1
|
Kerckhof P, Ambrocio GPL, Beeckmans H, Kaes J, Geudens V, Bos S, Willems L, Vermaut A, Vermant M, Goos T, De Fays C, Aversa L, Mohamady Y, Vanstapel A, Orlitová M, Van Slambrouck J, Jin X, Varghese V, Josipovic I, Boone MN, Dupont LJ, Weynand B, Dubbeldam A, Van Raemdonck DE, Ceulemans LJ, Gayan-Ramirez G, De Sadeleer LJ, McDonough JE, Vanaudenaerde BM, Vos R. Ventilatory capacity in CLAD is driven by dysfunctional airway structure. EBioMedicine 2024; 101:105030. [PMID: 38394744 PMCID: PMC10897920 DOI: 10.1016/j.ebiom.2024.105030] [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: 11/15/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) encompasses three main phenotypes: bronchiolitis obliterans syndrome (BOS), restrictive allograft syndrome (RAS) and a Mixed phenotype combining both pathologies. How the airway structure in its entirety is affected in these phenotypes is still poorly understood. METHODS A detailed analysis of airway morphometry was applied to gain insights on the effects of airway remodelling on the distribution of alveolar ventilation in end-stage CLAD. Ex vivo whole lung μCT and tissue-core μCT scanning of six control, six BOS, three RAS and three Mixed explant lung grafts (9 male, 9 female, 2014-2021, Leuven, Belgium) were used for digital airway reconstruction and calculation of airway dimensions in relation to luminal obstructions. FINDINGS BOS and Mixed explants demonstrated airway obstructions of proximal bronchioles (starting at generation five), while RAS explants particularly had airway obstructions in the most distal bronchioles (generation >12). In BOS and Mixed explants 76% and 84% of bronchioles were obstructed, respectively, while this was 22% in RAS. Bronchiolar obstructions were mainly caused by lymphocytic inflammation of the airway wall or fibrotic remodelling, i.e. constrictive bronchiolitis. Proximal bronchiolectasis and imbalance in distal lung ventilation were present in all CLAD phenotypes and explain poor lung function and deterioration of specific lung function parameters. INTERPRETATION Alterations in the structure of conducting bronchioles revealed CLAD to affect alveolar ventilatory distribution in a regional fashion. The significance of various obstructions, particularly those associated with mucus, is highlighted. FUNDING This research was funded with the National research fund Flanders (G060322N), received by R.V.
Collapse
Affiliation(s)
- Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Gene P L Ambrocio
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Division of Pulmonary Medicine, Department of Internal Medicine, University of the Philippines - Philippine General Hospital, Manilla, The Philippines
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Saskia Bos
- Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Astrid Vermaut
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Tinne Goos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Charlotte De Fays
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Lucia Aversa
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Yousry Mohamady
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | | | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vimi Varghese
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Heart and Lung Transplant, Yashoda Hospitals, Hyderabad, India
| | - Iván Josipovic
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Matthieu N Boone
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Lieven J Dupont
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Adriana Dubbeldam
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | | | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Laurens J De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Cell Circuits in Systems Medicine of Lung Disease (Schiller Lab), Institute of Lung Health and Immunity (LHI) / Comprehensive Pneumology Centre (CPC), German Centre for Lung Research, Helmholtz Zentrum München, München, Germany
| | - John E McDonough
- Department of Medicine, McMaster University, Firestone Institute of Respiratory Health, Hamilton, Canada
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Papazoglou AS, Karagiannidis E, Liatsos A, Bompoti A, Moysidis DV, Arvanitidis C, Tsolaki F, Tsagkaropoulos S, Theocharis S, Tagarakis G, Michaelson JS, Herrmann MD. Volumetric Tissue Imaging of Surgical Tissue Specimens Using Micro-Computed Tomography: An Emerging Digital Pathology Modality for Nondestructive, Slide-Free Microscopy-Clinical Applications of Digital Pathology in 3 Dimensions. Am J Clin Pathol 2023; 159:242-254. [PMID: 36478204 DOI: 10.1093/ajcp/aqac143] [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: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Micro-computed tomography (micro-CT) is a novel, nondestructive, slide-free digital imaging modality that enables the acquisition of high-resolution, volumetric images of intact surgical tissue specimens. The aim of this systematic mapping review is to provide a comprehensive overview of the available literature on clinical applications of micro-CT tissue imaging and to assess its relevance and readiness for pathology practice. METHODS A computerized literature search was performed in the PubMed, Scopus, Web of Science, and CENTRAL databases. To gain insight into regulatory and financial considerations for performing and examining micro-CT imaging procedures in a clinical setting, additional searches were performed in medical device databases. RESULTS Our search identified 141 scientific articles published between 2000 and 2021 that described clinical applications of micro-CT tissue imaging. The number of relevant publications is progressively increasing, with the specialties of pulmonology, cardiology, otolaryngology, and oncology being most commonly concerned. The included studies were mostly performed in pathology departments. Current micro-CT devices have already been cleared for clinical use, and a Current Procedural Terminology (CPT) code exists for reimbursement of micro-CT imaging procedures. CONCLUSIONS Micro-CT tissue imaging enables accurate volumetric measurements and evaluations of entire surgical specimens at microscopic resolution across a wide range of clinical applications.
Collapse
Affiliation(s)
| | - Efstratios Karagiannidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Liatsos
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreana Bompoti
- Diagnostic Imaging, Peterborough City Hospital, North West Anglia NHS Foundation Trust, Peterborough, UK
| | - Dimitrios V Moysidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Arvanitidis
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, Heraklion, Crete, Greece.,LifeWatch ERIC, Sector II-II, Seville, Spain
| | - Fani Tsolaki
- Department of Cardiothoracic Surgery, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Stamatios Theocharis
- First Department of Pathology, National and Kapoditrian University of Athens, Athens, Greece
| | - Georgios Tagarakis
- Department of Cardiothoracic Surgery, AHEPA University Hospital, Thessaloniki, Greece
| | - James S Michaelson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Biomarkers for Chronic Lung Allograft Dysfunction: Ready for Prime Time? Transplantation 2023; 107:341-350. [PMID: 35980878 PMCID: PMC9875844 DOI: 10.1097/tp.0000000000004270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle impairing lung transplant outcome. Parallel to the better clinical identification and characterization of CLAD and CLAD phenotypes, there is an increasing urge to find adequate biomarkers that could assist in the earlier detection and differential diagnosis of CLAD phenotypes, as well as disease prognostication. The current status and state-of-the-art of biomarker research in CLAD will be discussed with a particular focus on radiological biomarkers or biomarkers found in peripheral tissue, bronchoalveolar lavage' and circulating blood' in which significant progress has been made over the last years. Ultimately, although a growing number of biomarkers are currently being embedded in the follow-up of lung transplant patients, it is clear that one size does not fit all. The future of biomarker research probably lies in the rigorous combination of clinical information with findings in tissue, bronchoalveolar lavage' or blood. Only by doing so, the ultimate goal of biomarker research can be achieved, which is the earlier identification of CLAD before its clinical manifestation. This is desperately needed to improve the prognosis of patients with CLAD after lung transplantation.
Collapse
|
4
|
Fifteen-Year Surveillance of LTR Receiving Pre-Emptive Therapy for CMV Infection: Prevention of CMV Disease and Incidence of CLAD. Microorganisms 2022; 10:microorganisms10122339. [PMID: 36557592 PMCID: PMC9788487 DOI: 10.3390/microorganisms10122339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The efficacy of pre-emptive therapy in the prevention of cytomegalovirus (CMV) disease and the potential association of CMV infection with the occurrence of chronic lung allograft dysfunction (CLAD) was evaluated in 129 lung transplant recipients receiving pre-emptive therapy based on pp65-antigenemia or CMV-DNA in the blood and in the bronchoalveolar lavage. Seventy-one (55%) patients received pre-emptive ganciclovir/valganciclovir (GCV/VGCV) for CMV infection for a median of 28 (9-191) days. Possible CMV disease occurred in six (5%) patients and was healed after the GCV/VGCV therapy. The cumulative incidence of CLAD was 38% and 54% at 5 and 10 years. Acute rejection and CMV load in the blood (but not in the lung) were independent predictors of the occurrence of CLAD. Pre-emptive therapy is highly effective in preventing CMV disease in lung recipients and does not induce a superior incidence of CLAD compared to what reported for other cohorts of patients who received an extended antiviral prophylaxis.
Collapse
|
5
|
Calabrese F, Roden AC, Pavlisko E, Lunardi F, Neil D, Adam B, Hwang D, Goddard M, Berry GJ, Ivanovic M, Thüsen JVD, Gibault L, Lin CY, Wassilew K, Glass C, Westall G, Zeevi A, Levine DJ, Roux A. LUNG ALLOGRAFT STANDARDIZED HISTOLOGICAL ANALYSIS (LASHA) TEMPLATE: A RESEARCH CONSENSUS PROPOSAL. J Heart Lung Transplant 2022; 41:1487-1500. [DOI: 10.1016/j.healun.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
|
6
|
Verleden SE, Braubach P, Werlein C, Plucinski E, Kuhnel MP, Snoeckx A, El Addouli H, Welte T, Haverich A, Laenger FP, Dettmer S, Pauwels P, Verplancke V, Van Schil PE, Lapperre T, Kwakkel-Van-Erp JM, Ackermann M, Hendriks JMH, Jonigk D. From Macroscopy to Ultrastructure: An Integrative Approach to Pulmonary Pathology. Front Med (Lausanne) 2022; 9:859337. [PMID: 35372395 PMCID: PMC8965844 DOI: 10.3389/fmed.2022.859337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022] Open
Abstract
Pathology and radiology are complimentary tools, and their joint application is often crucial in obtaining an accurate diagnosis in non-neoplastic pulmonary diseases. However, both come with significant limitations of their own: Computed Tomography (CT) can only visualize larger structures due to its inherent–relatively–poor resolution, while (histo) pathology is often limited due to small sample size and sampling error and only allows for a 2D investigation. An innovative approach of inflating whole lung specimens and subjecting these subsequently to CT and whole lung microCT allows for an accurate matching of CT-imaging and histopathology data of exactly the same areas. Systematic application of this approach allows for a more targeted assessment of localized disease extent and more specifically can be used to investigate early mechanisms of lung diseases on a morphological and molecular level. Therefore, this technique is suitable to selectively investigate changes in the large and small airways, as well as the pulmonary arteries, veins and capillaries in relation to the disease extent in the same lung specimen. In this perspective we provide an overview of the different strategies that are currently being used, as well as how this growing field could further evolve.
