Next Article in Journal
The Longitudinal Influence of Parent–Grandparent Coparenting Relationships on Preschoolers’ Eating Behaviors in Chinese Urban Families: The Mediating Roles of Caregivers’ Feeding Behaviors
Previous Article in Journal
Synbiotic Supplementation with Probiotics and Omega-3 Fatty Acids Enhances Upper-Body Muscle Strength in Elite Swimmers: Evidence for Gut–Muscle Axis Modulation During Race-Pace Training
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Is There a Future Without Gluten Restrictions for Celiac Patients? Update on Current Treatments

by
Marina Girbal-González
1,2 and
Francisco J. Pérez-Cano
1,2,*
1
Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain
2
Nutrition and Food Safety Research Institute (INSA-UB), University of Barcelona (UB), 08921 Santa Coloma de Gramenet, Spain
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(18), 2960; https://doi.org/10.3390/nu17182960
Submission received: 31 July 2025 / Revised: 11 September 2025 / Accepted: 12 September 2025 / Published: 15 September 2025
(This article belongs to the Section Nutritional Immunology)

Abstract

Celiac disease (CeD) is a chronic autoimmune enteropathy triggered by dietary gluten in genetically predisposed individuals. Along with other disorders such as non-celiac gluten/wheat sensitivity and gluten allergy, adherence to a strict gluten-free diet (GFD) is required as the only effective treatment for CeD. To this end, and partially due to the burdensome nature and limited efficacy in some patients of a GFD, significant research into alternative therapies has been catalyzed. This review gives a perspective on current and emerging treatment strategies targeting different aspects of CeD pathogenesis. These include gluten-degrading enzymes (e.g., AN-PEP, Latiglutenase, Zamaglutenase), gluten-sequestering agents (e.g., AGY-010, BL-7010), modulators of intestinal permeability (e.g., Larazotide acetate, IMU-856), immune-modulating agents (e.g., ZED1227, AMG 714, EQ102), and strategies for immune tolerization (e.g., TAK-101, KAN-101, Nexvax2). Newer approaches are also targeting probiotics to modulate the gut microbiota (e.g., VSL#3, Lactobacillus plantarum HEAL9), nutraceuticals (e.g., polyphenols, vitamins), or food modifications to remove the gluten from naturally gluten-containing foodstuffs (e.g., gluten transamidation, Gluten Friendly™ technology). Despite encouraging results in preclinical and clinical trials, no treatment has yet been conclusively proven to serve as an effective alternative to the GFD. Continued research is essential to validate efficacy, optimize dosing, and ensure safety in broader patient populations. Here, we provide a comprehensive overview of the therapeutic landscape for CeD, analyze the main strengths and limitations of each treatment and highlight promising directions for future management of CeD, altogether evidencing the urgent need to develop effective alternatives for these patients.

Graphical Abstract

1. Celiac Disease Pathophysiology and Limitations of a Gluten-Free Diet

Celiac disease (CeD) is a chronic autoimmune enteropathy mediated by exposure to dietary gluten in genetically predisposed individuals [1,2]. Gluten encompasses a group of related storage proteins found mainly in wheat and in other cereals including rye, barley and oat. In wheat, the main proteins are gliadin and glutenin, which belong to the prolamin family and contain high amounts of glutamine and proline residues. These can resist degradation by regular digestive enzymes, which can mediate the immune reaction triggered after gluten intake in the context of CeD [3]. One of the most immunoreactive peptides in CeD is 33-mer, resulting from degradation of α2-gliadin by intestinal enzymes and which contains three T-cell-reactive epitopes [4]. Such partially digested peptides, also named gluten immunogenic peptides (GIPs), are mainly transported through transcellular pathways and reach the lamina propria, where they can be deamidated by the tissue transglutaminase 2 enzyme (tTG2 or TG2). Then, either these deamidated gluten peptides (DGPs), alone or complexed to tTG2, can bind to the Human leukocyte antigen (HLA)-DQ2 or DQ8 molecules in antigen-presenting cells (APCs). These activate gluten-specific CD4+ T cells, which leads to the development of tTG2- and gluten-specific B cells that evolve into anti-tTG2 and anti-DGP antibody-producing plasma cells. Simultaneously, an overall inflammatory milieu is created through secretion of interleukin (IL)-15 by APCs and epithelial cells or IL-21 and interferon (IFN)-ɣ by activated CD4+ T cells, among others. These pro-inflammatory cytokines also promote infiltration of lymphocytes into the gut epithelium (IELs) and cause intestinal atrophy [5].
The worldwide prevalence of CeD ranges from 1.1 to 1.7% [6], and it affects more women than men in a 1.79:1 ratio [7]. The seroprevalence of CeD is higher in Europe and Asia, and it is thought to be able to increase in the coming years since most patients remain undiagnosed and new screening policies are being implemented [8]. Nevertheless, the only currently effective treatment for CeD is strict adherence to a GFD, which can be difficult to maintain and poses a great socioeconomic burden [9]. Hence, finding a treatment for CeD is presently a very active line of research in the biomedical field. However, even with this commitment, many of the investigations performed and technologies developed failed to finally reach patients and help in day-to-day life.
In this perspective, we aim to thoroughly present all the current approaches to prevent or treat CeD. Although this topic is being approached from many different, sometimes complementary, outlooks, the options that are currently under research fall into eight main categories based on their mechanism of action. First, gluten-degrading enzymes, one of the most promising strategies, aim to digest gliadin and GIPs to obtain smaller peptides, which can then potentially go unrecognized by the immune system. Alternatively, gluten-sequestering or -neutralizing compounds can complex to gluten or gliadin and prevent them from crossing into the lamina propria. Many kinds of immune modulators have also been investigated, which intend to stop the gluten-derived immune cascade at different stages. For example, TG2 inhibitors try to prevent deamidation of GIPs and the subsequent interaction with the HLA-DQ2/8 receptors in CD4+ T cells. Other strategies within this category include IL-15 inhibitors, which aim to prevent the establishment of an inflammatory milieu and IEL infiltration, or HLA-DQ2 inhibitors, to prevent interaction between these and deamidated GIPs. Gluten tolerization has also been researched with the objective to prevent an immune response to gluten ingestion altogether. Moreover, since gut dysbiosis is a hallmark of CeD, two alternative strategies, i.e., promotion of intestinal integrity and gut microbiota modulation, have been targeted through intestinal permeability (IP) modulators and pro-, pre-, synbiotics and fecal microbiota transplantation (FMT), respectively. The working hypothesis for these treatments is that an improvement in overall gut health might help decrease the CeD-associated symptoms. Simultaneously, the use of a wide range of naturally derived bioactive compounds (nutraceuticals) has also been researched to prevent or ameliorate CeD symptomatology, since this can exert anti-inflammatory, antioxidant and prebiotic actions. Finally, several strategies have been directed at achieving gluten-free foods from gluten-containing cereals, through modification of gliadin, breeding strategies or enzymatic degradation. A schematic overview of CeD pathophysiology and the mechanistic target of all the reviewed therapeutic agents is shown in Figure 1.
Initially, the inclusion criteria for the current perspective comprised scientific articles primarily published within the last ten years, which were identified through a comprehensive search of the literature and subsequently categorized according to the treatment field (e.g., gluten-degrading enzymes, gluten neutralization strategies, immune-modulating therapies). However, given our aim to provide an integrative overview of approaches that have failed, evolved, or remain under active development, earlier publications predating this timeframe, including seminal studies foundational to current research directions, were also considered.

2. Gluten-Degrading Enzymes

This approach has been researched for over 20 years, and multiple clinical trials are still going on. For an enzyme to be considered a good therapeutic option for CeD, it must meet several criteria simultaneously: (1) efficiently target and break down the proline- and glutamine-rich peptides, (2) remain active in the acidic environment of the stomach (low pH), (3) resist proteolysis by digestive enzymes, (4) be active at the body’s temperature and (5) be non-toxic and non-immunogenic [10]. Active, completed and terminated trials, as well as in vitro and preclinical studies on the topic, are summarized in Table 1 and Figure 2.

2.1. Fungi-Derived Enzymes

Among the most prominent options within this category, we can find AN-PEP (Aspergillus niger prolyl endopeptidase), a fungus-derived enzyme with activity at the stomach’s pH values and resistant to degradation by pepsin. Although this enzyme is already available on the market, conflicting studies exist on its efficacy. In initial studies, AN-PEP was shown to accelerate gluten digestion [11], to partially degrade gluten in a gastric simulator [12] and to significantly digest gluten in two clinical trials (NCT02060864 and NCT01335503) [13,14]. Nevertheless, in subsequent phase 1 and 2 pilot studies (NCT00810654) [15] and an exploratory phase 4 study (NCT04788797) [16], no difference in the presence of GIPs and other relevant biomarkers between the placebo and AN-PEP groups was found. The authors stated in a previous publication that “AN-PEP should not be used to replace a gluten free diet, but rather to support digestion of occasional and/or inadvertent gluten consumption”. To this end, current research around the AN-PEP enzyme is targeted at achieving gluten-free foodstuffs, especially bread [17,18], beer [19] and pizza [20].
Another recent fungi-derived enzyme aiming to treat CeD is AMYNOPEP, a combination of two polypeptides from Trichophyton rubrum, a leucine aminopeptidase 2 displaying aminopeptidase activity (AP) and a dipeptidylpeptidase IV (DPPIV) [21]. A study on AMYNOPEP reported full degradation (99.5%) of 33-mer into single and dipeptide amino acids in vitro when used in a 1:10 enzyme/33-mer ratio [22]. In healthy volunteers (German Register of Clinical Studies, DRKS00033099), 33-mer was administered alone or mimicking more complex meals through addition of apple juice, a whey protein drink or wheat gluten, with or without AMYNOPEP. Results from this proof-of-concept study showed faster and larger degradation of 33-mer when AMYNOPEP was administered [22].
Other fungi-derived enzymes are a DPPIV from Aspergillus oryzae or an aspergillopepsin from Aspergillus niger (ASP) [23]. While both DPPIV and ASP alone failed to efficiently cleave GIPs, a phase 1 and 2 clinical trial (NCT00962182) using a cocktail of both enzymes (STAN1) reported moderate gluten detoxification properties [23].
Table 1. Gluten-degrading enzymes.
Table 1. Gluten-degrading enzymes.
OriginSpeciesNameTypePhase, Clinical Trial, SponsorRef.
FungiA. nigerAN-PEPSerine prolyl endopeptidaseInterventional (NCT02060864,
NCT01335503), Phase 1 and Phase 2 (NCT00810654), Phase 4 (NCT04788797)/DSM Food Specialties
[11,12,13,14,15,16]
A. niger
and A. oryzae
STAN1Aspartate aspergillopepsin (ASP) + serine dipeptidyl-peptidase IV (DPPIV)Phase 1 and phase 2 (NCT00962182)/Heim Pal Children’s Hospital[23]
A. oryzaeFlavourzymeMix of endo- and exo-peptidasesIn vitro[24]
T. rubrumAMYNOPEPLeucine aminopeptidase 2 and DPPIVGerman Register of Clinical
Studies, DRKS00033099
[21,22]
BacteriaR. aeria and R. mucilaginosaR. aeria and R. mucilaginosaSubtilisin-type serine endopeptidasesIn vitro[25]
A. sendaiensisZamaglutenase/TAK-062 (from KumaMax and Kuma062)Serine endopeptidasePhase 1 (NCT03701555), Phase 2 (NCT05353985)/Takeda[26,27,28]
B. gladioliBga1903Serine endopeptidaseIn vitro[29]
A. A8Endopeptidase 40 (E40)EndopeptidaseIn vitro[30]
M. xanthusMX PEPProlyl endopeptidaseIn vitro[31]
F. meningosepticumFM PEPProlyl endopeptidaseIn vitro[32,33]
S. capsulateSC-PEPProlyl endoproteaseIn vitro[34]
B. licheniformisAlcalaseSerine endopeptidaseIn vitro and in vivo[24,35]
Plant- and food-derived enzymesN. ventrataCeliacase (neprosin)Prolyl endopeptidaseIn vitro and in vivo[36,37]
Pineapple (A. comosus)BromelainCysteine proteaseIn vitro[38]
Fig latex (F. carica)FicinCysteine proteaseIn vitro[38]
Hayward Kiwi (A. deliciosa cv. Hayward)ActinidinCysteine proteaseIn vitro and in vivo[39,40,41]
Papaya latex (C. papaya)Papain/Caricain (crude or purified)Cysteine proteaseIn vitro and in vivo[24,35,42]
T. aestivumTriticain-αCysteine proteaseIn vitro[43]
Hordeym vulgare (Barley)EP-B2Cysteine endoproteaseIn vitro and in vivo[32]
Combined (Bacteria + Plants)S.
Capsulate + H. vulgare (Barley)
Latiglutenase (ALV003 or IMX003)Glutamine-specific cysteine peptidase + modified serine prolyl-specific oligopeptidasePhase 1 (NCT00626184, NCT00669825), Phase 2 (NCT00959114, NCT01255696, NCT01917630, NCT03585478)/Immunogenics, LLC[44]

2.2. Bacteria-Derived Enzymes

An early study identified around 60 bacteria naturally present in the oral cavity with gluten-hydrolyzing properties, from which Rothia mucilaginosa and Rothia aeria displayed the highest activity towards gluten degradation [25]. Later on, it was discovered that R. aeria achieves gluten detoxification through the BAV86562.1 subtilisin. In a preclinical study, addition of live or dead R. aeria to regular chow led to a 20–33% larger degradation of gliadins and immunogenic epitopes in vivo in BALB/c mice compared to regular chow [45].
Zamaglutenase (TAK-062, previously Kuma-062 or KumaMax) [26,27] is a serine endopeptidase derived and modified from the bacterial enzyme kumamolisin-As produced by Alicyclobacillus sendaiensis. Its main strength is that it targets the proline-glutamine (P-Q) motif instead of just one of either amino acid residues. It has proteolytic activity at a large pH range (2.5–6.0) and has been shown to degrade a 49.0–99.9% of the gluten present in a complex meal (1–6 g) 20–65 min after its intake. The phase 1 clinical trial (NCT03701555) also demonstrated its safety and tolerability at doses ranging from 100 to 900 mg [26]. A phase 2 trial (NCT05353985) has been completed but results have not been made available [28].
Bga1903 is a serine endopeptidase enzyme originating from Burkholderia gladioli [29]. In a previous study, it was reported to effectively degrade the 33- and 26-mer gluten-derived immunotoxic peptides in vitro and in beer in conjunction with peptidases from A. niger. While the mature version of Bga1903 is not considered to be ideal for gluten degradation, its optimum pH of 7.0 highly differs from the acidic conditions in the stomach, and the authors indicated that the enzyme could be modified in order to better perform as an oral therapy for CeD or so as to be targeted to reduce gluten content in foods.
Another bacterial-derived enzyme, Endoprotease 40 (E40), secreted by Actinoallomurus A8, was reported to be pepsin and trypsin resistant and activated at a pH ranging from 3 to 6 [30]. E40 achieved complete in vitro degradation of 33-mer and whole gliadin at 1:48 and 1:96 molar ratios, after 30 and 240 min, respectively, as assessed via high-performance liquid chromatography (HPLC). Moreover, E40-treated gliadin samples contained gluten levels below 20 ppm, when analyzed by an enzyme-linked immunosorbent assay (ELISA). Treatment of T cells obtained from CeD patients with TG2-deamidated E40-digested gliadin did not lead to the secretion of IFN-ɣ, contrary to what was observed for untreated samples.
Other enzymes which were researched in the past but are no longer being worked on include the bacterial prolyl endopeptidases from Myxococcus xanthus (MX PEP) [31] and Flavobacterium meningosepticum (FM PEP) [33,46].

2.3. Plant- and Food-Derived Enzymes

Neprosin is a prolyl endopeptidase naturally secreted by the carnivorous plant Nepenthes × ventrata. A previous study [37] showed that a combination of two proteolytic components from N. ventrata, nepenthesin and neprosin (in a 4:1 rate) displayed high gluten-degrading efficacy and prevented accumulation of IELs in vivo using HLA-DQ8 non-obese diabetic (NOD/DQ8) mice. Moreover, the proteolytic efficiency in vitro of the combined enzymes was found to be much higher than the readily available AN-PEP enzyme, thus requiring smaller amounts to achieve similar degradation rates. In a subsequent study [36], neprosin was shown to be capable of degrading gliadin in a 1:500–1000 enzyme/gliadin ratio. Moreover, it was able to efficiently function at the temperature and pH of the stomach both in vitro and in vivo, where it degraded the gluten administered to mice by up to 90%. Celiacase is a new gluten-degrading enzyme originating from Neprosin, which has been modified to achieve higher peptidolytic activity, as well as improved expression and stability [47].
Several studies have focused on enzymes naturally present in the fruit, latex or root from fruit trees. Two of them are Bromelain, an enzyme encompassing multiple cysteine proteases originating from pineapple (Ananas comosus), and ficin, a cysteine protease derived from the latex of the Ficus tree (Ficus carica). Digestion of gliadin and gliadin-derived 19-mer peptide by a mixture of bromelain and ficin (BF) led to significantly lower toxicity on Caco-2 cells and decreased intestinal permeability, similar to that of treatment with Larazotide acetate [38] (see Section 2.3. Intestinal permeability modulators).
Another food-derived enzyme is actinidin, a cysteine protease from Hayward kiwifruit (Actinidia deliciosa cv. Hayward), which showed small gluten degradation properties in rats, specifically for ω-gliadin [39], and a maximum degree of 15% gluten hydrolysis in a gastrointestinal simulator. Nevertheless, actinidin was the best-performing enzyme when compared to other naturally sourced enzymatic compounds like papain or bromelain [40,41].
A comparative study assessed the gluten-degrading capacity of three enzymes from natural origins: papain from the papaya-fruit latex (Carica papaya), flavourzyme from the fungus Aspergillus oryzae and alcalase from the bacterium Bacillus licheniformis, which were compared to animal trypsin, α-chymotrypsin and pepsin. The authors concluded that sequential treatment of wheat flour with alcalase and papain was the most effective combination, leading to the lowest immunoglobulin (Ig)E-binding [24]. In a subsequent study, BALB/c mice were sensitized with either regular gliadins, phosphorylated gliadins or alcalase- or papain-hydrolyzed gliadins. All pre-treatments significantly decreased serum total IgE, histamine, IFN-γ and IL-4 levels, compared to those in mice receiving untreated gliadin [35]. Another study obtained crude caricain/papain from papaya latex, purified it, and demonstrated that treatment of wholegrain bread fermented at 30–37 °C from 5 to 7 h with purified caricain/papain managed to reduce up to 98% of the gliadin originally present, and that the purified enzyme worked better than its crude version [42]. Another comparative study evaluated wheat-, rye- and barley-gluten degradation by either papain, bromelain, alcalase, flavourzyme, pepsin and trypsin [48]. The results showed that papain was capable of digesting gluten below the limit of detection for all food matrices, and the same was observed for alcalase-, flavourzyme- and pepsin-digested wheat and rye. The poorest-performing enzymes were bromelain and trypsin.
In terms of cereal-derived enzymes, triticain-α is a cysteine protease present in wheat (Triticum aestivum) [43] with potential gluten-hydrolyzing properties based on its cleavage sites and stability in acidic conditions. Nevertheless, these have not been proved in in vitro or in vivo experiments.

2.4. Combined Bacterial- and Plant-Derived Enzymes

Latiglutenase [44] (formerly ALV003 or IMX003) is composed of two glutenases active at the stomach’s pH in a 1:1 ratio, namely EP-B2, a cysteine endoprotease originating from germinating barley seeds [32], and SC-PEP, a prolyl endoprotease (PEP) from Sphingomonas capsulate able to cleave gluten at the proline residues [34]. Previous studies have shown that pre-treatment of gluten with this enzyme reduced the IFN-γ levels in blood but was not linked to a change in symptoms [44]. Two phase 1 (NCT00626184 and NCT00669825) and phase 2 (NCT00959114 and NCT01255696) clinical trials demonstrated that Latiglutenase was well tolerated at doses ranging from 100 to 1800 mg [49] and was able to prevent development of villous atrophy in patients following a GFD containing traces (2 g gluten per day) [50]. Nevertheless, in another phase 2 clinical trial (NCT01917630), Latiglutenase was not capable of reducing histologic and symptom scores in symptomatic CeD patients following a GFD [51]. In a more recent phase 2 trial (NCT03585478), it was demonstrated that Latiglutenase intake at a 1200 mg dose was capable of reducing the damage associated with daily intake of gluten traces (2 g/day) in CeD patients otherwise following a GFD, as observed by both a decrease in symptom- and histological-severity values [52]. While some results of the aforementioned trials are promising, others are conflicting, which may indicate that Latiglutenase could be an option to avoid histological damage derived from the presence of gluten traces in meals but would not be an alternative to a GFD.

2.5. Limitations and Strengths of Current Gluten-Degrading Enzymes

  • Strengths:
  • Precise target: Gluten-degrading enzymes directly target the root problem in CeD, i.e., the GIPs that are resistant to digestion and then trigger immune responses. Their mechanism is rational and well defined, making them appealing therapeutic candidates.
  • High safety and tolerability: Due to enzyme specificity, these are usually easily tolerated and do not interfere with other biological pathways.
  • Oral delivery: Most researched enzymes are proposed to be delivered orally in a pill format before meals, which is easy and practical for patients.
  • Role as a GFD-adjuvant: Some of the enzymes show potential in supporting digestion of inadvertent gluten exposure, which is a practical issue for patients strictly adhering to a GFD.
  • Limitations:
  • Physiological barriers: Maintaining enzyme activity through the acidic, protease-rich environment of the stomach and achieving effective gluten degradation in real-meal contexts remains challenging, especially in mixed meals with complex matrices. From all the reviewed gluten-degrading enzymes, only a few have managed to overcome this frequent hindrance.
  • High gliadin/enzyme ratio: Most of the enzymes developed required a high amount of the active proteins in order to properly degrade gluten in an effective manner.
  • Site-specific enzyme activation: Effective gluten-degrading enzymes must be activated at the stomach’s acidic pH, where complete gliadin degradation should occur, and revert to an inactive state as pH rises along the gastrointestinal tract (GIT), as other ways GIPs may persist and trigger downstream immune responses.
  • Inconsistent clinical efficacy: Despite promising in vitro and preclinical results, many enzymes have failed to show consistent benefit in human trials. Hence, since no enzyme therapy to date can serve as a standalone alternative to a GFD, even the most promising candidates are currently seen as adjunctive, not curative.
Due to their strengths and despite their limitations, which do not apply to all enzymes currently under investigation, gluten-degrading enzymes represent one of the most promising therapeutic approaches to CeD, given their high target specificity, safety, and ease of administration.

3. Gluten Direct Neutralization

Another strategy that has been proposed in the search for alternatives to a GFD is that of gluten neutralization, either through the use of targeted antibodies (Abs) or by means of complexing gluten to other molecules, thus rendering it unavailable to interact with the host’s immune system (Table 2). The main neutralization strategy consists of the IgY Ab from the egg yolk (AGY) of hens, which have been previously hyper-immunized against gliadin [53]. An important advantage of this method is that anti-gliadin IgY is not absorbed into the blood by humans; thus, it can act in the GIT while going undetected by the immune system. Moreover, it is easily producible in large amounts. A previous study showed in vivo with BALB/c mice that, after AGY-010 intake at a 1:5 AGY/gliadin ratio, only less than 1% of the gliadin administered reached the animal’s GIT [53]. Posteriorly, AGY-010 was reported to be safe for consumption in a phase 1 4-week clinical study (NCT01765647) [54] with biopsy-proven CeD patients following a GFD. A phase 2 (NCT03707730) [55] study has been completed but the results are not available yet.
Alternatively, BL-7010, a poly(hydroxyethyl methacrylate-co-styrene sulfonate) (P(HEMA-co-SS)) copolymer, was shown to effectively perform gliadin sequestration in both in vitro cell cultures and HLA-HCD4/DQ8 or NOD-DQ8 mice [56]. As a result of gliadin complexation with BL-7010 and the smaller amounts of GIPs consequently generated, the authors reported a decrease in cell mortality, IEL infiltration and paracellular permeability. BL-7010 also led to a higher villous height/crypt depth ratio (Vh:Cd) and was shown to be safe in rats at high doses [57]. While a phase 1 and 2 study was registered in 2013 (NCT01990885), its results have not been made available yet [58].

Limitations and Strengths of Current Gluten Neutralization Treatments

  • Strengths:
  • Early incidence: Since these strategies aim to block gluten passing through the epithelial layer, they act on the very first step of CeD pathophysiology, preventing the derived immune reaction before its initiation.
  • Safety and tolerability: The anti-gliadin IgY antibodies from egg yolk are not absorbed systemically, significantly reducing the risk of immune reactions. This makes them particularly safe as oral therapies.
  • Ease of production: Anti-gliadin IgY antibodies can be produced in large quantities at relatively low cost, offering a practical advantage over more complex biologics.
  • Oral delivery: Both proposed treatments would be delivered orally, which generally show better acceptance scores by patients than other methods (e.g., vaccines).
  • Limitations:
  • Uncertain site of action: It remains unclear where gluten neutralization primarily occurs, whether in the stomach or further along the GIT, which would allow for GIPs to be generated and absorbed into the lamina propria, thus triggering an immune reaction. Additionally, the stability and resistance of these agents to gastric pH remain insufficiently characterized.
  • Limited clinical data: While preclinical studies show promise, robust efficacy data in humans are still lacking. The results of critical phase 2 trials are not yet published, creating uncertainty about real-world effectiveness.

