51
|
Cice G, Monzo L, Calo L. The uraemic hypertensive patient: a therapeutic challenge—right you are (if you think so). Eur Heart J Suppl 2020; 22:L44-L48. [PMID: 33654466 PMCID: PMC7904065 DOI: 10.1093/eurheartj/suaa133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
High blood pressure (BP) is a leading cause of chronic kidney disease (CKD) and at the same time represents its most frequent complication. High BP is an independent risk factor for advanced CKD; on the other hand, at least 40% of patients with normal glomerular filtration rate (GFR) and virtually all patients with GFR <30 mL/min are hypertensive. CKD and microalbuminuria are powerful risk factors for cardiovascular morbidity and mortality. Consequently, in uraemic hypertension, it is of utmost importance to carefully manage both high BP and microalbuminuria, in order to slow down the progression of kidney damage and to reduce the incidence of cardiovascular events. The first purpose of the medical treatment in hypertensive patients is to normalize BP, regardless of the drug used. Nevertheless, some drugs have an ‘additional’ nephroprotective effect at the same BP target achieved. In this regard, first-line drugs are definitely renin–angiotensin–aldosterone inhibitors, mainly for their proved efficacy in reducing hypertension-related kidney damage and proteinuria. Anyway, a combined approach (two or more drugs) is usually needed to achieve the optimal BP target and reduce the worsening of CKD.
Collapse
Affiliation(s)
- Gennaro Cice
- Department of Cardiology, Policlinico Casilino, Rome, Italy
| | - Luca Monzo
- Department of Cardiology, Policlinico Casilino, Rome, Italy
- Sapienza University, Rome, Italy
| | - Leonardo Calo
- Department of Cardiology, Policlinico Casilino, Rome, Italy
| |
Collapse
|
52
|
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: 6.6] [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
|
53
|
Cosimato C, Agoritsas T, Mavrakanas TA. Mineralocorticoid receptor antagonists in patients with chronic kidney disease. Pharmacol Ther 2020; 219:107701. [PMID: 33027644 DOI: 10.1016/j.pharmthera.2020.107701] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/17/2020] [Indexed: 11/24/2022]
Abstract
Mineralocorticoid receptor antagonists (MRA) can reduce cardiovascular morbidity and mortality in patients with heart failure and ischemic heart disease. In addition, these agents have been used in patients with diabetic nephropathy to control proteinuria and slow down chronic kidney disease (CKD) progression. Current guidelines recommend against the use of MRAs in patients with advanced CKD. However, there is growing interest on their use in this population that has unmet needs (high cardiovascular morbidity and mortality) and unique challenges (risk of acute kidney injury or hyperkalemia). This narrative review discusses the emerging role of MRAs for the management of cardiovascular disease and/or the prevention of CKD progression, highlighting results from randomized controlled trials and presenting real-world data from available registries. Results from recent trials in patients on maintenance dialysis are also discussed.
Collapse
Affiliation(s)
- Cosimo Cosimato
- Division of General Internal Medicine, Department of Medicine, University Hospitals of Geneva & Faculty of Medicine, Geneva, Switzerland
| | - Thomas Agoritsas
- Division of General Internal Medicine, Department of Medicine, University Hospitals of Geneva & Faculty of Medicine, Geneva, Switzerland; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Thomas A Mavrakanas
- Division of General Internal Medicine, Department of Medicine, University Hospitals of Geneva & Faculty of Medicine, Geneva, Switzerland; Division of Nephrology, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
54
|
Minakuchi H, Wakino S, Urai H, Kurokochi A, Hasegawa K, Kanda T, Tokuyama H, Itoh H. The effect of aldosterone and aldosterone blockade on the progression of chronic kidney disease: a randomized placebo-controlled clinical trial. Sci Rep 2020; 10:16626. [PMID: 33024237 PMCID: PMC7538950 DOI: 10.1038/s41598-020-73638-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/10/2020] [Indexed: 01/13/2023] Open
Abstract
The progression of chronic kidney disease (CKD) cannot be completely inhibited. We first explored factors contributing to CKD progression in patients with CKD in a prospective observational study. In the next phase, we focused on the effects of aldosterone, conducting a single-blinded placebo-controlled study using the selective mineralocorticoid receptor antagonist (MRA), eplerenone (25 mg/day). We recruited patients with CKD stage 2 and 3 whose plasma aldosterone concentration was above 15 ng/dL based on the prior data of a prospective observational study. In the CKD cohort study (n = 141), baseline plasma aldosterone concentration was identified as an independent contributory factor for the future rate of change in estimated glomerular filtration rate (eGFR). When the cut-off value for aldosterone was set at 14.5 ng/dL, the decline rate was significantly higher in patients with higher plasma aldosterone concentration (− 1.22 ± 0.39 ml/min/1.73 m2/year vs. 0.39 ± 0.40 ml/min/1.73 m2/year, p = 0.0047). In the final intervention study, in the eplerenone group, eGFR dropped at 6 months after the initiation of the study, and thereafter eGFR was maintained until the end of the study. At 24 months and 36 months, eGFR was significantly higher in the eplerenone group than in the placebo group. In conclusion, MRA can be an effective strategy in preventing CKD progression, especially in patients with high plasma aldosterone.
Collapse
Affiliation(s)
- Hitoshi Minakuchi
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shu Wakino
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Hidenori Urai
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Arata Kurokochi
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuhiro Hasegawa
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Kanda
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hirobumi Tokuyama
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
55
|
Shantsila E, Shahid F, Sun Y, Deeks J, Calvert M, Fisher JP, Kirchhof P, Gill PS, Lip GYH. Spironolactone in Atrial Fibrillation With Preserved Cardiac Fraction: The IMPRESS-AF Trial. J Am Heart Assoc 2020; 9:e016239. [PMID: 32909497 PMCID: PMC7726985 DOI: 10.1161/jaha.119.016239] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Patients with permanent atrial fibrillation have poor outcomes, exercise capacity, and quality of life even on optimal anticoagulation. Based on mechanistic and observational data, we tested whether the mineralocorticoid receptor antagonist spironolactone can improve exercise capacity, E/e' ratio, and quality of life in patients with permanent atrial fibrillation and preserved ejection fraction. Methods and Results The double-masked, placebo-controlled IMPRESS-AF (Improved Exercise Tolerance in Heart Failure With Preserved Ejection Fraction by Spironolactone on Myocardial Fibrosis in Atrial Fibrillation) trial (NCT02673463) randomized 250 stable patients with permanent atrial fibrillation and preserved left ventricular ejection fraction to spironolactone 25 mg daily or placebo. Patients were followed for 2 years. The primary efficacy outcome was peak oxygen consumption on cardiopulmonary exercise testing at 2 years. Secondary end points included 6-minute walk distance, E/e' ratio, quality of life, and hospital admissions. Spironolactone therapy did not improve peak oxygen consumption at 2 years (14.0 mL/min per kg [SD, 5.4]) compared with placebo (14.5 [5.1], adjusted treatment effect, -0.28; 95% CI, -1.27 to 0.71]; P=0.58). The findings were consistent across all sensitivity analyses. There were no differences in the 6-minute walking distance (adjusted treatment effect, -8.47 m; -31.9 to 14.9; P=0.48), E/e' ratio (adjusted treatment effect, -0.68; -1.52 to 0.17, P=0.12), or quality of life (P=0.74 for EuroQol-5 Dimensions, 5-level version quality of life questionnaire and P=0.84 for Minnesota Living with Heart Failure). At least 1 hospitalization occurred in 15% of patients in the spironolactone group and 23% in the placebo group (P=0.15). Estimated glomerular filtration rate was reduced by 6 mL/min in the spironolactone group with <1-unit reduction in controls (P<0.001). Systolic blood pressure was reduced by 7.2 mm Hg (95% CI, 2.2-12.3) in the spironolactone group versus placebo (P=0.005). Conclusions Spironolactone therapy does not improve exercise capacity, E/e' ratio, or quality of life in patients with chronic atrial fibrillation and preserved ejection fraction. Registration UTL: https://www.clinicaltrial.gov; Unique identifier: NCT02673463. EudraCT number 2014-003702-33.
Collapse
Affiliation(s)
- Eduard Shantsila
- North Worcestershire VTSSt Helens and Knowsley Teaching Hospitals NHS TrustPrescotMerseysideUnited Kingdom
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Farhan Shahid
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
| | - Yongzhong Sun
- Birmingham Clinical Trials UnitInstitute of Applied Health ResearchUniversity of BirminghamUnited Kingdom
| | - Jonathan Deeks
- Birmingham Clinical Trials UnitInstitute of Applied Health ResearchUniversity of BirminghamUnited Kingdom
- NIHR Birmingham Biomedical Research CentreUniversity of BirminghamUnited Kingdom
| | - Melanie Calvert
- NIHR Birmingham Biomedical Research CentreUniversity of BirminghamUnited Kingdom
- Centre for Patient Reported Outcomes Research (CPROR)Institute of Applied Health ResearchUniversity of BirminghamUnited Kingdom
- National Institute for Health Research (NIHR) Applied Research Centre West MidlandsUniversity of BirminghamUnited Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research CentreUniversity of BirminghamUnited Kingdom
- Birmingham Health Partners Centre for Regulatory Science and InnovationUniversity of BirminghamUnited Kingdom
| | - James P. Fisher
- Department of PhysiologyFaculty of Medical and Health SciencesUniversity of AucklandNew Zealand
| | - Paulus Kirchhof
- Institute of Cardiovascular SciencesUniversity of BirminghamUnited Kingdom
- University Hospitals Birmingham NHS Foundation TrustBirminghamUnited Kingdom
- Sandwell and West Birmingham Hospitals NHS TrustBirminghamUnited Kingdom
- University Heart and Vascular CenterUKE HamburgHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LübeckHamburgGermany
| | - Paramjit S. Gill
- Academic Unit of Primary CareWarwick Medical SchoolUniversity of WarwickCoventryUnited Kingdom
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular ScienceUniversity of Liverpool and Liverpool Heart & Chest HospitalLiverpoolUnited Kingdom
- Aalborg Thrombosis Research UnitDepartment of Clinical MedicineAalborg UniversityAalborgDenmark
| |
Collapse
|
56
|
Rico-Mesa JS, White A, Ahmadian-Tehrani A, Anderson AS. Mineralocorticoid Receptor Antagonists: a Comprehensive Review of Finerenone. Curr Cardiol Rep 2020; 22:140. [PMID: 32910349 DOI: 10.1007/s11886-020-01399-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW We aim to review the mechanism of action and safety profile of mineralocorticoid receptor antagonists (MRAs) and discuss the differences between selective and non-selective MRAs. More specifically, finerenone is a new medication that is currently under investigation for its promising cardiovascular and nephrological effects. RECENT FINDINGS MRAs are well known for their utility in treating heart failure, refractory hypertension, and diverse nephropathies, namely, diabetic nephropathy. As their name denotes, MRAs inhibit the action of aldosterone at the mineralocorticoid receptor, preventing receptor activation. This prevents remodeling, decreases inflammation, and improves proteinuria. There are not significant differences in outcomes between selective and non-selective MRAs. A new selective MRA named finerenone (originally BAY 94-8862) has shown promising results in several trials (ARTS-HF and ARTS-DN) and smaller studies. Finerenone may have a dose-dependent benefit over older MRAs, decreasing rates of albuminuria and levels of BNP and NT-ProBNP without causing a significant increase in serum potassium levels. This medication is not yet approved as it is still in phase 3 clinical trials (FIGARO-DKD and FIDELIO-DKD trials). MRAs are beneficial in several disease states. Newer medications, such as finerenone, should be considered in patients with heart failure and diabetic nephropathy who may benefit from a reduction in albuminuria and BNP/NT-ProBNP. Data surrounding finerenone are limited to date. However, results from ongoing clinical trials, as well as new trials to evaluate use in other pathologies, could validate the implementation of this medication in daily practice.
