1
|
Gali S, Kundu A, Sharma S, Ahn MY, Puia Z, Kumar V, Kim IS, Kwak JH, Palit P, Kim HS. Therapeutic potential of bark extracts from Macaranga denticulata on renal fibrosis in streptozotocin-induced diabetic rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:911-933. [PMID: 39306745 DOI: 10.1080/15287394.2024.2394586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Macaranga denticulata (MD) bark is commonly utilized in traditional medicine for diabetes prevention and treatment. The bark extract of MD is rich in prenyl or farnesyl flavonoids and stilbenes, which possess antioxidant properties. Although data suggest the potential therapeutic benefits of the use of MD in treating diabetic nephropathy (DN), the precise mechanisms underlying MD-initiated protective effects against DN are not well understood. This study aimed to assess the renoprotective properties of MD extract by examining renofibrosis inhibition, oxidative stress, and inflammation utilizing streptozotocin-induced DN male Sprague - Dawley rats. Diabetic rats were intraperitoneally injected with streptozotocin (STZ) to induce diabetes. After 6 days, these rats were orally administered MD extract (200 mg/kg/day) or metformin (200 mg/kg/day) for 14 days. The administration of MD extract significantly lowered blood glucose levels, restored body weight, and reduced urine levels of various biomarkers associated with kidney functions. Histopathological analysis revealed protective effects in both kidneys and pancreas. Further, MD extract significantly restored abnormalities in advanced glycation end products, oxidative stress biomarkers, and proinflammatory cytokine levels in STZ-treated rats. MD extract markedly reduced renal fibrosis biomarker levels, indicating recovery from renal injury, and reversed dysregulation of sirtuins and claudin-1 in the kidneys of rats with STZ-induced diabetes. In conclusion, data demonstrated the renoprotective role of MD extract, indicating plant extract's ability to suppress oxidative stress and regulate proinflammatory pathways during pathological changes in diabetic nephropathy.
Collapse
Affiliation(s)
- Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Amit Kundu
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
- Department of Pharmacology, GITAM School of Pharmacy, GITAM Deemed to be University, Visakhapatnam, India
| | - Swati Sharma
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Mee-Young Ahn
- Department of Biochemistry and Health Science, Changwon National University, Changwon-si, Republic of Korea
| | - Zothan Puia
- Department of Pharmacy, Regional Institute of Paramedical & Nursing Sciences, Aizawl, India
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom Institute of Agriculture, Technology & Sciences, Allahabad, India
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Jeong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Partha Palit
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Assam University, Silchar, India
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| |
Collapse
|
2
|
Tetti M, Burrello J, Hureaux M, Billon C, Clauser E, Veglio F, Rabbia F, Pasini B, Crisetti A, Jeunemaitre X, Mulatero P, Monticone S. Prevalence of Hyperkalemia and Familial Hyperkalemic Hypertension in 5100 Patients Referred to a Tertiary Hypertension Unit. Hypertension 2024; 81:2275-2285. [PMID: 39229746 DOI: 10.1161/hypertensionaha.124.23500] [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: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Hyperkalemia is a frequent electrolyte alteration whose prevalence varies widely, depending on the adopted cutoff, the setting (inpatients versus outpatients), and the characteristics of the study population. Familial hyperkalemic hypertension (FHH) is a rare cause of hypertension, hyperkalemia, and hyperchloremic metabolic acidosis. METHODS In this retrospective observational study, we investigated the prevalence of hyperkalemia (serum K+ >5.2 mmol/L on 2 repeated measurements) in 5100 referred patients affected by arterial hypertension, the potential causes, and the associated cardiovascular risk profile. RESULTS Overall, 374 (7.3%) patients had hyperkalemia. This was associated with drugs known to increase K+ levels (74.6%), chronic kidney disease (33.7%), or both (24.3%). Among the 60 patients with unexplained hyperkalemia, 3 displayed a clinical and biochemical phenotype suggestive of FHH that was genetically confirmed in 2 of them (0.04% in the entire cohort). FHH prevalence rose to 3.3% in patients with unexplained hyperkalemia and up to 29% (2/7) if they had serum K+>5.8 mmol/L. The genetic cause of FHH was a missense variant affecting the acidic motif of WNK1 in 1 family and a rare CUL3 splicing variant, whose functional significance was confirmed by a minigene assay, in another. Finally, we observed a significant association between hyperkalemia and the occurrence of cardiovascular events, metabolic syndrome, and organ damage, independent of potential confounding factors. CONCLUSIONS The identification of hyperkalemia in patients with hypertension has prognostic implications. A timely diagnosis of FHH is important for effective management of hypertension, electrolyte imbalance correction with tailored treatment, and genetic counseling.
Collapse
Affiliation(s)
- Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| | - Jacopo Burrello
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| | - Marguerite Hureaux
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Génétique et Centre de Référence des Maladies Rénales Rares, Hôpital Européen Georges Pompidou, Paris, France (M.H., C.B., E.C., X.J.)
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) U970 Paris Cardiovascular Research Center, France (M.H., C.B., E.C., X.J.)
| | - Clarisse Billon
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Génétique et Centre de Référence des Maladies Rénales Rares, Hôpital Européen Georges Pompidou, Paris, France (M.H., C.B., E.C., X.J.)
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) U970 Paris Cardiovascular Research Center, France (M.H., C.B., E.C., X.J.)
| | - Eric Clauser
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Génétique et Centre de Référence des Maladies Rénales Rares, Hôpital Européen Georges Pompidou, Paris, France (M.H., C.B., E.C., X.J.)
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) U970 Paris Cardiovascular Research Center, France (M.H., C.B., E.C., X.J.)
| | - Franco Veglio
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| | - Franco Rabbia
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| | - Barbara Pasini
- Medical Genetics Unit, Department of Medical Science (B.P.), University of Torino, Italy
- SC Genetica Medica U, Azienda Ospedaliero-Universitaria (AOU) Città della Salute e della Scienza di Torino, Italy (B.P.)
| | - Annalisa Crisetti
- Nephrology and Dialisis Unit, Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy (A.C.)
| | - Xavier Jeunemaitre
- Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Génétique et Centre de Référence des Maladies Rénales Rares, Hôpital Européen Georges Pompidou, Paris, France (M.H., C.B., E.C., X.J.)
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) U970 Paris Cardiovascular Research Center, France (M.H., C.B., E.C., X.J.)
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences (M.T., J.B., F.V., F.R., P.M., S.M.), University of Torino, Italy
| |
Collapse
|
3
|
Zachariah T, Radhakrishnan J. Potential Role of Mineralocorticoid Receptor Antagonists in Nondiabetic Chronic Kidney Disease and Glomerular Disease. Clin J Am Soc Nephrol 2024; 19:1499-1512. [PMID: 39037799 PMCID: PMC11556932 DOI: 10.2215/cjn.0000000000000540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Glomerular disease is a leading cause of CKD and ESKD. Although diabetic kidney disease is the most common cause of glomerular disease, nondiabetic causes include malignancy, systemic autoimmune conditions, drug effects, or genetic conditions. Nondiabetic glomerular diseases are rare diseases, with a paucity of high-quality clinical trials in this area. Furthermore, late referral can result in poor patient outcomes. This article reviews the current management of nondiabetic glomerular disease and explores the latest developments in drug treatment in this area. Current treatment of nondiabetic glomerular disease aims to manage complications (edema, hypertension, proteinuria, hyperlipidemia, hypercoagulability, and thrombosis) as well as target the underlying cause of glomerular disease. Treatment options include renin-angiotensin-aldosterone system inhibitors, statins/nonstatin alternatives, loop diuretics, anticoagulation agents, immunosuppressives, and lifestyle and dietary modifications. Effective treatment of nondiabetic glomerular disease is limited by heterogeneity and a lack of understanding of the disease pathogenesis. Sodium-glucose cotransporter-2 inhibitors and nonsteroidal mineralocorticoid receptor antagonists (ns-MRAs, such as finerenone), with their broad anti-inflammatory and antifibrotic effects, have emerged as valuable therapeutic options for a range of cardiorenal conditions, including CKD. ns-MRAs are an evolving drug class of particular interest for the future treatment of nondiabetic glomerular disease, and there is evidence that these agents may improve kidney prognosis in various subgroups of patients with CKD. The benefits offered by ns-MRAs may present an opportunity to reduce the progression of CKD from a spectrum of glomerular disease. Several novel ns-MRA are in clinical development for both diabetic and nondiabetic CKD.
Collapse
|
4
|
Mortensen LA, Jespersen B, Helligsoe ASL, Tougaard B, Cibulskyte-Ninkovic D, Egfjord M, Boesby L, Marcussen N, Madsen K, Jensen BL, Petersen I, Bistrup C, Thiesson HC. Effect of Spironolactone on Kidney Function in Kidney Transplant Recipients (the SPIREN trial): A Randomized Placebo-Controlled Clinical Trial. Clin J Am Soc Nephrol 2024; 19:755-766. [PMID: 38416033 PMCID: PMC11168825 DOI: 10.2215/cjn.0000000000000439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
Key Points Spironolactone is safe for kidney transplant patients. Spironolactone reduces kidney function by an acute effect, whereafter it remains stable. Spironolactone does not affect the progression of interstitial fibrosis in protocol biopsies. Background Long-term kidney allograft survival is hampered by progressive interstitial fibrosis and tubular atrophy. The SPIREN trial tested the hypothesis that the mineralocorticoid receptor antagonist spironolactone stabilizes kidney function and attenuates glomerular barrier injury in kidney transplant patients treated with calcineurin inhibitors. Methods We conducted a randomized, placebo-controlled, double-blind clinical trial including 188 prevalent kidney transplant patients. Patients were randomized to spironolactone or placebo for 3 years. GFR was measured along with proteinuria and kidney fibrosis. The primary end point was change in measured GFR. Secondary outcomes were 24-hour proteinuria, kidney allograft fibrosis, and cardiovascular events. Measured GFRs, 24-hour proteinuria, and BP were determined yearly. Kidney biopsies were collected at baseline and after 2 years (n =48). Fibrosis was evaluated by quantitative stereology and classified according to Banff. Results The groups were comparable at baseline except for slightly older allografts in the spironolactone group. Spironolactone reduced measured GFRs (up to –7.6 [95% confidence interval, −10.9 to −4.3] ml/min compared with placebo) independently of time since transplantation and BP with no effect on the kidney function curve over time and reduced 24-hour proteinuria after 1 year. There was no significant effect of spironolactone on the development of interstitial fibrosis. Conclusions Spironolactone added to standard therapy for 3 years in kidney transplant patients did not improve kidney function, long-term proteinuria, or interstitial fibrosis. Clinical Trial registration number NCT01602861 .
Collapse
Affiliation(s)
- Line A. Mortensen
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Bente Jespersen
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | - Martin Egfjord
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Boesby
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Kirsten Madsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Boye L. Jensen
- Cardiovascular and Renal Research Unit, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Inge Petersen
- Odense University Hospital, OPEN, Open Patient data Explorative Network, Odense, Denmark
| | - Claus Bistrup
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Helle C. Thiesson
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Ikemoto M, Morimoto S, Ichihara A. Prediction of endogenous mineralocorticoid receptor activity by depressor effects of mineralocorticoid receptor antagonists in patients with primary aldosteronism. Hypertens Res 2024; 47:1707-1718. [PMID: 38548912 DOI: 10.1038/s41440-024-01651-5] [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: 12/04/2023] [Revised: 02/17/2024] [Accepted: 03/01/2024] [Indexed: 06/06/2024]
Abstract
Patients with primary aldosteronism have an increased risk of developing cardiovascular disease. The response to mineralocorticoid receptor antagonists varies among individuals, indicating diverse mineralocorticoid receptor activities in these patients. This study explored the factors linked to the efficacy of blood pressure reduction through mineralocorticoid receptor antagonists in patients with primary aldosteronism. We examined the relationship between the reduction in blood pressure and patient characteristics in a group of 41 patients with primary aldosteronism (24 males, mean age 55 ± 13 years, including 34 patients diagnosed with bilateral primary aldosteronism) before and after undergoing treatment with mineralocorticoid receptor antagonists. Significant reductions in office blood pressure were observed 3 and 6 months after treatment initiation. Single correlation analyses showed that the urinary chloride-to-potassium ratio displayed the strongest positive association with blood pressure reduction, surpassing plasma aldosterone concentration, plasma renin activity, and urinary sodium-to-potassium ratio, at 3 and 6 months. Multiple correlation analyses revealed a consistent and independent positive correlation between the urinary chloride-to-potassium ratio and blood pressure reduction at 3 and 6 months. The optimal threshold for the urinary chloride-to-potassium ratio with respect to its ability to lower blood pressure, was determined as 3.18. These results imply that the urinary chloride-to-potassium ratio may be independently associated with the effectiveness of blood pressure reduction facilitated by mineralocorticoid receptor antagonists. Moreover, it could potentially serve as a valuable predictor of the effectiveness of these agents and function as an indicator of endogenous mineralocorticoid receptor activity in patients with primary aldosteronism.
Collapse
Affiliation(s)
- Makiko Ikemoto
- Department of Internal Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoshi Morimoto
- Department of Internal Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| | - Atsuhiro Ichihara
- Department of Internal Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
6
|
Lim RS, Yeo SC, Barratt J, Rizk DV. An Update on Current Therapeutic Options in IgA Nephropathy. J Clin Med 2024; 13:947. [PMID: 38398259 PMCID: PMC10889409 DOI: 10.3390/jcm13040947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Immunoglobulin A nephropathy (IgAN) remains the leading cause of primary glomerular disease worldwide. Outcomes are poor with high rates of progressive chronic kidney disease and kidney failure, which contributes to global healthcare costs. Although this disease entity has been described, there were no disease-specific treatments until recently, with the current standard of care focusing on optimal supportive measures including lifestyle modifications and optimization of the renin-angiotensin-aldosterone blockade. However, with significant advances in the understanding of the pathogenesis of IgAN in the past decade, and the acceptance of surrogate outcomes for accelerated drug approval, there have been many new investigational agents tested to target this disease. As these agents become available, we envision a multi-pronged treatment strategy that simultaneously targets the consequences of ongoing nephron loss, stopping any glomerular inflammation, inhibiting pro-fibrotic signals in the glomerulus and tubulo-interstitium, and inhibiting the production of pathogenic IgA molecules. This review is an update on a previous review published in 2021, and we aim to summarize the developments and updates in therapeutic strategies in IgAN and highlight the promising discoveries that are likely to add to our armamentarium.
