1
|
Engole Mompango Y, Bukabau Busanga J, Makulo Rissassy JR, Nlandu Mayamba Y, Makanzu B, Nkodila A, Tshiswaka T, Mokoli Momeme V, Longo Luzayadio A, Mboliasa Ingole MF, Kajingulu Musungayi F, Fwana S, Ilunga Kabemba C, Nkondi Nsenga C, Zinga Vuvu C, Nseka Mangani N, Sumaili Kiswaya E. Prevalence and associated factors of glomerular hyperfiltration among adult stable sickle cells in Kinshasa, DR Congo. Ren Fail 2024; 46:2407888. [PMID: 39329176 PMCID: PMC11441020 DOI: 10.1080/0886022x.2024.2407888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
INTRODUCTION Glomerular hyperfiltration is highly frequent, theoretically dependent on cardiac output, low systemic vascular resistance and hemolysis markers. In sickle cell disease (SCD), hyperfiltration is an extremely common phenomenon and occurred in young and early adult patients. Despite the fact that the glomerular hyperfiltration is known as the early manifestations of sickle cell nephropathy, its burden among adult sickle cell disease in sub-Saharan is poor studied. This study aimed to determine the prevalence and associated factors of hyperfiltration. METHODS This was an analytical multicentric cross-sectional study involving stable adult sickle cell patients in Kinshasa, recruited between March and October 2023. Parameters of interest encompasses demographic, clinical, biological, echocardiographic and pulse wave measurement data. Hyperfiltration was defined using the CDK-EPI equation based on cystatin C; eGFR >130 for women and >140 ml/min/1.73m2 for men. We used multivariate logistic regression analysis to search determinants of glomerular hyperfiltration. RESULTS Two hundred and fourty six (246) patients with SCD were enrolled. The prevalence of hyperfiltration was 20.7%. In multiple logistic regression analysis, hyperfiltration status was independently associated with age (< 25 years) [3.57 (1.78-7.49); p = 0.027)], female sex [4.36 (2.55-5.62); p = 0.031), CRP (< 6 mg/l) [0.77 (0.61-0.97); p = 0.028)], central systolic pressure (< 100 mmHg) and central diastolic pressure (< 60 mmHg) [0.86(0.74-0.98), p = 0.028)], [(0.83 (0.71-0.98); p = 0.032)]. CONCLUSION One out of five SS adults exhibits hyperfiltration, which is associated with young age and female sex, whereas low CRP and blood pressure were negative risk factors.
Collapse
Affiliation(s)
- Yannick Engole Mompango
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
- Specialized Clinics in Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Justine Bukabau Busanga
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | | | - Yannick Nlandu Mayamba
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
- Specialized Clinics in Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Brady Makanzu
- Specialized Clinics in Kinshasa, Kinshasa, Democratic Republic of the Congo
- Cardiology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Aliocha Nkodila
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Tresor Tshiswaka
- Cardiology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Vieux Mokoli Momeme
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | | | | | | | - Shekinah Fwana
- Specialized Clinics in Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Cedric Ilunga Kabemba
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Clarisse Nkondi Nsenga
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Chantal Zinga Vuvu
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Nazaire Nseka Mangani
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| | - Ernest Sumaili Kiswaya
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, XI, Democratic Republic of the Congo
| |
Collapse
|
2
|
Zheng G, Cheng Y, Wang C, Wang B, Zou X, Zhou J, Peng L, Zeng T. Elucidating the causal nexus and immune mediation between frailty and chronic kidney disease: integrative multi-omics analysis. Ren Fail 2024; 46:2367028. [PMID: 39010723 PMCID: PMC11265307 DOI: 10.1080/0886022x.2024.2367028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Empirical research has consistently documented the concurrent manifestation of frailty and chronic kidney disease (CKD). However, the existence of a reverse causal association or the influence of confounding variables on these correlations remains ambiguous. METHODS Our analysis of 7,078 participants from National Health and Nutrition Examination Survey(NHANES) (1999-2018) applied weighted logistic regression and Mendelian Randomization (MR) to investigate the correlation between the frailty index (FI) and renal function. The multivariate MR analysis was specifically adjusted for type 2 diabetes and hypertension. Further analysis explored 3282 plasma proteins to link FI to CKD. A two-step network MR highlighted immune cells' mediating roles in the FI-CKD relationship. RESULT Genetically inferred FI and various renal function markers are significantly correlated, as supported by NHANES analyses. Multivariate MR analysis revealed a direct causal association between the FI and CKD. Additionally, our investigation into plasma proteins identified Tmprss11D and MICB correlated with FI and CKD, respectively. A two-step network MR to reveal 15 immune cell types, notably Central Memory CD4+ T cells and Lymphocytes, as crucial mediators between FI and CKD. CONCLUSION Our work establishes a causal connection between frailty and CKD, mediated by specific immune cell profiles. These findings highlight the importance of immune mechanisms in the frailty-CKD interplay and suggest that targeting shared risk factors and immune pathways could improve management strategies for these conditions. Our research contributes to a more nuanced understanding of frailty and CKD, offering new avenues for intervention and patient care in an aging population.
Collapse
Affiliation(s)
- Guanghao Zheng
- Department of Medicine, Graduate School of Nanchang University, Nanchang, China
| | - Yu Cheng
- Department of Medicine, Graduate School of Nanchang University, Nanchang, China
| | - Chenlong Wang
- Department of Central Laboratory, The Affiliated Huaian No.1 Peopele’s Hospital, Nanjing Medical University, Huai’an, China
| | - Bin Wang
- Department of Medicine, Graduate School of Nanchang University, Nanchang, China
| | - Xinchang Zou
- Department of Medicine, Graduate School of Nanchang University, Nanchang, China
| | - Jie Zhou
- Department of Medicine, Graduate School of Nanchang University, Nanchang, China
| | - Lifen Peng
- Molecular Experiment Center, Jiangxi Provincial People’s Hospital Affiliated to Nanchang University, Nanchang, China
| | - Tao Zeng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Svigelj R, de Marco A. Biological and technical factors affecting the point-of-care diagnostics in not-oncological chronic diseases. Biosens Bioelectron 2024; 264:116669. [PMID: 39146770 DOI: 10.1016/j.bios.2024.116669] [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: 07/15/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Inexpensive point-of-care (POC) analytical solutions have the potential to allow the implementation of large-scale screening campaigns aimed at identifying the initial stages of pathologies in the population, reducing morbidity, mortality and, indirectly, also the costs for the healthcare system. At global level, the most common preventive screening schemes address some cancer pathologies or are used to monitor the spread of some infective diseases. However, systematic testing might become decisive to improve the care response even in the case of chronic pathologies and, in this review, we analyzed the state-of-the-art of the POC diagnostics for Chronic Kidney Disease, Chronic Obstructive Pulmonary Disease and Multiple Sclerosis. The different technological options used to manufacture the biosensors and evaluate the produced data have been described and this information has been integrated with the present knowledge relatively to the biomarkers that have been proposed to monitor such diseases, namely their availability and reliability. Finally, the nature of the macromolecules used to capture the biomarkers has been discussed in relation to the biomarker nature.
Collapse
Affiliation(s)
- Rossella Svigelj
- Department of Agrifood, Environmental and Animal Sciences, University of Udine, Via Cotonificio 108, 33100, Udine, Italy
| | - Ario de Marco
- Lab of Environmental and Life Sciences, University of Nova Gorica, Vipavska Cesta 13, 5000, Nova Gorica, Slovenia.
| |
Collapse
|
4
|
Nakano FK, Åkesson A, de Boer J, Dedja K, D'hondt R, Haredasht FN, Björk J, Courbebaisse M, Couzi L, Ebert N, Eriksen BO, Dalton RN, Derain-Dubourg L, Gaillard F, Garrouste C, Grubb A, Jacquemont L, Hansson M, Kamar N, Legendre C, Littmann K, Mariat C, Melsom T, Rostaing L, Rule AD, Schaeffner E, Sundin PO, Bökenkamp A, Berg U, Åsling-Monemi K, Selistre L, Larsson A, Nyman U, Lanot A, Pottel H, Delanaye P, Vens C. Comparison between the EKFC-equation and machine learning models to predict Glomerular Filtration Rate. Sci Rep 2024; 14:26383. [PMID: 39487227 PMCID: PMC11530427 DOI: 10.1038/s41598-024-77618-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
In clinical practice, the glomerular filtration rate (GFR), a measurement of kidney functioning, is normally calculated using equations, such as the European Kidney Function Consortium (EKFC) equation. Despite being the most general equation, EKFC, just like previously proposed approaches, can still struggle to achieve satisfactory performance, limiting its clinical applicability. As a possible solution, recently machine learning (ML) has been investigated to improve GFR prediction, nonetheless the literature still lacks a general and multi-center study. Using a dataset with 19,629 patients from 13 cohorts, we investigate if ML can improve GFR prediction in comparison to EKFC. More specifically, we compare diverse ML methods, which were allowed to use age, sex, serum creatinine, cystatin C, height, weight and BMI as features, in internal and external cohorts against EKFC. The results show that the most performing ML method, random forest (RF), and EKFC are very competitive where RF and EKFC achieved respectively P10 and P30 values of 0.45 (95% CI 0.44;0.46) and 0.89 (95% CI 0.88;0.90), whereas EKFC yielded 0.44 (95% CI 0.43; 0.44) and 0.89 (95% CI 0.88; 0.90), considering the entire cohort. Small differences were, however, observed in patients younger than 12 years where RF slightly outperformed EKFC.
Collapse
Affiliation(s)
- Felipe Kenji Nakano
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium.
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium.
| | - Anna Åkesson
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Jasper de Boer
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium
| | - Klest Dedja
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium
| | - Robbe D'hondt
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium
| | - Fateme Nateghi Haredasht
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium
| | - Jonas Björk
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Marie Courbebaisse
- Physiology Department, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris, INSERM U1151-CNRS UMR8253, Paris Descartes University, Paris, France
| | - Lionel Couzi
- CNRS-UMR 5164 Immuno ConcEpT, CHU de Bordeaux, Nephrologie-Transplantation-Dialyse, Université de Bordeaux, Bordeaux, France
| | - Natalie Ebert
- Institute of Public Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Björn O Eriksen
- Metabolic and Renal Research Group, UiT the Arctic University of Norway, Tromsö, Norway
| | - R Neil Dalton
- The Wellchild Laboratory, Evelina London Children's Hospital, London, UK
| | - Laurence Derain-Dubourg
- Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Hôpital Edouard Herriot, Hospices Civils de Lyon, France
| | - Francois Gaillard
- Renal Transplantation Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Cyril Garrouste
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund University, Lund, Sweden
| | - Lola Jacquemont
- Renal Transplantation Department, CHU Nantes, Nantes University, Nantes, France
| | - Magnus Hansson
- Function Area Clinical Chemistry, Karolinska University Laboratory, Karolinska Institute, Karolinska University Hospital Huddinge and Department of Laboratory Medicine, Stockholm, Sweden
| | - Nassim Kamar
- Department of Nephrology, Dialysis and Organ Transplantation, CHU Rangueil, INSERM U1043, IFR-BMT, University Paul Sabatier, Toulouse, France
| | | | - Karin Littmann
- Institute om Medicine Huddinge (Med H), Karolinska Institute, Solna, Sweden
| | - Christophe Mariat
- Service de Néphrologie, Dialyse et Transplantation Rénale, Hôpital Nord, CHU de Saint-Etienne, Saint-Priest-en-Jarez, France
| | - Toralf Melsom
- Metabolic and Renal Research Group, UiT the Arctic University of Norway, Tromsö, Norway
| | - Lionel Rostaing
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, Hôpital Michallon, CHU Grenoble-Alpes, Tronche, France
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Elke Schaeffner
- Institute of Public Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Per-Ola Sundin
- Karla Healthcare Centre, Faculty of Medicine and Health, Örebro University, 70182, Örebro, SE, Sweden
| | - Arend Bökenkamp
- Department of Paediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ulla Berg
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Kajsa Åsling-Monemi
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luciano Selistre
- Mestrado Em Ciências da Saúde-Universidade Caxias do Sul Foundation CAPES, Caxias Do Sul, Brazil
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Ulf Nyman
- Department of Translational Medicine, Division of Medical Radiology, Lund University, Malmö, Sweden
| | - Antoine Lanot
- Normandie Université, Unicaen, CHU de Caen Normandie, Néphrologie, Caen, France
- Normandie Université, Unicaen, UFR de Médecine, 2 Rue Des Rochambelles, Caen, France
- ANTICIPE U1086 INSERM-UCN, Centre François Baclesse, Caen, France
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hopital Universitaire Caremeau, Nimes, France
| | - Celine Vens
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
- Itec, Imec Research Group at KU Leuven, Kortrijk, Belgium
| |
Collapse
|
5
|
Zhang C, Zhang H, Zhang B, Lindenberg J, Amat MJ, Rice MB, Mukamal KJ. Marijuana Use and Hemoglobin Concentrations in NHANES 2009-2018: Implications for Subclinical Hypoxemia. Ann Am Thorac Soc 2024; 21:1488-1495. [PMID: 38985494 DOI: 10.1513/annalsats.202404-357oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 07/11/2024] Open
Abstract
Rationale: Cannabis use is rapidly growing in the United States, but its health implications are poorly understood, particularly when compared with cigarette smoking. Previous research conducted on animal models or nonrepresentative populations with small sample sizes has yielded mixed results on the impact of marijuana use on hemoglobin concentrations, which might reflect subclinical hypoxemia and/or carbon monoxide exposure. Objectives: We evaluated the association between marijuana use and hemoglobin concentrations in a nationally representative sample of U.S. adults. Methods: This cross-sectional study included 16,038 individuals 18-59 years of age enrolled in National Health and Nutrition Examination Survey (NHANES) from 2009 to 2018. We related current and former marijuana use with measured hemoglobin concentrations, with adjustment for demographics, education, housing, and cigarette smoking status in multivariable analyses that incorporated complex survey weights. As candidate positive and negative control exposures, we used similar methods to relate cigarette smoking and benzodiazepine use, respectively, with hemoglobin concentrations. Results: Current marijuana use was associated with significantly higher hemoglobin concentrations. After multivariable adjustment, compared with never use, current marijuana use was associated with a 0.111 (95% confidence interval, 0.021 to 0.201) g/dl higher hemoglobin concentration, whereas former use was associated with a 0.047 (95% confidence interval, -0.018 to 0.113) g/dl higher concentration (linear trend P = 0.01). As hypothesized, cigarette smoking was also associated with higher hemoglobin concentrations, whereas benzodiazepine use was not. Conclusions: Among American adults, current marijuana use was associated with higher hemoglobin concentrations, as is cigarette smoking but not benzodiazepine use. These results suggest the possibility that marijuana smoking induces subclinical hypoxemia stimulating hemoglobin production. Further confirmation of this observational finding is needed in light of the increasing medical and recreational use of smoked marijuana products.
