1
|
Lendermon EA, Hage CA. Pulmonary Immunocompromise in Solid Organ Transplantation. Clin Chest Med 2025; 46:149-158. [PMID: 39890285 DOI: 10.1016/j.ccm.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
This article reviews the multitude of factors contributing to immune dysfunction and pulmonary infection risk in solid organ transplant recipients and references relevant clinical scientific reports. The mechanisms of action of individual immunosuppressive agents are explained, and the clinical effects of these drugs are compared. In addition, specialized methods to assess the net state of immunosuppression in individual transplant recipients and their limitations are discussed.
Collapse
Affiliation(s)
- Elizabeth A Lendermon
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh Medical Center, 3459 Fifth Avenue, MUH NW 628, Pittsburgh, PA 15213, USA
| | - Chadi A Hage
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh Medical Center, 3459 Fifth Avenue, MUH NW 628, Pittsburgh, PA 15213, USA; Lung Transplant, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite C-901, Pittsburgh, PA 15213, USA.
| |
Collapse
|
2
|
Tharmaraj D, Mulley WR, Dendle C. Current and emerging tools for simultaneous assessment of infection and rejection risk in transplantation. Front Immunol 2024; 15:1490472. [PMID: 39660122 PMCID: PMC11628869 DOI: 10.3389/fimmu.2024.1490472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Infection and rejection are major complications that impact transplant longevity and recipient survival. Balancing their risks is a significant challenge for clinicians. Current strategies aimed at interrogating the degree of immune deficiency or activation and their attendant risks of infection and rejection are imprecise. These include immune (cell counts, function and subsets, immunoglobulin levels) and non-immune (drug levels, viral loads) markers. The shared risk factors between infection and rejection and the bidirectional and intricate relationship between both entities further complicate transplant recipient care and decision-making. Understanding the dynamic changes in the underlying net state of immunity and the overall risk of both complications in parallel is key to optimizing outcomes. The allograft biopsy is the current gold standard for the diagnosis of rejection but is associated with inherent risks that warrant careful consideration. Several biomarkers, in particular, donor derived cell-free-DNA and urinary chemokines (CXCL9 and CXCL10), show significant promise in improving subclinical and clinical rejection risk prediction, which may reduce the need for allograft biopsies in some situations. Integrating conventional and emerging risk assessment tools can help stratify the individual's short- and longer-term infection and rejection risks in parallel. Individuals identified as having a low risk of rejection may tolerate immunosuppression wean to reduce medication-related toxicity. Serial monitoring following immunosuppression reduction or escalation with minimally invasive tools can help mitigate infection and rejection risks and allow for timely diagnosis and treatment of these complications, ultimately improving allograft and patient outcomes.
Collapse
Affiliation(s)
- Dhakshayini Tharmaraj
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - William R. Mulley
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Claire Dendle
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
- Monash Infectious Diseases, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
3
|
Pan X, Peng J, Zhu R, An N, Pei J. Non-invasive biomarkers of acute rejection in pediatric kidney transplantation: New targets and strategies. Life Sci 2024; 348:122698. [PMID: 38710278 DOI: 10.1016/j.lfs.2024.122698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Kidney transplantation is the preferred treatment for pediatric end-stage renal disease. However, pediatric recipients face unique challenges due to their prolonged need for kidney function to accommodate growth and development. The continual changes in the immune microenvironment during childhood development and the heightened risk of complications from long-term use of immunosuppressive drugs. The overwhelming majority of children may require more than one kidney transplant in their lifetime. Acute rejection (AR) stands as the primary cause of kidney transplant failure in children. While pathologic biopsy remains the "gold standard" for diagnosing renal rejection, its invasive nature raises concerns regarding potential functional impairment and the psychological impact on children due to repeated procedures. In this review, we outline the current research status of novel biomarkers associated with AR in urine and blood after pediatric kidney transplantation. These biomarkers exhibit superior diagnostic and prognostic performance compared to conventional ones, with the added advantages of being less invasive and highly reproducible for long-term graft monitoring. We also integrate the limitations of these novel biomarkers and propose a refined monitoring model to optimize the management of AR in pediatric kidney transplantation.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jinpu Peng
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Rong Zhu
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Nini An
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jun Pei
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| |
Collapse
|
4
|
Gaiffe E, Colladant M, Desmaret M, Bamoulid J, Leroux F, Laheurte C, Brouard S, Giral M, Saas P, Courivaud C, Degauque N, Ducloux D. Pre-transplant immune profile defined by principal component analysis predicts acute rejection after kidney transplantation. Front Immunol 2023; 14:1192440. [PMID: 37497224 PMCID: PMC10367005 DOI: 10.3389/fimmu.2023.1192440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Background Acute rejection persists as a frequent complication after kidney transplantation. Defining an at-risk immune profile would allow better preventive approaches. Methods We performed unsupervised hierarchical clustering analysis on pre-transplant immunological phenotype in 1113 renal transplant recipients from the ORLY-EST cohort. Results We identified three immune profiles correlated with clinical phenotypes. A memory immune cluster was defined by memory CD4+T cell expansion and decreased naïve CD4+T cell. An activated immune cluster was characterized by an increase in CD8+T cells and a decreased CD4/CD8 ratio. A naïve immune cluster was mainly defined by increased naïve CD4+T cells. Patients from the memory immune profile tend to be older and to have diabetes whereas those from the activated immune profile were younger and more likely to have pre-transplant exposure to CMV. Patients from the activated immune profile were more prone to experience acute rejection than those from other clusters [(HR=1.69, 95%IC[1.05-2.70], p=0.030) and (HR=1.85; 95%IC[1.16-3.00], p=0.011). In the activated immune profile, those without previous exposure to CMV (24%) were at very high risk of acute rejection (27 vs 16%, HR=1.85; 95%IC[1.04-3.33], p=0.039). Conclusion Immune profile determination based on principal component analysis defines clinically different sub-groups and discriminate a population at high-risk of acute rejection.
