1
|
Kim Y, Yeo Y, Kim M, Son YW, Kim J, Kim KL, Kim S, Oh S, Kim Y, Lee H, Park HW, Lee D, Lee SJ, Kang C, Choi H, Park CS, Lee SP, Suh W, Jang JH. A highly mobile adeno-associated virus targeting vascular smooth muscle cells for the treatment of pulmonary arterial hypertension. Nat Biomed Eng 2025:10.1038/s41551-025-01379-8. [PMID: 40301691 DOI: 10.1038/s41551-025-01379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
In pulmonary arterial hypertension (PAH), a phenotypic switch in pulmonary arterial smooth muscle cells (PASMCs) that is primarily caused by aberrant gene regulatory networks can lead to dysregulated vascular remodelling, heart failure or death. No curative therapies for PAH are currently available, presumably because of a lack of viral vectors specifically targeting PASMCs. Here we show that a highly mobile and PASMC-tropic adeno-associated virus variant developed via directed evolution overcomes physical barriers that inhibit its transfer from bronchial airways to vascular layers, ultimately boosting therapeutic efficacy in murine models of PAH. Intratracheal administration of the adeno-associated virus variant carrying a transgene for fibroblast growth factor 12-a key factor regulating the PASMC phenotype-suppressed pulmonary vascular remodelling, prevented the development of PAH in mice and reversed established PAH in rats. The variant's mobility and enhanced tropism for PASMCs may enable curative treatments for PAH.
Collapse
Affiliation(s)
- Yoojin Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
- Nano Science Technology Institute, Yonsei University, Seoul, Korea
| | - Yeongju Yeo
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Minju Kim
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Yong-Wook Son
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Joowon Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Koung Li Kim
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Seohee Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Seokmin Oh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Yunha Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Hyowoo Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Hyun-Woo Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Dongsoo Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Sung Jin Lee
- R&D Center, GluGene Therapeutics Inc., Seoul, Korea
| | | | | | - Chan Soon Park
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seung-Pyo Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Wonhee Suh
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea.
- College of Pharmacy, Chung-Ang University, Seoul, Korea.
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea.
- R&D Center, GluGene Therapeutics Inc., Seoul, Korea.
| |
Collapse
|
2
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
3
|
Fadón-Padilla L, Miranda-Pérez de Alejo C, Miguel-Coello AB, Beraza M, Di Silvio D, Urkola-Arsuaga A, Sánchez-Guisado MJ, Aiestaran-Zelaia I, Fernández-Méndez L, Martinez-Parra L, Ismalaj E, Berra E, Carregal-Romero S, Ruíz-Cabello J. Magnetic nanoradiotracers for targeted neutrophil detection in pulmonary arterial hypertension. J Nanobiotechnology 2024; 22:709. [PMID: 39543655 PMCID: PMC11562838 DOI: 10.1186/s12951-024-03000-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a severe disease characterized by elevated blood pressure in the pulmonary artery that can ultimately damage the right ventricle of the heart. PAH is pathophysiologically heterogeneous, which makes early diagnosis and treatment difficult. Inflammation is thought to be an important factor in the development and progression of this disease and may explain some of the observed interindividual differences. In the context of both acute and chronic inflammation, neutrophil recruitment to the lung has been suggested as a potential biomarker for studying PAH progression. However, there are currently no specific probes for its non-invasive in vivo detection. The imaging-based gold standard for assessing inflammation is [18F] fluorodeoxyglucose (18F-FDG), which is not cell specific. This highlights the urgent need for more specific molecular probes to support personalized medicine. METHODS This study investigated the potential of magnetic nanoradiotracers based on ultrasmall iron oxide nanoparticles, functionalized with N-cinnamoyl-F-(D)L-F-(D)L-F peptide, to detect increased neutrophil infiltration in vivo in different PAH animal models via positron emission tomography. These nanoprobes target formyl peptide receptor 1, which is abundantly expressed in the cell membrane of neutrophils. To assess the benefit of these nanoprobes, their biodistribution was first assessed via magnetic resonance imaging and histology. Then, their lung uptake was compared by positron emission tomography with that of 18F-FDG in two types of PAH animal models with different profiles of inflammation and neutrophil infiltration: monocrotaline and double-hit Sugen-chronic hypoxia PAH rat models. RESULTS Our targeted magnetic nanoradiotracer detected an increase in pulmonary neutrophil infiltration in both PAH models and distinguished between them, which was not possible with 18F-FDG PET. CONCLUSIONS This study underscores the importance of targeted imaging in providing an individualized and longitudinal evaluation of heterogeneous and multifactorial diseases such as PAH. The use of targeted multimodal nanoprobes, for magnetic resonance/positron emission tomography imaging has the potential to facilitate the diagnosis and monitoring of diseases, as well as the development of novel therapies.
