1
|
Geraci TC, Chan JC, Niroomand A, Chang SH. Post Lung Transplant Primary Graft Dysfunction. Semin Thorac Cardiovasc Surg 2025:S1043-0679(25)00050-4. [PMID: 40268260 DOI: 10.1053/j.semtcvs.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/18/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
Primary graft dysfunction (PGD) is a major source of morbidity and mortality following lung transplantation, presenting as acute lung injury within 72 hours post-transplantation. Despite advances in surgical techniques and perioperative care, the complex interplay of donor, recipient, and perioperative factors contributes to its development, underscoring the multifactorial nature of PGD. Clinical management of recipients with PGD relies on supportive care strategies, including lung-protective ventilation, inhaled nitric oxide, and extracorporeal membrane oxygenation (ECMO). Severe cases of PGD may result in significant short- and long-term adverse outcomes, including early mortality. Even for patients who recover from PGD, there is also an associated increased risk of chronic lung allograft dysfunction, further compounding its clinical impact. This review provides a brief review of current knowledge regarding PGD, detailing risk factors, diagnostic criteria, and management approaches while identifying critical gaps in understanding its pathophysiology. Ongoing research is essential to develop innovative therapeutic strategies and improve outcomes for lung transplant recipients.
Collapse
Affiliation(s)
- Travis C Geraci
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY USA
| | - Justin Cy Chan
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY USA
| | - Anna Niroomand
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY USA
| | - Stephanie H Chang
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY USA.
| |
Collapse
|
2
|
Miceli V, Ferrigno P, Centi C, Carcione C, Iannolo G, Agnese V, Lo Iacono G, Liotta R, Conaldi PG, Pinzani M, De Monte L, Bertani A. Differentially expressed microRNAs in pre-transplant lung biopsies target immune checkpoint proteins and can predict primary graft dysfunction in lung transplantation. Heliyon 2025; 11:e42515. [PMID: 40028527 PMCID: PMC11869042 DOI: 10.1016/j.heliyon.2025.e42515] [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: 05/10/2024] [Revised: 01/18/2025] [Accepted: 02/06/2025] [Indexed: 03/02/2025] Open
Abstract
Lung transplantation (LTx) significantly improves outcomes for patients with end-stage respiratory failure. However, primary graft dysfunction (PGD) remains one of the most relevant hurdles. Although PGD is attributed to ischemia-reperfusion injury (IRI), immune responses, primarily T cell-mediated, may play a pivotal role in its pathogenesis. Additionally, innate immune activation following IRI links PGD to adaptive alloimmunity, highlighting the impact of early events on LTx outcomes. Immune checkpoints (ICPs) such as PD-1/PD-L1, CD40/CD40LG, and OX40/OX40L, regulate post-LTx T cell responses, and dysregulation of microRNAs (miRNAs) has been implicated in altering ICP expression, influencing the amplification of immune responses. In this preliminary study, we used the taqMan low-density array (TLDA) cards to investigate miRNA dysregulation's prognostic potential as a PGD marker in pre-transplant back-table lung biopsies. Our analysis revealed differential miRNA expression in donor lung tissues, potentially associated with PGD onset, targeting immune regulatory pathways. Specifically, deregulated miRNAs targeted key ICP proteins, including PD-L1, CD40LG, and OX40L. Moreover, the differential expression of these miRNAs was observed in grafts with future PGD compared to grafts without PGD, suggesting a potential prognostic benefit and a possible role for lung tissue miRNAs in the onset of early graft dysfunction. These findings provide a basis for future investigations into their mechanistic roles and therapeutic potential for PGD. Although based on a limited number of cases, our results imply that miRNAs might be involved in early graft dysfunction. While requiring validation in larger cohorts, our data raise the possibility that the evaluation of the aforementioned markers during the pre-transplant phase, might offer a prognostic benefit in monitoring the onset of PGD. Additionally, the use of compounds that can modulate the function of these molecules could be evaluated for the management of LTx patients.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Pia Ferrigno
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| | - Claudio Centi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | | | - Gioacchin Iannolo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Valentina Agnese
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Giovanna Lo Iacono
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Rosa Liotta
- Pathology Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Massimo Pinzani
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Lavinia De Monte
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
| | - Alessandro Bertani
- Division of Thoracic Surgery and Lung Transplantation, Chest Center, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo, Italy
- UPMCI (University of Pittsburgh Medical Center Italy), Palermo, Italy
| |
Collapse
|
3
|
Ma W, Oh I, Luo Y, Kumar S, Gupta A, Lai AM, Puri V, Kreisel D, Gelman AE, Nava R, Witt CA, Byers DE, Halverson L, Vazquez-Guillamet R, Payne PRO, Sotiras A, Lu H, Niazi K, Gurcan MN, Hachem RR, Michelson AP. Developing approaches to incorporate donor-lung computed tomography images into machine learning models to predict severe primary graft dysfunction after lung transplantation. Am J Transplant 2025:S1600-6135(25)00045-0. [PMID: 39924113 DOI: 10.1016/j.ajt.2025.01.039] [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: 06/11/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
Primary graft dysfunction (PGD) is a common complication after lung transplantation associated with poor outcomes. Although risk factors have been identified, the complex interactions between clinical variables affecting PGD risk are not well understood, which can complicate decisions about donor-lung acceptance. Previously, we developed a machine learning model to predict grade 3 PGD using donor and recipient electronic health record data, but it lacked granular information from donor-lung computed tomography (CT) scans, which are routinely assessed during offer review. In this study, we used a gated approach to determine optimal methods for analyzing donor-lung CT scans among patients receiving first-time, bilateral lung transplants at a single center over 10 years. We assessed 4 computer vision approaches and fused the best with electronic health record data at 3 points in the machine learning process. A total of 160 patients had donor-lung CT scans for analysis. The best imaging-only approach employed a 3D ResNet model, yielding median (interquartile range) areas under the receiver operating characteristic and precision-recall curves of 0.63 (0.49-0.72) and 0.48 (0.35-0.6), respectively. Combining imaging with clinical data using late fusion provided the highest performance, with median areas under the receiver operating characteristic and precision-recall curves of 0.74 (0.59-0.85) and 0.61 (0.47-0.72), respectively.
Collapse
Affiliation(s)
- Weiwei Ma
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Inez Oh
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Yixuan Luo
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Sayantan Kumar
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Aditi Gupta
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA; Division of Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Albert M Lai
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Varun Puri
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ruben Nava
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Chad A Witt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Laura Halverson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Rodrigo Vazquez-Guillamet
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Philip R O Payne
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Aristeidis Sotiras
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Hao Lu
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Khalid Niazi
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Metin N Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ramsey R Hachem
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Andrew P Michelson
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
4
|
Sella N, Pettenuzzo T, Congedi S, Bisi M, Gianino G, De Carolis A, Bertoncello CA, Roccaforte M, Zarantonello F, Persona P, Petranzan E, Roca G, Biamonte E, Carron M, Dell'Amore A, Rea F, Boscolo A, Navalesi P. Early Prone Positioning As a Rescue Therapy for Moderate-to-severe Primary Graft Dysfunction After Bilateral Lung Transplant. J Cardiothorac Vasc Anesth 2025; 39:479-488. [PMID: 39675928 DOI: 10.1053/j.jvca.2024.11.018] [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: 06/27/2024] [Revised: 10/24/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVES Primary graft dysfunction (PGD) affects survival after lung transplant (LT). The current hypothesis was that prone positioning (PP), proposed as a rescue maneuver to treat refractory hypoxemia due to PGD, may improve LT outcomes, especially when applied early. DESIGN Bilateral LT recipients developing moderate-to-severe PGD within 24 hours from intensive care unit admission were enrolled. From January 2020 to November 2021, patients developing PGD after LT were turned prone between 24 and 48 hours after diagnosis, only in case of radiological or oxygenation worsening ("late PP" group). After November 2021, patients were routinely turned prone within 24 hours from PGD diagnosis ("early PP"). A propensity score-weighted analysis, adjusted for clinically relevant covariates, was applied. SETTING Intensive care unit. PARTICIPANTS Bilateral LT recipients. INTERVENTIONS Early PP, late PP, or supine position. MEASUREMENTS AND MAIN RESULTS 130 LT patients were screened and 67 were enrolled. A total of 25 (37%) recipients were treated in the supine position, 24 (36%) in early PP, and 18 (27%) in late PP. After propensity score weighting, both supine treatment (estimated effect for 1 ventilator-free day = 8.23, standard error: 2.97, p = 0.007) and early PP treatment (estimated effect = 9.42, standard error: 2.59, p < 0.001) were associated with greater 28-day ventilator-free days than late PP treatment (reference). Compared with late PP, early PP was also associated with better oxygenation, driving pressure, and static respiratory system compliance. Compared with supine recipients, the early PP group showed better oxygenation at 72 hours after PGD diagnosis. CONCLUSIONS Early PP in LT recipients with moderate-to-severe PGD seems to be associated with better 28-day ventilator-free days, oxygenation, and driving pressure than late PP.
Collapse
Affiliation(s)
- Nicolò Sella
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy
| | - Tommaso Pettenuzzo
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy
| | | | - Maria Bisi
- Department of Medicine, University of Padua, Padua, Italy
| | - Giulio Gianino
- Department of Medicine, University of Padua, Padua, Italy
| | - Agnese De Carolis
- Department of Anesthesia, Critical Care and Emergency, University of Milan, Milan, Italy
| | | | | | | | - Paolo Persona
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy
| | - Enrico Petranzan
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy
| | - Gabriella Roca
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padua, Italy
| | - Eugenio Biamonte
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| | - Michele Carron
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy; Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Dell'Amore
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padua, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padua, Italy
| | - Annalisa Boscolo
- Anesthesia and Intensive Care, University Hospital of Padua, Padua, Italy; Department of Medicine, University of Padua, Padua, Italy; Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padua, Italy.
| | - Paolo Navalesi
- Department of Medicine, University of Padua, Padua, Italy
| |
Collapse
|
5
|
Beeckmans H, Kerckhof P, Acet Öztürk N, Zajacova A, Van Slambrouck J, Bos S, Vermant M, Van Dieren LO, Goeminne T, Vandervelde C, Bardyn J, Willems E, Lauriers S, Brusselmans M, Langenhoven LV, Emonds MP, De Pelsmaeker S, Kerkhofs J, Sadeleer LD, Godinas L, Dupont LJ, Raemdonck DEV, Ceulemans LJ, Vanaudenaerde BM, Vos R. Clinical predictors for restrictive allograft syndrome: a nested case-control study. Am J Transplant 2025:S1600-6135(25)00042-5. [PMID: 39892791 DOI: 10.1016/j.ajt.2025.01.036] [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: 07/24/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Risk factors for restrictive allograft syndrome (RAS), a severe phenotype of chronic lung allograft dysfunction (CLAD) after lung transplantation, are currently not well known. In this retrospective nested case-control-study, we analyzed 69 patients with RAS and 69 matched non-CLAD controls to identify clinical risk factors for RAS. Patients with RAS demonstrated overall higher blood eosinophils (P = .02), increased bronchoalveolar eosinophils (P < .001) and lymphocytes (P = .03), and higher incidence of infections, particularly Pseudomonas species infection (P = .003), invasive fungal disease (P < .001, mainly due to Aspergillus species), SARS-CoV-2 (P < .001), and cytomegalovirus infection (P = .04), compared with non-CLAD controls. Antihuman leukocyte antigen (anti-HLA) antibodies, especially persistent donor-specific antibodies (P < 0.001), specifically targeting HLA-DQ and HLA-DR loci, and antibody-mediated rejection (P < .001), were strongly associated with later RAS. Histopathologic lung injury patterns on transbronchial biopsy (P < .001), and persistent chest computed tomography opacities in absence of pulmonary dysfunction (P < .001) were identified as early indicators of later RAS. Proactive detection and management of these risk factors could help mitigate future decline in allograft function and reduce progression to clinical RAS. Future studies should explore early treatment strategies targeting these modifiable factors to preserve allograft function and improve long-term outcomes for lung transplant recipients.
