Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
OPEN ACCESS

Fecal, Blood, and Urinary Biomarkers in Inflammatory Bowel Diseases

  • Sarah Bencardino1,
  • Ferdinando D’Amico1,2,* ,
  • Alessandra Zilli1,
  • Tommaso Lorenzo Parigi1,
  • Mariangela Allocca1,
  • Gionata Fiorino1,3,
  • Silvio Danese1 and
  • Federica Furfaro1
Journal of Translational Gastroenterology   2024;2(2):61-75

doi: 10.14218/JTG.2024.00001

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Bencardino S, D’Amico F, Zilli A, Parigi TL, Allocca M, Fiorino G, et al. Fecal, Blood, and Urinary Biomarkers in Inflammatory Bowel Diseases. J Transl Gastroen. 2024;2(2):61-75. doi: 10.14218/JTG.2024.00001.

Abstract

The incidence and prevalence of inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn’s disease, are rapidly increasing. Currently, colonoscopy is the gold standard for diagnosing and monitoring the course of IBD. However, the recently implemented “treat-to-target” strategy, which involves meticulously pursuing multiple therapeutic objectives, has opened avenues for non-invasive diagnostic and monitoring tools. These tools aim to assess disease activity and predict the likelihood of recurrence. Research studies into serum and fecal biomarkers in IBD have been ongoing for several years. Among the most relevant biomarkers, fecal calprotectin and C-reactive protein have shown the best accuracy, with good-to-optimal correlation with endoscopic, histologic, or transmural activity. Numerous studies have explored the potential advantages of using multi-target tools that combine serum and fecal biomarkers with clinical activity indexes to enhance diagnostic and monitoring effectiveness. Encouraging findings have emerged for fecal lactoferrin, autoantibodies, micro-RNA, gene expression, and many other serological and fecal markers. However, limited evidence has hindered their widespread adoption in routine clinical practice. This review aimed to summarize the available data on the utilization of biomarkers in IBD.

Keywords

Biomarkers, Ulcerative colitis, Crohn’s disease, Inflammatory bowel diseases, IBD, Fecal markers, Calprotectin, C-reactive protein

Introduction

The burden of inflammatory bowel diseases (IBD), primarily Crohn’s disease (CD) and ulcerative colitis (UC), is spreading worldwide.1 Recent incidence rates are steadily rising, reaching up to 17.8 cases per 100,000 person-years for CD and even higher for UC (up to 28.4 per 100,000 person-years).2 Over the past 20 years, there has been a shift in the incidence and prevalence of IBD, with the onset occurring in older populations and varying geographically.3 In highly developed countries such as those in North America, age-standardized prevalence rates are significantly higher than in less developed regions.4 Currently, endoscopic assessment through ileo-colonoscopy remains the most viable and guideline-advised tool for assessing disease activity and monitoring treatment response in IBD patients.5 Monitoring new disease flares and predicting disease course and long-term outcomes are crucial in IBD management.6 Symptom-based tools, such as the partial Mayo score or the Harvey Bradshaw index (HBI),7,8 have not shown optimal correlation with disease activity and endoscopic remission, presenting low negative and positive predictive values.9 In the first STRIDE consensus by the International Organization for the Study of IBD, short-term endoscopic response and long-term endoscopic remission were identified as the primary therapeutic targets of IBD work-up.10 Nevertheless, several cross-sectional imaging techniques, such as magnetic resonance enterography and intestinal ultrasound, have become essential for diagnosing and monitoring IBD patients.11 These imaging modalities offer the advantage of equal accuracy with less invasiveness, enabling comprehensive evaluation of disease status and therapeutic response.12 While cross-sectional imaging and endoscopy serve as primary methods for patient monitoring, their inconsistent availability often leads to prolonged waiting lists, requiring patients to visit hospitals for these investigations. Thus, relying on serum, fecal, and urinary biomarkers of disease activity is crucial for implementing close monitoring of IBD, with some biomarkers potentially being analyzed directly from home.13 A biomarker is defined as a measurable compound or substance that can be objectively identified and quantitatively evaluated in a biological sample, such as blood, urine, tissue, or feces.14 Recently, the novel “treat-to-target” approach in managing IBD patients has heightened the significance of these biomarkers, as it fully depends on the periodic monitoring of markers of disease activity, leading to treatment optimization or therapy changes.6 The CALM trial was the first to demonstrate that promptly escalating treatment, guided by both clinical symptoms and biomarkers, yields superior clinical and endoscopic outcomes in individuals with early CD compared to decisions based on clinical symptoms alone.15 The STRIDE-II consensus initiative confirmed that, in IBD patients, clinical response and remission, endoscopic healing, and normalization of C-reactive protein (CRP)/erythrocyte sedimentation rate and fecal calprotectin (FC) represent the most crucial targets.16 Other serological, fecal, urine, and tissue transcriptomic markers have been tested and are still being studied to assess disease activity or predict response to therapies in IBD (Fig. 1). This review aimed to provide an overview of available biomarkers in IBD, focusing on their role in diagnosis, disease activity evaluation, prediction of response to therapy, endoscopic and histological healing, and disease recurrence.

Summary of fecal, urinary, blood, and histologic biomarkers.
Fig. 1  Summary of fecal, urinary, blood, and histologic biomarkers.

FC, Calprotectin; FL, Lactoferrin; MPO, Myeloperoxidase; CRP, C-reactive-protein; ESR, Erythrocyte sedimentation rate; ST2, Suppression of tumorigenicity 2, TNFAIP6, TNF-α-induced protein 6.

Review criteria

PubMed, Embase, and Scopus databases were screened up to November 30, 2023, to identify studies reporting the accuracy, sensitivity, specificity, and overall feasibility of different biomarkers in IBD. The following search terms were used: ‘biomarkers’, ‘fecal calprotectin’, ‘calprotectin’, ‘polymerase chain reaction (PCR)’, ‘C-reactive protein’, ‘protein C’, ‘erythrocyte sedimentation rate’, ‘anti-neutrophil cytoplasmic antibodies’, ‘auto-antibodies’, ‘non-invasive markers’, combined with ‘Crohn’, ‘Crohn’s disease’, ‘terminal ileitis’, ‘IBD’, ‘post-surgical CD’, ‘peri-anal’, ‘peri-anal disease’, ‘peri-anal CD’, ‘UC’, ‘ulcerative colitis’, ‘colitis’, ‘segmental colitis’. Only articles published in English were considered. Four authors (SB, FD, FF, and AZ) independently reviewed titles and abstracts to identify eligible studies. The full texts of the selected articles were examined for inclusion, and relevant references in their lists were hand-searched to identify studies missed by the electronic search. Abstracts and articles were included based on their relevance.

Fecal biomarkers

FC

FC is a cytosolic calcium and zinc-binding protein composed of two monomeric subunits, S100A8 and S100A9, expressed by neutrophils, dendritic cells, monocytes, macrophages, and squamous cells.17 FC is a stable protein, capable of persisting in a fecal sample at room temperature for one week18 due to its calcium-binding capacity, which confers resistance to proteolysis. The concentration of FC in feces is about six times higher than in plasma and urine samples.19 FC is a hallmark of neutrophil activation during various inflammatory processes in the gut, and its presence in feces is recognized as a marker of gastrointestinal organic disorders,20 such as IBD, non-steroidal anti-inflammatory drug enteropathy,21 and even colorectal cancer.22 The quantification of FC in stools is determined by different types of assays, mainly based on the use of mono or polyclonal antibodies that bind specific epitopes of the FC heterodimer via various techniques (such as immunofluorescence or chemiluminescence).23 The concentration of FC in the stool may vary depending on patient-related characteristics and intrinsic biases encountered during the two phases of FC determination: the pre-analytical phase (stool storage and FC extraction) and the analytical phase during which the test and thresholds are decided.24 To address these issues, an international consensus recently standardized practical tips to homogenize FC assessment: the preference for quantitative FC determination and the consistent use of the same quantification technique whenever possible.25

Fecal calprotectin and diagnosis

FC is the most studied biomarker in the field of IBD. FC was first quantified in IBD patients by Roseth and colleagues in 1992.26 In a subsequent study, the same authors reported higher levels of FC in patients with low-to-mild UC compared to healthy controls (HC).27 Several pooled analyses have comprehensively aggregated FC accuracy data, distinguishing between IBD and functional patients. Von Roon and colleagues,28 pooling data from 5,983 patients, reported a sensitivity of 0.95 (95% CI 0.93–0.97) and a specificity of 0.91 (95% CI 0.86–0.91) in discriminating IBD from HC. Menees et al. identified an optimal cut-off value for FC around 50 µg/L, which could exclude organic inflammation from IBS patients with fair accuracy.29 A more recent meta-analysis, which retrieved data from 2,145 patients subgrouped into organic versus functional gastrointestinal disorders (of which 1,059 had IBD), reported a pooled FC sensitivity and specificity of 88% (95% CI, 80–93%) and 72% (95% CI, 59–82%) in differentiating IBD from IBS.29

Fecal calprotectin and endoscopy

Non-invasive measurement of FC concentration accurately predicts endoscopic disease activity. Petryszyn et al. demonstrated that colonoscopy could be avoided in 66.7% of cases by incorporating FC into the diagnostic work-up for IBD.30 Mosli et al. pooled data from 2,499 patients, equally divided between UC and CD, reporting an overall sensitivity of 0.88 (95% CI 0.84–0.90) and specificity of 0.73 (95% CI 0.66–0.79) for predicting endoscopic activity in IBD patients.31 The authors identified a relatively low cut-off threshold for endoscopic activity assessment at 50 µg/g, demonstrating a high negative predictive value of FC determination with a pre-test probability of IBD of 25%.31 A recent study by Rokkas and colleagues set optimal cut-off values for FC at 50 µg/g for best sensitivity and 100 µg/g for best specificity in detecting IBD activity,32 while other studies found higher concentrations in patients in remission.33 Schoepfer et al. extensively evaluated the role of FC in describing disease activity, reporting higher FC levels (396 ± 351 versus 18.1 ± 5 µg/g) in active versus non-active UC patients.34 The correlation between FC and the Mayo endoscopic score was strong (Spearman score 0.834).34 The authors identified an overall accuracy of 89% for FC in detecting endoscopic activity in UC.34 Similarly, Walsh and colleagues defined thresholds of 71 µg/g, 91 µg/g, and 67.7 µg/g of FC, correlating well with endoscopic, histologic, and combined activity in 39 UC patients.35 The area under the curve (AUC) of FC for combined activity prediction was above 0.90.35 In CD, the correlation between FC levels and endoscopic activity was stronger when the disease was located in the colon rather than the ileum.36 The most recent large-cohort (273 CD patients) analysis on the clinical prediction value of FC in CD, provided by Li et al., reported optimal sensitivity and specificity in predicting endoscopic remission (sensitivity 68.02% and specificity 85.53%; AUC 0.83 with a cutoff value of 80.84 µg/g).37 Brand and colleagues conducted an intricate analysis, extrapolating 27 prediction models for assessing endoscopic activity in CD patients.38 Specifically, the researchers calculated that by setting a cut-off value of < 100 µg/g for FC, up to 72.5% of ileo-colonoscopies could be avoided, while potentially overlooking active CD in about 19.8% of cases as per the application of this predictive model.38 Schoepfer and colleagues also analyzed FC in CD, evidencing similar results, with a slightly lower correlation between FC and the SES-CD score (Spearman 0.75) but comparable accuracy of 87% with an FC cut-off of 70 µg/g.39 In both studies, FC performed better than clinical activity scores and blood component evaluations, such as blood leukocyte count.34,39

Fecal calprotectin and histology

FC has also been evaluated as a potential predictor of histologic activity in UC patients by Guardiola et al. More elevated levels of FC were found in histologically active UC patients (278 mg/g) compared to histologically inactive ones (68 mg/g; p < 0.002).40 In early retrospective evaluations of new rapid tests, FC has been found to correlate closely with both endoscopic and histologic activity for CD41 and UC.42

Fecal calprotectin and disease monitoring

Considering the severity of the disease, specifically for UC, FC values were also evaluated in acute-severe UC (ASUC). An FC > 800 µg/g at admission independently predicted the need for medical rescue therapy (OR 2.61, 95% CI, 1.12–6.12) and surgery within three months (OR 2.88, 95% CI, 1.01–8.17).43