Collapse
Affiliation(s)
| | - Hui Zhang
- Division of Biostatistics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and
| | - Bo Zhang
- Department of Neurology and
- Institutional Centers of Clinical and Translational Research Biostatistics and Research Design Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Mary B Rice
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | |
Collapse
|
6
|
Weidmann L, Laux C, Castrezana Lopez K, Harmacek D, George B, von Moos S, Schachtner T. Immunosuppression and transplantation-related characteristics affect the difference between eGFR equations based on creatinine compared to cystatin C in kidney transplant recipients. Clin Kidney J 2024; 17:sfae253. [PMID: 39502371 PMCID: PMC11536772 DOI: 10.1093/ckj/sfae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Previous studies show heterogeneity when applying estimated glomerular filtration (eGFR) equations to kidney transplant recipients (KTRs). However, research on the impact of transplantation-related characteristics on eGFR equations using creatinine (eGFRcr) compared to cystatin C (eGFRcys) is scarce. Methods We conducted a comprehensive analysis with three eGFRcr equations (Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) 2009, European Kidney Function Consortium (EKFC) 2021, kidney recipient specific-glomerular filtration rate KRS-GFR) 2023), comparing them to two eGFRcys (CKD-EPI 2012 and EKFC 2023) in 596 KTRs. Bland-Altman plots demonstrated relative differences according to different eGFR-stages. Multivariable logistic regression identified transplantation-related characteristics independently associated with smaller or greater differences between eGFRcr and eGFRcys equations. Results 94.3% of the cohort were White individuals. Median eGFR differed as much as 9 ml/min/1.73 m2 between equations. The median relative differences (Q2) were greater (more negative) when comparing the eGFRcr equations to eGFRcys CKD-EPI 2012, than when comparing them to eGFRcys EKFC 2023 (P < .001). Better average eGFR was associated with smaller mean relative differences in all comparisons but eGFRcr CKD-EPI 2009 with eGFR EKFC 2023 and eGFRcr EKFC 2021 with eGFRcys EKFC 2023. Living kidney donation and belatacept use were independent factors associated with a smaller difference (≥Q3) between eGFRcr and eGFRcys equations, while prednisone use or higher HbA1c were independently associated with a greater difference (≤Q1) between equations. Conclusion Different eGFR-stages, donor, or recipient characteristics, along with immunosuppression such as belatacept or prednisone, contribute to differences between eGFRcr and eGFRcys. These effects need to be considered in the clinical management of KTRs.
Collapse
Affiliation(s)
- Lukas Weidmann
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| | - Catherine Laux
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| | | | - Dusan Harmacek
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| | - Britta George
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| | - Seraina von Moos
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
- Department of Nephrology, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Thomas Schachtner
- Department of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Akbari A, El Wadia H, Knoll GA, White CA, Sood MM, Massicotte-Azarniouch D, McCudden C, Deschenes MJ, Salman M, Ramsay T, Hundemer GL. Comparison of eGFR Equations to Guide Dosing of Medications for Kidney Transplant Recipients. Transplantation 2024; 108:2270-2277. [PMID: 38831493 DOI: 10.1097/tp.0000000000005098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
BACKGROUND Clinicians caring for kidney transplant recipients (KTRs) most commonly use estimated glomerular filtration rate (eGFR) to guide medication dosing as it is the most readily available measure of kidney function. Which eGFR equations provide the most accurate medication dosing guidance for KTRs remains uncertain. METHODS We studied 415 stable KTRs in Canada and New Zealand. Participants completed same-day measurements of creatinine and cystatin C and measured GFR (diethylenetriaminepentaacetic acid). Chronic Kidney Disease Epidemiology Collaboration, European Kidney Function Consortium, and transplant-specific eGFR equations were compared with both Cockcroft-Gault creatinine clearance (CrCl) and measured GFR. eGFR equations were assessed both indexed to a standardized body surface area (BSA) of 1.73 m 2 (milliliter per minute per 1.73 m 2 , as is conventional reporting from most clinical laboratories) and nonindexed (milliliter per minute) accounting for actual BSA. The primary outcome was the proportion of medication dosing discordance relative to Cockcroft-Gault CrCl or measured GFR for 8 commonly prescribed medications. Stratified analyses were performed on the basis of obesity status. RESULTS Nonindexed eGFR equations (milliliter per minute) resulted in substantially lower medication dosing discordance compared with indexed eGFR equations (milliliter per minute per 1.73 m 2 ). These findings were most pronounced among KTRs with obesity, in whom underdosing was frequent. When compared with Cockcroft-Gault CrCl, the lowest proportion of discordance was found with the nonindexed 2023 transplant-specific equation. When compared with measured GFR, the lowest proportion of discordance was found with the nonindexed 2021 Chronic Kidney Disease Epidemiology Collaboration Cr/CysC equation. CONCLUSIONS Nonindexed eGFR values accounting for actual BSA should be used by clinicians for medication dosing in KTRs. These findings may inform KT providers about which eGFR equations provide the safest, most accurate medication dosing guidance for KTRs.
Collapse
Affiliation(s)
- Ayub Akbari
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Hajar El Wadia
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Greg A Knoll
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Christine A White
- Division of Nephrology, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Manish M Sood
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - David Massicotte-Azarniouch
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Christopher McCudden
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Marie-Josee Deschenes
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Maria Salman
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Tim Ramsay
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Gregory L Hundemer
- Division of Nephrology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
8
|
Mayne KJ, Hanlon P, Lees JS. Detecting and managing the patient with chronic kidney disease in primary care: A review of the latest guidelines. Diabetes Obes Metab 2024; 26 Suppl 6:43-54. [PMID: 38699995 DOI: 10.1111/dom.15625] [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: 03/29/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Chronic kidney disease (CKD) is a major global health problem, affecting about 9.5% of the population and 850 million people worldwide. In primary care, most CKD is caused by diabetes and/or hypertension, but a substantial proportion of cases may have alternative causes. During the early stages, CKD is asymptomatic, and many people are unaware that they are living with the disease. Despite the lack of symptoms, CKD is associated with elevated risks of cardiovascular disease, progressive kidney disease, kidney failure and premature mortality. Risk reduction strategies are effective and cost-effective but require early diagnosis through testing of the estimated glomerular filtration rate and albuminuria in high-risk populations. Once diagnosed, the treatment of CKD centres around lifestyle interventions, blood pressure and glycaemic control, and preventative treatments for cardiovascular disease and kidney disease progression. Most patients with CKD should be managed with statins, renin-angiotensin-aldosterone system inhibitors and sodium-glucose cotransporter-2 inhibitors. Additional treatment options to reduce cardiorenal risk are available in patients with diabetes, including glucagon-like peptide-1 receptor agonists and non-steroidal mineralocorticoid receptor antagonists. The Kidney Failure Risk Equation is a new tool that can support the identification of patients at high risk of progressive kidney disease and kidney failure and can be used to guide referrals to nephrology. This review summarizes the latest guidance relevant to managing adults with, or at risk of, CKD and provides practical advice for managing patients with CKD in primary care.
Collapse
Affiliation(s)
- Kaitlin J Mayne
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Glasgow Renal and Transplant Unit, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Peter Hanlon
- School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Jennifer S Lees
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Glasgow Renal and Transplant Unit, NHS Greater Glasgow and Clyde, Glasgow, UK
| |
Collapse
|
9
|
Herold JM, Wiegrebe S, Nano J, Jung B, Gorski M, Thorand B, Koenig W, Zeller T, Zimmermann ME, Burkhardt R, Banas B, Küchenhoff H, Stark KJ, Peters A, Böger CA, Heid IM. Population-based reference values for kidney function and kidney function decline in 25- to 95-year-old Germans without and with diabetes. Kidney Int 2024; 106:699-711. [PMID: 39084259 DOI: 10.1016/j.kint.2024.06.024] [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: 01/26/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
Understanding normal aging of kidney function is pivotal to help distinguish individuals at particular risk for chronic kidney disease. Glomerular filtration rate (GFR) is typically estimated via serum creatinine (eGFRcrea) or cystatin C (eGFRcys). Since population-based age-group-specific reference values for eGFR and eGFR-decline are scarce, we aimed to provide such reference values from population-based data of a wide age range. In four German population-based cohorts (KORA-3, KORA-4, AugUR, DIACORE), participants underwent medical exams, interview, and blood draw up to five times within up to 25 years. We analyzed eGFRcrea and eGFRcys cross-sectionally and longitudinally (12,000 individuals, age 25-95 years). Cross-sectionally, we found age-group-specific eGFRcrea to decrease approximately linearly across the full age range, for eGFRcys up to the age of 60 years. Within age-groups, there was little difference by sex or diabetes status. Longitudinally, linear mixed models estimated an annual eGFRcrea decline of -0.80 [95% confidence interval -0.82, -0.77], -0.79 [-0.83, -0.76], and -1.20 mL/min/1.73m2 [-1.33, -1.08] for the general population, "healthy" individuals, or individuals with diabetes, respectively. Reference values for eGFR using cross-sectional data were shown as percentile curves for "healthy" individuals and for individuals with diabetes. Reference values for eGFR-decline using longitudinal data were presented as 95% prediction intervals for "healthy" individuals and for individuals with diabetes, obesity, and/or albuminuria. Thus, our results can help clinicians to judge eGFR values in individuals seen in clinical practice according to their age and to understand the expected range of annual eGFR-decline based on their risk profile.
Collapse
Affiliation(s)
- Janina M Herold
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Simon Wiegrebe
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany; Statistical Consulting Unit StaBLab, Department of Statistics, LMU Munich, Munich, Germany
| | - Jana Nano
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Bettina Jung
- Department of Nephrology, University of Regensburg, University Hospital Regensburg, Regensburg, Germany; Department of Nephrology, Diabetology, and Rheumatology, Traunstein Hospital, Southeast Bavarian Clinics, Traunstein, Germany; KfH Kidney Center Traunstein, Traunstein, Germany
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany; Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Tanja Zeller
- University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Hamburg, Germany
| | - Martina E Zimmermann
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University of Regensburg, University Hospital Regensburg, Regensburg, Germany
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, Department of Statistics, LMU Munich, Munich, Germany
| | - Klaus J Stark
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Carsten A Böger
- Department of Nephrology, University of Regensburg, University Hospital Regensburg, Regensburg, Germany; Department of Nephrology, Diabetology, and Rheumatology, Traunstein Hospital, Southeast Bavarian Clinics, Traunstein, Germany; KfH Kidney Center Traunstein, Traunstein, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
10
|
Flaherty CM, Surapaneni A, Seegmiller JC, Coresh J, Grams ME, Ballew SH. CKD Prevalence and Incidence in Older Adults Using Estimated GFR With Different Filtration Markers: The Atherosclerosis Risk in Communities Study. Kidney Med 2024; 6:100893. [PMID: 39319210 PMCID: PMC11420509 DOI: 10.1016/j.xkme.2024.100893] [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] [Indexed: 09/26/2024] Open
Abstract
Rationale & Objective The prevalence of chronic kidney disease (CKD) is known to increase with age; however, creatinine may be a less reliable filtration marker in older adults. Few studies have investigated the prevalence and progression of CKD using different filtration markers for estimating glomerular filtration rate (GFR). Study Design A prospective observational cohort study. Setting & Participants 6,393 White and African American participants aged 65-100 years from the Atherosclerosis Risk in Communities Study (ARIC) at Visit 5, followed longitudinally at Visits 6 and 7. Exposure and Outcome The eGFR was estimated either by creatinine (eGFRcr), cystatin C (eGFRcys), creatinine and cystatin C (eGFRcr-cys), or using creatinine, cystatin C, and β-2-microglobulin (eGFRcr-cys-b2m). CKD progression was defined as 30% decline in eGFR at follow-up visits. Analytical Approach Logistic regression models, adjusted for sex, race and study center, diabetes, blood pressure, body mass index, prevalent cardiovascular disease, and heart failure. Results At Visit 5, the mean age in the study population was 75.8 years, and the mean eGFR ranged from 71.2 to 61.2 mL/min/1.73m2 using eGFRcr or eGFRcys, respectively. The proportion with eGFR < 60 mL/min/1.73m2 was lowest with eGFRcr and highest with eGFRcys for all age groups, and prevalence increased with age for all markers. For example, the prevalence of eGFRcr < 60 mL/min/1.73m2 in ages 70-74 years ranged from 15% to 21% and in ages 85-89 years ranged from 38% to 46% at the different visits. The proportion with a 30% eGFR decline over a mean of 8 years in people who were originally aged 65-69 years ranged from 9% (eGFRcr)-18% (eGFRcys). More people with eGFRcr ≥ 60 mL/min/1.73m2 were reclassified to < 60 mL/min/1.73m2 when using eGFRcys (33%) compared with eGFRcr-cys (12%) or eGFRcr-cys-b2m (18%). The proportion with 30% eGFR decline was lowest with eGFRcr and highest with eGFRcys, with greater incidence in older age groups for all markers. Limitations No direct measurement of GFR. Not all participants survived or attended subsequent follow-up visits. Conclusions The prevalence and progression of CKD increase with age, but estimates vary with the filtration marker used. The eGFRcr gave the lowest estimate of CKD at 15% for people aged 65-69 years at Visit 5 while eGFRcys gave the highest estimates of CKD at 26% for that same population.
