3,410
Views
1
CrossRef citations to date
0
Altmetric
Review

Current and emerging biomarkers for ulcerative colitis

, &
Pages 1107-1119 | Received 10 Feb 2023, Accepted 01 Nov 2023, Published online: 07 Nov 2023

References

  • Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2017;390(10114):2769–2778.
  • Agrawal M, Christensen HS, Bøgsted M, et al. The rising burden of inflammatory bowel disease in Denmark over two decades: a nationwide cohort study. Gastroenterology. 2022;163(6):1547–1554.e5.
  • Alatab S, Sepanlou SG, Ikuta K, et al. 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.
  • Graham DB, Xavier RJ. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature. 2020;578(7796):527–539. doi: 10.1038/s41586-020-2025-2
  • Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68(Suppl 3):s1–s106.
  • Zilbauer M, Heuschkel R. Disease prognostic biomarkers in inflammatory bowel diseases—A reality check. J Crohns Colitis. 2022;16(1):162–165. doi: 10.1093/ecco-jcc/jjab118
  • Honig G, Heller C, Hurtado-Lorenzo A. Defining the path forward for biomarkers to address unmet needs in inflammatory bowel diseases. Inflamm Bowel Dis. 2020;26(10):1451–1462. doi: 10.1093/ibd/izaa210
  • Honig G. Towards a common validated methodology for histologic measurement of mucosal healing in ulcerative colitis: introducing a new project of the biomarkers consortium of the foundation for the nih. Gastroenterology. 2022;162(3):S35. doi: 10.1053/j.gastro.2021.12.071
  • Ma C, Hanzel J, Panaccione R, et al. CORE-IBD: a multidisciplinary international consensus initiative to develop a core outcome set for randomized controlled trials in inflammatory bowel disease. Gastroenterology. 2022;163(4):950–964.
  • Plevris N, Lees CW. Disease monitoring in inflammatory bowel disease: evolving principles and possibilities. Gastroenterology. 2022;162(5):1456–1475.e1. doi: 10.1053/j.gastro.2022.01.024
  • Jukic A, Bakiri L, Wagner EF, et al. Calprotectin: from biomarker to biological function. Gut. 2021;70(10):1978–1988.
  • Udegbune M, Sharrod–Cole H, Townsend S, et al. Diagnostic performance of serum calprotectin in discriminating active from inactive ulcerative colitis in an outpatient setting. Ann Clin Biochem. 2022;59(6):404–409.
  • Shimizu H, Ebana R, Kudo T, et al. Both fecal calprotectin and fecal immunochemical tests are useful in children with inflammatory bowel disease. J Gastroenterol. 2022;57(5):344–356.
  • Kim ES, Lee HS, Kim SK, et al. Fecal calprotectin is more accurate than fecal immunochemical test for predicting mucosal healing in quiescent ulcerative colitis: a prospective multicenter study. Scand J Gastroenterol. 2020;55(2):163–168.
  • Kawashima K, Oshima N, Kishimoto K, et al. Low fecal calprotectin predicts histological healing in patients with ulcerative colitis with endoscopic remission and leads to prolonged clinical remission. Inflamm Bowel Dis. 2023;29(3):359–366.
  • Turner D, Ruemmele FM, Orlanski-Meyer E, et al. Management of paediatric ulcerative colitis, part 1: ambulatory care—an evidence-based Guideline from European Crohn’s and colitis organization and European Society of paediatric Gastroenterology, hepatology and nutrition. J Pediatr Gastroenterol Nutr. 2018;67(2):257–291.
  • Guardiola J, Lobatón T, Cerrillo E, et al. Recomendaciones del Grupo Español de Trabajo en Enfermedad de Crohn y Colitis Ulcerosa (GETECCU) sobre la utilidad de la determinación de calprotectina fecal en la enfermedad inflamatoria intestinal. Gastroenterol Hepatol. 2018;41(8):514–529.
  • Koninckx CR, Donat E, Benninga MA, et al. The use of fecal calprotectin testing in paediatric disorders: a position paper of the European Society for paediatric Gastroenterology and nutrition gastroenterology committee. J Pediatr Gastroenterol Nutr. 2021;72(4):617–640.
