1
|
Rudzik KN, Lyster H. Management of pharmacotherapy in lung transplant candidates. Curr Opin Pulm Med 2025; 31:387-396. [PMID: 40265512 DOI: 10.1097/mcp.0000000000001172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW Lung transplantation is a common treatment for end-stage lung disease (ESLD). Patients present to lung transplantation evaluation on various medications that could impact their candidacy and posttransplant course. In this review, we will discuss pretransplant optimization of pharmacotherapy to minimize complications while waiting for transplant and increase posttransplant success. We will also discuss important considerations for posttransplant immunosuppression, antimicrobial prophylaxis, and complex drug interactions. RECENT FINDINGS Prior to lung transplantation, several medications should be optimized to promote posttransplant success including minimization of corticosteroids, opioids, and benzodiazepines. Lung transplantation candidates should be up to date on vaccinations. Most medications for ESLD are well tolerated to continue up until the point of transplant including antifibrotics, CFTR modulators, and pulmonary vasodilators. Mammalian target of rapamycin inhibitors and other immunosuppressants may need to be stopped or minimized before lung transplantation to minimize posttransplant infection and would healing complications. Medications that increase risk of posttransplant bleeding, thrombosis, or aspiration should be stopped prior to listing. SUMMARY In this article, we discuss management of pharmacotherapy for lung transplantation candidates to minimize posttransplant complications. Changes in medications for ESLD should be done cautiously to prevent worsening of native disease while waiting for lung transplantation.
Collapse
Affiliation(s)
| | - Haifa Lyster
- Department of Pharmacy, Royal Brompton and Harefield Hospitals, part of Guy's and St Thomas' NHS Foundation Trust
- King's College London, London, UK
| |
Collapse
|
2
|
Ronen L, Keshavjee S, Sage AT. Advancing lung transplantation through machine learning and artificial intelligence. Curr Opin Pulm Med 2025; 31:381-386. [PMID: 40152900 DOI: 10.1097/mcp.0000000000001168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
PURPOSE OF REVIEW To explore the current applications of artificial intelligence and machine learning in lung transplantation, including outcome prediction, drug dosing, and the potential future uses and risks as the technology continues to evolve. RECENT FINDINGS While the use of artificial intelligence (AI) and machine learning (ML) in lung transplantation is relatively new, several groups have developed models to predict short-term outcomes, such as primary graft dysfunction and time-to-extubation, as well as long-term outcomes related to survival and chronic lung allograft dysfunction. Additionally, drug dosing models for Tacrolimus levels have been designed, demonstrating proof of concept for modelling treatment as a time-series problem. SUMMARY The integration of ML models with clinical decision-making has shown promise in improving post-transplant survival and optimizing donor lung utilization. As technology advances, the field will continue to evolve, with enhanced datasets supporting more sophisticated ML models, particularly through real-time monitoring of biological, biochemical, and physiological data.
Collapse
Affiliation(s)
- Lielle Ronen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network
- Institute of Medical Science
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network
- Institute of Medical Science
- Department of Surgery, Temerty Faculty of Medicine
| | - Andrew T Sage
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network
- Institute of Medical Science
- Department of Surgery, Temerty Faculty of Medicine
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Chan EG, Deitz RL, Ryan JP, Suzuki Y, Hage CA, Furukawa M, Noda K, Subramaniam K, Sanchez PG. Bloodless lung transplantation: Comparison between 2 central venoarterial extracorporeal membrane oxygenation anticoagulation strategies and their impact on lung transplant outcomes. J Thorac Cardiovasc Surg 2025; 169:1620-1628. [PMID: 39393627 DOI: 10.1016/j.jtcvs.2024.09.055] [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: 04/25/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE To report differences between 2 anticoagulation protocols during venoarterial extracorporeal membrane oxygenation (VA-ECMO) intraoperative support and their effects on outcomes after lung transplantation. METHODS We performed a retrospective analysis of patients undergoing double-lung transplantation with intraoperative VA-ECMO from January 1, 2016, to December 30, 2023. Two distinct anticoagulation protocols were in place during this period. One included targeted activated clotting time >180 seconds at all times with protamine reversal after decannulation. The second included 75 units per kilogram of heparin at the time of cannulation with no redosing plus a tranexamic acid infusion after ECMO initiation. RESULTS A total of 116 patients (46 low heparin, 70 standard) were included in the analysis. Cannulation strategies and ECMO circuit were equivalent between the groups. The low-dose heparin protocol group had a shorter surgical time (7.28 hours vs 8.53 hours, P < .001) and required significantly less intraoperative packed red blood cells (median 0 vs 4.37 units, P < .001), fresh-frozen plasma (median 0 vs 2 units, P < .001), platelets (median 0 vs 1 units, P < .001), cryoprecipitate (median 0 vs 0 units, P < .001), and total blood products (median 0 vs 9 units, P < .001) compared with the standard group. There were no differences in rates of deep vein thrombosis (P = .13), airway dehiscence (P > .99), pneumonia (P = .38), or acute kidney injury requiring renal-replacement therapy (P = .59). There was no difference in rates of severe grade 3 primary graft dysfunction at 72 hours after transplant (P = .42). CONCLUSIONS Our low-dose heparin VA-ECMO protocol for intraoperative support during lung transplantation led to a significant reduction of blood product use. Although this did not translate to a reduced rates of grade 3 primary graft dysfunction, the low-dose heparin protocol was associated with similar postoperative outcomes.
Collapse
Affiliation(s)
- Ernest G Chan
- Section of Thoracic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Ill
| | - Rachel L Deitz
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - John P Ryan
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Yota Suzuki
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Chadi A Hage
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Masashi Furukawa
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Kathirvel Subramaniam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Pablo G Sanchez
- Section of Thoracic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Ill.
| |
Collapse
|
4
|
Braithwaite SA, Blankman P, van der Kaaij NP. New Techniques for the Optimization of Donor Lungs/Hearts. Anesthesiol Clin 2025; 43:243-265. [PMID: 40348542 DOI: 10.1016/j.anclin.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Optimization of donor heart and lung allografts is vital for the post-transplant outcome of the recipient, in terms of survival and quality of life. This requires a holistic approach to the transplant process ensuring optimal donor management, organ procurement and allograft preservation. Advances are being made in static cold storge techniques and in ex vivo organ perfusion. Normothermic perfusion techniques enable functional and/ or biochemical assessment of allografts. Future applications of ex vivo perfusion will enable a platform facilitating the advanced diagnostic assessment of allografts, the administration of advanced therapy to attenuate organ injury and to immunomodulate allografts.
Collapse
Affiliation(s)
- Sue A Braithwaite
- Department of Anesthesiology, University Medical Center Utrecht, Q04.2.317, Postbus 85500, Utrecht, 3508 GA, The Netherlands.
| | - Paul Blankman
- Department of Anesthesiology, University Medical Center Utrecht, Q04.2.317, Postbus 85500, Utrecht, 3508 GA, The Netherlands
| | - Niels P van der Kaaij
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, E03.511, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
5
|
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; 25:1339-1349. [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] [MESH Headings] [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; 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
|
6
|
Wu J, Gao P, Yang C, Zhuang F, Luo Y, Wen F, Zhang P, Wang L, Xie H, Dai C, Zhao D, Li C, Deng H, Deng Z, Chen C. Targeting mitochondrial complex I of CD177 + neutrophils alleviates lung ischemia-reperfusion injury. Cell Rep Med 2025; 6:102140. [PMID: 40398393 DOI: 10.1016/j.xcrm.2025.102140] [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: 06/20/2024] [Revised: 01/01/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025]
Abstract
Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality following lung transplantation, with neutrophils playing a central role in its inflammatory pathology. Here, we employ single-cell RNA sequencing and spatial transcriptomics to investigate neutrophil subtypes in the lung ischemia-reperfusion injury (IRI) model. We identify CD177+ neutrophils as an activated subpopulation that significantly contributes to lung injury and serves as an early biomarker for predicting severe PGD in human lung transplant recipients (area under the curve [AUC] = 0.871). CD177+ neutrophils exhibit elevated oxidative phosphorylation and increased mitochondrial complex I activity, driving inflammation and the formation of neutrophil extracellular traps. Targeting mitochondrial function with the complex I inhibitor IACS-010759 reduces CD177+ neutrophil activation and alleviates lung injury in both mouse IRI and rat left lung transplant models. These findings provide a comprehensive landscape of CD177+ neutrophil-driven inflammation in lung IRI and highlight its potential value for future early diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenlu Yang
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Yunzhe Luo
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | | | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chongwu Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | | | | | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
7
|
Hillebrand C, Benazzo A. [Immunological Aspects after Lung Transplantation]. Zentralbl Chir 2025. [PMID: 40359989 DOI: 10.1055/a-2590-9933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Since the 1980 s, lung transplantation has evolved into an established therapeutic procedure, due to advancements in surgical techniques and the introduction of immunosuppressants such as cyclosporine. Despite improved short-term outcomes, the long-term prognosis remains limited, primarily due to immunological complications. With a median survival of approximately six years, the lung is the most immunogenic solid organ, owing to its constant exposure to environmental antigens and its extensive vascular endothelial surface. After lung transplantation, various forms of alloreactivity, including T cell-mediated acute and chronic rejection, play a central role. Additionally, humoral immune responses, characterised by the production of donor-specific and non-HLA antibodies, contribute significantly to graft injury. Recurrent tissue damage, such as ischemia reperfusion injury, leads to the exposure of cryptic antigens, promotes autoreactive processes, and facilitates the formation of tertiary lymphoid organs. These mechanisms sustain persistent inflammation, ultimately resulting in chronic graft dysfunction. Rejection reactions remain a major challenge. Acute forms, such as cellular and humoral rejection, require rapid and targeted therapies to prevent irreversible damage. Chronic rejection, particularly chronic lung allograft dysfunction (CLAD), progressively impairs lung function. In the main phenotypes of CLAD, bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS), are crucial for prognosis and treatment. Nevertheless, therapeutic options remain limited, and retransplantation is often the last resort. Immunosuppressive therapy forms the cornerstone of rejection prevention, and typically employs a triple combination of calcineurin inhibitors, antiproliferative agents, and corticosteroids. Induction therapy frequently involves monoclonal or polyclonal antibodies. Modern strategies aim to effectively suppress immune responses while minimising severe side effects, such as infections, malignancies, and nephrotoxicity. Future research will focus on personalised immunosuppressive strategies, optimised diagnostics, and innovative therapies to improve the long-term prognosis of lung transplant recipients.
Collapse
Affiliation(s)
| | - Alberto Benazzo
- Thoraxchirurgie, Medizinische Universität Wien, Wien, Österreich
| |
Collapse
|
8
|
Chuachao P, Devaquet J, Sage E, Vallée A, Guen ML, Fischler M, Fessler J. Prediction of Postoperative Lung Graft Dysfunction During the Procedure: A Single-Center Cohort Study of Cystic Fibrosis Patients. J Cardiothorac Vasc Anesth 2025:S1053-0770(25)00351-9. [PMID: 40414788 DOI: 10.1053/j.jvca.2025.04.033] [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: 02/14/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVES To predict severe primary graft dysfunction (PGD3) after double-lung transplantation in cystic fibrosis (CF) patients using intraoperative data. DESIGN A retrospective single-center cohort study. SETTING University Hospital, France. PARTICIPANTS CF patients who underwent double-lung transplantation between 2012 and 2019. Patients younger than age 18 and those with multiorgan transplants, retransplantation, or intraoperative cardiopulmonary bypass were excluded. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Sixty-nine variables were recorded in real-time across the nine time-points. PGD3 occurred in 24 patients (15.5%). PGD3 WAS ASSESSED ON POSTOPERATIVE DAY 3: A logistic regression model to predict PGD3 was developed using data collected at nine predefined time-points during surgery, from start (recipient and donor variables) to finish. The model's area under the curve improved progressively during surgery, rising from 0.764 to 0.892. The optimal model incorporated five variables: three associated with reduced PGD3 risk (preoperative pulmonary hypertension, donor body mass index, and PaO₂/FiO₂ ratio at surgery's end) and two were linked to increased risk (lactate level at second pulmonary artery clamping and extracorporeal membrane oxygenation [ECMO] use at surgery's end). The risk of PGD3 increased by a factor of 11.48 (95% CI 4.48-29.39; p < 0.001) when ECMO was required at the end of surgery and by 1.29 (95% CI: 1.02-1.63; p = 0.035) for each 1 mEq/L rise in lactate concentration at time-point 7 (second pulmonary artery clamping). CONCLUSIONS This predictive model underscores the adverse impact of sustained ECMO placed at the end of surgery and elevated intraoperative lactate levels on PGD3 risk.
