Next Article in Journal
Different Approaches to Study Molecular Blueprint and Biological Behavior of Brain Tumors: Editorial to the Special Issue “Advances in Molecular Genetics of Brain Tumors”
Next Article in Special Issue
Meta-Analysis and Systematic Review of HLA DQ2/DQ8 in Adults with Celiac Disease
Previous Article in Journal
Localized Infections with P. aeruginosa Strains Defective in Zinc Uptake Reveal That Zebrafish Embryos Recapitulate Nutritional Immunity Responses of Higher Eukaryotes
Previous Article in Special Issue
Toll-like Receptors and Celiac Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Developments in Celiac Disease Treatment

by
Mariana Verdelho Machado
1,2
1
Gastroenterology Department, Hospital de Vila Franca de Xira, Estrada Carlos Lima Costa, Nª 2, 2600-009 Vila Franca de Xira, Portugal
2
Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
Int. J. Mol. Sci. 2023, 24(2), 945; https://doi.org/10.3390/ijms24020945
Submission received: 30 November 2022 / Revised: 18 December 2022 / Accepted: 1 January 2023 / Published: 4 January 2023
(This article belongs to the Special Issue Celiac Disease: Genetics, Pathogenesis and Therapy)

Abstract

:
Celiac disease (CD) is a common autoimmune disease affecting around 1% of the population. It consists of an immune-mediated enteropathy, triggered by gluten exposure in susceptible patients. All patients with CD, irrespective of the presence of symptoms, must endure a lifelong gluten-free diet (GFD). This is not an easy task due to a lack of awareness of the gluten content in foods and the extensive incorporation of gluten in processed foods. Furthermore, a GFD imposes a sense of limitation and might be associated with decreased quality of life in CD patients. This results in gluten contamination in the diet of four out of five celiac patients adhering to a GFD. Furthermore, one in three adult patients will report persistent symptoms and two in three will not achieve full histological recovery when on a GFD. In recent years, there has been extensive research conducted in the quest to find the holy grail of pharmacological treatment for CD. This review will present a concise description of the current rationale and main clinical trials related to CD drug therapy.

1. Introduction

Celiac disease (CD) is an autoimmune disease in which, in susceptible subjects, the ingestion of gluten triggers an immune attack on the small bowel, as well as a serological response [1]. Unlike other autoimmune diseases, the immunogenic antigens that trigger the immune response in CD have already been identified and highly characterized [2]. As such, removing those antigens by enduring a gluten-free diet (GFD) is a known effective treatment for CD.
The prevalence of CD is estimated to be 1% of the population [3]. Considering a global population of eight billion people, there are roughly eighty million patients with CD. The classical presentation of CD is malabsorption manifestations such as diarrhea, weight loss, and nutrition deficits, although most patients will remain asymptomatic or with non-specific and extraintestinal symptoms [4]. Even though not consistent among all cohorts [5], patients with CD seem to present increased mortality [6,7], which might be mitigated by achieving mucosal healing through a GFD [8,9].
In the last decade, intense efforts have been applied in the search for pharmacological treatments for CD. The bar is high as dietary treatment is already available, which is effective and has no predictable adverse effects. However, adhering to a strict GFD is challenging, resulting in self-reported adherence rates ranging from 42% to 91% [10]. Furthermore, up to 80% of GFD-adherent patients might have inadvertent gluten contamination in their diet [11]. Lastly, mucosal healing after a GFD seems to occur in less than half of adults with CD [12]. Up to 0.5% of patients with CD will progress to refractory celiac disease (RCD) [13], which when associated with aberrant monoclonal T cell infiltration (RCD type 2) presents a high risk of progression to enteropathy-associated T cell lymphoma (EATL) [14].
Drug discovery for treating CD, as a complement to a GFD, aims to intervene in different scenarios, including (a) maintenance therapy, (b) rescue therapy after acute gluten exposure, and (c) the mitigation of inadvertent chronic gluten exposure [15].
This review will summarize the rationale that has been applied to CD drug development, as well as the clinical advances in its research.

2. Gluten-Free Diet: The Current Standard of Care

Sensum strictum, gluten refers to proteins present in wheat that contain two major protein components: monomeric water-soluble gliadins and multimeric water-insoluble glutenins. The proteins secalin, in rye, and hordein, in barley, share similar immunogenic properties and have also been called gluten in sensum latum [16]. Oats are phylogenetically more distant and are usually tolerated by CD patients [17].
Patients with CD independent of the presence of symptoms, symptomatic potential CD (that is patients with positive anti-transglutaminase antibody and normal duodenal histology), dermatitis herpetiformis, and gluten ataxia should follow a lifelong GFD, since a GFD is currently the only proven treatment for CD [18]. Regarding patients with asymptomatic potential CD, only a minority will develop villous atrophy and should be followed rather than be recommended a GFD [19].
CD is associated with a panoply of extraintestinal manifestations and autoimmune diseases, such as neuropsychiatric [20,21,22] and dermatological manifestations [23], unexplained abnormal liver enzymes [24], type 1 diabetes mellitus, and autoimmune thyroid disorders [25]. Adherence to a GFD may improve most of those manifestations. Regarding neuropsychiatric manifestations, it is known to improve headache [26] and gluten ataxia as long as there is no irreversible loss of Purkinje cells in the cerebellum [27]. A GFD may also improve response to drug-resistant epilepsy [28], gluten-induced cognitive impairment [29], psychiatric disorders (particularly anxiety) [26], and fatigue in up to 50% the patients [30], while response to peripheral neuropathy is variable with lower response rates in patients presenting anti-neuronal antibodies [31]. Dermatitis herpetiformis typically responds to a GFD [32]. Psoriasis [33], chronic urticaria [34], and recurrent aphthous stomatitis may also improve in patients with CD, while dental enamel defects are irreversible [35]. Furthermore, a GFD may improve metabolic control in CD patients with type 1 diabetes mellitus [33], bone density [36], menstrual disturbances and fertility [37], and unexplained transaminitis [38].
A GFD will only be effective if the patient complies. In most cases, less than 50% of CD patients achieve long-term adherence to a GFD [10]. Indeed, dietary regimens are the least appealing medical strategies, being one of the treatment regimens with the lowest rate of adherence (as opposed to medications that have the highest adherence rate) [39]. A GFD is particularly difficult to follow due to the omnipresence of gluten in processed foods, even in unlikely foods (such as yogurt and frozen fish) and products (such as toothpaste and lipstick). Gluten is the Latin word for “glue”, and its viscoelastic properties make it highly appealing to the food industry. The removal of gluten changes the physical properties and durability of foods. To compensate for gluten removal, the food industry tends to deliver unhealthily high fat and sugar content [40] and decrease healthy supplementation, such as the addition of fiber, iron, folate, and zinc [15]. Gluten-free products are more expensive than their gluten-containing counterparts, even though the recent popularity, and hence widespread availability, of gluten-free products has translated into a decrease in price [41]. Dining out may be challenging as it is difficult to control for gluten contamination, which may subsequently lead to social isolation, anxiety, and impaired quality of life [42].
Another barrier to a GFD is inadvertent gluten contamination. Even lifelong GFD-adherent patients struggle to correctly ascertain gluten content from commercial product labels [43]. Indeed, 70–80% of patients adherent to a GFD present gluten contamination in their diet [11,12,44], with an average gluten exposure of around 150 mg/day [45], which is much higher than the considered safe amount of up to 10 mg/day [46] and higher than the 50 mg/day cutoff (equivalent to 1% of a slice of bread) that is known to be able to induce mucosal atrophy [47]. Of note, the gluten content of a typical Western diet is around 15–20 g/day [48].
Finally, up to 30% of patients report persistent symptoms [49], and one to two-thirds of adults will not achieve histological recovery after one year on a strict GFD [50]. Of note, monitoring of anti-actin IgA antibodies may help to predict GFD-induced resolution of intestinal mucosa damage [51].

3. Pathophysiology-Driven Strategies to Treat Celiac Disease

3.1. Non-Immunogenic Gluten Delivery

Gluten proteins are rich in proline (15% of amino acid composition) and glutamine (35% of amino acid composition), which confers high resistance to the action of human proteases in the intestinal lumen [16] and results in the production of peptides up to 30–40 amino acids in length that are highly immunogenic [52]. One strategy for the treatment of CD, besides removing gluten from the diet, would be to present non-immunogenic variants of gluten. Different natural wheat variants present different T cell immunogenicity [53]. However, all variants, even ancient ones, seem to present toxic potential [54,55]. Another strategy would be to genetically engineer non-immunogenic wheat. This is not an easy task as around 100 genes encode gluten, and hence silencing one gene would not be enough. Additionally, silencing genes responsible for gluten immunogenicity might hamper its viscoelastic properties [56]. One example is the E82 wheat line produced by RNAi technology that blocks relevant gliadin genes [57]. A pilot study with 21 CD patients eating E82 wheat did show decreased interferon-γ (INF-γ) production in peripheral blood mononuclear cells and a very low level of gluten immunogenic peptides (GIP) in stool samples from those patients, suggesting low exposure to immunogenic epitopes [58].