Collapse
Affiliation(s)
- Emilie Gaiffe
- Besançon University Hospital, INSERM CIC-1431, Besançon, France
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Mathilde Colladant
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Besançon University Hospital, Department of Nephrology, Besançon, France
| | - Maxime Desmaret
- Besançon University Hospital, INSERM CIC-1431, Besançon, France
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Jamal Bamoulid
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Besançon University Hospital, Department of Nephrology, Besançon, France
| | - Franck Leroux
- Besançon University Hospital, INSERM CIC-1431, Besançon, France
| | - Caroline Laheurte
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Sophie Brouard
- Centre Hospitalier Universitaire (CHU) Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Université de Nantes (ITUN), Nantes, France
| | - Magali Giral
- Centre Hospitalier Universitaire (CHU) Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Université de Nantes (ITUN), Nantes, France
| | - Philippe Saas
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Cécile Courivaud
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Besançon University Hospital, Department of Nephrology, Besançon, France
| | - Nicolas Degauque
- Centre Hospitalier Universitaire (CHU) Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, Unité Mixte de Recherche (UMR) 1064, Institut de Transplantation Université de Nantes (ITUN), Nantes, France
| | - Didier Ducloux
- Besançon University Hospital, INSERM CIC-1431, Besançon, France
- Univ. Franche-Comté, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, Unité Mixte de Recherche (UMR) 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Besançon University Hospital, Department of Nephrology, Besançon, France
| |
Collapse
|
5
|
Sparding N, Genovese F, Rasmussen DGK, Karsdal MA, Krogstrup NV, Nielsen MB, Hornum M, Nagarajah S, Birn H, Jespersen B, Tepel M, Nørregaard R. Endotrophin Levels Are Associated with Allograft Outcomes in Kidney Transplant Recipients. Biomolecules 2023; 13:biom13050792. [PMID: 37238662 DOI: 10.3390/biom13050792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/29/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Early prediction of kidney graft function may assist clinical management, and for this, reliable non-invasive biomarkers are needed. We evaluated endotrophin (ETP), a novel non-invasive biomarker of collagen type VI formation, as a prognostic marker in kidney transplant recipients. ETP levels were measured with the PRO-C6 ELISA in the plasma (P-ETP) of 218 and urine (U-ETP/Cr) of 172 kidney transplant recipients, one (D1) and five (D5) days, as well as three (M3) and twelve (M12) months, after transplantation. P-ETP and U-ETP/Cr at D1 (P-ETP AUC = 0.86, p < 0.0001; U-ETP/Cr AUC = 0.70, p = 0.0002) were independent markers of delayed graft function (DGF) and P-ETP at D1 had an odds ratio of 6.3 (p < 0.0001) for DGF when adjusted for plasma creatinine. The results for P-ETP at D1 were confirmed in a validation cohort of 146 transplant recipients (AUC = 0.92, p < 0.0001). U-ETP/Cr at M3 was negatively associated with kidney graft function at M12 (p = 0.007). This study suggests that ETP at D1 can identify patients at risk of delayed graft function and that U-ETP/Cr at M3 can predict the future status of the allograft. Thus, measuring collagen type VI formation could aid in predicting graft function in kidney transplant recipients.
Collapse
Affiliation(s)
- Nadja Sparding
- Nordic Bioscience, 2730 Herlev, Denmark
- Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | | | | | | | - Marie Bodilsen Nielsen
- Department of Renal Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Subagini Nagarajah
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik Birn
- Department of Renal Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Bente Jespersen
- Department of Renal Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Martin Tepel
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
6
|
Tepel M, Nagarajah S, Saleh Q, Thaunat O, Bakker SJL, van den Born J, Karsdal MA, Genovese F, Rasmussen DGK. Pretransplant characteristics of kidney transplant recipients that predict posttransplant outcome. Front Immunol 2022; 13:945288. [PMID: 35958571 PMCID: PMC9357871 DOI: 10.3389/fimmu.2022.945288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Better characterization of the potential kidney transplant recipient using novel biomarkers, for example, pretransplant plasma endotrophin, will lead to improved outcome after transplantation. This mini-review will focus on current knowledge about pretransplant recipients’ characteristics, biomarkers, and immunology. Clinical characteristics of recipients including age, obesity, blood pressure, comorbidities, and estimated survival scores have been introduced for prediction of recipient and allograft survival. The pretransplant immunologic risk assessment include histocompatibility leukocyte antigens (HLAs), anti-HLA donor-specific antibodies, HLA-DQ mismatch, and non-HLA antibodies. Recently, there has been the hope that pretransplant determination of markers can further improve the prediction of posttransplant complications, both short-term and long-term outcomes including rejections, allograft loss, and mortality. Higher pretransplant plasma endotrophin levels were independently associated with posttransplant acute allograft injury in three prospective European cohorts. Elevated numbers of non-synonymous single-nucleotide polymorphism mismatch have been associated with increased allograft loss in a multivariable analysis. It is concluded that there is a need for integration of clinical characteristics and novel molecular and immunological markers to improve future transplant medicine to reach better diagnostic decisions tailored to the individual patient.