Collapse
Affiliation(s)
- Stijn E. Verleden
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), Antwerp University, Antwerp, Belgium
- Division of Pneumology, University Hospital Antwerp, Edegem, Belgium
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, Edegem, Belgium
| | - Peter Braubach
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Edith Plucinski
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Mark P. Kuhnel
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Annemiek Snoeckx
- Division of Radiology, University Hospital Antwerp and University of Antwerp, Edegem, Belgium
| | - Haroun El Addouli
- Division of Radiology, University Hospital Antwerp and University of Antwerp, Edegem, Belgium
| | - Tobias Welte
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Division of Pneumology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Division of Thoracic Surgery, Hannover Medical School, Hannover, Germany
| | - Florian P. Laenger
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Sabine Dettmer
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Department of Radiology, Hannover Medical School, Hannover, Germany
| | - Patrick Pauwels
- Division of Pathology, University Hospital Antwerp, Edegem, Belgium
| | | | - Paul E. Van Schil
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), Antwerp University, Antwerp, Belgium
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, Edegem, Belgium
| | - Therese Lapperre
- Division of Pneumology, University Hospital Antwerp, Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Antwerp University, Antwerp, Belgium
| | - Johanna M. Kwakkel-Van-Erp
- Division of Pneumology, University Hospital Antwerp, Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Antwerp University, Antwerp, Belgium
| | - Maximilian Ackermann
- Institute of Pathology and Department of Molecular Pathology, Helios University Clinic Wuppertal, University of Witten-Herdecke, Witten, Germany
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jeroen M. H. Hendriks
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), Antwerp University, Antwerp, Belgium
- Department of Thoracic and Vascular Surgery, University Hospital Antwerp, Edegem, Belgium
| | - Danny Jonigk
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Institute for Pathology, Hannover Medical School, Hannover, Germany
- *Correspondence: Danny Jonigk
| |
Collapse
|
7
|
Ram S, Verleden SE, Bell AJ, Hoff BA, Labaki WW, Murray S, Vanaudenaerde BM, Vos R, Verleden GM, Kazerooni EA, Galbán S, Hatt CR, Han MK, Lama VN, Galbán CJ. Quantitative CT Correlates with Local Inflammation in Lung of Patients with Subtypes of Chronic Lung Allograft Dysfunction. Cells 2022; 11:699. [PMID: 35203345 PMCID: PMC8870691 DOI: 10.3390/cells11040699] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/03/2023] Open
Abstract
Chronic rejection of lung allografts has two major subtypes, bronchiolitis obliterans syndrome (BOS) and restrictive allograft syndrome (RAS), which present radiologically either as air trapping with small airways disease or with persistent pleuroparenchymal opacities. Parametric response mapping (PRM), a computed tomography (CT) methodology, has been demonstrated as an objective readout of BOS and RAS and bears prognostic importance, but has yet to be correlated to biological measures. Using a topological technique, we evaluate the distribution and arrangement of PRM-derived classifications of pulmonary abnormalities from lung transplant recipients undergoing redo-transplantation for end-stage BOS (N = 6) or RAS (N = 6). Topological metrics were determined from each PRM classification and compared to structural and biological markers determined from microCT and histopathology of lung core samples. Whole-lung measurements of PRM-defined functional small airways disease (fSAD), which serves as a readout of BOS, were significantly elevated in BOS versus RAS patients (p = 0.01). At the core-level, PRM-defined parenchymal disease, a potential readout of RAS, was found to correlate to neutrophil and collagen I levels (p < 0.05). We demonstrate the relationship of structural and biological markers to the CT-based distribution and arrangement of PRM-derived readouts of BOS and RAS.
Collapse
Affiliation(s)
- Sundaresh Ram
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| | - Stijn E. Verleden
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (B.M.V.); (R.V.); (G.M.V.)
| | - Alexander J. Bell
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| | - Benjamin A. Hoff
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (W.W.L.); (M.K.H.); (V.N.L.)
| | - Susan Murray
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Bart M. Vanaudenaerde
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (B.M.V.); (R.V.); (G.M.V.)
| | - Robin Vos
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (B.M.V.); (R.V.); (G.M.V.)
- Respiratory Division, University Hospital Leuven, 3000 Leuven, Belgium
| | - Geert M. Verleden
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (B.M.V.); (R.V.); (G.M.V.)
- Respiratory Division, University Hospital Leuven, 3000 Leuven, Belgium
| | - Ella A. Kazerooni
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| | - Stefanie Galbán
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| | | | - Meilan K. Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (W.W.L.); (M.K.H.); (V.N.L.)
| | - Vibha N. Lama
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (W.W.L.); (M.K.H.); (V.N.L.)
| | - Craig J. Galbán
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (S.R.); (A.J.B.); (B.A.H.); (E.A.K.); (S.G.)
| |
Collapse
|
8
|
Bompoti A, Papazoglou AS, Moysidis DV, Otountzidis N, Karagiannidis E, Stalikas N, Panteris E, Ganesh V, Sanctuary T, Arvanitidis C, Sianos G, Michaelson JS, Herrmann MD. Volumetric Imaging of Lung Tissue at Micrometer Resolution: Clinical Applications of Micro-CT for the Diagnosis of Pulmonary Diseases. Diagnostics (Basel) 2021; 11:diagnostics11112075. [PMID: 34829422 PMCID: PMC8625264 DOI: 10.3390/diagnostics11112075] [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: 09/17/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Micro-computed tomography (micro-CT) is a promising novel medical imaging modality that allows for non-destructive volumetric imaging of surgical tissue specimens at high spatial resolution. The aim of this study is to provide a comprehensive assessment of the clinical applications of micro-CT for the tissue-based diagnosis of lung diseases. This scoping review was conducted in accordance with the PRISMA Extension for Scoping Reviews, aiming to include every clinical study reporting on micro-CT imaging of human lung tissues. A literature search yielded 570 candidate articles, out of which 37 were finally included in the review. Of the selected studies, 9 studies explored via micro-CT imaging the morphology and anatomy of normal human lung tissue; 21 studies investigated microanatomic pulmonary alterations due to obstructive or restrictive lung diseases, such as chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and cystic fibrosis; and 7 studies examined the utility of micro-CT imaging in assessing lung cancer lesions (n = 4) or in transplantation-related pulmonary alterations (n = 3). The selected studies reported that micro-CT could successfully detect several lung diseases providing three-dimensional images of greater detail and resolution than routine optical slide microscopy, and could additionally provide valuable volumetric insight in both restrictive and obstructive lung diseases. In conclusion, micro-CT-based volumetric measurements and qualitative evaluations of pulmonary tissue structures can be utilized for the clinical management of a variety of lung diseases. With micro-CT devices becoming more accessible, the technology has the potential to establish itself as a core diagnostic imaging modality in pathology and to enable integrated histopathologic and radiologic assessment of lung cancer and other lung diseases.
Collapse
Affiliation(s)
- Andreana Bompoti
- Department of Radiology, Peterborough City Hospital, Northwest Anglia NHS Foundation Trust, Peterborough PE3 9GZ, UK;
| | - Andreas S. Papazoglou
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - Dimitrios V. Moysidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - Nikolaos Otountzidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - Efstratios Karagiannidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - Nikolaos Stalikas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - Eleftherios Panteris
- Biomic_AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd., P.O. Box 8318, GR 57001 Thessaloniki, Greece;
| | | | - Thomas Sanctuary
- Respiratory Department, Medway NHS Foundation Trust, Kent ME7 5NY, UK;
| | - Christos Arvanitidis
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), 70013 Heraklion, Greece;
- LifeWatch ERIC, Sector II-II, Plaza de España, 41071 Seville, Spain
| | - Georgios Sianos
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (A.S.P.); (D.V.M.); (N.O.); (E.K.); (N.S.); (G.S.)
| | - James S. Michaelson
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Markus D. Herrmann
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
- Correspondence: ; Tel.: +6-17-724-1896
| |
Collapse
|
9
|
Müller C, Rosmark O, Åhrman E, Brunnström H, Wassilew K, Nybom A, Michaliková B, Larsson H, Eriksson LT, Schultz HH, Perch M, Malmström J, Wigén J, Iversen M, Westergren-Thorsson G. Protein Signatures of Remodeled Airways in Transplanted Lungs with Bronchiolitis Obliterans Syndrome Obtained Using Laser-Capture Microdissection. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1398-1411. [PMID: 34111430 DOI: 10.1016/j.ajpath.2021.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 10/25/2022]
Abstract
Bronchiolitis obliterans syndrome, a common form of chronic lung allograft dysfunction, is the major limitation to long-term survival after lung transplantation. The histologic correlate is progressive, fibrotic occlusion of small airways, obliterative bronchiolitis lesions, which ultimately lead to organ failure. The molecular composition of these lesions is unknown. In this sutdy, the protein composition of the lesions in explanted lungs from four end-stage bronchiolitis obliterans syndrome patients was analyzed using laser-capture microdissection and optimized sample preparation protocols for mass spectrometry. Immunohistochemistry and immunofluorescence were used to determine the spatial distribution of commonly identified proteins on the tissue level, and protein signatures for 14 obliterative bronchiolitis lesions were established. A set of 39 proteins, identified in >75% of lesions, included distinct structural proteins (collagen types IV and VI) and cellular components (actins, vimentin, and tryptase). Each respective lesion exhibited a unique composition of proteins (on average, n = 66 proteins), thereby mirroring the morphologic variation of the lesions. Antibody-based staining confirmed these mass spectrometry-based findings. The 14 analyzed obliterative bronchiolitis lesions showed variations in their protein content, but also common features. This study provides molecular and morphologic insights into the development of chronic rejection after lung transplantation. The protein patterns in the lesions were correlated to pathways of extracellular matrix organization, tissue development, and wound healing processes.
Collapse
Affiliation(s)
- Catharina Müller
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oskar Rosmark
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Emma Åhrman
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden; Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Division of Laboratory Medicine, Department of Genetics and Pathology, Region Skåne, Lund, Sweden
| | - Katharina Wassilew
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Annika Nybom
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Barbora Michaliková
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hillevi Larsson
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Leif T Eriksson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden; Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Hans H Schultz
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jenny Wigén
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Martin Iversen
- Department of Cardiology, Section for Lung Transplantation, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
10
|
Brugiere O, Verleden SE. Putting the spotlight on macrophage-derived cathepsin in the pathophysiology of obliterative bronchiolitis. Eur Respir J 2021; 57:57/5/2004607. [PMID: 33985982 DOI: 10.1183/13993003.04607-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/19/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Olivier Brugiere
- Lung Transplant Dept, Foch Hospital, Suresnes, France .,Inserm UMR S 1152, Physiopathologie et Epidémiologie des Maladies Respiratoires, Paris, France
| | | |
Collapse
|
11
|
Hage CA, Klesney-Tait J, Wille K, Arcasoy S, Yung G, Hertz M, Chan KM, Morrell M, Goldberg H, Vedantham S, Derfler MC, Commean P, Berman K, Spitznagel E, Atkinson J, Despotis G. Extracorporeal photopheresis to attenuate decline in lung function due to refractory obstructive allograft dysfunction. Transfus Med 2021; 31:292-302. [PMID: 33955079 PMCID: PMC8453798 DOI: 10.1111/tme.12779] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/18/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND This study was designed to prospectively evaluate the efficacy of extracorporeal photopheresis (ECP) to attenuate the rate of decline of FEV1 in lung transplant recipients with refractory bronchiolitis obliterans. Due to an observed higher than expected early mortality, a preliminary analysis was performed. STUDY DESIGN AND METHODS Subjects from 10 lung transplant centres were assigned to ECP treatment or to observation based on spirometric criteria, with potential crossover for those under observation. The primary endpoint of this study was to assess response to ECP (i.e., greater than a 50% decrease in the rate of FEV1 decline) before and 6 months after initiation of ECP. Mortality was also evaluated 6 and 12 months after enrolment as a secondary endpoint. RESULTS Of 44 enrolled subjects, 31 were assigned to ECP treatment while 13 were initially assigned to observation on a non-random basis using specific spirometric inclusion criteria (seven of the observation patients subsequently crossed over to receive ECP). Of evaluable patients, 95% of patients initially assigned to treatment responded to ECP with rates of FEV1 decline that were reduced by 93% in evaluable ECP-treated patients. Mortality rates (percentages) at 6 and 12 months after enrolment was 32% and 41%, respectively. The most common (92%) primary cause of death was respiratory or graft failure. Significantly (p = 0.002) higher rates of FEV1 decline were observed in the non-survivors (-212 ± 177 ml/month) when compared to the survivors (-95 ± 117 ml/month) 12 months after enrolment. In addition, 18 patients with bronchiolitis obliterans syndrome (BOS) diagnosis within 6 months of enrolment had lost 38% of their baseline lung function at BOS diagnosis and 50% of their lung function at enrolment. CONCLUSIONS These analyses suggest that earlier detection and treatment of BOS should be considered to appreciate improved outcomes with ECP.
