1
|
Iordan L, Gaita L, Timar R, Avram V, Sturza A, Timar B. The Renoprotective Mechanisms of Sodium-Glucose Cotransporter-2 Inhibitors (SGLT2i)-A Narrative Review. Int J Mol Sci 2024; 25:7057. [PMID: 39000165 PMCID: PMC11241663 DOI: 10.3390/ijms25137057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Chronic kidney disease (CKD) is a noncommunicable condition that has become a major healthcare burden across the globe, often underdiagnosed and associated with low awareness. The main cause that leads to the development of renal impairment is diabetes mellitus and, in contrast to other chronic complications such as retinopathy or neuropathy, it has been suggested that intensive glycemic control is not sufficient in preventing the development of diabetic kidney disease. Nevertheless, a novel class of antidiabetic agents, the sodium-glucose cotransporter-2 inhibitors (SGLT2i), have shown multiple renoprotective properties that range from metabolic and hemodynamic to direct renal effects, with a major impact on reducing the risk of occurrence and progression of CKD. Thus, this review aims to summarize current knowledge regarding the renoprotective mechanisms of SGLT2i and to offer a new perspective on this innovative class of antihyperglycemic drugs with proven pleiotropic beneficial effects that, after decades of no significant progress in the prevention and in delaying the decline of renal function, start a new era in the management of patients with CKD.
Collapse
Affiliation(s)
- Liana Iordan
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Second Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Laura Gaita
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Second Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Romulus Timar
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Second Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Vlad Avram
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Second Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adrian Sturza
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Bogdan Timar
- “Pius Brinzeu” Emergency County Hospital, 300723 Timisoara, Romania; (L.I.); (R.T.); (V.A.); (A.S.); (B.T.)
- Second Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
2
|
Berezin AE, Berezina TA. Plausible prediction of renoprotective effects of sodium-glucose cotransporter-2 inhibitors in patients with chronic kidney diseases. J Int Med Res 2024; 52:3000605241227659. [PMID: 38329077 PMCID: PMC10854388 DOI: 10.1177/03000605241227659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/20/2023] [Indexed: 02/09/2024] Open
Abstract
This narrative review was conducted due to uncertainty in predicting the beneficial impact of sodium-glucose cotransporter-2 (SGLT2) inhibitors on a dip of estimated glomerular filtration rate (eGFR), regardless of albuminuria presence, with the aim of elucidating plausible predictors of kidney function outcome among patients treated with SGLT2 inhibitors. The PubMed and Web of Science databases were searched in May 2023 for relevant articles published in English between 2013 and 2023. A total of 25 full-length scientific publications (comprising 11 large randomized trials and two cohort studies) were included for analysis. The majority of studies demonstrated a limited value of conventional biomarkers, such as initial decline in eGFR, a trajectory of eGFR during SGLT2 inhibitor administration, and urine albumin-to-creatinine ratio (UACR), in prediction of renoprotection. Included studies showed that the tendency to decreased eGFR, UACR, hemoglobin, glycosylated hemoglobin, lipid profile, serum uric acid, inflammatory biomarkers and natriuretic peptides did not predict clinical outcomes in groups without heart failure (HF) treated with SGLT2 inhibitors. In HF groups, biomarkers of inflammation, kidney injury, oxidative stress, mitochondrial dysfunction, ketogenesis, energy metabolism, and adipose tissue dysfunction (adropin and irisin), were detected with the aim of finding potential biomarkers. Biomarkers of adipose tissue dysfunction and inflammation may be promising for predicting SGLT2 inhibitor benefit compared with N-terminal pro-B-type natriuretic peptide and energy metabolism indicators.
Collapse
Affiliation(s)
- Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, Ukraine
| |
Collapse
|
3
|
Ito H, Inoue H, Izutsu T, Matsumoto S, Antoku S, Yamasaki T, Mori T, Togane M. Changes in the estimated glomerular filtration rate and predictors of the renal prognosis in Japanese patients with type 2 diabetes: A retrospective study during the 12 months after the initiation of tofogliflozin. PLoS One 2023; 18:e0292014. [PMID: 37733761 PMCID: PMC10513294 DOI: 10.1371/journal.pone.0292014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND The changes in the estimated glomerular filtration rate (eGFR) and predictors of the renal prognosis were retrospectively assessed over the 12 months after the initiation of tofogliflozin, which has the shortest half-life among sodium-glucose cotransporter 2 (SGLT2) inhibitors, in Japanese patients with type 2 diabetes and renal impairment. METHODS In total, 158 patients treated with tofogliflozin between 2019 and 2021 were studied as the safety analysis set. One hundred and thirty subjects whose medication was continued over 12 months were investigated as the full analysis set. The subjects were divided into two groups based on the eGFR: normal- (eGFR ≥60 mL/min/1.73 m2, n = 87) and low- (eGFR <60 mL/min/1.73 m2, n = 43) eGFR groups. RESULTS The body weight, blood pressure, urinary protein excretion, and serum uric acid concentration decreased from baseline in both eGFR groups while the hemoglobin level increased. The eGFR did not significantly differ over time, except for the initial dip (-4.3±9.6 mL/min/1.73 m2 in the normal-eGFR group and -1.5±5.3 mL/min/1.73 m2 in the low-eGFR group). The change in the eGFR at 12 months after the initiation of tofogliflozin was -1.9±9.0 mL/min/1.73 m2 and 0.2±6.0 mL/min/1.73 m2 in the normal- and low-eGFR group, respectively. In the normal-eGFR group, the change in the eGFR showed a significant negative correlation with the HbA1c and eGFR at baseline, according to a multiple regression analysis. In the low-eGFR group, the change in the eGFR showed a significant negative correlation with urate-lowering agent use. The frequencies of adverse events specific for SGLT2 inhibitors were not significantly different between the normal- and low-eGFR groups. CONCLUSIONS Tofogliflozin may preserve renal function in the medium term in patients with type 2 diabetes and kidney impairment without an increase in specific adverse events.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Hideyuki Inoue
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Takuma Izutsu
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Suzuko Matsumoto
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Shinichi Antoku
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Tomoko Yamasaki
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Toshiko Mori
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| | - Michiko Togane
- Department of Diabetes, Metabolism and Kidney Disease, Edogawa Hospital, Edogawa-Ku, Tokyo, Japan
| |
Collapse
|
4
|
Villarreal D, Ramírez H, Sierra V, Amarís JS, Lopez-Salazar AM, González-Robledo G. Sodium-Glucose Cotransporter 2 Inhibitors in Frail Patients with Heart Failure: Clinical Experience of a Heart Failure Unit. Drugs Aging 2023; 40:293-299. [PMID: 36811172 DOI: 10.1007/s40266-022-01004-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2022] [Indexed: 02/23/2023]
Abstract
OBJECTIVE The objective of this study was to determine the difference in tolerance for sodium glucose cotransporter 2 inhibitors between patients with heart failure classified as frail according to the FRAIL questionnaire, compared to those with heart failure without frailty. METHODS A prospective cohort study was performed between 2021 and 2022 that included patients with heart failure at a heart failure unit in Bogotá who were being treated with a sodium glucose cotransporter 2 inhibitor. Clinical and laboratory data were collected during an initial visit and 12-48 weeks after that. The FRAIL questionnaire was applied to all participants through a phone call or during the follow-up visit. The primary outcome was the adverse effect rate and as a secondary outcome we compared the estimated glomerular filtration rate change between frail and non-frail patients. RESULTS One hundred and twelve patients were included in the final analysis. Frail patients had a more than twice increased risk of having adverse effects (95% confidence interval 1.5-3.9). Age was also a risk factor for the appearance of these. The estimated glomerular filtration rate decrease was inversely correlated with the age, left ventricular ejection fraction, and renal function before the use of sodium glucose cotransporter 2 inhibitors. CONCLUSIONS When prescribing in heart failure, it is important to remember that frail patients are more likely to have adverse effects with the use of sodium glucose cotransporter 2 inhibitors, of which the most common are those related to osmotic diuresis. Nonetheless, these do not appear to increase the risk of discontinuation or abandonment of therapy in this population.
