Whether GLP-1 RAs Significantly Delay Gastric Emptying Called into Question

Article Type
Changed
Thu, 07/11/2024 - 10:26

 

TOPLINE:

Patients taking a glucagon-like peptide 1 receptor agonist (GLP-1 RA) experience only a modest delay in gastric emptying of solid foods and no significant delay for liquids, compared with those receiving placebo, indicating that patients may not need to discontinue these medications before surgery.

METHODOLOGY:

  • GLP-1 RAs, while effective in managing diabetes and obesity, are linked to delayed gastric emptying, which may pose risks during procedures requiring anesthesia or sedation due to potential aspiration of gastric contents.
  • Researchers conducted a meta-analysis to quantify the duration of delay in gastric emptying caused by GLP-1 RAs in patients with diabetes and/or excessive body weight, which could guide periprocedural management decisions in the future.
  • The primary outcome was halftime, the time required for 50% of solid gastric contents to empty, measured using scintigraphy. This analysis included data from five studies involving 247 patients who received either a GLP-1 RA or placebo.
  • The secondary outcome was gastric emptying of liquids measured using the acetaminophen absorption test. Ten studies including 411 patients who received either a GLP-1 RA or placebo were included in this analysis.

TAKEAWAY:

  • The mean gastric emptying halftime for solid foods was 138.4 minutes with a GLP-1 RA and 95.0 minutes with placebo, resulting in a pooled mean difference of 36.0 minutes (P < .01).
  • No significant difference was found in the gastric emptying time for liquids between the GLP-1 RA and placebo groups. Furthermore, the amount of gastric emptying noted at 4 or 5 hours on the acetaminophen absorption test was comparable between these groups.
  • The gastric emptying time for both solids and liquids did not differ between GLP-1 RA formulations or between short-acting or long-acting GLP-1 RAs.

IN PRACTICE:

“Based on current evidence, a conservative approach with a liquid diet on the day before procedures while continuing GLP-1 RA therapy would represent the most sensible approach until more conclusive data on a solid diet are available,” the authors wrote.

SOURCE:

The study, led by Brent Hiramoto, MD, MPH, of the Center for Gastrointestinal Motility at Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in The American Journal of Gastroenterology.

LIMITATIONS:

The small number of studies utilizing some diagnostic modalities, such as breath testing, precluded a formal meta-analysis of these subgroups. The results could not be stratified by indication for GLP-1 RA (diabetes or obesity) because of insufficient studies in each category.

DISCLOSURES:

The lead author was supported by the National Institute of Diabetes and Digestive and Kidney Diseases. One author declared serving on the advisory boards of three pharmaceutical companies.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

Patients taking a glucagon-like peptide 1 receptor agonist (GLP-1 RA) experience only a modest delay in gastric emptying of solid foods and no significant delay for liquids, compared with those receiving placebo, indicating that patients may not need to discontinue these medications before surgery.

METHODOLOGY:

  • GLP-1 RAs, while effective in managing diabetes and obesity, are linked to delayed gastric emptying, which may pose risks during procedures requiring anesthesia or sedation due to potential aspiration of gastric contents.
  • Researchers conducted a meta-analysis to quantify the duration of delay in gastric emptying caused by GLP-1 RAs in patients with diabetes and/or excessive body weight, which could guide periprocedural management decisions in the future.
  • The primary outcome was halftime, the time required for 50% of solid gastric contents to empty, measured using scintigraphy. This analysis included data from five studies involving 247 patients who received either a GLP-1 RA or placebo.
  • The secondary outcome was gastric emptying of liquids measured using the acetaminophen absorption test. Ten studies including 411 patients who received either a GLP-1 RA or placebo were included in this analysis.

TAKEAWAY:

  • The mean gastric emptying halftime for solid foods was 138.4 minutes with a GLP-1 RA and 95.0 minutes with placebo, resulting in a pooled mean difference of 36.0 minutes (P < .01).
  • No significant difference was found in the gastric emptying time for liquids between the GLP-1 RA and placebo groups. Furthermore, the amount of gastric emptying noted at 4 or 5 hours on the acetaminophen absorption test was comparable between these groups.
  • The gastric emptying time for both solids and liquids did not differ between GLP-1 RA formulations or between short-acting or long-acting GLP-1 RAs.

IN PRACTICE:

“Based on current evidence, a conservative approach with a liquid diet on the day before procedures while continuing GLP-1 RA therapy would represent the most sensible approach until more conclusive data on a solid diet are available,” the authors wrote.

SOURCE:

The study, led by Brent Hiramoto, MD, MPH, of the Center for Gastrointestinal Motility at Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in The American Journal of Gastroenterology.

LIMITATIONS:

The small number of studies utilizing some diagnostic modalities, such as breath testing, precluded a formal meta-analysis of these subgroups. The results could not be stratified by indication for GLP-1 RA (diabetes or obesity) because of insufficient studies in each category.

DISCLOSURES:

The lead author was supported by the National Institute of Diabetes and Digestive and Kidney Diseases. One author declared serving on the advisory boards of three pharmaceutical companies.

A version of this article first appeared on Medscape.com.

 

TOPLINE:

Patients taking a glucagon-like peptide 1 receptor agonist (GLP-1 RA) experience only a modest delay in gastric emptying of solid foods and no significant delay for liquids, compared with those receiving placebo, indicating that patients may not need to discontinue these medications before surgery.

METHODOLOGY:

  • GLP-1 RAs, while effective in managing diabetes and obesity, are linked to delayed gastric emptying, which may pose risks during procedures requiring anesthesia or sedation due to potential aspiration of gastric contents.
  • Researchers conducted a meta-analysis to quantify the duration of delay in gastric emptying caused by GLP-1 RAs in patients with diabetes and/or excessive body weight, which could guide periprocedural management decisions in the future.
  • The primary outcome was halftime, the time required for 50% of solid gastric contents to empty, measured using scintigraphy. This analysis included data from five studies involving 247 patients who received either a GLP-1 RA or placebo.
  • The secondary outcome was gastric emptying of liquids measured using the acetaminophen absorption test. Ten studies including 411 patients who received either a GLP-1 RA or placebo were included in this analysis.

TAKEAWAY:

  • The mean gastric emptying halftime for solid foods was 138.4 minutes with a GLP-1 RA and 95.0 minutes with placebo, resulting in a pooled mean difference of 36.0 minutes (P < .01).
  • No significant difference was found in the gastric emptying time for liquids between the GLP-1 RA and placebo groups. Furthermore, the amount of gastric emptying noted at 4 or 5 hours on the acetaminophen absorption test was comparable between these groups.
  • The gastric emptying time for both solids and liquids did not differ between GLP-1 RA formulations or between short-acting or long-acting GLP-1 RAs.

IN PRACTICE:

“Based on current evidence, a conservative approach with a liquid diet on the day before procedures while continuing GLP-1 RA therapy would represent the most sensible approach until more conclusive data on a solid diet are available,” the authors wrote.

SOURCE:

The study, led by Brent Hiramoto, MD, MPH, of the Center for Gastrointestinal Motility at Brigham and Women’s Hospital and Harvard Medical School, Boston, was published online in The American Journal of Gastroenterology.

LIMITATIONS:

The small number of studies utilizing some diagnostic modalities, such as breath testing, precluded a formal meta-analysis of these subgroups. The results could not be stratified by indication for GLP-1 RA (diabetes or obesity) because of insufficient studies in each category.

DISCLOSURES:

The lead author was supported by the National Institute of Diabetes and Digestive and Kidney Diseases. One author declared serving on the advisory boards of three pharmaceutical companies.

A version of this article first 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>168687</fileName> <TBEID>0C050EF6.SIG</TBEID> <TBUniqueIdentifier>MD_0C050EF6</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240710T132050</QCDate> <firstPublished>20240710T132224</firstPublished> <LastPublished>20240710T132224</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240710T132224</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>SHRABASTI BHATTACHARYA</byline> <bylineText>SHRABASTI BHATTACHARYA</bylineText> <bylineFull>SHRABASTI BHATTACHARYA</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>No significant difference was found in the gastric emptying time for liquids between the GLP-1 RA and placebo groups.</metaDescription> <articlePDF/> <teaserImage/> <teaser>Study finds that GLP-1 RAs do not slow gastric emptying significantly, removing need to stop meds before surgery.</teaser> <title>Whether GLP-1 RAs Significantly Delay Gastric Emptying Called into Question</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>mdsurg</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement>2018 Frontline Medical Communications Inc.,</copyrightStatement> </publicationData> <publicationData> <publicationCode>mdendo</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement>2018 Frontline Medical Communications Inc.,</copyrightStatement> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">52226</term> <term>51940</term> <term>15</term> <term>21</term> </publications> <sections> <term>27970</term> <term canonical="true">39313</term> </sections> <topics> <term canonical="true">351</term> <term>205</term> <term>261</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Whether GLP-1 RAs Significantly Delay Gastric Emptying Called into Question</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE:</h2> <p>Patients taking a glucagon-like peptide 1 receptor agonist (GLP-1 RA) experience only a modest delay in gastric emptying of solid foods and no significant delay for liquids, compared with those receiving placebo, indicating that patients may not need to discontinue these medications before surgery.</p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>GLP-1 RAs, while effective in managing diabetes and obesity, are linked to delayed gastric emptying, which may pose risks during procedures requiring anesthesia or sedation due to potential aspiration of gastric contents.</li> <li>Researchers conducted a meta-analysis to quantify the duration of delay in gastric emptying caused by GLP-1 RAs in patients with diabetes and/or excessive body weight, which could guide periprocedural management decisions in the future.</li> <li>The primary outcome was halftime, the time required for 50% of solid gastric contents to empty, measured using scintigraphy. This analysis included data from five studies involving 247 patients who received either a GLP-1 RA or placebo.</li> <li>The secondary outcome was gastric emptying of liquids measured using the acetaminophen absorption test. Ten studies including 411 patients who received either a GLP-1 RA or placebo were included in this analysis.</li> </ul> <h2>TAKEAWAY:</h2> <ul class="body"> <li>The mean gastric emptying halftime for solid foods was 138.4 minutes with a GLP-1 RA and 95.0 minutes with placebo, resulting in a pooled mean difference of 36.0 minutes (<em>P</em> &lt; .01).</li> <li><span class="tag metaDescription">No significant difference was found in the gastric emptying time for liquids between the GLP-1 RA and placebo groups.</span> Furthermore, the amount of gastric emptying noted at 4 or 5 hours on the acetaminophen absorption test was comparable between these groups.</li> <li>The gastric emptying time for both solids and liquids did not differ between GLP-1 RA formulations or between short-acting or long-acting GLP-1 RAs.</li> </ul> <h2>IN PRACTICE:</h2> <p>“Based on current evidence, a conservative approach with a liquid diet on the day before procedures while continuing GLP-1 RA therapy would represent the most sensible approach until more conclusive data on a solid diet are available,” the authors wrote.</p> <h2>SOURCE:</h2> <p>The study, led by Brent Hiramoto, MD, MPH, of the Center for Gastrointestinal Motility at Brigham and Women’s Hospital and Harvard Medical School, Boston, was <span class="Hyperlink"><a href="https://journals.lww.com/ajg/abstract/2024/06000/quantified_metrics_of_gastric_emptying_delay_by.26.aspx">published online</a></span> in <em>The American Journal of Gastroenterology</em>.</p> <h2>LIMITATIONS:</h2> <p>The small number of studies utilizing some diagnostic modalities, such as breath testing, precluded a formal meta-analysis of these subgroups. The results could not be stratified by indication for GLP-1 RA (diabetes or obesity) because of insufficient studies in each category.</p> <h2>DISCLOSURES:</h2> <p>The lead author was supported by the National Institute of Diabetes and Digestive and Kidney Diseases. One author declared serving on the advisory boards of three pharmaceutical companies.<span class="end"/></p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://medscape.com/viewarticle/do-glp-1-ras-significantly-delay-gastric-emptying-maybe-not-2024a1000cnk">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

Long-Term Assessment of Weight Loss Medications in a Veteran Population

Article Type
Changed
Wed, 07/10/2024 - 09:31

The Centers for Disease Control and Prevention (CDC) classifies individuals with a body mass index (BMI) of 25 to 29.9as overweight and those with a BMI > 30 as obese (obesity classes: I, BMI 30 to 34.9; II, BMI 35 to 39.9; and III, BMI ≥ 40).1 In 2011, the CDC estimated that 27.4% of adults in the United States were obese; less than a decade later, that number increased to 31.9%.1 In that same period, the percentage of adults in Indiana classified as obese increased from 30.8% to 36.8%.1 About 1 in 14 individuals in the US have class III obesity and 86% of veterans are either overweight or obese.2

High medical expenses can likely be attributed to the long-term health consequences of obesity. Compared to those with a healthy weight, individuals who are overweight or obese are at an increased risk for high blood pressure, high low-density lipoprotein cholesterol levels, low high-density lipoprotein cholesterol levels, high triglyceride levels, type 2 diabetes mellitus (T2DM), coronary heart disease, stroke, gallbladder disease, osteoarthritis, sleep apnea, cancer, mental health disorders, body pain, low quality of life, and death.3 Many of these conditions lead to increased health care needs, medication needs, hospitalizations, and overall health care system use.

Guidelines for the prevention and treatment of obesity have been produced by the American Heart Association, American College of Cardiology, and The Obesity Society; the Endocrine Society; the American Diabetes Association; and the US Departments of Veterans Affairs (VA) and Defense. Each follows a general algorithm to manage and prevent adverse effects (AEs) related to obesity. General practice is to assess a patient for elevated BMI (> 25), implement intense lifestyle modifications including calorie restriction and exercise, reassess for a maintained 5% to 10% weight loss for cardiovascular benefits, and potentially assess for pharmacological or surgical intervention to assist in weight loss.2,4-6

While some weight loss medications (eg, phentermine/topiramate, naltrexone/bupropion, orlistat, and lorcaserin) tend to have unfavorable AEs or mixed efficacy, glucagon-like peptide-1 receptor agonists (GLP-1RAs) have provided new options.7-10 Lorcaserin, for example, was removed from the market in 2020 due to its association with cancer risks.11 The GLP-1RAs liraglutide and semaglutide received US Food and Drug Administration (FDA) approval for weight loss in 2014 and 2021, respectively.12,13 GLP-1RAs have shown the greatest efficacy and benefits in reducing hemoglobin A1c (HbA1c); they are the preferred agents for patients who qualify for pharmacologic intervention for weight loss, especially those with T2DM. However, these studies have not evaluated the long-term outcomes of using these medications for weight loss and may not reflect the veteran population.14,15

 

At Veteran Health Indiana (VHI), clinicians may use several weight loss medications for patients to achieve 5% to 10% weight loss. The medications most often used include liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone. However, more research is needed to determine which weight loss medication is the most beneficial for veterans, particularly following FDA approval of GLP-1RAs. At VHI, phentermine/topiramate is the preferred first-line agent unless patients have contraindications for use, in which case naltrexone/bupropion is recommended. These are considered first-line due to their ease of use in pill form, lower cost, and comparable weight loss to the GLP-1 medication class.2 However, for patients with prediabetes, T2DM, BMI > 40, or BMI > 35 with specific comorbid conditions, liraglutide is preferred because of its beneficial effects for both weight loss and blood glucose control.2

This study aimed to expand on the 2021 Hood and colleagues study that examined total weight loss and weight loss as a percentage of baseline weight in patients with obesity at 3, 6, 12, and > 12 months of pharmacologic therapy by extending the time frame to 48 months.16 This study excluded semaglutide because few patients were prescribed the medication for weight loss during the study.

 

 

METHODS

We conducted a single-center, retrospective chart review of patients prescribed weight loss medications at VHI. A patient list was generated based on prescription fills from June 1, 2017, to July 31, 2021. Data were obtained from the Computerized Patient Record System; patients were not contacted. This study was approved by the Indiana University Health Institutional Review Board and VHI Research and Development Committee.

At the time of this study, liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone were available at VHI for patients who met the clinical criteria for use. All patients must have been enrolled in dietary and lifestyle management programs, including the VA MOVE! program, to be approved for these medications. After the MOVE! orientation, patients could participate in group or individual 12-week programs that included weigh-ins, goal-setting strategies, meal planning, and habit modification support. If patients could not meet in person, phone and other telehealth opportunities were available.

Patients were included in the study if they were aged ≥ 18 years, received a prescription for any of the 5 available medications for weight loss during the enrollment period, and were on the medication for ≥ 6 consecutive months. Patients were excluded if they received a prescription, were treated outside the VA system, or were pregnant. The primary indication for the included medication was not weight loss; the primary indication for the GLP-1RA was T2DM, or the weight loss was attributed to another disease. Adherence was not a measured outcome of this study; if patients were filling the medication, it was assumed they were taking it. Data were collected for each instance of medication use; as a result, a few patients were included more than once. Data collection for a failed medication ended when failure was documented. New data points began when new medication was prescribed; all data were per medication, not per patient. This allowed us to account for medication failure and provide accurate weight loss results based on medication choice within VHI.

Primary outcomes included total weight loss and weight loss as a percentage ofbaseline weight during the study period at 3, 6, 12, 24, 36, and 48 months of therapy. Secondary outcomes included the percentage of patients who lost 5% to 10% of their body weight from baseline; the percentage of patients who maintained ≥ 5% weight loss from baseline to 12, 24, 36, and 48 months if maintained on medication for that duration; duration of medication treatment in weeks; medication discontinuation rate; reason for medication discontinuation; enrollment in the MOVE! clinic and the time enrolled; percentage of patients with a BMI of 18 to 24.9 at the end of the study; and change in HbA1c at 3, 6, 12, 24, 36, and 48 months.

Demographic data included race, age, sex, baseline weight, height, baseline BMI, and comorbid conditions (collected based on the most recent primary care clinical note before initiating medication). Medication data collected included medications used to manage comorbidities. Data related to weight management medication included prescribing clinic, maintenance dose of medication, duration of medication during the study period, the reason for medication discontinuation, or bariatric surgery intervention if applicable.

 


Basic descriptive statistics were used to characterize study participants. For continuous data, analysis of variance tests were used; if those results were not normal, then nonparametric tests were used, followed by pairwise tests between medication groups if the overall test was significant using the Fisher significant differences test. For nominal data, χ2 or Fisher exact tests were used. For comparisons of primary and secondary outcomes, if the analyses needed to include adjustment for confounding variables, analysis of covariance was used for continuous data. A 2-sided 5% significance level was used for all tests.

 

 

RESULTS

ruwophabrogudanatukasachacobesteswilejesp

A total of 228 instances of medication use were identified based on prescription fills; 123 did not meet inclusion criteria (117 for < 6 consecutive months of medication use) (Figure). The study included 105 participants with a mean age of 56 years; 80 were male (76.2%), and 85 identified as White race (81.0%). Mean (SD) weight was 130.1 kg (26.8) and BMI was 41.6 (7.2). The most common comorbid disease states among patients included hypertension, dyslipidemia, obstructive sleep apnea, and T2DM (Table 1). The baseline characteristics were comparable to those of Hood and colleagues.16

sputhuphilatikuslepatavitribestifruseslulachatafrurutridusputrevecladoteslikokojufronocrisleclimishishacrecraclemareslodrimostoprapatraprebajafriveuocleswofruthanoswachemawristetotrapovubedruswacliswujewustespemamenupacluthoneuobraprisewecrac

Most patients at VHI started on liraglutide (63%) or phentermine/topiramate (28%). For primary and secondary outcomes, statistics were calculated to determine whether the results were statistically significant for comparing the liraglutide and phentermine/topiramate subgroups. Sample sizes were too small for statistical analysis for bupropion/naltrexone, phentermine, and orlistat.

Primary Outcomes

The mean (SD) weight of participants dropped 8.1% from 130.1 kg to 119.5 kg over the patient-specific duration of weight management medication therapy for an absolute difference of 10.6 kg (9.7). Duration of individual medication use varied from 6 to 48 months. Weight loss was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. Patient weight was not recorded after the medication was discontinued.

ralashaclulauiclelivespolabrubehesheclawroslucripratrophuchithutrigebeliminebruludrospegevukukubramaphistukuchichopiphifrirothogavubiwrushibuwarefrouotiphonipuhospispichaueslubobrebrathislitra

When classified by medication choice, the mean change in weight over the duration of the study was −23.9 kg for 2 patients using orlistat, −10.2 kg for 46 patients using liraglutide, −11.0 kg for 25 patients using phentermine/topiramate, -7.4 kg for 1 patient using phentermine, and -13.0 kg for 4 patients using naltrexone/bupropion. Patients without a weight documented at the end of their therapy or at the conclusion of the data collection period were not included in the total weight loss at the end of therapy. There were 78 documented instances of weight loss at the end of therapy (Table 2).

kiwistecrohecredribimothaslof

Body weight loss percentage was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. The mean (SD) body weight loss percentage over the duration of the study was 9.2% (11.2). When classified by medication choice, the mean percentage of body weight loss was 16.8% for 2 patients using orlistat, 9.4% for 46 patients using liraglutide, 8.2% for 25 patients using phentermine/topiramate, 6.0% for 1 patient using phentermine alone, and 10.6% for 4 patients using naltrexone/bupropion (Table 3).

Secondary Outcomes

While none of the secondary outcomes were statistically significant, the results of this study suggest that both medications may contribute to weight loss in many patients included in this study. Almost two-thirds of the included patients analyzed lost ≥ 5% of weight from baseline while taking weight management medication. Sixty-six patients (63%) lost ≥ 5% of body weight at any time during the data collection period. When stratified by liraglutide and phentermine/topiramate, 41 patients (63%) taking liraglutide and 20 patients (67%) taking phentermine/topiramate lost ≥ 5% of weight from baseline. Of the 66 patients who lost ≥ 5% of body weight from baseline, 36 (55%) lost ≥ 10% of body weight from baseline at any time during the data collection period.

vothouastinedruwaspuuuslithipruuostubrinicobrurusliherichatidocetecewrohekushasadrelufrobruuuwrasebreprucocedalisirejutubrevohiphedurinishefruwrostutrama

The mean (SD) duration for weight management medication use was 23 months (14.9). Phentermine/topiramate was tolerated longer than liraglutide: 22.7 months vs 21.7 months, respectively (Table 4).

 

The average overall documented medication discontinuation rate was 35.2%. Reasons for discontinuation included 21 patient-elected discontinuations, 8 patients no longer met criteria for use, 4 medications were no longer indicated, and 4 patients experienced AEs. It is unknown whether weight management medication was discontinued or not in 18 patients (17.2%).

 

 

DISCUSSION

This study evaluated the use and outcomes of weight loss medications over a longer period (up to 48 months) than what was previously studied among patients at VHI (12 months). The study aimed to better understand the long-term effect of weight loss medications, determine which medication had better long-term outcomes, and examine the reasons for medication discontinuation.

The results of this study displayed some similarities and differences compared with the Hood and colleagues study.16 Both yielded similar results for 5% of body weight loss and 10% of body weight loss. The largest difference was mean weight loss over the study period. In this study, patients lost a mean 10.6 kg over the course of weight loss medication use compared to 15.8 kg found by Hood and colleagues.16 A reason patients in the current study lost less weight overall could be the difference in time frames. The current study encompassed the COVID-19 pandemic, meaning fewer overall in-person patient appointments, which led to patients being lost to follow-up, missing weigh-ins during the time period, and gaps in care. For some patients, the pandemic possibly contributed to depression, missed medication doses, and a more sedentary lifestyle, leading to more weight gain.17 Telemedicine services at VHI expanded during the pandemic in an attempt to increase patient monitoring and counseling. It is unclear whether this expansion was enough to replace the in-person contact necessary to promote a healthy lifestyle.

VA pharmacists now care for patients through telehealth and are more involved in weight loss management. Since the conclusion of the Hood and colleagues study and start of this research, 2 pharmacists at VHI have been assigned to follow patients for obesity management to help with adherence to medication and lifestyle changes, management of AEs, dispense logistics, interventions for medications that may cause weight gain, and case management of glycemic control and weight loss with GLP-1RAs. Care management by pharmacists at VHI helps improve the logistics of titratable orders and save money by improving the use of high-cost items like GLP-1RAs. VA clinical pharmacy practitioners already monitor GLP-1RAs for patients with T2DM, so they are prepared to educate and assist patients with these medications.

It is important to continue developing a standardized process for weight loss medication management across the VA to improve the quality of patient care and optimize prescription outcomes. VA facilities differ in how weight loss management care is delivered and the level at which pharmacists are involved. Given the high rate of obesity among patients at the VA, the advent of new prescription options for weight loss, and the high cost associated with these medications, there has been increased attention to obesity care. Some Veterans Integrated Service Networks are forming a weight management community of practice groups to create standard operating procedures and algorithms to standardize care. Developing consistent processes is necessary to improve weight loss and patient care for veterans regardless where they receive treatment.

Limitations

The data used in this study were dependent on clinician documentation. Because of a lack of documentation in many instances, it was difficult to determine the full efficacy of the medications studied due to missing weight recordings. The lack of documentation made it difficult to determine whether patients were enrolled and active in the MOVE! program. It is required that patients enroll in MOVE! to obtain medications, but many did not have any follow-up MOVE! visits after initially obtaining their weight loss medication.

In this study, differences in the outcomes of patients with and without T2DM were not compared. It is the VA standard of care to prefer liraglutide over phentermine/topiramate in patients with T2DM or prediabetes.2 This makes it difficult to assess whether phentermine/topiramate or liraglutide is more effective for weight loss in patients with T2DM. Weight gain after the discontinuation of weight loss medications was not assessed. Collecting this data may help determine whether a certain weight loss medication is less likely to cause rebound weight gain when discontinued.

Other limitations to this study consisted of excluding patients who discontinued therapy within 6 months, small sample sizes on some medications, and lack of data on adherence. Adherence was based on medication refills, which means that if a patient refilled the medication, it was assumed they were taking it. This is not always the case, and while accurate data on adherence is difficult to gather, it can impact how results may be interpreted. These additional limitations make it difficult to accurately determine the efficacy of the medications in this study.

 

CONCLUSIONS

This study found similar outcomes to what has been observed in larger clinical trials regarding weight loss medications. Nevertheless, there was a lack of accurate clinical documentation for most patients, which limits the conclusions. This lack of documentation potentially led to inaccurate results. It revealed that many patients at VHI did not uniformly receive consistent follow-up after starting a weight loss medication during the study period. With more standardized processes implemented at VA facilities, increased pharmacist involvement in weight loss medication management, and increased use of established telehealth services, patients could have the opportunity for closer follow-up that may lead to better weight loss outcomes. With these changes, there is more reason for additional studies to be conducted to assess follow-up, medication management, and weight loss overall.

