A randomized, double-blind study of larazotide acetate to prevent the activation of celiac disease during gluten challenge

Daniel A Leffler, C P Kelly, H Z Abdallah, A M Colatrella, L A Harris, F Leon, L A Arterburn, B M Paterson, Z H Lan, J A Murray, Daniel A Leffler, C P Kelly, H Z Abdallah, A M Colatrella, L A Harris, F Leon, L A Arterburn, B M Paterson, Z H Lan, J A Murray

Abstract

Objectives: In patients with celiac disease, enteropathy is caused by the entry of gluten peptides into the lamina propria of the intestine, in which their immunogenicity is potentiated by tissue transglutaminase (tTG) and T-helper type 1-mediated immune responses are triggered. Tight junction disassembly and paracellular permeability are believed to have an important role in the transport of gluten peptides to the lamina propria. Larazotide acetate is a tight-junction regulator peptide that, in vitro, prevents the opening of intestinal epithelial tight junctions. The aim of this study was to evaluate the efficacy and tolerability of larazotide acetate in protecting against gluten-induced intestinal permeability and gastrointestinal symptom severity in patients with celiac disease.

Methods: In this dose-ranging, placebo-controlled study, 86 patients with celiac disease controlled through diet were randomly assigned to larazotide acetate (0.25, 1, 4, or 8 mg) or placebo three times per day with or without gluten challenge (2.4 g/day) for 14 days. The primary efficacy outcome was the urinary lactulose/mannitol (LAMA) fractional excretion ratio. Secondary endpoints included gastrointestinal symptom severity, quality-of-life measures, and antibodies to tTG.

Results: LAMA measurements were highly variable in the outpatient setting. The increase in LAMA ratio associated with the gluten challenge was not statistically significantly greater than the increase in the gluten-free control. Among patients receiving the gluten challenge, the difference in the LAMA ratios for the larazotide acetate and placebo groups was not statistically significant. However, larazotide acetate appeared to limit gluten-induced worsening of gastrointestinal symptom severity as measured by the Gastrointestinal Symptom Rating Scale at some lower doses but not at the higher dose. Symptoms worsened significantly in the gluten challenge-placebo arm compared with the placebo-placebo arm, suggesting that 2.4 g of gluten per day is sufficient to induce reproducible gluten toxicity. Larazotide acetate was generally well tolerated. No serious adverse events were observed. The most common adverse events were headache and urinary tract infection.

Conclusions: LAMA variability in the outpatient setting precluded accurate assessment of the effect of larazotide acetate on intestinal permeability. However, some lower doses of larazotide acetate appeared to prevent the increase in gastrointestinal symptom severity induced by gluten challenge.

Figures

Figure 1
Figure 1
Schema of overall study design and participant allocation. (a) Study design and (b) disposition of patients. LAMA, lactulose/mannitol.
Figure 2
Figure 2
Change from baseline LAMA levels in the individual treatment groups. Mean change from baseline in the urinary lactulose-to-mannitol (LAMA) ratio. Values are the geometric mean fold-ratio on day 14 over baseline (day 0). Vertical bars represent 95% confidence intervals. Dashed horizontal line indicates level for gluten-free control. G, gluten; P, placebo.
Figure 3
Figure 3
Change from baseline in gastrointestinal symptoms in the individual treatment groups as measured by the GSRS and CeD-GSRS. Mean (95% confidence interval) changes from baseline to day 14 in the total (a) Gastrointestinal Symptom Rating Scale (GSRS) scores and (b) Celiac Disease GSRS (CeD-GSRS) scores. n=9–13 per group. P values comparing larazotide acetate/gluten-challenge (G) groups to the placebo (P) drug/gluten-challenge group were calculated using an analysis of covariance model, with treatment as a fixed effect and the corresponding baseline value as a covariate.
Figure 4
Figure 4
Change in gastrointestinal symptoms as measured by the GSRS and CeD-GSRS during and after gluten challenge in the gluten challenge/placebo drug cohort compared to the aggregated gluten-free control cohorts and the Larazotide-treated gluten challenge groups. Time course of mean change from baseline in the total (a) Gastrointestinal Symptom Rating Scale (GSRS) score and (b) Celiac Disease (CeD-GSRS) scores. The gluten control group includes patients who received placebo drug and the gluten challenge (n=13). Gluten-free control includes patients who received placebo drug or 8 mg larazotide acetate and gluten placebo (n=20). Patients who received larazotide acetate and the gluten challenge were also combined into one group (n=38). P values were calculated using an analysis of covariance model with treatment as a fixed effect and the corresponding baseline value as a covariate.