  • Dulai PS, Feagan BG, Sands BE, et al. Prognostic value of fecal calprotectin to inform treat-to-target monitoring in ulcerative colitis. Clin Gastroenterol Hepatol. 2023;21(2):456–466.e7.
  • Kamat N, Vuyyuru SK, Kedia S, et al. Correlation of fecal calprotectin and patient-reported outcome measures in patients with ulcerative colitis. Intest Res. 2022;20(2):269–273.
  • Sakuraba A, Nemoto N, Hibi N, et al. Extent of disease affects the usefulness of fecal biomarkers in ulcerative colitis. BMC Gastroenterol. 2021;21(1):197.
  • Pavlidis P, Joshi D, Sherif YE, et al. Faecal calprotectin is a surrogate marker of biliary inflammation in primary sclerosing cholangitis associated inflammatory bowel disease. Frontline Gastroenterol. 2022;13(6):497–502.
  • Kalla R, Boyapati R, Vatn S, et al. Patients’ perceptions of faecal calprotectin testing in inflammatory bowel disease: results from a prospective multicentre patient-based survey. Scand J Gastroenterol. 2018;53(12):1437–1442.
  • Maréchal C, Aimone-Gastin I, Baumann C, et al. Compliance with the faecal calprotectin test in patients with inflammatory bowel disease. United Eur Gastroenterol J. 2017;5(5):702–707.
  • Lasson A, Stotzer P-O, Öhman L, et al. The intra-individual variability of faecal calprotectin: a prospective study in patients with active ulcerative colitis. J Crohns Colitis. 2015;9:26–32. doi: 10.1016/j.crohns.2014.06.002
  • Kalla R, Kennedy NA, Ventham NT, et al. Serum Calprotectin: a Novel diagnostic and prognostic marker in inflammatory bowel diseases. Am J Gastroenterol. 2016;111(12):1796–1805.
  • Malham M, Carlsen K, Riis L, et al. Plasma calprotectin is superior to serum calprotectin as a biomarker of intestinal inflammation in ulcerative colitis. Scand J Gastroenterol. 2019;54(10):1214–1219.
  • Kessel C, Lavric M, Weinhage T, et al. Serum biomarkers confirming stable remission in inflammatory bowel disease. Sci Rep. 2021;11(1):6690.
  • Mortensen JH, Sinkeviciute D, Manon-Jensen T, 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. doi: 10.1093/ecco-jcc/jjac047
  • Alexdottir MS, Bourgonje AR, Karsdal MA, et al. Serological biomarkers of extracellular matrix turnover and neutrophil activity are associated with long-term use of vedolizumab in patients with Crohn’s disease. Int J Mol Sci. 2022;23(15):8137.
  • Iwańczak B, Ruczka M, Matusiewicz M, et al. Correlation between biomarkers (calprotectin, seromucoid, metalloproteinase-3 and CRP) and clinical and endoscopic activity of ulcerative colitis in children. Adv Med Sci. 2020;65(2):259–264.
  • Yoon JY, Park SJ, Hong SP, et al. Correlations of C-reactive protein levels and erythrocyte sedimentation rates with endoscopic activity indices in patients with ulcerative colitis. Dig Dis Sci. 2014;59(4):829–837.
  • Karoui S, Laz S, Serghini M, et al. Correlation of C-reactive protein with clinical and endoscopic activity in patients with ulcerative colitis. Dig Dis Sci. 2011;56(6):1801–1805.
  • Ishida N, Higuchi T, Miyazu T, et al. C-reactive protein is superior to fecal biomarkers for evaluating colon-wide active inflammation in ulcerative colitis. Sci Rep. 2021;11(1):12431.
  • Voiosu T, Benguş A, Bălănescu P, et al. Rapid fecal calprotectin Testing predicts mucosal healing better than C-reactive protein and serum tumor necrosis factor α in patients with ulcerative colitis. Romanian J Intern Med Rev Roum Med Interne. 2015;53(3):253–260.