Collapse
Affiliation(s)
- Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| | - Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Nilufer Acet Öztürk
- Department of Pulmonology, Uludağ University Faculty of Medicine, Bursa, Turkey
| | - Andrea Zajacova
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Saskia Bos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Lyne O Van Dieren
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Tessa Goeminne
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Christelle Vandervelde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Josephine Bardyn
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Elisabeth Willems
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Sam Lauriers
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Marie Paule Emonds
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium; Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Steffi De Pelsmaeker
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium
| | - Johan Kerkhofs
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross Flanders, Mechelen, Belgium
| | - Laurens De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Laurent Godinas
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Lieven J Dupont
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Miyashita Y, Kaiho T, Kurihara C. Unveiling the potential of lung transplantation for situs inversus. Expert Rev Respir Med 2025; 19:43-54. [PMID: 39718540 DOI: 10.1080/17476348.2024.2447513] [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: 09/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Situs inversus is a rare congenital condition where the organs in the chest and abdomen are reversed, thus complicating surgeries such as lung transplantation. Kartagener syndrome (KS), associated with situs inversus, includes chronic sinusitis and bronchiectasis, which can progress to end-stage lung disease requiring transplantation. This review discusses the unique surgical considerations, technical challenges, and outcomes of lung transplantation in patients with situs inversus, particularly KS. AREAS COVERED The review highlights anatomical and physiological challenges in lung transplantation due to reversed organ positioning, requiring customized surgical approaches and intraoperative modifications. Preoperative imaging, anesthesia adjustments, and tailored surgical techniques are crucial for successful transplantation. Postoperative care focuses on managing complications such as primary graft dysfunction, infections, and anastomotic issues. Literature on survival rates, chronic lung allograft dysfunction, and quality of life is analyzed, indicating outcomes comparable to other lung transplant recipients. EXPERT OPINION Despite significant challenges, lung transplantation in patients with situs inversus and KS is feasible with outcomes similar to traditional cases. Advances in imaging, surgical planning, and minimally invasive techniques offer promise for improved outcomes. Ongoing research, collaboration, and ethical considerations are essential to optimizing care and expand treatment possibilities for this high-risk patient population.
Collapse
Affiliation(s)
- Yudai Miyashita
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Taisuke Kaiho
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chitaru Kurihara
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
7
|
Yamanashi K, Wang A, Bellissimo CA, Siebiger G, Oliveira P, Zhang Y, Montagne J, Garza G, Furie N, Pal P, Liu M, Goligher EC, Keshavjee S, Cypel M. Protective effects of 10 °C preservation on donor lungs with lipopolysaccharide-induced acute lung injury. J Thorac Cardiovasc Surg 2024:S0022-5223(24)00829-8. [PMID: 39321867 DOI: 10.1016/j.jtcvs.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/27/2024] [Accepted: 09/14/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVES Hypothermic lung preservation at 10 °C has been recently shown to enhance quality of healthy donor lungs during ischemia. This study aims to show generalizability of the 10 °C lung preservation using an endotoxin-induced lung injury with specific focus on the benefits of post-transplant lung function and mitochondrial preservation. METHODS Lipopolysaccharide (3 mg/kg) was injected intratracheally in rats to induce lung injury. Injured lungs were flushed with preservation solution and allocated to 3 groups (n = 6 each): minimum cold storage, 6-hour storage on ice (ice), and 6-hour storage at 10 °C (10 °C). Left lungs were transplanted and reperfused for 2 hours. After storage, lung tissue was used to evaluate the effects of hypothermic storage on the mitochondrial function: mitochondrial membrane potential was assessed by JC-1 staining; mitochondrial oxygen consumption was assessed using high-resolution respirometry. RESULTS Two hours after reperfusion, the oxygen tension/inspired oxygen fraction ratio from the graft was significantly greater in the 10 °C group than in the Ice group (P = .015), whereas the wet-to-dry weight ratio was significantly lower (P = .041). Levels of interleukin-8 in lung tissues were significantly lower in the 10 °C group than in the Ice group (P = .004). Mechanistically, we noted greater mitochondrial membrane potential and elevated state III respiration in the 10 °C group than in the Ice group (P = .015 and P = .002, respectively), implying higher metabolic activities may be maintained during 10 °C preservation. CONCLUSIONS Favorable metabolism during 10 °C preservation prevented ischemia-induced mitochondrial damages in injured lungs, leading to better post-transplant outcomes.
Collapse
Affiliation(s)
- Keiji Yamanashi
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Thoracic and Cardio-Vascular Surgery, Nara Medical University School of Medicine, Kashihara, Japan
| | - Aizhou Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Catherine A Bellissimo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Gabriel Siebiger
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Paolo Oliveira
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Instituto do Coração, Faculdade de Medicina HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Yu Zhang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Juan Montagne
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Guillermo Garza
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nadav Furie
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Prodipto Pal
- Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ewan C Goligher
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Nord D, Brunson JC, Langerude L, Moussa H, Gill B, Machuca T, Rackauskas M, Sharma A, Lin C, Emtiazjoo A, Atkinson C. Predicting Primary Graft Dysfunction in Lung Transplantation: Machine Learning-Guided Biomarker Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595368. [PMID: 39386627 PMCID: PMC11463600 DOI: 10.1101/2024.05.24.595368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
BACKGROUND – There is an urgent need to better understand the pathophysiology of primary graft dysfunction (PGD) so that point-of-care methods can be developed to predict those at risk. Here we utilize a multiplex multivariable approach to define cytokine, chemokines, and growth factors in patient-matched biospecimens from multiple biological sites to identify factors predictive of PGD. METHODS – Biospecimens were collected from patients undergoing bilateral LTx from three distinct sites: donor lung perfusate, post-transplant bronchoalveolar lavage (BAL) fluid (2h), and plasma (2h and 24h). A 71-multiplex panel was performed on each biospecimen. Cross-validated logistic regression (LR) and random forest (RF) machine learning models were used to determine whether analytes in each site or from combination of sites, with or without clinical data, could discriminate between PGD grade 0 (n = 9) and 3 (n = 8). RESULTS – Using optimal AUROC, BAL fluid at 2h was the most predictive of PGD (LR, 0.825; RF, 0.919), followed by multi-timepoint plasma (LR, 0.841; RF, 0.653), then perfusate (LR, 0.565; RF, 0.448). Combined clinical, BAL, and plasma data yielded strongest performance (LR, 1.000; RF, 1.000). Using a LASSO of the predictors obtained using LR, we selected IL-1RA, BCA-1, and Fractalkine, as most predictive of severe PGD. CONCLUSIONS – BAL samples collected 2h post-transplant were the strongest predictors of severe PGD. Our machine learning approach not only identified novel cytokines not previously associated with PGD, but identified analytes that could be used as a point-of-care cytokine panel aimed at identifying those at risk for developing severe PGD.
Collapse
Affiliation(s)
- Dianna Nord
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL
| | | | - Logan Langerude
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL
| | - Hassan Moussa
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL
| | - Blake Gill
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL
| | - Tiago Machuca
- Department of Surgery, University of Miami, Miami, FL
| | | | - Ashish Sharma
- Department of Surgery, University of Florida, Gainesville, FL
| | - Christine Lin
- Department of Medicine, University of California San Diego, San Diego, CA
| | - Amir Emtiazjoo
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL
| | - Carl Atkinson
- Department of Surgery, Northwestern University, Chicago, IL
| |
Collapse
|
9
|
Kurihara C, Kaiho T, Thomae B, Cerier E, Lung K, Avella Patino D, Toyoda T, Yan Y, Budinger GRS, Bharat A. Contrasting predictors of severe primary graft dysfunction following transplant for chronic and acute respiratory failure. J Thorac Dis 2024; 16:5050-5062. [PMID: 39268121 PMCID: PMC11388248 DOI: 10.21037/jtd-24-100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/19/2024] [Indexed: 09/15/2024]
Abstract
Background Lung transplantation represents a pivotal intervention for individuals grappling with end-stage lung diseases, and the role of lung transplantation in acute respiratory distress syndrome (ARDS) patients has garnered increased attention especially after the coronavirus disease 2019 (COVID-19) pandemic. Multiple studies have demonstrated a high incidence of primary graft dysfunction (PGD) in patients with ARDS compared to contemporaneous controls undergoing transplantation for chronic end-stage lung diseases although underlying mechanisms or risk factors remain unknown. This retrospective study investigates the contrasting risk factors for PGD grade 3 in patients with ARDS and chronic respiratory failure undergoing lung transplantation. Methods The study included 293 patients who underwent lung transplantation from January 2018 through June 2023. We performed a multivariate logistic regression analysis using variables from the univariate logistic regression analyses to predict PGD grade 3. Results Our findings reveal distinct predictors for PGD grade 3 in the two cohorts. ARDS patients had higher incidence of PGD grade 3 than non-ARDS patients (30.2% vs. 9.6%, P<0.001). Multivariate logistic regression analysis showed ischemic time [odds ratio (OR) =0.60; 95% confidence interval (CI): 0.40-0.90; P=0.01] as predictor of PGD grade 3 for non-ARDS patients, and age (OR =0.72; 95% CI: 0.52-0.99; P=0.048), pre-operative albumin (OR <0.01; 95% CI: <0.01-0.74; P=0.042) for ARDS patients. Interestingly, there was no notable difference in post-transplant survival between the two groups. Conclusions This study highlights differing risk profiles for severe PGD in ARDS and non-ARDS lung transplant recipients, underscoring the need for tailored approaches in managing these patients. It paves the way for further research to refine strategies aimed at reducing PGD incidence and enhancing transplant outcomes in these distinct populations.
Collapse
Affiliation(s)
- Chitaru Kurihara
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Taisuke Kaiho
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin Thomae
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily Cerier
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kalvin Lung
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Diego Avella Patino
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Takahide Toyoda
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yuanqing Yan
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G. R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
10
|
Novo M, Nordén R, Westin J, Dellgren G, Böhmer J, Ricksten A, Magnusson JM. Donor Fractions of Cell-Free DNA Are Elevated During CLAD But Not During Infectious Complications After Lung Transplantation. Transpl Int 2024; 37:12772. [PMID: 39114640 PMCID: PMC11303165 DOI: 10.3389/ti.2024.12772] [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: 01/30/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
During the last few years, cell-free DNA (cfDNA) has emerged as a possible non-invasive biomarker for prediction of complications after lung transplantation. We previously published a proof-of-concept study using a digital droplet polymerase chain reaction (ddPCR)-based method for detection of cfDNA. In the current study, we aimed to further evaluate the potential clinical usefulness of detecting chronic lung allograft dysfunction (CLAD) using three different ddPCR applications measuring and calculating the donor fraction (DF) of cfDNA as well as one method using the absolute amount of donor-derived cfDNA. We analyzed 246 serum samples collected from 26 lung transplant recipients. Nine of the patients had ongoing CLAD at some point during follow-up. All four methods showed statistically significant elevation of the measured variable in the CLAD samples compared to the non-CLAD samples. The results support the use of ddPCR-detected cfDNA as a potential biomarker for prediction of CLAD. These findings need to be validated in a subsequent prospective study.
