1
|
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
|
2
|
Brunet-Ratnasingham E, Yellamilli S, Guo R, Mohanty RP, Duong A, Kolaitis NA, Hays SR, Shah RJ, Venado A, Maheshwari JA, Kleinhenz ME, Leard LE, McDyer J, Martinu T, Combes AJ, Calabrese DR, Singer JP, Greenland JR. Persistent and progressive acute lung allograft dysfunction is linked to cell compositional and transcriptional changes in small airways. J Heart Lung Transplant 2025:S1053-2498(25)01842-X. [PMID: 40293382 DOI: 10.1016/j.healun.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Acute lung allograft dysfunction (ALAD) is a clinical syndrome of forced expiratory volume in 1-second (FEV1) decline concerning for chronic lung allograft dysfunction (CLAD) onset. Novel diagnostic tools are needed to identify those with ALAD who will progress to CLAD and to target appropriate therapies. We hypothesized that progressive ALAD would be associated with changes in small airway cell composition and cell-specific transcription. METHODS We prospectively identified recipients with undifferentiated ALAD and controls with stable allograft function for small airway brushing and single-cell RNA sequencing analysis. ALAD outcome group was categorized as (1) control (n = 8), or ALAD with (2) recovered (n = 4), (3) persistent (n = 5), or (4) progressive (n = 3) FEV1 decline. Cell compositional changes, pseudobulk Reactome pathways, and the AI2 score, previously linked to CLAD in airway brush transcriptomes, were assessed as a function of ALAD outcome group. RESULTS Across 68,140 cells, the distribution of cell composition was linked to ALAD outcome group (PERMANOVA, p = 0.004). Worse ALAD outcomes correlated with loss of basal cells, changes in club and ciliated subsets, a loss of macrophages, and expansion of cytotoxic T cells. The AI2 gene score was positively associated with ALAD outcome group, particularly in epithelial cell subsets (p < 0.001). Pathway analysis showed increased interferon signaling and inhibition of cell proliferation in epithelial cells. CONCLUSIONS In this pilot study, persistent and progressive ALAD was associated with changes in bronchiolar cell composition and transcriptional programs. Molecular phenotyping may help identify and characterize individuals with ALAD at increased risk for progression.
Collapse
Affiliation(s)
| | - Shivaram Yellamilli
- Department of Medicine, University of California, San Francisco, San Francisco, California; UCSF CoLabs, San Francisco, California
| | - Ruyin Guo
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Rashmi Prava Mohanty
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Allen Duong
- Toronto Lung Transplant Program, Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Nicholas A Kolaitis
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Steven R Hays
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Rupal J Shah
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Aida Venado
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Julia A Maheshwari
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Mary Ellen Kleinhenz
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Lorriana E Leard
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tereza Martinu
- Toronto Lung Transplant Program, Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Alexis J Combes
- Department of Medicine, University of California, San Francisco, San Francisco, California; UCSF CoLabs, San Francisco, California; Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, San Francisco, California; Medical Service, San Francisco VA Health Care System, San Francisco, California
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, San Francisco, California; Medical Service, San Francisco VA Health Care System, San Francisco, California.
| |
Collapse
|
3
|
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
|
4
|
Snell GI, Ennis SL, Levvey BJ. Inhaled immunosuppressants after lung transplantation -real potential to enhance patient outcomes. Expert Rev Respir Med 2024; 18:925-927. [PMID: 39604127 DOI: 10.1080/17476348.2024.2434152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Affiliation(s)
- Gregory I Snell
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Health, Melbourne, Australia
| | - Samantha L Ennis
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Health, Melbourne, Australia
| | - Bronwyn J Levvey
- Lung Transplant Service, Department of Respiratory Medicine, Alfred Health, Melbourne, Australia
| |
Collapse
|
5
|
Mohanty RP, Moghbeli K, Singer JP, Calabrese DR, Hays SR, Iasella C, Lieber S, Leard LE, Shah RJ, Venado A, Kleinhenz ME, Golden JA, Martinu T, Love C, Ward R, Langelier CR, McDyer J, Greenland JR. Small airway brush gene expression predicts chronic lung allograft dysfunction and mortality. J Heart Lung Transplant 2024; 43:1820-1832. [PMID: 39115489 DOI: 10.1016/j.healun.2024.07.010] [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: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) limits survival following lung transplant, but substantial lung damage occurs before diagnosis by traditional methods. We hypothesized that small airway gene expression patterns could identify CLAD risk before spirometric diagnosis and predict subsequent graft failure. METHODS Candidate genes from 4 rejection-associated transcript sets were assessed for associations with CLAD or graft failure in a derivation cohort of 156 small airway brushes from 45 CLAD cases and 37 time-matched controls with >1-year stable lung function. Candidate genes not associated with CLAD and time to graft failure were excluded, yielding the Airway Inflammation 2 (AI2) gene set. Area under the receiver operating curve (AUC) for CLAD and competing risks of death or graft failure were assessed in an independent validation cohort of 37 CLAD cases and 37 controls. RESULTS Thirty-two candidate genes were associated with CLAD and graft failure, comprising the AI2 score, which clustered into 3 subcomponents. The AI2 score identified CLAD before its onset, in early and late post-CLAD brushes, as well as in the validation cohort (AUC 0.69-0.88). The AI2 score association with CLAD was independent of positive microbiology, CLAD stage, or CLAD subtype. However, transcripts most associated with CLAD evolved over time from CLAD onset. The AI2 score predicted time to graft failure and retransplant-free survival in both cohorts (p ≤ 0.03). CONCLUSIONS This airway inflammation gene score is associated with CLAD development, graft failure, and death. Future studies defining the molecular heterogeneity of airway inflammation could lead to endotype-targeted therapies.
Collapse
Affiliation(s)
- Rashmi Prava Mohanty
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Kaveh Moghbeli
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - Daniel R Calabrese
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Steven R Hays
- Department of Medicine, University of California, San Francisco, California
| | - Carlo Iasella
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sophia Lieber
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lorriana E Leard
- Department of Medicine, University of California, San Francisco, California
| | - Rupal J Shah
- Department of Medicine, University of California, San Francisco, California
| | - Aida Venado
- Department of Medicine, University of California, San Francisco, California
| | - Mary E Kleinhenz
- Department of Medicine, University of California, San Francisco, California
| | - Jeffery A Golden
- Department of Medicine, University of California, San Francisco, California
| | - Tereza Martinu
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Christina Love
- Department of Medicine, University of California, San Francisco, California
| | - Ryan Ward
- Department of Medicine, University of California, San Francisco, California
| | | | - John McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, California; Medical Service, Veterans Affairs Health Care System, San Francisco, California.
| |
Collapse
|
6
|
Sindu D, Bay C, Grief K, Walia R, Tokman S. Clinical utility of plasma percent donor-derived cell-free DNA for lung allograft surveillance: A real-world single-center experience. JHLT OPEN 2024; 6:100141. [PMID: 40145063 PMCID: PMC11935403 DOI: 10.1016/j.jhlto.2024.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background Plasma percent donor-derived cell-free DNA (%dd-cfDNA) has been investigated as a biomarker of allograft injury after lung transplantation. We sought to determine the clinical utility of %dd-cfDNA as a screen for acute cellular rejection (ACR) and respiratory infections (RIs) among lung transplant recipients (LTRs). Methods We retrospectively analyzed %dd-cfDNA results from 95 plasma samples collected from 81 bilateral LTRs >45 days after transplant with a paired transbronchial biopsy performed within 24 hours after sample collection. We calculated sensitivity, specificity, negative predictive value (NPV), and positive predictive value of %dd-cfDNA to detect ACR and RIs and used a generalized estimating equation model to compare %dd-cfDNA between groups. Results A dd-cfDNA threshold of 0.5% had low sensitivity to detect ACR among LTRs (41.67%), as did a 70% increase in %dd-cfDNA (50.00%). The NPV was high (88.89% and 87.50%, respectively) but driven by the low prevalence of ACR (12/95 [12.6%]). The area under the receiver operating characteristic curve (AUC) was 0.499 (95% confidence interval [CI] [0.326-0.672]) and 0.360 (95%CI [0.132-0.588]) for the detection of ACR and ACR grade ≥A2, respectively. The adjusted mean %dd-cfDNA trended higher in LTRs with a definite or possible RI (1.218, 95%CI [0.671-2.212]) than in LTRs without microbial isolation (0.731, 95%CI [0.525-1.017], p = 0.059), but was not significantly different from those with microbial colonization (0.873, 95%CI [0.538-1.415], p = 0.390). The AUC for the detection of allograft dysfunction due to ACR and/or RI was 0.573 (95%CI [0.431-0.716]). Conclusions %dd-cfDNA may have limited utility as a screening tool to detect ACR and/or RI among LTRs.
Collapse
Affiliation(s)
- Devika Sindu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Curt Bay
- A. T. Still University, Phoenix, Arizona
| | - Katherine Grief
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
- Creighton University School of Medicine, Phoenix Health Sciences Campus, Phoenix, Arizona
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
- Creighton University School of Medicine, Phoenix Health Sciences Campus, Phoenix, Arizona
| |
Collapse
|
7
|
Dunn CT, Brandon W, Jacob A, Zhang S, Gao A, Torres F, Lawrence A, Timofte I, Bollineni S, Mohanka M, Wait M, Peltz M, Heid C, Huffman L, Ring S, Murala J, Keshavamurthy S, Weston AJ, Kaza V. Anti-Thymocyte Globulin as a Therapy for Chronic Lung Allograft Dysfunction: A Single-Center Experience. Clin Transplant 2024; 38:e15461. [PMID: 39365102 DOI: 10.1111/ctr.15461] [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: 10/25/2023] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Anti-thymocyte globulin (ATG) is a polyclonal antibody formulation which has been used as a second-line therapy for chronic lung allograft dysfunction (CLAD). Limited data exist evaluating its efficacy; however, several single-center retrospective studies have variably demonstrated either improvement or stabilization of spirometry parameters after administration of ATG. ATG has been in use at UT Southwestern for treatment of CLAD since at least 2010; here, we seek to evaluate the effectiveness of this intervention at our center. METHODS: A retrospective chart review was conducted of a total of 136 patients who underwent lung transplantation at UT Southwestern Medical Center between 2010 and 2022. Of these, 72 patients had received ATG specifically for treatment of CLAD, and the remaining 64 had never received ATG. Two separate analyses were performed: in the first, among those who received ATG for CLAD, spirometry data from the 6 months preceding and following ATG administration were reviewed and rates of change in FEV1 were calculated for each time period. Descriptive statistics were performed to summarize the baseline clinical characteristics and outcomes after ATG, with patients classified as having either a full response (positive rate of change in FEV1) or partial response (>20% attenuation in rate of FEV1 decline) to ATG. In the second analysis, survival was described among those who received ATG for CLAD and comparison was provided between propensity-score matched cohorts from the ATG and non-ATG groups. RESULTS Of the 63 patients who received ATG for treatment of CLAD (and had adequate spirometry measurements available to trend FEV1), 49 (77.8%) had at least a partial response to therapy; 8 (12.7%) experienced an overall improvement in FEV1. Response to ATG was found to be associated with a more rapid rate of pre-ATG decline in FEV1; no other baseline parameters were found to be predictive of a response to ATG. Median post-CLAD graft survival was 31.7 months among those who received ATG, and only baseline absolute neutrophil count was found to be associated with worse post-CLAD graft survival among this group. CONCLUSION Anti-thymocyte globulin therapy, when given for CLAD, was associated with at least a modest attenuation in rate of FEV1 decline in most patients but only rarely preceded an absolute improvement in FEV1. Further study is warranted to better define the role for ATG in treatment of CLAD, a challenging disease state with limited therapeutics available.