3.2. Blocking Immunogenic Gluten Exposure

In order to induce an immune response, gluten needs to overcome the intestinal epithelial barrier and reach the lamina propria, where it will be presented to the immune system by antigen-presenting cells (APCs). Three strategies have been proposed to neutralize gluten after dietary exposure: (a) digesting gluten through the delivery of exogenous peptidases, (b) sequestering gluten in the intestinal lumen, and (c) decreasing epithelial permeability. These strategies intend to mitigate immune responses to chronic inadvertent low levels of gluten exposure as an adjunctive to a GFD (Table 1).
Regarding the first strategy, human proteases are ineffective at degrading proline- and glutamine-rich gluten proteins. As such, gluten degradation could be achieved by the administration of exogenous endopeptidases that would digest, in the stomach, the gluten proteins into small non-immunogenic peptides before they reach the duodenum. Those endopeptidases must fulfill several requisites: (a) be able to degrade all different gluten immunogenic sequences, (b) be stable and active in the low pH gastric environment and escape gastric protease degradation, and (c) not induce adverse effects in the patient [71]. Several microorganisms express prolyl endopeptidases that have been shown in vitro and in vivo to be able to degrade gluten proteins [72], such as Aspergillus niger, Flavobacterium meningosepticum, Myxococcus xanthus, and Sphingomonas capsulate [71]. Similarly, glutamine-specific endoproteases can have a complementary action on gluten degradation. One such example is EP-B2, a cysteine endoprotease expressed in germinating barley (Hordeum vulgare) seeds [73].
The most studied endoprotease drug for CD is latiglutenase, formally dubbed ALV003. Latiglutenase is an orally administered mixture of two gluten proteases: ALV001 (a modified recombinant version of glutamine endopeptidase EP-B2) and ALV002 (a modified recombinant version of prolyl endopeptidase from S. capsulata). These peptidases have complementary peptidase activity in terms of substrate sequence and length [72]. In 2010, a placebo-controlled pilot study of oral latiglutenase administered with large gluten meals (16 g/day) over 3 days in 20 CD patients abrogated gluten immune responses, with decreased INF-γ secretion by gluten-specific T cells in peripheral blood from those patients [59]. A phase 2a dose-ranging trial administered different doses of latiglutenase (from 100mg to 900mg/day, for 12 or 24 weeks) to 494 CD patients presenting persistent moderate to severe symptoms despite being on a strict GFD for at least one year. Globally, latiglutenase, compared to placebo, did not improve symptoms or histology, i.e., intraepithelial lymphocytosis (IEL) or villous height to crypt depth ratio (Vh:Cd) [61]. However, in the subgroup of seropositive patients, the highest dose of latiglutenase (900 mg) was associated with an improvement in abdominal pain and bloating when compared to placebo [62], suggesting a benefit for patients with gluten contamination in their diet. Subsequently, two phase 2 randomized controlled trials (RCT) in 41 and 43 patients with CD subjected to a 2 g/day gluten challenge for 6 weeks showed that the administration of high doses of latiglutenase (at least 900 mg) was able to prevent mucosal deterioration (abrogated Vh:Cd decrease and IEL infiltration) and improve symptoms (at 1200 mg/day) [60,63]. An ongoing phase 2 RCT (NCT 04243551) in patients with CD subjected to periodic gluten exposure for 6 months is estimated to be completed in May 2023.
Another prolyl endopeptidase that is derived from Aspergillus niger, AN-PEP, also showed in vitro gluten-degrading activity [74,75]. Two pilot studies involving 12 healthy subjects and 19 gluten-sensitive patients showed AN-PEP to be capable of achieving significant gastric degradation of gluten [76,77]. A short phase 2 RCT involving 12 CD patients subjected to a 7 g/day gluten challenge failed to demonstrate symptomatic advantages over placebo. However, it should be noted that even the group on placebo did not have worsened symptoms due the gluten challenge [64]. An ongoing phase 4 RCT (NCT 04788797) in patients with CD on a strict GFD was estimated to be completed in December 2022.
More recently, TAK-62, a glutenase effective in vitro [78], was shown to be well tolerated and capable of degrading 97% of gluten in gastric aspirates from CD patients after a gluten challenge [65].
The second strategy is to sequester and neutralize gluten proteins in the intestinal lumen, preventing its digestion in immunogenic gluten peptides. Two main therapies have been studied: AGY, an oral egg yolk anti-gliadin polyclonal antibody [66], and BL-7010, a non-absorbable high molecular weight copolymer of hydroxyethyl methacrylate and styrene sulfonate—P(HEMA-co-SS) [79].
AGY was shown to neutralize gluten proteins in mouse models [80] and decrease symptoms, serology, and intestinal permeability (assessed with lactulose:mannitol excretion ratio, LMER) in a phase 1 open-label single-arm trial involving 10 CD patients on a GFD [66]. An ongoing phase 2 RCT on AGY in symptomatic CD patients on a GFD (NCT 03707730) was estimated to be completed in December 2022.
BL-7010 was shown to bind with high affinity to gliadin in vitro [79], to abrogate gluten-induced intestinal injury in vivo in rodent models [79,81], and to decrease tumor necrosis factor-α (TNF-α) secretion ex vivo in mucosal biopsies from CD patients in the presence of partially digested gliadin [81]. A phase 1 RCT in CD patients was completed in 2014, however no data have been published so far (NCT 01990885).
Lastly, intestinal permeability to gluten has been addressed through modulation of tight junctions. Larazotide acetate, formerly dubbed AT1001, is a synthetic octapeptide structurally related to the zonula occludens toxin (ZOT) produced by the bacterium Vibrio cholera [82]. Larazotide improves intestinal barrier function by acting as an inhibitor of zonulin by blocking its receptor [83,84]. Gluten induces zonulin secretion after binding to receptor CXCR3 in enterocytes. Zonulin will then bind to the receptor complex epidermal growth factor receptor (EGFR) and protease-activated receptor-2 (PAR2) in enterocytes, initiating a signaling pathway that is Myd88-dependent and which leads to the polymerization of actin microfilaments and subsequent tight junction disassembly, hence increasing intestinal permeability [85]. A phase 1 double-blind RCT compared larazotide at a dosage of 12mg to placebo administered over 3 days in 21 patients with CD who were subjected to one day of a 2.5 g gluten challenge. Larazotide was associated with fewer symptoms and INF-γ immune response, even though it failed to demonstrate improvements in intestinal permeability (assessed by LMER) [67]. Subsequently, three short-term (14 days [84], 6 weeks [69], and 12 weeks [70]) phase 2 RCTs and one meta-analysis [82] enrolling 626 CD patients, treated with larazotide doses that ranged from 1mg to 8mg three times per day, showed improvements in symptoms after a gluten challenge, but not in patients on a strict GFD. While no data on histology were described, those trials did not demonstrate an improvement in terms of LMER, which might be explained by LMER baseline variability [68]. Interestingly, the authors found an inverse dose–response relationship (lower doses presenting better results), which might be explained by an increase in larazotide fragmentation at higher doses (with larazotide fragments being competitively less effective) or by peptide aggregation compromising its function at higher doses [70]. A phase 3 RCT among symptomatic CD patients on a GFD (NCT 03569007) named CeDLara (Celiac Disease Larazotide) planned to enroll 525 patients. However, an interim analysis of half of the initial target enrollment showed disappointing results, leading to discontinuation of the study in June 2022. Strategies aiming to decrease intestinal permeability are hindered by transcellular routes for gluten which allow it to trespass from the lumen into the lamina propria [1].

3.3. Transglutaminase Inhibition

Once gluten reaches the lamina propria, it undergoes deamidation by the enzyme tissue transglutaminase-2 (TG-2), which converts glutamine residues in glutamic acid [16]. This is a critical step for increasing gluten immunogenicity by increasing the stability of gluten-HLA DQ2/8 complex [86]. TG-2 also induces degradation of the anti-inflammatory PPAR-γ and promotes transcellular intestinal permeability to gluten [87]. This enzyme is also the autoantigen for classical serology in CD diagnosis [1]. One strategy that aims to decrease gluten immunogenicity is the inhibition of TG-2 activity.
Inhibition of TG-2 has been shown to abrogate gluten-induced immune activation in vitro [88,89] and ex vivo in intestinal biopsies from CD patients [89], and has also been shown to decrease enteropathy in animal models [90]. Of note, mice deficient in TG-2 develop systemic autoimmunity with splenomegaly and glomerulonephritis due to impaired clearance of apoptotic cells during thymus involution, which should prompt further investigation of the possible side effects of TG-2 inhibition [91]. Furthermore, TG has a role in extracellular matrix remodeling and the repair of mucosal damage, which must be taken into account in therapeutic strategies based on TG inhibition [92].
ZED1227 is an oral first-in-class selective inhibitor of TG-2. Phase 1 clinical trials among 100 healthy female and male volunteers (EudraCT 2014-003044-13 and 2015-005283-42), treated with up to 500 mg of ZED1227, proved it to be safe and well tolerated [93]. Recently, a phase 2 proof-of-concept trial tested increasing doses of ZED1227 (10 mg, 50 mg, or 100 mg) for 6 weeks and compared them to placebo in 160 CD patients undergoing a gluten challenge (3 g/day) [94]. ZED1227 abrogated intestinal mucosa injury (blunting the gluten-induced decrease in Vh:Cd and increase in IEL) and improved symptoms and quality of life. It was well tolerated, with the development of a cutaneous rash in only 8% of patients on 100 mg of ZED1227.

