Faecal calprotectin – a useful tool in the management of inflammatory bowel disease

DOI: https://doi.org/10.4414/smw.2012.13557

Emanuel Burri, Christoph Beglinger

Summary

Inflammatory bowel disease (IBD) should be suspected in any patient presenting with chronic or recurrent abdominal pain and diarrhoea. Current guidelines suggest performing invasive endoscopy with histological sampling for further diagnosis. Measuring calprotectin, a neutrophilic protein, in faeces has been proposed as a surrogate marker of intestinal inflammation. Calprotectin values have been shown to reliably differentiate between IBD and non-organic disease in symptomatic patients and, when elevated, warrant early endoscopic investigation to rule out IBD and other organic pathologies.

Endoscopy with histological sampling is also used to evaluate disease activity and here, too, faecal calprotectin values seem to correlate well. In a number of studies, faecal calprotectin values have consistently shown to better assess mucosal inflammation than clinical indices and serum markers. Calprotectin’s advantage of non-invasive monitoring of disease activity is especially beneficial when considering the dynamics of repeated measurements.

Mucosal healing (MH) has been associated with sustained clinical remission, reduced rates of hospitalisation and of surgical resection, both in Crohn’s disease and ulcerative colitis patients. Elevated faecal calprotectin levels in patients in clinical remission are associated with increased risk of disease relapse within 12 months follow-up. In most clinically quiescent IBD, residual mucosal inflammation is still present; it appears that faecal calprotectin can detect subclinical mucosal inflammation and thus might identify patients at risk for relapse.

In summary, measuring faecal calprotectin can be highly useful in the diagnosis and disease management of patients with IBD and could help predict disease course.

Introduction

Inflammatory bowel disease (IBD) is a life-long disorder that includes two major forms of chronic intestinal inflammation: Ulcerative colitis (UC) and Crohn’s disease (CD) (table 1). The aetiology of IBD is not yet fully understood, but the disorder seems to arise from interactions between genetic and environmental factors [1–4]. IBD is more prevalent in developed countries, affecting approximately 1–2 per 1000 people, and its incidence is increasing both in adults and children [5–7]. UC occurs more frequently in men aged 30–40 years, whereas CD is most often seen in women between 20–30 years. Paediatric IBD accounts for 7–20% of all IBD cases and, here, CD seems to be more prevalent [8, 9].

UC and CD both have distinct pathological features. However, the clinical presentation of IBD depends on the disease location and its extent and can thus be inconsistent, showing symptoms that overlap with both disorders. Suspicion should be raised when patients present with chronic or recurrent episodes of abdominal pain and/or diarrhoea. The probability of IBD increases when alarm signs, such as rectal bleeding, fever, anorexia, or anaemia, are reported [10]. A single parameter or laboratory value for diagnosing UC or CD is not yet available; diagnosis is confirmed by clinical evaluation and a combination of biochemical, radiological, endoscopic and histological analysis [11, 12]. A change in diagnosis from UC to CD, or vice versa, occurs in 10% of UC patients and 5% of CD patients [13].

Some 10% of patients have CD of the small bowel and up to 15% may have penetrating lesions (fistulae, phlegmonous disease, or abcesses) at the time of diagnosis [14]. Extraintestinal manifestations in IBD are common and occur in up to 43% of patients [15–18]. However, this might be an over-estimation that arises from high-volume referral centre data. Community studies suggest a lower prevalence of extraintestinal manifestations [17]. Guidelines for the treatment of IBD have been published [19–23].

Table 1: Definitions of Inflammatory Bowel Disease (IBD) and Irritable Bowel Syndrome (IBS).
Inflammatory Bowel Disease
Inflammatory bowel disease encompasses a multisystem group of disorders of chronic inflammation with specific clinical and pathological features that primarily affect the gastrointestinal tract. Extraintestinal manifestations include: musculoskeletal (peripheral arthritis, sacroiliitis, ankylosing spondylitis, osteoporosis), dermatological (erythema nodosum, pyoderma gangrenosum, aphthous stomatitis), and ocular (uveitis, scleritis, episcleritis) afflictions, as well as primary sclerosing cholangitis, thromboembolic events, and nephrolithiasis.
Ulcerative colitis (UC) is a chronic inflammatory condition causing continuous mucosal inflammation of the colon, without granulomas on biopsy, affecting the rectum and a variable extent of the colon in continuity, which is characterised by a relapsing and remitting course.
Crohn’s disease (CD) is characterised by patchy, transmural inflammation, which may affect any part of the gastrointestinal tract. It may be defined by location (terminal ileal, colonic, ileocolic, upper gastrointestinal), or by pattern of disease (inflammatory, fistulating, or stricturing).
Indeterminate colitis (IC): Because features of both conditions are present, about 5% of patients with IBD affecting the colon cannot be classified after considering clinical, radiological, endoscopic, and pathological criteria.
Irritable Bowel Syndrome
Recurrent abdominal pain or discomfort (uncomfortable sensation not described as pain) at least 3 days/month in the last 3 months, with symptom onset at least 6 months before diagnosis, and associated with two or more of the following: 1. Improvement with defecation 2. Onset associated with a change in frequency of stool 3. Onset associated with a change in form (appearance) of stool
Adapted from current guidelines [11, 12, 137].

Diagnostic challenges

The clinical manifestations of IBD are not specific and no pathognomonic sign or symptom exists. In patients with chronic or recurrent abdominal pain and diarrhoea, suspicion of IBD should be raised, and endoscopy with histological sampling should be performed. Nevertheless, selecting patients for endoscopy solely based on symptoms is not reliable, and many patients with suspected IBD will have negative findings on endoscopy [24]. A substantial portion of patients with negative findings will, suffer from functional gastrointestinal disorders, e.g., irritable bowel syndrome (IBS) (table 1) [25, 26]. Patients with IBS should be promptly identified, as they tend to undergo repeated endoscopies for abdominal complaints that are, in fact, unnecessary. On the other hand, diagnosing IBS using Rome criteria could lead to misdiagnosis. As many as 1/3 of patients with IBD also fulfil Rome criteria for IBS [27]. Although, in general, month-long diagnostic delays in IBD seem to be common, a considerable portion of patients experience long delays (>12 months), especially those with CD [28]. In children, timely diagnosis is of even greater importance as IBD may affect growth and sexual maturation [29].

The evaluation and risk stratification of patients using a simple, non-invasive and inexpensive test would be highly desirable. An ideal marker would be sensitive, thereby reliably detecting intestinal inflammation, yet still afford a good specificity that avoids unnecessary investigations. More than a decade ago, measurement of calprotectin in faeces was proposed as a surrogate marker of intestinal inflammation and has been extensively studied since.

Distinguishing organic disease from non-organic disorders

Several studies have investigated the value of faecal calprotectin in distinguishing organic from non-organic gastrointestinal disease in symptomatic patients [27, 29–43]. In a pioneer study, Tibble et al. measured the faecal calprotectin values of 602 patients with symptoms suggestive of IBS or organic intestinal disease who underwent invasive diagnostic imaging with barium enteroclysis, barium enema and/or colonoscopy [27]. Patients with non-organic disease, mainly IBS, had lower faecal calprotectin than did patients with organic disease, e.g., IBD, small bowel enteropathy, microscopic/collagenous colitis, infectious diarrhoea, diverticular disease, or cancer. The sensitivity and specificity of faecal calprotectin in identifying organic disease was 89% and 79%, respectively, and equally valid to Rome I criteria. In another study, Carroccio et al. investigated 120 patients with chronic diarrhoea [34]. Faecal calprotectin identified organic causes of diarrhoea with a 64% sensitivity and an 80% specificity. False positive results were associated with the use of aspirin or non-steroidal anti-inflammatory drugs while false negative results mainly included patients suffering from coeliac disease. In a recent prospective multicentre study by Meucci et al. including 870 unselected patients referred for colonocscopy, faecal calprotectin had an 89% sensitivity and a 62% specificity in identifying any organic disease. In the 416 patients with abnormal findings, colorectal cancer was found in 34, colorectal polyps in 244 (1 cm in 192, ≥1 cm in 52), inflammatory bowel disease in 102 (43 active UC, 30 UC in remission, 19 active CD, 10 CD in remission), diverticula with peri-diverticular inflammation in 12, ischaemic colitis in 2 patients, and miscellaneous diagnoses in the remaining 24 [42]. In a meta-analysis that included 2475 patients, Gisbert et al. calculated a mean sensitivity and specificity of 83% and 84%, respectively, for faecal calprotectin in distinguishing between organic and non-organic disease [44]. Faecal calprotectin’s diagnostic accuracy has been reported as being higher than that of C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), or a combination of the two [27, 31].

