LayerRx Mapping ID
315
Slot System
Featured Buckets
Featured Buckets Admin

May 2024 – ICYMI

Article Type
Changed
Fri, 05/03/2024 - 16:28

 

Gastroenterology

January 2024

Hirano I, et al; ASCENT WORKING GROUP. Ascending to New Heights for Novel Therapeutics for Eosinophilic Esophagitis. Gastroenterology. 2024 Jan;166(1):1-10. doi: 10.1053/j.gastro.2023.09.004. Epub 2023 Sep 9. PMID: 37690772; PMCID: PMC10872872.



Åkerström JH, et al. Antireflux Surgery Versus Antireflux Medication and Risk of Esophageal Adenocarcinoma in Patients With Barrett’s Esophagus. Gastroenterology. 2024 Jan;166(1):132-138.e3. doi: 10.1053/j.gastro.2023.08.050. Epub 2023 Sep 9. PMID: 37690771.



Barnes EL, et al; AGA Clinical Guidelines Committee. AGA Clinical Practice Guideline on the Management of Pouchitis and Inflammatory Pouch Disorders. Gastroenterology. 2024 Jan;166(1):59-85. doi: 10.1053/j.gastro.2023.10.015. PMID: 38128971.

February 2024

Yoo HW, et al. Helicobacter pylori Treatment and Gastric Cancer Risk After Endoscopic Resection of Dysplasia: A Nationwide Cohort Study. Gastroenterology. 2024 Feb;166(2):313-322.e3. doi: 10.1053/j.gastro.2023.10.013. Epub 2023 Oct 18. PMID: 37863270.



Yang J, et al. High Soluble Fiber Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites in Mice. Gastroenterology. 2024 Feb;166(2):323-337.e7. doi: 10.1053/j.gastro.2023.10.012. Epub 2023 Oct 18. PMID: 37858797.



Young E, et al. Texture and Color Enhancement Imaging Improves Colonic Adenoma Detection: A Multicenter Randomized Controlled Trial. Gastroenterology. 2024 Feb;166(2):338-340.e3. doi: 10.1053/j.gastro.2023.10.008. Epub 2023 Oct 14. PMID: 37839498.
 

Clinical Gastroenterology and Hepatology

January 2024

Overbeek KA, et al; Dutch Familial Pancreatic Cancer Surveillance Study work group. Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: Incidence, Growth Rate, and Malignancy Risk. Clin Gastroenterol Hepatol. 2024 Jan;22(1):62-71.e7. doi: 10.1016/j.cgh.2023.03.035. Epub 2023 Apr 7. PMID: 37031711.



Reddy CA, et al. Achalasia is Strongly Associated With Eosinophilic Esophagitis and Other Allergic Disorders. Clin Gastroenterol Hepatol. 2024 Jan;22(1):34-41.e2. doi: 10.1016/j.cgh.2023.06.013. Epub 2023 Jun 28. PMID: 37391057; PMCID: PMC10753026.

Thiruvengadam NR, et al. The Clinical Impact and Cost-Effectiveness of Surveillance of Incidentally Detected Gastric Intestinal Metaplasia: A Microsimulation Analysis. Clin Gastroenterol Hepatol. 2024 Jan;22(1):51-61. doi: 10.1016/j.cgh.2023.05.028. Epub 2023 Jun 9. Erratum in: Clin Gastroenterol Hepatol. 2024 Jan 19;: PMID: 37302442.

February 2024

Goodoory VC, et al. Systematic Review and Meta-analysis: Efficacy of Mesalamine in Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):243-251.e5. doi: 10.1016/j.cgh.2023.02.014. Epub 2023 Feb 27. PMID: 36858143.

Brenner DM, et al. Development and Current State of Digital Therapeutics for Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):222-234. doi: 10.1016/j.cgh.2023.09.013. Epub 2023 Sep 22. PMID: 37743035.
 

Techniques and Innovations in Gastrointestinal Endoscopy

January 2024

Ramirez PR, et al. Gaps and Improvement Opportunities in Post-Colonoscopy Communication. Tech Innov Gastrointest Endosc. 2024 Jan;26(1):90-92. doi: 10.1016/j.tige.2023.10.001. Epub 2023 Oct 22.



Gonzaga ER, et al. Gastric Peroral Endoscopic Myotomy (G-POEM) for the Management of Gastroparesis. Tech Innov Gastrointest Endosc. 2024 Jan; 26(1): 46-55. doi: 10.1016/j.tige.2023.09.002. Epub 2023 Oct 13.



Wang D, et al. Sphincterotomy vs Sham Procedure for Pain Relief in Sphincter of Oddi Dysfunction: Systematic Review and Meta-analysis. Tech Innov Gastrointest Endosc. 2024 Jan;26(1): 30-37. doi: 10.1016/j.tige.2023.10.003. Epub 2023 Nov 8.
 

Gastro Hep Advances

January 2024

Adeniran E, et al. Intense and Sustained Alcohol Consumption Associated With Acute Pancreatitis Warrants Early Intervention. Gastro Hep Advances. 2024 Jan;3(1):61-63. doi: 10.1016/j.gastha.2023.08.017. Epub 2023 Sep 2.



Alkhouri N, et al. A Novel Prescription Digital Therapeutic Option for the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease. Gastro Hep Advances. 2024 Jan;3(1): 9-16. doi: 10.1016/j.gastha.2023.08.019. Epub 2023 Oct 1.

Publications
Topics
Sections

 

Gastroenterology

January 2024

Hirano I, et al; ASCENT WORKING GROUP. Ascending to New Heights for Novel Therapeutics for Eosinophilic Esophagitis. Gastroenterology. 2024 Jan;166(1):1-10. doi: 10.1053/j.gastro.2023.09.004. Epub 2023 Sep 9. PMID: 37690772; PMCID: PMC10872872.



Åkerström JH, et al. Antireflux Surgery Versus Antireflux Medication and Risk of Esophageal Adenocarcinoma in Patients With Barrett’s Esophagus. Gastroenterology. 2024 Jan;166(1):132-138.e3. doi: 10.1053/j.gastro.2023.08.050. Epub 2023 Sep 9. PMID: 37690771.



Barnes EL, et al; AGA Clinical Guidelines Committee. AGA Clinical Practice Guideline on the Management of Pouchitis and Inflammatory Pouch Disorders. Gastroenterology. 2024 Jan;166(1):59-85. doi: 10.1053/j.gastro.2023.10.015. PMID: 38128971.

February 2024

Yoo HW, et al. Helicobacter pylori Treatment and Gastric Cancer Risk After Endoscopic Resection of Dysplasia: A Nationwide Cohort Study. Gastroenterology. 2024 Feb;166(2):313-322.e3. doi: 10.1053/j.gastro.2023.10.013. Epub 2023 Oct 18. PMID: 37863270.



Yang J, et al. High Soluble Fiber Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites in Mice. Gastroenterology. 2024 Feb;166(2):323-337.e7. doi: 10.1053/j.gastro.2023.10.012. Epub 2023 Oct 18. PMID: 37858797.



Young E, et al. Texture and Color Enhancement Imaging Improves Colonic Adenoma Detection: A Multicenter Randomized Controlled Trial. Gastroenterology. 2024 Feb;166(2):338-340.e3. doi: 10.1053/j.gastro.2023.10.008. Epub 2023 Oct 14. PMID: 37839498.
 

Clinical Gastroenterology and Hepatology

January 2024

Overbeek KA, et al; Dutch Familial Pancreatic Cancer Surveillance Study work group. Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: Incidence, Growth Rate, and Malignancy Risk. Clin Gastroenterol Hepatol. 2024 Jan;22(1):62-71.e7. doi: 10.1016/j.cgh.2023.03.035. Epub 2023 Apr 7. PMID: 37031711.



Reddy CA, et al. Achalasia is Strongly Associated With Eosinophilic Esophagitis and Other Allergic Disorders. Clin Gastroenterol Hepatol. 2024 Jan;22(1):34-41.e2. doi: 10.1016/j.cgh.2023.06.013. Epub 2023 Jun 28. PMID: 37391057; PMCID: PMC10753026.

Thiruvengadam NR, et al. The Clinical Impact and Cost-Effectiveness of Surveillance of Incidentally Detected Gastric Intestinal Metaplasia: A Microsimulation Analysis. Clin Gastroenterol Hepatol. 2024 Jan;22(1):51-61. doi: 10.1016/j.cgh.2023.05.028. Epub 2023 Jun 9. Erratum in: Clin Gastroenterol Hepatol. 2024 Jan 19;: PMID: 37302442.

February 2024

Goodoory VC, et al. Systematic Review and Meta-analysis: Efficacy of Mesalamine in Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):243-251.e5. doi: 10.1016/j.cgh.2023.02.014. Epub 2023 Feb 27. PMID: 36858143.

Brenner DM, et al. Development and Current State of Digital Therapeutics for Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):222-234. doi: 10.1016/j.cgh.2023.09.013. Epub 2023 Sep 22. PMID: 37743035.
 

Techniques and Innovations in Gastrointestinal Endoscopy

January 2024

Ramirez PR, et al. Gaps and Improvement Opportunities in Post-Colonoscopy Communication. Tech Innov Gastrointest Endosc. 2024 Jan;26(1):90-92. doi: 10.1016/j.tige.2023.10.001. Epub 2023 Oct 22.



Gonzaga ER, et al. Gastric Peroral Endoscopic Myotomy (G-POEM) for the Management of Gastroparesis. Tech Innov Gastrointest Endosc. 2024 Jan; 26(1): 46-55. doi: 10.1016/j.tige.2023.09.002. Epub 2023 Oct 13.



Wang D, et al. Sphincterotomy vs Sham Procedure for Pain Relief in Sphincter of Oddi Dysfunction: Systematic Review and Meta-analysis. Tech Innov Gastrointest Endosc. 2024 Jan;26(1): 30-37. doi: 10.1016/j.tige.2023.10.003. Epub 2023 Nov 8.
 

Gastro Hep Advances

January 2024

Adeniran E, et al. Intense and Sustained Alcohol Consumption Associated With Acute Pancreatitis Warrants Early Intervention. Gastro Hep Advances. 2024 Jan;3(1):61-63. doi: 10.1016/j.gastha.2023.08.017. Epub 2023 Sep 2.



Alkhouri N, et al. A Novel Prescription Digital Therapeutic Option for the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease. Gastro Hep Advances. 2024 Jan;3(1): 9-16. doi: 10.1016/j.gastha.2023.08.019. Epub 2023 Oct 1.

 

Gastroenterology

January 2024

Hirano I, et al; ASCENT WORKING GROUP. Ascending to New Heights for Novel Therapeutics for Eosinophilic Esophagitis. Gastroenterology. 2024 Jan;166(1):1-10. doi: 10.1053/j.gastro.2023.09.004. Epub 2023 Sep 9. PMID: 37690772; PMCID: PMC10872872.



Åkerström JH, et al. Antireflux Surgery Versus Antireflux Medication and Risk of Esophageal Adenocarcinoma in Patients With Barrett’s Esophagus. Gastroenterology. 2024 Jan;166(1):132-138.e3. doi: 10.1053/j.gastro.2023.08.050. Epub 2023 Sep 9. PMID: 37690771.



Barnes EL, et al; AGA Clinical Guidelines Committee. AGA Clinical Practice Guideline on the Management of Pouchitis and Inflammatory Pouch Disorders. Gastroenterology. 2024 Jan;166(1):59-85. doi: 10.1053/j.gastro.2023.10.015. PMID: 38128971.

February 2024

Yoo HW, et al. Helicobacter pylori Treatment and Gastric Cancer Risk After Endoscopic Resection of Dysplasia: A Nationwide Cohort Study. Gastroenterology. 2024 Feb;166(2):313-322.e3. doi: 10.1053/j.gastro.2023.10.013. Epub 2023 Oct 18. PMID: 37863270.



Yang J, et al. High Soluble Fiber Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites in Mice. Gastroenterology. 2024 Feb;166(2):323-337.e7. doi: 10.1053/j.gastro.2023.10.012. Epub 2023 Oct 18. PMID: 37858797.



Young E, et al. Texture and Color Enhancement Imaging Improves Colonic Adenoma Detection: A Multicenter Randomized Controlled Trial. Gastroenterology. 2024 Feb;166(2):338-340.e3. doi: 10.1053/j.gastro.2023.10.008. Epub 2023 Oct 14. PMID: 37839498.
 

Clinical Gastroenterology and Hepatology

January 2024

Overbeek KA, et al; Dutch Familial Pancreatic Cancer Surveillance Study work group. Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: Incidence, Growth Rate, and Malignancy Risk. Clin Gastroenterol Hepatol. 2024 Jan;22(1):62-71.e7. doi: 10.1016/j.cgh.2023.03.035. Epub 2023 Apr 7. PMID: 37031711.



Reddy CA, et al. Achalasia is Strongly Associated With Eosinophilic Esophagitis and Other Allergic Disorders. Clin Gastroenterol Hepatol. 2024 Jan;22(1):34-41.e2. doi: 10.1016/j.cgh.2023.06.013. Epub 2023 Jun 28. PMID: 37391057; PMCID: PMC10753026.

Thiruvengadam NR, et al. The Clinical Impact and Cost-Effectiveness of Surveillance of Incidentally Detected Gastric Intestinal Metaplasia: A Microsimulation Analysis. Clin Gastroenterol Hepatol. 2024 Jan;22(1):51-61. doi: 10.1016/j.cgh.2023.05.028. Epub 2023 Jun 9. Erratum in: Clin Gastroenterol Hepatol. 2024 Jan 19;: PMID: 37302442.

February 2024

Goodoory VC, et al. Systematic Review and Meta-analysis: Efficacy of Mesalamine in Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):243-251.e5. doi: 10.1016/j.cgh.2023.02.014. Epub 2023 Feb 27. PMID: 36858143.

Brenner DM, et al. Development and Current State of Digital Therapeutics for Irritable Bowel Syndrome. Clin Gastroenterol Hepatol. 2024 Feb;22(2):222-234. doi: 10.1016/j.cgh.2023.09.013. Epub 2023 Sep 22. PMID: 37743035.
 

Techniques and Innovations in Gastrointestinal Endoscopy

January 2024

Ramirez PR, et al. Gaps and Improvement Opportunities in Post-Colonoscopy Communication. Tech Innov Gastrointest Endosc. 2024 Jan;26(1):90-92. doi: 10.1016/j.tige.2023.10.001. Epub 2023 Oct 22.



Gonzaga ER, et al. Gastric Peroral Endoscopic Myotomy (G-POEM) for the Management of Gastroparesis. Tech Innov Gastrointest Endosc. 2024 Jan; 26(1): 46-55. doi: 10.1016/j.tige.2023.09.002. Epub 2023 Oct 13.



Wang D, et al. Sphincterotomy vs Sham Procedure for Pain Relief in Sphincter of Oddi Dysfunction: Systematic Review and Meta-analysis. Tech Innov Gastrointest Endosc. 2024 Jan;26(1): 30-37. doi: 10.1016/j.tige.2023.10.003. Epub 2023 Nov 8.
 

Gastro Hep Advances

January 2024

Adeniran E, et al. Intense and Sustained Alcohol Consumption Associated With Acute Pancreatitis Warrants Early Intervention. Gastro Hep Advances. 2024 Jan;3(1):61-63. doi: 10.1016/j.gastha.2023.08.017. Epub 2023 Sep 2.



Alkhouri N, et al. A Novel Prescription Digital Therapeutic Option for the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease. Gastro Hep Advances. 2024 Jan;3(1): 9-16. doi: 10.1016/j.gastha.2023.08.019. Epub 2023 Oct 1.

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>167942</fileName> <TBEID>0C04FEDB.SIG</TBEID> <TBUniqueIdentifier>MD_0C04FEDB</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240503T160409</QCDate> <firstPublished>20240503T162424</firstPublished> <LastPublished>20240503T162424</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240503T162424</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>(None)</byline> <bylineText/> <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>In case you missed it: Recent studies published in the journals of the American Gastroenterology Association.</metaDescription> <articlePDF/> <teaserImage/> <teaser> <span class="tag metaDescription">In case you missed it: Recent studies published in the journals of the American Gastroenterology Association.</span> </teaser> <title>May 2024 – ICYMI</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>47431</term> <term canonical="true">46646</term> </sections> <topics> <term canonical="true">347</term> <term>344</term> <term>345</term> <term>39703</term> <term>39702</term> <term>343</term> <term>346</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>May 2024 – ICYMI</title> <deck/> </itemMeta> <itemContent> <h2>Gastroenterology</h2> <p> <strong>January 2024</strong> </p> <p>Hirano I, et al; ASCENT WORKING GROUP. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.09.004">Ascending to New Heights for Novel Therapeutics for Eosinophilic Esophagitis</a></span>. Gastroenterology. 2024 Jan;166(1):1-10. doi: 10.1053/j.gastro.2023.09.004. Epub 2023 Sep 9. PMID: 37690772; PMCID: PMC10872872.<br/><br/><br/><br/>Åkerström JH, et al. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.08.050">Antireflux Surgery Versus Antireflux Medication and Risk of Esophageal Adenocarcinoma in Patients With Barrett’s Esophagus</a></span>. Gastroenterology. 2024 Jan;166(1):132-138.e3. doi: 10.1053/j.gastro.2023.08.050. Epub 2023 Sep 9. PMID: 37690771.<br/><br/><br/><br/>Barnes EL, et al; AGA Clinical Guidelines Committee. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.10.015">AGA Clinical Practice Guideline on the Management of Pouchitis and Inflammatory Pouch Disorders</a></span>. Gastroenterology. 2024 Jan;166(1):59-85. doi: 10.1053/j.gastro.2023.10.015. PMID: 38128971.</p> <p><strong>February 2024</strong><br/><br/>Yoo HW, et al. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.10.013">Helicobacter pylori Treatment and Gastric Cancer Risk After Endoscopic Resection of Dysplasia: A Nationwide Cohort Study</a></span>. Gastroenterology. 2024 Feb;166(2):313-322.e3. doi: 10.1053/j.gastro.2023.10.013. Epub 2023 Oct 18. PMID: 37863270.<br/><br/><br/><br/>Yang J, et al. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.10.012">High Soluble Fiber Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites in Mice</a></span>. Gastroenterology. 2024 Feb;166(2):323-337.e7. doi: 10.1053/j.gastro.2023.10.012. Epub 2023 Oct 18. PMID: 37858797.<br/><br/><br/><br/>Young E, et al. <span class="Hyperlink"><a href="https://doi.org/10.1053/j.gastro.2023.10.008">Texture and Color Enhancement Imaging Improves Colonic Adenoma Detection: A Multicenter Randomized Controlled Trial</a></span>. Gastroenterology. 2024 Feb;166(2):338-340.e3. doi: 10.1053/j.gastro.2023.10.008. Epub 2023 Oct 14. PMID: 37839498.<br/><br/></p> <h2>Clinical Gastroenterology and Hepatology</h2> <p><strong>January 2024</strong><br/><br/>Overbeek KA, et al; Dutch Familial Pancreatic Cancer Surveillance Study work group. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.03.035">Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: Incidence, Growth Rate, and Malignancy Risk</a></span>. Clin Gastroenterol Hepatol. 2024 Jan;22(1):62-71.e7. doi: 10.1016/j.cgh.2023.03.035. Epub 2023 Apr 7. PMID: 37031711.</p> <p><br/><br/>Reddy CA, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.06.013">Achalasia is Strongly Associated With Eosinophilic Esophagitis and Other Allergic Disorders</a></span>. Clin Gastroenterol Hepatol. 2024 Jan;22(1):34-41.e2. doi: 10.1016/j.cgh.2023.06.013. Epub 2023 Jun 28. PMID: 37391057; PMCID: PMC10753026.<br/><br/>Thiruvengadam NR, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.05.028">The Clinical Impact and Cost-Effectiveness of Surveillance of Incidentally Detected Gastric Intestinal Metaplasia: A Microsimulation Analysis</a></span>. Clin Gastroenterol Hepatol. 2024 Jan;22(1):51-61. doi: 10.1016/j.cgh.2023.05.028. Epub 2023 Jun 9. Erratum in: Clin Gastroenterol Hepatol. 2024 Jan 19;: PMID: 37302442.</p> <p><strong>February 2024</strong><br/><br/>Goodoory VC, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.02.014">Systematic Review and Meta-analysis: Efficacy of Mesalamine in Irritable Bowel Syndrome</a></span>. Clin Gastroenterol Hepatol. 2024 Feb;22(2):243-251.e5. doi: 10.1016/j.cgh.2023.02.014. Epub 2023 Feb 27. PMID: 36858143.<br/><br/>Brenner DM, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.cgh.2023.09.013">Development and Current State of Digital Therapeutics for Irritable Bowel Syndrome</a></span>. Clin Gastroenterol Hepatol. 2024 Feb;22(2):222-234. doi: 10.1016/j.cgh.2023.09.013. Epub 2023 Sep 22. PMID: 37743035.<br/><br/></p> <h2>Techniques and Innovations in Gastrointestinal Endoscopy</h2> <p><strong>January 2024</strong><br/><br/>Ramirez PR, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.tige.2023.10.001">Gaps and Improvement Opportunities in Post-Colonoscopy Communication</a></span>. Tech Innov Gastrointest Endosc. 2024 Jan;26(1):90-92. doi: 10.1016/j.tige.2023.10.001. Epub 2023 Oct 22.</p> <p><br/><br/>Gonzaga ER, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.tige.2023.09.002">Gastric Peroral Endoscopic Myotomy (G-POEM) for the Management of Gastroparesis</a></span>. Tech Innov Gastrointest Endosc. 2024 Jan; 26(1): 46-55. doi: 10.1016/j.tige.2023.09.002. Epub 2023 Oct 13.<br/><br/><br/><br/>Wang D, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.tige.2023.10.003">Sphincterotomy vs Sham Procedure for Pain Relief in Sphincter of Oddi Dysfunction: Systematic Review and Meta-analysis</a></span>. Tech Innov Gastrointest Endosc. 2024 Jan;26(1): 30-37. doi: 10.1016/j.tige.2023.10.003. Epub 2023 Nov 8.<br/><br/></p> <h2>Gastro Hep Advances</h2> <p><strong>January 2024</strong><br/><br/>Adeniran E, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.gastha.2023.08.017">Intense and Sustained Alcohol Consumption Associated With Acute Pancreatitis Warrants Early Intervention</a></span>. Gastro Hep Advances. 2024 Jan;3(1):61-63. doi: 10.1016/j.gastha.2023.08.017. Epub 2023 Sep 2.</p> <p><br/><br/>Alkhouri N, et al. <span class="Hyperlink"><a href="https://doi.org/10.1016/j.gastha.2023.08.019">A Novel Prescription Digital Therapeutic Option for the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease</a></span>. Gastro Hep Advances. 2024 Jan;3(1): 9-16. doi: 10.1016/j.gastha.2023.08.019. Epub 2023 Oct 1.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

Late-Stage Incidence Rates Support CRC Screening From Age 45

Article Type
Changed
Mon, 04/29/2024 - 10:34

In the setting of conflicting national screening guidelines, the incidence of distant- and regional-stage colorectal adenocarcinoma (CRC) has been increasing in individuals aged 46-49 years, a cross-sectional study of stage-stratified CRC found.

It is well known that CRC is becoming more prevalent generally in the under 50-year population, but stage-related analyses have not been done.

Staging analysis in this age group is important, however, as an increasing burden of advance-staged disease would provide further evidence for earlier screening initiation, wrote Eric M. Montminy, MD, a gastroenterologist at John H. Stroger Hospital of County Cook, Chicago, Illinois, and colleagues in JAMA Network Open.

Montminy_Eric_IL_web.jpg
%3Cp%3EDr.%20Eric%20M.%20Montminy%3C%2Fp%3E


The United States Preventive Services Task Force (USPSTF) has recommended that average-risk screening begin at 45 years of age, as do the American Gastroenterological Association and other GI societies, although the American College of Physicians last year published clinical guidance recommending 50 years as the age to start screening for CRC for patients with average risk.

“Patients aged 46-49 may become confused on which guideline to follow, similar to confusion occurring with prior breast cancer screening changes,” Dr. Montminy said in an interview. “We wanted to demonstrate incidence rates with stage stratification to help clarify the incidence trends in this age group. Stage stratification is a key because it provides insight into the relationship between time and cancer incidence, ie, is screening finding early cancer or not?”