Collapse
Affiliation(s)
- Martin Tepel
- Department of Nephrology, Odense University Hospital, Odense, Denmark, and Cardiovascular and Renal Research, Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, Odense, Denmark
- *Correspondence: Martin Tepel,
| | - Subagini Nagarajah
- Department of Nephrology, Odense University Hospital, Odense, Denmark, and Cardiovascular and Renal Research, Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Qais Saleh
- Department of Nephrology, Odense University Hospital, Odense, Denmark, and Cardiovascular and Renal Research, Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Olivier Thaunat
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique, Lyon, France
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | | |
Collapse
|
7
|
Yatim KM, Azzi JR. Novel Biomarkers in Kidney Transplantation. Semin Nephrol 2022; 42:2-13. [DOI: 10.1016/j.semnephrol.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
8
|
Roberts MB, Fishman JA. Immunosuppressive Agents and Infectious Risk in Transplantation: Managing the "Net State of Immunosuppression". Clin Infect Dis 2020; 73:e1302-e1317. [PMID: 32803228 DOI: 10.1093/cid/ciaa1189] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Successful solid organ transplantation reflects meticulous attention to the details of immunosuppression, balancing risks for graft rejection against risks for infection. The 'net state of immune suppression' is a conceptual framework of all factors contributing to infectious risk. Assays which measure immune function in the immunosuppressed transplant recipient relative to infectious risk and allograft function are lacking. The best measures of integrated immune function may be quantitative viral loads to assess the individual's ability to control latent viral infections. Few studies address adjustment of immunosuppression during active infections. Thus, confronted with infection in solid organ recipients, the management of immunosuppression is based largely on clinical experience. This review examines known measures of immune function and the immunologic effects of common immunosuppressive drugs and available studies reporting modification of drug regimens for specific infections. These data provide a conceptual framework for the management of immunosuppression during infection in organ recipients.
Collapse
Affiliation(s)
- Matthew B Roberts
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston MA
| | - Jay A Fishman
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Fernández-Ruiz M, López-Medrano F, Aguado JM. Predictive tools to determine risk of infection in kidney transplant recipients. Expert Rev Anti Infect Ther 2020; 18:423-441. [PMID: 32084326 DOI: 10.1080/14787210.2020.1733976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Infection represents a major complication after kidney transplantation (KT). Therapeutic drug monitoring is essentially the only approach for the adjustment of immunosuppression in current practice, with suboptimal results. The implementation of immune monitoring strategies may contribute to minimizing the risk of adverse events attributable to over-immunosuppression without compromising graft outcomes.Areas covered: The present review (based on PubMed/MEDLINE searches from database inception to November 2019) is focused on immune biomarkers with no antigen specificity (non-pathogen-specific), including serum levels of immunoglobulins and complement factors, peripheral blood lymphocyte subpopulations, soluble CD30, intracellular ATP production by stimulated CD4+ T-cells, and other cell-based immune assays. We also summarized recent advances in the use of replication kinetics of latent viruses to assess the functionality of T-cell immunity, with focus on the nonpathogenic anelloviruses. Finally, the composite risk scores reported in the literature are critically discussed.Expert opinion: Notable efforts have been made to develop an enlarging repertoire of immune biomarkers and prediction models, although most of them still lack technical standardization and external validation. Preventive interventions based on these tools (prolongation of prophylaxis, tapering of immunosuppression, or immunoglobulin replacement therapy in hypogammaglobulinemic patients) remain to be defined, ideally in the context of controlled trials.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (Imas12), Madrid, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0002), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (Imas12), Madrid, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0002), Instituto de Salud Carlos III, Madrid, Spain.,School of Medicine, Universidad Complutense, Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (Imas12), Madrid, Spain.,Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0002), Instituto de Salud Carlos III, Madrid, Spain.,School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
10
|
Soluble CD30, Acute Rejection, and Graft Survival: Pre- and 6-Month Post-Transplant Determinations-When Is the Best Time to Measure? Transplant Proc 2018; 50:728-736. [PMID: 29661425 DOI: 10.1016/j.transproceed.2018.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Pretransplantation soluble CD30 (sCD30) has been shown to be a good predictor of acute rejection (AR) and graft loss. This study aimed to evaluate the effectiveness of sCD30 measured pretransplant and up to 6 months after transplantation as a predictor of AR, graft loss, and survival at 5 years post-transplantation. Subjects were patients receiving living donor renal transplants at Bonsucesso Federal Hospital (Rio de Janeiro) in 2006 and between August 2010 and May 2011. METHODS sCD30 was analyzed in samples collected pretransplantation and 7, 14, and 21, 28 days and 3, 4, 5, and 6 months post-transplantation from 73 kidney recipients. RESULTS Patients in the AR group did not present a positive correlation with the sCD30 levels pretransplant (P = .54); in the post-transplant period, the 7- to 14-day samples showed patients with AR had higher levels of this biomarker (P = .036). The graft survival in 5 years of follow-up was not different between groups. CONCLUSIONS The best time to predict AR using sCD30 is the 7- to 14-day sample; however, identifying and following the decrease of this biomarker from pre- to post-transplant seems to be better than just 1 measurement. The sCD30 post-transplant is another tool that may be used in monitoring patients after renal transplantation.
Collapse
|
11
|
Abstract
Detecting acute rejection in kidney transplantation has been traditionally done using histological analysis of invasive allograft biopsies, but this method carries a risk and is not perfect. Transplant professionals have been working to develop more accurate or less invasive biomarkers that can predict acute rejection or subsequent worse allograft survival. These biomarkers can use tissue, blood or urine as a source. They can comprise individual molecules or panels, singly or in combination, across different components or pathways of the immune system. This review highlights the most recent evidence for biomarker efficacy, especially from multicenter trials.
Collapse
Affiliation(s)
- Vikas R Dharnidharka
- Division of Pediatric Nephrology, Hypertension and Pheresis, Washington University in St Louis & St. Louis Children's Hospital, St Louis, MO, USA.