4. Intestinal Permeability Modulators

Intestinal permeability (IP) is a common trait of CeD, although the causal effect is still under discussion: does having a “leaky gut” contribute to development of CeD, or does the overall inflammation characteristic of CeD lead to increased gut permeability? Despite what comes first, reducing the CeD-associated IP could not only help reduce the number of GIPs that cross from the intestinal lumen to the lamina propria, but it would also prevent microbial antigens and toxins from trespassing and causing inflammation [64]. To this end, multiple treatments have been researched, which aim to keep the tight junctions (TJs) between intestinal epithelial cells actually “tight” (Table 2).
One of the forerunners on this race is Larazotide acetate (previously AT-1001), a synthetic octapeptide that inhibits paracellular permeability through its function as a zonulin inhibitor [60]. Briefly, zonulin is a protein that modulates TJs and, in normal physiological conditions, it regulates their temporary opening to facilitate nutrient absorption and immune surveillance. Nevertheless, zonulin overexpression in CeD leads to chronic opening of the TJs [65]. Previous clinical studies have shown conflicting results; an initial study consisting of the administration of 2.5 g of gluten and 12 mg of Larazotide acetate for 7 days (n = 21) reported a prevention of IP (measured in lactulose-to-mannitol, LAMA ratios) and a decrease in both IFN-ɣ secretion and Gastrointestinal Symptoms Rating Scale (GSRS) values (NCT00362856, NCT00386165) [60]. A second trial (n = 86) testing four different treatment doses (0.25, 1, 4 or 8 mg) and a similar gluten intake of 2.4 g for 14 days reported no difference in the LAMA ratio or anti-tTG2 levels between the placebo and treatment groups but observed a better GSRS score in Larazotide vs. control patients undergoing a gluten challenge (NCT00362856) [61]. The same cohort was also used in a phase 1 study to evaluate Larazotide’s safety and tolerance (NCT00386490), which was deemed to be well tolerated. In a subsequent phase 2 cohort (NCT00492960 and NCT00889473), patients received 900 mg of gluten thrice per day, to a daily amount of 2.7 g of gluten, and 1, 4 or 8 mg of Larazotide 15 min before each meal for 6 weeks [62]. The authors reported a significant decrease in anti-tTG2 IgA levels in all the treatment groups, compared to the placebo one, but no differences in LAMA ratios. On a different direction, Larazotide acetate was tested in CeD patients (n = 342) following a GFD but with ongoing symptoms. Four doses were tested (0.5, 1 or 2 mg thrice daily) for 4 weeks and only the 0.5 mg group achieved the primary end point, i.e., a significant decrease in GSRS scores (NCT01396213) [59]. A 2019 phase 3 clinical trial (NCT03569007) was terminated without completion by the sponsor (9 Meters Biopharma, Inc.) [63].
Another recent IP regulator for the treatment of CeD is IMU-856, a modulator of sirtuin 6 (SIRT6) [66]. SIRT6 overexpression has been reported to display a protective effect against the loss of function by TJs and histopathological damage in a mouse model of colitis [67]. In a phase 1 clinical trial (ACTRN12620000901909, Australian New Zealand Clinical Trials Registry and CT-2020-CTN-01997-1, Therapeutic Goods Administration Clinical Trial Notification), IMU-856 administration at different doses alongside gluten was found to cause mild but frequent treatment-emergent adverse events (TEAEs). Moreover, no significant improvements in histology parameters because of IMU-856 administration, and regardless of dose, were found.

Limitations and Strengths of Current Intestinal Permeability Modulators

  • Strengths:
  • Disease target: The focus on intestinal permeability directly addresses a core feature of celiac pathophysiology, i.e., the disruption of TJs, which will then prevent access of GIPs into the lamina propria. Moreover, since these treatments typically act locally within the gut, systemic immune suppression and the risk of broader side effects common with immunomodulators are minimized.
  • Safety and tolerability: Both existent strategies have demonstrated good tolerability in phase 1 and 2 trials, with mild or manageable adverse events, making them promising from a safety standpoint.
  • Oral delivery: Both proposed treatments would be delivered orally, which generally show better acceptance scores by patients than other methods (e.g., vaccines).
  • Symptom relief in some trials: Larazotide acetate has shown symptomatic benefit in several trials, even when histological or biomarker outcomes are inconsistent. This suggests potential value as an adjunct therapy for patients with persistent symptoms despite adherence to a GFD.
  • Limitations:
  • Indirect activity: IP modulators do not alter gliadin itself; therefore, gliadin levels and GIP production remain unchanged. Their action is limited to preventing peptide translocation into the lamina propria, meaning that even minimal peptide crossing may still be sufficient to initiate CeD pathophysiology.
  • Limited clinical effectiveness and inconsistent outcomes: No current IP modulator has demonstrated the ability to fully prevent villous atrophy or histological damage in response to gluten exposure, which is a key therapeutic goal in celiac disease. Moreover, while some studies reported reduced antibody levels or symptoms, others failed to demonstrate significant improvements in intestinal permeability or histological recovery. Results are often dose-dependent and inconsistent across studies.
  • Uncertain causality in pathogenesis: It remains unclear whether increased IP is a cause or a consequence of CeD. This ambiguity complicates the rationale for prioritizing it as a standalone therapeutic target.

5. Immune Modulators

5.1. TG2 Inhibitors

As previously mentioned, the enzyme TG2 is one of the key players in the autoimmune reaction elicited by exposure to dietary gluten in CeD patients. Briefly, the GIPs that reach the lamina propria are deamidated by TG2, which significantly enhances its affinity to the HLA-DQ2/DQ8 receptors in APCs. Hence, TG2 inhibition is one of the most evident strategies to prevent CeD. To this end, ZED1227 (TAK-227), a selective oral TG2 inhibitor, was administered to CeD patients at one of three doses (10, 50 or 100 mg) alongside a 3 g gluten challenge for 6 weeks. Results of a phase 2 clinical trial (2017-00224-30, CEC-3 EudraCT number) showed that ZED1227 intake significantly decreased the extent of histological damage (measured as Vh:Cd), as compared to the placebo, but was not capable of completely preventing intestinal epithelial injury [68]. It was also deemed safe in two phase 1 trials (2014-003044-13 and 2015-005283-42) [69].
Similarly, another TG2 inhibitor, GSK-3915393, was being studied for CeD, but after phase 1 completion, the study was discontinued by the sponsor (NCT04604795) [70] and is currently only being studied for Idiopathic Pulmonary Fibrosis [71].

5.2. Anti-IL-15 Treatments

IL-15 has a crucial role in the pathogenesis of CeD. It has been shown that overexpression of IL-15 in the lamina propria and in intestinal epithelial cells (IECs) is essential for the development of villous atrophy in CeD [72]. Briefly, the conjunction of IL-15 and other pro-inflammatory cytokines such as IFN-ɣ promotes the acquisition of a CD8+ cytotoxic phenotype by IELs, which then mediate tissue damage after acquiring a killer phenotype. Moreover, it prevents the differentiation of regulatory T cells and promotes loss of oral tolerance [5,73]. Hence, targeting IL-15 is one of the strategies being investigated for treatment of CeD (Table 3). AMG 714 (or PRV-015) is an injection-administered IgG1κ-type human monoclonal antibody (moAb) against IL-15. It is hypothesized that it could inhibit the T-cell activation induced by IL-15 and the immune reaction that follows. AMG 714 has been studied in three phase 2 studies (NCT02637141, NCT02633020 and NCT04424927) for its efficacy and safety in adult patients with CeD [74], type II refractory CeD (RCeD-II) [75] and non-responsive CeD [76], respectively. Results from the first two trials showed no significant difference in the percentage of IELs, Vh/Cd, anti-TG2 or anti-DGP Abs between the treatment and placebo groups. In patients with non-responsive CeD, an improvement in diarrhea as measured by the Bristol stool scale was reported.
Another potential treatment targeting IL-15 inhibition is CALY-002, a moAb that neutralizes IL-15 through prevention of its interaction with IL-15Rβ [77], a receptor in IL-15-responder cells [78]. In a preclinical study regarding RCeD and eosinophilic esophagitis (EoE), the authors reported that treatment with CALY-002 significantly reduced the IELs in IL-15TgE mice (a mouse model of IL-15 overexpression) and the natural killer (NK) cells in cynomolgus monkeys. A downside of CALY-002 is that it does not prevent IL-15 recognition by receptor IL-15α. While CeD and EoE are not directly associated, they can co-occur [79]. A phase 1 study was recently completed (NCT04593251), but results are not available yet [80].
TEV-53408, an alternative anti-IL-15 moAb, managed to reduce the proliferation of mature NK cells, which have increased cytotoxicity and produce more pro-inflammatory cytokines [81]. A phase 1 study concluded that TEV-53408 was safe in healthy volunteers [82], and a phase 2 study was initiated in 2025 and is currently in the recruitment phase (NCT06807463) [83].
Alternatively, EQ102 is a gamma-chain receptor antagonist that inhibits both IL-15 and IL-21 [84]. Its sponsor company, Equillium, announced a phase 1 study of EQ102 in 2022 (ACTRN12622001449729) [85,86] and phase 2 study on EQ101 (IL-2, IL-9, and IL-15 inhibitor) and EQ102 in 2023 for Alopecia Areata (AA) and CeD (NCT05589610) [87]. While the results of this trial have not been made available to date, it was reported that EQ102 displayed lower bioavailability than expected. Hence, EQ302, an alternative treatment with increased potency, was developed from EQ102, its parent peptide [88]. As a result, work with EQ102 was terminated. Research on EQ302 is currently in the preclinical stage [89].
A recent study evaluated ex vivo the effect of aIL-15, an anti-IL-15 moAb, on the immune response responsible for intestinal epithelial damage from untreated CeD patients’ jejunal biopsies (Ethics committee of the San G. Moscati Hospital, Avellino, Italy, CECN/809) [90]. Results showed that those samples receiving aIL-15 displayed significantly smaller levels from molecules involved in tissue atrophy, including Fas, HLA-E, perforin and IFN-γ, compared to controls. While results are promising, further research on the effect of aIL-15 on relevant biomarkers of CeD, such as TG2 levels or intestinal epithelium histological measurements, is needed.
Table 3. Immune modulation strategies.
Table 3. Immune modulation strategies.
StrategyActive PrincipleNamePhase, Clinical TrialSponsorRef.
TG2 inhibitorsSIRT6 modulatorIMU-856Phase 1 (ACTRN12620000901909 or CT-2020-CTN-01997-1)Immunic Australia Pty Ltd.[66]
TG2 inhibitorZED1227Phase 1 (2014-003044-13)
Phase 2 (2015-005283-42 and 2017-00224-30)
Dr. Falk Pharma GmbH[68,69]
TG2 inhibitorGSK3915393Phase 1–Discontinued (NCT04604795)GlaxoSmithKline[70]
IL-15 inhibitorsIL-15 inhibitor (moAb)AMG 714 (PRV-015)Phase 2 (NCT02633020, NCT02637141 and NCT04424927)Amgen and Provention Bio (Sanofi) [74,75,76,91]
IL-15 inhibitor (moAb)CALY-002Phase 1 (NCT04593251)Calypso Biotech BV[77,80]
IL-15 inhibitor (moAb)TEV-53408Phase 1 (NCT06807463)Teva Branded Pharmaceutical Products R&D LLC[82,83]
IL-15 and IL-21 inhibitor (gamma chain receptor antagonist)EQ102Phase 1 and 2 (ACTRN12622001449729)Equillium[84,85,86,87,88]
IL-15 inhibitor (moAb)aIL-15Ex vivoISA-CNR, ELFID *[90]
Lymphocyte migration inhibitorsCCR9 antagonistGSK1605786A/vercirnon (formerly CCX282-B)Phase 2 (NCT00540657)Amgen (formerly ChemoCentryx)[92,93]
α4β7 integrin inhibitorPTG-100Phase 1 (NCT04524221)Nielsen Fernandez-Becker[94,95]
HLA-DQ2 inhibitorHLA-DQ2 inhibitorAzidoprolineIn vitro [96,97]
JAK inhibitionPan-JAK inhibitorTofacitinib (Xeljanz)Phase 2 (EudraCT 2018-001678-10)VU Medical Center[98]
JAK3 and TEC kinase family inhibitorRitlecitinib (LITFULO)Phase 2 (NCT05636293)Massachusetts General Hospital/Pfizer[99,100,101]
CFTR potentiatiorCFTR potentiatorIvacaftor (Kalydeco)Case studies [102,103,104]
* ISA-CNR = Institute of Food Science, National Research Council, ELFID = European Laboratory for the Investigation of Food-Induced Diseases.

5.3. Lymphocyte Migration Inhibitors

Lymphocyte migration plays a key role in CeD pathogenesis. Briefly, when GIPs enter the lamina propria and epitopes are taken up by APCs, lymphocytes migrate to the intestinal tissue, where they interact with APCs, are activated and initiate the gliadin-derived immune reaction. Current treatments focusing on the inhibition of lymphocyte migration in the context of CeD are available in Table 3. The CC motif chemokine receptor 9 (CCR9), primarily expressed in lymphocytes, plays an important role in CeD pathogenesis. Briefly, it promotes migration and homing of immune cells to the small intestine through interaction with the CC motif chemokine ligand 25 (CCL25, also known as TECK) receptor, highly expressed in both thymic and small intestine cells [105]. Previous studies have shown an increase in both CCR9 and CCL25 expression in CeD. Since, in the context of CeD, T cells in the gut contribute to damage of the epithelial tissue, it is hypothesized that any medication targeting the CCR9-CCL25 interaction could have a positive impact on reductions in intestinal inflammation. To this end, a phase 2 clinical study was undertaken to evaluate if CCX282-B, an antagonist of CCR9, could mitigate the effects of gluten ingestion in patients with CeD [92]. While a previous preclinical study in a mouse model of Crohn’s disease (TNF∆ARE) had shown that CCX282-B was capable of inhibiting the CCL25-induced chemotaxis mediated by CCR9A and B, and thus exert a preventive effect on intestinal inflammation [93], results in the context of CeD have not been published yet.
In a similar manner, PTG-100 is an inhibitor of the α4β7 integrin expressed in effector memory T cells, leading to their homing to inflamed areas in the GIT through interaction with mucosal addressin cell adhesion molecule 1 (MasCAM-1) [95]. Since a previous study showed a significant increase in MasCAM-1 expression in active CeD [106], blocking the α4β7 integrin/MasCAM-1 axis could be an effective strategy to prevent homing of effector T cells to the gut. While the results of a current phase 1 study on the safety and efficacy of PTG-100 in preventing gluten-induced inflammatory injury to the small intestine in patients with CeD have not been published yet (NCT04524221) [94], a previous clinical study in inflammatory bowel disease (IBS) showed an improvement in histological measurements [95].

5.4. HLA-DQ2 Inhibitors

Since the presence of the Class II HLA genes DQ2.2, DQ2.5 or DQ8 is essential for (but not indicative of) the development of CeD [107], one potential strategy for the prevention of this pathology is their inhibition, which would prevent the initiation of the ensuing immunological reaction. With this goal, one study substituted the proline residues in GIPs by azidoprolines, with the aim to use them to block the HLA-DQ2 molecules and prevent its interaction with regular GIPs [97]. Unfortunately, since the modified gluten peptides displayed similar binding affinity to HLA-DQ2 as normal gluten peptides, they could only partially prevent gluten binding to the receptors and the gluten-derived T-cell activation and immune reaction. Still, an encouraging observation was that the modified peptides complexed to HLA-DQ2 were not recognized by CD4+ T cells. In a following study, the authors actively looked for peptides with higher binding affinity to HLA-DQ2 than that of regular GIPs [96]. The peptide sequence ADAYDYESEELFAA displayed superior binding properties and was modified with non-natural peptides. Altogether, the newly developed sequences achieved a binding affinity that was up to 200-fold higher than that of gluten peptides. Nevertheless, after the last study concluded in 2010, this topic was not investigated further.

5.5. Janus Kinase (JAK) Inhibition

Intestinal permeability is considered a predisposing factor for the development of inflammatory diseases, including CeD. JAKs are a family of intracellular tyrosine kinases that are critical in cell signaling, especially in immune and blood cell functions [108]. The JAK-Signal Transducer and Activator of Transcription (STAT) pathway has a role in the regulation of IP through its selective activation or repression of genes involved in inflammation, cell proliferation, survival and differentiation. Specifically, in CeD, binding of cytokines such as IL-15 or IFN-γ to their corresponding receptors in epithelial cells can trigger the JAK-STAT pathway. Briefly, the JAK1 and JAK3 enzymes are activated and phosphorylated, and they then recruit and phosphorylate STAT proteins, which translocate into the nucleus and promote the expression of pore-forming proteins such as claudin-2, while decreasing the expression of barrier-sealing claudins and other promoters of intestinal integrity, such as occludin or zonula occludens-1 (ZO-1) [108]. Hence, some of the treatments being investigated for the prevention of CeD are JAK inhibitors (Table 3). Tofacitinib, a pan-JAK inhibitor, was subcutaneously delivered to T3b-hIL-15 transgenic (tg) mice in a 30 mg/(kg·day) dose for 42 days. The authors reported a decrease in CD8+NKG2D+ T cells in blood and the intestinal mucosa, reduction in IELs and prevention of villous atrophy. Nevertheless, a significant increase in visceral adiposity was also noted [109]. While many clinical studies have been conducted for Tofacitinib and a wide array of pathologies, the first phase 2 study to assess its effect on CeD, and more specifically RCeD-II, was developed in 2019 (EudraCT 2018-001678-10) [98]. Patients received 10 mg twice daily for 12 weeks, and their symptoms (diarrhea, abdominal pain, weight loss) rapidly improved but reappeared on discontinuation of treatment. Nevertheless, a significant decrease in IELs was not observed, and mucosal improvement was reported for only half of the patients. Moreover, TEAEs were reported for all patients. Limitations of this study include the low patient numbers (n = 4) and no placebo group. Previous case studies on tofacitinib have reported histologic and serologic remission of CeD in a gluten-containing diet (GCD) [110], significant improvement in CeD-related dermatitis herpetiformis (DH) after failed response to first-line treatment (dapsone) [111] and CeD and microscopic colitis clinical and histological remission with and without a GFD [112].
Simultaneously, Ritlecitinib is a JAK3 and TEC (tyrosine kinase expressed in hepatocellular carcinoma) kinase family inhibitor [99]. Its use was approved in the USA and Japan in 2023 for the treatment of severe AA and has long been under investigation for vitiligo, ulcerative colitis and Crohn’s disease. Just recently, a phase 2 clinical trial was developed to evaluate the potential effects of Ritlecitinib in the context of CeD (NCT05636293) [100], which is still in the recruitment phase.

5.6. Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Promoter

CFTR is a protein that functions as a chloride channel on the surface of epithelial cells and, at the intestinal level, it is linked to the regulation of fluid secretion, maintenance of proper mucosal barrier function and protection against pathogens and inflammation. A previous study observed that the P31-43 α-gliadin 13-mer peptide (LGQQQPFPPQQPY) inhibits the function of the CFTR, which leads to epithelial stress, TG2 and IL-15 production, among others [104]. Ivacaftor (Table 3) is a CFTR potentiator commercialized for cystic fibrosis (CF) treatment, which, based on previous findings, could have a therapeutical effect on CeD [113]. While there is no ongoing trial on this topic, a few case studies from CF patients diagnosed with CeD and treated with Ivacaftor reported that an improvement in GI symptoms was only observed after adherence to a GFD and not drug related [102,103].

5.7. Limitations and Strengths of Current Immune Modulators

  • Strengths:
  • Disease target: Immune modulators aim to intervene directly in the autoimmune cascade triggered by gluten exposure, including T-cell activation, cytokine release, and tissue damage. By targeting elements like IL-15, TG2, JAK-STAT signaling, or HLA-DQ2 binding, these therapies address the core immunopathology of CeD rather than just symptoms.
  • Promising early results in specific areas: ZED1227 (TG2 inhibitor) has shown histological improvement in phase 2 trials, marking one of the most promising outcomes in disease-modifying therapy. Tofacitinib and Ritlecitinib (JAK inhibitors) showed a clinical improvement in RCeD-II, suggesting utility in hard-to-treat CeD. AMG 714 provided symptom relief in some cohorts, particularly for patients with non-responsive CeD.
  • Mode of delivery: Most treatments are designed to be delivered orally.
  • Limitations:
  • Safety and tolerability: Tofacitinib was associated with increased visceral adiposity and TEAEs in all patients studied. Long-term safety data for JAK inhibitors and systemic moAbs are still limited, particularly regarding immune suppression and infection risk.
  • Mode of delivery: While some treatments are designed to be delivered orally, others, mainly the IL-15 inhibitors, should be administered through an injection, which may decrease patient acceptability.
  • Site of action: Most immune modulators act downstream, after gliadin peptides have crossed the epithelial barrier and entered the lamina propria, where they can already trigger the autoimmune cascade. This timing allows for disease activation before therapeutic intervention can occur.
  • Symptom relief without mucosal healing: Several immune modulators improved subjective symptoms (e.g., diarrhea, abdominal pain) but failed to correlate with objective markers like IEL counts or villous architecture.

6. Gluten Tolerization

Since exposure to dietary gluten is essential for the development of CeD, many strategies have focused on achieving immune tolerance to GIPs (Table 4). One such approach is Nexvax2, a vaccine whose main active principles are three 15/16-aminoacid-long peptides containing five immunodominant epitopes that interact with the HLA-DQ2.5+ receptor in CD4+ T cells [114,115]. While this treatment was considered safe after various phase I clinical trials (NCT00879749, NCT02528799 and NCT03543540) [114,115,116], a phase 2 study (NCT03644069) [117] concluded that after administration of escalating doses (from 1 µg to 750 µg for 5 weeks and then 900 µg for 11 weeks), Nexvax2 was not capable of reducing the symptoms associated with gluten consumption in CeD in a posterior 10 g gluten challenge at week 14. As a result, the study was terminated by the authors [118].
Another therapy under investigation is TAK-101 (or TIMP-GLIA), consisting of negatively charged gliadin-encapsulating nanoparticles [119]. In a phase 1 (NCT03486990) and phase 2 study (NCT03738475), patients following a GFD were administered ascending doses of TAK-101 (0.1–8 mg/kg up to 650 mg) through two 30 min intravenous infusions on days 1 and 8 within the 2 weeks prior to the start of the challenge and were then subjected to a 14-day oral gluten challenge (12 g/day for 3 days and 6 g/day the remaining time). Results from both trials concluded that TAK-101 was capable of reducing 88% of gliadin-specific IFN-γ-producing cells, and it also significantly prevented the degree of villous atrophy, among other indicators. Moreover, no serious TEAEs were reported. An additional phase 2 clinical trial with alternative doses/dosing strategies is currently under development, but the results are not available yet [120].
KAN-101 consists of a conjugation between a liver-targeting glycosylation signature and an α-gliadin deamidated peptide (KAN0009). It aims to induce gluten tolerance through the promotion of tolerogenic pathways, which then prevent T-cell activation after gluten exposure. In a phase 1 trial (NCT04248855) [121], KAN-101 was administered thrice intravenously in single ascending doses ranging from 0.15 to 1.5 mg/kg, followed by a gluten challenge of 9 g/day for 3 days. During the study, 67–79% of participants receiving KAN-101 reported TEAEs of mild to moderate severity. All doses significantly decreased plasma IL-2 after gluten exposure compared to the placebo group. While a subsequent phase 2 study was terminated by the sponsor (NCT05574010) [122], another phase 2 study, from which results are unavailable, was developed and recently completed (NCT06001177) [123].
Another potential treatment for CeD is TPM502, consisting of nanoparticles coupled with CeD-derived relevant antigens [124]. Although results of the phase 2 clinical trial have not been published yet (NCT05660109) [125], the project sponsor communicated in a press release that the drug was safe and that a decrease in T-cell-derived IL-2 and IFN-γ was observed at the highest dose. A total of 38 patients received two doses of TPM502 (ranging from 0.72 to 7.2 μmol) on days 1 and 15 and were subjected to a gluten challenge (6 g gluten) 7 days after the last infusion [124].