FC is also a useful tool in disease monitoring, correlating with endoscopic and clinical remission,44 or predicting disease relapses during stable remission.40 Considering UC, a post hoc analysis of the GEMINI 1 study showed that an FC < 150 µg/g was associated with clinical and endoscopic remission (AUC range 0.70–0.77). Endoscopic evaluation should be indicated for patients with FC levels ≥250 µg/g, but not for those with FC values < 100 µg/g.45 An FC value of 130 µg/g predicted remission, while an FC > 300 µg/g was associated with relapse.46 These data are confirmed by the i-Support Therapy-Access to Rapid Treatment approach for patient-centered therapy in mild-to-moderate UC, which emphasizes the importance of regularly conducting FC measurements to monitor patients and promptly detect relapses to initiate suitable treatment.47 Exploring the latest studies on the use of FC to assess therapeutic response, Bertani and colleagues found a statistically significant correlation between FC levels and the prediction of mucosal healing (Mayo endoscopic score ≤1) in UC, with a sensitivity of 75% and specificity of 88.9%. This correlation was observed 8 weeks after the initiation of anti-TNF or Vedolizumab therapy.48 Similar prior studies have corroborated these findings.49,50 In UC, a recent standardization on the use of biomarkers to monitor inflammation during remission of treatment response has been achieved.51 The American Gastroenterology Association developed a clinical practice guideline, advising an FC cut-off of >150 µg/g as optimal for specificity and sensitivity in predicting endoscopic activity in patients with moderate to severe symptoms of UC.51 Considering CD, Haisma et al. found that early pursuit (< 12 weeks) of FC normalization was associated with lower disease relapse and better long-term outcomes.52 For patients with persistently high levels of FC, the probability of experiencing a relapse could potentially reach 83%.53 This data was confirmed in subsequent studies, which reported that high FC values predicted relapse after anti-TNF therapy discontinuation, in both CD and UC.46,54 Guidi et al. assessed FC values before and one year after induction and maintenance therapy with anti-TNF α in CD patients, finding a mean decrease of FC from 308 µg/g to 106 µg/g (p < 0.0001) for the sustained clinically responsive group, with no significant reduction in those who did not improve clinically (sensitivity of 83% and specificity of 74% for post-induction FC).55 A recent study by Magro and colleagues, which collected prospective data from 289 CD patients treated with infliximab (IFX) for two years, analyzed the progression of the disease through two composite outcomes. The first composite outcome integrated clinical factors such as initial occurrence of surgery or hospitalization related to IBD, or the emergence of new fistulas, abscesses, or strictures, and drug-related factors such as initial prescription of either at least one course of oral corticosteroids or more than 10 mg of prednisolone per day, or the initiation of de novo azathioprine or methotrexate, or a swap/switch to biological therapy (to adalimumab, golimumab, vedolizumab, or ustekinumab), or an increase in azathioprine dose unrelated to weight fluctuation, or an escalation in IFX dose or a reduction in dosing intervals. The second composite outcome considered adjustments in IFX dose and/or frequency.56 The authors demonstrated that FC > 500.0 µg/g was associated with complications of CD and the need for corticosteroid treatment during biological therapy (43.1% in composite outcome 1 and 26.9% in composite outcome 2; OR 3.069).56 Mild elevations (250.1–500.0 µg/g) were relevant when observed in at least two consecutive visits (33.3 % in composite outcome 1 and 15.2 % in composite outcome 2; p < 0.001 and p = 0.04 respectively). These results were confirmed by Cao et al., who demonstrated that high FC levels (>238 µg/g) during IFX maintenance treatment in CD patients predicted endoscopic activity within a one-year follow-up (p < 0.001).57

Considering the relevance of FC in clinical practice, easy-to-use and feasible kits for rapid assessment of FC in stools, such as “CalproSmart,” “QuantoCal,” and “IBDoc,” have been developed and can be performed directly at home.58 Sjoukje-Marije Haisma et al. compared these three home tests and found that within a low calprotectin range (≤500 µg/g), IBDoc, QuantOnCal, and CalproSmart demonstrated agreement rates of 87%, 82%, and 76%, respectively, with their corresponding ELISA. In the elevated range (>500 µg/g), the agreement stood at 37%, 19%, and 37%, respectively. The imprecision in the high range is of lesser concern because any calprotectin result exceeding 500 µg/g is interpreted as indicative of active disease, regardless of the specific concentration. Moreover, the CalproSmart and QuantOnCal smartphone applications experienced significantly higher rates of reading errors compared to the IBDoc application, with rates of 5.8% and 4.8% versus 1.9% (p = 0.002 and p = 0.012), respectively.59

Fecal calprotectin in the post-operative recurrence (POR)

Fecal biomarkers have demonstrated accuracy in predicting POR in CD patients.60 Initially, FC < 272 µg/g revealed a strong correlation with endoscopic remission following surgery in CD patients (area under the receiver operating characteristic curve [AUROC] 0.93). The correlation between FC and the Crohn’s disease index of severity was stronger (r = 0.722; p < 0.001) compared to CRP (r = 0.362; p < 0.001) and leukocyte count (r = 0.327; p = 0.003).41 Additionally, an analysis by Yamamoto and colleagues demonstrated a positive correlation between FC and endoscopic recurrence in CD patients who underwent ileocolonic resection (p = 0.0001).61 Conversely, blood markers such as CRP and leukocyte count did not perform as well. In the POCER trial, a subgroup analysis revealed higher FC values in patients who experienced a recurrence (275 µg/g vs. 72 µg/g, p < 0.001).62 This data was confirmed in a real-world cohort of patients with CD and endoscopic recurrence after ileocolic resection.63 Specifically, with an FC threshold of 50 µg/g, sensitivity was 90% and negative predictive value was 93%, while specificity and positive predictive value were 48% and 38%, respectively.63 Recently, it was identified that a combination of FC > 50 µg/g and bowel wall thickness measured by standard intestinal ultrasound had a very high positive predictive value(94%), with an OR of 8.58 (p < 0.001) in predicting post-operative recurrence in adult CD patients.64

In conclusion, the STRIDE-II consensus has revisited and refined key messages from the initial consensus, emphasizing the importance of biochemical non-invasive evaluation as an early to intermediate objective. This approach aims to enhance and expedite the follow-up of IBD, not only in terms of treatment response but also in specific scenarios such as POR.16

Fecal Lactoferrin (FL)

FL is an 80 kilodaltons (kDa) monomeric glycoprotein that exerts the main function of binding iron in its Fe3+ form and transporting it in the blood (with the capacity to also bind other ions like magnesium and zinc).65 FL plays a key role in innate immunity processes, being released by several immune cells, primarily neutrophils (from secondary granules).66 FL exerts both anti-bacterial and antifungal activities thanks to its Fe3+ binding capacity.67 Even anti-parasitic effects (mostly against Entamoeba histolytica due to lipid membrane binding and disruption68) and antiviral effects (towards non-enveloped RNA viruses by viral entry inhibition69) of FL have been described. Overall, FL plays an anti-inflammatory role by reducing the production of pro-inflammatory cytokines,66 promoting apoptosis of uncontrolled damaged cells, and blocking the cellular cycle of malignant cells, thus exhibiting potential anti-carcinogenic activity.70

FL has been proven as a useful fecal biomarker during various inflammatory conditions,71 with recent increasing interest in IBD pathogenesis and diagnosis.66 Menees et al., evaluating the available literature on FL, were unable to conclusively determine its discriminative power of FL for distinguishing IBD from HC or IBS patients.29 In the context of IBD, the highest predictive likelihood for disease using FL was 20.4% at a concentration of 1,810 µg/g. A level of 10 µg/g was associated with a 2% probability of IBD. Among all individual markers, FL exhibited the highest predictability for IBS at 74% with a concentration of 2,960 µg/g.29 Zhou and colleagues analyzed data from 1,012 patients, revealing a pooled sensitivity of 0.78 (95% CI 0.75, 0.82), specificity of 0.94 (95% CI 0.91, 0.96), combining for an AUC of 0.94 (95% CI: 0.90, 0.98), and OR of 52.65 (95% CI: 25.69, 107.91) in discriminating IBS from IBD using FL.72 Similar results were found in subsequent reports.73

FL has shown a correlation with endoscopic and histologic activity in colonic CD,74 even in pediatric patients.75 The most recent pooled analysis of 10 available studies identified good sensitivity and specificity values for FL in detecting activity. The combined sensitivity and specificity values for evaluating UC activity were 0.81 (95% CI 0.64–0.92) and 0.82 (95% CI 0.61–0.93), respectively. Additionally, the pooled sensitivity and specificity values for assessing CD activity were 0.82 (95% CI 0.73–0.88) and 0.71 (95% CI 0.63–0.78), respectively. Notably, the diagnostic performance of the FL assay in UC patients appeared to be superior to that in CD patients.76 In some studies, FL levels correlated with endoscopic disease activity. A meta-analysis of patients with CD reported combined sensitivity and specificity values for FL in detecting endoscopic activity at 75% and 80%, respectively.77

FL has also gained recognition as a fecal biomarker in monitoring IBD.66 It has shown a strong correlation, particularly following anti-TNF therapy in CD.78 A cutoff of 10 mg/g correlated with endoscopic response defined as a Crohn’s disease index of severity (≤3) (Spearman’s r 0.773, p < 0.001).78 In a recent post-hoc analysis of UNIFI and PURSUIT trials, FL has been targeted as a potential marker of worse long-term outcomes following anti-TNFα or anti-IL12-23 therapy in UC.79 FL levels above 84.5 µg/mL predicted a low likelihood of clinical (OR 0.43; p < 0.001), endoscopic (OR [95% CI]: 0.40 [0.29, 0.56]; p < 0.001), and histological (OR [95% CI]: 0.27 [0.14, 0.53]; p < 0.001) remission.79 Despite this, the use of FL is not widespread.

Moreover, FL’s role in predicting response (assessed with HBI and partial Mayo score) to biological agents during induction in both UC and CD has been demonstrated.80 Baseline FL values were higher in the non-responder group compared to responders (2,221 ± 1,910 vs. 773 ± 1,054 µg/mL, p = 0.02). Similarly, the initial FL drop was less on average in non-responders, although the difference was not statistically significant (62 ± 36 vs. 83 ± 16%, p = 0.09).80

Serum biomarkers

CRP

CRP is an acute-phase inflammatory protein, which can increase by 1000-fold during an acute response in various settings.81 CRP is a pentameric protein with five non-covalently linked subunits of 206 amino acids and a molecular weight of 23 kDa.82 CRP has numerous functions within the human immune system. CRP is produced by the liver in response to increased levels of pro-inflammatory cytokines such as interleukin (IL)-1, IL-6, or TNFα.83 IL-6 has been identified as the primary and most significant stimulator of CRP production by hepatocytes during inflammatory states.84 CRP rises within the first few hours following the initial causative stress (such as tissue injury or infection), typically reaching an initial concentration of around 0.8 mg/L.85 Moreover, CRP shows a late-onset curve in serum, especially during acute bacterial infections, compared to other infectious biomarkers like white blood cells.86 The main role of CRP in immune system activation and defense is to promote the activation of the complement cascade.87 Additionally, it plays a crucial role in cell-mediated immunity by binding to the FcεRi receptor of immunoglobulin-G (IgG) during antigen presentation.88

CRP has been studied as a tool to aid in the differential diagnosis between IBD and IBS since the mid-1980s.89 In a landmark population-based study by Fagan and colleagues, mean CRP values were found to be elevated in both CD and UC patients. Specifically, CRP levels were consistently higher in patients with moderate to severe disease compared to those with mild to moderate disease. Furthermore, CRP values were higher in CD compared to UC at the same disease activity level (4, 0–65 mg/L in mild CD vs 0, 0–15 mg/L in mild UC; 15, 1–100 mg/L in moderate CD vs 3, 0–29 mg/L in moderate UC; 85, 15–183 mg/L in severe CD vs 12, 2–33 mg/L in severe UC).90 The work by Menees and colleagues evaluated the accuracy of CRP in differentiating IBS, IBD, and HC from four available studies using a Bayesian predictive algorithm, demonstrating a 90% likelihood probability of suffering from IBD for a threshold CRP level above 2.7 mg/L.29 Despite some limitations in CRP accuracy,91 its usefulness as a biomarker in IBD remains relevant.92 Particularly when combined with other biomarkers, CRP can significantly improve diagnostic performance.93

CRP is traditionally recognized as one of the principal serum biomarkers of disease activity in IBD.94 Schoepfer et al. extensively evaluated the role of CRP in describing disease activity, reporting higher levels (16 ± 13 versus 3 ± 2 mg/L) in active UC versus non-active patients, with 69% accuracy in detecting endoscopic activity.34

The role of CRP is significant in ASUC. Particularly, the CRP/albumin ratio (CAR) is a better predictor of response to steroid therapy in ASUC than CRP or albumin values alone (AUC = 0.75; p < 0.001). The optimal CAR to predict response to steroids on day 3 is 0.85 (sensitivity 70%, specificity 76%).95 Moreover, following IFX rescue therapy for ASUC, CAR emerged as a straightforward biomarker, demonstrating robust predictive capabilities for the likelihood of colectomy. A day 3 CAR cutoff of 0.47 had 79% sensitivity, 80% specificity, and 94% negative predictive value to predict colectomy.96

CRP showed lower accuracy values compared to FC in detecting endoscopic activity. Specifically, in an intermediate pre-test scenario, an 11.5% rate of false positives for elevated CRP levels has been determined.48

CRP is also a useful marker in disease monitoring, allowing therapy adjustment according to the treat-to-target strategy.10 In patients with CD treated with IFX, an increase in CRP levels >10.0 mg/L in at least one visit was associated with a higher need for oral corticosteroid treatment during biological therapy [(44.3% in composite outcome 1 and 25.7% in composite outcome 2; OR 3.187); composite outcome 1 and 2 are described in the section 3.1.4].56 Mild elevations (3.1–10.0 mg/L) were only considered relevant when observed in at least two visits, whether consecutive or not (35.2% in composite outcome 1 and 18% in composite outcome 2; p < 0.001 and p = 0.007, respectively). These results were confirmed by Cao et al., who demonstrated that high CRP levels (>3.00 mg/L) during IFX maintenance treatment in CD patients predicted moderate endoscopic activity outcomes within a one-year follow-up (p = 0.012).57

Despite its utility as a non-invasive biomarker for IBD, it is important to note that 20–25% of IBD patients do not produce CRP during a flare.97 Additionally, CRP demonstrates a weaker correlation with disease activity in UC patients compared to those with CD.98

The accuracy in diagnosis, disease activity, and disease monitoring of FC, FL, and CRP is shown in Table 1.

