1
|
Mayne KJ, Sardell RJ, Staplin N, Judge PK, Zhu D, Sammons E, Cherney DZI, Green JB, Levin A, Pontremoli R, Hauske SJ, Emberson J, Preiss D, Landray MJ, Baigent C, Wanner C, Haynes R, Herrington WG. Empagliflozin lowers serum uric acid in chronic kidney disease: exploratory analyses from the EMPA-KIDNEY trial. Nephrol Dial Transplant 2025; 40:720-730. [PMID: 39277784 PMCID: PMC7616479 DOI: 10.1093/ndt/gfae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Hyperuricaemia and gout are common in chronic kidney disease (CKD). We aimed to assess the effects of sodium-glucose co-transporter-2 (SGLT2) inhibition on uric acid (urate) and gout in patients with CKD. METHODS The EMPA-KIDNEY trial randomised 6609 patients with CKD to receive either empagliflozin 10 mg daily or matching placebo over a median of 2 years of follow-up. Serum uric acid was measured at randomisation then at 2 and 18 months of follow-up and the effects of empagliflozin were analysed using a pre-specified mixed model repeated measures approach. Participant-reported gout events were analysed in Cox regression models (first events) with the Andersen-Gill extension (total events). A post hoc composite outcome included new initiation of uric acid-lowering therapy or colchicine. EMPA-KIDNEY primary and kidney disease progression outcomes were also assessed in subgroups of baseline serum uric acid. RESULTS Baseline mean ± standard deviation serum uric acid concentration was 431 ± 114 µmol/l. Allocation to empagliflozin resulted in a study-average between-group difference in serum uric acid of -25.6 µmol/l [95% confidence interval (CI) -30.3 to -21.0], with larger effects in those with higher eGFR (trend P < .001) and without diabetes (heterogeneity P < .001). Compared with placebo, empagliflozin did not significantly reduce first or total gout events [hazard ratio 0.87 (95% CI 0.74-1.02) for the 595 first events and 0.86 (0.72-1.03) for the 869 total events] with similar hazard ratios for the post hoc composite and across subgroups, including by diabetes and eGFR. The effect of empagliflozin on the primary outcome and kidney disease progression outcomes were similar irrespective of the baseline level of uric acid. CONCLUSIONS SGLT2 inhibition reduces serum uric acid in patients with CKD, with larger effects at higher eGFR and in the absence of diabetes. However, the effect on uric acid is modest and did not translate into reduced risk of gout in EMPA-KIDNEY.
Collapse
Affiliation(s)
- Kaitlin J Mayne
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rebecca J Sardell
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Natalie Staplin
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Parminder K Judge
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Oxford Kidney Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Doreen Zhu
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Oxford Kidney Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Emily Sammons
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | | | - Adeera Levin
- University of British Columbia, Vancouver, BC, Canada
| | - Roberto Pontremoli
- Università degli Studi and IRCCS Ospedale Policlinico San Martino di Genova, Genova, Italy
| | - Sibylle J Hauske
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany & Vth Department of Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jonathan Emberson
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David Preiss
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Martin J Landray
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Colin Baigent
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christoph Wanner
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Richard Haynes
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Oxford Kidney Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - William G Herrington
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Oxford Kidney Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
2
|
Takata T, Taniguchi S, Mae Y, Kageyama K, Fujino Y, Iyama T, Hikita K, Sugihara T, Isomoto H. Comparative assessment of the effects of dotinurad and febuxostat on the renal function in chronic kidney disease patients with hyperuricemia. Sci Rep 2025; 15:8990. [PMID: 40089552 PMCID: PMC11910530 DOI: 10.1038/s41598-025-94020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Although hyperuricemia is associated with chronic kidney disease (CKD), the impact of uric acid (UA)-lowering drugs on CKD has been controversial. Previous investigations have primarily included xanthine oxidase inhibitors; therefore, research of dotinurad, a recently developed selective urate reabsorption inhibitor, is necessary. This retrospective study included 58 patients with CKD; of these, 29 newly initiated dotinurad and 29 initiated febuxostat. The effects of dotinurad and febuxostat on the serum UA, urinary UA-to-creatinine ratio (UUCR), and estimated glomerular filtration rate (eGFR) during 3 months were analyzed to compare their impacts on renal function. Dotinurad and febuxostat decreased serum UA (8.40 ± 1.11 to 6.50 ± 0.80 mg/dL [p < 0.001] and 8.91 ± 1.21 to 6.05 ± 1.28 mg/dL [p = < 0.001], respectively). The UUCR increased after dotinurad (0.35 ± 0.15 to 0.40 ± 0.21 g/gCr [p = 0.024]); however, it decreased after febuxostat (0.33 ± 0.12 to 0.21 ± 0.06 g/gCr [p = 0.002]). The eGFR improved after dotinurad (33.9 ± 15.2 to 36.2 ± 15.9 mL/min/1.73 m2 [p < 0.001]). No change was observed after febuxostat treatment (33.4 ± 19.6 to 34.1 ± 21.6 mL/min/1.73 m2). Renal function improved only with dotinurad, thus highlighting its renoprotective effects beyond the reduction of serum UA.
Collapse
Affiliation(s)
- Tomoaki Takata
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan.
| | - Sosuke Taniguchi
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Kana Kageyama
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Yudai Fujino
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Takuji Iyama
- Kidney Center, Tottori University Hospital, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Katsuya Hikita
- Kidney Center, Tottori University Hospital, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Takaaki Sugihara
- School of Health Science, Major in Clinical Laboratory Science, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| |
Collapse
|
3
|
Yang H, Ying J, Zu T, Meng XM, Jin J. Insights into renal damage in hyperuricemia: Focus on renal protection (Review). Mol Med Rep 2025; 31:59. [PMID: 39717954 PMCID: PMC11711934 DOI: 10.3892/mmr.2024.13424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
The incidence of hyperuricemia has increased recently, posing a serious threat to public health. Hyperuricemia is associated with an increased risk of gout, chronic kidney disease (CKD), obesity, metabolic syndrome, type 2 diabetes mellitus, hypertension, hypertriglyceridaemia, metabolic dysfunction‑associated steatotic liver disease, acute kidney injury, coronary heart disease and cardiovascular disease (CVD). These diseases are commonly accompanied by varying degrees of kidney damage. A number of randomized controlled clinical trials have investigated the effectiveness of UA‑lowering therapies in preventing kidney disease progression. The present review provided fundamental insights into the pathogenesis, principles and therapeutic approaches for managing hyperuricemia in patients with aforementioned diseases and assesses the effect of uric acid‑lowering therapy on diabetic nephropathy, systemic lupus erythematosus, CKD, CVD and obesity progression.
Collapse
Affiliation(s)
- Hang Yang
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jie Ying
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Tong Zu
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei, Anhui 230032, P.R. China
| | - Juan Jin
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
4
|
Li X, Huang B, Liu Y, Wang M, Cui JQ. Uric acid in diabetic microvascular complications: Mechanisms and therapy. J Diabetes Complications 2025; 39:108929. [PMID: 39689504 DOI: 10.1016/j.jdiacomp.2024.108929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Uric acid (UA) is mainly synthesized in the liver, intestine, and vascular endothelium and excreted by the kidney (70 %) and intestine (30 %). Hyperuricemia (HUA) occurs when UA production exceeds excretion. Many studies have found that elevated UA is associated with diabetic microvascular complications (DMC), including diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic peripheral neuropathy (DPN). In addition, too high or too low UA levels will promote the occurrence and development of chronic diseases, but the relationship between UA and diabetic microvascular complications (DMC) is not clear. Therefore, the rational treatment of UA in patients with diabetes is essential. In this review, we summarize and discuss the mechanism and treatment of UA and DMC and may provide potential advice for rational drug selection.
Collapse
Affiliation(s)
- Xin Li
- Tianjin Medical University General Hospital, People's Republic of China
| | - Bo Huang
- Tianjin Medical University General Hospital, People's Republic of China
| | - Yue Liu
- Tianjin Medical University General Hospital, People's Republic of China
| | - Meng Wang
- Tianjin Medical University General Hospital, People's Republic of China
| | - Jing-Qiu Cui
- Tianjin Medical University General Hospital, People's Republic of China.
| |
Collapse
|
5
|
Gu W, Zhao J, Xu Y. Hyperuricemia-induced complications: dysfunctional macrophages serve as a potential bridge. Front Immunol 2025; 16:1512093. [PMID: 39935474 PMCID: PMC11810932 DOI: 10.3389/fimmu.2025.1512093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
With the changes in modern life, hyperuricemia (HUA) has become a serious universal health issue, leading to rising morbidity and mortality. Characterized by elevated levels of UA, HUA has become an independent risk factor for gout, chronic kidney disease, insulin resistance, cardiovascular disease, nonalcoholic fatty liver disease, etc. As HUA is a metabolic syndrome, the immune response is likely to play an active role throughout the whole process. Moreover, macrophages, as an indispensable component of the immune system, may serve as a promising target for addressing hyperuricemia-induced inflammation. Along with their precursor cells, monocytes, macrophages play a key role in the pathogenesis of HUA, primarily through three specific aspects, all of which are associated with inflammatory cytokines. The first mechanism involves direct action on urate transporters, such as URAT1 and ABCG2. The second mechanism is the modulation of inflammation, including targeting toll-like receptors (TLRs) and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. The third mechanism pertains to the effects on oxidative stress mediators. In this review, we summarize the underlying mechanisms of hyperuricemia, focusing on the effects of macrophages, therapeutic approaches, and clinical trials addressing hyperuricemia-caused dysfunction. Additionally, we highlight directions for future development, aiming to support future theoretical studies.