Collapse
Affiliation(s)
- Lucía Fadón-Padilla
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, 21201, USA
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Claudia Miranda-Pérez de Alejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Ana Beatriz Miguel-Coello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Marta Beraza
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Desiré Di Silvio
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Ainhize Urkola-Arsuaga
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - María Jesús Sánchez-Guisado
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Irati Aiestaran-Zelaia
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Laura Fernández-Méndez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), Donostia, 20018, Spain
| | - Lydia Martinez-Parra
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
| | - Ermal Ismalaj
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Edurne Berra
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, 4810, Spain
- Centro de investigación en red cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain
| | - Susana Carregal-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain.
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain.
| | - Jesús Ruíz-Cabello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia, 20014, Spain.
- Centro de investigación en red de enfermedades respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Ikerbasque, Basque Foundation for Science Ikerbasque, Bilbao, 48013, Spain.
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| |
Collapse
|
4
|
Rajagopal S, Bogaard HJ, Elbaz MSM, Freed BH, Remy-Jardin M, van Beek EJR, Gopalan D, Kiely DG. Emerging multimodality imaging techniques for the pulmonary circulation. Eur Respir J 2024; 64:2401128. [PMID: 39209480 PMCID: PMC11525339 DOI: 10.1183/13993003.01128-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) remains a challenging condition to diagnose, classify and treat. Current approaches to the assessment of PH include echocardiography, ventilation/perfusion scintigraphy, cross-sectional imaging using computed tomography and magnetic resonance imaging, and right heart catheterisation. However, these approaches only provide an indirect readout of the primary pathology of the disease: abnormal vascular remodelling in the pulmonary circulation. With the advent of newer imaging techniques, there is a shift toward increased utilisation of noninvasive high-resolution modalities that offer a more comprehensive cardiopulmonary assessment and improved visualisation of the different components of the pulmonary circulation. In this review, we explore advances in imaging of the pulmonary vasculature and their potential clinical translation. These include advances in diagnosis and assessing treatment response, as well as strategies that allow reduced radiation exposure and implementation of artificial intelligence technology. These emerging modalities hold the promise of developing a deeper understanding of pulmonary vascular disease and the impact of comorbidities. They also have the potential to improve patient outcomes by reducing time to diagnosis, refining classification, monitoring treatment response and improving our understanding of disease mechanisms.
Collapse
Affiliation(s)
| | - Harm J Bogaard
- Department of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Mohammed S M Elbaz
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin H Freed
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Edwin J R van Beek
- Edinburgh Imaging, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Deepa Gopalan
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit and NIHR Biomedical Research Centre Sheffield, Royal Hallamshire Hospital, Sheffield, UK
| |
Collapse
|
5
|
Andriana P, Fair-Mäkelä R, Liljenbäck H, Kärnä S, Iqbal I, Makrypidi K, Rajander J, Pirmettis I, Li XG, Jalkanen S, Saraste A, Salmi M, Roivainen A. Macrophage mannose receptor CD206 targeting of fluoride-18 labeled mannosylated dextran: A validation study in mice. Eur J Nucl Med Mol Imaging 2024; 51:2216-2228. [PMID: 38532026 PMCID: PMC11178572 DOI: 10.1007/s00259-024-06686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE Aluminum fluoride-18-labeled 1,4,7-triazacyclononane-1,4,7-triacetic acid-conjugated mannosylated dextran derivative (Al[18F]F-NOTA-D10CM) is a new tracer for PET imaging. We report here on in vitro and in vivo validation of the tracer's ability to target the macrophage mannose receptor