Collapse
Affiliation(s)
- Juan Simon Rico-Mesa
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Averi White
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Ashkan Ahmadian-Tehrani
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Allen S Anderson
- Department of Medicine, Division of Cardiovascular Diseases, University of Texas Health San Antonio, 7703 Floyd Curl Drive, MC 7872, San Antonio, TX, 78229, USA.
| |
Collapse
|
57
|
Effects of mineralocorticoid receptor antagonists in proteinuric kidney disease: a systematic review and meta-analysis of randomized controlled trials. J Hypertens 2020; 37:2307-2324. [PMID: 31688290 DOI: 10.1097/hjh.0000000000002187] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Reductions in albuminuria of more than 30% are considered a strong marker of delay of chronic kidney disease (CKD) progression. Single renin-angiotensin system (RAS) blockade represents the cornerstone of CKD treatment. However, as CKD progression still occurs, other nephroprotective options were explored; mineralocorticoid receptor antagonists (MRA) were tested with generally positive results. METHODS We conducted a systematic review and meta-analysis on the effects of MRAs on albuminuria/proteinuria, and adverse events, such as change in renal function and hyperkalemia incidence. A detailed search in electronic databases, clinical trial registries and grey literature was performed to retrieve randomized controlled trials (RCTs) in which administration of an MRA alone or on-top of ACEi/ARB was compared with placebo or active treatment. RESULTS Of the 45 initially identified reports, 31, with 2767 participants, were included in analysis of the primary outcome. The use of MRAs (alone or on top of RAS blockade) compared with placebo decreased urine albumin-to-creatinine ratio (UACR) by -24.55% (95% CI -29.57 to -19.53%), urine protein-to-creatinine ratio (UPCR) by -53.93% (95% CI -79% to -28.86%) and 24 h albumin excretion by -32.47% (95% CI -41.1 to -23.85%). MRAs also reduced UACR by -22.48% (95% CI -24.51 to -20.44%) compared with calcium-channel-blockers (CCBs), whereas no differences were found compared with a second ACEi/ARB or nonpotassium-sparing diuretics. Addition of an MRA was associated with change in estimated glomerular filtration rate (eGFR) of -2.38 ml/min per 1.73 m (95% CI -3.51 to -1.25), rise in potassium by 0.22 mEq/l (95% CI 0.16-0.28 mEq/l) and a 2.6-fold increase in hyperkalemia risk (RR 2.63, 95% CI 1.69-4.08) compared with placebo/active control. CONCLUSION Use of MRAs alone or on top of RAS blockade confers important antiproteinuric effects in patients with CKD, with a slight increase in mean potassium levels.
Collapse
|
58
|
Morimoto S, Ichihara A. Management of primary aldosteronism and mineralocorticoid receptor-associated hypertension. Hypertens Res 2020; 43:744-753. [PMID: 32424201 DOI: 10.1038/s41440-020-0468-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/11/2020] [Accepted: 04/11/2020] [Indexed: 12/31/2022]
Abstract
Resistant hypertension is associated with a poor prognosis due to organ damage caused by prolonged suboptimal blood pressure control. The concomitant use of mineralocorticoid receptor (MR) antagonists with other antihypertensives has been shown to improve blood pressure control in some patients with resistant hypertension, and such patients are considered to have MR-associated hypertension. MR-associated hypertension is classified into two subtypes: one with a high plasma aldosterone level, which includes primary aldosteronism (PA), and the other with a normal aldosterone level. In patients with unilateral PA, adrenalectomy may be the first-choice procedure, while in patients with bilateral PA, MR antagonists are selected. In addition, in patients with other types of MR-associated hypertension with high aldosterone levels, MR antagonists may be selected as a first-line therapy. In patients with normal aldosterone levels, ARBs or ACE inhibitors are used as a first-line therapy, and MR antagonists may be used as an add-on agent. Since MR antagonist therapy may have efficacy as a first-line or add-on agent in these patients, it is important to recognize this type of hypertension. Further studies are needed to elucidate the pathogenesis and management of MR-associated hypertension in more detail to improve the clinical outcomes of patients with MR-associated hypertension.
Collapse
Affiliation(s)
- Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan.
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
59
|
Spencer S, Wheeler‐Jones C, Elliott J. Aldosterone and the mineralocorticoid receptor in renal injury: A potential therapeutic target in feline chronic kidney disease. J Vet Pharmacol Ther 2020; 43:243-267. [PMID: 32128854 PMCID: PMC8614124 DOI: 10.1111/jvp.12848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/20/2020] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
There is a growing body of experimental and clinical evidence supporting mineralocorticoid receptor (MR) activation as a powerful mediator of renal damage in laboratory animals and humans. Multiple pathophysiological mechanisms are proposed, with the strongest evidence supporting aldosterone-induced vasculopathy, exacerbation of oxidative stress and inflammation, and increased growth factor signalling promoting fibroblast proliferation and deranged extracellular matrix homeostasis. Further involvement of the MR is supported by extensive animal model experiments where MR antagonists (such as spironolactone and eplerenone) abrogate renal injury, including ischaemia-induced damage. Additionally, clinical trials have shown MR antagonists to be beneficial in human chronic kidney disease (CKD) in terms of reducing proteinuria and cardiovascular events, though current studies have not evaluated primary end points which allow conclusions to made about whether MR antagonists reduce mortality or slow CKD progression. Although differences between human and feline CKD exist, feline CKD shares many characteristics with human disease including tubulointerstitial fibrosis. This review evaluates the evidence for the role of the MR in renal injury and summarizes the literature concerning aldosterone in feline CKD. MR antagonists may represent a promising therapeutic strategy in feline CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| | | | - Jonathan Elliott
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| |
Collapse
|
60
|
Galceran I, Vázquez S, Durán X, Outón S, Pascual J, Oliveras A. Renal safety outcomes of spironolactone in patients with resistant hypertension. Nefrologia 2020; 40:414-420. [PMID: 31898989 DOI: 10.1016/j.nefro.2019.10.004] [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: 06/05/2019] [Revised: 09/18/2019] [Accepted: 10/20/2019] [Indexed: 10/25/2022] Open
Abstract
INTRODUCTION Resistant hypertension (RH) is a significant health problem with complex management. The aim of this study was to evaluate the risks and benefits of adding spironolactone to treat RH. MATERIAL AND METHODS In total, 216 patients with RH in whom spironolactone (12.5-25mg daily) was added as an antihypertensive were evaluated. One-hundred and twenty-five (125) were analysed retrospectively and 91 prospectively. Blood pressure (BP) and laboratory parameters (serum creatinine [sCrea], estimated glomerular filtration rate [eGFR] and serum potassium [sK]) were analysed at baseline and at 3-6-12 months after introducing spironolactone. RESULTS A change of systolic/diastolic BP (mean±standard deviation) of -10.9±2.7/-4.3±1.6mmHg at 3 months and -13.6±2.8/-6.0±1.6mmHg at 12 months; p<0.001 was observed. These values were confirmed with ambulatory-BP monitoring at 12 months. At 3 months, an increase in sCrea of 0.10±0.04mg/dl, a decrease in eGFR of -5.4±1.9ml/min/1.73m2 and an increase in sK of 0.3±0.1mmol/l; p<0.001 was observed for all cases. These changes were maintained after 12 months. There were no significant differences in changes of BP, sCrea, eGFR and sK between 3 and 12 months. Results of the retrospective and prospective cohorts separately were superimposable. In the prospective cohort, spironolactone was withdrawn in 9 patients (9.9%) because of adverse effects. CONCLUSIONS After 3 months with spironolactone, a decrease in BP associated with a decrease in the eGFR and an increase in sCrea and sK was observed. These changes were maintained at 12 months. Spironolactone is an effective and safe treatment for RH in patients with baseline eGFR ≥30ml/min/1.73m2.
Collapse
Affiliation(s)
- Isabel Galceran
- Servicio de Nefrología, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España; Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España.
| | - Susana Vázquez
- Servicio de Nefrología, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España; Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España
| | - Xavier Durán
- Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España
| | - Sara Outón
- Servicio de Nefrología, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España; Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España
| | - Julio Pascual
- Servicio de Nefrología, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España; Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España
| | - Anna Oliveras
- Servicio de Nefrología, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España; Institut Hospital del Mar d' Investigacions Mèdiques, Universitat Autònoma de Barcelona, Barcelona, España
| |
Collapse
|
61
|
Sarafidis PA, Memmos E, Alexandrou ME, Papagianni A. Mineralocorticoid Receptor Antagonists for Nephroprotection: Current Evidence and Future Perspectives. Curr Pharm Des 2019; 24:5528-5536. [PMID: 30848187 DOI: 10.2174/1381612825666190306162658] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/26/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The use of single RAS-blockade is currently the recommended first-line treatment for proteinuric diabetic or non-diabetic nephropathy, as these agents were repeatedly shown in studies with hard renal outcomes to retard the progression of renal injury. However, CKD will continue to progress on optimum single RAS-blockade, and other options to ameliorate renal injury were explored. Dual RAS-blockade was associated with an increased risk of adverse-events with no apparent benefits and, therefore, is currently abandoned. Based on the phenomenon of aldosterone escape and the well-documented harmful effects of aldosterone on renal tissue, several randomized trials have studied the effects of a MRA in diabetic and non-diabetic nephropathy. METHOD This is a review of the literature in relevance to data evaluating the effect of MRA on renal outcomes. RESULTS Studies with spironolactone and eplerenone added to single RAS-blockade showed that these agents are associated with greater reductions in urine albumin or protein excretion compared to either placebo or dual RASblockade. However, studies with these agents on hard renal outcomes are currently missing and the reasonable skepticism of physicians on the real-world incidence of hyperkalemia in CKD patients are limiting their use. A non-steroidal MRA, finerenone, has also great potency in decreasing albuminuria in diabetic nephropathy with possibly lower rates of hyperkalemia. Two multi-center clinical trials examining the effect of finerenone on hard cardiovascular and renal outcomes are currently ongoing. CONCLUSION MRAs are able to reduce albuminuria and proteinuria on top of single RAS-blockade in patients with proteinuric CKD. Ongoing clinical trials are expected to clarify whether such an effect is accompanied by delay in CKD progression.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelos Memmos
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria-Eleni Alexandrou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aikaterini Papagianni
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
62
|
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: 0.3] [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
|
63
|
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: 1.7] [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
|
64
|
Sato A. Does the temporary decrease in the estimated glomerular filtration rate (eGFR) after initiation of mineralocorticoid receptor (MR) antagonist treatment lead to a long-term renal protective effect? Hypertens Res 2019; 42:1841-1847. [DOI: 10.1038/s41440-019-0320-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/30/2019] [Accepted: 08/02/2019] [Indexed: 01/13/2023]
|
65
|
Verdalles U, Goicoechea M, García de Vinuesa S, Torres E, Hernández A, Verde E, Pérez de José A, Luño J. Chronic kidney disease progression in patients with resistant hypertension subject to 2 therapeutic strategies: Intensification with loop diuretics vs aldosterone antagonists. Nefrologia 2019; 40:65-73. [PMID: 31451203 DOI: 10.1016/j.nefro.2019.04.012] [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: 10/04/2017] [Revised: 04/14/2018] [Accepted: 04/29/2019] [Indexed: 10/26/2022] Open
Abstract
INTRODUCTION Actualy, there are few data about glomerular filtration rate (eGFR) drop in patients with resistant hypertension and how diferent therapies can modify chronic kidney disease progression (CKD). OBJECTIVE To evaluate CKD progression in patients with resistant hypertension undergoing 2diferent therapies: treatment with spironolactone or furosemide. METHODS We included 30 patients (21M, 9W) with a mean age of 66.3±9.1 years, eGFR 55.8±16.5ml/min/1.73 m2, SBP 162.8±8.2 and DBP 90.2±6.2mmHg: 15 patients received spironolactone and 15 furosemide and we followed up them a median of 32 months (28-41). RESULTS The mean annual eGFR decrease was -2.8±5.4ml/min/1.73 m2. In spironolactone group was -2.1±4.8ml/min/1.73 m2 and in furosemide group was -3.2±5.6ml/min/1.73 m2, P<0.01. In patients received spironolactone, SBP decreased 23±9mmHg and in furosemide group decreased 16±3mmHg, P<.01. DBP decreased 10±8mmHg and 6±2mmHg, respectively (P<.01). Treatment with spironolactone reduced albuminuria from a serum albumin/creatine ratio of 210 (121-385) mg/g to 65 (45-120) mg/g at the end of follow-up, P<.01. There were no significant changes in the albumin/creatinine ratio in the furosemide group. The slower drop in kidney function was associated with lower SBP (P=.04), higher GFR (P=.01), lower albuminuria (P=.01), not diabetes mellitus (P=.01) and treatment with spironolactone (P=.02). Treatment with spironolactone (OR 2.13, IC 1.89-2.29) and lower albuminuria (OR 0.98, CI 0.97-0.99) maintain their independent predictive power in a multivariate model. CONCLUSION Treatment with spironolactone is more effective reducing BP and albuminuria in patients with resistant hypertension compared with furosemide and it is associated with a slower progression of CKD in the long term follow up.
