Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 28, 2024; 30(24): 3022-3035
Published online Jun 28, 2024. doi: 10.3748/wjg.v30.i24.3022
Fecal calprotectin and endoscopic scores: The cornerstones in clinical practice for evaluating mucosal healing in inflammatory bowel disease
Marcia Henriques de Magalhães Costa, Department of Clinical Medicine, Fluminense Federal University, Rio de Janeiro 24210-200, Brazil
Ligia Yukie Sassaki, Department of Internal Medicine, Medical School, São Paulo State University (Unesp), Botucatu 18618-686, São Paulo, Brazil
Júlio Maria Fonseca Chebli, Division of Gastroenterology, Department of Medicine, University Hospital of The Federal University of Juiz de Fora, University of Juiz de Fora School of Medicine, Juiz de Fora 36036-247, Minas Gerais, Brazil
ORCID number: Marcia Henriques de Magalhães Costa (0000-0002-5636-7501); Ligia Yukie Sassaki (0000-0002-7319-8906); Júlio Maria Fonseca Chebli (0000-0003-1527-0663).
Author contributions: Costa MHM, Sassaki LY, and Chebli JMF contributed to the conception and design of the study, acquisition of data, drafting of the article, and making critical revisions related to the important intellectual content of the manuscript; All of the authors approved the final version of the article to be published.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: Https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Júlio Maria Fonseca Chebli, MD, PhD, Full Professor, Division of Gastroenterology, Department of Medicine, University Hospital of The Federal University of Juiz de Fora, University of Juiz de Fora School of Medicine, Rua Maria Jose Leal, 296, Juiz de Fora 36036-247, Minas Gerais, Brazil. julio.chebli@medicina.ufjf.br
Received: March 3, 2024
Revised: May 1, 2024
Accepted: May 27, 2024
Published online: June 28, 2024
Processing time: 113 Days and 14.6 Hours

Abstract

Managing inflammatory bowel disease (IBD) is becoming increasingly complex and personalized, considering the advent of new advanced therapies with distinct mechanisms of action. Achieving mucosal healing (MH) is a pivotal therapeutic goal in IBD management and can prevent IBD progression and reduce flares, hospitalization, surgery, intestinal damage, and colorectal cancer. Employing proactive disease and therapy assessment is essential to achieve better control of intestinal inflammation, even if subclinical, to alter the natural course of IBD. Periodic monitoring of fecal calprotectin (FC) levels and interval endoscopic evaluations are cornerstones for evaluating response/remission to advanced therapies targeting IBD, assessing MH, and detecting subclinical recurrence. Here, we comment on the article by Ishida et al Moreover, this editorial aimed to review the role of FC and endoscopic scores in predicting MH in patients with IBD. Furthermore, we intend to present some evidence on the role of these markers in future targets, such as histological and transmural healing. Additional prospective multicenter studies with a stricter MH criterion, standardized endoscopic and histopathological analyses, and virtual chromoscopy, potentially including artificial intelligence and other biomarkers, are desired.

Key Words: Fecal calprotectin, Endoscopic scores, Mucosal healing, Histological healing, Ulcerative colitis, Inflammatory bowel diseases

Core Tip: Mucosal healing (MH) is a pivotal goal in inflammatory bowel disease (IBD) management and can prevent IBD relapse. However, assessing MH is challenging due to the poor correlation between symptoms and intestinal inflammation. Fecal calprotectin (FC) levels and interval endoscopic evaluation are cornerstones for achieving this goal. The editorial updates the best predictor tools of IBD relapse in 12 mo concerning FC and endoscopic scores (ESs). In the retrospective single-center study, all three ESs and FC were useful in predicting ulcerative colitis relapse, suggesting the Ulcerative Colitis Endoscopic Index of Severity as the preferred choice for combining feasibility and accuracy.



INTRODUCTION

The management of inflammatory bowel disease (IBD) is becoming more complex and personalized, given the numerous advanced therapies recently incorporated into the therapeutic arsenal for controlling IBD. Achieving mucosal healing (MH) is a pivotal therapeutic goal in IBD management, and it can prevent IBD progression and reduce disease flares, hospitalization rates, the need for surgery, irreversible intestinal damage, and colorectal cancer development[1,2]. However, evaluating an individual therapy-induced MH is difficult. For example, the relapse of intestinal symptoms is usually non-specific and cannot confirm disease activity or a late event indicating persistent or recurrent intestinal inflammation[3]. A critical point is to not rely solely on symptoms to assess therapy-induced MH. Consequently, employing proactive disease and therapy assessment is essential to better control intestinal inflammation, even if subclinical, to alter the natural course of IBD[4]. Furthermore, the periodic monitoring of fecal calprotectin (FC) levels and interval endoscopic evaluation are cornerstones for evaluating response/remission to advanced therapies targeting IBD, assessing MH, and detecting subclinical recurrence[5]

Therefore, in this editorial, we comment on the article by Ishida et al[6], published in a recent issue of the World Journal of Gastroenterology in 2023. They studied the usefulness of FC and endoscopic scores for predicting clinical relapse within 12 mo in 75 patients with ulcerative colitis (UC) in clinical remission. The endoscopic scores studied were the Mayo Endoscopic Subscore (MES), UC Endoscopic Index of Severity (UCEIS), and UC Colonoscopic Index of Severity (UCCIS). In total, 18 patients (24%) experienced clinical relapse during the 12 mo. The cutoff values for predicting relapse were an FC and UCCIS score of ≥ 323 mg/kg and ≥ 10.2, respectively. In summary, the accuracy of the endoscopic scores and biomarkers for predicting relapse was 86.7%, 85.3%, 76.0%, and 73.3% for UCCIS, UCEIS, FC, and MES, respectively. Therefore, the authors concluded that the endoscopic scores studied and FC may predict disease relapse in 12 mo among patients with UC in clinical remission.

In their article[6], the authors included patients with UC in clinical remission measured using the Rachmilewitz score [clinical activity index (CAI ≤ 4)][7]. However, the criteria used to define MH were not overly strict and included patients with mild endoscopic activity, as shown by the MES, UCEIS score, and UCEIS score of 1 (42.7%), 2 (17.3%), and 3 (8.0%), respectively. Furthermore, the criteria used to define clinical relapse, which was the primary outcome, were simply an increase in CAI above baseline, determined by the worsening of diarrhea, abdominal pain, or bloody stools requiring modification or the addition of treatment. To the best of our knowledge and in accord with the consensus and guidelines on IBD[8], the best definition for disease activity, disease remission, or relapse should be based on clinical parameters and endoscopic evaluation or even with the use of the FC, which was not reported in the article. This could underestimate or overestimate the association between the scores and disease activity as the correlation between the presence of symptoms and endoscopic activity is not 100%. Moreover, the authors did not assess FC levels during clinical relapse. Other important comments can be highlighted. First, as a retrospective study, it is crucial to determine whether the scores were explicitly provided in the report or were calculated based on the descriptive findings or image review. Therefore, information regarding the experience of the endoscopists with IBD scores should be considered as significant interobserver variability exists in scoring IBD activity[9]. Lastly, an observation regarding the timing of stool sampling should be considered as some samples were collected the day after the endoscopic procedure (colonic preparation and possible biopsies), which could have influenced the FC results[10].

Despite these limitations, the article by Ishida et al[6] provides important information regarding the association between endoscopic scores and FC for predicting clinical relapse in patients with UC. These findings should encourage disease monitoring, especially in patients with evidence of residual disease activity, such as FC values > 150 mg/kg and mild activity on endoscopy. Another relevant point of the study was the evaluation of the relationship between FC, UCEIS score, and UCCIS score. The data were strengthened by the fact that the researchers analyzed FC and endoscopic scores simultaneously[6].

This editorial aimed to review the role of FC and endoscopic scores in predicting MH in patients with IBD, given the importance of MH as a target in IBD, following the recommendation of the Selecting Therapeutic Targets in IBD II (STRIDE-II) consensus[11]. Furthermore, we intend to present some evidence on the role of these markers in future targets, such as histological and transmural healing (TH).

FECAL BIOMARKERS AND MUCOSAL HEALING IN PATIENTS WITH IBD

FC is a calcium-binding protein comprising a complex of two proteins, S100A8 and S100A9[12], primarily derived from the cytoplasm of neutrophils. It has antibacterial activity and a role in the innate immune response[13]. It is also expressed by monocytes, dendritic cells, activated macrophages, keratinocytes, and some mucosal epithelial cells[14]. FC is an early marker of neutrophil degranulation and appears to be the most sensitive marker of intestinal inflammation in IBD, with concentrations correlated with the extent of mucosal inflammation. This biomarker is closely correlated with endoscopy score and MH[8] and can be measured with several commercially available assays. According to the European Crohn’s and Colitis Organization (ECCO) guidelines[8], FC is indicated for the initial diagnosis of IBD, to differentiate between IBD flare and irritable bowel syndrome (IBS) symptoms, confirm an IBD relapse, and monitor IBD treatment, and is closely associated with MH[15-17] and histological healing[8,11].

In IBD assessment, a recent systematic review and meta-analysis showed that FC was a validated biomarker to distinguish between IBD and IBS with a sensitivity of 85.8% and a specificity of 91.7% . A cutoff value of ≤ 50 μg/g had better sensitivity than a cutoff of > 50 μg/g (87% vs 79%)[18]. Another meta-analysis concluded that the probability of having IBD when the FC value was ≤ 40 μg/g was relatively low (≤ 1%)[19].

FC can also be used to monitor IBD flares in patients in clinical and biochemical remission[8]. Therefore, repeated monitoring of the patient every 3-4 mo is indicated when the FC value is in the estimated target. However, the therapeutic plan must be discussed if the FC exceeds the desired value, and the FC should be remeasured within 1 mo to confirm the previous result. Moreover, repeating the test is recommended to confirm the value before optimizing therapy in case of doubt[8].

