Celiac Dz - Peds

download Celiac Dz - Peds

of 12

Transcript of Celiac Dz - Peds

  • 8/12/2019 Celiac Dz - Peds

    1/12

    Silent CD describes the situation of an individualwithout signs or symptoms of CD, but who has small

    bowel biopsy evidence of CD; usually these patients

    have an associated condition or a family history of

    CD, and are identified on screening as having CD-

    associated antibodies. Latent CD applies to individuals without signs or

    symptoms of CD, but who have some risk for future

    development of CD, such as expression of CD-

    related antibodies or DQ2 or DQ8 permissive genes,

    family history of CD, or having an associated condi-

    tion. These individuals do not have small bowel

    biopsy changes, but may have CD-associated anti-

    bodies. Refractory CD describes an individual with defined

    CD who continues to have signs or symptoms of

    active CD despite pursuing a GFD. In this situation,

    considerations include considered exposure to gluten,

    enteropathy-associated T-cell lymphoma (EATL), or

    possibly another condition such as allergy or inflam-

    matory bowel disease.

    Incidence and Prevalence

    The prevalence in the United States and Europe is

    roughly 313 cases per 1000 individuals (1:300 to

    1:80).1There is a female predominance with a ratio of

    roughly 2:1.2 These estimates indicate that there areapproximately 3 million people with CD in the United

    States alone, and a roughly equal number in Europe, of

    which 90% are undiagnosed (Table 181). Recent

    screening studies suggest that in developing countries

    in Africa, parts of Asia, and South America, the fre-

    quency is similar to that of the U.S. and European

    countries.3To date, there are very little data exploring

    DEFINITIONS AND EPIDEMIOLOGYCeliac disease (CD) is a permanent sensitivity to gluten

    in wheat and related proteins found in barley and rye,

    occurring in genetically susceptible individuals, and

    manifesting as an immune mediated enteropathy as

    defined by characteristic changes seen on intestinal his-

    tology.1A conservative definitionrequires the following:

    typical signs or symptoms; presence of CD-associated antibodies; a small intestinal biopsy showing villous atrophy; resolution of clinical manifestations with a gluten-

    free diet (GFD), including complete healing of the

    intestinal mucosa; reduction or disappearance of the CD-associated

    antibodies on a GFD.

    In practice, it is questionable whether it is neces-

    sary to meet all aspects of this definition. Controversy

    continues about whether a small bowel biopsy is required

    to diagnose CD. Because of numerous reports of CD-

    associated seropositive individuals with no signs or

    symptoms of CD, guidelines continue to require a

    biopsy to confirm the diagnosis and need for

    treatment.

    Different terms have been applied to common

    clinical situations:

    Classic CDrefers to a presentation with typical clini-cal features such as diarrhea, abdominal pain, failure

    to thrive, or abdominal distention. Atypical CDdescribes a non-traditional presentation,

    primarily with extraintestinal manifestations, such as

    arthritis or iron deficiency anemia. In older children

    and adults, the atypical presentation may be more

    common than the classic presentation.

    Celiac DiseaseEdward Hoffenberg

    CHAPTER18

    Bishop_Ch18_265 276.indd 265Bishop_Ch18_265-276.indd 265 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    2/12

    266 Section 3: Disorders of the Stomach and Intestine

    the rates of CD in China, Japan, and Southeast Asian

    countries, and these populations are thought to be at

    lower genetic risk for CD. A number of conditions are

    associated with an increased risk of CD in children and

    adults (Table 182).

    Genetic Considerations

    Specific HLA types are permissive for development of

    CD. HLA molecules bind peptide antigens and presentthem to CD4helper T cells. An estimated 95% of CD

    patients express the HLA-DQ2 gene and the remain-

    der express DQ8.5HLA II molecules are made up of

    dimers, expressing one alpha and one beta chain. The

    vast majority of CD patients express the HLA II sub-

    type DQ2 coded by alleles DQA1*0501 and DQB*0201,

    or DQ8 coded by alleles DRB*04-DQA1*03-

    DQB1*0302. The DQ2 dimer contains specific pockets

    that bind deamidated gluten peptides and present

    them to CD4lymphocytes. Expressing two copies of

    HLA-DQ2 (homozygous state) increases the risk for

    developing CD, and an estimated 25% of CD patients

    are DQ2 homozygous. The genes coding for HLA II

    are located on chromosome 6p21, termed the CELIAC1locus.

    However, other genetic factors are important,

    and the absolute necessity of DQ2/DQ8 for develop-

    ment of CD has been questioned, as apparent cases of

    CD occur in individuals who do not express DQ2 or

    DQ8.2,4The DQ2 gene is expressed by approximately

    30% of Caucasians, but only 3% of these develop CD.

    Having a first-degree relative with CD increases the

    risk of CD to between 5% and 18%.4There is an esti-

    mated 7086% concordance in monozygotic twins

    with CD, but only 3040% with HLA-matched twins,

    and less than 20% concordance in dizygotic twins and

    first-degree relatives.6Expression of DQ2 or DQ8 onlyaccounts for approximately 3653% of the disease

    risk. Therefore, other genetic and environmental fac-

    tors are important.7,8 Gene linkage analysis and

    genetic-associated studies, including genome-wide

    association studies, have identified a number of other

    loci of possible importance in CD5,9 (Tables 183

    and 184).