Collapse
Affiliation(s)
- U Verdalles
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España.
| | - M Goicoechea
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - S García de Vinuesa
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - E Torres
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - A Hernández
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - E Verde
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - A Pérez de José
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - J Luño
- Departamento Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, España
| |
Collapse
|
66
|
Ito S, Shikata K, Nangaku M, Okuda Y, Sawanobori T. Efficacy and Safety of Esaxerenone (CS-3150) for the Treatment of Type 2 Diabetes with Microalbuminuria: A Randomized, Double-Blind, Placebo-Controlled, Phase II Trial. Clin J Am Soc Nephrol 2019; 14:1161-1172. [PMID: 31248950 PMCID: PMC6682830 DOI: 10.2215/cjn.14751218] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/28/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES The progression of kidney disease in some patients with type 2 diabetes mellitus may not be adequately suppressed by renin-angiotensin system inhibitors. Esaxerenone (CS-3150) is a nonsteroidal mineralocorticoid receptor blocker that has shown kidney protective effects in preclinical studies, and it is a potential add-on therapy to treat diabetic kidney disease. This phase 2 study evaluated the efficacy and safety of esaxerenone in Japanese patients with type 2 diabetes mellitus and microalbuminuria. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This multicenter, randomized, double-blind, placebo-controlled trial enrolled 365 hypertensive or normotensive patients with type 2 diabetes mellitus and microalbuminuria (urinary albumin-to-creatinine ratio ≥45 to <300 mg/g creatinine) treated with renin-angiotensin system inhibitor who had eGFR≥30 ml/min per 1.73 m2. Participants were randomized to receive 0.625, 1.25, 2.5, or 5 mg/d esaxerenone or placebo for 12 weeks. The primary end point was the change in urinary albumin-to-creatinine ratio from baseline to week 12 (with last observation carried forward). RESULTS Esaxerenone treatment at 1.25, 2.5, and 5 mg/d significantly reduced urinary albumin-to-creatinine ratio by the end of treatment (38%, 50%, and 56%, respectively) compared with placebo (7%; all P<0.001). The urinary albumin-to-creatinine ratio remission rate (defined as urinary albumin-to-creatinine ratio <30 mg/g creatinine at the end of treatment and ≥30% decrease from baseline) was 21% in the 2.5- and 5-mg/d groups versus 3% for placebo (both P<0.05). Adverse events occurred slightly more frequently with esaxerenone versus placebo, but the frequencies of drug-related adverse events and discontinuation rates were similar in the placebo and the 0.625-, 1.25-, and 2.5-mg/d groups. Drug-related adverse events and treatment discontinuations were marginally higher in the 5-mg/d group. The most common drug-related adverse event was hyperkalemia, which was dose proportional. CONCLUSIONS Adding esaxerenone at 1.25, 2.5, and 5 mg/d for 12 weeks to an ongoing renin-angiotensin system inhibitor significantly reduces urinary albumin-to-creatinine ratio in patients with type 2 diabetes mellitus and microalbuminuria.
Collapse
Affiliation(s)
- Sadayoshi Ito
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University School of Medicine, Sendai, Japan;
| | - Kenichi Shikata
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; and
| | | | - Tomoko Sawanobori
- Clinical Development Department Daiichi-Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
67
|
Barrera-Chimal J, Girerd S, Jaisser F. Mineralocorticoid receptor antagonists and kidney diseases: pathophysiological basis. Kidney Int 2019; 96:302-319. [PMID: 31133455 DOI: 10.1016/j.kint.2019.02.030] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) represents a global health concern, and its prevalence is increasing. The ultimate therapeutic option for CKD is kidney transplantation. However, the use of drugs that target specific pathways to delay or halt CKD progression, such as angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors is limited in clinical practice. Mineralocorticoid receptor activation in nonclassical tissues, such as the endothelium, smooth muscle cells, inflammatory cells, podocytes, and fibroblasts may have deleterious effects on kidney structure and function. Several preclinical studies have shown that mineralocorticoid receptor antagonists (MRAs) ameliorate or cure kidney injury and dysfunction in different models of kidney disease. In this review, we present the preclinical evidence showing a benefit of MRAs in acute kidney injury, the transition from acute kidney injury to CKD, hypertensive and diabetic nephropathy, glomerulonephritis, and kidney toxicity induced by calcineurin inhibitors. We also discuss the molecular mechanisms responsible for renoprotection related to MRAs that lead to reduced oxidative stress, inflammation, fibrosis, and hemodynamic alterations. The available clinical data support a benefit of MRA in reducing proteinuria in diabetic kidney disease and improving cardiovascular outcomes in CKD patients. Moreover, a benefit of MRAs in kidney transplantation has also been observed. The past and present clinical trials describing the effect of MRAs on kidney injury are presented, and the risk of hyperkalemia and use of other options, such as potassium binding agents or nonsteroidal MRAs, are also addressed. Altogether, the available preclinical and clinical data support a benefit of using MRAs in CKD, an approach that should be further explored in future clinical trials.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Sophie Girerd
- Transplant Unit, Nephrology Department, Nancy University Hospital, Lorraine University, Vandoeuvre-lès-Nancy, France; Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France
| | - Frederic Jaisser
- Institut national de la santé et de la recherche médicale U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, French-Clinical Research Infrastructure Network, Nancy, France; Institut national de la santé et de la recherche médicale, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris Descartes University, Paris, France.
| |
Collapse
|
68
|
Keppel MH, Piecha G, März W, Cadamuro J, Auer S, Felder TK, Mrazek C, Oberkofler H, Trummer C, Grübler MR, Schwetz V, Verheyen N, Pandis M, Borzan V, Haschke-Becher E, Tomaschitz A, Pilz S. The endogenous cardiotonic steroid Marinobufagenin and decline in estimated glomerular filtration rate at follow-up in patients with arterial hypertension. PLoS One 2019; 14:e0212973. [PMID: 30817774 PMCID: PMC6394930 DOI: 10.1371/journal.pone.0212973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 02/12/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Marinobufagenin (MBG) is an endogenous cardiotonic steroid (CTS) that inhibits the Na+/K+-ATPase. Human MBG is significantly increased in end-stage renal disease and immunization against MBG attenuates cardiovascular fibrosis in a rat model of uremic cardiomyopathy. Mineralocorticoid antagonists (MRA) block MBG binding sites and decrease proteinuria in chronic kidney disease (CKD) patients. We therefore aimed to investigate the association of MBG and albuminuria, as a marker of renal damage, as well as MBG and decline of glomerular filtration rate (GFR). METHODS The Graz endocrine causes of hypertension (GECOH) study is a single center study of adults routinely referred for screening of endocrine hypertension. Plasma MBG was measured by an enzyme-linked immunoassay, and in a post-hoc analysis, follow-up creatinine levels were obtained. Patients with proteinuria >3.5g/day at baseline were excluded from further evaluation. RESULTS We measured MBG concentrations in 40 hypertensive subjects and excluded one patient due to pre-existing proteinuria. Plasma MBG was significantly correlated with albuminuria (Spearman ρ = .357; p = .028) and proteinuria (ρ = .336; p = .039). In linear regression analysis, the association remained significant after adjustment for age, sex, and BMI (β = .306; p = .036), and for mean systolic blood pressure (β = .352; p = .034). In follow-up analyses (N = 30), MBG was significantly associated with decline in GFR after adjustment for time-to-follow-up (β = -.374; p = .042). CONCLUSION The findings suggest that MBG plasma concentrations were associated with albuminuria as well as decline in kidney function. Whether MBG predicts hard renal endpoints warrants further investigations.
Collapse
Affiliation(s)
- Martin H. Keppel
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Grzegorz Piecha
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Winfried März
- Synlab Academy, Mannheim, Germany
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics Medical, University of Graz, Graz, Austria
| | - Janne Cadamuro
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Simon Auer
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Thomas K. Felder
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Cornelia Mrazek
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Hannes Oberkofler
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | - Christian Trummer
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Martin R. Grübler
- Department of Cardiology, Swiss Cardiovascular Center Bern, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Verena Schwetz
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Nicolas Verheyen
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Marlene Pandis
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Valentin Borzan
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Elisabeth Haschke-Becher
- University Institute for Medical and Chemical Laboratory Diagnostics, Paracelsus Medical University, Salzburg, Austria
| | | | - Stefan Pilz
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| |
Collapse
|
69
|
Abstract
PURPOSE OF REVIEW Type 2 diabetes (T2D) is associated with an increased risk of diabetic kidney disease (DKD), cardiovascular disease, and heart failure, in part through activation of the renin-angiotensin-aldosterone system (RAAS). Although recent cardiovascular outcome trials have identified newer therapeutic agents such as sodium-glucose cotransporter-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1)-receptor agonists that reduce the risk of these complications, patients still exhibit residual cardiorenal morbidity and mortality. Accordingly, the identification of pharmacological agents that attenuate micro- and macrovascular complications related to T2D is a major priority. Our aim was to review evidence for the role of novel mineralocorticoid receptor antagonists (MRAs) that are being developed as adjunctive therapies to reduce the risk of DKD and cardiovascular disease in the setting of T2D. RECENT FINDINGS Dual RAAS blockade with angiotensin-converting enzyme (ACE) inhibitor plus angiotensin receptor blockade (ARB) or ARB plus renin inhibition increases serious adverse events such as acute kidney injury and stroke. Due to the potential for these serious side effects, more recent interest has focused on newer, more selective non-steroidal MRAs such as finerenone as cardiorenal protective therapies. Finerenone reduces albuminuria in the setting of DKD in patients with T2D and has a lower risk of hyperkalemia compared to currently available MRAs. Novel MRAs such as finerenone have the potential to reduce the risk of DKD progression in patients with T2D. The impact of finerenone on hard, long-term cardiorenal endpoints is being examined in the FIGARO and FIDELIO trials in patients with DKD.