A 2020 study in JAMA Network Open demonstrated a 46.1% increase in CRC incidence rates (IRs) in persons aged 49-50 years. This steep increase is consistent with the presence of a large preexisting and undetected case burden.

“Our results demonstrate that adults aged 46-49 years, who are between now-conflicting guidelines on whether to start screening at age 45 or 50 years, have an increasing burden of more advanced-stage CRC and thus may be at an increased risk if screening is not initiated at age 45 years,” Dr. Montminy’s group wrote.

Using incidence data per 100,000 population from the National Cancer Institute’s Surveillance, Epidemiology, and End Results registry, the investigators observed the following IRs for early-onset CRC in the age group of 46-49 years:

  • Distant adenocarcinoma IRs increased faster than other stages: annual percentage change (APC), 2.2 (95% CI, 1.8-2.6).
  • Regional IRs also significantly increased: APC, 1.3 (95% CI, 0.8-1.7).
  • Absolute regional IRs of CRC in the age bracket of 46-49 years are similar to total pancreatic cancer IRs in all ages and all stages combined (13.2 of 100,000) over similar years. When distant IRs for CRC are included with regional IRs, those for IRs for CRC are double those for pancreatic cancer of all stages combined.
  • The only decrease was seen in localized IRs: APC, -0.6 (95% CI, -1 to -0.2).

“My best advice for clinicians is to provide the facts from the data to patients so they can make an informed health decision,” Dr. Montminy said. “This includes taking an appropriate personal and family history and having the patient factor this aspect into their decision on when and how they want to perform colon cancer screening.”

His institution adheres to the USPSTF recommendation of initiation of CRC screening at age 45 years.
 

 

 

Findings From 2000 to 2020

During 2000-2020 period, 26,887 CRCs were diagnosed in adults aged 46-49 years (54.5% in men).

As of 2020, the localized adenocarcinoma IR decreased to 7.7 of 100,000, but regional adenocarcinoma IR increased to 13.4 of 100,000 and distant adenocarcinoma IR increased to 9.0 of 100,000.

Regional adenocarcinoma IR remained the highest of all stages in 2000-2020. From 2014 to 2020, distant IRs became similar to localized IRs, except in 2017 when distant IRs were significantly higher than localized.
 

Why the CRC Uptick?

“It remains an enigma at this time as to why we’re seeing this shift,” Dr. Montminy said, noting that etiologies from the colonic microbiome to cellphones have been postulated. “To date, no theory has substantially provided causality. But whatever the source is, it is affecting Western countries in unison with data demonstrating a birth cohort effect as well,” he added. “We additionally know, based on the current epidemiologic data, that current screening practices are failing, and a unified discussion must occur in order to prevent young patients from developing advanced colon cancer.”

Meyer_Joshua_PA_web.jpg
%3Cp%3EDr.%20Joshua%20Meyer%3C%2Fp%3E

Offering his perspective on the findings, Joshua Meyer, MD, vice chair of translational research in the Department of Radiation Oncology at Fox Chase Cancer Center in Philadelphia, said the findings reinforce the practice of offering screening to average-risk individuals starting at age 45 years, the threshold at his institution. “There are previously published data demonstrating an increase in advanced stage at the time of screening initiation, and these data support that,” said Dr. Meyer, who was not involved in the present analysis.

More research needs to be done, he continued, not just on optimal age but also on the effect of multiple other factors impacting risk. “These may include family history and genetic risk as well as the role of blood- and stool-based screening assays in an integrated strategy to screen for colorectal cancer.”

There are multiple screening tests, and while colonoscopy, the gold standard, is very safe, it is not completely without risks, Dr. Meyer added. “And the question of the appropriate allocation of limited societal resources continues to be discussed on a broader level and largely explains the difference between the two guidelines.”

This study received no specific funding. Co-author Jordan J. Karlitz, MD, reported personal fees from GRAIL (senior medical director) and an equity position from Gastro Girl/GI On Demand outside f the submitted work. Dr. Meyer disclosed no conflicts of interest relevant to his comments.

Publications
Topics
Sections

In the setting of conflicting national screening guidelines, the incidence of distant- and regional-stage colorectal adenocarcinoma (CRC) has been increasing in individuals aged 46-49 years, a cross-sectional study of stage-stratified CRC found.

It is well known that CRC is becoming more prevalent generally in the under 50-year population, but stage-related analyses have not been done.

Staging analysis in this age group is important, however, as an increasing burden of advance-staged disease would provide further evidence for earlier screening initiation, wrote Eric M. Montminy, MD, a gastroenterologist at John H. Stroger Hospital of County Cook, Chicago, Illinois, and colleagues in JAMA Network Open.

Montminy_Eric_IL_web.jpg
%3Cp%3EDr.%20Eric%20M.%20Montminy%3C%2Fp%3E


The United States Preventive Services Task Force (USPSTF) has recommended that average-risk screening begin at 45 years of age, as do the American Gastroenterological Association and other GI societies, although the American College of Physicians last year published clinical guidance recommending 50 years as the age to start screening for CRC for patients with average risk.

“Patients aged 46-49 may become confused on which guideline to follow, similar to confusion occurring with prior breast cancer screening changes,” Dr. Montminy said in an interview. “We wanted to demonstrate incidence rates with stage stratification to help clarify the incidence trends in this age group. Stage stratification is a key because it provides insight into the relationship between time and cancer incidence, ie, is screening finding early cancer or not?”

A 2020 study in JAMA Network Open demonstrated a 46.1% increase in CRC incidence rates (IRs) in persons aged 49-50 years. This steep increase is consistent with the presence of a large preexisting and undetected case burden.

“Our results demonstrate that adults aged 46-49 years, who are between now-conflicting guidelines on whether to start screening at age 45 or 50 years, have an increasing burden of more advanced-stage CRC and thus may be at an increased risk if screening is not initiated at age 45 years,” Dr. Montminy’s group wrote.

Using incidence data per 100,000 population from the National Cancer Institute’s Surveillance, Epidemiology, and End Results registry, the investigators observed the following IRs for early-onset CRC in the age group of 46-49 years:

  • Distant adenocarcinoma IRs increased faster than other stages: annual percentage change (APC), 2.2 (95% CI, 1.8-2.6).
  • Regional IRs also significantly increased: APC, 1.3 (95% CI, 0.8-1.7).
  • Absolute regional IRs of CRC in the age bracket of 46-49 years are similar to total pancreatic cancer IRs in all ages and all stages combined (13.2 of 100,000) over similar years. When distant IRs for CRC are included with regional IRs, those for IRs for CRC are double those for pancreatic cancer of all stages combined.
  • The only decrease was seen in localized IRs: APC, -0.6 (95% CI, -1 to -0.2).

“My best advice for clinicians is to provide the facts from the data to patients so they can make an informed health decision,” Dr. Montminy said. “This includes taking an appropriate personal and family history and having the patient factor this aspect into their decision on when and how they want to perform colon cancer screening.”

His institution adheres to the USPSTF recommendation of initiation of CRC screening at age 45 years.
 

 

 

Findings From 2000 to 2020

During 2000-2020 period, 26,887 CRCs were diagnosed in adults aged 46-49 years (54.5% in men).

As of 2020, the localized adenocarcinoma IR decreased to 7.7 of 100,000, but regional adenocarcinoma IR increased to 13.4 of 100,000 and distant adenocarcinoma IR increased to 9.0 of 100,000.

Regional adenocarcinoma IR remained the highest of all stages in 2000-2020. From 2014 to 2020, distant IRs became similar to localized IRs, except in 2017 when distant IRs were significantly higher than localized.
 

Why the CRC Uptick?

“It remains an enigma at this time as to why we’re seeing this shift,” Dr. Montminy said, noting that etiologies from the colonic microbiome to cellphones have been postulated. “To date, no theory has substantially provided causality. But whatever the source is, it is affecting Western countries in unison with data demonstrating a birth cohort effect as well,” he added. “We additionally know, based on the current epidemiologic data, that current screening practices are failing, and a unified discussion must occur in order to prevent young patients from developing advanced colon cancer.”

Meyer_Joshua_PA_web.jpg
%3Cp%3EDr.%20Joshua%20Meyer%3C%2Fp%3E

Offering his perspective on the findings, Joshua Meyer, MD, vice chair of translational research in the Department of Radiation Oncology at Fox Chase Cancer Center in Philadelphia, said the findings reinforce the practice of offering screening to average-risk individuals starting at age 45 years, the threshold at his institution. “There are previously published data demonstrating an increase in advanced stage at the time of screening initiation, and these data support that,” said Dr. Meyer, who was not involved in the present analysis.

More research needs to be done, he continued, not just on optimal age but also on the effect of multiple other factors impacting risk. “These may include family history and genetic risk as well as the role of blood- and stool-based screening assays in an integrated strategy to screen for colorectal cancer.”

There are multiple screening tests, and while colonoscopy, the gold standard, is very safe, it is not completely without risks, Dr. Meyer added. “And the question of the appropriate allocation of limited societal resources continues to be discussed on a broader level and largely explains the difference between the two guidelines.”

This study received no specific funding. Co-author Jordan J. Karlitz, MD, reported personal fees from GRAIL (senior medical director) and an equity position from Gastro Girl/GI On Demand outside f the submitted work. Dr. Meyer disclosed no conflicts of interest relevant to his comments.

In the setting of conflicting national screening guidelines, the incidence of distant- and regional-stage colorectal adenocarcinoma (CRC) has been increasing in individuals aged 46-49 years, a cross-sectional study of stage-stratified CRC found.

It is well known that CRC is becoming more prevalent generally in the under 50-year population, but stage-related analyses have not been done.

Staging analysis in this age group is important, however, as an increasing burden of advance-staged disease would provide further evidence for earlier screening initiation, wrote Eric M. Montminy, MD, a gastroenterologist at John H. Stroger Hospital of County Cook, Chicago, Illinois, and colleagues in JAMA Network Open.

Montminy_Eric_IL_web.jpg
%3Cp%3EDr.%20Eric%20M.%20Montminy%3C%2Fp%3E


The United States Preventive Services Task Force (USPSTF) has recommended that average-risk screening begin at 45 years of age, as do the American Gastroenterological Association and other GI societies, although the American College of Physicians last year published clinical guidance recommending 50 years as the age to start screening for CRC for patients with average risk.

“Patients aged 46-49 may become confused on which guideline to follow, similar to confusion occurring with prior breast cancer screening changes,” Dr. Montminy said in an interview. “We wanted to demonstrate incidence rates with stage stratification to help clarify the incidence trends in this age group. Stage stratification is a key because it provides insight into the relationship between time and cancer incidence, ie, is screening finding early cancer or not?”

A 2020 study in JAMA Network Open demonstrated a 46.1% increase in CRC incidence rates (IRs) in persons aged 49-50 years. This steep increase is consistent with the presence of a large preexisting and undetected case burden.

“Our results demonstrate that adults aged 46-49 years, who are between now-conflicting guidelines on whether to start screening at age 45 or 50 years, have an increasing burden of more advanced-stage CRC and thus may be at an increased risk if screening is not initiated at age 45 years,” Dr. Montminy’s group wrote.

Using incidence data per 100,000 population from the National Cancer Institute’s Surveillance, Epidemiology, and End Results registry, the investigators observed the following IRs for early-onset CRC in the age group of 46-49 years:

  • Distant adenocarcinoma IRs increased faster than other stages: annual percentage change (APC), 2.2 (95% CI, 1.8-2.6).
  • Regional IRs also significantly increased: APC, 1.3 (95% CI, 0.8-1.7).
  • Absolute regional IRs of CRC in the age bracket of 46-49 years are similar to total pancreatic cancer IRs in all ages and all stages combined (13.2 of 100,000) over similar years. When distant IRs for CRC are included with regional IRs, those for IRs for CRC are double those for pancreatic cancer of all stages combined.
  • The only decrease was seen in localized IRs: APC, -0.6 (95% CI, -1 to -0.2).

“My best advice for clinicians is to provide the facts from the data to patients so they can make an informed health decision,” Dr. Montminy said. “This includes taking an appropriate personal and family history and having the patient factor this aspect into their decision on when and how they want to perform colon cancer screening.”

His institution adheres to the USPSTF recommendation of initiation of CRC screening at age 45 years.
 

 

 

Findings From 2000 to 2020

During 2000-2020 period, 26,887 CRCs were diagnosed in adults aged 46-49 years (54.5% in men).

As of 2020, the localized adenocarcinoma IR decreased to 7.7 of 100,000, but regional adenocarcinoma IR increased to 13.4 of 100,000 and distant adenocarcinoma IR increased to 9.0 of 100,000.

Regional adenocarcinoma IR remained the highest of all stages in 2000-2020. From 2014 to 2020, distant IRs became similar to localized IRs, except in 2017 when distant IRs were significantly higher than localized.
 

Why the CRC Uptick?

“It remains an enigma at this time as to why we’re seeing this shift,” Dr. Montminy said, noting that etiologies from the colonic microbiome to cellphones have been postulated. “To date, no theory has substantially provided causality. But whatever the source is, it is affecting Western countries in unison with data demonstrating a birth cohort effect as well,” he added. “We additionally know, based on the current epidemiologic data, that current screening practices are failing, and a unified discussion must occur in order to prevent young patients from developing advanced colon cancer.”

Meyer_Joshua_PA_web.jpg
%3Cp%3EDr.%20Joshua%20Meyer%3C%2Fp%3E

Offering his perspective on the findings, Joshua Meyer, MD, vice chair of translational research in the Department of Radiation Oncology at Fox Chase Cancer Center in Philadelphia, said the findings reinforce the practice of offering screening to average-risk individuals starting at age 45 years, the threshold at his institution. “There are previously published data demonstrating an increase in advanced stage at the time of screening initiation, and these data support that,” said Dr. Meyer, who was not involved in the present analysis.

More research needs to be done, he continued, not just on optimal age but also on the effect of multiple other factors impacting risk. “These may include family history and genetic risk as well as the role of blood- and stool-based screening assays in an integrated strategy to screen for colorectal cancer.”

There are multiple screening tests, and while colonoscopy, the gold standard, is very safe, it is not completely without risks, Dr. Meyer added. “And the question of the appropriate allocation of limited societal resources continues to be discussed on a broader level and largely explains the difference between the two guidelines.”

This study received no specific funding. Co-author Jordan J. Karlitz, MD, reported personal fees from GRAIL (senior medical director) and an equity position from Gastro Girl/GI On Demand outside f the submitted work. Dr. Meyer disclosed no conflicts of interest relevant to his comments.

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>167483</fileName> <TBEID>0C04F390.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F390</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>IRs up for advanced CRCs</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240426T182455</QCDate> <firstPublished>20240429T091047</firstPublished> <LastPublished>20240429T092924</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240429T091047</CMSDate> <articleSource>FROM JAMA NETWORK OPEN</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>In the setting of conflicting national screening guidelines, the incidence of distant- and regional-stage colorectal adenocarcinoma (CRC) has been increasing in</metaDescription> <articlePDF/> <teaserImage/> <teaser>Stage stratification is key because it provides insight into the relationship between time and cancer incidence, said Dr. Eric Montminy.</teaser> <title>Late-Stage Incidence Rates Support CRC Screening From Age 45</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>3</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>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> <term>21</term> <term>15</term> <term>31</term> </publications> <sections> <term canonical="true">27970</term> <term>39313</term> </sections> <topics> <term canonical="true">344</term> <term>213</term> <term>263</term> <term>67020</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Late-Stage Incidence Rates Support CRC Screening From Age 45</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">In the setting of conflicting national screening guidelines, the incidence of distant- and regional-stage colorectal adenocarcinoma (CRC) has been increasing in individuals aged 46-49 years</span>, a cross-sectional study of stage-stratified CRC found.</p> <p>It is well known that CRC is becoming more prevalent generally in the under 50-year population, but stage-related analyses have not been done.<br/><br/>Staging analysis in this age group is important, however, as an increasing burden of advance-staged disease would provide further evidence for earlier screening initiation, wrote Eric M. Montminy, MD, a gastroenterologist at John H. Stroger Hospital of County Cook, Chicago, Illinois, and colleagues <a href="https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2816222"><span class="Hyperlink">in</span><em> JAMA Network Open</em></a>.<br/><br/>The United States Preventive Services Task Force (USPSTF) has recommended that average-risk screening begin at 45 years of age, as do the American Gastroenterological Association and other GI societies, although the American College of Physicians last year published clinical guidance recommending 50 years as the age to start screening for CRC for patients with average risk.<br/><br/>“Patients aged 46-49 may become confused on which guideline to follow, similar to confusion occurring with prior breast cancer screening changes,” Dr. Montminy said in an interview. “We wanted to demonstrate incidence rates with stage stratification to help clarify the incidence trends in this age group. Stage stratification is a key because it provides insight into the relationship between time and cancer incidence, ie, is screening finding early cancer or not?”<br/><br/>A 2020 study <a href="https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2759846"><span class="Hyperlink">in </span><em>JAMA Network Open</em></a> demonstrated a 46.1% increase in CRC incidence rates (IRs) in persons aged 49-50 years. This steep increase is consistent with the presence of a large preexisting and undetected case burden.<br/><br/>“Our results demonstrate that adults aged 46-49 years, who are between now-conflicting guidelines on whether to start screening at age 45 or 50 years, have an increasing burden of more advanced-stage CRC and thus may be at an increased risk if screening is not initiated at age 45 years,” Dr. Montminy’s group wrote.<br/><br/>Using incidence data per 100,000 population from the National Cancer Institute’s Surveillance, Epidemiology, and End Results registry, the investigators observed the following IRs for early-onset CRC in the age group of 46-49 years:</p> <ul class="body"> <li>Distant adenocarcinoma IRs increased faster than other stages: annual percentage change (APC), 2.2 (95% CI, 1.8-2.6). </li> <li>Regional IRs also significantly increased: APC, 1.3 (95% CI, 0.8-1.7).</li> <li>Absolute regional IRs of CRC in the age bracket of 46-49 years are similar to total pancreatic cancer IRs in all ages and all stages combined (13.2 of 100,000) over similar years. When distant IRs for CRC are included with regional IRs, those for IRs for CRC are <span class="Hyperlink">double those for pancreatic cancer</span> of all stages combined.</li> <li>The only decrease was seen in localized IRs: APC, -0.6 (95% CI, -1 to -0.2).</li> </ul> <p>“My best advice for clinicians is to provide the facts from the data to patients so they can make an informed health decision,” Dr. Montminy said. “This includes taking an appropriate personal and family history and having the patient factor this aspect into their decision on when and how they want to perform colon cancer screening.”<br/><br/>His institution adheres to the USPSTF recommendation of initiation of CRC screening at age 45 years.<br/><br/></p> <h2>Findings From 2000 to 2020</h2> <p>During 2000-2020 period, 26,887 CRCs were diagnosed in adults aged 46-49 years (54.5% in men).</p> <p>As of 2020, the localized adenocarcinoma IR decreased to 7.7 of 100,000, but regional adenocarcinoma IR increased to 13.4 of 100,000 and distant adenocarcinoma IR increased to 9.0 of 100,000.<br/><br/>Regional adenocarcinoma IR remained the highest of all stages in 2000-2020. From 2014 to 2020, distant IRs became similar to localized IRs, except in 2017 when distant IRs were significantly higher than localized.<br/><br/></p> <h2>Why the CRC Uptick?</h2> <p>“It remains an enigma at this time as to why we’re seeing this shift,” Dr. Montminy said, noting that etiologies from the colonic microbiome to cellphones have been postulated. “To date, no theory has substantially provided causality. But whatever the source is, it is affecting Western countries in unison with data demonstrating a birth cohort effect as well,” he added. “We additionally know, based on the current epidemiologic data, that current screening practices are failing, and a unified discussion must occur in order to prevent young patients from developing advanced colon cancer.”</p> <p>Offering his perspective on the findings, Joshua Meyer, MD, vice chair of translational research in the Department of Radiation Oncology at Fox Chase Cancer Center in Philadelphia, said the findings reinforce the practice of offering screening to average-risk individuals starting at age 45 years, the threshold at his institution. “There are previously published data demonstrating an increase in advanced stage at the time of screening initiation, and these data support that,” said Dr. Meyer, who was not involved in the present analysis.<br/><br/>More research needs to be done, he continued, not just on optimal age but also on the effect of multiple other factors impacting risk. “These may include family history and genetic risk as well as the role of blood- and stool-based screening assays in an integrated strategy to screen for colorectal cancer.”<br/><br/>There are multiple screening tests, and while colonoscopy, the gold standard, is very safe, it is not completely without risks, Dr. Meyer added. “And the question of the appropriate allocation of limited societal resources continues to be discussed on a broader level and largely explains the difference between the two guidelines.”<br/><br/>This study received no specific funding. Co-author Jordan J. Karlitz, MD, reported personal fees from GRAIL (senior medical director) and an equity position from Gastro Girl/GI On Demand outside f the submitted work. Dr. Meyer disclosed no conflicts of interest relevant to his comments.<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 JAMA NETWORK OPEN

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

Liquid Biopsy Has Near-Perfect Accuracy for Early Pancreatic Cancer

Article Type
Changed
Mon, 04/15/2024 - 17:34

— A liquid biopsy assay that combines a microRNA signature and a well-known biomarker for pancreatic cancer has demonstrated an accuracy of 97% for detecting stage I/II pancreatic ductal adenocarcinoma, the most common type of pancreatic cancer.

It is quite encouraging to know we have a blood test that could potentially find this disease early, said Ajay Goel, PhD, a molecular diagnostics specialist at City of Hope in Duarte, California, who presented the findings at the annual meeting of the American Association for Cancer Research (AACR).

Dr. Goel and colleagues developed a signature for pancreatic cancer based on microRNAs identified in the exomes shed from pancreatic cancers and cell-free DNA markers found in the blood of patients with the disease.

Their initial assay tested blood samples for this signature in a training cohort of 252 people in Japan, approximately 60% of whom had pancreatic cancer. The rest were healthy controls. The assay was then tested in validation cohorts of 400 subjects, half with pancreatic cancer and half controls, in China and South Korea.

In both the initial and validation tests, the microRNA assay had an accuracy of about 90% for stage I/II pancreatic cancer, already far better than commercially available assays.

In an additional validation cohort in the United States with 139 patients with pancreatic cancer and 193 controls at six centers across the country, the researchers found that adding carbohydrate antigen 19-9 — a well-known marker of pancreatic cancer — to the assay boosted the test’s accuracy to 97%.

The test performed the same whether the tumor was in the head or tail of the pancreas.

“We are very excited about this data,” said Dr. Goel.

The technology was recently licensed to Pharus Diagnostics for commercial development, which will likely include a prospective screening trial, he told this news organization.

Because pancreatic cancer is fairly uncommon, Dr. Goel did not anticipate the test being used for general screening but rather for screening high-risk patients such as those with newly diagnosed type 2 diabetes, a family history of pancreatic cancer, or predisposing genetic mutations.

“It should be a very inexpensive test; it doesn’t cost us much to do in the lab,” he added.

Study moderator Ryan Corcoran, MD, PhD, a gastrointestinal (GI) oncologist at Massachusetts General Hospital, Boston, saw the potential.

“As a GI oncologist, I know how lethal and hard to treat pancreatic cancer is,” he said. A test that could reliably detect pancreatic cancer early, with an acceptable false-positive rate, would be extremely useful.

“The cure rate is many, many times higher,” if we detect it before it has a chance to spread, he explained.

In the meantime, Dr. Goel said there’s more work to be done.

Almost 4,000 subjects have been enrolled in ongoing validation efforts, and efforts are underway to use the test to screen thousands of banked blood samples from the PLCO, a prospective cancer screening trial in healthy subjects.

The researchers also want to see if the test can distinguish benign pancreatic cysts from ones that turn cancerous.

The idea is to find the earliest possible signs of this disease to see if we can find it not “at the moment of clinical diagnosis, but possibly 6 months, 1 year, 2 years earlier” than with radiologic imaging, Dr. Goel said.

The work was funded by the National Cancer Institute and others. Dr. Goel is a consultant for Pharus Diagnostics and Cellomics. Dr. Corcoran is a consultant for, has grants from, and/or holds stock in numerous companies, including Pfizer, Novartis, Eli Lilly, and Revolution Medicines.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

— A liquid biopsy assay that combines a microRNA signature and a well-known biomarker for pancreatic cancer has demonstrated an accuracy of 97% for detecting stage I/II pancreatic ductal adenocarcinoma, the most common type of pancreatic cancer.