CD206. METHODS First, the uptake of intravenously (i.v.) administered Al[18F]F-NOTA-D10CM was compared between wild-type (WT) and CD206-/- knockout (KO) mice. C57BL/6N mice were injected with complete Freund's adjuvant (CFA) in the left hind leg and the uptake of Al[18F]F-NOTA-D10CM after i.v. or intradermal (i.d.) injection was studied at 5 and 14 days after CFA induction of inflammation. Healthy C57BL/6N mice were studied as controls. Mice underwent PET/CT on consecutive days with [18F]FDG, i.v. Al[18F]F-NOTA-D10CM, and i.d. Al[18F]F-NOTA-D10CM. After the last imaging, Al[18F]F-NOTA-D10CM was i.v. injected for an ex vivo biodistribution study and autoradiography of inflamed tissues. Blood plasma samples were analyzed using high-performance liquid chromatography. To evaluate the specificity of Al[18F]F-NOTA-D10CM binding, an in vitro competitive displacement study was performed on inflamed tissue sections using autoradiography. CD206 expression was assessed by immunohistochemical staining. RESULTS Compared with WT mice, the uptake of Al[18F]F-NOTA-D10CM was significantly lower in several CD206-/- KO mice tissues, including liver (SUV 8.21 ± 2.51 vs. 1.06 ± 0.16, P < 0.001) and bone marrow (SUV 1.63 ± 0.37 vs. 0.22 ± 0.05, P < 0.0001). The uptake of i.v. injected Al[18F]F-NOTA-D10CM was significantly higher in inflamed ankle joint (SUV 0.48 ± 0.13 vs. 0.18 ± 0.05, P < 0.0001) and inflamed foot pad skin (SUV 0.41 ± 0.10 vs. 0.04 ± 0.01, P < 0.0001) than in the corresponding tissues in healthy mice. The i.d.-injected Al[18F]F-NOTA-D10CM revealed differences between CFA-induced lymph node activation and lymph nodes in healthy mice. Ex vivo γ-counting, autoradiography, and immunohistochemistry supported the results, and a decrease of ~ 80% in the binding of Al[18F]F-NOTA-D10CM in the displacement study with excess NOTA-D10CM confirmed that tracer binding was specific. At 60 min after i.v. injection, an average 96.70% of plasma radioactivity was derived from intact Al[18F]F-NOTA-D10CM, indicating good in vivo stability. The uptake of Al[18F]F-NOTA-D10CM into inflamed tissues was positively associated with the area percentage of CD206-positive staining. CONCLUSION The uptake of mannosylated dextran derivative Al[18F]F-NOTA-D10CM correlated with CD206 expression and the tracer appears promising for inflammation imaging.
Collapse
Affiliation(s)
- Putri Andriana
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Ruth Fair-Mäkelä
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku Center of Disease Modeling, University of Turku, Turku, Finland
| | - Salli Kärnä
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Imran Iqbal
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Konstantina Makrypidi
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Science and Technology, Energy and Safety, NCSR "Demokritos", Athens, Greece
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Turku Center of Disease Modeling, University of Turku, Turku, Finland.
- Turku PET Centre, Turku University Hospital, Turku, Finland.
| |
Collapse
|
6
|
Zuo Y, Li B, Gao M, Xiong R, He R, Li N, Geng Q. Novel insights and new therapeutic potentials for macrophages in pulmonary hypertension. Respir Res 2024; 25:147. [PMID: 38555425 PMCID: PMC10981837 DOI: 10.1186/s12931-024-02772-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Inflammation and immune processes underlie pulmonary hypertension progression. Two main different activated phenotypes of macrophages, classically activated M1 macrophages and alternatively activated M2 macrophages, are both involved in inflammatory processes related to pulmonary hypertension. Recent advances suggest that macrophages coordinate interactions among different proinflammatory and anti-inflammatory mediators, and other cellular components such as smooth muscle cells and fibroblasts. In this review, we summarize the current literature on the role of macrophages in the pathogenesis of pulmonary hypertension, including the origin of pulmonary macrophages and their response to triggers of pulmonary hypertension. We then discuss the interactions among macrophages, cytokines, and vascular adventitial fibroblasts in pulmonary hypertension, as well as the potential therapeutic benefits of macrophages in this disease. Identifying the critical role of macrophages in pulmonary hypertension will contribute to a comprehensive understanding of this pathophysiological abnormality, and may provide new perspectives for pulmonary hypertension management.