Collapse
Affiliation(s)
- Yuliya Lytvyn
- Toronto General Hospital Research Institute, UHN, 585 University Ave, 8N-845, Toronto, Ontario, M5G 2N2, Canada.
| | - Lucas C Godoy
- Peter Munk Cardiac Centre, University of Toronto, Toronto, Canada
- Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Rosalie A Scholtes
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, The Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, The Netherlands
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Canada
- Banting and Best Diabetes Centre, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
70
|
Shatat IF, Becton LJ, Woroniecki RP. Hypertension in Childhood Nephrotic Syndrome. Front Pediatr 2019; 7:287. [PMID: 31380323 PMCID: PMC6646680 DOI: 10.3389/fped.2019.00287] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/26/2019] [Indexed: 11/20/2022] Open
Abstract
Arterial hypertension (HTN) is commonly encountered by clinicians treating children with steroid sensitive (SSNS) and steroid resistant nephrotic syndrome (SRNS). Although the prevalence of HTN in SSNS is less documented than in SRNS, recent studies reported high prevalence in both. Studies have estimated the prevalence of HTN in different patient populations with NS to range from 8 to 59.1%. Ambulatory HTN, abnormalities in BP circadian rhythm, and measures of BP variability are prevalent in patients with NS. Multiple mechanisms and co-morbidities contribute to the pathophysiology of HTN in children with NS. Some contributing factors are known to cause acute and episodic elevations in blood pressure such as fluid shifts, sodium retention, and medication side effects (steroids, CNIs). Others are associated with chronic and more sustained HTN such as renal fibrosis, decreased GFR, and progression of chronic kidney disease. Children with NS are more likely to suffer from other cardiovascular disease risk factors, such as obesity, increased measures of arterial stiffness [increased carotid intima-media thickness (cIMT), endothelial dysfunction, increased pulse wave velocity (PWV)], impaired glucose metabolism, dyslipidemia, left ventricular hypertrophy (LVH), left ventricular dysfunction, and atherosclerosis. Those risk factors have been associated with premature death in adults. In this review on HTN in patients with NS, we will discuss the epidemiology and pathophysiology of hypertension in patients with NS, as well as management aspects of HTN in children with NS.
Collapse
Affiliation(s)
- Ibrahim F Shatat
- Pediatric Nephrology and Hypertension, Sidra Medicine, Doha, Qatar.,Department of Pediatrics, Weill Cornell College of Medicine-Qatar, Doha, Qatar.,College of Nursing, Medical University of South Carolina, Charleston, SC, United States
| | - Lauren J Becton
- Private Practice Practitioner, Pediatric Nephrology, Seattle, WA, United States
| | - Robert P Woroniecki
- Pediatric Nephrology and Hypertension, Stony Brook Children's Hospital, Stony Brook, NY, United States
| |
Collapse
|
71
|
Abstract
Aldosterone is a mineralocorticoid hormone, as its main renal effect has been considered as electrolyte and water homeostasis in the distal tubule, thus maintaining blood pressure and extracellular fluid homeostasis through the activation of mineralocorticoid receptor (MR) in epithelial cells. However, over the past decade, numerous studies have documented the significant role of aldosterone in the progression of chronic kidney disease (CKD) which has become a subject of interest. It is being studied that aldosterone can affect cardiovascular and renal system, thereby contributing to tissue inflammation, injury, glomerulosclerosis, and interstitial fibrosis. Aldosterone acts on renal vessels, renal cells (glomerular mesangial cells, podocytes, vascular smooth muscle cells, tubular epithelial cells, and interstitial fibroblasts), and infiltrating inflammatory cells, inducing reactive oxygen species (ROS) production, upregulated epithelial growth factor receptor (EGFR), and type 1 angiotensin (AT1) receptor expressions, and activating nuclear factor kappa B (NF-κB), activator protein-1 (AP-1), and EGFR to further promote cell proliferation, apoptosis, and proliferation. Phenotypic transformation of epithelial cells stimulates the expression of transforming growth factor-β (TGF-β), connective tissue growth factor (CTGF), osteopontin (OPN), and plasminogen activator inhibitor-1 (PAI-1), eventually leading to renal fibrosis. MR antagonisms are related to inhibition of aldosterone-mediated pro-inflammatory and pro-fibrotic effect. In this review, we will summarize the important role of aldosterone in the pathogenesis of renal injury and fibrosis, emphasizing on its multiple underlying mechanisms and advances in aldosterone research along with the potential therapeutics for targeting MR in a renal fibrosis.
Collapse
|
72
|
Granberg KL, Yuan ZQ, Lindmark B, Edman K, Kajanus J, Hogner A, Malmgren M, O’Mahony G, Nordqvist A, Lindberg J, Tångefjord S, Kossenjans M, Löfberg C, Brånalt J, Liu D, Selmi N, Nikitidis G, Nordberg P, Hayen A, Aagaard A, Hansson E, Hermansson M, Ivarsson I, Jansson-Löfmark R, Karlsson U, Johansson U, William-Olsson L, Hartleib-Geschwindner J, Bamberg K. Identification of Mineralocorticoid Receptor Modulators with Low Impact on Electrolyte Homeostasis but Maintained Organ Protection. J Med Chem 2018; 62:1385-1406. [DOI: 10.1021/acs.jmedchem.8b01523] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Dongmei Liu
- Pharmaron Beijing Co., Ltd., No. 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Lachaux M, Barrera-Chimal J, Nicol L, Rémy-Jouet I, Renet S, Dumesnil A, Wecker D, Richard V, Kolkhof P, Jaisser F, Ouvrard-Pascaud A, Mulder P. Short- and long-term administration of the non-steroidal mineralocorticoid receptor antagonist finerenone opposes metabolic syndrome-related cardio-renal dysfunction. Diabetes Obes Metab 2018; 20:2399-2407. [PMID: 29862614 DOI: 10.1111/dom.13393] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 12/22/2022]
Abstract
AIM To determine whether non-steroidal mineralocorticoid receptor (MR) antagonists oppose metabolic syndrome-related end-organ, i.e. cardiac, damage. MATERIALS AND METHODS In Zucker fa/fa rats, a rat model of metabolic syndrome, we assessed the effects of the non-steroidal MR antagonist finerenone (oral 2 mg/kg/day) on left ventricular (LV) function, haemodynamics and remodelling (using echocardiography, magnetic resonance imaging and biochemical methods). RESULTS Long-term (90 days) finerenone modified neither systolic blood pressure nor heart rate, but reduced LV end-diastolic pressure and LV end-diastolic pressure-volume relationship, without modifying LV end-systolic pressure and LV end-systolic pressure-volume relationship. Simultaneously, long-term finerenone reduced both LV systolic and diastolic diameters, associated with reductions in LV weight and LV collagen density, while proteinuria and renal nGAL expression were reduced. Short-term (7 days) finerenone improved LV haemodynamics and reduced LV systolic diameter, without modifying LV diastolic diameter. Moreover, short-term finerenone increased myocardial tissue perfusion and reduced myocardial reactive oxygen species, while plasma nitrite levels, an indicator of nitric oxide (NO) bio-availability, were increased. CONCLUSIONS In rats with metabolic syndrome, the non-steroidal MR antagonist finerenone opposed metabolic syndrome-related diastolic cardiac dysfunction and nephropathy. This involved acute effects, such as improved myocardial perfusion, reduced oxidative stress/increased NO bioavailability, as well as long-term effects, such as modifications in the myocardial structure.
Collapse
Affiliation(s)
- Marianne Lachaux
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Lionel Nicol
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Isabelle Rémy-Jouet
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Sylvanie Renet
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Anais Dumesnil
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Vincent Richard
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | | | - Frederic Jaisser
- Institut National de la Santé et de la Recherche Médicale U1138, Paris, France
| | - Antoine Ouvrard-Pascaud
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, FHU- REMOD-VHF, 76000 Rouen, France
| |
Collapse
|
74
|
Sun WY, Bai B, Luo C, Yang K, Li D, Wu D, Félétou M, Villeneuve N, Zhou Y, Yang J, Xu A, Vanhoutte PM, Wang Y. Lipocalin-2 derived from adipose tissue mediates aldosterone-induced renal injury. JCI Insight 2018; 3:120196. [PMID: 30185654 DOI: 10.1172/jci.insight.120196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Lipocalin-2 is not only a sensitive biomarker, but it also contributes to the pathogenesis of renal injuries. The present study demonstrates that adipose tissue-derived lipocalin-2 plays a critical role in causing both chronic and acute renal injuries. Four-week treatment with aldosterone and high salt after uninephrectomy (ANS) significantly increased both circulating and urinary lipocalin-2, and it induced glomerular and tubular injuries in kidneys of WT mice. Despite increased renal expression of lcn2 and urinary excretion of lipocalin-2, mice with selective deletion of lcn2 alleles in adipose tissue (Adipo-LKO) are protected from ANS- or aldosterone-induced renal injuries. By contrast, selective deletion of lcn2 alleles in kidney did not prevent aldosterone- or ANS-induced renal injuries. Transplantation of fat pads from WT donors increased the sensitivity of mice with complete deletion of Lcn2 alleles (LKO) to aldosterone-induced renal injuries. Aldosterone promoted the urinary excretion of a human lipocalin-2 variant, R81E, in turn causing renal injuries in LKO mice. Chronic treatment with R81E triggered significant renal injuries in LKO, resembling those observed in WT mice following ANS challenge. Taken in conjunction, the present results demonstrate that lipocalin-2 derived from adipose tissue causes acute and chronic renal injuries, largely independent of local lcn2 expression in kidney.