Recently, the American Gastroenterological Association (AGA) published guidelines on the role of biomarkers, including C-reactive protein (CRP) and FC, and endoscopy, in the management of Crohn’s Disease (CD)[20] and UC[21]. In CD, monitoring based on biomarkers may be performed every 6-12 mo in patients in symptomatic remission[20].

Despite the recommendation of using FC as a reliable marker, no consensus exists on the ideal cutoff value for IBD monitoring[8]. A meta-analysis published in 2015 included 744 UC patients and 727 CD patients from 13 studies. The FC cutoff value ranged from 30 to 274 μg/g for clinical disease activity[22]. The pooled sensitivity and specificity were 0.92 (0.90-0.94) and 0.60 (0.52-0.67), respectively, for a cutoff value of 50 μg/g, and 0.80 (0.76-0.84) and 0.82 (0.77-0.86), respectively, for a cutoff value of 250 μg/g[22]. Some studies have shown that FC can predict IBD recurrence. A systematic review by Heida et al[23] published in 2017 revealed that an increase in FC level in patients in clinical remission was associated with a 53%-83% likelihood of recurrence within 2-3 mo. Conversely, patients with consecutive normal FC levels had a 67%-94% probability of maintaining clinical remission over the next 2-3 mo[23]. A cutoff value of 150 μg/g was associated with the risk of disease recurrence in both UC and CD. Specifically, patients with an FC of ≥ 150 μg/g had 14-fold and 2-fold increased risks of UC recurrence and CD relapse, respectively, compared with levels below the threshold[24]. A prospective study involving 164 patients with UC with MH observed that an FC cutoff value of 115 μg/g had a sensitivity and specificity of 83% and 81%, respectively, to predict endoscopic relapse within 12 mo and lactoferrin, another possible biomarker, had a sensitivity and specificity of 70% and 79%, respectively at a threshold of 145 μg/g[25]. For patients with UC, the AGA recommends monitoring asymptomatic patients with FC every 6-12 mo. An FC value of < 150 μg/g in patients in symptomatic remission may rule out active inflammation and prevent routine endoscopic assessment of the disease. In contrast, an FC value of < 50 μg/g may be preferred over < 150 μg/g to detect endoscopic improvement in patients with UC who have recently achieved symptomatic remission after treatment adjustment. Repeating the examination within 3-6 mo or performing endoscopic assessment to confirm disease activity is recommended in asymptomatic patients with an FC value of > 150 μg/g. However, for symptomatic patients with UC and with moderate-to-severe symptoms suggestive of a flare-up, an FC value of > 150 μg/g indicates disease activity, and endoscopic assessment can be avoided. An endoscopic assessment is recommended before treatment adjustment in patients with mild symptoms and an FC value of > 150 μg/g[21].

For patients with CD, the AGA recommends using an FC value of < 150 μg/g in patients in symptomatic remission (with recent endoscopic remission) to rule out active inflammation and avoid endoscopic assessment of disease activity. The AGA suggests confirming disease activity in patients without recent confirmation of endoscopic remission by endoscopic evaluation. An endoscopic assessment of disease activity rather than empiric treatment adjustment is recommended in patients presenting with an FC value of > 150 μg/g. Similarly, in patients with CD symptoms and a normal FC value of < 150 μg/g, the AGA suggests endoscopic assessment of disease activity rather than empiric treatment adjustment. This similar approach is suggested for symptomatic patients with CD with a normal FC value of < 150 μg/g[20].

Despite the lack of a published established cutoff value for disease activity in IBD, FC is a more sensitive marker of disease activity than hemoglobin, CRP, and albumin[26,27] for in colonic and small bowel diseases[28,29]. FC, in symptomatic patients, has a good correlation (r > 0.8) with endoscopic disease activity in CD and UC[30,31]. Additionally, FC can predict treatment responses in patients with UC[32,33] and with CD[34]. An FC value of ≤ 168 μg/g was associated with a sustained clinical response at 1 year (sensitivity and specificity of 83% and 74%. respectively), and a value of ≤ 121 μg/g was associated with MH (sensitivity and specificity of 79% and 57%, respectively) in patients with UC after 1 year of treatment with anti-tumor necrosis factor agents[32].

The ECCO consensus recommends the use of FC as a marker of MH in patients with UC who clinically respond to medical therapy approximately 3-6 mo after treatment initiation[8]. Regarding the ideal FC value associated with MH, a systemic review including nine studies showed that the cutoff values of FC ranged between 58 μg/g (sensitivity and specificity of 89.7% and 93.3%, respectively) and 490 μg/g (sensitivity and specificity of 100% and 62%, respectively) for MH detection in patients with UC[35]. In contrast, the cutoff values of FC in patients with CD ranged from 71 μg/g (sensitivity and specificity of 95.9% and 52.3%, respectively) to 918 μg/g (sensitivity and specificity of 50% and 100%, respectively) for MH detection[35]. According to the STRIDE-II guidelines, an FC cutoff value of 150 μg/g can be used to identify MH, and an FC value of < 600 μg/g is associated with minimal inflammation in patients with CD[11]. A meta-analysis published in 2021 included 17 colonic CD and 49 UC studies[36]. In the CD studies, the authors observed a sensitivity and specificity of 0.828 (0.769-0.874) and 0.759 (0.683-0.821), respectively, for FC in MH diagnosis. In the analysis of the receiver operating characteristic (ROC) curve, the area under the ROC curve (AUC) was 0.829. In contrast, the sensitivity and specificity were 0.804 (0.757-0.843) and 0.817 (0.780-0.848), respectively, in the UC studies, and the analysis of the ROC curve showed an AUC of 0.858. Therefore, the authors concluded that FC was a reliable biomarker of MH in IBD with good diagnostic accuracy. Notably, the included studies used various FC cutoff values to correspond to MH (13.9-251 μg/g) in UC, although the most common values were between 150 and 250 μg/g. Similarly, no universal cutoff value for FC in CD was identified, with the reported range relatively wide, ranging from 54-918 μg/g, with a mean cutoff of 205 μg/g[36].

Another meta-analysis that included 1682 and 221 patients with UC and CD, respectively, investigated the optimal FC range for predicting MH[37]. The optimal performance in UC was at a cutoff range of 60-75 μg/g, with high sensitivity (0.87, 95%CI: 0.80-0.91) and specificity (0.81, 95%CI: 0.76-0.85). Among the 221 patients with CD, an FC value of 180-250 μg/g had moderate sensitivity (0.67, 95%CI: 0.41-0.86) and high specificity (0.76, 95%CI: 0.65-0.85). The authors evaluated seven different ranges of FC cutoff values according to the included studies but only one cutoff in CD as follows: 25-50, 60-75, 96-125, 150-180, 192-201, 170-200, and 250-259 μg/g[37]. Recently, another meta-analysis that included 33 studies showed that both FC and fecal lactoferrin were highly sensitive and specific for distinguishing endoscopic activity from MH in patients with CD[38]. Regarding endoscopic activity, FC had pooled sensitivity, specificity, and AUC of 81% (77%-84%), 74% (70%-80%), and 0.85, respectively. The diagnostic odds ratio (DOR) was 13.93 (10.89-17.81). For MH, FC had a sensitivity, specificity, AUC, and DOR of 88% (84%-90%), 72% (64%-79%), 0.88, and 18.17 (11.08-29.82), respectively. Fecal lactoferrin had a sensitivity, specificity, AUC, and DOR of 75% (65%-83%), 80% (57%-92%), 0.81, and 13.41 (5.74-31.32), respectively, for endoscopic activity. Furthermore, the authors noted that 15 of the 25 studies (60%) reported FC values of > 200 μg/g as the optimal cutoff when discriminating between endoscopically active and inactive disease[38].

In small bowel CD, a study that included 165 patients showed that the optimal cutoff value of FC for predicting MH was 76.99 μg/g (sensitivity, specificity, and AUC of 79.45%, 84.21%, and 0.877, respectively). FC was also strongly correlated with the Simplified Endoscopic Score for CD (SES-CD) in these patients (r = 0.753, P < 0.001)[39]. Histological healing in UC has been associated with better prognosis, reduction in clinical recurrence rates, need for steroid use, hospitalization, and surgery, in addition to reducing the risk of dysplasia and colonic neoplasia[40]. Although histological healing is recognized as an important outcome, it is not yet considered a treatment target in UC or CD[11]. However, it could be implemented as an additional target in the UC scenario, combined with MH, to achieve a deeper level of healing. A systematic review published in 2020 that included 12 studies and 1,168 patients with UC found a correlation between FC and histological activity. Eleven different FC cutoff points were identified to distinguish histological remission, ranging from 40.5-200 μg/g, and cutoff values indicating histologic activity varied from 72-250 μg/g[41].

A recent study aimed to determine the optimal FC cutoff value for predicting histological healing in patients with UC in clinical and endoscopic remission (partial Mayo score of ≤ 2 points and MES of 0-1)[42]. The authors included 76 patients, and the FC value in patients with histological healing was 56.2 μg/g compared with 118.1 μg/g in those with histological activity (P = 0.002). Therefore, the optimal cutoff value for predicting histological healing was 82.7 μg/g (sensitivity, specificity, and AUC of 73%, 64%, and 0.71, respectively), and FC values increased with to the histological grade of severity. FC value was also lower in patients with MES of 0 (53.4 μg/g) than in those with MES of 1 (171.8 μg/g; P < 0.001)[42]. Similar FC values were observed in a study published in 2020, which included 185 patients with UC. An FC value of ≥ 135 μg/g predicted histological activity [Geboes score (GS) of ≥ 3.1] with a sensitivity, specificity, and AUC of 54%, 69%, and 0.627, respectively[43]. Patients in histological remission had a median FC of 79.5 μg/g compared with a median FC of 148.5 μg/g for GS of ≥ 3.1 (P = 0.003)[43]. Another study found that an FC value of < 100 μg/g had a high sensitivity to identify endoscopic (81.5%), histological (91.7%), and deep (82.4%) remissions[44]. Cannatelli et al[45] in 2021 published results from a group of patients with CD, where they found that the value of FC for predicting endoscopic healing measured as an SES-CD of ≤ 2 was 96 μg/g (sensitivity, specificity, accuracy, and AUC of 75.0%, 84.4%, 82.9%, and 75.0, respectively). In this study, the optimal cutoff of FC for predicting histological remission assessed with modified Riley = 0 was 225 μg/g (sensitivity, specificity, accuracy, and AUC of 88.9%, 71.9%, 75.6%, and 80.9, respectively)[45].

