Nothing Special   »   [go: up one dir, main page]

Enfermeda Celiaca

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Diagnosis and management of adult coeliac disease:

guidelines from the British Society


of Gastroenterology
Jonas F Ludvigsson,
1,2
Julio C Bai,
3
Federico Biagi,
4
Timothy R Card,
5
Carolina Ciacci,
6
Paul J Ciclitira,
7
Peter H R Green,
8
Marios Hadjivassiliou,
9
Anne Holdoway,
10
David A van Heel,
11
Katri Kaukinen,
12,13,14
Daniel A Lefer,
15
Jonathan N Leonard,
16
Knut E A Lundin,
17
Norma McGough,
18
Mike Davidson,
19
Joseph A Murray,
20
Gillian L Swift,
21
Marjorie M Walker,
22
Fabiana Zingone,
23
David S Sanders,
24
Authors of the BSG Coeliac Disease Guidelines Development Group
For numbered afliations see
end of article.
Correspondence to
David S Sanders,
Gastroenterology and Liver
Unit, Royal Hallamshire
Hospital & University of
Shefeld, Shefeld S10 2JF,
UK; david.sanders@sth.nhs.uk
Received 12 December 2013
Revised 23 April 2014
Accepted 25 April 2014
To cite: Ludvigsson JF,
Bai JC, Biagi F, et al. Gut
Published Online First:
[ please include Day Month
Year] doi:10.1136/gutjnl-
2013-306578
ABSTRACT
A multidisciplinary panel of 18 physicians and 3 non-
physicians from eight countries (Sweden, UK, Argentina,
Australia, Italy, Finland, Norway and the USA) reviewed
the literature on diagnosis and management of adult
coeliac disease (CD). This paper presents the
recommendations of the British Society of
Gastroenterology. Areas of controversies were explored
through phone meetings and web surveys. Nine working
groups examined the following areas of CD diagnosis
and management: classication of CD; genetics and
immunology; diagnostics; serology and endoscopy;
follow-up; gluten-free diet; refractory CD and
malignancies; quality of life; novel treatments; patient
support; and screening for CD.
INTRODUCTION
Objective
The aim was to create updated guidelines for the
management of adult coeliac disease (CD), but
non-coeliac gluten sensitivity (NCGS) was not
considered.
Development of the guidelines
The British Society of Gastroenterology (BSG)
guidelines on the management of adult CD were
originally published in 1996. Recently the
European Society for Paediatric Gastroenterology,
Hepatology and Nutrition (ESPGHAN) published
updated guidelines for paediatric CD,
1
but inter-
national guidelines for adult CD are scarce
25
since
the NIH consensus
6
on CD in 2005 (despite more
than 4000 PubMed publications about CD in the
last 8 years). As a result, the Clinical Services and
Standards Committee of the BSG commissioned
these guidelines, subject to rigorous peer review
and based on a comprehensive review of the recent
literature, including data from any available rando-
mised controlled trials, systematic reviews,
meta-analyses, cohort studies, prospective and
retrospective studies.
A multidisciplinary panel of 18 physicians from
eight countries (Sweden, UK, Argentina, Australia,
Italy, Finland, Norway and the USA), a dietitian
and a representative and a patient advocate from
Coeliac UK reviewed the literature on the manage-
ment of CD. These individuals were involved in
the original stakeholder meetings and with revision
of the manuscript.
Intent and levels of evidence
All aspects of the contemporary diagnosis and man-
agement of patients with adult CD were consid-
ered. PubMed literature was searched from 1900 to
2012 to obtain evidence for these guidelines. Also
there was input from all authors who have consid-
erable expertise and experience in diagnosis and
management of CD. The panel of international
experts previously collaborated in the publication
of denitions of CD
7
and were invited by the BSG
through coauthor DSS. Our task force contained
representatives from the clinical disciplines gastro-
enterology, paediatrics, histopathology, neurology,
dermatology, genetics and immunology.
The current literature of review papers was
examined, focusing on 10 reviews
817
to explore
gaps in current reviews on CD. Nine working sub-
groups were then formed that examined the follow-
ing areas of CD management: classication of CD:
FB, MH, DSS, CC; genetics and immunology:
KEAL, DaVH, PJC; diagnostic criteria, serology
and endoscopy in the investigation of CD: MMW,
JAM, FB, PHRG, JFL, KEAL; follow-up: DAL,
PHRG, JCB, JFL; gluten-free diet (GFD): PJC, KK,
CC, GLS; refractory CD (RCD) and complications:
FZ, FB, DAL, PHRG; quality of life (QoL): GLS,
JCB, TRC, FZ; novel therapy: JCB, KEAL; screen-
ing for CD: TRC, KK, JAM, JFL. The working
groups wrote the sections, which were subsequently
internally reviewed. Each nal fully written and
Recommendations
Diagnosis of CD requires duodenal biopsy when
the patient is on a gluten-containing diet and
for the vast majority of adult patients also
positive serology. (Grade B)
Biopsy remains essential for the diagnosis of
adult CD and cannot be replaced by serology.
Follow-up should aim at strict adherence to a
gluten-free diet. (Grade B)
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 1
Guidelines
Gut Online First, published on June 10, 2014 as 10.1136/gutjnl-2013-306578
Copyright Article author (or their employer) 2014. Produced by BMJ Publishing Group Ltd (& BSG) under licence.
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
referenced section was then released to all group members for
review by teleconference and email correspondence. Thereafter
JFL created the rst draft of the guidelines by amalgamating all
documents. All authors then helped revise this draft until nal
document consensus was reached.
Between January 2012 and February 2013, six web surveys
were performed using the web site survey console (http://www.
surveyconsole.com) to explore issues including coeliac topics of
controversy; the role of endoscopy; the role of histopathology
and serology in the diagnosis of CD; and follow-up of patients,
including the use of follow-up biopsy. The web surveys were for
the coauthors/Guidelines Development Group (GDG) members.
Survey results were then discussed at teleconference and used to
inform the direction of recommendations and outline areas
where the GDG were not concordant. Disagreements were
solved through discussion.
Studies used as a basis for these guidelines are graded accord-
ing to the quality of evidence using the Oxford Centre for
Evidence-based Medicine levels of evidence.
18
Strength of recommendations
A. Directly based on category I evidence, for example, from
systematic reviews and randomised controlled trials. This is
the strongest recommendation of the four grades listed.
B. Directly based on category II or III evidence or extrapolated
recommendation from category I evidence. This includes
evidence from controlled non-randomised studies or time
series; or indirect evidence from systematic reviews or ran-
domised controlled trials.
C. Directly based on category IV evidence or extrapolated rec-
ommendation from category II or III evidence. This also
includes evidence from non-experimental studies such as
cohort studies or casecontrol studies.
D. Directly based on category V evidence or inconsistent or
inconclusive studies of any level. This includes evidence
from expert committees and respected authorities.
BACKGROUND
CD is an immune-mediated small intestinal enteropathy that is
triggered by exposure to dietary gluten in genetically predis-
posed individuals.
7
Samuel Gee is credited with the rst clinical
description of CD in 1887, although Aretaeus of Cappadocia
may have described the disease in the rst century AD. In the
1930s and 1940s, Dicke demonstrated (then published in
1953
19
) that a wheat-free diet was the key to management. The
discovery of antigliadin antibodies in 1961 was the rst non-
invasive serological marker for CD.
20
Until the 1980s, CD was considered a rare disease affecting
mainly children (exceptions occurred
21
), but subsequently has
been shown to occur at any age.
22
Although the prevalence seems
to vary considerably (from <0.25%
23
to >1%
24
), a large-scale
screening study in subjects from Finland, Italy, the UK and
Germany found a prevalence of CD of around 1%,
2527
with a
recent US study showing a prevalence of 0.71%.
28
CD is more
frequently diagnosed in women than in men with a ratio
between 1.5 and 2,
29
but this gender imbalance may vanish
with age and has been absent in some screening studies.
30
Traditionally patients with CD presented with malabsorption
dominated by diarrhoea, steatorrhoea, weight loss or failure to
thrive (classical CD),
7
but over time the proportion of newly
diagnosed patients with malabsorptive symptoms has
decreased,
31
and non-classical CD
7
and even asymptomatic
CD have gained prominence. Newly diagnosed patients with
CD can present with a wide range of symptoms and signs,
including anaemia,
32
vague abdominal symptoms (often similar
to irritable bowel syndrome (IBS)
33
), neuropathy,
34 35
ataxia,
36
depression,
37
short stature,
38
osteomalacia and osteoporosis,
39
liver disease,
40
adverse pregnancy outcomes
41
and lymphoma.
42
Asymptomatic patients are typically diagnosed through screen-
ing. Screening may be initiated because the individual has a
CD-associated disorder or has symptoms and is a rst-degree
relative to a patient with CD.
The diversity of the clinical presentation of CD emphasises
the need for robust diagnostic criteria and careful disease
work-up. It is therefore natural that over the years, several
efforts have been made to dene CD,
1 7 4345
and how it
should be managed.
816
While all these reviews discussed critical
aspects of diagnostics, including the small intestinal biopsy and
serology, there are scant reports discussing the genetic and
immunological background,
814
the role of human leucocyte
antigen (HLA) testing,
11 12 15 16 46
and even fewer have dis-
cussed issues such as QoL
11 15
and patient support.
15
During our previous review on denitions of CD and related
concepts,
7
we realised that there was a need for a consensus
paper on modern management and diagnosis of CD. In this
project, participants of the so-called Oslo group who rst con-
vened at the 2011 meeting on CD in Oslo
7
collaborated with
representatives of the BSG to write guidelines for the manage-
ment of CD in adults. These guidelines will enable physicians,
dietitian and other healthcare personnel to provide better care
for their patients.
GENETICS, IMMUNOLOGY AND TRIGGER FACTORS
Environmental factors are important in CD and ingestion of
gluten is a prerequisite for the development of CD. Children
breastfed at and beyond gluten introduction may be at lower
risk of developing CD in childhood, although research is not
consistent.
47
Conversely, large amounts of gluten or gluten
exposure without ongoing breastfeeding may increase the risk of
future CD.
48 49
Gastrointestinal infections, drugs, interferon
and surgery have also been implicated as trigger factors.
47 50 51
The factors leading to the breakdown of tolerance to gluten are
not known, but local pro-inammatory changes are of para-
mount importance.
52
A high prevalence (10%) among rst-degree relatives of
patients with CD
53
and a greater concordance rate in monozy-
gotic twins (75%)
54
than in dizygotic twins indicates a strong
genetic component.
CD shows a very strong association with a particular HLA
variant termed HLA-DQ2.5.
55
This molecule is encoded by the
DQA1*05:01 and DQB1*02:01 genes in cis conguration on
the DR3 haplotype. Thus most patients are DR3, DQ2 positive.
A smaller subset of patients with CD express a very similar
DQ2.5 molecule encoded by a combination of DR5 and DR7
positive haplotypes, in this situation by the genes in trans pos-
ition. While the proportion of individuals with CD who are
DQ2.5 positive varies in different geographical areas it is gener-
ally 90%. The majority of the remaining patients are
HLA-DQ8 positive (DR4, DQ8 haplotype).
56
Genome wide association studies have so far identied 40 loci
outside of HLA with genes predisposing to or protecting against
CD.
57 58
Most of these genes have immunological functions and
are related to B-cell and T-cell functions. Each of the non-HLA
genes contributes little to genetic risk. With a population preva-
lence of 1% and a heritability of 50%, the 39 non-HLA loci
account for 14% of the genetic variance whereas HLA in com-
parison accounts for 40%.
57
2 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
The most striking morphological features of the active coeliac
lesion are the inltration in the epithelium of T-cell receptor /
and / expressing CD8 positive T cells, natural killer cell like T
cells, and the dense population in the lamina propria of plasma
cells and activated antigen presenting cells.
59 60
Although
numerically not dominant, CD4 gluten reactive T cells (T-cell
receptor / expressing)
61 62
are crucial in the pathogenesis of
CD and are not found in people without CD.
6365
The CD4, gluten-specic T cells in the lamina propria invari-
ably recognise gluten presented by the disease-associated
HLA-DQ2 and HLA-DQ8 molecules
6165
and recognise gliadin
peptides that have been modied by tissue transglutaminase 2
(TG2). This modication (glutamine to glutamic acid deamida-
tion process) introduces negative charges.
66
These modications
produce peptides that bind with much higher afnity to the
HLA-DQ2 and HLA-DQ8 molecules.
67
Patients with CD who
are untreated typically have high titres of antibodies against the
endomysium antigen (EMA test), also shown to be the extracel-
lular enzyme tissue TG2.
68
On exposure to gluten, plasma cells
produce antibodies to tissue transglutaminase (IgA-TG2)
69
and
deamidated gliadin peptides (IgA-DGP
70
and IgG-DGP
71
).
Untreated and treated CD are characterised by an increase in
/ intraepithelial lymphocytes (IELs), although in treated
disease, the IEL count is close to that in healthy individuals.
72
DIAGNOSTICS
Diagnosis of CD is by serology and duodenal biopsy, ideally
with the patient on a normal, that is, gluten-containing diet.
Biopsy remains essential for the diagnosis of adult CD and
cannot be replaced by serology. Exceptions are patients with
coagulation disorders and pregnant women, in whom biopsy
may not be feasible or should be postponed until postpartum.
To state denite diagnosis of CD, villous atrophy is required.
However, lesser degrees of damage (25 IELs but no villous
atrophy) combined with positive serology (IgA-EMA, tissue
transglutaminase (TTG) or IgG-DGP) may also represent CD
(probable CD), and in these circumstances a trial with GFD
may be considered to further support the diagnosis of CD. HLA
status may also aid diagnosis. Differential diagnoses of lympho-
cytic duodenosis should be ruled out if there is no response to
GFD (see table 2).
The diagnosis of CD is readily established in those who,
while consuming a gluten-containing diet, have positive serology
and a duodenal biopsy with obvious coeliac histology (increased
intraepithelial lymphocytosis, crypt hyperplasia and villous
atrophy; table 1). These patients can immediately initiate a GFD
with condence.
CD can also be suspected in patients with mild gastrointes-
tinal symptoms, associated conditions or those at genetic risk.
3
Such patients should be investigated initially with serology and
if this is positive (or if there is still a high index of suspicion as
among symptomatic rst-degree relatives
73
), then undergo
upper endoscopy and duodenal biopsies.
However, in some cases the diagnosis of CD may not be
straightforward, for example, patients are already on a GFD and
therefore antibodies are negative, biopsies were not oriented
correctly (this could lead to false-negative or false-positive
villous atrophy) or show solely intraepithelial lymphocytosis
(lymphocytic duodenosis)
74
without architectural changes. In
these situations, the patient needs to be maintained on a gluten-
containing diet and further evaluated with additional testing
and, if necessary, referred to a centre or clinician with a specic
interest in CD.
The diagnosis of CD is illustrated in table 1.
Serology in CD diagnosis
Serological detection depends on the presence of specic endo-
mysial antibodies (EMAs, also called AEAs), IgA anti-tissue
Table 1 An algorithm for the diagnosis of coeliac disease
IgA-TG2 Total IgA IgG-TG2- IgG-DGP EMA HLA
Biopsy
showing CD type
histology* Coeliac status
Patient on a gluten-containing diet, IgA-TG2 or IgA-DGP, or IgG-DGP, EMA positive and the
biopsy shows villous atrophy
Coeliac
For patients for whom the above criteria are not fulfilled, please see below
+ Normal or ND ND ND ND ND + Coeliac if VA, otherwise probably coeliac
+ ND ND ND + + + Coeliac if VA, otherwise probably coeliac
+ ND ND ND + + Potential celiac
+ (usually
low titre)
ND ND ND Not coeliac
Normal + Not coeliac
Normal Not coeliac
Normal + Unclear. Check serology when the patient is on a gluten-containing diet.
Evaluate response to gluten-free diet, and consider other causes of VA
Normal + + Unclear. Check serology when the patient is on a gluten-containing diet.
Evaluate response to gluten-free diet, and consider other causes of VA
Low/absent + + + + IgA deficient and celiac if biopsy shows VA.
Otherwise IgA deficient and probable coeliac
Low/absent IgA deficient, not coeliac
Low/absent + + ?post infectious diarrhoea
Combined immune deficiency syndrome
*Biopsy showing CD type histology = villous atrophy, crypt hyperplasia but must have intraepithelial lymphocytosis, IELs25/100 enterocytes.
A duodenal biopsy may be negative when the lesion is only present in the jejunum.
And the patient does not respond to a gluten-free diet.
CD, coeliac disease; DGP, deamidated gliadin peptide; EMA, endomysium antigen; HLA, human leucocyte antigen; IEL, intraepithelial lymphocyte; ND, not done;
TG2, transglutaminase 2; VA, villous atrophy (Marsh 3).