    Worldwide Disease Prevalence Based on

    Screening Data

    Argentina

    Australia

    Brazil

    Egypt

    Finland

    India

    Iran

    Israel

    Italy

    Netherlands

    Russia

    Spain

    Sweden

    Tunisia

    Turkey

    United KingdomUnited States

    1:167

    1:251

    1:66 to 1:119

    1:187

    1:99 to 1:67

    1:310

    1:166 to 1:104

    1:157

    1:106

    1:198

    1:133

    1:118

    1:77

    1:157

    1:115

    1:1001:105

    Table 181.

    Conditions Associated with Increased Risk for CD

    in Children and Adults1,4,11

    Condition Prevalence of CD (%)

    Type 1 diabetes

    Down syndrome

    Turner syndrome

    Williams syndrome

    IgA deficiency

    Autoimmune thyroiditis

    First-degree relatives with CD

    Second-degree relatives

    Dermatitis herpetiformis

    Short stature/delayed puberty

    Dental enamel defects

    Adults specifically: Irritable bowel syndrome Persistent aphthous stomatitis Peripheral neuropathy Unexplained cerebellar ataxia Elevated transaminases Unexplained iron deficiency

    anemia Decreased bone mineral density Infertility

    816

    315

    48

    10

    28

    8

    518

    3

    80100

    Table 182.

    Genetic Linkage Studies5,9

    Locus Gene Region Function

    CELIAC1 6p21 HLA II coding region

    CELIAC2 5q315q33 Cytokine gene cluster

    CELIAC3 2q33 T-cell regulatory genes

    CELIAC4 19p13.1 MYO9Bunconventional

    myosin molecule

    Table 183.

    Bishop_Ch18_265 276.indd 266Bishop_Ch18_265-276.indd 266 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    3/12

    CHAPTER 18 Celiac Disease 267

    Pathogenesis

    The exact mechanism by which ingestion of gluten pro-

    teins by genetically susceptible individuals leads to

    immune-mediated intestinal injury remains unclear

    (Figure 181). An inciting event such as a viral infec-

    tion, plus additional genetic factors, may be important

    to development of CD. Both innate and adaptive immu-

    nity are thought to be involved.

    Gluten proteins are found in wheat and related

    grains, and can be divided into gliadin and glutenin

    components. Hordeins in barley and secalins in rye are

    equivalent proteins that promote the CD process. The

    oat avedin protein is more distantly related to Triticeae

    Gluten

    Gut lumen Brush border

    Deamidated

    peptideAPC

    T cell

    TG2

    Mucosal

    epithelium

    TG2

    CD4 + T cell

    CD4 TCR

    HLA-DQ2or -DQ8

    APC

    Lamina propria

    FIGURE 181 Cartoon highlighting steps involved in pathogenesis of CD.41 Gluten peptides survive digestion and

    cross the mucosal epithelium, where deamidation by tissue transglutaminase (TG2) occurs. HLA-DQ2 or -DQ8 molecules

    on antigen-presenting cells present these peptides in the intestinal lamina propria where they are recognized by specific

    CD4T cells Adapted by permission from Ref.41(Macmillan Publishers Ltd).

    Genetic Association Studies5,4345

    Gene Region Function/Association

    1q31 Contains RGS1, regulator of G protein signaling molecule. Involved in B-cell act ivation/proliferation; found

    in intestinal intraepithelial lymphocytes

    2q112q12 2 genes coding for receptor for IL-18 that stimulates T-cell synthesis of IFN-gamma

    3p21 Large cluster of chemokine receptor genes CCR1, CCR2, CCR3, CCR5, CCRL2, and XCR1

    3q253q26 Immediately 5of IL-12 gene. IL-12 induces Th1 IFN-gamma secretion

    3q28 Associated with LPP gene of unclear significance

    4q27 Strongest current association with CD outside HLA locus. Cluster of 8 associated single nucleotide

    polymorphisms; 2 contain genes for IL2 and IL21, involved in T-cell activation. Associated with type 1

    diabetes and rheumatoid arthritis

    6q25 Contains TAGAP gene (T-cell activation GTPase-activating protein) expressed in act ivated T cel ls

    12q24 Contains LNK and ATXN2strongly expressed in monocytes, dendritic cel ls, and small intest ine

    Table 184.

    Bishop_Ch18_265 276.indd 267Bishop_Ch18_265-276.indd 267 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    4/12

    268 Section 3: Disorders of the Stomach and Intestine

    glutens, and seems to be non-immunogenic for most

    CD patients.10

    Several properties of gluten may be important in

    stimulating the immune response of CD:

    Gliadin stimulation of IL-15 secretion through a non-

    HLA-binding mechanism. Gliadin induction of zonulin expression in small

    intestinal epithelium, with subsequent increase in

    intestinal permeability, CD4T-cell antigen exposure,

    and alteration in cell morphology. The high proline (15%) and glutamine (35%) content

    confers important chemical properties to gluten. First,

    gluten peptides resist human acid and peptic diges-

    tion, and reach the intestinal mucosa in antigenic seg-

    ments. Second, constitutively expressed tissue trans-

    glutaminase (TTG) in the small intestine alters gliadin

    peptides by deamidating specific glutamine residues

    to the negatively charged glutamate. Individuals with

    CD express antibodies to both TTG and deamidated

    gliadin peptides; these antibodies are specific sero-logic markers for CD. Deamidated gliadin peptides

    show enhanced binding to specific pockets in the DQ2

    molecule, resulting in enhanced presentation to

    CD4T cells by local antigen-presenting cells

    (APCs).

