You are on page 1of 11

Human Pathology (2023) 132, 20e30

www.elsevier.com/locate/humpath

Review

Histologic evaluation in the diagnosis and


management of celiac disease: practical
challenges, current best practice
recommendations and beyond*
Zongming Eric Chen M.D, Ph.D, Hee Eun Lee M.D, Ph.D,
Tsung-Teh Wu M.D, Ph.D*
Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic,
Rochester, MN, 55905, USA

Received 22 June 2022; revised 13 July 2022; accepted 14 July 2022

Available online 4 August 2022

Keywords: Summary Celiac disease (CD) is an immunoallergic enteropathy affecting genetically susceptible in-
Celiac disease; dividuals upon dietary exposure to gluten. In current clinical practice, the diagnosis of CD is based
Ultrashort celiac disease; on a combination of clinical, serologic, and histologic factors with the possible exception of pediatric
Refractory celiac disease; patients. Histopathologic evaluation of small intestinal tissue plays a critical role in the disease diag-
Collagenous sprue; nosis and management, despite many practical challenges. Recently published best practice guidelines
Q-MARSH help to standardize biopsy sample procurement, tissue preparation, histology interpretation, and report-
ing, to optimize patient care. In addition, an increasing demand for monitoring the disease course,
particularly demonstrating the efficacy of dietary and nondietary interventions for disease manage-
ment, calls for the use of quantitative histology. With the advent of a gradual transition
toward digital pathology in routine diagnostic practice, quantitative histopathologic evaluation in
CD shows a promising future.
© 2022 Elsevier Inc. All rights reserved.

1. Introduction

Celiac disease (CD) is an immune-mediated enteropathy


occurring in genetically susceptible individuals after expo-
sure to gluten in their diets. Its prevalence is increasing [1]. A
*
Competing interests: None. recent meta-analysis study of epidemiologic surveys showed
* Corresponding author. Mayo Clinic, 200 First Street SW, Rochester, an estimated seroprevalence of 1.4% and biopsy-proven
MN, 55905, USA. prevalence of 0.7% [2]. Increased incidences occurred
E-mail address: wu.tsungteh@mayo.edu (T.-T. Wu).

https://doi.org/10.1016/j.humpath.2022.07.017
0046-8177/© 2022 Elsevier Inc. All rights reserved.
Diagnosis and management of celiac disease 21

globally, even in traditionally low incidence geographic re- sensitive and reproducible method necessary to address this
gions such as Asia [3]. Many contributing factors have been issue [12e14].
identified including the common use of gluten in food, In current clinical practice, the diagnosis of CD is based
increased disease awareness in the population, and improved on a combination of clinical, serologic, and histologic
detection methodology. There are also emerging data to factors, with one exception [15e17]. European pediatric
suggest other environmental factors may play a role, such as a guidelines suggest that, in a subset of children, duodenal
microbial impact on disease development [4]. Nevertheless, biopsies can be avoided in the presence of strict symp-
because of the variable clinical presentation of the disease, tomatic and serological criteria (serum IgA anti-tTG anti-
which includes many asymptomatic patients, and limited body 10 times or greater than the upper limit of normal,
accessibility to endoscopic biopsy and sensitive and specific positive anti-EMA, and the presence of HLA-DQ2/8) [17].
serologic tests in many regions of the world, underdiagnosis The no-biopsy approach, however, remains controversial.
and misdiagnosis of CD remains an important worldwide There are recent reports concerning the reliability of serum
challenge [5]. Currently, the only effective treatment for CD biomarker performance in the real world [18,19]. In addi-
is life-long gluten-free diet (GFD), which is costly and tion, the lack of direct assessment of mucosal injury makes
difficult to strictly follow. There is still an unmet clinical need histologic comparisons of before and after GFD treatment
for highly effective novel therapies [6]. impossible and may miss or delay the diagnosis of sero-
There have been great advances in understanding the negative or refractory CD (RCD) [20].
major immunopathogenetic mechanisms of CD [7,8]. The On the other hand, to make the best use of histologic
antigenic stimulators are a group of proteins collectively evaluation in CD diagnosis and management, there are
called gluten, including gliadins in wheat, hordeins in many challenging issues to consider, beyond the scope of
barley, and secalins in barley. They are rich in proline, the histology itself. A holistic approach is often necessary
which makes them resistant to gastric acid digestion. When to deal with perplexing clinical scenarios. Recently pub-
these proteins pass down into duodenum and permeate lished best practice guidelines help to standardize tissue
through the mucosa, they can become immunogenic, procurement, specimen preparation, and histologic inter-
particularly after their binding to tissue transglutaminase pretation, as well as reporting to optimize patient care [21].
(tTG) in the duodenal mucosa. This process releases These best practice principles are also mentioned in
deaminated peptides that can be presented by human recently published excellent review articles [22,23].
leukocyte antigen (HLA) to activate specific CD4þ T cells, The focus of the current review is to highlight these best
a critical step in initiating a cascade of innate and adaptive practice recommendations, considering the most frequently
immune responses. So far, HLA haplotypes of D2 (2.5), encountered practical challenges, particularly focusing on
D8, and rarely D7 are responsible for the genetic suscep- histologic variants of CD and their specific differential di-
tibility of CD [5]. The cytokines released from the initial agnoses. The use of quantitative histology is not recom-
immune reaction, particularly interleukin (IL)-15 and mended in the current best practice guidelines for several
interferon, are important in recruiting cytotoxic CD8þ T reasons, including a lack of definitive clinical value and the
cells (more gd subtype than ab subtype) into the epithe- process being too time-consuming for routine diagnostic
lium, causing enterocyte death and a breakdown of the practice. However, its value for disease management,
mucosal barrier, leading to escalating mucosal inflamma- including monitoring disease course and particularly in
tion and injury [9,10]. demonstrating the efficacy of dietary and nondietary in-
With recent advances in bioengineering capacity, it is terventions, has been clearly demonstrated [12]. With
now possible to synthesize specific HLA-DQ molecules to advent adoption of whole slide imaging technology and
form tetramer complexes with known peptides that cause digital imaging analysis applications into routine surgical
CD. These artificial molecular complexes are extremely pathology practice [24], quantitative histopathologic eval-
sensitive and specific to identify gluten-responsive CD4þ T uation in CD has a promising future. Therefore, also
cells in the blood or small intestinal mucosa of susceptible included is the Quantitative-Mucosal Algorithmic Rules for
individuals. As such, they have the potential to be a novel, Scoring Histology (Q-MARSH) [12] and potential chal-
noninvasive biomarker for CD and revolutionize the current lenges for its clinical adoption.
clinical diagnostic algorithm [11]. On the therapeutic front,
several emerging therapies targeting specific immunopa- 2. Practical challenges of tissue histologic
thogenic mechanisms of CD have shown promising results. evaluation for CD diagnosis
The number of clinical trials for CD novel therapies has
also grown significantly in the last few years [6]. However, The histologic changes of CD occur primarily in the
a common challenge for all of these trials is how to reliably small intestine. However, systemic presentations, such as
demonstrate therapeutic efficacy. Currently, quantitative dermatitis herpetiformis [25], lymphocytic and collagenous
histologic evaluation to demonstrate mucosal changes gastritis [26], and colitis [27], also occur and are beyond the
before and after therapeutic intervention appears the only scope of this review. The most characteristic histologic
22 Z.E. Chen et al.