Collapse
Affiliation(s)
- Regina Shaoying Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore; (R.S.L.); (S.C.Y.)
| | - See Cheng Yeo
- Department of Renal Medicine, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore; (R.S.L.); (S.C.Y.)
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK;
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester LE5 4PW, UK
| | - Dana V. Rizk
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, ZRB 614, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Chen W, Zheng L, Wang J, Lin Y, Zhou T. Overview of the safety, efficiency, and potential mechanisms of finerenone for diabetic kidney diseases. Front Endocrinol (Lausanne) 2023; 14:1320603. [PMID: 38174337 PMCID: PMC10762446 DOI: 10.3389/fendo.2023.1320603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Diabetic kidney disease (DKD) is a common disorder with numerous severe clinical implications. Due to a high level of fibrosis and inflammation that contributes to renal and cardiovascular disease (CVD), existing treatments have not effectively mitigated residual risk for patients with DKD. Excess activation of mineralocorticoid receptors (MRs) plays a significant role in the progression of renal and CVD, mostly by stimulating fibrosis and inflammation. However, the application of traditional steroidal MR antagonists (MRAs) to DKD has been limited by adverse events. Finerenone (FIN), a third-generation non-steroidal selective MRA, has revealed anti-fibrotic and anti-inflammatory effects in pre-clinical studies. Current clinical trials, such as FIDELIO-DKD and FIGARO-DKD and their combined analysis FIDELITY, have elucidated that FIN reduces the kidney and CV composite outcomes and risk of hyperkalemia compared to traditional steroidal MRAs in patients with DKD. As a result, FIN should be regarded as one of the mainstays of treatment for patients with DKD. In this review, the safety, efficiency, and potential mechanisms of FIN treatment on the renal system in patients with DKD is reviewed.
Collapse
Affiliation(s)
| | | | | | | | - Tianbiao Zhou
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
8
|
Bobkova IN. [The role of mineralocorticoid receptors hyperactivation in the development of cardiorenal complications in patients with diabetes mellitus, perspective of the selective nonsteroidal mineralocorticoid receptors antagonist's treatment: A review]. TERAPEVT ARKH 2023; 95:796-801. [PMID: 38158924 DOI: 10.26442/00403660.2023.09.202367] [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: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 01/03/2024]
Abstract
The renin-angiotensin-aldosterone system (RAAS) activation plays a key role in the chronic kidney disease (CKD) progression and in the cardiovascular complications (CVC) development in patients with diabetes mellitus (DM). RAAS blockers alone are not sufficient to prevent CVC and CVC progression. RAAS upregulation in CKD associated with DM triggers the mineralocorticoid receptors (MCR) hyperactivation which results in fibrosis and inflammation in the heart and kidneys. This review presents the current data about the variety of MCR hyperactivation manifestations, as well as about of multiplicity of MCR hyperactivation ways in DM. The efficacy and safety of finerenone, a new MCR nonsteroidal selective antagonist, are discussed.
Collapse
Affiliation(s)
- I N Bobkova
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
9
|
Mima A, Lee R, Murakami A, Gotoda H, Akai R, Kidooka S, Nakamoto T, Kido S, Lee S. Effect of finerenone on diabetic kidney disease outcomes with estimated glomerular filtration rate below 25 mL/min/1.73 m 2. Metabol Open 2023; 19:100251. [PMID: 37497038 PMCID: PMC10366575 DOI: 10.1016/j.metop.2023.100251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
Background In the Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease trial, finerenone reduced the risk of cardiovascular events in patients with chronic kidney disease (CKD) and type 2 diabetes, while in the Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease trial, it improved renal and cardiovascular outcomes in patients with advanced CKD. However, no previous studies have assessed patients with CKD and type 2 diabetes with an estimated glomerular filtration rate (eGFR) below 25 mL/min/1.73 m2. Methods Nine patients with CKD and type 2 diabetes who received finerenone 10 mg/day were analyzed retrospectively. Changes in eGFR, urinary protein, and serum potassium levels were studied from 1 year before administration of finerenone until 6 months after administration. Results The mean baseline eGFR slope was -7.63 ± 9.84 (mL/min/1.73 m2/year). After finerenone treatment, the mean eGFR slope significantly improved -1.44 ± 3.17 (mL/min/1.73 m2/6 months, P=0.038). However, finerenone treatment did not significantly reduce proteinuria. Furthermore, finerenone did not increase serum potassium levels. Conclusions Patients treated with finerenone showed a significantly slower decline in eGFR. Furthermore, aside from the present study, no reports have indicated the effectiveness of finerenone in patients with advanced CKD with an eGFR below 25 mL/min/1.73 m2. As confirmed in our clinical trials, the finding that finerenone is effective in a wide range of renal functions can be generalized to clinical practice. However, sample size in this study was small. Thus, further large-scale investigations will be needed.
Collapse
Affiliation(s)
- Akira Mima
- Corresponding author. Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, 569-8686, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Tuot DS, Powe NR. Care of the Patient With Abnormal Kidney Test Results. Ann Intern Med 2023; 176:ITC65-ITC80. [PMID: 37155988 DOI: 10.7326/aitc202305160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Blood and urine tests are commonly performed by clinicians in both ambulatory and hospital settings that detect chronic and acute kidney disease. Thresholds for these tests have been established that signal the presence and severity of kidney injury or dysfunction. In the appropriate clinical context of a patient's history and physical examination, an abnormal test result should trigger specific actions for clinicians, including reviewing patient medication use, follow-up testing, prescribing lifestyle modifications, and specialist referral. Tests for kidney disease can also be used to determine the future risk for kidney failure as well as cardiovascular death.
Collapse
Affiliation(s)
- Delphine S Tuot
- Division of Nephrology and Department of Medicine, Priscilla Chan and Mark Zuckerberg San Francisco General Hospital, and Department of Medicine, University of California San Francisco, San Francisco, California (D.S.T.)
| | - Neil R Powe
- Department of Medicine, Priscilla Chan and Mark Zuckerberg San Francisco General Hospital, and Department of Medicine, University of California San Francisco, San Francisco, California (N.R.P.)
| |
Collapse
|
11
|
Figueroa SM, Bertocchio JP, Nakamura T, El-Moghrabi S, Jaisser F, Amador CA. The Mineralocorticoid Receptor on Smooth Muscle Cells Promotes Tacrolimus-Induced Renal Injury in Mice. Pharmaceutics 2023; 15:pharmaceutics15051373. [PMID: 37242615 DOI: 10.3390/pharmaceutics15051373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Tacrolimus (Tac) is a calcineurin inhibitor commonly used as an immunosuppressor after solid organ transplantation. However, Tac may induce hypertension, nephrotoxicity, and an increase in aldosterone levels. The activation of the mineralocorticoid receptor (MR) is related to the proinflammatory status at the renal level. It modulates the vasoactive response as they are expressed on vascular smooth muscle cells (SMC). In this study, we investigated whether MR is involved in the renal damage generated by Tac and if the MR expressed in SMC is involved. Littermate control mice and mice with targeted deletion of the MR in SMC (SMC-MR-KO) were administered Tac (10 mg/Kg/d) for 10 days. Tac increased the blood pressure, plasma creatinine, expression of the renal induction of the interleukin (IL)-6 mRNA, and expression of neutrophil gelatinase-associated lipocalin (NGAL) protein, a marker of tubular damage (p < 0.05). Our study revealed that co-administration of spironolactone, an MR antagonist, or the absence of MR in SMC-MR-KO mice mitigated most of the unwanted effects of Tac. These results enhance our understanding of the involvement of MR in SMC during the adverse reactions of Tac treatment. Our findings provided an opportunity to design future studies considering the MR antagonism in transplanted subjects.
Collapse
Affiliation(s)
- Stefanny M Figueroa
- Institute of Biomedical Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Jean-Philippe Bertocchio
- INSERM UMRS1138, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Toshifumi Nakamura
- INSERM UMRS1138, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Soumaya El-Moghrabi
- INSERM UMRS1138, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Frédéric Jaisser
- INSERM UMRS1138, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Cristián A Amador
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510156, Chile
| |
Collapse
|
12
|
Salukhov VV, Shamkhalova MS, Duganova AV. [Finerenone cardiorenal effects and its placement in treatment of chronic kidney disease in patients with type 2 diabetes mellitus: A review]. TERAPEVT ARKH 2023; 95:261-273. [PMID: 37167149 DOI: 10.26442/00403660.2023.03.202152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
Chronic kidney disease (CKD) is one of the most common complications of diabetes mellitus and an independent risk factor for cardiovascular disease. Despite guideline-directed therapy of CKD in patients with type 2 diabetes, the risk of renal failure and cardiovascular events still remains high. To date, current medications for CKD haven't reduced enough the residual risk associated with inflammation and fibrosis in patients with type 2 diabetes. Here, in this review we present the results of FIDELIO-DKD, FIGARO-DKD trials and their pooled analysis FIDELITY, aimed to evaluate the effectiveness and safety of selective non-steroidal mineralocorticoid receptor antagonist finerenone in patients with type 2 diabetes with wide range stages of CKD. Modern pathophysiological aspects of mineralocorticoid receptor hyperactivation and features of their blockade by steroidal and nonsteroidal mineralocorticoid receptor antagonists are considered, differences in pharmacological effects between them are also discussed, finerenone benefits and its adverse events, demonstrated in randomized clinical trials are considered here. The probable mechanisms of early and delayed action of finerenone, which were realized in beneficial cardiovascular and renal outcomes in patients with type 2 diabetes with CKD, are presented here. Practical points for finerenone initiation and titration are indicated, aimed to minimize the hyperkalemia risk. Current guidelines for CKD treatment in patients with type 2 diabetes are analyzed, the finerenone placement in combined nephroprotective therapy is determined.
Collapse
|
13
|
Cherney DZI, Bell A, Girard L, McFarlane P, Moist L, Nessim SJ, Soroka S, Stafford S, Steele A, Tangri N, Weinstein J. Management of Type 2 Diabetic Kidney Disease in 2022: A Narrative Review for Specialists and Primary Care. Can J Kidney Health Dis 2023; 10:20543581221150556. [PMID: 36726361 PMCID: PMC9884958 DOI: 10.1177/20543581221150556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/03/2022] [Indexed: 01/26/2023] Open
Abstract
Purpose of review Kidney disease is present in almost half of Canadian patients with type 2 diabetes (T2D), and it is also the most common first cardiorenal manifestation of T2D. Despite clear guidelines for testing, opportunities are being missed to identify kidney diseases, and many Canadians are therefore not receiving the best available treatments. This has become even more important given recent clinical trials demonstrating improvements in both kidney and cardiovascular (CV) endpoints with sodium-glucose cotransporter 2 (SGLT2) inhibitors and a nonsteroidal mineralocorticoid receptor antagonist, finerenone. The goal of this document is to provide a narrative review of the current evidence for the treatment of diabetic kidney disease (DKD) that supports this new standard of care and to provide practice points. Sources of information An expert panel of Canadian clinicians was assembled, including 9 nephrologists, an endocrinologist, and a primary care practitioner. The information the authors used for this review consisted of published clinical trials and guidelines, selected by the authors based on their assessment of their relevance to the questions being answered. Methods Panelists met virtually to discuss potential questions to be answered in the review and agreed on 10 key questions. Two panel members volunteered as co-leads to write the summaries and practice points for each of the identified questions. Summaries and practice points were distributed to the entire author list by email. Through 2 rounds of online voting, a second virtual meeting, and subsequent email correspondence, the authors reached consensus on the contents of the review, including all the practice points. Key findings It is critical that DKD be identified as early as possible in the course of the disease to optimally prevent disease progression and associated complications. Patients with diabetes should be routinely screened for DKD with assessments of both urinary albumin and kidney function. Treatment decisions should be individualized based on the risks and benefits, patients' needs and preferences, medication access and cost, and the degree of glucose lowering needed. Patients with DKD should be treated to achieve targets for A1C and blood pressure. Renin-angiotensin-aldosterone system blockade and treatment with SGLT2 inhibitors are also key components of the standard of care to reduce the risk of kidney and CV events for these patients. Finerenone should also be considered to further reduce the risk of CV events and chronic kidney disease progression. Education of patients with diabetes prescribed SGLT2 inhibitors and/or finerenone is an important component of treatment. Limitations No formal guideline process was used. The practice points are not graded and are not intended to be viewed as having the weight of a clinical practice guideline or formal consensus statement. However, most practice points are well aligned with current clinical practice guidelines.
Collapse
Affiliation(s)
- David Z. I. Cherney
- Division of Nephrology, Department of
Medicine, Toronto General Hospital, University of Toronto, ON, Canada
- Temerty Faculty of Medicine, University
of Toronto, ON, Canada
| | - Alan Bell
- Department of Family & Community
Medicine, University of Toronto, ON, Canada
| | - Louis Girard
- Division of Nephrology, Department of
Medicine, Cumming School of Medicine, University of Calgary, AB, Canada
| | - Philip McFarlane
- Division of Nephrology, Department of
Medicine, Toronto General Hospital, University of Toronto, ON, Canada
| | - Louise Moist
- Division of Nephrology, Department of
Medicine, Schulich School of Medicine & Dentistry, Western University, London,
ON, Canada
| | - Sharon J. Nessim
- Division of Nephrology, Jewish General
Hospital, McGill University, Montreal, QC, Canada
| | - Steven Soroka
- QEII Health Sciences Centre, Nova
Scotia Health, Halifax, Canada
| | - Sara Stafford
- Fraser Health Division of
Endocrinology, University of British Columbia, Surrey, Canada
| | | | - Navdeep Tangri
- Departments of Medicine and Community
Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Jordan Weinstein
- Division of Nephrology, St. Michael’s
Hospital, University of Toronto, ON, Canada
| |
Collapse
|
14
|
Jain M, Mohan S, van Dijk EHC. Central serous chorioretinopathy: Pathophysiology, systemic associations, and a novel etiological classification. Taiwan J Ophthalmol 2022; 12:381-393. [PMID: 36660127 PMCID: PMC9843580 DOI: 10.4103/2211-5056.362601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/04/2022] [Indexed: 12/08/2022] Open
Abstract
Central serous chorioretinopathy (CSC) has remained an enigmatic disease since its initial description by Von Graefe. Over the years, multiple risk factors have been recognized: these include psychological stress, behavioral traits, and corticosteroids. The basic pathophysiology of CSC involves choroidal thickening, vascular congestion, altered choroidal blood flow (ChBF), and choroidal hyperpermeability, leading to retinal pigment epithelium decompensation and subsequent neurosensory detachment. Multiple organ systems, mainly the nervous, cardiovascular, endocrinal, and renal systems participate in the control of the vascular tone and the ChBF via hypothalamus-pituitary-adrenal axis and renin-angiotensin-aldosterone system, while others such as the hepatic system regulate the enzymatic degradation of corticosteroids. Many vasoactive and psychotropic drugs also modulate the ocular perfusion. In addition, there are anatomical and genetic predispositions that determine its progression to the chronic or recurrent form, through cellular response and angiogenesis. We herein review the basic pathophysiology and immunogenetics in CSC along with the role of multiple organ systems. With this background, we propose an etiological classification that should provide a framework for customized therapeutic interventions.
