Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

FIB-4 Index Misclassifies Many Patients

Article Type
Changed
Tue, 06/11/2024 - 16:35

The Fibrosis-4 (FIB-4) index shows high discordance with liver stiffness measurement (LSM) via transient elastography, suggesting that many patients are misclassified, potentially impacting clinical decisions, according to investigators.

These findings call for a cautious interpretation of low-risk FIB-4 results among patients at greatest risk of misclassification, and/or use of alternative assessment strategies, reported Mazen Noureddin, MD, MHSc, of Houston Methodist Hospital, and coauthors.

swesabobogathobegeuecucheticrithucrufromocaphemaluclemulawromilokuswagucinobegitorilaspouaposididefrolowraprobeseswewrigivepr
Dr. Mazen Noureddin

“Currently, the AGA/AASLD Pathways recommends identifying patients at risk for metabolic dysfunction-associated steatotic liver disease (MASLD), then using sequential testing with FIB-4 followed by FibroScan to risk-stratify patients,” the investigators wrote in Clinical Gastroenterology and Hepatology.

Yet the performance of the FIB-4 index in this context remains unclear.

“Previous studies have shown FIB-4 to have low accuracy for screening liver fibrosis, especially among obese and diabetic patients,” the investigators wrote. “Thus, there is a concern that classifying patients with FIB-4 can lead to misclassification and missed diagnosis.”

To explore this concern, Dr. Noureddin and colleagues turned to data from the 2017-2020 National Health and Nutrition Examination Surveys, including 5285 subjects at risk for MASLD. Exclusions were made for those with excessive alcohol intake or other liver diseases, resulting in a final cohort of 3741 individuals.

All subjects were classified as low-, indeterminate-, or high-risk for advanced liver fibrosis based on FIB-4 scores. These scores were then compared with liver stiffness measurements (LSM) obtained through transient elastography (FibroScan).

Out of 2776 subjects classified as low-risk by FIB-4, 277 (10%) were reclassified as higher risk by LSM, including 75 (2.7%) who were found to be at high risk. Out of 879 subjects with indeterminate FIB-4 scores, 37 (4.2%) were at high risk according to LSM. Finally, among the 86 subjects classified as high risk by FIB-4, 68 (79.1%) were reclassified as lower risk by LSM, including 54 (62.8%) who were deemed low risk.

Subjects misclassified as low risk by FIB-4 were typically older and had higher waist circumferences, body mass indices, glycohemoglobin A1c levels, fasting glucose levels, liver enzyme levels, diastolic blood pressures, controlled attenuation parameter scores, white blood cell counts, and alkaline phosphatase levels, but lower high-density lipoprotein and albumin levels (all P less than .05). They were also more likely to have prediabetes or diabetes.

“[I]t is important to acknowledge that 10% of the subjects were misclassified as low risk by FIB-4,” Dr. Noureddin and colleagues wrote, including 2.7% of patients who were actually high risk. “This misclassification of high-risk patients can lead to missed diagnoses, delaying crucial medical treatments or lifestyle interventions.”

They therefore suggested cautious interpretation of low-risk FIB-4 results among patients with factors predicting misclassification, or even use of alternative diagnostic strategies.

“Some possible alternatives to FIB-4 include new serum tests such NIS-2+, MASEF, SAFE score, and machine learning methods,” Dr. Noureddin and colleagues wrote. “However, additional confirmatory and cost-effective studies are required to validate the effectiveness of these tests, including studies conducted on the general population.”

The investigators disclosed relationships with AbbVie, Corcept, Galectin, and others.

Publications
Topics
Sections

The Fibrosis-4 (FIB-4) index shows high discordance with liver stiffness measurement (LSM) via transient elastography, suggesting that many patients are misclassified, potentially impacting clinical decisions, according to investigators.

These findings call for a cautious interpretation of low-risk FIB-4 results among patients at greatest risk of misclassification, and/or use of alternative assessment strategies, reported Mazen Noureddin, MD, MHSc, of Houston Methodist Hospital, and coauthors.

swesabobogathobegeuecucheticrithucrufromocaphemaluclemulawromilokuswagucinobegitorilaspouaposididefrolowraprobeseswewrigivepr
Dr. Mazen Noureddin

“Currently, the AGA/AASLD Pathways recommends identifying patients at risk for metabolic dysfunction-associated steatotic liver disease (MASLD), then using sequential testing with FIB-4 followed by FibroScan to risk-stratify patients,” the investigators wrote in Clinical Gastroenterology and Hepatology.

Yet the performance of the FIB-4 index in this context remains unclear.

“Previous studies have shown FIB-4 to have low accuracy for screening liver fibrosis, especially among obese and diabetic patients,” the investigators wrote. “Thus, there is a concern that classifying patients with FIB-4 can lead to misclassification and missed diagnosis.”

To explore this concern, Dr. Noureddin and colleagues turned to data from the 2017-2020 National Health and Nutrition Examination Surveys, including 5285 subjects at risk for MASLD. Exclusions were made for those with excessive alcohol intake or other liver diseases, resulting in a final cohort of 3741 individuals.

All subjects were classified as low-, indeterminate-, or high-risk for advanced liver fibrosis based on FIB-4 scores. These scores were then compared with liver stiffness measurements (LSM) obtained through transient elastography (FibroScan).

Out of 2776 subjects classified as low-risk by FIB-4, 277 (10%) were reclassified as higher risk by LSM, including 75 (2.7%) who were found to be at high risk. Out of 879 subjects with indeterminate FIB-4 scores, 37 (4.2%) were at high risk according to LSM. Finally, among the 86 subjects classified as high risk by FIB-4, 68 (79.1%) were reclassified as lower risk by LSM, including 54 (62.8%) who were deemed low risk.

Subjects misclassified as low risk by FIB-4 were typically older and had higher waist circumferences, body mass indices, glycohemoglobin A1c levels, fasting glucose levels, liver enzyme levels, diastolic blood pressures, controlled attenuation parameter scores, white blood cell counts, and alkaline phosphatase levels, but lower high-density lipoprotein and albumin levels (all P less than .05). They were also more likely to have prediabetes or diabetes.

“[I]t is important to acknowledge that 10% of the subjects were misclassified as low risk by FIB-4,” Dr. Noureddin and colleagues wrote, including 2.7% of patients who were actually high risk. “This misclassification of high-risk patients can lead to missed diagnoses, delaying crucial medical treatments or lifestyle interventions.”

They therefore suggested cautious interpretation of low-risk FIB-4 results among patients with factors predicting misclassification, or even use of alternative diagnostic strategies.

“Some possible alternatives to FIB-4 include new serum tests such NIS-2+, MASEF, SAFE score, and machine learning methods,” Dr. Noureddin and colleagues wrote. “However, additional confirmatory and cost-effective studies are required to validate the effectiveness of these tests, including studies conducted on the general population.”

The investigators disclosed relationships with AbbVie, Corcept, Galectin, and others.

The Fibrosis-4 (FIB-4) index shows high discordance with liver stiffness measurement (LSM) via transient elastography, suggesting that many patients are misclassified, potentially impacting clinical decisions, according to investigators.

These findings call for a cautious interpretation of low-risk FIB-4 results among patients at greatest risk of misclassification, and/or use of alternative assessment strategies, reported Mazen Noureddin, MD, MHSc, of Houston Methodist Hospital, and coauthors.

swesabobogathobegeuecucheticrithucrufromocaphemaluclemulawromilokuswagucinobegitorilaspouaposididefrolowraprobeseswewrigivepr
Dr. Mazen Noureddin

“Currently, the AGA/AASLD Pathways recommends identifying patients at risk for metabolic dysfunction-associated steatotic liver disease (MASLD), then using sequential testing with FIB-4 followed by FibroScan to risk-stratify patients,” the investigators wrote in Clinical Gastroenterology and Hepatology.

Yet the performance of the FIB-4 index in this context remains unclear.

“Previous studies have shown FIB-4 to have low accuracy for screening liver fibrosis, especially among obese and diabetic patients,” the investigators wrote. “Thus, there is a concern that classifying patients with FIB-4 can lead to misclassification and missed diagnosis.”

To explore this concern, Dr. Noureddin and colleagues turned to data from the 2017-2020 National Health and Nutrition Examination Surveys, including 5285 subjects at risk for MASLD. Exclusions were made for those with excessive alcohol intake or other liver diseases, resulting in a final cohort of 3741 individuals.

All subjects were classified as low-, indeterminate-, or high-risk for advanced liver fibrosis based on FIB-4 scores. These scores were then compared with liver stiffness measurements (LSM) obtained through transient elastography (FibroScan).

Out of 2776 subjects classified as low-risk by FIB-4, 277 (10%) were reclassified as higher risk by LSM, including 75 (2.7%) who were found to be at high risk. Out of 879 subjects with indeterminate FIB-4 scores, 37 (4.2%) were at high risk according to LSM. Finally, among the 86 subjects classified as high risk by FIB-4, 68 (79.1%) were reclassified as lower risk by LSM, including 54 (62.8%) who were deemed low risk.

Subjects misclassified as low risk by FIB-4 were typically older and had higher waist circumferences, body mass indices, glycohemoglobin A1c levels, fasting glucose levels, liver enzyme levels, diastolic blood pressures, controlled attenuation parameter scores, white blood cell counts, and alkaline phosphatase levels, but lower high-density lipoprotein and albumin levels (all P less than .05). They were also more likely to have prediabetes or diabetes.

“[I]t is important to acknowledge that 10% of the subjects were misclassified as low risk by FIB-4,” Dr. Noureddin and colleagues wrote, including 2.7% of patients who were actually high risk. “This misclassification of high-risk patients can lead to missed diagnoses, delaying crucial medical treatments or lifestyle interventions.”

They therefore suggested cautious interpretation of low-risk FIB-4 results among patients with factors predicting misclassification, or even use of alternative diagnostic strategies.

“Some possible alternatives to FIB-4 include new serum tests such NIS-2+, MASEF, SAFE score, and machine learning methods,” Dr. Noureddin and colleagues wrote. “However, additional confirmatory and cost-effective studies are required to validate the effectiveness of these tests, including studies conducted on the general population.”

The investigators disclosed relationships with AbbVie, Corcept, Galectin, and others.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168271</fileName> <TBEID>0C05057E.SIG</TBEID> <TBUniqueIdentifier>MD_0C05057E</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CGH_Chang_FIB4</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240611T160847</QCDate> <firstPublished>20240611T163223</firstPublished> <LastPublished>20240611T163223</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240611T163223</CMSDate> <articleSource>FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY </articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The Fibrosis-4 (FIB-4) index shows high discordance with liver stiffness measurement (LSM) via transient elastography, suggesting that many patients are misclas</metaDescription> <articlePDF/> <teaserImage>301902</teaserImage> <teaser>Out of 2776 subjects classified as low-risk by FIB-4, 277 (10%) were reclassified as higher risk by liver stiffness measurements.</teaser> <title>FIB-4 Index Misclassifies Many Patients</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">346</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240129f9.jpg</altRep> <description role="drol:caption">Dr. Mazen Noureddin</description> <description role="drol:credit">Houston Methodist Hospital</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>FIB-4 Index Misclassifies Many Patients</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">The Fibrosis-4 (FIB-4) index shows high discordance with liver stiffness measurement (LSM) via transient elastography, suggesting that many patients are misclassified</span>, potentially impacting clinical decisions, according to investigators.</p> <p>These findings call for a cautious interpretation of low-risk FIB-4 results among patients at greatest risk of misclassification, and/or use of alternative assessment strategies, reported Mazen Noureddin, MD, MHSc, of Houston Methodist Hospital, and coauthors.[[{"fid":"301902","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Mazen Noureddin, Houston Methodist Hospital","field_file_image_credit[und][0][value]":"Houston Methodist Hospital","field_file_image_caption[und][0][value]":"Dr. Mazen Noureddin"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]“Currently, the AGA/AASLD Pathways recommends identifying patients at risk for metabolic dysfunction-associated steatotic liver disease (MASLD), then using sequential testing with FIB-4 followed by FibroScan to risk-stratify patients,” the <span class="Hyperlink"><a href="https://www.cghjournal.org/article/S1542-3565(24)00213-1/abstract">investigators wrote</a></span> in <em>Clinical Gastroenterology and Hepatology</em>.<br/><br/>Yet the performance of the FIB-4 index in this context remains unclear.<strong><br/><br/> </strong>“Previous studies have shown FIB-4 to have low accuracy for screening liver fibrosis, especially among obese and diabetic patients,” the investigators wrote. “Thus, there is a concern that classifying patients with FIB-4 can lead to misclassification and missed diagnosis.”<br/><br/>To explore this concern, Dr. Noureddin and colleagues turned to data from the 2017-2020 National Health and Nutrition Examination Surveys, including 5285 subjects at risk for MASLD. Exclusions were made for those with excessive alcohol intake or other liver diseases, resulting in a final cohort of 3741 individuals.<br/><br/>All subjects were classified as low-, indeterminate-, or high-risk for advanced liver fibrosis based on FIB-4 scores. These scores were then compared with liver stiffness measurements (LSM) obtained through transient elastography (FibroScan).<br/><br/>Out of 2776 subjects classified as low-risk by FIB-4, 277 (10%) were reclassified as higher risk by LSM, including 75 (2.7%) who were found to be at high risk. Out of 879 subjects with indeterminate FIB-4 scores, 37 (4.2%) were at high risk according to LSM. Finally, among the 86 subjects classified as high risk by FIB-4, 68 (79.1%) were reclassified as lower risk by LSM, including 54 (62.8%) who were deemed low risk.<br/><br/>Subjects misclassified as low risk by FIB-4 were typically older and had higher waist circumferences, body mass indices, glycohemoglobin A1c levels, fasting glucose levels, liver enzyme levels, diastolic blood pressures, controlled attenuation parameter scores, white blood cell counts, and alkaline phosphatase levels, but lower high-density lipoprotein and albumin levels (all <em>P</em> less than .05). They were also more likely to have prediabetes or diabetes.<br/><br/>“[I]t is important to acknowledge that 10% of the subjects were misclassified as low risk by FIB-4,” Dr. Noureddin and colleagues wrote, including 2.7% of patients who were actually high risk. “This misclassification of high-risk patients can lead to missed diagnoses, delaying crucial medical treatments or lifestyle interventions.”<br/><br/>They therefore suggested cautious interpretation of low-risk FIB-4 results among patients with factors predicting misclassification, or even use of alternative diagnostic strategies.<br/><br/>“Some possible alternatives to FIB-4 include new serum tests such NIS-2+, MASEF, SAFE score, and machine learning methods,” Dr. Noureddin and colleagues wrote. “However, additional confirmatory and cost-effective studies are required to validate the effectiveness of these tests, including studies conducted on the general population.”<br/><br/>The investigators disclosed relationships with AbbVie, Corcept, Galectin, and others.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Autonomous AI Outperforms Humans in Optical Diagnosis of Colorectal Polyps

‘Automatic’ CADx in Colonoscopy
Article Type
Changed
Tue, 06/11/2024 - 15:06

Autonomous artificial intelligence (AI) can achieve similar accuracy to AI-assisted humans (AI-H) in the optical diagnosis of diminutive colorectal polyps, while providing greater alignment with pathology-based surveillance intervals, based on a randomized controlled trial.

These findings suggest that autonomous AI may one day replace histologic assessment of diminutive polyps, reported lead author Roupen Djinbachian, MD, of the Montreal University Hospital Research Center, Montreal, Quebec, Canada, and colleagues.Optical diagnosis of diminutive colorectal polyps has been proposed as a cost-effective alternative to histologic diagnosis, but its implementation in general clinical practice has been hindered by endoscopists’ concerns about incorrect diagnoses, the investigators wrote in Gastroenterology.“AI-based systems (CADx) have been proposed as a solution to these barriers to implementation, with studies showing high adherence to Preservation and Incorporation of Valuable Endoscopic Innovations (PIVI) thresholds when using AI-H,” they wrote. “However, the efficacy and safety of autonomous AI-based diagnostic platforms have not yet been evaluated.”

To address this knowledge gap, Dr. Djinbachian and colleagues conducted a randomized controlled noninferiority trial involving 467 patients, all of whom underwent elective colonoscopies at a single academic institution.

Participants were randomly assigned to one of two groups. The first group received an optical diagnosis of diminutive (1-5 mm) colorectal polyps using an autonomous AI-based CADx system without any human input. The second group had diagnoses performed by endoscopists who used AI-H to make their optical diagnoses.

The primary outcome was the accuracy of optical diagnosis compared with the gold standard of histologic evaluation. Secondarily, the investigators explored associations between pathology-based surveillance intervals and various measures of accuracy, including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV).

The results showed that the accuracy of optical diagnosis for diminutive polyps was similar between the two groups, supporting noninferiority. Autonomous AI achieved an accuracy rate of 77.2%, while the AI-H group had an accuracy of 72.1%, which was not statistically significant (P = .86).

But when it came to pathology-based surveillance intervals, autonomous AI showed a clear advantage; the autonomous AI system achieved a 91.5% agreement rate, compared with 82.1% for the AI-H group (P = .016).

“These findings indicate that autonomous AI not only matches but also surpasses AI-H in accuracy for determining surveillance intervals,” the investigators wrote, noting that this finding highlights the “complexities of human interaction with AI modules where human intervention could lead to worse outcomes.”

Further analysis revealed that the sensitivity of autonomous AI for identifying adenomas was 84.8%, slightly higher than the 83.6% sensitivity of the AI-H group. Specificity was 64.4% for autonomous AI vs 63.8% for AI-H. While PPV was higher in the autonomous AI group (85.6%), compared with the AI-H group (78.6%), NPV was lower for autonomous AI than AI-H (63.0% vs 71.0%).

Dr. Djinbachian and colleagues suggested that future research should focus on larger, multicenter trials to validate these findings and further explore the integration of autonomous AI systems in clinical practice. They also noted that improving AI algorithms to accurately diagnose sessile serrated lesions could enhance the overall effectiveness of AI-based optical diagnosis.

“The performance of autonomous AI in accurately diagnosing diminutive polyps and determining appropriate surveillance intervals suggests that it could play a crucial role in streamlining colorectal cancer screening processes, reducing the burden on pathologists, and potentially lowering healthcare costs,” the investigators concluded.The study was supported by Fujifilm, which had no role in the study design or data analysis. Dr. von Renteln reported additional research funding from Vantage and Fujifilm.

Body

 

In the era of computer vision for endoscopy and colonoscopy, current paradigms rely on AI as a co-pilot or second observer, with the physician serving as the final arbiter in procedure-related decision-making. This study by Djinbachian and Haumesser et al brings up the interesting wrinkle of autonomous AI as a potentially superior (or noninferior) option in narrow, task-specific use cases.

In this study, human input from the endoscopist after CADx diagnosis led to lower agreement between the AI-predicted diagnosis and corresponding surveillance intervals; human oversight more often incorrectly changed the resultant diagnosis and led to shorter than recommended surveillance intervals.

This study offers a small but very important update to the growing body of literature on CADx in colonoscopy. So far, prospective validation of CADx compared with the human eye for in-situ diagnosis of polyps has provided mixed results. This study is one of the first to examine the potential role of “automatic” CADx without additional human input and sheds light on the importance of the AI-human hybrid in medical care. How do the ways in which humans interact with the user interface and output of AI lead to changes in outcome? How can we optimize the AI-human interaction in order to provide optimal results?

[[{"fid":"301890","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"}},"attributes":{"alt":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","class":"media-element file-medstat-image-flush-left","data-delta":"1"}}]]In this case, the suggestion is that less is more when it comes to human interference with optical diagnosis, but further research is needed on how to best optimize this important relationship as well as how AI might (or might not) support diagnose-and-leave and diagnose-and-discard strategies in the United States and worldwide.

Jeremy R. Glissen Brown is an assistant professor in the Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina. He has served as a consultant for Medtronic and Olympus, and on the advisory board for Odin Vision.

Publications
Topics
Sections
Body

 

In the era of computer vision for endoscopy and colonoscopy, current paradigms rely on AI as a co-pilot or second observer, with the physician serving as the final arbiter in procedure-related decision-making. This study by Djinbachian and Haumesser et al brings up the interesting wrinkle of autonomous AI as a potentially superior (or noninferior) option in narrow, task-specific use cases.

In this study, human input from the endoscopist after CADx diagnosis led to lower agreement between the AI-predicted diagnosis and corresponding surveillance intervals; human oversight more often incorrectly changed the resultant diagnosis and led to shorter than recommended surveillance intervals.

This study offers a small but very important update to the growing body of literature on CADx in colonoscopy. So far, prospective validation of CADx compared with the human eye for in-situ diagnosis of polyps has provided mixed results. This study is one of the first to examine the potential role of “automatic” CADx without additional human input and sheds light on the importance of the AI-human hybrid in medical care. How do the ways in which humans interact with the user interface and output of AI lead to changes in outcome? How can we optimize the AI-human interaction in order to provide optimal results?

[[{"fid":"301890","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"}},"attributes":{"alt":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","class":"media-element file-medstat-image-flush-left","data-delta":"1"}}]]In this case, the suggestion is that less is more when it comes to human interference with optical diagnosis, but further research is needed on how to best optimize this important relationship as well as how AI might (or might not) support diagnose-and-leave and diagnose-and-discard strategies in the United States and worldwide.

Jeremy R. Glissen Brown is an assistant professor in the Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina. He has served as a consultant for Medtronic and Olympus, and on the advisory board for Odin Vision.

Body

 

In the era of computer vision for endoscopy and colonoscopy, current paradigms rely on AI as a co-pilot or second observer, with the physician serving as the final arbiter in procedure-related decision-making. This study by Djinbachian and Haumesser et al brings up the interesting wrinkle of autonomous AI as a potentially superior (or noninferior) option in narrow, task-specific use cases.

In this study, human input from the endoscopist after CADx diagnosis led to lower agreement between the AI-predicted diagnosis and corresponding surveillance intervals; human oversight more often incorrectly changed the resultant diagnosis and led to shorter than recommended surveillance intervals.

This study offers a small but very important update to the growing body of literature on CADx in colonoscopy. So far, prospective validation of CADx compared with the human eye for in-situ diagnosis of polyps has provided mixed results. This study is one of the first to examine the potential role of “automatic” CADx without additional human input and sheds light on the importance of the AI-human hybrid in medical care. How do the ways in which humans interact with the user interface and output of AI lead to changes in outcome? How can we optimize the AI-human interaction in order to provide optimal results?

[[{"fid":"301890","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"}},"attributes":{"alt":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","class":"media-element file-medstat-image-flush-left","data-delta":"1"}}]]In this case, the suggestion is that less is more when it comes to human interference with optical diagnosis, but further research is needed on how to best optimize this important relationship as well as how AI might (or might not) support diagnose-and-leave and diagnose-and-discard strategies in the United States and worldwide.

Jeremy R. Glissen Brown is an assistant professor in the Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina. He has served as a consultant for Medtronic and Olympus, and on the advisory board for Odin Vision.

Title
‘Automatic’ CADx in Colonoscopy
‘Automatic’ CADx in Colonoscopy

Autonomous artificial intelligence (AI) can achieve similar accuracy to AI-assisted humans (AI-H) in the optical diagnosis of diminutive colorectal polyps, while providing greater alignment with pathology-based surveillance intervals, based on a randomized controlled trial.

These findings suggest that autonomous AI may one day replace histologic assessment of diminutive polyps, reported lead author Roupen Djinbachian, MD, of the Montreal University Hospital Research Center, Montreal, Quebec, Canada, and colleagues.Optical diagnosis of diminutive colorectal polyps has been proposed as a cost-effective alternative to histologic diagnosis, but its implementation in general clinical practice has been hindered by endoscopists’ concerns about incorrect diagnoses, the investigators wrote in Gastroenterology.“AI-based systems (CADx) have been proposed as a solution to these barriers to implementation, with studies showing high adherence to Preservation and Incorporation of Valuable Endoscopic Innovations (PIVI) thresholds when using AI-H,” they wrote. “However, the efficacy and safety of autonomous AI-based diagnostic platforms have not yet been evaluated.”

To address this knowledge gap, Dr. Djinbachian and colleagues conducted a randomized controlled noninferiority trial involving 467 patients, all of whom underwent elective colonoscopies at a single academic institution.

Participants were randomly assigned to one of two groups. The first group received an optical diagnosis of diminutive (1-5 mm) colorectal polyps using an autonomous AI-based CADx system without any human input. The second group had diagnoses performed by endoscopists who used AI-H to make their optical diagnoses.

The primary outcome was the accuracy of optical diagnosis compared with the gold standard of histologic evaluation. Secondarily, the investigators explored associations between pathology-based surveillance intervals and various measures of accuracy, including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV).

The results showed that the accuracy of optical diagnosis for diminutive polyps was similar between the two groups, supporting noninferiority. Autonomous AI achieved an accuracy rate of 77.2%, while the AI-H group had an accuracy of 72.1%, which was not statistically significant (P = .86).

But when it came to pathology-based surveillance intervals, autonomous AI showed a clear advantage; the autonomous AI system achieved a 91.5% agreement rate, compared with 82.1% for the AI-H group (P = .016).

“These findings indicate that autonomous AI not only matches but also surpasses AI-H in accuracy for determining surveillance intervals,” the investigators wrote, noting that this finding highlights the “complexities of human interaction with AI modules where human intervention could lead to worse outcomes.”