References

1. Overweight & obesity. Centers for Disease Control and Prevention. Updated September 21, 2023. Accessed April 23, 2024. https://www.cdc.gov/obesity/index.html

2. US Department of Defense, US Department of Veterans Affairs. The Management of Adult Overweight and Obesity Working Group. VA/DoD Clinical Practice Guideline for the Management of Adult Overweight and Obesity. Updated July 2020. Accessed April 23, 2024. https://www.healthquality.va.gov/guidelines/CD/obesity/VADoDObesityCPGFinal5087242020.pdf

3. Health effects of overweight and obesity. Centers for Disease Control and Prevention. Updated September 24, 2022. Accessed April 23, 2024. https://www.cdc.gov/healthyweight/effects/index.html

4. Jensen MD, Ryan DH, Apovian CM, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol. 2014;63(25 Pt B):2985-3023. doi:10.1016/j.jacc.2013.11.004

5. Apovian CM, Aronne LJ, Bessesen DH, et al. Pharmacological management of obesity: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(2):342-362. doi:10.1210/jc.2014-3415

6. American Diabetes Association Professional Practice Committee. 3. Prevention or delay of type 2 diabetes and associated comorbidities: standards of medical care in diabetes-2022. Diabetes Care. 2022;45(Suppl 1):S39-S45. doi:10.2337/dc22-S003

7. Phentermine and topiramate extended-release. Package insert. Vivus, Inc; 2012. Accessed April 23, 2024. https://qsymia.com/patient/include/media/pdf/prescribing-information.pdf

8. Naltrexone and bupropion extended-release. Package insert. Orexigen Therapeutics, Inc; 2014. Accessed April 23, 2024. https://contrave.com/wp-content/uploads/2024/01/Contrave-label-113023.pdf

9. Orlistat. Package insert. Roche Laboratories, Inc; 2009. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020766s026lbl.pdf

10. Lorcaserin. Package insert. Arena Pharmaceuticals; 2012. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022529lbl.pdf

11. FDA requests the withdrawal of the weight-loss drug Belviq, Belviq XR (lorcaserin) from the market. News release. US Food & Drug Administration. February 13, 2020. Accessed April 23, 2024. https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market

12. Saxenda Injection (Liraglutide [rDNA origin]). Novo Nordisk, Inc. October 1, 2015. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206321Orig1s000TOC.cfm

13. FDA approves new drug treatment for chronic weight management, first since 2014. News release. US Food & Drug Administration. June 4, 2021. Accessed April 23, 2024. https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014

14. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. New Engl J Med. 2015;373:11-22. doi:10.1056/NEJMoa1411892

15. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. New Engl J Med 2021;384:989-1002. doi:10.1056/NEJMoa2032183

16. Hood SR, Berkeley AW, Moore EA. Evaluation of pharmacologic interventions for weight management in a veteran population. Fed Pract. 2021;38(5):220-226. doi:10.12788/fp.0117

17. Melamed OC, Selby P, Taylor VH. Mental health and obesity during the COVID-19 pandemic. Curr Obes Rep. 2022;11(1):23-31. doi:10.1007/s13679-021-00466-6

Article PDF
Author and Disclosure Information

Allison D. Rodriguez, PharmDa; Amanda P. Ifeachor, PharmD, MPH, BCPSa; Emily A. Moore, PharmD, BCACPa;   Cassandra F. Otte, PharmD, BCACPa; M. Joseph Schopper, PharmDb; Suthat Liangpunsakul, MD, MPHa,c; Amale A. Lteif, MDd

Correspondence:  Allison Rodriguez  (smitherman.allison@gmail.com)

aVeteran Health Indiana, Indianapolis

bCommunity Health Network, Anderson, Indiana

cDivision of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis

dPittsburgh Veterans Affairs Medical Center, Pennsylvania

Acknowledgments

This study was presented at the American Society of Health System Pharmacists Midyear Clinical Meeting and Exhibition in December 2022 in Las Vegas, Nevada. It was also presented at the Great Lakes Pharmacy Resident Conference at Purdue University in April 2023.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review thecomplete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

This study was reviewed by the Indiana University Human Research Protection Program Institutional Review Board and determined to be exempt.

Issue
Federal Practitioner - 41(7)a
Publications
Topics
Page Number
202-207
Sections
Author and Disclosure Information

Allison D. Rodriguez, PharmDa; Amanda P. Ifeachor, PharmD, MPH, BCPSa; Emily A. Moore, PharmD, BCACPa;   Cassandra F. Otte, PharmD, BCACPa; M. Joseph Schopper, PharmDb; Suthat Liangpunsakul, MD, MPHa,c; Amale A. Lteif, MDd

Correspondence:  Allison Rodriguez  (smitherman.allison@gmail.com)

aVeteran Health Indiana, Indianapolis

bCommunity Health Network, Anderson, Indiana

cDivision of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis

dPittsburgh Veterans Affairs Medical Center, Pennsylvania

Acknowledgments

This study was presented at the American Society of Health System Pharmacists Midyear Clinical Meeting and Exhibition in December 2022 in Las Vegas, Nevada. It was also presented at the Great Lakes Pharmacy Resident Conference at Purdue University in April 2023.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review thecomplete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

This study was reviewed by the Indiana University Human Research Protection Program Institutional Review Board and determined to be exempt.

Author and Disclosure Information

Allison D. Rodriguez, PharmDa; Amanda P. Ifeachor, PharmD, MPH, BCPSa; Emily A. Moore, PharmD, BCACPa;   Cassandra F. Otte, PharmD, BCACPa; M. Joseph Schopper, PharmDb; Suthat Liangpunsakul, MD, MPHa,c; Amale A. Lteif, MDd

Correspondence:  Allison Rodriguez  (smitherman.allison@gmail.com)

aVeteran Health Indiana, Indianapolis

bCommunity Health Network, Anderson, Indiana

cDivision of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis

dPittsburgh Veterans Affairs Medical Center, Pennsylvania

Acknowledgments

This study was presented at the American Society of Health System Pharmacists Midyear Clinical Meeting and Exhibition in December 2022 in Las Vegas, Nevada. It was also presented at the Great Lakes Pharmacy Resident Conference at Purdue University in April 2023.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review thecomplete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

This study was reviewed by the Indiana University Human Research Protection Program Institutional Review Board and determined to be exempt.

Article PDF
Article PDF

The Centers for Disease Control and Prevention (CDC) classifies individuals with a body mass index (BMI) of 25 to 29.9as overweight and those with a BMI > 30 as obese (obesity classes: I, BMI 30 to 34.9; II, BMI 35 to 39.9; and III, BMI ≥ 40).1 In 2011, the CDC estimated that 27.4% of adults in the United States were obese; less than a decade later, that number increased to 31.9%.1 In that same period, the percentage of adults in Indiana classified as obese increased from 30.8% to 36.8%.1 About 1 in 14 individuals in the US have class III obesity and 86% of veterans are either overweight or obese.2

High medical expenses can likely be attributed to the long-term health consequences of obesity. Compared to those with a healthy weight, individuals who are overweight or obese are at an increased risk for high blood pressure, high low-density lipoprotein cholesterol levels, low high-density lipoprotein cholesterol levels, high triglyceride levels, type 2 diabetes mellitus (T2DM), coronary heart disease, stroke, gallbladder disease, osteoarthritis, sleep apnea, cancer, mental health disorders, body pain, low quality of life, and death.3 Many of these conditions lead to increased health care needs, medication needs, hospitalizations, and overall health care system use.

Guidelines for the prevention and treatment of obesity have been produced by the American Heart Association, American College of Cardiology, and The Obesity Society; the Endocrine Society; the American Diabetes Association; and the US Departments of Veterans Affairs (VA) and Defense. Each follows a general algorithm to manage and prevent adverse effects (AEs) related to obesity. General practice is to assess a patient for elevated BMI (> 25), implement intense lifestyle modifications including calorie restriction and exercise, reassess for a maintained 5% to 10% weight loss for cardiovascular benefits, and potentially assess for pharmacological or surgical intervention to assist in weight loss.2,4-6

While some weight loss medications (eg, phentermine/topiramate, naltrexone/bupropion, orlistat, and lorcaserin) tend to have unfavorable AEs or mixed efficacy, glucagon-like peptide-1 receptor agonists (GLP-1RAs) have provided new options.7-10 Lorcaserin, for example, was removed from the market in 2020 due to its association with cancer risks.11 The GLP-1RAs liraglutide and semaglutide received US Food and Drug Administration (FDA) approval for weight loss in 2014 and 2021, respectively.12,13 GLP-1RAs have shown the greatest efficacy and benefits in reducing hemoglobin A1c (HbA1c); they are the preferred agents for patients who qualify for pharmacologic intervention for weight loss, especially those with T2DM. However, these studies have not evaluated the long-term outcomes of using these medications for weight loss and may not reflect the veteran population.14,15

 

At Veteran Health Indiana (VHI), clinicians may use several weight loss medications for patients to achieve 5% to 10% weight loss. The medications most often used include liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone. However, more research is needed to determine which weight loss medication is the most beneficial for veterans, particularly following FDA approval of GLP-1RAs. At VHI, phentermine/topiramate is the preferred first-line agent unless patients have contraindications for use, in which case naltrexone/bupropion is recommended. These are considered first-line due to their ease of use in pill form, lower cost, and comparable weight loss to the GLP-1 medication class.2 However, for patients with prediabetes, T2DM, BMI > 40, or BMI > 35 with specific comorbid conditions, liraglutide is preferred because of its beneficial effects for both weight loss and blood glucose control.2

This study aimed to expand on the 2021 Hood and colleagues study that examined total weight loss and weight loss as a percentage of baseline weight in patients with obesity at 3, 6, 12, and > 12 months of pharmacologic therapy by extending the time frame to 48 months.16 This study excluded semaglutide because few patients were prescribed the medication for weight loss during the study.

 

 

METHODS

We conducted a single-center, retrospective chart review of patients prescribed weight loss medications at VHI. A patient list was generated based on prescription fills from June 1, 2017, to July 31, 2021. Data were obtained from the Computerized Patient Record System; patients were not contacted. This study was approved by the Indiana University Health Institutional Review Board and VHI Research and Development Committee.

At the time of this study, liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone were available at VHI for patients who met the clinical criteria for use. All patients must have been enrolled in dietary and lifestyle management programs, including the VA MOVE! program, to be approved for these medications. After the MOVE! orientation, patients could participate in group or individual 12-week programs that included weigh-ins, goal-setting strategies, meal planning, and habit modification support. If patients could not meet in person, phone and other telehealth opportunities were available.

Patients were included in the study if they were aged ≥ 18 years, received a prescription for any of the 5 available medications for weight loss during the enrollment period, and were on the medication for ≥ 6 consecutive months. Patients were excluded if they received a prescription, were treated outside the VA system, or were pregnant. The primary indication for the included medication was not weight loss; the primary indication for the GLP-1RA was T2DM, or the weight loss was attributed to another disease. Adherence was not a measured outcome of this study; if patients were filling the medication, it was assumed they were taking it. Data were collected for each instance of medication use; as a result, a few patients were included more than once. Data collection for a failed medication ended when failure was documented. New data points began when new medication was prescribed; all data were per medication, not per patient. This allowed us to account for medication failure and provide accurate weight loss results based on medication choice within VHI.

Primary outcomes included total weight loss and weight loss as a percentage ofbaseline weight during the study period at 3, 6, 12, 24, 36, and 48 months of therapy. Secondary outcomes included the percentage of patients who lost 5% to 10% of their body weight from baseline; the percentage of patients who maintained ≥ 5% weight loss from baseline to 12, 24, 36, and 48 months if maintained on medication for that duration; duration of medication treatment in weeks; medication discontinuation rate; reason for medication discontinuation; enrollment in the MOVE! clinic and the time enrolled; percentage of patients with a BMI of 18 to 24.9 at the end of the study; and change in HbA1c at 3, 6, 12, 24, 36, and 48 months.

Demographic data included race, age, sex, baseline weight, height, baseline BMI, and comorbid conditions (collected based on the most recent primary care clinical note before initiating medication). Medication data collected included medications used to manage comorbidities. Data related to weight management medication included prescribing clinic, maintenance dose of medication, duration of medication during the study period, the reason for medication discontinuation, or bariatric surgery intervention if applicable.

 


Basic descriptive statistics were used to characterize study participants. For continuous data, analysis of variance tests were used; if those results were not normal, then nonparametric tests were used, followed by pairwise tests between medication groups if the overall test was significant using the Fisher significant differences test. For nominal data, χ2 or Fisher exact tests were used. For comparisons of primary and secondary outcomes, if the analyses needed to include adjustment for confounding variables, analysis of covariance was used for continuous data. A 2-sided 5% significance level was used for all tests.

 

 

RESULTS

ruwophabrogudanatukasachacobesteswilejesp

A total of 228 instances of medication use were identified based on prescription fills; 123 did not meet inclusion criteria (117 for < 6 consecutive months of medication use) (Figure). The study included 105 participants with a mean age of 56 years; 80 were male (76.2%), and 85 identified as White race (81.0%). Mean (SD) weight was 130.1 kg (26.8) and BMI was 41.6 (7.2). The most common comorbid disease states among patients included hypertension, dyslipidemia, obstructive sleep apnea, and T2DM (Table 1). The baseline characteristics were comparable to those of Hood and colleagues.16

sputhuphilatikuslepatavitribestifruseslulachatafrurutridusputrevecladoteslikokojufronocrisleclimishishacrecraclemareslodrimostoprapatraprebajafriveuocleswofruthanoswachemawristetotrapovubedruswacliswujewustespemamenupacluthoneuobraprisewecrac

Most patients at VHI started on liraglutide (63%) or phentermine/topiramate (28%). For primary and secondary outcomes, statistics were calculated to determine whether the results were statistically significant for comparing the liraglutide and phentermine/topiramate subgroups. Sample sizes were too small for statistical analysis for bupropion/naltrexone, phentermine, and orlistat.

Primary Outcomes

The mean (SD) weight of participants dropped 8.1% from 130.1 kg to 119.5 kg over the patient-specific duration of weight management medication therapy for an absolute difference of 10.6 kg (9.7). Duration of individual medication use varied from 6 to 48 months. Weight loss was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. Patient weight was not recorded after the medication was discontinued.

ralashaclulauiclelivespolabrubehesheclawroslucripratrophuchithutrigebeliminebruludrospegevukukubramaphistukuchichopiphifrirothogavubiwrushibuwarefrouotiphonipuhospispichaueslubobrebrathislitra

When classified by medication choice, the mean change in weight over the duration of the study was −23.9 kg for 2 patients using orlistat, −10.2 kg for 46 patients using liraglutide, −11.0 kg for 25 patients using phentermine/topiramate, -7.4 kg for 1 patient using phentermine, and -13.0 kg for 4 patients using naltrexone/bupropion. Patients without a weight documented at the end of their therapy or at the conclusion of the data collection period were not included in the total weight loss at the end of therapy. There were 78 documented instances of weight loss at the end of therapy (Table 2).

kiwistecrohecredribimothaslof

Body weight loss percentage was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. The mean (SD) body weight loss percentage over the duration of the study was 9.2% (11.2). When classified by medication choice, the mean percentage of body weight loss was 16.8% for 2 patients using orlistat, 9.4% for 46 patients using liraglutide, 8.2% for 25 patients using phentermine/topiramate, 6.0% for 1 patient using phentermine alone, and 10.6% for 4 patients using naltrexone/bupropion (Table 3).

Secondary Outcomes

While none of the secondary outcomes were statistically significant, the results of this study suggest that both medications may contribute to weight loss in many patients included in this study. Almost two-thirds of the included patients analyzed lost ≥ 5% of weight from baseline while taking weight management medication. Sixty-six patients (63%) lost ≥ 5% of body weight at any time during the data collection period. When stratified by liraglutide and phentermine/topiramate, 41 patients (63%) taking liraglutide and 20 patients (67%) taking phentermine/topiramate lost ≥ 5% of weight from baseline. Of the 66 patients who lost ≥ 5% of body weight from baseline, 36 (55%) lost ≥ 10% of body weight from baseline at any time during the data collection period.

vothouastinedruwaspuuuslithipruuostubrinicobrurusliherichatidocetecewrohekushasadrelufrobruuuwrasebreprucocedalisirejutubrevohiphedurinishefruwrostutrama

The mean (SD) duration for weight management medication use was 23 months (14.9). Phentermine/topiramate was tolerated longer than liraglutide: 22.7 months vs 21.7 months, respectively (Table 4).

 

The average overall documented medication discontinuation rate was 35.2%. Reasons for discontinuation included 21 patient-elected discontinuations, 8 patients no longer met criteria for use, 4 medications were no longer indicated, and 4 patients experienced AEs. It is unknown whether weight management medication was discontinued or not in 18 patients (17.2%).

 

 

DISCUSSION

This study evaluated the use and outcomes of weight loss medications over a longer period (up to 48 months) than what was previously studied among patients at VHI (12 months). The study aimed to better understand the long-term effect of weight loss medications, determine which medication had better long-term outcomes, and examine the reasons for medication discontinuation.

The results of this study displayed some similarities and differences compared with the Hood and colleagues study.16 Both yielded similar results for 5% of body weight loss and 10% of body weight loss. The largest difference was mean weight loss over the study period. In this study, patients lost a mean 10.6 kg over the course of weight loss medication use compared to 15.8 kg found by Hood and colleagues.16 A reason patients in the current study lost less weight overall could be the difference in time frames. The current study encompassed the COVID-19 pandemic, meaning fewer overall in-person patient appointments, which led to patients being lost to follow-up, missing weigh-ins during the time period, and gaps in care. For some patients, the pandemic possibly contributed to depression, missed medication doses, and a more sedentary lifestyle, leading to more weight gain.17 Telemedicine services at VHI expanded during the pandemic in an attempt to increase patient monitoring and counseling. It is unclear whether this expansion was enough to replace the in-person contact necessary to promote a healthy lifestyle.

VA pharmacists now care for patients through telehealth and are more involved in weight loss management. Since the conclusion of the Hood and colleagues study and start of this research, 2 pharmacists at VHI have been assigned to follow patients for obesity management to help with adherence to medication and lifestyle changes, management of AEs, dispense logistics, interventions for medications that may cause weight gain, and case management of glycemic control and weight loss with GLP-1RAs. Care management by pharmacists at VHI helps improve the logistics of titratable orders and save money by improving the use of high-cost items like GLP-1RAs. VA clinical pharmacy practitioners already monitor GLP-1RAs for patients with T2DM, so they are prepared to educate and assist patients with these medications.

It is important to continue developing a standardized process for weight loss medication management across the VA to improve the quality of patient care and optimize prescription outcomes. VA facilities differ in how weight loss management care is delivered and the level at which pharmacists are involved. Given the high rate of obesity among patients at the VA, the advent of new prescription options for weight loss, and the high cost associated with these medications, there has been increased attention to obesity care. Some Veterans Integrated Service Networks are forming a weight management community of practice groups to create standard operating procedures and algorithms to standardize care. Developing consistent processes is necessary to improve weight loss and patient care for veterans regardless where they receive treatment.

Limitations

The data used in this study were dependent on clinician documentation. Because of a lack of documentation in many instances, it was difficult to determine the full efficacy of the medications studied due to missing weight recordings. The lack of documentation made it difficult to determine whether patients were enrolled and active in the MOVE! program. It is required that patients enroll in MOVE! to obtain medications, but many did not have any follow-up MOVE! visits after initially obtaining their weight loss medication.

In this study, differences in the outcomes of patients with and without T2DM were not compared. It is the VA standard of care to prefer liraglutide over phentermine/topiramate in patients with T2DM or prediabetes.2 This makes it difficult to assess whether phentermine/topiramate or liraglutide is more effective for weight loss in patients with T2DM. Weight gain after the discontinuation of weight loss medications was not assessed. Collecting this data may help determine whether a certain weight loss medication is less likely to cause rebound weight gain when discontinued.

Other limitations to this study consisted of excluding patients who discontinued therapy within 6 months, small sample sizes on some medications, and lack of data on adherence. Adherence was based on medication refills, which means that if a patient refilled the medication, it was assumed they were taking it. This is not always the case, and while accurate data on adherence is difficult to gather, it can impact how results may be interpreted. These additional limitations make it difficult to accurately determine the efficacy of the medications in this study.

 

CONCLUSIONS

This study found similar outcomes to what has been observed in larger clinical trials regarding weight loss medications. Nevertheless, there was a lack of accurate clinical documentation for most patients, which limits the conclusions. This lack of documentation potentially led to inaccurate results. It revealed that many patients at VHI did not uniformly receive consistent follow-up after starting a weight loss medication during the study period. With more standardized processes implemented at VA facilities, increased pharmacist involvement in weight loss medication management, and increased use of established telehealth services, patients could have the opportunity for closer follow-up that may lead to better weight loss outcomes. With these changes, there is more reason for additional studies to be conducted to assess follow-up, medication management, and weight loss overall.

The Centers for Disease Control and Prevention (CDC) classifies individuals with a body mass index (BMI) of 25 to 29.9as overweight and those with a BMI > 30 as obese (obesity classes: I, BMI 30 to 34.9; II, BMI 35 to 39.9; and III, BMI ≥ 40).1 In 2011, the CDC estimated that 27.4% of adults in the United States were obese; less than a decade later, that number increased to 31.9%.1 In that same period, the percentage of adults in Indiana classified as obese increased from 30.8% to 36.8%.1 About 1 in 14 individuals in the US have class III obesity and 86% of veterans are either overweight or obese.2

High medical expenses can likely be attributed to the long-term health consequences of obesity. Compared to those with a healthy weight, individuals who are overweight or obese are at an increased risk for high blood pressure, high low-density lipoprotein cholesterol levels, low high-density lipoprotein cholesterol levels, high triglyceride levels, type 2 diabetes mellitus (T2DM), coronary heart disease, stroke, gallbladder disease, osteoarthritis, sleep apnea, cancer, mental health disorders, body pain, low quality of life, and death.3 Many of these conditions lead to increased health care needs, medication needs, hospitalizations, and overall health care system use.

Guidelines for the prevention and treatment of obesity have been produced by the American Heart Association, American College of Cardiology, and The Obesity Society; the Endocrine Society; the American Diabetes Association; and the US Departments of Veterans Affairs (VA) and Defense. Each follows a general algorithm to manage and prevent adverse effects (AEs) related to obesity. General practice is to assess a patient for elevated BMI (> 25), implement intense lifestyle modifications including calorie restriction and exercise, reassess for a maintained 5% to 10% weight loss for cardiovascular benefits, and potentially assess for pharmacological or surgical intervention to assist in weight loss.2,4-6

While some weight loss medications (eg, phentermine/topiramate, naltrexone/bupropion, orlistat, and lorcaserin) tend to have unfavorable AEs or mixed efficacy, glucagon-like peptide-1 receptor agonists (GLP-1RAs) have provided new options.7-10 Lorcaserin, for example, was removed from the market in 2020 due to its association with cancer risks.11 The GLP-1RAs liraglutide and semaglutide received US Food and Drug Administration (FDA) approval for weight loss in 2014 and 2021, respectively.12,13 GLP-1RAs have shown the greatest efficacy and benefits in reducing hemoglobin A1c (HbA1c); they are the preferred agents for patients who qualify for pharmacologic intervention for weight loss, especially those with T2DM. However, these studies have not evaluated the long-term outcomes of using these medications for weight loss and may not reflect the veteran population.14,15

 

At Veteran Health Indiana (VHI), clinicians may use several weight loss medications for patients to achieve 5% to 10% weight loss. The medications most often used include liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone. However, more research is needed to determine which weight loss medication is the most beneficial for veterans, particularly following FDA approval of GLP-1RAs. At VHI, phentermine/topiramate is the preferred first-line agent unless patients have contraindications for use, in which case naltrexone/bupropion is recommended. These are considered first-line due to their ease of use in pill form, lower cost, and comparable weight loss to the GLP-1 medication class.2 However, for patients with prediabetes, T2DM, BMI > 40, or BMI > 35 with specific comorbid conditions, liraglutide is preferred because of its beneficial effects for both weight loss and blood glucose control.2

This study aimed to expand on the 2021 Hood and colleagues study that examined total weight loss and weight loss as a percentage of baseline weight in patients with obesity at 3, 6, 12, and > 12 months of pharmacologic therapy by extending the time frame to 48 months.16 This study excluded semaglutide because few patients were prescribed the medication for weight loss during the study.

 

 

METHODS

We conducted a single-center, retrospective chart review of patients prescribed weight loss medications at VHI. A patient list was generated based on prescription fills from June 1, 2017, to July 31, 2021. Data were obtained from the Computerized Patient Record System; patients were not contacted. This study was approved by the Indiana University Health Institutional Review Board and VHI Research and Development Committee.

At the time of this study, liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone were available at VHI for patients who met the clinical criteria for use. All patients must have been enrolled in dietary and lifestyle management programs, including the VA MOVE! program, to be approved for these medications. After the MOVE! orientation, patients could participate in group or individual 12-week programs that included weigh-ins, goal-setting strategies, meal planning, and habit modification support. If patients could not meet in person, phone and other telehealth opportunities were available.

Patients were included in the study if they were aged ≥ 18 years, received a prescription for any of the 5 available medications for weight loss during the enrollment period, and were on the medication for ≥ 6 consecutive months. Patients were excluded if they received a prescription, were treated outside the VA system, or were pregnant. The primary indication for the included medication was not weight loss; the primary indication for the GLP-1RA was T2DM, or the weight loss was attributed to another disease. Adherence was not a measured outcome of this study; if patients were filling the medication, it was assumed they were taking it. Data were collected for each instance of medication use; as a result, a few patients were included more than once. Data collection for a failed medication ended when failure was documented. New data points began when new medication was prescribed; all data were per medication, not per patient. This allowed us to account for medication failure and provide accurate weight loss results based on medication choice within VHI.

Primary outcomes included total weight loss and weight loss as a percentage ofbaseline weight during the study period at 3, 6, 12, 24, 36, and 48 months of therapy. Secondary outcomes included the percentage of patients who lost 5% to 10% of their body weight from baseline; the percentage of patients who maintained ≥ 5% weight loss from baseline to 12, 24, 36, and 48 months if maintained on medication for that duration; duration of medication treatment in weeks; medication discontinuation rate; reason for medication discontinuation; enrollment in the MOVE! clinic and the time enrolled; percentage of patients with a BMI of 18 to 24.9 at the end of the study; and change in HbA1c at 3, 6, 12, 24, 36, and 48 months.

Demographic data included race, age, sex, baseline weight, height, baseline BMI, and comorbid conditions (collected based on the most recent primary care clinical note before initiating medication). Medication data collected included medications used to manage comorbidities. Data related to weight management medication included prescribing clinic, maintenance dose of medication, duration of medication during the study period, the reason for medication discontinuation, or bariatric surgery intervention if applicable.

 


Basic descriptive statistics were used to characterize study participants. For continuous data, analysis of variance tests were used; if those results were not normal, then nonparametric tests were used, followed by pairwise tests between medication groups if the overall test was significant using the Fisher significant differences test. For nominal data, χ2 or Fisher exact tests were used. For comparisons of primary and secondary outcomes, if the analyses needed to include adjustment for confounding variables, analysis of covariance was used for continuous data. A 2-sided 5% significance level was used for all tests.

 

 

RESULTS

ruwophabrogudanatukasachacobesteswilejesp

A total of 228 instances of medication use were identified based on prescription fills; 123 did not meet inclusion criteria (117 for < 6 consecutive months of medication use) (Figure). The study included 105 participants with a mean age of 56 years; 80 were male (76.2%), and 85 identified as White race (81.0%). Mean (SD) weight was 130.1 kg (26.8) and BMI was 41.6 (7.2). The most common comorbid disease states among patients included hypertension, dyslipidemia, obstructive sleep apnea, and T2DM (Table 1). The baseline characteristics were comparable to those of Hood and colleagues.16

sputhuphilatikuslepatavitribestifruseslulachatafrurutridusputrevecladoteslikokojufronocrisleclimishishacrecraclemareslodrimostoprapatraprebajafriveuocleswofruthanoswachemawristetotrapovubedruswacliswujewustespemamenupacluthoneuobraprisewecrac

Most patients at VHI started on liraglutide (63%) or phentermine/topiramate (28%). For primary and secondary outcomes, statistics were calculated to determine whether the results were statistically significant for comparing the liraglutide and phentermine/topiramate subgroups. Sample sizes were too small for statistical analysis for bupropion/naltrexone, phentermine, and orlistat.

Primary Outcomes

The mean (SD) weight of participants dropped 8.1% from 130.1 kg to 119.5 kg over the patient-specific duration of weight management medication therapy for an absolute difference of 10.6 kg (9.7). Duration of individual medication use varied from 6 to 48 months. Weight loss was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. Patient weight was not recorded after the medication was discontinued.

ralashaclulauiclelivespolabrubehesheclawroslucripratrophuchithutrigebeliminebruludrospegevukukubramaphistukuchichopiphifrirothogavubiwrushibuwarefrouotiphonipuhospispichaueslubobrebrathislitra

When classified by medication choice, the mean change in weight over the duration of the study was −23.9 kg for 2 patients using orlistat, −10.2 kg for 46 patients using liraglutide, −11.0 kg for 25 patients using phentermine/topiramate, -7.4 kg for 1 patient using phentermine, and -13.0 kg for 4 patients using naltrexone/bupropion. Patients without a weight documented at the end of their therapy or at the conclusion of the data collection period were not included in the total weight loss at the end of therapy. There were 78 documented instances of weight loss at the end of therapy (Table 2).

kiwistecrohecredribimothaslof

Body weight loss percentage was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. The mean (SD) body weight loss percentage over the duration of the study was 9.2% (11.2). When classified by medication choice, the mean percentage of body weight loss was 16.8% for 2 patients using orlistat, 9.4% for 46 patients using liraglutide, 8.2% for 25 patients using phentermine/topiramate, 6.0% for 1 patient using phentermine alone, and 10.6% for 4 patients using naltrexone/bupropion (Table 3).

Secondary Outcomes

While none of the secondary outcomes were statistically significant, the results of this study suggest that both medications may contribute to weight loss in many patients included in this study. Almost two-thirds of the included patients analyzed lost ≥ 5% of weight from baseline while taking weight management medication. Sixty-six patients (63%) lost ≥ 5% of body weight at any time during the data collection period. When stratified by liraglutide and phentermine/topiramate, 41 patients (63%) taking liraglutide and 20 patients (67%) taking phentermine/topiramate lost ≥ 5% of weight from baseline. Of the 66 patients who lost ≥ 5% of body weight from baseline, 36 (55%) lost ≥ 10% of body weight from baseline at any time during the data collection period.

vothouastinedruwaspuuuslithipruuostubrinicobrurusliherichatidocetecewrohekushasadrelufrobruuuwrasebreprucocedalisirejutubrevohiphedurinishefruwrostutrama

The mean (SD) duration for weight management medication use was 23 months (14.9). Phentermine/topiramate was tolerated longer than liraglutide: 22.7 months vs 21.7 months, respectively (Table 4).

 

The average overall documented medication discontinuation rate was 35.2%. Reasons for discontinuation included 21 patient-elected discontinuations, 8 patients no longer met criteria for use, 4 medications were no longer indicated, and 4 patients experienced AEs. It is unknown whether weight management medication was discontinued or not in 18 patients (17.2%).

 

 

DISCUSSION

This study evaluated the use and outcomes of weight loss medications over a longer period (up to 48 months) than what was previously studied among patients at VHI (12 months). The study aimed to better understand the long-term effect of weight loss medications, determine which medication had better long-term outcomes, and examine the reasons for medication discontinuation.

The results of this study displayed some similarities and differences compared with the Hood and colleagues study.16 Both yielded similar results for 5% of body weight loss and 10% of body weight loss. The largest difference was mean weight loss over the study period. In this study, patients lost a mean 10.6 kg over the course of weight loss medication use compared to 15.8 kg found by Hood and colleagues.16 A reason patients in the current study lost less weight overall could be the difference in time frames. The current study encompassed the COVID-19 pandemic, meaning fewer overall in-person patient appointments, which led to patients being lost to follow-up, missing weigh-ins during the time period, and gaps in care. For some patients, the pandemic possibly contributed to depression, missed medication doses, and a more sedentary lifestyle, leading to more weight gain.17 Telemedicine services at VHI expanded during the pandemic in an attempt to increase patient monitoring and counseling. It is unclear whether this expansion was enough to replace the in-person contact necessary to promote a healthy lifestyle.

VA pharmacists now care for patients through telehealth and are more involved in weight loss management. Since the conclusion of the Hood and colleagues study and start of this research, 2 pharmacists at VHI have been assigned to follow patients for obesity management to help with adherence to medication and lifestyle changes, management of AEs, dispense logistics, interventions for medications that may cause weight gain, and case management of glycemic control and weight loss with GLP-1RAs. Care management by pharmacists at VHI helps improve the logistics of titratable orders and save money by improving the use of high-cost items like GLP-1RAs. VA clinical pharmacy practitioners already monitor GLP-1RAs for patients with T2DM, so they are prepared to educate and assist patients with these medications.

It is important to continue developing a standardized process for weight loss medication management across the VA to improve the quality of patient care and optimize prescription outcomes. VA facilities differ in how weight loss management care is delivered and the level at which pharmacists are involved. Given the high rate of obesity among patients at the VA, the advent of new prescription options for weight loss, and the high cost associated with these medications, there has been increased attention to obesity care. Some Veterans Integrated Service Networks are forming a weight management community of practice groups to create standard operating procedures and algorithms to standardize care. Developing consistent processes is necessary to improve weight loss and patient care for veterans regardless where they receive treatment.

Limitations

The data used in this study were dependent on clinician documentation. Because of a lack of documentation in many instances, it was difficult to determine the full efficacy of the medications studied due to missing weight recordings. The lack of documentation made it difficult to determine whether patients were enrolled and active in the MOVE! program. It is required that patients enroll in MOVE! to obtain medications, but many did not have any follow-up MOVE! visits after initially obtaining their weight loss medication.

In this study, differences in the outcomes of patients with and without T2DM were not compared. It is the VA standard of care to prefer liraglutide over phentermine/topiramate in patients with T2DM or prediabetes.2 This makes it difficult to assess whether phentermine/topiramate or liraglutide is more effective for weight loss in patients with T2DM. Weight gain after the discontinuation of weight loss medications was not assessed. Collecting this data may help determine whether a certain weight loss medication is less likely to cause rebound weight gain when discontinued.

Other limitations to this study consisted of excluding patients who discontinued therapy within 6 months, small sample sizes on some medications, and lack of data on adherence. Adherence was based on medication refills, which means that if a patient refilled the medication, it was assumed they were taking it. This is not always the case, and while accurate data on adherence is difficult to gather, it can impact how results may be interpreted. These additional limitations make it difficult to accurately determine the efficacy of the medications in this study.

 

CONCLUSIONS

This study found similar outcomes to what has been observed in larger clinical trials regarding weight loss medications. Nevertheless, there was a lack of accurate clinical documentation for most patients, which limits the conclusions. This lack of documentation potentially led to inaccurate results. It revealed that many patients at VHI did not uniformly receive consistent follow-up after starting a weight loss medication during the study period. With more standardized processes implemented at VA facilities, increased pharmacist involvement in weight loss medication management, and increased use of established telehealth services, patients could have the opportunity for closer follow-up that may lead to better weight loss outcomes. With these changes, there is more reason for additional studies to be conducted to assess follow-up, medication management, and weight loss overall.

References

1. Overweight & obesity. Centers for Disease Control and Prevention. Updated September 21, 2023. Accessed April 23, 2024. https://www.cdc.gov/obesity/index.html

2. US Department of Defense, US Department of Veterans Affairs. The Management of Adult Overweight and Obesity Working Group. VA/DoD Clinical Practice Guideline for the Management of Adult Overweight and Obesity. Updated July 2020. Accessed April 23, 2024. https://www.healthquality.va.gov/guidelines/CD/obesity/VADoDObesityCPGFinal5087242020.pdf

3. Health effects of overweight and obesity. Centers for Disease Control and Prevention. Updated September 24, 2022. Accessed April 23, 2024. https://www.cdc.gov/healthyweight/effects/index.html

4. Jensen MD, Ryan DH, Apovian CM, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol. 2014;63(25 Pt B):2985-3023. doi:10.1016/j.jacc.2013.11.004

5. Apovian CM, Aronne LJ, Bessesen DH, et al. Pharmacological management of obesity: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(2):342-362. doi:10.1210/jc.2014-3415

6. American Diabetes Association Professional Practice Committee. 3. Prevention or delay of type 2 diabetes and associated comorbidities: standards of medical care in diabetes-2022. Diabetes Care. 2022;45(Suppl 1):S39-S45. doi:10.2337/dc22-S003

7. Phentermine and topiramate extended-release. Package insert. Vivus, Inc; 2012. Accessed April 23, 2024. https://qsymia.com/patient/include/media/pdf/prescribing-information.pdf

8. Naltrexone and bupropion extended-release. Package insert. Orexigen Therapeutics, Inc; 2014. Accessed April 23, 2024. https://contrave.com/wp-content/uploads/2024/01/Contrave-label-113023.pdf

9. Orlistat. Package insert. Roche Laboratories, Inc; 2009. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020766s026lbl.pdf

10. Lorcaserin. Package insert. Arena Pharmaceuticals; 2012. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022529lbl.pdf

11. FDA requests the withdrawal of the weight-loss drug Belviq, Belviq XR (lorcaserin) from the market. News release. US Food & Drug Administration. February 13, 2020. Accessed April 23, 2024. https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market

12. Saxenda Injection (Liraglutide [rDNA origin]). Novo Nordisk, Inc. October 1, 2015. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206321Orig1s000TOC.cfm

13. FDA approves new drug treatment for chronic weight management, first since 2014. News release. US Food & Drug Administration. June 4, 2021. Accessed April 23, 2024. https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014

14. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. New Engl J Med. 2015;373:11-22. doi:10.1056/NEJMoa1411892

15. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. New Engl J Med 2021;384:989-1002. doi:10.1056/NEJMoa2032183

16. Hood SR, Berkeley AW, Moore EA. Evaluation of pharmacologic interventions for weight management in a veteran population. Fed Pract. 2021;38(5):220-226. doi:10.12788/fp.0117

17. Melamed OC, Selby P, Taylor VH. Mental health and obesity during the COVID-19 pandemic. Curr Obes Rep. 2022;11(1):23-31. doi:10.1007/s13679-021-00466-6

References

1. Overweight & obesity. Centers for Disease Control and Prevention. Updated September 21, 2023. Accessed April 23, 2024. https://www.cdc.gov/obesity/index.html

2. US Department of Defense, US Department of Veterans Affairs. The Management of Adult Overweight and Obesity Working Group. VA/DoD Clinical Practice Guideline for the Management of Adult Overweight and Obesity. Updated July 2020. Accessed April 23, 2024. https://www.healthquality.va.gov/guidelines/CD/obesity/VADoDObesityCPGFinal5087242020.pdf

3. Health effects of overweight and obesity. Centers for Disease Control and Prevention. Updated September 24, 2022. Accessed April 23, 2024. https://www.cdc.gov/healthyweight/effects/index.html

4. Jensen MD, Ryan DH, Apovian CM, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol. 2014;63(25 Pt B):2985-3023. doi:10.1016/j.jacc.2013.11.004

5. Apovian CM, Aronne LJ, Bessesen DH, et al. Pharmacological management of obesity: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(2):342-362. doi:10.1210/jc.2014-3415

6. American Diabetes Association Professional Practice Committee. 3. Prevention or delay of type 2 diabetes and associated comorbidities: standards of medical care in diabetes-2022. Diabetes Care. 2022;45(Suppl 1):S39-S45. doi:10.2337/dc22-S003

7. Phentermine and topiramate extended-release. Package insert. Vivus, Inc; 2012. Accessed April 23, 2024. https://qsymia.com/patient/include/media/pdf/prescribing-information.pdf

8. Naltrexone and bupropion extended-release. Package insert. Orexigen Therapeutics, Inc; 2014. Accessed April 23, 2024. https://contrave.com/wp-content/uploads/2024/01/Contrave-label-113023.pdf

9. Orlistat. Package insert. Roche Laboratories, Inc; 2009. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020766s026lbl.pdf

10. Lorcaserin. Package insert. Arena Pharmaceuticals; 2012. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022529lbl.pdf

11. FDA requests the withdrawal of the weight-loss drug Belviq, Belviq XR (lorcaserin) from the market. News release. US Food & Drug Administration. February 13, 2020. Accessed April 23, 2024. https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market

12. Saxenda Injection (Liraglutide [rDNA origin]). Novo Nordisk, Inc. October 1, 2015. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206321Orig1s000TOC.cfm

13. FDA approves new drug treatment for chronic weight management, first since 2014. News release. US Food & Drug Administration. June 4, 2021. Accessed April 23, 2024. https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014

14. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. New Engl J Med. 2015;373:11-22. doi:10.1056/NEJMoa1411892

15. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. New Engl J Med 2021;384:989-1002. doi:10.1056/NEJMoa2032183

16. Hood SR, Berkeley AW, Moore EA. Evaluation of pharmacologic interventions for weight management in a veteran population. Fed Pract. 2021;38(5):220-226. doi:10.12788/fp.0117

17. Melamed OC, Selby P, Taylor VH. Mental health and obesity during the COVID-19 pandemic. Curr Obes Rep. 2022;11(1):23-31. doi:10.1007/s13679-021-00466-6

Issue
Federal Practitioner - 41(7)a
Issue
Federal Practitioner - 41(7)a
Page Number
202-207
Page Number
202-207
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>0724 FED Weight Loss</fileName> <TBEID>0C02F7D1.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F7D1</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240709T163554</firstPublished> <LastPublished>20240709T163554</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240709T163554</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Allison D. Rodriguez, PharmDa; Amanda P. Ifeachor, PharmD, MPH, BCPSa; Emily A. Moore, PharmD, BCACPa; Cassandra F. Otte, PharmD, BCACPa; M. Joseph Schopper, PharmDb; Suthat Liangpunsakul, MD, MPHa,c; Amale A. Lteif, MDd</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The Centers for Disease Control and Prevention (CDC) classifies individuals with a body mass index (BMI) of 25 to 29.9 as overweight and those with a BMI &gt; 30 a</metaDescription> <articlePDF/> <teaserImage/> <title>Long-Term Assessment of Weight Loss Medications in a Veteran Population</title> <deck/> <eyebrow>Original Research</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>7</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>7</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2951</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>July 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Feature | 2951<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">27442</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Long-Term Assessment of Weight Loss Medications in a Veteran Population</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Overweight and obesity are common in the veteran population. Medical management with semaglutide, orlistat, liraglutide, phentermine, phentermine/topiramate, and naltrexone/bupropion is increasingly common. This study expands on a 2021 study and evaluates medication effectiveness. <br/><br/><b>Methods:</b> This single-center retrospective study analyzed patients prescribed weight loss medications at Veteran Health Indiana. Primary outcomes included body weight loss and total weight loss with each medication at 3, 6, 12, 24, 36, and 48 months. Secondary outcomes were also used to determine the efficacy of the current weight loss medications.<br/><br/><b>Results:</b> Of 105 included patients, 66 were treated with liraglutide, 30 with phentermine/topiramate, 5 with naltrexone/bupropion, 3 with orlistat, and 1 with phentermine. The absolute weight loss for all medications was 10.6 kg over the patient-specific duration of weight management therapy. The mean body weight loss was 9.2%. There were no statistically significant differences in primary or secondary outcomes between liraglutide and phentermine/topiramate. The group sizes were too small to analyze the other medication groups. <br/><br/><b>Conclusions:</b> Patients in this study lost weight while using medications. However, there were no statistically significant differences among the medications. Patients did not receive uniformly consistent follow-up care, suggesting the need for more standardized processes that could lead to better weight loss outcomes.</p> <p><span class="Drop">T</span>he Centers for Disease Control and Prevention (CDC) classifies individuals with a body mass index (BMI) of 25 to 29.9<sup> </sup>as overweight and those with a BMI &gt; 30 as obese (obesity classes: I, BMI 30 to 34.9; II, BMI 35 to 39.9; and III, BMI ≥ 40).<sup>1</sup> In 2011, the CDC estimated that 27.4% of adults in the United States were obese; less than a decade later, that number increased to 31.9%.<sup>1</sup> In that same period, the percentage of adults in Indiana classified as obese increased from 30.8% to 36.8%.<sup>1</sup> About 1 in 14 individuals in the US have class III obesity and 86% of veterans are either overweight or obese.<sup>2</sup></p> <p>High medical expenses can likely be attributed to the long-term health consequences of obesity. Compared to those with a healthy weight, individuals who are overweight or obese are at an increased risk for high blood pressure, high low-density lipoprotein cholesterol levels, low high-density lipoprotein cholesterol levels, high triglyceride levels, type 2 diabetes mellitus (T2DM), coronary heart disease, stroke, gallbladder disease, osteoarthritis, sleep apnea, cancer, mental health disorders, body pain, low quality of life, and death.<sup>3</sup> Many of these conditions lead to increased health care needs, medication needs, hospitalizations, and overall health care system use.<br/><br/>Guidelines for the prevention and treatment of obesity have been produced by the American Heart Association, American College of Cardiology, and The Obesity Society; the Endocrine Society; the American Diabetes Association; and the US Departments of Veterans Affairs (VA) and Defense. Each follows a general algorithm to manage and prevent adverse effects (AEs) related to obesity. General practice is to assess a patient for elevated BMI (&gt; 25), implement intense lifestyle modifications including calorie restriction and exercise, reassess for a maintained 5% to 10% weight loss for cardiovascular benefits, and potentially assess for pharmacological or surgical intervention to assist in weight loss.<sup>2,4-6<br/><br/></sup>While some weight loss medications (eg, phentermine/topiramate, naltrexone/bupropion, orlistat, and lorcaserin) tend to have unfavorable AEs or mixed efficacy, glucagon-like peptide-1 receptor agonists (GLP-1RAs) have provided new options.<sup>7-10</sup> Lorcaserin, for example, was removed from the market in 2020 due to its association with cancer risks.<sup>11</sup> The GLP-1RAs liraglutide and semaglutide received US Food and Drug Administration (FDA) approval for weight loss in 2014 and 2021, respectively.<sup>12,13</sup> GLP-1RAs have shown the greatest efficacy and benefits in reducing hemoglobin A<sub>1c</sub> (HbA<sub>1c</sub>); they are the preferred agents for patients who qualify for pharmacologic intervention for weight loss, especially those with T2DM. However, these studies have not evaluated the long-term outcomes of using these medications for weight loss and may not reflect the veteran population.<sup>14,15<br/><br/></sup>At Veteran Health Indiana (VHI), clinicians may use several weight loss medications for patients to achieve 5% to 10% weight loss. The medications most often used include liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone. However, more research is needed to determine which weight loss medication is the most beneficial for veterans, particularly following FDA approval of GLP-1RAs. At VHI, phentermine/topiramate is the preferred first-line agent unless patients have contraindications for use, in which case naltrexone/bupropion is recommended. These are considered first-line due to their ease of use in pill form, lower cost, and comparable weight loss to the GLP-1 medication class.<sup>2</sup> However, for patients with prediabetes, T2DM, BMI &gt; 40, or BMI &gt; 35 with specific comorbid conditions, liraglutide is preferred because of its beneficial effects for both weight loss and blood glucose control.<sup>2<br/><br/></sup>This study aimed to expand on the 2021 Hood and colleagues study that examined total weight loss and weight loss as a percentage of baseline weight in patients with obesity at 3, 6, 12, and &gt; 12 months of pharmacologic therapy by extending the time frame to 48 months.<sup>16</sup> This study excluded semaglutide because few patients were prescribed the medication for weight loss during the study.</p> <h2>METHODS</h2> <p>We conducted a single-center, retrospective chart review of patients prescribed weight loss medications at VHI. A patient list was generated based on prescription fills from June 1, 2017, to July 31, 2021. Data were obtained from the Computerized Patient Record System; patients were not contacted. This study was approved by the Indiana University Health Institutional Review Board and VHI Research and Development Committee.</p> <p>At the time of this study, liraglutide, phentermine/topiramate, naltrexone/bupropion, orlistat, and phentermine alone were available at VHI for patients who met the clinical criteria for use. All patients must have been enrolled in dietary and lifestyle management programs, including the VA MOVE! program, to be approved for these medications. After the MOVE! orientation, patients could participate in group or individual 12-week programs that included weigh-ins, goal-setting strategies, meal planning, and habit modification support. If patients could not meet in person, phone and other telehealth opportunities were available.<br/><br/>Patients were included in the study if they were aged ≥ 18 years, received a prescription for any of the 5 available medications for weight loss during the enrollment period, and were on the medication for ≥ 6 consecutive months. Patients were excluded if they received a prescription, were treated outside the VA system, or were pregnant. The primary indication for the included medication was not weight loss; the primary indication for the GLP-1RA was T2DM, or the weight loss was attributed to another disease. Adherence was not a measured outcome of this study; if patients were filling the medication, it was assumed they were taking it. Data were collected for each instance of medication use; as a result, a few patients were included more than once. Data collection for a failed medication ended when failure was documented. New data points began when new medication was prescribed; all data were per medication, not per patient. This allowed us to account for medication failure and provide accurate weight loss results based on medication choice within VHI.<br/><br/>Primary outcomes included total weight loss and weight loss as a percentage ofbaseline weight during the study period at 3, 6, 12, 24, 36, and 48 months of therapy. Secondary outcomes included the percentage of patients who lost 5% to 10% of their body weight from baseline; the percentage of patients who maintained ≥ 5% weight loss from baseline to 12, 24, 36, and 48 months if maintained on medication for that duration; duration of medication treatment in weeks; medication discontinuation rate; reason for medication discontinuation; enrollment in the MOVE! clinic and the time enrolled; percentage of patients with a BMI of 18 to 24.9 at the end of the study; and change in HbA<sub>1c</sub> at 3, 6, 12, 24, 36, and 48 months.<br/><br/>Demographic data included race, age, sex, baseline weight, height, baseline BMI, and comorbid conditions (collected based on the most recent primary care clinical note before initiating medication). Medication data collected included medications used to manage comorbidities. Data related to weight management medication included prescribing clinic, maintenance dose of medication, duration of medication during the study period, the reason for medication discontinuation, or bariatric surgery intervention if applicable.<br/><br/>Basic descriptive statistics were used to characterize study participants. For continuous data, analysis of variance tests were used; if those results were not normal, then nonparametric tests were used, followed by pairwise tests between medication groups if the overall test was significant using the Fisher significant differences test. For nominal data, χ<sup>2</sup> or Fisher exact tests were used. For comparisons of primary and secondary outcomes, if the analyses needed to include adjustment for confounding variables, analysis of covariance was used for continuous data. A 2-sided 5% significance level was used for all tests. </p> <h2>RESULTS</h2> <p>A total of 228 instances of medication use were identified based on prescription fills; 123 did not meet inclusion criteria (117 for &lt; 6 consecutive months of medication use)(Figure). The study included 105 participants with a mean age of 56 years; 80 were male (76.2%), and 85 identified as White race (81.0%). Mean (SD) weight was 130.1 kg (26.8) and BMI was 41.6 (7.2). The most common comorbid disease states among patients included hypertension, dyslipidemia, obstructive sleep apnea, and T2DM (Table 1). The baseline characteristics were comparable to those of Hood and colleagues.<sup>16</sup></p> <p>Most patients at VHI started on liraglutide (63%) or phentermine/topiramate (28%). For primary and secondary outcomes, statistics were calculated to determine whether the results were statistically significant for comparing the liraglutide and phentermine/topiramate subgroups. Sample sizes were too small for statistical analysis for bupropion/naltrexone, phentermine, and orlistat. </p> <h3>Primary Outcomes</h3> <p>The mean (SD) weight of participants dropped 8.1% from 130.1 kg to 119.5 kg over the patient-specific duration of weight management medication therapy for an absolute difference of 10.6 kg (9.7). Duration of individual medication use varied from 6 to 48 months. Weight loss was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. Patient weight was not recorded after the medication was discontinued.</p> <p>When classified by medication choice, the mean change in weight over the duration of the study was −23.9 kg for 2 patients using orlistat, −10.2 kg for 46 patients using liraglutide, −11.0 kg for 25 patients using phentermine/topiramate, -7.4 kg for 1 patient using phentermine, and -13.0 kg for 4 patients using naltrexone/bupropion. Patients without a weight documented at the end of their therapy or at the conclusion of the data collection period were not included in the total weight loss at the end of therapy. There were 78 documented instances of weight loss at the end of therapy (Table 2).<br/><br/>Body weight loss percentage was recorded at 6, 12, 24, 36, and 48 months of weight management therapy. The mean (SD) body weight loss percentage over the duration of the study was 9.2% (11.2). When classified by medication choice, the mean percentage of body weight loss was 16.8% for 2 patients using orlistat, 9.4% for 46 patients using liraglutide, 8.2% for 25 patients using phentermine/topiramate, 6.0% for 1 patient using phentermine alone, and 10.6% for 4 patients using naltrexone/bupropion (Table 3).</p> <h3>Secondary Outcomes </h3> <p>While none of the secondary outcomes were statistically significant, the results of this study suggest that both medications may contribute to weight loss in many patients included in this study. Almost two-thirds of the included patients analyzed lost ≥ 5% of weight from baseline while taking weight management medication. Sixty-six patients (63%) lost ≥ 5% of body weight at any time during the data collection period. When stratified by liraglutide and phentermine/topiramate, 41 patients (63%) taking liraglutide and 20 patients (67%) taking phentermine/topiramate lost ≥ 5% of weight from baseline. Of the 66 patients who lost ≥ 5% of body weight from baseline, 36 (55%) lost ≥ 10% of body weight from baseline at any time during the data collection period.</p> <p>The mean (SD) duration for weight management medication use was 23 months (14.9). Phentermine/topiramate was tolerated longer than liraglutide: 22.7 months vs 21.7 months, respectively (Table 4). <br/><br/>The average overall documented medication discontinuation rate was 35.2%. Reasons for discontinuation included 21 patient-elected discontinuations, 8 patients no longer met criteria for use, 4 medications were no longer indicated, and 4 patients experienced AEs. It is unknown whether weight management medication was discontinued or not in 18 patients (17.2%).</p> <h2>DISCUSSION</h2> <p>This study evaluated the use and outcomes of weight loss medications over a longer period (up to 48 months) than what was previously studied among patients at VHI (12 months). The study aimed to better understand the long-term effect of weight loss medications, determine which medication had better long-term outcomes, and examine the reasons for medication discontinuation. </p> <p>The results of this study displayed some similarities and differences compared with the Hood and colleagues study.<sup>16</sup> Both yielded similar results for 5% of body weight loss and 10% of body weight loss. The largest difference was mean weight loss over the study period. In this study, patients lost a mean 10.6 kg over the course of weight loss medication use compared to 15.8 kg found by Hood and colleagues.<sup>16</sup> A reason patients in the current study lost less weight overall could be the difference in time frames. The current study encompassed the COVID-19 pandemic, meaning fewer overall in-person patient appointments, which led to patients being lost to follow-up, missing weigh-ins during the time period, and gaps in care. For some patients, the pandemic possibly contributed to depression, missed medication doses, and a more sedentary lifestyle, leading to more weight gain.<sup>17</sup> Telemedicine services at VHI expanded during the pandemic in an attempt to increase patient monitoring and counseling. It is unclear whether this expansion was enough to replace the in-person contact necessary to promote a healthy lifestyle.<br/><br/>VA pharmacists now care for patients through telehealth and are more involved in weight loss management. Since the conclusion of the Hood and colleagues study and start of this research, 2 pharmacists at VHI have been assigned to follow patients for obesity management to help with adherence to medication and lifestyle changes, management of AEs, dispense logistics, interventions for medications that may cause weight gain, and case management of glycemic control and weight loss with GLP-1RAs. Care management by pharmacists at VHI helps improve the logistics of titratable orders and save money by improving the use of high-cost items like GLP-1RAs. VA clinical pharmacy practitioners already monitor GLP-1RAs for patients with T2DM, so they are prepared to educate and assist patients with these medications. <br/><br/>It is important to continue developing a standardized process for weight loss medication management across the VA to improve the quality of patient care and optimize prescription outcomes. VA facilities differ in how weight loss management care is delivered and the level at which pharmacists are involved. Given the high rate of obesity among patients at the VA, the advent of new prescription options for weight loss, and the high cost associated with these medications, there has been increased attention to obesity care. Some Veterans Integrated Service Networks are forming a weight management community of practice groups to create standard operating procedures and algorithms to standardize care. Developing consistent processes is necessary to improve weight loss and patient care for veterans regardless where they receive treatment.</p> <h3>Limitations</h3> <p>The data used in this study were dependent on clinician documentation. Because of a lack of documentation in many instances, it was difficult to determine the full efficacy of the medications studied due to missing weight recordings. The lack of documentation made it difficult to determine whether patients were enrolled and active in the MOVE! program. It is required that patients enroll in MOVE! to obtain medications, but many did not have any follow-up MOVE! visits after initially obtaining their weight loss medication.</p> <p>In this study, differences in the outcomes of patients with and without T2DM were not compared. It is the VA standard of care to prefer liraglutide over phentermine/topiramate in patients with T2DM or prediabetes.<sup>2</sup> This makes it difficult to assess whether phentermine/topiramate or liraglutide is more effective for weight loss in patients with T2DM. Weight gain after the discontinuation of weight loss medications was not assessed. Collecting this data may help determine whether a certain weight loss medication is less likely to cause rebound weight gain when discontinued.<br/><br/>Other limitations to this study consisted of excluding patients who discontinued therapy within 6 months, small sample sizes on some medications, and lack of data on adherence. Adherence was based on medication refills, which means that if a patient refilled the medication, it was assumed they were taking it. This is not always the case, and while accurate data on adherence is difficult to gather, it can impact how results may be interpreted. These additional limitations make it difficult to accurately determine the efficacy of the medications in this study.</p> <h2>CONCLUSIONS</h2> <p>This study found similar outcomes to what has been observed in larger clinical trials regarding weight loss medications. Nevertheless, there was a lack of accurate clinical documentation for most patients, which limits the conclusions. This lack of documentation potentially led to inaccurate results. It revealed that many patients at VHI did not uniformly receive consistent follow-up after starting a weight loss medication during the study period. With more standardized processes implemented at VA facilities, increased pharmacist involvement in weight loss medication management, and increased use of established telehealth services, patients could have the opportunity for closer follow-up that may lead to better weight loss outcomes. With these changes, there is more reason for additional studies to be conducted to assess follow-up, medication management, and weight loss overall. </p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Veteran Health Indiana, Indianapolis<br/><br/><sup>b</sup>Community Health Network, Anderson, Indiana<sup>c</sup>Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis<br/><br/><sup>d</sup>Pittsburgh Veterans Affairs Medical Center, Pennsylvania</em> </p> <p class="isub">Acknowledgments</p> <p> <em>This study was presented at the American Society of Health System Pharmacists Midyear Clinical Meeting and Exhibition in December 2022 in Las Vegas, Nevada. It was also presented at the Great Lakes Pharmacy Resident Conference at Purdue University in April 2023.</em> </p> <p class="isub">Author disclosures </p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner</i>, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review thecomplete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.</em> </p> <p class="isub">Ethics and consent</p> <p> <em>This study was reviewed by the Indiana University Human Research Protection Program Institutional Review Board and determined to be exempt.</em> </p> <h2>References</h2> <p class="reference"> 1. Overweight &amp; obesity. Centers for Disease Control and Prevention. Updated September 21, 2023. Accessed April 23, 2024. https://www.cdc.gov/obesity/index.html<br/><br/> 2. US Department of Defense, US Department of Veterans Affairs. The Management of Adult Overweight and Obesity Working Group. VA/DoD Clinical Practice Guideline for the Management of Adult Overweight and Obesity. Updated July 2020. Accessed April 23, 2024. https://www.healthquality.va.gov/guidelines/CD/obesity/VADoDObesityCPGFinal5087242020.pdf<br/><br/> 3. Health effects of overweight and obesity. Centers for Disease Control and Prevention. Updated September 24, 2022. Accessed April 23, 2024. https://www.cdc.gov/healthyweight/effects/index.html<br/><br/> 4. Jensen MD, Ryan DH, Apovian CM, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. <i>J Am Coll Cardiol</i>. 2014;63(25 Pt B):2985-3023. doi:10.1016/j.jacc.2013.11.004<br/><br/> 5. Apovian CM, Aronne LJ, Bessesen DH, et al. Pharmacological management of obesity: an endocrine society clinical practice guideline. <i>J Clin Endocrinol Metab.</i> 2015;100(2):342-362. doi:10.1210/jc.2014-3415<br/><br/> 6. American Diabetes Association Professional Practice Committee. 3. Prevention or delay of type 2 diabetes and associated comorbidities: standards of medical care in diabetes-2022. <i>Diabetes Care</i>. 2022;45(Suppl 1):S39-S45. doi:10.2337/dc22-S003<br/><br/> 7. Phentermine and topiramate extended-release. Package insert. Vivus, Inc; 2012. Accessed April 23, 2024. https://qsymia.com/patient/include/media/pdf/prescribing-information.pdf<br/><br/> 8. Naltrexone and bupropion extended-release. Package insert. Orexigen Therapeutics, Inc; 2014. Accessed April 23, 2024. https://contrave.com/wp-content/uploads/2024/01/Contrave-label-113023.pdf<br/><br/> 9. Orlistat. Package insert. Roche Laboratories, Inc; 2009. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020766s026lbl.pdf<br/><br/>10. Lorcaserin. Package insert. Arena Pharmaceuticals; 2012. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/022529lbl.pdf<br/><br/>11. FDA requests the withdrawal of the weight-loss drug Belviq, Belviq XR (lorcaserin) from the market. News release. US Food &amp; Drug Administration. February 13, 2020. Accessed April 23, 2024. https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market<br/><br/>12. Saxenda Injection (Liraglutide [rDNA origin]). Novo Nordisk, Inc. October 1, 2015. Accessed April 23, 2024. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206321Orig1s000TOC.cfm<br/><br/>13. FDA approves new drug treatment for chronic weight management, first since 2014. News release. US Food &amp; Drug Administration. June 4, 2021. Accessed April 23, 2024. https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014<br/><br/>14. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. <i>New Engl J Med</i>.<i> </i>2015;373:11-22. doi:10.1056/NEJMoa1411892<br/><br/>15. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. <i>New Engl J Med</i> 2021;384:989-1002. doi:10.1056/NEJMoa2032183<br/><br/>16. Hood SR, Berkeley AW, Moore EA. Evaluation of pharmacologic interventions for weight management in a veteran population. <i>Fed Pract</i>. 2021;38(5):220-226. doi:10.12788/fp.0117<br/><br/>17. Melamed OC, Selby P, Taylor VH. Mental health and obesity during the COVID-19 pandemic. <i>Curr Obes Rep.</i> 2022;11(1):23-31. doi:10.1007/s13679-021-00466-6</p> </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
Article PDF Media
Image
Disable zoom
Off

Common Antidepressants Ranked by Potential for Weight Gain

Article Type
Changed
Wed, 07/10/2024 - 14:08

 

Eight commonly used antidepressants have been ranked by their weight gain potential. 

Results of a large observational study showed small differences in short- and long-term weight change in patients prescribed one of eight antidepressants, with bupropion associated with the lowest weight gain and escitalopramparoxetine, and duloxetine associated with the greatest. 

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with those taking sertraline, which was used as a comparator. 

Investigators noted that the more clinicians and patients know about how a particular antidepressant may affect patients’ weight, the better informed they can be about which antidepressants to prescribe. 

“Patients and their clinicians often have several options when starting an antidepressant for the first time. This study provides important real-world evidence regarding the amount of weight gain that should be expected after starting some of the most common antidepressants,” lead author Joshua Petimar, ScD, assistant professor of population medicine in the Harvard Pilgrim Health Care Institute at Harvard Medical School, Boston, said in a press release. 

The findings were published online in Annals of Internal Medicine


 

Real-World Data

Though weight gain is a commonly reported side effect of antidepressant use and may lead to medication nonadherence and worse outcomes, there is a lack of real-world data about weight change across specific medications. 

Investigators used electronic health records from eight health care systems across the United States spanning from 2010 to 2019. The analysis included information on 183,118 adults aged 20-80 years who were new users of one of eight common first-line antidepressants. Investigators measured their weight at baseline and at 6, 12, and 24 months after initiation to estimate intention-to-treat (ITT) effects of weight change.

At baseline, participants were randomly assigned to begin sertraline, citalopram, escitalopram, fluoxetine, paroxetine, bupropion, duloxetine, or venlafaxine

The most common antidepressants prescribed were sertraline, citalopram, and bupropion. Approximately 36% of participants had a diagnosis of depression, and 39% were diagnosed with anxiety.

Among selective serotonin reuptake inhibitors (SSRIs), escitalopram and paroxetine were associated with the greatest 6-month weight gain, whereas bupropion was associated with the least weight gain across all analyses.

Using sertraline as a comparator, 6-month weight change was lower for bupropion (difference, 0.22 kg) and higher for escitalopram (difference, 0.41 kg), duloxetine (difference, 0.34 kg), paroxetine (difference, 0.37 kg), and venlafaxine (difference, 0.17 kg).

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with sertraline users.

Investigators noted little difference in adherence levels between medications during the study except at 6 months, when it was higher for those who took bupropion (41%) than for those taking other antidepressants (28%-36%).

The study included data only on prescriptions and investigators could not verify whether the medications were dispensed or taken as prescribed. Other limitations included missing weight information because most patients did not encounter the health system at exactly 6, 12, and 24 months; only 15%-30% had weight measurements in those months. 

Finally, the low adherence rates made it difficult to attribute relative weight change at the 12- and 24-month time points to the specific medications of interest.

“Clinicians and patients could consider these differences when making decisions about specific antidepressants, especially given the complex relationships of obesity and depression with health, quality of life, and stigma,” the authors wrote. 

The study was funded by the National Institute of Diabetes and Digestive and Kidney Diseases. Disclosures are noted in the original article. 

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

Eight commonly used antidepressants have been ranked by their weight gain potential. 

Results of a large observational study showed small differences in short- and long-term weight change in patients prescribed one of eight antidepressants, with bupropion associated with the lowest weight gain and escitalopramparoxetine, and duloxetine associated with the greatest. 

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with those taking sertraline, which was used as a comparator. 

Investigators noted that the more clinicians and patients know about how a particular antidepressant may affect patients’ weight, the better informed they can be about which antidepressants to prescribe. 

“Patients and their clinicians often have several options when starting an antidepressant for the first time. This study provides important real-world evidence regarding the amount of weight gain that should be expected after starting some of the most common antidepressants,” lead author Joshua Petimar, ScD, assistant professor of population medicine in the Harvard Pilgrim Health Care Institute at Harvard Medical School, Boston, said in a press release. 

The findings were published online in Annals of Internal Medicine


 

Real-World Data

Though weight gain is a commonly reported side effect of antidepressant use and may lead to medication nonadherence and worse outcomes, there is a lack of real-world data about weight change across specific medications. 

Investigators used electronic health records from eight health care systems across the United States spanning from 2010 to 2019. The analysis included information on 183,118 adults aged 20-80 years who were new users of one of eight common first-line antidepressants. Investigators measured their weight at baseline and at 6, 12, and 24 months after initiation to estimate intention-to-treat (ITT) effects of weight change.

At baseline, participants were randomly assigned to begin sertraline, citalopram, escitalopram, fluoxetine, paroxetine, bupropion, duloxetine, or venlafaxine

The most common antidepressants prescribed were sertraline, citalopram, and bupropion. Approximately 36% of participants had a diagnosis of depression, and 39% were diagnosed with anxiety.

Among selective serotonin reuptake inhibitors (SSRIs), escitalopram and paroxetine were associated with the greatest 6-month weight gain, whereas bupropion was associated with the least weight gain across all analyses.

Using sertraline as a comparator, 6-month weight change was lower for bupropion (difference, 0.22 kg) and higher for escitalopram (difference, 0.41 kg), duloxetine (difference, 0.34 kg), paroxetine (difference, 0.37 kg), and venlafaxine (difference, 0.17 kg).

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with sertraline users.

Investigators noted little difference in adherence levels between medications during the study except at 6 months, when it was higher for those who took bupropion (41%) than for those taking other antidepressants (28%-36%).

The study included data only on prescriptions and investigators could not verify whether the medications were dispensed or taken as prescribed. Other limitations included missing weight information because most patients did not encounter the health system at exactly 6, 12, and 24 months; only 15%-30% had weight measurements in those months. 

Finally, the low adherence rates made it difficult to attribute relative weight change at the 12- and 24-month time points to the specific medications of interest.

“Clinicians and patients could consider these differences when making decisions about specific antidepressants, especially given the complex relationships of obesity and depression with health, quality of life, and stigma,” the authors wrote. 

The study was funded by the National Institute of Diabetes and Digestive and Kidney Diseases. Disclosures are noted in the original article. 

A version of this article appeared on Medscape.com.

 

Eight commonly used antidepressants have been ranked by their weight gain potential. 

Results of a large observational study showed small differences in short- and long-term weight change in patients prescribed one of eight antidepressants, with bupropion associated with the lowest weight gain and escitalopramparoxetine, and duloxetine associated with the greatest. 

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with those taking sertraline, which was used as a comparator. 

Investigators noted that the more clinicians and patients know about how a particular antidepressant may affect patients’ weight, the better informed they can be about which antidepressants to prescribe. 

“Patients and their clinicians often have several options when starting an antidepressant for the first time. This study provides important real-world evidence regarding the amount of weight gain that should be expected after starting some of the most common antidepressants,” lead author Joshua Petimar, ScD, assistant professor of population medicine in the Harvard Pilgrim Health Care Institute at Harvard Medical School, Boston, said in a press release. 

The findings were published online in Annals of Internal Medicine


 

Real-World Data

Though weight gain is a commonly reported side effect of antidepressant use and may lead to medication nonadherence and worse outcomes, there is a lack of real-world data about weight change across specific medications. 

Investigators used electronic health records from eight health care systems across the United States spanning from 2010 to 2019. The analysis included information on 183,118 adults aged 20-80 years who were new users of one of eight common first-line antidepressants. Investigators measured their weight at baseline and at 6, 12, and 24 months after initiation to estimate intention-to-treat (ITT) effects of weight change.

At baseline, participants were randomly assigned to begin sertraline, citalopram, escitalopram, fluoxetine, paroxetine, bupropion, duloxetine, or venlafaxine

The most common antidepressants prescribed were sertraline, citalopram, and bupropion. Approximately 36% of participants had a diagnosis of depression, and 39% were diagnosed with anxiety.

Among selective serotonin reuptake inhibitors (SSRIs), escitalopram and paroxetine were associated with the greatest 6-month weight gain, whereas bupropion was associated with the least weight gain across all analyses.

Using sertraline as a comparator, 6-month weight change was lower for bupropion (difference, 0.22 kg) and higher for escitalopram (difference, 0.41 kg), duloxetine (difference, 0.34 kg), paroxetine (difference, 0.37 kg), and venlafaxine (difference, 0.17 kg).

Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with sertraline users.

Investigators noted little difference in adherence levels between medications during the study except at 6 months, when it was higher for those who took bupropion (41%) than for those taking other antidepressants (28%-36%).

The study included data only on prescriptions and investigators could not verify whether the medications were dispensed or taken as prescribed. Other limitations included missing weight information because most patients did not encounter the health system at exactly 6, 12, and 24 months; only 15%-30% had weight measurements in those months. 

Finally, the low adherence rates made it difficult to attribute relative weight change at the 12- and 24-month time points to the specific medications of interest.

“Clinicians and patients could consider these differences when making decisions about specific antidepressants, especially given the complex relationships of obesity and depression with health, quality of life, and stigma,” the authors wrote. 

The study was funded by the National Institute of Diabetes and Digestive and Kidney Diseases. Disclosures are noted in the original article. 

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>168665</fileName> <TBEID>0C050E90.SIG</TBEID> <TBUniqueIdentifier>MD_0C050E90</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240709T142425</QCDate> <firstPublished>20240709T145310</firstPublished> <LastPublished>20240709T145310</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240709T145310</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Eve Bender</byline> <bylineText>EVE BENDER</bylineText> <bylineFull>EVE BENDER</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>Investigators noted that the more clinicians and patients know about how a particular antidepressant may affect patients’ weight, the better informed they can b</metaDescription> <articlePDF/> <teaserImage/> <teaser>Patients receiving escitalopram, paroxetine, and duloxetine 10%-15% more likely to gain at least 5% of baseline weight.</teaser> <title>Common Antidepressants Ranked by Potential for Weight Gain</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>cpn</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> </publications_g> <publications> <term canonical="true">9</term> <term>21</term> <term>15</term> </publications> <sections> <term canonical="true">39313</term> </sections> <topics> <term canonical="true">202</term> <term>261</term> <term>248</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Common Antidepressants Ranked by Potential for Weight Gain</title> <deck/> </itemMeta> <itemContent> <p>Eight commonly used antidepressants have been ranked by their weight gain potential. </p> <p>Results of a large observational study showed small differences in short- and long-term weight change in patients prescribed one of eight antidepressants, with <span class="Hyperlink">bupropion</span> associated with the lowest weight gain and <span class="Hyperlink">escitalopram</span>, <span class="Hyperlink">paroxetine</span>, and <span class="Hyperlink">duloxetine</span> associated with the greatest. <br/><br/>Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with those taking <span class="Hyperlink">sertraline</span>, which was used as a comparator. <br/><br/><span class="tag metaDescription">Investigators noted that the more clinicians and patients know about how a particular antidepressant may affect patients’ weight, the better informed they can be about which antidepressants to prescribe.</span> <br/><br/>“Patients and their clinicians often have several options when starting an antidepressant for the first time. This study provides important real-world evidence regarding the amount of weight gain that should be expected after starting some of the most common antidepressants,” lead author Joshua Petimar, ScD, assistant professor of population medicine in the Harvard Pilgrim Health Care Institute at Harvard Medical School, Boston, said in a press release. <br/><br/>The findings were <span class="Hyperlink"><a href="https://www.acpjournals.org/doi/10.7326/M23-2742">published online</a></span> in <em>Annals of Internal Medicine</em>. <br/><br/><br/><br/></p> <h2>Real-World Data</h2> <p>Though weight gain is a commonly reported side effect of antidepressant use and may lead to medication nonadherence and worse outcomes, there is a lack of real-world data about weight change across specific medications. <br/><br/>Investigators used electronic health records from eight health care systems across the United States spanning from 2010 to 2019. The analysis included information on 183,118 adults aged 20-80 years who were new users of one of eight common first-line antidepressants. Investigators measured their weight at baseline and at 6, 12, and 24 months after initiation to estimate intention-to-treat (ITT) effects of weight change.<br/><br/>At baseline, participants were randomly assigned to begin sertraline, citalopram, escitalopram, <span class="Hyperlink">fluoxetine</span>, paroxetine, bupropion, duloxetine, or <span class="Hyperlink">venlafaxine</span>. <br/><br/>The most common antidepressants prescribed were sertraline, citalopram, and bupropion. Approximately 36% of participants had a diagnosis of <span class="Hyperlink">depression</span>, and 39% were diagnosed with anxiety.<br/><br/>Among selective serotonin reuptake inhibitors (SSRIs), escitalopram and paroxetine were associated with the greatest 6-month weight gain, whereas bupropion was associated with the least weight gain across all analyses.<br/><br/>Using sertraline as a comparator, 6-month weight change was lower for bupropion (difference, 0.22 kg) and higher for escitalopram (difference, 0.41 kg), duloxetine (difference, 0.34 kg), paroxetine (difference, 0.37 kg), and venlafaxine (difference, 0.17 kg).<br/><br/>Escitalopram, paroxetine, and duloxetine users were 10%-15% more likely to gain at least 5% of their baseline weight compared with sertraline users.<br/><br/>Investigators noted little difference in adherence levels between medications during the study except at 6 months, when it was higher for those who took bupropion (41%) than for those taking other antidepressants (28%-36%).<br/><br/>The study included data only on prescriptions and investigators could not verify whether the medications were dispensed or taken as prescribed. Other limitations included missing weight information because most patients did not encounter the health system at exactly 6, 12, and 24 months; only 15%-30% had weight measurements in those months. <br/><br/>Finally, the low adherence rates made it difficult to attribute relative weight change at the 12- and 24-month time points to the specific medications of interest.<br/><br/>“Clinicians and patients could consider these differences when making decisions about specific antidepressants, especially given the complex relationships of <span class="Hyperlink">obesity</span> and depression with health, quality of life, and stigma,” the authors wrote. <br/><br/>The study was funded by the National Institute of Diabetes and Digestive and Kidney Diseases. Disclosures are noted in the original article.<span class="end"/> </p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/eight-common-antidepressants-ranked-weight-gain-potential-2024a1000c9v">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

Semaglutide May Increase Risk of Disease Causing Vision Loss

Article Type
Changed
Tue, 07/09/2024 - 14:50

 

TOPLINE: 

Patients with type 2 diabetes, overweight, or obesity taking the glucagon-like peptide receptor agonist (GLP-1 RA) semaglutide appear to have an increased risk for an uncommon condition that can cause vision loss. 
 

METHODOLOGY:

  • Researchers conducted a retrospective study of 16,827 patients at Massachusetts Eye and Ear in Boston.
  • Their analysis focused on 710 patients with type 2 diabetes (194 of whom had been prescribed semaglutide) and 979 patients with overweight or obesity (361 prescribed semaglutide).
  • The researchers compared patients prescribed semaglutide with those prescribed a medication other than a GLP-1 agent. They matched patients by factors such as age and sex and whether they had hypertension, obstructive sleep apnea, or coronary artery disease.
  • They assessed the cumulative incidence of nonarteritic anterior ischemic optic neuropathy (NAION) during 36 months of follow-up. 
  •  

TAKEAWAY: 

  • Semaglutide use was associated with a higher risk for NAION in patients with type 2 diabetes (hazard ratio [HR], 4.28; 95% CI, 1.62-11.29).
  • In patients with overweight or obesity, semaglutide again was linked to a higher risk for NAION (HR, 7.64; 95% CI, 2.21-26.36).
  • Among patients with type 2 diabetes, the cumulative incidence of NAION over 36 months was 8.9% for those prescribed semaglutide vs 1.8% among those taking non–GLP-1 medications.
  • For patients with overweight or obesity, the cumulative incidence of NAION over 36 months was 6.7% for the semaglutide cohort vs 0.8% for those in the other group. 

IN PRACTICE:

Semaglutide has “provided very significant benefits in many ways, but future discussions between a patient and their physician should include NAION as a potential risk,” study leader Joseph Rizzo, MD, with Mass Eye and Ear and Harvard Medical School, said in a news release about the findings. “It is important to appreciate, however, that the increased risk relates to a disorder that is relatively uncommon.”

“Given the numbers of participants who have been recruited to clinical trials and the large number of people globally who use GLP-1 RAs, we should be confident that if corroborated, the absolute risk of developing NAION in direct relation to taking semaglutide must indeed be rare,” Susan P. Mollan, MBcHB, of University Hospitals Birmingham NHS Foundation Trust, in England, wrote in a commentary published with the study. 
 

SOURCE:

The study was published online on July 3 in JAMA Ophthalmology.
 

LIMITATIONS: 

The patients were seen at a hospital that specializes in ophthalmology and has a specialized neuro-ophthalmology service, so the results may not fully apply to other settings. The results were driven by a relatively small number of NAION cases in the patients exposed to semaglutide. The study does not establish that semaglutide directly causes NAION, the researchers noted. “The best approaches to confirm, refute, or refine our findings would be to conduct a much larger, retrospective, multicenter population-based cohort study; a prospective, randomized clinical study; or a postmarket analysis of all GLP-1 RA drugs,” they wrote.

 

DISCLOSURES:

The study was supported by a grant from Research to Prevent Blindness. 
 

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE: 

Patients with type 2 diabetes, overweight, or obesity taking the glucagon-like peptide receptor agonist (GLP-1 RA) semaglutide appear to have an increased risk for an uncommon condition that can cause vision loss. 
 

METHODOLOGY:

  • Researchers conducted a retrospective study of 16,827 patients at Massachusetts Eye and Ear in Boston.
  • Their analysis focused on 710 patients with type 2 diabetes (194 of whom had been prescribed semaglutide) and 979 patients with overweight or obesity (361 prescribed semaglutide).
  • The researchers compared patients prescribed semaglutide with those prescribed a medication other than a GLP-1 agent. They matched patients by factors such as age and sex and whether they had hypertension, obstructive sleep apnea, or coronary artery disease.
  • They assessed the cumulative incidence of nonarteritic anterior ischemic optic neuropathy (NAION) during 36 months of follow-up. 
  •  

TAKEAWAY: 

  • Semaglutide use was associated with a higher risk for NAION in patients with type 2 diabetes (hazard ratio [HR], 4.28; 95% CI, 1.62-11.29).
  • In patients with overweight or obesity, semaglutide again was linked to a higher risk for NAION (HR, 7.64; 95% CI, 2.21-26.36).
  • Among patients with type 2 diabetes, the cumulative incidence of NAION over 36 months was 8.9% for those prescribed semaglutide vs 1.8% among those taking non–GLP-1 medications.
  • For patients with overweight or obesity, the cumulative incidence of NAION over 36 months was 6.7% for the semaglutide cohort vs 0.8% for those in the other group. 

IN PRACTICE:

Semaglutide has “provided very significant benefits in many ways, but future discussions between a patient and their physician should include NAION as a potential risk,” study leader Joseph Rizzo, MD, with Mass Eye and Ear and Harvard Medical School, said in a news release about the findings. “It is important to appreciate, however, that the increased risk relates to a disorder that is relatively uncommon.”

“Given the numbers of participants who have been recruited to clinical trials and the large number of people globally who use GLP-1 RAs, we should be confident that if corroborated, the absolute risk of developing NAION in direct relation to taking semaglutide must indeed be rare,” Susan P. Mollan, MBcHB, of University Hospitals Birmingham NHS Foundation Trust, in England, wrote in a commentary published with the study. 
 

SOURCE:

The study was published online on July 3 in JAMA Ophthalmology.
 

LIMITATIONS: 

The patients were seen at a hospital that specializes in ophthalmology and has a specialized neuro-ophthalmology service, so the results may not fully apply to other settings. The results were driven by a relatively small number of NAION cases in the patients exposed to semaglutide. The study does not establish that semaglutide directly causes NAION, the researchers noted. “The best approaches to confirm, refute, or refine our findings would be to conduct a much larger, retrospective, multicenter population-based cohort study; a prospective, randomized clinical study; or a postmarket analysis of all GLP-1 RA drugs,” they wrote.

 

DISCLOSURES:

The study was supported by a grant from Research to Prevent Blindness. 
 

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on Medscape.com.

 

TOPLINE: 

Patients with type 2 diabetes, overweight, or obesity taking the glucagon-like peptide receptor agonist (GLP-1 RA) semaglutide appear to have an increased risk for an uncommon condition that can cause vision loss. 
 

METHODOLOGY:

  • Researchers conducted a retrospective study of 16,827 patients at Massachusetts Eye and Ear in Boston.
  • Their analysis focused on 710 patients with type 2 diabetes (194 of whom had been prescribed semaglutide) and 979 patients with overweight or obesity (361 prescribed semaglutide).
  • The researchers compared patients prescribed semaglutide with those prescribed a medication other than a GLP-1 agent. They matched patients by factors such as age and sex and whether they had hypertension, obstructive sleep apnea, or coronary artery disease.
  • They assessed the cumulative incidence of nonarteritic anterior ischemic optic neuropathy (NAION) during 36 months of follow-up. 
  •  

TAKEAWAY: 

  • Semaglutide use was associated with a higher risk for NAION in patients with type 2 diabetes (hazard ratio [HR], 4.28; 95% CI, 1.62-11.29).
  • In patients with overweight or obesity, semaglutide again was linked to a higher risk for NAION (HR, 7.64; 95% CI, 2.21-26.36).
  • Among patients with type 2 diabetes, the cumulative incidence of NAION over 36 months was 8.9% for those prescribed semaglutide vs 1.8% among those taking non–GLP-1 medications.
  • For patients with overweight or obesity, the cumulative incidence of NAION over 36 months was 6.7% for the semaglutide cohort vs 0.8% for those in the other group. 

IN PRACTICE:

Semaglutide has “provided very significant benefits in many ways, but future discussions between a patient and their physician should include NAION as a potential risk,” study leader Joseph Rizzo, MD, with Mass Eye and Ear and Harvard Medical School, said in a news release about the findings. “It is important to appreciate, however, that the increased risk relates to a disorder that is relatively uncommon.”

“Given the numbers of participants who have been recruited to clinical trials and the large number of people globally who use GLP-1 RAs, we should be confident that if corroborated, the absolute risk of developing NAION in direct relation to taking semaglutide must indeed be rare,” Susan P. Mollan, MBcHB, of University Hospitals Birmingham NHS Foundation Trust, in England, wrote in a commentary published with the study. 
 

SOURCE:

The study was published online on July 3 in JAMA Ophthalmology.
 

LIMITATIONS: 

The patients were seen at a hospital that specializes in ophthalmology and has a specialized neuro-ophthalmology service, so the results may not fully apply to other settings. The results were driven by a relatively small number of NAION cases in the patients exposed to semaglutide. The study does not establish that semaglutide directly causes NAION, the researchers noted. “The best approaches to confirm, refute, or refine our findings would be to conduct a much larger, retrospective, multicenter population-based cohort study; a prospective, randomized clinical study; or a postmarket analysis of all GLP-1 RA drugs,” they wrote.

 

DISCLOSURES:

The study was supported by a grant from Research to Prevent Blindness. 
 

This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. 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>168667</fileName> <TBEID>0C050E94.SIG</TBEID> <TBUniqueIdentifier>MD_0C050E94</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240709T141439</QCDate> <firstPublished>20240709T144707</firstPublished> <LastPublished>20240709T144707</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240709T144707</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Edited by Jake Remaly</byline> <bylineText>EDITED JAKE REMALY</bylineText> <bylineFull>EDITED JAKE REMALY</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>For patients with overweight or obesity, the cumulative incidence of NAION over 36 months was 6.7% for the semaglutide cohort vs 0.8% for those in the other gro</metaDescription> <articlePDF/> <teaserImage/> <teaser>Retrospective study finds increased risk for NAION for patients with T2D and overweight or obesity taking semaglutide.</teaser> <title>Semaglutide May Increase Risk of Disease Causing Vision Loss</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>endo</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term>21</term> <term>15</term> <term canonical="true">34</term> </publications> <sections> <term canonical="true">39313</term> <term>27970</term> </sections> <topics> <term canonical="true">261</term> <term>280</term> <term>205</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Semaglutide May Increase Risk of Disease Causing Vision Loss</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE: </h2> <p>Patients with type 2 diabetes, overweight, or <span class="Hyperlink">obesity</span> taking the <span class="Hyperlink">glucagon</span>-like peptide receptor agonist (GLP-1 RA) semaglutide appear to have an increased risk for an uncommon condition that can cause vision loss. <br/><br/></p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>Researchers conducted a retrospective study of 16,827 patients at Massachusetts Eye and Ear in Boston.</li> <li>Their analysis focused on 710 patients with type 2 diabetes (194 of whom had been prescribed semaglutide) and 979 patients with overweight or obesity (361 prescribed semaglutide).</li> <li>The researchers compared patients prescribed semaglutide with those prescribed a medication other than a GLP-1 agent. They matched patients by factors such as age and sex and whether they had <span class="Hyperlink">hypertension</span>, obstructive sleep apnea, or <span class="Hyperlink">coronary artery disease</span>.</li> <li>They assessed the cumulative incidence of nonarteritic <span class="Hyperlink">anterior ischemic optic neuropathy</span> (NAION) during 36 months of follow-up. </li> <li/> </ul> <h2>TAKEAWAY: </h2> <ul class="body"> <li>Semaglutide use was associated with a higher risk for NAION in patients with type 2 diabetes (hazard ratio [HR], 4.28; 95% CI, 1.62-11.29).</li> <li>In patients with overweight or obesity, semaglutide again was linked to a higher risk for NAION (HR, 7.64; 95% CI, 2.21-26.36).</li> <li>Among patients with type 2 diabetes, the cumulative incidence of NAION over 36 months was 8.9% for those prescribed semaglutide vs 1.8% among those taking non–GLP-1 medications.</li> <li><span class="tag metaDescription">For patients with overweight or obesity, the cumulative incidence of NAION over 36 months was 6.7% for the semaglutide cohort vs 0.8% for those in the other group.</span> </li> </ul> <h2>IN PRACTICE:</h2> <p>Semaglutide has “provided very significant benefits in many ways, but future discussions between a patient and their physician should include NAION as a potential risk,” study leader Joseph Rizzo, MD, with Mass Eye and Ear and Harvard Medical School, <span class="Hyperlink"><a href="https://www.eurekalert.org/news-releases/1050055">said in a news release</a></span> about the findings. “It is important to appreciate, however, that the increased risk relates to a disorder that is relatively uncommon.”<br/><br/>“Given the numbers of participants who have been recruited to clinical trials and the large number of people globally who use GLP-1 RAs, we should be confident that if corroborated, the absolute risk of developing NAION in direct relation to taking semaglutide must indeed be rare,” Susan P. Mollan, MBcHB, of University Hospitals Birmingham NHS Foundation Trust, in England, wrote <span class="Hyperlink"><a href="https://jamanetwork.com/journals/jamaophthalmology/fullarticle/10.1001/jamaophthalmol.2024.2514?guestAccessKey=d529160a-60b5-4ca0-87ac-278b71c943ba&amp;utm_source=for_the_media&amp;utm_medium=referral&amp;utm_campaign=ftm_links&amp;utm_content=tfl&amp;utm_term=070324">in a commentary</a></span> published with the study. <br/><br/></p> <h2>SOURCE:</h2> <p>The study <span class="Hyperlink"><a href="https://jamanetwork.com/journals/jamaophthalmology/fullarticle/10.1001/jamaophthalmol.2024.2296?guestAccessKey=bb0f187b-7fb2-4e73-8cc1-52cd65b44324&amp;utm_source=For_The_Media&amp;utm_medium=referral&amp;utm_campaign=ftm_links&amp;utm_content=tfl&amp;utm_term=070324">was published online</a></span> on July 3 in <em>JAMA Ophthalmolog</em>y.<br/><br/></p> <h2>LIMITATIONS: </h2> <p>The patients were seen at a hospital that specializes in ophthalmology and has a specialized neuro-ophthalmology service, so the results may not fully apply to other settings. The results were driven by a relatively small number of NAION cases in the patients exposed to semaglutide. The study does not establish that semaglutide directly causes NAION, the researchers noted. “The best approaches to confirm, refute, or refine our findings would be to conduct a much larger, retrospective, multicenter population-based cohort study; a prospective, randomized clinical study; or a postmarket analysis of all GLP-1 RA drugs,” they wrote.<br/><br/></p> <h2>DISCLOSURES:</h2> <p>The study was supported by a grant from Research to Prevent Blindness. <br/><br/></p> <p> <em>This article was created using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/semaglutide-linked-cause-vision-loss-2024a1000cd7">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

Weight Loss Drugs Cut Cancer Risk in Diabetes Patients

Article Type
Changed
Mon, 07/08/2024 - 12:40

Recent research on popular weight loss drugs has uncovered surprising benefits beyond their intended use, like lowering the risk of fatal heart attacks. And now there may be another unforeseen advantage: People with type 2 diabetes who took these drugs had a lower risk of having 10 out of 13 obesity-related cancers, compared to those who used insulin therapy.

That’s according to a study published July 5 in JAMA Network Open where researchers studied glucagon-like peptide receptor agonists (known as GLP-1RAs), a class of drugs used to treat diabetes and obesity. Ozempic, Wegovy, Mounjaro, and Zepbound, which have become well-known recently because they are linked to rapid weight loss, contain GLP-1RAs.

For the study, they looked at electronic health records of 1.7 million patients who had type 2 diabetes, no prior diagnosis of obesity-related cancers, and had been prescribed GLP-1RAs, insulins, or metformin from March 2005 to November 2018.

The scientists found that compared to patients who took insulin, people who took GLP-1RAs had a “significant risk reduction” in 10 of 13 obesity-related cancers. Those 10 cancers were esophageal, colorectal, endometrial, gallbladder, kidney, liver, ovarian, and pancreatic cancers, as well as meningioma and multiple myeloma.

Compared with patients taking insulin, patients taking GLP-1RAs showed no statistically significant reduction in stomach cancer and no reduced risk of breast and thyroid cancers, the study said.

But the study found no decrease in cancer risk with GLP-1RAs compared with metformin.

While the study results suggest that these drugs may reduce the risk of certain obesity-related cancers better than insulins, more research is needed, they said.

A version of this article appeared on WebMD.com.

Publications
Topics
Sections

Recent research on popular weight loss drugs has uncovered surprising benefits beyond their intended use, like lowering the risk of fatal heart attacks. And now there may be another unforeseen advantage: People with type 2 diabetes who took these drugs had a lower risk of having 10 out of 13 obesity-related cancers, compared to those who used insulin therapy.

That’s according to a study published July 5 in JAMA Network Open where researchers studied glucagon-like peptide receptor agonists (known as GLP-1RAs), a class of drugs used to treat diabetes and obesity. Ozempic, Wegovy, Mounjaro, and Zepbound, which have become well-known recently because they are linked to rapid weight loss, contain GLP-1RAs.

For the study, they looked at electronic health records of 1.7 million patients who had type 2 diabetes, no prior diagnosis of obesity-related cancers, and had been prescribed GLP-1RAs, insulins, or metformin from March 2005 to November 2018.

The scientists found that compared to patients who took insulin, people who took GLP-1RAs had a “significant risk reduction” in 10 of 13 obesity-related cancers. Those 10 cancers were esophageal, colorectal, endometrial, gallbladder, kidney, liver, ovarian, and pancreatic cancers, as well as meningioma and multiple myeloma.

Compared with patients taking insulin, patients taking GLP-1RAs showed no statistically significant reduction in stomach cancer and no reduced risk of breast and thyroid cancers, the study said.

But the study found no decrease in cancer risk with GLP-1RAs compared with metformin.

While the study results suggest that these drugs may reduce the risk of certain obesity-related cancers better than insulins, more research is needed, they said.

A version of this article appeared on WebMD.com.

Recent research on popular weight loss drugs has uncovered surprising benefits beyond their intended use, like lowering the risk of fatal heart attacks. And now there may be another unforeseen advantage: People with type 2 diabetes who took these drugs had a lower risk of having 10 out of 13 obesity-related cancers, compared to those who used insulin therapy.

That’s according to a study published July 5 in JAMA Network Open where researchers studied glucagon-like peptide receptor agonists (known as GLP-1RAs), a class of drugs used to treat diabetes and obesity. Ozempic, Wegovy, Mounjaro, and Zepbound, which have become well-known recently because they are linked to rapid weight loss, contain GLP-1RAs.

For the study, they looked at electronic health records of 1.7 million patients who had type 2 diabetes, no prior diagnosis of obesity-related cancers, and had been prescribed GLP-1RAs, insulins, or metformin from March 2005 to November 2018.

The scientists found that compared to patients who took insulin, people who took GLP-1RAs had a “significant risk reduction” in 10 of 13 obesity-related cancers. Those 10 cancers were esophageal, colorectal, endometrial, gallbladder, kidney, liver, ovarian, and pancreatic cancers, as well as meningioma and multiple myeloma.

Compared with patients taking insulin, patients taking GLP-1RAs showed no statistically significant reduction in stomach cancer and no reduced risk of breast and thyroid cancers, the study said.

But the study found no decrease in cancer risk with GLP-1RAs compared with metformin.

While the study results suggest that these drugs may reduce the risk of certain obesity-related cancers better than insulins, more research is needed, they said.

A version of this article appeared on WebMD.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>168649</fileName> <TBEID>0C050E39.SIG</TBEID> <TBUniqueIdentifier>MD_0C050E39</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240708T122810</QCDate> <firstPublished>20240708T123625</firstPublished> <LastPublished>20240708T123625</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240708T123625</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Ralph Ellis</byline> <bylineText>RALPH ELLIS</bylineText> <bylineFull>RALPH ELLIS</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>People with type 2 diabetes who took these drugs had a lower risk of having 10 out of 13 obesity-related cancers, compared to those who used insulin therapy.</metaDescription> <articlePDF/> <teaserImage/> <teaser>Researchers examine electronic health records of patients who had type 2 diabetes, no prior diagnosis of obesity-related cancers, and had been prescribed GLP-1RAs, insulins, or metformin.</teaser> <title>Weight Loss Drugs Cut Cancer Risk in Diabetes Patients</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>oncr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>ob</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>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>hemn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>endo</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>chph</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>nr</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Neurology Reviews</journalTitle> <journalFullTitle>Neurology Reviews</journalFullTitle> <copyrightStatement>2018 Frontline Medical Communications Inc.,</copyrightStatement> </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>23</term> <term>21</term> <term>15</term> <term>13</term> <term>18</term> <term>34</term> <term>6</term> <term>22</term> </publications> <sections> <term canonical="true">27970</term> <term>39313</term> </sections> <topics> <term canonical="true">280</term> <term>192</term> <term>198</term> <term>61821</term> <term>59244</term> <term>67020</term> <term>214</term> <term>217</term> <term>221</term> <term>238</term> <term>240</term> <term>242</term> <term>244</term> <term>39570</term> <term>27442</term> <term>256</term> <term>245</term> <term>270</term> <term>31848</term> <term>292</term> <term>261</term> <term>206</term> <term>205</term> <term>263</term> <term>181</term> <term>179</term> <term>178</term> <term>59374</term> <term>195</term> <term>196</term> <term>197</term> <term>37637</term> <term>233</term> <term>243</term> <term>250</term> <term>49434</term> <term>303</term> <term>210</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Weight Loss Drugs Cut Cancer Risk in Diabetes Patients</title> <deck/> </itemMeta> <itemContent> <p>Recent research on popular weight loss drugs has uncovered surprising benefits beyond their intended use, like lowering the risk of fatal heart attacks. And now there may be another unforeseen advantage:<span class="tag metaDescription"> People with type 2 diabetes who took these drugs had a lower risk of having 10 out of 13 obesity-related cancers, compared to those who used insulin therapy.</span></p> <p>That’s according to a <span class="Hyperlink"><a href="https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2820833">study published</a></span> July 5 in <em>JAMA Network Open</em> where researchers studied glucagon-like peptide receptor agonists (known as GLP-1RAs), a class of drugs used to treat diabetes and obesity. Ozempic, Wegovy, Mounjaro, and Zepbound, which have become well-known recently because they are linked to rapid weight loss, contain GLP-1RAs.<br/><br/>For the study, they looked at electronic health records of 1.7 million patients who had type 2 diabetes, no prior diagnosis of obesity-related cancers, and had been prescribed GLP-1RAs, insulins, or metformin from March 2005 to November 2018.<br/><br/>The scientists found that compared to patients who took insulin, people who took GLP-1RAs had a “significant risk reduction” in 10 of 13 obesity-related cancers. Those 10 cancers were esophageal, colorectal, endometrial, gallbladder, kidney, liver, ovarian, and pancreatic cancers, as well as meningioma and multiple myeloma.<br/><br/>Compared with patients taking insulin, patients taking GLP-1RAs showed no statistically significant reduction in stomach cancer and no reduced risk of breast and thyroid cancers, the study said.<br/><br/>But the study found no decrease in cancer risk with GLP-1RAs compared with metformin. <br/><br/>While the study results suggest that these drugs may reduce the risk of certain obesity-related cancers better than insulins, more research is needed, they said.</p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.webmd.com/diabetes/news/20240705/weight-loss-drugs-lower-cancer-risk-in-type-2-diabetes-patients">WebMD.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

Can Response to Semaglutide Be Predicted With a Genetic Test?

Article Type
Changed
Mon, 07/08/2024 - 10:29

— An analysis of data from 137 patients suggested testing whether people have a trait known as abnormal postprandial satiety (APS), or hungry gut, can predict how well they may respond to the obesity drug semaglutide, although it failed to establish this link for the somewhat similar tirzepatide.

At the American Diabetes Association (ADA) Scientific Sessions, Maria Daniela Hurtado Andrade, MD, PhD, of the Mayo Clinic, Jacksonville, Florida, presented results of a study using the MyPhenome Hungry Gut test, which was developed through machine learning, a form of artificial intelligence. 

The test is part of the MyPhenome obesity phenotyping portfolio from Phenomix Sciences, a company founded by Mayo Clinic physicians, scientists, and researchers Andres Acosta, MD, PhD, and Michael Camilleri, MD, DSc. 

At the ADA meeting, Dr. Hurtado Andrade discussed a test of 137 adults: 91 were considered to have a positive biomarker for abnormal postprandial satiety (APS+), and 46 who did not have it were classified as APS−. These were patients of the Mayo Clinic who were already taking obesity drugs and agreed to phenotyping. Of this group, 113 were on semaglutide and 24 on tirzepatide.

At 12 months, among those taking semaglutide, patients classified as APS+ achieved a mean 18% body weight loss compared with 10% in those classified as APS−. But the test didn’t find these kinds of differences for the tirzepatide group, with a mean 19.4% body weight loss in the APS+ group and a mean loss of 22.1% in the APS− group.

Further studies are warranted to assess the clinical utility of these biomarkers, Dr. Hurtado Andrade said. But these findings do support “the use of precision medicine for obesity based on an individual’s genetic background,” she said.

Dr. Hurtado Andrade’s presentation impressed fellow researchers who noted it as an early step toward the long-sought goal of more personalized medicine.

Daniel S. Hsia, MD, of Emory University, Atlanta, who led the ADA session at which Dr. Hurtado Andrade presented, said it was good to see new information being presented about using genetic risk scoring in obesity.

“The numbers were very small for the tirzepatide group as compared to the semaglutide group, so it’s a little hard to really come to any significant conclusions,” Dr. Hsia said in an interview.

At the ADA meeting, Ajay D. Rao, MD, MMSc, of Temple University, Philadelphia, said clinicians are excited about the idea of having biomarkers to aid in decisions about approaches to obesity.

In a follow-up interview with this news organization, Dr. Rao said he too is looking to see more testing of this approach to care, while describing Hurtado Andrade’s work as a “very well-done study.”

“We still need to see more large-scale studies of responsiveness to certain interventions,” he said. 

Dr. Hurtado Andrade noted that researchers at academic centers such as Mayo can try to hone in the combination of genetic and other factors that led to obesity, such as emotional eating patterns and abnormal postprandial satiety. 

But this approach is not widely scalable, as it demands resources of time and staffing that not all clinicians and patients enjoy.

“To overcome this challenge, our team has been working on developing biomarkers” such as the machine-learning gene risk score used to predict abnormal postprandial satiety, she said.

Findings for a related project were presented in May at Digestive Disease Week, as this news organization reported earlier. In that study, researchers calculated the genetic risk score for 84 adults undergoing weight loss interventions at Mayo Clinic who were prescribed the glucagon-like peptide 1 receptor agonist semaglutide.

This news organization separately asked Phenomix about the sales of MyPhenome Test kits. These cost $499, and about 500 tests have been sold since commercialization started last year, a spokesperson said.

The study was funded by Phenomix Sciences. Separately, Dr. Hurtado Andrade has worked as a consultant for Novo Nordisk and received research support from the National Institutes of Health. 

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

— An analysis of data from 137 patients suggested testing whether people have a trait known as abnormal postprandial satiety (APS), or hungry gut, can predict how well they may respond to the obesity drug semaglutide, although it failed to establish this link for the somewhat similar tirzepatide.

At the American Diabetes Association (ADA) Scientific Sessions, Maria Daniela Hurtado Andrade, MD, PhD, of the Mayo Clinic, Jacksonville, Florida, presented results of a study using the MyPhenome Hungry Gut test, which was developed through machine learning, a form of artificial intelligence. 

The test is part of the MyPhenome obesity phenotyping portfolio from Phenomix Sciences, a company founded by Mayo Clinic physicians, scientists, and researchers Andres Acosta, MD, PhD, and Michael Camilleri, MD, DSc. 

At the ADA meeting, Dr. Hurtado Andrade discussed a test of 137 adults: 91 were considered to have a positive biomarker for abnormal postprandial satiety (APS+), and 46 who did not have it were classified as APS−. These were patients of the Mayo Clinic who were already taking obesity drugs and agreed to phenotyping. Of this group, 113 were on semaglutide and 24 on tirzepatide.

At 12 months, among those taking semaglutide, patients classified as APS+ achieved a mean 18% body weight loss compared with 10% in those classified as APS−. But the test didn’t find these kinds of differences for the tirzepatide group, with a mean 19.4% body weight loss in the APS+ group and a mean loss of 22.1% in the APS− group.

Further studies are warranted to assess the clinical utility of these biomarkers, Dr. Hurtado Andrade said. But these findings do support “the use of precision medicine for obesity based on an individual’s genetic background,” she said.

Dr. Hurtado Andrade’s presentation impressed fellow researchers who noted it as an early step toward the long-sought goal of more personalized medicine.

Daniel S. Hsia, MD, of Emory University, Atlanta, who led the ADA session at which Dr. Hurtado Andrade presented, said it was good to see new information being presented about using genetic risk scoring in obesity.

“The numbers were very small for the tirzepatide group as compared to the semaglutide group, so it’s a little hard to really come to any significant conclusions,” Dr. Hsia said in an interview.

At the ADA meeting, Ajay D. Rao, MD, MMSc, of Temple University, Philadelphia, said clinicians are excited about the idea of having biomarkers to aid in decisions about approaches to obesity.

In a follow-up interview with this news organization, Dr. Rao said he too is looking to see more testing of this approach to care, while describing Hurtado Andrade’s work as a “very well-done study.”

“We still need to see more large-scale studies of responsiveness to certain interventions,” he said. 

Dr. Hurtado Andrade noted that researchers at academic centers such as Mayo can try to hone in the combination of genetic and other factors that led to obesity, such as emotional eating patterns and abnormal postprandial satiety. 

But this approach is not widely scalable, as it demands resources of time and staffing that not all clinicians and patients enjoy.

“To overcome this challenge, our team has been working on developing biomarkers” such as the machine-learning gene risk score used to predict abnormal postprandial satiety, she said.

Findings for a related project were presented in May at Digestive Disease Week, as this news organization reported earlier. In that study, researchers calculated the genetic risk score for 84 adults undergoing weight loss interventions at Mayo Clinic who were prescribed the glucagon-like peptide 1 receptor agonist semaglutide.

This news organization separately asked Phenomix about the sales of MyPhenome Test kits. These cost $499, and about 500 tests have been sold since commercialization started last year, a spokesperson said.

The study was funded by Phenomix Sciences. Separately, Dr. Hurtado Andrade has worked as a consultant for Novo Nordisk and received research support from the National Institutes of Health. 

A version of this article first appeared on Medscape.com.

— An analysis of data from 137 patients suggested testing whether people have a trait known as abnormal postprandial satiety (APS), or hungry gut, can predict how well they may respond to the obesity drug semaglutide, although it failed to establish this link for the somewhat similar tirzepatide.

At the American Diabetes Association (ADA) Scientific Sessions, Maria Daniela Hurtado Andrade, MD, PhD, of the Mayo Clinic, Jacksonville, Florida, presented results of a study using the MyPhenome Hungry Gut test, which was developed through machine learning, a form of artificial intelligence. 

The test is part of the MyPhenome obesity phenotyping portfolio from Phenomix Sciences, a company founded by Mayo Clinic physicians, scientists, and researchers Andres Acosta, MD, PhD, and Michael Camilleri, MD, DSc. 

At the ADA meeting, Dr. Hurtado Andrade discussed a test of 137 adults: 91 were considered to have a positive biomarker for abnormal postprandial satiety (APS+), and 46 who did not have it were classified as APS−. These were patients of the Mayo Clinic who were already taking obesity drugs and agreed to phenotyping. Of this group, 113 were on semaglutide and 24 on tirzepatide.

At 12 months, among those taking semaglutide, patients classified as APS+ achieved a mean 18% body weight loss compared with 10% in those classified as APS−. But the test didn’t find these kinds of differences for the tirzepatide group, with a mean 19.4% body weight loss in the APS+ group and a mean loss of 22.1% in the APS− group.

Further studies are warranted to assess the clinical utility of these biomarkers, Dr. Hurtado Andrade said. But these findings do support “the use of precision medicine for obesity based on an individual’s genetic background,” she said.

Dr. Hurtado Andrade’s presentation impressed fellow researchers who noted it as an early step toward the long-sought goal of more personalized medicine.

Daniel S. Hsia, MD, of Emory University, Atlanta, who led the ADA session at which Dr. Hurtado Andrade presented, said it was good to see new information being presented about using genetic risk scoring in obesity.

“The numbers were very small for the tirzepatide group as compared to the semaglutide group, so it’s a little hard to really come to any significant conclusions,” Dr. Hsia said in an interview.

At the ADA meeting, Ajay D. Rao, MD, MMSc, of Temple University, Philadelphia, said clinicians are excited about the idea of having biomarkers to aid in decisions about approaches to obesity.

In a follow-up interview with this news organization, Dr. Rao said he too is looking to see more testing of this approach to care, while describing Hurtado Andrade’s work as a “very well-done study.”

“We still need to see more large-scale studies of responsiveness to certain interventions,” he said. 

Dr. Hurtado Andrade noted that researchers at academic centers such as Mayo can try to hone in the combination of genetic and other factors that led to obesity, such as emotional eating patterns and abnormal postprandial satiety. 

But this approach is not widely scalable, as it demands resources of time and staffing that not all clinicians and patients enjoy.

“To overcome this challenge, our team has been working on developing biomarkers” such as the machine-learning gene risk score used to predict abnormal postprandial satiety, she said.

Findings for a related project were presented in May at Digestive Disease Week, as this news organization reported earlier. In that study, researchers calculated the genetic risk score for 84 adults undergoing weight loss interventions at Mayo Clinic who were prescribed the glucagon-like peptide 1 receptor agonist semaglutide.

This news organization separately asked Phenomix about the sales of MyPhenome Test kits. These cost $499, and about 500 tests have been sold since commercialization started last year, a spokesperson said.

The study was funded by Phenomix Sciences. Separately, Dr. Hurtado Andrade has worked as a consultant for Novo Nordisk and received research support from the National Institutes of Health. 

A version of this article first 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>168626</fileName> <TBEID>0C050DF2.SIG</TBEID> <TBUniqueIdentifier>MD_0C050DF2</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240708T101812</QCDate> <firstPublished>20240708T102631</firstPublished> <LastPublished>20240708T102632</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240708T102631</CMSDate> <articleSource>FROM ADA 2024</articleSource> <facebookInfo/> <meetingNumber/> <byline>Kerry Dooley Young</byline> <bylineText>KERRY DOOLEY YOUNG</bylineText> <bylineFull>KERRY DOOLEY YOUNG</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>At 12 months, among those taking semaglutide, patients classified as APS+ achieved a mean 18% body weight loss compared with 10% in those classified as APS−. Bu</metaDescription> <articlePDF/> <teaserImage/> <teaser>AI genetic testing may help identify patients who would benefit from semaglutide, by identifying abnormal postprandial satiety.</teaser> <title>Can Response to Semaglutide Be Predicted With a Genetic Test?</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>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>dbh</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> </publications_g> <publications> <term canonical="true">34</term> <term>15</term> <term>21</term> <term>358</term> </publications> <sections> <term canonical="true">39313</term> <term>27970</term> </sections> <topics> <term canonical="true">205</term> <term>261</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Can Response to Semaglutide Be Predicted With a Genetic Test?</title> <deck/> </itemMeta> <itemContent> <p><span class="dateline">ORLANDO, FLORIDA</span> — An analysis of data from 137 patients suggested testing whether people have a trait known as abnormal postprandial satiety (APS), or hungry gut, can predict how well they may respond to the obesity drug semaglutide, although it failed to establish this link for the somewhat similar tirzepatide.</p> <p>At the <a href="https://www.medscape.com/viewcollection/37466">American Diabetes Association (ADA) Scientific Sessions</a>, Maria Daniela Hurtado Andrade, MD, PhD, of the Mayo Clinic, Jacksonville, Florida, presented results of a study using the MyPhenome Hungry Gut test, which was developed through machine learning, a form of artificial intelligence. <br/><br/>The test is part of the MyPhenome obesity phenotyping portfolio from Phenomix Sciences, a company founded by Mayo Clinic physicians, scientists, and researchers Andres Acosta, MD, PhD, and Michael Camilleri, MD, DSc. <br/><br/>At the ADA meeting, Dr. Hurtado Andrade discussed a test of 137 adults: 91 were considered to have a positive biomarker for abnormal postprandial satiety (APS+), and 46 who did not have it were classified as APS−. These were patients of the Mayo Clinic who were already taking obesity drugs and agreed to phenotyping. Of this group, 113 were on semaglutide and 24 on tirzepatide.<br/><br/><span class="tag metaDescription">At 12 months, among those taking semaglutide, patients classified as APS+ achieved a mean 18% body weight loss compared with 10% in those classified as APS−. But the test didn’t find these kinds of differences for the tirzepatide group</span>, with a mean 19.4% body weight loss in the APS+ group and a mean loss of 22.1% in the APS− group.<br/><br/>Further studies are warranted to assess the clinical utility of these biomarkers, Dr. Hurtado Andrade said. But these findings do support “the use of precision medicine for obesity based on an individual’s genetic background,” she said.<br/><br/>Dr. Hurtado Andrade’s presentation impressed fellow researchers who noted it as an early step toward the long-sought goal of more personalized medicine.<br/><br/>Daniel S. Hsia, MD, of Emory University, Atlanta, who led the ADA session at which Dr. Hurtado Andrade presented, said it was good to see new information being presented about using genetic risk scoring in obesity.<br/><br/>“The numbers were very small for the tirzepatide group as compared to the semaglutide group, so it’s a little hard to really come to any significant conclusions,” Dr. Hsia said in an interview.<br/><br/>At the ADA meeting, Ajay D. Rao, MD, MMSc, of Temple University, Philadelphia, said clinicians are excited about the idea of having biomarkers to aid in decisions about approaches to obesity.<br/><br/>In a follow-up interview with this news organization, Dr. Rao said he too is looking to see more testing of this approach to care, while describing Hurtado Andrade’s work as a “very well-done study.”<br/><br/>“We still need to see more large-scale studies of responsiveness to certain interventions,” he said. <br/><br/>Dr. Hurtado Andrade noted that researchers at academic centers such as Mayo can try to hone in the combination of genetic and other factors that led to obesity, such as emotional eating patterns and abnormal postprandial satiety. <br/><br/>But this approach is not widely scalable, as it demands resources of time and staffing that not all clinicians and patients enjoy.<br/><br/>“To overcome this challenge, our team has been working on developing biomarkers” such as the machine-learning gene risk score used to predict abnormal postprandial satiety, she said.<br/><br/>Findings for a related project were presented in May at Digestive Disease Week, as this news organization <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/genetic-test-can-predict-response-semaglutide-weight-loss-2024a10009k6">reported earlier</a></span>. In that study, researchers calculated the genetic risk score for 84 adults undergoing weight loss interventions at Mayo Clinic who were prescribed the glucagon-like peptide 1 receptor agonist semaglutide.<br/><br/>This news organization separately asked Phenomix about the sales of MyPhenome Test kits. These cost $499, and about 500 tests have been sold since commercialization started last year, a spokesperson said.<br/><br/>The study was funded by Phenomix Sciences. Separately, Dr. Hurtado Andrade has worked as a consultant for Novo Nordisk and received research support from the National Institutes of Health. </p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/can-genetic-test-predict-response-semaglutide-2024a1000cat">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 ADA 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

Should South Park: The End of Obesity Be Required Viewing in Medical School?

Article Type
Changed
Wed, 07/03/2024 - 13:00
Display Headline
Should South Park: The End of Obesity Be Required Viewing in Medical School?

Yes, there’s still much to find offensive, but South Park: The End of Obesity, in just 51 minutes, does more to explain some of obesity’s realities, its pharmacotherapy, and weight bias than the mainstream media has done perhaps ever. 

The mini-movie follows the plight of Eric Cartman, the fictional South Parkian child with severe obesity. 

South Park got everything right. The movie starts in a medical center where discussions with Cartman, his mother, and his doctor make it clear that obesity isn’t something that Cartman chose and is perhaps the most distressing aspect of his life. This certainly echoes study findings which report that quality-of-life scores in children with severe obesity are lower than those of children with newly diagnosed on-treatment cancers. As to how obesity erodes a child’s quality of life, no doubt part of its impact stems from obesity being a top source of schoolyard bullying, which is reflected by Cartman as he imagines his life without it. 

Cartman’s mother explains that of course they’ve tried diet and exercise, but that intentional behavior change alone hasn’t been sufficient to sustainably move the scale’s needle — a truth for the vast majority of people with obesity. But here, unlike in many actual doctors’ offices, Cartman’s doctor doesn’t spend time doubting or cajoling; instead, he does his job — which is to inform his patient, without judgment, about a pharmaceutical option that has proved to be beneficial. He accurately describes these medications as ushering in “a whole new era of medicine, a miracle really” that can “help people lose vast amounts of weight.”

The kicker, though, comes next. The doctor explains that insurance companies cover the medications only for patients with diabetes, “so if you can’t afford them, you’re just kind of out of luck.” This is changing somewhat now, at least here in Canada, where two of our main private insurers have changed their base coverages to make antiobesity medications something employers need to opt out of rather than opt into, but certainly they’re not covered by US Medicare for weight management, nor by our version of the same here in Canada.

But even for those who have coverage, there are hoops to jump through, which is highlighted by the incredible efforts made by Cartman and his friends to get his insurance plan to cover the medications. Thwarted at every turn, despite the undeniable benefits of these medications to health and quality of life, they are forced to turn to compounding — a phenomenon certainly pervasive here in North America whereby compounding pharmacies claim to be able to provide glucagon-like peptide-1 (GLP-1) analogs with comparable efficacy at a fraction of the price, but without the same rigor of proof of purity or efficacy. 

Also covered by South Park is that the GLP-1 analog supply is impacted by use by people who don’t meet approved medical criteria and are using the medications for aesthetic purposes. This speaks to the incredible societal pressure to be thin and to the comfort of some physicians to inappropriately prescribe these medications. This is covered by the subplot of South Park’s weed farmer, Randy, who in turn delivers an important insight into how it feels to use a GLP-1 analog: “I think there’s something wrong with these drugs ... I feel satisfied. With any drugs I want to do more and more, but with these drugs I feel like I want things less. With these drugs you don’t really crave anything.” The sentiment is echoed by Cartman, who exclaims, “I think I’m full. I’ve never known that feeling before in my life, but I’m full.”

It’s remarkable that South Park, a show built on serving up politically incorrect offense, covers obesity and its treatment with more accuracy, nuance, and compassion than does society as a whole. The show notes that obesity is a biological condition (it is), that when it comes to health (in America) “you have to have some f-ing willpower.” But where they explicitly mean having willpower in terms of filing and pursing insurance claims (you do), explains that drug companies are making antiobesity medications more expensive in America than anywhere else in the world (they are), and finally delivers this quote, which, while missing the biological basis of behavior and hunger with respect to obesity, certainly sums up why blame has no place in the discourse:

“We have sugar companies, pharmaceutical companies, and insurance companies all just trying to figure out how to make money off our health. It isn’t fair to put the blame on anyone for their weight.”

No, it’s not.

This movie should be required viewing in medical schools.
 

Dr. Freedhoff is associate professor, department of family medicine, University of Ottawa, and medical director, Bariatric Medical Institute, Ottawa, Ontario, Canada. He disclosed ties with Bariatric Medical Institute, Constant Health, Novo Nordisk, and Weighty Matters.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

Yes, there’s still much to find offensive, but South Park: The End of Obesity, in just 51 minutes, does more to explain some of obesity’s realities, its pharmacotherapy, and weight bias than the mainstream media has done perhaps ever. 

The mini-movie follows the plight of Eric Cartman, the fictional South Parkian child with severe obesity. 

South Park got everything right. The movie starts in a medical center where discussions with Cartman, his mother, and his doctor make it clear that obesity isn’t something that Cartman chose and is perhaps the most distressing aspect of his life. This certainly echoes study findings which report that quality-of-life scores in children with severe obesity are lower than those of children with newly diagnosed on-treatment cancers. As to how obesity erodes a child’s quality of life, no doubt part of its impact stems from obesity being a top source of schoolyard bullying, which is reflected by Cartman as he imagines his life without it. 

Cartman’s mother explains that of course they’ve tried diet and exercise, but that intentional behavior change alone hasn’t been sufficient to sustainably move the scale’s needle — a truth for the vast majority of people with obesity. But here, unlike in many actual doctors’ offices, Cartman’s doctor doesn’t spend time doubting or cajoling; instead, he does his job — which is to inform his patient, without judgment, about a pharmaceutical option that has proved to be beneficial. He accurately describes these medications as ushering in “a whole new era of medicine, a miracle really” that can “help people lose vast amounts of weight.”

The kicker, though, comes next. The doctor explains that insurance companies cover the medications only for patients with diabetes, “so if you can’t afford them, you’re just kind of out of luck.” This is changing somewhat now, at least here in Canada, where two of our main private insurers have changed their base coverages to make antiobesity medications something employers need to opt out of rather than opt into, but certainly they’re not covered by US Medicare for weight management, nor by our version of the same here in Canada.

But even for those who have coverage, there are hoops to jump through, which is highlighted by the incredible efforts made by Cartman and his friends to get his insurance plan to cover the medications. Thwarted at every turn, despite the undeniable benefits of these medications to health and quality of life, they are forced to turn to compounding — a phenomenon certainly pervasive here in North America whereby compounding pharmacies claim to be able to provide glucagon-like peptide-1 (GLP-1) analogs with comparable efficacy at a fraction of the price, but without the same rigor of proof of purity or efficacy. 

Also covered by South Park is that the GLP-1 analog supply is impacted by use by people who don’t meet approved medical criteria and are using the medications for aesthetic purposes. This speaks to the incredible societal pressure to be thin and to the comfort of some physicians to inappropriately prescribe these medications. This is covered by the subplot of South Park’s weed farmer, Randy, who in turn delivers an important insight into how it feels to use a GLP-1 analog: “I think there’s something wrong with these drugs ... I feel satisfied. With any drugs I want to do more and more, but with these drugs I feel like I want things less. With these drugs you don’t really crave anything.” The sentiment is echoed by Cartman, who exclaims, “I think I’m full. I’ve never known that feeling before in my life, but I’m full.”

It’s remarkable that South Park, a show built on serving up politically incorrect offense, covers obesity and its treatment with more accuracy, nuance, and compassion than does society as a whole. The show notes that obesity is a biological condition (it is), that when it comes to health (in America) “you have to have some f-ing willpower.” But where they explicitly mean having willpower in terms of filing and pursing insurance claims (you do), explains that drug companies are making antiobesity medications more expensive in America than anywhere else in the world (they are), and finally delivers this quote, which, while missing the biological basis of behavior and hunger with respect to obesity, certainly sums up why blame has no place in the discourse:

“We have sugar companies, pharmaceutical companies, and insurance companies all just trying to figure out how to make money off our health. It isn’t fair to put the blame on anyone for their weight.”

No, it’s not.

This movie should be required viewing in medical schools.
 

Dr. Freedhoff is associate professor, department of family medicine, University of Ottawa, and medical director, Bariatric Medical Institute, Ottawa, Ontario, Canada. He disclosed ties with Bariatric Medical Institute, Constant Health, Novo Nordisk, and Weighty Matters.

A version of this article appeared on Medscape.com.

Yes, there’s still much to find offensive, but South Park: The End of Obesity, in just 51 minutes, does more to explain some of obesity’s realities, its pharmacotherapy, and weight bias than the mainstream media has done perhaps ever. 

The mini-movie follows the plight of Eric Cartman, the fictional South Parkian child with severe obesity. 

South Park got everything right. The movie starts in a medical center where discussions with Cartman, his mother, and his doctor make it clear that obesity isn’t something that Cartman chose and is perhaps the most distressing aspect of his life. This certainly echoes study findings which report that quality-of-life scores in children with severe obesity are lower than those of children with newly diagnosed on-treatment cancers. As to how obesity erodes a child’s quality of life, no doubt part of its impact stems from obesity being a top source of schoolyard bullying, which is reflected by Cartman as he imagines his life without it. 

Cartman’s mother explains that of course they’ve tried diet and exercise, but that intentional behavior change alone hasn’t been sufficient to sustainably move the scale’s needle — a truth for the vast majority of people with obesity. But here, unlike in many actual doctors’ offices, Cartman’s doctor doesn’t spend time doubting or cajoling; instead, he does his job — which is to inform his patient, without judgment, about a pharmaceutical option that has proved to be beneficial. He accurately describes these medications as ushering in “a whole new era of medicine, a miracle really” that can “help people lose vast amounts of weight.”

The kicker, though, comes next. The doctor explains that insurance companies cover the medications only for patients with diabetes, “so if you can’t afford them, you’re just kind of out of luck.” This is changing somewhat now, at least here in Canada, where two of our main private insurers have changed their base coverages to make antiobesity medications something employers need to opt out of rather than opt into, but certainly they’re not covered by US Medicare for weight management, nor by our version of the same here in Canada.

But even for those who have coverage, there are hoops to jump through, which is highlighted by the incredible efforts made by Cartman and his friends to get his insurance plan to cover the medications. Thwarted at every turn, despite the undeniable benefits of these medications to health and quality of life, they are forced to turn to compounding — a phenomenon certainly pervasive here in North America whereby compounding pharmacies claim to be able to provide glucagon-like peptide-1 (GLP-1) analogs with comparable efficacy at a fraction of the price, but without the same rigor of proof of purity or efficacy. 

Also covered by South Park is that the GLP-1 analog supply is impacted by use by people who don’t meet approved medical criteria and are using the medications for aesthetic purposes. This speaks to the incredible societal pressure to be thin and to the comfort of some physicians to inappropriately prescribe these medications. This is covered by the subplot of South Park’s weed farmer, Randy, who in turn delivers an important insight into how it feels to use a GLP-1 analog: “I think there’s something wrong with these drugs ... I feel satisfied. With any drugs I want to do more and more, but with these drugs I feel like I want things less. With these drugs you don’t really crave anything.” The sentiment is echoed by Cartman, who exclaims, “I think I’m full. I’ve never known that feeling before in my life, but I’m full.”

It’s remarkable that South Park, a show built on serving up politically incorrect offense, covers obesity and its treatment with more accuracy, nuance, and compassion than does society as a whole. The show notes that obesity is a biological condition (it is), that when it comes to health (in America) “you have to have some f-ing willpower.” But where they explicitly mean having willpower in terms of filing and pursing insurance claims (you do), explains that drug companies are making antiobesity medications more expensive in America than anywhere else in the world (they are), and finally delivers this quote, which, while missing the biological basis of behavior and hunger with respect to obesity, certainly sums up why blame has no place in the discourse:

“We have sugar companies, pharmaceutical companies, and insurance companies all just trying to figure out how to make money off our health. It isn’t fair to put the blame on anyone for their weight.”

No, it’s not.

This movie should be required viewing in medical schools.
 

Dr. Freedhoff is associate professor, department of family medicine, University of Ottawa, and medical director, Bariatric Medical Institute, Ottawa, Ontario, Canada. He disclosed ties with Bariatric Medical Institute, Constant Health, Novo Nordisk, and Weighty Matters.

A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Display Headline
Should South Park: The End of Obesity Be Required Viewing in Medical School?
Display Headline
Should South Park: The End of Obesity Be Required Viewing in Medical School?
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>168590</fileName> <TBEID>0C050D1E.SIG</TBEID> <TBUniqueIdentifier>MD_0C050D1E</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>353</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240703T124613</QCDate> <firstPublished>20240703T124954</firstPublished> <LastPublished>20240703T124954</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240703T124954</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Yoni Freedhoff, MD</byline> <bylineText>YONI FREEDHOFF, MD</bylineText> <bylineFull>YONI FREEDHOFF, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <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>Yes, there’s still much to find offensive, but South Park: The End of Obesity, in just 51 minutes, does more to explain some of obesity’s realities, its pharmac</metaDescription> <articlePDF/> <teaserImage/> <teaser>It’s remarkable that <span class="Emphasis">South Park</span>, a show built on serving up politically incorrect offense, covers obesity and its treatment with more accuracy, nuance, and compassion than does society as a whole.</teaser> <title>Should South Park: The End of Obesity Be Required Viewing in Medical School?</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>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> </publications_g> <publications> <term canonical="true">34</term> <term>15</term> <term>21</term> </publications> <sections> <term canonical="true">52</term> </sections> <topics> <term>38029</term> <term canonical="true">261</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Should South Park: The End of Obesity Be Required Viewing in Medical School?</title> <deck/> </itemMeta> <itemContent> <p>Yes, there’s still much to find offensive, but <span class="Emphasis">South Park: The End of Obesity</span>, in just 51 minutes, does more to explain some of <span class="Hyperlink">obesity</span>’s realities, its pharmacotherapy, and weight bias than the mainstream media has done perhaps ever. </p> <p>The mini-movie follows the plight of Eric Cartman, the fictional South Parkian child with severe obesity. <br/><br/><span class="Emphasis">South Park </span>got everything right. The movie starts in a medical center where discussions with Cartman, his mother, and his doctor make it clear that obesity isn’t something that Cartman chose and is perhaps the most distressing aspect of his life. This certainly echoes study findings which report that quality-of-life scores in children with severe obesity <span class="Hyperlink"><a href="https://link.springer.com/article/10.1186/1477-7525-5-43">are lower than those of children with newly diagnosed on-treatment cancers</a></span>. As to how obesity erodes a child’s quality of life, no doubt part of its impact stems from obesity <span class="Hyperlink"><a href="https://www.nature.com/articles/ijo2014117">being a top source of schoolyard bullying</a></span>, which is reflected by Cartman as he imagines his life without it. <br/><br/>Cartman’s mother explains that of course they’ve tried diet and exercise, but that intentional behavior change alone hasn’t been sufficient to sustainably move the scale’s needle — a truth for the vast majority of people with obesity. But here, unlike in many actual doctors’ offices, Cartman’s doctor doesn’t spend time doubting or cajoling; instead, he does his job — which is to inform his patient, without judgment, about a pharmaceutical option that has proved to be beneficial. He accurately describes these medications as ushering in “a whole new era of medicine, a miracle really” that can “help people lose vast amounts of weight.”<br/><br/>The kicker, though, comes next. The doctor explains that insurance companies cover the medications only for patients with diabetes, “so if you can’t afford them, you’re just kind of out of luck.” This is changing somewhat now, at least here in Canada, where two of our main private insurers have changed their base coverages to make antiobesity medications something employers need to opt out of rather than opt into, but certainly they’re not covered by US Medicare for weight management, nor by our version of the same here in Canada.<br/><br/>But even for those who have coverage, there are hoops to jump through, which is highlighted by the incredible efforts made by Cartman and his friends to get his insurance plan to cover the medications. Thwarted at every turn, despite the undeniable benefits of these medications to health and quality of life, they are forced to turn to compounding — a phenomenon certainly pervasive here in North America whereby compounding pharmacies claim to be able to provide <span class="Hyperlink">glucagon</span>-like peptide-1 (GLP-1) analogs with comparable efficacy at a fraction of the price, but without the same rigor of proof of purity or efficacy. <br/><br/>Also covered by <span class="Emphasis">South Park</span> is that the GLP-1 analog supply is impacted by use by people who don’t meet approved medical criteria and are using the medications for aesthetic purposes. This speaks to the incredible societal pressure to be thin and to the comfort of some physicians to inappropriately prescribe these medications. This is covered by the subplot of South Park’s weed farmer, Randy, who in turn delivers an important insight into how it feels to use a GLP-1 analog: “I think there’s something wrong with these drugs ... I feel satisfied. With any drugs I want to do more and more, but with these drugs I feel like I want things less. With these drugs you don’t really crave anything.” The sentiment is echoed by Cartman, who exclaims, “I think I’m full. I’ve never known that feeling before in my life, but I’m full.”<br/><br/>It’s remarkable that <span class="Emphasis">South Park</span>, a show built on serving up politically incorrect offense, covers obesity and its treatment with more accuracy, nuance, and compassion than does society as a whole. The show notes that obesity is a biological condition (it is), that when it comes to health (in America) “you have to have some f-ing willpower.” But where they explicitly mean having willpower in terms of filing and pursing insurance claims (you do), explains that drug companies are making antiobesity medications more expensive in America than anywhere else in the world (they are), and finally delivers this quote, which, while missing the biological basis of behavior and hunger with respect to obesity, certainly sums up why blame has no place in the discourse:<br/><br/>“We have sugar companies, pharmaceutical companies, and insurance companies all just trying to figure out how to make money off our health. It isn’t fair to put the blame on anyone for their weight.”<br/><br/>No, it’s not.<br/><br/>This movie should be required viewing in medical schools.<br/><br/></p> <p> <em>Dr. Freedhoff is associate professor, department of family medicine, University of Ottawa, and medical director, Bariatric Medical Institute, Ottawa, Ontario, Canada. He disclosed ties with Bariatric Medical Institute, Constant Health, Novo Nordisk, and Weighty Matters.</em> </p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/should-south-park-end-obesity-be-required-viewing-medical-2024a1000by5">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

What Should Be Prioritized in Managing Early Diabetes?

Article Type
Changed
Wed, 07/03/2024 - 10:54

— What to prioritize first in managing early diabetes? That was the question debated on an expert panel at the American Diabetes Association (ADA) 84th Scientific Sessions, with impassioned responses ranging from a plea to “treat obesity first,” to a James Carville–inspired counterpoint of “it’s the glucose, stupid.” 

With a focus on preventing complications and inducing remission rounding out the four positions argued, Session Moderator Ravi Retnakaran, MD, of the University of Toronto, noted that the options reflect the tricky choices clinicians treating patients with diabetes are pressed to make on a daily basis.

“In clinical decision-making [for early diabetes], we are faced with weighing each of these variables for the individual patient, and while all are good options, strong arguments can be made for prioritizing each — with the potential of each choice to influence or improve all of the others,” Dr. Retnakaran told this news organization.
 

Which to Prioritize First?

Making the obesity first argument, Ania M. Jastreboff, MD, PhD, associate professor and director of the Yale Obesity Research Center at Yale School of Medicine, New Haven, Connecticut, noted the striking statistic that nearly 90% of people with type 2 diabetes have overweight or obesity and discussed the ever-expanding data showing the benefits of drugs including glucagon-like peptide 1 (GLP-1) receptor agonists not just in weight loss but also in kidney, cardiovascular, and, as presented at the meeting, sleep apnea improvement.

She contrasted the experiences of two patients with obesity: One treated for the obesity upon type 2 diagnosis — who had a quick normalization of lipids and hypertension soon after the obesity treatment — and the other presenting after 10 years with type 2 diabetes — who was on therapy for hypertension and hyperlipidemia but not for obesity and whose diseases were not as easily treated by that point.

“Why are we treating all the downstream effects and we’re not treating the disease that is potentially the root cause of all these other diseases?” Dr. Jastreboff said.
 

Complications?

Arguing in favor of focusing on complications, Roopa Mehta, MD, PhD, with the department of endocrinology and metabolism at Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, made the case that stakes don’t get any higher in diabetes than when it comes the looming threat of potentially fatal complications.

Acute myocardial infarction, stroke, amputation, and end-stage renal disease are all on the list of unwanted outcomes and need to be considered even in the earliest stages, as data show early onset type 2 diabetes is linked to life expectancy.

“The main goal of management has always been to prevent complications,” she noted. Citing ADA guidelines, Dr. Mehta underscored the benefits of first- and second-line therapy of metformin, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and GLP-1 receptor agonists for most patients.
 

Remission?

Discussing the priority of putting patients into disease remission, Roy Taylor, MD, professor of medicine and metabolism at Newcastle University and Newcastle Hospitals NHS in Newcastle upon Tyne, England, and author of the book Life Without Diabetes, focused on an evidence-based alternative to achieving remission — a nonpharmacologic approach that avoids costly and sometimes inaccessible drugs.

In the intervention, described in the DiRECT randomized trial and subsequently in the UK National Health Service Type 2 Diabetes Path to Remission Program, patients with overweight or obesity were placed on a highly restrictive diet of just 800-900 calories a day for 12-20 weeks, followed by maintenance for 12 months, and they not only achieved weight loss but also achieved diabetes remission, in some cases long term.

Acknowledging that “this is not for everyone,” Dr. Taylor asserted that “we have to realize there is a substantial minority of people who want to be healthy but who don’t want to be medicalized,” he said.

“They want their health, and they can do extremely well.”
 

Glucose?

In taking his self-titled “it’s the glucose, stupid” stand, David M. Nathan, MD, of the Diabetes Center, Massachusetts General Hospital, Harvard Medical School, in Boston, cited extensive evidence showing that early intensive blood glucose control with treatment including sulfonylureas, insulin, or metformin significantly reduced the risk for complications in type 2 diabetes 15 or more years later, including renal failure, blindness, amputation, and myocardial infarctions, in addition to a reduction in diabetes-related death.

“In many of these studies, you saw the benefit even in the setting of weight-gain,” Dr. Nathan underscored.

He further noted the “sobering” findings of the Look AHEAD study, which had to be stopped due to futility when an intensive lifestyle/weight loss intervention showed no significant benefits in terms of cardiovascular disease in people with type 2 diabetes at a median follow-up of 9.6 years.

Ultimately, “diabetes, type 1 and type 2, remains a gluco-centric disease,” Dr. Nathan asserted. “Hyperglycemia is the only universal link between all forms of diabetes and mortality, and the long-term complications of diabetes are intimately associated with hyperglycemia.”
 

Tackling the Caveats

The ensuing panel discussion did not fail to deliver in delving into key areas of contention, particularly in terms of GLP-1 treatment.

Regarding a lack of data on the potential long-term effects of GLP-1s: “Yes, there are a huge number of studies [on GLP-1 receptor agonists], but they are, in general, over short periods of time and driven by pharma, who get in and get out as quickly as they can and have little in the way of interest to do comparative effectiveness studies,” Dr. Nathan argued.

“Meanwhile, this is like the crack cocaine of medications — patients have to stay on it for a lifetime or they will regain the weight — are you concerned at all about a lifetime of exposure to GLP-1 [drugs]?” he asked the panel.

Dr. Jastreboff responded that the first GLP-1 receptor agonist medications were approved in 2005, nearly 20 years ago, by the US Food and Drug Administration.

“Do I think we need long-term lifetime data? Absolutely,” she said. “We need to do our due diligence, we need to be careful, we need to monitor patients, and when and if there are signals, we need to follow them.”

What about the notorious gastrointestinal side effects of the drugs? “A majority of them are mitigated by slow up-titration,” Dr. Jastreboff noted.

“If patients have nausea, I do not go up [in dose]. I invite patients to tell me if they’re having vomiting because I don’t want anybody to have it, and I can count on one hand how many of my patients do.”

Dr. Mehta added the concern that as the drugs’ popularity soars, “a lot of doctors don’t know when they need to put the brakes on [weight coming off too quickly].”

She underscored that “we are not treating obesity for weight loss or for cosmetic reasons — this is about optimizing health.”

Dr. Jastreboff noted that in her practice, “I down-titrate if they’re losing weight too quickly.”

“If the patient is losing more than 1% per week of their body weight, then I slow down to make sure they’re getting the nutrients that they need, that they have enough energy to exercise, and that they’re prioritizing protein and fruits and vegetables in their diet.

“We just need to go slow, and yes, we need to follow them long term,” she said.

Chiming in from the audience, Julio Rosenstock, MD, a recognized thought leader in type 2 diabetes, offered his own take on the issues, describing Dr. Taylor’s very low–calorie diet suggestion as “not realistic” and Dr. Nathan’s glucose-first argument to be “stuck in the past.”

Based on modern-day evidence, “there is no reason on earth to start [diabetes treatment] with only metformin,” asserted Dr. Rosenstock, director of the Velocity Clinical Research center at Medical City and clinical professor of medicine at the University of Texas Southwestern Medical Center, Dallas.

“We need to start at the very least with metformin and a sodium-glucose cotransporter 2 (SGLT2) inhibitor from day 1, and then, if it’s affordable and there is access, with a GLP-1 receptor agonist,” he said.

“There is nothing better these days than those agents that consistently have shown a reduction of cardiovascular events and slowing of kidney disease progression.”

Overall, however, “I think you are all right,” he added, a sentiment shared by most.

Noting that the discussion as a whole represents a virtual sea change from the evidence-based options that would have been discussed only a decade ago, Dr. Retnakaran summed up his take-home message: “Stay tuned.

“You could easily see things changing in the next decade to come as we get more data and evidence to support what we ultimately should prioritize an early type 2 diabetes, so this is an exciting time.”

Dr. Retnakaran disclosed ties with Novo Nordisk, Boehringer Ingelheim, Novartis, Sanofi, and Eli Lilly. Dr. Jastreboff disclosed ties with Amgen, AstraZeneca, Boehringer Ingelheim, Biohaven, Eli Lilly, Intellihealth, Novo Nordisk, Pfizer, Regeneron, Scholar Rock, Structure Therapeutics, Terms Pharmaceutical, Weight Watchers, and Zealand Pharmaceuticals. Dr. Roopa had relationships with Novo Nordisk, Boehringer Ingelheim, Amgen, AstraZeneca, Eli Lilly, Silanes, and Sanofi. Dr. Taylor received lecture fees from Novartis, Lilly, Abbott, and Nestle Health and research funding from Diabetes UK and is an advisor to Fast800. Dr. Rosenstock reported relationships with Applied Therapeutics, AstraZeneca, Biomea Fusion, Boehringer Ingelheim, Eli Lilly and Company, Hanmi, Merck, Oramed, Structure Therapeutics, Novartis, Novo Nordisk, Pfizer, Ragor, and Sanofi. Dr. Nathan had no disclosures to report.
 

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

— What to prioritize first in managing early diabetes? That was the question debated on an expert panel at the American Diabetes Association (ADA) 84th Scientific Sessions, with impassioned responses ranging from a plea to “treat obesity first,” to a James Carville–inspired counterpoint of “it’s the glucose, stupid.” 

With a focus on preventing complications and inducing remission rounding out the four positions argued, Session Moderator Ravi Retnakaran, MD, of the University of Toronto, noted that the options reflect the tricky choices clinicians treating patients with diabetes are pressed to make on a daily basis.

“In clinical decision-making [for early diabetes], we are faced with weighing each of these variables for the individual patient, and while all are good options, strong arguments can be made for prioritizing each — with the potential of each choice to influence or improve all of the others,” Dr. Retnakaran told this news organization.
 

Which to Prioritize First?

Making the obesity first argument, Ania M. Jastreboff, MD, PhD, associate professor and director of the Yale Obesity Research Center at Yale School of Medicine, New Haven, Connecticut, noted the striking statistic that nearly 90% of people with type 2 diabetes have overweight or obesity and discussed the ever-expanding data showing the benefits of drugs including glucagon-like peptide 1 (GLP-1) receptor agonists not just in weight loss but also in kidney, cardiovascular, and, as presented at the meeting, sleep apnea improvement.

She contrasted the experiences of two patients with obesity: One treated for the obesity upon type 2 diagnosis — who had a quick normalization of lipids and hypertension soon after the obesity treatment — and the other presenting after 10 years with type 2 diabetes — who was on therapy for hypertension and hyperlipidemia but not for obesity and whose diseases were not as easily treated by that point.

“Why are we treating all the downstream effects and we’re not treating the disease that is potentially the root cause of all these other diseases?” Dr. Jastreboff said.
 

Complications?

Arguing in favor of focusing on complications, Roopa Mehta, MD, PhD, with the department of endocrinology and metabolism at Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, made the case that stakes don’t get any higher in diabetes than when it comes the looming threat of potentially fatal complications.

Acute myocardial infarction, stroke, amputation, and end-stage renal disease are all on the list of unwanted outcomes and need to be considered even in the earliest stages, as data show early onset type 2 diabetes is linked to life expectancy.

“The main goal of management has always been to prevent complications,” she noted. Citing ADA guidelines, Dr. Mehta underscored the benefits of first- and second-line therapy of metformin, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and GLP-1 receptor agonists for most patients.
 

Remission?

Discussing the priority of putting patients into disease remission, Roy Taylor, MD, professor of medicine and metabolism at Newcastle University and Newcastle Hospitals NHS in Newcastle upon Tyne, England, and author of the book Life Without Diabetes, focused on an evidence-based alternative to achieving remission — a nonpharmacologic approach that avoids costly and sometimes inaccessible drugs.

In the intervention, described in the DiRECT randomized trial and subsequently in the UK National Health Service Type 2 Diabetes Path to Remission Program, patients with overweight or obesity were placed on a highly restrictive diet of just 800-900 calories a day for 12-20 weeks, followed by maintenance for 12 months, and they not only achieved weight loss but also achieved diabetes remission, in some cases long term.

Acknowledging that “this is not for everyone,” Dr. Taylor asserted that “we have to realize there is a substantial minority of people who want to be healthy but who don’t want to be medicalized,” he said.

“They want their health, and they can do extremely well.”
 

Glucose?

In taking his self-titled “it’s the glucose, stupid” stand, David M. Nathan, MD, of the Diabetes Center, Massachusetts General Hospital, Harvard Medical School, in Boston, cited extensive evidence showing that early intensive blood glucose control with treatment including sulfonylureas, insulin, or metformin significantly reduced the risk for complications in type 2 diabetes 15 or more years later, including renal failure, blindness, amputation, and myocardial infarctions, in addition to a reduction in diabetes-related death.

“In many of these studies, you saw the benefit even in the setting of weight-gain,” Dr. Nathan underscored.

He further noted the “sobering” findings of the Look AHEAD study, which had to be stopped due to futility when an intensive lifestyle/weight loss intervention showed no significant benefits in terms of cardiovascular disease in people with type 2 diabetes at a median follow-up of 9.6 years.

Ultimately, “diabetes, type 1 and type 2, remains a gluco-centric disease,” Dr. Nathan asserted. “Hyperglycemia is the only universal link between all forms of diabetes and mortality, and the long-term complications of diabetes are intimately associated with hyperglycemia.”
 

Tackling the Caveats

The ensuing panel discussion did not fail to deliver in delving into key areas of contention, particularly in terms of GLP-1 treatment.

Regarding a lack of data on the potential long-term effects of GLP-1s: “Yes, there are a huge number of studies [on GLP-1 receptor agonists], but they are, in general, over short periods of time and driven by pharma, who get in and get out as quickly as they can and have little in the way of interest to do comparative effectiveness studies,” Dr. Nathan argued.

“Meanwhile, this is like the crack cocaine of medications — patients have to stay on it for a lifetime or they will regain the weight — are you concerned at all about a lifetime of exposure to GLP-1 [drugs]?” he asked the panel.

Dr. Jastreboff responded that the first GLP-1 receptor agonist medications were approved in 2005, nearly 20 years ago, by the US Food and Drug Administration.

“Do I think we need long-term lifetime data? Absolutely,” she said. “We need to do our due diligence, we need to be careful, we need to monitor patients, and when and if there are signals, we need to follow them.”

What about the notorious gastrointestinal side effects of the drugs? “A majority of them are mitigated by slow up-titration,” Dr. Jastreboff noted.

“If patients have nausea, I do not go up [in dose]. I invite patients to tell me if they’re having vomiting because I don’t want anybody to have it, and I can count on one hand how many of my patients do.”

Dr. Mehta added the concern that as the drugs’ popularity soars, “a lot of doctors don’t know when they need to put the brakes on [weight coming off too quickly].”

She underscored that “we are not treating obesity for weight loss or for cosmetic reasons — this is about optimizing health.”

Dr. Jastreboff noted that in her practice, “I down-titrate if they’re losing weight too quickly.”

“If the patient is losing more than 1% per week of their body weight, then I slow down to make sure they’re getting the nutrients that they need, that they have enough energy to exercise, and that they’re prioritizing protein and fruits and vegetables in their diet.

“We just need to go slow, and yes, we need to follow them long term,” she said.

Chiming in from the audience, Julio Rosenstock, MD, a recognized thought leader in type 2 diabetes, offered his own take on the issues, describing Dr. Taylor’s very low–calorie diet suggestion as “not realistic” and Dr. Nathan’s glucose-first argument to be “stuck in the past.”

Based on modern-day evidence, “there is no reason on earth to start [diabetes treatment] with only metformin,” asserted Dr. Rosenstock, director of the Velocity Clinical Research center at Medical City and clinical professor of medicine at the University of Texas Southwestern Medical Center, Dallas.

“We need to start at the very least with metformin and a sodium-glucose cotransporter 2 (SGLT2) inhibitor from day 1, and then, if it’s affordable and there is access, with a GLP-1 receptor agonist,” he said.

“There is nothing better these days than those agents that consistently have shown a reduction of cardiovascular events and slowing of kidney disease progression.”

Overall, however, “I think you are all right,” he added, a sentiment shared by most.

Noting that the discussion as a whole represents a virtual sea change from the evidence-based options that would have been discussed only a decade ago, Dr. Retnakaran summed up his take-home message: “Stay tuned.

“You could easily see things changing in the next decade to come as we get more data and evidence to support what we ultimately should prioritize an early type 2 diabetes, so this is an exciting time.”

Dr. Retnakaran disclosed ties with Novo Nordisk, Boehringer Ingelheim, Novartis, Sanofi, and Eli Lilly. Dr. Jastreboff disclosed ties with Amgen, AstraZeneca, Boehringer Ingelheim, Biohaven, Eli Lilly, Intellihealth, Novo Nordisk, Pfizer, Regeneron, Scholar Rock, Structure Therapeutics, Terms Pharmaceutical, Weight Watchers, and Zealand Pharmaceuticals. Dr. Roopa had relationships with Novo Nordisk, Boehringer Ingelheim, Amgen, AstraZeneca, Eli Lilly, Silanes, and Sanofi. Dr. Taylor received lecture fees from Novartis, Lilly, Abbott, and Nestle Health and research funding from Diabetes UK and is an advisor to Fast800. Dr. Rosenstock reported relationships with Applied Therapeutics, AstraZeneca, Biomea Fusion, Boehringer Ingelheim, Eli Lilly and Company, Hanmi, Merck, Oramed, Structure Therapeutics, Novartis, Novo Nordisk, Pfizer, Ragor, and Sanofi. Dr. Nathan had no disclosures to report.
 

A version of this article first appeared on Medscape.com.

— What to prioritize first in managing early diabetes? That was the question debated on an expert panel at the American Diabetes Association (ADA) 84th Scientific Sessions, with impassioned responses ranging from a plea to “treat obesity first,” to a James Carville–inspired counterpoint of “it’s the glucose, stupid.” 

With a focus on preventing complications and inducing remission rounding out the four positions argued, Session Moderator Ravi Retnakaran, MD, of the University of Toronto, noted that the options reflect the tricky choices clinicians treating patients with diabetes are pressed to make on a daily basis.

“In clinical decision-making [for early diabetes], we are faced with weighing each of these variables for the individual patient, and while all are good options, strong arguments can be made for prioritizing each — with the potential of each choice to influence or improve all of the others,” Dr. Retnakaran told this news organization.
 

Which to Prioritize First?

Making the obesity first argument, Ania M. Jastreboff, MD, PhD, associate professor and director of the Yale Obesity Research Center at Yale School of Medicine, New Haven, Connecticut, noted the striking statistic that nearly 90% of people with type 2 diabetes have overweight or obesity and discussed the ever-expanding data showing the benefits of drugs including glucagon-like peptide 1 (GLP-1) receptor agonists not just in weight loss but also in kidney, cardiovascular, and, as presented at the meeting, sleep apnea improvement.

She contrasted the experiences of two patients with obesity: One treated for the obesity upon type 2 diagnosis — who had a quick normalization of lipids and hypertension soon after the obesity treatment — and the other presenting after 10 years with type 2 diabetes — who was on therapy for hypertension and hyperlipidemia but not for obesity and whose diseases were not as easily treated by that point.

“Why are we treating all the downstream effects and we’re not treating the disease that is potentially the root cause of all these other diseases?” Dr. Jastreboff said.
 

Complications?

Arguing in favor of focusing on complications, Roopa Mehta, MD, PhD, with the department of endocrinology and metabolism at Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, made the case that stakes don’t get any higher in diabetes than when it comes the looming threat of potentially fatal complications.

Acute myocardial infarction, stroke, amputation, and end-stage renal disease are all on the list of unwanted outcomes and need to be considered even in the earliest stages, as data show early onset type 2 diabetes is linked to life expectancy.

“The main goal of management has always been to prevent complications,” she noted. Citing ADA guidelines, Dr. Mehta underscored the benefits of first- and second-line therapy of metformin, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and GLP-1 receptor agonists for most patients.
 

Remission?

Discussing the priority of putting patients into disease remission, Roy Taylor, MD, professor of medicine and metabolism at Newcastle University and Newcastle Hospitals NHS in Newcastle upon Tyne, England, and author of the book Life Without Diabetes, focused on an evidence-based alternative to achieving remission — a nonpharmacologic approach that avoids costly and sometimes inaccessible drugs.

In the intervention, described in the DiRECT randomized trial and subsequently in the UK National Health Service Type 2 Diabetes Path to Remission Program, patients with overweight or obesity were placed on a highly restrictive diet of just 800-900 calories a day for 12-20 weeks, followed by maintenance for 12 months, and they not only achieved weight loss but also achieved diabetes remission, in some cases long term.

Acknowledging that “this is not for everyone,” Dr. Taylor asserted that “we have to realize there is a substantial minority of people who want to be healthy but who don’t want to be medicalized,” he said.

“They want their health, and they can do extremely well.”
 

Glucose?

In taking his self-titled “it’s the glucose, stupid” stand, David M. Nathan, MD, of the Diabetes Center, Massachusetts General Hospital, Harvard Medical School, in Boston, cited extensive evidence showing that early intensive blood glucose control with treatment including sulfonylureas, insulin, or metformin significantly reduced the risk for complications in type 2 diabetes 15 or more years later, including renal failure, blindness, amputation, and myocardial infarctions, in addition to a reduction in diabetes-related death.

“In many of these studies, you saw the benefit even in the setting of weight-gain,” Dr. Nathan underscored.

He further noted the “sobering” findings of the Look AHEAD study, which had to be stopped due to futility when an intensive lifestyle/weight loss intervention showed no significant benefits in terms of cardiovascular disease in people with type 2 diabetes at a median follow-up of 9.6 years.

Ultimately, “diabetes, type 1 and type 2, remains a gluco-centric disease,” Dr. Nathan asserted. “Hyperglycemia is the only universal link between all forms of diabetes and mortality, and the long-term complications of diabetes are intimately associated with hyperglycemia.”
 

Tackling the Caveats

The ensuing panel discussion did not fail to deliver in delving into key areas of contention, particularly in terms of GLP-1 treatment.

Regarding a lack of data on the potential long-term effects of GLP-1s: “Yes, there are a huge number of studies [on GLP-1 receptor agonists], but they are, in general, over short periods of time and driven by pharma, who get in and get out as quickly as they can and have little in the way of interest to do comparative effectiveness studies,” Dr. Nathan argued.

“Meanwhile, this is like the crack cocaine of medications — patients have to stay on it for a lifetime or they will regain the weight — are you concerned at all about a lifetime of exposure to GLP-1 [drugs]?” he asked the panel.

Dr. Jastreboff responded that the first GLP-1 receptor agonist medications were approved in 2005, nearly 20 years ago, by the US Food and Drug Administration.

“Do I think we need long-term lifetime data? Absolutely,” she said. “We need to do our due diligence, we need to be careful, we need to monitor patients, and when and if there are signals, we need to follow them.”

What about the notorious gastrointestinal side effects of the drugs? “A majority of them are mitigated by slow up-titration,” Dr. Jastreboff noted.

“If patients have nausea, I do not go up [in dose]. I invite patients to tell me if they’re having vomiting because I don’t want anybody to have it, and I can count on one hand how many of my patients do.”

Dr. Mehta added the concern that as the drugs’ popularity soars, “a lot of doctors don’t know when they need to put the brakes on [weight coming off too quickly].”

She underscored that “we are not treating obesity for weight loss or for cosmetic reasons — this is about optimizing health.”

Dr. Jastreboff noted that in her practice, “I down-titrate if they’re losing weight too quickly.”

“If the patient is losing more than 1% per week of their body weight, then I slow down to make sure they’re getting the nutrients that they need, that they have enough energy to exercise, and that they’re prioritizing protein and fruits and vegetables in their diet.

“We just need to go slow, and yes, we need to follow them long term,” she said.

Chiming in from the audience, Julio Rosenstock, MD, a recognized thought leader in type 2 diabetes, offered his own take on the issues, describing Dr. Taylor’s very low–calorie diet suggestion as “not realistic” and Dr. Nathan’s glucose-first argument to be “stuck in the past.”

Based on modern-day evidence, “there is no reason on earth to start [diabetes treatment] with only metformin,” asserted Dr. Rosenstock, director of the Velocity Clinical Research center at Medical City and clinical professor of medicine at the University of Texas Southwestern Medical Center, Dallas.

“We need to start at the very least with metformin and a sodium-glucose cotransporter 2 (SGLT2) inhibitor from day 1, and then, if it’s affordable and there is access, with a GLP-1 receptor agonist,” he said.

“There is nothing better these days than those agents that consistently have shown a reduction of cardiovascular events and slowing of kidney disease progression.”

Overall, however, “I think you are all right,” he added, a sentiment shared by most.

Noting that the discussion as a whole represents a virtual sea change from the evidence-based options that would have been discussed only a decade ago, Dr. Retnakaran summed up his take-home message: “Stay tuned.

“You could easily see things changing in the next decade to come as we get more data and evidence to support what we ultimately should prioritize an early type 2 diabetes, so this is an exciting time.”

Dr. Retnakaran disclosed ties with Novo Nordisk, Boehringer Ingelheim, Novartis, Sanofi, and Eli Lilly. Dr. Jastreboff disclosed ties with Amgen, AstraZeneca, Boehringer Ingelheim, Biohaven, Eli Lilly, Intellihealth, Novo Nordisk, Pfizer, Regeneron, Scholar Rock, Structure Therapeutics, Terms Pharmaceutical, Weight Watchers, and Zealand Pharmaceuticals. Dr. Roopa had relationships with Novo Nordisk, Boehringer Ingelheim, Amgen, AstraZeneca, Eli Lilly, Silanes, and Sanofi. Dr. Taylor received lecture fees from Novartis, Lilly, Abbott, and Nestle Health and research funding from Diabetes UK and is an advisor to Fast800. Dr. Rosenstock reported relationships with Applied Therapeutics, AstraZeneca, Biomea Fusion, Boehringer Ingelheim, Eli Lilly and Company, Hanmi, Merck, Oramed, Structure Therapeutics, Novartis, Novo Nordisk, Pfizer, Ragor, and Sanofi. Dr. Nathan had no disclosures to report.
 

A version of this article first 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>168616</fileName> <TBEID>0C050DA0.SIG</TBEID> <TBUniqueIdentifier>MD_0C050DA0</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240703T104552</QCDate> <firstPublished>20240703T105107</firstPublished> <LastPublished>20240703T105107</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240703T105107</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Nancy Melville</byline> <bylineText>NANCY A. MELVILLE</bylineText> <bylineFull>NANCY A. MELVILLE</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>Session Moderator Ravi Retnakaran, MD, of the University of Toronto, noted that the options reflect the tricky choices clinicians treating patients with diabete</metaDescription> <articlePDF/> <teaserImage/> <teaser>ADA meeting debate on what to treat first in early diabetes included obesity, glucose, remission as priorities.</teaser> <title>What Should Be Prioritized in Managing Early Diabetes?</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>endo</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> </publications_g> <publications> <term canonical="true">34</term> <term>21</term> <term>15</term> </publications> <sections> <term canonical="true">39313</term> </sections> <topics> <term canonical="true">205</term> <term>261</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>What Should Be Prioritized in Managing Early Diabetes?</title> <deck/> </itemMeta> <itemContent> <p><span class="dateline">ORLANDO, FLORIDA</span> — What to prioritize first in managing early diabetes? That was the question debated on an expert panel at the <a href="https://www.medscape.com/viewcollection/37466">American Diabetes Association (ADA) 84th Scientific Sessions</a>, with impassioned responses ranging from a plea to “treat obesity first,” to a James Carville–inspired counterpoint of “it’s the glucose, stupid.” </p> <p>With a focus on preventing complications and inducing remission rounding out the four positions argued, <span class="tag metaDescription">Session Moderator Ravi Retnakaran, MD, of the University of Toronto, noted that the options reflect the tricky choices clinicians treating patients with diabetes are pressed to make on a daily basis.</span><br/><br/>“In clinical decision-making [for early diabetes], we are faced with weighing each of these variables for the individual patient, and while all are good options, strong arguments can be made for prioritizing each — with the potential of each choice to influence or improve all of the others,” Dr. Retnakaran told this news organization.<br/><br/></p> <h2>Which to Prioritize First?</h2> <p>Making the obesity first argument, Ania M. Jastreboff, MD, PhD, associate professor and director of the Yale Obesity Research Center at Yale School of Medicine, New Haven, Connecticut, noted the striking statistic that nearly 90% of people with type 2 diabetes have overweight or obesity and discussed the ever-expanding data showing the benefits of drugs including glucagon-like peptide 1 (GLP-1) receptor agonists not just in weight loss but also in kidney, cardiovascular, and, as presented at the meeting, sleep apnea improvement.<br/><br/>She contrasted the experiences of two patients with obesity: One treated for the obesity upon type 2 diagnosis — who had a quick normalization of lipids and hypertension soon after the obesity treatment — and the other presenting after 10 years with type 2 diabetes — who was on therapy for hypertension and hyperlipidemia but not for obesity and whose diseases were not as easily treated by that point.<br/><br/>“Why are we treating all the downstream effects and we’re not treating the disease that is potentially the root cause of all these other diseases?” Dr. Jastreboff said.<br/><br/></p> <h2>Complications?</h2> <p>Arguing in favor of focusing on complications, Roopa Mehta, MD, PhD, with the department of endocrinology and metabolism at Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), Mexico City, made the case that stakes don’t get any higher in diabetes than when it comes the looming threat of potentially fatal complications.</p> <p>Acute myocardial infarction, stroke, amputation, and end-stage renal disease are all on the list of unwanted outcomes and need to be considered even in the earliest stages, as data show early onset type 2 diabetes is linked to life expectancy.<br/><br/>“The main goal of management has always been to prevent complications,” she noted. Citing ADA guidelines, Dr. Mehta underscored the benefits of first- and second-line therapy of metformin, sodium-glucose cotransporter 2 (SGLT2) inhibitors, and GLP-1 receptor agonists for most patients.<br/><br/></p> <h2>Remission?</h2> <p>Discussing the priority of putting patients into disease remission, Roy Taylor, MD, professor of medicine and metabolism at Newcastle University and Newcastle Hospitals NHS in Newcastle upon Tyne, England, and author of <span class="Hyperlink"><a href="https://www.harpercollins.com/products/life-without-diabetes-roy-taylor?variant=32126577672226">the book</a></span> <em>Life Without Diabetes</em>, focused on an evidence-based alternative to achieving remission — a nonpharmacologic approach that avoids costly and sometimes inaccessible drugs.</p> <p>In the intervention, described in <a href="https://www.thelancet.com/journals/landia/article/PIIS2213-8587(23)00385-6/fulltext">the DiRECT randomized trial</a> and subsequently in the UK National Health Service Type 2 Diabetes Path to Remission Program, patients with overweight or obesity were placed on a highly restrictive diet of just 800-900 calories a day for 12-20 weeks, followed by maintenance for 12 months, and they not only achieved weight loss but also achieved diabetes remission, in some cases long term.<br/><br/>Acknowledging that “this is not for everyone,” Dr. Taylor asserted that “we have to realize there is a substantial minority of people who want to be healthy but who don’t want to be medicalized,” he said.<br/><br/>“They want their health, and they can do extremely well.”<br/><br/></p> <h2>Glucose?</h2> <p>In taking his self-titled “it’s the glucose, stupid” stand, David M. Nathan, MD, of the Diabetes Center, Massachusetts General Hospital, Harvard Medical School, in Boston, cited extensive evidence showing that early intensive blood glucose control with treatment including sulfonylureas, insulin, or metformin significantly reduced the risk for complications in type 2 diabetes 15 or more years later, including renal failure, blindness, amputation, and myocardial infarctions, in addition to a reduction in diabetes-related death.</p> <p>“In many of these studies, you saw the benefit even in the setting of weight-gain,” Dr. Nathan underscored.<br/><br/>He further noted the “sobering” findings of the <a href="https://www.nejm.org/doi/full/10.1056/NEJMoa1212914">Look AHEAD</a> study, which had to be stopped due to futility when an intensive lifestyle/weight loss intervention showed no significant benefits in terms of cardiovascular disease in people with type 2 diabetes at a median follow-up of 9.6 years.<br/><br/>Ultimately, “diabetes, type 1 and type 2, remains a gluco-centric disease,” Dr. Nathan asserted. “Hyperglycemia is the only universal link between all forms of diabetes and mortality, and the long-term complications of diabetes are intimately associated with hyperglycemia.”<br/><br/></p> <h2>Tackling the Caveats</h2> <p>The ensuing panel discussion did not fail to deliver in delving into key areas of contention, particularly in terms of GLP-1 treatment.</p> <p>Regarding a lack of data on the potential long-term effects of GLP-1s: “Yes, there are a huge number of studies [on GLP-1 receptor agonists], but they are, in general, over short periods of time and driven by pharma, who get in and get out as quickly as they can and have little in the way of interest to do comparative effectiveness studies,” Dr. Nathan argued.<br/><br/>“Meanwhile, this is like the crack cocaine of medications — patients have to stay on it for a lifetime or they will regain the weight — are you concerned at all about a lifetime of exposure to GLP-1 [drugs]?” he asked the panel.<br/><br/>Dr. Jastreboff responded that the first GLP-1 receptor agonist medications were approved in 2005, nearly 20 years ago, by the US Food and Drug Administration.<br/><br/>“Do I think we need long-term lifetime data? Absolutely,” she said. “We need to do our due diligence, we need to be careful, we need to monitor patients, and when and if there are signals, we need to follow them.”<br/><br/>What about the notorious gastrointestinal side effects of the drugs? “A majority of them are mitigated by slow up-titration,” Dr. Jastreboff noted.<br/><br/>“If patients have nausea, I do not go up [in dose]. I invite patients to tell me if they’re having vomiting because I don’t want anybody to have it, and I can count on one hand how many of my patients do.”<br/><br/>Dr. Mehta added the concern that as the drugs’ popularity soars, “a lot of doctors don’t know when they need to put the brakes on [weight coming off too quickly].”<br/><br/>She underscored that “we are not treating obesity for weight loss or for cosmetic reasons — this is about optimizing health.”<br/><br/>Dr. Jastreboff noted that in her practice, “I down-titrate if they’re losing weight too quickly.”<br/><br/>“If the patient is losing more than 1% per week of their body weight, then I slow down to make sure they’re getting the nutrients that they need, that they have enough energy to exercise, and that they’re prioritizing protein and fruits and vegetables in their diet.<br/><br/>“We just need to go slow, and yes, we need to follow them long term,” she said.<br/><br/>Chiming in from the audience, Julio Rosenstock, MD, a recognized thought leader in type 2 diabetes, offered his own take on the issues, describing Dr. Taylor’s very low–calorie diet suggestion as “not realistic” and Dr. Nathan’s glucose-first argument to be “stuck in the past.”<br/><br/>Based on modern-day evidence, “there is no reason on earth to start [diabetes treatment] with only metformin,” asserted Dr. Rosenstock, director of the Velocity Clinical Research center at Medical City and clinical professor of medicine at the University of Texas Southwestern Medical Center, Dallas.<br/><br/>“We need to start at the very least with metformin and a sodium-glucose cotransporter 2 (SGLT2) inhibitor from day 1, and then, if it’s affordable and there is access, with a GLP-1 receptor agonist,” he said.<br/><br/>“There is nothing better these days than those agents that consistently have shown a reduction of cardiovascular events and slowing of kidney disease progression.”<br/><br/>Overall, however, “I think you are all right,” he added, a sentiment shared by most.<br/><br/>Noting that the discussion as a whole represents a virtual sea change from the evidence-based options that would have been discussed only a decade ago, Dr. Retnakaran summed up his take-home message: “Stay tuned.<br/><br/>“You could easily see things changing in the next decade to come as we get more data and evidence to support what we ultimately should prioritize an early type 2 diabetes, so this is an exciting time.”<br/><br/>Dr. Retnakaran disclosed ties with Novo Nordisk, Boehringer Ingelheim, Novartis, Sanofi, and Eli Lilly. Dr. Jastreboff disclosed ties with Amgen, AstraZeneca, Boehringer Ingelheim, Biohaven, Eli Lilly, Intellihealth, Novo Nordisk, Pfizer, Regeneron, Scholar Rock, Structure Therapeutics, Terms Pharmaceutical, Weight Watchers, and Zealand Pharmaceuticals. Dr. Roopa had relationships with Novo Nordisk, Boehringer Ingelheim, Amgen, AstraZeneca, Eli Lilly, Silanes, and Sanofi. Dr. Taylor received lecture fees from Novartis, Lilly, Abbott, and Nestle Health and research funding from Diabetes UK and is an advisor to Fast800. Dr. Rosenstock reported relationships with Applied Therapeutics, AstraZeneca, Biomea Fusion, Boehringer Ingelheim, Eli Lilly and Company, Hanmi, Merck, Oramed, Structure Therapeutics, Novartis, Novo Nordisk, Pfizer, Ragor, and Sanofi. Dr. Nathan had no disclosures to report.<span class="end"><br/><br/></span></p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/debate-what-should-be-priority-managing-early-diabetes-2024a1000c8e">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

Triple Therapy May Be Effective in Drug-Naive T2D

Article Type
Changed
Wed, 07/03/2024 - 10:12

 

TOPLINE:

A triple combination therapy (TCT) of metformindapagliflozin, and saxagliptin is an effective and safe treatment option for drug-naive patients with type 2 diabetes (T2D) compared with stepwise add-on therapy.

