1
|
Waikar SS, Mogg R, Baker AF, Frendl G, Topper M, Adler S, Sultana S, Zhao R, King NMP, Piccoli SP, Sauer JM, Hoffmann S, Nunes I, Sistare FD. Urinary Kidney Injury Biomarker Profiles in Healthy Individuals and After Nephrotoxic and Ischemic Injury. Clin Pharmacol Ther 2025. [PMID: 39754474 DOI: 10.1002/cpt.3531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Two observational studies were conducted to support an initiative to qualify translational kidney safety biomarkers as clinical drug development tools that identify tubular injury prior to changes in estimated glomerular filtration rate (eGFR). Normal healthy volunteers provided three morning spot urine collections over 4 weeks. Patients undergoing surgical resection and intrathoracic cisplatin for malignant pleural mesothelioma provided urine samples pre- and postoperatively at 4, 8, and 12 hours and daily for 6 days. Using receiver-operating characteristics curves, "statistically significant thresholds" established peak longitudinal changes for 8 biomarkers to differentiate mesothelioma patients who developed acute kidney injury (AKI) from normal healthy volunteers. We also assessed "medically significant thresholds" to differentiate mesothelioma patients who did vs. did not develop AKI. Statistically and medically significant thresholds for a fold-change from baseline of urine creatinine (UCr)-normalized values were established for 6 biomarkers: clusterin (2.2, 5.1); osteopontin (3.1, 7.1); N-acetyl-ß-D-glucosaminidase (2.7, 8.1); kidney injury molecule-1 (4.3, 7.5); cystatin C (1.8, 4.5); neutrophil gelatinase-associated lipocalin (2.9, 7.8). For urine albumin and total protein, thresholds were established based on UCr-normalized absolute values: (> upper limit normal, > 10× upper limit normal). Statistically significant thresholds for all biomarkers outperformed eGFR at discriminating mesothelioma subjects exposed to cisplatin from healthy volunteers, demonstrating their utility for enhancing safe drug development. Medically significant thresholds provide perspective on when patients begin to exhibit AKI. These studies have established guideposts for confirmatory studies with additional cohorts and nephrotoxicants to formally qualify the selected biomarkers with worldwide regulatory authorities.
Collapse
Affiliation(s)
- Sushrut S Waikar
- Department of Medicine and Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Robin Mogg
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | - Gyorgy Frendl
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | - Runqi Zhao
- Department of Medicine and Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Steve Hoffmann
- Foundation for the National Institutes of Health, North Bethesda, Maryland, USA
| | | | | |
Collapse
|
2
|
Bhayana S, Schytz PA, Bisgaard Olesen ET, Soh K, Das V. Single-Cell Advances in Investigating and Understanding Chronic Kidney Disease and Diabetic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:55-68. [PMID: 39097167 DOI: 10.1016/j.ajpath.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
Chronic kidney disease (CKD) and its subset diabetic kidney disease are progressive conditions that affect >850 million people worldwide. Diabetes, hypertension, and glomerulonephritis are the most common causes of CKD, which is associated with significant patient morbidity and an increased risk of cardiovascular events, such as heart failure, ultimately leading to premature death. Despite newly approved drugs, increasing evidence shows that patients respond to treatment differently given the complexity of disease heterogeneity and complicated pathophysiology. This review article presents an integrative approach to understanding and addressing CKD through the lens of precision medicine and therapeutics. Advancements in single-cell omics technologies and artificial intelligence can be leveraged to explore the intricate cellular mechanisms underlying CKD and diabetic kidney disease pathogenesis. Dissecting the cellular heterogeneity and identifying rare cell populations using single-cell approaches will facilitate uncovering novel therapeutic targets and biomarkers for personalized treatment strategies. Finally, we discuss the potential of artificial intelligence-driven analyses in predicting disease progression and treatment response, thereby paving the way for tailored interventions.
Collapse
Affiliation(s)
- Sagar Bhayana
- Kidney Biology, Global Drug Development, Novo Nordisk A/S, Søborg, Denmark
| | - Philip A Schytz
- Cardiovascular, Kidney and Alzheimer Disease, Medical and Science, Novo Nordisk A/S, Søborg, Denmark
| | - Emma T Bisgaard Olesen
- Cardiovascular, Kidney and Alzheimer Disease, Medical and Science, Novo Nordisk A/S, Søborg, Denmark
| | - Keng Soh
- Integrated Omics, AI and Analytics, Development, Novo Nordisk A/S, Søborg, Denmark
| | - Vivek Das
- Integrated Omics, AI and Analytics, Development, Novo Nordisk A/S, Søborg, Denmark.
| |
Collapse
|
3
|
Claudel SE, Waikar SS, Schmidt IM, Vasan RS, Verma A. The relationship between low levels of albuminuria and mortality among adults without major cardiovascular risk factors. Eur J Prev Cardiol 2024; 31:2046-2055. [PMID: 38825979 PMCID: PMC11629963 DOI: 10.1093/eurjpc/zwae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
AIMS The aim of this study is to determine whether elevated levels of albuminuria within the low range [urinary albumin-to-creatinine ratio (UACR) <30 mg/g] are linked to cardiovascular death in adults lacking major cardiovascular risk factors. METHODS AND RESULTS The association between UACR and cardiovascular mortality was investigated among 12 835 participants in the 1999-2014 National Health and Nutrition Examination Survey using Cox proportional hazard models and confounder-adjusted survival curves. We excluded participants with baseline cardiovascular disease, hypertension, diabetes, pre-diabetes, an estimated glomerular filtration rate <60 mL/min/1.73 m2, currently pregnant, and those who received dialysis last year. Over a median follow-up of 12.3 years, 110 and 621 participants experienced cardiovascular and all-cause mortality. In multivariable-adjusted models, each doubling of UACR was associated with a 36% higher risk of cardiovascular death [hazard ratio (HR) 1.36, 95% confidence interval (CI) 1.02-1.82] and a 24% higher risk of all-cause mortality (HR 1.24, 95% CI 1.10-1.39). The 15-year adjusted cumulative incidences of cardiovascular mortality were 0.91, 0.99, and 2.1% for UACR levels of <4.18, 4.18 to <6.91, and ≥6.91 mg/g, respectively. The 15-year adjusted cumulative incidences of all-cause mortality were 5.1, 6.1, and 7.4% for UACR levels of <4.18, 4.18 to <6.91, and ≥6.91 mg/g, respectively. CONCLUSION Adults with elevated levels of albuminuria within the low range (UACR <30 mg/g) and no major cardiovascular risk factors had elevated risks of cardiovascular and all-cause mortality. The risk increased linearly with higher albuminuria levels. This emphasizes a risk gradient across all albuminuria levels, even within the supposedly normal range, adding to the existing evidence.
Collapse
Affiliation(s)
- Sophie E Claudel
- Department of Medicine, Boston Medical Center, 72 E. Concord Street, Boston, MA 02118, USA
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, X-521, 650 Albany Street, Boston, MA 02118, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, X-521, 650 Albany Street, Boston, MA 02118, USA
- Section of Nephrology, Department of Medicine, Boston Medical Center, 650 Albany Street, Boston, MA 02118, USA
| | - Insa M Schmidt
- Section of Nephrology, Department of Medicine, Boston Medical Center, 650 Albany Street, Boston, MA 02118, USA
- Hamburg Center for Kidney Health, University Medical Center Hamburg, Martinistrase 52 Campus Research N27, Hamburg 20246, Germany
| | - Ramachandran S Vasan
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, X-521, 650 Albany Street, Boston, MA 02118, USA
- Department of Quantitative and Qualitative Health Sciences, University of Texas School of Public Health and University of Texas Health Science Center in San Antonio, San Antonio, TX, USA
| | - Ashish Verma
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, X-521, 650 Albany Street, Boston, MA 02118, USA
- Section of Nephrology, Department of Medicine, Boston Medical Center, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
4
|
McCoy IE, Oates A, Hsu CY. Urine Protein-to-Creatinine Ratio Remains Informative Despite Nonsteady State Serum Creatinine During Acute Kidney Injury. Kidney Int Rep 2024; 9:3455-3463. [PMID: 39698350 PMCID: PMC11652066 DOI: 10.1016/j.ekir.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Experts have cautioned that assessment of proteinuria using urine protein-to-creatinine ratios (UPCRs) are not valid during acute kidney injury (AKI) because reduced urine creatinine in the denominator may artificially inflate the ratio. However, there is little empiric data assessing this theoretical concern. Methods Here, we retrospectively examined changes in UPCRs measured during episodes of severe AKI and assessed whether the magnitude and direction of these changes associate with how the serum creatinine level is changing at the time of UPCR collection. We repeated these analyses comparing hospitalization UPCRs with prehospitalization or posthospitalization UPCRs, where available. Results Among 329 adults hospitalized with stage 2 or 3 AKI (defined as peak:nadir serum creatinine during hospitalization ≥ 2) at the University of California, San Francisco from January 1, 2014 to December 31, 2022 with multiple UPCRs measured during AKI hospitalization, UPCR values were similar whether the serum creatinine was increasing or decreasing at the time of measurement (median difference, 0.06 g/g; interquartile range [IQR], -0.26 to 0.50 g/g). There was no association between the difference in serum creatinine slopes when the UPCRs were collected and the difference in UPCR values (UPCR 0.05 g/g higher per mg/dl/d serum creatinine slope; 95% confidence interval [CI], -0.36 to 0.47, P = 0.80). UPCRs measured during hospitalization demonstrated positive and negative predictive values suggesting utility in appraising clinically relevant outpatient UPCR levels. Conclusion Despite nonsteady state serum creatinine at the time of collection, UPCRs measured during AKI hospitalizations may be more informative than previously believed and should not be wholly disregarded.
Collapse
Affiliation(s)
- Ian E. McCoy
- Division of Nephrology, University of California San Francisco, San Francisco, California, USA
| | - Aris Oates
- Division of Pediatric Nephrology, University of California San Francisco, San Francisco, California, USA
| | - Chi-yuan Hsu
- Division of Nephrology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
5
|
Min K, Matsumoto Y, Asakura M, Ishihara M. Rediscovery of the implication of albuminuria in heart failure: emerging classic index for cardiorenal interaction. ESC Heart Fail 2024; 11:3470-3487. [PMID: 38725278 PMCID: PMC11631258 DOI: 10.1002/ehf2.14811] [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: 11/29/2023] [Revised: 03/09/2024] [Accepted: 04/02/2024] [Indexed: 12/12/2024] Open
Abstract
The development of new drugs and device therapies has led to remarkable advancements in heart failure (HF) treatment in the past couple of decades. However, it becomes increasingly evident that guideline-directed medical therapy cannot be one-size-fits-all across a wide range of ejection fractions (EFs) and various aetiologies. Therefore, classifications solely relying on EF and natriuretic peptide make optimization of treatment challenging, and there is a growing exploration of new indicators that enable efficient risk stratification of HF patients. Particularly when considering HF as a multi-organ interaction syndrome, the cardiorenal interaction plays a central role in its pathophysiology, and albuminuria has gained great prominence as its biomarker, independent from glomerular filtration rate. Albuminuria has been shown to exhibit a linear correlation with cardiovascular disease and HF prognosis in multiple epidemiological studies, ranging from normal (<30 mg/g) to high levels (>300 mg/g). However, on the other hand, it is only recently that the details of the pathological mechanisms that give rise to albuminuria have begun to be elucidated, including the efficient compaction/tightening of the glomerular basement membrane by podocytes and mesangial cells. Interestingly, renal disease, diabetes, and HF damage these components associated with albuminuria, and experimental models have demonstrated that recently developed HF drugs reduce albuminuria by ameliorating these pathological phenotypes. In this review, facing the rapid expansion of horizons in HF treatment, we aim to clarify the current understanding of the pathophysiology of albuminuria and explore the comprehensive understanding of albuminuria by examining the clinically established evidence to date, the pathophysiological mechanisms leading to its occurrence, and the outcomes of clinical studies utilizing various drug classes committed to specific pathological mechanisms to put albuminuria as a novel axis to depict the pathophysiology of HF.
Collapse
Affiliation(s)
- Kyung‐Duk Min
- Department of Cardiovascular and Renal MedicineHyogo Medical University1‐1 Mukogawa‐choNishinomiya663‐8501HyogoJapan
| | - Yuki Matsumoto
- Department of Cardiovascular and Renal MedicineHyogo Medical University1‐1 Mukogawa‐choNishinomiya663‐8501HyogoJapan
| | - Masanori Asakura
- Department of Cardiovascular and Renal MedicineHyogo Medical University1‐1 Mukogawa‐choNishinomiya663‐8501HyogoJapan
| | - Masaharu Ishihara
- Department of Cardiovascular and Renal MedicineHyogo Medical University1‐1 Mukogawa‐choNishinomiya663‐8501HyogoJapan
| |
Collapse
|
6
|
Lin YT, Wuopio J, Larsson A, Malinovschi A, Feldreich T, Engström G, Fall T, Ärnlöv J. The association between novel urinary kidney damage biomarkers and coronary atherosclerosis in an apparently healthy population. Sci Rep 2024; 14:29215. [PMID: 39587192 PMCID: PMC11589585 DOI: 10.1038/s41598-024-80321-5] [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: 02/27/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
Several novel urinary kidney damage biomarkers predict the progression of kidney disease. However, the relations of these biomarkers to atherosclerosis, a major consequence of kidney disease, are less studied. Urinary levels of several biomarkers, including kidney injury molecule-1 (KIM-1), osteopontin, epidermal growth factor, and Dickkopf-3, were assessed in participants enrolled in the Swedish CArdioPulmonary BioImage Study. The study included 9,628 individuals with a mean age of 57.5 years, of which 52.4% were women. The presence of coronary artery stenosis and the coronary artery calcium score (CACS) were determined using coronary computed tomography angiography. To analyze the associations between coronary atherosclerosis and urinary biomarker levels, an ordered logistic regression model adusting for confounding factors was employed. KIM-1 was the only biomarker associated with both coronary stenosis and CACS after adjusting for established cardiovascular risk factors (odds ratio [95% confidence intervals], 1.23[1.05-1.44] and 1.25[1.07-1.47]). These results were consistent in sensitivity analyses of individuals without hypertension, diabetes, or known cardiovascular disease and with normal kidney function. Urinary KIM-1, a specific marker of proximal tubular damage, was robustly linked to coronary atherosclerosis even in apparently healthy individuals, which suggests that the detrimental interplay between the kidney and cardiovascular system begins before clinically overt kidney disease. Additional studies are warranted to evaluate the urinary KIM-1 to predict kidney and cardiovascular disease.