| | - Andrew Malone
- Division of Nephrology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
12
|
Affiliation(s)
- Arjun Chakraborty
- Department of Surgery, University of California San Francisco, San Francisco, USA
| | - Minnie Sarwal
- Director of Precision Transplant Medicine, University of California San Francisco, San Francisco, USA
| |
Collapse
|
13
|
Trailin AV, Ostapenko TI, Nykonenko TN, Nesterenko SN, Nykonenko OS. Peritransplant Soluble CD30 as a Risk Factor for Slow Kidney Allograft Function, Early Acute Rejection, Worse Long-Term Allograft Function, and Patients' Survival. DISEASE MARKERS 2017; 2017:9264904. [PMID: 28694560 PMCID: PMC5485490 DOI: 10.1155/2017/9264904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/27/2017] [Accepted: 04/11/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND We aimed to determine whether serum soluble CD30 (sCD30) could identify recipients at high risk for unfavorable early and late kidney transplant outcomes. METHODS Serum sCD30 was measured on the day of kidney transplantation and on the 4th day posttransplant. We assessed the value of these measurements in predicting delayed graft function, slow graft function (SGF), acute rejection (AR), pyelonephritis, decline of allograft function after 6 months, and graft and patient survival during 5 years of follow-up in 45 recipients. RESULTS We found the association between low pretransplant serum levels of sCD30 and SGF. The absence of significant decrease of sCD30 on the 4th day posttransplant was characteristic for SGF, early AR (the 8th day-6 months), late AR (>6 months), and early pyelonephritis (the 8th day-2 months). Lower pretransplant and posttransplant sCD30 predicted worse allograft function at 6 months and 2 years, respectively. Higher pretransplant sCD30 was associated with higher frequency of early AR, and worse patients' survival, but only in the recipients of deceased-donor graft. Pretransplant sCD30 also allowed to differentiate patients with early pyelonephritis and early AR. CONCLUSIONS Peritransplant sCD30 is useful in identifying patients at risk for unfavorable early and late transplant outcomes.
Collapse
Affiliation(s)
- Andriy V. Trailin
- Department of Laboratory Diagnostics and General Pathology, State Institution “Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine”, 20 Winter Boulevard, Zaporizhzhia 69096, Ukraine
| | - Tetyana I. Ostapenko
- Department of Transplantology, Endocrine Surgery and Cardiovascular Surgery, State Institution “Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine”, Zaporizhzhia Regional Hospital, 10 Orikhiv Highway, Zaporizhzhia 69050, Ukraine
| | - Tamara N. Nykonenko
- Institute of Cardiovascular Surgery and Transplantology, State Institution “Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine”, 20 Winter Boulevard, Zaporizhzhia 69096, Ukraine
| | - Svitlana N. Nesterenko
- Immunological Laboratory, Zaporizhzhia Regional Hospital, State Institution “Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine”, 10 Orikhiv Highway, Zaporizhzhia 69050, Ukraine
| | - Olexandr S. Nykonenko
- Department of Transplantology, Endocrine Surgery and Cardiovascular Surgery, State Institution “Zaporizhzhia Medical Academy of Postgraduate Education Ministry of Health of Ukraine”, Zaporizhzhia Regional Hospital, 10 Orikhiv Highway, Zaporizhzhia 69050, Ukraine
| |
Collapse
|
14
|
Salvadori M, Tsalouchos A. Pre-transplant biomarkers and prediction of post-transplant outcomes in kidney transplantation. J Renal Inj Prev 2017; 6:222-230. [DOI: 10.15171/jrip.2017.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
|
15
|
Fernández-Ruiz M, Parra P, López-Medrano F, Ruiz-Merlo T, González E, Polanco N, Origüen J, San Juan R, Andrés A, Aguado JM. Serum sCD30: A promising biomarker for predicting the risk of bacterial infection after kidney transplantation. Transpl Infect Dis 2017; 19. [PMID: 28122147 DOI: 10.1111/tid.12668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The transmembrane glycoprotein CD30 contributes to regulate the balance between Th1 and Th2 responses. Previous studies have reported conflicting results on the utility of its soluble form (sCD30) to predict post-transplant infection. METHODS Serum sCD30 was measured by a commercial ELISA assay at baseline and post-transplant months 1, 3, and 6 in 100 kidney transplant (KT) recipients (279 monitoring points). The impact of sCD30 levels on the incidence of overall, bacterial and opportunistic infection during the first 12 months after transplantation was assessed by Cox regression. RESULTS There were no differences in serum sCD30 according to the occurrence of overall or opportunistic infection. However, sCD30 levels were higher in patients with bacterial infection compared to those without at baseline (P=.038) and months 1 (P<.0001), 3 (P=.043), and 6 after transplantation (P=.006). Patients with baseline sCD30 levels ≥13.5 ng/mL had lower 12-month bacterial infection-free survival (35.0% vs 80.0%; P<.0001). After adjusting for potential confounders, baseline sCD30 levels ≥13.5 ng/mL remained as an independent risk factor for bacterial infection (adjusted hazard ratio [aHR]: 4.65; 95% confidence interval [CI]: 2.05-10.53; <.001). Analogously, sCD30 levels ≥6.0 ng/mL at month 1 acted as a risk factor for subsequent bacterial infection (aHR: 5.29; 95% CI: 1.11-25.14; P=.036). CONCLUSION Higher serum sCD30 levels were associated with an increased risk of bacterial infection after KT. We hypothesize that this biomarker reflects a Th2 -polarized T-cell response, which exerts a detrimental effect on the immunity against bacterial pathogens.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Esther González
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Julia Origüen
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - Amado Andrés
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Hospital "12 de Octubre" (i+12), School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
16
|
Menon MC, Murphy B, Heeger PS. Moving Biomarkers toward Clinical Implementation in Kidney Transplantation. J Am Soc Nephrol 2017; 28:735-747. [PMID: 28062570 DOI: 10.1681/asn.2016080858] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Long-term kidney transplant outcomes remain suboptimal, delineating an unmet medical need. Although current immunosuppressive therapy in kidney transplant recipients is effective, dosing is conventionally adjusted empirically on the basis of time after transplant or altered in response to detection of kidney dysfunction, histologic evidence of allograft damage, or infection. Such strategies tend to detect allograft rejection after significant injury has already occurred, fail to detect chronic subclinical inflammation that can negatively affect graft survival, and ignore specific risks and immune mechanisms that differentially contribute to allograft damage among transplant recipients. Assays and biomarkers that reliably quantify and/or predict the risk of allograft injury have the potential to overcome these deficits and thereby, aid clinicians in optimizing immunosuppressive regimens. Herein, we review the data on candidate biomarkers that we contend have the highest potential to become clinically useful surrogates in kidney transplant recipients, including functional T cell assays, urinary gene and protein assays, peripheral blood cell gene expression profiles, and allograft gene expression profiles. We identify barriers to clinical biomarker adoption in the transplant field and suggest strategies for moving biomarker-based individualization of transplant care from a research hypothesis to clinical implementation.