Collapse
Affiliation(s)
- Daniel Villarreal
- School of Medicine, Universidad de los Andes, Cra. 7 #116-5, 110121, Bogotá, Colombia.
| | - Hernán Ramírez
- Heart Failure Unit, Internal Medicine Department, University Hospital Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | - Juan S Amarís
- School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Ana M Lopez-Salazar
- Geriatrics Team and Palliative Care Unit, Internal Medicine Department, University Hospital Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | - Gina González-Robledo
- Heart Failure Unit, Internal Medicine Department, University Hospital Fundación Santa Fe de Bogotá, Bogotá, Colombia
| |
Collapse
|
5
|
Wolf L, Föller M, Feger M. The impact of SGLT2 inhibitors on αKlotho in renal MDCK and HK-2 cells. Front Endocrinol (Lausanne) 2023; 14:1069715. [PMID: 36967770 PMCID: PMC10032406 DOI: 10.3389/fendo.2023.1069715] [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: 10/14/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
αKlotho is a transmembrane protein predominantly expressed in the kidney serving as a co-receptor for phosphate homeostasis-regulating hormone FGF23 and has an extracellular domain that can be cleaved off and is a hormone. αKlotho deficiency results in accelerated aging and early onset of aging-associated diseases while its overexpression strongly expands the lifespan of mice. Moreover, αKlotho exerts health-beneficial anti-inflammatory, anti-neoplastic, anti-fibrotic, and anti-oxidant effects. Higher αKlotho levels are associated with better outcomes in renal and cardiovascular diseases. SGLT2 inhibitors are novel drugs in the treatment of diabetes by inhibiting renal glucose transport and have additional nephro- and cardioprotective effects. We explored whether SGLT2 inhibitors affect αKlotho gene expression and protein secretion. Experiments were performed in renal MDCK and HK-2 cells, and αKlotho transcripts were determined by qRT-PCR and Klotho protein by ELISA. SGLT2 inhibitors canagliflozin, sotagliflozin, and dapagliflozin enhanced whereas empagliflozin reduced αKlotho gene expression in MDCK cells. By the same token, canagliflozin, sotagliflozin, dapagliflozin, but not empagliflozin down-regulated p65 subunit of pro-inflammatory NFκB. In HK-2 cells, all SGLT2 inhibitors reduced αKlotho transcripts. Canagliflozin and sotagliflozin, however, increased Klotho protein concentration in the cell culture supernatant, an effect paralleled by up-regulation of ADAM17. Taken together, our investigations demonstrate complex effects of different SGLT2 inhibitors on αKlotho gene expression and protein secretion in renal MDCK and HK-2 cells.
Collapse
|
6
|
Forycka J, Hajdys J, Krzemińska J, Wilczopolski P, Wronka M, Młynarska E, Rysz J, Franczyk B. New Insights into the Use of Empagliflozin-A Comprehensive Review. Biomedicines 2022; 10:biomedicines10123294. [PMID: 36552050 PMCID: PMC9775057 DOI: 10.3390/biomedicines10123294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Empagliflozin is a relatively new drug that, as an inhibitor of the sodium−glucose cotransporter 2 (SGLT2), causes increased urinary glucose excretion and thus contributes to improved glycemic control, better glucose metabolism, reduced glucotoxicity and insulin resistance. Although its original use was to induce a hypoglycemic effect in patients with type 2 diabetes mellitus (T2DM), empagliflozin has also shown a number of other beneficial effects by demonstrating a nephroprotective effect, and it has proven to be a breakthrough in the treatment of heart failure (HF). Empagliflozin has been shown to reduce hospitalizations for HF and the number of deaths from cardiovascular causes. Empagliflozin treatment also reduces the incidence of renal events, including death from renal causes, as well as the risk of end-stage renal failure. Empagliflozin appears to be a fairly well-tolerated and safe drug. In patients with inadequate glycemic control, empagliflozin used in monotherapy or as an adjunct to therapy effectively lowers fasting blood glucose, postprandial blood glucose, average daily glucose levels, glycated hemoglobin A1C (HbA1C) and also leads to significant weight reduction in patients with T2DM. Unfortunately, there are some limitations, e.g., severe hypersensitivity reaction to the drug and a glomerular filtration rate (GFR) < 30 mL/min/1.73 m2. As with any drug, empagliflozin is also characterized by several side effects among which symptomatic hypotension, troublesome genital fungal infections, urinary tract infections and rare ketoacidosis are characteristic.
Collapse
Affiliation(s)
- Joanna Forycka
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Hajdys
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Julia Krzemińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Piotr Wilczopolski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Wronka
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
- Correspondence: ; Tel.: +48-(042)-639-37-50
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
7
|
Sędzikowska A, Szablewski L. Human Glucose Transporters in Renal Glucose Homeostasis. Int J Mol Sci 2021; 22:13522. [PMID: 34948317 PMCID: PMC8708129 DOI: 10.3390/ijms222413522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022] Open
Abstract
The kidney plays an important role in glucose homeostasis by releasing glucose into the blood stream to prevent hypoglycemia. It is also responsible for the filtration and subsequent reabsorption or excretion of glucose. As glucose is hydrophilic and soluble in water, it is unable to pass through the lipid bilayer on its own; therefore, transport takes place using carrier proteins localized to the plasma membrane. Both sodium-independent glucose transporters (GLUT proteins) and sodium-dependent glucose transporters (SGLT proteins) are expressed in kidney tissue, and mutations of the genes coding for these glucose transporters lead to renal disorders and diseases, including renal cancers. In addition, several diseases may disturb the expression and/or function of renal glucose transporters. The aim of this review is to describe the role of the kidney in glucose homeostasis and the contribution of glucose transporters in renal physiology and renal diseases.
Collapse
Affiliation(s)
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| |
Collapse
|
8
|
Abstract
Over the past 40 years there has been a steady rise in the number of people with chronic kidney disease due mainly to a significant increase in the number of people with diabetic kidney disease (DKD). Current treatments (blood pressure control, blood sugar control, and renin-angiotensin-aldosterone system inhibitors) have had a significant impact on slowing progression of DKD. But the continued rise illustrates that there is a great need for new medications. Recently, a number of potentially reno-protective medicines have been studied. In this review, these new medications are discussed with respect to both their reported benefits and possible risks.
Collapse
|
9
|
Escott GM, da Silveira LG, Cancelier VDA, Dall'Agnol A, Silveiro SP. Monitoring and management of hyperglycemia in patients with advanced diabetic kidney disease. J Diabetes Complications 2021; 35:107774. [PMID: 33168397 DOI: 10.1016/j.jdiacomp.2020.107774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/10/2020] [Accepted: 10/10/2020] [Indexed: 11/19/2022]
Abstract
Diabetes mellitus is the leading cause of end-stage renal disease, and uncontrolled hyperglycemia is directly related to the increased mortality in this setting. As kidney function decreases, it becomes more challenging to control blood glucose since the risk of hypoglycemia increases. Decreased appetite, changes in glycaemia homeostasis, along with reduced renal excretion of anti-hyperglycemic drugs tend to facilitate the occurrence of hypoglycemia, despite the paradoxical occurrence of insulin resistance in advanced kidney disease. Thus, in patients using insulin and/or oral anti-hyperglycemic agents, dynamic adjustments with drug dose reduction or drug switching are often necessary. Furthermore, in addition to consider these pharmacokinetics alterations, it is of utmost importance to choose drugs with proven cardio-renal benefits in this setting, such as sodium-glucose co-transporter 2 inhibitors and glucagon-like peptide 1 receptor agonists. In this review, we summarize the indications and contraindications, titration of doses and side effects of the available anti-hyperglycemic agents in the presence of advanced diabetic kidney disease (DKD) and dialysis, highlighting the risks and benefits of the different agents. Additionally, basic renal function assessment and monitoring of glycemic control in DKD will be evaluated in order to guide the use of drugs and define the glycemic targets to be achieved.
Collapse
Affiliation(s)
- Gustavo Monteiro Escott
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Brazil
| | | | | | - Angélica Dall'Agnol
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Brazil
| | - Sandra Pinho Silveiro
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Brazil; Endocrine Unit, Hospital de Clínicas de Porto Alegre, RS, Brazil.
| |
Collapse
|
10
|
Genuardi MV, Mather PJ. The dawn of the four-drug era? SGLT2 inhibition in heart failure with reduced ejection fraction. Ther Adv Cardiovasc Dis 2021; 15:17539447211002678. [PMID: 33779401 PMCID: PMC8010852 DOI: 10.1177/17539447211002678] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Sodium-glucose cotransporter type 2 (SGLT2) inhibitors are a relatively new class of antihyperglycemic drug with salutary effects on glucose control, body weight, and blood pressure. Emerging evidence now indicates that these drugs may have a beneficial effect on outcomes in heart failure with reduced ejection fraction (HFrEF). Post-approval cardiovascular outcomes data for three of these agents (canagliflozin, empagliflozin, and dapagliflozin) showed an unexpected improvement in cardiovascular endpoints, including heart failure hospitalization and mortality, among patients with type 2 diabetes mellitus (T2DM) and established cardiovascular disease or risk factors. These studies were followed by a placebo controlled trial of dapagliflozin in patients with HFrEF both with and without T2DM, showing a reduction in all-cause mortality comparable to current guideline-directed HFrEF medical therapies such as angiotensin-converting enzyme inhibitors and beta-blockers. In this review, we discuss the current landscape of evidence, safety and adverse effects, and proposed mechanisms of action for use of these agents for patients with HFrEF. The United States (US) and European guidelines are reviewed, as are the current US federally approved indications for each SGLT2 inhibitor. Use of these agents in clinical practice may be limited by an uncertain insurance environment, especially in patients without T2DM. Finally, we discuss practical considerations for the cardiovascular clinician, including within-class differences of the SGLT2 inhibitors currently available on the US market (217/300).
Collapse
Affiliation(s)
- Michael V. Genuardi
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul J. Mather
- Perelman School of Medicine, University of Pennsylvania, 2 East Perelman Center for Advanced Medicine, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Takahashi K, Nakamura A, Furusawa S, Yokozeki K, Sugawara H, Yanagisawa H, Akikawa K, Kikuchi H. Initial dip predicts renal protective effects after the administration of sodium-glucose cotransporter 2 inhibitors in patients with type 2 diabetes and chronic kidney disease with normoalbuminuria. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2020; 22:100244. [PMID: 33318949 PMCID: PMC7726333 DOI: 10.1016/j.jcte.2020.100244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 01/14/2023]
Abstract
Investigated renoprotective effect of SGLT2is in T2DM and CKD with normoalbuminuria. More favorable effects of SGLT2is on renal function were observed. Higher initial dip was possible marker of worsening renal function.