Collapse
Affiliation(s)
| | - Chadi A Hage
- Department of Medicine, Division of Pulmonology, Indiana University, Bloomington, Indiana, USA
| | - Julia Klesney-Tait
- Department of Medicine, Division of Pulmonology, University of Iowa, Iowa City, Iowa, USA
| | - Keith Wille
- Department of Medicine, Division of Pulmonology, University of Alabama, Tuscaloosa, Alabama, USA
| | - Selim Arcasoy
- Department of Medicine, Division of Pulmonology, Columbia University of Alabama, Orange Beach, Alabama, USA
| | - Gordon Yung
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, California, USA
| | - Marshall Hertz
- Department of Medicine, Division of Pulmonology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kevin M Chan
- Department of Medicine, Division of Pulmonology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matt Morrell
- Department of Medicine, Division of Pulmonology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hilary Goldberg
- Harvard Medical School, Department of Medicine, Division of Pulmonology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Suresh Vedantham
- Clinical Coordinating Center, Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Mary Clare Derfler
- Clinical Coordinating Center, Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Paul Commean
- Data Coordinating Center, Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Keith Berman
- Health Research Associates, Mountlake Terrace, Washington, USA
| | - Ed Spitznagel
- Department of Mathematics, Washington University, St. Louis, Missouri, USA
| | - Jeff Atkinson
- Department of Internal Medicine, Division of Pulmonary Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - George Despotis
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Department of Anesthesiology, Division of Cardiothoracic Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Klouda T, Vargas SO, Midyat L. Restrictive allograft syndrome after lung transplantation. Pediatr Transplant 2021; 25:e14000. [PMID: 33728767 DOI: 10.1111/petr.14000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/07/2021] [Accepted: 02/22/2021] [Indexed: 11/29/2022]
Abstract
Despite recent advances over the past decade in lung transplantation including improved surgical technique and immunotherapy, the diagnosis and treatment of chronic lung allograft dysfunction remains a significant barrier to recipient survival. Aside from bronchiolitis obliterans syndrome, a restrictive phenotype called restrictive allograft syndrome has recently been recognized and affects up to 35% of all patients with CLAD. The main characteristics of RAS include a persistent and unexplained decline in lung function compared to baseline and persistent parenchymal infiltrates on imaging. The median survival after diagnosis of RAS is 6 to 18 months, significantly shorter than other forms of CLAD. Treatment options are limited, as therapies used for BOS are typically ineffective at halting disease progression. Specific medications such as fibrinolytics are lacking large, multicenter prospective studies. In this manuscript, we discuss the definition, mechanism, and characteristics of RAS while highlighting the similarities and differences between other forms of CLAD. We also review the diagnoses along with current and potential treatment options that are available for patients. Finally, we discuss the existing knowledge gaps and areas for future research to improve patient outcomes and understanding of RAS.
Collapse
Affiliation(s)
- Timothy Klouda
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Levent Midyat
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Pezzuto F, Lunardi F, Vadori M, Zampieri D, Casiraghi F, Azzollini N, Vuljan SE, Mammana M, Vedovelli L, Schiavon M, Gregori D, Cozzi E, Rea F, Calabrese F. Chronic lung allograft pathology lesions in two rat strain combinations. J Thorac Dis 2021; 13:2833-2843. [PMID: 34164175 PMCID: PMC8182524 DOI: 10.21037/jtd-20-3415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Chronic lung allograft dysfunction remains an obstacle to long-term survival after lung transplantation. Two phenotypes have been described: obliterative bronchiolitis and restrictive allograft syndrome. Preclinical models are essential to analyze chronic lung allograft dysfunction pathophysiology. Methods Orthotopic lung transplants from 38 Lewis into Fischer 344 (Lew→F344) and 67 Brown-Norway into Lewis (BN→Lew) rats were performed in our center in the last decade. We carefully reviewed and quantified all grafts with chronic rejection (40 cases) (18 Lew→F344, 22 BN→Lew) with the aim to investigate if histological changes of chronic lung allograft dysfunction could be also detected in rat grafts. Results All animals showed human reminiscent histological lesions. Early chronic rejection lesions were detected in BN→Lew. End-stage chronic rejection with features of obliterative bronchiolitis was observed in 33% of Lew→F344; end-stage with restrictive allograft syndrome chronic rejection in 67% and 80% of Lew→F344 and BN→Lew, respectively. BN→Lew showed higher grades of endotheliitis, vascular fibrosis, and lower grades of lymphoid aggregates than Lew→F344 (P=0.007, P=0.043, P=0.004, respectively). Conclusions Chronic rejection lesions in rat lung allografts mimic those in humans. The frequent occurrence of restrictive allograft syndrome-like lesions in BN→Lew may be related to a higher degree of mismatch in this strain combination. These animal models could allow future mechanistic studies to better understand chronic lung allograft dysfunction pathogenesis.
Collapse
Affiliation(s)
- Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Davide Zampieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | | | - Nadia Azzollini
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Mammana
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Emanuele Cozzi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| |
Collapse
|
14
|
Brun AL, Chabi ML, Picard C, Mellot F, Grenier PA. Lung Transplantation: CT Assessment of Chronic Lung Allograft Dysfunction (CLAD). Diagnostics (Basel) 2021; 11:diagnostics11050817. [PMID: 33946544 PMCID: PMC8147203 DOI: 10.3390/diagnostics11050817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/05/2023] Open
Abstract
Chronic lung allograft rejection remains one of the major causes of morbi-mortality after lung transplantation. The term Chronic Lung Allograft Dysfunction (CLAD) has been proposed to describe the different processes that lead to a significant and persistent deterioration in lung function without identifiable causes. The two main phenotypes of CLAD are Bronchiolitis Obliterans Syndrome (BOS) and Restrictive Allograft Syndrome (RAS), each of them characterized by particular functional and imaging features. These entities can be associated (mixed phenotype) or switched from one to the other. If CLAD remains a clinical diagnosis based on spirometry, computed tomography (CT) scan plays an important role in the diagnosis and follow-up of CLAD patients, to exclude identifiable causes of functional decline when CLAD is first suspected, to detect early abnormalities that can precede the diagnosis of CLAD (particularly RAS), to differentiate between the obstructive and restrictive phenotypes, and to detect exacerbations and evolution from one phenotype to the other. Recognition of early signs of rejection is crucial for better understanding of physiopathologic pathways and optimal management of patients.
Collapse
Affiliation(s)
- Anne-Laure Brun
- Radiology Department, Hôpital Foch, 92150 Suresnes, France; (M.-L.C.); (F.M.)
- Correspondence: (A.-L.B.); (P.A.G.)
| | - Marie-Laure Chabi
- Radiology Department, Hôpital Foch, 92150 Suresnes, France; (M.-L.C.); (F.M.)
| | - Clément Picard
- Respiratory Department, Hôpital Foch, 92150 Suresnes, France;
| | - François Mellot
- Radiology Department, Hôpital Foch, 92150 Suresnes, France; (M.-L.C.); (F.M.)
| | - Philippe A. Grenier
- Department of Clinical Research and Innovation, Hôpital Foch, 92150 Suresnes, France
- Correspondence: (A.-L.B.); (P.A.G.)
| |
Collapse
|
15
|
Byrne D, Nador RG, English JC, Yee J, Levy R, Bergeron C, Swiston JR, Mets OM, Muller NL, Bilawich AM. Chronic Lung Allograft Dysfunction: Review of CT and Pathologic Findings. Radiol Cardiothorac Imaging 2021; 3:e200314. [PMID: 33778654 PMCID: PMC7978021 DOI: 10.1148/ryct.2021200314] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 04/14/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is the most common cause of mortality in lung transplant recipients after the 1st year of transplantation. CLAD has traditionally been classified into two distinct obstructive and restrictive forms: bronchiolitis obliterans syndrome and restrictive allograft syndrome. However, CLAD may manifest with a spectrum of imaging and pathologic findings and a combination of obstructive and restrictive physiologic abnormalities. Although the initial CT manifestations of CLAD may be nonspecific, the progression of findings at follow-up should signal the possibility of CLAD and may be present on imaging studies prior to the development of functional abnormalities of the lung allograft. This review encompasses the evolution of CT findings in CLAD, with emphasis on the underlying pathogenesis and pathologic condition, to enhance understanding of imaging findings. The purpose of this article is to familiarize the radiologist with the initial and follow-up CT findings of the obstructive, restrictive, and mixed forms of CLAD, for which early diagnosis and treatment may result in improved survival. Supplemental material is available for this article. © RSNA, 2021.
Collapse
|
16
|
Werlein C, Ackermann M, Hoffmann TL, Laenger F, Jonigk D. [Fibrotic remodeling of the lung following lung and stem-cell transplantation]. DER PATHOLOGE 2021; 42:17-24. [PMID: 33416936 DOI: 10.1007/s00292-020-00898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
Transplantation of solid organs and hematopoietic stem cells represents an important therapeutic option for a variety of end-stage pulmonary diseases, aggressive hematopoietic neoplasms, or severe immunodeficiencies. Although the overall survival following transplantation has generally improved over recent decades, long-time survival of lung and stem-cell transplant recipients is still alarmingly low with an average 5‑year survival rate of only 50-60%. Chronic allo-immunoreactions in general and pulmonary allo-immunoreactions with subsequent fibrosis in particular are major reasons for this poor outcome. Comparable patterns of fibrotic lung remodeling are observed following both lung and hematopoietic stem-cell transplantation. Besides the meanwhile well-established obliterative and functionally obstructive remodeling of the small airways - obliterative bronchiolitis - a specific restrictive subform of fibrosis, namely alveolar fibroelastosis, has been identified. Despite their crucial impact on patient outcome, both entities can be very challenging to detect by conventional histopathological analysis. Their underlying mechanisms are considered overreaching aberrant repair attempts to acute lung injuries with overactivation of (myo-) fibroblasts and excessive and irreversible deposition of extracellular matrix. Of note, the underlying molecular mechanisms are widely divergent between these two morphological entities and are independent of the underlying clinical setting.Further comprehensive investigations of these fibrotic alterations are key to the development of much-needed predictive diagnostics and curative concepts, considering the high mortality of pulmonary fibrosis following transplantation.
Collapse
Affiliation(s)
- Christopher Werlein
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland
| | - Max Ackermann
- Institut für Pathologie und Molekularpathologie, Helios Universitätsklinikum Wuppertal, Universität Witten-Herdecke, Wuppertal, Deutschland.,Institut für Funktionelle und Klinische Anatomie, Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Mainz, Deutschland
| | - Thia Leandra Hoffmann
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland
| | - Florian Laenger
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Standort Hannover, Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland
| | - Danny Jonigk
- Institut für Pathologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, OE 5110, 30625, Hannover, Deutschland. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Standort Hannover, Deutsches Zentrum für Lungenforschung (DZL), Hannover, Deutschland.
| |
Collapse
|
17
|
Werlein C, Seidel A, Warnecke G, Gottlieb J, Laenger F, Jonigk D. Lung Transplant Pathology: An Overview on Current Entities and Procedures. Surg Pathol Clin 2020; 13:119-140. [PMID: 32005428 DOI: 10.1016/j.path.2019.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alloimmune reactions are, besides various infections, the major cause for impaired lung allograft function following transplant. Acute cellular rejection is not only a major trigger of acute allograft failure but also contributes to development of chronic lung allograft dysfunction. Analogous to other solid organ transplants, acute antibody-mediated rejection has become a recognized entity in lung transplant pathology. Adequate sensitivity and specificity in the diagnosis of alloimmune reactions in the lung can only be achieved by synoptic analysis of histopathologic, clinical, and radiological findings together with serologic and microbiologic findings.
Collapse
Affiliation(s)
- Christopher Werlein
- Institute for Pathology, OE 5110, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany.
| | - Allison Seidel
- Institute for Pathology, OE 5110, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)
| | - Gregor Warnecke
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH); Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, OE6210, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Jens Gottlieb
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH); Department of Pneumology, OE6210, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Florian Laenger
- Institute for Pathology, OE 5110, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)
| | - Danny Jonigk
- Institute for Pathology, OE 5110, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)
| |
Collapse
|
18
|
Verleden SE, Von der Thüsen J, Roux A, Brouwers ES, Braubach P, Kuehnel M, Laenger F, Jonigk D. When tissue is the issue: A histological review of chronic lung allograft dysfunction. Am J Transplant 2020; 20:2644-2651. [PMID: 32185874 DOI: 10.1111/ajt.15864] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 01/25/2023]
Abstract
Although chronic lung allograft dysfunction (CLAD) remains the major life-limiting factor following lung transplantation, much of its pathophysiology remains unknown. The discovery that CLAD can manifest both clinically and morphologically in vastly different ways led to the definition of distinct subtypes of CLAD. In this review, recent advances in our understanding of the pathophysiological mechanisms of the different phenotypes of CLAD will be discussed with a particular focus on tissue-based and molecular studies. An overview of the current knowledge on the mechanisms of the airway-centered bronchiolitis obliterans syndrome, as well as the airway and alveolar injuries in the restrictive allograft syndrome and also the vascular compartment in chronic antibody-mediated rejection is provided. Specific attention is also given to morphological and molecular markers for early CLAD diagnosis or histological changes associated with subsequent CLAD development. Evidence for a possible overlap between different forms of CLAD is presented and discussed. In the end, "tissue remains the (main) issue," as we are still limited in our knowledge about the actual triggers and specific mechanisms of all late forms of posttransplant graft failure, a shortcoming that needs to be addressed in order to further improve the outcome of lung transplant recipients.