Collapse
Affiliation(s)
- Yi-Ting Lin
- Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jonas Wuopio
- Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Center for Clinical Research Dalarna, Uppsala University, Falun, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | | | - Gunnar Engström
- Department of Clinical Sciences, Cardiovascular Epidemiology, Lund University, Malmö, Sweden
| | - Tove Fall
- Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.
- Center for Clinical Research Dalarna, Uppsala University, Falun, Sweden.
- School of Health and Welfare, Dalarna University, Falun, Sweden.
| |
Collapse
|
7
|
Degenaar A, Kruger R, Jacobs A, Mels CMC. Phenotyping Kidney Function in Young Adults With High Blood Pressure: The African-PREDICT Study. J Clin Hypertens (Greenwich) 2024; 26:1291-1300. [PMID: 39368068 PMCID: PMC11555542 DOI: 10.1111/jch.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024]
Abstract
Biomarkers of kidney function, including glomerular, tubular, and fibrotic markers, have been associated with blood pressure in elderly populations and individuals with kidney and cardiovascular diseases. However, limited information is available in young adults. In this study, we compared levels of several kidney function biomarkers between normotensive and hypertensive young adults and explored the associations of these biomarkers with blood pressure within these groups. In this cross-sectional assessment, twenty-four-hour (24-h) blood pressure measurements of 1055 participants (mean age = 24.6 years) were used to classify hypertension as per the 2018 ESC/ESH guidelines. Biomarkers of kidney function included estimated glomerular filtration rate, urinary albumin, alpha-1 microglobulin (uA1M), neutrophil gelatinase-associated lipocalin (uNGAL), uromodulin (uUMOD), and the CKD273 classifier. All urinary biomarkers, except for the CKD273 classifier, were standardized for urinary creatinine (Cr). In the hypertensive group (61.0% White; 73.2% men), urinary albumin-to-creatinine ratio (uACR), uNGAL/Cr and uUMOD/Cr were lower than the normotensive group. In multiple regression analyses, 24-h systolic blood pressure (SBP) (β = 0.14; p = 0.042), 24-h diastolic blood pressure (DBP) (β = 0.14; p = 0.040), and 24-h mean arterial pressure (MAP) (β = 0.16; p = 0.020) associated positively with uA1M/Cr in the hypertensive group, while 24-h MAP positively associated with uACR (β = 0.17; p = 0.017). In exploratory factor analysis, positive associations of 24-h DBP and 24-h MAP with a factor pattern including tubular biomarkers were observed in the hypertensive group (24-h DBP: β = 0.18; p = 0.026, 24-h MAP: β = 0.17; p = 0.032). In the setting of hypertension, high perfusion pressure in the kidneys may play a role in the development of proximal tubule damage and promote early deterioration in kidney function in young adults. Trial Registration: ClinicalTrials.gov identifier: NCT03292094.
Collapse
Affiliation(s)
- Anja Degenaar
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| | - Adriaan Jacobs
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| | - Catharina M. C. Mels
- Hypertension in Africa Research Team (HART)North‐West UniversityPotchefstroomSouth Africa
- Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, North‐West UniversityPotchefstroomSouth Africa
| |
Collapse
|
8
|
Kouri TT, Hofmann W, Falbo R, Oyaert M, Schubert S, Gertsen JB, Merens A, Pestel-Caron M. The EFLM European Urinalysis Guideline 2023. Clin Chem Lab Med 2024; 62:1653-1786. [PMID: 38534005 DOI: 10.1515/cclm-2024-0070] [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: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND The EFLM Task and Finish Group Urinalysis has updated the ECLM European Urinalysis Guidelines (2000) on urinalysis and urine bacterial culture, to improve accuracy of these examinations in European clinical laboratories, and to support diagnostic industry to develop new technologies. RECOMMENDATIONS Graded recommendations were built in the following areas. MEDICAL NEEDS AND TEST REQUISITION Strategies of urine testing are described to patients with complicated or uncomplicated urinary tract infection (UTI), and high or low-risk to kidney disease. SPECIMEN COLLECTION Patient preparation, and urine collection are supported with two quality indicators: contamination rate (cultures), and density of urine (chemistry, particles). CHEMISTRY Measurements of both urine albumin and α1-microglobulin are recommended for sensitive detection of kidney disease in high-risk patients. Performance specifications are given for urine protein measurements and quality control of multiproperty strip tests. PARTICLES Procedures for microscopy are reviewed for diagnostic urine particles, including urine bacteria. Technologies in automated particle counting and visual microscopy are updated with advice how to verify new instruments with the reference microscopy. BACTERIOLOGY Chromogenic agar is recommended as primary medium in urine cultures. Limits of significant growth are reviewed, with an optimised workflow for routine specimens, using leukocyturia to reduce less important antimicrobial susceptibility testing. Automation in bacteriology is encouraged to shorten turn-around times. Matrix assisted laser desorption ionization time-of-flight mass spectrometry is applicable for rapid identification of uropathogens. Aerococcus urinae, A. sanguinicola and Actinotignum schaalii are taken into the list of uropathogens. A reference examination procedure was developed for urine bacterial cultures.
Collapse
Affiliation(s)
- Timo T Kouri
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
- HUSLAB, HUS Diagnostic Center, Hospital District of Helsinki and Uusimaa, 00014 Helsinki, Finland
| | | | - Rosanna Falbo
- University Department of Laboratory Medicine, ASST Brianza, Pio XI Hospital, 20832 Desio (MB), Italy
| | - Matthijs Oyaert
- Department of Laboratory Medicine, University Hospital Ghent, 9000 Ghent, Belgium
| | - Sören Schubert
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Jan Berg Gertsen
- Department of Clinical Microbiology, Aarhus University Hospital, Skejby, 8200 Aarhus N, Denmark
| | - Audrey Merens
- Service de Biologie Médicale, Hôpital d'Instruction des Armées Bégin, 94160 Saint-Mandé, France
| | - Martine Pestel-Caron
- Department of Microbiology, CHU Rouen, University of Rouen Normandie, INSERM, DYNAMICURE UMR 1311, 76000 Rouen, France
| |
Collapse
|
9
|
Lamb EJ, Barratt J, Brettell EA, Cockwell P, Dalton RN, Deeks JJ, Eaglestone G, Pellatt-Higgins T, Kalra PA, Khunti K, Loud FC, Ottridge RS, Potter A, Rowe C, Scandrett K, Sitch AJ, Stevens PE, Sharpe CC, Shinkins B, Smith A, Sutton AJ, Taal MW. Accuracy of glomerular filtration rate estimation using creatinine and cystatin C for identifying and monitoring moderate chronic kidney disease: the eGFR-C study. Health Technol Assess 2024; 28:1-169. [PMID: 39056437 PMCID: PMC11331378 DOI: 10.3310/hyhn1078] [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: 07/28/2024] Open
Abstract
Background Estimation of glomerular filtration rate using equations based on creatinine is widely used to manage chronic kidney disease. In the UK, the Chronic Kidney Disease Epidemiology Collaboration creatinine equation is recommended. Other published equations using cystatin C, an alternative marker of kidney function, have not gained widespread clinical acceptance. Given higher cost of cystatin C, its clinical utility should be validated before widespread introduction into the NHS. Objectives Primary objectives were to: (1) compare accuracy of glomerular filtration rate equations at baseline and longitudinally in people with stage 3 chronic kidney disease, and test whether accuracy is affected by ethnicity, diabetes, albuminuria and other characteristics; (2) establish the reference change value for significant glomerular filtration rate changes; (3) model disease progression; and (4) explore comparative cost-effectiveness of kidney disease monitoring strategies. Design A longitudinal, prospective study was designed to: (1) assess accuracy of glomerular filtration rate equations at baseline (n = 1167) and their ability to detect change over 3 years (n = 875); (2) model disease progression predictors in 278 individuals who received additional measurements; (3) quantify glomerular filtration rate variability components (n = 20); and (4) develop a measurement model analysis to compare different monitoring strategy costs (n = 875). Setting Primary, secondary and tertiary care. Participants Adults (≥ 18 years) with stage 3 chronic kidney disease. Interventions Estimated glomerular filtration rate using the Chronic Kidney Disease Epidemiology Collaboration and Modification of Diet in Renal Disease equations. Main outcome measures Measured glomerular filtration rate was the reference against which estimating equations were compared with accuracy being expressed as P30 (percentage of values within 30% of reference) and progression (variously defined) studied as sensitivity/specificity. A regression model of disease progression was developed and differences for risk factors estimated. Biological variation components were measured and the reference change value calculated. Comparative costs of monitoring with different estimating equations modelled over 10 years were calculated. Results Accuracy (P30) of all equations was ≥ 89.5%: the combined creatinine-cystatin equation (94.9%) was superior (p < 0.001) to other equations. Within each equation, no differences in P30 were seen across categories of age, gender, diabetes, albuminuria, body mass index, kidney function level and ethnicity. All equations showed poor (< 63%) sensitivity for detecting patients showing kidney function decline crossing clinically significant thresholds (e.g. a 25% decline in function). Consequently, the additional cost of monitoring kidney function annually using a cystatin C-based equation could not be justified (incremental cost per patient over 10 years = £43.32). Modelling data showed association between higher albuminuria and faster decline in measured and creatinine-estimated glomerular filtration rate. Reference change values for measured glomerular filtration rate (%, positive/negative) were 21.5/-17.7, with lower reference change values for estimated glomerular filtration rate. Limitations Recruitment of people from South Asian and African-Caribbean backgrounds was below the study target. Future work Prospective studies of the value of cystatin C as a risk marker in chronic kidney disease should be undertaken. Conclusions Inclusion of cystatin C in glomerular filtration rate-estimating equations marginally improved accuracy but not detection of disease progression. Our data do not support cystatin C use for monitoring of glomerular filtration rate in stage 3 chronic kidney disease. Trial registration This trial is registered as ISRCTN42955626. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 11/103/01) and is published in full in Health Technology Assessment; Vol. 28, No. 35. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- Edmund J Lamb
- Clinical Biochemistry, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Elizabeth A Brettell
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Paul Cockwell
- Renal Medicine, Queen Elizabeth Hospital Birmingham and Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - R Nei Dalton
- WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital, London, UK
| | - Jon J Deeks
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Gillian Eaglestone
- Kent Kidney Care Centre, East Kent Hospitals University NHS Foundation Trust, Kent, UK
| | | | - Philip A Kalra
- Department of Renal Medicine, Salford Royal Hospital Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | | | - Ryan S Ottridge
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Aisling Potter
- Clinical Biochemistry, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK
| | - Ceri Rowe
- Clinical Biochemistry, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK
| | - Katie Scandrett
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Alice J Sitch
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Paul E Stevens
- Kent Kidney Care Centre, East Kent Hospitals University NHS Foundation Trust, Kent, UK
| | - Claire C Sharpe
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Bethany Shinkins
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Alison Smith
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Andrew J Sutton
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Maarten W Taal
- Department of Renal Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| |
Collapse
|
10
|
Claudel SE, Waikar SS. Imprecise, Measure it Twice? Dealing With the Biological Variability of Albuminuria in Diabetes. Am J Kidney Dis 2024; 84:1-3. [PMID: 38661613 DOI: 10.1053/j.ajkd.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/23/2024] [Accepted: 03/02/2024] [Indexed: 04/26/2024]
Affiliation(s)
- Sophie E Claudel
- Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts.
| |
Collapse
|
11
|
Zannad F, Sanyal AJ, Butler J, Ferreira JP, Girerd N, Miller V, Pandey A, Parikh CR, Ratziu V, Younossi ZM, Harrison SA. MASLD and MASH at the crossroads of hepatology trials and cardiorenal metabolic trials. J Intern Med 2024; 296:24-38. [PMID: 38738988 DOI: 10.1111/joim.13793] [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] [Indexed: 05/14/2024]
Abstract
Steatotic liver disease (SLD) is a worldwide public health problem, causing considerable morbidity and mortality. Patients with SLD are at increased risk for major adverse cardiovascular (CV) events, type 2 diabetes mellitus and chronic kidney disease. Conversely, patients with cardiometabolic conditions have a high prevalence of SLD. In addition to epidemiological evidence linking many of these conditions, there is evidence of shared pathophysiological processes. In December 2022, a unique multi-stakeholder, multi-specialty meeting, called MOSAIC (Metabolic multi Organ Science Accelerating Innovation in Clinical Trials) was convened to foster collaboration across metabolic, hepatology, nephrology and CV disorders. One of the goals of the meeting was to consider approaches to drug development that would speed regulatory approval of treatments for multiple disorders by combining liver and cardiorenal endpoints within a single study. Non-invasive tests, including biomarkers and imaging, are needed in hepatic and cardiorenal trials. They can be used as trial endpoints, to enrich trial populations, to diagnose and risk stratify patients and to assess treatment efficacy and safety. Although they are used in proof of concept and phase 2 trials, they are often not acceptable for regulatory approval of therapies. The challenge is defining the optimal combination of biomarkers, imaging and morbidity/mortality outcomes and ensuring that they are included in future trials while minimizing the burden on patients, trialists and trial sponsors. This paper provides an overview of some of the wide array of CV, liver and kidney measurements that were discussed at the MOSAIC meeting.
