1
|
McFarlin BE, Duffin KL, Konkar A. Incretin and glucagon receptor polypharmacology in chronic kidney disease. Am J Physiol Endocrinol Metab 2024; 326:E747-E766. [PMID: 38477666 PMCID: PMC11551006 DOI: 10.1152/ajpendo.00374.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/10/2024] [Indexed: 03/14/2024]
Abstract
Chronic kidney disease is a debilitating condition associated with significant morbidity and mortality. In recent years, the kidney effects of incretin-based therapies, particularly glucagon-like peptide-1 receptor agonists (GLP-1RAs), have garnered substantial interest in the management of type 2 diabetes and obesity. This review delves into the intricate interactions between the kidney, GLP-1RAs, and glucagon, shedding light on their mechanisms of action and potential kidney benefits. Both GLP-1 and glucagon, known for their opposing roles in regulating glucose homeostasis, improve systemic risk factors affecting the kidney, including adiposity, inflammation, oxidative stress, and endothelial function. Additionally, these hormones and their pharmaceutical mimetics may have a direct impact on the kidney. Clinical studies have provided evidence that incretins, including those incorporating glucagon receptor agonism, are likely to exhibit improved kidney outcomes. Although further research is necessary, receptor polypharmacology holds promise for preserving kidney function through eliciting vasodilatory effects, influencing volume and electrolyte handling, and improving systemic risk factors.
Collapse
Affiliation(s)
- Brandon E McFarlin
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| | - Kevin L Duffin
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| | - Anish Konkar
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| |
Collapse
|
2
|
Sun Z, Liu Y, Zhao Y, Xu Y. Animal Models of Type 2 Diabetes Complications: A Review. Endocr Res 2024; 49:46-58. [PMID: 37950485 DOI: 10.1080/07435800.2023.2278049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Diabetes mellitus is a multifactorial metabolic disease, of which type 2 diabetes (T2D) is one of the most common. The complications of diabetes are far more harmful than diabetes itself. Type 2 diabetes complications include diabetic nephropathy (DN), diabetic heart disease, diabetic foot ulcers (DFU), diabetic peripheral neuropathy (DPN), and diabetic retinopathy (DR) et al. Many animal models have been developed to study the pathogenesis of T2D and discover an effective strategy to treat its consequences. In this sense, it is crucial to choose the right animal model for the corresponding diabetic complication. This paper summarizes and classifies the animal modeling approaches to T2D complications and provides a comprehensive review of their advantages and disadvantages. It is hopeful that this paper will provide theoretical support for animal trials of diabetic complications.
Collapse
Affiliation(s)
- Zhongyan Sun
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
| | - Yadi Liu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, Taipa, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine,Macau University of Science and Technology, Zhuhai, PR China
- Macau University of Science and Technology, Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, PR China
| |
Collapse
|
3
|
Muskiet MHA, Elders PJM, van Raalte DH. Diabetes care in older people: a call for action. THE LANCET. HEALTHY LONGEVITY 2023; 4:e657-e659. [PMID: 38042157 DOI: 10.1016/s2666-7568(23)00234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 12/04/2023] Open
Affiliation(s)
- Marcel H A Muskiet
- Diabetes Centre, Department of Internal Medicine, Amsterdam University Medical Centers, VU University, 1081 HV Amsterdam, Netherlands.
| | - Petra J M Elders
- Department of General Practice and Elderly Care Medicine, and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, VU University, Amsterdam, Netherlands
| | - Daniël H van Raalte
- Diabetes Centre, Department of Internal Medicine, Amsterdam University Medical Centers, VU University, 1081 HV Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, VU University, Amsterdam, Netherlands
| |
Collapse
|
4
|
Advances in the Pharmacological Management of Diabetic Nephropathy: A 2022 International Update. Biomedicines 2023; 11:biomedicines11020291. [PMID: 36830828 PMCID: PMC9953496 DOI: 10.3390/biomedicines11020291] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/24/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD) worldwide. Its pathogenesis encompasses functional alterations involving elevated intraglomerular and systemic pressure, increased activity of the renin-angiotensin system (RAS) and oxidative stress, and the eventual development of renal fibrosis. The management of DN involves the optimization of blood pressure (BP) and blood glucose targets. However, treatment of these risk factors slows down but does not stop the progression of DN. Innovative pharmacologic therapies for dyslipidemia and type 2 diabetes mellitus (T2DM) could play a key role in bridging this gap and attenuating the residual risk of DN beyond traditional risk factor management. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), sodium-glucose cotransporter-2 inhibitors (SGLT-2is), and inhibitors of mineralocorticoid receptor-mediated sodium reabsorption are recently introduced drug classes that have been shown to have positive effects on kidney function in individuals with T2DM. The aim of this review is to provide an update on the therapeutic options available in order to prevent or slow the onset and progression of DN in diabetic patients.
Collapse
|
5
|
Kugathasan L, Dubrofsky L, Advani A, Cherney DZI. The anti-hypertensive effects of sodium-glucose cotransporter-2 inhibitors. Expert Rev Cardiovasc Ther 2023; 21:15-34. [PMID: 36524239 DOI: 10.1080/14779072.2023.2159810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Hypertension is a well-established risk factor for cardiovascular (CV) events in patients with chronic kidney disease (CKD), heart failure, obesity, and diabetes. Despite the usual prescribed antihypertensive therapies, many patients fail to achieve the recommended blood pressure (BP) targets. AREAS COVERED This review summarizes the clinical BP-lowering data presented in major CV and kidney outcome trials for sodium-glucose cotransporter-2 (SGLT2) inhibitors, as well as smaller dedicated BP trials in high-risk individuals with and without diabetes. We have also highlighted potential mechanisms that may contribute to the antihypertensive effects of SGLT2 inhibitors, including natriuresis and hemodynamic changes, a loop diuretic-like effect, and alterations in vascular physiology. EXPERT OPINION The antihypertensive properties of SGLT2 inhibitors are generally modest but may be larger in certain patient populations. SGLT2 inhibitors may have an additional role as an adjunctive BP-lowering therapy in patients with hypertension at high risk of CV disease or kidney disease.
Collapse
Affiliation(s)
- Luxcia Kugathasan
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Cardiovascular Sciences Collaborative Specialization, University of Toronto, Toronto, Ontario, Canada
| | - Lisa Dubrofsky
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Nephrology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Division of Nephrology, Women's College Hospital, Toronto, Ontario, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Cardiovascular Sciences Collaborative Specialization, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Nomogram-Based Chronic Kidney Disease Prediction Model for Type 1 Diabetes Mellitus Patients Using Routine Pathological Data. J Pers Med 2022; 12:jpm12091507. [PMID: 36143293 PMCID: PMC9501949 DOI: 10.3390/jpm12091507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) patients are a significant threat to chronic kidney disease (CKD) development during their life. However, there is always a high chance of delay in CKD detection because CKD can be asymptomatic, and T1DM patients bypass traditional CKD tests during their routine checkups. This study aims to develop and validate a prediction model and nomogram of CKD in T1DM patients using readily available routine checkup data for early CKD detection. This research utilized 1375 T1DM patients’ sixteen years of longitudinal data from multi-center Epidemiology of Diabetes Interventions and Complications (EDIC) clinical trials conducted at 28 sites in the USA and Canada and considered 17 routinely available features. Three feature ranking algorithms, extreme gradient boosting (XGB), random forest (RF), and extremely randomized trees classifier (ERT), were applied to create three feature ranking lists, and logistic regression analyses were performed to develop CKD prediction models using these ranked feature lists to identify the best performing top-ranked features combination. Finally, the most significant features were selected to develop a multivariate logistic regression-based CKD prediction model for T1DM patients. This model was evaluated using sensitivity, specificity, accuracy, precision, and F1 score on train and test data. A nomogram of the final model was further generated for easy application in clinical practices. Hypertension, duration of diabetes, drinking habit, triglycerides, ACE inhibitors, low-density lipoprotein (LDL) cholesterol, age, and smoking habit were the top-8 features ranked by the XGB model and identified as the most important features for predicting CKD in T1DM patients. These eight features were selected to develop the final prediction model using multivariate logistic regression, which showed 90.04% and 88.59% accuracy in internal and test data validation. The proposed model showed excellent performance and can be used for CKD identification in T1DM patients during routine checkups.
Collapse
|
7
|
Zhang J, Xian TZ, Wu MX, Li C, Wang W, Man F, Zhang X, Wang X, Pan Q, Guo L. Comparing the effects of twice-daily exenatide and insulin on renal function in patients with type 2 diabetes mellitus: secondary analysis of a randomized controlled trial. J Investig Med 2022; 70:1529-1535. [PMID: 35725020 DOI: 10.1136/jim-2021-002237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 11/03/2022]
Abstract
This is a secondary analysis of a randomized controlled trial (RCT) on the effects of the glucagon-like peptide-1 receptor agonists exenatide and insulin aspartate 30 injection on carotid intima-media thickness. Here, we report the renal outcomes of the intervention in patients with type 2 diabetes mellitus (T2DM). Data from the RCT study was used to evaluate the effect of exenatide or insulin given for 52 weeks on estimated glomerular filtration rate (eGFR) in patients with T2DM. The primary end point was the change in the eGFR from baseline between the exenatide and insulin groups in normal versus overweight patients and patients with obesity. The secondary end point was the correlation between change in eGFR and oxidative stress, glycemic control, and dyslipidemia. There was a significant difference in eGFR between the insulin and exenatide groups at 52 weeks (p=0.0135). Within the insulin group, the eGFR remained below baseline at 52 weeks in all patients, and there was an increase in body weight in the normal group compared with the overweight patients and patients with obesity. The opposite was observed in the exenatide group. A decrease in body weight was prominent in the exenatide group at 52 weeks (p<0.05), the eGFR was below baseline in overweight patients and patients with obesity and significantly above baseline in the normal group (p<0.05). The eGFR was positively correlated to 8-oxo-7,8-dihydroguanosine in the insulin group (p<0.05) but not the exenatide group. It can be concluded that compared with insulin, exenatide may improve renal function in overweight patients and patients with obesity more than in normal-weight patients with T2DM, but a further RCT is needed to confirm this effect.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Tong-Zhang Xian
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Ming-Xiao Wu
- Department of Ultrasound, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Chen Li
- Department of Ultrasound, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Weihao Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Fuli Man
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Xianbo Zhang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Xiaoxia Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, Beijing, China
| |
Collapse
|
8
|
van Ruiten CC, Hesp AC, van Raalte DH. Sodium glucose cotransporter-2 inhibitors protect the cardiorenal axis: Update on recent mechanistic insights related to kidney physiology. Eur J Intern Med 2022; 100:13-20. [PMID: 35414444 DOI: 10.1016/j.ejim.2022.03.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022]
Abstract
Sodium glucose cotransporter-2 (SGLT2) inhibitors have acquired a central role in the treatment of type 2 diabetes, chronic kidney disease including diabetic kidney disease, and heart failure with reduced ejection fraction. SGLT2 inhibitors lower glucose levels by inducing glycosuria. In addition, SGLT2 inhibitors improve cardiovascular outcomes (3-point MACE), end-stage kidney disease, hospitalization for heart failure, and cardiovascular mortality in people with and without diabetes. The mechanisms underlying these benefits have been extensively investigated, but remain poorly understood. In this review, we first summarize recent trial evidence and subsequently focus on (1) the mechanisms by which SGLT2 inhibitors improve kidney outcomes and (2) the potential role of the kidneys in mediating the cardioprotective effects of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Charlotte C van Ruiten
- Amsterdam Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers (Amsterdam UMC), location VU University Medical Center, De Boelelaan 1117 (room ZH 4A63), Amsterdam 1081 HV, the Netherland.
| | - Anne C Hesp
- Amsterdam Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers (Amsterdam UMC), location VU University Medical Center, De Boelelaan 1117 (room ZH 4A63), Amsterdam 1081 HV, the Netherland
| | - Daniël H van Raalte
- Amsterdam Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers (Amsterdam UMC), location VU University Medical Center, De Boelelaan 1117 (room ZH 4A63), Amsterdam 1081 HV, the Netherland; Department of Vascular Medicine Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, the Netherland
| |
Collapse
|
9
|
Mohammad Zadeh Gharabaghi MA, Rezvanfar MR, Saeedi N, Aghajani F, Alirezaei M, Yarahmadi P, Nakhostin-Ansari A. Comparison of effects of Empagliflozin and Linagliptin on renal function and glycaemic control: a double-blind, randomized clinical trial. Clin Diabetes Endocrinol 2022; 8:5. [PMID: 35610696 PMCID: PMC9131518 DOI: 10.1186/s40842-022-00142-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background This study aimed to compare the effects of Linagliptin and Empagliflozin on renal function and glycaemic control in patients with type 2 diabetes mellitus (DM). Method We conducted a randomized, double-blind, parallel trial on patients aged 30 to 80 years with type 2 DM and HbA1c ≤ 9%, regardless of background medical therapy, to compare the effects of Empagliflozin and Linagliptin on albuminuria, FBS, HbA1c, and eGFR. Participants were given the mentioned drugs for 12 weeks. Statistical analysis was performed using appropriate tests in IBM™SPSS® statistics software for windows version 24. Results In total, 60 patients participated in the study, thirty patients in each group. The mean age of participants was 56.8 (SD = 8.15) in the Empagliflozin group and 60.9 (SD = 7.22) in the Linagliptin group. Before the intervention, FBS, HbA1C, and albuminuria values were significantly higher in the Empagliflozin group than those in the Linagliptin group (P < 0.05), but there was no significant difference between groups regarding eGFR (P = 0.271). Changes in the FBS, HbA1C, and eGFR were not significantly different between groups (P > 0.05), but there was more decrease in albuminuria in the Empagliflozin group compared to the Linagliptin group (P = 0.001, Cohen’s d = 0.98). Conclusions Regardless of baseline albuminuria, eGFR, or HbA1c, Empagliflozin 10 mg daily significantly reduced albuminuria at 12 weeks compared to Linagliptin 5 mg daily in patients with type 2 diabetes. Trial registration Iranian Registry of Clinical Trials, IRCT20200722048176N1. Registered 3 August 2020.