In recent years, several studies have highlighted the importance of faecal calprotectin testing, not only for organic disease of the colon, but also of the small-bowel [45–47] and the upper gastrointestinal tract [43]. In the only study that systematically investigated the use of faecal calprotectin in the upper gastrointestinal tract, its diagnostic accuracy was reported as being highly valuable, though with a slightly lower sensitivity and specificity than for colonic inflammation [43]. In this lively field of research, more data on the diagnostic performance of faecal calprotectin will emerge and further define its role as a non-invasive test for the presence of gastrointestinal tract inflammation.

Key Message Calprotectin in faeces is a reliable surrogate marker of intestinal inflammation throughout the gastrointestinal tract. It is useful in differentiating between organic and non-organic gastrointestinal disease.

Identifying inflammatory bowel disease

Consistently higher faecal calprotectin levels have been reported in both adult and paediatric patients with IBD compared to patients with IBS or healthy controls [27, 29, 31, 32, 34–37, 48–66]. Table 2 summarises all studies that investigated the diagnostic performance of faecal calprotectin in identifying patients with IBD. Tibble et al. first validated calprotectin in faeces as a marker of intestinal inflammation by comparing the faecal excretion of 111indium-labelled leukocytes to faecal calprotectin in 22 patients with CD; they then assessed the sensitivity of faecal calprotectin in detecting established CD in 111 patients [31]. In the same study, they further investigated the diagnostic accuracy of faecal calprotectin in 220 consecutive patients who were referred for assessing whether they had IBD or IBS. Using a cut-off of 30 mg/l, calprotectin had a 100% sensitivity and a 97% specificity in discriminating between CD and IBS. In another study including 148 patients referred for lower gastrointestinal symptoms, Chung-Faye et al. found a slightly lower sensitivity and specificity (80% and 74%, respectively), for faecal calprotectin in identifying IBD [57]. Langhorst et al. assessed faecal levels of calprotectin in 139 (54 IBS, 42 UC, 43 CD) patients undergoing diagnostic ileocolonoscopy [62]. Calprotectin had a high sensitivity and specificity in identifying IBD (81.7% and 83.5, respectively), with a slightly superior diagnostic accuracy for CD (81.4%) compared to UC (78.6%). In a recent meta-analysis, von Roon et al. summarised data from 30 studies that included 5983 patients [27, 31–37, 48–51, 53–55, 67–76, 77]. Faecal calprotectin was higher in IBD patients than in non-IBD patients (by 219 μg/g), and showed excellent pooled sensitivity and specificity rates in distinguishing between these groups (95% and 91%, respectively). Other studies [27, 29] have reported higher faecal calprotectin levels for CD than for UC (p = 0.04), but the clinical value of this finding seems questionable as levels varied widely among patients and studies, and no clear-cut distinction between the two disorders were identified.

Most of these studies compared IBD patients with either IBS patients or healthy volunteers, i.e., the extremes of the clinical spectrum. This could overestimate the diagnostic accuracy of the test and impair its usefulness in clinical practice. Van Rheenen et al., in an excellent meta-analysis, compared the diagnostic accuracy of faecal calprotectin in the evaluation of patients with suspected IBD [78]. Thirteen studies summarising data of 1041 patients (670 adults, 371 children) were included [29, 31–33, 37, 56, 60, 61, 63–65, 79, 80]. Studies were selected for their methodological robustness and had to present a paired design where faecal calprotectin values were measured prior to endoscopy. Pooled sensitivity and specificity rates of calprotectin testing were 93% and 96%, respectively. The specificity in children and teenagers was significantly lower (76%). In adults, using faecal calprotectin as a diagnostic test in suspected IBD for deciding upon endoscopy would result in a 67% reduction in patients requiring endoscopy, but would also result in a delayed diagnosis for 6% of patients due to false negative test results.

The diagnostic accuracy of faecal calprotectin for IBD appears to be fairly similar to lactoferrin, another neutrophilic marker measured in faeces [59, 62], and is greatly superior to serum markers, such as CRP, ESR, anti-neutrophil cytoplasmatic antibody (ANCA) and anti-Saccharomyces cerevisiae antibody (ASCA). A number of studies have evaluated ESR and CRP [29, 31, 52] in the diagnosis of IBD and have reported high specificity, both for CRP (78–100%) and ESR (78–100%), though with a markedly lower sensitivity than faecal markers (35–40% and 18–52%, respectively). Schoepfer et al. prospectively compared faecal calprotectin levels with measurement of CRP and IBD antibodies (ANCA, ASCA) in 136 patients [63]. The goal of this study was the non-invasive discrimination between IBD and IBS patients. The overall diagnostic ability of CRP was only moderate (64%) and had a lower sensitivity for UC (52%) than for CD (73%). ANCA and ASCA were highly specific for the presence of IBD, but their diagnostic ability was limited by their low sensitivity. Even when IBD antibody and faecal calprotectin testing were combined, the overall accuracy in distinguishing between IBD and IBS patients improved only minimally.

Key Message Faecal calprotectin levels are elevated in patients with active IBD. Calprotectin testing shows excellent diagnostic accuracy in patients with suspected IBD.

Table 2: Diagnostic precision of studies investigating the use of faecal calprotectin in distinguishing inflammatory bowel disease (IBD) from non-IBD.
Author No. of patients Patient population Cut-off value (µg/g) Sensitivity Specificity PPV (%) NPV (%)
Adult patients              
Tibble, JA [31] 220 CD 30 1.00 0.97 86 100
Limburg, PJ [32] 110 UC/CD 100 0.94 0.83 63 93
Carroccio, A [34] 70 CD 170 1.00 0.95 75 100
Costa, F [35] 239 UC/CD 50 0.81 0.82 88 74
Wassell, J [54] 50 CD 90 0.85 100 100 87
Chung-Faye, G [57] 148 UC/CD 25 0.80 0.74 87 65
Kaiser, T [58] 171 UC/CD 50 0.63 0.86 90 51
D’Incà, R [59] 144 UC/CD 80 0.79 0.74 92 53
Schroder, O [60] 88 UC/CD 15 0.93 1.00 100 91
Schoepfer, AM [61] 74 UC/CD 50 0.83 1.00 100 77
Langhorst, J [62] 139 UC/CD 50 0.82 0.84 89 74
Schoepfer, AM [63] 136 UC/CD 50 0.83 1.00 100 74
Paediatric patients              
Canani, RB [29] 49 UC/CD 95 0.93 0.89 91 91
Fagerberg, UL [37] 36 UC/CD 50 0.95 0.93 95 93
Bunn, SK [51] 68 UC/CD 50 0.65 1.00 100 71
Kolho, KL [56] 57 UC/CD 50 100 0.48 69 100
Sidler, MA [64] 61 UC/CD 50 1.00 0.64 72 100
Ashorn, S [65] 73 UC/CD 100 0.89 0.90 97 67
Diamanti, A [66] 626 UC/CD 160 1.00 0.80 54 100
For each study, the number of included patients (No. of patients) and the type of inflammatory bowel disease (IBD) is given: Ulcerative colitis (UC) or Crohn’s Disease (CD), or both (UC/CD). Cut-off values for the following are given to distinguish between IBD and non-IBD: calprotectin, sensitivities, specificities and positive (PPV, %) and negative predictive value (NPV, %).