  • Schoepfer AM, Beglinger C, Straumann A, et al. Fecal calprotectin more accurately reflects endoscopic activity of ulcerative colitis than the Lichtiger index, C-reactive protein, platelets, hemoglobin, and blood leukocytes. Inflamm Bowel Dis. 2013;19(2):332–341.
  • Deva Rajoo G, Tan L, Lopez A, et al. Early response to corticosteroid and baseline C-Reactive protein predicts outcomes in children with moderate to severe ulcerative colitis. Dig Dis Sci. 2019;64(7):1929–1937.
  • Con D, Andrew B, Nicolaides S, et al. 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.
  • Dubinsky MC, Magro F, Steinwurz F, et al. Association of C-reactive protein and partial Mayo score with response to tofacitinib induction therapy: results from the ulcerative colitis clinical program. Inflamm Bowel Dis. 2022;29(1):51–61.
  • Croft A, Lord A, Radford-Smith G. Markers of systemic inflammation in acute attacks of ulcerative colitis: what level of C-reactive protein constitutes severe colitis? J Crohns Colitis. 2022;16(7):1089–1096. doi: 10.1093/ecco-jcc/jjac014
  • Bakkaloglu OK, Eskazan T, Celik S, et al. Can we predict mucosal remission in ulcerative colitis more precisely with a redefined cutoff level of C-reactive protein? Colorectal Dis Off J Assoc Coloproctology G B Irel. 2022;24(1):77–84.
  • Imakiire S, Takedatsu H, Mitsuyama K, et al. Role of serum proteinase 3 antineutrophil cytoplasmic antibodies in the diagnosis, evaluation of disease severity, and clinical course of ulcerative colitis. Gut Liver. 2022;16(1):92–100.
  • Horn MP, Peter AM, Righini Grunder F, et al. PR3-ANCA and panel diagnostics in pediatric inflammatory bowel disease to distinguish ulcerative colitis from Crohn’s disease. PLoS One. 2018;13:e0208974. doi: 10.1371/journal.pone.0208974
  • Xu Y, Xu F, Li W, et al. The diagnostic role and clinical association of serum proteinase 3 anti-neutrophil cytoplasmic antibodies in Chinese patients with inflammatory bowel disease. Scand J Gastroenterol. 2020;55(7):806–813.
  • Lee W-I, Subramaniam K, Hawkins CA, et al. The significance of ANCA positivity in patients with inflammatory bowel disease. Pathology. 2019;51(6):634–639.
  • Laass MW, Ziesmann J, de Laffolie J, et al. Anti-proteinase 3 antibodies as a biomarker for ulcerative colitis and primary sclerosing cholangitis in children. J Pediatr Gastroenterol Nutr. 2022;74(4):463–470.
  • Rydell N, Ekoff H, Hellström PM, et al. 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.
  • Kuwada T, Shiokawa M, Kodama Y, et al. Identification of an anti–integrin αvβ6 autoantibody in patients with ulcerative colitis. Gastroenterology. 2021;160(7):2383–2394.e21.
  • Livanos AE, Dunn A, Fischer J, et al. Anti-Integrin αvβ6 Autoantibodies Are a Novel Biomarker That Antedate Ulcerative Colitis. Gastroenterology. 2023;164(4):619–629. doi: 10.1053/j.gastro.2022.12.042
  • Swaminathan A, Borichevsky GM, Edwards TS, et al. Faecal myeloperoxidase as a biomarker of endoscopic activity in inflammatory bowel disease. J Crohns Colitis. 2022;16(12):1862–1873.
  • Czub E, Nowak JK, Szaflarska-Poplawska A, et al. Comparison of fecal pyruvate kinase isoform M2 and calprotectin in assessment of pediatric inflammatory bowel disease severity and activity. Acta Biochim Pol. 2014;61(1):99–102.
  • Amcoff K, Cao Y, Zhulina Y, et al. Prognostic significance of faecal eosinophil granule proteins in inflammatory bowel disease. Scand J Gastroenterol. 2019;54(10):1237–1244.
  • Abedin N, Seemann T, Kleinfeld S, et al. Fecal eosinophil cationic protein is a diagnostic and predictive biomarker in young adults with inflammatory bowel disease. J Clin Med. 2019;8(12):2025.