Limitations and Strengths of Current Gluten Tolerization Strategies

  • Strengths:
  • Disease target: These therapies aim to reprogram the immune system to tolerate gluten by directly targeting antigen-specific T-cell responses, rather than broadly suppressing immunity. This makes them highly specific and theoretically curative, addressing the disease’s underlying cause.
  • Promising early results in specific areas: Agents like TAK-101 and KAN-101 have shown clear immunologic effects in clinical trials, including reductions in gliadin-specific IFN-γ-producing T cells, decreased IL-2 levels following gluten challenge and prevention of or reduction in villous atrophy.
  • Safety and tolerability: Most candidates, including TAK-101, KAN-101, TPM502, and Nexvax2, demonstrated acceptable safety profiles, with only mild to moderate TEAEs and no serious safety concerns reported.
  • Limitations:
  • Mode of delivery: Most agents are currently delivered intravenously (e.g., TAK-101, KAN-101, TPM502), which may limit patient acceptability and clinical practicality for long-term treatment or maintenance use.
  • Limited clinical effectiveness and inconsistent outcomes: Nexvax2, despite strong immunological rationale and early safety, failed to reduce symptoms during gluten challenge in a phase 2 trial, so it was discontinued. Results of multiple phase 2 trials have not been made available.
Table 4. Gluten tolerization strategies.
Table 4. Gluten tolerization strategies.
StrategyActive PrincipleNamePhase, Clinical TrialSponsorRef.
Gluten tolerizationVaccine containing 5 immunodominant gluten epitopesNexvax2Phase 1 (NCT00879749, NCT02528799, NCT03543540)
Phase 2–terminated (NCT03644069)
Nexpep Pty Ltd. and ImmusanT, Inc.[114,115,116,117]
Gliadin-encapsulated nanoparticlesTAK-101 or TIMP-GLIAPhase 1 (NCT03486990), Phase 2 (NCT03738475 and NCT04530123)Takeda[119,120,126]
Liver-targeting glycosylation signature conjugated to an α-gliadin deamidated peptideKAN-101Phase 1 (NCT04248855, NCT05574010), Phase 2 (NCT05574010 and NCT06001177)Kanyos Bio, Inc. (Anokion SA)[121,122,123]
Nanoparticles coupled with CeD-derived relevant antigensTPM502Phase 2 (NCT05660109)Topas Therapeutics GmbH[124,125]

7. Probiotics, Prebiotics, Synbiotics and Postbiotics

With CeD being a chronic enteropathy, it is not surprising that the gut microbiota plays a big role in its pathophysiology. It is widely recognized that most patients with CeD suffer from gut dysbiosis, including IP, inflammation of the intestinal epithelium and many gut-related symptoms such as diarrhea, bloating or constipation, even when on a GFD [127]. While more research is still needed to truly understand the numerous pathways in which the gut microbiota influences CeD pathogenesis, some of them have already been elucidated. A healthy intestinal microbiota produces beneficial metabolites crucial in overall body health, such as short-chain fatty acids (SCFAs) or mucous, which are known to help prevent IP [128]. Studies have also shown that the gut microbiota can exert immunomodulatory functions, like an increase in proliferation of IECs, in the production of antimicrobial peptides or regulation of the inflammatory response triggered by Toll-like receptors (TLRs) [128,129]. Moreover, functions of specific taxa have also been uncovered. Several studies have reported that, in CeD, a reduction in the abundance of several bacterial groups such as Bifidobacterium, Lactobacillus or B. longum and an increase in Bacteroides, Staphylococcus, enterobacteria, B. vulgatus or E. coli have frequently been found, when compared to control subjects [130,131,132,133]. It has also been reported that patients with active CeD with or without symptoms, with DH or with recurring symptoms after initiation of a GFD, present clearly differentiated gut microbiota profiles [134,135]. Because of the observed decrease in Bifidobacterium and Lactobacillus, and due to their recognized beneficial effects, many interventional studies target them, as well as other probiotics (live microorganisms that, when administered in adequate amounts, confer a health benefit on the host) [136]. Simultaneously, a lot of research is now focused on prebiotics (a substrate which is selectively utilized by host microorganisms conferring a health benefit), synbiotics (a combination of prebiotics and probiotics) and postbiotics (preparation of inanimate microorganisms and/or their components that confers a health benefit on the host) [136]. Nevertheless, only one study on prebiotics, and none for synbiotics and postbiotics, has focused on the potential effects of these compounds in CeD pathogenesis. The current existing research on pre- and probiotics in relation to CeD is summarized in Table 5.

7.1. Bifidobacterium

One candidate probiotic for CeD is Bifidobacterium infantis natren life start super strain (NLS-SS or Lifestart 2). In an initial interventional clinical trial (NCT01257620) [137], patients received 2 × 109 colony-forming units (CFU)/capsule twice a day alongside 12 g of gluten daily. Results of the study suggested that NLS-SS was safe for consumption, but it did not achieve a significant decrease in serological markers. Even so, GSRS scores slightly improved. In a posterior phase 2 clinical study (NCT03271138) [138], the effect of NLS-SS on CeD patients on a GFD with persistent symptomatology was evaluated. Results showed a decrease in CeD symptoms and differential β-diversity of the stool microbiota between treatment and placebo groups. The presence of GIPs in fecal and urine samples did not vary between groups.
Within the Bifidobacterium genus, the probiotic B. breve BR03 and B632 strains have also been investigated in the context of CeD. As a result of a phase 1 and 2 clinical trial (NCT02244047) [139] with children on a GFD receiving 2 × 109 CFU/daily for 3 months, a significant decrease in serum TNF-α levels was reported, which increased back on treatment discontinuation. The authors also observed a decrease in the Firmicutes/Bacteroidetes ratio and in Actinobacteria, which were normalized after probiotic intake [140,141].
B. longum CECT 7347 or ES1 is another strain that has been widely researched. An initial preclinical study used an animal model of gliadin-induced enteropathy and reported that CECT 7347 administration alongside gluten prevented cellular infiltration, caused a reduction in villi height and width [142] and led to an improvement in gliadin-derived iron deficiency in the liver [143]. Subsequently, an exploratory study with children on a GFD was undertaken [144]. Patients received 109 CFU/daily for 3 months and, as a result, a greater height percentile, decreased peripheral CD3+ T-cell levels and less secretory IgA in stools were achieved. A pilot study was then conducted on non-celiac gluten sensitivity (NCGS) (Maggiore Polyclinic Hospital, study registration number: 1370) [145], where a decrease in all gluten-associated symptoms, except for anxiety, was reported after coadministration of ES1 alongside a GFD. In 2016, another study to assess the effect of ES1 on NCGS was registered (NCT02810301), but its status is unknown [146]. Two recent interventional clinical trials have evaluated the effects of B. longum CECT 7347 in healthy adults with mild to moderate digestive symptoms (NCT05367427) [147] or in patients with diarrhea-predominant IBS (IBS-D) (NCT05339243) [148]. In IBS-D, a decrease in symptom severity and an improvement in stool consistency after ES1 intake were reported.
Along another line, a previous study demonstrated that supplementation with a serine protease inhibitor (Srp) helped decrease gluten-related immunopathology [149]. A study with sensitized NOD/DQ8 tg mice demonstrated that administration of 109 CFU of B. longum NCC2705, which produces a serine protease inhibitor, once a week during a 3-week period, partially prevented IEL infiltration and villi atrophy [150,151]. An interventional trial was started in 2018 for NCGS consisting of administration of two capsules twice daily for four days followed by a 3 g gluten challenge, but the results are not available yet [151].
In an in vitro study using Caco-2 cells, the ability of specific strains of L. fermentum and B. lactis to prevent intestinal damage caused by GIPs was evaluated [152]. The results showed that B. lactis decreased gliadin-induced intestinal permeability and reinforced TJs, as observed by a higher ZO-1 expression.

7.2. Lactobacillus

A probiotic mix composed of Lactobacillus plantarum HEAL9 and L. paracasei 8700:2 was evaluated in an interventional clinical study (NCT03176095) [153,154,155]. The results showed that probiotic administration at 1010 CFU/day for six months did not cause a significant change in tTG2-IgA or –IgG levels between groups, but a decrease in both NK and NKT cells was observed in the probiotic group. A subsequent interventional study on these probiotics is active but not recruiting (NCT04014660) [156].
While no other clinical trials have been undertaken for Lactobacillus and CeD, one study did evaluate the gluten-degrading capacity from Lactobacilli obtained from the small intestine of gluten-fed pigs [157]. Among them, L. ruminis, L. johnsonii, L. amylovorus, and L. salivarius showed the highest gluten degradation activities. Another study researched L. brevis KT16-2 and reported that it displayed in vitro gluten-hydrolyzing abilities [158]. Several other studies have focused on the potential beneficial effects of various Lactobacillus sp. in CeD, including L. casei ATCC 9595, L. plantarum ITM21B, L. paracasei IMPC2.1 and L. fermentum, as well as Bifidobacterium lactis [159,160,161]. Probiotic administration (1010 CFU/4 times a week) to HLA-DQ8 tg gluten-sensitized mice led to an enhanced CD4+ T-cell response, as seen by increased IFN-ɣ and TNF-α expression. Later on, using the same mouse model with addition of cyclo-oxygenase inhibition, the administration of L. casei ATCC 9595 induced recovery of gliadin-derived villi atrophy and a decrease in TNF-α but not IL-2, levels [160].

7.3. Others

A previous study obtained and characterized gluten-hydrolyzing bacteria from wheat sourdough and curd. The Bacillus sp. GS 33, 143, 181 and 188 demonstrated survival properties at pH 2 and in the presence of 0.3% bile acids [162,163]. Similarly, another work identified four Enterococcus faecalis strains, as well as Enterococcus mundtii, Bacillus cereus, Bacillus megaterium and Wickerhamomyces anomalus strains present in fermented sourdoughs, which displayed gluten-hydrolyzing properties [164]. Among these, only E. mundtii QAUSD01 and W. anomalus QAUWA03 showed sufficient pH and bile salt tolerance to be considered for future research as probiotics in CeD.
A novel strain belonging to the Bacteroides vulgatus species (20220303-A2) and present in healthy but not in CeD subjects was isolated. Using a human gut organoid, the authors observed that B. vulgatus 20220303-A2 mitigated the increased cell death, IP and levels of inflammatory cytokines caused by gliadin exposure [165].
Table 5. Probiotic and prebiotic treatments.
Table 5. Probiotic and prebiotic treatments.
Active PrincipleNamePhase, Clinical TrialSponsorRef.
BifidobacteriaB. infantis NLS-SSInterventional (NCT01257620)
Phase 2 (NCT03271138)
Bai, Julio M.D. and
Global Institute of Probiotics
[137,138]
B. breve BR03 and B632Phase 1 and 2 (NCT02244047)University Medical Centre Maribor[139,140,141]
B. longum CECT 7347 (ES1)Interventional (NCT05339243 and NCT05367427)
Phase 2–unknow status (NCT02810301), Maggiore Polyclinic Hospital clinical trial 1370
Vedic Lifesciences Pvt. Ltd.,
Instituto de Investigación Hospital Universitario La Paz, Maggiore Polyclinic Hospital clinical, Exzell Pharma Inc.
[142,143,144,145,146,147,148,166]
B. longum NCC2705Interventional (NCT03775499)Société des Produits Nestlé (SPN)[149,150]
B. lactisIn vitro and in vivo [152,161]
LactobacillusL. plantarum HEAL9 and L. paracasei 8700:2Interventional (NCT03176095 and NCT04014660)Lund University[153,154,155]
L. brevis KT16-2In vitro [158]
L. casei ATCC 9595In vitro and in vivo [159,160,161]
L. plantarum ITM21B, L. paracasei IMPC2.1, L. fermentum,In vitro and in vivo [161]
BacillusBacillus sp. GS 33, 143, 181 and 188In vitro [162,163]
B. vulgatus 20220303-A2Ex vivo [165]
Multispecies probioticsVSL#3Phase 2 (ACTRN12610000630011)Metametrix Clinical Laboratory, Diagnostic Insight and Sigma Pharmaceuticals Pty[167,168]
Multispecies probiotic
Interventional (NCT01699191)University of Bari[169]
PentabiocelInterventional (NCT03857360)Università Politecnica delle Marche[170]
P1: B. breve B632 and BR03, P2: L. plantarum LP14, L. casei subsp. paracasei LPC09, L. rhamnosus LR04In vitro and in vivoPobiotical SpA[171]
Bacteria & yeastE. mundtii QAUSD01 and W. anomalus QAUWA03In vitro [164]
YeastS. boulardii KK1In vivo [172]
HelminthN. americanusPhase 1 (NCT02754609), Phase 1 and 2 (NCT01661933)
Phase 2 (NCT00671138
Princess Alexandra Hospital, The Prince Charles Hospital and James Cook University[173,174,175]
Prebiotics and PostbioticsSynergy 1 (Oligofructose-enriched inulin)Interventional (NCT03064997)Polish Academy of Sciences[176,177,178,179]
Heat-treated B. longum CECT 7347 (HI-ES1)Interventional (NCT05339243 and NCT05367427)
Phase 2–unknow status (NCT02810301), Maggiore Polyclinic Hospital clinical trial 1370
Vedic Lifesciences Pvt. Ltd.,
Instituto de Investigación Hospital Universitario La Paz, Maggiore Polyclinic Hospital clinical, Exzell Pharma Inc.
[142,143,144,145,146,147,148,166]

7.4. Multispecies Probiotics

The VSL#3 probiotic formulation, containing Streptococcus thermophilus, L. plantarum, L. acidophilus, L. casei, L. delbrueckii sp. bulgaricus, Bifidobacterium breve, B. longum and B. infantis, demonstrated a gluten-degrading capacity during a long fermentation of wheat flour. Moreover, addition of VSL#3-digested gliadins achieved smaller zonulin production by rat intestinal epithelial cells (IEC-6), compared to that of regular gliadin, and smaller CeD3+ cell infiltration in the mucosa of biopsies from CeD patients. Nevertheless, in BALB/c mice, VSL#3-digested gliadins were not capable of preventing the reduction in tissue transepithelial electrical resistance (TEER) caused by gliadin, thus leading to increased IP [168]. While many clinical trials have been developed for VSL#3 and a wide array of diseases, including IBS or Asthma, only one has researched its potential effect on CeD pathogenesis (ACTRN12610000630011) [167], and it concluded that VSL#3 intake did not cause a significant change in the fecal microbiota of CeD patients.
A few multispecies probiotics have also been researched for their effects on CeD. A probiotic combination containing two Lactobacillus and three Bifidobacterium sp. (L. casei LMG 101/37 P-17504, L. plantarum CECT 4528, B. animalis subsp. lactis Bi1 LMG P-17502, B. breve Bbr8 LMG P-17501, B. breve Bl10 LMG P-17500) was administered in a clinical trial (NCT01699191) at concentrations ranging from 5 to 10 × 109 CFU/day, for 6 weeks in patients with CeD and IBS, while following a GFD. The results reported a decrease in GSRS scores [169]. The same authors had previously investigated the peptidase activity of 18 commercial probiotic lactobacilli strains (L. casei BGP93; L. delbrueckii subsp. bulgaricus SP5; L. paracasei LPC01 and BGP2; and L. plantarum BGP12, LP27, LP35, LP40, LP47, and SP1), reporting that pooling of 10 of these strains was capable of completely degrading all the GIPs present in a sample using a gastrointestinal simulator [180]. Moreover, when wheat bread was pre-digested with the same probiotic mix and applied to biopsies from CeD patients, IL-2, IL-10 and IFN-ɣ levels remained at baseline.
Another probiotic cocktail named Pentabiocel and composed of L. paracasei 101/37 LMG P-17504, L. plantarum 14D CECT 4528, Bifidobacterium animalis subsp. lactis Bi1 LMG P-17502, B. breve Bbr8 LMG P-17501 and B. breve BL10 LMG P-17500 was evaluated in a clinical trial (NCT03857360) [170]. Children newly diagnosed with CeD took the probiotic for 12 weeks, but no significant differences between the treatment and placebo groups in terms of anti-TG2 IgA Abs, fecal calprotectin or hemoglobin were found. Only the BMI Z-score was significantly higher in the probiotic group.
Two different probiotic combinations from the company Probiotical SpA were assayed both in vitro and in vivo in gliadin-sensitized BALB/c mice [171], showing promising results. Both P1 and P2 formulations, containing ~106 active fluorescent units (AFUs) of B. breve B632 and BR03 (P1) and L. plantarum LP14, L. casei subsp. paracasei LPC09 and L. rhamnosus LR04 (P2), were administered daily to mice, along with gliadin. The results showed that probiotic administration significantly decreased TG2 expression and the levels of inflammatory cytokines in the small intestine down to baseline levels. It also achieved restoration of intestinal morphology and gliadin-mediated IP, as observed by decreased claudin-2 and -15 and increased occludin levels.

7.5. Yeast

In a gluten-sensitized BALB/c mouse model, administration of 108 CFU/day Saccharomyces boulardii KK1 for 7 days alongside gluten partially prevented villi atrophy, as measured using the Vh/Cd ratio, IEL infiltration and both IFN-ɣ and TNF-α production. Nevertheless, it was not capable of decreasing IL-15 and TG2 production. The same study assayed joined administration of L. paracasei DC205 and DC412, which did not display the same preventive effect [172].

7.6. Helminth

Previous studies have shown that helminth infection can attenuate the severity of autoimmune and inflammatory diseases, such as autoimmune liver disease [181], IBS [182], colitis [183,184], multiple sclerosis [185] or allergic rhinitis [186]. Although the exact mechanism for these effects is not very well known, it is hypothesized they could exert beneficial effects through modification of the intestinal microbiota [175]. In order to elucidate the potential role of a helminth infection in the amelioration of CeD, a phase 2 clinical study (NCT00671138) was undertaken consisting of the infection of 20 CeD patients with the hookworm Necator americanus [174]. Unfortunately, the results showed no improvement in the pathology. In a subsequent phase 1 and 2 study (NCT01661933) [175], infection with N. americanus in CeD patients followed by a gluten challenge (10–50 mg/day) was associated with increased abundance of species belonging to the Bacteroides phylum and with an increase in microbiota richness and diversity. Although biopsies were collected, the study did not report data on gluten-related biomarkers such as villous atrophy. An ensuing phase 1 study (NCT02754609) reported similar results to the initial one, where infection with the hookworm did not have a positive impact on reducing gluten-associated symptomatology in CeD patients but also decreased their quality of life [173].

7.7. Prebiotics, Synbiotics and Postbiotics

An untapped research area in regard to CeD involves prebiotics, symbiotics and postbiotics. Since all of them target the intestinal microbiota, and it is acknowledged that a lot of investigation is still needed to better understand the specific, and thus modifiable, pathways in which the gut microbiota prevents, promotes or alleviates CeD, it is only logical that very few projects have been targeted to elucidate the role of pre-, syn- and postbiotics on CeD pathogenesis. Currently, there are only two projects researching the effect of prebiotic intake on CeD. As mentioned before, decreased amounts of Lactobacillus and Bifidobacterium are commonly found in both active and treated CeD. To this end, the goal of one study was to increase such beneficial bacteria through the administration of the oligofructose-enriched inulin probiotic Sinergy 1. A clinical trial was conducted in CeD children following a GFD (NCT03064997) [177], which consisted of 1 intake/day of a 10 g Synergy 1 dose for 3 months. Results from the study showed that prebiotic supplementation did not cause side effects and significantly increased Bifidobacterium, acetate and butyrate fecal levels [179] and glutamine plasma concentration [178]. Conversely, Synergy 1 did not increase barrier integrity, with the limitation that most study subjects had normal IP values at study initiation [187]. Moreover, a decrease in serum hepcidin, which is involved in intestinal iron absorption, was observed [176].
Simultaneously, heat-treated B. longum CECT 7347 (HT-ES1) has also been researched in two clinical trials (while compared to its active counterpart) focusing on IBS (NCT05339243) [148] and healthy adults with moderate gastrointestinal symptoms (NCT05367427) [147]. For the latter, HT-ES1 intake did not have an effect on GSRS scores, but it caused a decrease in total and non-high-density lipoprotein (HDL) cholesterol and an increase in the butyrate-producing Faecalibacterium genera. In IBS, both an improvement in stool consistency and a decrease in symptom severity were reported as a result of HT-ES1 intake.

7.8. Fecal Microbiota Transplantation (FMT)

Along the same line as all aforementioned strategies, FMT aims to modify the intestinal microbial composition to prevent dysbiosis and its associated detrimental effects. While not many studies have aimed to uncover the potential role of FMT in CeD pathogenesis, one case study did report that, after performing FMT on a patient with RCeD-II due to persistent Clostridium difficile infection (CDI), remission of CeD symptoms and recovery of intestinal morphology were observed [188]. While FMT is currently only recommended for CDI, an existing clinical trial (NCT04014413) is currently recruiting patients with intestinal diseases, including CeD, to evaluate the safety and efficacy of FMT [189].

7.9. Limitations and Strengths of Current Probiotic, Prebiotic, Synbiotic and Postbiotic Strategies

  • Strengths:
  • Disease target: CeD is increasingly recognized as a condition involving gut dysbiosis, even in patients adhering to a GFD. The microbial imbalance is associated with inflammation, increased IP and persistent symptoms, making microbiota-targeted therapies highly relevant.
  • Uncertain mechanisms of action: A key limitation of probiotic use in CeD is the lack of clear evidence regarding the exact mechanism of action. While some studies suggest potential benefits, the specific pathways through which probiotics exert these effects in the context of gluten-induced autoimmunity remain largely speculative. This uncertainty hampers the development of targeted formulations and limits confidence in their therapeutic efficacy.
  • Broad immunomodulatory potential: Certain strains (e.g., B. breve, B. longum, L. casei) have shown abilities to decrease pro-inflammatory cytokines, improve intestinal barrier function, modulate immune cell populations or increase beneficial metabolites like SCFA.
  • Symptom relief in some studies: Several probiotic combinations (e.g., B. infantis NLS-SS, ES1 or some multispecies formulations) showed improvements in GI symptoms, including bloating, stool consistency, and GSRS scores, especially in patients on a GFD with persistent symptomatology.
  • Safety and tolerability: Across most trials, no serious TEAEs were reported. Even novel combinations or postbiotics (e.g., heat-treated ES1) were well tolerated, including in children.
  • Complementary to existing therapies: Generally, these strategies do not aim to replace a GFD but rather enhance its effectiveness by improving symptom control, supporting mucosal healing, and addressing non-responsive or partially responsive CeD.
  • Mode of delivery: Most treatments are designed to be delivered orally.
  • Limitations:
  • Limited clinical efficacy and inconsistent outcomes: Many interventions showed no significant changes in serological markers, histological damage or IP. Some promising in vitro or in vivo studies failed to translate into measurable clinical benefits in humans (e.g., VSL#3, Pentabiocel, S. boulardii).
  • Uncertain causality in pathogenesis: Similar to intestinal permeability modulators, it remains unclear whether increased gut dysbiosis is a cause or a consequence of CeD. Hence, the extent of the effect of improvement in the gut microbiota profile in CeD symptomatology and disease biomarkers is unclear.
  • Few rigorous or large-scale trials: Most trials were small, exploratory or short in duration, with few phase 2 studies. Critical endpoints like villous atrophy, IEL infiltration and anti-TG2 levels are rarely assessed.

8. Nutraceuticals

The term nutraceuticals encompasses bioactive compounds from natural or food sources with potential health benefits [190]. Some of these functional compounds include phytochemicals (e.g., polyphenols, terpenoids, alkaloids, etc.), fatty acids, vitamins, minerals or amino acids, among others. Nutraceuticals can exert functions like gliadin sequestration, which prevents its degradation and the subsequent formation of GIPs, a decrease in IP, inhibition of TG2 enzyme or modulation of the gut microbiota, as well as display anti-inflammatory, antioxidant and immunoregulatory effects [191]. Nevertheless, more studies are still needed to elucidate the pathways through which nutraceuticals can exert beneficial effects in the pathophysiology of CeD. A list of studies regarding nutraceuticals and CeD can be found in Table 6 (polyphenol-related strategies) and Table 7 (other bioactives).

8.1. Polyphenols

Polyphenols are a type of phytochemical structurally related to phenolic compounds, which possess strong antioxidant properties. Polyphenols can be mainly classified into flavonoids and non-flavonoids. Within the flavonoids, relevant subcategories are flavanols, anthocyanidins, anthocyanins, isoflavones, flavones, flavonols, flavanones and flavanonols. In the non-flavonoid family, relevant sub-types are phenolic acids, stilbenes, lignans and tannins [192]. In the context of CeD, polyphenols can exert beneficial effects through multiple mechanisms. One of them is gliadin sequestration [193], where polyphenols have been hypothesized to be capable of agglomerating and forming 3D structures, which can complex to gliadin and prevent its crossing into the lamina propria and the immune reaction that ensues.
A study on green tea extract, which contains high amounts of polyphenols, confirmed that it was capable of attaching to gliadin in vitro, as seen by an increase in turbidity [194], and, thus, inhibit gliadin digestion. Moreover, in Caco-2 cells, green tea extract’s complexation with gliadin decreased IP and IL-6 and IL-8 secretion. In another study on green tea polyphenols, mainly (-)-epigallocatechin (EGC) and (-)-epigallocatechin-3-gallate (ECGC), it was shown that ECGC, but not EGC, was capable of reducing the in vitro digestion of a gluten + wheat starch mixture, which was attributed to the polyphenol’s crosslinking to both molecules [195].
In a project that aimed to uncover the mechanisms by which polyphenols can have a positive impact in reducing CeD symptomatology, the interaction between the 32-mer GIP and dietary polyphenols (catechin, procyanidin B3, procyanidin C2, EGC and ECGC) was evaluated [196]. The authors reported that procyanidin B3, but, even more so, C2, significantly prevented transport of the 32-mer peptide from the apical to the basolateral side of the intestine using Caco-2 cells. Along the same line, another study evaluated the capacity of polyphenols sourced from artichoke and green tea leaves, as well as cranberries and apples, to complex with gliadin and decrease gliadin-derived immunogenicity and allergenicity [197]. It was found that all but green tea leaves’ polyphenols formed insoluble complexes with gliadin and that cranberry and artichoke extracts did partially prevent basophil degranulation. The same pattern was observed in a study using Raman spectroscopy, which reported conformational changes in gliadin after interaction with the anthocyanins malvin, kuromanin and callistephin [198]; the anthocyanidin cyanidin; the coumarin-derivative 3-ethoxycarbonylcoumarin (3-EcC) [199]; and the flavonoid quercetin [200]. This effect was not observed for the anthocyanins pelargonin, oenin and cyanin [200]. Another study on quercetin [201] reported decreased TG2 and increased glutathione (GSH) and TEER after quercetin treatment alongside pepsin-trypsin-digested gliadin (PT-G) in Caco-2 cells. Quercetin intake in C57BL/6N gliadin-sensitized mice caused a small prevention of the gliadin-associated decrease in the Vh/Cd ratio and a significant decrease in nitric oxide (NO) and TG2 levels but did not affect GSH. Quercetin decreased the % of T-regulatory cells, the T-helper (Th)1/Th2 ratio and many inflammatory cytokines (IL-1β, TNF-α, IL-2 and IL-12p70). An increase in beneficial bacteria such as B. longum was also noted.
An alternative study reported that three procyanidins naturally present in grape seeds (i.e., B3, B6 and T2) were all capable of molecularly binding to the 32-mer peptide and that the B6 and T2 compounds strongly prevented 32-mer transport from the intestinal lumen to the lamina propria [202]. Similarly, a project researching the impact of peanut skin proanthocyanidins (PSPc) on CeD showed that treatment of PT–G-sensitized Caco-2 cells with PSPc decreased reactive oxygen species (ROS), TG2, IL-1β, IL-6 and TNF-α levels, especially at high concentrations, indicating a strong antioxidant and anti-inflammatory effect [203]. Moreover, addition of PSPc also increased occludin expression, a marker of intestinal integrity. A very similar experiment was performed using anthocyanin-rich sour cherry extract (AC), where it was reported that AC prevented the gliadin-induced IP. This was concluded after observation that AC intake led to an increase in TEER values, compared to those in the Cacao-2 cells exclusively treated with gliadin, and by a decrease in the permeability coefficient of Lucifer Yellow, a small paracellular marker [204]. Addition of AC alongside gliadin also limited apical TNF-α, IFN-ɣ and IL-8 levels. While ROS levels decreased in the AC + gliadin group, compared to the gliadin-only group, these were still significantly higher than in the control samples.
Propolis is a resin-like substance produced by honeybees, which contains high amounts of polyphenols. The effects of propolis supplementation ex vivo in peripheral blood mononuclear cells (PBMCs) and fecal samples, both from CeD donors, were also evaluated. Culture of PBMCs with ethanolic extract of propolis (EEP) significantly reduced the levels of the oxidative stress markers NO and inducible nitric oxide synthase (iNOS) enzyme and the pro-inflammatory cytokine IFN-ɣ, simultaneously with an increase in the anti-inflammatory cytokine IL-10 [205]. Moreover, it also down-regulated the expression of the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) and phosphorylated STAT 3 (pSTAT-3) transcription factors, which can be involved in pro-inflammatory processes. Culture of fecal material from CeD patients with propolis significantly increased the levels of SCFA and of beneficial bacteria such as Bifidobacterium [206].
A study on the protective role of curcumin in vitro using the HCT-116 and -29 human intestinal cell lines reported that pre-treatment with curcumin before gliadin exposure prevented an increase in inflammatory and oxidative markers, such as advanced oxidation protein products (AOPPs), Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase (NOX) and myeloperoxidase (MPO), as well as the levels of multiple inflammatory cytokines (IL-1β, IFN-ɣ, TNF-α…) [207]. A similar study showed both in vitro and in vivo that exposure/administration of resveratrol to gliadin-sensitized Caco-2 and C57BL/6N mice caused a decrease in inflammatory malondialdehyde (MDA) and an increase in activity of the anti-inflammatory superoxide dismutase (SOP) enzyme, as well as of GSH levels [208]. Supplementation with resveratrol also led to a significant decrease in TG2, iNOS and cyclooxygenase-2 (COX-2) expression, an increase in occludin activity and a decrease in IP.