A recent study evaluated the accuracy of FC for predicting clinical, endoscopic, and histological remission in patients with UC treated with biological therapy[46]. When comparing FC values of < 250 and > 250 μg/g, the group with FC levels of ≤ 250 μg/g had a higher probability of achieving clinical [odds ratio (OR): 4.03; 95%CI: 2.78-5.85], endoscopic (OR: 4.26; 95%CI: 2.83-6.40), and histologic (OR: 6.42; 95%CI: 4.02-10.26) remission at week 52 compared with the group with FC levels of ≥ 250 μg/g. The study also observed a reduced probability of risk of colectomy and hospitalization[46].

A study evaluated the association between FC and TH using intestinal ultrasound[47]. The study included 118 patients with CD treated with biological therapy for 2 years. Specifically, the clinical remission rate, MH, and TH were 62.7%, 44.1%, and 32.2%, respectively. Mean FC concentration decreased from 307.5 ± 248.2 to 45.4 ± 31.3 μg/g (P < 0.001) and from 384.5 ± 355.8 to 44.9 ± 27.82 μg/g (P < 0.001) in patients with TH and those with MH, respectively. An FC cutoff value of 94 μg/g was associated with MH (sensitivity, specificity, and AUC of 94.2%, 84.8%, and 0.95, respectively) and TH (sensitivity, specificity, and AUC of 92.1%, 70%, and 0.88, respectively)[47].

FC can also identify postoperative recurrence in patients with CD who underwent ileocecal resection[48]. A meta-analysis including 10 articles and 613 postoperative patients reported sensitivity and specificity values of 0.82 and 0.61 for FC, respectively[49]. In a prospective study, an FC level of > 100 μg/g was associated with a sensitivity, specificity, and accuracy of 95%, 54%, and 77%, respectively, for predicting endoscopic recurrence[50]. Considering patients with CD in surgically induced remission in the first 12 mo, the AGA recommends an FC value of < 150 μg/g to avoid endoscopic assessment of disease activity in patients at low risk of postoperative disease recurrence. However, for patients at high risk of postoperative disease recurrence, the AGA recommends endoscopic evaluation for assessing endoscopic recurrence rather than relying only on biomarkers[20].

Studies have also shown that FC values were significantly elevated in pouchitis[51]. A retrospective study that included 26 patients with and 40 patients without pouchitis, found that the optimal FC value for predicting a high risk of pouchitis was 143.25 μg/g (AUC: 0.876)[51]. In contrast, a prospective study that included 170 patients (72 with pouchitis) found that the FC cutoff value associated with the presence of pouchitis was 246 μg/g (sensitivity, specificity, and AUC of 83.9%, 71.0%, and 0.85, respectively). Furthermore, the authors reported a good correlation coefficient between calprotectin and the modified pouchitis disease activity index (r2 = 0.279, P < 0.001)[52]. An FC value of < 125 μg/g (sensitivity and specificity of 35.29 and 83.33, respectively) or < 100 μg/g (sensitivity and specificity of 29.41 and 94.44, respectively) was associated with endoscopic remission in patients with ileal pouch-anal anastomosis[53]. A systematic clinical review published in 2020 included 117 studies that enrolled 256 patients (100 with pouchitis) and reported an FC cutoff value ranging from 56 to 494 μg/g among the studies[54].

Although FC is an easy, non-invasive, cost-effective, and reliable biomarker, it has some limitations that should be mentioned. Some factors can affect FC levels, by increasing the concentration[55,56]. Specifically, some factors should be highlighted, particularly variability with age, lifestyle, obesity[55], and the influence of some medications, such as non-steroidal anti-inflammatory drugs, acetylsalicylic acid, and proton pump inhibitors[56]. As FC levels are a measure of mucosal inflammation, elevated FC concentrations can also be found in other disorders with an inflammatory component, such as infectious enteritis, ischemic colitis, diverticulitis, or colorectal cancer[14]. Therefore, when evaluating FC levels, the patient's clinical characteristics should be considered to interpret the test accurately.

Notably, for the optimal use of this inflammatory biomarker in clinical practice, care should be taken in the pre-analytical and analytical phases of FC measurement. The stool sample should be collected at home and in a minimum quantity of 100 mg. If possible, the sample should be collected from the first bowel movement of the day, and formed or semi-formed stools are preferable. Preventing contamination of the fecal sample with urine or toilet water is important. Analysis of only one stool sample is necessary and limiting storage at 4 °C and 2-3 d is preferable. When possible, taking non-steroidal anti-inflammatory drugs and proton-pump inhibitors, which could induce an increase in FC levels, should be discontinued at least 2 wk before performing the assay. In the laboratory, the samples should be processed within 3 d and no later than 1 wk. An enzyme-linked immunosorbent assay should be used to conduct quantitative FC measurements, as well as choosing the same laboratory and technique to perform serial FC measurements, but point-of-care tests and home-based measurements are valid alternatives[10,57].

In addition to FC, other fecal markers have been studied in IBD. These, include alpha-1-antitrypsin, beta-glucuronidase, chitinase 3-like-1, fecal eosinophil proteins, fecal hemoglobin, high-mobility group box 1 (HMGB1), human beta-defensin-2, human neutrophil peptides, lysozyme, M2-pyruvate kinase, matrix metalloproteinase 9, myeloperoxidase, neopterin, neutrophil gelatinase-associated lipocalin, polymorphonuclear neutrophil elastase, and S100A12[58]. S100A12 is a protein released during granulocyte activation and is associated with clinical activity, while HMGB1 has a good correlation with clinical and endoscopic activity[58]. However, future studies are needed to validate these fecal biomarkers and for their use in the clinical management of patients with IBD.

In conclusion, FC is a reliable biomarker that could be used in clinical practice for patients with IBD, from diagnosis to monitoring therapy and predicting relapse. Despite its well-established role in the literature, some limitations exist on its use, particularly within the range of 100-250 μg/g, where a combination or confirmation with other methods to assess disease activity is needed. Therefore, new fecal biomarkers require further study to validate their use as substitute or complementary tests for FC.

ENDOSCOPIC SCORES AND MH IN PATIENTS WITH IBD

Endoscopy has a fundamental role in the journey of patients with IBD. It is indicated for the diagnosis, management, prognosis, surveillance of dysplasia, and even treatment of some IBD complications, such as fistulae and strictures[59]. Table 1 outlines the recommendations for the use of FC and endoscopy in patients with IBD. Table 2 lists the advantages and limitations of these procedures in this clinical setting. The availability of biologics and small molecules in the therapeutic arsenal of IBD brought a new prospective with more ambitious goals that could genuinely alter the natural course of the disease[60]. Endoscopic healing, recently proposed by the STRIDE-II consensus as the therapeutic target for IBD, is associated with improved long-term outcomes[11]. The presence of mucosal inflammation, even in the absence of symptoms, can lead to bowel damage, complications, and higher rates of hospitalization and surgery[61]. Therefore, the greatest challenge lies in determining the criteria for defining endoscopic and/or MH and to what extent we should prioritize it, given the limited pipeline of drugs.

Table 1 Main indications for the use of fecal calprotectin and endoscopy in patients with inflammatory bowel disease.
Fecal calprotectin
Endoscopy
Differential diagnosis of IBD and irritable bowel syndromeDifferential diagnosis with mimics
Monitoring therapeutic responseEvaluation of disease extension
Monitoring mucosal healingMonitoring therapeutic response
Monitoring histological healingMonitoring mucosal healing
Prediction of disease activity and postoperative recurrenceMonitoring histological healing
Prediction of disease activity and postoperative recurrence
Dysplasia surveillance
Treatment of some complications (e.g., strictures)
Table 2 Advantages and limitations of fecal calprotectin and endoscopy in patients with inflammatory bowel disease.