Collapse
Affiliation(s)
- Wai Yan Sun
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Bo Bai
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Cuiting Luo
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Kangmin Yang
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Dahui Li
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | | | | | - Yang Zhou
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Paul M Vanhoutte
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology and.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
75
|
Agarwal R, Rossignol P, Garza D, Mayo MR, Warren S, Arthur S, Romero A, White WB, Williams B. Patiromer to Enable Spironolactone Use in the Treatment of Patients with Resistant Hypertension and Chronic Kidney Disease: Rationale and Design of the AMBER Study. Am J Nephrol 2018; 48:172-180. [PMID: 30176673 DOI: 10.1159/000492622] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/01/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND While chronic kidney disease (CKD) is common in resistant hypertension (RHTN), prior studies -evaluating mineralocorticoid receptor antagonists excluded patients with reduced kidney function due to risk of hyperkalemia. AMBER (ClinicalTrials.gov identifier NCT03071263) will evaluate if the potassium-binding polymer patiromer used concomitantly with spironolactone in patients with RHTN and CKD prevents hyperkalemia and allows more persistent spironolactone use for hypertension management. METHODS Randomized, double-blind, placebo-controlled parallel group 12-week study of patiromer and spironolactone versus placebo and spironolactone in patients with uncontrolled RHTN and CKD. RHTN is defined as unattended systolic automated office blood pressure (AOBP) of -135-160 mm Hg during screening despite taking ≥3 antihypertensives, including a diuretic, and an angiotensin-converting enzyme inhibitor or an angiotensin receptor blocker -(unless not tolerated or contraindicated). The CKD inclusion criterion is an estimated glomerular filtration rate (eGFR) of 25 to ≤45 mL/min/1.73 m2. Screening serum potassium must be 4.3-5.1 mEq/L. The primary efficacy endpoint is the between-group difference (spironolactone plus patiromer versus spironolactone plus placebo) in the proportion of patients remaining on spironolactone at Week 12. RESULTS Baseline characteristics have been analyzed as of March 2018 for 146 (of a targeted 290) patients. Mean (SD) baseline age is 69.3 (10.9) years; 52.1% are male, 99.3% White, and 47.3% have diabetes. Mean (SD) baseline serum potassium is 4.68 (0.25) mEq/L, systolic AOBP is 144.3 (6.8) mm Hg, eGFR is 35.7 (7.7) mL/min/1.73 m2. CONCLUSION AMBER will define the ability of patiromer to facilitate the use of spironolactone, an effective antihypertensive therapy for patients with RHTN and CKD.
Collapse
Affiliation(s)
- Rajiv Agarwal
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Patrick Rossignol
- Inserm 1433 CIC-P CHRU de Nancy, University of Lorraine and FCRIN INI-CRCT, Nancy, France
| | - Dahlia Garza
- Relypsa, Inc., a Vifor Pharma Group Company, Redwood City, California, USA
| | - Martha R Mayo
- Relypsa, Inc., a Vifor Pharma Group Company, Redwood City, California, USA
| | - Suzette Warren
- Relypsa, Inc., a Vifor Pharma Group Company, Redwood City, California, USA
| | - Susan Arthur
- Relypsa, Inc., a Vifor Pharma Group Company, Redwood City, California, USA
| | - Alain Romero
- Relypsa, Inc., a Vifor Pharma Group Company, Redwood City, California, USA
| | - William B White
- Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Bryan Williams
- Institute of Cardiovascular Sciences University College London (UCL) and National Institute for Health Research (NIHR) UCL/UCL Hospitals Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
76
|
Erlandsson F, Albayaty M, Chialda L, Ericsson H, Amilon C, Nelander K, Jansson‐Löfmark R, Wernevik L, Kjaer M, Bamberg K, Hartleib‐Geschwindner J. Clinical safety, tolerability, pharmacokinetics and effects on urinary electrolyte excretion of AZD9977, a novel, selective mineralocorticoid receptor modulator. Br J Clin Pharmacol 2018; 84:1486-1493. [PMID: 29468715 PMCID: PMC6005625 DOI: 10.1111/bcp.13562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 11/24/2017] [Accepted: 01/28/2018] [Indexed: 11/28/2022] Open
Abstract
AIMS AZD9977 is the first mineralocorticoid receptor modulator in clinical development exerting similar organ protection as eplerenone with minimal urinary electrolyte effects in preclinical studies. The aim was to perform the initial clinical assessment of AZD9977. METHODS A first-in-human trial explored doses from 5 to 1200 mg. To study effects on urinary electrolyte excretion an additional randomized placebo controlled cross-over four-period clinical trial was performed. Twenty-three healthy volunteers were administered fludrocortisone alone or in combination with AZD9977, eplerenone or both. AZD9977/eplerenone combination was given to assess if AZD9977 can attenuate eplerenone induced natriuresis. RESULTS AZD9977 at doses from 5 to 1200 mg was safe and well tolerated and pharmacokinetics were compatible with further development. AZD9977 exhibited similar effects on urinary ln [Na+ ]/[K+ ] as eplerenone when using fludrocortisone as mineralocorticoid receptor agonist, and the combination had an additive effect on ln [Na+ K+ ]. CONCLUSIONS The results in man contradict the results in rodent models driven by aldosterone, in which AZD9977 has minimal electrolyte effects. Future clinical studies with AZD9977 should be performed in presence of endogenous or exogenous aldosterone to assess potential benefit of AZD9977 in patients.
Collapse
Affiliation(s)
- Fredrik Erlandsson
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Muna Albayaty
- PAREXEL Early Phase Clinical Unit, Northwick Park HospitalWatford RoadHarrowHA1 3UJUK
| | - Ligia Chialda
- PAREXEL Early Phase Clinical Unit, Northwick Park HospitalWatford RoadHarrowHA1 3UJUK
| | - Hans Ericsson
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Carl Amilon
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Karin Nelander
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Rasmus Jansson‐Löfmark
- Cardiovascular and Metabolic Disease Innovative Medicine Unit, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1Mölndal431 83Sweden
| | - Linda Wernevik
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Magnus Kjaer
- Early Clinical Development, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1, Mölndal 431 83Sweden
| | - Krister Bamberg
- Cardiovascular and Metabolic Disease Innovative Medicine Unit, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1Mölndal431 83Sweden
| | - Judith Hartleib‐Geschwindner
- Cardiovascular and Metabolic Disease Innovative Medicine Unit, Innovative Medicines and Early Development Biotech UnitAstraZenecaPepparedsleden 1Mölndal431 83Sweden
| |
Collapse
|
77
|
Vanholder R, Van Laecke S, Glorieux G, Verbeke F, Castillo-Rodriguez E, Ortiz A. Deleting Death and Dialysis: Conservative Care of Cardio-Vascular Risk and Kidney Function Loss in Chronic Kidney Disease (CKD). Toxins (Basel) 2018; 10:E237. [PMID: 29895722 PMCID: PMC6024824 DOI: 10.3390/toxins10060237] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
The uremic syndrome, which is the clinical expression of chronic kidney disease (CKD), is a complex amalgam of accelerated aging and organ dysfunctions, whereby cardio-vascular disease plays a capital role. In this narrative review, we offer a summary of the current conservative (medical) treatment options for cardio-vascular and overall morbidity and mortality risk in CKD. Since the progression of CKD is also associated with a higher cardio-vascular risk, we summarize the interventions that may prevent the progression of CKD as well. We pay attention to established therapies, as well as to novel promising options. Approaches that have been considered are not limited to pharmacological approaches but take into account lifestyle measures and diet as well. We took as many randomized controlled hard endpoint outcome trials as possible into account, although observational studies and post hoc analyses were included where appropriate. We also considered health economic aspects. Based on this information, we constructed comprehensive tables summarizing the available therapeutic options and the number and kind of studies (controlled or not, contradictory outcomes or not) with regard to each approach. Our review underscores the scarcity of well-designed large controlled trials in CKD. Nevertheless, based on the controlled and observational data, a therapeutic algorithm can be developed for this complex and multifactorial condition. It is likely that interventions should be aimed at targeting several modifiable factors simultaneously.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Steven Van Laecke
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Francis Verbeke
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain.
| |
Collapse
|
78
|
Zheng Y, Tang L, Chen X, Cai G, Li W, Ni Z, Shi W, Ding X, Lin H. Resistant and undertreated hypertension in patients with chronic kidney disease: data from the PATRIOTIC survey. Clin Exp Hypertens 2018; 40:784-791. [PMID: 29509105 DOI: 10.1080/10641963.2018.1433193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Hypertension is prevalent in chronic kidney disease (CKD), but the control of hypertension is suboptimal. We reported the prevalence and characteristics of resistant and undertreated hypertension based on a nationwide survey aiming to improve blood pressure (BP) control. METHODS Resistant hypertension (RH) was defined as BP above the target (<140/90 mm Hg) despite the use of 3 antihypertensive drugs or achieving the target BP by using ≥4 antihypertensive drugs. Undertreated hypertension was defined as uncontrolled hypertension (unCH) using ≤2 drugs. We compared the characteristics and antihypertensive treatment among different groups (including RH and unCH using ≤2 drugs). Multivariable logistic regression was used to detect factors associated with unCH using ≤2 drugs and RH. RESULTS 4,435 nondialysis CKD patients with hypertension were analyzed, and 36.9% of participants achieved controlled hypertension (CH) using ≤3 drugs, 11.1% met the criteria for RH, and 52% had unCH despite the use of ≤ 2 antihypertensive drugs. Participants with unCH using ≤ 2 drugs had low usage of renin-angiotensin system blockers (36.8%) and diuretics (5.5%), which was much lower than participants with CH using ≤3 drugs and RH (P< 0.05). After multivariable adjustment, obesity, advanced CKD stages, urinary protein level of ≥1.5 g/24 h, diabetes, and cardiovascular disease were associated with RH in CKD patients (P< 0.05). CONCLUSION Compared with RH, undertreated hypertension contributes more to the unCH in Chinese CKD patients. It is important to ensure adequate antihypertensive treatment, including choosing antihypertensive drugs, that guidelines recommended.
Collapse
Affiliation(s)
- Ying Zheng
- a Department of Nephrology , Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, State Key Laboratory of Kidney Diseases, Chinese PLA Institute of Nephrology , Beijing , China
| | - Li Tang
- a Department of Nephrology , Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, State Key Laboratory of Kidney Diseases, Chinese PLA Institute of Nephrology , Beijing , China
| | - Xiangmei Chen
- a Department of Nephrology , Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, State Key Laboratory of Kidney Diseases, Chinese PLA Institute of Nephrology , Beijing , China
| | - Guangyan Cai
- a Department of Nephrology , Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, State Key Laboratory of Kidney Diseases, Chinese PLA Institute of Nephrology , Beijing , China
| | - Wenge Li
- b Department of Nephrology , China-Japan Friendship Hospital , Beijing 100029 , China
| | - Zhaohui Ni
- c Department of Nephrology , Renji Hospital, School of Medicine, Shanghai Jiaotong University , Shanghai , China
| | - Wei Shi
- d Department of Nephrology , Guangdong Provincial People's Hospital, Guangzhou , Guangdong , China
| | - Xiaoqiang Ding
- e Department of Nephrology , Zhongshan Hospital, Fudan University , Shanghai , China
| | - Hongli Lin
- f Department of Nephrology , First Affiliated Hospital of Dalian Medical University, Dalian , Liaoning , China
| |
Collapse
|
79
|
Bamberg K, Johansson U, Edman K, William-Olsson L, Myhre S, Gunnarsson A, Geschwindner S, Aagaard A, Björnson Granqvist A, Jaisser F, Huang Y, Granberg KL, Jansson-Löfmark R, Hartleib-Geschwindner J. Preclinical pharmacology of AZD9977: A novel mineralocorticoid receptor modulator separating organ protection from effects on electrolyte excretion. PLoS One 2018; 13:e0193380. [PMID: 29474466 PMCID: PMC5825103 DOI: 10.1371/journal.pone.0193380] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 02/11/2018] [Indexed: 11/23/2022] Open
Abstract
Excess mineralocorticoid receptor (MR) activation promotes target organ dysfunction, vascular injury and fibrosis. MR antagonists like eplerenone are used for treating heart failure, but their use is limited due to the compound class-inherent hyperkalemia risk. Here we present evidence that AZD9977, a first-in-class MR modulator shows cardio-renal protection despite a mechanism-based reduced liability to cause hyperkalemia. AZD9977 in vitro potency and binding mode to MR were characterized using reporter gene, binding, cofactor recruitment assays and X-ray crystallopgraphy. Organ protection was studied in uni-nephrectomised db/db mice and uni-nephrectomised rats administered aldosterone and high salt. Acute effects of single compound doses on urinary electrolyte excretion were tested in rats on a low salt diet. AZD9977 and eplerenone showed similar human MR in vitro potencies. Unlike eplerenone, AZD9977 is a partial MR antagonist due to its unique interaction pattern with MR, which results in a distinct recruitment of co-factor peptides when compared to eplerenone. AZD9977 dose dependently reduced albuminuria and improved kidney histopathology similar to eplerenone in db/db uni-nephrectomised mice and uni-nephrectomised rats. In acute testing, AZD9977 did not affect urinary Na+/K+ ratio, while eplerenone increased the Na+/K+ ratio dose dependently. AZD9977 is a selective MR modulator, retaining organ protection without acute effect on urinary electrolyte excretion. This predicts a reduced hyperkalemia risk and AZD9977 therefore has the potential to deliver a safe, efficacious treatment to patients prone to hyperkalemia.