Collapse
Affiliation(s)
- Carina M Flaherty
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Aditya Surapaneni
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jesse C Seegmiller
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Josef Coresh
- Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY
| | - Morgan E Grams
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Shoshana H Ballew
- Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
11
|
Tungsanga S, Fung W, Okpechi IG, Ye F, Ghimire A, Li PKT, Shlipak MG, Tummalapalli SL, Arruebo S, Caskey FJ, Damster S, Donner JA, Jha V, Levin A, Saad S, Tonelli M, Bello AK, Johnson DW. Organization and Structures for Detection and Monitoring of CKD Across World Countries and Regions: Observational Data From a Global Survey. Am J Kidney Dis 2024; 84:457-468.e1. [PMID: 38788792 DOI: 10.1053/j.ajkd.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 05/26/2024]
Abstract
RATIONALE & OBJECTIVE Established therapeutic interventions effectively mitigate the risk and progression of chronic kidney disease (CKD). Countries and regions have a compelling need for organizational structures that enable early identification of people with CKD who can benefit from these proven interventions. We report the current global status of CKD detection programs. STUDY DESIGN A multinational cross-sectional survey. SETTING & PARTICIPANTS Stakeholders, including nephrologist leaders, policymakers, and patient advocates from 167 countries, participating in the International Society of Nephrology (ISN) survey from June to September 2022. OUTCOME Structures for the detection and monitoring of CKD, including CKD surveillance systems in the form of registries, community-based detection programs, case-finding practices, and availability of measurement tools for risk identification. ANALYTICAL APPROACH Descriptive statistics. RESULTS Of all participating countries, 19% (n=31) reported CKD registries, and 25% (n=40) reported implementing CKD detection programs as part of their national policies. There were variations in CKD detection program, with 50% (n=20) using a reactive approach (managing cases as identified) and 50% (n=20) actively pursuing case-finding in at-risk populations. Routine case-finding for CKD in high-risk populations was widespread, particularly for diabetes (n=152; 91%) and hypertension (n=148; 89%). Access to diagnostic tools, estimated glomerular filtration rate (eGFR), and urine albumin-creatinine ratio (UACR) was limited, especially in low-income (LICs) and lower-middle-income (LMICs) countries, at primary (eGFR: LICs 22%, LMICs 39%, UACR: LICs 28%, LMICs 39%) and secondary/tertiary health care levels (eGFR: LICs 39%, LMICs 73%, UACR: LICs 44%, LMICs 70%), potentially hindering CKD detection. LIMITATIONS A lack of detailed data prevented an in-depth analysis. CONCLUSIONS This comprehensive survey highlights a global heterogeneity in the organization and structures (surveillance systems and detection programs and tools) for early identification of CKD. Ongoing efforts should be geared toward bridging such disparities to optimally prevent the onset and progression of CKD and its complications. PLAIN-LANGUAGE SUMMARY Early detection and management of chronic kidney disease (CKD) is crucial to prevent progression to kidney failure. A multinational survey across 167 countries revealed disparities in CKD detection programs. Only 19% reported CKD registries, and 25% implemented detection programs as part of their national policy. Half used a reactive approach while others actively pursued case-finding in at-risk populations. Routine case-finding was common for individuals with diabetes and hypertension. However, limited access to gold standard tools such as estimated glomerular filtration rate (eGFR) and urine albumin-creatinine ratio (UACR), especially in low-income and lower-middle income countries, may hinder CKD detection. A global effort to bridge these disparities is needed to optimally prevent the onset and progression of CKD and its complications.
Collapse
Affiliation(s)
- Somkanya Tungsanga
- Division of Nephrology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Division of General Internal Medicine-Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Winston Fung
- Department of Medicine & Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Ikechi G Okpechi
- Division of Nephrology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Division of Nephrology and Hypertension and Kidney and Hypertension Research Unit, University of Cape Town, Cape Town, South Africa
| | - Feng Ye
- Division of Nephrology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anukul Ghimire
- Division of Nephrology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Philip Kam-Tao Li
- Department of Medicine & Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, University of California at San Francisco, San Francisco, California; General Internal Medicine Division, Medical Service, San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Sri Lekha Tummalapalli
- Division of Healthcare Delivery Science and Innovation, Department of Population Health Sciences, Weill Cornell Medicine, New York, New York; Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, New York
| | | | - Fergus J Caskey
- Population Health Sciences, Bristol Medical School, University of Bristol, United Kingdom
| | | | - Jo-Ann Donner
- International Society of Nephrology, Brussels, Belgium
| | - Vivekanand Jha
- School of Public Health, Imperial College, London, United Kingdom; George Institute for Global Health, University of New South Wales, New Delhi, India; Manipal Academy of Higher Education, Manipal, India
| | - Adeera Levin
- Division of Nephrology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Syed Saad
- Division of Nephrology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Marcello Tonelli
- Canada and Pan-American Health Organization/World Health Organization's Collaborating Centre in Prevention and Control of Chronic Kidney Disease, University of Calgary, Calgary, Alberta, Canada; Division of Nephrology, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Aminu K Bello
- Division of Nephrology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - David W Johnson
- Department of Kidney and Transplant Services and Centre for Kidney Disease Research, University of Queensland at Princess Alexandra Hospital, Translational Research Institute, Brisbane, Australia; Australasian Kidney Trials Network at the University of Queensland, Brisbane, Australia
| |
Collapse
|
12
|
Ebert N, Schaeffner E, Seegmiller JC, van Londen M, Bökenkamp A, Cavalier E, Delanaye P, Derain-Dubourg L, Eriksen BO, Indridason OS, Palsson R, Shafi T, Christensson A, Bevc S, Carrara F, Courbebaisse M, Dalton RN, van der Giet M, Melsom T, Methven S, Nordin G, Pottel H, Rule AD, Trillini M, White CA. Iohexol plasma clearance measurement protocol standardization for adults: a consensus paper of the European Kidney Function Consortium. Kidney Int 2024; 106:583-596. [PMID: 39097002 DOI: 10.1016/j.kint.2024.06.029] [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: 02/20/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 08/05/2024]
Abstract
International consensus supports the development of standardized protocols for measured glomerular filtration rate (mGFR) to facilitate the integration of mGFR testing in both clinical and research settings. To this end, the European Kidney Function Consortium convened an international group of experts with relevant experience in mGFR. The working group performed an extensive literature search to inform the development of recommendations for mGFR determination using 1-compartment plasma clearance models and iohexol as the exogenous filtration marker. Iohexol was selected as it is non-radio labeled, inexpensive, and safe, can be assayed at a central laboratory, and the other commonly used non-radio-labeled tracers have been (inulin) or are soon to be (iothalamate) discontinued. A plasma clearance model was selected over urine clearance as it requires no urine collection. A 1 compartment was preferred to 2 compartments as it requires fewer samples. The recommendations are based on published evidence complemented by expert opinion. The consensus paper covers practical advice for patients and health professionals, preparation, administration, and safety aspects of iohexol, laboratory analysis, blood sample collection and sampling times using both multiple and single-sample protocols, description of the mGFR mathematical calculations, as well as implementation strategies. Supplementary materials include patient and provider information sheets, standard operating procedures, a study protocol template, and support for mGFR calculation.
Collapse
Affiliation(s)
- Natalie Ebert
- Charité Universitätsmedizin Berlin, Institute of Public Health, Berlin, Germany
| | - Elke Schaeffner
- Charité Universitätsmedizin Berlin, Institute of Public Health, Berlin, Germany
| | - Jesse C Seegmiller
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Marco van Londen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, the Netherlands
| | - Arend Bökenkamp
- Department of Pediatric Nephrology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, Centre Hospitalier Universitaire du Sart-Tilman, Liège, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, Centre Hospitalier Universitaire du Sart-Tilman, Liège (ULiege), Belgium; Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| | - Laurence Derain-Dubourg
- Service de Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Centre de Référence des Maladies Rénales Rares, Service de Néphrologie et Rhumatologie Pédiatriques, Hospices Civils de Lyon, Lyon, France
| | - Bjørn O Eriksen
- Section of Nephrology, University Hospital of North Norway and Metabolic and Renal Research Group, UiT, The Arctic University of Norway, Tromsø, Norway
| | | | - Runolfur Palsson
- Division of Nephrology, Landspitali University Hospital, Reykavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Tariq Shafi
- Division of Kidney Diseases, Hypertension and Transplantation, Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Anders Christensson
- Department of Nephrology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Sebastjan Bevc
- Department of Nephrology, Department of Pharmacology, University Medical Centre Maribor, Maribor, Slovenia; Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Fabiola Carrara
- Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marie Courbebaisse
- Université Paris Cité; Physiology Department, Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - R Neil Dalton
- The WellChild Laboratory, Evelina London Children's Hospital, London, UK
| | - Markus van der Giet
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Toralf Melsom
- Section of Nephrology, University Hospital of North Norway and Metabolic and Renal Research Group, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Shona Methven
- Department of Renal Medicine, Aberdeen Royal Infirmary, Scotland, UK
| | | | - Hans Pottel
- Department of Public Health and Primary Care, Katholieke Universiteit Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Matias Trillini
- Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Christine A White
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
13
|
Liyanarachi KV, Mohus RM, Rogne T, Gustad LT, Åsvold BO, Romundstad S, Solligård E, Hallan S, Damås JK. Chronic kidney disease and risk of bloodstream infections and sepsis: a 17-year follow-up of the population-based Trøndelag Health Study in Norway. Infection 2024; 52:1983-1993. [PMID: 38679665 PMCID: PMC11499395 DOI: 10.1007/s15010-024-02265-2] [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: 12/05/2023] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE Bloodstream infections (BSI) and sepsis are important causes of hospitalization, loss of health, and death globally. Targetable risk factors need to be identified to improve prevention and treatment. In this study, we aimed to evaluate the association of chronic kidney disease (CKD) and risk of and mortality from BSI and sepsis in the general population during a 22-year period. METHODS We conducted a prospective cohort study among participants in the population-based Norwegian HUNT Study, where 68,438 participated. The median follow-up time was 17.4 years. The exposures were estimated glomerular filtration rate (eGFR) and albumin-creatinine ratio (ACR) in urine. The outcomes were hazard ratios (HR) of hospital admission or death due to BSI or sepsis. The associations were adjusted for age, sex, diabetes, obesity, systolic blood pressure, smoking status, and cardiovascular disease. RESULTS Participants with eGFR < 30 ml/min/1.732 had HR 3.35 for BSI (95% confidence intervals (CI) 2.12-5.3) and HR 2.94 for sepsis (95% CI 1.82-4.8) compared to normal eGFR (≥ 90 ml/min/1.732). HRs of death from BSI and sepsis were 4.2 (95% CI 1.71-10.4) and 4.1 (95% CI 1.88-8.9), respectively. Participants with severely increased albuminuria (ACR > 30 mg/mmol) had HR 3.60 for BSI (95% CI 2.30-5.6) and 3.14 for sepsis (95% CI 1.94-5.1) compared to normal albumin excretion (ACR < 3 mg/mmol). HRs of death were 2.67 (95% CI 0.82-8.7) and 2.16 (95% CI 0.78-6.0), respectively. CONCLUSION In this large population-based cohort study, CKD was clearly associated with an increased risk of BSI and sepsis and related death.
Collapse
Affiliation(s)
- Kristin Vardheim Liyanarachi
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.
- Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Randi Marie Mohus
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tormod Rogne
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Yale Department of Chronic Disease Epidemiology and Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Lise Tuset Gustad
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Bjørn Olav Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Solfrid Romundstad
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Erik Solligård
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Helse Møre Og Romsdal Hospital Trust, Ålesund, Norway
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jan Kristian Damås
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
14
|
Villain C, Ebert N, Glassock RJ, Mielke N, Bothe T, Barghouth MH, Pöhlmann A, Fietz AK, Gill JS, Schaeffner E. Medical Suitability and Willingness for Living Kidney Donation Among Older Adults. Am J Kidney Dis 2024:S0272-6386(24)00983-1. [PMID: 39362396 DOI: 10.1053/j.ajkd.2024.07.010] [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: 03/27/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 10/05/2024]
Abstract
RATIONALE & OBJECTIVE The benefits of kidney transplantation compared to treatment with dialysis, including in older adult, are primarily limited by the number of donated kidneys. We studied the potential to expand the use of older living kidney donors. STUDY DESIGN Secondary analysis of the Berlin Initiative Study, a population-based cohort. SETTING & Participants: 2069 adults aged ≥70 years in Germany. EXPOSURES Age and sex. OUTCOMES Suitability for living donation assessed by the absence of kidney-related exclusions for donation including albuminuria and low estimated glomerular filtration rate (eGFR) as well as absence of other medical exclusions. Willingness for living and deceased kidney donation assessed by participant survey. ANALYTICAL APPROACH Descriptive analysis. RESULTS Among the 2069 participants (median age 80 years, 53% women, median eGFR 63 ml/min/1.73m2), 93% had ≥1 medical contraindication for living donation at study entry unrelated to eGFR or albuminuria. Using two published eGFR and albuminuria thresholds for donor acceptance, 38% to 54% of participants had kidney-related exclusions for donation. Among the 5% to 6% of participants with neither medical nor kidney-related exclusions for living donation at baseline, 11% to 12% remained suitable for donation during 8 years of follow-up. Willingness for living or deceased donation was high (73% and 60%, respectively). LIMITATIONS GFR was not measured and medical exclusions unrelated to eGFR and albuminuria were assessed using a cohort database complemented by claims data. CONCLUSIONS One in twenty older adults were potentially suitable for living kidney donation and willingness for living donation was high. Further studies are warranted to define the feasibility of expanding living kidney donation among older adults.
Collapse
Affiliation(s)
- Cédric Villain
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany; Normandie Univ UNICAEN, INSERM U1075 COMETE, service de Gériatrie, CHU de Caen, Caen, France
| | - Natalie Ebert
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Richard J Glassock
- Department of Medicine, Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Nina Mielke
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Tim Bothe
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Muhammad Helmi Barghouth
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Anna Pöhlmann
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Anne-Katrin Fietz
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Charitéplatz 1, 10117 Berlin, Germany
| | - John S Gill
- Division of Nephrology, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Elke Schaeffner
- Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Public Health, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
15
|
Grosyeux C, Alla A, Barbé F, Dubourg LD, Chardon L, Guéant JL, Frimat L, Oussalah A, Vrillon I. The EKFC equation outperforms the CKD-EPI and CKiD equations for GFR estimation in adolescent and young adult kidney transplant patients. Nephrology (Carlton) 2024; 29:680-687. [PMID: 38803085 DOI: 10.1111/nep.14328] [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: 02/20/2024] [Revised: 04/28/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
AIM This study evaluated the bias and accuracy of the CKD-EPI/CKiD and EKFC equations compared with the reference exogenous tracer-based assessment of glomerular filtration rate (GFR) in adult and pediatric patients according to their renal transplant status. METHODS We assessed the bias and P30 accuracy of the CKD-EPI/CKiD and EKFC equations compared with iohexol-based GFR measurement. RESULTS In the overall population (n = 59), the median age was 29 years (IQR, 16.0-46.0) and the median measured GFR was 73.9 mL/min/1.73m2 (IQR, 57.3-84.6). Among non-kidney transplant patients, the median was 77.7 mL/min/1.73m2 (IQR, 59.3-86.5), while among kidney transplant patients, it was 60.5 mL/min/1.73m2 (IQR, 54.2-66.8). The bias associated with the EKFC and CKD-EPI/CKiD equations was significantly higher among kidney transplant patients than among non-kidney transplant patients, with a difference between medians (Hodges-Lehmann) of +10.4 mL/min/1.73m2 (95% CI, 2.2-18.9; p = .02) for the EKFC and +12.1 mL/min/1.73m2 (95% CI, 4.2-21.4; p = .006) for the CKD-EPI/CKiD equations. In multivariable analysis, kidney transplant status emerged as an independent factor associated with a bias of >3.4 mL/min/1.73m2 (odds ratio, 7.7; 95% CI, 1.4-43.3; p = .02) for the EKFC equation and a bias of >13.4 mL/min/1.73m2 (odds ratio, 15.0; 95% CI, 2.6-85.7; p = .002) for the CKD-EPI/CKiD equations. CONCLUSION In our study, which included adolescent and young adult kidney transplant patients, both the CKD-EPI/CKiD and EKFC equations tended to overestimate the measured glomerular filtration rate, with the EKFC equation exhibiting less bias. Renal transplant status significantly influenced the degree of estimation bias.