Collapse
Affiliation(s)
- Stijn E Verleden
- Lab of Respiratory Diseases, BREATH, Department of CHROMETA, KU Leuven, Leuven, Belgium.,Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany
| | - Jan Von der Thüsen
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Antoine Roux
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - Emily S Brouwers
- Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), The German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Hannover Medical School (MHH), Hannover, Germany
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), The German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Hannover Medical School (MHH), Hannover, Germany
| | - Mark Kuehnel
- Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), The German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Hannover Medical School (MHH), Hannover, Germany
| | - Florian Laenger
- Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), The German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Hannover Medical School (MHH), Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School (MHH), Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), The German Center for Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
19
|
Verleden SE, Vanstapel A, De Sadeleer L, Weynand B, Boone M, Verbeken E, Piloni D, Van Raemdonck D, Ackermann M, Jonigk DD, Verschakelen J, Wuyts WA. Quantitative analysis of airway obstruction in lymphangioleiomyomatosis. Eur Respir J 2020; 56:13993003.01965-2019. [PMID: 32108050 PMCID: PMC7330132 DOI: 10.1183/13993003.01965-2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/03/2020] [Indexed: 02/02/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare, cystic lung disease with progressive pulmonary function loss caused by progressively proliferating LAM cells. The degree of airway obstruction has not been well investigated within the pathogenesis of LAM.Using a combination of ex vivo computed tomography (CT), microCT and histology, the site and nature of airway obstruction in LAM explant lungs was compared with matched control lungs (n=5 each). The total number of airways per generation, total airway counts, terminal bronchioles number and surface density were compared in LAM versus control.Ex vivo CT analysis demonstrated a reduced number of airways from generation 7 on (p<0.0001) in LAM compared with control, whereas whole-lung microCT analysis confirmed the three- to four-fold reduction in the number of airways. Specimen microCT analysis further demonstrated a four-fold decrease in the number of terminal bronchioles (p=0.0079) and a decreased surface density (p=0.0079). Serial microCT and histology images directly showed the loss of functional airways by collapse of airways on the cysts and filling of the airway by exudate.LAM lungs show a three- to four-fold decrease in the number of (small) airways, caused by cystic destruction which is the likely culprit for the progressive loss of pulmonary function.
Collapse
Affiliation(s)
- Stijn E Verleden
- Respiratory Diseases, Dept of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium .,Both authors contributed equally
| | - Arno Vanstapel
- Respiratory Diseases, Dept of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium.,Both authors contributed equally
| | - Laurens De Sadeleer
- Respiratory Diseases, Dept of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | | | - Matthieu Boone
- Dept of Physics and Astronomy, Radiation Physics-Centre for X-ray Tomography, Ghent University, Ghent, Belgium
| | | | - Davide Piloni
- The Respiratory Disease Unit, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Dirk Van Raemdonck
- Thoracic Surgery, Dept of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Institute of Pathology and Molecular Pathology, Helios University Hospital Wuppertal, Witten-Herdecke University, Wuppertal, Germany
| | - Danny D Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | - Wim A Wuyts
- Respiratory Diseases, Dept of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Verleden SE, Tanabe N, McDonough JE, Vasilescu DM, Xu F, Wuyts WA, Piloni D, De Sadeleer L, Willems S, Mai C, Hostens J, Cooper JD, Verbeken EK, Verschakelen J, Galban CJ, Van Raemdonck DE, Colby TV, Decramer M, Verleden GM, Kaminski N, Hackett TL, Vanaudenaerde BM, Hogg JC. Small airways pathology in idiopathic pulmonary fibrosis: a retrospective cohort study. THE LANCET. RESPIRATORY MEDICINE 2020; 8:573-584. [PMID: 32061334 PMCID: PMC7292784 DOI: 10.1016/s2213-2600(19)30356-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND The observation that patients with idiopathic pulmonary fibrosis (IPF) can have higher than normal expiratory flow rates at low lung volumes led to the conclusion that the airways are spared in IPF. This study aimed to re-examine the hypothesis that airways are spared in IPF using a multiresolution imaging protocol that combines multidetector CT (MDCT), with micro-CT and histology. METHODS This was a retrospective cohort study comparing explanted lungs from patients with severe IPF treated by lung transplantation with a cohort of unused donor (control) lungs. The donor control lungs had no known lung disease, comorbidities, or structural lung injury, and were deemed appropriate for transplantation on review of the clinical files. The diagnosis of IPF in the lungs from patients was established by a multidisciplinary consensus committee according to existing guidelines, and was confirmed by video-assisted thoracic surgical biopsy or by pathological examination of the contralateral lung. The control and IPF groups were matched for age, sex, height, and bodyweight. Samples of lung tissue were compared using the multiresolution imaging approach: a cascade of clinical MDCT, micro-CT, and histological imaging. We did two experiments: in experiment 1, all the lungs were randomly sampled; in experiment 2, samples were selected from regions of minimal and established fibrosis. The patients and donors were recruited from the Katholieke Universiteit Leuven (Leuven, Belgium) and the University of Pennsylvania Hospital (Philadelphia, PA, USA). The study took place at the Katholieke Universiteit Leuven, and the University of British Columbia (Vancouver, BC, Canada). FINDINGS Between Oct 5, 2009, and July 22, 2016, explanted lungs from patients with severe IPF (n=11), were compared with a cohort of unused donor (control) lungs (n=10), providing 240 samples of lung tissue for comparison using the multiresolution imaging approach. The MDCT specimen scans show that the number of visible airways located between the ninth generation (control 69 [SD 22] versus patients with IPF 105 [33], p=0·0023) and 14th generation (control 9 [6] versus patients with IPF 49 [28], p<0·0001) of airway branching are increased in patients with IPF, which we show by micro-CT is due to thickening of their walls and distortion of their lumens. The micro-CT analysis showed that compared with healthy (control) lung anatomy (mean 5·6 terminal bronchioles per mL [SD 1·6]), minimal fibrosis in IPF tissue was associated with a 57% loss of the terminal bronchioles (mean 2·4 terminal bronchioles per mL [SD 1·0]; p<0·0001), the appearance of fibroblastic foci, and infiltration of the tissue by inflammatory immune cells capable of forming lymphoid follicles. Established fibrosis in IPF tissue had a similar reduction (66%) in the number of terminal bronchioles (mean 1·9 terminal bronchioles per mL [SD 1·4]; p<0·0001) and was dominated by increased airspace size, Ashcroft fibrosis score, and volume fractions of tissue and collagen. INTERPRETATION Small airways disease is a feature of IPF, with significant loss of terminal bronchioles occuring within regions of minimal fibrosis. On the basis of these findings, we postulate that the small airways could become a potential therapeutic target in IPF. FUNDING Katholieke Universiteit Leuven, US National Institutes of Health, BC Lung Association, and Genentech.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Naoya Tanabe
- University of British Columbia, Department of Pathology and Center for Heart and Lung Innovation at St Paul's Hospital, Vancouver, BC, Canada; Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - John E McDonough
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Dragoş M Vasilescu
- University of British Columbia, Department of Pathology and Center for Heart and Lung Innovation at St Paul's Hospital, Vancouver, BC, Canada
| | - Feng Xu
- University of British Columbia, Department of Pathology and Center for Heart and Lung Innovation at St Paul's Hospital, Vancouver, BC, Canada
| | - Wim A Wuyts
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Davide Piloni
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium; The Respiratory Disease Unit, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Laurens De Sadeleer
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Stijn Willems
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Cindy Mai
- Department of Radiology, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | | | - Joel D Cooper
- Department of Thoracic Surgery University of Pennsylvania, Philadelphia, PA, USA
| | - Erik K Verbeken
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Johny Verschakelen
- Department of Radiology, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Craig J Galban
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Dirk E Van Raemdonck
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Thomas V Colby
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Marc Decramer
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University, New Haven, CT, USA
| | - Tillie-Louise Hackett
- University of British Columbia, Department of Pathology and Center for Heart and Lung Innovation at St Paul's Hospital, Vancouver, BC, Canada
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Katholieke Universiteit Leuven, University Hospitals Leuven, Leuven, Belgium
| | - James C Hogg
- University of British Columbia, Department of Pathology and Center for Heart and Lung Innovation at St Paul's Hospital, Vancouver, BC, Canada.
| |
Collapse
|
21
|
Combs MP, Xia M, Wheeler DS, Belloli EA, Walker NM, Braeuer RR, Lyu DM, Murray S, Lama VN. Fibroproliferation in chronic lung allograft dysfunction: Association of mesenchymal cells in bronchoalveolar lavage with phenotypes and survival. J Heart Lung Transplant 2020; 39:815-823. [PMID: 32360292 DOI: 10.1016/j.healun.2020.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD), the primary cause of poor outcome after lung transplantation, arises from fibrotic remodeling of the allograft and presents as diverse clinical phenotypes with variable courses. Here, we investigate whether bronchoalveolar lavage (BAL) mesenchymal cell activity at CLAD onset can inform regarding disease phenotype, progression, and survival. METHODS Mesenchymal cell colony-forming units (CFUs) were measured in BAL obtained at CLAD onset (n = 77) and CLAD-free time post-transplant matched controls (n = 77). CFU counts were compared using Wilcoxon's rank-sum test. Cox proportional hazards and restricted means models were utilized to investigate post-CLAD survival. RESULTS Higher mesenchymal CFU counts were noted in BAL at the time of CLAD onset than in CLAD-free controls. Patients with restrictive allograft syndrome had higher BAL mesenchymal CFU count at CLAD onset than patients with bronchiolitis obliterans syndrome (p = 0.011). Patients with high mesenchymal CFU counts (≥10) at CLAD onset had worse outcomes than those with low (<10) CFU counts, with shorter average survival (2.64 years vs 4.25 years; p = 0.027) and shorter progression-free survival, defined as time to developing either CLAD Stage 3 or death (0.97 years vs 2.70 years; p < 0.001). High CFU count remained predictive of decreased overall survival and progression-free survival after accounting for the CLAD phenotype and other clinical factors in multivariable analysis. CONCLUSIONS Fulminant fibroproliferation with higher mesenchymal CFU counts in BAL is noted in restrictive allograft syndrome and is independently associated with poor survival after CLAD onset.
Collapse
Affiliation(s)
- Michael P Combs
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Meng Xia
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - David S Wheeler
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth A Belloli
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Natalie M Walker
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Russell R Braeuer
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Dennis M Lyu
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan
| | - Susan Murray
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Vibha N Lama
- Department of Internal Medicine, Division of Pulmonary & Critical Care, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
22
|
Verleden SE, Von Der Thüsen J, Van Herck A, Weynand B, Verbeken E, Verschakelen J, Dubbeldam A, Vanaudenaerde BM, Vos R, Verleden GM. Identification and characterization of chronic lung allograft dysfunction patients with mixed phenotype: A single-center study. Clin Transplant 2020; 34:e13781. [PMID: 31958356 DOI: 10.1111/ctr.13781] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 11/29/2022]
Abstract
RATIONALE Patients can change chronic lung allograft dysfunction (CLAD) phenotype, especially from BOS to mixed phenotype. Our aim was to further characterize these patients. METHOD Mixed CLAD was defined as a restrictive physiology with persistent CT opacities, after initial bronchiolitis obliterans syndrome (BOS) diagnosis. The incidence, prognosis, pulmonary function, radiology, pathology, and airway inflammation were compared between patients with restrictive allograft syndrome (RAS) and mixed CLAD. RESULT A total of 268 (44%) patients developed CLAD of which 47 (18%) were diagnosed with RAS "ab initio," 215 (80%) with BOS, and 6 (2%) an undefined phenotype. Twenty-five patients developed a mixed CLAD phenotype (24 BOS to mixed and 1 RAS to mixed). Survival after mixed phenotype diagnosis was comparable (P = .39) to RAS. More emphysema patients developed a mixed phenotype (P = .020) compared to RAS ab initio, while mixed CLAD patients had a lower FEV1 (P < .0001) and FEV1 /FVC (P = .0002) at diagnosis compared to RAS ab initio. CT scans in patients with the mixed phenotype demonstrated apical predominance of the opacities (P = .0034) with pleuroparenchymal fibroelastosis on histopathology. CONCLUSION We further characterized patients with a mixed phenotype of CLAD. Although the survival after diagnosis was comparable to RAS ab initio patients, there was a difference in demography, pulmonary function, radiology, and pathology.