Fecal calprotectin
Endoscopy
AdvantagesNoninvasiveDirect evaluation of the mucosa
Low-cost, cost-effectiveGold standard method to evaluate the goal of IBD treatment (mucosal healing)
Easy collection and storagePossibility of obtaining samples (biopsies)
Validated in UC and CDValidated in IBD diagnostic, monitoring, and prediction of disease activity
Validated in both colonic and small bowel diseaseValidated in adults and pediatric population
Validated in IBD diagnostic, monitoring, and prediction of disease activityValidated scores for both UC and CD
Validated in adults and pediatric population
Distinguish patients with IBD from those with IBS
LimitationsNot specific for IBDInvasive
Not differentiated UC from CDCost
No validated cutoff to define disease activityAvailability
Presence of a “gray zone” level between 100 and 250 μg/g, which is difficult to interpretInter-observer variability
Lower accuracy in detecting inflammatory activity in patients with CD of the small intestine or of the upper gastrointestinal tract compared to predominant or extensive colonic involvement
Variation depending on patient age, presence of obesity, and lifestyle
The presence of mucus and blood can interfere with FC result
High day-to-day variability
Despite the low cost, it is not available in some locations

Endoscopic activity assessment is crucial in IBD management, and many scores have been proposed to evaluate mucosal improvement. The most used scores in clinical practice for CD are the CD Endoscopic Index of Severity[62] and the SES-CD[63]. Each has advantages and disadvantages, and the simplified score is more suitable for routine use. Several endoscopic scoring systems are currently in use for UC. Two of the most routinely used scoring systems are the MES[64] and UCEIS[65]. In a study by Ishida et al[6], only patients with MES 0 or 1 were enrolled because is no clear definition of endoscopic remission by UCEIS[6]. MES is a simple score used in large-scale clinical trials and daily practice, but it has some limitations in the description of both active and inactive UC. Despite some similarities with MES, the UCEIS considers the depth of ulcers. The more detailed description of the ulcers by UCEIS makes it slightly more laborious. This might explain the better performance of UCEIS compared with MES in most published studies, with higher reliability and a potential prognostic role[66]. UCEIS is not as complicated a score to measure and can reveal the most severe colitis area better and more precisely than the MES[67]. However, both scores described above solely focus on the most active area of inflammation and do not consider the entire colon. Therefore, the UCCIS score was developed to fill this gap and include the evaluation of disease progression. It provides more detailed information about the inflammatory condition of the entire colonic mucosa[68]. This is relevant as patients with UC undergoing treatment can have erratic mucosal healing with “patchy” inflammation or histologic rectal sparing[69]. The UCCIS score evaluates the vascular pattern, granularity, friability, and erosions/ulcers in the five segments of the colon (ascendant, transverse, descendent, colon, and rectum). Subsequently, they are applied to a formula that, unfortunately, is difficult to use. Therefore, calculating the UCCIS score requires time, effort, and a complete colonoscopy, making its daily use almost impractical[70]. A recent study by Ishida et al[6] determined the cutoff values of the UCCIS score for predicting a 5-year clinical relapse in patients with UC. Relapse rates were significantly lower in patients with a UCCIS score of ≥ 9.8 than in those with a UCCIS score of < 9.8 (log-rank test, P < 0.001). Therefore, prospective multicenter studies are needed to compare the UCCIS score with biomarkers or histological indices and evaluate its potential prognostic role[71].

A significant challenge concerning the assessment of endoscopic disease activity is the relative subjectivity of the evaluated parameters, which may lead to lower rates of interobserver agreement, particularly among unexperienced endoscopists[9]. To minimize these variabilities, a central endoscopy reading of the recorded examinations by trained readers has been proposed and adopted, at least in most clinical trials[72]. Recently, artificial intelligence has been added to the endoscopic arsenal to improve the quality and definition of images, aiding a better assessment of disease activity, mainly in UC. The use of a computer-aided diagnostic system can overcome the subjectivity of the scores and overcome the bias of interobserver variability[73]. A computerized system, developed by Maeda et al[74], which analyzed approximately 13000 images from 87 patients, determined active histological inflammation with a sensitivity, specificity, and accuracy of 74%, 97%, and 91%, respectively[74]. Other computer-assisted diagnostic systems have also been developed. Ozawa et al[75] used a convolutional neural network that had an AUROC of 0.86 and 0.98 to identify MES of 0 and 1, respectively[75]. Takenaka et al[76] used another deep neural network system and reported accuracies of 90.1% and 92.9% in rating endoscopic remission and histologic healing, respectively[76].

MH, defined as MES of ≤ 1, has been the treatment target in UC for many years. However, in observational studies and post hoc analysis of the infliximab trials, when patients with MES of 0 and 1 were subdivided, the group with MES of 0 at week 8 showed a higher steroid-free remission rate at week 54 than those with MES of 1 (63% and 46%, respectively)[77]. Carvalho et al[78] evaluated 138 patients with UC in steroid-free remission, and patients with MES of 1 had a significant three-fold increased risk of relapse compared with those with MES of 0[78]. In a meta-analysis published in 2020, including 2608 patients with UC in clinical remission from 17 studies, MES of 0 was associated with a 52% lower risk of relapse than MES of 1 (relative risk: 0.48, 95%CI: 0.37-0.62)[79]. Currently, there is a trend toward considering endoscopic remission and improvement in UC as MES of 0 and ≤ 1, respectively, representing MH[64]. Based on these findings, the 2021 update of the STRIDE-II guidelines began considering endoscopic healing in CD as SES-CD score of < 3 points or the absence of ulcerations, while in UC, a MES of 0 or UCEIS score of ≤ 1[11].

In the previous year, a study by George et al[80], including 445 patients with UC, aimed to evaluate the risk of relapse among patients with MES of 0 or 1 and determine if the Robarts Histopathologic Index (RHI) was predictive of clinical relapse. Most patients (95%) with MES of 0 were in histologic remission based on the RHI compared with only 35% with MES of 1. Within 1 year of colonoscopy, 26% of patients experienced a clinical relapse. Patients with MES of 1 or RHI of > 3 were significantly more likely to relapse (P < 0.01) than those with MES of 0 or RHI of ≤ 3. When stratified into four groups by MES and histologic activity, defined by an RHI of > 3 (MES of 0, RHI of ≤ 3; MES of 0, RHI of > 3; MES of 1, RHI of ≤ 3; and MES of 1, RHI of > 3), an RHI of > 3 was the better predictor of clinical relapse (P = 0.008)[80].

The histological index, RHI, used in the previous study assesses four characteristics of mucosal activity, including inflammatory infiltration, neutrophils in the lamina propria or the epithelium, and erosion or ulcers. Each of these items is rated on a scale from 0 to 3, depending on their intensity. Additionally, each parameter has a different weight and is multiplied by 1, 2, 3, and 5 to in a total possible score ranging from 0 (inactive) to 33 (severely active disease)[81]. RHI is not the only histopathological index used to evaluate the activity of patients with UC. The Nancy Index (NI) and GS are also studied for this purpose. The NI was the first validated index to assess histological disease activity in UC. The first criteria are the presence of mucosal ulceration (loss of colonic crypts with “immature” granulation tissue with disorganized blood vessels and extravasated neutrophils) or the presence of fibrinopurulent exudate. The second criterion is the acute inflammatory cells infiltrate (presence of neutrophils in the lamina propria or epithelium). The last criterion is evaluation of the presence and intensity of chronic inflammatory infiltrate (presence of lymphocytes and/or plasmocytes and/or eosinophils in the lamina propria)[82]. In a phase III clinical trial of adalimumab, a moderate (r ≥ 0.3) positive correlation between NI and MES (r = 0.39 for the endoscopy subscore) was reported by Peyrin-Biroulet et al[83] at week 8. In the same study, this correlation increased (r = 0.53) when evaluated in week 52[83]. GS is a complex and difficult score that assesses features relevant to histological inflammation in UC to distinguish among quiescent disease (inactive disease or grade 1), mildly active disease (presence of polymorphonuclear cells or neutrophils or grades 2 and 3), and moderate to severely active disease (epithelial cell damage or grades 4 and 5)[84]. In the abovementioned adalimumab, study, comparing the three histological indices found significant differences among the mean scores of all three indices observed across the known groups based on MESs and full Mayo scores at weeks 8 and 52 (P < 0.001)[83].

For CD, TH is an ambitious and powerful treatment goal and appears to be associated with improvement in all clinical outcomes, with better long-term results even after the discontinuation of biologics; however, we still do not have sufficient data to support optimizing therapy when TH is not achieved[85]. The evolution of endoscopic equipment with the advent of virtual chromoendoscopy (VCE) brought an improvement in endoscopic image analyzing mucosal and vascular components. This new technique enabled the development of the Paddington International VCE ScOre (PICaSSO score), a validated score that showed optimal performance in defining deep remission, a strong correlation with histological activity, and a high interobserver agreement and reliability[86]. Most previous studies showed a relatively strong correlation between PICaSSO and UCEIS, with similar short-term prognostic performance. However, data comparing with UCCIS are scarce. A comparison among a modified PICaSSO, MES, and a probe-based confocal laser endomicroscopy in the prediction of histological healing (RHI of ≤ 3) was conducted in a prospective study by Iacucci et al[87] and published in 2020. The modified PICaSSO simplified and structured by thresholds of severity from the original score, had optimal performances (AUROC: 0.96, accuracy: 91.5%) with a cutoff threshold of 4[87].

Preliminary unpublished reports suggest that the score, originally validated using the iSCAN platform, appeared to be reproducible when used with other VCE techniques, such as the narrow-band imaging near focus (Olympus) or blue-light imaging (Fujifilm) platforms[88]. Despite their weaknesses, mostly due to poor use in practice, the UCEIS, UCCIS, and PICaSSO have been described as useful and reliable endoscopic scores with superior strength to the MES. Table 3 summarizes the endoscopic indices for UC activity, highlighting their strengths and limitations[89].

Table 3 Endoscopic indices of ulcerative colitis activity with their strengths and limitations.
Indices
Endoscopic technique
Validation
Strengths
Limitations
MESWLENoThe easiest to useSubjectivity
Used in clinical trials and daily practiceModerate reproducibility
Not appropriate description of inflammation and severity
Ambiguous definition of endoscopic remission
UCEISWLEYesEasy to useNo thresholds for mild, moderate and severe disease
Good reproducibility and agreementNo definition of superficial or deep ulcer
High correlation with clinical, and histological indices and biomarkers
Clear definition or ER/MH
Clinically relevant outcomes
UCCISWLEYesGood reproducibility and agreementNo definition of MH
Provides details about the status of inflammation of the entire colonic mucosaNo thresholds for mild, moderate and severe disease
Few evidence
PICaSSOVCE-iSCANYesHigh reproducibility and agreementEndoscopy experience and training required.
Strong accuracy discriminating quiescent from mild disease.No long-term clinical outcome
Highest correlation with MH

Although endoscopy is an invasive tool for monitoring UC, it remains cost-effective. However, it is the gold standard method for evaluating MH, which is the desired target of IBD treatment. In this context, the use of endoscopy scores is essential to standardize reports, allowing a comparison of examinations before and after treatment. Endoscopic findings in patients with UC in clinical remission can also predict the risk of relapse, which can be improved by rigorous remission definitions of endoscopic (MES of 0/UCEIS score of ≤ 1) and histologic remission. In the analysis of Ishida et al[6], all three endoscopic indices and FC were useful in predicting disease relapse[6]. Despite the better results with UCCIS, its complexity makes routine use difficult and impractical. Therefore, UCEIS appears to be the scoring system that combines simplicity with better accuracy in the prediction of clinical relapse for clinical practice. The PICaSSO score shows promise; however, the equipment required and the need for endoscopist training are some gaps in most centers.