Collapse
Affiliation(s)
- Chloé Grosyeux
- Pediatric Nephrology Department, University Hospital of Nancy, Nancy, France
| | - Asma Alla
- Department of Nephrology, University of Lorraine, Nancy, France
| | - Françoise Barbé
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France
- Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy, France
| | - Laurence Derain Dubourg
- Nephrology, Dialysis, Hypertension and Functional Renal Exploration, Edouard Herriot Hospital, Hospices Civils de Lyon and Université Lyon 1, Lyon, France
| | - Laurence Chardon
- Department of Biology and Hormonology, Lyon-Est Hospital, Bron, France
| | - Jean-Louis Guéant
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France
- Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy, France
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Nancy, France
| | - Luc Frimat
- Department of Nephrology, University of Lorraine, Nancy, France
- INSERM CIC-EC CIE6, University of Lorraine, Nancy, France
| | - Abderrahim Oussalah
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France
- Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy, France
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Nancy, France
| | - Isabelle Vrillon
- Pediatric Nephrology Department, University Hospital of Nancy, Nancy, France
| |
Collapse
|
16
|
Liguori A, D'Ambrosio F, Napodano C, Gentili V, Giustiniani MC, Pompili M, Grieco A, Rapaccini G, Urbani A, Gasbarrini A, Basile U, Miele L. Serum-free light chains as a dependable biomarker for stratifying patients with metabolic dysfunction-associated steatotic liver disease. Liver Int 2024; 44:2625-2638. [PMID: 39016540 DOI: 10.1111/liv.16036] [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: 11/04/2023] [Revised: 05/29/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND AND AIMS Adaptive immunity is gaining a significant role in progression of metabolic dysfunction-associated steatotic liver disease (MASLD). B-cell activity can be assessed by serum-free light chains (sFLCs) k and λ levels. The objective of the present investigation is to examine the utility of sFLCs as non-invasive biomarkers for the stratification of MASLD. METHODS We enrolled a consecutive cohort from an outpatient liver unit. Diagnosis of metabolic dysfunction-associated steatohepatitis (MASH) was made with liver biopsy according to current guidelines. Compensated advanced chronic liver disease (cACLD) and clinically significant portal hypertension (CSPH) were defined according to Baveno VII criteria. sFLCs were measured by turbidimetry using an immunoassay. RESULTS We evaluated 254 patients, 162/254 (63.8%) were male. Median age was 54 years old, and the median body mass index was 28.4 kg/m2. A total of 157/254 (61.8%) subjects underwent liver biopsy: 88 had histological diagnosis of MASH, 89 were considered as simple metabolic dysfunction-associated steatotic liver (MASL) and 77/254 (30.3%) patients with compensated metabolic dysfunction-associated cirrhosis. By using Baveno VII criteria, 101/254 (39.7%) patients had cACLD; among them, 45/101 (44.5%) had CSPH. Patients with cACLD showed higher sFLC levels compared with patients without cACLD (p < .01), and patients with CSPH showed higher sFLC levels than patients without CSPH (p < .01). At multivariable analysis, sFLCs were associated with cACLD (p < .05) independently from γ-globulins and other known dysmetabolic risk factors. κFLC was associated with CSPH (p < .05) independently from γ-globulins and other known dysmetabolic risk factors. CONCLUSION sFLCs could be a simple biomarker for stratification of cACLD in MASLD patients.
Collapse
Affiliation(s)
- Antonio Liguori
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- CEMAD, Digestive Disease Center, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Francesca D'Ambrosio
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cecilia Napodano
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanessa Gentili
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Cristina Giustiniani
- Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Maurizio Pompili
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Grieco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gianludovico Rapaccini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Urbani
- Department of Laboratory and Infectious Diseases Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti HospitalAUSL Latina, Latina, Italy
| | - Luca Miele
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Internal Medicine and Liver Transplant Unit, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- CEMAD, Digestive Disease Center, Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Turino Miranda K, Dumanski SM, Saad N, Inker LA, White CA, Delanaye P, Collister D, Greene DN, Whitley CT, Harrison TG, Rytz CL, Peace L, Sola DY, Ahmed SB. Glomerular filtration rate estimation in transgender and gender-diverse adults using gender-affirming hormone therapy: an exploratory cross-sectional study. Kidney Int 2024; 106:753-756. [PMID: 39142563 DOI: 10.1016/j.kint.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/29/2024] [Accepted: 06/14/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Keila Turino Miranda
- Cardiovascular Health and Autonomic Regulation Laboratory, Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Sandra M Dumanski
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Nathalie Saad
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Tufts University, Boston, Massachusetts, USA
| | - Christine A White
- Division of Nephrology, Queen's University, Kingston, Ontario, Canada
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium; Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| | - David Collister
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Dina N Greene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Cameron T Whitley
- Department of Sociology, Western Washington University, Bellingham, Washington, USA
| | - Tyrone G Harrison
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Chantal L Rytz
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | | | - Darlene Y Sola
- Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sofia B Ahmed
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
18
|
Stehlé T, Delanaye P. Which is the best glomerular filtration marker: Creatinine, cystatin C or both? Eur J Clin Invest 2024; 54:e14278. [PMID: 38949475 DOI: 10.1111/eci.14278] [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: 04/25/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND The glomerular filtration rate (GFR) is estimated by the serum or plasma concentration of creatinine and/or cystatin C using equations that include demographic data. The equations worldwide most widely used are those of the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) consortium and updated in 2021 to remove the Afro-American racial correction factor. In 2021 and then in 2023, the European Kidney Function Consortium also developed equations based on creatinine and cystatin C, usable across the full age spectrum, and constructed by including the Q value (i.e. the median creatinine or cystatin C in healthy men and women, which is customizable for specific populations). METHODS The aim of this narrative review is to examine the strengths and weaknesses of each biomarker. RESULTS Both biomarkers have non-GFR determinants, namely muscle mass, protein intake and tubular secretion for creatinine; dysthyroidism and systemic corticosteroids for cystatin C, as well as other more debated determinants (diabetes, obesity, proteinuria, inflammatory syndrome). These non-GFR determinants are the reason why no equation based on a single endogenous biomarker has an accuracy within 30% greater than 90% over the entire age spectrum (in at least one patient in 10, estimated GFR is at least 30% higher or at least 30% lower than the measured GFR). CONCLUSION Equations combining the two biomarkers provide a better estimate of GFR, particularly in the subgroup of patients whose estimates based on each of the biomarkers are highly discordant. These patients must also be identified as being at increased risk of morbidity, particularly cardiovascular, and mortality.
Collapse
Affiliation(s)
- Thomas Stehlé
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Fédération Hospitalo-Universitaire «Innovative therapy for immune disorders», Créteil, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U955, Institut Mondor de Recherche Biomédicale (IMRB), Univ. Paris Est Créteil, Créteil, France
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| |
Collapse
|
19
|
Chen L, Jiang Y, Cheng X. Associations Between CYP3A5 (c.6986A>G) Gene Polymorphism and Kidney Impairment in Hypertensive Adults Without Cystatin C Elevation. Cardiovasc Toxicol 2024; 24:1047-1052. [PMID: 39095622 DOI: 10.1007/s12012-024-09902-0] [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: 04/25/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE This study aimed to explore the potential role of CYP3A5 (c. 6986A>G) gene polymorphism in predicting kidney function impairment in patients with hypertension who did not have elevated serum cystatin C. METHODS We recruited a group of patients with hypertension who did not have elevated cystatin C and analyzed the CYP3A5 (c. 6986A>G) gene polymorphism. Chi-square tests were used to compare the clinical characteristics and genotypic distribution between the two groups. Logistic regression analysis was used to explore the association between CYP3A5 (c.6986A>G) gene polymorphism and renal function impairment in hypertension with non-elevated cystatin. RESULTS In patients with hypertension who participated in the study, there was a significant association between CYP3A5 (c. 6986A>G) gene polymorphism and kidney function impairment (p < 0.05). Patients with the CYP3A5 (c. 6986A>G) mutation display a greater risk of kidney function impairment. CONCLUSION CYP3A5 (c. 6986A>G) gene AA homozygote polymorphism significantly increases risk of kidney function impairment in patients with hypertension with normal cystatin C. However, further studies are needed to validate this association and to further understand the mechanism of CYP3A5 (c. 6986A>G) gene polymorphism in kidney function impairment in patients with hypertension.
Collapse
Affiliation(s)
- Ling Chen
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou City, 215000, Jiangsu Province, People's Republic of China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou City, 215125, Jiangsu Province, People's Republic of China.
| | - Xingbo Cheng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou City, 215000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
20
|
Maringhini S, Zoccali C. Chronic Kidney Disease Progression-A Challenge. Biomedicines 2024; 12:2203. [PMID: 39457516 PMCID: PMC11505431 DOI: 10.3390/biomedicines12102203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition characterized by a continuous decline in renal function, independent of the initial cause of damage or external factors such as infection, inflammation, or toxins. The accurate measurement of renal function, typically assessed using the glomerular filtration rate (GFR), is crucial for managing CKD. The most accepted hypothesis for CKD progression is glomerular damage caused by hyperfiltration. Various factors can accelerate CKD progression, and several biomarkers have been identified to monitor this progression. Numerous studies have explored the risk factors associated with CKD progression, and some of these factors can be modified. Additionally, several drugs are now available that can reduce CKD progression. This review summarizes recent publications and highlights potential future research directions in CKD progression. It discusses the evolution of GFR measurement methods, the mechanisms driving CKD progression, and the latest findings on biomarkers and risk factors. Furthermore, it explores therapeutic strategies, including dietary modifications and pharmacological interventions, to slow CKD progression. Understanding these mechanisms and interventions is crucial for developing effective therapeutic strategies to prevent or slow CKD progression.
Collapse
Affiliation(s)
- Silvio Maringhini
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), 90127 Palermo, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, NY 10065, USA;
- Institute of Molecular Biology and Genetics (Biogem), 83031 Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, 89124 Reggio Calabria, Italy
| |
Collapse
|
21
|
Wang ZS, Wang SF, Zhao MY, He QN. [Current clinical application of glomerular filtration rate assessment methods in pediatric populations]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1002-1008. [PMID: 39267519 PMCID: PMC11404467 DOI: 10.7499/j.issn.1008-8830.2401011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Glomerular filtration rate (GFR) is a critical indicator of renal function assessment, which exhibits age-dependency in children and may differ from adults under various disease conditions. In recent years, there has been a growing focus on GFR among scholars, with an increasing number of clinical studies dedicated to refining and optimizing GFR estimation to span all pediatric age groups. However, the methods and assessment equations for estimating GFR may vary under different disease conditions, affecting the accuracy and applicability of assessments. This article reviews the peculiarities of renal function in children, explores GFR measurement methods, and evaluates the application of various GFR assessment equations in pediatric clinical practice, providing a reference for clinical assessment of renal function in children.
Collapse
Affiliation(s)
- Zi-Sai Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Sheng-Feng Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Ming-Yi Zhao
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Qing-Nan He
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| |
Collapse
|
22
|
Muraglia A, Utyro O, Nardini M, Santolini M, Ceresa D, Agostini V, Nencioni A, Filaci G, Cancedda R, Mastrogiacomo M. A simple cell proliferation assay and the inflammatory protein content show significant differences in human plasmas from young and old subjects. Front Bioeng Biotechnol 2024; 12:1408499. [PMID: 39351061 PMCID: PMC11440192 DOI: 10.3389/fbioe.2024.1408499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Some studies showed a "rejuvenating" effect of exposing aging tissues to a young environment. In mouse heterochronic parabiosis experiments, in response to young organisms, old animals lived longer than isochrony old age-matched conjoint animals. Comparable "rejuvenating" effects were obtained by injecting young plasma in old mice. This raised great hopes of slowing down the senescence process in humans by the injection of young plasma, as well as to prevent or cure age-related diseases. Some clinical trials are currently being performed or were recently completed. However, these studies are small and of limited duration, and we still lack convincing evidence to support the effectiveness of young plasma injection. It is urgent to perform additional investigations, including the development of an assay to measure the cell proliferation induction capability of different human plasmas, before one can seriously think of a large-scale treatment of humans. We adopted a simple method to measure the potential of different plasmas in supporting cell line proliferation, regardless of the co-presence of a platelet lysate. By comparing plasmas from young and old subjects, we observed a decreased activity in plasmas from old individuals. The young plasma effect may be attributed to specific proteins and growth factors more abundant in younger individuals that could decrease with age. Alternatively, or at the same time, the reduced cell proliferation support could be due to inhibitors present in the old plasma. Studying the different protein content of young and old plasmas was out of the scope of this article. Such differences should be adequately investigated by proteomics using many samples. However, a preliminary study of the different protein content of young and old plasmas was part of the assay validation using a commercially available cytokine array for parallel determination of the relative levels of 105 selected human proteins. We could show the existence of specific differences between young and old plasmas and that plasmas from old individuals presented a higher concentration of "inflammatory" proteins.
Collapse
Affiliation(s)
- A. Muraglia
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
| | - O. Utyro
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
| | - M. Nardini
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
| | - M. Santolini
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
| | - D. Ceresa
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - V. Agostini
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - A. Nencioni
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - G. Filaci
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - R. Cancedda
- Dipartimento di Medicina Sperimentale (DIMES), Università Degli Studi di Genova, Genova, Italy
| | - M. Mastrogiacomo
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
23
|
Delanaye P, Derain-Dubourg L, Björk J, Courbebaisse M, Couzi L, Gaillard F, Garrouste C, Grubb A, Jacquemont L, Hansson M, Kamar N, Legendre C, Littmann K, Mariat C, Rostaing L, Rule AD, Sundin PO, Bökenkamp A, Berg U, Åsling-Monemi K, Åkesson A, Larsson A, Nyman U, Pottel H. Estimating glomerular filtration in young people. Clin Kidney J 2024; 17:sfae261. [PMID: 39314869 PMCID: PMC11418036 DOI: 10.1093/ckj/sfae261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 09/25/2024] Open
Abstract
Background Creatinine-based equations are the most used to estimate glomerular filtration rate (eGFR). The Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI), the re-expressed Lund-Malmö Revised (r-LMR) and the European Kidney Function Consortium (EKFC) equations are the most validated. The EKFC and r-LMR equations have been suggested to have better performances in young adults, but this is debated. Methods We collected data (GFR) measured by clearance of an exogenous marker (reference method), serum creatinine, age and sex from 2366 young adults (aged between 18 and 25 years) both from Europe and the USA. Results In the European cohorts (n = 1892), the bias (in mL/min/1.73 m²) was systematically better for the EKFC and r-LMR equations compared with the CKD-EPI equation [2.28, 95% confidence interval (1.59; 2.91), -2.50 (-3.85; -1.76), 17.41 (16.49; 18.47), respectively]. The percentage of estimated GFR within 30% of measured GFR (P30) was also better for EKFC and r-LMR equations compared with the CKD-EPI equation [84.4% (82.8; 86.0), 87.2% (85.7; 88.7) and 65.4% (63.3; 67.6), respectively]. In the US cohorts (n = 474), the bias for the EKFC and r-LMR equations was better than for the CKD-EPI equation in the non-Black population [0.97 (-1.69; 3.06), -2.62 (-5.14; -1.43) and 7.74 (5.97; 9.63), respectively], whereas the bias was similar in Black US individuals. P30 results were not different between the three equations in US cohorts. Analyses in sub-populations confirmed these results, except in individuals with high GFR levels (GFR ≥120 mL/min/1.73 m²) for whom the CKD-EPI equation might have a lower bias. Conclusions We demonstrated that both the EKFC and r-LMR creatinine-based equations have a better performance than the CKD-EPI equation in a young population. The only exception might be in patients with hyperfiltration.