Collapse
Affiliation(s)
- Stijn E Verleden
- Lung Transplant Unit, Department of Chronic diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | - Jan Von Der Thüsen
- Department of Pathology, University Hospital Rotterdam, Rotterdam, The Netherlands
| | - Anke Van Herck
- Lung Transplant Unit, Department of Chronic diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | | | | | | | | | - Bart M Vanaudenaerde
- Lung Transplant Unit, Department of Chronic diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Department of Chronic diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Department of Chronic diseases, Metabolism and Aging, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
23
|
Peripheral CD19+CD24highCD38high B-regulatory cells in lung transplant recipients. Transpl Immunol 2019; 57:101245. [DOI: 10.1016/j.trim.2019.101245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 10/26/2022]
|
24
|
Abstract
Lung transplantation is an accepted therapeutic option for end-stage lung diseases. Its history starts in the 1940s, initially hampered by early deaths due to perioperative problems and acute rejection. Improvement of surgical techniques and the introduction of immunosuppressive drugs resulted in longer survival. Chronic lung allograft dysfunction (CLAD), a new complication appeared and remains the most serious complication today. CLAD, the main reason why survival after lung transplantation is impaired compared to other solid-organ transplantations is characterized by a gradually increasing shortness of breath, reflected in a deterioration of pulmonary function status, respiratory insufficiency and possibly death.
Collapse
|
25
|
Phenotypical diversity of airway morphology in chronic lung graft vs. host disease after stem cell transplantation. Mod Pathol 2019; 32:817-829. [PMID: 30723292 DOI: 10.1038/s41379-019-0203-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023]
Abstract
Pulmonary graft vs. host disease is a diverse and underestimated complication following allogenic hematopoietic stem cell transplantation. We aimed to compare the airway architecture with chronic lung allograft dysfunction post lung transplantation. Inflated explant lungs from graft vs. host disease patients were compared with lungs with chronic lung allograft dysfunction following lung transplantation, and control lungs using a combination of CT, microCT, and histology (n = 6 per group) and pathology in the (small) airways was further quantified and analyzed. Following allogenic hematopoietic stem cell transplantation, three patients presented as bronchiolitis obliterans syndrome and three patients showed interstitial changes and restriction. The CT analysis demonstrated a strong similarity between bronchiolitis obliterans syndrome after lung transplantation and post allogenic hematopoietic stem cell transplantation, evidenced by severe ( > 50%) airway obstruction from generation 9, with 70.8% of the airways ending in obstruction. Further analysis indicated that the airways either collapsed or accumulated matrix along a segment of the airway. In patients with restriction and interstitial changes following allogenic hematopoietic stem cell transplantation, the degree of airway obstruction was lower compared with bronchiolitis obliterans syndrome post allogenic hematopoietic stem cell transplantation, but similar to restrictive allograft syndrome post lung transplantation, showing a lower proportion of airway obstruction (20-35%), decreased number of terminal bronchioles per lung (p < 0.01), and parenchymal fibrosis. We observed similarities in the airway and parenchymal morphometric changes in lung graft vs. host disease and with chronic lung allograft dysfunction following lung transplantation, suggesting similar pathophysiological mechanisms.
Collapse
|
26
|
Wohlschlaeger J, Laenger F, Gottlieb J, Hager T, Seidel A, Jonigk D. Lungentransplantation. DER PATHOLOGE 2019; 40:281-291. [DOI: 10.1007/s00292-019-0598-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
27
|
Glanville AR, Verleden GM, Todd JL, Benden C, Calabrese F, Gottlieb J, Hachem RR, Levine D, Meloni F, Palmer SM, Roman A, Sato M, Singer LG, Tokman S, Verleden SE, von der Thüsen J, Vos R, Snell G. Chronic lung allograft dysfunction: Definition and update of restrictive allograft syndrome-A consensus report from the Pulmonary Council of the ISHLT. J Heart Lung Transplant 2019; 38:483-492. [PMID: 31027539 DOI: 10.1016/j.healun.2019.03.008] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023] Open
Affiliation(s)
- Allan R Glanville
- Lung Transplant Unit, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | | | - Jamie L Todd
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | | | - Fiorella Calabrese
- Department of Cardiothoracic and Vascular Sciences, University of Padova Medical School, Padova, Italy
| | - Jens Gottlieb
- Department of Respiratory Medicine, Hannover Medical School, Member of the German Center for Lung Research, Hannover, Germany
| | - Ramsey R Hachem
- Division of Pulmonary & Critical Care, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Deborah Levine
- Pulmonary Disease and Critical Care Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Federica Meloni
- Department of Respiratory Diseases Policlinico San Matteo Foundation & University of Pavia, Pavia, Italy
| | - Scott M Palmer
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Antonio Roman
- Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Masaaki Sato
- Department of Thoracic Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Lianne G Singer
- Toronto Lung Transplant Program, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | | - Jan von der Thüsen
- Department of Pathology, University Medical Center, Rotterdam, The Netherlands
| | - Robin Vos
- University Hospital Gasthuisberg, Leuven, Belgium
| | - Gregory Snell
- Lung Transplant Service, The Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Vos R, Wuyts WA, Gheysens O, Goffin KE, Schaevers V, Verleden SE, Van Herck A, Sacreas A, Heigl T, McDonough JE, Yserbyt J, Godinas L, Dupont LJ, Neyrinck AP, Van Raemdonck DE, Verbeken EK, Vanaudenaerde BM, Verleden GM. Pirfenidone in restrictive allograft syndrome after lung transplantation: A case series. Am J Transplant 2018; 18:3045-3059. [PMID: 30019840 DOI: 10.1111/ajt.15019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 01/25/2023]
Abstract
Pirfenidone may attenuate the decline of pulmonary function in restrictive allograft syndrome (RAS) after lung transplantation. We retrospectively assessed all lung transplant recipients with RAS who were treated with pirfenidone for at least 3 months (n = 11) in our lung transplant center and report on their long-term outcomes following initiation of pirfenidone. Main outcome parameters included evolution of pulmonary function and overall survival. Pirfenidone appears to attenuate the decline in forced vital capacity and forced expiratory volume in 1 second. Notably, 3 patients were bridged to redo-transplantation with pirfenidone for 11 (5-12) months and are currently alive, while 3 other patients demonstrate long-term stabilization of pulmonary function after 26.6 (range 18.4-46.6) months of treatment. Median overall 3-year survival after RAS diagnosis was 54.5%. Subjective intolerance, mainly anorexia and nausea, necessitating pirfenidone dose de-escalation in 55% of patients, as well as calcineurin dose increase requirements with about 20% are important complications during pirfenidone treatment after lung transplantation. Our findings provide further evidence that pirfenidone appears to be safe and may attenuate the rate of decline in lung function in patients with RAS, but the actual clinical benefit cannot be assessed in the context of this study design and requires further investigation in a larger randomized trial.
Collapse
Affiliation(s)
- Robin Vos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Wim A Wuyts
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Olivier Gheysens
- Department of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Karolien E Goffin
- Department of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Veronique Schaevers
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Anke Van Herck
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Tobias Heigl
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - John E McDonough
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Jonas Yserbyt
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Laurent Godinas
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Lieven J Dupont
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Bart M Vanaudenaerde
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), Division of Respiratory Diseases, KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Chionis L, Grossman BJ, Hachem R, Commean P, Derfler MC, Vedantham S, Dodds K, Spitznagel E, Atkinson J, Despotis G. The efficacy of extracorporeal photopheresis to arrest bronchiolitis obliterans in lung allograft recipients was compared between two automated photopheresis instruments. Transfusion 2018; 58:2933-2941. [PMID: 30312482 DOI: 10.1111/trf.14913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND The most common instruments used for extracorporeal photopheresis (ECP) treatment in the United States are the UVAR XTS and the CELLEX devices (Therakos, West Chester, PA). When compared to the UVAR XTS instrument, the efficacy of the CELLEX instrument to arrest the decline in lung function in patients with chronic lung allograft dysfunction (CLAD) related to bronchiolitis obliterans (BOS) has not been previously evaluated. METHODS The relative efficacy of the CELLEX vs UVAR XTS ECP instruments was assessed by comparing the difference in rates of FEV1 decline before and after ECP treatment and survival in two series of lung allograft recipients with BOS who had been treated with these instruments. RESULTS Similar Slope Difference values for change in rate of decline (6 months Post ECP - Pre ECP) were observed between the two cohorts (UVAR XTS: 85 ± 109 mL/month vs CELLEX: 76 ± 128 mL/month, p=0.72). A similar percentage of patients responded to ECP (UVAR XTS: 77% vs CELLEX: 89%; p=0.36) i.e., as defined as a positive difference in slope between the rate of decline of FEV1 before and 6 months after ECP. Survival at either 6 (p=0.89) or 12 (p=0.8) months after the start of ECP was not associated with instrument used despite a trend in higher early mortality (34% vs 17%, p=0.054) in the patients who were predominately treated with the CELLEX. CONCLUSIONS Our data support the use of the CELLEX for prospective studies designed to evaluate the merits of ECP in this population.
Collapse
Affiliation(s)
| | - Brenda J Grossman
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Ramsey Hachem
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Washington University School of Medicine, St Louis, Missouri
| | - Paul Commean
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Mary Clare Derfler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Suresh Vedantham
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Kathy Dodds
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Edward Spitznagel
- Department of Mathematics, Washington University, St Louis, Missouri
| | - Jeff Atkinson
- Department of Medicine, Division of Pulmonology, Washington University School of Medicine, St Louis, Missouri
| | - George Despotis
- Department of Pathology & Immunology, Division of Laboratory & Genomic Medicine, Washington University School of Medicine, St Louis, Missouri.,Department of Anesthesiology, Division of Cardiothoracic Anesthesiology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
30
|
Parker WF, Bag R. Chronic Lung Allograft Dysfunction. CURRENT PULMONOLOGY REPORTS 2018. [DOI: 10.1007/s13665-018-0208-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
31
|
Orfanos S, Gomez C, Baron S, Akkisetty R, Dufeu N, Coltey B, Thomas PA, Rolain JM, Reynaud-Gaubert M. Impact of gram negative bacteria airway recolonization on the occurrence of chronic lung allograft dysfunction after lung transplantation in a population of cystic fibrosis patients. BMC Microbiol 2018; 18:88. [PMID: 30126365 PMCID: PMC6102836 DOI: 10.1186/s12866-018-1231-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Background Chronic Lung Allograft Dysfunction (CLAD) is the main cause of morbidity and mortality after the first year following lung transplantation (LTx). Risk factors of CLAD have been extensively studied, but the association between gram-negative bacteria (GNB) bronchial colonization and the development of CLAD is controversial. The purpose of our study was to investigate the association between post-transplant recolonization with the same species or de-novo colonization with a new GNB species and CLAD. The same analysis was performed on a sub-group of patients at the strain level using Matrix Assisted Laser Desorption Ionization-Time of Flight Mass Spectrometry technique. Results Forty adult cystic fibrosis (CF) patients who underwent a first bilateral LTx in the University Hospital of Marseille, between January 2010 and December 2014, were included in the study. Patients with GNB de-novo colonization had a higher risk of developing CLAD (OR = 6.72, p = 0.04) and a lower rate of CLAD-free survival (p = 0.005) compared to patients with GNB recolonization. No conclusion could be drawn from the subgroup MALDI-TOF MS analysis at the strain level. Conclusion Post-LTx GNB airway recolonization seems to be a protective factor against CLAD, whereas de-novo colonization with a new species of GNB seems to be a risk factor for CLAD. Electronic supplementary material The online version of this article (10.1186/s12866-018-1231-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarah Orfanos
- Aix-Marseille University, Faculté de médecine, Marseille, France.
