Open access peer-reviewed chapter

Celiac Disease, Management, and Follow-Up

Written By

Ángela Ruiz-Carnicer, Verónica Segura, Carolina Sousa and Isabel Comino

Submitted: 01 March 2022 Reviewed: 24 March 2022 Published: 20 May 2022

DOI: 10.5772/intechopen.104652

From the Edited Volume

Immunology of the GI Tract - Recent Advances

Edited by Luis Rodrigo

Chapter metrics overview

134 Chapter Downloads

View Full Metrics

Abstract

Celiac disease (CD) is a systemic immune-mediated disorder characterized by a specific serological and histological profile triggered by gluten ingestion, which is given in genetically predisposed subjects. Heterogeneous clinical presentation is characteristic in CD, affecting any organ or tissue with gastrointestinal, extraintestinal, seronegative, or nonresponsive manifestations. CD diagnosis is based on several criteria, including genetic and serological tests, clinical symptoms and/or risk conditions, and duodenal biopsy. Currently, the available treatment for CD is a strict gluten-free diet (GFD) that essentially relies on the consumption of naturally gluten-free foods, such as animal-based products, fruits, vegetables, legumes, and nuts, as well as gluten-free dietary products that may not contain more than 20 mg of gluten per kg of food according to Codex Alimentarius. However, it is difficult to maintain a strict oral diet for life and at least one-third of patients with CD are exposed to gluten. Difficulties adhering to a GFD have led to new tools to monitor the correct adherence to GFD and alternative forms of treatment.

Keywords

  • celiac disease
  • gluten-free diet
  • gluten immunogenic peptides
  • dietary adherence
  • non-dietary therapies

1. Introduction

Celiac disease (CD) is a chronic immune-mediated enteropathy triggered by exposure to dietary gluten in genetically predisposed individuals [1]. The diagnosis rate of this pathology has increased in the last 10 years [2], so worldwide epidemiologic data are now available showing that CD is ubiquitous, with a prevalence of 1.4% [3], higher in female than male individuals [2, 3, 4, 5, 6, 7].

Clinically, CD presents with a wide variety of gastrointestinal and extraintestinal symptoms that differ considerably according to the age of presentation [8] or even be an asymptomatic disease. Digestive symptoms and growth retardation are frequent in the pediatric population diagnosed within the first years of life [9]. However, in adults, symptoms can be nonspecific gastrointestinal or extraintestinal of various kinds.

Currently, the only available treatment for CD is a strict, lifelong gluten-free diet (GFD), which requires significant patient education, motivation, and follow-up [10]. Adherence to a GFD is not easy, with the ubiquitous nature of gluten, cross-contamination of foods, inadequate food-labeling regulations, and social constraints [11]. Current methods to evaluate adherence to a GFD include the use of a dietary questionnaire and monitoring of serological findings or clinical symptoms; however, neither of these methods generates a direct nor an accurate measurement of dietary adherence [1, 11, 12]. Small bowel biopsy is the “gold standard” for CD diagnosis, but according to most clinical guidelines, its role in the follow-up of patients with CD is limited to cases involving a lack of clinical response or symptom recurrence [13, 14, 15, 16, 17]. Nonresponsive CD occurs frequently, particularly in those diagnosed in adulthood. Persistent or recurring symptoms should lead to a review of patients to exclude alternative diagnoses and a review of GFD to ensure there is no obvious gluten contamination and confirm adherence to GFD [1]. Possible causes include age at diagnosis, follow-up time, the existence of social differences, the intake of certain drugs (PPIs, NSAIDs), severe clinical symptoms at diagnosis, inadequate adherence to diet, or the presence of inadvertent contamination of the diet [18, 19].

In this chapter, we synthesized the latest research findings and evidence related to the management of CD and GFD, including emerging tools to monitor the correct adherence to GFD and the development of non-dietary therapies.

Advertisement

2. Pathogeny

Pathogeny development of CD is due to a combination of environmental (gluten and other factors), genetic (HLA system), and immunological factors (response of intestinal T lymphocytes).

2.1 Gluten

The major environmental factor responsible for the development of CD is gluten, which is a complex mixture of prolamin and glutelin storage proteins of certain cereals, such as wheat, barley, rye, oats, and their derivates. These common dietary proteins have unusual biochemical properties that include a high abundance of glutamine and proline residues, which render them resistant to degradation by gastrointestinal proteases [20], leaving large peptides. These peptides enter the lamina propria of the small intestine via transcellular or paracellular routes where, in affected individuals, an immune reaction occurs.

2.2 Immunological factors

The most accepted model for explaining CD immunopathogenesis is the two-signal model mediated by a first innate immune response (direct toxic effect of gluten on the epithelium) followed by a secondary antigen-specific adaptive response (through CD4+ T lymphocytes of the lamina propria) [20, 21]. Some peptides, such as, the 19-mer gliadin peptide, trigger an innate immune response mainly characterized by the production of IL-15 by epithelial cells. Result is the disruption of the epithelial barrier, by increasing the permeability and inducing enterocyte apoptosis [20]. These peptides enter the lamina propria of the small intestine via transcellular or paracellular routes [20] where, in affected individuals, an adaptive immune reaction occurs that is facilitated by increased intestinal permeability that allows the passage of immunogenic peptides, such as 33-mer, to the lamina propria. At the same time, some glutamine residues of these peptides are catalytically deaminated by tissue transglutaminase (tTG). This deamination, in turn, increases the immunogenicity of peptides due to high-affinity interactions between modified residues and ligand binding sites of HLA-DQ2 and HLA-DQ8 molecules [22] expressed by dendritic cells. Gliadin peptides are then presented to gliadin-reactive CD4+ T cells. During this process, antibodies against tTG, gliadin, and actin are made through unclear mechanisms. These antibodies might contribute to extra-intestinal manifestations of CD, such as dermatitis herpetiformis and gluten ataxia. Moreover, the immune response initiates a cascade of reactions that degenerate into crypt hyperplasia and flattening of the intestinal villi.

2.3 Genetic factors

The importance of a genetic component for the development of CD is evident, based on the familial occurrence and the high concordance among identical twins [2324]. Almost 100% of patients with CD possess specific variants of the HLA class II genes HLA-DQA1 and HLA-DQB1 that, together, encode the two chains (α and β) of CD-associated heterodimer proteins DQ2 and DQ8 that are expressed on the surface of antigen-presenting cells [25]. More than 90% of patients with CD are DQ2 positive and most of the others are DQ8 positive [26]. HLA-DQ2 and HLA-DQ8 risk heterodimers are present in approximately 30–40% of the general population, and of these, approximately 1% develop the disease, so HLA DQ2/8 seems necessary, but not sufficient for the development of CD [27].

Several studies have been carried out to identify non-HLA susceptibility genes. Among these are a large number of CD-associated genes basically encode interleukins, interleukin receptors, and tumor necrosis factors or receptors that are involved in innate immunity and epithelial stress signals (COELIAC2, COELIAC 3, CTLA4, and COELIAC4) [28].

2.4 Other environmental factors

Other environmental factors that could contribute to the development of CD have also been studied, such as the time and manner of introduction of gluten, the type of delivery, the start and duration of breastfeeding, the microbiome or early exposure to antibiotics, among others [29, 30]. However, the studies carried out to date do not confirm the different hypotheses proposed. Recently, the link between viral infections and loss of oral gluten tolerance has been investigated, since infections caused by rotavirus, reovirus, astrovirus, enterovirus, and adenovirus are very common in childhood. This opens the door to a new field of knowledge that could allow the design of preventive strategies in the future of CD [31, 32].

Advertisement

3. Clinical manifestations

Clinical characteristics of CD differ considerably depending on the age of presentation, and it can also be profuse or simply present analytical abnormalities [33, 34, 35, 36]. It can manifest clinically with a wide variety of symptoms that affect multiple organs and systems and that can be both gastrointestinal (diarrhea, vomiting, abdominal pain, bloating, bloating, constipation, gastroesophageal reflux, among others) and extra-intestinal (tiredness, dermatitis herpetiformis, anemia, osteoporosis, infertility, growth retardation, neuropathy, ataxia, delayed puberty, etc.) [8, 25]. Symptomatic CD can be classified into classic and non-classic. Any case presenting with malabsorption is classified as a classic CD. Although the clinical presentation is changing toward an affectation of older individuals with milder symptoms. The symptomatic classical disease was previously the most common presentation, and although it remains a prominent mode of presentation, subclinical and nonclassical cases now make up roughly 30% and 40–60% of new cases, respectively [37, 38].

