Skip to content
Publicly Available Published by De Gruyter October 9, 2013

Inflammatory bowel diseases: from pathogenesis to laboratory testing

  • Daniela Basso

    Daniela Basso is responsible of the Molecular Biology Section of the Department of Laboratory Medicine at the University-Hospital of Padua, Italy. She received her medical degree from the University of Padua (Italy). She completed her Residency and obtained the following Fellowships at the same University: 1. Gastroenterology and Digestive Endoscopy and 2. Clinical Pathology. Dr. Basso is one of the leading research experts in test selection for the diagnosis of celiac disease, pancreatic ductal adenocarcinoma and H. pylori infection. She is also research expert in the pathophysiology of pancreatic cancer-associated diabetes mellitus, tumor/stroma interactions, H. pylori/host genetics interactions and inflammatory bowel diseases. She has participated in National and European review committees and projects in the gastroenterology field.

    EMAIL logo
    , Carlo-Federico Zambon

    Carlo-Federico Zambon is a researcher at the Department of Medicine-DIMED of the University of Padua, Italy. At the same University he received the Degree in Biology, the PhD in Molecular and Cellular Pharmacology and the Postdoctoral Specialty in Biochemistry and Clinical Chemistry. Dr. Zambon is one of the leading research experts in the pathophysiology of H. pylori infection with a particular interest in the interplay between host and bacterium in determining its clinical outcome. He is also a research expert in molecular biology testing for the diagnosis of pancreatic ductal adenocarcinoma and H. pylori infection as well as in pharmacogenetic testing. He has participated in National and European projects in the gastroenterology field.

    and Mario Plebani

    Mario Plebani obtained his medical degree summa cum laude from the Medical School of the University of Padua in 1975. He completed residency training and specialization in Laboratory Medicine (1978), and subsequently in Gastroenterology (1983), at the same University. He is full Professor of Clinical Biochemistry and Clinical Molecular Biology at the University of Padua, School of Medicine, Chief of the Department of Laboratory Medicine at the University-Hospital of Padua, and Chief of the Center of Biomedical Research (a specialized Center for quality in laboratory medicine for the Veneto Region). Currently, he is also Director of the Post-graduate School in Clinical Biochemistry at the Medical School of the Padua University and President of the Course for Medical Technologists at the same Medical School. He served as President of the International Society of Enzymology (ISE) for four years (2004–2008) and as President of the Italian Society of Clinical Biochemistry and Molecular Clinical Biology for five years (in 2003 and from 2007 to 2009). He is the Chairman of the IFCC Working group on “Laboratory errors and patient safety” (WG LEPS) and in 2008 received the AACC Award for Outstanding Clinical Laboratory Contributions to Improving Patient Safety. Dr. Plebani is Editor in Chief of CCLM (Clinical Chemistry and Laboratory Medicine), and Associate editor of CRC Clinical Laboratory Sciences and International Journal of Biological Markers. His main areas of research are quality in laboratory medicine, biomarkers in cancer and cardiovascular diseases, and in vitro allergy diagnostics.

Abstract

Inflammatory bowel diseases (IBDs), which comprise the two major clinical subtypes, Crohn’s disease and ulcerative colitis, incur high morbidity and potential mortality. The present study reviews data on the pathogenesis and diagnosis of IBDs. The pathogenesis depends on complex interactions between susceptibility genes, environmental factors, and innate and adaptive immunity, the understanding of which is crucial to discovering novel laboratory biomarkers. Traditional laboratory tests for the diagnosis, prognosis and assessment of disease activity of IBDs are reported on, and the biochemical properties, pre-analytical and analytical aspects and clinical utility of the fecal markers lactoferrin and calprotectin are described. DNA testing and established (ASCA and pANCA) and emerging (ACCA, ALCA, AMCA, OmpC) serum markers are described; a further aspect to be addressed is the clinical use of pharmacogenetics for the treatment of IBDs.

Inflammatory bowel diseases (IBDs) – epidemiology, genetics and pathogenesis

Inflammatory bowel diseases (IBDs) comprise a heterogeneous group of chronic inflammatory disorders of the intestinal tract, most cases having an onset during young adulthood, although about 20%–25% of patients are diagnosed during childhood [1–3]. Unlike adult IBDs, pediatric IBDs have a particularly severe clinical activity, and are predominant in males, the most frequent clinical presentation being ileocolonic inflammation in cases of Crohn’s disease (CD) and pan-colitis in ulcerative colitis (UC), the two major clinical subtypes. IBDs, one of the largest healthcare challenges in developed countries, affect over one million people in Europe, with a North-South incidence gradient [1]. While the incidence of IBDs in western Europe has progressively increased since the World War II, apparently plateauing and now affecting 0.5%–1.0% of the population, it is on the increase in areas with a low incidence (eastern and southern Europe, the Middle East, Asia, Latin America and Indian subcontinent). IBDs have higher morbidity and mortality rates than most other diseases, and incur an important risk of complications (in children 29% at diagnosis and 59% at follow-up; in adults 16% after diagnosis) [2]. Currently, pediatric IBDs are considered a distinct disease rather than simply a particularly severe and early presentation of a common form of IBDs evolving from pediatric to adult age. The exact cause of IBDs is still unknown as no effective genetic and/or biochemical IBDs markers are available, the diagnosis calling for colonoscopy and histology [1]. It is believed that the pathogenesis of IBDs depends on complex interactions between susceptibility genes, environmental factors, and innate and adaptive immunity [4–11].

In the last decade, genome-wide association (GWA) studies have been conducted to shed light on genetic loci potentially underlying susceptibility to IBDs. The majority of studies have been made on adult IBDs, but two large studies have been performed on pediatric IBDs [12–18]. Overall, it has been confirmed that CD and UC share a common genetic ground, but each has distinct genetic features, the IBD5 locus (5q31-33 region) being strongly associated with UC, and the NOD2 (16q12) and major histocompatibility complex (MHC) (6p21) locus with CD [19, 20]. Furthermore, adult and pediatric IBDs have some susceptibility loci in common but each also has distinct genetic markers. Imielinski et al. [14, 16], who recently conducted the largest GWA study among children, validated 31/49 known adult loci and found five new loci associated with early-onset IBDs. Several genetic polymorphsims associated with IBDs depend on proteins entering the inflammasome multiprotein complexes formation, with the consequent activation of the cysteine protease caspase-1 and the processing and activation of pro-IL1‧, pro-IL18 and pro-IL33 [21]. This complex is formed by a nucleotide binding domain leucine rich repeat containing receptors (NLRs), the adaptor molecule apoptosis associated speck-like protein (ASC) containing the caspase recruitment domain (CARD) and pro-caspase-1 (Figure 1). The specific name of inflammasome (i.e., NLRP1 inflammasome or NLRP3 inflammasome) stems from the individual NLR number, which might be one of the 22 known molecules included in the complex [21]. NOD2 may also be required to activate NLRP1 inflammasome. Three NOD2 polymorphisms [rs2066844 (R702W), rs2066845 (G908R), rs5743293 (L1007fsinsC)], leading to loss of function, may be associated with CD [6], while NLRP1 polymorphism rs12150220 (Leu155His) has been associated with resistance to steroid treatment in young IBDs patients [22]. The above data indicate that NLRP1 inflammasome is implicated in IBDs, but epidemiological and experimental data suggest that NLRP3 inflammasome is also involved [23]. Decreased NLRP3 expression and IL1‧ production, associated with the rs6672995 and rs4353135 risk alleles, has been linked to increased CD susceptibility [24]; as demonstrated in an experimental animal model, this decrease in NLRP3 expression enhances susceptibility to dextran sodium sulphate induced colitis [25]. In brief, a number of genetic polymorphisms associated with an increased IBDs risk are associated with reduced inflammasome complex proteins expression, and consequently with a reduced caspase-1 activity.

