1
|
Campbell DE, Mehr S, Moscatelli OG, Anderson RP, Tye-Din JA. Immune therapies in coeliac disease and food allergies: Advances, challenges, and opportunities. Semin Immunol 2025; 78:101960. [PMID: 40273881 DOI: 10.1016/j.smim.2025.101960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/19/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
Coeliac disease and food allergy management primarily relies on the strict avoidance of dietary antigens. This approach is challenging to maintain in real-world settings and in food allergy carries the risk of life-threatening anaphylaxis. Despite their distinct pathogenesis, both disorders are driven by maladaptive responses to dietary proteins, creating opportunities for shared treatment strategies. In food allergy, desensitisation therapies such as oral, sublingual, and epicutaneous immunotherapy are well-established, complemented by biologics like omalizumab and dupilumab. However, the induction of sustained tolerance remains challenging. In contrast, therapeutic advancements for coeliac disease are still in their early stages. Current efforts focus on gluten detoxification or modification, immune blockade or modulation, tolerogenic approaches, and barrier restoration. Emerging therapies, including JAK and BTK inhibitors and microbiome-targeted interventions, support further targeted treatment options for both conditions. Biomarkers tracking gluten-specific T cells have emerged as valuable tools for immunomonitoring and symptom assessment in coeliac disease, although standardisation of patient-reported outcome measures and gluten challenge protocols is still needed. Food allergy trials are reliant on double-blind placebo-controlled food challenges to measure allergen reactivity, but these are time-consuming, carry risks, and underscore the need for surrogate biomarkers. The successful development of immune-targeted therapies will require building an immune toolset to optimally assess systemic responses to antigens in both conditions. Clinically, this could lead to better outcomes for patients who might otherwise remain undiagnosed or untreated due to the absence of significant enteropathy or allergen-specific symptoms.
Collapse
Affiliation(s)
- Dianne E Campbell
- Children's Hospital at Westmead, Sydney, New South Wales, Australia; University of Sydney, Sydney, New South Wales, Australia; National Allergy Centre of Excellence, Murdoch Children's Research Institute, Parkville, Victora, Australia
| | - Sam Mehr
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Olivia G Moscatelli
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Robert P Anderson
- Gastroenterology Service, Mackay Base Hospital, West Mackay, Queensland, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Victoria, Australia; Department of Gastroenterology, the Royal Melbourne Hospital, Parkville, Victoria, Australia; The Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| |
Collapse
|
2
|
Gómez-Aguililla S, Farrais S, López-Palacios N, Arau B, Senosiain C, Corzo M, Fernandez-Jimenez N, Ruiz-Carnicer Á, Fernández-Bañares F, González-García BP, Tristán E, Montero-Calle A, Garranzo-Asensio M, Casado I, Pujals M, Hernández JM, Infante-Menéndez J, Roy G, Sousa C, Núñez C. Diagnosis of celiac disease on a gluten-free diet: a multicenter prospective quasi-experimental clinical study. BMC Med 2025; 23:182. [PMID: 40140887 PMCID: PMC11948932 DOI: 10.1186/s12916-025-04008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Diagnosing celiac disease (CD) in individuals adhering to a gluten-free diet (GFD) presents significant challenges. Current guidelines recommend a gluten challenge (GC) lasting at least 6-8 weeks, which has several limitations. Our aim was to compare four approaches previously proposed for diagnosing CD on a GFD: IL-2 serum levels, gut-homing CD8+ T cells, % TCRγδ+ intraepithelial lymphocytes (IELs), and UBE2L3 gene expression. Additionally, we evaluated the CD8+ T-cell-based method with a 3-day GC against the standard GC protocol. METHODS We conducted a multicenter prospective quasi-experimental clinical study. Two subsets of individuals were considered: (1) 20 patients with CD previously diagnosed and 15 non-CD controls, to evaluate the first aim; (2) 41 individuals with uncertain diagnosis who were on a GFD and required GC following current clinical guidelines, to assess the second aim. All participants underwent a 3-day GC (10 g gluten/day). RESULTS Among CD patients and non-CD controls, the sensitivity and specificity of IL-2, gut-homing CD8+ T cells, and UBE2L3 were 82.4% and 83.3%, 88.2% and 100%, and 52.9% and 100%, respectively. The percentage of TCRγδ+ IELs showed 88.2% sensitivity. In the uncertain diagnosis group, a CD8+ T-cell positive response was observed in 8 of the 41 subjects. CONCLUSIONS The percentage of TCRγδ+ IELs and the analysis of IL-2 levels and gut-homing CD8+ T cells are promising diagnostic methods for CD on a GFD. Notably, our results suggest that the CD8+ T-cell assay may provide a consistent and reliable alternative to the extended GC, eliminating the need for invasive procedures to obtain duodenal samples and prolonged gluten ingestion. However, further research with larger cohorts are necessary to validate these findings and establish their definitive clinical utility.
Collapse
Affiliation(s)
- Sara Gómez-Aguililla
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain.
| | - Sergio Farrais
- Servicio de Aparato Digestivo, Hospital Universitario Fundación Jiménez Díaz, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, 28040, Spain
| | - Natalia López-Palacios
- Servicio de Aparato Digestivo, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Beatriz Arau
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Carla Senosiain
- Servicio de Aparato Digestivo, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - María Corzo
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Nora Fernandez-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute, University of the Basque Country (UPV/EHU), Leioa, Basque Country, Spain
| | - Ángela Ruiz-Carnicer
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, 41012, Spain
| | - Fernando Fernández-Bañares
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Bárbara P González-García
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute, University of the Basque Country (UPV/EHU), Leioa, Basque Country, Spain
| | - Eva Tristán
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Ana Montero-Calle
- Functional Proteomics Unit, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - María Garranzo-Asensio
- Functional Proteomics Unit, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda, Madrid, 28220, Spain
| | - Isabel Casado
- Servicio de Anatomía Patológica, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Mar Pujals
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Juana María Hernández
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa (Barcelona), Spain
| | - Jorge Infante-Menéndez
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain
| | - Garbiñe Roy
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | - Carolina Sousa
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, 41012, Spain
| | - Concepción Núñez
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, 28040, Spain.