Collapse
Affiliation(s)
- Madhav C Menon
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Barbara Murphy
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter S Heeger
- Renal Division, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
17
|
Barcelona Consensus on Biomarker-Based Immunosuppressive Drugs Management in Solid Organ Transplantation. Ther Drug Monit 2016; 38 Suppl 1:S1-20. [PMID: 26977997 DOI: 10.1097/ftd.0000000000000287] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
With current treatment regimens, a relatively high proportion of transplant recipients experience underimmunosuppression or overimmunosuppression. Recently, several promising biomarkers have been identified for determining patient alloreactivity, which help in assessing the risk of rejection and personal response to the drug; others correlate with graft dysfunction and clinical outcome, offering a realistic opportunity for personalized immunosuppression. This consensus document aims to help tailor immunosuppression to the needs of the individual patient. It examines current knowledge on biomarkers associated with patient risk stratification and immunosuppression requirements that have been generally accepted as promising. It is based on a comprehensive review of the literature and the expert opinion of the Biomarker Working Group of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology. The quality of evidence was systematically weighted, and the strength of recommendations was rated according to the GRADE system. Three types of biomarkers are discussed: (1) those associated with the risk of rejection (alloreactivity/tolerance), (2) those reflecting individual response to immunosuppressants, and (3) those associated with graft dysfunction. Analytical aspects of biomarker measurement and novel pharmacokinetic-pharmacodynamic models accessible to the transplant community are also addressed. Conventional pharmacokinetic biomarkers may be used in combination with those discussed in this article to achieve better outcomes and improve long-term graft survival. Our group of experts has made recommendations for the most appropriate analysis of a proposed panel of preliminary biomarkers, most of which are currently under clinical evaluation in ongoing multicentre clinical trials. A section of Next Steps was also included, in which the Expert Committee is committed to sharing this knowledge with the Transplant Community in the form of triennial updates.
Collapse
|
18
|
Pike R, Thomas N, Workman S, Ambrose L, Guzman D, Sivakumaran S, Johnson M, Thorburn D, Harber M, Chain B, Stauss HJ. PD1-Expressing T Cell Subsets Modify the Rejection Risk in Renal Transplant Patients. Front Immunol 2016; 7:126. [PMID: 27148254 PMCID: PMC4827377 DOI: 10.3389/fimmu.2016.00126] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/21/2016] [Indexed: 12/01/2022] Open
Abstract
We tested whether multi-parameter immune phenotyping before or after renal transplantation can predict the risk of rejection episodes. Blood samples collected before and weekly for 3 months after transplantation were analyzed by multi-parameter flow cytometry to define 52 T cell and 13 innate lymphocyte subsets in each sample, producing more than 11,000 data points that defined the immune status of the 28 patients included in this study. Principle component analysis suggested that the patients with histologically confirmed rejection episodes segregated from those without rejection. Protein death 1 (PD-1)-expressing subpopulations of regulatory and conventional T cells had the greatest influence on the principal component segregation. We constructed a statistical tool to predict rejection using a support vector machine algorithm. The algorithm correctly identified 7 out of 9 patients with rejection, and 14 out of 17 patients without rejection. The immune profile before transplantation was most accurate in determining the risk of rejection, while changes of immune parameters after transplantation were less accurate in discriminating rejection from non-rejection. The data indicate that pretransplant immune subset analysis has the potential to identify patients at risk of developing rejection episodes, and suggests that the proportion of PD1-expressing T cell subsets may be a key indicator of rejection risk.
Collapse
Affiliation(s)
- Rebecca Pike
- Institute of Immunity and Transplantation, University College London , London , UK
| | - Niclas Thomas
- Institute of Immunity and Transplantation, University College London , London , UK
| | - Sarita Workman
- Institute of Immunity and Transplantation, University College London , London , UK
| | - Lyn Ambrose
- Institute of Immunity and Transplantation, University College London , London , UK
| | - David Guzman
- Institute of Immunity and Transplantation, University College London , London , UK
| | | | - Margaret Johnson
- Department of HIV Medicine, Royal Free London NHS Foundation Trust , London , UK
| | - Douglas Thorburn
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust , London , UK
| | - Mark Harber
- Department of Renal Medicine, Royal Free London NHS Foundation Trust , London , UK
| | - Benny Chain
- Institute of Immunity and Transplantation, University College London , London , UK
| | - Hans J Stauss
- Institute of Immunity and Transplantation, University College London , London , UK
| |
Collapse
|
19
|
Bamoulid J, Courivaud C, Crepin T, Carron C, Gaiffe E, Roubiou C, Laheurte C, Moulin B, Frimat L, Rieu P, Mousson C, Durrbach A, Heng AE, Rebibou JM, Saas P, Ducloux D. Pretransplant thymic function predicts acute rejection in antithymocyte globulin-treated renal transplant recipients. Kidney Int 2016; 89:1136-1143. [PMID: 27083287 DOI: 10.1016/j.kint.2015.12.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/26/2015] [Accepted: 12/17/2015] [Indexed: 12/25/2022]
Abstract
Lack of clear identification of patients at high risk of acute rejection hampers the ability to individualize immunosuppressive therapy. Here we studied whether thymic function may predict acute rejection in antithymocyte globulin (ATG)-treated renal transplant recipients in 482 patients prospectively studied during the first year post-transplant of which 86 patients experienced acute rejection. Only CD45RA(+)CD31(+)CD4(+) T cell (recent thymic emigrant [RTE]) frequency (RTE%) was marginally associated with acute rejection in the whole population. This T-cell subset accounts for 26% of CD4(+) T cells. Pretransplant RTE% was significantly associated with acute rejection in ATG-treated patients (hazard ratio, 1.04; 95% confidence interval, 1.01-1.08) for each increased percent in RTE/CD4(+) T cells), but not in anti-CD25 monoclonal (αCD25 mAb)-treated patients. Acute rejection was significantly more frequent in ATG-treated patients with high pretransplant RTE% (31.2% vs. 16.4%) or absolute number of RTE/mm(3) (31.7 vs. 16.1). This difference was not found in αCD25 monclonal antibody-treated patients. Highest values of both RTE% (>31%, hazard ratio, 2.50; 95% confidence interval, 1.09-5.74) and RTE/mm(3) (>200/mm(3), hazard ratio, 3.71; 95% confidence interval, 1.59-8.70) were predictive of acute rejection in ATG-treated patients but not in patients having received αCD25 monoclonal antibody). Results were confirmed in a retrospective cohort using T-cell receptor excision circle levels as a marker of thymic function. Thus, pretransplant thymic function predicts acute rejection in ATG-treated patients.