Figure 1 NLRP1 and NLRP3 inflammasomes multiprotein complexes.These complexes are formed by a nucleotide binding domain leucine rich repeat containing receptors (NLRP), the adaptor molecule apoptosis associated speck-like protein (ASC) containing caspase recruitment domain and pro-caspase-1. The activation of NLRP1 inflammasome requires NOD2. The cysteine protease caspase-1 activates pro-IL1β, pro-IL18 and pro-IL33.
Figure 1

NLRP1 and NLRP3 inflammasomes multiprotein complexes.

These complexes are formed by a nucleotide binding domain leucine rich repeat containing receptors (NLRP), the adaptor molecule apoptosis associated speck-like protein (ASC) containing caspase recruitment domain and pro-caspase-1. The activation of NLRP1 inflammasome requires NOD2. The cysteine protease caspase-1 activates pro-IL1β, pro-IL18 and pro-IL33.

The gut mucosa, an extensive surface deputed to nutrient absorption, is primarily involved in maintaining a delicate balance between host defence and uncontrolled inflammation. Immune system cells, extensively present in the gut mucosa, regulate the balance between tolerance and inflammation in response to the huge bulk of environmental and intestinal microbial antigens reaching this large tissue-environmental interface. Intraepithelial lymphocytes (IELs), the primary mediators of the innate immune response to antigens in the epithelial layer of the small intestine, constitute a unique T cell population in that they differ from circulating, spleen and lymph node T cells (MHC class II restricted CD4+ αβ and MHC class I restricted CD8+ αβ). IELs are not MHC class II restricted, mainly being made up of (>80%) CD8+ T cells, which include CD8+ αα‧T cell (absent in blood), and 10% of CD8-CD4- T cells (double negative T cells, rare in blood) [26]. IELs are involved in the primary response to foreign antigens (e.g., food and microbial proteins), but are also autoreactive to “self-engendered” molecules deriving from infection or cell transformation; these molecules are independent of foreign epitopes [27]. The adaptive immune response taking place in the lamina propria, which has been well characterized in celiac disease with CD4+ T cells responding to antigens in a MHC class II restricted manner [28–31], is also implicated in IBDs. Innate and adaptive immune response are mediated by dendritic cells in the intestinal mucosa which, through the release of different cytokines, may either induce tolerance or evoke a cascade of inflammatory events ultimately leading to chronic inflammation. Dendritic cells in the gut mucosa may favor tolerance by releasing high amounts of the anti-inflammatory cytokine IL10 or promote inflammation through the release of IL12. The finding that IL10 and IL12 genetic polymorphisms are associated with the risk of developing IBDs supports the hypothesis that the adaptive immune response plays a primary role in the pathogenesis of IBDs [12, 14, 19, 32]; this is further supported by the observation that several of the more recently identified genetic susceptibility genes (IL23R, IL12B, IL27, IL18R1, IL18RAP, JAK2, IL10, TNFRSF6B and STAT3) are involved in innate and adaptive immunity [16].

CD-related inflammation in the gut mucosa is characterized by a marked infiltration of CD4+ T cells secreting T helper type 1 (Th1) and Th17 cytokines, whereas in UC the local immune response is less polarized, although IL5, IL13 and Th17 cytokines production may be enhanced [33]. Th17 cells and related cytokines may be involved in the expansion and accumulation of myeloid-derived suppressive cells (MDSCs), which have been described in animal models of experimental colitis, and in patients with IBDs [34]. Th17 and MDSCs cells appear to be implicated in the pathogenesis of autoimmune diseases, and it has been suggested that autoimmunity can also contribute to the onset of IBDs [35]. This hypothesis is supported by the observation that in sera of patients with CD pancreatic autoantibodies recognizing the major zymogen granule membrane glycoprotein 2 (GP-2) are detected in about 30% of cases [36].

However, despite the numerous genetic studies conducted so far in large patient cohorts, as much as 80% of hereditability in IBDs has yet to be clarified. The belief that environmental factors play a crucial role in IBDs expression is in line with the epidemiological observations of a rise in the incidence of these diseases proportionate to improvement in socio-economic conditions, and the observed surge of the incidence of IBDs in South Asian children migrating to Canada or the UK, mirroring that of the host country [37]. One of the most important environmental factors to be suspected is eating habit, particularly the western and “westernized” diet, which may contribute to IBDs pathogenesis through several mechanisms, including the direct effect of dietary antigens, alterations in gene expression and in the composition of the enteric flora and the consequent effects on gastrointestinal permeability [37]. An increased consumption of refined carbohydrates, high total fat and protein intake have been associated with CD and/or UC, while a high intake of dietary fibers (vegetables and fruits) has been associated with a decreased risk of IBDs [8]. Animal protein consumption has been cited as potentially involved in the pathogenesis of this disease in Europe, US and Japan [8, 37]. In only a few epidemiological studies have different sources of animal protein been investigated to ascertain the risk of IBDs; Bernstein et al. [38] found a higher risk of CD and UC in pork than in chicken meat consumers, while Sakamoto et al. [39] found an increased risk of CD, not UC, in fish consumers.

IBDs – laboratory diagnosis

Fecal markers

In the presence of suggestive symptoms, blood and stool laboratory tests are integral to the overall diagnostic work-up, which includes a physical exam, clinical history, endoscopy, biopsy and imaging [1]. A complete blood count, ESR and CRP, which may indicate the presence of intestinal inflammation, are, however, unreliable indexes of disease activity in patients with confirmed IBDs. Other blood parameters, including electrolytes, ferritin, calcium, magnesium, cobalamin, liver enzymes and function tests (INR, albumin, bilirubin), may indicate malabsorption and/or intestinal protein loss. Calprotectin and lactoferrin are the most reliable available fecal markers to be used as indexes of intestinal inflammation [1, 40–44]; both proteins, produced mainly by polymorphonuclear and monocytic inflammatory cells, are shaded in the intestinal lumen in the presence of mucosal inflammation. Fecal calprotectin/lactoferrin, considered hallmarks of neutrophilic intestinal inflammation, are invariably present in IBDs, thus explaining the high sensitivity of these markers in detecting the condition. Other organic intestinal diseases (e.g., celiac disease, diverticulosis, colorectal carcinoma) are also associated with neutrophilic intestinal inflammation, and consequently with an increased fecal calprotectin/lactoferrin output (Table 1). Therefore fecal calprotectin, with a sensitivity of 64%–95% and a specificity of 79%–93%, has been suggested as an index of organic bowel disease [43]. Nonetheless, an increased fecal level of calprotectin and/or lactoferrin leads to the identification of patients most likely to have IBDs and calls for immediate endoscopy; moreover, the use of fecal calprotectin/lactoferrin for screening allows a reduction in the number of negative findings at endoscopy in both adults and young patients with suspected IBDs [45]. In adults, the overall sensitivity of these markers in discriminating between patients with IBDs and healthy subjects is 93%, and the specificity 96%; the sensitivity is also high (92%) in children and teenagers, but the specificity is less satisfactory (76%) [45, 46]. The reasons for the discrepancy in specificity between adults and children probably depends on both age-related variations and the higher prevalence among children of intestinal disorders that can lead to augmented calprotectin fecal levels. Overall, fecal calprotectin and lactoferrin, which are correlated with each other, perform equally well in diagnosing IBDs and therefore should be used as an alternative, rather than an addition, to the diagnostic flow-chart.

Table 1

Causes of abnormal results for fecal calprotectin/lactoferrin other than IBDs.

InfectionsDrugs
Giardia lambliaNon-steroidal anti-inflammatory drugs
Bacterial or viral gastroenteritisProton pump inhibitors
Helicobacter pylori gastritis
MalignancyOther gastrointestinal diseases
Colorectal cancerUntreated food allergy
Gastric cancerUntreated celiac disease
Intestinal lymphomaGastro-esophageal reflux disease
Colorectal adenomaDiverticular disease
Juvenile polypProtein losing enteropathy
Cystic fibrosis
Microscopic colitis

In patients with a confirmed diagnosis of IBDs, fecal markers are also sensitive and specific markers of disease activity. Table 2 reports the sensitivity and specificity of lactoferrin, calprotectin and of CRP in discriminating between active and inactive IBDs [47–54]. High levels of calprotectin/lactoferrin are also predictive of relapse for both UC and CD [55, 56]. The calprotectin thresholds specified by different authors for predicting relapse vary, ranging from 50 to 400 μg/g (Table 3). In their recent paper, Louis et al. [60] demonstrated that a simple algorithm based on six clinical and laboratory parameters (no previous surgical resection, male gender, Hb ‧145 g/L, leucocyte count >6×109/L, hsCRP ‧5 mg/L, fecal calprotectin ‧300 μg/g) allows the stratification of CD patients into four categories according to risk of relapse.

Table 2

Sensitivity and specificity of lactoferrin, calprotectin and of C reactive protein (CRP) in discriminating between active and inactive IBDs.

Author [ref.]IBDs typeLactoferrinCalprotectinCRP
Sens/SpecSens/SpecSens/Spec
Røseth et al. [47]CD and UC77%/100%
Solem et al. [48]CD54%/75%
D’Incà et al. [49]CD77%/80%81%/80%
D’Incà et al. [49]UC75%/60%78%/70%
Sipponen et al. [50]CD77%/100%87%/100%
Schoepfer et al. [51]UC86%–93%/71%–88%
Schoepfer et al. [52]CD89%/58%68%/58%
Sipponen et al. [53]CD80%/67%80%/89%
Lobatòn et al. [54]UC74%/90%84%/38%

CD, Crohn’s disease; Sens, sensitivity; Spec, specificity; UC, ulcerative colitis.

Table 3

Fecal calprotectin thresholds for predicting IBDs relapse rate.

Author [ref.]IBDs typeCut-off, μg/gRelapse rate with low calprotectin, %Relapse rate with high calprotectin, %
Tibble et al. [55]UC501085
CD501585
Sipponen et al. [53]UC+CD1002539
D’Incà et al. [57]UC1303079
Gisbert et al. [46]UC150931
Costa et al. [58]UC1501081
CD1505787
Walkiewicz et al. [59]CD4001156

CD, Crohn’s disease; UC, ulcerative colitis.

Lactoferrin and calprotectin: biochemical properties, pre-analytical and analytical aspects

Lactoferrin, an 80 kDa protein, stored in secondary granules of neutrophils, is released upon cell activation. Calprotectin, a 36.5 kDa hetero-trimer, consisting of one S100A8 light chain and two S100A9 heavy chains belonging to the S100 calcium binding proteins family, is present in the cytoplasm of granulocytes and accounts for about 60% of soluble proteins; it is secreted extracellularly from stimulated neutrophils and monocytes or released by cell disruption or death [44]. Both lactoferrin and calprotectin have antimicrobial properties: histidine-based zinc-binding sequences (His-X-X-X-His motif) are involved in the antimicrobial activity of calprotectin, while the amino-terminal domain (lactoferricin) liberated by pepsin cleavage is involved in the antimicrobial activity of lactoferrin. Both proteins are resistant to bacterial proteolysis, their integrity being maintained in stools, where they can easily be identified; since they remain stable in feces for up to 1 week when stored at room temperature, and for months when stored at −20°C [43], it may be possible to adopt low stringency transport conditions for fecal material, although transport in refrigerated boxes is advisable for a better overall standardization of the pre-analytical phase. Laboratory processing of the samples includes: 1) sampling and dilution; and 2) analysis. In the initial pre-analytical phase, 100 mg of feces should be collected from the fecal mass and diluted into an exact w/v ratio. The time-consuming weighting of fecal samples has now been obviated by the use of devices allowing the easy and rapid obtaining of standardized w/v fecal sample dilutions. However, the devices cause an under-recovery of calprotectin with respect to the classical manual procedure, and this appears to be particularly relevant in the case of liquid stool samples [61]. We obtained similar results for lactoferrin, when manual weighting was compared with the approach using the Schebo device; under-recovery was particularly evident when levels were below the upper normal limit. These findings suggest that each laboratory should investigate the transferability of published and/or manufacturers’ cut-off values not only to its own patients’ population but also to its own pre-analytical procedures for sample preparation. The upper normal limits for both calprotectin and lactoferrin, which have mainly been established by using manual processing of the stool samples, appear to be age-dependent: while for adults and adolescents it is estimated at 50 μg/g for calprotectin and 7–11 μg/g for lactoferrin [44, 62], in children younger than 10 years, values three to four-fold the reference cut-off are suggested [63]. Likewise, a separate reference range two-fold higher than the reference cut-off is required in patients over 60 years of age [63].

The commercially available ELISAs for calprotectin have a total assay imprecision with mean % CV values of <12% [61]. By contrast the biological variability may span from low (<10%) to very high (58%) values [64], suggesting that a single stool test result should be interpreted with great caution in clinical practice.

The complex scenario of calprotectin laboratory testing includes several rapid immunochromatographic tests [point-of-care tests (FC-POCT)]. Three types of FC-POCT (qualitative, semi-quantitative and quantitative) are available – and good correlations between the latter and ELISAs have been reported [54, 65–68]. However, it has yet to be established whether FC-POCT results call for ELISA confirmation, and the way in which quality should be monitored. Since FC-POCT is not suitable for monitoring IBDs in patients likely to have high concentrations, it is reasonable to suggest that ELISA should be used to confirm positive FC-POCT findings, although this is time-consuming and incurs additional costs. Furthermore, the set of results of each ELISA plate can be monitored by quality control of the manufacturers’ and patients’ extracts [61], but this does not apply to any FC-POCT. These considerations indicate that the uncontrolled use of FC-POCT is inadvisable and that batch ELISA testing is preferable in high volumes centralized laboratories.

Serum markers to define type of IBDs

Although the two main IBDs clinical subtypes, UC and CD, have distinct clinical and pathological features, IBDs remain unclassified in about 6% of pediatric and 8% of adult onset cases [2]. Serum markers of clinical utility in diagnosing IBDs and aiding the differentiation between CD and UC are: 1) IgA and IgG class antibodies anti-Saccharomyces cerevisiae (ASCA), which can be determined by means of enzyme immunoassays; and 2) IgG class antineutrophil cytoplasmic antibodies (ANCA) demonstrating atypical perinuclear staining (pANCA), which can be assessed by indirect fluorescent antibody assays. While ASCA are associated with CD, positive pANCA are more frequently encountered in UC [69–71]. The antigenic target of ASCA has been identified as the mannose residue from the phosphopeptidomannan of the S. cerevisiae cell wall. In making a diagnosis of CD, ASCA have a high specificity (96%–100%), but a limited sensitivity (50%–63%) [72, 73]. If CD is suspected, ASCA IgA and IgG should be measured: while about two-thirds of CD patients with ASCA IgG are also positive for ASCA IgA, one-third of these patients are IgA ASCA negative, and 0%–19% have only ASCA IgA antibodies. The prevalence of ASCA is much higher in cases of sporadic CD and family members with CD alone (63%) than in those with both CD and UC (33%). Furthermore, ASCA is detected more frequently in healthy relatives of CD patients (20%–25%), and may therefore be considered a familial and hereditary quantitative trait. pANCA are spontaneously produced by the lamina propria and mesenteric node lymphocytes with the antigenic target present on the inner side of the nuclear periphery [74]. Positive pANCA antibody detects 60%–80% of patients with UC, but will also identify 6%–20% of subjects with CD. The combined analysis of pANCA and ASCA, indicated for defining the IBDs type, raises the specificity of the test to 94.3% with a sensitivity of 51.3% in adults [69]. In the pediatric population pANCA combined with ASCA raises sensitivity to 70.3% with no significant variation in specificity (93.4%) [69]. If IBDs patients with ASCA positive/pANCA negative findings are more likely to have CD, while those with ASCA negative/pANCA positive are more likely to have UC, double negative or double positive ASCA and pANCA results do not support one diagnosis or another. A study was conducted in 2006 to clarify this aspect, a platform with 384 new antibodies anti-glycans, the predominant components of the cell wall surface of many micro-organisms such as fungi, yeast and bacteria, being tested: those directed against laminaribioside (ALCA), chitobioside (ACCA) and mannobioside (AMCA) were found more frequently and in higher levels in CD patients than in subjects with UC and healthy controls [75]. The sensitivity and specificity of these serum markers in distinguishing between CD subjects, healthy controls and UC patients are reported in Table 4 [76]. ANCA, ALCA, ACCA and AMCA levels, increased in both CD patients and in their first degree relatives, have been found to be associated with CD-associated NOD2 mutations [77–79]; this suggests that they may be the expression of an increased stimulation of the immune system in CD predisposed subjects.

Table 4

Performance of individual anti-glycans antibodies for the diagnosis of Crohn’s disease (CD) in comparison with ulcerative colitis (UC) and healthy controls.

Anti-glycan

antibody markers
Sensitivity %

Mean (min-max)
Specificity %

Mean (min-max)
CD vs. UC
 ASCA57 (52–61)88 (86–90)
 ALCA18 (12–27)92 (85–96)
 AMCA24 (18–31)92 (88–95)
 ACCA16 (11–22)92 (85–96)
CD vs. controls
 ASCA53 (45–61)70 (86–90)
 ALCA17 (9–31)72 (22–99)
 AMCA26 (18–37)87 (56–97)
 ACCA15 (9–23)81 (22–99)

Other potential serum biomarkers of CD, with respective clinical uses, sensitivity and specificity, are reported in Table 5. Although none of them is currently recommended instead of or in addition to ASCA determination, a combination of pANCA, ASCA, anti-CBir1 and anti-OmpC were shown to predict CD and UC in individuals from a low-risk population several years before any clinical sign of the disease [80]. Moreover, it has been suggested that anti-GP2 IgG and IgA serum autoantibodies combined with ASCA are of potential clinical utility in distinguishing CD from UC [36, 81], but that they may also be helpful in distinguishing between ileocolonic and colonic CD [35].

Table 5

Potential new serum biomarkers for IBDs differential diagnosis.

BiomarkerAntigenDisease indicationPrevalenceSens, %Spec, %
CDUCHC
Anti-CBir1 IgGFlagellin, NCir (Clostridium subphylum)CD

May help differentiate CD from UC in pANCA+
50%<5%8%5053
Anti-I2 IgABacterial DNA sequence derived from Pseudomonas fluorescensCD54%10%4%4276
Anti-OmpC Iga, IgGOuter membrane porin, originally isolated from E. coliCD

May identify CD <15% of ASCA-
20%–55%10%5%20–5589

CD, Crohn’s disease; HC, healthy controls; Sens, sensitivity; Spec, specificity; UC, ulcerative colitis.

Antibodies against intestinal bacterial components detected in the sera of CD patients and of their first degree relatives might be the expression of a dysfunctional intestinal barrier function that may lead to the dysfunctional handling of macromolecules and intestinal bacterial antigens, resulting in an excessive immunological intestinal mucosal response.

Intestinal permeability

Increased gastrointestinal permeability, known to play a role in the pathogenesis of IBDs, has been found in a large number of familial studies on CD patients and in their asymptomatic first-degree relatives [82–84]; it can be evaluated by sugar testing, which is based on the measurement of the urinary excretion of orally administered non-metabolized sugar probe molecules (lactulose/mannitole). Normal intestinal epithelium allows the trans-cellular absorption of about 10% of the monosaccharide mannitole, while only traces (<1%) of the disaccharide lactulose are absorbed through the paracellular pathway. A leaky intestinal epithelium causes increased paracellular lactulose absorption and urinary excretion with respect to mannitole, thus leading to an increased lactulose/mannitole urinary excretion ratio that, in excess of 0.025, indicates the presence of increased intestinal permeability [85].

Genetic markers

The development of IBDs depends on complex interactions between environmental factors, genetic predisposition, and innate and adaptive immune response. A series of GWA studies identified more than 70 loci for CD and 47 for UC, each locus typically spanning about 150 kb and encompassing an average of 3–4 genes. NOD2 (16q12) and MHC loci (6p21) are associated with an increased risk of CD, while the IBD5 locus (5q31-33 region) is associated with an increased risk of UC [12–19]. The CARD15/NOD2 proteins enter the formation of inflammasomes multiprotein complexes that, upon activation, lead to the activation of the cysteine protease caspase-1 and the resultant processing and activation of pro-IL1‧, pro-IL18 and pro-IL33 [21]. Although multiple variants in the gene can increase CD susceptibility, the most commonly identified mutations of NOD2 are R702W, G908R and L1007fsinsC. NOD2 mutations, found in approximately 30% of CD patients, are associated with a more severe form of the disease, early age at onset, and ileal lesions [86, 87]. Heterozygosity increases the risk two to three-fold, whereas homozygosity is associated with a 20–40-fold higher risk of developing CD. The IBD5 locus contains immunoregulatory genes, including IL4, IL5, IL13 and the interferon regulatory factor-1 [88]. Despite the numerous efforts made to identify predisposing genes, the reported percentage for IBDs heredity, as high as 80% has yet to be explained, and further prospective studies must be undertaken before any attempt is made to translate GWA study findings to clinical practice.

IBDs – pharmacognetics

The drugs employed for treating IBDs include anti-inflammatory agents (5-aminosalicylic acid, sulfasalazine, corticosteroids), immune modifiers (azathioprine and 6-mercaptopurine (6-MP), calcineurin inhibitors, methotrexate), anti-TNF agents (infliximab, adalimumab, cartolizumab), antibiotics (most commonly metronidazole and ciprofloxacin) and probiotics. Experimental agents and symptomatic therapy complete the IBDs treatment panel. Before starting thiopurines AZA or 6-MP administration, thiopurine methyl transferase (TPMT) phenotyping (enzyme levels) or genotyping is recommended in order to direct dosing thus maximizing patient safety [1, 89]. TPMT plays an important role in the metabolism of AZA and 6-MP, which it converts into inactive metabolites. Genetically determined differences in the enzymatic activity of TPMT may influence the likelihood of AZA- and 6-MP-induced myelotoxicity. More than 20 single nucleotide polymorphisms can cause a reduction in TPMT production; however, the most prevalent allele among whites associated with a reduced TPMT enzyme activity is TPMT*3A (0.035), followed by TPMT*3C (0.0042) and TPMT*2 (0.0019). To diagnose TPMT deficiency, phenotype or genotype analyses may be performed [90]. Phenotype analysis has the advantage of verifying the effects of different factors (e.g., blood transfusions, medications, alcohol, food, post-myelotoxicity), which might affect the genetically-based expected TPMT enzyme activity, but must be repeated throughout treatment and has a high inter-laboratory variability mainly due to differences in assay types (HPLC followed by enzymatic assay, tandem mass spectrometry or radiochemical assay). The sensitivity of genotype analysis, which is not influenced by exogenous factors and need not be repeated, maybe limited, but exceeds 90% when this assay method is used for detecting the major alleles (i.e., wild type TPMT*1 and the most frequent mutant alleles TPMT*2, *3A and *3C), rather than for rare mutations [91]. Treatment with AZA or 6-MT is contraindicated in homozygous TPMT mutant allele carriers, since these subjects have an extremely low TPMT enzyme activity and are therefore at a high risk of myelotoxicity [1, 89, 92]. It has not yet been demonstrated in heterozygote patients, for whom a reduced enzyme activity should be expected, whether dosage adjustments might improve the outcome of treatment or reduce myelotoxicity, although a regimen starting with a 50% standard dose reduction followed by adjustment based on the degree of myelosuppression (continued for 2–4 weeks to reach steady state) is advisable in these cases. In all cases periodic monitoring of cell blood count is mandatory for the early identification of myelotoxicity, and for the patient’s safety.

In conclusion, several genetic and biochemical laboratory testing strategies are currently available for supporting a diagnosis of IBDs, differentiating between IBDs types, predicting relapse and establishing the risk of adverse events from therapy.

Conflict of interest statement

Authors’ conflict of interest disclosure: The authors stated that there are no conflicts of interest regarding the publication of this article.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.


Corresponding author: Daniela Basso, MD, Department of Laboratory Medicine, University Hospital of Padua, Via Giustiniani 2, 35128 Padua, Italy, Phone: +39 0498212801, Fax: +39 0498211981, E-mail:

About the authors

Daniela Basso

Daniela Basso is responsible of the Molecular Biology Section of the Department of Laboratory Medicine at the University-Hospital of Padua, Italy. She received her medical degree from the University of Padua (Italy). She completed her Residency and obtained the following Fellowships at the same University: 1. Gastroenterology and Digestive Endoscopy and 2. Clinical Pathology. Dr. Basso is one of the leading research experts in test selection for the diagnosis of celiac disease, pancreatic ductal adenocarcinoma and H. pylori infection. She is also research expert in the pathophysiology of pancreatic cancer-associated diabetes mellitus, tumor/stroma interactions, H. pylori/host genetics interactions and inflammatory bowel diseases. She has participated in National and European review committees and projects in the gastroenterology field.

Carlo-Federico Zambon

Carlo-Federico Zambon is a researcher at the Department of Medicine-DIMED of the University of Padua, Italy. At the same University he received the Degree in Biology, the PhD in Molecular and Cellular Pharmacology and the Postdoctoral Specialty in Biochemistry and Clinical Chemistry. Dr. Zambon is one of the leading research experts in the pathophysiology of H. pylori infection with a particular interest in the interplay between host and bacterium in determining its clinical outcome. He is also a research expert in molecular biology testing for the diagnosis of pancreatic ductal adenocarcinoma and H. pylori infection as well as in pharmacogenetic testing. He has participated in National and European projects in the gastroenterology field.

Mario Plebani

Mario Plebani obtained his medical degree summa cum laude from the Medical School of the University of Padua in 1975. He completed residency training and specialization in Laboratory Medicine (1978), and subsequently in Gastroenterology (1983), at the same University. He is full Professor of Clinical Biochemistry and Clinical Molecular Biology at the University of Padua, School of Medicine, Chief of the Department of Laboratory Medicine at the University-Hospital of Padua, and Chief of the Center of Biomedical Research (a specialized Center for quality in laboratory medicine for the Veneto Region). Currently, he is also Director of the Post-graduate School in Clinical Biochemistry at the Medical School of the Padua University and President of the Course for Medical Technologists at the same Medical School. He served as President of the International Society of Enzymology (ISE) for four years (2004–2008) and as President of the Italian Society of Clinical Biochemistry and Molecular Clinical Biology for five years (in 2003 and from 2007 to 2009). He is the Chairman of the IFCC Working group on “Laboratory errors and patient safety” (WG LEPS) and in 2008 received the AACC Award for Outstanding Clinical Laboratory Contributions to Improving Patient Safety. Dr. Plebani is Editor in Chief of CCLM (Clinical Chemistry and Laboratory Medicine), and Associate editor of CRC Clinical Laboratory Sciences and International Journal of Biological Markers. His main areas of research are quality in laboratory medicine, biomarkers in cancer and cardiovascular diseases, and in vitro allergy diagnostics.

References

1. Bernstein CN, Fried M, Krabshuis JH, Cohen H, Eliakim R, Fedail S, et al. World gastroenterology organization practice guidelines for the diagnosis and management of IBD in 2010. Inflamm Bowel Dis 2010;16:112–24.10.1002/ibd.21048Search in Google Scholar PubMed

2. Guariso G, Gasparetto M, Visonà Dalla Pozza L, D’Incà R, Zancan L, Sturniolo G, et al. Inflammatory bowel disease developing in paediatric and adult age. J Pediatr Gastroenterol Nutr 2010;51:698–707.10.1097/MPG.0b013e3181da1db8Search in Google Scholar PubMed

3. Ruemmele FM. Pediatric inflammatory bowel diseases: coming of age. Curr Opin Gastroenterol 2010;26:332–6.10.1097/MOG.0b013e328339ec2dSearch in Google Scholar PubMed

4. Ferguson LR, Shelling AN, Browning BL, Huebner C, Petermann I. Genes, diet and inflammatory bowel disease. Mutat Res 2007;622:70–83.10.1016/j.mrfmmm.2007.05.011Search in Google Scholar PubMed

5. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007;448:427–34.10.1038/nature06005Search in Google Scholar PubMed

6. de Mesquita MB, Civitelli F, Levine A. Epidemiology, genes and inflammatory bowel diseases in childhood. Dig Liver Dis 2008;40:3–11.10.1016/j.dld.2007.07.165Search in Google Scholar PubMed

7. Renz H, von Mutius E, Brandtzaeg P, Cookson WO, Autenrieth IB, Haller D. Gene-environment interactions in chronic inflammatory disease. Nat Immunol 2011;12:273–7.10.1038/ni0411-273Search in Google Scholar PubMed

8. Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am J Gastroenterol 2011;106:563–73.10.1038/ajg.2011.44Search in Google Scholar PubMed

9. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011;474:307–17.10.1038/nature10209Search in Google Scholar PubMed PubMed Central

10. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 2011;474:298–306.10.1038/nature10208Search in Google Scholar PubMed

11. Kotlarz D, Beier R, Murugan D, Diestelhorst J, Jensen O, Boztug K, et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology 2012;143:347–55.10.1053/j.gastro.2012.04.045Search in Google Scholar PubMed

12. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet 2008;40:955–62.10.1038/ng.175Search in Google Scholar PubMed PubMed Central

13. Kugathasan S, Baldassano RN, Bradfield JP, Sleiman PM, Imielinski M, Guthery SL, et al. Loci on 20q13 and 21q22 are associated with pediatric-onset inflammatory bowel disease. Nat Genet 2008;40:1211–5.10.1038/ng.203Search in Google Scholar PubMed PubMed Central

14. Imielinski M, Baldassano RN, Griffiths A, Russell RK, Annese V, Dubinsky M, et al. Common variants at five new loci associated with early-onset inflammatory bowel disease. Nat Genet 2009;41:1335–40.10.1038/ng.489Search in Google Scholar PubMed PubMed Central

15. Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet 2010;42:1118–25.10.1038/ng.717Search in Google Scholar PubMed PubMed Central

16. Imielinski M, Hakonarson H. Breaking new ground in inflammatory bowel disease genetics: genome-wide association studies and beyond. Pharmacogenomics 2010;11:663–5.10.2217/pgs.10.56Search in Google Scholar PubMed

17. Anderson CA, Boucher G, Lees CW, Franke A, D’Amato M, Taylor KD, et al. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet 2011;43:246–52.10.1038/ng.764Search in Google Scholar PubMed PubMed Central

18. Rivas MA, Beaudoin M, Gardet A, Stevens C, Sharma Y, Zhang CK, et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet 2011;43:1066–73.10.1038/ng.952Search in Google Scholar PubMed PubMed Central

19. Anderson CA, Massey DC, Barrett JC, Prescott NJ, Tremelling M, Fisher SA, et al. Investigation of Crohn’s disease risk loci in ulcerative colitis further defines their molecular relationship. Gastroenterology 2009;136:523–9.10.1053/j.gastro.2008.10.032Search in Google Scholar PubMed PubMed Central

20. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491:119–24.10.1038/nature11582Search in Google Scholar PubMed PubMed Central

21. Pedra JH, Cassel SL, Sutterwala FS. Sensing pathogens and danger signals by the inflammasome. Curr Opin Immunol 2009;21:10–6.10.1016/j.coi.2009.01.006Search in Google Scholar PubMed PubMed Central

22. De Iudicibus S, Stocco G, Martelossi S, Londero M, Ebner E, Pontillo A, et al. Genetic predictors of glucocorticoid response in pediatric patients with inflammatory bowel diseases. J Clin Gastroenterol 2011;45:e1–7.10.1097/MCG.0b013e3181e8ae93Search in Google Scholar PubMed

23. Zaki MH, Lamkanfi M, Kanneganti TD. The Nlrp3 inflammasome: contributions to intestinal homeostasis. Trends Immunol 2011;32:171–9.10.1016/j.it.2011.02.002Search in Google Scholar PubMed PubMed Central

24. Villani AC, Lemire M, Fortin G, Louis E, Silverberg MS, Collette C, et al. Common variants in the NLRP3 region contribute to Crohn’s disease susceptibility. Nat Genet 2009;41:71–6.10.1038/ng.285Search in Google Scholar PubMed PubMed Central

25. Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti TD. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity 2010;32:379–91.10.1016/j.immuni.2010.03.003Search in Google Scholar PubMed PubMed Central

26. Chang F, Mahadeva U, Deere H. Pathological and clinical significance of increased intraepithelial lymphocytes (IELs) in small bowel mucosa. APMIS 2005;113:385–99.10.1111/j.1600-0463.2005.apm_204.xSearch in Google Scholar PubMed

27. Hayday A, Theodoridis E, Ramsburg E, Shires J. Intraepithelial lymphocytes: exploring the Third Way in immunology. Nat Immunol 2001;2:997–1003.10.1038/ni1101-997Search in Google Scholar PubMed

28. Koning F, Gilissen L, Wijmenga C. Gluten: a two-edged sword. Immunopathogenesis of celiac disease. Springer Semin Immunopathol 2005;27:217–32.10.1007/s00281-005-0203-9Search in Google Scholar PubMed

29. Green PH, Cellier C. Celiac disease. N Engl J Med 2007;357: 1731–43.10.1056/NEJMra071600Search in Google Scholar PubMed

30. Kagnoff MF. Celiac disease: pathogenesis of a model immunogenetic disease. J Clin Invest 2007;117:41–9.10.1172/JCI30253Search in Google Scholar PubMed PubMed Central

31. Briani C, Samaroo D, Alaedini A. Celiac disease: from gluten to autoimmunity. Autoimmun Rev 2008;7:644–50.10.1016/j.autrev.2008.05.006Search in Google Scholar PubMed

32. Wang AH, Lam WJ, Han DY, Ding Y, Hu R, Fraser AG, et al. The effect of IL-10 genetic variation and interleukin 10 serum levels on Crohn’s disease susceptibility in a New Zealand population. Hum Immunol 2011;72:431–5.10.1016/j.humimm.2011.02.014Search in Google Scholar PubMed

33. Monteleone G, Pallone F, MacDonald TT. Emerging immunological targets in inflammatory bowel disease. Curr Opin Pharmacol 2011;11:640–5.10.1016/j.coph.2011.09.013Search in Google Scholar PubMed

34. Ostanin DV, Bhattacharya D. Myeloid-derived suppressor cells in the inflammatory bowel diseases. Inflamm Bowel Dis 2013;19:2468–77.10.1097/MIB.0b013e3182902b11Search in Google Scholar PubMed

35. Somma V, Ababneh H, Ababneh A, Gatti S, Romagnoli V, Bendia E, et al. The novel Crohn’s disease marker anti-GP2 antibody is associated with ileocolonic location of disease. Gastroenterol Res Pract 2013;2013:683824.10.1155/2013/683824Search in Google Scholar PubMed PubMed Central

36. Roggenbuck D, Hausdorf G, Martinez-Gamboa L, Reinhold D, Büttner T, Jungblut PR, et al. Identification of GP2, the major zymogen granule membrane glycoprotein, as the autoantigen of pancreatic antibodies in Crohn’s disease. Gut 2009;58:1620–8.10.1136/gut.2008.162495Search in Google Scholar PubMed

37. Issa M, Saeian K. Diet in inflammatory bowel disease. Nutr Clin Pract 2011;26:151–4.10.1177/0884533611400233Search in Google Scholar PubMed

38. Bernstein CN, Rawsthorne P, Cheang M, Blanchard JF. A population-based case control study of potential risk factors for IBD. Am J Gastroenterol 2006;101:993–1002.10.1111/j.1572-0241.2006.00381.xSearch in Google Scholar PubMed

39. Sakamoto N, Kono S, Wakai K, Fukuda Y, Satomi M, Shimoyama T, et al. Dietary risk factors for inflammatory bowel disease: a multicenter case-control study in Japan. Inflamm Bowel Dis 2005;11:154–63.10.1097/00054725-200502000-00009Search in Google Scholar PubMed

40. Langhorst J, Elsenbruch S, Koelzer J, Rueffer A, Michalsen A, Dobos GJ. Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol 2008;103:162–9.10.1111/j.1572-0241.2007.01556.xSearch in Google Scholar PubMed

41. Wong A, Bass D. Laboratory evaluation of inflammatory bowel disease. Curr Opin Pediatr 2008;20:566–70.10.1097/MOP.0b013e32830d3aafSearch in Google Scholar PubMed

42. Joishy M, Davies I, Ahmed M, Wassel J, Davies K, Sayers A, et al. Fecal calprotectin and lactoferrin as noninvasive markers of pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2009;48:48–54.10.1097/MPG.0b013e31816533d3Search in Google Scholar PubMed