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Madrid, 28029, Spain.
| |
Collapse
|
3
|
Neumann FAO, Müller M, Mattert G, Liebig S, Herbst V, Zapf D, Kiderlen TR, Linke C, Arp F, Deckert PM, Lüth S, Schwarzlose-Schwarck S, Dammermann W, Reinwald M. Pneumocystis Jirovecii Pneumonia: The Potential of KEX1, MSG1, and MSG2 as Key Antigens in Cytokine Release Assays. Diagnostics (Basel) 2025; 15:793. [PMID: 40218143 PMCID: PMC11989143 DOI: 10.3390/diagnostics15070793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives:Pneumocystis jirovecii pneumonia (PJP) is the most frequently diagnosed AIDS-defining illness in Europe, with especially high mortality in HIV-negative patients caused by delayed diagnosis and low awareness. This study aims to evaluate cytokine release assays (CRA) to facilitate a less invasive and resource-efficient PJP specific diagnostic test. We focus on the P. jirovecii antigens Kexin 1 (KEX1), MSG1, and MSG2, which were identified in prior studies as immunologically relevant. Methods: Whole blood samples from 50 participants-22 healthy individuals and 28 immunocompromised individuals, including 8 with proven PJP-were stimulated in vitro with full-length and partial KEX1, MSG1, MSG2, and a combination of all three antigens (PJ-MIX). Following 24 h incubation at 37 °C, cytokine levels of IL-2, IFN-γ, IL-17A, and IL-17F were measured. Results: Stimulation with full-length KEX1, MSG1, MSG2, and PJ-MIX antigens induced higher IL-2 concentrations in the healthy control group compared to the groups IL-2 baseline levels and to the group of proven PJP cases. Similarly, stimulation with full-length KEX1, MSG1, and PJ-MIX elevated IFN-γ levels in the healthy control group compared to baseline IFN-γ levels. Conclusions: Our findings highlight the potential of IL-2 and IFN-γ release following stimulation with PJ antigens, with PJ-MIX eliciting the strongest and most significant responses, suggesting a cumulative antigen effect. This pilot study establishes a foundation for a PJP-specific CRA, deepening our knowledge of T-cell immunity against PJP. Clinically, such a test could, among other applications, evaluate at-risk patients who should receive prophylaxis and may consequently reduce PJP-related morbidity and mortality.
Collapse
Affiliation(s)
- F. A. Ottilie Neumann
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - Markus Müller
- Department of Infectiology, Academic Medical Teaching Hospital, St. Joseph Krankenhaus Berlin Tempelhof, 12101 Berlin, Germany
| | - Gregor Mattert
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - Sven Liebig
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Victor Herbst
- Euroimmun Medizinische Labordiagnostika AG, 23560 Lübeck, Germany
| | - Dorinja Zapf
- Euroimmun Medizinische Labordiagnostika AG, 23560 Lübeck, Germany
| | - Til R. Kiderlen
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
- Department of Hematology and Oncology, Vivantes Auguste-Viktoria-Klinikum, 12157 Berlin, Germany
| | - Christian Linke
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - Franziska Arp
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - P. Markus Deckert
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - Stefan Lüth
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
- Department of Gastroenterology, Center for Translational Medicine, University Hospital Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany
| | - Sandra Schwarzlose-Schwarck
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| | - Werner Dammermann
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
- Department of Gastroenterology, Center for Translational Medicine, University Hospital Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany
| | - Mark Reinwald
- Department of Hematology, Oncology and Palliative Care, and Center for Translational Medicine, University Hospital Brandenburg an der Havel, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (P.M.D.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 14770 Brandenburg an der Havel, Germany (W.D.)
| |
Collapse
|
4
|
Lacorcia M, Bhattacharjee P, Foster A, Hardy MY, Tye-Din JA, Karas JA, Wentworth JM, Cameron FJ, Mannering SI. BASTA, a simple whole-blood assay for measuring β cell antigen-specific CD4 + T cell responses in type 1 diabetes. Sci Transl Med 2025; 17:eadt2124. [PMID: 40106580 DOI: 10.1126/scitranslmed.adt2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease where T cells mediate the destruction of the insulin-producing β cells found within the islets of Langerhans in the pancreas. Autoantibodies to β cell antigens are the only tests available to detect β cell autoimmunity. T cell responses to β cell antigens, which are known to cause T1D, can only be measured in research settings because of the complexity of assays and the large blood volumes required. Here, we describe the β cell antigen-specific T cell assay (BASTA). BASTA is a simple whole-blood assay that can detect human CD4+ T cell responses to β cell antigens by measuring antigen-stimulated interleukin-2 (IL-2) production. BASTA is both more sensitive and specific than the CFSE (carboxyfluorescein diacetate succinimidyl ester)-based proliferation assay. We used BASTA to identify the regions of preproinsulin that stimulated T cell responses specifically in blood from people with T1D. BASTA can be done with as little as 2 to 3 milliliters of blood. We found that effector memory CD4+ T cells are the primary producers of IL-2 in response to preproinsulin peptides. We then evaluated responses to individual and pooled preproinsulin peptides in a cross-sectional study of pediatric patients: without T1D, without T1D but with a first-degree relative with T1D, or diagnosed with T1D. In contrast with other preproinsulin peptides, full-length C-peptide (PI33-63) showed high specificity for T1D [area under the curve (AUC) = 0.86)]. We suggest that BASTA will be a useful tool for monitoring changes in β cell-specific CD4+ T cell responses both in research and clinical settings.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Pushpak Bhattacharjee
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Abby Foster
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Melinda Y Hardy
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - John A Karas
- School of Chemistry, University of Melbourne, Parkville, Victoria 3010, Australia
| | - John M Wentworth
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
- Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
5
|
Kemppainen E, Albó O, Kaunisto H, Siukola E, Lindfors K. Differential immune responses behind different celiac disease manifestations. Semin Immunol 2025; 78:101941. [PMID: 40086411 DOI: 10.1016/j.smim.2025.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
In celiac disease (CeD), dietary gluten serves as the driver for a comparatively well characterized small bowel mucosal immune response that generally results in small bowel mucosal villous atrophy and crypt hyperplasia along with a disease-specific transglutaminase 2 (TG2) targeting autoantibody response. Individuals with positive TG2 autoantibodies but normal small intestinal mucosal morphology are regarded at increased risk of developing CeD and represent patients with potential CeD. The removal of gluten from the diet leads to disappearance of the autoantibodies and normalization of the mucosal architecture in most cases. However, refractory CeD patients deviate from this dogma as they present with abnormal T cell compartment, persistent symptoms and villous atrophy despite a strict gluten-free diet. The heterogeneity of CeD presentation is further diversified by varying symptomatology. Gastrointestinal symptoms are the most canonical signs of CeD, and they include for instance diarrhea, vomiting, constipation and abdominal pain. Yet, a great portion of the patients manifest the disease at extraintestinal sites such as skin, musculoskeletal system or neuronal tissues. Beyond the involvement of various transglutaminase autoantibodies, the detailed immune mechanisms contributing to the development of these manifestations remains elusive, though. This review highlights the current understanding of the immunological differences in various manifestations of CeD. As the immunological basis of the different CeD phenotypes is at present insufficiently understood, more research on the subject is warranted before such data could be maximally applied to clinical practice.