change in small intestinal mucosa is increased intra-


Table 1 A list of common histologic differential diagnoses
epithelial lymphocytosis (IEL), which occurs in essentially of CD.
all affected individuals. However, it is also the most
Common and rare entities Common and rare entities
nonspecific histologic finding, one that may occur in many
show duodenal IEL and show duodenal mucosal
conditions unrelated to CD [28,29]. Mucosal architectural preserved villous atrophy with or without IEL
changes, including villous blunting and crypt hyperplasia, architecture
are also characteristic findings, representing a dynamic
 Infectious etiologies  Tropical sprue
balance between mucosal injury and regeneration. These
including H. pylori  Collagenous sprue
changes are almost always associated with increased lam- gastritis, bacterial over-  Refractory CD
ina propria lymphoplasmacytic infiltration. In fact, the growth syndrome, giardia,  Whipple’s disease
absence of plasma cells can indicate common variable cryptosporidiosis, and viral  Mycobacterial infection
immunodeficiency (CVID) [30]. Cryptal architectural infection including HIV  Drugs including angio-
destruction, with or without missing normal cell compo-  Drugs including NSAIDs tensin 2 receptor blockers
nents such as goblet cells and endocrine cells, almost never  Food hypersensitivity such as Olmesartan and
occurs in CD and should lead to evaluation for autoimmune including Cow’s milk, soy, immune checkpoint inhibi-
diseases (including autoimmune enteropathy and inflam- etc. tor such as Nivolumab
matory bowel disease) or drug-induced injury [31e34].  Peptic duodenitis  Autoimmune enteropathy
 Immunodeficiency  GVHD
Frequently, neutrophils and eosinophils are also present in
including CVID, IgA  Crohn’s disease
the inflammatory milieu of the lamina propria with CD, and
deficiency  Eosinophilic enteritis
rarely neutrophilic or eosinophilic cryptitis may also exist,  Systemic autoimmune dis-  Microvillous inclusion
particularly in cases with significant mucosal flattening. ease including rheumatoid disease
However, the presence of significant neutrophilic or arthritis, SLE, Hashimoto  Tufting enteropathy
eosinophilic cryptitis with crypt destruction is rare, and thyroiditis  Nutritional deficiency
when present should be considered as a potential red flag  Chemotherapy
for nonceliac injury [23]. Other common but nonspecific  Lymphoma
cytologic features in CD include enterocyte vacuolar Abbreviations: CD, celiac disease; IEL, intraepithelial lymphocytosis;
changes, focal gastric foveolar metaplasia, and slightly HIV, human immunodeficiency virus; NSAID, nonsteroidal anti-
increased apoptosis in crypt cells [35]. Markedly increased inflammatory drug; CVID, common variable immunodeficiency;
crypt cell apoptosis is rare for CD, however, and should SLE, systemic lupus erythema; GVHD, graft-versus-host disease.
lead to exclusion of drug-induced injury or graft-versus-
host disease [36].
As already noted, the characteristic histologic alterations ctly taking care of the patients are necessary. For example, a
seen in CD are not pathognomonic for CD. Other etiologies CD patient may also suffer from Crohn’s disease and/or H.
can cause similar mucosal histologic changes, including pylori gastritis. These challenging clinical situations require
infection, autoimmune enteropathy, drug effect, and immu- close clinical follow-up and monitoring and may require
nodeficiency. For example, Helicobacter pylori gastritis may further tissue sampling to assess histologic changes during
induce significant IEL in the duodenum mucosa, mimicking the disease course, such as after GFD or other relevant
CD. Some drugs, particularly angiotensin 2 receptor blockers treatments.
such as Olmesartan, are known to induce IEL with villous The histological changes of CD are often unevenly
atrophy, mimicking severe active CD [31]. More recently, distributed and of variable severity [38]. Limited tissue
immune checkpoint inhibitors (nivolumab, ipilimumab, and sampling, along with suboptimal orientation, is an important
pembrolizumab) have been reported to cause duodenal injury practical challenge for histologic evaluation. The misinter-
that can share similarities with severe active CD [37]. To pretation of mucosal injury patterns may occur when tissue
differentiate CD from these morphologic mimickers can be a sampling is limited, resulting in underdiagnosis or a missed
major challenge in some cases. Table 1 summarizes common diagnosis [39,40]. The recently described CD patterns of
differential diagnoses that need to be considered when duodenal bulb-only or bulb-predominant CD injury are seen
evaluating tissue histology for CD. These entities can be in a subset of patients and clearly illustrate the clinical
roughly divided into 2 categories based on the predominant importance of obtaining bulb tissue for evaluation (see
histologic patterns, ie, (1) IEL with preserved villous archi- detailed discussion below). Tissue fragmentation and crush
tecture, and (2) villous atrophy with or without IEL. In most artifact secondary to endoscopic biopsy handling techniques
cases, with proper clinical information including medication can also severely impair the pathologists’ ability to discern
use and CD serologic testing results, pathologists can histologic details and make an accurate diagnosis. These
reasonably confidently make an accurate diagnosis. How- preanalytical variables have a direct impact on proper his-
ever, for complicated clinical scenarios, such as when mul- tologic evaluation and its outcome.
tiple disorders may coexist in individual patients, close How to properly apply CD histologic classification
communications between pathologists and clinicians dire- grading schemes is another practical challenge. Since the
Diagnosis and management of celiac disease 23