Collapse
Affiliation(s)
- Manish Jain
- Department of Ophthalmology, Al Dhannah Hospital, Abu Dhabi, United Arab Emirates,Address for correspondence: Dr. Manish Jain, Department of Ophthalmology, Al Dhannah Hospital, Abu Dhabi, United Arab Emirates. E-mail:
| | - Sashwanthi Mohan
- Department of Vitreous and Retina, Rajan Eye Care Hospital, Chennai, Tamil Nadu, India
| | - Elon H. C. van Dijk
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
15
|
Trubitsyna NP, Zaitseva NV, Severinа AS, Shamkhalova MS. Chronic kidney disease in patients with type 2 diabetes: new targets of medicine action. DIABETES MELLITUS 2022. [DOI: 10.14341/dm12944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Diabetes mellitus type 2 (DM2) is socially important disease, becoming non-infectious epidemic due to increasing prevalence. Chronic kidney disease (CKD) is one of the most common diabetic complications. Kidney injury signs and/or estimated glomerular filtration rate (eGFR) decrease are seen in 40-50% of patients with DM2. Three groups of factors are considered to be the basis of CKD development and progression in DM2: metabolic, hemodynamic, inflammation and fibrosis. Existing drugs that are used in patients with CKD and DM2 first of all target hemodynamic and metabolic disturbances, but their action against inflammation and fibrosis is indirect. Hyperactivation of mineralocorticoid receptors (MR) is considered as one of the main trigger factors of end-organ damage in patients with DM2 due to inflammation and fibrosis. Development of selective nonsteroidal MR antagonists (MRA) as a new class of medications is directed to demonstrate positive effects from blocking this pathophysiological pathway of CKD development and overcome the steroidal MRAs’ shortcomings. Hence pathophysiological hyperactivation of MR with subsequent inflammation and fibrosis in patients with CKD in DM2 is considered a promising therapeutic target for the new drugs with cardionephroprotective effect.
Collapse
|
16
|
Ameer OZ. Hypertension in chronic kidney disease: What lies behind the scene. Front Pharmacol 2022; 13:949260. [PMID: 36304157 PMCID: PMC9592701 DOI: 10.3389/fphar.2022.949260] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022] Open
Abstract
Hypertension is a frequent condition encountered during kidney disease development and a leading cause in its progression. Hallmark factors contributing to hypertension constitute a complexity of events that progress chronic kidney disease (CKD) into end-stage renal disease (ESRD). Multiple crosstalk mechanisms are involved in sustaining the inevitable high blood pressure (BP) state in CKD, and these play an important role in the pathogenesis of increased cardiovascular (CV) events associated with CKD. The present review discusses relevant contributory mechanisms underpinning the promotion of hypertension and their consequent eventuation to renal damage and CV disease. In particular, salt and volume expansion, sympathetic nervous system (SNS) hyperactivity, upregulated renin–angiotensin–aldosterone system (RAAS), oxidative stress, vascular remodeling, endothelial dysfunction, and a range of mediators and signaling molecules which are thought to play a role in this concert of events are emphasized. As the control of high BP via therapeutic interventions can represent the key strategy to not only reduce BP but also the CV burden in kidney disease, evidence for major strategic pathways that can alleviate the progression of hypertensive kidney disease are highlighted. This review provides a particular focus on the impact of RAAS antagonists, renal nerve denervation, baroreflex stimulation, and other modalities affecting BP in the context of CKD, to provide interesting perspectives on the management of hypertensive nephropathy and associated CV comorbidities.
Collapse
Affiliation(s)
- Omar Z. Ameer
- Department of Pharmaceutical Sciences, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
- Department of Biomedical Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
- *Correspondence: Omar Z. Ameer,
| |
Collapse
|
17
|
Epidemiology and risk of cardiovascular disease in populations with chronic kidney disease. Nat Rev Nephrol 2022; 18:696-707. [DOI: 10.1038/s41581-022-00616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/08/2022]
|
18
|
Parfianowicz D, Shah S, Nguyen C, Maitz TN, Hajra A, Goel A, Sreenivasan J, Aronow WS, Vyas A, Gupta R. Finerenone: A New Era for Mineralocorticoid Receptor Antagonism and Cardiorenal Protection. Curr Probl Cardiol 2022; 47:101386. [PMID: 36057315 DOI: 10.1016/j.cpcardiol.2022.101386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/03/2022]
|
19
|
Agarwal R, Anker SD, Bakris G, Filippatos G, Pitt B, Rossing P, Ruilope L, Gebel M, Kolkhof P, Nowack C, Joseph A. Investigating new treatment opportunities for patients with chronic kidney disease in type 2 diabetes: the role of finerenone. Nephrol Dial Transplant 2022; 37:1014-1023. [PMID: 33280027 PMCID: PMC9130026 DOI: 10.1093/ndt/gfaa294] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Despite the standard of care, patients with chronic kidney disease (CKD) and type 2 diabetes (T2D) progress to dialysis, are hospitalized for heart failure and die prematurely. Overactivation of the mineralocorticoid receptor (MR) causes inflammation and fibrosis that damages the kidney and heart. Finerenone, a nonsteroidal, selective MR antagonist, confers kidney and heart protection in both animal models and Phase II clinical studies; the effects on serum potassium and kidney function are minimal. Comprising the largest CKD outcomes program to date, FIDELIO-DKD (FInerenone in reducing kiDnEy faiLure and dIsease prOgression in Diabetic Kidney Disease) and FIGARO-DKD (FInerenone in reducinG cArdiovascular moRtality and mOrbidity in Diabetic Kidney Disease) are Phase III trials investigating the efficacy and safety of finerenone on kidney failure and cardiovascular outcomes from early to advanced CKD in T2D. By including echocardiograms and biomarkers, they extend our understanding of pathophysiology; by including quality of life measurements, they provide patient-centered outcomes; and by including understudied yet high-risk cardiorenal subpopulations, they have the potential to widen the scope of therapy in T2D with CKD. Trial registration number: FIDELIO-DKD (NCT02540993) and FIGARO-DKD (NCT02545049).
Collapse
Affiliation(s)
- Rajiv Agarwal
- Richard L. Roudebush VA Medical Center and Indiana University, Indianapolis, IN, USA
| | - Stefan D Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - George Bakris
- Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens, School of Medicine, Department of Cardiology, Attikon University Hospital, Athens, Greece
| | - Bertram Pitt
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Luis Ruilope
- Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research imas12, Madrid, Spain
- IBER-CV, Hospital Universitario, 12 de Octubre, Madrid, Spain
- Faculty of Sport Sciences, European University of Madrid, Madrid, Spain
| | - Martin Gebel
- Research and Development, Statistics and Data Insights, Bayer AG, Berlin, Germany
| | - Peter Kolkhof
- Research and Development, Preclinical Research Cardiovascular, Bayer AG, Wuppertal, Germany
| | - Christina Nowack
- Research and Development, Clinical Development Operations, Bayer AG, Wuppertal, Germany
| | - Amer Joseph
- Cardiology and Nephrology Clinical Development, Bayer AG, Berlin, Germany
| | | |
Collapse
|
20
|
Leon SJ, Tangri N. Balancing Hyperkalemia Risks with Clinical Benefits of Renin-Angiotensin-Aldosterone Inhibitors/Mineralocorticoid Receptor Antagonists Blockade: It's Apples and Oranges. KIDNEY360 2022; 3:1442-1444. [PMID: 36176652 PMCID: PMC9416839 DOI: 10.34067/kid.0000952022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/15/2022] [Indexed: 01/11/2023]
Affiliation(s)
- Silvia J. Leon
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Canada
| | - Navdeep Tangri
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
21
|
Liu J, Jia W, Yu C. Safety and Efficacy of Spironolactone in Dialysis-Dependent Patients: Meta-Analysis of Randomized Controlled Trials. Front Med (Lausanne) 2022; 9:828189. [PMID: 35372414 PMCID: PMC8970057 DOI: 10.3389/fmed.2022.828189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background Patients with end-stage renal disease (ESRD) are characterized with high risk of heart failure. Although mineralocorticoid receptor antagonists have beneficial effect on relieving cardiac fibrosis and, thus, reduce the incidence of cardiovascular disease and cardiac death, the therapeutic benefits and adverse effects are still controversial. We conducted a meta-analysis to measure the safety and efficacy of spironolactone in patients undergoing dialysis. Methods A systematic search for randomized controlled trials (RCTs) was performed in PubMed, Embase, and Cochrane databases. Primary outcomes included changes in all-cause mortality (ACM), serum potassium concentration, incidence of hyperkalemia and gynecomastia (GYN). Secondary outcomes included changes in blood pressure (BP), left ventricular mass index (LVMI) and left ventricular ejection fraction (LVEF). Subgroup analysis and sensitivity analysis were further conducted. This research was registered with PROSPERO (International Prospective Register of Systematic Reviews; No. CRD42021287493). Results Fifteen RCTs with 1,258 patients were enrolled in this pooled-analysis. Spironolactone treatment significantly decreased ACM (RR = 0.42, P < 0.0001), CCV (RR = 0.54, P = 0.008) and LVMI (MD = −6.28, P = 0.002), also increased occurrence of GYN (RR = 4.36, P = 0.0005). However, LVEF (MD = 2.63, P = 0.05), systolic BP (MD = −4.61, P = 0.14) and diastolic BP (MD = −0.12, P = 0.94) did not change between two groups after treatment. Although serum potassium concentration was increased (MD = 0.22, P < 0.0001) after spironolactone supplement, the risk of hyperkalemia remained unchanged (RR = 1.21, P = 0.31). Further subgroup analysis found more obvious advantageous as well as disadvantageous effects in Asian subjects than European or American ones. Also, with more than 9 months of treatment duration, patients achieved more favorable influence than shorter duration. Conclusions These results highlight the therapeutic effects of spironolactone on cardiovascular indexes, including ACM, CCV, and LVMI. However, the unignorable increase of GYN incidence and serum potassium level indicate that close monitor in dialysis-dependent patients, especially Asian patients, is essential.
Collapse
Affiliation(s)
- Jing Liu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - WanYu Jia
- Department of Pediatrics, Clinical Center of Pediatric Nephrology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Chen Yu
| |
Collapse
|
22
|
Li Y, Chakraborty A, Broughton BRS, Ferens D, Widdop RE, Ricardo SD, Samuel CS. Comparing the renoprotective effects of BM-MSCs versus BM-MSC-exosomes, when combined with an anti-fibrotic drug, in hypertensive mice. Biomed Pharmacother 2021; 144:112256. [PMID: 34607108 DOI: 10.1016/j.biopha.2021.112256] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 12/29/2022] Open
Abstract
Fibrosis, a hallmark of chronic kidney disease (CKD), impairs the viability of human bone marrow derived-mesenchymal stromal cells (BM-MSCs) post-transplantation. To address this, we demonstrated that combining BM-MSCs with the anti-fibrotic drug, serelaxin (RLX), enhanced BM-MSC-induced renoprotection in preclinical CKD models. Given the increased interest and manufacturing advantages to using stem cell-derived exosomes (EXO) as therapeutics, this study determined whether RLX could enhance the therapeutic efficacy of BM-MSC-EXO, and compared the renoprotective effects of RLX and BM-MSC-EXO versus RLX and BM-MSCs in mice with hypertensive CKD. Adult male C57BL/6 mice were uninephrectomised, received deoxycorticosterone acetate and given saline to drink (1K/DOCA/salt) for 21 days. Control mice were uninephrectomised and given normal drinking water for the same time-period. Subgroups of 1K/DOCA/salt-hypertensive mice were then treated with either RLX (0.5 mg/kg/day) or BM-MSC-EXO (25 μg/mouse; equivalent to 1-2 × 106 BM-MSCs/mouse) alone; combinations of RLX and BM-MSC-EXO or BM-MSCs (1 × 106/mouse); or the mineralocorticoid receptor antagonist, spironolactone (20 mg/kg/day), from days 14-21. 1K/DOCA/salt-hypertensive mice developed kidney tubular damage, inflammation and fibrosis, and impaired kidney function 21 days post-injury. Whilst RLX alone attenuated the 1K/DOCA/salt-induced fibrosis, BM-MSC-EXO alone only diminished measures of tissue inflammation post-treatment. Comparatively, the combined effects of RLX and BM-MSC-EXO or BM-MSCs demonstrated similar anti-fibrotic efficacy, but RLX and BM-MSCs offered broader renoprotection over RLX and/or BM-MSC-EXO, and comparable effects to spironolactone. Only RLX and BM-MSCs, but not RLX and/or BM-MSC-EXO, also attenuated the 1K/DOCA/salt-induced hypertension. Hence, although RLX improved the renoprotective effects of BM-MSC-EXO, combining RLX with BM-MSCs provided a better therapeutic option for hypertensive CKD.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Amlan Chakraborty
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Dorota Ferens
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Sharon D Ricardo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
23
|
Kintscher U, Bakris GL, Kolkhof P. Novel Non-Steroidal Mineralocorticoid Receptor Antagonists in Cardiorenal Disease. Br J Pharmacol 2021; 179:3220-3234. [PMID: 34811750 DOI: 10.1111/bph.15747] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Mineralocorticoid receptor (MR) antagonists (MRAs) are key agents in guideline-oriented drug therapy for cardiovascular (CV) diseases such as chronic heart failure with reduced ejection fraction (HFrEF) and resistant hypertension. Currently available steroidal MRAs are efficacious in reducing morbidity and mortality, however, they can be associated with intolerable side effects including hyperkalemia in everyday clinical practice. Recently, a new class of non-steroidal MRAs including esaxerenone, AZD9977, apararenone, KBP-5074, and finerenone have been developed with an improved benefit-risk profile and a novel indication for finerenone for diabetic kidney disease. To better understand the non-steroidal MRAs, this review provides information on the molecular pharmacology as well as relevant current preclinical and clinical data on cardiorenal outcomes. A comparative review of all compounds in the class is discussed with regard to clinical efficacy and safety as well as a perspective outlining their future use in clinical practice.