| | - Carine Gomez
- Aix-Marseille University, Faculté de médecine, Marseille, France.,Department of Respiratory Diseases, Lung Transplant Team, University Hospital of Marseille, Marseille, France
| | - Sophie Baron
- Aix-Marseille University, Faculté de médecine, Marseille, France.,URMITE CNRS IRD UMR 6236, IHU Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Ritesh Akkisetty
- Department of Biology, Drexel University College of Art and Sciences, Philadelphia, USA
| | - Nadine Dufeu
- Department of Respiratory Diseases, Lung Transplant Team, University Hospital of Marseille, Marseille, France
| | - Bérengère Coltey
- Department of Respiratory Diseases, Lung Transplant Team, University Hospital of Marseille, Marseille, France
| | - Pascal Alexandre Thomas
- Aix-Marseille University, Faculté de médecine, Marseille, France.,Department of Thoracic Surgery, Lung Transplant Team APHM, University Hospital of Marseille, Marseille, France
| | - Jean Marc Rolain
- Aix-Marseille University, Faculté de médecine, Marseille, France.,URMITE CNRS IRD UMR 6236, IHU Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Martine Reynaud-Gaubert
- Aix-Marseille University, Faculté de médecine, Marseille, France.,Department of Respiratory Diseases, Lung Transplant Team, University Hospital of Marseille, Marseille, France
| |
Collapse
|
32
|
von der Thüsen JH, Vandermeulen E, Vos R, Weynand B, Verbeken EK, Verleden SE. The histomorphological spectrum of restrictive chronic lung allograft dysfunction and implications for prognosis. Mod Pathol 2018; 31:780-790. [PMID: 29327719 DOI: 10.1038/modpathol.2017.180] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022]
Abstract
Chronic lung allograft dysfunction continues to be the main contributor to poor long-term allograft survival after lung transplantation. The restrictive phenotype of chronic lung allograft dysfunction carries a particularly poor prognosis. Little is known about the pathogenetic mechanisms involved in restrictive chronic lung allograft dysfunction. In this study, we performed histomorphological and immunohistochemical analysis of restrictive chronic lung allograft dysfunction lungs. Explant lung tissue from 21 restrictive chronic lung allograft dysfunction patients was collected and histopathologic patterns of rejection, fibrosis and vascular changes were scored after routine histochemical stains and additional immunohistochemistry for endothelial markers and C4d. In all, 75% of cases showed evidence of acute cellular rejection; lymphocytic bronchiolitis was absent in most lungs, whereas in 55% there was obliterative bronchiolitis. Almost half of the cases showed a pattern consistent with pleuroparenchymal fibro-elastosis (n=10), and a subset showed nonspecific interstitial pneumonia (n=5) or irregular emphysema (n=5). Fibrinous alveolar exudates were frequently seen in association with fibrosis (n=6), but no diffuse alveolar damage was found. Evidence of microvascular damage was present in most cases. An emphysematous pattern of fibrosis was associated with a better survival (P=0.0030), whereas fibrinous exudates were associated with a worse survival (P=0.0007). In addition to the previously described nonspecific interstitial pneumonia and pleuroparenchymal fibro-elastosis patterns in restrictive chronic lung allograft dysfunction, we are the first to describe a pattern of fibrosis-induced subpleural/paraseptal emphysema. This pattern confers a better survival, whereas fibrinous exudates are associated with a worse survival. We believe that our findings offer a pathogenetic theory for pleuroparenchymal fibro-elastosis in restrictive chronic lung allograft dysfunction, and show that restrictive chronic lung allograft dysfunction is an increasingly heterogeneous disease with presumably different mechanisms of subpattern formation.
Collapse
Affiliation(s)
| | - Elly Vandermeulen
- Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | | | - Stijn E Verleden
- Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Durand M, Lacoste P, Danger R, Jacquemont L, Brosseau C, Durand E, Tilly G, Loy J, Foureau A, Royer PJ, Tissot A, Roux A, Reynaud-Gaubert M, Kessler R, Mussot S, Dromer C, Brugière O, Mornex JF, Guillemain R, Claustre J, Degauque N, Magnan A, Brouard S. High circulating CD4 +CD25 hiFOXP3 + T-cell sub-population early after lung transplantation is associated with development of bronchiolitis obliterans syndrome. J Heart Lung Transplant 2018; 37:770-781. [PMID: 29571601 DOI: 10.1016/j.healun.2018.01.1306] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/07/2017] [Accepted: 01/24/2018] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND Chronic bronchiolitis obliterans syndrome (BOS) remains a major limitation for long-term survival after lung transplantation. The immune mechanisms involved and predictive biomarkers have yet to be identified. The purpose of this study was to determine whether peripheral blood T-lymphocyte profile could predict BOS in lung transplant recipients. METHODS An in-depth profiling of CD4+ and CD8+ T cells was prospectively performed on blood cells from stable (STA) and BOS patients with a longitudinal follow-up. Samples were analyzed at 1 and 6 months after transplantation, at the time of BOS diagnosis, and at an intermediate time-point at 6 to 12 months before BOS diagnosis. RESULTS Although no significant difference was found for T-cell compartments at BOS diagnosis or several months beforehand, we identified an increase in the CD4+CD25hiFoxP3+ T-cell sub-population in BOS patients at 1 and 6 months after transplantation (3.39 ± 0.40% vs 1.67 ± 0.22% in STA, p < 0.001). A CD4+CD25hiFoxP3+ T-cell threshold of 2.4% discriminated BOS and stable patients at 1 month post-transplantation. This was validated on a second set of patients at 6 months post-transplantation. Patients with a proportion of CD4+CD25hiFoxP3+ T cells up to 2.4% in the 6 months after transplantation had a 2-fold higher risk of developing BOS. CONCLUSIONS This study is the first to report an increased proportion of circulating CD4+CD25hiFoxP3+ T cells early post-transplantation in lung recipients who proceed to develop BOS within 3 years, which supports its use as a BOS predictive biomarker.
Collapse
Affiliation(s)
- Maxim Durand
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France
| | - Philippe Lacoste
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Richard Danger
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Lola Jacquemont
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Carole Brosseau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Eugénie Durand
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Gaelle Tilly
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Jennifer Loy
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Aurore Foureau
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Pierre-Joseph Royer
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Adrien Tissot
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Antoine Roux
- Hôpital Foch, Suresnes, Université de Versailles, Saint-Quentin-en-Yvelines, France
| | | | | | - Sacha Mussot
- Centre Chirurgical Marie Lannelongue, Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardiopulmonaire, Le Plessis Robinson, France
| | | | - Olivier Brugière
- Hôpital Bichat, Service de Pneumologie et Transplantation Pulmonaire, Paris, France
| | | | | | - Johanna Claustre
- Clinique Universitaire de Pneumologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Université Grenoble Alpes, Inserm U1055, Grenoble, France
| | - Nicolas Degauque
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Antoine Magnan
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France; Institut du thorax, CHU de Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France; Centre d'Investigation Clinique Biothérapie, CHU Nantes, Nantes, France.
| | | |
Collapse
|
34
|
Verleden SE, Vos R, Vanaudenaerde BM, Verleden GM. Chronic lung allograft dysfunction phenotypes and treatment. J Thorac Dis 2017; 9:2650-2659. [PMID: 28932572 DOI: 10.21037/jtd.2017.07.81] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) remains a major hurdle limiting long-term survival post lung transplantation. Given the clinical heterogeneity of CLAD, recently two phenotypes of CLAD have been defined [bronchiolitis obliterans syndrome (BOS) vs. restrictive allograft syndrome (RAS) or restrictive CLAD (rCLAD)]. BOS is characterized by an obstructive pulmonary function, air trapping on CT and obliterative bronchiolitis (OB) on histopathology, while RAS/rCLAD patients show a restrictive pulmonary function, persistent pleuro-parenchymal infiltrates on CT and pleuroparenchymal fibro-elastosis on biopsies. Importantly, the patients with RAS/rCLAD have a severely limited survival post diagnosis of 6-18 months compared to 3-5 years after BOS diagnosis. In this review, we will review historical evidence for this heterogeneity and we will highlight the clinical, radiological, histopathological characteristics of both phenotypes, as well as their risk factors. Treatment of CLAD remains troublesome, nevertheless, we will give an overview of different treatment strategies that have been tried with some success. Adequate phenotyping remains difficult but is clearly needed for both clinical and scientific purposes.
Collapse
Affiliation(s)
- Stijn E Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Department of Clinical and Experimental Medicine, Lung Transplant Unit, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Belloli EA, Degtiar I, Wang X, Yanik GA, Stuckey LJ, Verleden SE, Kazerooni EA, Ross BD, Murray S, Galbán CJ, Lama VN. Parametric Response Mapping as an Imaging Biomarker in Lung Transplant Recipients. Am J Respir Crit Care Med 2017; 195:942-952. [PMID: 27779421 DOI: 10.1164/rccm.201604-0732oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
RATIONALE The predominant cause of chronic lung allograft failure is small airway obstruction arising from bronchiolitis obliterans. However, clinical methodologies for evaluating presence and degree of small airway disease are lacking. OBJECTIVES To determine if parametric response mapping (PRM), a novel computed tomography voxel-wise methodology, can offer insight into chronic allograft failure phenotypes and provide prognostic information following spirometric decline. METHODS PRM-based computed tomography metrics quantifying functional small airways disease (PRMfSAD) and parenchymal disease (PRMPD) were compared between bilateral lung transplant recipients with irreversible spirometric decline and control subjects matched by time post-transplant (n = 22). PRMfSAD at spirometric decline was evaluated as a prognostic marker for mortality in a cohort study via multivariable restricted mean models (n = 52). MEASUREMENTS AND MAIN RESULTS Patients presenting with an isolated decline in FEV1 (FEV1 First) had significantly higher PRMfSAD than control subjects (28% vs. 15%; P = 0.005), whereas patients with concurrent decline in FEV1 and FVC had significantly higher PRMPD than control subjects (39% vs. 20%; P = 0.02). Over 8.3 years of follow-up, FEV1 First patients with PRMfSAD greater than or equal to 30% at spirometric decline lived on average 2.6 years less than those with PRMfSAD less than 30% (P = 0.004). In this group, PRMfSAD greater than or equal to 30% was the strongest predictor of survival in a multivariable model including bronchiolitis obliterans syndrome grade and baseline FEV1% predicted (P = 0.04). CONCLUSIONS PRM is a novel imaging tool for lung transplant recipients presenting with spirometric decline. Quantifying underlying small airway obstruction via PRMfSAD helps further stratify the risk of death in patients with diverse spirometric decline patterns.
Collapse
Affiliation(s)
| | | | - Xin Wang
- 2 Department of Biostatistics, and
| | - Gregory A Yanik
- 3 Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan; and
| | | | - Stijn E Verleden
- 5 Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Ella A Kazerooni
- 6 Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan
| | - Brian D Ross
- 6 Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan
| | | | - Craig J Galbán
- 6 Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan
| | - Vibha N Lama
- 1 Division of Pulmonary and Critical Care Medicine
| |
Collapse
|
36
|
Analysis of long term CD4+CD25highCD127- T-reg cells kinetics in peripheral blood of lung transplant recipients. BMC Pulm Med 2017; 17:102. [PMID: 28720146 PMCID: PMC5516333 DOI: 10.1186/s12890-017-0446-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Background The role of CD4+CD25highCD127− T-reg cells in solid-organ Transplant (Tx) acceptance has been extensively studied. In previous studies on kidney and liver recipients, peripheral T-reg cell counts were associated to graft survival, while in lung Tx, there is limited evidence for similar findings. This study aims to analyze long term peripheral kinetics of T-reg-cells in a cohort of lung recipients and tests its association to several clinical variables. Methods From jan 2009 to dec 2014, 137 lung Tx recipients were submitted to an immunological follow up (median: 105.9 months (6.7–310.5)). Immunological follow up consisted of a complete blood peripheral immuno-phenotype, inclusive of CD4+CD25highCD127− T and FOXP3+ cells. We tested the association between T-reg and relevant variables by linear OR regression models for repeated measures, adjusting for time from Tx. Also, by ordered logistic models for panel data, the association between Chronic Lung Allograft Dysfuncton (CLAD) onset/progression and T-reg counts in the previous 3 months was tested. Results Among all variables analyzed at multivariate analysis: Bronchiolitis Obliterans Syndrome (OR −6.51, p < 0.001), Restrictive Allograft Syndrome (OR −5.19, p = 0.04) and Extracorporeal photopheresis (OR −5.65, p < 0.001) were significantly associated to T-reg cell. T-reg cell counts progressively decreased according to the severity of CLAD. Furthermore, patients with higher mean T-reg counts in a trimester had a significantly lower risk (OR 0.97, p = 0.012) of presenting CLAD or progressing in the graft dysfunction in the following trimester. Conclusions Our present data confirm animal observations on the possible role of T-reg in the evolution of CLAD.
