1
|
Désy O, Thivierge MP, Béland S, Desgagnés JS, Bouchard-Boivin F, Gama A, Houde I, Lapointe I, Côté I, Lesage J, De Serres SA. A Risk Score Using a Cell-based Assay Predicts Long-term Over-immunosuppression Events in Kidney Transplant Recipients. Transplantation 2024:00007890-990000000-00952. [PMID: 39665497 DOI: 10.1097/tp.0000000000005279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
BACKGROUND Infections and cancer are major causes of premature death in organ recipients. However, there have been few advances in personalized immunosuppressive therapy. We previously reported that a cell-based assay measuring CD14+16+tumor necrosis factor-α+ monocytes after peripheral blood mononuclear cell (PBMC) incubation with Epstein-Barr virus peptides has a high sensitivity for detecting over-immunosuppression (OIS) events in kidney recipients in the short term. We aimed to develop a risk score for predicting long-term events. METHODS We studied 551 PBMC samples from 118 kidney recipients. Following random allocation to a testing and training set, we derived a risk function for the delineated tertiles of low, intermediate, and high risk of OIS based on age and CD14+16+tumor necrosis factor-α+ cells. RESULTS Patients were followed for a median of 6.3 y (25th-75th percentiles: 3.7-8.3 y). Of these, 40 (34%) experienced an OIS event. The validation indicated that the risk score was well calibrated, with an absolute risk of 21%, 41%, and 61% in the low-, intermediate-, and high-risk categories, respectively (P = 0.03). In sensitivity analyses, the risk score was robust to alternative definitions of OIS ranging from mild to severe. In particular, when restricted to life-threatening OIS, the proportion of events varied from 5% to 27% among the low- and high-risk categories, respectively (P = 0.01). CONCLUSIONS Using a combination of age and in vitro PBMC response to Epstein-Barr virus peptides allows a substantial shift in the estimated risk of OIS events.
Collapse
Affiliation(s)
- Olivier Désy
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - Marie-Pier Thivierge
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - Stéphanie Béland
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - Jean-Simon Desgagnés
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - François Bouchard-Boivin
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - Alcino Gama
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
| | - Isabelle Houde
- Transplantation Unit, Renal Division, Department of Medicine, Faculty of Medicine, University Health Center of Quebec, Laval University, QC, Canada
| | - Isabelle Lapointe
- Transplantation Unit, Renal Division, Department of Medicine, Faculty of Medicine, University Health Center of Quebec, Laval University, QC, Canada
| | - Isabelle Côté
- Transplantation Unit, Renal Division, Department of Medicine, Faculty of Medicine, University Health Center of Quebec, Laval University, QC, Canada
| | - Julie Lesage
- Transplantation Unit, Renal Division, Department of Medicine, Faculty of Medicine, University Health Center of Quebec, Laval University, QC, Canada
| | - Sacha A De Serres
- Department of Medicine, Faculty of Medicine, University Health Center (CHU) of Quebec Research Center, Laval University, QC, Canada
- Transplantation Unit, Renal Division, Department of Medicine, Faculty of Medicine, University Health Center of Quebec, Laval University, QC, Canada
| |
Collapse
|
2
|
Ettenger RB, Seifert ME, Blydt-Hansen T, Briscoe DM, Holman J, Weng PL, Srivastava R, Fleming J, Malekzadeh M, Pearl M. Detection of Subclinical Rejection in Pediatric Kidney Transplantation: Current and Future Practices. Pediatr Transplant 2024; 28:e14836. [PMID: 39147695 DOI: 10.1111/petr.14836] [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: 04/18/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
INTRODUCTION The successes in the field of pediatric kidney transplantation over the past 60 years have been extraordinary. Year over year, there have been significant improvements in short-term graft survival. However, improvements in longer-term outcomes have been much less apparent. One important contributor has been the phenomenon of low-level rejection in the absence of clinical manifestations-so-called subclinical rejection (SCR). METHODS Traditionally, rejection has been diagnosed by changes in clinical parameters, including but not limited to serum creatinine and proteinuria. This review examines the shortcomings of this approach, the effects of SCR on kidney allograft outcome, the benefits and drawbacks of surveillance biopsies to identify SCR, and new urine and blood biomarkers that define the presence or absence of SCR. RESULTS Serum creatinine is an unreliable index of SCR. Surveillance biopsies are the method most utilized to detect SCR. However, these have significant drawbacks. New biomarkers show promise. These biomarkers include blood gene expression profiles and donor derived-cell free DNA; urine gene expression profiles; urinary cytokines, chemokines, and metabolomics; and other promising blood and urine tests. CONCLUSION Specific emphasis is placed on studies carried out in pediatric kidney transplant recipients. TRIAL REGISTRATION ClinicalTrials.gov: NCT03719339.
Collapse
Affiliation(s)
- Robert B Ettenger
- Division of Nephrology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michael E Seifert
- Division of Pediatric Nephrology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Tom Blydt-Hansen
- Multi-Organ Transplant Program, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - David M Briscoe
- Division of Nephrology, Department of Pediatrics Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John Holman
- Transplant Genomics Inc., Framingham, Massachusetts, USA
| | - Patricia L Weng
- Division of Nephrology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rachana Srivastava
- Division of Nephrology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - James Fleming
- Transplant Genomics Inc., Framingham, Massachusetts, USA
| | - Mohammed Malekzadeh
- Division of Nephrology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Meghan Pearl
- Division of Nephrology, Department of Pediatrics, UCLA Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
3
|
Ho J, Schaub S, Jackson AM, Balshaw R, Carroll R, Cun S, De Serres SA, Fantus D, Handschin J, Hönger G, Jevnikar AM, Kleiser M, Lee JH, Li Y, Nickerson P, Pei R, Pochinco D, Shih R, Trinh M, Wang J, Nguyen J, Knechtle S. Multicenter Validation of a Urine CXCL10 Assay for Noninvasive Monitoring of Renal Transplants. Transplantation 2023; 107:1630-1641. [PMID: 36949034 DOI: 10.1097/tp.0000000000004554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
BACKGROUND Urine CXCL10 (C-X-C motif chemokine ligand 10, interferon gamma-induced protein 10 [IP10]) outperforms standard-of-care monitoring for detecting subclinical and early clinical T-cell-mediated rejection (TCMR) and may advance TCMR therapy development through biomarker-enriched trials. The goal was to perform an international multicenter validation of a CXCL10 bead-based immunoassay (Luminex) for transplant surveillance and compare with an electrochemiluminescence-based (Meso Scale Discovery [MSD]) assay used in transplant trials. METHODS Four laboratories participated in the Luminex assay development and evaluation. Urine CXCL10 was measured by Luminex and MSD in 2 independent adult kidney transplant trial cohorts (Basel and TMCT04). In an independent test and validation set, a linear mixed-effects model to predict (log 10 -transformed) MSD CXCL10 from Luminex CXCL10 was developed to determine the conversion between assays. Net reclassification was determined after mathematical conversion. RESULTS The Luminex assay was precise, with an intra- and interassay coefficient of variation 8.1% and 9.3%; showed modest agreement between 4 laboratories (R 0.96 to 0.99, P < 0.001); and correlated with known CXCL10 in a single- (n = 100 urines, R 0.94 to 0.98, P < 0.001) and multicenter cohort (n = 468 urines, R 0.92, P < 0.001) but the 2 assays were not equivalent by Passing-Bablok regression. Linear mixed-effects modeling demonstrated an intercept of -0.490 and coefficient of 1.028, showing Luminex CXCL10 are slightly higher than MSD CXCL10, but the agreement is close to 1.0. After conversion of the biopsy thresholds, the decision to biopsy would be changed for only 6% (5/85) patients showing acceptable reclassification. CONCLUSIONS These data demonstrate this urine CXCL10 Luminex immunoassay is robust, reproducible, and accurate, indicating it can be readily translated into clinical HLA laboratories for serial posttransplant surveillance.