  • Trasolini R, Zhu K, Klemm N, et al. Fecal leukocyte esterase, an alternative biomarker to fecal calprotectin in inflammatory bowel disease: a Pilot series. Gastro Hep Adv. 2022;1(1):45–51.
  • Shinzaki S, Matsuoka K, Iijima H, et al. Leucine-rich alpha-2 glycoprotein is a serum biomarker of mucosal healing in ulcerative colitis. J Crohns Colitis. 2017;11(1):84–91.
  • Nakov R, Velikova T, Nakov V, et al. Trefoil factor 3 is highly predictive of complete mucosal healing independently and in combination with C-Reactive protein in patients with ulcerative colitis. J Gastrointest Liver Dis JGLD. 2019;28:169–174. doi: 10.15403/jgld-177
  • Wakai M, Hayashi R, Tanaka S, et al. Serum amyloid a is a better predictive biomarker of mucosal healing than C-reactive protein in ulcerative colitis in clinical remission. BMC Gastroenterol. 2020;20(1):85.
  • Jaenisch SE, Abbott CA, Gorrell MD, et al. Circulating Dipeptidyl peptidase activity is a potential biomarker for inflammatory bowel disease. Clin Transl Gastroenterol. 2022;13(1):e00452.
  • Pinto-Lopes P, Afonso J, Pinto-Lopes R, et al. Serum Dipeptidyl peptidase 4: a predictor of disease activity and prognosis in inflammatory bowel disease. Inflamm Bowel Dis. 2020;26(11):1707–1719.
  • Maeda K, Nakamura M, Yamamura T, et al. Gelsolin as a potential biomarker for endoscopic activity and mucosal healing in ulcerative colitis. Biomedicines. 2022;10(4):872.
  • Shiraishi K, Furukawa S, Yagi S, et al. Serum globulin is associated with endoscopic findings and mucosal healing in Japanese patients with ulcerative colitis. Dig Dis Sci. 2022;67(1):233–240.
  • Verstockt S, Verstockt B, Machiels K, et al. Oncostatin M is a biomarker of diagnosis, worse disease prognosis, and therapeutic nonresponse in inflammatory bowel disease. Inflamm Bowel Dis. 2021;27(10):1564–1575. doi: 10.1093/ibd/izab032
  • Guo A, Ross C, Chande N, et al. High oncostatin M predicts lack of clinical remission for patients with inflammatory bowel disease on tumor necrosis factor α antagonists. Sci Rep. 2022;12(1):1185.
  • West NR, Hegazy AN, Owens BMJ, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor–neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017;23(5):579–589.
  • Nishida Y, Hosomi S, Watanabe K, et al. Serum interleukin-6 level is associated with response to infliximab in ulcerative colitis. Scand J Gastroenterol. 2018;53(5):579–585.
  • Bertani L, Rossari F, Barberio B, et al. Novel prognostic biomarkers of mucosal healing in ulcerative colitis patients treated with anti-TNF: neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio. Inflamm Bowel Dis. 2020;26(10):1579–1587.
  • Mortensen JH, Godskesen LE, Jensen MD, et al. Fragments of citrullinated and MMP-degraded vimentin and MMP-degraded type III collagen are Novel serological biomarkers to differentiate Crohn’s disease from ulcerative colitis. J Crohns Colitis. 2015;9(10):863–872.
  • Domislovic V, Høg Mortensen J, Lindholm M, 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.
  • Kalla R, Adams AT, Bergemalm D, et al. Serum proteomic profiling at diagnosis predicts clinical course, and need for intensification of treatment in inflammatory bowel disease. J Crohns Colitis. 2021;15(5):699–708.
  • Sudhakar P, Salomon B, Verstockt B, et al. DOP79 biomarkers for IBD using OLINK proteomics inflammation panel: preliminary results from the COLLIBRI consortium. J Crohns Colitis. 2022;16(Supplement_1):i123–i124.
  • Bergemalm D, Andersson E, Hultdin J, et al. Systemic Inflammation in Preclinical Ulcerative Colitis. Gastroenterology. 2021;161(5):1526–1539.e9.