8.2. Combination of Polyphenols and Other Bioactives

Some studies have also focused on potential synergistic effects between multiple bioactives. The anti-inflammatory and antioxidant effects of the carotenoid lycopene and the polyphenols quercetin and tyrosol were evaluated in an in vitro model of gliadin and IFN-γ-stimulated RAW 264.7 macrophages [209]. All treatments significantly decreased nitrite (NO2) and prostaglandin (PGE2) production, iNOS and COX-2 expression and NF-κB, interferon regulatory factor -1(IRF-1) and STAT-1α activation.
Regarding this topic, one food, which naturally presents a combination of health-promoting compounds, is cocoa. The beneficial effects of cocoa as a whole [210,211], as well as of cocoa-derived polyphenols [212,213], theobromine [214,215], caffeine [216] and fiber [217], have been studied individually in health and disease. To this end, procyanidin B2-enriched cocoa extract (PCE) was investigated for its potential preventive role in CeD using an in vitro model of gliadin-sensitized Caco-2 cells [218].
Table 6. Polyphenol-containing strategies in CeD.
Table 6. Polyphenol-containing strategies in CeD.
StrategyActive PrincipleSourceMechanism of ActionPhase *Ref.
PolyphenolsPolyphenolsGreen tea extractGliadin sequestrationIn vitro[194]
(-)-epigallocatechin and (-)-epigallocatechin-3-gallateGreen teaGliadin sequestrationIn vitro[195]
Catechin, Procyanidin B3, Procyanidin C2, Epigallocatechin and Epigallocatechin Gallate Gliadin sequestrationIn vitro[196]
PolyphenolsArtichoke leaves, cranberries, apples, green tea leavesGliadin sequestrationIn vitro[197]
Kuromanin, Callistephin, Oenin, Cyanin, Pelargonin, Malvin (Anthocyanins) Gliadin sequestrationIn vitro[198]
Procyanidin B3, B6 and T2 (Tannins)Grape seedGliadin sequestrationIn vitro[202]
Quercetin (Flavonoid) Gliadin sequestrationIn vitro[200]
Cyanidin (Anthocyanidin)CoumarinGliadin sequestrationIn vitro[199]
ProanthocyanidinsPeanut skinGliadin sequestrationIn vitro[203]
AnthocyaninsSour cherry extractImmune and intestinal permeability modulationIn vitro[204]
FlavonoidsPropolis dry extract ESIT 12®Immune and gut microbiota modulationEx vivo[205,206]
Curcumin (Diarylheptanoid)Turmeric (Curcuma longa)Immune modulationIn vitro[207]
Resveratrol (Stilbene)Grapes, red wineImmune, intestinal permeability and gut microbial modulationIn vitro and in vivo[208]
Combination of polyphenols & other bioactivesLycopene (carotenoid), quercetin (flavonoid) and tyrosol (phenolic alcohol)Tomatoes/Onions, extra virgin olive oil (EVOO), broccoli/white wine, EVOOImmune modulationIn vitro[209]
Procyanidin B2, theobromine, caffeine (alkaloids)Cocoa extractImmune modulationIn vitro[210]
Epicatechin (flavanol), theobromine (alkaloid)ChocolateImmune modulationPilot study, ECCEL2 nº 43.18: 4, 2018[211]
* ECCEL2 = Ethics Committee of the Comitato Etico Lazio 2.
The results showed that both theobromine and caffeine, individually, significantly decreased TG2 levels (40–60%) and that the reduction was even higher when using the PCE (77% reduction). The addition of PCE to the sensitized model led to a significant decrease in many inflammatory biomarkers, including TG2, COX-2, IL-15, IL-1β, IL-6 and IL-8 levels. Along the same line, another study evaluated the lymphocyte-to-monocyte (LMR) and the platelet-to-lymphocyte (PLR) ratios in CeD patients with habitual dark chocolate consumption. The results showed that they displayed a higher LMR compared to non-chocolate-consuming controls [219]. Since an association between low LMR and autoimmune disease has previously been reported [220], this finding could indicate a beneficial role of chocolate consumption in CeD. Another preclinical study using DQ8-Dd-villin-IL-15tg mice with predisposition to CeD reported that cocoa supplementation alongside gliadin managed to prevent the increase in anti-gliadin and anti-TG2 Abs, the latter only in male animals, where a decrease in fecal humidity was also found. Cocoa intake also prevented an increase in villi width, induced a change in β-diversity and an increase in beneficial probiotic species such as Akkermansia muciniphila or the decrease in the opportunistic genera Chlamydia, indicating the potential multispectral effect of cocoa intake in the context of CeD [221,222].

8.3. Vitamins

Another group of bioactive compounds is vitamins. These have long been studied in relation to CeD, since Vitamin B deficiency is a common trait of this disease. As far back as 1944, one study reported an improvement in weight gain and height in children with CeD after supplementation with Vitamin B complex [223]. Many studies have researched the vitamin status of CeD patients, mostly reporting deficiencies in folate [224,225], vitamin D and A, and a tendency to lower B12 [226], which tends to normalize after gluten exclusion from the diet. Nevertheless, only few works have investigated the potential effect of vitamin supplementation on CeD pathogenesis. One study evaluated Vitamin D3’s (1,25-dihydroxy vitamin D3, VD3) TJs modulation properties in both PT-G-sensitized Caco-2 cells and BALB/c mice [227]. The results of simultaneous application of PT-G and VD3 for 7 days to Caco-2 cells showed an increase in TEER, occludin, claudin-1 and ZO-1 values and a decrease in zonulin expression. In mice, a decrease in serum FITC-dextran 4000 (FD-4) flux after VD3 intake also indicated an improvement in the integrity of the intestinal barrier, which was supported by increases in the relative levels of occludin, claudin-1 and ZO-1. In another study, CeD seronegative patients on a GFD were supplemented with 400 mg of vitamin E, twice daily, for a period of 3 months (KBET/174/B/2013) [228]. The authors reported that, although no significant difference in Vitamin E levels was found before and after supplementation, serum NO and alanine aminotransferase (ALT) levels, as well as glutathione peroxidase-3 (GPx3) activity, did decrease, while Vitamin D levels increased, altogether indicating a decrease in oxidative stress and inflammation.

8.4. Fatty Acids

Polyunsaturated fatty acids (PUFAs), especially omega-3 (n-3) fatty acids (FAs), have many recognized health benefits, mainly due to their capacity to exert anti-inflammatory effects through the regulation of T cells, macrophages and other immune cells [229]. While multiple studies have evaluated the characteristic FA levels in CeD, only two of them have focused on the effect of FA administration in CeD pathophysiology. A characteristic feature of CeD is villi atrophy, mainly driven by the acquisition of a cytotoxic phenotype by IELs. Since this is mediated by the release of arachidonic acid (AA), which is dependent on cytosolic phospholipase A2 (cPLA2), one study aimed to assess if the n-3 long-chain PUFA docosahexanoic acid (DHA) was able to down-regulate the release of AAc by IECs [230]. Results from the study showed that the addition of DHA to a PT-G-sensitized-Caco2 monolayer effectively decreased the release of multiple inflammatory markers: cPLA2, AAc, COX-2 and PGE2. This prevented the establishment of an inflammatory milieu, as observed by a decreased secretion of the pro-inflammatory cytokine IL-8.
In a clinical study (Ethics Committee of the Assis Gurgacz University Center number 2,315,783), CeD patients followed a GFD supplemented with fish oil or the latter and exercise for 12 weeks [231]. The results showed a decrease in C-reactive protein and IL-6 in the group that included exercise.

8.5. Terpenes

Terpenes are a type of phytochemical widely found in plants, especially in essential oils but also in some animals and microorganisms. The oil from black cumin seed (Nigella sativa, NS) was studied for its potential beneficial effect in the context of RCeD, due to its high content of thymoquinone and monoterpenes [232]. The authors reported that the group with NS intake alongside a GFD in a 450 mg dose daily achieved a significantly higher histological remission than the group that exclusively followed a GFD without supplementation.

8.6. Glucosinolates/Isothiocyanates

Glucosinolates are sulfur- and nitrogen-containing compounds present in plants from the Brassicaceae family. At consumption, glucoraphanin, the main glucosinolate found in broccoli, is transformed to sulforaphane, an isothyocyanate-type compound with many reported health benefits but low stability. Hence, a study was performed in a Caco-2 model of inflammation, oxidation and gliadin sensitization to analyze the potential effects of glucoraphanin intake [233]. The results showed that the bioactive compound sulforaphane prevented the secretion of monocyte chemoattractant protein-1 (MCP-1), C_X_C motif chemokine ligand 10 (CXCL10) and IL-8, typically upregulated during inflammation. Moreover, sulforaphane modulated the activity of NF-κB but was not capable of preventing a decrease in TEER values.

8.7. Algae

The effect of algae as an adjuvant to the diet is being investigated for multiple diseases, including cardiovascular diseases like obesity or diabetes [234]. One study analyzed the effect of the addition of Chlorella pyrenoidosa in a gut simulator fed with the feces of either a healthy subject or a CeD patient following a GFD. The results showed a difference in β-diversity abundance between the CeD and healthy samples [235]. The addition of C. pyrenoidosa caused an increase in the CeD sample of the generally recognized as beneficial genera Faecalibacterium, Bifidobacterium and Megasphaera and a decrease in the abundance of the Enterobacteriaceae family, which contains opportunistic bacteria. Moreover, C. pyrenoidosa supplementation was linked to an increase in butyrate and propionate. A strong limitation of this study is that only one sample was used for each group (n = 1).

8.8. Limitations and Strengths of Current Nutraceutical Strategies

  • Strengths:
  • Wide range of bioactive effects: Nutraceuticals offer multiple mechanisms of action relevant to CeD pathogenesis, including gliadin degradation or sequestration, inhibition of TG2 enzyme activity, modulation of IP, anti-inflammatory and antioxidant effects and gut microbiota modulation.
  • Safety and tolerability: Since nutraceuticals are naturally occurring, they are generally recognized as safe and are well tolerated, even at higher doses.
  • Complementary to existing therapies: Generally, these strategies do not aim to replace a GFD but rather enhance its effectiveness by improving symptom control, supporting mucosal healing and decreasing gluten-derived inflammation.
  • Mode of delivery: Most treatments are designed to be delivered orally.
  • Limitations:
  • Predominance of in vitro and preclinical evidence: Most data originated from in vitro and animal models, with only a few human trials.
  • Low bioavailability of certain compounds: Some nutraceuticals suffer from poor stability or absorption, limiting their in vivo efficacy unless specially formulated. This remains a challenge for translation into real-world therapies.
  • Confounding from GFD adherence: Several studies are conducted in patients already on a GFD, making it difficult to isolate the effect of nutraceutical intervention from the baseline benefits of dietary gluten exclusion.
  • Heterogeneity of compounds and protocols: The category “nutraceuticals” includes vastly diverse compounds, delivery forms, and dosages, making standardization, reproducibility, and comparison between studies difficult.
Table 7. Non-polyphenol nutraceuticals in CeD.
Table 7. Non-polyphenol nutraceuticals in CeD.
StrategyActive PrincipleSourceMechanism of ActionPhase *Ref.
VitaminsVitamin D, 1,25-dihydroxy vitamin D3 Immune and intestinal permeability modulationIn vitro, in vivo, clinical trial, CIEC 53,043,469/050.04–52[226,227]
Fatty acidsDHA Immune and intestinal permeability modulationIn vitro[230]
DHA and EPAFish Oil Clinical trial, CEP-FAG 2,315,783[231]
TerpenesThymoquinone and monoterpenesBlack cumin (N. sativa) oilImmune modulationClinical trial, ECCM-UB[232]
Glucosinolates/ isothiocyanatesGlucoraphanin/SulforaphaneBroccoli sprouts (B. oleracea var. italica
Planck)
Immune and intestinal permeability modulationIn vitro[233]
AlgaeC. pyrenoidosaChlorella sp. (algae)Gut microbial modulationEx vivo[234]
* CIEC = Clinical investigations ethics committee, ECCM-UB = Ethics Committee of College of Medicine/ University of Baghdad CEP-FAG = Ethics Committee of the Assis Gurgacz University Center.

9. Food Modifications

Multiple strategies have aimed to prevent CeD through alterations in gluten-containing foods. To this end, a wide range of approaches have been essayed, from gluten modification to decrease its toxicity to its removal from naturally gluten-containing foodstuffs, like wheat flour, or its processed-derivatives like bread or pasta.

9.1. Bacterial and Enzymatic Degradation of Gluten-Contaning Foodstuffs

In order to obtain gluten-free foods, a frequent method used has been the pre-treatment of wheat with either gluten-degrading enzymes or bacteria [236]. Hence, some of the microorganisms and enzymes described in Section 2 and Section 7 have been redirected to food-processing purposes, like AN-PEP, which has been reportedly used for the obtention of gluten-free beers [237] and doughs [238] or papain and alcalase [236], which displayed gluten-hydrolyzing activity while maintaining optimal rheological properties in bread. Two other commercially available proteases, Flavourzyme (A. oryzae-containing) and Protamex (Bacillus lichenformis and B. amyloliquefaciens), were evaluated for their gluten-digesting properties in both wheat flour and vital wheat gluten, demonstrating up to 30% of gluten hydrolysis for Flavourzyme and 15% for Protamex [239]. A subsequent study evaluating the gluten-degrading capacity of three alternative preparations, Flavourzyme 100L, Bioprase SP-20FG (B. lichenformis) and Thermoase PC10F (B. thermoproteolyticus), validated the prior results, with Flavourzyme again displaying the highest gluten-degrading activity and achieving over 40% hydrolyzation of wheat gluten after 500 min [240]. ASP, DPPIV and EP-B2, alone or together, successfully degraded from 22 to 98% of the gluten present in whole wheat [23]. In another work, CeD patients on a GFD received either regular wheat flour or lactobacilli and fungal protease-pre-digested wheat gluten [241]. Lactobacilli used included L. alimentarius 15M, L. brevis 14G, L. sanfranciscensis 7A and L. hilgardii, while fungi were A. oryzae and A. niger. The results showed that hydrolyzed wheat did not cause an increase in IFN-ɣ levels in CeD patients, contrary to regular wheat. Another study reported that exposure of Caco-2 cells to L. paracasei CBA L74-fermented rice flour alongside the 33-mer (P31–43) peptide from α-gliadin significantly reduced its entrance into the basolateral side [242]. Other studies have also used such microorganisms to improve quality, nutritional or rheological properties from gluten-free foods [243,244].

9.2. Gluten Genetic Modifications

An alternative strategy that has been investigated is that of gluten modification before its use to generate processed foods. This can be achieved through mutation breeding, γ-irradiation, line deletion, transgenic modifications, gene silencing or targeted mutagenesis [245]. Along this line, one approach to reduce gluten content in wheat and other cereals has been through breeding wheat cultivars with naturally reduced gluten amounts or which display lower immunogenicity [246]. One study reported that deletion of the Gli-2 loci in wheat, which encodes for 49 α-gliadin expressing genes, removed the presence of 33-mer [247]. Parallelly, another project obtained hypoimmunogenic gluten through gene editing techniques [248]. The authors designed six single-guide RNA sequences to target both α- and γ-gliadins, which contain immunoreactive epitopes, through the CRISPR/Cas9 technology. The results showed increased efficiency and precision as compared to that obtained with γ-irradiation, a method which is accepted in the European Union, contrary to genetic modification [249]. Gene silencing is a post-transcriptional mechanism achieved through RNA interference, where the complementary messenger RNA is degraded, thus preventing the production of targeted proteins [250]. This process was used to down-regulate the expression of gliadins from bread wheat. The results obtained showed a decreased T-cell response from cells obtained from CeD patients’ damaged intestinal tissue [251].

9.3. Gluten Transamidation

Since one of the main steps in catalyzing the gluten-derived immune response is deamidation of the glutamine residues present in gliadins, some efforts have focused on preventing this process. Transamidation, contrary to deamidation where the amide group from glutamine is removed, does not lead to the elimination of the amide but its substitution with an amine group. To this end, for this process to occur, both an amine donor, such as a lysine, and a transamidating enzyme, i.e., a microbial or animal transglutaminase, are needed. Several studies have demonstrated that transamidation of gliadin or flour leads to decreased immunogenicity, observed by a smaller IFN-γ response by CeD-biopsy-derived gliadin-specific T-cell lines, as well as a decreased binding affinity to HLA-DQ2 by transamidated gliadins [252]. Similar results have been observed in Caco-2 cells, bone-marrow-derived dendritic cells and BALB/c mice, where transamidated gliadin achieved reduced intestinal damage and permeability, as well as decreased levels of pro-inflammatory cytokines, including IL-6, IL-8 and TNF-α [250]. Moreover, a clinical study with CeD patients in a GFD, whom either consumed regular or transamidated gliadin (Ethical Committee of San G. Moscati Hospital OsSc registry n.06/09, trial n.234, and Ethical Committee ASL Salerno, OsSc registry n.318; trial n.118/AA.GG), reported significant decreases in clinical relapse, intestinal permeability, anti-transglutaminase IgA levels, Vh/Cd and IFN-γ expression in the group with transamidated gliadin intake [253]. As a result, a few studies have investigated the production of bread with decreased immunoreactivity through the use of microbial transglutaminase [254,255].

9.4. The Gluten Friendly™ Technology

Multiple studies have been performed using the registered Gluten Friendly (GF)™ technology, which aims to obtain gluten-detoxified flours through structural modifications of gluten proteins, which then do not trigger the regular gluten-derived immune reaction [256]. Briefly, this technology mixes physical and chemical modifications of the wheat kernels through cycles of controlled heat, humidity and resting, which then induces conformational changes that make the gliadins potentially unrecognizable by the immune system [257]. A clinical trial was developed, which consisted of the intake of 3 or 6 g of GF/day (or none in the placebo group) for 12 weeks (NCT03137862) [258]. While the results of the study showed similar Vh/Cd or %IELs infiltration between the three groups, a significant limitation of the study is the lack of a positive control group. While a simultaneous clinical trial, consisting of the administration of none, low (GF bread ~1.5 g), medium (3 g) or high (6 g) daily gluten amounts for 2 weeks (NCT03168490) [259], was completed, results have not been made available.

9.5. Limitations and Strengths of Current Gluten-Containing Food Modifications

  • Strengths:
  • Source targeting: These strategies aim to prevent the immune response from initiating by modifying gluten before ingestion, with clear health and economic benefits.
  • Potential to improve quality of life: By detoxifying gluten in common foods like bread and pasta, these methods could enhance dietary flexibility, reduce cross-contamination risks, and improve patient adherence and satisfaction.
  • Cost-effective and scalable: Compared to pharmaceutical development, food-processing interventions are relatively inexpensive, can be integrated into existing manufacturing systems, and scale efficiently, making them a viable solution in broader populations and low-resource settings.
  • Safety and tolerability: Most studies report that modified gluten products are well tolerated in CeD patients, with no major adverse effects or symptom relapses when consumed under controlled conditions. This supports their potential for safe dietary integration, although more safety studies should be performed.
  • Mode of delivery: By incorporating modified gluten directly into commonly consumed foods (like bread, pasta, or flour), these strategies offer a non-invasive, patient-friendly alternative to pills or injections. This enhances patient acceptability and compliance, especially for long-term management.
  • Limitations:
  • Predominance of preclinical evidence: Most data originate from in vitro and animal models, with only a few human trials. Many promising findings lack validation in clinical settings, especially with well-defined CeD endpoints like villous atrophy or IEL counts.
  • Incomplete or inconsistent detoxification: Not all methods fully remove or neutralize gluten peptides, which may still trigger an immune response in sensitive individuals. Moreover, gluten modification methods may differ in effectiveness depending on the food matrix, processing conditions, and enzyme source, making it challenging to standardize protocols for broad use.
  • Mode of delivery: Although modified gluten foods offer a convenient route of delivery, they still require CeD patients to consume specialized or alternative food products. This contrasts with pharmaceutical approaches (like enzymes or immune modulators), which may one day allow patients to eat unmodified gluten-containing foods, potentially offering greater freedom and social normalization.
  • Regulatory and consumer acceptance barriers: Strategies involving genetic modification (e.g., CRISPR/Cas9, RNA interference) face regulatory hurdles in many regions (notably the EU), as well as potential consumer resistance.
  • Limited clinical validation: Most strategies have not progressed beyond small trials or preclinical models, leaving efficacy in real-world patients uncertain.

10. Conclusions

Despite decades of research and the rising global prevalence of CeD, the therapeutic landscape remains remarkably limited. A strict GFD is still the only approved treatment, yet it is both socially restrictive and clinically insufficient for many patients. Numerous alternative strategies, ranging from gluten-degrading enzymes and permeability modulators to immune tolerance therapies and microbiota-targeted interventions, have been explored. However, none have yet demonstrated consistent, reproducible efficacy or proven to be viable, long-term substitutes for a GFD.
What emerges from this study is a sense of insufficiency and urgency. While a lot of work is being conducted and has been carried out on the topic for the last two to three decades, a feasible treatment for CeD still remains out of reach. This brings leads to the question of where the problem lies: Have sufficient efforts been targeted on CeD? Is there a lack of investment? Are we focusing on the wrong pathways? Should we try to avoid the current fragmentation in therapeutic efforts? A pattern that seems to be repeating itself is that, initially, many approaches show promise in early-stage trials or preclinical models but then fail to translate into substantial, lasting benefits in humans. Treatments like enzyme therapies, immune modulators and tolerization vaccines have yielded inconsistent or underperforming outcomes, often hampered by issues of low bioavailability, modest effect sizes or safety concerns.
At the core of this challenge is the complex, multifactorial nature of CeD, which involves not only adaptive and innate immune responses but also gut barrier dysfunction, environmental triggers and the gut microbiome alteration. This complexity makes it unlikely that a single therapeutic strategy will offer a universal solution. In conclusion, although scientific advances have greatly expanded our understanding of CeD pathophysiology and therapeutic targets, we are still far from delivering a practical, scalable treatment. Hence, despite all the efforts focused on this research field, the only current treatment for CD is not any of the abovementioned strategies, but it still remains a lifelong string adherence to a GFD. This underscores the urgent need for better-designed trials, more collaborative efforts, investment and patient-centered outcomes, as well as focus on multimodal therapies that move beyond theoretical benefit and towards real, meaningful improvements in daily life for patients with CeD.

