1
|
Cecrdlova E, Krupickova L, Fialova M, Novotny M, Tichanek F, Svachova V, Mezerova K, Viklicky O, Striz I. Insights into IL-1 family cytokines in kidney allograft transplantation: IL-18BP and free IL-18 as emerging biomarkers. Cytokine 2024; 180:156660. [PMID: 38801805 DOI: 10.1016/j.cyto.2024.156660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/15/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Proinflammatory cytokines and their inhibitors are involved in the regulation of multiple immune reactions including response to transplanted organs. In this prospective study, we evaluated changes in serum concentrations of six IL-1 family cytokines (IL-1 alpha, IL-1 beta, IL-1RA, IL-18, IL-18BP, and IL-36 beta) in 138 kidney allograft recipients and 48 healthy donors. Samples were collected before transplantation and then after one week, three months and one year, additional sera were obtained at the day of biopsy positive for acute rejection. We have shown, that concentrations of proinflammatory members of the IL-1 family (IL-1β, IL-18, IL-36 β) and anti-inflammatory IL-18BP decreased immediately after the transplantation. The decline of serum IL-1RA and IL-1α was not observed in subjects with acute rejection. IL-18, including specifically its free form, is the only cytokine which increase serum concentrations in the period between one week and three months in both groups of patients without upregulation of its inhibitor, IL-18BP. Serum concentrations of calculated free IL-18 were upregulated in the acute rejection group at the time of acute rejection. We conclude that IL-1 family cytokines are involved mainly in early phases of the response to kidney allograft. Serum concentrations of free IL-18 and IL-18BP represent possible biomarkers of acute rejection, and targeting IL-18 might be of therapeutic value.
Collapse
Affiliation(s)
- E Cecrdlova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - L Krupickova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - M Fialova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - M Novotny
- Institute for Clinical and Experimental Medicine, Transplant Center, Department of Nephrology, Prague, Czech Republic
| | - F Tichanek
- Institute for Clinical and Experimental Medicine, Department of Data Science, Prague, Czech Republic
| | - V Svachova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - K Mezerova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - O Viklicky
- Institute for Clinical and Experimental Medicine, Transplant Center, Department of Nephrology, Prague, Czech Republic
| | - I Striz
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic.
| |
Collapse
|
2
|
Swolinsky JS, Hinz RM, Markus CE, Singer E, Bachmann F, Halleck F, Kron S, Naik MG, Schmidt D, Obermeier M, Gebert P, Rauch G, Kropf S, Haase M, Budde K, Eckardt KU, Westhoff TH, Schmidt-Ott KM. Plasma NGAL levels in stable kidney transplant recipients and the risk of allograft loss. Nephrol Dial Transplant 2024; 39:483-495. [PMID: 37858309 PMCID: PMC11024820 DOI: 10.1093/ndt/gfad226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The objective of this study was to investigate the utility of neutrophil gelatinase-associated lipocalin (NGAL) and calprotectin (CPT) to predict long-term graft survival in stable kidney transplant recipients (KTR). METHODS A total of 709 stable outpatient KTR were enrolled >2 months post-transplant. The utility of plasma and urinary NGAL (pNGAL, uNGAL) and plasma and urinary CPT at enrollment to predict death-censored graft loss was evaluated during a 58-month follow-up. RESULTS Among biomarkers, pNGAL showed the best predictive ability for graft loss and was the only biomarker with an area under the curve (AUC) > 0.7 for graft loss within 5 years. Patients with graft loss within 5 years (n = 49) had a median pNGAL of 304 [interquartile range (IQR) 235-358] versus 182 (IQR 128-246) ng/mL with surviving grafts (P < .001). Time-dependent receiver operating characteristic analyses at 58 months indicated an AUC for pNGAL of 0.795, serum creatinine-based Chronic Kidney Disease Epidemiology Collaboration estimated glomerular filtration rate (eGFR) had an AUC of 0.866. pNGAL added to a model based on conventional risk factors for graft loss with death as competing risk (age, transplant age, presence of donor-specific antibodies, presence of proteinuria, history of delayed graft function) had a strong independent association with graft loss {subdistribution hazard ratio (sHR) for binary log-transformed pNGAL [log2(pNGAL)] 3.4, 95% confidence interval (CI) 2.24-5.15, P < .0001}. This association was substantially attenuated when eGFR was added to the model [sHR for log2(pNGAL) 1.63, 95% CI 0.92-2.88, P = .095]. Category-free net reclassification improvement of a risk model including log2(pNGAL) in addition to conventional risk factors and eGFR was 54.3% (95% CI 9.2%-99.3%) but C-statistic did not improve significantly. CONCLUSIONS pNGAL was an independent predictor of renal allograft loss in stable KTR from one transplant center but did not show consistent added value when compared with baseline predictors including the conventional marker eGFR. Future studies in larger cohorts are warranted.
Collapse
Affiliation(s)
- Jutta S Swolinsky
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ricarda M Hinz
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Carolin E Markus
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eugenia Singer
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Susanne Kron
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Marcel G Naik
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | | | - Pimrapat Gebert
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology
| | - Geraldine Rauch
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology
| | - Siegfried Kropf
- Institute of Biometry and Medical Informatics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Michael Haase
- Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
- Diaverum Renal Services, MVZ Potsdam, Potsdam, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Timm H Westhoff
- Medical Department I, Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, Bochum, Germany
| | - Kai M Schmidt-Ott
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
3
|
Lai M, Scherzer R, Shlipak MG, Madden E, Vittinghoff E, Tse W, Parikh CR, Villalobos CPC, Monroy-Trujillo JM, Moore RD, Estrella MM. Ambulatory urine biomarkers associations with acute kidney injury and hospitalization in people with HIV. AIDS 2023; 37:2339-2348. [PMID: 37650762 PMCID: PMC10843826 DOI: 10.1097/qad.0000000000003705] [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] [Indexed: 09/01/2023]
Abstract
BACKGROUND People with HIV (PWH) generally have worse ambulatory levels of kidney injury biomarkers and excess risk of acute kidney injury (AKI) compared to persons without HIV. We evaluated whether ambulatory measures of subclinical kidney injury among PWH are associated with subsequent AKI. METHODS In the Predictors of Acute Renal Injury Study (PARIS), which enrolled 468 PWH from April 2016 to August 2019, we measured 10 urine biomarkers of kidney health (albumin, a1m, b2M, NGAL, IL18, KIM-1, EGF, UMOD, MCP-1, YKL40) at baseline and annually during follow-up. Using multivariable Cox regression models, we evaluated baseline and time-updated biomarker associations with the primary outcome of AKI (≥0.3 mg/dl or ≥1.5-times increase in serum creatinine from baseline) and secondary outcome of all-cause hospitalization. RESULTS At baseline, the mean age was 53 years old, and 45% self-identified as female. In time-updated models adjusting for sociodemographic factors, comorbidities, albuminuria, estimated glomerular filtration rate, and HIV-associated factors, higher KIM-1 [hazard ratio (HR) = 1.30 per twofold higher; 95% confidence interval (CI) 1.03-1.63] and NGAL concentrations (HR = 1.24, 95% CI 1.06-1.44) were associated with higher risk of hospitalized AKI. Additionally, in multivariable, time-updated models, higher levels of KIM-1 (HR = 1.19, 95% CI 1.00, 1.41), NGAL (HR = 1.13, 95% CI 1.01-1.26), and MCP-1 (HR = 1.20, 95% CI 1.00, 1.45) were associated with higher risk of hospitalization. CONCLUSIONS Urine biomarkers of kidney tubular injury, such as KIM-1 and NGAL, are strongly associated with AKI among PWH, and may hold potential for risk stratification of future AKI.