Collapse
Affiliation(s)
- Pimnara Chuachao
- Department of Epidemiology, Data, Biostatistics, Delegation of Clinical Research and Innovation, Hôpital Foch, Suresnes, France
| | - Jérome Devaquet
- Department of Intensive Care Medicine, Hôpital Foch, Suresnes, France
| | - Edouard Sage
- Department of Thoracic Surgery, Hôpital Foch, Suresnes, France; Université Versailles-Saint-Quentin-en-Yvelines, Versailles, France
| | - Alexandre Vallée
- Department of Epidemiology and Public Health, Hôpital Foch, Suresnes, France
| | - Morgan Le Guen
- Université Versailles-Saint-Quentin-en-Yvelines, Versailles, France; Department of Anesthesiology, Hôpital Foch, Suresnes, France
| | - Marc Fischler
- Department of Anesthesiology, Hôpital Foch, Suresnes, France
| | - Julien Fessler
- Department of Anesthesiology, Hôpital Foch, Suresnes, France.
| |
Collapse
|
9
|
Schäfer M, Selzman CH, DiChiacchio L, Heiler JC, Ramakrishna S, Tumarkin E, Hill B, Kagawa H, Contreras N, Raman S, Frye L, Cahill B, Morrell M, Goodwin ML. Reduced left atrial compliance exacerbates primary graft dysfunction after lung transplantation. J Thorac Cardiovasc Surg 2025:S0022-5223(25)00309-5. [PMID: 40328425 DOI: 10.1016/j.jtcvs.2025.04.020] [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: 02/11/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVE Primary graft dysfunction (PGD) after lung transplantation (LTx) heralds significantly worse short- and long-term outcomes. The preoperative presence of recipient left ventricular diastolic dysfunction elevates postcapillary hydrostatic pressures and increases the risk for PGD. In this study, we investigated the role of the left atrial strain (LAS), a recently established sensitive marker of left atrial compliance, as a predictor of PGD. METHODS Preoperative echocardiography of all patients who underwent bilateral LTx at a single center from 2014 to 2024 was analyzed for global myocardial deformation, including the standard phases of LAS reservoir, conduit, and pump as well as left ventricular global longitudinal strain. The presence of PGD grade 3 was defined as P:F <200 at 48 or 72 hours after the operation. Right heart catheterization, standard echocardiographic, and strain indices were subjected to univariable and multivariable analysis to predict PGD. RESULTS In total, 132 patients were analyzed, from whom 35 (26.5%) developed PGD. There were no differences in traditional echocardiographic left ventricular diastolic dysfunction biomarkers, including Doppler and tissue Doppler indices, between the PGD (+) and PGD (-) groups. Preoperative right heart catheterization revealed increased mean pulmonary arterial pressure (36 vs 26 mm Hg, P = .003) and median pulmonary vascular resistance (7.8 vs 4.6, P = .001) in the PGD group. Reservoir LAS was reduced in PGD (22.7 ± 7.7 vs 31.5 ± 10.7%, P < .001), followed by reduced conduit LAS (-11.4 ± 6.6 vs -16.0 ± 8.2%, P = .002) and reduced LV GLS (-13.9 ± 3.6 vs -15.8 ± 3.7%, P = .014). In final multivariable model, conduit LAS was independently associated with a greater risk of PGD (odds ratio, 0.88; 95% confidence interval, 0.81-0.95; P = .002) along with greater pulmonary vascular resistance index (odds ratio, 1.13; 95% confidence interval, 1.05-1.25; P = .003). The final model yielded a c-statistic of 0.82, specificity of 93.8%, sensitivity of 40.0%, positive predictive value of 80.8%, and negative predictive value of 70.6%. CONCLUSIONS Patients with decreased preoperative left atrial compliance assessed by LAS who undergo LTx have a greater risk of developing PGD in the setting of normal LV systolic and diastolic function. Given the increasing use of strain indices, LAS should be considered a risk factor for PGD in prospective studies.
Collapse
Affiliation(s)
- Michal Schäfer
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah.
| | - Craig H Selzman
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| | - Laura DiChiacchio
- Division of Cardiothoracic Surgery, Cedars-Sinai, Los Angeles, Calif
| | - Joseph C Heiler
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| | | | - Ethan Tumarkin
- Division of Cardiology, University of Utah, Salt Lake City, Utah
| | - Bryce Hill
- Department of Anesthesiology, University of Utah, Salt Lake City, Utah
| | - Hiroshi Kagawa
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| | - Nicolas Contreras
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| | - Sanjeev Raman
- Division of Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Laura Frye
- Division of Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Barbara Cahill
- Division of Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Matt Morrell
- Division of Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Matthew L Goodwin
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| |
Collapse
|
10
|
Liu X, Chen W, Du W, Li P, Wang X. Application of artificial intelligence and machine learning in lung transplantation: a comprehensive review. Front Digit Health 2025; 7:1583490. [PMID: 40376618 PMCID: PMC12078212 DOI: 10.3389/fdgth.2025.1583490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Lung transplantation (LTx) is an effective method for treating end-stage lung disease. The management of lung transplant recipients is a complex, multi-stage process that involves preoperative, intraoperative, and postoperative phases, integrating multidimensional data such as demographics, clinical data, pathology, imaging, and omics. Artificial intelligence (AI) and machine learning (ML) excel in handling such complex data and contribute to preoperative assessment and postoperative management of LTx, including the optimization of organ allocation, assessment of donor suitability, prediction of patient and graft survival, evaluation of quality of life, and early identification of complications, thereby enhancing the personalization of clinical decision-making. However, these technologies face numerous challenges in real-world clinical applications, such as the quality and reliability of datasets, model interpretability, physicians' trust in the technology, and legal and ethical issues. These problems require further research and resolution so that AI and ML can more effectively enhance the success rate of LTx and improve patients' quality of life.
Collapse
Affiliation(s)
- Xiting Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Wenqian Chen
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Wenwen Du
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Pengmei Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
- Department of Pharmacy Administration, Clinical Pharmacy School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
11
|
Yin V, Rodman JCS, Atay SM, Wightman SC, Rosenberg GM, Udelsman BV, Ganesh S, Chung P, Kim AW, Harano T. Outcomes of single-lung retransplantation after double-lung transplantation. J Thorac Cardiovasc Surg 2025; 169:1398-1406.e2. [PMID: 39357566 DOI: 10.1016/j.jtcvs.2024.09.039] [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: 04/15/2024] [Revised: 08/21/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE To compare outcomes of single-lung retransplantation (SLRTx) and double-lung retransplantation (DLRTx) after an initial double-lung transplantation. METHODS The Organ Procurement and Transplantation Network/United Network for Organ Sharing database between May 2005 and December 2022 was retrospectively analyzed. Multiorgan transplantations, repeated retransplantations, and lung retransplantations when the status of the initial transplantation was unknown were excluded. RESULTS A total of 891 patients were included in the analysis, included 698 (78.3%) with DLRTx and 193 (21.7%) with SLRTx. The mean lung allocation score was higher in the DLRTx group (59.6 ± 20.7 vs 55.1 ± 19.3; P = .007). The use of extracorporeal membrane oxygenation (ECMO) bridge to lung transplantation was similar in the 2 groups (P = .125), as was waitlist time (P = .610). The need for mechanical ventilation (54.6% vs 35.8%; P = .005) and ECMO (17.9% vs 9.0%; P = .069) at 72 hours post-transplantation was greater in the DLRTx group. However, median post-transplantation hospital stay (21.5 [interquartile range (IQR), 12-35] days versus 20 [IQR, 12-35] days; P = .119) and in-hospital mortality (10.9% [n = 76/698] vs 12.4% [n = 24/193]; P = .547) were comparable in the 2 groups. Long-term survival was significantly better in the DLRTx group (P < .001, log-rank test). In the propensity score-weighted multivariable model, the DLRTx group had 28% lower risk of mortality at any point during follow-up compared to the SLRTx group (hazard ratio, 0.72; 95% confidence interval, 0.57-0.91; P = .006). CONCLUSIONS The less invasiveness of single-lung transplantation in the retransplantation setting has minimal short-term benefit and is associated with significantly worse long-term survival. Double-lung retransplantation should remain the standard for lung retransplantation after initial double-lung transplantation.
Collapse
Affiliation(s)
- Victoria Yin
- Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - John C S Rodman
- Biostatistics, Epidemiology, and Research Design, Southern California Clinical and Translational Science Institute, University of Southern California, Los Angeles, Calif
| | - Scott M Atay
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Sean C Wightman
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Graeme M Rosenberg
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Brooks V Udelsman
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Sivagini Ganesh
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Peter Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Anthony W Kim
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Takashi Harano
- Division of Thoracic Surgery, Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, Calif.
| |
Collapse
|
12
|
Xu X, Lou Z, Li J, Liang F, Yu Y, Wu M. Inhibition of Hsp90 Alleviates Necroptosis and Inflammation in Lung Epithelial Cells During Pulmonary Ischemia-Reperfusion Injury. Clin Exp Pharmacol Physiol 2025; 52:e70037. [PMID: 40169254 DOI: 10.1111/1440-1681.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/12/2025] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Lung ischemia-reperfusion injury (LIRI) is a critical pathological process associated with various clinical conditions, characterised by excessive inflammatory responses and cell death, which can lead to severe respiratory dysfunction and even mortality. However, no effective therapeutic strategy is currently available. This study investigates the protective effects and underlying mechanisms of the Hsp90 inhibitor 17-dimethylaminoethylamino (17-DMAG) in LIRI. An in vivo mouse model of LIRI was established by transiently occluding the left pulmonary hilum with a microvascular clamp, followed by reperfusion. In vitro, necroptosis was induced in BEAS-2B cells using TSZ (TNF-α, Smac mimetic and z-VAD-FMK). Our results demonstrate that 17-DMAG significantly attenuates lung injury, inflammation and epithelial cell necroptosis in mice. Additionally, 17-DMAG mitigates TSZ-induced cell death and inflammatory responses in BEAS-2B cells. Mechanistically, 17-DMAG inhibits the phosphorylation of RIPK1, RIPK3 and MLKL-key necroptotic regulators and client proteins of Hsp90-thereby suppressing necroptosis and reducing the associated inflammatory response. In conclusion, 17-DMAG alleviates LIRI by inhibiting necroptosis and its consequent acute inflammatory cascade. These findings suggest that 17-DMAG may serve as a promising therapeutic candidate for LIRI treatment.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Lung Surgery, Zhejiang Cancer Hospital, Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Zhiling Lou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Jinsheng Li
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Fuxiang Liang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Yifan Yu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Ming Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| |
Collapse
|
13
|
Park BS, Kim DJ, Lee CY, Park BJ, Kim HE, Yang YH, Park MS, Kim SY, La Woo A, Kim EY, Lee JG. Outcomes of Donor/Recipient Size-mismatched Lung Transplantation. Transplant Proc 2025:S0041-1345(25)00228-3. [PMID: 40312209 DOI: 10.1016/j.transproceed.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/05/2025] [Accepted: 04/16/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Donor/recipient (D/R) size matching is crucial to achieve proper organ allocation and outcome for lung transplantation (LT). However, studies in this regard have not shown consistent results. We analyzed the results of size-mismatched LT focusing on primary graft dysfunction (PGD) and survival. METHODS A total of 446 patients underwent LT between January 2010 and December 2022. After exclusion, the patients were divided into 3 groups according to the donor/recipient size; D/R ratio >120% was grouped as Over (n = 87), 120%≥D/R ratio≥80% was grouped as Normal (n = 271), 80%>D/R was grouped as Under (n = 19). Early and long-term outcomes were analyzed. RESULTS Recipient height, weight, and proportion of male were the highest in the Under group, followed by the Normal and Over groups (P < 0.001). The ratio of extracorporeal membrane oxygenation weaning in the operating room was highest in the Under group, followed by the Normal and Over groups (P : 0.04). The proportions of PGD grade 3 within 48 h and 72 h were highest in the Over group, followed by the Normal and Under groups (P : 0.007 and 0.016, respectively). There was no statistical difference in the pulmonary function test results between the groups at 12 months postoperative follow-up. The 5-year survival rate did not differ among the groups (60.9% vs 56.8% vs 54.7%, Under vs Normal vs Over, P : 0.833) CONCLUSIONS: Although oversized D/R-matched LT demonstrated late recovery during the early postoperative period, their long-term results were non-inferior in terms of the D/R size ratio.