Monitoring disease activity

As the clinical course of these chronic, remitting and relapsing conditions often changes, the assessment of the severity of IBD and the monitoring of disease activity are important issues, both in CD and UC [81]. Symptoms of colonic inflammation are often unspecific, complicating the evaluation of IBD’s clinical activity. A variety of activity indices, such as the Crohn’s Disease Activity Index (CDAI) and the Harvey-Bradshaw index, have been developed [82, 83]. These scores use a combination of symptoms, clinical examination results, and laboratory values to quantify IBD activity, but they have two major drawbacks: First, the collection of data is tedious and, second, they depend strongly on subjective patient symptoms. Accordingly, they are used in clinical studies, but rarely in clinical practice. Laboratory values measuring systemic inflammatory parameters (leukocytes, CRP, ESR) to detect IBD show a low sensitivity and specificity and the correlation with symptoms and activity indices is poor [62, 75, 84–89]. Currently, endoscopy with biopsy is still considered the gold standard for the evaluation of mucosal inflammation [11, 12], and a number of scores exist to assess endoscopic activity in CD [90–92] and UC [93–102]. However, endoscopy is invasive, costly, and uncomfortable for patients. To overcome these limitations, a non-invasive marker to monitor activity of intestinal inflammation in IBD patients would be welcome.

In recent years, growing interest in the value of faecal calprotectin for disease monitoring has led to a number of studies investigating its correlation with the degree of IBD activity, as measured by endoscopy (summarised in table 3) and histology [31, 33, 35, 38, 49, 51, 59, 62, 85–89, 103–113]. Langhorst et al. reported faecal calprotectin to be valuable in differentiating between active and inactive IBD, and between IBD and IBS patients [62]. Calprotectin was able to identify active IBD and was superior to CRP and activity indices in detecting endoscopic inflammation. The location of colonic inflammation seems to be irrelevant, though the correlation is better for colonic than for ileal disease activity [49, 108, 109]. In a recent study by Ricanek et al., faecal calprotectin levels correlated highly with endoscopic activity scores in patients with suspected IBD, but correlated inconsistently with clinical activity scores, especially in CD [89]. Schoepfer et al. reported similar results in patients with UC [88]. Using the Rachmilewitz index [99], endoscopic disease activity correlated best with faecal calprotectin (R = 0.834), followed by the Clinical Activity Index (R = 0.672), CRP (R = 0.503), and blood leukocytes (R = 0.461). The overall accuracy for calprotectin in detecting endoscopically active disease was 89%, and was the only marker to discriminate inactive, mild, moderate, and highly active disease. In another study, Sipponen et al. investigated 77 patients with CD who underwent colonoscopy, and compared endoscopic disease activity, as scored by the Crohn’s Disease Index of Severity (CDEIS), with a clinical index (CDAI) [109]. Faecal calprotectin correlated well with CDEIS (R = 0.729) and had a 70% sensitivity and a 92% specificity in predicting endoscopically active disease (CDEIS ≥3), whereas the sensitivity of CDAI ≥150 was only 27%, but with a somewhat higher specificity (94%). In general, faecal calprotectin values correlate better with endoscopic findings than with clinical activity. Accordingly, this sensitive marker may detect residual inflammatory activity in patients with presumably quiescent disease [114].

Key Message Faecal calprotectin levels correlate well with endoscopic and histological disease activity. In CD, the correlation is better for colonic than for ileal disease.

Table 3: Studies investigating the correlation between faecal calprotectin levels and endoscopic IBD activity.
Author No. of patients/ endoscopies Patient population Endoscopic Activity Index Correlation with calprotectin
Bunn, SK [33] 22/22 UC/CD Saverymuttu score 0.75
D’Incà, R [59] 46/46 UC Mayo score 0.51
D’Incà, R [59] 31/31 CD SES-CD 0.48
Langhorst, J [62] 42/42 UC Rachmilewitz index 0.49
Langhorst, J [62] 43/43 CD SES-CD 0.35
Røseth, AG [49] 62/64 UC Mayo score 0.57
Jones, J [87] 164/164 CD SES-CD 0.72
Schoepfer, AM [88] 140/140 CD CDEIS 0.75
Denis, MA [85] 28/28 CD CDEIS not significant
Xiang, JY [86] 66/66 UC Sutherland criteria 0.87
Hanai, H [104] 31/31 UC Matts’ index 0.81
Aomatsu, T [112] 17/17 UC Matts’ index 0.84
Aomatsu, T [112] 18/18 CD SES-CD 0.76
Langhorst, J [106] 31/31 UC Rachmilewitz index 0.51
Sipponen, T [108] 61/87 CD SES-CD 0.64
Fagerberg, UL [107] 39/39 UC/CD Study score * 0.52
Sipponen, T [109] 77/106 CD CDEIS 0.73
Schoepfer, AM [111] 134/134 UC Rachmilewitz index 0.83
Sipponen, T [110] 15/15 CD CDEIS 0.83
For each study, the number of included patients and the number of endoscopies (No. of patients/endoscopies), the type of inflammatory bowel disease (IBD), the endoscopic activity index (for UC: Mayo score [97], Matts’ index [94], Sutherland criteria [98], Saverymuttu score [138], Rachmilewitz index [99]; for CD: Crohn’s Disease endoscopic index of severity [CDEIS] [90], simple endoscopic score for Crohn’s Disease [SES-CD] [92]), and the correlation coefficient (Spearman’s correlation, R) are given. *Colonoscopy scoring system of macroscopic inflammation in 8 colonic segments. Adapted from [139].

Monitoring response to treatment

The evaluation of treatment response in IBD has been based primarily on symptoms, clinical scores and serum markers of inflammation. Data on the value of faecal calprotectin in this setting have long been scarce [103, 115] and are, only now, becoming evident [110, 116–120]. Wagner et al. investigated 38 (11 CD, 27 UC) patients with active IBD and measured their response to treatment using the Harvey-Bradshaw clinical activity index for CD, and for UC, a semi-quantitative four-grade (normal, mild, moderate and severe) scale to assess endoscopic activity [116]. Patients were treated with 5-aminosalicyclic acid (5-ASA) or various combinations of 5-ASA, prednisone, and azathioprine. None received anti-tumour necrosis factor alpha (TNFα) agents. After 8 weeks, 82% of patients had normal endoscopy and normalisation of calprotectin levels were 100% predictive for complete response to treatment.

The response of CD patients to anti-TNF agents was investigated by Sipponen et al. [110]. In 15 patients from a previously-published cross-sectional study [109] who were considered in need of anti-TNF treatment for acute flare-ups (N = 6), chronic active disease (N = 6) or rapid reccurrence of the disease postoperatively (N = 3), colonoscopy was performed at baseline and 12 weeks after induction treatment with 5 mg/kg intravenous Infliximab (week 0 and week 8) in 14 patients, and 40 mg Adalimumab subcutaneously (week 0, 2, 4, 6, 8) in 1 patient. The endoscopic post-treatment activity (by CDEIS) correlated highly with faecal calprotectin values (R = 0.831). Using 200 μg/g as a cut-off value, calprotectin had an 87% sensitivity and a 100% specificity in predicting endoscopically active disease (CDEIS ≥3). In 90 patients with acute severe ulcerative colitis, Ho et al. studied how well faecal calprotectin could predict those patients requiring colectomy and those who would not respond to corticosteroid or Infliximab treament [118]. Calprotectin levels were higher only in patients requiring colectomy (P = 0.04), but not in corticosteroid (P = 0.08) and Infliximab non-responders (P = 0.06). In two studies involving paediatric patients with UC (N = 24 and N = 128), the Pediatric UC Activity Index (PUCAI), a clinical score, predicted treatment response and long-term outcome more accurately than did faecal calprotectin [119, 120]. However, in these studies by Turner et al., treatment decisions might have been guided by symptoms also assessed by the PUCAI, and it might therefore be difficult for calprotectin to outperform the prognostic value of clinical symptoms. This contrasts with results cited by Langhorst [62] from adult patients where calprotectin better identified active vs inactive IBD than did activity indices.

Key Message Low faecal calprotectin levels after treatment indicate response of endoscopic disease activity better among adult than paediatric patients.

