1
|
Zajacova A, Alkhouri M, Guney M, Ferrao G, Rezac D, Vyskocilova K, Kotowski T, Dutkova A, Dvorackova E, Lischke R, Fila L, Havlin J. Exploring acute cellular rejection in lung transplantation: insights from donor-derived cell-free DNA analysis. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2025. [PMID: 40391837 DOI: 10.5507/bp.2025.016] [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: 05/22/2025] Open
Abstract
BACKGROUND Acute cellular rejection (ACR) is a frequent complication following lung transplantation, yet standardized guidelines for ACR screening remain lacking. This study aimed to compare the current gold standard for ACR evaluation - histological assessment of transbronchial biopsies - with a novel biomarker for allograft monitoring: donor-derived cell-free DNA (dd-cfDNA). Specifically, we investigated whether total cell-free DNA (cfDNA) and both the absolute and percentage values of dd-cfDNA (dd-cfDNA and dd-cfDNA%) could provide valuable insights into detecting ACR and assessing allograft health. METHODS Patients after bilateral lung transplantation between May 2021 and March 2024 were included. Clinically significant ACR cases (ACR+) were defined as samples with histological ACR grade ≥A2 or ACR grade A1 in patients who have received antirejection therapy due to symptoms, CT findings, or lung function decline. Samples with A0 or A1 rejection in clinically stable, untreated patients were classified as controls. Measurements of dd-cfDNA%, dd-cfDNA (cp/mL) and total cfDNA (cp/mL) were obtained at the time of biopsy and compared between cohorts. RESULTS The median dd-cfDNA concentration was significantly higher in the ACR+ group (61.2 cp/mL, IQR: 38.7-114.1) compared to controls (25.8 cp/mL, IQR: 10.7-65.7; P=0.04). However, no significant differences were observed for dd-cfDNA% and cfDNA. CONCLUSION Dd-cfDNA shows promise as a valuable tool for ruling out ACR; however, further research is necessary in order to validate its clinical utility and optimize its implementation. Its negative predictive value supports dd-cfDNA as an effective screening tool for allograft health, nevertheless, further investigation is required.
Collapse
Affiliation(s)
- Andrea Zajacova
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Majd Alkhouri
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Miray Guney
- Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Goncalo Ferrao
- Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - David Rezac
- 7th Field Hospital of the Army of the Czech Republic, Hradec Kralove, Czechia
| | - Kristyna Vyskocilova
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Tereza Kotowski
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Alzbeta Dutkova
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Eliska Dvorackova
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Robert Lischke
- Prague Lung Transplant Program, 3rd Department of Surgery, First Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Libor Fila
- Prague Lung Transplant Program, Department of Pneumology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Jan Havlin
- Prague Lung Transplant Program, 3rd Department of Surgery, First Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| |
Collapse
|
2
|
Charya AV, Jang MK, Kong H, Park W, Tian X, Keller M, Phipps K, Sanders A, Shah P, Mathew J, Aryal S, Berry GJ, Marboe C, Orens JB, Nathan SD, Agbor-Enoh S. Donor-derived cell-free DNA is associated with the degree of immunosuppression in lung transplantation. Am J Transplant 2025:S1600-6135(25)00205-9. [PMID: 40252922 DOI: 10.1016/j.ajt.2025.04.011] [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: 11/07/2024] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Donor-derived cell-free DNA (ddcfDNA) is increasingly used in clinical practice to monitor lung transplant patients for acute rejection (AR). However, its association with conventional approaches to monitor immunosuppression remains unclear. This multicenter observational cohort study examines the association of ddcfDNA with surrogate measures of immunosuppression. We collected serial plasma samples to quantify ddcfDNA and anellovirus abundance using shotgun and metagenomic sequencing. Adjudication committees reviewed clinical data to define acute cellular and antibody-mediated rejection. The association between ddcfDNA, anellovirus abundance, and tacrolimus trough concentrations in serum over the study period and during episodes of AR was examined using linear mixed-effects modeling. ddcfDNA demonstrated a significant inverse association with tacrolimus troughs (P = .027) and anellovirus abundance (P < .001) over time. AR episodes were associated with significantly decreased anellovirus abundance (median, 0.042 vs 0.708, P < .001) and higher ddcfDNA levels (1.49% vs 0.26%, P < .001) compared with stable control time points. However, tacrolimus levels were similar between AR and controls (10.1 vs 10.3 ng/mL, P = .13). Our findings suggest ddcfDNA correlates with measures of immunosuppression in lung transplant patients. Additional studies are needed to assess the utility of ddcfDNA to assess immunosuppression adequacy.
Collapse
Affiliation(s)
- Ananth V Charya
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Moon K Jang
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Woojin Park
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Michael Keller
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Division of Pulmonary and Critical Care, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Kellie Phipps
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Auriel Sanders
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Pali Shah
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Joby Mathew
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shambhu Aryal
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia, USA
| | - Gerald J Berry
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Charles Marboe
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Department of Pathology, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York, USA
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia, USA
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA.
| |
Collapse
|
3
|
Keller MB, Newman D, Alnababteh M, Bon A, Ponor L, Shah P, Mathew J, Kong H, Andargie T, Park W, Charya A, Luikart H, Intrieri T, Aryal S, Nathan SD, Orens JB, Khush KK, Jang M, Agbor-Enoh S. Molecular criteria for pulmonary antibody-mediated rejection are associated with an increased risk of allograft failure. J Heart Lung Transplant 2025:S1053-2498(25)01853-4. [PMID: 40120999 DOI: 10.1016/j.healun.2025.03.015] [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: 10/24/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Current International Society for Heart and Lung Transplantation (ISHLT) criteria for pulmonary antibody-mediated rejection (AMR) is predicated on a constellation of clinical, laboratory and histopathological parameters, including the presence of donor-specific antibodies (DSA). However, molecular evidence of allograft injury is not considered. The aim of this study was to investigate if allograft injury on the molecular level, as measured by donor-derived cell-free DNA (dd-cfDNA), identifies DSA positive patients experiencing a form of AMR associated with increased risk of chronic lung allograft dysfunction (CLAD) or death. METHODS This multicenter, observational analysis included adult lung transplant recipients from 2 prospective cohort studies. Serial plasma samples were collected for dd-cfDNA measurement by shotgun sequencing. Molecular AMR was defined as the presence of DSA and dd-cfDNA level >1% occurring >30 days post-transplant. Clinical AMR was defined using ISHLT criteria. Time-dependent multivariable Cox regression models were used to determine the association of Clinical AMR or Molecular AMR with the composite outcome of CLAD or death. RESULTS The final analysis included 209 subjects. Sixty-one subjects met criteria for molecular AMR. Molecular AMR captured 42/46 (91%) of patients who experienced Clinical AMR. Molecular AMR was associated with an increased risk of CLAD or death (HR 2.00, 95% CI: 1.18-3.38, p = 0.010). The results remained consistent analyzing Molecular AMR subjects without concomitant ISHLT Clinical AMR, acute rejection, or infection (HR 2.45, 95% CI: 1.01-5.94, p = 0.047). CONCLUSIONS Molecular AMR identifies a population of lung transplant recipients potentially experiencing antibody-mediated rejection not captured by current ISHLT criteria.
Collapse
Affiliation(s)
- Michael B Keller
- Division of Pulmonary, Critical Care & Sleep Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - David Newman
- College of Nursing, Florida Atlantic University, Boca Raton, Florida
| | - Muhtadi Alnababteh
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Ann Bon
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Lucia Ponor
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Pali Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Joby Mathew
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Hyesik Kong
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Temesgen Andargie
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Woojin Park
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Ananth Charya
- Division of Pulmonary, Critical Care & Sleep Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Helen Luikart
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California; Department of Pathology, Stanford University School of Medicine, Palo Alto, California
| | - Tyler Intrieri
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California; Department of Pathology, Stanford University School of Medicine, Palo Alto, California
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary Division, Inova Fairfax Hospital, Falls Church, Virginia
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary Division, Inova Fairfax Hospital, Falls Church, Virginia
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Kiran K Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Moon Jang
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Sean Agbor-Enoh
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland.
| |
Collapse
|
4
|
Liao F, Zhou D, Cano M, Liu Z, Scozzi D, Tague LK, Byers DE, Li W, Sivapackiam J, Sharma V, Krupnick AS, Frank DW, Kreisel D, Kulkarni HS, Hachem RR, Gelman AE. Pseudomonas aeruginosa infection induces intragraft lymphocytotoxicity that triggers lung transplant antibody-mediated rejection. Sci Transl Med 2025; 17:eadp1349. [PMID: 39908350 DOI: 10.1126/scitranslmed.adp1349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
How pathogens inhibit transplant tolerance remains unclear. Here, we found that Pseudomonas aeruginosa infection, but not other common bacterial respiratory infections, increases antibody-mediated rejection (AMR) risk in recipients of lung transplants. To explore this relationship, we performed orthotopic lung transplants in mice, infected recipients with P. aeruginosa, and observed for the development of AMR. Intravital two-photon microscopy showed that P. aeruginosa rapidly invaded bronchial-associated lymphoid tissues, which resulted in acute lymphocytotoxicity, including the death of forkhead box P3 (Foxp3)+CD4+ T cells that are required to suppress AMR. P. aeruginosa-mediated AMR required expression of the type III secretion system (T3SS), which injects exotoxins into the cell cytoplasm. Through a combination of mutagenesis and epitope tagging experiments, we revealed that T3SS exotoxin T ADP ribosyl-transferase activity was sufficient for graft-resident Foxp3+CD4+ T cell apoptosis, leading to myeloid differentiation primary response 88 (Myd88)-dependent generation of T-box expressed in T cells (T-bet)- and C-X-C motif chemokine receptor 3 (CXCR3)-positive germinal center and memory B cells with high donor antigen avidity. We also found that T-bet+ and CXCR3+ B cells were elevated in biopsies from recipients of lung transplants who were diagnosed with AMR. In mice, CXCR3 deficiency restricted to B cells or CXCR3 blockade prevented AMR despite P. aeruginosa infection. Our work has identified a previously unrecognized role of bacterial virulence in lung allograft rejection and suggests potential strategies to prevent AMR for those at high risk of P. aeruginosa infection after transplant.
Collapse
Affiliation(s)
- Fuyi Liao
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dequan Zhou
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marlene Cano
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhiyi Liu
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Davide Scozzi
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laneshia K Tague
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjun Li
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vijay Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander S Krupnick
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dara W Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel Kreisel
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hrishikesh S Kulkarni
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramsey R Hachem
- Department of Internal Medicine, Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84108, USA
| | - Andrew E Gelman
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Millán O, Julian J, Brunet M. miRNAs, dd-cf-DNA, and Chemokines as Potential Noninvasive Biomarkers for the Assessment of Clinical Graft Evolution and Personalized Immunosuppression Requirement in Solid Organ Transplantation. Ther Drug Monit 2025; 47:77-97. [PMID: 39503575 DOI: 10.1097/ftd.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/20/2024] [Indexed: 11/08/2024]
Abstract
ABSTRACT The use of noninvasive biomarkers may reduce the need for biopsy and guide immunosuppression adjustments during transplantation. The scientific community in solid organ transplantation currently considers that chemokines, T- and B-cell immunophenotypes, and gene expression, among other molecular biomarkers, have great potential as diagnostic and predictive biomarkers for graft evolution; however, in clinical practice, few valid early biomarkers have emerged. This review focuses on the most relevant scientific advances in this field in the last 5 years regarding the role of 3 biomarkers: miRNAs, chemokines, and ddcf-DNA, in both adult and pediatric populations. An update was provided on the scores based on the combination of these biomarkers. The most-featured articles were identified through a literature search of the PubMed database. This review provides a comprehensive analysis of the potential clinical applications of these biomarkers in the diagnosis and prediction of graft outcomes and discusses the reasons why none have been implemented in clinical practice to date. Translating these biomarkers into routine clinical practice and combining them with pharmacogenetics and pharmacokinetic monitoring is challenging; however, it is the key to present/future individualized immunosuppressive therapies. It is essential that they be shown to be applicable and robust in real-life patient conditions and properly evaluate their added value when combined with the standard-of-care factor monitoring for graft clinical assessment. Partnership strategies among scientists, academic institutions, consortia, including expert working groups and scientific societies, and pharmaceutical and/or biotechnology companies should promote the development of prospective, randomized, multicenter intervention studies for adequate clinical validation of these biomarkers and their monitoring frequency, and their commercialization to make them available to transplant physicians.