It is quite encouraging to know we have a blood test that could potentially find this disease early, said Ajay Goel, PhD, a molecular diagnostics specialist at City of Hope in Duarte, California, who presented the findings at the annual meeting of the American Association for Cancer Research (AACR).

Dr. Goel and colleagues developed a signature for pancreatic cancer based on microRNAs identified in the exomes shed from pancreatic cancers and cell-free DNA markers found in the blood of patients with the disease.

Their initial assay tested blood samples for this signature in a training cohort of 252 people in Japan, approximately 60% of whom had pancreatic cancer. The rest were healthy controls. The assay was then tested in validation cohorts of 400 subjects, half with pancreatic cancer and half controls, in China and South Korea.

In both the initial and validation tests, the microRNA assay had an accuracy of about 90% for stage I/II pancreatic cancer, already far better than commercially available assays.

In an additional validation cohort in the United States with 139 patients with pancreatic cancer and 193 controls at six centers across the country, the researchers found that adding carbohydrate antigen 19-9 — a well-known marker of pancreatic cancer — to the assay boosted the test’s accuracy to 97%.

The test performed the same whether the tumor was in the head or tail of the pancreas.

“We are very excited about this data,” said Dr. Goel.

The technology was recently licensed to Pharus Diagnostics for commercial development, which will likely include a prospective screening trial, he told this news organization.

Because pancreatic cancer is fairly uncommon, Dr. Goel did not anticipate the test being used for general screening but rather for screening high-risk patients such as those with newly diagnosed type 2 diabetes, a family history of pancreatic cancer, or predisposing genetic mutations.

“It should be a very inexpensive test; it doesn’t cost us much to do in the lab,” he added.

Study moderator Ryan Corcoran, MD, PhD, a gastrointestinal (GI) oncologist at Massachusetts General Hospital, Boston, saw the potential.

“As a GI oncologist, I know how lethal and hard to treat pancreatic cancer is,” he said. A test that could reliably detect pancreatic cancer early, with an acceptable false-positive rate, would be extremely useful.

“The cure rate is many, many times higher,” if we detect it before it has a chance to spread, he explained.

In the meantime, Dr. Goel said there’s more work to be done.

Almost 4,000 subjects have been enrolled in ongoing validation efforts, and efforts are underway to use the test to screen thousands of banked blood samples from the PLCO, a prospective cancer screening trial in healthy subjects.

The researchers also want to see if the test can distinguish benign pancreatic cysts from ones that turn cancerous.

The idea is to find the earliest possible signs of this disease to see if we can find it not “at the moment of clinical diagnosis, but possibly 6 months, 1 year, 2 years earlier” than with radiologic imaging, Dr. Goel said.

The work was funded by the National Cancer Institute and others. Dr. Goel is a consultant for Pharus Diagnostics and Cellomics. Dr. Corcoran is a consultant for, has grants from, and/or holds stock in numerous companies, including Pfizer, Novartis, Eli Lilly, and Revolution Medicines.

A version of this article appeared on Medscape.com.

— A liquid biopsy assay that combines a microRNA signature and a well-known biomarker for pancreatic cancer has demonstrated an accuracy of 97% for detecting stage I/II pancreatic ductal adenocarcinoma, the most common type of pancreatic cancer.

It is quite encouraging to know we have a blood test that could potentially find this disease early, said Ajay Goel, PhD, a molecular diagnostics specialist at City of Hope in Duarte, California, who presented the findings at the annual meeting of the American Association for Cancer Research (AACR).

Dr. Goel and colleagues developed a signature for pancreatic cancer based on microRNAs identified in the exomes shed from pancreatic cancers and cell-free DNA markers found in the blood of patients with the disease.

Their initial assay tested blood samples for this signature in a training cohort of 252 people in Japan, approximately 60% of whom had pancreatic cancer. The rest were healthy controls. The assay was then tested in validation cohorts of 400 subjects, half with pancreatic cancer and half controls, in China and South Korea.

In both the initial and validation tests, the microRNA assay had an accuracy of about 90% for stage I/II pancreatic cancer, already far better than commercially available assays.

In an additional validation cohort in the United States with 139 patients with pancreatic cancer and 193 controls at six centers across the country, the researchers found that adding carbohydrate antigen 19-9 — a well-known marker of pancreatic cancer — to the assay boosted the test’s accuracy to 97%.

The test performed the same whether the tumor was in the head or tail of the pancreas.

“We are very excited about this data,” said Dr. Goel.

The technology was recently licensed to Pharus Diagnostics for commercial development, which will likely include a prospective screening trial, he told this news organization.

Because pancreatic cancer is fairly uncommon, Dr. Goel did not anticipate the test being used for general screening but rather for screening high-risk patients such as those with newly diagnosed type 2 diabetes, a family history of pancreatic cancer, or predisposing genetic mutations.

“It should be a very inexpensive test; it doesn’t cost us much to do in the lab,” he added.

Study moderator Ryan Corcoran, MD, PhD, a gastrointestinal (GI) oncologist at Massachusetts General Hospital, Boston, saw the potential.

“As a GI oncologist, I know how lethal and hard to treat pancreatic cancer is,” he said. A test that could reliably detect pancreatic cancer early, with an acceptable false-positive rate, would be extremely useful.

“The cure rate is many, many times higher,” if we detect it before it has a chance to spread, he explained.

In the meantime, Dr. Goel said there’s more work to be done.

Almost 4,000 subjects have been enrolled in ongoing validation efforts, and efforts are underway to use the test to screen thousands of banked blood samples from the PLCO, a prospective cancer screening trial in healthy subjects.

The researchers also want to see if the test can distinguish benign pancreatic cysts from ones that turn cancerous.

The idea is to find the earliest possible signs of this disease to see if we can find it not “at the moment of clinical diagnosis, but possibly 6 months, 1 year, 2 years earlier” than with radiologic imaging, Dr. Goel said.

The work was funded by the National Cancer Institute and others. Dr. Goel is a consultant for Pharus Diagnostics and Cellomics. Dr. Corcoran is a consultant for, has grants from, and/or holds stock in numerous companies, including Pfizer, Novartis, Eli Lilly, and Revolution Medicines.

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>167647</fileName> <TBEID>0C04F827.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F827</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240411T132645</QCDate> <firstPublished>20240411T134005</firstPublished> <LastPublished>20240411T134005</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240411T134005</CMSDate> <articleSource>FROM AACR 2024</articleSource> <facebookInfo/> <meetingNumber/> <byline>M. Alexander Otto, PA</byline> <bylineText>M. ALEXANDER OTTO, PA, MMS</bylineText> <bylineFull>M. ALEXANDER OTTO, PA, MMS</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>SAN DIEGO — A liquid biopsy assay that combines a microRNA signature and a well-known biomarker for pancreatic cancer has demonstrated an accuracy of 97% for de</metaDescription> <articlePDF/> <teaserImage/> <teaser>Researchers develop signature for pancreatic cancer based on microRNAs and cell-free DNA markers in the blood of patients with the disease.</teaser> <title>Liquid Biopsy Has Near-Perfect Accuracy for Early Pancreatic Cancer</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>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>endo</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>GIHOLD</publicationCode> <pubIssueName>January 2014</pubIssueName> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> </publications_g> <publications> <term canonical="true">31</term> <term>34</term> <term>15</term> <term>21</term> </publications> <sections> <term canonical="true">53</term> <term>39313</term> </sections> <topics> <term>270</term> <term>280</term> <term canonical="true">67020</term> <term>213</term> <term>205</term> <term>210</term> <term>277</term> <term>263</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Liquid Biopsy Has Near-Perfect Accuracy for Early Pancreatic Cancer</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription"><span class="dateline">SAN DIEGO</span> — A liquid biopsy assay that combines a microRNA signature and a well-known biomarker for <span class="Hyperlink"><a href="https://emedicine.medscape.com/article/280605-overview">pancreatic cancer</a></span> has demonstrated an accuracy of 97% for detecting stage I/II pancreatic ductal adenocarcinoma,</span> the most common type of pancreatic cancer.</p> <p>It is quite encouraging to know we have a <span class="Hyperlink">blood test</span> that could potentially find this disease early, said <span class="Hyperlink"><a href="https://www.cityofhope.org/ajay-goel">Ajay Goel, PhD</a></span>, a molecular diagnostics specialist at City of Hope in Duarte, California, who presented the findings at the annual meeting of the <span class="Hyperlink"><a href="https://www.medscape.com/viewcollection/37452">American Association for Cancer Research (AACR)</a></span>.<br/><br/>Dr. Goel and colleagues developed a signature for pancreatic cancer based on microRNAs identified in the exomes shed from pancreatic cancers and cell-free DNA markers found in the blood of patients with the disease.<br/><br/>Their initial assay tested blood samples for this signature in a training cohort of 252 people in Japan, approximately 60% of whom had pancreatic cancer. The rest were healthy controls. The assay was then tested in validation cohorts of 400 subjects, half with pancreatic cancer and half controls, in China and South Korea.<br/><br/>In both the initial and validation tests, the microRNA assay had an accuracy of about 90% for stage I/II pancreatic cancer, already far better than commercially available assays.<br/><br/>In an additional validation cohort in the United States with 139 patients with pancreatic cancer and 193 controls at six centers across the country, the researchers found that adding carbohydrate antigen 19-9 — a well-known marker of pancreatic cancer — to the assay boosted the test’s accuracy to 97%.<br/><br/>The test performed the same whether the tumor was in the head or tail of the pancreas.<br/><br/>“We are very excited about this data,” said Dr. Goel.<br/><br/>The technology was recently licensed to <span class="Hyperlink"><a href="https://www.prnewswire.com/news-releases/pharus-diagnostics-signs-worldwide-exclusive-license-agreement-with-city-of-hope-for-novel-biomarkers-to-be-used-in-liquid-biopsy-screening-for-early-pancreatic-cancer-diagnosis-302062754.html">Pharus Diagnostics</a></span> for commercial development, which will likely include a prospective screening trial, he told this news organization.<br/><br/>Because pancreatic cancer is fairly uncommon, Dr. Goel did not anticipate the test being used for general screening but rather for screening high-risk patients such as those with newly diagnosed <span class="Hyperlink"><a href="https://emedicine.medscape.com/article/117853-overview">type 2 diabetes</a></span>, a family history of pancreatic cancer, or predisposing genetic mutations.<br/><br/>“It should be a very inexpensive test; it doesn’t cost us much to do in the lab,” he added.<br/><br/>Study moderator <span class="Hyperlink"><a href="https://www.massgeneral.org/doctors/23096/ryan-corcoran">Ryan Corcoran</a></span>, MD, PhD, a gastrointestinal (GI) oncologist at Massachusetts General Hospital, Boston, saw the potential.<br/><br/>“As a GI oncologist, I know how lethal and hard to treat pancreatic cancer is,” he said. A test that could reliably detect pancreatic cancer early, with an acceptable false-positive rate, would be extremely useful.<br/><br/>“The cure rate is many, many times higher,” if we detect it before it has a chance to spread, he explained.<br/><br/>In the meantime, Dr. Goel said there’s more work to be done.<br/><br/>Almost 4,000 subjects have been enrolled in ongoing validation efforts, and efforts are underway to use the test to screen thousands of banked blood samples from the <span class="Hyperlink"><a href="https://prevention.cancer.gov/major-programs/prostate-lung-colorectal-and-ovarian-cancer-screening-trial-plco">PLCO</a></span>, a prospective cancer screening trial in healthy subjects.<br/><br/>The researchers also want to see if the test can distinguish benign pancreatic cysts from ones that turn cancerous.<br/><br/>The idea is to find the earliest possible signs of this disease to see if we can find it not “at the moment of clinical diagnosis, but possibly 6 months, 1 year, 2 years earlier” than with radiologic imaging, Dr. Goel said.<br/><br/>The work was funded by the National Cancer Institute and others. Dr. Goel is a consultant for Pharus Diagnostics and Cellomics. Dr. Corcoran is a consultant for, has grants from, and/or holds stock in numerous companies, including Pfizer, Novartis, Eli Lilly, and Revolution Medicines.<span class="end"/></p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/liquid-biopsy-has-near-perfect-accuracy-early-pancreatic-2024a10006ut">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 AACR 2024

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

Power-Washing Moves Beyond Home Improvement, Into Gastroenterology

A Promising Approach, but Challenges Remain
Article Type
Changed
Wed, 04/10/2024 - 09:35

Power-washing is no longer just for blasting grimy driveways and stripping flaky paint. It’s good for work inside the gut, too.

In a proof-of-concept study, a “novel systematically directed high-pressure liquid spray,” delivered via the ERBEJET flexible probe, showed promise for collecting cytology specimens from the stomachs of patients undergoing endoscopy for gastric cancer screening or surveillance, reported lead author Charles J. Lightdale, MD, of Columbia University Irving Medical Center, New York City, and colleagues.

“Systematic random biopsies (updated Sydney protocol) have been recommended to increase detection of gastric intestinal metaplasia (GIM) and dysplasia,” the investigators wrote in Techniques and Innovations in Gastrointestinal Endoscopy. “However, random biopsies can be laborious, time consuming, costly, and susceptible to sampling error owing to the large surface area of the stomach.”

Power-washing, in contrast, with the pressure dial turned to 10 bar, involves spraying the gut in a systematic fashion “using sweeping and painting motions” to dislodge cells from the mucosa. These specimens are then suctioned from the resultant pools of liquid, mixed 1:1 with 10% formalin, and shipped to the lab.
 

Boom! Cytology!

Just to be sure, however, the nine patients involved in the study also underwent standard-of-care biopsy collection from areas of interest, followed by random sampling according to the updated Sydney protocol. Two of the patients were power-washed again 12 months later for endoscopic surveillance.

Power-washing added 7-10 minutes to standard endoscopy time and generated 60-100 mL of liquid for collection. Post suction, a closer look at the gastric mucosa revealed “scattered superficial erosions,” while blood loss was deemed “minimal.” The procedure appeared well tolerated, with no aspiration or esophageal reflux during endoscopy, or adverse events reported by patients after 1 week of follow-up.

Cytopathology samples were deemed satisfactory and yielded “multiple strips and large clusters of cells.” These were sufficient to diagnose GIM in three patients and reactive glandular changes with inflammation in one patient, with findings confirmed on biopsy. In contrast, the power-washed cells from one patient were “highly suspicious” for dysplasia, but biopsies were negative.

Although the study was too small for a reliable comparison with the Sydney protocol, Dr. Lightdale and colleagues concluded that the power-wash approach deserves further investigation.

Use of power-wash to obtain cytology has the potential to improve endoscopic screening and surveillance protocols for detecting GIM and dysplasia and to reduce morbidity and mortality from gastric cancer,” they wrote.

The investigators predicted that power-washing is likely safe in most patients, although it may be unsuitable for those with noncorrectable coagulopathies or in patients who cannot stop anticoagulants. Postsurgical patients, on the other hand, should tolerate the procedure just fine.

Patients with risk of gastric cancer “might be an important group” for evaluating the power-wash procedure, the investigators wrote, noting that combining the approach with artificial intelligence could one day yield even better results.

In the meantime, Dr. Lightdale and colleagues — like so many weekend warriors wielding a power-washer — are going to see if a different nozzle will take their work to the next level.

“We are actively studying a catheter with a broader stream and the potential to increase efficiency and decrease procedure time,” they wrote. “Another catheter design might allow for simultaneous spray and suction, so that cytology samples from specific regions of the stomach could be separately analyzed.”

This study was funded by Dalio Philanthropies, the Price Family Foundation, and the Frederic and Patricia Salerno Foundation. The investigators disclosed relationships with Boston Scientific, Interscope, Medtronic, and others.

Body

 

The optimal surveillance endoscopic modality for gastric intestinal metaplasia (GIM) is yet to be determined. Although the updated Sydney System, a comprehensive endoscopic biopsy protocol, has been advocated for GIM mapping, challenges are the heterogeneous distribution of GIM, suboptimal diagnostic accuracy of endoscopy to detect GIM, and the cost burden of multiple biopsies.

This study by Lightdale et al. demonstrated the technical feasibility and safety of obtaining cytology for the detection of gastric intestinal metaplasia by using a systemic endoscopy-guided high-pressure spray “power-wash” method. In this study, all cytophathology samples in nine subjects were deemed satisfactory for evaluation. All three subjects who were cytology positive for GIM on H&E stain and confirmed with positive immunohistochemistry (IHC) showed GIM on biopsy, and one subject had cells highly suspicious for dysplasia on cytology but biopsy was negative. Although all patients showed multiple superficial erosions after power-wash, bleeding was minimal and no adverse events related to power-wash were observed.

Applying cytology for detection of GIM appears promising as the way of collecting samples from the large surface area of the stomach. As clinicians, however, we are still left with some challenges. Even if cells collected are suspicious for dysplasia/neoplasia by this power-wash method, it would not be useful unless we precisely localize the area as we can not provide a focal curative endoscopic treatment. It is critical to increase the yield of localization of cytology sampling. Further research is also needed to standardize the cytopathologic diagnostic criteria of GIM and cost-effectiveness of the cytology-based approach compared to the current gold-standard biopsy protocol for the diagnosis of GIM.

Yutaka Tomizawa, MD, MSc, is a therapeutic endoscopist and clinical associate professor of medicine, Division of Gastroenterology, University of Washington, Seattle. He has no conflicts related to this report.

Publications
Topics
Sections
Body

 

The optimal surveillance endoscopic modality for gastric intestinal metaplasia (GIM) is yet to be determined. Although the updated Sydney System, a comprehensive endoscopic biopsy protocol, has been advocated for GIM mapping, challenges are the heterogeneous distribution of GIM, suboptimal diagnostic accuracy of endoscopy to detect GIM, and the cost burden of multiple biopsies.

This study by Lightdale et al. demonstrated the technical feasibility and safety of obtaining cytology for the detection of gastric intestinal metaplasia by using a systemic endoscopy-guided high-pressure spray “power-wash” method. In this study, all cytophathology samples in nine subjects were deemed satisfactory for evaluation. All three subjects who were cytology positive for GIM on H&E stain and confirmed with positive immunohistochemistry (IHC) showed GIM on biopsy, and one subject had cells highly suspicious for dysplasia on cytology but biopsy was negative. Although all patients showed multiple superficial erosions after power-wash, bleeding was minimal and no adverse events related to power-wash were observed.

Applying cytology for detection of GIM appears promising as the way of collecting samples from the large surface area of the stomach. As clinicians, however, we are still left with some challenges. Even if cells collected are suspicious for dysplasia/neoplasia by this power-wash method, it would not be useful unless we precisely localize the area as we can not provide a focal curative endoscopic treatment. It is critical to increase the yield of localization of cytology sampling. Further research is also needed to standardize the cytopathologic diagnostic criteria of GIM and cost-effectiveness of the cytology-based approach compared to the current gold-standard biopsy protocol for the diagnosis of GIM.

Yutaka Tomizawa, MD, MSc, is a therapeutic endoscopist and clinical associate professor of medicine, Division of Gastroenterology, University of Washington, Seattle. He has no conflicts related to this report.

Body

 

The optimal surveillance endoscopic modality for gastric intestinal metaplasia (GIM) is yet to be determined. Although the updated Sydney System, a comprehensive endoscopic biopsy protocol, has been advocated for GIM mapping, challenges are the heterogeneous distribution of GIM, suboptimal diagnostic accuracy of endoscopy to detect GIM, and the cost burden of multiple biopsies.

This study by Lightdale et al. demonstrated the technical feasibility and safety of obtaining cytology for the detection of gastric intestinal metaplasia by using a systemic endoscopy-guided high-pressure spray “power-wash” method. In this study, all cytophathology samples in nine subjects were deemed satisfactory for evaluation. All three subjects who were cytology positive for GIM on H&E stain and confirmed with positive immunohistochemistry (IHC) showed GIM on biopsy, and one subject had cells highly suspicious for dysplasia on cytology but biopsy was negative. Although all patients showed multiple superficial erosions after power-wash, bleeding was minimal and no adverse events related to power-wash were observed.

Applying cytology for detection of GIM appears promising as the way of collecting samples from the large surface area of the stomach. As clinicians, however, we are still left with some challenges. Even if cells collected are suspicious for dysplasia/neoplasia by this power-wash method, it would not be useful unless we precisely localize the area as we can not provide a focal curative endoscopic treatment. It is critical to increase the yield of localization of cytology sampling. Further research is also needed to standardize the cytopathologic diagnostic criteria of GIM and cost-effectiveness of the cytology-based approach compared to the current gold-standard biopsy protocol for the diagnosis of GIM.

Yutaka Tomizawa, MD, MSc, is a therapeutic endoscopist and clinical associate professor of medicine, Division of Gastroenterology, University of Washington, Seattle. He has no conflicts related to this report.

Title
A Promising Approach, but Challenges Remain
A Promising Approach, but Challenges Remain

Power-washing is no longer just for blasting grimy driveways and stripping flaky paint. It’s good for work inside the gut, too.

In a proof-of-concept study, a “novel systematically directed high-pressure liquid spray,” delivered via the ERBEJET flexible probe, showed promise for collecting cytology specimens from the stomachs of patients undergoing endoscopy for gastric cancer screening or surveillance, reported lead author Charles J. Lightdale, MD, of Columbia University Irving Medical Center, New York City, and colleagues.

“Systematic random biopsies (updated Sydney protocol) have been recommended to increase detection of gastric intestinal metaplasia (GIM) and dysplasia,” the investigators wrote in Techniques and Innovations in Gastrointestinal Endoscopy. “However, random biopsies can be laborious, time consuming, costly, and susceptible to sampling error owing to the large surface area of the stomach.”

Power-washing, in contrast, with the pressure dial turned to 10 bar, involves spraying the gut in a systematic fashion “using sweeping and painting motions” to dislodge cells from the mucosa. These specimens are then suctioned from the resultant pools of liquid, mixed 1:1 with 10% formalin, and shipped to the lab.
 

Boom! Cytology!

Just to be sure, however, the nine patients involved in the study also underwent standard-of-care biopsy collection from areas of interest, followed by random sampling according to the updated Sydney protocol. Two of the patients were power-washed again 12 months later for endoscopic surveillance.

Power-washing added 7-10 minutes to standard endoscopy time and generated 60-100 mL of liquid for collection. Post suction, a closer look at the gastric mucosa revealed “scattered superficial erosions,” while blood loss was deemed “minimal.” The procedure appeared well tolerated, with no aspiration or esophageal reflux during endoscopy, or adverse events reported by patients after 1 week of follow-up.

Cytopathology samples were deemed satisfactory and yielded “multiple strips and large clusters of cells.” These were sufficient to diagnose GIM in three patients and reactive glandular changes with inflammation in one patient, with findings confirmed on biopsy. In contrast, the power-washed cells from one patient were “highly suspicious” for dysplasia, but biopsies were negative.

Although the study was too small for a reliable comparison with the Sydney protocol, Dr. Lightdale and colleagues concluded that the power-wash approach deserves further investigation.

Use of power-wash to obtain cytology has the potential to improve endoscopic screening and surveillance protocols for detecting GIM and dysplasia and to reduce morbidity and mortality from gastric cancer,” they wrote.

The investigators predicted that power-washing is likely safe in most patients, although it may be unsuitable for those with noncorrectable coagulopathies or in patients who cannot stop anticoagulants. Postsurgical patients, on the other hand, should tolerate the procedure just fine.

Patients with risk of gastric cancer “might be an important group” for evaluating the power-wash procedure, the investigators wrote, noting that combining the approach with artificial intelligence could one day yield even better results.

In the meantime, Dr. Lightdale and colleagues — like so many weekend warriors wielding a power-washer — are going to see if a different nozzle will take their work to the next level.

“We are actively studying a catheter with a broader stream and the potential to increase efficiency and decrease procedure time,” they wrote. “Another catheter design might allow for simultaneous spray and suction, so that cytology samples from specific regions of the stomach could be separately analyzed.”

This study was funded by Dalio Philanthropies, the Price Family Foundation, and the Frederic and Patricia Salerno Foundation. The investigators disclosed relationships with Boston Scientific, Interscope, Medtronic, and others.