Collapse
Affiliation(s)
- Ulrich Kintscher
- Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal Research Center, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - George L Bakris
- Department of Medicine, American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL, USA
| | - Peter Kolkhof
- Research & Early Development, Cardiovascular Research, Bayer AG, Wuppertal, Germany
| |
Collapse
|
24
|
Kawanami D, Takashi Y, Muta Y, Oda N, Nagata D, Takahashi H, Tanabe M. Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease. Front Pharmacol 2021; 12:754239. [PMID: 34790127 PMCID: PMC8591525 DOI: 10.3389/fphar.2021.754239] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/13/2021] [Indexed: 01/19/2023] Open
Abstract
Diabetic kidney disease (DKD) is a major cause of end-stage kidney disease (ESKD) worldwide. Mineralocorticoid receptor (MR) plays an important role in the development of DKD. A series of preclinical studies revealed that MR is overactivated under diabetic conditions, resulting in promoting inflammatory and fibrotic process in the kidney. Clinical studies demonstrated the usefulness of MR antagonists (MRAs), such as spironolactone and eplerenone, on DKD. However, concerns regarding their selectivity for MR and hyperkalemia have remained for these steroidal MRAs. Recently, nonsteroidal MRAs, including finerenone, have been developed. These agents are highly selective and have potent anti-inflammatory and anti-fibrotic properties with a low risk of hyperkalemia. We herein review the current knowledge and future perspectives of MRAs in DKD treatment.
Collapse
Affiliation(s)
- Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yuichi Takashi
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yoshimi Muta
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Naoki Oda
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Dai Nagata
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Hiroyuki Takahashi
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Makito Tanabe
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
25
|
Barrera-Chimal J, Jaisser F, Anders HJ. The mineralocorticoid receptor in chronic kidney disease. Br J Pharmacol 2021; 179:3152-3164. [PMID: 34786690 DOI: 10.1111/bph.15734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/11/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022] Open
Abstract
Chronic kidney disease (CKD) is a major public health concern, affecting approximately 10% of the population worldwide. CKD of glomerular or tubular origin leads to the activation of stress mechanisms, including the renin angiotensin aldosterone system and mineralocorticoid receptor (MR) activation. Over the last two decades, blockade of the MR has arisen as a potential therapeutic approach against various forms of kidney disease. In this review, we summarize the experimental studies that have shown a protective effect of MR antagonists (MRAs) in non-diabetic and diabetic CKD animal models. Moreover, we review the main clinical trials that have shown the clinical application of MRAs to reduce albuminuria and, importantly, to slow CKD progression. Recent evidence from the FIDELIO trial showed that the MRA finerenone can reduce hard kidney outcomes when added to the standard of care in CKD associated with type 2 diabetes. Finally, we discuss the effects of MRAs relative to those of SGLT2 inhibitors, as well as the potential benefit of combination therapy to maximize organ protection.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Frederic Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Université de Lorraine, INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ziemssenstr. 1, D-80336, München
| |
Collapse
|
26
|
Edwards NC, Price AM, Mehta S, Hiemstra TF, Kaur A, Greasley PJ, Webb DJ, Dhaun N, MacIntyre IM, Farrah T, Melville V, Herrey AS, Slinn G, Wale R, Ives N, Wheeler DC, Wilkinson I, Steeds RP, Ferro CJ, Townend JN. Effects of Spironolactone and Chlorthalidone on Cardiovascular Structure and Function in Chronic Kidney Disease: A Randomized, Open-Label Trial. Clin J Am Soc Nephrol 2021; 16:1491-1501. [PMID: 34462286 PMCID: PMC8499017 DOI: 10.2215/cjn.01930221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 08/16/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES In a randomized double-blind, placebo-controlled trial, treatment with spironolactone in early-stage CKD reduced left ventricular mass and arterial stiffness compared with placebo. It is not known if these effects were due to BP reduction or specific vascular and myocardial effects of spironolactone. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A prospective, randomized, open-label, blinded end point study conducted in four UK centers (Birmingham, Cambridge, Edinburgh, and London) comparing spironolactone 25 mg to chlorthalidone 25 mg once daily for 40 weeks in 154 participants with nondiabetic stage 2 and 3 CKD (eGFR 30-89 ml/min per 1.73 m2). The primary end point was change in left ventricular mass on cardiac magnetic resonance imaging. Participants were on treatment with an angiotensin-converting enzyme inhibitor or angiotensin receptor blocker and had controlled BP (target ≤130/80 mm Hg). RESULTS There was no significant difference in left ventricular mass regression; at week 40, the adjusted mean difference for spironolactone compared with chlorthalidone was -3.8 g (95% confidence interval, -8.1 to 0.5 g, P=0.08). Office and 24-hour ambulatory BPs fell in response to both drugs with no significant differences between treatment. Pulse wave velocity was not significantly different between groups; at week 40, the adjusted mean difference for spironolactone compared with chlorthalidone was 0.04 m/s (-0.4 m/s, 0.5 m/s, P=0.90). Hyperkalemia (defined ≥5.4 mEq/L) occurred more frequently with spironolactone (12 versus two participants, adjusted relative risk was 5.5, 95% confidence interval, 1.4 to 22.1, P=0.02), but there were no patients with severe hyperkalemia (defined ≥6.5 mEq/L). A decline in eGFR >30% occurred in eight participants treated with chlorthalidone compared with two participants with spironolactone (adjusted relative risk was 0.2, 95% confidence interval, 0.05 to 1.1, P=0.07). CONCLUSIONS Spironolactone was not superior to chlorthalidone in reducing left ventricular mass, BP, or arterial stiffness in nondiabetic CKD.
Collapse
Affiliation(s)
- Nicola C. Edwards
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom,Department of Cardiology, Green Lane Cardiovascular Unit, Auckland, New Zealand
| | - Anna M. Price
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom,Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Samir Mehta
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Thomas F. Hiemstra
- Cambridge Clinical Trials Unit, Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, United Kingdom,GlaxoSmithKline, England, United Kingdom
| | - Amreen Kaur
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom
| | - Peter J. Greasley
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - David J. Webb
- Center for Cardiovascular Science and Clinical Research Center, University of Edinburgh, United Kingdom
| | - Neeraj Dhaun
- Center for Cardiovascular Science and Clinical Research Center, University of Edinburgh, United Kingdom,Department of Nephrology, National Health Services Lothian, Edinburgh, United Kingdom
| | - Iain M. MacIntyre
- Center for Cardiovascular Science and Clinical Research Center, University of Edinburgh, United Kingdom,Department of Nephrology, National Health Services Lothian, Edinburgh, United Kingdom
| | - Tariq Farrah
- Center for Cardiovascular Science and Clinical Research Center, University of Edinburgh, United Kingdom,Department of Nephrology, National Health Services Lothian, Edinburgh, United Kingdom
| | - Vanessa Melville
- Center for Cardiovascular Science and Clinical Research Center, University of Edinburgh, United Kingdom
| | - Anna S. Herrey
- UCL Institute of Cardiovascular Science and Department of Cardiology, Barts Heart Centre, St Bartholomew’s Hospital, London, United Kingdom
| | - Gemma Slinn
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Rebekah Wale
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - Natalie Ives
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | - David C. Wheeler
- Department of Renal Medicine, University College London, United Kingdom,George Institute for Global Health, Sydney, Australia
| | - Ian Wilkinson
- Cambridge Clinical Trials Unit, Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, United Kingdom,GlaxoSmithKline, England, United Kingdom
| | - Richard P. Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom,Department of Cardiology, Queen Elizabeth Hospital Birmingham, United Kingdom
| | - Charles J. Ferro
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom,Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Jonathan N. Townend
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom,Department of Cardiology, Queen Elizabeth Hospital Birmingham, United Kingdom
| |
Collapse
|
27
|
Yan MT, Chao CT, Lin SH. Chronic Kidney Disease: Strategies to Retard Progression. Int J Mol Sci 2021; 22:ijms221810084. [PMID: 34576247 PMCID: PMC8470895 DOI: 10.3390/ijms221810084] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD), defined as the presence of irreversible structural or functional kidney damages, increases the risk of poor outcomes due to its association with multiple complications, including altered mineral metabolism, anemia, metabolic acidosis, and increased cardiovascular events. The mainstay of treatments for CKD lies in the prevention of the development and progression of CKD as well as its complications. Due to the heterogeneous origins and the uncertainty in the pathogenesis of CKD, efficacious therapies for CKD remain challenging. In this review, we focus on the following four themes: first, a summary of the known factors that contribute to CKD development and progression, with an emphasis on avoiding acute kidney injury (AKI); second, an etiology-based treatment strategy for retarding CKD, including the approaches for the common and under-recognized ones; and third, the recommended approaches for ameliorating CKD complications, and the final section discusses the novel agents for counteracting CKD progression.
Collapse
Affiliation(s)
- Ming-Tso Yan
- Department of Medicine, Division of Nephrology, Cathay General Hospital, School of Medicine, Fu-Jen Catholic University, Taipei 106, Taiwan;
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei 114, Taiwan
| | - Chia-Ter Chao
- Department of Internal Medicine, Nephrology Division, National Taiwan University Hospital, Taipei 104, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 104, Taiwan
- Department of Internal Medicine, Nephrology Division, National Taiwan University College of Medicine, Taipei 104, Taiwan
| | - Shih-Hua Lin
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei 114, Taiwan
- Department of Internal Medicine, Nephrology Division, National Defense Medical Center, Taipei 104, Taiwan
- Correspondence: or
| |
Collapse
|
28
|
Georgianos PI, Agarwal R. Mineralocorticoid Receptor Antagonism in Chronic Kidney Disease. Kidney Int Rep 2021; 6:2281-2291. [PMID: 34514191 PMCID: PMC8418944 DOI: 10.1016/j.ekir.2021.05.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/25/2022] Open
Abstract
The overactivation of the mineralocorticoid receptor (MR) in animal models of chronic kidney disease (CKD) increases sodium retention and hypertension and provokes inflammation and fibrosis in the kidneys, blood vessels, and the heart; these processes play an important role in the progression of cardiorenal disease. Accordingly, blockade of the MR is an attractive therapeutic intervention to retard the progression of CKD and improve cardiovascular morbidity and mortality. Finerenone is a novel, nonsteroidal MR antagonist (MRA) with a unique mode of action that is distinct from currently available steroidal MRAs. In animal models of CKD, finerenone has a more favorable benefit/risk ratio as compared with the steroidal MRAs such as spironolactone and eplerenone. In patients with type 2 diabetes and heart and/or kidney disease, phase II trials have revealed that compared with spironolactone, eplerenone, or placebo, finerenone displays benefits that exceed the risks of MR antagonism. In patients with CKD and type 2 diabetes, a large phase III trial has shown that, compared with placebo, finerenone improved kidney failure and cardiovascular outcomes. In the first part of this article, we explore the safety and efficacy of spironolactone and eplerenone in early- and late-stage CKD. In the second part, we describe the mechanism of action of finerenone and discuss the promising role of this nonsteroidal MRA as a novel therapeutic opportunity to improve clinical outcomes in patients with CKD.
Collapse
Affiliation(s)
- Panagiotis I. Georgianos
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Rajiv Agarwal
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine and Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
29
|
Trujillo H, Caravaca-Fontán F, Caro J, Morales E, Praga M. The Forgotten Antiproteinuric Properties of Diuretics. Am J Nephrol 2021; 52:435-449. [PMID: 34233330 DOI: 10.1159/000517020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although diuretics are one of the most widely used drugs by nephrologists, their antiproteinuric properties are not generally taken into consideration. SUMMARY Thiazide diuretics have been shown to reduce proteinuria by >35% in several prospective controlled studies, and these values are markedly increased when combined with a low-salt diet. Thiazide-like diuretics (indapamide and chlorthalidone) have shown similar effectiveness. The antiproteinuric effect of mineralocorticoid receptor antagonists (spironolactone, eplerenone, and finerenone) has been clearly established through prospective and controlled studies, and treatment with finerenone reduces the risk of chronic kidney disease progression in type-2 diabetic patients. The efficacy of other diuretics such as amiloride, triamterene, acetazolamide, or loop diuretics has been less explored, but different investigations suggest that they might share the same antiproteinuric properties of other diuretics that should be evaluated through controlled studies. Although the inclusion of sodium-glucose cotransporter-2 inhibitors (SGLT2i) among diuretics is a controversial issue, their renoprotective and cardioprotective properties, confirmed in various landmark trials, constitute a true revolution in the treatment of patients with kidney disease. Recent subanalyses of these trials have shown that the early antiproteinuric effect induced by SGLT2i predicts long-term preservation of kidney function. Key Message: Whether the early reduction in proteinuria induced by diuretics other than finerenone and SGLT2i, as summarized in this review, also translates into long-term renoprotection requires further prospective and observational studies. In any case, it is important for the clinician to be aware of the antiproteinuric properties of drugs so often used in daily clinical practice.