original Marsh criteria proposed in 1990s [41], there have is also recommended to have one biopsy per pass as the
been several classification grading schemes developed standard endoscopic tissue sampling technique. Impor-
through years, with the basic purpose of capturing the dy- tantly, it is formally recommended to have clinicians to
namic mucosal changes of CD and to semiquantitatively supply relevant clinical information when submitting tissue
document disease status and activity. Table 2 summarizes for histologic evaluation. This is extremely helpful in set-
commonly used classification grading schemes. Currently, tings where a pathologist’s access to electronic medical
the most popular grading scheme is the so-called Marsh- records is limited.
Oberhuber system [42]. It is a semiquantitative descrip- Tissue orientation is a major preanalytical factor impacting
tion of IEL and villous atrophy, with or without crypt hy- proper histologic evaluation and applying CD classification
perplasia. The classification relies on subjective judgment grading schemes. It was recognized that, although there are
based on global assessment. The interobserver agreement is insufficient data to recommend special orientation efforts dur-
generally low and the reproducibility poor. To simplify the ing tissue preparation and embedding, using serial sectioning
grading method, Corazza-Villanacci developed a 3-grade to obtain well-oriented villus-crypt units is still recommended.
system: grade A shows normal crypts and villus architec- Identifying reasonable numbers of well-oriented villus-crypt
ture but increase IEL (>25 IELs per 100 enterocytes); units is also a key determinant for successful quantitative
grade B1 shows villous atrophy but clearly detectable villi tissue evaluation (see more discussion below).
are still present, whereas grade B2 shows villous atrophy to For histologic analysis, the recommendations also pro-
the extent of complete flattening, with no detectable villi vide helpful standardization. For counting IELs, the recom-
[43]. This simplified system showed better agreement be- mendations recognize several practical scenarios: (1) A
tween pathologists and is commonly used in clinical normal or “non-CD” pattern with occasional IEL, up to 25/
practice. However, the subjectivity of individual patholo- 100 enterocytes, with a distribution skewed toward the lateral
gists at the time of tissue evaluation still exists. aspects of the villi and decrease lymphocytes toward the
villous tips, the so-called “decrescendo pattern”; (2) IEL in
3. Current best practice recommendations for CD patients with normal villi, showing >25/100 enterocytes
histopathology evaluation in CD with an even distribution over the entire villous or more
numerous lymphocytes in the villous tips (>6/20 enter-
To address and overcome these practical challenges, ocytes); (3) The number of IEL in CD patients with abnormal
experts from the Rodger C. Haggitt Gastrointestinal Pa- villi almost always exceeds 40/100 enterocytes. It is also
thology society and the North American Association for the formally recognized that routinely applying CD3 immuno-
Study of Celiac Disease recently published a set of best staining for IEL counting is not necessary [44], and therefore
practice recommendations [21]. These help to standardize discouraged. The recommendations also call for increased
preanalytical and analytical factors impacting tissue histo- awareness of unusual histologic changes that may indicate
logic evaluation for CD. Table 3 summarizes the key rec- non-CD etiology during histologic evaluation. In addition,
ommendations. It is formally recommended to obtain care must be taken to avoid overinterpreting villous abnor-
adequate biopsy specimens when CD is considered to be malities in areas of Brunner gland hyperplasia or lympho-
clinically possible, regardless of the endoscopic mucosal glandular complexes.
appearance. The biopsy series should contain at least 2 These recommendations also clearly recognize the clin-
biopsies from the duodenal bulb and 4 biopsies from the ical necessity of comparing mucosal histologic changes with
distal (postbulbar) duodenum. To ensure tissue integrity, it previous biopsies for certain indications, and the utility of

Table 2 Comparing different histologic classification grading schemes used for celiac disease.
Marsh Marsh Corazza QMARSH
-Oberhuber -Villanacci
IEL Mucosal Architectural Vh:Cd ratio
Abnormality
Normal Normal 0 0 Normal 3.0
Increased Normal 1 1 Grade A 3.0
Increased Normal villous height 2 2.0 to <3.0
but increased crypt depth
Increased Partial villous atrophy 3 3a Grade B1 1.0 to <2.0
Increased Subtotal villous atrophy 3b 0.5 to <1.0
Increased Total villous atrophy 3c Grade B2 <0.5
Increased Hypoplastic 4
Abbreviations: IEL, intraepithelial lymphocytosis; Vh, villous height; Cd, crypt depth.
24 Z.E. Chen et al.

Table 3 A summary of GISP-NAASCD best practice recommendations.


Tissue procurement and Tissue preparation and Histologic evaluation Pathology reporting
clinical information orientation
submission
Recommendations  Taking biopsy regardless  Special effort at tissue  Occasional IEL (up to  Pathology reports should
of endoscopic appearance orientation in the labo- 25/100 enterocytes) are mention semi-
 At least 4 specimens from ratory are not required present in non-CD quantitatively the degree
the distal (postbulbar) for the diagnosis of ce- samples with a of villous blunting and
duodenum and 2 speci- liac disease “decrescendo pattern” should compare the
mens from the duodenal  Adequate numbers of (more numerous along villous architecture with
bulb biopsies, along with the the lateral aspects of villi existing previous bi-
 Obtaining one specimen appropriate use of serial and decrease toward the opsies if clinically indi-
per pass of the biopsy sectioning typically villous tips) cated in patients with
forceps result in a sufficient  IEL in CD samples are suspected or proven CD
 Supply relevant clinical number of well-oriented evenly distributed over  A named classification
information including villus crypt units to the entire villous (>25/ system score may be
signs and symptoms, accurately determine 100 enterocytes) or are included in pathology
endoscopic findings, architecture in the more numerous in the reports if it is understood
medications, patient and majority of cases with villous tips (>6/20 by, and enhances
family history, current randomly embedded enterocytes); may communication with
adherence to GFD, and tissue prompt additional sero- clinicians
serological or genetic test logic studies if they have  No specific system is
results not already been carried endorsed as superior
out.
 Immunohistochemical
stains for T-lymphocyte
markers do not improve
the detection of CD; “up
front” ordering of
immunohistochemical
stains not recommended
 Histologic changes are
sensitive but not specific
for CD; differential
diagnosis should be
clinically and histologi-
cally excluded
 Avoid overinterpreting
villous abnormalities in
areas of Brunner gland
hyperplasia or lympho-
glandular complexes
Abbreviations: CD, celiac disease; IEL, intraepithelial lymphocytosis; GFD, gluten-free diet.