Collapse
Affiliation(s)
- Colin T Dunn
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - William Brandon
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Alan Jacob
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Song Zhang
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ang Gao
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Fernando Torres
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Adrian Lawrence
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Irina Timofte
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Srinivas Bollineni
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Manish Mohanka
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Michael Wait
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Heid
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Lynn Huffman
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Steve Ring
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - John Murala
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Suresh Keshavamurthy
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Alex Jaye Weston
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Vaidehi Kaza
- Division of Pulmonary and Critical Care Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
8
|
January SE, Hubbard J, Fester KA, Dubrawka CA, Vazquez Guillamet R, Kulkarni HS, Hachem RR. Impact of Angiotensin Blockade on Development of Chronic Lung Allograft Dysfunction. J Pharm Pract 2024; 37:1170-1174. [PMID: 37923307 PMCID: PMC11994155 DOI: 10.1177/08971900231213699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Background: The renin-angiotensin-aldosterone system (RAAS) is responsible for a multitude of physiological functions, including immunological effects such as promotion of TGF-β and upregulation of IL-6 and IL-8 which are also implicated in the development of chronic lung allograft dysfunction (CLAD). Blockade of the RAAS pathway in pre-clinical models has demonstrated a decrease in these cytokines and pulmonary neutrophil recruitment. Objective: This study sought to evaluate whether use of RAAS inhibitor (RAASi) in lung transplant recipients impacted CLAD-free survival. Methods: In this retrospective, single-center study, 35 lung transplant recipients who received a RAASi post-transplant were compared to 70 lung transplant recipients not exposed to a RAASi and were followed for up to 5 years post-transplant. Results: The incidence of CLAD did not differ based on RAASi treatment (34.3% in RAASi vs 38.6%, P-value .668). This was confirmed with a multivariable Cox proportional hazards model with RAASi initiation as a time-varying covariate (RAASi hazard ratio of 1.01, P-value .986). Incidence of hyperkalemia and acute kidney injury were low in the RAASi group. Conclusions: This study demonstrated no association between post-transplant RAASi use and decreased risk of CLAD development. RAASi were also well tolerated in this patient population.
Collapse
Affiliation(s)
- Spenser E January
- Department of Pharmacy, Barnes-Jewish Hospital, Saint Louis, MO, USA
| | - Julie Hubbard
- Department of Pharmacy, Barnes-Jewish Hospital, Saint Louis, MO, USA
| | - Keith A Fester
- Department of Pharmacy, Barnes-Jewish Hospital, Saint Louis, MO, USA
| | - Casey A Dubrawka
- Department of Pharmacy, Barnes-Jewish Hospital, Saint Louis, MO, USA
| | - Rodrigo Vazquez Guillamet
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ramsey R Hachem
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
9
|
Renaud-Picard B, Berra G, Hwang D, Huszti E, Miyamoto E, Berry GJ, Pal P, Juvet S, Keshavjee S, Martinu T. Spectrum of chronic lung allograft dysfunction pathology in human lung transplantation. J Heart Lung Transplant 2024; 43:1701-1715. [PMID: 38663465 DOI: 10.1016/j.healun.2024.04.002] [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: 08/23/2023] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Long-term survival after lung transplantation (LTx) remains limited by chronic lung allograft dysfunction (CLAD), which includes 2 main phenotypes: bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS), with possible overlap. We aimed to detail and quantify pathological features of these CLAD sub-types. METHODS Peripheral and central paraffin-embedded explanted lung samples were obtained from 20 consecutive patients undergoing a second LTx for CLAD, from 3 lobes. Thirteen lung samples, collected from non-transplant lobectomies or donor lungs, were used as controls. Blinded semi-quantitative grading was performed to assess airway fibrotic changes, parenchymal and pleural fibrosis, and epithelial and vascular abnormalities. RESULTS CLAD lung samples had higher scores for all airway- and lung-related parameters compared to controls. There was a notable overlap in histologic scores between BOS and RAS, with a wide range of scores in both conditions. Parenchymal and vascular fibrosis scores were significantly higher in RAS compared to BOS (p = 0.003 for both). We observed a significant positive correlation between the degree of inflammation around each airway, the severity of epithelial changes, and airway fibrosis. Immunofluorescence staining demonstrated a trend toward a lower frequency of club cells in CLAD and a higher frequency of apoptotic club cells in BOS samples (p = 0.01). CONCLUSIONS CLAD is a spectrum of airway, parenchymal, and pleural fibrosis, as well as epithelial, vascular, and inflammatory pathologic changes, where BOS and RAS overlap significantly. Our semi-quantitative grading score showed a generally high inter-reader reliability and may be useful for future CLAD histologic assessments.
Collapse
Affiliation(s)
- Benjamin Renaud-Picard
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; INSERM Unité Mixte de Recherche 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Gregory Berra
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Service de Pneumologie, Département de Médecine, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - David Hwang
- Department of Pathology, Sunnybrook Hospital, Toronto, Ontario, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Ei Miyamoto
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Prodipto Pal
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada; Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Milesi J, Gras D, Chanez P, Coiffard B. Airway epithelium in lung transplantation: a potential actor for post-transplant complications? Eur Respir Rev 2024; 33:240093. [PMID: 39603662 PMCID: PMC11600126 DOI: 10.1183/16000617.0093-2024] [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: 04/26/2024] [Accepted: 09/20/2024] [Indexed: 11/29/2024] Open
Abstract
Lung transplantation, a critical intervention for end-stage lung diseases, is frequently challenged by post-transplant complications. Indeed, primary graft dysfunction, anastomotic complications, infections and acute and chronic rejections pose significant hurdles in lung transplantation. While evidence regarding the role of airway epithelium after lung transplantation is still emerging, its importance is becoming increasingly recognised. This review looks at the complex involvement of airway epithelium in various post-transplant complications, while emphasising the utility of airway epithelial culture as a research model. In summary, by elucidating the involvement of airway epithelium in each post-transplant complication and explaining these intricate processes, the review aims to guide specific future research efforts and therapeutic strategies aimed at improving lung transplant outcomes and enhancing patient care.
Collapse
Affiliation(s)
- Jules Milesi
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Delphine Gras
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pascal Chanez
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Benjamin Coiffard
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
11
|
Barten MJ, Fisher AJ, Hertig A. The use of extracorporeal photopheresis in solid organ transplantation-current status and future directions. Am J Transplant 2024; 24:1731-1741. [PMID: 38490642 DOI: 10.1016/j.ajt.2024.03.012] [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: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/10/2024] [Indexed: 03/17/2024]
Abstract
Prevention and management of allograft rejection urgently require more effective therapeutic solutions. Current immunosuppressive therapies used in solid organ transplantation, while effective in reducing the risk of acute rejection, are associated with substantial adverse effects. There is, therefore, a need for agents that can provide immunomodulation, supporting graft tolerance, while minimizing the need for immunosuppression. Extracorporeal photopheresis (ECP) is an immunomodulatory therapy currently recommended in international guidelines as an adjunctive treatment for the prevention and management of organ rejection in heart and lung transplantations. This article reviews clinical experience and ongoing research with ECP for organ rejection in heart and lung transplantations, as well as emerging findings in kidney and liver transplantation. ECP, due to its immunomodulatory and immunosuppressive-sparing effects, offers a potential therapeutic option in these settings, particularly in high-risk patients with comorbidities, infectious complications, or malignancies.
Collapse
Affiliation(s)
- Markus J Barten
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg; University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Andrew J Fisher
- Transplant and Regnerative Medicine Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexandre Hertig
- Department of Nephrology, University Versailles Saint Quentin, Foch Hospital, Suresnes, France
| |
Collapse
|
12
|
Yeo HJ, Kang J, Kim YH, Cho WH. Periostin in Bronchiolitis Obliterans Syndrome after Lung Transplant. Int J Mol Sci 2024; 25:10423. [PMID: 39408746 PMCID: PMC11477235 DOI: 10.3390/ijms251910423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The utility of measuring serum periostin levels for predicting the occurrence of bronchiolitis obliterans syndrome (BOS) after lung transplantation remains underexplored. We analyzed differentially expressed genes (DEGs) between initially transplanted lung tissue and lung tissue with BOS from four patients. Periostin levels were assessed in 97 patients who had undergone lung transplantation 1 year post-transplantation and at the onset of BOS. The association between periostin levels and BOS, as well as their correlation with the decline in forced expiratory volume in one second (FEV1), was evaluated. Periostin levels in the BOS group were significantly higher than those in the control group (p < 0.001) and the stable group (p < 0.001). Periostin levels at the onset of BOS were significantly higher than those 1 year post-transplantation in the BOS group (p < 0.001). The serum periostin levels at the time of BOS diagnosis showed a positive correlation with the reduction in FEV1 (%) (r = 0.745, p < 0.001). The increase in the serum periostin levels at the time of BOS diagnosis compared with those 1 year post-transplantation was positively correlated with reduction in FEV1 (%) (r = 0.753, p < 0.001). Thus, serum periostin levels may serve as biomarkers for predicting a decline in lung function in patients with BOS after lung transplantation.
Collapse
Affiliation(s)
- Hye Ju Yeo
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
- Department of Internal Medicine, School of Medicine, Pusan National University, Busan 43241, Republic of Korea
| | - Junho Kang
- Department of research, Keimyung University Donsan Medical Center, Daegu 42601, Republic of Korea;
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woo Hyun Cho
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Internal Medicine, Transplant Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
- Department of Internal Medicine, School of Medicine, Pusan National University, Busan 43241, Republic of Korea
| |
Collapse
|
13
|
Aburahma K, de Manna ND, Kuehn C, Salman J, Greer M, Ius F. Pushing the Survival Bar Higher: Two Decades of Innovation in Lung Transplantation. J Clin Med 2024; 13:5516. [PMID: 39337005 PMCID: PMC11432129 DOI: 10.3390/jcm13185516] [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: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Survival after lung transplantation has significantly improved during the last two decades. The refinement of the already existing extracorporeal life support (ECLS) systems, such as extracorporeal membrane oxygenation (ECMO), and the introduction of new techniques for donor lung optimization, such as ex vivo lung perfusion (EVLP), have allowed the extension of transplant indication to patients with end-stage lung failure after acute respiratory distress syndrome (ARDS) and the expansion of the donor organ pool, due to the better evaluation and optimization of extended-criteria donor (ECD) lungs and of donors after circulatory death (DCD). The close monitoring of anti-HLA donor-specific antibodies (DSAs) has allowed the early recognition of pulmonary antibody-mediated rejection (AMR), which requires a completely different treatment and has a worse prognosis than acute cellular rejection (ACR). As such, the standardization of patient selection and post-transplant management has significantly contributed to this positive trend, especially at high-volume centers. This review focuses on lung transplantation after ARDS, on the role of EVLP in lung donor expansion, on ECMO as a principal cardiopulmonary support system in lung transplantation, and on the diagnosis and therapy of pulmonary AMR.
Collapse
Affiliation(s)
- Khalil Aburahma
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Nunzio Davide de Manna
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Kuehn
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| | - Jawad Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| | - Mark Greer
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, 30625 Hannover, Germany
| | - Fabio Ius
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| |
Collapse
|
14
|
Matheson AM, Tanimoto A, Woods JC. Imaging in Pediatric Lung Disease: Current Practice and Future Directions. Clin Chest Med 2024; 45:569-585. [PMID: 39069322 DOI: 10.1016/j.ccm.2024.03.005] [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: 07/30/2024]
Abstract
Pediatric diseases present differently from adult diseases and imaging forms a cornerstone of modern pediatric care through differential diagnosis, disease monitoring, and measuring response to therapy. Imaging is especially well suited to providing novel insights into the underlying mechanisms driving disease through structural and functional imaging. In this review, we describe key imaging findings in standard-of-care and state-of-the-art techniques in pediatric and adult diseases with origins in childhood. We examine applications in small airways disease, large airway disease, diseases of maturity, interstitial lung disease, neuromuscular disease, congenital disease, and pulmonary infection.
