Effect of hookworm infection on wheat challenge in celiac disease--a randomised double-blinded placebo controlled trial

A James Daveson, Dianne M Jones, Soraya Gaze, Henry McSorley, Andrew Clouston, Andrew Pascoe, Sharon Cooke, Richard Speare, Graeme A Macdonald, Robert Anderson, James S McCarthy, Alex Loukas, John Croese, A James Daveson, Dianne M Jones, Soraya Gaze, Henry McSorley, Andrew Clouston, Andrew Pascoe, Sharon Cooke, Richard Speare, Graeme A Macdonald, Robert Anderson, James S McCarthy, Alex Loukas, John Croese

Abstract

Background and aims: The association between hygiene and prevalence of autoimmune disease has been attributed in part to enteric helminth infection. A pilot study of experimental infection with the hookworm Necator americanus was undertaken among a group of otherwise healthy people with celiac disease to test the potential of the helminth to suppress the immunopathology induced by gluten.

Methods: In a 21-week, double-blinded, placebo-controlled study, we explored the effects of N. americanus infection in 20 healthy, helminth-naïve adults with celiac disease well controlled by diet. Staged cutaneous inoculations with 10 and 5 infective 3(rd) stage hookworm larvae or placebo were performed at week-0 and -12 respectively. At week-20, a five day oral wheat challenge equivalent to 16 grams of gluten per day was undertaken. Primary outcomes included duodenal Marsh score and quantification of the immunodominant α-gliadin peptide (QE65)-specific systemic interferon-γ-producing cells by ELISpot pre- and post-wheat challenge.

Results: Enteric colonisation with hookworm established in all 10 cases, resulting in transiently painful enteritis in 5. Chronic infection was asymptomatic, with no effect on hemoglobin levels. Although some duodenal eosinophilia was apparent, hookworm-infected mucosa retained a healthy appearance. In both groups, wheat challenge caused deterioration in both primary and several secondary outcomes.

Conclusions: Experimental N. americanus infection proved to be safe and enabled testing its effect on a range of measures of the human autoimmune response. Infection imposed no obvious benefit on pathology.

Trial registration: ClinicalTrials.gov NCT00671138.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist. Envoi Specialist Pathologists, Brisbane, QLD, Australia contributed to the processing and the interpretation (Prof. Clouston) of the histological material, but neither the organisation nor any employee has any commercial interest beyond this support nor a competing interest. Envoi's involvement does not affect or alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. CONSORT flowchart.
Figure 1. CONSORT flowchart.
Figure 2. Recruitment and Protocol.
Figure 2. Recruitment and Protocol.
Recruitment and summation of those excluded post screening (A), and trial outline (B).
Figure 3. Representative photographs of parasitic infection…
Figure 3. Representative photographs of parasitic infection (A) Adult hookworm (B) Inoculation site.
Figure 4. Symptom responses to hookworm infection.
Figure 4. Symptom responses to hookworm infection.
Control group is indicated by black circles, hookworm group by grey squares, values are mean +/− SEM. (A) pain events, (B) bowel motions/day, (C) flatulence, (D) skin rash, (E) lethargy, (F) wellbeing. Area under the curve analysis followed by Mann-Whitney U test to compare between groups showed that pain events (A) was significantly different between the groups.
Figure 5. Blood cell and marker levels.
Figure 5. Blood cell and marker levels.
(A) white cell, (B) eosinophil (C) hemoglobin. Control group is indicated by black circles, hookworm group by grey squares, values are mean +/− SEM. Data was analyzed by two-way ANOVA: significant effects of time and interaction was shown in (B), and post-hoc one-way ANOVA on each group showed the differences between timepoints as indicated.
Figure 6. Histological and immunological results.
Figure 6. Histological and immunological results.
(A) Marsh score and (B) IFN-γ ELISpot. Data was analyzed by two-way ANOVA: significant effects of time only were shown as indicated, no significant effect of group or interaction was found in (A) or (B).