CONCLUSION

Achieving MH is a pivotal therapeutic goal in the management of IBD. Employing proactive disease and therapy assessment is crucial to achieve better control of intestinal inflammation, even if subclinical, to alter the natural history of the disease. Furthermore, periodic monitoring of FC levels and interval endoscopic evaluations are cornerstones for evaluating response and remission to advanced therapies targeting IBD and detecting subclinical recurrence. Therefore, this editorial aimed to review the role of FC and endoscopic scores in predicting MH in patients with IBD. In addition, Tables 4 and 5 present practical tips to assist the clinician caring for patients with IBD in interpreting and optimizing the use of FC in patients with CD and UC. Despite the UCEIS being more reliable than the MES for assessing endoscopic healing, the widespread use and simplicity of MES justify its routine use in predicting UC relapse. Even with excellent prediction rates of relapse, the value of histological healing and TH as a therapeutic target in IBD continue to evolve and need further study. Data to support treatment intensification based only on histologic and transmural activity remains insufficient, and these patients at least require a closer follow-up.

Table 4 Practical pearls for the use of fecal calprotectin in Crohn’s disease.
FC is a reliable test in distinguishing patients with IBD from those with IBS. A cutoff of ≤50 μg/g appears to have a better sensitivity and a negative predictive value of > 95% for IBD in Western countries[18]
FC should be measured before starting or optimizing any therapy for CD, at the end of the induction phase, every 2–4 mo in patients being treated for active disease, and every 6–12 mo during the maintenance therapy in those in symptomatic remission; and in case of clinical relapse of disease[14,20]
In patients with CD in symptomatic remission with confirmation of endoscopic remission within the last 3 years, an FC of < 150 µg/g can reliably rule out active inflammation and avoid routine endoscopic reassessment with relatively low false negatives[20]
Where there is a symptom-biomarker disconnect, or in patients with mild symptoms, an FC value of > 150 µg/g is insufficient to identify endoscopically active inflammation. In this context, endoscopic or radiologic assessment is necessary to truly define the presence of active disease before making empiric treatment adjustments[20]
In the presence of moderate to severe symptoms, an elevated FC strongly suggests endoscopically active disease and can be used to make decisions regarding most changes in therapy. However, normal FC is insufficient to dismiss inflammation, and endoscopic or radiologic assessment is important in this setting[20]
In small bowel CD, where only a short segment of involvement may exist, as well as in relatively proximal disease (upper gastrointestinal, stomach, and esophagus), the FC concentrations may not be elevated to that degree and produce false negatives[57]
Many times, FC is highly effective in detecting endoscopic ulcerations regardless of the CD location. A cutoff value of > 200 μg/g in patients with isolated ileal involvement and > 250 μg/g for ileocolonic or colonic disease may be the optimal threshold to detect endoscopic ulcerations[90]
Normalization of FC (e.g., < 250 μg/g) within 12 mo of starting therapy is associated with a reduced risk of CD progression[91]
In patients with CD in surgically induced remission within the past 12 mo at a low risk of postoperative recurrence, an FC value of < 50 μg/g reliably rules out postoperative recurrence. In patients at high risk (e.g., smokers, more than one intestinal resection, surgery due to penetrating disease, perianal disease, and long segments of small bowel resection), FC cannot be used to rule out or confirm endoscopic recurrence[20]
After ileocecal resection, an FC cutoff value of > 150 μg/g is likely to have the best overall accuracy in predicting postoperative endoscopic recurrence, with a sensitivity of approximately 70%[92]
FC concentration from an ileostomy effluent can be used for assessing and monitoring small bowel inflammation and disease recurrence. An FC level of > 60 μg/g is strongly suggestive of the presence of small bowel inflammation[93]
Table 5 Practical pearls to the use of fecal calprotectin in ulcerative colitis.
FC should be measured before starting or optimizing any therapy for UC, at the end of induction therapy, every 2–4 mo in patients being treated for active disease, and every 6–12 mo during the maintenance therapy in patients in symptomatic remission; and in case of clinical relapse of disease[94]
FC values of < 150 μg/g typically reflect remission, FC values ranging from 150–250 μg/g are a grey zone, and cutoff values of > 200–300 μg/g suggest the presence of active disease[11]
Prior to symptoms based on the diagnosis of a flare, FC is reported to be elevated approximately 8 wk in advance. Conversely, patients who maintain remission usually present FC concentrations persistently < 60 μg/g[95]
FC is a reliable biomarker to evaluate the response to treatment. A post-induction FC concentration of ≤ 250 µg/g vs > 250 µg/g is associated with a substantially higher probability of achieving clinical, endoscopic, and histologic remission[46]
FC is a valuable marker of endoscopic inflammation, being useful in distinguishing Mayo endoscopic subscores of 0 from 1–3 using the FC cutoff of 60 μg/g[96]
Persistent high values of FC are an important predictor of disease flare in asymptomatic patients[94]
In patients with mild-to-moderate UC who achieve complete endoscopic healing, a FC cutoff value between 75 and 100 µg/g can be used to discriminate patients with ongoing microscopic inflammation from those with histologic remission[97]
In patients with ileal pouch-anal anastomosis, FC values of > 100 µg/g are suggestive of endoscopic or histological inflammation of the pouch (e.g., pouchitis)[57]
Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Brazil

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade C

Creativity or Innovation: Grade C

Scientific Significance: Grade B

P-Reviewer: Kim JE, South Korea S-Editor: Li L L-Editor: Filipodia P-Editor: Zheng XM