  • Torres J, Petralia F, Sato T, et al. Serum biomarkers identify patients who will develop inflammatory bowel diseases up to 5 years before diagnosis. Gastroenterology. 2020;159(1):96–104.
  • Bourgonje AR, von Martels JZH, Gabriëls RY, et al. A combined set of four serum inflammatory biomarkers reliably predicts endoscopic disease activity in inflammatory bowel disease. Front Med. 2019;6:251. doi: 10.3389/fmed.2019.00251
  • Baldan-Martin M, Azkargorta M, Iloro I, et al. P018 proteomic profile of serum and urine in newly diagnosed patients with inflammatory bowel disease: new approach for biomarker discovery. J Crohns Colitis. 2022;16(Supplement_1):i145.
  • Andersson E, Bergemalm D, Kruse R, et al. Subphenotypes of inflammatory bowel disease are characterized by specific serum protein profiles. PLoS One. 2017;12(10):e0186142.
  • Heier CR, Fiorillo AA, Chaisson E, et al. Identification of pathway-specific serum biomarkers of response to glucocorticoid and infliximab treatment in children with inflammatory bowel disease. Clin Transl Gastroenterol. 2016;7(9):e192.
  • Adams A, Gupta V, Mohsen W, et al. Early management of acute severe UC in the biologics era: development and international validation of a prognostic clinical index to predict steroid response. Gut. 2023;72(3):433–442.
  • Grant RK, Jones G-R, Plevris N, et al. The ACE (albumin, CRP and endoscopy) index in acute colitis: a simple clinical index on admission that predicts outcome in patients with acute ulcerative colitis. Inflamm Bowel Dis. 2021;27(4):451–457.
  • Gibson DJ, Hartery K, Doherty J, et al. Crp/albumin ratio: an early predictor of steroid responsiveness in acute severe ulcerative colitis. J Clin Gastroenterol. 2018;52(6):e48–e52.
  • Header DA, Aboelwafa RA, Elkeleny MR, et al. C-reactive protein/albumin ratio (CAR) as a marker for detecting acute severe ulcerative colitis in Egyptian patients. Rev Gastroenterol Mex Engl. 2022;87(4):447–454.
  • Yagi S, Furukawa S, Shiraishi K, et al. The albumin to globulin ratio is associated with clinical outcome in Japanese patients with ulcerative colitis. Ann Coloproctology. 2023;39(2):155–163.
  • Kalla R, Adams AT, Nowak JK, et al. Analysis of systemic epigenetic alterations in inflammatory bowel disease: defining geographical, genetic and immune-inflammatory influences on the circulating methylome. J Crohns Colitis. 2023;17(2):170–184.
  • Gasparetto M, Payne F, Nayak K, et al. Transcription and DNA methylation patterns of blood-derived CD8+ T cells are associated with age and inflammatory bowel disease but do not predict prognosis. Gastroenterology. 2020;160(1):232–244.e7.
  • Adams AT, Kennedy NA, Hansen R, et al. Two-stage genome-wide methylation profiling in childhood-onset Crohnʼs disease implicates epigenetic alterations at the VMP1/MIR21 and HLA loci. Inflamm Bowel Dis. 2014;20(10):1784–1793.
  • Kalla R, Adams AT, Satsangi J. Blood-based DNA methylation in Crohn’s disease and severity of intestinal inflammation. Transl Gastroenterol Hepatol. 2019;4:76. doi: 10.21037/tgh.2019.10.03
  • Ventham NT, Kennedy NA, Adams AT, et al. Integrative epigenome-wide analysis demonstrates that DNA methylation may mediate genetic risk in inflammatory bowel disease. Nat Commun. 2016;7(1):13507.
  • Joustra V, Hageman IL, Satsangi J, et al. Systematic review and meta-analysis of peripheral blood DNA methylation studies in inflammatory bowel disease. J Crohns Colitis. 2023;17(2):185–198.
  • Venkateswaran S, Somineni HK, Matthews JD, et al. Longitudinal DNA methylation profiling of the rectal mucosa identifies cell-specific signatures of disease status, severity and clinical outcomes in ulcerative colitis cell-specific DNA methylation signatures of UC. Clin Epigenetics. 2023;15(1):50.