Collapse
Affiliation(s)
- Olga Millán
- Biomedical Research Center in Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III (ISCII), c/Sinesio Delgado, Madrid; and
- Pharmacology and Toxicology, Biochemistry and Molecular Genetics, Biomedical Diagnostic Center (CDB), Hospital Clinic of Barcelona, IDIBAPS, University of Barcelona, c/Villarroel, Barcelona, Spain
| | - Judit Julian
- Pharmacology and Toxicology, Biochemistry and Molecular Genetics, Biomedical Diagnostic Center (CDB), Hospital Clinic of Barcelona, IDIBAPS, University of Barcelona, c/Villarroel, Barcelona, Spain
| | - Mercè Brunet
- Biomedical Research Center in Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III (ISCII), c/Sinesio Delgado, Madrid; and
- Pharmacology and Toxicology, Biochemistry and Molecular Genetics, Biomedical Diagnostic Center (CDB), Hospital Clinic of Barcelona, IDIBAPS, University of Barcelona, c/Villarroel, Barcelona, Spain
| |
Collapse
|
6
|
Yavuz G, Walter J, Hirv K, Wachter O, Dick A, Kovacs J, Zimmermann J, Glueck OM, Vorstandlechner M, Samm N, Fertmann JM, Sienel W, Michel S, Irlbeck M, Kneidinger N, Hatz R, Behr J, Schneider C, Kauke T. Donor-derived cell-free DNA for detection of acute rejection in lung transplant recipients. Front Immunol 2025; 16:1531774. [PMID: 39944692 PMCID: PMC11814210 DOI: 10.3389/fimmu.2025.1531774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/13/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Acute rejection is a significant risk factor for developing chronic lung allograft dysfunction. Current monitoring tools, transbronchial biopsies and HLA antibody determination, have limitations in detecting acute rejection. This study aims to explore the potential utility of donor-derived cell-free DNA (ddcfDNA) as a non-invasive biomarker for detecting acute rejection in lung transplant recipients (LTR). Methods We developed a molecular method based on digital droplet PCR to determine the total amount and the proportion of ddcfDNA. Using blood samples collected sequentially post-transplant from a cohort of 81 LTR, we compared median levels of %ddcfDNA in patients with acute cellular rejection (ACR), antibody-mediated rejection (AMR), infection, or decline in pulmonary function (FEV1). Results Median %ddcfDNA levels were significantly higher in groups with ACR (1.92% [0.70%, 2.30%], p=0.0006), AMR (1.27% [0.34%, 2.29%], p=0.0009), isolated lymphocytic bronchiolitis (0.54% [0.23%, 2.18%], p=0.03), and infection or prolonged ventilation over 30 days (0.50% [0.22%, 2.35%], p=0.005) versus stable allograft function group (0.26% [0.09%, 0.60%]). %ddcfDNA levels were also elevated in patients with FEV1 loss compared to those with stable or improving FEV1 after 12 months (1.98% vs. 1.36%, p=0.04). An optimal cut-off of 0.73% for %ddcfDNA was calculated to detect ACR and AMR with 80% specificity and 68% sensitivity. Discussion %ddcfDNA is a promising biomarker for identifying allograft injury due to acute rejection in LTR and could be a valuable tool for monitoring allograft health.
Collapse
Affiliation(s)
- Gökce Yavuz
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Julia Walter
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
- Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Andrea Dick
- Division of Transfusion Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Kovacs
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Julia Zimmermann
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Olaf M. Glueck
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | - Nicole Samm
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Jan M. Fertmann
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Wulf Sienel
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Michel
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Michael Irlbeck
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rudolf Hatz
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christian Schneider
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Teresa Kauke
- Division of Thoracic Surgery, University Hospital, LMU Munich, Munich, Germany
- Transplant Center, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Smith JB, Peterson RA, Pomponio R, Steele M, Gray AL. Donor derived cell free DNA in lung transplant recipients rises in setting of allograft instability. FRONTIERS IN TRANSPLANTATION 2024; 3:1497374. [PMID: 39737412 PMCID: PMC11683734 DOI: 10.3389/frtra.2024.1497374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025]
Abstract
Purpose The purpose of this study was to evaluate the correlation between longitudinal monitoring of donor-derived cell free DNA (dd-cfDNA) in lung transplant recipients and a "gold standard" of existing tools (pulmonary function testing, radiographic imaging, laboratory and bronchoscopy data, clinical judgment) to assess allograft function. Methods 24 consecutive transplant recipients were prospectively enrolled in this study measuring dd-cfDNA levels monthly in the first year after bilateral lung transplant. Blinded clinical adjudications were performed at the same timepoints to categorize allograft function as stable (FEV1 within 10% of prior value or when compared to best two averaged post-transplant values) or unstable. When deemed unstable, etiology of unstable graft function was elicited based on available clinical data. We then evaluated the association between dd-cfDNA and the clinical impression of allograft function using linear mixed models which adjusted for patient-level covariates and time since transplant. Results Unstable allografts were associated with 54.4% higher measures of dd-cfDNA, controlling for time since transplant and demographic covariates [adjusted mean ratio (aMR) = 1.54, 95% CI: 1.25-1.91]. Females tended to have higher measures of dd-cfDNA (aMR = 1.90 95%CI: 1.14-3.16). A two-fold increase in dd-cfDNA was associated with declines in FEV1 and FVC of 0.047 and 0.066 L, respectively, controlling for time since transplant and demographic covariates (slope: -0.047 95%CI: -0.076 to -0.019, and slope: -0.066 95%CI: -0.097 to -0.035, respectively). Discussion: Donor derived cell free DNA presents a potential additional minimally invasive clinical tool in lung transplant allograft monitoring within the first year of transplant, with unstable allografts correlating with higher dd-cfDNA values.
Collapse
Affiliation(s)
- Joshua B. Smith
- Division of Pulmonary Sciences and Critical Care, University of Colorado School of Medicine, Aurora, CO, United States
| | - Ryan A. Peterson
- Department of Biostatistics and Informatics, Center for Innovative Design and Analysis, University of Colorado School of Public Health, Aurora, CO, United States
| | - Raymond Pomponio
- Department of Biostatistics and Informatics, Center for Innovative Design and Analysis, University of Colorado School of Public Health, Aurora, CO, United States
| | - Mark Steele
- Division of Pulmonary Sciences and Critical Care, University of Colorado School of Medicine, Aurora, CO, United States
| | - Alice L. Gray
- Division of Pulmonary Sciences and Critical Care, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
8
|
Sindu D, Bay C, Grief K, Walia R, Tokman S. Clinical utility of plasma percent donor-derived cell-free DNA for lung allograft surveillance: A real-world single-center experience. JHLT OPEN 2024; 6:100141. [PMID: 40145063 PMCID: PMC11935403 DOI: 10.1016/j.jhlto.2024.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background Plasma percent donor-derived cell-free DNA (%dd-cfDNA) has been investigated as a biomarker of allograft injury after lung transplantation. We sought to determine the clinical utility of %dd-cfDNA as a screen for acute cellular rejection (ACR) and respiratory infections (RIs) among lung transplant recipients (LTRs). Methods We retrospectively analyzed %dd-cfDNA results from 95 plasma samples collected from 81 bilateral LTRs >45 days after transplant with a paired transbronchial biopsy performed within 24 hours after sample collection. We calculated sensitivity, specificity, negative predictive value (NPV), and positive predictive value of %dd-cfDNA to detect ACR and RIs and used a generalized estimating equation model to compare %dd-cfDNA between groups. Results A dd-cfDNA threshold of 0.5% had low sensitivity to detect ACR among LTRs (41.67%), as did a 70% increase in %dd-cfDNA (50.00%). The NPV was high (88.89% and 87.50%, respectively) but driven by the low prevalence of ACR (12/95 [12.6%]). The area under the receiver operating characteristic curve (AUC) was 0.499 (95% confidence interval [CI] [0.326-0.672]) and 0.360 (95%CI [0.132-0.588]) for the detection of ACR and ACR grade ≥A2, respectively. The adjusted mean %dd-cfDNA trended higher in LTRs with a definite or possible RI (1.218, 95%CI [0.671-2.212]) than in LTRs without microbial isolation (0.731, 95%CI [0.525-1.017], p = 0.059), but was not significantly different from those with microbial colonization (0.873, 95%CI [0.538-1.415], p = 0.390). The AUC for the detection of allograft dysfunction due to ACR and/or RI was 0.573 (95%CI [0.431-0.716]). Conclusions %dd-cfDNA may have limited utility as a screening tool to detect ACR and/or RI among LTRs.
Collapse
Affiliation(s)
- Devika Sindu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Curt Bay
- A. T. Still University, Phoenix, Arizona
| | - Katherine Grief
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
- Creighton University School of Medicine, Phoenix Health Sciences Campus, Phoenix, Arizona
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
- Creighton University School of Medicine, Phoenix Health Sciences Campus, Phoenix, Arizona
| |
Collapse
|
9
|
Arunachalam A, Anjum F, Rosenheck JP, Rampolla R, Girgis R, Huang HJ, Crabtree K, McCormick S, Zhang Z, Bhorade S, Ross DJ. Donor-derived cell-free DNA is a valuable monitoring tool after single lung transplantation: Multicenter analysis. JHLT OPEN 2024; 6:100155. [PMID: 40145054 PMCID: PMC11935494 DOI: 10.1016/j.jhlto.2024.100155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background Donor-derived cell-free DNA (dd-cfDNA) is a nonspecific plasma biomarker for tissue injury that has been validated for monitoring acute rejection (AR) after lung transplantation (LT). However, no studies to date have focused specifically on single lung transplantation (SLT). Herein, we report the performance of dd-cfDNA in detecting AR in SLT from 6 academic centers that implemented this biomarker surveillance in their standard of practice (SOP). Methods dd-cfDNA test results were corrected for SLT by an algorithm in the Clinical Laboratory Improvement Amendments (CLIA) laboratory to permit comparison against the same 1.0% threshold used in double-lung transplant. Investigators reviewed patient SOP electronic medical record clinical data to assign test results into cohorts based on clinical allograft health status. To avoid ambiguity in interpretation, samples drawn after a prior AR or infection event or without histopathologic confirmation of AR were excluded from further analysis. Diagnostic cohorts included AR (N=25 samples), healthy (STABLE, N=137), allograft infection (INFXN, N=41), chronic lung allograft dysfunction (CLAD, N=7), and "OTHER" types of graft injury (N=12). Results The study included a total of 257 dd-cfDNA results from 103 SLT patients with one patient excluded due to active cancer. Samples were drawn a median of 233 days (interquartile range: 96-489) after SLT. Laterality for SLT (R vs L) and median dd-cfDNA fraction in AR and STABLE cohort were not statistically different. The median dd-cfDNA fraction was elevated with AR (1.8%) and INFXN (1.1%) vs STABLE (0.46%; p < 0.0001). dd-cfDNA with CLAD was also significantly higher than STABLE cohort (p = 0.0155). The area under receiver operator characteristics curve was 0.850 (95% confidence interval: 0.72-0.95, p < 0.0001) for AR vs STABLE cohort. Applying the dd-cfDNA threshold ≥1.0% for detection of AR yielded a sensitivity = 77.8%, specificity = 84.6%, positive predictive value = 38.31%, and negative predictive value = 96.83%. Conclusions These multicenter data, incorporating real-world experiences, support the clinical validity and utility of dd-cfDNA monitoring of SLT recipients. Additional studies of the impact of biomarker surveillance on clinically meaningful outcomes should be forthcoming from robust, prospective, and clinical trials already in progress.
Collapse
Affiliation(s)
- Ambalavanan Arunachalam
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Fatima Anjum
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Justin P. Rosenheck
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio
| | - Reinaldo Rampolla
- Cedars-Sinai Medical Center, Department of Cardiac Surgery, Los Angeles, California
| | - Reda Girgis
- Corewell HealthCare and Michigan State University College of Human Medicine, Grand Rapids, Michigan
| | - Howard J. Huang
- Houston Methodist Lung Transplant Center, Houston Methodist Hospital, Houston, Texas
| | | | | | | | | | | |
Collapse
|
10
|
Zhang W, Liu B, Jia D, Wang R, Cao H, Wu H, Ye Z, Gao B. Application of graft-derived cell-free DNA for solid organ transplantation. Front Immunol 2024; 15:1461480. [PMID: 39376561 PMCID: PMC11456428 DOI: 10.3389/fimmu.2024.1461480] [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: 07/08/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024] Open
Abstract
Monitoring the status of grafts and the occurrence of postoperative complications, such as rejection, is crucial for ensuring the success and long-term survival of organ transplants. Traditional histopathological examination, though effective, is an invasive procedure and poses risks of complications, making frequent use impractical. In recent years, graft-derived cell-free DNA (gd-cfDNA) has emerged as a promising non-invasive biomarker. It not only provides early warnings of rejection and other types of graft injury but also offers important information about the effectiveness of immunosuppressive therapy and prognosis. gd-cfDNA shows potential in the monitoring of organ transplants. The early, real-time information on graft injury provided by gd-cfDNA facilitates timely individualized treatment and improves patient outcomes. However, the progress of research on gd-cfDNA varies across different organs. Therefore, this article will comprehensively review the application and findings of gd-cfDNA in monitoring various solid organs, discussing the advantages, limitations, and some future research directions to aid in its clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Baoshan Gao
- Department of Urology II, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Fricke K, Sievi NA, Schmidt FP, Schuurmans MM, Kohler M. Efficacy of surveillance bronchoscopy versus clinically indicated bronchoscopy for detection of acute lung transplant rejection: a systematic review and meta-analysis. ERJ Open Res 2024; 10:00404-2024. [PMID: 39377093 PMCID: PMC11456970 DOI: 10.1183/23120541.00404-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 10/09/2024] Open
Abstract
Background Acute allograft rejection after lung transplantation significantly increases the risk of developing bronchiolitis obliterans syndrome, a form of chronic lung allograft dysfunction and the leading cause of mortality beyond the initial post-transplantation year. There are two diagnostic approaches available for monitoring lung transplant recipients: clinically indicated bronchoscopy (CIB) and surveillance bronchoscopy (SB). The efficacy of both methods and their relative superiority in detecting acute rejection have not been conclusively determined. Methods We systematically searched the MEDLINE, Embase, Cochrane and Scopus databases from inception until 10 October 2023 for prospective studies comparing the efficacy of SB and CIB. Meta-analysis using a random effects model was performed for three observational cohort studies, totalling 122 patients with 527 bronchoscopies. Results Overall, neither SB nor CIB had a higher likelihood of detecting acute lung transplant rejection of any grade. Subsequent subgroup analyses showed no advantage for SB in detecting minimal rejection (grade A1), but an inverse association was observed for higher-grade rejection. Conclusion In conclusion, our study found no significant difference in detecting acute lung transplant rejection between SB and CIB. However, due to the limited number of studies and small sample sizes, larger prospective studies are urgently needed to definitely determine whether there truly exists no difference between SB and CIB in detecting acute rejection, particularly A1 minimal rejection.