Collapse
Affiliation(s)
- Mason Lai
- Kidney Health Research Collaborative, Department of Medicine
- Department of Medicine, University of California San Francisco
| | | | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine
- Department of Medicine, University of California San Francisco
- San Francisco VA Healthcare System
- Department of Epidemiology and Biostatistics
| | - Erin Madden
- Kidney Health Research Collaborative, Department of Medicine
- San Francisco VA Healthcare System
| | - Eric Vittinghoff
- Kidney Health Research Collaborative, Department of Medicine
- Department of Epidemiology and Biostatistics
| | - Warren Tse
- Kidney Health Research Collaborative, Department of Medicine
- San Francisco VA Healthcare System
| | - Chirag R Parikh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Richard D Moore
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine
- Department of Medicine, University of California San Francisco
- San Francisco VA Healthcare System
- Department of Medicine, Division of Nephrology, University of California San Francisco, San Francisco, California
| |
Collapse
|
4
|
Rybi Szumińska A, Wasilewska A, Kamianowska M. Protein Biomarkers in Chronic Kidney Disease in Children-What Do We Know So Far? J Clin Med 2023; 12:3934. [PMID: 37373629 DOI: 10.3390/jcm12123934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic kidney disease (CKD) in children is a major concern of medical care and public health as it is related to high morbidity and mortality due to progression to end-stage kidney disease (ESKD). It is essential to identify patients with a risk of developing CKD to implement therapeutic interventions. Unfortunately, conventional markers of CKD, such as serum creatinine, glomerular filtration rate (GFR) and proteinuria, have many limitations in serving as an early and specific diagnostic tool for this condition. Despite the above, they are still the most frequently utilized as we do not have better. Studies from the last decade identified multiple CKD blood and urine protein biomarkers but mostly assessed the adult population. This article outlines some recent achievements and new perspectives in finding a set of protein biomarkers that might improve our ability to prognose CKD progression in children, monitor the response to treatment, or even become a potential therapeutic target.
Collapse
Affiliation(s)
- Agnieszka Rybi Szumińska
- Department of Peadiatrics and Nephrology, Medical University of Bialystok, Waszyngtona 17, 15-297 Bialystok, Poland
| | - Anna Wasilewska
- Department of Peadiatrics and Nephrology, Medical University of Bialystok, Waszyngtona 17, 15-297 Bialystok, Poland
| | - Monika Kamianowska
- Department of Peadiatrics and Nephrology, Medical University of Bialystok, Waszyngtona 17, 15-297 Bialystok, Poland
| |
Collapse
|
5
|
Wlodek E, Kirkpatrick RB, Andrews S, Noble R, Schroyer R, Scott J, Watson CJE, Clatworthy M, Harrison EM, Wigmore SJ, Stevenson K, Kingsmore D, Sheerin NS, Bestard O, Stirnadel-Farrant HA, Abberley L, Busz M, DeWall S, Birchler M, Krull D, Thorneloe KS, Weber A, Devey L. A pilot study evaluating GSK1070806 inhibition of interleukin-18 in renal transplant delayed graft function. PLoS One 2021; 16:e0247972. [PMID: 33684160 PMCID: PMC7939287 DOI: 10.1371/journal.pone.0247972] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/11/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Delayed graft function (DGF) following renal transplantation is a manifestation of acute kidney injury (AKI) leading to poor long-term outcome. Current treatments have limited effectiveness in preventing DGF. Interleukin-18 (IL18), a biomarker of AKI, induces interferon-γ expression and immune activation. GSK1070806, an anti-IL18 monoclonal antibody, neutralizes activated (mature) IL18 released from damaged cells following inflammasome activation. This phase IIa, single-arm trial assessed the effect of a single dose of GSK1070806 on DGF occurrence post donation after circulatory death (DCD) kidney transplantation. METHODS The 3 mg/kg intravenous dose was selected based on prior studies and physiologically based pharmacokinetic (PBPK) modeling, indicating the high likelihood of a rapid and high level of IL18 target engagement when administered prior to kidney allograft reperfusion. Utilization of a Bayesian sequential design with a background standard-of-care DGF rate of 50% based on literature, and confirmed via extensive registry data analyses, enabled a statistical efficacy assessment with a minimal sample size. The primary endpoint was DGF frequency, defined as dialysis requirement ≤7 days post transplantation (except for hyperkalemia). Secondary endpoints included safety, pharmacokinetics and pharmacodynamic biomarkers. RESULTS GSK1070806 administration was associated with IL18-GSK1070806 complex detection and increased total serum IL18 levels due to IL18 half-life prolongation induced by GSK1070806 binding. Interferon-γ-induced chemokine levels declined or remained unchanged in most patients. Although the study was concluded prior to the Bayesian-defined stopping point, 4/7 enrolled patients (57%) had DGF, exceeding the 50% standard-of-care rate, and an additional two patients, although not reaching the protocol-defined DGF definition, demonstrated poor graft function. Six of seven patients experienced serious adverse events (SAEs), including two treatment-related SAEs. CONCLUSION Overall, using a Bayesian design and extensive PBPK dose modeling with only a small sample size, it was deemed unlikely that GSK1070806 would be efficacious in preventing DGF in the enrolled DCD transplant population. TRIAL REGISTRATION NCT02723786.
Collapse
Affiliation(s)
- E. Wlodek
- GlaxoSmithKline, Clinical Unit Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - R. B. Kirkpatrick
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - S. Andrews
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - R. Noble
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - R. Schroyer
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - J. Scott
- JMS Statistics Ltd, Pinner, United Kingdom
| | - C. J. E. Watson
- University of Cambridge and the NIHR Cambridge Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge, Cambridge, United Kingdom
| | - M. Clatworthy
- University of Cambridge and the NIHR Cambridge Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge, Cambridge, United Kingdom
| | | | - S. J. Wigmore
- Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - K. Stevenson
- Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - D. Kingsmore
- Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - N. S. Sheerin
- Newcastle Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Newcastle University, Newcastle, United Kingdom
| | - O. Bestard
- L’Hospitalet de Llobregat, Bellvitge University Hospital, Kidney Transplant Unit, Barcelona, Spain
| | | | - L. Abberley
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - M. Busz
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - S. DeWall
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - M. Birchler
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - D. Krull
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - K. S. Thorneloe
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - A. Weber
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - L. Devey
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
6
|
Nagarajah S, Xia S, Rasmussen M, Tepel M. Endogenous intronic antisense long non-coding RNA, MGAT3-AS1, and kidney transplantation. Sci Rep 2019; 9:14743. [PMID: 31611608 PMCID: PMC6791892 DOI: 10.1038/s41598-019-51409-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 09/28/2019] [Indexed: 11/09/2022] Open
Abstract
β-1,4-mannosylglycoprotein 4-β-N-acetylglucosaminyltransferase (MGAT3) is a key molecule for the innate immune system. We tested the hypothesis that intronic antisense long non-coding RNA, MGAT3-AS1, can predict delayed allograft function after kidney transplantation. We prospectively assessed kidney function and MGAT3-AS1 in 129 incident deceased donor kidney transplant recipients before and after transplantation. MGAT3-AS1 levels were measured in mononuclear cells using qRT-PCR. Delayed graft function was defined by at least one dialysis session within 7 days of transplantation. Delayed graft function occurred in 22 out of 129 transplant recipients (17%). Median MGAT3-AS1 after transplantation was significantly lower in patients with delayed graft function compared to patients with immediate graft function (6.5 × 10−6, IQR 3.0 × 10−6 to 8.4 × 10−6; vs. 8.3 × 10−6, IQR 5.0 × 10−6 to 12.8 × 10−6; p < 0.05). The median preoperative MGAT3-AS1 was significantly lower in kidney recipients with delayed graft function (5.1 × 10−6, IQR, 2.4 × 10−6 to 6.8 × 10−6) compared to recipients with immediate graft function (8.9 × 10−6, IQR, 6.8 × 10−6 to 13.4 × 10−6; p < 0.05). Receiver-operator characteristics showed that preoperative MGAT3-AS1 predicted delayed graft function (area under curve, 0.83; 95% CI, 0.65 to 1.00; p < 0.01). We observed a positive predictive value of 0.57, and a negative predictive value of 0.95. Long non-coding RNA, MGAT3-AS1, indicates short-term outcome in patients with deceased donor kidney transplantation.