Further analysis revealed that the sensitivity of autonomous AI for identifying adenomas was 84.8%, slightly higher than the 83.6% sensitivity of the AI-H group. Specificity was 64.4% for autonomous AI vs 63.8% for AI-H. While PPV was higher in the autonomous AI group (85.6%), compared with the AI-H group (78.6%), NPV was lower for autonomous AI than AI-H (63.0% vs 71.0%).

Dr. Djinbachian and colleagues suggested that future research should focus on larger, multicenter trials to validate these findings and further explore the integration of autonomous AI systems in clinical practice. They also noted that improving AI algorithms to accurately diagnose sessile serrated lesions could enhance the overall effectiveness of AI-based optical diagnosis.

“The performance of autonomous AI in accurately diagnosing diminutive polyps and determining appropriate surveillance intervals suggests that it could play a crucial role in streamlining colorectal cancer screening processes, reducing the burden on pathologists, and potentially lowering healthcare costs,” the investigators concluded.The study was supported by Fujifilm, which had no role in the study design or data analysis. Dr. von Renteln reported additional research funding from Vantage and Fujifilm.

Autonomous artificial intelligence (AI) can achieve similar accuracy to AI-assisted humans (AI-H) in the optical diagnosis of diminutive colorectal polyps, while providing greater alignment with pathology-based surveillance intervals, based on a randomized controlled trial.

These findings suggest that autonomous AI may one day replace histologic assessment of diminutive polyps, reported lead author Roupen Djinbachian, MD, of the Montreal University Hospital Research Center, Montreal, Quebec, Canada, and colleagues.Optical diagnosis of diminutive colorectal polyps has been proposed as a cost-effective alternative to histologic diagnosis, but its implementation in general clinical practice has been hindered by endoscopists’ concerns about incorrect diagnoses, the investigators wrote in Gastroenterology.“AI-based systems (CADx) have been proposed as a solution to these barriers to implementation, with studies showing high adherence to Preservation and Incorporation of Valuable Endoscopic Innovations (PIVI) thresholds when using AI-H,” they wrote. “However, the efficacy and safety of autonomous AI-based diagnostic platforms have not yet been evaluated.”

To address this knowledge gap, Dr. Djinbachian and colleagues conducted a randomized controlled noninferiority trial involving 467 patients, all of whom underwent elective colonoscopies at a single academic institution.

Participants were randomly assigned to one of two groups. The first group received an optical diagnosis of diminutive (1-5 mm) colorectal polyps using an autonomous AI-based CADx system without any human input. The second group had diagnoses performed by endoscopists who used AI-H to make their optical diagnoses.

The primary outcome was the accuracy of optical diagnosis compared with the gold standard of histologic evaluation. Secondarily, the investigators explored associations between pathology-based surveillance intervals and various measures of accuracy, including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV).

The results showed that the accuracy of optical diagnosis for diminutive polyps was similar between the two groups, supporting noninferiority. Autonomous AI achieved an accuracy rate of 77.2%, while the AI-H group had an accuracy of 72.1%, which was not statistically significant (P = .86).

But when it came to pathology-based surveillance intervals, autonomous AI showed a clear advantage; the autonomous AI system achieved a 91.5% agreement rate, compared with 82.1% for the AI-H group (P = .016).

“These findings indicate that autonomous AI not only matches but also surpasses AI-H in accuracy for determining surveillance intervals,” the investigators wrote, noting that this finding highlights the “complexities of human interaction with AI modules where human intervention could lead to worse outcomes.”

Further analysis revealed that the sensitivity of autonomous AI for identifying adenomas was 84.8%, slightly higher than the 83.6% sensitivity of the AI-H group. Specificity was 64.4% for autonomous AI vs 63.8% for AI-H. While PPV was higher in the autonomous AI group (85.6%), compared with the AI-H group (78.6%), NPV was lower for autonomous AI than AI-H (63.0% vs 71.0%).

Dr. Djinbachian and colleagues suggested that future research should focus on larger, multicenter trials to validate these findings and further explore the integration of autonomous AI systems in clinical practice. They also noted that improving AI algorithms to accurately diagnose sessile serrated lesions could enhance the overall effectiveness of AI-based optical diagnosis.

“The performance of autonomous AI in accurately diagnosing diminutive polyps and determining appropriate surveillance intervals suggests that it could play a crucial role in streamlining colorectal cancer screening processes, reducing the burden on pathologists, and potentially lowering healthcare costs,” the investigators concluded.The study was supported by Fujifilm, which had no role in the study design or data analysis. Dr. von Renteln reported additional research funding from Vantage and Fujifilm.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168264</fileName> <TBEID>0C050575.SIG</TBEID> <TBUniqueIdentifier>MD_0C050575</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Gastro_Djinbachian_AI</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240611T140656</QCDate> <firstPublished>20240611T150325</firstPublished> <LastPublished>20240611T150325</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240611T150325</CMSDate> <articleSource>FROM GASTROENTEROLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Autonomous artificial intelligence (AI) can achieve similar accuracy to AI-assisted humans (AI-H) in the optical diagnosis of diminutive colorectal polyps</metaDescription> <articlePDF/> <teaserImage>301890</teaserImage> <teaser>Autonomous AI may one day replace histologic assessment of diminutive polyps.</teaser> <title>Autonomous AI Outperforms Humans in Optical Diagnosis of Colorectal Polyps</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">39702</term> <term>344</term> <term>345</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240129e5.jpg</altRep> <description role="drol:caption">Dr. Jeremy R. Glissen Brown</description> <description role="drol:credit">Duke University Medical Center</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Autonomous AI Outperforms Humans in Optical Diagnosis of Colorectal Polyps</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Autonomous artificial intelligence (AI) can achieve similar accuracy to AI-assisted humans (AI-H) in the optical diagnosis of diminutive colorectal polyps</span>, while providing greater alignment with pathology-based surveillance intervals, based on a randomized controlled trial.</p> <p>These findings suggest that autonomous AI may one day replace histologic assessment of diminutive polyps, reported lead author <a href="https://www.researchgate.net/profile/Roupen-Djinbachian">Roupen Djinbachian, MD</a>, of the Montreal University Hospital Research Center, Montreal, Quebec, Canada, and colleagues.Optical diagnosis of diminutive colorectal polyps has been proposed as a cost-effective alternative to histologic diagnosis, but its implementation in general clinical practice has been hindered by endoscopists’ concerns about incorrect diagnoses, the investigators wrote in<strong> </strong><em><a href="https://www.gastrojournal.org/article/S0016-5085(24)00131-8/fulltext">Gastroenterology</a></em>.“AI-based systems (CADx) have been proposed as a solution to these barriers to implementation, with studies showing high adherence to Preservation and Incorporation of Valuable Endoscopic Innovations (PIVI) thresholds when using AI-H,” they wrote. “However, the efficacy and safety of autonomous AI-based diagnostic platforms have not yet been evaluated.”<br/><br/>To address this knowledge gap, Dr. Djinbachian and colleagues conducted a randomized controlled noninferiority trial involving 467 patients, all of whom underwent elective colonoscopies at a single academic institution.<br/><br/>Participants were randomly assigned to one of two groups. The first group received an optical diagnosis of diminutive (1-5 mm) colorectal polyps using an autonomous AI-based CADx system without any human input. The second group had diagnoses performed by endoscopists who used AI-H to make their optical diagnoses.<br/><br/>The primary outcome was the accuracy of optical diagnosis compared with the gold standard of histologic evaluation. Secondarily, the investigators explored associations between pathology-based surveillance intervals and various measures of accuracy, including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV).<br/><br/>The results showed that the accuracy of optical diagnosis for diminutive polyps was similar between the two groups, supporting noninferiority. Autonomous AI achieved an accuracy rate of 77.2%, while the AI-H group had an accuracy of 72.1%, which was not statistically significant (<em>P</em> = .86). <br/><br/>But when it came to pathology-based surveillance intervals, autonomous AI showed a clear advantage; the autonomous AI system achieved a 91.5% agreement rate, compared with 82.1% for the AI-H group (<em>P</em> = .016).<br/><br/>“These findings indicate that autonomous AI not only matches but also surpasses AI-H in accuracy for determining surveillance intervals,” the investigators wrote, noting that this finding highlights the “complexities of human interaction with AI modules where human intervention could lead to worse outcomes.”<br/><br/>Further analysis revealed that the sensitivity of autonomous AI for identifying adenomas was 84.8%, slightly higher than the 83.6% sensitivity of the AI-H group. Specificity was 64.4% for autonomous AI vs 63.8% for AI-H. While PPV was higher in the autonomous AI group (85.6%), compared with the AI-H group (78.6%), NPV was lower for autonomous AI than AI-H (63.0% vs 71.0%).<br/><br/>Dr. Djinbachian and colleagues suggested that future research should focus on larger, multicenter trials to validate these findings and further explore the integration of autonomous AI systems in clinical practice. They also noted that improving AI algorithms to accurately diagnose sessile serrated lesions could enhance the overall effectiveness of AI-based optical diagnosis.<br/><br/>“The performance of autonomous AI in accurately diagnosing diminutive polyps and determining appropriate surveillance intervals suggests that it could play a crucial role in streamlining colorectal cancer screening processes, reducing the burden on pathologists, and potentially lowering healthcare costs,” the investigators concluded.The study was supported by Fujifilm, which had no role in the study design or data analysis. Dr. von Renteln reported additional research funding from Vantage and Fujifilm.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>views</itemRole> <itemClass>text</itemClass> <title>‘Automatic’ CADx in Colonoscopy</title> <deck/> </itemMeta> <itemContent> <p>In the era of computer vision for endoscopy and colonoscopy, current paradigms rely on AI as a co-pilot or second observer, with the physician serving as the final arbiter in procedure-related decision-making. This study by Djinbachian and Haumesser et al brings up the interesting wrinkle of autonomous AI as a potentially superior (or noninferior) option in narrow, task-specific use cases.</p> <p>In this study, human input from the endoscopist after CADx diagnosis led to lower agreement between the AI-predicted diagnosis and corresponding surveillance intervals; human oversight more often incorrectly changed the resultant diagnosis and led to shorter than recommended surveillance intervals.<br/><br/>This study offers a small but very important update to the growing body of literature on CADx in colonoscopy. So far, prospective validation of CADx compared with the human eye for in-situ diagnosis of polyps has provided mixed results. This study is one of the first to examine the potential role of “automatic” CADx without additional human input and sheds light on the importance of the AI-human hybrid in medical care. How do the ways in which humans interact with the user interface and output of AI lead to changes in outcome? How can we optimize the AI-human interaction in order to provide optimal results?<br/><br/>[[{"fid":"301890","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jeremy R. Glissen Brown, Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina","field_file_image_credit[und][0][value]":"Duke University Medical Center","field_file_image_caption[und][0][value]":"Dr. Jeremy R. Glissen Brown"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]In this case, the suggestion is that less is more when it comes to human interference with optical diagnosis, but further research is needed on how to best optimize this important relationship as well as how AI might (or might not) support diagnose-and-leave and diagnose-and-discard strategies in the United States and worldwide.</p> <p><em> <em>Jeremy R. Glissen Brown is an assistant professor in the Department of Internal Medicine and Division of Gastroenterology at Duke University Medical Center, Durham, North Carolina. He has served as a consultant for Medtronic and Olympus, and on the advisory board for Odin Vision.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Targeting Enteroendocrine Cells Could Hold Promise for IBD

Valuable Insights into Small Intestine Dysmotility
Article Type
Changed
Tue, 06/11/2024 - 10:48

Colitis-induced small intestinal hypomotility is closely linked to the loss of enteroendocrine cells (EECs) in mice, revealing a potential therapeutic strategy for patients with inflammatory bowel disease (IBD), according to investigators.

These findings suggest that restoring EEC function could alleviate some of the more general abdominal symptoms associated with IBD, reported lead author Zachariah Raouf, MD, of Johns Hopkins University School of Medicine, Baltimore, and colleagues.

“The symptoms experienced by patients with IBD, especially ulcerative colitis, may include those that are colonic in nature, such as bloody stools, abdominal pain, and weight loss, as well as those that are more general in nature, such as severe nausea and abdominal bloating,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology . “Although the first set of symptoms may be attributable to the effects of colonic inflammation itself, those that are more vague seem to overlap with the symptoms that patients with small intestinal dysmotility experience, such as occur in response to medications, or diabetes.”

Supporting this notion, several previous studies have reported the onset of intestinal dysmotility in experimental models of colitis, which is believed to be caused by impaired enteric nervous system function. But the precise mechanisms behind the impaired intestinal motility observed in colitis patients remain unclear.

To learn more, Dr. Raouf and colleagues conducted experiments involving three groups of mice: wild-type mice, mice genetically engineered to overexpress EECs, and mice lacking EECs.

To induce colitis, the mice were administered dextran sulfate sodium (DSS) in drinking water at concentrations of 2.5% or 5% for 7 days. Small intestinal motility was evaluated by measuring the transit of fluorescein isothiocyanate (FITC)-dextran. Immunohistochemical analyses were conducted to assess EEC number and differentiation, while quantitative reverse-transcriptase polymerase chain reaction was used to examine the expression of genes related to serotonin synthesis and transport.

The researchers examined colon length and signs of colonic inflammation, monitored weight loss, and measured the expression of proinflammatory cytokines. Histological analyses of colon and small intestine tissues were performed to further understand the effects of colitis. The presence and number of EEC cells was evaluated using chromogranin A (ChgA) staining, while apoptosis in EECs was measured via TUNEL staining. The expression of serotonin-related genes was also assessed.

These experiments revealed that DSS-induced colitis led to significant small-bowel hypomotility and a reduction in EEC density. Of note, genetic overexpression of EECs or treatment with prucalopride, a 5-hydroxytryptamine receptor 4 agonist, improved small intestinal motility.

“It is noteworthy that there were no significant changes in the density of other intestinal epithelial cells, or in other cell types that are linked to motility, such as enteric glia and neurons, suggesting the specificity of the effect,” the investigators wrote. “Importantly, treatment with a serotonin agonist ameliorated the colitis-induced, small-bowel hypomotility and attenuated the severity of colitis, providing potential clinical relevance of the current findings. Taken together, these results identify mechanisms to explain the intestinal hypomotility observed in the setting of colitis.”Dr. Raouf and colleagues called for human clinical trials to their findings. Specifically, they suggested exploring therapies targeting enteroendocrine cells or serotonin pathways and examining the role of different EEC types in gut motility during inflammation. The study was supported by the National Institutes of Health. The investigators disclosed no conflicts of interest.

Body

Inflammatory bowel disease (IBD) typically manifests with colonic symptoms but is also associated with intestinal inflammation and dysmotility of the small intestine. Clinical research debates whether IBD causes small intestine hypermotility or hypomotility, but these motility dysfunctions are often attributed to alterations of the gut’s intrinsic nervous system.

Dr. Raouf and colleagues focus on the role of enteroendocrine cells, an epithelial cell subtype with neuron-like features that secrete serotonin, one of the most important regulators of intestinal motility. Their population is reduced in colitis, and the subsequent alteration of serotonin signaling induces small intestine dysmotility. The observed loss of enteroendocrine cells in the small bowel may result from low-grade local inflammation increasing enteroendocrine cell apoptosis, or impaired gene expression in their differentiation pathways. However, more research is required to elucidate the underlying mechanisms of this loss.

[[{"fid":"301892","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"}},"attributes":{"alt":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","class":"media-element file-medstat-image-flush-right","data-delta":"1"}}]]Nevertheless, their findings provide valuable insights into small intestine dysmotility associated with IBD pathologies and suggest a therapeutic approach based on a pharmacologic serotonin agonist. Treatment with prucalopride, a serotonin type 4 receptor agonist already used in clinics with minimal adverse effects, restores small intestine motility and offers therapeutic benefits. Although the results are promising in DSS models of colitis, the observed improvement in small intestinal motility needs to be confirmed in IBD patients.

This study enhances our understanding of the small intestine dysfunction associated with colitis and raises the exciting possibility of enteroendocrine cell-based therapeutic approaches in IBD.

Jacques A. Gonzales, PhD, is a postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing. He has no conflicts of interest.

Publications
Topics
Sections
Body

Inflammatory bowel disease (IBD) typically manifests with colonic symptoms but is also associated with intestinal inflammation and dysmotility of the small intestine. Clinical research debates whether IBD causes small intestine hypermotility or hypomotility, but these motility dysfunctions are often attributed to alterations of the gut’s intrinsic nervous system.

Dr. Raouf and colleagues focus on the role of enteroendocrine cells, an epithelial cell subtype with neuron-like features that secrete serotonin, one of the most important regulators of intestinal motility. Their population is reduced in colitis, and the subsequent alteration of serotonin signaling induces small intestine dysmotility. The observed loss of enteroendocrine cells in the small bowel may result from low-grade local inflammation increasing enteroendocrine cell apoptosis, or impaired gene expression in their differentiation pathways. However, more research is required to elucidate the underlying mechanisms of this loss.

[[{"fid":"301892","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"}},"attributes":{"alt":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","class":"media-element file-medstat-image-flush-right","data-delta":"1"}}]]Nevertheless, their findings provide valuable insights into small intestine dysmotility associated with IBD pathologies and suggest a therapeutic approach based on a pharmacologic serotonin agonist. Treatment with prucalopride, a serotonin type 4 receptor agonist already used in clinics with minimal adverse effects, restores small intestine motility and offers therapeutic benefits. Although the results are promising in DSS models of colitis, the observed improvement in small intestinal motility needs to be confirmed in IBD patients.

This study enhances our understanding of the small intestine dysfunction associated with colitis and raises the exciting possibility of enteroendocrine cell-based therapeutic approaches in IBD.

Jacques A. Gonzales, PhD, is a postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing. He has no conflicts of interest.

Body

Inflammatory bowel disease (IBD) typically manifests with colonic symptoms but is also associated with intestinal inflammation and dysmotility of the small intestine. Clinical research debates whether IBD causes small intestine hypermotility or hypomotility, but these motility dysfunctions are often attributed to alterations of the gut’s intrinsic nervous system.

Dr. Raouf and colleagues focus on the role of enteroendocrine cells, an epithelial cell subtype with neuron-like features that secrete serotonin, one of the most important regulators of intestinal motility. Their population is reduced in colitis, and the subsequent alteration of serotonin signaling induces small intestine dysmotility. The observed loss of enteroendocrine cells in the small bowel may result from low-grade local inflammation increasing enteroendocrine cell apoptosis, or impaired gene expression in their differentiation pathways. However, more research is required to elucidate the underlying mechanisms of this loss.

[[{"fid":"301892","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"},"type":"media","field_deltas":{"1":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"}},"attributes":{"alt":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","class":"media-element file-medstat-image-flush-right","data-delta":"1"}}]]Nevertheless, their findings provide valuable insights into small intestine dysmotility associated with IBD pathologies and suggest a therapeutic approach based on a pharmacologic serotonin agonist. Treatment with prucalopride, a serotonin type 4 receptor agonist already used in clinics with minimal adverse effects, restores small intestine motility and offers therapeutic benefits. Although the results are promising in DSS models of colitis, the observed improvement in small intestinal motility needs to be confirmed in IBD patients.

This study enhances our understanding of the small intestine dysfunction associated with colitis and raises the exciting possibility of enteroendocrine cell-based therapeutic approaches in IBD.

Jacques A. Gonzales, PhD, is a postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing. He has no conflicts of interest.

Title
Valuable Insights into Small Intestine Dysmotility
Valuable Insights into Small Intestine Dysmotility

Colitis-induced small intestinal hypomotility is closely linked to the loss of enteroendocrine cells (EECs) in mice, revealing a potential therapeutic strategy for patients with inflammatory bowel disease (IBD), according to investigators.

These findings suggest that restoring EEC function could alleviate some of the more general abdominal symptoms associated with IBD, reported lead author Zachariah Raouf, MD, of Johns Hopkins University School of Medicine, Baltimore, and colleagues.

“The symptoms experienced by patients with IBD, especially ulcerative colitis, may include those that are colonic in nature, such as bloody stools, abdominal pain, and weight loss, as well as those that are more general in nature, such as severe nausea and abdominal bloating,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology . “Although the first set of symptoms may be attributable to the effects of colonic inflammation itself, those that are more vague seem to overlap with the symptoms that patients with small intestinal dysmotility experience, such as occur in response to medications, or diabetes.”

Supporting this notion, several previous studies have reported the onset of intestinal dysmotility in experimental models of colitis, which is believed to be caused by impaired enteric nervous system function. But the precise mechanisms behind the impaired intestinal motility observed in colitis patients remain unclear.

To learn more, Dr. Raouf and colleagues conducted experiments involving three groups of mice: wild-type mice, mice genetically engineered to overexpress EECs, and mice lacking EECs.

To induce colitis, the mice were administered dextran sulfate sodium (DSS) in drinking water at concentrations of 2.5% or 5% for 7 days. Small intestinal motility was evaluated by measuring the transit of fluorescein isothiocyanate (FITC)-dextran. Immunohistochemical analyses were conducted to assess EEC number and differentiation, while quantitative reverse-transcriptase polymerase chain reaction was used to examine the expression of genes related to serotonin synthesis and transport.

The researchers examined colon length and signs of colonic inflammation, monitored weight loss, and measured the expression of proinflammatory cytokines. Histological analyses of colon and small intestine tissues were performed to further understand the effects of colitis. The presence and number of EEC cells was evaluated using chromogranin A (ChgA) staining, while apoptosis in EECs was measured via TUNEL staining. The expression of serotonin-related genes was also assessed.

These experiments revealed that DSS-induced colitis led to significant small-bowel hypomotility and a reduction in EEC density. Of note, genetic overexpression of EECs or treatment with prucalopride, a 5-hydroxytryptamine receptor 4 agonist, improved small intestinal motility.

“It is noteworthy that there were no significant changes in the density of other intestinal epithelial cells, or in other cell types that are linked to motility, such as enteric glia and neurons, suggesting the specificity of the effect,” the investigators wrote. “Importantly, treatment with a serotonin agonist ameliorated the colitis-induced, small-bowel hypomotility and attenuated the severity of colitis, providing potential clinical relevance of the current findings. Taken together, these results identify mechanisms to explain the intestinal hypomotility observed in the setting of colitis.”Dr. Raouf and colleagues called for human clinical trials to their findings. Specifically, they suggested exploring therapies targeting enteroendocrine cells or serotonin pathways and examining the role of different EEC types in gut motility during inflammation. The study was supported by the National Institutes of Health. The investigators disclosed no conflicts of interest.

Colitis-induced small intestinal hypomotility is closely linked to the loss of enteroendocrine cells (EECs) in mice, revealing a potential therapeutic strategy for patients with inflammatory bowel disease (IBD), according to investigators.

These findings suggest that restoring EEC function could alleviate some of the more general abdominal symptoms associated with IBD, reported lead author Zachariah Raouf, MD, of Johns Hopkins University School of Medicine, Baltimore, and colleagues.

“The symptoms experienced by patients with IBD, especially ulcerative colitis, may include those that are colonic in nature, such as bloody stools, abdominal pain, and weight loss, as well as those that are more general in nature, such as severe nausea and abdominal bloating,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology . “Although the first set of symptoms may be attributable to the effects of colonic inflammation itself, those that are more vague seem to overlap with the symptoms that patients with small intestinal dysmotility experience, such as occur in response to medications, or diabetes.”

Supporting this notion, several previous studies have reported the onset of intestinal dysmotility in experimental models of colitis, which is believed to be caused by impaired enteric nervous system function. But the precise mechanisms behind the impaired intestinal motility observed in colitis patients remain unclear.

To learn more, Dr. Raouf and colleagues conducted experiments involving three groups of mice: wild-type mice, mice genetically engineered to overexpress EECs, and mice lacking EECs.

To induce colitis, the mice were administered dextran sulfate sodium (DSS) in drinking water at concentrations of 2.5% or 5% for 7 days. Small intestinal motility was evaluated by measuring the transit of fluorescein isothiocyanate (FITC)-dextran. Immunohistochemical analyses were conducted to assess EEC number and differentiation, while quantitative reverse-transcriptase polymerase chain reaction was used to examine the expression of genes related to serotonin synthesis and transport.

The researchers examined colon length and signs of colonic inflammation, monitored weight loss, and measured the expression of proinflammatory cytokines. Histological analyses of colon and small intestine tissues were performed to further understand the effects of colitis. The presence and number of EEC cells was evaluated using chromogranin A (ChgA) staining, while apoptosis in EECs was measured via TUNEL staining. The expression of serotonin-related genes was also assessed.

These experiments revealed that DSS-induced colitis led to significant small-bowel hypomotility and a reduction in EEC density. Of note, genetic overexpression of EECs or treatment with prucalopride, a 5-hydroxytryptamine receptor 4 agonist, improved small intestinal motility.