Advertisement

4. Diagnosis

The diagnosis of CD may require genetic and serological tests and a duodenal biopsy.

4.1 Genetic risk markers

The main genetic risk factor for CD is the presence of HLA-DQ2 and -DQ8 heterodimers, which are identified in 90% and 5–7% of patients with CD, respectively [7]. Since these alleles are found in 30–40% of the general population (HLA-DQ2 being the most common) [39], their absence is important due to their negative predictive value (NPV). Therefore, the HLA-DQ2/HLA-DQ8 test plays an important role in CD diagnosis and is recommended in the following situations [40]—(a) exclusion of the disease, especially in patients who have started GFD; (b) in situations of uncertain diagnosis due to negative serology, but histology suggestive of CD; (c) to differentiate siblings in whom it is intended to ensure that it is unlikely that they will develop the disease from those who will need monitoring; (d) in subjects with autoimmune diseases and other diseases in which CD should be investigated.

A negative result for HLA-DQ2/HLA-DQ8 means a very low probability of developing the disease. Therefore, this test can be used to support the diagnosis of CD, since it has a high NPV, allowing exclusion with 99% certainty [41]. However, it has little positive predictive value (PPV) (only around 12%), so its determination has no diagnostic value in situations with elevated antibodies directed against tTG and should be reserved as second-line in patients with diagnostic doubt [42, 43].

4.2 Specific serum antibodies

Various serological tests have been developed to detect CD—antigliadin antibodies (anti-AGA), antibodies against deaminated gliadin peptides (anti-DGP), anti-endomysia antibodies (anti-EMA), and anti-transglutaminase antibodies (anti-tTG). Serological tests are important for two reasons—(1) they select patients in whom duodenal biopsy should be indicated to confirm clinical suspicion, and (2) they confirm the diagnosis in cases in which enteropathy has been observed [43].

Anti-AGA has been used for decades and is reasonably safe when the probability of suffering from CD is very high. However, it has been shown that these antibodies present variability in their diagnostic precision, due to the fact that they have low sensitivity and specificity; therefore, they should not be included in routine tests for the diagnosis of CD [41, 44].

Anti-EMA has a relatively low sensitivity (80–90%), but its specificity is close to 100%. However, they require more complex laboratory techniques and depend on the experience of the laboratory staff, remaining as a second-line test adequate to confirm clinical suspicion [1].

Anti-tTG IgA has a sensitivity and specificity of 95 and 90%, respectively [41, 45]. Anti-DGP has shown good precision, although lower than anti-tTG IgA, so an isolated positive result for IgA and/or IgG-DGP in patients at low risk for CD, predicts the disease only in 15%, being in the rest of the cases false positives. Therefore, in a first approximation, anti-tTG are the preferred antibodies for the diagnosis of CD according to the ESPGHAN diagnostic criteria [46, 47]. Anti-DGP is considered less sensitive or specific for the detection of CD compared to anti-tTG and anti-EMA. However, these last two antibodies are less sensitive in children under 2 years of age. It should also be taken into account that anti-tTG can be negative in 5–16% of patients with histologically confirmed CD [48]. Therefore, there is no serological test with perfect sensitivity and specificity [44]. In case of general IgA deficiency, which is observed in 2–3% of patients with CD, the IgG-based test (anti-DGP IgG and anti-tTG IgG) should be performed. IgG anti-tTG has diagnostic utility in patients with selective IgA deficiency (IgA < 0.07 mg/dl). Regarding anti-DGP IgG, there is no evidence of greater efficacy compared to anti-tTG IgG or anti-EMA IgG [41].

4.3 Intestinal biopsy

Duodenal biopsy of the small intestine is a key point in the diagnosis of CD. A distinctive pattern of histological abnormalities has been identified in this disease, including partial or total villous atrophy, elongated crypts, decreased villus/crypt ratio, increased crypt mitotic index, increased crypt density of intraepithelial lymphocytes (IELs), and infiltration of plasma cells in the lamina propria. An increase in IELs tends to be located at the tips of the villi and are usually CD8+ [37]. The presence of a diffuse and uniform infiltrate of these lymphocytes is the most sensitive finding, but it is not specific to CD. A count of at least 25 IELs/100 enterocytes represents a definitive increase in IELs [49, 50]. Immunohistochemical studies have shown that the increase in IELs represents an expansion of cytotoxic T cells alpha-beta and gamma-delta. Gamma-delta T cells are observed in 1–10% of the normal small intestinal mucosa but increase in patients with CD, where they may represent 15–30% of all IELs [1]. In addition, the absence of the brush border can be identified, as well as alterations in epithelial cells.

There are three grading systems to establish the severity of histological damage proposed by Marsh, Oberhuber [51], and Corazza-Villanaci [52]. Marsh system, with three types of grades, was replaced in 1999 by Oberhuber [51], which proposes a better standardization with six types [51]. In 2007, a new, simpler classification was published by Corazza-Villanacci [52]. These classifications are qualitative and subjective [1, 37]. Marsh-Oberhuber classification is used by most pathologists both for diagnosis and to ensure regression of the lesion after GFD [1]. Generally, six stages are distinguished—type 0 without lesion, type 1 (infiltrative lesion), type 2 (crypt hyperplasia), type 3 (villi atrophy: 3a: partial; 3b: subtotal; 3c: total) [51]. Furthermore, these lesions are not pathognomonic for CD, and there is a wide spectrum of diseases that can produce indistinguishable microscopic lesions.

Currently, it is considered that, in patients with high levels of antibodies, the diagnosis could be based on the combination of symptoms, antibodies determination, and genetics, omitting in this case the duodenal biopsy [11, 46], unlike what was established in the previous ESPGHAN guidelines for the diagnosis of CD. However, confirmation of CD by biopsy is considered the gold standard in the diagnosis of CD in certain types of patients.

The biopsy can be used to diagnose and monitor, but CD is a burden for patients. Therefore, less invasive and objective biomarkers are required to assess the disease. In addition, in certain patients, a challenge with gluten is necessary to make a correct diagnosis of CD. Based on this, Leonard et al. [53] investigated the ability of different biomarkers to diagnose CD after provocation. These biomarkers could, complement or replace histology in the diagnosis of CD. These authors evaluated traditional diagnostic techniques, such as biopsy, antibodies, symptomatology, as well as different biomarkers to measure the response to two levels of gluten exposure, studying interleukin-2 (IL-2), the tetramer test, and the dot enzyme-linked immunosorbent assay (Enzyme-Linked ImmunoSpot Assay, ELISpot), among others. Results showed that the measurement of IL-2 in plasma might be the first and most sensitive marker for the evaluation of gluten exposure in patients with CD. This study provides a framework for the rational design and selection of biomarkers in future gluten challenge studies with the goal of incorporating them into clinical practice.

Advertisement

5. Treatment of celiac disease

5.1 Diet therapy: gluten-free diet

Only effective treatment available for CD consists of following a strict GFD, excluding gluten proteins from the diet from wheat, barley, rye, and oats, as well as hybrids of these cereals such as triticale and their derivatives (starch, flour, etc.) [14]. Nevertheless, such a diet is difficult to follow due to the unintended contamination of “gluten-free” products, improper labeling, social constraints, and ubiquity of gluten proteins in raw or cooked foods and pharmaceuticals. Thus, accidental gluten encounters are likely. Most patients with CD can safely tolerate approximately 10 mg of gluten cross-contamination daily. However, there is a tremendous degree of variability within this population, and some patients may have worsening histological changes with very low daily gluten exposure [1, 10].

Strict adherence to GFD leads to remission of gastrointestinal and extra-intestinal symptoms, normalization of serological tests, and recovery of the intestinal mucosa, in most cases [14]. Initiation of strict GFD generally results in a rapid improvement of clinical symptoms, while recovery of the intestinal villi requires several years of a strict GFD (around 2 years in 34% and 5 years in 66%) [44]. Therefore, it is essential that patient with CD is aware of adherence to GFD to avoid future complications.