References
1.  Wang M, Shi J, Yu C, Zhang X, Xu G, Xu Z, Ma Y. Emerging strategy towards mucosal healing in inflammatory bowel disease: what the future holds? Front Immunol. 2023;14:1298186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
2.  Imbrizi M, Baima JP, Azevedo MFC, Andrade AR, Queiroz NSF, Chebli JMF, Chebli LA, Argollo MC, Sassaki LY, Parra RS, Quaresma AB, Vieira A, Damião AOMC, Moraes ACDS, Flores C, Zaltman C, Vilela EG, Morsoletto EM, Gonçalves Filho FA, Penna FGCE, Santana GO, Zabot GP, Parente JML, Costa MHM, Zerôncio MA, Machado MB, Cassol OS, Kotze PG, Fróes RSB, Miszputen SJ, Ambrogini Junior O, Saad-Hossne R, Coy CSR. Second Brazilian consensus on the management of Crohn's disease in adults: a consensus of the Brazilian organization for Crohn’s disease and colitis (GEDIIB). Arq Gastroenterol. 2023;59:20-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
3.  Chebli JMF, Queiroz NSF, Damião AOMC, Chebli LA, Costa MHM, Parra RS. How to manage inflammatory bowel disease during the COVID-19 pandemic: A guide for the practicing clinician. World J Gastroenterol. 2021;27:1022-1042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
4.  Garcia KS, de Azevedo MFC, Carlos AS, Barros LL, Oba J, Sobrado Junior CW, Sipahi AM, Alves ODC, Navarro-Rodriguez T, Parra RS, Chebli JMF, Chebli LA, Flores C, Vieira A, do Ceará CDA, Queiroz NSF, Damião AOMC. Efficacy of Early Optimization of Infliximab Guided by Therapeutic Drug Monitoring during Induction-A Prospective Trial. Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Baima JP, Imbrizi M, Andrade AR, Chebli LA, Argollo MC, Queiroz NSF, Azevedo MFC, Vieira A, Costa MHM, Fróes RSB, Penna FGCE, Quaresma AB, Damião AOMC, Moraes ACDS, Santos CHMD, Flores C, Zaltman C, Vilela EG, Morsoletto E, Gonçalves Filho FA, Santana GO, Zabot GP, Parente JML, Sassaki LY, Zerôncio MA, Machado MB, Cassol OS, Kotze PG, Parra RS, Miszputen SJ, Coy CSR, Ambrogini Junior O, Chebli JMF, Saad-Hossne R. Second Brazilian consensus on the management of ulcerative colitis in adults: a consensus of the brazilian organization for crohn's disease and colitis (GEDIIB). Arq Gastroenterol. 2023;59:51-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  Ishida N, Ito T, Takahashi K, Asai Y, Miyazu T, Higuchi T, Tamura S, Tani S, Yamade M, Iwaizumi M, Hamaya Y, Osawa S, Sugimoto K. Comparison of fecal calprotectin levels and endoscopic scores for predicting relapse in patients with ulcerative colitis in remission. World J Gastroenterol. 2023;29:6111-6121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (2)]
7.  Rachmilewitz D. Coated mesalazine (5-aminosalicylic acid) versus sulphasalazine in the treatment of active ulcerative colitis: a randomised trial. BMJ. 1989;298:82-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 771]  [Cited by in F6Publishing: 787]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
8.  Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, Calabrese E, Baumgart DC, Bettenworth D, Borralho Nunes P, Burisch J, Castiglione F, Eliakim R, Ellul P, González-Lama Y, Gordon H, Halligan S, Katsanos K, Kopylov U, Kotze PG, Krustinš E, Laghi A, Limdi JK, Rieder F, Rimola J, Taylor SA, Tolan D, van Rheenen P, Verstockt B, Stoker J; European Crohn’s and Colitis Organisation [ECCO] and the European Society of Gastrointestinal and Abdominal Radiology [ESGAR]. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis. 2019;13:144-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 633]  [Cited by in F6Publishing: 857]  [Article Influence: 171.4]  [Reference Citation Analysis (0)]
9.  Hindryckx P, Baert F, Hart A, Magro F, Armuzzi A, Peyrin-Biroulet L; Clinical Trial Committee Clincom of the European Crohn’s and Colitis Organisation (ECCO). Clinical trials in ulcerative colitis: a historical perspective. J Crohns Colitis. 2015;9:580-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
10.  D'Amico F, Rubin DT, Kotze PG, Magro F, Siegmund B, Kobayashi T, Olivera PA, Bossuyt P, Pouillon L, Louis E, Domènech E, Ghosh S, Danese S, Peyrin-Biroulet L. International consensus on methodological issues in standardization of fecal calprotectin measurement in inflammatory bowel diseases. United European Gastroenterol J. 2021;9:451-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
11.  Turner D, Ricciuto A, Lewis A, D'Amico F, Dhaliwal J, Griffiths AM, Bettenworth D, Sandborn WJ, Sands BE, Reinisch W, Schölmerich J, Bemelman W, Danese S, Mary JY, Rubin D, Colombel JF, Peyrin-Biroulet L, Dotan I, Abreu MT, Dignass A; International Organization for the Study of IBD. STRIDE-II: An Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology. 2021;160:1570-1583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 1010]  [Article Influence: 336.7]  [Reference Citation Analysis (0)]
12.  Nakashige TG, Zhang B, Krebs C, Nolan EM. Human calprotectin is an iron-sequestering host-defense protein. Nat Chem Biol. 2015;11:765-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 192]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
13.  Ayling RM, Kok K. Fecal Calprotectin. Adv Clin Chem. 2018;87:161-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 90]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
14.  Kapel N, Ouni H, Benahmed NA, Barbot-Trystram L. Fecal Calprotectin for the Diagnosis and Management of Inflammatory Bowel Diseases. Clin Transl Gastroenterol. 2023;14:e00617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
15.  Gisbert JP, Bermejo F, Pérez-Calle JL, Taxonera C, Vera I, McNicholl AG, Algaba A, López P, López-Palacios N, Calvo M, González-Lama Y, Carneros JA, Velasco M, Maté J. Fecal calprotectin and lactoferrin for the prediction of inflammatory bowel disease relapse. Inflamm Bowel Dis. 2009;15:1190-1198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 182]  [Article Influence: 12.1]  [Reference Citation Analysis (3)]
16.  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;14:32-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 187]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
17.  Vinding KK, Elsberg H, Thorkilgaard T, Belard E, Pedersen N, Elkjaer M, Marker D, Carlsen K, Burisch J, Munkholm P. Fecal Calprotectin Measured By Patients at Home Using Smartphones--A New Clinical Tool in Monitoring Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22:336-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 67]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
18.  Dajti E, Frazzoni L, Iascone V, Secco M, Vestito A, Fuccio L, Eusebi LH, Fusaroli P, Rizzello F, Calabrese C, Gionchetti P, Bazzoli F, Zagari RM. Systematic review with meta-analysis: Diagnostic performance of faecal calprotectin in distinguishing inflammatory bowel disease from irritable bowel syndrome in adults. Aliment Pharmacol Ther. 2023;58:1120-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Menees SB, Powell C, Kurlander J, Goel A, Chey WD. A meta-analysis of the utility of C-reactive protein, erythrocyte sedimentation rate, fecal calprotectin, and fecal lactoferrin to exclude inflammatory bowel disease in adults with IBS. Am J Gastroenterol. 2015;110:444-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 213]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
20.  Ananthakrishnan AN, Adler J, Chachu KA, Nguyen NH, Siddique SM, Weiss JM, Sultan S, Velayos FS, Cohen BL, Singh S; AGA Clinical Guidelines Committee. AGA Clinical Practice Guideline on the Role of Biomarkers for the Management of Crohn's Disease. Gastroenterology. 2023;165:1367-1399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Reference Citation Analysis (0)]
21.  Singh S, Ananthakrishnan AN, Nguyen NH, Cohen BL, Velayos FS, Weiss JM, Sultan S, Siddique SM, Adler J, Chachu KA; AGA Clinical Guidelines Committee. Electronic address: clinicalpractice@gastro.org. AGA Clinical Practice Guideline on the Role of Biomarkers for the Management of Ulcerative Colitis. Gastroenterology. 2023;164:344-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
22.  Lin JF, Chen JM, Zuo JH, Yu A, Xiao ZJ, Deng FH, Nie B, Jiang B. Meta-analysis: fecal calprotectin for assessment of inflammatory bowel disease activity. Inflamm Bowel Dis. 2014;20:1407-1415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 220]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
23.  Heida A, Park KT, van Rheenen PF. Clinical Utility of Fecal Calprotectin Monitoring in Asymptomatic Patients with Inflammatory Bowel Disease: A Systematic Review and Practical Guide. Inflamm Bowel Dis. 2017;23:894-902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 96]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
24.  Costa F, Mumolo MG, Ceccarelli L, Bellini M, Romano MR, Sterpi C, Ricchiuti A, Marchi S, Bottai M. Calprotectin is a stronger predictive marker of relapse in ulcerative colitis than in Crohn's disease. Gut. 2005;54:364-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in F6Publishing: 400]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
25.  Yamamoto T, Shimoyama T, Umegae S, Matsumoto K. Endoscopic score vs. fecal biomarkers for predicting relapse in patients with ulcerative colitis after clinical remission and mucosal healing. Clin Transl Gastroenterol. 2018;9:136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
26.  Schoepfer AM, Beglinger C, Straumann A, Safroneeva E, Romero Y, Armstrong D, Schmidt C, Trummler M, Pittet V, Vavricka SR. Fecal calprotectin more accurately reflects endoscopic activity of ulcerative colitis than the Lichtiger Index, C-reactive protein, platelets, hemoglobin, and blood leukocytes. Inflamm Bowel Dis. 2013;19:332-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 207]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
27.  Sipponen T, Kärkkäinen P, Savilahti E, Kolho KL, Nuutinen H, Turunen U, Färkkilä M. Correlation of faecal calprotectin and lactoferrin with an endoscopic score for Crohn's disease and histological findings. Aliment Pharmacol Ther. 2008;28:1221-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 233]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
28.  Arai T, Takeuchi K, Miyamura M, Ishikawa R, Yamada A, Katsumata M, Igarashi Y, Suzuki Y. Level of Fecal Calprotectin Correlates With Severity of Small Bowel Crohn's Disease, Measured by Balloon-assisted Enteroscopy and Computed Tomography Enterography. Clin Gastroenterol Hepatol. 2017;15:56-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
29.  Egea Valenzuela J, Pereñíguez López A, Pérez Fernández V, Alberca de Las Parras F, Carballo Álvarez F. Fecal calprotectin and C-reactive protein are associated with positive findings in capsule endoscopy in suspected small bowel Crohn's disease. Rev Esp Enferm Dig. 2016;108:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
30.  Moniuszko A, Głuszek S, Rydzewska G. Rapid fecal calprotectin test for prediction of mucosal inflammation in ulcerative colitis and Crohn disease: a prospective cohort study. Pol Arch Intern Med. 2017;127:312-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