“It is noteworthy that there were no significant changes in the density of other intestinal epithelial cells, or in other cell types that are linked to motility, such as enteric glia and neurons, suggesting the specificity of the effect,” the investigators wrote. “Importantly, treatment with a serotonin agonist ameliorated the colitis-induced, small-bowel hypomotility and attenuated the severity of colitis, providing potential clinical relevance of the current findings. Taken together, these results identify mechanisms to explain the intestinal hypomotility observed in the setting of colitis.”Dr. Raouf and colleagues called for human clinical trials to their findings. Specifically, they suggested exploring therapies targeting enteroendocrine cells or serotonin pathways and examining the role of different EEC types in gut motility during inflammation. The study was supported by the National Institutes of Health. The investigators disclosed no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168272</fileName> <TBEID>0C050583.SIG</TBEID> <TBUniqueIdentifier>MD_0C050583</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CMGH_Raouf_colitis dysmotility</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240611T104353</QCDate> <firstPublished>20240611T104535</firstPublished> <LastPublished>20240611T104535</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240611T104535</CMSDate> <articleSource>FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Colitis-induced small intestinal hypomotility is closely linked to the loss of enteroendocrine cells (EECs) in mice, revealing a potential therapeutic strategy </metaDescription> <articlePDF/> <teaserImage>301892</teaserImage> <teaser>The findings suggest that restoring EEC function could alleviate some abdominal symptoms associated with IBD.</teaser> <title>Targeting Enteroendocrine Cells Could Hold Promise for IBD</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">345</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240129e7.jpg</altRep> <description role="drol:caption">Dr. Jacques A. Gonzales</description> <description role="drol:credit">Dr. Gonzales</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Targeting Enteroendocrine Cells Could Hold Promise for IBD</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Colitis-induced small intestinal hypomotility is closely linked to the loss of enteroendocrine cells (EECs) in mice, revealing a potential therapeutic strategy for patients with inflammatory bowel disease (IBD)</span>, according to investigators.</p> <p>These findings suggest that restoring EEC function could alleviate some of the more general abdominal symptoms associated with IBD, reported lead author <a href="https://uk.linkedin.com/in/zachariah-zac-raouf-23626a211?original_referer=https%3A%2F%2Fwww.google.com%2F">Zachariah Raouf, MD</a>, of Johns Hopkins University School of Medicine, Baltimore, and colleagues.<br/><br/>“The symptoms experienced by patients with IBD, especially ulcerative colitis, may include those that are colonic in nature, such as bloody stools, abdominal pain, and weight loss, as well as those that are more general in nature, such as severe nausea and abdominal bloating,” the investigators <span class="Hyperlink"><a href="https://www.cmghjournal.org/article/S2352-345X(24)00051-1/fulltext">wrote</a></span> in<strong> </strong><em>Cellular and Molecular Gastroenterology and Hepatology</em> . “Although the first set of symptoms may be attributable to the effects of colonic inflammation itself, those that are more vague seem to overlap with the symptoms that patients with small intestinal dysmotility experience, such as occur in response to medications, or diabetes.”<br/><br/>Supporting this notion, several previous studies have reported the onset of intestinal dysmotility in experimental models of colitis, which is believed to be caused by impaired enteric nervous system function. But the precise mechanisms behind the impaired intestinal motility observed in colitis patients remain unclear.<br/><br/>To learn more, Dr. Raouf and colleagues conducted experiments involving three groups of mice: wild-type mice, mice genetically engineered to overexpress EECs, and mice lacking EECs.<br/><br/>To induce colitis, the mice were administered dextran sulfate sodium (DSS) in drinking water at concentrations of 2.5% or 5% for 7 days. Small intestinal motility was evaluated by measuring the transit of fluorescein isothiocyanate (FITC)-dextran. Immunohistochemical analyses were conducted to assess EEC number and differentiation, while quantitative reverse-transcriptase polymerase chain reaction was used to examine the expression of genes related to serotonin synthesis and transport.<br/><br/>The researchers examined colon length and signs of colonic inflammation, monitored weight loss, and measured the expression of proinflammatory cytokines. Histological analyses of colon and small intestine tissues were performed to further understand the effects of colitis. The presence and number of EEC cells was evaluated using chromogranin A (ChgA) staining, while apoptosis in EECs was measured via TUNEL staining. The expression of serotonin-related genes was also assessed.<br/><br/>These experiments revealed that DSS-induced colitis led to significant small-bowel hypomotility and a reduction in EEC density. Of note, genetic overexpression of EECs or treatment with prucalopride, a 5-hydroxytryptamine receptor 4 agonist, improved small intestinal motility.<br/><br/>“It is noteworthy that there were no significant changes in the density of other intestinal epithelial cells, or in other cell types that are linked to motility, such as enteric glia and neurons, suggesting the specificity of the effect,” the investigators wrote. “Importantly, treatment with a serotonin agonist ameliorated the colitis-induced, small-bowel hypomotility and attenuated the severity of colitis, providing potential clinical relevance of the current findings. Taken together, these results identify mechanisms to explain the intestinal hypomotility observed in the setting of colitis.”Dr. Raouf and colleagues called for human clinical trials to their findings. Specifically, they suggested exploring therapies targeting enteroendocrine cells or serotonin pathways and examining the role of different EEC types in gut motility during inflammation. The study was supported by the National Institutes of Health. The investigators disclosed no conflicts of interest.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>views</itemRole> <itemClass>text</itemClass> <title>Valuable Insights into Small Intestine Dysmotility</title> <deck/> </itemMeta> <itemContent> <p>Inflammatory bowel disease (IBD) typically manifests with colonic symptoms but is also associated with intestinal inflammation and dysmotility of the small intestine. Clinical research debates whether IBD causes small intestine hypermotility or hypomotility, but these motility dysfunctions are often attributed to alterations of the gut’s intrinsic nervous system.</p> <p>Dr. Raouf and colleagues focus on the role of enteroendocrine cells, an epithelial cell subtype with neuron-like features that secrete serotonin, one of the most important regulators of intestinal motility. Their population is reduced in colitis, and the subsequent alteration of serotonin signaling induces small intestine dysmotility. The observed loss of enteroendocrine cells in the small bowel may result from low-grade local inflammation increasing enteroendocrine cell apoptosis, or impaired gene expression in their differentiation pathways. However, more research is required to elucidate the underlying mechanisms of this loss.<br/><br/>[[{"fid":"301892","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Jacques A. Gonzales, postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing, Michigan","field_file_image_credit[und][0][value]":"Dr. Gonzales","field_file_image_caption[und][0][value]":"Dr. Jacques A. Gonzales"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]Nevertheless, their findings provide valuable insights into small intestine dysmotility associated with IBD pathologies and suggest a therapeutic approach based on a pharmacologic serotonin agonist. Treatment with prucalopride, a serotonin type 4 receptor agonist already used in clinics with minimal adverse effects, restores small intestine motility and offers therapeutic benefits. Although the results are promising in DSS models of colitis, the observed improvement in small intestinal motility needs to be confirmed in IBD patients.<br/><br/>This study enhances our understanding of the small intestine dysfunction associated with colitis and raises the exciting possibility of enteroendocrine cell-based therapeutic approaches in IBD.</p> <p><em> <em>Jacques A. Gonzales, PhD, is a postdoctoral fellow in the Gulbransen laboratory at Michigan State University, East Lansing. He has no conflicts of interest.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

High-Alcohol Intake in MASLD Increases Risk of Cirrhosis

Critical Need for Early Tx of High-Risk Alcohol Use
Article Type
Changed
Fri, 05/10/2024 - 13:41

One in nine patients with steatotic liver disease reported concurrent alcohol use, and more than 11% reported high-risk consumption, a national study of more than a million US veterans found.

Moreover, the combination of steatotic liver disease and high-risk alcohol intake carried a more than 43% higher long-term risk of liver cirrhosis compared with no alcohol use, according to researchers led by Robert J. Wong, MD, MS, of the Division of Gastroenterology and Hepatology, Veterans Affairs Healthcare System Palo Alto, at Stanford University School of Medicine in Palo Alto, California.

However, the study found that “reducing alcohol use lowers risk of cirrhosis, emphasizing the importance of timely alcohol use assessment and early interventions to address high-risk alcohol use in steatotic liver disease,” Dr. Wong and associates wrote in Gastroenterology.

Although concurrent moderate to heavy alcohol intake would be expected to lead more rapidly to liver disease progression, the existing literature has been conflicting, the authors noted. Several studies have even found moderate alcohol associated with a lower risk of advanced liver disease among MASLD patients, including that by Dunn et al. .
 

The Study

MASLD patients were identified through the US Veterans Affairs Corporate Data Warehouse from January 1, 2010, through December 31, 2017, with follow-up through December 31, 2022.

Alcohol use was assessed by Alcohol Use Disorders Identification Test–Concise (AUDIT-C) scores and was categorized as follows: no alcohol (AUDIT-C = 0), low-risk alcohol use (AUDIT-C 1-2 for women and 1–3 for men), and high-risk alcohol (AUDIT-C ≥ 3 for women and ≥ 4 for men).

Among the 1,156,189 veterans with MASLD, 54.2% reported no alcohol, 34.6% low-risk alcohol, and 11.2% high-risk alcohol use. In median follow-up of nine to 10 years, incidence rates of cirrhosis were .53 per 100 person-years for no use, .42 for low-risk use, and .76 for high-risk use.

In contrast to patients with baseline high-risk alcohol intake who reported no change in use, those who decreased their alcohol intake during follow-up experienced a 39% reduction in the long-term risk of cirrhosis, for a hazard ratio of .61 (95% CI, .45-.83, P < .01).

tusludastawushechuhauajarosuframachuslejurodrowraboslusibabofratrathijubraniphudeswopohokasajestusathodruswiphisliprodorastimuspasoslislauostesholephububraswugikitrebriwrostodoclislefruspotrecraslucugerecredrafrulephates
Dr. Robert J. Wong


About 70% of patients were non-Hispanic Whites and more than 90% were male in all consumption categories. The no-alcohol group was older than the high-risk alcohol group: 64 years vs 59.9 years (P < .0001). Compared with the high-risk alcohol group, the no-alcohol group had a significantly greater proportion of comorbid diabetes (62.3% vs 42.5%), hypertension (77.9% vs 69.1%), or cardiovascular disease (40.2% vs 25.9%, P < .0001 for all comparisons).

In a significant study observation, fewer than 5% of patients with high-risk use received behavioral or pharmacologic therapy and of those who did, most were referred for or received treatment at or near the time of cirrhosis diagnosis. “This highlights a major gap in linking patients with high-risk alcohol use to appropriate behavioral or pharmacologic therapy in a timely manner and may reflect missed opportunities to prevent further alcohol-related morbidity and mortality,” Dr. Wong and colleagues wrote.

They called for studies of novel interventions for timely assessment of alcohol use with linkage to addiction services. They cited the need to understand the interaction between levels of alcohol use and underlying MASLD, adding, “More research is also needed to understand whether this interaction varies across different populations.”

This study received no specific funding. Dr. Wong reported funding through his institution from Gilead Sciences, Exact Sciences, and Thera Technologies.

Body

 

Recent consensus in defining metabolic dysfunction-associated steatotic liver disease (MASLD) has raised awareness for the combined impact of cardiometabolic risk factors and alcohol consumption on liver disease progression. This study by Wong et al. highlights the undeniable influence of high-risk alcohol use on the development of advanced liver disease.

In a national cohort of over 1 million US veterans with steatotic liver disease (SLD), patients with high-risk alcohol use based on AUDIT-C assessment exhibited > 43% greater risk of cirrhosis compared to those with no alcohol use. The relationship between alcohol and liver severity in SLD was observed even after excluding patients meeting classification for predominant alcohol-associated liver disease. While increased alcohol use was associated with increased incidence of cirrhosis, decreased alcohol use led to a notable 39% reduction in cirrhosis risk over time.

Reducing alcohol consumption remains best practice guidelines for mitigating risk of progression in steatotic liver disease. However, results of this study emphasize the critical need for early identification and treatment of high-risk alcohol use in all patients with SLD. While universal recommendations for alcohol abstinence provides pragmatic implementation, there is a significant need to better understand the interaction of specific metabolic risk factors and patterns of alcohol use across the spectrum of MetALD to guide personalized recommendations for patient education and management.[[{"fid":"301427","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"}},"attributes":{"alt":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]

Further research using prospective clinical trial design is needed to evaluate the interplay of alcohol consumption and metabolic risk factors across variable age, sex, genetics, and environmental exposures that are increasingly being recognized as vital drivers of health and disease.

Tiffany Wu, MD, MS, is a fellow in Transplant Hepatology at Mayo Clinic in Rochester, Minnesota. She has no conflicts.

Publications
Topics
Sections
Body

 

Recent consensus in defining metabolic dysfunction-associated steatotic liver disease (MASLD) has raised awareness for the combined impact of cardiometabolic risk factors and alcohol consumption on liver disease progression. This study by Wong et al. highlights the undeniable influence of high-risk alcohol use on the development of advanced liver disease.

In a national cohort of over 1 million US veterans with steatotic liver disease (SLD), patients with high-risk alcohol use based on AUDIT-C assessment exhibited > 43% greater risk of cirrhosis compared to those with no alcohol use. The relationship between alcohol and liver severity in SLD was observed even after excluding patients meeting classification for predominant alcohol-associated liver disease. While increased alcohol use was associated with increased incidence of cirrhosis, decreased alcohol use led to a notable 39% reduction in cirrhosis risk over time.

Reducing alcohol consumption remains best practice guidelines for mitigating risk of progression in steatotic liver disease. However, results of this study emphasize the critical need for early identification and treatment of high-risk alcohol use in all patients with SLD. While universal recommendations for alcohol abstinence provides pragmatic implementation, there is a significant need to better understand the interaction of specific metabolic risk factors and patterns of alcohol use across the spectrum of MetALD to guide personalized recommendations for patient education and management.[[{"fid":"301427","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"}},"attributes":{"alt":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]

Further research using prospective clinical trial design is needed to evaluate the interplay of alcohol consumption and metabolic risk factors across variable age, sex, genetics, and environmental exposures that are increasingly being recognized as vital drivers of health and disease.

Tiffany Wu, MD, MS, is a fellow in Transplant Hepatology at Mayo Clinic in Rochester, Minnesota. She has no conflicts.

Body

 

Recent consensus in defining metabolic dysfunction-associated steatotic liver disease (MASLD) has raised awareness for the combined impact of cardiometabolic risk factors and alcohol consumption on liver disease progression. This study by Wong et al. highlights the undeniable influence of high-risk alcohol use on the development of advanced liver disease.

In a national cohort of over 1 million US veterans with steatotic liver disease (SLD), patients with high-risk alcohol use based on AUDIT-C assessment exhibited > 43% greater risk of cirrhosis compared to those with no alcohol use. The relationship between alcohol and liver severity in SLD was observed even after excluding patients meeting classification for predominant alcohol-associated liver disease. While increased alcohol use was associated with increased incidence of cirrhosis, decreased alcohol use led to a notable 39% reduction in cirrhosis risk over time.

Reducing alcohol consumption remains best practice guidelines for mitigating risk of progression in steatotic liver disease. However, results of this study emphasize the critical need for early identification and treatment of high-risk alcohol use in all patients with SLD. While universal recommendations for alcohol abstinence provides pragmatic implementation, there is a significant need to better understand the interaction of specific metabolic risk factors and patterns of alcohol use across the spectrum of MetALD to guide personalized recommendations for patient education and management.[[{"fid":"301427","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"}},"attributes":{"alt":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]

Further research using prospective clinical trial design is needed to evaluate the interplay of alcohol consumption and metabolic risk factors across variable age, sex, genetics, and environmental exposures that are increasingly being recognized as vital drivers of health and disease.

Tiffany Wu, MD, MS, is a fellow in Transplant Hepatology at Mayo Clinic in Rochester, Minnesota. She has no conflicts.

Title
Critical Need for Early Tx of High-Risk Alcohol Use
Critical Need for Early Tx of High-Risk Alcohol Use

One in nine patients with steatotic liver disease reported concurrent alcohol use, and more than 11% reported high-risk consumption, a national study of more than a million US veterans found.

Moreover, the combination of steatotic liver disease and high-risk alcohol intake carried a more than 43% higher long-term risk of liver cirrhosis compared with no alcohol use, according to researchers led by Robert J. Wong, MD, MS, of the Division of Gastroenterology and Hepatology, Veterans Affairs Healthcare System Palo Alto, at Stanford University School of Medicine in Palo Alto, California.

However, the study found that “reducing alcohol use lowers risk of cirrhosis, emphasizing the importance of timely alcohol use assessment and early interventions to address high-risk alcohol use in steatotic liver disease,” Dr. Wong and associates wrote in Gastroenterology.

Although concurrent moderate to heavy alcohol intake would be expected to lead more rapidly to liver disease progression, the existing literature has been conflicting, the authors noted. Several studies have even found moderate alcohol associated with a lower risk of advanced liver disease among MASLD patients, including that by Dunn et al. .
 

The Study

MASLD patients were identified through the US Veterans Affairs Corporate Data Warehouse from January 1, 2010, through December 31, 2017, with follow-up through December 31, 2022.

Alcohol use was assessed by Alcohol Use Disorders Identification Test–Concise (AUDIT-C) scores and was categorized as follows: no alcohol (AUDIT-C = 0), low-risk alcohol use (AUDIT-C 1-2 for women and 1–3 for men), and high-risk alcohol (AUDIT-C ≥ 3 for women and ≥ 4 for men).

Among the 1,156,189 veterans with MASLD, 54.2% reported no alcohol, 34.6% low-risk alcohol, and 11.2% high-risk alcohol use. In median follow-up of nine to 10 years, incidence rates of cirrhosis were .53 per 100 person-years for no use, .42 for low-risk use, and .76 for high-risk use.

In contrast to patients with baseline high-risk alcohol intake who reported no change in use, those who decreased their alcohol intake during follow-up experienced a 39% reduction in the long-term risk of cirrhosis, for a hazard ratio of .61 (95% CI, .45-.83, P < .01).

tusludastawushechuhauajarosuframachuslejurodrowraboslusibabofratrathijubraniphudeswopohokasajestusathodruswiphisliprodorastimuspasoslislauostesholephububraswugikitrebriwrostodoclislefruspotrecraslucugerecredrafrulephates
Dr. Robert J. Wong


About 70% of patients were non-Hispanic Whites and more than 90% were male in all consumption categories. The no-alcohol group was older than the high-risk alcohol group: 64 years vs 59.9 years (P < .0001). Compared with the high-risk alcohol group, the no-alcohol group had a significantly greater proportion of comorbid diabetes (62.3% vs 42.5%), hypertension (77.9% vs 69.1%), or cardiovascular disease (40.2% vs 25.9%, P < .0001 for all comparisons).

In a significant study observation, fewer than 5% of patients with high-risk use received behavioral or pharmacologic therapy and of those who did, most were referred for or received treatment at or near the time of cirrhosis diagnosis. “This highlights a major gap in linking patients with high-risk alcohol use to appropriate behavioral or pharmacologic therapy in a timely manner and may reflect missed opportunities to prevent further alcohol-related morbidity and mortality,” Dr. Wong and colleagues wrote.

They called for studies of novel interventions for timely assessment of alcohol use with linkage to addiction services. They cited the need to understand the interaction between levels of alcohol use and underlying MASLD, adding, “More research is also needed to understand whether this interaction varies across different populations.”

This study received no specific funding. Dr. Wong reported funding through his institution from Gilead Sciences, Exact Sciences, and Thera Technologies.

One in nine patients with steatotic liver disease reported concurrent alcohol use, and more than 11% reported high-risk consumption, a national study of more than a million US veterans found.

Moreover, the combination of steatotic liver disease and high-risk alcohol intake carried a more than 43% higher long-term risk of liver cirrhosis compared with no alcohol use, according to researchers led by Robert J. Wong, MD, MS, of the Division of Gastroenterology and Hepatology, Veterans Affairs Healthcare System Palo Alto, at Stanford University School of Medicine in Palo Alto, California.

However, the study found that “reducing alcohol use lowers risk of cirrhosis, emphasizing the importance of timely alcohol use assessment and early interventions to address high-risk alcohol use in steatotic liver disease,” Dr. Wong and associates wrote in Gastroenterology.

Although concurrent moderate to heavy alcohol intake would be expected to lead more rapidly to liver disease progression, the existing literature has been conflicting, the authors noted. Several studies have even found moderate alcohol associated with a lower risk of advanced liver disease among MASLD patients, including that by Dunn et al. .
 

The Study

MASLD patients were identified through the US Veterans Affairs Corporate Data Warehouse from January 1, 2010, through December 31, 2017, with follow-up through December 31, 2022.

Alcohol use was assessed by Alcohol Use Disorders Identification Test–Concise (AUDIT-C) scores and was categorized as follows: no alcohol (AUDIT-C = 0), low-risk alcohol use (AUDIT-C 1-2 for women and 1–3 for men), and high-risk alcohol (AUDIT-C ≥ 3 for women and ≥ 4 for men).

Among the 1,156,189 veterans with MASLD, 54.2% reported no alcohol, 34.6% low-risk alcohol, and 11.2% high-risk alcohol use. In median follow-up of nine to 10 years, incidence rates of cirrhosis were .53 per 100 person-years for no use, .42 for low-risk use, and .76 for high-risk use.

In contrast to patients with baseline high-risk alcohol intake who reported no change in use, those who decreased their alcohol intake during follow-up experienced a 39% reduction in the long-term risk of cirrhosis, for a hazard ratio of .61 (95% CI, .45-.83, P < .01).

tusludastawushechuhauajarosuframachuslejurodrowraboslusibabofratrathijubraniphudeswopohokasajestusathodruswiphisliprodorastimuspasoslislauostesholephububraswugikitrebriwrostodoclislefruspotrecraslucugerecredrafrulephates
Dr. Robert J. Wong


About 70% of patients were non-Hispanic Whites and more than 90% were male in all consumption categories. The no-alcohol group was older than the high-risk alcohol group: 64 years vs 59.9 years (P < .0001). Compared with the high-risk alcohol group, the no-alcohol group had a significantly greater proportion of comorbid diabetes (62.3% vs 42.5%), hypertension (77.9% vs 69.1%), or cardiovascular disease (40.2% vs 25.9%, P < .0001 for all comparisons).

In a significant study observation, fewer than 5% of patients with high-risk use received behavioral or pharmacologic therapy and of those who did, most were referred for or received treatment at or near the time of cirrhosis diagnosis. “This highlights a major gap in linking patients with high-risk alcohol use to appropriate behavioral or pharmacologic therapy in a timely manner and may reflect missed opportunities to prevent further alcohol-related morbidity and mortality,” Dr. Wong and colleagues wrote.

They called for studies of novel interventions for timely assessment of alcohol use with linkage to addiction services. They cited the need to understand the interaction between levels of alcohol use and underlying MASLD, adding, “More research is also needed to understand whether this interaction varies across different populations.”