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 3
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
transglutaminase antibodies (IgA-TG2, also called a-TTG, TTA)
and/or deamidated antigliadin antibodies (DGP, either IgA or
IgG isotype).
7
IgG-TG2 is primarily useful in patients with
known IgA deciency.
75 76
There is continuing debate on the sole use of non-invasive
tests to diagnose CD. Recently ESPGHAN proposed new guide-
lines for the diagnosis of CD in children. It suggests that in
symptomatic paediatric patients
1
in whom the IgA-TG2 level
exceeds 10 times the upper limit of normal, EMA antibodies are
positive on a separately taken blood sample, and HLA-DQ2 or
HLA-DQ8 are positive, then biopsies do not need to be per-
formed to conrm the diagnosis of CD.
1
In adults, this strategy
has also been proposed
77
; however, there are very strong argu-
ments for retaining the biopsy as gold standard for the diagnosis
of CD. A recent study from the (UK) National External Quality
Assessment Service centre states that not all commercial
IgA-TG2 kits are reliable and the ESPGHAN guidelines are
therefore not translatable for use in all centres and should not
be used in the UK.
78
Also, 2% of patients with CD are IgA de-
cient (0.2% of the population in general) and as usual serology
tests for IgA-TG2 and EMA are IgA based, this may lead to false
negatives and a reduction in test sensitivity. If patients are
known to be IgA decient, IgG-TG2 or IgG-DGP antibodies can
be used, or alternatively, such patients should proceed directly
to biopsy.
79
A combination of immunoassays offers the best sensitivity if
either a positive IgA-TG2 or IgG-DGP is considered a positive
detection test. The combination of IgG-DGP and IgA-TG2 is
particularly useful as an addition to detection of patients with
CD who are IgA decient, IgG-DGP was able to detect a few
more IgA-sufcient patients who were missed by IgA-TG2
alone.
76
There are now several point-of-care tests commercially avail-
able, which allow both immediacy and the ability to use them in
a physician ofce/primary care setting. However, there is a rela-
tive paucity of data on the sensitivity and specicity of such tests
by comparison to the gold standard of duodenal biopsy.
80
There
is concern regarding the use of these tests as patients may start
on a GFD without a rm diagnosiswhich includes biopsy
and further studies should be performed before considering the
use of these in everyday practice. One study utilising community
health nurses demonstrated a lower than expected sensitivity
for CD.
81
Endoscopy in seronegative individuals
The prevalence of seronegative CD is 622% of all diagnosed
cases.
71 82 83 84
One study also found a high degree of variabil-
ity in EMA values for sensitivity between laboratories,
85
and
upper endoscopy is generally well tolerated and safe.
86
Individuals of white European, Middle Eastern, North African
or North Indian origin who undergo upper endoscopy for
anaemia, weight loss or diarrhoea should therefore have duo-
denal biopsies performed, irrespective of whether they have had
serology for CD. These features may well indicate that CD or
an alternative mucosal cause of malabsorption is present.
87
In
Table 2 Histological mimics of CD in seronegative patientsconditions to be considered for investigation in an appropriate clinical context
Duodenal histology: normal architecture and increased IELs (25/100 enterocytes) or villous atrophyincreased IELs (25/100 enterocytes)
Immune disorders Common variable immunodeficiency syndrome
Glomerulonephritis
Hypogammaglobulaemia
IgA deficiency
Autoimmune disease
These patients may have concurrent CD,
check serology and HLA status if appropriate*
Autoimmune enteropathy (adults and children)
Graves disease*
Haemolytic anaemia
Hashimotos thyroiditis*
Multiple sclerosis
Psoriasis
Rheumatoid arthritis
Sjgrens syndrome*
Systemic lupus erythematosus
Thymoma-associated autoimmune enteropathy
Type I diabetes mellitus*
Hypersensitivity/non-gluten protein intolerance Non-coeliac gluten sensitivity
Protein intolerance (cows milk, soy,
eggs, peanuts, cereals)
Infection AIDS
Cryptosporidium
Giardiasis
Helicobacter pylori gastritis
Postinfectious diarrhoea
Small intestinal bacterial overgrowth
Tropical sprue
Tuberculosis (including atypical TB)
Viral
Whipples disease (for example, HIV)
Drugs Chemotherapy
Non-steroidal anti-inflammatory drugs
Olmesartan
Mycophenolate mofetil
Neoplasia Enteropathy-associated T-cell lymphoma
Immunoproliferative small intestinal disease
Refractory CD type 2
CD 4 T-cell proliferation
Other Abetalipoproteinaemia
Collagenous colitis
Collagenous duodenitis
Crohns disease
Eosinophilic gastroenteritis
Glycogen storage disease
Microscopic colitis
Radiation enteritis
Small bowel ischaemia
*For definition please see text and the Oslo definitions.
7
Common.
CD, coeliac disease; HLA, human leucocyte antigen; IEL, intraepithelial lymphocyte.
4 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
fact, it has been suggested that duodenal biopsy should be con-
sidered in any individual undergoing endoscopy, because CD is
common and has many varied clinical manifestations, including
reux, a common indication for endoscopy.
88
Recommendation
In individuals undergoing an upper endoscopy in whom
laboratory tests or symptoms or endoscopic features suggest
CD, duodenal biopsy should be considered. (Grade C)
Role of HLA in the diagnosis of CD
CD is associated with specic HLA types in virtually all popula-
tions in which this has been tested, and is associated with the
carriage of the gene pairs that encode DQ2.5 and DQ8.
46 89
The diagnostic value of HLA genotyping in patients who may
have CD revolves around its high negative predictive value,
meaning that patients who lack the appropriate HLA genotype
pairs described above are very unlikely to have CD.
90
However,
the positive predictive value of the HLA genotyping for CD sus-
ceptibility is very low as a large proportion of individuals
without CD carry either HLA-DQ2 or HLA-DQ8 (the preva-
lence of DQ2 in the general population varies between 0% and
40% while that of DQ8 varies between 0% and 20% between
countries
91
). In family screening, DQ2-positive or DQ8-positive
relatives (especially siblings) are at a higher risk of CD,
53 92
with
one study suggesting that DQ2 positivity was associated with a
16-fold increased risk of CD among rst-degree relatives.
53
Specic use of HLA typing
HLA genotyping may be used in patients with suspected CD but
who fail to respond to a GFD. A negative test in this circum-
stance would indicate that patients are highly unlikely to have
CD (<1% of patients with CD are negative for DQ2 and
DQ8
93
) and thus the clinician can direct diagnostic efforts else-
where. HLA typing may similarly be used in patients who are
self-treated on a GFD and never had appropriate testing for CD
before changing their diet. HLA typing may have an adjunctive
role to identifying individuals who are not genetically at risk of
CD and in whom further evaluation for CD is not necessary,
saving a large number of repeated tests for CD in patients who
would otherwise have to undergo testing because they have
symptoms and a rst-degree relative with CD.
94
Recommendations
HLA typing should be used to rule out CD. A positive DQ2.5
or DQ8 can never conrm the diagnosis. (Grade B)
HLA typing should be used in individuals who are
self-treated on a GFD and never had appropriate testing for
CD before changing their diet. (Grade B)
HLA typing can be used to rule out CD, and minimise future
testing, in high-risk individuals with CD, for example,
rst-degree relatives. (Grade B)
Biopsy and endoscopy in CD
There are endoscopic markers of villous atrophy described
scalloping or reduction of duodenal folds and nodularitybut
these are not sensitive enough to preclude a biopsy,
95
and a
normal endoscopic appearance may occur in the presence of
villous atrophy.
9698
Therefore, the endoscopic appearance of
the duodenum should not determine whether biopsy is
performed.
Biopsy of the duodenum for a diagnosis of CD should be per-
formed irrespective of the prior performance of serological
tests, if the patient exhibits symptoms or signs of CD, such as
diarrhoea, weight loss or anaemia. Biopsies can be mounted on
bre-free paper to aid orientation,
99
or alternatively biopsies
could be free oated in formalin. Consultation with the histo-
pathology laboratory is recommended to agree on specimen
presentation.
The villous atrophy may be patchy in CD; hence multiple
biopsies from the bulb and the more distal duodenum are recom-
mended. The taking of at least four biopsy specimens is asso-
ciated with a doubling of the diagnostic rate compared with
patients undergoing a lower number of biopsies (less than
four).
100
In patients with persistently positive coeliac serology
but a normal mucosa, repeat small intestinal biopsy should be
considered, including biopsies from the jejunum.
101
Video
capsule endoscopy may support a CD diagnosis in this setting.
102
A diagnosis of CD has implications for family members, as
overall, around 10% of rst-degree relatives
53
may be affected
and there could be uncertainty in pursuing this diagnosis in the
family if the index case does not have a denite CD diagnosis.
Within an adult population the patient may have other indica-
tions for an upper endoscopy, for example anaemia, and thus
exclusion of other diseases is essential. Upper endoscopy is gen-
erally well tolerated by adults and, in contrast to children, can
usually be readily performed with mild or even no sedation.
Finally histological appearance of the small intestinal mucosa
may also predict the risk of certain future complications, such as
lymphoma ( patients with villous atrophy are at statistically sig-
nicantly higher risk of future lymphoma than patients with a
normal mucosa but positive coeliac serology).
103
Nevertheless there are still adult patients who may be unable
or unwilling to undergo an endoscopy. Under these circum-
stances, assessment of the serological assay and/or level of
IgA-TG2 (if 10 times the upper limit of normal), positive DGP/
EMA, or the use of capsule endoscopy may have a supportive
role. The sensitivity of capsule endoscopy to detect CD is
similar to that of conventional endoscopy when combined with
biopsies. However, it is less invasive, with good specicity and
may provide endoscopic images that can then be used to
support the diagnosis of CD in conjunction with positive ser-
ology. This approach may also be taken in equivocal
cases.
104 105
Recommendations
The diagnosis of CD requires duodenal biopsy when the
patient is on a gluten-containing diet and for the vast
majority of adult patients also positive serology. (Grade B)
Duodenal biopsy should be retained as the mainstay for the
diagnosis of adult CD and cannot be replaced by serology.
(Grade B)
At endoscopy, if there is suspicion of CD, then at least four
biopsy specimens should be obtained, including a duodenal
bulb biopsy. (Grade C)
In serologically negative patients showing signs of
malabsorption (such as anaemia or diarrhoea) or a family
history of CD, a duodenal biopsy should be considered.
(Grade C)
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 5
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
Histopathology diagnosis of CD
It is important that pathologists and clinicians appreciate that
patients with CD benet from a GFD regardless of the degree
of damage in the small intestine and that minor degrees of histo-
logical change suggestive of CD should not be ignored.
106
Marsh
107
described a commonly used classication of this
spectrum. This classication has been modied subsequently by
Oberhuber
108
and simplied by Corazza and Villanacci.
109
Recently, Rostami and Villanacci
110
dened microscopic enteritis
(also called lymphocytic duodenosis,
111
lymphocytic enterop-
athy
111
) and Villanacci et al
112
published a practical classication
with a user-friendly checklist for the histology report of CD.
Some clinicians would prefer a descriptive report, and collab-
oration between pathologists and gastroenterologists as to the
content of the report is valuable. It is important that if serology
has not been performed prior to the biopsy, then this must be
carried out by the requesting physician on receipt of a histology
report suggesting a diagnosis of CD. A biopsy nding of villous
atrophy is not specic for CD. Although CD is the commonest
cause of villous atrophy, there are other causes (table 2); for this
reason the addition of coeliac-specic serology seals the diagno-
sis. The biopsies must be properly oriented (usually by an
experienced laboratory technician) as correct orientation is
necessary for assessment of villous height crypt depth ratio
(derived from the well oriented elds of the biopsies) (gure 1).
The following features should be stated in the report:
Number of biopsies (including those from the duodenal
bulb)
113
and orientation.
The architectural features (normal, partial, sub-total or total
villous atrophy).
Comment on the content of the lamina propria (in CD these
are lymphocytes, plasma cells and eosinophils, and occasion-
ally neutrophils, but cryptitis and crypt abscesses should
suggest other pathology).
Presence of Brunners glands.
Presence of crypt hyperplasia, villous height: crypt depth
ratio (3:1).
112
The absence of plasma cells suggests common
variable immunodeciency.
Evaluation of IELs (with immunocytochemical staining for T
cells (CD3) in equivocal cases
114
) is vital. Counting IELs
should be time efcientsimply counting IELs/20 entero-
cytes at the tips of ve villi,
115 116
or IELs per 50 enterocytes
in two villi and summing these
111
are both reliable and sensi-
tive methods using H&E staining methods. The normal
count has been variably cited; however, in evidence-based
practice
111
and in recent classications,
109 112
<25IELs/100
enterocytes should be taken as the norm.
Use of a simple classication system greatly enhances
intra-observer agreement.
117
Other causes of lymphocytic duodenosis and villous atrophy
Lymphocytic duodenosis
74
is a common condition (3.8% of a
population negative for coeliac serology
111
) seen in association
with infection ( particularly Helicobacter pylori), altered
immune states, for example, common variable immunode-
ciency, autoimmune and chronic inammatory disorders, drugs
and neoplasia.
16 118120
The villous architecture is normal, typ-
ically there is no crypt hyperplasia and IELs are 25/100
enterocytes. Of note, in a single study 16% of cases of lympho-
cytic duodenosis were found to have CD, and in 66% a known
association was found by further investigation.
121
Similarly
there are other causes of villous atrophy in duodenal biopsies,
including immune disorders and deciency, food hypersensitiv-
ity, infection, drugs, neoplasia and miscellaneous disor-
ders
120 122
(table 2). In a study of non-coeliac enteropathy,
70% of patients with this condition were initially diagnosed as
having CD.
123
NOVEL DIAGNOSTIC METHODS
While the current standard tests of serology and conventional
histology are usually adequate to reach a diagnosis of CD, there
are patients whose tests are equivocal and diagnostic uncertainty
remains. Several novel diagnostic approaches have been under-
taken. The deposition of IgA antibodies in close proximity to
TG2 in the small intestine has shown promise as a way of den-
ing early or potential CD in patients who are seropositive but
lack any of the usual histological markers for CD. Recent work
from Finland on IgA-TG2 autoantibody deposition in the small
intestine in such patients shows promise in delivery of an early
Figure 1 Relationship between
villous height and crypt depth. CD,
crypt depth; IEL, intraepithelial
lymphocyte; LD, lymphocytic
duodenosis; PVA, partial villous
atrophy; TVA, total villous atrophy; VH,
villous height. The dots represent IELs.
6 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
prediction of development of CD. However, this is currently
experimental and the methodology requires tissue sections
frozen in liquid nitrogen.
124
Another diagnostic method merit-
ing further evaluation is EmA assay in the culture medium of
small intestinal biopsies.
125 126
Other investigators have
reported their ndings using new techniques associated with
endoscopy to enhance the diagnosis of CD. These include con-
focal microscopy, high-resolution magnication endoscopy,
optical band imaging
127
and optimal coherence tomography.
These novel techniques are still limited by availability, tolerabil-
ity and cost.
98
However, the immersion technique and dye
enhancement in which the endoscopist instills water or a con-
trast dye (for example, indigo carmine or methylene blue) into
the bowel lumen, with or without the assistance of magnica-
tion endoscopy, enhancing the visualisation of the villus
128
can
be readily used and improves visualisation of villi, thus increas-
ing the sensitivity for detection of villous atrophy.
98
Dermatitis herpetiformis
Dermatitis herpetiformis (DH) is the cutaneous manifestation of
gluten-sensitive enteropathy precipitated by exposure to dietary
gluten.
129
It is characterised clinically by herpetiform clusters of
intensely itchy urticated papules and small blisters distributed
on the extensor aspects of the elbows and knees and over the
buttocks and on the scalp. The commonest age of onset is
between the third and fourth decade, though the condition may
occur at any age after weaning. Male patients are affected twice
as often as female patients. For the majority of patients the
disease is lifelong with varying periods of activity, potentially
due to varying degrees of dietary adherence.
The major diagnostic criterion for diagnosis is the presence of
granular IgA deposits in the dermal papillae of uninvolved peri-
lesional skin as shown by direct immunouorescence, and the
diagnosis should not be made unless this has been conrmed.
130
Less than 10% of patients with DH have symptoms or signs
of malabsorption but most have evidence of CD that responds
to a GFD and relapses on gluten challenge. Patients with DH
present with their skin manifestations and are not usually
troubled by the underlying small bowel problem at the time of
presentation.
129 131
Abnormality of the small intestinal mucosa
with either total or subtotal villous atrophy is found in approxi-
mately 70% of patients with DH.
132
A further 25% have
normal villous architecture with increased IELs.