Collapse
Affiliation(s)
- Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center at Institut Lorrain du Coeur et des Vaisseaux, University Hospital of Nancy, Nancy, France
| | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, USA
- University of Mississippi, Jackson, Mississippi, USA
| | - João Pedro Ferreira
- UnIC@RISE, Cardiovascular Research and Development Center, Department Surgery Physiology, University of Porto, Porto, Portugal
- Centre d'Investigations Cliniques Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
- F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre Hospitalier Régional Universitaire de Nancy, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Veronica Miller
- Forum for Collaborative Research, Washington, District of Columbia, USA
- University of California Berkeley School of Public Health, Berkeley, California, USA
| | | | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vlad Ratziu
- Sorbonne Université, Hôpital Pitié-Salpêtrière, Institute for Cardiometabolism and Nutrition, INSERM UMRS, Paris, France
| | | | - Stephen A Harrison
- Visiting Professor of Hepatology Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Rasaratnam N, Salim A, Blackberry I, Cooper ME, Magliano DJ, van Wijngaarden P, Varadarajan S, Sacre JW, Shaw JE. Urine Albumin-Creatinine Ratio Variability in People With Type 2 Diabetes: Clinical and Research Implications. Am J Kidney Dis 2024; 84:8-17.e1. [PMID: 38551531 DOI: 10.1053/j.ajkd.2023.12.018] [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: 07/10/2023] [Revised: 11/27/2023] [Accepted: 12/09/2023] [Indexed: 06/23/2024]
Abstract
RATIONALE & OBJECTIVE Evidence has demonstrated that albuminuria is a key diagnostic and prognostic marker of diabetic chronic kidney disease, but the impact of its day-to-day variability has not been adequately considered. This study quantified within-individual variability of albuminuria in people with type 2 diabetes to inform clinical albuminuria monitoring. STUDY DESIGN Descriptive cross-sectional analysis. SETTING & PARTICIPANTS People with type 2 diabetes (n=826, 67.1 [IQR, 60.3-72.4] years, 64.9% male) participating in the Progression of Diabetic Complications (PREDICT) cohort study. EXPOSURE Four spot urine collections for measurement of urinary albumin-creatinine ratio (UACR) within 4 weeks. OUTCOME Variability of UACR. ANALYTICAL APPROACH We characterized within-individual variability (coefficient of variation [CV], 95% limits of random variation, intraclass correlation coefficient), developed a calculator displaying probabilities that any observed difference between a pair of UACR values truly exceeded a 30% difference, and estimated the ranges of diagnostic uncertainty to inform a need for additional UACR collections to exclude or confirm albuminuria. Multiple linear regression examined factors influencing UACR variability. RESULTS We observed high within-individual variability (CV 48.8%; 95% limits of random variation showed a repeated UACR to be as high/low as 3.78/0.26 times the first). If a single-collection UACR increased from 2 to 5mg/mmol, the probability that UACR actually increased by at least 30% was only 50%, rising to 97% when 2 collections were obtained at each time point. The ranges of diagnostic uncertainty were 2.0-4.0mg/mmol after an initial UACR test, narrowing to 2.4-3.2 and 2.7-2.9mg/mmol for the mean of 2 and 3 collections, respectively. Some factors correlated with higher (female sex; moderately increased albuminuria) or lower (reduced estimated glomerular filtration rate and sodium-glucose cotransporter 2 inhibitor/angiotensin-converting enzyme inhibitor/angiotensin receptor blocker treatment) within-individual UACR variability. LIMITATIONS Reliance on the mean of 4 UACR collections as the reference standard for albuminuria. CONCLUSIONS UACR demonstrates a high degree of within-individual variability among individuals with type 2 diabetes. Multiple urine collections for UACR may improve capacity to monitor changes over time in clinical and research settings but may not be necessary for the diagnosis of albuminuria. PLAIN-LANGUAGE SUMMARY Albuminuria (albumin in urine) is a diagnostic and prognostic marker of diabetic chronic kidney disease. However, albuminuria can vary within an individual from day to day. We compared 4 random spot urinary albumin-creatinine ratio (UACR) samples from 826 participants. We found that a second UACR collection may be as small as a fourth or as large as almost 4 times the first sample's UACR level. This high degree of variability presents a challenge to our ability to interpret changes in albuminuria. Multiple collections have been suggested as a solution. We have constructed tools that may aid clinicians in deciding how many urine collections are required to monitor and diagnose albuminuria. Multiple urine collections may be required for individual monitoring but not necessarily for diagnosis.
Collapse
Affiliation(s)
- Natasha Rasaratnam
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia; School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | - Irene Blackberry
- Care Economy Research Institute, La Trobe University, Wodonga, Australia
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
| | | | - Julian W Sacre
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, Australia; School of Life Sciences, La Trobe University, Melbourne, Australia
| |
Collapse
|
13
|
Huang JX, Copeland TP, Pitts CE, Myers SR, Kilberg MJ, Ku E, Glaser N. Transient albuminuria in the setting of short-term severe hyperglycemia in type 1 diabetes. J Diabetes Complications 2024; 38:108762. [PMID: 38703638 DOI: 10.1016/j.jdiacomp.2024.108762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
In a cohort of 1817 children with type 1 diabetes (T1D), short-term hyperglycemia was associated with transient albuminuria (11 % during new-onset T1D without diabetic ketoacidosis (DKA), 12 % during/after DKA, 6 % during routine screening). Our findings have implications regarding future risk of diabetic kidney disease and further investigation is needed.
Collapse
Affiliation(s)
- Jia Xin Huang
- Department of Pediatrics, University of California San Francisco, United States of America.
| | - Timothy P Copeland
- Department of Medicine, University of California San Francisco, United States of America
| | - Casey E Pitts
- Department of Medicine, University of California San Francisco, United States of America
| | - Sage R Myers
- Department of Medicine, University of California San Francisco, United States of America
| | - Marissa J Kilberg
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, United States of America
| | - Elaine Ku
- Department of Pediatrics, University of California San Francisco, United States of America; Department of Medicine, University of California San Francisco, United States of America
| | - Nicole Glaser
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, United States of America
| |
Collapse
|
14
|
Wang Q, Meeusen JW. Clinical Impacts of Implementing the 2021 Race-Free Chronic Kidney Disease Epidemiology Collaboration Estimated Glomerular Filtration Rate. J Appl Lab Med 2024; 9:586-598. [PMID: 38366867 DOI: 10.1093/jalm/jfad137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 02/18/2024]
Abstract
BACKGROUND Estimated glomerular filtration rate (eGFR) has become incorporated into multiple clinical management situations. Historically, equations included a Black race coefficient, which lacked biological plausibility and created potential to exacerbate health disparities. A new equation created in 2021 changed the weighting of age, sex, and creatinine by modeling against a diverse cohort and removing the Black race coefficient. CONTENT A variety of clinical outcomes including kidney disease risk stratification, medication dosing, patient eligibility for clinical trials, and kidney donation are impacted by implementation of the new equation. Nearly 2 years after its initial publication, many studies have reported on observed analytical performance of the 2021 eGFR determined as diagnostic concordance and percentage of estimates within 30% of measured GFR. Additionally, the potential clinical impacts following adoption of the new eGFR among different patient populations has also been reported. Here we review these studies with a focus on assessing the data associated with the transition from 2009 to 2021 Chronic Kidney Disease Epidemiology Collaboration equations. SUMMARY The reported interindividual variation in eGFR performance is significantly larger than any potential benefit derived from race coefficients. Both the 2021 eGFR and the 2009 eGFR analytical performance fall short of the validation cohort performance in most cohorts. However, the 2021 analytical is similar or better than the 2009 eGFR in most cohorts. Implementing the 2021 eGFR will remove a systematic overestimation of kidney function among Black patients.
Collapse
Affiliation(s)
- Qian Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN, United States
| | - Jeffrey W Meeusen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN, United States
| |
Collapse
|
15
|
Hwang JH, Hsu CY. A "Fit for Purpose" Approach to CKD Classification? Am J Kidney Dis 2024; 83:564-565. [PMID: 38300185 DOI: 10.1053/j.ajkd.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024]
Affiliation(s)
- Jin Ho Hwang
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea; Division of Nephrology, University of California San Francisco, San Francisco, California
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California San Francisco, San Francisco, California.
| |
Collapse
|
16
|
Bock F, Isermann B. Does it matter how we measure urinary creatinine in patients taking SGLT2 inhibitors? Nephrol Dial Transplant 2024; 39:739-741. [PMID: 38218592 DOI: 10.1093/ndt/gfae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Indexed: 01/15/2024] Open
Affiliation(s)
- Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| |
Collapse
|
17
|
Verma A, Schmidt IM, Claudel S, Palsson R, Waikar SS, Srivastava A. Association of Albuminuria With Chronic Kidney Disease Progression in Persons With Chronic Kidney Disease and Normoalbuminuria : A Cohort Study. Ann Intern Med 2024; 177:467-475. [PMID: 38560911 DOI: 10.7326/m23-2814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Albuminuria is a major risk factor for chronic kidney disease (CKD) progression, especially when categorized as moderate (30 to 300 mg/g) or severe (>300 mg/g). However, there are limited data on the prognostic value of albuminuria within the normoalbuminuric range (<30 mg/g) in persons with CKD. OBJECTIVE To estimate the increase in the cumulative incidence of CKD progression with greater baseline levels of albuminuria among persons with CKD who had normoalbuminuria (<30 mg/g). DESIGN Multicenter prospective cohort study. SETTING 7 U.S. clinical centers. PARTICIPANTS 1629 participants meeting criteria from the CRIC (Chronic Renal Insufficiency Cohort) study with CKD (estimated glomerular filtration rate [eGFR], 20 to 70 mL/min/1.73 m2) and urine albumin-creatinine ratio (UACR) less than 30 mg/g. MEASUREMENTS Baseline spot urine albumin divided by spot urine creatinine to calculate UACR as the exposure variable. The 10-year adjusted cumulative incidences of CKD progression (composite of 50% eGFR decline or kidney failure [dialysis or kidney transplantation]) from confounder adjusted survival curves using the G-formula. RESULTS Over a median follow-up of 9.8 years, 182 of 1629 participants experienced CKD progression. The 10-year adjusted cumulative incidences of CKD progression were 8.7% (95% CI, 5.9% to 11.6%), 11.5% (CI, 8.8% to 14.3%), and 19.5% (CI, 15.4% to 23.5%) for UACR levels of 0 to less than 5 mg/g, 5 to less than 15 mg/g, and 15 mg/g or more, respectively. Comparing persons with UACR 15 mg/g or more to those with UACR 5 to less than 15 mg/g and 0 to less than 5 mg/g, the absolute risk differences were 7.9% (CI, 3.0% to 12.7%) and 10.7% (CI, 5.8% to 15.6%), respectively. The 10-year adjusted cumulative incidence increased linearly based on baseline UACR levels. LIMITATION UACR was measured once. CONCLUSION Persons with CKD and normoalbuminuria (<30 mg/g) had excess risk for CKD progression, which increased in a linear fashion with higher levels of albuminuria. PRIMARY FUNDING SOURCE None.
Collapse
Affiliation(s)
- Ashish Verma
- Boston Medical Center and Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (A.V., S.S.W.)
| | - Insa M Schmidt
- Boston Medical Center and Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; and Hamburg Center for Kidney Health, University Medical Center Hamburg, Hamburg, Germany (I.M.S.)
| | - Sophie Claudel
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (S.C.)
| | - Ragnar Palsson
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts (R.P.)
| | - Sushrut S Waikar
- Boston Medical Center and Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts (A.V., S.S.W.)
| | - Anand Srivastava
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois (A.S.)
| |
Collapse
|
18
|
Rupprecht H, Catanese L, Amann K, Hengel FE, Huber TB, Latosinska A, Lindenmeyer MT, Mischak H, Siwy J, Wendt R, Beige J. Assessment and Risk Prediction of Chronic Kidney Disease and Kidney Fibrosis Using Non-Invasive Biomarkers. Int J Mol Sci 2024; 25:3678. [PMID: 38612488 PMCID: PMC11011737 DOI: 10.3390/ijms25073678] [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: 02/07/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Effective management of chronic kidney disease (CKD), a major health problem worldwide, requires accurate and timely diagnosis, prognosis of progression, assessment of therapeutic efficacy, and, ideally, prediction of drug response. Multiple biomarkers and algorithms for evaluating specific aspects of CKD have been proposed in the literature, many of which are based on a small number of samples. Based on the evidence presented in relevant studies, a comprehensive overview of the different biomarkers applicable for clinical implementation is lacking. This review aims to compile information on the non-invasive diagnostic, prognostic, and predictive biomarkers currently available for the management of CKD and provide guidance on the application of these biomarkers. We specifically focus on biomarkers that have demonstrated added value in prospective studies or those based on prospectively collected samples including at least 100 subjects. Published data demonstrate that several valid non-invasive biomarkers of potential value in the management of CKD are currently available.
Collapse
Affiliation(s)
- Harald Rupprecht
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (H.R.); (L.C.)
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
| | - Lorenzo Catanese
- Department of Nephrology, Angiology and Rheumatology, Klinikum Bayreuth GmbH, 95445 Bayreuth, Germany; (H.R.); (L.C.)
- Department of Nephrology, Medizincampus Oberfranken, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Kuratorium for Dialysis and Transplantation (KfH) Bayreuth, 95445 Bayreuth, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Felicitas E. Hengel
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Tobias B. Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | | | - Maja T. Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.E.H.); (T.B.H.); (M.T.L.)
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (A.L.); (H.M.); (J.S.)