3.4. Immune Modulation

At the lamina propria, immunogenic deamidated gluten peptides will be exhibited at the surface of APCs, bound to major histocompatibility complex (MHC) class II proteins in a lock–key fashion. The subtype of human leukocyte antigen (HLA)-DQ2/DQ8 is the lock for that key, and carrying those haplotypes is a necessary albeit not sufficient condition for the development of CD [95]. Those APCs will present gluten epitopes to CD4+ T cells, triggering a Th1/Th17 phenotype with the production of proinflammatory cytokines such as TNF-α, INF-γ, interleukin (IL)-18, and IL-21, as well as a B cell response [96].
Strategies to block HLA-DQ2/DQ8 are being studied but remain in the preclinical phase. One such strategy is to deliver competitive inhibitors with gluten peptide analogues that present higher binding affinity than gluten while also not being recognized by T cells. Those inhibitors showed mild blunting of T cell activation in silico [97] and in vitro [98,99]. The quest for such therapy has been challenging, with issues regarding rapid degradation of the peptide ligand and possible interference with other vital MHC immune surveillance functions [100,101].
Another technique for immune modulation is the inhibition of lymphocyte trafficking and homing to the small bowel. This can be achieved by targeting the adhesion molecules on gut endothelial cells (mucosal addressin cellular adhesion molecule-1, MadCAM-1) or their counterpart integrin receptors in lymphocytes (integrin receptor α4β7), as well as by targeting tissue-specific chemokine receptors on lymphocytes (chemokine receptor-9, CCR9) (Table 2).
PTG-100 is an orally administered, potent, and selective α4β7 peptide antagonist. It was already tested in a phase 2a RCT in patients with ulcerative colitis and showed high gastrointestinal exposure, limited systemic exposure, and a dose–response improvement in endoscopy and histology [104]. A phase 1b study (NCT 04524221) evaluating PTG-100 at a dose of 600 mg twice a day versus placebo in 30 CD patients on a gluten challenge was completed in April 2022. Vedolizumab is an anti-α4β7 antibody that is widely used in inflammatory bowel disease. A large epidemiological study found that patients with inflammatory bowel disease treated with steroids, 5-aminosalicylates, and immunomodulators (but not vedolizumab) had a lower risk of developing CD compared to untreated patients, which may suggest a lack of efficacy when using vedolizumab in the treatment of CD. A phase 2 RCT evaluating vedolizumab in CD started in 2016 but was terminated in 2018 due to lack of enrollment (NCT02929316).
Vercirnon is an oral selective antagonist of CCR9 that has shown promising results in early studies of its use in the treatment of Crohn’s disease [105], even though subsequently a phase 3 RCT failed to show efficacy when used for induction therapy [106]. A phase 2 study of vercirnon in CD was completed in 2008 (NCT00540657), but its results were not published.
IL-15 seems to be a pivotal cytokine in the pathogenesis of CD [90]. IL-15 is produced by APCs and epithelial cells and induces activation and proliferation of IEL, thus promoting villous atrophy [102]. The IL-15 receptor consists of three chains: (a) a common cytokine receptor γ-chain that is shared with the receptors for IL-2, IL-4, IL-7, IL-9, and IL-21; a β-chain (IL-15Rβ) that is shared with the IL-2 receptor; and an IL-15 specific α-chain (IL-15Rα) [71]. The IL-15 receptor signals through the Janus kinase–signal transducer and activator of transcription (JAK/STAT) [107].
PRN-015, previously dubbed AMG714, was the first anti-IL-15 evaluated in CD in a proof-of-concept phase 2a RCT [102]. PRN-015 is a fully human IgG1k monoclonal antibody that binds to IL-15. This study evaluated 64 CD patients on a 2–4 g daily gluten challenge who were subcutaneously administered the antibody every other week for 10 weeks. According to the study, the antibody was well tolerated. Compared to placebo, PRN-015 treatment was associated with a decrease in IEL and a symptomatic benefit (less diarrhea development), but failed to show a benefit in terms of serology or Vh:Cd [102]. PRN-015 was also evaluated in 28 patients with type II RCD in a phase 2a RCT, and while it was associated with improvement in diarrhea, it showed no histological benefit over placebo [103]. Importantly, in those patients, PRN-015 was associated with significant adverse events, such as tuberculosis and cerebellar syndrome. An ongoing phase 2b RCT (NCT 04424927) in patients with CD who are non-responsive to a GFD is estimated to be completed in December 2023.
Regarding targeting the IL-15 receptor, a phase 1 RCT of a humanized monoclonal antibody against IL-15Rβ (Hu-Myk-β1) was completed among five patients with RCD in December 2019, but its results have not been published (NCT 01893775).
Tofacitinib is an oral small molecule pan-JAK inhibitor. In preclinical studies with transgenic mice that overexpressed IL-15 and thus developed the pathologic features of CD, tofacitinib was able to revert enteropathy [108]. Case reports also showed histological improvement in a CD patient on a gluten-containing diet [109], as well as in a patient with type II RCD [110]. A phase 2 open-label trial of tofacitinib in type II RCD is ongoing (Eudra CT: 2018-001678-10). (Table 2).
Infliximab, an anti-TNF-α antibody and a cornerstone in the treatment of inflammatory bowel disease, showed encouraging results in case reports of steroid-unresponsive RCD patients [111,112,113,114].
Steroids, particularly budesonide, may have a role in the treatment of non-responsive patients or those with RCD despite the frequent relapse after tapering suggested by case reports and case series [115,116,117]. In newly diagnosed CD however, a pilot study among 27 patients failed to show any benefit from budesonide as an adjunctive treatment to a GFD when attempting to accelerate intestinal mucosal recovery [118].

3.5. Inducing Immune Tolerance

CD is characterized by a loss of immune tolerance, with T regulatory cells (Treg) unable to suppress effector T cells [119,120,121]. Four strategies have been studied to re-establish immune gluten tolerance in patients with CD: (a) desensitization through the presentation of gliadin proteins in nanoparticles [122] or (b) red-blood cell moieties [123], (c) therapeutic vaccination [124], or (d) infestation with helminths [125]. These approaches have the advantage of avoiding impairments to systemic immune function [15] (Table 3).
TAK-101, previously dubbed TIMP-GLIA, consists of gliadin encapsulated in negatively charged poly(dl-lactide-co-glycolic acid) nanoparticles. After intravenous administration, TAK-101 is uptaken by APCs in the liver and spleen, which modulates their transcription towards an anti-inflammatory phenotype with (a) downregulation of costimulatory molecules CD80 and CD86, (b) induction of inhibitory PD-L1, and (c) enhanced production of the regulatory cytokines IL-10 and tumor growth factor-β (TGF-β) [122]. In mouse models of CD, TAK-101 induced effector CD4+ T cell anergy and CD4+ Treg activation, as well as, gliadin-specific unresponsiveness, thus preventing gluten-associated enteropathy [122]. In humans, TAK-101 also downregulates the expression of gut-homing (α4β7+ CD4+) and gut-retaining (αEβ7+ CD8+) integrins in circulating T cells [126]. Recently, a phase 2 study evaluating TAK-101 in 33 HLA-DQ2/8 CD patients subjected to a gluten challenge was conducted as a proof-of-concept, consequently showing the induction of antigen-specific immune tolerance in an autoimmune disease. Compared to placebo, infusions of TAK-101 on day 1 and day 8 blunted the growth of a population of gluten-specific INF-γ producing cells by 88%. It also abrogated the flattening of Vh:Cd [126]. An ongoing phase 2 dose-ranging trial (NCT 04530123) among 168 CD patients with a gluten-supplemented diet is estimated to be completed in January 2024.
KAN-101 is still in development and combines erythrocytes and gluten moieties. Red blood cells undergo early apoptosis. Dying erythrocytes combined with gluten are recognized by immune cells, which might induce gluten-specific tolerance [123]. A phase 1 study evaluating the safety and tolerability of KAN-101 is currently ongoing (NCT 04248855).
Nexvax-2 is a therapeutic vaccine that is an adjuvant-free mixture of three peptides (NPL001, NPL002, and NPL003) with immunodominant epitopes for gluten-specific CD4+ cells [124] and is used with the aim of rendering those cells unresponsive to further antigen exposure. Phase 1 studies have shown nexvax-2 to be safe and well tolerated after intradermal administration, even though it induced gastrointestinal symptoms such as diarrhea and nausea that are similar to the effects of gluten exposure [127]. However, a subsequent phase 1 RCT with increasing doses, ranging from 60 µg to 150 µg twice weekly for 8 weeks, could not prevent intestinal histological deterioration (assessed by Vh:Cd) in 108 CD patients exposed to a gluten challenge [124]. The intradermal administration may have hit the wrong target, being uptaken by APCs in the skin rather than the spleen and liver [126]. More recently, a phase 2 study with a similar design was terminated prematurely after an interim analysis determined futility [128].
Inoculation with helminths, namely Necator americanus, is a strategy that has also been studied in the treatment of inflammatory bowel disease [130]. In celiac disease, two small phase 1 single arm studies showed that N. americanus inoculation in CD patients exposed to progressively higher gluten challenges induced a shift in the adaptive T cell response to gluten towards a type 2 phenotype, while also leading to decreased INF-γ and IL-17 response to gluten exposure. Furthermore, compared to historical controls, infestation blunted serological and histological gluten-induced injury [131]. However, infestation failed to improve symptoms [125]. N. americanus was well tolerated, except for short-term pruritus at the inoculation site and self-limited abdominal pain [125].
More recently, a phase 2 RCT controlled with placebo that used a similar design among 54 CD patients was disappointing. Infestation did not protect from gluten-induced mucosal deterioration and was associated with a decrease in quality of life scores [129].