Collapse
Affiliation(s)
- Subagini Nagarajah
- Odense University Hospital, Department of Nephrology, Odense, Denmark.,University of Southern Denmark, Institute of Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research, Odense, Denmark
| | - Shengqiang Xia
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | | | - Martin Tepel
- Odense University Hospital, Department of Nephrology, Odense, Denmark. .,University of Southern Denmark, Institute of Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research, Odense, Denmark.
| |
Collapse
|
7
|
Rangaswamy D, Sud K. Acute kidney injury and disease: Long-term consequences and management. Nephrology (Carlton) 2019; 23:969-980. [PMID: 29806146 DOI: 10.1111/nep.13408] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 01/31/2023]
Abstract
With increasing longevity and the presence of multiple comorbidities, a significant proportion of hospitalized patients, and an even larger population in the community, is at increased risk of developing an episode of acute kidney injury (AKI). Because of improvements in short-term outcomes following an episode of AKI, survivors of an episode of AKI are now predisposed to develop its long-term sequel. The identification of risk for progression to chronic kidney disease (CKD) is complicated by the absence of good biomarkers that identify this risk and the variability of risk associated with clinical factors including, but not limited to, the number of AKI episodes, severity, duration of previous AKI and pre-existing CKD that has made the prediction for long-term outcomes in survivors of AKI more difficult. Being a significant contributor to the growing incidence of CKD, there is a need to implement measures to prevent AKI in both the community and hospital settings, target interventions to treat AKI that are also associated with better long-term outcomes, accurately identify patients at risk of adverse consequences following an episode of AKI and institute therapeutic strategies to improve these long-term outcomes. We discuss the lasting renal and non-renal consequences following an episode of AKI, available biomarkers and non-invasive testing to identify ongoing intra-renal pathology and review the currently available and future treatment strategies to help reduce these adverse long-term outcomes.
Collapse
Affiliation(s)
- Dharshan Rangaswamy
- Department of Nephrology, Kasturba Medical College and Hospital, Manipal Academy of Higher Education, Karnataka, India.,Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| | - Kamal Sud
- Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia.,Department of Renal Medicine, Nepean Hospital, New South Wales, Australia.,Nepean Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Vanmassenhove J, Van Biesen W, Vanholder R, Lameire N. Subclinical AKI: ready for primetime in clinical practice? J Nephrol 2018; 32:9-16. [PMID: 30523562 DOI: 10.1007/s40620-018-00566-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
Abstract
There has been considerable progress over the last decade in the standardization of the acute kidney injury (AKI) definition with the publication of the RIFLE, AKIN, KDIGO and ERBP classification criteria. However, these classification criteria still rely on imperfect parameters such as serum creatinine and urinary output. The use of timed urine collections, kinetic eGFR (estimated glomerular filtration rate), real time measurement of GFR and direct measures of tubular damage can theoretically aid in a more timely diagnosis of AKI and improve patients' outcome. There has been an extensive search for new biomarkers indicative of structural tubular damage but it remains controversial whether these new markers should be included in the current classification criteria. The use of these markers has also led to the creation of a new concept called subclinical AKI, a condition where there is an increase in biomarkers but without clinical AKI, defined as an increase in serum creatinine and/or a decrease in urinary output. In this review we provide a framework on how to critical appraise biomarker research and on how to position the concept of subclinical AKI. The evaluation of biomarker performance and the usefulness of the concept 'subclinical AKI' requires careful consideration of the context these biomarkers are used in (clinical versus research setting) and the goal we want to achieve (risk assessment versus prediction versus early diagnosis versus prognostication). It remains currently unknown whether an increase in biomarkers levels without functional repercussion is clinically relevant and whether including biomarkers in classification criteria will improve patients' outcome.
Collapse
Affiliation(s)
- Jill Vanmassenhove
- Renal Division, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Wim Van Biesen
- Renal Division, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Raymond Vanholder
- Renal Division, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Norbert Lameire
- Renal Division, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
9
|
Barriers and Advances in Kidney Preservation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9206257. [PMID: 30643824 PMCID: PMC6311271 DOI: 10.1155/2018/9206257] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/15/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022]
Abstract
Despite the fact that a significant fraction of kidney graft dysfunctions observed after transplantation is due to ischemia-reperfusion injuries, there is still no clear consensus regarding optimal kidney preservation strategy. This stems directly from the fact that as of yet, the mechanisms underlying ischemia-reperfusion injury are poorly defined, and the role of each preservation parameter is not clearly outlined. In the meantime, as donor demography changes, organ quality is decreasing which directly increases the rate of poor outcome. This situation has an impact on clinical guidelines and impedes their possible harmonization in the transplant community, which has to move towards changing organ preservation paradigms: new concepts must emerge and the definition of a new range of adapted preservation method is of paramount importance. This review presents existing barriers in transplantation (e.g., temperature adjustment and adequate protocol, interest for oxygen addition during preservation, and clear procedure for organ perfusion during machine preservation), discusses the development of novel strategies to overcome them, and exposes the importance of identifying reliable biomarkers to monitor graft quality and predict short and long-term outcomes. Finally, perspectives in therapeutic strategies will also be presented, such as those based on stem cells and their derivatives and innovative models on which they would need to be properly tested.
Collapse
|
10
|
Wiebe C, Ho J, Gibson IW, Rush DN, Nickerson PW. Carpe diem-Time to transition from empiric to precision medicine in kidney transplantation. Am J Transplant 2018; 18:1615-1625. [PMID: 29603637 DOI: 10.1111/ajt.14746] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 02/06/2023]
Abstract
The current immunosuppressive pipeline in kidney transplantation is limited. In part, this is due to excellent one-year allograft outcomes with the current standard of care (ie, calcineurin inhibitor in combination with anti-proliferative agents). Despite this success, a recent Federal government-sponsored systematic review has identified gaps/limits in the evidence of what constitutes optimal calcineurin inhibitor use in the short- and long-term. Moreover, recent empiric approaches to minimize/withdraw/convert from calcineurin inhibitors have come with the price of increased alloreactivity. As the time horizon to replace calcineurin inhibitors on a global scale may be distant, the transplant community should seize the opportunity to develop ways to personalize calcineurin inhibitor immunosuppression to the individual-transitioning from empiricism to precision. The authors argue in this viewpoint that the path to precision will require measures capable of detecting subclinical alloreactivity to define adequacy of immunosuppression, as well as novel genetic analytics to accurately define alloimmune risk at the individual level-both approaches will require validation in clinical trials.