Collapse
Affiliation(s)
- Esko Kemppainen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Olga Albó
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Helka Kaunisto
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emilia Siukola
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
6
|
Kowalski MK, Domżał-Magrowska D, Małecka-Wojciesko E. Celiac Disease-Narrative Review on Progress in Celiac Disease. Foods 2025; 14:959. [PMID: 40231983 PMCID: PMC11941517 DOI: 10.3390/foods14060959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 04/16/2025] Open
Abstract
Celiac disease is defined as a systemic immunological disorder caused by gluten (gliadin and other prolamin) in genetically predisposed individuals, who present with a variety of gluten-dependent symptoms, specific antibodies, the presence of the HLA DQ2 and DQ8 histocompatibility antigen, and enteropathy. Its prevalence, depending on the studied population and methodology, is estimated at 0.75-1.6% of the general population. During the complex immune reaction it induces, most cells involved in inflammatory processes are activated, which leads to the gradual atrophy of intestinal villi and the proliferation of enterocytes within intestinal crypts. The pathogenesis of celiac disease is extremely complicated and is still the subject of research. According to the current diagnostic guidelines, the following criteria should be taken into account: clinical symptoms (intestinal and extraintestinal), the presence of antibodies against tissue transglutaminase in the IgA class, the level of total IgA, and the presence of typical histological changes in duodenal biopsies. Diet-resistant celiac disease is one of the most important clinical challenges, causing serious complications. Currently, the basic method for treating celiac disease is an elimination diet (i.e., the exclusion of products that may contain gluten from the diet), however, new therapeutic strategies are still being sought, mainly based on supplementation with exogenous endopeptidases, modification of the immune response, and the use of zonulin inhibitors and transglutaminase 2 inhibitors. Clinical trials of new drugs are ongoing. The gradually expanding knowledge about the pathogenesis of celiac disease may allow for the development of new therapeutic strategies for both patients with a mild disease course, as well as those that are diet-resistant.
Collapse
Affiliation(s)
| | | | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Norbert Barlicki University Hospital, 90-153 Lodz, Poland; (M.K.K.); (D.D.-M.)
| |
Collapse
|
7
|
Lin TC, Lacorcia M, Mannering SI. Current and Emerging Assays for Measuring Human T-Cell Responses Against Beta-Cell Antigens in Type 1 Diabetes. Biomolecules 2025; 15:384. [PMID: 40149920 PMCID: PMC11939970 DOI: 10.3390/biom15030384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by T-cell mediated destruction of the pancreatic insulin-producing beta cells. Currently, the development of autoantibodies is the only measure of beta-cell autoimmunity used in the clinic. Despite T-cells' well-accepted role in the autoimmune pathogenesis of human T1D, autoimmune T-cell responses against beta cells remain very difficult to measure. An assay capable of measuring beta-cell antigen-specific T-cell responses has been a long-sought goal. Such an assay would facilitate the direct monitoring of T1D-associated T-cell responses facilitating, earlier diagnosis and rapid evaluation of candidate immune therapies in clinical trials. In addition, a simple and robust assay for beta-cell antigen-specific T-cell responses would be a powerful tool for dissecting the autoimmune pathogenesis of human T1D. Here, we review the challenges associated with measuring beta-cell antigen-specific T-cell responses, the current assays which are used to achieve this and, finally, we discuss BASTA, a promising emerging assay for measuring human beta-cell antigen-specific CD4+ T-cell responses.
Collapse
Affiliation(s)
| | | | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia; (T.-C.L.); (M.L.)
| |
Collapse
|
8
|
Galipeau HJ, Verdu EF. Is the microbiome important in celiac disease? J Can Assoc Gastroenterol 2025; 8:S51-S55. [PMID: 39990507 PMCID: PMC11842898 DOI: 10.1093/jcag/gwae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Celiac disease (CeD) is an autoimmune condition driven by gluten in genetically predisposed individuals. CeD is characterized by small intestinal villous atrophy but presents with a spectrum of gastrointestinal and systemic manifestations. Its only treatment is a strict, life-long adherence to a gluten-free diet, which is difficult to manage and does not always lead to symptomatic or mucosal recovery. About 40% of the population express the CeD-associated risk genes, but only 1%-2% of the worldwide population has CeD. This, along with the rising prevalence of CeD suggests other cofactors in disease pathogenesis. The gut microbiome has been implicated in CeD based on epidemiology studies and clinical associations. Mechanistic studies using relevant in vitro and in vivo preclinical models have begun to elucidate mechanisms through which microbes can influence CeD. Ultimately, a better understanding of these cofactors and their mechanisms will provide rationale intervention strategies and novel therapeutic targets to prevent or treat CeD.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton L8K4K1, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton L8K4K1, Canada
| |
Collapse
|
9
|
Repo M, Koskimaa S, Paavola S, Kurppa K. Serological testing for celiac disease in children. Expert Rev Gastroenterol Hepatol 2025; 19:155-164. [PMID: 39893645 DOI: 10.1080/17474124.2025.2462245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Celiac disease is one of the most common chronic immune-mediated gastrointestinal conditions, characterized by the presence of disease-specific serum antibodies against self-antigen transglutaminase 2. Even though modern serological assays can identify most untreated celiac disease patients and are also increasingly being used to establish a diagnosis, several challenges are associated with the tests, including a lack of standardization, the variable sensitivity and specificity of commercial assays, and inadequate sensitivity for monitoring adherence to a gluten-free diet. AREAS COVERED This narrative review outlines the current use of serological tests in case-finding and screening, as well as in the follow-up of dietary treatment. Additionally, the possible challenges and pitfalls of serological tests, along with future directions, are addressed. EXPERT OPINION The excellent accuracy of modern autoantibody tests, especially for greatly elevated levels of transglutaminase 2 antibodies and positive endomysial antibodies, enables using serological testing in establishing a diagnosis. However, better international standardization of the assays is required, the necessity of endomysial antibody testing needs to be further scrutinized, and additional research is needed to improve noninvasive tools for follow-up and to further expand the no-biopsy criteria for celiac disease.
Collapse
Affiliation(s)
- Marleena Repo
- Department of Pediatrics, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Sara Koskimaa
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Saana Paavola
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Kalle Kurppa
- Department of Pediatrics, Tampere University Hospital and Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
- The University Consortium of Seinäjoki, Seinäjoki, Finland
| |
Collapse
|
10
|
Tye‐Din JA. Evolution in coeliac disease diagnosis and management. JGH Open 2024; 8:e13107. [PMID: 38957478 PMCID: PMC11217771 DOI: 10.1002/jgh3.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/04/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
The traditional gut-centric view of coeliac disease is evolving as immune and genetic insights underscore the central importance of a systemic, T cell immune response to gluten in disease pathogenesis. As the field increasingly recognize the limitations of small intestinal histology as the diagnostic standard, data supporting the accuracy of an immune (serologic) diagnosis of coeliac disease - well demonstrated in children - are growing for adults. Novel biomarkers such as interleukin-2 that identify the gluten-specific T cell demonstrate high sensitivity and specificity for coeliac disease and offer the potential for a diagnostic approach that avoids the need for gluten challenge. Asymptomatic disease and manifestations outside the gut pose considerable challenges for diagnosis using a case-finding strategy and enthusiasm for population screening is growing. The gluten-free diet remains a highly restrictive treatment and there is a paucity of controlled data to inform a safe gluten intake threshold. Ongoing symptoms and enteropathy are common and require systematic evaluation. Slowly-responsive disease is prevalent in the older patient diagnosed with coeliac disease, and super-sensitivity to gluten is an emerging concept that may explain many cases of nonresponsive disease. While there is great interest in developing novel therapies for coeliac disease, no drug has yet been registered. Efficacy studies are generally assessing drugs in patients with treated coeliac disease who undergo gluten challenge or in patients with nonresponsive disease; however, substantial questions remain around specific endpoints relevant for patients, clinicians and regulatory agencies and optimal trial design. Novel immune tools are providing informative readouts for clinical trials and are now shaping their design.