Collapse
MESH Headings
- Administration, Oral
- Aldosterone
- Animals
- Benzoates/chemistry
- Benzoates/pharmacokinetics
- Benzoates/pharmacology
- Cell Line, Tumor
- Dose-Response Relationship, Drug
- Drug Evaluation, Preclinical
- Eplerenone
- Humans
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Male
- Mice, Mutant Strains
- Mineralocorticoid Receptor Antagonists/chemistry
- Mineralocorticoid Receptor Antagonists/pharmacokinetics
- Mineralocorticoid Receptor Antagonists/pharmacology
- Molecular Structure
- Oxazines/chemistry
- Oxazines/pharmacokinetics
- Oxazines/pharmacology
- Potassium/urine
- Rats, Sprague-Dawley
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Sodium/urine
- Sodium, Dietary
- Spironolactone/analogs & derivatives
- Spironolactone/chemistry
- Spironolactone/pharmacokinetics
- Spironolactone/pharmacology
Collapse
Affiliation(s)
- Krister Bamberg
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ulrika Johansson
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Karl Edman
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lena William-Olsson
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Susanna Myhre
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anders Gunnarsson
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Stefan Geschwindner
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna Aagaard
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna Björnson Granqvist
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM U1138 Team 1, Paris, France
| | - Yufeng Huang
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Kenneth L. Granberg
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson-Löfmark
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Judith Hartleib-Geschwindner
- Cardiovascular, Renal and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
80
|
Laursen SB, Finsen S, Marcussen N, Quaggin SE, Hansen PBL, Dimke H. Endothelial mineralocorticoid receptor ablation does not alter blood pressure, kidney function or renal vessel contractility. PLoS One 2018; 13:e0193032. [PMID: 29466427 PMCID: PMC5821352 DOI: 10.1371/journal.pone.0193032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 02/02/2018] [Indexed: 12/18/2022] Open
Abstract
Aldosterone blockade confers substantial cardiovascular and renal protection. The effects of aldosterone on mineralocorticoid receptors (MR) expressed in endothelial cells (EC) within the renal vasculature have not been delineated. We hypothesized that lack of MR in EC may be protective in renal vasculature and examined this by ablating the Nr3c2 gene in endothelial cells (EC-MR) in mice. Blood pressure, heart rate and PAH clearance were measured using indwelling catheters in conscious mice. The role of the MR in EC on contraction and relaxation was investigated in the renal artery and in perfused afferent arterioles. Urinary sodium excretion was determined by use of metabolic cages. EC-MR transgenics had markedly decreased MR expression in isolated aortic endothelial cells as compared to littermates (WT). Blood pressure and effective renal plasma flow at baseline and following AngII infusion was similar between groups. No differences in contraction and relaxation were observed between WT and EC-MR KO in isolated renal arteries during baseline or following 2 or 4 weeks of AngII infusion. The constriction or dilatations of afferent arterioles between genotypes were not different. No changes were found between the groups with respect to urinary excretion of sodium after 4 weeks of AngII infusion, or in urinary albumin excretion and kidney morphology. In conclusion, deletion of the EC-MR does not confer protection towards the development of hypertension, endothelial dysfunction of renal arteries or renal function following prolonged AngII-infusion.
Collapse
Affiliation(s)
- Sidsel B. Laursen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stine Finsen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| | - Susan E. Quaggin
- Feinberg Cardiovascular Research Institute and Division of Nephrology and Hypertension, Northwestern University, Chicago, IL, United States of America
| | - Pernille B. L. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Cardiovascular and Metabolic Disease, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- * E-mail:
| |
Collapse
|
81
|
RAGE-aptamer attenuates deoxycorticosterone acetate/salt-induced renal injury in mice. Sci Rep 2018; 8:2686. [PMID: 29422652 PMCID: PMC5805738 DOI: 10.1038/s41598-018-21176-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
The mineralocorticoid receptor (MR) and its downstream signaling play an important role in hypertensive renal injury. The interaction of advanced glycation end products (AGE) with their receptor (RAGE) is involved in the progression of renal disease. However, the pathological crosstalk between AGE–RAGE axis and MR system in kidney derangement remains unclear. We screened DNA-aptamer directed against RAGE (RAGE-apt) in vitro and examined its effects on renal injury in uninephrectomized deoxycorticosterone acetate (DOCA)/salt-induced hypertensive mice. RAGE, GTP-bound Rac-1 (Rac1), and MR were co-localized in the podocytes of DOCA mice. The deletion of RAGE gene significantly inhibited mesangial matrix expansion and tubulointerstitial fibrosis in DOCA mice, which was associated with the reduction of glomerular oxidative stress, MR, Rac1, and urinary albumin excretion (UAE) levels. RAGE-apt attenuated the increase in carboxymethyllysine (CML), RAGE, nitrotyrosine, Rac1, and MR levels in the kidneys and reduced UAE in DOCA mice. Aldosterone (Aldo) increased nitrotyrosine, CML, and RAGE gene expression in murine podocytes, whereas CML stimulated MR and Rac1 levels, which were blocked by RAGE-apt. The present study indicates the crosstalk between the AGE–RAGE axis and Aldo–MR system, suggesting that RAGE-apt may be a novel therapeutic tool for the treatment of MR-associated renal diseases.
Collapse
|
82
|
Antihypertensive therapy in nondiabetic chronic kidney disease: a review and update. ACTA ACUST UNITED AC 2018; 12:154-181. [PMID: 29396103 DOI: 10.1016/j.jash.2018.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 01/02/2018] [Accepted: 01/12/2018] [Indexed: 01/06/2023]
Abstract
Hypertension is an important contributor to progression of nondiabetic chronic kidney disease (CKD). Compelling observational evidence indicates that the divergence of blood pressure (BP) away from an ideal range in either direction is associated with a progressive rise in the risk of mortality and cardiovascular and renal disease progression. To date, various clinical trials and meta-analyses examining strict versus less intensive BP control in nondiabetic CKD have not conclusively demonstrated a renal advantage of one BP-lowering approach over another, except in certain subgroups such as proteinuric patients where evidence is circumstantial. As recent data have come to light suggesting that intensive BP control yields superior survival and cardiovascular outcomes in patients at high risk for cardiovascular disease, interest in the prospect of whether such benefit extends to individuals with CKD has surged. This review is a comprehensive analysis of antihypertensive literature in nondiabetic renal disease, with a particular emphasis on BP target.
Collapse
|
83
|
Sloan-Lancaster J, Raddad E, Deeg MA, Eli M, Flynt A, Tumlin J. Evaluation of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy After Single and Multiple Dosings of LY3016859 in Healthy Subjects and Patients With Diabetic Nephropathy. Clin Pharmacol Drug Dev 2018; 7:759-772. [PMID: 29385323 DOI: 10.1002/cpdd.436] [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: 06/28/2017] [Accepted: 12/07/2017] [Indexed: 11/06/2022]
Abstract
Two phase 1 studies (TGAA and TGAB) evaluated the safety, pharmacokinetics, pharmacodynamics, and efficacy of LY3016859 (LY), a monoclonal antibody that binds epiregulin and transforming growth factor α (TGF-α), administered intravenously or subcutaneously. In TGAA, 56 healthy subjects received a single dose of LY (0.1-750 mg intravenously, 50 mg subcutaneously) or placebo. In TGAB part A, 15 patients with diabetic nephropathy (DN) received 2 doses of LY (10-750 mg intravenously) or placebo, and in TGAB part B, 45 patients with DN received 5 doses of LY (50-750 mg intravenously) or placebo. Pharmacokinetics, pharmacodynamics, anti-LY antibodies, and change in proteinuria and albuminuria were evaluated. Single and multiple doses of LY administered 3 weeks apart were well tolerated. Pharmacokinetics were nonlinear in healthy subjects and patients with DN, indicating target-mediated drug disposition. Epiregulin level increased in both studies, and TGF-α levels increased in the TGAB study, consistent with target engagement; however, LY treatment did not significantly reduce proteinuria or albuminuria in patients with DN. There was no obvious effect of LY on the disease-related biomarkers monocyte chemoattractant protein-1, synaptopodin, or transferrin. Although LY administration resulted in a high frequency of anti-LY antibodies, pharmacokinetics, target engagement, and efficacy were not impacted.
Collapse
Affiliation(s)
| | - Eyas Raddad
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark A Deeg
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Michelle Eli
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Amy Flynt
- PharPoint Research, Inc., Durham, NC, USA
| | | |
Collapse
|
84
|
Non-uniform relationship between salt status and aldosterone activity in patients with chronic kidney disease. Clin Sci (Lond) 2018; 132:285-294. [PMID: 29321218 DOI: 10.1042/cs20171603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/28/2017] [Accepted: 01/07/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hypertension is prevalent in chronic kidney disease (CKD). Studies suggest that reduction in dietary salt intake reduces blood pressure (BP). We studied relationships between salt intake, BP and renin-angiotensin system regulation in order to establish if it is disordered in CKD. METHODS Mechanistic crossover study of CKD patients versus non-CKD controls. Participants underwent modified saline suppression test prior to randomization to either low or high salt diet for 5 days and then crossed over to the alternate diet. Angiotensin-II stimulation testing was performed in both salt states. BP, urea and electrolytes, and plasma aldosterone concentration (PAC) were measured. RESULTS Twenty-seven subjects were recruited (12 CKD, 15 control). There was no difference in age and baseline BP between the groups. Following administration of intravenous saline, systolic BP increased in CKD but not controls (131 ± 16 to 139 ± 14 mmHg, P=0.016 vs 125 ± 20 to 128 ± 22 mmHg, P=0.38). Median PAC reduced from 184 (124,340) to 95 (80,167) pmol in controls (P=0.003), but failed to suppress in CKD (230 (137,334) to 222 (147,326) pmol (P=0.17)). Following dietary salt modification, there was no change in BP in either group. Median PAC was lower following high salt compared with low salt diet in CKD and controls. There was a comparable increase in systolic BP in response to angiotensin-II in both groups. DISCUSSION We demonstrate dysregulation of aldosterone in CKD in response to salt loading with intravenous saline, but not to dietary salt modification.