Collapse
Affiliation(s)
- Kai Fricke
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | - Malcolm Kohler
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Keller MB, Newman D, Alnababteh M, Ponor L, Shah P, Mathew J, Kong H, Andargie T, Park W, Charya A, Luikart H, Aryal S, Nathan SD, Orens JB, Khush KK, Jang M, Agbor-Enoh S. Extreme elevations of donor-derived cell-free DNA increases the risk of chronic lung allograft dysfunction and death, even without clinical manifestations of disease. J Heart Lung Transplant 2024; 43:1374-1382. [PMID: 38705500 DOI: 10.1016/j.healun.2024.04.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Lung transplant recipients are traditionally monitored with pulmonary function testing (PFT) and lung biopsy to detect post-transplant complications and guide treatment. Plasma donor-derived cell free DNA (dd-cfDNA) is a novel molecular approach of assessing allograft injury, including subclinical allograft dysfunction. The aim of this study was to determine if episodes of extreme molecular injury (EMI) in lung transplant recipients increases the risk of chronic lung allograft dysfunction (CLAD) or death. METHODS This multicenter prospective cohort study included 238 lung transplant recipients. Serial plasma samples were collected for dd-cfDNA measurement by shotgun sequencing. EMI was defined as a dd-cfDNA above the third quartile of levels observed for acute rejection (dd-cfDNA level of ≥5% occurring after 45 days post-transplant). EMI was categorized as Secondary if associated with co-existing acute rejection, infection or PFT decline; or Primary if not associated with these conditions. RESULTS EMI developed in 16% of patients at a median 343.5 (IQR: 177.3-535.5) days post-transplant. Over 50% of EMI episodes were classified as Primary. EMI was associated with an increased risk of severe CLAD or death (HR: 2.78, 95% CI: 1.26-6.22, p = 0.012). The risk remained consistent for the Primary EMI subgroup (HR: 2.34, 95% CI 1.18-4.85, p = 0.015). Time to first EMI episode was a significant predictor of the likelihood of developing CLAD or death (AUC=0.856, 95% CI=0.805-0.908, p < 0.001). CONCLUSIONS Episodes of EMI in lung transplant recipients are often isolated and may not be detectable with traditional clinical monitoring approaches. EMI is associated with an increased risk of severe CLAD or death, independent of concomitant transplant complications.
Collapse
Affiliation(s)
- Michael B Keller
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - David Newman
- College of Nursing, Florida Atlantic University, Boca Raton, Florida
| | - Muhtadi Alnababteh
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Lucia Ponor
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Pali Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Joby Mathew
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Hyesik Kong
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Temesgen Andargie
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Woojin Park
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Ananth Charya
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, Maryland
| | - Helen Luikart
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California; Department of Pathology, Stanford University School of Medicine, Palo Alto, California
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Kiran K Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Moon Jang
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Sean Agbor-Enoh
- Laborarory of Applied Precision Omics (APO) National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland.
| |
Collapse
|
13
|
Messika J, Belousova N, Parquin F, Roux A. Antibody-Mediated Rejection in Lung Transplantation: Diagnosis and Therapeutic Armamentarium in a 21st Century Perspective. Transpl Int 2024; 37:12973. [PMID: 39170865 PMCID: PMC11336419 DOI: 10.3389/ti.2024.12973] [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: 03/11/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024]
Abstract
Humoral immunity is a major waypoint towards chronic allograft dysfunction in lung transplantation (LT) recipients. Though allo-immunization and antibody-mediated rejection (AMR) are well-known entities, some diagnostic gaps need to be addressed. Morphological analysis could be enhanced by digital pathology and artificial intelligence-based companion tools. Graft transcriptomics can help to identify graft failure phenotypes or endotypes. Donor-derived cell free DNA is being evaluated for graft-loss risk stratification and tailored surveillance. Preventative therapies should be tailored according to risk. The donor pool can be enlarged for candidates with HLA sensitization, with strategies combining plasma exchange, intravenous immunoglobulin and immune cell depletion, or with emerging or innovative therapies such as imlifidase or immunoadsorption. In cases of insufficient pre-transplant desensitization, the effects of antibodies on the allograft can be prevented by targeting the complement cascade, although evidence for this strategy in LT is limited. In LT recipients with a humoral response, strategies are combined, including depletion of immune cells (plasmapheresis or immunoadsorption), inhibition of immune pathways, or modulation of the inflammatory cascade, which can be achieved with photopheresis. Altogether, these innovative techniques offer promising perspectives for LT recipients and shape the 21st century's armamentarium against AMR.
Collapse
Affiliation(s)
- Jonathan Messika
- Thoracic Intensive Care Unit, Foch Hospital, Suresnes, France
- Physiopathology and Epidemiology of Respiratory Diseases, UMR1152 INSERM and Université de Paris, Paris, France
- Paris Transplant Group, Paris, France
| | - Natalia Belousova
- Paris Transplant Group, Paris, France
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - François Parquin
- Thoracic Intensive Care Unit, Foch Hospital, Suresnes, France
- Paris Transplant Group, Paris, France
| | - Antoine Roux
- Paris Transplant Group, Paris, France
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| |
Collapse
|
14
|
Keller MB, Sun J, Alnababteh M, Ponor L, D. Shah P, Mathew J, Kong H, Charya A, Luikart H, Aryal S, Nathan SD, Orens JB, Khush KK, Kyoo Jang M, Agbor-Enoh S. Baseline Lung Allograft Dysfunction After Bilateral Lung Transplantation Is Associated With an Increased Risk of Death: Results From a Multicenter Cohort Study. Transplant Direct 2024; 10:e1669. [PMID: 38953039 PMCID: PMC11216668 DOI: 10.1097/txd.0000000000001669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 07/03/2024] Open
Abstract
Background A prior single-center, retrospective cohort study identified baseline lung allograft dysfunction (BLAD) as a risk factor for death in bilateral lung transplant recipients. In this multicenter prospective cohort study, we test the association of BLAD with death in bilateral lung transplant recipients, identify clinical risk factors for BLAD, and assess its association with allograft injury on the molecular level. Methods This multicenter, prospective cohort study included 173 bilateral lung transplant recipients that underwent serial pulmonary function testing and plasma collection for donor-derived cell-free DNA at prespecified time points. BLAD was defined as failure to achieve ≥80% predicted for both forced expiratory volume in 1 s and forced vital capacity after lung transplant, on 2 consecutive measurements at least 3 mo apart. Results BLAD was associated with increased risk of death (hazard ratio, 1.97; 95% confidence interval [CI], 1.05-3.69; P = 0.03) but not chronic lung allograft dysfunction alone (hazard ratio, 1.60; 95% CI, 0.87-2.95; P = 0.13). Recipient obesity (odds ratio, 1.69; 95% CI, 1.15-2.80; P = 0.04) and donor age (odds ratio, 1.03; 95% CI, 1.02-1.05; P = 0.004) increased the risk of developing BLAD. Patients with BLAD did not demonstrate higher log10(donor-derived cell-free DNA) levels compared with no BLAD (slope [SE]: -0.0095 [0.0007] versus -0.0109 [0.0007]; P = 0.15). Conclusions BLAD is associated with an increased risk of death following lung transplantation, representing an important posttransplant outcome with valuable prognostic significance; however, early allograft specific injury on the molecular level does not increase the risk of BLAD, supporting further mechanistic insight into disease pathophysiology.
Collapse
Affiliation(s)
- Michael B. Keller
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Junfeng Sun
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Muhtadi Alnababteh
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Lucia Ponor
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Pali D. Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Joby Mathew
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Hyesik Kong
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
| | - Ananth Charya
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD
| | - Helen Luikart
- Genome Transplant Genomics (GTD), Stanford University School of Medicine, Palo Alto, CA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Steven D. Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Jonathan B. Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Kiran K. Khush
- Genome Transplant Genomics (GTD), Stanford University School of Medicine, Palo Alto, CA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Moon Kyoo Jang
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
| | - Sean Agbor-Enoh
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
15
|
Nikolova A, Agbor-Enoh S, Bos S, Crespo-Leiro M, Ensminger S, Jimenez-Blanco M, Minervini A, Perch M, Segovia J, Vos R, Khush K, Potena L. European Society for Organ Transplantation (ESOT) Consensus Statement on the Use of Non-invasive Biomarkers for Cardiothoracic Transplant Rejection Surveillance. Transpl Int 2024; 37:12445. [PMID: 38962472 PMCID: PMC11221358 DOI: 10.3389/ti.2024.12445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/04/2024] [Indexed: 07/05/2024]
Abstract
While allograft rejection (AR) continues to threaten the success of cardiothoracic transplantation, lack of accurate and repeatable surveillance tools to diagnose AR is a major unmet need in the clinical management of cardiothoracic transplant recipients. Endomyocardial biopsy (EMB) and transbronchial biopsy (TBBx) have been the cornerstone of rejection monitoring since the field's incipience, but both suffer from significant limitations, including poor concordance of biopsy interpretation among pathologists. In recent years, novel molecular tools for AR monitoring have emerged and their performance characteristics have been evaluated in multiple studies. An international working group convened by ESOT has reviewed the existing literature and provides a series of recommendations to guide the use of these biomarkers in clinical practice. While acknowledging some caveats, the group recognized that Gene-expression profiling and donor-derived cell-free DNA (dd-cfDNA) may be used to rule out rejection in heart transplant recipients, but they are not recommended for cardiac allograft vasculopathy screening. Other traditional biomarkers (NT-proBNP, BNP or troponin) do not have sufficient evidence to support their use to diagnose AR. Regarding lung transplant, dd-cfDNA could be used to rule out clinical rejection and infection, but its use to monitor treatment response is not recommended.
Collapse
Affiliation(s)
- Andriana Nikolova
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT) and Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, United States
- Lung Transplantation, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle uponTyne, United Kingdom
- Institute of Transplantation, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle uponTyne, United Kingdom
| | - Marisa Crespo-Leiro
- Cardiology Department, Complexo Hospitalario Universitario A Coruna (CHUAC), Instituto de Investigación Biomédica A Coruña (INIBIC), Universitade de Coruna (UDC), Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares/Network Biomedical Research Center—Cardiovascular Diseases (CIBERCV), La Coruna, Spain
| | - Stephan Ensminger
- Klinik für Herz- und Thorakale Gefäßchirurgie, Universitäres Herzzentrum Lübeck, Lübeck, Germany
| | - Marta Jimenez-Blanco
- Cardiology Department, University Hospital Ramón y Cajal (Madrid), Centro de Investigación Biomedica en Red—Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Annamaria Minervini
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michael Perch
- Department of Cardiology, Section for Lung Transplantation, Righospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Javier Segovia
- Cardiology Department, Puerta de Hierro Majadahonda University Hospital, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana/Puerta de Hierro Health Research Institute—Segovia de Arana (IDIPHISA), Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares/Network Biomedical Research Center—Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Robin Vos
- Department of Respiratory Diseases, UZ Leuven, and Lung Transplant Unit, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Kiran Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Luciano Potena
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
16
|
Wannes Daou A, Wallace C, Barker M, Ambrosino T, Towe C, Morales DLS, Wikenheiser-Brokamp KA, Hayes D, Burg G. Flexible bronchoscopy in pediatric lung transplantation. Pediatr Transplant 2024; 28:e14757. [PMID: 38695266 DOI: 10.1111/petr.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/09/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024]
Abstract
Pediatric lung transplantation represents a treatment option for children with advanced lung disease or pulmonary vascular disorders who are deemed an appropriate candidate. Pediatric flexible bronchoscopy is an important and evolving field that is highly relevant in the pediatric lung transplant population. It is thus important to advance our knowledge to better understand how care for children after lung transplant can be maximally optimized using pediatric bronchoscopy. Our goals are to continually improve procedural skills when performing bronchoscopy and to decrease the complication rate while acquiring adequate samples for diagnostic evaluation. Attainment of these goals is critical since allograft assessment by bronchoscopic biopsy is required for histological diagnosis of acute cellular rejection and is an important contributor to establishing chronic lung allograft dysfunction, a common complication after lung transplant. Flexible bronchoscopy with bronchoalveolar lavage and transbronchial lung biopsy plays a key role in lung transplant graft assessment. In this article, we discuss the application of bronchoscopy in pediatric lung transplant evaluation including historical approaches, our experience, and future directions not only in bronchoscopy but also in the evolving pediatric lung transplantation field. Pediatric flexible bronchoscopy has become a vital modality for diagnosing lung transplant complications in children as well as assessing therapeutic responses. Herein, we review the value of flexible bronchoscopy in the management of children after lung transplant and discuss the application of novel techniques to improve care for this complex pediatric patient population and we provide a brief update about new diagnostic techniques applied in the growing lung transplantation field.