5.1.1 Difficulties in following a gluten-free diet: transgressions

Although adherence to GFD is the cornerstone of the treatment of patients with CD, there are conditions that prevent it from being carried out and mean that a significant percentage of patients with CD do not adhere and commit voluntary or involuntary transgressions [10]. Among the conditions that can prevent the GFD monitoring, we highlight the high economic cost of gluten-free products, which are not accessible to a large number of people with CD. Another factor to highlight that can favor its involuntary intake is the ubiquity of gluten in a high percentage of manufactured products since many of the foods that are marketed contain gluten from wheat, barley, rye, or oats, including those that intervene only as a thickener or binder. In fact, several studies carried out to determine the gluten content in natural (unprocessed) gluten-free foods or in foods labeled gluten-free reveal relatively high contamination rates, present in 9–22% of the samples analyzed [54, 55, 56]. In addition, many products contain hidden gluten, mainly due to cross-contamination with other gluten-containing foods that are processed or stored in the same place. The risk that these foods pose for patients with CD makes rigorous control of gluten content convenient [57]. Therefore, accurate detection and quantification of gluten in food are essential [10]. The Codex Alimentarius [58] has established that a food classified as “gluten-free” should not exceed 20 mg of gluten per kg of food, that is, 20 parts per million (ppm). Currently, several methods are used for the detection and quantification of gluten in foods. Enzyme-Linked ImmunoSorbent Assays (ELISAs) are the most widely used methods, as they are sensitive, rapid, and relatively easy to perform. Most commercial ELISAs use monoclonal antibodies (moAbs) such as R5 and G12 [59, 60, 61, 62, 63, 64]. Other methods, such as the Polymerase Chain Reaction (PCR), developed mainly for research, are far from being able to replace ELISA, as they are not suitable for the detection of gluten in highly processed or hydrolyzed samples due to DNA degradation. Lastly, liquid chromatography/mass spectrometry methods require expensive equipment and expertise [65].

All the factors described above cause nonadherence to GFD among patients with CD. Recent studies have indicated that inadvertent gluten ingestion occurs more frequently than intentional ingestion, and gluten contamination in naturally gluten-free foods is likely to be one of the most important factors in inadvertent nonadherence [66]. Other investigations based on the study of intestinal biopsies of patients with CD on GFD for more than 2 years have suggested that transgressions are relatively frequent, detecting a lack of recovery of the intestinal villi in 36–55% of the population studied [67, 68, 69]. These inadvertent or intentional violations are the main reason for uncontrolled CD in adult patients with CD [70]. Likewise, there is a small percentage of patients with CD (approximately 0.3–10%) who do not respond to GFD and have persistent symptoms of malabsorption and intestinal villi atrophy, which is known as refractory CD (RCD) [7, 71, 72, 73, 74].

5.1.2 Gluten-free diet monitoring methods

The existence of a reliable method that makes it possible to verify whether or not patients with CD are following a GFD is undoubtedly useful not only in monitoring the patient to avoid long-term complications, but also when diagnosing RCD [16, 44]. Among the methods to monitor adherence to GFD is the determination of specific antibodies, dietary interviews, control of symptoms, biopsies, and the detection of gluten immunogenic peptides (GIP) in stool and urine (Table 1) [1, 41, 47, 75].

5.1.2.1 Serological tests

Anti-tTG and anti-DGP have been used frequently to assess CD follow-up [76]. Use of these serological tests has revealed that it takes several months for the specific serology of CD to return to normal values. A significant decrease in levels during the first year suggests adherence to the diet and, therefore, patients with CD whose serology tests do not improve should be reassessed regarding their exposure to gluten [16]. However, negative serological markers do not reflect strict adherence to a GFD and are a poor predictor of dietary transgressions [17, 43, 77]. Although serology shows high accuracy for the diagnosis of CD, these tests are not as useful in follow-up, since they do not correlate with histological findings or symptoms [78]. It is important to note that a negative serology in a patient with CD on GFD does not necessarily guarantee the recovery of the intestinal mucosa [14, 43]. In a recent meta-analysis, PPV of persistently positive determination of anti-tTG IgA was very low and showed a sensitivity of 38% in adults. NPV of serology in adult patients with CD on GFD for one year or more was higher, with a specificity of 80%. Therefore, the usefulness of serology in the follow-up of adult patients with CD is very limited [1].

5.1.2.2 Symptomatology

Among the most widely used methods to assess the presence of gastrointestinal symptoms in patients with CD is the Gastrointestinal Symptom Rating Scale (GSRS) questionnaire [79, 80]. This questionnaire serves to check symptoms and determine the improvement and evolution of CD. However, there are a large number of patients with CD who are asymptomatic or minimally symptomatic at the time of presentation and, in these cases, it would not be feasible to use the clinical response as an indicator of intestinal mucosal recovery and adherence to GFD [13, 70].

5.1.2.3 Intestinal biopsy

Histological lesion remains the gold standard test for the diagnosis of CD, recovery of the mucosa is the main marker of response to diet. The only method to verify this normalization of the duodenum is by performing an oral endoscopy with an intestinal biopsy, an aggressive and costly follow-up method. However, an intestinal biopsy is a method used clinically, especially in the evaluation of patients with persistent symptoms [81]. It seems advisable to perform a follow-up endoscopy in adults 1–2 years after starting GFD to ensure recovery of the mucosa [82]. In this way, it would be possible to differentiate patients who are at low risk and in whom follow-up periods can be extended, from those at high risk who may need special supervision to maintain adherence to GFD [83].

5.1.2.4 Dietary questionnaires and interviews

Adherence to GFD can be assessed through dietary interviews or questionnaires conducted by a specialist. Dietitian has an important role in providing practical advice on lifestyle and food choices [16]. Evaluation of adherence to the diet through dietary interviews has been suggested because of its low cost and because it is not invasive; however, they are difficult to standardize and are subjective.

Different questionnaires assess the frequency of food and self-reported adherence to GFD [84]. Some of the more specific questionnaires are—(a) Gluten Free Score by Biagi et al. [85], whose four items provide a score from 0 to IV and in which levels 0 and I indicate poor adherence to the diet and, (b) the Celiac Dietary Adherence Test (CDAT) developed by Leffler et al. [86], which is a brief questionnaire that allows a rapid and standardized evaluation. This last questionnaire comprises seven easy-to-apply questions with optimal psychometric characteristics that assess CD symptomatology, self-efficacy expectations, reasons for maintaining GFD, knowledge of the disease, associated risk behaviors, and the perceived degree of adherence.

Nevertheless, there is considerable controversy about the validity of dietary questionnaires in the assessment of GFD because some patients with CD do not record the actual gluten consumed intentionally in some cases. Therefore, the measurement of adherence to GFD through questionnaires appears to be subjective and imprecise and does not allow involuntary infractions to be identified [25, 84].

5.1.2.5 Detection of immunogenic gluten peptides in human samples

Recently, new noninvasive methodologies have been developed to monitor gluten exposure in patients with CD based on the detection and quantification of GIP in stool and urine samples [87, 88, 89, 90]. These immunological methodologies (ELISA and immunochromatographic strips) based on G12 and A1 moAbs are capable of detecting GIP, which are gluten fragments resistant to gastrointestinal digestion, and mainly responsible for the immune response of patients with CD [60, 61, 91, 92, 93, 94, 95]. These tools make it possible to monitor adherence to GFD and detect violations cases, helping to identify the origin of clinical symptoms and avoid complications derived from gluten intake (anemia, osteoporosis, increased risk of lymphoma, etc.). These techniques have represented a revolutionary worldwide advance in the clinical practice of CD and have been introduced in the new guidelines, both European and Spanish, for monitoring the GFD of patients with CD [141]. Numerous rigorous studies have evaluated the use of GIP determination in stool and/or urine to monitor adherence to GFD compared to other tools (Table 2). The studies included children and adults diagnosed with CD and healthy volunteers. Overall, these studies indicated that this novel technique was highly sensitive for the detection of GFD transgressions and therefore could facilitate the follow-up of patients with CD.

StrengthsWeak points
Serological tests
  • High accuracy for the diagnosis of CD

  • Late positives (6–24 months to normalize)

  • False positives and negatives, for follow-up, no correlation with biopsy and symptoms

  • I need a blood draw

Dietary questionnaires, symptomatology questionnaires, and dietary interviews
  • Non-invasive

  • Low cost

  • Forgetfulness, omissions

  • Falsified

  • Tedious

  • Non-objective

Intestinal biopsy
  • Gold standard test for the diagnosis of CD

  • Invasive

  • Expensive, consumes hospital resources

  • Uncomfortable for the patient

Detection of GIP in human samples
  • Simple and fast method

  • Non-invasive

  • Correlation with gluten intake

Other bookmarks (Calprotectin)
  • Simple and fast method

  • Non-invasive

  • Non-CD specific

Table 1.