Collapse
Affiliation(s)
- Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hopital Universitaire Caremeau, Nimes, France
| | - Laurence Derain-Dubourg
- Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Jonas Björk
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Marie Courbebaisse
- Physiology Department, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris, Paris Cité University, INSERM U1151-CNRS UMR8253, Paris, France
| | - Lionel Couzi
- CHU de Bordeaux, Nephrologie – Transplantation – Dialyse, Université de Bordeaux, CNRS-UMR 5164 Immuno ConcEpT, Bordeaux, France
| | - Francois Gaillard
- AURAL, Association pour l'utilisation du rein artificiel dans la région lyonnaise, Lyon, France
| | - Cyril Garrouste
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund University, Lund, Sweden
| | - Lola Jacquemont
- Renal Transplantation Department, CHU Nantes, Nantes University, Nantes, France
| | - Magnus Hansson
- Function area Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital Huddinge and Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Nassim Kamar
- Department of Nephrology, Dialysis and Organ Transplantation, CHU Rangueil, INSERM U1043, IFR – BMT, University Paul Sabatier, Toulouse, France
| | | | - Karin Littmann
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Christophe Mariat
- Service de Néphrologie, Dialyse et Transplantation Rénale, Hôpital Nord, CHU de Saint-Etienne, Saint-Etienne, France
| | - Lionel Rostaing
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, Hôpital Michallon, CHU Grenoble-Alpes, Grenoble, France
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Per-Ola Sundin
- Karla Healthcare Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Arend Bökenkamp
- Department of Paediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ulla Berg
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Kajsa Åsling-Monemi
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Anna Åkesson
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Ulf Nyman
- Department of Translational Medicine, Division of Medical Radiology, Lund University, Malmö, Sweden
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| |
Collapse
|
24
|
Ashouri R, Lertkitcharoenpon A, Maidaa M, Brunner B, Velazquez O, Lipshultz L, Campbell K. Creatinine and Cystatin C: A Measure of Renal Function in Men With Testosterone-Induced Muscle Hypertrophy. Am J Mens Health 2024; 18:15579883241286654. [PMID: 39402920 PMCID: PMC11483778 DOI: 10.1177/15579883241286654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Creatinine (Cr) is often used as a standalone gold standard marker of kidney function. Cystatin C (Cys C) is a less physiologically labile marker of renal function, particularly in certain subgroups. Herein, we analyze trends in cystatin C as compared to creatinine in men on testosterone replacement therapy with varying body mass indices and percent body fat (PBF). This retrospective analysis observes 227 men with testosterone-induced muscle hypertrophy who visited a men's health tertiary care clinic. All participants were characterized as competitive or recreational athletes. In patients with a normal body mass index (BMI), there was no clinically significant correlation between Cr and Cys C. Slight correlation was seen with overweight (R2 = .27) patients (p < .0001) and obese (R2 = .29) patients (p < .0001). Patients with PBF of 0%-10% (n = 22) exhibited minimal (R2 = .23) positive correlation between Cys C and Cr (p = .03). Positive correlation between Cys C and Cr in patients with PBF of 10%-20% was clinically negligible (R2 = .17, n = 87), modest (R2 = .49) in patients with PBF of 20%-30% (n = 42), and evident (R2 = 1.00) in patients >30% (n = 3) (p < .0001, respectively). Cystatin C measurements display less variance compared with creatinine at differing BMI distinctions. At the upper limit of BMI or PBF in our patient population, cystatin C exhibits minimal to moderate variability compared with creatinine.
Collapse
Affiliation(s)
- Rani Ashouri
- Department of Urology, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | - Michael Maidaa
- Department of Urology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bobby Brunner
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Oscar Velazquez
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Larry Lipshultz
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Kevin Campbell
- Department of Urology, College of Medicine, University of Florida, Gainesville, FL, USA
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Costa E Silva VT, Gil LA, Inker LA, Caires RA, Costalonga E, Coura-Filho G, Sapienza MT, Castro G, Estevez-Diz MDP, Zanetta DMT, Antonângelo L, Marçal L, Tighiouart H, Miao S, Mathew P, Levey AS, Burdmann EA. Glomerular Filtration Rate Estimation Using β 2-Microglobulin and β-Trace Protein in Adults With Solid Tumors: A Prospective Cross-Sectional Study. Am J Kidney Dis 2024; 84:339-348.e1. [PMID: 38537905 DOI: 10.1053/j.ajkd.2024.01.532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/06/2023] [Accepted: 01/02/2024] [Indexed: 05/26/2024]
Abstract
RATIONALE & OBJECTIVE β2-Microglobulin (B2M) and β-trace protein (BTP) are novel endogenous filtration markers that may improve the accuracy of estimated glomerular filtration rate (eGFR) beyond creatinine and cystatin C (eGFRcr-cys), but they have not been assessed in patients with cancer. STUDY DESIGN Cross-sectional analysis. SETTING & PARTICIPANTS Prospective cohort of 1,200 patients with active solid tumors recruited between April 2015 and September 2017. EXPOSURE CKD-EPI equations without race combining B2M and/or BTP with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR). OUTCOME Performance of equations compared with eGFRcr-cys and non-GFR determinants of serum B2M and BTP (SB2M, and SBTP, respectively). Measured GFR (mGFR) was determined using the plasma clearance of chromium-51 labeled ethylenediamine tetraacetic acid (51Cr-EDTA). ANALYTICAL APPROACH Bias was defined as the median of the differences between mGFR and eGFR, and 1-P30 was defined as the percentage of estimates that differed by more than 30% from the mGFR (1-P30). Linear regression was used to assess association of clinical and laboratory variables with SB2M, and SBTP after adjustment for mGFR. RESULTS Mean age and mGFR were 58.8±13.2 SD years and 78.4±21.7 SD mL/min/1.73m2, respectively. Performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys (lesser bias and 1-P30). Performance of 2-marker panel equations was as good as eGFRcr-cys (lesser bias and similar 1-P30). SB2M and SBTP were not strongly influenced by cancer site. LIMITATIONS Participants may have had better clinical performance status than the general population of patients with solid tumors. CONCLUSIONS B2M and BTP can improve the accuracy of eGFR and may be useful as confirmatory tests in patients with solid tumors, either by inclusion in a multimarker panel equation with creatinine and cystatin C, or by substituting for cystatin C in combination with creatinine. PLAIN-LANGUAGE SUMMARY The most accurate method to assess estimate kidney function is estimated glomerular filtration rate (eGFR) using creatinine and cystatin C (eGFRcr-cys). We studied whether using β2-microglobulin (B2M) and/or β-trace protein (BTP) with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR) might be useful in patients with active solid tumors. The performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys. Performance of 2-marker panel equations was as good as eGFRcr-cys. We conclude that B2M and BTP can improve the accuracy of eGFR and may be useful as a confirmatory test in patients with solid tumors either by inclusion in multimarker panel equation with creatinine and cystatin C or by substituting for cystatin C in combination with creatinine.
Collapse
Affiliation(s)
- Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica (LIM) 16, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Luiz A Gil
- Laboratório de Investigação Médica (LIM) 66, Serviço de Geriatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Elerson Costalonga
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - George Coura-Filho
- Serviço de Medicina Nuclear, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo T Sapienza
- Radiology and Oncology Department, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto Castro
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Maria D P Estevez-Diz
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Dirce Maria T Zanetta
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Leila Antonângelo
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lia Marçal
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Biostatistics, Epidemiology, and Research Design Center, Tufts Medical Center, Boston, Massachusetts
| | - Shiyuan Miao
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Paul Mathew
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica (LIM) 12, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Fujii R, Melotti R, Köttgen A, Teumer A, Giardiello D, Pattaro C. Integrating multiple kidney function markers to predict all-cause and cardiovascular disease mortality: prospective analysis of 366 758 UK Biobank participants. Clin Kidney J 2024; 17:sfae207. [PMID: 39135936 PMCID: PMC11317837 DOI: 10.1093/ckj/sfae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Indexed: 08/15/2024] Open
Abstract
Background Reduced kidney function is a risk factor of cardiovascular and all-cause mortality. This association was demonstrated for several kidney function markers, but it is unclear whether integrating multiple measured markers may improve mortality risk prediction. Methods We conducted an exploratory factor analysis (EFA) of serum creatinine- and cystatin C-based estimated glomerular filtration rate [eGFRcre and eGFRcys; derived by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) and European Kidney Function Consortium (EKFC) equations], blood urea nitrogen (BUN), uric acid and serum albumin among 366 758 participants in the UK Biobank without a history of kidney failure. Fitting Cox proportional hazards models, we compared the ability of the identified latent factors to predict overall mortality and mortality by cardiovascular disease (CVD), also considering CVD-specific causes like coronary heart disease (CHD) and cerebrovascular disease. Results During 12.5 years of follow-up, 26 327 participants died from any cause, 5376 died from CVD, 2908 died from CHD and 1116 died from cerebrovascular disease. We identified two latent factors, EFA1 and EFA2, both representing kidney function variations. When using the CKD-EPI equation, EFA1 performed like eGFRcys, with EFA1 showing slightly larger hazard ratios for overall and CVD-related mortality. At 10 years of follow-up, EFA1 and eGFRcys showed moderate discrimination performance for CVD-related mortality, outperforming all other kidney indices. eGFRcre was the least predictive marker across all outcomes. When using the EKFC equation, eGFRcys performed better than EFA1 while all other results remaining similar. Conclusions While EFA is an attractive approach to capture the complex effects of kidney function, eGFRcys remains the most practical and effective measurement for all-cause and CVD mortality risk prediction.
Collapse
Affiliation(s)
- Ryosuke Fujii
- Institute for Biomedicine, Eurac Research, Bolzano/Bozen, Italy
- Department of Preventive Medical Science, Fujita Health University School of Medical Sciences, Toyoake, Japan
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Roberto Melotti
- Institute for Biomedicine, Eurac Research, Bolzano/Bozen, Italy
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany
| | | | | |
Collapse
|
27
|
Chen Y, Ma Y, Yong Z, Wei L, Pei X, Zhu B, Zhao W. Assessment of the 2023 European Kidney Function Consortium (EKFC) equations in a Chinese adult population. Clin Chem Lab Med 2024; 62:1570-1579. [PMID: 38336773 DOI: 10.1515/cclm-2024-0080] [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: 07/25/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES The European Kidney Function Consortium (EKFC) developed two novel equations in 2023 for estimating glomerular filtration rate (GFR): one sex-free cystatin C-based equation (EKFCCys) and one creatinine-cystatin C combined equation (EKFCCr-Cys). This study compared their performance with the previous creatinine-based EKFC equation (EKFCCr) and commonly used Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) and Berlin Initiative Study (BIS) equations in Chinese adults. METHODS A total of 2,438 Chinese adults (mean age=53.04 years) who underwent the 99mTc-DTPA renal dynamic imaging for reference GFR (rGFR) were included. Diagnostic value was evaluated using correlation coefficients, sensitivity, specificity, and area under the receiver operating characteristic curve (ROCAUC). Performance was assessed in terms of bias, precision (interquartile range of the median difference [IQR]), accuracy (percentage of estimates ±30 % of rGFR [P30], and root-mean-square error [RMSE]) across age, sex, and rGFR subgroups. Gender differences in bias and P30 were also analyzed. RESULTS Average rGFR was 73.37 mL/min/1.73 m2. EKFC equations showed stronger correlations and larger AUCs compared to the parallel CKD-EPI equations, with EKFCCr-Cys demonstrating the greatest improvement (R=0.771, ROCAUC=0.913). Concerning bias, precision, and accuracy, EKFC equations consistently outperformed CKD-EPI equations. EKFCCr-Cys and EKFCCr performed acceptably well in the entire population and were equivalent to BIS equations in the elderly. All equations, including EKFCCys, showed similar P30 accuracy across sexes. CONCLUSIONS EKFC equations provided a reasonable alternative for estimating GFR in the Chinese adult population. While EKFCCys did not outperform EKFCCr, EKFCCr-Cys improved the accuracy of single-marker equations.