Author Contributions

Conceptualization, data curation, writing—original draft preparation, writing—review and editing, M.G.-G. and F.J.P.-C.; supervision, F.J.P.-C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. The authors want to thank the Institute of Nutrition and Food Safety (INSA) Maria de Maeztu Unit of Excellence grant (CEX2021-001234-M) funded by MICIN/AEI/FEDER, UE as well as the University of Barcelona Predoctoral Researcher Recruitment Program (PREDOCS-UB 2022).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
3-EcC3-ethoxycarbonylcoumarin
AAAlopecia areata
AAcArachidonic acid
AbsAntibodies
AFUActive fluorescent units
AGYImmunoglobulin Y antibody from the egg yolk
ALTAlanine aminotransferase
AOPPAdvanced oxidation protein product
APCsAntigen presenting cells
ASPAspergillus niger
BFBromelain and ficin
CCLCC motif chemokine ligand
CCRCC motif chemokine receptor
CeDCeliac disease
CFCystic fibrosis
CFTRCystic fibrosis transmembrane conductance regulator
CFUColony-forming units
COX-2Cyclooxygenase-2
cPLA2Cytosolic phospholipase A2
CXCLCXC motif chemokine ligand
DGPDeamidated gluten peptides
DHDermatitis herpetiformis
DHADocosahexanoic acid
DPPIVDipeptidyl peptidase IV
ECGC(-)-Epigallocatechin-3-gallate
EEPEthanolic extract of propolis
EGC(-)-Epigallocatechin
ELISAEnzyme-linked immunosorbent assay
EoEEosinophilic esophagitis
FAFatty acid
FD-4FITC-dextran 4000
FM PEPFlavobacterium meningosepticum
GCDGluten-containing diet
GFDGluten-free diet
GIPGluten immunogenic peptide
GITGastrointestinal tract
GPx3Glutathione peroxidase-3
GSHGlutathione
GSRSGastrointestinal symptoms rating scale
HDLHigh-density lipoprotein
HLAHuman leukocyte antigen
HPLCHigh-performance liquid chromatography
IBSInflammatory bowel disease
IBS-DDiarrhea-predominant inflammatory bowel disease
IECIntestinal epithelial cell
IELIntestinal epithelial lymphocyte
IFNInterferon
IgImmunoglobulin
ILInterleukin
iNOSInducible nitric oxide synthase
IPIntestinal permeability
IRFInterferon regulatory factor
JAKJanus kinase
LAMALactulose to mannitol
LMRLymphocyte-to-monocyte ratio
MasCAMMucosal addressin cell adhesion molecule
MCPMonocyte chemoattractant protein
MDAMalondialdehyde
MHCMajor histocompatibility complex
MoAbMonoclonal antibody
MPOMyeloperoxidase
MX PEPMyxococcus xanthus
n-3Omega-3
NADPHNicotinamide Adenine Dinucleotide Phosphate
NCGSNon-celiac gluten sensitivity
NF-κBNuclear factor kappa-light-chain enhancer of activated B cells
NKNatural killer
NKTNatural killer T cells
NONitric oxide
NOD/DQ8HLA-DQ8 non-obese diabetic
NOXNADPH oxidase
P(HEMA-co-SS)Poly(hydroxyethyl methacrylate-co-styrene sulfonate)
PBMCPeripheral blood mononuclear cells
PCEProcyanidin B2-enriched cocoa extract
PGE2Prostaglandin E2
PLRPlatelet-to-lymphocyte ratio
PSPcProanthocyanidins
pSTATPhosphorylated STAT
PT-GPepsin-trypsin-digested gliadin
PUFAPolyunsaturated fatty acid
RCeD-IIType II refractory CeD
ROSReactive oxygen species
SCFAShort-chain fatty acids
SIRT6Sirtuin 6
SOPSuperoxide dismutase
SrpSerine protease inhibitor
STATSignal transducer and activator of transcription
TEAETreatment-emergent adverse event
TECTyrosine kinase expressed in hepatocellular carcinoma
TEERTransepithelial electrical resistance
tgTransgenic
ThT-helper
TLRToll-like receptor
tTG2Tissue transglutaminase 2
VD3Vitamin D3
Vh/CdVillous height/crypt depth ratio