A comparison of the strengths and weaknesses of the tools used to monitor GFD in patients with CD. CD, celiac disease; GFD, gluten-free diet; GIP, gluten immunogenic peptides.

PopulationStudy designReferences
StoolChildrenCase-control study[87]
Cohort study[96]
Prospective study[89]
Transversal study[97]
Systematic revision[98]
Prospective study[99]
Observational descriptive study[100]
Children and adultsProspective study[88]
Transversal study[101, 102]
AdultsObservational prospective study[103]
Prospective study[104]
Prospective study[105]
UrineChildren and adultsControlled study[106]
Randomized controlled study[90]
AdultsTransversal study[107]
Prospective study[108]
Prospective study[109]
Prospective study[110]
Stool and urineChildren and adultsMeta-analysis[111]
AdultsProspective study[80]
Prospective study[17, 77]
Prospective study[112]

Table 2.

Studies based on GIP determination in stool and/or urine for monitoring of gluten-free diet. GIP, gluten immunogenic peptides.

5.1.2.6 Other bookmarks

Other markers have been proposed for monitoring GFD, such as the permeability test [113] or fecal calprotectin [114, 115]. Determination of fecal calprotectin concentration has established itself in recent years as a new useful marker of gastrointestinal pathologies. Several studies show that there is an association between calprotectin levels and the degree of inflammation, so it can be used to monitor response to treatment and predict the risk of recurrence. In addition, results obtained by Oribe et al. [116] have shown that patients with positive anti-tTG IgA antibodies, that is, those in contact with gluten, showed significantly higher values of fecal calprotectin than patients undergoing GFD and non-celiac patients. These methods, by demonstrating the presence of intestinal inflammatory processes, are generally not specific for CD and, therefore, if their values are modified, it could also be due to other causes such as infectious diseases, inflammatory bowel disease (IBD), or allergic processes.

5.2 Non-dietary therapies

Since strict follow-up of GFD presents many difficulties for patients with CD, additional treatments are needed for this disease. In recent years, CD research has focused on the search for non-dietary therapies to control GFD [17, 77]. Emerging therapeutic options for CD can be broadly classified into one of the following strategies—(1) removal of toxic gluten peptides before reaching the intestinal tract, (2) regulation of the immunostimulatory effects of toxic gluten peptides, (3) modulation of intestinal permeability, (4) immune modulation and induction of gluten tolerance, and (5) restoration of imbalance in the intestinal microbiota (Figure 1).

Figure 1.

Clinical and preclinical trials in the development of new non-dietary therapies in CD. CD: celiac disease; PEP: prolyl endopeptidases; TNF: tumor necrosis factor; and tTG: tissue transglutaminase [128].

To date, only larazotide acetate is in phase III studies. Larazotide is an oral peptide that modulates tight junctions and prevents the passage of gluten peptides to the lamina propria by closing the intercellular junctions of enterocytes. Therefore, it could help prevent the development of the immune cascade in patients with CD, showing a reduction in symptoms as well as a reduction in anti-tTG antibody levels. In addition, some very promising therapies are PRV-015 immunotherapy, the use of oral glutenases, as well as vaccine therapies (phase II). There are many other exciting drugs that are in the early stages of research, such as tTG inhibitors, HLA blockers, and probiotics [20, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128]. Similarly, some therapies are being evaluated in preclinical trials and are postulated as promising treatments for the pathogenesis of CD (Figure 1). Thus, we are faced with many promising and emerging options for the treatment of CD.

Advertisement

6. Conclusions

Research on CD is changing rapidly due to a steady increase in knowledge that addresses its pathophysiology, diagnosis, follow-up, and therapeutic options. Diagnosis of CD is based on several criteria, including positive serology, a spectrum of duodenal damage, clinical symptoms and/or risk conditions, and response to a GFD in susceptible individuals. In the absence of some of these criteria, the diagnosis of CD becomes challenging. In this regard, studies based on gluten reintroduction combined with IL-2 measurements could provide a new clinical alternative to diagnose and monitor patients who already have a GFD.

Several patients have difficulty controlling their diet they regularly consume sufficient gluten to trigger symptoms. Despite the availability of diverse traditional GFD adherence markers, such as diet tests or serology, none of them is an accurate diet evaluation method. Thus, use of GIP detection in stool and/or urine has been developed as a direct and specific test for GFD monitoring. Furthermore, non-dietary therapies have shown encouraging preliminary results in phase II and III clinical trials, such as larazotide acetate, PRV-015, IMGX-003, vaccine, and drug therapy. However, a GFD is the mainstay of CD therapy for the immediate future. For all these reasons, a health-oriented lifestyle should be promoted for better management and control of CD, responding to the growing demand of society and the empowerment of patients with CD.

Advertisement

Acknowledgments

This work was supported by grants from Knowledge Transfer Activities and Fundación Progreso y Salud, Consejería de Salud, Junta de Andalucía (project PI-0053-2018).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Appendices and nomenclature