References

    1. Fasano A, Berti I, Gerarduzzi T, et al. Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. Arch Intern Med. 2003;163:286–292.
    1. Green PH, Cellier C. Celiac disease. N Engl J Med. 2007;357:1731–1743.
    1. Fasano A. European and North American populations should be screened for coeliac disease. Gut. 2003;52:168–169.
    1. Catassi C, Fabiani E, Iacono G, et al. A prospective, double-blind, placebo-controlled trial to establish a safe gluten threshold for patients with celiac disease. Am J Clin Nutr. 2007;85:160–166.
    1. Green PH. The many faces of celiac disease: clinical presentation of celiac disease in the adult population. Gastroenterology. 2005;128:S74–S78.
    1. Murray JA, Van Dyke C, Plevak MF, et al. Trends in the identification and clinical features of celiac disease in a North American community, 1950–2001. Clin Gastroenterol Hepatol. 2003;1:19–27.
    1. Farrell RJ, Kelly CP. Celiac sprue. N Engl J Med. 2002;346:180–188.
    1. Rubio-Tapia A, Murray JA. Celiac disease. Curr Opin Gastroenterol. 2010;26:116–122.
    1. Ludvigsson JF, Green PH. Clinical management of coeliac disease. J Int Med. 2011;269:560–571.
    1. Green PH, Fleischauer AT, Bhagat G, et al. Risk of malignancy in patients with celiac disease. Am J Med. 2003;115:191–195.
    1. Jabri B, Kasarda DD, Green PH. Innate and adaptive immunity: the yin and yang of celiac disease. Immunol Rev. 2005;206:219–231.
    1. Leffler DA, Edwards-George J, Dennis M, et al. Factors that influence adherence to a gluten-free diet in adults with celiac disease. Dig Dis Sci. 2008;53:1573–1581.
    1. Gibert A, Espadaler M, Angel Canela M, et al. Consumption of gluten-free products: should the threshold value for trace amounts of gluten be at 20, 100 or 200 p.p.m. Eur J Gastroenterol Hepatol. 2006;18:1187–1195.
    1. Mayer M, Greco L, Troncone R, et al. Compliance of adolescents with coeliac disease with a gluten free diet. Gut. 1991;32:881–885.
    1. Collin P, Thorell L, Kaukinen K, et al. The safe threshold for gluten contamination in gluten-free products. Can trace amounts be accepted in the treatment of coeliac disease. Aliment Pharmacol Ther. 2004;19:1277–1283.
    1. Midhagen G, Hallert C. High rate of gastrointestinal symptoms in celiac patients living on a gluten-free diet: controlled study. Am J Gastroenterol. 2003;98:2023–2026.
    1. Ciacci C, Cirillo M, Cavallaro R, et al. Long-term follow-up of celiac adults on gluten-free diet: prevalence and correlates of intestinal damage. Digestion. 2002;66:178–185.
    1. Lee SK, Lo W, Memeo L, et al. Duodenal histology in patients with celiac disease after treatment with a gluten-free diet. Gastrointest Endosc. 2003;57:187–191.
    1. Rubio-Tapia A, Rahim MW, See JA, et al. Mucosal recovery and mortality in adults with celiac disease after treatment with a gluten-free diet. Am J Gastroenterol. 2010;105:1412–1420.
    1. Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel therapies. Gastroenterology. 2009;137:1912–1933.
    1. NIH National Institutes of Health Consensus Development Conference Statement on Celiac Disease, June 28–30, 2004. Gastroenterology. 2005;128:S1–S9.
    1. Zimmer KP, Poremba C, Weber P, et al. Translocation of gliadin into HLA-DR antigen containing lysosomes in coeliac disease enterocytes. Gut. 1995;36:703–709.
    1. Matysiak-Budnik T, Candalh C, Dugave C, et al. Alterations of the intestinal transport and processing of gliadin peptides in celiac disease. Gastroenterology. 2003;125:696–707.
    1. Matysiak-Budnik T, Moura IC, Arcos-Fajardo M, et al. Secretory IgA mediates retrotranscytosis of intact gliadin peptides via the transferrin receptor in celiac disease. J Exp Med. 2008;205:143–154.
    1. Schumann M, Richter JF, Wedell I, et al. Mechanisms of epithelial translocation of the alpha(2)-gliadin-33mer in coeliac sprue. Gut. 2008;57:747–754.
    1. Koning F. Celiac disease: caught between a rock and a hard place. Gastroenterology. 2005;129:1294–1301.