Collapse
Affiliation(s)
- Mirza Novo
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rickard Nordén
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Westin
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Göran Dellgren
- Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jens Böhmer
- Pediatric Heart Center, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Clinic Frankfurt-Höchst, Frankfurt, Germany
| | - Anne Ricksten
- Department of Clinical Genetics and Genomics, Sahlgrenska Academy, University of Gothenbururg, Gothenburg, Sweden
| | - Jesper M. Magnusson
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
11
|
Ikeda M, Aoyama A, Fukuyama J, Okuda M, Yamazaki K, Minatoya K, Chen-Yoshikawa TF, Kayawake H, Tanaka S, Yamada Y, Yutaka Y, Ohsumi A, Nakajima D, Hamaji M, Date H. Beneficial effects of extracorporeal membrane oxygenation over cardiopulmonary bypass in living-donor lobar lung transplantation. JHLT OPEN 2024; 4:100070. [PMID: 40144253 PMCID: PMC11935482 DOI: 10.1016/j.jhlto.2024.100070] [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 Extracorporeal membrane oxygenation (ECMO) has been frequently used instead of cardiopulmonary bypass (CPB) as extracorporeal circulatory support during cadaveric lung transplantation. This study compared the outcomes of intraoperative CPB or ECMO in living-donor lobar lung transplantation (LDLLT). Methods CPB and ECMO were performed in 23 and 53 patients, respectively, who underwent initial bilateral LDLLT in our institution from 2008 to 2019. We retrospectively compared the short- and long-term outcomes between the 2 groups. Results Patient background, graft size-matching data, operation time, extracorporeal circulation time, and bleeding amount were not significantly different in the 2 groups. However, the CPB group required more transfusion than the ECMO group (6,860 vs 3,840 ml, respectively; p = 0.002). The rate of increase in body weight through LDLLT was 7.4% and 4.9% in CPB and ECMO groups, respectively (p = 0.040), and primary graft dysfunction scores were significantly worse in the CPB group. Postoperative ECMO support was required in 4 cases, and hospital death occurred in 1 patient exclusively in the CPB group. Chronic lung allograft dysfunction (CLAD) was diagnosed in 43.5% and 17.0% of patients in the CPB and ECMO groups, respectively (p = 0.021), and the 5-year CLAD-free survival was 55.8% and 72.7% of patients, respectively (p = 0.013). Conclusions Intraoperative ECMO reduced primary graft dysfunction, possibly due to the lower requirement for intraoperative transfusion and less intraoperative weight gain causing systemic edema. The beneficial effect of ECMO in the early phase may result in less CLAD development in the long-term follow-up after LDLLT.
Collapse
Affiliation(s)
- Masaki Ikeda
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Aoyama
- Department of Thoracic Surgery, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Junya Fukuyama
- Department of Medical Equipment, Kyoto University Hospital, Kyoto, Japan
| | - Masanori Okuda
- Department of Medical Equipment, Kyoto University Hospital, Kyoto, Japan
| | - Kazuhiro Yamazaki
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Hidenao Kayawake
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yojiro Yutaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
Niroomand A, Hirdman G, Bèchet N, Ghaidan H, Stenlo M, Kjellström S, Isaksson M, Broberg E, Pierre L, Hyllén S, Olm F, Lindstedt S. Proteomic Analysis of Primary Graft Dysfunction in Porcine Lung Transplantation Reveals Alveolar-Capillary Barrier Changes Underlying the High Particle Flow Rate in Exhaled Breath. Transpl Int 2024; 37:12298. [PMID: 38741700 PMCID: PMC11089893 DOI: 10.3389/ti.2024.12298] [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: 10/25/2023] [Accepted: 03/19/2024] [Indexed: 05/16/2024]
Abstract
Primary graft dysfunction (PGD) remains a challenge for lung transplantation (LTx) recipients as a leading cause of poor early outcomes. New methods are needed for more detailed monitoring and understanding of the pathophysiology of PGD. The measurement of particle flow rate (PFR) in exhaled breath is a novel tool to monitor and understand the disease at the proteomic level. In total, 22 recipient pigs underwent orthotopic left LTx and were evaluated for PGD on postoperative day 3. Exhaled breath particles (EBPs) were evaluated by mass spectrometry and the proteome was compared to tissue biopsies and bronchoalveolar lavage fluid (BALF). Findings were confirmed in EBPs from 11 human transplant recipients. Recipients with PGD had significantly higher PFR [686.4 (449.7-8,824.0) particles per minute (ppm)] compared to recipients without PGD [116.6 (79.7-307.4) ppm, p = 0.0005]. Porcine and human EBP proteins recapitulated proteins found in the BAL, demonstrating its utility instead of more invasive techniques. Furthermore, adherens and tight junction proteins were underexpressed in PGD tissue. Histological and proteomic analysis found significant changes to the alveolar-capillary barrier explaining the high PFR in PGD. Exhaled breath measurement is proposed as a rapid and non-invasive bedside measurement of PGD.
Collapse
Affiliation(s)
- Anna Niroomand
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Rutgers Robert Wood Johnson University Hospital, New Brunswick, NJ, United States
| | - Gabriel Hirdman
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Nicholas Bèchet
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Haider Ghaidan
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transpantation, Skåne University Hospital, Lund, Sweden
| | - Martin Stenlo
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | | | - Marc Isaksson
- Department of Clinical Sciences, BioMS, Lund, Sweden
| | - Ellen Broberg
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Leif Pierre
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transpantation, Skåne University Hospital, Lund, Sweden
| | - Snejana Hyllén
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Franziska Olm
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transpantation, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
13
|
Gouchoe DA, Whitson BA, Rosenheck J, Henn MC, Mokadam NA, Ramsammy V, Kirkby S, Nunley D, Ganapathi AM. Long-Term Survival Following Primary Graft Dysfunction Development in Lung Transplantation. J Surg Res 2024; 296:47-55. [PMID: 38219506 DOI: 10.1016/j.jss.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/14/2023] [Accepted: 12/17/2023] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Primary graft dysfunction (PGD) is a known risk factor for early mortality following lung transplant (LT). However, the outcomes of patients who achieve long-term survival following index hospitalization are unknown. We aimed to determine the long-term association of PGD grade 3 (PGD3) in patients without in-hospital mortality. METHODS LT recipients were identified from the United Network for Organ Sharing Database. Patients were stratified based on the grade of PGD at 72 h (No PGD, Grade 1/2 or Grade 3). Groups were assessed with comparative statistics. Long-term survival was evaluated using Kaplan-Meier methods and a multivariable shared frailty model including recipient, donor, and transplant characteristics. RESULTS The PGD3 group had significantly increased length of stay, dialysis, and treated rejection post-transplant (P < 0.001). Unadjusted survival analysis revealed a significant difference in long-term survival (P < 0.001) between groups; however, following adjustment, PGD3 was not independently associated with long-term survival (hazard ratio: 0.972; 95% confidence interval: 0.862-1.096). Increased mortality was significantly associated with increased recipient age and treated rejection. Decreased mortality was significantly associated with no donor diabetes, bilateral LT as compared to single LT, transplant in 2015-2016 and 2017-2018, and no post-transplant dialysis. CONCLUSIONS While PGD3 remains a challenge post LT, PGD3 at 72 h is not independently associated with decreased long-term survival, while complications such as dialysis and rejection are, in patients who survive index hospitalization. Transplant providers should be aggressive in preventing further complications in recipients with severe PGD to minimize the negative association on long-term survival.
Collapse
Affiliation(s)
- Doug A Gouchoe
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; 88th Surgical Operations Squadron, Wright-Patterson Medical Center, WPAFB, Columbus, Ohio
| | - Bryan A Whitson
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Justin Rosenheck
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Matthew C Henn
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Nahush A Mokadam
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Verai Ramsammy
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Stephen Kirkby
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - David Nunley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Asvin M Ganapathi
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
14
|
Michelson AP, Oh I, Gupta A, Puri V, Kreisel D, Gelman AE, Nava R, Witt CA, Byers DE, Halverson L, Vazquez-Guillamet R, Payne PRO, Hachem RR. Developing machine learning models to predict primary graft dysfunction after lung transplantation. Am J Transplant 2024; 24:458-467. [PMID: 37468109 DOI: 10.1016/j.ajt.2023.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
Primary graft dysfunction (PGD) is the leading cause of morbidity and mortality in the first 30 days after lung transplantation. Risk factors for the development of PGD include donor and recipient characteristics, but how multiple variables interact to impact the development of PGD and how clinicians should consider these in making decisions about donor acceptance remain unclear. This was a single-center retrospective cohort study to develop and evaluate machine learning pipelines to predict the development of PGD grade 3 within the first 72 hours of transplantation using donor and recipient variables that are known at the time of donor offer acceptance. Among 576 bilateral lung recipients, 173 (30%) developed PGD grade 3. The cohort underwent a 75% to 25% train-test split, and lasso regression was used to identify 11 variables for model development. A K-nearest neighbor's model showing the best calibration and performance with relatively small confidence intervals was selected as the final predictive model with an area under the receiver operating characteristics curve of 0.65. Machine learning models can predict the risk for development of PGD grade 3 based on data available at the time of donor offer acceptance. This may improve donor-recipient matching and donor utilization in the future.
Collapse
Affiliation(s)
- Andrew P Michelson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA; Institute for Informatics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Inez Oh
- Institute for Informatics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Aditi Gupta
- Institute for Informatics, Washington University School of Medicine, Saint Louis, Missouri, USA; Division of Biostatistics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Varun Puri
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Andrew E Gelman
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ruben Nava
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Chad A Witt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Laura Halverson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Rodrigo Vazquez-Guillamet
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Philip R O Payne
- Institute for Informatics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ramsey R Hachem
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
15
|
Yu H, Wang J, Liu M, Hu C, Sun J, Xu B, Lu S, Huang D, Pang Q, Hu C. Metformin alleviates lung ischemia-reperfusion injury in a rat lung transplantation model. Exp Lung Res 2024; 50:15-24. [PMID: 38317565 DOI: 10.1080/01902148.2023.2301615] [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: 02/24/2023] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Background: Lung ischemia-reperfusion injury (LIRI) is among the complications observed after lung transplantation and is associated with morbidity and mortality. Preconditioning of the donor lung before organ retrieval may improve organ quality after transplantation. We investigated whether preconditioning with metformin (Met) ameliorates LIRI after lung transplantation. Methods: Twenty Lewis rats were randomly divided into the sham, LIRI, and Met groups. The rats in the LIRI and Met groups received saline and Met, respectively, via oral gavage. Subsequently, a donor lung was harvested and kept in cold storage for 8 h. The LIRI and Met groups then underwent left lung transplantation. After 2 h of reperfusion, serum and transplanted lung tissues were examined. Results: The partial pressure of oxygen (PaO2) was greater in the Met group than in the LIRI group. In the Met group, wet-to-dry (W/D) weight ratios, inflammatory factor levels, oxidative stress levels and apoptosis levels were notably decreased. Conclusions: Met protects against ischemia-reperfusion injury after lung transplantation in rats, and its therapeutic effect is associated with its anti-inflammatory, antioxidative, and antiapoptotic properties.