Collapse
Affiliation(s)
| | - Mohammad Reza Rezvanfar
- Internal Medicine Department, Arak University of Medical Sciences, A'lam-Al-Hoda Street, Shahid Shiroodi Street, Arak, Iran
| | - Nasser Saeedi
- Internal Medicine Department, Arak University of Medical Sciences, A'lam-Al-Hoda Street, Shahid Shiroodi Street, Arak, Iran
| | - Faezeh Aghajani
- Research Development Center, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Pourya Yarahmadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Nakhostin-Ansari
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Huang Y, Wang Y, Liu C, Zhou Y, Wang X, Cheng B, Kui C, Wang Y. C-peptide, glycaemic control, and diabetic complications in type 2 diabetes mellitus: A real-world study. Diabetes Metab Res Rev 2022; 38:e3514. [PMID: 34841643 DOI: 10.1002/dmrr.3514] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/02/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To explore the relationship between C-peptide and glycaemic control rate and diabetic complications (microvascular complication and cerebral infarction) and provide evidence for stratified treatment of type 2 diabetes mellitus (T2DM)-based C-peptide. METHOD This is a cross-sectional real-world observational study. According to the inclusion and exclusion criteria, we studied 1377 patients with T2DM, grouped by fasting C-peptide and HOMA-IR. Blood samples were collected after fasting overnight. Logistic regression was used to analyse the relationship among fasting C-peptide, HOMA-IR, C2/C0 ratio (the ratio of 2 h postprandial C-peptide to fasting C-peptide), glycaemic control rate, and occurrence of diabetic complications. Restricted cubic spline (RCS) curves based on logistic regression were used to evaluate the relationship between C-peptide, glycaemic control rate, and diabetic kidney disease (DKD). RESULTS Patients were subdivided according to their fasting C-peptide in 4 groups (Q1,Q2,Q3,Q4). Patients of group Q3 (1.71 ≤ C-peptide < 2.51 ng/ml) showed the lowest incidence of DKD, diabetic retinopathy (DR), and rate of insulin absorption as welll as higher glycaemic control rate. Logistic regression shows that the probability of reaching glycemic control increased with higher levels of C-peptide, compared with group Q1, after adjusting for age, gender, duration of diabetes, body mass index, systolic blood pressure, diastolic blood pressure, creatinine, low-density lipoprotein, triglyceride, total cholesterol, and high-density lipoprotein. RCS curve shows that, when C-peptide is ≤2.68 ng/ml, the incidence of not reaching glycaemic control decreases with increasing C-peptide. The possibility of not reaching glycaemic control decreased with increasing C2/C0, when C-peptide is ≥1.71 ng/ml. RCS curve shows that the relationship between C-peptide and DKD follows a U-style curve. When C-peptide is <2.84 ng/ml, the incidence of DKD decreased with increasing C-peptide. With the increase in the C2/C0 ratio, the incidence of DKD, DR, and fatty liver did not decrease. CONCLUSION When C-peptide is ≥ 1.71 and < 2.51 ng/ml, patients with T2DM had a higher glycemic control rate. Excessive C-peptide plays different roles in DKD and DR; C-peptide may promote the incidence of DKD but protects patients from DR. Higher C2/C0 ratio is important for reaching glycaemic control but cannot reduce the risk of DKD, DR, and fatty liver.
Collapse
Affiliation(s)
- Yajing Huang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yahao Wang
- Medicine College, Qingdao University, Qingdao, China
| | - Chuanfeng Liu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Zhou
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bingfei Cheng
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Che Kui
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
The effect of sodium-glucose cotransporter 2 inhibitor (tofogliflozin) on renal tubular damage in diabetic patients without albuminuria. Int Urol Nephrol 2021; 54:1907-1914. [PMID: 34843041 DOI: 10.1007/s11255-021-03064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 11/11/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE The sodium-glucose cotransporter 2 (SGLT2) inhibitors comprise a new class of glucose-lowering drugs for individuals with diabetes. Large-scale clinical trials indicated that SGLT2 inhibitors have both a cardiovascular-protective and renal-protective effects. A reduction in glomerular hyperfiltration and a decrease in albuminuria are suspected as the main causes of SGLT2 inhibitors' renoprotective effect. The effects of SGLT2 inhibitors on tubular damage in non-albuminuric diabetic patients are unclear. METHODS The SGLT2 inhibitor tofogliflozin (20 mg, 1 × /day) was orally administered to 14 non-albuminuric diabetic patients. Serum and urine samples were collected at baseline (before) and after the start of tofogliflozin treatment. Hemoglobin A1c, hemoglobin, estimated glomerular filtration rate (eGFR), body weight, and blood pressure (BP) were analyzed as clinical parameters at baseline and 1, 3, and 6 months later. Urinary neutrophil gelatinase-associated lipocalin (NGAL) and N-acetyl-β-D-glucosaminidase were measured as tubular damage markers and the urinary 8-hidroxydeoxyguanosine (8-OHdG) values were measured as an oxidative stress marker at baseline and at 1 and 3 months. RESULTS Compared to baseline, the patients' HbA1c values and body weights were significantly decreased post-tofogliflozin administration, and their eGFR values were decreased at 3 months but recovered at 6 months; the hemoglobin concentrations were significantly increased at 3 and 6 months and the urinary NGAL level tended to be decreased at 3 months. No significant changes in blood urea nitrogen, BP, NAG, urine sodium concentration, or urinary 8-OHdG values occurred. The effect of this SGLT2 inhibitor was not influenced by the use of an angiotensin receptor blocker or dipeptidyl-peptidase 4 inhibitor. CONCLUSION For individuals with non-albuminuric diabetes, tofogliflozin has a good glucose-lowering effect and might have a tubular-protective effect.
Collapse
|
12
|
Xu Y, Fu EL, Clase CM, Mazhar F, Jardine MJ, Carrero JJ. GLP-1 receptor agonist versus DPP-4 inhibitor and kidney and cardiovascular outcomes in clinical practice in type-2 diabetes. Kidney Int 2021; 101:360-368. [PMID: 34826514 DOI: 10.1016/j.kint.2021.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
Whether glucagon-like peptide-1 receptor agonists (GLP1-RA) reduce detrimental kidney outcomes is uncertain. In secondary analyses, trials have shown consistent reductions in macroalbuminuria, but inconclusive results about kidney function decline. To help clarify this, we conducted a cohort study to compare kidney and cardiovascular outcomes in individuals who started GLP1-RA or dipeptidyl peptidase-4 inhibitors (DPP4i) (reduces degradation of endogenous GLP1). The primary outcome was a composite of sustained doubling of creatinine, kidney failure or kidney death. The secondary outcomes were three-point major adverse cardiovascular events (MACE) and its individual components. Propensity score weighted Cox regression was used to balance 53 confounders. A total of 19,766 individuals were included, of whom 5,699 initiated GLP1-RA, and were followed for a median 2.9 years. Mean age was 63 years, 26.2% had atherosclerotic cardiovascular disease and 16.0% had an estimated glomerular filtration rate (eGFR) under 60 ml/min/1.73m2. The adjusted hazard ratio for GLP1-RA vs. DPP4i was 0.72 (95% confidence interval 0.53-0.98) for the composite kidney outcome and 0.85 (0.73-0.99) for MACE, with absolute five-year risk reductions of 0.8% (0.1%-1.5%) and 1.6% (0.2%-2.9%), respectively. Hazard ratios were 0.79 (0.60-1.05) for cardiovascular death, 0.86 (0.68-1.09) for myocardial infarction and 0.74 (0.59-0.93) for stroke. Results were consistent within subgroups, including age, sex, eGFR and baseline metformin use. Thus, in our analysis of patients from routine clinical practice, the use of GLP1-RA was associated with a lower risk of kidney outcomes compared with DPP4i. Reductions in both kidney outcomes and MACE were similar in magnitude to those reported in large cardiovascular outcome trials.
Collapse
Affiliation(s)
- Yang Xu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Edouard L Fu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catherine M Clase
- Department of Medicine and Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Faizan Mazhar
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Meg J Jardine
- Kidney Health Research, NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Juan J Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
GLP-1 Receptor Agonists in Diabetic Kidney Disease: From Physiology to Clinical Outcomes. J Clin Med 2021; 10:jcm10173955. [PMID: 34501404 PMCID: PMC8432108 DOI: 10.3390/jcm10173955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/28/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common complications in type 2 diabetes mellitus (T2D) and a major cause of morbidity and mortality in diabetes. Despite the widespread use of nephroprotective treatment of T2D, the incidence of DKD is increasing, and it is expected to become the fifth cause of death worldwide within 20 years. Previous studies have demonstrated that GLP-1 receptor agonists (GLP-1 RA) have improved macrovascular and microvascular outcomes independent of glycemic differences, including DKD. GLP-1Ras’ improvement on kidney physiology is mediated by natriuresis, reduction in hyperfiltration and renin-angiotensin-aldosterone system (RAAS) activity and anti-inflammatory properties. These findings translate into improved clinical outcomes such as an enhanced urine albumin-to-creatinine ratio (UACR) and a reduction in renal impairment and the need for renal replacement therapies (RRT). In this article, we review the role of GLP-1RAs on the mechanisms and effect in DKD and their clinical efficacy.
Collapse
|
14
|
Tommerdahl KL, Nadeau KJ, Bjornstad P. Mechanisms of Cardiorenal Protection of Glucagon-Like Peptide-1 Receptor Agonists. Adv Chronic Kidney Dis 2021; 28:337-346. [PMID: 34922690 DOI: 10.1053/j.ackd.2021.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 05/03/2021] [Accepted: 06/01/2021] [Indexed: 11/11/2022]
Abstract
The worldwide prevalence of type 2 diabetes (T2D) is steadily increasing, and it remains a challenging public health problem for populations in both developing and developed countries around the world. Despite the recent advances in novel antidiabetic agents, diabetic kidney disease and cardiovascular disease remain the leading causes of morbidity and mortality in T2D. Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs), incretin hormones that stimulate postprandial insulin secretion, serve as a promising avenue for treatment of T2D as they result in a variety of antihyperglycemic effects including increased endogenous insulin secretion, decreased gluconeogenesis, inhibition of pancreatic α-cell glucagon production, decreased pancreatic β-cell apoptosis, and increased β-cell proliferation. GLP-1RAs have also been found to delay gastric emptying, promote weight loss, increase satiety, decrease hypertension, improve dyslipidemia, reduce inflammation, improve albuminuria, induce natriuresis, improve cardiovascular function, and prevent thrombogenesis. In this review, we will present risk factors for the development of cardiac and kidney disease in individuals with T2D and discuss possible mechanisms for the cardiorenal protective effects seen with GLP-1RAs. We will also present the possibility of dual- and tri-receptor agonist therapies with GLP-1, gastric inhibitory peptide, and glucagon RAs as an area of possible mechanistic synergy in the treatment of T2D and the prevention of cardiorenal complications.