    1. Clemente MG, De Virgiliis S, Kang JS, et al. Early effects of gliadin on enterocyte intracellular signalling involved in intestinal barrier function. Gut. 2003;52:218–223.
    1. Vorobjova T, Uibo O, Ojakivi I, et al. Lower expression of tight junction protein 1 gene and increased FOXP3 expression in the small bowel mucosa in coeliac disease and associated type 1 diabetes mellitus. Inter Arch Allergy Immunol. 2011;156:451–461.
    1. Schumann M, Gunzel D, Buergel N, et al. Cell polarity-determining proteins Par-3 and PP-1 are involved in epithelial tight junction defects in coeliac disease. Gut. 2012;61:220–228.
    1. van Elburg RM, Uil JJ, Mulder CJ, et al. Intestinal permeability in patients with coeliac disease and relatives of patients with coeliac disease. Gut. 1993;34:354–357.
    1. Wapenaar MC, Monsuur AJ, van Bodegraven AA, et al. Associations with tight junction genes PARD3 and MAGI2 in Dutch patients point to a common barrier defect for coeliac disease and ulcerative colitis. Gut. 2008;57:463–467.
    1. van Heel DA, Franke L, Hunt KA, et al. A genome-wide association study for celiac disease identifies risk variants in the region harboring IL2 and IL21. Nat Genet. 2007;39:827–829.
    1. Hrdlickova B, Westra HJ, Franke L, et al. Celiac disease: moving from genetic associations to causal variants. Clin Genet. 2011;80:203–313.
    1. Gopalakrishnan S, Tripathi A, Tamiz AP, et al. Larazotide acetate promotes tight junction assembly in epithelial cells. Peptides. 2012;35:95–101.
    1. Gopalakrishnan S, Durai M, Kitchens K, et al. Larazotide acetate regulates epithelial tight junctions in vitro and in vivo. Peptides. 2012;35:86–94.
    1. Di Pierro M, Lu R, Uzzau S, et al. Zonula occludens toxin structure-function analysis. Identification of the fragment biologically active on tight junctions and of the zonulin receptor binding domain. J Biol Chem. 2001;276:19160–19165.
    1. Paterson BM, Lammers KM, Arrieta MC, et al. 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.
    1. Meddings JB, Gibbons I. Discrimination of site-specific alterations in gastrointestinal permeability in the rat. Gastroenterology. 1998;114:83–92.
    1. Dimenas E, Carlsson G, Glise H, et al. Relevance of norm values as part of the documentation of quality of life instruments for use in upper gastrointestinal disease. Scand J Gastroenterol Suppl. 1996;221:8–13.
    1. Dimenas E, Glise H, Hallerback B, et al. Well-being and gastrointestinal symptoms among patients referred to endoscopy owing to suspected duodenal ulcer. Scand J Gastroenterol. 1995;30:1046–1052.
    1. Revicki DA, Wood M, Wiklund I, et al. Reliability and validity of the Gastrointestinal symptom rating scale in patients with gastroesophageal reflux disease. Qual Life Res. 1998;7:75–83.
    1. Mustalahti K, Lohiniemi S, Collin P, et al. Gluten-free diet and quality of life in patients with screen-detected celiac disease. Eff Clin Pract. 2002;5:105–113.
    1. Dupuy HJ.The psychological general well-being (PGWB) indexIn: Wenger NK, Mattson ME, Furberg CF, et al., (eds). Assessment of Quality of Life in Clinical Trials of Cardiovascular Therapies Le Jacq Publishing: New York; 1984170–183.
    1. Lohiniemi S, Maki M, Kaukinen K, et al. Gastrointestinal symptoms rating scale in coeliac disease patients on wheat starch-based gluten-free diets. Scand J Gastroenterol Suppl. 2000;35:947–949.
    1. Hallert C, Granno C, Grant C, et al. Quality of life of adult coeliac patients treated for 10 years. Scand J Gastroenterol Suppl. 1998;33:933–938.
    1. Ukkola A, Maki M, Kurppa K, et al. Diet improves perception of health and well-being in symptomatic, but not asymptomatic, patients with celiac disease. Clin Gastroenterol Hepatol. 2011;9:118–123.
    1. Hallert C, Svensson M, Tholstrup J, et al. Clinical trial: B vitamins improve health in patients with coeliac disease living on a gluten-free diet. Aliment Pharmacol Ther. 2009;29:811–816.

Source: PubMed

3
구독하다