Collapse
Affiliation(s)
- Huizhi Yu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jing Wang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Mingzhao Liu
- Department of Lung Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Chunlan Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jiaojiao Sun
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Bo Xu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shunmei Lu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Dongxiao Huang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Qingfeng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chunxiao Hu
- Department of Transplant Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
16
|
Wong KHM, Hsin KYM. Primary graft dysfunction in lung transplantation: still a thorn in the side of lung transplant. J Thorac Dis 2024; 16:1-5. [PMID: 38410540 PMCID: PMC10894369 DOI: 10.21037/jtd-23-1618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/12/2023] [Indexed: 02/28/2024]
Affiliation(s)
- Kwun Hung Max Wong
- Department of Cardiothoracic Surgery, Queen Mary Hospital, Hong Kong, China
| | | |
Collapse
|
17
|
Anderson MR, Cantu E, Shashaty M, Benvenuto L, Kalman L, Palmer SM, Singer JP, Gallop R, Diamond JM, Hsu J, Localio AR, Christie JD. Body Mass Index and Cause-Specific Mortality after Lung Transplantation in the United States. Ann Am Thorac Soc 2023; 20:825-833. [PMID: 36996331 PMCID: PMC10257034 DOI: 10.1513/annalsats.202207-613oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/29/2023] [Indexed: 04/01/2023] Open
Abstract
Rationale: Low and high body mass index (BMI) are associated with increased mortality after lung transplantation. Why extremes of BMI might increase risk of death is unknown. Objectives: To estimate the association of extremes of BMI with causes of death after transplantation. Methods: We performed a retrospective study of the United Network for Organ Sharing database, including 26,721 adults who underwent lung transplantation in the United States between May 4, 2005, and December 2, 2020. We mapped 76 reported causes of death into 16 distinct groups. We estimated cause-specific hazards for death from each cause using Cox models. Results: Relative to a subject with a BMI of 24 kg/m2, a subject with a BMI of 16 kg/m2 had 38% (hazard ratio [HR], 1.38; 95% confidence interval [95% CI], 0.99-1.90), 82% (HR, 1.82; 95% CI, 1.34-2.46), and 62% (HR, 1.62; 95% CI, 1.18-2.22) increased hazards of death from acute respiratory failure, chronic lung allograft dysfunction (CLAD), and infection, respectively, and a subject with a BMI of 36 kg/m2 had 44% (HR, 1.44; 95% CI, 0.97-2.12), 42% (HR, 1.42; 95% CI, 0.93-2.15), and 185% (HR, 2.85; 95% CI, 1.28-6.33) increased hazards of death from acute respiratory failure, CLAD, and primary graft dysfunction, respectively. Conclusions: Low BMI is associated with increased risk of death from infection, acute respiratory failure, and CLAD after lung transplantation, whereas high BMI is associated with increased risk of death from primary graft dysfunction, acute respiratory failure, and CLAD.
Collapse
Affiliation(s)
| | | | - Michael Shashaty
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Luke Benvenuto
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Columbia University, New York, New York
| | - Laurel Kalman
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Scott M. Palmer
- Division of Pulmonary Medicine, Department of Medicine, Duke University, Durham, North Carolina
| | - Jonathan P. Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California; and
| | - Robert Gallop
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Mathematics, West Chester University, West Chester, Pennsylvania
| | - Joshua M. Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Jesse Hsu
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - A. Russell Localio
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| |
Collapse
|
18
|
Wong W, Johnson B, Cheng PC, Josephson MB, Maeda K, Berg RA, Kawut SM, Harhay MO, Goldfarb SB, Yehya N, Himebauch AS. Primary graft dysfunction grade 3 following pediatric lung transplantation is associated with chronic lung allograft dysfunction. J Heart Lung Transplant 2023; 42:669-678. [PMID: 36639317 PMCID: PMC10811698 DOI: 10.1016/j.healun.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Severe primary graft dysfunction (PGD) is associated with the development of bronchiolitis obliterans syndrome (BOS), the most common form of chronic lung allograft dysfunction (CLAD), in adults. However, PGD associations with long-term outcomes following pediatric lung transplantation are unknown. We hypothesized that PGD grade 3 (PGD 3) at 48- or 72-hours would be associated with shorter CLAD-free survival following pediatric lung transplantation. METHODS This was a single center retrospective cohort study of patients ≤ 21 years of age who underwent bilateral lung transplantation between 2005 and 2019 with ≥ 1 year of follow-up. PGD and CLAD were defined by published criteria. We evaluated the association of PGD 3 at 48- or 72-hours with CLAD-free survival by using time-to-event analyses. RESULTS Fifty-one patients were included (median age 12.7 years; 51% female). The most common transplant indications were cystic fibrosis (29%) and pulmonary hypertension (20%). Seventeen patients (33%) had PGD 3 at either 48- or 72-hours. In unadjusted analysis, PGD 3 was associated with an increased risk of CLAD or mortality (HR 2.10, 95% CI 1.01-4.37, p=0.047). This association remained when adjusting individually for multiple potential confounders. There was evidence of effect modification by sex (interaction p = 0.055) with the association of PGD 3 and shorter CLAD-free survival driven predominantly by males (HR 4.73, 95% CI 1.44-15.6) rather than females (HR 1.23, 95% CI 0.47-3.20). CONCLUSIONS PGD 3 at 48- or 72-hours following pediatric lung transplantation was associated with shorter CLAD-free survival. Sex may be a modifier of this association.
Collapse
Affiliation(s)
- Wai Wong
- Department of Pediatrics, Division of Pulmonary Medicine and Respiratory Diseases, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Brandy Johnson
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Pi Chun Cheng
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Division of Pediatric Pulmonology, Allergy, and Sleep Medicine, Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, Indiana
| | - Maureen B Josephson
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Katsuhide Maeda
- Department of Surgery, Division of Cardiothoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert A Berg
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael O Harhay
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samuel B Goldfarb
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Minnesota, Masonic Children's Hospital, Minneapolis, Minnesota
| | - Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adam S Himebauch
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Yamanashi K, Ohsumi A, Oda H, Tanaka S, Yamada Y, Nakajima D, Date H. Reduction of donor mononuclear phagocytes with clodronate-liposome during ex vivo lung perfusion attenuates ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2023; 165:e181-e203. [PMID: 36404143 DOI: 10.1016/j.jtcvs.2022.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Clodronate-liposome is used for depleting mononuclear phagocytes associated with ischemia-reperfusion injury. We hypothesized that administration of clodronate-liposome into the perfusate during ex vivo lung perfusion could reduce mononuclear phagocytes and attenuate ischemia-reperfusion injury. METHODS First, the number of mononuclear phagocytes in flushed grafts (minimum cold ischemic time, 6-hour cold ischemic time, 15-hour cold ischemic time, and 18-hour cold ischemic time; n = 6 each) was determined using flow cytometry. Second, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 5 each). In the clodronate group, clodronate-liposome is administered into the perfusate. After 4 hours of ex vivo lung perfusion, the number of mononuclear phagocytes in the perfusate and lung tissues was measured. Third, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 6 each). After 4 hours of ex vivo lung perfusion, the left lungs were transplanted and reperfused for 2 hours. Lung function was evaluated, and samples were analyzed. RESULTS First, mononuclear phagocytes remain in flushed grafts after prolonged cold ischemia. Second, the number of mononuclear phagocytes in lung tissues after ex vivo lung perfusion was significantly reduced in the clodronate group (P = .008). Third, lung compliance and vascular resistance during ex vivo lung perfusion were significantly improved in the clodronate group (P < .001 for both). Blood oxygenation and pulmonary edema were significantly improved in the clodronate group after 2 hours of reperfusion (P = .015 and P = .026, respectively). Histological findings showed reduced lung injury in the clodronate group (P = .013). CONCLUSIONS Administration of clodronate-liposome into the perfusate during ex vivo lung perfusion resulted in a significant reduction of mononuclear phagocytes in donor lungs, leading to attenuation of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Keiji Yamanashi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiromi Oda
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
20
|
Beeckmans H, Bos S, Vos R, Glanville AR. Acute Rejection and Chronic Lung Allograft Dysfunction: Obstructive and Restrictive Allograft Dysfunction. Clin Chest Med 2023; 44:137-157. [PMID: 36774160 DOI: 10.1016/j.ccm.2022.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Lung transplantation is an established treatment of well-selected patients with end-stage respiratory diseases. However, lung transplant recipients have the highest rates of acute and chronic rejection among transplanted solid organs. Owing to ongoing alloimmune recognition and associated immune-driven airway/vascular remodeling, precipitated by multifactorial, endogenous or exogenous, post-transplant injuries to the bronchovascular axis of the secondary pulmonary lobule, most lung transplant recipients will suffer from a pathophysiological decline of their allograft, either functionally and/or structurally. This review discusses current knowledge, barriers, and gaps in acute cellular rejection and chronic lung allograft dysfunction-the greatest impediment to long-term post-transplant survival.
Collapse
Affiliation(s)
- Hanne Beeckmans
- Department of Chronic Diseases and Metabolism, KU Leuven, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Leuven, Belgium
| | - Saskia Bos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium; Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Robin Vos
- Department of Chronic Diseases and Metabolism, KU Leuven, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Leuven, Belgium; Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| | | |
Collapse
|
21
|
Toyoda T, Cerier EJ, Manerikar AJ, Kandula V, Bharat A, Kurihara C. Recipient, donor, and surgical factors leading to primary graft dysfunction after lung transplant. J Thorac Dis 2023; 15:399-409. [PMID: 36910052 PMCID: PMC9992558 DOI: 10.21037/jtd-22-974] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/05/2022] [Indexed: 02/10/2023]
Abstract
Background Primary graft dysfunction is a major cause of early mortality following lung transplantation. The International Society for Heart and Lung Transplantation subdivides it into 4 grades of increasing severity. Methods A retrospective review of the institutional lung transplant database from March 2018 to September 2021 was performed. Patients were stratified into three groups: primary graft dysfunction grade 0 patients, grade 1 or 2 patients, and grade 3 patients. Recipient, donor, and surgical variables were analyzed by logistic regression analysis to identify risk factors for primary graft dysfunction grade 1 or 2 and grade 3. Results Primary graft dysfunction grade 1 to 3 occurred in 45.0% of the cohort (n=68) of whom 33.3% (n=23) had primary graft dysfunction grade 3. Longer operative time was more common in primary graft dysfunction grade 1 to 3 patients (P<0.001). The 1-year survival of the patients with primary graft dysfunction grade 3 was lower than the others (grade 0-2 vs. 3, 93.7% vs. 65.2%, P=0.0006). Univariate analysis showed that acute respiratory distress syndrome, operative time, and intraoperative veno-arterial extracorporeal membrane oxygenation use were risk factors for primary graft dysfunction grades 1 or 2 and grade 3. Multivariate analysis identified that intraoperative veno-arterial extracorporeal membrane oxygenation use was an independent risk factor of primary graft dysfunction grade 1 or 2. Patients with an operative time of more than 8.18 hours had significantly higher incidence of primary graft dysfunction grade 3, acute kidney injury, and digital ischemia. Conclusions The calculated predictors of primary graft dysfunction grade 1 or 2 were similar to those of primary graft dysfunction grade 3.
Collapse
Affiliation(s)
- Takahide Toyoda
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily Jeong Cerier
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adwaiy Jayant Manerikar
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Viswajit Kandula
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ankit Bharat
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chitaru Kurihara
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
22
|
Takahashi T, Terada Y, Pasque MK, Nava RG, Kozower BD, Meyers BF, Patterson GA, Kreisel D, Puri V, Hachem RR. Outcomes of Extracorporeal Membrane Oxygenation for Primary Graft Dysfunction After Lung Transplantation. Ann Thorac Surg 2023; 115:1273-1280. [PMID: 36634836 DOI: 10.1016/j.athoracsur.2022.12.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Primary graft dysfunction (PGD) is the leading cause of death in the first 30 days after lung transplantation and is also associated with worse long-term outcomes. Outcomes of patients with PGD grade 3 requiring extracorporeal membrane oxygenation (ECMO) support after lung transplantation have yet to be well described. We sought to describe short- and long-term outcomes for patients with PGD grade 3 who required ECMO support. METHODS This is a single-center retrospective cohort study of patients undergoing lung transplantation. We stratified patients with PGD grade 3 into non-ECMO, venoarterial (VA) ECMO, and venovenous (VV) ECMO groups after transplantation. We then compared the outcomes between the groups. RESULTS Of 773 lung transplant recipients, PGD grade 3 developed in 204 (26%) at any time in the first 72 hours after lung transplantation. Of these, 13 (5%) required VA ECMO and 25 (10%) required VV ECMO support. The 30-day, 1-year, and 5-year survival in the VA ECMO group was 62%, 54%, and 43% compared with 96%, 84%, and 65% in the VV ECMO group and 99%, 94%, and 71% in the non-ECMO group. Multivariable Cox regression analysis showed that VA ECMO was associated with increased mortality (hazard ratio, 2.37; 95% CI, 1.06-5.28; P = .04). CONCLUSIONS Patients who required VA ECMO support for PGD grade 3 have significantly worse survival compared with those who did not require ECMO and those who required VV ECMO support. This suggests that VA ECMO treatment of patients with PGD grade 3 after lung transplantation can be a predictable risk factor for mortality.