Collapse
Affiliation(s)
- Chris Wiebe
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Diagnostic Services of Manitoba, Winnipeg, MB, Canada
| | - Julie Ho
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Ian W Gibson
- Diagnostic Services of Manitoba, Winnipeg, MB, Canada.,Department of Pathology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - David N Rush
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Peter W Nickerson
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Diagnostic Services of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
11
|
Abstract
Chronic kidney disease (CKD) has become a significant public health concern, as it is associated with substantial morbidity. Prior research has evaluated multiple novel CKD biomarkers to supplement serum creatinine and proteinuria. The ultimate goal of this research is to find biomarkers that can be used to accurately predict CKD progression and to better time outpatient follow-up, and referral for transplant. Also, an optimal panel of biomarkers can augment the predictive value of proteinuria and serum creatinine by enriching patient enrollment in clinical trials. In this review, we discuss salient findings on 12 candidate plasma and urine biomarkers and their reported association with CKD. We explore the common pathways of CKD progression and the pathophysiologic processes of tubulointerstitial injury, inflammation, repair, and fibrosis that are potentially classified by specific biomarkers. We describe both pediatric and adult findings and highlight the paucity of pediatric research in CKD progression. It will be important for cohorts with longitudinal follow-up to evaluate these CKD biomarkers for potential use in pediatric clinical trials and routine CKD management.
Collapse
|
12
|
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) leading to delayed graft function (DGF), defined by the United Network for Organ Sharing as dialysis in the first week (UNOS-DGF), associates with poor kidney transplant outcomes. Controversies remain, however, about dialysis initiation thresholds and the utility for other criteria to denote less severe IRI, or slow graft function (SGF). METHODS Multicenter, prospective study of deceased-donor kidney recipients to compare UNOS-DGF to a definition that combines impaired creatinine reduction in the first 48 hours or greater than 1 dialysis session for predicting 12-month estimated glomerular filtration rate (eGFR). We also assessed 10 creatinine and urine output-based SGF definitions relative to 12-month eGFR. RESULTS In 560 recipients, 215 (38%) had UNOS-DGF, 330 (59%) met the combined definition, 14 (3%) died, and 23 (4%) had death-censored graft failure by 12 months. Both DGF definitions were associated with lower adjusted 12-month eGFR (95% confidence interval)-by 7.3 (3.6-10.9) and 7.4 (3.8-11.0) mL/min per 1.73 m, respectively. Adjusted relative risks for 12-month eGFR less than 30 mL/min per 1.73 m were 1.9 (1.2-3.1) and 2.1 (1.1-3.7), with unadjusted areas under the curve of 0.618 and 0.627, respectively. For SGF definitions, postoperative day (POD) 7 creatinine had the strongest association with 12-month eGFR, and POD5 creatinine and creatinine reduction between POD1 and POD2 demonstrated modest separations in 12-month eGFR. CONCLUSIONS Although UNOS-DGF does not adequately predict 12-month function on its own, our findings do not support changing the definition. Postoperative day 7 creatinine is correlated with 12-month eGFR, but large translational studies are needed to understand the biological link between IRI severity at transplant and longer-term outcomes.
Collapse
|
13
|
Abstract
BACKGROUND Kidneys with "high" Kidney Donor Profile Index (KDPI) are often biopsied and pumped, yet frequently discarded. METHODS In this multicenter study, we describe the characteristics and outcomes of kidneys with KDPI of 80 or greater that were procured from 338 deceased donors. We excluded donors with anatomical kidney abnormalities. RESULTS Donors were categorized by the number of kidneys discarded: (1) none (n = 154, 46%), (2) 1 discarded and 1 transplanted (n = 48, 14%), (3) both discarded (n = 136, 40%). Donors in group 3 were older, more often white, and had higher terminal creatinine and KDPI than group 1 (all P < 0.05). Biopsy was performed in 92% of all kidneys, and 47% were pumped. Discard was associated with biopsy findings and first hour renal resistance. Kidney injury biomarker levels (neutrophil gelatinase-associated lipocalin, IL-18, and kidney injury molecule-1 measured from donor urine at procurement and from perfusate soon after pump perfusion) were not different between groups. There was no significant difference in 1-year estimated glomerular filtration rate or graft failure between groups 1 and 2 (41.5 ± 18 vs 41.4 ± 22 mL/min per 1.73 m; P = 0.97 and 9% vs 10%; P = 0.76). CONCLUSIONS Kidneys with KDPI of 80 or greater comprise the most resource consuming fraction of our donor kidney pool and have the highest rates of discard. Our data suggest that some discarded kidneys with KDPI of 80 or greater are viable; however, current tools and urine and perfusate biomarkers to identify these viable kidneys are not satisfactory. We need better methods to assess viability of kidneys with high KDPI.
Collapse
|
14
|
Abstract
AKI is an increasingly common disorder that is strongly linked to short- and long-term morbidity and mortality. Despite a growing heterogeneity in its causes, providing a timely and certain diagnosis of AKI remains challenging. In this review, we summarize the evolution of AKI biomarker studies over the past few years, focusing on two major areas of investigation: the early detection and prognosis of AKI. We highlight some of the lessons learned in conducting AKI biomarker studies, including ongoing attempts to address the limitations of creatinine as a reference standard and the recent shift toward evaluating the prognostic potential of these markers. Lastly, we suggest current gaps in knowledge and barriers that may be hindering their incorporation into care and a full ascertainment of their value.
Collapse
Affiliation(s)
- Rakesh Malhotra
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, San Diego, California
| | - Edward D. Siew
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical center, Nashville, Tennessee
- Tennessee Valley Healthcare System, Veteran's Administration Medical Center, Veterans Health Administration, Nashville, Tennessee; and
- Vanderbilt Center for Kidney Disease and Integrated Program for Acute Kidney Injury Research, Nashville, Tennessee
| |
Collapse
|
15
|
Jafari A, Khatami MR, Dashti-Khavidaki S, Lessan-Pezeshki M, Abdollahi A. Plasma Neutrophil Gelatinase-Associated Lipocalin as a Marker for Prediction of 3-Month Graft Survival after Kidney Transplantation. Int J Organ Transplant Med 2017; 8:17-27. [PMID: 28299024 PMCID: PMC5347402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Ischemic injury during organ transplantation increases the risk of acute and chronic rejections by promoting alloimmune responses. Measurement of neutrophil gelatinase-associated lipocalin (NGAL) immediately after kidney transplantation may be promising for early detection of ischemic injuries to allograft. OBJECTIVE This study assessed possible predictive values of plasma NGAL levels during first hours after kidney transplantation for graft loss within the first 3 months after transplantation. METHODS 45 kidney transplant recipients were classified into those without graft loss or with graft loss during 3 months after transplantation. Plasma NGAL levels were measured before and at 2, 6, 12, 24 and 96 hours after transplantation. Serum creatinine concentration was assessed daily during hospitalization and at 1, 2, and 3 months post-transplantation. RESULTS Serum creatinine and plasma NGAL levels were consistently higher in patients with graft loss compared with those without graft loss. At 2, 24, and 96 hours after transplantation, plasma NGAL concentration was significantly higher in patients who developed allograft loss within 3 months post-transplantation. The cutoff point of plasma NGAL at 2, 24, and 96 hours after transplantation for prediction of graft loss was 304.5 ng/mL (sensitivity of 71.4%, and specificity of 73.7%), 207.8 ng/mL(sensitivity of 85.7%, and specificity of 60.5%), and 184 ng/mL (sensitivity of 85.7%, and specificity of 71.1%), respectively. CONCLUSION Plasma NGAL levels at 2, 24, and 96 hours after transplantation can predict 3-month graft loss with fair sensitivity and specificity.