Collapse
Affiliation(s)
- Julie Ho
- Department of Internal Medicine and Immunology, University of Manitoba, Winnipeg, Canada
- Transplant Manitoba, Shared Health Manitoba, Winnipeg, Canada
| | - Stefan Schaub
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
- HLA-Diagnostic and Immunogenetics, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | | | - Robert Balshaw
- George and Fay Yee Center for Healthcare Innovation, Manitoba, Canada
| | - Robert Carroll
- Royal Adelaide Hospital, University of Adelaide, SA, Australia
| | - Sylvia Cun
- Thermo Fisher Scientific, Los Angeles, CA
| | | | - Daniel Fantus
- Division of Nephrology, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) and Centre de Recherche du CHUM (CRCHUM), Montréal, Québec, Canada
| | - Joelle Handschin
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gideon Hönger
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
- HLA-Diagnostic and Immunogenetics, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Anthony M Jevnikar
- Department of Medicine, Western University and Multiorgan Transplant Program, London, ON, Canada
| | - Marc Kleiser
- HLA-Diagnostic and Immunogenetics, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | | | - Yan Li
- Department of Surgery and Immunology, Duke University, Durham, NC
| | - Peter Nickerson
- Department of Internal Medicine and Immunology, University of Manitoba, Winnipeg, Canada
- Transplant Manitoba, Shared Health Manitoba, Winnipeg, Canada
- Canadian Blood Services HLA Laboratory, Diagnostic Services of Manitoba, Canada
| | - Rui Pei
- Thermo Fisher Scientific, Los Angeles, CA
| | - Denise Pochinco
- Canadian Blood Services HLA Laboratory, Diagnostic Services of Manitoba, Canada
| | - Remi Shih
- Terasaki Innovation Center, Los Angeles, CA
| | | | - Jason Wang
- Thermo Fisher Scientific, Los Angeles, CA
| | | | - Stuart Knechtle
- Department of Surgery and Immunology, Duke University, Durham, NC
| |
Collapse
|
4
|
Brunet M, Millán O. Getting immunosuppression just right: the role of clinical biomarkers in predicting patient response post solid organ transplantation. Expert Rev Clin Pharmacol 2021; 14:1467-1479. [PMID: 34607521 DOI: 10.1080/17512433.2021.1987882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Actually, immunosuppression selection isn't based on individual immune alloreactivity, and immunosuppressive drug dosing is mainly based on the development of toxicity and the achievement of specific target concentrations. Since a successful outcome requires optimal patient risk stratification and treatment, several groups have evaluated candidate biomarkers that have shown promise in the assessment of individual immune responses, the prediction of personal pharmacodynamic effects of immunosuppressive drugs and the prognosis and diagnosis of graft outcomes.. AREAS COVERED This review includes biomarkers that the Scientific Community in Solid Organ Transplantation currently considers to have potential as diagnostic and prognostic biomarkers of graft evolution. We have focused on recent scientific advances and expert recommendations regarding the role of specific and non-specific pharmacodynamic biomarkers that are mainly involved in the T-cell-mediated response. EXPERT OPINION Integral pharmacologic monitoring that combines pharmacokinetics, pharmacogenetics and predictive pharmacodynamic biomarkers may provide crucial information and allow personal adjustment of immunosuppressive drugs at an early stage before severe adverse events ensue. Multicentre, randomized, prospective and interventional trials are needed to fine tune the established cut-off values for each biomarker and the optimal monitoring frequency for each biomarker and to accurately evaluate possible clinical confounding factors to enable correct clinical qualification.
Collapse
Affiliation(s)
- Mercè Brunet
- Pharmacology and Toxicology Section, CDB, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain.,Biomedical Research Center in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Millán
- Pharmacology and Toxicology Section, CDB, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain.,Biomedical Research Center in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Jang HR, Kim M, Hong S, Lee K, Park MY, Yang KE, Lee CJ, Jeon J, Lee KW, Lee JE, Park JB, Kim K, Kwon GY, Kim YG, Kim DJ, Huh W. Early postoperative urinary MCP-1 as a potential biomarker predicting acute rejection in living donor kidney transplantation: a prospective cohort study. Sci Rep 2021; 11:18832. [PMID: 34552150 PMCID: PMC8458304 DOI: 10.1038/s41598-021-98135-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the clinical relevance of urinary cytokines/chemokines reflecting intrarenal immunologic micromilieu as prognostic markers and the optimal measurement timing after living donor kidney transplantation (LDKT). This prospective cohort study included 77 LDKT patients who were followed for ≥ 5 years. Patients were divided into control (n = 42) or acute rejection (AR, n = 35) group. Early AR was defined as AR occurring within 3 months. Serum and urine cytokines/chemokines were measured serially as follows: intraoperative, 8/24/72 h, 1 week, 3 months, and 1 year after LDKT. Intrarenal total leukocytes, T cells, and B cells were analyzed with immunohistochemistry followed by tissueFAXS. Urinary MCP-1 and fractalkine were also analyzed in a validation cohort. Urinary MCP-1 after one week was higher in the AR group. Urinary MCP-1, fractalkine, TNF-α, RANTES, and IL-6 after one week were significantly higher in the early AR group. Intrarenal total leukocytes and T cells were elevated in the AR group compared with the control group. Urinary fractalkine, MCP-1, and IL-10 showed positive correlation with intrarenal leukocyte infiltration. Post-KT 1 week urinary MCP-1 showed predictive value in the validation cohort. One-week post-KT urinary MCP-1 may be used as a noninvasive diagnostic marker for predicting AR after LDKT.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Minjung Kim
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Sungjun Hong
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Mee Yeon Park
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Kyeong Eun Yang
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Cheol-Jung Lee
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Junseok Jeon
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Kyo Won Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Eun Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyunga Kim
- Statistics and Data Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon Goo Kim
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Dae Joong Kim
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Wooseong Huh
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
6
|
Handschin J, Wehmeier C, Amico P, Hopfer H, Dickenmann M, Schaub S, Hirt-Minkowski P. Urinary CXCL10 Measurement in Late Renal Allograft Biopsies Predicts Outcome Even in Histologically Quiescent Patients. Transplant Proc 2021; 53:2168-2179. [PMID: 34419254 DOI: 10.1016/j.transproceed.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/27/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND CXCL10 is a promising early noninvasive diagnostic marker for allograft rejection and predictive for long-term outcomes. However, its value when measured later in the posttransplant course has not yet been accurately analyzed. METHODS We investigated urinary CXCL10 in 141 patients from a prospective, observational renal transplant cohort with 182 clinically indicated allograft biopsies performed >12 months posttransplant and corresponding urines. Urinary CXCL10 was retrospectively quantified on stored urines using the MSD V-Plex Chemokine Panel 1 sandwich immunoassay (Meso Scale Discovery). The primary outcome was a composite of allograft loss/renal function decline (>30% estimated glomerular filtration rate [eGFR]-decrease between index biopsy and last follow-up). RESULTS Seventy-two patients (51%) reached the primary outcome, and their urinary CXCL10 levels were significantly higher at the time of their biopsy compared with patients with stable allograft function (median 9.3 ng/mmol vs 3.3 ng/mmol, P < .0001). Time-to-endpoint analyses according to high/low urinary CXCL10 demonstrated that low urinary CXCL10 (≤7.0 ng/mmol) was associated with 73% 5-year event-free graft survival compared with 48% with high urinary CXCL10 (>7.0 ng/mmol; P = .0001). Even in histologically quiescent patients, high urinary CXCL10 was associated with inferior endpoint-free graft survival (P = .003), and it was an independent predictor of the primary outcome (P = .03). CONCLUSIONS This study demonstrates that urinary CXCL10 has a promising diagnostic performance for detection of late allograft rejection and is an independent predictor of long-term renal allograft outcomes, even in histologically quiescent patients.
Collapse
Affiliation(s)
- Joelle Handschin
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Caroline Wehmeier
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Patrizia Amico
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Helmut Hopfer
- lnstitute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Michael Dickenmann
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Stefan Schaub
- Transplantation Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland; Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland; HLA-Diagnostic and lmmunogenetics, Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Patricia Hirt-Minkowski
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
7
|
Millán O, Rovira J, Guirado L, Espinosa C, Budde K, Sommerer C, Piñeiro GJ, Diekmann F, Brunet M. Advantages of plasmatic CXCL-10 as a prognostic and diagnostic biomarker for the risk of rejection and subclinical rejection in kidney transplantation. Clin Immunol 2021; 229:108792. [PMID: 34217849 DOI: 10.1016/j.clim.2021.108792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/22/2022]
Abstract
This study evaluate the potential of plasmatic CXCL-10 (pCXCL-10) as a pre&post transplantation prognostic and diagnostic biomarker of T-cell-mediated rejection (TCMR), antibody-mediated rejection (ABMR) and subclinical rejection (SCR) risk in adult kidney recipients considering BKV and CMV infections as possible clinical confounder factors. Twenty-eight of 100 patients included experienced rejection (TCMR:14; ABMR:14); 8 SCR; 13 and 16 were diagnosed with BKV and CMV infection, respectively. Pre-transplantation pCXCL-10 was significantly increased in TCMR and ABMR and post-transplantation in TCMR, ABMR and SCR compared with nonrejectors. All CMV+ patients showed pCXCL-10 levels above the cutoff values established for rejection whereas the 80% of BKV+ patients showed pCXCL-10 concentration < 100 pg/mL. pCXCL-10 could improve pre-transplantation patient stratification and immunosuppressive treatment selection according to rejection risk; and after kidney transplantation could be a potential early prognostic biomarker for rejection. Clinical confounding factor in BKV+ and particularly in CMV+ patients must be discarded.