Collapse
Affiliation(s)
- Tsuyoshi Takahashi
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri.
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Michael K Pasque
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Ruben G Nava
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Benjamin D Kozower
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Bryan F Meyers
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - G Alexander Patterson
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Varun Puri
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Ramsey R Hachem
- Division of Pulmonary & Critical Care, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
23
|
McGinniss JE, Whiteside SA, Deek RA, Simon-Soro A, Graham-Wooten J, Oyster M, Brown MD, Cantu E, Diamond JM, Li H, Christie JD, Bushman FD, Collman RG. The Lung Allograft Microbiome Associates with Pepsin, Inflammation, and Primary Graft Dysfunction. Am J Respir Crit Care Med 2022; 206:1508-1521. [PMID: 36103583 PMCID: PMC9757091 DOI: 10.1164/rccm.202112-2786oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/14/2022] [Indexed: 12/24/2022] Open
Abstract
Rationale: Primary graft dysfunction (PGD) is the principal cause of early morbidity and mortality after lung transplantation. The lung microbiome has been implicated in later transplantation outcomes but has not been investigated in PGD. Objectives: To define the peritransplant bacterial lung microbiome and relationship to host response and PGD. Methods: This was a single-center prospective cohort study. Airway lavage samples from donor lungs before organ procurement and recipient allografts immediately after implantation underwent bacterial 16S ribosomal ribonucleic acid gene sequencing. Recipient allograft samples were analyzed for cytokines by multiplex array and pepsin by ELISA. Measurements and Main Results: We enrolled 139 transplant subjects and obtained donor lung (n = 109) and recipient allograft (n = 136) samples. Severe PGD (persistent grade 3) developed in 15 subjects over the first 72 hours, and 40 remained without PGD (persistent grade 0). The microbiome of donor lungs differed from healthy lungs, and recipient allograft microbiomes differed from donor lungs. Development of severe PGD was associated with enrichment in the immediate postimplantation lung of oropharyngeal anaerobic taxa, particularly Prevotella. Elevated pepsin, a gastric biomarker, and a hyperinflammatory cytokine profile were present in recipient allografts in severe PGD and strongly correlated with microbiome composition. Together, immediate postimplantation allograft Prevotella/Streptococcus ratio, pepsin, and indicator cytokines were associated with development of severe PGD during the 72-hour post-transplantation period (area under the curve = 0.81). Conclusions: Lung allografts that develop PGD have a microbiome enriched in anaerobic oropharyngeal taxa, elevated gastric pepsin, and hyperinflammatory phenotype. These findings suggest a possible role for peritransplant aspiration in PGD, a potentially actionable mechanism that warrants further investigation.
Collapse
Affiliation(s)
- John E. McGinniss
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | | | - Aurea Simon-Soro
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Michelle Oyster
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Melanie D. Brown
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Joshua M. Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Hongzhe Li
- Department of Epidemiology, Biostatistics, and Informatics
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ronald G. Collman
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Van De Wauwer C, van Suylen V, Zhang ZL, Verschuuren EAM, van der Bij W, Gan CT, Ubbink R, Erasmus ME. Is logistically motivated ex vivo lung perfusion a good idea? FRONTIERS IN TRANSPLANTATION 2022; 1:988950. [PMID: 38994392 PMCID: PMC11235284 DOI: 10.3389/frtra.2022.988950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/14/2022] [Indexed: 07/13/2024]
Abstract
Ex vivo lung perfusion (EVLP) is a technique for reconditioning and evaluating lungs. However, the use of EVLP for logistical reasons is still under discussion. In this retrospective study, all EVLPs performed between July 2012 and October 2019 were analyzed for ventilation and perfusion data. After transplantation, primary graft dysfunction (PGD), lung function, chronic lung allograft dysfunction (CLAD)-free survival, and overall survival were analyzed. Fifty EVLPs were performed: seventeen logistic EVLPs led to 15 lung transplantations (LT) and two rejections (LR), and 33 medical EVLPs resulted in 26 lung transplantations (MT) and seven rejections (MR). Pre-EVLP PaO2 was lower for MT than LT (p < 0.05). Dynamic lung compliance remained stable in MT and LT but decreased in MR and LR. Plateau airway pressure started at a higher level in MR (p < 0.05 MT vs. MR at T60) and increased further in LR. After transplantation, there were no differences between MT and LT in PGD, lung function, CLAD-free survival, and overall survival. In addition, the LT group was compared with a cohort group receiving standard donor lungs without EVLP (LTx). There were no significant differences between LT and LTx for PGD, CLAD-free survival, and overall survival. FVC was significantly lower in LT than in LTx after 1 year (p = 0.005). We found that LT lungs appear to perform better than MT lungs on EVLP. In turn, the outcome in the LT group was comparable with the LTx group. Overall, lung transplantation after EVLP for logistic reasons is safe and makes transplantation timing controllable.
Collapse
Affiliation(s)
- Caroline Van De Wauwer
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Vincent van Suylen
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Zhang L. Zhang
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Erik A. M. Verschuuren
- Department of Pulmonary Diseases and Lung Transplantation, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Wim van der Bij
- Department of Pulmonary Diseases and Lung Transplantation, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - C. Tji Gan
- Department of Pulmonary Diseases and Lung Transplantation, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Rinse Ubbink
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Michiel E. Erasmus
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| |
Collapse
|
25
|
Shepherd HM, Gauthier JM, Terada Y, Li W, Krupnick AS, Gelman AE, Kreisel D. Updated Views on Neutrophil Responses in Ischemia-Reperfusion Injury. Transplantation 2022; 106:2314-2324. [PMID: 35749228 PMCID: PMC9712152 DOI: 10.1097/tp.0000000000004221] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ischemia-reperfusion injury is an inevitable event during organ transplantation and represents a primary risk factor for the development of early graft dysfunction in lung, heart, liver, and kidney transplant recipients. Recent studies have implicated recipient neutrophils as key mediators of this process and also have found that early innate immune responses after transplantation can ultimately augment adaptive alloimmunity and affect late graft outcomes. Here, we discuss signaling pathways involved in neutrophil recruitment and activation after ischemia-mediated graft injury in solid organ transplantation with an emphasis on lung allografts, which have been the focus of recent studies. These findings suggest novel therapeutic interventions that target ischemia-reperfusion injury-mediated graft dysfunction in transplant recipients.
Collapse
Affiliation(s)
- Hailey M. Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Yuriko Terada
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | | | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
26
|
Cantu E, Diamond JM, Cevasco M, Suzuki Y, Crespo M, Clausen E, Dallara L, Ramon CV, Harmon MT, Bermudez C, Benvenuto L, Anderson M, Wille KM, Weinacker A, Dhillon GS, Orens J, Shah P, Merlo C, Lama V, McDyer J, Snyder L, Palmer S, Hartwig M, Hage CA, Singer J, Calfee C, Kukreja J, Greenland JR, Ware LB, Localio R, Hsu J, Gallop R, Christie JD. Contemporary trends in PGD incidence, outcomes, and therapies. J Heart Lung Transplant 2022; 41:1839-1849. [PMID: 36216694 PMCID: PMC9990084 DOI: 10.1016/j.healun.2022.08.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We sought to describe trends in extracorporeal membrane oxygenation (ECMO) use, and define the impact on PGD incidence and early mortality in lung transplantation. METHODS Patients were enrolled from August 2011 to June 2018 at 10 transplant centers in the multi-center Lung Transplant Outcomes Group prospective cohort study. PGD was defined as Grade 3 at 48 or 72 hours, based on the 2016 PGD ISHLT guidelines. Logistic regression and survival models were used to contrast between group effects for event (i.e., PGD and Death) and time-to-event (i.e., death, extubation, discharge) outcomes respectively. Both modeling frameworks accommodate the inclusion of potential confounders. RESULTS A total of 1,528 subjects were enrolled with a 25.7% incidence of PGD. Annual PGD incidence (14.3%-38.2%, p = .0002), median LAS (38.0-47.7 p = .009) and the use of ECMO salvage for PGD (5.7%-20.9%, p = .007) increased over the course of the study. PGD was associated with increased 1 year mortality (OR 1.7 [95% C.I. 1.2, 2.3], p = .0001). Bridging strategies were not associated with increased mortality compared to non-bridged patients (p = .66); however, salvage ECMO for PGD was significantly associated with increased mortality (OR 1.9 [1.3, 2.7], p = .0007). Restricted mean survival time comparison at 1-year demonstrated 84.1 days lost in venoarterial salvaged recipients with PGD when compared to those without PGD (ratio 1.3 [1.1, 1.5]) and 27.2 days for venovenous with PGD (ratio 1.1 [1.0, 1.4]). CONCLUSIONS PGD incidence continues to rise in modern transplant practice paralleled by significant increases in recipient severity of illness. Bridging strategies have increased but did not affect PGD incidence or mortality. PGD remains highly associated with mortality and is increasingly treated with salvage ECMO.
Collapse
Affiliation(s)
- Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marisa Cevasco
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yoshi Suzuki
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Crespo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily Clausen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laura Dallara
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christian V Ramon
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael T Harmon
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christian Bermudez
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Luke Benvenuto
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University School of Medicine, New York, New York
| | - Michaela Anderson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University School of Medicine, New York, New York
| | - Keith M Wille
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ann Weinacker
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Palo Alto, California
| | - Gundeep S Dhillon
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Palo Alto, California
| | - Jonathan Orens
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University Medical Center, Baltimore, Maryland
| | - Pali Shah
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University Medical Center, Baltimore, Maryland
| | - Christian Merlo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University Medical Center, Baltimore, Maryland
| | - Vibha Lama
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | - John McDyer
- Division of Pulmonary, Allergy, and Critical Care, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laurie Snyder
- Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina
| | - Scott Palmer
- Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina
| | - Matt Hartwig
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Chadi A Hage
- Division of Pulmonary, Allergy, Critical Care, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jonathan Singer
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, California
| | - Carolyn Calfee
- Department of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Russel Localio
- Division of Biostatistics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jesse Hsu
- Division of Biostatistics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert Gallop
- Department of Mathematics, West Chester University, West Chester, Pennsylvania
| | - Jason D Christie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Liu CR, Heid CA, Hauptmann E, Ali M, Pruszynski J, Pillai A, Banga A, Wait MA, Huffman LC, Peltz M, Hackmann AE, Jessen ME, Ring WS, Murala JS. Donor substance use and lung transplantation: A single center experience. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Perez AA, Shah RJ. Critical Care of the Lung Transplant Patient. Clin Chest Med 2022; 43:457-470. [PMID: 36116814 DOI: 10.1016/j.ccm.2022.04.007] [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: 11/03/2022]
Abstract
Lung transplantation is a therapeutic option for end-stage lung disease that improves survival and quality of life. Prelung transplant admission to the intensive care unit (ICU) for bridge to transplant with mechanical ventilation and extracorporeal membrane oxygenation (ECMO) is common. Primary graft dysfunction is an important immediate complication of lung transplantation with short- and long-term morbidity and mortality. Later transplant-related causes of respiratory failure necessitating ICU admission include acute cellular rejection, atypical infections, and chronic lung allograft dysfunction. Lung transplantation for COVID-19-related ARDS is increasingly common..