References

  1. Ludvigsson, J.F.; Leffler, D.A.; Bai, J.C.; Biagi, F.; Fasano, A.; Green, P.H.R.; Hadjivassiliou, M.; Kaukinen, K.; Kelly, C.P.; Leonard, J.N.; et al. The Oslo Definitions for Coeliac Disease and Related Terms. Gut 2013, 62, 43–52. [Google Scholar] [CrossRef]
  2. Al-Toma, A.; Volta, U.; Auricchio, R.; Castillejo, G.; Sanders, D.S.; Cellier, C.; Mulder, C.J.; Lundin, K.E.A. European Society for the Study of Coeliac Disease (ESsCD) Guideline for Coeliac Disease and Other Gluten-Related Disorders. United Eur. Gastroenterol. J. 2019, 7, 583–613. [Google Scholar] [CrossRef]
  3. Biesiekierski, J.R. What Is Gluten? J. Gastroenterol. Hepatol. 2017, 32, 78–81. [Google Scholar] [CrossRef]
  4. Schalk, K.; Lang, C.; Wieser, H.; Koehler, P.; Scherf, K.A. Quantitation of the Immunodominant 33-Mer Peptide from α-Gliadin in Wheat Flours by Liquid Chromatography Tandem Mass Spectrometry. Sci. Rep. 2017, 7, srep45092. [Google Scholar] [CrossRef] [PubMed]
  5. Catassi, C.; Verdu, E.F.; Bai, J.C.; Lionetti, E. Coeliac Disease. Lancet 2022, 399, 2413–2426. [Google Scholar] [CrossRef] [PubMed]
  6. Makharia, G.K.; Singh, P.; Catassi, C.; Sanders, D.S.; Leffler, D.; Ali, R.A.R.; Bai, J.C. The Global Burden of Coeliac Disease: Opportunities and Challenges. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 313–327. [Google Scholar] [CrossRef]
  7. Jansson-Knodell, C.L.; Hujoel, I.A.; West, C.P.; Taneja, V.; Prokop, L.J.; Rubio-Tapia, A.; Murray, J.A. Sex Difference in Celiac Disease in Undiagnosed Populations: A Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2019, 17, 1954–1968.e13. [Google Scholar] [CrossRef]
  8. Heijdra Suasnabar, J.; Meijer, C.R.; Smit, L.; van Overveld, F.; Thom, H.; Keeney, E.; Mearin, M.L.; van den Akker-van Marle, M.E. Long-Term Cost-Effectiveness of Case Finding and Mass Screening for Celiac Disease in Children. Gastroenterology 2024, 167, 1129–1140. [Google Scholar] [CrossRef]
  9. Bascuñán, K.A.; Vespa, M.C.; Araya, M. Celiac Disease: Understanding the Gluten-Free Diet. Eur. J. Nutr. 2017, 56, 449–459. [Google Scholar] [CrossRef]
  10. Wei, G.; Helmerhorst, E.J.; Darwish, G.; Blumenkranz, G.; Schuppan, D. Gluten Degrading Enzymes for Treatment of Celiac Disease. Nutrients 2020, 12, 2095. [Google Scholar] [CrossRef] [PubMed]
  11. Mitea, C.; Havenaar, R.; Wouter Drijfhout, J.; Edens, L.; Dekking, L.; Koning, F.; Dekking, E.H.A. Efficient Degradation of Gluten by a Prolyl Endoprotease in a Gastrointestinal Model: Implications for Coeliac Disease. Gut 2008, 57, 25–32. [Google Scholar] [CrossRef]
  12. Montserrat, V.; Bruins, M.J.; Edens, L.; Koning, F. Influence of Dietary Components on Aspergillus Niger Prolyl Endoprotease Mediated Gluten Degradation. Food Chem. 2015, 174, 440–445. [Google Scholar] [CrossRef]
  13. König, J.; Holster, S.; Bruins, M.J.; Brummer, R.J. Randomized Clinical Trial: Effective Gluten Degradation by Aspergillus Niger-Derived Enzyme in a Complex Meal Setting. Sci. Rep. 2017, 7, 13100. [Google Scholar] [CrossRef]
  14. Salden, B.N.; Monserrat, V.; Troost, F.J.; Bruins, M.J.; Edens, L.; Bartholomé, R.; Haenen, G.R.; Winkens, B.; Koning, F.; Masclee, A.A. Randomised Clinical Study: Aspergillus Niger-Derived Enzyme Digests Gluten in the Stomach of Healthy Volunteers. Aliment. Pharmacol. Ther. 2015, 42, 273–285. [Google Scholar] [CrossRef] [PubMed]
  15. Tack, G.J.; van de Water, J.M.W.; Bruins, M.J.; Kooy-Winkelaar, E.M.C.; van Bergen, J.; Bonnet, P.; Vreugdenhil, A.C.E.; Korponay-Szabo, I.; Edens, L.; von Blomberg, B.M.E.; et al. Consumption of Gluten with Gluten-Degrading Enzyme by Celiac Patients: A Pilot-Study. World J. Gastroenterol. 2013, 19, 5837–5847. [Google Scholar] [CrossRef]
  16. Stefanolo, J.P.; Segura, V.; Grizzuti, M.; Heredia, A.; Comino, I.; Costa, A.F.; Puebla, R.; Temprano, M.P.; Niveloni, S.I.; de Diego, G.; et al. Effect of Aspergillus Niger Prolyl Endopeptidase in Patients with Celiac Disease on a Long-Term Gluten-Free Diet. World J. Gastroenterol. 2024, 30, 1545–1555. [Google Scholar] [CrossRef] [PubMed]
  17. Mohan Kumar, B.V.; Vijaykrishnaraj, M.; Kurrey, N.K.; Shinde, V.S.; Prabhasankar, P. Prolyl Endopeptidase-Degraded Low Immunoreactive Wheat Flour Attenuates Immune Responses in Caco-2 Intestinal Cells and Gluten-Sensitized BALB/c Mice. Food Chem. Toxicol. 2019, 129, 466–475. [Google Scholar] [CrossRef] [PubMed]
  18. Walter, T.; Wieser, H.; Koehler, P. Production of Gluten-Free Wheat Starch by Peptidase Treatment. J. Cereal. Sci. 2014, 60, 202–209. [Google Scholar] [CrossRef]
  19. Knorr, V.; Wieser, H.; Koehler, P. Production of Gluten-Free Beer by Peptidase Treatment. Eur. Food Res. Technol. 2016, 242, 1129–1140. [Google Scholar] [CrossRef]
  20. Camarca, A.; D’Auria, G.; Rotondi Aufiero, V.; Nitride, C.; Giardullo, N.; Sapone, A.; Leffler, D.; Ferranti, P.; Mazzarella, G. Digestion Supplemented with Commercial Proteases: Evaluation of the Fate of Gluten Immunogenic Peptides in Pizza. Food Res. Int. 2025, 220, 117027. [Google Scholar] [CrossRef]
  21. Tschollar, W.; Mourabit, S. Pharmaceutical Compositions Comprising Aminopeptidase and Dipeptidylpeptidase Polypeptides and Uses Thereof 2023. Available online: https://patents.google.com/patent/WO2025067633A1/en?assignee=AMYRA&oq=AMYRA (accessed on 30 July 2025).
  22. Mourabit, S.; Römer, S.; Bonner, E.R.; Winter, F.; Tschollar, J.; Tzvetkov, M.V.; Weitschies, W.; Engeli, S.; Tschollar, W. Exopeptidase Combination Enhances the Degradation of Isotopically Labelled Gluten Immunogenic Peptides in Humans. Front. Immunol. 2024, 15, 1425982. [Google Scholar] [CrossRef]
  23. Ehren, J.; Móron, B.; Martin, E.; Bethune, M.T.; Gray, G.M.; Khosla, C. A Food-Grade Enzyme Preparation with Modest Gluten Detoxification Properties. PLoS ONE 2009, 4, e6313. [Google Scholar] [CrossRef]
  24. Li, Y.; Yu, J.; Goktepe, I.; Ahmedna, M. The Potential of Papain and Alcalase Enzymes and Process Optimizations to Reduce Allergenic Gliadins in Wheat Flour. Food Chem. 2016, 196, 1338–1345. [Google Scholar] [CrossRef]
  25. Zamakhchari, M.; Wei, G.; Dewhirst, F.; Lee, J.; Schuppan, D.; Oppenheim, F.G.; Helmerhorst, E.J. Identification of Rothia Bacteria as Gluten-Degrading Natural Colonizers of the Upper Gastro-Intestinal Tract. PLoS ONE 2011, 6, e24455. [Google Scholar] [CrossRef]
  26. Pultz, I.S.; Hill, M.; Vitanza, J.M.; Wolf, C.; Saaby, L.; Liu, T.; Winkle, P.; Leffler, D.A. Gluten Degradation, Pharmacokinetics, Safety, and Tolerability of TAK-062, an Engineered Enzyme to Treat Celiac Disease. Gastroenterology 2021, 161, 81–93.e3. [Google Scholar] [CrossRef]
  27. Wolf, C.; Siegel, J.B.; Tinberg, C.; Camarca, A.; Gianfrani, C.; Paski, S.; Guan, R.; Montelione, G.; Baker, D.; Pultz, I.S. Engineering of Kuma030: A Gliadin Peptidase That Rapidly Degrades Immunogenic Gliadin Peptides in Gastric Conditions. J. Am. Chem. Soc. 2015, 137, 13106–13113. [Google Scholar] [CrossRef] [PubMed]
  28. National Library of Medicine (U.S.). A Study of TAK-062 in Treatment of Active Celiac Disease in Participants Attempting a Gluten-Free Diet. Available online: https://www.clinicaltrials.gov/study/NCT05353985 (accessed on 7 July 2025).
  29. Liu, Y.Y.; Lee, C.C.; Hsu, J.H.; Leu, W.M.; Meng, M. Efficient Hydrolysis of Gluten-Derived Celiac Disease-Triggering Immunogenic Peptides by a Bacterial Serine Protease from Burkholderia Gladioli. Biomolecules 2021, 11, 451. [Google Scholar] [CrossRef] [PubMed]
  30. Cavaletti, L.; Taravella, A.; Carrano, L.; Carenzi, G.; Sigurtà, A.; Solinas, N.; De Caro, S.; Di Stasio, L.; Picascia, S.; Laezza, M.; et al. E40, a Novel Microbial Protease Efficiently Detoxifying Gluten Proteins, for the Dietary Management of Gluten Intolerance. Sci. Rep. 2019, 9, 13147. [Google Scholar] [CrossRef] [PubMed]
  31. Gass, J.; Ehren, J.; Strohmeier, G.; Isaacs, I.; Khosla, C. Fermentation, Purification, Formulation, and Pharmacological Evaluation of a Prolyl Endopeptidase from Myxococcus Xanthus: Implications for Celiac Sprue Therapy. Biotechnol. Bioeng. 2005, 92, 674–684. [Google Scholar] [CrossRef]
  32. Gass, J.; Vora, H.; Bethune, M.T.; Gray, G.M.; Khosla, C. Effect of Barley Endoprotease EP-B2 on Gluten Digestion in the Intact Rat. J. Pharmacol. Exp. Ther. 2006, 318, 1178–1186. [Google Scholar] [CrossRef]
  33. Bordusa, F.; Jakubke, H.-D. The Specificity of Prolyl Endopeptidase from Flavobacterium Meningoseptum: Mapping the S H Subsites by Positional Scanning via Acyl Transfer. Bioorganic Med. Chem. 1998, 6, 1775–1780. [Google Scholar] [CrossRef]
  34. Siegel, M.; Bethune, M.T.; Gass, J.; Ehren, J.; Xia, J.; Johannsen, A.; Stuge, T.B.; Gray, G.M.; Lee, P.P.; Khosla, C. Rational Design of Combination Enzyme Therapy for Celiac Sprue. Chem. Biol. 2006, 13, 649–658. [Google Scholar] [CrossRef]
  35. Xue, L.; Li, Y.; Li, T.; Pan, H.; Liu, J.; Fan, M.; Qian, H.; Zhang, H.; Ying, H.; Wang, L. Phosphorylation and Enzymatic Hydrolysis with Alcalase and Papain Effectively Reduce Allergic Reactions to Gliadins in Normal Mice. J. Agric. Food Chem. 2019, 67, 6313–6323. [Google Scholar] [CrossRef]
  36. del Amo-Maestro, L.; Mendes, S.R.; Rodríguez-Banqueri, A.; Garzon-Flores, L.; Girbal, M.; Rodríguez-Lagunas, M.J.; Guevara, T.; Franch, À.; Pérez-Cano, F.J.; Eckhard, U.; et al. Molecular and in Vivo Studies of a Glutamate-Class Prolyl-Endopeptidase for Coeliac Disease Therapy. Nat. Commun. 2022, 13, 4446. [Google Scholar] [CrossRef] [PubMed]
  37. Rey, M.; Yang, M.; Lee, L.; Zhang, Y.; Sheff, J.G.; Sensen, C.W.; Mrazek, H.; Halada, P.; Man, P.; McCarville, J.L.; et al. Addressing Proteolytic Efficiency in Enzymatic Degradation Therapy for Celiac Disease. Sci. Rep. 2016, 6, 30980. [Google Scholar] [CrossRef] [PubMed]
  38. Mousavi Maleki, M.S.; Aghamirza Moghim Ali Abadi, H.; Vaziri, B.; Shabani, A.A.; Ghavami, G.; Madanchi, H.; Sardari, S. Bromelain and Ficin Proteolytic Effects on Gliadin Cytotoxicity and Expression of Genes Involved in Cell-Tight Junctions in Caco-2 Cells. Amino Acids 2023, 55, 1601–1619. [Google Scholar] [CrossRef]
  39. Rutherfurd, S.M.; Montoya, C.A.; Zou, M.L.; Moughan, P.J.; Drummond, L.N.; Boland, M.J. Effect of Actinidin from Kiwifruit (Actinidia Deliciosa Cv. Hayward) on the Digestion of Food Proteins Determined in the Growing Rat. Food Chem. 2011, 129, 1681–1689. [Google Scholar] [CrossRef]
  40. Jayawardana, I.A.; Boland, M.J.; Higgs, K.; Zou, M.; Loo, T.; Mcnabb, W.C.; Montoya, C.A. The Kiwifruit Enzyme Actinidin Enhances the Hydrolysis of Gluten Proteins during Simulated Gastrointestinal Digestion. Food Chem. 2021, 341, 128239. [Google Scholar] [CrossRef] [PubMed]
  41. Jayawardana, I.A.; Boland, M.J.; Loo, T.S.; McNabb, W.C.; Montoya, C.A. Actinidin Reduces Gluten-Derived Immunogenic Peptides Reaching the Small Intestine in an in Vitro Semi-Dynamic Gastrointestinal Tract Digestion Model. Food Res. Int. 2022, 159, 111560. [Google Scholar] [CrossRef]
  42. Buddrick, O.; Cornell, H.J.; Small, D.M. Reduction of Toxic Gliadin Content of Wholegrain Bread by the Enzyme Caricain. Food Chem. 2015, 170, 343–347. [Google Scholar] [CrossRef]
  43. Savvateeva, L.V.; Gorokhovets, N.V.; Makarov, V.A.; Serebryakova, M.V.; Solovyev, A.G.; Morozov, S.Y.; Reddy, V.P.; Zernii, E.Y.; Zamyatnin, A.A.; Aliev, G. Glutenase and Collagenase Activities of Wheat Cysteine Protease Triticain-α: Feasibility for Enzymatic Therapy Assays. Int. J. Biochem. Cell Biol. 2015, 62, 115–124. [Google Scholar] [CrossRef]
  44. Tye-Din, J.A.; Anderson, R.P.; Ffrench, R.A.; Brown, G.J.; Hodsman, P.; Siegel, M.; Botwick, W.; Shreeniwas, R. The Effects of ALV003 Pre-Digestion of Gluten on Immune Response and Symptoms in Celiac Disease in Vivo. Clin. Immunol. 2010, 134, 289–295. [Google Scholar] [CrossRef]
  45. Wei, G.; Darwish, G.; Oppenheim, F.G.; Schuppan, D.; Helmerhorst, E.J. Commensal Bacterium Rothia Aeria Degrades and Detoxifies Gluten via a Highly Effective Subtilisin Enzyme. Nutrients 2020, 12, 3724. [Google Scholar] [CrossRef]
  46. Gass, J.; Khosla, C. Prolyl Endopeptidases. Cell. Mol. Life Sci. 2007, 64, 345–355. [Google Scholar] [CrossRef]
  47. Gomis-Rüth, F.X.; Del Amo Maestro, L.; Dos Mendes Rei, S.I.; Eckhard, U.; Rodríguez Banqueri, A.; Guevara Puig, T.; Pérez Cano, F.; Rodríguez Lagunas, M.J.; Franch Masferrer, À.; Girbal González, M. Proteína Degradadora de Gluten y Usos de La Misma 2023. Available online: https://patents.google.com/patent/ES3008845A1/es (accessed on 8 July 2025).
  48. Pahlavan, A.; Sharma, G.M.; Pereira, M.; Williams, K.M. Effects of Grain Species and Cultivar, Thermal Processing, and Enzymatic Hydrolysis on Gluten Quantitation. Food Chem. 2016, 208, 264–271. [Google Scholar] [CrossRef] [PubMed]
  49. Siegel, M.; Garber, M.E.; Spencer, A.G.; Botwick, W.; Kumar, P.; Williams, R.N.; Kozuka, K.; Shreeniwas, R.; Pratha, V.; Adelman, D.C. Safety, Tolerability, and Activity of ALV003: Results from Two Phase 1 Single, Escalating-Dose Clinical Trials. Dig. Dis. Sci. 2012, 57, 440–450. [Google Scholar] [CrossRef] [PubMed]
  50. Lähdeaho, M.L.; Kaukinen, K.; Laurila, K.; Vuotikka, P.; Koivurova, O.P.; Kärjä-Lahdensuu, T.; Marcantonio, A.; Adelman, D.C.; Mäki, M. Glutenase ALV003 Attenuates Gluten-Induced Mucosal Injury in Patients with Celiac Disease. Gastroenterology 2014, 146, 1649–1658. [Google Scholar] [CrossRef] [PubMed]
  51. Murray, J.A.; Kelly, C.P.; Green, P.H.R.; Marcantonio, A.; Wu, T.T.; Mäki, M.; Adelman, D.C.; Ansari, S.; Ayub, K.; Basile, A.; et al. No Difference Between Latiglutenase and Placebo in Reducing Villous Atrophy or Improving Symptoms in Patients with Symptomatic Celiac Disease. Gastroenterology 2017, 152, 787–798.e2. [Google Scholar] [CrossRef]
  52. Murray, J.A.; Syage, J.A.; Wu, T.T.; Dickason, M.A.; Ramos, A.G.; Van Dyke, C.; Horwath, I.; Lavin, P.T.; Mäki, M.; Hujoel, I.; et al. Latiglutenase Protects the Mucosa and Attenuates Symptom Severity in Patients with Celiac Disease Exposed to a Gluten Challenge. Gastroenterology 2022, 163, 1510–1521.e6. [Google Scholar] [CrossRef]
  53. Gujral, N.; Löbenberg, R.; Suresh, M.; Sunwoo, H. In-Vitro and in-Vivo Binding Activity of Chicken Egg Yolk Immunoglobulin y (IgY) against Gliadin in Food Matrix. J. Agric. Food Chem. 2012, 60, 3166–3172. [Google Scholar] [CrossRef]
  54. Sample, D.A.; Sunwoo, H.H.; Huynh, H.Q.; Rylance, H.L.; Robert, C.L.; Xu, B.W.; Kang, S.H.; Gujral, N.; Dieleman, L.A. AGY, a Novel Egg Yolk-Derived Anti-Gliadin Antibody, Is Safe for Patients with Celiac Disease. Dig. Dis. Sci. 2017, 62, 1277–1285. [Google Scholar] [CrossRef]
  55. National Library of Medicine (U.S.). A Randomized, Double-Blind, Placebo Controlled, Crossover Trial to Evaluate Safety and Efficacy of AGY in Celiac Disease (AGY-010). Available online: https://clinicaltrials.gov/study/NCT03707730?term=agy-010&rank=1 (accessed on 8 July 2025).
  56. Pinier, M.; Verdu, E.F.; Nasser–Eddine, M.; David, C.S.; Vézina, A.; Rivard, N.; Leroux, J. Polymeric Binders Suppress Gliadin-Induced Toxicity in the Intestinal Epithelium. Gastroenterology 2009, 136, 288–298. [Google Scholar] [CrossRef]
  57. McCarville, J.L.; Nisemblat, Y.; Galipeau, H.J.; Jury, J.; Tabakman, R.; Cohen, A.; Naftali, E.; Neiman, B.; Halbfinger, E.; Murray, J.A.; et al. BL-7010 Demonstrates Specific Binding to Gliadin and Reduces Gluten-Associated Pathology in a Chronic Mouse Model of Gliadin Sensitivity. PLoS ONE 2014, 9, e109972. [Google Scholar] [CrossRef] [PubMed]
  58. National Library of Medicine (U.S.). A Two-Part, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Systemic Exposure of Single Escalating Administrations and Repeated Administration of BL-7010 in Well-Controlled Celiac Patients. Available online: https://clinicaltrials.gov/study/NCT01990885?intr=BL-7010&rank=1 (accessed on 18 June 2025).
  59. Leffler, D.A.; Kelly, C.P.; Green, P.H.R.; Fedorak, R.N.; Dimarino, A.; Perrow, W.; Rasmussen, H.; Wang, C.; Bercik, P.; Bachir, N.M.; et al. Larazotide Acetate for Persistent Symptoms of Celiac Disease despite a Gluten-Free Diet: A Randomized Controlled Trial. Gastroenterology 2015, 148, 1311–1319.e6. [Google Scholar] [CrossRef]
  60. Paterson, B.M.; Lammers, K.M.; Arrieta, M.C.; Fasano, A.; Meddings, J.B. The Safety, Tolerance, Pharmacokinetic and Pharmacodynamic Effects of Single Doses of AT-1001 in Coeliac Disease Subjects: A Proof of Concept Study. Aliment. Pharmacol. Ther. 2007, 26, 757–766. [Google Scholar] [CrossRef]
  61. Leffler, D.A.; Kelly, C.P.; Abdallah, H.Z.; Colatrella, A.M.; Harris, L.A.; Leon, F.; Arterburn, L.A.; Paterson, B.M.; Lan, Z.H.; Murray, J.A. A Randomized, Double-Blind Study of Larazotide Acetate to Prevent the Activation of Celiac Disease during Gluten Challenge. Am. J. Gastroenterol. 2012, 107, 1554–1562. [Google Scholar] [CrossRef] [PubMed]
  62. Kelly, C.P.; Green, P.H.R.; Murray, J.A.; Dimarino, A.; Colatrella, A.; Leffler, D.A.; Alexander, T.; Arsenescu, R.; Leon, F.; Jiang, J.G.; et al. Larazotide Acetate in Patients with Coeliac Disease Undergoing a Gluten Challenge: A Randomised Placebo-Controlled Study. Aliment. Pharmacol. Ther. 2013, 37, 252–262. [Google Scholar] [CrossRef] [PubMed]
  63. National Library of Medicine (U.S.). Study to Evaluate the Efficacy and Safety of Larazotide Acetate for the Relief of CeD Symptoms. Available online: https://clinicaltrials.gov/study/NCT03569007?cond=ced%20in%20syndrome&viewType=Table&checkSpell=&rank=2 (accessed on 8 July 2025).
  64. Schumann, M.; Siegmund, B.; Schulzke, J.D.; Fromm, M. Celiac Disease: Role of the Epithelial Barrier. CMGH 2017, 3, 150–162. [Google Scholar] [CrossRef] [PubMed]
  65. Hoilat, G.J.; Altowairqi, A.K.; Ayas, M.F.; Alhaddab, N.T.; Alnujaidi, R.A.; Alharbi, H.A.; Alyahyawi, N.; Kamal, A.; Alhabeeb, H.; Albazee, E.; et al. Larazotide Acetate for Treatment of Celiac Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Clin. Res. Hepatol. Gastroenterol. 2022, 46, 101782. [Google Scholar] [CrossRef] [PubMed]
  66. Daveson, A.J.M.; Stubbs, R.; Polasek, T.M.; Isola, J.; Anderson, R.; Tye-Din, J.A.; Schoeman, M.; Lionnet, C.; Mei, S.L.C.Y.; Mihajlović, J.; et al. Safety, Clinical Activity, Pharmacodynamics, and Pharmacokinetics of IMU-856, a SIRT6 Modulator, in Coeliac Disease: A First-in-Human, Randomised, Double-Blind, Placebo-Controlled, Phase 1 Trial. Lancet Gastroenterol. Hepatol. 2024, 10, 44–54. [Google Scholar] [CrossRef] [PubMed]
  67. Xu, K.; Guo, Y.; Ping, L.; Qiu, Y.; Liu, Q.; Li, Z.; Wang, Z. Protective Effects of SIRT6 Overexpression against DSS-Induced Colitis in Mice. Cells 2020, 9, 1513. [Google Scholar] [CrossRef]
  68. Schuppan, D.; Mäki, M.; Lundin, K.E.A.; Isola, J.; Friesing-Sosnik, T.; Taavela, J.; Popp, A.; Koskenpato, J.; Langhorst, J.; Hovde, Ø.; et al. A Randomized Trial of a Transglutaminase 2 Inhibitor for Celiac Disease. N. Engl. J. Med. 2021, 385, 35–45. [Google Scholar] [CrossRef]
  69. Büchold, C.; Hils, M.; Gerlach, U.; Weber, J.; Pelzer, C.; Heil, A.; Aeschlimann, D.; Pasternack, R. Features of ZED1227: The First-in-Class Tissue Transglutaminase Inhibitor Undergoing Clinical Evaluation for the Treatment of Celiac Disease. Cells 2022, 11, 1667. [Google Scholar] [CrossRef]
  70. National Library of Medicine (U.S.). Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of GSK3915393 in Healthy Participants and to Evaluate the Interaction Between GSK3915393 and Grapefruit Juice and Itraconazole. Available online: https://clinicaltrials.gov/study/NCT04604795?cond=%22Celiac%20disease%22&rank=9 (accessed on 9 July 2025).
  71. National Library of Medicine (U.S.). A Study to Evaluate the Efficacy and Safety of GSK3915393 in Participants with Idiopathic Pulmonary Fibrosis (IPF). Available online: https://clinicaltrials.gov/study/NCT06317285?term=Labrador%20Lung&viewType=Table&rank=10 (accessed on 9 July 2025).
  72. Abadie, V.; Kim, S.M.; Lejeune, T.; Palanski, B.A.; Ernest, J.D.; Tastet, O.; Voisine, J.; Discepolo, V.; Marietta, E.V.; Hawash, M.B.F.; et al. IL-15, Gluten and HLA-DQ8 Drive Tissue Destruction in Coeliac Disease. Nature 2020, 578, 600–604. [Google Scholar] [CrossRef]
  73. Dunne, M.R.; Byrne, G.; Chirdo, F.G.; Feighery, C. Coeliac Disease Pathogenesis: The Uncertainties of a Well-Known Immune Mediated Disorder. Front. Immunol. 2020, 11, 1374. [Google Scholar] [CrossRef] [PubMed]
  74. Lähdeaho, M.L.; Scheinin, M.; Vuotikka, P.; Taavela, J.; Popp, A.; Laukkarinen, J.; Koffert, J.; Koivurova, O.P.; Pesu, M.; Kivelä, L.; et al. Safety and Efficacy of AMG 714 in Adults with Coeliac Disease Exposed to Gluten Challenge: A Phase 2a, Randomised, Double-Blind, Placebo-Controlled Study. Lancet Gastroenterol. Hepatol. 2019, 4, 948–959. [Google Scholar] [CrossRef] [PubMed]
  75. Cellier, C.; Bouma, G.; van Gils, T.; Khater, S.; Malamut, G.; Crespo, L.; Collin, P.; Green, P.H.; Crowe, S.E.; Tsuji, W.; et al. Safety and Efficacy of AMG 714 in Patients with Type 2 Refractory Coeliac Disease: A Phase 2a, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study. Lancet Gastroenterol. Hepatol. 2019, 4, 960–970. [Google Scholar] [CrossRef]
  76. National Library of Medicine (U.S.). PRV-015 in Gluten-Free Diet Non-Responsive Celiac Disease (PROACTIVE). Available online: https://clinicaltrials.gov/study/NCT04424927 (accessed on 9 July 2025).
  77. Vicari, A.P.; Schoepfer, A.M.; Meresse, B.; Goffin, L.; Léger, O.; Josserand, S.; Guégan, N.; Yousefi, S.; Straumann, A.; Cerf-Bensussan, N.; et al. Discovery and Characterization of a Novel Humanized Anti-IL-15 Antibody and Its Relevance for the Treatment of Refractory Celiac Disease and Eosinophilic Esophagitis. mAbs 2017, 9, 927–944. [Google Scholar] [CrossRef] [PubMed]
  78. Abadie, V.; Jabri, B. IL-15: A Central Regulator of Celiac Disease Immunopathology. Immunol. Rev. 2014, 260, 221–234. [Google Scholar] [CrossRef]
  79. Lucendo, A.J.; Arias, Á.; Tenias, J.M. Systematic Review: The Association between Eosinophilic Oesophagitis and Coeliac Disease. Aliment. Pharmacol. Ther. 2014, 40, 422–434. [Google Scholar] [CrossRef]
  80. National Library of Medicine (U.S.). Dose Escalation Study to Evaluate an Experimental New Treatment (CALY-002) in Healthy Subjects and Subjects with Celiac Disease and Eosinophilic Esophagitis. Available online: https://clinicaltrials.gov/study/NCT04593251?intr=CALY-002&rank=1 (accessed on 10 July 2025).
  81. Koenig, L.; Ben-Eliezer, I.; Tao, T.P.; Winter, A.; Grossman, M. Modeling Human Natural Killer Cell Development and Drug Response in a Microfluidic Bone Marrow Model. Front. Immunol. 2025, 16, 1499397. [Google Scholar] [CrossRef] [PubMed]
  82. Schnir, V.; Rigby, M.; Calota, M.R.; Gross, N.; Echenique, I.; Lammerich, A.; Liu, Y.; Robinson, R.; Wangsa, J.; Morrow, V. Su1326 Developing TEV-53408 for the Treatment of Celiac Disease: Summary of Preliminary Results from the First in Human Phase 1 Study in Healthy Volunteers. Gastroenterology 2024, 166, S-730. [Google Scholar] [CrossRef]
  83. National Library of Medicine (U.S.). A Trial to Assess the Efficacy and Safety of TEV-53408 in Adults with Celiac Disease ClinicalTrials.Gov ID NCT06807463. Available online: https://clinicaltrials.gov/study/NCT06807463 (accessed on 10 July 2025).
  84. Tiet, P.; Giovannone, A.J.; Al-Mawsawi, L.; Ampudia, J.; Marrocco, V.; Connelly, S.; Ng, C. Γc Receptor Antagonist, EQ102, Prevents the NK and T Cell-Mediated Responses Driven by IL-15 and IL-21. J. Immunol. 2023, 210, 219–243. [Google Scholar] [CrossRef]
  85. Equillium Equillium Announces Initiation of Phase 1 Study of EQ102, A First-in-Class Multi-Cytokine Inhibitor of IL-15 and IL-21 Targeting Celiac Disease. Available online: https://www.equilliumbio.com/investors/press-releases/news-details/2022/Equillium-Announces-Initiation-of-Phase-1-study-of-EQ102-A-First-in-Class-Multi-Cytokine-Inhibitor-of-IL-15-and-IL-21-Targeting-Celiac-Disease/default.aspx (accessed on 10 July 2022).
  86. Australian New Zealand Clinical Trials Registry a Randomized, Double-Blind, Placebo-Controlled Phase I Study to Assess the Safety and Tolerability of Subcutaneous EQ102 Administered in a Single and Multiple Ascending Dose Schedule. Available online: https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=384358 (accessed on 10 July 2022).
  87. National Library of Medicine (U.S.). Study of EQ101 in Adult Subjects with Moderate to Severe Alopecia Areata. Available online: https://clinicaltrials.gov/study/NCT05589610?term=EQ101&rank=1 (accessed on 10 July 2022).
  88. Equillium Equillium Announces Update on Multi-Cytokine Inhibitors EQ101 & EQ102 in Development for Alopecia Areata and Celiac Disease. Available online: https://www.equilliumbio.com/investors/press-releases/news-details/2023/Equillium-Announces-Update-on-Multi-Cytokine-Inhibitors-EQ101--EQ102-in-Development-for-Alopecia-Areata-and-Celiac-Disease/default.aspx (accessed on 10 July 2025).
  89. Equillium Equillium Announces Preclinical Data from New Orally Deliverable Multi-Cytokine Inhibitor in Presentation at the 18th Annual Peptide Therapeutics Symposium. Available online: https://www.equilliumbio.com/investors/press-releases/news-details/2023/Equillium-Announces-Preclinical-Data-from-New-Orally-Deliverable-Multi-Cytokine-Inhibitor-in-Presentation-at-the-18th-Annual-Peptide-Therapeutics-Symposium/default.aspx (accessed on 11 July 2025).
  90. Rotondi Aufiero, V.; Iacomino, G.; De Chiara, G.; Picariello, E.; Iaquinto, G.; Troncone, R.; Mazzarella, G. Neutralizing IL-15 Inhibits Tissue-Damaging Immune Response in Ex Vivo Cultured Untreated Celiac Intestinal Mucosa. Cells 2025, 14, 234. [Google Scholar] [CrossRef] [PubMed]