Collapse
Affiliation(s)
- Antoinette Wannes Daou
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carolyn Wallace
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mitzi Barker
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Teresa Ambrosino
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Christopher Towe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - David L S Morales
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Don Hayes
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Gregory Burg
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Paraskeva MA, Snell GI. Advances in lung transplantation: 60 years on. Respirology 2024; 29:458-470. [PMID: 38648859 DOI: 10.1111/resp.14721] [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: 02/11/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
Lung transplantation is a well-established treatment for advanced lung disease, improving survival and quality of life. Over the last 60 years all aspects of lung transplantation have evolved significantly and exponential growth in transplant volume. This has been particularly evident over the last decade with a substantial increase in lung transplant numbers as a result of innovations in donor utilization procurement, including the use donation after circulatory death and ex-vivo lung perfusion organs. Donor lungs have proved to be surprisingly robust, and therefore the donor pool is actually larger than previously thought. Parallel to this, lung transplant outcomes have continued to improve with improved acute management as well as microbiological and immunological insights and innovations. The management of lung transplant recipients continues to be complex and heavily dependent on a tertiary care multidisciplinary paradigm. Whilst long term outcomes continue to be limited by chronic lung allograft dysfunction improvements in diagnostics, mechanistic understanding and evolutions in treatment paradigms have all contributed to a median survival that in some centres approaches 10 years. As ongoing studies build on developing novel approaches to diagnosis and treatment of transplant complications and improvements in donor utilization more individuals will have the opportunity to benefit from lung transplantation. As has always been the case, early referral for transplant consideration is important to achieve best results.
Collapse
Affiliation(s)
- Miranda A Paraskeva
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Gregory I Snell
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Keller MB, Tian X, Jang MK, Meda R, Charya A, Berry GJ, Marboe CC, Kong H, Ponor IL, Aryal S, Orens JB, Shah PD, Nathan SD, Agbor-Enoh S. Higher Molecular Injury at Diagnosis of Acute Cellular Rejection Increases the Risk of Lung Allograft Failure: A Clinical Trial. Am J Respir Crit Care Med 2024; 209:1238-1245. [PMID: 38190701 PMCID: PMC11146548 DOI: 10.1164/rccm.202305-0798oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024] Open
Abstract
Rationale: The association of acute cellular rejection (ACR) with chronic lung allograft dysfunction (CLAD) in lung transplant recipients has primarily been described before consensus recommendations incorporating restrictive phenotypes. Furthermore, the association of the degree of molecular allograft injury during ACR with CLAD or death remains undefined. Objectives: To investigate the association of ACR with the risk of CLAD or death and to further investigate if this risk depends on the degree of molecular allograft injury. Methods: This multicenter, prospective cohort study included 188 lung transplant recipients. Subjects underwent serial plasma collections for donor-derived cell-free DNA (dd-cfDNA) at prespecified time points and bronchoscopy. Multivariable Cox proportional-hazards analysis was conducted to analyze the association of ACR with subsequent CLAD or death as well as the association of dd-cfDNA during ACR with risk of CLAD or death. Additional outcomes analyses were performed with episodes of ACR categorized as "high risk" (dd-cfDNA ⩾ 1%) and "low risk" (dd-cfDNA < 1%). Measurements and Main Results: In multivariable analysis, ACR was associated with the composite outcome of CLAD or death (hazard ratio [HR], 2.07 [95% confidence interval (CI), 1.05-4.10]; P = 0.036). Elevated dd-cfDNA ⩾ 1% at ACR diagnosis was independently associated with increased risk of CLAD or death (HR, 3.32; 95% CI, 1.31-8.40; P = 0.012). Patients with high-risk ACR were at increased risk of CLAD or death (HR, 3.13; 95% CI, 1.41-6.93; P = 0.005), whereas patients with low-risk status ACR were not. Conclusions: Patients with ACR are at higher risk of CLAD or death, but this may depend on the degree of underlying allograft injury at the molecular level. Clinical trial registered with www.clinicaltrials.gov (NCT02423070).
Collapse
Affiliation(s)
- Michael B. Keller
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Laboratory of Applied Precision Omics and
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Xin Tian
- Office of Biostatistics Research, NHLBI, NIH, Bethesda, Maryland
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Laboratory of Applied Precision Omics and
| | - Rohan Meda
- Laboratory of Applied Precision Omics and
| | - Ananth Charya
- University of Maryland Medical Center, Baltimore, Maryland
| | - Gerald J. Berry
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- School of Medicine, Stanford University, Stanford, California
| | - Charles C. Marboe
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Laboratory of Applied Precision Omics and
| | - Ileana L. Ponor
- Department of Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland; and
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Jonathan B. Orens
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Pali D. Shah
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Steven D. Nathan
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Laboratory of Applied Precision Omics and
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Calabrese DR, Shaver CM. The Emperor Has Some Clothes: Emerging Molecular Diagnostics in Post-Lung Transplant Decision-Making. Am J Respir Crit Care Med 2024; 209:1182-1184. [PMID: 38387008 PMCID: PMC11146531 DOI: 10.1164/rccm.202401-0059ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/22/2024] [Indexed: 02/24/2024] Open
Affiliation(s)
- Daniel R Calabrese
- Department of Medicine University of California, San Francisco San Francisco, California
- San Francisco Veterans Affairs Medical Center San Francisco, California
| | - Ciara M Shaver
- Department of Medicine Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
20
|
Pavlisko EN, Adam BA, Berry GJ, Calabrese F, Cortes-Santiago N, Glass CH, Goddard M, Greenland JR, Kreisel D, Levine DJ, Martinu T, Verleden SE, Weigt SS, Roux A. The 2022 Banff Meeting Lung Report. Am J Transplant 2024; 24:542-548. [PMID: 37931751 DOI: 10.1016/j.ajt.2023.10.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
The Lung Session of the 2022 16th Banff Foundation for Allograft Pathology Conference-held in Banff, Alberta-focused on non-rejection lung allograft pathology and novel technologies for the detection of allograft injury. A multidisciplinary panel reviewed the state-of-the-art of current histopathologic entities, serologic studies, and molecular practices, as well as novel applications of digital pathology with artificial intelligence, gene expression analysis, and quantitative image analysis of chest computerized tomography. Current states of need as well as prospective integration of the aforementioned tools and technologies for complete assessment of allograft injury and its impact on lung transplant outcomes were discussed. Key conclusions from the discussion were: (1) recognition of limitations in current standard of care assessment of lung allograft dysfunction; (2) agreement on the need for a consensus regarding the standardized approach to the collection and assessment of pathologic data, inclusive of all lesions associated with graft outcome (eg, non-rejection pathology); and (3) optimism regarding promising novel diagnostic modalities, especially minimally invasive, which should be integrated into large, prospective multicenter studies to further evaluate their utility in clinical practice for directing personalized therapies to improve graft outcomes.
Collapse
Affiliation(s)
- Elizabeth N Pavlisko
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA.
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Gerald J Berry
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Nahir Cortes-Santiago
- Department of Pathology and Immunology, Texas Children's Hospital, Houston, Texas, USA
| | - Carolyn H Glass
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Martin Goddard
- Pathology Department, Royal Papworth Hospital, NHS Trust, Papworth Everard, Cambridge, UK
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, USA; Veterans Affairs Health Care System, San Francisco, California, USA
| | - Daniel Kreisel
- Department of Surgery, Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Deborah J Levine
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, California, USA
| | - Tereza Martinu
- Division of Respirology, Department of Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada; Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Stijn E Verleden
- Lung Transplant Unit, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of ASTARC, University of Antwerp, Wilrijk, Belgium
| | - S Sam Weigt
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Antoine Roux
- Department of Respiratory Medicine, Foch Hospital, Suresnes, France
| |
Collapse
|
21
|
Keller MB, Tian X, Jang MK, Meda R, Charya A, Ozisik D, Berry GJ, Marboe CC, Kong H, Ponor IL, Aryal S, Orens JB, Shah PD, Nathan SD, Agbor-Enoh S. Organizing pneumonia is associated with molecular allograft injury and the development of antibody-mediated rejection. J Heart Lung Transplant 2024; 43:563-570. [PMID: 37972825 DOI: 10.1016/j.healun.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/28/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The association between organizing pneumonia (OP) after lung transplantation with the development of acute rejection (AR) remains undefined. In addition, molecular allograft injury, as measured by donor-derived cell-free DNA (dd-cfDNA), during episodes of OP and its relationship to episodes of AR, chronic lung allograft dysfunction (CLAD), or death is unknown. METHODS This multicenter, prospective cohort study collected serial plasma samples from 188 lung transplant recipients for dd-cfDNA at the time of bronchoscopy with biopsy. Multivariable Cox regression was used to analyze the association between OP with the development of AR (antibody-mediated rejection (AMR) and acute cellular rejection (ACR)), CLAD, and death. Multivariable models were performed to test the association of dd-cfDNA at OP with the risk of AR, CLAD, or death. RESULTS In multivariable analysis, OP was associated with increased risk of AMR (hazard ratio (HR) = 2.26, 95% confidence interval (CI) 1.04-4.92, p = 0.040) but not ACR (HR = 1.29, 95% CI: 0.66-2.5, p = 0.45) or the composite outcome of CLAD or death (HR = 0.88, 95% CI, 0.47-1.65, p = 0.69). Median levels of dd-cfDNA were higher in OP compared to stable controls (1.33% vs 0.43%, p = 0.0006). Multivariable analysis demonstrated that levels of dd-cfDNA at diagnosis of OP were associated with increased risk of both AMR (HR = 1.29, 95% CI 1.03-1.62, p = 0.030) and death (HR = 1.16, 95% CI, 1.02-1.31, p = 0.026). CONCLUSIONS OP is independently associated with an increased risk of AMR but not CLAD or death. The degree of molecular allograft injury at the diagnosis of OP may further predict the risk of AMR and death.
Collapse
Affiliation(s)
- Michael B Keller
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Rohan Meda
- Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ananth Charya
- University of Maryland Medical Center, Baltimore, Maryland
| | - Deniz Ozisik
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Gerald J Berry
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Stanford University School of Medicine, Stanford, California
| | - Charles C Marboe
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ileana L Ponor
- Department of Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Pali D Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
22
|
Balasubramanian S, Richert ME, Kong H, Fu S, Jang MK, Andargie TE, Keller MB, Alnababteh M, Park W, Apalara Z, Sun J, Redekar N, Orens J, Aryal S, Bush EL, Cantu E, Diamond J, Shah P, Yu K, Nathan SD, Agbor-Enoh S. Cell-Free DNA Maps Tissue Injury and Correlates with Disease Severity in Lung Transplant Candidates. Am J Respir Crit Care Med 2024; 209:727-737. [PMID: 38117233 PMCID: PMC10945061 DOI: 10.1164/rccm.202306-1064oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023] Open
Abstract
Rationale: Plasma cell-free DNA levels correlate with disease severity in many conditions. Pretransplant cell-free DNA may risk stratify lung transplant candidates for post-transplant complications. Objectives: To evaluate if pretransplant cell-free DNA levels and tissue sources identify patients at high risk of primary graft dysfunction and other pre- and post-transplant outcomes. Methods: This multicenter, prospective cohort study recruited 186 lung transplant candidates. Pretransplant plasma samples were collected to measure cell-free DNA. Bisulfite sequencing was performed to identify the tissue sources of cell-free DNA. Multivariable regression models determined the association between cell-free DNA levels and the primary outcome of primary graft dysfunction and other transplant outcomes, including Lung Allocation Score, chronic lung allograft dysfunction, and death. Measurements and Main Results: Transplant candidates had twofold greater cell-free DNA levels than healthy control patients (median [interquartile range], 23.7 ng/ml [15.1-35.6] vs. 12.9 ng/ml [9.9-18.4]; P < 0.0001), primarily originating from inflammatory innate immune cells. Cell-free DNA levels and tissue sources differed by native lung disease category and correlated with the Lung Allocation Score (P < 0.001). High pretransplant cell-free DNA increased the risk of primary graft dysfunction (odds ratio, 1.60; 95% confidence interval [CI], 1.09-2.46; P = 0.0220), and death (hazard ratio, 1.43; 95% CI, 1.07-1.92; P = 0.0171) but not chronic lung allograft dysfunction (hazard ratio, 1.37; 95% CI, 0.97-1.94; P = 0.0767). Conclusions: Lung transplant candidates demonstrate a heightened degree of tissue injury with elevated cell-free DNA, primarily originating from innate immune cells. Pretransplant plasma cell-free DNA levels predict post-transplant complications.