Collapse
|
15
|
Bouman EJ, Smits MM, van Bommel EJ, Muskiet MHA, Hesp AC, Serné EH, Joles JA, van Raalte DH. Skin microvascular function and renal hemodynamics in overweight patients with type 2 diabetes: A cross-sectional study. Microcirculation 2021; 28:e12700. [PMID: 33864418 PMCID: PMC8459253 DOI: 10.1111/micc.12700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Diabetic kidney disease is a microvascular complication of diabetes. Here, we assessed the association between skin microvascular function and renal hemodynamic function in a cohort of well-phenotyped adults with type 2 diabetes (T2D). METHODS We included 81 overweight/obese adults (age: 62 ± 8 years; BMI: 32 ± 4 kg/m2 ) with well-controlled T2D and no renal impairment. Skin microvascular function was assessed by nailfold capillary density in rest and after arterial occlusion (ie, peak capillary density). Renal hemodynamic functions (ie, measured glomerular filtration rate [mGFR], effective renal blood flow [ERBF], filtration fraction [FF], and effective renal vascular resistance [ERVR]) were assessed by combined inulin and para-aminohippurate clearances and blood pressure measurements. RESULTS Skin capillary density was 45 ± 10 capillaries/mm2 at baseline and 57 ± 11 capillaries/mm2 during post-occlusive peak; mGFR averaged 108 ± 20 ml/min. In multivariable regression analyses, positive associations between capillary density during post-occlusive peak and mGFR (β = 0.224; p = 0.022) and ERBF (β = 0.203; p = 0.020) and a positive trend for hyperemia and mGFR (β = 0.391; p = 0.053) were observed, while a negative association for post-occlusive capillary density with ERVR (β = -0.196; p = 0.027) was found. CONCLUSION These findings indicate that microvascular dysfunction in overweight adults with T2D is associated with lower mGFR and ERPF and higher ERVR. We hypothesize that increased renal vascular resistance may contribute to glomerular dysfunction due to impaired renal perfusion.
Collapse
Affiliation(s)
- Emma J Bouman
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Erik J van Bommel
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Anne C Hesp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Erik H Serné
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Scholtes RA, van Baar MJB, Kok MD, Bjornstad P, Cherney DZI, Joles JA, van Raalte DH. Renal haemodynamic and protective effects of renoactive drugs in type 2 diabetes: Interaction with SGLT2 inhibitors. Nephrology (Carlton) 2021; 26:377-390. [PMID: 33283420 PMCID: PMC8026736 DOI: 10.1111/nep.13839] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022]
Abstract
Diabetic kidney disease remains the leading cause of end-stage kidney disease and a major risk factor for cardiovascular disease. Large cardiovascular outcome trials and dedicated kidney trials have shown that sodium-glucose cotransporter (SGLT)2 inhibitors reduce cardiovascular morbidity and mortality and attenuate hard renal outcomes in patients with type 2 diabetes (T2D). Underlying mechanisms explaining these renal benefits may be mediated by decreased glomerular hypertension, possibly by vasodilation of the post-glomerular arteriole. People with T2D often receive several different drugs, some of which could also impact the renal vasculature, and could therefore modify both renal efficacy and safety of SGLT2 inhibition. The most commonly prescribed drugs that could interact with SGLT2 inhibitors on renal haemodynamic function include renin-angiotensin system inhibitors, calcium channel blockers and diuretics. Herein, we review the effects of these drugs on renal haemodynamic function in people with T2D and focus on studies that measured glomerular filtration rate (GFR) and effective renal plasma flow (ERPF) with gold-standard techniques. In addition, we posit, based on these observations, potential interactions with SGLT2 inhibitors with an emphasis on efficacy and safety.
Collapse
Affiliation(s)
- Rosalie A. Scholtes
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Michaël J. B. van Baar
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Megan D. Kok
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Petter Bjornstad
- Department of Pediatrics, Division of EndocrinologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of Medicine, Division of NephrologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - David Z. I. Cherney
- Department of Medicine and Department of Physiology, Division of Nephrology, University Health NetworkUniversity of TorontoTorontoOntarioCanada
| | - Jaap A. Joles
- Department of Nephrology and HypertensionUniversity Medical CenterUtrechtThe Netherlands
| | - Daniël H. van Raalte
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
- Department of Vascular Medicine, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
17
|
van Velzen DM, Smits MM, van Bommel EJM, Muskiet MHA, Tonneijck L, Kramer MHH, Joles JA, den Heijer M, Nokoff N, Bjornstad P, van Raalte DH. Kidney hemodynamic function in men and postmenopausal women with type 2 diabetes and preserved kidney function. Am J Physiol Renal Physiol 2021; 320:F1152-F1158. [PMID: 33900855 DOI: 10.1152/ajprenal.00660.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The progression of kidney disease may differ between sexes in type 2 diabetes (T2D), with previous studies reporting a slower decline in women. Glomerular hyperfiltration is a key factor driving the kidney function decline. The current study aimed to investigate the differences in kidney hemodynamic function between men and women with T2D. A cross-sectional analysis of pooled data from three studies compared kidney hemodynamic function between men and postmenopausal women with T2D without overt nephropathy. The outcome measures were glomerular filtration rate (GFR; inulin clearance), effective renal plasma flow (ERPF; p-aminohippurate clearance), filtration fraction (GFR/ERPF), and renal vascular resistance (RVR; mean arterial pressure/renal blood flow). Glomerular hydraulic pressure (PGLO) as well as afferent and efferent vascular resistance were estimated by Gomez formulae. Sex differences were assessed with linear regression models adjusted for systolic blood pressure, glucose, use of renin-angiotensin system blockers, and body mass index. In total, 101 men [age: 63 (58-68) yr, body mass index: 31.5 ± 3.9 kg/m2, GFR: 111 ± 18 mL/min, HbA1c: 7.4 ± 0.7%] and 27 women [age: 66 (62-69) yr, body mass index: 30.9 ± 4.5 kg/m2, GFR: 97 ± 11 mL/min, HbA1c: 7.1 ± 0.5%] were included. GFR was higher in men versus women [11.0 mL/min (95% confidence interval: 3.6, 18.4)]. Although statistically nonsignificant, PGLO trended higher in men [1.9 mmHg (95% confidence interval: -0.1, 4.0)], whereas RVR [-0.012 mmHg/L/min (95% confidence interval: -0.022, -0.002)] and afferent vascular resistance were lower [-361 dyn/s/cm5 (95% confidence interval: -801, 78)]. In conclusion, in adults without overt nephropathy, GFR was higher in men compared with women. PGLO also trended to be higher in men. Both findings are possibly related to afferent vasodilation and suggest greater prevalence of hyperfiltration. This could contribute to accelerated GFR loss over time in men with T2D.NEW & NOTEWORTHY In adults with type 2 diabetes, men had higher markers of hyperfiltration, which could potentially explain the accelerated progression of diabetic kidney disease in men compared with women.
Collapse
Affiliation(s)
- Daan M van Velzen
- Section of Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Erik J M van Bommel
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Mark H H Kramer
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Martin den Heijer
- Section of Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Natalie Nokoff
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado.,Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniël H van Raalte
- Section of Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands.,Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Dong W, Wan EYF, Fong DYT, Kwok RLP, Chao DVK, Tan KCB, Hui EMT, Tsui WWS, Chan KH, Fung CSC, Lam CLK. Prediction models and nomograms for 10-year risk of end-stage renal disease in Chinese type 2 diabetes mellitus patients in primary care. Diabetes Obes Metab 2021; 23:897-909. [PMID: 33319467 DOI: 10.1111/dom.14292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/28/2020] [Accepted: 12/07/2020] [Indexed: 12/30/2022]
Abstract
AIMS To develop and validate 10-year risk prediction models, nomograms and charts for end-stage renal disease (ESRD) in Chinese patients with type 2 diabetes mellitus (T2DM) in primary care, in order to guide individualized treatment. MATERIALS AND METHODS This was a 10-year population-based observational cohort study. A total of 141 516 Chinese T2DM patients without history of cardiovascular disease or ESRD who were managed in public primary care clinics in 2008 were included and followed up until December 2017. Two-thirds of these patients were randomly selected to develop sex-specific ESRD risk prediction models using Cox regressions. The validity and accuracy of the models were tested on the remaining third of patients using Harrell's C-index. We selected variables based on their clinical and statistical importance to construct the nomograms and charts. RESULTS The median follow-up period was 9.75 years. The cumulative incidence of ESRD was 6.0% (men: 6.1%, women: 5.9%). Age, diabetes duration, systolic blood pressure (SBP), SBP variability, diastolic blood pressure, triglycerides, glycated haemoglobin (HbA1c), HbA1c variability, urine albumin to creatinine ratio (UACR), and estimated glomerular filtration rate (eGFR) were significant predictors for both sexes. Smoking and total cholesterol to HDL cholesterol ratio were additional significant predictors for men and women, respectively. The models showed Harrell's C-statistics of 0.889/0.889 (women/men). Age, eGFR, UACR, SBP and HbA1c were selected for both sexes to develop nomograms and charts. CONCLUSIONS Using routinely available variables, the 10-year ESRD risk of Chinese T2DM patients in primary care can be predicted with approximately 90% accuracy. We have developed different tools to facilitate routine ESRD risk prediction in primary care, so that individualized care can be provided to prevent or delay ESRD in T2DM patients.
Collapse
Affiliation(s)
- Weinan Dong
- Department of Family Medicine and Primary Care, University of Hong Kong, Hong Kong
| | - Eric Yuk Fai Wan
- Department of Family Medicine and Primary Care, University of Hong Kong, Hong Kong
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong
| | | | - Ruby Lai Ping Kwok
- Department of Primary and Community Services, Hospital Authority, Hong Kong
| | - David Vai Kiong Chao
- Department of Family Medicine and Primary Health Care, Kowloon East Cluster, Hospital Authority, Hong Kong
| | | | - Eric Ming Tung Hui
- Department of Family Medicine, New Territories East Cluster, Hospital Authority, Hong Kong
| | - Wendy Wing Sze Tsui
- Family Medicine and Primary Healthcare, QMH, Hong Kong West Cluster, Hospital Authority, Hong Kong
| | | | | | - Cindy Lo Kuen Lam
- Department of Family Medicine and Primary Care, University of Hong Kong, Hong Kong
| |
Collapse
|
19
|
Fang M, Selvin E. Thirty-Year Trends in Complications in U.S. Adults With Newly Diagnosed Type 2 Diabetes. Diabetes Care 2021; 44:699-706. [PMID: 33419932 PMCID: PMC7896270 DOI: 10.2337/dc20-2304] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/28/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the prevalence of and trends in complications among U.S. adults with newly diagnosed diabetes. RESEARCH DESIGN AND METHODS We included 1,486 nonpregnant adults (aged ≥20 years) with newly diagnosed diabetes (diagnosed within the past 2 years) from the 1988-1994 and 1999-2018 National Health and Nutrition Examination Survey. We estimated trends in albuminuria (albumin-to-creatinine ratio ≥30 mg/g), reduced estimated glomerular filtration rate (eGFR <60 mL/min/1.73 m2), retinopathy (any retinal microaneurysms or blot hemorrhages), and self-reported cardiovascular disease (history of congestive heart failure, heart attack, or stroke). RESULTS From 1988-1994 to 2011-2018, there was a significant decrease in the prevalence of albuminuria (38.9 to 18.7%, P for trend <0.001) but no change in the prevalence of reduced eGFR (7.5 to 9.9%, P for trend = 0.30), retinopathy (1988-1994 to 1999-2008 only; 13.2 to 12.1%, P for trend = 0.86), or self-reported cardiovascular disease (19.0 to 16.5%, P for trend = 0.64). There were improvements in glycemic, blood pressure, and lipid control in the population, and these partially explained the decline in albuminuria. Complications were more common at the time of diabetes diagnosis for adults who were older, lower income, less educated, and obese. CONCLUSIONS Over the past three decades, there have been encouraging reductions in albuminuria and risk factor control in adults with newly diagnosed diabetes. However, the overall burden of complications around the time of the diagnosis remains high.
Collapse
Affiliation(s)
- Michael Fang
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
20
|
Skov-Jeppesen SM, Yderstraede KB, Bistrup C, Jensen BL, Marcussen N, Hanna M, Lund L. Low-intensity shockwave therapy in the treatment of diabetic nephropathy: a prospective Phase 1 study. Nephrol Dial Transplant 2020; 35:1385-1392. [PMID: 30590575 DOI: 10.1093/ndt/gfy375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/09/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Low-intensity shockwave therapy (LI-SWT) is suggested as a therapy for promoting tissue regeneration. In pigs, it was recently found that LI-SWT improved renal function after ischaemic injury. Our objectives were to study glomerular filtration rate (GFR) and albuminuria in diabetic nephropathy (DN) after treatment with LI-SWT. The present pilot study reports on the clinical safety of LI-SWT in DN. METHODS A total of 14 patients with diabetes mellitus and Stage 3 chronic kidney disease were recruited for this prospective, one-arm Phase 1 study. The patients were treated with six sessions of LI-SWT during a 3-week period. At each session, 3000 shockwaves were applied to each kidney with 0.265 mJ/mm2, extended focal size and 4 Hz. Follow-up visits were performed at 1, 3 and 6 months. RESULTS In general, the treatment was well tolerated. Transient macroscopic haematuria was observed in three patients immediately after LI-SWT. The majority of patients experienced lower back tenderness lasting up to 2 days after treatment. There was no need for analgesic treatment. LI-SWT showed no negative effect on GFR and albuminuria. At baseline, median (interquartile range) GFR was 33.5 mL/min/1.73 m2 (27.8-43.8) compared with 36.0 mL/min/1.73 m2 (27.5-52.0) at 6 months follow-up. In parallel, median albuminuria was 256 mg/24 h (79-619) at baseline and tended to decrease to 137 mg/24 h (41-404) 6 months after LI-SWT. There was no statistical difference between baseline and follow-up results. CONCLUSIONS LI-SWT is a safe treatment for DN. Inclusion of more patients is needed to determine whether LI-SWT can improve renal functional outcomes.