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (A.L.); (H.M.); (J.S.)
| | - Ralph Wendt
- Department of Nephrology, Hospital St. Georg, 04129 Leipzig, Germany;
| | - Joachim Beige
- Department of Nephrology, Hospital St. Georg, 04129 Leipzig, Germany;
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, 04129 Leipzig, Germany
- Department of Internal Medicine II, Martin-Luther-University Halle/Wittenberg, 06108 Halle (Saale), Germany
| |
Collapse
|
19
|
Fu Y, Song C, Qin Y, Zheng T, Zhou X, Zhao X, Zou J, Huang B. Clinical value of serum MMP-3 in chronic kidney disease. Clin Chim Acta 2024; 553:117725. [PMID: 38128817 DOI: 10.1016/j.cca.2023.117725] [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: 09/02/2023] [Revised: 11/26/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is defined as the progressive deterioration of renal parenchyma and decline in renal unit function. In the early stages of CKD(G1 + G2), symptoms are usually not obvious and cannot be effectively recognized on the basis of available clinical markers. Progression to the middle and late stages of CKD results in severe kidney damage with multiple complications causing adverse outcomes, including death. Therefore, the early diagnosis and monitoring of CKD is critical. Matrix metalloproteinase-3 (MMP-3), an extracellular matrix-degrading enzyme, plays an important role in kidney diseases. However, the clinical significance of serum MMP-3 levels in CKD has rarely been reported. METHODS We quantified the serum MMP-3 levels of 237 patients with CKD and 96 healthy individuals by using a highly sensitive time-resolved fluorescence immunoassay and analyzed differences in MMP-3 levels among the stages of CKD and the correlations of these changes with clinical indicators. RESULTS The serum MMP-3 concentrations of patients with CKD (171.76 ± 165.22 ng/mL) were significantly higher than those of healthy controls (34.05 ± 22.93 ng/mL; P < 0.0001). In CKD, serum MMP-3 levels were significantly correlated with estimated glomerular filtration rate (eGFR) (r = - 0.5804, P < 0.0001), serum creatinine (CREA) (r = 0.5823, P < 0.0001), blood urea nitrogen (BUN) (r = 0.6106, P < 0.0001), and protein-to-creatinine ratio (r = 0.4992, P < 0.0001). Randomized forest analysis finds CREA, BUN, and MMP-3 most significant influences on CKD disease severity. The critical value of MMP-3 concentration of 40.39 ng/mL combined with eGFR was effective in diagnosing positive patients in the early (G1 + G2) stage of CKD and showed a positivity rate of 73.45 %. Moreover, in the early stages of CKD, patients with CKD who had serum MMP-3 concentration > 100 ng/mL had more severe renal impairment and inflammation than those with CKD who have lower serum MMP-3 concentrations. CONCLUSION Elevated serum MMP-3 levels are correlated with decreased kidney function in CKD progression, and patients with concomitant inflammation may express high levels of serum MMP-3. Serum MMP-3 may assist eGFR in improving the diagnosis of patients with early CKD.
Collapse
Affiliation(s)
- Yulin Fu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Cheng Song
- The Taihu Sanatorium of Jiangsu Province (The Taihu Rehabilitation Hospital of Jiangsu Province), Wuxi, Jiangsu 214086, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Tianyu Zheng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xueqin Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jian Zou
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
| |
Collapse
|
20
|
Claudel SE, Waikar SS, Schmidt IM, Vasan RS, Verma A. The relationship between low levels of albuminuria and cardiovascular mortality among apparently healthy adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.21.23300378. [PMID: 38196576 PMCID: PMC10775339 DOI: 10.1101/2023.12.21.23300378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background Albuminuria is associated with cardiovascular events among adults with underlying cardiovascular disease and diabetes, even at low levels of urinary albumin excretion. We hypothesized that low levels of albuminuria in the 'normal' range (urinary albumin-to-creatine ratio (UACR) <30 mg/g) are associated with cardiovascular death among apparently healthy adults. Methods We studied adults who participated in the 1999-2014 National Health and Nutrition Examination Survey. We excluded participants with baseline cardiovascular disease, hypertension, diabetes, estimated glomerular filtration rate (eGFR) <60ml/min/1.73m2, those who were currently pregnant, and those who had received dialysis in the last year. After excluding these conditions, only 5.0% of the remaining population had UACR ≥30 mg/g (N=873) and were excluded. The final sample size was 16,247. We assessed the relationship between UACR and cardiovascular and all-cause mortality using multivariable-adjusted Cox proportional hazards models. Models were adjusted for age, sex, race or ethnicity, smoking status, systolic blood pressure, hemoglobin A1c, total cholesterol, health insurance, food insecurity, serum albumin, body mass index, use of statins, and eGFR. Results Mean age was 38.9 years (SD 13.6) and 53.7% were women. The median length of follow-up was 12.2 years. In multivariable-adjusted models, each doubling of UACR (within the <30 mg/g range) was associated with a 36% higher risk of cardiovascular death [HR 1.36 (95% confidence interval (CI) 1.11-1.65)] and a 28% higher risk of all-cause mortality [HR 1.28 (95%CI 1.17-1.41)]. The highest tertile of UACR (7.1-29.9 mg/g) was associated with an 87% higher risk of cardiovascular death [HR 1.87 (95%CI 1.20-2.92)] and 59% higher risk of all-cause mortality [HR 1.59 (95%CI 1.28-1.96)], compared with the lowest tertile (< 4.3 mg/g). Conclusions In a nationally representative sample of relatively healthy community-dwelling adults, higher levels of albuminuria in the conventionally "normal" range <30 mg/g in healthy individuals are associated with greater mortality. Overall, our findings contribute to the growing body of evidence on the existence of a risk gradient across all levels of albuminuria, even in the so-called normal range.
Collapse
Affiliation(s)
- Sophie E Claudel
- Department of Internal Medicine, Boston Medical Center, Boston, MA, USA
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Department of Medicine, Boston Medical Center, Boston, MA, USA
| | - Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston Medical Center, Boston, MA, USA
- Hamburg Center for Kidney Health, University Medical Center Hamburg, Hamburg, Germany
| | - Ramachandran S. Vasan
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- University of Texas School of Public Health, and University of Texas Health Science Center in San Antonio, TX, USA
| | - Ashish Verma
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Department of Medicine, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
21
|
Mussap M, Sortino M, Monteverde E, Tomaiuolo R, Banfi G, Locatelli M, Carobene A. Review on adherence of the literature to official recommendations on albuminuria harmonization and standardization. Clin Chem Lab Med 2023; 61:2076-2083. [PMID: 37325992 DOI: 10.1515/cclm-2023-0408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Albuminuria standardization is a key issue to produce reliable and equivalent results between laboratories. We investigated whether official recommendations on albuminuria harmonization are followed in the literature. The PubMed database was searched from June 1 to September 26, 2021. The search terms included urine albumin, urine albumin-to-creatinine ratio (uACR), and albuminuria. A total of 159 articles were considered eligible; 50.9 % reported the type of urine collection. Specifically, 58.1 % collected a random spot urine specimen, 21 % collected a first morning void, and 6.2 % collected a 24-h specimen. Overall, 15 % of articles reported data on sample shipping, storage, and centrifugation and 13.3 % mentioned the preanalytical phase without any data on albuminuria. The method for albuminuria was properly described in 31.4 % of articles; of these, 54.9 % used immunological methods, and 8.9 % contained errors or missing data. Most articles (76.7 %) expressed test results as albuminuria-to-creatininuria ratio. Different decision levels were utilized in 130 articles; of these, 36 % used a decision level of ≤30 mg/g creatininuria and 23.7 % used three decision levels (≤30, 30-300, and ≥300 mg/g). The failure to follow guidelines on albuminuria harmonization was mainly found in the preanalytical phase. The poor awareness of the importance of preanalytical steps on test result may be a possible explanation.
Collapse
Affiliation(s)
- Michele Mussap
- Molecular Unit, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | | | | | | | - Giuseppe Banfi
- University Vita-Salute San Raffaele, Milan, Italy
- IRCCS Galeazzi-Sant'Ambrogio Hospital, Milan, Italy
| | - Massimo Locatelli
- Laboratory Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Carobene
- Laboratory Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Møller AL, Thöni S, Keller F, Sharifli S, Rasmussen DGK, Genovese F, Karsdal MA, Mayer G. Combination Therapy of RAS Inhibition and SGLT2 Inhibitors Decreases Levels of Endotrophin in Persons with Type 2 Diabetes. Biomedicines 2023; 11:3084. [PMID: 38002084 PMCID: PMC10669010 DOI: 10.3390/biomedicines11113084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
We investigated for the first time the effect of combination therapy of renin-angiotensin system inhibition (RASi) and sodium-glucose co-transporter-2 inhibitors (SGLT2is) on endotrophin (ETP), a pro-fibrotic signaling molecule reflecting collagen type VI formation, measured in the plasma of persons with type 2 diabetes (T2D). ETP was measured using the PRO-C6 ELISA in 294 individuals from the "Drug combinations for rewriting trajectories of renal pathologies in type 2 diabetes" (DC-ren) project. In the DC-ren study, kidney disease progression was defined as a >10% decline in the estimated glomerular filtration rate (eGFR) to an eGFR < 60 mL/min/1.73 m2. Among the investigated circulating markers, ETP was the most significant predictor of future eGFR. Combination therapy of RASi and SGLT2is led to a significant reduction in ETP levels compared to RASi monotherapy (p for slope difference = 0.002). Higher levels of baseline plasma ETP were associated with a significantly increased risk of kidney disease progression (p = 0.007). In conclusion, plasma ETP identified individuals at higher risk of kidney disease progression. The observed decreased levels of plasma ETP with combination therapy of RASi and SGLT2is in persons with T2D may reflect a reduced risk of kidney disease progression following treatment with SGLT2is.
Collapse
Affiliation(s)
- Alexandra Louise Møller
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stefanie Thöni
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Felix Keller
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Samir Sharifli
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, 6020 Innsbruck, Austria
| | | | | | | | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
23
|
Rotgers E, Lamberg T, Pihlajamaa T, Pussinen C, Joutsi-Korhonen L, Kouri TT. Verifying measurements on Siemens Atellica® instruments using clinically acceptable analytical performance specifications. Scand J Clin Lab Invest 2023; 83:408-416. [PMID: 37671917 DOI: 10.1080/00365513.2023.2253422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
Measurements on clinical chemistry analysers must be verified to demonstrate applicability to their intended clinical use. We verified the performance of measurements on the Siemens Atellica® Solution chemistry analysers against the clinically acceptable analytical performance specifications, CAAPS, including the component of intra-individual biological variation, CVI. The relative standard uncertainty of measurement, i.e. analytical variation, CVA, was estimated for six example measurands, haemoglobin A1c in whole blood (B-HbA1c), albumin in urine (U-Alb), and the following measurands in plasma: sodium (P-Na), pancreatic amylase (P-AmylP), low-density lipoprotein cholesterol (P-LDL-C), and creatinine (P-Crea). Experimental CVA was calculated from single-instrument imprecision using control samples, variation between measurements on parallel instruments, and estimation of bias with pooled patient specimens. Each obtained CVA was compared with previously developed CAAPS. The calculated CVA was 1.4% for B-HbA1c (CAAPS 1.9% for single diagnostic testing, CAAPS 2.0% for monitoring after duplicate tests; IFCC units), 10.9% for U-Alb (CAAPS 44.9%), 1.2% for P-Na (CAAPS 0.6%, after triplicate testing 1.5%), 8.2% for P-AmylP (CAAPS 22.9%). The CVA was 4.9% for P-LDL-C (CAAPS for cardiovascular risk stratification 4.9% after four replicates), and 4.2% for P-Crea (CAAPS 8.0%). Three of the six measurands fulfilled the estimated clinical need. Results from P-Na measurements indicate a general need for improving the P-Na assays for emergency patients. It is necessary to consider CVI when creating diagnostic targets for laboratory tests, as emphasised by the CAAPS estimates of B-HbA1c and P-LDL-C.
Collapse
Affiliation(s)
- Emmi Rotgers
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| | - Tea Lamberg
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| | - Tero Pihlajamaa
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| | - Christel Pussinen
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| | - Timo T Kouri
- Department of Clinical Chemistry, University of Helsinki, and HUS Diagnostic Center, Helsinki and Uusimaa Hospital District, Helsinki, Finland
| |
Collapse
|
24
|
Chan KW, Kwong ASK, Tan KCB, Lui SL, Chan GC, Ip TP, Yiu WH, Cowling BJ, Taam Wong V, Lao L, Feng Y, Lai KN, Tang SC. Add-on Rehmannia-6-Based Chinese Medicine in Type 2 Diabetes and CKD: A Multicenter Randomized Controlled Trial. Clin J Am Soc Nephrol 2023; 18:1163-1174. [PMID: 37307005 PMCID: PMC10564374 DOI: 10.2215/cjn.0000000000000199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/03/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Diabetes is the leading cause of CKD and kidney failure. We assessed the real-world effectiveness of Rehmannia-6-based Chinese medicine treatment, the most used Chinese medicine formulation, on the change in eGFR and albuminuria in patients with diabetes and CKD with severely increased albuminuria. METHODS In this randomized, assessor-blind, standard care-controlled, parallel, multicenter trial, 148 adult patients from outpatient clinics with type 2 diabetes, an eGFR of 30-90 ml/min per 1.73 m 2 , and a urine albumin-to-creatinine ratio (UACR) of 300-5000 mg/g were randomized 1:1 to a 48-week add-on protocolized Chinese medicine treatment program (using Rehmannia-6-based formulations in the granule form taken orally) or standard care alone. Primary outcomes were the slope of change in eGFR and UACR between baseline and end point (48 weeks after randomization) in the intention-to-treat population. Secondary outcomes included safety and the change in biochemistry, biomarkers, and concomitant drug use. RESULTS The mean age, eGFR, and UACR were 65 years, 56.7 ml/min per 1.73 m 2 , and 753 mg/g, respectively. Ninety-five percent ( n =141) of end point primary outcome measures were retrievable. For eGFR, the estimated slope of change was -2.0 (95% confidence interval [CI], -0.1 to -3.9) and -4.7 (95% CI, -2.9 to -6.5) ml/min per 1.73 m 2 in participants treated with add-on Chinese medicine or standard care alone, resulting in a 2.7 ml/min per 1.73 m 2 per year (95% CI, 0.1 to 5.3; P = 0.04) less decline with Chinese medicine. For UACR, the estimated proportion in the slope of change was 0.88 (95% CI, 0.75 to 1.02) and 0.99 (95% CI, 0.85 to 1.14) in participants treated with add-on Chinese medicine or standard care alone, respectively. The intergroup proportional difference (0.89, 11% slower increment in add-on Chinese medicine, 95% CI, 0.72 to 1.10; P = 0.28) did not reach statistical significance. Eighty-five adverse events were recorded from 50 participants (add-on Chinese medicine versus control: 22 [31%] versus 28 [36%]). CONCLUSIONS Rehmannia-6-based Chinese medicine treatment stabilized eGFR on top of standard care alone after 48 weeks in patients with type 2 diabetes, stage 2-3 CKD, and severely increased albuminuria. CLINICAL TRIAL REGISTRY Semi-individualized Chinese Medicine Treatment as an Adjuvant Management for Diabetic Nephropathy (SCHEMATIC), NCT02488252 .