Collapse
Affiliation(s)
- Shanti Balasubramanian
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland
| | - Mary E. Richert
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sheng Fu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Temesgen E. Andargie
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Department of Biology, Howard University, Washington, District of Columbia
| | - Michael B. Keller
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Muhtadi Alnababteh
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland
| | - Woojin Park
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Zainab Apalara
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Integrated Data Science Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jian Sun
- Integrated Data Science Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Neelam Redekar
- Integrated Data Science Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jonathan Orens
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Errol L. Bush
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Edward Cantu
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua Diamond
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pali Shah
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Steven D. Nathan
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation, Bethesda, Maryland
- Division of Intramural Research, Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
23
|
Trindade AJ. A Hospitable Home? Cell-Free DNA and the Inflammatory Milieu in Lung Transplant Candidates. Am J Respir Crit Care Med 2024; 209:627-628. [PMID: 38226872 PMCID: PMC10945066 DOI: 10.1164/rccm.202312-2242ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Affiliation(s)
- Anil J Trindade
- Division of Allergy, Pulmonary, and Critical Care Medicine and Vanderbilt Transplant Center Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
24
|
Pradère P, Zajacova A, Bos S, Le Pavec J, Fisher A. Molecular monitoring of lung allograft health: is it ready for routine clinical use? Eur Respir Rev 2023; 32:230125. [PMID: 37993125 PMCID: PMC10663940 DOI: 10.1183/16000617.0125-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 11/24/2023] Open
Abstract
Maintenance of long-term lung allograft health in lung transplant recipients (LTRs) requires a fine balancing act between providing sufficient immunosuppression to reduce the risk of rejection whilst at the same time not over-immunosuppressing individuals and exposing them to the myriad of immunosuppressant drug side-effects that can cause morbidity and mortality. At present, lung transplant physicians only have limited and rather blunt tools available to assist them with this task. Although therapeutic drug monitoring provides clinically useful information about single time point and longitudinal exposure of LTRs to immunosuppressants, it lacks precision in determining the functional level of immunosuppression that an individual is experiencing. There is a significant gap in our ability to monitor lung allograft health and therefore tailor optimal personalised immunosuppression regimens. Molecular diagnostics performed on blood, bronchoalveolar lavage or lung tissue that can detect early signs of subclinical allograft injury, differentiate rejection from infection or distinguish cellular from humoral rejection could offer clinicians powerful tools in protecting lung allograft health. In this review, we look at the current evidence behind molecular monitoring in lung transplantation and ask if it is ready for routine clinical use. Although donor-derived cell-free DNA and tissue transcriptomics appear to be the techniques with the most immediate clinical potential, more robust data are required on their performance and additional clinical value beyond standard of care.
Collapse
Affiliation(s)
- Pauline Pradère
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Department of Respiratory Diseases, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph and Paris Saclay University, Paris, France
| | - Andrea Zajacova
- Prague Lung Transplant Program, Department of Pneumology, Motol University Hospital and 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Institute of Transplantation, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Jérôme Le Pavec
- Department of Respiratory Diseases, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph and Paris Saclay University, Paris, France
| | - Andrew Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Institute of Transplantation, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
25
|
Li Y, Liang B. Circulating donor-derived cell-free DNA as a marker for rejection after lung transplantation. Front Immunol 2023; 14:1263389. [PMID: 37885888 PMCID: PMC10598712 DOI: 10.3389/fimmu.2023.1263389] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Objective Recently, circulating donor-derive cell free DNA (dd-cfDNA) has gained growing attention in the field of solid organ transplantation. The aim of the study was to analyze circulating dd-cfDNA levels in graft rejection, ACR and AMR separately for each rejection type compared with non-rejection, and assessed the diagnostic potential of dd-cfDNA levels in predicting graft rejection after lung transplantation. Methods A systematic search for relevant articles was conducted on Medline, Web of Science, China National Knowledge Infrastructure (CNKI), and Wanfang databases without restriction of languages. The search date ended on June 1, 2023. STATA software was used to analyze the difference between graft rejection, ACR, AMR and stable controls, and evaluate the diagnostic performance of circulating dd-cfDNA in detecting graft rejection. Results The results indicated that circulating dd-cfDNA levels in graft rejection, ACR, and AMR were significantly higher than non-rejection (graft rejection: SMD=1.78, 95% CI: 1.31-2.25, I2 = 88.6%, P< 0.001; ACR: SMD=1.03, 95% CI: 0.47-1.59, I2 = 89.0%, P < 0.001; AMR: SMD= 1.78, 95% CI: 1.20-2.35, I2 = 89.8%, P < 0.001). Circulating dd-cfDNA levels distinguished graft rejection from non-rejection with a pooled sensitivity of 0.87 (95% CI: 0.80-0.92) and a pooled specificity of 0.82 (95% CI: 0.76-0.86). The corresponding SROC yield an AUROC of 0.90 (95% CI: 0.87-0.93). Conclusion Circulating dd-cfDNA could be used as a non-invasive biomarker to distinguish the patients with graft rejection from normal stable controls. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023440467.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Laboratory Medical Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Bin Liang
- Bioinformatics of Department, Key laboratory of Cell Biology, School of Life Sciences, China Medical University, Shenyang, China
| |
Collapse
|
26
|
Brandon W, Dunn C, Bollineni S, Joerns J, Lawrence A, Mohanka M, Timofte I, Torres F, Kaza V. Management of donor-specific antibodies in lung transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1248284. [PMID: 38993917 PMCID: PMC11235237 DOI: 10.3389/frtra.2023.1248284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/23/2023] [Indexed: 07/13/2024]
Abstract
The formation of antibodies against donor human leukocyte antigens poses a challenging problem both for donor selection as well as postoperative graft function in lung transplantation. These donor-specific antibodies limit the pool of potential donor organs and are associated with episodes of antibody-mediated rejection, chronic lung allograft dysfunction, and increased mortality. Optimal management strategies for clearance of DSAs are poorly defined and vary greatly by institution; most of the data supporting any particular strategy is limited to small-scale retrospective cohort studies. A typical approach to antibody depletion may involve the use of high-dose steroids, plasma exchange, intravenous immunoglobulin, and possibly other immunomodulators or small-molecule therapies. This review seeks to define the current understanding of the significance of DSAs in lung transplantation and outline the literature supporting strategies for their management.
Collapse
Affiliation(s)
- William Brandon
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Colin Dunn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Srinivas Bollineni
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John Joerns
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Adrian Lawrence
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Manish Mohanka
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Irina Timofte
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fernando Torres
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Vaidehi Kaza
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
27
|
Raszeja-Wyszomirska J, Macech M, Kolanowska M, Krawczyk M, Nazarewski S, Wójcicka A, Małyszko J. Free-Circulating Nucleic Acids as Biomarkers in Patients After Solid Organ Transplantation. Ann Transplant 2023; 28:e939750. [PMID: 37580899 PMCID: PMC10439677 DOI: 10.12659/aot.939750] [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: 02/07/2023] [Accepted: 05/17/2023] [Indexed: 08/16/2023] Open
Abstract
A number types of extracellular DNA (eg, cell-free, cfDNA) circulate in human blood, including mitochondrial, transcriptome, and regulatory DNA, usually at low concentrations. Larger amounts of cfDNA appear in any inflammatory condition, including organ damage due to a variety of reasons. The role of cfDNA in solid organ transplantation is discussed in this review as a valuable additional tool in the standard of care of transplant patients. Post-transplant monitoring requires the use of high-quality biomarkers for early detection of graft damage or rejection to be able to apply early therapeutic intervention. CfDNA complements the traditional monitoring strategies, being a risk stratification tool and an important prognostic marker. However, improving the sensitivity and specificity of cfDNA detection is necessary to facilitate personalized patient management, warranting further research in terms of measurement, test standardization, and storage, processing, and shipping. A diagnostic test (Allosure, CareDx, Inc., Brisbane, CA) for kidney, heart and lung transplant patients is now commercially available, and validation for other organs (eg, liver) is pending. To date, donor-derived cfDNA in combination with other biomarkers appears to be a promising tool in graft rejection as it is minimally invasive, time-sensitive, and cost-effective. However, improvement of sensitivity and specificity is required to facilitate personalized patient management. Whether it could be an alternate to graft biopsy remains unclear.
Collapse
Affiliation(s)
- Joanna Raszeja-Wyszomirska
- Department of Hepatology, Transplantology, and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Michał Macech
- Department of General, Vascular, and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | | | - Marek Krawczyk
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Nazarewski
- Department of General, Vascular, and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | | | - Jolanta Małyszko
- Department of Nephrology, Dialysis, and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
28
|
Trindade AJ, Chapin KC, Gray JN, Furuya Y, Mullican A, Hoy H, Demarest CT, Robbins IM, Bacchetta M, Erasmus DB, Shaver CM. Relative Change in Donor-Derived Cell-free DNA is Superior to Absolute Values for Diagnosis of Acute Lung Allograft Dysfunction. Transplant Direct 2023; 9:e1487. [PMID: 37250487 PMCID: PMC10212612 DOI: 10.1097/txd.0000000000001487] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/31/2023] Open
Abstract
Donor-derived cell-free DNA (dd-cfDNA%) is a biomarker of early acute lung allograft dysfunction (ALAD), with a value of ≥1.0% indicating injury. Whether dd-cfDNA% is a useful biomarker in patients >2 y posttransplant is unknown. Our group previously demonstrated that median dd-cfDNA% in lung recipients ≥2 y posttransplant without ALAD was 0.45%. In that cohort, biologic variability of dd-cfDNA% was estimated by a reference change value (RCV) of 73%, suggesting that change exceeding 73% may be pathologic. In this study, we aimed to determine whether dd-cfDNA% variability or absolute thresholds are optimal for detecting ALAD. Methods We prospectively measured plasma dd-cfDNA% every 3 to 4 mo in patients ≥2 y post-lung transplant. ALAD was defined as infection, acute cellular rejection, possible antibody-mediated rejection, or change in forced expiratory volume in 1 s >10%, and was adjudicated retrospectively. We analyzed area under the curve for RCV and absolute dd-cfDNA% and reported performance of RCV ≥73% versus absolute value >1% for discriminating ALAD. Results Seventy-one patients had ≥2 baseline measurements of dd-cfDNA%; 30 developed ALAD. RCV of dd-cfDNA% at ALAD had a greater area under the receiver operator characteristic curve than absolute dd-cfDNA% values (0.87 versus 0.69, P = 0.018). Test characteristics of RCV >73% for ALAD diagnosis were sensitivity 87%, specificity 78%, positive predictive value 74%, and negative predictive value 89%. In contrast, dd-cfDNA% ≥1% had sensitivity 50%, specificity 78%, positive predictive value 63%, and negative predictive value 68%. Conclusions Relative change in dd-cfDNA% has improved test characteristics for diagnosing ALAD compared with absolute values.