Collapse
Affiliation(s)
- Wenyi Gu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Zhao
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yu Xu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for Traditional Chinese Medicine New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Yamamoto T, Kasahara M, Ueshima K, Uemura S, Kashihara N, Kimura K, Konta T, Shoji T, Mima A, Mukoyama M, Saito Y. Multicenter randomized controlled trial of intensive uric acid lowering therapy for CKD patients with hyperuricemia: TARGET-UA. Clin Exp Nephrol 2024; 28:764-772. [PMID: 38530491 PMCID: PMC11266370 DOI: 10.1007/s10157-024-02483-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND We investigate whether Intensive uric acid (UA)-lowering therapy (ULT) provides increased renal protection compared with standard therapy in chronic kidney disease (CKD) patients. METHODS This was a multicenter randomized controlled trial. Only CKD patients with hyperuricemia were included in this study. The participants were randomly assigned to either the Intensive therapy group (target serum UA level ≥ 4.0 mg/dL and < 5.0 mg/dL) or the standard therapy group (serum UA level ≥ 6.0 mg/dL and < 7.0 mg/dL). ULT was performed using topiroxostat, a non-purine-type selective xanthine oxidase inhibitor. The primary endpoint was change in the logarithmic value of urine albumin to the creatinine ratio (ACR) between baseline and week 52 of the treatment. RESULTS Three hundred fifty-two patients were included in the full analysis set. In the Standard therapy group, mean serum UA was 8.23 mg/dL at baseline and 6.13 mg/dL at 52 weeks. In the Intensive therapy group, mean serum UA was 8.15 mg/dL at baseline and 5.25 mg/dL at 52 weeks. There was no significant difference in changes in log ACR at 52 weeks between the Intensive therapy and the Standard therapy groups. CONCLUSION This study did not reveal the benefit of Intensive ULT to improve albuminuria levels. (UMIN000026741 and jRCTs051180146).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Akira Mima
- Osaka Medical and Pharmaceutical University, Osaka, Japan
| | | | | |
Collapse
|
7
|
Hofherr A, Liarte Marin E, Musial B, Seth A, Slidel T, Conway J, Baker D, Hansen PB, Challis B, Bartesaghi S, Bhat M, Pecoits-Filho R, Tu X, Selvarajah V, Woollard K, Heerspink HJ. Inhibition of Interleukin-33 to Reduce Glomerular Endothelial Inflammation in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1876-1891. [PMID: 38899206 PMCID: PMC11184260 DOI: 10.1016/j.ekir.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Inflammation is a significant contributor to cardiorenal morbidity and mortality in diabetic kidney disease (DKD). The pathophysiological mechanisms linking systemic, subacute inflammation and local, kidney injury-initiated immune maladaptation is partially understood. Methods Here, we explored the expression of proinflammatory cytokines in patients with DKD; investigated mouse models of type 1 and type 2 diabetes (T2D); evaluated glomerular signaling in vitro; performed post hoc analyses of systemic and urinary markers of inflammation; and initiated a phase 2b clinical study (FRONTIER-1; NCT04170543). Results Transcriptomic profiling of kidney biopsies from patients with DKD revealed significant glomerular upregulation of interleukin-33 (IL-33). Inhibition of IL-33 signaling reduced glomerular damage and albuminuria in the uninephrectomized db/db mouse model (T2D/DKD). On a cellular level, inhibiting IL-33 improved glomerular endothelial health by decreasing cellular inflammation and reducing release of proinflammatory cytokines. Therefore, FRONTIER-1 was designed to test the safety and efficacy of the IL-33-targeted monoclonal antibody tozorakimab in patients with DKD. So far, 578 patients are enrolled in FRONTIER-1. The baseline inflammation status of participants (N > 146) was assessed in blood and urine. Comparison to independent reference cohorts (N > 200) validated the distribution of urinary tumor necrosis factor receptor 1 (TNFR1) and C-C motif chemokine ligand 2 (CCL2). Treatment with dapagliflozin for 6 weeks did not alter these biomarkers significantly. Conclusion We show that blocking the IL-33 pathway may mitigate glomerular endothelial inflammation in DKD. The findings from the FRONTIER-1 study will provide valuable insights into the therapeutic potential of IL-33 inhibition in DKD.
Collapse
Affiliation(s)
- Alexis Hofherr
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elena Liarte Marin
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Barbara Musial
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Asha Seth
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Tim Slidel
- Bioinformatics, Oncology R&D, AstraZeneca, Cambridge, UK
| | - James Conway
- Bioinformatics, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - David Baker
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Pernille B.L. Hansen
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benjamin Challis
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefano Bartesaghi
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Bhat
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Roberto Pecoits-Filho
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
- School of Medicine, Pontificia Universidade de Catolica do Parana, Curitiba, Brazil
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Xiao Tu
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Viknesh Selvarajah
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kevin Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Hiddo J.L. Heerspink
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
8
|
Xue X, Sun M, Yan F, Dalbeth N, He Y, Li X, Qi H, Chen Y, Yuan X, Li M, Ji A, Terkeltaub R, Li C. Superiority of Low-Dose Benzbromarone Add-On to Low-Dose Febuxostat Compared With Febuxostat Monotherapy in Gout With Combined-Type Hyperuricemia. Arthritis Care Res (Hoboken) 2024; 76:703-711. [PMID: 38130040 PMCID: PMC11039362 DOI: 10.1002/acr.25283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/17/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE There is an unmet need for simpler urate-lowering therapy (ULT) regimens that achieve the serum urate target and improve the overall quality of gout care. We report a comparative effectiveness trial of febuxostat monotherapy versus benzbromarone add-on to low-dose febuxostat in gout specifically with combined renal urate underexcretion and overload. METHODS A prospective randomized trial was conducted on patients with combined-type hyperuricemia and estimated glomerular filtration rate >60 mL/min/1.73 m2 1:1 randomly assigned to febuxostat and benzbromarone combination therapy (initially febuxostat at 20 mg/day, with benzbromarone at 25 mg/day added onto 20 mg/day of febuxostat if not at target) or febuxostat monotherapy (initially 20 mg/day, escalating to 40 mg/day if not at target). The primary end point at 12 weeks was the proportion achieving a serum urate (SU) level <360 μmol/L. Other outcomes included altered liver and kidney function, new-onset urolithiasis, and gout flares. RESULTS There were 250 participants randomized; 219 completed 12-week treatment. More patients in the febuxostat and benzbromarone combination group achieved the SU target compared to patients in the febuxostat monotherapy group (75.5% vs 47.7%; odds ratio 3.37 [95% confidence interval 1.90-5.98]). Safety profiles were comparable between the two groups. CONCLUSION Simply adding on low-dose benzbromarone (25 mg/day) to low-dose (20 mg/day) febuxostat showed superior urate lowering compared to febuxostat monotherapy in gout with a combined-type hyperuricemia. For selected patients, expedited achievement of the SU target in more than 75% of patients using one titration step and low xanthine oxidase inhibitor and uricosuric doses is a potential alternative to standard ULT regimens.
Collapse
Affiliation(s)
- Xiaomei Xue
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingshu Sun
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- Department of Rheumatology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fei Yan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Yuwei He
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinde Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Qi
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Chen
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| | - Xuan Yuan
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Maichao Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aichang Ji
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Robert Terkeltaub
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Changgui Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Heerspink HJ, Stack AG, Terkeltaub R, Jongs N, Inker LA, Bjursell M, Maklad N, Perl S, Eklund O, Rikte T, Sjöström CD, Perkovic V. Combination Treatment with Verinurad and Allopurinol in CKD: A Randomized Placebo and Active Controlled Trial. J Am Soc Nephrol 2024; 35:594-606. [PMID: 38564654 PMCID: PMC11149044 DOI: 10.1681/asn.0000000000000326] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
Key Points The SAPPHIRE trial was designed to assess albuminuria-lowering effects of the urate transporter 1 inhibitor verinurad combined with allopurinol in patients with CKD. Verinurad 3, 7.5, and 12 mg in combination with allopurinol 300 mg did not reduce albuminuria during 34 weeks treatment compared with allopurinol alone or placebo. Verinurad/allopurinol combination dose-dependently reduced serum urate concentrations compared with placebo. Background Hyperuricemia is associated with elevated risks of cardiovascular and chronic kidney disease (CKD). Since inhibition of urate transporter 1 has been suggested to be potentially nephroprotective, we performed a phase 2b study to assess albuminuria-lowering effects of the urate transporter 1 inhibitor verinurad combined with the xanthine oxidase inhibitor allopurinol in patients with CKD and hyperuricemia. Methods In this randomized placebo and active controlled trial, we enrolled participants with serum urate concentrations ≥6.0 mg/dl, eGFR ≥25 ml/min per 1.73 m2, and a urinary albumin-creatinine ratio (UACR) 30–5000 mg/g to one of five treatment arms: placebo, placebo+allopurinol 300 mg/day, verinurad 3 mg+allopurinol 300 mg/day, verinurad 7.5 mg+allopurinol 300 mg/day, or verinurad 12 mg+allopurinol 300 mg/day in a 1:1:1:1:1 ratio. The primary end point was the change in UACR from baseline to 34 weeks. Secondary end points were changes from baseline in UACR at week 60 and changes in serum urate and eGFR at weeks 34 and 60. Results Between August 2019 and November 2021, 861 adults with CKD (mean age 65 years, 33.0% female, mean eGFR 48 ml/min per 1.73 m2, median UACR 217 mg/g) were enrolled. At 34 weeks, the geometric mean percentage change in UACR from baseline did not differ among treatment groups (16.7%, 95% confidence interval [CI], −0.6 to 37.1 in the 3 mg group, 15.0% [95% CI, −1.85 to 34.6] in the 7.5 mg group, 14.0% [95% CI, −3.4 to 34.4] in the 12 mg group versus 9.9% [95% CI, −6.6 to 29.4] in the allopurinol group, and 37.3% [95% CI, 16.6 to 61.8] in the placebo group). UACR and eGFR change from baseline did not differ among treatment groups after 60 weeks. Verinurad/allopurinol combination dose-dependently reduced serum urate concentrations compared with placebo. The proportion of patients with adverse events and serious adverse events was balanced among treatment groups. Conclusions Verinurad in combination with allopurinol did not decrease UACR or eGFR decline, but further reduced serum urate compared with allopurinol alone or placebo. Clinical Trial registry name and registration number: SAPPHIRE Trial registration number, NCT03990363 .
Collapse
Affiliation(s)
- Hiddo J.L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- The George Institute for Global Health, Sydney, New South Wales, Australia
| | - Austin G. Stack
- School of Medicine & Health Research Institute, University of Limerick, Limerick, Ireland
| | | | - Niels Jongs
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lesley A. Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Magnus Bjursell
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Noha Maklad
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gaithersburg, Maryland
| | - Shira Perl
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gaithersburg, Maryland
| | - Olof Eklund
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Tord Rikte
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - C. David Sjöström
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca, Gothenburg, Sweden
| | - Vlado Perkovic
- The George Institute for Global Health, Sydney, New South Wales, Australia
- University New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Fenta ET, Kidie AA, Tiruneh MG, Anagaw TF, Bogale EK, Dessie AA, Worku NK, Amera MG, Tesfa H, Limenh LW, Delie AM, Ayal BG. Exploring barriers of health literacy on non-communicable disease prevention and care among patients in north wollo zone public hospitals; Northeast, Ethiopia, 2023: application of socio-ecological model. BMC Public Health 2024; 24:971. [PMID: 38581006 PMCID: PMC10998356 DOI: 10.1186/s12889-024-18524-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/04/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Health literacy is the important for the prevention of non-communicable disease to make informed health decisions, and practice healthy and protective behaviours. Therefore, application of socioecological model to this study aimed to identify multilevel factors on health literacy among patients and develop scientific health communication interventional strategies to improve health literacy on non-communicable disease prevention and care. OBJECTIVE To explore barriers of health literacy on non-communicable disease prevention and care among patients in north wollo zone public Hospitals, Northeast Ethiopia, 2023. METHOD In this study phenomenological study design was conducted from February 5 to 30/2023.We have used purposive sampling technique to select study participants from chronic follow up clinics. Data were collected using in-depth interview and focused group discussion in which audio was recorded, transcribed verbatim and translated to English. Thematic analysis was performed with atlas ti. 7 software. RESULT In this study four main themes with seven subthemes were developed. The main themes were factors at the organizational, community, interpersonal, and intra-personal factors. The poor knowledge, lack of enough money for transportation and medication at the hospital were identified as barrier to get early diagnosis and treatment. Some participants explored that they have no any support from family or others. The cultural norms like weeding and funeral ceremonies enforce patients to consume prohibited substances like alcohol and salty foods. CONCLUSION In this study different barriers of health literacy were explored. Lack of knowledge, economic problems, lack of social support, poor communication with health care providers, cultural influences, lack of regular health education, lack of access to health care services and poor infrastructure were main barriers of health literacy in patients with NCD. Therefore, we recommended all concerned bodies to work on social and behavioral change communication intervention focusing on awareness creation, supply of drugs and create supportive environment to get accessible and affordable health care service to decrease the impact of non-communicable disease at personal, community and national level.