DH shares with CD an increased risk of developing lymphomas
but this seems to be conned to those with severe gut involve-
ment. The risk similarly declines with time on a strict GFD.
133
Due to rash and itch, dapsone is often initiated. More than
70% of patients on a strict GFD are however able to slowly
wean off dapsone over a period of 24 months.
131
FOLLOW-UP
There is a paucity of data pertaining to adherence to a GFD
being improved by follow-up in patients with CD.
17 134138
Only one previous historical study has assessed the impact of
regular follow-up (annual review) at a dedicated doctor-led
coeliac clinic.
136
The investigators suggested that adherence was
improved by having access and regular follow-up within the
setting of a specialist coeliac clinic (improvement in adherence
was 97.5% for those under clinic follow-up vs 40.4% for those
no longer under follow-up); however as this was an observa-
tional study and there were likely to be marked biases in referral
of cases, it is not possible to be condent that the associations
seen were causal. There are no published data assessing the
value of this approach or whether adherence to a GFD, QoL,
avoidance of complications, or satisfaction with the service is
improved by offering a dietitian-led coeliac clinic. One of the
key factors relating to adherence is dietetic input and regular
follow-up.
17 134
Optimally, the clinic should have gastrointes-
tinal and dietetic expertise.
139
Patients should be encouraged to
join disease-specic patient support groups if applicable.
140
Once the disease is stable and the patients manage their diet
without any problems, annual follow-ups should be initiated. The
physician should check on intact small intestinal absorption (full
blood count, ferritin,
32
serum folate,
141
vitamin B12,
142
calcium,
143
alkaline phosphatase
144
), associated autoimmune con-
ditions (thyroid-stimulating hormone and thyroid hormone(s),
145
and serum glucose),
146 147
liver disease (aspartate aminotransfer-
ase/alanine aminotransferase)
40
and dietary adherence (anti-TG2
or EMA/DGP), although the sensitivity and specicity of the
latter
148 149
cannot substitute for structured dietary interview.
In follow-up of CD, the key endpoints are normalisation of
the health of patients judged by an absence of symptoms, and
mucosal healing.
17
A lack of symptoms
72 150 151
or negative
serological markers are not reliable or responsive surrogates of
mucosal response to diet. Dickey et al reported that among 32
patients with CD and persistent villous atrophy, EMA had nor-
malised in 27 (84%); while another British study found that
7/16 (44%) individuals with persistent villous atrophy at
follow-up biopsy had a normalised TTG.
149 152154
The proportion of patients who do not achieve full histo-
logical recovery on diet varies, with most reports suggesting
mucosal healing in 5776%.
150 155159
Some experts favour repeat intestinal biopsy after 1 year of
dietary therapy; others, however, do not believe a repeat biopsy
is essential for coeliac management in typical cases. It is univer-
sally acknowledged that there is little evidence to address
whether clinical outcomes are signicantly altered as a result of
re-biopsy and that the costbenet analysis of such an approach
has yet to be fully established.
Recommendations
Follow-up biopsies may be considered in patients with CD,
and are potentially helpful in identifying patients at
increased risk of lymphoma. (Grade B)
Follow-up biopsies are not mandatory if the patient with CD
is asymptomatic on a GFD, and has no other features that
suggest an increased risk of complications. (Grade C)
Follow-up biopsies should be undertaken in patients with CD
whose condition does not respond to a GFD. (Grade C)
Assessing adherence to the GFD
Whilst the panel of experts agree adherence to the GFD is most
important for the health of a patient with CD, there are no
evidence-based grade A recommendations regarding the most
useful way to assess this. Dietary adherence should guarantee
mucosal healing and at least remission of most gastrointestinal
symptoms. At present there are no non-invasive biomarkers that
indicate complete mucosal recovery and a number of studies
indicate a high prevalence of villous atrophy in adult patients
with CD who appear to be adherent (see table 3). When specif-
ically questioned, the experts agreed there is a difference
between the rst-year follow-up of a newly diagnosed patient
and long-term follow-up of an adherent patient with stable
disease. An adherent patient with stable disease needs less
follow-up and testing than a patient with newly diagnosed CD
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 7
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
in whom the GFD has just commenced, and neither the mucosa
nor biochemical aberrations have yet normalised.
There are four steps to assess dietary adherence: clinical
assessment of symptoms, dietetic review, serum antibodies and
follow-up biopsy.
Symptoms
A meta-analysis of seven studies including more than 3000 sub-
jects showed that the presence of gastrointestinal IBS-like symp-
toms is common in CD. IBS-type symptoms are more common
in patients with CD who are not adherent to a GFD (OR 2.69;
95% CI 0.75 to 9.56).
161
However, patients with CD who are
also adherent to a GFD are more likely to experience ( persist-
ent) symptoms than controls.
161
Dietetic review
The second step is a careful dietetic review conducted by a diet-
itian or dedicated physician. Apart from a visual analogue score
scale which consists of an unmarked line with the anchor sentences
I never adhere to my diet and I always adhere to my diet at each
end,
162
there are a number of questionnaires evaluating self-
reported GFD adherence and food frequency in the English lan-
guage
163166
that are also available in other languages.
167171
These questionnaires should be augmented by a dietetic review,
which is a useful tool to tease out inadvertent gluten intake and to
provide education for a balanced and adequate nutrient intake.
There is no standard or quality control for dietetic review because
local diets and habits require a specic structured interview, which
is related to the quality of the diet. Currently, no data are available
on GFD review outcomes in different countries, and there is no
evidence that a careful review can substitute for other tools (eg,
biopsy) to predict mucosal damage.
Studies report that poor dietary adherence due to occasional
lapses is frequent and it is inuenced by a number of factors,
such as age at diagnosis, knowledge of disease and psychological
factors.
172 173
Serology
The third step in the rst year is to check the IgA-TG2 or appro-
priate serology. Despite contradictory results,
148 149
it is reason-
able to assume that positive antibody titres correspond to some
gluten intake and there is also some evidence that low TTG
titres do not accurately predict mucosal recovery.
148 149
Tursi
et al
148
reported that out of 17 patients with persistent villous
atrophy 1 year after diagnosis, only 1 (6%) was anti-TG2-
positive and 3 (18%) EMA-positive. Vahedi et al
149
reported a
Table 3 Histological recovery of duodenal mucosa in CD
Study Country
Number of
patients
Female
patients
(%)
Median
(M)/average
(A) follow-up,
years
Positive
correlation
between dietary
adherence and
mucosal
improvement
Symptoms
assessed
Main
reason for
mucosal
damage
Histological recovery
of duodenal mucosa
Normal Improved
No
change/
worse
Hutchinson
et al
156
UK 284 71 1.9 (M) p=0.014 No 9% poor
adherence
35% 40% 20.10%
Rubio Tapia
et al
150
USA 241 73 * p<0.01 Yes Poor
adherence/
severe CD
at diagnosis
66%
(5 years)
19%
(25
years)
Lanzini
et al
159
Italy 465 77 1.3 (A) p=0.029 Yes 25% poor
adherence
8% 65% 27%
Ciacci
et al
158
Italy 390 77 6.9 (A) p<0.001 Yes Poor
adherence
43.60% 32.60%
Wahab
et al
160
The
Netherlands
158 72 12 No data NRCD=symptoms 65% 17.1%
(5 years)
Kaukinen
(specific
study of
NRCD)
151
Finland 591
13 with
NRCD
69% of
those
with
NRCD
0.7 (M) p=0.02 Became
symptomatic if
NRCD
46% poor
adherence
1.90%
Tuire et al
72
Finland 177 73 710 No correlation** Patients
asymptomatic
Clinical relevance of
persistent IELs with
normal villi
85%
Lebwohl
et al
155
Sweden 7648 63 1.3 (M) No data no No data 57% 43%
This table is restricted to studies involving at least 100 patients and presents available data on histological recovery of the duodenal mucosa.
Comment on table: In adult studies with >100 patients, non-adherence to a gluten-free diet is a major reason for poor outcome. Symptoms are not a reliable predictor of mucosal
healing. Antibodies are not good enough to predict small intestinal damage,
149
so a follow-up biopsy is important. Lymphocytic duodenosis is common, but not significant in
contribution to symptoms, although it correlates with transgression of adherence to diet.
*Authors present mucosal recovery rate according to KaplanMeier at 2-year and 5-year follow-up.
The authors do not present an exact percentage (or absolute number of female patients). The percentage 77% is based on reported data that the female:male ratio was 3.3:1.
No absolute follow-up time is reported but first follow-up biopsies were carried out between 1 and 2 years after coeliac diagnosis.
Median duration in individuals with persistent villous atrophy. The paper contains no data on the follow-up of the 580 with improved mucosa.
Median duration was 7 years in those with persistent villous atrophy but 10 years in those with normal mucosa. The abstract of the paper states an average follow-up of 11 years but
that figure is not reported in the paper.
**All individuals, also those with persistent mucosal villous atrophy, had a good dietary adherence. Hence, there can be no positive correlation between dietary adherence and mucosal
improvement.
p value (Fishers exact test) calculated by us based on 6/13 versus 0/18 with poor dietary adherence; see table 1 in original publication for explanation.
CD, coeliac disease; IEL, intraepithelial lymphocyte; NRCD, non-responsive CD.
8 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
substantially higher sensitivity for persistent total villous
atrophy (73% and 91% for IgA-TG2 and EMA, respectively),
but reported no data for partial villous atrophy.
Follow-up biopsy
The last step is the follow-up biopsy. Some authors suggested
that it is important to perform a duodenal biopsy to assess the
recovery of intestinal mucosa and to exclude RCD and malig-
nancies. However, one recent study of 7648 individuals failed
to show that overall mortality was increased in patients with CD
with persistent villous atrophy at follow-up biopsy in patients
with a median follow-up of more than 11 years.
174
In many cases 1 year is too brief a timespan to obtain com-
plete recovery of duodenal mucosa. Tuire et al
72
found that
IELs were more frequent even 25 years after coeliac diagnosis
compared with thereafter. Some experts do not routinely
perform a follow-up biopsy in asymptomatic patients with nega-
tive serology and good adherence. Currently there are no
studies indicating an absolute necessity of follow-up biopsy for
all patients, but of eight major studies, ve
150 151 159 160 175
examined follow-up biopsies at roughly 25 years (table 3). This
may be related to costs (economic and psychological) of per-
forming a procedure that in theory should be repeated many
times over the years of follow-up as dietary adherence may vary
over time.
The authors of this review underline the necessity of distin-
guishing asymptomatic patients in whom clinical improvement,
negative serology, and potentially a follow-up biopsy and good
adherence assessed by dietetic review are considered sufcient
from symptomatic patients in whom repeated biopsies are
needed to rule out RCD or malignancies. Studies report that
poor dietary adherence due to occasional lapses is frequent and
it is inuenced by a number of factors, such as age at diagnosis,
knowledge of disease and psychological factors.
172 173
Histological recovery of duodenal mucosa in adult patients
with CD
In adults, neither symptoms
72 150 151 156 158160
nor serology
149
is reliable to predict small intestinal damage;
149
assessing
mucosal healing by biopsy is the key. Serum antibodies have
poor sensitivity for persistent villous atrophy, especially 1 year
or more after diagnosis and institution of a GFD.
Lymphocytic duodenosis is commonly seen on biopsy of
follow-up patients. It is rarely symptomatic, although it may also
correlate with transgression of adherence from the GFD.
72
Early biopsy (at 6 months) is not considered to be optimal.
156
Recommendations
When adherence is questioned, it should be reviewed by a
dietitian. (Grade C)
Symptomatic patients should be evaluated more thoroughly
than asymptomatic patients. (Grade C)
Gluten challenge
To perform a gluten challenge, a recent study recommends a
14-day gluten intake at 3 g of gluten/day (two slices of wheat
bread per day) to induce histological and serological changes in
the majority of adults with CD.
176
The challenge can be pro-
longed to 8 weeks if serology remains negative at 2 weeks (in
the Lefer et al
176
study, serology was negative after 2 weeks in
all cases, but positive after another 2 weeks).
Medical management during follow-up
Long-term follow-up can be in secondary care clinics or in
primary care as long as the expertise is available.
177
However,
prompt access to specialist centres or secondary care is recom-
mended if any problems arise, and it should be noted that the
need for long-term follow-up is controversial.
17 178
The risk of osteoporosis
144 179183
and bone fracture
184190
is
increased with CD,
2
with one Swedish study showing an excess
risk of any fracture of 481/100 000 person-years in adults with
CD
189
and a British study (13% of individuals were children)
320/100 000 person-years.
184
The excess risk is reduced with
good dietary adherence and reduction in intestinal villous
atrophy, and bone density increases during the rst year of GFD
adherence.
191195
However, one population-based study found a
similar excess risk for fractures before and after coeliac diagnosis
(eg, the incidence ratio 510 years before CD diagnosis was 1.8
compared with 2.2 some 510 years after diagnosis).
189
On the basis of current evidence, the suggestion should there-
fore be to measure calcium, alkaline phosphatase and vitamin D
levels (and parathyroid hormone for compensatory increase) at
diagnosis and replace as necessary. Calcium intake should be
maintained at or above 1000 mg per day.
196
Bone density
should be measured in those at high risk of osteoporosis; appro-
priate criteria for judging this are given by the BSG (http://www.
bsg.org.uk/images/stories/clinical/ost_coe_ibd.pdf ). Repeat bone
density investigations (generally after an interval of 2 years)
should otherwise be considered in patients who have low bone
density on index measurement following initiation of appropri-
ate treatment, or who have evidence of ongoing villous atrophy
or poor dietary adherence. Postmenopausal women with CD
may require supplementation in addition to the GFD.
197
Loss of
bone density at a greater than expected rate should prompt
measurement of vitamin D levels, dietary review of adherence,
consideration of repeat intestinal mucosal biopsy and review of
additional risk factors such as hypogonadism.
Hyposplenism
198
associated with CD may result in impaired
immunity to encapsulated bacteria, and an increase in such
infections has been demonstrated in CD.
199201
Hyposplenism
does not seem to correlate with duration of GFD.
198
Vaccination against Pneumococcus is therefore recommended.
202
However, it is unclear whether vaccination with the conjugated
vaccine is preferable in this setting and whether additional vac-
cination against Haemophilus, Meningococcus and Inuenza
203
should be considered if not previously given.
204
It should also
be noted that patients with CD may have a weaker response to
hepatitis B vaccination than normal.
205 206
Recommendations
Newly diagnosed patients should have vaccination for
Pneumococcus. (Grade C)
Bone density should be measured after 1 year of diet in
patients who have additional risk factors for osteoporosis or
if over the age of 55 years. (Grade D)
Adult patients with CD should have a calcium intake of at
least 1000 mg per day. (Grade D)
Patients with CD require follow-up by a dietitian and/or
clinician with an interest or expertise in this eld. (Grade D)
Patients should have annual haematological and biochemical
proles. (Grade D)
A GFD is the core management strategy for prevention of
osteoporosis. (Grade D)
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 9
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
SCREENING FOR CD
Table 4 lists the WHO criteria for general population screening.
CD fulls many of these criteria but not all of them as yet. Our
recommendation is active case nding but not mass screening.
Given that CD is a common disease (about 1% of the western
population
22 24
) and that there is a therapy (GFD) that most
often relieves symptoms, and may even have an effect on the
risk of future complications, any patient with signs or symptoms
of CD should undergo testing.
In addition, testing should be carried out in high-risk groups
such as those with iron deciency anaemia,
224
Downs syn-
drome,
225
type 1 diabetes mellitus,
147
osteoporosis
184 189 226
and IBS when CD is suspected. The coeliac prevalence in these
groups typically varies between 2% and 5%.
144 227 228
Recommendations
There is insufcient evidence to recommend population
screening for CD, however there should be a low threshold
for case nding in clinical practice as per National Institute
for Health and Care Excellence guidelines.
3
(Grade B)
Symptomatic rst-degree relatives of patients with CD
should undergo CD testing. (Grade C)
QUALITY OF LIFE
There has been a growing interest in how patients with CD per-
ceive the impact of their diagnosis, how their psychological state
is inuenced by the disease and the effect of GFD. Several
studies report that patients with CD have lower QoL scores
than the general population.
229232
Though some studies have found improved QoL with GFD
treatment in symptomatic and screening-detected patients,
222 233
others suggest that any benet of diet is restricted to those pre-
senting with symptomatic disease.
214 220 221
Since mood disor-
ders such as anxiety, depression and fatigue are often linked
with CD, before and after diagnosis, it is likely that they may
contribute to the effect upon QoL. A recent meta-analysis
234
suggested that depression, but not anxiety, is more common in
adults with CD. Also fatigue has been linked to undetected
CD.