Collapse
Affiliation(s)
- Kam Wa Chan
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Alfred Siu Kei Kwong
- Department of Family Medicine and Primary Healthcare, Hong Kong West Cluster, Hospital Authority, Hong Kong SAR, China
| | - Kathryn Choon Beng Tan
- Division of Endocrinology & Metabolism, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sing Leung Lui
- Department of Medicine, Tung Wah Hospital, Hong Kong SAR, China
| | - Gary C.W. Chan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China
| | - Tai Pang Ip
- Department of Medicine, Tung Wah Hospital, Hong Kong SAR, China
| | - Wai Han Yiu
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Benjamin John Cowling
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Vivian Taam Wong
- School of Chinese Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lixing Lao
- School of Chinese Medicine, The University of Hong Kong, Hong Kong SAR, China
- Virginia University of Integrative Medicine, Fairfax, Virginia
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kar Neng Lai
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sydney C.W. Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Ahmad MI, Chen LY, Singh S, Luqman-Arafath TK, Kamel H, Soliman EZ. Interrelations between albuminuria, electrocardiographic left atrial abnormality, and incident atrial fibrillation in the Multi-Ethnic Study of Atherosclerosis (MESA) cohort. Int J Cardiol 2023; 383:102-109. [PMID: 37100232 DOI: 10.1016/j.ijcard.2023.04.036] [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: 03/18/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND The objective of the study was to examine the joint associations of albuminuria and electrocardiographic left atrial abnormality (ECG-LAA) with incident atrial fibrillation (AF) and whether this relationship varies by race. METHODS This analysis included 6670 participants free of clinical cardiovascular disease (CVD), including atrial fibrillation (AF), from the Multi-Ethnic Study of Atherosclerosis. ECG-LAA was defined as P-wave terminal force in V1 [PTFV1] >5000 μV × ms. Albuminuria was defined as urine albumin-creatinine ratio (UACR) ≥30 mg/g. Incident AF events through 2015 were ascertained from hospital discharge records and study-scheduled electrocardiograms. Cox proportional hazard models were used to examine the association of "no albuminuria + no ECG-LAA (reference)", "isolated albuminuria", "isolated ECG-LAA" and "albuminuria + ECG-LAA" with incident AF. RESULTS Over a median follow-up of 13.8 years, 979 incident cases of AF occurred. In adjusted models, the concomitant presence of ECG-LAA and albuminuria was associated with a higher risk of AF than either ECG-LAA or albuminuria in isolation (HR (95% CI): 2.43 (1.65-3.58), 1.33 (1.05-1.69), and 1.55 (1.27-1.88), respectively (interaction p-value = 0.50). Effect modification by race was observed with a 4-fold greater AF risk in Black participants with albuminuria + ECG-LAA (HR (95%CI): 4.37 (2.38-8.01) but no significant association in White participants (HR (95% CI) 0.60 (0.19-1.92) respectively; (interaction p-value for race x albuminuria-ECG-LAA combination = 0.05). CONCLUSIONS Concomitant presence of ECG-LAA and albuminuria confers a higher risk of AF compared to either one in isolation with a stronger association in Blacks than Whites.
Collapse
Affiliation(s)
- Muhammad Imtiaz Ahmad
- Department of Internal Medicine, Section on Hospital Medicine, Medical College of Wisconsin, Wauwatosa, WI, United States of America.
| | - Lin Y Chen
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, United States of America
| | - Sanjay Singh
- Department of Internal Medicine, Section on Hospital Medicine, Medical College of Wisconsin, Wauwatosa, WI, United States of America
| | - T K Luqman-Arafath
- Department of Internal Medicine, Section on Hospital Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, New York, NY, United States of America
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center (EPICARE), Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| |
Collapse
|
26
|
Friedli I, Baid-Agrawal S, Unwin R, Morell A, Johansson L, Hockings PD. Magnetic Resonance Imaging in Clinical Trials of Diabetic Kidney Disease. J Clin Med 2023; 12:4625. [PMID: 37510740 PMCID: PMC10380287 DOI: 10.3390/jcm12144625] [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: 05/29/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic kidney disease (CKD) associated with diabetes mellitus (DM) (known as diabetic kidney disease, DKD) is a serious and growing healthcare problem worldwide. In DM patients, DKD is generally diagnosed based on the presence of albuminuria and a reduced glomerular filtration rate. Diagnosis rarely includes an invasive kidney biopsy, although DKD has some characteristic histological features, and kidney fibrosis and nephron loss cause disease progression that eventually ends in kidney failure. Alternative sensitive and reliable non-invasive biomarkers are needed for DKD (and CKD in general) to improve timely diagnosis and aid disease monitoring without the need for a kidney biopsy. Such biomarkers may also serve as endpoints in clinical trials of new treatments. Non-invasive magnetic resonance imaging (MRI), particularly multiparametric MRI, may achieve these goals. In this article, we review emerging data on MRI techniques and their scientific, clinical, and economic value in DKD/CKD for diagnosis, assessment of disease pathogenesis and progression, and as potential biomarkers for clinical trial use that may also increase our understanding of the efficacy and mode(s) of action of potential DKD therapeutic interventions. We also consider how multi-site MRI studies are conducted and the challenges that should be addressed to increase wider application of MRI in DKD.
Collapse
Affiliation(s)
- Iris Friedli
- Antaros Medical, BioVenture Hub, 43183 Mölndal, Sweden
| | - Seema Baid-Agrawal
- Transplant Center, Sahlgrenska University Hospital, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Robert Unwin
- AstraZeneca R&D BioPharmaceuticals, Translational Science and Experimental Medicine, Early Cardiovascular, Renal & Metabolic Diseases (CVRM), Granta Park, Cambridge CB21 6GH, UK
| | - Arvid Morell
- Antaros Medical, BioVenture Hub, 43183 Mölndal, Sweden
| | | | - Paul D Hockings
- Antaros Medical, BioVenture Hub, 43183 Mölndal, Sweden
- MedTech West, Chalmers University of Technology, 41345 Gothenburg, Sweden
| |
Collapse
|
27
|
Pierre CC, Marzinke MA, Ahmed SB, Collister D, Colón-Franco JM, Hoenig MP, Lorey T, Palevsky PM, Palmer OP, Rosas SE, Vassalotti J, Whitley CT, Greene DN. AACC/NKF Guidance Document on Improving Equity in Chronic Kidney Disease Care. J Appl Lab Med 2023:jfad022. [PMID: 37379065 DOI: 10.1093/jalm/jfad022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Kidney disease (KD) is an important health equity issue with Black, Hispanic, and socioeconomically disadvantaged individuals experiencing a disproportionate disease burden. Prior to 2021, the commonly used estimated glomerular filtration rate (eGFR) equations incorporated coefficients for Black race that conferred higher GFR estimates for Black individuals compared to non-Black individuals of the same sex, age, and blood creatinine concentration. With a recognition that race does not delineate distinct biological categories, a joint task force of the National Kidney Foundation and the American Society of Nephrology recommended the adoption of the CKD-EPI 2021 race-agnostic equations. CONTENT This document provides guidance on implementation of the CKD-EPI 2021 equations. It describes recommendations for KD biomarker testing, and opportunities for collaboration between clinical laboratories and providers to improve KD detection in high-risk populations. Further, the document provides guidance on the use of cystatin C, and eGFR reporting and interpretation in gender-diverse populations. SUMMARY Implementation of the CKD-EPI 2021 eGFR equations represents progress toward health equity in the management of KD. Ongoing efforts by multidisciplinary teams, including clinical laboratorians, should focus on improved disease detection in clinically and socially high-risk populations. Routine use of cystatin C is recommended to improve the accuracy of eGFR, particularly in patients whose blood creatinine concentrations are confounded by processes other than glomerular filtration. When managing gender-diverse individuals, eGFR should be calculated and reported with both male and female coefficients. Gender-diverse individuals can benefit from a more holistic management approach, particularly at important clinical decision points.
Collapse
Affiliation(s)
- Christina C Pierre
- Department of Pathology and Laboratory Medicine, Penn Medicine Lancaster General Hospital, Lancaster, PA, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark A Marzinke
- Departments of Pathology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sofia B Ahmed
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David Collister
- Division of Nephrology, University of Alberta, Edmonton, AB, Canada
- Population Health Research Institute, Hamilton, ON, Canada
| | | | - Melanie P Hoenig
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Division of Nephrology and Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Thomas Lorey
- Kaiser Permanante, The Permanante Medical Group Regional Laboratory, Berkeley, CA, United States
| | - Paul M Palevsky
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Kidney Medicine Program and Kidney Medicine Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States
- The National Kidney Foundation, Inc., New York, NY, United States
| | - Octavia Peck Palmer
- Departments of Pathology, Critical Care Medicine, and Clinical and Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Sylvia E Rosas
- The National Kidney Foundation, Inc., New York, NY, United States
- Kidney and Hypertension Unit, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Joseph Vassalotti
- The National Kidney Foundation, Inc., New York, NY, United States
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cameron T Whitley
- Department of Sociology, Western Washington University, Bellingham, WA, United States
| | - Dina N Greene
- Department of Laboratory Medicine and Pathology, University of Washington Medicine, Seattle, WA, United States
- LetsGetChecked Laboratories, Monrovia, CA, United States
| |
Collapse
|
28
|
Sandokji I, Xu Y, Denburg M, Furth S, Abraham AG, Greenberg JH. Current and Novel Biomarkers of Progression Risk in Children with Chronic Kidney Disease. Nephron Clin Pract 2023; 148:1-10. [PMID: 37232009 PMCID: PMC10840447 DOI: 10.1159/000530918] [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: 09/15/2022] [Accepted: 02/18/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Due to the complexity of chronic kidney disease (CKD) pathophysiology, biomarkers representing different mechanistic pathways have been targeted for the study and development of novel biomarkers. The discovery of clinically useful CKD biomarkers would allow for the identification of those children at the highest risk of kidney function decline for timely interventions and enrollment in clinical trials. SUMMARY Glomerular filtration rate and proteinuria are traditional biomarkers to classify and prognosticate CKD progression in clinical practice but have several limitations. Over the recent decades, novel biomarkers have been identified from blood or urine with metabolomic screening studies, proteomic screening studies, and an improved knowledge of CKD pathophysiology. This review highlights promising biomarkers associated with the progression of CKD that could potentially serve as future prognostic markers in children with CKD. KEY MESSAGES Further studies are needed in children with CKD to validate putative biomarkers, particularly candidate proteins and metabolites, for improving clinical management.
Collapse
Affiliation(s)
- Ibrahim Sandokji
- Department of Pediatrics, Taibah University College of Medicine, Medina, Saudi Arabia,
| | - Yunwen Xu
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Denburg
- Division of Nephrology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Furth
- Division of Nephrology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alison G Abraham
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jason H Greenberg
- Department of Pediatrics, Section of Nephrology, Clinical and Translational Research Accelerator, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
29
|
Kukkar D, Chhillar M, Kim KH. Application of SERS-based nanobiosensors to metabolite biomarkers of CKD. Biosens Bioelectron 2023; 232:115311. [PMID: 37086564 DOI: 10.1016/j.bios.2023.115311] [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: 03/06/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 04/24/2023]
Abstract
A clinical diagnosis of chronic kidney disease (CKD) is commonly achieved by estimating the serum levels of urea and creatinine (CR). Given the limitations of the conventional diagnostic assays, it is imperative to seek alternative, economical strategies for the detection of CKD-specific biomarkers with high specificity and selectivity. In this respect, surface-enhanced Raman spectroscopy (SERS) can be regarded as an ideal choice. SERS signals can be greatly amplified by noble metal nanoparticles (e.g., gold nanoparticles (GNPs)) of numerous sizes, shapes, and configurations to help achieve ultra-sensitive single molecule-level detection at 10-15 M (up to 10 orders of magnitude more sensitive than fluorescence-based detection). The irregular geometry of GNPs with spike-like tips, dimers, and aggregates with small nanogaps (i.e., due to plasmon coupling such as Raman hot spots) play a pivotal role in enhancing the specificity and sensitivity of SERS. This review critically outlines the performance of SERS-based biosensors in the ultrasensitive detection of CKD biomarkers in various body fluids in terms of basic quality assurance parameters (e.g., limit of detection, figure of merit, enhancement factor, and stability of the biosensor). Moreover, the challenges and perspectives are described with respect to the expansion of such sensing techniques in practical clinical settings.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India
| | - Monika Chhillar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul, 04763, South Korea.
| |
Collapse
|
30
|
Palmer BF. Change in albuminuria as a surrogate endpoint for cardiovascular and renal outcomes in patients with diabetes. Diabetes Obes Metab 2023; 25:1434-1443. [PMID: 36809555 DOI: 10.1111/dom.15030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/10/2023] [Accepted: 02/19/2023] [Indexed: 02/23/2023]
Abstract
For the purpose of predicting clinical outcomes in patients with diabetes and chronic kidney disease, change in albuminuria is a good candidate to be a surrogate marker for future cardiovascular events and progression of kidney disease. Spot urine albumin/creatinine ratio is convenient and recognized as a viable alternative to 24-h albumin, with some limitations. Although there is sufficient evidence to validate its use in clinical trials as a surrogate endpoint for renal outcomes, this is not yet the case for cardiovascular outcomes. While change in albuminuria as a primary or secondary endpoint is trial-specific, its use should be encouraged, nonetheless.