Collapse
Affiliation(s)
- Bong Suk Park
- Department of Thoracic and Cardiovascular Surgery, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Dae Joon Kim
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chang Young Lee
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Jo Park
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ha Eun Kim
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ho Yang
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moo Suk Park
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Song Yee Kim
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - A La Woo
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Kim
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Geraci TC, Chan JCY, 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
| | - Justin C Y Chan
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY
| | - Anna Niroomand
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY
| | - Stephanie H Chang
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY.
| |
Collapse
|
15
|
Park HJ, Choi SM, Na KJ, Park S, Lee HJ, Kim YT, Lim WH, Yoon SH, Lee JH, Park J. Prognostic impact of low muscle mass on clinical outcomes in patients who undergo lung transplant. J Thorac Cardiovasc Surg 2025:S0022-5223(25)00282-X. [PMID: 40187556 DOI: 10.1016/j.jtcvs.2025.03.030] [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: 12/27/2024] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Low muscle mass (LMM) is recognized as a poor prognostic factor in various chronic lung diseases. However, its prognostic impact on recipients of lung transplants remains inconclusive. METHODS We retrospectively analyzed patients who underwent lung transplantation at a tertiary referral center in South Korea. Pretransplant skeletal muscle mass was quantified at the L1 vertebral level by computed tomography scans of the chest using a commercially available body composition analysis software. Patients were classified into LMM and non-LMM group using a threshold for LMM that had been previously validated in the South Korean population. We then evaluated the prognostic impact of preoperative LMM on clinical outcomes after lung transplantation. RESULTS A total of 107 patients were included in this analysis, of whom 44 (41.1%) were classified into the LMM group. The median follow-up duration was 958 days posttransplantation. A preoperative LMM was identified as an independent factor associated with a greater risk of overall mortality (adjusted hazard ratio, 2.15; 95% confidence interval, 1.07-4.34). In addition, patients with LMM had a greater risk of developing primary graft dysfunction (adjusted odds ratio, 3.56; 95% confidence interval, 1.25-10.18). At the 1-year follow-up, 37.5% of the patients with baseline LMM had recovered and were reclassified into the non-LMM group, and this improvement was found to mitigate the negative impact of preoperative LMM. CONCLUSIONS Pretransplant LMM was significantly associated with poor clinical outcomes in recipients of lung transplants. These findings highlight the importance of maintaining adequate muscle mass during the waiting period for lung transplantation.
Collapse
Affiliation(s)
- Hyun-Jun Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sun Mi Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kwon Joong Na
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Republic of Korea; Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Samina Park
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyun Joo Lee
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Republic of Korea; Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Woo Hyeon Lim
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soon Ho Yoon
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jong Hyuk Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Jimyung Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Ware LB, Koyama T, Shaver CM, Swain S, Nguyen J, Salehi A, Dhillon G, Wickersham N, Maheshwari J, Singer JP, Weigt SS, Kukreja J, Matthay MA. A randomized trial of open lung protective ventilation compared to conventional mechanical ventilation in deceased organ donors. J Heart Lung Transplant 2025:S1053-2498(25)01865-0. [PMID: 40187505 DOI: 10.1016/j.healun.2025.03.027] [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: 01/24/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND We conducted a randomized trial of open lung protective ventilation (OLPV) compared to conventional ventilation (CV) in deceased donors. The primary outcome was lung utilization for transplantation. METHODS Eligible donors were ≥13 years with PaO2/FiO2 between 150 and 400 mmHg. Donors were randomized to volume control with OLPV [tidal volume (TV) 8 ml/kg, PEEP 10 cmH2O, protocolized recruitment maneuvers (RM)] or CV [TV 10 ml/kg, PEEP 5 cm H2O, RM only after vent disconnect] for duration of donor management. Lungs were evaluated for transplantation on standardized ventilator settings in both arms [TV 10 ml/kg, PEEP 5 cm H2O, FiO2 1.0]. RESULTS One hundred and fifty three donors were randomized (74 to OLPV, 79 to CV) and included in the final analysis. Median duration of treatment was 50 hours and did not differ by arm. Donor lung utilization was 23% in the OLPV arm and 22% in the CV arm, p = 0.85. Change in PaO2/FiO2 from randomization to procurement did not differ by treatment; median increase (quartiles) in OLPV versus CV was 68 mmHg (18, 127) vs 74 (-27, 170), p = 0.72. There was no difference in need for vasopressors or serious adverse events between arms. Among 28 lung recipients in whom detailed outcomes were available, duration of mechanical ventilation, ICU stay and hospital stay were not different by treatment arm. CONCLUSIONS An open lung protective ventilator strategy was safe but did not improve donor lung utilization or oxygenation compared to a conventional ventilator strategy in a population of US organ donors. NCT03439995.
Collapse
Affiliation(s)
- Lorraine B Ware
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Tatsuki Koyama
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ciara M Shaver
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Gundeep Dhillon
- Department of Medicine, Stanford University, Palo Alto, California
| | - Nancy Wickersham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julia Maheshwari
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - S Samuel Weigt
- Department of Medicine, University of California, Los Angeles, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Michael A Matthay
- Department of Medicine, University of California, San Francisco, California; Department of Anesthesia and the Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
17
|
Kolaitis NA, Barnes H, Levine DJ, Castillo H, Arcasoy SM, Bacchetta M, Benvenuto L, Berman‐Rosenzweig E, Cevasco M, Demarest CT, Dewachter C, Erasmus ME, Glanville AR, Granton J, Keshavjee S, Khangoora V, Krishnan S, Mercier O, Miltiades AN, Montani D, Murphy E, Robbins I, Rahaghi FF, Saddoughi SA, Savale L, Simon MA, Vachiery J, Ventetuolo CE, Whitford HM, Girgis RE. Approach to Lung Transplantation in Pulmonary Arterial Hypertension: A Delphi Consensus on Behalf of the Transplant Task Force of the Pulmonary Vascular Research Institute. Pulm Circ 2025; 15:e70088. [PMID: 40276473 PMCID: PMC12018530 DOI: 10.1002/pul2.70088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Lung transplantation is indicated for selected patients with advanced pulmonary arterial hypertension (PAH). We used a modified Delphi process to develop recommendations on care of patients with PAH undergoing lung transplantation. This Delphi panel was recruited from the Pulmonary Vascular Research Institute's Innovative Drug Discovery Initiative - Lung Transplantation Workstream, consisting of clinical and research experts in PAH and lung transplantation. In this process, 29 panelists were given open-ended questions, querying topics related to lung transplantation in PAH. A steering group converted the responses into discrete statements. Panelists then rated agreement using a Likert scale in two further survey rounds: -5 (strongly disagree) to 5 (strongly agree). Consensus was defined as mean ≥ 2.5 or ≤ -2.5, with a standard deviation not crossing zero. Consensus was reached on 141 of 223 statements. Notable areas of consensus were for early discussions about transplantation, and agreement with previously published referral and listing criteria. There was agreement that lung transplantation could be offered in sick candidates, including those with concurrent renal or hepatic insufficiency. Bilateral lung transplantation was considered the procedure of choice for most patients, with rare indications for heart-lung transplantation. Consensus on bridging strategies included use of veno-arterial extracorporeal membrane oxygenation and preemptive awake cannulation in those with severe right ventricular dysfunction. Consensus was also achieved on intraoperative use of invasive hemodynamic monitoring, and prolonged postoperative circulatory support guided by hemodynamic response and echocardiography. Patients with PAH undergoing transplantation require specialized management, which differs somewhat from other candidates.
Collapse
Affiliation(s)
| | | | | | | | - Selim M. Arcasoy
- Columbia University Irving Medical Center, New York‐Presbyterian HospitalNew YorkNew YorkUSA
| | | | - Luke Benvenuto
- Columbia University Irving Medical Center, New York‐Presbyterian HospitalNew YorkNew YorkUSA
| | - Erika Berman‐Rosenzweig
- Columbia University Irving Medical Center, New York‐Presbyterian HospitalNew YorkNew YorkUSA
| | | | | | | | | | | | | | | | | | | | - Olaf Mercier
- Department of thoracic surgery and Heart‐lung transplantation, Marie Lannelongue HospitalUniversité Paris‐SaclayLe Plessis RobinsonFrance
| | - Andrea N. Miltiades
- Columbia University Irving Medical Center, New York‐Presbyterian HospitalNew YorkNew YorkUSA
| | - David Montani
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de BicêtreUniversité Paris–Saclay, AP‐HP, INSERM UMR_S 999Le Kremlin BicêtreFrance
| | | | - Ivan Robbins
- Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | | | - Laurent Savale
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de BicêtreUniversité Paris–Saclay, AP‐HP, INSERM UMR_S 999Le Kremlin BicêtreFrance
| | - Marc A. Simon
- University of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | | | | | | |
Collapse
|
18
|
Prada G, Daubenspeck D, Chan EG, Sanchez PG, Martin AK. Take a Deep Breath: Operating Room Extubation After Bilateral Lung Transplantation on Venoarterial Extracorporeal Membrane Oxygenation. J Cardiothorac Vasc Anesth 2025; 39:836-848. [PMID: 39788803 DOI: 10.1053/j.jvca.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025]
Affiliation(s)
- Gabriel Prada
- Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC.
| | - Danisa Daubenspeck
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL
| | - Ernest G Chan
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Pablo G Sanchez
- Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Archer Kilbourne Martin
- Division of Cardiovascular and Thoracic Anesthesiology, Mayo Clinic College of Medicine and Science, Mayo Clinic Florida, Jacksonville, FL
| |
Collapse
|
19
|
Nelson NC, Wong KK, Mahoney IJ, Malik T, Rudym D, Lesko MB, Qayum S, Lewis TC, Chang SH, Chan JCY, Geraci TC, Li Y, Pamar P, Schnier J, Singh R, Collazo D, Chang M, Kyeremateng Y, McCormick C, Borghi S, Patel S, Darawshy F, Barnett CR, Sulaiman I, Kugler MC, Brosnahan SB, Singh S, Tsay JCJ, Wu BG, Pass HI, Angel LF, Segal LN, Natalini JG. Lung allograft dysbiosis associates with immune response and primary graft dysfunction. J Heart Lung Transplant 2025; 44:422-434. [PMID: 39561864 PMCID: PMC11956144 DOI: 10.1016/j.healun.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Lower airway enrichment with oral commensals has been previously associated with severe primary graft dysfunction (PGD) after lung transplantation (LT). We aimed to determine whether this dysbiotic signature is present across all PGD severity grades and whether it is associated with a distinct host inflammatory endotype. METHODS Lower airway samples from 96 LT recipients were used to evaluate the lung allograft microbiota via 16S rRNA gene sequencing. Bronchoalveolar lavage (BAL) cytokine concentrations and cell differential percentages were compared across PGD grades. In a subset of samples, we evaluated the lower airway host transcriptome using RNA sequencing methods. RESULTS Differential analyses demonstrated lower airway enrichment with supraglottic-predominant taxa (SPT) in moderate and severe PGD. Dirichlet multinomial mixtures modeling identified 2 distinct microbial clusters. A greater percentage of subjects with moderate-severe PGD than no PGD were identified within the dysbiotic cluster (C-SPT, 48% and 29%, respectively) though this did not reach statistical significance (p = 0.06). PGD severity associated with increased BAL neutrophil concentration (p = 0.03) and correlated with BAL concentrations of MCP-1/CCL2, IP-10/CXCL10, IL-10, and TNF-α (p < 0.05). Furthermore, signatures of dysbiosis correlated with neutrophils, MCP-1/CCL-2, IL-10, and TNF-α (p < 0.05). C-SPT exhibited differential expression of TNF, SERPINE1, MPO, and MMP1 genes and upregulation of MAPK pathways, host signling associated with neutrophilic inflammation. CONCLUSIONS Lower airway dysbiosis within the lung allograft is associated with a neutrophilic inflammatory endotype, an immune profile commonly recognized as the hallmark for PGD. These data highlight a putative role of lower airway microbial dysbiosis in the pathogenesis of this syndrome.
Collapse
Affiliation(s)
- Nathaniel C Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Kendrew K Wong
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Ian J Mahoney
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Tahir Malik
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Darya Rudym
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Melissa B Lesko
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Seema Qayum
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Tyler C Lewis
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Stephanie H Chang
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Justin C Y Chan
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Travis C Geraci
- NYU Langone Transplant Institute, NYU Langone Health, New York, New York; Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Yonghua Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Prerna Pamar
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Joseph Schnier
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Rajbir Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Destiny Collazo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Miao Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Yaa Kyeremateng
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Colin McCormick
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Sara Borghi
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Shrey Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Fares Darawshy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; The Institute of Pulmonology, Hadassah Medical Center, Jerusalem, Israel; Department of Medicine, The Faculty of Medicine at the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Clea R Barnett
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Imran Sulaiman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Department of Respiratory Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Respiratory Medicine, Beaumont Hospital, Dublin, Ireland
| | - Matthias C Kugler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Shari B Brosnahan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Shivani Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Jun-Chieh J Tsay
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, New York
| | - Benjamin G Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, New York
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Grossman School of Medicine, New York, New York
| | - Luis F Angel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York
| | - Leopoldo N Segal
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Jake G Natalini
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, New York; NYU Langone Transplant Institute, NYU Langone Health, New York, New York.
| |
Collapse
|
20
|
Bechet NB, Celik A, Mittendorfer M, Wang Q, Huzevka T, Kjellberg G, Boden E, Hirdman G, Pierre L, Niroomand A, Olm F, McCully JD, Lindstedt S. Xenotransplantation of mitochondria: A novel strategy to alleviate ischemia-reperfusion injury during ex vivo lung perfusion. J Heart Lung Transplant 2025; 44:448-459. [PMID: 39536924 DOI: 10.1016/j.healun.2024.10.033] [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: 06/05/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) plays a crucial role in the development of primary graft dysfunction (PGD) following lung transplantation. A promising novel approach to optimize donor organs before transplantation and reduce the incidence of PGD is mitochondrial transplantation. METHODS In this study, we explored the delivery of isolated mitochondria in 4 hours ex vivo lung perfusion (EVLP) before transplantation as a means to mitigate IRI. To provide a fresh and viable source of mitochondria, as well as to streamline the workflow without the need for donor muscle biopsies, we investigated the impact of autologous, allogeneic, and xenogeneic mitochondrial transplantation. In the xenogeneic settings, isolated mitochondria from mouse liver were utilized while autologous and allogeneic sources came from pig skeletal muscle biopsies. RESULTS Treatment with mitochondrial transplantation increased the P/F ratio and reduced pulmonary peak pressure of the lungs during EVLP, compared to lungs without any mitochondrial transplantation, indicating IRI mitigation. Extensive investigations using advanced light and scanning electron microscopy did not reveal evidence of acute rejection in any of the groups, indicating safe xenotransplantation of mitochondria. CONCLUSIONS Future work is needed to further explore this novel therapy for combating IRI in lung transplantation, where xenotransplantation of mitochondria may serve as a fresh, viable source to reduce IRI.