31.  Schoepfer AM, Beglinger C, Straumann A, Trummler M, Vavricka SR, Bruegger LE, Seibold F. 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;105:162-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 398]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
32.  Guidi L, Marzo M, Andrisani G, Felice C, Pugliese D, Mocci G, Nardone O, De Vitis I, Papa A, Rapaccini G, Forni F, Armuzzi A. Faecal calprotectin assay after induction with anti-Tumour Necrosis Factor α agents in inflammatory bowel disease: Prediction of clinical response and mucosal healing at one year. Dig Liver Dis. 2014;46:974-979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 63]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
33.  Bertani L, Blandizzi C, Mumolo MG, Ceccarelli L, Albano E, Tapete G, Baiano Svizzero G, Zanzi F, Coppini F, de Bortoli N, Bellini M, Morganti R, Marchi S, Costa F. Fecal Calprotectin Predicts Mucosal Healing in Patients With Ulcerative Colitis Treated With Biological Therapies: A Prospective Study. Clin Transl Gastroenterol. 2020;11:e00174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 33]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
34.  Narula N, Wong ECL, Dulai PS, Marshall JK, Colombel JF, Reinisch W. Week 6 Calprotectin Best Predicts Likelihood of Long-term Endoscopic Healing in Crohn's Disease: A Post-hoc Analysis of the UNITI/IM-UNITI Trials. J Crohns Colitis. 2021;15:462-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
35.  State M, Negreanu L, Voiosu T, Voiosu A, Balanescu P, Mateescu RB. Surrogate markers of mucosal healing in inflammatory bowel disease: A systematic review. World J Gastroenterol. 2021;27:1828-1840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (1)]
36.  Bromke MA, Neubauer K, Kempiński R, Krzystek-Korpacka M. Faecal Calprotectin in Assessment of Mucosal Healing in Adults with Inflammatory Bowel Disease: A Meta-Analysis. J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
37.  Xiang BJ, Jiang M, Sun MJ, Dai C. Optimal Range of Fecal Calprotectin for Predicting Mucosal Healing in Patients with Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Visc Med. 2021;37:338-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
38.  Bohra A, Mohamed G, Vasudevan A, Lewis D, Van Langenberg DR, Segal JP. The Utility of Faecal Calprotectin, Lactoferrin and Other Faecal Biomarkers in Discriminating Endoscopic Activity in Crohn's Disease: A Systematic Review and Meta-Analysis. Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  Han W, Wu J, Zhang P, Hu N, Mei Q, Hu J. Fecal calprotectin predicts endoscopic activity and mucosal healing of small bowel Crohn's disease evaluated by double-balloon endoscopy. Int J Colorectal Dis. 2022;37:1953-1961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
40.  Neurath MF, Vieth M. Different levels of healing in inflammatory bowel diseases: mucosal, histological, transmural, barrier and complete healing. Gut. 2023;72:2164-2183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
41.  D'Amico F, Bonovas S, Danese S, Peyrin-Biroulet L. Review article: faecal calprotectin and histologic remission in ulcerative colitis. Aliment Pharmacol Ther. 2020;51:689-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
42.  Kawashima K, Oshima N, Kishimoto K, Kataoka M, Fukunaga M, Kotani S, Sonoyama H, Oka A, Mishima Y, Kazumori H, Ishikawa N, Araki A, Ishihara S. Low Fecal Calprotectin Predicts Histological Healing in Patients with Ulcerative Colitis with Endoscopic Remission and Leads to Prolonged Clinical Remission. Inflamm Bowel Dis. 2023;29:359-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
43.  Hart L, Chavannes M, Kherad O, Maedler C, Mourad N, Marcus V, Afif W, Bitton A, Lakatos PL, Brassard P, Bessissow T. Faecal Calprotectin Predicts Endoscopic and Histological Activity in Clinically Quiescent Ulcerative Colitis. J Crohns Colitis. 2020;14:46-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
44.  Malvão LDR, Madi K, Esberard BC, de Amorim RF, Silva KDS, Farias E Silva K, de Souza HSP, Carvalho ATP. Fecal calprotectin as a noninvasive test to predict deep remission in patients with ulcerative colitis. Medicine (Baltimore). 2021;100:e24058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
45.  Cannatelli R, Bazarova A, Zardo D, Nardone OM, Shivaji U, Smith SCL, Gkoutos G, Ricci C, Gui XS, Ghosh S, Iacucci M. Fecal Calprotectin Thresholds to Predict Endoscopic Remission Using Advanced Optical Enhancement Techniques and Histological Remission in IBD Patients. Inflamm Bowel Dis. 2021;27:647-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
46.  Dulai PS, Feagan BG, Sands BE, Chen J, Lasch K, Lirio RA. Prognostic Value of Fecal Calprotectin to Inform Treat-to-Target Monitoring in Ulcerative Colitis. Clin Gastroenterol Hepatol. 2023;21:456-466.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
47.  Castiglione F, Imperatore N, Testa A, de Sire R, Nardone OM, Ricciolino S, Di Luna I, Patturelli M, Villani GD, Olmo O, Rispo A. Exploring the concept of deep remission in Crohn's disease: correlation between transmural healing and biomarkers. Therap Adv Gastroenterol. 2022;15:17562848221110643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
48.  Yamamoto T, Shiraki M, Bamba T, Umegae S, Matsumoto K. Faecal calprotectin and lactoferrin as markers for monitoring disease activity and predicting clinical recurrence in patients with Crohn's disease after ileocolonic resection: A prospective pilot study. United European Gastroenterol J. 2013;1:368-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
49.  Qiu Y, Mao R, Chen BL, He Y, Zeng ZR, Xue L, Song XM, Li ZP, Chen MH. Fecal calprotectin for evaluating postoperative recurrence of Crohn's disease: a meta-analysis of prospective studies. Inflamm Bowel Dis. 2015;21:315-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
50.  Boschetti G, Laidet M, Moussata D, Stefanescu C, Roblin X, Phelip G, Cotte E, Passot G, Francois Y, Drai J, Del Tedesco E, Bouhnik Y, Flourie B, Nancey S. Levels of Fecal Calprotectin Are Associated With the Severity of Postoperative Endoscopic Recurrence in Asymptomatic Patients With Crohn's Disease. Am J Gastroenterol. 2015;110:865-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 99]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
51.  Li RB, Li CQ, Zhang SY, Li KY, Zhao ZC, Liu G. Fecal calprotectin as an indicator in risk stratification of pouchitis following ileal pouch-anal anastomosis for ulcerative colitis. Ann Med. 2023;55:305-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Uchino M, Horio Y, Kuwahara R, Kusunoki K, Nagano K, Ikeuchi H. Associations between Pouchitis and Fecal Calprotectin after Restorative Proctocolectomy in Patients with Ulcerative Colitis. Inflamm Intest Dis. 2023;8:77-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
53.  Ollech JE, Bannon L, Maharshak N, Bar N, Goren I, Tulchinsky H, Yanai H, Dotan I. Fecal Calprotectin Is Increased in Pouchitis and Progressively Increases With More Severe Endoscopic and Histologic Disease. Clin Gastroenterol Hepatol. 2022;20:1839-1846.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
54.  McKechnie T, Lee Y, Kruse C, Ramji K, Springer JE, Wood T, Doumouras AG, Hong D, Eskicioglu C. The role of fecal calprotectin in the diagnosis of acute pouchitis following IPAA for ulcerative colitis: a systematic clinical review. Int J Colorectal Dis. 2020;35:1619-1628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
55.  Caviglia GP, Ribaldone DG, Rosso C, Saracco GM, Astegiano M, Pellicano R. Fecal calprotectin: beyond intestinal organic diseases. Panminerva Med. 2018;60:29-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
56.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 450]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
57.  D'Amico F, Nancey S, Danese S, Peyrin-Biroulet L. A Practical Guide for Faecal Calprotectin Measurement: Myths and Realities. J Crohns Colitis. 2021;15:152-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
58.  Di Ruscio M, Vernia F, Ciccone A, Frieri G, Latella G. Surrogate Fecal Biomarkers in Inflammatory Bowel Disease: Rivals or Complementary Tools of Fecal Calprotectin? Inflamm Bowel Dis. 2017;24:78-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
59.  Bourreille A, Ignjatovic A, Aabakken L, Loftus EV Jr, Eliakim R, Pennazio M, Bouhnik Y, Seidman E, Keuchel M, Albert JG, Ardizzone S, Bar-Meir S, Bisschops R, Despott EJ, Fortun PF, Heuschkel R, Kammermeier J, Leighton JA, Mantzaris GJ, Moussata D, Lo S, Paulsen V, Panés J, Radford-Smith G, Reinisch W, Rondonotti E, Sanders DS, Swoger JM, Yamamoto H, Travis S, Colombel JF, Van Gossum A; World Organisation of Digestive Endoscopy (OMED) and the European Crohn's and Colitis Organisation (ECCO). Role of small-bowel endoscopy in the management of patients with inflammatory bowel disease: an international OMED-ECCO consensus. Endoscopy. 2009;41:618-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 223]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
60.  Neurath MF, Travis SP. Mucosal healing in inflammatory bowel diseases: a systematic review. Gut. 2012;61:1619-1635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 587]  [Article Influence: 48.9]  [Reference Citation Analysis (0)]
61.  Ungaro RC, Yzet C, Bossuyt P, Baert FJ, Vanasek T, D'Haens GR, Joustra VW, Panaccione R, Novacek G, Reinisch W, Armuzzi A, Golovchenko O, Prymak O, Goldis A, Travis SP, Hébuterne X, Ferrante M, Rogler G, Fumery M, Danese S, Rydzewska G, Pariente B, Hertervig E, Stanciu C, Serrero M, Diculescu M, Peyrin-Biroulet L, Laharie D, Wright JP, Gomollón F, Gubonina I, Schreiber S, Motoya S, Hellström PM, Halfvarson J, Butler JW, Petersson J, Petralia F, Colombel JF. Deep Remission at 1 Year Prevents Progression of Early Crohn's Disease. Gastroenterology. 2020;159:139-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 130]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
62.  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 Thérapeutiques des Affections Inflammatoires du Tube Digestif (GETAID). Gut. 1989;30:983-989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 634]  [Cited by in F6Publishing: 619]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
63.  Daperno M, D'Haens G, Van Assche G, Baert F, Bulois P, Maunoury V, Sostegni R, Rocca R, Pera A, Gevers A, Mary JY, Colombel JF, Rutgeerts P. Development and validation of a new, simplified endoscopic activity score for Crohn's disease: the SES-CD. Gastrointest Endosc. 2004;60:505-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 999]  [Cited by in F6Publishing: 1128]  [Article Influence: 56.4]  [Reference Citation Analysis (0)]
64.  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;317:1625-1629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1958]  [Cited by in F6Publishing: 2045]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
