367.
  • 488 Andresen V, Enck P, Frieling T. et al. [S2k guideline for chronic constipation: definition, pathophysiology, diagnosis and therapy]. Zeitschrift fur Gastroenterologie 2013; 51 (07) 651-672
  • 489 Muller-Lissner S, Tack J, Feng Y. et al. Levels of satisfaction with current chronic constipation treatment options in Europe – an internet survey. Alimentary pharmacology & therapeutics 2013; 37 (01) 137-145
  • 490 Welstead L. The Gluten-Free Diet in the 3rd Millennium: Rules, Risks and Opportunities. Diseases (Basel, Switzerland) 2015; 3 (03) 136-149
  • 491 Tovoli F, Negrini G, Fari R. et al. Increased risk of nonalcoholic fatty liver disease in patients with coeliac disease on a gluten-free diet: beyond traditional metabolic factors. Alimentary pharmacology & therapeutics 2018; 48 (05) 538-546
  • 492 Reilly NR, Lebwohl B, Hultcrantz R. et al. Increased risk of non-alcoholic fatty liver disease after diagnosis of celiac disease. Journal of hepatology 2015; 62 (06) 1405-1411
  • 493 Vici G, Belli L, Biondi M. et al. Gluten free diet and nutrient deficiencies: A review. Clinical nutrition (Edinburgh, Scotland) 2016; 35 (06) 1236-1241
  • 494 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: j1892
  • 495 Fry L, Madden AM, Fallaize R. An investigation into the nutritional composition and cost of gluten-free versus regular food products in the UK. Journal of human nutrition and dietetics: the official journal of the British Dietetic Association 2017; DOI: 10.1111/jhn.12502.
  • 496 Caio G, Lungaro L, Segata N. et al. Effect of Gluten-Free Diet on Gut Microbiota Composition in Patients with Celiac Disease and Non-Celiac Gluten/Wheat Sensitivity. Nutrients 2020; 12 (06) DOI: 10.3390/nu12061832.
  • 497 Shah S, Akbari M, Vanga R. et al. Patient perception of treatment burden is high in celiac disease compared with other common conditions. The American journal of gastroenterology 2014; 109 (09) 1304-1311
  • 498 Pfeiffer K, Kohlenberg-Müller K. Was kostet eine glutenfreie Ernährung bei Zöliakie?Verzehrserhebungen und Selbsteinschätzungen zum diätetisch bedingten Aufwand. Aktuel Ernahrungsmed 2015; 40 (03) P1-P6
  • 499 Bulka CM, Davis MA, Karagas MR. et al. The Unintended Consequences of a Gluten-free Diet. Epidemiology (Cambridge, Mass) 2017; 28 (03) e24-e25
  • 500 Raehsler SL, Choung RS, Marietta EV. et al. Accumulation of Heavy Metals in People on a Gluten-Free Diet. Clinical Gastroenterology and Hepatology 2018; 16 (02) 244-251