Collapse
|
85
|
Lang F, Leibrock C, Pelzl L, Gawaz M, Pieske B, Alesutan I, Voelkl J. Therapeutic Interference With Vascular Calcification-Lessons From Klotho-Hypomorphic Mice and Beyond. Front Endocrinol (Lausanne) 2018; 9:207. [PMID: 29780355 PMCID: PMC5945862 DOI: 10.3389/fendo.2018.00207] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Medial vascular calcification, a major pathophysiological process associated with cardiovascular disease and mortality, involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). In chronic kidney disease (CKD), osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification is mainly driven by hyperphosphatemia, resulting from impaired elimination of phosphate by the diseased kidneys. Hyperphosphatemia with subsequent vascular calcification is a hallmark of klotho-hypomorphic mice, which are characterized by rapid development of multiple age-related disorders and early death. In those animals, hyperphosphatemia results from unrestrained formation of 1,25(OH)2D3 with subsequent retention of calcium and phosphate. Analysis of klotho-hypomorphic mice and mice with vitamin D3 overload uncovered several pathophysiological mechanisms participating in the orchestration of vascular calcification and several therapeutic opportunities to delay or even halt vascular calcification. The present brief review addresses the beneficial effects of bicarbonate, carbonic anhydrase inhibition, magnesium supplementation, mineralocorticoid receptor (MR) blockage, and ammonium salts. The case is made that bicarbonate is mainly effective by decreasing intestinal phosphate absorption, and that carbonic anhydrase inhibition leads to metabolic acidosis, which counteracts calcium-phosphate precipitation and VSMC transdifferentiation. Magnesium supplementation, MR blockage and ammonium salts are mainly effective by interference with osteo-/chondrogenic signaling in VSMCs. It should be pointed out that the, by far, most efficient substances are ammonium salts, which may virtually prevent vascular calcification. Future research will probably uncover further therapeutic options and, most importantly, reveal whether these observations in mice can be translated into treatment of patients suffering from vascular calcification, such as patients with CKD.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- *Correspondence: Florian Lang,
| | - Christina Leibrock
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- Fresenius Kabi Deutschland GmbH, Bad Homburg, Germany
| | - Lisann Pelzl
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Eberhard Karls-University, Tübingen, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
86
|
Okada K, Yamaguchi Y, Kamada N, Abe K, Wagou M, Katoh S, Sasaki H, Okada M, Saheki N. [The relationship between death from renal insufficiency and diuretic therapy among older adults at two nursing homes catering atomic bomb survivors]. Nihon Ronen Igakkai Zasshi 2018; 55:640-649. [PMID: 30542030 DOI: 10.3143/geriatrics.55.640] [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] [Indexed: 06/09/2023]
Abstract
AIM In Geriatrics Gerontology International we previously reported the efficacy of reducing diuretics to prevent falls and fractures in older adults. We have since noticed another important problem, regarding the diuretic therapy for older adults with decreased muscle and water volumes. We performed a study on renal insufficiency and diuretic therapy in an attempt to confirm the need for case control study between standard diuretic therapy administered, according to guidelines and "NY-mode" diuretic therapy, which involves the administration of the mineral-corticoid receptor inhibitor spironolactone at 12.5 mg orally every other day. METHODS We reviewed the causes of death among 1,855 residents living at 2 atomic bomb survivors nursing homes, with a focus on the death certification and diuretic therapy status of 48 older adults who died due to renal insufficiency. We also evaluated the relationship between the estimated glomerular filtration rate of 407 residents using serum creatinine data and the level of independence in daily life of disabled older adults. RESULTS We found that deaths due to chronic renal insufficiency were concentrated within certain period in two nursing homes examined and in patients receiving standard diuretic therapy (continuous daily loop diuretics or combination of loop diuretics with mineral-corticoid receptor inhibitor). Older adults with a relatively low level of independence in daily life showed a relatively higher estimated glomerular filtration rate, depending on their muscle volume. CONCLUSION These results suggest the need for a case control study of standard diuretic therapy and "NY-mode" therapy among older adults with decreased muscle and water volumes to not only prevent falls and fall-related fractures but also protect the kidney from damage.
Collapse
Affiliation(s)
- Kosuke Okada
- Department of Internal Medicine, COOP Saeki Hospital
- Former Hiroshima Atomic Bomb Survivors Nursing Home, Kandayama Yasuragien
| | - Yumiko Yamaguchi
- Former Hiroshima Atomic Bomb Survivors Nursing Home, Kurakake Nozomien
| | - Nanao Kamada
- Department of Internal Medicine, Hongou Central Hospital
- Former Hiroshima Atomic Bomb Survivors Relief Foundation
| | - Kazuhiro Abe
- Hiroshima Atomic Bomb Survivors Nursing Home, Kandayama Yasuragien
| | - Masakuni Wagou
- Hiroshima Atomic Bomb Survivors Nursing Home, Kurakake Nozomien
| | - Shigeko Katoh
- Faculty of Nursing, Hiroshima Bunnka Gakuen University
| | - Hidemi Sasaki
- Faculty of Nursing, Hiroshima Bunnka Gakuen University
| | - Masahiro Okada
- Department of Food and Dietetics, Two-year College of Hiroshima Bunka Gakuen
| | | |
Collapse
|
87
|
Cooper LB, Lippmann SJ, Greiner MA, Sharma A, Kelly JP, Fonarow GC, Yancy CW, Heidenreich PA, Hernandez AF. Use of Mineralocorticoid Receptor Antagonists in Patients With Heart Failure and Comorbid Diabetes Mellitus or Chronic Kidney Disease. J Am Heart Assoc 2017; 6:e006540. [PMID: 29275368 PMCID: PMC5779000 DOI: 10.1161/jaha.117.006540] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/26/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Perceived risks of hyperkalemia and acute renal insufficiency may limit use of mineralocorticoid receptor antagonist (MRA) therapy in patients with heart failure, especially those with diabetes mellitus or chronic kidney disease. METHODS AND RESULTS Using clinical registry data linked to Medicare claims, we analyzed patients hospitalized with heart failure between 2005 and 2013 with a history of diabetes mellitus or chronic kidney disease. We stratified patients by MRA use at discharge. We used inverse probability-weighted proportional hazards models to assess associations between MRA therapy and 30-day, 1-year, and 3-year mortality, all-cause readmission, and readmission for heart failure, hyperkalemia, and acute renal insufficiency. We performed interaction analyses for differential effects on 3-year outcomes for reduced, borderline, and preserved ejection fraction. Of 16 848 patients, 12.3% received MRA therapy at discharge. Higher serum creatinine was associated with lower odds of MRA use (odds ratio, 0.66; 95% confidence interval, 0.61-0.71); serum potassium was not (odds ratio, 1.00; 95% confidence interval, 0.90-1.11). There was no mortality difference between groups. MRA therapy was associated with greater risks of readmission for hyperkalemia and acute renal insufficiency and lower risks of long-term all-cause readmission. Patients on MRA therapy with borderline or preserved ejection fraction had greater risks of readmission for hyperkalemia (P=0.02) and acute renal insufficiency (P<0.001); patients with reduced ejection fraction did not. CONCLUSIONS Among patients with heart failure and diabetes mellitus or chronic kidney disease, MRA use was associated with lower risk of all-cause readmission despite greater risk of hyperkalemia and acute renal insufficiency.
Collapse
Affiliation(s)
- Lauren B Cooper
- Heart Failure and Transplant Program, Inova Heart & Vascular Institute, Inova Fairfax Hospital, Falls Church, VA
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Steven J Lippmann
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Melissa A Greiner
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Abhinav Sharma
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jacob P Kelly
- Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Gregg C Fonarow
- Ahmanson-UCLA Cardiomyopathy Center, University of California, Los Angeles, Los Angeles, CA
| | - Clyde W Yancy
- Department of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Adrian F Hernandez
- Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
88
|
Microalbuminuria and plasma aldosterone levels in nondiabetic treatment-naïve patients with hypertension. J Hypertens 2017; 35:2510-2516. [DOI: 10.1097/hjh.0000000000001476] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
89
|
Belden Z, Deiuliis JA, Dobre M, Rajagopalan S. The Role of the Mineralocorticoid Receptor in Inflammation: Focus on Kidney and Vasculature. Am J Nephrol 2017; 46:298-314. [PMID: 29017166 PMCID: PMC6863172 DOI: 10.1159/000480652] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The remarkable success of clinical trials in mineralocorticoid receptor (MR) inhibition in heart failure has driven research on the physiological and pathological role(s) of nonepithelial MR expression. MR is widely expressed in the cardiovascular system and is a major determinant of endothelial function, smooth muscle tone, vascular remodeling, fibrosis, and blood pressure. An important new dimension is the appreciation of the role MR plays in immune cells and target organ damage in the heart, kidney and vasculature, and in the development of insulin resistance. SUMMARY The mechanism for MR activation in tissue injury continues to evolve with the evidence to date suggesting that activation of MR results in a complex repertoire of effects involving both macrophages and T cells. MR is an important transcriptional regulator of macrophage phenotype and function. Another important feature of MR activation is that it can occur even with normal or low aldosterone levels in pathological conditions. Tissue-specific conditional models of MR expression in myeloid cells, endothelial cells, smooth muscle cells and cardiomyocytes have been very informative and have firmly demonstrated a critical role of MR as a key pathophysiologic variable in cardiac hypertrophy, transition to heart failure, adipose inflammation, and atherosclerosis. Finally, the central nervous system activation of MR in permeable regions of the blood-brain barrier may play a role in peripheral inflammation. Key Message: Ongoing clinical trials will help clarify the role of MR blockade in conditions, such as atherosclerosis and chronic kidney disease.
Collapse
Affiliation(s)
- Zachary Belden
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey A. Deiuliis
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mirela Dobre
- Division of Nephrology and Hypertension, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sanjay Rajagopalan
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW Several interrelated mechanisms promote the development of hypertension in obesity, often contributing to end organ damage including cardiovascular disease and chronic kidney disease. RECENT FINDINGS The treatment of hypertension in obesity is complicated by a high prevalence of resistant hypertension, as well as unpredictable hemodynamic effects of many medications. Weight loss stabilizes neurohormonal activity and causes clinically significant reductions in blood pressure. While lifestyle interventions can improve blood pressure, they fail to consistently yield sustained weight loss and have not demonstrated long-term benefits. Bariatric surgery provides more permanent weight reduction, corresponding with dramatic declines in blood pressure and attenuation of long-term cardiovascular risk. Hypertension is closely linked to the prevalence, pathophysiology, and morbidity of obesity. There are multiple barriers to managing hypertension in obesity. Surgical weight loss offers the most promise in reducing blood pressure and decreasing end organ damage in this patient population.
Collapse
|
91
|
Abstract
Proteinuria and decline of renal function are associated with progression of kidney disease. The Renin Angiotensin Aldosterone System (RAAS) plays an important role in blood pressure regulation, fluid volume, and sodium balance. Overactivity of RAAS contributes to the pathogenesis of a variety of clinical conditions including progress of chronic kidney disease (CKD). This review summarizes the use of RAAS inhibitors as dual therapy or monotherapy in different stages of kidney disease. Experimental and clinical studies have demonstrated RAAS inhibitors prevent proteinuria, kidney fibrosis and slow decline of renal function and thus play a protective role in both early and end stages of kidney disease. While combination use of RAAS inhibitors showed higher efficiency compared with monotherapy, it is also associated with higher incidence of adverse events. Besides ACEI/ARBs, more mechanism research of mineralocorticoid receptor antagonists in kidney disease should be performed.