Collapse
Affiliation(s)
- Olga Millán
- Pharmacology and Toxicology Section, CDB, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, c/Villarroel, 170, 08036 Barcelona, Spain; Biomedical Research Center in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, c/Sinesio Delgado 4, 28029 Madrid, Spain.
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), IDIBAPS, Barcelona, Spain; Red de Investigación Renal (REDINREN), Plaza de las Cortes, 11, 28014 Madrid, Spain.
| | - Lluis Guirado
- Renal Transplant Unit, Nephrology Department, Fundació Puigvert, Carrer de Cartagena, 340, 08025 Barcelona, Spain.
| | - Cristina Espinosa
- Pharmacology and Toxicology Section, CDB, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, c/Villarroel, 170, 08036 Barcelona, Spain.
| | - Klemens Budde
- Medizinische Klinik mit Schwerpunkt Nephrologie, Charité Universitätsmedizin Berlin, Campus Charité Mitte Luisenstraße 13, 10117 Berlin, Germany.
| | - Claudia Sommerer
- Department of Nephrology, University of Heidelberg, University Hospital of Heidelberg and Mannheim, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany.
| | - Gaston J Piñeiro
- Department of Nephrology and Kidney Transplantation, ICNU, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), IDIBAPS, Barcelona, Spain; Red de Investigación Renal (REDINREN), Plaza de las Cortes, 11, 28014 Madrid, Spain; Department of Nephrology and Kidney Transplantation, ICNU, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain.
| | - Mercè Brunet
- Pharmacology and Toxicology Section, CDB, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, c/Villarroel, 170, 08036 Barcelona, Spain; Biomedical Research Center in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, c/Sinesio Delgado 4, 28029 Madrid, Spain.
| |
Collapse
|
8
|
Tinel C, Anglicheau D. [Urinary biomarkers in kidney transplant recipients: From technological innovations to clinical development]. Nephrol Ther 2021; 17S:S83-S87. [PMID: 33910704 DOI: 10.1016/j.nephro.2020.03.003] [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: 01/31/2020] [Accepted: 03/01/2020] [Indexed: 11/16/2022]
Abstract
In kidney transplantation, the assessment of individual risks remains highly imperfect and highlights the need for robust noninvasive biomarkers with the overall goal to improve patient and graft outcomes. In the field of noninvasive biomarkers discovery, urinary biomarkers are promising tools which use easily accessible biological fluid. During the past decades, the technical revolution in the fields of genetics and molecular biology, and advances in chemistry and data analysis have led to a wealth of studies using urinary cell pellets or supernatants from kidney transplant recipients. Transcriptomic, proteomic and metabonomic analyses have suggested numerous signatures for the diagnoses of acute rejection, delayed-graft function or interstitial fibrosis. Nevertheless, the translation and validation of exploratory findings and their implementation into standard clinical practice remain challenging. This requires dedicated prospective interventional trials demonstrating that the use of these biomarkers avoids invasive procedures and improves patient or transplant outcomes.
Collapse
Affiliation(s)
- Claire Tinel
- Service de néphrologie et transplantation rénale, Assistance publique-Hôpitaux de Paris, 149, rue de Sèvres, 75015 Paris, France; Faculte de médecine Necker, Institut Necker-Enfants-Malades, 156-160, rue de Vaugirard, 75015 Paris, France; Inserm, U1151, 156-160, rue de Vaugirard, 75015 Paris, France.
| | - Dany Anglicheau
- Service de néphrologie et transplantation rénale, Assistance publique-Hôpitaux de Paris, 149, rue de Sèvres, 75015 Paris, France; Faculte de médecine Necker, Institut Necker-Enfants-Malades, 156-160, rue de Vaugirard, 75015 Paris, France; Inserm, U1151, 156-160, rue de Vaugirard, 75015 Paris, France
| |
Collapse
|
9
|
Arnau A, Benito-Hernández A, Ramos-Barrón MA, García-Unzueta MT, Gómez-Román JJ, Gómez-Ortega JM, López-Hoyos M, San Segundo D, Ruiz JC, Rodrigo E. Urinary C-X-C Motif Chemokine 10 Is Related to Acute Graft Lesions Secondary to T Cell- and Antibody-Mediated Damage. Ann Transplant 2021; 26:e929491. [PMID: 33686050 PMCID: PMC7955576 DOI: 10.12659/aot.929491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Non-invasive biomarkers of graft rejection are needed to optimize the management and outcomes of kidney transplant recipients. Urinary excretion of IFN-γ-related chemokine CXCL10 is clearly associated with clinical and subclinical T cell-mediated graft inflammation, but its relationship with antibody-mediated damage has not been fully addressed. Further, the variables influencing levels of urinary CXCL10 excretion are unknown. Material/Methods A total of 151 kidney graft biopsies (92 surveillance and 59 indication biopsies) and 151 matched urine samples obtained before biopsy were prospectively analyzed. T cell-mediated rejection (TCMR) and antibody-mediated rejection (AbMR) were defined according to the 2017 Banff classification criteria. Urinary CXCL10 levels were measured by ELISA and corrected by urinary creatinine. Results Banff scores ‘t’, ‘i’, ‘g’, and ‘ptc’ were significantly related to urinary CXCL10 levels. Multivariate analysis showed that ‘t’ (β=0.107, P=0.001) and ‘ptc’ (β=0.093, P=0.002) were significantly associated with urinary CXCL10. Donor-specific antibodies (DSAs) were related to the high excretion of urinary CXCL10 at 1 year after transplantation (odds ratio [OR] 17.817, P=0.003). Urinary CXCL10 showed good discrimination ability for AbMR (AUC-ROC 0.760, P=0.001). The third tertile of urinary CXCL10 remained significantly associated with AbMR (OR 4.577, 95% confidence interval 1.799–11.646, P=0.001) after multivariate regression analysis. Conclusions DSA was the only variable clearly related to high urinary CXCL10 levels. Urinary CXCL10 is a good non-invasive candidate biomarker of AbMR and TCMR, supplying information independent of renal function and other variables normally used to monitor kidney transplants.
Collapse
Affiliation(s)
- Alvaro Arnau
- Nephrology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Adalberto Benito-Hernández
- Nephrology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - María Angeles Ramos-Barrón
- Nephrology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - María Teresa García-Unzueta
- Department of Clinical Biochemistry, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - José Javier Gómez-Román
- Pathology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - José María Gómez-Ortega
- Pathology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - David San Segundo
- Immunology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Juan Carlos Ruiz
- Nephrology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Emilio Rodrigo
- Nephrology Service, University Hospital Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| |
Collapse
|
10
|
Rocchetti MT, Rascio F, Castellano G, Fiorentino M, Netti GS, Spadaccino F, Ranieri E, Gallone A, Gesualdo L, Stallone G, Pontrelli P, Grandaliano G. Altered Phosphorylation of Cytoskeleton Proteins in Peripheral Blood Mononuclear Cells Characterizes Chronic Antibody-Mediated Rejection in Kidney Transplantation. Int J Mol Sci 2020; 21:ijms21186509. [PMID: 32899575 PMCID: PMC7556000 DOI: 10.3390/ijms21186509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022] Open
Abstract
Chronic antibody-mediated rejection (CAMR) is the major cause of kidney transplant failure. The molecular mechanisms underlying this event are still poorly defined and this lack of knowledge deeply influences the potential therapeutic strategies. The aim of our study was to analyze the phosphoproteome of peripheral blood mononuclear cells (PBMCs), to identify cellular signaling networks differentially activated in CAMR. Phosphoproteins isolated from PBMCs of biopsy proven CAMR, kidney transplant recipients with normal graft function and histology and healthy immunocompetent individuals, have been investigated by proteomic analysis. Phosphoproteomic results were confirmed by Western blot and PBMCs’ confocal microscopy analyses. Overall, 38 PBMCs samples were analyzed. A differential analysis of PBMCs’ phosphoproteomes revealed an increase of lactotransferrin, actin-related protein 2 (ARPC2) and calgranulin-B in antibody-mediated rejection patients, compared to controls. Increased expression of phosphorylated ARPC2 and its correlation to F-actin filaments were confirmed in CAMR patients. Our results are the first evidence of altered cytoskeleton organization in circulating immune cells of CAMR patients. The increased expression of phosphorylated ARPC2 found in the PBMCs of our patients, and its association with derangement of F-actin filaments, might suggest that proteins regulating actin dynamics in immune cells could be involved in the mechanism of CAMR of kidney grafts.