4. Conclusions

Currently, the only proven effective treatment for CD is a lifelong GFD. Drug development faces many challenges when there is already an established non-pharmacological therapy for a disease. For a new drug to be able to replace a GFD in diet-responsive CD patients, it must be devoid of potential adverse effects, must be simple to administer (preferably as an oral therapy), and must be inexpensive.
Pharmacological treatments for CD may be more useful, at least in the short term, for patients unresponsive to a GFD and may also be useful as an adjunctive treatment in association with a GFD, especially considering the high rate of gluten contamination in the diet due to either inadvertent consumption or non-compliance. To mitigate gluten contamination in the diet, two drugs currently present the most advanced clinical research: larazotide and latiglutenase. Larazotide is a drug that stabilizes enterocyte tight junctions with the aim of decreasing intestinal permeability. While RCTs could not demonstrate a decrease in intestinal permeability due to high variability in the assay, it did decrease symptoms and serology in phase 2 studies, suggesting an effective decrease in the amount of gluten the immune system is exposed to. However, in 2022, a phase 3 trial was suspended after an interim analysis showed no meaningful effect. Latiglutenase is a mix of glutenases that, in phase 2 studies, has been shown to prevent mucosal degradation and symptom development as a result of gluten contamination or challenge. Latiglutenase is a strong contender to become a standard adjunctive therapy in the treatment of CD.
For patients who are unresponsive to a GFD or those with RCD, one key focal point of research is the IL-15 pathway. Blocking IL-15 with a directed monoclonal antibody (PRN15) showed somewhat disappointing results, with the potential for meaningful adverse side effects. More promising seems to be tofacitinib, a pan-JAK inhibitor that acts on the IL-15 receptor signaling pathway. The use of monoclonal antibodies acting on different cytokines and lymphocyte trafficking are still only in embrionary phases of research.
An appealing strategy would be to induce immune tolerance to gluten, and hence avoid the systemic suppression of the immune system. However, two strategies were disappointing: therapeutic vaccines and hookworm infestation. Nevertheless, desensitization to antigen presentation is still in the running.
Lastly, it is still yet to be determined whether the drugs in the pipeline for the treatment of CD may have a role in the treatment of extraintestinal manifestations and conditions associated with CD, such as neuropsychiatric and autoimmune diseases.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

APCs: antigen-presenting cells; CCR-9, chemokine receptor-9; CD, celiac disease; EATL, enteropathy-associated T cell lymphoma; EGFR, epidermal growth factor receptor; GFD, gluten-free diet; GIP, gluten immunogenic peptides; IEL, intraepithelial lymphocytosis; IL, interleukin; INF-γ, interferon-γ; LMER, lactulose:mannitol excretion ratio; MadCAM-1, mucosal addressin cellular adhesion molecule-1; PAR2, protease-activated receptor-2; RCD, refractory celiac disease; RCT, randomized controlled trials; TG-2, transglutaminase-2; TGF-β, tumor growth factor-β, TNF-α, tumor necrosis factor-α; Treg, T regulatory cells; Vh:Cd, villous height to crypt depth ratio; ZOT, zonula occludens toxin.