  91. National Library of Medicine (U.S.). A Study to Evaluate the Efficacy and Safety of AMG 714 in Adult Patients with Celiac Disease. Available online: https://clinicaltrials.gov/study/NCT02637141?cond=celiac&intr=prv-015&rank=2&tab=results (accessed on 9 July 2025).
  92. National Library of Medicine (U.S.). A Phase II Study of CCX282-B in Patients with Celiac Disease. Available online: https://clinicaltrials.gov/study/NCT00540657?term=NCT00540657&rank=1 (accessed on 10 July 2022).
  93. Walters, M.J.; Wang, Y.; Lai, N.; Baumgart, T.; Zhao, B.N.; Dairaghi, D.J.; Bekker, P.; Ertl, L.S.; Penfold, M.E.T.; Jaen, J.C.; et al. Characterization of CCX282-B, an Orally Bioavailable Antagonist of the CCR9 Chemokine Receptor, for Treatment of Inflammatory Bowel Disease. J. Pharmacol. Exp. Ther. 2010, 335, 61–69. [Google Scholar] [CrossRef]
  94. National Library of Medicine (U.S.). PTG-100 for Patients with Celiac Disease. Available online: https://www.clinicaltrials.gov/study/NCT04524221 (accessed on 11 July 2022).
  95. Sandborn, W.J.; Mattheakis, L.C.; Modi, N.B.; Pugatch, D.; Bressler, B.; Lee, S.; Bhandari, R.; Kanwar, B.; Shames, R.; D’Haens, G.; et al. PTG-100, an Oral A4β7 Antagonist Peptide: Preclinical Development and Phase 1 and 2a Studies in Ulcerative Colitis. Gastroenterology 2021, 161, 1853–1864.e10. [Google Scholar] [CrossRef]
  96. Kapoerchan, V.V.; Wiesner, M.; Hillaert, U.; Drijfhout, J.W.; Overhand, M.; Alard, P.; van der Marel, G.A.; Overkleeft, H.S.; Koning, F. Design, Synthesis and Evaluation of High-Affinity Binders for the Celiac Disease Associated HLA-DQ2 Molecule. Mol. Immunol. 2010, 47, 1091–1097. [Google Scholar] [CrossRef]
  97. Kapoerchan, V.V.; Wiesner, M.; Overhand, M.; van der Marel, G.A.; Koning, F.; Overkleeft, H.S. Design of Azidoproline Containing Gluten Peptides to Suppress CD4+ T-Cell Responses Associated with Celiac Disease. Bioorg. Med. Chem. 2008, 16, 2053–2062. [Google Scholar] [CrossRef]
  98. Dieckman, T.; Schumann, M.; Beaumont, H.; Bontkes, H.J.; Koning, F.; Bouma, G.; Byrnes, V.; McCarthy, J.; Neefjes-Borst, A.; Loddenkemper, C.; et al. Enduring Clinical Remission in Refractory Celiac Disease Type II with Tofacitinib: An Open-Label Clinical Study. Clin. Gastroenterol. Hepatol. 2024, 22, 2334–2336. [Google Scholar] [CrossRef]
  99. Blair, H.A. Ritlecitinib: First Approval. Drugs 2023, 83, 1315–1321. [Google Scholar] [CrossRef]
  100. National Library of Medicine (U.S.). Double Blind, Placebo-Controlled Trial to Establish Safety and Efficacy of Ritlecitinib in Celiac Disease Patients in Remission. Available online: https://clinicaltrials.gov/study/NCT05636293?cond=celiac%20disease&intr=ritlecitinib&rank=1 (accessed on 11 July 2025).
  101. Choung, R.S.; Ramakrishna, J.; Pradhan, V.; King, B.; Guttman-Yassky, E.; Peeva, E.; Murray, J.A. Ritlecitinib, a JAK3 /TEC Inhibitor, Modulates the Markers of Celiac Autoimmunity in Alopecia Areata and Vitiligo Patients. Arch. Dermatol. Res. 2025, 317, 280. [Google Scholar] [CrossRef]
  102. Hjelm, M.; Shaikhkhalil, A.K. Celiac Disease in Patients with Cystic Fibrosis on Ivacaftor: A Case Series. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 257–260. [Google Scholar] [CrossRef] [PubMed]
  103. Safe, M.; Gifford, A.J.; Jaffe, A.; Ooi, C.Y. Resolution of Intestinal Histopathology Changes in Cystic Fibrosis after Treatment with Ivacaftor. Ann. Am. Thorac. Soc. 2016, 13, 297–298. [Google Scholar] [CrossRef]
  104. Villella, V.R.; Venerando, A.; Cozza, G.; Esposito, S.; Ferrari, E.; Monzani, R.; Spinella, M.C.; Oikonomou, V.; Renga, G.; Tosco, A.; et al. A Pathogenic Role for Cystic Fibrosis Transmembrane Conductance Regulator in Celiac Disease. EMBO J. 2019, 38, e100101. [Google Scholar] [CrossRef]
  105. Escudero-Hernández, C.; Martín, Á.; de Pedro Andrés, R.; Fernández-Salazar, L.; Garrote, J.A.; Bernardo, D.; Arranz, E. Circulating Dendritic Cells from Celiac Disease Patients Display a Gut-Homing Profile and Are Differentially Modulated by Different Gliadin-Derived Peptides. Mol. Nutr. Food Res. 2020, 64, e1900989. [Google Scholar] [CrossRef]
  106. Di Sabatino, A.; Rovedatti, L.; Rosado, M.M.; Carsetti, R.; Corazza, G.R.; MacDonald, T.T. Increased Expression of Mucosal Addressin Cell Adhesion Molecule 1 in the Duodenum of Patients with Active Celiac Disease Is Associated with Depletion of Integrin A4β7-Positive T Cells in Blood. Hum. Pathol. 2009, 40, 699–704. [Google Scholar] [CrossRef]
  107. Brown, N.K.; Guandalini, S.; Semrad, C.; Kupfer, S.S. A Clinician’s Guide to Celiac Disease HLA Genetics. Am. J. Gastroenterol. 2019, 114, 1587–1592. [Google Scholar] [CrossRef]
  108. Lei, H.; Crawford, M.S.; McCole, D.F. Jak-Stat Pathway Regulation of Intestinal Permeability: Pathogenic Roles and Therapeutic Opportunities in Inflammatory Bowel Disease. Pharmaceuticals 2021, 14, 840. [Google Scholar] [CrossRef] [PubMed]
  109. Yokoyama, S.; Perera, P.Y.; Waldmann, T.A.; Hiroi, T.; Perera, L.P. Tofacitinib, a Janus Kinase Inhibitor Demonstrates Efficacy in an IL-15 Transgenic Mouse Model That Recapitulates Pathologic Manifestations of Celiac Disease. J. Clin. Immunol. 2013, 33, 586–594. [Google Scholar] [CrossRef] [PubMed]
  110. Wauters, L.; Vanuytsel, T.; Hiele, M. Celiac Disease Remission with Tofacitinib: A Case Report. Ann. Intern. Med. 2020, 173, 585. [Google Scholar] [CrossRef]
  111. Kahn, J.S.; Moody, K.; Rosmarin, D. Significant Improvement of Dermatitis Herpetiformis with Tofacitinib. Dermatol. Online J. 2021, 27, 1–6. [Google Scholar] [CrossRef] [PubMed]
  112. Lenfant, M.; De Hertogh, G.; Verstockt, B. Tofacitinib for Celiac Disease and Microscopic Colitis: Killing Two Birds with One Stone. Acta Gastroenterol. Belg. 2023, 86, 374–376. [Google Scholar] [CrossRef]
  113. Maiuri, L.; Villella, V.R.; Raia, V.; Kroemer, G. The Gliadin-CFTR Connection: New Perspectives for the Treatment of Celiac Disease. Ital. J. Pediatr. 2019, 45, 40. [Google Scholar] [CrossRef]
  114. Daveson, A.J.M.; Ee, H.C.; Andrews, J.M.; King, T.; Goldstein, K.E.; Dzuris, J.L.; MacDougall, J.A.; Williams, L.J.; Treohan, A.; Cooreman, M.P.; 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. [Google Scholar] [CrossRef]
  115. Goel, G.; King, T.; Daveson, A.J.; Andrews, J.M.; Krishnarajah, J.; Krause, R.; Brown, G.J.E.; Fogel, R.; Barish, C.F.; Epstein, R.; et al. Epitope-Specific Immunotherapy Targeting CD4-Positive T Cells in Coeliac Disease: Two Randomised, Double-Blind, Placebo-Controlled Phase 1 Studies. Lancet Gastroenterol. Hepatol. 2017, 2, 479–493. [Google Scholar] [CrossRef] [PubMed]
  116. Truitt, K.E.; Daveson, A.J.M.; Ee, H.C.; Goel, G.; MacDougall, J.; Neff, K.; Anderson, R.P. Randomised Clinical Trial: A Placebo-Controlled Study of Subcutaneous or Intradermal NEXVAX2, an Investigational Immunomodulatory Peptide Therapy for Coeliac Disease. Aliment. Pharmacol. Ther. 2019, 50, 547–555. [Google Scholar] [CrossRef]
  117. Tye-Din, J.A.; Daveson, A.J.M.; Goel, G.; Goldstein, K.E.; Hand, H.L.; Neff, K.M.; Popp, A.; Taavela, J.; Maki, M.; Isola, J.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  118. Tye-Din, J. Discontinuation of the Nexvax2 Phase 2 Trial. Aust. Coeliac 2019, 21. [Google Scholar]
  119. Kelly, C.P.; Murray, J.A.; Leffler, D.A.; Getts, D.R.; Bledsoe, A.C.; Smithson, G.; First, M.R.; Morris, A.; Boyne, M.; Elhofy, A.; et al. TAK-101 Nanoparticles Induce Gluten-Specific Tolerance in Celiac Disease: A Randomized, Double-Blind, Placebo-Controlled Study. Gastroenterology 2021, 161, 66–80.e8. [Google Scholar] [CrossRef]
  120. National Library of Medicine (U.S.). Dose-Ranging Study of the Efficacy and Safety of TAK-101 for Prevention of Gluten-Specific T Cell Activation in Participants with Celiac Disease on a Gluten-Free Diet. Available online: https://clinicaltrials.gov/study/NCT04530123?intr=TAK%20101&rank=1 (accessed on 11 July 2025).
  121. Murray, J.A.; Wassaf, D.; Dunn, K.; Arora, S.; Winkle, P.; Stacey, H.; Cooper, S.; Goldstein, K.E.; Manchanda, R.; Kontos, S.; et al. Safety and Tolerability of KAN-101, a Liver-Targeted Immune Tolerance Therapy, in Patients with Coeliac Disease (ACeD): A Phase 1 Trial. Lancet Gastroenterol. Hepatol. 2023, 8, 735–747. [Google Scholar] [CrossRef]
  122. National Library of Medicine (U.S.). A Study of Safety, Tolerability, Pharmacodynamics, and Pharmacokinetics of KAN-101 in Celiac Disease (AceD-It). Available online: https://www.clinicaltrials.gov/study/NCT05574010 (accessed on 14 July 2025).
  123. National Library of Medicine (U.S.). A Study of Efficacy, Safety, and Tolerability of KAN-101 in People with Celiac Disease (SynCeD). Available online: https://clinicaltrials.gov/study/NCT06001177 (accessed on 14 July 2025).
  124. Topas Therapeutics Topas Therapeutics’ TPM502 Achieves Gluten-Specific Tolerance Induction, Positive Safety Profile in Phase 2a Trial in Celiac Disease Patients. Available online: https://topas-therapeutics.com/topas-therapeutics-tpm502-achieves-gluten-specific-tolerance-induction-positive-safety-profile-in-phase-2a-trial-in-celiac-disease-patients/ (accessed on 14 July 2025).
  125. National Library of Medicine (U.S.). A Study to Assess the Safety of TPM502 in Adults with Celiac Disease. Available online: https://clinicaltrials.gov/study/NCT05660109?term=TPM502&rank=1 (accessed on 14 July 2025).
  126. Corrêa, J. A Randomized, Double-Blind, Placebo-Controlled, Phase 1/2a Trial Protocol to Assess the Safety and Efficacy of TAK-101 Administered by Microneedles in Patients with Celiac Disease. Princ. Pract. Clin. Res. J. 2022, 8, 37–48. [Google Scholar] [CrossRef]
  127. Cenit, M.C.; Olivares, M.; Codoñer-Franch, P.; Sanz, Y. Intestinal Microbiota and Celiac Disease: Cause, Consequence or Co-Evolution? Nutrients 2015, 7, 6900–6923. [Google Scholar] [CrossRef]
  128. Kamada, N.; Seo, S.U.; Chen, G.Y.; Núñez, G. Role of the Gut Microbiota in Immunity and Inflammatory Disease. Nat. Rev. Immunol. 2013, 13, 321–335. [Google Scholar] [CrossRef]
  129. Reikvam, D.H.; Erofeev, A.; Sandvik, A.; Grcic, V.; Jahnsen, F.L.; Gaustad, P.; McCoy, K.D.; Macpherson, A.J.; Meza-Zepeda, L.A.; Johansen, F.E. Depletion of Murine Intestinal Microbiota: Effects on Gut Mucosa and Epithelial Gene Expression. PLoS ONE 2011, 6, e17996. [Google Scholar] [CrossRef] [PubMed]
  130. Collado, M.C.; Donat, E.; Ribes-Koninckx, C.; Calabuig, M.; Sanz, Y. Specific Duodenal and Faecal Bacterial Groups Associated with Paediatric Coeliac Disease. J. Clin. Pathol. 2009, 62, 264–269. [Google Scholar] [CrossRef]
  131. Collado, M.C.; Donat, E.; Ribes-Koninckx, C.; Calabuig, M.; Sanz, Y. Imbalances in Faecal and Duodenal Bifidobacterium Species Composition in Active and Non-Active Coeliac Disease. BMC Microbiol. 2008, 8, 232. [Google Scholar] [CrossRef]
  132. Di Cagno, R.; De Angelis, M.; De Pasquale, I.; Ndagijimana, M.; Vernocchi, P.; Ricciuti, P.; Gagliardi, F.; Laghi, L.; Crecchio, C.; Guerzoni, M.; et al. Duodenal and Faecal Microbiota of Celiac Children: Molecular, Phenotype and Metabolome Characterization. BMC Microbiol. 2011, 11, 219. [Google Scholar] [CrossRef] [PubMed]
  133. Pisarello, M.L.J.; Vintiñi, E.O.; González, S.N.; Pagani, F.; Medina, M.S. Decrease in Lactobacilli in the Intestinal Microbiota of Celiac Children with a Gluten-Free Diet, And Selection of Potentially Probiotic Strains. Can J. Microbiol. 2014, 61, 32–37. [Google Scholar] [CrossRef]
  134. Wacklin, P.; Laurikka, P.; Lindfors, K.; Collin, P.; Salmi, T.; Lähdeaho, M.L.; Saavalainen, P.; Mäki, M.; Mättö, J.; Kurppa, K.; et al. Altered Duodenal Microbiota Composition in Celiac Disease Patients Suffering from Persistent Symptoms on a Long-Term Gluten-Free Diet. Am. J. Gastroenterol. 2014, 109, 1933–1941. [Google Scholar] [CrossRef] [PubMed]
  135. Wacklin, P.; Kaukinen, K.; Tuovinen, E.; Collin, P.; Lindfors, K.; Partanen, J.; Mäki, M.; Mättuö, J. The Duodenal Microbiota Composition of Adult Celiac Disease Patients Is Associated with the Clinical Manifestation of the Disease. Inflamm. Bowel. Dis. 2013, 19, 934–941. [Google Scholar] [CrossRef]
  136. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  137. Smecuol, E.; Hwang, H.J.; Sugai, E.; Corso, L.; Cherñavsky, A.C.; Bellavite, F.P.; Gonza’lez, A.; Voda’novich, F.; Moreno, M.L.; Va’zquez, H.; et al. Exploratory, Randomized, Double-Blind, Placebo-Controlled Study on the Effects of Bifidobacterium Infantis Natren Life Start Strain Super Strain in Active Celiac Disease. J. Clin. Gastroenterol. 2013, 47, 139–147. [Google Scholar] [CrossRef] [PubMed]
  138. Smecuol, E.; Constante, M.; Temprano, M.P.; Costa, A.F.; Moreno, M.L.; Pinto-Sanchez, M.I.; Vázquez, H.; Stefanolo, J.P.; Gonzalez, A.F.; D’Adamo, C.R.; et al. Effect of Bifidobacterium Infantis NLS Super Strain in Symptomatic Coeliac Disease Patients on Long-Term Gluten-Free Diet—An Exploratory Study. Benef. Microbes 2020, 11, 527–534. [Google Scholar] [CrossRef]
  139. Klemenak, M.; Dolinšek, J.; Langerholc, T.; Di Gioia, D.; Mičetić-Turk, D. Administration of Bifidobacterium Breve Decreases the Production of TNF-α in Children with Celiac Disease. Dig. Dis. Sci. 2015, 60, 3386–3392. [Google Scholar] [CrossRef]
  140. Quagliariello, A.; Aloisio, I.; Cionci, N.B.; Luiselli, D.; D’Auria, G.; Martinez-Priego, L.; Pérez-Villarroya, D.; Langerholc, T.; Primec, M.; Mičetić-Turk, D.; et al. Effect of Bifidobacterium Breve on the Intestinal Microbiota of Coeliac Children on a Gluten Free Diet: A Pilot Study. Nutrients 2016, 8, 660. [Google Scholar] [CrossRef] [PubMed]
  141. Primec, M.; Klemenak, M.; Di Gioia, D.; Aloisio, I.; Bozzi Cionci, N.; Quagliariello, A.; Gorenjak, M.; Mičetić-Turk, D.; Langerholc, T. Clinical Intervention Using Bifidobacterium Strains in Celiac Disease Children Reveals Novel Microbial Modulators of TNF-α and Short-Chain Fatty Acids. Clin. Nutr. 2019, 38, 1373–1381. [Google Scholar] [CrossRef]
  142. Laparra, J.M.; Olivares, M.; Gallina, O.; Sanz, Y. Bifidobacterium Longum CECT 7347 Modulates Immune Responses in a Gliadin-Induced Enteropathy Animal Model. PLoS ONE 2012, 7, e30744. [Google Scholar] [CrossRef] [PubMed]
  143. Laparra, J.M.; Olivares, M.; Sanz, Y. Oral Administration of Bifidobacterium Longum CECT 7347 Ameliorates Gliadin-Induced Alterations in Liver Iron Mobilisation. Br. J. Nutr. 2013, 110, 1828–1836. [Google Scholar] [CrossRef]
  144. Olivares, M.; Castillejo, G.; Varea, V.; Sanz, Y. Double-Blind, Randomised, Placebo-Controlled Intervention Trial to Evaluate the Effects of Bifidobacterium Longum CECT 7347 in Children with Newly Diagnosed Coeliac Disease. Br. J. Nutr. 2014, 112, 30–40. [Google Scholar] [CrossRef]
  145. Di Pierro, F.; Bergomas, F.; Marraccini, P.; Ingenito, M.R.; Ferrari, L.; Vigna, L. Pilot Study on Non-Celiac Gluten Sensitivity: Effects of Bifidobacterium Longum Es1 Co-Administered with a Gluten-Free Diet. Minerva Gastroenterol. Dietol. 2020, 66, 187–193. [Google Scholar] [CrossRef]
  146. National Library of Medicine. Efficacy of Probiotic ES1 for the Treatment of Non-Celiac Gluten Sensitivity. Available online: https://clinicaltrials.gov/study/NCT02810301 (accessed on 15 July 2025).
  147. Naghibi, M.; Pont-Beltran, A.; Lamelas, A.; Llobregat, L.; Martinez-Blanch, J.F.; Rojas, A.; Álvarez, B.; López Plaza, B.; Arcos Castellanos, L.; Chenoll, E.; et al. Effect of Postbiotic Bifidobacterium Longum CECT 7347 on Gastrointestinal Symptoms, Serum Biochemistry, and Intestinal Microbiota in Healthy Adults: A Randomised, Parallel, Double-Blind, Placebo-Controlled Pilot Study. Nutrients 2024, 16, 3952. [Google Scholar] [CrossRef]
  148. Srivastava, S.; Basak, U.; Naghibi, M.; Vijayakumar, V.; Parihar, R.; Patel, J.; Jadon, P.S.; Pandit, A.; Dargad, R.R.; Khanna, S.; et al. A Randomized Double-Blind, Placebo-Controlled Trial to Evaluate the Safety and Efficacy of Live Bifidobacterium Longum CECT 7347 (ES1) and Heat-Treated Bifidobacterium Longum CECT 7347 (HT-ES1) in Participants with Diarrhea-Predominant Irritable Bowel Syndrome. Gut Microbes 2024, 16, 2338322. [Google Scholar] [CrossRef] [PubMed]
  149. Galipeau, H.J.; Wiepjes, M.; Motta, J.P.; Schulz, J.D.; Jury, J.; Natividad, J.M.; Pinto-Sanchez, I.; Sinclair, D.; Rousset, P.; Martin-Rosique, R.; et al. Novel Role of the Serine Protease Inhibitor Elafin in Gluten-Related Disorders. Am. J. Gastroenterol. 2014, 109, 748–756. [Google Scholar] [CrossRef]
  150. McCarville, J.L.; Dong, J.; Caminero, A.; Bermudez-Brito, M.; Jury, J.; Murray, J.A.; Duboux, S.; Steinmann, M.; Delley, M.; Tangyu, M.; et al. A Commensal Bifidobacterium Longum Strain Prevents Gluten-Related Immunopathology in Mice through Expression of a Serine Protease Inhibitor. Appl. Environ. Microbiol. 2017, 83, e01323-17. [Google Scholar] [CrossRef]
  151. National Library of Medicine (U.S.). Probiotic BL NCC 2705 and Gluten Sensitivity. Available online: https://clinicaltrials.gov/study/NCT03775499?intr=BL%20NCC&rank=1 (accessed on 15 July 2025).
  152. Lindfors, K.; Blomqvist, T.; Juuti-Uusitalo, K.; Stenman, S.; Venäläinen, J.; Mäki, M.; Kaukinen, K. Live Probiotic Bifidobacterium Lactis Bacteria Inhibit the Toxic Effects Induced by Wheat Gliadin in Epithelial Cell Culture. Clin. Exp. Immunol. 2008, 152, 552–558. [Google Scholar] [CrossRef]
  153. Oscarsson, E.; Håkansson, Å.; Andrén Aronsson, C.; Molin, G.; Agardh, D. Effects of Probiotic Bacteria Lactobacillaceae on the Gut Microbiota in Children with Celiac Disease Autoimmunity: A Placebo-Controlled and Randomized Clinical Trial. Front. Nutr. 2021, 8, 680771. [Google Scholar] [CrossRef] [PubMed]
  154. Håkansson, Å.; Aronsson, C.A.; Brundin, C.; Oscarsson, E.; Molin, G.; Agardh, D. Effects of Lactobacillus Plantarum and Lactobacillus Paracasei on the Peripheral Immune Response in Children with Celiac Disease Autoimmunity: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2019, 11, 1925. [Google Scholar] [CrossRef]
  155. Jenickova, E.; Andrén Aronsson, C.; Mascellani Bergo, A.; Cinek, O.; Havlik, J.; Agardh, D. Effects of Lactiplantibacillus Plantarum and Lacticaseibacillus Paracasei Supplementation on the Faecal Metabolome in Children with Coeliac Disease Autoimmunity: A Randomised, Double-Blinded Placebo-Controlled Clinical Trial. Front. Nutr. 2023, 10, 1183963. [Google Scholar] [CrossRef]
  156. National Library of Medicine (U.S.). Prevention Av Autoimmunitet Med Laktobaciller (PAL). Available online: https://clinicaltrials.gov/study/NCT04014660?intr=laktobaciller&rank=1 (accessed on 15 July 2025).
  157. Duar, R.M.; Clark, K.J.; Patil, P.B.; Hernández, C.; Brüning, S.; Burkey, T.E.; Madayiputhiya, N.; Taylor, S.L.; Walter, J. Identification and Characterization of Intestinal Lactobacilli Strains Capable of Degrading Immunotoxic Peptides Present in Gluten. J. Appl. Microbiol. 2015, 118, 515–527. [Google Scholar] [CrossRef]
  158. Kunduhoglu, B.; Hacioglu, S. Probiotic Potential and Gluten Hydrolysis Activity of Lactobacillus Brevis KT16-2. Probiotics Antimicrob. Proteins 2021, 13, 720–733. [Google Scholar] [CrossRef] [PubMed]
  159. D’Arienzo, R.; Maurano, F.; Luongo, D.; Mazzarella, G.; Stefanile, R.; Troncone, R.; Auricchio, S.; Ricca, E.; David, C.; Rossi, M. Adjuvant Effect of Lactobacillus Casei in a Mouse Model of Gluten Sensitivity. Immunol. Lett. 2008, 119, 78–83. [Google Scholar] [CrossRef]
  160. D’Arienzo, R.; Stefanile, R.; Maurano, F.; Mazzarella, G.; Ricca, E.; Troncone, R.; Auricchio, S.; Rossi, M. Immunomodulatory Effects of Lactobacillus Casei Administration in a Mouse Model of Gliadin-Sensitive Enteropathy. Scand. J. Immunol. 2011, 74, 335–341. [Google Scholar] [CrossRef]
  161. D’Arienzo, R.; Maurano, F.; Lavermicocca, P.; Ricca, E.; Rossi, M. Modulation of the Immune Response by Probiotic Strains in a Mouse Model of Gluten Sensitivity. Cytokine 2009, 48, 254–259. [Google Scholar] [CrossRef]
  162. Rashmi, B.S.; Gayathri, D. Draft Genome Sequence of the Gluten-Hydrolyzing Bacterium Bacillus Subtilis GS 188, Isolated from Wheat Sourdough. Genome Announc. 2017, 5, 10–1128. [Google Scholar] [CrossRef]
  163. Rashmi, B.S.; Gayathri, D. Molecular Characterization of Gluten Hydrolysing Bacillus Sp. and Their Efficacy and Biotherapeutic Potential as Probiotics Using Caco-2 Cell Line. J. Appl. Microbiol. 2017, 123, 759–772. [Google Scholar] [CrossRef]
  164. Sakandar, H.A.; Usman, K.; Imran, M. Isolation and Characterization of Gluten-Degrading Enterococcus Mundtii and Wickerhamomyces Anomalus, Potential Probiotic Strains from Indigenously Fermented Sourdough (Khamir). LWT 2018, 91, 271–277. [Google Scholar] [CrossRef]
  165. Tran, T.; Senger, S.; Baldassarre, M.; Brosnan, R.A.; Cristofori, F.; Crocco, M.; De Santis, S.; Elli, L.; Faherty, C.S.; Francavilla, R.; et al. Novel Bacteroides Vulgatus Strain Protects against Gluten-Induced Break of Human Celiac Gut Epithelial Homeostasis: A Pre-Clinical Proof-of-Concept Study. Pediatr. Res. 2024, 95, 1254–1264. [Google Scholar] [CrossRef] [PubMed]
  166. Martorell, P.; Alvarez, B.; Llopis, S.; Navarro, V.; Ortiz, P.; Gonzalez, N.; Balaguer, F.; Rojas, A.; Chenoll, E.; Ramón, D.; et al. Heat-Treated Bifidobacterium Longum CECT-7347: A Whole-Cell Postbiotic with Antioxidant, Anti-Inflammatory, and Gut-Barrier Protection Properties. Antioxidants 2021, 10, 536. [Google Scholar] [CrossRef] [PubMed]
  167. Harnett, J.; Myers, S.P.; Rolfe, M. Probiotics and the Microbiome in Celiac Disease: A Randomised Controlled Trial. Evid.-Based Complement. Altern. Med. 2016, 2016, 9048574. [Google Scholar] [CrossRef]
  168. De Angelis, M.; Rizzello, C.G.; Fasano, A.; Clemente, M.G.; De Simone, C.; Silano, M.; De Vincenzi, M.; Losito, I.; Gobbetti, M. VSL#3 Probiotic Preparation Has the Capacity to Hydrolyze Gliadin Polypeptides Responsible for Celiac Sprue. Biochim. Biophys. Acta Mol. Basis Dis. 2006, 1762, 80–93. [Google Scholar] [CrossRef]
  169. Francavilla, R.; Piccolo, M.; Francavilla, A.; Polimeno, L.; Semeraro, F.; Cristofori, F.; Castellaneta, S.; Barone, M.; Indrio, F.; Gobbetti, M.; et al. Clinical and Microbiological Effect of a Multispecies Probiotic Supplementation in Celiac Patients with Persistent IBS-Type Symptoms: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. J. Clin. Gastroenterol. 2019, 53, E117–E125. [Google Scholar] [CrossRef]
  170. Lionetti, E.; Dominijanni, V.; Iasevoli, M.; Cimadamore, E.; Acquaviva, I.; Gatti, S.; Monachesi, C.; Catassi, G.; Pino, A.; Faragalli, A.; et al. Effects of the Supplementation with a Multispecies Probiotic on Clinical and Laboratory Recovery of Children with Newly Diagnosed Celiac Disease: A Randomized, Placebo-Controlled Trial. Dig. Liver Dis. 2023, 55, 1328–1337. [Google Scholar] [CrossRef] [PubMed]
  171. Ferrari, E.; Monzani, R.; Saverio, V.; Gagliardi, M.; Pańczyszyn, E.; Raia, V.; Villella, V.R.; Bona, G.; Pane, M.; Amoruso, A.; et al. Probiotics Supplements Reduce Er Stress and Gut Inflammation Associated with Gliadin Intake in a Mouse Model of Gluten Sensitivity. Nutrients 2021, 13, 1221. [Google Scholar] [CrossRef] [PubMed]
  172. Papista, C.; Gerakopoulos, V.; Kourelis, A.; Sounidaki, M.; Kontana, A.; Berthelot, L.; Moura, I.C.; Monteiro, R.C.; Yiangou, M. Gluten Induces Coeliac-like Disease in Sensitised Mice Involving IgA, CD71 and Transglutaminase 2 Interactions That Are Prevented by Probiotics. Lab. Investig. 2012, 92, 625–635. [Google Scholar] [CrossRef] [PubMed]
  173. Croese, J.; Miller, G.C.; Marquart, L.; Llewellyn, S.; Gupta, R.; Becker, L.; Clouston, A.D.; Welch, C.; Sidorenko, J.; Wallace, L.; et al. Randomized, Placebo Controlled Trial of Experimental Hookworm Infection for Improving Gluten Tolerance in Celiac Disease. Clin. Transl. Gastroenterol. 2020, 11, e00274. [Google Scholar] [CrossRef]
  174. Daveson, A.J.; Jones, D.M.; Gaze, S.; McSorley, H.; Clouston, A.; Pascoe, A.; Cooke, S.; Speare, R.; Macdonald, G.A.; Anderson, R.; et al. Effect of Hookworm Infection on Wheat Challenge in Celiac Disease—A Randomised Double-Blinded Placebo Controlled Trial. PLoS ONE 2011, 6, e17366. [Google Scholar] [CrossRef]
  175. Giacomin, P.; Zakrzewski, M.; Jenkins, T.P.; Su, X.; Al-Hallaf, R.; Croese, J.; De Vries, S.; Grant, A.; Mitreva, M.; Loukas, A.; et al. Changes in Duodenal Tissue-Associated Microbiota Following Hookworm Infection and Consecutive Gluten Challenges in Humans with Coeliac Disease. Sci. Rep. 2016, 6, 36797. [Google Scholar] [CrossRef]
  176. Feruś, K.; Drabińska, N.; Krupa-Kozak, U.; Jarocka-Cyrta, E. A Randomized, Placebo-Controlled, Pilot Clinical Trial to Evaluate the Effect of Supplementation with Prebiotic Synergy 1 on Iron Homeostasis in Children and Adolescents with Celiac Disease Treated with a Gluten-Free Diet. Nutrients 2018, 10, 1818. [Google Scholar] [CrossRef]
  177. Krupa-Kozak, U.; Drabińska, N.; Jarocka-Cyrta, E. The Effect of Oligofructose-Enriched Inulin Supplementation on Gut Microbiota, Nutritional Status and Gastrointestinal Symptoms in Paediatric Coeliac Disease Patients on a Gluten-Free Diet: Study Protocol for a Pilot Randomized Controlled Trial. Nutr. J. 2017, 16, 47. [Google Scholar] [CrossRef]
  178. Drabińska, N.; Krupa-Kozak, U.; Ciska, E.; Jarocka-Cyrta, E. Plasma Profile and Urine Excretion of Amino Acids in Children with Celiac Disease on Gluten-Free Diet after Oligofructose-Enriched Inulin Intervention: Results of a Randomised Placebo-Controlled Pilot Study. Amino Acids 2018, 50, 1451–1460. [Google Scholar] [CrossRef]
  179. Drabińska, N.; Jarocka-Cyrta, E.; Markiewicz, L.H.; Krupa-Kozak, U. The Effect of Oligofructose-Enriched Inulin on Faecal Bacterial Counts and Microbiota-Associated Characteristics in Celiac Disease Children Following a Gluten-Free Diet: Results of a Randomized, Placebo-Controlled Trial. Nutrients 2018, 10, 201. [Google Scholar] [CrossRef] [PubMed]
