Diabetic Charcot neuroarthropathy (DCN), first described in 1936, occurs in less than 1% of diabetic patients, but in those diabetic subjects with distal symmetrical polyneuropathy, the overall incidence increases to 30% and the risk is even greater in those with type 1 diabetes. Factors that precipitate DCN are trauma, ischaemia due to arterio-venous shunting, increased osteoclastic activity and inflammation. DCN usually presents with a painless swollen foot and/or ankle which is 'hot to the touch'. These clinical findings are soon followed by characteristic magnetic resonance imaging (MRI) abnormalities and later X-ray changes. The joints that are most typically involved in chronological order are the tarsometatarsals followed by the naviculocuniform, sub-tarsal, talonavicular and metatarsal and tarsophalangeal. The cornerstone of therapy is prolonged (3-12 months) offloading with immobilization. Bisphosphonates may possibly accelerate recovery, whereas other unproven possible therapies include rhPTH, 1-34, calcitonin and methylprednisolone, which are not only ineffective but in some cases may also prolong the time to healing. Denosumab is potentially an efficacious, if unproven, therapy to accelerate healing. The risk of amputation is high and increases in the presence of a foot ulcer. DCN is associated with manifestations of autonomic neuropathy, including cardiac denervation, so that the risks of a cardiac event and heart failure are increased with DCN. Mortality is also increased with DCN, especially in the presence of a foot ulcer. To avoid the recurrence of DCN and especially to lower the risk of the recurrence of a foot ulcer recurrence reconstructive, surgery may be needed.
{"title":"Diabetic Charcot neuroarthropathy: A threat to both limb and life.","authors":"David S H Bell, Terri Jerkins","doi":"10.1111/dom.15994","DOIUrl":"https://doi.org/10.1111/dom.15994","url":null,"abstract":"<p><p>Diabetic Charcot neuroarthropathy (DCN), first described in 1936, occurs in less than 1% of diabetic patients, but in those diabetic subjects with distal symmetrical polyneuropathy, the overall incidence increases to 30% and the risk is even greater in those with type 1 diabetes. Factors that precipitate DCN are trauma, ischaemia due to arterio-venous shunting, increased osteoclastic activity and inflammation. DCN usually presents with a painless swollen foot and/or ankle which is 'hot to the touch'. These clinical findings are soon followed by characteristic magnetic resonance imaging (MRI) abnormalities and later X-ray changes. The joints that are most typically involved in chronological order are the tarsometatarsals followed by the naviculocuniform, sub-tarsal, talonavicular and metatarsal and tarsophalangeal. The cornerstone of therapy is prolonged (3-12 months) offloading with immobilization. Bisphosphonates may possibly accelerate recovery, whereas other unproven possible therapies include rhPTH, 1-34, calcitonin and methylprednisolone, which are not only ineffective but in some cases may also prolong the time to healing. Denosumab is potentially an efficacious, if unproven, therapy to accelerate healing. The risk of amputation is high and increases in the presence of a foot ulcer. DCN is associated with manifestations of autonomic neuropathy, including cardiac denervation, so that the risks of a cardiac event and heart failure are increased with DCN. Mortality is also increased with DCN, especially in the presence of a foot ulcer. To avoid the recurrence of DCN and especially to lower the risk of the recurrence of a foot ulcer recurrence reconstructive, surgery may be needed.</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386690","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aim: To examine the cost-effectiveness of adding finerenone to standard of care (SoC) for treating type 2 diabetes mellitus (T2DM)-related chronic kidney disease (CKD) in the United States.
Materials and methods: Based on the clinical data analysed by FIDELITY, we referenced the validated FINE-CKD model (Markov model) to evaluate the cost-effectiveness of SoC versus SoC + finerenone from the perspective of US payers. The model was cycled for 35 years in 4-month cycles, with cost and utility values derived from the published literature. The primary outcomes were incremental cost-effectiveness ratio (ICER) and quality-adjusted life years (QALYs). Deterministic and probabilistic sensitivity analyses were conducted to assess the robustness of the base-case results.
Results: The treatment strategy of finerenone plus SoC led to gains of 6.95 QALYs and had a lifetime cost of $491 745.31. Compared to SoC, that strategy yielded 0.48 more QALYs at an added cost of $65 305.72. The ICER for finerenone was $135 257.06 per QALY, which is below the willingness-to-pay threshold of United States ($150 000/QALY). The results were sensitive to the hazard ratios associated with the efficacy of finerenone and its cost. Probabilistic sensitivity analyses showed that the probability that finerenone plus SoC would be cost-effective was 57.6%.