235
GLUTEN-FREE DIET
The mainstay of treatment of CD and DH is a GFD. The term
gluten should be used to indicate not only wheat-based proteins
(gliadins), but it also includes those from barley (hordeins) and
rye (secalins), and cereal hybrids such as triticale.
236
Originally
oats were also avoided in the GFD. Earlier research indicates
that oats uncontaminated by gluten are probably safe for
patients with CD.
237 238
This is important because oats contain
soluble bre, are able to lower blood glucose and attenuate
insulin response.
239
The appearance of symptoms related to
introducing oats might be due to cross contamination. Also a
small percentage of patients with CD may be sensitive to oats
240
and develop symptoms or even mucosal damage.
241 242
Patients
with CD should be educated to avoid cereals and food contain-
ing gluten (breakfast cereals, ours, pasta, cakes, biscuits, sauces
etc) derived from wheat, barley or rye and food made from
gluten-contaminated oats, and encouraged to eat naturally
occurring gluten-free foods and alternative sources of starch
(corn, rice, potatoes etc). Levels of susceptibility to gluten con-
tamination of food vary among patients with CD. Although it
has also been suggested that the acceptable threshold for gluten
content in gluten-free products can from the clinical point of
view be set at 100 ppm ( mg/kg),
243
the Codex Alimentarius
Commission of the WHO issued new guidelines for gluten
content of processed food in 2008 and a law from the
European Commission (EC41/2009), effective since January
2012, stipulated that foods labelled as gluten free should
contain 20 parts per million of gluten, and that this gluten
content is safe for the coeliac population. The 20 ppm threshold
for gluten-free food is also accepted by the US Food and Drug
Administration, effective since August 2013. Newly diagnosed
patients should be referred to a dietitian to discuss dietary man-
agement.
243
It is important that they are educated not only to
avoid gluten but also to have a sufcient intake of nutrients,
vitamins, bre and calcium present in their GFD.
244
Recent data
also indicate that a strict GFD might be of help in reaching ideal
body weight, whether an individual is underweight or obese at
diagnosis.
245
Data suggest that adherence to the GFD is better
achieved when the patient is well educated and supported by
carers and families. However, there is a wide variation in provi-
sion of dietary consultation services for patients diagnosed with
CD in the world. A survey indicates that dietetic support may
be underprovided in the UK.
246
Table 4 CD as a candidate for general population screening
WHO criteria Comment
That the disease is common and well defined CD occurs in approximately 1% of the western population,
24 111
and is even more frequent in selected
populations.
207 208
Screening tests are simple, safe and accurate IgA-TG2 screening offers high sensitivity and specificity but the positive predictive value does not attain 100%,
83
with a consequent risk of false-positive cases.
92 209
The screening test should be culturally acceptable Screening seems to be culturally accepted in most parts of the world
Treatment is available GFD offers symptomatic relief and will often lead to mucosal healing.
Clinical detection is difficult The clinical picture of CD varies, and many patients only have minor symptoms,
31 208 210
making it difficult to
diagnose CD.
If undiagnosed and untreated the disease will lead
to severe complications
The only available treatment is a GFD. The GFD seems to reduce symptoms and the risk of complications in
symptomatic patients,
211213
but it is doubtful whether the GFD leads to health gains in asymptomatic
patients,
81 214218 219
although it should be noted that a proportion of patients who first claim to be asymptomatic in
fact have symptoms.
216
It is also unclear if QoL in patients with screen-detected asymptomatic CD is
different
81 214 217 220223
from that of the control populations. Without a decrease in pre-diagnostic QoL, institution of
a GFD is unlikely to result in improved QoL after diagnosis.
Testing and treatment is cost effective Cost effectiveness depends on the duration of symptoms and the risk of complications in untreated versus treated CD.
It is still not clear if the risk of complications is substantially lower in diagnosed CD than in undiagnosed CD.
CD, coeliac disease; GFD, gluten-free diet; QoL, quality of life; TG2, transglutaminase 2.
10 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
Safe gluten intake?
As shown previously, diagnosis and advice on a gluten-free diet
often lead to mucosal recovery, while poor adherence to diet
slows or hinders complete recovery.
Whilst contamination of the diet by gluten may be unavoid-
able and increased IEL counts are associated with less severe
nutritional and metabolic consequences than classic CD with
villous atrophy, patients may have signicant signs and symp-
toms related to this disorder. A clinical response and mucosal
recovery can be achieved by strict adherence to a gluten-free
diet. On balance, data support treatment of patients with CD
regardless of the degree of mucosal damage.
247
A recent review on safe gluten levels argues that daily
intakes of <10 mg have no effect on mucosal histology,
248
whereas denite alterations are caused by a daily intake of
500 mg and observable alterations by 100 mg. A calculated daily
intake of 30 mg seems not to harm the mucosa. Therefore, at
present, a safe limit could be set between 10 and 100 mg.
249
The most comprehensive systematic review (35 studies) suggests
that while the amount of tolerable gluten varies among people
with CD, a daily gluten intake of <10 mg is unlikely to cause
signicant histological abnormalities.
248
There is extensive research on GFD and how this may inuence
the clinical course. While a recent study of more than 7000 indivi-
duals undergoing follow-up biopsy found no association between
persistent villous atrophy (likely to signal poorer dietary adherence)
and overall mortality,
174
this does not rule out that poor dietary
adherence is negative for specic health outcome, such as auto-
immune disease,
250 251
pregnancy outcome (two studies have found
poor foetal outcome in pregnant women with undiagnosed CD but
not in diagnosed CD
41 252
), and especially the risk of lymphoma.
Most studies on lymphoma risk according to dietary adherence
point towards a protective effect from GFD but the studies have so
far been small in size,
212 253255
preventing rm conclusions.
However, recently Lebwohl et al
175
found a statistically signicantly
increased risk of lymphoma in patients with CD with persistent
villous atrophy compared with those with mucosal healing.
A frequent complaint of patients with CD is that they experience
limitations in their social life because of difculty accessing gluten-
free meals or concern about the safety of food when eating out. The
patients worries are justied as a survey shows that chefs knowl-
edge about CD is lower than that of the general public.
256
Therefore, education about a GFD needs to be directed to catering
personnel. Another criticism is that the availability of gluten-free
food is clearly limited in more rural areas and shopping for gluten-
free food is time consuming. In most countries, high-quality gluten-
free products are available in supermarkets or in special health food
stores and on the internet, but the cost of gluten-free food is much
greater than the equivalent wheat-based foods.
257 258
Recommendations
Patients should adhere to a GFD and have an intake of less
than 10 mg gluten per day. (Grade B)
Gluten challenge is not recommended in the ordinary
patient with CD, but in patients in whom the diagnosis
remains unclear despite a follow-up biopsy, gluten challenge
should be performed. (Grade C)
Patients may commence gluten-free oats at diagnosis. (Grade D)
A GFD is recommended to decrease the excess risk of
adverse foetal outcome and of lymphoma among patients
with CD. (Grade C)
Patient information and support
Patient support should not be a monologue by a physician, but
instead a two-way communication involving the patient, and his/
her family. Ideally, collaboration between the patient, the patients
family, an expert dietitian and interested physician should be the
setting in which the GFD should be initiated. Family involvement
is very important as the disease (including GFD) will inevitably
affect family members, but also joining a national coeliac support
group can help patients cope with their disease.
Patients need information, reassurance and the opportunity to
learn at their pace about the rather challenging demands of a
GFD. They particularly need to be encouraged and motivated
to adapt to and maintain a GFD. Continued professional
engagement in their follow-up care is likely to help sustain that
motivation. Unfortunately, many patients report that they are
not satised with the amount and quality of the information
offered by their physician.
216
Physicians should inform patients
even before the CD diagnosis, offering information about sero-
logical testing, and what it means to undergo a small intestinal
biopsy. Worries are common in patients awaiting a biopsy
259
and the physician should address this anxiety.
Patients may want information about when and where CD
occurs, its aetiology and whether it is a common disease or not.
Understanding the vital role of GFD in the management of CD
is important for dietary adherence.
140
Attaining dietary adher-
ence may be particularly difcult in patients identied through
screening. Patients with CD often report reduced QoL because
of dietetic restrictions and this must be taken into
account.
260263
Part of the reduced QoL may be due to social
restrictions, such as not being able to eat out with friends, and
the economic burden from the more expensive GFD.
258 264
Patients should also be informed that while most symptoms are
likely to go away on a GFD, some symptoms may persist (see
section on Non-responsive CD).
Recommendation
At diagnosis, patients should be encouraged to join their
national coeliac support group. In the UK, patients should be
advised about gluten-free items on prescription (FP10),
details of which are available from http://www.coeliac.org.uk.
Non-responsive CD
After adoption of the GFD, 430% of patients with CD report
persisting symptoms and are considered to be affected by non-
responsive CD (NRCD).
265
Once the initial diagnosis of CD has
been conrmed, adherence to the GFD should be assessed by an
expert dietitian as inadvertent or deliberate gluten exposure is
the most frequent cause of NRCD.
265 266
After these initial steps, evaluation should be individualised;
however assessment for ongoing enteropathy plays a central
role, therefore a follow-up biopsy is needed. Small bowel
imaging should be performed in any patient with abdominal
pain, persisting fever, obstruction, anaemia, gastrointestinal
bleeding or unexplained weight loss.
267
If duodenal biopsy does
not reveal a persistent enteropathy, symptoms are likely to be
due to a second condition.
265 268
While the GFD is efcacious at controlling the signs and
symptoms of CD and improving intestinal histology in most
patients, treatment of CD is currently imperfect. Over time, vir-
tually all individuals with CD will have symptomatic exacerba-
tions due to gluten exposure and up to 30% of patients will
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 11
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
have symptoms severe or chronic enough to visit their treating
physician
265 266
Comprehensive monitoring of patients with CD requires
assessment for NRCD and efcient evaluation for potential aeti-
ologies. NRCD may be considered in three categories.
265 266 269
First and most commonly, NRCD is due to continued dietary
exposure to gluten.
265 266 269
Second, it may be due to a pre-
existing or coincidental condition causing symptoms that resem-
ble CD which may have led to the detection of otherwise
asymptomatic CDthis includes IBS
33
or colonic malignancy
causing anaemia.
270
Finally, it may be due to conditions asso-
ciated with CD,
268
such as secondary lactose intolerance,
265
pancreatic exocrine insufciency,
271
small bowel bacterial over-
growth, microscopic colitis
272
and cows milk protein sensitivity.
Very occasionally a state refractory to gluten withdrawal
occurs, referred to as RCD. Figure 2 suggests an outline for
investigating patients with NRCD.
In symptomatic patients with ongoing enteropathy and RCD,
coeliac-related malignancies and disorders that mimic CD (table
2) must be excluded. RCD is dened as persistent or recurrent
malabsorptive symptoms and/or signs with villous atrophy
despite a strict GFD for more than 12 months in the absence of
other causes of villous atrophy or malignant complications and
after conrmation of CD.
7
RCD is subdivided into type I
(RCDI) and type II (RCDII). The most important aspect of dif-
ferentiating RCDI and RCDII is demonstration of a monoclonal
population of T cells or aberrant T cells in the latter. There are
different methods available for this, including genetic analysis of
T-cell receptor clonality, immunohistochemistry and ow cyto-
metry. Most laboratories will employ at least two methods but
their relative contributions remain uncertain. Currently there is
variation in the criteria used to diagnose RCDII. The two
factors that support the diagnosis of RCDII include loss of
normal surface markers CD3 and CD8 with preserved expres-
sion of intracytoplasmic CD3 and detection of monoclonal
rearrangement of T-cell receptor chain.
120
Areas of uncertainty
include the use of immunohistochemistry versus ow cytometry
for IEL classication and the relative prognosis of patients with
discordant IEL and T-cell studies.
Patients with normal CD3 and CD8 expression and no evi-
dence of T-cell monoclonality have RCDI with a good progno-
sis. Patients with RCDII have a poorer prognosis, due
predominantly to nutritional complications and transformation
into enteropathy-associated T-cell lymphoma (EATL).
273 274
Ulcerative jejunoileitis (UJI) is a rare condition characterised by
inammatory ulceration of the small bowel that arises in
RCD.
274276
The nding of UJI should raise suspicion for
lymphoma.
Figure 2 Investigation of the patient
with non-responsive coeliac disease
(NRCD). Based on a gure by Mooney
et al.
305
FODMAPs, fermentable
oligosaccharides, disaccharides,
monosaccharides, and polyols; GI,
gastrointestinal; HLA, human leucocyte
antigen; RCD, refractory coeliac
disease; SIBO, small intestine bacterial
overgrowth.
12 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
There is no standard treatment for RCD. Elemental diets, sys-
temic steroids, oral budesonide, oral thioguanines including
azathioprine are used in RCDI (and sometimes are benecial),
but have limited benet in RCDII.
277 278
In RCDII, cyclospor-
ine, cladribine and high-dose chemotherapy with autologous
stem cell support have been reported; however, therapy must be
individualised and include surveillance for EATL.
120
EATL is a rare lymphoma strongly associated with RCDII,
273
which carries a poor prognosis with a cumulative 5-year survival
of less than 20%.
279 280
Currently, two groups of EATL are
recognised
281
: EATL type I accounts for 8090% of all cases and
is a large cell lymphoma exclusively associated with CD.
282
In
contrast, EATL type II has not been associated with CD.
281 283
The poor prognosis of EATL is determined by extent of
disease at diagnosis, multifocal small bowel involvement, poor
general health and presence of complications including perfor-
ation that preclude chemotherapy.
120
Presence of RCDII is asso-
ciated with a poor prognosis compared with isolated EATL in
CD without RCDII. There is no proven effective treatment in
RCDII, though a number of strategies have been proposed, and
patients should be referred to a tertiary centre to optimise their
management.
Finally, numerous studies have conrmed the association
between CD and B-cell lymphoma
284286
and CD and small
intestinal adenocarcinoma.
42 254 287289
Recommendations
Patients with persistent symptoms despite a GFD should
have a follow-up biopsy. (Grade B)
In symptomatic patients with ongoing enteropathy and RCD,
coeliac-related malignancies and disorders that mimic CD
must be excluded. (Grade C)
Small bowel imaging should be performed in any patient with
abdominal pain, fever, obstruction, anaemia, gastrointestinal
bleeding or unexplained weight loss. (Grade D)
Patients with RCD should be referred to a tertiary centre to
optimise their management. (Grade D)
NOVEL TREATMENT
The role of non-dietary therapiesas an adjunct or as an alter-
native to the GFDhas yet to be ascertained. Of the candidate
approaches, immunotherapy (including hookworm expos-
ure)
290 291
is currently explored as an alternative to GFD but,
even if successful, is unlikely to benet all patients. The role of
glutenases ( propyl endopeptidases) and tight junction regulators
(table 5) are likely to be in reducing the threshold response and
optimising the benets of gluten restriction rather than allowing
a normal gluten-containing diet.
Table 5 contains a list of potential novel treatments. None of
the available novel treatments can as yet ( January 2014) be
recommended for use outside clinical trials.
Recommendation
None of the available novel treatments can as yet be
recommended for use outside clinical trials. (Grade D)
DISCUSSION
We recommend testing for CD in those with suggested symp-
toms or syndromes, especially if they have a rst-degree relative
with CD.
53
There is not yet sufcient evidence to support indis-
criminate general population screening. Specic serology such as
IgA-TG2 and IgG-DGP with or without a strategy for determin-
ation of total IgA level should be the preferred serologic strategy
for detection of CD. Ideally a combination of serology and
biopsies done on a gluten-containing diet will then provide the
most robust diagnosis of CD. Additional testing may be neces-
sary for those with less than clear-cut results.
We recommend a duodenal biopsy before the diagnosis of
CD. This contrasts with the recent ESPGHAN recommenda-
tions where a duodenal biopsy is optional in symptomatic
paediatric patients in whom the IgA-TG2 level exceeds 10
times the upper limit of normal, EMA antibodies are positive
on a separately taken blood sample and HLA typing is positive
for DQ2 or DQ8.
1
CD has been linked to a large number of
symptoms (table 1 in the ESPGHAN review on paediatric CD
Table 5 Novel treatment in CD
Treatment Comment
Cereal genomics The high copy numbers in gliadin genes have so far limited attempts to genetically modify cultivars incapable of
expressing immunotoxic peptides
292
RNA interference of protein translation may reduce gliadin expression, with evidence of reduced proliferation of
lymphocytes challenged in vitro
293
Prolyl endopeptidases (PEPs) These endopeptidases have been isolated from microbial sources and may be capable of enzymatic cleavage of the
immunotoxic gluten peptides ex vivo.
294 295 296
A combination of a glutamine-specific endoprotease (EP-B2 from barley)
and a prolyl endopeptidase (subcutaneous PEP from Sphingomonas capsulata)
297
acts synergistically and has been
evaluated in a 6-week phase IIA clinical trial against 2 g gluten taken daily
298
Larazotide acetate Larazotide is a tight junction regulator
299
and maintains intestinal barrier function after gluten challenge.