Table 1

Summary of accuracy of principal biomarkers in diagnosis, disease activity, and disease monitoring of IBD

DiagnosisDisease activityDisease monitoring
FCIBD from HC: se: 0.95 (95% CI 0.93 0.97); sp: 0.91 95% CI 0.86–0.91). IBD from IBS: se: 88% (95% CI, 80–93%); sp: 72% (95% CI, 59–82%).Predicting endoscopic activity in IBD: se: 0.88 (95% CI 0.84–0.90); sp: 0.73 (95% CI 0.66–0.79). UC: correlation between FC and MES (Spearman score 0.834): accuracy of 89% in predicting endoscopic activity; AUC of 0.90. CD: correlation between FC and the SES-CD score (Spearman 0.75); accuracy of 87% in predicting endoscopic activity.IBD: repeated FC measurements above the study’s cutoff level had a 53% to 83% probability of developing disease relapse. UC: AUC range 0.70–0.77 for FC reduction >90%, FC ≤ 150 µg/g that indicates clinical and endoscopic remission; se: 75% and sp: 88.9% in predict mucosal healing after 8 weeks of treatment. CD: se: 83% and sp: 74% for FC post-induction treatment.
FLIBD from IBS: se: 0.78 (95% CI 0.75, 0.82), sp: 0.94 (95% CI 0.91, 0.96).UC: se: 0.81 (95% CI, 0.64–0.92); sp: 0.82 (95% CI, 0.61–0.93); CD: se: 0.82 (95% CI, 0.73–0.88) sp: 0.71 (95% CI, 0.63–0.78).FL > 84.5 µg/mL predicted a low likelihood of clinical (OR 0.43) endoscopic (OR 0.40) and histological (OR 0.27) remission.
CRPDifferentiating IBS, IBD and HC: CRP > 2.7 mg/L probability of 90% to suffer from IBD.UC: 69% accuracy in detecting endoscopic activity.

Autoantibodies in IBD assessment

There has been a suggestion regarding the potential involvement of antibodies in the diagnostic differential process of IBD.99 Antibodies against neutrophil cytoplasmic antigens (ANCA) are currently serum biomarkers useful in the diagnostic assessment of several inflammatory diseases.100 ANCA can be divided into two sub-groups: cytomplasmic ANCA and preinuclear ANCA (pANCA), depending on the location within the cell where the antigens bound by these autoantibodies are detected by ELISA staining.101 pANCA are directed against myeloperoxidase, while cytomplasmic ANCA target proteinase 3.102

These autoantibodies were first evaluated in IBD by Rump and colleagues in 1990. They found elevated titers of p-ANCA in UC patients with active disease, while no increase was reported in the CD cohort.103 Ruemmele and colleagues found a specificity of 100% for both IgA and IgG pANCA levels for UC diagnosis versus non-IBD controls, while lower levels of this marker were reported in CD patients.104 Other authors evaluated high levels of pANCA in CD patients with UC-like colitis characteristics, suggesting a potential clinical subgroup distinction using this biomarker.105 In contrast, in CD, an increased titer of anti-Saccharomyces cerevisiae (ASCA) antibodies has been reported for years.106 ASCA are specific autoantibodies known to directly bind the mannose residue on the cell surface mannan of Saccharomyces Cerevisiae. They were described long ago in coeliac disease.107 Other microbial species, namely Candida albicans, have also been shown to be immunogenic for ASCA production in CD.108 Peeters and colleagues in 2001 investigated the accuracy of ASCA and pANCA in the differential diagnosis of IBD, achieving considerable levels of specificity (up to 92%) but very low sensitivity (around 60%), even when combined, indicating limited diagnostic feasibility in IBD.99

Other antibodies associated with the diagnosis of IBD include anti-outer-membrane porin C and anti-CBir1 antibodies.109 Their detection is associated with the diagnosis of IBD.110,111

A recent study reported a correlation between primary non-response to anti-TNF therapy in UC patients and high levels of pANCA antibodies (p > 0.0002), suggesting a role for autoantibodies in predicting treatment response in IBD.112 The previously mentioned low sensitivity of autoantibodies as biomarkers of inflammation has severely limited their use as diagnostic non-invasive tools, reducing research interest in them.99 Nonetheless, individually assessing the feasibility of all these potential biomarkers could be debated. It is crucial to consider the utilization of a panel of multiple biomarkers when aiming to enhance the accuracy of disease prediction in IBD patients.113 A recent validation of this approach is represented by the study of Plevy and colleagues, which explored the diagnostic performance of a multi-panel test based on inflammatory markers (such as CRP) in combination with several serological markers (especially ASCA, ANCA, outer-membrane porin C, CBir1). This study demonstrated a substantial increase of the AUC in discriminating IBD from non-IBD patients (from 0.80 to 0.87; 95% CI, −0.4 to 0.4; p < 0.001) and between CD and UC (from 0.78 to 0.93; 95% CI, −0.4 to 0.4; p < 0.001).114

Erythrocyte sedimentation rate (ESR)

The ESR is a serological biomarker of inflammation, reflecting the degree of blood serum aggregation influenced by increased production of acute inflammatory proteins and metabolites.115 ESR values are affected by hematocrit and red blood cell aggregation.116 The most comprehensive examination of ESR as a diagnostic marker for IBD stems from the aforementioned study by Menees et al. In this study, no ESR levels demonstrated statistically significant prediction of IBD, indicating that ESR did not exhibit notable accuracy compared to FC and CRP.29 Even ESR has been evaluated in active IBD cohorts, showing no significant differences between the two disease phenotypes, UC versus CD.117 However, there was concordance with CRP and FC in predicting endoscopic and clinical activity.118

Data available from more recent meta-analyses in the literature on the feasibility of the principal biomarkers assessed in the diagnosis of IBD, both in adult and pediatric settings, are summarized in Table 2.27,29–31,76

Table 2

Studies focusing on diagnostic accuracy of biomarkers in IBD diagnosis in adults and children

Author and dateStudy designCohort N.Patients typeDiseaseAssessed biomarkerAccuracy
Von Roon et al.27Metanalysis5,983 (CD = 663, UC = 361, IBD = 186)AdultsIBD vs HCFCSe 0.95 (95% CI 0.93–0.97); Sp 0.91 (95% CI 0.86–0.91)
Menees et al.29Metanalysis2,145 (IBD = 1,059)AdultsIBD vs IBS vs HCFC, FL, ESR, CRPMaximal predictive value of IBD for FC: 78.7% at 1,000 µ/g; IBS exclusion for FC < 40 µ/g; Maximal predictive value of IBD for CRP: 90% at 2.7 mg/dl; FL and ESR no predictive values
Mosli et al.31Metanalysis2,499 (UC = 1,069, CD = 1,033)AdultsIBDFC, FL, CRPCRP: Se 0.49 (95% CI 0.34–0.64); Sp 0.92 (95% CI 0.72–0.96); FC: Se 0.88 (95% CI 0.84–0.90); Sp 0.73 (95% CI 0.66–0.79); FL: Se 0.82 (95% CI 0.73–0.88); Sp 0.79 (95% CI 0.62–0.89)
Petryszyn et al.30Metanalysis5,032 (IBD = 620)Adults and childrenIBD vs HCFCSe: 0.88 (95% CI, 0.827–0.921); Sp: 0.79 (95% CI, 0.693–0.875)
Dai et al.76Metanalysis936 (IBD = 773)AdultsIBDFLUC: Se 0.81 95% CI, 0.64–0.92; Sp 0.82 95% CI, 0.61–0.93; CD: Se 0.82 95% CI, 0.73–0.88; Sp 0.71 95% CI, 0.63–0.78

Urine markers

Urine biomarkers have shown limited utility in assessing IBD activity; however, certain studies have explored urine as a potential source of non-invasive biochemical indicators for disease activity.119 Prostaglandins are synthesized through the action of A2 phospholipase, which catalyzes the phosphorylation of arachidonic acid. This process is triggered by various inflammatory triggers.120 Prostaglandins serve numerous functions, particularly in mucosal inflammation and the recruitment of inflammatory cells.121 PGE2 is excreted in urine in the form of several processed metabolites, primarily Tetranor-prostaglandin E metabolite. Arai and colleagues evaluated urinary concentrations of Tetranor-prostaglandin E metabolite, demonstrating good agreement with FC in describing active inflammation in UC (p < 0.01), and also predicting histological and endoscopic remission.122 In pediatric patients, urinary PGE2 showed a positive correlation with both endoscopic and clinical activity indexes in UC (r = 0.594 and r = 0.462, respectively).123 Recently, the role of PGE2 in predicting disease relapse has been investigated, with a reported predictive value of 25.2 µg/g and an AUC of 0.721 (95% confidence interval: 0.556–0.886).124 Patients with PGE2 values ≥25.2 µg/g experienced a significantly shorter relapse-free period (log-rank test: p < 0.001).124 The feasibility of PGE2 and its metabolites in urine warrants further research efforts. Future studies are needed to explore this potential avenue.

Unveiling novel IBD indicators: Exploring promising markers on the horizon

Several biomarkers have been evaluated since the end of the 1990s to assess the presence of CD or UC.18 Other biomarkers with promising results are currently under investigation but still need to be incorporated into clinical practice.

Myeloperoxidase

Myeloperoxidase (MPO) is a heme-containing enzyme primarily found in azurophil granules of neutrophils and in monocyte lysosomes.125 MPO is a crucial component of the cytoplasmic antimicrobial compartment in phagocytic cells. MPO produces oxygen reactants, particularly hypochlorous acid.126 One of the first influential studies on fecal MPO and IBD was conducted in 1998 by Saiki T. The study enrolled a total of 33 UC patients, 32 CD patients, and 15 HC, demonstrating significantly elevated levels of MPO concentration in stools among IBD patients compared to the control group (p < 0.001).127 Regarding disease activity, a paired analysis highlighted reduced levels of MPO in stools of patients with UC in remission compared to those with active disease (p < 0.001).127 Anezaki and colleagues found concordant levels of MPO and IL-8 (measured with ELISA testing) in the stools of active UC patients versus inactive ones.128 Regarding disease monitoring, Sangfelt et al. evaluated the decrease in MPO levels after seven days of corticosteroid treatment in patients with distal and rectal UC.129 However, there is currently insufficient evidence to support the use of MPO as a biomarker, and it is not recommended for clinical practice.

Calgranulin C

Calgranulin C, a calcium-binding protein related to the S100 superfamily, has been found to increase in stools during inflammatory flares.130 Calgranulin C is expressed exclusively by neutrophils and has the ability to target the RAGE protein, inducing NF-κB and MAP-kinase pathways, leading to increased production of proinflammatory cytokines.131 In 2003, Foell and colleagues reported a statistically significant increase in calgranulin C levels in both CD and UC compared to HC using sandwich ELISA testing (p < 0.0001).132 A recent study aimed to analyze the concentration of calgranulin and FC in IBD pediatric patients.133 The authors found a direct positive correlation between FC and calgranulin C levels, as well as significant consistency in calgranulin levels assessed during active disease versus remission (p = 0.02).133 However, despite these findings, the use of calgranulin C as a biomarker is not authorized in clinical practice.