  • Burakoff R, Chao S, Perencevich M, et al. Blood-based biomarkers can differentiate ulcerative colitis from crohnʼs disease and noninflammatory diarrhea. Inflamm Bowel Dis. 2011;17(8):1719–1725.
  • Biasci D, Lee JC, Noor NM, et al. A blood-based prognostic biomarker in IBD. Gut. 2019;68(8):1386–1395.
  • Nowak JK, Adams AT, Kalla R, et al. Characterization of the circulating transcriptomic landscape in inflammatory bowel disease provides evidence for dysregulation of multiple transcription factors including NFE2, SPI1, CEBPB, and IRF2. J Crohns Colitis. 2022;16(8):1255–1268.
  • Kalla R, Adams AT, Ventham NT, et al. Whole blood profiling of T-cell derived miRNA allows the development of prognostic models in inflammatory bowel disease. J Crohns Colitis. 2020;14(12):1724–1733.
  • Yarani R, Shojaeian A, Palasca O, et al. Differentially expressed miRnas in ulcerative colitis and Crohn’s disease. Front Immunol. 2022;13:865777. doi: 10.3389/fimmu.2022.865777
  • Zhou J, Liu J, Gao Y, et al. miRNA-Based potential biomarkers and new molecular insights in ulcerative colitis. Front Pharmacol. 2021 [cited 2023 Aug 13]; 12. Internet: 10.3389/fphar.2021.707776
  • Vatn SS, Lindstrøm JC, Moen AEF, et al. Mucosal gene transcript signatures in treatment naïve inflammatory bowel disease: a comparative analysis of disease to symptomatic and healthy controls in the European IBD-Character cohort. Clin Exp Gastroenterol. 2022;15:5–25. doi: 10.2147/CEG.S343468
  • Ostrowski J, Dabrowska M, Lazowska I, et al. Redefining the practical utility of blood transcriptome biomarkers in inflammatory bowel diseases. J Crohns Colitis. 2019;13(5):626–633.
  • Wu Z, Bi Y. Potential role of MALT1 as a candidate biomarker of disease surveillance and treatment response prediction in inflammatory bowel disease patients. J Clin Lab Anal. 2022;36(2):e24130. doi: 10.1002/jcla.24130
  • Mo A, Nagpal S, Gettler K, et al. Stratification of risk of progression to colectomy in ulcerative colitis via measured and predicted gene expression. Am J Hum Genet. 2021;108(9):1765–1779.
  • Clerc F, Novokmet M, Dotz V, et al. Plasma N-Glycan signatures are associated with features of inflammatory bowel diseases. Gastroenterology. 2018;155(3):829–843.
  • Shubhakar A, Jansen B, Adams A, et al. DOP10 serum N-glycomic biomarkers predict treatment escalation in inflammatory bowel disease. J Crohns Colitis. 2019;13(Supplement_1):S032–S033.
  • Shubhakar A, Jansen BC, Adams AT, et al. Serum N-Glycomic biomarkers predict treatment escalation in inflammatory bowel disease. J Crohns Colitis. 2023;17(6):919–932.
  • Pereira MS, Maia L, Azevedo LF, et al. A [Glyco]biomarker that predicts failure to Standard therapy in ulcerative colitis patients. J Crohns Colitis. 2019;13(1):39–49.
  • Probert F, Walsh A, Jagielowicz M, et al. Plasma nuclear magnetic resonance Metabolomics discriminates between high and low endoscopic activity and predicts progression in a prospective cohort of patients with ulcerative colitis. J Crohns Colitis. 2018;12(11):1326–1337.
  • Ghimire H, Viennois E, Hu X, et al. Infrared spectrometric biomarkers for ulcerative colitis screening using human serum samples. J Biophoto. 2022;15(6):e202100307.
  • Berry N, Sinha SK, Bhattacharya A, et al. Role of positron emission tomography in assessing disease activity in ulcerative colitis: comparison with biomarkers. Dig Dis Sci. 2018;63(6):1541–1550.
  • Borren NZ, Plichta D, Joshi AD, et al. Multi-“-omics” profiling in patients with quiescent inflammatory bowel disease identifies biomarkers predicting relapse. Inflamm Bowel Dis. 2020;26(10):1524–1532.