Collapse
Affiliation(s)
- Eneyew Talie Fenta
- Department of Public Health, College of Medicine and Health Sciences, Injibara University, Injibara, Ethiopia.
| | - Atitegeb Abera Kidie
- Department of Public Health, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Misganaw Guadie Tiruneh
- Department of Health Systems and Policy, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tadele Fentabel Anagaw
- Department of Health Promotion and Behavioral Science, School of Public Health, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Eyob Ketema Bogale
- Department of Health Promotion and Behavioral Science, School of Public Health, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Amanuel Addisu Dessie
- Department of Public Health, College of Medicine and Health Sciences, Injibara University, Injibara, Ethiopia
| | - Nigus Kassie Worku
- Department of Public Health, College of Medicine and Health Science, Dire Dawa University, Dire Dawa, Ethiopia
| | - Mastewal Giza Amera
- Department of Public Health, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Hiwot Tesfa
- Department of Public Health, College of Medicine and Health Sciences, Injibara University, Injibara, Ethiopia
| | | | - Amare Mebrate Delie
- Department of Public Health, College of Medicine and Health Sciences, Injibara University, Injibara, Ethiopia
| | - Birtukan Gizachew Ayal
- Department of Public Health, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| |
Collapse
|
11
|
Casanova AG, Morales AI, Vicente-Vicente L, López-Hernández FJ. Effect of uric acid reduction on chronic kidney disease. Systematic review and meta-analysis. Front Pharmacol 2024; 15:1373258. [PMID: 38601468 PMCID: PMC11005459 DOI: 10.3389/fphar.2024.1373258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Accumulating evidence suggests that hyperuricemia is a pathological factor in the development and progression of chronic kidney disease. However, the potential benefit afforded by the control of uric acid (UA) is controversial. Individual studies show discrepant results, and most existing meta-analysis, especially those including the larger number of studies, lack a placebo or control group as they aim to compare efficacy between drugs. On these grounds, we performed a me-ta-analysis restricted to studies including the action of any anti-gout therapies referenced to a control or placebo arm. This approach allows for a clearer association between UA reduction and renal effect. Of the twenty-nine papers included, most used allopurinol and febuxostat and, therefore, solid conclusions could only be obtained for these drugs. Both were very effective in reducing UA, but only allopurinol was able to significantly improve glomerular filtration rate (GFR), although not in a dose-dependent manner. These results raised doubts as to whether it is the hypouricemic effect of anti-gout drugs, or a pleiotropic effect, what provides protection of kidney function. Accordingly, in a correlation study that we next performed between UA reduction and GFR improvement, no association was found, which suggests that additional mechanisms may be involved. Of note, most trials show large inter-individual response variability, probably because they included patients with heterogeneous phenotypes and pathological characteristics, including different stages of CKD and comorbidities. This highlights the need to sub classify the effect of UA-lowering therapies according to the pathological scenario, in order to identify those CKD patients that may benefit most from them. Systematic Review Registration: CRD42022306646 https://www.crd.york.ac.uk/prospero/.
Collapse
Affiliation(s)
- Alfredo G. Casanova
- Toxicology Unit, Universidad de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL) del Instituto de Ciencias de la Salud de Castilla y León (ICSCYL), Salamanca, Spain
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain
- National Network for Kidney Research REDINREN, RICORS2040 RD21/0005/0004-Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I. Morales
- Toxicology Unit, Universidad de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL) del Instituto de Ciencias de la Salud de Castilla y León (ICSCYL), Salamanca, Spain
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain
- National Network for Kidney Research REDINREN, RICORS2040 RD21/0005/0004-Instituto de Salud Carlos III, Madrid, Spain
- Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| | - Laura Vicente-Vicente
- Toxicology Unit, Universidad de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL) del Instituto de Ciencias de la Salud de Castilla y León (ICSCYL), Salamanca, Spain
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain
- National Network for Kidney Research REDINREN, RICORS2040 RD21/0005/0004-Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J. López-Hernández
- Instituto de Investigación Biomédica de Salamanca (IBSAL) del Instituto de Ciencias de la Salud de Castilla y León (ICSCYL), Salamanca, Spain
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain
- National Network for Kidney Research REDINREN, RICORS2040 RD21/0005/0004-Instituto de Salud Carlos III, Madrid, Spain
- Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| |
Collapse
|
12
|
Asahina Y, Sakaguchi Y, Oka T, Hattori K, Kawaoka T, Doi Y, Yamamoto R, Matsui I, Mizui M, Kaimori JY, Isaka Y. Association between urinary uric acid excretion and kidney outcome in patients with CKD. Sci Rep 2024; 14:5119. [PMID: 38429496 PMCID: PMC10907602 DOI: 10.1038/s41598-024-55809-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/28/2024] [Indexed: 03/03/2024] Open
Abstract
Inhibiting tubular urate reabsorption may protect the kidney from urate-induced tubular injury. However, this approach may promote intratubular uric acid crystallization, especially in acidified urine, which could be toxic to the kidney. To assess how tubular urate handling affects kidney outcomes, we conducted a retrospective cohort study including 1042 patients with estimated glomerular filtration rates (eGFR) of 15-60 mL/min/1.73 m2. The exposures were fractional excretion of uric acid (FEUA) and urinary uric acid-to-creatinine ratio (UUCR). The kidney outcome was defined as a halving of eGFR from baseline or initiating kidney replacement therapy. The median FEUA and UUCR were 7.2% and 0.33 g/gCre, respectively. During a median follow-up of 1.9 years, 314 kidney outcomes occurred. In a multivariate Cox model, the lowest FEUA quartile exhibited a 1.68-fold higher rate of kidney outcome than the highest FEUA quartile (95% confidence interval, 1.13-2.50; P = 0.01). Similarly, lower UUCR was associated with a higher rate of kidney outcome. Notably, patients in the highest quartile of FEUA and UUCR were at the lowest risk of kidney outcome even among those with aciduria. In conclusion, lower FEUA and UUCR were associated with a higher risk of kidney failure, suggesting that increased urate reabsorption is harmful to the kidney.
Collapse
Affiliation(s)
- Yuta Asahina
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koki Hattori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takayuki Kawaoka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yohei Doi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryohei Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun-Ya Kaimori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
13
|
Yip K, Braverman G, Yue L, Fields T. Pipeline Therapies for Gout. Curr Rheumatol Rep 2024; 26:69-80. [PMID: 38133712 DOI: 10.1007/s11926-023-01128-3] [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] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE OF REVIEW Despite effective available treatments, gout management is often unsuccessful in getting patients to target serum urate goal and in managing flares in the setting of comorbidities. Studies addressing future treatment options for short- and long-term management are reviewed. RECENT FINDINGS URAT-1 blocking agents have been helpful but have had limitations related to effects on renal function, lack of efficacy with renal impairment, and potential to increase renal stones. Dotinurad may function in the setting of decreased renal function. Arhalofenate has anti-URAT-1 activity and may also blunt gout flares. A new xanthine oxidase inhibitor (XOI), tigulixostat, is under study. New uricase treatments manufactured in combination with agents that can reduce immunogenicity may make uricase treatment simpler. A unique strategy of inhibiting gut uricase may offer the benefits of avoiding systemic absorption. For gout flares, IL-1β inhibitor studies in progress include different dosing schedules. Dapansutrile, an oral agent under investigation, inhibits activation of the NLRP3 inflammasome and may be an effective anti-inflammatory. New treatments for gout that are under study may work in the setting of comorbidities, simplify management, utilize new mechanisms, or have reduced side effects.
Collapse
Affiliation(s)
- Kevin Yip
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA.
| | - Genna Braverman
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Linda Yue
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Theodore Fields
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
14
|
Kurihara O, Yamada T, Kato K, Miyauchi Y. Efficacy of dotinurad in patients with severe renal dysfunction. Clin Exp Nephrol 2024; 28:208-216. [PMID: 37864678 DOI: 10.1007/s10157-023-02419-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Although hyperuricemia is associated with the progression of chronic kidney disease (CKD), a reduction in CKD progression by uric acid (UA)-lowering therapy has been controversial. Recently, dotinurad, a uricosuric drug with selective urate reabsorption inhibitory properties, has been developed. However, its efficacy in lowering serum UA levels and its effects on renal function in patients with severe renal dysfunction are unclear. Thus, this study aimed to determine the effects of dotinurad on renal function in patients with severe renal dysfunction. METHODS Data from 53 outpatients with hyperuricemia who newly received dotinurad between December 2020 and October 2022 were retrospectively analyzed. The mean baseline estimated glomerular filtration rate (eGFR) was 38.7 ± 17.0 mL/min/1.73 m2. The patients were divided into three groups based on their baseline eGFR: eGFR < 30 (n = 17), 30 ≤ eGFR < 45 (n = 17), and eGFR ≥ 45 (n = 19). RESULTS The mean follow-up period was 9.8 ± 4.5 (range, 3-21) months. Serum UA levels significantly decreased in all groups. Although eGFR did not significantly change in patients with 30 ≤ eGFR < 45 and eGFR ≥ 45 (P = 0.918 and P = 0.535, respectively), it improved significantly in patients with eGFR < 30 (P = 0.032). The proportion of patients with improved eGFR was significantly higher in patients with eGFR < 30 (P = 0.038) than in patients with 30 ≤ eGFR < 45 and eGFR ≥ 45. In the multivariate logistic regression analysis, baseline eGFR < 30 and achieving a serum UA level of ≤ 6.0 mg/dL were significantly associated with improved eGFR (P = 0.033 and P = 0.015, respectively). CONCLUSIONS Dotinurad may have UA-lowering effects and the potential to improve kidney function in patients with severe renal dysfunction.
Collapse
Affiliation(s)
- Osamu Kurihara
- Cardiovascular Center, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamakari, Inzai, Chiba, 270-1694, Japan.
| | - Takehisa Yamada
- Department of Nephrology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba, Japan
| | - Katsuhito Kato
- Department of Hygiene and Public Health, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yasushi Miyauchi
- Cardiovascular Center, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamakari, Inzai, Chiba, 270-1694, Japan
| |
Collapse
|
15
|
Moszczuk B, Życińska K, Mucha K. Asymptomatic Hyperuricemia: A Nephro-Rheumatological Perspective. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0024. [PMID: 39612508 DOI: 10.2478/aite-2024-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/18/2024] [Indexed: 12/01/2024]
Abstract
Hyperuricemia (HU) is a common disorder associated with gout, kidney injury, and high cardiovascular risk. However, whether high serum uric acid (sUA) is a causative factor or just comorbidity remains unclear. When asked if asymptomatic hyperuricemic patients need treatment, even artificial intelligence in the form of the GPT chat provides an ambivalent answer and refers us to a healthcare provider. We believe that such discrepancies stem from an incomplete understanding of the role that uric acid (UA) plays inside and outside the cell. With the rapid development of genomics, proteomics, immunology, and novel biomarkers, we are armed with new data to help us better understand the weight of inborn and environmental factors on an individual's UA concentrations. This review sums up the latest progress that has been made in the field of asymptomatic HU, compares the results presented by various research teams, and indicates new directions that emerge for future studies.