Establishing mucosal healing

Growing evidence suggests that MH indicates controlled IBD activity with a more favourable course; surprisingly, episodic clinical remission is a poorer marker of clinical outcome [121–125]. Baert et al. showed that, after 2 years of treatment, endoscopic activity of mucosal inflammation in CD could predict the clinical course for the next 2 years [126]. The relapse rate in patients with MH was lower than in patients with residual mucosal inflammation (32% vs 65%, P = 0.004). In 214 patients with CD (31 (14.5%) primary non-responders excluded) receiving anti-TNFα therapy, MH (achieved in 68%) predicted long-term sustained clinical benefit (65% of the 68% achieving MH vs 40% of those who did not, P <0.001 by logrank), reduced the need for major abdominal surgery (14% among those patients with MH vs 38% of those without MH, P <0.001) and hospitalisation (42% with MH vs 59% of those without MH, P = 0.002) during a median of 69 months follow-up [127]. In a large population-based, cohort study with 495 (354 UC, 141 CD) patients who had endoscopy at baseline and after 1 and 5 years, MH was associated with a 60% reduction in surgery among CD patients, and with a lower rate of colectomy among UC patients (2% vs 7%, P = 0.02) [128]. As explained above, faecal calprotectin correlates highly with endoscopic activity of IBD, especially when calprotectin values are normal. Accordingly, it might be considered a surrogate marker of MH. Røseth et al. performed colonoscopies in IBD patients in clinical remission that had normal faecal calprotectin values and found normal mucosal histology, eg, mucosal healing, in 38 of 45 [103]. Similarly, Sipponen et al. showed that faecal calprotectin values normalised in CD patients who achieved endoscopic remission after anti-TNFα treatment [110, 117].

Key Message Mucosal healing seems to indicate controlled IBD activity. It has been associated with sustained clinical remission as well as reduced rates of hospitalisation and surgical resection. Data on faecal calprotectin as a surrogate marker of MH are emerging, but the evidence is not yet conclusive.

Prediction of IBD relapse

The natural course of IBD is typically characterised by recurrent episodes of disease relapse with exacerbated intestinal inflammation and remissions. However, the disease course may vary substantially and include patients suffering from chronic active disease, patients with recurrent episodes of disease relapse, and even patients who remain in remission. Most IBD patients with clinically quiescent disease seem to have some degree of residual mucosal inflammation [129] and elevated faecal calprotectin levels have been detected in patients in clinical remission [114]. Several other studies have shown that values of faecal calprotectin predict relapse in patients with IBD within 12 months (table 4) [68, 74, 114, 130–134]. In a pioneer study, Tibble et al. demonstrated that, among 80 IBD (43 CD, 37 UC) patients in clinical remission, faecal calprotectin levels of patients who experienced clinical relapse (as measured by clinical activity scores) were higher than those who remained in remission [68]. Calprotectin predicted clinical relapse with a 90% sensitivity and an 83% specificity. In a prospective multicentre study, Gisbert et al. included 163 (89 CD, 74 UC) IBD patients in clinical remission [133]. Sixteen patients (9.8%) experienced a clinical relapse within the 12 months follow-up, and faecal calprotectin values at inclusion were higher in those patients with clinical relapse later on (239 μg/g vs 136 μg/g, p <0.001). The risk of relapse was 30% if calprotectin levels were >150 μg/g and 7% if values were <150 μg/g (p <0.001). In a study by Costa et al., median faecal calprotectin levels were higher only in UC patients who relapsed, but not in those with CD. Accordingly, the risk of relapse within 12 months was increased 14-fold in UC patients with faecal calprotectin levels >150 μg/g but only two-fold in CD patients [74]. Results from another study by D’Incà et al. report that median faecal calprotectin levels in CD patients experiencing a clinical relapse within 12 months did not differ from non-relapse patients (P = 0.055) [130]. Only in the subgroup of patients with colonic CD were calprotectin levels significantly different (177 mg/kg vs 75 mg/kg, P = 0.04). Recently, Kallel et al. studied 53 CD patients in clinical remission, specifically excluding patients with small-bowel CD [134]. Within 12 months follow-up, 18% developed clinical relapse. Calprotectin values were higher in the relapse group (381 μg/g vs 155 μg/g, respectively, p <0.001). Using 340 μg/g as the cut-off, faecal calprotectin provided an 80% sensitivity and a 91% specificity in predicting clinical relapse, which corresponded to an 18-fold risk increase.

Key Message Faecal calprotectin levels <150 μg/g indicate IBD remission with a low risk of relapse. Reports from prospective intervention studies using calprotectin-guided therapy strategies to investigate the long-term outcome of IBD are not yet available.

Table 4: Studies investigating the use of faecal calprotectin to predict relapse of IBD activity.
Author No. of patients Patient population Calprotectin cut-off (μg/g) Relapse rate below cut-off (%) Relapse rate above cut-off (%)
Costa, F [74] 41 UC >150 10 81
Costa, F [74] 38 CD >150 57 87
Tibble, J [68] 37 UC >50 10 85
Tibble, J [68] 43 CD >50 15 85
Sipponen, T [114] 72 UC/CD >100 25 39
D’Incà R [130] 97 UC >130 21 59
D’Incà R [130] 65 CD >130 20 43
Gisbert, JP [133] 163 UC >150 9 31
Gisbert, JP [133] 163 CD >150 8 30
Diamanti, A [132] 73 UC/CD >275 1 84
Walkiewicz, D [131] 32 CD >400 11 56
Kallel, L [134] 53 CD >340 5 67
For each study, the number of included patients (No. of patients), the patient population (ulcerative colitis [UC], Crohn’s disease [CD], or both [UC/CD]), the calprotectin value (μg/g) used as cut-off, and relapse rates (%) of patients below and above the calprotectin cut-off are given. Adapted from [139].

Discussion

Measurement of faecal calprotectin has been shown to differentiate IBD from IBS. Its high sensitivity and high negative predictive value have proven especially useful in ruling out IBD in undiagnosed, symptomatic patients with abdominal pain or diarrhoea. Unfortunately, the specificity of faecal calprotectin testing in identifying IBD is lower than desirable, as several other organic intestinal disorders can also show increased calprotectin levels. Difficulties in comparing results from published studies about the diagnostic value of faecal calprotectin arise from different limits for defining a positive test. Most studies used 50 µg/g as a cut-off, as recommended by the test manufacturers [35, 37, 51, 56, 58, 61, 63, 64], while others based their cut-off on receiver operating characteristics analysis [31, 32, 34, 57, 59, 66], on the 95th percentile of values in healthy subjects [29, 60], or on previous investigations [65]. Most recent studies [58, 61–64] have used 50 µg/g as the cut-off to define a positive test result and to decide on endoscopy in patients with abdominal discomfort to rule out IBD or other organic pathologies. It has been calculated that the use of faecal calprotectin as a diagnostic test in suspected IBD would result in a 67% reduction in patients requiring endoscopy. However, the consequences of misdiagnosis leading to a delayed start of appropriate treatment must be balanced against the number of ultimately unnecessary invasive endoscopic procedures. Figure 1 offers an algorithm to investigate patients with suspected inflammatory bowel disease that includes faecal calprotectin measurement before endoscopy.

Figure 1

A diagnostic algorithm for the evaluation of patients with suspected inflammatory bowel disease that includes faecal calprotectin measurement before endoscopy.

To date, data on the use of faecal calprotectin in IBD diagnosis are almost exclusively gathered from studies carried out in large GI clinics and referral centres. Data on the value of calprotectin testing in primary care are scarce, and can only be extrapolated from data available from tertiary care facilities. In primary care, the emphasis is on ruling out IBD and test characteristics focus on sensitivity. Estimating a lower disease prevalence in primary care than in referral centres, the negative predictive value of normal calprotectin values would significantly increase and allow one to rule out IBD or to adopt a strategy of watchful waiting. Recently, the cost-effectiveness of faecal calprotectin testing to rule out IBD has been demonstrated in hypothetical economic models [135, 136]. Using cut-off values of 50 µg/g and 100 µg/g, the estimated demand for colonoscopies was reduced by 50% and 67%, respectively [135]. However, these data need confirmation from large multi-centre studies including primary care populations.