Collapse
Affiliation(s)
- Nicholas B Bechet
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Aybuke Celik
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden; Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Margareta Mittendorfer
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Qi Wang
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Tibor Huzevka
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden; Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Gunilla Kjellberg
- Department of Thoracic Surgery and Anesthesiology, Uppsala University Hospital, Uppsala, Sweden
| | - Embla Boden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Gabriel Hirdman
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Leif Pierre
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Anna Niroomand
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Franziska Olm
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - James D McCully
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Sandra Lindstedt
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
21
|
He J, Xu X, Yang C, Peng G, Shi J, Ju C, Liu H, Lan L, Liu X, Sang L, Liu X, Wang L, Liang H, Huang D, Zhong N. New classification and precise prevention strategies for donor lung injury in lung transplantation. J Thorac Dis 2025; 17:1118-1121. [PMID: 40083517 PMCID: PMC11898357 DOI: 10.21037/jtd-24-1320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Jianxing He
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Xin Xu
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Chao Yang
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Guilin Peng
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Jiang Shi
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Chunrong Ju
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Hui Liu
- Department of Anesthesiology, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Centre for Respiratory Disease, Guangzhou, China
| | - Lan Lan
- Department of Anesthesiology, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Centre for Respiratory Disease, Guangzhou, China
| | - Xiaoyou Liu
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ling Sang
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Centre for Respiratory Disease, Guangzhou, China
| | - Xuesong Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, National Clinical Centre for Respiratory Disease, Guangzhou, China
| | - Lulin Wang
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Hengrui Liang
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Danxia Huang
- Department of Organ Transplantation, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
22
|
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
|
23
|
Gao X, Tang M, Li J, Ma J, Liu Z, Liu W. Activation of Nrf2 pathway by 4-Octyl itaconate enhances donor lung function in cold preservation settings. Respir Res 2025; 26:69. [PMID: 40016745 PMCID: PMC11869626 DOI: 10.1186/s12931-025-03151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Lung transplantation is the primary treatment for end-stage lung diseases. However, ischemia-reperfusion injury (IRI) significantly impacts transplant outcomes. 4-Octyl itaconate (4-OI) has shown potential in mitigating organ IRI, although its effects in lung transplantation require further exploration. METHODS BEAS-2B cells were used to model transplantation, assessing the effects of 4-OI through viability, apoptosis, and ROS assays. qRT-PCR analyzed cytokine transcription post-cold ischemia/reperfusion (CI/R). RNA sequencing and Gene Ontology analysis elucidated 4-OI's mechanisms of action, confirmed by Western blotting. ALI-airway and lung transplantation organoid models evaluated improvements in bronchial epithelial morphology and function due to 4-OI. ELISA measured IL-6 and IL-8 levels. Rat models of extended cold preservation and non-heart-beating transplantation assessed 4-OI's impact on lung function, injury, and inflammation. RESULTS Our findings indicate that 4-OI (100 µM) during cold preservation effectively maintained cell viability, decreased apoptosis, and reduced ROS production in BEAS-2B cells under CI/R conditions. It also downregulated pro-inflammatory cytokine transcription, including IL1B, IL6, and TNF. Inhibition of Nrf2 partially reversed these protective effects. In cold preservation solutions, 4-OI upregulated Nrf2 target genes such as NQO1, HMOX1, and SLC7A11. In ALI airway models, 4-OI enhanced bronchial epithelial barrier integrity and ciliary beat function after CI/R. In rat models, 4-OI administration improved lung function and reduced pulmonary edema, tissue injury, apoptosis, and systemic inflammation following extended cold preservation or non-heart-beating lung transplantation. CONCLUSIONS Incorporating 4-OI into cold preservation solutions appears promising for alleviating CI/R-induced bronchial epithelial injury and enhancing lung transplant outcomes via Nrf2 pathway activation.
Collapse
Affiliation(s)
- Xinliang Gao
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jianzun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhengrui Liu
- Changchun Yifu Jilin Province Academician Workstation, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
24
|
Niethamer TK, Planer JD, Morley MP, Babu A, Zhao G, Basil MC, Cantu E, Frank DB, Diamond JM, Nottingham AN, Li S, Sharma A, Hallquist H, Levin LI, Zhou S, Vaughan AE, Morrisey EE. Longitudinal single-cell profiles of lung regeneration after viral infection reveal persistent injury-associated cell states. Cell Stem Cell 2025; 32:302-321.e6. [PMID: 39818203 PMCID: PMC11805657 DOI: 10.1016/j.stem.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
Functional regeneration of the lung's gas exchange surface following injury requires the coordination of a complex series of cell behaviors within the alveolar niche. Using single-cell transcriptomics combined with lineage tracing of proliferating progenitors, we examined mouse lung regeneration after influenza injury, demonstrating an asynchronously phased response across different cellular compartments. This longitudinal atlas of injury responses has produced a catalog of transient and persistent transcriptional alterations in cells as they transit across axes of differentiation. These cell states include an injury-induced capillary endothelial cell (iCAP) that arises after injury, persists indefinitely, and shares hallmarks with developing lung endothelium and endothelial aberrations found in degenerative human lung diseases. This dataset provides a foundational resource to understand the complexity of cellular and molecular responses to injury and correlations to responses found in human development and disease.
Collapse
Affiliation(s)
- Terren K Niethamer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Joseph D Planer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Apoorva Babu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gan Zhao
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Cantu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Frank
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana N Nottingham
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arnav Sharma
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Hannah Hallquist
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lillian I Levin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
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
|
26
|
Kawana S, Okazaki M, Sakaue T, Hashimoto K, Nakata K, Choshi H, Tanaka S, Miyoshi K, Ohtani S, Ohara T, Sugimoto S, Matsukawa A, Toyooka S. Loss of Nr4a1 ameliorates endothelial cell injury and vascular leakage in lung transplantation from circulatory-death donor. J Heart Lung Transplant 2025; 44:249-260. [PMID: 39369968 DOI: 10.1016/j.healun.2024.09.028] [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: 02/22/2024] [Revised: 09/20/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) stands as a major trigger for primary graft dysfunction (PGD) in lung transplantation (LTx). Especially in LTx from donation after cardiac death (DCD), effective control of IRI following warm ischemia (WIRI) is crucial to prevent PGD. This study aimed to identify the key factors affecting WIRI in LTx from DCD. METHODS Previously reported RNA-sequencing dataset of lung WIRI was reanalyzed to identify nuclear receptor subfamily 4 group A member 1 (NR4A1) as the immediate early gene for WIRI. Dynamics of NR4A1 expression were verified using a mouse hilar clamp model. To investigate the role of NR4A1 in WIRI, a mouse model of LTx from DCD was established using Nr4a1 knockout (Nr4a1-/-) mice. RESULTS NR4A1 was located around vascular cells, and its protein levels in the lungs increased rapidly and transiently during WIRI. LTx from Nr4a1-/- donors significantly improved pulmonary graft function compared to wild-type donors. Histological analysis showed decreased microvascular endothelial cell death, neutrophil infiltration, and albumin leakage. Evans blue permeability assay demonstrated maintained pulmonary microvascular barrier integrity in grafts from Nr4a1-/- donors, correlating with diminished pulmonary edema. However, NR4A1 did not significantly affect the inflammatory response during WIRI, and IRI was not suppressed when a wild-type donor lung was transplanted into the Nr4a1-/- recipient. CONCLUSIONS Donor NR4A1 plays a specialized role in the positive regulation of endothelial cell injury and microvascular hyperpermeability. These findings demonstrate the potential of targeting NR4A1 interventions to alleviate PGD and improve outcomes in LTx from DCD.
Collapse
Affiliation(s)
- Shinichi Kawana
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Tomohisa Sakaue
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan; Department of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Kohei Hashimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Nakata
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Haruki Choshi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Tanaka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaroh Miyoshi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Ohtani
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
27
|
Carlier FM, Dumonceaux M, Planté-Bordeneuve T, Evrard P, Marchand E, Belhaj A, Rondelet B, Demeure F. Transcatheter mitral valve repair with MitraClip® enabling single lung transplantation: A case report. Heliyon 2025; 11:e41222. [PMID: 39802031 PMCID: PMC11721235 DOI: 10.1016/j.heliyon.2024.e41222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Post-capillary hypertension resulting from mitral regurgitation is typically considered a contraindication for single lung transplantation due to heightened risks of primary graft dysfunction. This case report highlights a 66-year-old COPD patient with severe mitral regurgitation who was deemed ineligible for surgical mitral replacement. As an alternative, transcatheter mitral valve replacement was successfully performed, resulting in the normalization of pulmonary artery pressures. Consequently, the patient became eligible for single lung transplantation, which was conducted successfully in the subsequent months. Eighteen months post-lung transplantation, the patient now experiences a normal functional status and excellent lung function. In conclusion, transcatheter mitral valve replacement appears to be a safe alternative to surgery for normalizing post-capillary pulmonary hypertension in patients with chronic respiratory diseases. This approach could potentially facilitate lung transplantation (LTx) in eligible candidates.
Collapse
Affiliation(s)
- François M. Carlier
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Department of Pneumology, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Michel Dumonceaux
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Department of Pneumology, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Thomas Planté-Bordeneuve
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Department of Pneumology, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Patrick Evrard
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Intensive Care Unit, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Eric Marchand
- Department of Pneumology, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Asmae Belhaj
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Department of Cardio-Thoracic Surgery, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Benoît Rondelet
- Lung Transplant Centre, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
- Department of Cardio-Thoracic Surgery, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| | - Fabian Demeure
- Department of Cardiology, CHU Mont-Godinne UCL Namur, Yvoir, Belgium
| |
Collapse
|
28
|
Xia W, Liu W, He Z, Song C, Liu J, Chen R, Chen J, Wang X, Xu H, Mao W. Machine Learning for Predicting Primary Graft Dysfunction After Lung Transplantation: An Interpretable Model Study. Transplantation 2025:00007890-990000000-00978. [PMID: 39789697 DOI: 10.1097/tp.0000000000005326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Primary graft dysfunction (PGD) develops within 72 h after lung transplantation (Lung Tx) and greatly influences patients' prognosis. This study aimed to establish an accurate machine learning (ML) model for predicting grade 3 PGD (PGD3) after Lung Tx. METHODS This retrospective study incorporated 802 patients receiving Lung Tx between July 2018 and October 2023 (640 in the derivation cohort and 162 in the external validation cohort), and 640 patients were randomly assigned to training and internal validation cohorts in a 7:3 ratio. Independent risk factors for PGD3 were determined by integrating the univariate logistic regression and least absolute shrinkage and selection operator regression analyses. Subsequently, 9 ML models were used to construct prediction models for PGD3 based on selected variables. Their prediction performances were further evaluated. Besides, model stratification performance was assessed with 3 posttransplant metrics. Finally, the SHapley Additive exPlanations algorithm was used to understand the predictive importance of selected variables. RESULTS We identified 9 independent clinical risk factors as selected variables. Among 9 ML models, the random forest (RF) model displayed optimal performance (area under the curve [AUC] = 0.9415, sensitivity [Se] = 0.8972, specificity [Sp] = 0.8795 in the training cohort; AUC = 0.7975, Se = 0.7520, Sp = 0.7313 in the internal validation cohort; and AUC = 0.8214, Se = 0.8235, Sp = 0.6667 in the external validation cohort). Further assessments on calibration and clinical usefulness indicated the promising applicability of the RF model in PGD3 prediction. Meanwhile, the RF model also performed best in terms of risk stratification for postoperative support (extracorporeal membrane oxygenation time: P < 0.001, mechanical ventilation time: P = 0.006, intensive care unit time: P < 0.001). CONCLUSIONS The RF model had the optimal performance in PGD3 prediction and postoperative risk stratification for patients after Lung Tx.