Collapse
|
92
|
Alesutan I, Voelkl J, Feger M, Kratschmar DV, Castor T, Mia S, Sacherer M, Viereck R, Borst O, Leibrock C, Gawaz M, Kuro-O M, Pilz S, Tomaschitz A, Odermatt A, Pieske B, Wagner CA, Lang F. Involvement Of Vascular Aldosterone Synthase In Phosphate-Induced Osteogenic Transformation Of Vascular Smooth Muscle Cells. Sci Rep 2017; 7:2059. [PMID: 28515448 PMCID: PMC5435689 DOI: 10.1038/s41598-017-01882-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/04/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification resulting from hyperphosphatemia is a major determinant of mortality in chronic kidney disease (CKD). Vascular calcification is driven by aldosterone-sensitive osteogenic transformation of vascular smooth muscle cells (VSMCs). We show that even in absence of exogenous aldosterone, silencing and pharmacological inhibition (spironolactone, eplerenone) of the mineralocorticoid receptor (MR) ameliorated phosphate-induced osteo-/chondrogenic transformation of primary human aortic smooth muscle cells (HAoSMCs). High phosphate concentrations up-regulated aldosterone synthase (CYP11B2) expression in HAoSMCs. Silencing and deficiency of CYP11B2 in VSMCs ameliorated phosphate-induced osteogenic reprogramming and calcification. Phosphate treatment was followed by nuclear export of APEX1, a CYP11B2 transcriptional repressor. APEX1 silencing up-regulated CYP11B2 expression and stimulated osteo-/chondrogenic transformation. APEX1 overexpression blunted the phosphate-induced osteo-/chondrogenic transformation and calcification of HAoSMCs. Cyp11b2 expression was higher in aortic tissue of hyperphosphatemic klotho-hypomorphic (kl/kl) mice than in wild-type mice. In adrenalectomized kl/kl mice, spironolactone treatment still significantly ameliorated aortic osteoinductive reprogramming. Our findings suggest that VSMCs express aldosterone synthase, which is up-regulated by phosphate-induced disruption of APEX1-dependent gene suppression. Vascular CYP11B2 may contribute to stimulation of VSMCs osteo-/chondrogenic transformation during hyperphosphatemia.
Collapse
Affiliation(s)
- Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Jakob Voelkl
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
| | - Martina Feger
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Tatsiana Castor
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Michael Sacherer
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
| | - Robert Viereck
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | | | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria
| | - Andreas Tomaschitz
- Div. of Cardiology, Medical University of Graz and Ludwig Boltzmann Institute for Translational Heart Failure Research, Graz, Austria
- Bad Gleichenberg Clinic, Bad Gleichenberg, Austria
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, and the National Center for Excellence in Research NCCR Kidney.CH, University of Basel, Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Department of Cardiology, University of Graz, Graz, Austria; Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, and the National Center for Excellence in Research NCCR Kidney, Zurich, Switzerland
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
93
|
Advances in the management of hyperkalemia in chronic kidney disease. Curr Opin Nephrol Hypertens 2017; 26:235-239. [DOI: 10.1097/mnh.0000000000000320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
94
|
Cooper LB, Hammill BG, Peterson ED, Pitt B, Maciejewski ML, Curtis LH, Hernandez AF. Characterization of Mineralocorticoid Receptor Antagonist Therapy Initiation in High-Risk Patients With Heart Failure. Circ Cardiovasc Qual Outcomes 2017; 10:e002946. [PMID: 28073850 PMCID: PMC5228387 DOI: 10.1161/circoutcomes.116.002946] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/21/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Heart failure guidelines recommend routine monitoring of serum potassium, and renal function in patients treated with a mineralocorticoid receptor antagonist (MRA). How these recommendations are implemented in high-risk patients or according to setting of drug initiation is poorly characterized. METHODS AND RESULTS We conducted a retrospective cohort study of Medicare beneficiaries linked to laboratory data in 10 states with prevalent heart failure as of July 1, 2011, and incident MRA use between May 1 and September 30, 2011. Outcomes included laboratory testing before MRA initiation and in the early (days 1-10) and extended (days 11-90) post-initiation periods, based on setting of drug initiation and the presence of renal insufficiency. Additional outcomes included abnormal laboratory results and adverse events proximate to MRA initiation. Of 10 443 Medicare beneficiaries with heart failure started on an MRA, 19.7% were initiated during a hospitalization. Appropriate follow-up laboratory testing across all time periods occurred in 25.2% of patients with inpatient initiation compared with 2.8% of patients begun as an outpatient. Patients with chronic kidney disease had higher rates of both hyperkalemia and acute kidney failure in the early (1.3% and 2.7%, respectively) and extended (5.6% and 9.8%, respectively) post-initiation periods compared with those without chronic kidney disease. CONCLUSIONS Patients initiated on MRA therapy as an outpatient had extremely poor rates of guideline indicated follow-up laboratory monitoring after drug initiation. In particular, patients with chronic kidney disease are at high risk for adverse events after MRA initiation. Quality improvement initiatives focused on systems to improve appropriate laboratory monitoring are needed.
Collapse
Affiliation(s)
- Lauren B Cooper
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.).
| | - Bradley G Hammill
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| | - Eric D Peterson
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| | - Bertram Pitt
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| | - Matthew L Maciejewski
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| | - Lesley H Curtis
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| | - Adrian F Hernandez
- From the Inova Heart and Vascular Institute, Falls Church, VA (L.B.C.); Department of Medicine (L.B.C., E.D.P., M.L.M., L.H.C., A.F.H.) and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (B.G.H., E.D.P., L.H.C., A.F.H.); Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.); and Center for Health Services Research in Primary Care, Durham Veterans Affairs Medical Center, NC (M.L.M.)
| |
Collapse
|
95
|
|
96
|
Sinnott SJ, Tomlinson LA, Root AA, Mathur R, Mansfield KE, Smeeth L, Douglas IJ. Comparative effectiveness of fourth-line anti-hypertensive agents in resistant hypertension: A systematic review and meta-analysis. Eur J Prev Cardiol 2016; 24:228-238. [DOI: 10.1177/2047487316675194] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Sarah-Jo Sinnott
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Laurie A Tomlinson
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Adrian A Root
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Rohini Mathur
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Kathryn E Mansfield
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Liam Smeeth
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| | - Ian J Douglas
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, UK
| |
Collapse
|
97
|
Vaden SL, Elliott J. Management of Proteinuria in Dogs and Cats with Chronic Kidney Disease. Vet Clin North Am Small Anim Pract 2016; 46:1115-30. [DOI: 10.1016/j.cvsm.2016.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
98
|
Abstract
PURPOSE OF REVIEW This review will highlight recent developments in mineralocorticoid receptor research which impact aldosterone-associated vascular and cardiometabolic dysfunction. RECENT FINDINGS The mineralocorticoid receptor is also expressed in vascular smooth muscle and vascular endothelium, and contributes to vascular function and remodeling. Adipocyte-derived leptin stimulates aldosterone secretion, which may explain the observed link between obesity and hyperaldosteronism. Adipocyte mineralocorticoid receptor overexpression produces systemic changes consistent with metabolic syndrome. Ongoing studies with novel nonsteroidal mineralocorticoid receptor antagonists may provide a novel treatment for diabetic nephropathy and heart failure in patients with chronic kidney disease, with reduced risk of hyperkalemia. SUMMARY Ongoing research continues to demonstrate novel roles of the vascular and adipocyte mineralocorticoid receptor function, which may explain the beneficial metabolic and vascular benefits of mineralocorticoid receptor antagonists.
Collapse
|
99
|
Currie G, Taylor AHM, Fujita T, Ohtsu H, Lindhardt M, Rossing P, Boesby L, Edwards NC, Ferro CJ, Townend JN, van den Meiracker AH, Saklayen MG, Oveisi S, Jardine AG, Delles C, Preiss DJ, Mark PB. Effect of mineralocorticoid receptor antagonists on proteinuria and progression of chronic kidney disease: a systematic review and meta-analysis. BMC Nephrol 2016; 17:127. [PMID: 27609359 PMCID: PMC5015203 DOI: 10.1186/s12882-016-0337-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 08/25/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Hypertension and proteinuria are critically involved in the progression of chronic kidney disease. Despite treatment with renin angiotensin system inhibition, kidney function declines in many patients. Aldosterone excess is a risk factor for progression of kidney disease. Hyperkalaemia is a concern with the use of mineralocorticoid receptor antagonists. We aimed to determine whether the renal protective benefits of mineralocorticoid antagonists outweigh the risk of hyperkalaemia associated with this treatment in patients with chronic kidney disease. METHODS We conducted a meta-analysis investigating renoprotective effects and risk of hyperkalaemia in trials of mineralocorticoid receptor antagonists in chronic kidney disease. Trials were identified from MEDLINE (1966-2014), EMBASE (1947-2014) and the Cochrane Clinical Trials Database. Unpublished summary data were obtained from investigators. We included randomised controlled trials, and the first period of randomised cross over trials lasting ≥4 weeks in adults. RESULTS Nineteen trials (21 study groups, 1 646 patients) were included. In random effects meta-analysis, addition of mineralocorticoid receptor antagonists to renin angiotensin system inhibition resulted in a reduction from baseline in systolic blood pressure (-5.7 [-9.0, -2.3] mmHg), diastolic blood pressure (-1.7 [-3.4, -0.1] mmHg) and glomerular filtration rate (-3.2 [-5.4, -1.0] mL/min/1.73 m(2)). Mineralocorticoid receptor antagonism reduced weighted mean protein/albumin excretion by 38.7 % but with a threefold higher relative risk of withdrawing from the trial due to hyperkalaemia (3.21, [1.19, 8.71]). Death, cardiovascular events and hard renal end points were not reported in sufficient numbers to analyse. CONCLUSIONS Mineralocorticoid receptor antagonism reduces blood pressure and urinary protein/albumin excretion with a quantifiable risk of hyperkalaemia above predefined study upper limit.
Collapse
Affiliation(s)
- Gemma Currie
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Center, 126 University Place, Glasgow, UK.
| | - Alison H M Taylor
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Center, 126 University Place, Glasgow, UK
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ohtsu
- Department of Clinical Study and Informatics, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Peter Rossing
- Steno Diabetes Center, Niels Steensens Vej, Gentofte, Denmark
- Health, Aarhus University, Aarhus, Denmark
- NNF Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Lene Boesby
- Department of Nephrology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Nicola C Edwards
- Departments of Cardiology and Nephrology, University Hospital Birmingham and School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK
| | - Charles J Ferro
- Departments of Cardiology and Nephrology, University Hospital Birmingham and School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK
| | - Jonathan N Townend
- Departments of Cardiology and Nephrology, University Hospital Birmingham and School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK
| | | | | | - Sonia Oveisi
- Metabolic Diseases Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Alan G Jardine
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Center, 126 University Place, Glasgow, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Center, 126 University Place, Glasgow, UK
| | - David J Preiss
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Oxford, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Center, 126 University Place, Glasgow, UK
| |
Collapse
|
100
|
Malachias MVB, Amodeo C, Paula RB, Cordeiro AC, Magalhães LBNC, Bodanese LC. 7th Brazilian Guideline of Arterial Hypertension: Chapter 8 - Hypertension and Associated Clinical Conditions. Arq Bras Cardiol 2016; 107:44-48. [PMID: 27819387 PMCID: PMC5319465 DOI: 10.5935/abc.20160158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|