Collapse
Affiliation(s)
- Alyssa A Perez
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, 400 Parnassus Street, 5th Floor, San Francisco, CA 94143, USA.
| | - Rupal J Shah
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, 400 Parnassus Street, 5th Floor, San Francisco, CA 94143, USA
| |
Collapse
|
29
|
Ravichandran R, Bansal S, Rahman M, Sureshbabu A, Sankpal N, Fleming T, Bharat A, Mohanakumar T. Extracellular Vesicles Mediate Immune Responses to Tissue-Associated Self-Antigens: Role in Solid Organ Transplantations. Front Immunol 2022; 13:861583. [PMID: 35572510 PMCID: PMC9094427 DOI: 10.3389/fimmu.2022.861583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Transplantation is a treatment option for patients diagnosed with end-stage organ diseases; however, long-term graft survival is affected by rejection of the transplanted organ by immune and nonimmune responses. Several studies have demonstrated that both acute and chronic rejection can occur after transplantation of kidney, heart, and lungs. A strong correlation has been reported between de novo synthesis of donor-specific antibodies (HLA-DSAs) and development of both acute and chronic rejection; however, some transplant recipients with chronic rejection do not have detectable HLA-DSAs. Studies of sera from such patients demonstrate that immune responses to tissue-associated antigens (TaAgs) may also play an important role in the development of chronic rejection, either alone or in combination with HLA-DSAs. The synergistic effect between HLA-DSAs and antibodies to TaAgs is being established, but the underlying mechanism is yet to be defined. We hypothesize that HLA-DSAs damage the transplanted donor organ resulting in stress and leading to the release of extracellular vesicles, which contribute to chronic rejection. These vesicles express both donor human leukocyte antigen (HLA) and non-HLA TaAgs, which can activate antigen-presenting cells and lead to immune responses and development of antibodies to both donor HLA and non-HLA tissue-associated Ags. Extracellular vesicles (EVs) are released by cells under many circumstances due to both physiological and pathological conditions. Primarily employing clinical specimens obtained from human lung transplant recipients undergoing acute or chronic rejection, our group has demonstrated that circulating extracellular vesicles display both mismatched donor HLA molecules and lung-associated Ags (collagen-V and K-alpha 1 tubulin). This review focuses on recent studies demonstrating an important role of antibodies to tissue-associated Ags in the rejection of transplanted organs, particularly chronic rejection. We will also discuss the important role of extracellular vesicles released from transplanted organs in cross-talk between alloimmunity and autoimmunity to tissue-associated Ags after solid organ transplantation.
Collapse
Affiliation(s)
| | - Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Mohammad Rahman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Narendra Sankpal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Ankit Bharat
- Department of Surgery-Thoracic, Northwestern University, Chicago, IL, United States
| | | |
Collapse
|
30
|
Trinh BN, Brzezinski M, Kukreja J. Early Postoperative Management of Lung Transplant Recipients. Thorac Surg Clin 2022; 32:185-195. [PMID: 35512937 DOI: 10.1016/j.thorsurg.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The early postoperative period after lung transplantation is a critical time. Prompt recognition and treatment of primary graft dysfunction can alter long-term allograft function. Cardiovascular, gastrointestinal, renal, and hematologic derangements are common and require close management to limit their negative sequelae.
Collapse
Affiliation(s)
- Binh N Trinh
- Division of Cardiothoracic Surgery, University of California, San Francisco, 500 Parnassus Avenue, Suite MUW-405, San Francisco, CA 94143-0118, USA
| | - Marek Brzezinski
- Department of Anesthesia, University of California, San Francisco, 500 Parnassus Avenue, Suite MUW-405, San Francisco, CA 94143-0118, USA
| | - Jasleen Kukreja
- Division of Cardiothoracic Surgery, University of California, San Francisco, 500 Parnassus Avenue, Suite MUW-405, San Francisco, CA 94143-0118, USA.
| |
Collapse
|
31
|
Magnusson JM, Ricksten A, Dellgren G, Wasslavik C, Nordén R, Westin J, Boehmer J. Cell‐free DNA as a biomarker after lung transplantation: A proof‐of‐concept study. Immun Inflamm Dis 2022; 10:e620. [PMID: 35478446 PMCID: PMC9017613 DOI: 10.1002/iid3.620] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 02/28/2022] [Accepted: 03/21/2022] [Indexed: 11/12/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Jesper M. Magnusson
- Transplant Institute Sahlgrenska University Hospital Gothenburg Sweden
- Department of Respiratory Medicine, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Anne Ricksten
- Department of Clinical Chemistry Sahlgrenska University Hospital Gothenburg Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Göran Dellgren
- Transplant Institute Sahlgrenska University Hospital Gothenburg Sweden
- Department of Cardiothoracic Surgery, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Carina Wasslavik
- Department of Clinical Chemistry Sahlgrenska University Hospital Gothenburg Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Rickard Nordén
- Department of Clinical Microbiology Sahlgrenska University Hospital, Region Västra Götaland Gothenburg Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Johan Westin
- Department of Clinical Microbiology Sahlgrenska University Hospital, Region Västra Götaland Gothenburg Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| | - Jens Boehmer
- Department of Pediatrics, Queen Silvias Children's Hospital Sahlgrenska University Hospital Gothenburg Sweden
- Department of Cardiology, Institute of Medicine, Sahlgrenska University Hospital University of Gothenburg Gothenburg Sweden
| |
Collapse
|
32
|
Alder JK, Sutton RM, Iasella CJ, Nouraie M, Koshy R, Hannan SJ, Chan EG, Chen X, Zhang Y, Brown M, Popescu I, Veatch M, Saul M, Berndt A, Methé BA, Morris A, Pilewski JM, Sanchez PG, Morrell MR, Shapiro SD, Lindell KO, Gibson KF, Kass DJ, McDyer JF. Lung transplantation for idiopathic pulmonary fibrosis enriches for individuals with telomere-mediated disease. J Heart Lung Transplant 2022; 41:654-663. [PMID: 34933798 PMCID: PMC9038609 DOI: 10.1016/j.healun.2021.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is the most common indication for lung transplantation in North America and variants in telomere-maintenance genes are the most common identifiable cause of IPF. We reasoned that younger IPF patients are more likely to undergo lung transplantation and we hypothesized that lung transplant recipients would be enriched for individuals with telomere-mediated disease due to the earlier onset and more severe disease in these patients. METHODS Individuals with IPF who underwent lung transplantation or were evaluated in an interstitial lung disease specialty clinic who did not undergo lung transplantation were examined. Genetic evaluation was completed via whole genome sequencing (WGS) of 426 individuals and targeted sequencing for 5 individuals. Rare variants in genes previously associated with IPF were classified using the American College of Medical Genetics guidelines. Telomere length from WGS data was measured using TelSeq software. Patient characteristics were collected via medical record review. RESULTS Of 431 individuals, 149 underwent lung transplantation for IPF. The median age of diagnosis of transplanted vs non-transplanted individuals was significantly younger (60 years vs 70 years, respectively, p<0.0001). IPF lung transplant recipients (IPF-LTRs) were twice as likely to have telomere-related rare variants compared to non-transplanted individuals (24% vs 12%, respectively, p=0.0013). IPF-LTRs had shorter telomeres than non-transplanted IPF patients (p=0.0028) and >85% had telomeres below the age-adjusted mean. Post-transplant survival and CLAD were similar amongst IPF-LTRs with rare variants in telomere-maintenance genes compared to those without, as well as in those with short telomeres versus longer telomeres. CONCLUSIONS There is an enrichment for telomere-maintenance gene variants and short telomeres among IPF-LTRs. However, transplant outcomes of survival and CLAD do not differ by gene variants or telomere length within IPF-LTRs. Our findings support individual with telomere-mediated disease should not be excluded from lung transplantation and focusing research efforts on therapies directed toward individuals with short-telomere mediated disease.
Collapse
Affiliation(s)
- Jonathan K Alder
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Rachel M Sutton
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carlo J Iasella
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mehdi Nouraie
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ritchie Koshy
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stefanie J Hannan
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ernest G Chan
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoping Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yingze Zhang
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark Brown
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iulia Popescu
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melinda Veatch
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melissa Saul
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Annerose Berndt
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Barbara A Methé
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pablo G Sanchez
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew R Morrell
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steven D Shapiro
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kathleen O Lindell
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; College of Nursing, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin F Gibson
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel J Kass
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John F McDyer
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
33
|
Niroomand A, Qvarnström S, Stenlo M, Malmsjö M, Ingemansson R, Hyllén S, Lindstedt S. The role of mechanical ventilation in primary graft dysfunction in the postoperative lung transplant recipient: A single center study and literature review. Acta Anaesthesiol Scand 2022; 66:483-496. [PMID: 35014027 PMCID: PMC9303877 DOI: 10.1111/aas.14025] [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: 02/25/2021] [Revised: 12/10/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022]
Abstract
Background Primary graft dysfunction (PGD) is still a major complication in patients undergoing lung transplantation (LTx). Much is unknown about the effect of postoperative mechanical ventilation on outcomes, with debate on the best approach to ventilation. Aim/Purpose The goal of this study was to generate hypotheses on the association between postoperative mechanical ventilation settings and allograft size matching in PGD development. Method This is a retrospective study of LTx patients between September 2011 and September 2018 (n = 116). PGD was assessed according to the International Society of Heart and Lung Transplantation (ISHLT) criteria. Data were collected from medical records, including chest x‐ray assessments, blood gas analysis, mechanical ventilator parameters and spirometry. Results Positive end‐expiratory pressures (PEEP) of 5 cm H2O were correlated with lower rates of grade 3 PGD. Graft size was important as tidal volumes calculated according to the recipient yielded greater rates of PGD when low volumes were used, a correlation that was lost when donor metrics were used. Conclusion Our results highlight a need for greater investigation of the role donor characteristics play in determining post‐operative ventilation of a lung transplant recipient. The mechanical ventilation settings on postoperative LTx recipients may have an implication for the development of acute graft dysfunction. Severe PGD was associated with the use of a PEEP higher than 5 and lower tidal volumes and oversized lungs were associated with lower long‐term mortality. Lack of association between ventilatory settings and survival may point to the importance of other variables than ventilation in the development of PGD.