Collapse
Affiliation(s)
- Wei Xia
- Department of Intensive Care Unit, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Weici Liu
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhao He
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Chenghu Song
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jiwei Liu
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ruo Chen
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jingyu Chen
- Department of Lung Transplantation, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xiaokun Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Hongyang Xu
- Department of Intensive Care Unit, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Wenjun Mao
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
29
|
Koyama T, Zhao Z, Balmes JR, Calfee CS, Matthay MA, Reilly JP, Porteous MK, Diamond JM, Christie JD, Cantu E, Ware LB. Long-term air pollution exposure and the risk of primary graft dysfunction after lung transplantation. J Heart Lung Transplant 2025; 44:64-74. [PMID: 39019353 DOI: 10.1016/j.healun.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Primary graft dysfunction (PGD) contributes substantially to both short- and long-term mortality after lung transplantation, but the mechanisms that lead to PGD are not well understood. Exposure to ambient air pollutants is associated with adverse events during waitlisting for lung transplantation and chronic lung allograft dysfunction, but its association with PGD has not been studied. We hypothesized that long-term exposure of the lung donor and recipient to high levels of ambient air pollutants would increase the risk of PGD in lung transplant recipients. METHODS Using data from 1428 lung transplant recipients and their donors enrolled in the Lung Transplant Outcomes Group observational cohort study, we evaluated the association between the development of PGD and zip-code-based estimates of long-term exposure to 6 major air pollutants (ozone, nitrogen dioxide, sulfur dioxide, carbon monoxide, particulate matter 2.5, and particulate matter 10) in both the lung donor and the lung recipient. Exposure estimates used daily EPA air pollutant monitoring data and were based on the geographic centroid of each subject's residential zip code. Associations were tested in both univariable and multivariable models controlling for known PGD risk factors. RESULTS We did not find strong associations between air pollutant exposures in either the donor or the recipient and PGD. CONCLUSIONS Exposure to ambient air pollutants, at the levels observed in this study, may not be sufficiently harmful to prime the donor lung or the recipient to develop PGD, particularly when considering the robust associations with other established PGD risk factors.
Collapse
Affiliation(s)
- Tatsuki Koyama
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zhiguo Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John R Balmes
- Department of Medicine, University of California, San Francisco, California
| | - Carolyn S Calfee
- Department of Medicine, University of California, San Francisco, California; Department of Anesthesia and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Michael A Matthay
- Department of Medicine, University of California, San Francisco, California; Department of Anesthesia and Cardiovascular Research Institute, University of California, San Francisco, California
| | - John P Reilly
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary K Porteous
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D Christie
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
30
|
Ju YN, Li H, Zhuo ZP, Yang Q, Gao W. Mitochondrial DNA from endothelial cells activated the cGAS-STING pathway and regulated pyroptosis in lung ischaemia reperfusion injury after lung transplantation. Immunobiology 2025; 230:152865. [PMID: 39826223 DOI: 10.1016/j.imbio.2024.152865] [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: 08/28/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Cell dysfunction and death induced by lung ischaemia-reperfusion injury (LIRI) are the main causes of death in transplant patients. Activation of the cGAS-STING-induced immune response and death plays a critical role in multiple organ injuries. However, no study has yet investigated the role of the cGAS-STING pathway in LIRI after lung transplantation. METHODS Sprague-Dawley (SD) rats were subjected to left lung transplantation and administered inhibitors of cGAS and STING. The expression of cGAS-STING-TBK1-IRF3, histological injury, pulmonary permeability, and the levels of cytokines and pyroptotic proteins in transplanted lungs were tested. Endothelial cells were subjected to hypoxemia and reoxygenation and treated with inhibitors of cGAS and STING. Mitochondrial DNA (mtDNA), the cGAS-STING axis and cytokine levels in cells, cellular activity and death were evaluated. Moreover, after the administration of deoxyribonuclease (DNase) I, the reoxygenated endothelial cells were also examined for cellular function and inflammatory factor expression. Finally, we administered an agonist of STING and an inhibitor of cathepsin B to the normal endothelium and investigated pyroptosis and pyroptotic proteins. RESULTS After 24 h of reperfusion, the expression of cGAS-STING-TBK1-IRF3 and pyroptotic proteins was significantly increased, and inhibitors of cGAS or STING ameliorated lung injury and reduced pyroptotic protein levels. In vitro, the inhibition of cGAS and STING reduced the activation of TBK and IRF3 and reduced cellular injury and death. The activation of cGAS-STING and cellular inflammation were suppressed by DNase I. Cathepsin B and NLRP3 were upregulated by an agonist of STING, and an inhibitor of cathepsin B reduced NLRP3 levels. CONCLUSION cGAS-STING participated in LIRI by promoting endothelial cell pyroptosis via cathepsin B.
Collapse
Affiliation(s)
- Ying-Nan Ju
- Department of Intensive Care Unit, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570 311, China
| | - Hu Li
- Department of Critical Care Medicine, Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| | - Zi-Peng Zhuo
- Department of Critical Care Medicine, Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| | - Qing Yang
- Department of Critical Care Medicine, Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| | - Wei Gao
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570311, China.
| |
Collapse
|
31
|
Scaravilli V, Scansani S, Meani P, Turconi G, Guzzardella A, Bosone M, Bonetti C, Vicenzi M, Morlacchi LC, Rossetti V, Rosso L, Blasi F, Nosotti M, Grasselli G. Right ventricle free wall longitudinal strain screening of lung transplant candidates. PLoS One 2024; 19:e0314235. [PMID: 39705303 PMCID: PMC11661623 DOI: 10.1371/journal.pone.0314235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/25/2024] [Indexed: 12/22/2024] Open
Abstract
BACKGROUND Lung transplant (LUTX) candidates have subclinical right ventricular (RV) dysfunction, which has not yet been assessed by speckle-tracking echocardiography (STE)-derived RV free-wall longitudinal strain (RVFWLS). To evaluate the prevalence of RV dysfunction by RVFWLS and its relationship with conventional RV echocardiographic indexes in LUTX candidates. METHODS In a single-center prospective observational cohort study, from January 2021 to March 2023 consecutive LUTX candidates underwent cardiac catheterization, radionuclide ventriculography, standard and STE. The diagnostic accuracy of RV ejection fraction by ventriculography (RVEF), tricuspid annular plane excursion (TAPSE), fractional area change (FAC), tricuspid peak annulus systolic velocity (S') versus RVFWS were computed. RESULTS Thirty-four patients (female, 41%) with a mean age of 48 [36-59] years old enlisted for pulmonary fibrosis (35%) and cystic fibrosis (30%) were included. At cardiac catheterization, only 7 (23%) had pulmonary hypertension. Around 15-25% presented right heart enlargement. Tricuspid regurgitation was present in 20 (60%) of the patients. Median RVFWLS was -20.1% [-22.5%--17%], being impaired (> -20%) in 16 (47%) of the patients. RVFWLS identified the highest percentage (47%) of RV dysfunction, compared to TAPSE (32%), S' (27%), FAC (26%), and ventriculography (15%), which had very low sensitivity for detecting RV dysfunction compared to RVFWLS. CONCLUSIONS In patients enlisted for LUTX, RV dysfunction assessed by STE-derived RVFWLS is highly prevalent. STE can detect RV dysfunction better than standard two-dimensional echocardiography and ventriculography. Further studies are urgently needed to define the clinical implications and the prognostic value of RV dysfunction measured with RVFWLS.
Collapse
Affiliation(s)
- Vittorio Scaravilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan (MI), Italy
| | - Silvia Scansani
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Paolo Meani
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
- Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Gloria Turconi
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Amedeo Guzzardella
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Marco Bosone
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Claudia Bonetti
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| | - Marco Vicenzi
- Department of Cardio-thoraco-vascular diseases, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
- Dipartimento di Scienze Cliniche e di Comunità, University of Milan, Milan (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
| | - Valeria Rossetti
- Department of Internal Medicine, Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Lorenzo Rosso
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
- Department of Cardio-thoraco-vascular diseases, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (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
| | - Mario Nosotti
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
- Department of Cardio-thoraco-vascular diseases, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan (MI), Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan (MI), Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan (MI), Italy
| |
Collapse
|
32
|
Wang JY, Michki SN, Sitaraman S, Banaschewski BJ, Jamal R, Gokey JJ, Lin SM, Katzen JB, Basil MC, Cantu E, Kropski JA, Zepp JA, Frank DB, Young LR. Dysregulated alveolar epithelial cell progenitor function and identity in Hermansky-Pudlak syndrome. JCI Insight 2024; 10:e183483. [PMID: 39699958 PMCID: PMC11948584 DOI: 10.1172/jci.insight.183483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024] Open
Abstract
Hermansky-Pudlak syndrome (HPS) is a genetic disorder of endosomal protein trafficking associated with pulmonary fibrosis in specific subtypes, including HPS-1 and HPS-2. Single-mutant HPS1 and HPS2 mice display increased fibrotic sensitivity while double-mutant HPS1/2 mice exhibit spontaneous fibrosis with aging, which has been attributed to HPS mutations in alveolar epithelial type II (AT2) cells. We utilized HPS mouse models and human lung tissue to investigate mechanisms of AT2 cell dysfunction driving fibrotic remodeling in HPS. Starting at 8 weeks of age, HPS mice exhibited progressive loss of AT2 cell numbers. HPS AT2 cell function was impaired ex vivo and in vivo. Incorporating AT2 cell lineage tracing in HPS mice, we observed aberrant differentiation with increased AT2-derived alveolar epithelial type I cells. Transcriptomic analysis of HPS AT2 cells revealed elevated expression of genes associated with aberrant differentiation and p53 activation. Lineage-tracing and organoid-modeling studies demonstrated that HPS AT2 cells were primed to persist in a Keratin-8-positive reprogrammed transitional state, mediated by p53 activity. Intrinsic AT2 progenitor cell dysfunction and p53 pathway dysregulation are mechanisms of disease in HPS-related pulmonary fibrosis, with the potential for early targeted intervention before the onset of fibrotic lung disease.
Collapse
Affiliation(s)
- Joanna Y. Wang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sylvia N. Michki
- Division of Cardiology, Department of Pediatrics, and
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sneha Sitaraman
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brandon J. Banaschewski
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Reshma Jamal
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jason J. Gokey
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Susan M. Lin
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
| | - Jeremy B. Katzen
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
| | - Maria C. Basil
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward Cantu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Jarod A. Zepp
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
| | - David B. Frank
- Division of Cardiology, Department of Pediatrics, and
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lisa R. Young
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Lung Biology Institute and
| |
Collapse
|
33
|
Wang JY, Michki SN, Sitaraman S, Banaschewski BJ, Jamal R, Gokey JJ, Lin SM, Katzen JB, Basil MC, Cantu E, Kropski JA, Zepp JA, Frank DB, Young LR. Dysregulated alveolar epithelial cell progenitor function and identity in Hermansky-Pudlak syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.17.545390. [PMID: 38496421 PMCID: PMC10942273 DOI: 10.1101/2023.06.17.545390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Hermansky-Pudlak syndrome (HPS) is a genetic disorder of endosomal protein trafficking associated with pulmonary fibrosis in specific subtypes, including HPS-1 and HPS-2. Single mutant HPS1 and HPS2 mice display increased fibrotic sensitivity while double mutant HPS1/2 mice exhibit spontaneous fibrosis with aging, which has been attributed to HPS mutations in alveolar epithelial type II (AT2) cells. We utilized HPS mouse models and human lung tissue to investigate mechanisms of AT2 cell dysfunction driving fibrotic remodeling in HPS. Starting at 8 weeks of age, HPS mice exhibited progressive loss of AT2 cell numbers. HPS AT2 cell function was impaired ex vivo and in vivo . Incorporating AT2 cell lineage tracing in HPS mice, we observed aberrant differentiation with increased AT2-derived alveolar epithelial type I cells. Transcriptomic analysis of HPS AT2 cells revealed elevated expression of genes associated with aberrant differentiation and p53 activation. Lineage tracing and organoid modeling studies demonstrated that HPS AT2 cells were primed to persist in a Krt8 + reprogrammed transitional state, mediated by p53 activity. Intrinsic AT2 progenitor cell dysfunction and p53 pathway dysregulation are novel mechanisms of disease in HPS-related pulmonary fibrosis, with the potential for early targeted intervention before the onset of fibrotic lung disease.