Anti-AGA

antigliadin antibodies

Anti-DGP

antibodies deaminated gliadin peptides

Anti-EMA

anti-endomysia antibodies

Anti-tTG

anti-transglutaminase antibodies

CD

celiac disease

CDAT

celiac dietary adherence test

ELISA

enzyme-linked immunosorbent assays

GFD

gluten-free diet

GIP

gluten immunogenic peptides

IBD

inflammatory bowel disease

IELs

intraepithelial lymphocytes

IL-2

interleukin-2

NPV

negative predictive value

PPV

positive predictive value

PCR

polymerase chain reaction

RCD

refractory celiac disease

tTG

tissue transglutaminase

References

  1. 1. Al-Toma A, Volta U, Auricchio R, Castillejo G, Sanders DS, Cellier C, 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(5):583-613. DOI: 10.1177/2050640619844125
  2. 2. King JA, Jeong J, Underwood FE, Quan J, Panaccione N, Windsor JW, et al. Incidence of celiac disease is increasing over time: A systematic review and meta-analysis. American Journal of Gastroenterology. 2020;115(4):507-525
  3. 3. Singh P, Arora A, Strand TA, Leffler DA, Catassi C, Green PH, et al. Global prevalence of celiac disease: Systematic review and meta-analysis. Clinical Gastroenterology Hepatology. 2018;16(6):823-836
  4. 4. Mardini HE, Westgate P, Grigorian AY. Racial differences in the prevalence of celiac disease in the US Population: National Health and Nutrition Examination Survey (NHANES) 2009-2012. Digestive Diseases and Sciences. 2015;60(6):1738-1742. DOI: 10.1007/s10620-014-3514-7
  5. 5. Lebwohl B, Sanders DS, Green PHR. Coeliac disease. Lancet. 2018;391(10115):70-81. DOI: 10.1016/S0140-6736(17)31796-8
  6. 6. Glissen JRB, Singh P. Coeliac disease. Paediatric International Child Health. 2019;39:23-31. DOI: 10.1080/20469047.2018.1504431
  7. 7. Ludvigsson JF, Murray JA. Epidemiology of celiac disease. Gastroenterology Clinics of North America. 2019;48(1):1-18. DOI: 10.1016/j.gtc.2018.09.004
  8. 8. Therrien A, Kelly CP, Silvester JA. Celiac disease: Extraintestinal manifestations and associated conditions. Journal of Clinical Gastroenterology. 2020;54(1):8-21. DOI: 10.1097/MCG.0000000000001267
  9. 9. Sahin Y. Celiac disease in children: A review of the literature. World Journal of Clinical Pediatrics. 2021;10(4):53-71. DOI: 10.5409/wjcp.v10.i4.53
  10. 10. Itzlinger A, Branchi F, Elli L, Schumann M. Gluten-free diet in celiac disease-forever and for all? Nutrients. 2018;10(11):1796. DOI: 10.3390/nu10111796
  11. 11. Wolf J, Petroff D, Richter T, Auth MKH, Uhlig HH, Laass MW, et al. Validation of antibody-based strategies for diagnosis of pediatric celiac disease without biopsy. Gastroenterology. 2017;153(2):410-419
  12. 12. Wieser H, Ruiz-Carnicer Á, Segura V, Comino I, Sousa C. Challenges of monitoring the gluten-free diet adherence in the management and follow-up of patients with celiac disease. Nutrients. 2021;13(7):2274. DOI: 10.3390/nu13072274
  13. 13. Sharkey LM, Corbett G, Currie E, Lee J, Sweeney N, Woodward JM. Optimising delivery of care in coeliac disease—Comparison of the benefits of repeat biopsy and serological follow-up. Alimentary Pharmacology Theraphy. 2013;38(10):1278-1291. DOI: 10.1111/apt.12510
  14. 14. Ludvigsson JF, Bai JC, Biagi F, Card TR, Ciacci C, Ciclitira PJ, et al. BSG Coeliac Disease Guidelines Development Group, British Society of Gastroenterology. Diagnosis and management of adult coeliac disease: Guidelines from the British Society of Gastroenterology. Gut. 2014;63(8):1210-1228. DOI: 10.1136/gutjnl-2013-306578
  15. 15. Silvester JA, Kurada S, Szwajcer A, Kelly CP, Leffler DA, Duerksen DR. Tests for serum transglutaminase and endomysial antibodies do not detect most patients with celiac disease and persistent villous atrophy on gluten-free diets: A meta-analysis. Gastroenterology. 2017;153(3):689-701
  16. 16. Husby S, Bai JC. Follow-up of celiac disease. Gastroenterology Clinics of North America. 2019;48(1):127-136. DOI: 10.1016/j.gtc.2018.09.009
  17. 17. Silvester JA, Comino I, Kelly CP, Sousa C, Duerksen DR, DOGGIE BAG Study Group. Most patients with celiac disease on gluten-free diets consume measurable amounts of gluten. Gastroenterology. 2020;158(5):1497-1499
  18. 18. Francavilla R, Cristofori F, Stella M, Borrelli G, Naspi G, Castellaneta S. Treatment of celiac disease: From gluten-free diet to novel therapies. Minerva Pediatrica. 2014;66(5):501-516
  19. 19. Leonard MM, Cureton P, Fasano A. Indications and use of the gluten contamination elimination diet for patients with nonresponsive celiac disease. Nutrients. 2017;9(10):1129. DOI: 10.3390/nu9101129
  20. 20. Lindfors K, Ciacci C, Kurppa K, Lundin KEA, Makharia GK, Mearin ML, et al. Coeliac disease. Nature Reviews. 2019;5(1):3. DOI: 10.1038/s41572-018-0054-z
  21. 21. Sharma N, Bhatia S, Chunduri V, Kaur S, Sharma S, Kapoor P, et al. Pathogenesis of celiac disease and other gluten related disorders in wheat and strategies for mitigating them. Frontiers in Nutrition. 2020;7:6. DOI: 10.3389/fnut.2020.00006
  22. 22. Sollid LM, Khosla C. Novel therapies for coeliac disease. Journal of Internal Medicine. 2011;269(6):604-613
  23. 23. Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, et al. Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: A large multicenter study. Archives of Internal Medicine. 2003;163(3):286-292. DOI: 10.1001/archinte.163.3.286
  24. 24. Kuja-Halkola R, Lebwohl B, Halfvarson J, Wijmenga C, Magnusson PK, Ludvigsson JF. Heritability of non-HLA genetics in coeliac disease: A population-based study in 107 000 twins. Gut. 2016;65:1793-1798. DOI: 10.1136/gutjnl-2016-311713
  25. 25. Caio G, Volta U, Sapone A, Leffler DA, De Giorgio R, Catassi C, et al. Celiacdisease: A comprehensivecurrentreview. BMC Medicine. 2019;17(1):142. DOI: 10.1186/s12916-019-1380-z
  26. 26. Megiorni F, Mora B, Bonamico M, Barbato M, Nenna R, Maiella G, et al. HLA-DQ and risk gradient for celiac disease. Human Immunology. 2009;70(1):55-59. DOI: 10.1016/j.humimm.2008.10.018
  27. 27. Bourgey M, Calcagno G, Tinto N, Gennarelli D, Margaritte-Jeannin P, Greco L, et al. HLA related genetic risk for coeliac disease. Gut. 2007;56(8):1054-1059. DOI: 10.1136/gut.2006.108530
  28. 28. Piscaglia AC. Intestinal stem cells and celiac disease. World Journal of Stem Cells. 2014;6(2):213-229. DOI: 10.4252/wjsc.v6.i2.213
  29. 29. Whyte LA, Kotecha S, Watkins WJ, Jenkins HR. Coeliac disease is more common in children with high socio-economic status. Acta Paediatrics. 2014;103(3):289-294. DOI: 10.1111/apa.12494
  30. 30. Dydensborg Sander S, Nybo Andersen AM, Murray JA, Karlstad Ø, Husby S, Størdal K. Association between antibiotics in the first year of life and celiac disease. Gastroenterology. 2019;156(8):2217-2229. DOI: 10.1053/j.gastro.2019.02.039
  31. 31. Gómez-Rial J, Rivero-Calle I, Salas A, Martinón-Torres F. Rotavirus and autoimmunity. The Journal of Infection. 2020;81(2):183-189. DOI: 10.1016/j.jinf.2020.04.041
  32. 32. Sánchez D, Hoffmanová I, Szczepanková A, Hábová V, Tlaskalová-Hogenová H. Contribution of infectious agents to the development of celiac disease. Microorganisms. 2021;9(3):547. DOI: 10.3390/microorganisms9030547
  33. 33. Jericho H, Sansotta N, Guandalini S. Extraintestinal manifestations of celiac disease: Effectiveness of the gluten-free diet. Journal of Pediatric and Gastroenterology Nutrition. 2017;65(1):75-79. DOI: 10.1097/MPG.0000000000001420
  34. 34. Jericho H, Guandalini S. Extra-intestinal manifestation of celiac disease in children. Nutrients. 2018;10(6):755. DOI: 10.3390/nu10060755