Collapse
Affiliation(s)
- A. Jafari
- Assistant Professor of Clinical Pharmacy, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - M. R. Khatami
- Professor of Nephrology, Nephrology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - S. Dashti-Khavidaki
- Professor of Clinical Pharmacy, Nephrology Research Center, Tehran University of Medical Sciences, Tehran, Iran,Correspondence: Simin Dashti-Khavidaki, Professor of Clinical Pharmacy, Nephrology Research Center, Tehran University of Medical Sciences, Tehran, Iran. Center of Excellence in Nephrology, Tehran University of Medical Sciences, Tehran 1417614411, Iran,PO Box: 14155-6451, Tel/Fax:+98-21-6658-1568 , E-mail:
| | - M. Lessan-Pezeshki
- Professor of Nephrology, Nephrology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - A. Abdollahi
- Associate Professor of Pathology, Vali-e-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Does Delayed Graft Function Still Herald a Poorer Outcome in Kidney Transplantation? CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0110-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
17
|
Abstract
Acute kidney injury is strongly associated with increased mortality and other adverse outcomes. Medical researchers have intensively investigated novel biomarkers to predict short- and long-term outcomes of acute kidney injury in many patient care settings, such as cardiac surgery, intensive care units, heart failure, and transplant. Future research should focus on leveraging this relationship to improve enrollment for clinical trials of acute kidney injury.
Collapse
Affiliation(s)
- Jennifer A Schaub
- Program of Applied Translational Research, Yale University, New Haven, CT, USA
| | - Chirag R Parikh
- Program of Applied Translational Research, Yale University, New Haven, CT, USA
| |
Collapse
|
18
|
Lohkamp LN, Öllinger R, Chatzigeorgiou A, Illigens BMW, Siepmann T. Intraoperative biomarkers in renal transplantation. Nephrology (Carlton) 2016; 21:188-199. [PMID: 26132511 DOI: 10.1111/nep.12556] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 12/11/2022]
Abstract
The emerging need for biomarkers in the management of renal transplantation is highlighted by the severity of related complications such as acute renal failure and ischaemia/reperfusion injury (IRI) and by the increasing efforts to identify novel markers of these events to predict and monitor delayed graft function (DGF) and long-term outcome. In clinical studies candidate markers such as kidney injury molecule-1, neutrophil gelatinase-associated lipocalin and interleukin-18 have been demonstrated to be valid biomarkers with high predictive value for DFG in a post-transplant setting. However, studies investigating biomarkers for early diagnosis of IRI and assumable DGF as well as identification of potential graft recipients at increased risk at the time point of transplantation lack further confirmation and translation into clinical practice. This review summarizes the current literature on the value of IRI biomarkers in outcome prediction following renal transplantation as well their capacity as surrogate end points from an intraoperative perspective.
Collapse
Affiliation(s)
- Laura-Nanna Lohkamp
- Department of Neurosurgery with Pediatric Neurosurgery, Charité-University Medicine, Campus Virchow, Berlin, Germany
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany
| | - Robert Öllinger
- Department for General, Visceral and Transplantation Surgery, Charité-University Medicine, Campus Virchow, Berlin, Germany
| | - Antonios Chatzigeorgiou
- Department of Clinical Pathobiochemistry, Medical Faculty Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
- Paul-Langerhans Institute Dresden, German Center for Diabetes Research, Dresden, Germany
| | - Ben Min-Woo Illigens
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Timo Siepmann
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Bansal N, Carpenter MA, Weiner DE, Levey AS, Pfeffer M, Kusek JW, Cai J, Hunsicker LG, Park M, Bennett M, Liu KD, Hsu CY. Urine Injury Biomarkers and Risk of Adverse Outcomes in Recipients of Prevalent Kidney Transplants: The Folic Acid for Vascular Outcome Reduction in Transplantation Trial. J Am Soc Nephrol 2015; 27:2109-21. [PMID: 26538631 DOI: 10.1681/asn.2015030292] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022] Open
Abstract
Recipients of kidney transplants (KTR) are at increased risk for cardiovascular events, graft failure, and death. It is unknown whether urine kidney injury biomarkers are associated with poor outcomes among KTRs. We conducted a post hoc analysis of the Folic Acid for Vascular Outcome Reduction in Transplantation (FAVORIT) Trial using a case-cohort study design, selecting participants with adjudicated cardiovascular events, graft failure, or death. Urine neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), IL-18, and liver-type fatty acid binding protein (L-FABP) were measured in spot urine samples and standardized to urine creatinine concentration. We adjusted for demographics, cardiovascular risk factors, eGFR, and urine albumin-to-creatinine ratio. Patients had 291 cardiovascular events, 257 graft failure events, and 359 deaths. Each log increase in urine NGAL/creatinine independently associated with a 24% greater risk of cardiovascular events (adjusted hazard ratio [aHR], 1.24; 95% confidence interval [95% CI], 1.06 to 1.45), a 40% greater risk of graft failure (aHR, 1.40; 95% CI, 1.16 to 1.68), and a 44% greater risk of death (aHR, 1.44; 95% CI, 1.26 to 1.65). Urine KIM-1/creatinine and IL-18/creatinine independently associated with greater risk of death (aHR, 1.29; 95% CI, 1.03 to 1.61 and aHR, 1.25; 95% CI, 1.04 to 1.49 per log increase, respectively) but not with risk of cardiovascular events or graft failure. Urine L-FABP did not associate with any study outcomes. In conclusion, among prevalent KTRs, higher urine NGAL, KIM-1, and IL-18 levels independently and differentially associated with greater risk of adverse outcomes.