299 300
Phase
IIA clinical trials have demonstrated limited effects on intestinal permeability after gluten ingestion, but beneficial effects
on symptoms and signs.
301
TG2 inhibitors Candidate peptidomimetic blockers are currently entering clinical trials but no data are available yet.
302
A potential
limitation of this drug candidate is that TG2 activity occurs in a number of diseases and a TG2 inhibitor may therefore
have unwanted side effects
Blocking of the antigen presenting groove of
HLA-DQ molecules
No trials yet. Regarded as unpredictable
Subcutaneous injection of dominant immunotoxic
gliadin peptides
65
Stimulates an immunoregulatory T-cell response or deplete or anergise antigen specific memory T cells.
Responses would be specific to the HLA haplotype DQ2 or 8. Ongoing phase II trials.
Polymer binding agents
303
No clinical trials performed yet
CD, coeliac disease; HLA, human leucocyte antigen; TG2, transglutaminase 2.
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 13
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
lists 16 such symptoms and signs with additional symptoms in
the text).
1
There is a risk that all symptoms, independent of
their origin, may be taken as a sign of CD when the sensitivity
and specicity of even gastrointestinal symptoms are moderate
in CD.
304
Other reasons to require a small intestinal biopsy
prior to diagnosis is that not all commercial IgA-TG2 kits are
of high quality,
78
and that alternative diagnoses may be more
common, and sometimes serious, in adults with suspected CD.
Finally, an initial biopsy is important for the follow-up of
patients, especially those whose condition is non-responsive to
a GFD.
Adequate (more than four) biopsies should be taken, from the
distal duodenum and the duodenal bulb to maximise diagnosis.
The threshold for abnormal IELs is 25/100 enterocytes, but
for a denite diagnosis villous atrophy is required. However,
lesser degrees of damage (25 IELs but no villous atrophy) com-
bined with positive serology (IgA-EMA, TTG or IgGDGP) may
also represent CD (probable CD),
The treatment of CD is a lifelong and strict GFD. The goal of
treatment is to relieve symptoms, achieve mucosal healing, avoid
complications of CD, and have a good QoL with a nutritionally
complete GFD. This is best achieved when patients are moti-
vated and receive expert information in a collaborative way,
with resources including expert dietitians and interested medical
care. Follow-up of CD is needed to ensure response to symp-
toms, prevention of consequences, and continued maintenance
of motivation to remain gluten free.
Discussions upon the issue of repeating duodenal biopsies
were intense in our group. There is no conclusive evidence of
the benet of universal follow-up biopsy, and we were unable to
reach a consensus. Some would undertake follow-up biopsies in
all patients with CD after 25 years on a GFD (table 3). There
are others who reserve follow-up biopsies for those in whom
there are persistent and recurrent symptoms or those for whom
the follow-up biopsies are necessary to help conrm the diagno-
sis in the setting of continued diagnostic uncertainty.
There are novel techniques that may enhance the sensitivity
of endoscopic examination, of which the immersion technique
is probably the most feasible currently. An understanding of the
precise value of various serologic strategies in the detection of
CD is continuing to evolve, as are advances in therapies that
may ultimately provide some mitigation of the impairment of
the QoL that is inherent in a strict GFD.
Ultimately, this review should not be regarded as xed. Instead
it represents our understanding of what is best in adult CD man-
agement according to current knowledge. It is likely, and indeed
we hope, that there will be substantial progress in diagnosis,
evaluation and management of CD to reduce the burden on the
patient and society. We anticipate that future updated versions of
these guidelines will be based on evolving published literature
that may have an audit or research base relevant to adult CD.
Data in this review were mostly retrieved through searches of
PubMed. We acknowledge that there are other medical databases
and we cannot rule out that had we searched more than one data-
base we may have identied additional relevant studies that have
now been left out of this review.
Areas for future research
The panel of experts recognise that the main challenge in the
future is to allocate available resources effectively to reduce the
burden of disease from CD.
Future research should focus on the following areas:
how to induce long-term remission without a GFD, that is,
novel therapies and vaccine;
better understanding of the disease processes, including gen-
etics and antigen presentation;
prevent and cure extra-intestinal manifestation and complica-
tions, including infections;
be able to assess tolerable amount of gluten for individual
patients;
dene the role of duodenal biopsy, serology and point of
care testing at diagnosis and follow-up;
nd a robust and valid blood marker for diagnosis and moni-
toring of the disease;
understand the pathogenesis of RCDI and II.
Author afliations
1
Department of Medical Epidemiology and Biostatistics, Karolinska Institutet,
Stockholm, Sweden
2
Department of Paediatrics, rebro University Hospital, rebro, Sweden
3
Department of Medicine, Dr C. Bonorino Udaondo Gastroenterology Hospital, Del
Salvador University, Buenos Aires, Argentina
4
Coeliac Centre/1st Department of Internal Medicine, University of Pavia, Fondazione
IRCCS Policlinico San Matteo, Pavia, Italy
5
University of Nottingham, Department of Epidemiology and Public Health,
Nottingham City Hospital, Nottingham, UK
6
Department of Medicine and Surgery, University of Salerno, Salerno, Italy
7
Gastroenterology, Division of Nutritional Sciences, Kings College London, The
Rayne Institute, St Thomas Hospital, London, UK
8
Coeliac Disease Center at Columbia University, New York, New York, USA
9
Department of Neurology, Royal Hallamshire Hospital, Shefeld, UK
10
Registered dietitian and representative of the British Dietetic Association, Bath,
Somerset, UK
11
Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen
Mary University of London, London, UK
12
School of Medicine, University of Tampere, Tampere, Finland
13
Department of Gastroenterology and Alimentary Tract Surgery, Tampere University
Hospital, Tampere, Finland
14
Department of Medicine, Seinjoki Central Hospital, Finland
15
Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard
Medical School, Boston, Massachusetts, USA
16
Department of Dermatology, Imperial College NHS Healthcare Trust, St Marys
Hospital, London, UK
17
Department of Gastroenterology, Centre for Immune Regulation, Oslo University
Hospital Rikshospitalet, Oslo, Norway
18
Coeliac UK, Apollo Centre, London, UK
19
Patient Representative & Regional Chairman for Coeliac UK, Shefeld, UK
20
Division of Gastroenterology and Hepatology, Department of Immunology Mayo
Clinic, Rochester, Minnesota, USA
21
Department of Gastroenterology, University Hospital Llandough, Wales, UK
22
Anatomical Pathology, University of Newcastle, Faculty of Health and Medicine,
School of Medicine & Public Health, Callaghan, Australia
23
Department of Medicine and Surgery, University of Salerno, Salerno, Italy
24
Gastroenterology and Liver Unit, Royal Hallamshire Hospital & University of
Shefeld, Shefeld, UK
Acknowledgements We thank all the patients involved in the related focus
groups and discussions.
Contributors JFL and DSS initiated the study. JFL coordinated the project and
conducted the web surveys on CD. DSS supervised the project and served as the
representative of the British Society of Gastroenterology (BSG). All authors
contributed to the literature searches, contributed to the writing of the manuscript,
and approved the nal version of the manuscript.
Funding JFL was supported by the Swedish Research Council (522-2A09-195) and
the Swedish Society of Medicine while writing the draft of this paper. JCB was
supported by Consejo en Investigacion en Salud GCABA. KK was supported by The
Academy of Finland, the Competitive Research Funding of the Tampere University
Hospital (9N062), and The Sigrid Juselius Foundation. JAM was supported by DK
57892. DSS has received an educational grant from Biocard and Simtomax to
undertake an investigator-led research studies on CD and/or gluten sensitivity, and
an educational grant from Dr Schr (a gluten-free food manufacturer) to undertake
an investigator-led research study on gluten sensitivity.
Competing interests DAvH, TRC: received grant support from Coeliac UK. PHRG:
scientic advisory board of Alvine Pharmaceuticals and ImmusanT. DAL: Ironwood
Pharmaceuticals, Alvine Therapeutics, Alba Pharmaceuticals, Shire Therapeutics, In
Nova Diagnostics and CRICO Risk Management Foundation. JAM: consultant for
Alba Pharmaceuticals, Alvine Therapeutics, Flamentera, 2GPharma Inc and
ImmusanT. KEAL: ImmusanT, Regeneron and Alvine Pharmaceuticals. DSS: received
14 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
educational grants from Coeliac UK, Biocard, Simtomax and Dr Schr (a gluten-free
food manufacturer) to undertake an investigator-led research study on CD and/or
gluten sensitivity.
Ethics approval British Society of Gastroenterology.
Provenance and peer review Not commissioned; externally peer reviewed.
Open Access This is an Open Access article distributed in accordance with the
Creative Commons Attribution Non Commercial (CC BY-NC 3.0) license, which
permits others to distribute, remix, adapt, build upon this work non-commercially,
and license their derivative works on different terms, provided the original work is
properly cited and the use is non-commercial. See: http://creativecommons.org/
licenses/by-nc/3.0/
REFERENCES
1 Husby S, Koletzko S, Korponay-Szabo IR, et al. European Society for Pediatric
Gastroenterology, Hepatology, and Nutrition guidelines for the diagnosis of coeliac
disease. J Pediatr Gastroenterol Nutr 2012;54:13660.
2 Ciclitira P, Dewar DH, McLaughlin SD, et al. The Management of Adults with
Coeliac Disease. London: British Society of Gastroenterology, 2010.
3 NICE. Coeliac Disease. Recognition and Assessment of Celiac Disease. London,
2009.
4 Bai JC, Fried M, Corazza GR, et al. World gastroenterology organisation global
guidelines on celiac disease. J Clin Gastroenterol 2013;47:1216.
5 Rubio-Tapia A, Hill ID, Kelly CP, et al. ACG clinical guidelines: diagnosis and
management of celiac disease. Am J Gastroenterol 2013;108:65676; quiz 77.
6 National Institutes of Health Consensus Development Conference Statement on
Celiac Disease, June 2830, 2004. Gastroenterology 2005;128(Suppl 1):S19.
7 Ludvigsson JF, Lefer DA, Bai JC, et al. The Oslo denitions for coeliac disease
and related terms. Gut 2013;62:4352.
8 Fasano A, Catassi C. Current approaches to diagnosis and treatment of celiac
disease: an evolving spectrum. Gastroenterology 2001;120:63651.
9 Farrell RJ, Kelly CP. Celiac sprue. N Engl J Med 2002;346:1808.
10 Green PH, Cellier C. Celiac disease. N Engl J Med 2007;357:173143.
11 Green PH, Jabri B. Coeliac disease. Lancet 2003;362:38391.
12 Di Sabatino A, Corazza GR. Coeliac disease. Lancet 2009;373:148093.
13 Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel
therapies. Gastroenterology 2009;137:191233.
14 van Heel DA, West J. Recent advances in coeliac disease. Gut 2006;55:103746.
15 Lefer D. Celiac disease diagnosis and management: a 46-year-old woman with
anemia. JAMA 2011;306:158292.
16 Walker MM, Murray JA. An update in the diagnosis of coeliac disease.
Histopathology 2011;59:16679.
17 Haines ML, Anderson RP, Gibson PR. Systematic review: the evidence base for
long-term management of coeliac disease. Aliment Pharmacol Ther
2008;28:104266.
18 Oxford Centre for Evidence-based MedicineLevels of Evidence. Oxford: Centre
for Evidence-based Medicine, Oxford University, 2009.
19 Dicke WK, Weijers HA, Van De Kamer JH. Coeliac disease. II. The presence in
wheat of a factor having a deleterious effect in cases of coeliac disease. Acta
Paediatr 1953;42:3442.
20 Taylor KB, Truelove SC, Thomson DL, et al. An immunological study of coeliac
disease and idiopathic steatorrhoea. Serological reactions to gluten and milk
proteins. Br Med J 1961;2:172731.
21 Cooke WT, Fone DJ, Cox EV, et al. Adult coeliac disease. Gut 1963;4:27991.
22 Dube C, Rostom A, Sy R, et al. The prevalence of celiac disease in average-risk
and at-risk Western European populations: a systematic review. Gastroenterology
2005;128:S5767.
23 Corazza GR, Andreani ML, Biagi F, et al. The smaller size of the coeliac iceberg
in adults. Scand J Gastroenterol 1997;32:91719.
24 Maki M, Mustalahti K, Kokkonen J, et al. Prevalence of celiac disease among
children in Finland. N Engl J Med 2003;348:251724.
25 Mustalahti K, Catassi C, Reunanen A, et al. The prevalence of celiac disease in
Europe: results of a centralized, international mass screening project. Ann Med
2010;42:58795.
26 Bingley PJ, Williams AJ, Norcross AJ, et al. Undiagnosed coeliac disease at age
seven: population based prospective birth cohort study. BMJ 2004;328:3223.
27 West J, Logan RF, Hill PG, et al. Seroprevalence, correlates, and characteristics of
undetected coeliac disease in England. Gut 2003;52:9605.
28 Rubio-Tapia A, Ludvigsson JF, Brantner TL, et al. The prevalence of celiac disease
in the United States. Am J Gastroenterol 2012;107:153844; quiz 37, 45.
29 Murray JA, Van Dyke C, Plevak MF, et al. Trends in the identication and clinical
features of celiac disease in a North American community, 19502001. Clin
Gastroenterol Hepatol 2003;1:1927.
30 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;163:28692.
31 Rampertab SD, Pooran N, Brar P, et al. Trends in the presentation of celiac
disease. Am J Med 2006;119:355 e914.
32 Bergamaschi G, Markopoulos K, Albertini R, et al. Anemia of chronic disease and
defective erythropoietin production in patients with celiac disease. Haematologica
2008;93:178591.
33 Sanders DS, Carter MJ, Hurlstone DP, et al. Association of adult coeliac disease
with irritable bowel syndrome: a case-control study in patients fullling ROME II
criteria referred to secondary care. Lancet 2001;358:15048.
34 Hadjivassiliou M, Gibson A, Davies-Jones GA, et al. Does cryptic gluten sensitivity
play a part in neurological illness? [see comments]. Lancet 1996;347:36971.
35 Ludvigsson JF, Olsson T, Ekbom A, et al. A population-based study of coeliac
disease, neurodegenerative and neuroinammatory diseases. Aliment Pharmacol
Ther 2007;25:131727.
36 Hadjivassiliou M, Grunewald R, Sharrack B, et al. Gluten ataxia in perspective:
epidemiology, genetic susceptibility and clinical characteristics. Brain
2003;126:68591.
37 Ludvigsson JF, Reutfors J, Osby U, et al. Coeliac disease and risk of mood
disordersa general population-based cohort study. J Affect Disord
2007;99:11726.
38 Groll A, Candy DC, Preece MA, et al. Short stature as the primary manifestation of
coeliac disease. Lancet 1980;2:10979.
39 Olmos M, Antelo M, Vazquez H, et al. Systematic review and meta-analysis of
observational studies on the prevalence of fractures in coeliac disease. Dig Liver
Dis 2008;40:4653.
40 Ludvigsson JF, Elfstrom P, Broome U, et al.Celiac disease and risk of liver
disease: a general population-based study. Clin Gastroenterol Hepatol
2007;5:6369 e1.
41 Ludvigsson JF, Montgomery SM, Ekbom A. Celiac disease and risk of adverse fetal
outcome: a population-based cohort study. Gastroenterology 2005;129:
45463.
42 West J, Logan RF, Smith CJ, et al. Malignancy and mortality in people with coeliac
disease: population based cohort study. BMJ 2004;329:71619.
43 Meeuwisse GW. Round table discussion. Diagnostic criteria in coeliac disease. Acta
Paediatr 1970;59:46163.
44 McNeish AS, Harms HK, Rey J, et al. The diagnosis of coeliac disease. A
commentary on the current practices of members of the European Society for
Paediatric Gastroenterology and Nutrition (ESPGAN). Arch Dis Child
1979;54:7836.
45 Revised Criteria for Diagnosis of Coeliac Disease. Report of Working Group of
European Society of Paediatric Gastroenterology and Nutrition. Arch Dis Child
1990;65:90911.
46 Sollid LM, Lie BA. Celiac disease genetics: current concepts and practical
applications. Clin Gastroenterol Hepatol 2005;3:84351.
47 Welander A, Tjernberg AR, Montgomery SM, et al. Infectious disease and risk of
later celiac disease in childhood. Pediatrics 2010;125:e5306.
48 Greco L, Auricchio S, Mayer M, et al. Case control study on nutritional risk factors
in celiac disease. J Pediatr Gastroenterol Nutr 1988;7:3959.
49 Ivarsson A, Persson LA, Nystrom L, et al. Epidemic of coeliac disease in Swedish
children. Acta Paediatr 2000;89:16571.