The correlation between faecal calprotectin levels and endoscopic and histological IBD disease activity has been well established. Calprotectin consistently performed better than clinical indices and serum markers in assessing mucosal inflammation. Currently, endoscopy with mucosal biopsy is considered the gold standard for evaluating the extent and severity of disease activity. However, endoscopy is an expensive and invasive procedure that is onerous to the patient. Faecal calprotectin allows a non-invasive monitoring of disease activity, especially advantageous when the dynamics of repeated measurements are considered. Recently, symptom-based clinical activity indices for defining IBD remission have been challenged and, among both CD and UC patients, MH has been proposed as better identifying controlled disease activity. MH has been associated with sustained clinical remission, as well as reduced rates of hospitalisation and surgical resection. Several smaller studies have shown normal faecal calprotectin values in patients with endoscopic remission. Nevertheless, the current data are not yet conclusive enough to establish faecal calprotectin as a surrogate marker for MH.

It has also been shown that elevated faecal calprotectin levels in patients in clinical remission increase the risk of disease relapse within 12 months follow-up. In most clinically quiescent IBD, residual mucosal inflammation is still present to some extent. When disease activity increases, clinical symptoms are usually not present during the early relapse stage, and patients become symptomatic only later, when intestinal inflammation has been well established. Faecal calprotectin seems to be able to detect subclinical mucosal inflammation, and thus might earlier identify those patients at risk for IBD relapse.

In conclusion, measurement of faecal calprotectin is highly useful for the diagnosis and disease monitoring of patients with IBD, and might additionally predict disease outcome. Future studies should evaluate the value of faecal calprotectin testing to guide treatment decisions and assess their effect on long-term outcome. Precisely this topic is currently being investigated with the CALM Study, a 56-week randomised, open-label, multi-centre efficacy and safety study that evaluates two treatment algorithms in subjects with moderate to severe Crohn’s disease. Patients are treated with prednisone, azathioprine and adalimumab using tight control management, including measurement of faecal calprotectin, or a clinically-driven management route. The primary endpoint of the study is MH after 56 weeks. Pending results from this and similar studies likely to be carried out in the near future, faecal calprotectin awaits confirmation of its value in changing disease outcome through earlier recognition, and treatment monitoring.

Acknowledgements:We thank Mrs Kathleen Bucher for excellent editorial assistance.

References

  1 Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology. 2011;140(6):1704–12 e1702.

  2 Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140(6):1720–28 e1723.

  3 Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology. 2011;140(6):1729–37.

  4 Kaser A, Blumberg RS. Autophagy, microbial sensing, endoplasmic reticulum stress, and epithelial function in inflammatory bowel disease. Gastroenterology. 2011;140(6):1738–47 e1732.

  5 Lakatos PL. Recent trends in the epidemiology of inflammatory bowel diseases: up or down? World J Gastroenterol. 2006;12(38):6102–8.

  6 Gismera CS, Aladren BS. Inflammatory bowel diseases: a disease (s) of modern times? Is incidence still increasing? World J Gastroenterol. 2008;14(36):5491–8.

  7 Benchimol EI, Guttmann A, Griffiths AM, Rabeneck L, Mack DR, Brill H, et al. Increasing incidence of paediatric inflammatory bowel disease in Ontario, Canada: evidence from health administrative data. Gut. 2009;58(11):1490–7.

  8 Auvin S, Molinie F, Gower-Rousseau C, Brazier F, Merle V, Grandbastien B, et al. Incidence, clinical presentation and location at diagnosis of pediatric inflammatory bowel disease: a prospective population-based study in northern France (1988–1999). J Pediatr Gastroenterol Nutr. 2005;41(1):49–55.

  9 Kelsen J, Baldassano RN. Inflammatory bowel disease: the difference between children and adults. Inflamm Bowel Dis. 2008;14(Suppl 2):S9–11.

10 Stange EF, Travis SP, Vermeire S, Beglinger C, Kupcinkas L, Geboes K, et al. European evidence based consensus on the diagnosis and management of Crohn’s disease: definitions and diagnosis. Gut. 2006;55(Suppl 1):i1–15.

11 Stange EF, Travis SP, Vermeire S, Reinisch W, Geboes K, Barakauskiene A, et al. European evidence-based Consensus on the diagnosis and management of ulcerative colitis: Definitions and diagnosis. J Crohns Colitis. 2008;2(1):1–23.

12 Van Assche G, Dignass A, Panes J, Beaugerie L, Karagiannis J, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Definitions and diagnosis. J Crohns Colitis. 2010;4(1):7–27.

13 Henriksen M, Jahnsen J, Lygren I, Sauar J, Schulz T, Stray N, et al. Change of diagnosis during the first five years after onset of inflammatory bowel disease: results of a prospective follow-up study (the IBSEN Study). Scand J Gastroenterol. 2006;41(9):1037–43.

14 Bruining DH, Siddiki HA, Fletcher JG, Tremaine WJ, Sandborn WJ, Loftus EV, Jr.: Prevalence of penetrating disease and extraintestinal manifestations of Crohn’s disease detected with CT enterography. Inflamm Bowel Dis. 2008;14(12):1701–6.

15 Bernstein CN, Blanchard JF, Rawsthorne P, Yu N. The prevalence of extraintestinal diseases in inflammatory bowel disease: a population-based study. Am J Gastroenterol. 2001;96(4):1116–22.

16 Danese S, Semeraro S, Papa A, Roberto I, Scaldaferri F, Fedeli G, et al. Extraintestinal manifestations in inflammatory bowel disease. World J Gastroenterol. 2005;11(46):7227–36.

17 Larsen S, Bendtzen K, Nielsen OH. Extraintestinal manifestations of inflammatory bowel disease: epidemiology, diagnosis, and management. Ann Med. 2010;42(2):97–114.

18 Vavricka SR, Brun L, Ballabeni P, Pittet V, Prinz Vavricka BM, Zeitz J, et al. Frequency and risk factors for extraintestinal manifestations in the Swiss inflammatory bowel disease cohort. Am J Gastroenterol. 2011;106(1):110–9.

19 Dignass A, Van Assche G, Lindsay JO, Lemann M, Soderholm J, Colombel JF, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Current management. J Crohns Colitis. 2010;4(1):28–62.

20 Travis SP, Stange EF, Lemann M, Oresland T, Bemelman WA, Chowers Y, et al. European evidence-based Consensus on the management of ulcerative colitis: Current management. J Crohns Colitis. 2008;2(1):24–62.

21 Lichtenstein GR, Hanauer SB, Sandborn WJ. Practice Parameters Committee of American College of G: Management of Crohn’s disease in adults. Am J Gastroenterol. 2009;104(2):465–83; quiz 464, 484.

22 Lichtenstein GR, Abreu MT, Cohen R, Tremaine W, American Gastroenterological A: American Gastroenterological Association Institute medical position statement on corticosteroids, immunomodulators, and infliximab in inflammatory bowel disease. Gastroenterology. 2006;130(3):935–9.

23 Manz M, Michetti P, Seibold F, Rogler G, Beglinger C. Treatment algorithm for moderate to severe ulcerative colitis. Swiss Med Wkly. 2011, 141:w13235.

24 Lasson A, Kilander A, Stotzer PO. Diagnostic yield of colonoscopy based on symptoms. Scand J Gastroenterol. 2008;43(3):356–62.

25 Tack J, Talley NJ, Camilleri M, Holtmann G, Hu P, Malagelada J-R, et al. Functional gastroduodenal disorders. Gastroenterology. 2006;130(5):1466–79.

26 Hillila MT, Farkkila MA. Prevalence of irritable bowel syndrome according to different diagnostic criteria in a non-selected adult population. Aliment Pharmacol Ther. 2004;20(3):339–45.

27 Tibble JA, Sigthorsson G, Foster R, Forgacs I, Bjarnason I. Use of surrogate markers of inflammation and Rome criteria to distinguish organic from nonorganic intestinal disease. Gastroenterology. 2002;123(2):450–60.

28 Vavricka SR, Spigaglia SM, Rogler G, Pittet V, Michetti P, Felley C, et al. Systematic evaluation of risk factors for diagnostic delay in inflammatory bowel disease. Inflamm Bowel Dis. 2011.