References

  1. Calado, J.; Machado, M.V. Celiac disease revisited. GE Port. J. Gastroenterol. 2021, 29, 111–124. [Google Scholar] [CrossRef] [PubMed]
  2. Sollid, L.M.; Tye-Din, J.A.; Qiao, S.-W.; Anderson, R.P.; Gianfrani, C.; Koning, F. Update 2020: Nomenclature and listing of celiac disease–relevant gluten epitopes recognized by CD4+ T cells. Immunogenetics 2020, 72, 85–88. [Google Scholar] [CrossRef] [PubMed]
  3. Singh, P.; Arora, A.; Strand, T.A.; Leffler, D.A.; Catassi, C.; Green, P.H.; Kelly, C.P.; Ahuja, V.; Makharia, G.K. Global Prevalence of Celiac Disease: Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 823–836.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Leonard, M.M.; Sapone, A.; Catassi, C.; Fasano, A. Celiac disease and nonceliac gluten sensitivity: A review. JAMA 2017, 318, 647–656. [Google Scholar] [CrossRef]
  5. Koskinen, I.; Virta, L.J.; Huhtala, H.; Ilus, T.; Kaukinen, K.; Collin, P. Overall and Cause-Specific Mortality in Adult Celiac Disease and Dermatitis Herpetiformis Diagnosed in the 21st Century. Am. J. Gastroenterol. 2020, 115, 1117–1124. [Google Scholar] [CrossRef]
  6. Lebwohl, B.; Green, P.H.R.; Söderling, J.; Roelstraete, B.; Ludvigsson, J.F. Association Between Celiac Disease and Mortality Risk in a Swedish Population. JAMA 2020, 323, 1277–1285. [Google Scholar] [CrossRef]
  7. Quarpong, W.; Card, T.R.; West, J.; Solaymani-Dodaran, M.; Logan, R.F.; Grainge, M.J. Mortality in people with coeliac disease: Long-term follow-up from a Scottish cohort. United Eur. Gastroenterol. J. 2019, 7, 377–387. [Google Scholar] [CrossRef] [Green Version]
  8. Corrao, G.; Corazza, G.R.; Bagnardi, V.; Brusco, G.; Ciacci, C.; Cottone, M.; Guidetti, C.S.; Usai, P.; Cesari, P.; Pelli, M.A.; et al. Mortality in patients with coeliac disease and their relatives: A cohort study. Lancet 2001, 358, 356–361. [Google Scholar] [CrossRef]
  9. Rubio-Tapia, A.; Rahim, M.W.; See, J.A.; Lahr, B.D.; Wu, T.T.; Murray, J.A. Mucosal recovery and mortality in adults with celiac disease after treatment with a gluten-free die. Am. J. Gastroenterol. 2010, 105, 1412–1420. [Google Scholar] [CrossRef] [Green Version]
  10. Hall, N.; Rubin, G.; Charnock, A. Systematic review: Adherence to a gluten-free diet in adult patients with coeliac disease. Aliment. Pharmacol. Ther. 2009, 30, 315–330. [Google Scholar] [CrossRef]
  11. Hollon, J.R.; Cureton, P.A.; Martin, M.L.; Puppa, E.L.L.; Fasano, A. Trace gluten contamination may play a role in mucosal and clinical recovery in a subgroup of diet-adherent non-responsive celiac disease patien. BMC Gastroenterol. 2013, 13, 40. [Google Scholar] [CrossRef] [Green Version]
  12. Silvester, J.A.; Comino, I.; Kelly, C.P.; Sousa, C.; Duerksen, D.R.; Bernstein, C.N.; Cebolla, A.; Dominguez, M.R.; Graff, L.A.; Green, K.H.; et al. Most Patients with Celiac Disease on Gluten-Free Diets Consume Measurable Amounts of Gluten. Gastroenterology 2020, 158, 1497–1499.e1. [Google Scholar] [CrossRef] [PubMed]
  13. Rowinski, S.A.; Christensen, E. Epidemiologic and therapeutic aspects of refractory coeliac disease—A systematic review. Dan. Med. J. 2016, 63, A5307. [Google Scholar]
  14. Green, P.H.; Paski, S.; Ko, C.W.; Rubio-Tapia, A. AGA Clinical Practice Update on Management of Refractory Celiac Disease: Expert Review. Gastroenterology 2022, 163, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  15. Klonarakis, M.; Andrews, C.N.; Raman, M.; Panaccione, R.; Ma, C. Review article: Therapeutic targets for the pharmacologic management of coeliac disease—The future beyond a gluten-free diet. Aliment. Pharmacol. Ther. 2022, 55, 1277–1296. [Google Scholar] [CrossRef] [PubMed]
  16. Stamnaes, J.; Sollid, L. Celiac disease: Autoimmunity in response to food antigen. Semin. Immunol. 2015, 27, 343–352. [Google Scholar] [CrossRef] [PubMed]
  17. Kagnoff, M.F. Celiac disease: Pathogenesis of a model immunogenetic disease. J. Clin. Investig. 2007, 117, 41–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Raiteri, A.; Granito, A.; Giamperoli, A.; Catenaro, T.; Negrini, G.; Tovoli, F. Current guidelines for the management of celiac disease: A systematic review with comparative analysis. World J. Gastroenterol. 2022, 28, 154–176. [Google Scholar] [CrossRef]
  19. Volta, U.; Caio, G.; Giancola, F.; Rhoden, K.J.; Ruggeri, E.; Boschetti, E.; Stanghellini, V.; De Giorgio, R. Features and Progression of Potential Celiac Disease in Adults. Clin. Gastroenterol. Hepatol. 2016, 14, 686–693.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Granito, A.; Tovoli, F.; Raiteri, A.; Volta, U. Anti-ganglioside antibodies and celiac disease. Allergy Asthma Clin. Immunol. 2021, 17, 53. [Google Scholar] [CrossRef]
  21. Volta, U.; De Giorgio, R.; Granito, A.; Stanghellini, V.; Barbara, G.; Avoni, P.; Liguori, R.; Petrolini, N.; Fiorini, E.; Montagna, P.; et al. Anti-ganglioside antibodies in coeliac disease with neurological disorders. Dig. Liver Dis. 2006, 38, 183–187. [Google Scholar] [CrossRef] [PubMed]
  22. Cervio, E.; Volta, U.; Verri, M.; Boschi, F.; Pastoris, O.; Granito, A.; Barbara, G.; Parisi, C.; Felicani, C.; Tonini, M.; et al. Sera of Patients with Celiac Disease and Neurologic Disorders Evoke a Mitochondrial-Dependent Apoptosis In Vitro. Gastroenterology 2007, 133, 195–206. [Google Scholar] [CrossRef] [PubMed]
  23. Zauli, D.; Grassi, A.; Granito, A.; Foderaro, S.; De Franceschi, L.; Ballardini, G.; Bianchi, F.; Volta, U. Prevalence of silent coeliac disease in atopics. Dig. Liver Dis. 2000, 32, 775–779. [Google Scholar] [CrossRef]
  24. Volta, U.; Granito, A.; De Franceschi, L.; Petrolini, N.; Bianchi, F.B. Anti tissue transglutaminase antibodies as predictors of silent coeliac disease in patients with hypertransaminasaemia of unknown origin. Dig. Liver Dis. 2001, 33, 420–425. [Google Scholar] [CrossRef] [PubMed]
  25. Volta, U.; Ravaglia, G.; Granito, A.; Forti, P.; Maioli, F.; Petrolini, N.; Zoli, M.; Bianchi, F.B. Coeliac Disease in Patients with Autoimmune Thyroiditis. Digestion 2001, 64, 61–65. [Google Scholar] [CrossRef] [PubMed]
  26. Jericho, H.; Sansotta, N.; Guandalini, S. Extraintestinal Manifestations of Celiac Disease: Effectiveness of the Gluten-Free Diet. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 75–79. [Google Scholar] [CrossRef]
  27. Hadjivassiliou, M.; Davies-Jones, G.A.B.; Sanders, D.S.; Grünewald, R.A. Dietary treatment of gluten ataxia. J. Neurol. Neurosurg. Psychiatry 2003, 74, 1221–1224. [Google Scholar] [CrossRef] [PubMed]
  28. Swartwood, S.; Wilkes, J.; Bonkowsky, J.L.; Trandafir, C.C. Celiac Disease in Children: An Association with Drug-Resistant Epilepsy. Pediatr. Neurol. 2021, 120, 12–17. [Google Scholar] [CrossRef]
  29. Lichtwark, I.T.; Newnham, E.D.; Robinson, S.R.; Shepherd, S.J.; Hosking, P.; Gibson, P.R.; Yelland, G.W. Cognitive impairment in coeliac disease improves on a gluten-free diet and correlates with histological and serological indices of disease severity. Aliment. Pharmacol. Ther. 2014, 40, 160–170. [Google Scholar] [CrossRef]
  30. Siniscalchi, M.; Iovino, P.; Tortora, R.; Forestiero, S.; Somma, A.; Capuano, L.; Franzese, M.D.; Sabbatini, F.; Ciacci, C. Fatigue in adult coeliac disease. Aliment. Pharmacol. Ther. 2005, 22, 489–494. [Google Scholar] [CrossRef]
  31. Tursi, A.; Giorgetti, G.M.; Iani, C.; Arciprete, F.; Brandimarte, G.; Capria, A.; Fontana, L. Peripheral Neurological Disturbances, Autonomic Dysfunction, and Antineuronal Antibodies in Adult Celiac Disease before and after a Gluten-Free Diet. Dig. Dis. Sci. 2006, 51, 1869–1874. [Google Scholar] [CrossRef]
  32. Fry, L.; Riches, D.; Seah, P.; Hoffbrand, A. Clearance of skin lesions in dermatitis herpetiformis after gluten withdrawal. Lancet 1973, 301, 288–291. [Google Scholar] [CrossRef] [PubMed]
  33. Passali, M.; Josefsen, K.; Frederiksen, J.L.; Antvorskov, J.C. Current Evidence on the Efficacy of Gluten-Free Diets in Multiple Sclerosis, Psoriasis, Type 1 Diabetes and Autoimmune Thyroid Diseases. Nutrients 2020, 12, 2316. [Google Scholar] [CrossRef] [PubMed]
  34. Caminiti, L.; Passalacqua, G.; Magazzu, G.; Comisi, F.; Vita, D.; Barberio, G.; Sferlazzas, C.; Pajno, G.B. Chronic urticaria and associated coeliac disease in children: A case-control study. Pediatr. Allergy Immunol. 2005, 16, 428–432. [Google Scholar] [CrossRef] [PubMed]
  35. Pastore, L.; Carroccio, A.; Compilato, D.; Panzarella, V.; Serpico, R.; Muzio, L.L. Oral Manifestations of Celiac Disease. J. Clin. Gastroenterol. 2008, 42, 224–232. [Google Scholar] [CrossRef] [PubMed]