Collapse
Affiliation(s)
- Sune Moeller Skov-Jeppesen
- Department of Urology, Odense University Hospital, Odense, Denmark.,OPEN, Odense Patient data Explorative Network, Odense University Hospital, Odense, Denmark.,Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Knud Bonnet Yderstraede
- Clinical Institute, University of Southern Denmark, Odense, Denmark.,Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Claus Bistrup
- Clinical Institute, University of Southern Denmark, Odense, Denmark.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Boyle L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Niels Marcussen
- Clinical Institute, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Milad Hanna
- Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark.,Clinical Institute, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
21
|
Wheeler DC, James J, Patel D, Viljoen A, Ali A, Evans M, Fernando K, Hicks D, Milne N, Newland-Jones P, Wilding J. SGLT2 Inhibitors: Slowing of Chronic Kidney Disease Progression in Type 2 Diabetes. Diabetes Ther 2020; 11:2757-2774. [PMID: 32996085 PMCID: PMC7524028 DOI: 10.1007/s13300-020-00930-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is a topic of increasing concern among clinicians involved in the management of type 2 diabetes mellitus (T2DM). It is a progressive and costly complication associated with increased risk of adverse cardiovascular (CV) and renal outcomes and mortality. Ongoing monitoring of the estimated glomerular filtration (eGFR) rate alongside the urine albumin:creatinine ratio (ACR) is recommended during regular T2DM reviews to enable a prompt DKD diagnosis or to assess disease progression, providing an understanding of adverse risk for each individual. Many people with DKD will progress to end-stage kidney disease (ESKD), requiring renal replacement therapy (RRT), typically haemodialysis or kidney transplantation. A range of lifestyle and pharmacological interventions is recommended to help lower CV risk, slow the advancement of DKD and prevent or delay the need for RRT. Emerging evidence concerning sodium-glucose co-transporter-2 inhibitor (SGLT2i) agents suggests a role for these medicines in slowing eGFR decline, enabling regression of albuminuria and reducing progression to ESKD. Improvements in renal end points observed in SGLT2i CV outcome trials (CVOTs) highlighted the possible impact of these agents in the management of DKD. Data from the canagliflozin CREDENCE trial (Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation) have since demonstrated the effectiveness of this medicine in reducing the risk of kidney failure and CV events in a population comprising individuals with T2DM and renal disease. CREDENCE was the first SGLT2i study to examine renal outcomes as the primary end point. Real-world studies have reaffirmed these outcomes in routine clinical practice. This article summarises the evidence regarding the use of SGLT2i medicines in slowing the progression of DKD and examines the possible mechanisms underpinning the renoprotective effects of these agents. The relevant national and international guidance for monitoring and treatment of DKD is also highlighted to help clinicians working to support this vulnerable group.
Collapse
Affiliation(s)
| | - June James
- University Hospitals of Leicester NHS Trust, University of Leicester, Leicester, UK
| | | | | | - Amar Ali
- Oakenhurst Medical Practice, Blackburn, UK
| | - Marc Evans
- University Hospital Llandough, Penarth, UK
| | | | | | - Nicola Milne
- Manchester University NHS Foundation Trust, Manchester, UK
| | | | | |
Collapse
|
22
|
Azimi F, Ghasemi JB, Azizian H, Najafi M, Faramarzi MA, Saghaei L, Sadeghi-Aliabadi H, Larijani B, Hassanzadeh F, Mahdavi M. Design and synthesis of novel pyrazole-phenyl semicarbazone derivatives as potential α-glucosidase inhibitor: Kinetics and molecular dynamics simulation study. Int J Biol Macromol 2020; 166:1082-1095. [PMID: 33157144 DOI: 10.1016/j.ijbiomac.2020.10.263] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 01/17/2023]
Abstract
A series of novel pyrazole-phenyl semicarbazone derivatives were designed, synthesized, and screened for in vitro α-glucosidase inhibitory activity. Given the importance of hydrogen bonding in promoting the α-glucosidase inhibitory activity, pharmacophore modification was established. The docking results rationalized the idea of the design. All newly synthesized compounds exhibited excellent in vitro yeast α-glucosidase inhibition (IC50 values in the range of 65.1-695.0 μM) even much more potent than standard drug acarbose (IC50 = 750.0 μM). Among them, compounds 8o displayed the most potent α-glucosidase inhibitory activity (IC50 = 65.1 ± 0.3 μM). Kinetic study of compound 8o revealed that it inhibited α-glucosidase in a competitive mode (Ki = 87.0 μM). Limited SAR suggested that electronic properties of substitutions have little effect on inhibitory potential of compounds. Cytotoxic studies demonstrated that the active compounds (8o, 8k, 8p, 8l, 8i, and 8a) compounds are also non-cytotoxic. The binding modes of the most potent compounds 8o, 8k, 8p, 8l and 8i was studied through in silico docking studies. Molecular dynamic simulations have been performed in order to explain the dynamic behavior and structural changes of the systems by the calculation of the root mean square deviation (RMSD) and root mean square fluctuation (RMSF).
Collapse
Affiliation(s)
- Fateme Azimi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461, Isfahan, Iran
| | - Jahan B Ghasemi
- School of Chemistry, University College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Science, Tehran, Iran
| | - Mohammad Najafi
- Department of Chemistry, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
| | - Lotfollah Saghaei
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461, Isfahan, Iran.
| | - Hojjat Sadeghi-Aliabadi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461, Isfahan, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Hezar Jerib, 817416-73461, Isfahan, Iran.
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Mosterd CM, Bjornstad P, van Raalte DH. Nephroprotective effects of GLP-1 receptor agonists: where do we stand? J Nephrol 2020; 33:965-975. [PMID: 32356231 PMCID: PMC7560915 DOI: 10.1007/s40620-020-00738-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/11/2020] [Indexed: 12/12/2022]
Abstract
Glucagon-like peptide (GLP)-1 receptor agonists are the cornerstone in the treatment of hyperglycemia in many people suffering from type 2 diabetes (T2D). These drugs have potent glucose-lowering actions and, additionally, lower body weight through satiety induction while reducing blood pressure and dyslipidemia. Partly through these actions, GLP-1 receptor agonism was shown to reduce cardiovascular disease (CVD) in people with T2D with previous CVD or at high-risk thereof. In these cardiovascular safety trials, in secondary or exploratory analyses, GLP-1 receptor agonists were also shown to reduce macro-albuminuria, an accepted surrogate marker for diabetic kidney disease (DKD), a condition that still represents a major unmet medical need. In this review we will discuss the evidence which suggests renoprotection induced by GLP-1 receptor agonists and the potential mechanisms that may be involved. These include mitigation of hyperglycemia, overweight and insulin resistance, systemic and glomerular hypertension, dyslipidemia, sodium retention, inflammation and renal hypoxia. The recently initiated large-sized FLOW trial investigating the effects of semaglutide on hard renal outcomes in patients with DKD will provide clarity whether GLP-1 receptor agonists may reduce the burden of DKD in addition to their other beneficial metabolic and cardiovascular effects.
Collapse
Affiliation(s)
- Charlotte M Mosterd
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics and Division of Nephrology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Piperidou A, Loutradis C, Sarafidis P. SGLT-2 inhibitors and nephroprotection: current evidence and future perspectives. J Hum Hypertens 2020; 35:12-25. [PMID: 32778748 DOI: 10.1038/s41371-020-00393-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is a major public health issue and an independent risk factor for cardiovascular and all-cause mortality. Diabetic kidney disease develops in 30-50% of diabetic patients and it is the leading cause of end-stage renal disease in the Western world. Strict blood pressure control and renin-angiotensin system (RAS) blocker use are the cornerstones of CKD treatment; however, their application in everyday clinical practice is not always ideal and in many patients CKD progression still occurs. Accumulated evidence in the past few years clearly suggests that sodium-glucose co-transporter-2 (SGLT-2) inhibitors present potent nephroprotective properties. In clinical trials in patients with type 2 diabetes mellitus, these agents were shown to reduce albuminuria and proteinuria by 30-50% and the incidence of composite hard renal outcomes by 40-50%. Furthermore, their mechanism of action appears rather solid, as they interfere with the major mechanism of proteinuric CKD progression, i.e., glomerular hypertension and hyperfiltration. The present review summarizes the current evidence from human trials on the effects of SGLT-2 inhibitors on nephroprotection and discusses their position in everyday clinical practice.
Collapse
Affiliation(s)
- Alexia Piperidou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charalampos Loutradis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
25
|
Pasternak B, Wintzell V, Eliasson B, Svensson AM, Franzén S, Gudbjörnsdottir S, Hveem K, Jonasson C, Melbye M, Svanström H, Ueda P. Use of Glucagon-Like Peptide 1 Receptor Agonists and Risk of Serious Renal Events: Scandinavian Cohort Study. Diabetes Care 2020; 43:1326-1335. [PMID: 32295809 DOI: 10.2337/dc19-2088] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/20/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the association between use of glucagon-like peptide 1 (GLP-1) receptor agonists and risk of serious renal events in routine clinical practice. RESEARCH DESIGN AND METHODS This was a cohort study using an active-comparator, new-user design and nationwide register data from Sweden, Denmark, and Norway during 2010-2016. The cohort included 38,731 new users of GLP-1 receptor agonists (liraglutide 92.5%, exenatide 6.2%, lixisenatide 0.7%, and dulaglutide 0.6%), matched 1:1 on age, sex, and propensity score to a new user of the active comparator, dipeptidyl peptidase 4 (DPP-4) inhibitors. The main outcome was serious renal events, a composite including renal replacement therapy, death from renal causes, and hospitalization for renal events. Secondary outcomes were the individual components of the main outcome. Hazard ratios (HRs) were estimated using Cox models and an intention-to-treat exposure definition. Mean (SD) follow-up time was 3.0 (1.7) years. RESULTS Mean (SD) age of the study population was 59 (10) years, and 18% had cardiovascular disease. A serious renal event occurred in 570 users of GLP-1 receptor agonists (incidence rate 4.8 events per 1,000 person-years) and in 722 users of DPP-4 inhibitors (6.3 events per 1,000 person-years, HR 0.76 [95% CI 0.68-0.85], absolute difference -1.5 events per 1,000 person-years [-2.1 to -0.9]). Use of GLP-1 receptor agonists was associated with a significantly lower risk of renal replacement therapy (HR 0.73 [0.62-0.87]) and hospitalization for renal events (HR 0.73 [0.65-0.83]) but not death from renal causes (HR 0.72 [0.48-1.10]). When we used an as-treated exposure definition in which patients were censored at treatment cessation or switch to the other study drug, the HR for the primary outcome was 0.60 (0.49-0.74). CONCLUSIONS In this large cohort of patients seen in routine clinical practice in three countries, use of GLP-1 receptor agonists, as compared with DPP-4 inhibitors, was associated with a reduced risk of serious renal events.
Collapse
Affiliation(s)
- Björn Pasternak
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Viktor Wintzell
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Björn Eliasson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ann-Marie Svensson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden
| | - Stefan Franzén
- Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden.,Health Metrics, Department of Public Health and Community Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Soffia Gudbjörnsdottir
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Swedish National Diabetes Register, Västra Götalandsregionen, Gothenburg, Sweden
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Science, Norwegian University of Science and Technology, Trondheim, Norway.,HUNT Research Center, Faculty of Medicine, Norwegian University of Science and Technology, Levanger, Norway
| | - Christian Jonasson
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Science, Norwegian University of Science and Technology, Trondheim, Norway.,HUNT Research Center, Faculty of Medicine, Norwegian University of Science and Technology, Levanger, Norway
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.,Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Henrik Svanström
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Ueda
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Idzerda NMA, Clegg LE, Hernandez AF, Bakris G, Penland RC, Boulton DW, Bethel MA, Holman RR, Heerspink HJL. Prediction and validation of exenatide risk marker effects on progression of renal disease: Insights from EXSCEL. Diabetes Obes Metab 2020; 22:798-806. [PMID: 31912603 PMCID: PMC7187441 DOI: 10.1111/dom.13958] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/23/2022]
Abstract
AIM To assess whether the previously developed multivariable risk prediction framework (PRE score) could predict the renal effects observed in the EXSCEL cardiovascular outcomes trial using short-term changes in cardio-renal risk markers. MATERIALS AND METHODS Changes from baseline to 6 months in HbA1c, systolic blood pressure (SBP), body mass index (BMI), haemoglobin, total cholesterol, and new micro- or macroalbuminuria were evaluated. The renal outcomes were defined as a composite of a sustained 30% or 40% decline in estimated glomerular filtration rate (eGFR) or end-stage renal disease (ESRD). Relationships between risk markers and long-term renal outcomes were determined in patients with type 2 diabetes from the ALTITUDE study using multivariable Cox regression analysis, and then applied to short-term changes in risk markers observed in EXSCEL to predict the exenatide-induced impact on renal outcomes. RESULTS Compared with placebo, mean HbA1c, BMI, SBP and total cholesterol were lower at 6 months with exenatide, as was the incidence of new microalbuminuria. The PRE score predicted a relative risk reduction for the 30% eGFR decline + ESRD endpoint of 11.3% (HR 0.89; 95% CI 0.83-0.94), compared with 12.7% (HR 0.87; 0.77-0.99) observed risk reduction. For the 40% eGFR decline + ESRD endpoint, the predicted and observed risk reductions were 11.0% (HR 0.89; 0.82-0.97) and 13.7% (HR 0.86, 0.72-1.04), respectively. CONCLUSIONS Integrating short-term risk marker changes into a multivariable risk score predicted the magnitude of renal risk reduction observed in EXSCEL.