Collapse
Affiliation(s)
- Barbara Moszczuk
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Życińska
- Department of Rheumatology, Systemic Connective Tissue Diseases and Rare Diseases, Central Clinical Hospital MSWiA in Warsaw, Warsaw, Poland
- Department of Family Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Mucha
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
16
|
Wang Y, Lu J. The Management of Diabetes with Hyperuricemia: Can We Hit Two Birds with One Stone? J Inflamm Res 2023; 16:6431-6441. [PMID: 38161355 PMCID: PMC10757772 DOI: 10.2147/jir.s433438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Serum urate (SU) is an independent predictor for the incidence of diabetes. In current diabetes treatment regimens, there is insufficient appreciation of the importance of hyperuricemia (HU) in disease control and prevention. To summarize the updated knowledge on the effects of SU on β-cell function, insulin resistance and chronic diabetic complications, as well as to evaluate the management of patients with both HU and diabetes, we searched the MEDLINE PubMed database, and included 285 journal articles. An inverted U-shaped relationship between fasting plasma glucose and SU levels was established in this review. Elevated SU levels may enhance the development of chronic diabetic complications, including macrovascular and microvascular dysfunction. Diet and exercise are essential parts of the lifestyle changes necessary for HU and diabetes management. Glucose- and urate-lowering drug selection and combination should be made with the principle of ameliorating, and at least not deteriorating, diabetes and HU. Medical artificial intelligence technology and monitoring systems can help to improve the effectiveness of long-term management of HU and diabetes through digital healthcare. This study comprehensively reviews and provides a scientific and reliable basis for and viewpoints on the clinical management of diabetes and HU.
Collapse
Affiliation(s)
- Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jie Lu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
17
|
Qu Y, Yu Y, Pan J, Li H, Cui C, Liu D. Systematic review and model-based analysis to identify whether renal safety risks of URAT1 inhibitors are fully determined by uric acid-lowering efficacies. Semin Arthritis Rheum 2023; 63:152279. [PMID: 37866004 DOI: 10.1016/j.semarthrit.2023.152279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVE Renal safety risk is currently an important factor that hinders the development of uric acid transporter 1 (URAT1) inhibitors. This study aimed to compare the renal safety and uric acid-lowering efficacy of different URAT1 inhibitors and clarify the association between them. METHODS A systematic review of published randomized controlled trials on URAT1 inhibitors was conducted to investigate the incidence of renal safety events. A model-based analysis was performed to predict the uric acid-lowering efficacy of representative URAT1 inhibitors. RESULTS The overall renal safety event incidences of lesinurad, verinurad, dotinurad, SHR4640, and benzbromarone in patients with hyperuricemia were 11.2 % (142/1264), 12.0 % (34/284), 0.5 % (2/421), 2.3 % (5/213), and 1.3 % (5/393), respectively. A semi-mechanistic pharmacokinetic/pharmacodynamic model was used to establish the dose-exposure-effect relationship of lesinurad, verinurad, dotinurad, and SHR4640 with or without the combination of xanthine oxidase inhibitors (XOIs). The efficacy ranking of the intermediate dose of URAT1 inhibitors with once-daily dosing was 2 mg dotinurad > 10 mg verinurad > 5 mg SHR4640 > 400 mg lesinurad. The combination of 80 mg febuxostat and 600 mg allopurinol reduced the 24-h cumulative renal uric acid excretion by 48.4 % and 48.3 %, respectively. CONCLUSION Uric acid-lowering efficacy is not an independent factor for the renal safety risk of different URAT1 inhibitors, and structural differences could be responsible for the difference. The adverse renal effects of URAT1 inhibitors are dose-dependent, and the combination with high doses of XOIs can significantly reduce the renal safety risk by reducing uric acid excretion by the kidneys.
Collapse
Affiliation(s)
- Yuchen Qu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunli Yu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiyan Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China; Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China; Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China; Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China; Department of Cardiology, Peking University Third Hospital, Beijing, China.
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China; Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
18
|
Singh A, Singh K, Sharma A, Kaur K, Chadha R, Singh Bedi PM. Past, present and future of xanthine oxidase inhibitors: design strategies, structural and pharmacological insights, patents and clinical trials. RSC Med Chem 2023; 14:2155-2191. [PMID: 37974965 PMCID: PMC10650961 DOI: 10.1039/d3md00316g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/06/2023] [Indexed: 11/19/2023] Open
Abstract
Xanthine oxidase, a molybdo-flavoenzyme, and an isoform of xanthine dehydrogenase both exist as xanthine oxidoreductase and are responsible for purine catabolism. Xanthine oxidase is more involved in pathological conditions when extensively modulated. Elevation of xanthine oxidase is not only the prime cause of gout but is also responsible for various hyperuricemia associated pathological conditions like diabetes, chronic wounds, cardiovascular disorders, Alzheimer's disease, etc. Currently available xanthine oxidase inhibitors in clinical practice (allopurinol, febuxostat and topiroxostat) suffer from fatal side effects that pose a serious problem to the healthcare system, raising global emergency to develop novel, potent and safer xanthine oxidase inhibitors. This review will provide key and systematic information about: a. design strategies (inspired from both marketed drugs in clinical practice and natural products), structural insights and pharmacological output (xanthine oxidase inhibition and associated activities) of various pre-clinical candidates reported by various research groups across the globe in the past two decades; b. patented xanthine oxidase inhibitors published in the last three decades and c. clinical trials and their outcomes on approved drug candidates. Information generated in this review has suggested fragment-based drug design (FBDD) and molecular hybridization techniques to be most suitable for development of desired xanthine oxidase inhibitors as one provides high selectivity toward the enzyme and the other imparts multifunctional properties to the structure and both may possess capabilities to surpass the limitations of currently available clinical drugs. All in combination will exclusively update researchers working on xanthine oxidase inhibitors and allied areas and potentially help in designing rational, novel, potent and safer xanthine oxidase inhibitors that can effectively tackle xanthine oxidase related disease conditions and disorders.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Renu Chadha
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh 160014 India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
- Drug and Pollution Testing Laboratory, Guru Nanak Dev University Amritsar Punjab 143005 India
| |
Collapse
|
19
|
Terkeltaub R. Emerging Urate-Lowering Drugs and Pharmacologic Treatment Strategies for Gout: A Narrative Review. Drugs 2023; 83:1501-1521. [PMID: 37819612 DOI: 10.1007/s40265-023-01944-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/13/2023]
Abstract
Hyperuricemia with consequent monosodium urate crystal deposition leads to gout, characterized by painful, incapacitating inflammatory arthritis flares that are also associated with increased cardiovascular event and related mortality risk. This narrative review focuses on emerging pharmacologic urate-lowering treatment (ULT) and management strategies in gout. Undertreated, gout can progress to palpable tophi and joint damage. In oral ULT clinical trials, target serum urate of < 6.0 mg/dL can be achieved in ~ 80-90% of subjects, with flare burden reduction by 1-2 years. However, real-world ULT results are far less successful, due to both singular patient nonadherence and prescriber undertreatment, particularly in primary care, where most patients are managed. Multiple dose titrations commonly needed to optimize first-line allopurinol ULT monotherapy, and substantial potential toxicities and other limitations of approved, marketed oral monotherapy ULT drugs, promote hyperuricemia undertreatment. Common gout comorbidities with associated increased mortality (e.g., moderate-severe chronic kidney disease [CKD], type 2 diabetes, hypertension, atherosclerosis, heart failure) heighten ULT treatment complexity and emphasize unmet needs for better and more rapid clinically significant outcomes, including attenuated gout flare burden. The gout drug armamentarium will be expanded by integrating sodium-glucose cotransporter-2 (SGLT2) inhibitors with uricosuric and anti-inflammatory properties as well as clinically indicated antidiabetic, nephroprotective, and/or cardioprotective effects. The broad ULT developmental pipeline is loaded with multiple uricosurics that selectively target uric acid transporter 1 (URAT1). Evolving ULT approaches include administering selected gut anaerobic purine degrading bacteria (PDB), modulating intestinal urate transport, and employing liver-targeted xanthine oxidoreductase mRNA knockdown. Last, emerging measures to decrease the immunogenicity of systemically administered recombinant uricases should simplify treatment regimens and further improve outcomes in managing the most severe gout phenotypes.
Collapse
Affiliation(s)
- Robert Terkeltaub
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
21
|
Hao SH, Ye LY, Yang C. The landscape of pathophysiology guided therapeutic strategies for gout treatment. Expert Opin Pharmacother 2023; 24:1993-2003. [PMID: 38037803 DOI: 10.1080/14656566.2023.2291073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023]
Abstract
INTRODUCTION Gout is a common autoinflammatory disease caused by hyperuricemia with acute and/or chronic inflammation as well as tissue damage. Currently, urate-lowering therapy (ULT) and anti-inflammatory therapy are used as first-line strategies for gout treatment. However, traditional drugs for gout treatment exhibit some unexpected side effects and are not suitable for certain patients due to their comorbidity with other chronic disease. AREAS COVERED In this review, we described the pathophysiology of hyperuricemia and monosodium urate (MSU) crystal induced inflammatory response during gout development in depth and comprehensively summarized the advances in the investigation of promising ULT drugs as well as anti-inflammatory drugs that might be safer and more effective for gout treatment. EXPERT OPINION New drugs that are developed based on these molecular mechanisms exhibited great efficacy on reduction of disease burden both in vitro and in vivo, implying their potential for clinical application. Moreover, hyperthermia also showed regulation effect on MSU crystals formation and the signaling pathways involved in inflammation.
Collapse
Affiliation(s)
- Sai Heng Hao
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lin Yan Ye
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Rossing P, Frimodt-Møller M, Persson F. Precision Medicine and/or Biomarker Based Therapy in T2DM: Ready for Prime Time? Semin Nephrol 2023; 43:151430. [PMID: 37862744 DOI: 10.1016/j.semnephrol.2023.151430] [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: 10/22/2023]
Abstract
Approximately 30-40% of people with type 2 diabetes mellitus develop chronic kidney disease. This is characterised by elevated blood pressure, declining kidney function and enhanced cardiovascular morbidity and mortality. Increased albuminuria and decreasing estimated glomerular function has to be evaluated regularly to diagsnose kidney disease. New biomarkers may facilitate early diagnosis and provide infomation on undlying pathology thereby supporting early precision intervention for the optimal benefit. A number of biomarkers have been suggested but are not yet implemented in clinical practice. iI the future such bimarkers may pave the way for personalized treatment.
Collapse
Affiliation(s)
- Peter Rossing
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | | - Frederik Persson
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|
23
|
Zhang Y, Song R, Hua Y, Su X, Wang L. Cardiovascular and kidney outcomes of uric acid-lowering therapy in patients with different kidney functions: study protocol for a systematic review, pairwise and network meta-analysis. BMJ Open 2023; 13:e059096. [PMID: 36754561 PMCID: PMC9923315 DOI: 10.1136/bmjopen-2021-059096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/29/2022] [Indexed: 02/10/2023] Open
Abstract
INTRODUCTION Hyperuricaemia has been implicated in the development of kidney function in populations with chronic kidney disease; however, the benefits of urate-lowering therapy (ULT) remain uncertain in different clinical studies. The different kidney functions of enrolled populations and distinct pharmacokinetic characteristics of ULT might be of the essence for the contrasting results. In this study, we will synthesise all available data from randomised controlled trials (RCTs) and cohort studies, then evaluate the outcomes of ULT in patients stratified by different estimated glomerular filtration rate (eGFR) stratifications. Furthermore, we will attempt to explore a relatively optimal ULT regimen using a Bayesian network meta-analysis in different eGFRs. METHODS AND ANALYSIS We searched published and unpublished data from MEDLINE, EMBASE, the Cochrane Central Register of Controlled trials and ClinicalTrials.gov website (before March 2022) for RCTs and cohort studies without language restriction. In the pairwise meta-analysis, all regimens of ULT will be pooled as a whole and compared with controls in different eGFRs. The random-effects model will be applied to generate the summary values using the software Stata V.12.0 (StataCorp). Network meta-analysis within a Bayesian framework will be conducted to explore the relative efficacy profiles of different ULTs and to find optimal ULT in different eGFRs. The software of WinBUGS V.1.4.3 and R2WinBUGS package of R V.3.1.1 will be used in the network meta-analysis. Primary outcomes will be the occurrence of major cardiovascular events and kidney failure events. Secondary outcomes will include the rate of change in eGFR per year, all-cause death, changes in serum uric acid level and major adverse events. Two authors will independently review study selection, data extraction and quality assessment. ETHICS AND DISSEMINATION The meta-analysis does not require ethical certification. The results will be disseminated through publication in a peer-reviewed journal and through presentations at academic conferences. PROSPERO REGISTRATION NUMBER CRD42021226163.