Introduction We investigated the renoprotective effects of sodium-glucose cotransporter 2 inhibitors (SGLT2is) on renal function in patients with type 2 diabetes and chronic kidney disease (CKD) with normoalbuminuria. Methods A retrospective review of clinical records of Japanese participants with type 2 diabetes and CKD (estimated glomerular filtration rate [eGFR] < 60 mL/min/1.73 m2) with normoalbuminuria (urine albumin to creatinine ratio < 30 mg/g Cr and/or urinary protein to creatinine ratio < 150 mg/g Cr) was conducted. Participants were categorized into two groups depending on whether they had started using SGLT2is. The main study outcome was a comparison of the change in renal function evaluated by eGFR after 1 year between the two groups. Then, we identified predictors that were associated with the outcome. Results Among the 46 participants, 21 were treated with SGLT2is (SGLT2 group) and 25 were treated with other antidiabetic medications (control group). Although eGFR was significantly decreased at 1 year in the control group, the decline in eGFR was not observed in the SGLT2 group. The decrease in eGFR was significantly smaller in the SGLT2 group than in the control group. Additionally, multiple linear regression analysis showed that an initial dip was an independent factor associated with the worsening of renal function in the SGLT2 group. Conclusions Although more favorable effects of SGLT2is on renal function were observed in patients with type 2 diabetes and CKD with normoalbuminuria, the higher initial dip was a possible marker of worsening renal function after the initiation of SGLT2is.
Collapse
Affiliation(s)
- Kiyohiko Takahashi
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Akinobu Nakamura
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sho Furusawa
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Kei Yokozeki
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Hajime Sugawara
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Hideyuki Yanagisawa
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Kazumasa Akikawa
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Hideaki Kikuchi
- Third Department of Internal Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| |
Collapse
|
12
|
Hiramatsu T, Ito H, Okumura S, Asano Y, Iguchi D, Furuta S. Impact of glucagon like peptide-1 receptor agonist and sodium glucose cotransporter 2 inhibitors on type 2 diabetes patients with renal impairment. Diab Vasc Dis Res 2020; 17:1479164120971220. [PMID: 33371732 PMCID: PMC7919219 DOI: 10.1177/1479164120971220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Diabetes mellitus is a progressive disease with cardiovascular complications. We evaluated the impact of a glucagon like peptide-1 (GLP-1) receptor agonist and sodium glucose cotransporter 2 (SGLT-2) inhibitors dapagliflozin and empagliflozin on renal and cardiac function in type 2 diabetes patients with renal impairment. MATERIALS AND METHODS A total of 156 patients referred with suboptimal glycemic control were assigned to Group G (GLP-1): n = 72 or Group S (SGLT-2 inhibitor)-dapagliflozin (n = 52) or empagliflozin (n = 32). Renal function was assessed every 3 months for 36 months. Cardiovascular parameters were evaluated every 12 months for 36 months. RESULTS Compared with baseline, HbA1c and systolic blood pressure significantly decreased in both groups (p < 0.05). The estimated glomerular filtration rate decreased, but without significance. Albuminuria decreased significantly in both groups and then subsequently increased after 30 months in Group S. Diastolic cardiac function, assessed by E/e' or left atrial volume index, decreased only in Group G at 36 months. CONCLUSIONS The GLP-1 receptor agonist and SGLT-2 inhibitors were effective for glycemic and blood pressure control and for maintaining renal function. The GLP-1 receptor agonist improved diastolic function at 36 months.
Collapse
Affiliation(s)
- Takeyuki Hiramatsu
- Takeyuki Hiramatsu, Department of Nephrology, Aichi Welfare Cooperative Agricultural Federation, Konan-Kosei Hospital, 137 Omatsubara, Takaya-cho, Konan-city, Aichi 483-8704, Japan.
| | | | | | | | | | | |
Collapse
|
13
|
Anderson S, Cotiguala L, Tischer S, Park JM, McMurry K. Review of Newer Antidiabetic Agents for Diabetes Management in Kidney Transplant Recipients. Ann Pharmacother 2020; 55:496-508. [PMID: 32795145 DOI: 10.1177/1060028020951955] [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] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE This systematic review describes the efficacy, safety, and drug interactions of dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1 RAs), and sodium-glucose transport protein 2 (SGLT2) inhibitors in kidney transplant recipients (KTRs). DATA SOURCES Articles were identified by English-language MEDLINE search, published prior to May 2020, using the terms kidney transplant, OR PTDM, OR NODAT, AND metformin, OR DPP4, OR GLP1, OR SGLT2. STUDY SELECTION AND DATA EXTRACTION All selected studies were included if the study population was composed of adult KTRs who were diagnosed with either impaired glucose tolerance, diabetes mellitus (DM), new-onset diabetes after transplant (NODAT), or posttransplantation diabetes mellitus (PTDM). DATA SYNTHESIS In KTRs, there is evidence for safety with DPP-4 inhibitors, GLP-1 RAs, and SGLT2 inhibitors. However, urinary tract infections and a slight initial decrease in renal function may limit use of SGLT2 inhibitors. As compared with the nontransplant type 2 DM population, SGLT2 inhibitors are not as efficacious in KTRs. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE This review provides an overview of the current literature on newer antidiabetic agents, addressing efficacy, safety, and drug interactions to help guide clinical decision-making for their use in KTRs. CONCLUSION Newer antidiabetic agents have been recommended by the American Diabetes Association for potential cardiovascular, renal, and hypoglycemic benefits. Particular agents, such as DPP-4 inhibitors and GLP-1 RAs may play a role in correcting PTDM-related defects. Clinicians need to take into account both patient-specific and drug-specific characteristics when initiating these agents in KTRs.
Collapse
|
14
|
Cai Y, Liu X, Xu G. Combination therapy with SGLT2 inhibitors for diabetic kidney disease. Biomed Pharmacother 2020; 127:110192. [PMID: 32559844 DOI: 10.1016/j.biopha.2020.110192] [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: 03/22/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/25/2022] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a novel class of oral antihyperglycemic agents developed in recent years. They could block most glucose reabsorption in renal proximal tubules, thereby exerting glucose lowering effects through glycosuric ways. The renal and cardiovascular protection effects of SGLT2 inhibitors have also been demonstrated both in preclinical studies and clinical trials. However, SGLT2 inhibitors alone could induce an increase in endogenous/hepatic glucose production as well as in fasting plasma glucose levels; a sharp decrease of blood glucose concentration induced by SGLT2 inhibitors could also promote the secretion of counter-regulatory hormones such as glucagon, which has been reported to be associated with the occurrence of glycemic ketoacidosis. Therefore, coadministration of SGLT2 inhibitors and other antihyperglycemic agents should be considered when the therapeutic effect of SGLT2 inhibitors alone was unsatisfactory.
Collapse
Affiliation(s)
- Yuwen Cai
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China; Grade 2016, The Second Clinical Medical College of Nanchang University, Jiangxi, China
| | - Xin Liu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China.
| |
Collapse
|
15
|
Patel DM, Bose M, Cooper ME. Glucose and Blood Pressure-Dependent Pathways-The Progression of Diabetic Kidney Disease. Int J Mol Sci 2020; 21:ijms21062218. [PMID: 32210089 PMCID: PMC7139394 DOI: 10.3390/ijms21062218] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
The major clinical associations with the progression of diabetic kidney disease (DKD) are glycemic control and systemic hypertension. Recent studies have continued to emphasize vasoactive hormone pathways including aldosterone and endothelin which suggest a key role for vasoconstrictor pathways in promoting renal damage in diabetes. The role of glucose per se remains difficult to define in DKD but appears to involve key intermediates including reactive oxygen species (ROS) and dicarbonyls such as methylglyoxal which activate intracellular pathways to promote fibrosis and inflammation in the kidney. Recent studies have identified a novel molecular interaction between hemodynamic and metabolic pathways which could lead to new treatments for DKD. This should lead to a further improvement in the outlook of DKD building on positive results from RAAS blockade and more recently newer classes of glucose-lowering agents such as SGLT2 inhibitors and GLP1 receptor agonists.
Collapse
Affiliation(s)
- Devang M. Patel
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
- Correspondence: (D.M.P.); (M.E.C.)
| | - Madhura Bose
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
| | - Mark E. Cooper
- Department of Diabetes, Monash University Central, Clinical School, Melbourne, VIC 3004, Australia;
- Department of Endocrinology and Diabetes, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Correspondence: (D.M.P.); (M.E.C.)
| |
Collapse
|
16
|
Beitelshees AL, Leslie BR, Taylor SI. Sodium-Glucose Cotransporter 2 Inhibitors: A Case Study in Translational Research. Diabetes 2019; 68:1109-1120. [PMID: 31109940 PMCID: PMC6610013 DOI: 10.2337/dbi18-0006] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are the most recently approved class of diabetes drugs. Unlike other agents, SGLT2 inhibitors act on the kidney to promote urinary glucose excretion. SGLT2 inhibitors provide multiple benefits, including decreased HbA1c, body weight, and blood pressure. These drugs have received special attention because they decrease the risk of major adverse cardiovascular events and slow progression of diabetic kidney disease (1-3). Balanced against these impressive benefits, the U.S. Food and Drug Administration-approved prescribing information describes a long list of side effects: genitourinary infections, ketoacidosis, bone fractures, amputations, acute kidney injury, perineal necrotizing fasciitis, and hyperkalemia. This review provides a physiological perspective to understanding the multiple actions of these drugs complemented by a clinical perspective toward balancing benefits and risks.