  35. 35. Mearin ML, Montoro-Huguet M, Polanco I, Ribes-Koninckx C, Santolaria S. Manifestaciones clínicas de la enfermedad celíaca y criterios Diagnósticos: Diferencias entre niños, adolescentes y adultos. In: Arranz E, Fernández-Bañares F, Rosell CM, Rodrigo L, Peña AS, editors. Avances en el conocimiento de las patologías relacionadas con el gluten y evolución de los alimentos sin gluten. 2018. pp. 259-303
  36. 36. Sansotta N, Amirikian K, Guandalini S, Jericho H. Celiac disease symptom resolution: Effectiveness of the gluten-free diet. Journal of Pediatric and Gastroenterology Nutrition. 2018;66(1):48-52. DOI: 10.1097/MPG.0000000000001634
  37. 37. Hujoel IA, Reilly NR, Rubio-Tapia A. Celiac disease: Clinical features and diagnosis. Gastroenterology Clinics of North America. 2019;48(1):19-37. DOI: 10.1016/j.gtc.2018.09.001
  38. 38. Lebwohl B, Rubio-Tapia A. Epidemiology, presentation, and diagnosis of celiac disease. Gastroenterology. 2021;160(1):63-75. DOI: 10.1053/j.gastro.2020.06.098
  39. 39. Sallese M, Lopetuso LR, Efthymakis K, Neri M. Beyond the HLA genes in gluten-related disorders. Frontiers in Nutrition. 2020;7:575844. DOI: 10.3389/fnut.2020.575844
  40. 40. Liu E, Lee HS, Aronsson CA, Hagopian WA, Koletzko S, Rewers MJ, et al. Risk of pediatric celiac disease according to HLA haplotype and country. The New England Journal of Medicine. 2014;371(1):42-49. DOI: 10.1056/NEJMoa1313977
  41. 41. Grupo de trabajo del Protocolo para el diagnóstico precoz de la enfermedad celíaca. Protocolo para el diagnóstico precoz de la enfermedad celíaca. Ministerio de Sanidad, Servicios Sociales e Igualdad. Servicio de Evaluación del Servicio Canario de la Salud (SESCS). 2018
  42. 42. Brown NK, Guandalini S, Semrad C, Kupfer SS. A clinician’s guide to celiac disease HLA genetics. American Joural of Gastroenterology. 2019;114(10):1587-1592
  43. 43. Husby S, Murray JA, Katzka DA. AGA clinical practice update on diagnosis and monitoring of celiac disease-changing utility of serology and histologic measures: Expert review. Gastroenterology. 2019;156(4):885-889. DOI: 10.1053/j.gastro.2018.12.010
  44. 44. Rubio-Tapia A, Hill ID, Kelly CP, Calderwood AH, Murray JA, American College of Gastroenterology. ACG clinical guidelines: Diagnosis and management of celiac disease. The American Journal of Gastroenterology. 2013;108(5):656-676
  45. 45. Lewis NR, Scott BB. Meta-analysis: Deamidated gliadin peptide antibody and tissue transglutaminase antibody compared as screening tests for coeliac disease. Alimentary Pharmacology Theraphy. 2010;31(1):73-81. DOI: 10.1111/j.1365-2036.2009.04110.x
  46. 46. Husby S, Koletzko S, Korponay-Szabó IR, Mearin ML, Phillips A, Shamir R, et al. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of coeliac disease. Journal of Pediatric and Gastroenterology Nutrition. 2012;54(1):136-160
  47. 47. Husby S, Koletzko S, Korponay-Szabó I, Kurppa K, Mearin ML, Ribes-Koninckx C, et al. European Society Paediatric Gastroenterology, Hepatology and Nutrition Guidelines for Diagnosing Coeliac Disease 2020. Journal of Pediatric and Gastroenterology Nutritio. 2020;70(1):141-156. DOI: 10.1097/MPG.0000000000002497
  48. 48. De Leo L, Bramuzzo M, Ziberna F, Villanacci V, Martelossi S, Leo GD, et al. Diagnostic accuracy and applicability of intestinal auto-antibodies in the wide clinical spectrum of coeliac disease. eBio Medicine. 2020;51:102567. DOI: 10.1016/j.ebiom.2019.11.028
  49. 49. Eiras P, Camarero C, León F, Roldán E, Asensio A, Baragaño M, et al. Linfocitos intraepiteliales en la enfermedad celíaca [Intraepitheliallymphocytes in celiacdisease]. Anales Españoles de Pediatría. 2002;56(3):224-232
  50. 50. Olivencia P, Cano A, Martín MA, León F, Roy G, Redondo C. Enfermedad celíaca del adulto y linfocitos intraepiteliales. ¿Nuevas opciones para el diagnóstico? [Adultceliacdisease and intraepitheliallymphocytes. New options for diagnosis?]. Gastroenterología y Hepatología. 2008;31(9):555-559. DOI: 10.1157/13128293
  51. 51. 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. DOI: 10.1097/00042737-199910000-00019
  52. 52. Corazza GR, Villanacci V, Zambelli C, Milione M, Luinetti O, Vindigni C, et al. Comparison of the interobserver reproducibility with different histologic criteria used in celiac disease. Clinical Gastroenterology and Hepatology. 2007;5(7):838-843. DOI: 10.1016/j.cgh.2007.03.019
  53. 53. Leonard MM, Silvester JA, Leffler D, Fasano A, Kelly CP, Lewis SK, et al. Evaluating responses to gluten challenge: A randomized, double-blind, 2-dose gluten challenge trial. Gastroenterology. 2021;160(3):720-733
  54. 54. Bustamante MÁ, Fernández-Gil MP, Churruca I, Miranda J, Lasa A, Navarro V, et al. Evolution of gluten content in cereal-based gluten-free products: An overview from 1998 to 2016. Nutrients. 2017;9(1):21. DOI: 10.3390/nu9010021
  55. 55. Farage P, de MedeirosNóbrega YK, Pratesi R, Gandolfi L, Assunção P, Zandonadi RP. Gluten contamination in gluten-free bakery products: A risk for coeliac disease patients. Public Health Nutrition. 2017;20(3):413-416. DOI: 10.1017/S1368980016002433
  56. 56. Raju N, Joshi AKR, Vahini R, Deepika T, Bhaskarachari K, Devindra S. Gluten contamination in labelled and naturally gluten-free grain products in southern India. Food Additive Contamination. 2020;37(4):531-538. DOI: 10.1080/19440049.2020.1711970
  57. 57. Mena MC, Sousa C. Analytical tools for gluten detection. Policies and regulation. In: Arranz E, Fernández-Bañares F, editors. Advances in the Understanding of Gluten Related Pathology and the Evolution of Gluten-free Foods. 2015. pp. 527-564
  58. 58. Codex Standard 118-1979. Codex standard for foods for special dietary use for persons intolerant to gluten. Codex Alimentarius Commission; revision 2008, amendment; 2015
  59. 59. Valdés I, García E, Llorente M, Méndez E. Innovative approach to low-level gluten determination in foods using a novel sandwich enzyme-linked immunosorbent assay protocol. European Journal of Gastroenterology & Hepatology. 2003;15(5):465-474. DOI: 10.1097/01.meg.0000059119.41030.df
  60. 60. Morón B, Cebolla A, Manyani H, Alvarez-Maqueda M, Megías M, Thomas Mdel C, et al. Sensitive detection of cereal fractions that are toxic to celiac disease patients by using monoclonal antibodies to a main immunogenic wheat peptide. American Journal of Clinical Nutrition. 2008;87(2):405-414. DOI: 10.1093/ajcn/87.2.405
  61. 61. Morón B, Bethune MT, Comino I, Manyani H, Ferragud M, López MC,et al. Toward the assessment of food toxicity for celiac patients: Characterization of monoclonal antibodies to a main immunogenic gluten peptide. PLoS One. 2008;3(5):e2294. DOI: 10.1371/journal.pone.0002294
  62. 62. Rosell CM, Barro F, Sousa C, Mena MC. Cereals for developing gluten-free products and analytical tools for gluten detection. Journal of Cereal Science. 2014;59(3):354-364. DOI: 10.1016/j.jcs.2013.10.001
  63. 63. Melini F, Melini V. Immunological methods in gluten risk analysis: A snapshot. Safety. 2018;4(4):56. DOI: 10.3390/safety4040056
  64. 64. Panda R, Garber EAE. Detection and quantitation of gluten in fermented-hydrolyzed foods by antibody-based methods: Challenges, progress, and a potential path forward. Frontiers in Nutrition. 2019;6:97. DOI: 10.3389/fnut.2019.00097
  65. 65. Scherf KA, Poms RE. Recent developments in analytical methods for tracing gluten. Journal of Cereal Science. 2016;67:112-122. DOI: 10.1016/j.jcs.2015.08.006
  66. 66. Muhammad H, Reeves S, Jeanes YM. Identifying and improving adherence to the gluten-free diet in people with coeliac disease. Proceedings of the Nutrition Society. 2019;78(3):418-425. DOI: 10.1017/S002966511800277X