Collapse
Affiliation(s)
- Yaqing Zhang
- Renal Division, Shanxi Medical University Second Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| | - Runxia Song
- Renal Division, Shanxi Medical University Second Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| | - Ying Hua
- Renal Division, Shanxi Medical University Second Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| | - Xiaole Su
- Renal Division, Shanxi Medical University Second Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| | - Lihua Wang
- Renal Division, Shanxi Medical University Second Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| |
Collapse
|
24
|
Yang Y, Hu Y, Yao F, Yang J, Ge L, Wang P, Xu X. Virtual screening and activity evaluation of human uric acid transporter 1 (hURAT1) inhibitors. RSC Adv 2023; 13:3474-3486. [PMID: 36756549 PMCID: PMC9871872 DOI: 10.1039/d2ra07193b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/13/2022] [Indexed: 01/25/2023] Open
Abstract
Hyperuricemia is a disease caused by disorder of purine metabolism, mainly due to insufficient renal excretion of uric acid. Urate transporter 1 (URAT1) is the most widely studied target of urate transporters, and used for uric acid (UA) reabsorption. This study used the AlphaFold2 algorithm to predict the structure of URAT1. Virtual screening and biological evaluation were used to discover novel URAT1 inhibitors that target the critical amino acids. Seven compounds were screened from the T2220 database and validated as URAT1 inhibitors by cell biology experiments. The IC50 values of benbromarone, NP023335, TN1148, and TN1008 were 6.878, 18.46, 24.64, and 53.04 μM, respectively. Molecular dynamics simulation was used to investigate the binding mechanism of URAT1 to NP023335, which forms stable contact with Ser35, Phe365, and Arg477. These interactions are essential for maintaining the biological activity of NP023335. The three compounds' pharmacokinetic characteristics were predicted, and NP023335's properties matched those of an empirical medication with the benefits of high solubility, low cardiotoxicity, good membrane permeability, and oral absorption. The natural product NP023335 will serve as a promising hit compound for facilitating the further design of novel URAT1 inhibitors.
Collapse
Affiliation(s)
- Yacong Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| | - Yu Hu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| | - Fengli Yao
- College of Food Science and Engineering, Ocean University of China Qingdao 266071 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
- School of Life Science, Lanzhou University Lanzhou 730000 China
| | - Leilei Ge
- Qingdao Vland Biotech Group Co., Ltd 266102 China
| | - Peng Wang
- College of Food Science and Engineering, Ocean University of China Qingdao 266071 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
| | - Ximing Xu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| |
Collapse
|
25
|
Bignardi PR, Ido DH, Garcia FAL, Braga LM, Delfino VDA. Does uric acid-lowering treatment slow the progression of chronic kidney disease? A meta-analysis of randomized controlled trials. Nefrologia 2022:S2013-2514(22)00177-8. [PMID: 36564223 DOI: 10.1016/j.nefroe.2022.04.005] [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: 11/05/2021] [Accepted: 04/02/2022] [Indexed: 06/17/2023] Open
Abstract
INTRODUCTION Hyperuricemia has been proposed as an independent factor in the development and progression of chronic kidney disease (CKD). However, the effect of uric acid-lowering therapies on delaying CKD progression is still uncertain. Therefore, this systemic review aims to assess the effect of uric acid-lowering therapies on renal outcomes in pre-dialysis CKD patients. METHODS PubMed, Cochrane Library, and Lilacs databases were searched until April 24, 2021, for randomized clinical trials of CKD patients on uric acid-lowering treatment with xanthine-oxidase (XO) inhibitors. The weighted mean difference (WMD) or standard mean difference (SMD) with confidence interval (CI) were pooled using a random-effects model. RESULTS Among 567 studies found, eighteen met the inclusion criteria (n=2463 participants). Compared to the patient's control group, the WMD for the glomerular filtration ratio (GFR) and serum creatinine changes of the treated group was 2.02ml/min/1.73m2 (95%CI 0.41 to 3.63, P=0.014) and -0.19mg/dl (95%CI -0.34 to -0.04, I2=86.2%, P=0.011), respectively. Subgroup analyses showed that the difference in follow-up time and CKD population type in the studies may explain the controversy about the role of uric acid-lowering therapies in CKD progression. The GFR and creatinine outcomes analysis by types of XO inhibitors showed no difference between the control and treated groups. Uric acid-lowering therapies were strongly associated with decreased serum uric acid and urinary protein-creatinine ratio and urinary albumin-creatinine ratio. CONCLUSIONS These findings suggest that uric acid-lowering treatment may slow CKD progress and reduce protein and albumin excretion. However, larger and properly powered randomized clinical trials with specific CKD populations are needed to confirm these findings.
Collapse
Affiliation(s)
| | - Danielle Harumi Ido
- School of Medicine, Pontifícia Universidade Católica do Paraná, Londrina, Brazil
| | | | - Lucas Mendes Braga
- School of Medicine, Pontifícia Universidade Católica do Paraná, Londrina, Brazil
| | - Vinicius Daher Alvares Delfino
- School of Medicine, Pontifícia Universidade Católica do Paraná, Londrina, Brazil; Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
26
|
Considerations for Choosing First-Line Urate-Lowering Treatment in Older Patients with Comorbid Conditions. Drugs Aging 2022; 39:923-933. [PMID: 36437395 DOI: 10.1007/s40266-022-00986-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/29/2022]
Abstract
Gout is the most common inflammatory arthritis in adults. The prevalence of gout increases with age. Urate-lowering treatment (ULT) among older patients is often challenging in that patients frequently suffer insufficient effectiveness or adverse events due to comorbidities, concurrent medications, and altered pharmacokinetics. The large-scale randomized controlled trials (RCTs) directly investigating gout patients regarding cardiovascular (CV) safety have only recently been introduced; CARES and FAST compared the CV safety of the two xanthine oxidase inhibitors (XOis), febuxostat versus allopurinol, in patients with gout. Based on the CARES trial that showed CV concerns with febuxostat, the current international guidelines recommend allopurinol as first-line ULT in gout, while preserving other agents as a second-line treatment, despite a higher potency of febuxostat. XOis would be more suitable than uricosurics to treat older patients with gout due to the high prevalence of chronic kidney disease (CKD) in older patients. However, allopurinol alone might not achieve the target serum uric acid levels below 6 mg/dL and CKD might confer an increased risk of allopurinol induced cutaneous adverse reactions in older patients. Furthermore, as well as the later analysis of CARES participants who were lost to follow-up, data from the FAST trial and real-world studies suggest non-inferior CV safety for febuxostat compared to allopurinol even in the presence of CV diseases. Thus, febuxostat use in older patients with renal impairment may be more positively considered. The combination therapy of a novel uricosuric, verinurad, plus febuxostat reduced albuminuria in hyperuricemic patients with type 2 diabetes and CKD in a phase 2a trial, and further RCTs are awaited. Finally, the sodium-glucose cotransporter-2 inhibitor class of oral hypoglycemic agents, known to exert beneficial CV and renal effects independent of glycemic control, have shown a uricosuric effect and could be used as adjunctive therapy in older patients with cardiorenal comorbidities.
Collapse
|
27
|
Stanley IK, Phoon RK, Toussaint ND, Cullen V, Kearns J, Dalbeth N, Johnson DW, Krishnasamy R, Tunnicliffe DJ, CARI Guidelines Steering Committee 13. Caring for Australians and New Zealanders With Kidney Impairment Guidelines: Rapid Development of Urate Lowering Therapy Guidelines for People With CKD. Kidney Int Rep 2022; 7:2563-2574. [PMID: 36506231 PMCID: PMC9727528 DOI: 10.1016/j.ekir.2022.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction The slow transformation of new research findings into clinical guidelines is a barrier to providing evidence-based care. The Caring for Australians and New Zealanders with Kidney Impairment (CARI) guidelines are developing models to improve guideline production, one methodology involves more functional concordance between trial groups, such as the Australian Kidney Trials Network (AKTN) and CARI. The objective of this project was to rapidly produce an evidence-based guideline on urate-lowering therapy in patients with chronic kidney disease (CKD), in response to new clinical trial publications on the topic by the AKTN. Methods To produce a guideline as rapidly as possible, an existing systematic review was utilized as the evidence base, and then updated with the inclusion of clinical trials that had been published subsequently. A Work Group was convened to review the evidence and compose an appropriate guideline using CARI/GRADE methodology. The group met 3 times over 45 days to formulate the guideline. Results The result was a strong recommendation against the use urate-lowering therapies in individuals with CKD (not receiving dialysis) and asymptomatic hyperuricemia. The process of identifying an appropriate existing systematic review, updating the literature search, and synthesizing the evidence, was done by 2 individuals over 15 days. The Work Group was formulated and composed the guideline over 45 days. In all, a new guideline incorporating the most up-to-date evidence was formulated in 60 days. Conclusion This method of guideline development represents a potentially new way of releasing guidelines that encapsulates all available evidence in a time-efficient manner.