50 Stene LC, Honeyman MC, Hoffenberg EJ, et al. Rotavirus infection frequency and
risk of celiac disease autoimmunity in early childhood: a longitudinal study. Am J
Gastroenterol 2006;101:233340.
51 Riddle MS, Murray JA, Porter CK. The incidence and risk of celiac disease in a
healthy US adult population. Am J Gastroenterol 2012;107:124855.
52 Jabri B, Sollid LM. Tissue-mediated control of immunopathology in coeliac disease.
Nat Rev Immunol 2009;9:85870.
53 Rubio-Tapia A, Van Dyke CT, Lahr BD, et al. Predictors of family risk for celiac
disease: a population-based study. Clin Gastroenterol Hepatol 2008;6:
9837.
54 Greco L, Romino R, Coto I, et al. The rst large population based twin study of
coeliac disease. Gut 2002;50:6248.
55 Monsuur AJ, Wijmenga C. Understanding the molecular basis of celiac disease:
what genetic studies reveal. Ann Med 2006;38:57891.
56 Spurkland A, Sollid LM, Polanco I, et al. HLA-DR and -DQ genotypes of celiac
disease patients serologically typed to be non-DR3 or non-DR5/7. Hum Immunol
1992;35:18892.
57 Trynka G, Hunt KA, Bockett NA, et al. Dense genotyping identies and localizes
multiple common and rare variant association signals in celiac disease. Nat Genet
2011;43:1193201.
58 Garner CP, Murray JA, Ding YC, et al. Replication of celiac disease UK
genome-wide association study results in a US population. Hum Mol Genet
2009;18:421925.
59 Beitnes AC, Raki M, Lundin KE, et al. Density of CD163+ CD11c+ dendritic cells
increases and CD103+ dendritic cells decreases in the coeliac lesion. Scand J
Immunol 2011;74:18694.
60 Raki M, Tollefsen S, Molberg O, et al. A unique dendritic cell subset accumulates
in the celiac lesion and efciently activates gluten-reactive T cells. Gastroenterology
2006;131:42838.
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 15
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
61 Lundin KE, Scott H, Hansen T, et al. Gliadin-specic, HLA-DQ(alpha 1*0501,beta
1*0201) restricted T cells isolated from the small intestinal mucosa of celiac
disease patients. J Exp Med 1993;178:18796.
62 Lundin KE, Scott H, Fausa O, et al. T cells from the small intestinal mucosa of a
DR4, DQ7/DR4, DQ8 celiac disease patient preferentially recognize gliadin when
presented by DQ8. Hum Immunol 1994;41:28591.
63 van de Wal Y, Kooy YM, van Veelen PA, et al. Small intestinal T cells of celiac
disease patients recognize a natural pepsin fragment of gliadin. Proc Natl Acad Sci
U S A 1998;95:100504.
64 Ellis HJ, Pollock EL, Engel W, et al. Investigation of the putative immunodominant
T cell epitopes in coeliac disease. Gut 2003;52:2127.
65 Camarca A, Anderson RP, Mamone G, et al. Intestinal T cell responses to gluten
peptides are largely heterogeneous: implications for a peptide-based therapy in
celiac disease. J Immunol 2009;182:415866.
66 Molberg O, McAdam SN, Korner R, et al. Tissue transglutaminase selectively
modies gliadin peptides that are recognized by gut-derived T cells in celiac
disease [see comments] [ published erratum appears in Nat Med 1998 Aug;4
(8):974]. Nat Med 1998;4:7137.
67 Tollefsen S, Arentz-Hansen H, Fleckenstein B, et al. HLA-DQ2 and -DQ8
signatures of gluten T cell epitopes in celiac disease. J Clin Invest 2006;116:
222636.
68 Dieterich W, Ehnis T, Bauer M, et al. Identication of tissue transglutaminase
as the autoantigen of celiac disease [see comments]. Nat Med 1997;3:
797801.
69 Sulkanen S, Halttunen T, Laurila K, et al. Tissue transglutaminase autoantibody
enzyme-linked immunosorbent assay in detecting celiac disease [see comments].
Gastroenterology 1998;115:13228.
70 Di Niro R, Mesin L, Zheng NY, et al. High abundance of plasma cells secreting
transglutaminase 2-specic IgA autoantibodies with limited somatic hypermutation
in celiac disease intestinal lesions. Nat Med 2012;18:4415.
71 Rashtak S, Ettore MW, Homburger HA, et al. Comparative usefulness of
deamidated gliadin antibodies in the diagnosis of celiac disease. Clin Gastroenterol
Hepatol 2008;6:42632; quiz 370.
72 Tuire I, Marja-Leena L, Teea S, et al. Persistent duodenal intraepithelial
lymphocytosis despite a long-term strict gluten-free diet in celiac disease. Am J
Gastroenterol 2012;107:15639.
73 Rostami K, Mulder CJ, van Overbeek FM, et al. Should relatives of coeliacs with
mild clinical complaints undergo a small-bowel biopsy despite negative serology?
Eur J Gastroenterol Hepatol 2000;12:515.
74 Hammer ST, Greenson JK. The clinical signicance of duodenal lymphocytosis with
normal villus architecture. Arch Pathol Lab Med 2013;137:121619.
75 Dahlbom I, Olsson M, Forooz NK, et al. Immunoglobulin G (IgG) anti-tissue
transglutaminase antibodies used as markers for IgA-decient celiac disease
patients. Clin Diagn Lab Immunol 2005;12:2548.
76 Rashtak S, Ettore MW, Homburger HA, et al. Combination testing for antibodies in
the diagnosis of coeliac disease: comparison of multiplex immunoassay and ELISA
methods. Aliment Pharmacol Ther 2008;28:80513.
77 Hill PG, Holmes GK. Coeliac disease: a biopsy is not always necessary for
diagnosis. Aliment Pharmacol Ther 2008;27:5727.
78 Egner W, Shrimpton A, Sargur R, et al. ESPGHAN guidance on coeliac disease
2012: multiples of ULN for decision making do not harmonise assay performance
across centres. J Pediatr Gastroenterol Nutr 2012;55:7335.
79 Lewis NR, Scott BB. Meta-analysis: deamidated gliadin peptide antibody and
tissue transglutaminase antibody compared as screening tests for coeliac disease.
Aliment Pharmacol Ther 2010;31:7381.
80 Raivio T, Kaukinen K, Nemes E, et al. Self transglutaminase-based rapid coeliac
disease antibody detection by a lateral ow method. Aliment Pharmacol Ther
2006;24:14754.
81 Korponay-Szabo IR, Szabados K, Pusztai J, et al. Population screening for coeliac
disease in primary care by district nurses using a rapid antibody test: diagnostic
accuracy and feasibility study. BMJ 2007;335:12447.
82 Collin P, Kaukinen K, Vogelsang H, et al. Antiendomysial and antihuman
recombinant tissue transglutaminase antibodies in the diagnosis of coeliac disease:
a biopsy-proven European multicentre study. Eur J Gastroenterol Hepatol
2005;17:8591.
83 Hopper AD, Cross SS, Hurlstone DP, et al. Pre-endoscopy serological testing for
coeliac disease: evaluation of a clinical decision tool. BMJ 2007;334:729.
84 Dickey W, Hughes DF, McMillan SA. Reliance on serum endomysial antibody
testing underestimates the true prevalence of coeliac disease by one fth. Scand J
Gastroenterol 2000;35:1813.
85 Murray JA, Herlein J, Mitros F, et al. Serologic testing for celiac disease in the
United States: results of a multilaboratory comparison study. Clin Diagn Lab
Immunol 2000;7:5847.
86 Quine MA, Bell GD, McCloy RF, et al. Prospective audit of upper gastrointestinal
endoscopy in two regions of England: safety, stafng, and sedation methods. Gut
1995;36:4627.
87 Lebwohl B, Tennyson CA, Holub JL, et al. Sex and racial disparities in duodenal
biopsy to evaluate for celiac disease. Gastrointest Endosc 2012;76:77985.
88 Nachman F, Vazquez H, Gonzalez A, et al. Gastroesophageal reux symptoms in
patients with celiac disease and the effects of a gluten-free diet. Clin Gastroenterol
Hepatol 2011;9:2149.
89 Margaritte-Jeannin P, Babron MC, Bourgey M, et al. HLA-DQ relative risks for
coeliac disease in European populations: a study of the European Genetics Cluster
on Coeliac Disease. Tissue Antigens 2004;63:5627.
90 Karell K, Louka AS, Moodie SJ, et al. HLA types in celiac disease patients not
carrying the DQA1*05-DQB1*02 (DQ2) heterodimer: results from the European
Genetics Cluster on Celiac Disease. Hum Immunol 2003;64:46977.
91 Abadie V, Sollid LM, Barreiro LB, et al. Integration of genetic and immunological
insights into a model of celiac disease pathogenesis. Annu Rev Immunol
2011;29:493525.
92 Hadithi M, von Blomberg BM, Crusius JB, et al. Accuracy of serologic tests and
HLA-DQ typing for diagnosing celiac disease. Ann Intern Med 2007;147:
294302.
93 Polvi A, Arranz E, Fernandez-Arquero M, et al. HLA-DQ2-negative celiac disease in
Finland and Spain. Hum Immunol 1998;59:16975.
94 Rashtak S, Murray JA. Tailored testing for celiac disease. Ann Intern Med
2007;147:33941.
95 Dickey W, Hughes D. Disappointing sensitivity of endoscopic markers for villous
atrophy in a high-risk population: implications for celiac disease diagnosis during
routine endoscopy. Am J Gastroenterol 2001;96:21268.
96 Olds G, McLoughlin R, OMorian C, et al. Celiac disease for the endoscopist.
Gastrointest Endosc 2002;56:40715.
97 Oxentenko AS, Grisolano SW, Murray JA, et al. The insensitivity of endoscopic
markers in celiac disease. Am J Gastroenterol 2002;97:9338.
98 Cammarota G, Fedeli P, Gasbarrini A. Emerging technologies in upper
gastrointestinal endoscopy and celiac disease. Nat Clin Pract Gastroenterol Hepatol
2009;6:4756.
99 Serra S, Jani PA. An approach to duodenal biopsies. J Clin Pathol
2006;59:113350.
100 Lebwohl B, Kapel RC, Neugut AI, et al. Adherence to biopsy guidelines increases
celiac disease diagnosis. Gastrointest Endosc 2011;74:1039.
101 Horoldt BS, McAlindon ME, Stephenson TJ, et al. Making the diagnosis of coeliac
disease: is there a role for push enteroscopy? Eur J Gastroenterol Hepatol
2004;16:11436.
102 Chang MS, Rubin M, Lewis SK, et al. Diagnosing celiac disease by video capsule
endoscopy (VCE) when esophogastroduodenoscopy (EGD) and biopsy is unable to
provide a diagnosis: a case series. BMC Gastroenterol 2012;12:90.
103 Elfstrom P, Granath F, Ekstrom Smedby K, et al. Risk of lymphoproliferative
malignancy in relation to small intestinal histopathology among patients with
celiac disease. J Natl Cancer Inst 2011;103:43644.
104 Tennyson CA, Ciaccio EJ, Lewis SK. Video capsule endoscopy in celiac disease.
Gastrointest Endosc Clin N Am 2012;22:74758.
105 Atlas DS, Rubio-Tapia A, Van Dyke CT, et al. Capsule endoscopy in nonresponsive
celiac disease. Gastrointest Endosc 2011;74:131522.
106 Kurppa K, Collin P, Viljamaa M, et al. Diagnosing mild enteropathy celiac disease:
a randomized, controlled clinical study. Gastroenterology 2009;136:81623.
107 Marsh MN. Gluten, major histocompatibility complex, and the small intestine. A
molecular and immunobiologic approach to the spectrum of gluten sensitivity
(celiac sprue). Gastroenterology 1992;102:33054.
108 Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease:
time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol
1999;11:118594.
109 Corazza GR, Villanacci V. Coeliac disease. J Clin Pathol 2005;58:5734.
110 Rostami K, Villanacci V. Microscopic enteritis: novel prospect in coeliac disease
clinical and immuno-histogenesis. Evolution in diagnostic and treatment strategies.
Dig Liver Dis 2009;41:24552.
111 Walker MM, Murray JA, Ronkainen J, et al. Detection of celiac disease and
lymphocytic enteropathy by parallel serology and histopathology in a
population-based study. Gastroenterology 2010;139:11219.
112 Villanacci V, Ceppa P, Tavani E, et al. Coeliac disease: the histology report. Dig
Liver Dis 2011;43(Suppl 4):S38595.
113 Arguelles-Grande C, Tennyson CA, Lewis SK, et al. Variability in small bowel
histopathology reporting between different pathology practice settings: impact on
the diagnosis of coeliac disease. J Clin Pathol 2012;65:2427.
114 Ensari A. Gluten-sensitive enteropathy (celiac disease): controversies in diagnosis
and classication. Arch Pathol Lab Med 2010;134:82636.
115 Biagi F, Luinetti O, Campanella J, et al. Intraepithelial lymphocytes in the villous
tip: do they indicate potential coeliac disease? J Clin Pathol 2004;57:8359.
116 Jarvinen TT, Collin P, Rasmussen M, et al. Villous tip intraepithelial lymphocytes
as markers of early-stage coeliac disease. Scand J Gastroenterol 2004;39:
42833.
117 Corazza GR, Villanacci V, Zambelli C, et al. Comparison of the interobserver
reproducibility with different histologic criteria used in celiac disease. Clin
Gastroenterol Hepatol 2007;5:83843.
118 Brown I, Mino-Kenudson M, Deshpande V, et al. Intraepithelial lymphocytosis in
architecturally preserved proximal small intestinal mucosa: an increasing diagnostic
16 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
problem with a wide differential diagnosis. Arch Pathol Lab Med
2006;130:10205.
119 Daum S, Cellier C, Mulder CJ. Refractory coeliac disease. Best Pract Res Clin
Gastroenterol 2005;19:41324.
120 Rubio-Tapia A, Murray JA. Classication and management of refractory coeliac
disease. Gut 2010;59:54757.
121 Aziz I, Evans KE, Hopper AD, et al. A prospective study into the aetiology of
lymphocytic duodenosis. Aliment Pharmacol Ther 2010;32:13927.
122 Rubio-Tapia A, Herman ML, Ludvigsson JF, et al. Severe spruelike enteropathy
associated with olmesartan. Mayo Clin Proc 2012;87:7328.
123 Pallav K, Lefer DA, Tariq S, et al. Noncoeliac enteropathy: the differential
diagnosis of villous atrophy in contemporary clinical practice. Aliment Pharmacol
Ther 2012;35:38090.
124 Koskinen O, Collin P, Lindfors K, et al. Usefulness of small-bowel mucosal
transglutaminase-2 specic autoantibody deposits in the diagnosis and follow-up
of celiac disease. J Clin Gastroenterol 2010;44:4838.
125 Carroccio A, Iacono G, DAmico D, et al. Production of anti-endomysial antibodies
in cultured duodenal mucosa: usefulness in coeliac disease diagnosis. Scand J
Gastroenterol 2002;37:328.
126 Carroccio A, Di Prima L, Pirrone G, et al. Anti-transglutaminase antibody assay of
the culture medium of intestinal biopsy specimens can improve the accuracy of
celiac disease diagnosis. Clin Chem 2006;52:117580.
127 Banerjee R, Reddy DN. High-resolution narrow-band imaging can identify patchy
atrophy in celiac disease: targeted biopsy can increase diagnostic yield.
Gastrointest Endosc 2009;69:9845.
128 Siegel LM, Stevens PD, Lightdale CJ, et al. Combined magnication endoscopy
with chromoendoscopy in the evaluation of patients with suspected
malabsorption. Gastrointest Endosc 1997;46:22630.
129 Leonard J, Haffenden G, Tucker W, et al. Gluten challenge in dermatitis
herpetiformis. N Engl J Med 1983;308:81619.
130 Seah PP, Fry L. Immunoglobulins in the skin in dermatitis herpetiformis and their
relevance in diagnosis. Br J Dermatol 1975;92:15766.
131 Fry L, Leonard JN, Swain F, et al. Long term follow-up of dermatitis
herpetiformis with and without dietary gluten withdrawal. Br J Dermatol
1982;107:63140.
132 Fry L, Seah PP, Harper PG, et al. The small intestine in dermatitis herpetiformis.
J Clin Pathol 1974;27:81724.
133 Lewis HM, Renaula TL, Garioch JJ, et al. Protective effect of gluten-free diet
against development of lymphoma in dermatitis herpetiformis. Br J Dermatol
1996;135:3637.
134 Hall NJ, Rubin G, Charnock A. Systematic review: adherence to a gluten-free diet
in adult patients with coeliac disease. Aliment Pharmacol Ther 2009;30:31530.