Conclusions: For patients with T2DM-related CKD, adding finerenone to SoC may be a cost-effective option in the United States. Reasonable price reductions for finerenone could potentially benefit more patients.
{"title":"Cost-effectiveness of finerenone added to standard of care for patients with type 2 diabetes-related chronic kidney disease in the United States.","authors":"Cailin Zheng, Jinneng Wu, Na Li, Xiaoxia Wei, Zhixiong Huang, Lingbin Chen, Zhou Chen","doi":"10.1111/dom.15997","DOIUrl":"https://doi.org/10.1111/dom.15997","url":null,"abstract":"<p><strong>Aim: </strong>To examine the cost-effectiveness of adding finerenone to standard of care (SoC) for treating type 2 diabetes mellitus (T2DM)-related chronic kidney disease (CKD) in the United States.</p><p><strong>Materials and methods: </strong>Based on the clinical data analysed by FIDELITY, we referenced the validated FINE-CKD model (Markov model) to evaluate the cost-effectiveness of SoC versus SoC + finerenone from the perspective of US payers. The model was cycled for 35 years in 4-month cycles, with cost and utility values derived from the published literature. The primary outcomes were incremental cost-effectiveness ratio (ICER) and quality-adjusted life years (QALYs). Deterministic and probabilistic sensitivity analyses were conducted to assess the robustness of the base-case results.</p><p><strong>Results: </strong>The treatment strategy of finerenone plus SoC led to gains of 6.95 QALYs and had a lifetime cost of $491 745.31. Compared to SoC, that strategy yielded 0.48 more QALYs at an added cost of $65 305.72. The ICER for finerenone was $135 257.06 per QALY, which is below the willingness-to-pay threshold of United States ($150 000/QALY). The results were sensitive to the hazard ratios associated with the efficacy of finerenone and its cost. Probabilistic sensitivity analyses showed that the probability that finerenone plus SoC would be cost-effective was 57.6%.</p><p><strong>Conclusions: </strong>For patients with T2DM-related CKD, adding finerenone to SoC may be a cost-effective option in the United States. Reasonable price reductions for finerenone could potentially benefit more patients.</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386689","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yutong Liu, Xiatiguli Shamaitijiang, Loren Skudder-Hill, Wandia Kimita, Ivana R Sequeira-Bisson, Maxim S Petrov
Aim: To investigate the associations of high-density lipoprotein (HDL) subfractions and apolipoprotein A-I (apo A-I) with fat in the pancreas.
Methods: A total of 170 individuals were studied. All participants underwent magnetic resonance imaging on a single 3.0-Tesla scanner to determine the presence/absence of fatty pancreas. HDL subfractions were measured using a commercially available lipoprotein subfractions testing system and classed as large, intermediate and small HDL. Both unadjusted and adjusted (accounting for demographics, anthropometrics, insulin resistance and other covariates) logistic regression models were built.
Results: Individuals with fatty pancreas had significantly lower circulating levels of the large HDL class and apo A-I. Every unit decrease in the large HDL class was associated with a 93% increase in the likelihood of fatty pancreas in the most adjusted model (P < .001). Every unit decrease in apo A-I was associated with a 45% increase in the likelihood of fatty pancreas in the most adjusted model (P = .012). The intermediate and small HDL classes were not significantly associated with fatty pancreas.
Conclusions: Fat in the pancreas is inversely associated with the circulating levels of large HDL particles and apo A-I. Purposely designed studies are warranted to investigate the potential of fatty pancreas as an indicator of the risk of cardiovascular diseases.