Power-washing is no longer just for blasting grimy driveways and stripping flaky paint. It’s good for work inside the gut, too.

In a proof-of-concept study, a “novel systematically directed high-pressure liquid spray,” delivered via the ERBEJET flexible probe, showed promise for collecting cytology specimens from the stomachs of patients undergoing endoscopy for gastric cancer screening or surveillance, reported lead author Charles J. Lightdale, MD, of Columbia University Irving Medical Center, New York City, and colleagues.

“Systematic random biopsies (updated Sydney protocol) have been recommended to increase detection of gastric intestinal metaplasia (GIM) and dysplasia,” the investigators wrote in Techniques and Innovations in Gastrointestinal Endoscopy. “However, random biopsies can be laborious, time consuming, costly, and susceptible to sampling error owing to the large surface area of the stomach.”

Power-washing, in contrast, with the pressure dial turned to 10 bar, involves spraying the gut in a systematic fashion “using sweeping and painting motions” to dislodge cells from the mucosa. These specimens are then suctioned from the resultant pools of liquid, mixed 1:1 with 10% formalin, and shipped to the lab.
 

Boom! Cytology!

Just to be sure, however, the nine patients involved in the study also underwent standard-of-care biopsy collection from areas of interest, followed by random sampling according to the updated Sydney protocol. Two of the patients were power-washed again 12 months later for endoscopic surveillance.

Power-washing added 7-10 minutes to standard endoscopy time and generated 60-100 mL of liquid for collection. Post suction, a closer look at the gastric mucosa revealed “scattered superficial erosions,” while blood loss was deemed “minimal.” The procedure appeared well tolerated, with no aspiration or esophageal reflux during endoscopy, or adverse events reported by patients after 1 week of follow-up.

Cytopathology samples were deemed satisfactory and yielded “multiple strips and large clusters of cells.” These were sufficient to diagnose GIM in three patients and reactive glandular changes with inflammation in one patient, with findings confirmed on biopsy. In contrast, the power-washed cells from one patient were “highly suspicious” for dysplasia, but biopsies were negative.

Although the study was too small for a reliable comparison with the Sydney protocol, Dr. Lightdale and colleagues concluded that the power-wash approach deserves further investigation.

Use of power-wash to obtain cytology has the potential to improve endoscopic screening and surveillance protocols for detecting GIM and dysplasia and to reduce morbidity and mortality from gastric cancer,” they wrote.

The investigators predicted that power-washing is likely safe in most patients, although it may be unsuitable for those with noncorrectable coagulopathies or in patients who cannot stop anticoagulants. Postsurgical patients, on the other hand, should tolerate the procedure just fine.

Patients with risk of gastric cancer “might be an important group” for evaluating the power-wash procedure, the investigators wrote, noting that combining the approach with artificial intelligence could one day yield even better results.

In the meantime, Dr. Lightdale and colleagues — like so many weekend warriors wielding a power-washer — are going to see if a different nozzle will take their work to the next level.

“We are actively studying a catheter with a broader stream and the potential to increase efficiency and decrease procedure time,” they wrote. “Another catheter design might allow for simultaneous spray and suction, so that cytology samples from specific regions of the stomach could be separately analyzed.”

This study was funded by Dalio Philanthropies, the Price Family Foundation, and the Frederic and Patricia Salerno Foundation. The investigators disclosed relationships with Boston Scientific, Interscope, Medtronic, 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>167500</fileName> <TBEID>0C04F4F8.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F4F8</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>TIGE_Lightdale_powerwash</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240409T173235</QCDate> <firstPublished>20240410T090547</firstPublished> <LastPublished>20240410T093150</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240410T090547</CMSDate> <articleSource>FROM TECHNIQUES AND INNOVATIONS IN GASTROINTESTINAL ENDOSCOPY</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>Use of power-wash to obtain cytology has the potential to improve endoscopic screening and surveillance protocols for detecting GIM and dysplasia</metaDescription> <articlePDF/> <teaserImage/> <teaser>Patients with risk of gastric cancer “might be an important group” for evaluating the power-wash procedure, the investigators wrote.</teaser> <title>Power-Washing Moves Beyond Home Improvement, Into Gastroenterology</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</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>39702</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Power-Washing Moves Beyond Home Improvement, Into Gastroenterology</title> <deck/> </itemMeta> <itemContent> <p>Power-washing is no longer just for blasting grimy driveways and stripping flaky paint. It’s good for work inside the gut, too.</p> <p>In a proof-of-concept study, a “novel systematically directed high-pressure liquid spray,” delivered via the ERBEJET flexible probe, showed promise for collecting cytology specimens from the stomachs of patients undergoing endoscopy for gastric cancer screening or surveillance, reported lead author <a href="https://doctors.columbia.edu/us/ny/new-york/charles-j-lightdale-md-180-fort-washington-avenue">Charles J. Lightdale, MD</a>, of Columbia University Irving Medical Center, New York City, and colleagues.<br/><br/>“Systematic random biopsies (updated Sydney protocol) have been recommended to increase detection of gastric intestinal metaplasia (GIM) and dysplasia,” the investigators wrote in <em><a href="https://www.tigejournal.org/article/S2590-0307(24)00001-1/fulltext">Techniques and Innovations in Gastrointestinal Endoscopy</a></em>. “However, random biopsies can be laborious, time consuming, costly, and susceptible to sampling error owing to the large surface area of the stomach.”<br/><br/>Power-washing, in contrast, with the pressure dial turned to 10 bar, involves spraying the gut in a systematic fashion “using sweeping and painting motions” to dislodge cells from the mucosa. These specimens are then suctioned from the resultant pools of liquid, mixed 1:1 with 10% formalin, and shipped to the lab.<br/><br/></p> <h2>Boom! Cytology!</h2> <p>Just to be sure, however, the nine patients involved in the study also underwent standard-of-care biopsy collection from areas of interest, followed by random sampling according to the updated Sydney protocol. Two of the patients were power-washed again 12 months later for endoscopic surveillance.</p> <p>Power-washing added 7-10 minutes to standard endoscopy time and generated 60-100 mL of liquid for collection. Post suction, a closer look at the gastric mucosa revealed “scattered superficial erosions,” while blood loss was deemed “minimal.” The procedure appeared well tolerated, with no aspiration or esophageal reflux during endoscopy, or adverse events reported by patients after 1 week of follow-up.<br/><br/>Cytopathology samples were deemed satisfactory and yielded “multiple strips and large clusters of cells.” These were sufficient to diagnose GIM in three patients and reactive glandular changes with inflammation in one patient, with findings confirmed on biopsy. In contrast, the power-washed cells from one patient were “highly suspicious” for dysplasia, but biopsies were negative.<br/><br/>Although the study was too small for a reliable comparison with the Sydney protocol, Dr. Lightdale and colleagues concluded that the power-wash approach deserves further investigation.<br/><br/>“<span class="tag metaDescription">Use of power-wash to obtain cytology has the potential to improve endoscopic screening and surveillance protocols for detecting GIM and dysplasia</span> and to reduce morbidity and mortality from gastric cancer,” they wrote.<br/><br/>The investigators predicted that power-washing is likely safe in most patients, although it may be unsuitable for those with noncorrectable coagulopathies or in patients who cannot stop anticoagulants. Postsurgical patients, on the other hand, should tolerate the procedure just fine.<br/><br/>Patients with risk of gastric cancer “might be an important group” for evaluating the power-wash procedure, the investigators wrote, noting that combining the approach with artificial intelligence could one day yield even better results. <br/><br/>In the meantime, Dr. Lightdale and colleagues — like so many weekend warriors wielding a power-washer — are going to see if a different nozzle will take their work to the next level.<br/><br/>“We are actively studying a catheter with a broader stream and the potential to increase efficiency and decrease procedure time,” they wrote. “Another catheter design might allow for simultaneous spray and suction, so that cytology samples from specific regions of the stomach could be separately analyzed.”<br/><br/>This study was funded by Dalio Philanthropies, the Price Family Foundation, and the Frederic and Patricia Salerno Foundation. The investigators disclosed relationships with Boston Scientific, Interscope, Medtronic, 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>A Promising Approach, but Challenges Remain</title> <deck/> </itemMeta> <itemContent> <p>The optimal surveillance endoscopic modality for gastric intestinal metaplasia (GIM) is yet to be determined. Although the updated Sydney System, a comprehensive endoscopic biopsy protocol, has been advocated for GIM mapping, challenges are the heterogeneous distribution of GIM, suboptimal diagnostic accuracy of endoscopy to detect GIM, and the cost burden of multiple biopsies. </p> <p>This study by Lightdale et al. demonstrated the technical feasibility and safety of obtaining cytology for the detection of gastric intestinal metaplasia by using a systemic endoscopy-guided high-pressure spray “power-wash” method. In this study, all cytophathology samples in nine subjects were deemed satisfactory for evaluation. All three subjects who were cytology positive for GIM on H&amp;E stain and confirmed with positive immunohistochemistry (IHC) showed GIM on biopsy, and one subject had cells highly suspicious for dysplasia on cytology but biopsy was negative. Although all patients showed multiple superficial erosions after power-wash, bleeding was minimal and no adverse events related to power-wash were observed.<br/><br/>Applying cytology for detection of GIM appears promising as the way of collecting samples from the large surface area of the stomach. As clinicians, however, we are still left with some challenges. Even if cells collected are suspicious for dysplasia/neoplasia by this power-wash method, it would not be useful unless we precisely localize the area as we can not provide a focal curative endoscopic treatment. It is critical to increase the yield of localization of cytology sampling. Further research is also needed to standardize the cytopathologic diagnostic criteria of GIM and cost-effectiveness of the cytology-based approach compared to the current gold-standard biopsy protocol for the diagnosis of GIM.</p> <p><em> <em>Yutaka Tomizawa, MD, MSc, is a therapeutic endoscopist and clinical associate professor of medicine, Division of Gastroenterology, University of Washington, Seattle. He has no conflicts related to this report.</em> </em></p> </itemContent> </newsItem> </itemSet></root>
Article Source

FROM TECHNIQUES AND INNOVATIONS IN GASTROINTESTINAL ENDOSCOPY

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

New Quality Measure Improves Follow-Up for CRC Screening

Article Type
Changed
Mon, 04/15/2024 - 12:09

A newly developed quality performance measure that tracks completion of colorectal cancer (CRC) screening with a colonoscopy within 6 months of an abnormal stool-based screening test (SBT) in adults could help address high rates of incomplete CRC screening, the developers said.

As part of their work, the researchers conducted a retrospective study of 20,581 adults aged 50-75 years from 38 health systems that showed that fewer than half (48%) had a follow-up colonoscopy within 180 days of an initial abnormal SBT for CRC.

“The low follow-up rates to an abnormal SBT were initially surprising,” first author Elizabeth L. Ciemins, PhD, MPH, MA, Research and Analytics, American Medical Group Association (AMGA), Alexandria, Virginia, told this news organization.

“However, once we interviewed clinicians and learned that this was not a measure they were tracking, along with their own incorrect assumptions of a much higher follow-up rate, the low rates made sense. As is commonly said, ‘you can’t change what you don’t measure,’” she said.

The CRC screening completion measure the researchers propose “builds on and addresses an important shortcoming in an existing measure and will help ensure complete screening for CRC,” they noted in their JAMA Network Open paper.

The key elements of the follow-up measure are the date and result of a SBT and the date of the follow-up colonoscopy — if it occurred, Dr. Ciemins explained.

“Currently, health systems are not consistently tracking this measure, but they have the data elements to do so, especially if they are doing colonoscopies in-house,” she said.

Field testing showed that use of this new measure is “feasible, valid, and reliable,” the authors said. Dr. Ciemins believed this CRC screening completion measure could be widely implemented.

“Three AMGA member health systems feasibility tested the data elements and found that they could reliably abstract the required elements from electronic health records (EHRs),” she told this news organization.

The researchers are currently testing the measure among 20 AMGA member health systems, that are submitting quarterly data on a version of the specified measure.

“Advancing this measure as a quality performance measure could significantly increase the early detection of CRC, thereby improving health and ultimately saving lives,” the authors concluded in their paper.

The Right Direction, But Questions Remain

The coauthors of a linked commentary said this research highlights the “suboptimal” rates of a timely follow-up colonoscopy after positive SBT results. They applauded the authors for “focusing attention on a meaningful approach to measuring high-quality CRC screening and providing guidance for standardized measurement.”

However, several questions arise from this study, “including whether 6 months is the ideal interval for colonoscopy completion after a positive SBT result, where this measure fits in the context of existing CRC screening measures, and how to implement it in practice,” Jennifer K. Maratt, MD, with Indiana University School of Medicine, Indianapolis, and coauthors wrote.

“This measure alone does not address all the gaps in the screening process, nor does it address barriers to colonoscopy completion, but it points us in the right direction for measuring the success of screening programs,” Dr. Maratt and her colleagues added.

The study was supported by a grant from the AARP. The authors and editorial writers had no relevant disclosures.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

A newly developed quality performance measure that tracks completion of colorectal cancer (CRC) screening with a colonoscopy within 6 months of an abnormal stool-based screening test (SBT) in adults could help address high rates of incomplete CRC screening, the developers said.

As part of their work, the researchers conducted a retrospective study of 20,581 adults aged 50-75 years from 38 health systems that showed that fewer than half (48%) had a follow-up colonoscopy within 180 days of an initial abnormal SBT for CRC.

“The low follow-up rates to an abnormal SBT were initially surprising,” first author Elizabeth L. Ciemins, PhD, MPH, MA, Research and Analytics, American Medical Group Association (AMGA), Alexandria, Virginia, told this news organization.

“However, once we interviewed clinicians and learned that this was not a measure they were tracking, along with their own incorrect assumptions of a much higher follow-up rate, the low rates made sense. As is commonly said, ‘you can’t change what you don’t measure,’” she said.

The CRC screening completion measure the researchers propose “builds on and addresses an important shortcoming in an existing measure and will help ensure complete screening for CRC,” they noted in their JAMA Network Open paper.

The key elements of the follow-up measure are the date and result of a SBT and the date of the follow-up colonoscopy — if it occurred, Dr. Ciemins explained.

“Currently, health systems are not consistently tracking this measure, but they have the data elements to do so, especially if they are doing colonoscopies in-house,” she said.

Field testing showed that use of this new measure is “feasible, valid, and reliable,” the authors said. Dr. Ciemins believed this CRC screening completion measure could be widely implemented.

“Three AMGA member health systems feasibility tested the data elements and found that they could reliably abstract the required elements from electronic health records (EHRs),” she told this news organization.

The researchers are currently testing the measure among 20 AMGA member health systems, that are submitting quarterly data on a version of the specified measure.

“Advancing this measure as a quality performance measure could significantly increase the early detection of CRC, thereby improving health and ultimately saving lives,” the authors concluded in their paper.

The Right Direction, But Questions Remain

The coauthors of a linked commentary said this research highlights the “suboptimal” rates of a timely follow-up colonoscopy after positive SBT results. They applauded the authors for “focusing attention on a meaningful approach to measuring high-quality CRC screening and providing guidance for standardized measurement.”

However, several questions arise from this study, “including whether 6 months is the ideal interval for colonoscopy completion after a positive SBT result, where this measure fits in the context of existing CRC screening measures, and how to implement it in practice,” Jennifer K. Maratt, MD, with Indiana University School of Medicine, Indianapolis, and coauthors wrote.

“This measure alone does not address all the gaps in the screening process, nor does it address barriers to colonoscopy completion, but it points us in the right direction for measuring the success of screening programs,” Dr. Maratt and her colleagues added.

The study was supported by a grant from the AARP. The authors and editorial writers had no relevant disclosures.

A version of this article appeared on Medscape.com.

A newly developed quality performance measure that tracks completion of colorectal cancer (CRC) screening with a colonoscopy within 6 months of an abnormal stool-based screening test (SBT) in adults could help address high rates of incomplete CRC screening, the developers said.

As part of their work, the researchers conducted a retrospective study of 20,581 adults aged 50-75 years from 38 health systems that showed that fewer than half (48%) had a follow-up colonoscopy within 180 days of an initial abnormal SBT for CRC.

“The low follow-up rates to an abnormal SBT were initially surprising,” first author Elizabeth L. Ciemins, PhD, MPH, MA, Research and Analytics, American Medical Group Association (AMGA), Alexandria, Virginia, told this news organization.

“However, once we interviewed clinicians and learned that this was not a measure they were tracking, along with their own incorrect assumptions of a much higher follow-up rate, the low rates made sense. As is commonly said, ‘you can’t change what you don’t measure,’” she said.

The CRC screening completion measure the researchers propose “builds on and addresses an important shortcoming in an existing measure and will help ensure complete screening for CRC,” they noted in their JAMA Network Open paper.

The key elements of the follow-up measure are the date and result of a SBT and the date of the follow-up colonoscopy — if it occurred, Dr. Ciemins explained.

“Currently, health systems are not consistently tracking this measure, but they have the data elements to do so, especially if they are doing colonoscopies in-house,” she said.

Field testing showed that use of this new measure is “feasible, valid, and reliable,” the authors said. Dr. Ciemins believed this CRC screening completion measure could be widely implemented.

“Three AMGA member health systems feasibility tested the data elements and found that they could reliably abstract the required elements from electronic health records (EHRs),” she told this news organization.

The researchers are currently testing the measure among 20 AMGA member health systems, that are submitting quarterly data on a version of the specified measure.

“Advancing this measure as a quality performance measure could significantly increase the early detection of CRC, thereby improving health and ultimately saving lives,” the authors concluded in their paper.

The Right Direction, But Questions Remain

The coauthors of a linked commentary said this research highlights the “suboptimal” rates of a timely follow-up colonoscopy after positive SBT results. They applauded the authors for “focusing attention on a meaningful approach to measuring high-quality CRC screening and providing guidance for standardized measurement.”

However, several questions arise from this study, “including whether 6 months is the ideal interval for colonoscopy completion after a positive SBT result, where this measure fits in the context of existing CRC screening measures, and how to implement it in practice,” Jennifer K. Maratt, MD, with Indiana University School of Medicine, Indianapolis, and coauthors wrote.

“This measure alone does not address all the gaps in the screening process, nor does it address barriers to colonoscopy completion, but it points us in the right direction for measuring the success of screening programs,” Dr. Maratt and her colleagues added.

The study was supported by a grant from the AARP. The authors and editorial writers had 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>167606</fileName> <TBEID>0C04F760.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F760</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240408T161425</QCDate> <firstPublished>20240409T094032</firstPublished> <LastPublished>20240409T094032</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240409T094032</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Megan Brooks</byline> <bylineText>MEGAN BROOKS</bylineText> <bylineFull>MEGAN BROOKS</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>A newly developed quality performance measure that tracks completion of colorectal cancer (CRC) screening with a colonoscopy within 6 months of an abnormal stoo</metaDescription> <articlePDF/> <teaserImage/> <teaser>Field testing shows that this CRC screening tracker is “feasible, valid, and reliable.”</teaser> <title>New Quality Measure Improves Follow-Up for CRC Screening</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>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>GIHOLD</publicationCode> <pubIssueName>January 2014</pubIssueName> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> </publications_g> <publications> <term>21</term> <term>15</term> <term canonical="true">31</term> </publications> <sections> <term canonical="true">27970</term> <term>39313</term> </sections> <topics> <term>213</term> <term canonical="true">280</term> <term>263</term> <term>67020</term> <term>278</term> <term>38029</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>New Quality Measure Improves Follow-Up for CRC Screening</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">A newly developed quality performance measure that tracks completion of colorectal cancer (CRC) screening with a colonoscopy within 6 months of an abnormal stool-based screening test (SBT) in adults could help address high rates of incomplete CRC screening,</span> the developers said.</p> <p>As part of their work, the researchers conducted a retrospective study of 20,581 adults aged 50-75 years from 38 health systems that showed that fewer than half (48%) had a follow-up colonoscopy within 180 days of an initial abnormal SBT for CRC.<br/><br/>“The low follow-up rates to an abnormal SBT were initially surprising,” first author Elizabeth L. Ciemins, PhD, MPH, MA, Research and Analytics, American Medical Group Association (AMGA), Alexandria, Virginia, told this news organization.<br/><br/>“However, once we interviewed clinicians and learned that this was not a measure they were tracking, along with their own incorrect assumptions of a much higher follow-up rate, the low rates made sense. As is commonly said, ‘you can’t change what you don’t measure,’” she said.<br/><br/>The CRC screening completion measure the researchers propose “builds on and addresses an important shortcoming in an existing measure and will help ensure complete screening for CRC,” they noted in their <a href="https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2816737"><span class="Emphasis">JAMA Network Open</span> paper</a>.<br/><br/>The key elements of the follow-up measure are the date and result of a SBT and the date of the follow-up colonoscopy — if it occurred, Dr. Ciemins explained.<br/><br/>“Currently, health systems are not consistently tracking this measure, but they have the data elements to do so, especially if they are doing colonoscopies in-house,” she said.<br/><br/>Field testing showed that use of this new measure is “feasible, valid, and reliable,” the authors said. Dr. Ciemins believed this CRC screening completion measure could be widely implemented.<br/><br/>“Three AMGA member health systems feasibility tested the data elements and found that they could reliably abstract the required elements from electronic health records (EHRs),” she told this news organization.<br/><br/>The researchers are currently testing the measure among 20 AMGA member health systems, that are submitting quarterly data on a version of the specified measure.<br/><br/>“Advancing this measure as a quality performance measure could significantly increase the early detection of CRC, thereby improving health and ultimately saving lives,” the authors concluded in their paper.</p> <h2>The Right Direction, But Questions Remain</h2> <p>The coauthors of a <a href="https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2816739">linked commentary</a> said this research highlights the “suboptimal” rates of a timely follow-up colonoscopy after positive SBT results. They applauded the authors for “focusing attention on a meaningful approach to measuring high-quality CRC screening and providing guidance for standardized measurement.”</p> <p>However, several questions arise from this study, “including whether 6 months is the ideal interval for colonoscopy completion after a positive SBT result, where this measure fits in the context of existing CRC screening measures, and how to implement it in practice,” Jennifer K. Maratt, MD, with Indiana University School of Medicine, Indianapolis, and coauthors wrote.<br/><br/>“This measure alone does not address all the gaps in the screening process, nor does it address barriers to colonoscopy completion, but it points us in the right direction for measuring the success of screening programs,” Dr. Maratt and her colleagues added.<br/><br/>The study was supported by a grant from the AARP. The authors and editorial writers had 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/new-quality-measure-improves-follow-crc-screening-2024a10006lb">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

Computer-Aided Colonoscopy Falls Short in Real-World Practice

Jury is still out on CADe
Article Type
Changed
Mon, 04/15/2024 - 11:30

Colonoscopy with computer-aided detection (CADe) fails to improve adenoma detection rate (ADR) in real-world, nonrandomized trials, according to investigators.

Although CADe did not increase burden of colonoscopy in the real-world, these real-world detection rates casts doubt on the generalizability of positive findings from randomized trials, reported lead author Harsh K. Patel, MD, of the University of Kansas Medical Center, Kansas City, Missouri, and colleagues.

CADe-assisted colonoscopy has gained increasing attention for its potential to improve ADR, particularly with the recent publication of a meta-analysis involving 20 randomized controlled trials (RCTs), Dr. Patel and colleagues wrote in Clinical Gastroenterology and Hepatology. “However, results of RCTs are not necessarily reproducible in clinical practice.”

RCTs evaluating this technology are susceptible to various issues with validity, they noted, such as psychological bias stemming from lack of blinding to the possibility that CADe could reduce operator attention, paradoxically “deskilling” endoscopists.

The present meta-analysis aimed to overcome these potential shortfalls by analyzing nonrandomized data from eight studies involving 9,782 patients.

Patel_Harsh_K_MO_web.jpg
Dr. Harsh K. Patel

“The lack of a highly controlled setting reduces the psychological pressure of the endoscopists to demonstrate a possible benefit of CADe (i.e., the operator bias) and allows endoscopists to use CADe according to their preferences and attitudes which we usually experience in a real-world clinical practice,” the investigators wrote. “On the other hand, noncontrolled factors may affect the outcome of the study, especially when considering that an equivalent distribution of prevalence of disease is required for a fair assessment of the effectiveness of the intervention.”

This approach revealed less favorable outcomes than those reported by RCTs.