Collapse
Affiliation(s)
- Nisha Bansal
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington;
| | - Myra A Carpenter
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Daniel E Weiner
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Marc Pfeffer
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - John W Kusek
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Lawrence G Hunsicker
- College of Medicine, Department of Medicine, University of Iowa, Iowa City, Iowa
| | - Meyeon Park
- Division of Nephrology, University of California, San Francisco, California; and
| | - Michael Bennett
- Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Kathleen D Liu
- Division of Nephrology, University of California, San Francisco, California; and
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California, San Francisco, California; and
| |
Collapse
|
20
|
Reese PP, Hall IE, Weng FL, Schröppel B, Doshi MD, Hasz RD, Thiessen-Philbrook H, Ficek J, Rao V, Murray P, Lin H, Parikh CR. Associations between Deceased-Donor Urine Injury Biomarkers and Kidney Transplant Outcomes. J Am Soc Nephrol 2015; 27:1534-43. [PMID: 26374609 DOI: 10.1681/asn.2015040345] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/23/2015] [Indexed: 12/13/2022] Open
Abstract
Assessment of deceased-donor organ quality is integral to transplant allocation practices, but tools to more precisely measure donor kidney injury and better predict outcomes are needed. In this study, we assessed associations between injury biomarkers in deceased-donor urine and the following outcomes: donor AKI (stage 2 or greater), recipient delayed graft function (defined as dialysis in first week post-transplant), and recipient 6-month eGFR. We measured urinary concentrations of microalbumin, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), IL-18, and liver-type fatty acid binding protein (L-FABP) from 1304 deceased donors at organ procurement, among whom 112 (9%) had AKI. Each biomarker strongly associated with AKI in adjusted analyses. Among 2441 kidney transplant recipients, 31% experienced delayed graft function, and mean±SD 6-month eGFR was 55.7±23.5 ml/min per 1.73 m(2) In analyses adjusted for donor and recipient characteristics, higher donor urinary NGAL concentrations associated with recipient delayed graft function (highest versus lowest NGAL tertile relative risk, 1.21; 95% confidence interval, 1.02 to 1.43). Linear regression analyses of 6-month recipient renal function demonstrated that higher urinary NGAL and L-FABP concentrations associated with slightly lower 6-month eGFR only among recipients without delayed graft function. In summary, donor urine injury biomarkers strongly associate with donor AKI but provide limited value in predicting delayed graft function or early allograft function after transplant.
Collapse
Affiliation(s)
- Peter P Reese
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, Department of Biostatistics and Epidemiology, and Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Isaac E Hall
- Program of Applied Translational Research, Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | - Rick D Hasz
- Gift of Life Institute, Philadelphia, Pennsylvania
| | | | - Joseph Ficek
- Program of Applied Translational Research, Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Veena Rao
- Program of Applied Translational Research, Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Patrick Murray
- School of Medicine & Medical Science, University College Dublin, Dublin, Ireland; and
| | - Haiqun Lin
- Program of Applied Translational Research, Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Chirag R Parikh
- Program of Applied Translational Research, Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut; Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut
| |
Collapse
|
21
|
|
22
|
Pianta TJ, Peake PW, Pickering JW, Kelleher M, Buckley NA, Endre ZH. Evaluation of biomarkers of cell cycle arrest and inflammation in prediction of dialysis or recovery after kidney transplantation. Transpl Int 2015; 28:1392-404. [DOI: 10.1111/tri.12636] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/30/2014] [Accepted: 07/07/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Timothy J. Pianta
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
- Northern Clinical School; Melbourne Medical School; University of Melbourne; Epping Vic Australia
| | - Philip W. Peake
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
| | - John W. Pickering
- Department of Medicine; University of Otago; Christchurch New Zealand
| | - Michaela Kelleher
- Department of Nephrology; Prince of Wales Hospital; Sydney NSW Australia
| | | | - Zoltan H. Endre
- Prince of Wales Clinical School; University of New South Wales; Sydney NSW Australia
- Department of Medicine; University of Otago; Christchurch New Zealand
| |
Collapse
|
23
|
Huen SC, Parikh CR. Molecular phenotyping of clinical AKI with novel urinary biomarkers. Am J Physiol Renal Physiol 2015; 309:F406-13. [PMID: 26084933 DOI: 10.1152/ajprenal.00682.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/10/2015] [Indexed: 01/09/2023] Open
Abstract
Acute kidney injury (AKI) is a common hospital complication. There are no effective treatments to minimize kidney injury or limit associated morbidity and mortality. Currently, serum creatinine and urine output remain the gold standard used clinically in the diagnosis of AKI. Several novel biomarkers can diagnose AKI earlier than elevations of serum creatinine and changes in urine output. Recent long-term observational studies have elucidated a subgroup of patients who have positive biomarkers of AKI but do not meet criteria for AKI by serum creatinine or urine output, termed subclinical AKI. These patients with subclinical AKI have increased risk of both short- and long-term mortality. In this review, we will highlight the implications of what these patients may represent and the need for better phenotyping of AKI by etiology, severity of injury, and ability to recover. We will discuss two AKI biomarkers, neutrophil gelatinase-associated lipocalin (NGAL) and breast regression protein-39 (BRP-39)/YKL-40, that exemplify the need to characterize the complexity of the biological meaning behind the biomarker, beyond elevated levels reporting on tissue injury. Ultimately, careful phenotyping of AKI will lead to identification of therapeutic targets and appropriate patient populations for clinical trials.
Collapse
Affiliation(s)
- Sarah C Huen
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut; and
| | - Chirag R Parikh
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut; and Program of Applied Translational Research, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Hall IE, Schröppel B, Doshi MD, Ficek J, Weng FL, Hasz RD, Thiessen-Philbrook H, Reese PP, Parikh CR. Associations of deceased donor kidney injury with kidney discard and function after transplantation. Am J Transplant 2015; 15:1623-31. [PMID: 25762442 PMCID: PMC4563988 DOI: 10.1111/ajt.13144] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 02/06/2023]
Abstract
Deceased donor kidneys with acute kidney injury (AKI) are often discarded due to fear of poor outcomes. We performed a multicenter study to determine associations of AKI (increasing admission-to-terminal serum creatinine by AKI Network stages) with kidney discard, delayed graft function (DGF) and 6-month estimated glomerular filtration rate (eGFR). In 1632 donors, kidney discard risk increased for AKI stages 1, 2 and 3 (compared to no AKI) with adjusted relative risks of 1.28 (1.08-1.52), 1.82 (1.45-2.30) and 2.74 (2.0-3.75), respectively. Adjusted relative risk for DGF also increased by donor AKI stage: 1.27 (1.09-1.49), 1.70 (1.37-2.12) and 2.25 (1.74-2.91), respectively. Six-month eGFR, however, was similar across AKI categories but was lower for recipients with DGF (48 [interquartile range: 31-61] vs. 58 [45-75] ml/min/1.73m(2) for no DGF, p < 0.001). There was significant favorable interaction between donor AKI and DGF such that 6-month eGFR was progressively better for DGF kidneys with increasing donor AKI (46 [29-60], 49 [32-64], 52 [36-59] and 58 [39-71] ml/min/1.73m(2) for no AKI, stage 1, 2 and 3, respectively; interaction p = 0.05). Donor AKI is associated with kidney discard and DGF, but given acceptable 6-month allograft function, clinicians should consider cautious expansion into this donor pool.
Collapse
Affiliation(s)
- Isaac E. Hall
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT,Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Bernd Schröppel
- Section of Nephrology, Department of Internal Medicine 1, University Hospital, Ulm, Germany
| | | | - Joseph Ficek
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | | | | | | | | | - Chirag R. Parikh
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT,Section of Nephrology, Yale University School of Medicine, New Haven, CT,Veterans Affairs Connecticut Healthcare System, New Haven, CT
| |
Collapse
|
25
|
Clusterin in kidney transplantation: novel biomarkers versus serum creatinine for early prediction of delayed graft function. Transplantation 2015; 99:171-9. [PMID: 25083615 DOI: 10.1097/tp.0000000000000256] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES Current methods for rapid detection of delayed graft function (DGF) after kidney transplantation are unreliable. Urinary clusterin is a biomarker of kidney injury but its utility for prediction of graft dysfunction is unknown. METHODS In a single-center, prospective cohort study of renal transplant recipients (N=81), urinary clusterin was measured serially between 4 hr and 7 days after transplantation. The utility of clusterin for prediction of DGF (hemodialysis within 7 days of transplantation) was compared with urinary interleukin (IL)-18, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1, serum creatinine, and clinical variables. RESULTS At 4 hr after reperfusion, anuria was highly specific, but of low sensitivity for detection of DGF. At 4 hr, receiver operating characteristic analysis suggested that urinary clusterin, IL-18, kidney injury molecule-1, and NGAL concentration were predictive of DGF. After adjusting for preoperative clinical variables and anuria, clusterin and IL-18 independently enhanced the clinical model for prediction of DGF. Kidney injury molecule-1 only modestly improved the prediction of DGF, whereas NGAL, serum creatinine, and the creatinine reduction ratio did not improve on the clinical model. At 12 hr, the creatinine reduction ratio independently predicted DGF. CONCLUSION Both urinary clusterin and IL-18 are useful biomarkers and may allow triaging of patients with DGF within 4 hr of transplantation. Relative performance of biomarkers for prediction of graft function is time-dependant. Early and frequent measurements of serum creatinine and calculation of the creatinine reduction ratio also predict DGF within 12 hr of reperfusion.