including semiquantitative descriptions of villous architec- 4. Histologic variants of CD


ture in pathology reports. Although there is no endorsement
for a specific CD classification grading scheme, the appro- 4.1. CD restrict to duodenal bulb only or ultrashort
priate use is encouraged and contingent to the clinical setting CD
where the scheme enhances communication with clinicians.
The recommendations also provide standardized pathology As noted earlier, it is not unusual to see patchy mucosal
report templates for CD to illustrate the optimal delivery of injury with variable severity in CD patients. Patchy disease is
the most valuable histologic information. Pathology reports more frequent in children than in adults [38,45,46]. At the
that incorporate clinical and pathologic data personalized to extreme end of the spectrum, some CD patients only show
individual patients provide the best value for treating clini- mucosal inflammation and injury, particularly villous atro-
cians, rather than pathology reports that blindly include every phy, within the duodenal bulb (D1 region), without more
possible cause of mucosal histologic changes a pathologist distal involvement (Fig. 1). Several prospective studies
can think of [22]. confirmed the existence of such patients and illustrate the
Diagnosis and management of celiac disease 25

Fig. 1 An example of ultrashort celiac disease (USCD). A, Localized typical histologic changes of celiac disease (CD) in duodenal bulb
with increased intraepithelial lymphocytosis (IEL), partial villous atrophy, and crypt hyperplasia with increased lamina propria inflam-
mation. B, Normal histology in the second portion of duodenum biopsied during the same procedure.

importance of including duodenal bulb tissue sampling to 4.2. Refractory CD


ensure their detection [39,40]. According to a recent meta-
analysis, in 2.5e30% of CD cases, inflammation is restricted RCD is a rare complication of CD and an important
to the duodenal bulb [47] and the study found that duodenal clinicopathologic diagnosis [52,53]. The current definition is
bulb biopsies improved the rate of detection of CD by 4% in that of persistent or recurrent malabsorption and continued
children and by 8% in adults. Therefore, all published society villous atrophy on duodenal biopsies, despite strict avoid-
guidelines currently recommend examination of bulb bi- ance of gluten for a minimum of 12 months, in the absence of
opsies. However, the effectiveness of screening bulb tissue to other causes of nonresponsive treated CD and overt lym-
improve the detection of CD in some settings is still phoma. RCD is further divided into 2 subtypes: RCD type 1
controversial. A study by Stoven et al., for example, analyzed shows IELs without an abnormal T cell phenotype, and RCD
biopsies from duodenal bulbs in a cohort of patients with a type 2 shows IELs with an abnormal T cell phenotype, or a
low pretest probability for CD and found only 1 of 679 in- monoclonal population of T cells. The distinction between
dividuals had CD disease restricted to bulb [48]. According the 2 types is based on clinical, histologic, and molecular
to this study, there was only 0.1% improvement in CD factors [52,53]. The diagnosis is of paramount importance
detection in the setting of low pretest probability. Neverthe- because the treatment options and prognosis differ signifi-
less, the practical importance for pathologists is to pay close cantly, as RCD type 2 is a severe complication that is
attention to CD-related mucosal histologic changes that are considered to be a precursor of T cells lymphoma, with
limited to duodenal bulb, because they may be obscured by 32e52% of patients transforming to enteropathy-associated
histologic changes unrelated to CD, such as peptic duode- T cells lymphoma in 5 years [53]. It is critical for pathologists
nitis, Brunner gland hyperplasia, and prominent lymphoid to perform appropriate tests and molecular workup to help
follicles. In general, marked IEL in bulb biopsies with or with identify these patients at an early stage.
villous atrophy should always be considered as potentially According to a recent systematic review [54], among CD
representing CD, regardless of the other background fea- patients, RCD has a cumulative incidence of 1e4% over a
tures. 10-year period and a prevalence of 0.31e0.38%. It usually
This D1-only presentation of CD is also referred to as presents in 2 clinical scenarios: primary RCD patients form a
ultrashort CD (USCD) [49e51]. There are only limited minority group who never respond to GFD therapy after the
studies focusing on this topic. From these studies, the initial CD diagnosis, while secondary RCD patients make up
prevalence of USCD ranges from 5 to 10% in both chil- the majority of cases and are patients who responded to GFD
dren and adults. The patients often show milder symp- for years but then subsequently developed new symptoms or
toms, less endoscopic findings, milder histologic abno- recurrence of diarrhea. For a primary RCD diagnosis, it
rmalities, and lower serum IgA-tTG titers, when compared should be noted that the 12-month should not be a rigid time
to cases of conventional CD. They also tend to respond frame because there are slow-responders who might need a
faster to GFD with a complete normalization of mucosal longer period on a GFD to achieve mucosal healing. In one
injury. USCD is generally regarded as a milder phenotype series [55], persistent villous atrophy was found in 42% of
of CD. Interestingly, a recent study showed a lower fre- patients at 1 year but that reduced to 5% over 2e5 years of
quency of HLA DQ2 and less NK cell suppression in follow-up. Continued “gluten” exposure is one of the main
USCD patients [49], elucidating potential distinct genetic causes for the slow responsiveness to GFD [52]. CD sero-
and immunologic features. Currently, few data exist logic tests are helpful to investigate this possibility. Other
regarding the long-term disease course and prognosis of nonceliac etiologies of mucosal injury should also be clini-
USCD. cally excluded.
26 Z.E. Chen et al.