Collapse
Affiliation(s)
- Hernando Trujillo
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain,
| | | | - Jara Caro
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Enrique Morales
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
30
|
Morales E, Cravedi P, Manrique J. Management of Chronic Hyperkalemia in Patients With Chronic Kidney Disease: An Old Problem With News Options. Front Med (Lausanne) 2021; 8:653634. [PMID: 34150795 PMCID: PMC8213200 DOI: 10.3389/fmed.2021.653634] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Hyperkalemia is one of the main electrolyte disorders in patients with chronic kidney disease (CKD). The prevalence of hyperkalemia increases as the Glomerular Filtration Rate (GFR) declines. Although chronic hyperkalemia is not a medical emergency, it can have negative consequences for the adequate cardio-renal management in the medium and long term. Hyperkalemia is common in patients on renin-angiotensin-aldosterone system inhibitors (RAASi) or Mineralocorticoid Receptor Antagonists (MRAs) and can affect treatment optimization for hypertension, diabetes mellitus, heart failure (HF), and CKD. Mortality rates are higher with suboptimal dosing among patients with CKD, diabetes or HF compared with full RAASi dosing, and are the highest among patients who discontinue RAASis. The treatment of chronic hyperkalemia is still challenging. Therefore, in the real world, discontinuation or reduction of RAASi therapy may lead to adverse cardiorenal outcomes, and current guidelines differ with regard to recommendations on RAASi therapy to enhance cardio and reno-protective effects. Treatment options for hyperkalemia have not changed much since the introduction of the cation exchange resin over 50 years ago. Nowadays, two new potassium binders, Patiromer Sorbitex Calcium, and Sodium Zirconium Cyclosilicate (SZC) already approved by FDA and by the European Medicines Agency, have demonstrated their clinical efficacy in reducing serum potassium with a good safety profile. The use of the newer potassium binders may allow continuing and optimizing RAASi therapy in patients with hyperkalemia keeping the cardio-renal protective effect in patients with CKD and cardiovascular disease. However, further research is needed to address some questions related to potassium disorders (definition of chronic hyperkalemia, monitoring strategies, prediction score for hyperkalemia or length for treatment).
Collapse
Affiliation(s)
- Enrique Morales
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Joaquin Manrique
- Nephrology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| |
Collapse
|
31
|
Update on Treatment of Hypertension After Renal Transplantation. Curr Hypertens Rep 2021; 23:25. [PMID: 33961145 DOI: 10.1007/s11906-021-01151-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW To incorporate novel findings on pathophysiology and treatment of posttransplant hypertension. RECENT FINDINGS (1) The sodium retaining effects of CNIs are mediated by stimulation of the thiazide-sensitive sodium chloride co-transporter in the distal convoluted tubule and in this regard chlorthalidone was proven to be an effective antihypertensive drug in renal transplantation. (2) Local and not systemic activation of the renin-angiotensin-aldosterone system plays a crucial role in the pathogenesis of posttransplant hypertension. (3) Recent randomized controlled trials failed to prove the presumed superiority of renin-angiotensin blockers in kidney transplantation. (4) Steroid-free and mammalian target of rapamycin-based immunosuppressive drug combinations did not show favorable effects on blood pressure control. (5) In a recent report the risk of non-melanoma skin cancer was higher with thiazide diuretics. But the increased cancer risk in transplant recipients is mainly attributed to comorbidities, such as diabetes and hypertension and of course to the transplantation condition itself or the obligatory application of immunosuppression, and has little to do with the antihypertensive medication Actual recommendations about BP targets in adult renal transplant recipients are coming from a post hoc analysis of a large randomized trial with another primary endpoint. Unless convincing studies on treatment of hypertension after renal transplantation are available, the ESC/ESH Guidelines 2018 should apply for these patients.
Collapse
|
32
|
Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors. Nat Rev Nephrol 2021; 17:319-334. [PMID: 33547417 DOI: 10.1038/s41581-021-00393-8] [Citation(s) in RCA: 263] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2021] [Indexed: 01/30/2023]
Abstract
Diabetic kidney disease is the leading cause of kidney failure worldwide; in the USA, it accounts for over 50% of individuals entering dialysis or transplant programmes. Unlike other complications of diabetes, the prevalence of diabetic kidney disease has failed to decline over the past 30 years. Hyperglycaemia is the primary aetiological factor responsible for the development of diabetic kidney disease. Once hyperglycaemia becomes established, multiple pathophysiological disturbances, including hypertension, altered tubuloglomerular feedback, renal hypoxia, lipotoxicity, podocyte injury, inflammation, mitochondrial dysfunction, impaired autophagy and increased activity of the sodium-hydrogen exchanger, contribute to progressive glomerular sclerosis and the decline in glomerular filtration rate. The quantitative contribution of each of these abnormalities to the progression of diabetic kidney disease, as well as their role in type 1 and type 2 diabetes mellitus, remains to be determined. Sodium-glucose co-transporter 2 (SGLT2) inhibitors have a beneficial impact on many of these pathophysiological abnormalities; however, as several pathophysiological disturbances contribute to the onset and progression of diabetic kidney disease, multiple agents used in combination will likely be required to slow the progression of disease effectively.
Collapse
|
33
|
Scholtes RA, van Baar MJB, Kok MD, Bjornstad P, Cherney DZI, Joles JA, van Raalte DH. Renal haemodynamic and protective effects of renoactive drugs in type 2 diabetes: Interaction with SGLT2 inhibitors. Nephrology (Carlton) 2021; 26:377-390. [PMID: 33283420 PMCID: PMC8026736 DOI: 10.1111/nep.13839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022]
Abstract
Diabetic kidney disease remains the leading cause of end-stage kidney disease and a major risk factor for cardiovascular disease. Large cardiovascular outcome trials and dedicated kidney trials have shown that sodium-glucose cotransporter (SGLT)2 inhibitors reduce cardiovascular morbidity and mortality and attenuate hard renal outcomes in patients with type 2 diabetes (T2D). Underlying mechanisms explaining these renal benefits may be mediated by decreased glomerular hypertension, possibly by vasodilation of the post-glomerular arteriole. People with T2D often receive several different drugs, some of which could also impact the renal vasculature, and could therefore modify both renal efficacy and safety of SGLT2 inhibition. The most commonly prescribed drugs that could interact with SGLT2 inhibitors on renal haemodynamic function include renin-angiotensin system inhibitors, calcium channel blockers and diuretics. Herein, we review the effects of these drugs on renal haemodynamic function in people with T2D and focus on studies that measured glomerular filtration rate (GFR) and effective renal plasma flow (ERPF) with gold-standard techniques. In addition, we posit, based on these observations, potential interactions with SGLT2 inhibitors with an emphasis on efficacy and safety.
Collapse
Affiliation(s)
- Rosalie A. Scholtes
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Michaël J. B. van Baar
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Megan D. Kok
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
| | - Petter Bjornstad
- Department of Pediatrics, Division of EndocrinologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of Medicine, Division of NephrologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - David Z. I. Cherney
- Department of Medicine and Department of Physiology, Division of Nephrology, University Health NetworkUniversity of TorontoTorontoOntarioCanada
| | - Jaap A. Joles
- Department of Nephrology and HypertensionUniversity Medical CenterUtrechtThe Netherlands
| | - Daniël H. van Raalte
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical CenterVU University Medical CenterAmsterdamThe Netherlands
- Department of Vascular Medicine, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
34
|
Jan RL, Weng SF, Wang JJ, Chang YS. Association between chronic kidney disease and the most common corneal ectasia disease (keratoconus): a nationwide cohort study. BMC Nephrol 2021; 22:109. [PMID: 33765939 PMCID: PMC7992838 DOI: 10.1186/s12882-021-02307-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 03/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Both keratoconus (KCN) and chronic kidney disease (CKD) are multifactorial conditions with multiple aetiologies and share several common pathophysiologies. However, the few studies that have described the relationship between KCN and CKD are limited to case reports and small case series. This study aimed to evaluate the association between KCN and CKD. METHODS The study cohort included 4,609 new-onset keratoconus patients ≥ 12 years identified by the International Classification of Diseases, Ninth Revision, Clinical Modification, code 371.6 and recruited between 2004 and 2011 from the Taiwan National Health Insurance Research Database. The age-, sex-, and comorbidity-matched control group included 27,654 non-KCN patients, selected from the Taiwan Longitudinal Health Insurance Database, 2000. Information for each patient was collected and tracked from the index date until December 2013. The incidence and risk of CKD were compared between the two groups. The adjusted hazard ratios (HRs) for CKD were calculated with Cox proportional hazard regression analysis. Kaplan-Meier analysis was used to calculate the cumulative CKD incidence rate. RESULTS The incidence rate of CKD was 1.36 times higher in KCN patients than in controls without statistically significant difference (95 % confidence interval [CI] = 0.99-1.86, p = 0.06). In total, 29 male KCN patients and 90 male controls developed CKD during the follow-up period. The incidence rate of CKD was 1.92 times (95 % [CI] = 1.26-2.91; p = 0.002) higher in male KCN patients than in controls. After adjusting for potential confounders, including age, hypertension, hyperlipidaemia, and diabetes mellitus, male KCN patients were 1.75 times (adjusted HR = 1.75, 95 % [CI] = 1.14-2.68, p < 0.05) more likely to develop CKD. CONCLUSIONS We found that male KCN patients have an increased risk of CKD. Therefore, it is recommended that male KCN patients should be aware of CKD.
Collapse
Affiliation(s)
- Ren-Long Jan
- Graduate Institute of Medical Science, College of Health Science, Chang Jung Christian University, Tainan, Taiwan
- Department of Pediatrics, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Medical Informatics and Statistics, Office of R&D, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jhi-Joung Wang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
- AI Biomed Center, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Yuh-Shin Chang
- Graduate Institute of Medical Science, College of Health Science, Chang Jung Christian University, Tainan, Taiwan
- Department of Ophthalmology, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
35
|
Hasegawa T, Nishiwaki H, Ota E, Levack WM, Noma H. Aldosterone antagonists for people with chronic kidney disease requiring dialysis. Cochrane Database Syst Rev 2021; 2:CD013109. [PMID: 33586138 PMCID: PMC8094170 DOI: 10.1002/14651858.cd013109.pub2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND People with chronic kidney disease (CKD) requiring dialysis are at a particularly high risk of cardiovascular death and morbidity. Several clinical studies suggested that aldosterone antagonists would be a promising treatment option for people undergoing dialysis. However, the clinical efficacy and potential harm of aldosterone antagonists for people with CKD on dialysis has yet to be determined. OBJECTIVES This review aimed to evaluate the benefits and harms of aldosterone antagonists, both non-selective (spironolactone) and selective (eplerenone), in comparison to control (placebo or standard care) in people with CKD requiring haemodialysis (HD) or peritoneal dialysis (PD). SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 5 August 2020 using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA We included parallel randomised controlled trials (RCTs), cross-over RCTs, and quasi-RCTs (where group allocation is by a method that is not truly random, such as alternation, assignment based on alternate medical records, date of birth, case record number, or other predictable methods) that compared aldosterone antagonists with placebo or standard care in people with CKD requiring dialysis. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed risk of bias for included studies. We used a random-effects model meta-analysis to perform a quantitative synthesis of the data. We used the I² statistic to measure heterogeneity among the studies in each analysis. We indicated summary estimates as a risk ratio (RR) for dichotomous outcomes, mean difference (MD) for continuous outcomes, or standardised mean differences (SMD) if different scales were used, with their 95% confidence interval (CI). We assessed the certainty of the evidence for each of the main outcomes using the GRADE (Grades of Recommendation, Assessment, Development, and Evaluation) approach. MAIN RESULTS We included 16 studies (14 parallel RCTs and two cross-over RCTs) involving a total of 1446 participants. Thirteen studies compared spironolactone to placebo or standard care and one study compared eplerenone to a placebo. Most included studies had an unclear or high risk of bias. Compared to control, aldosterone antagonists probably reduced the risk of death (any cause) for people with CKD requiring dialysis (9 studies, 1119 participants: RR 0.45, 95% CI 0.30 to 0.67; I² = 0%; moderate certainty of evidence). Aldosterone antagonist probably decreased the risk of death due to cardiovascular disease (6 studies, 908 participants: RR 0.37, 95% CI 0.22 to 0.64; I² = 0%; moderate certainty of evidence) and cardiovascular and cerebrovascular morbidity (3 studies, 328 participants: RR 0.38, 95% CI 0.18 to 0.76; I² = 0%; moderate certainty of evidence). While aldosterone antagonists probably increased risk of gynaecomastia compared with control (4 studies, 768 participants: RR 5.95, 95% CI 1.93 to 18.3; I² = 0%; moderate certainty of evidence), aldosterone antagonists may make little or no difference to the risk of hyperkalaemia (9 studies, 981 participants: RR 1.41, 95% CI 0.72 to 2.78; I² = 47%; low certainty of evidence). Aldosterone antagonists had a marginal effect on left ventricular mass among participants undergoing dialysis (8 studies, 633 participants: SMD -0.42, 95% CI -0.78 to 0.05; I² = 77%). In people with CKD requiring dialysis received aldosterone antagonists compared to control, there were 72 fewer deaths from all causes per 1000 participants (95% CI 47 to 98) with a number needed to treat for an additional beneficial outcome (NNTB) of 14 (95% CI 10 to 21) and for gynaecomastia were 26 events per 1000 participants (95% CI 15 to 39) with a number need to treat for an additional harmful outcome (NNTH) of 38 (95% CI 26 to 68). AUTHORS' CONCLUSIONS Based on moderate certainty of the evidence, aldosterone antagonists probably reduces the risk of all-cause and cardiovascular death and probably reduces morbidity due to cardiovascular and cerebrovascular disease in people with CKD requiring dialysis. For the adverse effect of gynaecomastia, the risk was increased compared to control. For this outcome, the absolute risk was lower than the absolute risk of death. It is hoped the three large ongoing studies will provide better certainty of evidence.