Collapse
Affiliation(s)
- Nienke M. A. Idzerda
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Lindsay E. Clegg
- Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaGaithersburgMarylandUnited States
| | - Adrian F. Hernandez
- Duke Clinical Research Institute, Duke University School of MedicineDurhamNorth CarolinaUnited States
| | - George Bakris
- University of Chicago MedicineChicagoIllinoisUnited States
| | - Robert C. Penland
- Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaWalthamBoston, MassachusettsUnited States
| | - David W. Boulton
- Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaGaithersburgMarylandUnited States
| | - M. Angelyn Bethel
- Diabetes Trials Unit, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Rury R. Holman
- Diabetes Trials Unit, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | | |
Collapse
|
27
|
Sarafidis P, Ferro CJ, Morales E, Ortiz A, Malyszko J, Hojs R, Khazim K, Ekart R, Valdivielso J, Fouque D, London GM, Massy Z, Ruggenenti P, Porrini E, Wiecek A, Zoccali C, Mallamaci F, Hornum M. SGLT-2 inhibitors and GLP-1 receptor agonists for nephroprotection and cardioprotection in patients with diabetes mellitus and chronic kidney disease. A consensus statement by the EURECA-m and the DIABESITY working groups of the ERA-EDTA. Nephrol Dial Transplant 2020; 34:208-230. [PMID: 30753708 DOI: 10.1093/ndt/gfy407] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) in patients with diabetes mellitus (DM) is a major problem of public health. Currently, many of these patients experience progression of cardiovascular and renal disease, even when receiving optimal treatment. In previous years, several new drug classes for the treatment of type 2 DM have emerged, including inhibitors of renal sodium-glucose co-transporter-2 (SGLT-2) and glucagon-like peptide-1 (GLP-1) receptor agonists. Apart from reducing glycaemia, these classes were reported to have other beneficial effects for the cardiovascular and renal systems, such as weight loss and blood pressure reduction. Most importantly, in contrast to all previous studies with anti-diabetic agents, a series of recent randomized, placebo-controlled outcome trials showed that SGLT-2 inhibitors and GLP-1 receptor agonists are able to reduce cardiovascular events and all-cause mortality, as well as progression of renal disease, in patients with type 2 DM. This document presents in detail the available evidence on the cardioprotective and nephroprotective effects of SGLT-2 inhibitors and GLP-1 analogues, analyses the potential mechanisms involved in these actions and discusses their place in the treatment of patients with CKD and DM.
Collapse
Affiliation(s)
- Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charles J Ferro
- Department of Renal Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Enrique Morales
- Department of Nephrology, Hospital Universitario 12 de Octubre and Research Institute i+12, Madrid, Spain
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and REDINREN, Madrid, Spain
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
| | - Radovan Hojs
- Department of Nephrology, University Medical Center and Faculty of Medicine, Maribor University, Maribor, Slovenia
| | - Khaled Khazim
- Department of Nephrology and Hypertension, Galilee Medical Center, Nahariya, Israel
| | - Robert Ekart
- Department of Nephrology, University Medical Center and Faculty of Medicine, Maribor University, Maribor, Slovenia
| | - Jose Valdivielso
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, IRBLleida, Lleida and RedInRen, ISCIII, Spain
| | - Denis Fouque
- Department of Nephrology, Centre Hospitalier Lyon Sud, University of Lyon, Lyon, France
| | | | - Ziad Massy
- Hopital Ambroise Paré, Paris Ile de France Ouest (UVSQ) University, Paris, France
| | - Petro Ruggenenti
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Nephrology and Dialysis Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Esteban Porrini
- Faculty of Medicine, University of La Laguna, Instituto de Tecnología Biomédicas (ITB) Hospital Universitario de Canarias, Tenerife, Canary Islands, Spain
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Carmine Zoccali
- CNR-IFC, Clinical Epidemiology and Pathophysiology of Hypertension and Renal Diseases Unit, Ospedali Riuniti, Reggio Calabria, Italy
| | - Francesca Mallamaci
- CNR-IFC, Clinical Epidemiology and Pathophysiology of Hypertension and Renal Diseases Unit, Ospedali Riuniti, Reggio Calabria, Italy
| | - Mads Hornum
- Department of Nephrology, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
28
|
Zhu Y, Ling Y, Wang X. Alantolactone mitigates renal injury induced by diabetes via inhibition of high glucose-mediated inflammatory response and macrophage infiltration. Immunopharmacol Immunotoxicol 2020; 42:84-92. [PMID: 32064988 DOI: 10.1080/08923973.2020.1725039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Inflammatory response plays a crucial role in the occurrence and development of diabetic nephropathy (DN). Drugs that carry anti-inflammatory effects have the potential to treat diabetic nephropathy. It's reported that alantolactone (ALA), a natural product, has a variety of pharmacological effects against inflammation and oxidation. However, the specific effects of alantolactone on DN and the mechanisms underlying alantolactone remain elusive. Therefore, the present study aimed to probe whether ALA could mitigate inflammation as mediated by high glucose (HG) in NRK-52E cells and reduce renal injury caused by diabetic nephropathy.Materials and methods: The anti-inflammatory effect of ALA was evaluated in the present study using ELISA and RT-qPCR. Western blot and macrophage adhesion assay were then performed to confirm anti-macrophage adhesion and the protein expression of cell adhesion molecules. Finally, the effect of ALA and its underlying mechanism was evaluated in vivo.Results: Results showed that ALA curbed HG-stimulated expression of macrophage adhesion and pro-inflammatory cytokines in renal NRK-52E cells. In addition, both pro-inflammatory cytokines and NF-kappa B witnessed reduced expression or activity in oral administration with ALA in vivo, thus inhibiting the increase of serum creatinine and urea nitrogen (BUN) levels. This in consequence ameliorated fibrosis and stemmed pathological worsening of diabetic renal tissues.Conclusions: These findings suggest that ALA may hold promise in the treatment of DN, and importantly, the anti-inflammatory system may prove to be a new strategy to treat human DN.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Traditional Chinese Medicine, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Reserch Center for Child Health, Hangzhou, China
| | - Yuanliang Ling
- Community Health Service Center of Xiangfu, Hangzhou, China
| | - Xiao'ai Wang
- Department of Traditional Chinese Medicine, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Reserch Center for Child Health, Hangzhou, China
| |
Collapse
|
29
|
Wang M, Li J, Li Y, Yao S, Zhao M, Wang C, Wu S, Xue H. The effects of hypertension and diabetes on new‐onset chronic kidney disease: A prospective cohort study. J Clin Hypertens (Greenwich) 2019; 22:39-46. [PMID: 31873983 DOI: 10.1111/jch.13768] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Miao Wang
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
- The School of Medicine Nankai University Tianjin China
| | - Junjuan Li
- Department of Nephrology Kailuan Hospital Hebei United University Tangshan China
| | - Yao Li
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
| | - Siyu Yao
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
| | - Maoxiang Zhao
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
| | - Chi Wang
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
| | - Shouling Wu
- Department of Cardiology Kailuan Hospital Hebei United University Tangshan China
| | - Hao Xue
- Department of Cardiology Chinese People's Liberation Army General Hospital Beijing China
| |
Collapse
|
30
|
van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, Bozovic A, Danser AHJ, Geurts F, Hoorn EJ, Touw DJ, Larsen EL, Poulsen HE, Kramer MHH, Nieuwdorp M, Joles JA, van Raalte DH. The renal hemodynamic effects of the SGLT2 inhibitor dapagliflozin are caused by post-glomerular vasodilatation rather than pre-glomerular vasoconstriction in metformin-treated patients with type 2 diabetes in the randomized, double-blind RED trial. Kidney Int 2019; 97:202-212. [PMID: 31791665 DOI: 10.1016/j.kint.2019.09.013] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve hard renal outcomes in type 2 diabetes. This is possibly explained by the fact that SGLT2i normalize the measured glomerular filtration rate (mGFR) by increasing renal vascular resistance, as was shown in young people with type 1 diabetes and glomerular hyperfiltration. Therefore, we compared the renal hemodynamic effects of dapagliflozin with gliclazide in type 2 diabetes. The mGFR and effective renal plasma flow were assessed using inulin and para-aminohippurate clearances in the fasted state, during clamped euglycemia (5 mmol/L) and during clamped hyperglycemia (15 mmol/L). Filtration fraction and renal vascular resistance were calculated. Additionally, factors known to modulate renal hemodynamics were measured. In 44 people with type 2 diabetes on metformin monotherapy (Hemoglobin A1c 7.4%, mGFR 113 mL/min), dapagliflozin versus gliclazide reduced mGFR by 5, 10, and 12 mL/min in the consecutive phases while both agents similarly improved Hemoglobin A1c (-0.48% vs -0.65%). Dapagliflozin also reduced filtration fraction without increasing renal vascular resistance, and increased urinary adenosine and prostaglandin concentrations. Gliclazide did not consistently alter renal hemodynamic parameters. Thus, beyond glucose control, SGLT2i reduce mGFR and filtration fraction in type 2 diabetes. The fact that renal vascular resistance was not increased by dapagliflozin suggests that this is due to post-glomerular vasodilation rather than pre-glomerular vasoconstriction.
Collapse
Affiliation(s)
- Erik J M van Bommel
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands.
| | - Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Michaël J B van Baar
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Anna L Emanuel
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Andrea Bozovic
- Department of Clinical Biochemistry, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank Geurts
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Emil L Larsen
- Department of Clinical Pharmacology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Henrik E Poulsen
- Department of Clinical Pharmacology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mark H H Kramer
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Suijk DL, Smits MM, Muskiet MH, Tonneijck L, Kramer MH, Joles JA, van Raalte DH. Plasma uric acid and renal haemodynamics in type 2 diabetes patients. Nephrology (Carlton) 2019; 25:290-297. [PMID: 31429150 PMCID: PMC7065078 DOI: 10.1111/nep.13645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2019] [Indexed: 11/30/2022]
Abstract
Aim Increased plasma uric acid (PUA) concentrations are associated with chronic kidney disease in type 2 diabetes (T2D) patients. The mechanisms involved remain unclear. We investigated the relation between PUA and (intra)renal haemodynamics in T2D patients without overt kidney disease. Methods Eighty‐eight white men and women with T2D were included (age 64 (58–68) years; body mass index 30.9 (28.3–33.6) kg/m2; glycated haemoglobin 7.1 (6.8–7.6)%). Plasma UA and fractional excretion of UA were measured, while glomerular filtration rate (GFR) and effective renal plasma flow (ERPF) were assessed by inulin and PAH‐clearance techniques, respectively. Effective renal vascular resistance was calculated (ERVR). Renal afferent and efferent arteriolar resistances and glomerular hydrostatic pressure were estimated. Relationships between PUA and fractional excretion of UA and (intra)renal haemodynamic parameters were evaluated by multivariable linear regression analyses. Results Plasma UA concentrations were at the higher end of the normal range in most participants: 342 ± 68 μmol/L or 5.7 ± 1.1 mg/dL (mean ± SD). In multivariable analyses, PUA concentrations were negatively associated with GFR (r = −0.471; P = 0.001), ERPF (r = −0.436; P = 0.003) and glomerular hydrostatic pressure (r = −0.427; P = 0.003). In contrast, PUA concentrations had a positive correlation with ERVR (r = 0.474; P = 0.001), but not with efferent vascular resistance. Fractional excretion of UA was not related to renal haemodynamics. Conclusion Plasma UA was negatively associated to GFR, ERPF but positively related to ERVR in T2D patients without overt renal impairment. Plasma UA‐related increase in ERVR may be related to increased arterial afferent tone, which may put the kidney at risk for renal damage through ischaemia. This paper, which explored the relationship between plasma uric acid (UA) and renal haemodynamics in patients with type 2 diabetes without overt kidney disease showed that higher plasma UA is associated with lower glomerular filtration rate and effective renal plasma flow but higher effective renal vascular resistance, which may contribute to glomerular dysfunction due to impairment in kidney perfusion.