Collapse
Affiliation(s)
- Biff F Palmer
- Department of Internal Medicine, Divison of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
31
|
Rotgers E, Linko S, Theodorsson E, Kouri TT. Clinical decision limits as criteria for setting analytical performance specifications for laboratory tests. Clin Chim Acta 2023; 540:117233. [PMID: 36693582 DOI: 10.1016/j.cca.2023.117233] [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: 09/02/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND The biological (CVI), preanalytical (CVPRE), and analytical variation (CVA) are inherent to clinical laboratory testing and consequently, interpretation of clinical test results. METHODS The sum of the CVI, CVPRE, and CVA, called diagnostic variation (CVD), was used to derive clinically acceptable analytical performance specifications (CAAPS) for clinical chemistry measurands. The reference change concept was applied to clinically significant differences (CD) between two measurements, with the formula CD = z*√2* CVD. CD for six measurands were sought from international guidelines. The CAAPS were calculated by subtracting variances of CVI and CVPRE from CVD. Modified formulae were applied to consider statistical power (1-β) and repeated measurements. RESULTS The obtained CAAPS were 44.9% for urine albumin, 0.6% for plasma sodium, 22.9% for plasma pancreatic amylase, and 8.0% for plasma creatinine (z = 3, α = 2.5%, 1-β = 85%). For blood HbA1c and plasma low-density lipoprotein cholesterol, replicate measurements were necessary to reach CAAPS for patient monitoring. The derived CAAPS were compared with analytical performance specifications, APS, based on biological variation. CONCLUSIONS The CAAPS models pose a new tool for assessing APS in a clinical laboratory. Their usability depends on the relevance of CD limits, required statistical power and the feasibility of repeated measurements.
Collapse
Affiliation(s)
- Emmi Rotgers
- Department of Clinical Chemistry, University of Helsinki, and HUSLAB, HUS Diagnostic Center, Helsinki University Hospital, FIN-00029 Helsinki, Finland
| | | | - Elvar Theodorsson
- Department of Biomedical and Clinical Sciences, Division of Clinical Chemistry and Pharmacology, Linkoping University, SE-58183 Linkoping, Sweden
| | - Timo T Kouri
- Department of Clinical Chemistry, University of Helsinki, and HUSLAB, HUS Diagnostic Center, Helsinki University Hospital, FIN-00029 Helsinki, Finland.
| |
Collapse
|
32
|
Khan MS, Shahid I, Anker SD, Fonarow GC, Fudim M, Hall ME, Hernandez A, Morris AA, Shafi T, Weir MR, Zannad F, Bakris GL, Butler J. Albuminuria and Heart Failure: JACC State-of-the-Art Review. J Am Coll Cardiol 2023; 81:270-282. [PMID: 36653095 DOI: 10.1016/j.jacc.2022.10.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/03/2022] [Indexed: 01/18/2023]
Abstract
Although chronic kidney disease is characterized by low glomerular filtration rate (GFR) or albuminuria, estimated GFR (eGFR) is more widely utilized as a marker of risk profile in cardiovascular diseases, including heart failure (HF). The presence and magnitude of albuminuria confers a strong prognostic association in forecasting risk of incident HF as well as its progression, irrespective of eGFR. Despite the high prevalence of albuminuria in HF, whether it adds incremental prognostic information in clinical practice and serves as an independent risk marker, and whether there are any therapeutic implications of assessing albuminuria in patients with HF is less well-established. In this narrative review, we assess the potential role of albuminuria in risk profiling for development and progression of HF, strengths and limitations of utilizing albuminuria as a risk marker, its ability to serve in HF risk prediction models, and the implications of adopting albuminuria as an effective parameter in cardiovascular trials and practice.
Collapse
Affiliation(s)
- Muhammad Shahzeb Khan
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/ShahzebkhanMD
| | - Izza Shahid
- Division of Preventive Cardiology, Department of Cardiology, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Stefan D Anker
- Department of Cardiology (CVK), Charité-Universitätsmedizin Berlin; Berlin Institute of Health Center for Regenerative Therapies, German Center for Cardiovascular Research, Berlin, Germany
| | - Gregg C Fonarow
- Division of Cardiology, University of California, Los Angeles, California, USA
| | - Marat Fudim
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Adrian Hernandez
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Alanna A Morris
- Division of Cardiology, Emory University, Atlanta, Georgia, USA
| | - Tariq Shafi
- Division of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Matthew R Weir
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Faiez Zannad
- Université de Lorraine, CIC Inserm, CHRU, Nancy, France
| | - George L Bakris
- Department of Medicine, University of Chicago Medicine, Chicago, Illinois, USA
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA; Baylor Scott and White Research Institute, Dallas, Texas, USA.
| |
Collapse
|
33
|
Thöni S, Keller F, Denicolò S, Buchwinkler L, Mayer G. Biological variation and reference change value of the estimated glomerular filtration rate in humans: A systematic review and meta-analysis. Front Med (Lausanne) 2022; 9:1009358. [PMID: 36275823 PMCID: PMC9583397 DOI: 10.3389/fmed.2022.1009358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Background Knowledge of the biological variation of serum or plasma creatinine (Cr) and the estimated glomerular filtration rate (eGFR) is important for understanding disease dynamics in Chronic Kidney Disease (CKD). The aim of our study was to determine the magnitude of random fluctuation of eGFR by determining its reference change value (RCV). Methods We performed a systematic review and meta-analysis of studies on biological variation of Cr. Relevant studies were identified by systematic literature search on PubMed. Additional studies were retrieved from the European Federation of Clinical Chemistry and Laboratory Medicine (EFLM) Biological Variation Database. Random-effects meta-analysis was conducted to derive an overall estimate of intra-individual variation of creatinine (CVICr). Based on our estimate of CVICr and RCV for Cr, the RCV for the eGFR was determined. Results Among identified studies, 37 met our inclusion criteria. Meta-analysis of all studies yielded a CVICr of 5.2% (95% confidence interval [CI] 4.6–5.8%), however high between-study heterogeneity (I2 = 82.3%) was found. Exclusion of outliers led to a significant reduction of heterogeneity while still including 85% of all studies and resulted in a slightly lower CVICr of 5.0% (95% CI 4.7–5.4%). Assuming an analytical variation of CVA 1.1%, we found an overall RCV for eGFR of ±16.5%. After exclusion of outlier studies, we found a minimum conservative RCV for eGFR of ±12.5%. Conclusion The RCV of the eGFR represents a valuable tool for clinicians to discern true changes in kidney function from random fluctuation.
Collapse
|
34
|
Krupka E, Curtis S, Ferguson T, Whitlock R, Askin N, Millar AC, Dahl M, Fung R, Ahmed SB, Tangri N, Walsh M, Collister D. The Effect of Gender-Affirming Hormone Therapy on Measures of Kidney Function: A Systematic Review and Meta-Analysis. Clin J Am Soc Nephrol 2022; 17:1305-1315. [PMID: 35973728 PMCID: PMC9625103 DOI: 10.2215/cjn.01890222] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/27/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND OBJECTIVES Gender-affirming hormone therapy modifies body composition and lean muscle mass in transgender persons. We sought to characterize the change in serum creatinine, other kidney function biomarkers, and GFR in transgender persons initiating masculinizing and feminizing gender-affirming hormone therapy. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We searched PubMed, EMBASE, the Cochrane Library, and ClinicalTrials.gov from inception to September 16, 2020 for randomized controlled trials, observational studies, and case series that evaluated the change in serum creatinine, other kidney function biomarkers, and GFR before and after the initiation of gender-affirming hormone therapy in adult transgender persons. Two reviewers independently screened and abstracted data, and disagreements were resolved by a third reviewer. A random effects meta-analysis was performed to determine the change in outcomes over follow-up of 3, 6, and 12 months. RESULTS Of the 4758 eligible studies, 26 met the inclusion criteria, including nine studies that recruited 488 transgender men and 593 women in which data were meta-analyzed. There was heterogeneity in study design, populations, gender-affirming hormone therapy routes, and dosing. At 12 months after initiating gender-affirming hormone therapy, serum creatinine increased by 0.15 mg/dl (95% confidence interval, 0.00 to 0.29) in 370 transgender men and decreased by -0.05 mg/dl (95% confidence interval, -0.16 to 0.05) in 361 transgender women. No study reported the effect of gender-affirming hormone therapy on albuminuria, proteinuria, cystatin C, or measured GFR. CONCLUSIONS Gender-affirming hormone therapy increases serum creatinine in transgender men and does not affect serum creatinine in transgender women. The effect on gender-affirming hormone therapy on other kidney function biomarkers and measured GFR is unknown. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Change in Kidney Function Biomarkers in Transgender Persons on Gender Affirmation Hormone Therapy-A Systematic Review and Meta-Analysis, CRD42020214248.
Collapse
Affiliation(s)
- Emily Krupka
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Sarah Curtis
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Thomas Ferguson
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Reid Whitlock
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Nicole Askin
- University of Manitoba, Winnipeg, Manitoba, Canada
| | - Adam C. Millar
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marshall Dahl
- Division of Endocrinology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymond Fung
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Michael Garron Hospital, Toronto, Ontario, Canada
| | - Sofia B. Ahmed
- Division of Nephrology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Navdeep Tangri
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
- Max Rady Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael Walsh
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences/McMaster University, Hamilton, Ontario, Canada
| | - David Collister
- Chronic Disease Innovation Centre, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
- Max Rady Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Population Health Research Institute, Hamilton Health Sciences/McMaster University, Hamilton, Ontario, Canada
- Section of Nephrology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Liu C, Debnath N, Mosoyan G, Chauhan K, Vasquez-Rios G, Soudant C, Menez S, Parikh CR, Coca SG. Systematic Review and Meta-Analysis of Plasma and Urine Biomarkers for CKD Outcomes. J Am Soc Nephrol 2022; 33:1657-1672. [PMID: 35858701 PMCID: PMC9529190 DOI: 10.1681/asn.2022010098] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/02/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Sensitive and specific biomarkers are needed to provide better biologic insight into the risk of incident and progressive CKD. However, studies have been limited by sample size and design heterogeneity. METHODS In this assessment of the prognostic value of preclinical plasma and urine biomarkers for CKD outcomes, we searched Embase (Ovid), MEDLINE ALL (Ovid), and Scopus up to November 30, 2020, for studies exploring the association between baseline kidney biomarkers and CKD outcomes (incident CKD, CKD progression, or incident ESKD). We used random-effects meta-analysis. RESULTS After screening 26,456 abstracts and 352 full-text articles, we included 129 studies in the meta-analysis for the most frequently studied plasma biomarkers (TNFR1, FGF23, TNFR2, KIM-1, suPAR, and others) and urine biomarkers (KIM-1, NGAL, and others). For the most frequently studied plasma biomarkers, pooled RRs for CKD outcomes were 2.17 (95% confidence interval [95% CI], 1.91 to 2.47) for TNFR1 (31 studies); 1.21 (95% CI, 1.15 to 1.28) for FGF-23 (30 studies); 2.07 (95% CI, 1.82 to 2.34) for TNFR2 (23 studies); 1.51 (95% CI, 1.38 to 1.66) for KIM-1 (18 studies); and 1.42 (95% CI, 1.30 to 1.55) for suPAR (12 studies). For the most frequently studied urine biomarkers, pooled RRs were 1.10 (95% CI, 1.05 to 1.16) for KIM-1 (19 studies) and 1.12 (95% CI, 1.06 to 1.19) for NGAL (19 studies). CONCLUSIONS Studies of preclinical biomarkers for CKD outcomes have considerable heterogeneity across study cohorts and designs, limiting comparisons of prognostic performance across studies. Plasma TNFR1, FGF23, TNFR2, KIM-1, and suPAR were among the most frequently investigated in the setting of CKD outcomes.
Collapse
Affiliation(s)
- Caroline Liu
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Neha Debnath
- Department of Medicine, Icahn School of Medicine at Mount Sinai (Morningside/West), New York, New York
| | - Gohar Mosoyan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kinsuk Chauhan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - George Vasquez-Rios
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Celine Soudant
- Division of Technology, Memorial Sloan Kettering Cancer Center Medical Library, New York, New York
| | - Steve Menez
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chirag R. Parikh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven G. Coca
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
36
|
Chao H, He Y, Wang Q, Bai Y, Avolio A, Deng X, Zuo J. Comparison of Influence of Blood Pressure and Carotid-Femoral Pulse Wave Velocity on Target Organ Damage in Hypertension. Front Cardiovasc Med 2022; 9:934747. [PMID: 35865385 PMCID: PMC9294321 DOI: 10.3389/fcvm.2022.934747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Assessment of target organ damage (TOD) is an important part of the diagnosis and evaluation of hypertension. Carotid-femoral pulse wave velocity (cf-PWV) is considered to be the gold-standard for noninvasive arterial stiffness assessment. This study aims to analyze the risk of TOD in people with different phenotypes of peripheral blood pressure and cf-PWV. Methods The study cohort was recruited from December 2017 to September 2021 at Ruijin Hospital in Shanghai. It was divided into 4 groups according to peripheral blood pressure (pBP) and cf-PWV. TOD was assessed as carotid intima-media thickness (CIMT), chronic kidney disease (CKD), urinary albumin-creatinine ratio (ACR), estimated glomerular filtration rate (eGFR) and left ventricular mass index (LVMI). Results A total of 1,257 subjects (mean age 53.13 ± 12.65 years, 64.2% males) was recruited. Age, body mass index (BMI) and fasting blood glucose (FBG), as well as peripheral systolic blood pressure (pSBP), peripheral diastolic blood pressure (pDBP), peripheral pulse pressure (pPP) were significantly different in the four groups (P < 0.01). eGFR, ACR, LVMI and CIMT were significantly different among different groups (P < 0.01). The risk of ACR abnormality was significantly higher in the group with elevated pBP (P = 0.005, OR 2.264, 95%CI 1.277–4.016; and in the group with elevated pBP and cf-PWV (P = 0.003, OR 1.482, 95%CI 1.144–1.920), while left ventricular hypertrophy (LVH) was significantly higher in the group with elevated cf-PWV (P = 0.002, OR 1.868, 95%CI 1.249–2.793). Conclusion Different profiles based on the status of PBP and cf-PWV associated with different TOD. Individuals with higher pBP have an increased risk of ACR abnormality, while individuals with only cf-PWV elevated have a higher risk of LVH.