Collapse
Affiliation(s)
- Jason A Tye‐Din
- Immunology DivisionWalter and Eliza Hall InstituteParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of GastroenterologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Centre for Food & Allergy ResearchThe Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| |
Collapse
|
11
|
Anderson RP, Verma R, Schumann M. A Look Into the Future: Are We Ready for an Approved Therapy in Celiac Disease? Gastroenterology 2024; 167:183-193. [PMID: 38355059 DOI: 10.1053/j.gastro.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
As it appears that we are currently at the cusp of an era in which drugs that are new, re-purposed, or "supplements" will be introduced to the management of celiac disease, we need to reflect on whether the framework is set for celiac disease to be treated increasingly with pharmaceuticals as well as diet. This refers to reflecting on the rigor of current diagnostic practices; the limitations of the current standard of care, which is a gluten-free diet; and that we lack objective markers of disease severity. Investigating these issues will help us to identify gaps in technology and practices that could be critical for selecting patients with a well-defined need for an improved or alternative treatment. Both aspects, circumscribed limitations of the gluten-free diet and diagnostics helping to define celiac disease target groups, together with the guiding requirements by the responsible regulatory authorities, will contribute to defining the subgroups of patients with confirmed celiac disease eligible for distinct pharmacologic strategies. Because many patients with celiac disease are diagnosed in childhood, these aspects need to be differentially discussed for the pediatric setting. In this perspective, we aimed to describe these contextual issues and then looked ahead to the future. What might be the major challenges in celiac disease clinics in the coming years once drugs are an option alongside diet? And what will be the future objectives for researchers who further decipher the mucosal immunology of celiac disease? Speculating on the answers to these questions is as stimulating as it is fascinating to be part of this turning point.
Collapse
Affiliation(s)
- Robert P Anderson
- Gastroenterology Service, Mackay Base Hospital, West Mackay, Queensland, Australia
| | - Ritu Verma
- University of Chicago, Comer Children's Hospital, Chicago, Illinois
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Abadie V, Han AS, Jabri B, Sollid LM. New Insights on Genes, Gluten, and Immunopathogenesis of Celiac Disease. Gastroenterology 2024; 167:4-22. [PMID: 38670280 PMCID: PMC11283582 DOI: 10.1053/j.gastro.2024.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois.
| | - Arnold S Han
- Columbia Center for Translational Immunology, Columbia University, New York, New York; Department of Microbiology and Immunology, Columbia University, New York, New York; Department of Medicine, Digestive and Liver Diseases, Columbia University, New York, New York
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois; Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
13
|
Silvester JA, Elli L, Khosla C, Tye-Din JA. Past, Present, and Future of Noninvasive Tests to Assess Gluten Exposure, Celiac Disease Activity, and End-Organ Damage. Gastroenterology 2024; 167:159-171. [PMID: 38670279 PMCID: PMC11235091 DOI: 10.1053/j.gastro.2024.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 04/28/2024]
Abstract
Although many biomarkers have been proposed, and several are in widespread clinical use, there is no single readout or combination of readouts that correlates tightly with gluten exposure, disease activity, or end-organ damage in treated patients with celiac disease. Challenges to developing and evaluating better biomarkers include significant interindividual variability-related to immune amplification of gluten exposure and how effects of immune activation are manifest. Furthermore, the current "gold standard" for assessment of end-organ damage, small intestinal biopsy, is itself highly imperfect, such that a marker that is a better reflection of the "ground truth" may indeed appear to perform poorly. The goal of this review was to analyze past and present efforts to establish robust noninvasive tools for monitoring treated patients with celiac disease and to highlight emerging tools that may prove to be useful in clinical practice.
Collapse
Affiliation(s)
- Jocelyn A Silvester
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Harvard Celiac Disease Research Program, Harvard Medical School, Boston, Massachusetts.
| | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chaitan Khosla
- Sarafan ChEM-H, Departments of Chemistry and Chemical Engineering, Stanford University, Stanford, California
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia; Department of Gastroenterology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Raju SA, Shiha MG, Penny HA. Monitoring coeliac disease in 2024, time to change practice? Curr Opin Gastroenterol 2024; 40:190-195. [PMID: 38547329 DOI: 10.1097/mog.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Persistent villous atrophy is associated with morbidity in coeliac disease and most commonly due to ongoing gluten ingestion. Current methods for assessing gluten exposure and persisting villous atrophy include dietary questionnaires and repeat duodenal biopsy, which have limited accuracy or are invasive. This review discusses adjunctive and/or novel tests that could be used to overcome these challenges. RECENT FINDINGS Small bowel capsule endoscopy is well tolerated and helps to evaluate for persisting villous atrophy and importantly, complications associated with coeliac disease. Testing for urinary and/or stool gluten immunogenic peptides may help identify recent gluten exposure, but further studies are still warranted to evaluate the accuracy and applicability of this approach. Measuring spikes in circulating Interleukin-2 following gluten challenge has shown promise for coeliac disease diagnosis, and thus may serve as a useful confirmatory test in those with persisting symptoms but provides no information on mucosal inflammation. No specific gut microbial signature has been identified in coeliac disease; however, studies have shown a reduced microbial diversity in active disease, which with future refinement may prove clinically useful. SUMMARY There is no evidence to support alternative methods for assessing persisting villous atrophy in coeliac disease over performing an up-to-date duodenal biopsy. Monitoring for adherence to a gluten-free diet remains clinically challenging and should be a priority for future research.
Collapse
Affiliation(s)
- Suneil A Raju
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Mohamed G Shiha
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Hugo A Penny
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
15
|
Henneken LM, Loh TJ, Ciacchi L, Ciacchi L, Lim JJ, Reid HH, Tye-Din JA. Bridging science and accessibility: a tactile journey from gluten through to coeliac disease. Immunol Cell Biol 2024; 102:331-335. [PMID: 38111272 DOI: 10.1111/imcb.12716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
As part of the Monash Sensory Science Exhibition, our team guided participants through a multisensory journey unraveling coeliac disease development and pathology. Through tactile and sensory exhibits, we showed how benign dietary gluten can be transformed into a harmful entity for the 1 in 70 Australians with this illness. In contrast to the common misconception of coeliac disease as a food allergy, our exhibits revealed its closer association with autoimmune diseases such as type 1 diabetes, involving genetic susceptibility linked to specific human leukocyte antigens, crucial antigen-specific T- and B-cell responses and autoantibody production. Tactile models underscored the severe consequences of the proinflammatory immune response to gluten on patient health and quality of life. This educational event affirmed to us the value and importance of fostering inclusivity in science education.