    Both activated APCs and CD4T cells secrete

    increased amounts of inflammatory cytokines such as

    IFN-gamma and IL-15, causing local recruitment of

    fibroblasts and clonal expansion of cytotoxic intraepi-

    thelial lymphocytes (IELs). The increased numbers of

    IELs and the subsequent mucosal damage are the

    classic changes seen on intestinal biopsy of CD

    patients.

    CLINICAL PRESENTATION

    Well-recognized clinical presentations of pediatric CD

    include: failure to thrive, chronic diarrhea, distended

    abdomen, irritability, constipation, and growth or puber-

    tal delay (Table 185).1,11 Extraintestinal manifestations

    such as arthritis, iron deficiency anemia, dental enameldefects, and dermatitis herpetiformis (DH) are also com-

    mon. Asymptomatic children may be identified on

    screening because of increased risk due to having an

    associated condition or a family history of CD or diabe-

    tes (Table 182). Recently, a variety of presentations have

    been imputed to be related to undiagnosed CD in adults

    (Table 182). These include neuropsychiatric presenta-

    tions such as ataxia and epilepsy, as well as infertility.

    Because the signs and symptoms may be mild, vague,

    and non-specific, delays in diagnosis are common.1113

    Celiac crisis is a rare medical emergency charac-

    terized by explosive, watery diarrhea and dehydration/

    electrolyte imbalances, marked abdominal distension,

    hypotension, and lethargy. It is typically seen in toddlers

    and responds to corticosteroids within a few days.

    Clinical Course/Progression of Disease

    With institution of a GFD, symptoms often improve by

    the second week, but may take months to completely

    resolve. Normalization of the intestinal histology may

    take 612 months. Lactose intolerance tends to resolve

    within a few weeks. Treatment of nutritional deficien-

    cies, such as iron deficiency, is generally not needed over

    long term. Adults tend to improve more slowly.

    For those with silent or latent CD, continued expo-

    sure to gluten leads to eventual clinical manifestations in

    Manifestations of CD1,11

    Classic CD Extraintestinal Neuropsychiatric Symptoms

    Failure to thrive* Dermatitis herpetiformis* Ataxia

    Diarrhea* Dental enamel defects* Epilepsy/cerebral calcifications

    Abdominal distension* Anemia* Migraines

    Vomiting* Aphthous stomatitis* Depression

    Abdominal pain* Arthritis/arthralgias Fatigue/malaise

    Constipation* Abnormal liver function tests* Anxiety

    Pubertal delay/short stature* Peripheral neuropathy

    Osteoporosis/osteopenia*

    Infertility

    Recurrent fetal loss

    *Strong evidence for association.Less strong.

    Table 185.

    Bishop_Ch18_265 276.indd 268Bishop_Ch18_265-276.indd 268 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    5/12

    CHAPTER 18 Celiac Disease 269

    at least some individuals.14Data from the adult literature

    also show a mortality risk significantly above the stan-

    dard mortality rate (SMR) by a factor of two- to three-

    fold for patients not adhering to a GFD, not responding

    to the GFD, or with lengthy delay in diagnosis.15There is

    reasonable evidence that early compliance with a GFD

    reduces the risk of mortality to close to baseline.16

    Malignancy

    The two- to three-fold increase in all-cause mortality in

    CD adult patients, as compared to controls, is mostly

    due to gastrointestinal malignancies such as EATL, non-

    Hodgkin lymphoma (NHL), and small bowel adenocar-

    cinoma. CD is associated with an increased relative risk

    2.16.6 times above baseline for NHL and a 30-fold

    increased risk for small bowel adenocarcinoma.16

    Dermatitis Herpetiformis

    DH is the most common skin lesion associated with CD,

    occurring in up to 24% of adult patients with CD, but is

    rare in children. Classic DH is a chronic, pruritic, papu-

    lar/vesicular rash symmetrically located on the extensor

    surfaces of the elbows, knees, buttocks, back, and sacrum,

    and occasionally is seen on the face, neck, and trunk.

    Biopsy of skin lesions shows microabscesses with neu-

    trophils and eosinophils in the dermal papillae. Immu-

    nofluorescence of adjacent skin shows IgA deposits in

    the papillary dermis directed toward an unknown anti-

    gen, but theorized to possibly be epidermal transglu-

    taminase. Markers of CD are often elevated in DH, with

    positive TTG IgA, endomyseal antibody assay (EMA),

    and anti-gliadin antibody assays (AGA). The vast major-ity of DH patients will have villous flattening or increased

    IELs, even in the absence of overt gastrointestinal symp-

    toms. Most, 8590%, are HLA B8 positive, and there is a

    strong association with HLA DW3, and DRW3. A glu-

    ten-containing diet is necessary to manifest the lesions of

    DH, and symptoms often resolve with GFD. Dapsone is

    often used as a suppressive medication.1,17,18

    Skeletal ManifestationsThere is strong evidence supporting the increased risk

    for osteopenia and osteoporosis in untreated CD. Bone

    mineral density, area, and content may be reduced but

    assessment should adjust for growth and pubertal delay.