Collapse
Affiliation(s)
- Anil J. Trindade
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Nashville, TN
| | | | | | | | - Amy Mullican
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Haley Hoy
- Vanderbilt Transplant Center, Nashville, TN
| | - Caitlin T. Demarest
- Vanderbilt Transplant Center, Nashville, TN
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Ivan M. Robbins
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Nashville, TN
| | - Matthew Bacchetta
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, TN
| | - David B. Erasmus
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Nashville, TN
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Nashville, TN
| |
Collapse
|
29
|
Scarpa JR, Elemento O. Multi-omic molecular profiling and network biology for precision anaesthesiology: a narrative review. Br J Anaesth 2023:S0007-0912(23)00125-3. [PMID: 37055274 DOI: 10.1016/j.bja.2023.03.006] [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: 11/11/2022] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 04/15/2023] Open
Abstract
Technological advancement, data democratisation, and decreasing costs have led to a revolution in molecular biology in which the entire set of DNA, RNA, proteins, and various other molecules - the 'multi-omic' profile - can be measured in humans. Sequencing 1 million bases of human DNA now costs US$0.01, and emerging technologies soon promise to reduce the cost of sequencing the whole genome to US$100. These trends have made it feasible to sample the multi-omic profile of millions of people, much of which is publicly available for medical research. Can anaesthesiologists use these data to improve patient care? This narrative review brings together a rapidly growing literature in multi-omic profiling across numerous fields that points to the future of precision anaesthesiology. Here, we discuss how DNA, RNA, proteins, and other molecules interact in molecular networks that can be used for preoperative risk stratification, intraoperative optimisation, and postoperative monitoring. This literature provides evidence for four fundamental insights: (1) Clinically similar patients have different molecular profiles and, as a consequence, different outcomes. (2) Vast, publicly available, and rapidly growing molecular datasets have been generated in chronic disease patients and can be repurposed to estimate perioperative risk. (3) Multi-omic networks are altered in the perioperative period and influence postoperative outcomes. (4) Multi-omic networks can serve as empirical, molecular measurements of a successful postoperative course. With this burgeoning universe of molecular data, the anaesthesiologist-of-the-future will tailor their clinical management to an individual's multi-omic profile to optimise postoperative outcomes and long-term health.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
30
|
Osmanodja B, Akifova A, Oellerich M, Beck J, Bornemann-Kolatzki K, Schütz E, Budde K. Donor-Derived Cell-Free DNA for Kidney Allograft Surveillance after Conversion to Belatacept: Prospective Pilot Study. J Clin Med 2023; 12:jcm12062437. [PMID: 36983437 PMCID: PMC10051604 DOI: 10.3390/jcm12062437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Donor-derived cell-free DNA (dd-cfDNA) is used as a biomarker for detection of antibody-mediated rejection (ABMR) and other forms of graft injury. Another potential indication is guidance of immunosuppressive therapy when no therapeutic drug monitoring is available. In such situations, detection of patients with overt or subclinical graft injury is important to personalize immunosuppression. We prospectively measured dd-cfDNA in 22 kidney transplant recipients (KTR) over a period of 6 months after conversion to belatacept for clinical indication and assessed routine clinical parameters. Patient and graft survival was 100% after 6 months, and eGFR remained stable (28.7 vs. 31.1 mL/min/1.73 m2, p = 0.60). Out of 22 patients, 2 (9%) developed biopsy-proven rejection-one episode of low-grade TCMR IA and one episode of caABMR. While both episodes were detected by increase in creatinine, the caABMR episode led to increase in absolute dd-cfDNA (168 copies/mL) above the cut-off of 50 copies/mL, while the TCMR episode did show slightly increased relative dd-cfDNA (0.85%) despite normal absolute dd-cfDNA (22 copies/mL). Dd-cfDNA did not differ before and after conversion in a subgroup of 12 KTR with previous calcineurin inhibitor therapy and no rejection (12.5 vs. 25.3 copies/mL, p = 0.34). In this subgroup, 3/12 (25%) patients showed increase of absolute dd-cfDNA above the prespecified cut-off (50 copies/mL) despite improving eGFR. Increase in dd-cfDNA after conversion to belatacept is common and could point towards subclinical allograft injury. To detect subclinical TCMR changes without vascular lesions, additional biomarkers or urinary dd-cfDNA should complement plasma dd-cfDNA. Resolving CNI toxicity is unlikely to be detected by decreased dd-cfDNA levels. In summary, the sole determination of dd-cfDNA has limited utility in the guidance of patients after late conversion to belatacept. Further studies should focus on patients undergoing early conversion and include protocol biopsies at least for patients with increased dd-cfDNA.
Collapse
Affiliation(s)
- Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Aylin Akifova
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Oellerich
- Department of Clinical Pharmacology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Julia Beck
- Chronix Biomedical GmbH, 37073 Göttingen, Germany
| | | | | | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
31
|
Halverson LP, Hachem RR. Antibody-Mediated Rejection: Diagnosis and Treatment. Clin Chest Med 2023; 44:95-103. [PMID: 36774172 PMCID: PMC10148231 DOI: 10.1016/j.ccm.2022.10.008] [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: 02/11/2023]
Abstract
Antibody-mediated rejection (AMR) is a form of lung allograft rejection that is emerging as an important risk factor for chronic lung allograft dysfunction and decreased long-term survival. In this review, we provide a brief overview of our current understanding of its pathophysiology with an emphasis on donor-specific antibodies before moving on to focus on the current diagnostic criteria and treatment strategies. Our goal is to discuss the limitations of our current knowledge and explore how novel diagnostic and therapeutic options aim to improve outcomes through earlier definitive diagnosis and preemptive targeted treatment.
Collapse
Affiliation(s)
- Laura P Halverson
- Division of Pulmonary & Critical Care, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8052, Saint Louis, MO 63108, USA.
| | - Ramsey R Hachem
- Division of Pulmonary & Critical Care, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8052, Saint Louis, MO 63108, USA
| |
Collapse
|
32
|
Biomarkers for Chronic Lung Allograft Dysfunction: Ready for Prime Time? Transplantation 2023; 107:341-350. [PMID: 35980878 PMCID: PMC9875844 DOI: 10.1097/tp.0000000000004270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle impairing lung transplant outcome. Parallel to the better clinical identification and characterization of CLAD and CLAD phenotypes, there is an increasing urge to find adequate biomarkers that could assist in the earlier detection and differential diagnosis of CLAD phenotypes, as well as disease prognostication. The current status and state-of-the-art of biomarker research in CLAD will be discussed with a particular focus on radiological biomarkers or biomarkers found in peripheral tissue, bronchoalveolar lavage' and circulating blood' in which significant progress has been made over the last years. Ultimately, although a growing number of biomarkers are currently being embedded in the follow-up of lung transplant patients, it is clear that one size does not fit all. The future of biomarker research probably lies in the rigorous combination of clinical information with findings in tissue, bronchoalveolar lavage' or blood. Only by doing so, the ultimate goal of biomarker research can be achieved, which is the earlier identification of CLAD before its clinical manifestation. This is desperately needed to improve the prognosis of patients with CLAD after lung transplantation.
Collapse
|
33
|
Charya AV, Ponor IL, Cochrane A, Levine D, Philogene M, Fu YP, Jang MK, Kong H, Shah P, Bon AM, Krishnan A, Mathew J, Luikart H, Khush KK, Berry G, Marboe C, Iacono A, Orens JB, Nathan SD, Agbor-Enoh S. Clinical features and allograft failure rates of pulmonary antibody-mediated rejection categories. J Heart Lung Transplant 2023; 42:226-235. [PMID: 36319530 DOI: 10.1016/j.healun.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Pulmonary antibody-mediated rejection (AMR) consensus criteria categorize AMR by diagnostic certainty. This study aims to define the clinical features and associated outcomes of these recently defined AMR categories. METHODS Adjudication committees reviewed clinical data of 335 lung transplant recipients to define clinical or subclinical AMR based on the presence of allograft dysfunction, and the primary endpoints, time from transplant to allograft failure, a composite endpoint of chronic lung allograft dysfunction and/or death. Clinical AMR was subcategorized based on diagnostic certainty as definite, probable or possible AMR if 4, 3, or 2 characteristic features were present, respectively. Allograft injury was assessed via plasma donor-derived cell-free DNA (ddcfDNA). Risk of allograft failure and allograft injury was compared for AMR categories using regression models. RESULTS Over the 38.5 months follow-up, 28.7% of subjects developed clinical AMR (n = 96), 18.5% developed subclinical AMR (n = 62) or 58.3% were no AMR (n = 177). Clinical AMR showed higher risk of allograft failure and ddcfDNA levels compared to subclinical or no AMR. Clinical AMR included definite/probable (n = 21) or possible AMR (n = 75). These subcategories showed similar clinical characteristics, ddcfDNA levels, and risk of allograft failure. However, definite/probable AMR showed greater measures of AMR severity, including degree of allograft dysfunction and risk of death compared to possible AMR. CONCLUSIONS Clinical AMR showed greater risk of allograft failure than subclinical AMR or no AMR. Subcategorization of clinical AMR based on diagnostic certainty correlated with AMR severity and risk of death, but not with the risk of allograft failure.
Collapse
Affiliation(s)
- Ananth V Charya
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care, University of Maryland Medical Center, Baltimore, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Ileana L Ponor
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland; Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Adam Cochrane
- Advanced Lung Disease and Lung Transplantation Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Deborah Levine
- Lung Transplantation Program, University of Texas, San Antonio, Texas
| | - Mary Philogene
- Histocompatibility and Molecular Genetics Laboratory, Philadelphia, Pennsylvania
| | - Yi-Ping Fu
- Biostatistics, Division of Intramural Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Moon K Jang
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Pali Shah
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ann Mary Bon
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Aravind Krishnan
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - Joby Mathew
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Helen Luikart
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Kiran K Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Gerald Berry
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Charles Marboe
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Department of Pathology, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York
| | - Aldo Iacono
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care, University of Maryland Medical Center, Baltimore, Maryland
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplantation Program, Inova Fairfax Hospital, Fairfax, Virginia.
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics, Division of Intramural Research, National Heart, Lung and Blood Institute, Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
34
|
Kueng N, Arcioni S, Sandberg F, Kuhn C, Banz V, Largiadèr CR, Sidler D, Amstutz U. Comparison of methods for donor-derived cell-free DNA quantification in plasma and urine from solid organ transplant recipients. Front Genet 2023; 14:1089830. [PMID: 36777723 PMCID: PMC9916053 DOI: 10.3389/fgene.2023.1089830] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
In allograft monitoring of solid organ transplant recipients, liquid biopsy has emerged as a novel approach using quantification of donor-derived cell-free DNA (dd-cfDNA) in plasma. Despite early clinical implementation and analytical validation of techniques, direct comparisons of dd-cfDNA quantification methods are lacking. Furthermore, data on dd-cfDNA in urine is scarce and high-throughput sequencing-based methods so far have not leveraged unique molecular identifiers (UMIs) for absolute dd-cfDNA quantification. Different dd-cfDNA quantification approaches were compared in urine and plasma of kidney and liver recipients: A) Droplet digital PCR (ddPCR) using allele-specific detection of seven common HLA-DRB1 alleles and the Y chromosome; B) high-throughput sequencing (HTS) using a custom QIAseq DNA panel targeting 121 common polymorphisms; and C) a commercial dd-cfDNA quantification method (AlloSeq® cfDNA, CareDx). Dd-cfDNA was quantified as %dd-cfDNA, and for ddPCR and HTS using UMIs additionally as donor copies. In addition, relative and absolute dd-cfDNA levels in urine and plasma were compared in clinically stable recipients. The HTS method presented here showed a strong correlation of the %dd-cfDNA with ddPCR (R 2 = 0.98) and AlloSeq® cfDNA (R 2 = 0.99) displaying only minimal to no proportional bias. Absolute dd-cfDNA copies also correlated strongly (τ = 0.78) between HTS with UMI and ddPCR albeit with substantial proportional bias (slope: 0.25; 95%-CI: 0.19-0.26). Among 30 stable kidney transplant recipients, the median %dd-cfDNA in urine was 39.5% (interquartile range, IQR: 21.8-58.5%) with 36.6 copies/μmol urinary creatinine (IQR: 18.4-109) and 0.19% (IQR: 0.01-0.43%) with 5.0 copies/ml (IQR: 1.8-12.9) in plasma without any correlation between body fluids. The median %dd-cfDNA in plasma from eight stable liver recipients was 2.2% (IQR: 0.72-4.1%) with 120 copies/ml (IQR: 85.0-138) while the median dd-cfDNA copies/ml was below 0.1 in urine. This first head-to-head comparison of methods for absolute and relative quantification of dd-cfDNA in urine and plasma supports a method-independent %dd-cfDNA cutoff and indicates the suitability of the presented HTS method for absolute dd-cfDNA quantification using UMIs. To evaluate the utility of dd-cfDNA in urine for allograft surveillance, absolute levels instead of relative amounts will most likely be required given the extensive variability of %dd-cfDNA in stable kidney recipients.
Collapse
Affiliation(s)
- Nicholas Kueng
- Department of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Séverine Arcioni
- Department of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
- Division of Medical Genetics, Central Institute of Hospitals, Valais Hospital, Sion, Switzerland
| | - Fanny Sandberg
- Department of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Christian Kuhn
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Vanessa Banz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Carlo R. Largiadèr
- Department of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Daniel Sidler
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Ursula Amstutz
- Department of Clinical Chemistry, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
Pedini P, Coiffard B, Cherouat N, Casas S, Fina F, Boutonnet A, Baudey JB, Aho P, Basire A, Simon S, Frassati C, Chiaroni J, Reynaud-Gaubert M, Picard C. Clinical relevance of cell-free DNA quantification and qualification during the first month after lung transplantation. Front Immunol 2023; 14:1183949. [PMID: 37180126 PMCID: PMC10174290 DOI: 10.3389/fimmu.2023.1183949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
Background Many studies have reported the relevance of donor-derived cfDNA (dd-cfDNA) after lung transplantation (LTx) to diagnose and monitor acute rejection (AR) or chronic rejection or infection (INF). However, the analysis of cfDNA fragment size has not been studied. The aim of this study was to determine the clinical relevance of dd-cfDNA and cfDNA size profiles in events (AR and INF) during the first month after LTx. Methods This prospective, single-center study includes 62 LTx recipients at the Marseille Nord Hospital, France. Total cfDNA quantification was performed by fluorimetry and digital PCR, dd-cfDNA by NGS (AlloSeq cfDNA-CareDX®), and the size profile by BIABooster (Adelis®). A bronchoalveolar lavage and transbronchial biopsies at D30 established the following groups: not-injured and injured graft (AR, INF, or AR+INF). Results Quantification of total cfDNA was not correlated with the patient's status at D30. The percentage of dd-cfDNA was significantly higher for injured graft patients at D30 (p=0.0004). A threshold of 1.72% of dd-cfDNA correctly classified the not-injured graft patients (negative predictive value of 91.4%). Among recipients with dd-cfDNA >1.72%, the quantification of small sizes (80-120bp) >3.70% identified the INF with high performance (specificity and positive predictive value of 100%). Conclusion With the aim of considering cfDNA as a polyvalent non-invasive biomarker in transplantation, an algorithm combining the quantification of dd-cfDNA and small sizes of DNA may significantly classify the different types of allograft injuries.