Collapse
Affiliation(s)
- Yi Chen
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Yao Ma
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Zhenzhu Yong
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Lu Wei
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Xiaohua Pei
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Bei Zhu
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Weihong Zhao
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| |
Collapse
|
28
|
Carrara F, Gaspari F, Trillini M, Peracchi T, Fidone D, Stucchi N, Ferrari S, Cugini D, Perico N, Parvanova A, Remuzzi G, Ruggenenti P. GFR measurement in patients with CKD: Performance and feasibility of simplified iohexol plasma clearance techniques. PLoS One 2024; 19:e0306935. [PMID: 39018289 PMCID: PMC11253958 DOI: 10.1371/journal.pone.0306935] [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: 05/15/2023] [Accepted: 06/25/2024] [Indexed: 07/19/2024] Open
Abstract
Implementing shortened one-compartment iohexol plasma clearance models for GFR measurement is crucial since the gold standard inulin renal clearance technique and the reference two-compartment, 10-hour, 16-samplings iohexol plasma clearance method are clinically unfeasible. Inulin may precipitate anaphylactic shock. Four-hour and 8-hour one-compartment iohexol plasma clearance models with Bröchner-Mortensen correction provide accurate GFR measurements in patients with estimated GFR (eGFR) > or ≤40 mL/min/1.73m2, respectively. We compared the performance of the simplified 5-hour, 4-samplings, two-compartment population pharmacokinetic model (popPK) with the performance of the reference two-compartment 10-hour iohexol method in 16 patients with GFR 15.2 to 56.5 mL/min/1.73 m2. We also compared the performance of shortened (5, 6 and 7-hour) one-compartment models with the performance of the standard 8-hour one-compartment model in 101 patients with eGFR ≤40 mL/min/1.73 m2. The performance of popPK and shortened methods versus reference methods was evaluated by total deviation index (TDI), concordance correlation coefficient (CCC) and coverage probability (CP). TDI <10%, CCC ≥0.9 and CP >90% indicated adequate performance. TDI, CCC and CP of popPK were 11.11%, 0.809 and 54.10%, respectively. All shortened, one-compartment models overestimated the GFR (p <0.0001 for all) as compared to the 8-hour model. TDI, CCC and CP were 7.02%, 0.815, and 75.80% for the 7-hour model, 7.26%, 0.803, and 74.20% for the 6-hour model, and 8.85%, 0.729 and 64.70% for the 5-hour model. The agreement of popPK model was comparable to that obtained with the Chronic-Kidney-Disease-Collaboration-Epidemiology (CKD-Epi) and the Modification-of-Diet-in-Renal-Disease (MDRD) serum-creatinine based equations for GFR estimation. PopPK model is remarkably unreliable for GFR measurement in stage III-IV CKD patients. In patients with eGFR ≤40 mL/min/1.73m2, shortened one-compartment models, in particular the 5-hour model, are less performant than the reference 8-hour model. For accurate GFR measurements, the iohexol plasma clearance should be measured with appropriate protocols. Over-simplified procedures should be avoided.
Collapse
Affiliation(s)
- Fabiola Carrara
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Flavio Gaspari
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Matias Trillini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Tobia Peracchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Diego Fidone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Nadia Stucchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Silvia Ferrari
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Daniela Cugini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Aneliya Parvanova
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
| | - Piero Ruggenenti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases “Aldo e Cele Daccò”, Bergamo, Italy
- Unit of Nephrology, Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
29
|
Ng DK, Muñoz A. Assessing bias in GFR estimating equations: improper GFR stratification can yield misleading results. Pediatr Nephrol 2024; 39:2139-2145. [PMID: 38396091 PMCID: PMC11232499 DOI: 10.1007/s00467-024-06318-4] [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: 11/27/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Assessing bias (estimated - measured) is key to evaluating glomerular filtration rate (GFR). Stratification by subgroups can indicate where equations perform differently. However, there is a fallacy in the assessment of two instruments (e.g., eGFR and mGFR) when stratifying on the level of only one of those instruments. Here, we present statistical aspects of the problem and a solution for GFR stratification along with an empirical investigation using data from the CKiD study. METHODS Compared and contrasted biases (eGFR relative to mGFR) with 95% confidence intervals within strata of mGFR only, eGFR only, and the average of mGFR and eGFR using data from the Chronic Kidney Disease in Children (CKiD) study. RESULTS A total of 304 participants contributed 843 GFR studies with a mean mGFR of 48.46 (SD = 22.72) and mean eGFR of 48.67 (SD = 22.32) and correlation of 0.904. Despite strong agreement, eGFR significantly overestimated mGFR when mGFR < 30 (+ 6.2%; 95%CI + 2.9%, + 9.7%) and significantly underestimated when mGFR > 90 (-12.2%; 95%CI - 17.3%, - 7.0%). Significant biases in opposite direction were present when stratifying by eGFR only. In contrast, when stratifying by the average of eGFR and mGFR, biases were not significant (+ 1.3% and - 1.0%, respectively) congruent with strong agreement. CONCLUSIONS Stratifying by either mGFR or eGFR only to assess eGFR biases is ubiquitous but can lead to inappropriate inference due to intrinsic statistical issues that we characterize and empirically illustrate using data from the CKiD study. Using the average of eGFR and mGFR is recommended for valid inferences in evaluations of eGFR biases.
Collapse
Affiliation(s)
- Derek K Ng
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Room E7642, Baltimore, MD, 21205, USA.
| | - Alvaro Muñoz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Room E7642, Baltimore, MD, 21205, USA
| |
Collapse
|
30
|
Delanaye P, Pottel H, Cavalier E, Flamant M, Stehlé T, Mariat C. Diagnostic standard: assessing glomerular filtration rate. Nephrol Dial Transplant 2024; 39:1088-1096. [PMID: 37950562 DOI: 10.1093/ndt/gfad241] [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: 07/14/2023] [Indexed: 11/12/2023] Open
Abstract
Creatinine-based estimated glomerular filtration rate (eGFR) is imprecise at individual level, due to non-GFR-related serum creatinine determinants, including atypical muscle mass. Cystatin C has the advantage of being independent of muscle mass, a feature that led to the development of race- and sex-free equations. Yet, cystatin C-based equations do not perform better than creatinine-based equations for estimating GFR unless both variables are included together. The new race-free Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation had slight opposite biases between Black and non-Black subjects in the USA, but has poorer performance than that the previous version in European populations. The European Kidney Function Consortium (EKFC) equation developed in 2021 can be used in both children and adults, is more accurate in young and old adults, and is applicable to non-white European populations, by rescaling the Q factor, i.e. population median creatinine, in a potentially universal way. A sex- and race-free cystatin C-based EKFC, with the same mathematical design, has also be defined. New developments in the field of GFR estimation would be standardization of cystatin C assays, development of creatinine-based eGFR equations that incorporate muscle mass data, implementation of new endogenous biomarkers and the use of artificial intelligence. Standardization of different GFR measurement methods would also be a future challenge, as well as new technologies for measuring GFR. Future research is also needed into discrepancies between cystatin C and creatinine, which is associated with high risk of adverse events: we need to standardize the definition of discrepancy and understand its determinants.
Collapse
Affiliation(s)
- Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège (ULiege), CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège (ULiege), CHU Sart Tilman, Liège, Belgium
| | - Martin Flamant
- Assistance Publique-Hôpitaux de Paris, Bichat Hospital, and Université Paris Cité, UMR 1149, Paris, France
| | - Thomas Stehlé
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie et Transplantation, Fédération Hospitalo-Universitaire « Innovative therapy for immune disorders », Créteil, France
| | - Christophe Mariat
- Service de Néphrologie, Dialyse et Transplantation Rénale, Hôpital Nord, CHU de Saint-Etienne, France
| |
Collapse
|
31
|
Iversen E, Christensen KM, Walls AB, Eickhoff MK, von Scholten BJ, Frimodt-Møller M, Hansen TW, Persson F, Rossing P, Rotbain Curovic V, Houlind MB. Performance of new and panel CKD-EPI equations in European adults with type 2 diabetes. Diabetes Obes Metab 2024; 26:2501-2504. [PMID: 38449081 DOI: 10.1111/dom.15536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/08/2024]
Affiliation(s)
- Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | | | - Anne Byriel Walls
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Bernt Johan von Scholten
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Novo Nordisk A/S, Data Science, Copenhagen, Denmark
- Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | | | - Tine Willum Hansen
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Zoccali C, Mallamaci F. The evolving scenario of estimated glomerular filtration rate in clinical practice: the European kidney function consortium formulas. J Nephrol 2024; 37:1193-1195. [PMID: 38771518 DOI: 10.1007/s40620-024-01957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/11/2024] [Indexed: 05/22/2024]
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, NY, USA.
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy.
- Associazione Ipertensione Nefrologia Trapianto Renal (IPNET), C/O Nefrologia, Grande Ospedale Metropolitano, 8914, Reggio Calabria, Italy.
| | - Francesca Mallamaci
- Nephrology, Dialysis and Transplantation Unit Azienda Ospedaliera, Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
- Institute of Clinical Physiology, Research Unit of Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, CNR-IFC, Reggio Calabria, Italy
| |
Collapse
|
33
|
Ma Y, Yong Z, Wei L, Yuan H, Wan L, Pei X, Zhang F, Wen G, Jin C, Gu Y, Zhang Q, Zhao W, Zhu B. Data mining of reference intervals for serum creatinine: an improvement in glomerular filtration rate estimating equations based on Q-values. Clin Chem Lab Med 2024; 62:1138-1148. [PMID: 38205974 DOI: 10.1515/cclm-2023-1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
OBJECTIVES Glomerular filtration rate (GFR) estimating equations based on rescaled serum creatinine (SCr/Q) have shown better performance, where Q represents the median SCr for age- and sex-specific healthy populations. However, there remains a scarcity of investigations in China to determine this value. We aimed to develop Chinese age- and sex-specific reference intervals (RIs) and Q-values for SCr and to validate the equations incorporating new Q-values. METHODS We included 117,345 adults from five centers for establishing RIs and Q-values, and 3,692 participants with reference GFR (rGFR, 99mTc-DTPA renal dynamic imaging measurement) for validation. Appropriate age partitioning was determined using the decision tree method. Lower and upper reference limits and medians were calculated using the refineR algorithm, and Q-values were determined accordingly. We evaluated the full age spectrum (FAS) and European Kidney Function Consortium (EKFC) equations incorporating different Q-values considering bias, precision (interquartile range, IQR), and accuracy (percentage of estimates within ±20 % [P20] and ±30 % [P30] of rGFR). RESULTS RIs for males were: 18-79 years, 55.53-92.50 μmol/L; ≥80 years, 54.41-96.43 μmol/L. RIs for females were: 18-59 years, 40.42-69.73 μmol/L; 60-79 years, 41.16-73.69 μmol/L; ≥80 years, 46.50-73.20 μmol/L. Q-values were set at 73.82 μmol/L (0.84 mg/dL) for males and 53.80 μmol/L (0.61 mg/dL) for females. After validation, we found that the adjusted equations exhibit less bias, improved precision and accuracy, and increased agreement of GFR categories. CONCLUSIONS We determined Chinese age- and sex-specific RIs and Q-values for SCr. The adjustable Q-values provide an effective alternative to obtain valid equations for estimating GFR.
Collapse
Affiliation(s)
- Yao Ma
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Zhenzhu Yong
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Lu Wei
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Haichuan Yuan
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Lihong Wan
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Xiaohua Pei
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Feng Zhang
- Department of Nephrology, Sheyang People's Hospital, Yancheng, China
| | - Guohua Wen
- The First People's Hospital of Yancheng, Yancheng, P.R. China
| | - Cheng Jin
- Department of Geriatrics, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, P.R. China
| | - Yan Gu
- Department of Geriatrics, The First People's Hospital of Nantong, Nantong, P.R. China
| | - Qun Zhang
- Health Management Center, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Weihong Zhao
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| | - Bei Zhu
- Division of Nephrology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, P.R. China
| |
Collapse
|
34
|
Zhang L, Nizhamuding X, Zheng H, Zeng J, Yuan X, Ma Z, Zhou W, Zhang C, Zhang T, Zhang C. An LC-MS/MS method for serum cystatin C quantification and its comparison with two commercial immunoassays. Clin Chem Lab Med 2024; 62:1092-1100. [PMID: 38253403 DOI: 10.1515/cclm-2023-0821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVES The standardization of cystatin C (CysC) measurement has received increasing attention in recent years due to its importance in estimating glomerular filtration rate (GFR). Mass spectrometry-based assays have the potential to provide an accuracy base for CysC measurement. However, a precise, accurate and sustainable LC-MS/MS method for CysC is still lacking. METHODS The developed LC-MS/MS method quantified CysC by detecting signature peptide (T3) obtained from tryptic digestion. Stable isotope labeled T3 peptide (SIL-T3) was spiked to control matrix effects and errors caused by liquid handling. The protein denaturation, reduction and alkylation procedures were combined into a single step with incubation time of 1 h, and the digestion lasted for 3.5 h. In the method validation, digestion time-course, imprecision, accuracy, matrix effect, interference, limit of quantification (LOQ), carryover, linearity, and the comparability to two routine immunoassays were evaluated. RESULTS No significant matrix effect or interference was observed with the CysC measurement. The LOQ was 0.21 mg/L; the within-run and total imprecision were 1.33-2.05 % and 2.18-3.90 % for three serum pools (1.18-5.34 mg/L). The LC-MS/MS method was calibrated by ERM-DA471/IFCC and showed good correlation with two immunoassays traceable to ERM-DA471/IFCC. However, significant bias was observed for immunoassays against the LC-MS/MS method. CONCLUSIONS The developed LC-MS/MS method is robust and simpler and holds the promise to provide an accuracy base for routine immunoassays, which will promote the standardization of CysC measurement.
Collapse
Affiliation(s)
- Li Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Xiaerbanu Nizhamuding
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Hao Zheng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Jie Zeng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Xinyi Yuan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Zijia Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Weiyan Zhou
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Tianjiao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Chuanbao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| |
Collapse
|
35
|
Chang LH, Chang TT, Chu CH, Huang CC, Lin LY. Soluble tumor necrosis factor receptor type 1 is an alternative marker of urinary albumin-creatinine ratio and estimated glomerular filtration rate for predicting the decline of renal function in subjects with type 2 diabetes mellitus. Clin Chim Acta 2024; 558:117880. [PMID: 38555050 DOI: 10.1016/j.cca.2024.117880] [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: 01/03/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Urinary albumin-creatinine ratio (UACR) and estimated glomerular filtration rates (eGFR) help predict worsening diabetic kidney disease (DKD) but have their limitations. Soluble tumor necrosis factor receptor type 1 (sTNFR1) is a biomarker of DKD. The predictive abilities of sTNFR1 and UACR plus eGFR have not been compared. METHODS This prospective cohort study included patients with type 2 diabetes (T2D) to identify the risk factors of worsening DKD. Renal events were defined as > 30 % loss in eGFR based on consecutive tests after 6 months. The associations of sTNFR1, UACR, and eGFR levels and the risks of renal events were tested using a Cox regression model and the area under the curve (AUC) was compared between sTNFR1 levels and UACR plus eGFR using receiver-operating characteristic (ROC) analysis. The accuracy of stratification was evaluated using Kaplan-Meier analysis. RESULTS Levels of sTNFR1 and UACR were associated with risks of > 30 % decline in eGFR after adjusting for relevant factors. The association between sTNFR1 levels and renal outcomes was independent of UACR and eGFR at baseline. The AUC of sTNFR1 level was comparable with that of combined UACR and eGFR (0.73 vs. 0.71, respectively, p = 0.72) and the results persisted for quartile groups of sTNFR1 and risk categories of Kidney Disease: Improving Global Outcomes (KDIGO) (0.70 vs. 0.71, respectively, p = 0.84). Both stratifications by sTNFR1 levels and KDIGO were accurate. CONCLUSION sTNFR1 could be an alternative marker for identifying patients with diabetes at risk of declining renal function.