    In children, bone density normalizes within 12 years of

    treatment with GFD.19

    Association with OtherAutoimmune Diseases

    The most commonly seen concurrent autoimmune dis-

    eases are type 1 diabetes and Hashimoto thyroiditis.

    Less common are associations with juvenile arthritis,alopecia areata, vitiligo, hepatitis and cholangitis,

    Sjogrens syndrome, Addisons disease, peripheral neu-

    ropathy, psoriasis, and autoimmune cardiomyopathy.

    DIFFERENTIAL DIAGNOSIS

    Because of its many and varied clinical features, CD can

    be a great imitator of other disorders, and it can also be

    silent. The differential diagnosis depends on the way it

    manifests as well as the age of the patient (Table 186).

    The presentation of diarrhea, abdominal distention,

    irritability, and vomiting may mimic gastroesophageal

    reflux, milk or soy protein allergy, Giardia infection,malrotation, lymphangiectasia, and iron deficiency ane-

    mia in toddlers. The same presentation in school age

    children and adolescents should lead to considerations

    Differential Diagnosis

    Toddler School Age and Adolescent Flattened Mucosa (Villous Atrophy)

    Gastroesophageal reflux Irritable bowel syndrome Malnutrition

    Milk or soy protein allergy Functional abdominal pain Infectious enteropathy: GiardiaGiardia Post-infectious chronic diarrhea Tropical sprue

    Lymphangiectasia Small bowel bacterial overgrowth IgA/immunodeficiency

    Malrotation Immunodeficiency (IgA deficiency, HIV, other) Allergic enteropathy: cow/soy milk

    Iron deficiency anemia IBD (Crohns disease) Autoimmune enteropathy

    Chronic constipation/

    Hirschsprungs

    Growth hormone deficiency ZollingerEllison syndrome

    Fibromyalgia

    Acid peptic disease, ulcer

    Table 186.

    Bishop_Ch18_265 276.indd 269Bishop_Ch18_265-276.indd 269 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    6/12

    270 Section 3: Disorders of the Stomach and Intestine

    of irritable bowel syndrome, inflammatory bowel dis-

    ease, lactose intolerance, ulcer, post-infectious diarrhea/

    bacterial overgrowth, as well as parasite infection, and

    immunodeficiency such as IgA deficiency or HIV. Of

    note, lactose intolerance may be a consequence of the

    intestinal injury of CD and often resolves with therapy.

    The histological hallmark of CD is the small bowel

    biopsy showing increased numbers of IELs and villous

    atrophy. The differential diagnosis for this histological

    finding includes HIV and immunodeficiency, allergy,

    Giardia infection, viral gastroenteritis, autoimmune

    enteropathy, ZollingerEllison syndrome, tropical sprue,

    and malnutrition (Table 186).

    DIAGNOSIS

    The criteria for diagnosis of CD have been recently

    updated separately by the North American Society for

    Pediatric Gastroenterology, Hepatology and Nutrition

    (NASPGHAN),1the Federation of International Societ-ies of Pediatric Gastroenterology Hepatology and Nutri-

    tion,20 the National Institutes of Health (NIH),11 and

    the American Gastroenterological Society (AGA).21,15

    Unfortunately, these guidelines are not uniform. In gen-

    eral, the diagnostic algorithm depends on whether the

    presentation includes clinical signs or symptoms (Fig-

    ure 182), or whether identified because of associated

    conditions or risks (Figure 183).

    The current NASPGHAN and FISPGHAN rec-

    ommendations are to use TTG antibody assays or EMA

    as the single screening test. These tests have sensitivity

    and specificity above 0.95. The role of new serologic

    tests such as the deamidated gliadin peptide antibody

    test is not yet clear. There is no benefit to a celiac panel

    of multiple antibody tests. Because of the rare case of

    CD in a patient with IgA deficiency, and to increase the

    confidence in a negative result, quantitative IgA levels

    may be obtained, or IgA plus IgG-based TTG tests

    ordered. AGA are no longer recommended for any

    patient suspected to be at risk for CD as it has a low

    specificity and high false-positive rate.

    A positive serologic screening test should be fol-

    lowed by a small bowel biopsy showing villous atrophy

    in order to confirm the diagnosis of CD, and to exclude

    other causes for the symptoms. The presence of increased

    IELs alone, the Marsh 1 lesion, is commonly seen in

    other conditions and experts do not consider it enough

    for diagnosis of CD. Multiple biopsies (four to six)

    should be taken from the second portion and distal

    duodenum. Villous atrophy with increase in IELs is the

    characteristic finding (Figure 184), but is not specificfor CD (see Table 186). Finally, a complete resolution

    of symptoms within weeks of following a GFD clinches

    the diagnosis in the child with a clinical presentation

    of CD.

    Value of DQ2/DQ8 Testing

    Virtually all patients with CD will test positive for DQ2

    or DQ 8. However, 3040% of Caucasians will express

    DQ2 or DQ8, and fewer than 5% will ever develop CD.