Collapse
Affiliation(s)
- Alexander M Matheson
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Aki Tanimoto
- Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Jason C Woods
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Cincinnati Bronchopulmonary Dysplasia Center, Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
15
|
Napoli C, Benincasa G, Fiorelli A, Strozziero MG, Costa D, Russo F, Grimaldi V, Hoetzenecker K. Lung transplantation: Current insights and outcomes. Transpl Immunol 2024; 85:102073. [PMID: 38889844 DOI: 10.1016/j.trim.2024.102073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Until now, the ability to predict or retard immune-mediated rejection events after lung transplantation is still limited due to the lack of specific biomarkers. The pressing need remains to early diagnose or predict the onset of chronic lung allograft dysfunction (CLAD) and its differential phenotypes that is the leading cause of death. Omics technologies (mainly genomics, epigenomics, and transcriptomics) combined with advanced bioinformatic platforms are clarifying the key immune-related molecular routes that trigger early and late events of lung allograft rejection supporting the biomarker discovery. The most promising biomarkers came from genomics. Both unregistered and NIH-registered clinical trials demonstrated that the increased percentage of donor-derived cell-free DNA in both plasma and bronchoalveolar lavage fluid showed a good diagnostic performance for clinically silent acute rejection events and CLAD differential phenotypes. A further success arose from transcriptomics that led to development of Molecular Microscope® Diagnostic System (MMDx) to interpret the relationship between molecular signatures of lung biopsies and rejection events. Other immune-related biomarkers of rejection events may be exosomes, telomer length, DNA methylation, and histone-mediated neutrophil extracellular traps (NETs) but none of them entered in registered clinical trials. Here, we discuss novel and existing technologies for revealing new immune-mediated mechanisms underlying acute and chronic rejection events, with a particular focus on emerging biomarkers for improving precision medicine of lung transplantation field.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Alfonso Fiorelli
- Thoracic Surgery Unit, Department of Translation Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | | | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | | | - Vincenzo Grimaldi
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli,", Naples, Italy
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
16
|
Wannes Daou A, Wallace C, Barker M, Ambrosino T, Towe C, Morales DLS, Wikenheiser-Brokamp KA, Hayes D, Burg G. Flexible bronchoscopy in pediatric lung transplantation. Pediatr Transplant 2024; 28:e14757. [PMID: 38695266 DOI: 10.1111/petr.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/09/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024]
Abstract
Pediatric lung transplantation represents a treatment option for children with advanced lung disease or pulmonary vascular disorders who are deemed an appropriate candidate. Pediatric flexible bronchoscopy is an important and evolving field that is highly relevant in the pediatric lung transplant population. It is thus important to advance our knowledge to better understand how care for children after lung transplant can be maximally optimized using pediatric bronchoscopy. Our goals are to continually improve procedural skills when performing bronchoscopy and to decrease the complication rate while acquiring adequate samples for diagnostic evaluation. Attainment of these goals is critical since allograft assessment by bronchoscopic biopsy is required for histological diagnosis of acute cellular rejection and is an important contributor to establishing chronic lung allograft dysfunction, a common complication after lung transplant. Flexible bronchoscopy with bronchoalveolar lavage and transbronchial lung biopsy plays a key role in lung transplant graft assessment. In this article, we discuss the application of bronchoscopy in pediatric lung transplant evaluation including historical approaches, our experience, and future directions not only in bronchoscopy but also in the evolving pediatric lung transplantation field. Pediatric flexible bronchoscopy has become a vital modality for diagnosing lung transplant complications in children as well as assessing therapeutic responses. Herein, we review the value of flexible bronchoscopy in the management of children after lung transplant and discuss the application of novel techniques to improve care for this complex pediatric patient population and we provide a brief update about new diagnostic techniques applied in the growing lung transplantation field.
Collapse
Affiliation(s)
- Antoinette Wannes Daou
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carolyn Wallace
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mitzi Barker
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Teresa Ambrosino
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Christopher Towe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - David L S Morales
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Don Hayes
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Transplant Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Gregory Burg
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Schmucki K, Gaisl T, Hofmann P, Hage R, Steinack C, Fehr TH, Ulrich S, Schuurmans MM. mTOR inhibitors after lung transplantation: a real-life experience. J Thorac Dis 2024; 16:3007-3018. [PMID: 38883630 PMCID: PMC11170424 DOI: 10.21037/jtd-23-1623] [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: 11/01/2023] [Accepted: 03/22/2024] [Indexed: 06/18/2024]
Abstract
Background The mammalian target of rapamycin (mTOR) inhibitors in combination with calcineurin inhibitors (CNIs), antimetabolites and corticosteroids for immunosuppression after lung transplantation (TPL) have gained importance in patients with chronic kidney disease (CKD). The goal of this study was to characterize lung transplant recipients (LTR) treated with mTOR inhibitors, with a special focus on kidney function. Methods LTR transplanted at the University Hospital Zurich between December 1992 and April 2022 were analyzed. Demographics, estimated glomerular filtration rate (eGFR) before and after mTOR initiation, TPL circumstances, immunosuppressive regimens, and allograft function were recorded. We used linear regression to calculate the Mitch curves and a linear mixed-effects model to compare the eGFR. Results Of all LTR, 70/593 (12%) received mTOR inhibitors. Intolerance or adverse events of antimetabolites were the most common indications for mTOR inhibitor introduction. Discontinuation in 34/70 (49%) was often related to planned or urgent surgery to prevent impaired wound healing. The majority of patients had a preserved baseline eGFR at mTOR inhibitor introduction with CKD Kidney Disease Improving Global Outcomes (KDIGO) stage G1 or 2. The mean annual eGFR decline changed significantly from -16.19 mL/min/1.73 m2/year [95% confidence interval (CI): -22.27 to -10.11] 12 months before to -6.16 mL/min/1.73 m2/year (95% CI: -13.37 to 1.05) 12 months after mTOR initiation (P=0.009) showing better outcomes with earlier mTOR inhibitor initiation after lung TPL. Conclusions This retrospective study suggests stabilization of kidney function after mTOR inhibitor initiation in LTR documented by a slower eGFR decline after mTOR inhibitor introduction with better outcomes early after lung TPL. Intolerance or adverse events of antimetabolites are important indications for the introduction of mTOR inhibitors. A relatively high discontinuation rate (49%) can be explained by planned discontinuation of mTOR inhibitors prior to surgery to avoid impaired wound healing.
Collapse
Affiliation(s)
- Katja Schmucki
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
| | - Thomas Gaisl
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick Hofmann
- Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
| | - René Hage
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Carolin Steinack
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Hans Fehr
- Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Silvia Ulrich
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Macé M Schuurmans
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Fisher AJ, White M, Goudie N, Kershaw A, Phillipson J, Bardgett M, Lally J, Bevin-Nicholls A, Chadwick T, Bryant A, Russell S, Smith H, Frisby L, Errington R, Carby M, Thompson R, Santhanakrishnan K, Parmar J, Lordan JL, Vale L, Hancock H, Exley C, Gennery AR, Wason JM. Extracorporeal photopheresis (ECP) in the treatment of chronic lung allograft dysfunction (CLAD): a prospective, multicentre, open-label, randomised controlled trial studying the addition of ECP to standard care in the treatment of bilateral lung transplant patients with CLAD (E-CLAD UK). BMJ Open Respir Res 2024; 11:e001995. [PMID: 38724453 PMCID: PMC11086459 DOI: 10.1136/bmjresp-2023-001995] [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: 08/02/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Long-term survival after lung transplantation is limited compared with other organ transplants. The main cause is development of progressive immune-mediated damage to the lung allograft. This damage, which can develop via multiple immune pathways, is captured under the umbrella term chronic lung allograft dysfunction (CLAD). Despite the availability of powerful immunosuppressive drugs, there are presently no treatments proven to reverse or reliably halt the loss of lung function caused by CLAD. The aim of the E-CLAD UK trial is to determine whether the addition of immunomodulatory therapy, in the form of extracorporeal photopheresis (ECP), to standard care is more efficacious at stabilising lung function in CLAD compared with standard care alone. METHODS AND ANALYSIS E-CLAD UK is a Phase II clinical trial of an investigational medicinal product (Methoxsalen) delivered to a buffy coat prepared via an enclosed ECP circuit. Target recruitment is 90 bilateral lung transplant patients identified as having CLAD and being treated at one of the five UK adult lung transplant centres. Participants will be randomised 1:1 to intervention plus standard of care, or standard of care alone. Intervention will comprise nine ECP cycles spread over 20 weeks, each course involving two treatments of ECP on consecutive days. All participants will be followed up for a period of 24 weeks.The primary outcome is lung function stabilisation derived from change in forced expiratory volume in one second and forced vital capacity at 12 and 24 weeks compared with baseline at study entry. Other parameters include change in exercise capacity, health-related quality of life and safety. A mechanistic study will seek to identify molecular or cellular markers linked to treatment response and qualitative interviews will explore patient experiences of CLAD and the ECP treatment.A patient and public advisory group is integral to the trial from design to implementation, developing material to support the consent process and interview materials. ETHICS AND DISSEMINATION The East Midlands-Derby Research Ethics Committee has provided ethical approval (REC 22/EM/0218). Dissemination will be via publications, patient-friendly summaries and presentation at scientific meetings. TRIAL REGISTRATION NUMBER EudraCT number 2022-002659-20; ISRCTN 10615985.
Collapse
Affiliation(s)
- Andrew J Fisher
- Faculty of Medical Sciences, Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michael White
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Nicola Goudie
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Anneka Kershaw
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Julia Phillipson
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Michelle Bardgett
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Joanne Lally
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Alex Bevin-Nicholls
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Thomas Chadwick
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Andrew Bryant
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Sian Russell
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Hesther Smith
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Laura Frisby
- Joint Research Office, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rebecca Errington
- Joint Research Office, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Martin Carby
- Royal Brompton and Harefield Hospitals, London, UK
| | - Richard Thompson
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Jasvir Parmar
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - James L Lordan
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Luke Vale
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Helen Hancock
- Newcastle Clinical Trials Unit, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Catherine Exley
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| | - Andrew R Gennery
- Faculty of Medical Sciences, Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - James Ms Wason
- Newcastle University Population Health Sciences Institute, Newcastle upon Tyne, UK
| |
Collapse
|
19
|
Keller MB, Tian X, Jang MK, Meda R, Charya A, Ozisik D, Berry GJ, Marboe CC, Kong H, Ponor IL, Aryal S, Orens JB, Shah PD, Nathan SD, Agbor-Enoh S. Organizing pneumonia is associated with molecular allograft injury and the development of antibody-mediated rejection. J Heart Lung Transplant 2024; 43:563-570. [PMID: 37972825 DOI: 10.1016/j.healun.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/28/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The association between organizing pneumonia (OP) after lung transplantation with the development of acute rejection (AR) remains undefined. In addition, molecular allograft injury, as measured by donor-derived cell-free DNA (dd-cfDNA), during episodes of OP and its relationship to episodes of AR, chronic lung allograft dysfunction (CLAD), or death is unknown. METHODS This multicenter, prospective cohort study collected serial plasma samples from 188 lung transplant recipients for dd-cfDNA at the time of bronchoscopy with biopsy. Multivariable Cox regression was used to analyze the association between OP with the development of AR (antibody-mediated rejection (AMR) and acute cellular rejection (ACR)), CLAD, and death. Multivariable models were performed to test the association of dd-cfDNA at OP with the risk of AR, CLAD, or death. RESULTS In multivariable analysis, OP was associated with increased risk of AMR (hazard ratio (HR) = 2.26, 95% confidence interval (CI) 1.04-4.92, p = 0.040) but not ACR (HR = 1.29, 95% CI: 0.66-2.5, p = 0.45) or the composite outcome of CLAD or death (HR = 0.88, 95% CI, 0.47-1.65, p = 0.69). Median levels of dd-cfDNA were higher in OP compared to stable controls (1.33% vs 0.43%, p = 0.0006). Multivariable analysis demonstrated that levels of dd-cfDNA at diagnosis of OP were associated with increased risk of both AMR (HR = 1.29, 95% CI 1.03-1.62, p = 0.030) and death (HR = 1.16, 95% CI, 1.02-1.31, p = 0.026). CONCLUSIONS OP is independently associated with an increased risk of AMR but not CLAD or death. The degree of molecular allograft injury at the diagnosis of OP may further predict the risk of AMR and death.