  67. 67. Stoven S, Murray JA, Marietta E. Celiac disease: Advances in treatment via gluten modification. Clinical Gastroenterology and Hepatology. 2012;10(8):859-862. DOI: 10.1016/j.cgh.2012.06.005
  68. 68. Hall NJ, Rubin GP, Charnock A. Intentional and inadvertent non-adherence in adult coeliac disease. A cross-sectional survey. Appetite. 2013;68:56-62. DOI: 10.1016/j.appet.2013.04.016
  69. 69. Matoori S, Fuhrmann G, Leroux JC. Celiac disease: A challenging disease for pharmaceutical scientists. Pharmaceutical Research. 2013;30(3):619-626. DOI: 10.1007/s11095-012-0951-x
  70. 70. Mahadev S, Murray JA, Wu TT, Chandan VS, Torbenson MS, Kelly CP, et al. Factors associated with villus atrophy in symptomatic coeliac disease patients on a gluten-free diet. Alimentary Pharmacology Theraphy. 2017;45(8):1084-1093. DOI: 10.1111/apt.13988
  71. 71. Rubio-Tapia A, Murray JA. Classification and management of refractory coeliac disease. Gut. 2010;59(4):547-557. DOI: 10.1136/gut.2009.195131
  72. 72. Rishi AR, Rubio-Tapia A, Murray JA. Refractory celiac disease. Expert Review of Gastroenterology & Hepatology. 2016;10(4):537-546. DOI: 10.1586/17474124.2016.1124759
  73. 73. Malamut G, Cellier C. Refractory celiac disease. Gastroenterology Clinics of North America. 2019;48(1):137-144. DOI: 10.1016/j.gtc.2018.09.010
  74. 74. Penny HA, Baggus EMR, Rej A, Snowden JA, Sanders DS. Non-responsive coeliac disease: A comprehensive review from the NHS England National Centre for refractory coeliac disease. Nutrients. 2020;12(1):216. DOI: 10.3390/nu12010216
  75. 75. Rodrigo L, Pérez-Martinez I, Lauret-Braña E, Suárez-González A. Descriptive study of the different tools used to evaluate the adherence to a gluten-free diet in celiac disease patients. Nutrients. 2018;10(11):1777. DOI: 10.3390/nu10111777
  76. 76. Ludvigsson JF, Ciacci C, Green PH, Kaukinen K, Korponay-Szabo IR, Kurppa K, et al. Outcome measures in coeliac disease trials: The Tampere recommendations. Gut. 2018;67(8):1410-1424. DOI: 10.1136/gutjnl-2017-314853
  77. 77. Silvester JA, Comino I, Rigaux LN, Segura V, Green KH, Cebolla A, et al. Exposure sources, amounts and time course of gluten ingestion and excretion in patients with coeliac disease on a gluten-free diet. Alimentary Pharmacology Theraphy. 2020;52(9):1469-1479. DOI: 10.1111/apt.16075
  78. 78. Silvester JA, Kurada S, Szwajcer A, Kelly CP, Leffler DA, Duerksen DR. Tests for serum transglutaminase and endomysial antibodies do not detect most patients with celiac disease and persistent villous atrophy on gluten-free diets: A meta-analysis. Gastroenterology. 2017;153(3):689-701
  79. 79. Svedlund J, Sjödin I, Dotevall G. GSRS—A clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome and peptic ulcer disease. Digestive Diseases and Sciences. 1988;33(2):129-134. DOI: 10.1007/BF01535722
  80. 80. Costa AF, Sugai E, Temprano MP, Niveloni SI, Vázquez H, Moreno ML, et al. Gluten immunogenic peptide excretion detects dietary transgressions in treated celiac disease patients. World Journal of Gastroenterology. 2019;25(11):1409-1420. DOI: 10.3748/wjg.v25.i11.1409
  81. 81. Kelly CP, Bai JC, Liu E, Leffler DA. Advances in diagnosis and management of celiac disease. Gastroenterology. 2015;148(6):1175-1186. DOI: 10.1053/j.gastro.2015.01.044
  82. 82. Moscoso JF, Quera PR. Enfermedad celíaca. Revisión [Update on celiac disease]. The Child. 2016;144(2):211-221
  83. 83. Pekki H, Kurppa K, Mäki M, Huhtala H, Sievänen H, Laurila K, et al. Predictors and significance of incomplete mucosal recovery in celiac disease after 1 year on a gluten-free diet. The American Journal of Gastroenterology. 2015;110(7):1078-1085. DOI: 10.1038/ajg.2015.155
  84. 84. Silvester JA, Weiten D, Graff LA, Walker JR, Duerksen DR. Is it gluten-free? Relationship between self-reported gluten-free diet adherence and knowledge of gluten content of foods. Nutrition. 2016;32(7-8):777-783. DOI: 10.1016/j.nut.2016.01.021
  85. 85. Biagi F, Campanella J, Martucci S, Pezzimenti D, Ciclitira PJ, Ellis HJ, et al. A milligram of gluten a day keeps the mucosal recovery away: A case report. Nutrition Reviews. 2004;62(9):360-363. DOI: 10.1111/j.1753-4887.2004.tb00062.x
  86. 86. Leffler DA, Dennis M, Edwards George JB, Jamma S, Magge S, Cook EF, et al. A simple validated gluten-free diet adherence survey for adults with celiac disease. Clinical Gastroenterology Hepatol. 2009;7(5):530-536
  87. 87. Comino I, Real A, Vivas S, Síglez MÁ, Caminero A, Nistal E, et al. Monitoring of gluten-free diet compliance in celiac patients by assessment of gliadin 33-mer equivalent epitopes in feces. American Journal of Clinical Nutrition. 2012;95(3):670-677. DOI: 10.3945/ajcn.111.026708
  88. 88. Comino I, Fernández-Bañares F, Esteve M, Ortigosa L, Castillejo G, Fambuena B, et al. Fecal gluten peptidesreveallimitations of serologicaltests and foodquestionnairesformonitoring gluten-free diet in celiacdiseasepatients. American Journal of Gastroentrology. 2016;111(10):1456-1465
  89. 89. Comino I, Segura V, Ortigosa L, Espín B, Castillejo G, Garrote JA, et al. Prospective longitudinal study: Use of faecal gluten immunogenic peptides to monitor children diagnosed with coeliac disease during transition to a gluten-free diet. Alimentary Pharmacology Theraphy. 2019;49(12):1484-1492. DOI: 10.1111/apt.15277
  90. 90. Moreno ML, Cebolla Á, Muñoz-Suano A, Carrillo-Carrion C, Comino I, Pizarro Á, 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(2):250-257. DOI: 10.1136/gutjnl-2015-310148
  91. 91. Comino I, Real A, de Lorenzo L, Cornell H, López-Casado MÁ, Barro F, et al. Diversity in oat potential immunogenicity: Basis for the selection of oat varieties with no toxicity in coeliac disease. Gut. 2011;60(7):915-922. DOI: 10.1136/gut.2010.225268
  92. 92. Comino I, Real A, Gil-Humanes J, Pistón F, de Lorenzo L, Moreno Mde L, et al. Significant differences in coeliac immunotoxicity of barley varieties. Molecular Nutrition Food and Research. 2012;56(11):1697-1707. DOI: 10.1002/mnfr.201200358
  93. 93. Comino I, Real A, Moreno Mde L, Montes R, Cebolla A, Sousa C. Immunological determination of gliadin 33-mer equivalent peptides in beers as a specific and practical analytical method to assess safety for celiac patients. Journal of the Science of Food and Agriculture. 2013;93(4):933-943. DOI: 10.1002/jsfa.5830
  94. 94. Real A, Comino I, Moreno Mde L, López-Casado MÁ, Lorite P, Torres MI, et al. Identification and in vitro reactivity of celiac immunoactive peptides in an apparent gluten-free beer. PLoS One. 2014;9(6):e100917. DOI: 10.1371/journal.pone.0100917
  95. 95. Moreno Mde L, Muñoz-Suano A, López-Casado MÁ, Torres MI, Sousa C, Cebolla Á. Selective capture of most celiac immunogenic peptides from hydrolyzed gluten proteins. Food Chemistry. 2016;205:36-42. DOI: 10.1016/j.foodchem.2016.02.066
  96. 96. Gerasimidis K, Zafeiropoulou K, Mackinder M, Ijaz UZ, Duncan H, Buchanan E, et al. Comparison of clinical methods with the faecal gluten immunogenic peptide to assess gluten intake in coeliac disease. Journal of Pediatric and Gastroenterology Nutrition. 2018;67(3):356-360. DOI: 10.1097/MPG.0000000000002062
  97. 97. Roca M, Donat E, Masip E, Crespo Escobar P, Fornes-Ferrer V, Polo B, et al. Detection and quantification of gluten immunogenic peptides in feces of infants and their relationship with diet. Revista Espanol Enfermedades Digestivas. 2019;111(2):106-110
  98. 98. Myléus A, Reilly NR, Green PHR. Rate, risk factors, and outcomes of nonadherence in pediatric patients with celiac disease: A systematic review. Clinical Gastroenterology and Hepatology. 2020;18(3):562-573. DOI: 10.1016/j.cgh.2019.05.046