    Therefore, the value of HLA haplotype testing is in its

    Clinical signs or symptoms

    TTG and quantitative IgA

    Biopsy consistent

    with celiac disease

    Gluten free diet

    Biopsy uncertain

    Consider repeat biopsy, trialof gluten free diet, other

    diagnoses

    If normal, celiac disease

    unlikely, evaluate further

    If abnormal, refer topediatric GI for small

    bowel biopsy

    FIGURE 182 Diagnostic algorithm for evaluation of the symptomatic child.

    Bishop_Ch18_265 276.indd 270Bishop_Ch18_265-276.indd 270 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    7/12

    CHAPTER 18 Celiac Disease 271

    high negative predictive value. Situations to consider

    DQ2/8 testing include those individuals with an unclear

    diagnosis and those with a risk factor but negative initial

    screening. In this situation, if testing is negative for DQ2

    or DQ8, CD is unlikely.

    Current recommendations for biopsy-positive indi-

    viduals are to recheck TTG 6 months after initiating GFD

    to measure compliance and response, as serology should

    normalize during this time with good compliance. For the

    asymptomatic patient, TTG should be rechecked at inter-vals of 1 year or longer. Gluten challenge and repeat biopsy

    are no longer recommended if patient improves on GFD.

    If the patient has been on a GFD after serologic testing but

    prior to biopsy, or if the patient is receiving corticoster-

    oids or immunosuppressants, biopsy results may not

    reflect true level of disease. In this situation, a gluten

    load of 24 weeks of a gluten-containing diet usually is

    sufficient to produce typical CD changes on biopsy.

    CONTROVERSIES

    Stool Testing

    Fecal tests for CD-associated antibodies are commer-

    cially available but have not been well validated in adults

    and have not been studied in children.

    Rapid In-office Assays

    Rapid whole blood assays for TTG are also being com-

    mercialized, with as yet limited data on use in clinical

    practice.

    Asymptomatic, at-risk child

    TTG

    Biopsy consistentwith celiac disease

    Consider gluten-free diet

    Biopsy uncertain

    Consider repeat biopsy,trial of gluten-free diet

    If normal, consider re-screening every 2-3 years,sooner if clinical concerns

    arise

    If abnormal, either re-testto confirm, or refer topediatric GI for small

    bowel biopsy

    FIGURE 183 Diagnostic algorithm for evaluation of the asymptomatic child with

    a risk.

    FIGURE 184 Small bowel biopsy features of CD, hematoxylin and eosin stain. Panel A shows normal small bowel biopsy with tall slender

    villi, 10x magnification (Marsh score 0) Panel B shows tall villi with increased numbers of intra-epithelial lymphocytes, 40x magnification (Marsh

    score 1) Panel C shows total villous atrophy typical of celiac disease, 10x magnification (Marsh score 3).

    A B C

    Bishop_Ch18_265 276.indd 271Bishop_Ch18_265-276.indd 271 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    8/12

  • 8/12/2019 Celiac Dz - Peds

    9/12

    CHAPTER 18 Celiac Disease 273

    (FALCPA), which took effect on January 1, 2006,

    requires food labels to clearly identify eight common

    food allergens: wheat, eggs, fish, milk, peanuts, shell-

    fish, soybeans, and tree nuts. This law does not, how-

    ever, address the use of barley (malt), rye, or oats. FAL-CPA also requires the U.S. Food and Drug

    Administration to develop rules for the use of the term

    gluten-free on product labels. The final ruling is still

    pending, although the FDA proposal is available from

    the federal register at http://www.cfsan.fda.gov/%7Elrd/

    fr070123.html. The parameter for a naturally gluten-

    free product is 20 ppm per serving, and for a food ren-

    dered gluten-free is 200 ppm.

    NEW THERAPIES UNDER

    INVESTIGATION

    While the only current treatment for CD is lifelong adher-ence to a GFD, several alternative treatments are currently

    being investigated as possible adjunctive therapies.

    Bacterial Prolyl-endopeptidases

    One of the properties of gluten that makes it immuno-

    genic is the ability to reach the lamina propria in large,

    antigenically intact peptide fragments. This is due to

    lack of native peptidases capable of cleavage of peptide

    Food and Ingredient List for the Gluten-free Diet

    Food Item Foods/Ingredients Allowed Foods/Ingredients to Question Foods/Ingredients Not Allowed

    Pastas, potatoes,

    and other

    starches

    Pastas made from allowed grains, rice

    including white brown and wild, corn

    tacos and corn tortillas, potatoes,

    saifun (bean threads), rice noodles

    Commercial rice and pasta mixes,

    French fries, fried restaurant

    foods (gluten-contaminated

    grease), polenta

    All pastas made from grains not

    allowed, couscous, tabbouleh,

    soba noodles

    Fruits and fruit

    products

    Fresh fruit, pure fruit juices Thickened or prepared fruits and

    pie fillings, dried fruit mixes, jelly

    and jam

    Beverages Tea (black, green, and white), coffee

    (plain), pure fruit juices, cider

    Soy and rice beverages, instant

    tea and coffee, flavored tea and

    coffee, hot cocoa, hot chocolate,

    soda, sports drinks, nutritional

    supplements

    Malted beverages, ground

    coffee with added grains

    Vinegar Vinegars (apple cider, rice, wine,

    and balsamic)