Collapse
Affiliation(s)
- Amber L Beitelshees
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | | | - Simeon I Taylor
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
17
|
Tamargo J. Sodium-glucose Cotransporter 2 Inhibitors in Heart Failure: Potential Mechanisms of Action, Adverse Effects and Future Developments. Eur Cardiol 2019; 14:23-32. [PMID: 31131034 PMCID: PMC6523047 DOI: 10.15420/ecr.2018.34.2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heart failure is a common complication in patients with diabetes, and people with both conditions present a worse prognosis. Sodium–glucose cotransporter 2 inhibitors (SGLT2Is) increase urinary glucose excretion, improving glycaemic control. In type 2 diabetes (T2D), some SGLT2Is reduce major cardiovascular events, heart failure hospitalisations and worsening of kidney function independent of glycaemic control. Multiple mechanisms (haemodynamic, metabolic, hormonal and direct cardiac/renal effects) have been proposed to explain these cardiorenal benefits. SGLT2Is are generally well tolerated, but can produce rare serious adverse effects, and the benefit/risk ratio differs between SGLT2Is. This article analyses the mechanisms underlying the cardiorenal benefits and adverse effects of SGLT2Is in patients with T2D and heart failure and outlines some questions to be answered in the near future.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, CIBERCV Madrid, Spain
| |
Collapse
|
18
|
Klee NS, McCarthy CG, Lewis S, McKenzie JL, Vincent JE, Webb RC. Urothelial Senescence in the Pathophysiology of Diabetic Bladder Dysfunction-A Novel Hypothesis. Front Surg 2018; 5:72. [PMID: 30564582 PMCID: PMC6288180 DOI: 10.3389/fsurg.2018.00072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
Abstract
Diabetic bladder dysfunction (DBD) is a well-recognized and common symptom affecting up to 50% of all diabetic patients. DBD has a broad range of clinical presentations ranging from overactive to underactive bladder symptoms that develops in middle-aged to elderly patients with long standing and poorly controlled diabetes. Low efficacy of current therapeutics and lifestyle interventions combined with high national healthcare costs highlight the need for more research into bladder dysfunction pathophysiology and novel treatment options. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest induced by replicative exhaustion and damaging insults. While controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence is known to result in tissue dysfunction through enhanced secretion of inflammatory factors. This review presents previous scientific findings and current hypotheses that characterize diabetic bladder dysfunction. Further, we propose the novel hypothesis that cellular senescence within the urothelial layer of the bladder contributes to the pro-inflammatory/pro-oxidant environment and symptoms of diabetic bladder dysfunction. Our results show increased cellular senescence in the urothelial layer of the bladder; however, whether this phenomenon is the cause or effect of DBD is unknown. The urothelial layer of the bladder is made up of transitional epithelia specialized to contract and expand with demand and plays an active role in transmission by modulating afferent activity. Transition from normal functioning urothelial cells to secretory senescence cells would not only disrupt the barrier function of this layer but may result in altered signaling and sensation of bladder fullness; dysfunction of this layer is known to result in symptoms of frequency and urgency. Future DBD therapeutics may benefit from targeting and preventing early transition of urothelial cells to senescent cells.
Collapse
Affiliation(s)
- Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Steven Lewis
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jaine L McKenzie
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States.,Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Julie E Vincent
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - R Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
19
|
Zelniker TA, Braunwald E. Cardiac and Renal Effects of Sodium-Glucose Co-Transporter 2 Inhibitors in Diabetes. J Am Coll Cardiol 2018; 72:1845-1855. [DOI: 10.1016/j.jacc.2018.06.040] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
|
20
|
Keri KC, Samji NS, Blumenthal S. Diabetic nephropathy: newer therapeutic perspectives. J Community Hosp Intern Med Perspect 2018; 8:200-207. [PMID: 30181826 PMCID: PMC6116149 DOI: 10.1080/20009666.2018.1500423] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/29/2018] [Indexed: 12/24/2022] Open
Abstract
Diabetic nephropathy (DN is a dreaded consequence of diabetes mellitus, accounting for about 40% of end-stage renal disease (ESRD). It is responsible for significant morbidity and mortality, both directly by causing ESRD and indirectly by increasing cardiovascular risk. Extensive research in this field has thrown light on multiple pathways that can be pharmacologically targeted, to control or reverse the process of DN. Glomerulocentric approach of DN still continues to produce favourable results as evidenced by the recent data on SGLT-2 (sodium glucose co-transporter type 2) inhibitors. Beyond the glomerular mechanisms, numerous novel pathways have been discovered in the last decade. Some of these pathways target inflammatory and oxidative damage, while the others target more specific mechanisms such as AGE-RAGE (advanced glycation end products-receptors for advanced glycation end products), ASK (apoptotic signal-regulating kinase), and endothelin-associated pathways. As a result of the research, a handful of clinically relevant drugs have made it to the human trials which have been elucidated in the following review, bearing in the mind that there are many more to come over the next few years. Ongoing research is expected to inform the clinicians regarding the use of the newer drugs in DN. Abbreviations: USFDA: Unites States Food and Drug Administration; SGLT-2: Sodium glucose transporter type 2; GLP-1: Glucagon-like peptide-1; DDP-4: Dipeptidyl peptidase-4; UACR: urinary albumin creatinine ratio; eGFR: Estimated glomerular filtration rate; CKD: Chronic kidney disease; DN: Diabetic nephropathy; TGF: Tubuloglomerular feedback; RAAS: Renin angiotensin aldosterone system; T1DM: Type 1 diabetes mellitus; T2DM: Type 2 diabetes mellitus; RCT: Randomized controlled trial; AGE-RAGE: Advanced glycation end products-receptors for advanced glycation end products; ASK-1: Apoptotic signal-regulating kinase-1; Nrf-2: Nuclear 1 factor [erythroid derived-2]-related factor 2; ml/min/1.73m2: Millilitre/minute/1.73 square meters of body surface area; ~: Approximately.
Collapse
Affiliation(s)
- Krishna C Keri
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Naga S Samji
- Internal Medicine Department, Primary Care, Bellin Health, Marinette, WI, USA
| | - Samuel Blumenthal
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
21
|
Yaribeygi H, Butler AE, Atkin SL, Katsiki N, Sahebkar A. Sodium–glucose cotransporter 2 inhibitors and inflammation in chronic kidney disease: Possible molecular pathways. J Cell Physiol 2018; 234:223-230. [DOI: 10.1002/jcp.26851] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/10/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences Tehran Iran
| | | | | | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine Medical School, Aristotle University of Thessaloniki, Hippokration Hospital Thessaloniki Greece
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
22
|
Abstract
Sodium-glucose cotransporter 2 (SGLT2), which is specifically expressed on the apical side of proximal tubular cells, is involved in the reabsorption of most of the glucose filtered by the glomeruli, and its inhibitors are gaining publicity as potent antihyperglycemic drugs. In some clinical trials, SGLT2 inhibitors exerted cardiovascular and kidney protective effects, which appeared to be partly independent of the original glucose-lowering effect. SGLT2 inhibitors have both direct and indirect renoprotective effects. The direct effects involve the suppression of hyperplasia/hypertrophy, inflammation, and fibrosis in the proximal tubular cells, utilization of ketone bodies, restored tubuloglomerular feedback, decreased oxygen consumption, improvement in anemia, and preconditioning against ischemia/reperfusion. The indirect effects involve a reduction in insulin levels and resistance, uric acid concentration, body weight, and blood pressure. However, safety concerns remain, including consequences of an enhanced glucose load in the lower nephron, leg amputation, bone fractures, and therapeutic efficacy in patients with advanced chronic kidney disease.
Collapse
Affiliation(s)
- Marie Ito
- Division of Nephrology and Endocrinology, University of Tokyo School of Medicine, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, University of Tokyo School of Medicine, Japan
| |
Collapse
|
23
|
Mima A. Renal protection by sodium-glucose cotransporter 2 inhibitors and its underlying mechanisms in diabetic kidney disease. J Diabetes Complications 2018; 32:720-725. [PMID: 29880432 DOI: 10.1016/j.jdiacomp.2018.04.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022]
Abstract
AIM Diabetic kidney disease (DKD) is the most frequent cause of mortality and morbidity, leading a global health burden. This review will focus on the potential therapeutic interventions using Sodium-glucose cotransporter-2 (SGLT2) inhibitors that could prevent the development and progression of DKD. RESULTS SGLT2 inhibitors have been widely used as anti-diabetic drugs. Recent clinical studies have demonstrated that these drugs, which improve glycemic control and hypertension and decrease body weight, decrease the risk of renal function impairment and heart failure in patients with type 2 diabetes. With regard to long-term clinical outcomes, the Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes (EMPA-REG OUTCOME), the EMPA-REG Renal OUTCOME, and the CANagliflozin cardioVascular Assessment Study (CANVAS) program which have been integrated from CANVAS and CANVAS-Renal (CANVAS-R) trials reported significant risk reductions in primary combined major adverse cardiovascular events. Furthermore, regarding renal outcomes, the EMPA-REG Renal OUTCOME and CANVAS program clearly showed improvements in renal outcomes, including decreases in albuminuria and progression of nephropathy, doubling of serum creatinine levels, and initiation of renal replacement therapy. CONCLUSIONS Potential mechanisms of SGLT2 inhibitors related to renoprotection can be divided into two categories: hemodynamic actions and metabolic actions.