Collapse
Affiliation(s)
- Jamal Bamoulid
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Cécile Courivaud
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Thomas Crepin
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Clémence Carron
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France
| | - Emilie Gaiffe
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| | - Caroline Roubiou
- Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - Caroline Laheurte
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Bruno Moulin
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Strasbourg, Strasbourg, France
| | - Luc Frimat
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Nancy, Nancy, France
| | - Philippe Rieu
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Reims, Reims, France
| | - Christiane Mousson
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - Antoine Durrbach
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Kremlin-Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne-Elisabeth Heng
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Michel Rebibou
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - Philippe Saas
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France; EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Didier Ducloux
- INSERM, UMR1098, Federation hospitalo-universitaire INCREASE, Besançon, France; Faculté de Médecine et de Pharmacie, University Bourgogne Franche-Comté, Besançon, France; Structure Fédérative de Recherche, Besançon, France; Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France; CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France.
| |
Collapse
|
20
|
Abstract
Demographic changes are associated with a steady increase of older patients with end-stage organ failure in need for transplantation. As a result, the majority of transplant recipients are currently older than 50 years, and organs from elderly donors are more frequently used. Nevertheless, the benefit of transplantation in older patients is well recognized, whereas the most frequent causes of death among older recipients are potentially linked to side effects of their immunosuppressants.Immunosenescence is a physiological part of aging linked to higher rates of diabetes, bacterial infections, and malignancies representing the major causes of death in older patients. These age-related changes impact older transplant candidates and may have significant implications for an age-adapted immunosuppression. For instance, immunosenescence is linked to lower rates of acute rejections in older recipients, whereas the engraftment of older organs has been associated with higher rejection rates. Moreover, new-onset diabetes mellitus after transplantation is more frequent in the elderly, potentially related to corticosteroids, calcineurin inhibitors, and mechanistic target of rapamycin inhibitors.This review presents current knowledge for an age-adapted immunosuppression based on both, experimental and clinical studies in and beyond transplantation. Recommendations of maintenance and induction therapy may help to improve graft function and to design future clinical trials in the elderly.
Collapse
|
21
|
T-Cell Surface Antigens and sCD30 as Biomarkers of the Risk of Rejection in Solid Organ Transplantation. Ther Drug Monit 2015; 38 Suppl 1:S29-35. [PMID: 26495982 DOI: 10.1097/ftd.0000000000000259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
T-cell activation is a characteristic of organ rejection. T cells, located in the draining lymph nodes of the transplant recipient, are faced with non-self-molecules presented by antigen presenting cells and become activated. Activated T cells are characterized by up-regulated surface antigens, such as costimulatory molecules, adhesion molecules, chemokine receptors, and major histocompatibility complex class II molecules. Surface antigen expression can be followed by flow cytometry using monoclonal antibodies in either cell function assays using donor-specific or nonspecific stimulation of isolated cells or whole blood and without stimulation on circulating lymphocytes. Molecules such as CD30 can be proteolytically cleaved off the surface of activated cells in vivo, and the determination of the soluble protein (sCD30) in serum or plasma is performed by immunoassays. As promising biomarkers for rejection and long-term transplant outcome, CD28 (costimulatory receptor for CD80 and CD86), CD154 (CD40 ligand), and sCD30 (tumor necrosis factor receptor superfamily, member 8) have been identified. Whereas cell function assays are time-consuming laboratory-developed tests which are difficult to standardize, commercial assays are frequently available for soluble proteins. Therefore, more data from clinical trials have been published for sCD30 compared with the surface antigens on activated T cells. This short review summarizes the association between selected surface antigens and immunosuppression, and rejection in solid organ transplantation.
Collapse
|
22
|
Crespo E, Bestard O. Biomarkers to assess donor-reactive T-cell responses in kidney transplant patients. Clin Biochem 2015; 49:329-37. [PMID: 26279496 DOI: 10.1016/j.clinbiochem.2015.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/23/2015] [Accepted: 08/09/2015] [Indexed: 02/07/2023]
Abstract
Different to antibody-mediated rejection (ABMR), T-cell mediated rejection (TCMR) still unpredictably occurs after kidney transplantation in a great part because of a poor immunologic evaluation of the cellular allogeneic immune response. However, in the last years, important efforts have focused on the development of novel and more sensitive assays to monitor T-cell alloimmune responses at different biological levels that may improve the understanding of the functional status of the cellular immune compartment in patients undergoing organ transplantation. In this direction, immune assays evaluating T-cell proliferation, intracellular ATP release, multiparameter flow cytometry, profiling T-cell receptor repertoires and measurements of frequencies of cytokine-producing T-cells using an IFN-γ enzyme-linked immunospot assay (IFN-γ ELISPOT) have been reported showing interesting associations between the cellular alloimmune response and kidney transplant outcomes. In summary, an important progress has been made in the assessment of alloreactive T-cell responses in the context of organ transplantation using novel immune assays at different biological levels. However, there is an urgent need for prospective, randomized clinical studies to validate these encouraging preliminary data to ultimately introduce them in current clinical practice for refining current immune-risk stratification in kidney transplantation.