Collapse
Affiliation(s)
- Isabelle Kitty Stanley
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Queensland, Australia
- Center for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard K.S. Phoon
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Renal Medicine, Center for Transplant and Renal Research, Westmead Hospital, Sydney, Australia
| | - Nigel D. Toussaint
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Vanessa Cullen
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Queensland, Australia
| | - John Kearns
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicola Dalbeth
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - David W. Johnson
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Rathika Krishnasamy
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
- Department of Nephrology, Sunshine Coast University Hospital, Birtinya, Australia
| | - David J. Tunnicliffe
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children’s Hospital of Westmead, Sydney, Australia
| | - CARI Guidelines Steering Committee13
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Queensland, Australia
- Center for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Renal Medicine, Center for Transplant and Renal Research, Westmead Hospital, Sydney, Australia
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Department of Nephrology, Sunshine Coast University Hospital, Birtinya, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children’s Hospital of Westmead, Sydney, Australia
| |
Collapse
|
28
|
Siddiq MAB, Jansen TL, Rasker JJ. What is the Place for Uricosuric Agents in Gout Management? Curr Rheumatol Rev 2022; 18:279-285. [PMID: 35260069 DOI: 10.2174/1573397118666220308160124] [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: 08/31/2021] [Revised: 12/20/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Md Abu Bakar Siddiq
- Department of Physical Medicine and Rheumatology, Brahmanbaria Medical College, Brahmanbaria, Bangladesh.,School of Health Sport and Professional Practice, University of South Wales, Pontypridd, United Kingdom
| | - Tim L Jansen
- Department of Rheumatology, Viecuri MC, Venlo, The Netherlands
| | - Johannes J Rasker
- Faculty of Behavioral, Management and Social Sciences, Department Psychology, Health and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
29
|
Ma Y, Lin J, Xia P, Zheng H, Cheng X, Ji P, Wu W, Hou L, Wang L, Zhu G, Qiu L, Zheng Y, Chen L. Comparison between the profiles of patients defined by age-adapted and fixed threshold CKD criteria, a national-wide, cross-sectional study. Clin Kidney J 2022; 15:2312-2321. [PMID: 36381365 PMCID: PMC9664565 DOI: 10.1093/ckj/sfac188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Indexed: 12/05/2022] Open
Abstract
Background Kidney function declines naturally with advancing age. Therefore an age-adapted estimated glomerular filtration rate (eGFR) threshold has been proposed instead of the fixed threshold for CKD definition. This study aims to describe and compare the profile of CKD patients defined by these two criteria in a Chinese population. Method We recruited adult participants with selected biochemical tests from the Chinese Physiological Constant and Health Condition survey conducted from 2007 to 2011, with the GFR estimated by the Chronic Kidney Disease Epidemiology Collaboration formula. The age-adapted threshold of eGFR is 75, 60 and 45 ml/min/1.73 m2 for the population <40 years of age, 40–64 years and >64 years, respectively. The fixed threshold is 60 ml/min/1.73 m2 for all ages. Results Among the recruited 23 438 participants, 480 were diagnosed with CKD by fixed threshold criteria, while 391 were diagnosed with CKD by age-adapted criteria. Patients diagnosed by fixed threshold criteria were significantly older (66.4 versus 43.4 years; P < .001) and had a higher prevalence of all CVD risk factors compared with the non-CKD population. In contrast, age-adapted criteria defined a younger patient group and were not significantly associated with diabetes or obesity. When adjusted by age and gender, fixed threshold–defined CKD was not significantly associated with the number of coexisting CVD risk factors, while age-adapted-defined CKD was significantly associated. We also found that the CKD patients defined by age-adapted criteria matched well with the 2.5th percentile of eGFR in Chinese individuals. When compared with their age- and gender-matched controls, patients included by age-adapted criteria but excluded by fixed threshold criteria had a significantly higher prevalence of hypertension (23.2% versus 7.7%; P < .001) and hyperuricaemia (25.0% versus 5.5%; P < .001), while patients included only by the fixed threshold criteria were not significantly different in the prevalence of CVD risk factors and CKD-related disturbance except for hyperuricaemia (41.2% versus 14.0%; P < .001). Conclusion An age-adapted criterion is more closely associated with CVD risk factors and CKD-related diseases compared with fixed threshold criteria.
Collapse
Affiliation(s)
- Yixin Ma
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Jianfeng Lin
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Peng Xia
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Hua Zheng
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Xinqi Cheng
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Peili Ji
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Wei Wu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Lian Hou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Li Wang
- Department of Epidemiology and Biostatistics, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Guangjin Zhu
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| | - Yali Zheng
- Department of Nephrology, Affiliated Ningxia People's Hospital of Ningxia Medical University , Yinchuan , China
| | - Limeng Chen
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , China
| |
Collapse
|
30
|
Jenkins C, Hwang JH, Kopp JB, Winkler CA, Cho SK. Review of Urate-Lowering Therapeutics: From the Past to the Future. Front Pharmacol 2022; 13:925219. [PMID: 36081938 PMCID: PMC9445164 DOI: 10.3389/fphar.2022.925219] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
We reviewed all currently available ULT, as well as any medications in development using following databases: United States Food and Drug Administration (FDA), European Medicines Agency (EMA), Japanese Pharmaceutical and Medical Devices Agency (PMDA), and ClinicalTrials.gov. We identified a total of 36 drugs, including 10 approved drugs, 17 in clinical testing phases, and 9 in preclinical developmental phases. The 26 drugs currently undergoing testing and development include 5 xanthine oxidase inhibitors, 14 uricosurics, 6 recombinant uricases, and one with multiple urate-lowering mechanisms of action. Herein, we reviewed the benefit and risk of each drug summarizing currently available drugs. New trials of uricosuric agents are underway to develop the new indication. New drugs are going on to improve the potency of recombinant uricase and to develop the new route administration of such as oral formulation. This review will provide valuable information on the properties, indications, and limitations of ULTs.
Collapse
Affiliation(s)
- Christopher Jenkins
- Department of Internal Medicine, University of Connecticut Health Center, Farmington, CT, United States
- Department of Internal Medicine, The Hospital of Central Connecticut, New Britain, CT, United States
| | - Jennifer H. Hwang
- Department of Internal Medicine, University of Connecticut Health Center, Farmington, CT, United States
- Department of Internal Medicine, The Hospital of Central Connecticut, New Britain, CT, United States
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cheryl A. Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD, United States
| | - Sung Kweon Cho
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD, United States
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea
- *Correspondence: Sung Kweon Cho,
| |
Collapse
|
31
|
Shah N, Perkovic V, Kotwal S. Impact of SGLT2 inhibitors on the kidney in people with type 2 diabetes and severely increased albuminuria. Expert Rev Clin Pharmacol 2022; 15:827-842. [PMID: 35912871 DOI: 10.1080/17512433.2022.2108402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Diabetes is the most common cause of end stage kidney disease. Therapies such as sodium-glucose co-transporter-2 inhibitors have been identified over the last decade as effective oral hypoglycemic agents that also confer additional cardio and kidney protection. Knowledge of their mechanism of action and impact on patients with diabetes and albuminuria is vital in galvanizing prescriber confidence and increasing clinical uptake. AREAS COVERED This manuscript discusses the pathophysiology of diabetic kidney disease, patho-physiological mechanisms for sodium-glucose co-transporter-2 inhibitors, and their impact on patients with Type 2 diabetes mellitus and albuminuric kidney disease. EXPERT OPINION Sodium-glucose co-transporter-2 inhibitors reduce albuminuria with consequent benefits on cardiovascular and kidney outcomes in patients with diabetes and severe albuminuria. Whilst they have been incorporated into guidelines, the uptake of these agents into clinical practice has been slow. Increasing the uptake of these agents into clinical practice is necessary to improve outcomes for the large number of patients with diabetic kidney disease globally.
Collapse
Affiliation(s)
- Nasir Shah
- Faculty of Medicine, UNSW, Kensington, Sydney Australia 2052
| | - Vlado Perkovic
- Faculty of Medicine, UNSW, Kensington, Sydney Australia 2052.,The George Institute for Global Health, UNSW, 1 King Street, Newtown, Sydney, Australia 2042
| | - Sradha Kotwal
- The George Institute for Global Health, UNSW, 1 King Street, Newtown, Sydney, Australia 2042.,Prince of Wales Hospital, High Street, Sydney, Australia, 2031
| |
Collapse
|
32
|
Heerspink HJL, Stack AG, Terkeltaub R, Greene TA, Inker LA, Bjursell M, Perl S, Rikte T, Erlandsson F, Perkovic V. Rationale, design, demographics and baseline characteristics of the randomized, controlled, Phase 2b SAPPHIRE study of verinurad plus allopurinol in patients with chronic kidney disease and hyperuricaemia. Nephrol Dial Transplant 2022; 37:1461-1471. [PMID: 34383954 PMCID: PMC9317164 DOI: 10.1093/ndt/gfab237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Verinurad is a human uric acid (UA) transporter (URAT1) inhibitor known to decrease serum UA (sUA) levels and that may reduce albuminuria. In a Phase 2a study (NCT03118739), treatment with verinurad + febuxostat lowered urine albumin-to-creatinine ratio (UACR) at 12 weeks by 39% (90% confidence interval 4-62%) among patients with Type 2 diabetes mellitus, hyperuricaemia and albuminuria. The Phase 2b, randomized, placebo-controlled Study of verinurAd and alloPurinol in Patients with cHronic kIdney disease and hyperuRicaEmia (SAPPHIRE; NCT03990363) will examine the effect of verinurad + allopurinol on albuminuria and estimated glomerular filtration rate (eGFR) slope among patients with chronic kidney disease (CKD) and hyperuricaemia. METHODS Adults (≥18 years of age) with CKD, eGFR ≥25 mL/min/1.73 m2, UACR 30-5000 mg/g and sUA ≥6.0 mg/dL will be enrolled. Approximately 725 patients will be randomized 1:1:1:1:1 to 12, 7.5 or 3 mg verinurad + allopurinol, allopurinol or placebo. An 8-week dose-titration period will precede a 12-month treatment period; verinurad dose will be increased to 24 mg at Month 9 in a subset of patients in the 3 mg verinurad + allopurinol arm. The primary efficacy endpoint the is change from baseline in UACR at 6 months. Secondary efficacy endpoints include changes in UACR, eGFR and sUA from baseline at 6 and 12 months. CONCLUSIONS This study will assess the combined clinical effect of verinurad + allopurinol on kidney function in patients with CKD, hyperuricaemia and albuminuria, and whether this combination confers renoprotection beyond standard-of-care.
Collapse
Affiliation(s)
- Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- The George Institute for Global Health, Sydney, Australia
| | - Austin G Stack
- School of Medicine & Health Research Institute, University of Limerick, Limerick, Ireland
| | - Robert Terkeltaub
- San Francisco VA Health Care System, University of California San Diego, La Jolla, CA, USA
| | - Tom A Greene
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | | - Tord Rikte
- AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | | | - Vlado Perkovic
- The George Institute for Global Health, Sydney, Australia
| |
Collapse
|
33
|
Ji P, Zhu J, Feng J, Li H, Yu Q, Qin H, Wei L, Zhang J. Serum uric acid levels and diabetic kidney disease in patients with type 2 diabetes mellitus: A dose-response meta-analysis. Prim Care Diabetes 2022; 16:457-465. [PMID: 35305901 DOI: 10.1016/j.pcd.2022.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/26/2022] [Accepted: 03/06/2022] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Our study aimed to assess the existing evidence on whether serum uric acid (SUA) levels are associated with diabetic kidney disease (DKD) in patients with type 2 diabetes mellitus (T2DM). METHODS We conducted a systematic search of articles up to October 2021 in Medline, Embase, The Cochrane Library and Web of Science that estimated DKD by SUA levels in patients with T2DM. Pooled relative risks with 95% CI were calculated using random effects models RESULTS: A total of eight cohort studies involving 25,741 T2DM patients were included. Meta-analysis showed that compared the highest with the lowest category of SUA level, the summary risk ratios were 2.04 (95%CI 1.43-2.92, P < 0.001). The linear dose-response analysis revealed that the risk of DKD increased by 24% for each 1 mg/dl increase of SUA. The non-linear dose-response analysis also showed a significant relevance between SUA and the risk of DKD in patients with type 2 diabetes mellitus (P < 0.001). CONCLUSIONS Serum uric acid is associated with an increased risk of diabetic kidney disease in patients with type 2 diabetes mellitus. Serum uric acid level could be a good indicator for predicting diabetic kidney disease in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Pan Ji
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Jieyun Zhu
- International Medical Services, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China.
| | - Jihua Feng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Hongyuan Li
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Qiao Yu
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Han Qin
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Lile Wei
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| | - Jianfeng Zhang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China.