Collapse
Affiliation(s)
- Li-Hsin Chang
- Division of Endocrinology and Metabolism, Department of Medicine, Yeezen General Hospital, Taoyuan, Taiwan
| | - Ting-Ting Chang
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Huei Chu
- Department of Otorhinolaryngology-Head and Neck Surgery, Mackay Memorial Hospital, Taipei, Taiwan; Department of Audiology and Speech Language Pathology, Mackay Medical College, New Taipei City, Taiwan
| | - Chin-Chou Huang
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Liang-Yu Lin
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
36
|
Lees JS, Fabian J, Shlipak MG. Cystatin C should be routinely available for estimating kidney function. Curr Opin Nephrol Hypertens 2024; 33:337-343. [PMID: 38411195 DOI: 10.1097/mnh.0000000000000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW In this report, we summarize why the availability of cystatin C is important across a variety of clinical scenarios, the recent literature on when, why and in whom cystatin C testing should be considered, and how nephrologists can take practical steps to incorporate cystatin C testing into their practice. RECENT FINDINGS Large intra-individual discrepancies between estimated glomerular filtration rate by creatinine (eGFRcr) and estimated glomerular filtration rate by creatinine eGFRcys (known as eGFRdiff) are observed in at least 1 in 4 people. These differences are seen more commonly among more vulnerable individuals: older adults, females, non-White individuals and those living with multiple medical conditions. A large eGFRdiff, where eGFRcys is lower than eGFRcr, is associated with a plethora of adverse outcomes, including medication-associated adverse events, acute kidney injury, cardiovascular disease, kidney failure and all-cause mortality. Among studies that have measured GFR, eGFRcr-cys usually provides the most accurate estimation of kidney function compared to mGFR, including among participants with large discrepancies between eGFRcr and eGFRcys. SUMMARY Cystatin C improves sensitivity and specificity of chronic kidney disease diagnosis, improves detection of harmful acute and chronic changes in kidney function, improves precision of treatment eligibility and safety, and may reduce healthcare inequalities. Better education, curiosity, and motivation among nephrologists could substantially improve the availability and utilization of cystatin C.
Collapse
Affiliation(s)
- Jennifer S Lees
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - June Fabian
- Wits Donald Gordon Medical Centre, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, University of California San Francisco and San Francisco VA Healthcare System, San Francisco, California, USA
| |
Collapse
|
37
|
Jaimes Campos MA, Mavrogeorgis E, Latosinska A, Eder S, Buchwinkler L, Mischak H, Siwy J, Rossing P, Mayer G, Jankowski J. Urinary peptide analysis to predict the response to blood pressure medication. Nephrol Dial Transplant 2024; 39:873-883. [PMID: 37930730 PMCID: PMC11181870 DOI: 10.1093/ndt/gfad223] [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: 06/17/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The risk of diabetic kidney disease (DKD) progression is significant despite treatment with renin-angiotensin system (RAS) blocking agents. Current clinical tools cannot predict whether or not patients will respond to treatment with RAS inhibitors (RASi). We aimed to investigate whether proteome analysis could identify urinary peptides as biomarkers that could predict the response to angiotensin-converting enzyme inhibitor and angiotensin-receptor blockers treatment to avoid DKD progression. Furthermore, we investigated the comparability of the estimated glomerular filtration rate (eGFR), calculated using four different GFR equations, for DKD progression. METHODS We evaluated urine samples from a discovery cohort of 199 diabetic patients treated with RASi. DKD progression was defined based on eGFR percentage slope results between visits (∼1 year) and for the entire period (∼3 years) based on the eGFR values of each GFR equation. Urine samples were analysed using capillary electrophoresis-coupled mass spectrometry. Statistical analysis was performed between the uncontrolled (patients who did not respond to RASi treatment) and controlled kidney function groups (patients who responded to the RASi treatment). Peptides were combined in a support vector machine-based model. The area under the receiver operating characteristic curve was used to evaluate the risk prediction models in two independent validation cohorts treated with RASi. RESULTS The classification of patients into uncontrolled and controlled kidney function varies depending on the GFR equation used, despite the same sample set. We identified 227 peptides showing nominal significant difference and consistent fold changes between uncontrolled and controlled patients in at least three methods of eGFR calculation. These included fragments of collagens, alpha-1-antitrypsin, antithrombin-III, CD99 antigen and uromodulin. A model based on 189 of 227 peptides (DKDp189) showed a significant prediction of non-response to the treatment/DKD progression in two independent cohorts. CONCLUSIONS The DKDp189 model demonstrates potential as a predictive tool for guiding treatment with RASi in diabetic patients.
Collapse
Affiliation(s)
- Mayra Alejandra Jaimes Campos
- Mosaiques Diagnostics GmbH, Hannover, Germany
- University Hospital RWTH Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | - Emmanouil Mavrogeorgis
- Mosaiques Diagnostics GmbH, Hannover, Germany
- University Hospital RWTH Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | | | - Susanne Eder
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Lukas Buchwinkler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Complications Research, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Joachim Jankowski
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
38
|
Provenzano M, Hu L, Abenavoli C, Cianciolo G, Coppolino G, De Nicola L, La Manna G, Comai G, Baraldi O. Estimated glomerular filtration rate in observational and interventional studies in chronic kidney disease. J Nephrol 2024; 37:573-586. [PMID: 38347343 PMCID: PMC11150208 DOI: 10.1007/s40620-024-01887-x] [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: 03/20/2023] [Accepted: 05/08/2023] [Indexed: 06/05/2024]
Abstract
Estimated glomerular filtration rate is considered the principal measure of kidney function and, together with albuminuria, is a relevant prognostic factor for the development of end-stage kidney disease. Due to the strong association between estimated glomerular filtration rate and clinical events, such as commencement of dialysis, cardiovascular outcomes and all-cause death, estimated glomerular filtration rate is crucial for clinical decision-making in terms of scheduling follow-up and pharmacological interventions, and planning renal replacement therapies in advanced chronic kidney disease. In this review we discuss the available methods for measuring glomerular filtration rate and for estimating it through mathematical equations developed over the last few decades. We summarize the prognostic association of different percentages of estimated glomerular filtration rate decline and the main clinical outcomes, and how treatments modify estimated glomerular filtration rate decline and the risk of future endpoints. We also examine the role of pre-clinical trial slope and that of estimated glomerular filtration rate as a useful biomarker when evaluating patients for inclusion into both observational and interventional studies.
Collapse
Affiliation(s)
- Michele Provenzano
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Lilio Hu
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Chiara Abenavoli
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
| | - Giuseppe Coppolino
- Renal Unit, Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Luca De Nicola
- Renal Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Giorgia Comai
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy.
| | - Olga Baraldi
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico Sant'Orsola, Bologna, Italy
| |
Collapse
|
39
|
Hundemer GL, Sood MM, Akbari A. Beyond Creatinine: Is Cystatin C the New Global Standard for Estimated Glomerular Filtration Rate Evaluation? Kidney Med 2024; 6:100806. [PMID: 38495598 PMCID: PMC10943046 DOI: 10.1016/j.xkme.2024.100806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Affiliation(s)
- Gregory L. Hundemer
- Department of Medicine, Division of Nephrology, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Manish M. Sood
- Department of Medicine, Division of Nephrology, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ayub Akbari
- Department of Medicine, Division of Nephrology, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
40
|
Cho JM, Koh JH, Kim SG, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK, Park S. Associations of MRI-derived kidney volume, kidney function, body composition and physical performance in ≈38 000 UK Biobank participants: a population-based observational study. Clin Kidney J 2024; 17:sfae068. [PMID: 38660121 PMCID: PMC11040514 DOI: 10.1093/ckj/sfae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 04/26/2024] Open
Abstract
Background Kidney volume is used as a predictive and therapeutic marker for several clinical conditions. However, there is a lack of large-scale studies examining the relationship between kidney volume and various clinicodemographic factors, including kidney function, body composition and physical performance. Methods In this observational study, MRI-derived kidney volume measurements from 38 526 UK Biobank participants were analysed. Major kidney volume-related measures included body surface area (BSA)-adjusted total kidney volume (TKV) and the difference in bilateral kidneys. Multivariable-adjusted linear regression and cubic spline analyses were used to explore the association between kidney volume-related measures and clinicodemographic factors. Cox or logistic regression was used to identify the risks of death, non-kidney cancer, myocardial infarction, ischaemic stroke and chronic kidney disease (CKD). Results The median of BSA-adjusted TKV and the difference in kidney volume were 141.9 ml/m2 [interquartile range (IQR) 128.1-156.9] and 1.08-fold (IQR 1.04-1.15), respectively. Higher BSA-adjusted TKV was significantly associated with higher estimated glomerular filtration rate {eGFR; β = 0.43 [95% confidence interval (CI) 0.42-0.44]; P < .001}, greater muscle volume [β = 0.50 (95% CI 0.48-0.51); P < .001] and greater mean handgrip strength [β = 0.15 (95% CI 0.13-0.16); P < .001] but lower visceral adipose tissue volume [VAT; β = -0.09 (95% CI -0.11 to -0.07); P < .001] in adjusted models. A greater difference in bilateral kidney volumes was associated with lower eGFR, muscle volume and physical performance but with higher proteinuria and VAT. Higher BSA-adjusted TKV was significantly associated with a reduced risk of CKD [odds ratio (OR) 0.7 (95% CI 0.63-0.77); P < .001], while a greater difference in kidney volume was significantly associated with an increased risk of CKD [OR 1.13 (95% CI 1.07-1.20); P < .001]. Conclusion Higher BSA-adjusted TKV and lower differences in bilateral kidney volumes are associated with higher kidney function, muscle volume and physical performance and a reduced risk of CKD.
Collapse
Affiliation(s)
- Jeong Min Cho
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Korea
| | - Jung Hun Koh
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seong Geun Kim
- Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Soojin Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Uijeongbu, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Semin Cho
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Sehoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
41
|
Stevens PE, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancıoğlu R, Lamb E, Lin P, Madero M, McIntyre N, Morrow K, Roberts G, Sabanayagam D, Schaeffner E, Shlipak M, Shroff R, Tangri N, Thanachayanont T, Ulasi I, Wong G, Yang CW, Zhang L, Levin A. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int 2024; 105:S117-S314. [PMID: 38490803 DOI: 10.1016/j.kint.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 03/17/2024]
|
42
|
Pottel H, Nyman U, Björk J, Berg U, Bökenkamp A, Dubourg LD, Lemoine S, Goffin K, Grubb A, Hansson M, Larsson A, Littmann K, Åsling-Monemi K, Adeli K, Cavalier E, Delanaye P. Extending the cystatin C based EKFC-equation to children - validation results from Europe. Pediatr Nephrol 2024; 39:1177-1183. [PMID: 37875730 DOI: 10.1007/s00467-023-06192-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND A new cystatin C based European Kidney Function Consortium (EKFCCysC) equation was recently developed for adults, using the same mathematical form as the previously published full age spectrum creatinine based EKFC-equation (EKFCCrea). In the present study the cystatin C based EKFC-equation is extended to children, by defining the appropriate cystatin C rescaling factor QCysC. METHODS Rescaling factor QCysC for cystatin C was defined as: a) 0.83 mg/L, exactly as it was defined for young adults in the adult equation, and b) a more complex QCysC-age relationship based on 4th degree cystatin C-age polynomials after evaluation of data from Uppsala, Stockholm and Canada and aggregated data from Germany. The EKFCCysC equation was then validated in an independent dataset in European children (n = 2,293) with measured GFR, creatinine, cystatin C, age, height and sex available. RESULTS The EKFCCysC with the simple QCysC-value of 0.83 had a bias of -7.6 [95%CI -8.4;-6.5] mL/min/1.73 m2 and a P30-value of 85.8% [95%CI 84.4;87.3] equal to the EKFCCysC with the more complex 4th degree QCysC-value. The arithmetic mean of the EKFCCrea and EKFCCysC with the simple QCysC of 0.83 had a bias of -4.0 [95%CI -4.5;-3.1] mL/min/1.73 m2 and P30 of 90.4% [95%CI 89.2;91.6] similar to using the more complex 4th degree QCysC-polynomial. CONCLUSION Using exactly the same QCysC of 0.83 mg/L, the adult EKFCCysC can easily be extended to children, with some bias but acceptable P30-values. The arithmetic mean of EKFCCrea and EKFCCysC results in bias closer to zero and P30 slightly over 90%.
Collapse
Affiliation(s)
- Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium.
| | - Ulf Nyman
- Department of Translational Medicine, Division of Medical Radiology, Lund University, Malmö, Sweden
| | - Jonas Björk
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Ulla Berg
- Department of Clinical Science, Intervention and Technology, Division of Paediatrics, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Arend Bökenkamp
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Laurence Derain Dubourg
- Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Groupement Hospitalier Edouard Herriot, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, and Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, Université Claude Bernard Lyon 1, Lyon, France
| | - Sandrine Lemoine
- Néphrologie, Dialyse, Hypertension et Exploration Fonctionnelle Rénale, Groupement Hospitalier Edouard Herriot, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, and Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, Université Claude Bernard Lyon 1, Lyon, France
| | - Karolien Goffin
- Department of Nuclear Medicine, Division of Nuclear Medicine and Molecular Imaging, University Hospital Leuven, KU Leuven, Louvain, Belgium
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund University, Lund, Sweden
| | - Magnus Hansson
- Function Area Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital Huddinge and Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Karin Littmann
- Department of Medicine Huddinge, and Medical Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska Institutet, Stockholm, Sweden, Karolinska University Hospital, Stockholm, Sweden
| | - Kajsa Åsling-Monemi
- Department of Clinical Science, Intervention and Technology, Division of Paediatrics, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Khosrow Adeli
- Clinical Biochemistry, Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hopital Universitaire Caremeau, Nîmes, France
| |
Collapse
|
43
|
Fu EL, Levey AS, Coresh J, Grams ME, Faucon AL, Elinder CG, Dekker FW, Delanaye P, Inker LA, Carrero JJ. Accuracy of GFR estimating equations based on creatinine, cystatin C or both in routine care. Nephrol Dial Transplant 2024; 39:694-706. [PMID: 37813817 DOI: 10.1093/ndt/gfad219] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND New equations to estimate glomerular filtration rate based on creatinine (eGFRcr), cystatin C (eGFRcys) or both (eGFRcr-cys) have been developed by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) and the European Kidney Function Consortium (EKFC). There is a need to evaluate the performance of these equations in diverse European settings to inform implementation decisions, especially among people with key comorbid conditions. METHODS We performed a cross-sectional study including 6174 adults referred for single-point plasma clearance of iohexol in Stockholm, Sweden, with 9579 concurrent measurements of creatinine and cystatin C. We assessed the performance of the CKD-EPI 2009/2012/2021, EKFC 2021/2023, revised Lund-Malmö (RLM) 2011 and Caucasian, Asian, Pediatric and Adult (CAPA) 2014 equations against measured GFR (mGFR). RESULTS Mean age was 56 years, median mGFR was 62 mL/min/1.73 m2 and 40% were female. Comorbid conditions were common: cardiovascular disease (30%), liver disease (28%), diabetes (26%) and cancer (26%). All eGFRcr-cys equations had small bias and P30 (the percentage of estimated values within 30% of mGFR) close to 90%, and performed better than eGFRcr or eGFRcys equations. Among eGFRcr equations, CKD-EPI 2009 and CKD-EPI 2021 showed larger bias and lower P30 than EKFC 2021 and RLM. There were no meaningful differences in performance across eGFRcys equations. Findings were consistent across comorbid conditions, and eGFRcr-cys equations showed good performance in patients with liver disease, cancer and heart failure. CONCLUSIONS In conclusion, eGFRcr-cys equations performed best, with minimal variation among equations in this Swedish cohort. The lower performance of CKD-EPI eGFRcr equations compared with EKFC and RLM may reflect differences in population characteristics and mGFR methods. Implementing eGFRcr equations will require a trade-off between accuracy and uniformity across regions.