The most important practical challenge for pathologists IHC, normal IELs in RCD type 1 show a CD3þ/CD8þ
is to distinguish RCD type 1 from type 2. The histologic phenotype, whereas abnormal IELs in RCD type 2 show
features of RCD type 1 are the same as active CD, whereas CD3epsilonþ/CD8-phenotype (Fig. 2). The abnormal IELs
RCD type 2 is characterized by the presence of abnormal T in RCD type 2 also exhibit clonal TCR gene rearrange-
cells that do not express surface CD3 and CD8 molecules ments. However, the sensitivity and specificity of these tests
but retain intracellular CD3 expression. They also express in the diagnosis of RCD type 2 should be critically evalu-
NKp46 and IL15 receptors and show clonal T cell receptor ated. One recent study [58] found that clonal T cells could
(TCR) gene rearrangements. According to a recent study be seen in biopsies from several benign conditions with
[56], these cells are believed to derive from a subset of T IELs, and similar clonal populations were identified in both
cells in the normal mucosa and are activated to expand by RCD type 1 and type 2 patients, whereas no clonal T cells
IL15 and gain of function somatic mutations within Jak1 were detected in one RCD type 2 patient. The same study
and Stat3. Increased levels of Smad7 protein also contribute also found that IHC for CD3/CD8 ratio was not sensitive to
to the expansion of these abnormal T cells by sustaining reliably differentiate RCD type 1 from type 2. The potential
and amplifying the inflammatory cytokine effect [57]. pitfalls in relying on TCR clonal analysis alone in the
Currently, immunohistochemistry (IHC) and TCR gene diagnosis of RCD type 2 have also been highlighted by
rearrangement analysis are 2 of the most used ancillary others [59]. In comparison with IHC and TCR clonal
tests in clinical practice in the United States. Typically, by analysis methods, flow cytometry appears to show superior

Fig. 2 Examples of refractory CD type 1 (AeC) and type 2 (DeF). Hematoxylin and eosin (H&E) stains show increased IEL in A and D,
with partial villous atrophy in A but not D. CD3 immunostains show positivity in most IEL and T cells in lamina propria in both B and E.
CD8 immunostains show retained CD8 expression in most IELs in refractory CD type 1 as shown in C, but loss of CD8 expression in
refractory CD type 2 as shown in F.
Diagnosis and management of celiac disease 27

sensitivity and specificity in detecting RCD patients, (ARBs), ie, sartan family medications such as Olmesartan,
particularly with moderately increased number of abnormal can also induce the development of CS [67]. An accurate
IELs [60]. This method has been used clinically to identify diagnosis of CS is important for patient management and
abnormal IELs in duodenal mucosa from RCD type 2 pa- treatment [66,68,69].
tients and is more commonly performed in Europe than in
the United States. It is prudent for pathologists to under- 5. Quantitative histology for clinical
stand different methods and their analytical performance management of CD
characteristics to put them into best use for the clinical
diagnosis of RCD patients. CD is a chronic inflammatory process and clinical disease
management often requires a close monitoring of the disease
4.3. Collagenous sprue course and particularly demonstrating the efficacy of dietary
and nondietary interventions. The current best practice
This is an entity with characteristic morphologic features guidelines recognize this need to compare mucosal histo-
[61,62]. The subepithelial collagen layer of the duodenum is logic changes with previous biopsies in certain indications
prominently thickened, with a thickness often exceeding and recommend including semiquantitative descriptions of
10 mm, and shows entrapment and dilatation of capillaries villous architecture in the pathology reports. However,
(Fig. 3). The collagen deposition can be diffuse or patchy. subjectivity and reproducibility are major limitations of the
Typically, there is associated villus blunting, lamina propria semiquantitative approach. The Quantitative-Mucosal
inflammation including increased eosinophils, and a variable Algorithmic Rules for Scoring Histology (Q-MARSH) sys-
increase in the number of IELs. Surface epithelial detach- tem (Table 2) uses an averaged assessment of villous height
ment is quite common and the presence of acute inflamma- (Vh), crypt depth (Cd), and IEL count (per 100 enterocytes)
tion within the surface epithelium and crypts is also to provide objective measures of histological changes [12]. It
frequently seen. These features are pathognomonic and has been mainly used as a research tool and was successfully
essentially the same as seen in collagenous gastritis or colitis applied in several recent clinical trials of CD, some of which
[63]. In fact, these entities often coexist and, when this is the showed promising results [13,14,70].
case, may indicate a systemic gastrointestinal tract collage- The proposed rules in Q-MARSH require measurements
nosis. An increased expression of fibrogenic genes, such as of Vh and Cd from 3 to 5 well-orientated villusecrypt units
procollagen1 and tissue inhibitor of MMP-1 (TIMP-1), by (deeper levels may be required to achieve this) from at least
myofibroblastic cells is thought to be responsible for the 4 postbulbar duodenum biopsies. This helps to average out
abnormal collagen deposition [64]. the variability in disease severity. A Vh/Cd ratio of <3.0
Collagenous sprue (CS) occurs in women more than men, and an IEL count of 30/100 enterocytes are abnormal. A
with a 2:1 ratio, and typically affects middle-aged or elderly comparison of Q-MARSH with the other commonly used
women who present with severe diarrhea and weight loss CD semiquantitative classification grading systems is
[65,66]. There is a close association with CD, and CS is seen shown in Table 2. It should be noted that Vh:Cd ratio in Q-
with 30e50% of all RCD cases. However, it is important to MARSH does not discriminate between crypt elongation
know that CD is not the only cause of CS. There are multiple and shortening of the villi. In general, Vh:Cd ratio of 2.5
other potential causes, including autoimmune enteropathy, indicates normalization of mucosal injury.
CVID, hypothyroidism, IgG4-related sclerosing disease, and To apply Q-MARSH well, there are 2 critical steps: to
idiopathic collagenosis involving gastrointestinal tract [62]. identify enough well-orientated villus-crypt units and to
It is worth noting that angiotensin 2 receptor blockers determine the boundary between a villus and a crypt. A well-