| |
Collapse
|
34
|
Najafi S, Bahrami M, Butler AE, Sahebkar A. Cardiovascular protection conferred by glucagon-like peptide-1 receptor agonists: A role for serum uric acid reduction? Br J Clin Pharmacol 2022; 88:4237-4238. [PMID: 35574902 DOI: 10.1111/bcp.15381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Sara Najafi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Bahrami
- Student Research Committee, Faculty of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain, Bahrain
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Bignardi PR, Ido DH, Garcia FAL, Braga LM, Delfino VDA. Does uric acid-lowering treatment slow the progression of chronic kidney disease? A meta-analysis of randomized controlled trials. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
36
|
Liang X, Liu X, Li D, Qin W, Liu Y. Effectiveness of Urate-Lowering Therapy for Renal Function in Patients With Chronic Kidney Disease: A Meta-Analysis of Randomized Clinical Trials. Front Pharmacol 2022; 13:798150. [PMID: 35370725 PMCID: PMC8968869 DOI: 10.3389/fphar.2022.798150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/24/2022] [Indexed: 02/05/2023] Open
Abstract
Background and Objective: Hyperuricemia is closely related to chronic kidney disease (CKD). The effects of urate-lowering therapy (ULT) on renal outcomes are uncertain, and whether it is warranted in CKD patients is currently unclear. The aim of our meta-analysis of randomized clinical trials (RCTs) was to assess the effectiveness and safety of ULT for improving kidney function in patients with CKD. Methods: RCTs were retrieved from the PubMed, Embase, MEDLINE and Cochrane Central Register of Controlled Trials databases. The meta-analysis was performed using Review Manager and Stata/SE software. The outcomes were changes in renal function and serum uric acid (SUA), serum creatinine, and adverse events. Results: Twelve RCTs with 1,469 participants were included in the meta-analysis. ULT was found to effectively lower SUA (standard mean difference (SMD): -2.70; 95% confidence interval (CI): -3.71, -1.69) but the renoprotective effects were not superior to those of control therapy (placebo or usual therapy), which were stable in the subgroup analyses and sensitivity analyses. Regarding adverse events, their risks did not increase in the ULT group compared with the control group and were stable in the sensitivity analyses. Conclusion: The findings of our meta-analysis suggested that ULT can effectively lower SUA, but there is insufficient evidence to support the renoprotective effects of ULT in CKD patients. In addition, ULT is safe for patients with CKD. Systematic Review Registration:https://clinicaltrials.gov/, identifier PROSPERO (CRD42020200550).
Collapse
Affiliation(s)
- Xiuping Liang
- Department of Rheumatology and Immunology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiang Liu
- Department of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Duohui Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wei Qin
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Najafi S, Bahrami M, Butler AE, Sahebkar A. The effect of Glucagon-like peptide-1 receptor agonists on serum uric acid concentration: A systematic review and meta-analysis. Br J Clin Pharmacol 2022; 88:3627-3637. [PMID: 35384008 DOI: 10.1111/bcp.15344] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/12/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Abstract
AIMS Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are a class of medications mainly used for the treatment of type 2 diabetes. They improve glucose tolerance, increase insulin secretion, and induce weight loss. There is controversy about the effect of GLP-1RAs on serum uric acid (SUA) concentration. Our systematic review aims to objectively answer whether GLP-1RAs affect SUA levels. METHODS We performed a systematic search on PubMed, Web of Science, Embase, Scopus, and Google Scholar datasets up to 27August,2021 with a language restriction of English only. Randomized controlled trials, observational studies, uncontrolled trials, and conference abstracts were included. Studies with insufficient data, irrelevant types of study, and follow-up duration of less than a month were excluded from the review. After critical appraisal by the Joanna Briggs Institute checklists, articles underwent data extraction using a pre-specified Microsoft Excel sheet. RESULTS Of 1004 identified studies, 17 were eligible for inclusion in this systematic review. Pre- to post-administration analysis of GLP-1RA effects on SUA demonstrated that GLP-1RAs could significantly reduce SUA concentration (difference in means=-0.341;SE=0.063;P-value<0.001). However, when compared to placebo, GLP-1 RAs did not perform any better in lowering SUA concentration (difference in means=-0.455;SE=0.259;P-value=0.079). Surprisingly, the active controls, that included insulin, metformin, sodium-glucose co-transporter 2 (SGLT-2) inhibitors, and dipeptidyl-peptidase 4 (DPP-4) inhibitors, did outperform GLP-1RAs in reducing SUA concentration (difference in means=0.250;SE=0.038;P-value<0.001). CONCLUSIONS Administration of GLP-1RAs can result in a significant reduction in SUA concentration. However, this reduction is less than that seen with the use of insulin, metformin, and SGLT-2 inhibitors.
Collapse
Affiliation(s)
- Sara Najafi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Bahrami
- Student Research Committee, Faculty of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Suijk D, van Baar M, van Bommel E, Iqbal Z, Krebber M, Vallon V, Touw D, Hoorn E, Nieuwdorp M, Kramer M, Joles J, Bjornstad P, van Raalte D. SGLT2 Inhibition and Uric Acid Excretion in Patients with Type 2 Diabetes and Normal Kidney Function. Clin J Am Soc Nephrol 2022; 17:663-671. [PMID: 35322793 PMCID: PMC9269569 DOI: 10.2215/cjn.11480821] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/17/2022] [Indexed: 11/23/2022]
Abstract
Background and objectives: Sodium glucose transporter 2 (SGLT2)-inhibitor-induced uric acid lowering may contribute to kidney protective effects of the drug-class in people with type 2 diabetes. This study investigates mechanisms of plasma uric acid lowering by SGLT2-inhibitors in people with type 2 diabetes with a focus on urate transporter (URAT)1. Methods: We conducted an analysis of two randomized, clinical trials. First, in the Renoprotective Effects of Dapagliflozin in Type 2 Diabetes (RED) study, 44 people with type 2 diabetes were randomized to dapagliflozin or gliclazide for 12 weeks. Plasma uric acid, fractional uric acid excretion and hemodynamic kidney function were measured in the fasted state and during clamped eu- or hyperglycemia. Second, in the Uric Acid Excretion study (UREX) study, 10 people with type 2 diabetes received 1-week empagliflozin, benzbromarone and their combination in a cross-over design and effects on plasma uric acid, fractional uric acid excretion and 24-hr uric acid excretion were measured. Results: In the RED study, compared to the fasted state (5.3±1.1mg/dL), acute hyperinsulinemia and hyperglycemia significantly reduced plasma uric acid by 0.2±0.3 and 0.4±0.3 mg/dL (both p<0.001), while increasing fractional uric acid excretion (by 3.2±3.1% and 8.9±4.5% respectively (both p<0.001). Dapagliflozin reduced plasma uric acid by 0.8±0.8mg/dL, 1.0±1.0mg/dL and by 0.8±0.7mg/dL during fasting, hyperinsulinemic-euglycemic and hyperglycemic conditions (p<0.001), whereas fractional uric acid excretion in 24-hr urine increased by 3.0±2.1% (p<0.001) and 2.6±4.5% during hyperinsulinemic-euglycemic conditions (p=0.003). Fractional uric acid excretion strongly correlated to fractional glucose excretion (r= 0.35, p=0.02). In the UREX study, empagliflozin and benzbromarone both significantly reduced plasma uric acid and increased fractional uric acid excretion. Effects of combination therapy did not differ from benzbromarone monotherapy. Conclusion: In conclusion, SGLT2-inhibitors induce uric acid excretion, which is strongly linked to urinary glucose excretion and which is attenuated during concomitant pharmacological blockade of URAT1.
Collapse
Affiliation(s)
- Danii Suijk
- D Suijk, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Michaël van Baar
- M van Baar, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Erik van Bommel
- E van Bommel, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Zainab Iqbal
- Z Iqbal, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Merle Krebber
- M Krebber, Department of Nephrology and Hypertension, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Volker Vallon
- V Vallon, Division of Nephrology and hypertension, Department of Medicine, University of California San Diego, La Jolla, United States
| | - Daan Touw
- D Touw, Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, Netherlands
| | - Ewout Hoorn
- E Hoorn, Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Max Nieuwdorp
- M Nieuwdorp, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Mark Kramer
- M Kramer, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| | - Jaap Joles
- J Joles, Department of Nephrology and Hypertension, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Petter Bjornstad
- P Bjornstad, Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, United States
| | - Daniël van Raalte
- D van Raalte, Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centres, Duivendrecht, Netherlands
| |
Collapse
|
39
|
Guedes M, Pecoits-Filho R. Can we cure diabetic kidney disease? Present and future perspectives from a nephrologist's point of view. J Intern Med 2022; 291:165-180. [PMID: 34914852 DOI: 10.1111/joim.13424] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease (CKD) worldwide, contributing to a great burden across a variety of patient-reported and clinical outcomes. New interventions for DKD management have been established in recent years, unleashing a novel paradigm, in which kidney-dedicated trials yield informative and robust data to guide optimal clinical management. After unprecedented results from groundbreaking randomized controlled trials were released, a new scenario of evidence-based recommendations has evolved for the management of diabetic patients with CKD. The current guidelines place great emphasis on multidimensional and interdisciplinary approaches, but the challenges of implementation are just starting and will be pivotal to optimize clinical results and to understand the new threshold for residual risk in DKD. We thereby provide an updated review on recent advances in DKD management based on new guideline recommendations, summarizing recent evidence while projecting the landscape for innovative ongoing initiatives in the field. Specifically, we review current insights on the natural history, epidemiology, pathogenesis, and therapeutics of DKD, mapping the new scientific information into the recently released Kidney Disease - Improving Global Outcomes Guidelines translating results from major novel randomized controlled trials to the clinical practice. Additionally, we approach the landscape of new therapeutics in the field, summarizing ongoing phase IIb and III trials focused on DKD. Finally, reflecting on the past and looking into the future, we highlight unmet needs in the current DKD management based on real-world evidence and offer a nephrologist's perspective into the challenge of fostering continuous improvement on clinical and patient-reported outcomes for individuals living with DKD.
Collapse
Affiliation(s)
- Murilo Guedes
- School of Medicine, Pontificia Universidade Catolica do Parana, Curitiba, Parana, Brazil
| | - Roberto Pecoits-Filho
- School of Medicine, Pontificia Universidade Catolica do Parana, Curitiba, Parana, Brazil.,DOPPS Program Area, Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Nishizawa H, Maeda N, Shimomura I. Impact of hyperuricemia on chronic kidney disease and atherosclerotic cardiovascular disease. Hypertens Res 2022; 45:635-640. [PMID: 35046512 DOI: 10.1038/s41440-021-00840-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
Hyperuricemia is caused by reduced renal/extrarenal excretion and overproduction of uric acid. It is affected by genetic predisposition related to uric acid transporters and by visceral fat accumulation due to overnutrition. The typical symptomatic complication of hyperuricemia is gout caused by monosodium urate crystals. Accumulated evidence from epidemiological studies suggests that hyperuricemia is also a risk factor for hypertension, chronic kidney disease (CKD) and atherosclerotic cardiovascular disease (CVD). However, it remains to be determined whether urate-lowering therapy for asymptomatic patients with hyperuricemia is effective in preventing CKD or CVD progression. This mini review focuses mainly on recent papers investigating the relationship between hyperuricemia and CKD or CVD and studies of urate-lowering therapy. Accumulated studies have proposed mechanisms of renal damage and atherosclerosis in hyperuricemia, including inflammasome activation, decreased nitric oxide bioavailability and oxidative stress induced by uric acid, urate crystals and xanthine oxidoreductase (XOR)-mediated reactive oxygen species. Since patients with hyperuricemia are a heterogeneous population with complex pathologies, it may be important to assess whether an outcome is the result of decreasing serum uric acid levels or an inhibitory effect on XOR. To clarify the impact of hyperuricemia on CKD and CVD progression, high-quality and detailed clinical and basic science studies of hyperuricemia and purine metabolism are needed.