CADe-assisted ADR was not significantly different from ADR for standard colonoscopy (44% vs 38%; risk ratio, 1.11; 95% CI, 0.97-1.28), nor was mean number of adenomas detected per colonoscopy (0.93 vs 0.79; mean difference, 0.14; 95% CI, -0.04-0.32).

“Our study provides a contrasting perspective to those results previously known from the randomized studies,” the investigators wrote.

While detection benefits were not identified, burden of CADe-assisted colonoscopy was not elevated either.

Mean nonneoplastic lesions per colonoscopy was similar between modalities (0.52 vs 0.47; mean difference, 0.14; 95% CI, -0.07-0.34), as was withdrawal time (14.3 vs 13.4 minutes; mean difference, 0.8 minutes; 95% CI, -0.18-1.90).

Dr. Patel and colleagues described “a high level of heterogeneity that was qualitatively and quantitatively distinct from the heterogeneity discovered in the prior meta-analysis of RCTs.” Unlike the RCT meta-analysis, which had no studies with an ADR outcome favoring the control arm, the present meta-analysis found that one third of the included studies favored the control arm.

“This qualitative difference generates a much higher degree of ambiguity, as it does not apply only to the magnitude of the effect of CADe, but it puts in question the actual existence of any CADe-related benefit,” they wrote. “An important point to make is that the analysis of adenoma and serrated lesions per colonoscopy supported the qualitative heterogeneity, favoring the control arm over the CADe arm, in the direction of the effect.”

Dr. Patel and colleagues suggested that the concurrent lack of benefit and lack of harm associated with CADe in the present meta-analysis is “interesting,” and may point to underutilization or a lack of effect of CADe.

“To address the uncertainties in the current literature, we recommend conducting additional randomized studies in a more pragmatic setting,” they concluded.

This meta-analysis was supported by the European Commission and AIRC. The investigators disclosed relationships with NEC, Satisfy, Odin, and others.

Body

 

The advent of AI in colonoscopy through computer-aided detection (CADe) systems has been promising, with over 20 randomized controlled trials (RCTs) affirming its benefits. However, this enthusiasm has been tempered by several recent nonrandomized studies indicating no real-world advantage, as discussed in Patel et al.’s systematic review and meta-analysis in Clinical Gastroenterology and Hepatology.

[[{"fid":"300209","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"}},"attributes":{"alt":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","class":"media-element file-medstat-image-flush-left","data-delta":"2"}}]]The stark differences in the results of RCTs and nonrandomized studies with CADe are interesting and thought-provoking, highlighting issues like potential RCT bias (due to lack of blinding) and the critical role of the human-AI interaction. It may be that some endoscopists derive a benefit from CADe while others do not, and further studies looking into the performance of individual endoscopists with and without CADe may be helpful. The meta-analysis also reveals varying outcomes based on study design — prospective or retrospective — and the nature of the control arm, be it concurrent or historical.

In addition, a critical consideration with evaluating any AI/CADe system is they often undergo frequent updates, each promising improved accuracy, sensitivity, and specificity. This is an interesting dilemma and raises questions about the enduring relevance of studies conducted using outdated versions of CADe.

In my opinion, the jury is still out on the effectiveness of CADe for colonoscopy in a real-world setting. The definitive assessment of CADe’s real-world value necessitates larger, well-structured trials that mirror actual clinical environments and span extended periods of time, taking care to minimize biases that may have influenced the results of current published studies.

Nabil M. Mansour, MD, is assistant professor of medicine in the Section of Gastroenterology, Baylor College of Medicine, Houston. He has served as a consultant for Iterative Health.

Publications
Topics
Sections
Body

 

The advent of AI in colonoscopy through computer-aided detection (CADe) systems has been promising, with over 20 randomized controlled trials (RCTs) affirming its benefits. However, this enthusiasm has been tempered by several recent nonrandomized studies indicating no real-world advantage, as discussed in Patel et al.’s systematic review and meta-analysis in Clinical Gastroenterology and Hepatology.

[[{"fid":"300209","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"}},"attributes":{"alt":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","class":"media-element file-medstat-image-flush-left","data-delta":"2"}}]]The stark differences in the results of RCTs and nonrandomized studies with CADe are interesting and thought-provoking, highlighting issues like potential RCT bias (due to lack of blinding) and the critical role of the human-AI interaction. It may be that some endoscopists derive a benefit from CADe while others do not, and further studies looking into the performance of individual endoscopists with and without CADe may be helpful. The meta-analysis also reveals varying outcomes based on study design — prospective or retrospective — and the nature of the control arm, be it concurrent or historical.

In addition, a critical consideration with evaluating any AI/CADe system is they often undergo frequent updates, each promising improved accuracy, sensitivity, and specificity. This is an interesting dilemma and raises questions about the enduring relevance of studies conducted using outdated versions of CADe.

In my opinion, the jury is still out on the effectiveness of CADe for colonoscopy in a real-world setting. The definitive assessment of CADe’s real-world value necessitates larger, well-structured trials that mirror actual clinical environments and span extended periods of time, taking care to minimize biases that may have influenced the results of current published studies.

Nabil M. Mansour, MD, is assistant professor of medicine in the Section of Gastroenterology, Baylor College of Medicine, Houston. He has served as a consultant for Iterative Health.

Body

 

The advent of AI in colonoscopy through computer-aided detection (CADe) systems has been promising, with over 20 randomized controlled trials (RCTs) affirming its benefits. However, this enthusiasm has been tempered by several recent nonrandomized studies indicating no real-world advantage, as discussed in Patel et al.’s systematic review and meta-analysis in Clinical Gastroenterology and Hepatology.

[[{"fid":"300209","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"},"type":"media","field_deltas":{"2":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"}},"attributes":{"alt":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","class":"media-element file-medstat-image-flush-left","data-delta":"2"}}]]The stark differences in the results of RCTs and nonrandomized studies with CADe are interesting and thought-provoking, highlighting issues like potential RCT bias (due to lack of blinding) and the critical role of the human-AI interaction. It may be that some endoscopists derive a benefit from CADe while others do not, and further studies looking into the performance of individual endoscopists with and without CADe may be helpful. The meta-analysis also reveals varying outcomes based on study design — prospective or retrospective — and the nature of the control arm, be it concurrent or historical.

In addition, a critical consideration with evaluating any AI/CADe system is they often undergo frequent updates, each promising improved accuracy, sensitivity, and specificity. This is an interesting dilemma and raises questions about the enduring relevance of studies conducted using outdated versions of CADe.

In my opinion, the jury is still out on the effectiveness of CADe for colonoscopy in a real-world setting. The definitive assessment of CADe’s real-world value necessitates larger, well-structured trials that mirror actual clinical environments and span extended periods of time, taking care to minimize biases that may have influenced the results of current published studies.

Nabil M. Mansour, MD, is assistant professor of medicine in the Section of Gastroenterology, Baylor College of Medicine, Houston. He has served as a consultant for Iterative Health.

Title
Jury is still out on CADe
Jury is still out on CADe

Colonoscopy with computer-aided detection (CADe) fails to improve adenoma detection rate (ADR) in real-world, nonrandomized trials, according to investigators.

Although CADe did not increase burden of colonoscopy in the real-world, these real-world detection rates casts doubt on the generalizability of positive findings from randomized trials, reported lead author Harsh K. Patel, MD, of the University of Kansas Medical Center, Kansas City, Missouri, and colleagues.

CADe-assisted colonoscopy has gained increasing attention for its potential to improve ADR, particularly with the recent publication of a meta-analysis involving 20 randomized controlled trials (RCTs), Dr. Patel and colleagues wrote in Clinical Gastroenterology and Hepatology. “However, results of RCTs are not necessarily reproducible in clinical practice.”

RCTs evaluating this technology are susceptible to various issues with validity, they noted, such as psychological bias stemming from lack of blinding to the possibility that CADe could reduce operator attention, paradoxically “deskilling” endoscopists.

The present meta-analysis aimed to overcome these potential shortfalls by analyzing nonrandomized data from eight studies involving 9,782 patients.

Patel_Harsh_K_MO_web.jpg
Dr. Harsh K. Patel

“The lack of a highly controlled setting reduces the psychological pressure of the endoscopists to demonstrate a possible benefit of CADe (i.e., the operator bias) and allows endoscopists to use CADe according to their preferences and attitudes which we usually experience in a real-world clinical practice,” the investigators wrote. “On the other hand, noncontrolled factors may affect the outcome of the study, especially when considering that an equivalent distribution of prevalence of disease is required for a fair assessment of the effectiveness of the intervention.”

This approach revealed less favorable outcomes than those reported by RCTs.

CADe-assisted ADR was not significantly different from ADR for standard colonoscopy (44% vs 38%; risk ratio, 1.11; 95% CI, 0.97-1.28), nor was mean number of adenomas detected per colonoscopy (0.93 vs 0.79; mean difference, 0.14; 95% CI, -0.04-0.32).

“Our study provides a contrasting perspective to those results previously known from the randomized studies,” the investigators wrote.

While detection benefits were not identified, burden of CADe-assisted colonoscopy was not elevated either.

Mean nonneoplastic lesions per colonoscopy was similar between modalities (0.52 vs 0.47; mean difference, 0.14; 95% CI, -0.07-0.34), as was withdrawal time (14.3 vs 13.4 minutes; mean difference, 0.8 minutes; 95% CI, -0.18-1.90).

Dr. Patel and colleagues described “a high level of heterogeneity that was qualitatively and quantitatively distinct from the heterogeneity discovered in the prior meta-analysis of RCTs.” Unlike the RCT meta-analysis, which had no studies with an ADR outcome favoring the control arm, the present meta-analysis found that one third of the included studies favored the control arm.

“This qualitative difference generates a much higher degree of ambiguity, as it does not apply only to the magnitude of the effect of CADe, but it puts in question the actual existence of any CADe-related benefit,” they wrote. “An important point to make is that the analysis of adenoma and serrated lesions per colonoscopy supported the qualitative heterogeneity, favoring the control arm over the CADe arm, in the direction of the effect.”

Dr. Patel and colleagues suggested that the concurrent lack of benefit and lack of harm associated with CADe in the present meta-analysis is “interesting,” and may point to underutilization or a lack of effect of CADe.

“To address the uncertainties in the current literature, we recommend conducting additional randomized studies in a more pragmatic setting,” they concluded.

This meta-analysis was supported by the European Commission and AIRC. The investigators disclosed relationships with NEC, Satisfy, Odin, and others.

Colonoscopy with computer-aided detection (CADe) fails to improve adenoma detection rate (ADR) in real-world, nonrandomized trials, according to investigators.

Although CADe did not increase burden of colonoscopy in the real-world, these real-world detection rates casts doubt on the generalizability of positive findings from randomized trials, reported lead author Harsh K. Patel, MD, of the University of Kansas Medical Center, Kansas City, Missouri, and colleagues.

CADe-assisted colonoscopy has gained increasing attention for its potential to improve ADR, particularly with the recent publication of a meta-analysis involving 20 randomized controlled trials (RCTs), Dr. Patel and colleagues wrote in Clinical Gastroenterology and Hepatology. “However, results of RCTs are not necessarily reproducible in clinical practice.”

RCTs evaluating this technology are susceptible to various issues with validity, they noted, such as psychological bias stemming from lack of blinding to the possibility that CADe could reduce operator attention, paradoxically “deskilling” endoscopists.

The present meta-analysis aimed to overcome these potential shortfalls by analyzing nonrandomized data from eight studies involving 9,782 patients.

Patel_Harsh_K_MO_web.jpg
Dr. Harsh K. Patel

“The lack of a highly controlled setting reduces the psychological pressure of the endoscopists to demonstrate a possible benefit of CADe (i.e., the operator bias) and allows endoscopists to use CADe according to their preferences and attitudes which we usually experience in a real-world clinical practice,” the investigators wrote. “On the other hand, noncontrolled factors may affect the outcome of the study, especially when considering that an equivalent distribution of prevalence of disease is required for a fair assessment of the effectiveness of the intervention.”

This approach revealed less favorable outcomes than those reported by RCTs.

CADe-assisted ADR was not significantly different from ADR for standard colonoscopy (44% vs 38%; risk ratio, 1.11; 95% CI, 0.97-1.28), nor was mean number of adenomas detected per colonoscopy (0.93 vs 0.79; mean difference, 0.14; 95% CI, -0.04-0.32).

“Our study provides a contrasting perspective to those results previously known from the randomized studies,” the investigators wrote.

While detection benefits were not identified, burden of CADe-assisted colonoscopy was not elevated either.

Mean nonneoplastic lesions per colonoscopy was similar between modalities (0.52 vs 0.47; mean difference, 0.14; 95% CI, -0.07-0.34), as was withdrawal time (14.3 vs 13.4 minutes; mean difference, 0.8 minutes; 95% CI, -0.18-1.90).

Dr. Patel and colleagues described “a high level of heterogeneity that was qualitatively and quantitatively distinct from the heterogeneity discovered in the prior meta-analysis of RCTs.” Unlike the RCT meta-analysis, which had no studies with an ADR outcome favoring the control arm, the present meta-analysis found that one third of the included studies favored the control arm.

“This qualitative difference generates a much higher degree of ambiguity, as it does not apply only to the magnitude of the effect of CADe, but it puts in question the actual existence of any CADe-related benefit,” they wrote. “An important point to make is that the analysis of adenoma and serrated lesions per colonoscopy supported the qualitative heterogeneity, favoring the control arm over the CADe arm, in the direction of the effect.”

Dr. Patel and colleagues suggested that the concurrent lack of benefit and lack of harm associated with CADe in the present meta-analysis is “interesting,” and may point to underutilization or a lack of effect of CADe.

“To address the uncertainties in the current literature, we recommend conducting additional randomized studies in a more pragmatic setting,” they concluded.

This meta-analysis was supported by the European Commission and AIRC. The investigators disclosed relationships with NEC, Satisfy, Odin, 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>167486</fileName> <TBEID>0C04F4A6.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F4A6</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>CGH_Patel_CADe</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240402T101407</QCDate> <firstPublished>20240402T110127</firstPublished> <LastPublished>20240402T110127</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240402T110127</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>Colonoscopy with computer-aided detection (CADe) fails to improve adenoma detection rate (ADR) in real-world, nonrandomized trials</metaDescription> <articlePDF/> <teaserImage>300921</teaserImage> <teaser>CADe-assisted colonoscopy has gained increasing attention for its potential to improve adenoma detection rate.</teaser> <title>Computer-Aided Colonoscopy Falls Short in Real-World Practice</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>345</term> <term>344</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240127be.jpg</altRep> <description role="drol:caption">Dr. Harsh K. Patel</description> <description role="drol:credit">University of Kansas Medical Center</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24012632.jpg</altRep> <description role="drol:caption">Dr. Nabil M. Mansour</description> <description role="drol:credit">Baylor College of Medicine</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Computer-Aided Colonoscopy Falls Short in Real-World Practice</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Colonoscopy with computer-aided detection (CADe) fails to improve adenoma detection rate (ADR) in real-world, nonrandomized trials</span>, according to investigators.</p> <p>Although CADe did not increase burden of colonoscopy in the real-world, these real-world detection rates casts doubt on the generalizability of positive findings from randomized trials, reported lead author <a href="https://scholar.google.com/citations?user=8vZQbFwAAAAJ&amp;hl=en">Harsh K. Patel, MD</a>, of the University of Kansas Medical Center, Kansas City, Missouri, and colleagues.<br/><br/>CADe-assisted colonoscopy has gained increasing attention for its potential to improve ADR, particularly with the recent publication of a <a href="https://www.acpjournals.org/doi/10.7326/M22-3678">meta-analysis</a> involving 20 randomized controlled trials (RCTs), Dr. Patel and colleagues <span class="Hyperlink"><a href="https://www.cghjournal.org/article/S1542-3565(23)00968-0/abstract">wrote</a></span> in <em>Clinical Gastroenterology and Hepatology</em>. “However, results of RCTs are not necessarily reproducible in clinical practice.”<br/><br/>RCTs evaluating this technology are susceptible to various issues with validity, they noted, such as psychological bias stemming from lack of blinding to the possibility that CADe could reduce operator attention, paradoxically “deskilling” endoscopists.<br/><br/>The present meta-analysis aimed to overcome these potential shortfalls by analyzing nonrandomized data from eight studies involving 9,782 patients. <br/><br/>[[{"fid":"300921","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. Harsh K. Patel, University of Kansas Medical Center, Kansas City, Missouri","field_file_image_credit[und][0][value]":"University of Kansas Medical Center","field_file_image_caption[und][0][value]":"Dr. Harsh K. Patel"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]“The lack of a highly controlled setting reduces the psychological pressure of the endoscopists to demonstrate a possible benefit of CADe (i.e., the operator bias) and allows endoscopists to use CADe according to their preferences and attitudes which we usually experience in a real-world clinical practice,” the investigators wrote. “On the other hand, noncontrolled factors may affect the outcome of the study, especially when considering that an equivalent distribution of prevalence of disease is required for a fair assessment of the effectiveness of the intervention.”<br/><br/>This approach revealed less favorable outcomes than those reported by RCTs.<br/><br/>CADe-assisted ADR was not significantly different from ADR for standard colonoscopy (44% vs 38%; risk ratio, 1.11; 95% CI, 0.97-1.28), nor was mean number of adenomas detected per colonoscopy (0.93 vs 0.79; mean difference, 0.14; 95% CI, -0.04-0.32). <br/><br/>“Our study provides a contrasting perspective to those results previously known from the randomized studies,” the investigators wrote.<br/><br/>While detection benefits were not identified, burden of CADe-assisted colonoscopy was not elevated either.<br/><br/>Mean nonneoplastic lesions per colonoscopy was similar between modalities (0.52 vs 0.47; mean difference, 0.14; 95% CI, -0.07-0.34), as was withdrawal time (14.3 vs 13.4 minutes; mean difference, 0.8 minutes; 95% CI, -0.18-1.90).<br/><br/>Dr. Patel and colleagues described “a high level of heterogeneity that was qualitatively and quantitatively distinct from the heterogeneity discovered in the prior meta-analysis of RCTs.” Unlike the RCT meta-analysis, which had no studies with an ADR outcome favoring the control arm, the present meta-analysis found that one third of the included studies favored the control arm.<br/><br/>“This qualitative difference generates a much higher degree of ambiguity, as it does not apply only to the magnitude of the effect of CADe, but it puts in question the actual existence of any CADe-related benefit,” they wrote. “An important point to make is that the analysis of adenoma and serrated lesions per colonoscopy supported the qualitative heterogeneity, favoring the control arm over the CADe arm, in the direction of the effect.”<br/><br/>Dr. Patel and colleagues suggested that the concurrent lack of benefit and lack of harm associated with CADe in the present meta-analysis is “interesting,” and may point to underutilization or a lack of effect of CADe.<br/><br/>“To address the uncertainties in the current literature, we recommend conducting additional randomized studies in a more pragmatic setting,” they concluded.<br/><br/>This meta-analysis was supported by the European Commission and AIRC. The investigators disclosed relationships with NEC, Satisfy, Odin, 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>Jury is still out on CADe</title> <deck/> </itemMeta> <itemContent> <p>The advent of AI in colonoscopy through computer-aided detection (CADe) systems has been promising, with over 20 randomized controlled trials (RCTs) affirming its benefits. However, this enthusiasm has been tempered by several recent nonrandomized studies indicating no real-world advantage, as discussed in Patel et al.’s systematic review and meta-analysis in <em>Clinical Gastroenterology and Hepatology</em>. </p> <p>[[{"fid":"300209","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. Nabil M. Mansour, McNair General GI Clinic at Baylor College of Medicine in Houston","field_file_image_credit[und][0][value]":"Baylor College of Medicine","field_file_image_caption[und][0][value]":"Dr. Nabil M. Mansour"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]The stark differences in the results of RCTs and nonrandomized studies with CADe are interesting and thought-provoking, highlighting issues like potential RCT bias (due to lack of blinding) and the critical role of the human-AI interaction. It may be that some endoscopists derive a benefit from CADe while others do not, and further studies looking into the performance of individual endoscopists with and without CADe may be helpful. The meta-analysis also reveals varying outcomes based on study design — prospective or retrospective — and the nature of the control arm, be it concurrent or historical.<br/><br/>In addition, a critical consideration with evaluating any AI/CADe system is they often undergo frequent updates, each promising improved accuracy, sensitivity, and specificity. This is an interesting dilemma and raises questions about the enduring relevance of studies conducted using outdated versions of CADe.<br/><br/>In my opinion, the jury is still out on the effectiveness of CADe for colonoscopy in a real-world setting. The definitive assessment of CADe’s real-world value necessitates larger, well-structured trials that mirror actual clinical environments and span extended periods of time, taking care to minimize biases that may have influenced the results of current published studies.</p> <p><em> <em>Nabil M. Mansour, MD, is assistant professor of medicine in the Section of Gastroenterology, Baylor College of Medicine, Houston. He has served as a consultant for Iterative Health.</em> </em></p> </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

GI physician channels humor to incentivize cancer screenings

Article Type
Changed
Fri, 04/12/2024 - 12:42
Display Headline
GI Physician Channels Humor to Incentivize Cancer Screenings

Growing up in a household where GI issues dominated conversations, it’s no surprise that Shida Haghighat, MD, chose gastroenterology as her area of study in medicine.

She watched her father suffer from the complications of Crohn’s disease and her brother struggle with irritable bowel syndrome. “We always needed to know where the nearest bathroom was. I grew up with that around me, and I was always just fascinated by the gut and the digestive system,” said Dr. Haghighat, who just finished up her fellowship at the University of Miami and is now a gastroenterologist at University of California, Los Angeles. She also serves as social media editor for AGA’s Gastro Hep Advances.

Haghighat_Shida_CA_web.jpg
Dr. Shida Haghighat

As she got to know the personalities of the GI department in the first year of medical school, “I realized that our senses of humor and personalities kind of aligned, and I was like, ‘Oh yeah, this is where I’m supposed to be,’ ” said Dr. Haghighat, who can be found on X @DoctorShida.

Humor is something Dr. Haghighat has reached for throughout her life and career. She eventually channeled her gift for satire onto the stage and the internet, as a stand-up comedian. In an interview with GI & Hepatology News, she spoke about the connection between GI medicine and humor, and the creative ways she has helped promote cancer screening in underserved populations.
 

Q: What practice challenges have you faced in your career?

Dr. Haghighat: I trained in a county hospital, so I’ve always worked with underserved and vulnerable populations. One of the challenges has been just navigation of care, especially as it pertains to cancer diagnoses or cancer screening. A lot of the time, patients don’t understand why they have to do a test or something invasive like a colonoscopy for symptoms they don’t have. 

Q: A focus of yours has been improving uptake of screening in underserved communities. Please talk about the work you’ve done in this area.

Dr. Haghighat: I was at Los Angeles General Medical Center — a county hospital in Los Angeles — for residency, where we treated underserved, uninsured patients. I noticed in our primary care clinics a very low uptake of colon cancer screening. Patients didn’t want to bring the stool tests back or get colonoscopies. I surveyed a bunch of the patients and asked: How can we make colon cancer screening easier for you? About a third of the patients said, “If I can do it in the clinic before I go home, that would be great.”

So, I started this initiative called “Go Before You Go.” We would ask patients, “Hey, do you need to go to the bathroom right now, if you can?” Our nurses handed them the stool test to do in the bathroom before they left the clinic after their doctor visits.

We saw really good results with that. Surprisingly, a lot of people can go on demand. We saw increased screening rates, and that quality improvement project went on to win multiple first place awards in research competitions. So that’s what got me interested, and that’s where I had my beginnings of increasing preventative services in underserved communities on the ground.
 

 

 

Q: Can you discuss some health disparity studies you’ve done in this area?

Dr. Haghighat: As a GI at Jackson Memorial Hospital in Miami, I was seeing cancer disparities firsthand every day. I wanted to approach these disparities from a research funding standpoint on a federal level. I was particularly interested in gastric cancer because it’s not common enough in the United States to warrant universal screening, but it’s very common among certain racial and ethnic minorities, which would warrant targeted screening.

I evaluated cancer funding allocation from the National Cancer Institute among the most common cancers in the United States and found that cancer afflicting a higher proportion of racial and ethnic minorities was receiving lower funding. One of those cancers was stomach cancer. This study basically highlighted that, to decrease these disparities, a top-down policy approach is necessary to distribute cancer research funding equitably across these groups. 