Collapse
Affiliation(s)
- Elena Crespo
- Laboratory of Experimental Nephrology, IDIBELL, Barcelona, Spain
| | - Oriol Bestard
- Laboratory of Experimental Nephrology, IDIBELL, Barcelona, Spain; Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona University, Barcelona, Spain.
| |
Collapse
|
23
|
Wieland E, Shipkova M. Lymphocyte surface molecules as immune activation biomarkers. Clin Biochem 2015; 49:347-54. [PMID: 26247177 DOI: 10.1016/j.clinbiochem.2015.07.099] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 07/23/2015] [Accepted: 07/25/2015] [Indexed: 01/07/2023]
Abstract
Immunosuppression is mandatory after solid organ transplantation between HLA mismatched individuals. It is a lifelong therapy that needs to be closely monitored to avoid under- and over-immunosuppression. For many drugs, pharmacokinetic monitoring has been proven to be beneficial. However, the therapeutic ranges are statistically derived surrogate markers for the effects that cannot predict the individual response of single patients. Better tailored immunosuppression biomarkers are needed that indicate immune activation. T cells are critically involved in organ rejection, and the means to assess their activation state may be promising to individualize immunosuppressive therapies. Activated T cells can be monitored with flow cytometry based on surface molecules that are typically up regulated or with molecules that are cleaved off the cell surface. Among these molecules are the interleukin-2 receptor (CD25); transferrin receptor (CD71); the T cell co-stimulatory molecules CD28, CD69, and CD154 and sCD30, which is a member of the TNF-alpha family. The effect of immunosuppressive drugs on T cell activation can be recorded with indirect cell function assays or by directly monitoring activated T cells in whole blood. Soluble proteins can be measured with immunoassays. This review provides a summary of the experimental and clinical studies investigating the potential of surface molecules as a tool for immune monitoring. It critically discusses the obstacles and shortcomings from an analytical and diagnostic perspective that are currently preventing their use in multicenter trials and clinical routine monitoring of transplant patients.
Collapse
Affiliation(s)
- Eberhard Wieland
- Klinikum Stuttgart, Central Institute for Clinical Chemistry and Laboratory Medicine, Germany.
| | - Maria Shipkova
- Klinikum Stuttgart, Central Institute for Clinical Chemistry and Laboratory Medicine, Germany.
| |
Collapse
|
24
|
Soluble co-signaling molecules predict long-term graft outcome in kidney-transplanted patients. PLoS One 2014; 9:e113396. [PMID: 25478957 PMCID: PMC4257538 DOI: 10.1371/journal.pone.0113396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/23/2014] [Indexed: 12/25/2022] Open
Abstract
Co-signaling molecules are responsible for full T-cell activation after solid organ transplantation. Their increased expression can lead to the release of a soluble form that can modulate the immune response post-transplantation. We analyzed the presence of co-signaling molecules (sCD30, sCD40, sCD137, sCTLA-4, sCD80, sCD28, sCD40L, sPD-1, and sPD-L1) in serum from kidney-transplanted patients (n = 59) obtained at different times (before transplantation, and 15 days, 3 months and 1 year post-transplantation) and their contribution to graft outcome was evaluated using principal component analysis. Before transplantation, high levels of soluble co-signaling molecules (mainly sCD30, sCD137 and sCD40) were detected in all patients. These molecules were modulated soon after receiving an allograft but never attained similar levels to those of healthy controls. A signature based on the determination of six soluble co-stimulatory (sCD30, sCD40, sCD137 and sCD40L) and co-inhibitory (sPD-1 and sPD-L1) molecules at 3 months post-transplantation allowed a group of patients to be identified (27.12%) with a worse long-term graft outcome. Patients with high levels of soluble molecules showed a progressive and gradual deterioration of kidney function (increased creatinine and proteinuria levels and decreased estimated glomerular filtration rate) over time and a higher risk of graft loss at 6 years post-transplantation than patients with low levels of these molecules (62.55% versus 5.14%, p<0.001). Thus, our data show an aberrant expression of soluble co-signaling molecules in kidney-transplanted patients whose quantification at 3 months post-transplantation might be a useful biomarker of immune status and help to predict long-term graft evolution.
Collapse
|
25
|
Valke LLFG, van Cranenbroek B, Hilbrands LB, Joosten I. Soluble CD30 does not predict late acute rejection or safe tapering of immunosuppression in renal transplantation. Transpl Immunol 2014; 32:18-22. [PMID: 25446808 DOI: 10.1016/j.trim.2014.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Previous reports revealed the potential value of the soluble CD30 level (sCD30) as biomarker for the risk of acute rejection and graft failure after renal transplantation, here we examined its use for the prediction of safe tapering of calcineurin inhibitors as well as late acute rejection. METHODS In a cohort of renal transplant patients receiving triple immunosuppressive therapy we examined whether sCD30 can be used as a marker for safe (rejection-free) discontinuation of tacrolimus at six months after transplantation (TDS cohort: 24 rejectors and 44 non-rejecting controls). Also, in a second cohort of patients (n=22, rejectors n=11 and non-rejectors n=11), participating in a clinical trial of rituximab as induction therapy after renal transplantation (RITS cohort), we examined whether sCD30 could predict the occurrence of late (>3months post-transplant) acute rejection episodes. sCD30 was measured by ELISA in serum taken before and at several time points after transplantation. RESULTS Overall, in the TDS cohort sCD30 decreased after transplantation. No difference in sCD30 was observed between rejectors and non-rejecting controls at any of the time points measured. In addition, in the RITS cohort, sCD30 measured at three months after transplantation were not indicative for the occurrence of late acute rejection. CONCLUSION In two prospectively followed cohorts of renal transplant patients we found no association between sCD30 and the occurrence of either late acute rejection or acute rejection after reduction of immunosuppression.