Lipocalin 2

Another potential marker of inflammation in IBD patients is Lipocalin 2 (L2). L2 is an adipo-cytokine, a member of the lipocalin family, small proteins capable of binding hydrophobic molecules on cell surfaces and forming large aggregates.134 L2 has been studied in various diseases, including diabetes mellitus,135 and in IBD as well.136 In a study by Thorsvik et al., the L2 concentration levels in the stools of 73 IBD patients were compared to HC and IBS patients, with significantly higher values of this marker in IBD compared to the other groups (difference of 0.3 mg/kg in UC and CD vs HC, p < 0.001 for both; difference of 0.4 mg/kg in UC and CD vs IBS, p < 0.001 and p = 0.004, respectively).137 Another study by Magro and colleagues identified a cutoff level of L2 in stool samples (approximately 12 µg/g) to predict endoscopic and histologic remission in UC.138 L2 is relevant in gut inflammation also due to its interaction with another fecal biomarker: fecal matrix metalloprotease-9 (MMP-9), a member of zinc-dependent endopeptidases known for degrading extracellular matrix, crucial in cell-extracellular environment interactions.139 Moreover, L2 can physiologically form complexes with neutrophil gelatinase-associated lipocalin, intricate structures of 198 amino acids with anti-inflammatory functions, equipped with terminal domains capable of binding and stabilizing several proteins such as MMP9 itself.140 Recently, Buisson and colleagues evaluated the accuracy of MMP9 along with L2 in CD patients, finding good reliability of MMP9 (sensitivity up to 90%) and slightly lower for L2 (sensitivity up to 87.5%) in detecting endoscopic and clinical activity.141

Micro-RNA (miRNA)

A novel trend in the non-invasive evaluation of IBD is represented by miRNA assessment in fecal samples.142 miRNAs consist of a group of small non-coding RNAs with variable expression in various diseases, from neurological disorders to cancer.143 miRNAs can be assessed in serum as well as in all biological specimens, including stools, using various molecular techniques, primarily PCR and Next Generation Sequencing.143,144 miRNA 21 is one of the most important miRNAs found to be increased in IBD compared to non-IBD controls.145 miRNA 21 and miRNA 223 were evaluated in the stools of IBD patients, with higher levels observed in UC compared to CD (miRNA 223 was 5.7-fold higher for CD and 10.2-fold higher for UC compared to controls).146

Cytokines and interleukins

Serum pro-inflammatory cytokines and interleukins have been extensively characterized in IBD.147 TNFα has been found to be elevated in the colonic and ileal mucosa of patients suffering from IBD.148 Despite discordant results in previous studies on serological samples,149 Komatsu and colleagues used PCR methods and found a statistically significant increase in serum concentrations of TNFα in IBD patients (both CD and UC) compared to HC (p < 0.0001).150 These results suggest a potential use of TNFα serum levels as a biomarker for assessing IBD activity. Serum concentrations of IL-10 in IBD patients have also been evaluated by Kucharzik and colleagues, who reported higher concentrations of serum IL-10 in active CD and UC patients compared with HC (p < 0.001).151 Mitsuyama and colleagues conducted an analysis of three inflammatory biomarkers—IL-6, CRP, and IL-10—in patients with UC, CD, and HC. The authors found increased levels of serum IL-6 and CRP in active UC and CD compared to HC (p < 0.0001), while higher serum concentrations of IL-10 were observed only in active UC compared to HC (p = 0.0086).152 The role of serum interleukins in assessing and monitoring IBD requires further evaluation through larger observational studies.

Nitric oxide

Additionally, nitric oxide (NO) is a recognized mediator of several processes related to local and systemic inflammation.153 NO is synthesized through the oxidation of the amino acid L-arginine by a family of enzymes known as NO synthases.153 There exist three isoforms of NO synthase: neuronal, expressed in the brain and peripheral nervous system; endothelial, found in endothelial cells; and inducible NO synthase, which activates in response to microbial products such as IL-1 or TNF-α.154 Inducible NO synthase activity has been demonstrated in active UC.155 The potential role of NO as a mediator in the inflammatory processes of IBD has garnered significant interest. Various manifestations of IBD have been found to correlate with NO, including vasodilation and increased vascular permeability, directly or indirectly.153 Avdagić et al. conducted a study exploring the potential of serum NO as a biomarker for diagnosing UC and CD.156 The results indicated statistically significant differences in serum NO levels among UC patients, CD patients, and HC. The median NO concentrations in UC patients, CD patients, and HC were 15.3 µM, 14.5 µM, and 13.3 µM, respectively. Using a cutoff of 17.4 µM, both the sensitivity and specificity of NO in distinguishing between active and inactive UC patients were 100%. With a cutoff of 14 µM, the sensitivity and specificity of NO in distinguishing between active and inactive CD patients were 88% and 69%, respectively. These findings suggest that serum NO could potentially serve as a biomarker for IBD.

Neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and albumin-to-globulin ratio

There has been increasing attention on blood cell count-based ratios such as the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR), and their potential relation to various immune-mediated inflammatory disorders.157 A recent study by Soufli et al. assessed serum concentrations of NO, NLR, and PLR in complicated CD patients compared to HC, finding statistically significant differences in these markers between the two groups (p < 0.001) before and after corticosteroid or anti-TNF therapy. Cut-off values for disease prediction were identified (NLR > 2.43; PLR > 156.4, respectively).158 Additionally, the albumin-to-globulin ratio (AGR) may emerge as a promising tool to aid clinicians in differentiating IBD and evaluating disease activity.159 AGR is notably reduced in IBD patients compared to those without IBD. In individuals with UC, there is an inverse correlation between serum AGR and the Mayo score (r = −0.413, p < 0.001), whereas in patients with CD, serum AGR shows an association with HBI (r = −0.471, p < 0.001).159

Suppression of tumorigenicity 2 (ST2) and TNF-α-induced protein 6 (TNFAIP6)

ST2 and TNFAIP6 are two novel serological biomarkers recently studied in patients with IBD. ST2 is an interleukin belonging to the IL-1 superfamily.160 It has been reported that ST2 is positively correlated with endoscopic and clinical activity in UC and CD compared to HC.161 Furthermore, serum levels of ST2 were found to be lower in individuals with UC who responded to treatment (conventional therapies such as 5-ASA derivatives, corticosteroids, or immunomodulators) compared to non-responders (p < 0.05).162 Additionally, among patients who responded to treatment, the level of intestinal ST2 remained consistent within the cellular infiltrate of the lamina propria during the six-month follow-up. In contrast, patients experiencing reactivation showed an elevation in total ST2 within the inflamed mucosa, though it remained confined to the cellular infiltrate.162 Similarly, elevated TNFAIP6 levels (a 35 kDa glycoprotein with terminal halves sharing homologous regions with terminal peptides of other immune proteins such as C1r/C1s or EGF)163 have been detected during episodes of acute inflammation.164 Its potential for diagnosing and monitoring IBD appears promising, but further clarification is certainly warranted.165

Autoantibodies against malondialdehyde-acetaldehyde adduct

A promising serological marker for diagnosing IBD is autoantibodies against the malondialdehyde-acetaldehyde adduct (MAA). IBD is associated with immune responses that involve oxidative stress, where elevated levels of malondialdehyde contribute to the formation of a highly stable and immunogenic MAA.166 Specifically, Duryee et al. demonstrated that IgG anti-MAA antibody levels could accurately identify UC with a sensitivity of 75%, specificity of 71%, and an AUC of 0.81.166

Calprotectin neo-epitope (CPa9-HNE)

Another serological marker that correlates with disease activity in IBD is CPa9-HNE.167 Serological levels of this marker are higher in both CD and UC patients compared to HC (p < 0.0001 for both). CPa9-HNE shows a significant association with the SES-CD (r = 0.61, p < 0.0001) and the full Mayo score (r = 0.52, p = 0.0013). It can effectively differentiate between CD and UC patients in terms of endoscopic remission and moderate/severe disease activity (CD: AUC = 0.82, UC: AUC = 0.87). Moreover, other two serological markers of disease activity in IBD are dipeptidyl peptidase activity circulating and proteins of extracellular matrix remodeling such as biomarkers of type III collagen degradation and formation, type IV collagen degradation and formation, and type V collagen formation.168,169

Anti-integrin αvβ6 antibodies

Integrin αvβ6 functions as a receptor for extracellular matrix proteins, notably fibronectin, and its expression is limited to epithelial cells. It plays a significant role in maintaining epithelial barrier functions.170 Recent research, particularly in the Japanese population, has demonstrated elevated circulating levels of IgG against colonic epithelial integrin αvβ6 in adult patients with UC compared to those with CD and HC.171 These antibody levels were found to correlate with the severity of the disease.171 Subsequent studies in small Swedish and Italian cohorts confirmed these findings.172,173 Additionally, anti-integrin αvβ6 autoantibodies appear to precede the clinical diagnosis of UC and are associated with adverse UC-related outcomes.171

Transcriptomic markers

Considering the heterogeneity and varied therapeutic outcomes among IBD patients who exhibit similar clinical, endoscopic, and histologic activity, a more personalized approach is necessary.174 Perez et al., in a meta-analysis, examined transcriptomic profiles of 1,047 samples from five cohorts to distinguish UC, colonic CD, ileal CD, and pouchitis in comparison to normal colonic and ileal mucosa. They subsequently conducted a meta-analysis focusing on distinct transcriptomic signatures associated with ileal and colonic manifestations of these diseases.175 They identified specific markers indicating inflammation in the ileum (FOLH1, CA2) and colon (REG3A), and demonstrated that as the disease progresses, specific cells in the ileum begin expressing markers typically associated with the colon. Immunohistochemistry validated the specificity of these markers for ileal or colonic diseases. These findings highlighted that colonic CD resembles UC more than ileal CD, which shares similarities with pouchitis. Transcriptomic analysis, in addition to aiding diagnosis, may also predict endoscopic and histologic healing in IBD patients. Biopsies from 111 UC patients treated with ritlecitinib (an oral JAK3/TEC inhibitor) were analyzed by Hassan-Zahraee et al. Ten genes (CXCL1, FCAR, CKAP4, SPINK4, CXCL17, OSM, CD4, CXCL9, IL17A, and GZMB) exhibited significant alterations from baseline in responders compared to non-responders at week 8, particularly in terms of endoscopic improvement or histological remission. Additionally, these genes showed a marked increase at baseline between colon biopsies with inflammation and those without inflammation.176 New transcriptomic biomarkers can predict therapeutic response, as Abreu et al. identified immune cell phenotypic and gene expression patterns associated with vedolizumab response.177 In this study, Treg cells, especially from the ileum, showed the most transcriptional differences at baseline in responders vs non-responders to vedolizumab, irrespective of CD or UC diagnosis (p < 0.05). This evidence supports molecular disease stratification over reliance solely on clinical criteria, opening possibilities for designing drugs that target diseases more specifically.

Discussion

The assessment of IBD activity is crucial for disease monitoring and evaluating treatment responses.10 The pursuit of achieving disease remission should be guided by a “treat-to-target” strategy, involving regular evaluations of disease activity.178 This approach aims to streamline treatment adjustments and enhance long-term outcomes.6 Serum and fecal biomarkers are the fastest and easiest tools to monitor the course of IBD.18 FC remains the most accurate marker of gut inflammation, demonstrating exceptional accuracy, sensitivity, and specificity in assessing disease activity, treatment efficacy, and predicting relapse.20 Despite its recognition as an inflammation marker, FL still faces challenges such as low specificity and a lack of significant validation studies, preventing its widespread adoption in routine clinical practice.91 Regarding serum markers, CRP, ESR, and blood cell counts are routinely used.92 Enhancing diagnostic accuracy through multi-target panels of biomarkers has demonstrated improved sensitivity and specificity. Novel biochemical targets such as fecal MPO, serum pro-inflammatory cytokines, interleukins, serum or fecal miRNAs, Calgranulin C, Lipocalin 2, NO, CPa9-HNE, transcriptomic markers, and urinary components (especially PGE2) are gaining attention in IBD research. However, further studies are needed to integrate them into clinical practice.179 Expanding the repertoire of biomarkers could serve as a pivotal role in implementing the treat-to-target strategy for monitoring IBD.180 While the potential of blood, fecal, and urinary biomarkers in IBD is promising, challenges remain. Achieving standardization in terms of assays, defining appropriate cut-off values, and establishing uniform interpretation criteria are crucial for their smooth integration into routine clinical practice. Longitudinal studies are also essential to confirm their efficacy in predicting disease flares, treatment response, and long-term outcomes. The evolving landscape of blood, fecal, and urinary biomarkers in IBD presents an exciting opportunity to revolutionize diagnostic and monitoring methodologies for these complex diseases. The incorporation of non-invasive biomarkers into clinical practice offers the potential for early detection, evaluation of disease activity, and tailored treatment approaches. As ongoing research unveils the complexity of IBD, integrating biomarkers into patient care becomes increasingly feasible. Through collaborative efforts and rigorous research, we can pave the way towards more effective management and improved quality of life for individuals navigating the challenges of IBD.