A lot of stomach cancer comes from a bacteria called Helicobacter pylori, which can be more prevalent in certain countries. In another study, I looked at country of birth as a risk factor for stomach cancer, specifically for gastric intestinal metaplasia, which is a precursor for gastric cancer . 

We found that country of birth is a key risk factor for gastric intestinal metaplasia and that it should be incorporated into risk stratification for targeted screening. 
 

Q: Outside of medicine, you perform as a stand-up comedian. You have a popular satirical alias on social media. How did you get interested in stand-up comedy?

Dr. Haghighat: I gave my medical school’s commencement speech, and I had sprinkled a few jokes in there. Afterward, multiple people approached me and said, “You should really consider stand-up comedy. Your timing and delivery are great.” A few months later, I started my intern year in Los Angeles and simultaneously took stand-up comedy classes. I started performing at local clubs around town throughout residency, and I had two or three good sets that I could rely on. And so that’s how I got into stand-up comedy.

My intern year is also when I started this social media satire account. It was a way to cope with the anxieties and stress of residency. Before I knew it, the account gained multitudes of followers, doctors, and other medical professionals. And I joke that the more hours I work in a week, the more memes I make, the more posts I make. It’s kind of a creative outlet for me after a long day.
 

Q: What types of things do you talk about during your stand-up act?

Dr. Haghighat: A lot of it is about growing up in an immigrant household as a first-generation Iranian American. One of my favorite jokes is, my parents gave me so many options for a career. They said I could be a family doctor, a surgeon, a plastic surgeon, and if I worked hard, even a wife of a surgeon. But I talk a lot about being a woman in medicine. That always gets a lot of laughs. And now that I’ve graduated GI fellowship, I’m excited to incorporate some GI jokes because it turns out people love poop jokes.

 

 

Lightning Round

Texting or talking?

Text

Favorite city in U.S. besides the one you live in?

Denver

Cat or dog person

Dog

Best place you went on vacation

Patagonia

Favorite sport

Basketball

Favorite ice cream

Rocky Road

What song do you have to sing along with when you hear it?

Celine Dion’s My Heart Will Go On

Publications
Topics
Sections

Growing up in a household where GI issues dominated conversations, it’s no surprise that Shida Haghighat, MD, chose gastroenterology as her area of study in medicine.

She watched her father suffer from the complications of Crohn’s disease and her brother struggle with irritable bowel syndrome. “We always needed to know where the nearest bathroom was. I grew up with that around me, and I was always just fascinated by the gut and the digestive system,” said Dr. Haghighat, who just finished up her fellowship at the University of Miami and is now a gastroenterologist at University of California, Los Angeles. She also serves as social media editor for AGA’s Gastro Hep Advances.

Haghighat_Shida_CA_web.jpg
Dr. Shida Haghighat

As she got to know the personalities of the GI department in the first year of medical school, “I realized that our senses of humor and personalities kind of aligned, and I was like, ‘Oh yeah, this is where I’m supposed to be,’ ” said Dr. Haghighat, who can be found on X @DoctorShida.

Humor is something Dr. Haghighat has reached for throughout her life and career. She eventually channeled her gift for satire onto the stage and the internet, as a stand-up comedian. In an interview with GI & Hepatology News, she spoke about the connection between GI medicine and humor, and the creative ways she has helped promote cancer screening in underserved populations.
 

Q: What practice challenges have you faced in your career?

Dr. Haghighat: I trained in a county hospital, so I’ve always worked with underserved and vulnerable populations. One of the challenges has been just navigation of care, especially as it pertains to cancer diagnoses or cancer screening. A lot of the time, patients don’t understand why they have to do a test or something invasive like a colonoscopy for symptoms they don’t have. 

Q: A focus of yours has been improving uptake of screening in underserved communities. Please talk about the work you’ve done in this area.

Dr. Haghighat: I was at Los Angeles General Medical Center — a county hospital in Los Angeles — for residency, where we treated underserved, uninsured patients. I noticed in our primary care clinics a very low uptake of colon cancer screening. Patients didn’t want to bring the stool tests back or get colonoscopies. I surveyed a bunch of the patients and asked: How can we make colon cancer screening easier for you? About a third of the patients said, “If I can do it in the clinic before I go home, that would be great.”

So, I started this initiative called “Go Before You Go.” We would ask patients, “Hey, do you need to go to the bathroom right now, if you can?” Our nurses handed them the stool test to do in the bathroom before they left the clinic after their doctor visits.

We saw really good results with that. Surprisingly, a lot of people can go on demand. We saw increased screening rates, and that quality improvement project went on to win multiple first place awards in research competitions. So that’s what got me interested, and that’s where I had my beginnings of increasing preventative services in underserved communities on the ground.
 

 

 

Q: Can you discuss some health disparity studies you’ve done in this area?

Dr. Haghighat: As a GI at Jackson Memorial Hospital in Miami, I was seeing cancer disparities firsthand every day. I wanted to approach these disparities from a research funding standpoint on a federal level. I was particularly interested in gastric cancer because it’s not common enough in the United States to warrant universal screening, but it’s very common among certain racial and ethnic minorities, which would warrant targeted screening.

I evaluated cancer funding allocation from the National Cancer Institute among the most common cancers in the United States and found that cancer afflicting a higher proportion of racial and ethnic minorities was receiving lower funding. One of those cancers was stomach cancer. This study basically highlighted that, to decrease these disparities, a top-down policy approach is necessary to distribute cancer research funding equitably across these groups. 

A lot of stomach cancer comes from a bacteria called Helicobacter pylori, which can be more prevalent in certain countries. In another study, I looked at country of birth as a risk factor for stomach cancer, specifically for gastric intestinal metaplasia, which is a precursor for gastric cancer . 

We found that country of birth is a key risk factor for gastric intestinal metaplasia and that it should be incorporated into risk stratification for targeted screening. 
 

Q: Outside of medicine, you perform as a stand-up comedian. You have a popular satirical alias on social media. How did you get interested in stand-up comedy?

Dr. Haghighat: I gave my medical school’s commencement speech, and I had sprinkled a few jokes in there. Afterward, multiple people approached me and said, “You should really consider stand-up comedy. Your timing and delivery are great.” A few months later, I started my intern year in Los Angeles and simultaneously took stand-up comedy classes. I started performing at local clubs around town throughout residency, and I had two or three good sets that I could rely on. And so that’s how I got into stand-up comedy.

My intern year is also when I started this social media satire account. It was a way to cope with the anxieties and stress of residency. Before I knew it, the account gained multitudes of followers, doctors, and other medical professionals. And I joke that the more hours I work in a week, the more memes I make, the more posts I make. It’s kind of a creative outlet for me after a long day.
 

Q: What types of things do you talk about during your stand-up act?

Dr. Haghighat: A lot of it is about growing up in an immigrant household as a first-generation Iranian American. One of my favorite jokes is, my parents gave me so many options for a career. They said I could be a family doctor, a surgeon, a plastic surgeon, and if I worked hard, even a wife of a surgeon. But I talk a lot about being a woman in medicine. That always gets a lot of laughs. And now that I’ve graduated GI fellowship, I’m excited to incorporate some GI jokes because it turns out people love poop jokes.

 

 

Lightning Round

Texting or talking?

Text

Favorite city in U.S. besides the one you live in?

Denver

Cat or dog person

Dog

Best place you went on vacation

Patagonia

Favorite sport

Basketball

Favorite ice cream

Rocky Road

What song do you have to sing along with when you hear it?

Celine Dion’s My Heart Will Go On

Growing up in a household where GI issues dominated conversations, it’s no surprise that Shida Haghighat, MD, chose gastroenterology as her area of study in medicine.

She watched her father suffer from the complications of Crohn’s disease and her brother struggle with irritable bowel syndrome. “We always needed to know where the nearest bathroom was. I grew up with that around me, and I was always just fascinated by the gut and the digestive system,” said Dr. Haghighat, who just finished up her fellowship at the University of Miami and is now a gastroenterologist at University of California, Los Angeles. She also serves as social media editor for AGA’s Gastro Hep Advances.

Haghighat_Shida_CA_web.jpg
Dr. Shida Haghighat

As she got to know the personalities of the GI department in the first year of medical school, “I realized that our senses of humor and personalities kind of aligned, and I was like, ‘Oh yeah, this is where I’m supposed to be,’ ” said Dr. Haghighat, who can be found on X @DoctorShida.

Humor is something Dr. Haghighat has reached for throughout her life and career. She eventually channeled her gift for satire onto the stage and the internet, as a stand-up comedian. In an interview with GI & Hepatology News, she spoke about the connection between GI medicine and humor, and the creative ways she has helped promote cancer screening in underserved populations.
 

Q: What practice challenges have you faced in your career?

Dr. Haghighat: I trained in a county hospital, so I’ve always worked with underserved and vulnerable populations. One of the challenges has been just navigation of care, especially as it pertains to cancer diagnoses or cancer screening. A lot of the time, patients don’t understand why they have to do a test or something invasive like a colonoscopy for symptoms they don’t have. 

Q: A focus of yours has been improving uptake of screening in underserved communities. Please talk about the work you’ve done in this area.

Dr. Haghighat: I was at Los Angeles General Medical Center — a county hospital in Los Angeles — for residency, where we treated underserved, uninsured patients. I noticed in our primary care clinics a very low uptake of colon cancer screening. Patients didn’t want to bring the stool tests back or get colonoscopies. I surveyed a bunch of the patients and asked: How can we make colon cancer screening easier for you? About a third of the patients said, “If I can do it in the clinic before I go home, that would be great.”

So, I started this initiative called “Go Before You Go.” We would ask patients, “Hey, do you need to go to the bathroom right now, if you can?” Our nurses handed them the stool test to do in the bathroom before they left the clinic after their doctor visits.

We saw really good results with that. Surprisingly, a lot of people can go on demand. We saw increased screening rates, and that quality improvement project went on to win multiple first place awards in research competitions. So that’s what got me interested, and that’s where I had my beginnings of increasing preventative services in underserved communities on the ground.
 

 

 

Q: Can you discuss some health disparity studies you’ve done in this area?

Dr. Haghighat: As a GI at Jackson Memorial Hospital in Miami, I was seeing cancer disparities firsthand every day. I wanted to approach these disparities from a research funding standpoint on a federal level. I was particularly interested in gastric cancer because it’s not common enough in the United States to warrant universal screening, but it’s very common among certain racial and ethnic minorities, which would warrant targeted screening.

I evaluated cancer funding allocation from the National Cancer Institute among the most common cancers in the United States and found that cancer afflicting a higher proportion of racial and ethnic minorities was receiving lower funding. One of those cancers was stomach cancer. This study basically highlighted that, to decrease these disparities, a top-down policy approach is necessary to distribute cancer research funding equitably across these groups. 

A lot of stomach cancer comes from a bacteria called Helicobacter pylori, which can be more prevalent in certain countries. In another study, I looked at country of birth as a risk factor for stomach cancer, specifically for gastric intestinal metaplasia, which is a precursor for gastric cancer . 

We found that country of birth is a key risk factor for gastric intestinal metaplasia and that it should be incorporated into risk stratification for targeted screening. 
 

Q: Outside of medicine, you perform as a stand-up comedian. You have a popular satirical alias on social media. How did you get interested in stand-up comedy?

Dr. Haghighat: I gave my medical school’s commencement speech, and I had sprinkled a few jokes in there. Afterward, multiple people approached me and said, “You should really consider stand-up comedy. Your timing and delivery are great.” A few months later, I started my intern year in Los Angeles and simultaneously took stand-up comedy classes. I started performing at local clubs around town throughout residency, and I had two or three good sets that I could rely on. And so that’s how I got into stand-up comedy.

My intern year is also when I started this social media satire account. It was a way to cope with the anxieties and stress of residency. Before I knew it, the account gained multitudes of followers, doctors, and other medical professionals. And I joke that the more hours I work in a week, the more memes I make, the more posts I make. It’s kind of a creative outlet for me after a long day.
 

Q: What types of things do you talk about during your stand-up act?

Dr. Haghighat: A lot of it is about growing up in an immigrant household as a first-generation Iranian American. One of my favorite jokes is, my parents gave me so many options for a career. They said I could be a family doctor, a surgeon, a plastic surgeon, and if I worked hard, even a wife of a surgeon. But I talk a lot about being a woman in medicine. That always gets a lot of laughs. And now that I’ve graduated GI fellowship, I’m excited to incorporate some GI jokes because it turns out people love poop jokes.

 

 

Lightning Round

Texting or talking?

Text

Favorite city in U.S. besides the one you live in?

Denver

Cat or dog person

Dog

Best place you went on vacation

Patagonia

Favorite sport

Basketball

Favorite ice cream

Rocky Road

What song do you have to sing along with when you hear it?

Celine Dion’s My Heart Will Go On

Publications
Publications
Topics
Article Type
Display Headline
GI Physician Channels Humor to Incentivize Cancer Screenings
Display Headline
GI Physician Channels Humor to Incentivize Cancer Screenings
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>167044</fileName> <TBEID>0C04EA8E.SIG</TBEID> <TBUniqueIdentifier>MD_0C04EA8E</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>GI Hep April Member Spotlight</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240311T122643</QCDate> <firstPublished>20240311T123115</firstPublished> <LastPublished>20240311T123115</LastPublished> <pubStatus qcode="stat:"/> <embargoDate>20240401T070000</embargoDate> <killDate/> <CMSDate>20240401T070000</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Jennifer Lubell</byline> <bylineText>JENNIFER LUBELL</bylineText> <bylineFull>JENNIFER LUBELL</bylineFull> <bylineTitleText>MDedge News</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType>Feature</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>Growing up in a household where GI issues dominated conversations, it’s no surprise that Shida Haghighat, MD, chose gastroenterology as her area of study in med</metaDescription> <articlePDF/> <teaserImage>300651</teaserImage> <teaser>Humor is something Dr. Shida Haghighat has reached for throughout her life and career.</teaser> <title>GI physician channels humor to incentivize cancer screenings</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">73428</term> <term>39313</term> </sections> <topics> <term>344</term> <term canonical="true">28399</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401270f.jpg</altRep> <description role="drol:caption">Dr. Shida Haghighat</description> <description role="drol:credit">University of Miami</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>GI physician channels humor to incentivize cancer screenings</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Growing up in a household where GI issues dominated conversations, it’s no surprise that Shida Haghighat, MD, chose gastroenterology as her area of study in medicine.</span> </p> <p>She watched her father suffer from the complications of Crohn’s disease and her brother struggle with irritable bowel syndrome. “We always needed to know where the nearest bathroom was. I grew up with that around me, and I was always just fascinated by the gut and the digestive system,” said Dr. Haghighat, who just finished up her fellowship at the University of Miami and is now a gastroenterologist at University of California, Los Angeles. She also serves as social media editor for AGA’s <em>Gastro Hep Advances</em>.<br/><br/>[[{"fid":"300651","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"gastroenterologist at University of California, Los Angeles","field_file_image_credit[und][0][value]":"University of Miami","field_file_image_caption[und][0][value]":"Dr. Shida Haghighat"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]As she got to know the personalities of the GI department in the first year of medical school, “I realized that our senses of humor and personalities kind of aligned, and I was like, ‘Oh yeah, this is where I’m supposed to be,’ ” said Dr. Haghighat, who can be found on X <span class="Hyperlink"><a href="https://twitter.com/doctorshida">@DoctorShida</a></span>.<br/><br/>Humor is something Dr. Haghighat has reached for throughout her life and career. She eventually channeled her gift for satire onto the stage and the internet, as a stand-up comedian. In an interview with <em>GI &amp; Hepatology News</em>, she spoke about the connection between GI medicine and humor, and the creative ways she has helped promote cancer screening in underserved populations. <br/><br/></p> <h2>Q: What practice challenges have you faced in your career?</h2> <p>Dr. Haghighat: I trained in a county hospital, so I’ve always worked with underserved and vulnerable populations. One of the challenges has been just navigation of care, especially as it pertains to cancer diagnoses or cancer screening. A lot of the time, patients don’t understand why they have to do a test or something invasive like a colonoscopy for symptoms they don’t have. </p> <h2>Q: A focus of yours has been improving uptake of screening in underserved communities. Please talk about the work you’ve done in this area. </h2> <p>Dr. Haghighat: I was at Los Angeles General Medical Center — a county hospital in Los Angeles — for residency, where we treated underserved, uninsured patients. I noticed in our primary care clinics a very low uptake of colon cancer screening. Patients didn’t want to bring the stool tests back or get colonoscopies. I surveyed a bunch of the patients and asked: How can we make colon cancer screening easier for you? About a third of the patients said, “If I can do it in the clinic before I go home, that would be great.”</p> <p>So, I started this initiative called “Go Before You Go.” We would ask patients, “Hey, do you need to go to the bathroom right now, if you can?” Our nurses handed them the stool test to do in the bathroom before they left the clinic after their doctor visits. <br/><br/>We saw really good results with that. Surprisingly, a lot of people can go on demand. We saw increased screening rates, and that quality improvement project went on to win multiple first place awards in research competitions. So that’s what got me interested, and that’s where I had my beginnings of increasing preventative services in underserved communities on the ground. <br/><br/></p> <h2>Q: Can you discuss some health disparity studies you’ve done in this area?</h2> <p>Dr. Haghighat: As a GI at Jackson Memorial Hospital in Miami, I was seeing cancer disparities firsthand every day. I wanted to approach these disparities from a research funding standpoint on a federal level. I was particularly interested in gastric cancer because it’s not common enough in the United States to warrant universal screening, but it’s very common among certain racial and ethnic minorities, which would warrant targeted screening. </p> <p>I <span class="Hyperlink"><a href="https://www.researchgate.net/publication/371406488_Urgent_Need_to_Mitigate_Disparities_in_Federal_Funding_for_Cancer_Research">evaluated cancer funding allocation</a></span> from the National Cancer Institute among the most common cancers in the United States and found that cancer afflicting a higher proportion of racial and ethnic minorities was receiving lower funding. One of those cancers was stomach cancer. This study basically highlighted that, to decrease these disparities, a top-down policy approach is necessary to distribute cancer research funding equitably across these groups. <br/><br/>A lot of stomach cancer comes from a bacteria called <em>Helicobacter pylori</em>, which can be more prevalent in certain countries. <span class="Hyperlink"><a href="https://www.researchgate.net/publication/376129170_Association_between_country_of_birth_and_gastric_intestinal_metaplasia_a_retrospective_cohort_study">In another study</a></span>, I looked at country of birth as a risk factor for stomach cancer, specifically for gastric intestinal metaplasia, which is a precursor for gastric cancer . <br/><br/>We found that country of birth is a key risk factor for gastric intestinal metaplasia and that it should be incorporated into risk stratification for targeted screening. <br/><br/></p> <h2>Q: Outside of medicine, you perform as a stand-up comedian. You have a popular satirical alias on social media. How did you get interested in stand-up comedy?</h2> <p>Dr. Haghighat: I gave my medical school’s commencement speech, and I had sprinkled a few jokes in there. Afterward, multiple people approached me and said, “You should really consider stand-up comedy. Your timing and delivery are great.” A few months later, I started my intern year in Los Angeles and simultaneously took stand-up comedy classes. I started performing at local clubs around town throughout residency, and I had two or three good sets that I could rely on. And so that’s how I got into stand-up comedy. </p> <p>My intern year is also when I started this social media satire account. It was a way to cope with the anxieties and stress of residency. Before I knew it, the account gained multitudes of followers, doctors, and other medical professionals. And I joke that the more hours I work in a week, the more memes I make, the more posts I make. It’s kind of a creative outlet for me after a long day. <br/><br/></p> <h2>Q: What types of things do you talk about during your stand-up act?</h2> <p>Dr. Haghighat: A lot of it is about growing up in an immigrant household as a first-generation Iranian American. One of my favorite jokes is, my parents gave me so many options for a career. They said I could be a family doctor, a surgeon, a plastic surgeon, and if I worked hard, even a wife of a surgeon. But I talk a lot about being a woman in medicine. That always gets a lot of laughs. And now that I’ve graduated GI fellowship, I’m excited to incorporate some GI jokes because it turns out people love poop jokes.<span class="end"/></p> <h2>Lightning Round</h2> <p><strong>Texting or talking?</strong><br/><br/>Text</p> <p><strong>Favorite city in U.S. besides the one you live in?</strong><br/><br/>Denver</p> <p><strong>Cat or dog person</strong><br/><br/>Dog</p> <p><strong>Best place you went on vacation</strong><br/><br/>Patagonia </p> <p><strong>Favorite sport</strong><br/><br/>Basketball</p> <p><strong>Favorite ice cream</strong><br/><br/>Rocky Road</p> <p><strong>What song do you have to sing along with when you hear it?</strong><br/><br/>Celine Dion’s My Heart Will Go On</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

Liquid Biopsy for Colorectal Cancer Appears Promising But Still Lacks Robust Efficacy

Article Type
Changed
Wed, 03/27/2024 - 10:04

Blood-based screening for colorectal cancer (CRC), also known as a “liquid biopsy,” may be better than nothing among patients who skip established screening tests, but it can’t replace colonoscopy as the gold standard, according to two new modeling studies and an expert consensus commentary.

Although some patients find blood-based tests more convenient, the higher numbers of false positives and false negatives could lead to more CRC cases and deaths.

“Based on their current characteristics, blood tests should not be recommended to replace established colorectal cancer screening tests, since blood tests are neither as effective nor cost-effective and would worsen outcomes,” David Lieberman, MD, AGAF, chair of the American Gastroenterological Association’s CRC Workshop Panel, and lead author of the expert commentary, said in a statement.

Lieberman_David_Ore_web.jpg
Dr. David Lieberman

The blood tests detect circulating nucleotides, such as cell-free DNA or metabolic products associated with CRC and its precursors. Current tests are in development by Guardant Health and Freenome.

The two modeling studies, published in Gastroenterology on March 26, analyzed the effectiveness and cost-effectiveness of blood-based CRC screening that meets Centers for Medicare & Medicaid Services (CMS) coverage criteria, as well as the comparative effectiveness and cost-effectiveness of CRC screening with blood-based biomarkers versus fecal tests or colonoscopy.

Also published on March 26 in Clinical Gastroenterology and Hepatology, the expert commentary included key conclusions from the AGA CRC Workshop, which analyzed the two modeling studies.
 

Comparing CRC Screening Methods

In the first modeling study, an international team of researchers ran three microsimulation models for CRC to estimate the effectiveness and cost-effectiveness of triennial blood-based screening for ages 45-75, compared with no screening, annual fecal immunochemical testing (FIT), triennial stool DNA testing combined with a FIT assay, and colonoscopy screening every 10 years. The researchers used CMS coverage criteria for blood tests, with a sensitivity of at least 74% for detection of CRC and specificity of at least 90%.

Without screening, the models predicted between 77 and 88 CRC cases and between 32 and 36 deaths per 1,000 individuals, costing between $5.3 million to $5.8 million. Compared with no screening, blood-based screening was considered cost-effective, with an additional cost of $25,600 to $43,700 per quality-adjusted life-year gained (QALYG).

However, compared with the FIT, stool, and colonoscopy options, blood-based screening was not cost-effective, with both a decrease in QALYG and an increase in costs. FIT was more effective and less costly, with 5-24 QALYG and nearly $3.5 million cheaper than blood-based screening, even when blood-based uptake was 20 percentage points higher than FIT uptake.

In the second modeling study, US researchers compared triennial blood-based screening with established alternatives at the CMS thresholds of 74% sensitivity and 90% specificity.

Overall, a blood-based test at the CMS minimum reduced CRC incidence by 40% and CRC mortality by 52% versus no screening. However, a blood-based test was significantly less effective than triennial stool DNA testing, annual FIT, and colonoscopy every 10 years, which reduced CRC incidence by 68%-79% and CRC mortality by 73%-81%.

Assuming a blood-based test would cost the same as a multi-target stool test, the blood-based test would cost $28,500 per QALYG versus no screening. At the same time, FIT, colonoscopy, and stool DNA testing were less costly and more effective. In general, the blood-based test would match FIT’s clinical outcomes if it achieved 1.4- to 1.8-fold the participation rate for FIT.