    Flavored vinegars Malt vinegar

    Condiments Salt, pepper (black, white, and red), pure

    herbs, pure spices, pure flavoring

    extracts, wheat-free soy sauce, cream

    of tartar, pickles, honey

    Ground spices, seasoning and

    spice mixes, gravy cubes and

    mixes, bouillon cubes and

    powder, ketchup, mustard,

    mustard powder, Worcestershire

    sauce, salad dressings, soy

    sauce, teriyaki sauce, relish

    Most soy sauces contain wheat

    Miscellaneous Seaweed (algin, algae, alginate),

    alfalfa, aspic

    Yeast, yeast flakes, bicarbonate of

    soda, baking soda, baking

    powder, powdered sugar,

    molasses, rice paper,

    confectioner sugar

    Communion wafers,

    brewers yeast

    Non-food Lotions, creams, cosmetics, lip

    gloss, lip balm, sunscreen,

    toothpaste, mouthwash,

    products used in dental offices,

    medications: prescription

    and over the counter (many

    contain gluten), laxatives,

    vitamins, stamps, envelopes,

    and gummed labels, Play Doh,

    paper mache

    Table 187. (Continued)

    Bishop_Ch18_265 276.indd 273Bishop_Ch18_265-276.indd 273 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    10/12

    274 Section 3: Disorders of the Stomach and Intestine

    bonds of proline residues. Currently being investigated

    is the use of exogenous bacterial derived prolyl-

    endopeptidases to cleave these peptides into smaller,

    less immunogenic fragments prior to reaching the

    lamina propria. There are published data on in vitro

    and in vivo animal models, and ex vivo human T cells,

    with promising results. Controlled studies in CD

    patients using these enzymes are yet to be

    published.2329

    Inhibition of Intestinal tTG

    Early work is underway at developing a site-specific

    inhibitor of intestinal tTG, to prevent the local deami-dation and binding enhancement of gluten peptides.

    There are potential side effects with such an inhibitor,

    such as impaired wound healing and perturbation of

    the extracellular matrix.30,31

    DQ2/DQ8-binding Peptides

    Theoretically, a compound can be formulated that will

    occupy the binding groove on HLA-DQ2 or -DQ8 and

    prevent gluten peptide antigen presentation. This would

    effectively block the immunogenic reaction. To our

    knowledge, no such compound has yet been formu-

    lated.30

    T-cell Silencing

    Another theoretic therapy would be to stimulate

    apoptosis in gluten-specific T cells, or induce toler-

    ance in these T cells by targeting tolerogenic dendritic

    cells. These therapies again are still in theoretic

    stages.30

    Cytokine Therapy

    Several trials of anti-inflammatory cytokine therapy are

    underway for other chronic diseases such as RA and

    IBD. IL-15 antagonists have been developed by several

    pharmaceutical companies for treatment of RA, but

    have therapeutic potential for CD as well. IFN- antago-nists have been developed and are in phase II testing for

    reduction of inflammation in IBD, but are being recog-

    nized for potential in CD treatment as well. IL-10 has

    been tested in both refractory CD and IBD for its poten-

    tial ability to shift away from TH1 immune reaction.

    Unfortunately, neither trial has so far showed any

    success.30,3234

    Genetically Modified Wheat

    Work is currently underway to evaluate the feasibility of

    engineering and cultivating a modified, gluten-free

    strain of wheat that would lack immunogenic peptides.

    Barriers to this would include cost, palatability, and

    industrial quality of the flour derived from such a strain.

    This project is in its early stages, and would not be avail-

    able to the mass market for many years, if ever.

    Other Possibilities

    Monoclonal antibodies to the adhesion molecule integ-

    rin a4 are currently being used for multiple sclerosis and

    are also in phase II trials for IBD. These compounds

    may have potential for CD by prevention of T-cell

    migration into the lamina propria. Zonulin antagonists

    are being looked at for prevention of gluten-induced

    intestinal hyper-permeability. The concept of usingNGK2D antagonists to prevent the phenotypic conver-

    sion of CD4 T cells to cytotoxic T cells in the intestinal

    epithelium has also been raised.30

    PREVENTION

    With a more thorough understanding of the pathogen-

    esis of CD, more emphasis may start being placed on the

    prevention of development rather than treatment.

    In a recent meta-analysis of breastfeeding prac-

    tices and effect on CD, the authors concluded that dura-

    tion of breastfeeding and breastfeeding during introduc-

    tion of grains reduced future risk of CD development.35,36This is not uniformly accepted, however, and some inves-

    tigators feel that breastfeeding may delay or mask, but

    not prevent the development of CD.37The mechanism is

    speculative, but involvement of probiotic bacteria is

    thought to be important for development of tolerance to

    food proteins and maintenance of intact intestinal

    epithelial barrier in infancy. It is also speculated that

    Celiac Disease and Gluten Sensitivity Resources

    www.celiac.com: celiac and gluten-free information

    www.celiac.org: Celiac Disease Foundation

    www.gluten.net: Gluten Intolerance Group of North America

    www.celiac.nih.gov: Celiac Disease Awareness Campaign

    www.celiaccentral.org: National Foundation for Celiac

    Awareness

    www.celiachealth.org: Childrens Digestive Health and

    Nutrition Foundation

    www.americanceliac.org: American Celiac Disease Alliance

    www.eatright.org: American Dietetic Association

    www.csaceliacs.org: Celiac Sprue Association

    www.cdhnf.org: Childrens Digestive Health and Nutrition

    Foundation

    Table 188.