Collapse
Affiliation(s)
- Danii Ls Suijk
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Marcel Ha Muskiet
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Mark Hh Kramer
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Muskiet MHA, Bunck MC, Heine RJ, Cornér A, Yki-Järvinen H, Eliasson B, Joles JA, Diamant M, Tonneijck L, van Raalte DH. Exenatide twice-daily does not affect renal function or albuminuria compared to titrated insulin glargine in patients with type 2 diabetes mellitus: A post-hoc analysis of a 52-week randomised trial. Diabetes Res Clin Pract 2019; 153:14-22. [PMID: 31078666 DOI: 10.1016/j.diabres.2019.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/23/2022]
Abstract
AIMS To compare the effects of long-term treatment with the GLP-1RA exenatide twice-daily versus titrated insulin glargine (iGlar) on renal function and albuminuria in type 2 diabetes (T2DM) patients. METHODS We post-hoc evaluated renal outcome-data of 54 overweight T2DM patients (mean ± SD age 60 ± 8 years, HbA1c 7.5 ± 0.9%, eGFR 86 ± 16 mL/min/1.73 m2, median [IQR] urinary albumin-to-creatinine-ratio (UACR) 0.75 [0.44-1.29] mg/mmol) randomised to exenatide 10 µg twice-daily or titrated iGlar on-top-of metformin for 52-weeks. Renal efficacy endpoints were change in creatinine clearance (CrCl) and albuminuria (urinary albumin-excretion [UAE] and UACR) based on 24-h urines, collected at baseline and Week-52. eGFR and exploratory endpoints were collected throughout the intervention-period, and after a 4-week wash-out. RESULTS HbA1c-reductions were similar with exenatide (mean ± SEM -0.80 ± 0.10%) and iGlar (-0.79 ± 0.14%; treatment-difference 0.02%; 95% CI -0.31 to 0.42%). Change from baseline to Week-52 in CrCl, UAE or UACR did not statistically differ; only iGlar reduced albuminuria (P < 0.05; within-group). eGFR decreased from baseline to Week-4 with exenatide (-3.9 ± 2.1 mL/min/1.73 m2; P = 0.069) and iGlar (-2.7 ± 1.2 mL/min/1.73 m2; P = 0.034), without treatment-differences in ensuing trajectory. Exenatide versus iGlar reduced bodyweight (-5.4 kg; 2.9-7.9; P < 0.001), but did not affect blood pressure, lipids or plasma uric acid. CONCLUSIONS Among T2DM patients without overt nephropathy, one-year treatment with exenatide twice-daily does not affect renal function-decline or onset/progression of albuminuria compared to titrated iGlar. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT00097500.
Collapse
Affiliation(s)
- M H A Muskiet
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands.
| | - M C Bunck
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands; Eli Lilly and Co., Indianapolis, IN, USA
| | - R J Heine
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands; Eli Lilly and Co., Indianapolis, IN, USA
| | - A Cornér
- Research Programs' Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - H Yki-Järvinen
- Research Programs' Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - B Eliasson
- Lundberg Laboratory for Diabetes Research, Sahlgrenska University Hospital, Göteborg, Sweden
| | - J A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, the Netherlands
| | - M Diamant
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| | - L Tonneijck
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| | - D H van Raalte
- Diabetes Centre, Amsterdam University Medical Centers, location VUMC, Amsterdam, the Netherlands
| |
Collapse
|
33
|
The effect of SGLT-2 inhibitors on blood pressure: a pleiotropic action favoring cardio- and nephroprotection. Future Med Chem 2019; 11:1285-1303. [PMID: 31161798 DOI: 10.4155/fmc-2018-0514] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Strict blood pressure (BP) control in patients with diabetes is associated with reductions in cardiovascular and renal risk. SGLT-2 inhibitors act in the proximal tubule to reduce glucose reabsorption. They also have mild natriuretic and diuretic effects, combining properties of proximal tubule diuretics and osmotic diuretics, which are expected to reduce BP. Several lines of evidence suggests that SGLT-2 inhibitors produce mild but meaningful reductions in BP and also decrease the incidence of renal outcomes, cardiovascular events and mortality. Thus, recent guidelines for type 2 diabetes suggest that among oral agents to use together with metformin, SGLT-2 inhibitors should be preferred in patients at increased cardiovascular risk, kidney disease or heart failure. This review summarizes current literature on the effect of SGLT-2 inhibitors on BP, and its potential relationships with cardio- and nephroprotection.
Collapse
|
34
|
Zobel EH, von Scholten BJ, Goldman B, Persson F, Hansen TW, Rossing P. Pleiotropic effects of liraglutide in patients with type 2 diabetes and moderate renal impairment: Individual effects of treatment. Diabetes Obes Metab 2019; 21:1261-1265. [PMID: 30663196 PMCID: PMC6593738 DOI: 10.1111/dom.13638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/08/2019] [Accepted: 01/17/2019] [Indexed: 01/09/2023]
Abstract
Liraglutide has pleiotropic effects favouring cardiovascular and renal risks. We investigated individual responses to liraglutide in six cardio-renal risk factors to examine whether responses in one risk factor are associated with changes in other risk factors (cross-dependency). We performed secondary analysis of the LIRA-RENAL trial (n = 279) in type 2 diabetes. HbA1c, body weight, systolic blood pressure (SBP), low density lipoprotein (LDL)-cholesterol, urine albumin-to-creatinine ratio (UACR) and estimated glomerular filtration rate (eGFR) were measured at baseline and after 26 weeks of liraglutide/placebo treatment: "Good responders" had a change within the best quartile. In the liraglutide-treated group, good HbA1c responders showed similar changes in other risk factors analysed to low responders (P ≥ 0.17). Good body weight responders had a larger reduction in HbA1c than low body weight responders (-1.6 ± 0.94 vs. -1.0 ± 0.82%; P = 0.003), but similar changes in the other risk factors (P ≥ 0.11). Good and low responders in SBP, UACR, LDL-cholesterol or eGFR showed similar changes in other risk factors (P ≥ 0.07). Treatment response to liraglutide is largely individual; aside from an association between body weight and HbA1c reduction, there are no obvious cross-dependencies in risk factor response.
Collapse
Affiliation(s)
- Emilie H. Zobel
- Complications Research, Steno Diabetes Center CopenhagenGentofteDenmark
| | - Bernt J. von Scholten
- Complications Research, Steno Diabetes Center CopenhagenGentofteDenmark
- Novo Nordisk A/SSøborgDenmark
| | | | - Frederik Persson
- Complications Research, Steno Diabetes Center CopenhagenGentofteDenmark
| | - Tine W. Hansen
- Complications Research, Steno Diabetes Center CopenhagenGentofteDenmark
| | - Peter Rossing
- Complications Research, Steno Diabetes Center CopenhagenGentofteDenmark
- Department of Clinical Medicine, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
35
|
Muskiet MHA, Wheeler DC, Heerspink HJL. New pharmacological strategies for protecting kidney function in type 2 diabetes. Lancet Diabetes Endocrinol 2019; 7:397-412. [PMID: 30579729 DOI: 10.1016/s2213-8587(18)30263-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/16/2018] [Accepted: 08/21/2018] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes is the leading cause of impaired kidney function, albuminuria, and renal replacement therapy globally, thus placing a large burden on health-care systems. Current treatment strategies rely on intensive glucose lowering as well as strict blood pressure control through blockade of the renin-angiotensin-aldosterone system. Such approaches might slow decline in kidney function, but many patients progress to end-stage kidney failure despite optimal therapy. In recent clinical trials, new-generation glucose-lowering drug classes, the sodium-glucose co-transporter-2 inhibitors and agents that target the incretin pathway, have been shown to improve kidney outcomes in patients with type 2 diabetes. Other new approaches, which have been developed on the basis of an improved understanding of the mechanisms that contribute to kidney damage in the context of diabetes, include use of drugs that block endothelin receptors (eg, atrasentan) and non-steroidal mineralocorticoid receptors (eg, finerenone). In this Review, we provide an overview of recent clinical data relevant to these new therapeutic approaches for management of kidney disease in the context of type 2 diabetes.
Collapse
Affiliation(s)
- Marcel H A Muskiet
- Diabetes Centre, Department of Internal Medicine, VU University Medical Centre, Amsterdam, Netherlands
| | - David C Wheeler
- Centre for Nephrology, University College London, London, UK.
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| |
Collapse
|
36
|
van Baar MJB, van der Aart AB, Hoogenberg K, Joles JA, Heerspink HJL, van Raalte DH. The incretin pathway as a therapeutic target in diabetic kidney disease: a clinical focus on GLP-1 receptor agonists. Ther Adv Endocrinol Metab 2019; 10:2042018819865398. [PMID: 31384419 PMCID: PMC6657126 DOI: 10.1177/2042018819865398] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022] Open
Abstract
Diabetic kidney disease (DKD) remains the main cause for chronic kidney disease (CKD) and end-stage kidney disease (ESKD) worldwide. Both CKD and ESKD lead to major increases in risk of cardiovascular disease and death in people with diabetes. Despite optimal management of lifestyle, glucose levels and hypertension, residual risk remains high, indicating that additional therapies to mitigate the burden of the disease are desired. In past decades, new treatment options for the management of diabetes have emerged, of which some have showed promising renoprotective potential. This review discusses current understanding of the renal effects of glucagon-like peptide receptor agonists and their potential use in prevention and treatment of DKD.
Collapse
Affiliation(s)
- Michaël J. B. van Baar
- Department of Internal Medicine, Amsterdam University Medical Centers, VUMC, Amsterdam, The Netherlands
| | - Annemarie B. van der Aart
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy, Martini Hospital, Groningen, The Netherlands
| | - Klaas Hoogenberg
- Department of Internal Medicine, Martini Hospital, Groningen, The Netherlands
| | - Jaap A. Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | | |
Collapse
|
37
|
Delanaye P, Scheen AJ. Preventing and treating kidney disease in patients with type 2 diabetes. Expert Opin Pharmacother 2018; 20:277-294. [PMID: 30462565 DOI: 10.1080/14656566.2018.1551362] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Chronic kidney disease (CKD) represents a huge burden in patients with type 2 diabetes (T2DM). This review therefore has the aim of assessing the add-on value of new glucose-lowering agents compared or combined with inhibitors of the renin angiotensin aldosterone system (RAAS) on renal outcomes in T2DM patients. AREAS COVERED This article first summarizes the results reported with RAAS inhibitors, mainstay of nephroprotection in T2DM with albuminuria. Second, it describes the positive results with glucagon-like peptide-1 receptor agonists (GLP-1RAs) and, even more impressive, sodium-glucose cotransporter type 2 inhibitors (SGLT2is). Third, besides the potential of combined therapies, it briefly considers some new approaches currently in development. EXPERT OPINION RAAS inhibitors exert renoprotective effects beyond their blood pressure lowering effects while SGLT2is, and possibly GLP-1RAs, exert nephroprotection independently of their glucose-lowering activity. These effects were demonstrated not only on surrogate endpoints such as albuminuria and estimated glomerular filtration rate decline, but also on hard endpoints, including progression to end-stage renal disease requiring replacement therapy. The underlying mechanisms are different and potentially complementary on glomerular hemodynamics, arguing for combined therapies. Nevertheless, there is still room for new emerging drugs to tackle CKD in T2DM.
Collapse
Affiliation(s)
- Pierre Delanaye
- a Division of Nephrology, Dialysis and Transplantation, Department of Medicine , Liège , Belgium
| | - André J Scheen
- b Division of Clinical Pharmacology, Centre for Interdisciplinary Research on Medicines (CIRM) , University of Liège , Liège , Belgium.,c Department of Medicine, Division of Diabetes , Nutrition and Metabolic Disorders , Liège , Belgium
| |
Collapse
|
38
|
León Jiménez D, Cherney DZI, Bjornstad P, Castilla-Guerra L, Miramontes González JP. Antihyperglycemic agents as novel natriuretic therapies in diabetic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1406-F1415. [PMID: 30066584 PMCID: PMC6293300 DOI: 10.1152/ajprenal.00384.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 07/11/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023] Open
Abstract
While sodium-glucose cotransporter-2 (SGLT2) inhibitors have been used for the routine management of type 2 diabetes for several years, it is perhaps their natriuretic effects that are most important clinically. This natriuresis activates tubuloglomerular feedback, resulting in reduced glomerular hypertension and proteinuria, leading to renal protective effects in the EMPA-REG OUTCOME and CANVAS Program trials. In the cardiovascular system, it is likely that plasma volume contraction due to natriuresis in response to SGLT2 inhibition is at least in part responsible for the reduction in the risk of heart failure observed in these trials. We compare this mechanism of action with other antidiabetics. Importantly, other diuretic classes, including thiazide and loop diuretics, have not resulted in such robust clinical benefits in patients with type 2 diabetes, possibly because these older agents do not influence intraglomerular pressure directly. In contrast, SGLT2 inhibitors do have important physiological similarities with carbonic anhydrase inhibitors, which also act proximally, and have been shown to activate tubuloglomerular feedback.