Collapse
Affiliation(s)
- Lars L F G Valke
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Centre, PO Box 9101 6500HB Nijmegen, the Netherlands; Department of Nephrology, Radboud University Medical Centre, PO Box 9101 6500HB Nijmegen, the Netherlands.
| | - Bram van Cranenbroek
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Centre, PO Box 9101 6500HB Nijmegen, the Netherlands.
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Centre, PO Box 9101 6500HB Nijmegen, the Netherlands.
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Centre, PO Box 9101 6500HB Nijmegen, the Netherlands.
| |
Collapse
|
26
|
Vondran FWR, Timrott K, Kollrich S, Steinhoff AK, Kaltenborn A, Schrem H, Klempnauer J, Lehner F, Schwinzer R. Pre-transplant immune state defined by serum markers and alloreactivity predicts acute rejection after living donor kidney transplantation. Clin Transplant 2014; 28:968-79. [PMID: 24931031 DOI: 10.1111/ctr.12399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2014] [Indexed: 02/06/2023]
Abstract
Acute rejection (AR) remains a major cause for long-term kidney allograft failure. Reliable immunological parameters suitable to define the pre-transplant immune state and hence the individual risk of graft rejection are highly desired to preferably adapt the immunosuppressive regimen in advance. Donor and third party alloreactivities were determined by mixed lymphocyte cultures. Soluble forms of CD25, CD30, and CD44 were detected in patients' serum by ELISA. Various lymphocyte subpopulations were measured using flow cytometry. All patients received triple immunosuppression (tacrolimus/mycophenolate mofetil/steroids) and were grouped according to biopsy results within the first year: rejection-free (RF, n = 13), borderline (BL, n = 5), or acute rejection (AR, n = 7). Patients with AR showed the highest pre-transplant alloreactivities and serum levels (sCD25/sCD30/sCD44) according to the pattern RF < BL < AR. Relying on serum analysis only, multivariate logistic regression (logit link function) yielded a prognostic score for prediction of rejection with 75.0% sensitivity and 69.2% specificity. Patients with rejection showed markedly higher pre-transplant frequencies of CD4(+) /CD8(+) T cells lacking CD28, but lower numbers of CD8(+) CD161(bright) T cells and NK cells than RF individuals. Pre-transplant immune state defined by alloreactivity, serum markers, and particular lymphocyte subsets seems to correlate with occurrence of graft rejection after kidney transplantation. A prognostic score based on pre-transplant serum levels has shown great potential for prediction of rejection episodes and should be further evaluated.
Collapse
Affiliation(s)
- Florian W R Vondran
- Transplant Laboratory, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Clinical immune-monitoring strategies for predicting infection risk in solid organ transplantation. Clin Transl Immunology 2014; 3:e12. [PMID: 25505960 PMCID: PMC4232060 DOI: 10.1038/cti.2014.3] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 01/28/2014] [Accepted: 01/28/2014] [Indexed: 02/06/2023] Open
Abstract
Infectious complications remain a leading cause of morbidity and mortality after solid organ transplantation (SOT), and largely depend on the net state of immunosuppression achieved with current regimens. Cytomegalovirus (CMV) is a major opportunistic viral pathogen in this setting. The application of strategies of immunological monitoring in SOT recipients would allow tailoring of immunosuppression and prophylaxis practices according to the individual's actual risk of infection. Immune monitoring may be pathogen-specific or nonspecific. Nonspecific immune monitoring may rely on either the quantification of peripheral blood biomarkers that reflect the status of a given arm of the immune response (serum immunoglobulins and complement factors, lymphocyte sub-populations, soluble form of CD30), or on the functional assessment of T-cell responsiveness (release of intracellular adenosine triphosphate following a mitogenic stimulus). In addition, various methods are currently available for monitoring pathogen-specific responses, such as CMV-specific T-cell-mediated immune response, based on interferon-γ release assays, intracellular cytokine staining or main histocompatibility complex-tetramer technology. This review summarizes the clinical evidence to date supporting the use of these approaches to the post-transplant immune status, as well as their potential limitations. Intervention studies based on validated strategies for immune monitoring still need to be performed.
Collapse
|
28
|
Solgi G, Gadi V, Paul B, Mytilineos J, Pourmand G, Mehrsai A, Ranjbar M, Mohammadnia M, Nikbin B, Amirzargar AA. Five-year clinical effects of donor bone marrow cells infusions in kidney allograft recipients: improved graft function and higher graft survival. CHIMERISM 2013; 4:87-94. [PMID: 23639966 DOI: 10.4161/chim.24719] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Augmentation of microchimerism in solid organ transplant recipients by donor bone marrow cells (DBMC) infusion may promote immune hyporesponsiveness and consequently improve long-term allograft survival. Between March 2005 and July 2007, outcomes for 20 living unrelated donor (LURD) primary kidney recipients with concurrent DBMC infusion (an average of 2.19 ± 1.13 x 10⁹ donor cells consisting of 2.66 ± 1.70 x 10⁷ CD34⁺ cells) were prospectively compared with 20 non-infused control allograft recipients given similar conventional immunosuppressive regimens. With five years of clinical follow up, a total of 11 cases experienced rejection episodes (3 DBMI patients vs. 8 controls, p = 0.15). One DBMC-infused patient experienced chronic rejection vs. two episodes (1 biopsy-confirmed) in the control patients. Actuarial and death-censored 5-y graft survival was significantly higher in infused patients compared with controls (p = 0.01 and p = 0.03, respectively). Long-term graft survival was significantly associated with pre-transplant anti-HLA antibodies (p = 0.01), slightly with peripheral microchimerism (p = 0.09) and CD4⁺CD25⁺FoxP3⁺ T cells (p = 0.09). Immunosuppressant dosing was lower in infused patients than controls, particularly for mycophenolate mofetil (p = 0.001). The current findings as well as our previous reports on these patients indicates clinical improvement in long-term graft survival of renal transplant patients resulting from low-dose DBMC infusion given without induction therapy.
Collapse
Affiliation(s)
- Ghasem Solgi
- Immunology Department; School of Medicine; Hamadan University of Medical Sciences; Hamadan, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|