  36. Grace-Farfaglia, P. Bones of Contention: Bone Mineral Density Recovery in Celiac Disease—A Systematic Review. Nutrients 2015, 7, 3347–3369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Alecsandru, D.; López-Palacios, N.; Castaño, M.; Aparicio, P.; Garcia-Velasco, J.A.; Núñez, C. Exploring undiagnosed celiac disease in women with recurrent reproductive failure: The gluten-free diet could improve reproductive outcomes. Am. J. Reprod. Immunol. 2020, 83, e13209. [Google Scholar] [CrossRef] [PubMed]
  38. Castillo, N.E.; Vanga, R.R.; Theethira, T.G.; Rubio-Tapia, A.; Murray, J.A.; Villafuerte, J.; Bonder, A.; Mukherjee, R.; Hansen, J.; Dennis, M.; et al. Prevalence of Abnormal Liver Function Tests in Celiac Disease and the Effect of a Gluten-Free Diet in the US Population. Am. J. Gastroenterol. 2015, 110, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  39. DiMatteo, M.R. Variations in patients’ adherence to medical recommendations: A quantitative review of 50 years of research. Med. Care 2004, 42, 200–209. [Google Scholar] [CrossRef]
  40. Diez-Sampedro, A.; Olenick, M.; Maltseva, T.; Flowers, M. A Gluten-Free Diet, Not an Appropriate Choice without a Medical Diagnosis. J. Nutr. Metab. 2019, 2019, 2438934. [Google Scholar] [CrossRef]
  41. Lee, A.R.; Wolf, R.L.; Lebwohl, B.; Ciaccio, E.J.; Green, P.H. Persistent Economic Burden of the Gluten Free Diet. Nutrients 2019, 11, 399. [Google Scholar] [CrossRef]
  42. Shah, S.; Akbari, M.; Vanga, R.; Kelly, C.P.; Hansen, J.; Theethira, T.; Tariq, S.; Dennis, M.; Leffler, D.A. Patient Perception of Treatment Burden Is High in Celiac Disease Compared with Other Common Conditions. Am. J. Gastroenterol. 2014, 109, 1304–1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gutowski, E.D.; Weiten, D.; Green, K.H.; Rigaux, L.N.; Bernstein, C.N.; Graff, L.A.; Walker, J.R.; Duerksen, D.R.; Silvester, J.A. Can individuals with celiac disease identify gluten-free foods correctly? Clin. Nutr. ESPEN 2020, 36, 82–90. [Google Scholar] [CrossRef]
  44. Fernández-Bañares, F.; Beltrán, B.; Salas, A.; Comino, I.; Ballester-Clau, R.; Ferrer, C.; Molina-Infante, J.; Rosinach, M.; Modolell, I.; Rodríguez-Moranta, F.; et al. Persistent Villous Atrophy in De Novo Adult Patients with Celiac Disease and Strict Control of Gluten-Free Diet Adherence: A Multicenter Prospective Study (CADER Study). Am. J. Gastroenterol. 2021, 116, 1036–1043. [Google Scholar] [CrossRef] [PubMed]
  45. Syage, J.A.; Kelly, C.P.; Dickason, M.A.; Ramirez, A.C.; Leon, F.; Dominguez, R.; Sealey-Voyksner, J.A. Determination of gluten consumption in celiac disease patients on a gluten-free diet. Am. J. Clin. Nutr. 2018, 107, 201–207. [Google Scholar] [CrossRef] [Green Version]
  46. Akobeng, A.K.; Thomas, A.G. Systematic review: Tolerable amount of gluten for people with coeliac disease. Aliment. Pharmacol. Ther. 2008, 27, 1044–1052. [Google Scholar] [CrossRef]
  47. Catassi, C.; Fabiani, E.; Iacono, G.; D’Agate, C.; Francavilla, R.; Biagi, F.; Volta, U.; Accomando, S.; Picarelli, A.; De Vitis, I.; et al. A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. Am. J. Clin. Nutr. 2007, 85, 160–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. van Overbeek, F.M.; Uil-Dieterman, I.G.; Mol, I.W.; Köhler-Brands, L.; Heymans, H.S.; Mulder, C.J. The daily gluten intake in relatives of patients with coeliac disease compared with that of the general Dutch population. Eur. J. Gastroenterol. Hepatol. 1997, 9, 1097–1099. [Google Scholar] [CrossRef]
  49. Baggus, E.M.R.; Hadjivassiliou, M.; Cross, S.; Penny, H.; Urwin, H.; Watson, S.; Woodward, J.M.; Sanders, D.S. How to manage adult coeliac disease: Perspective from the NHS England Rare Diseases Collaborative Network for Non-Responsive and Refractory Coeliac Disease. Front. Gastroenterol. 2019, 11, 235–242. [Google Scholar] [CrossRef]
  50. Galli, G.; Esposito, G.; Lahner, E.; Pilozzi, E.; Corleto, V.D.; Di Giulio, E.; Spiriti, M.A.A.; Annibale, B. Histological recovery and gluten-free diet adherence: A prospective 1-year follow-up study of adult patients with coeliac disease. Aliment. Pharmacol. Ther. 2014, 40, 639–647. [Google Scholar] [CrossRef]
  51. Granito, A.; Muratori, P.; Cassani, F.; Pappas, G.; Agostinelli, D.; Veronesi, L.; Bortolotti, R.; Petrolini, N.; Bianchi, F.B.; Volta, U. Anti-actin IgA antibodies in severe coeliac disease. Clin. Exp. Immunol. 2004, 137, 386–392. [Google Scholar] [CrossRef]
  52. Bethune, M.T.; Khosla, C. Oral Enzyme Therapy for Celiac Sprue. Methods Enzymol. 2012, 502, 241–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Spaenij–Dekking, L.; Kooy–Winkelaar, Y.; van Veelen, P.; Drijfhout, J.W.; Jonker, H.; van Soest, L.; Smulders, M.J.; Bosch, D.; Gilissen, L.J.; Koning, F. Natural Variation in Toxicity of Wheat: Potential for Selection of Nontoxic Varieties for Celiac Disease Patients. Gastroenterology 2005, 129, 797–806. [Google Scholar] [CrossRef] [PubMed]
  54. Šuligoj, T.; Gregorini, A.; Colomba, M.; Ellis, H.J.; Ciclitira, P.J. Evaluation of the safety of ancient strains of wheat in coeliac disease reveals heterogeneous small intestinal T cell responses suggestive of coeliac toxicity. Clin. Nutr. 2013, 32, 1043–1049. [Google Scholar] [CrossRef] [PubMed]
  55. Colombo, F.; Di Lorenzo, C.; Biella, S.; Bani, C.; Restani, P. Ancient and Modern Cereals as Ingredients of the Gluten-Free Diet: Are They Safe Enough for Celiac Consumers? Foods 2021, 10, 906. [Google Scholar] [CrossRef]
  56. García-Molina, M.D.; Giménez, M.J.; Sánchez-León, S.; Barro, F. Gluten free wheat: Are we there? Nutrients 2019, 11, 487. [Google Scholar] [CrossRef] [Green Version]
  57. 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] [Green Version]
  58. Guzmán-López, M.H.; Sánchez-León, S.; Marín-Sanz, M.; Comino, I.; Segura, V.; Vaquero, L.; Rivero-Lezcano, O.M.; Pastor, J.; Sousa, C.; Vivas, S.; et al. Oral Consumption of Bread from an RNAi Wheat Line with Strongly Silenced Gliadins Elicits No Immunogenic Response in a Pilot Study with Celiac Disease Patients. Nutrients 2021, 13, 4548. [Google Scholar] [CrossRef]
  59. 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]
  60. 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] [Green Version]
  61. Murray, J.A.; Kelly, C.P.; Green, P.H.; 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] [PubMed]
  62. Syage, J.A.; Murray, J.A.; Green, P.H.R.; Khosla, C. Latiglutenase Improves Symptoms in Seropositive Celiac Disease Patients While on a Gluten-Free Diet. Dig. Dis. Sci. 2017, 62, 2428–2432. [Google Scholar] [CrossRef] [PubMed]
  63. 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; Epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
  64. Tack, G.J.; Van De Water, J.M.; Bruins, M.J.; Mc Kooy-Winkelaar, E.; Van Bergen, J.; Bonnet, P.; Vreugdenhil, A.C.; 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] [PubMed] [Green Version]
  65. 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]
  66. 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]
  67. 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]
  68. 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 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] [Green Version]
  69. 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]
  70. Leffler, D.A.; Kelly, C.P.; Green, P.H.; Fedorak, R.; 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] [Green Version]
  71. Kivelä, L.; Caminero, A.; Leffler, D.A.; Pinto-Sanchez, M.I.; Tye-Din, J.A.; Lindfors, K. Current and emerging therapies for coeliac disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 181–195. [Google Scholar] [CrossRef] [PubMed]
  72. Gass, J.; Bethune, M.T.; Siegel, M.; Spencer, A.; Khosla, C. Combination Enzyme Therapy for Gastric Digestion of Dietary Gluten in Patients with Celiac Sprue. Gastroenterology 2007, 133, 472–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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] [PubMed] [Green Version]
  74. Mitea, C.; Havenaar, R.; Drijfhout, J.W.; Edens, L.; Dekking, L.; Koning, F. Efficient degradation of gluten by a prolyl endoprotease in a gastrointestinal model: Implications for coeliac disease. Gut 2008, 57, 25–32. [Google Scholar] [CrossRef]
  75. Stepniak, D.; Spaenij-Dekking, L.; Mitea, C.; Moester, M.; De Ru, A.; Baak-Pablo, R.; van Veelen, P.; Edens, L.; Koning, F. Highly efficient gluten degradation with a newly identified prolyl endoprotease: Implications for celiac disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 291, G621–G629. [Google Scholar] [CrossRef]
  76. 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] [Green Version]
  77. König, J.; Holster, S.; Bruins, M.; 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] [Green Version]
  78. 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] [Green Version]