  • Integrated bile acid profile analysis for the non-invasive staging diagnosis of ulcerative colitis. medRxiv [Internet]. [cited 2022 Nov 8]. Available from: https://www.medrxiv.org/content/10.1101/2022.03.14.22272391v1
  • Sun Q, Tang Y, Dai L, et al. Serum bile acid metabolites predict the therapeutic effect of Mesalazine in patients with ulcerative colitis. J Proteome Res. 2023;22(4):1287–1297.
  • Jagt JZ, Struys EA, Ayada I, et al. Fecal amino acid analysis in newly diagnosed pediatric inflammatory bowel disease: a multicenter case-control study. Inflamm Bowel Dis. 2022;28(5):755–763.
  • Zhou G, Liu H, Wei P, et al. Amino acids-targeted metabolomics reveals novel diagnostic biomarkers for ulcerative colitis and Crohn’s disease. Amino Acids. 2023;55(3):349–358.
  • Liu Z, Tang H, Liang H, et al. Dyslipidaemia is associated with severe disease activity and poor prognosis in ulcerative colitis: a retrospective cohort study in China. Nutrients. 2022;14(15):3040.
  • Manfredi M, Conte E, Barberis E, et al. Integrated serum proteins and fatty acids analysis for putative biomarker discovery in inflammatory bowel disease. J Proteomics. 2019;195:138–149. doi: 10.1016/j.jprot.2018.10.017
  • Galipeau HJ, Caminero A, Turpin W, et al. Novel fecal biomarkers that precede clinical diagnosis of ulcerative colitis. Gastroenterology. 2021;160(5):1532–1545.
  • Iwasawa K, Suda W, Tsunoda T, et al. Dysbiosis of the salivary microbiota in pediatric-onset primary sclerosing cholangitis and its potential as a biomarker. Sci Rep. 2018;8(1):5480.
  • Olbjørn C, Cvancarova Småstuen M, Thiis-Evensen E, et al. Fecal microbiota profiles in treatment-naïve pediatric inflammatory bowel disease – associations with disease phenotype, treatment, and outcome. Clin Exp Gastroenterol. 2019;12:37–49. doi: 10.2147/CEG.S186235
  • Olbjørn C, Småstuen MC, Moen AEF. Targeted analysis of the Gut microbiome for diagnosis, prognosis and treatment individualization in pediatric inflammatory bowel disease. Microorganisms. 2022;10(7):1273. doi: 10.3390/microorganisms10071273
  • Vatn S, Carstens A, Kristoffersen AB, et al. Faecal microbiota signatures of IBD and their relation to diagnosis, disease phenotype, inflammation, treatment escalation and anti-TNF response in a European multicentre study (IBD-Character). Scand J Gastroenterol. 2020;55(10):1146–1156.
  • Liu J, Fang H, Hong N, et al. Gut microbiome and metabonomic profile predict early remission to anti-integrin therapy in patients with moderate to severe ulcerative colitis. Microbiol Spectr. 2023;11(3):e0145723.
  • Rees NP, Shaheen W, Quince C, et al. Systematic review of donor and recipient predictive biomarkers of response to faecal microbiota transplantation in patients with ulcerative colitis. EBioMedicine. 2022;81:104088. doi: 10.1016/j.ebiom.2022.104088
  • van Thiel IAM, Rahman S, Hakvoort TBM, et al. Fecal Filobasidium is associated with clinical remission and endoscopic response following fecal microbiota transplantation in mild-to-moderate ulcerative colitis. Microorganisms. 2022;10(4):737.
  • Nishida Y, Hosomi S, Yamagami H, et al. Novel prognostic biomarkers of pouchitis after ileal pouch-anal anastomosis for ulcerative colitis: neutrophil-to-lymphocyte ratio. PLoS One. 2020;15(10):e0241322.
  • Okba AM, Amin MM, Abdelmoaty AS, et al. Neutrophil/Lymphocyte ratio and lymphocyte/monocyte ratio in ulcerative colitis as non-invasive biomarkers of disease activity and severity. Autoimmun Highlights. 2019;10(1):4.