Collapse
|
34
|
Mittendorfer M, Pierre L, Huzevka T, Schofield J, Abrams ST, Wang G, Toh CH, Bèchet NB, Caprnja I, Kjellberg G, Aswani A, Olm F, Lindstedt S. Restoring discarded porcine lungs by ex vivo removal of neutrophil extracellular traps. J Heart Lung Transplant 2024; 43:1919-1929. [PMID: 39038563 DOI: 10.1016/j.healun.2024.07.007] [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: 02/12/2024] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND By causing inflammation and tissue damage, neutrophil extracellular traps (NETs) constitute an underlying mechanism of aspiration-induced lung injury, a major factor of the low utilization of donor lungs in lung transplantation (LTx). METHODS To determine whether NET removal during ex vivo lung perfusion (EVLP) can restore lung function and morphology in aspiration-damaged lungs, gastric aspiration lung injury was induced in 12 pigs. After confirmation of acute respiratory distress syndrome, the lungs were explanted and assigned to NET removal connected to EVLP (treated) (n = 6) or EVLP only (nontreated) (n = 6). Hemodynamic measurements were taken, and blood and tissue samples were collected to assess lung function, morphology, levels of cell-free DNA, extracellular histones, and nucleosomes as markers of NETs, as well as cytokine levels. RESULTS After EVLP and NET removal in porcine lungs, PaO2/FiO2 ratios increased significantly compared to those undergoing EVLP alone (p = 0.0411). Treated lungs had lower cell-free DNA (p = 0.0260) and lower levels of extracellular histones in EVLP perfusate (p= 0.0260) than nontreated lungs. According to histopathology, treated lungs showed less immune cell infiltration and less edema compared with nontreated lungs, which was reflected in decreased levels of proinflammatory cytokines in EVLP perfusate and bronchoalveolar lavage fluid. CONCLUSIONS To conclude, removing NETs during EVLP improved lung function and morphology in aspiration-damaged donor lungs. The ability to remove NETs during EVLP could represent a new therapeutic approach for LTx and potentially expand the donor pool for transplantation.
Collapse
Affiliation(s)
- Margareta Mittendorfer
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Leif Pierre
- Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Tibor Huzevka
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Jeremy Schofield
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Nicholas B Bèchet
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Ilma Caprnja
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Gunilla Kjellberg
- Department of Thoracic Surgery and Anaesthesiology, Uppsala University Hospital, Uppsala, Sweden
| | - Andrew Aswani
- Department of Critical Care, Guy's and St Thomas's NHS Foundation Trust, London, United Kingdom; Santersus AG, Zurich, Switzerland
| | - Franziska Olm
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden.
| |
Collapse
|
35
|
Hauser BR, Estafanos M, Ayyat KS, Yun JJ, Elgharably H. Current status of routine use of veno-arterial extracorporeal membrane oxygenation during lung transplantation. Expert Rev Med Devices 2024; 21:1153-1163. [PMID: 39670791 DOI: 10.1080/17434440.2024.2442485] [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: 09/11/2024] [Revised: 11/27/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Recently, there has been growing experience with utilizing a veno-arterial extracorporeal membrane oxygenator (VA ECMO) routinely during lung transplantation procedures. Yet, there is a lack of consensus on the protocols, benefits, and outcomes of routine VA ECMO use in lung transplantation. AREAS COVERED This article presents an overview of the current status of routine use of VA ECMO during lung transplantation, including rationale, protocols, applications, and outcomes. EXPERT OPINION Utilization of VA ECMO during lung transplantation has emerged as an alternative mechanical circulatory support modality to cardiopulmonary bypass, with growing evidence showing lower rates of peri-operative complications. Some groups took that further into routine application of VA ECMO during lung transplantation. The current available evidence suggests that routine utilization of VA ECMO during lung transplantation is associated with lower rates of primary graft dysfunction and improved early outcomes. Use of VA ECMO allows controlled reperfusion of the allograft and avoids an unplanned "crash" on pump in case of hemodynamic instability, which carries worse outcomes after lung transplantation. As a relatively new approach, further follow-up of growing experience, as well as prospective clinical trials, is necessary to develop a consensus about routine utilization of VA ECMO during lung transplantation.
Collapse
Affiliation(s)
- Benjamin R Hauser
- School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Mina Estafanos
- Department of Surgery, Division of Cardiac Surgery, University of Rochester, Rochester, NY, USA
| | - Kamal S Ayyat
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - James J Yun
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Haytham Elgharably
- Department of Thoracic & Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
36
|
Kim JS, Gupta R. Lung transplantation in pulmonary sarcoidosis. J Autoimmun 2024; 149:103135. [PMID: 37923622 DOI: 10.1016/j.jaut.2023.103135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
Sarcoidosis is a systemic inflammatory disease of unknown etiology and variable clinical course. Pulmonary sarcoidosis is the most common presentation and accounts for most morbidity and mortality related to sarcoidosis. While sarcoidosis generally has good outcomes, few patients experience chronic disease. A minority of patients progress to a specific phenotype of sarcoidosis referred to advanced pulmonary sarcoidosis (APS) which includes advanced fibrosis, pulmonary hypertension and respiratory failure, leading to high morbidity and mortality. In patients with advanced disease despite medical therapy, lung transplantation may be the last viable option for improvement in quality of life. Though post-transplant survival is similar to that of other end-stage lung diseases, it is imperative that patients are evaluated and referred early to transplant centers with experience in APS. A multidisciplinary approach and clinical experience are crucial in detecting the optimal timing of referral, initiating comprehensive transplantation evaluation and listing, discussing surgical approach, and managing perioperative and post-transplant care. This review article seeks to address these aspects of lung transplantation in APS.
Collapse
Affiliation(s)
- Jin Sun Kim
- Lewis Katz School of Medicine, Department of Thoracic Medicine and Surgery, Philadelphia, PA, USA.
| | - Rohit Gupta
- Lewis Katz School of Medicine, Department of Thoracic Medicine and Surgery, Philadelphia, PA, USA
| |
Collapse
|
37
|
Bennett RM, Reilly JP. Environmental Risk Factors for Acute Respiratory Distress Syndrome. Clin Chest Med 2024; 45:797-807. [PMID: 39442998 PMCID: PMC11969571 DOI: 10.1016/j.ccm.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Several environmental exposures increase susceptibility to the acute respiratory distress syndrome (ARDS). Specifically, chronic exposure to ambient air pollution, cigarette smoke, and alcohol "prime" the lung via epithelial injury, endothelial dysfunction, and immunomodulatory mechanisms, increasing the risk and severity of ARDS following an array of acute insults. Future research of these pathways may reveal therapeutic targets. Relevant emerging threats, such as electronic cigarettes and vaping, wildfire smoke, and the environmental hazards associated with climate change, may also be associated with ARDS. Building upon existing public policy interventions can prevent substantial morbidity and mortality from ARDS.
Collapse
Affiliation(s)
- Rachel M Bennett
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - John P Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA 19104, USA; Center for Translational Lung Biology, 5042 Gates Building, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
Edström D, Niroomand A, Stenlo M, Broberg E, Hirdman G, Ghaidan H, Hyllén S, Pierre L, Olm F, Lindstedt S. Amniotic fluid-derived mesenchymal stem cells reduce inflammation and improve lung function following transplantation in a porcine model. J Heart Lung Transplant 2024; 43:2018-2030. [PMID: 39182800 DOI: 10.1016/j.healun.2024.08.014] [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: 10/14/2023] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Lung transplantation is hindered by low donor lung utilization rates. Infectious complications are reasons to decline donor grafts due to fear of post-transplant primary graft dysfunction. Mesenchymal stem cells are a promising therapy currently investigated in treating lung injury. Full-term amniotic fluid-derived lung-specific mesenchymal stem cell treatment may regenerate damaged lungs. These cells have previously demonstrated inflammatory mediation in other respiratory diseases, and we hypothesized that treatment would improve donor lung quality and postoperative outcomes. METHODS In a transplantation model, donor pigs were stratified to either the treated or the nontreated group. Acute respiratory distress syndrome was induced in donor pigs and harvested lungs were placed on ex vivo lung perfusion (EVLP) before transplantation. Treatment consisted of 3 doses of 2 × 106 cells/kg: one during EVLP and 2 after transplantation. Donors and recipients were assessed on clinically relevant parameters and recipients were followed for 3 days before evaluation for primary graft dysfunction (PGD). RESULTS Repeated injection of the cell treatment showed reductions in inflammation seen through lowered immune cell counts, reduced histology signs of inflammation, and decreased cytokines in the plasma and bronchoalveolar lavage fluid. Treated recipients showed improved pulmonary function, including increased PaO2/FiO2 ratios and reduced incidence of PGD. CONCLUSIONS Repeated injection of lung-specific cell treatment during EVLP and post transplant was associated with improved function of previously damaged lungs. Cell treatment may be considered as a potential therapy to increase the number of lungs available for transplantation and the improvement of postoperative outcomes.
Collapse
Affiliation(s)
- Dag Edström
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Anna Niroomand
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, New York
| | - Martin Stenlo
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Ellen Broberg
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Gabriel Hirdman
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Haider Ghaidan
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Snejana Hyllén
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Leif Pierre
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Franziska Olm
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden; Lund Stem Cell Center, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
39
|
Amarelli C, Bello I, Aigner C, Berman M, Boffini M, Clark S, Dalvindt M, de Wolf J, Ensminger S, Gomez de Antonio D, Hoyos L, Palmieri L, Schweiger M, Sponga S, Wiegmann B, Neyrinck A, the ESOT Guidelines Taskforce. European Society of Organ Transplantation (ESOT) Consensus Statement on Machine Perfusion in Cardiothoracic Transplant. Transpl Int 2024; 37:13112. [PMID: 39649067 PMCID: PMC11620879 DOI: 10.3389/ti.2024.13112] [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: 04/08/2024] [Accepted: 09/26/2024] [Indexed: 12/10/2024]
Abstract
The machine perfusion (MP) of transplantable grafts has emerged as an upcoming field in Cardiothoracic (CT) transplantation during the last decade. This technology carries the potential to assess, preserve, and even recondition thoracic grafts before transplantation, so it is a possible game-changer in the field. This technology field has reached a critical turning point, with a growing number of publications coming predominantly from a few leading institutions, but still need solid scientific evidence. Due to the increasing need to expand the donor pool, especially in Europe, where the donor age is steeply increased, a consensus has been established to address the growing need and knowledge of machine perfusion in cardiothoracic transplantation, targeting the unmet scientific need in this growing field but also, priorities for development, and regional differences in utilization rates and organizational issues. To address MP in CT, the European Society of Organ Transplantation (ESOT) convened a dedicated Working group comprised of experts in CT to review literature about MP to develop guidelines that were subsequently discussed and voted on during the Consensus Conference that took place in person in Prague during the TLJ 3.0 in November 2022. The findings and recommendations of the Cardiothoracic Working Group on MP are presented in this article.
Collapse
Affiliation(s)
- Cristiano Amarelli
- Department of Cardiac Surgery and Transplants, Monaldi, Azienda dei Colli, Naples, Italy
| | - Irene Bello
- Institut Clínic Respiratorio, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Clemens Aigner
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Marius Berman
- Transplant Unit, Royal Papworth Hospital, NHS Foundation Trust, Cambridge, United Kingdom
| | - Massimo Boffini
- Cardiac Surgery Division, Surgical Sciences Department, Citta della Salute e della Scienza, University of Torino, Turin, Italy
| | - Stephen Clark
- Department Cardiothoracic Transplant, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marita Dalvindt
- Department of Cardiothoracic Surgery, Lund University, Lund, Sweden
| | - Julien de Wolf
- Department of Thoracic Surgery, Lung Heart Institute, University Hospital of Lille, Lille, France
| | - Stephan Ensminger
- Department of Cardiac and Thoracic Vascular Surgery, University Heart Center Lübeck, Lübeck, Germany
| | - David Gomez de Antonio
- Department of Thoracic Surgery, Puerta de Hierro University Hospital Majadahonda, Madrid, Spain
| | - Lucas Hoyos
- Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Lucrezia Palmieri
- Department of Translational Medical Sciences, Monaldi Hospital, University of Campania “Luigi Vanvitelli“, Naples, Italy
| | - Martin Schweiger
- Department of Congenital Cardiovascular Surgery, Pediatric Heart Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Sandro Sponga
- Division of Cardiac Surgery, Cardiothoracic Department, University Hospital of Udine, Udine, Italy
| | - Bettina Wiegmann
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Arne Neyrinck
- Department of Cardiovascular Sciences, Anesthesiology and Algology, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
40
|
Christie JD, Van Raemdonck D, Fisher AJ. Lung Transplantation. N Engl J Med 2024; 391:1822-1836. [PMID: 39536228 DOI: 10.1056/nejmra2401039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Jason D Christie
- From the Division of Pulmonary Allergy and Critical Care Medicine, Center for Clinical Epidemiology and Biostatistics, Lung Biology Institute at the University of Pennsylvania Perelman School of Medicine, Philadelphia (J.D.C.); the Department of Thoracic Surgery, University Hospitals Leuven, and the Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven University - both in Leuven, Belgium (D.V.R.); and the Department of Transplantation and Regenerative Medicine, Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom (A.J.F.)
| | - Dirk Van Raemdonck
- From the Division of Pulmonary Allergy and Critical Care Medicine, Center for Clinical Epidemiology and Biostatistics, Lung Biology Institute at the University of Pennsylvania Perelman School of Medicine, Philadelphia (J.D.C.); the Department of Thoracic Surgery, University Hospitals Leuven, and the Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven University - both in Leuven, Belgium (D.V.R.); and the Department of Transplantation and Regenerative Medicine, Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom (A.J.F.)