29 Canani RB, de Horatio LT, Terrin G, Romano MT, Miele E, Staiano A, et al. Combined use of noninvasive tests is useful in the initial diagnostic approach to a child with suspected inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2006;42(1):9–15.

30 Tibble JA, Sigthorsson G, Foster R, Scott D, Fagerhol MK, Roseth A, et al. High prevalence of NSAID enteropathy as shown by a simple faecal test. Gut. 1999;45(3):362–6.

31 Tibble J, Teahon K, Thjodleifsson B, Roseth A, Sigthorsson G, Bridger S, et al. A simple method for assessing intestinal inflammation in Crohn’s disease. Gut. 2000;47(4):506–13.

32 Limburg PJ, Ahlquist DA, Sandborn WJ, Mahoney DW, Devens ME, Harrington JJ, et al. Fecal calprotectin levels predict colorectal inflammation among patients with chronic diarrhea referred for colonoscopy. Am J Gastroenterol. 2000;95(10):2831–7.

33 Bunn SK, Bisset WM, Main MJ, Gray ES, Olson S, Golden BE. Fecal calprotectin: validation as a noninvasive measure of bowel inflammation in childhood inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2001;33(1):14-22.

34 Carroccio A, Iacono G, Cottone M, Di Prima L, Cartabellotta F, Cavataio F, et al. Diagnostic accuracy of fecal calprotectin assay in distinguishing organic causes of chronic diarrhea from irritable bowel syndrome: a prospective study in adults and children. Clin Chem. 2003;49(6 Pt 1):861–7.

35 Costa F, Mumolo MG, Bellini M, Romano MR, Ceccarelli L, Arpe P, et al. Role of faecal calprotectin as non-invasive marker of intestinal inflammation. Dig Liver Dis. 2003;35(9):642–7.

36 Dolwani S, Metzner M, Wassell JJ, Yong A, Hawthorne AB. Diagnostic accuracy of faecal calprotectin estimation in prediction of abnormal small bowel radiology. Aliment Pharmacol Ther. 2004;20(6):615–21.

37 Fagerberg UL, Loof L, Myrdal U, Hansson LO, Finkel Y. Colorectal inflammation is well predicted by fecal calprotectin in children with gastrointestinal symptoms. J Pediatr Gastroenterol Nutr. 2005;40(4):450–5.

38 Garcia Sanchez Mdel V, Gonzalez R, Iglesias Flores E, Gomez Camacho F, Casais Juanena L, Cerezo Ruiz A, et al. Diagnostic value of fecal calprotectin in predicting an abnormal colonoscopy]. Med Clin. (Barc) 2006;127(2):41–6.

39 D’Inca 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(4):429–37.

40 Chung-Faye G, Hayee B, Maestranzi S, Donaldson N, Forgacs I, Sherwood R. Fecal M2-pyruvate kinase (M2-PK): a novel marker of intestinal inflammation. Inflamm Bowel Dis. 2007;13(11):1374–8.

41 Jeffery J, Lewis SJ, Ayling RM. Fecal dimeric M2-pyruvate kinase (tumor M2-PK) in the differential diagnosis of functional and organic bowel disorders. Inflamm Bowel Dis. 2009;15(11):1630–4.

42 Meucci G, D’Inca R, Maieron R, Orzes N, Vecchi M, Visentini D, et al. Diagnostic value of faecal calprotectin in unselected outpatients referred for colonoscopy: A multicenter prospective study. Dig Liver Dis. 2010;42(3):191–5.

43 Manz M, Burri E, Rothen C, Tchanguizi N, Niederberger C, Rossi L, et al. Value of fecal calprotectin in the evaluation of patients with abdominal discomfort: an observational study. BMC Gastroenterol. 2012;12:5.

44 Gisbert JP, McNicholl AG. Questions and answers on the role of faecal calprotectin as a biological marker in inflammatory bowel disease. Dig Liver Dis. 2009;41(1):56–66.

45 Maiden L, Thjodleifsson B, Theodors A, Gonzalez J, Bjarnason I. A quantitative analysis of NSAID-induced small bowel pathology by capsule enteroscopy. Gastroenterology. 2005;128(5):1172–8.

46 Jensen MD, Kjeldsen J, Nathan T. Fecal calprotectin is equally sensitive in Crohn’s disease affecting the small bowel and colon. Scand J Gastroenterol. 2011;46(6):694–700.

47 Koulaouzidis A, Douglas S, Rogers MA, Arnott ID, Plevris JN. Fecal calprotectin: a selection tool for small bowel capsule endoscopy in suspected IBD with prior negative bi-directional endoscopy. Scand J Gastroenterol. 2011;46(5):561–6.

48 Roseth AG, Fagerhol MK, Aadland E, Schjonsby H. Assessment of the neutrophil dominating protein calprotectin in feces. A methodologic study. Scand J Gastroenterol. 1992;27(9):793–8.

49 Roseth AG, Aadland E, Jahnsen J, Raknerud N. Assessment of disease activity in ulcerative colitis by faecal calprotectin, a novel granulocyte marker protein. Digestion. 1997;58(2):176–80.

50 Roseth AG, Schmidt PN, Fagerhol MK. Correlation between faecal excretion of indium-111-labelled granulocytes and calprotectin, a granulocyte marker protein, in patients with inflammatory bowel disease. Scand J Gastroenterol. 1999;34(1):50–4.

51 Bunn SK, Bisset WM, Main MJ, Golden BE. Fecal calprotectin as a measure of disease activity in childhood inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2001;32(2):171–7.

52 Fagerberg UL, Lööf L, Merzoug RD, Hansson L-O, Finkel Y. Fecal calprotectin levels in healthy children studied with an improved assay. J Pediatr Gastroenterol Nutr. 2003;37(4):468–72.

53 Thjodleifsson B, Sigthorsson G, Cariglia N, Reynisdottir I, Gudbjartsson DF, Kristjansson K, et al. Subclinical intestinal inflammation: an inherited abnormality in Crohn’s disease relatives? Gastroenterology. 2003;124(7):1728–37.

54 Wassell J, Dolwani S, Metzner M, Losty H, Hawthorne A. Faecal calprotectin: a new marker for Crohn’s disease? Ann Clin Biochem. 2004;41(Pt 3):230–2.

55 Berni Canani R, Rapacciuolo L, Romano MT, Tanturri de Horatio L, Terrin G, Manguso F, et al. Diagnostic value of faecal calprotectin in paediatric gastroenterology clinical practice. Dig Liver Dis. 2004;36(7):467–70.

56 Kolho KL, Raivio T, Lindahl H, Savilahti E. Fecal calprotectin remains high during glucocorticoid therapy in children with inflammatory bowel disease. Scand J Gastroenterol. 2006;41(6):720–5.

57 Chung-Faye G, Hayee Bh, Maestranzi S, Donaldson N, Forgacs I, Sherwood R. Fecal M2-pyruvate kinase (M2-PK): a novel marker of intestinal inflammation. Inflamm Bowel Dis. 2007;13(11):1374–8.

58 Kaiser T, Langhorst J, Wittkowski H, Becker K, Friedrich AW, Rueffer A, et al. Faecal S100A12 as a non-invasive marker distinguishing inflammatory bowel disease from irritable bowel syndrome. Gut. 2007;56(12):1706–13.

59 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(4):429–37.

60 Schroder O, Naumann M, Shastri Y, Povse N, Stein J. Prospective evaluation of faecal neutrophil-derived proteins in identifying intestinal inflammation: combination of parameters does not improve diagnostic accuracy of calprotectin. Aliment Pharmacol Ther. 2007;26(7):1035–42.

61 Schoepfer AM, Trummler M, Seeholzer P, Criblez DH, Seibold F. Accuracy of four fecal assays in the diagnosis of colitis. Dis Colon Rectum. 2007;50(10):1697–706.

62 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(1):162–9.

63 Schoepfer AM, Trummler M, Seeholzer P, Seibold-Schmid B, Seibold F. Discriminating IBD from IBS: comparison of the test performance of fecal markers, blood leukocytes, CRP, and IBD antibodies. Inflamm Bowel Dis. 2008;14(1):32–9.