Collapse
Affiliation(s)
- Michael B Keller
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Rohan Meda
- Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ananth Charya
- University of Maryland Medical Center, Baltimore, Maryland
| | - Deniz Ozisik
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Gerald J Berry
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Stanford University School of Medicine, Stanford, California
| | - Charles C Marboe
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York
| | - Hyesik Kong
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ileana L Ponor
- Department of Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Jonathan B Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Pali D Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Steven D Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Fairfax, Virginia
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Laboratory of Applied Precision Omics (APO), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; Division of Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
20
|
Tissot A, Durand E, Goronflot T, Coiffard B, Renaud-Picard B, Roux A, Demant X, Mornex JF, Falque L, Salpin M, Le Pavec J, Villeneuve T, Boussaud V, Knoop C, Magnan A, Lair D, Berthelot L, Danger R, Brouard S. Blood MMP-9 measured at 2 years after lung transplantation as a prognostic biomarker of chronic lung allograft dysfunction. Respir Res 2024; 25:88. [PMID: 38336710 PMCID: PMC10858575 DOI: 10.1186/s12931-024-02707-3] [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: 11/29/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Long-term outcomes of lung transplantation (LTx) remain hampered by chronic lung allograft dysfunction (CLAD). Matrix metalloproteinase 9 (MMP-9) is a secretory endopeptidase identified as a key mediator in fibrosis processes associated with CLAD. The objective of this study was to investigate whether plasma MMP9 levels may be prognostic of CLAD development. METHODS Participants were selected from the Cohort in Lung Transplantation (COLT) for which a biocollection was associated. We considered two time points, year 1 (Y1) and year 2 (Y2) post-transplantation, for plasma MMP-9 measurements. We analysed stable recipients at those time points, comparing those who would develop a CLAD within the 2 years following the measurement to those who would remain stable 2 years after. RESULTS MMP-9 levels at Y1 were not significantly different between the CLAD and stable groups (230 ng/ml vs. 160 ng/ml, p = 0.4). For the Y2 analysis, 129 recipients were included, of whom 50 developed CLAD within 2 years and 79 remained stable within 2 years. MMP-9 plasma median concentrations were higher in recipients who then developed CLAD than in the stable group (230 ng/ml vs. 118 ng/ml, p = 0.003). In the multivariate analysis, the Y2 MMP-9 level was independently associated with CLAD, with an average increase of 150 ng/ml (95% CI [0-253], p = 0.05) compared to that in the stable group. The Y2 ROC curve revealed a discriminating capacity of blood MMP-9 with an area under the curve of 66%. CONCLUSION Plasmatic MMP-9 levels measured 2 years after lung transplantation have prognostic value for CLAD.
Collapse
Affiliation(s)
- Adrien Tissot
- CHU Nantes, INSERM, Service de Pneumologie, l'institut du thorax, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes Université, 44093, Nantes, France.
| | - Eugénie Durand
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Thomas Goronflot
- CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des données, INSERM, CIC 1413, Nantes Université, Nantes, France
| | - Benjamin Coiffard
- Department of Respiratory Medicine and Lung Transplantation, APHM, Hôpital Nord, Aix Marseille Univ, Marseille, France
| | - Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, Inserm UMR 1260, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - Xavier Demant
- Service de Pneumologie, Centre Hospitalier Universitaire de Bordeaux, Pessac, France
| | - Jean-François Mornex
- Université Lyon 1, PSL, EPHE, INRAE, IVPC, Hospices Civils de Lyon, groupement hospitalier est, service de pneumologie, Orphalung, RESPIFIL, Université de Lyon, Lyon, France
| | - Loïc Falque
- Service Hospitalier Universitaire de Pneumologie et Physiologie, CHU Grenoble Alpes, Pôle Thorax et Vaisseaux, Grenoble, France
| | - Mathilde Salpin
- APHP Nord-Université Paris Cité, Hôpital Bichat, Service de Pneumologie B et Transplantation Pulmonaire, PHERE UMRS 1152, Université Paris Cité, Paris, France
| | - Jérôme Le Pavec
- Service de Pneumologie et Transplantation Pulmonaire, Groupe hospitalier Marie-Lannelongue -Saint Joseph, Le Plessis-Robinson, Université Paris-Saclay, UMR_S 999, INSERM, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Thomas Villeneuve
- CHU Toulouse, Service de Pneumologie, Université Toulouse III-Paul Sabatier, Toulouse, France
| | | | | | - Antoine Magnan
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department Hôpital Foch, Suresnes, INRAe UMR 0892, Paris Transplant Group, Université de Versailles Saint Quentin Paris-Saclay, Paris, France
| | - David Lair
- CHU Nantes, Nantes Université, Institut du Thorax, Lung O2, Nantes, France
| | - Laureline Berthelot
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Richard Danger
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| | - Sophie Brouard
- CHU de Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie (CR2TI), Nantes Université, Nantes, France
| |
Collapse
|
21
|
Watanabe T, Juvet SC, Berra G, Havlin J, Zhong W, Boonstra K, Daigneault T, Horie M, Konoeda C, Teskey G, Guan Z, Hwang DM, Liu M, Keshavjee S, Martinu T. Donor IL-17 receptor A regulates LPS-potentiated acute and chronic murine lung allograft rejection. JCI Insight 2023; 8:e158002. [PMID: 37937643 PMCID: PMC10721268 DOI: 10.1172/jci.insight.158002] [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: 01/12/2022] [Accepted: 09/15/2023] [Indexed: 11/09/2023] Open
Abstract
Chronic lung allograft dysfunction (CLAD) is a major complication after lung transplantation that results from a complex interplay of innate inflammatory and alloimmune factors, culminating in parenchymal and/or obliterative airway fibrosis. Excessive IL-17A signaling and chronic inflammation have been recognized as key factors in these pathological processes. Herein, we developed a model of repeated airway inflammation in mouse minor alloantigen-mismatched single-lung transplantation. Repeated intratracheal LPS instillations augmented pulmonary IL-17A expression. LPS also increased acute rejection, airway epithelial damage, and obliterative airway fibrosis, similar to human explanted lung allografts with antecedent episodes of airway infection. We then investigated the role of donor and recipient IL-17 receptor A (IL-17RA) in this context. Donor IL-17RA deficiency significantly attenuated acute rejection and CLAD features, whereas recipient IL-17RA deficiency only slightly reduced airway obliteration in LPS allografts. IL-17RA immunofluorescence positive staining was greater in human CLAD lungs compared with control human lung specimens, with localization to fibroblasts and myofibroblasts, which was also seen in mouse LPS allografts. Taken together, repeated airway inflammation after lung transplantation caused local airway epithelial damage, with persistent elevation of IL-17A and IL-17RA expression and particular involvement of IL-17RA on donor structural cells in development of fibrosis.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Stephen C. Juvet
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory Berra
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Jan Havlin
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wenshan Zhong
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Kristen Boonstra
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Tina Daigneault
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | | | - Chihiro Konoeda
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
| | - Grace Teskey
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Zehong Guan
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
| | - David M. Hwang
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Center, University Health Network, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Trindade AJ. Can We See Clearly Now? Computed Tomography for Bronchiolitis Obliterans Syndrome Prognostication. Transplantation 2023; 107:e281-e282. [PMID: 37443411 PMCID: PMC10593147 DOI: 10.1097/tp.0000000000004727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Affiliation(s)
- Anil J. Trindade
- Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt Transplant Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Kolaitis NA. Lung Transplantation for Pulmonary Arterial Hypertension. Chest 2023; 164:992-1006. [PMID: 37150504 DOI: 10.1016/j.chest.2023.04.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/09/2023] Open
Abstract
TOPIC IMPORTANCE Even though patients with pulmonary arterial hypertension have multiple therapeutic options, the disease can be refractory despite appropriate management. In patients with end-stage pulmonary arterial hypertension, lung transplantation has the potential both to extend survival and improve health-related quality of life. Pulmonary arterial hypertension is the only major diagnostic indication for transplantation that is not a parenchymal pulmonary process, and thus the care of these patients is unique. REVIEW FINDINGS This review focuses on the complexities of lung transplantation for patients with pulmonary arterial hypertension, presents the updated referral and listing criteria, and discusses the inequities in the organ allocation process that impact this disease group and the strategies to optimize outcomes for patients with pulmonary arterial hypertension who require lung transplantation. SUMMARY Lung transplantation is an effective and lifesaving therapy for patients with end-stage lung disease. Sadly, patients with pulmonary arterial hypertension face many challenges as it relates to transplantation including higher perioperative risks, inequities in the allocation system, and less favorable long-term outcomes. This review covers the complexities of transplantation in patients with pulmonary vascular disease.
Collapse
Affiliation(s)
- Nicholas A Kolaitis
- Department of Medicine, University of California, San Francisco School of Medicine, San Francisco, CA.
| |
Collapse
|
24
|
Singer JP, Gao Y, Huang CY, Kordahl RC, Sriram A, Hays SR, Kukreja J, Venado A, Calabrese DR, Greenland JR. The Association Between Frailty and Chronic Lung Allograft Dysfunction After Lung Transplantation. Transplantation 2023; 107:2255-2261. [PMID: 37287095 PMCID: PMC10524113 DOI: 10.1097/tp.0000000000004672] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
BACKGROUND After lung transplantation, both frailty and chronic lung allograft dysfunction (CLAD) commonly develop, and when they do, are associated with poorer outcomes. Given their potential shared mechanisms, we sought to explore the temporal relationship between frailty and CLAD onset. METHODS In a single center, we prospectively measured frailty by the short physical performance battery (SPPB) repeatedly after transplant. Because of the nature of the relationship between frailty and CLAD is unknown, we tested the association between frailty, modeled as a time-dependent predictor, and CLAD development as well as CLAD development, modeled as a time-dependent predictor, and frailty development. To do so, we used Cox proportional cause-specific hazards and conditional logistic regression models adjusted for age, sex, race, diagnosis, cytomegalovirus serostatus, posttransplant body mass index, and acute cellular rejection episodes as time-dependent covariates. We tested SPPB frailty as a binary (≤9 points) and continuous predictor (12-point scale); as an outcome, we defined frailty as SPPB ≤9. RESULTS The 231 participants were a mean age of 55.7 y (SD 12.1). After adjusting for covariates, the development of frailty within 3 y after lung transplant was associated with cause-specific CLAD risk (adjusted cause-specific hazard ratio: 1.76; 95% confidence interval [CI], 1.05-2.92 when defining frailty as SPPB ≤9 and adjusted cause-specific hazard ratio: 1.10, 95% CI, 1.03-1.18 per 1-point worsening in SPPB). CLAD onset did not appear to be a risk factor for subsequent frailty (odds ratio, 4.0; 95% CI, 0.4-197.0). CONCLUSIONS Studying the mechanisms underlying frailty and CLAD could provide new insights into the pathobiology of both and potential targets for intervention.
Collapse
Affiliation(s)
- Jonthan P Singer
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Ying Gao
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Chiung-Yu Huang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA
| | - Rose C Kordahl
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Anya Sriram
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Steven R Hays
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jasleen Kukreja
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
| | - Aida Venado
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Daniel R Calabrese
- Department of Medicine, University of California San Francisco, San Francisco, CA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
| | - John R Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
| |
Collapse
|
25
|
Salami AC, Huddleston SJ. Beating the odds: long-term survival after lung transplantation. J Thorac Dis 2023; 15:4558-4560. [PMID: 37868854 PMCID: PMC10586948 DOI: 10.21037/jtd-23-1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/11/2023] [Indexed: 10/24/2023]
Affiliation(s)
- Aitua C. Salami
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, USA
| | | |
Collapse
|
26
|
Kandasamy S, Lau CL, Krupnick AS. Pulmonary transplantation-is it a long-term solution? J Thorac Dis 2023; 15:4148-4150. [PMID: 37691647 PMCID: PMC10482618 DOI: 10.21037/jtd-23-956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/21/2023] [Indexed: 09/12/2023]
Affiliation(s)
- Sivaveera Kandasamy
- Division of Thoracic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christine L Lau
- Division of Thoracic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexander Sasha Krupnick
- Division of Thoracic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Miggins JJ, Reul RM, Barrett S, Rana A, Alnajar A, Dunson J, Shafii A, Garcha P, Goss J, Loor G. Twenty-year survival following lung transplantation. J Thorac Dis 2023; 15:2997-3012. [PMID: 37426158 PMCID: PMC10323557 DOI: 10.21037/jtd-22-1414] [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: 11/10/2022] [Accepted: 03/03/2023] [Indexed: 07/11/2023]
Abstract
Background Lung transplantation median survival has seen improvements due to recognition of short-term survival factors but continues to trail behind other solid organs due to limited understanding of long-term survivorship. Given the creation of the United Network for Organ Sharing (UNOS) database in 1986, it was difficult to accrue data on long-term survivors until recently. This study characterizes factors impacting lung transplant survival beyond 20 years, conditional to 1-year survival. Methods Lung transplant recipients listed in UNOS from 1987 to 2002 who survived to 1 post-transplant year were reviewed. Kaplan-Meier and adjusted Cox regression analyses were performed at 20 and 10 years to identify risk factors associated with long-term outcomes independent of their short-term effects. Results A total of 6,172 recipients were analyzed, including 472 (7.6%) recipients who lived 20+ years. Factors associated with increased likelihood of 20-year survival were female-to-female gender match, recipient age 25-44, waitlist time >1 year, human leukocyte antigen (HLA) mismatch level 3, and donor cause of death: head trauma. Factors associated with decreased 20-year survival included recipient age ≥55, chronic obstructive pulmonary disease/emphysema (COPD/E) diagnosis, donor smoking history >20 pack-years, unilateral transplant, blood groups O&AB, recipient glomerular filtration rate (GFR) <10 mL/min, and donor GFR 20-29 mL/min. Conclusions This is the first study identifying factors associated with multiple-decade survival following lung transplant in the United States. Despite its challenges, long-term survival is possible and more likely in younger females in good waitlist condition without COPD/E who receive a bilateral allograft from a non-smoking, gender-matched donor of minimal HLA mismatch. Further analysis of the molecular and immunologic implications of these conditions are warranted.