Collapse
|
37
|
Cova E, Inghilleri S, Pandolfi L, Morosini M, Magni S, Colombo M, Piloni D, Finetti C, Ceccarelli G, Benedetti L, Cusella MG, Agozzino M, Corsi F, Allevi R, Mrakic-Sposta S, Moretti S, De Gregori S, Prosperi D, Meloni F. Bioengineered gold nanoparticles targeted to mesenchymal cells from patients with bronchiolitis obliterans syndrome does not rise the inflammatory response and can be safely inhaled by rodents. Nanotoxicology 2017; 11:534-545. [PMID: 28415888 DOI: 10.1080/17435390.2017.1317862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The use of gold nanoparticles (GNPs) as drug delivery system represents a promising issue for diseases without effective pharmacological treatment due to insufficient local drug accumulation and excessive systemic toxicity. Bronchiolitis obliterans syndrome (BOS) represents about 70% of cases of chronic lung allograft dysfunction, the main challenge to long-term lung transplantation. It is believed that due to repeated insults to epithelial bronchiolar cells local inflammatory response creates a milieu that favors epithelial-mesenchymal transition and activation of local mesenchymal cells (MCs) leading to airway fibro-obliteration. In a previous work, we engineered GNPs loaded with the mammalian target of rapamycin inhibitor everolimus, specifically decorated with an antibody against CD44, a surface receptor expressed by primary MCs isolated from bronchoalveolar lavage of BOS patients. We proved in vitro that these GNPs (GNP-HCe) were able to specifically inhibit primary MCs without affecting the bronchial epithelial cell. In the present work, we investigated the effect of these bioengineered nanoconstructs on inflammatory cells, given that a stimulating effect on macrophages, neutrophils or lymphocytes is strongly unwanted in graft airways since it would foster fibrogenesis. In addition, we administered GNP-HCe by the inhalatory route to normal mice for a preliminary assessment of their pulmonary and peripheral (liver, spleen and kidney) uptake. By these experiments, an evaluation of tissue toxicity was also performed. The present study proves that our bioengineered nanotools do not rise an inflammatory response and, under the tested inhalatory conditions that were used, are non-toxic.
Collapse
Affiliation(s)
- Emanuela Cova
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Simona Inghilleri
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Laura Pandolfi
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Monica Morosini
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Sara Magni
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Miriam Colombo
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Davide Piloni
- c Dipartimento di Medicina Interna, Unità di Pneumologia , Università degli Studi di Pavia , Pavia , Italy
| | - Chiara Finetti
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Gabriele Ceccarelli
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Laura Benedetti
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Maria Gabriella Cusella
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Manuela Agozzino
- e Centro per le Malattie Cardiovascolari Ereditarie , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Fabio Corsi
- f Dipartimento di Scienze Biomediche e Cliniche L. Sacco , Università degli Studi di Milano , Pavia , Italy.,g Chirurgia Senologica , ICS Maugeri S.p.A. SB , Pavia , Italy
| | - Raffaele Allevi
- f Dipartimento di Scienze Biomediche e Cliniche L. Sacco , Università degli Studi di Milano , Pavia , Italy
| | - Simona Mrakic-Sposta
- h Istituto di Bioimmagini e Fisiologia Molecolare , Consiglio Nazionale delle Ricerche (CNR) , Segrate , Milano , Italia
| | - Sarah Moretti
- h Istituto di Bioimmagini e Fisiologia Molecolare , Consiglio Nazionale delle Ricerche (CNR) , Segrate , Milano , Italia
| | - Simona De Gregori
- i S.S.di Farmacocinetica Clinica e Sperimentale , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Davide Prosperi
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Federica Meloni
- c Dipartimento di Medicina Interna, Unità di Pneumologia , Università degli Studi di Pavia , Pavia , Italy
| |
Collapse
|
38
|
Müller C, Andersson-Sjöland A, Schultz HH, Eriksson LT, Andersen CB, Iversen M, Westergren-Thorsson G. Early extracellular matrix changes are associated with later development of bronchiolitis obliterans syndrome after lung transplantation. BMJ Open Respir Res 2017; 4:e000177. [PMID: 28469930 PMCID: PMC5411729 DOI: 10.1136/bmjresp-2016-000177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022] Open
Abstract
Background Chronic lung allograft dysfunction in the form of bronchiolitis obliterans syndrome (BOS) is the main cause of death beyond 1-year post-lung transplantation. The disease-initiating triggers as well as the molecular changes leading to fibrotic alterations in the transplanted lung are largely unknown. The aim of this study was to identify potential early changes in the extracellular matrix (ECM) in different compartments of the transplanted lung prior to the development of BOS. Methods Transbronchial biopsies from a cohort of 58 lung transplantation patients at the Copenhagen University hospital between 2005 and 2006, with or without development of BOS in a 5-year follow-up, were obtained 3 and 12 months after transplantation. Biopsies were assessed for total collagen, collagen type IV and biglycan in the alveolar and small airway compartments using Masson's Trichrome staining and immunohistochemistry. Results A time-specific and compartment-specific pattern of ECM changes was detected. Alveolar total collagen (p=0.0190) and small airway biglycan (p=0.0199) increased between 3 and 12 months after transplantation in patients developing BOS, while collagen type IV (p=0.0124) increased in patients without BOS. Patients with early-onset BOS mirrored this increase. Patients developing grade 3 BOS showed distinct ECM changes already at 3 months. Patients with BOS with treated acute rejections displayed reduced alveolar total collagen (p=0.0501) and small airway biglycan (p=0.0485) at 3 months. Conclusions Patients with future BOS displayed distinct ECM changes compared with patients without BOS. Our data indicate an involvement of alveolar and small airway compartments in post-transplantation changes in the development of BOS.
Collapse
Affiliation(s)
- Catharina Müller
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Hans Henrik Schultz
- Section for Lung Transplantation, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Leif T Eriksson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Claus B Andersen
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Martin Iversen
- Section for Lung Transplantation, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
39
|
Nayak DK, Saravanan PB, Bansal S, Naziruddin B, Mohanakumar T. Autologous and Allogenous Antibodies in Lung and Islet Cell Transplantation. Front Immunol 2016; 7:650. [PMID: 28066448 PMCID: PMC5179571 DOI: 10.3389/fimmu.2016.00650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/14/2016] [Indexed: 01/02/2023] Open
Abstract
The field of organ transplantation has undoubtedly made great strides in recent years. Despite the advances in donor-recipient histocompatibility testing, improvement in transplantation procedures, and development of aggressive immunosuppressive regimens, graft-directed immune responses still pose a major problem to the long-term success of organ transplantation. Elicitation of immune responses detected as antibodies to mismatched donor antigens (alloantibodies) and tissue-restricted self-antigens (autoantibodies) are two major risk factors for the development of graft rejection that ultimately lead to graft failure. In this review, we describe current understanding on genesis and pathogenesis of antibodies in two important clinical scenarios: lung transplantation and transplantation of islet of Langerhans. It is evident that when compared to any other clinical solid organ or cellular transplant, lung and islet transplants are more susceptible to rejection by combination of allo- and autoimmune responses.
Collapse
Affiliation(s)
- Deepak Kumar Nayak
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | | | - Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | | | | |
Collapse
|
40
|
Jonigk D, Rath B, Borchert P, Braubach P, Maegel L, Izykowski N, Warnecke G, Sommer W, Kreipe H, Blach R, Anklamm A, Haverich A, Eder M, Stadler M, Welte T, Gottlieb J, Kuehnel M, Laenger F. Comparative analysis of morphological and molecular motifs in bronchiolitis obliterans and alveolar fibroelastosis after lung and stem cell transplantation. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 3:17-28. [PMID: 28138398 PMCID: PMC5259562 DOI: 10.1002/cjp2.60] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 12/14/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) remains the major obstacle to long‐term survival following lung transplantation (LuTx). Morphologically CLAD is defined by obliterative remodelling of the small airways (bronchiolitis obliterans, BO) as well as a more recently described collagenous obliteration of alveoli with elastosis summarised as alveolar fibroelastosis (AFE). Both patterns are not restricted to pulmonary allografts, but have also been reported following haematopoietic stem cell transplantation (HSCT) and radio chemotherapy (RC). In this study we performed compartment‐specific morphological and molecular analysis of BO and AFE lesions in human CLAD (n = 22), HSCT (n = 29) and RC (n = 6) lung explants, utilising conventional histopathology, laser‐microdissection, PCR techniques and immunohistochemistry to assess fibrosis‐associated gene and protein expression. Three key results emerged from our analysis of fibrosis‐associated genes: (i) generally speaking, “BO is BO”. Despite the varying clinical backgrounds, the molecular characteristics of BO lesions were found to be alike in all groups. (ii) “AFE is AFE”. In all groups of patients suffering from restrictive changes to lung physiology due to AFE there were largely – but not absolutely ‐ identical gene expression patterns. iii) BO concomitant to AFE after LuTx is characterised by an AFE‐like molecular microenvironment, representing the only exception to (i). Additionally, we describe an evolutionary model for the AFE pattern: a non‐specific fibrin‐rich reaction to injury pattern triggers a misguided resolution attempt and eventual progression towards manifest AFE. Our data point towards an absence of classical fibrinolytic enzymes and an alternative fibrin degrading mechanism via macrophages, resulting in fibrous remodelling and restrictive functional changes. These data may serve as diagnostic adjuncts and help to predict the clinical course of respiratory dysfunction in LuTx and HSCT patients. Moreover, analysis of the mechanism of fibrinolysis and fibrogenesis may unveil potential therapeutic targets to alter the course of the eventually fatal lung remodelling.
Collapse
Affiliation(s)
- Danny Jonigk
- Institute of Pathology, Hannover Medical School (MHH)HanoverGermany; The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany
| | - Berenice Rath
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Paul Borchert
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Lavinia Maegel
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Nicole Izykowski
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Gregor Warnecke
- The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany; Division of Cardiac, Thoracic, Transplantation and Vascular SurgeryMedical School HanoverHanoverGermany
| | - Wiebke Sommer
- The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany; Division of Cardiac, Thoracic, Transplantation and Vascular SurgeryMedical School HanoverHanoverGermany
| | - Hans Kreipe
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Robert Blach
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Adrian Anklamm
- Institute of Pathology, Hannover Medical School (MHH) Hanover Germany
| | - Axel Haverich
- The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany; Division of Cardiac, Thoracic, Transplantation and Vascular SurgeryMedical School HanoverHanoverGermany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Medical School Hanover Hanover Germany
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation Medical School Hanover Hanover Germany
| | - Tobias Welte
- The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany; Department of Respiratory Medicine, Medical School Hanover, Hanover, Germany
| | - Jens Gottlieb
- The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany; Department of Respiratory Medicine, Medical School Hanover, Hanover, Germany
| | - Mark Kuehnel
- Institute of Pathology, Hannover Medical School (MHH)HanoverGermany; The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany
| | - Florian Laenger
- Institute of Pathology, Hannover Medical School (MHH)HanoverGermany; The German Center for Lung Research (Deutsches Zentrum für Lungenforschung DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hanover (BREATH)HanoverGermany
| |
Collapse
|
41
|
Vandermeulen E, Lammertyn E, Verleden SE, Ruttens D, Bellon H, Ricciardi M, Somers J, Bracke KR, Van Den Eynde K, Tousseyn T, Brusselle GG, Verbeken EK, Verschakelen J, Emonds MP, Van Raemdonck DE, Verleden GM, Vos R, Vanaudenaerde BM. Immunological diversity in phenotypes of chronic lung allograft dysfunction: a comprehensive immunohistochemical analysis. Transpl Int 2016; 30:134-143. [PMID: 27933655 DOI: 10.1111/tri.12882] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 11/30/2022]
Abstract
Chronic rejection after organ transplantation is defined as a humoral- and cell-mediated immune response directed against the allograft. In lung transplantation, chronic rejection is nowadays clinically defined as a cause of chronic lung allograft dysfunction (CLAD), consisting of different clinical phenotypes including restrictive allograft syndrome (RAS) and bronchiolitis obliterans syndrome (BOS). However, the differential role of humoral and cellular immunity is not investigated up to now. Explant lungs of patients with end-stage BOS (n = 19) and RAS (n = 18) were assessed for the presence of lymphoid (B and T cells) and myeloid cells (dendritic cells, eosinophils, mast cells, neutrophils, and macrophages) and compared to nontransplant control lung biopsies (n = 21). All myeloid cells, with exception of dendritic cells, were increased in RAS versus control (neutrophils, eosinophils, and mast cells: all P < 0.05, macrophages: P < 0.001). Regarding lymphoid cells, B cells and cytotoxic T cells were increased remarkably in RAS versus control (P < 0.001) and in BOS versus control (P < 0.01). Interestingly, lymphoid follicles were restricted to RAS (P < 0.001 versus control and P < 0.05 versus BOS). Our data suggest an immunological diversity between BOS and RAS, with a more pronounced involvement of the B-cell response in RAS characterized by a structural organization of lymphoid follicles. This may impact future therapeutic approaches.
