1
|
Honap S, Jairath V, Sands BE, Dulai PS, Higgins PDR, De Cruz P, Gutiérrez A, Kotze PG, Ye BD, Kobayashi T, Gearry RB, Olivera PA, Amiot A, Mosli MH, Al Awadhi S, Halfvarson J, Patel KV, Sebastian S, Danese S, Peyrin-Biroulet L. Acute Severe Ulcerative Colitis: An International Delphi Consensus on Clinical Trial Design and Endpoints. Clin Gastroenterol Hepatol 2024:S1542-3565(24)01082-6. [PMID: 39681225 DOI: 10.1016/j.cgh.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND & AIMS Interventional clinical trials in acute severe ulcerative colitis (ASUC) are characterized by substantial heterogeneity due to a lack of consensus in several key areas of trial design-this impedes clinical research efforts to identify novel therapies. The objective of this initiative was to achieve the first consensus and provide clear position statements on ASUC trial design. METHODS A modified Delphi consensus approach was employed with a panel of 20 clinicians with international representation and expertise in ASUC trial design and delivery. Agreement was defined as at least 75% of participants voting as "agree" with each statement. RESULTS In total, 30 statements achieved consensus and were approved. Statements centred on proposing suitable eligibility criteria (disease extent, disease severity, prior therapy exposure), optimizing trial design (randomization, stratification, corticosteroid handling, timing of assessments), and recommending primary and secondary endpoints alongside defining key efficacy outcomes (clinical and endoscopic response and remission, treatment failure, quality of life). CONCLUSIONS The expansion of drugs to treat moderate-severe ulcerative colitis over the past decade, particularly the rapidly acting Janus kinase inhibitors, is promising and has reignited the interest in identifying suitable therapeutic candidates for ASUC. Clinical trials in this high-risk population are challenging to conduct and this consensus provides a framework for future trials to advance drug development.
Collapse
Affiliation(s)
- Sailish Honap
- Department of Gastroenterology, INFINY Institute, CHRU Nancy, Nancy, France; Department of Gastroenterology, St George's University Hospitals NHS Foundation Trust, London, United Kingdom; School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.
| | - Vipul Jairath
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University, London, Ontario, Canada; Lawson Health Research Institute, Western University, London, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Bruce E Sands
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Parambir S Dulai
- Division of Gastroenterology, Northwestern University, Chicago, Illinois
| | - Peter D R Higgins
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan
| | - Peter De Cruz
- Department of Gastroenterology, Austin Health, Melbourne, Victoria, Australia; Department of Medicine (Austin Health), University of Melbourne, Melbourne, Victoria, Australia
| | - Ana Gutiérrez
- Department of Gastroenterology, Hospital General Universitario Dr Balmis de Alicante, Alicante, Spain; Instituto de Instituto de Investigación Sanitaria y Biomédica de Alicante, Alicante, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Paulo G Kotze
- Colorectal Surgery Unit, Pontificia Universidade Católica do Paraná, Curitiba, Brazil
| | - Byong Duk Ye
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea; Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Richard B Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand; Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| | - Pablo A Olivera
- IBD Unit, Gastroenterology Section, Department of Internal Medicine, Centro de Educación Médica e Investigación Clínica, Buenos Aires, Argentina; Zane Cohen Centre for Digestive Diseases, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Aurélien Amiot
- Department of Gastroenterology, Hôpitaux Universitaires Bicêtre, Assistance Publique-Hôpitaux de Paris, Université Paris Saclay, Le Kremlin Bicêtre, France; Centre for Epidemiology and Population Health, INSERM, Université Paris Saclay, Villejuif, France
| | - Mahmoud H Mosli
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sameer Al Awadhi
- Digestive Diseases Unit, Rashid Hospital, Dubai, United Arab Emirates
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Kamal V Patel
- Department of Gastroenterology, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Shaji Sebastian
- IBD Unit, Department of Gastroenterology, Hull University Teaching Hospitals, Hull, United Kingdom
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, INFINY Institute, CHRU Nancy, INSERM NGERE, University of Lorraine, Nancy, France
| |
Collapse
|
2
|
Costa MHDM, Sassaki LY, Chebli JMF. Fecal calprotectin and endoscopic scores: The cornerstones in clinical practice for evaluating mucosal healing in inflammatory bowel disease. World J Gastroenterol 2024; 30:3022-3035. [PMID: 38983953 PMCID: PMC11230062 DOI: 10.3748/wjg.v30.i24.3022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/01/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
Managing inflammatory bowel disease (IBD) is becoming increasingly complex and personalized, considering the advent of new advanced therapies with distinct mechanisms of action. Achieving mucosal healing (MH) is a pivotal therapeutic goal in IBD management and can prevent IBD progression and reduce flares, hospitalization, surgery, intestinal damage, and colorectal cancer. Employing proactive disease and therapy assessment is essential to achieve better control of intestinal inflammation, even if subclinical, to alter the natural course of IBD. Periodic monitoring of fecal calprotectin (FC) levels and interval endoscopic evaluations are cornerstones for evaluating response/remission to advanced therapies targeting IBD, assessing MH, and detecting subclinical recurrence. Here, we comment on the article by Ishida et al Moreover, this editorial aimed to review the role of FC and endoscopic scores in predicting MH in patients with IBD. Furthermore, we intend to present some evidence on the role of these markers in future targets, such as histological and transmural healing. Additional prospective multicenter studies with a stricter MH criterion, standardized endoscopic and histopathological analyses, and virtual chromoscopy, potentially including artificial intelligence and other biomarkers, are desired.
Collapse
Affiliation(s)
| | - Ligia Yukie Sassaki
- Department of Internal Medicine, Medical School, São Paulo State University (Unesp), Botucatu 18618-686, São Paulo, Brazil
| | - Júlio Maria Fonseca Chebli
- Division of Gastroenterology, Department of Medicine, University Hospital of The Federal University of Juiz de Fora, University of Juiz de Fora School of Medicine, Juiz de Fora 36036-247, Minas Gerais, Brazil
| |
Collapse
|
3
|
Yang MM, Usiskin K, Ahmad HA, Ather S, Sreih A, Canavan JB, Farraye FA, Ma C. Considerations for Colorectal Neoplasia Detection in Inflammatory Bowel Disease Clinical Trials. Dig Dis 2023; 42:12-24. [PMID: 37757769 PMCID: PMC10836758 DOI: 10.1159/000533395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/25/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND High-quality colonoscopic surveillance can lead to earlier and increased detection of colorectal neoplasia in patients with inflammatory bowel disease (IBD). In IBD clinical trials, endoscopy is used to assess mucosal disease activity before and after treatment but also provides an opportunity to surveil for colorectal neoplasia during follow-up. SUMMARY Best practices for colorectal cancer identification in IBD clinical trials require engagement and collaboration between the clinical trial sponsor, site endoscopist and/or principal investigator, and central read team. Each team member has unique responsibilities for maximizing dysplasia detection in IBD trials. KEY MESSAGES Sponsors should work in accordance with scientific guidelines to standardize imaging procedures, design the protocol to ensure the trial population is safeguarded, and oversee trial conduct. The site endoscopist should remain updated on best practices to tailor sponsor protocol-required procedures to patient needs, examine the mucosa for disease activity and potential dysplasia during all procedures, and provide optimal procedure videos for central read analysis. Central readers may detect dysplasia or colorectal cancer and a framework to report these findings to trial sponsors is essential. Synergistic relationships between all team members in IBD clinical trials provide an important opportunity for extended endoscopic evaluation and colorectal neoplasia identification.
Collapse
Affiliation(s)
- Mira M Yang
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Keith Usiskin
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Harris A Ahmad
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Shabana Ather
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Antoine Sreih
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - James B Canavan
- Division of Immunology and Fibrosis Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Francis A Farraye
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, Jacksonville, Florida, USA
| | - Christopher Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, and Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Medical Research and Development, Alimentiv Inc (formerly Robarts Clinical Trials, Inc.), London, Ontario, Canada
| |
Collapse
|
4
|
Sandborn WJ, Panés J, Danese S, Sharafali Z, Hassanali A, Jacob-Moffatt R, Eden C, Daperno M, Valentine JF, Laharie D, Baía C, Atreya R, Panaccione R, Rydzewska G, Aguilar H, Vermeire S. Etrolizumab as induction and maintenance therapy in patients with moderately to severely active Crohn's disease (BERGAMOT): a randomised, placebo-controlled, double-blind, phase 3 trial. Lancet Gastroenterol Hepatol 2023; 8:43-55. [PMID: 36240801 DOI: 10.1016/s2468-1253(22)00303-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Etrolizumab is a gut-targeted anti-β7 monoclonal antibody targeting α4β7 and αEβ7 integrins. We aimed to compare the safety and efficacy of two doses of etrolizumab with placebo in patients with Crohn's disease. METHODS BERGAMOT was a randomised, placebo-controlled, double-blind, phase 3 study done at 326 treatment centres worldwide. We included patients aged 18-80 years with moderately to severely active Crohn's disease (Crohn's Disease Activity Index [CDAI] score of 220-480, and a mean daily stool frequency score of ≥6 or a mean daily stool frequency score of >3, and a mean daily abdominal pain score of >1, as well as the presence of active inflammation on screening ileocolonoscopy) who had intolerance, inadequate response, or no response to one or more of corticosteroids, immunosuppressants, or anti-TNF therapy within the past 5 years. BERGAMOT consisted of three induction cohorts (a placebo-controlled, double-blind exploratory cohort [cohort 1]; an active treatment cohort not containing a placebo control [cohort 2]; and a placebo-controlled, double-blind pivotal cohort [cohort 3]) and one maintenance cohort. In induction cohort 3, during the 14-week induction, patients were randomly assigned (2:3:3) to receive matched placebo, 105 mg etrolizumab subcutaneously every 4 weeks (at weeks 0, 4, 8, and 12) or 210 mg etrolizumab subcutaneously (at weeks 0, 2, 4, 8, and 12), stratified by concomitant treatment with oral corticosteroids, concomitant treatment with immunosuppressants, baseline disease activity, and previous exposure to anti-TNF therapy. To preserve masking, all patients received two injections at weeks 0, 4, 8, and 12 and one injection at week 2. Week 14 etrolizumab responders from all cohorts were re-randomly assigned (1:1) to receive 105 mg etrolizumab (etrolizumab maintenance group) or placebo (placebo maintenance group) every 4 weeks for 52 weeks; patients in the induction placebo group underwent a sham re-randomisation to preserve masking. During maintenance, randomisation was stratified by CDAI remission status, concomitant treatment with oral corticosteroids, induction dose regimen, and previous exposure to anti-TNF therapy. All participants and study site personnel were masked to treatment assignment for both induction and maintenance. Co-primary induction endpoints at week 14 (placebo vs 210 mg etrolizumab) were clinical remission (mean stool frequency ≤3 and mean abdominal pain ≤1, with no worsening) and endoscopic improvement (≥50% reduction in Simple Endoscopic Score for Crohn's Disease [SES-CD]). Co-primary maintenance endpoints at week 66 (placebo vs etrolizumab) were clinical remission and endoscopic improvement. Efficacy was analysed using a modified intention-to-treat (mITT) population, defined as all randomised patients who received at least one dose of study drug (induction) and as all patients re-randomised into maintenance who received at least one dose of study drug in the maintenance phase (maintenance). Safety analyses included all patients who received at least one dose of study drug. Maintenance safety analyses include all adverse events occurring in both induction and maintenance. This trial is registered with ClinicalTrials.gov, NCT02394028, and is closed to recruitment. FINDINGS Between March 20, 2015, and Sept 7, 2021, 385 patients (209 [54%] male and 326 [85%] white) were randomly assigned in induction cohort 3 to receive placebo (n=97), 105 mg etrolizumab (n=143), or 210 mg etrolizumab (n=145). 487 patients had a CDAI-70 response in any of the induction cohorts and were enrolled into the maintenance cohort, of whom 434 had a response to etrolizumab and were randomly assigned to placebo (n=217) or 105 mg etrolizumab (n=217). At week 14, 48 (33%) of 145 patients in the 210 mg induction etrolizumab group versus 28 (29%) of 96 patients in the placebo induction group were in clinical remission (adjusted treatment difference 3·8% [95% CI -8·3 to 15·3]; p=0·52), and 40 (27%) versus 21 (22%) showed endoscopic improvement (5·8% [-5·4 to 17·1]; p=0·32). At week 66, a significantly higher proportion of patients receiving etrolizumab than those receiving placebo had clinical remission (76 [35%] of 217 vs 52 [24%] of 217; adjusted treatment difference 11·3% [95% CI 2·7-19·7]; p=0·0088) and endoscopic improvement (51 [24%] vs 26 [12%]; 11·5% [4·1-18·8]; p=0·0026). Similar proportions of patients reported one or more adverse events during induction (95 [66%] of 143 in the 105 mg etrolizumab group, 85 [59%] of 145 in the 210 mg etrolizumab group, and 51 [53%] of 96 in the placebo group) and maintenance (189 [87%] of 217 in the etrolizumab group and 190 [88%] of 217 in the placebo group). During induction, the most common treatment-related adverse events were injection site erythema (six [4%] of 143 in the 105 mg etrolizumab group, four [3%] of 145 in the 210 mg etrolizumab group, and none of 96 in the placebo group), and arthralgia (two [1%], one [1%], and four [4%]). In the maintenance cohort, the most common treatment-related adverse events were injection site erythema (six [3%] of 217 in the etrolizumab group vs 14 [6%] of 217 in the placebo: group), arthralgia (five [2%] vs eight [4%]), and headache (five [2%] vs seven [3%]). The most common serious adverse event was exacerbation of Crohn's disease (14 [6%] of 217 patients taking placebo and four [2%] of 217 patients taking 105 mg etrolizumab in the maintenance cohort). INTERPRETATION A significantly higher proportion of patients with moderately to severely active Crohn's disease achieved clinical remission and endoscopic improvement with etrolizumab than placebo during maintenance, but not during induction. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- William J Sandborn
- Department of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Julian Panés
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, August Pi i Sunyer Biomedical Research Institute, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milan, Italy
| | | | | | | | | | - Marco Daperno
- SC Gastroenterologia AO Ordine Mauriziano di Torino, Turin, Italy
| | - John F Valentine
- Division of Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, UT, USA
| | - David Laharie
- Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, Service d'Hépato-gastroentérologie et Oncologie Digestive - Université de Bordeaux, Bordeaux, France
| | - Carolina Baía
- Médica Gastroenterologista em Belo Horizonte, Minas Gerais, Brazil
| | - Raja Atreya
- Medical Clinic 1, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Remo Panaccione
- Division of Gastroenterology and Hepatology, University of Calgary, Calgary, AB, Canada
| | - Grazyna Rydzewska
- Central Clinical Hospital of the Ministry of Interior in Warsaw, Warsaw, Poland
| | | | - Séverine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium.
| | | |
Collapse
|
5
|
Ozanimod as a novel oral small molecule therapy for the treatment of Crohn's disease: The YELLOWSTONE clinical trial program. Contemp Clin Trials 2022; 122:106958. [PMID: 36208720 PMCID: PMC10008122 DOI: 10.1016/j.cct.2022.106958] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/29/2022] [Accepted: 10/01/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Ozanimod, an oral sphingosine 1-phosphate receptor modulator currently approved for the treatment of moderately to severely active ulcerative colitis and relapsing multiple sclerosis, showed clinical, endoscopic, and histological benefit in the phase 2 STEPSTONE trial for Crohn's disease (CD). We aim to describe the trial design of the YELLOWSTONE phase 3 program evaluating the safety and efficacy of ozanimod in patients with moderately to severely active CD. METHODS The YELLOWSTONE program consists of phase 3, randomized, double-blind, placebo-controlled induction (NCT03440372 and NCT03440385) and maintenance (NCT03464097) trials and an open-label extension (OLE) study (NCT03467958). Patients with inadequate response or intolerance to ≥1 CD treatment are randomized to receive daily ozanimod 0.92 mg (equivalent to ozanimod HCl 1 mg) or placebo for 12 weeks during induction. Those who respond to ozanimod are rerandomized to continue ozanimod or placebo maintenance therapy for 52 weeks. Patients who do not meet criteria for maintenance, experience relapse during maintenance, or complete maintenance or ≥ 1 year of STEPSTONE are eligible for open-label treatment for up to 234 weeks. Efficacy endpoints include clinical, endoscopic, and histologic outcomes. RESULTS Expected 2023 (induction studies), 2024 (maintenance study), and 2026 (OLE). CONCLUSION YELLOWSTONE will provide pivotal phase 3 data on the safety and efficacy of ozanimod in patients with moderately to severely active CD using state-of-the-art methods, including centrally read endoscopic and histologic measurements, along with subjective assessments of symptom control based on the Crohn's Disease Activity Index. These studies could enable approval of ozanimod as a new CD therapy. CLINICAL TRIAL REGISTRATION NUMBERS NCT03440372, NCT03440385, NCT03464097, NCT03467958.
Collapse
|
6
|
Raine T, Pavey H, Qian W, Moran GW, Subramanian S, Swaby L, Travis SP, Din S, Irving PM, Lindsay JO, Parkes M, Kennedy NA. Establishment of a validated central reading system for ileocolonoscopy in an academic setting. Gut 2022; 71:661-664. [PMID: 35022265 DOI: 10.1136/gutjnl-2021-325575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/23/2021] [Indexed: 12/08/2022]
Affiliation(s)
- Tim Raine
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Holly Pavey
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - Wendi Qian
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - Gordon W Moran
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, Nottinghamshire, UK
| | - Sreedhar Subramanian
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lizzie Swaby
- Clinical Trials Research Unit, ScHARR, The University of Sheffield, Sheffield, Sheffield, UK
| | - Simon Pl Travis
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Shahida Din
- Edinburgh IBD Unit, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Peter M Irving
- IBD Unit, Guy's and St Thomas' Hospital NHS Trust, London, UK.,School of Immunology & Microbial Sciences, King's College London, London, London, UK
| | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Miles Parkes
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nicholas A Kennedy
- Gastroenterology, Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, UK
| |
Collapse
|
7
|
Novak KL, Nylund K, Maaser C, Petersen F, Kucharzik T, Lu C, Allocca M, Maconi G, de Voogd F, Christensen B, Vaughan R, Palmela C, Carter D, Wilkens R. Expert Consensus on Optimal Acquisition and Development of the International Bowel Ultrasound Segmental Activity Score [IBUS-SAS]: A Reliability and Inter-rater Variability Study on Intestinal Ultrasonography in Crohn's Disease. J Crohns Colitis 2021; 15:609-616. [PMID: 33098642 PMCID: PMC8023841 DOI: 10.1093/ecco-jcc/jjaa216] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Intestinal ultrasound [IUS] is an accurate, patient-centreed monitoring tool that objectively evaluates Crohn's disease [CD] activity. However no current, widely accepted, reproducible activity index exists to facilitate consistent IUS identification of inflammatory activity. The aim of this study is to identify key parameters of CD inflammation on IUS, evaluate their reliability, and develop an IUS index reflecting segmental activity. METHODS There were three phases: [1] expert consensus Delphi method to derive measures of IUS activity; [2] an initial, multi-expert case acquisition and expert interpretation of 20 blinded cases, to measure inter-rater reliability for individual measures; [3] refinement of case acquisition and interpretation by 12 international experts, with 30 blinded case reads with reliability assessment and development of a segmental activity score. RESULTS Delphi consensus: 11 experts representing seven countries identified four key parameters including: [1] bowel wall thickness [BWT]; [2] bowel wall stratification; [3] hyperaemia of the wall [colour Doppler imaging]; and [4] inflammatory mesenteric fat. Blind read: each variable exhibited moderate to substantial reliability. Optimal, standardised image and cineloop acquisition were established. Second blind read and score development: intra-class correlation coefficient [ICC] for BWT was almost perfect at 0.96 [0.94-0.98]. All four parameters correlated with the global disease activity assessment and were included in the final International Bowel Ultrasound Segmental Activity Score with almost perfect ICC (0.97 [0.95-0.99, p <0.001]). CONCLUSIONS Using expert consensus and standardised approaches, identification of key activity measurements on IUS has been achieved and a segmental activity score has been proposed, demonstrating excellent reliability.
Collapse
Affiliation(s)
- Kerri L Novak
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, AB, Canada
| | - Kim Nylund
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
- Institute of Clinical Medicine, University in Bergen, Klinisk institutt 1, Bergen, Norway
| | - Christian Maaser
- Outpatient Department of Gastroenterology, Department of Geriatric Medicine, University Teaching Hospital Lueneburg, Lueneburg, Germany
| | - Frauke Petersen
- Department of Gastroenterology, University Teaching Hospital Lueneburg, Lueneburg, Germany
| | - Torsten Kucharzik
- Department of Gastroenterology, University Teaching Hospital Lueneburg, Lueneburg, Germany
| | - Cathy Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, AB, Canada
| | - Mariangela Allocca
- Humanitas Clinical and Research Centre, Rozzano, Italy
- Humanitas University, Department of Biomedical Sciences, Milan, Italy
| | - Giovanni Maconi
- Gastroenterology Unit, Department of Biomedical and Clinical Sciences. FBF- L.Sacco University Hospital, Milan. Italy
| | - Floris de Voogd
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Britt Christensen
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC Australia
| | - Rose Vaughan
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC Australia
| | - Carolina Palmela
- Division of Gastroenterology, Department of General Surgery, Hospital Beatriz Ângelo, Loures, Portugal
| | - Dan Carter
- Department of Gastroenterology, Chaim Sheba Medical Centre, Tel Hashomer, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rune Wilkens
- Gastrounit, Division of Medicine, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
- Copenhagen Centre for Inflammatory Bowel Disease in Children, Adolescents and Adults, University of Copenhagen, Hvidovre Hospital, Copenhagen, Denmark
| |
Collapse
|
8
|
Gottlieb K, Requa J, Karnes W, Chandra Gudivada R, Shen J, Rael E, Arora V, Dao T, Ninh A, McGill J. Central Reading of Ulcerative Colitis Clinical Trial Videos Using Neural Networks. Gastroenterology 2021; 160:710-719.e2. [PMID: 33098883 DOI: 10.1053/j.gastro.2020.10.024] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Endoscopic disease activity scoring in ulcerative colitis (UC) is useful in clinical practice but done infrequently. It is required in clinical trials, where it is expensive and slow because human central readers are needed. A machine learning algorithm automating the process could elevate clinical care and facilitate clinical research. Prior work using single-institution databases and endoscopic still images has been promising. METHODS Seven hundred and ninety-five full-length endoscopy videos were prospectively collected from a phase 2 trial of mirikizumab with 249 patients from 14 countries, totaling 19.5 million image frames. Expert central readers assigned each full-length endoscopy videos 1 endoscopic Mayo score (eMS) and 1 Ulcerative Colitis Endoscopic Index of Severity (UCEIS) score. Initially, video data were cleaned and abnormality features extracted using convolutional neural networks. Subsequently, a recurrent neural network was trained on the features to predict eMS and UCEIS from individual full-length endoscopy videos. RESULTS The primary metric to assess the performance of the recurrent neural network model was quadratic weighted kappa (QWK) comparing the agreement of the machine-read endoscopy score with the human central reader score. QWK progressively penalizes disagreements that exceed 1 level. The model's agreement metric was excellent, with a QWK of 0.844 (95% confidence interval, 0.787-0.901) for eMS and 0.855 (95% confidence interval, 0.80-0.91) for UCEIS. CONCLUSIONS We found that a deep learning algorithm can be trained to predict levels of UC severity from full-length endoscopy videos. Our data set was prospectively collected in a multinational clinical trial, videos rather than still images were used, UCEIS and eMS were reported, and machine learning algorithm performance metrics met or exceeded those previously published for UC severity scores.
Collapse
Affiliation(s)
| | | | | | | | - Jie Shen
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Vipin Arora
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | |
Collapse
|
9
|
Reinisch W, Mishkin DS, Oh YS, Schreiber S, Hussain F, Jacob R, Hassanali A, Daperno M. Impact of various central endoscopy reading models on treatment outcome in Crohn's disease using data from the randomized, controlled, exploratory cohort arm of the BERGAMOT trial. Gastrointest Endosc 2021; 93:174-182.e2. [PMID: 32464142 DOI: 10.1016/j.gie.2020.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/01/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Endoscopic assessment of mucosal appearance by independent central reading has become the standard method to assess Crohn's disease activity in clinical trials. The performance characteristics of various endoscopy reading models have yet to be systematically evaluated. METHODS This substudy included patients with Crohn's disease in the exploratory induction cohort of the BERGAMOT trial (NCT02394028) randomly assigned to etrolizumab or placebo. Endoscopies conducted at baseline and week 14 were independently scored using the Simple Endoscopic Score for Crohn's Disease (SES-CD) by a local reader (LR) and 2 central readers (CRs). Five endoscopy reading models were compared: single LR, single CR, average of 2 CRs, and 2 models incorporating the LR and 1 or 2 CRs depending on alignment between the LR and the CR, defined according to a sliding scale applied to a range of scores. RESULTS Five hundred thirty-five videos were scored. Models involving 2 readers demonstrated lower placebo rates (3.4%) than the single LR (11.9%) and the single CR (6.8%) models. Treatment effect size based on endoscopic improvement (≥50% reduction in SES-CD from baseline) was highest with the 2 models incorporating the LR and 1 or 2 CRs (Δ = 16.2%). Further, in the etrolizumab arm, models with 2 readers demonstrated the lowest variability for the SES-CD. CONCLUSIONS Central endoscopy reading models in Crohn's disease have an impact on placebo response rates and effect size. Incorporating the LR appears to be important because models using both CRs and LRs resulted in the greatest treatment effect size for endoscopic improvement with etrolizumab, lower placebo rates, and reduced variability.
Collapse
Affiliation(s)
| | | | - Young S Oh
- Genentech, Inc, South San Francisco, California, USA
| | | | - Fez Hussain
- Gastroenterology Center of Excellence, IQVIA, Reading
| | - Rhian Jacob
- Roche Products Limited, Welwyn Garden City, United Kingdom
| | | | | |
Collapse
|
10
|
Hart L, Chavannes M, Lakatos PL, Afif W, Bitton A, Bressler B, Bessissow T. Do You See What I See? An Assessment of Endoscopic Lesions Recognition and Description by Gastroenterology Trainees and Staff Physicians. J Can Assoc Gastroenterol 2020; 3:216-221. [PMID: 32905160 PMCID: PMC7465549 DOI: 10.1093/jcag/gwz022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Gastroenterologists should accurately describe endoscopic findings and integrate them into management plans. We aimed to determine if trainees and staff are describing inflammatory bowel disease (IBD) lesions in a similar manner. Methods Using 20 ileocolonoscopy images, participants described IBD inflammatory burden based on physician severity rating, and Mayo endoscopic score (MES) (ulcerative colitis [UC]) or simple endoscopic score (SES-CD) (Crohn’s disease [CD]). Images were selected based on agreement by three IBD experts. Findings of varying severity were presented; 10 images included a question about management. We examined inter-observer agreement among trainees and staff, compared trainees to staff, and determined accuracy of response comparing both groups to IBD experts. Results One hundred and twenty-nine staff and 47 trainees participated from across Canada. There was moderate inter-rater agreement using physician severity rating (κ = 0.53 UC and 0.52 CD for staff, κ = 0.51 UC and 0.43 CD for trainees). There was moderate inter-rater agreement for MES for staff and trainees (κ = 0.49 and 0.48, respectively), but fair agreement for SES-CD (κ = 0.37 and 0.32, respectively). For accuracy of response, the mean score was 68.7% for staff and 63.7% for trainees (P = 0.028). Both groups identified healed bowel or severe disease better than mild/moderate (P < 0.05). There was high accuracy for management, but staff scored higher than trainees for UC (P < 0.01). Conclusion Inter-rater agreement on description of IBD lesions was moderate at best. Staff and trainees more accurately describe healed and severe disease, and better describe lesions in UC than CD.
Collapse
Affiliation(s)
- Lara Hart
- Division of Gastroenterology, McGill University Health Center, Montreal, Quebec, Canada
| | - Mallory Chavannes
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter L Lakatos
- Division of Gastroenterology, McGill University Health Center, Montreal, Quebec, Canada.,Division of Gastroenterology, Semmelweis University, Budapest, Hungary
| | - Waqqas Afif
- Division of Gastroenterology, McGill University Health Center, Montreal, Quebec, Canada
| | - Alain Bitton
- Division of Gastroenterology, McGill University Health Center, Montreal, Quebec, Canada
| | - Brian Bressler
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Talat Bessissow
- Division of Gastroenterology, McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Abadir AP, Ali MF, Karnes W, Samarasena JB. Artificial Intelligence in Gastrointestinal Endoscopy. Clin Endosc 2020; 53:132-141. [PMID: 32252506 PMCID: PMC7137570 DOI: 10.5946/ce.2020.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Artificial intelligence (AI) is rapidly integrating into modern technology and clinical practice. Although in its nascency, AI has become a hot topic of investigation for applications in clinical practice. Multiple fields of medicine have embraced the possibility of a future with AI assisting in diagnosis and pathology applications. In the field of gastroenterology, AI has been studied as a tool to assist in risk stratification, diagnosis, and pathologic identification. Specifically, AI has become of great interest in endoscopy as a technology with substantial potential to revolutionize the practice of a modern gastroenterologist. From cancer screening to automated report generation, AI has touched upon all aspects of modern endoscopy. Here, we review landmark AI developments in endoscopy. Starting with broad definitions to develop understanding, we will summarize the current state of AI research and its potential applications. With innovation developing rapidly, this article touches upon the remarkable advances in AI-assisted endoscopy since its initial evaluation at the turn of the millennium, and the potential impact these AI models may have on the modern clinical practice. As with any discussion of new technology, its limitations must also be understood to apply clinical AI tools successfully.
Collapse
Affiliation(s)
- Alexander P Abadir
- Department of Medicine, University of California Irvine, Orange, CA, USA
| | - Mohammed Fahad Ali
- Department of Medicine, University of California Irvine, Orange, CA, USA
| | - William Karnes
- Division of Gastroenterology & Hepatology, Department of Medicine, H. H. Chao Comprehensive Digestive Disease Center, University of California Irvine, Orange, CA, USA
| | - Jason B Samarasena
- Division of Gastroenterology & Hepatology, Department of Medicine, H. H. Chao Comprehensive Digestive Disease Center, University of California Irvine, Orange, CA, USA
| |
Collapse
|
12
|
Reinisch W, Gottlieb K, Colombel JF, Danese S, Panaccione R, Panes J, Peyrin-Biroulet L, Rubin D, Sands BE, Schreiber S, Vermeire S, Mulberg A, Sandborn B. Comparison of the EMA and FDA Guidelines on Ulcerative Colitis Drug Development. Clin Gastroenterol Hepatol 2019; 17:1673-1679.e1. [PMID: 31352970 DOI: 10.1016/j.cgh.2018.10.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/09/2018] [Accepted: 10/21/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS In 2016, the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) released revised EMA and new FDA draft guidelines related to the development of drugs intended for the treatment of ulcerative colitis. We sought to compare and contrast the EMA draft guideline with the FDA draft guidance to facilitate further discussion and perhaps harmonization between the 2 guidelines when they are finalized. METHODS A concordance document was created by arranging like or similar topics addressed by the guidelines side by side in a tabular format. This concordance table served as a source for writing the narrative. The first draft was subjected to repeated rounds of reviews and revisions by the authors and outside reviewers, all of them familiar with the subject matter from a regulatory science and/or academic perspective. RESULTS The FDA guidance focuses on end points, whereas the EMA guideline additionally supplies much useful information for trial design. FDA guidance appears more aspirational, suggesting the development of entirely new patient-reported outcome instruments and the incorporation of a not-yet-validated histology instrument into the definition of mucosal healing. CONCLUSIONS The guidelines by the FDA and the EMA complement each other and together are aimed to further practical drug development toward more clinically relevant end points in ulcerative colitis. Efforts are needed to harmonize the documents.