Collapse
Affiliation(s)
- Lee M Henneken
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Tiing Jen Loh
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Laura Ciacchi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lisa Ciacchi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jia-Jia Lim
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Hugh H Reid
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jason A Tye-Din
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC, Australia
- Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia
| |
Collapse
|
16
|
Sollid LM. Tolerance-inducing therapies in coeliac disease - mechanisms, progress and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:335-347. [PMID: 38336920 DOI: 10.1038/s41575-024-00895-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Coeliac disease is an autoinflammatory condition caused by immune reactions to cereal gluten proteins. Currently, the only available treatment for the condition is a lifelong avoidance of gluten proteins in the diet. There is an unmet need for alternative therapies. Coeliac disease has a strong association with certain HLA-DQ allotypes (DQ2.5, DQ2.2 and DQ8), and these disease-associated HLA-DQ molecules present deamidated gluten peptides to gluten-specific CD4+ T cells. The gluten-specific CD4+ T cells are the drivers of the immune reactions leading to coeliac disease. Once established, the clonotypes of gluten-specific CD4+ T cells persist for decades, explaining why patients must adhere to a gluten-free diet for life. Given the key pathogenic role of gluten-specific CD4+ T cells, tolerance-inducing therapies that target these T cells are attractive for treatment of the disorder. Lessons learned from coeliac disease might provide clues for treatment of other HLA-associated diseases for which the disease-driving antigens are unknown. Thus, intensive efforts have been and are currently implemented to bring an effective tolerance-inducing therapy for coeliac disease. This Review discusses mechanisms of the various approaches taken, summarizing the progress made, and highlights future directions in this field.
Collapse
Affiliation(s)
- Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
17
|
Crepaldi M, Palo M, Maniero D, Bertin L, Savarino EV, Anderson RP, Zingone F. Emerging Pharmaceutical Therapies to Address the Inadequacy of a Gluten-Free Diet for Celiac Disease. Pharmaceuticals (Basel) 2023; 17:4. [PMID: 38275990 PMCID: PMC10821495 DOI: 10.3390/ph17010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Celiac disease (CeD) is a chronic autoimmune disorder triggered by the ingestion of gluten, affecting around 1% of the global population. It is a multifactorial disease involving both genetics and environmental factors. Nowadays, the only available treatment for CeD is a life-long gluten-free diet (GFD), which can cause a significant burden for patients, since symptoms and mucosal injury can persist despite apparent compliance with a GFD. This could also lead to psychological consequences and affect the quality of life of these patients. Thankfully, recent advances in understanding the pathogenesis of CeD and the availability of various targets have made it feasible to explore pharmaceutical treatments specific to CeD. Recently, the FDA has highlighted the unmet needs of adult patients on a GFD who experience ongoing symptoms attributed to CeD and also show persistent duodenal villous atrophy. This review will outline the limitations of a GFD, describe the targets of potential novel treatment of CeD and provide an overview of the primary clinical trials involving oral and injectable agents for a non-dietary treatment of CeD.
Collapse
Affiliation(s)
- Martina Crepaldi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Michela Palo
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| | - Robert P. Anderson
- Gastroenterology Department, Mackay Base Hospital, Mackay, QLD 4740, Australia
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy; (M.C.); (M.P.); (D.M.); (L.B.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy
| |
Collapse
|
18
|
Syage J, Ramos A, Loskutov V, Norum A, Bledsoe A, Choung RS, Dickason M, Sealey-Voyksner J, Murray J. Dynamics of Serologic Change to Gluten in Celiac Disease Patients. Nutrients 2023; 15:5083. [PMID: 38140342 PMCID: PMC10746107 DOI: 10.3390/nu15245083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Serologic measures of tissue transglutaminase (tTG) immunoglobulin A (IgA) and deamidated gliadin peptide (DGP) IgA and immunoglobulin G (IgG) are hallmark tests utilized when diagnosing individuals for celiac disease (CeD) and for monitoring adherence to a gluten-free diet (GFD), currently the only available treatment for CeD. We address two issues in this study: (i) the relapse to seropositivity for CeD patients who resume a gluten containing diet and (ii) the correlation between two different tTG-IgA assays near the upper limit of normal (ULN) designated thresholds. Regarding the first issue, often a suspected CeD individual is put back on a gluten diet to return to their serologic levels. However, we show it requires a substantial amount of gluten for serology to return to a positive level. For example, in one study of 22 patients treated with placebo and taking 84 g of gluten over 6 weeks, only two converted from seronegative to seropositive for tTG-IgA. Regarding the second topic, we compare the relationship for different serologic assays, namely tTG-IgA AB (recombinant, ULN = 4 units/mL) vs. tTG-IgA (non-recombinant, ULN = 20 units). There is a strong correlation between both measurements as evidenced by a Pearson coefficient of R = 0.8584; however, we observed that the cross-correlation in terms of sensitivity and specificity improved substantially by using an ULN value of three instead of four for the tTG-IgA AB (recombinant) assay. This result suggests that assay thresholds used for initial diagnosis in patients who have not yet started a GFD may need to be adjusted for monitoring and in the setting of a diagnostic gluten challenge.
Collapse
Affiliation(s)
- Jack Syage
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Ana Ramos
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Vasiliy Loskutov
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Anna Norum
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Adam Bledsoe
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Rok Seon Choung
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew Dickason
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Jennifer Sealey-Voyksner
- ImmunogenX, Inc., 1600 Dove Street, Suite 330, Newport Beach, CA 92660, USA; (A.R.); (V.L.); (M.D.)
| | - Joseph Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Fowler A, FitzPatrick M, Shanmugarasa A, Ibrahim ASF, Kockelbergh H, Yang HC, Williams-Walker A, Luu Hoang KN, Evans S, Provine N, Klenerman P, Soilleux EJ. An Interpretable Classification Model Using Gluten-Specific TCR Sequences Shows Diagnostic Potential in Coeliac Disease. Biomolecules 2023; 13:1707. [PMID: 38136579 PMCID: PMC10742135 DOI: 10.3390/biom13121707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Coeliac disease (CeD) is a T-cell mediated enteropathy triggered by dietary gluten which remains substantially under-diagnosed around the world. The diagnostic gold-standard requires histological assessment of intestinal biopsies taken at endoscopy while consuming a gluten-containing diet. However, there is a lack of concordance between pathologists in histological assessment, and both endoscopy and gluten challenge are burdensome and unpleasant for patients. Identification of gluten-specific T-cell receptors (TCRs) in the TCR repertoire could provide a less subjective diagnostic test, and potentially remove the need to consume gluten. We review published gluten-specific TCR sequences, and develop an interpretable machine learning model to investigate their diagnostic potential. To investigate this, we sequenced the TCR repertoires of mucosal CD4+ T cells from 20 patients with and without CeD. These data were used as a training dataset to develop the model, then an independently published dataset of 20 patients was used as the testing dataset. We determined that this model has a training accuracy of 100% and testing accuracy of 80% for the diagnosis of CeD, including in patients on a gluten-free diet (GFD). We identified 20 CD4+ TCR sequences with the highest diagnostic potential for CeD. The sequences identified here have the potential to provide an objective diagnostic test for CeD, which does not require the consumption of gluten.
Collapse
Affiliation(s)
- Anna Fowler
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool L69 3GF, UK
| | - Michael FitzPatrick
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (M.F.); (P.K.)
| | | | - Amro Sayed Fadel Ibrahim
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| | - Hannah Kockelbergh
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool L69 3GF, UK
| | - Han-Chieh Yang
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| | - Amelia Williams-Walker
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| | - Kim Ngan Luu Hoang
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| | - Shelley Evans
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| | - Nicholas Provine
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (M.F.); (P.K.)
| | - Paul Klenerman
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (M.F.); (P.K.)