Collapse
Affiliation(s)
- Maria Teresa Rocchetti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Federica Rascio
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Giuseppe Castellano
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, Faculty of Medicine University of Foggia, 71122 Foggia, Italy; (G.C.); (G.S.)
| | - Marco Fiorentino
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.F.); (L.G.)
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Federica Spadaccino
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Anna Gallone
- Experimental Biology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Loreto Gesualdo
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.F.); (L.G.)
| | - Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, Faculty of Medicine University of Foggia, 71122 Foggia, Italy; (G.C.); (G.S.)
| | - Paola Pontrelli
- Experimental Biology, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
- Correspondence: ; Tel.: +39-08-05-478-868
| | - Giuseppe Grandaliano
- Nephrology Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
- Department of Translational Medicine and Surgery, Faculty of Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
11
|
Recent Advances on Biomarkers of Early and Late Kidney Graft Dysfunction. Int J Mol Sci 2020; 21:ijms21155404. [PMID: 32751357 PMCID: PMC7432796 DOI: 10.3390/ijms21155404] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
New biomarkers of early and late graft dysfunction are needed in renal transplant to improve management of complications and prolong graft survival. A wide range of potential diagnostic and prognostic biomarkers, measured in different biological fluids (serum, plasma, urine) and in renal tissues, have been proposed for post-transplant delayed graft function (DGF), acute rejection (AR), and chronic allograft dysfunction (CAD). This review investigates old and new potential biomarkers for each of these clinical domains, seeking to underline their limits and strengths. OMICs technology has allowed identifying many candidate biomarkers, providing diagnostic and prognostic information at very early stages of pathological processes, such as AR. Donor-derived cell-free DNA (ddcfDNA) and extracellular vesicles (EVs) are further promising tools. Although most of these biomarkers still need to be validated in multiple independent cohorts and standardized, they are paving the way for substantial advances, such as the possibility of accurately predicting risk of DGF before graft is implanted, of making a “molecular” diagnosis of subclinical rejection even before histological lesions develop, or of dissecting etiology of CAD. Identification of “immunoquiescent” or even tolerant patients to guide minimization of immunosuppressive therapy is another area of active research. The parallel progress in imaging techniques, bioinformatics, and artificial intelligence (AI) is helping to fully exploit the wealth of information provided by biomarkers, leading to improved disease nosology of old entities such as transplant glomerulopathy. Prospective studies are needed to assess whether introduction of these new sets of biomarkers into clinical practice could actually reduce the need for renal biopsy, integrate traditional tools, and ultimately improve graft survival compared to current management.
Collapse
|
12
|
Navarrete M, Korkmaz B, Guarino C, Lesner A, Lao Y, Ho J, Nickerson P, Wilkins JA. Activity-based protein profiling guided identification of urine proteinase 3 activity in subclinical rejection after renal transplantation. Clin Proteomics 2020; 17:23. [PMID: 32549867 PMCID: PMC7296916 DOI: 10.1186/s12014-020-09284-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/19/2020] [Indexed: 03/04/2023] Open
Abstract
Background The pathophysiology of subclinical versus clinical rejection remains incompletely understood given their equivalent histological severity but discordant graft function. The goal was to evaluate serine hydrolase enzyme activities to explore if there were any underlying differences in activities during subclinical versus clinical rejection. Methods Serine hydrolase activity-based protein profiling (ABPP) was performed on the urines of a case control cohort of patients with biopsy confirmed subclinical or clinical transplant rejection. In-gel analysis and affinity purification with mass spectrometry were used to demonstrate and identify active serine hydrolase activity. An assay for proteinase 3 (PR3/PRTN3) was adapted for the quantitation of activity in urine. Results In-gel ABPP profiles suggested increased intensity and diversity of serine hydrolase activities in urine from patients undergoing subclinical versus clinical rejection. Serine hydrolases (n = 30) were identified by mass spectrometry in subclinical and clinical rejection patients with 4 non-overlapping candidates between the two groups (i.e. ABHD14B, LTF, PR3/PRTN3 and PRSS12). Western blot and the use of a specific inhibitor confirmed the presence of active PR3/PRTN3 in samples from patients undergoing subclinical rejection. Analysis of samples from normal donors or from several serial post-transplant urines indicated that although PR3/PRTN3 activity may be highly associated with low-grade subclinical inflammation, the enzyme activity was not restricted to this patient group. Conclusions There appear to be limited qualitative and quantitative differences in serine hydrolase activity in patients with subclinical versus clinical renal transplant rejection. The majority of enzymes identified were present in samples from both groups implying that in-gel quantitative differences may largely relate to the activity status of shared enzymes. However qualitative compositional differences were also observed indicating differential activities. The PR3/PRTN3 analyses indicate that the activity status of urine in transplant patients is dynamic possibly reflecting changes in the underlying processes in the transplant. These data suggest that differential serine hydrolase pathways may be active in subclinical versus clinical rejection which requires further exploration in larger patient cohorts. Although this study focused on PR3/PRTN3, this does not preclude the possibility that other enzymes may play critical roles in the rejection process.
Collapse
Affiliation(s)
- Mario Navarrete
- Manitoba Centre for Proteomics and Systems Biology, 799 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E3P4 Canada
| | - Brice Korkmaz
- INSERM, UMR 1100, "Centre d'Etude des Pathologies Respiratoires", Université de Tours, 37032 Tours, France
| | - Carla Guarino
- INSERM, UMR 1100, "Centre d'Etude des Pathologies Respiratoires", Université de Tours, 37032 Tours, France
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland
| | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, 799 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E3P4 Canada
| | - Julie Ho
- Manitoba Centre for Proteomics and Systems Biology, 799 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E3P4 Canada.,Section Biomedical Proteomics, Dept. Internal Medicine, University of Manitoba, Winnipeg, MB Canada.,Section of Nephrology, Dept. Internal Medicine, University of Manitoba, Winnipeg, MB Canada.,Dept. Immunology, University of Manitoba, Winnipeg, MB Canada
| | - Peter Nickerson
- Manitoba Centre for Proteomics and Systems Biology, 799 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E3P4 Canada.,Section Biomedical Proteomics, Dept. Internal Medicine, University of Manitoba, Winnipeg, MB Canada.,Section of Nephrology, Dept. Internal Medicine, University of Manitoba, Winnipeg, MB Canada.,Dept. Immunology, University of Manitoba, Winnipeg, MB Canada
| | - John A Wilkins
- Manitoba Centre for Proteomics and Systems Biology, 799 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E3P4 Canada.,Section Biomedical Proteomics, Dept. Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| |
Collapse
|
13
|
Thongprayoon C, Vaitla P, Craici IM, Leeaphorn N, Hansrivijit P, Salim SA, Bathini T, Cabeza Rivera FH, Cheungpasitporn W. The Use of Donor-Derived Cell-Free DNA for Assessment of Allograft Rejection and Injury Status. J Clin Med 2020; 9:E1480. [PMID: 32423115 PMCID: PMC7290747 DOI: 10.3390/jcm9051480] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
Patient monitoring after kidney transplantation (KT) for early detection of allograft rejection remains key in preventing allograft loss. Serum creatinine has poor predictive value to detect ongoing active rejection as its increase is not sensitive, nor specific for acute renal allograft rejection. Diagnosis of acute rejection requires allograft biopsy and histological assessment, which can be logistically challenging in some cases and carries inherent risk for complications related to procedure. Donor-derived cell-free DNA (dd-cfDNA), DNA of donor origin in the blood of KT recipient arising from cells undergoing injury and death, has been examined as a potential surrogate marker for allograft rejection. A rise in dd-cfDNA levels precedes changes in serum creatinine allows early detections and use as a screening tool for allograft rejection. In addition, when used in conjunction with donor-specific antibodies (DSA), it increases the pre-biopsy probability of antibody-mediated rejection (ABMR) aiding the decision-making process. Advancements in noninvasive biomarker assays such as dd-cfDNA may offer the opportunity to improve and expand the spectrum of available diagnostic tools to monitor and detect risk for rejection and positively impact outcomes for KT recipients. In this this article, we discussed the evolution of dd-cfDNA assays and recent evidence of assessment of allograft rejection and injury status of KT by the use of dd-cfDNA.