Even still, the sensitivity for advanced precancerous lesion (APL) was a key determinant. A paradigm-changing blood-based test would need to have higher than 90% sensitivity for CRC and 80% for APL, 90% specificity, and cost less than $120 to $140, the study authors wrote.

“High APL sensitivity, which can result in CRC prevention, should be a top priority for screening test developers,” the authors wrote. “APL detection should not be penalized by a definition of test specificity that focuses on CRC only.”
 

 

 

Additional Considerations

The AGA CRC Workshop Panel met in September 2023 to review the two modeling studies and other data on blood-based tests for CRC. Overall, the group concluded that a triennial blood test that meets minimal CMS criteria would likely result in better outcomes than no screening and provide a simple process to encourage more people to participate in screening.

However, patients who may have declined colonoscopy should understand the need for a colonoscopy if blood-based tests show abnormal results, the commentary authors wrote.

In addition, because blood-based tests for CRC appear to be less effective and more costly than current screening options, they shouldn’t be recommended to replace established screening methods. Although these blood-based tests may improve screening rates and outcomes in unscreened people, substituting blood tests for other effective tests would increase costs and worsen patient outcomes.

Beyond that, they wrote, the industry should consider other potential benchmarks for an effective blood test, such as a sensitivity for stage I-III CRC of greater than 90% and sensitivity for advanced adenomas of 40%-50% or higher.

Carethers_John_CA.jpg
Dr. John M. Carethers

“Unless we have the expectation of high sensitivity and specificity, blood-based colorectal cancer tests could lead to false positive and false negative results, which are both bad for patient outcomes,” John M. Carethers, MD, AGAF, vice chancellor for health sciences at UC San Diego, AGA past president, and a member of the AGA CRC Workshop panel, said in a statement.

Several authors reported consultant roles and funding support from numerous companies, including Guardant Health and Freenome.

Publications
Topics
Sections

Blood-based screening for colorectal cancer (CRC), also known as a “liquid biopsy,” may be better than nothing among patients who skip established screening tests, but it can’t replace colonoscopy as the gold standard, according to two new modeling studies and an expert consensus commentary.

Although some patients find blood-based tests more convenient, the higher numbers of false positives and false negatives could lead to more CRC cases and deaths.

“Based on their current characteristics, blood tests should not be recommended to replace established colorectal cancer screening tests, since blood tests are neither as effective nor cost-effective and would worsen outcomes,” David Lieberman, MD, AGAF, chair of the American Gastroenterological Association’s CRC Workshop Panel, and lead author of the expert commentary, said in a statement.

Lieberman_David_Ore_web.jpg
Dr. David Lieberman

The blood tests detect circulating nucleotides, such as cell-free DNA or metabolic products associated with CRC and its precursors. Current tests are in development by Guardant Health and Freenome.

The two modeling studies, published in Gastroenterology on March 26, analyzed the effectiveness and cost-effectiveness of blood-based CRC screening that meets Centers for Medicare & Medicaid Services (CMS) coverage criteria, as well as the comparative effectiveness and cost-effectiveness of CRC screening with blood-based biomarkers versus fecal tests or colonoscopy.

Also published on March 26 in Clinical Gastroenterology and Hepatology, the expert commentary included key conclusions from the AGA CRC Workshop, which analyzed the two modeling studies.
 

Comparing CRC Screening Methods

In the first modeling study, an international team of researchers ran three microsimulation models for CRC to estimate the effectiveness and cost-effectiveness of triennial blood-based screening for ages 45-75, compared with no screening, annual fecal immunochemical testing (FIT), triennial stool DNA testing combined with a FIT assay, and colonoscopy screening every 10 years. The researchers used CMS coverage criteria for blood tests, with a sensitivity of at least 74% for detection of CRC and specificity of at least 90%.

Without screening, the models predicted between 77 and 88 CRC cases and between 32 and 36 deaths per 1,000 individuals, costing between $5.3 million to $5.8 million. Compared with no screening, blood-based screening was considered cost-effective, with an additional cost of $25,600 to $43,700 per quality-adjusted life-year gained (QALYG).

However, compared with the FIT, stool, and colonoscopy options, blood-based screening was not cost-effective, with both a decrease in QALYG and an increase in costs. FIT was more effective and less costly, with 5-24 QALYG and nearly $3.5 million cheaper than blood-based screening, even when blood-based uptake was 20 percentage points higher than FIT uptake.

In the second modeling study, US researchers compared triennial blood-based screening with established alternatives at the CMS thresholds of 74% sensitivity and 90% specificity.

Overall, a blood-based test at the CMS minimum reduced CRC incidence by 40% and CRC mortality by 52% versus no screening. However, a blood-based test was significantly less effective than triennial stool DNA testing, annual FIT, and colonoscopy every 10 years, which reduced CRC incidence by 68%-79% and CRC mortality by 73%-81%.

Assuming a blood-based test would cost the same as a multi-target stool test, the blood-based test would cost $28,500 per QALYG versus no screening. At the same time, FIT, colonoscopy, and stool DNA testing were less costly and more effective. In general, the blood-based test would match FIT’s clinical outcomes if it achieved 1.4- to 1.8-fold the participation rate for FIT.

Even still, the sensitivity for advanced precancerous lesion (APL) was a key determinant. A paradigm-changing blood-based test would need to have higher than 90% sensitivity for CRC and 80% for APL, 90% specificity, and cost less than $120 to $140, the study authors wrote.

“High APL sensitivity, which can result in CRC prevention, should be a top priority for screening test developers,” the authors wrote. “APL detection should not be penalized by a definition of test specificity that focuses on CRC only.”
 

 

 

Additional Considerations

The AGA CRC Workshop Panel met in September 2023 to review the two modeling studies and other data on blood-based tests for CRC. Overall, the group concluded that a triennial blood test that meets minimal CMS criteria would likely result in better outcomes than no screening and provide a simple process to encourage more people to participate in screening.

However, patients who may have declined colonoscopy should understand the need for a colonoscopy if blood-based tests show abnormal results, the commentary authors wrote.

In addition, because blood-based tests for CRC appear to be less effective and more costly than current screening options, they shouldn’t be recommended to replace established screening methods. Although these blood-based tests may improve screening rates and outcomes in unscreened people, substituting blood tests for other effective tests would increase costs and worsen patient outcomes.

Beyond that, they wrote, the industry should consider other potential benchmarks for an effective blood test, such as a sensitivity for stage I-III CRC of greater than 90% and sensitivity for advanced adenomas of 40%-50% or higher.

Carethers_John_CA.jpg
Dr. John M. Carethers

“Unless we have the expectation of high sensitivity and specificity, blood-based colorectal cancer tests could lead to false positive and false negative results, which are both bad for patient outcomes,” John M. Carethers, MD, AGAF, vice chancellor for health sciences at UC San Diego, AGA past president, and a member of the AGA CRC Workshop panel, said in a statement.

Several authors reported consultant roles and funding support from numerous companies, including Guardant Health and Freenome.

Blood-based screening for colorectal cancer (CRC), also known as a “liquid biopsy,” may be better than nothing among patients who skip established screening tests, but it can’t replace colonoscopy as the gold standard, according to two new modeling studies and an expert consensus commentary.

Although some patients find blood-based tests more convenient, the higher numbers of false positives and false negatives could lead to more CRC cases and deaths.

“Based on their current characteristics, blood tests should not be recommended to replace established colorectal cancer screening tests, since blood tests are neither as effective nor cost-effective and would worsen outcomes,” David Lieberman, MD, AGAF, chair of the American Gastroenterological Association’s CRC Workshop Panel, and lead author of the expert commentary, said in a statement.

Lieberman_David_Ore_web.jpg
Dr. David Lieberman

The blood tests detect circulating nucleotides, such as cell-free DNA or metabolic products associated with CRC and its precursors. Current tests are in development by Guardant Health and Freenome.

The two modeling studies, published in Gastroenterology on March 26, analyzed the effectiveness and cost-effectiveness of blood-based CRC screening that meets Centers for Medicare & Medicaid Services (CMS) coverage criteria, as well as the comparative effectiveness and cost-effectiveness of CRC screening with blood-based biomarkers versus fecal tests or colonoscopy.

Also published on March 26 in Clinical Gastroenterology and Hepatology, the expert commentary included key conclusions from the AGA CRC Workshop, which analyzed the two modeling studies.
 

Comparing CRC Screening Methods

In the first modeling study, an international team of researchers ran three microsimulation models for CRC to estimate the effectiveness and cost-effectiveness of triennial blood-based screening for ages 45-75, compared with no screening, annual fecal immunochemical testing (FIT), triennial stool DNA testing combined with a FIT assay, and colonoscopy screening every 10 years. The researchers used CMS coverage criteria for blood tests, with a sensitivity of at least 74% for detection of CRC and specificity of at least 90%.

Without screening, the models predicted between 77 and 88 CRC cases and between 32 and 36 deaths per 1,000 individuals, costing between $5.3 million to $5.8 million. Compared with no screening, blood-based screening was considered cost-effective, with an additional cost of $25,600 to $43,700 per quality-adjusted life-year gained (QALYG).

However, compared with the FIT, stool, and colonoscopy options, blood-based screening was not cost-effective, with both a decrease in QALYG and an increase in costs. FIT was more effective and less costly, with 5-24 QALYG and nearly $3.5 million cheaper than blood-based screening, even when blood-based uptake was 20 percentage points higher than FIT uptake.

In the second modeling study, US researchers compared triennial blood-based screening with established alternatives at the CMS thresholds of 74% sensitivity and 90% specificity.

Overall, a blood-based test at the CMS minimum reduced CRC incidence by 40% and CRC mortality by 52% versus no screening. However, a blood-based test was significantly less effective than triennial stool DNA testing, annual FIT, and colonoscopy every 10 years, which reduced CRC incidence by 68%-79% and CRC mortality by 73%-81%.

Assuming a blood-based test would cost the same as a multi-target stool test, the blood-based test would cost $28,500 per QALYG versus no screening. At the same time, FIT, colonoscopy, and stool DNA testing were less costly and more effective. In general, the blood-based test would match FIT’s clinical outcomes if it achieved 1.4- to 1.8-fold the participation rate for FIT.

Even still, the sensitivity for advanced precancerous lesion (APL) was a key determinant. A paradigm-changing blood-based test would need to have higher than 90% sensitivity for CRC and 80% for APL, 90% specificity, and cost less than $120 to $140, the study authors wrote.

“High APL sensitivity, which can result in CRC prevention, should be a top priority for screening test developers,” the authors wrote. “APL detection should not be penalized by a definition of test specificity that focuses on CRC only.”
 

 

 

Additional Considerations

The AGA CRC Workshop Panel met in September 2023 to review the two modeling studies and other data on blood-based tests for CRC. Overall, the group concluded that a triennial blood test that meets minimal CMS criteria would likely result in better outcomes than no screening and provide a simple process to encourage more people to participate in screening.

However, patients who may have declined colonoscopy should understand the need for a colonoscopy if blood-based tests show abnormal results, the commentary authors wrote.

In addition, because blood-based tests for CRC appear to be less effective and more costly than current screening options, they shouldn’t be recommended to replace established screening methods. Although these blood-based tests may improve screening rates and outcomes in unscreened people, substituting blood tests for other effective tests would increase costs and worsen patient outcomes.

Beyond that, they wrote, the industry should consider other potential benchmarks for an effective blood test, such as a sensitivity for stage I-III CRC of greater than 90% and sensitivity for advanced adenomas of 40%-50% or higher.

Carethers_John_CA.jpg
Dr. John M. Carethers

“Unless we have the expectation of high sensitivity and specificity, blood-based colorectal cancer tests could lead to false positive and false negative results, which are both bad for patient outcomes,” John M. Carethers, MD, AGAF, vice chancellor for health sciences at UC San Diego, AGA past president, and a member of the AGA CRC Workshop panel, said in a statement.

Several authors reported consultant roles and funding support from numerous companies, including Guardant Health and Freenome.

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>167468</fileName> <TBEID>0C04F43D.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F43D</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240327T092454</QCDate> <firstPublished>20240327T094059</firstPublished> <LastPublished>20240327T094059</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240327T094059</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Carolyn Crist</byline> <bylineText>CAROLYN CRIST</bylineText> <bylineFull>CAROLYN CRIST</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>Blood-based screening for colorectal cancer (CRC), also known as a “liquid biopsy,” may be better than nothing among patients who skip established screening tes</metaDescription> <articlePDF/> <teaserImage>252253</teaserImage> <teaser>Patients who may have declined colonoscopy should understand the need for a colonoscopy if blood-based tests show abnormal results.</teaser> <title>Liquid Biopsy for Colorectal Cancer Appears Promising But Still Lacks Robust Efficacy</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>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> <term>15</term> <term>21</term> <term>31</term> </publications> <sections> <term canonical="true">69</term> <term>27970</term> <term>39313</term> </sections> <topics> <term canonical="true">344</term> <term>345</term> <term>213</term> <term>263</term> <term>67020</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2400cda7.jpg</altRep> <description role="drol:caption">Dr. David Lieberman</description> <description role="drol:credit">AGA Institute</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/24011d12.jpg</altRep> <description role="drol:caption">Dr. John M. Carethers</description> <description role="drol:credit">University of California San Diego</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Liquid Biopsy for Colorectal Cancer Appears Promising But Still Lacks Robust Efficacy</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">Blood-based screening for colorectal cancer (CRC), also known as a “liquid biopsy,” may be better than nothing among patients who skip established screening tests, but it can’t replace colonoscopy as the gold standard</span>, according to two new modeling studies and an expert consensus commentary.</p> <p>Although some patients find blood-based tests more convenient, the higher numbers of false positives and false negatives could lead to more CRC cases and deaths.<br/><br/>“Based on their current characteristics, blood tests should not be recommended to replace established colorectal cancer screening tests, since blood tests are neither as effective nor cost-effective and would worsen outcomes,” David Lieberman, MD, AGAF, chair of the American Gastroenterological Association’s CRC Workshop Panel, and lead author of the expert commentary, said in a statement.<br/><br/>[[{"fid":"252253","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Dr. David Lieberman, Oregon Health and Science University, Portland","field_file_image_credit[und][0][value]":"AGA Institute","field_file_image_caption[und][0][value]":"Dr. David Lieberman"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]The blood tests detect circulating nucleotides, such as cell-free DNA or metabolic products associated with CRC and its precursors. Current tests are in development by Guardant Health and Freenome.<br/><br/>The two modeling studies, published in <em>Gastroenterology</em> on March 26, analyzed <span class="Hyperlink"><a href="https://www.gastrojournal.org/article/S0016-5085(24)00174-4/fulltext">the effectiveness and cost-effectiveness</a></span> of blood-based CRC screening that meets Centers for Medicare &amp; Medicaid Services (CMS) coverage criteria, as well as the <span class="Hyperlink"><a href="https://www.gastrojournal.org/article/S0016-5085(24)00293-2/abstract">comparative effectiveness and cost-effectiveness</a></span> of CRC screening with blood-based biomarkers versus fecal tests or colonoscopy. <br/><br/>Also published on March 26 in <em>Clinical Gastroenterology and Hepatology</em>, <span class="Hyperlink"><a href="https://www.cghjournal.org/article/S1542-3565(24)00162-9/abstract">the expert commentary</a></span> included key conclusions from the AGA CRC Workshop, which analyzed the two modeling studies.<br/><br/></p> <h2>Comparing CRC Screening Methods</h2> <p>In the first modeling study, an international team of researchers ran three microsimulation models for CRC to estimate the effectiveness and cost-effectiveness of triennial blood-based screening for ages 45-75, compared with no screening, annual fecal immunochemical testing (FIT), triennial stool DNA testing combined with a FIT assay, and colonoscopy screening every 10 years. The researchers used CMS coverage criteria for blood tests, with a sensitivity of at least 74% for detection of CRC and specificity of at least 90%.</p> <p>Without screening, the models predicted between 77 and 88 CRC cases and between 32 and 36 deaths per 1,000 individuals, costing between $5.3 million to $5.8 million. Compared with no screening, blood-based screening was considered cost-effective, with an additional cost of $25,600 to $43,700 per quality-adjusted life-year gained (QALYG).<br/><br/>However, compared with the FIT, stool, and colonoscopy options, blood-based screening was not cost-effective, with both a decrease in QALYG and an increase in costs. FIT was more effective and less costly, with 5-24 QALYG and nearly $3.5 million cheaper than blood-based screening, even when blood-based uptake was 20 percentage points higher than FIT uptake.<br/><br/>In the second modeling study, US researchers compared triennial blood-based screening with established alternatives at the CMS thresholds of 74% sensitivity and 90% specificity. <br/><br/>Overall, a blood-based test at the CMS minimum reduced CRC incidence by 40% and CRC mortality by 52% versus no screening. However, a blood-based test was significantly less effective than triennial stool DNA testing, annual FIT, and colonoscopy every 10 years, which reduced CRC incidence by 68%-79% and CRC mortality by 73%-81%.<br/><br/>Assuming a blood-based test would cost the same as a multi-target stool test, the blood-based test would cost $28,500 per QALYG versus no screening. At the same time, FIT, colonoscopy, and stool DNA testing were less costly and more effective. In general, the blood-based test would match FIT’s clinical outcomes if it achieved 1.4- to 1.8-fold the participation rate for FIT.<br/><br/>Even still, the sensitivity for advanced precancerous lesion (APL) was a key determinant. A paradigm-changing blood-based test would need to have higher than 90% sensitivity for CRC and 80% for APL, 90% specificity, and cost less than $120 to $140, the study authors wrote.<br/><br/>“High APL sensitivity, which can result in CRC prevention, should be a top priority for screening test developers,” the authors wrote. “APL detection should not be penalized by a definition of test specificity that focuses on CRC only.”<br/><br/></p> <h2>Additional Considerations</h2> <p>The AGA CRC Workshop Panel met in September 2023 to review the two modeling studies and other data on blood-based tests for CRC. Overall, the group concluded that a triennial blood test that meets minimal CMS criteria would likely result in better outcomes than no screening and provide a simple process to encourage more people to participate in screening.</p> <p>However, patients who may have declined colonoscopy should understand the need for a colonoscopy if blood-based tests show abnormal results, the commentary authors wrote.<br/><br/>In addition, because blood-based tests for CRC appear to be less effective and more costly than current screening options, they shouldn’t be recommended to replace established screening methods. Although these blood-based tests may improve screening rates and outcomes in unscreened people, substituting blood tests for other effective tests would increase costs and worsen patient outcomes.<br/><br/>Beyond that, they wrote, the industry should consider other potential benchmarks for an effective blood test, such as a sensitivity for stage I-III CRC of greater than 90% and sensitivity for advanced adenomas of 40%-50% or higher.<br/><br/>[[{"fid":"294809","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Dr. John M. Carethers, distinguished professor of medicine and vice chancellor for health sciences at the University of California, San Diego; AGA president John M. Carethers, MD, AGAF","field_file_image_credit[und][0][value]":"University of California San Diego","field_file_image_caption[und][0][value]":"Dr. John M. Carethers"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]“Unless we have the expectation of high sensitivity and specificity, blood-based colorectal cancer tests could lead to false positive and false negative results, which are both bad for patient outcomes,” John M. Carethers, MD, AGAF, vice chancellor for health sciences at UC San Diego, AGA past president, and a member of the AGA CRC Workshop panel, said in a statement.<br/><br/>Several authors reported consultant roles and funding support from numerous companies, including Guardant Health and Freenome.<span class="end"/></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

New CRC stool test beats FIT for sensitivity but not specificity

Article Type
Changed
Mon, 03/25/2024 - 09:15

A next-generation stool DNA test for colorectal cancer (CRC) screening had higher sensitivity for all screening-relevant lesions but lower specificity than a currently available fecal immunochemical test (FIT), according to the large prospective BLUE-C study.

The multi-target assay by Exact Sciences Corporation, the makers of Cologuard, includes new biomarkers designed to increase specificity without decreasing sensitivity. It showed a sensitivity for CRC of almost 94%, with more than 43% sensitivity for advanced precancerous lesions and nearly 91% specificity for advanced neoplasia, according to the study results, which were published in The New England Journal of Medicine.

Imperiale_Thomas_IN_web.jpg
Dr. Thomas F. Imperiale

Adherence to CRC screening in the United States is well below the 80% national target, and the quest continues for noninvasive screening assays that might improve screening adherence, noted lead author Thomas F. Imperiale, MD, AGAF, a professor of medicine at Indiana University School of medicine in Indianapolis, and colleagues.

“The test’s manufacturer developed a new version of its existing Cologuard FIT/DNA test because it took to heart the feedback from primary care providers and gastroenterologists about the test’s low specificity,” Dr. Imperiale said in an interview. “The goal of the new test was to improve specificity without losing, and perhaps even gaining, some sensitivity — a goal that is not easily accomplished when you’re trying to improve on a sensitivity for colorectal cancer that was already 92.3% in the current version of Cologuard.”

Compared with the earlier version of Cologuard, he added, the new generation retained sensitivity for CRC and advanced precancerous lesions or polyps while improving specificity by 30% (90.6% vs 86.6%) for advanced neoplasia — a combination of CRC and advanced precancerous lesions, he said. “This with the caveat, however, that the two versions were not compared head-to-head in this new study,” Dr. Imperiale said.

The higher specificity for advanced lesions is expected to translate to a lower false positive rate. Lowering false positive rates is crucial because that reduces the need for costly, invasive, and unnecessary colonoscopies, said Aasma Shaukat, MD, MPH, AGAF, director of outcomes research in NYU Langone Health’s division of gastroenterology and hepatology in New York City.

Shaukat_Aasma_NY_web.jpg
Dr. Aasma Shaukat

“Many physicians felt there were too many false positives with the existing version, and that is anxiety-provoking in patients and providers,” said Dr. Shaukat, who was not involved in the study.

In her view, however, the test’s moderate improvements in detecting certain lesions does not make it demonstrably superior to its predecessor, and there is always the possibility of higher cost to consider.

While acknowledging that a higher sensitivity for all advanced precancerous lesions would have been welcome, Dr. Imperiale said the test detected 75% of the most worrisome of such lesions — “the ones containing high-grade dysplastic cells and suggesting near-term conversion to cancer. And its ability to detect other advanced lesions improved as the size of the lesions increased.”
 

 

 

Testing details

Almost 21,000 asymptomatic participants age 40 years and older undergoing screening colonoscopy were evaluated at 186 US sites during the period 2019 to 2023. Of the cohort, 98 had CRC, 2144 had advanced precancerous lesions, 6973 had nonadvanced adenomas, and 10,961 had nonneoplastic findings or negative colonoscopy.

Advanced precancerous lesions included one or more adenomas or sessile serrated lesions measuring at least 1 cm in the longest dimension, lesions with villous histologic features, and high-grade dysplasia. The new DNA test identified 92 of 98 participants with CRC and 76 of 82 participants with screening-relevant cancers. Among the findings for the new assay:

  • Sensitivity for any-stage CRC was 93.9% (95% confidence interval [CI], 87.1- 97.7)
  • Sensitivity for advanced precancerous lesions was 43.4% (95% CI, 41.3-45.6)
  • Sensitivity for high-grade dysplasia was 74.6% (95% CI, 65.6-82.3)
  • Specificity for advanced neoplasia was 90.6% (95% CI, 90.1- 91.0).
  • Specificity for nonneoplastic findings or negative colonoscopy was 92.7% (95% CI, 92.2-93.1)
  • Specificity for negative colonoscopy was 93.3 (95% CI, 92.8-93.9)
  • No adverse events occurred.

In the comparator assay, OC-AUTO FIT by Polymedco, sensitivity was 67.3% (95% CI, 57.1-76.5) for CRC, 23.3% (95% CI, 21.5-25.2) for advanced precancerous lesions, and 47.4% (95% CI, 37.9-56.9) for high-grade dysplasia. In the comparator FIT, however, specificity was better across all age groups — at 94.8% (95% CI, 94.4-95.1) for advanced neoplasia, 95.7% (95% CI, 95.3- 96.1) for nonneoplastic findings, and 96.0% (95% CI, 95.5-96.4) for negative colonoscopy.

In another article in the same issue of NEJM, Guardant Health’s cell-free DNA blood-based test had 83% sensitivity for CRC, 90% specificity for advanced neoplasia, and 13% sensitivity for advanced precancerous lesions in an average-risk population.