{"title":"Relationship of high-density lipoprotein subfractions and apolipoprotein A-I with fat in the pancreas.","authors":"Yutong Liu, Xiatiguli Shamaitijiang, Loren Skudder-Hill, Wandia Kimita, Ivana R Sequeira-Bisson, Maxim S Petrov","doi":"10.1111/dom.15990","DOIUrl":"https://doi.org/10.1111/dom.15990","url":null,"abstract":"<p><strong>Aim: </strong>To investigate the associations of high-density lipoprotein (HDL) subfractions and apolipoprotein A-I (apo A-I) with fat in the pancreas.</p><p><strong>Methods: </strong>A total of 170 individuals were studied. All participants underwent magnetic resonance imaging on a single 3.0-Tesla scanner to determine the presence/absence of fatty pancreas. HDL subfractions were measured using a commercially available lipoprotein subfractions testing system and classed as large, intermediate and small HDL. Both unadjusted and adjusted (accounting for demographics, anthropometrics, insulin resistance and other covariates) logistic regression models were built.</p><p><strong>Results: </strong>Individuals with fatty pancreas had significantly lower circulating levels of the large HDL class and apo A-I. Every unit decrease in the large HDL class was associated with a 93% increase in the likelihood of fatty pancreas in the most adjusted model (P < .001). Every unit decrease in apo A-I was associated with a 45% increase in the likelihood of fatty pancreas in the most adjusted model (P = .012). The intermediate and small HDL classes were not significantly associated with fatty pancreas.</p><p><strong>Conclusions: </strong>Fat in the pancreas is inversely associated with the circulating levels of large HDL particles and apo A-I. Purposely designed studies are warranted to investigate the potential of fatty pancreas as an indicator of the risk of cardiovascular diseases.</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Prevalence of individuals with diabetes and chronic kidney disease indicated for finerenone treatment in the United States: A National Health and Nutrition Examination Survey-based estimation.","authors":"Xiufang Kong, Wei Wang","doi":"10.1111/dom.16006","DOIUrl":"https://doi.org/10.1111/dom.16006","url":null,"abstract":"","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Norbert Hermanns, Dominic Ehrmann, Bernhard Kulzer, Laura Klinker, Thomas Haak, Andreas Schmitt
Aim: To analyse the potential drivers (glucose level, complications, diabetes type, gender, age and mental health) of diabetes symptoms using continuous glucose monitoring (CGM) and ecological momentary assessment.
Materials and methods: Participants used a smartphone application to rate 25 diabetes symptoms in their daily lives over 8 days. These symptoms were grouped into four blocks so that each symptom was rated six times on 2 days (noon, afternoon and evening). The symptom ratings were associated with the glucose levels for the previous 2 hours, measured with CGM. Linear mixed-effects models were used, allowing for nested random effects and the conduct of N = 1 analysis of individual associations.
Results: In total, 192 individuals with type 1 diabetes and 179 with type 2 diabetes completed 6380 app check-ins. Four symptoms showed a significant negative association with glucose values, indicating higher ratings at lower glucose (speech difficulties, P = .003; coordination problems, P = .00005; confusion, P = .049; and food cravings, P = .0003). Four symptoms showed a significant positive association with glucose values, indicating higher scores at higher glucose (thirst, P = .0001; urination, P = .0003; taste disturbances, P = .021; and itching, P = .0120). There were also significant positive associations between microangiopathy and eight symptoms. Elevated depression and diabetes distress were associated with higher symptom scores. N = 1 analysis showed highly idiosyncratic associations between symptom reports and glucose levels.
Conclusions: The N = 1 analysis facilitated the creation of personalized symptom profiles related to glucose levels with consideration of factors such as complications, gender, body mass index, depression and diabetes distress. This approach can enhance precision monitoring for diabetes symptoms in precision medicine.