    Bishop_Ch18_265 276.indd 274Bishop_Ch18_265-276.indd 274 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    11/12

    CHAPTER 18 Celiac Disease 275

    probiotic therapy in at-risk individuals may help to pro-

    mote tolerance and prevent CD.

    Timing of introduction of gluten and quantity of

    gluten in the diet are other controversial topics in pre-

    vention of CD. Early (prior to 3 months of age) and late

    (after 7 months) introduction of gluten were associated

    with increased risk of CD in one prospective study.37

    Several recent papers documented a four-fold rise in

    incidence of CD in Sweden in children less than 2 years

    old, after a significant increase in gluten intake in this

    population. A later decrease in gluten consumption cor-

    responded to a drop in CD.38

    Prevention of infectious triggers such as rotavirus

    and the subsequent effect on CD risk is another area

    currently being researched. If the increased permeabil-

    ity and inflammation corresponding with infection can

    be avoided in at-risk individuals, perhaps breakdown of

    tolerance will not occur.36,39,40

    REFERENCES

    1. Hill ID, Dirks MH, Liptak GS, et al. Guideline for the diag-

    nosis and treatment of celiac disease in children: recom-

    mendations of the North American Society for Pediatric

    Gastroenterology, Hepatology and Nutrition. J Pediatr

    Gastroenterol Nutr. 2005;40:119.

    2. Megiorni F, Mora B, Bonamico M, et al. HLA-DQ and

    susceptibility to celiac disease: evidence for gender differ-

    ences and parent-of-origin effects. Am J Gastroenterol.

    2008;103:9971003.

    3. Cataldo F, Montalto G. Celiac disease in the developing

    countries: a new and challenging public health problem.

    World J Gastroenterol. 2007;13:21532159.

    4. Bonamico M, Ferri M, Mariani P, et al. Serologic and

    genetic markers of celiac disease: a sequential study in the

    screening of first degree relatives. J Pediatr Gastroenterol

    Nutr. 2006;42:150154.

    5. Wolters VM, Wijmenga C. Genetic background of celiac

    disease and its clinical implications. Am J Gastroenterol.

    2008;103:190195.

    6. Nistico L, Fagnani C, Coto I, et al. Concordance, disease

    progression, and heritability of coeliac disease in Italian

    twins. Gut. 2006;55:803808.

    7. Kagnoff MF. Celiac disease: pathogenesis of a model

    immunogenetic disease.J Clin Invest. 2007;117:4149.

    8. Sollid LM. Hunting for celiac disease genes. Gastroenterol-

    ogy. 2008;134:869871.

    9. Catassi C, Fasano A. Celiac disease. Curr Opin Gastroen-

    terol. 2008;24:687691.

    10. Kemppainen T, Janatuinen E, Holm K, et al. No observedlocal immunological response at cell level after five years

    of oats in adult coeliac disease. Scand J Gastroenterol.

    2007;42:5459.

    11. National Institutes of Health Consensus Development

    Conference Statement on Celiac Disease, June 2830,

    2004. Gastroenterology. 2005;128:S1S9.

    12. NIH Consensus Development Conference on Celiac Dis-

    ease. NIH Consens State Sci Statements. 2004;21:123.

    13. Fasano A. Clinical presentation of celiac disease in the

    pediatric population. Gastroenterology. 2005;128:

    S68S73.

    14. Troncone R, Auricchio R, Granata V. Issues related to

    gluten-free diet in coeliac disease. Curr Opin Clin Nutr

    Metab Care. 2008;11:329333.

    15. Rostom A, Murray JA, Kagnoff MF. American Gastroen-terological Association (AGA) Institute technical review

    on the diagnosis and management of celiac disease. Gas-

    troenterology. 2006;131:19812002.

    16. Halfdanarson TR, Litzow MR, Murray JA. Hematologic

    manifestations of celiac disease. Blood. 2007;109:412421.

    17. Collin P, Reunala T. Recognition and management of the

    cutaneous manifestations of celiac disease: a guide for

    dermatologists. Am J Clin Dermatol. 2003;4:1320.

    18. Zone JJ. Skin manifestations of celiac disease. Gastroenter-

    ology. 2005;128:S87S91.

    19. Zanchi C, Di Leo G, Ronfani L, Martelossi S, Not T, Ven-

    tura A. Bone metabolism in celiac disease. J Pediatr.

    2008;153:262265.

    20. Fasano A, Araya M, Bhatnagar S, et al. Federation of Inter-

    national Societies of Pediatric Gastroenterology, Hepatol-

    ogy, and Nutrition consensus report on celiac disease.J Pediatr Gastroenterol Nutr. 2008;47:214219.

    21. AGA Institute medical position statement on the diagno-

    sis and management of celiac disease. Gastroenterology.

    2006;131:19771980.

    22. Catassi C, Fabiani E, Iacono G, et al. A prospective, dou-

    ble-blind, placebo-controlled trial to establish a safe glu-

    ten threshold for patients with celiac disease. Am J Clin

    Nutr. 2007;85:160166.