| | - Andrew J Fisher
- From the Division of Pulmonary Allergy and Critical Care Medicine, Center for Clinical Epidemiology and Biostatistics, Lung Biology Institute at the University of Pennsylvania Perelman School of Medicine, Philadelphia (J.D.C.); the Department of Thoracic Surgery, University Hospitals Leuven, and the Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven University - both in Leuven, Belgium (D.V.R.); and the Department of Transplantation and Regenerative Medicine, Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom (A.J.F.)
| |
Collapse
|
41
|
Alderete IS, Medina CK, Pontula A, Halpern SE, Soto AL, Patel KJ, Klapper JA, Hartwig MG. Donor and recipient factors associated with primary graft dysfunction following lung transplantation: A donor management goal registry analysis. J Thorac Cardiovasc Surg 2024:S0022-5223(24)01000-6. [PMID: 39489331 DOI: 10.1016/j.jtcvs.2024.10.045] [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: 05/03/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Current risk-adjusted models for predicting primary graft dysfunction (PGD) following lung transplantation (LTx) do not include bedside donor critical care data. Donor management goals (DMGs) represent predefined critical care endpoints aimed at optimizing multiorgan donor management. Here we sought to identify novel predictors to better understand the relationship between donor management and PGD following LTx. METHODS We used the national DMG registry to identify a cohort of LTx recipients linked to their respective donors between January 1, 2015, and March 1, 2023. Grade 3 PGD (PGD3) was defined according to modified International Society for Heart and Lung Transplantation criteria. Multivariable modeling was performed to identify risk factors for the development of PGD3. RESULTS A total of 2704 eligible patients were identified, of whom 643 (23.8%) developed PGD3. After multivariable modeling, the likelihood of PGD3 was greater with increasing donor age (odds ratio [OR], 1.06; 95% confidence interval [CI], 1.02-1.10 per 5-year change; P = .003), increasing donor serum pH at the time of authorization (OR, 1.14; 95% CI, 1.02-1.25 per 0.1-point increase; P = .016), donor history of cocaine use (OR, 1.34; 95% CI, 1.05-1.71; P = .020), and increased recipient central venous pressure (OR, 1.03; 95% CI, 1.01-1.06; P = .005). Recipients who received donor lungs in which the DMG for PF ratio was met had a lower likelihood of developing PGD3 (OR, 0.63; 95% CI, 0.46-0.86; P = .006). CONCLUSIONS This study leverages a novel detailed donor management database to identify factors associated with the development of PGD3. These factors may be used to recognize donors and recipients who may benefit from early interventions to improve short-term outcomes.
Collapse
Affiliation(s)
- Isaac S Alderete
- Department of Surgery, Duke University School of Medicine, Durham, NC.
| | - Cathlyn K Medina
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Arya Pontula
- University of Manchester Medical School, Manchester, United Kingdom
| | | | - Alexandria L Soto
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Kunal J Patel
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University, Durham, NC
| | - Jacob A Klapper
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University, Durham, NC
| | - Matthew G Hartwig
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University, Durham, NC
| |
Collapse
|
42
|
Miano TA, Zuppa AF, Feng R, Griffiths S, Kalman L, Oyster M, Cantu E, Yang W, Diamond JM, Christie JD, Scheetz MH, Shashaty MG. Development and validation of a population pharmacokinetic model to guide perioperative tacrolimus dosing after lung transplantation. JHLT OPEN 2024; 6:100134. [PMID: 40145052 PMCID: PMC11935331 DOI: 10.1016/j.jhlto.2024.100134] [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 Tacrolimus therapy is standard of care for immunosuppression after lung transplantation. However, tacrolimus exposure variability during the early postoperative period may contribute to poor outcomes in this population. Few studies have examined tacrolimus pharmacokinetics (PK) during this high-risk period. Methods We conducted a retrospective pharmacokinetic study in lung transplant recipients at the University of Pennsylvania who were enrolled in the Lung Transplant Outcomes Group cohort. We used nonlinear mixed-effects regression to derive a population PK model in 270 patients and examined validity in a separate cohort of 114 patients. Covariates were examined with univariate analysis and a multivariable model was developed using forward and backward stepwise selection. The performance of the final model in the validation cohort was examined with calculation of prediction error (PE). Results We developed a 1-compartment base model with a fixed rate absorption constant. Covariates improving model fit were postoperative day, hematocrit, transplant type, CYP3A5 genotype, weight, and exposure to cytochrome p450 enzyme (CYP) inhibitor drugs. The strongest predictor of tacrolimus clearance was postoperative day, with median predicted clearance increasing more than 3-fold over the 14-day study period. In the validation cohort, the final model showed a mean PE of 36.4% (95% confidence interval 30.8%-41.9%) and a median PE of 7.2% (interquartile range -29.3% to 70.53%). Conclusions Tacrolimus clearance is highly dynamic during the early postlung transplant period. Population PK models that include lung-transplant-specific covariates may enable precision dosing algorithms that account for this highly dynamic clearance. Future multicenters studies including a broader set of covariates are warranted.
Collapse
Affiliation(s)
- Todd A. Miano
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Real-world Effectiveness and Safety of Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Rui Feng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen Griffiths
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laurel Kalman
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Oyster
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wei Yang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Real-world Effectiveness and Safety of Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joshua M. Diamond
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D. Christie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marc H. Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, Illinois
- Pharmacometrics Center of Excellence, Midwestern University, Downers Grove, Illinois
| | - Michael G.S. Shashaty
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Axel S, Moneke I, Autenrieth J, Baar W, Loop T. Analysis of Perioperative Factors Leading to Postoperative Pulmonary Complications, Graft Injury and Increased Postoperative Mortality in Lung Transplantation. J Cardiothorac Vasc Anesth 2024; 38:2712-2721. [PMID: 39214800 DOI: 10.1053/j.jvca.2024.08.002] [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: 04/17/2024] [Revised: 07/15/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Postoperative complications such as postoperative pulmonary complications (PPCs) and other organ complications are associated with increased morbidity and mortality after successful lung transplantation and have a detrimental effect on patient recovery. The aim of this study was to investigate perioperative risk factors for in-hospital mortality and postoperative complications with a focus on PPC and graft injury in patients undergoing lung transplantation DESIGN: Single-center retrospective cohort study of 173 patients undergoing lung transplantation SETTING: University Hospital, Medical Center Freiburg. MAIN RESULTS In the stepwise multivariate regression analysis, donor age >60 years (odds ratio [OR], 1.85; 95% confidence interval [CI], 1.27-2.81), intraoperative extracorporeal membrane oxygenation (OR, 2.4; 95% CI, 1.7-3.3), transfusion of >4 red blood cell concentrates (OR, 3.1; 95% CI, 1.82-5.1), mean pulmonary artery pressure of >30 mmHg at the end of surgery (OR, 3.5; 95% CI, 2-6.3), the occurrence of postoperative graft injury (OR, 4.1; 95% CI, 2.8-5.9), PPCs (OR, 2.1; 95% CI, 1.7-2.6), sepsis (OR, 4.5; 95% CI, 2.8-7.3), and Kidney disease Improving Outcome grading system stage 3 acute renal failure (OR, 4.3; 95% CI, 2.4-7.7) were associated with increased in hospital mortality, whereas patients with chronic obstructive pulmonary disease had a lower in-hospital mortality (OR, 1.6; 95% CI, 1.4-1.9). The frequency and number of PPCs correlated with postoperative mortality. CONCLUSIONS Clinical management and risk stratification focusing on the underlying identified factors that could help to improve patient outcomes.
Collapse
Affiliation(s)
- Semmelmann Axel
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Freiburg, Germany.
| | - Isabelle Moneke
- Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Autenrieth
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Freiburg, Germany
| | - Wolfgang Baar
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Freiburg, Germany
| | - Torsten Loop
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Schaenman JM, Weigt SS, Pan M, Lee JJ, Zhou X, Elashoff D, Shino MY, Reynolds JM, Budev M, Shah P, Singer LG, Todd JL, Snyder LD, Palmer S, Belperio J. Alterations in circulating measures of Th2 immune responses pre-lung transplant associates with reduced primary graft dysfunction. J Heart Lung Transplant 2024; 43:1869-1872. [PMID: 39029637 DOI: 10.1016/j.healun.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024] Open
Abstract
Primary graft dysfunction (PGD) is a complication of lung transplantation that continues to cause significant morbidity. The Th2 immune response has been shown to counteract tissue-damaging inflammation. We hypothesized that Th2 cytokines/chemokines in blood would be associated with protection from PGD. Utilizing pretransplant sera from the multicenter clinical trials in organ transplantation study, we evaluated Th2 cytokines/chemokines in 211 patients. Increased concentrations of Th2 cytokines were associated with freedom from PGD, namely IL-4 (odds ratio [OR] 0.66 [95% confidence interval {CI} 0.45-0.99], p = 0.043), IL-9 (OR 0.68 [95% CI 0.49-0.94], p = 0.019), IL-13 (OR 0.73 [95% CI 0.55-0.96], p = 0.023), and IL-6 (OR 0.74 [95% CI 0.56-0.98], p = 0.036). Multivariable regression performed for each cytokine, including clinically relevant covariables, confirmed these associations and additionally demonstrated association with IL-5 (OR 0.57 [95% CI 0.36-0.89], p = 0.014) and IL-10 (OR 0.55 [95% CI 0.32-0.96], p = 0.035). Higher levels of Th2 immune response before lung transplant appear to have a protective effect against PGD, which parallels the Th2 role in resolving inflammation and tissue injury. Pretransplant cytokine assessments could be utilized for recipient risk stratification.
Collapse
Affiliation(s)
- Joanna M Schaenman
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Stephen Samuel Weigt
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Mengtong Pan
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Joshua J Lee
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Xinkai Zhou
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - David Elashoff
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Michael Y Shino
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - John M Reynolds
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Marie Budev
- Department of Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Pali Shah
- Division of Pulmonary Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lianne G Singer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Jamie L Todd
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Laurie D Snyder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Scott Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, North Carolina
| | - John Belperio
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
45
|
Martin AK, Mercier O, Fritz AV, Gelzinis TA, Hoetzenecker K, Lindstedt S, Marczin N, Wilkey BJ, Schecter M, Lyster H, Sanchez M, Walsh J, Morrissey O, Levvey B, Landry C, Saatee S, Kotecha S, Behr J, Kukreja J, Dellgren G, Fessler J, Bottiger B, Wille K, Dave K, Nasir BS, Gomez-De-Antonio D, Cypel M, Reed AK. ISHLT consensus statement on the perioperative use of ECLS in lung transplantation: Part II: Intraoperative considerations. J Heart Lung Transplant 2024:S1053-2498(24)01830-8. [PMID: 39453286 DOI: 10.1016/j.healun.2024.08.027] [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: 08/07/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 10/26/2024] Open
Abstract
The use of extracorporeal life support (ECLS) throughout the perioperative phase of lung transplantation requires nuanced planning and execution by an integrated team of multidisciplinary experts. To date, no multidisciplinary consensus document has examined the perioperative considerations of how to best manage these patients. To address this challenge, this perioperative utilization of ECLS in lung transplantation consensus statement was approved for development by the International Society for Heart and Lung Transplantation Standards and Guidelines Committee. International experts across multiple disciplines, including cardiothoracic surgery, anesthesiology, critical care, pediatric pulmonology, adult pulmonology, pharmacy, psychology, physical therapy, nursing, and perfusion, were selected based on expertise and divided into subgroups examining the preoperative, intraoperative, and postoperative periods. Following a comprehensive literature review, each subgroup developed recommendations to examine via a structured Delphi methodology. Following 2 rounds of Delphi consensus, a total of 39 recommendations regarding intraoperative considerations for ECLS in lung transplantation met consensus criteria. These recommendations focus on the planning, implementation, management, and monitoring of ECLS throughout the entire intraoperative period.