Collapse
Affiliation(s)
- Pascal Pedini
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
- ADES UMR, Aix Marseille Univ, Marseille, France
- *Correspondence: Pascal Pedini,
| | - Benjamin Coiffard
- Aix-Marseille University, Lung Transplant Department, APHM, Marseille, France
| | - Nicem Cherouat
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Sylvia Casas
- Medical Direction, CareDx, Brisbane, CA, United States
| | | | | | | | - Printil Aho
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Agnes Basire
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Sophie Simon
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Coralie Frassati
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Jacques Chiaroni
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
- ADES UMR, Aix Marseille Univ, Marseille, France
| | | | - Christophe Picard
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
- ADES UMR, Aix Marseille Univ, Marseille, France
| |
Collapse
|
36
|
Trindade AJ, Chapin KC, Mullican A, Gray JN, Hoy H, Demarest CT, Lambright ES, McPherson KA, Norfolk SG, Robbins IM, Bacchetta M, Erasmus DB, Shaver CM. Assessment of dd-cfDNA Levels in Clinically Stable Lung Allograft Recipients Beyond the Initial 2 y Posttransplant. Transplant Direct 2022; 8:e1411. [PMID: 36406896 PMCID: PMC9671749 DOI: 10.1097/txd.0000000000001411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023] Open
Abstract
Donor-derived cell-free DNA (dd-cfDNA) is a useful biomarker for the diagnosis of acute allograft injury within the first 1 to 2 y after lung transplant, but its utility for diagnosing chronic lung allograft dysfunction (CLAD) has not yet been studied. Understanding baseline dd-cfDNA kinetics beyond the initial 2 y posttransplant is a necessary first step in determining the utility of dd-cfDNA as a CLAD biomarker. We seek to establish baseline dd-cfDNA% levels in clinically stable lung allograft recipients who are >2 y posttransplant. Methods We performed a prospective, single-center, observational study to identify plasma dd-cfDNA levels in clinically stable lung allograft recipients >2 y posttransplant. Results Fifty-one subjects were enrolled and ≥3 baseline dd-cfDNA measurements were acquired during a median of 252 d. The median baseline percent dd-cfDNA level in our cohort was 0.45% (interquartile range [IQR], 0.26-0.69). There were statistically significant differences in dd-cfDNA based on posttransplant duration (≤5 y posttransplant median 0.41% [IQR, 0.21-0.64] versus >5 y posttransplant median 0.50% [IQR, 0.33-0.76]; P < 0.02). However, the clinical significance of this small change in dd-cfDNA is uncertain because this magnitude of change is within the biologic test variation of 73%. Conclusions This study is the first to define levels of dd-cfDNA in clinically stable patients who are >2 y post-lung transplant. These findings lay the groundwork for the study of dd-cfDNA as a possible biomarker for CLAD.
Collapse
Affiliation(s)
- Anil J. Trindade
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Kaitlyn C. Chapin
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Amy Mullican
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Haley Hoy
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Caitlin T. Demarest
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Eric S. Lambright
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Katie A. McPherson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Stephanie G. Norfolk
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ivan M. Robbins
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew Bacchetta
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN
| | - David B. Erasmus
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ciara M. Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
37
|
Bazemore K, Permpalung N, Mathew J, Lemma M, Haile B, Avery R, Kong H, Jang MK, Andargie T, Gopinath S, Nathan SD, Aryal S, Orens J, Valantine H, Agbor-Enoh S, Shah P. Elevated cell-free DNA in respiratory viral infection and associated lung allograft dysfunction. Am J Transplant 2022; 22:2560-2570. [PMID: 35729715 DOI: 10.1111/ajt.17125] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 01/25/2023]
Abstract
Respiratory viral infection (RVI) in lung transplant recipients (LTRs) is a risk for chronic lung allograft dysfunction (CLAD). We hypothesize that donor-derived cell-free DNA (%ddcfDNA), at the time of RVI predicts CLAD progression. We followed 39 LTRs with RVI enrolled in the Genomic Research Alliance for Transplantation for 1 year. Plasma %ddcfDNA was measured by shotgun sequencing, with high %ddcfDNA as ≥1% within 7 days of RVI. We examined %ddcfDNA, spirometry, and a composite (progression/failure) of CLAD stage progression, re-transplant, and death from respiratory failure. Fifty-nine RVI episodes, 38 low and 21 high %ddcfDNA were analyzed. High %ddcfDNA subjects had a greater median %FEV1 decline at RVI (-13.83 vs. -1.83, p = .007), day 90 (-7.97 vs. 0.91, p = .04), and 365 (-20.05 vs. 1.09, p = .047), compared to those with low %ddcfDNA and experienced greater progression/failure within 365 days (52.4% vs. 21.6%, p = .01). Elevated %ddcfDNA at RVI was associated with an increased risk of progression/failure adjusting for symptoms and days post-transplant (HR = 1.11, p = .04). No difference in %FEV1 decline was seen at any time point when RVIs were grouped by histopathology result at RVI. %ddcfDNA delineates LTRs with RVI who will recover lung function and who will experience sustained decline, a utility not seen with histopathology.
Collapse
Affiliation(s)
- Katrina Bazemore
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nitipong Permpalung
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Division of Mycology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Joby Mathew
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Merte Lemma
- Advanced Lung Disease and Transplant Program, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, Virginia
| | | | - Robin Avery
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hyesik Kong
- Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Moon Kyoo Jang
- Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Temesgen Andargie
- Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Shilpa Gopinath
- Division of Transplant Oncology Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, Virginia.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Shambhu Aryal
- Advanced Lung Disease and Transplant Program, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, Virginia
| | - Jonathan Orens
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Hannah Valantine
- Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Sean Agbor-Enoh
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Laboratory of Applied Precision Omics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| | - Pali Shah
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland
| |
Collapse
|
38
|
Oellerich M, Budde K, Osmanodja B, Bornemann-Kolatzki K, Beck J, Schütz E, Walson PD. Donor-derived cell-free DNA as a diagnostic tool in transplantation. Front Genet 2022; 13:1031894. [PMID: 36339004 PMCID: PMC9634115 DOI: 10.3389/fgene.2022.1031894] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/11/2022] [Indexed: 11/22/2022] Open
Abstract
There is a need to improve personalized immunosuppression in organ transplantation to reduce premature graft loss. Biomarkers are needed to better detect rejection, asymptomatic graft injury, and under-immunosuppression. Assessment of minimal necessary exposure to guide tapering and prevent immune activation is also important. There is robust clinical evidence from a large number of published studies supporting the role of dd-cfDNA for monitoring graft integrity and detection or exclusion of rejection. Dd-cfDNA indicates graft cell death without being rejection specific. It can be determined in plasma through droplet digital PCR using preselected SNPs or next generation sequencing. Changes in recipient cfDNA (e.g., by infection) can affect the results of dd-cfDNA fractional determination. This limitation can be overcome using absolute dd-cfDNA quantification. The combination of fractional and absolute determination including total cfDNA is recommended for meaningful interpretation of the results. The value proposition for the patient includes earlier transplant injury detection and intervention, less full blown rejection risk, an alternative to invasive biopsies, and personalized immunosuppression with potential for improved long-term outcome. Transplant physicians benefit from better immunosuppressive guidance and having an alternative when biopsies are refused or contraindicated. Further advantages are improved biopsy interpretation, less trial and error changes in immunosuppression, and less time dealing with complications. The laboratory medicine specialist can provide more effective services. Hospital management and insurance companies could benefit from more cost-effective surveillance of transplant recipients. Potential cost savings would result from fewer biopsies as a result of the tests’ high negative predictive value, fewer re-transplantations, and less organ failure with return to dialysis. A pathway to implementation and metrics is suggested to measure the effectiveness of dd-cfDNA testing.
Collapse
Affiliation(s)
- Michael Oellerich
- Department of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
- *Correspondence: Michael Oellerich,
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Julia Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | | | - Philip D. Walson
- Department of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
39
|
Louis K, Lefaucheur C. DSA in solid organ transplantation: is it a matter of specificity, amount, or functional characteristics? Curr Opin Organ Transplant 2022; 27:392-398. [PMID: 35881421 DOI: 10.1097/mot.0000000000001006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The present review describes the clinical relevance of human leukocyte antigen (HLA) donor-specific antibodies (HLA-DSAs) as biomarkers of alloimmunity and summarizes recent improvements in their characterization that provide insights into immune risk assessment, precision diagnosis, and prognostication in transplantation. RECENT FINDINGS Recent studies have addressed the clinical utility of HLA-DSAs as biomarkers for immune risk assessment in pretransplant and peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and risk stratification. SUMMARY HLA-DSAs have proved to be the most advanced immune biomarkers in solid organ transplantation in terms of analytical validity, clinical validity and clinical utility. Recent studies are integrating multiple HLA-DSA characteristics including antibody specificity, HLA class, quantity, immunoglobulin G subclass, and complement-binding capacity to improve risk assessment peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and transplant prognosis evaluation. In addition, integration of HLA-DSAs to clinical, functional and histological transplant parameters has further consolidated the utility of HLA-DSAs as robust biomarkers and allows to build new tools for monitoring, precision diagnosis, and risk stratification for individual patients. However, prospective and randomized-controlled studies addressing the clinical benefit and cost-effectiveness of HLA-DSA-based monitoring and patient management strategies are required to demonstrate that the use of HLA-DSAs as biomarkers can improve current clinical practice and transplant outcomes.
Collapse
Affiliation(s)
- Kevin Louis
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Human Immunology and Immunopathology, Université de Paris
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| |
Collapse
|
40
|
Keller MB, Meda R, Fu S, Yu K, Jang MK, Charya A, Berry GJ, Marboe CC, Kong H, Luikart H, Ponor IL, Shah PD, Khush KK, Nathan SD, Agbor‐Enoh S. Comparison of donor-derived cell-free DNA between single versus double lung transplant recipients. Am J Transplant 2022; 22:2451-2457. [PMID: 35322546 PMCID: PMC9508279 DOI: 10.1111/ajt.17039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 01/25/2023]
Abstract
Plasma donor-derived cell-free DNA (dd-cfDNA) is a sensitive biomarker for the diagnosis of acute rejection in lung transplant recipients; however, differences in dd-cfDNA levels between single and double lung transplant remains unknown. We performed an observational analysis that included 221 patients from two prospective cohort studies who had serial measurements of plasma dd-cfDNA at the time of bronchoscopy and pulmonary function testing, and compared dd-cfDNA between single and double lung transplant recipients across a range of disease states. Levels of dd-cfDNA were lower for single vs. double lung transplant in stable controls (median [IQR]: 0.15% [0.07, 0.44] vs. 0.46% [0.23, 0.74], p < .01) and acute rejection (1.06% [0.75, 2.32] vs. 1.78% [1.18, 5.73], p = .05). Doubling dd-cfDNA for single lung transplant to account for differences in lung mass eliminated this difference. The area under the receiver operating curve (AUC) for the detection of acute rejection was 0.89 and 0.86 for single and double lung transplant, respectively. The optimal dd-cfDNA threshold for the detection of acute rejection was 0.54% in single lung and 1.1% in double lung transplant. In conclusion, accounting for differences in dd-cfDNA in single versus double lung transplant is key for the interpretation of dd-cfDNA testing in research and clinical settings.