References

    1. Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. The N Engl J Med. 2002;347:911–920.
    1. Weinstock JV, Elliott DE. Helminths and the IBD hygiene hypothesis. Inflamm Bowel Dis. 2009;15:128–133.
    1. Altmann DM. Review series on helminths, immune modulation and the hygiene hypothesis: nematode coevolution with adaptive immunity, regulatory networks and the growth of inflammatory diseases. Immunology. 2009;126:1–2.
    1. Jackson JA, Friberg IM, Little S, Bradley JE. Review series on helminths, immune modulation and the hygiene hypothesis: immunity against helminths and immunological phenomena in modern human populations: coevolutionary legacies? Immunology. 2009;126:18–27.
    1. Fumagalli M, Pozzoli U, Cagliani R, Comi GP, Riva S, et al. Parasites represent a major selective force for interleukin genes and shape the genetic predisposition to autoimmune conditions. J Exp Med. 2009;206(6):1395–1408.
    1. Aoyama H, Hirata T, Sakugawa H, Watanabe T, Miyagi S, et al. An inverse relationship between autoimmune liver diseases and Strongyloides stercoralis infection. Am J Trop Med Hyg. 2007;76:972–976.
    1. Correale J, Farez M. Association between parasite infection and immune responses in multiple sclerosis. Ann Neurol. 2007;61:97–108.
    1. Elliott DE, Weinstock JV. Helminthic therapy: using worms to treat immune-mediated disease. Adv Exp Med Biol. 2009;666:157–166.
    1. Ruyssers NE, De Winter BY, De Man JG, Ruyssers ND, Van Gils AJ, et al. Schistosoma mansoni proteins attenuate gastrointestinal motility disturbances during experimental colitis in mice. World J Gastroenterol. 2010;16:703–712.
    1. Musgrove P, Hotez PJ. Turning neglected tropical diseases into forgotten maladies. Health Aff (Millwood) 2009;28:1691–1706.
    1. Feary J, Venn A, Brown A, Hooi D, Falcone FH, et al. Safety of hookworm infection in individuals with measurable airway responsiveness: a randomized placebo-controlled feasibility study. Clin Exp Allergy. 2009;39:1060–1068.
    1. Bager P, Arnved J, Ronborg S, Wohlfahrt J, Poulsen LK, 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.
    1. Summers RW, Elliott DE, Qadir K, Urban JF, Jr, Thompson R, et al. Trichuris suis seems to be safe and possibly effective in the treatment of inflammatory bowel disease. Am J Gastroenterol. 2003;98:2034–2041.
    1. Summers RW, Elliott DE, Urban JF, Jr, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005;128:825–832.
    1. Hotez PJ, Brooker S, Bethony JM, Bottazzi ME, Loukas A, et al. Hookworm infection. N Engl J Med. 2004;351:799–807.
    1. Maxwell C, Hussain R, Nutman TB, Poindexter RW, Little MD, et al. The clinical and immunologic responses of normal human volunteers to low dose hookworm (Necator americanus) infection. Am J Trop Med Hyg. 1987;37:126–134.
    1. Wright V, Bickle Q. Immune responses following experimental human hookworm infection. Clin Exp Immunol. 2005;142:398–403.
    1. Croese J, Wood MJ, Melrose W, Speare R. Allergy controls the population density of Necator americanus in the small intestine. Gastroenterology. 2006;131:402–409.
    1. Mortimer K, Brown A, Feary J, Jagger C, Lewis S, et al. Dose-ranging study for trials of therapeutic infection with Necator americanus in humans. Am J Trop Med Hyg. 2006;75:914–920.
    1. Croese J, O'Neil J, Masson J, Cooke S, Melrose W, et al. A proof of concept study establishing Necator americanus in Crohn's patients and reservoir donors. Gut. 2006;55:136–137.
    1. Quinnell RJ, Bethony J, Pritchard DI. The immunoepidemiology of human hookworm infection. Parasite Immunol. 2004;26:443–454.
    1. Leffler DA, Dennis M, Edwards George J, Jamma S, Cook EF, et al. A validated disease-specific symptom index for adults with celiac disease. Clin Gastroenterol Hepatol. 2009;7:1328–1334.
    1. Green PH, Cellier C. Celiac disease. N Engl J Med. 2007;357:1731–1743.
    1. Catassi C, Fabiani E, Iacono G, D'Agate C, Francavilla R, 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. Marsh MN, Crowe PT. Morphology of the mucosal lesion in gluten sensitivity. Baillieres Clin Gastroenterol. 1995;9:273–293.
    1. Tye-Din J, Anderson R. Immunopathogenesis of celiac disease. Curr Gastroenterol Rep. 2008;10:458–465.
    1. Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AV. In vivo antigen challenge in celiac disease identifies a single transglutaminase-modified peptide as the dominant A-gliadin T-cell epitope. Nat Med. 2000;6:337–342.
    1. Kagnoff MF. Celiac disease: pathogenesis of a model immunogenetic disease. J Clin Invest. 2007;117:41–49.
    1. Blount D, Hooi D, Feary J, Venn A, Telford G, et al. Immunologic profiles of persons recruited for a randomized, placebo-controlled clinical trial of hookworm infection. Am J Trop Med Hyg. 2009;81:911–916.
    1. Palmer ED. Course of egg output over a 15 year period in a case of experimentally induced necatoriasis americanus, in the absence of hyperinfection. Am J Trop Med Hyg. 1955;4:756–757.
    1. Beaver PC. Light, long-lasting Necator infection in a volunteer. Am J Trop Med Hyg. 1988;39:369–372.
    1. Feary JR, Venn AJ, Mortimer K, Brown AP, Hooi D, et al. Experimental hookworm infection: a randomized placebo-controlled trial in asthma. Clin Exp Allergy. 2010;40:299–306.
    1. Summers RW, Elliott DE, Urban JF, Jr, Thompson R, Weinstock JV. Trichuris suis therapy in Crohn's disease. Gut. 2005;54:87–90.
    1. Glas J, Stallhofer J, Ripke S, Wetzke M, Pfennig S, et al. Novel genetic risk markers for ulcerative colitis in the IL2/IL21 region are in epistasis with IL23R and suggest a common genetic background for ulcerative colitis and celiac disease. Am J Gastroenterol. 2009;104:1737–1744.

Source: PubMed

3
Tilaa