65.  Travis SP, Schnell D, Krzeski P, Abreu MT, Altman DG, Colombel JF, Feagan BG, Hanauer SB, Lémann M, Lichtenstein GR, Marteau PR, Reinisch W, Sands BE, Yacyshyn BR, Bernhardt CA, Mary JY, Sandborn WJ. Developing an instrument to assess the endoscopic severity of ulcerative colitis: the Ulcerative Colitis Endoscopic Index of Severity (UCEIS). Gut. 2012;61:535-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 385]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
66.  Travis SP, Schnell D, Krzeski P, Abreu MT, Altman DG, Colombel JF, Feagan BG, Hanauer SB, Lichtenstein GR, Marteau PR, Reinisch W, Sands BE, Yacyshyn BR, Schnell P, Bernhardt CA, Mary JY, Sandborn WJ. Reliability and initial validation of the ulcerative colitis endoscopic index of severity. Gastroenterology. 2013;145:987-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 294]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
67.  Vuitton L, Peyrin-Biroulet L, Colombel JF, Pariente B, Pineton de Chambrun G, Walsh AJ, Panes J, Travis SP, Mary JY, Marteau P. Defining endoscopic response and remission in ulcerative colitis clinical trials: an international consensus. Aliment Pharmacol Ther. 2017;45:801-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 87]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
68.  Samuel S, Bruining DH, Loftus EV Jr, Thia KT, Schroeder KW, Tremaine WJ, Faubion WA, Kane SV, Pardi DS, de Groen PC, Harmsen WS, Zinsmeister AR, Sandborn WJ. Validation of the ulcerative colitis colonoscopic index of severity and its correlation with disease activity measures. Clin Gastroenterol Hepatol. 2013;11:49-54.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 97]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
69.  Kim B, Barnett JL, Kleer CG, Appelman HD. Endoscopic and histological patchiness in treated ulcerative colitis. Am J Gastroenterol. 1999;94:3258-3262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 156]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
70.  Neumann H, Neurath MF. Ulcerative colitis: UCCIS--a reproducible tool to assess mucosal healing. Nat Rev Gastroenterol Hepatol. 2012;9:692-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
71.  Ishida N, Onoue S, Miyazu T, Tamura S, Tani S, Yamade M, Iwaizumi M, Hamaya Y, Osawa S, Furuta T, Sugimoto K. Further research on the clinical relevance of the ulcerative colitis colonoscopic index of severity for predicting 5-year relapse. Int J Colorectal Dis. 2021;36:2661-2670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
72.  Panés J, Feagan BG, Hussain F, Levesque BG, Travis SP. Central Endoscopy Reading in Inflammatory Bowel Diseases. J Crohns Colitis. 2016;10 Suppl 2:S542-S547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
73.  Holmer AK, Dulai PS. Using Artificial Intelligence to Identify Patients With Ulcerative Colitis in Endoscopic and Histologic Remission. Gastroenterology. 2020;158:2045-2047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
74.  Maeda Y, Kudo SE, Ogata N, Misawa M, Iacucci M, Homma M, Nemoto T, Takishima K, Mochida K, Miyachi H, Baba T, Mori K, Ohtsuka K, Mori Y. Evaluation in real-time use of artificial intelligence during colonoscopy to predict relapse of ulcerative colitis: a prospective study. Gastrointest Endosc. 2022;95:747-756.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
75.  Ozawa T, Ishihara S, Fujishiro M, Saito H, Kumagai Y, Shichijo S, Aoyama K, Tada T. Novel computer-assisted diagnosis system for endoscopic disease activity in patients with ulcerative colitis. Gastrointest Endosc. 2019;89:416-421.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 107]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
76.  Takenaka K, Ohtsuka K, Fujii T, Negi M, Suzuki K, Shimizu H, Oshima S, Akiyama S, Motobayashi M, Nagahori M, Saito E, Matsuoka K, Watanabe M. Development and Validation of a Deep Neural Network for Accurate Evaluation of Endoscopic Images From Patients With Ulcerative Colitis. Gastroenterology. 2020;158:2150-2157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 128]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
77.  Colombel JF, Rutgeerts P, Reinisch W, Esser D, Wang Y, Lang Y, Marano CW, Strauss R, Oddens BJ, Feagan BG, Hanauer SB, Lichtenstein GR, Present D, Sands BE, Sandborn WJ. Early mucosal healing with infliximab is associated with improved long-term clinical outcomes in ulcerative colitis. Gastroenterology. 2011;141:1194-1201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 643]  [Cited by in F6Publishing: 676]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
78.  Boal Carvalho P, Dias de Castro F, Rosa B, Moreira MJ, Cotter J. Mucosal Healing in Ulcerative Colitis--When Zero is Better. J Crohns Colitis. 2016;10:20-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 62]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
79.  Yoon H, Jangi S, Dulai PS, Boland BS, Prokop LJ, Jairath V, Feagan BG, Sandborn WJ, Singh S. Incremental Benefit of Achieving Endoscopic and Histologic Remission in Patients With Ulcerative Colitis: A Systematic Review and Meta-Analysis. Gastroenterology. 2020;159:1262-1275.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 87]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
80.  George LA, Feldman HT, Alizadeh M, Abutaleb A, Zullow S, Hine A, Stashek K, Sarkar S, Sun K, Hudesman D, Axelrad J, Cross RK. Histologic Inflammation can Predict Future Clinical Relapse in Ulcerative Colitis Patients in Endoscopic Remission. Crohns Colitis 360. 2023;5:otad059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
81.  Mosli MH, Feagan BG, Zou G, Sandborn WJ, D'Haens G, Khanna R, Shackelton LM, Walker CW, Nelson S, Vandervoort MK, Frisbie V, Samaan MA, Jairath V, Driman DK, Geboes K, Valasek MA, Pai RK, Lauwers GY, Riddell R, Stitt LW, Levesque BG. Development and validation of a histological index for UC. Gut. 2017;66:50-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 228]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
82.  Marchal-Bressenot A, Scherl A, Salleron J, Peyrin-Biroulet L. A practical guide to assess the Nancy histological index for UC. Gut. 2016;65:1919-1920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
83.  Peyrin-Biroulet L, Arenson E, Rubin DT, Siegel CA, Lee S, Stephen Laroux F, Zhou W, Finney-Hayward T, Sanchez Gonzalez Y, Shields AL. A Comparative Evaluation of the Measurement Properties of Three Histological Indices of Mucosal Healing in Ulcerative Colitis: Geboes Score, Robarts Histopathology Index and Nancy Index. J Crohns Colitis. 2023;17:1733-1743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
84.  Geboes K, Riddell R, Ost A, Jensfelt B, Persson T, Löfberg R. A reproducible grading scale for histological assessment of inflammation in ulcerative colitis. Gut. 2000;47:404-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 575]  [Cited by in F6Publishing: 638]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
85.  Castiglione F, Imperatore N, Testa A, De Palma GD, Nardone OM, Pellegrini L, Caporaso N, Rispo A. One-year clinical outcomes with biologics in Crohn's disease: transmural healing compared with mucosal or no healing. Aliment Pharmacol Ther. 2019;49:1026-1039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 96]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
86.  Iacucci M, Smith SCL, Bazarova A, Shivaji UN, Bhandari P, Cannatelli R, Daperno M, Ferraz J, Goetz M, Gui X, Hayee B, De Hertogh G, Lazarev M, Li J, Nardone OM, Parra-Blanco A, Pastorelli L, Panaccione R, Occhipinti V, Rath T, Tontini GE, Vieth M, Villanacci V, Zardo D, Bisschops R, Kiesslich R, Ghosh S. An International Multicenter Real-Life Prospective Study of Electronic Chromoendoscopy Score PICaSSO in Ulcerative Colitis. Gastroenterology. 2021;160:1558-1569.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
87.  Iacucci M, Cannatelli R, Gui X, Zardo D, Bazarova A, Gkoutos GV, Lethebe BC, Kaplan GG, Panaccione R, Kiesslich R, Ghosh S. Assessment of Endoscopic Healing by Using Advanced Technologies Reflects Histological Healing in Ulcerative Colitis. J Crohns Colitis. 2020;14:1282-1289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
88.  Cannatelli R, Nardone O, Shivaji U, Smith SCL, Bazarova A, Zardo D, Ghosh S, Iacucci M. P148 Reproducibility of PICaSSO score by using narrow banding images (NBI) to assess mucosal and histological healing in ulcerative colitis (UC) patients. J Crohns Colitis. 2020;14 (Suppl 1):S211-S212.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Ruscio MD, Cedola M, Mangone M, Brighi S. How to assess endoscopic disease activity in ulcerative colitis in 2022. Ann Gastroenterol. 2022;35:462-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
90.  Buisson A, Mak WY, Andersen MJ, Lei D, Pekow J, Cohen RD, Kahn SA, Pereira B, Rubin DT. Fecal Calprotectin Is Highly Effective to Detect Endoscopic Ulcerations in Crohn's Disease Regardless of Disease Location. Inflamm Bowel Dis. 2021;27:1008-1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
91.  Plevris N, Fulforth J, Lyons M, Siakavellas SI, Jenkinson PW, Chuah CS, Lucaciu L, Pattenden RJ, Arnott ID, Jones GR, Lees CW. Normalization of Fecal Calprotectin Within 12 Months of Diagnosis Is Associated With Reduced Risk of Disease Progression in Patients With Crohn's Disease. Clin Gastroenterol Hepatol. 2021;19:1835-1844.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
92.  Tham YS, Yung DE, Fay S, Yamamoto T, Ben-Horin S, Eliakim R, Koulaouzidis A, Kopylov U. Fecal calprotectin for detection of postoperative endoscopic recurrence in Crohn's disease: systematic review and meta-analysis. Therap Adv Gastroenterol. 2018;11:1756284818785571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
93.  Daoud ND, Hashash JG, Picco MF, Farraye FA. Faecal Calprotectin from Ileostomy Output Is Sensitive and Specific for the Prediction of Small Bowel Inflammation in Patients with Crohn's Disease. J Crohns Colitis. 2022;16:601-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
94.  D'Amico F, Magro F, Caron B, Dignass A, Jairath V, Hart A, Kotze PG, Paridaens K, Awadhi SA, Kobayashi T, Siegmund B, Peyrin-Biroulet L, Danese S. iSTART-II: An Update on the i Support Therapy-Access to Rapid Treatment (iSTART) Approach for Patient-Centered Therapy in Mild-to-Moderate Ulcerative Colitis. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
95.  Yamamoto T, Shimoyama T, Matsumoto K. Consecutive monitoring of faecal calprotectin during mesalazine suppository therapy for active rectal inflammation in ulcerative colitis. Aliment Pharmacol Ther. 2015;42:549-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
96.  Suttichaimongkol T, Coelho-Prabhu N, Bruining DH, Tariq R, Snyder MR, Loftus EV Jr. Diagnostic Performance of a Fecal Calprotectin Assay as a Biomarker for Mayo Endoscopic Subscore in Ulcerative Colitis: Result From a Tertiary Referral Center. Inflamm Bowel Dis. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
97.  Stevens TW, Gecse K, Turner JR, de Hertogh G, Rubin DT, D'Haens GR. Diagnostic Accuracy of Fecal Calprotectin Concentration in Evaluating Therapeutic Outcomes of Patients With Ulcerative Colitis. Clin Gastroenterol Hepatol. 2021;19:2333-2342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]