  99. 99. Roca M, Donat E, Masip E, Crespo-Escobar P, Cañada-Martínez AJ, Polo B, et al. Analysis of gluten immunogenic peptides in feces to assess adherence to the gluten-free diet in pediatric celiac patients. European Journal of Nutrition. 2021;60(4):2131-2140. DOI: 10.1007/s00394-020-02404-z
  100. 100. Fernández Miaja M, Díaz Martín JJ, Jiménez Treviño S, Suárez González M, BousoñoGarcía C. Study of adherence to the gluten-free diet in coeliac patients. Anales de Pediatría (Barcelona, Spain). 2021;94(6):377-384. DOI: 10.1016/j.anpedi.2020.06.017
  101. 101. Porcelli B, Ferretti F, Biviano I, Santini A, Cinci F, Vascotto M, et al. Testing for fecal gluten immunogenic peptides: A useful tool to evaluate compliance with gluten-free diet by celiacs. Annals of Gastroenterology. 2020;33(6):631-637
  102. 102. Porcelli B, Ferretti F, Cinci F, Biviano I, Santini A, Grande E, et al. Fecal gluten immunogenic peptides as indicators of dietary compliance in celiac patients. Minerva Gastroenterology Dietology. 2020;66(3):201-207
  103. 103. Laserna-Mendieta EJ, Casanova MJ, Arias Á, Arias-González L, Majano P, Mate LA, et al. Poor sensitivity of fecal gluten immunogenic peptides and serum antibodies to detect duodenal mucosal damage in celiac disease monitoring. Nutrients. 2020;13(1):98. DOI: 10.3390/nu13010098
  104. 104. Fernández-Bañares F, Beltrán B, Salas A, Comino I, Ballester-Clau R, Ferrer C, et al. Persistent villous atrophy in de novo adult patients with celiac disease and strict control of gluten-free diet adherence: A Multicenter Prospective Study (CADER Study). American Journal of Gastroenterology. 2021;116(5):1036-1043
  105. 105. Coto L, Sousa C, Cebolla A. Individual variability in patterns and dynamics of fecal gluten immunogenic peptides excretion after low gluten intake. European Journal of Nutrition. 2022;7:1-17. DOI: 10.1007/s00394-021-02765-z
  106. 106. Soler M, Estevez MC, Moreno Mde L, Cebolla A, Lechuga LM. Label-free SPR detection of gluten peptides in urine for non-invasive celiac disease follow-up. BiosensBioelectron. 2016;79:158-164. DOI: 10.1016/j.bios.2015.11.097
  107. 107. Peláez EC, Estevez MC, Domínguez R, Sousa C, Cebolla A, Lechuga LM. A compact SPR biosensor device for the rapid and efficient monitoring of gluten-free diet directly in human urine. Analytical and Bioanalytical Chemistry. 2020;412(24):6407-6417. DOI: 10.1007/s00216-020-02616-6
  108. 108. Ruiz-Carnicer Á, Garzón-Benavides M, Fombuena B, Segura V, García-Fernández F, Sobrino-Rodríguez S, 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. American Journal of Clinical Nutrition. 2020;112(5):1240-1251
  109. 109. Moreno ML, Sánchez-Muñoz D, Sanders D, Rodríguez-Herrera A, Sousa C. Verifying diagnosis of refractory celiac disease with urine gluten immunogenic peptides as biomarker. Frontier in Medicine (Lausanne). 2021;7:601854
  110. 110. Coto L, Sousa C, Cebolla A. Dynamics and considerations in the determination of the excretion of gluten immunogenic peptides in urine: Individual variability at low gluten intake. Nutrients. 2021;13(8):2624. DOI: 10.3390/nu13082624
  111. 111. Syage JA, Kelly CP, Dickason MA, Ramirez AC, Leon F, Dominguez R, et al. Determination of gluten consumption in celiac disease patients on a gluten-free diet. American Journal of Clinical Nutrition. 2018;107(2):201-207. DOI: 10.1093/ajcn/nqx049
  112. 112. Stefanolo JP, Tálamo M, Dodds S, de la Paz TM, Costa AF, Moreno ML, et al. Real-World gluten exposure in patients with celiac disease on gluten-free diets, determined from gliadin immunogenic peptides in urine and fecal samples. Clinical Gastroenterology and Hepatology. 2021;19(3):484-491
  113. 113. Duerksen DR, Wilhelm-Boyles C, Parry DM. Intestinal permeability in long-term follow-up of patients with celiac disease on a gluten-free diet. Digestive Diseases and Sciences. 2005;50(4):785-790. DOI: 10.1007/s10620-005-2574-0
  114. 114. Ertekin V, Selimoğlu MA, Turgut A, Bakan N. Fecal calprotectin concentration in celiac disease. Journal of Clinical Gastroenterology. 2010;44(8):544-546. DOI: 10.1097/MCG.0b013e3181cadbc0
  115. 115. Szaflarska-Popławska A, Romańczuk B, Parzęcka M. Faecal calprotectin concentration in children with coeliac disease. Przeglad Gastroenterology. 2020;15(1):44-47. DOI: 10.5114/pg.2020.93630
  116. 116. Oribe J, Castro GE, Montañez SG, Pérez PC, Ruiz BB. Niveles de calprotectina fecal en pacientes con anticuerpos anti-transglutaminasa positivos y pacientes con dieta libre de gluten. Acta Bioquím Clín Latinoam. 2020;54(1):29-38
  117. 117. Veeraraghavan G, Leffler DA, Kaswala DH, Mukherjee R. Celiac disease 2015 update: New therapies. Expert Review of Gastroenterology & Hepatology. 2015;9(7):913-927. DOI: 10.1586/17474124.2015.1033399
  118. 118. Khaleghi S, Ju JM, Lamba A, Murray JA. The potential utility of tight junction regulation in celiac disease: Focus on larazotide acetate. Therapeutic Advances in Gastroenterology. 2016;9(1):37-49. DOI: 10.1177/1756283X15616576
  119. 119. Daveson AJM, Ee HC, Andrews JM, King T, Goldstein KE, Dzuris JL, et al. Epitope-specific immunotherapy targeting cd4-positive t cells in celiac disease: Safety, pharmacokinetics, and effects on intestinal histology and plasma cytokines with escalating dose regimens of nexvax2 in a randomized, double-blind, placebo-controlled phase 1 study. eBioMedicine. 2017;26:78-90. DOI: 10.1016/j.ebiom.2017.11.018
  120. 120. Syage JA, Murray JA, Green PHR, Khosla C. Latiglutenase improves symptoms in seropositive celiac disease patients while on a gluten-free diet. Digestive Diseases and Sciences. 2017;62(9):2428-2432. DOI: 10.1007/s10620-017-4687-7
  121. 121. Vaquero L, Rodríguez-Martín L, León F, Jorquera F, Vivas S. New coeliac disease treatments and their complications. Gastroenterology Hepatology. 2018;41(3):191-204
  122. 122. Cellier C, Bouma G, van Gils T, Khater S, Malamut G, Crespo L, 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. The Lancet Gastroenterology & Hepatology. 2019;4(12):960-970. DOI: 10.1016/S2468-1253(19)30265-1
  123. 123. Lähdeaho ML, Scheinin M, Vuotikka P, Taavela J, Popp A, Laukkarinen J, et al. Safety and efficacy of AMG 714 in adults with coeliac disease exposed to gluten challenge: A phase 2A, randomised, double-blind, placebo-controlled study. The Lancet Gastroenterology & Hepatology. 2019;4(12):948-959. DOI: 10.1016/S2468-1253(19)30264-X
  124. 124. Syage JA, Green PHR, Khosla C, Adelman DC, Sealey-Voyksner JA, Murray JA. Latiglutenase Treatment for celiac disease: Symptom and quality of life improvement for seropositive patients on a gluten-free diet. GastroHep. 2019;1(6):293-301. DOI: 10.1002/ygh2.371
  125. 125. Serena G, Kelly CP, Fasano A. Nondietary therapies for celiac disease. Gastroenterology Clinics of North America. 2019;48(1):145-163. DOI: 10.1016/j.gtc.2018.09.011
  126. 126. Caio G, Ciccocioppo R, Zoli G, De Giorgio R, Volta U. Therapeutic options for coeliac disease: What else beyond gluten-free diet? Digestive and Liver Disease. 2020;52(2):130-137. DOI: 10.1016/j.dld.2019.11.010
  127. 127. Kivelä L, Caminero A, Leffler DA, Pinto-Sanchez MI, Tye-Din JA, Lindfors K. Current and emerging therapies for coeliac disease. Nature Reviews. Gastroenterology & Hepatology. 2021;18(3):181-195. DOI: 10.1038/s41575-020-00378-1
  128. 128. Segura V, Ruiz-Carnicer Á, Sousa C, Moreno ML. New insights into non-dietary treatment in celiac disease: Emerging therapeutic options. Nutrients. 2021;13(7):2146. DOI: 10.3390/nu13072146

Written By

Ángela Ruiz-Carnicer, Verónica Segura, Carolina Sousa and Isabel Comino

Submitted: 01 March 2022 Reviewed: 24 March 2022 Published: 20 May 2022