Collapse
Affiliation(s)
- Charat Thongprayoon
- Division of Nephrology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (C.T.); (I.M.C.)
| | - Pradeep Vaitla
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Iasmina M. Craici
- Division of Nephrology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (C.T.); (I.M.C.)
| | - Napat Leeaphorn
- Renal Transplant Program, University of Missouri-Kansas City School of Medicine/Saint Luke’s Health System, Kansas City, MO 64111, USA;
| | - Panupong Hansrivijit
- Department of Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, PA 17105, USA;
| | - Sohail Abdul Salim
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Tarun Bathini
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Franco H. Cabeza Rivera
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| |
Collapse
|
14
|
Hanssen O, Weekers L, Lovinfosse P, Jadoul A, Bonvoisin C, Bouquegneau A, Grosch S, Huynen A, Anglicheau D, Hustinx R, Jouret F. Diagnostic yield of 18 F-FDG PET/CT imaging and urinary CXCL9/creatinine levels in kidney allograft subclinical rejection. Am J Transplant 2020; 20:1402-1409. [PMID: 31841263 DOI: 10.1111/ajt.15742] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 01/25/2023]
Abstract
Subclinical kidney allograft acute rejection (SCR) corresponds to "the unexpected histological evidence of acute rejection in a stable patient." SCR detection relies on surveillance biopsy. Noninvasive approaches may help avoid biopsy-associated complications. From November 2015 to January 2018, we prospectively performed positron emission tomography/computed tomography (PET/CT) after injection of F18 -fluorodeoxyglucose (18 F-FDG) in adult kidney transplant recipients with surveillance biopsy at ~3 months posttransplantation. The Banff-2017 classification was used. The ratio of the mean standard uptake value (mSUVR) between kidney cortex and psoas muscle was measured. Urinary levels of CXCL-9 were concomitantly quantified. Our 92-patient cohort was categorized upon histology: normal (n = 70), borderline (n = 16), and SCR (n = 6). No clinical or biological difference was observed between groups. The mSUVR reached 1.87 ± 0.55, 1.94 ± 0.35, and 2.41 ± 0.54 in normal, borderline, and SCR groups, respectively. A significant difference in mSUVR was found among groups. Furthermore, mSUVR was significantly higher in the SCR vs normal group. The area under the receiver operating characteristic curve (AUC) was 0.79, with 83% sensitivity using an mSUVR threshold of 2.4. The AUC of urinary CXCL-9/creatinine ratios comparatively reached 0.79. The mSUVR positively correlated with ti and acute composite Banff scores. 18 F-FDG-PET/CT helps noninvasively exclude SCR, with a negative predictive value of 98%. External validations are required.
Collapse
Affiliation(s)
- Oriane Hanssen
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium
| | - Laurent Weekers
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine, Department of Medical Physics, University of Liège Hospital, Liège, Belgium
| | - Alexandre Jadoul
- Division of Nuclear Medicine, Department of Medical Physics, University of Liège Hospital, Liège, Belgium
| | - Catherine Bonvoisin
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium
| | - Antoine Bouquegneau
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium
| | - Stéphanie Grosch
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium.,Division of Renal Pathology, Unilab, University of Liège Hospital, Liège, Belgium
| | - Alexandre Huynen
- Structural Engineering Division, Faculty of Applied Sciences, University of Liège, Liège, Belgium
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, RTRS Centaure, LabEx Transplantex, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Roland Hustinx
- Division of Nuclear Medicine, Department of Medical Physics, University of Liège Hospital, Liège, Belgium
| | - Francois Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium.,Groupe Interdisciplinaire de Géno-protéomique Appliquée, Cardiovascular Sciences, University of Liège, Liège, Belgium
| |
Collapse
|
15
|
Weseslindtner L, Hedman L, Wang Y, Strassl R, Helanterä I, Aberle SW, Bond G, Hedman K. Longitudinal assessment of the CXCL10 blood and urine concentration in kidney transplant recipients with BK polyomavirus replication-a retrospective study. Transpl Int 2020; 33:555-566. [PMID: 31981424 PMCID: PMC7216881 DOI: 10.1111/tri.13584] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/03/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
In kidney transplant recipients (KTRs), BK polyomavirus (BKPyV) replication may progress to polyomavirus‐associated nephropathy (PVAN). In this retrospective study, we assessed the chemokine CXCL10 in urine and blood samples consecutively acquired from 85 KTRs who displayed different stages of BKPyV replication and eventually developed PVAN. In parallel to progression toward PVAN, CXCL10 gradually increased in blood and urine, from baseline (prior to virus replication) to BKPyV DNAuria (median increase in blood: 42.15 pg/ml, P = 0.0156), from mere DNAuria to low‐ and high‐level BKPyV DNAemia (median increase: 52.60 and 87.26 pg/ml, P = 0.0010 and P = 0.0002, respectively) and peaked with histologically confirmed PVAN (median increase: 145.00 pg/ml, P < 0.0001). CXCL10 blood and urine levels significantly differed among KTRs with respect to simultaneous presence of human cytomegalovirus (P < 0.001) as well as in relation to the clinical severity of respective BKPyV DNAemia episodes (P = 0.0195). CXCL‐10 concentrations were particularly lower in KTRs in whom BKPyV DNAemia remained without clinical evidence for PVAN, as compared to individuals who displayed high decoy cell levels, decreased renal function and/or biopsy‐proven PVAN (median blood concentration: 266.97 vs. 426.42 pg/ml, P = 0.0282). In conclusion, in KTRs CXCL10 rises in parallel to BKPyV replication and correlates with the gradual development of PVAN.
Collapse
Affiliation(s)
- Lukas Weseslindtner
- Department of Virology, University of Helsinki, Helsinki, Finland.,Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Lea Hedman
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Yilin Wang
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Robert Strassl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ilkka Helanterä
- Division of Nephrology, Transplantation and Liver Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Stephan W Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Gregor Bond
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Klaus Hedman
- Department of Virology, University of Helsinki, Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
16
|
Human leukocyte antigen molecular mismatch to risk stratify kidney transplant recipients. Curr Opin Organ Transplant 2020; 25:8-14. [DOI: 10.1097/mot.0000000000000714] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
17
|
Ho J, Sharma A, Kroeker K, Carroll R, De Serres S, Gibson IW, Hirt-Minkowski P, Jevnikar A, Kim SJ, Knoll G, Rush DN, Wiebe C, Nickerson P. Multicentre randomised controlled trial protocol of urine CXCL10 monitoring strategy in kidney transplant recipients. BMJ Open 2019; 9:e024908. [PMID: 30975673 PMCID: PMC6500325 DOI: 10.1136/bmjopen-2018-024908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Subclinical inflammation is an important predictor of death-censored graft loss, and its treatment has been shown to improve graft outcomes. Urine CXCL10 outperforms standard post-transplant surveillance in observational studies, by detecting subclinical rejection and early clinical rejection before graft functional decline in kidney transplant recipients. METHODS AND ANALYSIS This is a phase ii/iii multicentre, international randomised controlled parallel group trial to determine if the early treatment of rejection, as detected by urine CXCL10, will improve kidney allograft outcomes. Incident adult kidney transplant patients (n~420) will be enrolled to undergo routine urine CXCL10 monitoring postkidney transplant. Patients at high risk of rejection, defined as confirmed elevated urine CXCL10 level, will be randomised 1:1 stratified by centre (n=250). The intervention arm (n=125) will undergo a study biopsy to check for subclinical rejection and biopsy-proven rejection will be treated per protocol. The control arm (n=125) will undergo routine post-transplant monitoring. The primary outcome at 12 months is a composite of death-censored graft loss, clinical biopsy-proven acute rejection, de novo donor-specific antibody, inflammation in areas of interstitial fibrosis and tubular atrophy (Banff i-IFTA, chronic active T-cell mediated rejection) and subclinical tubulitis on 12-month surveillance biopsy. The secondary outcomes include decline of graft function, microvascular inflammation at 12 months, development of IFTA at 12 months, days from transplantation to clinical biopsy-proven rejection, albuminuria, EuroQol five-dimension five-level instrument, cost-effectiveness analysis of the urine CXCL10 monitoring strategy and the urine CXCL10 kinetics in response to rejection therapy. ETHICS AND DISSEMINATION The study has been approved by the University of Manitoba Health Research Ethics Board (HS20861, B2017:076) and the local research ethics boards of participating centres. Recruitment commenced in March 2018 and results are expected to be published in 2023. De-identified data may be shared with other researchers according to international guidelines (International Committee of Medical Journal Editors [ICJME]). TRIAL REGISTRATION NUMBER NCT03206801; Pre-results.