This study received no specific funding. Dr. Wong reported funding through his institution from Gilead Sciences, Exact Sciences, and Thera Technologies.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167904</fileName> <TBEID>0C04FE2D.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FE2D</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Alcohol-cirrhosis in MASLD</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240510T132054</QCDate> <firstPublished>20240510T133724</firstPublished> <LastPublished>20240510T133724</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240510T133724</CMSDate> <articleSource>FROM GASTROENTEROLOGY </articleSource> <facebookInfo/> <meetingNumber/> <byline>Diana Swift dianaswift@rogers.com</byline> <bylineText>DIANA SWIFT</bylineText> <bylineFull>DIANA SWIFT</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>One in nine patients with steatotic liver disease reported concurrent alcohol use, and more than 11% reported high-risk consumption, a national study of more th</metaDescription> <articlePDF/> <teaserImage>301426</teaserImage> <teaser>In a significant study observation, fewer than 5% of US veterans with high-risk use received behavioral or pharmacologic therapy.</teaser> <title>High-Alcohol Intake in MASLD Increases Risk of Cirrhosis</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>39313</term> <term>27970</term> </sections> <topics> <term canonical="true">346</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012918.jpg</altRep> <description role="drol:caption">Dr. Robert J. Wong</description> <description role="drol:credit">Dr. Wong</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012919.jpg</altRep> <description role="drol:caption">Dr. Tiffany Wu</description> <description role="drol:credit">Mayo Clinic</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>High-Alcohol Intake in MASLD Increases Risk of Cirrhosis</title> <deck/> </itemMeta> <itemContent> <p> <span class="tag metaDescription">One in nine patients with steatotic liver disease reported concurrent alcohol use, and more than 11% reported high-risk consumption, a national study of more than a million US veterans found.</span> </p> <p>Moreover, the combination of steatotic liver disease and high-risk alcohol intake carried a more than 43% higher long-term risk of liver cirrhosis compared with no alcohol use, according to researchers led by Robert J. Wong, MD, MS, of the Division of Gastroenterology and Hepatology, Veterans Affairs Healthcare System Palo Alto, at Stanford University School of Medicine in Palo Alto, California.<br/><br/>However, <span class="Hyperlink"><a href="https://www.gastrojournal.org/article/S0016-5085(24)00238-5/abstract">the study</a></span> found that “reducing alcohol use lowers risk of cirrhosis, emphasizing the importance of timely alcohol use assessment and early interventions to address high-risk alcohol use in steatotic liver disease,” Dr. Wong and associates <span class="Hyperlink">wrote in </span><em>Gastroenterology</em>.<br/><br/>Although concurrent moderate to heavy alcohol intake would be expected to lead more rapidly to liver disease progression, the existing literature has been conflicting, the authors noted. Several studies have even found moderate alcohol associated with a lower risk of advanced liver disease among MASLD patients, including that <span class="Hyperlink"><a href="https://www.journal-of-hepatology.eu/article/S0168-8278(12)00279-6/abstract">by Dunn et al.</a></span> .<br/><br/></p> <h2>The Study</h2> <p>MASLD patients were identified through the US Veterans Affairs Corporate Data Warehouse from January 1, 2010, through December 31, 2017, with follow-up through December 31, 2022. </p> <p>Alcohol use was assessed by Alcohol Use Disorders Identification Test–Concise (AUDIT-C) scores and was categorized as follows: no alcohol (AUDIT-C = 0), low-risk alcohol use (AUDIT-C 1-2 for women and 1–3 for men), and high-risk alcohol (AUDIT-C ≥ 3 for women and ≥ 4 for men).<br/><br/>Among the 1,156,189 veterans with MASLD, 54.2% reported no alcohol, 34.6% low-risk alcohol, and 11.2% high-risk alcohol use. In median follow-up of nine to 10 years, incidence rates of cirrhosis were .53 per 100 person-years for no use, .42 for low-risk use, and .76 for high-risk use.<br/><br/>In contrast to patients with baseline high-risk alcohol intake who reported no change in use, those who decreased their alcohol intake during follow-up experienced a 39% reduction in the long-term risk of cirrhosis, for a hazard ratio of .61 (95% CI, .45-.83, <em>P</em> &lt; .01).[[{"fid":"301426","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Robert J. Wong, Division of Gastroenterology and Hepatology, Veterans Affairs Healthcare System Palo Alto, at Stanford University School of Medicine in Palo Alto, California","field_file_image_credit[und][0][value]":"Dr. Wong","field_file_image_caption[und][0][value]":"Dr. Robert J. Wong"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]<br/><br/>About 70% of patients were non-Hispanic Whites and more than 90% were male in all consumption categories. The no-alcohol group was older than the high-risk alcohol group: 64 years vs 59.9 years (<em>P</em> &lt; .0001). Compared with the high-risk alcohol group, the no-alcohol group had a significantly greater proportion of comorbid diabetes (62.3% vs 42.5%), hypertension (77.9% vs 69.1%), or cardiovascular disease (40.2% vs 25.9%, <em>P</em> &lt; .0001 for all comparisons).<br/><br/>In a significant study observation, fewer than 5% of patients with high-risk use received behavioral or pharmacologic therapy and of those who did, most were referred for or received treatment at or near the time of cirrhosis diagnosis. “This highlights a major gap in linking patients with high-risk alcohol use to appropriate behavioral or pharmacologic therapy in a timely manner and may reflect missed opportunities to prevent further alcohol-related morbidity and mortality,” Dr. Wong and colleagues wrote.<br/><br/>They called for studies of novel interventions for timely assessment of alcohol use with linkage to addiction services. They cited the need to understand the interaction between levels of alcohol use and underlying MASLD, adding, “More research is also needed to understand whether this interaction varies across different populations.”<br/><br/>This study received no specific funding. Dr. Wong reported funding through his institution from Gilead Sciences, Exact Sciences, and Thera Technologies.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>views</itemRole> <itemClass>text</itemClass> <title>Critical Need for Early Tx of High-Risk Alcohol Use</title> <deck/> </itemMeta> <itemContent> <p>Recent consensus in defining metabolic dysfunction-associated steatotic liver disease (MASLD) has raised awareness for the combined impact of cardiometabolic risk factors and alcohol consumption on liver disease progression. This study by Wong et al. highlights the undeniable influence of high-risk alcohol use on the development of advanced liver disease. </p> <p>In a national cohort of over 1 million US veterans with steatotic liver disease (SLD), patients with high-risk alcohol use based on AUDIT-C assessment exhibited &gt; 43% greater risk of cirrhosis compared to those with no alcohol use. The relationship between alcohol and liver severity in SLD was observed even after excluding patients meeting classification for predominant alcohol-associated liver disease. While increased alcohol use was associated with increased incidence of cirrhosis, decreased alcohol use led to a notable 39% reduction in cirrhosis risk over time.<br/><br/>Reducing alcohol consumption remains best practice guidelines for mitigating risk of progression in steatotic liver disease. However, results of this study emphasize the critical need for early identification and treatment of high-risk alcohol use in all patients with SLD. While universal recommendations for alcohol abstinence provides pragmatic implementation, there is a significant need to better understand the interaction of specific metabolic risk factors and patterns of alcohol use across the spectrum of MetALD to guide personalized recommendations for patient education and management.[[{"fid":"301427","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Tiffany Wu, Transplant Hepatology at Mayo Clinic in Rochester, Minnesota","field_file_image_credit[und][0][value]":"Mayo Clinic","field_file_image_caption[und][0][value]":"Dr. Tiffany Wu"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]<br/><br/>Further research using prospective clinical trial design is needed to evaluate the interplay of alcohol consumption and metabolic risk factors across variable age, sex, genetics, and environmental exposures that are increasingly being recognized as vital drivers of health and disease.</p> <p><em> <em>Tiffany Wu, MD, MS, is a fellow in Transplant Hepatology at Mayo Clinic in Rochester, Minnesota. She has no conflicts.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Genes May Govern Intestinal Sites of Pediatric Crohn’s

Article Type
Changed
Thu, 05/09/2024 - 16:43

Genetic predisposition likely directs intestinal disease location in pediatric Crohn’s disease (CD) — whether colon-predominant (C-CD) or small-bowel-predominant (SB-CD), a small analysis in Cellular and Molecular Gastroenterology and Hepatology suggests.

Richard Kellermayer, MD, PhD, professor of pediatrics in the Section of Pediatric Gastroenterology, Hepatology and Nutrition at Baylor College of Medicine in Houston, Texas, and colleagues compared the genetic makeup of patients based on their Crohn’s disease location — predominantly in the small bowel (L4) or predominantly in the colon (L2 and/or L3). They then generated bipartite networks of susceptibility genes to study the polygenic background of the disease subtypes. They hypothesize that such networks may govern where a patient develops Crohn’s disease.

gomugetrisadaclavuphuprospikehivotibothubauo
Dr. Richard Kellermayer

According to current understanding, as Dr. Kellermayer told GI & Hepatology News, most autoimmune disorders, CD included, develop in people with a genetic predisposition after serial environmental insults between conception and young adulthood. “As opposed to single-gene-associated genetic disorders, autoimmune diseases are linked to several hundred genes in which subtle anomalies can work in concert to predispose someone to a certain disorder,” he said. “We hope our findings will guide the development of personalized treatments based on the disease location at diagnosis to advance precision medicine.”
 

CD cases

Eight cases of SB-CD and 11 of C-CD met the inclusion criteria. Mean age at CD diagnosis was about 11 years for both subtypes, while 36.3% of patients with C-CD were female vs 25% of those with SB-CD. Ethnicity was 72.2% White in the C-CD group and 87.5% in the SB-CD group.

As to the main ileocolonic locations according to the Paris Classification of pediatric inflammatory bowel disease, 54.5% in the C-CD group had involvement at L2 and 45.5% at L3. In SB-CD cases, 100% had disease at L4b, 37.5% at L4, and 50% at L1.

The researchers identified 115 single-nucleotide polymorphisms (SNPs) with a combined annotation-dependent depletion (CADD) score on Phil’s Read Editor (PHRED) of >10 that was associated with 97 genes. PHRED is a computer program measuring the quality of the identification of nucleobases generated by automated DNA sequencing and scores the deleteriousness of single-nucleotide variants. The identified genes in this study had a significantly (P < .01) different allele variation between C-CD and SB-CD.

Among the top 28 candidates was an SNP in the EFNA3 gene with a CADD score > 20 for differentiating between the two phenotypically distinct CD groups. Furthermore, the EFNA3 rs17723260 (predicted to be deleterious) was found to have a significantly lower allele frequency (4.5%) in C-CD compared with its allele frequency of 37.5% in SB-CD (chi square P = .0097).

“This finding indicates that EFNA3 might play a role in modulating colonic inflammation, in which a deleterious genetic defect might provide protection against colitis (and direct autoimmunity against the proximal small bowel) in the polygenic background of CD,” the investigators wrote.

EFNA3 has been linked to both CD and ulcerative colitis. Another four genes associated with the top five SNP candidates had already been connected with IBD or mammalian intestinal inflammation. 

According to the authors, the biomedical literature and mouse model findings “implicate the translational relevance of our candidate gene compendium for directing colon- vs small-bowel–predominant CD development.” They hope the findings will be replicated in larger CD cohorts differentiated by disease location. “Our work may set the nidus for CD subtype–based precision medicine by guiding individualized treatment strategies,” they wrote.

This study was supported by the ProKIIDS Network of the Crohn’s and Colitis Foundation and the Public Health Service. It was also supported by the Wagner, Frugoni, and Klaasmeyer families’ Gutsy Kids Fund and by the DR and GL Laws Fund. The authors disclosed no conflicts of interest.

Publications
Topics
Sections

Genetic predisposition likely directs intestinal disease location in pediatric Crohn’s disease (CD) — whether colon-predominant (C-CD) or small-bowel-predominant (SB-CD), a small analysis in Cellular and Molecular Gastroenterology and Hepatology suggests.

Richard Kellermayer, MD, PhD, professor of pediatrics in the Section of Pediatric Gastroenterology, Hepatology and Nutrition at Baylor College of Medicine in Houston, Texas, and colleagues compared the genetic makeup of patients based on their Crohn’s disease location — predominantly in the small bowel (L4) or predominantly in the colon (L2 and/or L3). They then generated bipartite networks of susceptibility genes to study the polygenic background of the disease subtypes. They hypothesize that such networks may govern where a patient develops Crohn’s disease.

gomugetrisadaclavuphuprospikehivotibothubauo
Dr. Richard Kellermayer

According to current understanding, as Dr. Kellermayer told GI & Hepatology News, most autoimmune disorders, CD included, develop in people with a genetic predisposition after serial environmental insults between conception and young adulthood. “As opposed to single-gene-associated genetic disorders, autoimmune diseases are linked to several hundred genes in which subtle anomalies can work in concert to predispose someone to a certain disorder,” he said. “We hope our findings will guide the development of personalized treatments based on the disease location at diagnosis to advance precision medicine.”
 

CD cases

Eight cases of SB-CD and 11 of C-CD met the inclusion criteria. Mean age at CD diagnosis was about 11 years for both subtypes, while 36.3% of patients with C-CD were female vs 25% of those with SB-CD. Ethnicity was 72.2% White in the C-CD group and 87.5% in the SB-CD group.

As to the main ileocolonic locations according to the Paris Classification of pediatric inflammatory bowel disease, 54.5% in the C-CD group had involvement at L2 and 45.5% at L3. In SB-CD cases, 100% had disease at L4b, 37.5% at L4, and 50% at L1.

The researchers identified 115 single-nucleotide polymorphisms (SNPs) with a combined annotation-dependent depletion (CADD) score on Phil’s Read Editor (PHRED) of >10 that was associated with 97 genes. PHRED is a computer program measuring the quality of the identification of nucleobases generated by automated DNA sequencing and scores the deleteriousness of single-nucleotide variants. The identified genes in this study had a significantly (P < .01) different allele variation between C-CD and SB-CD.

Among the top 28 candidates was an SNP in the EFNA3 gene with a CADD score > 20 for differentiating between the two phenotypically distinct CD groups. Furthermore, the EFNA3 rs17723260 (predicted to be deleterious) was found to have a significantly lower allele frequency (4.5%) in C-CD compared with its allele frequency of 37.5% in SB-CD (chi square P = .0097).

“This finding indicates that EFNA3 might play a role in modulating colonic inflammation, in which a deleterious genetic defect might provide protection against colitis (and direct autoimmunity against the proximal small bowel) in the polygenic background of CD,” the investigators wrote.

EFNA3 has been linked to both CD and ulcerative colitis. Another four genes associated with the top five SNP candidates had already been connected with IBD or mammalian intestinal inflammation. 

According to the authors, the biomedical literature and mouse model findings “implicate the translational relevance of our candidate gene compendium for directing colon- vs small-bowel–predominant CD development.” They hope the findings will be replicated in larger CD cohorts differentiated by disease location. “Our work may set the nidus for CD subtype–based precision medicine by guiding individualized treatment strategies,” they wrote.

This study was supported by the ProKIIDS Network of the Crohn’s and Colitis Foundation and the Public Health Service. It was also supported by the Wagner, Frugoni, and Klaasmeyer families’ Gutsy Kids Fund and by the DR and GL Laws Fund. The authors disclosed no conflicts of interest.

Genetic predisposition likely directs intestinal disease location in pediatric Crohn’s disease (CD) — whether colon-predominant (C-CD) or small-bowel-predominant (SB-CD), a small analysis in Cellular and Molecular Gastroenterology and Hepatology suggests.

Richard Kellermayer, MD, PhD, professor of pediatrics in the Section of Pediatric Gastroenterology, Hepatology and Nutrition at Baylor College of Medicine in Houston, Texas, and colleagues compared the genetic makeup of patients based on their Crohn’s disease location — predominantly in the small bowel (L4) or predominantly in the colon (L2 and/or L3). They then generated bipartite networks of susceptibility genes to study the polygenic background of the disease subtypes. They hypothesize that such networks may govern where a patient develops Crohn’s disease.

gomugetrisadaclavuphuprospikehivotibothubauo
Dr. Richard Kellermayer

According to current understanding, as Dr. Kellermayer told GI & Hepatology News, most autoimmune disorders, CD included, develop in people with a genetic predisposition after serial environmental insults between conception and young adulthood. “As opposed to single-gene-associated genetic disorders, autoimmune diseases are linked to several hundred genes in which subtle anomalies can work in concert to predispose someone to a certain disorder,” he said. “We hope our findings will guide the development of personalized treatments based on the disease location at diagnosis to advance precision medicine.”
 

CD cases

Eight cases of SB-CD and 11 of C-CD met the inclusion criteria. Mean age at CD diagnosis was about 11 years for both subtypes, while 36.3% of patients with C-CD were female vs 25% of those with SB-CD. Ethnicity was 72.2% White in the C-CD group and 87.5% in the SB-CD group.

As to the main ileocolonic locations according to the Paris Classification of pediatric inflammatory bowel disease, 54.5% in the C-CD group had involvement at L2 and 45.5% at L3. In SB-CD cases, 100% had disease at L4b, 37.5% at L4, and 50% at L1.

The researchers identified 115 single-nucleotide polymorphisms (SNPs) with a combined annotation-dependent depletion (CADD) score on Phil’s Read Editor (PHRED) of >10 that was associated with 97 genes. PHRED is a computer program measuring the quality of the identification of nucleobases generated by automated DNA sequencing and scores the deleteriousness of single-nucleotide variants. The identified genes in this study had a significantly (P < .01) different allele variation between C-CD and SB-CD.

Among the top 28 candidates was an SNP in the EFNA3 gene with a CADD score > 20 for differentiating between the two phenotypically distinct CD groups. Furthermore, the EFNA3 rs17723260 (predicted to be deleterious) was found to have a significantly lower allele frequency (4.5%) in C-CD compared with its allele frequency of 37.5% in SB-CD (chi square P = .0097).

“This finding indicates that EFNA3 might play a role in modulating colonic inflammation, in which a deleterious genetic defect might provide protection against colitis (and direct autoimmunity against the proximal small bowel) in the polygenic background of CD,” the investigators wrote.

EFNA3 has been linked to both CD and ulcerative colitis. Another four genes associated with the top five SNP candidates had already been connected with IBD or mammalian intestinal inflammation. 

According to the authors, the biomedical literature and mouse model findings “implicate the translational relevance of our candidate gene compendium for directing colon- vs small-bowel–predominant CD development.” They hope the findings will be replicated in larger CD cohorts differentiated by disease location. “Our work may set the nidus for CD subtype–based precision medicine by guiding individualized treatment strategies,” they wrote.

This study was supported by the ProKIIDS Network of the Crohn’s and Colitis Foundation and the Public Health Service. It was also supported by the Wagner, Frugoni, and Klaasmeyer families’ Gutsy Kids Fund and by the DR and GL Laws Fund. The authors disclosed no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167921</fileName> <TBEID>0C04FE47.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FE47</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Location genes in pediatric CD</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240509T153928</QCDate> <firstPublished>20240509T163909</firstPublished> <LastPublished>20240509T163909</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240509T163909</CMSDate> <articleSource>FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY </articleSource> <facebookInfo/> <meetingNumber/> <byline>Diana Swift dianaswift@rpgers.cm</byline> <bylineText>DIANA SWIFT</bylineText> <bylineFull>DIANA SWIFT</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Genetic predisposition likely directs intestinal disease location in pediatric Crohn’s disease (CD) — whether colon-predominant (C-CD) or small-bowel-predominan</metaDescription> <articlePDF/> <teaserImage>301415</teaserImage> <teaser>The researchers hope the findings will be replicated in larger CD cohorts differentiated by disease location.</teaser> <title>Genes May Govern Intestinal Sites of Pediatric Crohn’s</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">345</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401290c.jpg</altRep> <description role="drol:caption">Dr. Richard Kellermayer</description> <description role="drol:credit">Baylor College of Medicine</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Genes May Govern Intestinal Sites of Pediatric Crohn’s</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Genetic predisposition likely directs intestinal disease location in pediatric Crohn’s disease (CD) — whether colon-predominant (C-CD) or small-bowel-predominant (SB-CD)</span>, <span class="Hyperlink"><a href="https://www.cmghjournal.org/article/S2352-345X(24)00038-9/fulltext">a small analysis</a></span> in <em>Cellular and Molecular Gastroenterology and Hepatology</em> suggests.</p> <p>Richard Kellermayer, MD, PhD, professor of pediatrics in the Section of Pediatric Gastroenterology, Hepatology and Nutrition at Baylor College of Medicine in Houston, Texas, and colleagues compared the genetic makeup of patients based on their Crohn’s disease location — predominantly in the small bowel (L4) or predominantly in the colon (L2 and/or L3). They then generated bipartite networks of susceptibility genes to study the polygenic background of the disease subtypes. They hypothesize that such networks may govern where a patient develops Crohn’s disease.<br/><br/>[[{"fid":"301415","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Richard Kellermayer, Section of Pediatric Gastroenterology, Hepatology and Nutrition at Baylor College of Medicine in Houston, Texas","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Richard Kellermayer"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]According to current understanding, as Dr. Kellermayer told <em>GI &amp; Hepatology News</em>, most autoimmune disorders, CD included, develop in people with a genetic predisposition after serial environmental insults between conception and young adulthood. “As opposed to single-gene-associated genetic disorders, autoimmune diseases are linked to several hundred genes in which subtle anomalies can work in concert to predispose someone to a certain disorder,” he said. “We hope our findings will guide the development of personalized treatments based on the disease location at diagnosis to advance precision medicine.”<br/><br/></p> <h2>CD cases</h2> <p>Eight cases of SB-CD and 11 of C-CD met the inclusion criteria. Mean age at CD diagnosis was about 11 years for both subtypes, while 36.3% of patients with C-CD were female vs 25% of those with SB-CD. Ethnicity was 72.2% White in the C-CD group and 87.5% in the SB-CD group.</p> <p>As to the main ileocolonic locations according to the <span class="Hyperlink"><a href="https://academic.oup.com/ibdjournal/article/17/6/1314/4631074">Paris Classification</a></span> of pediatric inflammatory bowel disease, 54.5% in the C-CD group had involvement at L2 and 45.5% at L3. In SB-CD cases, 100% had disease at L4b, 37.5% at L4, and 50% at L1.<br/><br/>The researchers identified 115 single-nucleotide polymorphisms (SNPs) with a combined annotation-dependent depletion (CADD) score on Phil’s Read Editor (<span class="Hyperlink"><a href="https://en.wikipedia.org/wiki/Phred_quality_score">PHRED</a></span>) of &gt;10 that was associated with 97 genes. PHRED is a computer program measuring the quality of the identification of nucleobases generated by automated DNA sequencing and scores the deleteriousness of single-nucleotide variants. The identified genes in this study had a significantly (<em>P</em> &lt; .01) different allele variation between C-CD and SB-CD. <br/><br/>Among the top 28 candidates was an SNP in the <em>EFNA3</em> gene with a CADD score &gt; 20 for differentiating between the two phenotypically distinct CD groups. Furthermore, the <em>EFNA3</em> rs17723260 (predicted to be deleterious) was found to have a significantly lower allele frequency (4.5%) in C-CD compared with its allele frequency of 37.5% in SB-CD (chi square <em>P</em> = .0097). <br/><br/>“This finding indicates that <em>EFNA3</em> might play a role in modulating colonic inflammation, in which a deleterious genetic defect might provide protection against colitis (and direct autoimmunity against the proximal small bowel) in the polygenic background of CD,” the investigators wrote.<br/><br/><em>EFNA3</em> has been linked to both CD and ulcerative colitis. Another four genes associated with the top five SNP candidates had already been connected with IBD or mammalian intestinal inflammation. <br/><br/>According to the authors, the biomedical literature and mouse model findings “implicate the translational relevance of our candidate gene compendium for directing colon- vs small-bowel–predominant CD development.” They hope the findings will be replicated in larger CD cohorts differentiated by disease location. “Our work may set the nidus for CD subtype–based precision medicine by guiding individualized treatment strategies,” they wrote.<br/><br/>This study was supported by the ProKIIDS Network of the Crohn’s and Colitis Foundation and the Public Health Service. It was also supported by the Wagner, Frugoni, and Klaasmeyer families’ Gutsy Kids Fund and by the DR and GL Laws Fund. The authors disclosed no conflicts of interest.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Composite Scale Better Gauges Mucosal Injury in Celiac Disease

Article Type
Changed
Thu, 05/09/2024 - 15:08

A new two-measure metric seems to improve accuracy and statistical precision in assessing celiac disease (CeD) histology compared with either of two components alone, according to a study in Clinical Gastroenterology and Hepatology.

The new morphometric duodenal biopsy mucosal scale joins together villous height-to-crypt depth ratio (Vh:Cd) and intraepithelial lymphocytes (IEL) — each key CeD histological measures of the small intestine — in a scale called VCIEL.

The authors believe the VCIEL will enable a broader and more accurate measurement of mucosal health in CeD. It will be particularly useful for population analysis in clinical trials and could improve the powering of trial design. “Use of VCIEL may lead to better outcome measures for potential new therapeutic treatments benefiting patients,” wrote Jocelyn A. Silvester, MD, PhD, a pediatrician at Boston Children’s Hospital and an assistant professor at Harvard Medical School, and colleagues.

chushosushachokinunudi
Dr. Jocelyn A. Silvester


This chronic enteropathy affects about 1% of the world’s population and requires a lifelong adherence to a gluten-free diet, the authors noted.

The authors pointed to weaknesses in the current quantitative and qualitative ways of measuring gluten-induced mucosal injury on biopsy for CeD. “Morphometry measures the injury continuum for architecture and inflammation, but these are used as separate outcomes,” they wrote. “The original Marsh-Oberhuber [M-O] classifications are rather contrived approaches to assess a biologic continuum, forcing the injury in categorical groups of unclear clinical relevance and where clinically significant changes may occur within one single category.”

Moreover, the quantitation of inflammation relies on binary assessment as normal or increased, which results in histology that is unscorable by M-O if villous atrophy persists without increased IELs, they added.
 

The Study

In the absence of a broadly accepted single measure of mucosal injury in CeD, the group assessed whether the composite metric could improve statistical precision for assessing histology.

Enter VCIEL, which combines the Vh:Cd and IEL for individual patients with equal weighting by converting each scale to a fraction of their standard deviation and summing the results.

The researchers applied the VCIEL formula in a reanalysis of four clinical gluten-challenge trials and compared the results for Vh:Cd and IEL separately with those for VCIEL for clinical significance (effect size) and statistical significance.

In reanalysis of the ALV003-1021 trial, for example, the researchers observed an effect size and P value (analysis of covariance) of 1.37 and .038 for a delta (difference) value of Vh:Cd 1.17 and .005 for IEL and 1.86 and .004 for VCIEL.

For the similar gluten-challenge IMGX003-NCCIH-1721 trial, the corresponding delta results were .76 and .057 for Vh:Cd, .98 and .018 for IEL, and 1.14 and .007 for VCIEL. Comparable improvements with VCIEL over individual Vh:Cd and IEL were observed for other studies, including a nontherapeutic gluten challenge study.

In NCT03409796 trial data, the computation of VCIEL values showed an improved statistical significance relative to the component values of Vh:Cd and IEL by the within-group paired 2-tailed t test P values from baseline to day 15, particularly at a 10-g gluten challenge dose: Vh:Cd, IEL, VCIEL = .0050, .0031, and .0014, respectively.

Little correlation emerged between baseline values and changes with intervention for Vh:Cd and IEL on an individual patient basis.

The greater accuracy and statistical precision of the VCIEL scale are presumably due to averaging over some of the measurement uncertainty in individual patient and timepoint Vh:Cd and IEL values and creating a composite of different histologic properties, the authors noted.