Collapse
Affiliation(s)
- Akira Mima
- Department of Nephrology, Kindai University Faculty of Medicine, Kindai University Nara Hospital, Nara, Japan.
| |
Collapse
|
24
|
Do effects of sodium-glucose cotransporter-2 inhibitors in patients with diabetes give insight into potential use in non-diabetic kidney disease? Curr Opin Nephrol Hypertens 2018; 26:358-367. [PMID: 28582367 DOI: 10.1097/mnh.0000000000000343] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Our aim was to review the rationale for the role of sodium-glucose cotransporter-2 inhibitors (SGLT-2i) as renoprotective therapy in patients with and without diabetes. RECENT FINDINGS SGLT-2i are antihyperglycemic agents, approved for treating type 2 diabetes to reduce glycosylated hemoglobin, type A1c. Primary glucoregulatory effects occur through selective inhibition of SGLT-2 at the renal proximal tubule promoting glucosuria leading to blood glucose lowering. From a hemodynamic perspective, SGLT-2 inhibition is also associated with decreased glomerular hyperfiltration, an effect that is mediated through natriuresis and tubuloglomerular feedback. With renal injury and progressive nephron loss, diabetic kidney disease, and nondiabetic chronic kidney diseases share overlapping phenotypes exhibiting single nephron hyperfiltration, along with increased proteinuria. Importantly, the impact of SGLT-2 inhibition on renal and systemic hemodynamic function, including effects on lowering blood pressure, hyperfiltration, and plasma volume, are independent of blood glucose lowering and instead are because of natriuresis. Accordingly, large clinical trials with SGLT-2i investigating the potential use of SGLT-2i in patients without diabetes are now underway. SUMMARY Based on prominent nonglycemic effects, the clinical use of SGLT-2i as renoprotective therapy may extend to nondiabetic chronic kidney diseases subtypes, which could help to address a large, unmet clinical need.
Collapse
|
25
|
Vallianou NG, Geladari E, Kazazis CE. SGLT-2 inhibitors: Their pleiotropic properties. Diabetes Metab Syndr 2017; 11:311-315. [PMID: 28011230 DOI: 10.1016/j.dsx.2016.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/08/2016] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes mellitus has become a global pandemic. Nowadays, it is estimated that approximately 415 million people all over the world have diabetes. The sodium glucose co-transporters 2 inhibitors are a new class of glucose-lowering agents, which act through a novel mechanism by producing a decline in glucose re-absorption in the kidney, thereby increasing glycosuria and decreasing serum glucose levels. Data suggest that apart from lowering HbA1c, they produce a small but significant weight loss and a small decrease in blood pressure. Also, they possess nephro-protective potential. These drugs are demonstrated to restore intra-glomerular pressure by increasing angiotensin (1-7), which exerts vasodilatory and anti-inflammatory effects. Their profile on cardiovascular events is still under investigation. In this review, the pleiotropic potential of this novel class of glucose-lowering levels will be discussed. Further research is warranted to determine their safety in the long term.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece.
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | | |
Collapse
|
26
|
Szablewski L. Distribution of glucose transporters in renal diseases. J Biomed Sci 2017; 24:64. [PMID: 28854935 PMCID: PMC5577680 DOI: 10.1186/s12929-017-0371-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 08/23/2017] [Indexed: 02/06/2023] Open
Abstract
Kidneys play an important role in glucose homeostasis. Renal gluconeogenesis prevents hypoglycemia by releasing glucose into the blood stream. Glucose homeostasis is also due, in part, to reabsorption and excretion of hexose in the kidney.Lipid bilayer of plasma membrane is impermeable for glucose, which is hydrophilic and soluble in water. Therefore, transport of glucose across the plasma membrane depends on carrier proteins expressed in the plasma membrane. In humans, there are three families of glucose transporters: GLUT proteins, sodium-dependent glucose transporters (SGLTs) and SWEET. In kidney, only GLUTs and SGLTs protein are expressed. Mutations within genes that code these proteins lead to different renal disorders and diseases. However, diseases, not only renal, such as diabetes, may damage expression and function of renal glucose transporters.
Collapse
Affiliation(s)
- Leszek Szablewski
- Medical University of Warsaw, Chair & Department of General Biology & Parasitology, Center for Biostructure Research, 5 Chalubinskiego Str., 02-004, Warsaw, Poland.
| |
Collapse
|
27
|
León Jiménez D, Castilla Guerra L, López Chozas JM, Miramontes González JP. Update concept of the dual blocking of the renin-angiotensin-aldosteron system. A new therapeutic option? Med Clin (Barc) 2017; 150:33-38. [PMID: 28668172 DOI: 10.1016/j.medcli.2017.05.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/26/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022]
Affiliation(s)
- David León Jiménez
- Unidad de Gestión Clínica de Medicina Interna, Hospital Universitario Virgen Macarena, Sevilla, España.
| | - Luis Castilla Guerra
- Unidad de Riesgo Vascular, Hospital Universitario Virgen Macarena, Sevilla, España
| | - José Manuel López Chozas
- Unidad de Gestión Clínica de Medicina Interna, Hospital Universitario Virgen Rocío, Sevilla, España
| | - José Pablo Miramontes González
- Servicio de Medicina Interna, Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, España
| |
Collapse
|
28
|
Zou H, Zhou B, Xu G. SGLT2 inhibitors: a novel choice for the combination therapy in diabetic kidney disease. Cardiovasc Diabetol 2017; 16:65. [PMID: 28511711 PMCID: PMC5434580 DOI: 10.1186/s12933-017-0547-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is the most common cause of end stage renal disease. The comprehensive management of DKD depends on combined target-therapies for hyperglycemia, hypertension, albuminuria, and hyperlipaemia, etc. Sodium-glucose co-transporter 2 (SGLT2) inhibitors, the most recently developed oral hypoglycemic agents acted on renal proximal tubules, suppress glucose reabsorption and increase urinary glucose excretion. Besides improvements in glycemic control, they presented excellent performances in direct renoprotective effects and the cardiovascular (CV) safety by decreasing albuminuria and the independent CV risk factors such as body weight and blood pressure, etc. Simultaneous use of SGLT-2 inhibitors and renin-angiotensin-aldosterone system (RAAS) blockers are novel strategies to slow the progression of DKD via reducing inflammatory and fibrotic markers induced by hyperglycaemia more than either drug alone. The available population and animal based studies have described SGLT2 inhibitors plus RAAS blockers. The present review was to systematically review the potential renal benefits of SGLT2 inhibitors combined with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, mineralocorticoid receptor antagonists, and especially the angiotensin-converting enzyme inhibitors/angiotensin receptor blockers.
Collapse
Affiliation(s)
- Honghong Zou
- Medical Center of the Graduate School, Nanchang University, Nanchang, People's Republic of China.,Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 1, Minde Road, Donghu District, Nanchang, Zip Code: 330006, People's Republic of China
| | - Baoqin Zhou
- Medical Center of the Graduate School, Nanchang University, Nanchang, People's Republic of China.,Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 1, Minde Road, Donghu District, Nanchang, Zip Code: 330006, People's Republic of China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 1, Minde Road, Donghu District, Nanchang, Zip Code: 330006, People's Republic of China.
| |
Collapse
|
29
|
Wang XX, Levi J, Luo Y, Myakala K, Herman-Edelstein M, Qiu L, Wang D, Peng Y, Grenz A, Lucia S, Dobrinskikh E, D'Agati VD, Koepsell H, Kopp JB, Rosenberg AZ, Levi M. SGLT2 Protein Expression Is Increased in Human Diabetic Nephropathy: SGLT2 PROTEIN INHIBITION DECREASES RENAL LIPID ACCUMULATION, INFLAMMATION, AND THE DEVELOPMENT OF NEPHROPATHY IN DIABETIC MICE. J Biol Chem 2017; 292:5335-5348. [PMID: 28196866 DOI: 10.1074/jbc.m117.779520] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Indexed: 12/21/2022] Open
Abstract
There is very limited human renal sodium gradient-dependent glucose transporter protein (SGLT2) mRNA and protein expression data reported in the literature. The first aim of this study was to determine SGLT2 mRNA and protein levels in human and animal models of diabetic nephropathy. We have found that the expression of SGLT2 mRNA and protein is increased in renal biopsies from human subjects with diabetic nephropathy. This is in contrast to db-db mice that had no changes in renal SGLT2 protein expression. Furthermore, the effect of SGLT2 inhibition on renal lipid content and inflammation is not known. The second aim of this study was to determine the potential mechanisms of beneficial effects of SGLT2 inhibition in the progression of diabetic renal disease. We treated db/db mice with a selective SGLT2 inhibitor JNJ 39933673. We found that SGLT2 inhibition caused marked decreases in systolic blood pressure, kidney weight/body weight ratio, urinary albumin, and urinary thiobarbituric acid-reacting substances. SGLT2 inhibition prevented renal lipid accumulation via inhibition of carbohydrate-responsive element-binding protein-β, pyruvate kinase L, SCD-1, and DGAT1, key transcriptional factors and enzymes that mediate fatty acid and triglyceride synthesis. SGLT2 inhibition also prevented inflammation via inhibition of CD68 macrophage accumulation and expression of p65, TLR4, MCP-1, and osteopontin. These effects were associated with reduced mesangial expansion, accumulation of the extracellular matrix proteins fibronectin and type IV collagen, and loss of podocyte markers WT1 and synaptopodin, as determined by immunofluorescence microscopy. In summary, our study showed that SGLT2 inhibition modulates renal lipid metabolism and inflammation and prevents the development of nephropathy in db/db mice.