Conclusion

Assessing IBD activity is crucial for understanding treatment responses and long-term outcomes. A “treat-to-target” strategy, supported by regular evaluations, guides remission targets. Biomarkers serve as non-invasive tools for diagnosing and monitoring IBD, yet further studies are required to fully integrate them and predict relapses and treatment outcomes. The evolving landscape of biomarkers presents an opportunity to improve IBD management and promote personalized medicine.

Declarations

Acknowledgement

None.

Data sharing statement

No new data were generated or analyzed in support of this research.

Funding

None.

Conflict of interest

SB, AZ, TLP declare no conflict of interest. FD has served as a speaker for Sandoz, Janssen, Galapagos, and Omega Pharma; he has also served as an advisory board member for Galapagos, AbbVie, and NFF received consulting fees from Amgen, AbbVie, Janssen, Pfizer, and Galapagos. GF received consultancy fees from Ferring, MSD, AbbVie, Takeda, Janssen, Amgen, Sandoz, Samsung Bioepis, and Celltrion. SD has served as a speaker, consultant, and advisory board member for Schering-Plough, AbbVie, Actelion, Alphawasserman, AstraZeneca, Cellerix, Cosmo Pharmaceuticals, Ferring, Genentech, Grunenthal, Johnson and Johnson, Millennium Takeda, MSD, Nikkiso Europe GmbH, Novo Nordisk, Nycomed, Pfizer, Pharmacosmos, UCB Pharma, and VMA received consulting fees from Nikkiso Europe, Mundipharma, Janssen, AbbVie, and Pfizer.

Authors’ contributions

Conceiving the article (MA, FF), writing the article and creating tables and figures (SB, FF), critically reviewing the content of the paper, and supervising the project (FD, AZ, TLP, MA, GF, SD). The manuscript was approved by all authors.