  79. 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. 2014BL-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] [Green Version]
  80. 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]
  81. Pinier, M.; Fuhrmann, G.; Galipeau, H.J.; Rivard, N.; Murray, J.A.; David, C.S.; Drasarova, H.; Tuckova, L.; Leroux, J.; Verdu, E.F. The Copolymer P(HEMA-co-SS) Binds Gluten and Reduces Immune Response in Gluten-Sensitized Mice and Human Tissues. Gastroenterology 2012, 142, 316–325.e12. [Google Scholar] [CrossRef] [PubMed]
  82. 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]
  83. Khaleghi, S.; Ju, J.M.; Lamba, A.; Murray, J.A. The potential utility of tight junction regulation in celiac disease: Focus on larazotide acetate. Ther. Adv. Gastroenterol. 2016, 9, 37–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Gopalakrishnan, S.; Durai, M.; Kitchens, K.; Tamiz, A.P.; Somerville, R.; Ginski, M.; Paterson, B.M.; Murray, J.A.; Verdu, E.F.; Alkan, S.S.; et al. Larazotide acetate regulates epithelial tight junctions in vitro and in vivo. Peptides 2012, 35, 86–94. [Google Scholar] [CrossRef] [PubMed]
  85. Fasano, A. Zonulin and Its Regulation of Intestinal Barrier Function: The Biological Door to Inflammation, Autoimmunity, and Cancer. Physiol. Rev. 2011, 91, 151–175. [Google Scholar] [CrossRef] [Green Version]
  86. Xia, J.; Sollid, L.M.; Khosla, C. Equilibrium and Kinetic Analysis of the Unusual Binding Behavior of a Highly Immunogenic Gluten Peptide to HLA-DQ2. Biochemistry 2005, 44, 4442–4449. [Google Scholar] [CrossRef] [PubMed]
  87. Elhence, A.; Ghoshal, U.C. A Trial of a Transglutaminase 2 Inhibitor in Celiac Disease. N. Engl. J. Med. 2021, 385, e57. [Google Scholar]
  88. Lebreton, C.; Ménard, S.; Abed, J.; Moura, I.C.; Coppo, R.; Dugave, C.; Monteiro, R.C.; Fricot, A.; Traore, M.G.; Griffin, M.; et al. Interactions among Secretory Immunoglobulin A, CD71, and Transglutaminase-2 Affect Permeability of Intestinal Epithelial Cells to Gliadin Peptides. Gastroenterology 2012, 143, 698–707.e4. [Google Scholar] [CrossRef] [PubMed]
  89. Maiuri, L.; Ciacci, C.; Ricciardelli, I.; Vacca, L.; Raia, V.; Rispo, A.; Griffin, M.; Issekutz, T.; Quaratino, S.; Londei, M. Unexpected Role of Surface Transglutaminase Type II in Celiac Disease. Gastroenterology 2005, 129, 1400–1413. [Google Scholar] [CrossRef]
  90. 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]
  91. Szondy, Z.; Sarang, Z.; Molnar, P.; Nemeth, T.; Piacentini, M.; Mastroberardino, P.G.; Falasca, L.; Aeschlimann, D.; Kovacs, J.; Kiss, I.; et al. Transglutaminase 2-/- mice reveal a phagocytosis-associated crosstalk between macrophages and apoptotic cells. Proc. Natl. Acad. Sci. USA 2003, 100, 7812–7817. [Google Scholar] [CrossRef] [Green Version]
  92. Paolella, G.; Sposito, S.; Romanelli, A.M.; Caputo, I. Type 2 Transglutaminase in Coeliac Disease: A Key Player in Pathogenesis, Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7513. [Google Scholar] [CrossRef] [PubMed]
  93. 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] [PubMed]
  94. Schuppan, D.; Mäki, M.; Lundin, K.E.; 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]
  95. Sollid, L.M. The roles of MHC class II genes and post-translational modification in celiac disease. Immunogenetics 2017, 69, 605–616. [Google Scholar] [CrossRef] [Green Version]
  96. Jabri, B.; Sollid, L.M. Tissue-mediated control of immunopathology in coeliac disease. Nat. Rev. Immunol. 2009, 9, 858–870. [Google Scholar] [CrossRef] [PubMed]
  97. Xia, J.; Bergseng, E.; Fleckenstein, B.; Siegel, M.; Kim, C.-Y.; Khosla, C.; Sollid, L.M. Cyclic and dimeric gluten peptide analogues inhibiting DQ2-mediated antigen presentation in celiac disease. Bioorg. Med. Chem. 2007, 15, 6565–6573. [Google Scholar] [CrossRef] [Green Version]
  98. 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]
  99. Huan, J.; Meza-Romero, R.; Mooney, J.L.; Vandenbark, A.A.; Offner, H.; Burrows, G.G. Single-chain recombinant HLA-DQ2.5/peptide molecules block α2-gliadin-specific pathogenic CD4+ T-cell proliferation and attenuate production of inflammatory cytokines: A potential therapy for celiac disease. Mucosal Immunol. 2011, 4, 112–120. [Google Scholar] [CrossRef] [Green Version]
  100. Ciccocioppo, R.; Di Sabatino, A.; Corazza, G.R. The immune recognition of gluten in coeliac disease. Clin. Exp. Immunol. 2005, 140, 408–416. [Google Scholar] [CrossRef]
  101. Haridy, J.; Lewis, D.; Newnham, E.D. Investigational drug therapies for coeliac disease—Where to from here? Expert Opin. Investig. Drugs 2018, 27, 225–233. [Google Scholar] [CrossRef] [PubMed]
  102. 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]
  103. Cellier, C.; Bouma, G.; van Gils, T.; Khater, S.; Malamut, G.; Crespo, L.; Collin, P.; Green, P.H.R.; Crowe, S.E.; Tsuji, W.; et al. Safety and efficacy of AMG 714 in patients with type 2 refractory coeliac disease: A phase 2a, randomised, double-blind, placebo-controlled, parallel-group study. Lancet Gastroenterol. Hepatol. 2019, 4, 960–970. [Google Scholar] [CrossRef] [PubMed]
  104. 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 α4β7 antagonist peptide: Preclinical development and phase 1 and 2a studies in ulcerative colitis. Gastroenterology 2022, 161, 1853–1864. [Google Scholar] [CrossRef] [PubMed]
  105. Keshav, S.; Vaňásek, T.; Niv, Y.; Petryka, R.; Howaldt, S.; Bafutto, M.; Rácz, I.; Hetzel, D.; Nielsen, O.H.; Vermeire, S.; et al. A Randomized Controlled Trial of the Efficacy and Safety of CCX282-B, an Orally-Administered Blocker of Chemokine Receptor CCR9, for Patients with Crohn’s Disease. PLoS ONE 2013, 8, e60094. [Google Scholar] [CrossRef] [PubMed]
  106. Feagan, B.G.; Sandborn, W.J.; D’Haens, G.; Lee, S.D.; Allez, M.; Fedorak, R.N.; Seidler, U.; Vermeire, S.; Lawrance, I.C.; Maroney, A.C.; et al. Randomised clinical trial: Vercirnon, an oral CCR9 antagonist, vs. placebo as induction therapy in active Crohn’s disease. Aliment. Pharmacol. Ther. 2015, 42, 1170–1181. [Google Scholar] [CrossRef] [Green Version]
  107. Levescot, A.; Malamut, G.; Cerf-Bensussan, N. Immunopathogenesis and environmental triggers in coeliac disease. Gut 2022, 71, 2337–2349. [Google Scholar] [CrossRef]
  108. 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] [Green Version]
  109. Wauters, L.; Vanuytsel, T.; Hiele, M. Celiac Disease Remission with Tofacitinib: A Case Report. Ann. Intern. Med. 2020, 173, 585. [Google Scholar] [CrossRef]
  110. Grewal, J.K.; Kassardjian, A.; Weiss, G.A. Successful novel use of tofacitinib for type II refractory coeliac disease. BMJ Case Rep. 2022, 15, e244692. [Google Scholar] [CrossRef]
  111. Gillett, H.R.; Arnott, I.D.; McIntyre, M.; Campbell, S.; Dahele, A.; Priest, M.; Jackson, R.; Ghosh, S. Successful infliximab treatment for steroid-refractory celiac disease: A case report. Gastroenterology 2002, 122, 800–805. [Google Scholar] [CrossRef] [PubMed]
  112. Turner, S.M.; Moorghen, M.; Probert, C.S. Refractory coeliac disease: Remission with infliximab and immunomodulators. Eur. J. Gastroenterol. Hepatol. 2005, 17, 667–669. [Google Scholar] [CrossRef] [PubMed]
  113. Costantino, G.; della Torre, A.; Presti, M.L.; Caruso, R.; Mazzon, E.; Fries, W. Treatment of life-threatening type I refractory coeliac disease with long-term infliximab. Dig. Liver Dis. 2008, 40, 74–77. [Google Scholar] [CrossRef] [PubMed]
  114. Rawal, N.; Twaddell, W.; Fasano, A.; Blanchard, S.; Safta, A. Remission of Refractory Celiac Disease with Infliximab in a Pediatric Patient. ACG Case Rep. J. 2015, 2, 121–123. [Google Scholar] [CrossRef]
  115. Brar, P.; Lee, S.; Lewis, S.; Egbuna, I.; Bhagat, G.; Green, P.H. Budesonide in the Treatment of Refractory Celiac Disease. Am. J. Gastroenterol. 2007, 102, 2265–2269. [Google Scholar] [CrossRef]
  116. Mukewar, S.S.; Sharma, A.; Rubio-Tapia, A.; Wu, T.-T.; Jabri, B.; Murray, J.A. Open-Capsule Budesonide for Refractory Celiac Disease. Am. J. Gastroenterol. 2017, 112, 959–967. [Google Scholar] [CrossRef]
  117. Therrien, A.; Silvester, J.A.; Leonard, M.M.; Leffler, D.A.; Fasano, A.; Kelly, C.P. Enteric-Release Budesonide May Be Useful in the Management of Non-Responsive Celiac Disease. Dig. Dis. Sci. 2020, 66, 1989–1997. [Google Scholar] [CrossRef]
  118. Newnham, E.D.; Clayton-Chubb, D.; Nagarethinam, M.; Hosking, P.; Gibson, P.R. Randomised clinical trial: Adjunctive induction therapy with oral effervescent budesonide in newly diagnosed coeliac disease. Aliment. Pharmacol. Ther. 2021, 54, 419–428. [Google Scholar] [CrossRef] [PubMed]
  119. Granzotto, M.; Bo, S.D.; Quaglia, S.; Tommasini, A.; Piscianz, E.; Valencic, E.; Ferrara, F.; Martelossi, S.; Ventura, A.; Not, T. Regulatory T-Cell Function Is Impaired in Celiac Disease. Dig. Dis. Sci. 2009, 54, 1513–1519. [Google Scholar] [CrossRef]