135 Mayer M, Greco L, Troncone R, et al. Compliance of adolescents with coeliac
disease with a gluten free diet. Gut 1991;32:8815.
136 Bardella MT, Molteni N, Prampolini L, et al. Need for follow up in coeliac disease.
Arch Dis Child 1994;70:2113.
137 Ljungman G, Myrdal U. Compliance in teenagers with coeliac diseasea Swedish
follow-up study. Acta Paediatr 1993;82:2358.
138 Kolsteren MM, Koopman HM, Schalekamp G, et al. Health-related quality of life in
children with celiac disease. J Pediatr 2001;138:5935.
139 Pietzak MM. Follow-up of patients with celiac disease: achieving compliance with
treatment. Gastroenterology 2005;128:S13541.
140 Lefer DA, Edwards-George J, Dennis M, et al. Factors that inuence adherence to
a gluten-free diet in adults with celiac disease. Dig Dis Sci 2008;53:157381.
141 Dickey W, Ward M, Whittle CR, et al. Homocysteine and related B-vitamin status
in coeliac disease: effects of gluten exclusion and histological recovery. Scand J
Gastroenterol 2008;43:6828.
142 Saibeni S, Lecchi A, Meucci G, et al. Prevalence of hyperhomocysteinemia in adult
gluten-sensitive enteropathy at diagnosis: role of B12, folate, and genetics. Clin
Gastroenterol Hepatol 2005;3:57480.
143 Ciacci C, Spagnuolo G, Tortora R, et al. Urinary stone disease in adults with celiac
disease: prevalence, incidence and urinary determinants. J Urol 2008;180:9749.
144 Stenson WF, Newberry R, Lorenz R, et al. Increased prevalence of celiac disease
and need for routine screening among patients with osteoporosis. Arch Intern Med
2005;165:3939.
145 Elfstrom P, Montgomery SM, Kampe O, et al. Risk of thyroid disease in individuals
with celiac disease. J Clin Endocrinol Metab 2008;93:391521.
146 Rapoport MJ, Bistritzer T, Vardi O, et al. Increased prevalence of diabetes-related
autoantibodies in celiac disease. J Pediatr Gastroenterol Nutr 1996;23:5247.
147 Ludvigsson JF, Ludvigsson J, Ekbom A, et al. Celiac disease and risk of subsequent
Type 1 diabetes: a general population cohort study of children and adolescents.
Diabetes Care 2006;29:24838.
148 Tursi A, Brandimarte G, Giorgetti GM. Lack of usefulness of anti-transglutaminase
antibodies in assessing histologic recovery after gluten-free diet in celiac disease.
J Clin Gastroenterol 2003;37:38791.
149 Vahedi K, Mascart F, Mary JY, et al. Reliability of antitransglutaminase antibodies
as predictors of gluten-free diet compliance in adult celiac disease. Am J
Gastroenterol 2003;98:107987.
150 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;105:141220.
151 Kaukinen K, Peraaho M, Lindfors K, et al. Persistent small bowel mucosal villous
atrophy without symptoms in coeliac disease. Aliment Pharmacol Ther
2007;25:123745.
152 Dickey W, Hughes DF, McMillan SA. Patients with serum IgA endomysial
antibodies and intact duodenal villi: clinical characteristics and management
options. Scand J Gastroenterol 2005;40:12403.
153 Hopper AD, Hadjivassiliou M, Hurlstone DP, et al. What is the role of serologic
testing in celiac disease? A prospective, biopsy-conrmed study with economic
analysis. Clin Gastroenterol Hepatol 2008;6:31420.
154 Dickey W, Hughes DF, McMillan SA. Disappearance of endomysial antibodies in
treated celiac disease does not indicate histological recovery [in process citation].
Am J Gastroenterol 2000;95:7124.
155 Lebwohl B, Murray JA, Rubio-Tapia A, et al. Predictors of persistent villous atrophy
in coeliac disease: a population-based study. Aliment Pharmacol Ther
2014;39:48895.
156 Hutchinson JM, West NP, Robins GG, et al. Long-term histological follow-up of
people with coeliac disease in a UK teaching hospital. QJM 2010;103:51117.
157 Selby WS, Painter D, Collins A, et al. Persistent mucosal abnormalities in coeliac
disease are not related to the ingestion of trace amounts of gluten. Scand J
Gastroenterol 1999;34:90914.
158 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;66:17885.
159 Lanzini A, Lanzarotto F, Villanacci V, et al. Complete recovery of intestinal mucosa
occurs very rarely in adult coeliac patients despite adherence to gluten-free diet.
Aliment Pharmacol Ther 2009;29:1299308.
160 Wahab PJ, Meijer JW, Mulder CJ. Histologic follow-up of people with celiac
disease on a gluten-free diet: slow and incomplete recovery. Am J Clin Pathol
2002;118:45963.
161 Sainsbury A, Sanders DS, Ford AC. Prevalence of irritable bowel syndrome-type
symptoms in patients with celiac disease: a meta-analysis. Clin Gastroenterol
Hepatol 2013;11:35965.e1.
162 Ciacci C, Iavarone A, Mazzacca G, et al. Depressive symptoms in adult coeliac
disease. Scand J Gastroenterol 1998;33:24750.
163 Edwards George JB, Lefer DA, Dennis MD, et al. Psychological correlates of
gluten-free diet adherence in adults with celiac disease. J Clin Gastroenterol
2009;43:3016.
164 Lefer DA, Dennis M, Edwards George JB, et al. A simple validated gluten-free
diet adherence survey for adults with celiac disease. Clin Gastroenterol Hepatol
2009;7:5306, 36 e12.
165 Sainsbury K, Mullan B. Measuring beliefs about gluten free diet adherence in adult
coeliac disease using the theory of planned behaviour. Appetite 2011;56:47683.
166 Chauhan JC, Kumar P, Dutta AK, et al. Assessment of dietary compliance to
gluten free diet and psychosocial problems in Indian children with celiac disease.
Indian J Pediatr 2010;77:64954.
167 Ciacci C, DAgate C, De Rosa A, et al. Self-rated quality of life in celiac disease.
Dig Dis Sci 2003;48:221620.
168 Hopman EG, Koopman HM, Wit JM, et al. Dietary compliance and health-related
quality of life in patients with coeliac disease. Eur J Gastroenterol Hepatol
2009;21:105661.
169 Hogberg L, Grodzinsky E, Stenhammar L. Better dietary compliance in patients
with coeliac disease diagnosed in early childhood. Scand J Gastroenterol
2003;38:7514.
170 Biagi F, Bianchi PI, Marchese A, et al. A score that veries adherence to a
gluten-free diet: a cross-sectional, multicentre validation in real clinical life. Br J
Nutr 2012;108:18848.
171 Charalampopoulos D, Panayiotou J, Chouliaras G, et al. Determinants of
adherence to gluten-free diet in Greek children with coeliac disease: a
cross-sectional study. Eur J Clin Nutr 2013;67:61519.
172 Kurppa K, Lauronen O, Collin P, et al. Factors associated with dietary adherence in
celiac disease: a nationwide study. Digestion 2012;86:30914.
173 van Hees NJ, Van der Does W, Giltay EJ. Coeliac disease, diet adherence and
depressive symptoms. J Psychosom Res 2013;74:15560.
174 Lebwohl B, Granath F, Ekbom A, et al. Mucosal healing and mortality in coeliac
disease. Aliment Pharmacol Ther 2013;37:3329.
175 Lebwohl B, Granath F, Ekbom A, et al. Mucosal healing and risk for
lymphoproliferative malignancy in celiac disease: a population-based cohort study.
Ann Intern Med 2013;159:16975.
176 Lefer D, Schuppan D, Pallav K, et al. Kinetics of the histological, serological and
symptomatic responses to gluten challenge in adults with coeliac disease. Gut
2013;62:9961004.
177 Herman ML, Rubio-Tapia A, Lahr BD, et al. Patients with celiac disease are not
followed up adequately. Clin Gastroenterol Hepatol 2012;10:89399 e1.
178 Bebb JR, Lawson A, Knight T, et al. Long-term follow-up of coeliac diseasewhat
do coeliac patients want? Aliment Pharmacol Ther 2006;23:82731.
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 17
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
179 Corazza GR, Di Sario A, Cecchetti L, et al. Bone mass and metabolism in patients
with celiac disease. Gastroenterology 1995;109:1228.
180 Corazza GR, Di Stefano M, Maurino E, et al. Bones in coeliac disease: diagnosis
and treatment. Best Pract Res Clin Gastroenterol 2005;19:45365.
181 McFarlane XA, Bhalla AK, Reeves DE, et al. Osteoporosis in treated adult coeliac
disease. Gut 1995;36:71014.
182 Passananti V, Santonicola A, Bucci C, et al. Bone mass in women with celiac
disease: role of exercise and gluten-free diet. Dig Liver Dis 2012;44:37983.
183 Capriles VD, Martini LA, Areas JA. Metabolic osteopathy in celiac disease:
importance of a gluten-free diet. Nutr Rev 2009;67:599606.
184 West J, Logan RF, Card TR, et al. Fracture risk in people with celiac disease: a
population-based cohort study. Gastroenterology 2003;125:42936.
185 Vestergaard P, Mosekilde L. Fracture risk in patients with celiac Disease, Crohns
disease, and ulcerative colitis: a nationwide follow-up study of 16,416 patients in
Denmark. Am J Epidemiol 2002;156:110.
186 Vasquez H, Mazure R, Gonzalez D, et al. Risk of fractures in celiac disease patients:
a cross-sectional, case- control study. Am J Gastroenterol 2000;95:1839.
187 Moreno ML, Vazquez H, Mazure R, et al. Stratication of bone fracture risk in
patients with celiac disease. Clin Gastroenterol Hepatol 2004;2:12734.
188 Davie MW, Gaywood I, George E, et al. Excess non-spine fractures in women over
50 years with celiac disease: a cross-sectional, questionnaire-based study.
Osteoporos Int 2005;16:11505.
189 Ludvigsson JF, Michaelsson K, Ekbom A, et al. Coeliac disease and the risk of
fracturesa general population-based cohort study. Aliment Pharmacol Ther
2007;25:27385.
190 Jafri MR, Nordstrom CW, Murray JA, et al. Long-term fracture risk in patients with
celiac disease: a population-based study in Olmsted County, Minnesota. Dig Dis
Sci 2007;53:96471.
191 Corazza GR, Di Sario A, Cecchetti L, et al. Inuence of pattern of clinical
presentation and of gluten-free diet on bone mass and metabolism in adult coeliac
disease. Bone 1996;18:52530.
192 Bai JC, Gonzalez D, Mautalen C, et al. Long-term effect of gluten restriction on
bone mineral density of patients with coeliac disease. Aliment Pharmacol Ther
1997;11:15764.
193 Valdimarsson T, Lofman O, Toss G, et al. Reversal of osteopenia with diet in adult
coeliac disease. Gut 1996;38:3227.
194 Ciacci C, Maurelli L, Klain M, et al. Effects of dietary treatment on bone mineral
density in adults with celiac disease: factors predicting response. Am J
Gastroenterol 1997;92:9926.
195 Mautalen C, Gonzalez D, Mazure R, et al. Effect of treatment on bone mass,
mineral metabolism, and body composition in untreated celiac disease patients.
Am J Gastroenterol 1997;92:31318.
196 Committee to Review Dietary Reference Intakes for Vitamin D and Calcium, Food
and Nutrition Board, Institute of Medicine. Dietary Reference Intakes for Calcium
and Vitamin D. Washington DC, 2010.
197 Malabanan AO, Holick MF. Vitamin D and bone health in postmenopausal
women. J Womens Health (Larchmt) 2003;12:1516.
198 Di Sabatino A, Rosado MM, Cazzola P, et al. Splenic hypofunction and the
spectrum of autoimmune and malignant complications in celiac disease. Clin
Gastroenterol Hepatol 2006;4:17986.
199 Ludvigsson JF, Olen O, Bell M, et al. Coeliac disease and risk of sepsis. Gut
2008;57:107480.
200 Thomas HJ, Wotton CJ, Yeates D, et al. Pneumococcal infection in patients with
coeliac disease. Eur J Gastroenterol Hepatol 2008;20:6248.
201 Corazza GR, Frisoni M, Vaira D, et al. Effect of gluten-free diet on splenic
hypofunction of adult coeliac disease. Gut 1983;24:22830.
202 Primary Care Society for Gastroenterology UK. The Management of Adults with
Coeliac Disease in Primary Care, 2006.
203 Marild K, Fredlund H, Ludvigsson JF. Increased risk of hospital admission for
inuenza in patients with celiac disease: a nationwide cohort study in Sweden. Am
J Gastroenterol 2010;105:246573.
204 McKinley M, Leibowitz S, Bronzo R, et al. Appropriate response to pneumococcal
vaccine in celiac sprue. J Clin Gastroenterol 1995;20:11316.
205 Noh KW, Poland GA, Murray JA. Hepatitis B vaccine nonresponse and celiac
disease. Am J Gastroenterol 2003;98:228992.
206 Zingone F, Morisco F, Zanetti A, et al. Long-term antibody persistence and
immune memory to hepatitis B virus in adult celiac patients vaccinated as
adolescents. Vaccine 2011;29:10058.
207 Myleus A, Ivarsson A, Webb C, et al. Celiac disease revealed in 3% of Swedish
12-year-olds born during an epidemic. J Pediatr Gastroenterol Nutr
2009;49:1706.
208 Vilppula A, Kaukinen K, Luostarinen L, et al. Increasing prevalence and high
incidence of celiac disease in elderly people: a population-based study. BMC
Gastroenterol 2009;9:49.
209 Katz KD, Rashtak S, Lahr BD, et al. Screening for celiac disease in a North
American population: sequential serology and gastrointestinal symptoms. Am J
Gastroenterol 2011;106:13339.
210 Collin P, Huhtala H, Virta L, et al. Diagnosis of celiac disease in clinical practice:
physicians alertness to the condition essential. J Clin Gastroenterol 2007;41:1526.
211 Murray JA, Watson T, Clearman B, et al. Effect of a gluten-free diet on
gastrointestinal symptoms in celiac disease. Am J Clin Nutr 2004;79:66973.
212 Holmes GK, Prior P, Lane MR, et al. Malignancy in coeliac diseaseeffect of a
gluten free diet. Gut 1989;30:3338.
213 Sanchez MI, Mohaidle A, Baistrocchi A, et al. Risk of fracture in celiac
disease: gender, dietary compliance, or both? World J Gastroenterol 2011;17:
303542.
214 Johnston SD, Rodgers C, Watson RG. Quality of life in screen-detected and typical
coeliac disease and the effect of excluding dietary gluten. Eur J Gastroenterol
Hepatol 2004;16:12816.
215 Whitaker JK, West J, Holmes GK, et al. Patient perceptions of the burden of
coeliac disease and its treatment in the UK. Aliment Pharmacol Ther
2009;29:11316.
216 Ukkola A, Maki M, Kurppa K, et al. Patients experiences and perceptions of living
with coeliac diseaseimplications for optimizing care. J Gastrointestin Liver Dis
2012;21:1722.
217 Nordyke K, Norstrom F, Lindholm L, et al. Health-related quality-of-life in children
with coeliac disease, measured prior to receiving their diagnosis through screening.
J Med Screen 2011;18:18792.
218 Johnston SD, Watson RG, McMillan SA, et al. Coeliac disease detected by
screening is not silentsimply unrecognized. QJM 1998;91:85360.
219 Rubio-Tapia A, Kyle RA, Kaplan EL, et al. Increased prevalence and mortality in
undiagnosed celiac disease. Gastroenterology 2009;137:8893.
220 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;9:11823.
221 Nachman F, Maurino E, Vazquez H, et al. Quality of life in celiac disease patients:
prospective analysis on the importance of clinical severity at diagnosis and the
impact of treatment. Dig Liver Dis 2009;41:1525.
222 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:
10513.
223 van Koppen EJ, Schweizer JJ, Csizmadia CG, et al. Long-term health and
quality-of-life consequences of mass screening for childhood celiac disease: a
10-year follow-up study. Pediatrics 2009;123:e5828.
224 Hin H, Bird G, Fisher P, et al. Coeliac disease in primary care: case nding study.
BMJ 1999;318:1647.
225 Marild K, Stephansson O, Grahnquist L, et al. Down syndrome is associated with
elevated risk of celiac disease: a nationwide case-control study. J Pediatr
2013;163:23742.
226 Vestergaard P, Mosekilde L. Fractures in patients with hyperthyroidism and
hypothyroidism: a nationwide follow-up study in 16,249 patients. Thyroid
2002;12:41119.