This study was funded by ImmunogenX, Inc. First author Jack A. Syage is a cofounder and shareholder in ImmunogenX Inc. Dr. Silvester has served on an advisory board for Takeda Pharmaceuticals and has received research funding from Biomedal S.L., Cour Pharmaceuticals, and Glutenostics LLC. Several coauthors disclosed various financial ties to multiple private-sector pharmaceutical and biomedical companies, including ImmunogenX.

Publications
Topics
Sections

A new two-measure metric seems to improve accuracy and statistical precision in assessing celiac disease (CeD) histology compared with either of two components alone, according to a study in Clinical Gastroenterology and Hepatology.

The new morphometric duodenal biopsy mucosal scale joins together villous height-to-crypt depth ratio (Vh:Cd) and intraepithelial lymphocytes (IEL) — each key CeD histological measures of the small intestine — in a scale called VCIEL.

The authors believe the VCIEL will enable a broader and more accurate measurement of mucosal health in CeD. It will be particularly useful for population analysis in clinical trials and could improve the powering of trial design. “Use of VCIEL may lead to better outcome measures for potential new therapeutic treatments benefiting patients,” wrote Jocelyn A. Silvester, MD, PhD, a pediatrician at Boston Children’s Hospital and an assistant professor at Harvard Medical School, and colleagues.

chushosushachokinunudi
Dr. Jocelyn A. Silvester


This chronic enteropathy affects about 1% of the world’s population and requires a lifelong adherence to a gluten-free diet, the authors noted.

The authors pointed to weaknesses in the current quantitative and qualitative ways of measuring gluten-induced mucosal injury on biopsy for CeD. “Morphometry measures the injury continuum for architecture and inflammation, but these are used as separate outcomes,” they wrote. “The original Marsh-Oberhuber [M-O] classifications are rather contrived approaches to assess a biologic continuum, forcing the injury in categorical groups of unclear clinical relevance and where clinically significant changes may occur within one single category.”

Moreover, the quantitation of inflammation relies on binary assessment as normal or increased, which results in histology that is unscorable by M-O if villous atrophy persists without increased IELs, they added.
 

The Study

In the absence of a broadly accepted single measure of mucosal injury in CeD, the group assessed whether the composite metric could improve statistical precision for assessing histology.

Enter VCIEL, which combines the Vh:Cd and IEL for individual patients with equal weighting by converting each scale to a fraction of their standard deviation and summing the results.

The researchers applied the VCIEL formula in a reanalysis of four clinical gluten-challenge trials and compared the results for Vh:Cd and IEL separately with those for VCIEL for clinical significance (effect size) and statistical significance.

In reanalysis of the ALV003-1021 trial, for example, the researchers observed an effect size and P value (analysis of covariance) of 1.37 and .038 for a delta (difference) value of Vh:Cd 1.17 and .005 for IEL and 1.86 and .004 for VCIEL.

For the similar gluten-challenge IMGX003-NCCIH-1721 trial, the corresponding delta results were .76 and .057 for Vh:Cd, .98 and .018 for IEL, and 1.14 and .007 for VCIEL. Comparable improvements with VCIEL over individual Vh:Cd and IEL were observed for other studies, including a nontherapeutic gluten challenge study.

In NCT03409796 trial data, the computation of VCIEL values showed an improved statistical significance relative to the component values of Vh:Cd and IEL by the within-group paired 2-tailed t test P values from baseline to day 15, particularly at a 10-g gluten challenge dose: Vh:Cd, IEL, VCIEL = .0050, .0031, and .0014, respectively.

Little correlation emerged between baseline values and changes with intervention for Vh:Cd and IEL on an individual patient basis.

The greater accuracy and statistical precision of the VCIEL scale are presumably due to averaging over some of the measurement uncertainty in individual patient and timepoint Vh:Cd and IEL values and creating a composite of different histologic properties, the authors noted.

This study was funded by ImmunogenX, Inc. First author Jack A. Syage is a cofounder and shareholder in ImmunogenX Inc. Dr. Silvester has served on an advisory board for Takeda Pharmaceuticals and has received research funding from Biomedal S.L., Cour Pharmaceuticals, and Glutenostics LLC. Several coauthors disclosed various financial ties to multiple private-sector pharmaceutical and biomedical companies, including ImmunogenX.

A new two-measure metric seems to improve accuracy and statistical precision in assessing celiac disease (CeD) histology compared with either of two components alone, according to a study in Clinical Gastroenterology and Hepatology.

The new morphometric duodenal biopsy mucosal scale joins together villous height-to-crypt depth ratio (Vh:Cd) and intraepithelial lymphocytes (IEL) — each key CeD histological measures of the small intestine — in a scale called VCIEL.

The authors believe the VCIEL will enable a broader and more accurate measurement of mucosal health in CeD. It will be particularly useful for population analysis in clinical trials and could improve the powering of trial design. “Use of VCIEL may lead to better outcome measures for potential new therapeutic treatments benefiting patients,” wrote Jocelyn A. Silvester, MD, PhD, a pediatrician at Boston Children’s Hospital and an assistant professor at Harvard Medical School, and colleagues.

chushosushachokinunudi
Dr. Jocelyn A. Silvester


This chronic enteropathy affects about 1% of the world’s population and requires a lifelong adherence to a gluten-free diet, the authors noted.

The authors pointed to weaknesses in the current quantitative and qualitative ways of measuring gluten-induced mucosal injury on biopsy for CeD. “Morphometry measures the injury continuum for architecture and inflammation, but these are used as separate outcomes,” they wrote. “The original Marsh-Oberhuber [M-O] classifications are rather contrived approaches to assess a biologic continuum, forcing the injury in categorical groups of unclear clinical relevance and where clinically significant changes may occur within one single category.”

Moreover, the quantitation of inflammation relies on binary assessment as normal or increased, which results in histology that is unscorable by M-O if villous atrophy persists without increased IELs, they added.
 

The Study

In the absence of a broadly accepted single measure of mucosal injury in CeD, the group assessed whether the composite metric could improve statistical precision for assessing histology.

Enter VCIEL, which combines the Vh:Cd and IEL for individual patients with equal weighting by converting each scale to a fraction of their standard deviation and summing the results.

The researchers applied the VCIEL formula in a reanalysis of four clinical gluten-challenge trials and compared the results for Vh:Cd and IEL separately with those for VCIEL for clinical significance (effect size) and statistical significance.

In reanalysis of the ALV003-1021 trial, for example, the researchers observed an effect size and P value (analysis of covariance) of 1.37 and .038 for a delta (difference) value of Vh:Cd 1.17 and .005 for IEL and 1.86 and .004 for VCIEL.

For the similar gluten-challenge IMGX003-NCCIH-1721 trial, the corresponding delta results were .76 and .057 for Vh:Cd, .98 and .018 for IEL, and 1.14 and .007 for VCIEL. Comparable improvements with VCIEL over individual Vh:Cd and IEL were observed for other studies, including a nontherapeutic gluten challenge study.

In NCT03409796 trial data, the computation of VCIEL values showed an improved statistical significance relative to the component values of Vh:Cd and IEL by the within-group paired 2-tailed t test P values from baseline to day 15, particularly at a 10-g gluten challenge dose: Vh:Cd, IEL, VCIEL = .0050, .0031, and .0014, respectively.

Little correlation emerged between baseline values and changes with intervention for Vh:Cd and IEL on an individual patient basis.

The greater accuracy and statistical precision of the VCIEL scale are presumably due to averaging over some of the measurement uncertainty in individual patient and timepoint Vh:Cd and IEL values and creating a composite of different histologic properties, the authors noted.

This study was funded by ImmunogenX, Inc. First author Jack A. Syage is a cofounder and shareholder in ImmunogenX Inc. Dr. Silvester has served on an advisory board for Takeda Pharmaceuticals and has received research funding from Biomedal S.L., Cour Pharmaceuticals, and Glutenostics LLC. Several coauthors disclosed various financial ties to multiple private-sector pharmaceutical and biomedical companies, including ImmunogenX.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167920</fileName> <TBEID>0C04FE6B.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FE6B</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Composite Celiac Scale</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240509T140639</QCDate> <firstPublished>20240509T150455</firstPublished> <LastPublished>20240509T150455</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240509T150455</CMSDate> <articleSource>FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY </articleSource> <facebookInfo/> <meetingNumber/> <byline>Diana Swift dianaswift@rogers.com</byline> <bylineText>DIANA SWIFT</bylineText> <bylineFull>DIANA SWIFT</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>A new two-measure metric seems to improve accuracy and statistical precision in assessing celiac disease (CeD) histology compared with either of two components </metaDescription> <articlePDF/> <teaserImage>301388</teaserImage> <teaser>The new scale should be particularly useful for population analysis in clinical trials and could improve the powering of trial design.</teaser> <title>Composite Scale Better Gauges Mucosal Injury in Celiac Disease</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>39313</term> <term>27970</term> </sections> <topics> <term canonical="true">345</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401290b.jpg</altRep> <description role="drol:caption">Dr. Jocelyn A. Silvester</description> <description role="drol:credit">Boston Children’s Hospital</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Composite Scale Better Gauges Mucosal Injury in Celiac Disease</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">A new two-measure metric seems to improve accuracy and statistical precision in assessing celiac disease (CeD) histology compared with either of two components alone</span>, according <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.10.031">to a study</a></span> in <em>Clinical Gastroenterology and Hepatology</em>.</p> <p>The new morphometric duodenal biopsy mucosal scale joins together villous height-to-crypt depth ratio (Vh:Cd) and intraepithelial lymphocytes (IEL) — each key CeD histological measures of the small intestine — in a scale called VCIEL. <br/><br/>The authors believe the VCIEL will enable a broader and more accurate measurement of mucosal health in CeD. It will be particularly useful for population analysis in clinical trials and could improve the powering of trial design. “Use of VCIEL may lead to better outcome measures for potential new therapeutic treatments benefiting patients,” wrote Jocelyn A. Silvester, MD, PhD, a pediatrician at Boston Children’s Hospital and an assistant professor at Harvard Medical School, and colleagues.[[{"fid":"301388","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Jocelyn A. Silvester, Boston Children’s Hospital and Harvard Medical School","field_file_image_credit[und][0][value]":"Boston Children’s Hospital","field_file_image_caption[und][0][value]":"Dr. Jocelyn A. Silvester"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]<br/><br/>This chronic enteropathy affects about 1% of <span class="Hyperlink"><a href="https://journals.lww.com/ajg/abstract/2015/03000/trends_and_racial_ethnic_disparities_in.19.aspx">the world’s population</a></span> and requires a lifelong adherence to a gluten-free diet, the authors noted.<br/><br/>The authors pointed to weaknesses in the current quantitative and qualitative ways of measuring gluten-induced mucosal injury on biopsy for CeD. “Morphometry measures the injury continuum for architecture and inflammation, but these are used as separate outcomes,” they wrote. “The original <span class="Hyperlink"><a href="https://diagnosticpathology.biomedcentral.com/articles/10.1186/s13000-023-01412-x">Marsh-Oberhuber [M-O] classifications</a></span> are rather contrived approaches to assess a biologic continuum, forcing the injury in categorical groups of unclear clinical relevance and where clinically significant changes may occur within one single category.” <br/><br/>Moreover, the quantitation of inflammation relies on binary assessment as normal or increased, which results in histology that is unscorable by M-O if villous atrophy persists without increased IELs, they added.<br/><br/></p> <h2>The Study</h2> <p>In the absence of a broadly accepted single measure of mucosal injury in CeD, the group assessed whether the composite metric could improve statistical precision for assessing histology. </p> <p>Enter VCIEL, which combines the Vh:Cd and IEL for individual patients with equal weighting by converting each scale to a fraction of their standard deviation and summing the results. <br/><br/>The researchers applied the VCIEL formula in a reanalysis of four clinical gluten-challenge trials and compared the results for Vh:Cd and IEL separately with those for VCIEL for clinical significance (effect size) and statistical significance.<br/><br/>In reanalysis of the ALV003-1021 trial, for example, the researchers observed an effect size and <em>P</em> value (analysis of covariance) of 1.37 and .038 for a delta (difference) value of Vh:Cd 1.17 and .005 for IEL and 1.86 and .004 for VCIEL.<br/><br/>For the similar gluten-challenge IMGX003-NCCIH-1721 trial, the corresponding delta results were .76 and .057 for Vh:Cd, .98 and .018 for IEL, and 1.14 and .007 for VCIEL. Comparable improvements with VCIEL over individual Vh:Cd and IEL were observed for other studies, including a nontherapeutic gluten challenge study.<br/><br/>In NCT03409796 trial data, the computation of VCIEL values showed an improved statistical significance relative to the component values of Vh:Cd and IEL by the within-group paired 2-tailed t test <em>P</em> values from baseline to day 15, particularly at a 10-g gluten challenge dose: Vh:Cd, IEL, VCIEL = .0050, .0031, and .0014, respectively.<br/><br/>Little correlation emerged between baseline values and changes with intervention for Vh:Cd and IEL on an individual patient basis.<br/><br/>The greater accuracy and statistical precision of the VCIEL scale are presumably due to averaging over some of the measurement uncertainty in individual patient and timepoint Vh:Cd and IEL values and creating a composite of different histologic properties, the authors noted.<br/><br/>This study was funded by ImmunogenX, Inc. First author Jack A. Syage is a cofounder and shareholder in ImmunogenX Inc. Dr. Silvester has served on an advisory board for Takeda Pharmaceuticals and has received research funding from Biomedal S.L., Cour Pharmaceuticals, and Glutenostics LLC. Several coauthors disclosed various financial ties to multiple private-sector pharmaceutical and biomedical companies, including ImmunogenX.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

GLP-1s May Increase Post-Endoscopy Aspiration Pneumonia Risk

Article Type
Changed
Thu, 04/25/2024 - 13:21

 

The use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may lead to an increased risk for aspiration pneumonia after endoscopic procedures, according to a new large population-based study.

In June 2023, the American Society of Anesthesiologists (ASA) recommended holding GLP-1 RAs before an endoscopic or surgical procedure to reduce the risk for complications associated with anesthesia and delayed stomach emptying.

In response, the American Gastroenterological Association (AGA) published a rapid clinical practice update in November 2023 that found insufficient evidence to support patients stopping the medications before endoscopic procedures.

“It is known that GLP-1 RAs significantly reduce the motility of the stomach and small bowel. As more and more patients are being started on GLP-1 RAs at higher doses and longer half-life, the question became whether the current recommended fasting durations are enough to reasonably assume the stomach is empty prior to procedures that require sedation,” said senior author Ali Rezaie, MD, medical director of the GI Motility Program at Cedars-Sinai Medical Center in Los Angeles.

“We wanted to see if these medications in fact increased the chance of aspiration before the ASA suggestion went into effect,” he said. “However, this is not an easy task, as aspiration is a rare event and a large sample size is needed to confidently answer that question. That is why we evaluated nearly 1 million cases.”

The study was published online in Gastroenterology.
 

Analyzing GLP-1 RA Use

Dr. Rezaie and colleagues conducted a population-based, retrospective cohort study of the TriNetX dataset, which includes 114 million deidentified individual health records from 80 healthcare organizations. The research team analyzed nearly 1 million records for adult patients between ages 21 and 70 who underwent upper and lower endoscopies between January 2018 and December 2020.

Rezaie_Ali_CA_web.jpg
Dr. Ali Rezaie

The researchers defined GLP-1 RA users as those who had the medication for more than 6 months and two or more refills within 6 months before the procedure. They adjusted for 59 factors that could affect gut motility or aspiration risks, such as obesity, numerous chronic diseases, and dozens of medications. The primary outcome was aspiration pneumonia within a month after the procedure.

Among 963,184 patients who underwent endoscopy, 46,935 (4.9%) were considered GLP-1 RA users. Among those, 20,099 GLP-1 RA users met the inclusion criteria and had their results compared with non-GLP-1 RA users.

After propensity score matching for the 59 potential confounders, GLP-1 RA use had a higher incidence rate of aspiration pneumonia (0.83% vs 0.63%) and was associated with a significantly higher risk for aspiration pneumonia, with a hazard ratio (HR) of 1.33.

An even higher risk was seen among patients with propofol-assisted endoscopies (HR, 1.49) but not among those without propofol (HR, 1.31).

In a subgroup analysis based on endoscopy type, an elevated risk was observed among patients who underwent upper endoscopy (HR, 1.82) and combined upper and lower endoscopy (HR, 2.26) but not lower endoscopy (HR, 0.56).

“The results were not necessarily surprising given the mechanism of action of GLP-1 RAs. However, for the first time, this was shown with a clinically relevant outcome, such as aspiration pneumonia,” Dr. Rezaie said. “Aspiration during sedation can have devastating consequences, and the 0.2% difference in risk of aspiration can have a significant effect on healthcare as well.”

More than 20 million endoscopies are performed across the United States annually. Based on the assumption that about 3% of those patients are taking GLP-1 RAs, about 1200 aspiration cases per year can be prevented by raising awareness, he said.
 

 

 

Considering Next Steps

The varying risk profiles observed with separate sedation and endoscopy types point to a need for more tailored guidance in managing GLP-1 RA use before a procedure, the study authors wrote.

Although holding the medications before endoscopy may disrupt diabetes management, the potential increased risk for aspiration could justify a change in practice, particularly for upper endoscopy and propofol-associated procedures, they added.

At the same time, additional studies are needed to understand the optimal drug withholding windows before endoscopies and other procedures, they concluded.

“We will need more data on what is the optimal duration of holding GLP-1 RAs,” Dr. Rezaie said. “But given our data and current ASA guidance, stopping these medications prior to elective procedures is the safe thing to do.”

For now, AGA guidance remains the same as offered in the November 2023 update, suggesting an individual approach for each patient on a GLP-1 RA rather than a “blanket statement” on how to manage all patients taking these medications.

“Overall, I believe that this study is important, but we require more high-level data to inform clinical decision-making regarding patients using GLP-1 receptor agonists prior to gastrointestinal endoscopy,” said Andrew Y. Wang, MD, AGAF, chief of gastroenterology and hepatology and director of interventional endoscopy at the University of Virginia in Charlottesville.

Dr. Wang, who wasn’t involved with this study, coauthored the AGA rapid clinical practice update. He and colleagues advised continuing with a procedure as planned for patients on GLP-1 RAs who followed standard preprocedure fasting instructions and didn’t have nausea, vomiting, dyspepsia, or abdominal distention.

Wang_Andrew_VA_web.jpg
Dr. Andrew Y. Wang


Among patients with symptoms that suggest retained gastric contents, rapid sequence intubation may be considered, though it may not be possible in ambulatory or office-based endoscopy settings, Dr. Wang and colleagues wrote. As another option in lieu of stopping GLP-1 RAs, patients can be placed on a liquid diet for 1 day before the procedure.

“While this study found a signal suggesting that patients using GLP-1 RAs had an increased risk of aspiration pneumonia within 1 month following upper endoscopy or combined upper and lower endoscopy, it does not inform us if having patients stop GLP-1 RAs before endoscopic procedures — especially for a single dose — will mitigate this potential risk,” Dr. Wang said.

“It was also interesting that these investigators found that patients taking GLP-1 RAs who underwent lower endoscopy alone were not at increased risk for aspiration pneumonia,” Dr. Wang noted.

The authors didn’t report a funding source and disclosed no potential conflicts. Dr. Wang reported no relevant disclosures.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

The use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may lead to an increased risk for aspiration pneumonia after endoscopic procedures, according to a new large population-based study.

In June 2023, the American Society of Anesthesiologists (ASA) recommended holding GLP-1 RAs before an endoscopic or surgical procedure to reduce the risk for complications associated with anesthesia and delayed stomach emptying.

In response, the American Gastroenterological Association (AGA) published a rapid clinical practice update in November 2023 that found insufficient evidence to support patients stopping the medications before endoscopic procedures.

“It is known that GLP-1 RAs significantly reduce the motility of the stomach and small bowel. As more and more patients are being started on GLP-1 RAs at higher doses and longer half-life, the question became whether the current recommended fasting durations are enough to reasonably assume the stomach is empty prior to procedures that require sedation,” said senior author Ali Rezaie, MD, medical director of the GI Motility Program at Cedars-Sinai Medical Center in Los Angeles.

“We wanted to see if these medications in fact increased the chance of aspiration before the ASA suggestion went into effect,” he said. “However, this is not an easy task, as aspiration is a rare event and a large sample size is needed to confidently answer that question. That is why we evaluated nearly 1 million cases.”

The study was published online in Gastroenterology.
 

Analyzing GLP-1 RA Use

Dr. Rezaie and colleagues conducted a population-based, retrospective cohort study of the TriNetX dataset, which includes 114 million deidentified individual health records from 80 healthcare organizations. The research team analyzed nearly 1 million records for adult patients between ages 21 and 70 who underwent upper and lower endoscopies between January 2018 and December 2020.

Rezaie_Ali_CA_web.jpg
Dr. Ali Rezaie

The researchers defined GLP-1 RA users as those who had the medication for more than 6 months and two or more refills within 6 months before the procedure. They adjusted for 59 factors that could affect gut motility or aspiration risks, such as obesity, numerous chronic diseases, and dozens of medications. The primary outcome was aspiration pneumonia within a month after the procedure.

Among 963,184 patients who underwent endoscopy, 46,935 (4.9%) were considered GLP-1 RA users. Among those, 20,099 GLP-1 RA users met the inclusion criteria and had their results compared with non-GLP-1 RA users.

After propensity score matching for the 59 potential confounders, GLP-1 RA use had a higher incidence rate of aspiration pneumonia (0.83% vs 0.63%) and was associated with a significantly higher risk for aspiration pneumonia, with a hazard ratio (HR) of 1.33.

An even higher risk was seen among patients with propofol-assisted endoscopies (HR, 1.49) but not among those without propofol (HR, 1.31).

In a subgroup analysis based on endoscopy type, an elevated risk was observed among patients who underwent upper endoscopy (HR, 1.82) and combined upper and lower endoscopy (HR, 2.26) but not lower endoscopy (HR, 0.56).

“The results were not necessarily surprising given the mechanism of action of GLP-1 RAs. However, for the first time, this was shown with a clinically relevant outcome, such as aspiration pneumonia,” Dr. Rezaie said. “Aspiration during sedation can have devastating consequences, and the 0.2% difference in risk of aspiration can have a significant effect on healthcare as well.”

More than 20 million endoscopies are performed across the United States annually. Based on the assumption that about 3% of those patients are taking GLP-1 RAs, about 1200 aspiration cases per year can be prevented by raising awareness, he said.
 

 

 

Considering Next Steps

The varying risk profiles observed with separate sedation and endoscopy types point to a need for more tailored guidance in managing GLP-1 RA use before a procedure, the study authors wrote.

Although holding the medications before endoscopy may disrupt diabetes management, the potential increased risk for aspiration could justify a change in practice, particularly for upper endoscopy and propofol-associated procedures, they added.

At the same time, additional studies are needed to understand the optimal drug withholding windows before endoscopies and other procedures, they concluded.

“We will need more data on what is the optimal duration of holding GLP-1 RAs,” Dr. Rezaie said. “But given our data and current ASA guidance, stopping these medications prior to elective procedures is the safe thing to do.”

For now, AGA guidance remains the same as offered in the November 2023 update, suggesting an individual approach for each patient on a GLP-1 RA rather than a “blanket statement” on how to manage all patients taking these medications.

“Overall, I believe that this study is important, but we require more high-level data to inform clinical decision-making regarding patients using GLP-1 receptor agonists prior to gastrointestinal endoscopy,” said Andrew Y. Wang, MD, AGAF, chief of gastroenterology and hepatology and director of interventional endoscopy at the University of Virginia in Charlottesville.

Dr. Wang, who wasn’t involved with this study, coauthored the AGA rapid clinical practice update. He and colleagues advised continuing with a procedure as planned for patients on GLP-1 RAs who followed standard preprocedure fasting instructions and didn’t have nausea, vomiting, dyspepsia, or abdominal distention.

Wang_Andrew_VA_web.jpg
Dr. Andrew Y. Wang


Among patients with symptoms that suggest retained gastric contents, rapid sequence intubation may be considered, though it may not be possible in ambulatory or office-based endoscopy settings, Dr. Wang and colleagues wrote. As another option in lieu of stopping GLP-1 RAs, patients can be placed on a liquid diet for 1 day before the procedure.

“While this study found a signal suggesting that patients using GLP-1 RAs had an increased risk of aspiration pneumonia within 1 month following upper endoscopy or combined upper and lower endoscopy, it does not inform us if having patients stop GLP-1 RAs before endoscopic procedures — especially for a single dose — will mitigate this potential risk,” Dr. Wang said.

“It was also interesting that these investigators found that patients taking GLP-1 RAs who underwent lower endoscopy alone were not at increased risk for aspiration pneumonia,” Dr. Wang noted.

The authors didn’t report a funding source and disclosed no potential conflicts. Dr. Wang reported no relevant disclosures.

A version of this article appeared on Medscape.com.

 

The use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may lead to an increased risk for aspiration pneumonia after endoscopic procedures, according to a new large population-based study.

In June 2023, the American Society of Anesthesiologists (ASA) recommended holding GLP-1 RAs before an endoscopic or surgical procedure to reduce the risk for complications associated with anesthesia and delayed stomach emptying.

In response, the American Gastroenterological Association (AGA) published a rapid clinical practice update in November 2023 that found insufficient evidence to support patients stopping the medications before endoscopic procedures.