43. Ayling RM. New faecal tests in gastroenterology. Ann Clin Biochem 2012;49:44–54.10.1258/acb.2011.011150Search in Google Scholar PubMed

44. Sherwood RA. Faecal markers of gastrointestinal inflammation. J Clin Pathol 2012;65:981–5.10.1136/jclinpath-2012-200901Search in Google Scholar PubMed

45. van Rheenen PF, Van de Vijver E, Fidler V. Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta-analysis. Br Med J 2010;341:c3369.10.1136/bmj.c3369Search in Google Scholar PubMed PubMed Central

46. Gisbert JP, McNicholl AG, Gomollon F. Questions and answers on the role of fecal lactoferrin as a biological marker in inflammatory bowel disease. Inflamm Bowel Dis 2009;15: 1746–54.10.1002/ibd.20920Search in Google Scholar PubMed

47. Røseth AG, Aadland E, Grzyb K. Normalization of faecal calprotectin: a predictor of mucosal healing in patients with inflammatory bowel disease. Scand J Gastroenterol 2004;39:1017–20.10.1080/00365520410007971Search in Google Scholar PubMed

48. Solem CA, Loftus EV Jr, Tremaine WJ, Harmsen WS, Zinsmeister AR, Sandborn WJ. Correlation of C-reactive protein with clinical, endoscopic, histologic, and radiographic activity in inflammatory bowel disease. Inflamm Bowel Dis 2005;11:707–12.10.1097/01.MIB.0000173271.18319.53Search in Google Scholar PubMed

49. D’Incà R, Dal Pont E, Di Leo V, Ferronato A, Fries W, Vettorato MG, et al. Calprotectin and lactoferrin in the assessment of intestinal inflammation and organic disease. Int J Colorectal Dis 2007;22:429–37.10.1007/s00384-006-0159-9Search in Google Scholar PubMed

50. Sipponen T, Kärkkäinen P, Savilahti E, Kolho KL, Nuutinen H, Turunen U, et al. Correlation of faecal calprotectin and lactoferrin with an endoscopic score for Crohn’s disease and histological findings. Aliment Pharmacol Ther 2008;28:1221–9.10.1111/j.1365-2036.2008.03835.xSearch in Google Scholar PubMed

51. Schoepfer AM, Beglinger C, Straumann A, Trummler M, Renzulli P, Seibold F. Ulcerative colitis: correlation of the Rachmilewitz endoscopic activity index with fecal calprotectin, clinical activity, C-reactive protein, and blood leukocytes. Inflamm Bowel Dis 2009;15:1851–8.10.1002/ibd.20986Search in Google Scholar PubMed

52. Schoepfer AM, Beglinger C, Straumann A, Trummler M, Vavricka SR, Bruegger LE, et al. Fecal calprotectin correlates more closely with the simple endoscopic score for Crohn’s disease (SES-CD) than CRP, blood leukocytes, and the CDAI. Am J Gastroenterol 2010;105:162–9.10.1038/ajg.2009.545Search in Google Scholar PubMed

53. Sipponen T, Björkesten CG, Färkkilä M, Nuutinen H, Savilahti E, Kolho KL. Faecal calprotectin and lactoferrin are reliable surrogate markers of endoscopic response during Crohn’s disease treatment. Scand J Gastroenterol 2010;45:325–31.10.3109/00365520903483650Search in Google Scholar PubMed

54. Lobatón T, Rodríguez-Moranta F, Lopez A, Sánchez E, Rodríguez-Alonso L, Guardiola J. A new rapid quantitative test for fecal calprotectin predicts endoscopic activity in ulcerative colitis. Inflamm Bowel Dis 2013;19:1034–42.10.1097/MIB.0b013e3182802b6eSearch in Google Scholar PubMed

55. Tibble JA, Sigthorsson G, Bridger S, Fagerhol MK, Bjarnason I. Surrogate markers of intestinal inflammation are predictive of relapse in patients with inflammatory bowel disease. Gastroenterology 2000;119:15–22.10.1053/gast.2000.8523Search in Google Scholar PubMed

56. Scarpa M, D’Incà R, Basso D, Ruffolo C, Polese L, Bertin E, et al. Fecal lactoferrin and calprotectin after ileocolonic resection for Crohn’s disease. Dis Colon Rectum 2007;50:861–9.10.1007/s10350-007-0225-6Search in Google Scholar PubMed

57. D’Incà R, Dal Pont E, Di Leo V, Benazzato L, Martinato M, Lamboglia F, et al. Can calprotectin predict relapse risk in inflammatory bowel disease? Am J Gastroenterol 2008;103:2007–14.10.1111/j.1572-0241.2008.01870.xSearch in Google Scholar PubMed

58. Costa F, Mumolo MG, Ceccarelli L, Bellini M, Romano MR, Sterpi C, et al. Calprotectin is a stronger predictive marker of relapse in ulcerative colitis than in Crohn’s disease. Gut 2005;54:364–8.10.1136/gut.2004.043406Search in Google Scholar PubMed PubMed Central

59. Walkiewicz D, Werlin SL, Fish D, Scanlon M, Hanaway P, Kugathasan S. Fecal calprotectin is useful in predicting disease relapse in pediatric inflammatory bowel disease. Inflamm Bowel Dis 2008;14:669–73.10.1002/ibd.20376Search in Google Scholar PubMed

60. Louis E, Mary JY, Vernier-Massouille G, Grimaud JC, Bouhnik Y, Laharie D, et al. Maintenance of remission among patients with Crohn’s disease on antimetabolite therapy after infliximab therapy is stopped. Gastroenterology 2012;142:63–70.10.1053/j.gastro.2011.09.034Search in Google Scholar PubMed

61. Whitehead SJ, French J, Brookes MJ, Ford C, Gama R. Between-assay variability of faecal calprotectin enzyme-linked immunosorbent assay kits. Ann Clin Biochem 2013;50:53–61.10.1258/acb.2012.011272Search in Google Scholar

62. Angriman I, Scarpa M, D’Incà R, Basso D, Ruffolo C, Polese L, et al. Enzymes in feces: useful markers of chronic inflammatory bowel disease. Clin Chim Acta 2007;381:63–8.10.1016/j.cca.2007.02.025Search in Google Scholar

63. Joshi S, Lewis SJ, Creanor S, Ayling RM. Age-related faecal calprotectin, lactoferrin and tumour M2-PK concentrations in healthy volunteers. Ann Clin Biochem 2010;47:259–63.10.1258/acb.2009.009061Search in Google Scholar

64. Husebye E, Tøn H, Johne B. Biological variability of fecal calprotectin in patients referred for colonoscopy without colonic inflammation or neoplasm. Am J Gastroenterol 2001;96:2683–7.10.1016/S0002-9270(01)02688-0Search in Google Scholar

65. Dolci A, Panteghini M. Comparative study of a new quantitative rapid test with an established ELISA method for faecal calprotectin. Clin Chim Acta 2012;413:350–1.10.1016/j.cca.2011.09.030Search in Google Scholar PubMed

66. Kolho KL, Turner D, Veereman-Wauters G, Sladek M, de Ridder L, Shaoul R, et al. Rapid test for fecal calprotectin levels in children with Crohn disease. J Pediatr Gastroenterol Nutr 2012;55:436–9.10.1097/MPG.0b013e318253cff1Search in Google Scholar PubMed

