Elsevier

Methods in Enzymology

Volume 502, 2012, Pages 241-271
Methods in Enzymology

Chapter ten - Oral Enzyme Therapy for Celiac Sprue

https://doi.org/10.1016/B978-0-12-416039-2.00013-6Get rights and content

Abstract

Celiac sprue is an inflammatory disease of the small intestine caused by dietary gluten and treated by adherence to a life-long gluten-free diet. The recent identification of immunodominant gluten peptides, the discovery of their cogent properties, and the elucidation of the mechanisms by which they engender immunopathology in genetically susceptible individuals have advanced our understanding of the molecular pathogenesis of this complex disease, enabling the rational design of new therapeutic strategies. The most clinically advanced of these is oral enzyme therapy, in which enzymes capable of proteolyzing gluten (i.e., glutenases) are delivered to the alimentary tract of a celiac sprue patient to detoxify ingested gluten in situ. In this chapter, we discuss the key challenges for discovery and preclinical development of oral enzyme therapies for celiac sprue. Methods for lead identification, assay development, gram-scale production and formulation, and lead optimization for next-generation proteases are described and critically assessed.

Introduction

Celiac sprue is a complex inflammatory disease of the small intestine involving both genetic and environmental factors. It is unique among chronic bowel disorders in that a specific dietary component—gluten from wheat and related protein content from rye and barley—has been identified as the proximate environmental cause of inflammation (Dicke et al., 1953). Gluten is a heterogeneous mixture of water insoluble storage proteins comprising monomeric gliadins and polymeric glutenin aggregates up to 10 MDa in size (Wieser, 2007). Both gliadins and glutenins contain repetitive sequences that are rich in proline (Pro; 15%) and glutamine (Gln; 35%), residues that are not preferred substrates for any human digestive enzyme. Consequently, gluten is relatively resistant to gastrointestinal proteolysis, releasing metastable Pro/Gln-rich peptides up to 30–40 amino acids in length into the gut lumen following digestion (Fig. 10.1; Shan et al., 2002, Shan et al., 2005b). Intestinal transglutaminase 2 (TG2) deamidates some of these peptides at specific Gln residues (Molberg et al., 1998, Shan et al., 2002), thereby increasing their affinity for human leukocyte antigen (HLA) DQ2 (Quarsten et al., 1999), a major histocompatibility class II molecule associated with over 90% of celiac patients (Sollid et al., 1989). Deamidated gluten peptide–DQ2 complexes on the surface of antigen-presenting cells (APCs) elicit a potent Th1 inflammatory response from gluten-specific intestinal T cells (Nilsen et al., 1995, Nilsen et al., 1998). Ultimately, this inflammatory response causes destruction of the intestinal architecture, malabsorption of nutrients, and, in many patients, numerous secondary symptoms including diarrhea, wasting, and anemia (Alaedini and Green, 2005). Complete and life-long adherence to a gluten-free diet reverses the signs and symptoms of celiac sprue in most patients. However, the ubiquity of gluten in human diets renders this an extraordinarily difficult prescription to follow, leading to frequent lapses and chronic morbidity in many patients (Ciacci et al., 2002, Cornell et al., 2005, Mayer et al., 1991, Pietzak, 2005). Nondietary therapies that detoxify ingested gluten would therefore improve quality of life considerably for the 1–2% of people afflicted with this life-long disease.

A number of therapeutic alternatives to the gluten-free diet are under development, including enzymatic detoxification of gluten, inhibition of TG2 to prevent peptide deamidation, antagonism of deamidated peptide binding to HLA-DQ2, or suppression of deleterious immune responses (reviewed in Schuppan et al., 2009). In this chapter, we focus on enzymatic detoxification of gluten by oral administration of enzymes capable of digesting gluten (i.e., glutenases).

Oral enzyme therapy is attractive for several reasons:

First, it follows the lead of the established gluten-free diet by targeting the exogenous pathogen, gluten, rather than targeting endogenous effectors. During clinical development, gluten intake and glutenase dose can be titrated in a complementary, dynamic, and individualized manner, thereby enabling systematic analysis of drug pharmacodynamics.

Second, by targeting ingested gluten, oral enzyme therapy avoids the shortcomings of strategies that target gluten at the level of exposure, including the gluten-free diet, selection or genetic engineering of less toxic grains (Molberg et al., 2005, Spaenij-Dekking et al., 2005), or pretreatment of gluten with glutenases, analogous to pretreatment of dairy products with lactase (De Angelis et al., 2006). Because the same proteins that cause toxicity in patients impart the desirable viscoelastic properties of dough, genetic or enzymatic removal of toxic epitopes from gluten will likely reduce baking quality. More importantly, such strategies do not address inadvertent exposure to ubiquitous gluten.