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, UK
| | - Elizabeth J. Soilleux
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; (A.S.F.I.); (H.-C.Y.); (A.W.-W.); (K.N.L.H.); (S.E.); (E.J.S.)
| |
Collapse
|
20
|
Popp A, Laurikka P, Czika D, Kurppa K. The role of gluten challenge in the diagnosis of celiac disease: a review. Expert Rev Gastroenterol Hepatol 2023; 17:691-700. [PMID: 37243608 DOI: 10.1080/17474124.2023.2219893] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/12/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Duodenal biopsy is the gold standard in the diagnosis of celiac disease, with increasing utilization of serology. A gluten challenge may be required, for example, when dietary gluten reduction precedes appropriate diagnostic evaluations. Evidence on the best challenge protocol is currently sparse. Pharmaceutical trials in recent years may have provided new insights into the challenge and advanced the development of novel sensitive histological and immunological methods. AREAS COVERED This review outlines the current perspectives on the use of gluten challenge in the diagnosis of celiac disease and explores future directions in this area. EXPERT OPINION Comprehensive elimination of celiac disease before dietary gluten restriction is essential to avoid diagnostic uncertainties. Gluten challenge continues to have an important role in certain clinical scenarios, although it is important to understand its limitations in the diagnostic evaluation. The evidence so far permits no unequivocal recommendation considering the timing, duration, and amount of gluten used in the challenge. Thus, these decisions should be made on a case-by-case basis. Further studies with more standardized protocols and outcome measures are called for. In the future novel immunological methods may help to shorten or even avoid gluten challenge.
Collapse
Affiliation(s)
- Alina Popp
- Department of Pediatrics, University of Medicine and Pharmacy Carol Davila and National Institute for Mother and Child Health, Bucharest, Romania
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pilvi Laurikka
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Diana Czika
- Department of Pediatrics, University of Medicine and Pharmacy Carol Davila and National Institute for Mother and Child Health, Bucharest, Romania
| | - Kalle Kurppa
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
- The University Consortium of Seinäjoki, Seinäjoki, Finland
| |
Collapse
|
21
|
Rodríguez-Sillke Y, Schumann M, Lissner D, Branchi F, Proft F, Steinhoff U, Siegmund B, Glauben R. Analysis of Circulating Food Antigen-Specific T-Cells in Celiac Disease and Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:ijms24098153. [PMID: 37175860 PMCID: PMC10179603 DOI: 10.3390/ijms24098153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
To demonstrate and analyze the specific T-cell response following barrier disruption and antigen translocation, circulating food antigen-specific effector T-cells isolated from peripheral blood were analyzed in patients suffering from celiac disease (CeD) as well as inflammatory bowel disease (IBD). We applied the antigen-reactive T-cell enrichment (ARTE) technique allowing for phenotypical and functional flow cytometric analyses of rare nutritional antigen-specific T-cells, including the celiac disease-causing gliadin (gluten). For CeD, patient groups, including treatment-refractory cases, differ significantly from healthy controls. Even symptom-free patients on a gluten-free diet were distinguishable from healthy controls, without being previously challenged with gluten. Moreover, frequency and phenotype of nutritional antigen-specific T-cells of IBD patients directly correlated to the presence of small intestinal inflammation. Specifically, the frequency of antigen specific T-cells as well as pro-inflammatory cytokines was increased in patients with active CeD or Crohn's disease, respectively. These results suggest active small intestinal inflammation as key for the development of a peripheral food antigen-specific T-cell response in Crohn's disease and celiac disease.
Collapse
Affiliation(s)
- Yasmina Rodríguez-Sillke
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
- Institute of Nutrition, University of Potsdam, 14558 Nuthetal, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| | - Donata Lissner
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| | - Federica Branchi
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| | - Fabian Proft
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hospital Hygiene, Philipps University of Marburg, 35043 Marburg, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| | - Rainer Glauben
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine Berlin, 13125 Berlin, Germany
| |
Collapse
|
22
|
Shiha MG, Raju SA, Sidhu R, Penny HA. The debate in the diagnosis of coeliac disease - time to go 'no-biopsy'? Curr Opin Gastroenterol 2023; 39:192-199. [PMID: 37144537 DOI: 10.1097/mog.0000000000000929] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
PURPOSE OF REVIEW Duodenal biopsies have been central to making a diagnosis of coeliac disease for the last 70 years. Recent paediatric guidelines have reduced the emphasis on duodenal biopsies with the incorporation of a 'no-biopsy' arm to the diagnostic pathway. This review discusses the no-biopsy approach in adults and highlights advances in alternative (non-biopsy) diagnostic modalities in coeliac disease. RECENT FINDINGS Evidence suggests that a no-biopsy approach for the diagnosis of adult coeliac disease is accurate. However, a number of factors still favour duodenal biopsy sampling in specific patient groups. Moreover, several factors need to be considered if this pathway is implemented into local gastroenterology services. SUMMARY Duodenal biopsies remain an important step in the diagnosis of adult coeliac disease. However, an alternative approach that removes the necessity for biopsies may be an option in selected adults. If further guidelines incorporate this pathway, then efforts should focus on supporting a dialogue between primary and secondary care to facilitate the appropriate implementation of this approach.