Collapse
Affiliation(s)
- Walter Reinisch
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| | | | - Jean-Frederic Colombel
- Dr Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Silvio Danese
- Inflammatory Bowel Disease Center, Humanitas University, Milano, Italy
| | - Remo Panaccione
- Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Canada
| | - Julian Panes
- Department of Gastroenterology, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Laurent Peyrin-Biroulet
- Inserm U954, Department of Gastroenterology, Nancy University Hospital, Lorraine University, Nancy, France
| | - David Rubin
- Department of Gastroenterology, Hepatology and Nutrition, University of Chicago Medicine, Chicago, Illinois
| | - Bruce E Sands
- Dr Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stefan Schreiber
- Klinik für Innere Medizin I, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Severine Vermeire
- Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | - Bill Sandborn
- University of California San Diego, La Jolla, California
| |
Collapse
|
13
|
Cucchiarelli S, Santopaolo F, Lamazza A, Lionetti R, Lenci I, Manzia TM, Angelico M, Milana M, Baiocchi L. Pitfalls in the reporting of upper endoscopy features in cirrhotic patients. Dig Liver Dis 2019; 51:382-385. [PMID: 30219669 DOI: 10.1016/j.dld.2018.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Upper endoscopy is the main tool for the accurate assessment of the risk of bleeding in cirrhotic patients. AIM To evaluate the diagnostic accuracy of upper endoscopy, in cirrhotic subjects, during common clinical practice. METHODS 120 endoscopic reports produced in different hospitals in our region were retrospectively and randomly selected. After a general evaluation, aimed at assessing the description of various endoscopic features, reports were evaluated by four expert endoscopists and four expert hepatologists. Experts were asked to fill in a questionnaire for each single endoscopic procedure, regarding the diagnostic accuracy of the report. RESULTS Endoscopic reports lacked descriptions of the size of esophageal varices and red signs in 14% and 29% of cases respectively. Presence (or absence) of gastric varices or portal hypertensive gastropathy were not reported in 62% and 34% of cases respectively. According to expert endoscopists 41% of the reports were incomplete, while, according to hepatologists, reports were incomplete and inadequate for clinical purposes in 36% of cases. CONCLUSION Our study clearly evidenced a significant lack of information in reports on upper endoscopy in cirrhotic patients, and supports the prompt adoption of corrective strategies.
Collapse
Affiliation(s)
| | | | | | - Raffaella Lionetti
- Infectious disease and Hepatology Unit, Lazzaro Spallanzani Hospital, Rome, Italy
| | - Ilaria Lenci
- Hepatology Unit, Tor Vergata University Hospital, Rome, Italy
| | | | - Mario Angelico
- Hepatology Unit, Tor Vergata University Hospital, Rome, Italy
| | - Martina Milana
- Hepatology Unit, Tor Vergata University Hospital, Rome, Italy
| | | |
Collapse
|
14
|
Bossuyt P, Louis E, Mary JY, Vermeire S, Bouhnik Y. Defining Endoscopic Remission in Ileocolonic Crohn's Disease: Let's Start from Scratch. J Crohns Colitis 2018; 12:1245-1248. [PMID: 29982528 DOI: 10.1093/ecco-jcc/jjy097] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endoscopic remission is the cornerstone of drug development and the guidance for daily clinical practice in Crohn's disease [CD]. However, although scoring systems for endoscopic activity in CD have been available for more than three decades, no consensus exists on the definition of endoscopic remission. In this viewpoint we describe the shortcomings of the current definition of endoscopic remission in ileocolonic CD and the essential requirements for a newly developed endoscopic scoring tool for endoscopic remission in CD.
Collapse
Affiliation(s)
- Peter Bossuyt
- Imelda GI Clinical Research Centre, Department of Gastroenterology, Bonheiden, Belgium.,University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Edouard Louis
- CHU de Liège, Department of Gastroenterology, Liège, Belgium
| | - Jean-Yves Mary
- INSERM UMR 1153, Paris Diderot University, Saint Louis Hospital, Paris, France
| | - Séverine Vermeire
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Yoram Bouhnik
- Beaujon Hospital, Department of Gastroenterology, APHP, Paris Diderot University, Clichy, France
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Since the first clinical trial of cortisone in ulcerative colitis in 1955, remarkable progress has been made in the design and conduct of clinical trials in inflammatory bowel diseases (IBD). In this review article, we will discuss evolution of clinical trials in IBD over the last 3-5 years. RECENT FINDINGS Recognizing limitations intrinsic to clinical disease activity indices in IBD, regulatory authorities have recommended evaluating co-primary endpoints of patient-reported outcomes and endoscopic disease activity in clinical trials. Biomarker-enriched trial enrolment and central endoscopy reading have emerged as critical events in trial recruitment and outcome assessment and have driven placebo response rates down. While trials of novel biologic therapies and advanced small molecules continue at an accelerated pace, pragmatic comparative efficacy trials of treatment strategy aimed at optimizing current therapies (such as early combined immunosuppression [REACT], tight disease control [CALM], therapeutic drug monitoring [TAXIT, TAILORIX]) have directly informed clinical practice. With emphasis on value-based care and population health management, multi-pronged remote monitoring, self-management, and telemedicine approaches in the era of smartphones have re-emerged with promise. Non-conventional therapies such as fecal microbiota transplantation, though still experimental, have provided insight into disease pathogenesis and offered hope for microbial manipulation strategies for treating these complex diseases. Clinical trials have rapidly evolved over the last 5 years not only focusing on novel therapies but also optimizing existing treatment approaches and population health management. Over the next decade, these trials will continue to advance the field, and be readily translatable into clinical practice.
Collapse
|
16
|
Horaist C, de Parades V, Abramowitz L, Benfredj P, Bonnaud G, Bouchard D, Fathallah N, Sénéjoux A, Siproudhis L, Staumont G, Viguier M, Marteau P. Elaboration and validation of Crohn’s disease anoperineal lesions consensual definitions. World J Gastroenterol 2017; 23:5371-5378. [PMID: 28839437 PMCID: PMC5550786 DOI: 10.3748/wjg.v23.i29.5371] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/11/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To establish consensual definitions of anoperineal lesions of Crohn’s (APLOC) disease and assess interobserver agreement on their diagnosis between experts.