  120. Hmida, N.B.; Ben Ahmed, M.; Moussa, A.; Rejeb, M.B.; Said, Y.; Kourda, N.; Meresse, B.; Abdeladhim, M.; Louzir, H.; Cerf-Bensussan, N. Impaired control of effector T cells by regulatory T cells: A clue to loss of oral tolerance and autoimmunity in celiac disease? Am. J. Gastroenterol. 2012, 104, 604–611. [Google Scholar] [CrossRef] [Green Version]
  121. Cook, L.; Munier, C.M.L.; Seddiki, N.; van Bockel, D.; Ontiveros, N.; Hardy, M.; Gillies, J.K.; Levings, M.; Reid, H.H.; Petersen, J.; et al. Circulating gluten-specific FOXP3+CD39+ regulatory T cells have impaired suppressive function in patients with celiac disease. J. Allergy Clin. Immunol. 2017, 140, 1592–1603.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Freitag, T.L.; Podojil, J.R.; Pearson, R.M.; Fokta, F.J.; Sahl, C.; Messing, M.; Andersson, L.C.; Leskinen, K.; Saavalainen, P.; Hoover, L.I.; et al. Gliadin nanoparticles induce immune tolerance to gliadin in mouse models of celiac disease. Gastroenterology 2020, 158, 1667–1681.e12. [Google Scholar] [CrossRef] [PubMed]
  123. Grimm, A.J.; Kontos, S.; Diaceri, G.; Quaglia-Thermes, X.; Hubbell, J.A. Memory of tolerance and induction of regulatory T cells by erythrocyte-targeted antigens. Sci. Rep. 2015, 5, 15907. [Google Scholar] [CrossRef] [Green Version]
  124. 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]
  125. 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]
  126. 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] [PubMed]
  127. 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] [Green Version]
  128. 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]
  129. 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]
  130. Croese, J.; O’neil, J.; Masson, J.; Cooke, S.; Melrose, W.; Pritchard, D.; Speare, R. A proof of concept study establishing Necator americanus in Crohn’s patients and reservoir donors. Gut 2006, 55, 136–137. [Google Scholar] [CrossRef] [Green Version]
  131. Croese, J.; Giacomin, P.; Navarro, S.; Clouston, A.; McCann, L.; Dougall, A.; Ferreira, I.; Susianto, A.; O’Rourke, P.; Howlett, M.; et al. Experimental hookworm infection and gluten microchallenge promote tolerance in celiac disease. J. Allergy Clin. Immunol. 2015, 135, 508–516.e5. [Google Scholar] [CrossRef] [PubMed]
Table 1. Relevant clinical trials on agents that block immunogenic gluten exposure.
Table 1. Relevant clinical trials on agents that block immunogenic gluten exposure.
AgentStudyTrial PhasePopulationTreatment DurationMain Results (vs. Placebo)
Endopeptidases
LatiglutenaseTye-Din, 2010 [59]120 CD patients on a gluten challenge (16 g/day)800 mg/day vs. placebo3 days
  • ↓ INF-γ secretion by gluten-specific T cells in peripheral blood
Lahdeaho, 2014 [60]2a41 CD patients on a gluten challenge (2 g/day)900 mg/day vs. placebo6 weeks
  • Prevented mucosal deterioration (no ↓ Vh:Cd or ↑ IEL)
  • No improvement in symptoms
Murray, 2017 [61]; Syage, 2017 [62]2b494 CD patients with moderate or severe symptoms on a GFD ≥ 1 year100 mg, 300 mg, 450 mg, 600 mg, or 900 mg/day vs. placebo12 or 24 weeks
  • No ≠ in Vh:Cd or ↑ IEL
  • No ≠ in serology
  • Improvement in the symptoms of seropositive patients with ≥600 mg/day
Murray, 2022 [63]2b43 CD patients on a gluten challenge (2 g/day)1200 mg/day vs. placebo6 weeks
  • Prevented mucosal deterioration (lower ↓ Vh:Cd)
  • Tendency to decrease symptoms
NCT 042435512b120 symptomatic CD patients undergoing periodic gluten exposurevs. placebo6 weeksOngoing
Estimated completion May 2023
AN-PEPTack, 2013 [64]214 CD patients on a gluten challenge (7 g/day)Topping with either AN-PEP or placebo2 weeks
  • No ≠ in Vh:Cd or ↑ IEL
  • No ≠ in terms of quality of life
NCT 04788797414 CD patients on a daily gluten challenge 2 capsules/day vs. placebo8 weeksOngoing
Completion December 2022
TAK-062Pultz, 2021 [65]1CD in GFD and healthy subjects after a gluten meal (3–9 g)100–900 mg6 weeks
  • Well tolerated
Gluten sequestration
AGYSample, 2017 [66]110 CD patients on a GFD1000 mg bid4 weeks
  • ↓ symptoms
  • ↓ serology
  • ↓ LMER
NCT 037077302Symptomatic CD patients on a GFDBefore meals vs. placebo14 weeksOngoing
Completion December 2022
BL-7010NCT 01990885140 asymptomatic CD patientsDose-findingSingle doseCompleted in 2014
No data published so far
Tight junction modulation
Larazotide acetatePaterson, 2007 [67]121 CD patients on a 1-day gluten challenge (2.5 g)12 mg vs. placebo3 days
  • ↓ INF-γ secretion
  • ↓ symptoms
  • No ≠ in LMAR
Leffler, 2012 [68]2a86 CD patients ± gluten challenge (2.4 g/day)0.25 mg, 1 mg, 4 mg, or 8 mg/day vs. placebo14 days
  • ↓ symptoms
  • No ≠ in LMAR
Kelly, 2013 [69]2b177 CD patients on a gluten challenge (2.7 g/day)1 mg, 4 mg, or 8 mg/day vs. placebo6 weeks
  • ↓ symptoms
  • ↓ serology
  • No ≠ in LMER
Leffler, 2015 [70]2b342 symptomatic CD patients on a GFD0.5 mg, 1 mg, or 2 mg/day vs. placebo12 weeks
  • ↓ symptoms
NCT 035690073307 symptomatic CD patients on a GFD0.5 mg or 1 mg vs. placebo12 weeksPrematurely interrupted after interim analysis in June 2022
Table 2. Relevant clinical trials on agents that induce immune modulation.
Table 2. Relevant clinical trials on agents that induce immune modulation.
AgentStudyTrial PhasePopulationTreatment DurationMain Results (vs. Placebo)
Lymphocyte trafficking
PTG-100
(anti-α4β7)
NCT 045242211b30 CD patients on a gluten challenge 600 mg bid vs. placebo42 daysCompleted in April 2022
No data published so far
Vedolizumab
(anti-α4β7)
NCT 029293162CD patients on a gluten challenge 300 mg vs. placebo6 weeksTerminated in 2018 due to lack of enrollment
Vercinon
(anti-CCR9)
NCT 00540657230 CD patients on a gluten challenge250 mg bid vs. placebo13 weeksCompleted in 2008
No data published so far
IL-15 targeting
PRN-015 or AMG714 (anti-IL-15)Lähdeaho, 2019 [102]2a64 CD patients on a gluten challenge (2–4 g/day)150 mg, 300 mg/day vs. placebo12 weeks
  • ↓ symptoms (diarrhea)
  • ↓ IEL at 300 mg
  • No ≠ in serology or Vh:Cd
Cellier, 2019 [103]2aType II RCD8 mg/kg 2×/week vs. placebo12 weeks
  • ↓ symptoms (diarrhea)
  • No ≠ in IEL, aberrant IEL, or Vh:Cd
  • Adverse events: 26% vs. 11%
NCT 044249272b220 CD patients non-responsive to a GFD3 ≠ arms vs. placebo28 weeksOngoing
Completion December 2023
Hu-Mik-β1
(anti-IL15Rβ1)
NCT 0189377515 RCD patientsEvery 3 weeks9 weeksCompleted in December 2019
No data published so far
Tofecitinib
(pan-JAK inhibitor)
Eudra CT: 2018-001678-102Type II RCD
(open-label) patients
10 mg bid12 weeksOngoing
Table 3. Relevant clinical trials on agents that induce immune tolerance.
Table 3. Relevant clinical trials on agents that induce immune tolerance.
AgentStudyTrial PhasePopulationTreatment DurationMain Results (vs. Placebo)
Nanoparticles for gliadin presentation
TAK-101 (TIMP-GLIA)Kelly, 2021 [126]233 CD patients on a gluten challenge 8 mg day 1 and 8 vs. placebo5 weeks
  • ↓ growth of gluten-specific INF-γ producing cells
  • ↓ Vh:Cd flattening
NCT 045301232168 CD patients on a gluten challenge 1–4 mg day 1 and 8 vs. placebo20 weeksOngoing
Completion January 2024
Gluten erythrocyte moiety on red blood cells
KAN-101NCT 042488552b41 CD patientsDose-ranging vs. placebo4 weeksCompleted in December 2021
No data published so far
Therapeutic vaccine
Nexvax-2Daveson, 2017 [127]136 HLA-DQ2.5 CD patients on a GFD3–900 µg 2×/week ID vs. placebo6 weeks
  • Transient symptoms resembling a gluten challenge
  • Safe and well tolerated
Goel, 2017 [124]2108 HLA-DQ2.5 CD patients on a gluten challenge60–150 µg 2×/week ID vs. placebo8 weeks
  • Transient symptoms resembling a gluten challenge
  • No ≠↓ Vh:Cd
Truitt, 2019 [128]2146 HLA-DQ2.5 CD patients on a gluten challenge32 doses SC vs. placebo26 weeksTerminated 2019 for futility
Helminth infestation
Necator americanusDaveson, 2011
[125]
110 CD patients on a gluten challenge (16 g/day)vs. historical controls21 weeks
  • Short-term pruritus at the inoculation site
  • No improvements in symptoms or histology
Croese, 2020 [129]254 CD patients subjected to gluten intakevs. placebo42 weeks
  • No histology improvement
  • ↓ quality of life
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

Machado, M.V. New Developments in Celiac Disease Treatment. Int. J. Mol. Sci. 2023, 24, 945. https://doi.org/10.3390/ijms24020945

AMA Style

Machado MV. New Developments in Celiac Disease Treatment. International Journal of Molecular Sciences. 2023; 24(2):945. https://doi.org/10.3390/ijms24020945

Chicago/Turabian Style

Machado, Mariana Verdelho. 2023. "New Developments in Celiac Disease Treatment" International Journal of Molecular Sciences 24, no. 2: 945. https://doi.org/10.3390/ijms24020945

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