Collapse
Affiliation(s)
- Mohamed G Shiha
- Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust
- Academic Unit of Gastroenterology, Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Suneil A Raju
- Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust
- Academic Unit of Gastroenterology, Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Reena Sidhu
- Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust
- Academic Unit of Gastroenterology, Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Hugo A Penny
- Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust
- Academic Unit of Gastroenterology, Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
23
|
Efficacy and safety of gluten peptide-based antigen-specific immunotherapy (Nexvax2) in adults with coeliac disease after bolus exposure to gluten (RESET CeD): an interim analysis of a terminated randomised, double-blind, placebo-controlled phase 2 study. Lancet Gastroenterol Hepatol 2023; 8:446-457. [PMID: 36898393 DOI: 10.1016/s2468-1253(22)00428-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 03/09/2023]
Abstract
BACKGROUND A gluten-free diet is insufficient to treat coeliac disease because intestinal injury persists and acute reactions with cytokine release follow gluten exposure. Nexvax2 is a specific immunotherapy using immunodominant peptides recognised by gluten-specific CD4+ T cells that might modify gluten-induced disease in coeliac disease. We aimed to assess the effects of Nexvax2 on gluten-induced symptoms and immune activation in patients with coeliac disease. METHODS This was a randomised, double-blind, placebo-controlled phase 2 trial done at 41 sites (29 community, one secondary, and 11 tertiary centres) in the USA, Australia, and New Zealand. Patients with coeliac disease aged 18-70 years who had excluded gluten for at least 1 year, were HLA-DQ2.5 positive, and had a worsening of symptoms after an unmasked 10 g vital gluten challenge were eligible for inclusion. Patients were stratified by HLA-DQ2.5 status (HLA-DQ2.5 non-homozygous vs homozygous). Patients who were non-homozygous were centrally (ICON; Dublin, Ireland) randomly assigned (1:1) to receive subcutaneous Nexvax2 (non-homozygous Nexvax2 group) or saline (0·9% sodium chloride; non-homozygous placebo group) twice a week escalating from 1 μg to 750 μg during the first 5 weeks followed by 11 weeks of maintenance therapy at 900 μg per dose. The exploratory homozygous group was centrally randomly assigned (2:1) to receive Nexvax2 (homozygous Nexvax2 group) or placebo (homozygous placebo group); patients who were homozygous received the same doseage as those who were non-homozygous. The primary endpoint was change in coeliac disease patient reported outcomes (total gastrointestinal domain) from pretreatment baseline to the day of masked bolus 10 g vital gluten challenge given in week 14 analysed in the non-homozygous intention-to-treat population. The trial is registered with ClinicalTrials.gov, NCT03644069. FINDINGS Between Sept 21, 2018, and April 24, 2019, 383 volunteers were screened for inclusion, of whom 179 (47%; 133 [74%] women, 46 [26%] men; median age 41 years [IQR 33-55]) were randomly assigned. One (1%) of 179 patients was excluded from analysis due to misassignment of genotype. The non-homozygous Nexvax2 group included 76 patients, the non-homozygous placebo group included 78 patients, the homozygous Nexvax2 group included 16 patients, and the homozygous placebo group included eight patients. The study was discontinued after planned interim analysis of 66 patients who were non-homozygous. We report an unmasked post-hoc analysis of all available data for the primary endpoint and secondary symptom-based endpoints combining data from 67 (66 were assessed in the planned interim analysis for the primary endpoint). Mean change from baseline to day of first masked gluten challenge in total gastrointestinal score for the non-homozygous Nexvax2 group was 2·86 (SD 2·28) compared with 2·63 (2·07) for the non-homozygous placebo group (p=0·43). Adverse events were similar between all patients who received Nexvax2 and those who received placebo. Serious adverse events were reported in five (3%) of 178 patients (two [2%] of 92 who received Nexvax2 and three [4%] of 82 who received placebo). One patient in the non-homozygous Nexvax2 group had a serious adverse event that occurred during gluten challenge (left-sided mid-back muscle strain with imaging suggestive of partial left kidney infarction). Serious adverse events were reported for three (4%) of 78 patients in the non-homozygous placebo group (one each with exacerbation of asthma and appendicitis, and one who had forehead abscess, conjunctivitis, and folliculitis) and one (1%) patient in the non-homozygous Nexvax2 group developed a pulmonary embolism. The most frequent adverse events in all 92 patients who received Nexvax2 compared with all 86 patients who received placebo were nausea (44 [48%] of 92 patients who received Nexvax2 vs 29 (34%) of 86 patients who received placebo), diarrhoea (32 [35%] vs 25 [29%]), abdominal pain (31 [34%] vs 27 [31%]), headache 32 [35%] vs 20 [23%]), and fatigue (24 [26%] vs 31 [36%]). INTERPRETATION Nexvax2 did not reduce acute gluten-induced symptoms. Masked bolus vital gluten challenge provides an alternative to extended gluten challenge in efficacy studies for coeliac disease. FUNDING ImmusanT.
Collapse
|
24
|
Anderson RP. Review article: Diagnosis of coeliac disease: a perspective on current and future approaches. Aliment Pharmacol Ther 2022; 56 Suppl 1:S18-S37. [PMID: 35815826 DOI: 10.1111/apt.16840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/09/2022]
Abstract
Diagnostics will play a central role in addressing the ongoing dramatic rise in global prevalence of coeliac disease, and in deploying new non-dietary therapeutics. Clearer understanding of the immunopathogenesis of coeliac disease and the utility of serology has led to partial acceptance of non-biopsy diagnosis in selected cases. Non-biopsy diagnosis may expand further because research methods for measuring gluten-specific CD4+ T cells and the acute recall response to gluten ingestion in patients is now relatively straightforward. This perspective on diagnosis in the context of the immunopathogenesis of coeliac disease sets out to highlight current consensus, limitations of current practices, gluten food challenge for diagnosis and the potential for diagnostics that measure the underlying cause for coeliac disease, gluten-specific immunity.
Collapse
|
25
|
Pinto-Sanchez MI, Silvester JA, Lebwohl B, Leffler DA, Anderson RP, Therrien A, Kelly CP, Verdu EF. Society for the Study of Celiac Disease position statement on gaps and opportunities in coeliac disease. Nat Rev Gastroenterol Hepatol 2021; 18:875-884. [PMID: 34526700 PMCID: PMC8441249 DOI: 10.1038/s41575-021-00511-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 02/08/2023]
Abstract
Progress has been made in understanding coeliac disease, a relatively frequent and underappreciated immune-mediated condition that occurs in genetically predisposed individuals. However, several gaps remain in knowledge related to diagnosis and management. The gluten-free diet, currently the only available management, is not curative or universally effective (some adherent patients have ongoing duodenal injury). Unprecedented numbers of emerging therapies, including some with novel tolerogenic mechanisms, are currently being investigated in clinical trials. In March 2020, the Celiac Disease Foundation and the Society for the Study of Celiac Disease convened a consensus workshop to identify high-yield areas of research that should be prioritized. Workshop participants included leading experts in clinical practice, academia, government and pharmaceutical development, as well as representatives from patient support groups in North America. This Roadmap summarizes key advances in the field of coeliac disease and provides information on important discussions from the consensus approach to address gaps and opportunities related to the pathogenesis, diagnosis and management of coeliac disease. The morbidity of coeliac disease is often underestimated, which has led to an unmet need to improve the management of these patients. Expanded research funding is needed as coeliac disease is a potentially curable disease.
Collapse
Affiliation(s)
- M Ines Pinto-Sanchez
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
- McMaster University Medical Center, Hamilton, Ontario, Canada
| | - Jocelyn A Silvester
- Harvard Medical School Celiac Research Program, Boston, MA, USA
- Boston Children's Hospital, Boston, MA, USA
- Celiac Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Daniel A Leffler
- Harvard Medical School Celiac Research Program, Boston, MA, USA
- Celiac Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Takeda Pharmaceuticals, Cambridge Massachusetts, Cambridge, MA, USA
| | - Robert P Anderson
- Wesley Medical Research, The Wesley Hospital, Auchenflower, Queensland, Australia
| | - Amelie Therrien
- Harvard Medical School Celiac Research Program, Boston, MA, USA
- Celiac Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ciaran P Kelly
- Harvard Medical School Celiac Research Program, Boston, MA, USA
- Celiac Center, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada.
- McMaster University Medical Center, Hamilton, Ontario, Canada.
| |
Collapse
|
26
|
Anderson RP. Emergence of an adaptive immune paradigm to explain celiac disease: a perspective on new evidence and implications for future interventions and diagnosis. Expert Rev Clin Immunol 2021; 18:75-91. [PMID: 34767744 DOI: 10.1080/1744666x.2021.2006636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Recent patient studies have shown that gluten-free diet is less effective in treating celiac disease than previously believed, and additionally patients remain vulnerable to gluten-induced acute symptoms and systemic cytokine release. Safe and effective pharmacological adjuncts to gluten-free diet are in preclinical and clinical development. Clear understanding of the pathogenesis of celiac disease is critical for drug target identification, establishing efficacy endpoints and to develop non-invasive biomarkers suitable to monitor and potentially diagnose celiac disease. AREAS COVERED The role and clinical effects of CD4+ T cells directed against deamidated gluten in the context of an "adaptive immune paradigm" are reviewed. Alternative hypotheses of gluten toxicity are discussed and contrasted. In the context of recent patient studies, implications of the adaptive immune paradigm for future strategies to prevent, diagnose, and treat celiac disease are outlined. EXPERT OPINION Effective therapeutics for celiac disease are likely to be approved and necessitate a variety of new clinical instruments and tests to stratify patient need, monitor remission, and confirm diagnosis in uncertain cases. Sensitive assessments of CD4+ T cells specific for deamidated gluten are likely to play a central role in clinical management, and to facilitate research and pharmaceutical development.