“It is known that GLP-1 RAs significantly reduce the motility of the stomach and small bowel. As more and more patients are being started on GLP-1 RAs at higher doses and longer half-life, the question became whether the current recommended fasting durations are enough to reasonably assume the stomach is empty prior to procedures that require sedation,” said senior author Ali Rezaie, MD, medical director of the GI Motility Program at Cedars-Sinai Medical Center in Los Angeles.

“We wanted to see if these medications in fact increased the chance of aspiration before the ASA suggestion went into effect,” he said. “However, this is not an easy task, as aspiration is a rare event and a large sample size is needed to confidently answer that question. That is why we evaluated nearly 1 million cases.”

The study was published online in Gastroenterology.
 

Analyzing GLP-1 RA Use

Dr. Rezaie and colleagues conducted a population-based, retrospective cohort study of the TriNetX dataset, which includes 114 million deidentified individual health records from 80 healthcare organizations. The research team analyzed nearly 1 million records for adult patients between ages 21 and 70 who underwent upper and lower endoscopies between January 2018 and December 2020.

Rezaie_Ali_CA_web.jpg
Dr. Ali Rezaie

The researchers defined GLP-1 RA users as those who had the medication for more than 6 months and two or more refills within 6 months before the procedure. They adjusted for 59 factors that could affect gut motility or aspiration risks, such as obesity, numerous chronic diseases, and dozens of medications. The primary outcome was aspiration pneumonia within a month after the procedure.

Among 963,184 patients who underwent endoscopy, 46,935 (4.9%) were considered GLP-1 RA users. Among those, 20,099 GLP-1 RA users met the inclusion criteria and had their results compared with non-GLP-1 RA users.

After propensity score matching for the 59 potential confounders, GLP-1 RA use had a higher incidence rate of aspiration pneumonia (0.83% vs 0.63%) and was associated with a significantly higher risk for aspiration pneumonia, with a hazard ratio (HR) of 1.33.

An even higher risk was seen among patients with propofol-assisted endoscopies (HR, 1.49) but not among those without propofol (HR, 1.31).

In a subgroup analysis based on endoscopy type, an elevated risk was observed among patients who underwent upper endoscopy (HR, 1.82) and combined upper and lower endoscopy (HR, 2.26) but not lower endoscopy (HR, 0.56).

“The results were not necessarily surprising given the mechanism of action of GLP-1 RAs. However, for the first time, this was shown with a clinically relevant outcome, such as aspiration pneumonia,” Dr. Rezaie said. “Aspiration during sedation can have devastating consequences, and the 0.2% difference in risk of aspiration can have a significant effect on healthcare as well.”

More than 20 million endoscopies are performed across the United States annually. Based on the assumption that about 3% of those patients are taking GLP-1 RAs, about 1200 aspiration cases per year can be prevented by raising awareness, he said.
 

 

 

Considering Next Steps

The varying risk profiles observed with separate sedation and endoscopy types point to a need for more tailored guidance in managing GLP-1 RA use before a procedure, the study authors wrote.

Although holding the medications before endoscopy may disrupt diabetes management, the potential increased risk for aspiration could justify a change in practice, particularly for upper endoscopy and propofol-associated procedures, they added.

At the same time, additional studies are needed to understand the optimal drug withholding windows before endoscopies and other procedures, they concluded.

“We will need more data on what is the optimal duration of holding GLP-1 RAs,” Dr. Rezaie said. “But given our data and current ASA guidance, stopping these medications prior to elective procedures is the safe thing to do.”

For now, AGA guidance remains the same as offered in the November 2023 update, suggesting an individual approach for each patient on a GLP-1 RA rather than a “blanket statement” on how to manage all patients taking these medications.

“Overall, I believe that this study is important, but we require more high-level data to inform clinical decision-making regarding patients using GLP-1 receptor agonists prior to gastrointestinal endoscopy,” said Andrew Y. Wang, MD, AGAF, chief of gastroenterology and hepatology and director of interventional endoscopy at the University of Virginia in Charlottesville.

Dr. Wang, who wasn’t involved with this study, coauthored the AGA rapid clinical practice update. He and colleagues advised continuing with a procedure as planned for patients on GLP-1 RAs who followed standard preprocedure fasting instructions and didn’t have nausea, vomiting, dyspepsia, or abdominal distention.

Wang_Andrew_VA_web.jpg
Dr. Andrew Y. Wang


Among patients with symptoms that suggest retained gastric contents, rapid sequence intubation may be considered, though it may not be possible in ambulatory or office-based endoscopy settings, Dr. Wang and colleagues wrote. As another option in lieu of stopping GLP-1 RAs, patients can be placed on a liquid diet for 1 day before the procedure.

“While this study found a signal suggesting that patients using GLP-1 RAs had an increased risk of aspiration pneumonia within 1 month following upper endoscopy or combined upper and lower endoscopy, it does not inform us if having patients stop GLP-1 RAs before endoscopic procedures — especially for a single dose — will mitigate this potential risk,” Dr. Wang said.

“It was also interesting that these investigators found that patients taking GLP-1 RAs who underwent lower endoscopy alone were not at increased risk for aspiration pneumonia,” Dr. Wang noted.

The authors didn’t report a funding source and disclosed no potential conflicts. Dr. Wang reported no relevant disclosures.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167772</fileName> <TBEID>0C04FB01.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FB01</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240425T115854</QCDate> <firstPublished>20240425T131527</firstPublished> <LastPublished>20240425T131527</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240425T131527</CMSDate> <articleSource>FROM GASTROENTEROLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Carolyn Crist</byline> <bylineText>CAROLYN CRIST</bylineText> <bylineFull>CAROLYN CRIST</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) may lead to an increased risk for aspiration pneumonia after endoscopic procedures</metaDescription> <articlePDF/> <teaserImage>301173</teaserImage> <teaser>Additional studies are needed to understand the optimal drug withholding windows before endoscopies and other procedures.</teaser> <title>GLP-1s May Increase Post-Endoscopy Aspiration Pneumonia Risk</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>chph</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> <term>21</term> <term>15</term> <term>6</term> </publications> <sections> <term canonical="true">69</term> <term>39313</term> <term>27970</term> </sections> <topics> <term canonical="true">39702</term> <term>347</term> <term>213</term> <term>284</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012882.jpg</altRep> <description role="drol:caption">Dr. Ali Rezaie</description> <description role="drol:credit">Cedars-Sinai Medical Center</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24010452.jpg</altRep> <description role="drol:caption">Dr. Andrew Y. Wang</description> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>GLP-1s May Increase Post-Endoscopy Aspiration Pneumonia Risk</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">The use of <span class="Hyperlink">glucagon</span>-like peptide-1 receptor agonists (GLP-1 RAs) may lead to an increased risk for <span class="Hyperlink">aspiration pneumonia</span> after endoscopic procedures</span>, according to a new large population-based study.</p> <p>In June 2023, the American Society of Anesthesiologists (ASA) <span class="Hyperlink"><a href="https://www.asahq.org/about-asa/newsroom/news-releases/2023/06/american-society-of-anesthesiologists-consensus-based-guidance-on-preoperative">recommended</a></span> holding GLP-1 RAs before an endoscopic or surgical procedure to reduce the risk for complications associated with <span class="Hyperlink">anesthesia</span> and delayed stomach emptying.<br/><br/>In response, the American Gastroenterological Association (AGA) <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.11.002">published</a></span> a rapid clinical practice update in November 2023 that found insufficient evidence to support patients stopping the medications before endoscopic procedures.<br/><br/>“It is known that GLP-1 RAs significantly reduce the motility of the stomach and small bowel. As more and more patients are being started on GLP-1 RAs at higher doses and longer half-life, the question became whether the current recommended fasting durations are enough to reasonably assume the stomach is empty prior to procedures that require sedation,” said senior author Ali Rezaie, MD, medical director of the GI Motility Program at Cedars-Sinai Medical Center in Los Angeles.<br/><br/>“We wanted to see if these medications in fact increased the chance of aspiration before the ASA suggestion went into effect,” he said. “However, this is not an easy task, as aspiration is a rare event and a large sample size is needed to confidently answer that question. That is why we evaluated nearly 1 million cases.”<br/><br/>The study was <span class="Hyperlink"><a href="https://www.gastrojournal.org/article/S0016-5085(24)00298-1/abstract">published online</a></span> in <em>Gastroenterology</em>.<br/><br/></p> <h2>Analyzing GLP-1 RA Use</h2> <p>Dr. Rezaie and colleagues conducted a population-based, retrospective cohort study of the TriNetX dataset, which includes 114 million deidentified individual health records from 80 healthcare organizations. The research team analyzed nearly 1 million records for adult patients between ages 21 and 70 who underwent upper and lower endoscopies between January 2018 and December 2020.[[{"fid":"301173","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Ali Rezaie, medical director of the GI Motility Program at Cedars-Sinai Medical Center in Los Angeles","field_file_image_credit[und][0][value]":"Cedars-Sinai Medical Center","field_file_image_caption[und][0][value]":"Dr. Ali Rezaie"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]</p> <p>The researchers defined GLP-1 RA users as those who had the medication for more than 6 months and two or more refills within 6 months before the procedure. They adjusted for 59 factors that could affect gut motility or aspiration risks, such as <span class="Hyperlink">obesity</span>, numerous chronic diseases, and dozens of medications. The primary outcome was aspiration pneumonia within a month after the procedure.<br/><br/>Among 963,184 patients who underwent endoscopy, 46,935 (4.9%) were considered GLP-1 RA users. Among those, 20,099 GLP-1 RA users met the inclusion criteria and had their results compared with non-GLP-1 RA users.<br/><br/>After propensity score matching for the 59 potential confounders, GLP-1 RA use had a higher incidence rate of aspiration pneumonia (0.83% vs 0.63%) and was associated with a significantly higher risk for aspiration pneumonia, with a hazard ratio (HR) of 1.33.<br/><br/>An even higher risk was seen among patients with <span class="Hyperlink">propofol</span>-assisted endoscopies (HR, 1.49) but not among those without propofol (HR, 1.31).<br/><br/>In a subgroup analysis based on endoscopy type, an elevated risk was observed among patients who underwent upper endoscopy (HR, 1.82) and combined upper and lower endoscopy (HR, 2.26) but not lower endoscopy (HR, 0.56).<br/><br/>“The results were not necessarily surprising given the mechanism of action of GLP-1 RAs. However, for the first time, this was shown with a clinically relevant outcome, such as aspiration pneumonia,” Dr. Rezaie said. “Aspiration during sedation can have devastating consequences, and the 0.2% difference in risk of aspiration can have a significant effect on healthcare as well.”<br/><br/><span class="Hyperlink"><a href="https://www.niddk.nih.gov/about-niddk/strategic-plans-reports/burden-of-digestive-diseases-in-united-states/indications-outcomes-gastrointestinal-endoscopy">More than 20 million endoscopies</a></span> are performed across the United States annually. Based on the assumption that about 3% of those patients are taking GLP-1 RAs, about 1200 aspiration cases per year can be prevented by raising awareness, he said.<br/><br/></p> <h2>Considering Next Steps</h2> <p>The varying risk profiles observed with separate sedation and endoscopy types point to a need for more tailored guidance in managing GLP-1 RA use before a procedure, the study authors wrote.</p> <p>Although holding the medications before endoscopy may disrupt diabetes management, the potential increased risk for aspiration could justify a change in practice, particularly for upper endoscopy and propofol-associated procedures, they added.<br/><br/>At the same time, additional studies are needed to understand the optimal drug withholding windows before endoscopies and other procedures, they concluded.<br/><br/>“We will need more data on what is the optimal duration of holding GLP-1 RAs,” Dr. Rezaie said. “But given our data and current ASA guidance, stopping these medications prior to elective procedures is the safe thing to do.”<br/><br/>For now, AGA guidance remains the same as offered in the November 2023 update, suggesting an individual approach for each patient on a GLP-1 RA rather than a “blanket statement” on how to manage all patients taking these medications.<br/><br/>“Overall, I believe that this study is important, but we require more high-level data to inform clinical decision-making regarding patients using GLP-1 receptor agonists prior to gastrointestinal endoscopy,” said Andrew Y. Wang, MD, AGAF, chief of gastroenterology and hepatology and director of interventional endoscopy at the University of Virginia in Charlottesville.<br/><br/>Dr. Wang, who wasn’t involved with this study, coauthored the AGA rapid clinical practice update. He and colleagues advised continuing with a procedure as planned for patients on GLP-1 RAs who followed standard preprocedure fasting instructions and didn’t have nausea, vomiting, dyspepsia, or abdominal distention.[[{"fid":"282039","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Andrew Y. Wang of the University of Virginia, Charlottesville","field_file_image_credit[und][0][value]":"","field_file_image_caption[und][0][value]":"Dr. Andrew Y. Wang"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]<br/><br/>Among patients with symptoms that suggest retained gastric contents, <span class="Hyperlink">rapid sequence intubation</span> may be considered, though it may not be possible in ambulatory or office-based endoscopy settings, Dr. Wang and colleagues wrote. As another option in lieu of stopping GLP-1 RAs, patients can be placed on a liquid diet for 1 day before the procedure.<br/><br/>“While this study found a signal suggesting that patients using GLP-1 RAs had an increased risk of aspiration pneumonia within 1 month following upper endoscopy or combined upper and lower endoscopy, it does not inform us if having patients stop GLP-1 RAs before endoscopic procedures — especially for a single dose — will mitigate this potential risk,” Dr. Wang said.<br/><br/>“It was also interesting that these investigators found that patients taking GLP-1 RAs who underwent lower endoscopy alone were not at increased risk for aspiration pneumonia,” Dr. Wang noted.<br/><br/>The authors didn’t report a funding source and disclosed no potential conflicts. Dr. Wang reported no relevant disclosures.<span class="end"/></p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/glp-1s-may-increase-post-endoscopy-aspiration-pneumonia-risk-2024a10007hv">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

AGA Defines Diagnostic, Treatment Approach to Cannabinoid Hyperemesis Syndrome

Article Type
Changed
Thu, 04/11/2024 - 13:11

A new American Gastroenterological Association (AGA) clinical practice update shines a light on cannabinoid hyperemesis syndrome (CHS).

CHS, which is triggered by chronic cannabis usage and manifests with GI and autonomic symptoms, is on the rise in the United States, yet underdiagnosis remains a challenge and clinical data are scarce, reported lead update panelist Alberto Rubio Tapia, MD, of Cleveland Clinic, Cleveland, Ohio, and colleagues.

RubioTapia_Alberto_OH_web.jpg
Dr. Alberto Rubio Tapia

“Although cannabis use has been reported for many decades, some of its unique adverse effects of nausea, vomiting, and abdominal pain, termed CHS, were noted relatively recently,” the panelists wrote in Gastroenterology. “The objective of this article was to help practitioners define the appropriate approach to the diagnosis and management of CHS.”

According to the update, the typical CHS patient is male with a years-long history of daily or near-daily cannabis use. Paradoxically, while cannabis use drives this condition, some patients with CHS report that cannabis use relieves their symptoms.The update describes CHS as a subtype of cyclical vomiting syndrome (CVS), and offers diagnostic criteria for CHS, reproduced below verbatim:

  • Clinical features: stereotypical episodic vomiting resembling CVS in terms of onset, with frequency 3 or more times annually;
  • Cannabis use patterns: duration of cannabis use more than 1 year before symptom onset; frequency more than 4 times per week, on average;
  • Cannabis cessation: resolution of symptoms after a period of abstinence from cannabis use for at least 6 months, or at least equal to the total duration of 3 typical vomiting cycles in that patient.

As CHS is a subtype of CVS, the update also provides an outline and management guide for this broader condition, which is characterized by four phases: inter-episodic, prodromal, emetic, and recovery.

During the inter-episodic phase, patients will have minimal or no symptoms, although almost one third will describe dyspepsia or nausea. Prophylactic medications in this period include tricyclics, mitochondrial supplements like CoQ10 and vitamin B12, NK1 antagonists, and anticonvulsants.

The prodromal phase is characterized by abdominal pain and nausea with a duration of 30-90 minutes. During this time patients may have autonomic symptoms like sweating and feeling hot or cold. Psychological symptoms may include feelings of panic and being “out of control.” Abortive medications are appropriate during this period, according to the update, like triptans and antiemetics.

Next comes the emetic phase, in which patients exhibit “relentless vomiting,” retching, abdominal pain, neurological symptoms and extreme thirst. Because an empty stomach may provide relief, inducing emesis may be considered, along with rest in a quiet dark room and supportive care.

Finally, the vomiting subsides during the recovery phase, when it is possible to restart oral intake and resume normal activities.

While this framework may be useful when managing patients with CHS, intervention should be centered around cannabis cessation, according to the update.

“For long-term management, counseling to achieve marijuana cessation and tricyclic antidepressants, such as amitriptyline, are the mainstay of therapy,” Dr. Rubio Tapia and colleagues wrote.

Advising patients to stop cannabis “cold turkey” is not recommended, they added, as this may bring on withdrawal symptoms, and it tends to be ineffective in this population, which has a high recidivism rate.

“Co-management with a psychologist or psychiatrist may be helpful for patients who have a lack of response to standard therapies or extensive psychiatric comorbidity,” the panelists wrote. “Anxiety and depression are very common associated conditions.”

Dr. Rubio Tapia and colleagues concluded with a call for more research.

“Further understanding of CHS pathophysiology and evidence-based therapies are urgently needed,” they wrote.

This update was commissioned and approved by the AGA. The update panelists disclosed relationships with Evoke Pharma, RedHill Biopharma, Takeda, and others.

Publications
Topics
Sections

A new American Gastroenterological Association (AGA) clinical practice update shines a light on cannabinoid hyperemesis syndrome (CHS).

CHS, which is triggered by chronic cannabis usage and manifests with GI and autonomic symptoms, is on the rise in the United States, yet underdiagnosis remains a challenge and clinical data are scarce, reported lead update panelist Alberto Rubio Tapia, MD, of Cleveland Clinic, Cleveland, Ohio, and colleagues.

RubioTapia_Alberto_OH_web.jpg
Dr. Alberto Rubio Tapia

“Although cannabis use has been reported for many decades, some of its unique adverse effects of nausea, vomiting, and abdominal pain, termed CHS, were noted relatively recently,” the panelists wrote in Gastroenterology. “The objective of this article was to help practitioners define the appropriate approach to the diagnosis and management of CHS.”

According to the update, the typical CHS patient is male with a years-long history of daily or near-daily cannabis use. Paradoxically, while cannabis use drives this condition, some patients with CHS report that cannabis use relieves their symptoms.The update describes CHS as a subtype of cyclical vomiting syndrome (CVS), and offers diagnostic criteria for CHS, reproduced below verbatim:

  • Clinical features: stereotypical episodic vomiting resembling CVS in terms of onset, with frequency 3 or more times annually;
  • Cannabis use patterns: duration of cannabis use more than 1 year before symptom onset; frequency more than 4 times per week, on average;
  • Cannabis cessation: resolution of symptoms after a period of abstinence from cannabis use for at least 6 months, or at least equal to the total duration of 3 typical vomiting cycles in that patient.

As CHS is a subtype of CVS, the update also provides an outline and management guide for this broader condition, which is characterized by four phases: inter-episodic, prodromal, emetic, and recovery.

During the inter-episodic phase, patients will have minimal or no symptoms, although almost one third will describe dyspepsia or nausea. Prophylactic medications in this period include tricyclics, mitochondrial supplements like CoQ10 and vitamin B12, NK1 antagonists, and anticonvulsants.

The prodromal phase is characterized by abdominal pain and nausea with a duration of 30-90 minutes. During this time patients may have autonomic symptoms like sweating and feeling hot or cold. Psychological symptoms may include feelings of panic and being “out of control.” Abortive medications are appropriate during this period, according to the update, like triptans and antiemetics.

Next comes the emetic phase, in which patients exhibit “relentless vomiting,” retching, abdominal pain, neurological symptoms and extreme thirst. Because an empty stomach may provide relief, inducing emesis may be considered, along with rest in a quiet dark room and supportive care.

Finally, the vomiting subsides during the recovery phase, when it is possible to restart oral intake and resume normal activities.

While this framework may be useful when managing patients with CHS, intervention should be centered around cannabis cessation, according to the update.

“For long-term management, counseling to achieve marijuana cessation and tricyclic antidepressants, such as amitriptyline, are the mainstay of therapy,” Dr. Rubio Tapia and colleagues wrote.

Advising patients to stop cannabis “cold turkey” is not recommended, they added, as this may bring on withdrawal symptoms, and it tends to be ineffective in this population, which has a high recidivism rate.

“Co-management with a psychologist or psychiatrist may be helpful for patients who have a lack of response to standard therapies or extensive psychiatric comorbidity,” the panelists wrote. “Anxiety and depression are very common associated conditions.”

Dr. Rubio Tapia and colleagues concluded with a call for more research.

“Further understanding of CHS pathophysiology and evidence-based therapies are urgently needed,” they wrote.

This update was commissioned and approved by the AGA. The update panelists disclosed relationships with Evoke Pharma, RedHill Biopharma, Takeda, and others.

A new American Gastroenterological Association (AGA) clinical practice update shines a light on cannabinoid hyperemesis syndrome (CHS).

CHS, which is triggered by chronic cannabis usage and manifests with GI and autonomic symptoms, is on the rise in the United States, yet underdiagnosis remains a challenge and clinical data are scarce, reported lead update panelist Alberto Rubio Tapia, MD, of Cleveland Clinic, Cleveland, Ohio, and colleagues.

RubioTapia_Alberto_OH_web.jpg
Dr. Alberto Rubio Tapia

“Although cannabis use has been reported for many decades, some of its unique adverse effects of nausea, vomiting, and abdominal pain, termed CHS, were noted relatively recently,” the panelists wrote in Gastroenterology. “The objective of this article was to help practitioners define the appropriate approach to the diagnosis and management of CHS.”

According to the update, the typical CHS patient is male with a years-long history of daily or near-daily cannabis use. Paradoxically, while cannabis use drives this condition, some patients with CHS report that cannabis use relieves their symptoms.The update describes CHS as a subtype of cyclical vomiting syndrome (CVS), and offers diagnostic criteria for CHS, reproduced below verbatim:

  • Clinical features: stereotypical episodic vomiting resembling CVS in terms of onset, with frequency 3 or more times annually;
  • Cannabis use patterns: duration of cannabis use more than 1 year before symptom onset; frequency more than 4 times per week, on average;
  • Cannabis cessation: resolution of symptoms after a period of abstinence from cannabis use for at least 6 months, or at least equal to the total duration of 3 typical vomiting cycles in that patient.

As CHS is a subtype of CVS, the update also provides an outline and management guide for this broader condition, which is characterized by four phases: inter-episodic, prodromal, emetic, and recovery.

During the inter-episodic phase, patients will have minimal or no symptoms, although almost one third will describe dyspepsia or nausea. Prophylactic medications in this period include tricyclics, mitochondrial supplements like CoQ10 and vitamin B12, NK1 antagonists, and anticonvulsants.

The prodromal phase is characterized by abdominal pain and nausea with a duration of 30-90 minutes. During this time patients may have autonomic symptoms like sweating and feeling hot or cold. Psychological symptoms may include feelings of panic and being “out of control.” Abortive medications are appropriate during this period, according to the update, like triptans and antiemetics.

Next comes the emetic phase, in which patients exhibit “relentless vomiting,” retching, abdominal pain, neurological symptoms and extreme thirst. Because an empty stomach may provide relief, inducing emesis may be considered, along with rest in a quiet dark room and supportive care.

Finally, the vomiting subsides during the recovery phase, when it is possible to restart oral intake and resume normal activities.

While this framework may be useful when managing patients with CHS, intervention should be centered around cannabis cessation, according to the update.

“For long-term management, counseling to achieve marijuana cessation and tricyclic antidepressants, such as amitriptyline, are the mainstay of therapy,” Dr. Rubio Tapia and colleagues wrote.

Advising patients to stop cannabis “cold turkey” is not recommended, they added, as this may bring on withdrawal symptoms, and it tends to be ineffective in this population, which has a high recidivism rate.

“Co-management with a psychologist or psychiatrist may be helpful for patients who have a lack of response to standard therapies or extensive psychiatric comorbidity,” the panelists wrote. “Anxiety and depression are very common associated conditions.”

Dr. Rubio Tapia and colleagues concluded with a call for more research.

“Further understanding of CHS pathophysiology and evidence-based therapies are urgently needed,” they wrote.