Collapse
Affiliation(s)
- Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
7
|
Gandhi SA, Min B, Fazio JC, Johannson KA, Steinmaus C, Reynolds CJ, Cummings KJ. The Impact of Occupational Exposures on the Risk of Idiopathic Pulmonary Fibrosis: A Systematic Review and Meta-Analysis. Ann Am Thorac Soc 2024; 21:486-498. [PMID: 38096107 PMCID: PMC10913770 DOI: 10.1513/annalsats.202305-402oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic pulmonary disorder of unknown etiology that is characterized by a usual interstitial pneumonia pattern. Previous meta-analyses have reported associations between occupational exposures and IPF, but higher-quality studies have been published in recent years, doubling the number of studied patients. Objectives: To provide a contemporary and comprehensive assessment of the relationship between occupational exposures and IPF. Methods: We searched PubMed, Embase, and Web of Science through July 2023 to identify all publications on occupational exposure and IPF. We conducted a meta-analysis of the occupational burden, odds ratio (OR), and population attributable fraction (PAF) of exposures. Five exposure categories were analyzed: vapors, gas, dust, and fumes (VGDF); metal dust; wood dust; silica dust; and agricultural dust. A comprehensive bias assessment was performed. The study protocol was registered in the International Prospective Register of Systematic Reviews (identifier CRD42021267808). Results: Our search identified 23,942 publications. Sixteen publications contained relative risks needed to calculate pooled ORs and PAFs, and 12 additional publications reported an occupational burden within a case series. The proportion of cases with occupational exposures to VGDF was 44% (95% confidence interval [CI], 36-53%), with a range of 8-17% within more specific exposure categories. The pooled OR was increased for VGDF at 1.8 (95% CI, 1.3-2.4), with a pooled PAF of 21% (95% CI, 15-28%). ORs and PAFs, respectively, were found to be 1.6 and 7% for metal dust, 1.6 and 3% for wood dust, 1.8 and 14% for agricultural dust, and 1.8 and 4% for silica dust. The pooled ORs and PAFs within specific exposure categories ranged from 1.6 to 1.8 and from 4% to 14%, respectively. We identified some publication bias, but it was not sufficient to diminish the association between occupational exposures and IPF based on sensitivity analysis and bias assessment. Conclusions: Our findings indicate that 21% of IPF cases (or approximately one in five) could be prevented by removal of occupational exposure (alongside a pooled OR of 1.8). Additionally, 44% of patients with IPF report occupational exposure to VGDF. This meta-analysis suggests that a considerable number of cases of IPF are attributable to inhaled occupational exposures and warrant increased consideration in the clinical care of patients and future prevention efforts.
Collapse
Affiliation(s)
- Sheiphali A. Gandhi
- Division of Occupational, Environmental, and Climate Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Bohyung Min
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jane C. Fazio
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | | | - Craig Steinmaus
- School of Public Health, University of California, Berkeley, Berkeley, California
| | - Carl J. Reynolds
- Faculty of Medicine, National Heart and Lung Institute, Imperial College of London, London, United Kingdom; and
| | - Kristin J. Cummings
- Occupational Health Branch, California Department of Public Health, Richmond, California
| |
Collapse
|
8
|
Gondry O, Xavier C, Raes L, Heemskerk J, Devoogdt N, Everaert H, Breckpot K, Lecocq Q, Decoster L, Fontaine C, Schallier D, Aspeslagh S, Vaneycken I, Raes G, Van Ginderachter JA, Lahoutte T, Caveliers V, Keyaerts M. Phase I Study of [ 68Ga]Ga-Anti-CD206-sdAb for PET/CT Assessment of Protumorigenic Macrophage Presence in Solid Tumors (MMR Phase I). J Nucl Med 2023; 64:1378-1384. [PMID: 37474271 PMCID: PMC10478821 DOI: 10.2967/jnumed.122.264853] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/05/2023] [Indexed: 07/22/2023] Open
Abstract
Macrophages play an important role throughout the body. Antiinflammatory macrophages expressing the macrophage mannose receptor (MMR, CD206) are involved in disease development, ranging from oncology to atherosclerosis and rheumatoid arthritis. [68Ga]Ga-NOTA-anti-CD206 single-domain antibody (sdAb) is a PET tracer targeting CD206. This first-in-human study, as its primary objective, evaluated the safety, biodistribution, and dosimetry of this tracer. The secondary objective was to assess its tumor uptake. Methods: Seven patients with a solid tumor of at least 10 mm, an Eastern Cooperative Oncology Group score of 0 or 1, and good renal and hepatic function were included. Safety was evaluated using clinical examination and blood sampling before and after injection. For biodistribution and dosimetry, PET/CT was performed at 11, 90, and 150 min after injection; organs showing tracer uptake were delineated, and dosimetry was evaluated. Blood samples were obtained at selected time points for blood clearance. Metabolites in blood and urine were assessed. Results: Seven patients were injected with, on average, 191 MBq of [68Ga]Ga-NOTA-anti-CD206-sdAb. Only 1 transient adverse event of mild severity was considered to be possibly, although unlikely, related to the study drug (headache, Common Terminology Criteria for Adverse Events grade 1). The blood clearance was fast, with less than 20% of the injected activity remaining after 80 min. There was uptake in the liver, kidneys, spleen, adrenals, and red bone marrow. The average effective dose from the radiopharmaceutical was 4.2 mSv for males and 5.2 mSv for females. No metabolites were detected. Preliminary data of tumor uptake in cancer lesions showed higher uptake in the 3 patients who subsequently progressed than in the 3 patients without progression. One patient could not be evaluated because of technical failure. Conclusion: [68Ga]Ga-NOTA-anti-CD206-sdAb is safe and well tolerated. It shows rapid blood clearance and renal excretion, enabling high contrast-to-noise imaging at 90 min after injection. The radiation dose is comparable to that of routinely used PET tracers. These findings and the preliminary results in cancer patients warrant further investigation of this tracer in phase II clinical trials.