An age-related decrease in specificity was observed with the new Cologuard test, but that did not concern Dr. Imperiale because the same observation was made with the current version. “In fact, the next-gen version appears to have less of an age-related decrease in specificity than the current version, although, again, the two versions were not tested head-to-head,” he noted.

The effect of age-related background methylation of DNA is well known, he explained. “Clinicians and older patients in the screening age range do need to be aware of this effect on specificity before ordering or agreeing to do the test. I do not see this as a stumbling block to implementation, but it does require discussion between patient and ordering provider.”

The new version of the DNA test is expected to be available in about a year.

According to Dr. Imperiale, further research is needed to ascertain the test’s acceptability and adherence rates and to quantify its yield in population-based screening. Determining its cost-effectiveness and making it easier to use are other goals. “And most importantly, the degree of reduction in the incidence and mortality from colorectal cancer,” he said.

Cost-effectiveness and the selection of the testing interval may play roles in adherence, particularly in populations with lower rates of screening adherence than the general population, John M. Carethers, MD, AGAF, of the University of California, San Diego, noted in a related editorial.

“Adherence to screening varies according to age group, including persons in the 45- to 49-year age group who are now eligible for average-risk screening,” he wrote. “It is hoped that these newer tests will increase use and adherence and elevate the percentage of the population undergoing screening in order to reduce deaths from colorectal cancer.”

This study was sponsored by Exact Sciences Corporation, which conducted the stool testing at its laboratories.

Dr. Imperiale had no competing interests to disclose. Several study co-authors reported employment with Exact Sciences, or stock and intellectual property ownership. Dr. Shaukat disclosed consulting for Freenome. Dr. Carethers reported ties to Avantor Inc. and Geneoscopy.

Publications
Topics
Sections

A next-generation stool DNA test for colorectal cancer (CRC) screening had higher sensitivity for all screening-relevant lesions but lower specificity than a currently available fecal immunochemical test (FIT), according to the large prospective BLUE-C study.

The multi-target assay by Exact Sciences Corporation, the makers of Cologuard, includes new biomarkers designed to increase specificity without decreasing sensitivity. It showed a sensitivity for CRC of almost 94%, with more than 43% sensitivity for advanced precancerous lesions and nearly 91% specificity for advanced neoplasia, according to the study results, which were published in The New England Journal of Medicine.

Imperiale_Thomas_IN_web.jpg
Dr. Thomas F. Imperiale

Adherence to CRC screening in the United States is well below the 80% national target, and the quest continues for noninvasive screening assays that might improve screening adherence, noted lead author Thomas F. Imperiale, MD, AGAF, a professor of medicine at Indiana University School of medicine in Indianapolis, and colleagues.

“The test’s manufacturer developed a new version of its existing Cologuard FIT/DNA test because it took to heart the feedback from primary care providers and gastroenterologists about the test’s low specificity,” Dr. Imperiale said in an interview. “The goal of the new test was to improve specificity without losing, and perhaps even gaining, some sensitivity — a goal that is not easily accomplished when you’re trying to improve on a sensitivity for colorectal cancer that was already 92.3% in the current version of Cologuard.”

Compared with the earlier version of Cologuard, he added, the new generation retained sensitivity for CRC and advanced precancerous lesions or polyps while improving specificity by 30% (90.6% vs 86.6%) for advanced neoplasia — a combination of CRC and advanced precancerous lesions, he said. “This with the caveat, however, that the two versions were not compared head-to-head in this new study,” Dr. Imperiale said.

The higher specificity for advanced lesions is expected to translate to a lower false positive rate. Lowering false positive rates is crucial because that reduces the need for costly, invasive, and unnecessary colonoscopies, said Aasma Shaukat, MD, MPH, AGAF, director of outcomes research in NYU Langone Health’s division of gastroenterology and hepatology in New York City.

Shaukat_Aasma_NY_web.jpg
Dr. Aasma Shaukat

“Many physicians felt there were too many false positives with the existing version, and that is anxiety-provoking in patients and providers,” said Dr. Shaukat, who was not involved in the study.

In her view, however, the test’s moderate improvements in detecting certain lesions does not make it demonstrably superior to its predecessor, and there is always the possibility of higher cost to consider.

While acknowledging that a higher sensitivity for all advanced precancerous lesions would have been welcome, Dr. Imperiale said the test detected 75% of the most worrisome of such lesions — “the ones containing high-grade dysplastic cells and suggesting near-term conversion to cancer. And its ability to detect other advanced lesions improved as the size of the lesions increased.”
 

 

 

Testing details

Almost 21,000 asymptomatic participants age 40 years and older undergoing screening colonoscopy were evaluated at 186 US sites during the period 2019 to 2023. Of the cohort, 98 had CRC, 2144 had advanced precancerous lesions, 6973 had nonadvanced adenomas, and 10,961 had nonneoplastic findings or negative colonoscopy.

Advanced precancerous lesions included one or more adenomas or sessile serrated lesions measuring at least 1 cm in the longest dimension, lesions with villous histologic features, and high-grade dysplasia. The new DNA test identified 92 of 98 participants with CRC and 76 of 82 participants with screening-relevant cancers. Among the findings for the new assay:

  • Sensitivity for any-stage CRC was 93.9% (95% confidence interval [CI], 87.1- 97.7)
  • Sensitivity for advanced precancerous lesions was 43.4% (95% CI, 41.3-45.6)
  • Sensitivity for high-grade dysplasia was 74.6% (95% CI, 65.6-82.3)
  • Specificity for advanced neoplasia was 90.6% (95% CI, 90.1- 91.0).
  • Specificity for nonneoplastic findings or negative colonoscopy was 92.7% (95% CI, 92.2-93.1)
  • Specificity for negative colonoscopy was 93.3 (95% CI, 92.8-93.9)
  • No adverse events occurred.

In the comparator assay, OC-AUTO FIT by Polymedco, sensitivity was 67.3% (95% CI, 57.1-76.5) for CRC, 23.3% (95% CI, 21.5-25.2) for advanced precancerous lesions, and 47.4% (95% CI, 37.9-56.9) for high-grade dysplasia. In the comparator FIT, however, specificity was better across all age groups — at 94.8% (95% CI, 94.4-95.1) for advanced neoplasia, 95.7% (95% CI, 95.3- 96.1) for nonneoplastic findings, and 96.0% (95% CI, 95.5-96.4) for negative colonoscopy.

In another article in the same issue of NEJM, Guardant Health’s cell-free DNA blood-based test had 83% sensitivity for CRC, 90% specificity for advanced neoplasia, and 13% sensitivity for advanced precancerous lesions in an average-risk population.

An age-related decrease in specificity was observed with the new Cologuard test, but that did not concern Dr. Imperiale because the same observation was made with the current version. “In fact, the next-gen version appears to have less of an age-related decrease in specificity than the current version, although, again, the two versions were not tested head-to-head,” he noted.

The effect of age-related background methylation of DNA is well known, he explained. “Clinicians and older patients in the screening age range do need to be aware of this effect on specificity before ordering or agreeing to do the test. I do not see this as a stumbling block to implementation, but it does require discussion between patient and ordering provider.”

The new version of the DNA test is expected to be available in about a year.

According to Dr. Imperiale, further research is needed to ascertain the test’s acceptability and adherence rates and to quantify its yield in population-based screening. Determining its cost-effectiveness and making it easier to use are other goals. “And most importantly, the degree of reduction in the incidence and mortality from colorectal cancer,” he said.

Cost-effectiveness and the selection of the testing interval may play roles in adherence, particularly in populations with lower rates of screening adherence than the general population, John M. Carethers, MD, AGAF, of the University of California, San Diego, noted in a related editorial.

“Adherence to screening varies according to age group, including persons in the 45- to 49-year age group who are now eligible for average-risk screening,” he wrote. “It is hoped that these newer tests will increase use and adherence and elevate the percentage of the population undergoing screening in order to reduce deaths from colorectal cancer.”

This study was sponsored by Exact Sciences Corporation, which conducted the stool testing at its laboratories.

Dr. Imperiale had no competing interests to disclose. Several study co-authors reported employment with Exact Sciences, or stock and intellectual property ownership. Dr. Shaukat disclosed consulting for Freenome. Dr. Carethers reported ties to Avantor Inc. and Geneoscopy.

A next-generation stool DNA test for colorectal cancer (CRC) screening had higher sensitivity for all screening-relevant lesions but lower specificity than a currently available fecal immunochemical test (FIT), according to the large prospective BLUE-C study.

The multi-target assay by Exact Sciences Corporation, the makers of Cologuard, includes new biomarkers designed to increase specificity without decreasing sensitivity. It showed a sensitivity for CRC of almost 94%, with more than 43% sensitivity for advanced precancerous lesions and nearly 91% specificity for advanced neoplasia, according to the study results, which were published in The New England Journal of Medicine.

Imperiale_Thomas_IN_web.jpg
Dr. Thomas F. Imperiale

Adherence to CRC screening in the United States is well below the 80% national target, and the quest continues for noninvasive screening assays that might improve screening adherence, noted lead author Thomas F. Imperiale, MD, AGAF, a professor of medicine at Indiana University School of medicine in Indianapolis, and colleagues.

“The test’s manufacturer developed a new version of its existing Cologuard FIT/DNA test because it took to heart the feedback from primary care providers and gastroenterologists about the test’s low specificity,” Dr. Imperiale said in an interview. “The goal of the new test was to improve specificity without losing, and perhaps even gaining, some sensitivity — a goal that is not easily accomplished when you’re trying to improve on a sensitivity for colorectal cancer that was already 92.3% in the current version of Cologuard.”

Compared with the earlier version of Cologuard, he added, the new generation retained sensitivity for CRC and advanced precancerous lesions or polyps while improving specificity by 30% (90.6% vs 86.6%) for advanced neoplasia — a combination of CRC and advanced precancerous lesions, he said. “This with the caveat, however, that the two versions were not compared head-to-head in this new study,” Dr. Imperiale said.

The higher specificity for advanced lesions is expected to translate to a lower false positive rate. Lowering false positive rates is crucial because that reduces the need for costly, invasive, and unnecessary colonoscopies, said Aasma Shaukat, MD, MPH, AGAF, director of outcomes research in NYU Langone Health’s division of gastroenterology and hepatology in New York City.

Shaukat_Aasma_NY_web.jpg
Dr. Aasma Shaukat

“Many physicians felt there were too many false positives with the existing version, and that is anxiety-provoking in patients and providers,” said Dr. Shaukat, who was not involved in the study.

In her view, however, the test’s moderate improvements in detecting certain lesions does not make it demonstrably superior to its predecessor, and there is always the possibility of higher cost to consider.

While acknowledging that a higher sensitivity for all advanced precancerous lesions would have been welcome, Dr. Imperiale said the test detected 75% of the most worrisome of such lesions — “the ones containing high-grade dysplastic cells and suggesting near-term conversion to cancer. And its ability to detect other advanced lesions improved as the size of the lesions increased.”
 

 

 

Testing details

Almost 21,000 asymptomatic participants age 40 years and older undergoing screening colonoscopy were evaluated at 186 US sites during the period 2019 to 2023. Of the cohort, 98 had CRC, 2144 had advanced precancerous lesions, 6973 had nonadvanced adenomas, and 10,961 had nonneoplastic findings or negative colonoscopy.

Advanced precancerous lesions included one or more adenomas or sessile serrated lesions measuring at least 1 cm in the longest dimension, lesions with villous histologic features, and high-grade dysplasia. The new DNA test identified 92 of 98 participants with CRC and 76 of 82 participants with screening-relevant cancers. Among the findings for the new assay:

  • Sensitivity for any-stage CRC was 93.9% (95% confidence interval [CI], 87.1- 97.7)
  • Sensitivity for advanced precancerous lesions was 43.4% (95% CI, 41.3-45.6)
  • Sensitivity for high-grade dysplasia was 74.6% (95% CI, 65.6-82.3)
  • Specificity for advanced neoplasia was 90.6% (95% CI, 90.1- 91.0).
  • Specificity for nonneoplastic findings or negative colonoscopy was 92.7% (95% CI, 92.2-93.1)
  • Specificity for negative colonoscopy was 93.3 (95% CI, 92.8-93.9)
  • No adverse events occurred.

In the comparator assay, OC-AUTO FIT by Polymedco, sensitivity was 67.3% (95% CI, 57.1-76.5) for CRC, 23.3% (95% CI, 21.5-25.2) for advanced precancerous lesions, and 47.4% (95% CI, 37.9-56.9) for high-grade dysplasia. In the comparator FIT, however, specificity was better across all age groups — at 94.8% (95% CI, 94.4-95.1) for advanced neoplasia, 95.7% (95% CI, 95.3- 96.1) for nonneoplastic findings, and 96.0% (95% CI, 95.5-96.4) for negative colonoscopy.

In another article in the same issue of NEJM, Guardant Health’s cell-free DNA blood-based test had 83% sensitivity for CRC, 90% specificity for advanced neoplasia, and 13% sensitivity for advanced precancerous lesions in an average-risk population.

An age-related decrease in specificity was observed with the new Cologuard test, but that did not concern Dr. Imperiale because the same observation was made with the current version. “In fact, the next-gen version appears to have less of an age-related decrease in specificity than the current version, although, again, the two versions were not tested head-to-head,” he noted.

The effect of age-related background methylation of DNA is well known, he explained. “Clinicians and older patients in the screening age range do need to be aware of this effect on specificity before ordering or agreeing to do the test. I do not see this as a stumbling block to implementation, but it does require discussion between patient and ordering provider.”

The new version of the DNA test is expected to be available in about a year.

According to Dr. Imperiale, further research is needed to ascertain the test’s acceptability and adherence rates and to quantify its yield in population-based screening. Determining its cost-effectiveness and making it easier to use are other goals. “And most importantly, the degree of reduction in the incidence and mortality from colorectal cancer,” he said.

Cost-effectiveness and the selection of the testing interval may play roles in adherence, particularly in populations with lower rates of screening adherence than the general population, John M. Carethers, MD, AGAF, of the University of California, San Diego, noted in a related editorial.

“Adherence to screening varies according to age group, including persons in the 45- to 49-year age group who are now eligible for average-risk screening,” he wrote. “It is hoped that these newer tests will increase use and adherence and elevate the percentage of the population undergoing screening in order to reduce deaths from colorectal cancer.”

This study was sponsored by Exact Sciences Corporation, which conducted the stool testing at its laboratories.

Dr. Imperiale had no competing interests to disclose. Several study co-authors reported employment with Exact Sciences, or stock and intellectual property ownership. Dr. Shaukat disclosed consulting for Freenome. Dr. Carethers reported ties to Avantor Inc. and Geneoscopy.

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>167365</fileName> <TBEID>0C04F0F9.SIG</TBEID> <TBUniqueIdentifier>MD_0C04F0F9</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname>Next-gen DNA stool test for CRC</storyname> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240321T160553</QCDate> <firstPublished>20240321T160740</firstPublished> <LastPublished>20240321T160741</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240321T160740</CMSDate> <articleSource>FROM NEW ENGLAND JOURNAL OF MEDICINE</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 next-generation stool DNA test for colorectal cancer (CRC) screening had higher sensitivity for all screening-relevant lesions but lower specificity than a cu</metaDescription> <articlePDF/> <teaserImage>300843</teaserImage> <teaser>The higher specificity for advanced lesions is expected to translate to a lower false positive rate.</teaser> <title>New CRC stool test beats FIT for sensitivity but not specificity</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>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">17</term> <term>15</term> <term>21</term> <term>31</term> </publications> <sections> <term canonical="true">27970</term> <term>39313</term> </sections> <topics> <term canonical="true">344</term> <term>213</term> <term>263</term> <term>67020</term> <term>280</term> </topics> <links> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/2401277b.jpg</altRep> <description role="drol:caption">Dr. Thomas F. Imperiale</description> <description role="drol:credit">Indiana University School of Medicine</description> </link> <link> <itemClass qcode="ninat:picture"/> <altRep contenttype="image/jpeg">images/240105b0.jpg</altRep> <description role="drol:caption">Dr. Aasma Shaukat</description> <description role="drol:credit">New York University</description> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>New CRC stool test beats FIT for sensitivity but not specificity</title> <deck/> </itemMeta> <itemContent> <p><span class="tag metaDescription">A next-generation stool DNA test for colorectal cancer (CRC) screening had higher sensitivity for all screening-relevant lesions but lower specificity than a currently available fecal immunochemical test (FIT)</span>, according to the large prospective BLUE-C study.</p> <p>The multi-target assay by Exact Sciences Corporation, the makers of Cologuard, includes new biomarkers designed to increase specificity without decreasing sensitivity. It showed a sensitivity for CRC of almost 94%, with more than 43% sensitivity for advanced precancerous lesions and nearly 91% specificity for advanced neoplasia, according to the study results, which <span class="Hyperlink"><a href="https://www.nejm.org/doi/full/10.1056/NEJMoa2310336">were published in</a></span> <em>The New England Journal of Medicine</em> .<br/><br/>[[{"fid":"300843","view_mode":"medstat_image_flush_right","fields":{"format":"medstat_image_flush_right","field_file_image_alt_text[und][0][value]":"Indiana University School of Medicine in Indianapolis","field_file_image_credit[und][0][value]":"Indiana University School of Medicine","field_file_image_caption[und][0][value]":"Dr. Thomas F. Imperiale"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_right"}}]]Adherence to CRC screening in the United States is well below the 80% national target, and the quest continues for noninvasive screening assays that might improve screening adherence, noted lead author Thomas F. Imperiale, MD, AGAF, a professor of medicine at Indiana University School of medicine in Indianapolis, and colleagues.<br/><br/>“The test’s manufacturer developed a new version of its existing Cologuard FIT/DNA test because it took to heart the feedback from primary care providers and gastroenterologists about the test’s low specificity,” Dr. Imperiale said in an interview. “The goal of the new test was to improve specificity without losing, and perhaps even gaining, some sensitivity — a goal that is not easily accomplished when you’re trying to improve on a sensitivity for colorectal cancer <span class="Hyperlink"><a href="https://www.nejm.org/doi/full/10.1056/nejmoa1311194">that was already 92.3%</a></span> in the current version of Cologuard.”<br/><br/>Compared with the earlier version of Cologuard, he added, the new generation retained sensitivity for CRC and advanced precancerous lesions or polyps while improving specificity by 30% (90.6% vs 86.6%) for advanced neoplasia — a combination of CRC and advanced precancerous lesions, he said. “This with the caveat, however, that the two versions were not compared head-to-head in this new study,” Dr. Imperiale said.<br/><br/>The higher specificity for advanced lesions is expected to translate to a lower false positive rate. Lowering false positive rates is crucial because that reduces the need for costly, invasive, and unnecessary colonoscopies, said Aasma Shaukat, MD, MPH, AGAF, director of outcomes research in NYU Langone Health’s division of gastroenterology and hepatology in New York City. <br/><br/>[[{"fid":"283023","view_mode":"medstat_image_flush_left","fields":{"format":"medstat_image_flush_left","field_file_image_alt_text[und][0][value]":"Aasma Shaukat, MD MPH AGAF, is Robert M. and Mary H. Glickman Professor of Medicine and Popula-tion Health and director of GI Outcomes Research at New York University.","field_file_image_credit[und][0][value]":"New York University","field_file_image_caption[und][0][value]":"Dr. Aasma Shaukat"},"type":"media","attributes":{"class":"media-element file-medstat_image_flush_left"}}]]“Many physicians felt there were too many false positives with the existing version, and that is anxiety-provoking in patients and providers,” said Dr. Shaukat, who was not involved in the study.<br/><br/>In her view, however, the test’s moderate improvements in detecting certain lesions does not make it demonstrably superior to its predecessor, and there is always the possibility of higher cost to consider.<br/><br/>While acknowledging that a higher sensitivity for all advanced precancerous lesions would have been welcome, Dr. Imperiale said the test detected 75% of the most worrisome of such lesions — “the ones containing high-grade dysplastic cells and suggesting near-term conversion to cancer. And its ability to detect other advanced lesions improved as the size of the lesions increased.” <br/><br/> </p> <h2>Testing details </h2> <p>Almost 21,000 asymptomatic participants age 40 years and older undergoing screening colonoscopy were evaluated at 186 US sites during the period 2019 to 2023. Of the cohort, 98 had CRC, 2144 had advanced precancerous lesions, 6973 had nonadvanced adenomas, and 10,961 had nonneoplastic findings or negative colonoscopy.</p> <p>Advanced precancerous lesions included one or more adenomas or sessile serrated lesions measuring at least 1 cm in the longest dimension, lesions with villous histologic features, and high-grade dysplasia. The new DNA test identified 92 of 98 participants with CRC and 76 of 82 participants with screening-relevant cancers. Among the findings for the new assay:</p> <ul class="body"> <li>Sensitivity for any-stage CRC was 93.9% (95% confidence interval [CI], 87.1- 97.7)</li> <li>Sensitivity for advanced precancerous lesions was 43.4% (95% CI, 41.3-45.6)</li> <li>Sensitivity for high-grade dysplasia was 74.6% (95% CI, 65.6-82.3)</li> <li>Specificity for advanced neoplasia was 90.6% (95% CI, 90.1- 91.0).</li> <li>Specificity for nonneoplastic findings or negative colonoscopy was 92.7% (95% CI, 92.2-93.1)</li> <li>Specificity for negative colonoscopy was 93.3 (95% CI, 92.8-93.9)</li> <li>No adverse events occurred.</li> </ul> <p>In the comparator assay, OC-AUTO FIT by Polymedco, sensitivity was 67.3% (95% CI, 57.1-76.5) for CRC, 23.3% (95% CI, 21.5-25.2) for advanced precancerous lesions, and 47.4% (95% CI, 37.9-56.9) for high-grade dysplasia. In the comparator FIT, however, specificity was better across all age groups — at 94.8% (95% CI, 94.4-95.1) for advanced neoplasia, 95.7% (95% CI, 95.3- 96.1) for nonneoplastic findings, and 96.0% (95% CI, 95.5-96.4) for negative colonoscopy.<br/><br/>In another <span class="Hyperlink"><a href="https://www.nejm.org/doi/full/10.1056/NEJMoa2304714">article in the same issue</a></span> of <em>NEJM</em>, Guardant Health’s cell-free DNA blood-based test had 83% sensitivity for CRC, 90% specificity for advanced neoplasia, and 13% sensitivity for advanced precancerous lesions in an average-risk population .<br/><br/>An age-related decrease in specificity was observed with the new Cologuard test, but that did not concern Dr. Imperiale because the same observation was made with the current version. “In fact, the next-gen version appears to have less of an age-related decrease in specificity than the current version, although, again, the two versions were not tested head-to-head,” he noted. <br/><br/>The effect of age-related background methylation of DNA is well known, he explained. “Clinicians and older patients in the screening age range do need to be aware of this effect on specificity before ordering or agreeing to do the test. I do not see this as a stumbling block to implementation, but it does require discussion between patient and ordering provider.”<br/><br/>The new version of the DNA test is expected to be available in about a year.<br/><br/>According to Dr. Imperiale, further research is needed to ascertain the test’s acceptability and adherence rates and to quantify its yield in population-based screening. Determining its cost-effectiveness and making it easier to use are other goals. “And most importantly, the degree of reduction in the incidence and mortality from colorectal cancer,” he said. <br/><br/>Cost-effectiveness and the selection of the testing interval may play roles in adherence, particularly in populations with lower rates of screening adherence than the general population, John M. Carethers, MD, AGAF, of the University of California, San Diego, noted in <span class="Hyperlink"><a href="https://www.nejm.org/doi/full/10.1056/NEJMe2400366?query=recirc_curatedRelated_article">a related editorial</a></span> . <br/><br/>“Adherence to screening varies according to age group, including persons in the 45- to 49-year age group who are now eligible for average-risk screening,” he wrote. “It is hoped that these newer tests will increase use and adherence and elevate the percentage of the population undergoing screening in order to reduce deaths from colorectal cancer.”<br/><br/>This study was sponsored by Exact Sciences Corporation, which conducted the stool testing at its laboratories.<br/><br/>Dr. Imperiale had no competing interests to disclose. Several study co-authors reported employment with Exact Sciences, or stock and intellectual property ownership. Dr. Shaukat disclosed consulting for Freenome. Dr. Carethers reported ties to Avantor Inc. and Geneoscopy.<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 NEW ENGLAND JOURNAL OF MEDICINE

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