This update was commissioned and approved by the AGA. The update panelists disclosed relationships with Evoke Pharma, RedHill Biopharma, Takeda, and others.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167488</fileName> <TBEID>0C04F4C6.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F4C6</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Gastro_Rubio_CHS</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240411T125900</QCDate> <firstPublished>20240411T130827</firstPublished> <LastPublished>20240411T130827</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240411T130826</CMSDate> <articleSource>FROM GASTROENTEROLOGY </articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>A new American Gastroenterological Association (AGA) clinical practice update shines a light on cannabinoid hyperemesis syndrome (CHS)</metaDescription> <articlePDF/> <teaserImage>301096</teaserImage> <teaser>Cannabinoid hyperemesis syndrome is triggered by chronic cannabis usage and manifests with GI and autonomic symptoms.</teaser> <title>AGA Defines Diagnostic, Treatment Approach to Cannabinoid Hyperemesis Syndrome</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">347</term> <term>27442</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401280d.jpg</altRep> <description role="drol:caption">Dr. Alberto Rubio Tapia</description> <description role="drol:credit">Courtesy Cleveland Clinic</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>AGA Defines Diagnostic, Treatment Approach to Cannabinoid Hyperemesis Syndrome</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">A new American Gastroenterological Association (AGA) clinical practice update shines a light on cannabinoid hyperemesis syndrome (CHS)</span>.</p> <p>CHS, which is triggered by chronic cannabis usage and manifests with GI and autonomic symptoms, is on the rise in the United States, yet underdiagnosis remains a challenge and clinical data are scarce, reported lead update panelist <a href="https://health.usnews.com/doctors/alberto-rubio-tapia-920838">Alberto Rubio Tapia, MD</a>, of Cleveland Clinic, Cleveland, Ohio, and colleagues.<br/><br/>[[{"fid":"301096","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Alberto Rubio Tapia, Cleveland Clinic","field_file_image_credit[und][0][value]":"Courtesy Cleveland Clinic","field_file_image_caption[und][0][value]":"Dr. Alberto Rubio Tapia"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]“Although cannabis use has been reported for many decades, some of its unique adverse effects of nausea, vomiting, and abdominal pain, termed CHS, were noted relatively recently,” the <a href="https://www.gastrojournal.org/article/S0016-5085(24)00127-6/fulltext">panelists wrote</a> in <em>Gastroenterology</em>. “The objective of this article was to help practitioners define the appropriate approach to the diagnosis and management of CHS.”<br/><br/>According to the update, the typical CHS patient is male with a years-long history of daily or near-daily cannabis use. Paradoxically, while cannabis use drives this condition, some patients with CHS report that cannabis use relieves their symptoms.The update describes CHS as a subtype of cyclical vomiting syndrome (CVS), and offers diagnostic criteria for CHS, reproduced below verbatim:</p> <ul class="body"> <li><strong>Clinical features:</strong> stereotypical episodic vomiting resembling CVS in terms of onset, with frequency 3 or more times annually;</li> <li><strong>Cannabis use patterns:</strong> duration of cannabis use more than 1 year before symptom onset; frequency more than 4 times per week, on average;</li> <li><strong>Cannabis cessation:</strong> resolution of symptoms after a period of abstinence from cannabis use for at least 6 months, or at least equal to the total duration of 3 typical vomiting cycles in that patient. </li> </ul> <p>As CHS is a subtype of CVS, the update also provides an outline and management guide for this broader condition, which is characterized by four phases: inter-episodic, prodromal, emetic, and recovery.<br/><br/>During the inter-episodic phase, patients will have minimal or no symptoms, although almost one third will describe dyspepsia or nausea. Prophylactic medications in this period include tricyclics, mitochondrial supplements like CoQ10 and vitamin B12, NK1 antagonists, and anticonvulsants.<br/><br/>The prodromal phase is characterized by abdominal pain and nausea with a duration of 30-90 minutes. During this time patients may have autonomic symptoms like sweating and feeling hot or cold. Psychological symptoms may include feelings of panic and being “out of control.” Abortive medications are appropriate during this period, according to the update, like triptans and antiemetics.<br/><br/>Next comes the emetic phase, in which patients exhibit “relentless vomiting,” retching, abdominal pain, neurological symptoms and extreme thirst. Because an empty stomach may provide relief, inducing emesis may be considered, along with rest in a quiet dark room and supportive care.<br/><br/>Finally, the vomiting subsides during the recovery phase, when it is possible to restart oral intake and resume normal activities.<br/><br/>While this framework may be useful when managing patients with CHS, intervention should be centered around cannabis cessation, according to the update. <br/><br/>“For long-term management, counseling to achieve marijuana cessation and tricyclic antidepressants, such as amitriptyline, are the mainstay of therapy,” Dr. Rubio Tapia and colleagues wrote.<br/><br/>Advising patients to stop cannabis “cold turkey” is not recommended, they added, as this may bring on withdrawal symptoms, and it tends to be ineffective in this population, which has a high recidivism rate.<br/><br/>“Co-management with a psychologist or psychiatrist may be helpful for patients who have a lack of response to standard therapies or extensive psychiatric comorbidity,” the panelists wrote. “Anxiety and depression are very common associated conditions.”<br/><br/>Dr. Rubio Tapia and colleagues concluded with a call for more research.<br/><br/>“Further understanding of CHS pathophysiology and evidence-based therapies are urgently needed,” they wrote.<br/><br/>This update was commissioned and approved by the AGA. The update panelists disclosed relationships with Evoke Pharma, RedHill Biopharma, Takeda, and others.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

TRAIL-targeting Therapies Still Hold Promise in Cholangiocarcinoma

Further Investigation is Warranted
Article Type
Changed
Thu, 04/11/2024 - 13:07

Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–targeting therapies still hold promise for treating cholangiocarcinoma (CCA) despite disappointing results in previous preclinical research, primarily due to the adaptive resistance and unexpected immune modulation, according to investigators.

Those prior studies evaluated a combination of immunotherapy and TRAIL agonism, but selective TRAIL antagonism shows greater potential via dual ligand/receptor (TRAIL/TRAIL-R) targeting to block immunosuppression, reported lead author Emilien J. Loeuillard, PhD, of Mayo Clinic, Rochester, Minnesota, and colleagues.

Loeuillard_Emilien_MN_web.jpg
Dr. Emilien J. Loeuillard

“The TRAIL/TRAIL-R system has garnered considerable interest in cancer biology, especially as a potential anticancer therapy,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology. “However, TRAIL-R agonists have had very limited anticancer activity in human beings, challenging this concept of TRAIL as an anticancer agent.”

This may be because they were working in the wrong direction, Dr. Loeuillard and colleagues suggested, citing recent work linking TRAIL with tumor proliferation and invasion, possibly via modification of the tumor immune microenvironment.

Exact mechanisms of modification, however, remain unclear. While TRAIL has been associated with tumor-promoting effects like induction of a promyeloid secretome in adenocarcinoma, it has also been linked with anticancer effects like activation of natural killer cells and cytotoxic T lymphocytes.

“Thus, the potency and hierarchy of TRAIL anticancer vs procancer processes in cancer biology has yet to be defined,” the investigators wrote.

While TRAIL ligation of cognate receptors has been previously investigated and shown to trigger proapoptotic signaling pathways, noncanonical TRAIL-mediated signaling remains largely unexplored, particularly in CCA.

The present study evaluated TRAIL biology in CCA using immunocompetent mouse models.

These experiments showed that noncanonical TRAIL signaling immunosuppresses the tumor microenvironment by increasing quantity and activity of myeloid-derived suppressor cells (MDSCs). Blocking noncanonical TRAIL signaling by selective deletion of TRAIL-R in immune cells had significantly reduced tumor volumes alongside fewer MDSCs, driven by FLICE inhibitory protein (cFLIP)-dependent nuclear factor kappa-B activation (NF-kappa-B) in MDSCs, which has antiapoptotic activity. While MDSCs present one possible target in this chain of immunosuppression, “therapeutic strategies for targeting MDSCs are limited,” the investigators wrote, noting that available myeloid modulators have fallen short in clinical trials.

Instead, cFLIP may be a convincing option, they suggested, as targeting cFLIP can sensitize cancer cells to proapoptotic TRAIL signaling. What’s more, cFLIP appears to protect MDSCs from TRAIL-mediated apoptosis, so taking out this barrier could render MDSCs susceptible to therapy.

“Our studies suggest that switching prosurvival/proliferation TRAIL signaling to canonical proapoptotic TRAIL signaling will promote MDSC apoptosis, which in turn has therapeutic implications for CCA suppression,” the investigators wrote.

Hope therefore remains for targeting TRAIL in patients with CCA, but with selective antagonism instead of agonism, as previously attempted.

“In summary, our findings support the role of selective therapeutic targeting of TRAIL-positive cancer cells in an effort to block TRAIL/TRAIL-R–mediated tumor immunosuppression,” Dr. Loeuillard and colleagues concluded.

This study was funded by the Cholangiocarcinoma Foundation and the Mayo Clinic Eagles 5th District Cancer Telethon Funds for Research Fellowship Program, the CTSA/National Center for Advancing Translational Science, the National Institutes of Health/National Cancer Institute, and others. The investigators disclosed no conflicts of interest.

Body

 

The dismal response of cholangiocarcinoma to immune checkpoint inhibitors (ICI) is particularly concerning, as it impedes the adoption of combination regimens, now standard in most solid tumors. Strategies modulating selective genes involved in the tumor inflammatory environment and tumor cell viability, including those within the tumor necrosis factor superfamily, parallel the mechanism of action of ICI and present a double-edged sword due to the context-dependent pro- and/or anticancer effects of their canonical and/or phantom roles.

Recent investigations suggest that selectively antagonizing TRAIL via (TRAIL/TRAIL-R) targeting may be more effective than agonism. Dr. Ilyas’ group from Mayo Clinic delved into the potential of TRAIL in cancer biology, particularly in CCA, shedding light on the complexities of TRAIL’s role in cancer, where both procancer and anticancer effects are observed.

[[{"fid":"301094","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"}},"attributes":{"alt":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Importantly, they unveiled that noncanonical TRAIL signaling contributes to suppressing the tumor microenvironment by promoting the accumulation of myeloid-derived suppressor cells which can be further mitigated by a novel strategy targeting FLICE inhibitory protein to increase cancer cell sensitivity to proapoptotic TRAIL signaling, presenting a potential avenue for therapeutic intervention as well as biomarkers predictive of TRAIL response for CCA.

Further investigation is warranted to explore how TRAIL/TRAIL-R therapy can be effectively combined with other broad-spectrum and/or targeted therapies to maximize selective toxicity to CCA cells, sparing the nonmalignant tissue, thereby extending the lifespan of CCA patients as well as assessing its preventive potential in predisposed premalignant stages, including cholestasis patients.
 

Sungjin Ko, DVM, PhD, is assistant professor in the Division of Experimental Pathology at the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. He is also a member of the Pittsburgh Liver Research Center. He reported no conflicts of interest.

Publications
Topics
Sections
Body

 

The dismal response of cholangiocarcinoma to immune checkpoint inhibitors (ICI) is particularly concerning, as it impedes the adoption of combination regimens, now standard in most solid tumors. Strategies modulating selective genes involved in the tumor inflammatory environment and tumor cell viability, including those within the tumor necrosis factor superfamily, parallel the mechanism of action of ICI and present a double-edged sword due to the context-dependent pro- and/or anticancer effects of their canonical and/or phantom roles.

Recent investigations suggest that selectively antagonizing TRAIL via (TRAIL/TRAIL-R) targeting may be more effective than agonism. Dr. Ilyas’ group from Mayo Clinic delved into the potential of TRAIL in cancer biology, particularly in CCA, shedding light on the complexities of TRAIL’s role in cancer, where both procancer and anticancer effects are observed.

[[{"fid":"301094","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"}},"attributes":{"alt":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Importantly, they unveiled that noncanonical TRAIL signaling contributes to suppressing the tumor microenvironment by promoting the accumulation of myeloid-derived suppressor cells which can be further mitigated by a novel strategy targeting FLICE inhibitory protein to increase cancer cell sensitivity to proapoptotic TRAIL signaling, presenting a potential avenue for therapeutic intervention as well as biomarkers predictive of TRAIL response for CCA.

Further investigation is warranted to explore how TRAIL/TRAIL-R therapy can be effectively combined with other broad-spectrum and/or targeted therapies to maximize selective toxicity to CCA cells, sparing the nonmalignant tissue, thereby extending the lifespan of CCA patients as well as assessing its preventive potential in predisposed premalignant stages, including cholestasis patients.
 

Sungjin Ko, DVM, PhD, is assistant professor in the Division of Experimental Pathology at the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. He is also a member of the Pittsburgh Liver Research Center. He reported no conflicts of interest.

Body

 

The dismal response of cholangiocarcinoma to immune checkpoint inhibitors (ICI) is particularly concerning, as it impedes the adoption of combination regimens, now standard in most solid tumors. Strategies modulating selective genes involved in the tumor inflammatory environment and tumor cell viability, including those within the tumor necrosis factor superfamily, parallel the mechanism of action of ICI and present a double-edged sword due to the context-dependent pro- and/or anticancer effects of their canonical and/or phantom roles.

Recent investigations suggest that selectively antagonizing TRAIL via (TRAIL/TRAIL-R) targeting may be more effective than agonism. Dr. Ilyas’ group from Mayo Clinic delved into the potential of TRAIL in cancer biology, particularly in CCA, shedding light on the complexities of TRAIL’s role in cancer, where both procancer and anticancer effects are observed.

[[{"fid":"301094","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"}},"attributes":{"alt":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Importantly, they unveiled that noncanonical TRAIL signaling contributes to suppressing the tumor microenvironment by promoting the accumulation of myeloid-derived suppressor cells which can be further mitigated by a novel strategy targeting FLICE inhibitory protein to increase cancer cell sensitivity to proapoptotic TRAIL signaling, presenting a potential avenue for therapeutic intervention as well as biomarkers predictive of TRAIL response for CCA.

Further investigation is warranted to explore how TRAIL/TRAIL-R therapy can be effectively combined with other broad-spectrum and/or targeted therapies to maximize selective toxicity to CCA cells, sparing the nonmalignant tissue, thereby extending the lifespan of CCA patients as well as assessing its preventive potential in predisposed premalignant stages, including cholestasis patients.
 

Sungjin Ko, DVM, PhD, is assistant professor in the Division of Experimental Pathology at the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. He is also a member of the Pittsburgh Liver Research Center. He reported no conflicts of interest.

Title
Further Investigation is Warranted
Further Investigation is Warranted

Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–targeting therapies still hold promise for treating cholangiocarcinoma (CCA) despite disappointing results in previous preclinical research, primarily due to the adaptive resistance and unexpected immune modulation, according to investigators.

Those prior studies evaluated a combination of immunotherapy and TRAIL agonism, but selective TRAIL antagonism shows greater potential via dual ligand/receptor (TRAIL/TRAIL-R) targeting to block immunosuppression, reported lead author Emilien J. Loeuillard, PhD, of Mayo Clinic, Rochester, Minnesota, and colleagues.

Loeuillard_Emilien_MN_web.jpg
Dr. Emilien J. Loeuillard

“The TRAIL/TRAIL-R system has garnered considerable interest in cancer biology, especially as a potential anticancer therapy,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology. “However, TRAIL-R agonists have had very limited anticancer activity in human beings, challenging this concept of TRAIL as an anticancer agent.”

This may be because they were working in the wrong direction, Dr. Loeuillard and colleagues suggested, citing recent work linking TRAIL with tumor proliferation and invasion, possibly via modification of the tumor immune microenvironment.

Exact mechanisms of modification, however, remain unclear. While TRAIL has been associated with tumor-promoting effects like induction of a promyeloid secretome in adenocarcinoma, it has also been linked with anticancer effects like activation of natural killer cells and cytotoxic T lymphocytes.

“Thus, the potency and hierarchy of TRAIL anticancer vs procancer processes in cancer biology has yet to be defined,” the investigators wrote.

While TRAIL ligation of cognate receptors has been previously investigated and shown to trigger proapoptotic signaling pathways, noncanonical TRAIL-mediated signaling remains largely unexplored, particularly in CCA.

The present study evaluated TRAIL biology in CCA using immunocompetent mouse models.

These experiments showed that noncanonical TRAIL signaling immunosuppresses the tumor microenvironment by increasing quantity and activity of myeloid-derived suppressor cells (MDSCs). Blocking noncanonical TRAIL signaling by selective deletion of TRAIL-R in immune cells had significantly reduced tumor volumes alongside fewer MDSCs, driven by FLICE inhibitory protein (cFLIP)-dependent nuclear factor kappa-B activation (NF-kappa-B) in MDSCs, which has antiapoptotic activity. While MDSCs present one possible target in this chain of immunosuppression, “therapeutic strategies for targeting MDSCs are limited,” the investigators wrote, noting that available myeloid modulators have fallen short in clinical trials.

Instead, cFLIP may be a convincing option, they suggested, as targeting cFLIP can sensitize cancer cells to proapoptotic TRAIL signaling. What’s more, cFLIP appears to protect MDSCs from TRAIL-mediated apoptosis, so taking out this barrier could render MDSCs susceptible to therapy.

“Our studies suggest that switching prosurvival/proliferation TRAIL signaling to canonical proapoptotic TRAIL signaling will promote MDSC apoptosis, which in turn has therapeutic implications for CCA suppression,” the investigators wrote.

Hope therefore remains for targeting TRAIL in patients with CCA, but with selective antagonism instead of agonism, as previously attempted.

“In summary, our findings support the role of selective therapeutic targeting of TRAIL-positive cancer cells in an effort to block TRAIL/TRAIL-R–mediated tumor immunosuppression,” Dr. Loeuillard and colleagues concluded.

This study was funded by the Cholangiocarcinoma Foundation and the Mayo Clinic Eagles 5th District Cancer Telethon Funds for Research Fellowship Program, the CTSA/National Center for Advancing Translational Science, the National Institutes of Health/National Cancer Institute, and others. The investigators disclosed no conflicts of interest.

Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–targeting therapies still hold promise for treating cholangiocarcinoma (CCA) despite disappointing results in previous preclinical research, primarily due to the adaptive resistance and unexpected immune modulation, according to investigators.

Those prior studies evaluated a combination of immunotherapy and TRAIL agonism, but selective TRAIL antagonism shows greater potential via dual ligand/receptor (TRAIL/TRAIL-R) targeting to block immunosuppression, reported lead author Emilien J. Loeuillard, PhD, of Mayo Clinic, Rochester, Minnesota, and colleagues.

Loeuillard_Emilien_MN_web.jpg
Dr. Emilien J. Loeuillard

“The TRAIL/TRAIL-R system has garnered considerable interest in cancer biology, especially as a potential anticancer therapy,” the investigators wrote in Cellular and Molecular Gastroenterology and Hepatology. “However, TRAIL-R agonists have had very limited anticancer activity in human beings, challenging this concept of TRAIL as an anticancer agent.”

This may be because they were working in the wrong direction, Dr. Loeuillard and colleagues suggested, citing recent work linking TRAIL with tumor proliferation and invasion, possibly via modification of the tumor immune microenvironment.

Exact mechanisms of modification, however, remain unclear. While TRAIL has been associated with tumor-promoting effects like induction of a promyeloid secretome in adenocarcinoma, it has also been linked with anticancer effects like activation of natural killer cells and cytotoxic T lymphocytes.

“Thus, the potency and hierarchy of TRAIL anticancer vs procancer processes in cancer biology has yet to be defined,” the investigators wrote.

While TRAIL ligation of cognate receptors has been previously investigated and shown to trigger proapoptotic signaling pathways, noncanonical TRAIL-mediated signaling remains largely unexplored, particularly in CCA.

The present study evaluated TRAIL biology in CCA using immunocompetent mouse models.

These experiments showed that noncanonical TRAIL signaling immunosuppresses the tumor microenvironment by increasing quantity and activity of myeloid-derived suppressor cells (MDSCs). Blocking noncanonical TRAIL signaling by selective deletion of TRAIL-R in immune cells had significantly reduced tumor volumes alongside fewer MDSCs, driven by FLICE inhibitory protein (cFLIP)-dependent nuclear factor kappa-B activation (NF-kappa-B) in MDSCs, which has antiapoptotic activity. While MDSCs present one possible target in this chain of immunosuppression, “therapeutic strategies for targeting MDSCs are limited,” the investigators wrote, noting that available myeloid modulators have fallen short in clinical trials.

Instead, cFLIP may be a convincing option, they suggested, as targeting cFLIP can sensitize cancer cells to proapoptotic TRAIL signaling. What’s more, cFLIP appears to protect MDSCs from TRAIL-mediated apoptosis, so taking out this barrier could render MDSCs susceptible to therapy.

“Our studies suggest that switching prosurvival/proliferation TRAIL signaling to canonical proapoptotic TRAIL signaling will promote MDSC apoptosis, which in turn has therapeutic implications for CCA suppression,” the investigators wrote.

Hope therefore remains for targeting TRAIL in patients with CCA, but with selective antagonism instead of agonism, as previously attempted.

“In summary, our findings support the role of selective therapeutic targeting of TRAIL-positive cancer cells in an effort to block TRAIL/TRAIL-R–mediated tumor immunosuppression,” Dr. Loeuillard and colleagues concluded.

This study was funded by the Cholangiocarcinoma Foundation and the Mayo Clinic Eagles 5th District Cancer Telethon Funds for Research Fellowship Program, the CTSA/National Center for Advancing Translational Science, the National Institutes of Health/National Cancer Institute, and others. The investigators disclosed no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167487</fileName> <TBEID>0C04F4B1.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F4B1</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CMGH_Loeuillard_CCA</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240411T124002</QCDate> <firstPublished>20240411T130244</firstPublished> <LastPublished>20240411T130244</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240411T130243</CMSDate> <articleSource>FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–targeting therapies still hold promise for treating cholangiocarcinoma (CCA) despite disappointi</metaDescription> <articlePDF/> <teaserImage>301095</teaserImage> <teaser>“Our findings support the role of selective therapeutic targeting of TRAIL-positive cancer cells,” said Dr. Emilien Loeuillard.</teaser> <title>TRAIL-targeting Therapies Still Hold Promise in Cholangiocarcinoma</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">344</term> <term>39703</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012809.jpg</altRep> <description role="drol:caption">Dr. Emilien J. Loeuillard</description> <description role="drol:credit">Courtesy Dr. Emilien J. Loeuillard</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012808.jpg</altRep> <description role="drol:caption">Dr. Sungjin Ko</description> <description role="drol:credit">Courtesy University of Pittsburgh School of Medicine</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>TRAIL-targeting Therapies Still Hold Promise in Cholangiocarcinoma</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL)–targeting therapies still hold promise for treating cholangiocarcinoma (CCA) despite disappointing results in previous preclinical research,</span> primarily due to the adaptive resistance and unexpected immune modulation, according to investigators.</p> <p>Those prior studies evaluated a combination of immunotherapy and TRAIL agonism, but selective TRAIL antagonism shows greater potential via dual ligand/receptor (TRAIL/TRAIL-R) targeting to block immunosuppression, reported lead author <a href="https://www.linkedin.com/in/emilien-loeuillard/">Emilien J. Loeuillard, PhD</a>, of Mayo Clinic, Rochester, Minnesota, and colleagues.<br/><br/>[[{"fid":"301095","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"Courtesy Dr. Emilien J. Loeuillard","field_file_image_caption[und][0][value]":"Dr. Emilien J. Loeuillard"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]“The TRAIL/TRAIL-R system has garnered considerable interest in cancer biology, especially as a potential anticancer therapy,” the investigators wrote in <em><a href="https://www.cmghjournal.org/article/S2352-345X(24)00005-5/fulltext">Cellular and Molecular Gastroenterology and Hepatology</a></em>. “However, TRAIL-R agonists have had very limited anticancer activity in human beings, challenging this concept of TRAIL as an anticancer agent.”<br/><br/>This may be because they were working in the wrong direction, Dr. Loeuillard and colleagues suggested, citing recent work linking TRAIL with tumor proliferation and invasion, possibly via modification of the tumor immune microenvironment.<br/><br/>Exact mechanisms of modification, however, remain unclear. While TRAIL has been associated with tumor-promoting effects like induction of a promyeloid secretome in adenocarcinoma, it has also been linked with anticancer effects like activation of natural killer cells and cytotoxic T lymphocytes.<br/><br/>“Thus, the potency and hierarchy of TRAIL anticancer vs procancer processes in cancer biology has yet to be defined,” the investigators wrote.<br/><br/>While TRAIL ligation of cognate receptors has been previously investigated and shown to trigger proapoptotic signaling pathways, noncanonical TRAIL-mediated signaling remains largely unexplored, particularly in CCA. <br/><br/>The present study evaluated TRAIL biology in CCA using immunocompetent mouse models.<br/><br/>These experiments showed that noncanonical TRAIL signaling immunosuppresses the tumor microenvironment by increasing quantity and activity of myeloid-derived suppressor cells (MDSCs). Blocking noncanonical TRAIL signaling by selective deletion of TRAIL-R in immune cells had significantly reduced tumor volumes alongside fewer MDSCs, driven by FLICE inhibitory protein (cFLIP)-dependent nuclear factor kappa-B activation (NF-kappa-B) in MDSCs, which has antiapoptotic activity. While MDSCs present one possible target in this chain of immunosuppression, “therapeutic strategies for targeting MDSCs are limited,” the investigators wrote, noting that available myeloid modulators have fallen short in clinical trials.<br/><br/>Instead, cFLIP may be a convincing option, they suggested, as targeting cFLIP can sensitize cancer cells to proapoptotic TRAIL signaling. What’s more, cFLIP appears to protect MDSCs from TRAIL-mediated apoptosis, so taking out this barrier could render MDSCs susceptible to therapy.<br/><br/>“Our studies suggest that switching prosurvival/proliferation TRAIL signaling to canonical proapoptotic TRAIL signaling will promote MDSC apoptosis, which in turn has therapeutic implications for CCA suppression,” the investigators wrote.<br/><br/>Hope therefore remains for targeting TRAIL in patients with CCA, but with selective antagonism instead of agonism, as previously attempted.<br/><br/>“In summary, our findings support the role of selective therapeutic targeting of TRAIL-positive cancer cells in an effort to block TRAIL/TRAIL-R–mediated tumor immunosuppression,” Dr. Loeuillard and colleagues concluded.<br/><br/>This study was funded by the Cholangiocarcinoma Foundation and the Mayo Clinic Eagles 5th District Cancer Telethon Funds for Research Fellowship Program, the CTSA/National Center for Advancing Translational Science, the National Institutes of Health/National Cancer Institute, and others. The investigators disclosed no conflicts of interest.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>views</itemRole> <itemClass>text</itemClass> <title>Further Investigation is Warranted</title> <deck/> </itemMeta> <itemContent> <p>The dismal response of cholangiocarcinoma to immune checkpoint inhibitors (ICI) is particularly concerning, as it impedes the adoption of combination regimens, now standard in most solid tumors. Strategies modulating selective genes involved in the tumor inflammatory environment and tumor cell viability, including those within the tumor necrosis factor superfamily, parallel the mechanism of action of ICI and present a double-edged sword due to the context-dependent pro- and/or anticancer effects of their canonical and/or phantom roles.</p> <p>Recent investigations suggest that selectively antagonizing TRAIL via (TRAIL/TRAIL-R) targeting may be more effective than agonism. Dr. Ilyas’ group from Mayo Clinic delved into the potential of TRAIL in cancer biology, particularly in CCA, shedding light on the complexities of TRAIL’s role in cancer, where both procancer and anticancer effects are observed.<br/><br/>[[{"fid":"301094","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Division of Experimental Pathology at the University of Pittsburgh School of Medicine. He is also a member of the Pittsburgh Liver Research Center","field_file_image_credit[und][0][value]":"Courtesy University of Pittsburgh School of Medicine","field_file_image_caption[und][0][value]":"Dr. Sungjin Ko"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]Importantly, they unveiled that noncanonical TRAIL signaling contributes to suppressing the tumor microenvironment by promoting the accumulation of myeloid-derived suppressor cells which can be further mitigated by a novel strategy targeting FLICE inhibitory protein to increase cancer cell sensitivity to proapoptotic TRAIL signaling, presenting a potential avenue for therapeutic intervention as well as biomarkers predictive of TRAIL response for CCA.<br/><br/>Further investigation is warranted to explore how TRAIL/TRAIL-R therapy can be effectively combined with other broad-spectrum and/or targeted therapies to maximize selective toxicity to CCA cells, sparing the nonmalignant tissue, thereby extending the lifespan of CCA patients as well as assessing its preventive potential in predisposed premalignant stages, including cholestasis patients.<br/><br/></p> <p><em> <em>Sungjin Ko, DVM, PhD, is assistant professor in the Division of Experimental Pathology at the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. He is also a member of the Pittsburgh Liver Research Center. He reported no conflicts of interest.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

IBD: Histologic Inflammation Linked With Lower Female Fertility

More Aggressive Treatment of Inflammation Is Appropriate
Article Type
Changed
Thu, 04/11/2024 - 12:34

Histologic inflammation in women with inflammatory bowel disease (IBD) may lead to reduced fertility, according to a Swedish nationwide cohort study.