{"title":"Somatic and mental symptoms associated with dysglycaemia, diabetes-related complications and mental conditions in people with diabetes: Assessments in daily life using continuous glucose monitoring and ecological momentary assessment.","authors":"Norbert Hermanns, Dominic Ehrmann, Bernhard Kulzer, Laura Klinker, Thomas Haak, Andreas Schmitt","doi":"10.1111/dom.15983","DOIUrl":"https://doi.org/10.1111/dom.15983","url":null,"abstract":"<p><strong>Aim: </strong>To analyse the potential drivers (glucose level, complications, diabetes type, gender, age and mental health) of diabetes symptoms using continuous glucose monitoring (CGM) and ecological momentary assessment.</p><p><strong>Materials and methods: </strong>Participants used a smartphone application to rate 25 diabetes symptoms in their daily lives over 8 days. These symptoms were grouped into four blocks so that each symptom was rated six times on 2 days (noon, afternoon and evening). The symptom ratings were associated with the glucose levels for the previous 2 hours, measured with CGM. Linear mixed-effects models were used, allowing for nested random effects and the conduct of N = 1 analysis of individual associations.</p><p><strong>Results: </strong>In total, 192 individuals with type 1 diabetes and 179 with type 2 diabetes completed 6380 app check-ins. Four symptoms showed a significant negative association with glucose values, indicating higher ratings at lower glucose (speech difficulties, P = .003; coordination problems, P = .00005; confusion, P = .049; and food cravings, P = .0003). Four symptoms showed a significant positive association with glucose values, indicating higher scores at higher glucose (thirst, P = .0001; urination, P = .0003; taste disturbances, P = .021; and itching, P = .0120). There were also significant positive associations between microangiopathy and eight symptoms. Elevated depression and diabetes distress were associated with higher symptom scores. N = 1 analysis showed highly idiosyncratic associations between symptom reports and glucose levels.</p><p><strong>Conclusions: </strong>The N = 1 analysis facilitated the creation of personalized symptom profiles related to glucose levels with consideration of factors such as complications, gender, body mass index, depression and diabetes distress. This approach can enhance precision monitoring for diabetes symptoms in precision medicine.</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386715","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nadine Beetz, Brigitte Kalsch, Thomas Forst, Bernhard Schmid, Armin Schultz, Anita M Hennige
Aims: Pharmacotherapeutic options for obesity treatment include glucagon-like peptide-1 receptor (GLP-1R) agonists, for example, liraglutide. However, an unmet need remains, particularly in patients with a high body mass index (BMI), as GLP-1R agonists are associated with gastrointestinal adverse events (AEs) and some patients do not respond to treatment. Neuropeptide Y (NPY) and peptide YY bind G-protein-coupled Y receptors and represent attractive targets for modulating bodyweight.
Materials and methods: This first-in-human, three-part, partially blinded phase I study (NCT04903509) investigated the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of single ascending doses of the peptidic NPY2R agonist BI 1820237, with/without low-dose liraglutide: part 1 (participants randomized to receive BI 1820237: 0.075-2.4 mg or placebo), part 2 (BI 1820237: 1.2 mg or placebo) and part 3 (BI 1820237: 0.025-1.2 mg + liraglutide 0.6 mg or placebo + liraglutide 0.6 mg). Primary endpoint is the proportion of participants with drug-related AEs. Secondary endpoints are tolerability, PK and PD.
Results: In total, 95 otherwise healthy men with increased BMI (25.0-34.9 kg/m2) were randomized. Drug-related AEs, mainly gastrointestinal events, were reported by 39.0% of participants (n = 23) in parts 1 + 2 and 30.6% of participants (n = 11) in part 3; one drug-related AE (11.1%, part 3) was reported in a participant receiving placebo with liraglutide. Post-dose paracetamol PK suggested that BI 1820237 and low-dose liraglutide exhibited additive effects on gastric emptying.
Conclusions: BI 1820237 treatment was associated with transient nausea and vomiting at higher doses. No differences in tolerability were observed when combined with liraglutide; effects on gastric emptying appeared additive.