Collapse
Affiliation(s)
- Takeshi Hasegawa
- Showa University Research Administration Center (SURAC), Showa University, Tokyo, Japan
- Division of Nephrology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Hiroki Nishiwaki
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Erika Ota
- Global Health Nursing, Graduate School of Nursing Science, St. Luke's International University, Tokyo, Japan
| | - William Mm Levack
- Rehabilitation Teaching and Research Unit, Department of Medicine, University of Otago, Wellington, New Zealand
| | - Hisashi Noma
- Department of Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
| |
Collapse
|
36
|
Khoo CM, Deerochanawong C, Chan SP, Matawaran B, Sheu WH, Chan J, Mithal A, Luk A, Suastika K, Yoon K, Ji L, Man NH, Pollock C. Use of sodium-glucose co-transporter-2 inhibitors in Asian patients with type 2 diabetes and kidney disease: An Asian perspective and expert recommendations. Diabetes Obes Metab 2021; 23:299-317. [PMID: 33155749 PMCID: PMC7839543 DOI: 10.1111/dom.14251] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/17/2020] [Accepted: 11/01/2020] [Indexed: 12/14/2022]
Abstract
Early onset of type 2 diabetes and a high prevalence of co-morbidities predispose the Asian population to a high risk for, and rapid progression of, diabetic kidney disease (DKD). Apart from renin-angiotensin system inhibitors, sodium-glucose co-transporter-2 (SGLT-2) inhibitors have been shown to delay renal disease progression in patients with DKD. In this review article, we consolidate the existing literature on SGLT-2 inhibitor use in Asian patients with DKD to establish contemporary guidance for clinicians. We extensively reviewed recommendations from international and regional guidelines, data from studies on Asian patients with DKD, global trials (DAPA-CKD, CREDENCE and DELIGHT) and cardiovascular outcomes trials. In patients with DKD, SGLT-2 inhibitor therapy significantly reduced albuminuria and the risk of hard renal outcomes (defined as the onset of end-stage kidney disease, substantial decline in renal function from baseline and renal death), cardiovascular outcomes and hospitalization for heart failure. In all the cardiovascular and renal outcomes trials, there was an initial decline in the estimated glomerular filtration rate (eGFR), which was followed by a slowing in the decline of renal function compared with that seen with placebo. Despite an attenuation in glucose-lowering efficacy in patients with low eGFR, there were sustained reductions in body weight and blood pressure, and an increase in haematocrit. Based on the available evidence, we conclude that SGLT-2 inhibitors represent an evidence-based therapeutic option for delaying the progression of renal disease in Asian patients with DKD and preserving renal function in patients at high risk of kidney disease.
Collapse
Affiliation(s)
| | | | - Siew Pheng Chan
- Department of MedicineUniversity of Malaya Medical CenterKuala LumpurMalaysia
| | - Bien Matawaran
- Department of Medicine, Section of Endocrinology, Diabetes and MetabolismUniversity of Santo Tomas HospitalManilaPhilippines
| | - Wayne Huey‐Herng Sheu
- Division of Endocrinology and Metabolism, Department of Internal MedicineTaichung Veterans General HospitalTaichungTaiwan
| | - Juliana Chan
- Department of Medicine and TherapeuticsHong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales HospitalHong Kong
| | | | - Andrea Luk
- Department of Medicine and TherapeuticsHong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales HospitalHong Kong
| | - Ketut Suastika
- Faculty of MedicineUdayana University, Sanglah General HospitalBaliIndonesia
| | - Kun‐Ho Yoon
- Department of Endocrinology & Metabolism, Seoul St Maryʼs HospitalThe Catholic University of KoreaSeoulSouth Korea
| | - Linong Ji
- Peking University Peopleʼs HospitalPekingChina
| | | | - Carol Pollock
- The University of Sydney School of MedicineSydneyNew South WalesAustralia
| |
Collapse
|
37
|
Agarwal R, Kolkhof P, Bakris G, Bauersachs J, Haller H, Wada T, Zannad F. Steroidal and non-steroidal mineralocorticoid receptor antagonists in cardiorenal medicine. Eur Heart J 2021; 42:152-161. [PMID: 33099609 PMCID: PMC7813624 DOI: 10.1093/eurheartj/ehaa736] [Citation(s) in RCA: 267] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/08/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
This review covers the last 80 years of remarkable progress in the development of mineralocorticoid receptor (MR) antagonists (MRAs) from synthesis of the first mineralocorticoid to trials of nonsteroidal MRAs. The MR is a nuclear receptor expressed in many tissues/cell types including the kidney, heart, immune cells, and fibroblasts. The MR directly affects target gene expression-primarily fluid, electrolyte and haemodynamic homeostasis, and also, but less appreciated, tissue remodelling. Pathophysiological overactivation of the MR leads to inflammation and fibrosis in cardiorenal disease. We discuss the mechanisms of action of nonsteroidal MRAs and how they differ from steroidal MRAs. Nonsteroidal MRAs have demonstrated important differences in their distribution, binding mode to the MR and subsequent gene expression. For example, the novel nonsteroidal MRA finerenone has a balanced distribution between the heart and kidney compared with spironolactone, which is preferentially concentrated in the kidneys. Compared with eplerenone, equinatriuretic doses of finerenone show more potent anti-inflammatory and anti-fibrotic effects on the kidney in rodent models. Overall, nonsteroidal MRAs appear to demonstrate a better benefit-risk ratio than steroidal MRAs, where risk is measured as the propensity for hyperkalaemia. Among patients with Type 2 diabetes, several Phase II studies of finerenone show promising results, supporting benefits on the heart and kidneys. Furthermore, finerenone significantly reduced the combined primary endpoint (chronic kidney disease progression, kidney failure, or kidney death) vs. placebo when added to the standard of care in a large Phase III trial.
Collapse
Affiliation(s)
- Rajiv Agarwal
- Indiana University School of Medicine and VA Medical Center, 1481 West 10th Street, 111N Indianapolis, IN 46202, USA
| | - Peter Kolkhof
- R&D Preclinical Research Cardiovascular, Bayer AG, Wuppertal, Germany
| | - George Bakris
- American Society of Hypertension's Comprehensive Hypertension Center at the University of Chicago Medicine, Chicago, IL, USA
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Faiez Zannad
- Centre d’Investigations Cliniques Plurithématique, University Henri Poincaré, Nancy, France
| |
Collapse
|
38
|
Chung EY, Ruospo M, Natale P, Bolignano D, Navaneethan SD, Palmer SC, Strippoli GF. Aldosterone antagonists in addition to renin angiotensin system antagonists for preventing the progression of chronic kidney disease. Cochrane Database Syst Rev 2020; 10:CD007004. [PMID: 33107592 PMCID: PMC8094274 DOI: 10.1002/14651858.cd007004.pub4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Treatment with angiotensin-converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) is used to reduce proteinuria and retard the progression of chronic kidney disease (CKD). However, resolution of proteinuria may be incomplete with these therapies and the addition of an aldosterone antagonist may be added to further prevent progression of CKD. This is an update of a Cochrane review first published in 2009 and updated in 2014. OBJECTIVES To evaluate the effects of aldosterone antagonists (selective (eplerenone), non-selective (spironolactone or canrenone), or non-steroidal mineralocorticoid antagonists (finerenone)) in adults who have CKD with proteinuria (nephrotic and non-nephrotic range) on: patient-centred endpoints including kidney failure (previously know as end-stage kidney disease (ESKD)), major cardiovascular events, and death (any cause); kidney function (proteinuria, estimated glomerular filtration rate (eGFR), and doubling of serum creatinine); blood pressure; and adverse events (including hyperkalaemia, acute kidney injury, and gynaecomastia). SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 13 January 2020 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs that compared aldosterone antagonists in combination with ACEi or ARB (or both) to other anti-hypertensive strategies or placebo in participants with proteinuric CKD. DATA COLLECTION AND ANALYSIS Two authors independently assessed study quality and extracted data. Data were summarised using random effects meta-analysis. We expressed summary treatment estimates as a risk ratio (RR) for dichotomous outcomes and mean difference (MD) for continuous outcomes, or standardised mean difference (SMD) when different scales were used together with their 95% confidence interval (CI). Risk of bias were assessed using the Cochrane tool. Evidence certainty was evaluated using GRADE. MAIN RESULTS Forty-four studies (5745 participants) were included. Risk of bias in the evaluated methodological domains were unclear or high risk in most studies. Adequate random sequence generation was present in 12 studies, allocation concealment in five studies, blinding of participant and investigators in 18 studies, blinding of outcome assessment in 15 studies, and complete outcome reporting in 24 studies. All studies comparing aldosterone antagonists to placebo or standard care were used in addition to an ACEi or ARB (or both). None of the studies were powered to detect differences in patient-level outcomes including kidney failure, major cardiovascular events or death. Aldosterone antagonists had uncertain effects on kidney failure (2 studies, 84 participants: RR 3.00, 95% CI 0.33 to 27.65, I² = 0%; very low certainty evidence), death (3 studies, 421 participants: RR 0.58, 95% CI 0.10 to 3.50, I² = 0%; low certainty evidence), and cardiovascular events (3 studies, 1067 participants: RR 0.95, 95% CI 0.26 to 3.56; I² = 42%; low certainty evidence) compared to placebo or standard care. Aldosterone antagonists may reduce protein excretion (14 studies, 1193 participants: SMD -0.51, 95% CI -0.82 to -0.20, I² = 82%; very low certainty evidence), eGFR (13 studies, 1165 participants, MD -3.00 mL/min/1.73 m², 95% CI -5.51 to -0.49, I² = 0%, low certainty evidence) and systolic blood pressure (14 studies, 911 participants: MD -4.98 mmHg, 95% CI -8.22 to -1.75, I² = 87%; very low certainty evidence) compared to placebo or standard care. Aldosterone antagonists probably increase the risk of hyperkalaemia (17 studies, 3001 participants: RR 2.17, 95% CI 1.47 to 3.22, I² = 0%; moderate certainty evidence), acute kidney injury (5 studies, 1446 participants: RR 2.04, 95% CI 1.05 to 3.97, I² = 0%; moderate certainty evidence), and gynaecomastia (4 studies, 281 participants: RR 5.14, 95% CI 1.14 to 23.23, I² = 0%; moderate certainty evidence) compared to placebo or standard care. Non-selective aldosterone antagonists plus ACEi or ARB had uncertain effects on protein excretion (2 studies, 139 participants: SMD -1.59, 95% CI -3.80 to 0.62, I² = 93%; very low certainty evidence) but may increase serum potassium (2 studies, 121 participants: MD 0.31 mEq/L, 95% CI 0.17 to 0.45, I² = 0%; low certainty evidence) compared to diuretics plus ACEi or ARB. Selective aldosterone antagonists may increase the risk of hyperkalaemia (2 studies, 500 participants: RR 1.62, 95% CI 0.66 to 3.95, I² = 0%; low certainty evidence) compared ACEi or ARB (or both). There were insufficient studies to perform meta-analyses for the comparison between non-selective aldosterone antagonists and calcium channel blockers, selective aldosterone antagonists plus ACEi or ARB (or both) and nitrate plus ACEi or ARB (or both), and non-steroidal mineralocorticoid antagonists and selective aldosterone antagonists. AUTHORS' CONCLUSIONS The effects of aldosterone antagonists when added to ACEi or ARB (or both) on the risks of death, major cardiovascular events, and kidney failure in people with proteinuric CKD are uncertain. Aldosterone antagonists may reduce proteinuria, eGFR, and systolic blood pressure in adults who have mild to moderate CKD but may increase the risk of hyperkalaemia, acute kidney injury and gynaecomastia when added to ACEi and/or ARB.
Collapse
Affiliation(s)
- Edmund Ym Chung
- Department of Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Marinella Ruospo
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Patrizia Natale
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Davide Bolignano
- Institute of Clinical Physiology, CNR - Italian National Council of Research, Reggio Calabria, Italy
| | | | - Suetonia C Palmer
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Giovanni Fm Strippoli
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
39
|
Abe T, Jujo K, Kametani M, Minami Y, Fukushima N, Saito K, Hagiwara N. Prognostic impact of additional mineralocorticoid receptor antagonists in octogenarian heart failure patients. ESC Heart Fail 2020; 7:2711-2724. [PMID: 32860346 PMCID: PMC7524245 DOI: 10.1002/ehf2.12862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/18/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS Guideline-directed medical therapy (GDMT) including beta-blockers and renin-angiotensin system inhibitors is shown to reduce mortality risk in patients with heart failure (HF) and reduced left ventricular ejection fraction (LVEF). However, there is little evidence about the efficacy of additional administration of mineralocorticoid receptor antagonists (MRAs) with GDMT in patients ≥80 years presenting with HF. We aimed to investigate the prognostic impact of GDMT with MRA in relation to the age of patients with HF. METHODS AND RESULTS This observational study included patients admitted for HF with reduced LVEF who were discharged alive; among them, 224 patients were ≥80 years, and 661 patients were <80 years. Both populations were divided into three groups depending on whether they received GDMT with or without MRA or single/no GDMT drugs (GDMT+MRA+, GDMT+MRA-, or non-GDMT, respectively). The primary endpoint was all-cause mortality. In patients ≥80 years, all-cause mortality was the lowest in the GDMT+MRA+ group (log-rank trend, P = 0.034), and no significant differences were observed between the GDMT+MRA- and non-GDMT groups. Multivariate Cox regression analysis revealed that GDMT+MRA+ was superior to GDMT+MRA-, even after adjusting for parameters at discharge (hazard ratio: 0.32, 95% confidence interval: 0.11-0.99). In patients <80 years, GDMT reduced all-cause mortality; however, additional MRA was not associated with an improved outcome. CONCLUSIONS The results of this study suggest that additional MRA to GDMT at discharge is one of the therapeutic options for elderly HF patients with reduced LVEF. This finding is not well documented in previous clinical trials.