Collapse
Affiliation(s)
- Xiaoxin X Wang
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220.,the Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado 80220
| | - Jonathan Levi
- the NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhuan Luo
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Komuraiah Myakala
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Michal Herman-Edelstein
- the Rabin Medical Center, Department of Nephrology and Hypertension, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Liru Qiu
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Dong Wang
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Yingqiong Peng
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Almut Grenz
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Scott Lucia
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Evgenia Dobrinskikh
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220
| | - Vivette D D'Agati
- the Department of Pathology, Columbia University, College of Physicians and Surgeons, New York, New York 10027
| | - Hermann Koepsell
- the Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, D-97082 Würzburg, Germany, and
| | - Jeffrey B Kopp
- the NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Avi Z Rosenberg
- the Department of Pathology, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Moshe Levi
- From the Departments of Medicine, Anesthesiology, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80220, .,the Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado 80220
| |
Collapse
|
30
|
Mudaliar S, Alloju S, Henry RR. Can a Shift in Fuel Energetics Explain the Beneficial Cardiorenal Outcomes in the EMPA-REG OUTCOME Study? A Unifying Hypothesis. Diabetes Care 2016; 39:1115-22. [PMID: 27289124 DOI: 10.2337/dc16-0542] [Citation(s) in RCA: 432] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/29/2016] [Indexed: 02/03/2023]
Abstract
Type 2 diabetes mellitus causes excessive morbidity and premature cardiovascular (CV) mortality. Although tight glycemic control improves microvascular complications, its effects on macrovascular complications are unclear. The recent publication of the EMPA-REG OUTCOME study documenting impressive benefits with empagliflozin (a sodium-glucose cotransporter 2 [SGLT2] inhibitor) on CV and all-cause mortality and hospitalization for heart failure without any effects on classic atherothrombotic events is puzzling. More puzzling is that the curves for heart failure hospitalization, renal outcomes, and CV mortality begin to separate widely within 3 months and are maintained for >3 years. Modest improvements in glycemic, lipid, or blood pressure control unlikely contributed significantly to the beneficial cardiorenal outcomes within 3 months. Other known effects of SGLT2 inhibitors on visceral adiposity, vascular endothelium, natriuresis, and neurohormonal mechanisms are also unlikely major contributors to the CV/renal benefits. We postulate that the cardiorenal benefits of empagliflozin are due to a shift in myocardial and renal fuel metabolism away from fat and glucose oxidation, which are energy inefficient in the setting of the type 2 diabetic heart and kidney, toward an energy-efficient super fuel like ketone bodies, which improve myocardial/renal work efficiency and function. Even small beneficial changes in energetics minute to minute translate into large differences in efficiency, and improved cardiorenal outcomes over weeks to months continue to be sustained. Well-planned physiologic and imaging studies need to be done to characterize fuel energetics-based mechanisms for the CV/renal benefits.
Collapse
Affiliation(s)
- Sunder Mudaliar
- Veterans Affairs Medical Center and University of California, San Diego School of Medicine, San Diego, CA
| | - Sindura Alloju
- Veterans Affairs Medical Center and University of California, San Diego School of Medicine, San Diego, CA
| | - Robert R Henry
- Veterans Affairs Medical Center and University of California, San Diego School of Medicine, San Diego, CA
| |
Collapse
|
31
|
Lam KSL, Chow CC, Tan KCB, Ma RCW, Kong APS, Tong PCY, Tsang MW, Chan TM, Tang SCW, Lee KK, So WY, Tomlinson B. Practical considerations for the use of sodium-glucose co-transporter type 2 inhibitors in treating hyperglycemia in type 2 diabetes. Curr Med Res Opin 2016; 32:1097-108. [PMID: 26933918 DOI: 10.1185/03007995.2016.1161608] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Sodium-glucose co-transporter type 2 (SGLT2) inhibitors are a new class of oral anti-diabetic agents with a unique, insulin-independent mode of action. In patients with diabetes who have adequate renal function, SGLT2 inhibitors reduce hyperglycemia by blocking renal glucose reabsorption and increasing urinary glucose excretion. These agents are indicated for the treatment of hyperglycemia in type 2 diabetes mellitus (T2DM), as an adjunct to diet and exercise. In terms of efficacy, they are comparable to most other oral agents, and carry a low risk of hypoglycemia unless combined with sulfonylureas or insulin. They may be used in combination regimens with metformin, sulfonylureas, or insulin. Beyond glucose lowering, SGLT2 inhibitors are associated with modest weight loss and mild anti-hypertensive effects. Emerging cardiovascular and renal outcomes data suggest other potentially beneficial non-glycemic effects, although these findings await confirmation from further studies. The main adverse effects are increased risk of volume depletion and of genitourinary infections, although these can be managed with standard interventions. Rare cases of euglycemic ketoacidosis have been reported in a subset of patients treated with these agents, an issue currently under investigation. SGLT2 inhibitors represent a promising alternative treatment option for T2DM patients in whom the effectiveness of oral anti-hyperglycemic therapy is limited by the risk of hypoglycemia, weight gain, or other adverse effects. Safety and efficacy (up to 4 years) have been demonstrated in a range of T2DM patient populations, although more studies will be needed to determine whether treatment with SGLT2 inhibitors improves patient-important outcomes in the longer term.
Collapse
Affiliation(s)
- Karen S L Lam
- a Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong SAR , China
| | - Chun Chung Chow
- b Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong SAR, China
| | - Kathryn C B Tan
- a Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong SAR , China
| | - Ronald C W Ma
- b Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong SAR, China
| | - Alice P S Kong
- b Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong SAR, China
| | - Peter C Y Tong
- c Qualigenics Diabetes Centre, The Chinese University of Hong Kong , Hong Kong SAR , China
| | | | - Tak Mao Chan
- a Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong SAR , China
| | - Sydney C W Tang
- a Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong SAR , China
| | - Ka Kui Lee
- a Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong SAR , China
| | - Wing Yee So
- b Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong SAR, China
| | - Brian Tomlinson
- b Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong SAR, China
| |
Collapse
|
32
|
Lytvyn Y, Bjornstad P, Pun N, Cherney DZI. New and old agents in the management of diabetic nephropathy. Curr Opin Nephrol Hypertens 2016; 25:232-9. [PMID: 26890303 PMCID: PMC5841607 DOI: 10.1097/mnh.0000000000000214] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Diabetic nephropathy is a long-standing complication of diabetes mellitus and is responsible for more than 40% of end-stage renal disease cases in developed countries. Unfortunately, conventional renin-angiotensin-aldosterone system (RAAS) inhibitor medications only partially protect against the development and progression of diabetic nephropathy. Moreover, RAAS inhibitors have failed as primary prevention therapy in type 1 diabetes. Thus, agents targeting alternative pathogenic mechanisms leading to diabetic nephropathy have been intensively investigated, which is the topic of this review. RECENT FINDINGS Promising emerging agents have targeted neurohormonal activation (alternative components of the RAAS and neprilysin inhibition), tubuloglomerular feedback mechanisms (sodium glucose cotransporter 2 inhibition and incretin-based therapy) and renal inflammation/fibrosis. SUMMARY Evidence demonstrating the potential of these agents to protect and prevent progression of diabetic nephropathy is summarized in this review. There are dedicated clinical trials ongoing with these therapies, which have the potential to change the clinical practice.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- aDivision of Nephrology, Department of Medicine, University Health Network bDepartment of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada cDepartment of Pediatric Endocrinology, University of Colorado School of Medicine dBarbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, USA *Drs Lytvyn, Bjornstad and Pun are the co-first authors of the article
| | | | | | | |
Collapse
|
33
|
Stinkens K, Mathieu C. Sodium-glucose Cotransporter 2 Inhibitors and Ketoacidosis - Clinical Implications in the Treatment of Patients with Type 2 Diabetes. EUROPEAN ENDOCRINOLOGY 2016; 12:33-34. [PMID: 29632585 PMCID: PMC5813456 DOI: 10.17925/ee.2016.12.01.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 03/10/2016] [Indexed: 11/25/2022]
Abstract
The use of sodium-glucose cotransporter 2 inhibitors is associated with an increased risk of diabetic ketoacidosis. This risk has been reported in particular in off-label use of these agents in type 1 diabetes, but reports of risks in type 2 diabetes patients also exist. In type 2 diabetes ketoacidosis is rare and is present particularly in patients who have undergone prolonged starvation, serious infection, alcohol abuse or surgery. The pleiotropic advantages of sodium-glucose cotransporter 2 inhibitors do not outweigh the risk for a diabetic ketoacidosis, but caution is warranted.