Collapse
Affiliation(s)
| | - Ross M. Reul
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Abbas Rana
- Division of Abdominal Transplantation, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Ahmed Alnajar
- Division of Cardiothoracic Surgery, Department of Surgery, University of Miami, Miami, FL, USA
| | - Jordan Dunson
- Department of Orthopaedic Surgery, UT College of Medicine Chattanooga, Chattanooga, TN, USA
| | - Alexis Shafii
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Puneet Garcha
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - John Goss
- Division of Abdominal Transplantation, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
28
|
Dégot T, Douvry B, Falque L, Bautin N, Frachon I, Mankikian J, Le Pavec J, Picard C. [Follow-up strategies for lung transplant recipients in France]. Rev Mal Respir 2023; 40:314-323. [PMID: 36868975 DOI: 10.1016/j.rmr.2023.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/24/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Lung transplantation (LT) requires sustained care for a frequently polypathological condition. Follow-up is focused on three main issues: 1/stability of respiratory function; 2/comorbidity management; 3/preventive medicine. About 3000 LT patients in France are treated in 11 LT centers. Given the increased size of the LT recipient cohort, follow-up might be partially shared with peripheral centers. METHODS This paper presents the suggestions of a working group of the SPLF (French-speaking respiratory medicine society) on possible modalities of shared follow-up. RESULTS While the main LT center is tasked with centralizing follow-up, particularly the choice of optimal immunosuppression, an identified peripheral center (PC) may serve as an alternative to deal with acute events, comorbidities and routine assessment. Communication between the different centers should be free-flowing. Shared follow-up may be offered from the 3rd postoperative year to stable and consenting patients, whereas unstable and non-observant patients are poor candidates. CONCLUSION These guidelines may serve as a reference for any pneumologist wishing to effectively contribute to follow-up, even and especially subsequent to lung transplant.
Collapse
Affiliation(s)
- T Dégot
- Groupe de transplantation pulmonaire, service de pneumologie, hôpitaux universitaires de Strasbourg, nouvel hôpital Civil, Strasbourg, France.
| | - B Douvry
- Service de pneumologie, centre hospitalier intercommunal, centre des maladies respiratoires rares (RESPIRARE®), CRCM, Créteil, France; Inserm, IMRB, université Paris-Est Créteil, 94010 Créteil, France
| | - L Falque
- Pôle thorax et vaisseaux, service hospitalier universitaire pneumologie physiologie, CHU de Grenoble-Alpes, Grenoble, France
| | - N Bautin
- Inserm, CHU de Lille, université de Lille, CNRS, institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - I Frachon
- Service de pneumologie, CHU, hôpital Cavale Blanche, Brest, France
| | - J Mankikian
- Service de pneumologie et d'explorations fonctionnelles respiratoires, CHU, hôpital Bretonneau, Tours, France
| | - J Le Pavec
- Service de pneumologie et transplantation pulmonaire, groupe hospitalier Marie-Lannelongue - Saint-Joseph, Le Plessis-Robinson, France; Université Paris-Saclay, Le Kremlin-Bicêtre, France; Inserm, UMR_S 999, université Paris-Sud, groupe hospitalier Marie-Lannelongue - Saint-Joseph, Le Plessis-Robinson, France
| | - C Picard
- Groupe de transplantation pulmonaire, service de pneumologie, hôpital Foch, Suresnes, France
| | | |
Collapse
|
29
|
Olson MT, Liu W, Mohanakumar T, Bremner RM. A potential mechanism by which aspiration of duodenogastric fluid augments the risk for bronchiolitis obliterans syndrome after lung transplantation. J Thorac Cardiovasc Surg 2023; 165:e23-e37. [PMID: 35428458 DOI: 10.1016/j.jtcvs.2022.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Aspiration of duodenogastric refluxate may damage the respiratory epithelium of lung allografts in transplant recipients. We sought to define a mechanism by which aspiration of duodenogastric fluid augments the risk of bronchiolitis obliterans syndrome after lung transplant in a murine model. METHODS We analyzed the immunological effects of acute aspiration of duodenogastric fluid (0.5 mL/kg) on transplant naive (strain DBA/2J) and transplanted mice (strain B6D2F1/J to strain DBA/2J). Serum antibodies to the lung self-antigens (SAgs) K-alpha1 tubulin and collagen-V were determined by enzyme-linked immunosorbent assay. Exosomes were isolated from serum, and immunoblot membranes were probed for antibodies to lung SAgs. Lung sections were assessed for fibrotic burden and obliterative bronchiolitis lesions by histologic and immunohistochemical analyses, including trichrome staining. RESULTS Transplanted mice that received duodenogastric fluid developed higher levels of antibodies to the lung SAgs K-alpha1 tubulin and collagen-V and exosomes with lung SAgs on posttransplant days 14 and 28 than transplanted mice with sham aspiration or transplant naive mice (with and without aspiration). All lung allografts demonstrated severe grade A4 rejection on posttransplant day 14, with the highest mean fibrotic burden and mean number of obliterative bronchiolitis-like lesions per microscopic field on day 28 in recipients with aspiration. CONCLUSIONS This study links aspiration of duodenogastric fluid after lung transplant to higher autoimmune responses to lung SAgs and the release of circulating exosomes with lung SAgs, which together promote sustained immune responses leading to extensive lung parenchymal damage and, ultimately, severe obliterative bronchiolitis-the histologic hallmark of bronchiolitis obliterans syndrome.
Collapse
Affiliation(s)
- Michael T Olson
- University of Arizona College of Medicine-Phoenix Campus, Phoenix, Ariz; Norton Thoracic Institute Research Laboratory, St Joseph's Hospital and Medical Center, Phoenix, Ariz; Division of Thoracic Surgery, Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Ariz
| | - Wei Liu
- Norton Thoracic Institute Research Laboratory, St Joseph's Hospital and Medical Center, Phoenix, Ariz
| | - Thalachallour Mohanakumar
- Norton Thoracic Institute Research Laboratory, St Joseph's Hospital and Medical Center, Phoenix, Ariz
| | - Ross M Bremner
- Division of Thoracic Surgery, Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, Ariz.
| |
Collapse
|
30
|
Rousselière A, Delbos L, Foureau A, Reynaud-Gaubert M, Roux A, Demant X, Le Pavec J, Kessler R, Mornex JF, Messika J, Falque L, Le Borgne A, Boussaud V, Tissot A, Hombourger S, Bressollette-Bodin C, Charreau B. Changes in HCMV immune cell frequency and phenotype are associated with chronic lung allograft dysfunction. Front Immunol 2023; 14:1143875. [PMID: 37187736 PMCID: PMC10175754 DOI: 10.3389/fimmu.2023.1143875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Background Human cytomegalovirus (HCMV) infection is common and often severe in lung transplant recipients (LTRs), and it is a risk factor associated with chronic lung allograft dysfunction (CLAD). The complex interplay between HCMV and allograft rejection is still unclear. Currently, no treatment is available to reverse CLAD after diagnosis, and the identification of reliable biomarkers that can predict the early development of CLAD is needed. This study investigated the HCMV immunity in LTRs who will develop CLAD. Methods This study quantified and phenotyped conventional (HLA-A2pp65) and HLA-E-restricted (HLA-EUL40) anti-HCMV CD8+ T (CD8 T) cell responses induced by infection in LTRs developing CLAD or maintaining a stable allograft. The homeostasis of immune subsets (B, CD4T, CD8 T, NK, and γδT cells) post-primary infection associated with CLAD was also investigated. Results At M18 post-transplantation, HLA-EUL40 CD8 T responses were less frequently found in HCMV+ LTRs (21.7%) developing CLAD (CLAD) than in LTRs (55%) keeping a functional graft (STABLE). In contrast, HLA-A2pp65 CD8 T was equally detected in 45% of STABLE and 47.8% of CLAD LTRs. The frequency of HLA-EUL40 and HLA-A2pp65 CD8 T among blood CD8 T cells shows lower median values in CLAD LTRs. Immunophenotype reveals an altered expression profile for HLA-EUL40 CD8 T in CLAD patients with a decreased expression for CD56 and the acquisition of PD-1. In STABLE LTRs, HCMV primary infection causes a decrease in B cells and inflation of CD8 T, CD57+/NKG2C+ NK, and δ2-γδT cells. In CLAD LTRs, the regulation of B, total CD8 T, and δ2+γδT cells is maintained, but total NK, CD57+/NKG2C+ NK, and δ2-γδT subsets are markedly reduced, while CD57 is overexpressed across T lymphocytes. Conclusions CLAD is associated with significant changes in anti-HCMV immune cell responses. Our findings propose that the presence of dysfunctional HCMV-specific HLA-E-restricted CD8 T cells together with post-infection changes in the immune cell distribution affecting NK and γδT cells defines an early immune signature for CLAD in HCMV+ LTRs. Such a signature may be of interest for the monitoring of LTRs and may allow an early stratification of LTRs at risk of CLAD.
Collapse
Affiliation(s)
- Amélie Rousselière
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Laurence Delbos
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Aurore Foureau
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
- Nantes Université, CHU Nantes, Service de Pneumologie, Institut du thorax, Nantes, France
| | - Martine Reynaud-Gaubert
- CHU de Marseille, APHM, Hôpital Nord, Service de Pneumologie et Equipe de Transplantation pulmonaire; Marseille, France; Aix-Marseille Université, Marseille, France
| | - Antoine Roux
- Hôpital Foch, Service de pneumologie, Suresnes, France
| | - Xavier Demant
- Hôpital Haut-Lévêque, Service de pneumologie, CHU de Bordeaux, Bordeaux, France
| | - Jérôme Le Pavec
- Service de Pneumologie et de Transplantation Pulmonaire, Groupe Hospitalier Marie-Lannelongue -Paris Saint Joseph, Le Plessis-Robinson, France
- Université Paris-Saclay, Le Kremlin Bicêtre, France
- UMR_S 999, Université Paris–Sud, Inserm, Groupe hospitalier Marie-Lannelongue-Saint Joseph, Le Plessis-Robinson, France
| | - Romain Kessler
- Groupe de transplantation pulmonaire des hôpitaux universitaires de Strasbourg, Inserm-Université de Strasbourg, Strasbourg, France
| | - Jean-François Mornex
- Université de Lyon, Université Lyon1, INRAE, IVPC, Lyon, France
- Hospices Civils de Lyon, GHE, Service de Pneumologie, Inserm, Lyon, France
| | - Jonathan Messika
- APHP, Nord-Université Paris Cité, Hôpital Bichat-Claude Bernard, Service de Pneumologie B et Transplantation Pulmonaire, Paris, France
- Physiopathology and Epidemiology of Respiratory Diseases, UMR1152 INSERM and Université de Paris, Paris, France
| | - Loïc Falque
- Service Hospitalier Universitaire Pneumologie et Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France
| | | | - Véronique Boussaud
- Service de Pneumologie, Hôpital Européen Georges-Pompidou, Paris, France
| | - Adrien Tissot
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
- Nantes Université, CHU Nantes, Service de Pneumologie, Institut du thorax, Nantes, France
| | - Sophie Hombourger
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Céline Bressollette-Bodin
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
- CHU Nantes, Nantes Université, Laboratoire de Virologie, Nantes, France
| | - Béatrice Charreau
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
- CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France
- *Correspondence: Béatrice Charreau,
| |
Collapse
|
31
|
Abstract
Lung transplantation provides a treatment option for many individuals with advanced lung disease due to cystic fibrosis (CF). Since the first transplants for CF in the 1980s, survival has improved and the opportunity for transplant has expanded to include individuals who previously were not considered candidates for transplant. Criteria to be a transplant candidate vary significantly among transplant programs, highlighting that the engagement in more than one transplant program may be necessary. Individuals with highly resistant CF pathogens, malnutrition, osteoporosis, CF liver disease, and other comorbidities may be suitable candidates for lung transplant, or if needed, multi-organ transplant. The transplant process involves several phases, from discussion of prognosis and referral to a transplant center, to transplant evaluation, to listing, transplant surgery, and care after transplant. While the availability of highly effective CF transmembrane conductance regulator (CFTR) modulators for many individuals with CF has improved lung function and slowed progression to respiratory failure, early discussion regarding transplant as a treatment option and referral to a transplant program are critical to maximizing opportunity and optimizing patient and family experience. The decision to be evaluated for transplant and to list for transplant are distinct, and early referral may provide a treatment option that can be urgently executed if needed. Survival after transplant for CF is improving, to a median survival of approximately 10 years, and most transplant survivors enjoy significant improvement in quality of life.