Collapse
Affiliation(s)
- Takuro Abe
- Department of CardiologyTokyo Women's Medical University8‐1 Kawadacho, Shinjuku‐kuTokyo162‐0054Japan
- Department of CardiologyNishiarai Heart Center HospitalTokyoJapan
| | - Kentaro Jujo
- Department of CardiologyTokyo Women's Medical University8‐1 Kawadacho, Shinjuku‐kuTokyo162‐0054Japan
- Department of CardiologyNishiarai Heart Center HospitalTokyoJapan
| | - Motoko Kametani
- Department of CardiologyTokyo Women's Medical University8‐1 Kawadacho, Shinjuku‐kuTokyo162‐0054Japan
| | - Yuichiro Minami
- Department of CardiologyTokyo Women's Medical University8‐1 Kawadacho, Shinjuku‐kuTokyo162‐0054Japan
| | - Noritoshi Fukushima
- Department of Preventive Medicine and Public HealthTokyo Medical UniversityTokyoJapan
| | - Katsumi Saito
- Department of CardiologyNishiarai Heart Center HospitalTokyoJapan
| | - Nobuhisa Hagiwara
- Department of CardiologyTokyo Women's Medical University8‐1 Kawadacho, Shinjuku‐kuTokyo162‐0054Japan
| |
Collapse
|
40
|
Georgianos PI, Agarwal R. Resistant Hypertension in Chronic Kidney Disease (CKD): Prevalence, Treatment Particularities, and Research Agenda. Curr Hypertens Rep 2020; 22:84. [PMID: 32880742 DOI: 10.1007/s11906-020-01081-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To explore the prevalence, treatment particularities, and research agenda in the management of resistant hypertension among patients with chronic kidney disease (CKD). RECENT FINDINGS The prevalence of resistant hypertension is reported to be 2-3 times higher in patients with CKD than in the general hypertensive population. Based in part on the results of the PATHWAY-2 trial showing add-on spironolactone to be superior to placebo or active treatment with an α- or β-blocker in reducing BP, international guidelines recommend the use of spironolactone as fourth-line agent in pharmacotherapy of resistant hypertension. Despite the several-fold higher burden of resistant hypertension among patients with stage 3b-4 CKD, the use of spironolactone in this population has been restricted, mainly due to the risk of hyperkalemia. The recently reported AMBER trial showed that among patients with uncontrolled resistant hypertension and an estimated glomerular filtration rate of 25-45 ml/min/1.73m2, the newer potassium-binder patiromer prevented the development of hyperkalemia and increased the proportion of participants who remained on add-on spironolactone over 12 weeks of follow-up. Administration of spironolactone was associated with a clinically meaningful reduction of 11-12 mmHg in unattended automated office systolic blood pressure (BP) over the course of the AMBER trial. Newer potassium-binding therapies overcome the barrier of hyperkalemia and facilitate the persistent use of spironolactone, which is an effective add-on therapy to control BP in patients with resistant hypertension and advanced CKD. Future trials are now warranted to explore whether this strategy confers benefits on "hard" clinical outcomes in this high-risk population.
Collapse
Affiliation(s)
- Panagiotis I Georgianos
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Rajiv Agarwal
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine and Richard L. Roudebush Veterans Administration Medical Center, 1481 West 10th Street, Indianapolis, IN, USA.
| |
Collapse
|
41
|
Athavale A, Roberts DM. Management of proteinuria: blockade of the renin-angiotensin-aldosterone system. Aust Prescr 2020; 43:121-125. [PMID: 32921887 PMCID: PMC7450775 DOI: 10.18773/austprescr.2020.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proteinuria, in particular albuminuria, is a potentially significant modifiable risk factor for cardiovascular disease and the progression of kidney disease. Current treatment guidelines for albuminuria recommend a single renin–angiotensin–aldosterone inhibitor. This can be an ACE inhibitor or an angiotensin receptor antagonist. The routine use of combined renin–angiotensin–aldosterone inhibition for albuminuria is not supported by current evidence. Combination therapy is associated with higher rates of adverse events such as hyperkalaemia and progressive renal impairment.
Collapse
Affiliation(s)
- Akshay Athavale
- Drug Health Services and Clinical Pharmacology and Toxicology, Royal Prince Alfred Hospital, Sydney
- Departments of Clinical Pharmacology and Toxicology, and Renal Medicine and Transplantation, St Vincent's Hospital, Sydney
| | - Darren M Roberts
- Drug Health Services and Clinical Pharmacology and Toxicology, Royal Prince Alfred Hospital, Sydney
- Departments of Clinical Pharmacology and Toxicology, and Renal Medicine and Transplantation, St Vincent's Hospital, Sydney
| |
Collapse
|
42
|
Blood pressure reduction and RAAS inhibition in diabetic kidney disease: therapeutic potentials and limitations. J Nephrol 2020; 33:949-963. [PMID: 32681470 PMCID: PMC7557495 DOI: 10.1007/s40620-020-00803-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022]
Abstract
Diabetic kidney disease (DKD) affects approximately one-third of patients with diabetes and taking into consideration the high cardiovascular risk burden associated to this condition a multifactorial therapeutic approach is traditionally recommended, in which glucose and blood pressure control play a central role. The inhibition of renin–angiotensin–aldosterone RAAS system represent traditionally the cornerstone of DKD. Clinical outcome trials have demonstrated clinical significant benefit in slowing nephropathy progression mainly in the presence of albuminuria. Thus, international guidelines mandate their use in such patients. Given the central role of RAAS activity in the pathogenesis and progression of renal and cardiovascular damage, a more profound inhibition of the system by the use of multiple agents has been proposed in the past, especially in the presence of proteinuria, however clinical trials have failed to confirm the usefulness of this therapeutic approach. Furthermore, whether strict blood pressure control and pharmacologic RAAS inhibition entails a favorable renal outcome in non-albuminuric patients is at present unclear. This aspect is becoming an important issue in the management of DKD since nonalbuminuric DKD is currently the prevailing presenting phenotype. For these reasons it would be advisable that blood pressure management should be tailored in each subject on the basis of the renal phenotype as well as related comorbidities. This article reviews the current literature and discusses potentials and limitation of targeting the RAAS in order to provide the greatest renal protection in DKD.
Collapse
|
43
|
Blankenburg M, Kovesdy CP, Fett AK, Griner RG, Gay A. Disease characteristics and outcomes in patients with chronic kidney disease and type 2 diabetes: a matched cohort study of spironolactone users and non-users. BMC Nephrol 2020; 21:61. [PMID: 32101152 PMCID: PMC7045439 DOI: 10.1186/s12882-020-01719-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 02/11/2020] [Indexed: 01/13/2023] Open
Abstract
Background Limited evidence has indicated that addition of a steroidal mineralocorticoid receptor antagonist (MRA) to the standard of care reduces proteinuria in patients with diabetic kidney disease (DKD); however, there are limited data regarding real-world MRA use in these patients. This study aimed to describe the characteristics of spironolactone users and non-users with DKD, and to explore their clinical outcomes. Methods This was a non-interventional, retrospective cohort study using demographic and clinical data from a US claims database (PharMetrics Plus) and the Experian consumer data asset during 2006–2015. Baseline characteristics (e.g. comorbidities) and post-inclusion clinical outcomes were described in matched cohorts of spironolactone users and non-users (n = 5465 per group). Results Although matching aligned key demographic and clinical characteristics of the cohorts, a significantly greater proportion of spironolactone users than non-users had oedema, proteinuria, and cardiovascular disease at baseline (P < 0.0001). During the post-inclusion period, disease progression and clinical events of interest such as acute kidney injury were more commonly observed in spironolactone users than non-users. Users also had higher healthcare resource utilization and costs than non-users; however, these differences diminished at later stages of disease. Conclusions In this study, spironolactone users had a greater comorbidity burden at baseline than matched non-users, suggesting that the presence of certain comorbidities may be contributing factors in the decision to prescribe spironolactone. High healthcare resource utilization and costs for patients at later stages of disease, irrespective of spironolactone use, highlight the need for new therapies for DKD.
Collapse
Affiliation(s)
- Michael Blankenburg
- Market Access, Public Affairs & Sustainability, Pharmaceuticals, Bayer AG, Berlin, Germany.
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | - Alain Gay
- Medical Affairs & Pharmacovigilance, Pharmaceuticals, Bayer AG, Berlin, Germany
| |
Collapse
|
44
|
Bădilă E. The expanding class of mineralocorticoid receptor modulators: New ligands for kidney, cardiac, vascular, systemic and behavioral selective actions. ACTA ENDOCRINOLOGICA-BUCHAREST 2020; 16:487-496. [PMID: 34084241 DOI: 10.4183/aeb.2020.487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This paper reviews the class of mineralocorticoid receptor (MR) modulators, especially new nonsteroidal antagonists. MR is a nuclear receptor expressed in many tissues and cell types. Aldosterone, the most important mineralocorticoid hormone and MR agonist, has many unfavorable effects, especially on the heart, blood vessels, and kidneys, by promoting fibrosis and tissue remodelling. Classical synthetic MR antagonists (spironolactone, eplerenone) have proven useful in clinical practice through their antihypertensive effects in resistant forms, and through benefits on morbidity and mortality in heart failure with reduced ejection fraction. These benefits are associated with important side effects, hyperkalemia being the main limitation. In the latest years, a new generation of MR modulators with a nonsteroidal structure has emerged. These compounds are more selective than classical MR antagonists, with much higher affinity for the MR than for the glucocorticoid, androgen, or progesterone receptors. Recent clinical and experimental observations suggest that nonsteroidal MR antagonists, especially finerenone, have proven superior renoprotective properties, antiproteinuric efficacy, inhibition of inflammation and heart fibrosis in animal models, without sharing the side effects of steroidal MR antagonists. Nonsteroidal MR modulators represent an interesting new therapeutic approach for the prevention and progression of chronic kidney disease and for patients with heart failure and renal disease. Despite these promising data, there are still many issues to be clarified and it is necessary to accumulate solid evidence from studies on larger numbers of patients and from head-to-head clinical trials.
Collapse
Affiliation(s)
- E Bădilă
- "Carol Davila" University of Medicine and Pharmacy, Clinical Emergency Hospital, Bucharest, Romania
| |
Collapse
|
45
|
Vidal-Petiot E, Metzger M, Faucon AL, Boffa JJ, Haymann JP, Thervet E, Houillier P, Geri G, Stengel B, Vrtovsnik F, Flamant M. Extracellular Fluid Volume Is an Independent Determinant of Uncontrolled and Resistant Hypertension in Chronic Kidney Disease: A NephroTest Cohort Study. J Am Heart Assoc 2019; 7:e010278. [PMID: 30371309 PMCID: PMC6404875 DOI: 10.1161/jaha.118.010278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Hypertension is highly prevalent during chronic kidney disease (CKD) and, in turn, worsens CKD prognosis. We aimed to describe the determinants of uncontrolled and resistant hypertension during CKD. Methods and Results We analyzed baseline data from patients with CKD stage 1 to 5 (NephroTest cohort) who underwent thorough renal explorations, including measurements of glomerular filtration rate (clearance of 51Cr‐EDTA) and of extracellular water (volume of distribution of the tracer). Hypertension was defined as blood pressure (BP; average of 3 office measurements) ≥140/90 mm Hg or the use of antihypertensive drugs. In 2015 patients (mean age, 58.7±15.3 years; 67% men; mean glomerular filtration rate, 42±15 mL/min per 1.73 m2), prevalence of hypertension was 88%. Among hypertensive patients, 44% and 32% had uncontrolled (≥140/90 mm Hg) and resistant (uncontrolled BP despite 3 drugs, including a diuretic, or ≥4 drugs, including a diuretic, regardless of BP level) hypertension, respectively. In multivariable analysis, extracellular water, older age, higher albuminuria, diabetic nephropathy, and the absence of aldosterone blockers were independently associated with uncontrolled BP. Extracellular water, older age, lower glomerular filtration rate, higher albuminuria and body mass index, male sex, African origin, diabetes mellitus, and diabetic and glomerular nephropathies were associated with resistant hypertension. Conclusions In this large population of patients with CKD, a lower glomerular filtration rate, a higher body mass index, diabetic status, and African origin were associated with hypertension severity but not with BP control. Higher extracellular water, older age, and higher albuminuria were independent determinants of both resistant and uncontrolled hypertension during CKD. Our results advocate for the large use of diuretics in this population.
Collapse
Affiliation(s)
- Emmanuelle Vidal-Petiot
- 1 Physiology Department Assistance Publique-Hôpitaux de Paris Hôpital Bichat and Inserm U1149 Paris France.,2 Paris Diderot University Sorbonne Paris Cité Paris France
| | - Marie Metzger
- 3 Inserm Centre de recherche en Epidéemiologie et Santée des Populations U1018 Hôpital Paul Brousse Villejuif France
| | - Anne-Laure Faucon
- 3 Inserm Centre de recherche en Epidéemiologie et Santée des Populations U1018 Hôpital Paul Brousse Villejuif France
| | - Jean-Jacques Boffa
- 4 Nephrology Department APHP, Hôpital Tenon Paris France.,5 Pierre et Marie Curie University Paris France
| | - Jean-Philippe Haymann
- 5 Pierre et Marie Curie University Paris France.,6 Physiology Department APHP, Hôpital Tenon Paris France
| | - Eric Thervet
- 7 Nephrology Department APHP HEGP Paris France.,8 Paris Descartes University Sorbonne Paris Cité Paris France
| | - Pascal Houillier
- 1 Physiology Department Assistance Publique-Hôpitaux de Paris Hôpital Bichat and Inserm U1149 Paris France.,8 Paris Descartes University Sorbonne Paris Cité Paris France.,9 Physiology Department APHP Hôpital Tenon, Georges Pompidou Paris France.,10 INSERM UMR_S1138 Paris France
| | - Guillaume Geri
- 3 Inserm Centre de recherche en Epidéemiologie et Santée des Populations U1018 Hôpital Paul Brousse Villejuif France.,11 Intensive Care Unit APHP, Hopital Ambroise Paré Boulogne France.,12 Versailles Saint Quentin University Versailles France
| | - Bénédicte Stengel
- 3 Inserm Centre de recherche en Epidéemiologie et Santée des Populations U1018 Hôpital Paul Brousse Villejuif France
| | - François Vrtovsnik
- 2 Paris Diderot University Sorbonne Paris Cité Paris France.,13 Nephrology Department APHP, Hôpital Bichat Paris France
| | - Martin Flamant
- 1 Physiology Department Assistance Publique-Hôpitaux de Paris Hôpital Bichat and Inserm U1149 Paris France.,2 Paris Diderot University Sorbonne Paris Cité Paris France
| | | |
Collapse
|
46
|
Mavrakanas TA, Giannetti N, Sapir-Pichhadze R, Alam A. Mineralocorticoid Receptor Antagonists and Renal Outcomes in Heart Failure Patients with and without Chronic Kidney Disease. Cardiorenal Med 2019; 10:32-41. [PMID: 31665724 DOI: 10.1159/000503223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/05/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The effect of mineralocorticoid receptor antagonists (MRAs) on chronic kidney disease (CKD) progression in patients with heart failure (HF) and with or without preexisting CKD has not been adequately studied. METHODS We conducted a retrospective cohort study including consecutive adult patients followed at the HF clinic of a tertiary care center who had already been on an angiotensin-converting enzyme inhibitor (ACEI) or an angiotensin receptor blocker (ARB). Exposure to MRAs was assessed at 6 months from registration. Patients who were never exposed to an MRA were the control group. RESULTS A total of 314 patients who were prescribed an MRA were compared to 1,116 patients who never received an MRA. Among them, 121 and 408 patients, respectively, had CKD (estimated glomerular filtration rate <60 mL/min/1.73 m2). MRAs had to be discontinued in 36/121 patients with CKD (29.8%) and 57/165 patients without CKD (34.5%) (p = 0.39). MRA treatment was associated with a higher risk for persistent creatinine doubling among patients without CKD (hazard ratio 4.07, 95% confidence interval 1.41-11.73). A numerically lower risk was identified among CKD patients (hazard ratio 0.33, 95% confidence interval 0.04-2.78) (p for interaction = 0.009). The primary safety outcome, a composite of any doubling of serum creatinine or any episode of serious hyperkalemia (K+ >6 mmol/L), occurred more commonly in MRA users compared with nonusers in the subgroup of patients without CKD, but not in CKD patients (p for interaction = 0.02). CONCLUSION MRA treatment in addition to an ACEI or an ARB could be safely prescribed in HF patients with CKD as it is not associated with persistent renal function decline, acute kidney injury, or serious hyperkalemia, compared with ACEI/ARB monotherapy.