Collapse
Affiliation(s)
- Kirsten Stinkens
- Department of Endocrinology UZ Leuven, UZ Gasthuisberg, Leuven, Belgium
| | - Chantal Mathieu
- Department of Endocrinology UZ Leuven, UZ Gasthuisberg, Leuven, Belgium
| |
Collapse
|
34
|
Pharmacokinetics, Pharmacodynamics and Clinical Use of SGLT2 Inhibitors in Patients with Type 2 Diabetes Mellitus and Chronic Kidney Disease. Clin Pharmacokinet 2016; 54:691-708. [PMID: 25805666 DOI: 10.1007/s40262-015-0264-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibitors of sodium-glucose cotransporters type 2 (SGLT2) are proposed as a novel approach for the management of type 2 diabetes mellitus. SGLT2 cotransporters are responsible for reabsorption of 90 % of the glucose filtered by the kidney. The glucuretic effect resulting from SGLT2 inhibition contributes to reduce hyperglycaemia and also assists weight loss and blood pressure reduction. Several SGLT2 inhibitors are already available in many countries (dapagliflozin, canagliflozin, empagliflozin) and in Japan (ipragliflozin, tofogliflozin). These SGLT2 inhibitors share similar pharmacokinetic characteristics with a rapid oral absorption, a long elimination half-life allowing once-daily administration, an extensive hepatic metabolism mainly via glucuronidation to inactive metabolites and a low renal elimination as a parent drug. Pharmacokinetic parameters are slightly altered in the case of chronic kidney disease (CKD). While no dose adjustment is required in the case of mild CKD, SGLT2 inhibitors may not be used or only at a lower daily dose in patients with moderate CKD. Furthermore, the pharmacodynamic response to SGLT2 inhibitors as assessed by urinary glucose excretion declines with increasing severity of renal impairment as assessed by a reduction in the estimated glomerular filtration rate. Nevertheless, the glucose-lowering efficacy and safety of SGLT2 inhibitors are almost comparable in patients with mild CKD as in patients with normal kidney function. In patients with moderate CKD, the efficacy tends to be dampened and safety concerns may occur. In patients with severe CKD, the use of SGLT2 inhibitors is contraindicated. Thus, prescribing information should be consulted regarding dosage adjustments or restrictions in the case of renal dysfunction for each SGLT2 inhibitor. The clinical impact of SGLT2 inhibitors on renal function and their potential to influence the course of diabetic nephropathy deserve attention because of preliminary favourable results in animal models.
Collapse
|
35
|
John M, Gopinath D, Jagesh R. Sodium-glucose cotransporter 2 inhibitors with insulin in type 2 diabetes: Clinical perspectives. Indian J Endocrinol Metab 2016; 20:22-31. [PMID: 26904465 PMCID: PMC4743379 DOI: 10.4103/2230-8210.172268] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The treatment of type 2 diabetes is a challenging problem. Most subjects with type 2 diabetes have progression of beta cell failure necessitating the addition of multiple antidiabetic agents and eventually use of insulin. Intensification of insulin leads to weight gain and increased risk of hypoglycemia. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a class of antihyperglycemic agents which act by blocking the SGLT2 in the proximal tubule of the kidney. They have potential benefits in terms of weight loss and reduction of blood pressure in addition to improvements in glycemic control. Further, one of the SGLT2 inhibitors, empagliflozin has proven benefits in reducing adverse cardiovascular (CV) outcomes in a CV outcome trial. Adding SGLT2 inhibitors to insulin in subjects with type 2 diabetes produced favorable effects on glycemic control without the weight gain and hypoglycemic risks associated with insulin therapy. The general risks of increased genital mycotic infections, urinary tract infections, volume, and osmosis-related adverse effects in these subjects were similar to the pooled data of individual SGLT2 inhibitors. There are subsets of subjects with type 2 diabetes who may have insulin deficiency, beta cell autoimmunity, or is prone to diabetic ketoacidosis. In these subjects, SGLT2 inhibitors should be used with caution to prevent the rare risks of ketoacidosis.
Collapse
Affiliation(s)
- Mathew John
- Department of Endocrinology and Diabetes, Providence Endocrine and Diabetes Specialty Centre, Thiruvananthapuram, Kerala, India
| | - Deepa Gopinath
- Department of Endocrinology and Diabetes, Providence Endocrine and Diabetes Specialty Centre, Thiruvananthapuram, Kerala, India
| | - Rejitha Jagesh
- Department of Endocrinology and Diabetes, Providence Endocrine and Diabetes Specialty Centre, Thiruvananthapuram, Kerala, India
| |
Collapse
|
36
|
Mohamed Assem SM, Nadia MSA, Sara AMA. Effect of canagliflozin or metformin on metabolic disorders in obese diabetic rats. ACTA ACUST UNITED AC 2015. [DOI: 10.5897/ajpp2015.4455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
37
|
Pharmacodynamics, efficacy and safety of sodium-glucose co-transporter type 2 (SGLT2) inhibitors for the treatment of type 2 diabetes mellitus. Drugs 2015; 75:33-59. [PMID: 25488697 DOI: 10.1007/s40265-014-0337-y] [Citation(s) in RCA: 358] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inhibitors of sodium-glucose co-transporter type 2 (SGLT2) are proposed as a novel approach for the management of type 2 diabetes mellitus (T2DM). Several compounds are already available in many countries (dapagliflozin, canagliflozin, empagliflozin and ipragliflozin) and some others are in a late phase of development. The available SGLT2 inhibitors share similar pharmacokinetic characteristics, with a rapid oral absorption, a long elimination half-life allowing once-daily administration, an extensive hepatic metabolism mainly via glucuronidation to inactive metabolites, the absence of clinically relevant drug-drug interactions and a low renal elimination as parent drug. SGLT2 co-transporters are responsible for reabsorption of most (90 %) of the glucose filtered by the kidneys. The pharmacological inhibition of SGLT2 co-transporters reduces hyperglycaemia by decreasing renal glucose threshold and thereby increasing urinary glucose excretion. The amount of glucose excreted in the urine depends on both the level of hyperglycaemia and the glomerular filtration rate. Results of numerous placebo-controlled randomised clinical trials of 12-104 weeks duration have shown significant reductions in glycated haemoglobin (HbA1c), resulting in a significant increase in the proportion of patients reaching HbA1c targets, and a significant lowering of fasting plasma glucose when SGLT2 inhibitors were administered as monotherapy or in addition to other glucose-lowering therapies including insulin in patients with T2DM. In head-to-head trials of up to 2 years, SGLT2 inhibitors exerted similar glucose-lowering activity to metformin, sulphonylureas or sitagliptin. The durability of the glucose-lowering effect of SGLT2 inhibitors appears to be better; however, this remains to be more extensively investigated. The risk of hypoglycaemia was much lower with SGLT2 inhibitors than with sulphonylureas and was similarly low as that reported with metformin, pioglitazone or sitagliptin. Increased renal glucose elimination also assists weight loss and could help to reduce blood pressure. Both effects were very consistent across the trials and they represent some advantages for SGLT2 inhibitors when compared with other oral glucose-lowering agents. The pharmacodynamic response to SGLT2 inhibitors declines with increasing severity of renal impairment, and prescribing information for each SGLT2 inhibitor should be consulted regarding dosage adjustments or restrictions in moderate to severe renal dysfunction. Caution is also recommended in the elderly population because of a higher risk of renal impairment, orthostatic hypotension and dehydration, even if the absence of hypoglycaemia represents an obvious advantage in this population. The overall effect of SGLT2 inhibitors on the risk of cardiovascular disease is unknown and will be evaluated in several ongoing prospective placebo-controlled trials with cardiovascular outcomes. The impact of SGLT2 inhibitors on renal function and their potential to influence the course of diabetic nephropathy also deserve more attention. SGLT2 inhibitors are generally well-tolerated. The most frequently reported adverse events are female genital mycotic infections, while urinary tract infections are less commonly observed and generally benign. In conclusion, with their unique mechanism of action that is independent of insulin secretion and action, SGLT2 inhibitors are a useful addition to the therapeutic options available for the management of T2DM at any stage in the natural history of the disease. Although SGLT2 inhibitors have already been extensively investigated, further studies should even better delineate the best place of these new glucose-lowering agents in the already rich armamentarium for the management of T2DM.