Collapse
|
32
|
Dillon WP, Acosta TP, Failla A, Corrales J, Alangaden G, Ramesh M. Utility of microbiologic testing in surveillance bronchoscopy following lung transplantation: A retrospective cohort study. Transpl Infect Dis 2022; 24:e13989. [PMID: 36380574 DOI: 10.1111/tid.13989] [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: 05/27/2021] [Revised: 09/28/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND The utility of surveillance bronchoscopy (SB) for the clinical management of lung transplant recipients (LTRs) is undefined. This study evaluates the role of SB in the monitoring and care of LTRs. METHODS We retrospectively analyzed all LTRs who had SB at Henry Ford Hospital in Detroit, Michigan between August 2014 and August 2019. Bronchoscopies performed for clinical symptoms, new radiographic abnormalities, and to assess stents or acute rejection were excluded. A total of 107 LTRs and 449 bronchoscopies were analyzed. The primary outcome was the rate of change in clinical care based on microbiologic and pathologic test results. Secondary outcomes were rates of microbiologic and pathologic test positivity and rates of adverse effects. RESULTS The most common microbiologic tests performed on bronchoalveolar lavage were bacterial (96.9%), fungal (95.3%), and acid-fast bacillus (95.1%) stains and cultures. Of 2560 microbiologic tests, 22.0% were positive and resulted in therapy changes for 2.9%. Positive galactomannan, acid-fast bacillus tests, and Pneumocystis jirovecii antigen/polymerase chain reaction did not result in therapy changes. Of the 370 transbronchial biopsies performed, 82.2% were negative for acute rejection and 13% were positive for A1/A2 rejection. Immunosuppressive therapy changes occurred after 15.8% with reduction in immunosuppression due to positive microbiologic tests in 16.9%. Adverse events occurred in 8.0% of patients. CONCLUSION Diagnostic stewardship is warranted when performing SB in LTRs.
Collapse
Affiliation(s)
- William P Dillon
- Division of Infectious Diseases, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tommy Parraga Acosta
- Division of Infectious Diseases, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Andrew Failla
- Division of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Julio Corrales
- Division of Pulmonary Medicine and Critical Care, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - George Alangaden
- Division of Infectious Diseases, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mayur Ramesh
- Division of Infectious Diseases, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
33
|
Functional Blockage of S100A8/A9 Ameliorates Ischemia–Reperfusion Injury in the Lung. Bioengineering (Basel) 2022; 9:bioengineering9110673. [DOI: 10.3390/bioengineering9110673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
(1) Background: Lung ischemia–reperfusion (IR) injury increases the mortality and morbidity of patients undergoing lung transplantation. The objective of this study was to identify the key initiator of lung IR injury and to evaluate pharmacological therapeutic approaches using a functional inhibitor against the identified molecule. (2) Methods: Using a mouse hilar clamp model, the combination of RNA sequencing and histological investigations revealed that neutrophil-derived S100A8/A9 plays a central role in inflammatory reactions during lung IR injury. Mice were assigned to sham and IR groups with or without the injection of anti-S100A8/A9 neutralizing monoclonal antibody (mAb). (3) Results: Anti-S100A8/A9 mAb treatment significantly attenuated plasma S100A8/A9 levels compared with control IgG. As evaluated by oxygenation capacity and neutrophil infiltration, the antibody treatment dramatically ameliorated the IR injury. The gene expression levels of cytokines and chemokines induced by IR injury were significantly reduced by the neutralizing antibody. Furthermore, the antibody treatment significantly reduced TUNEL-positive cells, indicating the presence of apoptotic cells. (4) Conclusions: We identified S100A8/A9 as a novel therapeutic target against lung IR injury.
Collapse
|
34
|
Fu A, Vasileva A, Hanafi N, Belousova N, Wu J, Rajyam SS, Ryan CM, Hantos Z, Chow CW. Characterization of chronic lung allograft dysfunction phenotypes using spectral and intrabreath oscillometry. Front Physiol 2022; 13:980942. [PMID: 36277208 PMCID: PMC9582781 DOI: 10.3389/fphys.2022.980942] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic lung allograft dysfunction (CLAD) is the major cause of death beyond 2 years after lung transplantation and develops in 50% of all patients by 5 years post-transplant. CLAD is diagnosed on the basis of a sustained drop of 20% for at least 3 months in the forced expiratory volume (FEV1), compared to the best baseline value achieved post-transplant. CLAD presents as two main phenotypes: bronchiolitis obliterans syndrome (BOS) is more common and has better prognosis than restrictive allograft syndrome (RAS). Respiratory oscillometry is a different modality of lung function testing that is highly sensitive to lung mechanics. The current study investigated whether spectral and intrabreath oscillometry can differentiate between CLAD-free, BOS- and RAS-CLAD at CLAD onset, i.e., at the time of the initial 20% drop in the FEV1. Methods: A retrospective, cross-sectional analysis of 263 double lung transplant recipients who underwent paired testing with oscillometry and spirometry at the Toronto General Pulmonary Function Laboratory from 2017 to 2022 was conducted. All pulmonary function testing and CLAD diagnostics were performed following international guidelines. Statistical analysis was conducted using multiple comparisons. Findings: The RAS (n = 6) spectral oscillometry pattern differs from CLAD-free (n = 225) by right-ward shift of reactance curve similar to idiopathic pulmonary fibrosis whereas BOS (n = 32) has a pattern similar to obstructive lung disease. Significant differences were found in most spectral and intrabreath parameters between BOS, RAS, and time-matched CLAD-free patients. Post-hoc analysis revealed these differences were primarily driven by BOS instead of RAS. While no differences were found between CLAD-free and RAS patients with regards to spectral oscillometry, the intrabreath metric of reactance at end-inspiration (XeI) was significantly different (p < 0.05). BOS and RAS were differentiated by spectral oscillometry measure R5, and intrabreath resistance at end expiration, ReE (p < 0.05 for both). Conclusion: Both spectral and intrabreath oscillometry can differentiate BOS-CLAD from CLAD-free states while intrabreath oscillometry, specifically XeI, can uniquely distinguish RAS-CLAD from CLAD-free. Spectral and intrabreath oscillometry offer complementary information regarding lung mechanics in CLAD patients to help distinguish the two phenotypes and could prove useful in prognostication.
Collapse
Affiliation(s)
- Anne Fu
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anastasiia Vasileva
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nour Hanafi
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Natalia Belousova
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Unit, University Health Network, Toronto, ON, Canada
| | - Joyce Wu
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Sarada Sriya Rajyam
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Clodagh M. Ryan
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
| | - Zoltán Hantos
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Chung-Wai Chow
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto Lung Transplant Program, Ajmera Multi-Organ Transplant Unit, University Health Network, Toronto, ON, Canada
- Toronto General Pulmonary Function Laboratory, University Health Network, Toronto, ON, Canada
- *Correspondence: Chung-Wai Chow,
| |
Collapse
|
35
|
The International Thoracic Organ Transplant Registry of the International Society for Heart and Lung Transplantation: Thirty-ninth adult lung transplantation report-2022; focus on lung transplant recipients with chronic obstructive pulmonary disease. J Heart Lung Transplant 2022; 41:1335-1347. [PMID: 36050206 DOI: 10.1016/j.healun.2022.08.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/21/2022] Open
|
36
|
Survival difference between high-risk and low-risk CFTR genotypes after lung transplant. J Heart Lung Transplant 2022; 41:1511-1519. [PMID: 35864004 PMCID: PMC10124119 DOI: 10.1016/j.healun.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/11/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND While cystic fibrosis transmembrane conductance regulator (CFTR) genotypes are associated with clinical outcomes in cystic fibrosis patients, it is unknown if genotype impacts lung transplant outcomes. We sought to compare lung transplant survival and time to bronchiolitis obliterans syndrome (BOS) between high-risk, low-risk, and not yet classified CFTR genotypes. METHODS We used merged data from the Organ Procurement and Transplantation Network (2005-2017) and United States Cystic Fibrosis Foundation Patient Registry (2005-2016). Cox Proportional Hazards models compared graft failure after lung transplant and time to BOS among high-risk, low-risk, and not yet classified risk CFTR genotype classes. RESULTS Among 1,830 cystic fibrosis lung transplant recipients, median survival for those with low-risk, high-risk, and not yet classified genotype was 9.83, 6.25, and 5.75 years, respectively. Adjusted Cox models showed recipients with a low-risk genotype had 39% lower risk of death or re-transplant compared to those with high-risk genotype (adjusted HR 0.61, 95% CI = 0.40, 0.91). A subset of 1,585 lung transplant recipients were included in the BOS subgroup analysis. Adjusted analyses showed no significant difference of developing BOS among high-risk, low-risk, or not yet classified genotypes. CONCLUSIONS Lung transplant recipients with low-risk CFTR genotype have better survival after transplant compared to recipients with high-risk or not yet classified genotypes. Given these differences, future studies evaluating the mechanism by which CFTR genotype affects post-transplant survival could identify potential targets for intervention.
Collapse
|
37
|
Perez AA, Shah RJ. Critical Care of the Lung Transplant Patient. Clin Chest Med 2022; 43:457-470. [PMID: 36116814 DOI: 10.1016/j.ccm.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Lung transplantation is a therapeutic option for end-stage lung disease that improves survival and quality of life. Prelung transplant admission to the intensive care unit (ICU) for bridge to transplant with mechanical ventilation and extracorporeal membrane oxygenation (ECMO) is common. Primary graft dysfunction is an important immediate complication of lung transplantation with short- and long-term morbidity and mortality. Later transplant-related causes of respiratory failure necessitating ICU admission include acute cellular rejection, atypical infections, and chronic lung allograft dysfunction. Lung transplantation for COVID-19-related ARDS is increasingly common..