Collapse
Affiliation(s)
- Julie Ho
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
- Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Atul Sharma
- Data Science, George and Fay Yee Centre for Healthcare Innovation, Winnipeg, Manitoba, Canada
| | - Kristine Kroeker
- Data Science, George and Fay Yee Centre for Healthcare Innovation, Winnipeg, Manitoba, Canada
| | - Robert Carroll
- Transplant Nephrology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Sacha De Serres
- Internal Medicine & Nephrology, Universite Laval, Québec, Québec, Canada
| | - Ian W Gibson
- Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Anthony Jevnikar
- Internal Medicine & Nephrology, Western University, London, Ontario, Canada
| | - S Joseph Kim
- Internal Medicine & Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - Greg Knoll
- Internal Medicine & Nephrology, University of Ottawa, Ottawa, Ontario, Canada
| | - David N Rush
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| | - Chris Wiebe
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Internal Medicine, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
18
|
Barner M, DeKoning J, Kashi Z, Halloran P. Recent Advancements in the Assessment of Renal Transplant Dysfunction with an Emphasis on Microarray Molecular Diagnostics. Clin Lab Med 2018; 38:623-635. [PMID: 30420057 DOI: 10.1016/j.cll.2018.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conventional assessment of renal transplant rejection and injury through use of histology, C4d staining, and HLA antibody testing, has been the standard approach to transplant management. By many measures, these methods of conventional assessment may be considered flawed, particularly with the subjective nature of histologic diagnoses. The Alberta Transplant Applied Genomics Center has developed the Molecular Microscope diagnostic system, which uses microarrays to measure gene expression. These data are analyzed using classifiers (weighted equations) that compare the tested biopsy to a proprietary reference set of biopsies to provide objective measures of the status of the renal transplant.
Collapse
Affiliation(s)
- Meagan Barner
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA
| | - Jenefer DeKoning
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA
| | - Zahra Kashi
- Kashi Clinical Laboratories, 10101 Southwest Barbur Boulevard Suite 200, Portland, OR 97219, USA.
| | - Phillip Halloran
- Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Alberta Transplant Applied Genomics Center, 250 Heritage Medical Research Centre, Edmonton, Alberta T6G 2S2, Canada; Transcriptome Sciences Inc, Edmonton, Alberta, Canada
| |
Collapse
|
19
|
Mockler C, Sharma A, Gibson IW, Gao A, Wong A, Ho J, Blydt-Hansen TD. The prognostic value of urinary chemokines at 6 months after pediatric kidney transplantation. Pediatr Transplant 2018; 22:e13205. [PMID: 29733487 DOI: 10.1111/petr.13205] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 11/29/2022]
Abstract
Pediatric kidney transplantation is lifesaving, but long-term allograft survival is still limited by injury processes mediated by alloimmune inflammation that may otherwise be clinically silent. Chemokines associated with alloimmune inflammation may offer prognostic value early post-transplant by identifying patients at increased risk of poor graft outcomes. We conducted a single-center prospective cohort study of consecutive pediatric kidney transplant recipients (<19 years). Urinary CCL2 and CXCL10 measured at 6 months post-transplant were evaluated for association with long-term eGFR decline, allograft survival, and concomitant acute cellular rejection histology. Thirty-eight patients with a mean age of 12.4 ± 4.6 years were evaluated. Urinary CCL2 was associated with eGFR decline until 6 months (ρ -0.43; P < .01), but not at later time points. Urinary CXCL10 was associated with eGFR decline at 36 months (ρ -0.49; P < .01), risk of 50% eGFR decline (HR = 1.04; P = .02), risk of allograft loss (HR = 1.05; P = .01), borderline rejection or rejection episodes 6-12 months post-transplant (r .41; P = .02), and Banff i + t score (r .47, P < .01). CCL2 and CXCL10 were also correlated with one another (ρ 0.54; P < .01). CCL2 and CXCL10 provide differing, but complementary, information that may be useful for early non-invasive prognostic testing in pediatric kidney transplant recipients.
Collapse
Affiliation(s)
- Claire Mockler
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Atul Sharma
- Department of Pediatrics and Child Health, Children's Hospital at Health Sciences Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Ian W Gibson
- Department of Pathology, Health Sciences Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Ang Gao
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, MB, Canada
| | - Alexander Wong
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Julie Ho
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, MB, Canada.,Section of Nephrology, Department of Internal Medicine, Health Sciences Centre, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Tom D Blydt-Hansen
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Despite modern immunosuppression, renal allograft rejection remains a major contributor to graft loss. Novel biomarkers may help improve posttransplant outcomes through the early detection and treatment of rejection. Our objective is to provide an overview of proteomics, review recent discovery-based rejection studies, and explore innovative approaches in biomarker development. RECENT FINDINGS Urine MMP7 was identified as a biomarker of subclinical and clinical rejection using two-dimensional liquid chromatography tandem-mass spectrometry (LC-MS/MS) and improved the overall diagnostic discrimination of urine CXCL10 : Cr alone for renal allograft inflammation. A novel peptide signature to classify stable allografts from acute rejection, chronic allograft injury, and polyoma virus (BKV) nephropathy was identified using isobaric tag for relative and absolute quantitation (TRAQ) and label-free MS, with independent validation by selected reaction monitoring mass spectrometry (SRM-MS). Finally, an in-depth exploration of peripheral blood mononuclear cells identified differential proteoform expression in healthy transplants versus rejection. SUMMARY There is still much in the human proteome that remains to be explored, and further integration of renal, urinary, and exosomal data may offer deeper insight into the pathophysiology of rejection. Functional proteomics may be more biologically relevant than protein/peptide quantity alone, such as assessment of proteoforms or activity-based protein profiling. Discovery-based studies have identified potential biomarker candidates, but external validation studies are required.
Collapse
|
21
|
Choi N, Rigatto C, Zappitelli M, Gao A, Christie S, Hiebert B, Arora RC, Ho J. Urinary Hepcidin-25 Is Elevated in Patients That Avoid Acute Kidney Injury Following Cardiac Surgery. Can J Kidney Health Dis 2018; 5:2054358117744224. [PMID: 29399365 PMCID: PMC5788097 DOI: 10.1177/2054358117744224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Acute kidney injury (AKI) following cardiac surgery leads to increased morbidity and mortality. Characterization and validation of early biomarkers of AKI may ultimately facilitate early therapeutic intervention. We have previously identified that elevated urinary hepcidin-25 is inversely and independently associated with the development of AKI in adult cardiac surgery patients. Hepcidin-25 is an antimicrobial peptide that sequesters iron intracellularly, and its elevation following human ischemia reperfusion injury may represent a renoprotective response to minimize renal injury. Objective: Our goal was to validate urinary hepcidin-25 as a non-invasive biomarker in an independent cardiac surgery cohort, within the context of clinical AKI predictors. Design: Prospective observational cohort study. Setting: Adult cardiac surgery program at St. Boniface Hospital, Winnipeg, Manitoba, Canada. Patients: Adult cardiac surgery patients undergoing cardiopulmonary bypass (CPB), n = 306. Measurements: Urine hepcidin-25, measured on post-operative day (POD) 1. Methods: A prospective, observational cohort of adult CPB patients (n = 306) was collected with serial perioperative urine samples. Urine hepcidin-25 at POD 1 was measured by competitive ELISA. Its diagnostic performance was evaluated in conjunction with clinical parameters and the Thakar clinical prediction score, using multivariate logistic regression. Results: Urinary hepcidin-25 is elevated following cardiac surgery in AKI and non-AKI patients. Elevated urinary hepcidin-25 concentration was inversely associated with AKI on both univariate (odds ratio [OR]: 0.61, 95% confidence interval [CI]: 0.45-0.83, P = .002) and multivariate analysis (OR: 0.67, 95% CI: 0.50-0.95, P = .02). A combined model with clinical risk factors demonstrated that baseline estimated glomerular filtration rate (eGFR), diabetes mellitus, and urinary hepcidin-25 concentration had an overall area under the curve (AUC) of 0.82 (0.75-0.88) for predicting subsequent AKI development, which was superior to clinical prediction alone as determined by the Thakar score. Limitations: (1) A single-center observational study. (2) Polyclonal antibody–based competitive ELISA. Conclusion: Hepcidin-25 is inversely associated with AKI in a multivariate model when combined with eGFR and diabetes mellitus, with an overall AUC of 0.82. Notably, urinary hepcidin-25 improves on clinical AKI prediction compared to the Thakar score alone.