目的:治疗肥胖症的药物治疗选择包括胰高血糖素样肽-1受体(GLP-1R)激动剂,例如利拉鲁肽。然而,由于GLP-1R激动剂与胃肠道不良反应(AEs)有关,而且一些患者对治疗没有反应,因此仍有未满足的需求,特别是对于体重指数(BMI)较高的患者。神经肽 Y(NPY)和肽 YY 与 G 蛋白偶联 Y 受体结合,是调节体重的诱人靶点:这项首次进行的人体三部分部分盲法 I 期研究(NCT04903509)调查了单次递增剂量肽类 NPY2R 激动剂 BI 1820237 的安全性、耐受性、药代动力学 (PK) 和药效学 (PD),以及低剂量利拉鲁肽/不使用低剂量利拉鲁肽的情况:第一部分(参与者随机接受 BI 1820237:0.075-2.4 mg 或安慰剂);第二部分(参与者随机接受 BI 1820237:0.075-2.4 mg 或安慰剂);第三部分(参与者随机接受 BI 1820237:0.075-2.4 mg 或安慰剂)。075-2.4毫克或安慰剂)、第2部分(BI 1820237:1.2毫克或安慰剂)和第3部分(BI 1820237:0.025-1.2毫克+利拉鲁肽0.6毫克或安慰剂+利拉鲁肽0.6毫克)。主要终点是出现药物相关不良反应的参与者比例。次要终点为耐受性、PK 和 PD:共有 95 名体重指数较高(25.0-34.9 kg/m2)的健康男性接受了随机治疗。第1+2部分中39.0%的参与者(n=23)和第3部分中30.6%的参与者(n=11)报告了与药物相关的AE,主要是胃肠道事件;一名接受安慰剂和利拉鲁肽的参与者报告了一次与药物相关的AE(11.1%,第3部分)。用药后扑热息痛的PK表明,BI 1820237和小剂量利拉鲁肽对胃排空具有相加效应:结论:BI 1820237治疗剂量较大时会出现短暂的恶心和呕吐。结论:BI 1820237 与利拉鲁肽联合治疗时会出现短暂的恶心和呕吐,但两者的耐受性没有差异;对胃排空的影响似乎是相加的。
{"title":"A randomized phase I study of BI 1820237, a novel neuropeptide Y receptor type 2 agonist, alone or in combination with low-dose liraglutide in otherwise healthy men with overweight or obesity.","authors":"Nadine Beetz, Brigitte Kalsch, Thomas Forst, Bernhard Schmid, Armin Schultz, Anita M Hennige","doi":"10.1111/dom.15984","DOIUrl":"https://doi.org/10.1111/dom.15984","url":null,"abstract":"<p><strong>Aims: </strong>Pharmacotherapeutic options for obesity treatment include glucagon-like peptide-1 receptor (GLP-1R) agonists, for example, liraglutide. However, an unmet need remains, particularly in patients with a high body mass index (BMI), as GLP-1R agonists are associated with gastrointestinal adverse events (AEs) and some patients do not respond to treatment. Neuropeptide Y (NPY) and peptide YY bind G-protein-coupled Y receptors and represent attractive targets for modulating bodyweight.</p><p><strong>Materials and methods: </strong>This first-in-human, three-part, partially blinded phase I study (NCT04903509) investigated the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of single ascending doses of the peptidic NPY2R agonist BI 1820237, with/without low-dose liraglutide: part 1 (participants randomized to receive BI 1820237: 0.075-2.4 mg or placebo), part 2 (BI 1820237: 1.2 mg or placebo) and part 3 (BI 1820237: 0.025-1.2 mg + liraglutide 0.6 mg or placebo + liraglutide 0.6 mg). Primary endpoint is the proportion of participants with drug-related AEs. Secondary endpoints are tolerability, PK and PD.</p><p><strong>Results: </strong>In total, 95 otherwise healthy men with increased BMI (25.0-34.9 kg/m<sup>2</sup>) were randomized. Drug-related AEs, mainly gastrointestinal events, were reported by 39.0% of participants (n = 23) in parts 1 + 2 and 30.6% of participants (n = 11) in part 3; one drug-related AE (11.1%, part 3) was reported in a participant receiving placebo with liraglutide. Post-dose paracetamol PK suggested that BI 1820237 and low-dose liraglutide exhibited additive effects on gastric emptying.</p><p><strong>Conclusions: </strong>BI 1820237 treatment was associated with transient nausea and vomiting at higher doses. No differences in tolerability were observed when combined with liraglutide; effects on gastric emptying appeared additive.</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142379662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Long-term glycaemic improvement with the initiation of an automated insulin delivery system in insulin pump-naïve older adults with type 1 diabetes.","authors":"Kagan E Karakus, Emma Mason, Halis K Akturk","doi":"10.1111/dom.15996","DOIUrl":"https://doi.org/10.1111/dom.15996","url":null,"abstract":"","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jun Hwa Hong, Myung Jin Kim, Kyung Wan Min, Jong Chul Won, Tae Nyun Kim, Byung-Wan Lee, Jun Goo Kang, Jae Hyeon Kim, Jung Hwan Park, Bon Jeong Ku, Chang Beom Lee, Sang Yong Kim, Ho Sang Shon, Woo Je Lee, Joong-Yeol Park
Aims: To evaluate the efficacy and safety of add-on dapagliflozin in patients with type 2 diabetes mellitus (T2D) who had inadequate glycaemic control with metformin and linagliptin.