Collapse
Affiliation(s)
- Huijuan Chao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan He
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaya Bai
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alberto Avolio
- Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Xueqin Deng
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Xueqin Deng
| | - Junli Zuo
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- *Correspondence: Junli Zuo
| |
Collapse
|
37
|
Waijer SW, Provenzano M, Mulder S, Rossing P, Persson F, Perkovic V, Heerspink HJL. Impact of random variation in albuminuria and estimated glomerular filtration rate on patient enrolment and duration of clinical trials in nephrology. Diabetes Obes Metab 2022; 24:983-990. [PMID: 35112455 PMCID: PMC9306498 DOI: 10.1111/dom.14660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 01/31/2022] [Indexed: 11/29/2022]
Abstract
AIM To test whether a screening approach with more flexible urinary albumin creatinine ratio (UACR) and estimated glomerular filtration rate (eGFR) thresholds would decrease screen failure rate without negatively impacting on the event rate and overall study duration. METHODS We performed a post-hoc analysis of the ALTITUDE trial. We selected participants randomized to placebo with a UACR of >300 mg/g and an eGFR between 30 mL/min/1.73 m2 and 60 mL/min/1.73 m2 at the first visit (pre-screening) for the trial. We then used less stringent lower UACR and higher eGFR thresholds for the following qualifying visit. For each scenario we calculated the number of eligible participants, the number of renal and cardiovascular endpoints, and the event rates. Based on this, we performed simulations for a future trial and estimated the duration of enrolment and total duration of this trial. RESULTS The base scenario consisted of 848 participants (median UACR 1239 mg/g; median eGFR 44 mL/min/1.73 m2 ). Lowering the UACR and/or raising eGFR qualification thresholds increased the number of eligible participants, decreased screen failures and resulted in only a modest decrease in renal and cardiovascular event rates. For example, relaxing the UACR criterion from 300 mg/g to 210 mg/g at the qualifying visit, increased the number of eligible patients from 848 to 923, and increased the number of renal events from 117 to 122 events. The event rate showed a moderate decrease from 5.6 (4.6-6.7) events per 100 patient-years to 5.3 (4.4-6.4) events per 100 patient-years. In simulations, lowering the UACR and raising eGFR thresholds for inclusion accelerated patient enrolment and did not increase in the overall trial duration. CONCLUSION More flexible albuminuria and eGFR-based inclusion criteria, in participants who met the inclusion criteria of a trial based on pre-screening values prior to the clinical trial, decreases screen failure rates and accelerated patient enrolment leading to more efficient trial conduct without impacting the overall trial duration.
Collapse
Affiliation(s)
- Simke W. Waijer
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
| | | | - Skander Mulder
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
- Pattern Recognition Laboratory, Faculty of Electrical Engineering, Mathematics and Computer ScienceUniversity of Technology DelftDelftThe Netherlands
| | - Peter Rossing
- Steno Diabetes Centre CopenhagenGentofteDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | | | - Vlado Perkovic
- George Institute for Global HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Centre GroningenGroningenThe Netherlands
- George Institute for Global HealthUniversity of New South WalesSydneyNew South WalesAustralia
| |
Collapse
|
38
|
Kong LR, Wei F, He DH, Zhou CQ, Li HC, Wu F, Luo Y, Luo JW, Xie QR, Peng H, Zhang Y. Biological variation in the serum and urine kidney injury markers of a healthy population measured within 24 hours. BMC Nephrol 2022; 23:195. [PMID: 35610615 PMCID: PMC9131627 DOI: 10.1186/s12882-022-02819-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND AIMS To explore the biological variation (BV) of kidney injury markers in serum and urine of healthy subjects within 24 hours to assist with interpretation of future studies using these biomarkers in the context of known BV. MATERIALS AND METHODS Serum and urine samples were collected every 4 hours (0, 4, 8, 12, 16 and 20 hours) from 31 healthy subjects within 24 hours and serum creatinine (s-Crea), serum β2-microglobin (s-β2MG), serum cystatin C (s-CYSC), serum neutrophil gelatinase-associated lipoprotein (s-NGAL), urine creatinine (u-Crea), urine β2-microglobin (u-β2MG), urine cystatin C (u-CYSC), urine neutrophil gelatinase-associated lipoprotein (u-NGAL) were measured. Outlier and variance homogeneity analyses were performed, followed by CV-ANOVA analysis on trend-corrected data (if relevant), and analytical (CVA), within-subject (CVI), and between-subject (CVG) biological variation were calculated. RESULTS The concentration of kidney injury markers in male was higher than that in female, except for u-CYSC and u-NGAL. There were no significant difference in serum and urine kidney injury markers concentration at different time points. Serum CVI was lower than urine CVI, serum CVG was higher than CVI, and urine CVG was lower than CVI. The individual index (II) of serum kidney injury markers was less than 0.6, while the II of urinary kidney injury markers was more than 1.0. CONCLUSIONS This study provides new short-term BV data for kidney injury markers in healthy subjects within 24 hours, which are of great significance in explaining other AKI / CKD studies.
Collapse
Affiliation(s)
- Li-Rui Kong
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Fei Wei
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Da-Hai He
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Chao-Qiong Zhou
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Hong-Chuan Li
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Feng Wu
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Yu Luo
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Jian-Wei Luo
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Qian-Rong Xie
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China
| | - Hai Peng
- Clinical Laboratory of Qinghai Provincial People's Hospital Xining, Xining, Qinghai, 810006, People's Republic of China
| | - Yan Zhang
- Traditional Chinese Medicine Hospital of Pidu District, No. 342, South Street, Pidu District, Chengdu, Sichuan, 611730, People's Republic of China.
| |
Collapse
|
39
|
Chen TK, Coca SG, Estrella MM, Appel LJ, Coresh J, Thiessen Philbrook H, Obeid W, Fried LF, Heerspink HJ, Ix JH, Shlipak MG, Kimmel PL, Parikh CR, Grams ME. Longitudinal TNFR1 and TNFR2 and Kidney Outcomes: Results from AASK and VA NEPHRON-D. J Am Soc Nephrol 2022; 33:996-1010. [PMID: 35314457 PMCID: PMC9063900 DOI: 10.1681/asn.2021060735] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/23/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Higher baseline levels of soluble TNF receptors (TNFR1 and TNFR2) have been associated with progressive CKD. Whether longitudinal changes in these biomarkers of inflammation are also associated with worse kidney outcomes has been less studied. METHODS We evaluated associations of longitudinal changes in TNFR1 and TNFR2 with ESKD in the African American Study of Kidney Disease and Hypertension (AASK; 38% female; 0% diabetes) and kidney function decline (first occurrence of ≥30 ml/min per 1.73 m2 or ≥50% eGFR decline if randomization eGFR ≥60 or <60 ml/min per 1.73 m2, respectively; ESKD) in the Veterans Affairs Nephropathy in Diabetes trial (VA NEPHRON-D; 99% male; 100% diabetes) using Cox models. Biomarkers were measured from samples collected at 0-, 12-, and 24-month visits for AASK (serum) and 0- and 12-month visits for VA NEPHRON-D (plasma). Biomarker slopes (AASK) were estimated using linear mixed-effects models. Covariates included sociodemographic/clinical factors, baseline biomarker level, and kidney function. RESULTS There were 129 ESKD events over a median of 7.0 years in AASK (n=418) and 118 kidney function decline events over a median of 1.5 years in VA NEPHRON-D (n=754). In AASK, each 1 SD increase in TNFR1 and TNFR2 slope was associated with 2.98- and 1.87-fold higher risks of ESKD, respectively. In VA NEPHRON-D, each 1 SD increase in TNFR1 and TNFR2 was associated with 3.20- and 1.43-fold higher risks of kidney function decline, respectively. CONCLUSIONS Among individuals with and without diabetes, longitudinal increases in TNFR1 and TNFR2 were each associated with progressive CKD, independent of initial biomarker level and kidney function.
Collapse
Affiliation(s)
- Teresa K. Chen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Steven G. Coca
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michelle M. Estrella
- Kidney Health Research Collaborative and Division of Nephrology, Department of Medicine, University of California and San Francisco VA Health Care System, San Francisco, California
| | - Lawrence J. Appel
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Wassim Obeid
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Linda F. Fried
- Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Departments of Medicine, Epidemiology, and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, and Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Michael G. Shlipak
- Kidney Health Research Collaborative and Division of Nephrology, Department of Medicine, University of California and San Francisco VA Health Care System, San Francisco, California
| | - Paul L. Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Morgan E. Grams
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
40
|
Makvandi K, Hockings PD, Jensen G, Unnerstall T, Leonhardt H, Jarl LV, Englund C, Francis S, Sundgren AK, Hulthe J, Baid-Agrawal S. Multiparametric Magnetic Resonance Imaging Allows Non-Invasive Functional and Structural Evaluation of Diabetic Kidney Disease. Clin Kidney J 2022; 15:1387-1402. [PMID: 35756740 PMCID: PMC9217657 DOI: 10.1093/ckj/sfac054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 11/15/2022] Open
Abstract
Background We sought to develop a novel non-contrast multiparametric MRI (mpMRI) protocol employing several complementary techniques in a single scan session for a comprehensive functional and structural evaluation of diabetic kidney disease (DKD). Methods In the cross-sectional part of this prospective observational study, 38 subjects ages 18‒79 years with type 2 diabetes and DKD [estimated glomerular filtration rate (eGFR) 15‒60 mL/min/1.73 m2] and 20 age- and gender-matched healthy volunteers (HVs) underwent mpMRI. Repeat mpMRI was performed on 23 DKD subjects and 10 HVs. By measured GFR (mGFR), 2 DKD subjects had GFR stage G2, 16 stage G3 and 20 stage G4/G5. A wide range of MRI biomarkers associated with kidney haemodynamics, oxygenation and macro/microstructure were evaluated. Their optimal sensitivity, specificity and repeatability to differentiate diabetic versus healthy kidneys and categorize various stages of disease as well as their correlation with mGFR/albuminuria was assessed. Results Several MRI biomarkers differentiated diabetic from healthy kidneys and distinct GFR stages (G3 versus G4/G5); mean arterial flow (MAF) was the strongest predictor (sensitivity 0.94 and 1.0, specificity 1.00 and 0.69; P = .04 and .004, respectively). Parameters significantly correlating with mGFR were specific measures of kidney haemodynamics, oxygenation, microstructure and macrostructure, with MAF being the strongest univariate predictor (r = 0.92; P < .0001). Conclusions A comprehensive and repeatable non-contrast mpMRI protocol was developed that, as a single, non-invasive tool, allows functional and structural assessment of DKD, which has the potential to provide valuable insights into underlying pathophysiology, disease progression and analysis of efficacy/mode of action of therapeutic interventions in DKD.
Collapse
Affiliation(s)
- Kianoush Makvandi
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Paul D Hockings
- Antaros Medical, Molndal, Sweden
- MedTech West, Chalmers University of Technology, Gothenburg, Sweden
| | - Gert Jensen
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tim Unnerstall
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Leonhardt
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | - Susan Francis
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Anna K Sundgren
- Late-Stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Seema Baid-Agrawal
- Department of Molecular and Clinical Medicine/Nephrology, The Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
- Transplant Center, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
41
|
Tokita J, Vega A, Sinfield C, Naik N, Rathi S, Martin S, Wang S, Amoruso L, Zabetian A, Coca SG, Nadkarni GN, Fleming F, Donovan MJ, Fields R. Real World Evidence and Clinical Utility of KidneyIntelX on Patients With Early-Stage Diabetic Kidney Disease: Interim Results on Decision Impact and Outcomes. J Prim Care Community Health 2022; 13:21501319221138196. [PMID: 36404761 PMCID: PMC9677284 DOI: 10.1177/21501319221138196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE The lack of precision to identify patients with early-stage diabetic kidney disease (DKD) at near-term risk for progressive decline in kidney function results in poor disease management often leading to kidney failure requiring unplanned dialysis. The KidneyIntelX is a multiplex, bioprognostic, immunoassay consisting of 3 plasma biomarkers and clinical variables that uses machine learning to generate a risk score for progressive decline in kidney function over 5-year in adults with early-stage DKD. Our objective was to assess the impact of KidneyIntelX on management and outcomes in a Health System in the real-world evidence (RWE) study. METHODS KidneyIntelX was introduced into a large metropolitan Health System via a population health-defined approved care pathway for patients with stages 1 to 3 DKD between [November 2020 to March 2022]. Decision impact on visit frequency, medication management, specialist referral, and selected lab values was assessed. We performed an interim analysis in patients through 6-months post-test date to evaluate the impact of risk level with clinical decision-making and outcomes. RESULTS A total of 1686 patients were enrolled in the RWE study and underwent KidneyIntelX testing and subsequent care pathway management. The median age was 68 years, 52% were female, 26% self-identified as Black, and 94% had hypertension. The median baseline eGFR was 59 ml/minute/1.73 m2, urine albumin-creatinine ratio was 69 mg/g, and HbA1c was 7.7%. After testing, a clinical encounter in the first month occurred in 13%, 43%, and 53% of low-risk, intermediate-risk, and high-risk patients, respectively and 46%, 61%, and 71% had at least 1 action taken within the first 6 months. High-risk patients were more likely to be placed on SGLT2 inhibitors (OR = 4.56; 95% CI 3.00-6.91 vs low-risk), and more likely to be referred to a specialist such as a nephrologist, endocrinologist, or dietician (OR = 2.49; 95% CI 1.53-4.01) compared to low-risk patients. CONCLUSIONS The combination of KidneyIntelX, clinical guidelines and educational support resulted in changes in clinical management by clinicians. After testing, there was an increase in visit frequency, referrals for disease management, and introduction to guideline-recommended medications. These differed by risk category, indicating an impact of KidneyIntelX risk stratification on clinical care.