Collapse
Affiliation(s)
- David León Jiménez
- Vascular Risk Unit, Internal Medicine Clinical Management Unit, Hospital Universitario Virgen Macarena , Seville , Spain
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology. Toronto General Hospital, University of Toronto , Toronto , Ontario, Canada
| | - Petter Bjornstad
- Department of Pediatrics, Division of Endocrinology and Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado , Aurora, Colorado
| | - Luis Castilla-Guerra
- Vascular Risk Unit, Internal Medicine Clinical Management Unit, Hospital Universitario Virgen Macarena , Seville , Spain
| | - José Pablo Miramontes González
- Service of Internal Medicine, Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca , Salamanca , Spain
| |
Collapse
|
39
|
Muskiet MHA, Tonneijck L, Huang Y, Liu M, Saremi A, Heerspink HJL, van Raalte DH. Lixisenatide and renal outcomes in patients with type 2 diabetes and acute coronary syndrome: an exploratory analysis of the ELIXA randomised, placebo-controlled trial. Lancet Diabetes Endocrinol 2018; 6:859-869. [PMID: 30292589 DOI: 10.1016/s2213-8587(18)30268-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/23/2018] [Accepted: 08/29/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND The results of the ELIXA trial demonstrated the cardiovascular safety of lixisenatide, a short-acting glucagon-like peptide-1 receptor agonist, in patients with type 2 diabetes and acute coronary syndrome. In this exploratory analysis of ELIXA, we investigate the effect of lixisenatide on renal outcomes. METHODS ELIXA was a randomised, double-blind, placebo-controlled trial, done at 828 sites in 49 countries. Patients with type 2 diabetes and a recent coronary artery event were randomly assigned (1:1) to a daily subcutaneous injection of lixisenatide (10-20 μg) or volume-matched placebo, in addition to usual care, until at least 844 patients had an adjudicated major adverse cardiovascular event included in the primary outcome. Patients, study staff, and individuals involved in analysis of trial data were masked to treatment assignment. The primary and secondary endpoints of this trial have been reported previously. Here, in an exploratory analysis of ELIXA, we investigated percentage change in urinary albumin-to-creatinine ratio (UACR) and estimated glomerular filtration rate (eGFR) according to prespecified albuminuria status at baseline (normoalbuminuria [UACR <30 mg/g]; microalbuminuria [≥30 to ≤300 mg/g]; and macroalbuminuria [>300 mg/g]) using a mixed-effect model with repeated measures. Time to new-onset macroalbuminuria and doubling of serum creatinine were also assessed with Cox proportional hazards models. The ELIXA trial is registered with ClinicalTrials.gov, number NCT01147250, and is completed. FINDINGS Of 6068 patients randomly allocated between July 9, 2010, and Aug 2, 2013, baseline UACR data were available for 5978 (99%). Median follow-up time was 108 weeks. 4441 (74%; 2191 assigned to placebo and 2250 assigned to lixisenatide) had normoalbuminuria, 1148 (19%; 596 assigned to placebo and 552 assigned to lixisenatide) had microalbuminuria, and 389 (7%; 207 assigned to placebo and 182 assigned to lixisenatide) had macroalbuminuria. After 108 weeks, the placebo-adjusted least-squares mean percentage change in UACR from baseline with lixisenatide was -1·69% (95% CI -11·69 to 8·30; p=0·7398) in patients with normoalbuminuria, -21·10% (-42·25 to 0·04; p=0·0502) in patients with microalbuminuria, and -39·18% (-68·53 to -9·84; p=0·0070) in patients with macroalbuminuria. Lixisenatide was associated with a reduced risk of new-onset macroalbuminuria compared with placebo when adjusted for baseline HbA1c (hazard ratio [HR] 0·808 [95% CI 0·660 to 0·991; p=0·0404]) or baseline and on-trial HbA1c (HR 0·815 [0·665 to 0·999; p=0·0491]); point estimates were similar when adjusted for other traditional renal risk factors. At week 108, the largest eGFR decline from baseline was observed in the macroalbuminuric group, but no significant differences were observed between the two treatment groups. No significant differences in eGFR decline were identified between treatment groups in any UACR subgroup. In the trial safety population, doubling of serum creatinine occurred in 35 (1%) of 3032 patients in the placebo group and 41 (1%) of 3031 patients in the lixisenatide group (HR 1·163, 95% CI 0·741-1·825; p=0·5127). As previously reported in the ELIXA trial, the proportion of patients with renal adverse events was low (48 [1·6%] of 3032 patients in the placebo group vs 48 [1·6%] of 3031 patients in the lixisenatide group) and did not significantly differ between treatment groups. INTERPRETATION Lixisenatide reduces progression of UACR in macroalbuminuric patients, and is associated with a lower risk of new-onset macroalbuminuria after adjustment for baseline and on-trial HbA1c and other traditional renal risk factors. FUNDING Sanofi.
Collapse
Affiliation(s)
- Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, Netherlands.
| | - Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
40
|
Tonneijck L, Muskiet MHA, Blijdorp CJ, Smits MM, Twisk JW, Kramer MHH, Danser AHJ, Diamant M, Joles JA, Hoorn EJ, van Raalte DH. Renal tubular effects of prolonged therapy with the GLP-1 receptor agonist lixisenatide in patients with type 2 diabetes mellitus. Am J Physiol Renal Physiol 2018; 316:F231-F240. [PMID: 30353743 DOI: 10.1152/ajprenal.00432.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) are well-established glucose-lowering drugs for type 2 diabetes mellitus (T2DM) management. Acute GLP-1RA administration increases urinary excretion of sodium and other electrolytes. However, the renal tubular effects of prolonged GLP-1RA treatment are largely unknown. In this secondary analysis of a randomized trial, we determined the renal tubular effects of 8-wk treatment with 20 μg lixisenatide, a short-acting (prandial) GLP-1RA, versus titrated once-daily insulin glulisine in 35 overweight T2DM-patients on stable insulin glargine background therapy (age: 62 ± 7 yr, glycated hemoglobin: 8.0 ± 0.9%, estimated glomerular filtration rate: >60 ml·min-1·1.73 m-2). After a standardized breakfast, lixisenatide increased absolute and fractional excretions of sodium, chloride, and potassium and increased urinary pH. In contrast, lixisenatide reduced absolute and fractional excretions of magnesium, calcium, and phosphate. At week 8, patients treated with lixisenatide had significantly more phosphorylated sodium-hydrogen exchanger isoform 3 (NHE3) in urinary extracellular vesicles than those on insulin glulisine treatment, which suggested decreased NHE3 activity in the proximal tubule. A rise in postprandial blood pressure with lixisenatide partly explained the changes in the urinary excretion of sodium, potassium, magnesium, and phosphate and the changes in urinary pH. In conclusion, lixisenatide affects postprandial urinary excretion of several electrolytes and increases urinary pH compared with insulin glulisine in T2DM patients after 8 wk of treatment. This is most likely explained by a drug-induced rise in blood pressure or direct inhibitory effects on NHE3 in the proximal tubule.
Collapse
Affiliation(s)
- Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| | - Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| | - Charles J Blijdorp
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| | - Jos W Twisk
- Department of Health Sciences and the EMGO Institute for Health and Care Research, VU University Amsterdam , Amsterdam , The Netherlands ; Department of Epidemiology and Biostatistics, VU University Medical Center , Amsterdam , The Netherlands
| | - Mark H H Kramer
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| | - A H J Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Michaela Diamant
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht , Utrecht , The Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
41
|
Rashid M, Verhoeven AJM, Mulder MT, Timman R, van Beek-Nieuwland Y, Athumani AA, Zandbergen AAM, van der Wiel HE, Sijbrands EJG, Berk KA. Use of monomeric and oligomeric flavanols in the dietary management of patients with type 2 diabetes mellitus and microalbuminuria (FLAVA trial): study protocol for a randomized controlled trial. Trials 2018; 19:379. [PMID: 30012185 PMCID: PMC6048822 DOI: 10.1186/s13063-018-2762-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/25/2018] [Indexed: 12/20/2022] Open
Abstract
Background Patients with type 2 diabetes mellitus (T2D) are prone to micro- and macro-vascular complications. Monomeric and oligomeric flavanols (MOF) isolated from grape seeds (Vitis vinifera) have been linked to improved endothelial function and vascular health. The aim of this study is to determine the effect of a daily supplementation of 200 mg MOF on renal endothelial function of patients with T2D and microalbuminuria. Methods/design For this double-blind, placebo-controlled, randomized, multicenter trial 96 individuals (ages 40–85 years) with T2D and microalbuminuria will be recruited. Participants will be randomly assigned to the intervention group, receiving 200 mg of MOF daily for 3 months, or to the control group, receiving a placebo. The primary endpoint is the evolution over time in albumin excretion rate (AER) until 3 months of intervention as compared with placebo. Secondary endpoints are the evolution over time in established plasma markers of renal endothelial function—asymmetric dimethylarginine (ADMA), soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular cell adhesion molecule-1 (sICAM-1), interleukin-6 (IL-6), and von Willebrand Factor (vWF)—until 3 months of intervention as compared with placebo. Mixed modeling will be applied for the statistical analysis of the data. Discussion We hypothesize that T2D patients with microalbuminuria have a medically determined requirement for MOF and that fulfilling this requirement will result in a decrease in AER and related endothelial biomarkers. If confirmed, this may lead to new insights in the dietary management of patients with T2D. Trial registration Nederlands Trial Register, NTR4669, registered on 7 July 2014. Electronic supplementary material The online version of this article (10.1186/s13063-018-2762-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mardin Rashid
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Adrie J M Verhoeven
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Reinier Timman
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Yvonne van Beek-Nieuwland
- Department of Internal Medicine, Havenziekenhuis, Haringvliet 2, Rotterdam, 3011 TD, The Netherlands
| | - Athumani A Athumani
- General Practitioners Group, Stichting Zorg op Zuid, Maashaven Oostzijde 155, Rotterdam, 3072 HS, The Netherlands
| | - Adrienne A M Zandbergen
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Internal Medicine, Ikazia Ziekenhuis, Montessoriweg 1, Rotterdam, 3083 HN, The Netherlands
| | - Hans E van der Wiel
- Department of Internal Medicine, IJsselland Ziekenhuis, Prins Constantijnweg 2, Capelle aan de Ijssel, 2906 ZC, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Kirsten A Berk
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands. .,Department of Dietetics, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
42
|
Lajara R. The MARLINA-T2D trial: putting the results into clinical perspective. Expert Rev Endocrinol Metab 2018; 13:173-176. [PMID: 30058901 DOI: 10.1080/17446651.2018.1455497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Chronic kidney disease (CKD) and type 2 diabetes mellitus (T2D) frequently coexist and are associated with poor clinical outcomes. Dipeptidyl peptidase-4 (DPP-4) inhibitors can be used for patients with T2D and CKD, and preclinical evidence suggests these agents may slow the progression of kidney disease in T2D. Although clinical evidence of the renal effects of DPP-4 inhibitors is limited, the recent publication of the MARLINA-T2D study provided important new data. In MARLINA-T2D, linagliptin was associated with significant improvements in glycemic control with a non-significant reduction in albuminuria and no evidence of renal adverse effects in a high-risk population of patients with T2D and early diabetic kidney disease. Although there was no conclusive evidence of renoprotective effects, previous research suggests that clinically apparent renal benefits might develop with longer term treatment. The results of ongoing trials with primary renal endpoints are awaited with interest.
Collapse
|
43
|
Tonneijck L, Muskiet MH, Smits MM, Bjornstad P, Kramer MH, Diamant M, Hoorn EJ, Joles JA, van Raalte DH. Effect of immediate and prolonged GLP-1 receptor agonist administration on uric acid and kidney clearance: Post-hoc analyses of four clinical trials. Diabetes Obes Metab 2018; 20:1235-1245. [PMID: 29341461 PMCID: PMC5899927 DOI: 10.1111/dom.13223] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/02/2018] [Accepted: 01/11/2018] [Indexed: 12/16/2022]
Abstract
AIMS To determine the effects of glucagon-like peptide (GLP)-1 receptor agonists (RA) on uric acid (UA) levels and kidney UA clearance. MATERIAL AND METHODS This study involved post-hoc analyses of 4 controlled clinical trials, which assessed actions of GLP-1RA administration on kidney physiology. The immediate effects of GLP-1RA exenatide infusion vs placebo were determined in 9 healthy overweight men (Study-A) and in 52 overweight T2DM patients (Study-B). The effects of 12 weeks of long-acting GLP-1RA liraglutide vs placebo in 36 overweight T2DM patients (Study-C) and of 8 weeks of short-acting GLP-1RA lixisenatide vs once-daily titrated insulin glulisine in 35 overweight T2DM patients (Study-D) were also examined. Plasma UA, fractional (inulin-corrected) and absolute urinary excretion of UA (UEUA ) and sodium (UENa ), and urine pH were determined. RESULTS Median baseline plasma UA level was 5.39 to 6.33 mg/dL across all studies (17%-22% of subjects were hyperuricaemic). In Study-A, exenatide infusion slightly increased plasma UA (+0.07 ± 0.02 mg/dL, P = .04), and raised absolute-UEUA (+1.58 ± 0.65 mg/min/1.73 m2 , P = .02), but did not affect fractional UEUA compared to placebo. Fractional UEUA and absolute UEUA correlated with increases in urine pH (r:0.86, P = .003 and r:0.92, P < .001, respectively). Fractional UEUA correlated with increased fractional UENa (r:0.76, P = .02). In Study-B, exenatide infusion did not affect plasma UA, but increased fractional UEUA (+0.76 ± 0.38%, P = .049) and absolute UEUA (+0.75 ± 0.27 mg/min/1.73 m2 , P = .007), compared to placebo. In regression analyses, both parameters were explained by changes in urine pH and, in part, by changes in UENa . In Study-C, liraglutide treatment did not affect plasma UA, UEUA, UENa or urine pH, compared to placebo. In Study-D, lixisenatide treatment increased UENa and urine pH from baseline, but did not affect plasma UA or UEUA . CONCLUSION Immediate exenatide infusion increases UEUA in overweight healthy men and in T2DM patients, probably by inhibiting Na+ /H+ -exchanger type-3 in the renal proximal tubule. Prolonged treatment with a long-acting or short-acting GLP-1RA does not affect plasma UA or UEUA in T2DM patients with normal plasma UA levels and at relatively low cardiovascular risk. Our results suggest that the cardio-renal benefits of GLP-1RA are not mediated through changes in UA.