Collapse
Affiliation(s)
- Alyssa A Perez
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, 400 Parnassus Street, 5th Floor, San Francisco, CA 94143, USA.
| | - Rupal J Shah
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, 400 Parnassus Street, 5th Floor, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Levine DJ, Demko ZP, Ross DJ. Variability in plasma donor-derived cell-free DNA levels with CLAD more than 5-years after Lung Transplantation: Pilot data. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
39
|
Ahmad D, O'Malley TJ, Jordan AM, Maynes EJ, Saxena A, Prochno KW, Rajab TK, Massey HT, Daly RC, Tchantchaleishvili V. Bronchial artery revascularization in lung transplantation: a systematic review and meta-analysis. J Thorac Dis 2022; 14:3285-3294. [PMID: 36245610 PMCID: PMC9562503 DOI: 10.21037/jtd-22-213] [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: 02/17/2022] [Accepted: 07/16/2022] [Indexed: 01/18/2023]
Abstract
Background Bronchial artery revascularization (BAR) during lung transplantation has been hypothesized to improve early tracheal healing and delay the onset of bronchiolitis obliterans syndrome (BOS). We aimed to assess the outcomes of BAR after lung transplantation. Methods Electronic search in Ovid Medline, Cumulative Index of Nursing and Allied Health Literature (CINAHL), Scopus, and Cochrane Controlled Trials Register (CCTR) databases was performed to identify all relevant studies published about lung transplantation with BAR. Studies discussing lung transplantation utilizing BAR were included while those without outcome data such as BOS and survival were excluded. Cohort-level data were extracted and pooled for analysis. A binary outcome meta-analysis of proportions with logit transformation was conducted. Newcastle-Ottawa scale was used for risk of bias assessment. Results Seven studies were selected for the analysis comprising 143 patients. Mean patient age was 47 (95% CI: 40-55) years. Sixty-one percent (48-72%) were male. Seventy-three percent (65-79%) of patients underwent double lung transplant while 27% (21-25%) underwent single lung transplant. In patients with postoperative angiography, successful BAR was demonstrated in 93% (82-97%) of all assessed conduits. The 30-day/in-hospital mortality was 6% (3-11%). Seventy-nine percent (63-89%) of patients were free from rejection at three months. Eighty-three percent (29-98%) of patients were free from signs of airway ischemia at three and six months. Pooled survival at one year and five years was 87% (78-92%) and 71% (46-87%), respectively, with a mean follow-up time of 21 (3-38) months. Pooled freedom from bronchiolitis obliterans was 86% (77-91%) at two years. Conclusions While this systematic review and meta-analysis is limited by the available surgeons, institutions, and papers discussing a highly specialized technique, it does show that BAR is a viable technique to minimize BOS and early anastomotic intervention following lung transplantation.
Collapse
Affiliation(s)
- Danial Ahmad
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Thomas J. O'Malley
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew M. Jordan
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elizabeth J. Maynes
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Abhiraj Saxena
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kyle W. Prochno
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Taufiek K. Rajab
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Howard T. Massey
- Division of Cardiac Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Richard C. Daly
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
40
|
Erasmus DB, Durand N, Alvarez FA, Narula T, Hodge DO, Zubair AC. Feasibility and Safety of Low-Dose Mesenchymal Stem Cell Infusion in Lung Transplant Recipients. Stem Cells Transl Med 2022; 11:891-899. [PMID: 35881142 PMCID: PMC9492292 DOI: 10.1093/stcltm/szac051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/12/2022] [Indexed: 12/02/2022] Open
Abstract
Background We have previously shown bone marrow-derived mesenchymal stem cells (MSCs) may shift immune responses toward anti-inflammatory pathways and stabilize the course of obstructive chronic lung allograft syndrome (o-CLAD) after lung transplantation. In this study, we measured the response of lower dose infusions. Methods We infused low-dose MSCs intravenously in 13 patients who had developed moderate-to-severe o-CLAD. Three had previously received an infusion of MSCs from a different donor and were re-dosed at 1 × 106 MSC/kg, while 5 received a first dose at 1 × 106 MSC/kg and five received an even lower dose at 0.5 × 106 MSC/kg. We recorded pulmonary function tests before and after infusion, and patients were followed clinically for 12 months. Results Infusions were well tolerated, and no significant adverse events were recorded in the first 30 days. There was significant decline (mean ± SD) in forced vital capacity (FVC) (3.49 ± 1.03 vs 3.18 ± 0.94 L, P = .03) and forced expiratory volume in 1 second (FEV1) (2.28 ± 0.86 vs 1.77 ± 0.49 L, P = .04) over the year preceding infusion. FVC (3.18 ± 0.94 vs 3.46 ± 0.99 L, P = .53) and FEV1 was not significantly changed (1.77 ± 0.49 vs 1.88 ± 0.75, P = .72) when comparing values immediately prior to infusion to those obtained 1 year after infusion, indicating a possible stabilizing effect on lung function decline due to o-CLAD. Conclusion Intravenous infusions of bone marrow-derived MSCs are well tolerated in lung transplant recipients with moderate-to-severe CLAD. Low-dose MSCs appear to slow progression of CLAD in some patients.
Collapse
|
41
|
[The bronchial arteries: a small but vital contribution to lung perfusion after lung transplantation]. Pneumologie 2022; 76:552-559. [PMID: 35878603 DOI: 10.1055/a-1845-0286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Blood supply to the lungs is carried out by the pulmonary and bronchial-arterial system. The bronchial-arterial vessels are involved in supplying the small airways all the way up to the terminal bronchioles. The bronchial-arterial system is also necessary for the regulation of airway temperature, humidity and mucociliary clearance. Chronic ischaemia of the small airways due to damage or injury to bronchial arterial supply increases the risk of fibrosis of the small airways (bronchiolitis obliteration), especially in lung transplantation (LTx). Although survival after LTx has improved over time, it is, with a 5-year survival rate of only 50 to 60%, still significantly worse than that of other organ transplants. It is likely that bronchial arterial revascularisation at the time of LTx plays an important transplant-preserving function.
Collapse
|
42
|
Hao X, Peng C, Lian W, Liu H, Fu G. Effect of azithromycin on bronchiolitis obliterans syndrome in posttransplant recipients: A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e29160. [PMID: 35839027 PMCID: PMC11132355 DOI: 10.1097/md.0000000000029160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/07/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) is a devastating complication that occurs after transplantation. Although azithromycin is currently used for the treatment of BOS, the evidence is sparse and controversial. The aim of this meta-analysis is to evaluate the effects of azithromycin on forced expiratory volume in 1 second (FEV1) and patient's survival. METHODS PubMed, Embase, Cochrane library, Web of Science databases, and the ClinicalTrials.gov registry were systematically searched from inception until December 2020 for relevant original research articles. Random-effects models were used to calculate pooled-effect estimates. RESULTS Searches identified 15 eligible studies involving 694 participants. For FEV1 (L), there was a significant increase after short-term (≤12 weeks; P = .00) and mid-term (12-24 weeks; P = .01) administration of azithromycin. For FEV1 (%) compared to baseline, there was a significant increase after short-term (≤12 weeks) administration of azithromycin (P = .02), while there were no statistically significant differences in the medium and long term. When pooled FEV1% was predicted, it exhibited a similar trend to FEV1 (%) compared to baseline. In addition, we discovered that azithromycin reduced the risk of death (hazard ratio = 0.26; 95% confidence interval = 0.17 to 0.40; P = .00) in patients with BOS post-lung transplantation. CONCLUSIONS Azithromycin therapy is both effective and safe for lung function improvement in patients with posttransplant BOS after the short- and medium-term administration. Additionally, it has been demonstrated a significant survival benefit among patients with BOS post-lung transplant. Higher quality randomized controlled trials and more extensive prospective cohort studies are needed to confirm the effect of azithromycin on patients with posttransplant BOS.
Collapse
Affiliation(s)
- Xiaohui Hao
- Department of Pharmacy, Medical Supplies Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Cheng Peng
- Department of Pharmacy, Medical Supplies Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Wenwen Lian
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Han Liu
- Department of Pharmacy, Medical Supplies Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Guiying Fu
- Department of Pharmacy, Medical Supplies Center of the Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
43
|
Glanville AR, Benden C, Bergeron A, Cheng GS, Gottlieb J, Lease ED, Perch M, Todd JL, Williams KM, Verleden GM. Bronchiolitis obliterans syndrome after lung or haematopoietic stem cell transplantation: current management and future directions. ERJ Open Res 2022; 8:00185-2022. [PMID: 35898810 PMCID: PMC9309343 DOI: 10.1183/23120541.00185-2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/18/2022] [Indexed: 11/05/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) may develop after either lung or haematopoietic stem cell transplantation (HSCT), with similarities in histopathological features and clinical manifestations. However, there are differences in the contributory factors and clinical trajectories between the two conditions. BOS after HSCT occurs due to systemic graft-versus-host-disease (GVHD), whereas BOS after lung transplantation is limited to the lung allograft. BOS diagnosis after HSCT is more challenging, as the lung function decline may occur due to extrapulmonary GVHD, causing sclerosis or inflammation in the fascia or muscles of the respiratory girdle. Treatment is generally empirical with no established effective therapies. This review provides rare insights and commonalities of both conditions, that are not well elaborated elsewhere in contemporary literature, and highlights the importance of cross disciplinary learning from experts in other transplant modalities. Treatment algorithms for each condition are presented, based on the published literature and consensus clinical opinion. Immunosuppression should be optimised, and other conditions or contributory factors treated where possible. When initial treatment fails, the ultimate therapeutic option is lung transplantation (or re-transplantation in the case of BOS after lung transplantation) in carefully selected candidates. Novel therapies under investigation include aerosolised liposomal cyclosporine, Janus kinase inhibitors, antifibrotic therapies, and (in patients with BOS after lung transplantation) B-cell–directed therapies. Effective novel treatments that have a tangible impact on survival and thereby avoid the need for lung transplantation or re-transplantation are urgently required.
Collapse
|
44
|
Blackett JW, Benvenuto L, Leiva-Juarez MM, D'Ovidio F, Arcasoy S, Jodorkovsky D. Risk Factors and Outcomes for Gastroparesis After Lung Transplantation. Dig Dis Sci 2022; 67:2385-2394. [PMID: 34524597 DOI: 10.1007/s10620-021-07249-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gastroparesis is common after lung transplantation and is associated with worse transplant outcomes, including the development of chronic lung allograft dysfunction (CLAD). This study sought to identify the prevalence, risk factors, and outcomes associated with a new diagnosis of gastroparesis after lung transplantation. METHODS This was a single-center retrospective study of patients who underwent lung transplantation in 2008-2018. The primary outcome was a new diagnosis of gastroparesis within 3 years of transplant. Secondary outcomes included a new diagnosis of gastroesophageal reflux and the association between gastroparesis and both post-transplant survival and CLAD-free survival. Multivariable logistic regression was used to compare diagnosis of gastroparesis and gastroesophageal reflux, while multivariable Cox proportional hazards models were used to analyze gastroparesis and post-transplant outcomes. RESULTS Of 616 patients with no prior history of gastroparesis, 107 (17.4%) were diagnosed with delayed gastric emptying within 3 years of transplant. On multivariable logistic regression, black race (OR 2.16, 95% CI 1.18-3.98, p = 0.013) was significantly associated with a new diagnosis of gastroparesis. Age, sex, history of diabetes, connective tissue disease, type of transplant, diagnosis group, renal function, and body mass index were not predictive of gastroparesis post-transplant. Gastroparesis was significantly associated with CLAD (HR 1.76, 95% CI 1.20-2.59, p = 0.004), but not with overall mortality (HR 1.16, p = 0.43). CONCLUSION While gastroparesis is common after lung transplantation, it remains difficult to predict which patients will develop these complications post-transplant. Black patients were more likely to be diagnosed with gastroparesis after adjusting for relevant confounders. Gastroparesis is associated with increased risk of CLAD, and further studies are needed to assess whether early detection and treatment can reduce the incidence of CLAD.
Collapse
Affiliation(s)
- John W Blackett
- Division of Digestive and Liver Diseases, Department of Medicine, New York Presbyterian Columbia University Medical Center, 622 West 168th Street, New York, NY, USA.
| | - Luke Benvenuto
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, New York Presbyterian Columbia University Medical Center, New York, NY, USA
| | - Miguel M Leiva-Juarez
- Division of Cardiac, Vascular, and Thoracic Surgery, Department of Surgery, New York Presbyterian Columbia University Medical Center, New York, NY, USA
| | - Frank D'Ovidio
- Division of Cardiac, Vascular, and Thoracic Surgery, Department of Surgery, New York Presbyterian Columbia University Medical Center, New York, NY, USA
| | - Selim Arcasoy
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, New York Presbyterian Columbia University Medical Center, New York, NY, USA
| | - Daniela Jodorkovsky
- Division of Digestive and Liver Diseases, Department of Medicine, New York Presbyterian Columbia University Medical Center, 622 West 168th Street, New York, NY, USA
| |
Collapse
|
45
|
Niroomand A, Ghaidan H, Hallgren O, Hansson L, Larsson H, Wagner D, Mackova M, Halloran K, Hyllén S, Lindstedt S. Corticotropin releasing hormone as an identifier of bronchiolitis obliterans syndrome. Sci Rep 2022; 12:8413. [PMID: 35589861 PMCID: PMC9120482 DOI: 10.1038/s41598-022-12546-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Lung transplantion (LTx) recipients have low long-term survival and a high incidence of bronchiolitis obliterans syndrome (BOS), an inflammation of the small airways in chronic rejection of a lung allograft. There is great clinical need for a minimally invasive biomarker of BOS. Here, 644 different proteins were analyzed to detect biomarkers that distinguish BOS grade 0 from grades 1–3. The plasma of 46 double lung transplant patients was analyzed for proteins using a high-component, multiplex immunoassay that enables analysis of protein biomarkers. Proximity Extension Assay (PEA) consists of antibody probe pairs which bind to targets. The resulting polymerase chain reaction (PCR) reporter sequence can be quantified by real-time PCR. Samples were collected at baseline and 1-year post transplantation. Enzyme-linked immunosorbent assay (ELISA) was used to validate the findings of the PEA analysis across both time points and microarray datasets from other lung transplantation centers demonstrated the same findings. Significant decreases in the plasma protein levels of CRH, FERC2, IL-20RA, TNFB, and IGSF3 and an increase in MMP-9 and CTSL1 were seen in patients who developed BOS compared to those who did not. In this study, CRH is presented as a novel potential biomarker in the progression of disease because of its decreased levels in patients across all BOS grades. Additionally, biomarkers involving the remodeling of the extracellular matrix (ECM), such as MMP-9 and CTSL1, were increased in BOS patients.
Collapse
Affiliation(s)
- Anna Niroomand
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,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, 221 85, Lund, Sweden
| | - Oskar Hallgren
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Lennart Hansson
- Department of Pulmonology and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Hillevi Larsson
- Department of Pulmonology and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Darcy Wagner
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Experimental Medical Sciences, Lung Bioengineering and Regeneration, Lund University, Lund, Sweden
| | - Martina Mackova
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Kieran Halloran
- Alberta Transplant Applied Genomics Center, University of Alberta, Edmonton, Canada
| | - Snejana Hyllén
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, 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, 221 85, Lund, Sweden.
| |
Collapse
|
46
|
Terada Y, Takahashi T, Hachem RR, Liu J, Witt CA, Byers DE, Guillamet RV, Kulkarni HS, Nava RG, Kozower BD, Meyers BF, Pasque MK, Patterson GA, Kreisel D, Puri V. Clinical Features and Outcomes of Unplanned Single Lung Transplants. J Thorac Cardiovasc Surg 2022; 164:1650-1659.e3. [DOI: 10.1016/j.jtcvs.2022.01.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 01/01/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022]
|
47
|
Greenland NY, Deiter F, Calabrese DR, Hays SR, Kukreja J, Leard LE, Kolaitis NA, Golden JA, Singer JP, Greenland JR. Inflammation on bronchoalveolar lavage cytology is associated with decreased chronic lung allograft dysfunction-free survival. Clin Transplant 2022; 36:e14639. [PMID: 35246990 DOI: 10.1111/ctr.14639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lung transplant recipients undergo bronchoalveolar lavage (BAL) to detect antecedents of chronic lung allograft dysfunction (CLAD), but routine assessment of BAL cytology is controversial. We hypothesized that inflammation on BAL cytology would predict CLAD-free survival. METHODS In a single-center retrospective cohort, associations between cytology results and clinical characteristics were compared using generalized-estimating equation-adjusted regression. The association between BAL inflammation and CLAD or death risk was assessed using time-dependent Cox models. RESULTS In 3,365 cytology reports from 451 subjects, inflammation was the most common finding (6.2%, 210 cases), followed by fungal forms (5.3%, 178 cases, including 24 cases of suspected Aspergillus). Inflammation on BAL cytology was more common in procedures for symptoms (8.5%) versus surveillance (3.2%, P<0.001). Inflammation on cytology was associated with automated neutrophil and lymphocyte counts, acute cellular rejection, infection, and portended a 2.2-fold hazard ratio (CI 1.2-4.0, P = 0.007) for CLAD or death. However, inflammation by cytology did not inform CLAD-free survival risk beyond automated BAL cell counts (P = 0.57). CONCLUSIONS Inflammation on BAL cytology is clinically significant, suggesting acute rejection or infection and increased risk of CLAD or death. However, other indicators of allograft inflammation can substitute for much of the information provided by BAL cytology. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nancy Y Greenland
- Department of Anatomic Pathology, University of California, San Francisco, USA.,Veterans Affairs Health Care System, San Francisco, California, USA
| | - Fred Deiter
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Daniel R Calabrese
- Veterans Affairs Health Care System, San Francisco, California, USA.,Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Steven R Hays
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, USA
| | - Lorriana E Leard
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Nicholas A Kolaitis
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jeffrey A Golden
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - Jonathan P Singer
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| | - John R Greenland
- Veterans Affairs Health Care System, San Francisco, California, USA.,Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep Medicine Division, University of California, San Francisco, USA
| |
Collapse
|
48
|
Sheshadri A, Makhnoon S, Alousi AM, Bashoura L, Andrade R, Miller CJ, Stolar KR, Arain MH, Noor L, Balagani A, Jain A, Blanco D, Ortiz A, Taylor MS, Stenzler A, Mehta R, Popat UR, Hosing C, Ost DE, Champlin RE, Dickey BF, Peterson SK. Home-Based Spirometry Telemonitoring After Allogeneic Hematopoietic Cell Transplantation: Mixed Methods Evaluation of Acceptability and Usability. JMIR Form Res 2022; 6:e29393. [PMID: 35129455 PMCID: PMC8861865 DOI: 10.2196/29393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/06/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Home-based spirometry (HS) allows for the early detection of lung complications in recipients of an allogeneic hematopoietic cell transplant (AHCT). Although the usability and acceptability of HS are critical for adherence, patient-reported outcomes of HS use remain poorly understood in this setting. OBJECTIVE The aim of this study is to design a longitudinal, mixed methods study to understand the usability and acceptability of HS among recipients of AHCT. METHODS Study participants performed HS using a Bluetooth-capable spirometer that transmitted spirometry data to the study team in real time. In addition, participants completed usability questionnaires and in-depth interviews and reported their experiences with HS. Analysis of interview data was guided by the constructs of performance expectancy, effort expectancy, and social influence from the Unified Theory of Acceptance and Use of Technology model. RESULTS Recipients of AHCT found HS to be highly acceptable despite modest technological barriers. On average, participants believed that the HS was helpful in managing symptoms related to AHCT (scores ranging from 2.22 to 2.68 on a scale of 0-4) and for early detection of health-related problems (score range: 2.88-3.12). Participants viewed HS favorably and were generally supportive of continued use. No significant barriers to implementation were identified from the patient's perspective. Age and gender were not associated with the patient perception of HS. CONCLUSIONS Study participants found HS acceptable and easy to use. Some modifiable technical barriers to performing HS were identified; however, wider implementation of pulmonary screening is feasible from the patient's perspective.
Collapse
Affiliation(s)
- Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sukh Makhnoon
- Department of Behavioral Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amin M Alousi
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lara Bashoura
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rene Andrade
- Department of Behavioral Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christopher J Miller
- Department of Behavioral Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Karen R Stolar
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Muhammad Hasan Arain
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Laila Noor
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amulya Balagani
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Akash Jain
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David Blanco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abel Ortiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Alex Stenzler
- Monitored Therapeutics, Inc, Dublin, OH, United States
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Uday R Popat
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chitra Hosing
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David E Ost
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Richard E Champlin
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Burton F Dickey
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Susan K Peterson
- Department of Behavioral Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
49
|
Jobst S, Schaefer J, Kleiser C, Kugler C. A Systematized Review of Professional Employment Following Thoracic Transplantation. Prog Transplant 2022; 32:55-66. [PMID: 35006009 DOI: 10.1177/15269248211064883] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Acknowledging the evolved landscape in thoracic transplantation, professional employment becomes an important outcome measure to quantify the success of this costly procedure. OBJECTIVE We aimed to assess rates of and characterize factors associated with professional employment in patients following thoracic transplantation, and create an evidence-base on the relationship between professional employment and relevant outcome parameters. METHODS We systematically searched Medline, Cinahl, and GoogleScholar to identify studies published between 1998 and 2021 reporting on professional employment following heart and lung transplantation. RESULTS Twenty-two studies from 11 countries with varying sample sizes (N = 27; 10 066) were included. Employment rates ranged from 19.7% to 69.4% for heart, and from 7.4% to 50.8% for lung transplant recipients. Most frequently reported positively associated factors with employment after transplant were younger age, higher education, and history of pretransplant employment. Longer duration of unemployment prior to transplantation and Medicaid coverage were the most frequently reported negatively associated factors. Relationships between professional employment and clinical outcomes included lower rates of acute and chronic rejection, less infection episodes, and better quality of life among working patients; one study reported a lower 5-year-mortality rate. Reasons not to work were "physical or mental health-related," "employment-related," "financial reasons," and "lifestyle choices." DISCUSSION Substantial proportions of patients following thoracic transplantation are not professionally employed, potentially diminishing the success of transplantation on individual and societal levels. Considering adverse clinical outcomes in employed transplant recipients were low, more efforts are needed to identify modifiable factors for employment in these populations.
Collapse
Affiliation(s)
- Stefan Jobst
- Institute of Nursing Science, University of Freiburg, Freiburg, Germany
| | - Jonas Schaefer
- Institute of Nursing Science, University of Freiburg, Freiburg, Germany
| | - Christina Kleiser
- Institute of Nursing Science, University of Freiburg, Freiburg, Germany
| | - Christiane Kugler
- Institute of Nursing Science, University of Freiburg, Freiburg, Germany
| |
Collapse
|
50
|
Neurohr C, Kneidinger N, Ghiani A, Monforte V, Knoop C, Jaksch P, Parmar J, Ussetti P, Sole A, Müller-Quernheim J, Kessler R, Wirtz H, Boerner G, Denk O, Prante Fernandes S, Behr J. A randomized controlled trial of liposomal cyclosporine A for inhalation in the prevention of bronchiolitis obliterans syndrome following lung transplantation. Am J Transplant 2022; 22:222-229. [PMID: 34587371 DOI: 10.1111/ajt.16858] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/20/2021] [Accepted: 09/11/2021] [Indexed: 01/25/2023]
Abstract
Long-term survival after lung transplantation is limited by chronic allograft dysfunction. The aim of this study was to investigate the effect of locally augmented immunosuppression with liposomal cyclosporine A for inhalation (L-CsA-i) for the prevention of bronchiolitis obliterans syndrome (BOS). In a randomized, double-blind, placebo-controlled, multi-center Phase 3 study, 180 LT recipients in BOS grade 0 were planned to receive L-CsA-i or placebo in addition to triple-drug immunosuppression. L-CsA-i was administered twice daily via an Investigational eFlow nebulizer to recipients of single (SLT) and bilateral lung transplants (BLT) within 6-32 weeks posttransplant, and continued for 2 years. The primary endpoint was BOS-free survival. 130 patients were enrolled before the study was prematurely terminated for business reasons. Despite a 2-year actuarial difference in BOS-free survival of 14.1% in favor of L-CsA-i in the overall study population, the primary endpoint was not met (p = .243). The pre-defined per protocol analysis of SLT recipients (n = 24) resulted in a treatment difference of 58.2% (p = .053). No difference was observed in the BLT (n = 48) subpopulation (p = .973). L-CsA-i inhalation was well tolerated. Although this study failed to meet its primary endpoint, the results warrant additional investigation of L-CsA-i in lung transplant recipients.
Collapse
Affiliation(s)
- Claus Neurohr
- Department of Medicine V, University Hospital, LMU Munich, German Center for Lung Research (DZL), Munich, Germany.,Department of Pulmonology and Respiratory Medicine, Robert-Bosch-Krankenhaus Stuttgart, Stuttgart, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, University Hospital, LMU Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Alessandro Ghiani
- Department of Pulmonology and Respiratory Medicine, Robert-Bosch-Krankenhaus Stuttgart, Stuttgart, Germany
| | | | | | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | - Amparo Sole
- Hospital Universitario La Fe, Valencia, Spain
| | - Joachim Müller-Quernheim
- Department of Pneumology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Romain Kessler
- Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hubert Wirtz
- Department of Respiratory Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Gerhard Boerner
- BREATH Therapeutics - a Zambon Group Company, Munich, Germany
| | - Oliver Denk
- BREATH Therapeutics - a Zambon Group Company, Munich, Germany
| | | | - Juergen Behr
- Department of Medicine V, University Hospital, LMU Munich, German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|