    23. Cerf-Bensussan N, Matysiak-Budnik T, Cellier C, Heyman

    M. Oral proteases: a new approach to managing coeliac

    disease. Gut. 2007;56:157160.

    24. Matysiak-Budnik T, Candalh C, Cellier C, et al. Limited

    efficiency of prolyl-endopeptidase in the detoxification of

    gliadin peptides in celiac disease. Gastroenterology.

    2005;129:786796.25. Gass J, Bethune MT, Siegel M, Spencer A, Khosla C. Com-

    bination enzyme therapy for gastric digestion of dietary

    gluten in patients with celiac sprue. Gastroenterology.

    2007;133:472480.

    26. Mitea C, Havenaar R, Drijfhout JW, Edens L, Dekking L,

    Koning F. Efficient degradation of gluten by a prolyl endo-

    protease in a gastrointestinal model: implications for coe-

    liac disease. Gut. 2008;57:2532.

    27. Stepniak D, Spaenij-Dekking L, Mitea C, et al. Highly effi-

    cient gluten degradation with a newly identified prolyl

    endoprotease: implications for celiac disease. Am J Physiol

    Gastrointest Liver Physiol. 2006;291:G621G629.

    28. Marti T, Molberg O, Li Q, Gray GM, Khosla C, Sollid LM.

    Prolyl endopeptidase-mediated destruction of T cell

    epitopes in whole gluten: chemical and immunologicalcharacterization.J Pharmacol Exp Ther. 2005;312:1926.

    29. Shan L, Marti T, Sollid LM, Gray GM, Khosla C. Com-

    parative biochemical analysis of three bacterial prolyl

    endopeptidases: implications for coeliac sprue. Biochem J.

    2004;383:311318.

    30. Sollid LM, Khosla C. Future therapeutic options for celiac

    disease. Nat Clin Pract Gastroenterol Hepatol. 2005;2:

    140147.

    Bishop_Ch18_265 276.indd 275Bishop_Ch18_265-276.indd 275 4/6/10 2:34/6/10 2:3

  • 8/12/2019 Celiac Dz - Peds

    12/12

    276 Section 3: Disorders of the Stomach and Intestine

    31. Schuppan D, Dieterich W. A molecular warhead and its

    target: tissue transglutaminase and celiac sprue. Chem

    Biol. 2003;10:199201.

    32. Gianfrani C, Auricchio S, Troncone R. Possible drug tar-

    gets for celiac disease. Expert Opin Ther Targets.

    2006;10:601611.

    33. Di Sabatino A, Ciccocioppo R, Cupelli F, et al. Epitheliumderived interleukin 15 regulates intraepithelial lymphocyte

    Th1 cytokine production, cytotoxicity, and survival in

    coeliac disease. Gut. 2006;55:469477.

    34. Salvati VM, Mazzarella G, Gianfrani C, et al. Recombinant

    human interleukin 10 suppresses gliadin dependent T cell

    activation in ex vivo cultured coeliac intestinal mucosa.

    Gut. 2005;54:4653.

    35. Akobeng AK, Heller RF. Assessing the population impact

    of low rates of breast feeding on asthma, coeliac disease

    and obesity: the use of a new statistical method. Arch Dis

    Child. 2007;92:483485.

    36. Troncone R, Auricchio S. Rotavirus and celiac disease:

    clues to the pathogenesis and perspectives on prevention.

    J Pediatr Gastroenterol Nutr. 2007;44:527528.

    37. Norris JM, Barriga K, Hoffenberg EJ, et al. Risk of celiac

    disease autoimmunity and timing of gluten introductionin the diet of infants at increased risk of disease. JAMA.

    2005;293:23432351.

    38. Ivarsson A. The Swedish epidemic of coeliac disease

    explored using an epidemiological approachsome les-

    sons to be learnt. Best Pract Res Clin Gastroenterol.

    2005;19:425440.

    39. Pavone P, Nicolini E, Taibi R, Ruggieri M. Rotavirus and

    celiac disease. Am J Gastroenterol. 2007;102:1831.

    40. Stene LC, Honeyman MC, Hoffenberg EJ, et al. Rotavirusinfection frequency and risk of celiac disease autoimmu-

    nity in early childhood: a longitudinal study. Am J Gastro-

    enterol. 2006;101:23332340.

    41. Sollid LM. Coeliac disease: dissecting a complex inflam-

    matory disorder. Nat Rev Immunol. 2002;2:647655.

    42. Hoffenberg EJ, Bao F, Eisenbarth GS, et al. Transglutami-

    nase antibodies in children with a genetic risk for celiac

    disease.J Pediatr. 2000;137:356360.

    43. Dubois PC, van Heel DA. Translational mini-review series

    on the immunogenetics of gut disease: immunogenetics

    of coeliac disease. Clin Exp Immunol. 2008;153:162173.

    44. Hunt KA, Zhernakova A, Turner G, et al. Newly identified

    genetic risk variants for celiac disease related to the

    immune response. Nat Genet. 2008;40:395402.

    45. Smyth DJ, Plagnol V, Walker NM, et al. Shared and dis-

    tinct genetic variants in type 1 diabetes and celiac disease.N Engl J Med. 2008;359:27672777.