Collapse
Affiliation(s)
- Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcel H.A. Muskiet
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark M. Smits
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO, United States; and Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO, United States
| | - Mark H.H. Kramer
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Michaela Diamant
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jaap A. Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Daniël H. van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Bjornstad P, Cherney DZ. Renal Hyperfiltration in Adolescents with Type 2 Diabetes: Physiology, Sex Differences, and Implications for Diabetic Kidney Disease. Curr Diab Rep 2018; 18:22. [PMID: 29557063 PMCID: PMC6344350 DOI: 10.1007/s11892-018-0996-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Compared to adult-onset type 2 diabetes (T2D), youth with T2D have a more aggressive phenotype with greater insulin resistance (IR), more rapid β cell decline, and higher prevalence of diabetic kidney disease (DKD). RECENT FINDINGS Hyperfiltration is common in youth with T2D and predicts progressive DKD. Hyperfiltration is a consequence of early changes in intrarenal hemodynamic function, including increased renal plasma flow (RPF) and glomerular pressure. Girls with T2D are disproportionally affected by DKD, with a 3-fold greater risk of developing hyperfiltration over 5 years compared to boys. Despite the high prevalence and gravity of DKD in youth-onset T2D, widely effective therapeutic options are lacking. In this review, we focus on pathophysiology underlying early DKD in T2D and sex differences and summarize promising novel medical therapies and bariatric surgery.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, 13123 East 16th Ave, Box B265, Aurora, CO, 80045, USA.
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
45
|
Deacon CF. A review of dipeptidyl peptidase-4 inhibitors. Hot topics from randomized controlled trials. Diabetes Obes Metab 2018; 20 Suppl 1:34-46. [PMID: 29364584 DOI: 10.1111/dom.13135] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/19/2017] [Accepted: 10/19/2017] [Indexed: 12/20/2022]
Abstract
The first clinical study to investigate effects of dipeptidyl peptidase-4 (DPP-4) inhibition was published in 2002, and since then, numerous randomized controlled trials (RCTs) have shown that DPP-4 inhibitors are efficacious, safe and well-tolerated. This review will focus upon RCTs which have investigated DPP-4 inhibitors in patient groups which are often under-represented or excluded from typical phase 3 clinical trials. Large cardiovascular (CV) safety outcome trials in patients with established CV disease have confirmed that DPP-4 inhibitors are not associated with any additional CV risk in these already-at-high-risk individuals, while raising awareness of any uncommon adverse events, such as heart failure hospitalization seen in one of the trials. Studies in patients with kidney disease have shown DPP-4 inhibitors to be efficacious without increasing the risk of hypoglycaemia, irrespective of the degree of renal impairment, while data from the large CV trials as well as smaller RCTs have even pointed towards potential renoprotective effects such individuals. The use of DPP-4 inhibitors with insulin when therapy requires intensification may be beneficial without affecting the incidence or severity of hypoglycaemia, with these effects also being replicated in patients with chronic kidney disease, for whom other agents may not be suitable. Attention is now turning towards exploring the potential utility of DPP-4 inhibitors in other circumstances, including for in-hospital management of hyperglycaemia and in other metabolic disorders. Together, these RCTs raise the possibility that in the future, DPP-4 inhibitors may have a broader use which may extend beyond glycaemic control in the typical type 2 diabetes mellitus (T2DM) patient seen in general practice and may encompass conditions other than T2DM.
Collapse
Affiliation(s)
- Carolyn F Deacon
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
Tonneijck L, Muskiet MHA, Smits MM, Hoekstra T, Kramer MHH, Danser AHJ, Diamant M, Joles JA, van Raalte DH. Postprandial renal haemodynamic effect of lixisenatide vs once-daily insulin-glulisine in patients with type 2 diabetes on insulin-glargine: An 8-week, randomised, open-label trial. Diabetes Obes Metab 2017; 19:1669-1680. [PMID: 28449402 DOI: 10.1111/dom.12985] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 01/17/2023]
Abstract
AIM To determine whether lixisenatide, a prandial short-acting glucagon-like peptide receptor agonist (GLP-1RA), ameliorates postprandial glomerular hyperfiltration in patients with type 2 diabetes mellitus (T2DM) compared with insulin-glulisine (iGlu). METHODS Postprandial renal haemodynamic effects of 8-week treatment with lixisenatide 20 µg vs once-daily titrated iGlu were measured in 35 overweight patients with T2DM inadequately controlled on insulin-glargine, with or without metformin [mean ± SD age 62 ± 7 years, HbA1c 8.0% ± 0.9%, estimated glomerular filtration rate (GFR) 85 ± 12 mL/min/1.73 m2 , median (IQR) urinary albumin/creatinine ratio 1.5 (0.9-3.0) mg/mmol]. After a standardised breakfast, GFR (primary endpoint) and effective renal plasma flow (ERPF) were determined by inulin and para-aminohippuric acid renal clearance, respectively, based on timed urine sampling. Intrarenal haemodynamic functions were estimated using Gomez equations. RESULTS Compared with iGlu, lixisenatide did not affect GFR [+0.1 mL/min/1.73 m2 (95% CI -9 to 9)], ERPF [-17 mL/min/1.73 m2 (-61 to 26)], other (intra-)renal haemodynamics or renal damage markers, but increased fractional sodium excretion [+0.25% (0.09-0.41)] and urinary pH [+0.7 (0.3-1.2)]. Plasma renin, angiotensin-II and aldosterone were unchanged. Lixisenatide and iGlu reduced HbA1c similarly, by 0.8% ± 0.1% and 0.6% ± 0.1%, respectively, while postprandial glucose was lower with lixisenatide (P = .002). Compared with iGlu, lixisenatide reduced bodyweight [-1.4 kg (-2.5 to -0.2)] and increased postprandial mean arterial pressure [+9 mm Hg (4-14)]. CONCLUSION Eight-week lixisenatide treatment does not affect postprandial (intra-)renal haemodynamics compared with iGlu when added to insulin-glargine in patients with T2DM without overt nephropathy. Prolonged lixisenatide treatment has a sustained natriuretic effect, which is in contrast to previous reports on long-acting GLP-1RA, reduces body weight and increases postprandial blood pressure compared with iGlu. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT02276196.
Collapse
Affiliation(s)
- Lennart Tonneijck
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcel H A Muskiet
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark M Smits
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Trynke Hoekstra
- Department of Health Sciences and the EMGO Institute for Health and Care Research, VU University Amsterdam, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark H H Kramer
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michaela Diamant
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, The Netherlands
| | - Daniël H van Raalte
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Affiliation(s)
| | | | - John B Buse
- University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
48
|
Renal outcomes with dipeptidyl peptidase-4 inhibitors. DIABETES & METABOLISM 2017; 44:101-111. [PMID: 29146035 DOI: 10.1016/j.diabet.2017.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 02/06/2023]
Abstract
Dipeptidyl peptidase-4 inhibitors (DPP-4is) are increasingly being used in the management of type 2 diabetes (T2D). The present review summarizes the current knowledge of the effects of DPP-4is on renal outcomes by analyzing the experimental preclinical data, the effects of DPP-4is on urinary albumin-creatinine ratios (UACRs) and estimated glomerular filtration rates (eGFRs) from observational studies and clinical trials, and renal events (including kidney failure requiring renal replacement therapy) in recent large prospective cardiovascular outcome trials. Renal protection has been demonstrated in various animal models that have implicated different underlying mechanisms independent of glucose control, whereas prevention of new onset microalbuminuria and/or progression of albuminuria has been reported in some clinical studies, but with no significant effects on eGFR in most of them. The long-term clinical effects of DPP-4is on renal outcomes and the development of end-stage renal disease remain largely unknown and, thus, demand further investigations in prospective trials and long-term observational studies. In conclusion, despite promising results in animal models, data on surrogate biological markers of renal function and clinical renal outcomes remain rather scanty in patients with T2D, and mostly demonstrate the safety rather than true efficacy of DPP-4is.
Collapse
|
49
|
Abstract
The gastrointestinal tract - the largest endocrine network in human physiology - orchestrates signals from the external environment to maintain neural and hormonal control of homeostasis. Advances in understanding entero-endocrine cell biology in health and disease have important translational relevance. The gut-derived incretin hormone glucagon-like peptide 1 (GLP-1) is secreted upon meal ingestion and controls glucose metabolism by modulating pancreatic islet cell function, food intake and gastrointestinal motility, amongst other effects. The observation that the insulinotropic actions of GLP-1 are reduced in type 2 diabetes mellitus (T2DM) led to the development of incretin-based therapies - GLP-1 receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors - for the treatment of hyperglycaemia in these patients. Considerable interest exists in identifying effects of these drugs beyond glucose-lowering, possibly resulting in improved macrovascular and microvascular outcomes, including in diabetic kidney disease. As GLP-1 has been implicated as a mediator in the putative gut-renal axis (a rapid-acting feed-forward loop that regulates postprandial fluid and electrolyte homeostasis), direct actions on the kidney have been proposed. Here, we review the role of GLP-1 and the actions of associated therapies on glucose metabolism, the gut-renal axis, classical renal risk factors, and renal end points in randomized controlled trials of GLP-1 receptor agonists and DPP-4 inhibitors in patients with T2DM.
Collapse
|
50
|
Kambara T, Shibata R, Osanai H, Nakashima Y, Asano H, Sakai K, Murohara T, Ajioka M. Use of sodium-glucose cotransporter 2 inhibitors in older patients with type 2 diabetes mellitus. Geriatr Gerontol Int 2017; 18:108-114. [PMID: 28861952 DOI: 10.1111/ggi.13149] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/18/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
AIM Sodium-glucose cotransporter 2 (SGLT2) inhibitors are antidiabetic agents that act on the proximal renal tubules to lower blood glucose levels by inhibiting glucose reabsorption and promoting urinary glucose excretion. The present study assessed the long-term use of SGLT2 inhibitors in older patients with diabetes. METHODS A total of 117 older patients with type 2 diabetes who were given SGLT2 inhibitors were enrolled from April 2014 to March 2016. RESULTS The mean age of the patients was 73.7 ± 10.0 years. During the follow-up period (mean 289.3 days), there was no event associated with oral administration of SGLT2 inhibitors. These drugs significantly lowered fasting blood glucose and glycosylated hemoglobin levels at 6 months, and did not affect the creatinine level, blood urea nitrogen/creatinine ratio or estimated glomerular filtration rate during treatment. Although the treatment significantly increased hemoglobin and hematocrit levels, it did not affect the ultrasonographically determined diameter of the inferior vena cava, and no signs of intravascular collapse were observed. Changes in brain natriuretic peptide levels during the follow-up period were assessed in 78 patients with a brain natriuretic peptide level exceeding the normal upper limit before treatment with SGLT2 inhibitors. The brain natriuretic peptide levels significantly decreased after 6 months of treatment. CONCLUSIONS In older Japanese patients with diabetes, treatment with SGLT2 inhibitors for 6 months exerted a favorable hypoglycemic effect, while no sign of dehydration was observed. Geriatr Gerontol Int 2018; 18: 108-114.
Collapse
Affiliation(s)
- Takahiro Kambara
- Department of Cardiovascular Medicine, Tosei General Hospital, Seto, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Osanai
- Department of Cardiovascular Medicine, Tosei General Hospital, Seto, Japan
| | | | - Hiroshi Asano
- Department of Cardiovascular Medicine, Tosei General Hospital, Seto, Japan
| | - Kazuyoshi Sakai
- Department of Cardiovascular Medicine, Tosei General Hospital, Seto, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masayoshi Ajioka
- Department of Cardiovascular Medicine, Tosei General Hospital, Seto, Japan
| |
Collapse
|