Collapse
Affiliation(s)
- Anna Niroomand
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
- Wallenberg Center for Molecular Medicine Lund University Lund Sweden
- Lund Stem Cell Center Lund University Lund Sweden
- Department of Clinical Sciences Lund University Lund Sweden
- Rutgers Robert University New Brunswick New Jersey USA
| | - Sara Qvarnström
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
| | - Martin Stenlo
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
- Lund Stem Cell Center Lund University Lund Sweden
- Department of Clinical Sciences Lund University Lund Sweden
| | - Malin Malmsjö
- Department of Clinical Sciences Lund University Lund Sweden
| | - Richard Ingemansson
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
- Department of Clinical Sciences Lund University Lund Sweden
| | - Snejana Hyllén
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
- Lund Stem Cell Center Lund University Lund Sweden
- Department of Clinical Sciences Lund University Lund Sweden
| | - Sandra Lindstedt
- Department of Cardiothoracic Anesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation Skåne University Hospital Lund University Lund Sweden
- Wallenberg Center for Molecular Medicine Lund University Lund Sweden
- Lund Stem Cell Center Lund University Lund Sweden
- Department of Clinical Sciences Lund University Lund Sweden
| |
Collapse
|
34
|
Van Slambrouck J, Van Raemdonck D, Vos R, Vanluyten C, Vanstapel A, Prisciandaro E, Willems L, Orlitová M, Kaes J, Jin X, Jansen Y, Verleden GM, Neyrinck AP, Vanaudenaerde BM, Ceulemans LJ. A Focused Review on Primary Graft Dysfunction after Clinical Lung Transplantation: A Multilevel Syndrome. Cells 2022; 11:cells11040745. [PMID: 35203392 PMCID: PMC8870290 DOI: 10.3390/cells11040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
Primary graft dysfunction (PGD) is the clinical syndrome of acute lung injury after lung transplantation (LTx). However, PGD is an umbrella term that encompasses the ongoing pathophysiological and -biological mechanisms occurring in the lung grafts. Therefore, we aim to provide a focused review on the clinical, physiological, radiological, histological and cellular level of PGD. PGD is graded based on hypoxemia and chest X-ray (CXR) infiltrates. High-grade PGD is associated with inferior outcome after LTx. Lung edema is the main characteristic of PGD and alters pulmonary compliance, gas exchange and circulation. A conventional CXR provides a rough estimate of lung edema, while a chest computed tomography (CT) results in a more in-depth analysis. Macroscopically, interstitial and alveolar edema can be distinguished below the visceral lung surface. On the histological level, PGD correlates to a pattern of diffuse alveolar damage (DAD). At the cellular level, ischemia-reperfusion injury (IRI) is the main trigger for the disruption of the endothelial-epithelial alveolar barrier and inflammatory cascade. The multilevel approach integrating all PGD-related aspects results in a better understanding of acute lung failure after LTx, providing novel insights for future therapies.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Cedric Vanluyten
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Elena Prisciandaro
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Pulmonary Circulation Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium;
| | - Michaela Orlitová
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yanina Jansen
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Geert M. Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arne P. Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
- Department of Anesthesiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
35
|
Chronic lung allograft dysfunction. Indian J Thorac Cardiovasc Surg 2021; 38:318-325. [DOI: 10.1007/s12055-021-01228-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 10/19/2022] Open
|
36
|
Li D, Weinkauf J, Kapasi A, Hirji A, Varughese R, Lien D, Nagendran J, Halloran K. Baseline lung allograft dysfunction in primary graft dysfunction survivors after lung transplantation. Respir Med 2021; 188:106617. [PMID: 34571454 DOI: 10.1016/j.rmed.2021.106617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Primary graft dysfunction (PGD) after lung transplantation has previously been associated with increased risk of death and chronic lung allograft dysfunction (CLAD), but the relationship to baseline lung allograft dysfunction (BLAD), where graft function fails to normalize, is not known. METHODS We reviewed all double lung transplant recipients transplanted in our program 2004-2016. We defined PGD and CLAD as per recent consensus definitions and BLAD as failure to achieve both FEV1 and FVC ≥80% predicted on 2 consecutive tests ≥3 weeks apart. We used logistic and proportional hazards regression to test the association between severe high-grade PGD (PGD3) with BLAD and CLAD respectively, adjusting for known and identified confounders. RESULTS 446 patients met inclusion criteria and 76 (17%) developed PGD3 at 48- or 72-h post-transplant. PGD3 occurred more frequently in patients with interstitial lung disease or pulmonary vascular disease, those with higher BMIs and recipients of older donors. PGD3 was associated with more frequent (58% vs. 36%; p = 0.0008) and more severe BLAD (p < 0.0001) and increased BLAD risk in an adjusted model (OR 2.00 [95% CI 1.13-3.60]; p = 0.0182). PGD3 was not associated with CLAD frequency, severity or time to CLAD onset in an adjusted model (HR 1.10 (95% CI 0.64-1.78), p = 0.7226). CONCLUSION Severe PGD was associated with increased risk and severity of BLAD but not CLAD. The mechanisms via which PGD may mediate baseline function warrant further investigation.
Collapse
Affiliation(s)
- David Li
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Justin Weinkauf
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Ali Kapasi
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Alim Hirji
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Rhea Varughese
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Dale Lien
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada
| | - Jayan Nagendran
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, T6G 2BY, Canada
| | - Kieran Halloran
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, T6G 2G3, Canada.
| |
Collapse
|
37
|
Ali A, Wang A, Ribeiro RVP, Beroncal EL, Baciu C, Galasso M, Gomes B, Mariscal A, Hough O, Brambate E, Abdelnour-Berchtold E, Michaelsen V, Zhang Y, Gazzalle A, Fan E, Brochard L, Yeung J, Waddell T, Liu M, Andreazza AC, Keshavjee S, Cypel M. Static lung storage at 10°C maintains mitochondrial health and preserves donor organ function. Sci Transl Med 2021; 13:eabf7601. [PMID: 34524862 DOI: 10.1126/scitranslmed.abf7601] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Aadil Ali
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Aizhou Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Rafaela V P Ribeiro
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Erika L Beroncal
- Departments of Pharmacology & Toxicology and Psychiatry, The Canada Mitochondrial Network, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Cristina Baciu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Marcos Galasso
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Bruno Gomes
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Andrea Mariscal
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Olivia Hough
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Edson Brambate
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Etienne Abdelnour-Berchtold
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Vinicius Michaelsen
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Yu Zhang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Anajara Gazzalle
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Eddy Fan
- Divisions of Respirology and Critical Care Medicine, University Health Network, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Laurent Brochard
- Divisions of Respirology and Critical Care Medicine, University Health Network, University of Toronto, Toronto, ON M5B 1W8, Canada.,Keenan Research Centre, St Michael's Hospital, Unity Health Toronto and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, M5B 1T8, Canada
| | - Jonathan Yeung
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, ON M5G 2C4, Canada
| | - Tom Waddell
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, ON M5G 2C4, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ana C Andreazza
- Departments of Pharmacology & Toxicology and Psychiatry, The Canada Mitochondrial Network, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, ON M5G 2C4, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto Lung Transplant Program, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
38
|
Venkata-Subramani M, Nunley DR, Roman J. Donor factors and risk of primary graft dysfunction and mortality post lung transplantation: A proposed conceptual framework. Clin Transplant 2021; 35:e14480. [PMID: 34516007 DOI: 10.1111/ctr.14480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 08/14/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022]
Abstract
Lung transplantation remains a therapeutic option in end-stage lung disease. However, despite advances in the field, early allograft function can be compromised by the development of primary graft dysfunction (PGD); this being the leading cause of morbidity and mortality immediately following the lung transplant procedure. Several recipient factors have been associated with increased risk of PGD, but less is known about donor factors. Aging, tobacco, and chronic alcohol use are donor factors implicated, but how these factors promote PGD remains unclear. Herein, we discuss the available clinical data that link these donor factors with outcomes after lung transplantation, and how they might render the recipient susceptible to PGD through a two-hit process.
Collapse
Affiliation(s)
- Mrinalini Venkata-Subramani
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Sidney Kimmel College of Medicine, and Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - David R Nunley
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Lung Transplantation Program, The Ohio State University, Columbus, Ohio, USA
| | - Jesse Roman
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Sidney Kimmel College of Medicine, and Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD) is a devastating complication in the acute postoperative lung transplant period, associated with high short-term mortality and chronic rejection. We review its definition, pathophysiology, risk factors, prevention, treatment strategies, and future research directions. RECENT FINDINGS New analyses suggest donation after circulatory death and donation after brain death donors have similar PGD rates, whereas donors >55 years are not associated with increased PGD risk. Recipient pretransplant diastolic dysfunction and overweight or obese recipients with predominant abdominal subcutaneous adipose tissue have increased PGD risk. Newly identified recipient biomarkers and donor and recipient genes increase PGD risk, but their clinical utility remains unclear. Mixed data still exists regarding cold ischemic time and PGD risk, and increased PGD risk with cardiopulmonary bypass remains confounded by transfusions. Portable ex vivo lung perfusion (EVLP) may prevent PGD, but its use is limited to a handful of centers. Although updates to current PGD treatment are lacking, future therapies are promising with targeted therapy and the use of EVLP to pharmacologically recondition donor lungs. SUMMARY There is significant progress in defining PGD and identifying its several risk factors, but effective prevention and treatment strategies are needed.
Collapse
|
40
|
Natalini JG, Diamond JM. Primary Graft Dysfunction. Semin Respir Crit Care Med 2021; 42:368-379. [PMID: 34030200 DOI: 10.1055/s-0041-1728794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Primary graft dysfunction (PGD) is a form of acute lung injury after transplantation characterized by hypoxemia and the development of alveolar infiltrates on chest radiograph that occurs within 72 hours of reperfusion. PGD is among the most common early complications following lung transplantation and significantly contributes to increased short-term morbidity and mortality. In addition, severe PGD has been associated with higher 90-day and 1-year mortality rates compared with absent or less severe PGD and is a significant risk factor for the subsequent development of chronic lung allograft dysfunction. The International Society for Heart and Lung Transplantation released updated consensus guidelines in 2017, defining grade 3 PGD, the most severe form, by the presence of alveolar infiltrates and a ratio of PaO2:FiO2 less than 200. Multiple donor-related, recipient-related, and perioperative risk factors for PGD have been identified, many of which are potentially modifiable. Consistently identified risk factors include donor tobacco and alcohol use; increased recipient body mass index; recipient history of pulmonary hypertension, sarcoidosis, or pulmonary fibrosis; single lung transplantation; and use of cardiopulmonary bypass, among others. Several cellular pathways have been implicated in the pathogenesis of PGD, thus presenting several possible therapeutic targets for preventing and treating PGD. Notably, use of ex vivo lung perfusion (EVLP) has become more widespread and offers a potential platform to safely investigate novel PGD treatments while expanding the lung donor pool. Even in the presence of significantly prolonged ischemic times, EVLP has not been associated with an increased risk for PGD.
Collapse
Affiliation(s)
- Jake G Natalini
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Himebauch AS, Wong W, Wang Y, McGowan FX, Berg RA, Mascio CE, Kilbaugh TJ, Lin KY, Goldfarb SB, Kawut SM, Mercer-Rosa L, Yehya N. Preoperative echocardiographic parameters predict primary graft dysfunction following pediatric lung transplantation. Pediatr Transplant 2021; 25:e13858. [PMID: 33073484 DOI: 10.1111/petr.13858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/13/2020] [Accepted: 09/02/2020] [Indexed: 11/30/2022]
Abstract
The importance of preoperative cardiac function in pediatric lung transplantation is unknown. We hypothesized that worse preoperative right ventricular (RV) systolic and worse left ventricular (LV) diastolic function would be associated with a higher risk of primary graft dysfunction grade 3 (PGD 3) between 48 and 72 hours. We performed a single center, retrospective pilot study of children (<18 years) who had echocardiograms <1 year prior to lung transplantation between 2006 and 2019. Conventional and strain echocardiography parameters were measured, and PGD was graded. Area under the receiver operating characteristic (AUROC) curves and logistic regression were performed. Forty-one patients were included; 14 (34%) developed PGD 3 and were more likely to have pulmonary hypertension (PH) as the indication for transplant (P = .005). PGD 3 patients had worse RV global longitudinal strain (P = .01), RV free wall strain (FWS) (P = .003), RV fractional area change (P = .005), E/e' (P = .01) and lateral e' velocity (P = .004) but not tricuspid annular plane systolic excursion (P = .61). RV FWS (AUROC 0.79, 95% CI 0.62-0.95) and lateral e' velocity (AUROC 0.87, 95% CI 0.68-1.00) best discriminated PGD 3 development and showed the strongest association with PGD 3 (RV FWS OR 3.87 [95% CI 1.59-9.43], P = .003; lateral e' velocity OR 0.10 [95% CI 0.01-0.70], P = .02). These associations remained when separately adjusting for age, weight, primary PH diagnosis, ischemic time, and bypass time. In this pilot study, worse preoperative RV systolic and worse LV diastolic function were associated with PGD 3 and may be modifiable recipient risk factors in pediatric lung transplantation.