Collapse
Affiliation(s)
- Nora Choi
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada.,Department of Immunology, University of Manitoba, Winnipeg, Canada.,Section of Cardiac Sciences, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Claudio Rigatto
- Department of Internal Medicine, Section of Nephrology, University of Manitoba, Winnipeg, Canada
| | - Michael Zappitelli
- Department of Pediatrics, Division of Nephrology, McGill University, Montréal, Quebec, Canada
| | - Ang Gao
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| | - Simon Christie
- Section of Cardiac Sciences, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Brett Hiebert
- Section of Cardiac Sciences, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Rakesh C Arora
- Section of Cardiac Sciences, St. Boniface Hospital, Winnipeg, Manitoba, Canada.,Department of Surgery, University of Manitoba, Winnipeg, Canada
| | - Julie Ho
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada.,Department of Immunology, University of Manitoba, Winnipeg, Canada.,Department of Internal Medicine, Section of Nephrology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
22
|
Salcido-Ochoa F, Allen Jr JC. Biomarkers and a tailored approach for immune monitoring in kidney transplantation. World J Transplant 2017; 7:276-284. [PMID: 29312857 PMCID: PMC5743865 DOI: 10.5500/wjt.v7.i6.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/29/2017] [Accepted: 11/22/2017] [Indexed: 02/05/2023] Open
Abstract
A literature review on immune monitoring in kidney transplantation produced dozens of research articles and a multitude of promising biomarkers, all in the quest for the much sought after - but perennially elusive - “holy grail” of kidney biomarkers able to unequivocally predict acute transplant rejection vs non-rejection. Detection methodologies and study designs were many and varied. Hence the motivation for this editorial, which espouses the notion that in today’s kidney transplantation milieu, the judicious use of disease classifiers tailored to specific patient immune risks may be more achievable and productive in the long run and confer a greater advantage for patient treatment than the pursuit of a single “omniscient” biomarker. In addition, we desire to direct attention toward greater scrutiny of biomarker publications and decisions to implement biomarkers in practice, standardization of methods in the development of biomarkers and consideration for adoption of “biomarker-driven” biopsies. We propose “biomarker-driven” biopsies as an adjunctive to and/or alternative to random surveillance (protocol) biopsies or belated indication biopsies. The discovery of a single kidney transplantation biomarker would represent a major breakthrough in kidney transplantation practice, but until that occurs - if ever it does occur, other approaches offer substantial potential for unlocking prognostic, diagnostic and therapeutic options. We conclude our editorial with suggestions and recommendations for productively incorporating current biomarkers into diagnostic algorithms and for testing future biomarkers of acute rejection in kidney transplantation.
Collapse
Affiliation(s)
- Francisco Salcido-Ochoa
- Tregs and HLA Research Force, Francisco Kidney and Medical Centre, Mount Elizabeth Novena Hospital, Singapore 329563, Singapore
| | - John Carson Allen Jr
- Centre for Quantitative Medicine, Duke-NUS Graduate Medical School, Singapore 169856, Singapore
| |
Collapse
|
23
|
Salvadori M, Tsalouchos A. Biomarkers in renal transplantation: An updated review. World J Transplant 2017; 7:161-178. [PMID: 28698834 PMCID: PMC5487307 DOI: 10.5500/wjt.v7.i3.161] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/07/2017] [Accepted: 04/18/2017] [Indexed: 02/05/2023] Open
Abstract
Genomics, proteomics and molecular biology lead to tremendous advances in all fields of medical sciences. Among these the finding of biomarkers as non invasive indicators of biologic processes represents a useful tool in the field of transplantation. In addition to define the principal characteristics of the biomarkers, this review will examine the biomarker usefulness in the different clinical phases following renal transplantation. Biomarkers of ischemia-reperfusion injury and of delayed graft function are extremely important for an early diagnosis of these complications and for optimizing the treatment. Biomarkers predicting or diagnosing acute rejection either cell-mediated or antibody-mediated allow a risk stratification of the recipient, a prompt diagnosis in an early phase when the histology is still unremarkable. The kidney solid organ response test detects renal transplant recipients at high risk for acute rejection with a very high sensitivity and is also able to make diagnosis of subclinical acute rejection. Other biomarkers are able to detect chronic allograft dysfunction in an early phase and to differentiate the true chronic rejection from other forms of chronic allograft nephropathies no immune related. Finally biomarkers recently discovered identify patients tolerant or almost tolerant. This fact allows to safely reduce or withdrawn the immunosuppressive therapy.
Collapse
|
24
|
Establishing Biomarkers in Transplant Medicine: A Critical Review of Current Approaches. Transplantation 2017; 100:2024-38. [PMID: 27479159 DOI: 10.1097/tp.0000000000001321] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the management of kidney transplant recipients has greatly improved over recent decades, the assessment of individual risks remains highly imperfect. Individualized strategies are necessary to recognize and prevent immune complications early and to fine-tune immunosuppression, with the overall goal to improve patient and graft outcomes. This review discusses current biomarkers and their limitations, and recent advancements in the field of noninvasive biomarker discovery. A wealth of noninvasive monitoring tools has been suggested that use easily accessible biological fluids such as urine and blood, allowing frequent and sequential assessments of recipient's immune status. This includes functional cell-based assays and the evaluation of molecular expression on a wide spectrum of platforms. Nevertheless, the translation and validation of exploratory findings and their implementation into standard clinical practice remain challenging. This requires dedicated prospective interventional trials demonstrating that the use of these biomarkers avoids invasive procedures and improves patient or transplant outcomes.
Collapse
|
25
|
Raza A, Firasat S, Khaliq S, Khan AR, Mahmood S, Aziz T, Mubarak M, Naqvi SAA, Rizvi SAH, Abid A. Monocyte Chemoattractant Protein-1 (MCP-1/CCL2) Levels and Its Association with Renal Allograft Rejection. Immunol Invest 2017; 46:251-262. [PMID: 27960564 DOI: 10.1080/08820139.2016.1248559] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND CCL2 is a chemoattractant for monocytes/macrophages, T cells, and natural killer cells. It is shown to be involved in the immunological responses against renal allograft. This study was conducted to access the role of urinary CCL2 expression in predicting the rejection episodes in renal transplant patients. METHOD A total of 409 urine samples included in this study. The samples were consisted of (a) biopsy-proven graft rejection (n = 165); (b) non-rejection (n = 93); (c) non-biopsy stable-graft (n = 42), and (d) healthy renal donors (n = 109). The samples were quantified for the CCL2 using the MCP-1/CCL2 ELISA kit. The data were analyzed using the Statistical Package for Social Sciences (SPSS®) and MedCalc® statistical software. RESULTS Results showed that the CCL2 levels were significantly increased in rejection group when compared with the non-rejection, stable-graft, and control, P < 0.05. The receiver operating curve's characteristics illustrated that the urinary CCL2 level is a good predictor for graft rejection, with an area under the curve of 0.81 ± 0.03 with optimum sensitivity and specificity of 87% and 62%, respectively, at a cut-off value of 198 pg/mL. Kaplan-Meier curve also showed better cumulative rejection-free graft survival time in group with less than 198 pg/mL of CCL2 as compared to those with expression levels of more than 198 pg/mL (30 weeks vs. 3 weeks; log-rank test, P < 0.001). CONCLUSION In our study, noninvasive investigation of CCL2 levels in urine has showed potential to predict rejection episodes. It is suggested that the CCL2, with others markers, may help in early detection and monitoring of graft rejection episodes.