Collapse
Affiliation(s)
- Joji Tokita
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aida Vega
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | | | - Nidhi Naik
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
| | - Shivani Rathi
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
| | | | - Stephanie Wang
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | - Leonard Amoruso
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | | | - Steven G. Coca
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Girish N. Nadkarni
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Michael J. Donovan
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
- Renalytix AI, Inc, New York, NY,
USA
| | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) is a silent disease, causing significant health and economic burden worldwide. It is of strong clinical value to identify novel prognostic, predictive, and pharmacodynamic biomarkers of kidney function, as current available measures have limitations. We reviewed the advances in biomarkers in CKD over the preceding year. RECENT FINDINGS The most frequently studied prognostic plasma biomarkers during recent year were plasma TNFR1, TNFR2, KIM1 and urinary MCP-1 and EGF. New biomarkers such as plasma WFDC2, MMP-7, EFNA4, EPHA2 may also have potential to serve as prognostic biomarkers. There is a shortage of data on biomarkers that are predictive of response to treatments. Data on novel biomarkers to serve as pharmacodynamic biomarkers are limited, but there are emerging data that plasmaTNFR1, TNFR2, KIM-1 are not only prognostic at baseline, but can also contribute to time-updated response signals in response to therapy. SUMMARY Data continue to emerge on applicable biomarkers for prognostic clinical risk stratification, prediction of therapeutic response and assessment of early efficacy of interventions. Although more studies are needed for refinement and specific clinical utility, there seems to be sufficient data to support clinical implementation for some biomarkers.
Collapse
Affiliation(s)
- Azadeh Zabetian
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
43
|
Ferumoxytol-enhanced ultrashort TE MRA and quantitative morphometry of the human kidney vasculature. Abdom Radiol (NY) 2021; 46:3288-3300. [PMID: 33666735 DOI: 10.1007/s00261-021-02984-2] [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: 12/04/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE To evaluate the feasibility of Quantitative Ultrashort-Time-to-Echo Contrast-Enhanced (QUTE-CE) MRA using ferumoxytol as a contrast agent for abdominal angiography in the kidney. METHODS Four subjects underwent ferumoxytol-enhanced MRA with the 3D UTE Spiral VIBE WIP sequence at 3 T. Image quality metrics were quantified, specifically the blood Signal-to-Noise Ratio (SNR), blood-tissue Contrast-to-Noise Ratio (CNR) and Intraluminal Signal Heterogeneity (ISH) from both the aorta and inferior vena cava (IVC). Morphometric analysis of the vessels was performed using manual approach and semi-automatic approach using Vascular Modeling ToolKit (VMTK). Image quality and branching order were compared between QUTE-CE MRA and the Gadolinium (Gd) CEMRA reference image. RESULTS QUTE-CE MRA provides a bright blood snapshot that delineates arteries and veins equally in the same scan. The maximum SNR and CNR values were 3,282 ± 1,218 and 1,295 ± 580, respectively - significantly higher than available literature values using other CEMRA techniques. QUTE-CE MRA had lower ISH and depicted higher vessel branching order (7th vs 3rd) within the kidney compared to a standard dynamic clinical Gd CEMRA scan. Morphometric analysis yielded quantitative results for the total kidney volume, total cyst volume and for diameters of the branching arterial network down to the 7th branch. Vessel curvature was significantly increased (p < 0.001) in the presence of a renal cyst compared to equivalent vessels in normal kidney regions. CONCLUSION QUTE-CE MRA is feasible for kidney angiography, providing greater detail of kidney vasculature, enabling quantitative morphometric analysis of the abdominal and intra-renal vessels and yielding metrics relevant to vascular diseases while using a contrast agent ferumoxytol that is safe for CKD patients.
Collapse
|
44
|
Coresh J, Grams ME, Chen TK. Using GFR, Albuminuria, and Their Changes in Clinical Trials and Clinical Care. Am J Kidney Dis 2021; 78:333-334. [PMID: 34059333 DOI: 10.1053/j.ajkd.2021.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/13/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Teresa K Chen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
45
|
Martin WP, Conroy C, Naicker SD, Cormican S, Griffin TP, Islam MN, McCole EM, McConnell I, Lamont J, FitzGerald P, Ferguson JP, Richardson C, Logue SE, Griffin MD. Multiplex Serum Biomarker Assays Improve Prediction of Renal and Mortality Outcomes in Chronic Kidney Disease. KIDNEY360 2021; 2:1225-1239. [PMID: 34849485 PMCID: PMC7612046 DOI: 10.34067/kid.0007552020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND We investigated the predictive value of 11 serum biomarkers for renal and mortality end points in people with CKD. METHODS Adults with CKD (n=139) were enrolled from outpatient clinics between February 2014 and November 2016. Biomarker quantification was performed using two multiplex arrays on a clinical-grade analyzer. Relationships between biomarkers and renal and mortality end points were investigated by random forests and Cox proportional hazards regression. RESULTS The cohort was 56% male. The mean age was 63 years and median (IQR) CKD-EPI eGFR was 33 (24-51) ml/min per BSA. A total of 56 (40%) people developed a composite end point defined as ≥40% decline in eGFR, doubling of serum creatinine, RRT, or death over median (IQR) follow-up of 5.4 (4.7-5.7) years. Prediction of the composite end point was better with random forests trained on serum biomarkers compared with clinical variables (area under the curve of 0.81 versus 0.78). The predictive performance of biomarkers was further enhanced when considered alongside clinical variables (area under the curve of 0.83 versus 0.81 for biomarkers alone). Patients (n=27, 19%) with high soluble TNF receptor-1 (≥3 ng/ml) and neutrophil gelatinase-associated lipocalin (≥156 ng/ml), coupled with low complement 3a des-arginine (<2368 ng/ml), almost universally (96%) developed the composite renal and mortality end point. C-reactive protein (adjusted hazard ratio, 1.4; 95% CI, 1.1 to 1.8), neutrophil gelatinase-associated lipocalin (adjusted hazard ratio, 2.8; 95% CI, 1.3 to 6.1) and complement 3a desarginine (adjusted hazard ratio, 0.6; 95% CI, 0.4 to 0.96) independently predicted time to the composite end point. CONCLUSIONS Outpatients with the triad of high soluble TNF receptor-1 and neutrophil gelatinase-associated lipocalin coupled with low complement 3a des-arginine had high adverse event rates over 5-year follow-up. Incorporation of serum biomarkers alongside clinical variables improved prediction of CKD progression and mortality. Our findings require confirmation in larger, more diverse patient cohorts.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Chloe Conroy
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Serika D. Naicker
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Sarah Cormican
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Nephrology Services, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Tomás P. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Md Nahidul Islam
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | - Ivan McConnell
- Randox Laboratories Limited, Crumlin, Antrim, Northern Ireland
| | - John Lamont
- Randox Laboratories Limited, Crumlin, Antrim, Northern Ireland
| | | | - John P. Ferguson
- Health Research Board Clinical Research Facility, National University of Ireland Galway, Galway, Ireland
| | | | - Susan E. Logue
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland,Nephrology Services, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| |
Collapse
|
46
|
Prischl FC, Rossing P, Bakris G, Mayer G, Wanner C. Major adverse renal events (MARE): a proposal to unify renal endpoints. Nephrol Dial Transplant 2021; 36:491-497. [PMID: 31711188 DOI: 10.1093/ndt/gfz212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In renal studies, various outcome endpoints are used with variable definitions, making it nearly impossible to perform meta-analyses and deduce meaningful conclusions. Increasing attention is directed towards standardization of renal outcome reporting. METHODS A working group was formed to produce a unifying definition of renal outcomes that can be used by all investigators. We propose major adverse renal events (MARE) as the term for a standardized composite of hard renal outcomes. We discuss the components for inclusion in MARE from existing evidence. RESULTS MARE could include three to five items, considered relevant to patients and regulators. New onset of kidney injury, that is persistent albuminuria/proteinuria and/or decreasing glomerular filtration rate (GFR) <60 ml/min/1.73 m2, persistent signs of worsening kidney disease, development of end-stage kidney disease with estimated GFR <15 ml/min/1.73 m2 without or with initiation of kidney replacement therapy, and death from renal cause are core items of MARE. Additionally, patient reported outcomes should be reported in parallel to MARE as a standard set of primary (or secondary) endpoints in studies on kidney disease of diabetic, hypertensive-vascular, or other origin. CONCLUSIONS MARE as a reporting standard will enhance the ability to compare studies and thus, facilitate meaningful meta-analyses. This will result in standardized endpoints that should result in guideline improvement to better individualize care of patients with kidney disease.
Collapse
Affiliation(s)
- Friedrich C Prischl
- Department of Nephrology, 4th Department of Internal Medicine, Klinikum WelsGrieskirchen, Wels, Austria
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - George Bakris
- Department of Medicine, American Heart Association Comprehensive Hypertension Center, University of Chicago, Chicago, IL, USA
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
47
|
Hsu CY, Go AS. Timing of Dialysis Initiation and End-Stage Kidney Disease Incidence-Reply. JAMA Intern Med 2021; 181:725-726. [PMID: 33587100 DOI: 10.1001/jamainternmed.2020.8806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Chi-Yuan Hsu
- Division of Nephrology, Department of Medicine, University of California, San Francisco.,Division of Research, Kaiser Permanente Northern California, Oakland
| | - Alan S Go
- Division of Nephrology, Department of Medicine, University of California, San Francisco.,Division of Research, Kaiser Permanente Northern California, Oakland.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Departments of Medicine, Health Research and Policy, Stanford University, Stanford, California
| |
Collapse
|
48
|
Insights from CREDENCE trial indicate an acute drop in estimated glomerular filtration rate during treatment with canagliflozin with implications for clinical practice. Kidney Int 2021; 99:999-1009. [DOI: 10.1016/j.kint.2020.10.042] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/10/2020] [Accepted: 10/29/2020] [Indexed: 01/13/2023]
|
49
|
Kraus BJ, Weir MR, Bakris GL, Mattheus M, Cherney DZ, Sattar N, Heerspink HJ, Ritter I, von Eynatten M, Zinman B, Inzucchi SE, Wanner C, Koitka-Weber A. Characterization and implications of the initial estimated glomerular filtration rate ‘dip’ upon sodium-glucose cotransporter-2 inhibition with empagliflozin in the EMPA-REG OUTCOME trial. Kidney Int 2021; 99:750-762. [DOI: 10.1016/j.kint.2020.10.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/30/2020] [Accepted: 10/15/2020] [Indexed: 01/02/2023]
|
50
|
Westreich KD, Isom S, Divers J, D'Agostino R, Lawrence JM, Kanakatti Shankar R, Dolan LM, Imperatore G, Dabelea D, Mayer-Davis EJ, Mottl AK. Trajectories in estimated glomerular filtration rate in youth-onset type 1 and type 2 diabetes: The SEARCH for Diabetes in Youth Study. J Diabetes Complications 2021; 35:107768. [PMID: 33168393 PMCID: PMC7855388 DOI: 10.1016/j.jdiacomp.2020.107768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 11/29/2022]
Abstract
AIMS We sought to characterize the direction and associated factors of eGFR change following diagnosis of youth-onset type 1 and type 2 diabetes. METHODS We assessed the direction of eGFR change at two visits (mean 6.6 years apart) in SEARCH, a longitudinal cohort study of youth-onset type 1 and type 2 diabetes. We used the CKiDCr-CysC equation to estimate GFR and categorized 'rising' and 'declining' eGFR as an annual change of ≥3 ml/min/1.73 m2 in either direction. Multivariable logistic regression evaluated factors associated with directional change in eGFR. RESULTS Estimated GFR declined in 23.8% and rose in 2.8% of participants with type 1 diabetes (N = 1225; baseline age 11.4 years), and declined in 18.1% and rose in 15.6% of participants with type 2 diabetes (N = 160; baseline age 15.0 years). Factors associated with rising and declining eGFR (versus stable) in both type 1 and type 2 diabetes included sex, age at diagnosis, baseline eGFR and difference in fasting glucose between study visits. Additional factors in type 1 diabetes included time from baseline visit, HbA1c and body mass index. CONCLUSIONS Over the first decade of diabetes, eGFR decline is more common in type 1 diabetes whereas eGFR rise is more common in type 2 diabetes.
Collapse
Affiliation(s)
- Katherine D Westreich
- University of North Carolina Kidney Center, UNC School of Medicine, Chapel Hill, NC, United States of America.
| | - Scott Isom
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America.
| | - Jasmin Divers
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America.
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States of America.
| | - Jean M Lawrence
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America.
| | - Roopa Kanakatti Shankar
- Division of Endocrinology, Children's National Medical Center, Washington, DC, United States of America
| | - Lawrence M Dolan
- Division of Endocrinology, Cincinnati Children's Hospital, Cincinnati, OH, United States of America.
| | - Giuseppina Imperatore
- Division of Diabetes Translation, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, GA, United States of America.
| | - Dana Dabelea
- Department of Epidemiology, School of Public Health, University of Colorado Denver, Aurora, CO, United States of America.
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, United States of America; Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.
| | - Amy K Mottl
- University of North Carolina Kidney Center, UNC School of Medicine, Chapel Hill, NC, United States of America.
| |
Collapse
|