Collapse
Affiliation(s)
- Michael B. Keller
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Laboratory of Applied Precision Omics (APO)National Heart, Lung and Blood InstituteBethesdaMarylandUSA,Division of Pulmonary and Critical Care MedicineThe Johns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Rohan Meda
- Laboratory of Applied Precision Omics (APO)National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Sheng Fu
- National Cancer InstituteRockvilleMarylandUSA
| | - Kai Yu
- National Cancer InstituteRockvilleMarylandUSA
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Laboratory of Applied Precision Omics (APO)National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Ananth Charya
- University of Maryland Medical CenterBaltimoreMarylandUSA
| | - Gerald J. Berry
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Stanford University School of MedicineStanfordCaliforniaUSA
| | - Charles C. Marboe
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Department of Pathology and Cell BiologyVagelos College of Physicians and Surgeons of Columbia UniversityNew YorkNew YorkUSA
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Laboratory of Applied Precision Omics (APO)National Heart, Lung and Blood InstituteBethesdaMarylandUSA
| | - Helen Luikart
- Stanford University School of MedicineStanfordCaliforniaUSA
| | - Ileana L. Ponor
- Department of MedicineJohns Hopkins Bayview Medical CenterBaltimoreMarylandUSA
| | - Pali D. Shah
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Division of Pulmonary and Critical Care MedicineThe Johns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Kiran K. Khush
- Stanford University School of MedicineStanfordCaliforniaUSA
| | - Steven D. Nathan
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Inova Fairfax HospitalFairfaxVAUSA
| | - Sean Agbor‐Enoh
- Genomic Research Alliance for Transplantation (GRAfT)BethesdaMarylandUSA,Laboratory of Applied Precision Omics (APO)National Heart, Lung and Blood InstituteBethesdaMarylandUSA,Division of Pulmonary and Critical Care MedicineThe Johns Hopkins School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
41
|
DeMaio A, Thiboutot J, Yarmus L. Applications of cryobiopsy in airway, pleural, and parenchymal disease. Expert Rev Respir Med 2022; 16:875-886. [PMID: 36069255 DOI: 10.1080/17476348.2022.2122444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION :Cryobiopsy is a novel diagnostic technique for thoracic diseases which has been extensively investigated over the past 20 years. It was originally proposed for the diagnosis of endobronchial lesions and diffuse parenchymal lung disease due to limitations of conventional sampling techniques including small size and presence of artifacts. AREAS COVERED :We will review recent evidence related to the expanding use of cryobiopsy in thoracic diseases. To identify references, the MEDLINE database was searched from database inception until May 2022 for case series, cohort studies, randomized controlled trials, systematic reviews and meta-analyses related to cryobiopsy. EXPERT OPINION Cryobiopsy has expanding applications in the field of thoracic diseases. Evidence to support transbronchial cryobiopsy as an alternative to surgical lung biopsy is increasing and was recently endorsed as a conditional recommendation by the latest American Thoracic Society guideline update for Idiopathic Pulmonary Fibrosis. Developments in technology and technique, in particular the availability of a 1.1 mm flexible cryoprobe, have extended applications to pulmonary diseases, including diagnosis of interstitial lung diseases, peripheral pulmonary lesions, and lung transplant rejection.
Collapse
Affiliation(s)
- Andrew DeMaio
- Section of Interventional Pulmonology, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine
| | - Jeffrey Thiboutot
- Section of Interventional Pulmonology, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine
| | - Lonny Yarmus
- Section of Interventional Pulmonology, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine.,Division of Pulmonary and Critical Care Medicine
| |
Collapse
|
42
|
Levine DJ, Demko ZP, Ross DJ. Variability in plasma donor-derived cell-free DNA levels with CLAD more than 5-years after Lung Transplantation: Pilot data. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
43
|
Rosenheck J, Keller B, Fehringer G, Demko Z, Bohrade S, Ross D. Why Cell-Free DNA Can Be a “Game Changer” for Lung Allograft Monitoring for Rejection and Infection. CURRENT PULMONOLOGY REPORTS 2022; 11:75-85. [PMID: 35910533 PMCID: PMC9315332 DOI: 10.1007/s13665-022-00292-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 02/06/2023]
Abstract
Purpose of Review Although there has been improvement in short-term clinical outcomes for patients following lung transplant (LT), advances have not translated into longer-term allograft survival. Furthermore, invasive biopsies are still standard of practice for monitoring LT recipients for allograft injury. We review the relevant literature supporting the role of using plasma donor-derived cell-free DNA (dd-cfDNA) as a non-invasive biomarker for LT allograft injury surveillance and discuss future research directions. Recent Findings Accumulating data has demonstrated that dd-cfDNA is associated with molecular and cellular injury due to acute (cellular and antibody-mediated) rejection, chronic lung allograft dysfunction, and relevant infectious pathogens. Strong performance in distinguishing rejection and allograft injury from stable patients has set the stage for clinical trials to assess dd-cfDNA utility for surveillance of LT patients. Research investigating the potential role of dd-cfDNA methylation signatures to map injured tissue and cell-free DNA in detecting allograft injury-related pathogens is ongoing. Summary There is an amassed breadth of clinical data to support a role for dd-cfDNA in monitoring rejection and other forms of allograft injury. Rigorously designed, robust clinical trials that encompass the diversity in patient demographics are paramount to furthering our understanding and adoption of plasma dd-cfDNA for surveillance of lung allograft health.
Collapse
Affiliation(s)
- J.P. Rosenheck
- Division of Pulmonary, Critical Care & Sleep Medicine, The Ohio State University, Columbus, OH USA
| | - B.C. Keller
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA USA
| | - G. Fehringer
- Medical Affairs in Organ Health, Natera, Inc., San Carlos, USA
| | - Z.P. Demko
- Medical Affairs in Organ Health, Natera, Inc., San Carlos, USA
| | - S.M. Bohrade
- Medical Affairs in Organ Health, Natera, Inc., San Carlos, USA
| | - D.J. Ross
- Medical Affairs in Organ Health, Natera, Inc., San Carlos, USA
- Lung Transplant & Molecular Diagnostics, Natera, Inc, San Carlos, CA USA
| |
Collapse
|
44
|
Sorbini M, Togliatto G, Mioli F, Simonato E, Marro M, Cappuccio M, Arruga F, Caorsi C, Mansouri M, Magistroni P, Gambella A, Delsedime L, Papotti MG, Solidoro P, Albera C, Boffini M, Rinaldi M, Amoroso A, Vaisitti T, Deaglio S. Validation of a Simple, Rapid, and Cost-Effective Method for Acute Rejection Monitoring in Lung Transplant Recipients. Transpl Int 2022; 35:10546. [PMID: 35755857 PMCID: PMC9221674 DOI: 10.3389/ti.2022.10546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
Abstract
Despite advances in immunosuppression therapy, acute rejection remains the leading cause of graft dysfunction in lung transplant recipients. Donor-derived cell-free DNA is increasingly being considered as a valuable biomarker of acute rejection in several solid organ transplants. We present a technically improved molecular method based on digital PCR that targets the mismatch between the recipient and donor at the HLA-DRB1 locus. Blood samples collected sequentially post-transplantation from a cohort of lung recipients were used to obtain proof-of-principle for the validity of the assay, correlating results with transbronchial biopsies and lung capacity tests. The results revealed an increase in dd-cfDNA during the first 2 weeks after transplantation related to ischemia-reperfusion injury (6.36 ± 5.36%, p < 0.0001). In the absence of complications, donor DNA levels stabilized, while increasing again during acute rejection episodes (7.81 ± 12.7%, p < 0.0001). Respiratory tract infections were also involved in the release of dd-cfDNA (9.14 ± 15.59%, p = 0.0004), with a positive correlation with C-reactive protein levels. Overall, the dd-cfDNA percentages were inversely correlated with the lung function values measured by spirometry. These results confirm the value of dd-cfDNA determination during post-transplant follow-up to monitor acute rejection in lung recipients, achieved using a rapid and inexpensive approach based on the HLA mismatch between donor and recipient.
Collapse
Affiliation(s)
- Monica Sorbini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Fiorenza Mioli
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Erika Simonato
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Matteo Marro
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | | | - Francesca Arruga
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristiana Caorsi
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Morteza Mansouri
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Paola Magistroni
- Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | | | - Luisa Delsedime
- Pathology Unit, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Mauro Giulio Papotti
- Pathology Unit, Città Della Salute e Della Scienza University Hospital, Turin, Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Paolo Solidoro
- Lung Transplantation and Advanced Airways Management, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Carlo Albera
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Boffini
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Mauro Rinaldi
- Cardiac Surgery Division, Surgical Sciences Department, Heart and Lung Transplant Center, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, University of Turin, Turin, Italy.,Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy.,Immunogenetics and Transplant Biology Service, Città Della Salute e Della Scienza University Hospital, Turin, Italy
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Recent evidence indicates that plasma donor-derived cell-free DNA (dd-cfDNA) is a sensitive biomarker for the detection of underlying allograft injury, including rejection and infection. In this review, we will cover the latest evidence revolving around dd-cfDNA in lung transplantation and its role in both advancing mechanistic insight into disease states in lung transplant recipients as well as its potential clinical utility. RECENT FINDINGS Plasma dd-cfDNA increases in the setting of allograft injury, including in primary graft dysfunction, acute cellular rejection, antibody-mediated rejection and infection. Dd-cfDNA has demonstrated good performance characteristics for the detection of various allograft injury states, most notably with a high negative-predictive value for detection of acute rejection. Elevated levels of dd-cfDNA in the early posttransplant period, reflecting molecular evidence of lung allograft injury, are associated with increased risk of chronic lung allograft dysfunction and death. SUMMARY As a quantitative, molecular biomarker of lung allograft injury, dd-cfDNA holds great promise in clinical and research settings for advancing methods of posttransplant surveillance monitoring, diagnosis of allograft injury states, monitoring adequacy of immunosuppression, risk stratification and unlocking pathophysiological mechanisms of various disease.
Collapse
|
46
|
Cell Death and Ischemia-Reperfusion Injury in Lung Transplantation. J Heart Lung Transplant 2022; 41:1003-1013. [DOI: 10.1016/j.healun.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
|
47
|
Clinical Validation of a Plasma Donor-derived Cell-free DNA Assay to Detect Allograft Rejection and Injury in Lung Transplant. Transplant Direct 2022; 8:e1317. [PMID: 35372675 PMCID: PMC8963832 DOI: 10.1097/txd.0000000000001317] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/04/2022] [Accepted: 02/26/2022] [Indexed: 01/29/2023] Open
|
48
|
Keller M, Mutebi C, Shah P, Levine D, Aryal S, Iacono A, Timofte I, Mathew J, Varghese A, Giner C, Agbor-Enoh S. Biological Variation of Donor-Derived Cell-Free DNA in Stable Lung Transplant Recipients. J Appl Lab Med 2022; 7:901-909. [PMID: 35024828 DOI: 10.1093/jalm/jfab171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Prior studies demonstrate that donor-derived cell-free DNA (dd-cfDNA) in lung transplant recipients may serve as a marker of allograft injury for detecting allograft rejection and infection. Clinical interpretation of dd-cfDNA requires understanding its biological variation in stable lung transplant patients in order to identify abnormal results suggesting underlying allograft dysfunction. This study establishes the biological variation and reference change values (RCV) of dd-cfDNA in stable lung transplant recipients using an analytically validated assay with an established analytic coefficient of variation (CVA). METHODS The AlloSure® assay, a targeted, sequencing-based approach, was used to measure plasma dd-cfDNA in a cohort of lung transplant patients at 4 centers that used dd-cfDNA to monitor for allograft dysfunction in preference to surveillance transbronchial biopsy. Patients with stable allograft function and ≥3 dd-cfDNA samples were included. Intraindividual coefficient of variation (CVI), interindividual CV (CVG), index of individuality (II) and the RCV were calculated. RESULTS Thirty-five patients with a combined 124 dd-cfDNA samples were included in the final analysis. The median dd-cfDNA was 0.31% (interquartile range 0.18%-0.68%), the 97.5th percentile and 95th percentile were 1.3% and 1.0%, respectively. In 30 stable patients with an average of 3.7 tests, the CVI was 25%, CVG 19%, II 1.33, and RCV 70%. CONCLUSION In stable lung transplant patients, fluctuations in dd-cfDNA levels of up to 70% or levels less than 1% are within normal biological variation. With further validation, these thresholds may be incorporated into surveillance monitoring algorithms to identify potentially abnormal results indicating allograft dysfunction.
Collapse
Affiliation(s)
- Michael Keller
- Laboratory of Applied Precision Omics (APO) and Genomic Research Alliance for Transplantation (GRAfT), National Institutes of Health, Bethesda, MD, USA.,Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Cedric Mutebi
- Laboratory of Applied Precision Omics (APO) and Genomic Research Alliance for Transplantation (GRAfT), National Institutes of Health, Bethesda, MD, USA
| | - Pali Shah
- Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Deborah Levine
- Pulmonary and Critical Care Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shambhu Aryal
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Aldo Iacono
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Irina Timofte
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Joby Mathew
- Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Anu Varghese
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Cassandra Giner
- Pulmonary and Critical Care Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Sean Agbor-Enoh
- Laboratory of Applied Precision Omics (APO) and Genomic Research Alliance for Transplantation (GRAfT), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Mackintosh JA, Chambers DC. Genomic Lung Allograft Surveillance – Is It Primer Time? J Heart Lung Transplant 2022; 41:467-469. [DOI: 10.1016/j.healun.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 10/19/2022] Open
|
50
|
Keller M, Sun J, Mutebi C, Shah P, Levine D, Aryal S, Iacono A, Timofte I, Mathew J, Varghese A, Giner C, Agbor-Enoh S. Donor-derived cell-free DNA as a composite marker of acute lung allograft dysfunction in clinical care. J Heart Lung Transplant 2021; 41:458-466. [DOI: 10.1016/j.healun.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/07/2021] [Accepted: 12/21/2021] [Indexed: 11/28/2022] Open
|