227 Halfdanarson TR, Litzow MR, Murray JA. Hematological manifestations of celiac
disease. Blood 2006;109:41221.
228 Ford AC, Chey WD, Talley NJ, et al. Yield of diagnostic tests for celiac disease in
individuals with symptoms suggestive of irritable bowel syndrome: systematic
review and meta-analysis. Arch Intern Med 2009;169:6518.
229 Green PHR, Stavropoulos SN, Panagi SG, et al. Characteristics of adult celiac
disease in the USA: results of a national survey. Am J Gastroenterol
2001;96:12631.
230 Ciacci C, Iavarone A, Siniscalchi M, et al. Psychological dimensions of celiac
disease: toward an integrated approach. Dig Dis Sci 2002;47:20827.
231 de Rosa A, Troncone A, Vacca M, et al. Characteristics and quality of illness
behaviour in celiac disease. Psychosomatics 2004;45:33642.
232 Hauser W, Gold J, Stein J, et al. Health-related quality of life in adult coeliac
disease in Germany: results of a national survey. Eur J Gastroenterol Hepatol
2006;18:74754.
233 Vilppula A, Kaukinen K, Luostarinen L, et al. Clinical benet of gluten-free
diet in screen-detected older celiac disease patients. BMC Gastroenterol
2011;11:136.
234 Smith DF, Gerdes LU. Meta-analysis on anxiety and depression in adult celiac
disease. Acta Psychiatr Scand 2012;125:18993.
235 Sanders DS, Patel D, Stephenson TJ, et al. A primary care cross-sectional study of
undiagnosed adult coeliac disease. Eur J Gastroenterol Hepatol 2003;15:
40713.
236 Platt SG, Kasarda DD. Separation and characterization of gliadin fractions.
Biochim Biophys Acta 1971;243:40715.
237 Janatuinen EK, Pikkarainen PH, Kemppainen TA, et al. A comparison of diets with
and without oats in adults with celiac disease. N Engl J Med 1995;333:10337.
238 Janatuinen EK, Kemppainen TA, Pikkarainen PH, et al. Lack of cellular and
humoral immunological responses to oats in adults with coeliac disease. Gut
2000;46:32731.
239 Sadiq Butt M, Tahir-Nadeem M, Khan MK, et al. Oat: unique among the cereals.
Eur J Nutr 2008;47:6879.
18 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
240 Comino I, Real A, de Lorenzo L, et al. Diversity in oat potential immunogenicity:
basis for the selection of oat varieties with no toxicity in coeliac disease. Gut
2011;60:91522.
241 Lundin KE, Nilsen EM, Scott HG, et al. Oats induced villous atrophy in coeliac
disease. Gut 2003;52:164952.
242 Arentz-Hansen H, Fleckenstein B, Molberg O, et al. The molecular basis for oat
intolerance in patients with celiac disease. PloS Med 2004;1:e1.
243 Case S. The gluten-free diet: how to provide effective education and resources.
Gastroenterology 2005;128:S12834.
244 Wild D, Robins GG, Burley VJ, et al. Evidence of high sugar intake, and low bre
and mineral intake, in the gluten-free diet. Aliment Pharmacol Ther
2010;32:57381.
245 Ukkola A, Maki M, Kurppa K, et al. Changes in body mass index on a gluten-free
diet in coeliac disease: a nationwide study. Eur J Intern Med 2012;23:3848.
246 Nelson M, Mendoza N, McGough N. A survey of provision of dietetic services for
coeliac disease in the UK. J Hum Nutr Diet 2007;20:40311.
247 Lefer D, Vanga R, Mukherjee R. Mild enteropathy celiac disease: a wolf in
sheeps clothing? Clin Gastroenterol Hepatol 2013;11:25961.
248 Akobeng AK, Thomas AG. Systematic review: tolerable amount of gluten for
people with coeliac disease. Aliment Pharmacol Ther 2008;27:104452.
249 Hischenhuber C, Crevel R, Jarry B, et al. Review article: safe amounts of gluten for
patients with wheat allergy or coeliac disease. Aliment Pharmacol Ther
2006;23:55975.
250 Ventura A, Magazzu G, Greco L. Duration of exposure to gluten and risk for
autoimmune disorders in patients with celiac disease. SIGEP Study Group for
Autoimmune Disorders in Celiac Disease. Gastroenterology 1999;117:297303.
251 Corrao G, Corazza GR, Bagnardi V, et al. Mortality in patients with coeliac disease
and their relatives: a cohort study. Lancet 2001;358:35661.
252 Khashan AS, Henriksen TB, Mortensen PB, et al. The impact of maternal celiac
disease on birthweight and preterm birth: a Danish population-based cohort study.
Hum Reprod 2010;25:52834.
253 Silano M, Volta U, Vincenzi AD, et al. Effect of a gluten-free diet on the risk of
enteropathy-associated T-cell lymphoma in celiac disease. Dig Dis Sci 2008;53:9726.
254 Green PH, Fleischauer AT, Bhagat G, et al. Risk of malignancy in patients with
celiac disease. Am J Med 2003;115:1915.
255 Olen O, Askling J, Ludvigsson JF, et al. Coeliac disease characteristics, compliance
to a gluten free diet and risk of lymphoma by subtype. Dig Liver Dis
2011;43:8628.
256 Karajeh MA, Hurlstone DP, Patel TM, et al. Chefs knowledge of coeliac disease
(compared to the public): a questionnaire survey from the United Kingdom. Clin
Nutr 2005;24:20610.
257 Singh J, Whelan K. Limited availability and higher cost of gluten-free foods. J Hum
Nutr Diet 2011;24:47986.
258 Lee AR, Ng DL, Zivin J, et al. Economic burden of a gluten-free diet. J Hum Nutr
Diet 2007;20:42330.
259 Drossman DA, Brandt LJ, Sears C, et al. A preliminary study of patients concerns
related to GI endoscopy. Am J Gastroenterol 1996;91:28791.
260 Ford S, Howard R, Oyebode J. Psychosocial aspects of coeliac disease: a
cross-sectional survey of a UK population. Br J Health Psychol 2012;17:74357.
261 Lee AR, Ng DL, Diamond B, et al. Living with coeliac disease: survey results from
the USA. J Hum Nutr Diet 2012;25:2338.
262 Bystrom IM, Hollen E, Falth-Magnusson K, et al. Health-related quality of life in
children and adolescents with celiac disease: from the perspectives of children and
parents. Gastroenterol Res Pract 2012;2012:986475.
263 Nachman F, del Campo MP, Gonzalez A, et al. Long-term deterioration of quality
of life in adult patients with celiac disease is associated with treatment
noncompliance. Dig Liver Dis 2010;42:68591.
264 Long KH, Rubio-Tapia A, Wagie AE, et al. The economics of coeliac disease: a
population-based study. Aliment Pharmacol Ther 2010;32:2619.
265 Lefer DA, Dennis M, Hyett B, et al. Etiologies and predictors of diagnosis in
nonresponsive celiac disease. Clin Gastroenterol Hepatol 2007;5:44550.
266 Abdulkarim AS, Burgart LJ, See J, et al. Etiology of nonresponsive celiac
disease: results of a systematic approach. Am J Gastroenterol 2002;97:201621.
267 Biagi F, Corazza GR. Dening gluten refractory enteropathy. Eur J Gastroenterol
Hepatol 2001;13:5615.
268 Fine KD, Meyer RL, Lee EL. The prevalence and causes of chronic diarrhea in
patients with celiac sprue treated with a gluten-free diet. Gastroenterology
1997;112:18308.
269 Dewar DH, Donnelly SC, McLaughlin SD, et al. Celiac disease: management of
persistent symptoms in patients on a gluten-free diet. World J Gastroenterol
2012;18:134856.
270 Dickey W. Colon neoplasia co-existing with coeliac disease in older patients:
coincidental, probably; important, certainly. Scand J Gastroenterol
2002;37:10546.
271 Leeds JS, Hopper AD, Hurlstone DP, et al. Is exocrine pancreatic insufciency in
adult coeliac disease a cause of persisting symptoms? Aliment Pharmacol Ther
2007;25:26571.
272 Olesen M, Eriksson S, Bohr J, et al. Microscopic colitis: a common diarrhoeal
disease. An epidemiological study in Orebro, Sweden, 19931998. Gut
2004;53:34650.
273 Cellier C, Delabesse E, Helmer C, et al. Refractory sprue, coeliac disease, and
enteropathy-associated T-cell lymphoma. French Coeliac Disease Study Group.
Lancet 2000;356:2038.
274 Rubio-Tapia A, Kelly DG, Lahr BD, et al. Clinical staging and survival in refractory
celiac disease: a single center experience. Gastroenterology 2009;136:99107;
quiz 3523.
275 Malamut G, Afchain P, Verkarre V, et al. Presentation and long-term follow-up of
refractory celiac disease: comparison of type I with type II. Gastroenterology
2009;136:8190.
276 Biagi F, Lorenzini P, Corazza GR. Literature review on the clinical relationship
between ulcerative jejunoileitis, coeliac disease, and enteropathy-associated T-cell.
Scand J Gastroenterol 2000;35:78590.
277 Goerres MS, Meijer JW, Wahab PJ, et al. Azathioprine and prednisone combination
therapy in refractory coeliac disease. Aliment Pharmacol Ther 2003;18:48794.
278 Maurino E, Niveloni S, Chernavsky A, et al. Azathioprine in refractory sprue: results
from a prospective, open-label study. Am J Gastroenterol 2002;97:2595602.
279 Gale J, Simmonds PD, Mead GM, et al. Enteropathy-type intestinal T-cell
lymphoma: clinical features and treatment of 31 patients in a single center. J Clin
Oncol 2000;18:795803.
280 Domizio P, Owen RA, Shepherd NA, et al. Primary lymphoma of the small intestine.
A clinicopathological study of 119 cases. Am J Surg Pathol 1993;17:42942.
281 Deleeuw RJ, Zettl A, Klinker E, et al. Whole-genome analysis and HLA genotyping
of enteropathy-type T-cell lymphoma reveals 2 distinct lymphoma subtypes.
Gastroenterology 2007;132:190211.
282 Kane EV, Newton R, Roman E. Non-Hodgkin lymphoma and gluten-sensitive
enteropathy: estimate of risk using meta-analyses. Cancer Causes Control
2011;22:143544.
283 Zettl A, deLeeuw R, Haralambieva E, et al. Enteropathy-type T-cell lymphoma. Am
J Clin Pathol 2007;127:7016.
284 Smedby KE, Akerman M, Hildebrand H, et al. Malignant lymphomas in coeliac
disease: evidence of increased risks for lymphoma types other than
enteropathy-type T cell lymphoma. Gut 2005;54:549.
285 Leslie LA, Lebwohl B, Neugut AI, et al. Incidence of lymphoproliferative disorders
in patients with celiac disease. Am J Hematol 2012;87:7549.
286 Olen O, Askling J, Ludvigsson JF, et al. Coeliac disease characteristics, compliance
to a gluten free diet and risk of lymphoma by subtype. Dig Liver Dis
2011;43:8628.
287 Moertel CG, Hargraves MM. Coexistence of adenocarcinoma of the jejunum and
nontropical sprue. JAMA 1961;176:6124.
288 Card TR, West J, Holmes GK. Risk of malignancy in diagnosed coeliac disease: a
24-year prospective, population-based, cohort study. Aliment Pharmacol Ther
2004;20:76975.
289 Elfstrom P, Granath F, Ye W, et al. Low risk of gastrointestinal cancer among
patients with celiac disease, inammation, or latent celiac disease. Clin
Gastroenterol Hepatol 2012;10:306.
290 McSorley HJ, Gaze S, Daveson J, et al. Suppression of inammatory immune
responses in celiac disease by experimental hookworm infection. PloS ONE
2011;6:e24092.
291 Daveson AJ, Jones DM, Gaze S, et al. Effect of hookworm infection on wheat
challenge in celiac diseasea randomised double-blinded placebo controlled trial.
PloS ONE 2011;6:e17366.
292 Brenchley R, Spannagl M, Pfeifer M, et al. Analysis of the bread wheat genome
using whole-genome shotgun sequencing. Nature 2012;491:70510.
293 Gil-Humanes J, Piston F, Tollefsen S, et al. Effective shutdown in the expression of
celiac disease-related wheat gliadin T-cell epitopes by RNA interference. Proc Natl
Acad Sci U S A 2010;107:170238.
294 Matysiak-Budnik T, Candalh C, Cellier C, et al. Limited efciency of
prolyl-endopeptidase in the detoxication of gliadin peptides in celiac disease.
Gastroenterology 2005;129:78696.
295 Mitea C, Havenaar R, Drijfhout JW, et al. Efcient degradation of gluten by a
prolyl endoprotease in a gastrointestinal model: implications for coeliac disease.
Gut 2008;57:2532.
296 Pyle GG, Paaso B, Anderson BE, et al. Effect of pretreatment of food gluten with
prolyl endopeptidase on gluten-induced malabsorption in celiac sprue. Clin
Gastroenterol Hepatol 2005;3:68794.
297 Gass J, Bethune MT, Siegel M, et al. Combination enzyme therapy for gastric
digestion of dietary gluten in patients with celiac sprue. Gastroenterology
2007;133:47280.
298 Tye-Din JA, Stewart JA, Dromey JA, et al. Comprehensive, quantitative mapping of
T cell epitopes in gluten in celiac disease. Sci Transl Med 2010;2:41ra51.
299 Gopalakrishnan S, Durai M, Kitchens K, et al. Larazotide acetate regulates
epithelial tight junctions in vitro and in vivo. Peptides 2012;35:8694.
300 Paterson BM, Lammers KM, Arrieta MC, et al. The safety, tolerance,
pharmacokinetic and pharmacodynamic effects of single doses of AT-1001 in
Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578 19
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
coeliac disease subjects: a proof of concept study. Aliment Pharmacol Ther
2007;26:75766.
301 Lefer DA, Kelly CP, Abdallah HZ, et al. A randomized, double-blind study of
larazotide acetate to prevent the activation of celiac disease during gluten
challenge. Am J Gastroenterol 2012;107:155462.
302 Hoffmann K, Alminger M, Andlid T, et al. Blocking peptides decrease tissue
transglutaminase processing of gliadin in vitro. J Agric Food Chem
2009;57:101505.
303 Stoven S, Murray JA, Marietta E. Celiac disease: advances in treatment via gluten
modication. Clin Gastroenterol Hepatol 2012;10:85962.
304 van der Windt DA, Jellema P, Mulder CJ, et al. Diagnostic testing for celiac disease
among patients with abdominal symptoms: a systematic review. JAMA
2010;303:173846.
305 Mooney PD, Evans KE, Singh S, et al. Treatment failure in coeliac disease: a
practical guide to investigation and treatment of non-responsive and refractory
coeliac disease. J Gastrointestin Liver Dis 2012;21:197203.
20 Ludvigsson JF, et al. Gut 2014;0:120. doi:10.1136/gutjnl-2013-306578
Guidelines
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
doi: 10.1136/gutjnl-2013-306578
published online June 10, 2014 Gut

Jonas F Ludvigsson, Julio C Bai, Federico Biagi, et al.

of Gastroenterology
disease: guidelines from the British Society
Diagnosis and management of adult coeliac
http://gut.bmj.com/content/early/2014/06/10/gutjnl-2013-306578.full.html
Updated information and services can be found at:
These include:
References
http://gut.bmj.com/content/early/2014/06/10/gutjnl-2013-306578.full.html#ref-list-1
This article cites 300 articles, 57 of which can be accessed free at:
Open Access
non-commercial. See: http://creativecommons.org/licenses/by-nc/3.0/
terms, provided the original work is properly cited and the use is
work non-commercially, and license their derivative works on different
license, which permits others to distribute, remix, adapt, build upon this
Creative Commons Attribution Non Commercial (CC BY-NC 3.0)
This is an Open Access article distributed in accordance with the
P<P
Published online June 10, 2014 in advance of the print journal.
service
Email alerting
the box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in
(DOIs) and date of initial publication.
publication. Citations to Advance online articles must include the digital object identifier
citable and establish publication priority; they are indexed by PubMed from initial
typeset, but have not not yet appeared in the paper journal. Advance online articles are
Advance online articles have been peer reviewed, accepted for publication, edited and
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from
Collections
Topic
(526 articles) Coeliac disease
(150 articles) Open access

Articles on similar topics can be found in the following collections


Notes
(DOIs) and date of initial publication.
publication. Citations to Advance online articles must include the digital object identifier
citable and establish publication priority; they are indexed by PubMed from initial
typeset, but have not not yet appeared in the paper journal. Advance online articles are
Advance online articles have been peer reviewed, accepted for publication, edited and
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
group.bmj.com on July 22, 2014 - Published by gut.bmj.com Downloaded from

You might also like