Reduced fertility was linked with histologic inflammation even in the absence of clinical disease activity, highlighting the importance of achieving deep remission in women planning pregnancy, reported lead author Karl Mårild, MD, PhD, of Sahlgrenska Academy, Gothenburg, Sweden, and colleagues.

“Reduced female fertility (ie, number of live births) is believed to be primarily confined to women with clinically active IBD, especially in Crohn’s disease (CD), where symptoms may inhibit sexual activity, and inflammation may affect the fallopian tubes and ovaries,” the investigators wrote in Gastroenterology. “Despite the increasing appreciation of histologic activity in IBD, its association with female fertility has not been clarified, including whether histologic activity in the absence of clinical disease activity impairs fertility.”

Marild_Karl_SWE_2_web.jpg
Dr. Karl Mårild

Dr. Mårild and colleagues aimed to address this knowledge gap by analyzing fertility rates and histologic inflammation or IBD activity in two cohorts of women with IBD aged 15-44 years. The first group included approximately 21,000 women with and without histologic inflammation from 1990 to 2016. The second group included approximately 25,000 women with or without IBD clinical activity from 2006 to 2020. In each group, the relationship between fertility and IBD was compared with fertility in matched general population comparator individuals.

This approach showed that clinical IBD activity was associated with an adjusted fertility rate ratio (aFRR) of 0.76 (95% CI, 0.72-0.79), which equates to one fewer child per six women with 10 years of clinical activity. Impacts on fertility were similar for UC (aFRR, 0.75) and CD (aFRR, 0.76).

“Fertility rates were notably reduced during periods of clinical IBD activity and, contrary to a generally accepted belief, equally reduced in clinically active UC and CD,” the investigators wrote. “Besides inflammation, clinically active IBD may reduce fertility through psychological mechanisms (eg, depression), dyspareunia (especially in perianal CD), bowel pain, urgency, and other symptoms that hinder sexual activity.”

Compared with histologic remission, histologic inflammation was also associated with reduced fertility (aFRR, 0.90). This means that in periods of histologic inflammation, 6.35 live births occurred per 100 person-years of follow-up, compared with 7.09 lives births for periods of histologic remission. This amounts to one fewer child per 14 women with 10 years of histologic inflammation.

Finally, the study revealed that, in women with clinically quiescent IBD, those with histologic inflammation had significantly reduced fertility, compared with those in histologic remission (aFRR, 0.85). This association persisted after controlling for contraceptive use.

“Even if histologic inflammation was associated with an overall modest fertility reduction … its impact on the individual might be substantial, with potential ramifications beyond reproductive health, given that reduced female fertility is linked to poor quality of life and mental health,” Dr. Mårild and colleagues wrote. “At a societal level, involuntary childlessness causes high and increasing costs, highlighting the need to focus on preventable causes of reduced fertility.”

The investigators suggested that inflammation may be driving infertility by reducing ovulation and fertilization, or by reducing endometrial receptivity, which increases risk of pregnancy loss.

“This is the first study, to our knowledge, to show reduced fertility during histologic inflammation in IBD compared to histologic remission,” the investigators wrote. “Our findings suggest that achieving histologic remission may improve the fertility of women with IBD, even in the absence of clinically defined disease activity.”

The investigators disclosed relationships with AbbVie, Pfizer, Janssen, and others.

Body

 

The importance of controlling inflammation to ensure a healthy pregnancy cannot be overstated. With regard to fertility, the literature has emphasized that surgery has been the major risk factor for decreasing fertility in both ulcerative colitis and Crohn’s disease. Disease activity has been more influential on Crohn’s disease versus ulcerative colitis. Other factors such as voluntary childlessness, premature ovarian failure, and malnutrition can also play a role. There have been data to show that anti–tumor necrosis factor use increases the chances of successful implantation for women with sub-fertility who do not have concomitant IBD, perhaps by decreasing inflammation in the pelvis.

[[{"fid":"301082","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"}},"attributes":{"alt":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Histologic activity has recently become the ultimate therapeutic goal. Up until now this has not been studied in the context of fertility. We know that clinical disease indices do not necessarily correlate with endoscopic appearance, and when trying to optimize pregnancy outcomes it might behoove us to know what our goal is — absence of clinical, endoscopic, or histologic inflammation. However, perfection might be the enemy of good: One fewer child per 14 women with 10 years of histologic inflammation is hard to put into clinical context. I think these results are important to again emphasize that we should not stop therapy in the preconception period, with a goal of controlling as much inflammation as possible. Perhaps the best way to use these data are to counsel women with unsuccessful attempts at pregnancy and, in the absence of any other factors, that more aggressive treatment of inflammation is appropriate.

Sunanda Kane, MD, MSPH, AGAF, is based in the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota. She reports serving as a consultant to Boehringer Ingelheim, Bristol Myers Squibb, Fresenius Kabi, Gilead, Janssen, and Takeda. She is also Section Editor for IBD for UptoDate.

Publications
Topics
Sections
Body

 

The importance of controlling inflammation to ensure a healthy pregnancy cannot be overstated. With regard to fertility, the literature has emphasized that surgery has been the major risk factor for decreasing fertility in both ulcerative colitis and Crohn’s disease. Disease activity has been more influential on Crohn’s disease versus ulcerative colitis. Other factors such as voluntary childlessness, premature ovarian failure, and malnutrition can also play a role. There have been data to show that anti–tumor necrosis factor use increases the chances of successful implantation for women with sub-fertility who do not have concomitant IBD, perhaps by decreasing inflammation in the pelvis.

[[{"fid":"301082","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"}},"attributes":{"alt":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Histologic activity has recently become the ultimate therapeutic goal. Up until now this has not been studied in the context of fertility. We know that clinical disease indices do not necessarily correlate with endoscopic appearance, and when trying to optimize pregnancy outcomes it might behoove us to know what our goal is — absence of clinical, endoscopic, or histologic inflammation. However, perfection might be the enemy of good: One fewer child per 14 women with 10 years of histologic inflammation is hard to put into clinical context. I think these results are important to again emphasize that we should not stop therapy in the preconception period, with a goal of controlling as much inflammation as possible. Perhaps the best way to use these data are to counsel women with unsuccessful attempts at pregnancy and, in the absence of any other factors, that more aggressive treatment of inflammation is appropriate.

Sunanda Kane, MD, MSPH, AGAF, is based in the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota. She reports serving as a consultant to Boehringer Ingelheim, Bristol Myers Squibb, Fresenius Kabi, Gilead, Janssen, and Takeda. She is also Section Editor for IBD for UptoDate.

Body

 

The importance of controlling inflammation to ensure a healthy pregnancy cannot be overstated. With regard to fertility, the literature has emphasized that surgery has been the major risk factor for decreasing fertility in both ulcerative colitis and Crohn’s disease. Disease activity has been more influential on Crohn’s disease versus ulcerative colitis. Other factors such as voluntary childlessness, premature ovarian failure, and malnutrition can also play a role. There have been data to show that anti–tumor necrosis factor use increases the chances of successful implantation for women with sub-fertility who do not have concomitant IBD, perhaps by decreasing inflammation in the pelvis.

[[{"fid":"301082","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"}},"attributes":{"alt":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","class":"media-element file-medstat-image-flush-right","data-delta":"2"}}]]Histologic activity has recently become the ultimate therapeutic goal. Up until now this has not been studied in the context of fertility. We know that clinical disease indices do not necessarily correlate with endoscopic appearance, and when trying to optimize pregnancy outcomes it might behoove us to know what our goal is — absence of clinical, endoscopic, or histologic inflammation. However, perfection might be the enemy of good: One fewer child per 14 women with 10 years of histologic inflammation is hard to put into clinical context. I think these results are important to again emphasize that we should not stop therapy in the preconception period, with a goal of controlling as much inflammation as possible. Perhaps the best way to use these data are to counsel women with unsuccessful attempts at pregnancy and, in the absence of any other factors, that more aggressive treatment of inflammation is appropriate.

Sunanda Kane, MD, MSPH, AGAF, is based in the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota. She reports serving as a consultant to Boehringer Ingelheim, Bristol Myers Squibb, Fresenius Kabi, Gilead, Janssen, and Takeda. She is also Section Editor for IBD for UptoDate.

Title
More Aggressive Treatment of Inflammation Is Appropriate
More Aggressive Treatment of Inflammation Is Appropriate

Histologic inflammation in women with inflammatory bowel disease (IBD) may lead to reduced fertility, according to a Swedish nationwide cohort study.

Reduced fertility was linked with histologic inflammation even in the absence of clinical disease activity, highlighting the importance of achieving deep remission in women planning pregnancy, reported lead author Karl Mårild, MD, PhD, of Sahlgrenska Academy, Gothenburg, Sweden, and colleagues.

“Reduced female fertility (ie, number of live births) is believed to be primarily confined to women with clinically active IBD, especially in Crohn’s disease (CD), where symptoms may inhibit sexual activity, and inflammation may affect the fallopian tubes and ovaries,” the investigators wrote in Gastroenterology. “Despite the increasing appreciation of histologic activity in IBD, its association with female fertility has not been clarified, including whether histologic activity in the absence of clinical disease activity impairs fertility.”

Marild_Karl_SWE_2_web.jpg
Dr. Karl Mårild

Dr. Mårild and colleagues aimed to address this knowledge gap by analyzing fertility rates and histologic inflammation or IBD activity in two cohorts of women with IBD aged 15-44 years. The first group included approximately 21,000 women with and without histologic inflammation from 1990 to 2016. The second group included approximately 25,000 women with or without IBD clinical activity from 2006 to 2020. In each group, the relationship between fertility and IBD was compared with fertility in matched general population comparator individuals.

This approach showed that clinical IBD activity was associated with an adjusted fertility rate ratio (aFRR) of 0.76 (95% CI, 0.72-0.79), which equates to one fewer child per six women with 10 years of clinical activity. Impacts on fertility were similar for UC (aFRR, 0.75) and CD (aFRR, 0.76).

“Fertility rates were notably reduced during periods of clinical IBD activity and, contrary to a generally accepted belief, equally reduced in clinically active UC and CD,” the investigators wrote. “Besides inflammation, clinically active IBD may reduce fertility through psychological mechanisms (eg, depression), dyspareunia (especially in perianal CD), bowel pain, urgency, and other symptoms that hinder sexual activity.”

Compared with histologic remission, histologic inflammation was also associated with reduced fertility (aFRR, 0.90). This means that in periods of histologic inflammation, 6.35 live births occurred per 100 person-years of follow-up, compared with 7.09 lives births for periods of histologic remission. This amounts to one fewer child per 14 women with 10 years of histologic inflammation.

Finally, the study revealed that, in women with clinically quiescent IBD, those with histologic inflammation had significantly reduced fertility, compared with those in histologic remission (aFRR, 0.85). This association persisted after controlling for contraceptive use.

“Even if histologic inflammation was associated with an overall modest fertility reduction … its impact on the individual might be substantial, with potential ramifications beyond reproductive health, given that reduced female fertility is linked to poor quality of life and mental health,” Dr. Mårild and colleagues wrote. “At a societal level, involuntary childlessness causes high and increasing costs, highlighting the need to focus on preventable causes of reduced fertility.”

The investigators suggested that inflammation may be driving infertility by reducing ovulation and fertilization, or by reducing endometrial receptivity, which increases risk of pregnancy loss.

“This is the first study, to our knowledge, to show reduced fertility during histologic inflammation in IBD compared to histologic remission,” the investigators wrote. “Our findings suggest that achieving histologic remission may improve the fertility of women with IBD, even in the absence of clinically defined disease activity.”

The investigators disclosed relationships with AbbVie, Pfizer, Janssen, and others.

Histologic inflammation in women with inflammatory bowel disease (IBD) may lead to reduced fertility, according to a Swedish nationwide cohort study.

Reduced fertility was linked with histologic inflammation even in the absence of clinical disease activity, highlighting the importance of achieving deep remission in women planning pregnancy, reported lead author Karl Mårild, MD, PhD, of Sahlgrenska Academy, Gothenburg, Sweden, and colleagues.

“Reduced female fertility (ie, number of live births) is believed to be primarily confined to women with clinically active IBD, especially in Crohn’s disease (CD), where symptoms may inhibit sexual activity, and inflammation may affect the fallopian tubes and ovaries,” the investigators wrote in Gastroenterology. “Despite the increasing appreciation of histologic activity in IBD, its association with female fertility has not been clarified, including whether histologic activity in the absence of clinical disease activity impairs fertility.”

Marild_Karl_SWE_2_web.jpg
Dr. Karl Mårild

Dr. Mårild and colleagues aimed to address this knowledge gap by analyzing fertility rates and histologic inflammation or IBD activity in two cohorts of women with IBD aged 15-44 years. The first group included approximately 21,000 women with and without histologic inflammation from 1990 to 2016. The second group included approximately 25,000 women with or without IBD clinical activity from 2006 to 2020. In each group, the relationship between fertility and IBD was compared with fertility in matched general population comparator individuals.

This approach showed that clinical IBD activity was associated with an adjusted fertility rate ratio (aFRR) of 0.76 (95% CI, 0.72-0.79), which equates to one fewer child per six women with 10 years of clinical activity. Impacts on fertility were similar for UC (aFRR, 0.75) and CD (aFRR, 0.76).

“Fertility rates were notably reduced during periods of clinical IBD activity and, contrary to a generally accepted belief, equally reduced in clinically active UC and CD,” the investigators wrote. “Besides inflammation, clinically active IBD may reduce fertility through psychological mechanisms (eg, depression), dyspareunia (especially in perianal CD), bowel pain, urgency, and other symptoms that hinder sexual activity.”

Compared with histologic remission, histologic inflammation was also associated with reduced fertility (aFRR, 0.90). This means that in periods of histologic inflammation, 6.35 live births occurred per 100 person-years of follow-up, compared with 7.09 lives births for periods of histologic remission. This amounts to one fewer child per 14 women with 10 years of histologic inflammation.

Finally, the study revealed that, in women with clinically quiescent IBD, those with histologic inflammation had significantly reduced fertility, compared with those in histologic remission (aFRR, 0.85). This association persisted after controlling for contraceptive use.

“Even if histologic inflammation was associated with an overall modest fertility reduction … its impact on the individual might be substantial, with potential ramifications beyond reproductive health, given that reduced female fertility is linked to poor quality of life and mental health,” Dr. Mårild and colleagues wrote. “At a societal level, involuntary childlessness causes high and increasing costs, highlighting the need to focus on preventable causes of reduced fertility.”

The investigators suggested that inflammation may be driving infertility by reducing ovulation and fertilization, or by reducing endometrial receptivity, which increases risk of pregnancy loss.

“This is the first study, to our knowledge, to show reduced fertility during histologic inflammation in IBD compared to histologic remission,” the investigators wrote. “Our findings suggest that achieving histologic remission may improve the fertility of women with IBD, even in the absence of clinically defined disease activity.”

The investigators disclosed relationships with AbbVie, Pfizer, Janssen, and others.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>167484</fileName> <TBEID>0C04F455.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F455</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Gastro_Marild_IBD fertility</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240411T115330</QCDate> <firstPublished>20240411T123052</firstPublished> <LastPublished>20240411T123052</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240411T123051</CMSDate> <articleSource>FROM GASTROENTEROLOGY</articleSource> <facebookInfo/> <meetingNumber/> <byline>Will Pass</byline> <bylineText>WILL PASS</bylineText> <bylineFull>WILL PASS</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>News</newsDocType> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Histologic inflammation in women with inflammatory bowel disease (IBD) may lead to reduced fertility</metaDescription> <articlePDF/> <teaserImage>301080</teaserImage> <teaser>In women with clinically quiescent IBD, those with histologic inflammation had significantly reduced fertility, compared with those in histologic remission.</teaser> <title>IBD: Histologic Inflammation Linked With Lower Female Fertility</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>gih</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">345</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240127fa.jpg</altRep> <description role="drol:caption">Dr. Karl Mårild</description> <description role="drol:credit">Courtesy University of Gothenburg</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240127f9.jpg</altRep> <description role="drol:caption">Dr. Sunanda Kane</description> <description role="drol:credit">CourtesyMayo Clinic</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>IBD: Histologic Inflammation Linked With Lower Female Fertility</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Histologic inflammation in women with inflammatory bowel disease (IBD) may lead to reduced fertility</span>, according to a Swedish nationwide cohort study.</p> <p>Reduced fertility was linked with histologic inflammation even in the absence of clinical disease activity, highlighting the importance of achieving deep remission in women planning pregnancy, reported lead author <a href="https://www.gu.se/en/about/find-staff/karlmarild">Karl Mårild, MD, PhD</a>, of Sahlgrenska Academy, Gothenburg, Sweden, and colleagues.<br/><br/>“Reduced female fertility (ie, number of live births) is believed to be primarily confined to women with clinically active IBD, especially in Crohn’s disease (CD), where symptoms may inhibit sexual activity, and inflammation may affect the fallopian tubes and ovaries,” the investigators wrote in <a href="https://www.gastrojournal.org/article/S0016-5085(24)00055-6/fulltext?referrer=https%3A%2F%2Fpubmed.ncbi.nlm.nih.gov%2F">G<em>astroenterology</em></a>. “Despite the increasing appreciation of histologic activity in IBD, its association with female fertility has not been clarified, including whether histologic activity in the absence of clinical disease activity impairs fertility.”<br/><br/>[[{"fid":"301080","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Karl Mårild, University of Gothenburg, Sweden","field_file_image_credit[und][0][value]":"Courtesy University of Gothenburg","field_file_image_caption[und][0][value]":"Dr. Karl Mårild"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]Dr. Mårild and colleagues aimed to address this knowledge gap by analyzing fertility rates and histologic inflammation or IBD activity in two cohorts of women with IBD aged 15-44 years. The first group included approximately 21,000 women with and without histologic inflammation from 1990 to 2016. The second group included approximately 25,000 women with or without IBD clinical activity from 2006 to 2020. In each group, the relationship between fertility and IBD was compared with fertility in matched general population comparator individuals.<br/><br/>This approach showed that clinical IBD activity was associated with an adjusted fertility rate ratio (aFRR) of 0.76 (95% CI, 0.72-0.79), which equates to one fewer child per six women with 10 years of clinical activity. Impacts on fertility were similar for UC (aFRR, 0.75) and CD (aFRR, 0.76).<br/><br/>“Fertility rates were notably reduced during periods of clinical IBD activity and, contrary to a generally accepted belief, equally reduced in clinically active UC and CD,” the investigators wrote. “Besides inflammation, clinically active IBD may reduce fertility through psychological mechanisms (eg, depression), dyspareunia (especially in perianal CD), bowel pain, urgency, and other symptoms that hinder sexual activity.”<br/><br/>Compared with histologic remission, histologic inflammation was also associated with reduced fertility (aFRR, 0.90). This means that in periods of histologic inflammation, 6.35 live births occurred per 100 person-years of follow-up, compared with 7.09 lives births for periods of histologic remission. This amounts to one fewer child per 14 women with 10 years of histologic inflammation. <br/><br/>Finally, the study revealed that, in women with clinically quiescent IBD, those with histologic inflammation had significantly reduced fertility, compared with those in histologic remission (aFRR, 0.85). This association persisted after controlling for contraceptive use.<br/><br/>“Even if histologic inflammation was associated with an overall modest fertility reduction … its impact on the individual might be substantial, with potential ramifications beyond reproductive health, given that reduced female fertility is linked to poor quality of life and mental health,” Dr. Mårild and colleagues wrote. “At a societal level, involuntary childlessness causes high and increasing costs, highlighting the need to focus on preventable causes of reduced fertility.”<br/><br/>The investigators suggested that inflammation may be driving infertility by reducing ovulation and fertilization, or by reducing endometrial receptivity, which increases risk of pregnancy loss.<br/><br/>“This is the first study, to our knowledge, to show reduced fertility during histologic inflammation in IBD compared to histologic remission,” the investigators wrote. “Our findings suggest that achieving histologic remission may improve the fertility of women with IBD, even in the absence of clinically defined disease activity.”<br/><br/>The investigators disclosed relationships with AbbVie, Pfizer, Janssen, and others.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>views</itemRole> <itemClass>text</itemClass> <title>More Aggressive Treatment of Inflammation Is Appropriate</title> <deck/> </itemMeta> <itemContent> <p>The importance of controlling inflammation to ensure a healthy pregnancy cannot be overstated. With regard to fertility, the literature has emphasized that surgery has been the major risk factor for decreasing fertility in both ulcerative colitis and Crohn’s disease. Disease activity has been more influential on Crohn’s disease versus ulcerative colitis. Other factors such as voluntary childlessness, premature ovarian failure, and malnutrition can also play a role. There have been data to show that anti–tumor necrosis factor use increases the chances of successful implantation for women with sub-fertility who do not have concomitant IBD, perhaps by decreasing inflammation in the pelvis. </p> <p>[[{"fid":"301082","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Sunanda Kane, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota","field_file_image_credit[und][0][value]":"CourtesyMayo Clinic","field_file_image_caption[und][0][value]":"Dr. Sunanda Kane"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]Histologic activity has recently become the ultimate therapeutic goal. Up until now this has not been studied in the context of fertility. We know that clinical disease indices do not necessarily correlate with endoscopic appearance, and when trying to optimize pregnancy outcomes it might behoove us to know what our goal is — absence of clinical, endoscopic, or histologic inflammation. However, perfection might be the enemy of good: One fewer child per 14 women with 10 years of histologic inflammation is hard to put into clinical context. I think these results are important to again emphasize that we should not stop therapy in the preconception period, with a goal of controlling as much inflammation as possible. Perhaps the best way to use these data are to counsel women with unsuccessful attempts at pregnancy and, in the absence of any other factors, that more aggressive treatment of inflammation is appropriate.</p> <p><em> <em>Sunanda Kane, MD, MSPH, AGAF, is based in the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota. She reports serving as a consultant to Boehringer Ingelheim, Bristol Myers Squibb, Fresenius Kabi, Gilead, Janssen, and Takeda. She is also Section Editor for IBD for UptoDate.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article