Collapse
Affiliation(s)
- Adam S Himebauch
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Wai Wong
- Division of Pulmonary Medicine, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yan Wang
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Francis X McGowan
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Robert A Berg
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher E Mascio
- Division of Cardiothoracic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Todd J Kilbaugh
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Y Lin
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel B Goldfarb
- Division of Pulmonary Medicine, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Laura Mercer-Rosa
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nadir Yehya
- Division of Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Kolaitis NA, Gao Y, Soong A, Greenland JR, Hays SR, Golden J, Leard LE, Shah RJ, Kleinhenz ME, Katz PP, Venado A, Kukreja J, Blanc PD, Singer JP. Primary graft dysfunction attenuates improvements in health-related quality of life after lung transplantation, but not disability or depression. Am J Transplant 2021; 21:815-824. [PMID: 32794295 DOI: 10.1111/ajt.16257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/17/2020] [Accepted: 07/31/2020] [Indexed: 01/25/2023]
Abstract
Disability, depressive symptoms, and impaired health-related quality of life (HRQL) are common among patients with life-threatening respiratory compromise. We sought to determine if primary graft dysfunction (PGD), a syndrome of acute lung injury, attenuates improvements in patient-reported outcomes after transplantation. In a single-center prospective cohort, we assessed disability, depressive symptoms, and HRQL before and at 3- to 6-month intervals after lung transplantation. We estimated the magnitude of change in disability, depressive symptoms, and HRQL with hierarchical segmented linear mixed-effects models. Among 251 lung transplant recipients, 50 developed PGD Grade 3. Regardless of PGD severity, participants had improvements in disability and depressive symptoms, as well as generic-physical, generic-mental, respiratory-specific, and health-utility HRQL, exceeding 1- to 4-fold the minimally clinically important difference across all instruments. Participants with PGD Grade 3 had a lower magnitude of improvement in generic-physical HRQL and health-utility than in all other participants. Among participants with PGD Grade 3, prolonged mechanical ventilation was associated with greater attenuation of improvements. PGD remains a threat to the 2 primary aims of lung transplantation, extending survival and improving HRQL. Attenuation of improvement persists long after hospital discharge. Future studies should assess if interventions can mitigate the impact of PGD on patient-reported outcomes.
Collapse
Affiliation(s)
- Nicholas A Kolaitis
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Ying Gao
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Allison Soong
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - John R Greenland
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Steven R Hays
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Jeffrey Golden
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Lorriana E Leard
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Rupal J Shah
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Mary Ellen Kleinhenz
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Patricia P Katz
- Division of Rheumatology, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Aida Venado
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Jasleen Kukreja
- Division of Thoracic Surgery, Department of Surgery, School of Medicine, University of California, San Francisco, California, USA
| | - Paul D Blanc
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| | - Jonathan P Singer
- Division of Pulmonary and Critical Care, Department of Medicine, School of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
43
|
Calabrese F, Schiavon M, Perissinotto E, Lunardi F, Marulli G, Di Gregorio G, Pezzuto F, Edith Vuljan S, Forin E, Wiegmann B, Jonigk D, Warnecke G, Rea F. Organ Care System Lung resulted in lower apoptosis and iNOS expression in donor lungs. Am J Transplant 2020; 20:3639-3648. [PMID: 32652873 DOI: 10.1111/ajt.16187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion (IR) injury after lung transplantation is still today an important complication in up to 25% of patients. The Organ Care System (OCS) Lung, an advanced normothermic ex vivo lung perfusion system, was found to be effective in reducing primary graft dysfunction compared to standard organ care (SOC) but studies on tissue/molecular pathways that could explain these more effective clinical results are lacking. This observational longitudinal study aimed to investigate IR injury in 68 tissue specimens collected before and after reperfusion from 17 OCS and 17 SOC preserved donor lungs. Several tissue analyses including apoptosis evaluation and inducible nitric oxide synthase (iNOS) expression (by immunohistochemistry and real-time reverse transcriptase-polymerase chain reaction) were performed. Lower iNOS expression and apoptotic index were distinctive of OCS preserved tissues at pre- and post-reperfusion times, independently from potential confounding factors. Moreover, OCS recipients had lower acute cellular rejection at the first 6-month follow-up. In conclusion, IR injury, in terms of apoptosis and iNOS expression, was less frequent in OCS- than in SOC-preserved lungs, which could eventually explain a better clinical outcome. Further studies are needed to validate our data and determine the role of iNOS expression as a predictive biomarker of the complex IR injury mechanism.
Collapse
Affiliation(s)
- Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Egle Perissinotto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Marulli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | | | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Edoardo Forin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Bettina Wiegmann
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Center of Lung Research, Hannover, Germany
| | - Danny Jonigk
- German Center of Lung Research, Hannover, Germany.,Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Center of Lung Research, Hannover, Germany
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
44
|
Regadenoson to the rescue: On the road toward prevention of primary graft dysfunction after lung transplantation. J Heart Lung Transplant 2020; 39:571-572. [PMID: 32303398 DOI: 10.1016/j.healun.2020.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022] Open
|
45
|
Sacreas A, Verleden SE. High-resolution human leukocyte antigen typing and early post-transplant outcomes: more than meets the eye. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:46. [PMID: 32154808 DOI: 10.21037/atm.2020.01.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Annelore Sacreas
- Leuven Lung Transplant Group, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Leuven Lung Transplant Group, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, Leuven, Belgium
| |
Collapse
|
46
|
Kawashima M, Juvet SC. The role of innate immunity in the long-term outcome of lung transplantation. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:412. [PMID: 32355856 PMCID: PMC7186608 DOI: 10.21037/atm.2020.03.20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Long-term survival after lung transplantation remains suboptimal due to chronic lung allograft dysfunction (CLAD), a progressive scarring process affecting the graft. Although anti-donor alloimmunity is central to the pathogenesis of CLAD, its underlying mechanisms are not fully elucidated and it is neither preventable nor treatable using currently available immunosuppression. Recent evidence has shown that innate immune stimuli are fundamental to the development of CLAD. Here, we examine long-standing assumptions and new concepts linking innate immune activation to late lung allograft fibrosis.
Collapse
Affiliation(s)
- Mitsuaki Kawashima
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Stephen C Juvet
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Burki S, Noda K, Philips BJ, Velayutham M, Shiva S, Sanchez PG, Kumar A, D'Cunha J. Impact of triptolide during ex vivo lung perfusion on grafts after transplantation in a rat model. J Thorac Cardiovasc Surg 2020; 161:S0022-5223(20)30191-4. [PMID: 32169373 DOI: 10.1016/j.jtcvs.2019.12.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/30/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Ex vivo lung perfusion creates a proinflammatory environment leading to deterioration in graft quality that may contribute to post-transplant graft dysfunction. Triptolide has been shown to have a therapeutic potential in various disease states because of its anti-inflammatory properties. On this basis, we investigated the impact of triptolide on graft preservation during ex vivo lung perfusion and associated post-transplant outcomes in a rat transplant model. METHODS We performed rat normothermic ex vivo lung perfusion with acellular Steen solution containing 100 nM triptolide for 4 hours and compared the data with untreated lungs. Orthotopic single lung transplantation after ex vivo lung perfusion was performed. RESULTS Physiologic and functional parameters of lung grafts on ex vivo lung perfusion with triptolide were better than those without treatment. Graft glucose consumption was significantly attenuated on ex vivo lung perfusion with triptolide via inhibition of hypoxia signaling resulting in improved mitochondrial function and reduced oxidative stress. Also, intragraft inflammation was markedly lower in triptolide-treated lungs because of inhibition of nuclear factor-κB signaling. Furthermore, post-transplant graft function and inflammatory events were significantly improved in the triptolide group compared with the untreated group. CONCLUSIONS Treatment of lung grafts with triptolide during ex vivo lung perfusion may serve to enhance graft preservation and improve graft protection resulting in better post-transplant outcomes.
Collapse
Affiliation(s)
- Sarah Burki
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Brian J Philips
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Murugesan Velayutham
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pa; Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Sruti Shiva
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Pablo G Sanchez
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Ajay Kumar
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Pheonix, Ariz.
| |
Collapse
|
48
|
Effect of left ventricular diastolic dysfunction on development of primary graft dysfunction after lung transplant. Curr Opin Anaesthesiol 2019; 33:10-16. [PMID: 31789901 DOI: 10.1097/aco.0000000000000811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD) is one of the most common complications after lung transplant and is associated with significant early and late morbidity and mortality. The cause of primary graft dysfunction is often multifactorial involving patient, donor, and operational factors. Diastolic dysfunction is increasingly recognized as an important risk factor for development of PGD after lung transplant and here we examine recent evidence on the topic. RECENT FINDINGS Patients with end-stage lung disease are more likely to suffer from cardiovascular disease including diastolic dysfunction. PGD as result of ischemia-reperfusion injury after lung transplant is exacerbated by increased left atrial pressure and pulmonary venous congestion impacted by diastolic dysfunction. Recent studies on relationship between diastolic dysfunction and PGD after lung transplant show that patients with diastolic dysfunction are more likely to develop PGD with worse survival outcome and complicated hospital course. SUMMARY Patients with diastolic dysfunction is more likely to suffer from PGD after lung transplant. From the lung transplant candidate selection to perioperative and posttransplant care, thorough evaluation and documentation diastolic dysfunction to guide patient care are imperative.
Collapse
|
49
|
The Comparable Efficacy of Lung Donation After Circulatory Death and Brain Death: A Systematic Review and Meta-analysis. Transplantation 2019; 103:2624-2633. [DOI: 10.1097/tp.0000000000002888] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
50
|
Scaravilli V, Morlacchi LC, Merrino A, Piacentino E, Marasco D, Zanella A, Nosotti M, Rosso L, Polli F, Blasi F, Pesenti A, Grasselli G. Intraoperative extracorporeal membrane oxygenation for lung transplantation in cystic fibrosis patients: Predictors and impact on outcome. J Cyst Fibros 2019; 19:659-665. [PMID: 31676344 DOI: 10.1016/j.jcf.2019.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Predictors and outcomes of intraoperative extracorporeal membrane oxygenation (ECMO) during lung transplantation (LUTX) for cystic fibrosis (CF) are unknown. METHODS We retrospectively collected the clinical data at enlistment of the CF patients who underwent double LUTX from January 2013 to December 2018 at an Italian tertiary referral center. We compared blood transfusions, incidence of primary graft dysfunction (PGD), duration of mechanical ventilation, intensive care unit (ICU) length of stay (LOS), hospital LOS and survival of ECMO and non-ECMO patients. Chi-square, Kruskal-Wallis, and log-rank tests were used. RESULTS Twenty-eight (40%) of the 70 included patients needed intraoperative central veno-arterial ECMO with postoperative veno-venous prolongation in 6 subjects. Lower right ventricle ejection fraction (p = 0.013, OR 0.92(0.86-0.98)), higher oxygen requirement (p = 0.026, OR 1.39(1.01-1.90)), lower body surface area (p = 0.044, OR 0.05(0.00-1.03)), and CF-related diabetes (p = 0.044, OR 2.81(1.03-7.66)) were associated with intraoperative ECMO. Compared to non-ECMO patients, ECMO patients needed almost fivefold intraoperative transfusion (2227 mL vs. 570 mL, p<0.001) and had PGD grade > 0 at 72 h more frequently (16/57% vs. 12/28%, p = 0.017, OR 3.33(1.22-9.09)). Mechanical ventilation, ICU LOS and hospital LOS were significantly longer in ECMO patients. Survival at follow-up (651(326-1277) days) of ECMO and non-ECMO patients was 78% vs. 83%, respectively (OR 0.73 (0.21-2.46), p = 0.616, log-rank test p = 0.498). CONCLUSION Pre-operative risk assessment and clinical planning should be done according to the predictors above. While undeniably useful as a life-saving procedure, ECMO during LUTX for CF is associated with worsened short-term outcomes. ECMO should be implemented weighing its risk and benefits.
Collapse
Affiliation(s)
- Vittorio Scaravilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, (MI) Italy.
| | - Letizia Corinna Morlacchi
- Department of Internal Medicine, Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Alessandra Merrino
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Edoardo Piacentino
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Davide Marasco
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Alberto Zanella
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, (MI) Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Mario Nosotti
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy; Thoracic Syrgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Lorenzo Rosso
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy; Thoracic Syrgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Federico Polli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, (MI) Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy; Department of Internal Medicine, Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Antonio Pesenti
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, (MI) Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, (MI) Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| |
Collapse
|