METHODOLOGY:

  • Current guidelines recommend early combination therapy to extend the time to treatment failure, reduce the risk for diabetic complications, and prevent clinical inertia in patients with T2D.
  • This randomized controlled open-label trial conducted at nine sites in South Korea included 105 drug-naive patients with T2D (mean age, 49.5 years; 32.4% women) who either received triple therapy (metformin, dapagliflozin, and saxagliptin) or stepwise add-on therapy (initiated with metformin, followed by glimepiride and sitagliptin for those with baseline hemoglobin A1c levels < 9.0% or with initial dual metformin and glimepiride in those with A1c levels ≥ 9.0% followed by sitagliptin).
  • The primary outcome was the proportion of patients who achieved A1c levels < 6.5% without hypoglycemia, weight gain ≥ 5%, or discontinuation of drugs because of adverse events at week 104.
  • The secondary outcomes were the proportion of patients whose A1c levels dropped to < 7.0% at weeks 56 and 104 and dropped to < 6.5% at week 56, all without hypoglycemia, weight gain, nor discontinuation due to adverse events.

TAKEAWAY:

  • At week 104, a higher proportion of patients in the triple therapy group achieved the primary outcome than those in the stepwise add-on therapy group (39.0% vs 17.1%; P = .027).
  • In both groups, a similar proportion of patients (46.3%) achieved A1c levels < 6.5% at week 104, but the proportion of patients without hypoglycemia, weight gain, or discontinuation because of adverse events was higher in the triple therapy group than those in the stepwise add-on therapy group (83.3% vs 38.0%; P < .001).

IN PRACTICE:

The authors wrote: “Although the glycemic efficacy of each drug in the TCT was modest, the combination of these drugs resulted in a 2-year durable glycemic efficacy, with greater than a 2.5% reduction in A1c levels from baseline. The overall results of this study suggest a novel strategy for initial combination therapy in newly diagnosed T2D patients.”

SOURCE:

The study was led by Nam Hoon Kim, MD, of the Department of Internal Medicine, Korea University College of Medicine, Seoul. It was published online in Diabetes, Obesity and Metabolism.

LIMITATIONS:

The study had a relatively small sample size as compared with previous clinical trials. More people in the standard therapy group had A1c levels ≥ 9.0%, which resulted in more than double the number of people receiving dual combination therapy over monotherapy in that group. The trial duration was insufficient to evaluate the cardiovascular outcomes.

DISCLOSURES:

The study was funded by AstraZeneca. Some authors reported financial ties with AstraZeneca and other pharmaceutical and medical device companies as members of advisory boards or recipients of grants, consulting fees, honoraria, or lecture fees.

A version of this article appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

A triple combination therapy (TCT) of metformindapagliflozin, and saxagliptin is an effective and safe treatment option for drug-naive patients with type 2 diabetes (T2D) compared with stepwise add-on therapy.

METHODOLOGY:

  • Current guidelines recommend early combination therapy to extend the time to treatment failure, reduce the risk for diabetic complications, and prevent clinical inertia in patients with T2D.
  • This randomized controlled open-label trial conducted at nine sites in South Korea included 105 drug-naive patients with T2D (mean age, 49.5 years; 32.4% women) who either received triple therapy (metformin, dapagliflozin, and saxagliptin) or stepwise add-on therapy (initiated with metformin, followed by glimepiride and sitagliptin for those with baseline hemoglobin A1c levels < 9.0% or with initial dual metformin and glimepiride in those with A1c levels ≥ 9.0% followed by sitagliptin).
  • The primary outcome was the proportion of patients who achieved A1c levels < 6.5% without hypoglycemia, weight gain ≥ 5%, or discontinuation of drugs because of adverse events at week 104.
  • The secondary outcomes were the proportion of patients whose A1c levels dropped to < 7.0% at weeks 56 and 104 and dropped to < 6.5% at week 56, all without hypoglycemia, weight gain, nor discontinuation due to adverse events.

TAKEAWAY:

  • At week 104, a higher proportion of patients in the triple therapy group achieved the primary outcome than those in the stepwise add-on therapy group (39.0% vs 17.1%; P = .027).
  • In both groups, a similar proportion of patients (46.3%) achieved A1c levels < 6.5% at week 104, but the proportion of patients without hypoglycemia, weight gain, or discontinuation because of adverse events was higher in the triple therapy group than those in the stepwise add-on therapy group (83.3% vs 38.0%; P < .001).

IN PRACTICE:

The authors wrote: “Although the glycemic efficacy of each drug in the TCT was modest, the combination of these drugs resulted in a 2-year durable glycemic efficacy, with greater than a 2.5% reduction in A1c levels from baseline. The overall results of this study suggest a novel strategy for initial combination therapy in newly diagnosed T2D patients.”

SOURCE:

The study was led by Nam Hoon Kim, MD, of the Department of Internal Medicine, Korea University College of Medicine, Seoul. It was published online in Diabetes, Obesity and Metabolism.

LIMITATIONS:

The study had a relatively small sample size as compared with previous clinical trials. More people in the standard therapy group had A1c levels ≥ 9.0%, which resulted in more than double the number of people receiving dual combination therapy over monotherapy in that group. The trial duration was insufficient to evaluate the cardiovascular outcomes.

DISCLOSURES:

The study was funded by AstraZeneca. Some authors reported financial ties with AstraZeneca and other pharmaceutical and medical device companies as members of advisory boards or recipients of grants, consulting fees, honoraria, or lecture fees.

A version of this article appeared on Medscape.com.

 

TOPLINE:

A triple combination therapy (TCT) of metformindapagliflozin, and saxagliptin is an effective and safe treatment option for drug-naive patients with type 2 diabetes (T2D) compared with stepwise add-on therapy.

METHODOLOGY:

  • Current guidelines recommend early combination therapy to extend the time to treatment failure, reduce the risk for diabetic complications, and prevent clinical inertia in patients with T2D.
  • This randomized controlled open-label trial conducted at nine sites in South Korea included 105 drug-naive patients with T2D (mean age, 49.5 years; 32.4% women) who either received triple therapy (metformin, dapagliflozin, and saxagliptin) or stepwise add-on therapy (initiated with metformin, followed by glimepiride and sitagliptin for those with baseline hemoglobin A1c levels < 9.0% or with initial dual metformin and glimepiride in those with A1c levels ≥ 9.0% followed by sitagliptin).
  • The primary outcome was the proportion of patients who achieved A1c levels < 6.5% without hypoglycemia, weight gain ≥ 5%, or discontinuation of drugs because of adverse events at week 104.
  • The secondary outcomes were the proportion of patients whose A1c levels dropped to < 7.0% at weeks 56 and 104 and dropped to < 6.5% at week 56, all without hypoglycemia, weight gain, nor discontinuation due to adverse events.

TAKEAWAY:

  • At week 104, a higher proportion of patients in the triple therapy group achieved the primary outcome than those in the stepwise add-on therapy group (39.0% vs 17.1%; P = .027).
  • In both groups, a similar proportion of patients (46.3%) achieved A1c levels < 6.5% at week 104, but the proportion of patients without hypoglycemia, weight gain, or discontinuation because of adverse events was higher in the triple therapy group than those in the stepwise add-on therapy group (83.3% vs 38.0%; P < .001).

IN PRACTICE:

The authors wrote: “Although the glycemic efficacy of each drug in the TCT was modest, the combination of these drugs resulted in a 2-year durable glycemic efficacy, with greater than a 2.5% reduction in A1c levels from baseline. The overall results of this study suggest a novel strategy for initial combination therapy in newly diagnosed T2D patients.”

SOURCE:

The study was led by Nam Hoon Kim, MD, of the Department of Internal Medicine, Korea University College of Medicine, Seoul. It was published online in Diabetes, Obesity and Metabolism.

LIMITATIONS:

The study had a relatively small sample size as compared with previous clinical trials. More people in the standard therapy group had A1c levels ≥ 9.0%, which resulted in more than double the number of people receiving dual combination therapy over monotherapy in that group. The trial duration was insufficient to evaluate the cardiovascular outcomes.

DISCLOSURES:

The study was funded by AstraZeneca. Some authors reported financial ties with AstraZeneca and other pharmaceutical and medical device companies as members of advisory boards or recipients of grants, consulting fees, honoraria, or lecture fees.

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>168587</fileName> <TBEID>0C050D18.SIG</TBEID> <TBUniqueIdentifier>MD_0C050D18</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240703T100337</QCDate> <firstPublished>20240703T100736</firstPublished> <LastPublished>20240703T100736</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240703T100735</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Javed Choudhury</byline> <bylineText>JAVED CHOUDHURY</bylineText> <bylineFull>JAVED CHOUDHURY</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 triple combination therapy (TCT) of metformin, dapagliflozin, and saxagliptin is an effective and safe treatment option for drug-naive patients with type 2 di</metaDescription> <articlePDF/> <teaserImage/> <teaser>Combo of metformin, dapagliflozin, and saxagliptin could reduce body weight and adverse events, compared with step-wise therapy.</teaser> <title>Triple Therapy May Be Effective in Drug-Naive T2D</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>endo</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>dbh</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term canonical="true">34</term> <term>358</term> <term>21</term> <term>15</term> </publications> <sections> <term canonical="true">39313</term> <term>27970</term> </sections> <topics> <term canonical="true">205</term> <term>261</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Triple Therapy May Be Effective in Drug-Naive T2D</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE:</h2> <p>A triple combination therapy (TCT) of <span class="Hyperlink">metformin</span>, <span class="Hyperlink">dapagliflozin</span>, and saxagliptin is an effective and safe treatment option for drug-naive patients with type 2 diabetes (T2D) compared with stepwise add-on therapy.</p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>Current guidelines recommend early combination therapy to extend the time to treatment failure, reduce the risk for diabetic complications, and prevent clinical inertia in patients with T2D.</li> <li>This randomized controlled open-label trial conducted at nine sites in South Korea included 105 drug-naive patients with T2D (mean age, 49.5 years; 32.4% women) who either received triple therapy (metformin, dapagliflozin, and saxagliptin) or stepwise add-on therapy (initiated with metformin, followed by <span class="Hyperlink">glimepiride</span> and sitagliptin for those with baseline hemoglobin <span class="Hyperlink">A1c</span> levels &lt; 9.0% or with initial dual metformin and glimepiride in those with A1c levels ≥ 9.0% followed by sitagliptin).</li> <li>The primary outcome was the proportion of patients who achieved A1c levels &lt; 6.5% without <span class="Hyperlink">hypoglycemia</span>, weight gain ≥ 5%, or discontinuation of drugs because of adverse events at week 104.</li> <li>The secondary outcomes were the proportion of patients whose A1c levels dropped to &lt; 7.0% at weeks 56 and 104 and dropped to &lt; 6.5% at week 56, all without hypoglycemia, weight gain, nor discontinuation due to adverse events.</li> </ul> <h2>TAKEAWAY:</h2> <ul class="body"> <li>At week 104, a higher proportion of patients in the triple therapy group achieved the primary outcome than those in the stepwise add-on therapy group (39.0% vs 17.1%; <em>P</em> = .027).</li> <li>In both groups, a similar proportion of patients (46.3%) achieved A1c levels &lt; 6.5% at week 104, but the proportion of patients without hypoglycemia, weight gain, or discontinuation because of adverse events was higher in the triple therapy group than those in the stepwise add-on therapy group (83.3% vs 38.0%; <em>P</em> &lt; .001).</li> <li> </li> </ul> <h2>IN PRACTICE:</h2> <p>The authors wrote: “Although the glycemic efficacy of each drug in the TCT was modest, the combination of these drugs resulted in a 2-year durable glycemic efficacy, with greater than a 2.5% reduction in A1c levels from baseline. The overall results of this study suggest a novel strategy for initial combination therapy in newly diagnosed T2D patients.”</p> <h2>SOURCE:</h2> <p>The study was led by Nam Hoon Kim, MD, of the Department of Internal Medicine, Korea University College of Medicine, Seoul. It was <span class="Hyperlink"><a href="https://doi.org/10.1111/dom.15705">published online</a></span> in Diabetes, Obesity and Metabolism.</p> <h2>LIMITATIONS:</h2> <p>The study had a relatively small sample size as compared with previous clinical trials. More people in the standard therapy group had A1c levels ≥ 9.0%, which resulted in more than double the number of people receiving dual combination therapy over monotherapy in that group. The trial duration was insufficient to evaluate the cardiovascular outcomes.</p> <h2>DISCLOSURES:</h2> <p>The study was funded by AstraZeneca. Some authors reported financial ties with AstraZeneca and other pharmaceutical and medical device companies as members of advisory boards or recipients of grants, consulting fees, honoraria, or lecture fees.<span class="end"/></p> <p> <em>A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/initial-triple-therapy-shows-promise-drug-naive-t2d-2024a1000c50">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

Trading TV Time for Physical Activity Boosts Healthy Aging

Article Type
Changed
Wed, 07/10/2024 - 13:54

 

TOPLINE:

Sedentary behavior, particularly sitting and watching television, is linked to lower odds of healthy aging, but substituting it with any physical activity — or even sleeping, in case of women with inadequate sleep — may lead to better overall health.

METHODOLOGY:

  • Previous studies have shown that replacing sedentary behavior with physical activity may improve mortality outcomes, but whether this increased lifespan is accompanied by better overall health remains an unanswered question.
  • To understand the impact of sedentary behavior and physical activity on healthy aging, researchers analyzed data from the prospective cohort Nurses’ Health Study.
  • They included 45,176 women aged > 50 years in 1992 (mean age, 59.2 years) who were free of major chronic diseases and were followed up for 20 years.
  • In 1992, validated questionnaires were used to record exposure to sedentary behavior, different levels of physical activity, and sleep. The time spent watching television was the primary exposure in the sedentary behavior category.
  • The main outcome was healthy aging, defined as survival to ≥ 70 years of age and maintenance of four domains of health — being free of 11 main chronic diseases and having no impairment of subjective memory, physical function, or mental health.

TAKEAWAY:

  • At 20 years of follow-up, 8.6% of the women achieved healthy aging, while 41.4% had none of the 11 chronic diseases, 16.1% had no physical function impairment, 44.1% had no mental health limitation, and 51.9% reported no memory impairment.
  • For each increase of 2 hours per day spent sitting and watching television, the odds of healthy aging dropped by 12% (95% confidence interval [CI], 7%-17%).
  • Conversely, every additional 2 hours per day of low-level physical activity at work upped the odds of healthy aging by 6% (95% CI, 3%-9%); furthermore, each extra hour per day of standardized moderate to vigorous physical activity (normal pace walking or the equivalent) was associated with 14% higher odds (95% CI, 11%-16%) of healthy aging.
  • In a theoretical modeling analysis, individuals could increase their odds of healthy aging by replacing 1 hour of television time per day with low levels of physical activity at home and work or with moderate to vigorous levels of physical activity — or even sleeping, for those who slept for ≤ 7 hours.

IN PRACTICE:

“These findings expand on the literature reporting that replacing sedentary behavior with light or moderate to vigorous physical activity is associated with decreased mortality by suggesting that this increased lifespan might be accompanied by better overall health,” the authors wrote.

SOURCE:

Hongying Shi, PhD, Department of Epidemiology and Health Statistics, School of Public Health, Wenzhou Medical University, Wenzhou, China, led this study, which was published online in JAMA Network Open.

LIMITATIONS:

The measures of different behaviors were self-reported and may, therefore, be less accurate than objective measurement methods. Measurement error may have attenuated the effect of low levels of physical activity. The single exposure assessment at baseline may not reflect the long-term pattern of these activities.

DISCLOSURES:

The lead author was supported by the National Social Science Foundation Project of China and the Zhejiang Provincial Philosophy and Social Sciences Planning Project. A co-author and the Nurses’ Health Study were supported by the US National Institutes of Health. The authors declared no conflicts of interest.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

 

TOPLINE:

Sedentary behavior, particularly sitting and watching television, is linked to lower odds of healthy aging, but substituting it with any physical activity — or even sleeping, in case of women with inadequate sleep — may lead to better overall health.

METHODOLOGY:

  • Previous studies have shown that replacing sedentary behavior with physical activity may improve mortality outcomes, but whether this increased lifespan is accompanied by better overall health remains an unanswered question.
  • To understand the impact of sedentary behavior and physical activity on healthy aging, researchers analyzed data from the prospective cohort Nurses’ Health Study.
  • They included 45,176 women aged > 50 years in 1992 (mean age, 59.2 years) who were free of major chronic diseases and were followed up for 20 years.
  • In 1992, validated questionnaires were used to record exposure to sedentary behavior, different levels of physical activity, and sleep. The time spent watching television was the primary exposure in the sedentary behavior category.
  • The main outcome was healthy aging, defined as survival to ≥ 70 years of age and maintenance of four domains of health — being free of 11 main chronic diseases and having no impairment of subjective memory, physical function, or mental health.

TAKEAWAY:

  • At 20 years of follow-up, 8.6% of the women achieved healthy aging, while 41.4% had none of the 11 chronic diseases, 16.1% had no physical function impairment, 44.1% had no mental health limitation, and 51.9% reported no memory impairment.
  • For each increase of 2 hours per day spent sitting and watching television, the odds of healthy aging dropped by 12% (95% confidence interval [CI], 7%-17%).
  • Conversely, every additional 2 hours per day of low-level physical activity at work upped the odds of healthy aging by 6% (95% CI, 3%-9%); furthermore, each extra hour per day of standardized moderate to vigorous physical activity (normal pace walking or the equivalent) was associated with 14% higher odds (95% CI, 11%-16%) of healthy aging.
  • In a theoretical modeling analysis, individuals could increase their odds of healthy aging by replacing 1 hour of television time per day with low levels of physical activity at home and work or with moderate to vigorous levels of physical activity — or even sleeping, for those who slept for ≤ 7 hours.

IN PRACTICE:

“These findings expand on the literature reporting that replacing sedentary behavior with light or moderate to vigorous physical activity is associated with decreased mortality by suggesting that this increased lifespan might be accompanied by better overall health,” the authors wrote.

SOURCE:

Hongying Shi, PhD, Department of Epidemiology and Health Statistics, School of Public Health, Wenzhou Medical University, Wenzhou, China, led this study, which was published online in JAMA Network Open.

LIMITATIONS:

The measures of different behaviors were self-reported and may, therefore, be less accurate than objective measurement methods. Measurement error may have attenuated the effect of low levels of physical activity. The single exposure assessment at baseline may not reflect the long-term pattern of these activities.

DISCLOSURES:

The lead author was supported by the National Social Science Foundation Project of China and the Zhejiang Provincial Philosophy and Social Sciences Planning Project. A co-author and the Nurses’ Health Study were supported by the US National Institutes of Health. The authors declared no conflicts of interest.

A version of this article first appeared on Medscape.com.

 

TOPLINE:

Sedentary behavior, particularly sitting and watching television, is linked to lower odds of healthy aging, but substituting it with any physical activity — or even sleeping, in case of women with inadequate sleep — may lead to better overall health.

METHODOLOGY:

  • Previous studies have shown that replacing sedentary behavior with physical activity may improve mortality outcomes, but whether this increased lifespan is accompanied by better overall health remains an unanswered question.
  • To understand the impact of sedentary behavior and physical activity on healthy aging, researchers analyzed data from the prospective cohort Nurses’ Health Study.
  • They included 45,176 women aged > 50 years in 1992 (mean age, 59.2 years) who were free of major chronic diseases and were followed up for 20 years.
  • In 1992, validated questionnaires were used to record exposure to sedentary behavior, different levels of physical activity, and sleep. The time spent watching television was the primary exposure in the sedentary behavior category.
  • The main outcome was healthy aging, defined as survival to ≥ 70 years of age and maintenance of four domains of health — being free of 11 main chronic diseases and having no impairment of subjective memory, physical function, or mental health.

TAKEAWAY:

  • At 20 years of follow-up, 8.6% of the women achieved healthy aging, while 41.4% had none of the 11 chronic diseases, 16.1% had no physical function impairment, 44.1% had no mental health limitation, and 51.9% reported no memory impairment.
  • For each increase of 2 hours per day spent sitting and watching television, the odds of healthy aging dropped by 12% (95% confidence interval [CI], 7%-17%).
  • Conversely, every additional 2 hours per day of low-level physical activity at work upped the odds of healthy aging by 6% (95% CI, 3%-9%); furthermore, each extra hour per day of standardized moderate to vigorous physical activity (normal pace walking or the equivalent) was associated with 14% higher odds (95% CI, 11%-16%) of healthy aging.
  • In a theoretical modeling analysis, individuals could increase their odds of healthy aging by replacing 1 hour of television time per day with low levels of physical activity at home and work or with moderate to vigorous levels of physical activity — or even sleeping, for those who slept for ≤ 7 hours.

IN PRACTICE:

“These findings expand on the literature reporting that replacing sedentary behavior with light or moderate to vigorous physical activity is associated with decreased mortality by suggesting that this increased lifespan might be accompanied by better overall health,” the authors wrote.

SOURCE:

Hongying Shi, PhD, Department of Epidemiology and Health Statistics, School of Public Health, Wenzhou Medical University, Wenzhou, China, led this study, which was published online in JAMA Network Open.

LIMITATIONS:

The measures of different behaviors were self-reported and may, therefore, be less accurate than objective measurement methods. Measurement error may have attenuated the effect of low levels of physical activity. The single exposure assessment at baseline may not reflect the long-term pattern of these activities.

DISCLOSURES:

The lead author was supported by the National Social Science Foundation Project of China and the Zhejiang Provincial Philosophy and Social Sciences Planning Project. A co-author and the Nurses’ Health Study were supported by the US National Institutes of Health. The authors declared no conflicts of interest.

A version of this article first 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>168601</fileName> <TBEID>0C050D4F.SIG</TBEID> <TBUniqueIdentifier>MD_0C050D4F</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240702T120553</QCDate> <firstPublished>20240702T125749</firstPublished> <LastPublished>20240702T125749</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240702T125749</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>SHRABASTI BHATTACHARYA</byline> <bylineText>SHRABASTI BHATTACHARYA</bylineText> <bylineFull>SHRABASTI BHATTACHARYA</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <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>Sedentary behavior, particularly sitting and watching television, is linked to lower odds of healthy aging</metaDescription> <articlePDF/> <teaserImage/> <teaser>For each increase of 2 hours per day spent sitting and watching television, the odds of healthy aging dropped by 12%.</teaser> <title>Trading TV Time for Physical Activity Boosts Healthy Aging</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>card</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>rn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term>5</term> <term>15</term> <term canonical="true">21</term> <term>26</term> </publications> <sections> <term>27970</term> <term canonical="true">39313</term> </sections> <topics> <term>215</term> <term>261</term> <term>280</term> <term canonical="true">322</term> <term>265</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Trading TV Time for Physical Activity Boosts Healthy Aging</title> <deck/> </itemMeta> <itemContent> <h2>TOPLINE:</h2> <p><span class="tag metaDescription">Sedentary behavior, particularly sitting and watching television, is linked to lower odds of healthy aging</span>, but substituting it with any physical activity — or even sleeping, in case of women with inadequate sleep — may lead to better overall health.</p> <h2>METHODOLOGY:</h2> <ul class="body"> <li>Previous studies have shown that replacing sedentary behavior with physical activity may improve mortality outcomes, but whether this increased lifespan is accompanied by better overall health remains an unanswered question.</li> <li>To understand the impact of sedentary behavior and physical activity on healthy aging, researchers analyzed data from the prospective cohort Nurses’ Health Study.</li> <li>They included 45,176 women aged &gt; 50 years in 1992 (mean age, 59.2 years) who were free of major chronic diseases and were followed up for 20 years.</li> <li>In 1992, validated questionnaires were used to record exposure to sedentary behavior, different levels of physical activity, and sleep. The time spent watching television was the primary exposure in the sedentary behavior category.</li> <li>The main outcome was healthy aging, defined as survival to ≥ 70 years of age and maintenance of four domains of health — being free of 11 main chronic diseases and having no impairment of subjective memory, physical function, or mental health.</li> </ul> <h2>TAKEAWAY:</h2> <ul class="body"> <li>At 20 years of follow-up, 8.6% of the women achieved healthy aging, while 41.4% had none of the 11 chronic diseases, 16.1% had no physical function impairment, 44.1% had no mental health limitation, and 51.9% reported no memory impairment.</li> <li>For each increase of 2 hours per day spent sitting and watching television, the odds of healthy aging dropped by 12% (95% confidence interval [CI], 7%-17%).</li> <li>Conversely, every additional 2 hours per day of low-level physical activity at work upped the odds of healthy aging by 6% (95% CI, 3%-9%); furthermore, each extra hour per day of standardized moderate to vigorous physical activity (normal pace walking or the equivalent) was associated with 14% higher odds (95% CI, 11%-16%) of healthy aging.</li> <li>In a theoretical modeling analysis, individuals could increase their odds of healthy aging by replacing 1 hour of television time per day with low levels of physical activity at home and work or with moderate to vigorous levels of physical activity — or even sleeping, for those who slept for ≤ 7 hours.</li> </ul> <h2>IN PRACTICE:</h2> <p>“These findings expand on the literature reporting that replacing sedentary behavior with light or moderate to vigorous physical activity is associated with decreased mortality by suggesting that this increased lifespan might be accompanied by better overall health,” the authors wrote.</p> <h2>SOURCE:</h2> <p>Hongying Shi, PhD, Department of Epidemiology and Health Statistics, School of Public Health, Wenzhou Medical University, Wenzhou, China, led this study, which was published <a href="https://doi.org/10.1001/jamanetworkopen.2024.16300">online</a> in <em>JAMA Network Open</em>.</p> <h2>LIMITATIONS:</h2> <p>The measures of different behaviors were self-reported and may, therefore, be less accurate than objective measurement methods. Measurement error may have attenuated the effect of low levels of physical activity. The single exposure assessment at baseline may not reflect the long-term pattern of these activities.</p> <h2>DISCLOSURES:</h2> <p>The lead author was supported by the National Social Science Foundation Project of China and the Zhejiang Provincial Philosophy and Social Sciences Planning Project. A co-author and the Nurses’ Health Study were supported by the US National Institutes of Health. The authors declared no conflicts of interest.<span class="end"/></p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/trading-tv-time-physical-activity-boosts-healthy-aging-2024a1000c67">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