References

  1. Torres J, Bonovas S, Doherty G, Kucharzik T, Gisbert JP, Raine T, Adamina M, et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical Treatment. J Crohns Colitis 2020;14(1):4-22 View Article PubMed/NCBI
  2. Agrawal M, Christensen HS, Bøgsted M, Colombel JF, Jess T, Allin KH. The Rising Burden of Inflammatory Bowel Disease in Denmark Over Two Decades: A Nationwide Cohort Study. Gastroenterology 2022;163(6):1547-1554.e5 View Article PubMed/NCBI
  3. Agrawal M, Jess T. Implications of the changing epidemiology of inflammatory bowel disease in a changing world. United European Gastroenterol J 2022;10(10):1113-1120 View Article PubMed/NCBI
  4. GBD 2017 Inflammatory Bowel Disease Collaborators. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2020;5(1):17-30 View Article PubMed/NCBI
  5. Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, et al. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis 2019;13(2):144-164 View Article PubMed/NCBI
  6. Garcia NM, Cohen NA, Rubin DT. Treat-to-target and sequencing therapies in Crohn’s disease. United European Gastroenterol J 2022;10(10):1121-1128 View Article PubMed/NCBI
  7. SSchroeder KW, Tremaine WJ, Ilstrup DM. Coated oral 5-aminosalicylic acid therapy for mildly to moderately active ulcerative colitis. A randomized study. N Engl J Med 1987;317(26):1625-9 View Article PubMed/NCBI
  8. Harvey RF, Bradshaw JM. A simple index of Crohn’s-disease activity. Lancet 1980;1(8167):514 View Article PubMed/NCBI
  9. Peyrin-Biroulet L, Reinisch W, Colombel JF, Mantzaris GJ, Kornbluth A, Diamond R, et al. Clinical disease activity, C-reactive protein normalisation and mucosal healing in Crohn’s disease in the SONIC trial. Gut 2014;63(1):88-95 View Article PubMed/NCBI
  10. Peyrin-Biroulet L, Sandborn W, Sands BE, Reinisch W, Bemelman W, Bryant RV, et al. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target. Am J Gastroenterol 2015;110(9):1324-1338 View Article PubMed/NCBI
  11. Bettenworth D, Bokemeyer A, Baker M, Mao R, Parker CE, Nguyen T, et al. Assessment of Crohn’s disease-associated small bowel strictures and fibrosis on cross-sectional imaging: a systematic review. Gut 2019;68(6):1115-1126 View Article PubMed/NCBI
  12. Rimola J, Torres J, Kumar S, Taylor SA, Kucharzik T. Recent advances in clinical practice: advances in cross-sectional imaging in inflammatory bowel disease. Gut 2022;71(12):2587-2597 View Article PubMed/NCBI
  13. Roda G, Chien Ng S, Kotze PG, Argollo M, Panaccione R, Spinelli A, et al. Crohn's disease. Nat Rev Dis Primers 2020;6(1):22 View Article PubMed/NCBI
  14. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001;69(3):89-95 View Article PubMed/NCBI
  15. Colombel JF, Panaccione R, Bossuyt P, Lukas M, Baert F, Vaňásek T, et al. Effect of tight control management on Crohn’s disease (CALM): a multicentre, randomised, controlled phase 3 trial. Lancet 2017;390(10114):2779-2789 View Article PubMed/NCBI
  16. Turner D, Ricciuto A, Lewis A, D’Amico F, Dhaliwal J, Griffiths AM, et al. 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(5):1570-1583 View Article PubMed/NCBI
  17. Jukic A, Bakiri L, Wagner EF, Tilg H, Adolph TE. Calprotectin: from biomarker to biological function. Gut 2021;70(10):1978-1988 View Article PubMed/NCBI
  18. Liu D, Saikam V, Skrada KA, Merlin D, Iyer SS. Inflammatory bowel disease biomarkers. Med Res Rev 2022;42(5):1856-1887 View Article PubMed/NCBI
  19. Chatzikonstantinou M, Konstantopoulos P, Stergiopoulos S, Kontzoglou K, Verikokos C, Perrea D, et al. Calprotectin as a diagnostic tool for inflammatory bowel diseases. Biomed Rep 2016;5(4):403-407 View Article PubMed/NCBI
  20. Laserna-Mendieta EJ, Lucendo AJ. Faecal calprotectin in inflammatory bowel diseases: A review focused on meta-analyses and routine usage limitations. Clin Chem Lab Med 2019;57(9):1295-1307 View Article PubMed/NCBI
  21. Tibble JA, Sigthorsson G, Foster R, Scott D, Fagerhol MK, Roseth A, et al. High prevalence of NSAID enteropathy as shown by a simple faecal test. Gut 1999;45(3):362-366 View Article PubMed/NCBI
  22. Røseth AG, Kristinsson J, Fagerhol MK, Schjønsby H, Aadland E, Nygaard K, et al. Faecal calprotectin: A novel test for the diagnosis of colorectal cancer?. Scand J Gastroenterol 1993;28(12):1073-1076 View Article PubMed/NCBI
  23. Tøn H, Brandsnes, Dale S, Holtlund J, Skuibina E, Schjønsby H, et al. Improved assay for fecal calprotectin. Clin Chim Acta 2000;292(1-2):41-54 View Article PubMed/NCBI
  24. 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(1):152-161 View Article PubMed/NCBI
  25. D’Amico F, Rubin DT, Kotze PG, Magro F, Siegmund B, Kobayashi T, et al. International consensus on methodological issues in standardization of fecal calprotectin measurement in inflammatory bowel diseases. United European Gastroenterol J 2021;9(4):451-460 View Article PubMed/NCBI
  26. Røseth AG, Fagerhol MK, Aadland E, Schjønsby H. Assessment of the neutrophil dominating protein calprotectin in feces. A methodologic study. Scand J Gastroenterol 1992;27(9):793-798 View Article PubMed/NCBI
  27. Røseth AG, Aadland E, Jahnsen J, Raknerud N. Assessment of disease activity in ulcerative colitis by faecal calprotectin, a novel granulocyte marker protein. Digestion 1997;58(2):176-180 View Article PubMed/NCBI
  28. von Roon AC, Karamountzos L, Purkayastha S, Reese GE, Darzi AW, Teare JP, et al. Diagnostic precision of fecal calprotectin for inflammatory bowel disease and colorectal malignancy. Am J Gastroenterol 2007;102(4):803-13 View Article PubMed/NCBI
  29. 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(3):444-454 View Article PubMed/NCBI
  30. Petryszyn P, Staniak A, Wolosianska A, Ekk-Cierniakowski P. Faecal calprotectin as a diagnostic marker of inflammatory bowel disease in patients with gastrointestinal symptoms: meta-analysis. Eur J Gastroenterol Hepatol 2019;31(11):1306-1312 View Article PubMed/NCBI
  31. Mosli MH, Zou G, Garg SK, et al. C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am J Gastroenterol 2015;110(6):802-819 View Article PubMed/NCBI
  32. Rokkas T, Portincasa P, Koutroubakis IE. Fecal calprotectin in assessing inflammatory bowel disease endoscopic activity: a diagnostic accuracy meta-analysis. J Gastrointestin Liver Dis 2018;27(3):299-306 View Article PubMed/NCBI
  33. Lin JF, Chen JM, Zuo JH, Yu A, Xiao ZJ, Deng FH, et al. Meta-analysis: fecal calprotectin for assessment of inflammatory bowel disease activity. Inflamm Bowel Dis 2014;20(8):1407-1415 View Article PubMed/NCBI
  34. Schoepfer AM, Beglinger C, Straumann A, Trummler M, Renzulli P, Seibold F. Ulcerative colitis: correlation of the Rachmilewitz endoscopic activity index with fecal calprotectin, clinical activity, C-reactive protein, and blood leukocytes. Inflamm Bowel Dis 2009;15(12):1851-1858 View Article PubMed/NCBI
  35. Walsh A, Kormilitzin A, Hinds C, Sexton V, Brain O, Keshav S, et al. Defining Faecal Calprotectin Thresholds as a Surrogate for Endoscopic and Histological Disease Activity in Ulcerative Colitis-a Prospective Analysis. J Crohns Colitis 2019;13(4):424-430 View Article PubMed/NCBI
  36. Sipponen T, Savilahti E, Kolho K-L, Nuutinen H, Turunen U, Färkkilä M. Crohn’s disease activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn’s disease activity index and endoscopic findings. Inflamm Bowel Dis 2008;14(1):40-46 View Article PubMed/NCBI
  37. Li J, Xu M, Qian W, Ling F, Chen Y, Li S, et al. Clinical value of fecal calprotectin for evaluating disease activity in patients with Crohn’s disease. Front Physiol 2023;14:1186665 View Article PubMed/NCBI
  38. Brand EC, Elias SG, Minderhoud IM, van der Veen JJ, Baert FJ, Laharie D, et al. Systematic Review and External Validation of Prediction Models Based on Symptoms and Biomarkers for Identifying Endoscopic Activity in Crohn’s Disease. Clin Gastroenterol Hepatol 2020;18(8):1704-1718 View Article PubMed/NCBI
  39. Schoepfer AM, Beglinger C, Straumann A, Trummler M, Vavricka SR, Bruegger LE, et al. Fecal calprotectin correlates more closely with the Simple Endoscopic Score for Crohn’s disease (SES-CD) than CRP, blood leukocytes, and the CDAI. Am J Gastroenterol 2010;105(1):162-169 View Article PubMed/NCBI
  40. Guardiola J, Lobatón T, Rodríguez-Alonso L, Ruiz-Cerulla A, Arajol C, Loayza C, et al. Fecal level of calprotectin identifies histologic inflammation in patients with ulcerative colitis in clinical and endoscopic remission. Clin Gastroenterol Hepatol 2014;12(11):1865-70 View Article PubMed/NCBI
  41. Lobatón T, López-García A, Rodríguez-Moranta F, Ruiz A, Rodríguez L, Guardiola J. A new rapid test for fecal calprotectin predicts endoscopic remission and postoperative recurrence in Crohn’s disease. J Crohns Colitis 2013;7(12):e641-51 View Article PubMed/NCBI
  42. Lobatón T, Rodríguez-Moranta F, Lopez A, Sánchez E, Rodríguez-Alonso L, Guardiola J. A new rapid quantitative test for fecal calprotectin predicts endoscopic activity in ulcerative colitis. Inflamm Bowel Dis 2013;19(5):1034-1042 View Article PubMed/NCBI
  43. Sasidharan S, Sasson AN, Shannon KM, Ananthakrishnan AN. Fecal Calprotectin Is a Predictor of Need for Rescue Therapy in Hospitalized Severe Colitis. Inflamm Bowel Dis 2022;28(12):1833-1837 View Article PubMed/NCBI
  44. Zittan E, Kelly OB, Kirsch R, Milgrom R, Burns J, Nguyen GC, et al. Low Fecal Calprotectin Correlates with Histological Remission and Mucosal Healing in Ulcerative Colitis and Colonic Crohn’s Disease. Inflamm Bowel Dis 2016;22(3):623-630 View Article PubMed/NCBI
  45. Reinisch W, Bressler B, Curtis R, Parikh A, Yang H, Rosario M, et al. Fecal Calprotectin Responses Following Induction Therapy With Vedolizumab in Moderate to Severe Ulcerative Colitis: A Post Hoc Analysis of GEMINI 1. Inflamm Bowel Dis 2019;25(4):803-810 View Article PubMed/NCBI
  46. Ferreiro-Iglesias R, Barreiro-de Acosta M, Lorenzo-Gonzalez A, Dominguez-Muñoz JE. Accuracy of Consecutive Fecal Calprotectin Measurements to Predict Relapse in Inflammatory Bowel Disease Patients Under Maintenance With Anti-TNF Therapy: A Prospective Longitudinal Cohort Study. J Clin Gastroenterol 2018;52(3):229-234 View Article PubMed/NCBI
  47. D’Amico F, Magro F, Caron B, Dignass A, Jairath V, Hart A, et al. 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(3):1142 View Article PubMed/NCBI
  48. Bertani L, Blandizzi C, Mumolo MG, Ceccarelli L, Albano E, Tapete G, et al. Fecal Calprotectin Predicts Mucosal Healing in Patients With Ulcerative Colitis Treated With Biological Therapies: A Prospective Study. Clin Transl Gastroenterol 2020;11(5):e00174 View Article PubMed/NCBI
  49. Toyonaga T, Kobayashi T, Nakano M, Saito E, Umeda S, Okabayashi S, et al. Usefulness of fecal calprotectin for the early prediction of short-term outcomes of remission-induction treatments in ulcerative colitis in comparison with two-item patient-reported outcome. PLoS One 2017;12(9):e0185131 View Article PubMed/NCBI
  50. Carlsen K, Riis LB, Elsberg H, Maagaard L, Thorkilgaard T, Sørbye SW, et al. The sensitivity of fecal calprotectin in predicting deep remission in ulcerative colitis. Scand J Gastroenterol 2018;53(7):825-830 View Article PubMed/NCBI
  51. Singh S, Ananthakrishnan AN, Nguyen NH, Cohen BL, Velayos FS, Weiss JM, et al. AGA Clinical Practice Guideline on the Role of Biomarkers for the Management of Ulcerative Colitis. Gastroenterology 2023;164(3):344-372 View Article PubMed/NCBI
  52. Haisma SM, Verkade HJ, Scheenstra R, van der Doef HPJ, Bodewes FAJA, van Rheenen PF. Time-to-reach Target Calprotectin Level in Newly Diagnosed Patients With Inflammatory Bowel Disease. J Pediatr Gastroenterol Nutr 2019;69(4):466-473 View Article PubMed/NCBI
  53. 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(6):894-902 View Article PubMed/NCBI
  54. Kennedy NA, Warner B, Johnston EL, Flanders L, Hendy P, Ding NS, et al. Relapse after withdrawal from anti-TNF therapy for inflammatory bowel disease: an observational study, plus systematic review and meta-analysis. Aliment Pharmacol Ther 2016;43(8):910-923 View Article PubMed/NCBI
  55. Guidi L, Marzo M, Andrisani G, Felice C, Pugliese D, Mocci G, et al. 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(11):974-979 View Article PubMed/NCBI
  56. Magro F, Estevinho MM, Catalano G, Patita M, Arroja B, Lago P, et al; GEDII (Grupo de Estudos da Doença Inflamatória Intestinal). How many biomarker measurements are needed to predict prognosis in Crohn’s disease patients under infliximab?-A prospective study. United European Gastroenterol J 2023;11(6):531-541 View Article PubMed/NCBI
  57. Cao WT, Huang R, Liu S, Fan YH, Xu MS, Xu Y, et al. Infliximab trough level combined with inflammatory biomarkers predict long-term endoscopic outcomes in Crohn’s disease under infliximab therapy. World J Gastroenterol 2022;28(23):2582-2596 View Article PubMed/NCBI
  58. Røer MJ, Småstuen MC, Røseth AG. Usability of IBDoc, a Novel Fecal Calprotectin Home-Based Rapid Test in Clinical Practice. Point of Care: The Journal of Near-Patient Testing & Technology 2019;18(3):85-91 View Article PubMed/NCBI
  59. Haisma SM, Galaurchi A, Almahwzi S, Adekanmi Balogun JA, Muller Kobold AC, van Rheenen PF. Head-to-head comparison of three stool calprotectin tests for home use. PLoS One 2019;14(4):e0214751 View Article PubMed/NCBI