Collapse
|
26
|
Pretransplantation Recipient Regulatory T cell Suppressive Function Predicts Delayed and Slow Graft Function after Kidney Transplantation. Transplantation 2014; 98:745-53. [DOI: 10.1097/tp.0000000000000219] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
Hall IE, Stern EP, Cantley LG, Elias JA, Parikh CR. Urine YKL-40 is associated with progressive acute kidney injury or death in hospitalized patients. BMC Nephrol 2014; 15:133. [PMID: 25128003 PMCID: PMC4144686 DOI: 10.1186/1471-2369-15-133] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 08/08/2014] [Indexed: 01/08/2023] Open
Abstract
Background A translational study in renal transplantation suggested YKL-40, a chitinase 3-like-1 gene product, plays an important role in acute kidney injury (AKI) and repair, but data are lacking about this protein in urine from native human kidneys. Methods This is an ancillary study to a single-center, prospective observational cohort of patients with clinically-defined AKI according to AKI Network serum creatinine criteria. We determined the association of YKL -40 ≥ 5 ng/ml, alone or combined with neutrophil gelatinase-associated lipocalin (NGAL), in urine collected on the first day of AKI with a clinically important composite outcome (progression to higher AKI stage and/or in-hospital death). Results YKL-40 was detectable in all 249 patients, but urinary concentrations were considerably lower than in previously measured deceased-donor kidney transplant recipients. Seventy-two patients (29%) progressed or died in-hospital, and YKL-40 ≥ 5 ng/ml had an adjusted odds ratio (95% confidence interval) for the outcome of 3.4 (1.5-7.7). The addition of YKL-40 to a clinical model for predicting the outcome resulted in a continuous net reclassification improvement of 29% (P = 0.04). In patients at high risk for the outcome based on NGAL concentrations in the upper quartile, YKL-40 further partitioned the cohort into moderate-risk and very high-risk groups. Conclusions Urine YKL-40 is associated with AKI progression and/or death in hospitalized patients and improves clinically determined risk reclassification. Combining YKL-40 with other AKI biomarkers like NGAL may further delineate progression risk, though additional studies are needed to determine whether YKL-40 has general applicability and to define its association with longer-term outcomes in AKI.
Collapse
Affiliation(s)
| | | | | | | | - Chirag R Parikh
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
28
|
Hall IE, Reese PP, Weng FL, Schröppel B, Doshi MD, Hasz RD, Reitsma W, Goldstein MJ, Hong K, Parikh CR. Preimplant histologic acute tubular necrosis and allograft outcomes. Clin J Am Soc Nephrol 2014; 9:573-82. [PMID: 24558049 PMCID: PMC3944773 DOI: 10.2215/cjn.08270813] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 12/04/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES The influence of deceased-donor AKI on post-transplant outcomes is poorly understood. The few published studies about deceased-donor preimplant biopsy have reported conflicting results regarding associations between AKI and recipient outcomes. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This multicenter study aimed to evaluate associations between deceased-donor biopsy reports of acute tubular necrosis (ATN) and delayed graft function (DGF), and secondarily for death-censored graft failure, first adjusting for the kidney donor risk index and then stratifying by donation after cardiac death (DCD) status. RESULTS Between March 2010 and April 2012, 651 kidneys (369 donors, 4 organ procurement organizations) were biopsied and subsequently transplanted, with ATN reported in 110 (17%). There were 262 recipients (40%) who experienced DGF and 38 (6%) who experienced graft failure. DGF occurred in 45% of kidneys with reported ATN compared with 39% without ATN (P=0.31) resulting in a relative risk (RR) of 1.13 (95% confidence interval [95% CI], 0.9 to 1.43) and a kidney donor risk index-adjusted RR of 1.11 (95% CI, 0.88 to 1.41). There was no significant difference in graft failure for kidneys with versus without ATN (8% versus 5%). In stratified analyses, the adjusted RR for DGF with ATN was 0.97 (95% CI, 0.7 to 1.34) for non-DCD kidneys and 1.59 (95% CI, 1.23 to 2.06) for DCD kidneys (P=0.02 for the interaction between ATN and DCD on the development of DGF). CONCLUSIONS Despite a modest association with DGF for DCD kidneys, this study reveals no significant associations overall between preimplant biopsy-reported ATN and the outcomes of DGF or graft failure. The potential benefit of more rigorous ATN reporting is unclear, but these findings provide little evidence to suggest that current ATN reports are useful for predicting graft outcomes or deciding to accept or reject allograft offers.
Collapse
Affiliation(s)
- Isaac E. Hall
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | - Rick D. Hasz
- Gift of Life Institute, Philadelphia, Pennsylvania
| | | | - Michael J. Goldstein
- Mount Sinai School of Medicine and New York Organ Donor Network, New York, New York; and
| | - Kwangik Hong
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Chirag R. Parikh
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
- Section of Nephrology, Veterans Affairs Medical Center, West Haven, Connecticut
| |
Collapse
|
29
|
Shabir S, Halimi JM, Cherukuri A, Ball S, Ferro C, Lipkin G, Benavente D, Gatault P, Baker R, Kiberd B, Borrows R. Predicting 5-year risk of kidney transplant failure: a prediction instrument using data available at 1 year posttransplantation. Am J Kidney Dis 2013; 63:643-51. [PMID: 24387794 DOI: 10.1053/j.ajkd.2013.10.059] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/18/2013] [Indexed: 11/11/2022]
Abstract
BACKGROUND Accurate prediction of kidney transplant failure remains imperfect. The objective of this study was to develop and validate risk scores predicting 5-year transplant failure, based on data available 12 months posttransplantation. STUDY DESIGN Development and then independent multicenter validation of risk scores predicting death-censored and overall transplant failure. SETTING & PARTICIPANTS Outcomes of kidney transplant recipients (n=651) alive with transplant function 12 months posttransplantation in Birmingham, United Kingdom, were used to develop models predicting transplant failure risk 5 years posttransplantation. The resulting risk scores were evaluated for prognostic utility (discrimination, calibration, and risk reclassification) in independent cohorts from Tours, France (n=736); Leeds, United Kingdom (n=787); and Halifax, Canada (n=475). PREDICTORS Weighted regression coefficients for baseline and 12-month demographic and clinical predictor characteristics. OUTCOMES Death-censored and overall transplant failure 5 years posttransplantation. MEASUREMENTS Baseline data and time to transplant failure. RESULTS Following model development, variables included in separate scores for death-censored and overall transplant failure included recipient age, sex, and race; acute rejection; transplant function; serum albumin level; and proteinuria. In the validation cohorts, these scores showed good to excellent discrimination for death-censored transplant failure (C statistics, 0.78-0.90) and moderate to good discrimination for overall transplant failure (C statistics, 0.75-0.81). Both scores demonstrated good calibration (Hosmer-Lemeshow P>0.05 in all cohorts). Compared with estimated glomerular filtration rate in isolation, application of the scores resulted in statistically significant and clinically relevant risk reclassification for death-censored transplant failure (net reclassification improvement [NRI], 36.1%-83.0%; all P<0.001) and overall transplant failure (NRI, 38.7%-53.5%; all P<0.001). Compared with the previously described US Renal Data System-based risk calculator, significant and relevant risk reclassification for overall transplant failure was seen (NRI, 30.0%; P<0.001). LIMITATIONS Validation is required in further populations. CONCLUSIONS These validated risk scores may be of prognostic utility in kidney transplantation, accurately identifying at-risk transplants, and informing clinicians and patients.