Fig. 3 An example of collagenous sprue. A, H&E stain shows total villous atrophy and slightly increased IEL. There is thickened
subepithelial collagen deposition (arrowheads) with entrapped capillaries and inflammatory cells. Note the detachment of epithelium from
the collagen table. B, Trichrome stain highlights subepithelial collagen deposition.
28 Z.E. Chen et al.

orientated villus-crypt unit is defined as a continuous cells shows a crispy texture, whereas it is absent or becomes
epithelial lining that is visible from the tip of the villous fuzzy on crypt cells. Recently, IHC for apolipoprotein A4
structure to the bottom of the crypt where it touches the (APOA4) has been shown to help define the villus-crypt
muscularis mucosae (Fig. 4). To find enough numbers of such border because it highlights the brush border of the villous
units, serial sectioning is the most reliable method and is cells but not the crypt cells [71]. The study demonstrated an
almost always required. There is no standard approach to improved interobserver variability of Vh and Cd measure-
determine the villus and crypt boundary. Some use the nar- ments by applying APO A4 IHC in challenging cases. Fig. 4
rowest point where a crypt opens to the surface as the illustrates how the measurement of Vh and Cd are performed
boundary, whereas others draw a virtual boundary line based on a well-orientated villus-crypt unit using a digitally scan-
on inspection of the overall mucosal topology of a specific ned whole slide image. The figure also shows that angulation
tissue fragment. Both are subjective assessments and tend to in the villi can be incorporated by measurement around the
work well for measuring Vh and Cd of healthy mucosa with angles. Essentially all digital pathology platforms have
slender villi and shallow crypts but face significant chal- specific annotation or measurement tools which make Vh and
lenges when there is partial villous atrophy and crypt hy- Cd assessment simple and quick. With a little effort, specific
perplasia. A more subjective way to determine the boundary applications can be developed to automatically calculate
is to carefully examine the brush border on enterocytes lining Vh:Cd ratio and export data to pathology reports. IELs can
the villi and crypts. The brush border associated with villous also be counted automatically by running commercially
available or self-brew lymphocyte enumeration algorithms.
These potential technical improvements, accompanied by
implantation of routine digital pathology workflows, would
make CD quantitative histology evaluation a real feasibility
in the future.

6. Conclusion
Tissue histologic evaluation plays a critical role in the
diagnosis and management of CD. There are many prac-
tical challenges, and the recent best practice recommen-
dations help to standardize some important pre-analytical
and analytical variants to optimize patient care. Patholo-
gists should be aware of histologic mimickers and variants
of CD to make an accurate diagnosis. With the advent of
transitioning into the digital pathology era, there is a po-
tential to adopt quantitative histologic evaluation of CD
into routine practice.

References
[1] King JA, Jeong J, Underwood FE, et al. Incidence of celiac disease is
increasing over time: a systematic review and meta-analysis. Am J
Gastroenterol 2020;115:507e25.
[2] Ludvigsson JF, Murray JA. Epidemiology of celiac disease. Gastro-
enterol Clin North Am 2019;48:1e18.
[3] Singh P, Arora S, Singh A, Strand TA, Makharia GK. Prevalence of
celiac disease in Asia: a systematic review and meta-analysis. J
Gastroenterol Hepatol 2016;31:1095e101.
[4] Lindfors K, Lin J, Lee HS, et al. Metagenomics of the faecal virome
indicate a cumulative effect of enterovirus and gluten amount on the
Fig. 4 An illustration of villous height (Vh; blue lines) and risk of coeliac disease autoimmunity in genetically at risk children:
crypt depth (Cd; red lines) measurements (in mm depicted in the the TEDDY study. Gut 2020;69:1416e22.
rectangular boxes) in 2 well-orientated villus-crypt units using a [5] Pinto-Sanchez MI, Silvester JA, Lebwohl B, et al. Society for the
standard digital pathology platform deployed for routine diag- Study of Celiac Disease position statement on gaps and opportunities
in coeliac disease. Nat Rev Gastroenterol Hepatol 2021;18:875e84.
nostic use at Mayo Clinic. Note the annotations and measurements
[6] Kivela L, Caminero A, Leffler DA, Pinto-Sanchez MI, Tye-Din JA,
made directly on the tissue section. The V/C boundary is deter- Lindfors K. Current and emerging therapies for coeliac disease. Nat
mined by subjective assessment according to the pathologist’s Rev Gastroenterol Hepatol 2021;18:181e95.
experience. (For interpretation of the references to color in this [7] Abadie V, Sollid LM, Barreiro LB, Jabri B. Integration of genetic and
figure legend, the reader is referred to the Web version of this immunological insights into a model of celiac disease pathogenesis.
article). Annu Rev Immunol 2011;29:493e525.
Diagnosis and management of celiac disease 29