67. Sydora MJ, Sydora BC, Fedorak RN. Validation of a point-of-care desk top device to quantitate fecal calprotectin and distinguish inflammatory bowel disease from irritable bowel syndrome. J Crohns Colitis 2012;6:207–14.10.1016/j.crohns.2011.08.008Search in Google Scholar PubMed

68. Wassell J, Wallage M, Brewer E. Evaluation of the Quantum Blue® rapid test for faecal calprotectin. Ann Clin Biochem 2012;49:55–8.10.1258/acb.2011.011106Search in Google Scholar PubMed

69. Reese GE, Constantinides VA, Simillis C, Darzi AW, Orchard TR, Fazio VW, et al. Diagnostic precision of anti-Saccharomyces cerevisiae antibodies and perinuclear antineutrophil cytoplasmic antibodies in inflammatory bowel disease. Am J Gastroenterol 2006;101:2410–22.10.1111/j.1572-0241.2006.00840.xSearch in Google Scholar PubMed

70. Peyrin-Biroulet L, Standaert-Vitse A, Branche J, Chamaillard M. IBD serological panels: facts and perspectives. Inflamm Bowel Dis 2007;13:1561–6.10.1002/ibd.20226Search in Google Scholar PubMed

71. Iskandar HN, Ciorba MA. Biomarkers in inflammatory bowel disease: current practices and recent advances. Transl Res 2012;159:313–25.10.1016/j.trsl.2012.01.001Search in Google Scholar PubMed PubMed Central

72. de Vries M, van der Horst-Bruinsma I, van Hoogstraten I, van Bodegraven A, von Blomberg BM, Ratnawati H, et al. pANCA, ASCA, and OmpC antibodies in patients with ankylosing spondylitis without inflammatory bowel disease. J Rheumatol 2010;37:2340–4.10.3899/jrheum.100269Search in Google Scholar PubMed

73. Lewis JD. The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology 2011;140:1817–26.10.1053/j.gastro.2010.11.058Search in Google Scholar PubMed PubMed Central

74. Kim WH, Choi PM, Landers CJ, Targan SR. Role of antineutrophil cytoplasmic antibodies in an ethnically distinct population: Korean patients with ulcerative colitis. Am J Gastroenterol 1995;90:1953–8.Search in Google Scholar

75. Kaul A, Hutfless S, Liu L, Bayless TM, Marohn MR, Li X. Serum anti-glycan antibody biomarkers for inflammatory bowel disease diagnosis and progression: a systematic review and meta-analysis. Inflamm Bowel Dis 2012;18:1872–84.10.1002/ibd.22862Search in Google Scholar PubMed PubMed Central

76. Dotan I. New serologic markers for inflammatory bowel disease diagnosis. Dig Dis 2010;28:418–23.10.1159/000320396Search in Google Scholar PubMed

77. Dassopoulos T, Frangakis C, Cruz-Correa M, Talor MV, Burek CL, Datta L, et al. Antibodies to Saccharomyces cerevisiae in Crohn’s disease: higher titers are associated with a greater frequency of mutant NOD2/CARD15 alleles and with a higher probability of complicated disease. Inflamm Bowel Dis 2007;13:143–51.10.1002/ibd.20031Search in Google Scholar PubMed

78. Papp M, Altorjay I, Dotan N, Palatka K, Foldi I, Tumpek J, et al. New serological markers for inflammatory bowel disease are associated with earlier age at onset, complicated disease behavior, risk for surgery, and NOD2/CARD15 genotype in a Hungarian IBD cohort. Am J Gastroenterol 2008;103:665–81.10.1111/j.1572-0241.2007.01652.xSearch in Google Scholar PubMed

79. Michielan A, Basso D, Martinato M, Pathak S, Banerjee A, Oliva L, et al. Increased antibody response to microbial antigens in patients with Crohn’s disease and their unaffected first-degree relatives. Dig Liver Dis 2013, in press.10.1016/j.dld.2013.01.017Search in Google Scholar PubMed

80. van Schaik FD, Oldenburg B, Hart AR, Siersema PD, Lindgren S, Grip O, et al. Serological markers predict inflammatory bowel disease years before the diagnosis. Gut 2013;62:683–8.10.1136/gutjnl-2012-302717Search in Google Scholar PubMed

81. Roggenbuck D, Reinhold D, Wex T, Goihl A, von Arnim U, Malfertheiner P, et al. Autoantibodies to GP2, the major zymogen granule membrane glycoprotein, are new markers in Crohn’s disease. Clin Chim Acta 2011;412:718–24.10.1016/j.cca.2010.12.029Search in Google Scholar PubMed

82. Hollander D. Permeability in Crohn’s disease: altered barrier functions in healthy relatives? Gastroenterology 1993;104:1848–51.10.1016/0016-5085(93)90668-3Search in Google Scholar

83. Peeters M, Geypens B, Claus D, Nevens H, Ghoos Y, Verbeke G, et al. Clustering of increased small intestinal permeability in families with Crohn’s disease. Gastroenterology 1997;113:802–7.10.1016/S0016-5085(97)70174-4Search in Google Scholar

84. Hollander D. Intestinal permeability barrier in Crohn’s disease: The difficulty in shifting the paradigm. Dig Dis Sci 2013;58:1827–9.10.1007/s10620-013-2721-ySearch in Google Scholar PubMed

85. Buhner S, Buning C, Genschel J, Kling K, Herrmann D, Dignass A, et al. Genetic basis for increased intestinal permeability in families with Crohn’s disease: role of CARD15 3020insC mutation? Gut 2006;55:342–7.10.1136/gut.2005.065557Search in Google Scholar PubMed PubMed Central

86. Economou M, Trikalinos TA, Loizou KT, Tsianos EV, Ioannidis JP. Differential effects of NOD2 variants on Crohn’s disease risk and phenotype in diverse populations: a metaanalysis. Am J Gastroenterol 2004;99:2393–404.10.1111/j.1572-0241.2004.40304.xSearch in Google Scholar PubMed

87. Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology 2011;140: 1704–12.10.1053/j.gastro.2011.02.046Search in Google Scholar PubMed PubMed Central

88. Georges M. The long and winding road from correlation to causation. Nat Genet 2011;43:180–1.10.1038/ng0311-180Search in Google Scholar PubMed

89. Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, et al. Clinical pharmacogenetics implementation consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther 2011;89:387–91.10.1038/clpt.2010.320Search in Google Scholar PubMed PubMed Central

90. Donnan JR, Ungar WJ, Mathews M, Rahman P. Systematic review of thiopurine methyltransferase genotype and enzymatic testing strategies. Ther Drug Monit 2011;33:192–9.10.1097/FTD.0b013e31820810cdSearch in Google Scholar PubMed

91. Schaeffeler E, Fischer C, Brockmeier D, Wernet D, Moerike K, Eichelbaum M, et al. Comprehensive analysis of thiopurine S-methyltransferase phenotype-genotype correlation in a large population of German-Caucasians and identification of novel TPMT variants. Pharmacogenetics 2004;14: 407–17.10.1097/01.fpc.0000114745.08559.dbSearch in Google Scholar PubMed

92. Chisick L, Oleschuk C, Bernstein CN. The utility of thiopurine methyltransferase enzyme testing in inflammatory bowel disease. Can J Gastroenterol 2013;27:39–43.10.1155/2013/280860Search in Google Scholar PubMed PubMed Central

Received: 2013-7-25
Accepted: 2013-9-10
Published Online: 2013-10-09
Published in Print: 2014-04-01

©2014 by Walter de Gruyter Berlin/Boston

Downloaded on 22.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2013-0588/html
Scroll to top button