Collapse
Affiliation(s)
- Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Norikazu Maeda
- Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
41
|
Guma M, Dadpey B, Coras R, Mikuls TR, Hamilton B, Quehenberger O, Thorisdottir H, Bittleman D, Lauro K, Reilly SM, Liu-Bryan R, Terkeltaub R. Xanthine oxidase inhibitor urate-lowering therapy titration to target decreases serum free fatty acids in gout and suppresses lipolysis by adipocytes. Arthritis Res Ther 2022; 24:175. [PMID: 35879786 PMCID: PMC9310412 DOI: 10.1186/s13075-022-02852-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/26/2022] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE Linked metabolic and cardiovascular comorbidities are prevalent in hyperuricemia and gout. For mechanistic insight into impact on inflammatory processes and cardiometabolic risk factors of xanthine oxidase inhibitor urate-lowering therapy (ULT) titration to target, we performed a prospective study of gout serum metabolomes from a ULT trial. METHODS Sera of gout patients meeting the 2015 ACR/EULAR gout classification criteria (n = 20) and with hyperuricemia were studied at time zero and weeks 12 and 24 of febuxostat or allopurinol dose titration ULT. Ultrahigh performance liquid chromatography-tandem mass spectroscopy acquired the serum spectra. Data were assessed using the Metabolon and Metaboloanalyst software. Lipolysis validation assays were done in febuxostat and/or colchicine-treated 3T3-L1 differentiated adipocytes. RESULTS Serum urate decreased from time zero (8.21 ±1.139 SD) at weeks 12 (5.965 ± 1.734 SD) and 24 (5.655 ±1.763 SD). Top metabolites generated by changes in nucleotide and certain amino acid metabolism and polyamine pathways were enriched at 12 and 24 weeks ULT, respectively. Decreases in multiple fatty acid metabolites were observed at 24 weeks, linked with obesity. In cultured adipocytes, febuxostat significantly decreased while colchicine increased the lipolytic response to β-adrenergic-agonism or TNF. CONCLUSION Metabolomic profiles linked xanthine oxidase inhibitor-based ULT titration to target with reduced serum free fatty acids. In vitro validation studies revealed that febuxostat, but not colchicine, reduced lipolysis in cultured adipocytes. Since soluble urate, xanthine oxidase inhibitor treatment, and free fatty acids modulate inflammation, our findings suggest that by suppressing lipolysis, ULT could regulate inflammation in gout and comorbid metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Monica Guma
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA ,grid.7080.f0000 0001 2296 0625Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona Spain
| | - Benyamin Dadpey
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA
| | - Roxana Coras
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA ,grid.7080.f0000 0001 2296 0625Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona Spain
| | - Ted R. Mikuls
- grid.266813.80000 0001 0666 4105University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Bartlett Hamilton
- grid.266813.80000 0001 0666 4105University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Oswald Quehenberger
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA
| | - Hilda Thorisdottir
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - David Bittleman
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Kimberly Lauro
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Shannon M. Reilly
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA ,grid.5386.8000000041936877XWeill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Ru Liu-Bryan
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Robert Terkeltaub
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| |
Collapse
|
42
|
Abstract
INTRODUCTION Gout is the most common form of inflammatory arthritis affecting millions of people around the world. Painful flares and tophaceous deposits can be debilitating, reducing quality of life among those affected and putting strain on health care systems. AREAS COVERED This review provides an overview of the treatment of gout for flare pain management and lowering serum urate. Firstline agents are discussed with emphasis on emerging evidence. Novel therapies are also covered. EXPERT OPINION Lifestyle modifications form a part of gout prevention. Regarding gout flare pharmacotherapy NSAIDs, colchicine and glucocorticoids are first line agents. The IL-1β antagonists also are highly effective for arresting flares but their cost-effectiveness render them as salvage therapies. Allopurinol is an agent of first choice for urate lowering therapy (ULT). In South East Asian and Black populations screening for HLA*B58:01 mutation is a cost-effective approach to decrease the occurrence of the rare but potentially very serious allopurinol hypersensitivity syndrome (AHS.). Febuxostat is another efficacious urate lowering therapy but it has received U.S. FDA black box warning for cardiovascular safety and careful consideration is warranted before its initiation in patients with high cardiovascular risk. Novel uricosurics are a class for continued drug development; verinurad and arhalofenate are agents with future promise. For patients with recalcitrant gout, pegloticase is another effective option in the rheumatologist's armamentarium. Its immunogenicity significantly threatens the achievement of sustained urate lowering responses. Abrogating pegloticase's immunogenicity with immunomodulatory co-therapy may lend to sustained efficacy.
Collapse
Affiliation(s)
- K E G Blake
- Clinical Fellow, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, 1720 2nd Ave South, FOT 839. Birmingham, AL 35294-3408
| | - Jordan L Saag
- Medical Student, University of Central Florida College of Medicine, 6850 Lake Nona Blvd. Orlando, FL 32827
| | - Kenneth G Saag
- Director, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, 1720 2nd Ave South, FOT 839. Birmingham, AL 35294-3408
| |
Collapse
|
43
|
Hyperuricemia and Progression of Chronic Kidney Disease: A Review from Physiology and Pathogenesis to the Role of Urate-Lowering Therapy. Diagnostics (Basel) 2021; 11:diagnostics11091674. [PMID: 34574015 PMCID: PMC8466342 DOI: 10.3390/diagnostics11091674] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/19/2022] Open
Abstract
The relationship between hyperuricemia, gout, and renal disease has been investigated for several years. From the beginning, kidney disease has been considered a complication of gout; however, the viewpoints changed, claiming that hypertension and elevated uric acid (UA) levels are caused by decreased urate excretion in patients with renal impairment. To date, several examples of evidence support the role of hyperuricemia in cardiovascular or renal diseases. Several mechanisms have been identified that explain the relationship between hyperuricemia and chronic kidney disease, including the crystal effect, renin-angiotensin-aldosterone system activation, nitric oxide synthesis inhibition, and intracellular oxidative stress stimulation, and urate-lowering therapy (ULT) has been proven to reduce renal disease progression in the past few years. In this comprehensive review, the source and physiology of UA are introduced, and the mechanisms that explain the reciprocal relationship between hyperuricemia and kidney disease are reviewed. Lastly, current evidence supporting the use of ULT to postpone renal disease progression in patients with hyperuricemia and gout are summarized.
Collapse
|
44
|
Fernandez-Prado R, Ortiz A. Verinurad/Febuxostat and Nephrotoxicity. Am J Kidney Dis 2021; 78:468. [DOI: 10.1053/j.ajkd.2021.03.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022]
|
45
|
Stack AG, Erlandsson F, Terkeltaub R. In Reply to 'Verinurad/Febuxostat and Nephrotoxicity'. Am J Kidney Dis 2021; 78:468-469. [PMID: 34126126 DOI: 10.1053/j.ajkd.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/28/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Austin G Stack
- Department of Nephrology, University Hospital, Limerick & Health Research Institute, University of Limerick, Limerick, Ireland
| | | | | |
Collapse
|
46
|
Leander J, Sunnåker M, Rekić D, Aksenov S, Eriksson UG, Johansson S, Parkinson J. A semi-mechanistic exposure-response model to assess the effects of verinurad, a potent URAT1 inhibitor, on serum and urine uric acid in patients with hyperuricemia-associated diseases. J Pharmacokinet Pharmacodyn 2021; 48:525-541. [PMID: 33728547 PMCID: PMC8225519 DOI: 10.1007/s10928-021-09747-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/02/2021] [Indexed: 01/08/2023]
Abstract
Verinurad, a uric acid transporter 1 (URAT1) inhibitor, lowers serum uric acid by promoting its urinary excretion. Co-administration with a xanthine oxidase inhibitor (XOI) to simultaneously reduce uric acid production rate reduces the potential for renal tubular precipitation of uric acid, which can lead to acute kidney injury. The combination is currently in development for chronic kidney disease and heart failure. The aim of this work was to apply and extend a previously developed semi-mechanistic exposure–response model for uric acid kinetics to include between-subject variability to verinurad and its combinations with XOIs, and to provide predictions to support future treatment strategies. The model was developed using data from 12 clinical studies from a total of 434 individuals, including healthy volunteers, patients with hyperuricemia, and renally impaired subjects. The model described the data well, taking into account the impact of various patient characteristics such as renal function, baseline fractional excretion of uric acid, and race. The potencies (EC50s) of verinurad (reducing uric acid reuptake), febuxostat (reducing uric acid production), and oxypurinol (reducing uric acid production) were: 29, 128, and 13,030 ng/mL, respectively. For verinurad, symptomatic hyperuricemic (gout) subjects showed a higher EC50 compared with healthy volunteers (37 ng/mL versus 29 ng/mL); while no significant difference was found for asymptomatic hyperuricemic patients. Simulations based on the uric acid model were performed to assess dose–response of verinurad in combination with XOI, and to investigate the impact of covariates. The simulations demonstrated application of the model to support dose selection for verinurad.
Collapse
Affiliation(s)
- Jacob Leander
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mikael Sunnåker
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Dinko Rekić
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sergey Aksenov
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Waltham, MA, USA
| | - Ulf G Eriksson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Susanne Johansson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Joanna Parkinson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
47
|
Jalal DI, Chertow GM. Urate Lowering With Combination Therapy in CKD: Reason for Optimism or Einstein's Definition of Insanity? Am J Kidney Dis 2021; 77:478-480. [PMID: 33568321 DOI: 10.1053/j.ajkd.2020.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/10/2020] [Indexed: 01/10/2023]
Affiliation(s)
- Diana I Jalal
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, Iowa City, Iowa.
| | - Glenn M Chertow
- Division of Nephrology, Departments of Medicine and Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
48
|
Rossing P, Persson F, Frimodt-Møller M, Hansen TW. Linking Kidney and Cardiovascular Complications in Diabetes-Impact on Prognostication and Treatment: The 2019 Edwin Bierman Award Lecture. Diabetes 2021; 70:39-50. [PMID: 33355308 PMCID: PMC7881849 DOI: 10.2337/dbi19-0038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In diabetes, increasing albuminuria and decreasing glomerular filtration rate are hallmarks of chronic kidney disease in diabetes and increase the risk of atherosclerotic cardiovascular events and mortality as well as the risk for end-stage kidney disease. For two decades, standard of care has been controlling risk factors, such as glucose, blood pressure, lipids, and lifestyle factors, and specifically use of agents blocking the renin-angiotensin system. This has improved outcome, but a large unmet need has been obvious. After many failed attempts to advance the therapeutic options, the past few years have provided several new promising treatment options such as sodium-glucose cotransporter 2 inhibitors, endothelin receptor antagonists, glucagon-like peptide 1 agonists, and nonsteroidal mineralocorticoid receptor antagonists. The benefits and side effects of these agents demonstrate the link between kidney and heart; some have beneficial effects on both, whereas for other potentially renoprotective agents, development of heart failure has been a limiting factor. They work on different pathways such as hemodynamic, metabolic, inflammatory, and fibrotic targets. We propose that treatment may be personalized if biomarkers or physiological investigations assessing activity in these pathways are applied. This could potentially pave the way for precision medicine, where treatment is optimized for maximal benefit and minimal adverse outcomes. At least it may help prioritizing agents for an individual subject.
Collapse
Affiliation(s)
- Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|