METHODS A database of digitally recorded pictures of APLOC was examined by a coordinating group who selected two series of 20 pictures illustrating the various aspects of APLOC. A reading group comprised: eight experts from the Société Nationale Française de Colo Proctologie group of study and research in proctology and one academic dermatologist. All members of the coordinating and reading groups participated in dedicated meetings. The coordinating group initially conducted a literature review to analyse verbatim descriptions used to evaluate APLOC. The study included two phases: establishment of consensual definitions using a formal consensus method and later assessment of interobserver agreement on the diagnosis of APLOC using photos of APLOC, a standardised questionnaire and Fleiss’s kappa test or descriptive statistics.
RESULTS Terms used in literature to evaluate visible APLOC did not include precise definitions or reference to definitions. Most of the expert reports on the first set of photos agreed with the main diagnosis but their verbatim reporting contained substantial variation. The definitions of ulceration (entity, depth, extension), anal skin tags (entity, inflammatory activity, ulcerated aspect), fistula (complexity, quality of drainage, inflammatory activity of external openings), perianal skin lesions (abscess, papules, edema, erythema) and anoperineal scars were validated. For fistulae, they decided to follow the American Gastroenterology Association’s guidelines definitions. The diagnosis of ulceration (κ = 0.70), fistulae (κ = 0.75), inflammatory activity of external fistula openings (86.6% agreement), abscesses (84.6% agreement) and erythema (100% agreement) achieved a substantial degree of interobserver reproducibility.
CONCLUSION This study constructed consensual definitions of APLOC and their characteristics and showed that experts have a fair level of interobserver agreement when using most of the definitions.
Collapse
|
17
|
Kahan BC, Feagan B, Jairath V. A comparison of approaches for adjudicating outcomes in clinical trials. Trials 2017; 18:266. [PMID: 28595589 PMCID: PMC5465459 DOI: 10.1186/s13063-017-1995-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/17/2017] [Indexed: 01/13/2023] Open
Abstract
Background Incorrect classification of outcomes in clinical trials can lead to biased estimates of treatment effect and reduced power. Ensuring appropriate adjudication methods to minimize outcome misclassification is therefore essential. While there are many reported adjudication approaches, there is little consensus over which approach is best. Methods Under the assumption of non-differential assessment (i.e. that misclassification rates are the same in each treatment arm, as would typically be the case when outcome assessors are blinded), we use simulation and theoretical results to address four different questions about outcome adjudication: (a) How many assessors should be used? (b) When is it better to use onsite or central assessment? (c) Should central assessors adjudicate all outcomes, or only suspected events? (d) Should central assessment with multiple assessors be done independently or through group consensus? Results No one adjudication approach performs optimally in all settings. The optimal approach depends on the misclassification rates of site and central assessors, and the correlation between assessors. We found: (a) there will generally be little incremental benefit to using more than three assessors and, for outcomes with very high correlation between assessors, using one assessor is sufficient; (b) when choosing between site and central assessors, the assessor with the smallest misclassification rate should be chosen; when these rates are unknown, a combination of one site assessor and two central assessors will provide good results across a range of scenarios; (c) having central assessors adjudicate only suspected events will typically increase bias, and should be avoided, unless the threshold for sending outcomes for central assessment is extremely low; (d) central assessors can adjudicate either independently or in a group, and the preferred option should be dictated by whichever is expected to have the lowest misclassification rate. Conclusions Outcome adjudication is of critical importance to ensure validity of trial results, although no one approach is optimal across all settings. Investigators should choose the best strategy based on the specific characteristics of their trial. Regardless of the adjudication strategy chosen, assessors should be qualified and receive appropriate training.
Collapse
Affiliation(s)
- Brennan C Kahan
- Pragmatic Clinical Trials Unit, Queen Mary University of London, 58 Turner St, London, E1 2AB, UK.
| | - Brian Feagan
- Robarts Clinical Trials, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - Vipul Jairath
- Robarts Clinical Trials, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
| |
Collapse
|
18
|
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by periods of remission and periods of relapse. Patients often present with symptoms such as rectal bleeding, diarrhea and weight loss, and may require hospitalization and even colectomy. Long-term complications of UC include decreased quality of life and productivity and an increased risk of colorectal cancer. Mucosal healing (MH) has gained progressive importance in the management of UC patients. In this article, we review the endoscopic findings that define both mucosal injury and MH, and the strengths and limitations of the scoring systems currently available in clinical practice. The basic mechanisms behind colonic injury and MH are covered, highlighting the pathways through which different drugs exert their effect towards reducing inflammation and promoting epithelial repair. A comprehensive review of the evidence for approved drugs for UC to achieve and maintain MH is provided, including a section on the pharmacokinetics of anti-tumor necrosis factor (TNF)-α drugs. Currently approved drugs with proven efficacy in achieving MH in UC include salicylates, corticosteroids (induction only), calcineurin inhibitors (induction only), thiopurines, vedolizumab and anti-TNFα drugs (infliximab, adalimumab, and golimumab). MH is of crucial relevance in the outcomes of UC, resulting in lower incidences of clinical relapse, the need for hospitalization and surgery, as well as reduced rates of dysplasia and colorectal cancer. Finally, we present recent evidence towards the need for a more strict definition of complete MH as the preferred endpoint for UC patients, using a combination of both endoscopic and histological findings.
Collapse
|