  180. Francavilla, R.; De Angelis, M.; Rizzello, C.G.; Cavallo, N.; Dal Bello, F.; Gobbetti, M. Selected Probiotic Lactobacilli Have the Capacity to Hydrolyze Gluten Peptides during Simulated Gastrointestinal Digestion. Appl. Environ. Microbiol. 2017, 83, e00376-17. [Google Scholar] [CrossRef] [PubMed]
  181. Aoyama, H.; Hirata, T.; Sakugawa, H.; Watanabe, T.; Miyagi, S.; Maeshiro, T.; Chinen, T.; Kawane, M.; Zaha, O.; Nakayoshi, T.; et al. An Inverse Relationship Between Autoimmune Liver Diseases and Strongyloides Stercoralis Infection. Am. J. Trop. Med. Hyg. 2007, 76, 972–976. [Google Scholar] [CrossRef] [PubMed]
  182. Summers, R. Trichuris Suis Seems to Be Safe and Possibly Effective in the Treatment of Inflammatory Bowel Disease. Am. J. Gastroenterol. 2003, 98, 2034–2041. [Google Scholar] [CrossRef]
  183. Ruyssers, N.E.; De Winter, B.Y.; De Man, J.G.; Ruyssers, N.D.; Van Gils, A.J.; Loukas, A.; Pearson, M.S.; Weinstock, J.V.; Pelckmans, P.A.; Moreels, T.G. Schistosoma Mansoni Proteins Attenuate Gastrointestinal Motility Disturbances during Experimental Colitis in Mice. World J. Gastroenterol. 2010, 16, 703–712. [Google Scholar] [CrossRef]
  184. Summers, R.W.; Elliott, D.E.; Urban, J.F.; Thompson, R.A.; Weinstock, J.V. Trichuris Suis Therapy for Active Ulcerative Colitis: A Randomized Controlled Trial. Gastroenterology 2005, 128, 825–832. [Google Scholar] [CrossRef]
  185. Correale, J.; Farez, M. Association between Parasite Infection and Immune Responses in Multiple Sclerosis. Ann. Neurol. 2007, 61, 97–108. [Google Scholar] [CrossRef]
  186. Bager, P.; Arnved, J.; Rønborg, S.; Wohlfahrt, J.; Poulsen, L.K.; Westergaard, T.; Petersen, H.W.; Kristensen, B.; Thamsborg, S.; Roepstorff, A.; et al. Trichuris Suis Ova Therapy for Allergic Rhinitis: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. J. Allergy Clin. Immunol. 2010, 125, 123–130.e3. [Google Scholar] [CrossRef]
  187. Drabińska, N.; Krupa-Kozak, U.; Jarocka-Cyrta, E. Intestinal Permeability in Children with Celiac Disease after the Administration of Oligofructose-Enriched Inulin into a Gluten-Free Diet—Results of a Randomized, Placebo-Controlled, Pilot Trial. Nutrients 2020, 12, 1736. [Google Scholar] [CrossRef]
  188. van Beurden, Y.H.; van Gils, T.; van Gils, N.A.; Kassam, Z.; Mulder, C.J.J.; Aparicio-Pagés, N. Serendipity in Refractory Celiac Disease: Full Recovery of Duodenal Villi and Clinical Symptoms after Fecal Microbiota Transfer. J. Gastrointest. Liver Dis. 2016, 25, 385–388. [Google Scholar] [CrossRef]
  189. National Library of Medicine (U.S.). Safety and Efficacy of Fecal Microbiota Transplantation. Available online: https://clinicaltrials.gov/study/NCT04014413?intr=fecal%20microbiota%20transplantation&page=2&rank=18 (accessed on 16 July 2025).
  190. Singh, S.; Razak, M.A.; Sangam, S.R.; Viswanath, B.; Begum, P.S.; Rajagopal, S. The Impact of Functional Food and Nutraceuticals in Health. In Therapeutic Foods; Elsevier: Amsterdam, The Netherlands, 2018; pp. 23–47. [Google Scholar]
  191. Jahdkaran, M.; Asri, N.; Esmaily, H.; Rostami-Nejad, M. Potential of Nutraceuticals in Celiac Disease. Tissue Barriers 2024, 13, 2374628. [Google Scholar] [CrossRef]
  192. Singla, R.K.; Dubey, A.K.; Garg, A.; Sharma, R.K.; Fiorino, M.; Ameen, S.M.; Haddad, M.A.; Al-Hiary, M. Natural Polyphenols: Chemical Classification, Definition of Classes, Subcategories, and Structures. J. AOAC Int. 2019, 102, 1397–1400. [Google Scholar] [CrossRef]
  193. Van Buiten, C.B.; Elias, R.J. Gliadin Sequestration as a Novel Therapy for Celiac Disease: A Prospective Application for Polyphenols. Int. J. Mol. Sci. 2021, 22, 595. [Google Scholar] [CrossRef] [PubMed]
  194. Van Buiten, C.B.; Lambert, J.D.; Elias, R.J. Green Tea Polyphenols Mitigate Gliadin-Mediated Inflammation and Permeability in Vitro. Mol. Nutr. Food Res. 2018, 62, e1700879. [Google Scholar] [CrossRef]
  195. Xie, F.; Huang, Q.; Fang, F.; Chen, S.; Wang, Z.; Wang, K.; Fu, X.; Zhang, B. Effects of Tea Polyphenols and Gluten Addition on in Vitro Wheat Starch Digestion Properties. Int. J. Biol. Macromol. 2019, 126, 525–530. [Google Scholar] [CrossRef] [PubMed]
  196. Dias, R.; Brás, N.F.; Pérez-Gregorio, M.; Fernandes, I.; Mateus, N.; Freitas, V. A Multi-Spectroscopic Study on the Interaction of Food Polyphenols with a Bioactive Gluten Peptide: From Chemistry to Biological Implications. Food Chem. 2019, 299, 125051. [Google Scholar] [CrossRef]
  197. Pérot, M.; Lupi, R.; Guyot, S.; Delayre-Orthez, C.; Gadonna-Widehem, P.; Thébaudin, J.Y.; Bodinier, M.; Larré, C. Polyphenol Interactions Mitigate the Immunogenicity and Allergenicity of Gliadins. J. Agric. Food Chem. 2017, 65, 6442–6451. [Google Scholar] [CrossRef]
  198. Taddei, P.; Zanna, N.; Tozzi, S. Raman Characterization of the Interactions between Gliadins and Anthocyanins. J. Raman Spectrosc. 2013, 44, 1435–1439. [Google Scholar] [CrossRef]
  199. Tozzi, S.; Zanna, N.; Taddei, P. Study on the Interaction between Gliadins and a Coumarin as Molecular Model System of the Gliadins-Anthocyanidins Complexes. Food Chem. 2013, 141, 3586–3597. [Google Scholar] [CrossRef]
  200. Wang, Q.; Tang, Y.; Yang, Y.; Zhao, J.; Zhang, Y.; Li, L.; Wang, Q.; Ming, J. Interaction between Wheat Gliadin and Quercetin under Different PH Conditions Analyzed by Multi-Spectroscopy Methods. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2020, 229, 117937. [Google Scholar] [CrossRef]
  201. Yu, T.; Xie, Y.; Wang, Z.; Li, J.; Shen, Y.; Yuan, J.; Gao, J.; Fakruddin, M.; Wu, Y.; Chen, H. Quercetin Ameliorates Celiac-Related Intestinal Inflammation Caused by Wheat Gluten through Modulating Oxidative Stress, Th1/Th2/Treg Balance, and Intestinal Microflora Structure. Food Funct. 2024, 15, 9343–9356. [Google Scholar] [CrossRef]
  202. Pérez-Gregorio, M.R.; Bessa Pereira, C.; Dias, R.; Mateus, N.; De Freitas, V. New-Level Insights into the Effects of Grape Seed Polyphenols on the Intestinal Processing and Transport of a Celiac Disease Immunodominant Peptide. J. Agric. Food Chem. 2021, 69, 13474–13486. [Google Scholar] [CrossRef]
  203. Wang, N.; Cui, C.; Xu, C.; Ren, H.; Wang, F.; Yu, Q.; Zhang, G. Effect and Mechanism of Peanut Skin Proanthocyanidins on Gliadin-Induced Caco-2 Celiac Disease Model Cells. Clin. Immunol. 2022, 245, 109100. [Google Scholar] [CrossRef]
  204. Klusóczki, Á.; Oláh, B.; Hosszú, D.; Fenyvesi, F.; Remenyik, J.; Homoki, J.; Gyöngyösi, A.; Bácskay, I.; Váradi, J. Effectiveness of Anthocyanin-Rich Sour Cherry Extract on Gliadin-Induced Caco-2 Barrier Damage. Nutrients 2023, 15, 4022. [Google Scholar] [CrossRef]
  205. Medjeber, O.; Touri, K.; Rafa, H.; Djeraba, Z.; Belkhelfa, M.; Boutaleb, A.F.; Arroul-Lammali, A.; Belguendouz, H.; Touil-Boukoffa, C. Ex Vivo Immunomodulatory Effect of Ethanolic Extract of Propolis during Celiac Disease: Involvement of Nitric Oxide Pathway. Inflammopharmacology 2018, 26, 1469–1481. [Google Scholar] [CrossRef]
  206. Garzarella, E.U.; Navajas-Porras, B.; Pérez-Burillo, S.; Ullah, H.; Esposito, C.; Santarcangelo, C.; Hinojosa-Nogueira, D.; Pastoriza, S.; Zaccaria, V.; Xiao, J.; et al. Evaluating the Effects of a Standardized Polyphenol Mixture Extracted from Poplar-Type Propolis on Healthy and Diseased Human Gut Microbiota. Biomed. Pharmacother. 2022, 148, 112759. [Google Scholar] [CrossRef] [PubMed]
  207. Gupta, K.B.; Mantha, A.K.; Dhiman, M. Mitigation of Gliadin-Induced Inflammation and Cellular Damage by Curcumin in Human Intestinal Cell Lines. Inflammation 2021, 44, 873–889. [Google Scholar] [CrossRef]
  208. Yu, T.; Xie, Y.; Yuan, J.; Gao, J.; Xiao, Z.; Wu, Y.; Chen, H. The Nutritional Intervention of Resveratrol Can Effectively Alleviate the Intestinal Inflammation Associated with Celiac Disease Induced by Wheat Gluten. Front. Immunol. 2022, 13, 878186. [Google Scholar] [CrossRef] [PubMed]
  209. De Stefano, D.; Maiuri, M.C.; Simeon, V.; Grassia, G.; Soscia, A.; Cinelli, M.P.; Carnuccio, R. Lycopene, Quercetin and Tyrosol Prevent Macrophage Activation Induced by Gliadin and IFN-γ. Eur. J. Pharmacol. 2007, 566, 192–199. [Google Scholar] [CrossRef]
  210. Périz, M.; Rodríguez-Lagunas, M.J.; Pérez-Cano, F.J.; Best, I.; Pastor-Soplin, S.; Castell, M.; Massot-Cladera, M. Influence of Consumption of Two Peruvian Cocoa Populations on Mucosal and Systemic Immune Response in an Allergic Asthma Rat Model. Nutrients 2022, 14, 410. [Google Scholar] [CrossRef] [PubMed]
  211. Abril-Gil, M.; Massot-Cladera, M.; Pérez-Cano, F.J.; Castellote, C.; Franch, À.; Castell, M. A Diet Enriched with Cocoa Prevents IgE Synthesis in a Rat Allergy Model. Pharmacol. Res. 2012, 65, 603–608. [Google Scholar] [CrossRef]
  212. Ramiro-Puig, E.; Casadesús, G.; Lee, H.G.; Zhu, X.; McShea, A.; Perry, G.; Pérez-Cano, F.J.; Smith, M.A.; Castell, M. Neuroprotective Effect of Cocoa Flavonids on in Vitro Oxidative Stress. Eur. J. Nutr. 2009, 48, 54–61. [Google Scholar] [CrossRef] [PubMed]
  213. Massot-Cladera, M.; Abril-Gil, M.; Torres, S.; Franch, À.; Castell, M.; Pérez-Cano, F.J. Impact of Cocoa Polyphenol Extracts on the Immune System and Microbiota in Two Strains of Young Rats. Br. J. Nutr. 2014, 112, 1944–1954. [Google Scholar] [CrossRef] [PubMed]
  214. Camps-Bossacoma, M.; Garcia-Aloy, M.; Saldana-Ruiz, S.; Cambras, T.; González-Domínguez, R.; Franch, À.; Pérez-Cano, F.J.; Andres-Lacueva, C.; Castell, M. Role of Theobromine in Cocoa’s Metabolic Properties in Healthy Rats. J. Agric. Food Chem. 2019, 67, 3605–3614. [Google Scholar] [CrossRef]
  215. Camps-Bossacoma, M.; Pérez-Cano, F.J.; Franch, À.; Castell, M. Theobromine Is Responsible for the Effects of Cocoa on the Antibody Immune Status of Rats. J. Nutr. 2018, 148, 464–471. [Google Scholar] [CrossRef]
  216. Franco, R.; Oñatibia-Astibia, A.; Martínez-Pinilla, E. Health Benefits of Methylxanthines in Cacao and Chocolate. Nutrients 2013, 5, 4159–4173. [Google Scholar] [CrossRef]
  217. Massot-Cladera, M.; Franch, À.; Pérez-Cano, F.J.; Castell, M. Cocoa and Cocoa Fibre Differentially Modulate IgA and IgM Production at Mucosal Sites. Br. J. Nutr. 2016, 115, 1539–1546. [Google Scholar] [CrossRef] [PubMed]
  218. Kramer, K.; Yeboah-Awudzi, M.; Magazine, N.; King, J.M.; Xu, Z.; Losso, J.N. Procyanidin B2 Rich Cocoa Extracts Inhibit Inflammation in Caco-2 Cell Model of in Vitro Celiac Disease by down-Regulating Interferon-Gamma- or Gliadin Peptide 31-43-Induced Transglutaminase-2 and Interleukin-15. J. Funct. Foods 2019, 57, 112–120. [Google Scholar] [CrossRef]
  219. Raguzzini, A.; Poce, G.; Consalvi, S.; Toti, E.; Palmacci, F.; Biava, M.; Peluso, I. Chocolate Consumers and Lymphocyte-to-Monocyte Ratio: A Working Hypothesis from a Preliminary Report of a Pilot Study in Celiac Subjects. Antioxidants 2019, 8, 440. [Google Scholar] [CrossRef]
  220. Du, J.; Chen, S.; Shi, J.; Zhu, X.; Ying, H.; Zhang, Y.; Chen, S.; Shen, B.; Li, J. The Association between the Lymphocyte-Monocyte Ratio and Disease Activity in Rheumatoid Arthritis. Clin. Rheumatol. 2017, 36, 2689–2695. [Google Scholar] [CrossRef]
  221. Girbal González, M.; Rodríguez-Banqueri, A.; Eckhard, U.; Gomis-Rüth, F.; Rodríguez-Lagunas, M.J.; Franch, À.; Pérez-Cano, F.J. Caracterización de La Composición y La Función de La Microbiota Intestinal En Un Modelo Preclínico de Celiaquía. XVI Workshop Soc. Española Microbiota Probióticos Y Prebióticos Barc. 2025, 6, 149. [Google Scholar]
  222. Girbal González, M.; Mendes, S.; Rodríguez-Banqueri, A.; Eckhard, U.; Gomis-Rüth, F.X.; Rodríguez-Lagunas, M.J.; Franch, À.; Pérez-Cano, F.J. Coeliac Disease Progression Is Preclinically Modulated by Cocoa Intake. In Proceedings of the 57th Annual Meeting of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN), Abstract E-books 2025, Helsinki, Finland, 14–17 May 2025; Volume 6, p. 997. [Google Scholar]
  223. Paterson, D.; Pierce, M.; Peck, E. The Treatment of Coeliac Disease with Vitamin B Complex and Concentrated Liver. Arch. Dis. Child. 1944, 19, 99–110. [Google Scholar] [CrossRef]
  224. Dickey, W.; Ward, M.; Whittle, C.R.; Kelly, M.T.; Pentieva, K.; Horigan, G.; Patton, S.; Mcnulty, H. Homocysteine and Related B-Vitamin Status in Coeliac Disease: Effects of Gluten Exclusion and Histological Recovery. Scand. J. Gastroenterol. 2008, 43, 682–688. [Google Scholar] [CrossRef] [PubMed]
  225. Valente, F.X.; Do Nascimento Campos, T.; De Sousa Moraes, L.F.; Miranda Hermsdorff, H.H.; De Morais Cardoso, L.; Pinheiro-Sant’ana, H.M.; Barros Gilberti, F.A.; Do Carmo Gouveia Peluzio, M. B Vitamins Related to Homocysteine Metabolism in Adults Celiac Disease Patients: A Cross-Sectional Study. Nutr. J. 2015, 14, 110. [Google Scholar] [CrossRef] [PubMed]
  226. Tokgöz, Y.; Terlemez, S.; Karul, A. Fat Soluble Vitamin Levels in Children with Newly Diagnosed Celiac Disease, a Case Control Study. BMC Pediatr. 2018, 18, 130. [Google Scholar] [CrossRef]
  227. Dong, S.; Singh, T.P.; Wei, X.; Yao, H.; Wang, H. Protective Effect of 1,25-Dihydroxy Vitamin D3 on Pepsin–Trypsin-Resistant Gliadin-Induced Tight Junction Injuries. Dig. Dis. Sci. 2018, 63, 92–104. [Google Scholar] [CrossRef]
  228. Piątek-Guziewicz, A.; Dąbek, A.; Przybylska-Feluś, M.; Zagrodzki, P.; Mach, T.; Zwolińska-Wcisło, M. Oral Vitamin E Supplementation in Reducing Nitrosative Stress in Adults Treated for Celiac Disease. Pol. Arch. Intern. Med. 2020, 130, 711–713. [Google Scholar] [CrossRef]
  229. Bascuñán, K.A.; Araya, M.; Rodríguez, J.M.; Roncoroni, L.; Elli, L.; Alvarez, J.D.P.L.; Valenzuela, R. Interplay of N-3 Polyunsaturated Fatty Acids, Intestinal Inflammation, and Gut Microbiota in Celiac Disease Pathogenesis. Nutrients 2025, 17, 621. [Google Scholar] [CrossRef] [PubMed]
  230. Vincentini, O.; Quaranta, M.G.; Viora, M.; Agostoni, C.; Silano, M. Docosahexaenoic Acid Modulates in Vitro the Inflammation of Celiac Disease in Intestinal Epithelial Cells via the Inhibition of CPLA 2. Clin. Nutr. 2011, 30, 541–546. [Google Scholar] [CrossRef]
  231. Costa, A.; Brito, G.A.P.D. Aerobic Exercise Associated with Fish Oil Supplementation Decreases C-Reactive Protein and Interleukin-6 in Celiac Disease Patients. J. Nutr. Metab. 2022, 2022, 3908675. [Google Scholar] [CrossRef]
  232. Osman, M.T.; Al-Duboni, G.; Taha, B.I.; Muhamed, L.A. Refractory Coeliac Disease; Role of Nigella Sativa as Immunomodulator. J. Adv. Med. Med. Res. 2012, 2, 527–535. [Google Scholar] [CrossRef]
  233. Sonzogni, E.; Martinelli, G.; Fumagalli, M.; Maranta, N.; Pozzoli, C.; Bani, C.; Marrari, L.A.; Di Lorenzo, C.; Sangiovanni, E.; Dell’Agli, M.; et al. In Vitro Insights into the Dietary Role of Glucoraphanin and Its Metabolite Sulforaphane in Celiac Disease. Nutrients 2024, 16, 2743. [Google Scholar] [CrossRef]
  234. Murray, M.; Dordevic, A.L.; Ryan, L.; Bonham, M.P. An Emerging Trend in Functional Foods for the Prevention of Cardiovascular Disease and Diabetes: Marine Algal Polyphenols. Crit. Rev. Food Sci. Nutr. 2018, 58, 1342–1358. [Google Scholar] [CrossRef]
  235. van der Linde, C.; Barone, M.; Turroni, S.; Brigidi, P.; Keleszade, E.; Swann, J.R.; Costabile, A. An in Vitro Pilot Fermentation Study on the Impact of Chlorella Pyrenoidosa on Gut Microbiome Composition and Metabolites in Healthy and Coeliac Subjects. Molecules 2021, 26, 2330. [Google Scholar] [CrossRef]
  236. Pourmohammadi, K.; Abedi, E. Hydrolytic Enzymes and Their Directly and Indirectly Effects on Gluten and Dough Properties: An Extensive Review. Food Sci. Nutr. 2021, 9, 3988–4006. [Google Scholar] [CrossRef] [PubMed]
  237. Akeroyd, M.; Van Zandycke, S.; Den Hartog, J.; Mutsaers, J.; Edens, L.; Van DenBerg, M.; Christis, C. AN-PEP, Proline-Specific Endopeptidase, Degrades All Known Immunostimulatory Gluten Peptides in Beer Made from Barley Malt. J. Am. Soc. Brew. Chem. 2016, 74, 91–99. [Google Scholar] [CrossRef]
  238. Hooshiyar, M.; Ebrahimipour, G.; Baghaei, K.; Saadi, M.I. Investigating the Effect of Cloned AN-PEP Enzyme in Yeast on Gluten Degradation and Rheological Features of Dough. Infect. Epidemiol Microbiol. 2021, 7, 337–345. [Google Scholar] [CrossRef]
  239. Hardt, N.A.; Boom, R.M.; van der Goot, A.J. Starch Facilitates Enzymatic Wheat Gluten Hydrolysis. LWT 2015, 61, 557–563. [Google Scholar] [CrossRef]
  240. Merz, M.; Kettner, L.; Langolf, E.; Appel, D.; Blank, I.; Stressler, T.; Fischer, L. Production of Wheat Gluten Hydrolysates with Reduced Antigenicity Employing Enzymatic Hydrolysis Combined with Downstream Unit Operations. J. Sci. Food Agric. 2016, 96, 3358–3364. [Google Scholar] [CrossRef] [PubMed]
  241. Mandile, R.; Picascia, S.; Parrella, C.; Camarca, A.; Gobbetti, M.; Greco, L.; Troncone, R.; Gianfrani, C.; Auricchio, R. Lack of Immunogenicity of Hydrolysed Wheat Flour in Patients with Coeliac Disease after a Short-Term Oral Challenge. Aliment. Pharmacol. Ther. 2017, 46, 440–446. [Google Scholar] [CrossRef]
  242. Gallo, M.; Nigro, F.; Passannanti, F.; Nanayakkara, M.; Lania, G.; Parisi, F.; Salameh, D.; Budelli, A.; Barone, M.V.; Nigro, R. Effect of PH Control during Rice Fermentation in Preventing a Gliadin P31-43 Entrance in Epithelial Cells. Int. J. Food. Sci. Nutr. 2019, 70, 950–958. [Google Scholar] [CrossRef] [PubMed]
  243. Păcularu-Burada, B.; Ceoromila (Cantaragiu), A.M.; Vasile, M.A.; Bahrim, G.E. Novel Insights into Different Kefir Grains Usefulness as Valuable Multiple Starter Cultures to Achieve Bioactive Gluten-Free Sourdoughs. LWT 2022, 165, 113670. [Google Scholar] [CrossRef]
  244. Zhou, Y.; Ouyang, B.; Duan, M.; Lv, X.; Zhou, X. Biological Characteristics of the Gluten-Free Sourdough System Fermented by Lactobacillus Plantarum ST-III and Its Effect on Dough Quality and Nutritional Value during Freezing. Food Chem. X 2022, 14, 100350. [Google Scholar] [CrossRef]
  245. Jouanin, A.; Gilissen, L.J.W.J.; Boyd, L.A.; Cockram, J.; Leigh, F.J.; Wallington, E.J.; van den Broeck, H.C.; van der Meer, I.M.; Schaart, J.G.; Visser, R.G.F.; et al. Food Processing and Breeding Strategies for Coeliac-Safe and Healthy Wheat Products. Food Res. Int. 2018, 110, 11–21. [Google Scholar] [CrossRef]
  246. Ribeiro, M.; Rodriguez-Quijano, M.; Nunes, F.M.; Carrillo, J.M.; Branlard, G.; Igrejas, G. New Insights into Wheat Toxicity: Breeding Did Not Seem to Contribute to a Prevalence of Potential Celiac Disease’s Immunostimulatory Epitopes. Food Chem. 2016, 213, 8–18. [Google Scholar] [CrossRef] [PubMed]
  247. Camerlengo, F.; Sestili, F.; Silvestri, M.; Colaprico, G.; Margiotta, B.; Ruggeri, R.; Lupi, R.; Masci, S.; Lafiandra, D. Production and Molecular Characterization of Bread Wheat Lines with Reduced Amount of α-Type Gliadins. BMC Plant Biol. 2017, 17, 11–21. [Google Scholar] [CrossRef]
  248. Jouanin, A.; Schaart, J.G.; Boyd, L.A.; Cockram, J.; Leigh, F.J.; Bates, R.; Wallington, E.J.; Visser, R.G.F.; Smulders, M.J.M. Outlook for Coeliac Disease Patients: Towards Bread Wheat with Hypoimmunogenic Gluten by Gene Editing of α- And γ-Gliadin Gene Families. BMC Plant Biol. 2019, 19, 33. [Google Scholar] [CrossRef]
  249. Jouanin, A.; Boyd, L.; Visser, R.G.F.; Smulders, M.J.M. Development of Wheat with Hypoimmunogenic Gluten Obstructed by the Gene Editing Policy in Europe. Front. Plant Sci. 2018, 9, 1523. [Google Scholar] [CrossRef]
  250. Zhang, Y.; Wu, H.; Huilian, C.; Fu, L. Detoxification Mechanism and the Impact of Transamidation-Modified Gliadin on Celiac-Based Gluten Sensitivity: The Potential of Unlocking Gluten Tolerance in Functional Food. J. Agric. Food Chem. 2025, 73, 1682–1693. [Google Scholar] [CrossRef] [PubMed]
  251. Gil-Humanes, J.; Pistón, F.; Tollefsen, S.; Sollid, L.M.; Barro, F. Effective Shutdown in the Expression of Celiac Disease-Related Wheat Gliadin T-Cell Epitopes by RNA Interference. Proc. Natl. Acad. Sci. USA 2010, 107, 17023–17028. [Google Scholar] [CrossRef]
  252. Gianfrani, C.; Siciliano, R.A.; Facchiano, A.M.; Camarca, A.; Mazzeo, M.F.; Costantini, S.; Salvati, V.M.; Maurano, F.; Mazzarella, G.; Iaquinto, G.; et al. Transamidation of Wheat Flour Inhibits the Response to Gliadin of Intestinal T Cells in Celiac Disease. Gastroenterology 2007, 133, 780–789. [Google Scholar] [CrossRef] [PubMed]
  253. Mazzarella, G.; Salvati, V.M.; Iaquinto, G.; Stefanile, R.; Capobianco, F.; Luongo, D.; Bergamo, P.; Maurano, F.; Giardullo, N.; Malamisura, B.; et al. Reintroduction of Gluten Following Flour Transamidation in Adult Celiac Patients: A Randomized, Controlled Clinical Study. Clin. Dev. Immunol. 2012, 2012, 329150. [Google Scholar] [CrossRef]
  254. Heredia-Sandoval, N.G.; Islas-Rubio, A.R.; Cabrera-Chávez, F.; Calderón De La Barca, A.M. Transamidation of Gluten Proteins during the Bread-Making Process of Wheat Flour to Produce Breads with Less Immunoreactive Gluten. Food Funct. 2014, 5, 1813–1818. [Google Scholar] [CrossRef]
  255. De Sena, V.; Mazzarella, G.; Rossi, M. Effects of Transamidation vs. Fermentation to Reduce the Gluten Content of the Triticum Monococcum Wheat Cultivar Hammurabi: Analysis of Biochemical, Baking and Sensory Parameters. Int. J. Food Sci. Technol. 2024, 59, 1927–1934. [Google Scholar] [CrossRef]
  256. Lamacchia, C.; Di Luccia, A.; Gianfrani, C. Method for the Detoxification of Gluten Proteins from Grains of Cereal 2015. U.S. Patent Application No. 14/432,461, 1 October 2015. [Google Scholar]
  257. Open Access Government Gluten Friendly Technology Fixes Proteins in Wheat Kernels. Available online: https://www.openaccessgovernment.org/gluten-friendly-technology-kernels/36411/ (accessed on 17 July 2025).
  258. Andriulli, A.; Bevilacqua, A.; Palmieri, O.; Latiano, A.; Fontana, R.; Gioffreda, D.; Castellana, S.; Mazza, T.; Panza, A.; Menzaghi, C.; et al. Healthy and Pro-Inflammatory Gut Ecology Plays a Crucial Role in the Digestion and Tolerance of a Novel Gluten FriendlyTM Bread in Celiac Subjects: A Randomized, Double Blind, Placebo Control in Vivo Study. Food Funct. 2022, 13, 1299–1315. [Google Scholar] [CrossRef] [PubMed]
  259. National Library of Medicine (U.S.). In Vivo Effects of the Gluten Friendly Bread in Coeliac Disease. Available online: https://clinicaltrials.gov/study/NCT03168490?intr=gluten%20friendly&rank=1disease (accessed on 17 July 2025).
Figure 1. Pathophysiology of CeD and site of action inhibited, modulated or promoted by all the existent therapeutic approaches to prevent, cure or ameliorate symptomatology of this disease. Current treatments can be classified into seven main groups based on their targeted step within the gluten-derived immunological cascade: gluten tolerization, gluten-degrading enzymes, gluten neutralization, IP modulators, immune modulators (CFTR promoters, TG2, IL-15, HLA-DQ2, JAK or lymphocyte migration inhibitors), pre-, pro- and synbiotics, nutraceuticals and modification of gluten-containing foodstuffs. The only currently accepted treatment for CeD is a GFD.
Figure 1. Pathophysiology of CeD and site of action inhibited, modulated or promoted by all the existent therapeutic approaches to prevent, cure or ameliorate symptomatology of this disease. Current treatments can be classified into seven main groups based on their targeted step within the gluten-derived immunological cascade: gluten tolerization, gluten-degrading enzymes, gluten neutralization, IP modulators, immune modulators (CFTR promoters, TG2, IL-15, HLA-DQ2, JAK or lymphocyte migration inhibitors), pre-, pro- and synbiotics, nutraceuticals and modification of gluten-containing foodstuffs. The only currently accepted treatment for CeD is a GFD.
Nutrients 17 02960 g001
Figure 2. Current research on gluten-degrading enzymes, origin and research status: (1) in vitro, (2) in vivo, (3) clinical trial or (4) commercialized.
Figure 2. Current research on gluten-degrading enzymes, origin and research status: (1) in vitro, (2) in vivo, (3) clinical trial or (4) commercialized.
Nutrients 17 02960 g002
Table 2. Gluten neutralization and intestinal permeability modulation strategies.
Table 2. Gluten neutralization and intestinal permeability modulation strategies.
StrategyActive PrincipleNamePhase, Clinical TrialSponsorRef.
Gluten neutralizationAnti-gliadin IgY from the egg yolk of hypersensitized hensAGY-010Phase 1 (NCT01765647),
Phase 2 (NCT03707730)
Igy Inc.[53,54,55]
(P(HEMA-co-SS)) copolymerBL-7010Phase 1 and 2 (NCT01990885)BioLineRx, Ltd.[56,57,58]
Intestinal permeability modulatorsOctapeptide inhibitor of paracellular
permeability (TJs modulator)
Larazotide acetate (AT-1001)Phase 1 (NCT00386165, NCT00386490), Phase 2 (NCT00362856, NCT00492960, NCT00620451, NCT00889473, NCT01396213), Phase 3–terminated (NCT03569007)9 Meters Biopharma, Inc.[59,60,61,62,63]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Girbal-González, M.; Pérez-Cano, F.J. Is There a Future Without Gluten Restrictions for Celiac Patients? Update on Current Treatments. Nutrients 2025, 17, 2960. https://doi.org/10.3390/nu17182960

AMA Style

Girbal-González M, Pérez-Cano FJ. Is There a Future Without Gluten Restrictions for Celiac Patients? Update on Current Treatments. Nutrients. 2025; 17(18):2960. https://doi.org/10.3390/nu17182960

Chicago/Turabian Style

Girbal-González, Marina, and Francisco J. Pérez-Cano. 2025. "Is There a Future Without Gluten Restrictions for Celiac Patients? Update on Current Treatments" Nutrients 17, no. 18: 2960. https://doi.org/10.3390/nu17182960

APA Style

Girbal-González, M., & Pérez-Cano, F. J. (2025). Is There a Future Without Gluten Restrictions for Celiac Patients? Update on Current Treatments. Nutrients, 17(18), 2960. https://doi.org/10.3390/nu17182960

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop