Novel approaches to treating NASH in diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:12

 

– The investigational oral agent cenicriviroc showed positive effects on liver fibrosis in adults with nonalcoholic steatohepatitis (NASH), many of whom had type 2 diabetes, in a phase 2b trial reported at the annual meeting of the European Association for the Study of Diabetes.

Dr. Henrik Landgren

Other data released at the meeting, which showed potential positive effects of novel or existing diabetes treatments on nonalcoholic fatty liver disease (NAFLD), included post hoc analyses of a phase 2b study with tirzepatide and a phase 3 study that combined exenatide and dapagliflozin.

Currently, no medications for NAFLD or NASH have been approved in the United States.

CENTAUR with cenicriviroc

Results of the previously reported CENTAUR trial showed that the antifibrotic effects of cenicriviroc, a dual chemokine receptor antagonist, were greatest in patients with more-severe liver disease (Hepatology. 2018;67[5]:1754-67). At the meeting, Henrik Landgren, PhD, of Allergan, presented data from the 2-year trial overall, and specifically in patients with advanced, stage 3 fibrosis.

CENTAUR was a randomized, double-blind, placebo-controlled, multinational study with 289 adults who had biopsy-confirmed NASH, an NAFLD Activity Score (NAS; range, 0-8; score of 5 or more diagnostic of NASH) of 4 or more, and stages 1-3 liver fibrosis as determined by the NASH clinical research network system (Contemp Clin Trials. 2016;47:356-65). The mean age of the patients enrolled at baseline was 54 years, the mean body mass index was 33.9 kg/m2, and just more than half the patients (52%) had type 2 diabetes.

The patients were randomized to three treatment arms: cenicriviroc 150 mg for 2 years; placebo for 1 year, then cenicriviroc 150 mg for 1 year; or placebo for 2 years. The primary endpoint was histologic improvement (reduction of 2 or more points in overall NAS, with reduction of 1 or more points in more than one category of the NAS without worsening of fibrosis at the end of year 1. The key secondary endpoint was complete NASH resolution without worsening of fibrosis at year 2.

Dr. Landgren reported that, at year 1, of the total number of patients, 28.6% of those receiving cenicriviroc achieved an improvement in fibrosis of one or more stages, compared with 19.0% of those receiving placebo. Of the 97 patients who had advanced fibrosis at baseline, 38.3% of those on cenicriviroc and 28.0% of those on placebo achieved the same endpoint.

Those effects were sustained at year 2, Dr. Landgren emphasized, with twice as many cenicriviroc- than placebo-treated patients achieving one or more stage improvement in fibrosis and no worsening of NASH at year 2 (60% and 30%, respectively), with more pronounced improvements in those who had advanced fibrosis at baseline (86% and 60%).

In addition, analyses of biomarkers suggested that cenicriviroc had systematic anti-inflammatory activity, with reductions observed in high-sensitivity C-reactive protein; fibrinogen; and levels of interleukin-6, IL-8, and IL-1-beta.

Dr. Landgren and colleagues noted that cenicriviroc provided antifibrotic benefit in patients with NASH and fibrosis. Those benefits were sustained through year 2 and were more pronounced in patients who had advanced fibrosis at baseline.

The safety of cenicriviroc was “comparable with placebo,” he said, suggesting that the data supported the phase 3 AURORA study that is currently recruiting.

 

 

Tirzepatide for NASH

Another approach worth exploring for the treatment of NASH, is the use of tirzepatide, a dual agonist of glucose-dependent insulinotropic polypeptide and the glucagonlike peptide–1 (GLP-1) receptor, according to Axel Haupt, MD, PhD, of Eli Lilly.

Tirzepatide (LY3298176) is currently under investigation for the treatment of type 2 diabetes, and Dr. Haupt reported data from a post hoc analysis of a double-blind, placebo-controlled, phase 2b study showing that “exploratory” serum markers of apoptosis and fibrosis – keratin-18 (K-18) and Pro-C3, respectively – were decreased from baseline to a greater extent in patients treated with tirzepatide than with placebo, while total adiponectin was increased. The latter is “thought to protect the liver from inflammation and fibrosis,” Dr. Haupt observed.

The main results of the trial were published last year (Lancet. 2018;392:2180-93) and showed that, after 26 weeks, there was a dose-dependent decrease in both glycated hemoglobin (HbA1c) and body weight with tirzepatide 10 mg and 15 mg, compared with placebo and an active comparator, dulaglutide 1.5 mg.

The study population was typical of type 2 diabetes: baseline HbA1c was 8.1%; the average body mass index was 32 kg/m2, with a diabetes duration of 5 years; and the main treatment (90%) had been metformin.

The rationale for the NASH-related biomarker analysis was that type 2 diabetes and NAFLD were known to be overlapping conditions, and weight loss had been shown to be an effective means of resolving NASH, Dr. Haupt said. In addition, a small “proof-of-concept” study with the GLP-1 receptor agonist liraglutide had suggested that these drugs may be effective in NASH.

Tirzepatide, at doses of 5, 10, and 15 mg, was associated with significant decreases in K-18 from baseline to week 26 and compared with placebo and the 1-mg tirzepatide dose. Mean baseline concentrations of K-18 were 394.4 U/L in the placebo group and reduced by 22.6 U/L by week 26. Corresponding baseline values for tirzepatide 5 mg were 375.8 U/L (change, –87.6 U/L); for 10 mg, 409.9 U/L (–157.8 U/L); and for 15 mg, 376.2 U/L (–110.6 U/L).

Dr. Haupt noted that a K-18 value of 250 U/L was considered a cutoff for a diagnosis of NASH. “So we really think that we have some NASH patients in this population,” he observed.

At week 26, Pro-C3 levels significantly decreased by 1.2 ng/mL from a baseline of 8.6 ng/mL with tirzepatide 15 mg, compared with an increase of 0.9 ng/mL from a baseline of 9.3 ng/mL for placebo (P less than .05). However, values of between 15-20 ng/mL would be expected for advanced fibrosis, Dr. Haupt said, “so we think we [don’t] have a lot of patients with advanced fibrosis, we have a lower grade of fibrosis or no fibrosis in our patient population.”

By week 26, adiponectin levels significantly increased by 0.9 mg/L from baseline, both with tirzepatide 10 mg (P less than .05) and 15 mg (P less than .05), compared with placebo (–0.1 mg/L; both P less than .05).

“This study was really designed as a type 2 diabetes efficacy study, so the NASH biomarker work is exploratory and only hypothesis generating,” Dr. Haupt noted. “We think there is overlap in type 2 diabetes and NASH, but it is not an ideal population to look into those biomarkers.” There are also other limitations, such as the baseline values across treatment groups not being matched, so there is likely to be some inconsistency in these data, he added.

That said, Dr. Haupt concluded that, “along with the weight-loss findings,” these exploratory biomarker findings supported the further evaluation of tirzepatide in patients with NASH.”

 

 

DURATION-8: Exenatide plus dapagliflozin

In another hypothesis-generating post hoc analysis, this time of the phase 3 DURATION-8 clinical trial, a combination of exenatide and dapagliflozin was found to have a beneficial effect on markers of hepatic steatosis and fibrosis in patients with type 2 diabetes.

Christian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
Sara Freeman/MDedge News
Dr. Christian Guja

“We have some good evidence that both GLP-1 receptor agonists and SGLT2 [sodium-glucose cotransporter 2] inhibitors may have benefits in reducing steatosis and even steatohepatitis in [patients with] type 2 diabetes. So the association of two diabetes drugs might provide an advantage. However, this had not previously been tested in a randomized, controlled trial,” observed Cristian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy in Bucharest, Romania.

The main aims of the DURATION-8 clinical trial, which ran for 104 weeks, was to compare the efficacy and safety of combining exenatide (2 mg, once a week) and dapagliflozin (10 mg, daily) with either exenatide 2 mg with placebo or dapagliflozin 10 mg with placebo. Results showed greater improved glycemic control and reductions in body weight and systolic BP with the exenatide-dapagliflozin combination.

A total of 685 patients were included in the post hoc analysis, of whom 228 had been treated with the combination, 228 with exenatide plus placebo, and 230 with dapagliflozin plus placebo. At baseline, levels of the markers of NAFLD and fibrosis that were assessed were similar between the groups. Between 81% and 93% of study participants had fatty liver or steatosis as defined by a Fatty Liver Index (FLI) of 60 or more or an overall NAFLD Liver Fat Score (NLFS) of –0.64 or higher. Between 9% and 13% of patients had liver fibrosis, as defined as an NAFLD Fibrosis Score (NFS) above 0.676, a Fibrosis-4 score (FIB-4) of 1.46 or more, or both.

At 28 weeks, the proportion of patients with biomarker scores suggestive of fatty liver disease or steatosis was significantly reduced from baseline with the exenatide-dapagliflozin combination (–10.5% for FLI of 60 or more; –6.5% for NLFS of –0.640 or more), Dr. Guja said, and biomarker scores suggestive of advanced fibrosis (NFS greater than 0.676; FIB-4 of 1.46 or more) were reduced by 4.1% and 3.6%, respectively.

At 28 and 52 weeks, the combination therapy showed stronger effects than exenatide and dapagliflozin alone in improving markers of hepatic steatosis (FLI: 28 weeks, –6.81, –3.90, –4.04; and 52 weeks, –6.23, –3.00, –4.58). The combination therapy also showed improvement for advanced fibrosis biomarkers at both time points (FIB-4: 28 weeks, –0.06, –0.03, –0.04; and 52 weeks, –0.05, –0.02, –0,04).

Dr. Guja noted that, although the study was not powered to assess the effect of on fatty liver, making all these data exploratory, this was the first analysis to describe improvements in biomarkers of fatty liver or steatosis and fibrosis from a large trial. “Some specific, dedicated, prospective trials are needed in the future to validate these findings.”

The CENTAUR study was funded by Allergan, of which Dr. Landgren is an employee. The phase 2b study with tirzepatide was supported by Eli Lilly. Dr. Haupt disclosed being an employee and also holding stocks in the company. The DURATION-8 study was sponsored by AstraZeneca. Dr. Guja disclosed that he had participated in scientific advisory boards and received consulting fees from AstraZeneca and other companies.

SOURCES: Landgren H et al. EASD 2019, Oral Presentation 179; Haupt A et al. EASD 2019, Oral Presentation 177; Guja C et al. EASD 2019, Oral Presentation 178.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– The investigational oral agent cenicriviroc showed positive effects on liver fibrosis in adults with nonalcoholic steatohepatitis (NASH), many of whom had type 2 diabetes, in a phase 2b trial reported at the annual meeting of the European Association for the Study of Diabetes.

Dr. Henrik Landgren

Other data released at the meeting, which showed potential positive effects of novel or existing diabetes treatments on nonalcoholic fatty liver disease (NAFLD), included post hoc analyses of a phase 2b study with tirzepatide and a phase 3 study that combined exenatide and dapagliflozin.

Currently, no medications for NAFLD or NASH have been approved in the United States.

CENTAUR with cenicriviroc

Results of the previously reported CENTAUR trial showed that the antifibrotic effects of cenicriviroc, a dual chemokine receptor antagonist, were greatest in patients with more-severe liver disease (Hepatology. 2018;67[5]:1754-67). At the meeting, Henrik Landgren, PhD, of Allergan, presented data from the 2-year trial overall, and specifically in patients with advanced, stage 3 fibrosis.

CENTAUR was a randomized, double-blind, placebo-controlled, multinational study with 289 adults who had biopsy-confirmed NASH, an NAFLD Activity Score (NAS; range, 0-8; score of 5 or more diagnostic of NASH) of 4 or more, and stages 1-3 liver fibrosis as determined by the NASH clinical research network system (Contemp Clin Trials. 2016;47:356-65). The mean age of the patients enrolled at baseline was 54 years, the mean body mass index was 33.9 kg/m2, and just more than half the patients (52%) had type 2 diabetes.

The patients were randomized to three treatment arms: cenicriviroc 150 mg for 2 years; placebo for 1 year, then cenicriviroc 150 mg for 1 year; or placebo for 2 years. The primary endpoint was histologic improvement (reduction of 2 or more points in overall NAS, with reduction of 1 or more points in more than one category of the NAS without worsening of fibrosis at the end of year 1. The key secondary endpoint was complete NASH resolution without worsening of fibrosis at year 2.

Dr. Landgren reported that, at year 1, of the total number of patients, 28.6% of those receiving cenicriviroc achieved an improvement in fibrosis of one or more stages, compared with 19.0% of those receiving placebo. Of the 97 patients who had advanced fibrosis at baseline, 38.3% of those on cenicriviroc and 28.0% of those on placebo achieved the same endpoint.

Those effects were sustained at year 2, Dr. Landgren emphasized, with twice as many cenicriviroc- than placebo-treated patients achieving one or more stage improvement in fibrosis and no worsening of NASH at year 2 (60% and 30%, respectively), with more pronounced improvements in those who had advanced fibrosis at baseline (86% and 60%).

In addition, analyses of biomarkers suggested that cenicriviroc had systematic anti-inflammatory activity, with reductions observed in high-sensitivity C-reactive protein; fibrinogen; and levels of interleukin-6, IL-8, and IL-1-beta.

Dr. Landgren and colleagues noted that cenicriviroc provided antifibrotic benefit in patients with NASH and fibrosis. Those benefits were sustained through year 2 and were more pronounced in patients who had advanced fibrosis at baseline.

The safety of cenicriviroc was “comparable with placebo,” he said, suggesting that the data supported the phase 3 AURORA study that is currently recruiting.

 

 

Tirzepatide for NASH

Another approach worth exploring for the treatment of NASH, is the use of tirzepatide, a dual agonist of glucose-dependent insulinotropic polypeptide and the glucagonlike peptide–1 (GLP-1) receptor, according to Axel Haupt, MD, PhD, of Eli Lilly.

Tirzepatide (LY3298176) is currently under investigation for the treatment of type 2 diabetes, and Dr. Haupt reported data from a post hoc analysis of a double-blind, placebo-controlled, phase 2b study showing that “exploratory” serum markers of apoptosis and fibrosis – keratin-18 (K-18) and Pro-C3, respectively – were decreased from baseline to a greater extent in patients treated with tirzepatide than with placebo, while total adiponectin was increased. The latter is “thought to protect the liver from inflammation and fibrosis,” Dr. Haupt observed.

The main results of the trial were published last year (Lancet. 2018;392:2180-93) and showed that, after 26 weeks, there was a dose-dependent decrease in both glycated hemoglobin (HbA1c) and body weight with tirzepatide 10 mg and 15 mg, compared with placebo and an active comparator, dulaglutide 1.5 mg.

The study population was typical of type 2 diabetes: baseline HbA1c was 8.1%; the average body mass index was 32 kg/m2, with a diabetes duration of 5 years; and the main treatment (90%) had been metformin.

The rationale for the NASH-related biomarker analysis was that type 2 diabetes and NAFLD were known to be overlapping conditions, and weight loss had been shown to be an effective means of resolving NASH, Dr. Haupt said. In addition, a small “proof-of-concept” study with the GLP-1 receptor agonist liraglutide had suggested that these drugs may be effective in NASH.

Tirzepatide, at doses of 5, 10, and 15 mg, was associated with significant decreases in K-18 from baseline to week 26 and compared with placebo and the 1-mg tirzepatide dose. Mean baseline concentrations of K-18 were 394.4 U/L in the placebo group and reduced by 22.6 U/L by week 26. Corresponding baseline values for tirzepatide 5 mg were 375.8 U/L (change, –87.6 U/L); for 10 mg, 409.9 U/L (–157.8 U/L); and for 15 mg, 376.2 U/L (–110.6 U/L).

Dr. Haupt noted that a K-18 value of 250 U/L was considered a cutoff for a diagnosis of NASH. “So we really think that we have some NASH patients in this population,” he observed.

At week 26, Pro-C3 levels significantly decreased by 1.2 ng/mL from a baseline of 8.6 ng/mL with tirzepatide 15 mg, compared with an increase of 0.9 ng/mL from a baseline of 9.3 ng/mL for placebo (P less than .05). However, values of between 15-20 ng/mL would be expected for advanced fibrosis, Dr. Haupt said, “so we think we [don’t] have a lot of patients with advanced fibrosis, we have a lower grade of fibrosis or no fibrosis in our patient population.”

By week 26, adiponectin levels significantly increased by 0.9 mg/L from baseline, both with tirzepatide 10 mg (P less than .05) and 15 mg (P less than .05), compared with placebo (–0.1 mg/L; both P less than .05).

“This study was really designed as a type 2 diabetes efficacy study, so the NASH biomarker work is exploratory and only hypothesis generating,” Dr. Haupt noted. “We think there is overlap in type 2 diabetes and NASH, but it is not an ideal population to look into those biomarkers.” There are also other limitations, such as the baseline values across treatment groups not being matched, so there is likely to be some inconsistency in these data, he added.

That said, Dr. Haupt concluded that, “along with the weight-loss findings,” these exploratory biomarker findings supported the further evaluation of tirzepatide in patients with NASH.”

 

 

DURATION-8: Exenatide plus dapagliflozin

In another hypothesis-generating post hoc analysis, this time of the phase 3 DURATION-8 clinical trial, a combination of exenatide and dapagliflozin was found to have a beneficial effect on markers of hepatic steatosis and fibrosis in patients with type 2 diabetes.

Christian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
Sara Freeman/MDedge News
Dr. Christian Guja

“We have some good evidence that both GLP-1 receptor agonists and SGLT2 [sodium-glucose cotransporter 2] inhibitors may have benefits in reducing steatosis and even steatohepatitis in [patients with] type 2 diabetes. So the association of two diabetes drugs might provide an advantage. However, this had not previously been tested in a randomized, controlled trial,” observed Cristian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy in Bucharest, Romania.

The main aims of the DURATION-8 clinical trial, which ran for 104 weeks, was to compare the efficacy and safety of combining exenatide (2 mg, once a week) and dapagliflozin (10 mg, daily) with either exenatide 2 mg with placebo or dapagliflozin 10 mg with placebo. Results showed greater improved glycemic control and reductions in body weight and systolic BP with the exenatide-dapagliflozin combination.

A total of 685 patients were included in the post hoc analysis, of whom 228 had been treated with the combination, 228 with exenatide plus placebo, and 230 with dapagliflozin plus placebo. At baseline, levels of the markers of NAFLD and fibrosis that were assessed were similar between the groups. Between 81% and 93% of study participants had fatty liver or steatosis as defined by a Fatty Liver Index (FLI) of 60 or more or an overall NAFLD Liver Fat Score (NLFS) of –0.64 or higher. Between 9% and 13% of patients had liver fibrosis, as defined as an NAFLD Fibrosis Score (NFS) above 0.676, a Fibrosis-4 score (FIB-4) of 1.46 or more, or both.

At 28 weeks, the proportion of patients with biomarker scores suggestive of fatty liver disease or steatosis was significantly reduced from baseline with the exenatide-dapagliflozin combination (–10.5% for FLI of 60 or more; –6.5% for NLFS of –0.640 or more), Dr. Guja said, and biomarker scores suggestive of advanced fibrosis (NFS greater than 0.676; FIB-4 of 1.46 or more) were reduced by 4.1% and 3.6%, respectively.

At 28 and 52 weeks, the combination therapy showed stronger effects than exenatide and dapagliflozin alone in improving markers of hepatic steatosis (FLI: 28 weeks, –6.81, –3.90, –4.04; and 52 weeks, –6.23, –3.00, –4.58). The combination therapy also showed improvement for advanced fibrosis biomarkers at both time points (FIB-4: 28 weeks, –0.06, –0.03, –0.04; and 52 weeks, –0.05, –0.02, –0,04).

Dr. Guja noted that, although the study was not powered to assess the effect of on fatty liver, making all these data exploratory, this was the first analysis to describe improvements in biomarkers of fatty liver or steatosis and fibrosis from a large trial. “Some specific, dedicated, prospective trials are needed in the future to validate these findings.”

The CENTAUR study was funded by Allergan, of which Dr. Landgren is an employee. The phase 2b study with tirzepatide was supported by Eli Lilly. Dr. Haupt disclosed being an employee and also holding stocks in the company. The DURATION-8 study was sponsored by AstraZeneca. Dr. Guja disclosed that he had participated in scientific advisory boards and received consulting fees from AstraZeneca and other companies.

SOURCES: Landgren H et al. EASD 2019, Oral Presentation 179; Haupt A et al. EASD 2019, Oral Presentation 177; Guja C et al. EASD 2019, Oral Presentation 178.

 

– The investigational oral agent cenicriviroc showed positive effects on liver fibrosis in adults with nonalcoholic steatohepatitis (NASH), many of whom had type 2 diabetes, in a phase 2b trial reported at the annual meeting of the European Association for the Study of Diabetes.

Dr. Henrik Landgren

Other data released at the meeting, which showed potential positive effects of novel or existing diabetes treatments on nonalcoholic fatty liver disease (NAFLD), included post hoc analyses of a phase 2b study with tirzepatide and a phase 3 study that combined exenatide and dapagliflozin.

Currently, no medications for NAFLD or NASH have been approved in the United States.

CENTAUR with cenicriviroc

Results of the previously reported CENTAUR trial showed that the antifibrotic effects of cenicriviroc, a dual chemokine receptor antagonist, were greatest in patients with more-severe liver disease (Hepatology. 2018;67[5]:1754-67). At the meeting, Henrik Landgren, PhD, of Allergan, presented data from the 2-year trial overall, and specifically in patients with advanced, stage 3 fibrosis.

CENTAUR was a randomized, double-blind, placebo-controlled, multinational study with 289 adults who had biopsy-confirmed NASH, an NAFLD Activity Score (NAS; range, 0-8; score of 5 or more diagnostic of NASH) of 4 or more, and stages 1-3 liver fibrosis as determined by the NASH clinical research network system (Contemp Clin Trials. 2016;47:356-65). The mean age of the patients enrolled at baseline was 54 years, the mean body mass index was 33.9 kg/m2, and just more than half the patients (52%) had type 2 diabetes.

The patients were randomized to three treatment arms: cenicriviroc 150 mg for 2 years; placebo for 1 year, then cenicriviroc 150 mg for 1 year; or placebo for 2 years. The primary endpoint was histologic improvement (reduction of 2 or more points in overall NAS, with reduction of 1 or more points in more than one category of the NAS without worsening of fibrosis at the end of year 1. The key secondary endpoint was complete NASH resolution without worsening of fibrosis at year 2.

Dr. Landgren reported that, at year 1, of the total number of patients, 28.6% of those receiving cenicriviroc achieved an improvement in fibrosis of one or more stages, compared with 19.0% of those receiving placebo. Of the 97 patients who had advanced fibrosis at baseline, 38.3% of those on cenicriviroc and 28.0% of those on placebo achieved the same endpoint.

Those effects were sustained at year 2, Dr. Landgren emphasized, with twice as many cenicriviroc- than placebo-treated patients achieving one or more stage improvement in fibrosis and no worsening of NASH at year 2 (60% and 30%, respectively), with more pronounced improvements in those who had advanced fibrosis at baseline (86% and 60%).

In addition, analyses of biomarkers suggested that cenicriviroc had systematic anti-inflammatory activity, with reductions observed in high-sensitivity C-reactive protein; fibrinogen; and levels of interleukin-6, IL-8, and IL-1-beta.

Dr. Landgren and colleagues noted that cenicriviroc provided antifibrotic benefit in patients with NASH and fibrosis. Those benefits were sustained through year 2 and were more pronounced in patients who had advanced fibrosis at baseline.

The safety of cenicriviroc was “comparable with placebo,” he said, suggesting that the data supported the phase 3 AURORA study that is currently recruiting.

 

 

Tirzepatide for NASH

Another approach worth exploring for the treatment of NASH, is the use of tirzepatide, a dual agonist of glucose-dependent insulinotropic polypeptide and the glucagonlike peptide–1 (GLP-1) receptor, according to Axel Haupt, MD, PhD, of Eli Lilly.

Tirzepatide (LY3298176) is currently under investigation for the treatment of type 2 diabetes, and Dr. Haupt reported data from a post hoc analysis of a double-blind, placebo-controlled, phase 2b study showing that “exploratory” serum markers of apoptosis and fibrosis – keratin-18 (K-18) and Pro-C3, respectively – were decreased from baseline to a greater extent in patients treated with tirzepatide than with placebo, while total adiponectin was increased. The latter is “thought to protect the liver from inflammation and fibrosis,” Dr. Haupt observed.

The main results of the trial were published last year (Lancet. 2018;392:2180-93) and showed that, after 26 weeks, there was a dose-dependent decrease in both glycated hemoglobin (HbA1c) and body weight with tirzepatide 10 mg and 15 mg, compared with placebo and an active comparator, dulaglutide 1.5 mg.

The study population was typical of type 2 diabetes: baseline HbA1c was 8.1%; the average body mass index was 32 kg/m2, with a diabetes duration of 5 years; and the main treatment (90%) had been metformin.

The rationale for the NASH-related biomarker analysis was that type 2 diabetes and NAFLD were known to be overlapping conditions, and weight loss had been shown to be an effective means of resolving NASH, Dr. Haupt said. In addition, a small “proof-of-concept” study with the GLP-1 receptor agonist liraglutide had suggested that these drugs may be effective in NASH.

Tirzepatide, at doses of 5, 10, and 15 mg, was associated with significant decreases in K-18 from baseline to week 26 and compared with placebo and the 1-mg tirzepatide dose. Mean baseline concentrations of K-18 were 394.4 U/L in the placebo group and reduced by 22.6 U/L by week 26. Corresponding baseline values for tirzepatide 5 mg were 375.8 U/L (change, –87.6 U/L); for 10 mg, 409.9 U/L (–157.8 U/L); and for 15 mg, 376.2 U/L (–110.6 U/L).

Dr. Haupt noted that a K-18 value of 250 U/L was considered a cutoff for a diagnosis of NASH. “So we really think that we have some NASH patients in this population,” he observed.

At week 26, Pro-C3 levels significantly decreased by 1.2 ng/mL from a baseline of 8.6 ng/mL with tirzepatide 15 mg, compared with an increase of 0.9 ng/mL from a baseline of 9.3 ng/mL for placebo (P less than .05). However, values of between 15-20 ng/mL would be expected for advanced fibrosis, Dr. Haupt said, “so we think we [don’t] have a lot of patients with advanced fibrosis, we have a lower grade of fibrosis or no fibrosis in our patient population.”

By week 26, adiponectin levels significantly increased by 0.9 mg/L from baseline, both with tirzepatide 10 mg (P less than .05) and 15 mg (P less than .05), compared with placebo (–0.1 mg/L; both P less than .05).

“This study was really designed as a type 2 diabetes efficacy study, so the NASH biomarker work is exploratory and only hypothesis generating,” Dr. Haupt noted. “We think there is overlap in type 2 diabetes and NASH, but it is not an ideal population to look into those biomarkers.” There are also other limitations, such as the baseline values across treatment groups not being matched, so there is likely to be some inconsistency in these data, he added.

That said, Dr. Haupt concluded that, “along with the weight-loss findings,” these exploratory biomarker findings supported the further evaluation of tirzepatide in patients with NASH.”

 

 

DURATION-8: Exenatide plus dapagliflozin

In another hypothesis-generating post hoc analysis, this time of the phase 3 DURATION-8 clinical trial, a combination of exenatide and dapagliflozin was found to have a beneficial effect on markers of hepatic steatosis and fibrosis in patients with type 2 diabetes.

Christian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
Sara Freeman/MDedge News
Dr. Christian Guja

“We have some good evidence that both GLP-1 receptor agonists and SGLT2 [sodium-glucose cotransporter 2] inhibitors may have benefits in reducing steatosis and even steatohepatitis in [patients with] type 2 diabetes. So the association of two diabetes drugs might provide an advantage. However, this had not previously been tested in a randomized, controlled trial,” observed Cristian Guja, MD, PhD, of Carol Davila University of Medicine and Pharmacy in Bucharest, Romania.

The main aims of the DURATION-8 clinical trial, which ran for 104 weeks, was to compare the efficacy and safety of combining exenatide (2 mg, once a week) and dapagliflozin (10 mg, daily) with either exenatide 2 mg with placebo or dapagliflozin 10 mg with placebo. Results showed greater improved glycemic control and reductions in body weight and systolic BP with the exenatide-dapagliflozin combination.

A total of 685 patients were included in the post hoc analysis, of whom 228 had been treated with the combination, 228 with exenatide plus placebo, and 230 with dapagliflozin plus placebo. At baseline, levels of the markers of NAFLD and fibrosis that were assessed were similar between the groups. Between 81% and 93% of study participants had fatty liver or steatosis as defined by a Fatty Liver Index (FLI) of 60 or more or an overall NAFLD Liver Fat Score (NLFS) of –0.64 or higher. Between 9% and 13% of patients had liver fibrosis, as defined as an NAFLD Fibrosis Score (NFS) above 0.676, a Fibrosis-4 score (FIB-4) of 1.46 or more, or both.

At 28 weeks, the proportion of patients with biomarker scores suggestive of fatty liver disease or steatosis was significantly reduced from baseline with the exenatide-dapagliflozin combination (–10.5% for FLI of 60 or more; –6.5% for NLFS of –0.640 or more), Dr. Guja said, and biomarker scores suggestive of advanced fibrosis (NFS greater than 0.676; FIB-4 of 1.46 or more) were reduced by 4.1% and 3.6%, respectively.

At 28 and 52 weeks, the combination therapy showed stronger effects than exenatide and dapagliflozin alone in improving markers of hepatic steatosis (FLI: 28 weeks, –6.81, –3.90, –4.04; and 52 weeks, –6.23, –3.00, –4.58). The combination therapy also showed improvement for advanced fibrosis biomarkers at both time points (FIB-4: 28 weeks, –0.06, –0.03, –0.04; and 52 weeks, –0.05, –0.02, –0,04).

Dr. Guja noted that, although the study was not powered to assess the effect of on fatty liver, making all these data exploratory, this was the first analysis to describe improvements in biomarkers of fatty liver or steatosis and fibrosis from a large trial. “Some specific, dedicated, prospective trials are needed in the future to validate these findings.”

The CENTAUR study was funded by Allergan, of which Dr. Landgren is an employee. The phase 2b study with tirzepatide was supported by Eli Lilly. Dr. Haupt disclosed being an employee and also holding stocks in the company. The DURATION-8 study was sponsored by AstraZeneca. Dr. Guja disclosed that he had participated in scientific advisory boards and received consulting fees from AstraZeneca and other companies.

SOURCES: Landgren H et al. EASD 2019, Oral Presentation 179; Haupt A et al. EASD 2019, Oral Presentation 177; Guja C et al. EASD 2019, Oral Presentation 178.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Cardiometabolic risk burden is high in under-50s with type 2 diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:12

– People diagnosed with type 2 diabetes when they are 18-39 years old have significantly higher cardiometabolic risk burden, compared with older people, according to the results of a large study from the United Kingdom presented at the annual meeting of the European Association for the Study of Diabetes.

Sanjoy Paul, PhD, director of the Melbourne EpiCentre at the University of Melbourne (Australia) at a press conference
Sara Freeman/MDedge News
Dr. Sanjoy Paul

Patients in that younger age group were found to have higher glycated hemoglobin (HbA1c) levels, along with higher levels of low-density lipoprotein cholesterol and higher body weight.

“We wanted to evaluate the population-level trend in the incidence of young-onset type 2 diabetes in the United Kingdom, compared with later-onset diabetes,” said senior study author Sanjoy Paul, PhD, the director of the Melbourne EpiCentre at the University of Melbourne at a press briefing during the meeting.

Other aims of the study were to compare temporal trends in the incidence of atherosclerotic cardiovascular disease in younger and older patients with type 2 diabetes, and to see how being “high risk” at diagnosis affected patients’ risk of ASCVD and subsequent risk of death.

High-risk status was defined as having at least two of the risk factors for ASCVD – smoking, high systolic blood pressure, high low-density lipoprotein cholesterol, or chronic kidney disease.

The investigators searched a nationally representative sample of primary care electronic medical records from The Health Improvement Network (THIN) database to find incident cases of type 2 diabetes that occurred between 2000 and 2017, with a total of 370,854 cases identified.

At diagnosis of type 2 diabetes, 8% of the sample (n = 29,678) was aged 18-39 years; 15% (n = 56,798), 40-49 years; 25% (n = 93,698), 50-59 years; 29% (n = 107,261), 60-69 years; and 23% (n = 83,419), 70-79 years. Follow-up was just more than 6 years.

Baseline HbA1c in the respective age groups was 8.6%, 8.4%, 8.1%, 7.8%, and 7.6%, with more than 55% of patients in the two youngest age groups having an HbA1c of 7.5% or higher, compared with 34%-47% in the three oldest age groups.

The percentage of patients with a high LDL cholesterol value (2.6 mmol/L or higher in those without ASCVD, and 1.8 or higher in those with ASCVD) was 71%, 75%, 74%, 69%, and 65%, from the youngest to oldest age groups. A respective 71%, 70%, 66%, 57%, and 44% of the patients had a body mass index of 35 kg/m2 or higher.

Few younger patients had ASCVD at diagnosis (2% of the 18-39 age group; 6% of the 40-49 group), with higher rates in the older age groups (13% of the 50-59 group; 23% of the 60-69 group; and 33% of the 70-79 group).

The percentage of patients considered to be at high risk of ASCVD at diagnosis was 23%, 37%, 45%, 50%, and 53%, respectively, across the five age groups.

Although high systolic blood pressure (SBP; 130 mmHg in those with ASCVD, 140 mmHg in those without) was more common in the older age groups (52% at 50-59 years; 60% at 60-69 years, and 64% at 70-79 years,) a substantial proportion of the younger patients also had a high SBP (27% at 18-39 years and 41% at 40-49 years).

Digsu Koye, PhD, a postdoctoral research fellow at the also of the Melbourne EpiCentre at the University of Melbourne (Australia) at a lectern
Sara Freeman/MDedge News
Dr. Digsu Koye


Digsu Koye, PhD, also of the Melbourne EpiCentre, presented the main findings of the study during the meeting, noting that the proportion of people diagnosed when they were younger than 50 years remained stable between 2000 and 2017, with a marginal increase in those diagnosed when they were aged 50-59 years, and a decline in those diagnosed when they were older than 70 years.

In the youngest and oldest age groups, equal numbers of men and women were diagnosed with type 2 diabetes, and more women than men were diagnosed in the 60-69 age group, Dr. Koye said. However, for the 40-49 and 50-59 age groups, there were more men than women diagnosed with type 2 diabetes.

Patients were followed for an average of just more than 6 years. “The rate of atherosclerotic cardiovascular disease was declining in all age categories during 2000-2006, but after that, we saw a stable and consistent pattern for all age categories after 2007,” Dr. Koye observed.

In regard to all-cause mortality, there was a 30% decline in the oldest age group (70-79 years), and a 20% decline in the 60-69 age group, but there was no significant decline in the younger age groups, he added.

The investigators determined the average time to event (ASCVD or all-cause mortality) by high-risk status at type 2 diabetes diagnosis for each age group. These analyses showed that there was little difference between the high- and low-risk groups for the average time to ASCVD or all-cause mortality in the youngest age group, with wider differences in the older patients of 1-2 years for ASCVD and 0.5-2 years for all-cause mortality.

Dr. Koye noted that people with young-onset type 2 diabetes had a risk of ASCVD or all-cause mortality that was similar to that of older people, irrespective of whether or not they were considered to be at high or low risk of events. “So we need a more focused treatment strategy for the youngest age group, irrespective of the cardiometabolic risk level at diagnosis,” he said.

Dr. Paul and Dr. Koye reported having no conflicts of interest.

SOURCE: Koye D et al. EASD 2019, Abstract 82.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– People diagnosed with type 2 diabetes when they are 18-39 years old have significantly higher cardiometabolic risk burden, compared with older people, according to the results of a large study from the United Kingdom presented at the annual meeting of the European Association for the Study of Diabetes.

Sanjoy Paul, PhD, director of the Melbourne EpiCentre at the University of Melbourne (Australia) at a press conference
Sara Freeman/MDedge News
Dr. Sanjoy Paul

Patients in that younger age group were found to have higher glycated hemoglobin (HbA1c) levels, along with higher levels of low-density lipoprotein cholesterol and higher body weight.

“We wanted to evaluate the population-level trend in the incidence of young-onset type 2 diabetes in the United Kingdom, compared with later-onset diabetes,” said senior study author Sanjoy Paul, PhD, the director of the Melbourne EpiCentre at the University of Melbourne at a press briefing during the meeting.

Other aims of the study were to compare temporal trends in the incidence of atherosclerotic cardiovascular disease in younger and older patients with type 2 diabetes, and to see how being “high risk” at diagnosis affected patients’ risk of ASCVD and subsequent risk of death.

High-risk status was defined as having at least two of the risk factors for ASCVD – smoking, high systolic blood pressure, high low-density lipoprotein cholesterol, or chronic kidney disease.

The investigators searched a nationally representative sample of primary care electronic medical records from The Health Improvement Network (THIN) database to find incident cases of type 2 diabetes that occurred between 2000 and 2017, with a total of 370,854 cases identified.

At diagnosis of type 2 diabetes, 8% of the sample (n = 29,678) was aged 18-39 years; 15% (n = 56,798), 40-49 years; 25% (n = 93,698), 50-59 years; 29% (n = 107,261), 60-69 years; and 23% (n = 83,419), 70-79 years. Follow-up was just more than 6 years.

Baseline HbA1c in the respective age groups was 8.6%, 8.4%, 8.1%, 7.8%, and 7.6%, with more than 55% of patients in the two youngest age groups having an HbA1c of 7.5% or higher, compared with 34%-47% in the three oldest age groups.

The percentage of patients with a high LDL cholesterol value (2.6 mmol/L or higher in those without ASCVD, and 1.8 or higher in those with ASCVD) was 71%, 75%, 74%, 69%, and 65%, from the youngest to oldest age groups. A respective 71%, 70%, 66%, 57%, and 44% of the patients had a body mass index of 35 kg/m2 or higher.

Few younger patients had ASCVD at diagnosis (2% of the 18-39 age group; 6% of the 40-49 group), with higher rates in the older age groups (13% of the 50-59 group; 23% of the 60-69 group; and 33% of the 70-79 group).

The percentage of patients considered to be at high risk of ASCVD at diagnosis was 23%, 37%, 45%, 50%, and 53%, respectively, across the five age groups.

Although high systolic blood pressure (SBP; 130 mmHg in those with ASCVD, 140 mmHg in those without) was more common in the older age groups (52% at 50-59 years; 60% at 60-69 years, and 64% at 70-79 years,) a substantial proportion of the younger patients also had a high SBP (27% at 18-39 years and 41% at 40-49 years).

Digsu Koye, PhD, a postdoctoral research fellow at the also of the Melbourne EpiCentre at the University of Melbourne (Australia) at a lectern
Sara Freeman/MDedge News
Dr. Digsu Koye


Digsu Koye, PhD, also of the Melbourne EpiCentre, presented the main findings of the study during the meeting, noting that the proportion of people diagnosed when they were younger than 50 years remained stable between 2000 and 2017, with a marginal increase in those diagnosed when they were aged 50-59 years, and a decline in those diagnosed when they were older than 70 years.

In the youngest and oldest age groups, equal numbers of men and women were diagnosed with type 2 diabetes, and more women than men were diagnosed in the 60-69 age group, Dr. Koye said. However, for the 40-49 and 50-59 age groups, there were more men than women diagnosed with type 2 diabetes.

Patients were followed for an average of just more than 6 years. “The rate of atherosclerotic cardiovascular disease was declining in all age categories during 2000-2006, but after that, we saw a stable and consistent pattern for all age categories after 2007,” Dr. Koye observed.

In regard to all-cause mortality, there was a 30% decline in the oldest age group (70-79 years), and a 20% decline in the 60-69 age group, but there was no significant decline in the younger age groups, he added.

The investigators determined the average time to event (ASCVD or all-cause mortality) by high-risk status at type 2 diabetes diagnosis for each age group. These analyses showed that there was little difference between the high- and low-risk groups for the average time to ASCVD or all-cause mortality in the youngest age group, with wider differences in the older patients of 1-2 years for ASCVD and 0.5-2 years for all-cause mortality.

Dr. Koye noted that people with young-onset type 2 diabetes had a risk of ASCVD or all-cause mortality that was similar to that of older people, irrespective of whether or not they were considered to be at high or low risk of events. “So we need a more focused treatment strategy for the youngest age group, irrespective of the cardiometabolic risk level at diagnosis,” he said.

Dr. Paul and Dr. Koye reported having no conflicts of interest.

SOURCE: Koye D et al. EASD 2019, Abstract 82.
 

– People diagnosed with type 2 diabetes when they are 18-39 years old have significantly higher cardiometabolic risk burden, compared with older people, according to the results of a large study from the United Kingdom presented at the annual meeting of the European Association for the Study of Diabetes.

Sanjoy Paul, PhD, director of the Melbourne EpiCentre at the University of Melbourne (Australia) at a press conference
Sara Freeman/MDedge News
Dr. Sanjoy Paul

Patients in that younger age group were found to have higher glycated hemoglobin (HbA1c) levels, along with higher levels of low-density lipoprotein cholesterol and higher body weight.

“We wanted to evaluate the population-level trend in the incidence of young-onset type 2 diabetes in the United Kingdom, compared with later-onset diabetes,” said senior study author Sanjoy Paul, PhD, the director of the Melbourne EpiCentre at the University of Melbourne at a press briefing during the meeting.

Other aims of the study were to compare temporal trends in the incidence of atherosclerotic cardiovascular disease in younger and older patients with type 2 diabetes, and to see how being “high risk” at diagnosis affected patients’ risk of ASCVD and subsequent risk of death.

High-risk status was defined as having at least two of the risk factors for ASCVD – smoking, high systolic blood pressure, high low-density lipoprotein cholesterol, or chronic kidney disease.

The investigators searched a nationally representative sample of primary care electronic medical records from The Health Improvement Network (THIN) database to find incident cases of type 2 diabetes that occurred between 2000 and 2017, with a total of 370,854 cases identified.

At diagnosis of type 2 diabetes, 8% of the sample (n = 29,678) was aged 18-39 years; 15% (n = 56,798), 40-49 years; 25% (n = 93,698), 50-59 years; 29% (n = 107,261), 60-69 years; and 23% (n = 83,419), 70-79 years. Follow-up was just more than 6 years.

Baseline HbA1c in the respective age groups was 8.6%, 8.4%, 8.1%, 7.8%, and 7.6%, with more than 55% of patients in the two youngest age groups having an HbA1c of 7.5% or higher, compared with 34%-47% in the three oldest age groups.

The percentage of patients with a high LDL cholesterol value (2.6 mmol/L or higher in those without ASCVD, and 1.8 or higher in those with ASCVD) was 71%, 75%, 74%, 69%, and 65%, from the youngest to oldest age groups. A respective 71%, 70%, 66%, 57%, and 44% of the patients had a body mass index of 35 kg/m2 or higher.

Few younger patients had ASCVD at diagnosis (2% of the 18-39 age group; 6% of the 40-49 group), with higher rates in the older age groups (13% of the 50-59 group; 23% of the 60-69 group; and 33% of the 70-79 group).

The percentage of patients considered to be at high risk of ASCVD at diagnosis was 23%, 37%, 45%, 50%, and 53%, respectively, across the five age groups.

Although high systolic blood pressure (SBP; 130 mmHg in those with ASCVD, 140 mmHg in those without) was more common in the older age groups (52% at 50-59 years; 60% at 60-69 years, and 64% at 70-79 years,) a substantial proportion of the younger patients also had a high SBP (27% at 18-39 years and 41% at 40-49 years).

Digsu Koye, PhD, a postdoctoral research fellow at the also of the Melbourne EpiCentre at the University of Melbourne (Australia) at a lectern
Sara Freeman/MDedge News
Dr. Digsu Koye


Digsu Koye, PhD, also of the Melbourne EpiCentre, presented the main findings of the study during the meeting, noting that the proportion of people diagnosed when they were younger than 50 years remained stable between 2000 and 2017, with a marginal increase in those diagnosed when they were aged 50-59 years, and a decline in those diagnosed when they were older than 70 years.

In the youngest and oldest age groups, equal numbers of men and women were diagnosed with type 2 diabetes, and more women than men were diagnosed in the 60-69 age group, Dr. Koye said. However, for the 40-49 and 50-59 age groups, there were more men than women diagnosed with type 2 diabetes.

Patients were followed for an average of just more than 6 years. “The rate of atherosclerotic cardiovascular disease was declining in all age categories during 2000-2006, but after that, we saw a stable and consistent pattern for all age categories after 2007,” Dr. Koye observed.

In regard to all-cause mortality, there was a 30% decline in the oldest age group (70-79 years), and a 20% decline in the 60-69 age group, but there was no significant decline in the younger age groups, he added.

The investigators determined the average time to event (ASCVD or all-cause mortality) by high-risk status at type 2 diabetes diagnosis for each age group. These analyses showed that there was little difference between the high- and low-risk groups for the average time to ASCVD or all-cause mortality in the youngest age group, with wider differences in the older patients of 1-2 years for ASCVD and 0.5-2 years for all-cause mortality.

Dr. Koye noted that people with young-onset type 2 diabetes had a risk of ASCVD or all-cause mortality that was similar to that of older people, irrespective of whether or not they were considered to be at high or low risk of events. “So we need a more focused treatment strategy for the youngest age group, irrespective of the cardiometabolic risk level at diagnosis,” he said.

Dr. Paul and Dr. Koye reported having no conflicts of interest.

SOURCE: Koye D et al. EASD 2019, Abstract 82.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Ketoacidosis is on the rise in children with type 1 diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:12

– As many as 40%-60% of children have diabetic ketoacidosis (DKA) at the time of being diagnosed with type 1 diabetes, according to data from two U.S. analyses – and the figures have been rising for the past 10 years.

Between 2010 and 2017, the prevalence of DKA at diagnosis in children who were followed up at the Barbara Davies Cancer Center in Denver (n = 2,429) went from 41% to 59%, with a 7% annual rise, Arleta Rewers, MD, PhD, of Children’s Hospital Colorado, Denver, reported at the annual meeting of the European Association for the Study of Diabetes.

Meanwhile, in another analysis that included multiple U.S. centers and about 7,600 cases of youth-onset type 1 diabetes, the overall prevalence of DKA at diagnosis was 38.5% between 2010 and 2016. However, the prevalence had increased from 35% in 2010 to 40.6% in 2016, according to Elizabeth T. Jensen, MPH, PhD, of Wake Forest University, Winston-Salem, N.C. The annual increase in prevalence of DKA at diagnosis of type 1 disease was 2%, adjusted for sociodemographic factors.
 

Rising prevalence

“DKA occurs most commonly at the time of type 1 diabetes diagnosis,” observed Dr. Jensen, who noted that “in the United States, among children, it’s younger children, uninsured or underinsured children, and children from minority racial or ethnic groups, who are at greatest risk.”

Dr. Elizabeth T. Jensen, Wake Forest University, Winston-Salem, N.C.
Sara Freeman/MDedge News
Dr. Elizabeth T. Jensen

Dr. Jensen and colleagues had previously shown that the prevalence of DKA at diagnosis was around 30% between 2002 and 2010, with no significant change in its prevalence. However, more recent reports from referral-based, single-center studies had suggested there was an increase, and that led her and her colleagues to take a closer look at the data.

To characterize the risk factors for DKA and the prevalence of DKA over time, Dr. Jensen and her team used the SEARCH for Diabetes in Youth database, which, she said, was “uniquely suited” for this purpose. SEARCH is a population-based, multicenter study conducted in centers in five U.S. states: South Carolina, Ohio, Colorado, California, and Washington.

A diagnosis of DKA was based on blood bicarbonate levels of less than 15 mmol/L, a venous pH of less than 7.25 or arterial or capillary pH of less than 7.3, or if there was any documentation of a DKA diagnosis.

As expected, the prevalence of DKA was highest in the youngest age group (0-4 years), Dr. Jensen said, but the increase in prevalence in that group was no different from the increases seen over time in the other age groups (5-9 years, 10-14 years, and 15 years or older).

There were no differences in the prevalence of DKA between the sexes, although there was a general increase over time. Similar trends were seen in DKA prevalence by race or ethnicity and by season, or time of year.

Of note, higher rates of DKA were seen in children who were covered by public health insurance, than in those covered by private insurance, although there was no difference in the rate of increase in DKA prevalence between the two groups. Dr. Jensen noted that only 64% of this study population had private insurance.

She said that future research in this area would need to look at the economic drivers and the “changing landscape of health insurance coverage in the United States.”
 

 

 

Expansion in health coverage

In presenting the findings of a study showing an increase in the prevalence of DKA at diagnosis of type 1 diabetes in children in Colorado from 2010 to 2017, Dr. Rewers said that the increase “paradoxically occurred” at a time of increasing health insurance coverage, a reference to the expansion of Medicaid during 2008-2012 and implementation in 2013 of the Affordable Care Act.

“Our group in Colorado has followed the frequency of DKA for almost 2 decades,” Dr. Rewers said. It’s important to study DKA as it is linked to worse glycemic control – with children with DKA having an HbA1c level of around 1% higher than those without DKA – and the potential for future, long-term complications.

Dr. Rewers noted that the increase in DKA at diagnosis of type 1 diabetes was more rapid in the children who had private rather than public health insurance. Of 1,187 patients with DKA, 57% had private health insurance, and 37% had public insurance, compared with 66% and 28%, respectively, in those without DKA. In 2010, the prevalence of DKA at diagnosis was 35.3% in those who were privately insured and 52.2% of those with public health insurance, but by 2017, a similar percentage of DKA was seen in the privately and publicly insured children (59.6% and 58.5%, respectively).

She said one possible explanation for that might be that “increased enrollment in high-deductible insurance plans could discourage families with private insurance from seeking timely care.”

Another explanation is that there is a low awareness of type 1 diabetes in the general population, she added. “Educational campaigns and autoimmunity screening have been shown to reduce DKA at diabetes diagnosis, but unfortunately they are not used widely at this point.”
 

Identifying at-risk children

“Diabetic ketoacidosis is a serious complication of diabetes [and] is difficult to diagnose because of the variability of the symptoms, said Angela Ibald-Mulli, PhD, who presented the findings of a retrospective cohort study in which she and her colleagues used a “discovery algorithm” called Q-Finder to identify the predictive factors for DKA in youth with type 1 diabetes, based on data from the Diabetes Prospective Follow-up Registry (DPV).

Dr. Angela Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi in Paris
Sara Freeman/MDedge News
Dr. Angela Ibald-Mulli

“The better we know the risk factors, the better we can care for our patients,” she emphasized.

The investigators obtained data on 108,223 patients with a diagnosis of type 1 disease and with more than two visits related to diabetes. The prevalence of DKA – defined as a pH of less than 7.3 during hospitalization occurring at least 10 days after the onset of type 1 diabetes – was 5.2%, said Dr. Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi, Paris.

A total of 129 different features were considered for their association with DKA – including comorbidities, sociodemographic factors, laboratory values, and concomitant medications – and were then used to identify, test, and the validate likely risk profiles.

After comparing the characteristics of patients with and without DKA, eight significant factors, all of which have been reported previously in the DPV cohort, were seen: younger age, lower body weight, higher HbA1c, younger age at onset of T1D; shorter disease duration; having a migration background; being less active; and having had more medical visits.

The investigators used the algorithm, and found 11 distinct profiles associated with DKA: an HbA1c higher than 8.87%; being aged 6-10 years; being aged 11-15 years; a diagnosis of nephropathy; DKA being present at onset; a prevalence of hypoglycemia with coma; a diagnosis of thyroiditis; a standardized body mass index lower than 16.9; not using short-acting insulin; younger than age 15 years; and not using continuous glucose monitoring.

Almost two-thirds of patients (64.7%) belonged to at least one of these risk profiles, Dr. Ibald-Mulli observed, with 7.1% of them having DKA, compared with 1.6% who belonged to none of the profiles.

Dr. Ibald-Mulli said it was important to note that the DKA risk profiles could overlap. “The more profiles a patient belongs to, the higher is the risk of having DKA,” she emphasized, adding that most patients (88.8%) with DKA belonged to just one profile, and fewer than 5% belonged to three or more profiles.

“Overall, the results of the algorithm confirmed known risk-factor profiles that had been previously identified by conventional statistical methods,” she concluded. It also provided “additional insights that can be further explored.”

SEARCH is funded by the Centers for Disease and Prevention and the National Institute of Diabetes and Digestive and Kidney Diseases. The DPV Registry is funded by multiple sponsors, including the European Federation for the Study of Diabetes and other academic institutions with the support of several commercial partners. Sanofi sponsored the study presented by Dr. Ibald-Mulli. Dr. Rewers made no disclosures, and Dr. Jensen did not have any conflicts of interest to declare. Dr. Ibald-Mulli is an employee of Sanofi.

 

SOURCE: Rewers A et al. EASD 2019, Abstract 115; Jensen E et al. EASD 2019, Abstract 116; Ibald-Mulli A et al. EASD 2019, Abstract 117.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– As many as 40%-60% of children have diabetic ketoacidosis (DKA) at the time of being diagnosed with type 1 diabetes, according to data from two U.S. analyses – and the figures have been rising for the past 10 years.

Between 2010 and 2017, the prevalence of DKA at diagnosis in children who were followed up at the Barbara Davies Cancer Center in Denver (n = 2,429) went from 41% to 59%, with a 7% annual rise, Arleta Rewers, MD, PhD, of Children’s Hospital Colorado, Denver, reported at the annual meeting of the European Association for the Study of Diabetes.

Meanwhile, in another analysis that included multiple U.S. centers and about 7,600 cases of youth-onset type 1 diabetes, the overall prevalence of DKA at diagnosis was 38.5% between 2010 and 2016. However, the prevalence had increased from 35% in 2010 to 40.6% in 2016, according to Elizabeth T. Jensen, MPH, PhD, of Wake Forest University, Winston-Salem, N.C. The annual increase in prevalence of DKA at diagnosis of type 1 disease was 2%, adjusted for sociodemographic factors.
 

Rising prevalence

“DKA occurs most commonly at the time of type 1 diabetes diagnosis,” observed Dr. Jensen, who noted that “in the United States, among children, it’s younger children, uninsured or underinsured children, and children from minority racial or ethnic groups, who are at greatest risk.”

Dr. Elizabeth T. Jensen, Wake Forest University, Winston-Salem, N.C.
Sara Freeman/MDedge News
Dr. Elizabeth T. Jensen

Dr. Jensen and colleagues had previously shown that the prevalence of DKA at diagnosis was around 30% between 2002 and 2010, with no significant change in its prevalence. However, more recent reports from referral-based, single-center studies had suggested there was an increase, and that led her and her colleagues to take a closer look at the data.

To characterize the risk factors for DKA and the prevalence of DKA over time, Dr. Jensen and her team used the SEARCH for Diabetes in Youth database, which, she said, was “uniquely suited” for this purpose. SEARCH is a population-based, multicenter study conducted in centers in five U.S. states: South Carolina, Ohio, Colorado, California, and Washington.

A diagnosis of DKA was based on blood bicarbonate levels of less than 15 mmol/L, a venous pH of less than 7.25 or arterial or capillary pH of less than 7.3, or if there was any documentation of a DKA diagnosis.

As expected, the prevalence of DKA was highest in the youngest age group (0-4 years), Dr. Jensen said, but the increase in prevalence in that group was no different from the increases seen over time in the other age groups (5-9 years, 10-14 years, and 15 years or older).

There were no differences in the prevalence of DKA between the sexes, although there was a general increase over time. Similar trends were seen in DKA prevalence by race or ethnicity and by season, or time of year.

Of note, higher rates of DKA were seen in children who were covered by public health insurance, than in those covered by private insurance, although there was no difference in the rate of increase in DKA prevalence between the two groups. Dr. Jensen noted that only 64% of this study population had private insurance.

She said that future research in this area would need to look at the economic drivers and the “changing landscape of health insurance coverage in the United States.”
 

 

 

Expansion in health coverage

In presenting the findings of a study showing an increase in the prevalence of DKA at diagnosis of type 1 diabetes in children in Colorado from 2010 to 2017, Dr. Rewers said that the increase “paradoxically occurred” at a time of increasing health insurance coverage, a reference to the expansion of Medicaid during 2008-2012 and implementation in 2013 of the Affordable Care Act.

“Our group in Colorado has followed the frequency of DKA for almost 2 decades,” Dr. Rewers said. It’s important to study DKA as it is linked to worse glycemic control – with children with DKA having an HbA1c level of around 1% higher than those without DKA – and the potential for future, long-term complications.

Dr. Rewers noted that the increase in DKA at diagnosis of type 1 diabetes was more rapid in the children who had private rather than public health insurance. Of 1,187 patients with DKA, 57% had private health insurance, and 37% had public insurance, compared with 66% and 28%, respectively, in those without DKA. In 2010, the prevalence of DKA at diagnosis was 35.3% in those who were privately insured and 52.2% of those with public health insurance, but by 2017, a similar percentage of DKA was seen in the privately and publicly insured children (59.6% and 58.5%, respectively).

She said one possible explanation for that might be that “increased enrollment in high-deductible insurance plans could discourage families with private insurance from seeking timely care.”

Another explanation is that there is a low awareness of type 1 diabetes in the general population, she added. “Educational campaigns and autoimmunity screening have been shown to reduce DKA at diabetes diagnosis, but unfortunately they are not used widely at this point.”
 

Identifying at-risk children

“Diabetic ketoacidosis is a serious complication of diabetes [and] is difficult to diagnose because of the variability of the symptoms, said Angela Ibald-Mulli, PhD, who presented the findings of a retrospective cohort study in which she and her colleagues used a “discovery algorithm” called Q-Finder to identify the predictive factors for DKA in youth with type 1 diabetes, based on data from the Diabetes Prospective Follow-up Registry (DPV).

Dr. Angela Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi in Paris
Sara Freeman/MDedge News
Dr. Angela Ibald-Mulli

“The better we know the risk factors, the better we can care for our patients,” she emphasized.

The investigators obtained data on 108,223 patients with a diagnosis of type 1 disease and with more than two visits related to diabetes. The prevalence of DKA – defined as a pH of less than 7.3 during hospitalization occurring at least 10 days after the onset of type 1 diabetes – was 5.2%, said Dr. Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi, Paris.

A total of 129 different features were considered for their association with DKA – including comorbidities, sociodemographic factors, laboratory values, and concomitant medications – and were then used to identify, test, and the validate likely risk profiles.

After comparing the characteristics of patients with and without DKA, eight significant factors, all of which have been reported previously in the DPV cohort, were seen: younger age, lower body weight, higher HbA1c, younger age at onset of T1D; shorter disease duration; having a migration background; being less active; and having had more medical visits.

The investigators used the algorithm, and found 11 distinct profiles associated with DKA: an HbA1c higher than 8.87%; being aged 6-10 years; being aged 11-15 years; a diagnosis of nephropathy; DKA being present at onset; a prevalence of hypoglycemia with coma; a diagnosis of thyroiditis; a standardized body mass index lower than 16.9; not using short-acting insulin; younger than age 15 years; and not using continuous glucose monitoring.

Almost two-thirds of patients (64.7%) belonged to at least one of these risk profiles, Dr. Ibald-Mulli observed, with 7.1% of them having DKA, compared with 1.6% who belonged to none of the profiles.

Dr. Ibald-Mulli said it was important to note that the DKA risk profiles could overlap. “The more profiles a patient belongs to, the higher is the risk of having DKA,” she emphasized, adding that most patients (88.8%) with DKA belonged to just one profile, and fewer than 5% belonged to three or more profiles.

“Overall, the results of the algorithm confirmed known risk-factor profiles that had been previously identified by conventional statistical methods,” she concluded. It also provided “additional insights that can be further explored.”

SEARCH is funded by the Centers for Disease and Prevention and the National Institute of Diabetes and Digestive and Kidney Diseases. The DPV Registry is funded by multiple sponsors, including the European Federation for the Study of Diabetes and other academic institutions with the support of several commercial partners. Sanofi sponsored the study presented by Dr. Ibald-Mulli. Dr. Rewers made no disclosures, and Dr. Jensen did not have any conflicts of interest to declare. Dr. Ibald-Mulli is an employee of Sanofi.

 

SOURCE: Rewers A et al. EASD 2019, Abstract 115; Jensen E et al. EASD 2019, Abstract 116; Ibald-Mulli A et al. EASD 2019, Abstract 117.

– As many as 40%-60% of children have diabetic ketoacidosis (DKA) at the time of being diagnosed with type 1 diabetes, according to data from two U.S. analyses – and the figures have been rising for the past 10 years.

Between 2010 and 2017, the prevalence of DKA at diagnosis in children who were followed up at the Barbara Davies Cancer Center in Denver (n = 2,429) went from 41% to 59%, with a 7% annual rise, Arleta Rewers, MD, PhD, of Children’s Hospital Colorado, Denver, reported at the annual meeting of the European Association for the Study of Diabetes.

Meanwhile, in another analysis that included multiple U.S. centers and about 7,600 cases of youth-onset type 1 diabetes, the overall prevalence of DKA at diagnosis was 38.5% between 2010 and 2016. However, the prevalence had increased from 35% in 2010 to 40.6% in 2016, according to Elizabeth T. Jensen, MPH, PhD, of Wake Forest University, Winston-Salem, N.C. The annual increase in prevalence of DKA at diagnosis of type 1 disease was 2%, adjusted for sociodemographic factors.
 

Rising prevalence

“DKA occurs most commonly at the time of type 1 diabetes diagnosis,” observed Dr. Jensen, who noted that “in the United States, among children, it’s younger children, uninsured or underinsured children, and children from minority racial or ethnic groups, who are at greatest risk.”

Dr. Elizabeth T. Jensen, Wake Forest University, Winston-Salem, N.C.
Sara Freeman/MDedge News
Dr. Elizabeth T. Jensen

Dr. Jensen and colleagues had previously shown that the prevalence of DKA at diagnosis was around 30% between 2002 and 2010, with no significant change in its prevalence. However, more recent reports from referral-based, single-center studies had suggested there was an increase, and that led her and her colleagues to take a closer look at the data.

To characterize the risk factors for DKA and the prevalence of DKA over time, Dr. Jensen and her team used the SEARCH for Diabetes in Youth database, which, she said, was “uniquely suited” for this purpose. SEARCH is a population-based, multicenter study conducted in centers in five U.S. states: South Carolina, Ohio, Colorado, California, and Washington.

A diagnosis of DKA was based on blood bicarbonate levels of less than 15 mmol/L, a venous pH of less than 7.25 or arterial or capillary pH of less than 7.3, or if there was any documentation of a DKA diagnosis.

As expected, the prevalence of DKA was highest in the youngest age group (0-4 years), Dr. Jensen said, but the increase in prevalence in that group was no different from the increases seen over time in the other age groups (5-9 years, 10-14 years, and 15 years or older).

There were no differences in the prevalence of DKA between the sexes, although there was a general increase over time. Similar trends were seen in DKA prevalence by race or ethnicity and by season, or time of year.

Of note, higher rates of DKA were seen in children who were covered by public health insurance, than in those covered by private insurance, although there was no difference in the rate of increase in DKA prevalence between the two groups. Dr. Jensen noted that only 64% of this study population had private insurance.

She said that future research in this area would need to look at the economic drivers and the “changing landscape of health insurance coverage in the United States.”
 

 

 

Expansion in health coverage

In presenting the findings of a study showing an increase in the prevalence of DKA at diagnosis of type 1 diabetes in children in Colorado from 2010 to 2017, Dr. Rewers said that the increase “paradoxically occurred” at a time of increasing health insurance coverage, a reference to the expansion of Medicaid during 2008-2012 and implementation in 2013 of the Affordable Care Act.

“Our group in Colorado has followed the frequency of DKA for almost 2 decades,” Dr. Rewers said. It’s important to study DKA as it is linked to worse glycemic control – with children with DKA having an HbA1c level of around 1% higher than those without DKA – and the potential for future, long-term complications.

Dr. Rewers noted that the increase in DKA at diagnosis of type 1 diabetes was more rapid in the children who had private rather than public health insurance. Of 1,187 patients with DKA, 57% had private health insurance, and 37% had public insurance, compared with 66% and 28%, respectively, in those without DKA. In 2010, the prevalence of DKA at diagnosis was 35.3% in those who were privately insured and 52.2% of those with public health insurance, but by 2017, a similar percentage of DKA was seen in the privately and publicly insured children (59.6% and 58.5%, respectively).

She said one possible explanation for that might be that “increased enrollment in high-deductible insurance plans could discourage families with private insurance from seeking timely care.”

Another explanation is that there is a low awareness of type 1 diabetes in the general population, she added. “Educational campaigns and autoimmunity screening have been shown to reduce DKA at diabetes diagnosis, but unfortunately they are not used widely at this point.”
 

Identifying at-risk children

“Diabetic ketoacidosis is a serious complication of diabetes [and] is difficult to diagnose because of the variability of the symptoms, said Angela Ibald-Mulli, PhD, who presented the findings of a retrospective cohort study in which she and her colleagues used a “discovery algorithm” called Q-Finder to identify the predictive factors for DKA in youth with type 1 diabetes, based on data from the Diabetes Prospective Follow-up Registry (DPV).

Dr. Angela Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi in Paris
Sara Freeman/MDedge News
Dr. Angela Ibald-Mulli

“The better we know the risk factors, the better we can care for our patients,” she emphasized.

The investigators obtained data on 108,223 patients with a diagnosis of type 1 disease and with more than two visits related to diabetes. The prevalence of DKA – defined as a pH of less than 7.3 during hospitalization occurring at least 10 days after the onset of type 1 diabetes – was 5.2%, said Dr. Ibald-Mulli, head of Medical Evidence Generation Primary Care at Sanofi, Paris.

A total of 129 different features were considered for their association with DKA – including comorbidities, sociodemographic factors, laboratory values, and concomitant medications – and were then used to identify, test, and the validate likely risk profiles.

After comparing the characteristics of patients with and without DKA, eight significant factors, all of which have been reported previously in the DPV cohort, were seen: younger age, lower body weight, higher HbA1c, younger age at onset of T1D; shorter disease duration; having a migration background; being less active; and having had more medical visits.

The investigators used the algorithm, and found 11 distinct profiles associated with DKA: an HbA1c higher than 8.87%; being aged 6-10 years; being aged 11-15 years; a diagnosis of nephropathy; DKA being present at onset; a prevalence of hypoglycemia with coma; a diagnosis of thyroiditis; a standardized body mass index lower than 16.9; not using short-acting insulin; younger than age 15 years; and not using continuous glucose monitoring.

Almost two-thirds of patients (64.7%) belonged to at least one of these risk profiles, Dr. Ibald-Mulli observed, with 7.1% of them having DKA, compared with 1.6% who belonged to none of the profiles.

Dr. Ibald-Mulli said it was important to note that the DKA risk profiles could overlap. “The more profiles a patient belongs to, the higher is the risk of having DKA,” she emphasized, adding that most patients (88.8%) with DKA belonged to just one profile, and fewer than 5% belonged to three or more profiles.

“Overall, the results of the algorithm confirmed known risk-factor profiles that had been previously identified by conventional statistical methods,” she concluded. It also provided “additional insights that can be further explored.”

SEARCH is funded by the Centers for Disease and Prevention and the National Institute of Diabetes and Digestive and Kidney Diseases. The DPV Registry is funded by multiple sponsors, including the European Federation for the Study of Diabetes and other academic institutions with the support of several commercial partners. Sanofi sponsored the study presented by Dr. Ibald-Mulli. Dr. Rewers made no disclosures, and Dr. Jensen did not have any conflicts of interest to declare. Dr. Ibald-Mulli is an employee of Sanofi.

 

SOURCE: Rewers A et al. EASD 2019, Abstract 115; Jensen E et al. EASD 2019, Abstract 116; Ibald-Mulli A et al. EASD 2019, Abstract 117.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Signs of adult diabetes apparent in very young children

Article Type
Changed
Tue, 05/03/2022 - 15:12

 

– Disturbed HDL cholesterol metabolism is one of the earliest features that may predispose individuals to the development of type 2 diabetes, according to data from a genetics and metabolomics study conducted in the United Kingdom.

Dr. Joshua Bell esearch fellow at the University of Bristol
Sara Freeman/MDedge News
Dr. Joshua Bell

Changes in HDL cholesterol metabolism were seen in children as young as 8 years, decades before the clinical onset of disease, Joshua Bell, PhD, a research fellow at the University of Bristol (England), reported at the annual meeting of the European Association for the Study of Diabetes.

“We know that type 2 diabetes certainly doesn’t develop overnight,” Dr. Bell said. Indeed, data exist showing that there are changes in glucose metabolism several years before a formal diagnosis may be made in adults. “What we don’t know is what the very earliest features of diabetes look like,” he added.

“The main assumption is that type 2 diabetes is a metabolic disease, and so disease features are visible in systemic metabolism,” explained Dr. Bell. What was not clear, however, was that if any metabolic features – seen mainly in observational studies and in adults – were caused by the disease itself or perhaps were independent causes of type 2 diabetes.To investigate, Dr. Bell and associates performed a study linking genetic liability with metabolomic data collected at four time points from 4,761 offspring from participants in the Avon Longitudinal Study of Parents and Children cohort, which is also known as the Children of the 90s cohort. More than 200 metabolic traits were considered, and a genetic risk score comprising more than 162 single nucleotide polymorphisms previously linked to adult type 2 diabetes was used.

The metabolomic traits considered included lipoprotein subclass-specific cholesterol and triglyceride content, amino and fatty acids, and inflammatory glycoprotein acetyls, which had been measured in childhood at the age of 8 years, in adolescence at 16 years, in young adulthood at 18 years, and in adulthood at 25 years.

Early metabolic features of type 2 diabetes liability were grouped together and one feature that stood out was the sizes of lipid particles. In particular, it was the size of HDL cholesterol particle subtypes in children at the age of 8 years. Before other types of changes in lipid particles were being seen, there were reductions in the lipid content of HDL cholesterol particle subtypes, notably those that were very large.

By age 16 years, strong associations remained with lower lipids in HDL cholesterol particle subtypes and type 2 diabetes liability, which became stronger with preglycemic traits, such as citrate, and with glycoprotein acetyls. By age 18 years, elevations were seen in branched amino acids, and by age 25, association had strengthened for the lipid content of very low–density lipoprotein cholesterol.

“Linking genetic liability to adult disease with traits measured much earlier in life can tell you something about how the disease activity unfolds over a lifetime,” Dr. Bell said, adding that the feature that was “most consistently tracked” could be evaluated and could help reveal whether or not an individual might go on to develop type 2 diabetes.

In a press release issued by the EASD, Dr. Bell observed: “It’s remarkable that we can see signs of adult diabetes in the blood from such a young age. Knowing what early features of type 2 diabetes look like, could help us to intervene much earlier to halt progression to full-blown diabetes and its complications.”

The study was funded by Diabetes U.K., Cancer Research U.K., the Elizabeth Blackwell Institute for Health Research, the Wellcome Trust, the Medical Research Council, and the University of Bristol. Dr. Bell said he had no conflicts of interest to declare.

SOURCE: Bell J et al. bioRxiv. 2019 Sep 17. doi: 10.1101/767756.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Disturbed HDL cholesterol metabolism is one of the earliest features that may predispose individuals to the development of type 2 diabetes, according to data from a genetics and metabolomics study conducted in the United Kingdom.

Dr. Joshua Bell esearch fellow at the University of Bristol
Sara Freeman/MDedge News
Dr. Joshua Bell

Changes in HDL cholesterol metabolism were seen in children as young as 8 years, decades before the clinical onset of disease, Joshua Bell, PhD, a research fellow at the University of Bristol (England), reported at the annual meeting of the European Association for the Study of Diabetes.

“We know that type 2 diabetes certainly doesn’t develop overnight,” Dr. Bell said. Indeed, data exist showing that there are changes in glucose metabolism several years before a formal diagnosis may be made in adults. “What we don’t know is what the very earliest features of diabetes look like,” he added.

“The main assumption is that type 2 diabetes is a metabolic disease, and so disease features are visible in systemic metabolism,” explained Dr. Bell. What was not clear, however, was that if any metabolic features – seen mainly in observational studies and in adults – were caused by the disease itself or perhaps were independent causes of type 2 diabetes.To investigate, Dr. Bell and associates performed a study linking genetic liability with metabolomic data collected at four time points from 4,761 offspring from participants in the Avon Longitudinal Study of Parents and Children cohort, which is also known as the Children of the 90s cohort. More than 200 metabolic traits were considered, and a genetic risk score comprising more than 162 single nucleotide polymorphisms previously linked to adult type 2 diabetes was used.

The metabolomic traits considered included lipoprotein subclass-specific cholesterol and triglyceride content, amino and fatty acids, and inflammatory glycoprotein acetyls, which had been measured in childhood at the age of 8 years, in adolescence at 16 years, in young adulthood at 18 years, and in adulthood at 25 years.

Early metabolic features of type 2 diabetes liability were grouped together and one feature that stood out was the sizes of lipid particles. In particular, it was the size of HDL cholesterol particle subtypes in children at the age of 8 years. Before other types of changes in lipid particles were being seen, there were reductions in the lipid content of HDL cholesterol particle subtypes, notably those that were very large.

By age 16 years, strong associations remained with lower lipids in HDL cholesterol particle subtypes and type 2 diabetes liability, which became stronger with preglycemic traits, such as citrate, and with glycoprotein acetyls. By age 18 years, elevations were seen in branched amino acids, and by age 25, association had strengthened for the lipid content of very low–density lipoprotein cholesterol.

“Linking genetic liability to adult disease with traits measured much earlier in life can tell you something about how the disease activity unfolds over a lifetime,” Dr. Bell said, adding that the feature that was “most consistently tracked” could be evaluated and could help reveal whether or not an individual might go on to develop type 2 diabetes.

In a press release issued by the EASD, Dr. Bell observed: “It’s remarkable that we can see signs of adult diabetes in the blood from such a young age. Knowing what early features of type 2 diabetes look like, could help us to intervene much earlier to halt progression to full-blown diabetes and its complications.”

The study was funded by Diabetes U.K., Cancer Research U.K., the Elizabeth Blackwell Institute for Health Research, the Wellcome Trust, the Medical Research Council, and the University of Bristol. Dr. Bell said he had no conflicts of interest to declare.

SOURCE: Bell J et al. bioRxiv. 2019 Sep 17. doi: 10.1101/767756.
 

 

– Disturbed HDL cholesterol metabolism is one of the earliest features that may predispose individuals to the development of type 2 diabetes, according to data from a genetics and metabolomics study conducted in the United Kingdom.

Dr. Joshua Bell esearch fellow at the University of Bristol
Sara Freeman/MDedge News
Dr. Joshua Bell

Changes in HDL cholesterol metabolism were seen in children as young as 8 years, decades before the clinical onset of disease, Joshua Bell, PhD, a research fellow at the University of Bristol (England), reported at the annual meeting of the European Association for the Study of Diabetes.

“We know that type 2 diabetes certainly doesn’t develop overnight,” Dr. Bell said. Indeed, data exist showing that there are changes in glucose metabolism several years before a formal diagnosis may be made in adults. “What we don’t know is what the very earliest features of diabetes look like,” he added.

“The main assumption is that type 2 diabetes is a metabolic disease, and so disease features are visible in systemic metabolism,” explained Dr. Bell. What was not clear, however, was that if any metabolic features – seen mainly in observational studies and in adults – were caused by the disease itself or perhaps were independent causes of type 2 diabetes.To investigate, Dr. Bell and associates performed a study linking genetic liability with metabolomic data collected at four time points from 4,761 offspring from participants in the Avon Longitudinal Study of Parents and Children cohort, which is also known as the Children of the 90s cohort. More than 200 metabolic traits were considered, and a genetic risk score comprising more than 162 single nucleotide polymorphisms previously linked to adult type 2 diabetes was used.

The metabolomic traits considered included lipoprotein subclass-specific cholesterol and triglyceride content, amino and fatty acids, and inflammatory glycoprotein acetyls, which had been measured in childhood at the age of 8 years, in adolescence at 16 years, in young adulthood at 18 years, and in adulthood at 25 years.

Early metabolic features of type 2 diabetes liability were grouped together and one feature that stood out was the sizes of lipid particles. In particular, it was the size of HDL cholesterol particle subtypes in children at the age of 8 years. Before other types of changes in lipid particles were being seen, there were reductions in the lipid content of HDL cholesterol particle subtypes, notably those that were very large.

By age 16 years, strong associations remained with lower lipids in HDL cholesterol particle subtypes and type 2 diabetes liability, which became stronger with preglycemic traits, such as citrate, and with glycoprotein acetyls. By age 18 years, elevations were seen in branched amino acids, and by age 25, association had strengthened for the lipid content of very low–density lipoprotein cholesterol.

“Linking genetic liability to adult disease with traits measured much earlier in life can tell you something about how the disease activity unfolds over a lifetime,” Dr. Bell said, adding that the feature that was “most consistently tracked” could be evaluated and could help reveal whether or not an individual might go on to develop type 2 diabetes.

In a press release issued by the EASD, Dr. Bell observed: “It’s remarkable that we can see signs of adult diabetes in the blood from such a young age. Knowing what early features of type 2 diabetes look like, could help us to intervene much earlier to halt progression to full-blown diabetes and its complications.”

The study was funded by Diabetes U.K., Cancer Research U.K., the Elizabeth Blackwell Institute for Health Research, the Wellcome Trust, the Medical Research Council, and the University of Bristol. Dr. Bell said he had no conflicts of interest to declare.

SOURCE: Bell J et al. bioRxiv. 2019 Sep 17. doi: 10.1101/767756.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Body weight influences SGLT2-inhibitor effects in type 1 diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:12

– Individuals with type 1 diabetes and a high body mass index gain the most benefit with the least risk when sodium-glucose cotransporter 2 (SGLT2) inhibitors are added to insulin therapy, according to data presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Thomas Dahne, Auf der Bult Hospital for Children and Adolescents, Hannover Medical School, Germany
Sara Freeman/MDedge News
Dr. Thomas Danne

Results from new analyses of the inTandem 1 and inTandem 2 trials with sotagliflozin (Zynquista), and the DEPICT-1 and DEPICT-2 trials with dapagliflozin (Farxiga), support the recent decision of the European Medicines Agency to license the use of the drugs only in patients with a BMI of 27 kg/m2 or higher.
 

inTandem with sotagliflozin

Weight gain is a challenge in patients with type 1 diabetes, said Thomas Danne, MD, who presented post hoc data from the two inTandem studies. “It’s a little bit counterintuitive,” he acknowledged, “but you have to realize, particularly in patients who have hypoglycemia, that they have to take in extra carbohydrates,” which may tip them to becoming overweight or obese.

SGLT2-inhibitor therapy with sotagliflozin or dapagliflozin added to insulin therapy has been shown to reduce body weight in individuals with type 1 diabetes, but there is an increased risk for diabetic ketoacidosis (DKA). That risk, however, seems to be lower in the higher body-weight categories.

Dr. Danne, director of the department of general pediatrics, endocrinology, and diabetology, and clinical research at the Auf der Bult Hospital for Children and Adolescents, at the Hannover (Germany) Medical School, presented data looking at the outcomes of patients treated with sotagliflozin or placebo based on their BMI.

In all, 1,575 patients were included in the analysis, of whom 659 were of normal weight (BMI of less than 27 kg/m2; average mean, 24 kg/m2 at baseline), and 916 had a higher weight (BMI of 27 kg/m2 or higher; average mean, 32 kg/m2 at baseline). The mean age of patients at study entry was 42 years for those with the lower BMI, and 45 years for those with the higher BMI.

Patients in the two inTandem trials had been treated with insulin plus placebo (n = 228, BMI less than 27 kg/m2; n = 298, BMI 27 kg/m2 or higher), or insulin plus sotagliflozin at a dose of 200 mg (n = 219, BMI less than 27 kg/m2; n = 305, BMI 27 kg/m2 or higher) or 400 mg (n = 212; BMI less than 27 kg/m2; n = 313, BMI 27 kg/m2 or higher).
 

Glycemic control and body weight

Greater reductions in glycated hemoglobin (HbA1c) were seen with sotagliflozin versus placebo, and even more so, if the BMI was 27 kg/m2 or higher. At week 24, the least-squares mean difference in HbA1c comparing sotagliflozin 200 mg and placebo was –0.32 in patients with the lower BMI, compared with –0.39 in those with the higher BMI. Corresponding values for the 400-mg sotagliflozin group in the higher-BMI group were –0.27 and –0.45, respectively (P less than .001 for all comparisons).

 

 

In the lower-BMI group, week 24 least-squares mean differences in body weight comparing sotagliflozin and placebo were –2.06 kg for the 200-mg group and –2.55 kg for the 400-mg group, and –2.27 kg and 3.32 kg in the higher-BMI group (P less than .001 for all comparisons).

“This is why this class of drugs holds so much of a promise, [because] it’s not only one good effect regarding improvement of glycemia judged by A1c,” Dr. Danne said.

He also reported that treatment with sotagliflozin was associated with an increased time in range, compared with placebo, again, with greater effects seen in the higher- versus lower-BMI groups. In those with a BMI of 27 kg/m2 or more, there was an additional 1 hour 58 minutes time in range for the 200-mg dose, and 3 hours 37 minutes for the 200-mg dose, compared with an extra 24 minutes and 1 hour 31 minutes, respectively, in the lower-BMI category.

“We also see a trend to improved reduction in systolic blood pressure in those with the higher BMI,” Dr. Danne said.
 

Risk for DKA

“The big charm of these drugs is that not only do you improve A1c and all the other good things, but also you do this without increasing the risk of hypoglycemia,” said Dr. Danne. “Again, you can see a trend of a lower risk of severe hypoglycemia for both sotagliflozin doses [compared with placebo] in the group with the body mass index of greater than 27 kg/m2 [versus BMI of less than 27 kg/m2].”

The risk of DKA was higher than placebo in both BMI groups, but the number of DKA events was very small when comparing the low and high body weight categories (0 and 1 events, respectively, in the placebo groups; 7 and 9, in the sotagliflozin 200-mg group; and 9 and 11, in the 400-mg group. The absolute risk difference in the exposure adjusted incidence rate was slightly lower in the lower-BMI group, he said, but the numbers were so small that it is difficult to draw conclusions from that finding.

“There is no doubt that we have an increase for the risk of DKA with this class of drugs in general ... but it is futile to discuss whether or not, just on the basis of a body mass index or something else, we will be able to reduce it in a big fashion,” Dr. Danne suggested.
 

Body weight and composition

Other data on the long-term effect of sotagliflozin on body weight and composition were presented by Sangeeta Sawhney, MD, vice-president of clinical development at Lexicon Pharmaceuticals, Chapel Hill, North Carolina.

She presented data from the DEXA substudy of the inTandem phase 3 studies in which 243 patients underwent fat mass and bone density scanning.

SGLT2 inhibitors are associated with weight loss through glycuresis and net caloric loss, Dr. Sawhney reminded the audience. As sotagliflozin is a dual inhibitor of SGLT1 and SGLT2, however, it is important to estimate the contribution of changes in fat mass and lean mass to the weight loss that could be achieved with the drug.

Pooled data from the inTandem 1 and inTandem 2 studies showed that at week 24, there were reductions in body weight of –1.7 kg and –2.6 kg with sotagliflozin 200 mg and 400 mg, respectively, and at 52 weeks, reductions of –1.9 kg and –2.9 kg. However, there was an increase in body weight with placebo (+0.5 and +0.8 kg, respectively).

For the substudy, patients underwent dual-energy x-ray absorptiometry at baseline and weeks 24 and 52. Fat mass was measured at all three time points, and bone density was evaluated at the start and end of the study.

The least-square mean change in total fat mass from baseline to week 24 and week 52 were +0.6 and +0.1 kg, respectively, for placebo, –1.6 and –1.6 kg for the sotagliflozin 200-mg dose; and –1.9 and –2.1 kg for the 400-mg dose, “which really parallels the reduction in total body weight,” Dr. Sawhney observed.

The changes in total lean mass were much smaller for sotagliflozin, she added, at –0.6 kg at week 24 and 0.3 kg at week 52 for the 200-mg dose, and –0.7 kg and –0.4 kg, respectively, for the 400-mg dose, and rises in lean mass of 0.2 kg and 0.4 kg, respectively, in placebo.

Taken together, these data show that “about 80% of the body weight reduction is really from the fat mass, and a much smaller proportion of the total body weight reduction is really coming from the lean fat mass,” said Dr. Sawhney.
 

 

 

DEPICT with dapagliflozin

In a poster, Paresh Dandona, MD, PhD, of the State University of New York at Buffalo, and associates presented data from a pooled analysis of the DEPICT-1 and DEPICT-2 studies looking at safety and efficacy outcomes with dapagliflozin according to five BMI categories: less than or equal to 23 kg/m2; greater than 23 kg/m2 to less than or equal to 25 kg/m2; greater than 25 kg/m2 to less than or equal to 27 kg/m2; greater than 27 kg/m2 to less than or equal to 30 kg/m2; and greater than 30 kg/m2.

The pooled analysis included 548 patients treated with dapagliflozin 5 mg and 532 who received placebo. The investigators found that patients with higher BMIs who were treated with dapagliflozin had greater weight loss, showed a trend toward achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) or more without the risk of severe hypoglycemia, and had fewer episodes of definite DKA, compared with those with those with lower BMIs.

The adjusted mean percentage change from baseline in body weight in the lowest BMI (less than or equal to 23 kg/m2) group at week 24 was +0.06 kg for placebo and –2.71 kg for dapagliflozin, and at week 52, +0.33 kg and –2.91 kg, respectively. Corresponding values comparing placebo and dapagliflozin at 24 and 52 weeks in the highest BMI group (greater than 30 kg/m2) were –0.30 kg and –3.03 kg, and +0.56 and –3.58 kg.

Odds ratios for achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) without severe hypoglycemia at week 24 with dapagliflozin, compared with placebo, were, in increasing order of BMI groups: 1.85, 1.93, 3.87, 2.91, and 4.20.

“Generally, more events of definite DKA were observed in patients treated with dapagliflozin than in those treated with placebo,” but there were fewer events as BMI increased, Dr. Dandona and associates reported. “These data should be interpreted with caution due to the low number of events in each subgroup,” they added.

The number of adjudicated DKA events comparing dapagliflozin and placebo across the BMI groups were: 4 versus 1 (BMI less than or equal to 23 kg/m2); 6 versus 1 (BMI greater than 23 kg/m2 to less than or equal to 25 kg/m2); 7 versus 1 (BMI greater than 25 kg/m2 to less than or equal to 27 kg/m2); 3 versus 1 (BMI greater than 27 kg/m2 to less than or equal to 30 kg/m2); and 2 versus 1 (BMI greater than 30 kg/m2).

In regard to limitations, “this was a post hoc analysis,” the investigators noted, adding that the studies were not originally powered for comparison between BMI subgroups, so the results should be considered exploratory. Moreover, DKA and hypoglycemia were strictly monitored in the trials, which “may differ from real-world situations,” they said.

The inTandem studies were sponsored by Lexicon and Sanofi. Dr. Danne disclosed receiving research funding and serving as a consultant, advisory board or steering committee member, or speaker for various companies, including Sanofi. Dr. Sawhney is an employee of and holds stoke in Lexicon. The DEPICT studies were sponsored by AstraZeneca. The lead author, Dr. Dandona, disclosed employment or consultancy services for multiple companies, including AstraZeneca.

SOURCE: Danne T et al. EASD 2018, Oral Presentation 2; Dandona P et al. EASD 2019, ePoster 720; Sawhney S et al. EASD 2019, Oral Presentation 3.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– Individuals with type 1 diabetes and a high body mass index gain the most benefit with the least risk when sodium-glucose cotransporter 2 (SGLT2) inhibitors are added to insulin therapy, according to data presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Thomas Dahne, Auf der Bult Hospital for Children and Adolescents, Hannover Medical School, Germany
Sara Freeman/MDedge News
Dr. Thomas Danne

Results from new analyses of the inTandem 1 and inTandem 2 trials with sotagliflozin (Zynquista), and the DEPICT-1 and DEPICT-2 trials with dapagliflozin (Farxiga), support the recent decision of the European Medicines Agency to license the use of the drugs only in patients with a BMI of 27 kg/m2 or higher.
 

inTandem with sotagliflozin

Weight gain is a challenge in patients with type 1 diabetes, said Thomas Danne, MD, who presented post hoc data from the two inTandem studies. “It’s a little bit counterintuitive,” he acknowledged, “but you have to realize, particularly in patients who have hypoglycemia, that they have to take in extra carbohydrates,” which may tip them to becoming overweight or obese.

SGLT2-inhibitor therapy with sotagliflozin or dapagliflozin added to insulin therapy has been shown to reduce body weight in individuals with type 1 diabetes, but there is an increased risk for diabetic ketoacidosis (DKA). That risk, however, seems to be lower in the higher body-weight categories.

Dr. Danne, director of the department of general pediatrics, endocrinology, and diabetology, and clinical research at the Auf der Bult Hospital for Children and Adolescents, at the Hannover (Germany) Medical School, presented data looking at the outcomes of patients treated with sotagliflozin or placebo based on their BMI.

In all, 1,575 patients were included in the analysis, of whom 659 were of normal weight (BMI of less than 27 kg/m2; average mean, 24 kg/m2 at baseline), and 916 had a higher weight (BMI of 27 kg/m2 or higher; average mean, 32 kg/m2 at baseline). The mean age of patients at study entry was 42 years for those with the lower BMI, and 45 years for those with the higher BMI.

Patients in the two inTandem trials had been treated with insulin plus placebo (n = 228, BMI less than 27 kg/m2; n = 298, BMI 27 kg/m2 or higher), or insulin plus sotagliflozin at a dose of 200 mg (n = 219, BMI less than 27 kg/m2; n = 305, BMI 27 kg/m2 or higher) or 400 mg (n = 212; BMI less than 27 kg/m2; n = 313, BMI 27 kg/m2 or higher).
 

Glycemic control and body weight

Greater reductions in glycated hemoglobin (HbA1c) were seen with sotagliflozin versus placebo, and even more so, if the BMI was 27 kg/m2 or higher. At week 24, the least-squares mean difference in HbA1c comparing sotagliflozin 200 mg and placebo was –0.32 in patients with the lower BMI, compared with –0.39 in those with the higher BMI. Corresponding values for the 400-mg sotagliflozin group in the higher-BMI group were –0.27 and –0.45, respectively (P less than .001 for all comparisons).

 

 

In the lower-BMI group, week 24 least-squares mean differences in body weight comparing sotagliflozin and placebo were –2.06 kg for the 200-mg group and –2.55 kg for the 400-mg group, and –2.27 kg and 3.32 kg in the higher-BMI group (P less than .001 for all comparisons).

“This is why this class of drugs holds so much of a promise, [because] it’s not only one good effect regarding improvement of glycemia judged by A1c,” Dr. Danne said.

He also reported that treatment with sotagliflozin was associated with an increased time in range, compared with placebo, again, with greater effects seen in the higher- versus lower-BMI groups. In those with a BMI of 27 kg/m2 or more, there was an additional 1 hour 58 minutes time in range for the 200-mg dose, and 3 hours 37 minutes for the 200-mg dose, compared with an extra 24 minutes and 1 hour 31 minutes, respectively, in the lower-BMI category.

“We also see a trend to improved reduction in systolic blood pressure in those with the higher BMI,” Dr. Danne said.
 

Risk for DKA

“The big charm of these drugs is that not only do you improve A1c and all the other good things, but also you do this without increasing the risk of hypoglycemia,” said Dr. Danne. “Again, you can see a trend of a lower risk of severe hypoglycemia for both sotagliflozin doses [compared with placebo] in the group with the body mass index of greater than 27 kg/m2 [versus BMI of less than 27 kg/m2].”

The risk of DKA was higher than placebo in both BMI groups, but the number of DKA events was very small when comparing the low and high body weight categories (0 and 1 events, respectively, in the placebo groups; 7 and 9, in the sotagliflozin 200-mg group; and 9 and 11, in the 400-mg group. The absolute risk difference in the exposure adjusted incidence rate was slightly lower in the lower-BMI group, he said, but the numbers were so small that it is difficult to draw conclusions from that finding.

“There is no doubt that we have an increase for the risk of DKA with this class of drugs in general ... but it is futile to discuss whether or not, just on the basis of a body mass index or something else, we will be able to reduce it in a big fashion,” Dr. Danne suggested.
 

Body weight and composition

Other data on the long-term effect of sotagliflozin on body weight and composition were presented by Sangeeta Sawhney, MD, vice-president of clinical development at Lexicon Pharmaceuticals, Chapel Hill, North Carolina.

She presented data from the DEXA substudy of the inTandem phase 3 studies in which 243 patients underwent fat mass and bone density scanning.

SGLT2 inhibitors are associated with weight loss through glycuresis and net caloric loss, Dr. Sawhney reminded the audience. As sotagliflozin is a dual inhibitor of SGLT1 and SGLT2, however, it is important to estimate the contribution of changes in fat mass and lean mass to the weight loss that could be achieved with the drug.

Pooled data from the inTandem 1 and inTandem 2 studies showed that at week 24, there were reductions in body weight of –1.7 kg and –2.6 kg with sotagliflozin 200 mg and 400 mg, respectively, and at 52 weeks, reductions of –1.9 kg and –2.9 kg. However, there was an increase in body weight with placebo (+0.5 and +0.8 kg, respectively).

For the substudy, patients underwent dual-energy x-ray absorptiometry at baseline and weeks 24 and 52. Fat mass was measured at all three time points, and bone density was evaluated at the start and end of the study.

The least-square mean change in total fat mass from baseline to week 24 and week 52 were +0.6 and +0.1 kg, respectively, for placebo, –1.6 and –1.6 kg for the sotagliflozin 200-mg dose; and –1.9 and –2.1 kg for the 400-mg dose, “which really parallels the reduction in total body weight,” Dr. Sawhney observed.

The changes in total lean mass were much smaller for sotagliflozin, she added, at –0.6 kg at week 24 and 0.3 kg at week 52 for the 200-mg dose, and –0.7 kg and –0.4 kg, respectively, for the 400-mg dose, and rises in lean mass of 0.2 kg and 0.4 kg, respectively, in placebo.

Taken together, these data show that “about 80% of the body weight reduction is really from the fat mass, and a much smaller proportion of the total body weight reduction is really coming from the lean fat mass,” said Dr. Sawhney.
 

 

 

DEPICT with dapagliflozin

In a poster, Paresh Dandona, MD, PhD, of the State University of New York at Buffalo, and associates presented data from a pooled analysis of the DEPICT-1 and DEPICT-2 studies looking at safety and efficacy outcomes with dapagliflozin according to five BMI categories: less than or equal to 23 kg/m2; greater than 23 kg/m2 to less than or equal to 25 kg/m2; greater than 25 kg/m2 to less than or equal to 27 kg/m2; greater than 27 kg/m2 to less than or equal to 30 kg/m2; and greater than 30 kg/m2.

The pooled analysis included 548 patients treated with dapagliflozin 5 mg and 532 who received placebo. The investigators found that patients with higher BMIs who were treated with dapagliflozin had greater weight loss, showed a trend toward achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) or more without the risk of severe hypoglycemia, and had fewer episodes of definite DKA, compared with those with those with lower BMIs.

The adjusted mean percentage change from baseline in body weight in the lowest BMI (less than or equal to 23 kg/m2) group at week 24 was +0.06 kg for placebo and –2.71 kg for dapagliflozin, and at week 52, +0.33 kg and –2.91 kg, respectively. Corresponding values comparing placebo and dapagliflozin at 24 and 52 weeks in the highest BMI group (greater than 30 kg/m2) were –0.30 kg and –3.03 kg, and +0.56 and –3.58 kg.

Odds ratios for achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) without severe hypoglycemia at week 24 with dapagliflozin, compared with placebo, were, in increasing order of BMI groups: 1.85, 1.93, 3.87, 2.91, and 4.20.

“Generally, more events of definite DKA were observed in patients treated with dapagliflozin than in those treated with placebo,” but there were fewer events as BMI increased, Dr. Dandona and associates reported. “These data should be interpreted with caution due to the low number of events in each subgroup,” they added.

The number of adjudicated DKA events comparing dapagliflozin and placebo across the BMI groups were: 4 versus 1 (BMI less than or equal to 23 kg/m2); 6 versus 1 (BMI greater than 23 kg/m2 to less than or equal to 25 kg/m2); 7 versus 1 (BMI greater than 25 kg/m2 to less than or equal to 27 kg/m2); 3 versus 1 (BMI greater than 27 kg/m2 to less than or equal to 30 kg/m2); and 2 versus 1 (BMI greater than 30 kg/m2).

In regard to limitations, “this was a post hoc analysis,” the investigators noted, adding that the studies were not originally powered for comparison between BMI subgroups, so the results should be considered exploratory. Moreover, DKA and hypoglycemia were strictly monitored in the trials, which “may differ from real-world situations,” they said.

The inTandem studies were sponsored by Lexicon and Sanofi. Dr. Danne disclosed receiving research funding and serving as a consultant, advisory board or steering committee member, or speaker for various companies, including Sanofi. Dr. Sawhney is an employee of and holds stoke in Lexicon. The DEPICT studies were sponsored by AstraZeneca. The lead author, Dr. Dandona, disclosed employment or consultancy services for multiple companies, including AstraZeneca.

SOURCE: Danne T et al. EASD 2018, Oral Presentation 2; Dandona P et al. EASD 2019, ePoster 720; Sawhney S et al. EASD 2019, Oral Presentation 3.

– Individuals with type 1 diabetes and a high body mass index gain the most benefit with the least risk when sodium-glucose cotransporter 2 (SGLT2) inhibitors are added to insulin therapy, according to data presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Thomas Dahne, Auf der Bult Hospital for Children and Adolescents, Hannover Medical School, Germany
Sara Freeman/MDedge News
Dr. Thomas Danne

Results from new analyses of the inTandem 1 and inTandem 2 trials with sotagliflozin (Zynquista), and the DEPICT-1 and DEPICT-2 trials with dapagliflozin (Farxiga), support the recent decision of the European Medicines Agency to license the use of the drugs only in patients with a BMI of 27 kg/m2 or higher.
 

inTandem with sotagliflozin

Weight gain is a challenge in patients with type 1 diabetes, said Thomas Danne, MD, who presented post hoc data from the two inTandem studies. “It’s a little bit counterintuitive,” he acknowledged, “but you have to realize, particularly in patients who have hypoglycemia, that they have to take in extra carbohydrates,” which may tip them to becoming overweight or obese.

SGLT2-inhibitor therapy with sotagliflozin or dapagliflozin added to insulin therapy has been shown to reduce body weight in individuals with type 1 diabetes, but there is an increased risk for diabetic ketoacidosis (DKA). That risk, however, seems to be lower in the higher body-weight categories.

Dr. Danne, director of the department of general pediatrics, endocrinology, and diabetology, and clinical research at the Auf der Bult Hospital for Children and Adolescents, at the Hannover (Germany) Medical School, presented data looking at the outcomes of patients treated with sotagliflozin or placebo based on their BMI.

In all, 1,575 patients were included in the analysis, of whom 659 were of normal weight (BMI of less than 27 kg/m2; average mean, 24 kg/m2 at baseline), and 916 had a higher weight (BMI of 27 kg/m2 or higher; average mean, 32 kg/m2 at baseline). The mean age of patients at study entry was 42 years for those with the lower BMI, and 45 years for those with the higher BMI.

Patients in the two inTandem trials had been treated with insulin plus placebo (n = 228, BMI less than 27 kg/m2; n = 298, BMI 27 kg/m2 or higher), or insulin plus sotagliflozin at a dose of 200 mg (n = 219, BMI less than 27 kg/m2; n = 305, BMI 27 kg/m2 or higher) or 400 mg (n = 212; BMI less than 27 kg/m2; n = 313, BMI 27 kg/m2 or higher).
 

Glycemic control and body weight

Greater reductions in glycated hemoglobin (HbA1c) were seen with sotagliflozin versus placebo, and even more so, if the BMI was 27 kg/m2 or higher. At week 24, the least-squares mean difference in HbA1c comparing sotagliflozin 200 mg and placebo was –0.32 in patients with the lower BMI, compared with –0.39 in those with the higher BMI. Corresponding values for the 400-mg sotagliflozin group in the higher-BMI group were –0.27 and –0.45, respectively (P less than .001 for all comparisons).

 

 

In the lower-BMI group, week 24 least-squares mean differences in body weight comparing sotagliflozin and placebo were –2.06 kg for the 200-mg group and –2.55 kg for the 400-mg group, and –2.27 kg and 3.32 kg in the higher-BMI group (P less than .001 for all comparisons).

“This is why this class of drugs holds so much of a promise, [because] it’s not only one good effect regarding improvement of glycemia judged by A1c,” Dr. Danne said.

He also reported that treatment with sotagliflozin was associated with an increased time in range, compared with placebo, again, with greater effects seen in the higher- versus lower-BMI groups. In those with a BMI of 27 kg/m2 or more, there was an additional 1 hour 58 minutes time in range for the 200-mg dose, and 3 hours 37 minutes for the 200-mg dose, compared with an extra 24 minutes and 1 hour 31 minutes, respectively, in the lower-BMI category.

“We also see a trend to improved reduction in systolic blood pressure in those with the higher BMI,” Dr. Danne said.
 

Risk for DKA

“The big charm of these drugs is that not only do you improve A1c and all the other good things, but also you do this without increasing the risk of hypoglycemia,” said Dr. Danne. “Again, you can see a trend of a lower risk of severe hypoglycemia for both sotagliflozin doses [compared with placebo] in the group with the body mass index of greater than 27 kg/m2 [versus BMI of less than 27 kg/m2].”

The risk of DKA was higher than placebo in both BMI groups, but the number of DKA events was very small when comparing the low and high body weight categories (0 and 1 events, respectively, in the placebo groups; 7 and 9, in the sotagliflozin 200-mg group; and 9 and 11, in the 400-mg group. The absolute risk difference in the exposure adjusted incidence rate was slightly lower in the lower-BMI group, he said, but the numbers were so small that it is difficult to draw conclusions from that finding.

“There is no doubt that we have an increase for the risk of DKA with this class of drugs in general ... but it is futile to discuss whether or not, just on the basis of a body mass index or something else, we will be able to reduce it in a big fashion,” Dr. Danne suggested.
 

Body weight and composition

Other data on the long-term effect of sotagliflozin on body weight and composition were presented by Sangeeta Sawhney, MD, vice-president of clinical development at Lexicon Pharmaceuticals, Chapel Hill, North Carolina.

She presented data from the DEXA substudy of the inTandem phase 3 studies in which 243 patients underwent fat mass and bone density scanning.

SGLT2 inhibitors are associated with weight loss through glycuresis and net caloric loss, Dr. Sawhney reminded the audience. As sotagliflozin is a dual inhibitor of SGLT1 and SGLT2, however, it is important to estimate the contribution of changes in fat mass and lean mass to the weight loss that could be achieved with the drug.

Pooled data from the inTandem 1 and inTandem 2 studies showed that at week 24, there were reductions in body weight of –1.7 kg and –2.6 kg with sotagliflozin 200 mg and 400 mg, respectively, and at 52 weeks, reductions of –1.9 kg and –2.9 kg. However, there was an increase in body weight with placebo (+0.5 and +0.8 kg, respectively).

For the substudy, patients underwent dual-energy x-ray absorptiometry at baseline and weeks 24 and 52. Fat mass was measured at all three time points, and bone density was evaluated at the start and end of the study.

The least-square mean change in total fat mass from baseline to week 24 and week 52 were +0.6 and +0.1 kg, respectively, for placebo, –1.6 and –1.6 kg for the sotagliflozin 200-mg dose; and –1.9 and –2.1 kg for the 400-mg dose, “which really parallels the reduction in total body weight,” Dr. Sawhney observed.

The changes in total lean mass were much smaller for sotagliflozin, she added, at –0.6 kg at week 24 and 0.3 kg at week 52 for the 200-mg dose, and –0.7 kg and –0.4 kg, respectively, for the 400-mg dose, and rises in lean mass of 0.2 kg and 0.4 kg, respectively, in placebo.

Taken together, these data show that “about 80% of the body weight reduction is really from the fat mass, and a much smaller proportion of the total body weight reduction is really coming from the lean fat mass,” said Dr. Sawhney.
 

 

 

DEPICT with dapagliflozin

In a poster, Paresh Dandona, MD, PhD, of the State University of New York at Buffalo, and associates presented data from a pooled analysis of the DEPICT-1 and DEPICT-2 studies looking at safety and efficacy outcomes with dapagliflozin according to five BMI categories: less than or equal to 23 kg/m2; greater than 23 kg/m2 to less than or equal to 25 kg/m2; greater than 25 kg/m2 to less than or equal to 27 kg/m2; greater than 27 kg/m2 to less than or equal to 30 kg/m2; and greater than 30 kg/m2.

The pooled analysis included 548 patients treated with dapagliflozin 5 mg and 532 who received placebo. The investigators found that patients with higher BMIs who were treated with dapagliflozin had greater weight loss, showed a trend toward achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) or more without the risk of severe hypoglycemia, and had fewer episodes of definite DKA, compared with those with those with lower BMIs.

The adjusted mean percentage change from baseline in body weight in the lowest BMI (less than or equal to 23 kg/m2) group at week 24 was +0.06 kg for placebo and –2.71 kg for dapagliflozin, and at week 52, +0.33 kg and –2.91 kg, respectively. Corresponding values comparing placebo and dapagliflozin at 24 and 52 weeks in the highest BMI group (greater than 30 kg/m2) were –0.30 kg and –3.03 kg, and +0.56 and –3.58 kg.

Odds ratios for achieving an HbA1c reduction of 5.5 mmol/mol (greater than or equal to 0.5%) without severe hypoglycemia at week 24 with dapagliflozin, compared with placebo, were, in increasing order of BMI groups: 1.85, 1.93, 3.87, 2.91, and 4.20.

“Generally, more events of definite DKA were observed in patients treated with dapagliflozin than in those treated with placebo,” but there were fewer events as BMI increased, Dr. Dandona and associates reported. “These data should be interpreted with caution due to the low number of events in each subgroup,” they added.

The number of adjudicated DKA events comparing dapagliflozin and placebo across the BMI groups were: 4 versus 1 (BMI less than or equal to 23 kg/m2); 6 versus 1 (BMI greater than 23 kg/m2 to less than or equal to 25 kg/m2); 7 versus 1 (BMI greater than 25 kg/m2 to less than or equal to 27 kg/m2); 3 versus 1 (BMI greater than 27 kg/m2 to less than or equal to 30 kg/m2); and 2 versus 1 (BMI greater than 30 kg/m2).

In regard to limitations, “this was a post hoc analysis,” the investigators noted, adding that the studies were not originally powered for comparison between BMI subgroups, so the results should be considered exploratory. Moreover, DKA and hypoglycemia were strictly monitored in the trials, which “may differ from real-world situations,” they said.

The inTandem studies were sponsored by Lexicon and Sanofi. Dr. Danne disclosed receiving research funding and serving as a consultant, advisory board or steering committee member, or speaker for various companies, including Sanofi. Dr. Sawhney is an employee of and holds stoke in Lexicon. The DEPICT studies were sponsored by AstraZeneca. The lead author, Dr. Dandona, disclosed employment or consultancy services for multiple companies, including AstraZeneca.

SOURCE: Danne T et al. EASD 2018, Oral Presentation 2; Dandona P et al. EASD 2019, ePoster 720; Sawhney S et al. EASD 2019, Oral Presentation 3.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

GABA falls short for type 1 diabetes prevention in children

Article Type
Changed
Tue, 05/03/2022 - 15:12

– Gamma aminobutyric acid (GABA) alone or given in combination with glutamic acid decarboxylase (GAD) had little to no effect on the progression of type 1 diabetes in children, according to early data presented at the annual meeting of the European Association for the Study of Diabetes.

There was no difference between the two active treatment groups and placebo for the primary outcome measure, which was the effect on meal-stimulated C-peptide secretion before and after 1 year of treatment, study investigator Kenneth L. McCormick, MD, reported, nor was there any difference in glycemic control, based on hemoglobin A1c (HbA1c) and insulin dose, between the children who received GABA alone (n = 39) or combined with GAD (n = 22), and those who received placebo (n = 30).

“However, the GABA–GAD combination tended to have greater efficacy [than placebo] in terms of the daily insulin dose and the fasting C-peptide–to–glucagon ratio,” said Dr. McCormick, a pediatric endocrinologist at the University of Alabama at Birmingham.

Some beneficial effects on glucagon were seen with the GABA–GAD combination. “At 12 months, fasting glucagon was reduced [P less than .013] in the GABA–GAD group, compared with placebo,” he said. This was a “novel observation,” because stimulated glucagon was also reduced in this cohort. “This could be a potential salutatory metabolic effect in diabetes.”

The data were the first to be reported from the trial, and results of the immunologic analyses should be available by the end of the year and might reveal more positive effects of GABA and GAD, Dr. McCormick suggested. Data from a “proinsulin analysis” will also be available later.

The inspiration for the trial was a study performed in mice showing that GABA exerted a protective and regenerative effect on the islet beta cells and “reversed diabetes” (Proc Natl Acad Sci USA. 2011;108:11692-7). It took almost 4 years from the publication of that study to enroll the first patient for the current study.

“GABA was intriguing ... first of all, it is available in health food stores and in supermarkets in the United States,” said Dr. McCormick. “It has a strong safety profile, it’s tasteless, and can be given orally – what better could you ask for in a trial of children with type 1 diabetes?”

GABA is thought to have multiple effects in the pancreas, from increasing insulin secretion and suppressing glucagon secretion, to altering inflammation and T-cell populations. “That’s what’s so important to emphasize, besides its metabolic effects, this compound also has immunosuppressant action,” Dr. McCormick noted.

 

 


The study hypothesis was that treatment with oral GABA, or a combination of GABA–GAD, would hinder the progression of new-onset type 1 diabetes. The double-blind trial was designed to run for 1 year (Contemp Clin Trials. 2019;82:93-100) and recruited 97 children with newly diagnosed type 1 diabetes, aged 4-18 years, who were randomized to the three study groups. They were evaluated at baseline and months 1, 5, 8, and 12.

The trial had several limitations, however, which might explain the findings. A key limitation was that the researchers used a low dose of GABA – 1 to 1.5 g/m2 a day, given as a twice-daily oral dose, as mandated by the Food and Drug Administration. “For the GABA dose and the response, we are at the threshold. I don’t believe we are overdosing these kids,” Dr. McCormick said, noting that this is the first study done with GABA in humans.

In fact, GABA has a short half-life of around 2.5-5 hours, so the dose may need to be much higher to show an effect and perhaps administered three times a day, he said.

Another limitation was compliance with the twice-daily medication, especially because 35% of the patients were teenagers, and that it was a young population, with about a third of the patients aged younger that 8 years.

GABA and GABA–GAD should still be studied further, Dr. McCormick concluded, but “additional studies with a higher dose of GABA [given] three times a day, and not twice, are warranted.” Such studies also need to have more participants in each group.

The University of Alabama at Birmingham sponsored the study. Collaborators included Diamyd, NOW Foods, Janssen, and the Juvenile Diabetes Research Foundation. Dr. McCormick did not have any disclosures.

SOURCE: McCormick KL et al. EASD 2019, Abstract S05.1.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– Gamma aminobutyric acid (GABA) alone or given in combination with glutamic acid decarboxylase (GAD) had little to no effect on the progression of type 1 diabetes in children, according to early data presented at the annual meeting of the European Association for the Study of Diabetes.

There was no difference between the two active treatment groups and placebo for the primary outcome measure, which was the effect on meal-stimulated C-peptide secretion before and after 1 year of treatment, study investigator Kenneth L. McCormick, MD, reported, nor was there any difference in glycemic control, based on hemoglobin A1c (HbA1c) and insulin dose, between the children who received GABA alone (n = 39) or combined with GAD (n = 22), and those who received placebo (n = 30).

“However, the GABA–GAD combination tended to have greater efficacy [than placebo] in terms of the daily insulin dose and the fasting C-peptide–to–glucagon ratio,” said Dr. McCormick, a pediatric endocrinologist at the University of Alabama at Birmingham.

Some beneficial effects on glucagon were seen with the GABA–GAD combination. “At 12 months, fasting glucagon was reduced [P less than .013] in the GABA–GAD group, compared with placebo,” he said. This was a “novel observation,” because stimulated glucagon was also reduced in this cohort. “This could be a potential salutatory metabolic effect in diabetes.”

The data were the first to be reported from the trial, and results of the immunologic analyses should be available by the end of the year and might reveal more positive effects of GABA and GAD, Dr. McCormick suggested. Data from a “proinsulin analysis” will also be available later.

The inspiration for the trial was a study performed in mice showing that GABA exerted a protective and regenerative effect on the islet beta cells and “reversed diabetes” (Proc Natl Acad Sci USA. 2011;108:11692-7). It took almost 4 years from the publication of that study to enroll the first patient for the current study.

“GABA was intriguing ... first of all, it is available in health food stores and in supermarkets in the United States,” said Dr. McCormick. “It has a strong safety profile, it’s tasteless, and can be given orally – what better could you ask for in a trial of children with type 1 diabetes?”

GABA is thought to have multiple effects in the pancreas, from increasing insulin secretion and suppressing glucagon secretion, to altering inflammation and T-cell populations. “That’s what’s so important to emphasize, besides its metabolic effects, this compound also has immunosuppressant action,” Dr. McCormick noted.

 

 


The study hypothesis was that treatment with oral GABA, or a combination of GABA–GAD, would hinder the progression of new-onset type 1 diabetes. The double-blind trial was designed to run for 1 year (Contemp Clin Trials. 2019;82:93-100) and recruited 97 children with newly diagnosed type 1 diabetes, aged 4-18 years, who were randomized to the three study groups. They were evaluated at baseline and months 1, 5, 8, and 12.

The trial had several limitations, however, which might explain the findings. A key limitation was that the researchers used a low dose of GABA – 1 to 1.5 g/m2 a day, given as a twice-daily oral dose, as mandated by the Food and Drug Administration. “For the GABA dose and the response, we are at the threshold. I don’t believe we are overdosing these kids,” Dr. McCormick said, noting that this is the first study done with GABA in humans.

In fact, GABA has a short half-life of around 2.5-5 hours, so the dose may need to be much higher to show an effect and perhaps administered three times a day, he said.

Another limitation was compliance with the twice-daily medication, especially because 35% of the patients were teenagers, and that it was a young population, with about a third of the patients aged younger that 8 years.

GABA and GABA–GAD should still be studied further, Dr. McCormick concluded, but “additional studies with a higher dose of GABA [given] three times a day, and not twice, are warranted.” Such studies also need to have more participants in each group.

The University of Alabama at Birmingham sponsored the study. Collaborators included Diamyd, NOW Foods, Janssen, and the Juvenile Diabetes Research Foundation. Dr. McCormick did not have any disclosures.

SOURCE: McCormick KL et al. EASD 2019, Abstract S05.1.

– Gamma aminobutyric acid (GABA) alone or given in combination with glutamic acid decarboxylase (GAD) had little to no effect on the progression of type 1 diabetes in children, according to early data presented at the annual meeting of the European Association for the Study of Diabetes.

There was no difference between the two active treatment groups and placebo for the primary outcome measure, which was the effect on meal-stimulated C-peptide secretion before and after 1 year of treatment, study investigator Kenneth L. McCormick, MD, reported, nor was there any difference in glycemic control, based on hemoglobin A1c (HbA1c) and insulin dose, between the children who received GABA alone (n = 39) or combined with GAD (n = 22), and those who received placebo (n = 30).

“However, the GABA–GAD combination tended to have greater efficacy [than placebo] in terms of the daily insulin dose and the fasting C-peptide–to–glucagon ratio,” said Dr. McCormick, a pediatric endocrinologist at the University of Alabama at Birmingham.

Some beneficial effects on glucagon were seen with the GABA–GAD combination. “At 12 months, fasting glucagon was reduced [P less than .013] in the GABA–GAD group, compared with placebo,” he said. This was a “novel observation,” because stimulated glucagon was also reduced in this cohort. “This could be a potential salutatory metabolic effect in diabetes.”

The data were the first to be reported from the trial, and results of the immunologic analyses should be available by the end of the year and might reveal more positive effects of GABA and GAD, Dr. McCormick suggested. Data from a “proinsulin analysis” will also be available later.

The inspiration for the trial was a study performed in mice showing that GABA exerted a protective and regenerative effect on the islet beta cells and “reversed diabetes” (Proc Natl Acad Sci USA. 2011;108:11692-7). It took almost 4 years from the publication of that study to enroll the first patient for the current study.

“GABA was intriguing ... first of all, it is available in health food stores and in supermarkets in the United States,” said Dr. McCormick. “It has a strong safety profile, it’s tasteless, and can be given orally – what better could you ask for in a trial of children with type 1 diabetes?”

GABA is thought to have multiple effects in the pancreas, from increasing insulin secretion and suppressing glucagon secretion, to altering inflammation and T-cell populations. “That’s what’s so important to emphasize, besides its metabolic effects, this compound also has immunosuppressant action,” Dr. McCormick noted.

 

 


The study hypothesis was that treatment with oral GABA, or a combination of GABA–GAD, would hinder the progression of new-onset type 1 diabetes. The double-blind trial was designed to run for 1 year (Contemp Clin Trials. 2019;82:93-100) and recruited 97 children with newly diagnosed type 1 diabetes, aged 4-18 years, who were randomized to the three study groups. They were evaluated at baseline and months 1, 5, 8, and 12.

The trial had several limitations, however, which might explain the findings. A key limitation was that the researchers used a low dose of GABA – 1 to 1.5 g/m2 a day, given as a twice-daily oral dose, as mandated by the Food and Drug Administration. “For the GABA dose and the response, we are at the threshold. I don’t believe we are overdosing these kids,” Dr. McCormick said, noting that this is the first study done with GABA in humans.

In fact, GABA has a short half-life of around 2.5-5 hours, so the dose may need to be much higher to show an effect and perhaps administered three times a day, he said.

Another limitation was compliance with the twice-daily medication, especially because 35% of the patients were teenagers, and that it was a young population, with about a third of the patients aged younger that 8 years.

GABA and GABA–GAD should still be studied further, Dr. McCormick concluded, but “additional studies with a higher dose of GABA [given] three times a day, and not twice, are warranted.” Such studies also need to have more participants in each group.

The University of Alabama at Birmingham sponsored the study. Collaborators included Diamyd, NOW Foods, Janssen, and the Juvenile Diabetes Research Foundation. Dr. McCormick did not have any disclosures.

SOURCE: McCormick KL et al. EASD 2019, Abstract S05.1.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

REPORTING FROM EASD 2019

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.

SUSTAIN 10: Weight loss, glycemic control better with semaglutide than liraglutide

Article Type
Changed
Tue, 05/03/2022 - 15:12

 

– Patients with type 2 diabetes who were treated with semaglutide achieved greater reductions in glycated hemoglobin (HbA1c) levels and body weight, compared with those receiving liraglutide, according to results presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Matthew Capehorn
Sara Freeman/MDEdge News
Dr. Matthew Capehorn

In the phase 3b SUSTAIN 10 trial, conducted in 11 European countries, mean glycated hemoglobin at 30 weeks decreased by 1.7% with once-weekly semaglutide and 1.0% for once-daily liraglutide, from the overall baseline level of 8.2%. The estimated treatment difference (ETD) between the two treatments was –0.69 percentage points (95% confidence interval, –0.82 to –0.56; P less than .0001).

Mean body weight decreased during the same period by 5.8 kg with semaglutide and 1.9 kg with liraglutide, from a baseline of 96.9 kg. The ETD was 3.83 kg (95% CI, –4.57 to –3.09; P less than .0001).

The doses of semaglutide and liraglutide used in the study were 1.0 mg and 1.2 mg, respectively, the latter being the dose that is used most commonly in clinical practice, study investigator Matthew Capehorn, MB, CAB, explained in an interview at the meeting.

“We know that at a dose of 1.8 mg, liraglutide is more effective than 1.2 mg, but it’s about whether it is deemed more cost effective,” said Dr. Capehorn, who is clinical manager at Rotherham (England) Institute for Obesity, Clifton Medical Centre. “Certainly, in the United Kingdom, we’re encouraged to use the 1.2-mg dose” according to guidance from the National Institute for Heath and Care Excellence, and “other European countries are the same.”

Dr. Capehorn noted that studies are being done with a higher dose of semaglutide to see if it has potential as a weight loss drug in its own right in patients who do not have type 2 diabetes. “I care as much about obesity and cardiovascular disease as I do about chasing the HbA1c level and getting that reduced, so I would rather pick an agent that covers all three [components], than just looking at the HbA1c,” he said.

In SUSTAIN 10,577 adults with type 2 diabetes and an HbA1c level of between 7.0% and 11.0% who were on stable doses of one to three oral antidiabetic drugs were randomized to receive semaglutide (n = 290) or liraglutide (n = 287) for 30 weeks.

The primary endpoint was the change in HbA1c from baseline to week 30, and the secondary confirmatory endpoint was change in body weight over the same period.

In presenting the findings, which were simultaneously published in Diabetes & Metabolism, Dr. Capehorn noted that the efficacy results were consistent with those of other SUSTAIN trials that compared semaglutide with other glucagonlike peptide–1 receptor antagonists, notably SUSTAIN 3 (with exenatide extended release) and SUSTAIN 7 (with dulaglutide).

Other efficacy findings from SUSTAIN 10 were that semaglutide produced greater mean changes than did liraglutide in both fasting plasma glucose and in a 7-point, self-monitoring of blood glucose profile.

A greater percentage of people treated with semaglutide, compared with liraglutide, also achieved their glycemic targets of less than 7.0% (80% vs. 46%, respectively) and of 6.5% or less (58% vs. 25%), and their weight loss targets of 5% or more (56% vs. 18%) and 10% or more (19% vs. 4%).

In addition, more semaglutide- than liraglutide-treated patients achieved an HbA1c target of less than 7.0% without severe or blood glucose–confirmed symptomatic hypoglycemia, with or without weight gain (76% vs. 37%; P less than .0001). There were also more semaglutide patients who achieved an HbA1c reduction of 1% or more and a weight loss reduction of 10% or more (17% vs. 4% for liraglutide, P less than .0001).

The safety profiles were similar for semaglutide and liraglutide, Dr. Capehorn noted, but gastrointestinal adverse events were more prevalent in patients receiving semaglutide, compared with liraglutide (43.9% vs. 38.3%), and more patients receiving semaglutide discontinued treatment prematurely because of those adverse events (11.4% vs. 6.6% for liraglutide).

Novo Nordisk sponsored the study. Dr. Capehorn reported receiving research funding from, providing advisory board support to, and speaker fees from Novo Nordisk and from several other companies.
 

SOURCE: Capehorn M et al. EASD 2019, Oral Presentation 53; Capehorn M et al. Diabetes Metab. 2019 Sep 17. doi: 10.1016/j.diabet.2019.101117.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Patients with type 2 diabetes who were treated with semaglutide achieved greater reductions in glycated hemoglobin (HbA1c) levels and body weight, compared with those receiving liraglutide, according to results presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Matthew Capehorn
Sara Freeman/MDEdge News
Dr. Matthew Capehorn

In the phase 3b SUSTAIN 10 trial, conducted in 11 European countries, mean glycated hemoglobin at 30 weeks decreased by 1.7% with once-weekly semaglutide and 1.0% for once-daily liraglutide, from the overall baseline level of 8.2%. The estimated treatment difference (ETD) between the two treatments was –0.69 percentage points (95% confidence interval, –0.82 to –0.56; P less than .0001).

Mean body weight decreased during the same period by 5.8 kg with semaglutide and 1.9 kg with liraglutide, from a baseline of 96.9 kg. The ETD was 3.83 kg (95% CI, –4.57 to –3.09; P less than .0001).

The doses of semaglutide and liraglutide used in the study were 1.0 mg and 1.2 mg, respectively, the latter being the dose that is used most commonly in clinical practice, study investigator Matthew Capehorn, MB, CAB, explained in an interview at the meeting.

“We know that at a dose of 1.8 mg, liraglutide is more effective than 1.2 mg, but it’s about whether it is deemed more cost effective,” said Dr. Capehorn, who is clinical manager at Rotherham (England) Institute for Obesity, Clifton Medical Centre. “Certainly, in the United Kingdom, we’re encouraged to use the 1.2-mg dose” according to guidance from the National Institute for Heath and Care Excellence, and “other European countries are the same.”

Dr. Capehorn noted that studies are being done with a higher dose of semaglutide to see if it has potential as a weight loss drug in its own right in patients who do not have type 2 diabetes. “I care as much about obesity and cardiovascular disease as I do about chasing the HbA1c level and getting that reduced, so I would rather pick an agent that covers all three [components], than just looking at the HbA1c,” he said.

In SUSTAIN 10,577 adults with type 2 diabetes and an HbA1c level of between 7.0% and 11.0% who were on stable doses of one to three oral antidiabetic drugs were randomized to receive semaglutide (n = 290) or liraglutide (n = 287) for 30 weeks.

The primary endpoint was the change in HbA1c from baseline to week 30, and the secondary confirmatory endpoint was change in body weight over the same period.

In presenting the findings, which were simultaneously published in Diabetes & Metabolism, Dr. Capehorn noted that the efficacy results were consistent with those of other SUSTAIN trials that compared semaglutide with other glucagonlike peptide–1 receptor antagonists, notably SUSTAIN 3 (with exenatide extended release) and SUSTAIN 7 (with dulaglutide).

Other efficacy findings from SUSTAIN 10 were that semaglutide produced greater mean changes than did liraglutide in both fasting plasma glucose and in a 7-point, self-monitoring of blood glucose profile.

A greater percentage of people treated with semaglutide, compared with liraglutide, also achieved their glycemic targets of less than 7.0% (80% vs. 46%, respectively) and of 6.5% or less (58% vs. 25%), and their weight loss targets of 5% or more (56% vs. 18%) and 10% or more (19% vs. 4%).

In addition, more semaglutide- than liraglutide-treated patients achieved an HbA1c target of less than 7.0% without severe or blood glucose–confirmed symptomatic hypoglycemia, with or without weight gain (76% vs. 37%; P less than .0001). There were also more semaglutide patients who achieved an HbA1c reduction of 1% or more and a weight loss reduction of 10% or more (17% vs. 4% for liraglutide, P less than .0001).

The safety profiles were similar for semaglutide and liraglutide, Dr. Capehorn noted, but gastrointestinal adverse events were more prevalent in patients receiving semaglutide, compared with liraglutide (43.9% vs. 38.3%), and more patients receiving semaglutide discontinued treatment prematurely because of those adverse events (11.4% vs. 6.6% for liraglutide).

Novo Nordisk sponsored the study. Dr. Capehorn reported receiving research funding from, providing advisory board support to, and speaker fees from Novo Nordisk and from several other companies.
 

SOURCE: Capehorn M et al. EASD 2019, Oral Presentation 53; Capehorn M et al. Diabetes Metab. 2019 Sep 17. doi: 10.1016/j.diabet.2019.101117.

 

– Patients with type 2 diabetes who were treated with semaglutide achieved greater reductions in glycated hemoglobin (HbA1c) levels and body weight, compared with those receiving liraglutide, according to results presented at the annual meeting of the European Association for the Study of Diabetes.

Dr. Matthew Capehorn
Sara Freeman/MDEdge News
Dr. Matthew Capehorn

In the phase 3b SUSTAIN 10 trial, conducted in 11 European countries, mean glycated hemoglobin at 30 weeks decreased by 1.7% with once-weekly semaglutide and 1.0% for once-daily liraglutide, from the overall baseline level of 8.2%. The estimated treatment difference (ETD) between the two treatments was –0.69 percentage points (95% confidence interval, –0.82 to –0.56; P less than .0001).

Mean body weight decreased during the same period by 5.8 kg with semaglutide and 1.9 kg with liraglutide, from a baseline of 96.9 kg. The ETD was 3.83 kg (95% CI, –4.57 to –3.09; P less than .0001).

The doses of semaglutide and liraglutide used in the study were 1.0 mg and 1.2 mg, respectively, the latter being the dose that is used most commonly in clinical practice, study investigator Matthew Capehorn, MB, CAB, explained in an interview at the meeting.

“We know that at a dose of 1.8 mg, liraglutide is more effective than 1.2 mg, but it’s about whether it is deemed more cost effective,” said Dr. Capehorn, who is clinical manager at Rotherham (England) Institute for Obesity, Clifton Medical Centre. “Certainly, in the United Kingdom, we’re encouraged to use the 1.2-mg dose” according to guidance from the National Institute for Heath and Care Excellence, and “other European countries are the same.”

Dr. Capehorn noted that studies are being done with a higher dose of semaglutide to see if it has potential as a weight loss drug in its own right in patients who do not have type 2 diabetes. “I care as much about obesity and cardiovascular disease as I do about chasing the HbA1c level and getting that reduced, so I would rather pick an agent that covers all three [components], than just looking at the HbA1c,” he said.

In SUSTAIN 10,577 adults with type 2 diabetes and an HbA1c level of between 7.0% and 11.0% who were on stable doses of one to three oral antidiabetic drugs were randomized to receive semaglutide (n = 290) or liraglutide (n = 287) for 30 weeks.

The primary endpoint was the change in HbA1c from baseline to week 30, and the secondary confirmatory endpoint was change in body weight over the same period.

In presenting the findings, which were simultaneously published in Diabetes & Metabolism, Dr. Capehorn noted that the efficacy results were consistent with those of other SUSTAIN trials that compared semaglutide with other glucagonlike peptide–1 receptor antagonists, notably SUSTAIN 3 (with exenatide extended release) and SUSTAIN 7 (with dulaglutide).

Other efficacy findings from SUSTAIN 10 were that semaglutide produced greater mean changes than did liraglutide in both fasting plasma glucose and in a 7-point, self-monitoring of blood glucose profile.

A greater percentage of people treated with semaglutide, compared with liraglutide, also achieved their glycemic targets of less than 7.0% (80% vs. 46%, respectively) and of 6.5% or less (58% vs. 25%), and their weight loss targets of 5% or more (56% vs. 18%) and 10% or more (19% vs. 4%).

In addition, more semaglutide- than liraglutide-treated patients achieved an HbA1c target of less than 7.0% without severe or blood glucose–confirmed symptomatic hypoglycemia, with or without weight gain (76% vs. 37%; P less than .0001). There were also more semaglutide patients who achieved an HbA1c reduction of 1% or more and a weight loss reduction of 10% or more (17% vs. 4% for liraglutide, P less than .0001).

The safety profiles were similar for semaglutide and liraglutide, Dr. Capehorn noted, but gastrointestinal adverse events were more prevalent in patients receiving semaglutide, compared with liraglutide (43.9% vs. 38.3%), and more patients receiving semaglutide discontinued treatment prematurely because of those adverse events (11.4% vs. 6.6% for liraglutide).

Novo Nordisk sponsored the study. Dr. Capehorn reported receiving research funding from, providing advisory board support to, and speaker fees from Novo Nordisk and from several other companies.
 

SOURCE: Capehorn M et al. EASD 2019, Oral Presentation 53; Capehorn M et al. Diabetes Metab. 2019 Sep 17. doi: 10.1016/j.diabet.2019.101117.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Obesity ups type 2 diabetes risk far more than lifestyle, genetics

Article Type
Changed
Tue, 05/03/2022 - 15:12

– Obesity, more so than having a poor lifestyle, significantly raised the odds of developing type 2 diabetes, independent of individuals’ genetic susceptibility, according to data from a Danish population-based, case-cohort study.

In fact, having a body mass index (BMI) of more than 30 kg/m2 was linked with a 480% risk of incident type 2 diabetes, compared with being of normal weight (BMI, 18.5-24.9 kg/m2). The 95% confidence interval was 5.16-6.55. Being overweight (BMI, 25-29.9 kg/m2) also carried a 100% increased risk of type 2 diabetes (hazard ratio, 2.37; 95% CI, 2.15-2.62).

Having an unfavorable lifestyle – which was defined as having no or only one of several healthy-living characteristics, from not smoking and moderating alcohol use to eating a well-balanced, nutritious diet and exercising regularly – increased the risk of diabetes by 18%, compared with having a favorable lifestyle (HR, 1.18; 95% CI, 1.06-1.30).

Individuals with a high genetic risk score (GRS) had a 100% increased risk of developing the disease versus those with a low GRS (HR, 2.0; 95% CI, 1.1-1.3).

“High genetic risk, obesity, and [an] unfavorable lifestyle increase the individual-level risk of incident type 2 diabetes,” Hermina Jakupovic and associates reported in a poster presentation at the annual meeting of the European Association for the Study of Diabetes. Their results suggest that “the effect of obesity on type 2 diabetes risk is dominant over other risk factors, highlighting the importance of weight management in type 2 diabetes prevention.”



Ms. Jakupovic, a PhD student at the Novo Nordisk Foundation Center for Basic Metabolic Research at the University of Copenhagen, and coauthors examined data on 9,555 participants of the Diet, Cancer, and Health cohort, a large, prospective study that has been running since the early 1990s.

Around half of the study sample were women and the mean age was 52 years. Just over one-fifth (22.8%) were obese, 43% were overweight, and the remaining 35.2% were of normal weight. A quarter (25.4%) had an unfavorable lifestyle, 40% a favorable lifestyle, and the remainder an “intermediate” lifestyle. Over a follow-up of almost 15 years, nearly half (49.5%) developed type 2 diabetes.

Genetic risk was assessed by a GRS comprising 193 genetic variants known to be strongly associated with type 2 diabetes, Ms. Jakupovic explained, adding that, using the GRS, patients were categorized into being at low (the lowest 20%), intermediate (middle 60%) and high risk (top 20%) of type 2 diabetes.

Considering individuals’ GRS and lifestyle score together showed an increasing risk of developing type 2 diabetes from the low GRS/favorable-lifestyle category (HR, 1.0; reference) upward to the high GRS/unfavorable lifestyle (HR, 2.22; 95% CI, 1.76-2.81).

The Diet, Cancer, and Health cohort is supported by the Danish Cancer Society. The Novo Nordisk Foundation Center for Basic Metabolic Research is an independent research center at the University of Copenhagen partially funded by an unrestricted donation from the Novo Nordisk Foundation. Ms. Jakupovic and associates are funded either directly or indirectly by the Novo Nordisk Foundation.

SOURCE: Jakupovic H et al. EASD 2019, Abstract 376.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– Obesity, more so than having a poor lifestyle, significantly raised the odds of developing type 2 diabetes, independent of individuals’ genetic susceptibility, according to data from a Danish population-based, case-cohort study.

In fact, having a body mass index (BMI) of more than 30 kg/m2 was linked with a 480% risk of incident type 2 diabetes, compared with being of normal weight (BMI, 18.5-24.9 kg/m2). The 95% confidence interval was 5.16-6.55. Being overweight (BMI, 25-29.9 kg/m2) also carried a 100% increased risk of type 2 diabetes (hazard ratio, 2.37; 95% CI, 2.15-2.62).

Having an unfavorable lifestyle – which was defined as having no or only one of several healthy-living characteristics, from not smoking and moderating alcohol use to eating a well-balanced, nutritious diet and exercising regularly – increased the risk of diabetes by 18%, compared with having a favorable lifestyle (HR, 1.18; 95% CI, 1.06-1.30).

Individuals with a high genetic risk score (GRS) had a 100% increased risk of developing the disease versus those with a low GRS (HR, 2.0; 95% CI, 1.1-1.3).

“High genetic risk, obesity, and [an] unfavorable lifestyle increase the individual-level risk of incident type 2 diabetes,” Hermina Jakupovic and associates reported in a poster presentation at the annual meeting of the European Association for the Study of Diabetes. Their results suggest that “the effect of obesity on type 2 diabetes risk is dominant over other risk factors, highlighting the importance of weight management in type 2 diabetes prevention.”



Ms. Jakupovic, a PhD student at the Novo Nordisk Foundation Center for Basic Metabolic Research at the University of Copenhagen, and coauthors examined data on 9,555 participants of the Diet, Cancer, and Health cohort, a large, prospective study that has been running since the early 1990s.

Around half of the study sample were women and the mean age was 52 years. Just over one-fifth (22.8%) were obese, 43% were overweight, and the remaining 35.2% were of normal weight. A quarter (25.4%) had an unfavorable lifestyle, 40% a favorable lifestyle, and the remainder an “intermediate” lifestyle. Over a follow-up of almost 15 years, nearly half (49.5%) developed type 2 diabetes.

Genetic risk was assessed by a GRS comprising 193 genetic variants known to be strongly associated with type 2 diabetes, Ms. Jakupovic explained, adding that, using the GRS, patients were categorized into being at low (the lowest 20%), intermediate (middle 60%) and high risk (top 20%) of type 2 diabetes.

Considering individuals’ GRS and lifestyle score together showed an increasing risk of developing type 2 diabetes from the low GRS/favorable-lifestyle category (HR, 1.0; reference) upward to the high GRS/unfavorable lifestyle (HR, 2.22; 95% CI, 1.76-2.81).

The Diet, Cancer, and Health cohort is supported by the Danish Cancer Society. The Novo Nordisk Foundation Center for Basic Metabolic Research is an independent research center at the University of Copenhagen partially funded by an unrestricted donation from the Novo Nordisk Foundation. Ms. Jakupovic and associates are funded either directly or indirectly by the Novo Nordisk Foundation.

SOURCE: Jakupovic H et al. EASD 2019, Abstract 376.

– Obesity, more so than having a poor lifestyle, significantly raised the odds of developing type 2 diabetes, independent of individuals’ genetic susceptibility, according to data from a Danish population-based, case-cohort study.

In fact, having a body mass index (BMI) of more than 30 kg/m2 was linked with a 480% risk of incident type 2 diabetes, compared with being of normal weight (BMI, 18.5-24.9 kg/m2). The 95% confidence interval was 5.16-6.55. Being overweight (BMI, 25-29.9 kg/m2) also carried a 100% increased risk of type 2 diabetes (hazard ratio, 2.37; 95% CI, 2.15-2.62).

Having an unfavorable lifestyle – which was defined as having no or only one of several healthy-living characteristics, from not smoking and moderating alcohol use to eating a well-balanced, nutritious diet and exercising regularly – increased the risk of diabetes by 18%, compared with having a favorable lifestyle (HR, 1.18; 95% CI, 1.06-1.30).

Individuals with a high genetic risk score (GRS) had a 100% increased risk of developing the disease versus those with a low GRS (HR, 2.0; 95% CI, 1.1-1.3).

“High genetic risk, obesity, and [an] unfavorable lifestyle increase the individual-level risk of incident type 2 diabetes,” Hermina Jakupovic and associates reported in a poster presentation at the annual meeting of the European Association for the Study of Diabetes. Their results suggest that “the effect of obesity on type 2 diabetes risk is dominant over other risk factors, highlighting the importance of weight management in type 2 diabetes prevention.”



Ms. Jakupovic, a PhD student at the Novo Nordisk Foundation Center for Basic Metabolic Research at the University of Copenhagen, and coauthors examined data on 9,555 participants of the Diet, Cancer, and Health cohort, a large, prospective study that has been running since the early 1990s.

Around half of the study sample were women and the mean age was 52 years. Just over one-fifth (22.8%) were obese, 43% were overweight, and the remaining 35.2% were of normal weight. A quarter (25.4%) had an unfavorable lifestyle, 40% a favorable lifestyle, and the remainder an “intermediate” lifestyle. Over a follow-up of almost 15 years, nearly half (49.5%) developed type 2 diabetes.

Genetic risk was assessed by a GRS comprising 193 genetic variants known to be strongly associated with type 2 diabetes, Ms. Jakupovic explained, adding that, using the GRS, patients were categorized into being at low (the lowest 20%), intermediate (middle 60%) and high risk (top 20%) of type 2 diabetes.

Considering individuals’ GRS and lifestyle score together showed an increasing risk of developing type 2 diabetes from the low GRS/favorable-lifestyle category (HR, 1.0; reference) upward to the high GRS/unfavorable lifestyle (HR, 2.22; 95% CI, 1.76-2.81).

The Diet, Cancer, and Health cohort is supported by the Danish Cancer Society. The Novo Nordisk Foundation Center for Basic Metabolic Research is an independent research center at the University of Copenhagen partially funded by an unrestricted donation from the Novo Nordisk Foundation. Ms. Jakupovic and associates are funded either directly or indirectly by the Novo Nordisk Foundation.

SOURCE: Jakupovic H et al. EASD 2019, Abstract 376.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

Liraglutide ‘option’ for treating pediatric type 2 diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:12

 

– The glucagon-like peptide-1 receptor agonist (GLP-1 RA) liraglutide added onto metformin with or without basal insulin effectively reduced hemoglobin A1c and fasting plasma glucose levels in children with type 2 diabetes in the 52-week ELLIPSE study.

Dr. Timothy Barrett, University of Birmingham, England
Sara Freeman/MDedge News
Dr. Timothy Barrett

The primary endpoint of the trial, which was the mean change in HbA1c from baseline to 26 weeks, was met, with a greater percentage point decrease with liraglutide (Victoza) than placebo (–0.64 vs. +0.42), with an estimated treatment difference of –1.06 percentage points (P less than .001). At the end of the study, the percentage point changes were –0.50 and +0.80, with a between-group difference of –1.30 in favor of liraglutide.

“Those of us working in pediatric practice are seeing an increasing demand for our clinical services in children with type 2 diabetes,” study investigator Timothy Barrett, PhD, MBBS, observed at the annual meeting of the European Association for the Study of Diabetes. This reflects the increasing prevalence of type 2 diabetes in this age group and is most likely linked to the rising rates of obesity and overweight that have been reported widely in young people in recent years, he added.

“Unfortunately, we look with envy upon our adult physician colleagues, and the range of treatments they have available to treat type 2 diabetes in adults.” In pediatrics, the only licensed treatments that have been available until recently were metformin and insulin, with the latter being an “illogical treatment to treat those with obesity-related diabetes.” The study’s findings, however, support liraglutide as another option to consider, said Dr. Barrett, a pediatric endocrinologist and professor of pediatrics and child health based at the University of Birmingham, England.

“Liraglutide at doses of up to 1.8 mg/day when added to metformin, and basal insulin if required, does seem to offer an additional treatment option for children and young people with type 2 diabetes who require improved glycemic control after they’ve reached a maximum dose of metformin,” he said.

ELLIPSE (Evaluation of Liraglutide in Pediatrics with Diabetes) was a multicenter, randomized, parallel group, placebo-controlled trial to assess the efficacy and safety of liraglutide as an add-on treatment to metformin, with or without basal insulin, in 134 overweight or obese children and adolescents (aged 10-17) with type 2 diabetes.

For inclusion, patients had to be able to complete the trial before their 18th birthday, and have an HbA1c of at least 7% if being treated with diet and exercise, or 6.5% or higher if already being treated with metformin, with or without insulin. Body mass index had to be above the 85the percentile for their age and sex.



Of 307 children and adolescents screened at 84 centers in 25 countries, 135 were randomized and 134 were treated between 2012 and 2018. Screening took place over a period of 2 weeks, after which time those eligible for the trial underwent a 3- to 4-week period where their dose of metformin was titrated if needed followed by an 8-week maintenance period. Only after that was randomization to liraglutide or placebo done, with the GLP-1R started at a subcutaneous dose of 0.6 mg and titrated up to 1.2 or 1.8 mg over 3 days to achieve a fasting plasma glucose (FPG) of less than 6.1 mmol/L (110 mg/dL). However, not all patients were escalated to the top dose, Dr. Barrett noted.

The mean age of patients in the trial was 14.5 years; about 60% of patients were female. The duration of diabetes was about 1.9 years and the average body weight and BMI a respective 91 kg and 33 kg/m2.

Over the course of the study, FPG fell by 1.06 mmol/L at week 26 and 1.03 mmol/L at week 52 in the liraglutide group but rose in the placebo group by 0.80 and 0.78 mmol/L, respectively. The estimated treatment difference was –1.88 (P = .002) and –1.81 at 26 an 52 weeks, respectively.

What was “a really gratifying to see,” said Dr. Barrett, was that the proportion of children and young people achieving a glycemic target of an HbA1c of less than 7% by the end of the double-blind treatment period was significantly higher in the liraglutide than placebo group, at 63.7% and 36.5%, respectively.

Most of the adverse effects seen in the study were gastrointestinal symptoms, including nausea, vomiting, and diarrhea, in about 20% of liraglutide-treated patients, compared with roughly 10% of placebo-treated patients. “This is really reflected in the adult studies as well, and many of these were thankfully transient.”

As for hypoglycemia, Dr. Barrett reported that there was a higher rate in liraglutide- than placebo-treated patients (45.5% vs. 25% for any event), although there were no severe episodes in the liraglutide group and one in the placebo group. Almost a third (31%) of hypoglycemic episodes were asymptomatic, versus 17.6% for the placebo group.

“This is the first successfully completed phase 3 trial showing efficacy of a noninsulin agent, in this case, for children who do not get managed solely on metformin monotherapy,” Dr. Barrett said.

The Food and Drug Administration has approved liraglutide for use in pediatric patients 10 years or older with type 2 diabetes, based in part on results of the ELLIPSE results, Novo Nordisk announced in June. The trial results were published prior to the EASD meeting (Tamborlane WV et al. N Engl J Med. 2019 Aug 15;381:637-46).

Novo Nordisk initiated and funded the trial, and most of the investigators reported receiving funds from the company outside the submitted work. Dr Barrett disclosed being a consultant to and/or receiving honoraria from AstraZeneca, Novo Nordisk and Servier.

SOURCE: Barrett T et al. EASD 2019. Abstract 84.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– The glucagon-like peptide-1 receptor agonist (GLP-1 RA) liraglutide added onto metformin with or without basal insulin effectively reduced hemoglobin A1c and fasting plasma glucose levels in children with type 2 diabetes in the 52-week ELLIPSE study.

Dr. Timothy Barrett, University of Birmingham, England
Sara Freeman/MDedge News
Dr. Timothy Barrett

The primary endpoint of the trial, which was the mean change in HbA1c from baseline to 26 weeks, was met, with a greater percentage point decrease with liraglutide (Victoza) than placebo (–0.64 vs. +0.42), with an estimated treatment difference of –1.06 percentage points (P less than .001). At the end of the study, the percentage point changes were –0.50 and +0.80, with a between-group difference of –1.30 in favor of liraglutide.

“Those of us working in pediatric practice are seeing an increasing demand for our clinical services in children with type 2 diabetes,” study investigator Timothy Barrett, PhD, MBBS, observed at the annual meeting of the European Association for the Study of Diabetes. This reflects the increasing prevalence of type 2 diabetes in this age group and is most likely linked to the rising rates of obesity and overweight that have been reported widely in young people in recent years, he added.

“Unfortunately, we look with envy upon our adult physician colleagues, and the range of treatments they have available to treat type 2 diabetes in adults.” In pediatrics, the only licensed treatments that have been available until recently were metformin and insulin, with the latter being an “illogical treatment to treat those with obesity-related diabetes.” The study’s findings, however, support liraglutide as another option to consider, said Dr. Barrett, a pediatric endocrinologist and professor of pediatrics and child health based at the University of Birmingham, England.

“Liraglutide at doses of up to 1.8 mg/day when added to metformin, and basal insulin if required, does seem to offer an additional treatment option for children and young people with type 2 diabetes who require improved glycemic control after they’ve reached a maximum dose of metformin,” he said.

ELLIPSE (Evaluation of Liraglutide in Pediatrics with Diabetes) was a multicenter, randomized, parallel group, placebo-controlled trial to assess the efficacy and safety of liraglutide as an add-on treatment to metformin, with or without basal insulin, in 134 overweight or obese children and adolescents (aged 10-17) with type 2 diabetes.

For inclusion, patients had to be able to complete the trial before their 18th birthday, and have an HbA1c of at least 7% if being treated with diet and exercise, or 6.5% or higher if already being treated with metformin, with or without insulin. Body mass index had to be above the 85the percentile for their age and sex.



Of 307 children and adolescents screened at 84 centers in 25 countries, 135 were randomized and 134 were treated between 2012 and 2018. Screening took place over a period of 2 weeks, after which time those eligible for the trial underwent a 3- to 4-week period where their dose of metformin was titrated if needed followed by an 8-week maintenance period. Only after that was randomization to liraglutide or placebo done, with the GLP-1R started at a subcutaneous dose of 0.6 mg and titrated up to 1.2 or 1.8 mg over 3 days to achieve a fasting plasma glucose (FPG) of less than 6.1 mmol/L (110 mg/dL). However, not all patients were escalated to the top dose, Dr. Barrett noted.

The mean age of patients in the trial was 14.5 years; about 60% of patients were female. The duration of diabetes was about 1.9 years and the average body weight and BMI a respective 91 kg and 33 kg/m2.

Over the course of the study, FPG fell by 1.06 mmol/L at week 26 and 1.03 mmol/L at week 52 in the liraglutide group but rose in the placebo group by 0.80 and 0.78 mmol/L, respectively. The estimated treatment difference was –1.88 (P = .002) and –1.81 at 26 an 52 weeks, respectively.

What was “a really gratifying to see,” said Dr. Barrett, was that the proportion of children and young people achieving a glycemic target of an HbA1c of less than 7% by the end of the double-blind treatment period was significantly higher in the liraglutide than placebo group, at 63.7% and 36.5%, respectively.

Most of the adverse effects seen in the study were gastrointestinal symptoms, including nausea, vomiting, and diarrhea, in about 20% of liraglutide-treated patients, compared with roughly 10% of placebo-treated patients. “This is really reflected in the adult studies as well, and many of these were thankfully transient.”

As for hypoglycemia, Dr. Barrett reported that there was a higher rate in liraglutide- than placebo-treated patients (45.5% vs. 25% for any event), although there were no severe episodes in the liraglutide group and one in the placebo group. Almost a third (31%) of hypoglycemic episodes were asymptomatic, versus 17.6% for the placebo group.

“This is the first successfully completed phase 3 trial showing efficacy of a noninsulin agent, in this case, for children who do not get managed solely on metformin monotherapy,” Dr. Barrett said.

The Food and Drug Administration has approved liraglutide for use in pediatric patients 10 years or older with type 2 diabetes, based in part on results of the ELLIPSE results, Novo Nordisk announced in June. The trial results were published prior to the EASD meeting (Tamborlane WV et al. N Engl J Med. 2019 Aug 15;381:637-46).

Novo Nordisk initiated and funded the trial, and most of the investigators reported receiving funds from the company outside the submitted work. Dr Barrett disclosed being a consultant to and/or receiving honoraria from AstraZeneca, Novo Nordisk and Servier.

SOURCE: Barrett T et al. EASD 2019. Abstract 84.

 

– The glucagon-like peptide-1 receptor agonist (GLP-1 RA) liraglutide added onto metformin with or without basal insulin effectively reduced hemoglobin A1c and fasting plasma glucose levels in children with type 2 diabetes in the 52-week ELLIPSE study.

Dr. Timothy Barrett, University of Birmingham, England
Sara Freeman/MDedge News
Dr. Timothy Barrett

The primary endpoint of the trial, which was the mean change in HbA1c from baseline to 26 weeks, was met, with a greater percentage point decrease with liraglutide (Victoza) than placebo (–0.64 vs. +0.42), with an estimated treatment difference of –1.06 percentage points (P less than .001). At the end of the study, the percentage point changes were –0.50 and +0.80, with a between-group difference of –1.30 in favor of liraglutide.

“Those of us working in pediatric practice are seeing an increasing demand for our clinical services in children with type 2 diabetes,” study investigator Timothy Barrett, PhD, MBBS, observed at the annual meeting of the European Association for the Study of Diabetes. This reflects the increasing prevalence of type 2 diabetes in this age group and is most likely linked to the rising rates of obesity and overweight that have been reported widely in young people in recent years, he added.

“Unfortunately, we look with envy upon our adult physician colleagues, and the range of treatments they have available to treat type 2 diabetes in adults.” In pediatrics, the only licensed treatments that have been available until recently were metformin and insulin, with the latter being an “illogical treatment to treat those with obesity-related diabetes.” The study’s findings, however, support liraglutide as another option to consider, said Dr. Barrett, a pediatric endocrinologist and professor of pediatrics and child health based at the University of Birmingham, England.

“Liraglutide at doses of up to 1.8 mg/day when added to metformin, and basal insulin if required, does seem to offer an additional treatment option for children and young people with type 2 diabetes who require improved glycemic control after they’ve reached a maximum dose of metformin,” he said.

ELLIPSE (Evaluation of Liraglutide in Pediatrics with Diabetes) was a multicenter, randomized, parallel group, placebo-controlled trial to assess the efficacy and safety of liraglutide as an add-on treatment to metformin, with or without basal insulin, in 134 overweight or obese children and adolescents (aged 10-17) with type 2 diabetes.

For inclusion, patients had to be able to complete the trial before their 18th birthday, and have an HbA1c of at least 7% if being treated with diet and exercise, or 6.5% or higher if already being treated with metformin, with or without insulin. Body mass index had to be above the 85the percentile for their age and sex.



Of 307 children and adolescents screened at 84 centers in 25 countries, 135 were randomized and 134 were treated between 2012 and 2018. Screening took place over a period of 2 weeks, after which time those eligible for the trial underwent a 3- to 4-week period where their dose of metformin was titrated if needed followed by an 8-week maintenance period. Only after that was randomization to liraglutide or placebo done, with the GLP-1R started at a subcutaneous dose of 0.6 mg and titrated up to 1.2 or 1.8 mg over 3 days to achieve a fasting plasma glucose (FPG) of less than 6.1 mmol/L (110 mg/dL). However, not all patients were escalated to the top dose, Dr. Barrett noted.

The mean age of patients in the trial was 14.5 years; about 60% of patients were female. The duration of diabetes was about 1.9 years and the average body weight and BMI a respective 91 kg and 33 kg/m2.

Over the course of the study, FPG fell by 1.06 mmol/L at week 26 and 1.03 mmol/L at week 52 in the liraglutide group but rose in the placebo group by 0.80 and 0.78 mmol/L, respectively. The estimated treatment difference was –1.88 (P = .002) and –1.81 at 26 an 52 weeks, respectively.

What was “a really gratifying to see,” said Dr. Barrett, was that the proportion of children and young people achieving a glycemic target of an HbA1c of less than 7% by the end of the double-blind treatment period was significantly higher in the liraglutide than placebo group, at 63.7% and 36.5%, respectively.

Most of the adverse effects seen in the study were gastrointestinal symptoms, including nausea, vomiting, and diarrhea, in about 20% of liraglutide-treated patients, compared with roughly 10% of placebo-treated patients. “This is really reflected in the adult studies as well, and many of these were thankfully transient.”

As for hypoglycemia, Dr. Barrett reported that there was a higher rate in liraglutide- than placebo-treated patients (45.5% vs. 25% for any event), although there were no severe episodes in the liraglutide group and one in the placebo group. Almost a third (31%) of hypoglycemic episodes were asymptomatic, versus 17.6% for the placebo group.

“This is the first successfully completed phase 3 trial showing efficacy of a noninsulin agent, in this case, for children who do not get managed solely on metformin monotherapy,” Dr. Barrett said.

The Food and Drug Administration has approved liraglutide for use in pediatric patients 10 years or older with type 2 diabetes, based in part on results of the ELLIPSE results, Novo Nordisk announced in June. The trial results were published prior to the EASD meeting (Tamborlane WV et al. N Engl J Med. 2019 Aug 15;381:637-46).

Novo Nordisk initiated and funded the trial, and most of the investigators reported receiving funds from the company outside the submitted work. Dr Barrett disclosed being a consultant to and/or receiving honoraria from AstraZeneca, Novo Nordisk and Servier.

SOURCE: Barrett T et al. EASD 2019. Abstract 84.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.

COMISAIR: CGM ‘makes the difference’ in type 1 diabetes

Article Type
Changed
Tue, 05/03/2022 - 15:13

 

– Real-time continuous glucose monitoring (rtCGM) was better than self-monitoring of blood glucose (SMBG) in reducing hemoglobin A1c (HbA1c) and other glycemic endpoints in people with type 1 diabetes, regardless of the type of insulin delivery method used in a 3-year follow-up of a prospective, real-world clinical trial.

Long-term results from the COMISAIR study showed that the end-of-study HbA1c values were significantly lower, compared with baseline values, in people with type 1 diabetes who used rtCGM with multiple daily injections (MDI) of insulin (7.0% [53 mmol/mol], P = .0002) or an insulin pump (6.9% [52 mmol/mol], P less than .0001). There was no significant difference between the two rtCGM delivery-method groups.

Final HbA1c values for those who used SMBG with multiple daily injections or an insulin pump were 8.0% (64 mmol/mol) and 7.7% (61 mmol/mol), respectively, but were not significantly different from baseline (P = .3574 and P = .1, respectively).

These findings could help guide physicians when discussing treatment and monitoring options with their patients, suggested study investigator Jan Šoupal, MD, PhD, of Charles University in Prague, when he presented the findings at the annual meeting of the European Association for the Study of Diabetes.

Dr. Šoupal and associates have previously reported data from the COMISAIR (Comparison of Different Treatment Modalities for Type 1 Diabetes Including Sensor-Augmented Insulin Regimens) study at 1 year of follow-up for 65 patients (Diabetes Technol Ther. 2016;18:532-8). The findings he presented at the EASD meeting, simultaneously published online in Diabetes Care, were for the full cohort of 94 patients and, with 3 years of follow-up, makes it “the longest CGM trial ever,” he said.

At the time the COMISAIR study was initiated, in 2013, “we knew that insulin pump therapy, especially in combination with real-time CGM, can improve several outcomes of patients with type 1 diabetes,” Dr. Šoupal observed. However, the effectiveness of CGM in patients with MDI was not widely described, and comparisons between continuous subcutaneous insulin infusion (CSII) and insulin MDI with rtCGM were lacking. “Moreover, we didn’t have any comparison between insulin pump therapy alone, without CGM, and MDI with CGM, and there were no long-term trials with real-time [continuous glucose monitoring].”

The aim of the COMISAIR study was therefore to compare four different treatment strategies in people with type 1 diabetes who had an HbA1c of 7%-­­10% (53-86 mmol/mol), despite MDI treatment with insulin analogues and SMBG. The treatment strategies tested were CSII plus rtCGM (n = 26), MDI plus rtCGM (n = 22), CSII plus SMBG (n = 25), and MDI plus SMBG (n = 21). Patients were not randomized to these treatment arms but exposed to all of them during a 4-day DAFNE-like training session and then allowed to choose which they would like to use according to their individual needs and preferences, reflecting real-life practice.

Dr. Šoupal pointed out that two different continuous glucose monitoring devices had been used in the trial, and that 100% of the CGM groups wore a sensor for more than 70% of the time, which was one of the prerequisites for inclusion in the trial. Good adherence was observed, with 93% of patients completing all study visits, and CGM users wearing their sensors on average 88% of the time. “This nice adherence may be connected to the pretty good results,” he observed.

In discussing the HbA1c results, Dr. Šoupal noted that “improvement observed in patients with [continuous glucose monitoring] is stable throughout 3 years, which is not always a reality for different types of treatment for diabetes.” In addition, “it is not so important how insulin is delivered, what is more important is how patients with type 1 diabetes monitor their glucose.”

Another key endpoint of the trial was time in range (70-180 mg/dL [3.9-10 mmol/L]). Results showed significantly more patients achieving this with rtCGM than with SMBG, regardless of whether they were using pump therapy or MDI. Comparing 3-year with baseline values, time in range was 72.3% versus 50.9% for rtCGM with CSII and 69% versus 48.7% for rtCGM with MDI (P less than .0001 for both). Results with SMBG with CSII or MDI were a respective 57.8% versus 50.6% (P = .0114) and 54.7% versus 51.8% (P = 1.0).

Glycemic variability was reduced in patients using insulin pumps with SBMG, and “not surprisingly, there was a reduction in both CGM-augmented groups,” Dr. Šoupal stated.

There was a reduction in the time spent in hypoglycemia from baseline to year 3 in all four groups, but that was significant only for the two rtCGM groups. Overall, there were seven severe hypoglycemia episodes, five in the SMBG groups (two in the CSII group, three in the MDI group) and two in the rtCGM groups (one each in the CSII and MDI groups), with one episode only occurring when the CGM sensor was not being worn.

Three episodes of ketoacidosis were reported – one each in the SMBG-pump, SMBG-MDI, and rtCGM-pump groups.

In summing up, Dr. Šoupal said that “real-time CGM, both with insulin pumps and with [multiple daily injections], provided significant, comparable, and stable improvement of glycemic outcomes.” He added that “treatment with CGM and MDI was more effective than treatment with insulin pump therapy alone, and that CGM and MDI can even be considered as a suitable alternative to treatment with insulin pumps and CMG for some patients.”

With many treatment options available, some will suit patients better than others, he suggested, but although “individualization of our treatment is important”, the COMISAIR data show that “it is CGM that makes the difference”.

The study was supported by the Agency for Healthcare Research and the Ministry of Health of the Czech Republic. Dr. Šoupal reported receiving honoraria from Abbott, AstraZeneca, Boehringer Ingelheim, Dexcom, Eli Lilly, Medtronic, Novo Nordisk, and Roche. Dexcom also paid for the development of the manuscript published in Diabetes Care.
 

SOURCES: Šoupal J et al. EASD 2019, Abstract 40; Šoupal J et al. Diabetes Care. 2019 Sep 17. doi: 10.2337/dc19-0888.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Real-time continuous glucose monitoring (rtCGM) was better than self-monitoring of blood glucose (SMBG) in reducing hemoglobin A1c (HbA1c) and other glycemic endpoints in people with type 1 diabetes, regardless of the type of insulin delivery method used in a 3-year follow-up of a prospective, real-world clinical trial.

Long-term results from the COMISAIR study showed that the end-of-study HbA1c values were significantly lower, compared with baseline values, in people with type 1 diabetes who used rtCGM with multiple daily injections (MDI) of insulin (7.0% [53 mmol/mol], P = .0002) or an insulin pump (6.9% [52 mmol/mol], P less than .0001). There was no significant difference between the two rtCGM delivery-method groups.

Final HbA1c values for those who used SMBG with multiple daily injections or an insulin pump were 8.0% (64 mmol/mol) and 7.7% (61 mmol/mol), respectively, but were not significantly different from baseline (P = .3574 and P = .1, respectively).

These findings could help guide physicians when discussing treatment and monitoring options with their patients, suggested study investigator Jan Šoupal, MD, PhD, of Charles University in Prague, when he presented the findings at the annual meeting of the European Association for the Study of Diabetes.

Dr. Šoupal and associates have previously reported data from the COMISAIR (Comparison of Different Treatment Modalities for Type 1 Diabetes Including Sensor-Augmented Insulin Regimens) study at 1 year of follow-up for 65 patients (Diabetes Technol Ther. 2016;18:532-8). The findings he presented at the EASD meeting, simultaneously published online in Diabetes Care, were for the full cohort of 94 patients and, with 3 years of follow-up, makes it “the longest CGM trial ever,” he said.

At the time the COMISAIR study was initiated, in 2013, “we knew that insulin pump therapy, especially in combination with real-time CGM, can improve several outcomes of patients with type 1 diabetes,” Dr. Šoupal observed. However, the effectiveness of CGM in patients with MDI was not widely described, and comparisons between continuous subcutaneous insulin infusion (CSII) and insulin MDI with rtCGM were lacking. “Moreover, we didn’t have any comparison between insulin pump therapy alone, without CGM, and MDI with CGM, and there were no long-term trials with real-time [continuous glucose monitoring].”

The aim of the COMISAIR study was therefore to compare four different treatment strategies in people with type 1 diabetes who had an HbA1c of 7%-­­10% (53-86 mmol/mol), despite MDI treatment with insulin analogues and SMBG. The treatment strategies tested were CSII plus rtCGM (n = 26), MDI plus rtCGM (n = 22), CSII plus SMBG (n = 25), and MDI plus SMBG (n = 21). Patients were not randomized to these treatment arms but exposed to all of them during a 4-day DAFNE-like training session and then allowed to choose which they would like to use according to their individual needs and preferences, reflecting real-life practice.

Dr. Šoupal pointed out that two different continuous glucose monitoring devices had been used in the trial, and that 100% of the CGM groups wore a sensor for more than 70% of the time, which was one of the prerequisites for inclusion in the trial. Good adherence was observed, with 93% of patients completing all study visits, and CGM users wearing their sensors on average 88% of the time. “This nice adherence may be connected to the pretty good results,” he observed.

In discussing the HbA1c results, Dr. Šoupal noted that “improvement observed in patients with [continuous glucose monitoring] is stable throughout 3 years, which is not always a reality for different types of treatment for diabetes.” In addition, “it is not so important how insulin is delivered, what is more important is how patients with type 1 diabetes monitor their glucose.”

Another key endpoint of the trial was time in range (70-180 mg/dL [3.9-10 mmol/L]). Results showed significantly more patients achieving this with rtCGM than with SMBG, regardless of whether they were using pump therapy or MDI. Comparing 3-year with baseline values, time in range was 72.3% versus 50.9% for rtCGM with CSII and 69% versus 48.7% for rtCGM with MDI (P less than .0001 for both). Results with SMBG with CSII or MDI were a respective 57.8% versus 50.6% (P = .0114) and 54.7% versus 51.8% (P = 1.0).

Glycemic variability was reduced in patients using insulin pumps with SBMG, and “not surprisingly, there was a reduction in both CGM-augmented groups,” Dr. Šoupal stated.

There was a reduction in the time spent in hypoglycemia from baseline to year 3 in all four groups, but that was significant only for the two rtCGM groups. Overall, there were seven severe hypoglycemia episodes, five in the SMBG groups (two in the CSII group, three in the MDI group) and two in the rtCGM groups (one each in the CSII and MDI groups), with one episode only occurring when the CGM sensor was not being worn.

Three episodes of ketoacidosis were reported – one each in the SMBG-pump, SMBG-MDI, and rtCGM-pump groups.

In summing up, Dr. Šoupal said that “real-time CGM, both with insulin pumps and with [multiple daily injections], provided significant, comparable, and stable improvement of glycemic outcomes.” He added that “treatment with CGM and MDI was more effective than treatment with insulin pump therapy alone, and that CGM and MDI can even be considered as a suitable alternative to treatment with insulin pumps and CMG for some patients.”

With many treatment options available, some will suit patients better than others, he suggested, but although “individualization of our treatment is important”, the COMISAIR data show that “it is CGM that makes the difference”.

The study was supported by the Agency for Healthcare Research and the Ministry of Health of the Czech Republic. Dr. Šoupal reported receiving honoraria from Abbott, AstraZeneca, Boehringer Ingelheim, Dexcom, Eli Lilly, Medtronic, Novo Nordisk, and Roche. Dexcom also paid for the development of the manuscript published in Diabetes Care.
 

SOURCES: Šoupal J et al. EASD 2019, Abstract 40; Šoupal J et al. Diabetes Care. 2019 Sep 17. doi: 10.2337/dc19-0888.

 

– Real-time continuous glucose monitoring (rtCGM) was better than self-monitoring of blood glucose (SMBG) in reducing hemoglobin A1c (HbA1c) and other glycemic endpoints in people with type 1 diabetes, regardless of the type of insulin delivery method used in a 3-year follow-up of a prospective, real-world clinical trial.

Long-term results from the COMISAIR study showed that the end-of-study HbA1c values were significantly lower, compared with baseline values, in people with type 1 diabetes who used rtCGM with multiple daily injections (MDI) of insulin (7.0% [53 mmol/mol], P = .0002) or an insulin pump (6.9% [52 mmol/mol], P less than .0001). There was no significant difference between the two rtCGM delivery-method groups.

Final HbA1c values for those who used SMBG with multiple daily injections or an insulin pump were 8.0% (64 mmol/mol) and 7.7% (61 mmol/mol), respectively, but were not significantly different from baseline (P = .3574 and P = .1, respectively).

These findings could help guide physicians when discussing treatment and monitoring options with their patients, suggested study investigator Jan Šoupal, MD, PhD, of Charles University in Prague, when he presented the findings at the annual meeting of the European Association for the Study of Diabetes.

Dr. Šoupal and associates have previously reported data from the COMISAIR (Comparison of Different Treatment Modalities for Type 1 Diabetes Including Sensor-Augmented Insulin Regimens) study at 1 year of follow-up for 65 patients (Diabetes Technol Ther. 2016;18:532-8). The findings he presented at the EASD meeting, simultaneously published online in Diabetes Care, were for the full cohort of 94 patients and, with 3 years of follow-up, makes it “the longest CGM trial ever,” he said.

At the time the COMISAIR study was initiated, in 2013, “we knew that insulin pump therapy, especially in combination with real-time CGM, can improve several outcomes of patients with type 1 diabetes,” Dr. Šoupal observed. However, the effectiveness of CGM in patients with MDI was not widely described, and comparisons between continuous subcutaneous insulin infusion (CSII) and insulin MDI with rtCGM were lacking. “Moreover, we didn’t have any comparison between insulin pump therapy alone, without CGM, and MDI with CGM, and there were no long-term trials with real-time [continuous glucose monitoring].”

The aim of the COMISAIR study was therefore to compare four different treatment strategies in people with type 1 diabetes who had an HbA1c of 7%-­­10% (53-86 mmol/mol), despite MDI treatment with insulin analogues and SMBG. The treatment strategies tested were CSII plus rtCGM (n = 26), MDI plus rtCGM (n = 22), CSII plus SMBG (n = 25), and MDI plus SMBG (n = 21). Patients were not randomized to these treatment arms but exposed to all of them during a 4-day DAFNE-like training session and then allowed to choose which they would like to use according to their individual needs and preferences, reflecting real-life practice.

Dr. Šoupal pointed out that two different continuous glucose monitoring devices had been used in the trial, and that 100% of the CGM groups wore a sensor for more than 70% of the time, which was one of the prerequisites for inclusion in the trial. Good adherence was observed, with 93% of patients completing all study visits, and CGM users wearing their sensors on average 88% of the time. “This nice adherence may be connected to the pretty good results,” he observed.

In discussing the HbA1c results, Dr. Šoupal noted that “improvement observed in patients with [continuous glucose monitoring] is stable throughout 3 years, which is not always a reality for different types of treatment for diabetes.” In addition, “it is not so important how insulin is delivered, what is more important is how patients with type 1 diabetes monitor their glucose.”

Another key endpoint of the trial was time in range (70-180 mg/dL [3.9-10 mmol/L]). Results showed significantly more patients achieving this with rtCGM than with SMBG, regardless of whether they were using pump therapy or MDI. Comparing 3-year with baseline values, time in range was 72.3% versus 50.9% for rtCGM with CSII and 69% versus 48.7% for rtCGM with MDI (P less than .0001 for both). Results with SMBG with CSII or MDI were a respective 57.8% versus 50.6% (P = .0114) and 54.7% versus 51.8% (P = 1.0).

Glycemic variability was reduced in patients using insulin pumps with SBMG, and “not surprisingly, there was a reduction in both CGM-augmented groups,” Dr. Šoupal stated.

There was a reduction in the time spent in hypoglycemia from baseline to year 3 in all four groups, but that was significant only for the two rtCGM groups. Overall, there were seven severe hypoglycemia episodes, five in the SMBG groups (two in the CSII group, three in the MDI group) and two in the rtCGM groups (one each in the CSII and MDI groups), with one episode only occurring when the CGM sensor was not being worn.

Three episodes of ketoacidosis were reported – one each in the SMBG-pump, SMBG-MDI, and rtCGM-pump groups.

In summing up, Dr. Šoupal said that “real-time CGM, both with insulin pumps and with [multiple daily injections], provided significant, comparable, and stable improvement of glycemic outcomes.” He added that “treatment with CGM and MDI was more effective than treatment with insulin pump therapy alone, and that CGM and MDI can even be considered as a suitable alternative to treatment with insulin pumps and CMG for some patients.”

With many treatment options available, some will suit patients better than others, he suggested, but although “individualization of our treatment is important”, the COMISAIR data show that “it is CGM that makes the difference”.

The study was supported by the Agency for Healthcare Research and the Ministry of Health of the Czech Republic. Dr. Šoupal reported receiving honoraria from Abbott, AstraZeneca, Boehringer Ingelheim, Dexcom, Eli Lilly, Medtronic, Novo Nordisk, and Roche. Dexcom also paid for the development of the manuscript published in Diabetes Care.
 

SOURCES: Šoupal J et al. EASD 2019, Abstract 40; Šoupal J et al. Diabetes Care. 2019 Sep 17. doi: 10.2337/dc19-0888.

Publications
Publications
Topics
Article Type
Sections
Article Source

REPORTING FROM EASD 2019

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.