Collapse
Affiliation(s)
- Ali Raza
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Sadaf Firasat
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Shagufta Khaliq
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
- b Department of Human Genetics , University of Health Sciences , Lahore , Pakistan
| | - Abdul Rafay Khan
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Shafaq Mahmood
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Tahir Aziz
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Muhammad Mubarak
- d Department of Histopathology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Syed Ali Anwar Naqvi
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Syed Adibul Hasan Rizvi
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Aiysha Abid
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| |
Collapse
|
26
|
Maier M, Takano T, Sapir-Pichhadze R. Changing Paradigms in the Management of Rejection in Kidney Transplantation: Evolving From Protocol-Based Care to the Era of P4 Medicine. Can J Kidney Health Dis 2017; 4:2054358116688227. [PMID: 28270929 PMCID: PMC5308536 DOI: 10.1177/2054358116688227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/17/2016] [Indexed: 12/30/2022] Open
Abstract
PURPOSE OF REVIEW P4 medicine denotes an evolving field of medicine encompassing predictive, preventive, personalized, and participatory medicine. Using the example of kidney allograft rejection because of donor-recipient incompatibility in human leukocyte antigens, this review outlines P4 medicine's relevance to the various stages of the kidney transplant cycle. SOURCES OF INFORMATION A search for English articles was conducted in Medline via OvidSP (up to August 18, 2016) using a combination of subject headings (MeSH) and free text in titles, abstracts, and author keywords for the concepts kidney transplantation and P4 medicine. The electronic database search was expanded further on particular subject headings. FINDINGS Available histocompatibility methods exemplify current applications of the predictive and preventive domains of P4 medicine in kidney transplant recipients' care. Pharmacogenomics are discussed as means to facilitate personalized immunosuppression regimens and promotion of active patient participation as a means to improve adherence. LIMITATIONS For simplicity, this review focuses on rejection. P4 medicine, however, should more broadly address health concerns in kidney transplant recipients, including competing outcomes such as infections, malignancies, and cardiovascular disease. This review highlights how biomarkers to evaluate these competing outcomes warrant validation and standardization prior to their incorporation into clinical practice. IMPLICATIONS Consideration of all 4 domains of the P4 medicine framework when caring for and/or studying kidney transplant recipients has the potential of increasing therapeutic efficiency, minimizing adverse effects, decreasing health care costs, and maximizing wellness. Technologies to gauge immune competency, immunosuppression requirements, and early/reversible immune-mediated injuries are required to optimize kidney transplant care.
Collapse
Affiliation(s)
- Mirela Maier
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Metabolic Disorders and Complications, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | - Tomoko Takano
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Metabolic Disorders and Complications, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | - Ruth Sapir-Pichhadze
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Metabolic Disorders and Complications, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
- Multi-Organ Transplant Program, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Centre for Outcomes Research and Evaluation, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Matz M, Lorkowski C, Fabritius K, Wu K, Rudolph B, Frischbutter S, Brakemeier S, Gaedeke J, Neumayer HH, Mashreghi MF, Budde K. The selective biomarker IL-8 identifies IFTA after kidney transplantation in blood cells. Transpl Immunol 2016; 39:18-24. [DOI: 10.1016/j.trim.2016.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 12/20/2022]
|
28
|
Abstract
INTRODUCTION Identification of allograft injury, including acute clinical and subclinical injury, is vital in increasing the longevity of the transplanted organ. Acute rejection, which occurs as a result of a variety of immune and non-immune factors including the infiltration of immune cells and antibodies to the donor specific epitopes, poses a significant risk to the organ. Recent years have marked an increase in the discovery of new genomic, transcriptomic, and proteomic biomarkers in molecular diagnostics, which offer better potential for personalized management of the transplanted organ by providing earlier detection of rejection episodes. Areas covered: This review was compiled from key word searches of full-text publications relevant to the field. Expert commentary: Many of the recent advancements in the molecular diagnostics of allograft injury show much promise, but before they can be fully realized further validation in larger sample sets must be conducted. Additionally, for better informed therapeutic decisions, more work must be completed to differentiate between different causes of injury. Moreover, the diagnostics field is looking at methodologies that allow for multiplexing, the ability to identify multiple targets simultaneously, in order to provide more robust biomarkers and better understanding.
Collapse
Affiliation(s)
- Michael Nasr
- Sarwal Lab, University of California, San Francisco
- University of California, San Francisco, Department of Bioengineering & Therapeutic Sciences
- University of California, Berkeley, Department of Bioengineering
| | - Tara Sigdel
- Sarwal Lab, University of California, San Francisco
- Unversity of California, San Francisco Department of Surgery
| | - Minnie Sarwal
- Sarwal Lab, University of California, San Francisco
- Unversity of California, San Francisco Department of Surgery
| |
Collapse
|
29
|
Erpicum P, Hanssen O, Weekers L, Lovinfosse P, Meunier P, Tshibanda L, Krzesinski JM, Hustinx R, Jouret F. Non-invasive approaches in the diagnosis of acute rejection in kidney transplant recipients, part II: omics analyses of urine and blood samples. Clin Kidney J 2016. [PMID: 28643819 PMCID: PMC5469577 DOI: 10.1093/ckj/sfw077] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidney transplantation (KTx) represents the best available treatment for patients with end-stage renal disease. Still, the full benefits of KTx are undermined by acute rejection (AR). The diagnosis of AR ultimately relies on transplant needle biopsy. However, such an invasive procedure is associated with a significant risk of complications and is limited by sampling error and interobserver variability. In the present review, we summarize the current literature about non-invasive approaches for the diagnosis of AR in kidney transplant recipients (KTRs), including in vivo imaging, gene-expression profiling and omics analyses of blood and urine samples. Most imaging techniques, such as contrast-enhanced ultrasound and magnetic resonance, exploit the fact that blood flow is significantly lowered in case of AR-induced inflammation. In addition, AR-associated recruitment of activated leucocytes may be detectable by 18F-fluorodeoxyglucose positron emission tomography. In parallel, urine biomarkers, including CXCL9/CXCL10 or a three-gene signature of CD3ε, CXCL10 and 18S RNA levels, have been identified. None of these approaches has yet been adopted in the clinical follow-up of KTRs, but standardization of analysis procedures may help assess reproducibility and comparative diagnostic yield in large, prospective, multicentre trials.
Collapse
Affiliation(s)
- Pauline Erpicum
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, Université de Liège, Liège, Belgium
| | - Oriane Hanssen
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), B-4000 Liège, Belgium
| | - Laurent Weekers
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), B-4000 Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Paul Meunier
- Division of Radiology, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Luaba Tshibanda
- Division of Radiology, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Jean-Marie Krzesinski
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, Université de Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - François Jouret
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, Université de Liège, Liège, Belgium
| |
Collapse
|
30
|
Hanssen O, Erpicum P, Lovinfosse P, Meunier P, Weekers L, Tshibanda L, Krzesinski JM, Hustinx R, Jouret F. Non-invasive approaches in the diagnosis of acute rejection in kidney transplant recipients. Part I. In vivo imaging methods. Clin Kidney J 2016. [PMID: 28643821 PMCID: PMC5469561 DOI: 10.1093/ckj/sfw062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Kidney transplantation (KTx) represents the best available treatment for patients with end-stage renal disease. Still, full benefits of KTx are undermined by acute rejection (AR). The diagnosis of AR ultimately relies on transplant needle biopsy. However, such an invasive procedure is associated with a significant risk of complications and is limited by sampling error and interobserver variability. In the present review, we summarize the current literature about non-invasive approaches for the diagnosis of AR in kidney transplant recipients (KTRs), including in vivo imaging, gene expression profiling and omics analyses of blood and urine samples. Most imaging techniques, like contrast-enhanced ultrasound and magnetic resonance, exploit the fact that blood flow is significantly lowered in case of AR-induced inflammation. In addition, AR-associated recruitment of activated leukocytes may be detectable by 18F-fluoro-deoxy-glucose positron emission tomography. In parallel, urine biomarkers, including CXCL9/CXCL10 or a three-gene signature of CD3ε, IP-10 and 18S RNA levels, have been identified. None of these approaches has been adopted yet in the clinical follow-up of KTRs, but standardization of procedures may help assess reproducibility and compare diagnostic yields in large prospective multicentric trials.
Collapse
Affiliation(s)
- Oriane Hanssen
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), Avenue Hippocrate, 13, B-4000 Liège, Belgium
| | - Pauline Erpicum
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), Avenue Hippocrate, 13, B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Paul Meunier
- Division of Radiology, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Laurent Weekers
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), Avenue Hippocrate, 13, B-4000 Liège, Belgium
| | - Luaba Tshibanda
- Division of Radiology, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - Jean-Marie Krzesinski
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), Avenue Hippocrate, 13, B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine, University of Liège Academic Hospital (ULg CHU), Liège, Belgium
| | - François Jouret
- Division of Nephrology, University of Liège Academic Hospital (ULg CHU), Avenue Hippocrate, 13, B-4000 Liège, Belgium.,GIGA Cardiovascular Sciences, University of Liège, Liège, Belgium
| |
Collapse
|