  60. Caccaro R, Angriman I, D’Incà R. Relevance of fecal calprotectin and lactoferrin in the post-operative management of inflammatory bowel diseases. World J Gastrointest Surg 2016;8(3):193-201 View Article PubMed/NCBI
  61. 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(5):368-374 View Article PubMed/NCBI
  62. De Cruz P, Kamm MA, Hamilton AL, Ritchie KJ, Krejany EO, Gorelik A, et al. Crohn’s disease management after intestinal resection: a randomised trial. Lancet 2015;385(9976):1406-1417 View Article PubMed/NCBI
  63. Li T, Shah R, Click B, Cohen BL, Barnes E, Joseph A, et al. American Gastroenterological Association-Proposed Fecal Calprotectin Cutoff of 50 ug/g is Associated With Endoscopic Recurrence in a Real-World Cohort of Patients With Crohn’s Disease Post-ileocolic Resection. Crohns Colitis 360 2024;6(1):otae016 View Article PubMed/NCBI
  64. Furfaro F, D’Amico F, Zilli A, Craviotto V, Aratari A, Bezzio C, et al. Noninvasive Assessment of Postoperative Disease Recurrence in Crohn’s Disease: A Multicenter, Prospective Cohort Study on Behalf of the Italian Group for Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2023;21(12):3143-3151 View Article PubMed/NCBI
  65. González-Chávez SA, Arévalo-Gallegos S, Rascón-Cruz Q. Lactoferrin: structure, function and applications. Int J Antimicrob Agents 2009;33(4):301.e1-e8 View Article PubMed/NCBI
  66. Kruzel ML, Zimecki M, Actor JK. Lactoferrin in a Context of Inflammation-Induced Pathology. Front Immunol 2017;8:1438 View Article PubMed/NCBI
  67. Kirkpatrick CH, Green I, Rich RR, Schade AL. Inhibition of growth of Candida albicans by iron-unsaturated lactoferrin: relation to host-defense mechanisms in chronic mucocutaneous candidiasis. J Infect Dis 1971;124(6):539-544 View Article PubMed/NCBI
  68. León-Sicairos N, López-Soto F, Reyes-López M, Godínez-Vargas D, Ordaz-Pichardo C, de la Garza M. Amoebicidal activity of milk, apo-lactoferrin, sIgA and lysozyme. Clin Med Res 2006;4(2):106-113 View Article PubMed/NCBI
  69. Superti F, Ammendolia MG, Valenti P, Seganti L. Antirotaviral activity of milk proteins: lactoferrin prevents rotavirus infection in the enterocyte-like cell line HT-29. Med Microbiol Immunol 1997;186(2-3):83-91 View Article PubMed/NCBI
  70. Wang WP, Iigo M, Sato J, Sekine K, Adachi I, Tsuda H. Activation of intestinal mucosal immunity in tumor-bearing mice by lactoferrin. Jpn J Cancer Res 2000;91(10):1022-1027 View Article PubMed/NCBI
  71. Legrand D. Lactoferrin, a key molecule in immune and inflammatory processes. Biochem Cell Biol 2012;90(3):252-268 View Article PubMed/NCBI
  72. Zhou XL, Xu W, Tang XX, Luo LS, Tu JF, Zhang CJ, et al. Fecal lactoferrin in discriminating inflammatory bowel disease from irritable bowel syndrome: a diagnostic meta-analysis. BMC Gastroenterol 2014;14:121 View Article PubMed/NCBI
  73. Wang Y, Pei F, Wang X, Sun Z, Hu C, Dou H. Diagnostic accuracy of fecal lactoferrin for inflammatory bowel disease: a meta-analysis. Int J Clin Exp Pathol 2015;8(10):12319-12332 View Article PubMed/NCBI
  74. Sipponen T, Kärkkäinen P, Savilahti E, Kolho KL, Nuutinen H, Turunen U, et al. Correlation of faecal calprotectin and lactoferrin with an endoscopic score for Crohn’s disease and histological findings. Aliment Pharmacol Ther 2008;28(10):1221-1229 View Article PubMed/NCBI
  75. Walker TR, Land ML, Kartashov A, Saslowsky TM, Lyerly DM, Boone JH, et al. Fecal lactoferrin is a sensitive and specific marker of disease activity in children and young adults with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2007;44(4):414-22 View Article PubMed/NCBI
  76. Dai C, Jiang M, Sun M-J, Cao Q. Fecal Lactoferrin for Assessment of Inflammatory Bowel Disease Activity: A Systematic Review and Meta-Analysis. J Clin Gastroenterol 2020;54(6):545-553 View Article PubMed/NCBI
  77. 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(5):1408 View Article PubMed/NCBI
  78. Sipponen T, Savilahti E, Kärkkäinen P, Kolho KL, Nuutinen H, Turunen U, et al. Fecal calprotectin, lactoferrin, and endoscopic disease activity in monitoring anti-TNF-alpha therapy for Crohn’s disease. Inflamm Bowel Dis 2008;14(10):1392-1398 View Article PubMed/NCBI
  79. Chen R, Tie Y, Zhang X, Li L, Chen M, Zhang S. Fecal lactoferrin early predicts long-term outcomes in ulcerative colitis: A post-hoc analysis of the UNIFI and PURSUIT trials. United European Gastroenterol J 2023;11(6):542-550 View Article PubMed/NCBI
  80. Sorrentino D, Nguyen VQ, Love K. Fecal Lactoferrin Predicts Primary Nonresponse to Biologic Agents in Inflammatory Bowel Disease. Dig Dis 2021;39(6):626-633 View Article PubMed/NCBI
  81. Kushner I. The phenomenon of the acute phase response. Ann N Y Acad Sci 1982;389:39-48 View Article PubMed/NCBI
  82. Pathak A, Agrawal A. Evolution of C-Reactive Protein. Front Immunol 2019;10:943 View Article PubMed/NCBI
  83. Kolb-Bachofen V. A review on the biological properties of C-reactive protein. Immunobiology 1991;183(1-2):133-145 View Article PubMed/NCBI
  84. Baumeister D, Akhtar R, Ciufolini S, Pariante CM, Mondelli V. Childhood trauma and adulthood inflammation: a meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-α. Mol Psychiatry 2016;21(5):642-649 View Article PubMed/NCBI
  85. Ford ES, Giles WH, Myers GL, Rifai N, Ridker PM, Mannino DM. C-reactive protein concentration distribution among US children and young adults: findings from the National Health and Nutrition Examination Survey, 1999-2000. Clin Chem 2003;49(8):1353-1357 View Article PubMed/NCBI
  86. Mold C, Nakayama S, Holzer TJ, Gewurz H, Du Clos TW. C-reactive protein is protective against Streptococcus pneumoniae infection in mice. J Exp Med 1981;154(5):1703-1708 View Article PubMed/NCBI
  87. Solmi M, Suresh Sharma M, Osimo EF, Fornaro M, Bortolato B, Croatto G, et al. Peripheral levels of C-reactive protein, tumor necrosis factor-α, interleukin-6, and interleukin-1β across the mood spectrum in bipolar disorder: A meta-analysis of mean differences and variability. Brain Behav Immun 2021;97:193-203 View Article PubMed/NCBI
  88. Du Clos TW. Function of C-reactive protein. Ann Med 2000;32(4):274-278 View Article PubMed/NCBI
  89. Shine B, Berghouse L, Jones JE, Landon J. C-reactive protein as an aid in the differentiation of functional and inflammatory bowel disorders. Clin Chim Acta 1985;148(2):105-109 View Article PubMed/NCBI
  90. Fagan EA, Dyck RF, Maton PN, Hodgson HJ, Chadwick VS, Petrie A, et al. Serum levels of C-reactive protein in Crohn’s disease and ulcerative colitis. Eur J Clin Invest 1982;12(4):351-359 View Article PubMed/NCBI
  91. Dai C, Jiang M, Sun M-J. 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(8):1242-1243 View Article PubMed/NCBI
  92. Sakurai T, Saruta M. Positioning and Usefulness of Biomarkers in Inflammatory Bowel Disease. Digestion 2023;104(1):30-41 View Article PubMed/NCBI
  93. Tilakaratne S, Lemberg DA, Leach ST, Day AS. C-reactive protein and disease activity in children with Crohn’s disease. Dig Dis Sci 2010;55(1):131-136 View Article PubMed/NCBI
  94. Ma C, Battat R, Parker CE, Khanna R, Jairath V, Feagan BG. Update on C-reactive protein and fecal calprotectin: are they accurate measures of disease activity in Crohn’s disease?. Expert Rev Gastroenterol Hepatol 2019;13(4):319-330 View Article PubMed/NCBI
  95. Gibson DJ, Hartery K, Doherty J, Nolan J, Keegan D, Byrne K, et al. CRP/Albumin Ratio: An Early Predictor of Steroid Responsiveness in Acute Severe Ulcerative Colitis. J Clin Gastroenterol 2018;52(6):e48-e52 View Article PubMed/NCBI
  96. Con D, Andrew B, Nicolaides S, van Langenberg DR, Vasudevan A. Biomarker dynamics during infliximab salvage for acute severe ulcerative colitis: C-reactive protein (CRP)-lymphocyte ratio and CRP-albumin ratio are useful in predicting colectomy. Intest Res 2022;20(1):101-113 View Article PubMed/NCBI
  97. Florin THJ, Paterson EWJ, Fowler EV, Radford-Smith GL. Clinically active Crohn’s disease in the presence of a low C-reactive protein. Scand J Gastroenterol 2006;41(3):306-311 View Article PubMed/NCBI
  98. Vermeire S, Van Assche G, Rutgeerts P. Laboratory markers in IBD: useful, magic, or unnecessary toys?. Gut 2006;55(3):426-431 View Article PubMed/NCBI
  99. Peeters M, Joossens S, Vermeire S, Vlietinck R, Bossuyt X, Rutgeerts P. Diagnostic value of anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease. Am J Gastroenterol 2001;96(3):730-734 View Article PubMed/NCBI
  100. Kallenberg CG, Mulder AH, Tervaert JW. Antineutrophil cytoplasmic antibodies: a still-growing class of autoantibodies in inflammatory disorders. Am J Med 1992;93(6):675-682 View Article PubMed/NCBI
  101. van der Woude FJ, Rasmussen N, Lobatto S, et al. Autoantibodies against neutrophils and monocytes: tool for diagnosis and marker of disease activity in Wegener’s granulomatosis. Lancet 1985;1(8426):425-429 View Article PubMed/NCBI
  102. Falk RJ, Jennette JC. Anti-neutrophil cytoplasmic autoantibodies with specificity for myeloperoxidase in patients with systemic vasculitis and idiopathic necrotizing and crescentic glomerulonephritis. N Engl J Med 1988;318(25):1651-1657 View Article PubMed/NCBI
  103. Rump JA, Schölmerich J, Gross V, Roth M, Helfesrieder R, Rautmann A, et al. A new type of perinuclear anti-neutrophil cytoplasmic antibody (p-ANCA) in active ulcerative colitis but not in Crohn’s disease. Immunobiology 1990;181(4-5):406-413 View Article PubMed/NCBI
  104. Ruemmele FM, Targan SR, Levy G, Dubinsky M, Braun J, Seidman EG. Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease. Gastroenterology 1998;115(4):822-829 View Article PubMed/NCBI
  105. Vasiliauskas EA, Plevy SE, Landers CJ, et al. Perinuclear antineutrophil cytoplasmic antibodies in patients with Crohn’s disease define a clinical subgroup. Gastroenterology 1996;110(6):1810-1819 View Article PubMed/NCBI
  106. Sandborn WJ, Feagan BG, Marano C, Zhang H, Strauss R, Johanns J, et al. Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2014;146(1):85-95 View Article PubMed/NCBI
  107. Makharia GK, Sachdev V, Gupta R, Lal S, Pandey RM. Anti-Saccharomyces cerevisiae antibody does not differentiate between Crohn’s disease and intestinal tuberculosis. Dig Dis Sci 2007;52(1):33-39 View Article PubMed/NCBI
  108. McKenzie H, Main J, Pennington CR, Parratt D. Antibody to selected strains of Saccharomyces cerevisiae (baker’s and brewer’s yeast) and Candida albicans in Crohn’s disease. Gut 1990;31(5):536-538 View Article PubMed/NCBI
  109. Lewis JD. The utility of biomarkers in the diagnosis and therapy of inflammatory bowel disease. Gastroenterology 2011;140(6):1817-1826.e2 View Article PubMed/NCBI
  110. Zholudev A, Zurakowski D, Young W, Leichtner A, Bousvaros A. Serologic testing with ANCA, ASCA, and anti-OmpC in children and young adults with Crohn’s disease and ulcerative colitis: diagnostic value and correlation with disease phenotype. Am J Gastroenterol 2004;99(11):2235-2241 View Article PubMed/NCBI
  111. Targan SR, Landers CJ, Yang H, Lodes MJ, Cong Y, Papadakis KA, et al. Antibodies to CBir1 flagellin define a unique response that is associated independently with complicated Crohn’s disease. Gastroenterology 2005;128(7):2020-2028 View Article PubMed/NCBI
  112. Yoshida A, Matsuoka K, Ueno F, Morizane T, Endo Y, Hibi T. Serum PR3-ANCA Is a Predictor of Primary Nonresponse to Anti-TNF-α Agents in Patients with Ulcerative Colitis. Inflamm Intest Dis 2021;6(2):117-122 View Article PubMed/NCBI
  113. Wolf DC, Abraham BP, Afzali A, Allegretti PD, Arai R. Community Perspectives: Combining Serology, Genetics, and Inflammation Markers for the Diagnosis of IBD and Differentiation Between CD and UC. Gastroenterol Hepatol (NY) 2012;8(6 Suppl 2):1-16 View Article PubMed/NCBI
  114. Plevy S, Silverberg MS, Lockton S, Stockfisch T, Croner L, Stachelski J, et al. Combined serological, genetic, and inflammatory markers differentiate non-IBD, Crohn’s disease, and ulcerative colitis patients. Inflamm Bowel Dis 2013;19(6):1139-1148 View Article PubMed/NCBI
  115. Saadeh C. The erythrocyte sedimentation rate: old and new clinical applications. South Med J 1998;91(3):220-225 View Article PubMed/NCBI
  116. Brigden M. The erythrocyte sedimentation rate. Still a helpful test when used judiciously. Postgrad Med 1998;103(5):257-262 View Article PubMed/NCBI
  117. Mak LY, Tong TSM, Cheung KS, Chen LJ, Lui KL, Lau KS, et al. Combined Use of Common Fecal and Blood Markers for Detection of Endoscopically Active Inflammatory Bowel Disease. Clin Transl Gastroenterol 2020;11(3):e00138 View Article PubMed/NCBI
  118. Alper A, Zhang L, Pashankar DS. Correlation of Erythrocyte Sedimentation Rate and C-Reactive Protein With Pediatric Inflammatory Bowel Disease Activity. J Pediatr Gastroenterol Nutr 2017;65(2):e25-e27 View Article PubMed/NCBI
  119. Sands BE. Biomarkers of inflammation in inflammatory bowel disease. Gastroenterology 2015;149(5):1275-1285.e2 View Article PubMed/NCBI
  120. Chinen T, Komai K, Muto G, Morita R, Inoue N, Yoshida H, et al. Prostaglandin E2 and SOCS1 have a role in intestinal immune tolerance. Nat Commun 2011;2:190 View Article PubMed/NCBI
  121. Goodwin JS, Ceuppens J. Regulation of the immune response by prostaglandins. J Clin Immunol 1983;3(4):295-315 View Article PubMed/NCBI