Third, the absence of a bona fide animal model for gluten sensitivity precludes preclinical safety and efficacy testing of drugs targeting endogenous effectors (Bethune and Khosla, 2008). By contrast, oral enzyme therapy can be evaluated in vivo using healthy or existing gluten-sensitive animal models (Bethune et al., 2008, Gass et al., 2006).

Fourth, glutenases that incompletely detoxify gluten as monotherapies can be coadministered with enzymes of complementary specificity to synergistic benefit (Gass et al., 2007a, Siegel et al., 2006).

This chapter describes methods for preclinical evaluation of oral enzyme therapy for celiac sprue. In Section 2, we provide strategies for identifying novel glutenase candidates. In Section 3, we provide protocols for characterizing the enzymatic properties of these candidates using gluten-derived and surrogate substrates. In Section 4, we describe methods for gram-scale production and formulation of promising candidates for delivery to alimentary sites of action. Finally, in Section 5, we discuss prospects for evolving next-generation glutenases and delivering them to the gut by various mechanisms.

Section snippets

Lead Identification

Oral enzyme therapy requires proteases that can digest the uniquely Pro- and Gln-rich sequences constituting immunogenic gluten epitopes. Moreover, these enzymes must be stable and active in the harsh environments of the stomach and/or upper small intestine. In this section, we describe strategies for identifying novel glutenases for oral enzyme therapy. These strategies conform to two general approaches. The first approach is to rationally select glutenase candidates based on therapeutically

Assay Development

A bona fide animal model for celiac sprue has not been discovered or engineered. Assays for glutenase efficacy are therefore conducted in vitro, ex vivo, and in surrogate animal models (e.g., in gluten-tolerant animals or in incomplete animal models of gluten sensitivity). Primary assays utilize biochemical tools such as spectrophotometry, reversed-phase HPLC, and liquid chromatography-assisted mass spectrometry (LC-MS) to identify disease-relevant gluten peptides and to preliminarily assess

Gram-Scale Production and Formulation

Lead glutenases that show promise in early (i.e., in vitro) experiments must be evaluated in more advanced gastrointestinal models and/or animal surrogates prior to human clinical trials. Such studies require gram-scale quantities of glutenase, necessitating high-yield production methods. In this section, we describe the heterologous expression, purification, formulation, and storage of gram-scale quantities of recombinant glutenases.

Lead Optimization for Next-Generation Proteases

In previous sections, we described strategies for identifying and characterizing existing proteases for use in oral enzyme therapy. In this section, we discuss strategies for improving on these enzymes, either by simply coadministering them as a combination glutenase or by augmenting their therapeutically desirable properties through protein engineering. Additionally, we discuss potential modes of delivery for enzyme therapy, oral, and otherwise.

Summary and Outlook

Since the identification in 1953 of gluten as the primary environmental trigger of celiac sprue (Dicke et al., 1953), we have gained an expansive understanding of the genetic, immunological, and biochemical basis for the pathogenesis of this complex disease. Nonetheless, over this same timespan, the prevalence of celiac sprue has increased fourfold to 1–2% of humans (Lohi et al., 2007, Rubio-Tapia et al., 2009) and the only treatment available to this growing population remains a life-long

References (91)

  • T. Kiyosaki et al.

    Wheat cysteine proteases triticain alpha, beta and gamma exhibit mutually distinct responses to gibberellin in germinating seeds

    J. Plant Physiol.

    (2009)
  • C.Y. Lui et al.

    Comparison of gastrointestinal pH in dogs and humans: Implications on the use of the beagle dog as a model for oral absorption in humans

    J. Pharm. Sci.

    (1986)
  • C. Mitea et al.

    Fine specificity of monoclonal antibodies against celiac disease-inducing peptides in the gluteome

    Am. J. Clin. Nutr.

    (2008)
  • O. Molberg et al.

    Mapping of gluten T-cell epitopes in the bread wheat ancestors: Implications for celiac disease

    Gastroenterology

    (2005)
  • B. Moron et al.

    Sensitive detection of cereal fractions that are toxic to celiac disease patients by using monoclonal antibodies to a main immunogenic wheat peptide

    Am. J. Clin. Nutr.

    (2008)
  • E.M. Nilsen et al.

    Gluten induces an intestinal cytokine response strongly dominated by interferon gamma in patients with celiac disease

    Gastroenterology

    (1998)
  • M.M. Pietzak

    Follow-up of patients with celiac disease: Achieving compliance with treatment

    Gastroenterology

    (2005)
  • G.G. Pyle et al.