Collapse
Affiliation(s)
- Thomas A Mavrakanas
- Divisions of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Québec, Canada, .,Department of Medicine, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland,
| | - Nadia Giannetti
- Division of Cardiology, Department of Medicine, McGill University Health Centre, Montreal, Québec, Canada
| | - Ruth Sapir-Pichhadze
- Divisions of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Québec, Canada
| | - Ahsan Alam
- Divisions of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Québec, Canada
| |
Collapse
|
47
|
Ruilope LM, Agarwal R, Anker SD, Bakris GL, Filippatos G, Nowack C, Kolkhof P, Joseph A, Mentenich N, Pitt B. Design and Baseline Characteristics of the Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease Trial. Am J Nephrol 2019; 50:345-356. [PMID: 31665733 DOI: 10.1159/000503712] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Among people with diabetes, those with kidney disease have exceptionally high rates of cardiovascular (CV) morbidity and mortality and progression of their underlying kidney disease. Finerenone is a novel, nonsteroidal, selective mineralocorticoid receptor antagonist that has shown to reduce albuminuria in type 2 diabetes (T2D) patients with chronic kidney disease (CKD) while revealing only a low risk of hyperkalemia. However, the effect of finerenone on CV and renal outcomes has not yet been investigated in long-term trials. PATIENTS AND METHODS The Finerenone in Reducing CV Mortality and Morbidity in Diabetic Kidney Disease (FIGARO-DKD) trial aims to assess the efficacy and safety of finerenone compared to placebo at reducing clinically important CV and renal outcomes in T2D patients with CKD. FIGARO-DKD is a randomized, double-blind, placebo-controlled, parallel-group, event-driven trial running in 47 countries with an expected duration of approximately 6 years. FIGARO-DKD randomized 7,437 patients with an estimated glomerular filtration rate ≥25 mL/min/1.73 m2 and albuminuria (urinary albumin-to-creatinine ratio ≥30 to ≤5,000 mg/g). The study has at least 90% power to detect a 20% reduction in the risk of the primary outcome (overall two-sided significance level α = 0.05), the composite of time to first occurrence of CV death, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure. CONCLUSIONS FIGARO-DKD will determine whether an optimally treated cohort of T2D patients with CKD at high risk of CV and renal events will experience cardiorenal benefits with the addition of finerenone to their treatment regimen. TRIAL REGISTRATION EudraCT number: 2015-000950-39; ClinicalTrials.gov identifier: NCT02545049.
Collapse
Affiliation(s)
- Luis M Ruilope
- Cardiorenal Translational Laboratory and Hypertension Unit, Institute of Research imas12, Madrid, Spain,
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain,
- Faculty of Sport Sciences, European University of Madrid, Madrid, Spain,
| | - Rajiv Agarwal
- Richard L. Roudebush VA Medical Center and Indiana University, Indianapolis, Indiana, USA
| | - Stefan D Anker
- Department of Cardiologsupply, and Berlin Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research Partner Site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - George L Bakris
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Gerasimos Filippatos
- Department of Cardiology, Attikon University Hospital, Athens, Greece, and University of Cyprus, Medical School, Nicosia, Cyprus
| | - Christina Nowack
- Research and Development, Clinical Development Operations, Bayer AG, Wuppertal, Germany
| | - Peter Kolkhof
- Research and Development, Preclinical Research Cardiovascular, Bayer AG, Wuppertal, Germany
| | - Amer Joseph
- Research and Development, Clinical Development, Bayer AG, Berlin, Germany
| | - Nicole Mentenich
- Research and Development, Statistics and Data Insights, Bayer AG, Wuppertal, Germany
| | - Bertram Pitt
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Whittaker A, Kragh ÅM, Hartleib-Geschwindner J, Albayaty M, Backlund A, Greasley PJ, Heijer M, Kjaer M, Forte P, Unwin R, Wernevik L, Ericsson H. Safety, Tolerability, and Pharmacokinetics of the Mineralocorticoid Receptor Modulator AZD9977 in Healthy Men: A Phase I Multiple Ascending Dose Study. Clin Transl Sci 2019; 13:275-283. [PMID: 31584739 PMCID: PMC7070793 DOI: 10.1111/cts.12705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/13/2019] [Indexed: 12/22/2022] Open
Abstract
Excessive activation of the mineralocorticoid receptor (MR) underlies the pathophysiology of heart failure and chronic kidney disease. Hyperkalemia risk limits the therapeutic use of conventional MR antagonists. AZD9977 is a nonsteroidal, selective MR modulator that may protect nonepithelial tissues without disturbing electrolyte balance. This phase I study investigated the safety, tolerability, pharmacokinetics, and pharmacodynamics of multiple oral doses of AZD9977 in healthy volunteers. Twenty‐seven male participants aged 23–45 years were randomized 3:1 to receive oral AZD9977 or placebo for 8 days (with twice‐daily dosing on days 2–7), in dose cohorts of 50, 150, and 300 mg (AZD9977, n = 6 per cohort; placebo, n = 3 per cohort). Adverse events occurred in 4 of 18 participants receiving AZD9977 (22.2%) and 6 of 9 receiving placebo (66.7%), all of mild or moderate severity; none were serious or led to withdrawal. AZD9977 was rapidly absorbed, with median time of maximum concentration of 0.50–0.84 hours across dose groups. Area under the curve and maximum concentration were approximately dose proportional but elimination and accumulation terminal half‐life increased with dose. Steady‐state was reached after 3–4 days, with dose‐dependent accumulation of 1.2–1.7‐fold. Renal clearance was 5.9–6.5 L/hour and 24–37% of AZD9977 was excreted in the urine. Serum aldosterone levels increased dose dependently from days −1 to 7 in participants receiving AZD9977, but serum potassium levels and urinary electrolyte excretion were unchanged. AZD9977 was generally well‐tolerated with no safety concerns. Exploratory outcomes suggested reduced hyperkalemia risk compared with MR antagonists. These findings support further clinical development of AZD9977.
Collapse
Affiliation(s)
- Andrew Whittaker
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Åsa M Kragh
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Judith Hartleib-Geschwindner
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Anna Backlund
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter J Greasley
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Heijer
- Clinical Pharmacology Biologics and Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Biometrics and Statistical Innovation, Data Science and AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Pablo Forte
- Early Phase Clinical Unit, PAREXEL, Harrow, UK
| | - Robert Unwin
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Wernevik
- Research and Early Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hans Ericsson
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
49
|
Aldosterone Antagonists Reduce the Risk of Cardiovascular Mortality in Dialysis Patients: A Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1925243. [PMID: 30941188 PMCID: PMC6421009 DOI: 10.1155/2019/1925243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/24/2019] [Indexed: 12/15/2022]
Abstract
Background and Purpose. Cardiovascular disease is the major cause of death in dialysis patients. Although aldosterone antagonists were considered a treatment for severe heart failure patients to reduce cardiac mortality, whether treating patients undergoing maintenance dialysis with aldosterone antagonists could reduce the risk of cardiocerebrovascular (CCV) remains unclear. We aim to systematically assess the efficacy and tolerability of the addition of aldosterone antagonists to conventional therapy in patients undergoing maintenance dialysis. Materials and Methods. We searched PubMed, EMBASE, the Cochrane Library, the Chinese Biomedical Literature Database (CBM), and the China National Knowledge Infrastructure (CNKI) for relevant articles. The primary endpoint of interest was CCV mortality. The secondary endpoints were all-cause mortality, left ventricular mass index (LVMI), and left ventricular ejection fraction (LVEF). Publication bias was evaluated using funnel plots and Egger's test. The meta-analysis was performed using Review Manager software version 5.3. Results. This analysis included 10 randomized controlled trials (RCTs) with 1172 total chronic dialysis patients. The use of aldosterone antagonists in the dialysis population resulted in a marked reduction in CCV mortality (RR 0.42, 95% CI 0.26-0.65, P=0.0002) and all-cause mortality (RR0.46, 95%CI 0.32-0.66, P<0.0001). The LVEF was improved by treatment with aldosterone antagonists (WMD 6.35%, P<0.00001). Moreover, aldosterone antagonists decreased the LVMI (WMD -8.69 g/m2, P=0.0006), whereas aldosterone antagonists increased the occurrence of hyperkalemia (RR1.70, 95%CI 1-2.88, P=0.05) and gynecomastia (RR 8.01, 95% CI 2.44- 26.27, P=0.0006). Conclusions. Addition of aldosterone antagonists to conventional treatment in chronic dialysis patients may reduce CCV mortality, improve cardiac function, and simultaneously decrease LVMI.
Collapse
|
50
|
Bramlage P, Lanzinger S, van Mark G, Hess E, Fahrner S, Heyer CHJ, Friebe M, Seufert J, Danne T, Holl RW. Patient and disease characteristics of type-2 diabetes patients with or without chronic kidney disease: an analysis of the German DPV and DIVE databases. Cardiovasc Diabetol 2019; 18:33. [PMID: 30878037 PMCID: PMC6420726 DOI: 10.1186/s12933-019-0837-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To evaluate the characteristics of type 2 diabetes (T2DM) patients with or without chronic kidney disease (CKD) in Germany. METHODS Using combined DPV/DIVE registry data, the analysis included patients with T2DM at least ≥ 18 years old who had an estimated glomerular filtration rate (eGFR) value available. CKD was defined as an eGFR < 60 mL/min/1.73 m2 or eGFR ≥ 60 mL/min/1.73 m2 and albuminuria (≥ 30 mg/g). Median values of the most recent treatment year per patient are reported. RESULTS Among 343,675 patients with T2DM 171,930 had CKD. Patients with CKD had a median eGFR of 48.9 mL/min/1.73 m2 and 51.2% had a urinary albumin level ≥ 30 mg/g. They were older, had a longer diabetes duration and a higher proportion was females compared to patients without CKD (all p < 0.001). More than half of CKD patients (53.5%) were receiving long-acting insulin-based therapy versus around 39.1% of those without (p < 0.001). CKD patients also had a higher rate of hypertension (79.4% vs 72.0%; p < 0.001). The most common antihypertensive drugs among CKD patients were renin-angiotensin-aldosteron system inhibitors (angiotensin converting enzyme inhibitors 33.8%, angiotensin receptor blockers 14.2%) and diuretics (40.2%). CKD patients had a higher rate of dyslipidemia (88.4% vs 86.3%) with higher triglyceride levels (157.9 vs 151.0 mg/dL) and lower HDL-C levels (men: 40.0 vs 42.0 mg/dL; women: 46.4 vs 50.0 mg/dL) (all p < 0.001) and a higher rate of hyperkalemia (> 5.5 mmol/L: 3.7% vs. 1.0%). Comorbidities were more common among CKD patients (p < 0.001). CONCLUSION The results illustrate the prevalence and morbidity burden associated with diabetic kidney disease in patients with T2DM in Germany. The data call for more attention to the presence of chronic kidney disease in patients with diabetes, should trigger intensified risk factor control up and beyond the control of blood glucose and HbA1c in these patients. They may also serve as a trigger for future investigations into this patient population asking for new treatment options to be developed.
Collapse
Affiliation(s)
- Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Bahnhofstrasse 20, 49661 Cloppenburg, Germany
| | - Stefanie Lanzinger
- Institut für Epidemiologie und medizinische Biometrie, ZIBMT; Universität Ulm, Ulm, Germany
- Deutsches Zentrum für Diabetesforschung e.V, Neuherberg, Munich, Germany
| | - Gesine van Mark
- Institute for Pharmacology and Preventive Medicine, Bahnhofstrasse 20, 49661 Cloppenburg, Germany
| | - Eva Hess
- Diabetologische Schwerpunktpraxis Dres. Hess, Worms, Germany
| | - Simon Fahrner
- Medizinische Klinik, SRH Klinik Sigmaringen, Pfullendorf, Germany
| | | | | | - Jochen Seufert
- Universitätsklinikum Freiburg, Medizinische Fakultät, Freiburg, Germany
| | - Thomas Danne
- Kinderkrankenhaus auf der Bult, Diabeteszentrum für Kinder und Jugendliche, Hannover, Germany
| | - Reinhard W. Holl
- Institut für Epidemiologie und medizinische Biometrie, ZIBMT; Universität Ulm, Ulm, Germany
- Deutsches Zentrum für Diabetesforschung e.V, Neuherberg, Munich, Germany
| |
Collapse
|