  122. Arai Y, Arihiro S, Matsuura T, Kato T, Matsuoka M, Saruta M, et al. Prostaglandin E-major urinary metabolite as a reliable surrogate marker for mucosal inflammation in ulcerative colitis. Inflamm Bowel Dis 2014;20(7):1208-1216 View Article PubMed/NCBI
  123. Hagiwara SI, Okayasu I, Fujiwara M, Matsuura M, Ohnishi H, Ito S, et al. Prostaglandin E-major Urinary Metabolite as a Biomarker for Pediatric Ulcerative Colitis Activity. J Pediatr Gastroenterol Nutr 2017;64(6):955-961 View Article PubMed/NCBI
  124. Ishida N, Sugiura K, Miyazu T, Tamura S, Suzuki S, Tani S, et al. Prostaglandin E-Major Urinary Metabolite Predicts Relapse in Patients With Ulcerative Colitis in Clinical Remission. Clin Transl Gastroenterol 2020;11(12):e00289 View Article PubMed/NCBI
  125. Rayner BS, Love DT, Hawkins CL. Comparative reactivity of myeloperoxidase-derived oxidants with mammalian cells. Free Radic Biol Med 2014;71:240-255 View Article PubMed/NCBI
  126. Nauseef WM. Myeloperoxidase in human neutrophil host defence. Cell Microbiol 2014;16(8):1146-1155 View Article PubMed/NCBI
  127. Saiki T. Myeloperoxidase concentrations in the stool as a new parameter of inflammatory bowel disease. Kurume Med J 1998;45(1):69-73 View Article PubMed/NCBI
  128. Anezaki K, Asakura H, Honma T, Ishizuka K, Funakoshi K, Tsukada Y, et al. Correlations between interleukin-8, and myeloperoxidase or luminol-dependent chemiluminescence in inflamed mucosa of ulcerative colitis. Intern Med 1998;37(3):253-8 View Article PubMed/NCBI
  129. Sangfelt P. Neutrophil and eosinophil granule proteins as markers of response to local prednisolone treatment in distal ulcerative colitis and proctitis. Am J Gastroenterol 2001;96(4):1085-1090 View Article PubMed/NCBI
  130. Manolakis AC, Kapsoritakis AN, Tiaka EK, Potamianos SP. Calprotectin, calgranulin C, and other members of the s100 protein family in inflammatory bowel disease. Dig Dis Sci 2011;56(6):1601-1611 View Article PubMed/NCBI
  131. Donato R. Intracellular and extracellular roles of S100 proteins. Microsc Res Tech 2003;60(6):540-551 View Article PubMed/NCBI
  132. Foell D, Kucharzik T, Kraft M, Vogl T, Sorg C, Domschke W, et al. Neutrophil derived human S100A12 (EN-RAGE) is strongly expressed during chronic active inflammatory bowel disease. Gut 2003;52(6):847-853 View Article PubMed/NCBI
  133. Cenni S, Casertano M, Trani M, Pacella D, Martinelli M, Staiano A, et al. The use of calgranulin-C (S100A12) and fecal zonulin as possible non-invasive markers in children with inflammatory bowel disease: a clinical study. Eur J Pediatr 2023;182(3):1299-1308 View Article PubMed/NCBI
  134. Jaberi SA, Cohen A, D’Souza C, Abdulrazzaq YM, Ojha S, Bastaki S, et al. Lipocalin-2: Structure, function, distribution and role in metabolic disorders. Biomed Pharmacother 2021;142:112002 View Article PubMed/NCBI
  135. Rashad NM, El-Shal AS, Etewa RL, Wadea FM. Lipocalin-2 expression and serum levels as early predictors of type 2 diabetes mellitus in obese women. IUBMB Life 2017;69(2):88-97 View Article PubMed/NCBI
  136. Stallhofer J, Friedrich M, Konrad-Zerna A, Wetzke M, Lohse P, Glas J, et al. Lipocalin-2 Is a Disease Activity Marker in Inflammatory Bowel Disease Regulated by IL-17A, IL-22, and TNF-α and Modulated by IL23R Genotype Status. Inflamm Bowel Dis 2015;21(10):2327-2340 View Article PubMed/NCBI
  137. Thorsvik S, Damås JK, Granlund AV, Flo TH, Bergh K, Østvik AE, et al. Fecal neutrophil gelatinase-associated lipocalin as a biomarker for inflammatory bowel disease. J Gastroenterol Hepatol 2017;32(1):128-135 View Article PubMed/NCBI
  138. Magro F, Lopes S, Coelho R, Cotter J, Dias de Castro F, Tavares de Sousa H, et al. Accuracy of Faecal Calprotectin and Neutrophil Gelatinase B-associated Lipocalin in Evaluating Subclinical Inflammation in UlceRaTIVE Colitis-the ACERTIVE study. J Crohns Colitis 2017;11(4):435-444 View Article PubMed/NCBI
  139. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463-516 View Article PubMed/NCBI
  140. Chakraborty S, Kaur S, Guha S, Batra SK. The multifaceted roles of neutrophil gelatinase associated lipocalin (NGAL) in inflammation and cancer. Biochim Biophys Acta 2012;1826(1):129-169 View Article PubMed/NCBI
  141. Buisson A, Vazeille E, Minet-Quinard R, Goutte M, Bouvier D, Goutorbe F, et al. Fecal Matrix Metalloprotease-9 and Lipocalin-2 as Biomarkers in Detecting Endoscopic Activity in Patients With Inflammatory Bowel Diseases. J Clin Gastroenterol 2018;52(7):e53-e62 View Article PubMed/NCBI
  142. Wan TM, Iyer DN, Ng L. Roles of microRNAs as non-invasive biomarker and therapeutic target in colorectal cancer. Histol Histopathol 2020;35(3):225-237 View Article PubMed/NCBI
  143. Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007;129(7):1401-1414 View Article PubMed/NCBI
  144. Correia CN, Nalpas NC, McLoughlin KE, Browne JA, Gordon SV, MacHugh DE, et al. Circulating microRNAs as Potential Biomarkers of Infectious Disease. Front Immunol 2017;8:118 View Article PubMed/NCBI
  145. Rashid H, Hossain B, Siddiqua T, Kabir M, Noor Z, Ahmed M, et al. Fecal MicroRNAs as Potential Biomarkers for Screening and Diagnosis of Intestinal Diseases. Front Mol Biosci 2020;7:181 View Article PubMed/NCBI
  146. Schönauen K, Le N, von Arnim U, Schulz C, Malfertheiner P, Link A. Circulating and Fecal microRNAs as Biomarkers for Inflammatory Bowel Diseases. Inflamm Bowel Dis 2018;24(7):1547-1557 View Article PubMed/NCBI
  147. Furfaro F, Ragaini E, Peyrin-Biroulet L, Danese S. Novel Therapies and Approaches to Inflammatory Bowel Disease (IBD). J Clin Med 2022;11(15):4374 View Article PubMed/NCBI
  148. Cuković-Cavka S, Vucelić B, Urek MC, Brinar M, Turk N. Mjesto anti-TNF terapije u lijecenju ulceroznog kolitisa [The role of anti-TNF therapy in ulcerative colitis]. Acta Med Croatica 2013;67(2):171-177 View Article PubMed/NCBI
  149. Avdagić N, Babić N, Šeremet M, Delić-Šarac M, Drače Z, Denjalić A, et al. Tumor necrosis factor-alpha serum level in assessment of disease activity in inflammatory bowel diseases. Med Glas (Zenica) 2013;10(2):211-216 View Article PubMed/NCBI
  150. Komatsu M, Kobayashi D, Saito K, Furuya D, Yagihashi A, Araake H, et al. Tumor necrosis factor-alpha in serum of patients with inflammatory bowel disease as measured by a highly sensitive immuno-PCR. Clin Chem 2001;47(7):1297-1301 View Article PubMed/NCBI
  151. Kucharzik T, Stoll R, Lügering N, Domschke W. Circulating antiinflammatory cytokine IL-10 in patients with inflammatory bowel disease (IBD). Clin Exp Immunol 1995;100(3):452-456 View Article PubMed/NCBI
  152. Mitsuyama K, Tomiyasu N, Takaki K, Masuda J, Yamasaki H, Kuwaki K, et al. Interleukin-10 in the pathophysiology of inflammatory bowel disease: increased serum concentrations during the recovery phase. Mediators Inflamm 2006;2006(6):26875 View Article PubMed/NCBI
  153. Kolios G, Valatas V, Ward SG. Nitric oxide in inflammatory bowel disease: a universal messenger in an unsolved puzzle. Immunology 2004;113(4):427-437 View Article PubMed/NCBI
  154. Korhonen R, Lahti A, Kankaanranta H, Moilanen E. Nitric oxide production and signaling in inflammation. Curr Drug Targets Inflamm Allergy 2005;4(4):471-479 View Article PubMed/NCBI
  155. Reynolds PD, Middleton SJ, Hansford GM, Hunter JO. Confirmation of nitric oxide synthesis in active ulcerative colitis by infra-red diode laser spectroscopy. Eur J Gastroenterol Hepatol 1997;9(5):463-466 View Article PubMed/NCBI
  156. Avdagić N, Zaćiragić A, Babić N, Hukić M, Seremet M, Lepara O, et al. Nitric oxide as a potential biomarker in inflammatory bowel disease. Bosn J Basic Med Sci 2013;13(1):5-9 View Article PubMed/NCBI
  157. Bernsmeier C, Cavazza A, Fatourou EM, Theocharidou E, Akintimehin A, Baumgartner B, et al. Leucocyte ratios are biomarkers of mortality in patients with acute decompensation of cirrhosis and acute-on-chronic liver failure. Aliment Pharmacol Ther 2020;52(5):855-865 View Article PubMed/NCBI
  158. Soufli I, Hablal A, Bessaad S, Amri M, Labsi M, Boussa RS, et al. Nitric Oxide, Neutrophil/Lymphocyte, and Platelet/Lymphocyte Ratios as Promising Inflammatory Biomarkers in Complicated Crohn’s Disease: Outcomes of Corticosteroids and Anti-TNF-α Therapies. Inflammation 2023;46(3):1091-1105 View Article PubMed/NCBI
  159. Wang Y, Li C, Wang W, Wang J, Li J, Qian S, et al. Serum Albumin to Globulin Ratio is Associated with the Presence and Severity of Inflammatory Bowel Disease. J Inflamm Res 2022;15:1907-1920 View Article PubMed/NCBI
  160. Pascual-Figal DA, Januzzi JL. The biology of ST2: the International ST2 Consensus Panel. Am J Cardiol 2015;115(7 Suppl):3B-7B View Article PubMed/NCBI
  161. Boga S, Alkim H, Koksal AR, Ozagari AA, Bayram M, Tekin Neijmann S, et al. Serum ST2 in inflammatory bowel disease: a potential biomarker for disease activity. J Investig Med 2016;64(5):1016-1024 View Article PubMed/NCBI
  162. Díaz-Jiménez D, De la Fuente M, Dubois-Camacho K, Landskron G, Fuentes J, Pérez T, et al. Soluble ST2 is a sensitive clinical marker of ulcerative colitis evolution. BMC Gastroenterol 2016;16(1):103 View Article PubMed/NCBI
  163. Milner CM, Day AJ. TSG-6: a multifunctional protein associated with inflammation. J Cell Sci 2003;116(Pt 10):1863-1873 View Article PubMed/NCBI
  164. Bárdos T, Kamath RV, Mikecz K, Glant TT. Anti-inflammatory and chondroprotective effect of TSG-6 (tumor necrosis factor-alpha-stimulated gene-6) in murine models of experimental arthritis. Am J Pathol 2001;159(5):1711-1721 View Article PubMed/NCBI
  165. Yu Q, Zhang S, Wang H, Zhang Y, Feng T, Chen B, et al. TNFAIP6 is a potential biomarker of disease activity in inflammatory bowel disease. Biomark Med 2016;10(5):473-483 View Article PubMed/NCBI
  166. Duryee MJ, Ahmad R, Eichele DD, Hunter CD, Mitra A, Talmon GA, et al. Identification of Immunoglobulin G Autoantibody Against Malondialdehyde-Acetaldehyde Adducts as a Novel Serological Biomarker for Ulcerative Colitis. Clin Transl Gastroenterol 2022;13(4):e00469 View Article PubMed/NCBI
  167. Mortensen JH, Sinkeviciute D, Manon-Jensen T, Domislović V, McCall K, Thudium CS, et al. A Specific Calprotectin Neo-epitope [CPa9-HNE] in Serum from Inflammatory Bowel Disease Patients Is Associated with Neutrophil Activity and Endoscopic Severity. J Crohns Colitis 2022;16(9):1447-1460 View Article PubMed/NCBI
  168. Jaenisch SE, Abbott CA, Gorrell MD, Bampton P, Butler RN, Yazbeck R. Circulating Dipeptidyl Peptidase Activity Is a Potential Biomarker for Inflammatory Bowel Disease. Clin Transl Gastroenterol 2022;13(1):e00452 View Article PubMed/NCBI
  169. Domislovic V, Høg Mortensen J, Lindholm M, Kaarsdal MA, Brinar M, Barisic A, et al. Inflammatory Biomarkers of Extracellular Matrix Remodeling and Disease Activity in Crohn’s Disease and Ulcerative Colitis. J Clin Med 2022;11(19):5907 View Article PubMed/NCBI
  170. Koivisto L, Bi J, Häkkinen L, Larjava H. Integrin αvβ6: Structure, function and role in health and disease. Int J Biochem Cell Biol 2018;99:186-196 View Article PubMed/NCBI
  171. Kuwada T, Shiokawa M, Kodama Y, Ota S, Kakiuchi N, Nannya Y, et al. Identification of an Anti-Integrin αvβ6 Autoantibody in Patients With Ulcerative Colitis. Gastroenterology 2021;160(7):2383-2394.e21 View Article PubMed/NCBI
  172. Rydell N, Ekoff H, Hellström PM, Movérare R. Measurement of Serum IgG Anti-Integrin αvβ6 Autoantibodies Is a Promising Tool in the Diagnosis of Ulcerative Colitis. J Clin Med 2022;11(7):1881 View Article PubMed/NCBI
  173. Marafini I, Laudisi F, Salvatori S, Lavigna D, Venuto C, Giannarelli D, et al. Diagnostic value of anti-integrin αvβ6 antibodies in ulcerative colitis. Dig Liver Dis 2024;56(1):55-60 View Article PubMed/NCBI
  174. Haberman Y. Tissue-based Gene Expression as Potential Biomarkers for IBD Course. Inflamm Bowel Dis 2020;26(10):1485-1489 View Article PubMed/NCBI
  175. Perez K, Ngollo M, Rabinowitz K, Hammoudi N, Seksik P, Xavier RJ, et al. Meta-Analysis of IBD Gut Samples Gene Expression Identifies Specific Markers of Ileal and Colonic Diseases. Inflamm Bowel Dis 2022;28(5):775-782 View Article PubMed/NCBI
  176. Danese S, Klopocka M, Scherl EJ, Romatowski J, Allegretti JR, Peeva E, et al. Anti-TL1A Antibody PF-06480605 Safety and Efficacy for Ulcerative Colitis: A Phase 2a Single-Arm Study. Clin Gastroenterol Hepatol 2021;19(11):2324-2332.e6 View Article PubMed/NCBI
  177. Abreu MT, Davies JM, Quintero MA, Delmas A, Diaz S, Martinez CD, et al. Transcriptional Behavior of Regulatory T Cells Predicts IBD Patient Responses to Vedolizumab Therapy. Inflamm Bowel Dis 2022;28(12):1800-1812 View Article PubMed/NCBI
  178. Serban ED. Treat-to-target in Crohn’s disease: Will transmural healing become a therapeutic endpoint?. World J Clin Cases 2018;6(12):501-513 View Article PubMed/NCBI
  179. Liu F, Lee SA, Riordan SM, Zhang L, Zhu L. Global Studies of Using Fecal Biomarkers in Predicting Relapse in Inflammatory Bowel Disease. Front Med (Lausanne) 2020;7:580803 View Article PubMed/NCBI
  180. Danese S, Vermeire S, D’Haens G, Panés J, Dignass A, Magro F, et al. Treat to target versus standard of care for patients with Crohn’s disease treated with ustekinumab (STARDUST): an open-label, multicentre, randomised phase 3b trial. Lancet Gastroenterol Hepatol 2022;7(4):294-306 View Article PubMed/NCBI