64 Sidler MA, Leach ST, Day AS. Fecal S100A12 and fecal calprotectin as noninvasive markers for inflammatory bowel disease in children. Inflamm Bowel Dis. 2008;14(3):359–66.

65 Ashorn S, Honkanen T, Kolho KL, Ashorn M, Valineva T, Wei B, et al. Fecal calprotectin levels and serological responses to microbial antigens among children and adolescents with inflammatory bowel disease. Inflamm Bowel Dis. 2009;15(2):199–205.

66 Diamanti A, Panetta F, Basso MS, Forgione A, Colistro F, Bracci F, et al. Diagnostic work-up of inflammatory bowel disease in children: the role of calprotectin assay. Inflamm Bowel Dis. 2010;16(11):1926–30.

67 Roseth AG, Kristinsson J, Fagerhol MK, Schjonsby H, Aadland E, Nygaard K, et al. Faecal calprotectin: a novel test for the diagnosis of colorectal cancer? Scand J Gastroenterol. 1993;28(12):1073-6.

68 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(1):15–22.

69 Kronborg O, Ugstad M, Fuglerud P, Johne B, Hardcastle J, Scholefield JH, et al. Faecal calprotectin levels in a high risk population for colorectal neoplasia. Gut. 2000;46(6):795–800.

70 Kristinsson J, Nygaard K, Aadland E, Barstad S, Sauar J, Hofstad B, et al. Screening of first degree relatives of patients operated for colorectal cancer: evaluation of fecal calprotectin vs. hemoccult II. Digestion. 2001;64(2):104–10.

71 Tibble J, Sigthorsson G, Foster R, Sherwood R, Fagerhol M, Bjarnason I. Faecal calprotectin and faecal occult blood tests in the diagnosis of colorectal carcinoma and adenoma. Gut. 200;49(3):402–8.

72 Limburg PJ, Devens ME, Harrington JJ, Diehl NN, Mahoney DW, et al. Prospective evaluation of fecal calprotectin as a screening biomarker for colorectal neoplasia. Am J Gastroenterol. 2003;98(10):2299–305.

73 Hoff G, Grotmol T, Thiis-Evensen E, Bretthauer M, Gondal G, Vatn MH. Testing for faecal calprotectin (PhiCal) in the Norwegian Colorectal Cancer Prevention trial on flexible sigmoidoscopy screening: comparison with an immunochemical test for occult blood (FlexSure OBT). Gut. 2004;53(9):1329–33.

74 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(3):364–8.

75 Gaya DR, Lyon TD, Duncan A, Neilly JB, Han S, Howell J, et al. Faecal calprotectin in the assessment of Crohn’s disease activity. QJM. 2005;98(6):435–41.

76 Bremner A, Roked S, Robinson R, Phillips I, Beattie M. Faecal calprotectin in children with chronic gastrointestinal symptoms. Acta Paediatr. 2005;94(12):1855–8.

77 von Roon AC, Karamountzos L, Purkayastha S, Reese GE, Darzi AW, Teare JP, et al. Diagnostic precision of fecal calprotectin for inflammatory bowel disease and colorectal malignancy. Am J Gastroenterol. 2007;102(4):803–13.

78 van Rheenen PF, Van de Vijver E, Fidler V. Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta-analysis. BMJ. 2010;341:c3369.

79 Otten CM, Kok L, Witteman BJ, Baumgarten R, Kampman E, Moons KG, et al. Diagnostic performance of rapid tests for detection of fecal calprotectin and lactoferrin and their ability to discriminate inflammatory from irritable bowel syndrome. Clin Chem Lab Med. 2008;46(9):1275–80.

80 Perminow G, Brackmann S, Lyckander LG, Franke A, Borthne A, Rydning A, et al. A characterization in childhood inflammatory bowel disease, a new population-based inception cohort from South-Eastern Norway, 2005-07, showing increased incidence in Crohn’s disease. Scand J Gastroenterol. 2009;44(4):446–56.

81 Vatn MH. Natural history and complications of IBD. Curr Gastroenterol Rep. 2009;11(6):481–7.

82 Best WR, Becktel JM, Singleton JW, Kern F, Jr.: Development of a Crohn’s disease activity index. National Cooperative Crohn’s Disease Study. Gastroenterology. 1976;70(3):439–44.

83 Harvey RF, Bradshaw JM. A simple index of Crohn’s-disease activity. Lancet. 1980;1(8167):514.

84 Nielsen OH, Vainer B, Madsen SM, Seidelin JB, Heegaard NH. Established and emerging biological activity markers of inflammatory bowel disease. Am J Gastroenterol. 2000;95(2):359–67.

85 Denis MA, Reenaers C, Fontaine F, Belaiche J, Louis E. Assessment of endoscopic activity index and biological inflammatory markers in clinically active Crohn’s disease with normal C-reactive protein serum level. Inflamm Bowel Dis. 2007;13(9):1100–5.

86 Xiang JY, Ouyang Q, Li GD, Xiao NP. Clinical value of fecal calprotectin in determining disease activity of ulcerative colitis. World J Gastroenterol. 2008;14(1):53–7.

87 Jones J, Loftus EV, Jr., Panaccione R, Chen LS, Peterson S, McConnell J, Baudhuin L, et al. Relationships between disease activity and serum and fecal biomarkers in patients with Crohn’s disease. Clin Gastroenterol Hepatol. 2008;6(11):1218–24.

88 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(1):162–9.

89 Ricanek P, Brackmann S, Perminow G, Lyckander LG, Sponheim J, Holme O, et al. Evaluation of disease activity in IBD at the time of diagnosis by the use of clinical, biochemical, and fecal markers. Scand J Gastroenterol. 2011.

90 Mary JY, Modigliani R. Development and validation of an endoscopic index of the severity for Crohn’s disease: a prospective multicentre study. Groupe d’Etudes Therapeutiques des Affections Inflammatoires du Tube Digestif (GETAID). Gut. 1989;30(7):983–9.

91 Rutgeerts P, Geboes K, Vantrappen G, Beyls J, Kerremans R, Hiele M. Predictability of the postoperative course of Crohn’s disease. Gastroenterology. 1990;99(4):956–63.

92 Daperno M, D’Haens G, Van Assche G, Baert F, Bulois P, Maunoury V, et al. Development and validation of a new, simplified endoscopic activity score for Crohn’s disease: the SES-CD. Gastrointest Endosc. 2004;60(4):505–12.

93 Truelove SC, Witts LJ. Cortisone in ulcerative colitis; final report on a therapeutic trial. Br Med J. 1955;2(4947):1041–8.

94 Matts SG. The value of rectal biopsy in the diagnosis of ulcerative colitis. Q J Med. 1961;30:393–407.

95 Baron JH, Connell AM, Lennard-Jones JE. Variation between Observers in Describing Mucosal Appearances in Proctocolitis. Br Med J. 1964;1(5375):89–92.

96 Powell-Tuck J, Bown RL, Lennard-Jones JE. A comparison of oral prednisolone given as single or multiple daily doses for active proctocolitis. Scand J Gastroenterol. 1978;13(7):833–7.

97 Schroeder KW, Tremaine WJ, Ilstrup DM. Coated oral 5-aminosalicylic acid therapy for mildly to moderately active ulcerative colitis. A randomized study. N Engl J Med. 1987;317(26):1625–9.

98 Sutherland LR, Martin F, Greer S, Robinson M, Greenberger N, Saibil F, et al. 5-Aminosalicylic acid enema in the treatment of distal ulcerative colitis, proctosigmoiditis, and proctitis. Gastroenterology. 1987;92(6):1894–8.

99 Rachmilewitz D. Coated mesalazine (5-aminosalicylic acid) versus sulphasalazine in the treatment of active ulcerative colitis: a randomised trial. BMJ. 1989;298(6666):82–6.

100 Hanauer S, Schwartz J, Robinson M, Roufail W, Arora S, Cello J, et al. Mesalamine capsules for treatment of active ulcerative colitis: results of a controlled trial. Pentasa Study Group. Am J Gastroenterol. 1993;88(8):1188–97.