Collapse
Affiliation(s)
- Archer Kilbourne Martin
- Division of Cardiovascular and Thoracic Anesthesiology, Mayo Clinic Florida, Jacksonville, Florida.
| | - Olaf Mercier
- Department of Thoracic Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite' Paris-Saclay, Le Plessis-Robinson, France
| | - Ashley Virginia Fritz
- Division of Cardiovascular and Thoracic Anesthesiology, Mayo Clinic Florida, Jacksonville, Florida
| | - Theresa A Gelzinis
- Division of Cardiovascular and Thoracic Anesthesiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Konrad Hoetzenecker
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Sandra Lindstedt
- Department of Cardiothoracic Surgery and Transplantation, Lund University, Lund, Sweden
| | - Nandor Marczin
- Department of Anaesthesia and Critical Care, Royal Brompton & Harefield Hospitals, Part of Guy's and St Thomas' NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Barbara J Wilkey
- Department of Anesthesiology, University of Colorado, Aurora, Colorado
| | - Marc Schecter
- Division of Pulmonary Medicine, University of Florida, Gainesville, Florida
| | - Haifa Lyster
- Department of Cardiothoracic Transplantation & Mechanical Circulatory Support, Royal Brompton & Harefield Hospitals, Part of Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Melissa Sanchez
- Department of Clinical Health Psychology, Kensington & Chelsea, West Middlesex Hospitals, London, United Kingdom
| | - James Walsh
- Department of Physiotherapy, The Prince Charles Hospital, Brisbane, Australia
| | - Orla Morrissey
- Division of Infectious Disease, Alfred Health and Monash University, Melbourne, Australia
| | - Bronwyn Levvey
- Faculty of Nursing & Health Sciences, The Alfred Hospital, Monah University, Melbourne, Australia
| | - Caroline Landry
- Division of Perfusion Services, Universite' de Montreal, Montreal, Quebec, Canada
| | - Siavosh Saatee
- Division of Cardiovascular and Thoracic Anesthesiology and Critical Care, University of Texas-Southwestern, Dallas, Texas
| | - Sakhee Kotecha
- Lung Transplant Service, Alfred Hospital and Monash University, Melbourne, Australia
| | - Juergen Behr
- Department of Medicine V, German Center for Lung Research, LMU University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jasleen Kukreja
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Francisco, California
| | - Göran Dellgren
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Julien Fessler
- Department of Anesthesiology and Pain Medicine, Hopital Foch, Universite' Versailles-Saint-Quentin-en-Yvelines, Suresnes, France
| | - Brandi Bottiger
- Division of Cardiothoracic Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Keith Wille
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kavita Dave
- Department of Cardiothoracic Transplantation & Mechanical Circulatory Support, Royal Brompton & Harefield Hospitals, Part of Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Basil S Nasir
- Division of Thoracic Surgery, Centre Hospitalier de l'Universite de Montreal (CHUM), Montreal, Quebec, Canada
| | - David Gomez-De-Antonio
- Department of Thoracic Surgery and Lung Transplantation, Hospital Universitario Puerta de Hierro-Majadahonda, Universidad Autonoma de Madria, Madrid, Spain
| | - Marcelo Cypel
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Anna K Reed
- Respiratory & Transplant Medicine, Royal Brompton and Harefield Hospitals, Part of Guy's and St Thomas' NHS Foundation Trust and Imperial College London, London, United Kingdom
| |
Collapse
|
46
|
Serna Villa V, Ren X. Lung Progenitor and Stem Cell Transplantation as a Potential Regenerative Therapy for Lung Diseases. Transplantation 2024; 108:e282-e291. [PMID: 38416452 DOI: 10.1097/tp.0000000000004959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Chronic lung diseases are debilitating illnesses ranking among the top causes of death globally. Currently, clinically available therapeutic options capable of curing chronic lung diseases are limited to lung transplantation, which is hindered by donor organ shortage. This highlights the urgent need for alternative strategies to repair damaged lung tissues. Stem cell transplantation has emerged as a promising avenue for regenerative treatment of the lung, which involves delivery of healthy lung epithelial progenitor cells that subsequently engraft in the injured tissue and further differentiate to reconstitute the functional respiratory epithelium. These transplanted progenitor cells possess the remarkable ability to self-renew, thereby offering the potential for sustained long-term treatment effects. Notably, the transplantation of basal cells, the airway stem cells, holds the promise for rehabilitating airway injuries resulting from environmental factors or genetic conditions such as cystic fibrosis. Similarly, for diseases affecting the alveoli, alveolar type II cells have garnered interest as a viable alveolar stem cell source for restoring the lung parenchyma from genetic or environmentally induced dysfunctions. Expanding upon these advancements, the use of induced pluripotent stem cells to derive lung progenitor cells for transplantation offers advantages such as scalability and patient specificity. In this review, we comprehensively explore the progress made in lung stem cell transplantation, providing insights into the current state of the field and its future prospects.
Collapse
Affiliation(s)
- Vanessa Serna Villa
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | | |
Collapse
|
47
|
Savale L, Benazzo A, Corris P, Keshavjee S, Levine DJ, Mercier O, Davis RD, Granton JT. Transplantation, bridging, and support technologies in pulmonary hypertension. Eur Respir J 2024; 64:2401193. [PMID: 39209471 PMCID: PMC11525343 DOI: 10.1183/13993003.01193-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024]
Abstract
Despite the progress made in medical therapies for treating pulmonary hypertension (PH), a subset of patients remain susceptible to developing a maladaptive right ventricular phenotype. The effective management of end-stage PH presents substantial challenges, necessitating a multidisciplinary approach and early identification of patients prone to acute decompensation. Identifying potential transplant candidates and assessing the feasibility of such a procedure are pivotal tasks that should be undertaken early in the treatment algorithm. Inclusion on the transplant list is contingent upon a comprehensive risk assessment, also considering the specific type of PH and various factors affecting waiting times, all of which should inform the decision-making process. While bilateral lung transplantation is the preferred option, it demands expert intra- and post-operative management to mitigate the heightened risks of pulmonary oedema and primary graft dysfunction in PH patients. Despite the availability of risk assessment tools, the occurrence of acute PH decompensation episodes can be unpredictable, potentially leading to refractory right ventricular failure even with optimal medical intervention, necessitating the use of rescue therapies. Advancements in right ventricular assist techniques and adjustments to graft allocation protocols for the most critically ill patients have significantly enhanced the survival in intensive care, affording the opportunity to endure while awaiting an urgent transplant. Given the breadth of therapeutic options available, specialised centres capable of delivering comprehensive care have become indispensable for optimising patient outcomes. These centres are instrumental in providing holistic support and management tailored to the complex needs of PH patients, ultimately enhancing their chances of a successful transplant and improved long-term prognosis.
Collapse
Affiliation(s)
- Laurent Savale
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, HPPIT, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Alberto Benazzo
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Paul Corris
- Newcastle University and Institute of Transplantation, Freeman Hospital, Newcastle, UK
| | - Shaf Keshavjee
- Toronto Lung Transplant Program, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Deborah Jo Levine
- Division of Pulmonary, Critical Care and Allergy, Stanford University, Palo Alto, CA, USA
| | - Olaf Mercier
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, HPPIT, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Le Kremlin-Bicêtre, France
- Marie Lannelongue Hospital, Dept of Thoracic Surgery and Heart-Lung Transplantation, Le Plessis Robinson, France
| | - R Duane Davis
- Thoracic and Cardiac Surgery, AdventHealth Transplant Institute, Orlando, FL, USA
| | - John T Granton
- Department of Medicine, Division of Respirology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
48
|
Midyat L, Muise ED, Visner GA. Pediatric Lung Transplantation for Pulmonary Vascular Diseases: Recent Advances and Challenges. Clin Chest Med 2024; 45:761-769. [PMID: 39069336 DOI: 10.1016/j.ccm.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Pediatric lung transplantation for pulmonary vascular diseases has seen notable advancements and trends. Medical therapies, surgical options, and bridging techniques like extracorporeal membrane oxygenation and different forms of transplants have expanded treatment possibilities. Current challenges include ensuring patient adherence to post-transplant therapies, addressing complications like primary graft dysfunction and rejection, and conducting further research in less common conditions like pulmonary veno-occlusive disease and pulmonary vein stenosis. In this review article, the authors will explore the advancements, emerging trends, and persistent challenges in pediatric lung transplantation for pulmonary vascular diseases.
Collapse
Affiliation(s)
- Levent Midyat
- Division of Pulmonary Medicine, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, AOB Suite 3300, Pittsburgh, PA 15224, USA.
| | - Eleanor D Muise
- Division of Pulmonary Medicine, NYU Grossman School of Medicine, Hassenfeld Children's Hospital, 240 East 38th Street, 14th Floor, New York, NY 10016, USA
| | - Gary A Visner
- Division of Pulmonary Medicine, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, BCH 3121, Boston, MA 02115, USA
| |
Collapse
|
49
|
Kaihou T, Toyoda T, Cerier E, Yagi Y, Manerikar A, Thomae BL, Kandula V, Bharat A, Kurihara C. The Risk of Pretransplant Blood Transfusion for Primary Graft Dysfunction After Lung Transplant. ANNALS OF THORACIC SURGERY SHORT REPORTS 2024; 2:573-577. [PMID: 39790394 PMCID: PMC11708632 DOI: 10.1016/j.atssr.2024.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 01/12/2025]
Abstract
Background Primary graft dysfunction (PGD) is the leading cause of short- and long-term mortality associated with lung transplantation. The impact of pretransplantation blood transfusions for recipients is not fully elucidated. Methods This is a retrospective review of 206 consecutive lung transplantations performed at a single academic center (Northwestern University Feinberg School of Medicine, Chicago, IL) from January 2018 to July 2022. Data on patient characteristics, pretransplantation laboratory values, transfusion requirements, and intraoperative and postoperative outcomes were collected. Results PGD grade 3 (PGD 3) occurred in 13.2% of the cohort (n = 28). A total of 33 patients received a blood transfusion within 4 weeks, whereas 21 patients received a blood transfusion a week before their lung transplant. Pretransplantation transfusions were strongly associated with a higher incidence of PGD 3 (48.5% vs 6.9%; P < .001). There was no significant difference in 1-year survival between the pretransplantation transfused group and the nontransfused group (77.7% vs 88.0%; P = .478). The 1year survival was reduced in recipients with PGD 3 compared with recipients without PGD 3 (63.5% vs 89.9%; P = .0012). In univariate analysis, pretransplant and intratransplant predictors of PGD 3 included younger age (P < .01), pretransplant extracorporeal membrane oxygenation (ECMO) use (P < .001), higher lung allocation score (P < .001), coronavirus disease 2019 (COVID-19)-related acute respiratory distress syndrome (P < .01), blood transfusion within 4 weeks (P < .001), longer operative time (P < .001), intratransplant blood transfusion (P < .001), and intratransplant venoarterial ECMO use (P < .001). Conclusions Pretransplantation blood transfusions could be associated with a higher rate of PGD. The findings indicated the potential risks of pretransplantation blood transfusions in lung transplant recipients.
Collapse
Affiliation(s)
- Taisuke Kaihou
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Takahide Toyoda
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Emily Cerier
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yuriko Yagi
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Adwaiy Manerikar
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Benjamin Louis Thomae
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Viswajit Kandula
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Chitaru Kurihara
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
50
|
Tsao T, Qiu L, Bharti R, Shemesh A, Hernandez AM, Cleary SJ, Greenland NY, Santos J, Shi R, Bai L, Richardson J, Dilley K, Will M, Tomasevic N, Sputova T, Salles A, Kang J, Zhang D, Hays SR, Kukreja J, Singer JP, Lanier LL, Looney MR, Greenland JR, Calabrese DR. CD94 + natural killer cells potentiate pulmonary ischaemia-reperfusion injury. Eur Respir J 2024; 64:2302171. [PMID: 39190789 DOI: 10.1183/13993003.02171-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/30/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Pulmonary ischaemia-reperfusion injury (IRI) is a major contributor to poor lung transplant outcomes. We recently demonstrated a central role of airway-centred natural killer (NK) cells in mediating IRI; however, there are no existing effective therapies for directly targeting NK cells in humans. METHODS We hypothesised that a depleting anti-CD94 monoclonal antibody (mAb) would provide therapeutic benefit in mouse and human models of IRI based on high levels of KLRD1 (CD94) transcripts in bronchoalveolar lavage samples from lung transplant patients. RESULTS We found that CD94 is highly expressed on mouse and human NK cells, with increased expression during IRI. Anti-mouse and anti-human mAbs against CD94 showed effective NK cell depletion in mouse and human models and blunted lung damage and airway epithelial killing, respectively. In two different allogeneic orthotopic lung transplant mouse models, anti-CD94 treatment during induction reduced early lung injury and chronic inflammation relative to control therapies. Anti-CD94 did not increase donor antigen-presenting cells that could alter long-term graft acceptance. CONCLUSIONS Lung transplant induction regimens incorporating anti-CD94 treatment may safely improve early clinical outcomes.
Collapse
Affiliation(s)
- Tasha Tsao
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- T. Tsao and L. Qiu contributed equally
| | - Longhui Qiu
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- T. Tsao and L. Qiu contributed equally
| | - Reena Bharti
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Avishai Shemesh
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
| | - Alberto M Hernandez
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Simon J Cleary
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Nancy Y Greenland
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jesse Santos
- Department of Surgery, University of California San Francisco - East Bay, Oakland, CA, USA
| | | | - Lu Bai
- Dren Bio, Foster City, CA, USA
| | | | | | | | | | | | | | | | - Dongliang Zhang
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Steven R Hays
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jasleen Kukreja
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan P Singer
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lewis L Lanier
- Parker Institute for Cancer Immunotherapy San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Mark R Looney
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John R Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Daniel R Calabrese
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA, USA
| |
Collapse
|