    Effect of pretreatment of food gluten with prolyl endopeptidase on gluten-induced malabsorption in celiac sprue

    Clin. Gastroenterol. Hepatol.

    (2005)
  • A. Rubio-Tapia et al.

    Increased prevalence and mortality in undiagnosed celiac disease

    Gastroenterology

    (2009)
  • D. Schuppan et al.

    Celiac disease: From pathogenesis to novel therapies

    Gastroenterology

    (2009)
  • M. Siegel et al.

    Rational design of combination enzyme therapy for celiac sprue

    Chem. Biol.

    (2006)
  • D.J. Simpson

    Proteolytic degradation of cereal prolamins—The problem with proline

    Plant Sci.

    (2001)
  • S. Souliman et al.

    A level A in vitro/in vivo correlation in fasted and fed states using different methods: Applied to solid immediate release oral dosage form

    Eur. J. Pharm. Sci.

    (2006)
  • L. Spaenij-Dekking et al.

    Natural variation in toxicity of wheat: Potential for selection of nontoxic varieties for celiac disease patients

    Gastroenterology

    (2005)
  • D. Stepniak et al.

    Enzymatic gluten detoxification: The proof of the pudding is in the eating!

    Trends Biotechnol.

    (2006)
  • R. Sturgess et al.

    Wheat peptide challenge in coeliac disease

    Lancet

    (1994)
  • J.A. Tye-Din et al.

    The effects of ALV003 pre-digestion of gluten on immune response and symptoms in celiac disease in vivo

    Clin. Immunol.

    (2010)
  • H. Wieser

    Chemistry of gluten proteins

    Food Microbiol.

    (2007)
  • A. Alaedini et al.

    Narrative review: Celiac disease: Understanding a complex autoimmune disorder

    Ann. Intern. Med.

    (2005)
  • A. Berger et al.

    Mapping the active site of papain with the aid of peptide substrates and inhibitors

    Philos. Trans. R. Soc. Lond. B. Biol. Sci.

    (1970)
  • M.T. Bethune et al.

    Parallels between pathogens and gluten peptides in celiac sprue

    PLoS Pathog.

    (2008)
  • M.T. Bethune et al.

    Transepithelial transport and enzymatic detoxification of gluten in gluten-sensitive rhesus macaques

    PLoS One

    (2008)
  • S. Blanquet et al.

    A dynamic artificial gastrointestinal system for studying the behavior of orally administered drug dosage forms under various physiological conditions

    Pharm. Res.

    (2004)
  • C. Catassi et al.

    Risk of non-Hodgkin lymphoma in celiac disease

    JAMA

    (2002)
  • N. Cerf-Bensussan et al.

    Oral proteases: A new approach to managing coeliac disease

    Gut

    (2007)
  • C. Ciacci et al.

    Long-term follow-up of celiac adults on gluten-free diet: Prevalence and correlates of intestinal damage

    Digestion

    (2002)
  • H.J. Cornell et al.

    Enzyme therapy for management of coeliac disease

    Scand. J. Gastroenterol.

    (2005)
  • A. Davy et al.

    Substrate specificity of barley cysteine endoproteases EP-A and EP-B

    Plant Physiol.

    (1998)
  • M. De Angelis et al.

    VSL#3 probiotic preparation has the capacity to hydrolyze gliadin polypeptides responsible for celiac sprue

    Biochim. Biophys. Acta

    (2006)
  • M. De Angelis et al.

    Mechanism of degradation of immunogenic gluten epitopes from Triticum turgidum L. var. durum by sourdough lactobacilli and fungal proteases

    Appl. Environ. Microbiol.

    (2010)
  • W.K. Dicke et al.

    Coeliac disease II. The presence in wheat of a factor having a deleterious effect in cases of coeliac disease

    Acta Paediatr.

    (1953)
  • J.B. Dressman

    Comparison of canine and human gastrointestinal physiology

    Pharm. Res.

    (1986)
  • F.P. Duan et al.

    Interrupting Vibrio cholerae infection of human epithelial cells with engineered commensal bacterial signaling

    Biotechnol. Bioeng.

    (2008)
  • F. Duan et al.

    Secretion of insulinotropic proteins by commensal bacteria: Rewiring the gut to treat diabetes

    Appl. Environ. Microbiol.

    (2008)
  • L. Edens et al.

    Extracellular prolyl endoprotease from Aspergillus niger and its use in the debittering of protein hydrolysates

    J. Agric. Food Chem.

    (2005)
  • Cited by (68)

    • New coeliac disease treatments and their complications

      2018, Gastroenterologia y Hepatologia
    View all citing articles on Scopus
    View full text