[8] Caio G, Volta U, Sapone A, et al. Celiac disease: a comprehensive intestinal mucosa: an increasing diagnostic problem with a wide
current review. BMC Med 2019;17:142. differential diagnosis. Arch Pathol Lab Med 2006;130:1020e5.
[9] Abadie V, Kim SM, Lejeune T, et al. IL-15, gluten and HLA-DQ8 [30] Agarwal S, Mayer L. Diagnosis and treatment of gastrointestinal
drive tissue destruction in coeliac disease. Nature 2020;578:600e4. disorders in patients with primary immunodeficiency. Clin Gastro-
[10] Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis enterol Hepatol 2013;11:1050e63.
to novel therapies. Gastroenterology 2009;137:1912e33. [31] Rubio-Tapia A, Herman ML, Ludvigsson JF, et al. Severe spruelike
[11] Sarna VK, Lundin KEA, Morkrid L, Qiao SW, Sollid LM, enteropathy associated with olmesartan. Mayo Clin Proc 2012;87:
Christophersen A. HLA-DQ-Gluten tetramer blood test accurately 732e8.
Identifies patients with and without celiac disease in absence of [32] Gentile NM, D’Souza A, Fujii LL, Wu TT, Murray JA. Association
gluten Consumption. Gastroenterology 2018;154:886e896 e6. between ipilimumab and celiac disease. Mayo Clin Proc 2013;88:
[12] Adelman DC, Murray J, Wu TT, Maki M, Green PH, Kelly CP. 414e7.
Measuring change in small intestinal histology in patients with celiac [33] Masia R, Peyton S, Lauwers GY, Brown I. Gastrointestinal biopsy
disease. Am J Gastroenterol 2018;113:339e47. findings of autoimmune enteropathy: a review of 25 cases. Am J Surg
[13] Kelly CP, Murray JA, Leffler DA, et al. TAK-101 Nanoparticles Pathol 2014;38:1319e29.
induce gluten-specific tolerance in celiac disease: a randomized, [34] Lee HE, Yuan L, Wu TT. Neuroendocrine cells are commonly absent
double-blind, placebo-Controlled study. Gastroenterology 2021;161: in the intestinal crypts in autoimmune enteropathy. Am J Surg Pathol
66e80 e8. 2020;44:1130e6.
[14] Schuppan D, Maki M, Lundin KEA, et al. A randomized trial of a [35] Shalimar DM, Das P, Sreenivas V, Gupta SD, Panda SK,
transglutaminase 2 inhibitor for celiac disease. N Engl J Med 2021; Makharia GK. Mechanism of villous atrophy in celiac disease: role of
385:35e45. apoptosis and epithelial regeneration. Arch Pathol Lab Med 2013;
[15] Rubio-Tapia A, Hill ID, Kelly CP, Calderwood AH, Murray JA, 137:1262e9.
American College of G. ACG clinical guidelines: diagnosis and [36] Matsuda K, Ono S, Tanaka I, et al. Histological and magnified
management of celiac disease. Am J Gastroenterol 2013;108: endoscopic evaluation of villous atrophy in gastrointestinal graft-
656e76. quiz 77. versus-host disease. Ann Hematol 2020;99:1121e8.
[16] Downey L, Houten R, Murch S, Longson D, Guideline [37] Irshaid L, Robert ME, Zhang X. Immune checkpoint inhibitor-
Development G. Recognition, assessment, and management of induced upper gastrointestinal tract inflammation shows morpho-
coeliac disease: summary of updated NICE guidance. BMJ 2015;351: logic similarities to, but is Immunologically distinct from, Heli-
h4513. cobacter pylori gastritis and celiac disease. Arch Pathol Lab Med
[17] Husby S, Koletzko S, Korponay-Szabo I, et al. European society 2021;145:191e200.
paediatric gastroenterology, hepatology and Nutrition guidelines for [38] Ravelli A, Villanacci V, Monfredini C, Martinazzi S, Grassi V,
diagnosing coeliac disease 2020. J Pediatr Gastroenterol Nutr 2020; Manenti S. How patchy is patchy villous atrophy?: distribution
70:141e56. pattern of histological lesions in the duodenum of children with ce-
[18] Stefanelli G, Navisse S, Valvano M, et al. Serum transglutaminase liac disease. Am J Gastroenterol 2010;105:2103e10.
antibodies do not always detect the persistent villous atrophy in pa- [39] Gonzalez S, Gupta A, Cheng J, et al. Prospective study of the role of
tients with celiac disease on a gluten-free diet. Eur J Gastroenterol duodenal bulb biopsies in the diagnosis of celiac disease. Gastrointest
Hepatol 2021;33:e650e5. Endosc 2010;72:758e65.
[19] Donat E, Roca M, Masip E, Polo B, Ramos D, Ribes-Koninckx C. [40] Evans KE, Aziz I, Cross SS, et al. A prospective study of duodenal
Screening-detected positive serology for celiac disease: a real clinical bulb biopsy in newly diagnosed and established adult celiac disease.
challenge. Eur J Gastroenterol Hepatol 2021;33:e999e1002. Am J Gastroenterol 2011;106:1837e42.
[20] Corazza GR, Lenti MV. Intestinal biopsy, coeliac disease and resis- [41] Marsh MN. Gluten, major histocompatibility complex, and the small
tance to change. Eur J Gastroenterol Hepatol 2021;33:e31e2. intestine. A molecular and immunobiologic approach to the spectrum of
[21] Robert ME, Crowe SE, Burgart L, et al. Statement on best practices gluten sensitivity (’celiac sprue’). Gastroenterology 1992;102:330e54.
in the use of pathology as a diagnostic tool for celiac disease: a guide [42] Oberhuber G. Histopathology of celiac disease. Biomed Pharmac-
for clinicians and pathologists. Am J Surg Pathol 2018;42:e44e58. other 2000;54:368e72.
[22] Lagana SM, Bhagat G. Biopsy diagnosis of celiac disease: the pa- [43] Corazza GR, Villanacci V, Zambelli C, et al. Comparison of the
thologist’s perspective in light of recent advances. Gastroenterol Clin interobserver reproducibility with different histologic criteria used in
North Am 2019;48:39e51. celiac disease. Clin Gastroenterol Hepatol 2007;5:838e43.
[23] Brown I, Bettington M, Rosty C. The role of histopathology in the [44] Hudacko R, Kathy Zhou X, Yantiss RK. Immunohistochemical stains
diagnosis and management of coeliac disease and other mal- for CD3 and CD8 do not improve detection of gluten-sensitive en-
absorptive conditions. Histopathology 2021;78:88e105. teropathy in duodenal biopsies. Mod Pathol 2013;26:1241e5.
[24] Kumar N, Gupta R, Gupta S. Whole slide imaging (WSI) in pa- [45] Hopper AD, Cross SS, Sanders DS. Patchy villous atrophy in adult
thology: current perspectives and future directions. J Digit Imaging patients with suspected gluten-sensitive enteropathy: is a multiple
2020;33:1034e40. duodenal biopsy strategy appropriate? Endoscopy 2008;40:219e24.
[25] Reunala T, Hervonen K, Salmi T. Dermatitis herpetiformis: an update [46] Prasad KK, Thapa BR, Nain CK, Singh K. The frequency of histo-
on diagnosis and management. Am J Clin Dermatol 2021;22: logic lesion variability of the duodenal mucosa in children with celiac
329e38. disease. World J Pediatr 2010;6:60e4.
[26] Stancu M, De Petris G, Palumbo TP, Lev R. Collagenous gastritis [47] McCarty TR, O’Brien CR, Gremida A, Ling C, Rustagi T. Efficacy of
associated with lymphocytic gastritis and celiac disease. Arch Pathol duodenal bulb biopsy for diagnosis of celiac disease: a systematic
Lab Med 2001;125:1579e84. review and meta-analysis. Endosc Int Open 2018;6:E1369e78.
[27] Green PH, Yang J, Cheng J, Lee AR, Harper JW, Bhagat G. An [48] Stoven SA, Choung RS, Rubio-Tapia A, et al. Analysis of biopsies
association between microscopic colitis and celiac disease. Clin from duodenal bulbs of all endoscopy patients increases detection of
Gastroenterol Hepatol 2009;7:1210e6. abnormalities but has a minimal effect on diagnosis of celiac disease.
[28] Robert ME. Gluten sensitive enteropathy and other causes of small Clin Gastroenterol Hepatol 2016;14:1582e8.
intestinal lymphocytosis. Semin Diagn Pathol 2005;22:284e94. [49] Mooney PD, Kurien M, Evans KE, et al. Clinical and immunologic
[29] Brown I, Mino-Kenudson M, Deshpande V, Lauwers GY. Intra- features of ultra-short celiac disease. Gastroenterology 2016;150:
epithelial lymphocytosis in architecturally preserved proximal small 1125e34.
30 Z.E. Chen et al.