Collapse
Affiliation(s)
- Elly Vandermeulen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Elise Lammertyn
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Stijn E Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - David Ruttens
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Hannelore Bellon
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Mario Ricciardi
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Jana Somers
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Ken R Bracke
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathleen Van Den Eynde
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Thomas Tousseyn
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Erik K Verbeken
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Johny Verschakelen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | | | - Dirk E Van Raemdonck
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| |
Collapse
|
42
|
Verleden SE, Vos R, Vandermeulen E, Ruttens D, Bellon H, Heigl T, Van Raemdonck DE, Verleden GM, Lama V, Ross BD, Galbán CJ, Vanaudenaerde BM. Parametric Response Mapping of Bronchiolitis Obliterans Syndrome Progression After Lung Transplantation. Am J Transplant 2016; 16:3262-3269. [PMID: 27367568 PMCID: PMC5083149 DOI: 10.1111/ajt.13945] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/07/2016] [Accepted: 06/12/2016] [Indexed: 01/25/2023]
Abstract
Bronchiolitis obliterans syndrome (BOS) remains a major complication after lung transplantation. Air trapping and mosaic attenuation are typical radiological features of BOS; however, quantitative evaluation remains troublesome. We evaluated parametric response mapping (PRM, voxel-to-voxel comparison of inspiratory and expiratory computed tomography [CT] scans) in lung transplant recipients diagnosed with BOS (n = 20) and time-matched stable lung transplant recipients (n = 20). Serial PRM measurements were performed prediagnosis, at time of BOS diagnosis, and postdiagnosis (Tpre , T0 , and Tpost , respectively), or at a postoperatively matched time in stable patients. PRM results were correlated with pulmonary function and confirmed by microCT analysis of end-stage explanted lung tissue. Using PRM, we observed an increase in functional small airway disease (fSAD), from Tpre to T0 (p = 0.006) and a concurrent decrease in healthy parenchyma (p = 0.02) in the BOS group. This change in PRM continued to Tpost , which was significantly different compared to the stable patients (p = 0.0002). At BOS diagnosis, the increase in fSAD was strongly associated with a decrease in forced expiratory volume in 1 s (p = 0.011). Micro-CT confirmed the presence of airway obliteration in a sample of a BOS patient identified with 67% fSAD by PRM. We demonstrated the use of PRM as an adequate output to monitor BOS progression in lung transplant recipients.
Collapse
Affiliation(s)
- Stijn E Verleden
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Robin Vos
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Elly Vandermeulen
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - David Ruttens
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Hannelore Bellon
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Tobias Heigl
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Dirk E Van Raemdonck
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Geert M Verleden
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| | - Vibha Lama
- Pneumology Department, University of Michigan, Ann Arbor, MI, USA
| | - Brian D Ross
- Radiology Department, University of Michigan, Ann Arbor, MI, USA
| | - Craig J Galbán
- Radiology Department, University of Michigan, Ann Arbor, MI, USA
| | - Bart M Vanaudenaerde
- Lung transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven Belgium
| |
Collapse
|
43
|
Yamada Y, Vandermeulen E, Heigl T, Somers J, Vaneylen A, Verleden SE, Bellon H, De Vleeschauwer S, Verbeken EK, Van Raemdonck DE, Vos R, Verleden GM, Jungraithmayr W, Vanaudenaerde BM. The role of recipient derived interleukin-17A in a murine orthotopic lung transplant model of restrictive chronic lung allograft dysfunction. Transpl Immunol 2016; 39:10-17. [PMID: 27737799 DOI: 10.1016/j.trim.2016.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/10/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023]
Abstract
The single most important cause of late mortality after lung transplantation is chronic lung allograft dysfunction (CLAD). However, the pathological development of CLAD was not as simple as previously presumed and subclassification phenotypes, bronchiolitis obliterans syndrome (BOS) and restrictive CLAD (rCLAD), have been introduced. We want to re-investigate how CLAD manifests in the murine orthotopic lung transplant model and investigate the role of interleukin 17A (IL-17A) within this model. Orthotopic LTx was performed in CB57BL/6, IL-17 WT and IL-17 KO mice. In a first experiment, CB57BL/6 mice receiving an isograft (CB57BL/6) or allograft (BALB/C) were compared. In a second experiment IL-17 WT and IL-17 KO mice (both CB57BL/6 background) received an allograft (BALB/C). Mice received daily immunosuppression with steroids and cyclosporine and were sacrificed 10weeks after transplantation for histopathological analysis by an experienced lung pathologist. After murine orthotopic lung transplantation, the allograft histopathologically presented features of human rCLAD (i.e. overt inflammation, pleural/parenchymal fibrosis and obliterative bronchiolitis). In the IL-17A KO group, less inflammation in the bronchovascular axis (p=0.03) was observed and a non-significant trend towards less bronchovascular fibrosis, pleural/septal inflammation and fibrosis, and parenchymal inflammation and fibrosis when compared to WT mice. The major mismatch orthotopic lung transplant model resembles features of human rCLAD. IL-17A mediated immunity is involved in the inflammatory component, but had little influence on the degree of fibrosis. Further mechanistic and therapeutic studies in this mouse model are needed to fully understand the mechanisms in rCLAD.
Collapse
Affiliation(s)
- Y Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - E Vandermeulen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - T Heigl
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - J Somers
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - A Vaneylen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S E Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - H Bellon
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S De Vleeschauwer
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - E K Verbeken
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - D E Van Raemdonck
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - R Vos
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - G M Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - W Jungraithmayr
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - B M Vanaudenaerde
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
44
|
Vandermeulen E, Verleden SE, Bellon H, Ruttens D, Lammertyn E, Claes S, Vandooren J, Ugarte-Berzal E, Schols D, Emonds MP, Van Raemdonck DE, Opdenakker G, Verleden GM, Vos R, Vanaudenaerde BM. Humoral immunity in phenotypes of chronic lung allograft dysfunction: A broncho-alveolar lavage fluid analysis. Transpl Immunol 2016; 38:27-32. [PMID: 27561239 DOI: 10.1016/j.trim.2016.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/18/2016] [Accepted: 08/20/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Recently, antibody mediated rejection (AMR) has been associated with a higher incidence of chronic lung allograft dysfunction (CLAD) and mortality after lung transplantation (LTx). We investigated markers related to AMR and matrix remodeling in CLAD, with special attention for its two phenotypes being bronchiolitis obliterans syndrome (BOS) and restrictive CLAD (rCLAD). METHODS Immunoglobulins (IgA, IgE, IgG1-IgG4, total IgG and IgM) and complement (C4d and C1q) were quantified in lung lavage samples at the moment of BOS (n=15) or RAS (n=16) diagnosis; and were compared to stable transplant patients who served as control (n=14). Also, airway remodeling and metalloproteinases (MMPs) were investigated via zymography and gelatin degradation. The presence of DSA was additionally assessed in blood. RESULTS Total IgG, IgG1-IgG4 and IgM were increased in rCLAD versus control (p<0.001) and BOS patients (p<0.01). IgA and IgE were increased in rCLAD compared to control (respectively p<0.05 and p<0.01), but not to BOS. Total IgG and IgE were increased in BOS versus control (respectively p<0.01 and p<0.05). Complement proteins were exclusively present in rCLAD and correlated positively with immunoglobulins. Additionally, in blood, DSA were more present in rCLAD (p=0.041). MMP-9 levels increased in RAS and BOS versus control (p<0.001) and MMP-9 induced gelatin degradation was only increased in BOS compared to control (p<0.01). CONCLUSION We demonstrated increased levels of immunoglobulins and complement proteins dominantly present in rCLAD. This leads to the belief that antibodies and AMR might play a more important role in rCLAD compared to BOS. Therefore, anti B-cell therapy could offer beneficial therapeutic effects in patients diagnosed with rCLAD, which needs further research.
Collapse
Affiliation(s)
- Elly Vandermeulen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Stijn E Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Hannelore Bellon
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - David Ruttens
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Elise Lammertyn
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Sandra Claes
- Laboratory of Virology and Chemotherapy (Rega Institute), Department of Microbiology and Immunology, KULeuven, Leuven, Belgium
| | - Jennifer Vandooren
- Laboratory of Immunobiology (Rega Institute), Department of Microbiology and Immunology, KULeuven, Leuven, Belgium
| | - Estafania Ugarte-Berzal
- Laboratory of Immunobiology (Rega Institute), Department of Microbiology and Immunology, KULeuven, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy (Rega Institute), Department of Microbiology and Immunology, KULeuven, Leuven, Belgium
| | | | - Dirk E Van Raemdonck
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology (Rega Institute), Department of Microbiology and Immunology, KULeuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium.
| |
Collapse
|
45
|
Verleden SE, Ruttens D, Vandermeulen E, Bellon H, Dubbeldam A, De Wever W, Dupont LJ, Van Raemdonck DE, Vanaudenaerde BM, Verleden GM, Benden C, Vos R. Predictors of survival in restrictive chronic lung allograft dysfunction after lung transplantation. J Heart Lung Transplant 2016; 35:1078-84. [PMID: 27212563 DOI: 10.1016/j.healun.2016.03.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/21/2016] [Accepted: 03/30/2016] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) is the main factor limiting long-term survival after lung transplantation. Besides bronchiolitis obliterans syndrome, a restrictive phenotype of CLAD (rCLAD) exists, which is associated with poor prognosis after diagnosis. However, survival determinants for rCLAD remain to be elucidated. Our aim in this study was to establish parameters predicting survival in patients with rCLAD. METHODS All patients diagnosed with rCLAD in 2 lung transplant centers were assessed in a retrospective manner. Various clinical parameters [demography, pulmonary function, bronchoalveolar lavage (BAL), histopathology, radiology and blood differentials] at rCLAD diagnosis were correlated with graft survival using unadjusted and adjusted analysis. RESULTS A total of 53 patients with rCLAD were included with a median graft survival after diagnosis of 1.1 years. Univariate analysis demonstrated that lower-lobe-dominant or diffuse infiltrates on chest computed tomography, presence of an identifiable trigger before rCLAD onset, lymphocytic bronchiolitis, increased BAL neutrophilia, increased BAL eosinophilia and increased blood eosinophils were associated with inferior graft survival after rCLAD diagnosis. Multivariate analysis confirmed the association of location of infiltrates and blood eosinophilia on graft survival. CONCLUSION In this study we have identified parameters associated with graft survival after rCLAD diagnosis that may be useful to predict prognosis.
Collapse
Affiliation(s)
- Stijn E Verleden
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium.
| | - David Ruttens
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Elly Vandermeulen
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Hannelore Bellon
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | | | - Lieven J Dupont
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Christian Benden
- Division of Pulmonary Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Robin Vos
- Leuven Lung Transplant Unit, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
46
|
Dorfmüller P, Kotsimbos T. Lung allograft loss: naming helps seeing... and vice versa! Eur Respir J 2015; 46:1242-6. [PMID: 26521274 DOI: 10.1183/13993003.01526-2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Peter Dorfmüller
- Dept of Pathology and INSERM UMR_S 999, LabEx LERMIT, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Tom Kotsimbos
- Dept of Medicine, Central Clinical School, Monash University, Dept of Allergy, Immunology and Respiratory Medicine, Alfred Hospital, Melbourne, Australia
| |
Collapse
|