Collapse
|
27
|
Fernández-Bañares F, López-Palacios N, Corzo M, Arau B, Rubio M, Fernández-Prieto M, Tristán E, Pujals M, Farrais S, Horta S, Hernández JM, Gomez-Perosanz M, Reche PA, Esteve M, Núñez C. Activated gut-homing CD8 + T cells for coeliac disease diagnosis on a gluten-free diet. BMC Med 2021; 19:237. [PMID: 34610833 PMCID: PMC8493675 DOI: 10.1186/s12916-021-02116-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The diagnosis of coeliac disease (CD) in individuals that have started a gluten-free diet (GFD) without an adequate previous diagnostic work-out is a challenge. Several immunological assays such as IFN-γ ELISPOT have been developed to avoid the need of prolonged gluten challenge to induce the intestinal damage. We aimed to evaluate the diagnostic accuracy of activated gut-homing CD8+ and TCRγδ+ T cells in blood after a 3-day gluten challenge and to compare it with the performance of IFN-γ ELISPOT in a HLA-DQ2.5 subsample. METHODS A total of 22 CD patients and 48 non-CD subjects, all of them following a GFD, underwent a 3-day 10-g gluten challenge. The percentage of two T cell subsets (CD8+ CD103+ β7hi CD38+/total CD8+ and TCRγδ+ CD103+ β7hi CD38+/total TCRγδ+) in fresh peripheral blood drawn baseline and 6 days after the challenge was determined by flow cytometry. IFN-γ ELISPOT assays were also performed in HLA-DQ2.5 participants. ROC curve analysis was used to assess the diagnostic performance of the CD8+ T cell response and IFN-γ ELISPOT. RESULTS Significant differences between the percentage of the two studied subsets of CD8+ and TCRγδ+ cells at days 0 and 6 were found only when considering CD patients (p < 10-3 vs. non-CD subjects). Measuring activated CD8+ T cells provided accurate CD diagnosis with 95% specificity and 97% sensitivity, offering similar results than IFN-γ ELISPOT. CONCLUSIONS The results provide a highly accurate blood test for CD diagnosis in patients on a GFD of easy implementation in daily clinical practice.
Collapse
Affiliation(s)
- Fernando Fernández-Bañares
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia López-Palacios
- Servicio de Aparato Digestivo, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - María Corzo
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Beatriz Arau
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Rubio
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Marta Fernández-Prieto
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Eva Tristán
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Pujals
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain
| | - Sergio Farrais
- Servicio de Aparato Digestivo, Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
| | - Saúl Horta
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Juana María Hernández
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain
| | - Marta Gomez-Perosanz
- Facultad de Medicina, Laboratorio de Inmunomedicina, Departamento de Inmunología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Pedro A Reche
- Facultad de Medicina, Laboratorio de Inmunomedicina, Departamento de Inmunología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - María Esteve
- Department of Gastroenterology, Hospital Universitari Mutua Terrassa, Terrassa, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Concepción Núñez
- Laboratorio de Investigación en Genética de enfermedades complejas, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain.
| |
Collapse
|
28
|
Hardy MY, Goel G, Russell AK, Chen Yi Mei SLG, Brown GJE, Wang S, Szymczak E, Zhang R, Goldstein KE, Neff KM, Williams LJ, Truitt KE, Dzuris JL, Tye-Din JA, Anderson RP. A Sensitive Whole Blood Assay Detects Antigen-Stimulated Cytokine Release From CD4+ T Cells and Facilitates Immunomonitoring in a Phase 2 Clinical Trial of Nexvax2 in Coeliac Disease. Front Immunol 2021; 12:661622. [PMID: 34093551 PMCID: PMC8171185 DOI: 10.3389/fimmu.2021.661622] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022] Open
Abstract
Improved blood tests assessing the functional status of rare gluten-specific CD4+ T cells are needed to effectively monitor experimental therapies for coeliac disease (CD). Our aim was to develop a simple, but highly sensitive cytokine release assay (CRA) for gluten-specific CD4+ T cells that did not require patients to undergo a prior gluten challenge, and would be practical in large, multi-centre clinical trials. We developed an enhanced CRA and used it in a phase 2 clinical trial (“RESET CeD”) of Nexvax2, a peptide-based immunotherapy for CD. Two participants with treated CD were assessed in a pilot study prior to and six days after a 3-day gluten challenge. Dye-dilution proliferation in peripheral blood mononuclear cells (PBMC) was assessed, and IL-2, IFN-γ and IL-10 were measured by multiplex electrochemiluminescence immunoassay (ECL) after 24-hour gluten-peptide stimulation of whole blood or matched PBMC. Subsequently, gluten-specific CD4+ T cells in blood were assessed in a subgroup of the RESET CeD Study participants who received Nexvax2 (maintenance dose 900 μg, n = 12) or placebo (n = 9). The pilot study showed that gluten peptides induced IL-2, IFN-γ and IL-10 release from PBMCs attributable to CD4+ T cells, but the PBMC CRA was substantially less sensitive than whole blood CRA. Only modest gluten peptide-stimulated IL-2 release could be detected without prior gluten challenge using PBMC. In contrast, whole blood CRA enabled detection of IL-2 and IFN-γ before and after gluten challenge. IL-2 and IFN-γ release in whole blood required more than 6 hours incubation. Delay in whole blood incubation of more than three hours from collection substantially reduced antigen-stimulated IL-2 and IFN-γ secretion. Nexvax2, but not placebo treatment in the RESET CeD Study was associated with significant reductions in gluten peptide-stimulated whole blood IL-2 and IFN-γ release, and CD4+ T cell proliferation. We conclude that using fresh whole blood instead of PBMC substantially enhances cytokine secretion stimulated by gluten peptides, and enables assessment of rare gluten-specific CD4+ T cells without requiring CD patients to undertake a gluten challenge. Whole blood assessment coupled with ultra-sensitive cytokine detection shows promise in the monitoring of rare antigen-specific T cells in clinical studies.
Collapse
Affiliation(s)
- Melinda Y Hardy
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Gautam Goel
- ImmusanT, Inc., Cambridge, MA, United States
| | - Amy K Russell
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | | | - Gregor J E Brown
- Department of Gastroenterology, Alfred Hospital, Prahran, VIC, Australia
| | - Suyue Wang
- ImmusanT, Inc., Cambridge, MA, United States
| | | | - Ruan Zhang
- ImmusanT, Inc., Cambridge, MA, United States
| | | | | | | | | | | | - Jason A Tye-Din
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | | |
Collapse
|