[50] Doyev R, Cohen S, Ben-Tov A, et al. Ultra-short celiac disease is a [61] Barry RE, Morris JS, Read AE. Collagenous sprue. N Engl J Med
distinct and milder phenotype of the disease in children. Dig Dis Sci 1971;284:1041.
2019;64:167e72. [62] Zhao X, Johnson RL. Collagenous sprue: a rare, severe small-
[51] Mata-Romero P, Martin-Holgado D, Ferreira-Nossa HC, et al. Ultra- bowel malabsorptive disorder. Arch Pathol Lab Med 2011;135:
short celiac disease exhibits differential genetic and immunopheno- 803e9.
typic features compared to conventional celiac disease. Gastroenterol [63] Kamimura K, Kobayashi M, Sato Y, Aoyagi Y, Terai S. Collagenous
Hepatol 2022. gastritis: review. World J Gastrointest Endosc 2015;7:265e73.
[52] Hujoel IA, Murray JA. Refractory celiac disease. Curr Gastroenterol [64] Daum S, Foss HD, Schuppan D, Riecken EO, Zeitz M, Ullrich R.
Rep 2020;22:18. Synthesis of collagen I in collagenous sprue. Clin Gastroenterol
[53] Malamut G, Cellier C. Refractory celiac disease. Gastroenterol Clin Hepatol 2006;4:1232e6.
North Am 2019;48:137e44. [65] Maguire AA, Greenson JK, Lauwers GY, et al. Collagenous sprue: a
[54] Rowinski SA, Christensen E. Epidemiologic and therapeutic aspects of clinicopathologic study of 12 cases. Am J Surg Pathol 2009;33:
refractory coeliac disease - a systematic review. Dan Med J 2016;63. 1440e9.
[55] Haere P, Hoie O, Schulz T, Schonhardt I, Raki M, Lundin KE. Long- [66] Vakiani E, Arguelles-Grande C, Mansukhani MM, et al. Collagenous
term mucosal recovery and healing in celiac disease is the rule - not sprue is not always associated with dismal outcomes: a clinicopath-
the exception. Scand J Gastroenterol 2016;51:1439e46. ological study of 19 patients. Mod Pathol 2010;23:12e26.
[56] Ettersperger J, Montcuquet N, Malamut G, et al. Interleukin-15- [67] Smyrk TC. Practical approach to the flattened duodenal biopsy. Surg
Dependent T-cell-like innate intraepithelial lymphocytes develop in Pathol Clin 2017;10:823e39.
the intestine and transform into lymphomas in celiac disease. Im- [68] Rubio-Tapia A, Talley NJ, Gurudu SR, Wu TT, Murray JA. Gluten-
munity 2016;45:610e25. free diet and steroid treatment are effective therapy for most patients
[57] Sedda S, De Simone V, Marafini I, et al. High Smad7 sustains in- with collagenous sprue. Clin Gastroenterol Hepatol 2010;8:344e349
flammatory cytokine response in refractory coeliac disease. Immu- e3.
nology 2017;150:356e63. [69] Choung RS, Sharma A, Chedid VG, Absah I, Chen ZE, Murray JA.
[58] Celli R, Hui P, Triscott H, et al. Clinical Insignficance of monoclonal Collagenous gastritis: characteristics and response to topical bude-
T-cell populations and duodenal intraepithelial T-cell phenotypes in sonide. Clin Gastroenterol Hepatol 2021.
celiac and Nonceliac patients. Am J Surg Pathol 2019;43:151e60. [70] Murray JA, Kelly CP, Green PHR, et al. No difference between lat-
[59] Hussein S, Gindin T, Lagana SM, et al. Clonal T cell receptor gene iglutenase and placebo in reducing villous atrophy or improving
rearrangements in coeliac disease: implications for diagnosing re- symptoms in patients with symptomatic celiac disease. Gastroenter-
fractory coeliac disease. J Clin Pathol 2018;71:825e31. ology 2017;152:787e98. e2.
[60] van Wanrooij RL, Muller DM, Neefjes-Borst EA, et al. Optimal [71] Taavela J, Viiri K, Valimaki A, et al. Apolipoprotein A4 defines the
strategies to identify aberrant intra-epithelial lymphocytes in re- villus-crypt border in duodenal specimens for celiac disease
fractory coeliac disease. J Clin Immunol 2014;34:828e35. morphometry. Front Immunol 2021;12:713854.

You might also like