101 Hanauer SB, Robinson M, Pruitt R, Lazenby AJ, Persson T, Nilsson LG, et al. Budesonide enema for the treatment of active, distal ulcerative colitis and proctitis: a dose-ranging study. U.S. Budesonide enema study group. Gastroenterology. 1998;115(3):525–32.

102 Feagan BG, Greenberg GR, Wild G, Fedorak RN, Pare P, McDonald JW, et al. Treatment of ulcerative colitis with a humanized antibody to the alpha4beta7 integrin. N Engl J Med. 2005;352(24):2499–507.

103 Roseth 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(10):1017–20.

104 Hanai H, Takeuchi K, Iida T, Kashiwagi N, Saniabadi AR, Matsushita I, et al. Relationship between fecal calprotectin, intestinal inflammation, and peripheral blood neutrophils in patients with active ulcerative colitis. Dig Dis Sci. 2004;49(9):1438–43.

105 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(8):707–12.

106 Langhorst J, Elsenbruch S, Mueller T, Rueffer A, Spahn G, Michalsen A, et al. Comparison of 4 neutrophil-derived proteins in feces as indicators of disease activity in ulcerative colitis. Inflamm Bowel Dis. 2005;11(12):1085–91.

107 Fagerberg UL, Loof L, Lindholm J, Hansson LO, Finkel Y. Fecal calprotectin: a quantitative marker of colonic inflammation in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2007;45(4):414–20.

108 Sipponen T, Karkkainen 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(10):1221–9.

109 Sipponen T, Savilahti E, Kolho KL, Nuutinen H, Turunen U, Farkkila M. Crohn’s disease activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn’s disease activity index and endoscopic findings. Inflamm Bowel Dis. 2008;14(1):40–6.

110 Sipponen T, Savilahti E, Karkkainen P, Kolho KL, Nuutinen H, Turunen U, et al. Fecal calprotectin, lactoferrin, and endoscopic disease activity in monitoring anti-TNF-alpha therapy for Crohn’s disease. Inflamm Bowel Dis. 2008;14(10):1392–8.

111 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(12):1851–8.

112 Aomatsu T, Yoden A, Matsumoto K, Kimura E, Inoue K, Andoh A, et al. Fecal calprotectin is a useful marker for disease activity in pediatric patients with inflammatory bowel disease. Dig Dis Sci. 2011.

113 Manolakis AC, Kapsoritakis AN, Tiaka EK, Potamianos SP. Calprotectin, calgranulin C, and other members of the s100 protein family in inflammatory bowel disease. Dig Dis Sci. 2011;56(6):1601–11.

114 Sipponen T, Kolho KL. Faecal calprotectin in children with clinically quiescent inflammatory bowel disease. Scand J Gastroenterol. 2010;45(7-8):872–7.

115 Aadland E, Fagerhol MK. Faecal calprotectin: a marker of inflammation throughout the intestinal tract. Eur J Gastroenterol Hepatol. 2002;14(8):823–5.

116 Wagner M, Peterson CGB, Ridefelt P, Sangfelt P, Carlson M. Fecal markers of inflammation used as surrogate markers for treatment outcome in relapsing inflammatory bowel disease. World J Gastroenterol. 2008;14(36):5584–9; discussion 5588.

117 Sipponen T, Bjorkesten CG, Farkkila 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. 2009.

118 Ho GT, Lee HM, Brydon G, Ting T, Hare N, Drummond H, et al. Fecal calprotectin predicts the clinical course of acute severe ulcerative colitis. Am J Gastroenterol. 2009;104(3):673–8.

119 Turner D, Leach ST, Mack D, Uusoue K, McLernon R, Hyams J, et al. Faecal calprotectin, lactoferrin, M2-pyruvate kinase and S100A12 in severe ulcerative colitis: a prospective multicentre comparison of predicting outcomes and monitoring response. Gut. 2010;59(9):1207–12.

120 Turner D, Mack D, Leleiko N, Walters TD, Uusoue K, Leach ST, et al. Severe pediatric ulcerative colitis: a prospective multicenter study of outcomes and predictors of response. Gastroenterology. 2010;138(7):2282–91.

121 Arnott ID, Watts D, Ghosh S. Review article: is clinical remission the optimum therapeutic goal in the treatment of Crohn’s disease? Aliment Pharmacol Ther. 2002;16(5):857–67.

122 Rutgeerts P, Vermeire S, Van Assche G. Mucosal healing in inflammatory bowel disease: impossible ideal or therapeutic target? Gut. 2007;56(4):453–5.

123 Vatn MH. Mucosal healing: impact on the natural course or therapeutic strategies. Dig Dis. 2009;27(4):470–5.

124 van Assche G, Vermeire S, Rutgeerts P. Mucosal healing and anti TNFs in IBD. Curr Drug Targets. 2010;11(2):227–33.

125 Pineton de Chambrun G, Peyrin-Biroulet L, Lemann M, Colombel JF. Clinical implications of mucosal healing for the management of IBD. Nat Rev Gastroenterol Hepatol. 2010;7(1):15–29.

126 Baert F, Moortgat L, Van Assche G, Caenepeel P, Vergauwe P, De Vos M, et al. Mucosal healing predicts sustained clinical remission in patients with early-stage Crohn’s disease. Gastroenterology. 2010;138(2):463–8; quiz e410–61.

127 Schnitzler F, Fidder H, Ferrante M, Noman M, Arijs I, Van Assche G, et al. Mucosal healing predicts long-term outcome of maintenance therapy with infliximab in Crohn’s disease. Inflamm Bowel Dis. 2009;15(9):1295–301.

128 Froslie KF, Jahnsen J, Moum BA, Vatn MH. Mucosal healing in inflammatory bowel disease: results from a Norwegian population-based cohort. Gastroenterology. 2007;133(2):412–22.

129 Saverymuttu SH. Clinical remission in Crohn’s disease – assessment using faecal 111In granulocyte excretion. Digestion. 1986;33(2):74–9.

130 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(8):2007–14.

131 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(5):669–73.

132 Diamanti A, Colistro F, Basso MS, Papadatou B, Francalanci P, Bracci F, et al. Clinical role of calprotectin assay in determining histological relapses in children affected by inflammatory bowel diseases. Inflamm Bowel Dis. 2008;14(9):1229–35.

133 Gisbert JP, Bermejo F, Perez-Calle JL, Taxonera C, Vera I, McNicholl AG, et al. Fecal calprotectin and lactoferrin for the prediction of inflammatory bowel disease relapse. Inflamm Bowel Dis. 2009;15(8):1190–8.

134 Kallel L, Ayadi I, Matri S, Fekih M, Mahmoud NB, Feki M, et al. Fecal calprotectin is a predictive marker of relapse in Crohn’s disease involving the colon: a prospective study. Eur J Gastroenterol Hepatol. 2010;22(3):340–5.

135 Mindemark M, Larsson A. Ruling out IBD: Estimation of the possible economic effects of pre-endoscopic screening with F-calprotectin. Clin Biochem. 2011.

136 Purchasing CfE-b: Economic report: value of calprotectin in screening out irritable bowel syndrome. 2009.

137 Longstreth GF, Thompson WG, Chey WD, Houghton LA, Mearin F, Spiller RC. Functional bowel disorders. Gastroenterology. 2006;130(5):1480–91.

138 Saverymuttu SH, Camilleri M, Rees H, Lavender JP, Hodgson HJ, Chadwick VS. Indium 111-granulocyte scanning in the assessment of disease extent and disease activity in inflammatory bowel disease. A comparison with colonoscopy, histology, and fecal indium 111-granulocyte excretion. Gastroenterology. 1986;90(5 Pt 1):1121–8.

139 Lewis JD. The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology. 2011;140(6):1817–26 e1812.

Notes

Funding / potential competing interests:Dr. Burri is supported by unrestricted research grants from the Freiwillige Akademische Gesellschaft (Basel, Switzerland) and the Gottfried und Julia Bangerter-Rhyner-Stiftung (Bern, Switzerland). Authors were independent of funding.