Collapse
Affiliation(s)
- Shazia Shabir
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | | | | | - Simon Ball
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Charles Ferro
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Graham Lipkin
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - David Benavente
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Philippe Gatault
- Service de Néphrologie-Immunologie clinique, CHU Tours, Tours, France
| | - Richard Baker
- Renal Transplant Unit, University of Leeds, Leeds, United Kingdom
| | - Bryce Kiberd
- Division of Nephrology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Richard Borrows
- Department of Nephrology, Queen Elizabeth Hospital, Birmingham, United Kingdom; Centre for Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
30
|
Coca SG, Garg AX, Thiessen-Philbrook H, Koyner JL, Patel UD, Krumholz HM, Shlipak MG, Parikh CR. Urinary biomarkers of AKI and mortality 3 years after cardiac surgery. J Am Soc Nephrol 2013; 25:1063-71. [PMID: 24357673 DOI: 10.1681/asn.2013070742] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Urinary biomarkers of AKI provide prognostic value for in-hospital outcomes, but little is known about their association with longer-term mortality after surgery. We sought to assess the association between kidney injury biomarkers and all-cause mortality in an international, multicenter, prospective long-term follow-up study from six clinical centers in the United States and Canada composed of 1199 adults who underwent cardiac surgery between 2007 and 2009 and were enrolled in the Translational Research in Biomarker Endpoints in AKI cohort. On postoperative days 1-3, we measured the following five urinary biomarkers: neutrophil gelatinase-associated lipocalin, IL-18, kidney injury molecule-1 (KIM-1), liver fatty acid binding protein, and albumin. During a median follow-up of 3.0 years (interquartile range, 2.2-3.6 years), 139 participants died (55 deaths per 1000 person-years). Among patients with clinical AKI, the highest tertiles of peak urinary neutrophil gelatinase-associated lipocalin, IL-18, KIM-1, liver fatty acid binding protein, and albumin associated independently with a 2.0- to 3.2-fold increased risk for mortality compared with the lowest tertiles. In patients without clinical AKI, the highest tertiles of peak IL-18 and KIM-1 also associated independently with long-term mortality (adjusted hazard ratios [95% confidence intervals] of 1.2 [1.0 to 1.5] and 1.8 [1.4 to 2.3] for IL-18 and KIM-1, respectively), and yielded continuous net reclassification improvements of 0.26 and 0.37, respectively, for the prediction of 3-year mortality. In conclusion, urinary biomarkers of kidney injury, particularly IL-18 and KIM-1, in the immediate postoperative period provide additional prognostic information for 3-year mortality risk in patients with and without clinical AKI.
Collapse
Affiliation(s)
- Steven G Coca
- Section of Nephrology, Yale University School of Medicine, Program of Applied Translational Research, Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut
| | - Amit X Garg
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | | | - Jay L Koyner
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Uptal D Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Harlan M Krumholz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, Connecticut; and
| | - Michael G Shlipak
- Division of General Internal Medicine, San Francisco Veterans Affairs Medical Center, University of California, San Francisco
| | - Chirag R Parikh
- Section of Nephrology, Yale University School of Medicine, Program of Applied Translational Research, Veterans Affairs Connecticut Healthcare System, New Haven, Connecticut;
| | | |
Collapse
|
31
|
Neutrophil gelatinase-associated lipocalin in kidney transplantation is an early marker of graft dysfunction and is associated with one-year renal function. J Transplant 2013; 2013:650123. [PMID: 24288591 PMCID: PMC3833111 DOI: 10.1155/2013/650123] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/07/2013] [Accepted: 09/20/2013] [Indexed: 12/13/2022] Open
Abstract
Urinary neutrophil gelatinase-associated lipocalin (uNGAL) has been suggested as potential early marker of delayed graft function (DGF) following kidney transplantation (KTx). We conducted a prospective study in 40 consecutive KTx recipients to evaluate serial changes of uNGAL within the first week after KTx and assess its performance in predicting DGF (dialysis requirement during initial posttransplant week) and graft function throughout first year. Urine samples were collected on post-KTx days 0, 1, 2, 4, and 7. Linear mixed and multivariable regression models, receiver-operating characteristic (ROC), and areas under ROC curves were used. At all-time points, mean uNGAL levels were significantly higher in patients developing DGF (n = 18). Shortly after KTx (3–6 h), uNGAL values were higher in DGF recipients (on average +242 ng/mL, considering mean dialysis time of 4.1 years) and rose further in following days, contrasting with prompt function recipients. Day-1 uNGAL levels accurately predicted DGF (AUC-ROC = 0.93), with a performance higher than serum creatinine (AUC-ROC = 0.76), and similar to cystatin C (AUC-ROC = 0.95). Multivariable analyses revealed that uNGAL levels at days 4 and 7 were strongly associated with one-year serum creatinine. Urinary NGAL is an early marker of graft injury and is independently associated with dialysis requirement within one week after KTx and one-year graft function.
Collapse
|
32
|
Koyner JL, Parikh CR. Clinical utility of biomarkers of AKI in cardiac surgery and critical illness. Clin J Am Soc Nephrol 2013; 8:1034-42. [PMID: 23471130 DOI: 10.2215/cjn.05150512] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AKI is a common and serious complication that is associated with several adverse outcomes in hospitalized patients. The past several years have seen a large number of multicenter investigations of biomarkers of AKI in the setting of cardiac surgery and critical illness. This review summarizes these biomarker results to identify applications for clinical use. The Translational Research Investigating Biomarker Endpoints in AKI (TRIBE-AKI) study showed that blood and urine biomarkers measured preoperatively, immediately postoperatively, and at the time of the clinical increase in serum creatinine in the setting of cardiac surgery all had the ability to improve patient risk stratification for a variety of important clinical end points. Analyses of biomarkers concentrations from the Acute Respiratory Distress Syndrome Network, EARLY ARF, and other studies of critically ill subjects have similarly shown that biomarkers measured early in the clinical course can forecast the development of AKI and need for renal replacement therapy as well as inpatient mortality. Although biomarkers have informed the diagnosis, prognosis, and treatment of AKI and are inching closer to clinical application, large multicenter interventional clinical trials to prevent AKI using biomarkers should continue to be an active area of clinical investigation.
Collapse
Affiliation(s)
- Jay L Koyner
- Section of Nephrology, Department of Medicine, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| | | |
Collapse
|