  • Kurimoto N, Nishida Y, Hosomi S, et al. Neutrophil-to-lymphocyte ratio may predict clinical relapse in ulcerative colitis patients with mucosal healing. PLoS One. 2023;18(1):e0280252.
  • Anderson A, Cherfane C, Click B, et al. Monocytosis is a biomarker of severity in inflammatory bowel disease: analysis of a 6-year prospective natural history registry. Inflamm Bowel Dis. 2022;28(1):70–78.
  • Gettler K, Levantovsky R, Moscati A, et al. Common and rare variant prediction and penetrance of IBD in a large, multi-ethnic Health System-Based Biobank Cohort. Gastroenterol. 2021;160(5):1546–1557.
  • Kikut J, Mokrzycka M, Drozd A, et al. Involvement of proinflammatory arachidonic acid (ARA) derivatives in Crohn’s disease (CD) and ulcerative colitis (UC). J Clin Med. 2022;11(7):1861.
  • Debourdeau E, Chamard C, Carriere I, et al. Retinal microcirculation changes in Crohn’s disease patients under biologics, a potential biomarker of severity: a Pilot study. J Pers Med. 2022;12(2):230.
  • Zittan E, Steinhart AH, Aran H, et al. The Toronto IBD Global endoscopic Reporting [TIGER] score: a single, easy to use endoscopic score for both Crohn’s disease and ulcerative colitis patients. J Crohns Colitis. 2022;16(4):544–553.
  • Rowan CR, Cullen G, Mulcahy HE, et al. DUBLIN [degree of ulcerative colitis burden of Luminal inflammation] score, a simple method to quantify inflammatory burden in ulcerative colitis. J Crohns Colitis. 2019;13(11):1365–1371.
  • Gordon IO, Agrawal N, Willis E, et al. Fibrosis in ulcerative colitis is directly linked to severity and chronicity of mucosal inflammation. Aliment Pharmacol Ther. 2018;47(7):922–939.
  • Mortensen JH, Lindholm M, Langholm LL, et al. The intestinal tissue homeostasis – the role of extracellular matrix remodeling in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2019;13(10):977–993.
  • Boyapati RK, Dorward DA, Tamborska A, et al. Mitochondrial DNA is a pro-inflammatory Damage-associated molecular Pattern Released During active IBD. Inflamm Bowel Dis. 2018;24(10):2113–2122.
  • Mitochondrial DAMPs as mechanistic biomarkers of mucosal inflammation in Crohn’s disease: study protocol for prospective longitudinal cohort study in Scotland. medRxiv [Internet]. [cited 2022 Nov 8]. Available from: https://www.medrxiv.org/content/10.1101/2022.03.21.22270313v1
  • Argmann C, Hou R, Ungaro RC, et al. Biopsy and blood-based molecular biomarker of inflammation in IBD. Gut [Internet]. 2022 [cited 2023 Jan 17]. Available from: https://gut.bmj.com/content/early/2022/09/14/gutjnl-2021-326451
  • Khanna R, Narula N, Feagan BG. The role of biomarkers in clinical trials of inflammatory bowel disease. Inflamm Bowel Dis. 2018;24(8):1619–1623. doi: 10.1093/ibd/izy195
  • Gabriëls RY, Bourgonje AR, von Martels J, et al. P577 Mucosal eosinophil abundance in non-inflamed colonic tissue predict response to vedolizumab induction therapy in inflammatory bowel disease. J Crohns Colitis. 2022;16(Supplement_1):i517–i518.
  • Roosenboom B, Wahab PJ, Smids C, et al. Mucosal α4β7+ Lymphocytes and MAdCAM+ Venules Predict Response to Vedolizumab in Ulcerative Colitis. Inflamm Bowel Dis. 2023;zad123. doi: 10.1093/ibd/izad123
  • Telesco SE, Brodmerkel C, Zhang H, et al. Gene expression signature for prediction of Golimumab response in a phase 2a open-label trial of patients with ulcerative colitis. Gastroenterology. 2018;155(4):1008–1011.e8.
  • Scheer J. Gould and Goldman: deadly deceit--a defense. Health Phys. 1991;61(2):279–280.