Collapse
|
38
|
Michel MC, Mayoux E, Vallon V. A comprehensive review of the pharmacodynamics of the SGLT2 inhibitor empagliflozin in animals and humans. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 388:801-16. [PMID: 26108304 PMCID: PMC5896322 DOI: 10.1007/s00210-015-1134-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 05/18/2015] [Indexed: 12/16/2022]
Abstract
Empagliflozin (formerly known as BI 10773) is a potent, competitive, and selective inhibitor of the sodium glucose transporter SGLT2, which mediates glucose reabsorption in the early proximal tubule and most of the glucose reabsorption by the kidney, overall. Accordingly, empagliflozin treatment increased urinary glucose excretion. This has been observed across multiple species including humans and was reported under euglycemic conditions, in obesity and, most importantly, in type 2 diabetic patients and multiple animal models of type 2 diabetes and of type 1 diabetes. This led to a reduction in blood glucose, smaller blood glucose excursions during oral glucose tolerance tests, and, upon chronic treatment, a reduction in HbA1c in animal models and patients. In rodents, such effects were observed in early and late phases of experimental diabetes and were associated with preservation of pancreatic β-cell function. Combination studies in animals demonstrated that beneficial metabolic effects of empagliflozin may also manifest when added to other types of anti-hyperglycemic treatments including linagliptin and pioglitazone. While some anti-hyperglycemic drugs lead to weight gain, empagliflozin treatment was associated with reduced body weight in normoglycemic obese and non-obese animals despite an increased food intake, largely due to a loss of adipose tissue; on the other hand, empagliflozin preserved body weight in models of type 1 diabetes. Empagliflozin improved endothelial dysfunction in diabetic rats and arterial stiffness, reduced blood pressure in diabetic patients, and attenuated early signs of nephropathy in diabetic animal models. Taken together, the SGLT2 inhibitor empagliflozin improves glucose metabolism by enhancing urinary glucose excretion; upon chronic administration, at least in animal models, the reductions in blood glucose levels are associated with beneficial effects on cardiovascular and renal complications of diabetes.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | | | | |
Collapse
|
39
|
Abstract
In type 2 diabetes (T2DM), glycaemic control delays the development and slows the progression of complications. Although there are numerous glucose-lowering agents in clinical use, only approximately half of T2DM patients achieve glycaemic control, while undesirable side-effects, such as hypoglycaemia and body weight gain, often impede treatment in those taking these medications. Thus, there is a need for novel agents and treatment options. Sodium-glucose cotransporter-2 inhibitors (SGLT-2-i) have recently been developed for the treatment of T2DM. The available data suggest a good tolerability profile for the three available drugs - canagliflozin, dapagliflozin and empagliflozin - approved by the US Food and Drug Administration (FDA) for the American market as well as in other countries. The most frequently reported adverse events with SGLT-2-i are female genital mycotic infections, urinary tract infections and increased urination. The pharmacodynamic response to SGLT-2-i declines with increasing severity of renal impairment, requiring dosage adjustments or restrictions with moderate-to-severe renal dysfunction. Most patients treated with SGLT-2-i also have a modest reduction in blood pressure and modest effects on serum lipid profiles, some of which are beneficial (increased high-density lipoprotein cholesterol and decreased triglycerides) and others which are not (increased low-density lipoprotein cholesterol, LDL-C). A number of large-scale and longer-term cardiovascular trials are now ongoing. In patients treated with dapagliflozin, a non-significant excess number of breast and bladder cancers has been reported; considered as due to a bias, this is nevertheless being followed in the ongoing trials. No other significant safety issues have been reported so far. Although there is some benefit for several cardiovascular risk factors such as HbA1c, high blood pressure, obesity and increases in LDL-C, adequately powered trials are still required to determine the effects of SGLT-2-i on macrovascular outcomes.
Collapse
Affiliation(s)
- S Halimi
- Scientific University Joseph-Fourier, and Diabetology Department Pavillon les Écrins, BP 217X, University Hospital Grenoble, 38043 Grenoble Cedex, France
| | - B Vergès
- Department of Endocrinology and Diabetology, University Hospital Dijon, and INSERM CRI 866, Dijon, France
| |
Collapse
|
40
|
Gnudi L, Karalliedde J. Beat it early: putative renoprotective haemodynamic effects of oral hypoglycaemic agents. Nephrol Dial Transplant 2015; 31:1036-43. [PMID: 25858586 PMCID: PMC4917060 DOI: 10.1093/ndt/gfv093] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/07/2015] [Indexed: 12/25/2022] Open
Abstract
Diabetic kidney disease represents a considerable burden; around one-third of patients with type 2 diabetes develop chronic kidney disease. In health, the kidneys play an important role in the regulation of glucose homeostasis via glucose utilization, gluconeogenesis and glucose reabsorption. In patients with diabetes, renal glucose homeostasis is significantly altered with an increase in both gluconeogenesis and renal tubular reabsorption of glucose. Environmental factors, both metabolic (hyperglycaemia, obesity and dyslipidaemia) and haemodynamic, together with a genetic susceptibility, lead to the activation of pro-oxidative, pro-inflammatory and pro-fibrotic pathways resulting in kidney damage. Hyperfiltration and its haemodynamic-driven insult to the kidney glomeruli is an important player in proteinuria and progression of kidney disease towards end-stage renal failure. Control of glycaemia and blood pressure are the mainstays to prevent kidney damage and slow its progression. There is emerging evidence that some hypoglycaemic agents may have renoprotective effects which are independent of their glucose-lowering effects. Sodium-glucose co-transporter-2 (SGLT-2) inhibitors may exert a renoprotective effect by a number of mechanisms including restoring the tubuloglomerular feedback mechanism and lowering glomerular hyperfiltration, reducing inflammatory and fibrotic markers induced by hyperglycaemia thus limiting renal damage. Simultaneous use of an SGLT-2 inhibitor and blockade of the renin-angiotensin-aldosterone system may be a strategy to slow progression of diabetic nephropathy more than either drug alone. The use of dipeptidyl peptidase-4 inhibitors and glucagon-like peptide 1 receptor agonists may exert a renoprotective effect by reducing inflammation, fibrosis and blood pressure. Given the burden of diabetic kidney disease, any additional renoprotective benefit with hypoglycaemic therapy is to be welcomed. Large randomized controlled trials are currently underway investigating if these new anti-diabetic agents can provide renoprotection in diabetes.
Collapse
Affiliation(s)
- Luigi Gnudi
- Cardiovascular Division, Department of Diabetes and Endocrinology, School of Life Science & Medicine, King's College, London, UK
| | - Janaka Karalliedde
- Cardiovascular Division, Department of Diabetes and Endocrinology, School of Life Science & Medicine, King's College, London, UK
| |
Collapse
|
41
|
White JR. Empagliflozin, an SGLT2 Inhibitor for the Treatment of Type 2 Diabetes Mellitus. Ann Pharmacother 2015; 49:582-98. [DOI: 10.1177/1060028015573564] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: To review available studies of empagliflozin, a sodium glucose co-transporter-2 (SGLT2) inhibitor approved in 2014 by the European Commission and the United States Food and Drug Administration for the treatment of type 2 diabetes mellitus (T2DM). Data Sources: PubMed was searched using the search terms empagliflozin, BI 10773, and BI10773, for entries between January 1, 2000, and December 1, 2014. Reference lists from retrieved articles were searched manually for additional peer-reviewed publications. Study Selection and Data Extraction: All publications reporting clinical trials of empagliflozin were eligible for inclusion. Data Synthesis: Empagliflozin is a new once-daily oral SGLT2 inhibitor with a mechanism of action that is independent of β-cell function and the insulin pathway. Data from a comprehensive phase III clinical trial program have demonstrated its efficacy as monotherapy, as add-on to other glucose-lowering agents, and in different patient populations. In these studies, empagliflozin resulted in improvements in blood glucose levels as well as reductions in body weight and blood pressure. Empagliflozin was well tolerated and was not associated with an increased risk of hypoglycemia versus placebo. Conclusion: The oral antidiabetes agent, empagliflozin, can be used as monotherapy or alongside other glucose-lowering treatments, including insulin, to treat T2DM.
Collapse
|
42
|
Lytvyn Y, Perkins BA, Cherney DZI. Uric acid as a biomarker and a therapeutic target in diabetes. Can J Diabetes 2015; 39:239-46. [PMID: 25600084 DOI: 10.1016/j.jcjd.2014.10.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/21/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy is a long-standing microvascular complication of diabetes mellitus and is the leading cause of end stage renal disease in developed countries. Current therapeutic strategies used to prevent or delay diabetic nephropathy exert limited clinical protective effects and can have serious adverse effects. Thus, identification of new pharmacologic agents that protect against the initiation and progression of complications of diabetes is of the utmost importance. Uric acid (UA) recently emerged as an inflammatory factor that increases oxidative stress and promotes activation of the renin angiotensin aldosterone system. As a consequence, higher UA levels are associated with various stages of the onset and progression of diabetic nephropathy, including metabolic, cardiovascular and kidney function abnormalities. If UA-lowering drugs, such as the xanthine oxidase inhibitors, block the mechanisms responsible for micro- and macrovascular injury in diabetes, these agents could represent a critical step toward preventing the progression of diabetes. This review focuses on the evidence that supports serum UA levels as a biomarker of renal and cardiovascular risk and as a potential additional therapeutic target in diabetes.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Bruce A Perkins
- Department of Medicine, Division of Endocrinology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Abstract
SGLT2 inhibition offers a novel mechanism to mitigate hyperglycemia in patients with diabetes and the introduction of SGLT2 has added a significant new tool to the antihyperglycemic armamentarium. At present, 2 agents are approved for use in the United States and several more are in development. SGLT2 inhibitors are generally associated with a reduction in A1C of between 0.5% and 1%. SGLT2 inhibitors are associated with an increased incidence of urinary tract and genital infections but these infections are typically mild, responsive to treatment, and are not use limiting.
Collapse
Affiliation(s)
- John R White
- Department of Pharmacotherapy, Washington State University, Health Sciences Building, Suite 210, PO Box 1459, Spokane, WA 99210, USA.
| |
Collapse
|
44
|
Cherney DZ, Perkins BA. Sodium-Glucose Cotransporter 2 Inhibition in Type 1 Diabetes: Simultaneous Glucose Lowering and Renal Protection? Can J Diabetes 2014; 38:356-63. [DOI: 10.1016/j.jcjd.2014.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/30/2014] [Accepted: 05/09/2014] [Indexed: 01/10/2023]
|