Materials and methods: A total of 235 patients with inadequate response to metformin (≥1000 mg/day) plus linagliptin (5 mg/day) were randomized to receive either dapagliflozin/linagliptin fixed-dose combination (FDC [AJU-A51]) 10/5 mg/day (n = 117) or linagliptin 5 mg plus placebo (n = 118) for 24 weeks. After the main treatment period, patients who received linagliptin plus placebo were treated with AJU-A51 for an additional 28 weeks. Change in glycated haemoglobin (HbA1c) from baseline to Week 24 was the primary endpoint.
Results: AJU-A51 significantly reduced HbA1c levels (from 7.93% ± 0.82% to 7.11% ± 0.61%) compared with linagliptin plus placebo (from 7.80% ± 0.71% to 7.87% ± 0.94%), with a least squares mean difference of -0.88% (95% confidence interval -1.07 to -0.68; p < 0.0001) at 24 weeks. The AJU-A51 group had a significantly higher proportion of patients who achieved HbA1c <7.0% at Week 24 than the control group (44.8% vs. 18.6%; p < 0.001). The AJU-A51 group maintained glycaemic efficacy up to 52 weeks, whereas the control group showed a substantial reduction in HbA1c after switching to AJU-A51 in the extension study period. Both groups had similar incidence of treatment-emergent and serious adverse events, and no cases of symptomatic hypoglycaemia were reported.
Conclusions: Dapagliflozin and linagliptin FDC (AJU-A51) showed potent glucose-lowering effects, with good tolerability, in patients with T2D who had poor glycaemic control on metformin and linagliptin (ClinicalTrials.gov [NCT06329674]).
{"title":"Efficacy and safety of a fixed-dose combination of dapagliflozin and linagliptin (AJU-A51) in patients with type 2 diabetes mellitus: A multicentre, randomized, double-blind, parallel-group, placebo-controlled phase III study.","authors":"Jun Hwa Hong, Myung Jin Kim, Kyung Wan Min, Jong Chul Won, Tae Nyun Kim, Byung-Wan Lee, Jun Goo Kang, Jae Hyeon Kim, Jung Hwan Park, Bon Jeong Ku, Chang Beom Lee, Sang Yong Kim, Ho Sang Shon, Woo Je Lee, Joong-Yeol Park","doi":"10.1111/dom.15985","DOIUrl":"https://doi.org/10.1111/dom.15985","url":null,"abstract":"<p><strong>Aims: </strong>To evaluate the efficacy and safety of add-on dapagliflozin in patients with type 2 diabetes mellitus (T2D) who had inadequate glycaemic control with metformin and linagliptin.</p><p><strong>Materials and methods: </strong>A total of 235 patients with inadequate response to metformin (≥1000 mg/day) plus linagliptin (5 mg/day) were randomized to receive either dapagliflozin/linagliptin fixed-dose combination (FDC [AJU-A51]) 10/5 mg/day (n = 117) or linagliptin 5 mg plus placebo (n = 118) for 24 weeks. After the main treatment period, patients who received linagliptin plus placebo were treated with AJU-A51 for an additional 28 weeks. Change in glycated haemoglobin (HbA1c) from baseline to Week 24 was the primary endpoint.</p><p><strong>Results: </strong>AJU-A51 significantly reduced HbA1c levels (from 7.93% ± 0.82% to 7.11% ± 0.61%) compared with linagliptin plus placebo (from 7.80% ± 0.71% to 7.87% ± 0.94%), with a least squares mean difference of -0.88% (95% confidence interval -1.07 to -0.68; p < 0.0001) at 24 weeks. The AJU-A51 group had a significantly higher proportion of patients who achieved HbA1c <7.0% at Week 24 than the control group (44.8% vs. 18.6%; p < 0.001). The AJU-A51 group maintained glycaemic efficacy up to 52 weeks, whereas the control group showed a substantial reduction in HbA1c after switching to AJU-A51 in the extension study period. Both groups had similar incidence of treatment-emergent and serious adverse events, and no cases of symptomatic hypoglycaemia were reported.</p><p><strong>Conclusions: </strong>Dapagliflozin and linagliptin FDC (AJU-A51) showed potent glucose-lowering effects, with good tolerability, in patients with T2D who had poor glycaemic control on metformin and linagliptin (ClinicalTrials.gov [NCT06329674]).</p>","PeriodicalId":158,"journal":{"name":"Diabetes, Obesity & Metabolism","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}