Collapse
Affiliation(s)
- Edouard L Fu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew S Levey
- Division of Nephrology, Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Morgan E Grams
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Anne-Laure Faucon
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- INSERM UMR 1018, Department of Clinical Epidemiology, Paris-Saclay University, Paris, France
| | - Carl-Gustaf Elinder
- Division of Renal Medicine, Department of Clinical Intervention, and Technology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| | - Lesley A Inker
- Division of Nephrology, Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Fino NF, Adingwupu OM, Coresh J, Greene T, Haaland B, Shlipak MG, Costa E Silva VT, Kalil R, Mindikoglu AL, Furth SL, Seegmiller JC, Levey AS, Inker LA. Evaluation of novel candidate filtration markers from a global metabolomic discovery for glomerular filtration rate estimation. Kidney Int 2024; 105:582-592. [PMID: 38006943 PMCID: PMC10932836 DOI: 10.1016/j.kint.2023.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/27/2023]
Abstract
Creatinine and cystatin-C are recommended for estimating glomerular filtration rate (eGFR) but accuracy is suboptimal. Here, using untargeted metabolomics data, we sought to identify candidate filtration markers for a new targeted assay using a novel approach based on their maximal joint association with measured GFR (mGFR) and with flexibility to consider their biological properties. We analyzed metabolites measured in seven diverse studies encompasing 2,851 participants on the Metabolon H4 platform that had Pearson correlations with log mGFR and used a stepwise approach to develop models to < -0.5 estimate mGFR with and without inclusion of creatinine that enabled selection of candidate markers. In total, 456 identified metabolites were present in all studies, and 36 had correlations with mGFR < -0.5. A total of 2,225 models were developed that included these metabolites; all with lower root mean square errors and smaller coefficients for demographic variables compared to estimates using untargeted creatinine. Seventeen metabolites were chosen, including 12 new candidate filtration markers. The selected metabolites had strong associations with mGFR and little dependence on demographic factors. Candidate metabolites were identified with maximal joint association with mGFR and minimal dependence on demographic variables across many varied clinical settings. These metabolites are excreted in urine and represent diverse metabolic pathways and tubular handling. Thus, our data can be used to select metabolites for a multi-analyte eGFR determination assay using mass spectrometry that potentially offers better accuracy and is less prone to non-GFR determinants than the current eGFR biomarkers.
Collapse
Affiliation(s)
- Nora F Fino
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Ogechi M Adingwupu
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Josef Coresh
- Department of Population Health, NYU Langone, New York, New York, USA
| | - Tom Greene
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Ben Haaland
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Michael G Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affair Medical Center and University of California, San Francisco, San Francisco, California, USA
| | - Veronica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica 16, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Kalil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ayse L Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA; Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L Furth
- Department of Pediatrics, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jesse C Seegmiller
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
45
|
Li CR, Deng YL, Miao Y, Zhang M, Zeng JY, Liu XY, Wu Y, Li YJ, Liu AX, Zhu JQ, Liu C, Zeng Q. Exposures to drinking water disinfection byproducts and kidney function in Chinese women. ENVIRONMENTAL RESEARCH 2024; 244:117925. [PMID: 38103773 DOI: 10.1016/j.envres.2023.117925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Disinfection byproducts (DBPs), the ubiquitous contaminants in drinking water, have been shown to impair renal function in experimental studies. However, epidemiological evidence is sparse. OBJECTIVE To investigate exposures to DBPs in associations with renal function among women. METHODS A total of 920 women from December 2018 to January 2020 were abstracted from the Tongji Reproductive and Environmental (TREE) Study, an ongoing cohort study in Wuhan, China. Urine samples were gathered at baseline recruitment and analyzed for dichloroacetic acid (DCAA) and trichloroacetic acid (TCAA) as biomarkers of DBP exposures. Serum uric acid (UA), creatinine, and estimated glomerular filtration rate (eGFR) were measured as indicators of renal function. Multivariate linear regression and restricted cubic spline (RCS) models were conducted to assess urinary DCAA and TCAA concentrations in associations with renal function indicators. Stratified analyses by age and body mass index (BMI) were also performed. RESULTS We found null evidence of urinary TCAA in associations with renal function indicators. However, elevated urinary DCAA tertiles were related to decreased eGFR (β = -1.78%, 95% CI: 3.21%, -0.36%, comparing the upper vs. lower tertile; P for trend = 0.01). This inverse association still existed when urinary DCAA concentration was treated as a continuous variable, and the dose-response relationship was linear based on the RCS model (P for overall association = 0.002 and P for non-linear associations = 0.44). In the stratified analyses, we found an association of urinary DCAA concentration with decreased UA level among women <30 years but an association with increased UA level among women ≥30 years (P for interaction = 0.04). CONCLUSION Urinary DCAA but not TCAA was associated with impaired renal function among women undergoing assisted reproductive technology.
Collapse
Affiliation(s)
- Cheng-Ru Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu Miao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Min Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jia-Yue Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiao-Ying Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yang Wu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yang-Juan Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - A-Xue Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jin-Qin Zhu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
46
|
Ebert N. [Novel equations for estimating renal function: significance for drug dose adjustment]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:280-285. [PMID: 38252158 DOI: 10.1007/s00108-023-01649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Important therapeutic decisions depend on kidney function, which is why its correct assessment is of great importance. It also plays an important role for drug dose adjustments in patients with impaired kidney function. OBJECTIVES In clinical practice, kidney function is almost always estimated using mathematical glomerular filtration rate (GFR) equations. To estimate GFR, the patient's age and gender as well as kidney-specific endogenous biomarkers are required. This work aims to provide an overview of the advantages and disadvantages of the biomarkers serum creatinine and cystatin C in assessing kidney function. Particularly in patients with significantly reduced or increased muscle mass, creatinine is not suitable for determining GFR, and cystatin C should be used. Currently recommended GFR estimating equations are described, illustrating for which patient groups they can be used. CURRENT DATA A large number of high-ranking publications are available investigating the validity of GFR estimating equations and the optimal choice of endogenous biomarkers. However, there are still large gaps when it comes to drug approval studies in older patients and children. CONCLUSION Estimated GFR (eGFR) is only a rough estimate of kidney function and should not be interpreted as an exact number. Drug dose adjustments may be necessary in patients with an eGFR of < 50 ml/min and should be verified particularly in severely impaired GFR (< 30 ml/min). There are tools available online for this purpose.
Collapse
Affiliation(s)
- Natalie Ebert
- Institut für Public Health, Charité - Universitätsmedizin Berlin, 10117, Berlin, Deutschland.
| |
Collapse
|
47
|
Fu EL, Carrero JJ, Sang Y, Evans M, Ishigami J, Inker LA, Grams ME, Levey AS, Coresh J, Ballew SH. Association of Low Glomerular Filtration Rate With Adverse Outcomes at Older Age in a Large Population With Routinely Measured Cystatin C. Ann Intern Med 2024; 177:269-279. [PMID: 38285982 PMCID: PMC11079939 DOI: 10.7326/m23-1138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND The commonly accepted threshold of glomerular filtration rate (GFR) to define chronic kidney disease (CKD) is less than 60 mL/min/1.73 m2. This threshold is based partly on associations between estimated GFR (eGFR) and the frequency of adverse outcomes. The association is weaker in older adults, which has created disagreement about the appropriateness of the threshold for these persons. In addition, the studies measuring these associations included relatively few outcomes and estimated GFR on the basis of creatinine level (eGFRcr), which may be less accurate in older adults. OBJECTIVE To evaluate associations in older adults between eGFRcr versus eGFR based on creatinine and cystatin C levels (eGFRcr-cys) and 8 outcomes. DESIGN Population-based cohort study. SETTING Stockholm, Sweden, 2010 to 2019. PARTICIPANTS 82 154 participants aged 65 years or older with outpatient creatinine and cystatin C testing. MEASUREMENTS Hazard ratios for all-cause mortality, cardiovascular mortality, and kidney failure with replacement therapy (KFRT); incidence rate ratios for recurrent hospitalizations, infection, myocardial infarction or stroke, heart failure, and acute kidney injury. RESULTS The associations between eGFRcr-cys and outcomes were monotonic, but most associations for eGFRcr were U-shaped. In addition, eGFRcr-cys was more strongly associated with outcomes than eGFRcr. For example, the adjusted hazard ratios for 60 versus 80 mL/min/1.73 m2 for all-cause mortality were 1.2 (95% CI, 1.1 to 1.3) for eGFRcr-cys and 1.0 (CI, 0.9 to 1.0) for eGFRcr, and for KFRT they were 2.6 (CI, 1.2 to 5.8) and 1.4 (CI, 0.7 to 2.8), respectively. Similar findings were observed in subgroups, including those with a urinary albumin-creatinine ratio below 30 mg/g. LIMITATION No GFR measurements. CONCLUSION Compared with low eGFRcr in older patients, low eGFRcr-cys was more strongly associated with adverse outcomes and the associations were more uniform. PRIMARY FUNDING SOURCE Swedish Research Council, National Institutes of Health, and Dutch Kidney Foundation.
Collapse
Affiliation(s)
- Edouard L. Fu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, and Division of Nephrology, Department of Clinical Sciences, Karolinska Institute, Danderyd Hospital, Stockholm, Sweden
| | - Yingying Sang
- Optimal Aging Institute and Division of Epidemiology, Department of Population Health, New York University Grossman School of Medicine, New York, New York
| | - Marie Evans
- Department of Clinical Intervention and Technology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Junichi Ishigami
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Lesley A. Inker
- Division of Nephrology, Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Morgan E. Grams
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Andrew S. Levey
- Division of Nephrology, Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Josef Coresh
- Optimal Aging Institute and Division of Epidemiology, Department of Population Health, New York University Grossman School of Medicine, New York, New York
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Shoshana H. Ballew
- Optimal Aging Institute and Division of Epidemiology, Department of Population Health, New York University Grossman School of Medicine, New York, New York
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
48
|
Salamin M, Segerer S, Buchkremer F. Estimating glomerular filtration rate with cystatin C: a systematic comparison of the new EKFC and the CKD-EPI equation. Clin Kidney J 2024; 17:sfad243. [PMID: 38468699 PMCID: PMC10926324 DOI: 10.1093/ckj/sfad243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Indexed: 03/13/2024] Open
Affiliation(s)
- Melanie Salamin
- Division of Nephrology, Kantonsspital Aarau, Aarau, Switzerland
| | - Stephan Segerer
- Division of Nephrology, Kantonsspital Aarau, Aarau, Switzerland
| | | |
Collapse
|
49
|
Pottel H, Delanaye P, Cavalier E. Exploring Renal Function Assessment: Creatinine, Cystatin C, and Estimated Glomerular Filtration Rate Focused on the European Kidney Function Consortium Equation. Ann Lab Med 2024; 44:135-143. [PMID: 37909162 PMCID: PMC10628758 DOI: 10.3343/alm.2023.0237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/25/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023] Open
Abstract
Serum creatinine and serum cystatin C are the most widely used renal biomarkers for calculating the estimated glomerular filtration rate (eGFR), which is used to estimate the severity of kidney damage. In this review, we present the basic characteristics of these biomarkers, their advantages and disadvantages, some basic history, and current laboratory measurement practices with state-of-the-art methodology. Their clinical utility is described in terms of normal reference intervals, graphically presented with age-dependent reference intervals, and their use in eGFR equations.
Collapse
Affiliation(s)
- Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
50
|
Delanaye P, Rule AD, Schaeffner E, Cavalier E, Shi J, Hoofnagle AN, Nyman U, Björk J, Pottel H. Performance of the European Kidney Function Consortium (EKFC) creatinine-based equation in United States cohorts. Kidney Int 2024; 105:629-637. [PMID: 38101514 DOI: 10.1016/j.kint.2023.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/28/2023] [Accepted: 11/05/2023] [Indexed: 12/17/2023]
Abstract
Estimating glomerular filtration rate (GFR) is important in daily practice to assess kidney function and adapting the best clinical care of patients with and without chronic kidney disease. The new creatinine-based European Kidney Function Consortium (EKFC) equation is used to estimate GFR. This equation was developed and validated mainly in European individuals and based on a rescaled creatinine, with the rescaling factor (Q-value) defined as the median normal value of serum creatinine in a given population. The validation was limited in Non-Black Americans and absent in Black Americans. Here, our cross-sectional analysis included 12,854 participants from nine studies encompassing large numbers of both non-Black and Black Americans with measured GFR by clearance of an exogenous marker (reference method), serum creatinine, age, sex, and self-reported race available. Two strategies were considered with population-specific Q-values in Black and non-Black men and women (EKFCPS) or a race-free Q-value (EKFCRF). In the whole population, only the EKFCPS equation showed no statistical median bias (0.14, 95% confidence interval [-0.07; 0.35] mL/min/1.73m2), and the bias for the EKFCRF (0.74, [0.51; 0.94] mL/min/1.73m2) was closer to zero than that for the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI2021) equation (1.22, [0.99; 1.47]) mL/min/1.73m2]. The percentage of estimated GFR within 30% of measured GFR was similar for CKD-EPI2021 (79.2% [78.5%; 79.9%]) and EKFCRF (80.1% [79.4%; 80.7%]), but improved for the EKFCPS equation (81.1% [80.5%; 81.8%]). Thus, our EKFC equations can be used to estimate GFR in the United States incorporating either self-reported race or unknown race at the patient's discretion per hospital registration records.
Collapse
Affiliation(s)
- Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium; Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France.
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elke Schaeffner
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Junyan Shi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA; Kidney Research Institute, Department of Medicine, University of Washington, Seattle, Washington, USA; Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ulf Nyman
- Department of Translational Medicine, Division of Medical Radiology, Lund University, Malmö, Sweden
| | - Jonas Björk
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden; Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| |
Collapse
|