Collapse
Affiliation(s)
- Odrade Gondry
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Catarina Xavier
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurens Raes
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johannes Heemskerk
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nick Devoogdt
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hendrik Everaert
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christel Fontaine
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Denis Schallier
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse Vaneycken
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology, Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; and
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Tony Lahoutte
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vicky Caveliers
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- MIMA, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
9
|
Sunwoo SH, Cha MJ, Han SI, Kang H, Cho YS, Yeom DH, Park CS, Park NK, Choi SW, Kim SJ, Cha GD, Jung D, Choi S, Oh S, Nam GB, Hyeon T, Kim DH, Lee SP. Ventricular tachyarrhythmia treatment and prevention by subthreshold stimulation with stretchable epicardial multichannel electrode array. SCIENCE ADVANCES 2023; 9:eadf6856. [PMID: 37000879 PMCID: PMC10065438 DOI: 10.1126/sciadv.adf6856] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/28/2023] [Indexed: 05/24/2023]
Abstract
The implantable cardioverter-defibrillator (ICD) is an effective method to prevent sudden cardiac death in high-risk patients. However, the transvenous lead is incompatible with large-area electrophysiological mapping and cannot accommodate selective multichannel precision stimulations. Moreover, it involves high-energy shocks, resulting in pain, myocardial damage, and recurrences of ventricular tachyarrhythmia (VTA). We present a method for VTA treatment based on subthreshold electrical stimulations using a stretchable epicardial multichannel electrode array, which does not disturb the normal contraction or electrical propagation of the ventricle. In rabbit models with myocardial infarction, the infarction was detected by mapping intracardiac electrograms with the stretchable epicardial multichannel electrode array. Then, VTAs could be terminated by sequential electrical stimuli from the epicardial multichannel electrode array beginning with low-energy subthreshold stimulations. Last, we used these subthreshold stimulations to prevent the occurrence of additional VTAs. The proposed protocol using the stretchable epicardial multichannel electrode array provides opportunities toward the development of innovative methods for painless ICD therapy.
Collapse
MESH Headings
- Rabbits
- Animals
- Tachycardia, Ventricular/therapy
- Tachycardia, Ventricular/epidemiology
- Tachycardia, Ventricular/etiology
- Defibrillators, Implantable/adverse effects
- Heart Ventricles
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Death, Sudden, Cardiac/epidemiology
- Myocardial Infarction/therapy
- Myocardial Infarction/etiology
Collapse
Affiliation(s)
- Sung-Hyuk Sunwoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Myung-Jin Cha
- Departments of Cardiology and Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Sang Ihn Han
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyejeong Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ye Seul Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Da-Hae Yeom
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Chan Soon Park
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Na Kyeong Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seong Woo Choi
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Gi Doo Cha
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongjun Jung
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Suji Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seil Oh
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Gi-Byoung Nam
- Departments of Cardiology and Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung-Pyo Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
10
|
Gu GJ, Chung H, Park JY, Yoo R, Im HJ, Choi H, Lee YS, Seok SH. Mannosylated-serum albumin nanoparticle imaging to monitor tumor-associated macrophages under anti-PD1 treatment. J Nanobiotechnology 2023; 21:31. [PMID: 36707872 PMCID: PMC9881286 DOI: 10.1186/s12951-023-01791-9] [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: 10/27/2022] [Accepted: 01/21/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors such as anti-programmed cell death protein 1 (PD1) block tumor growth by reinvigorating the immune system; however, determining their efficacy only by the changes in tumor size may prove inaccurate. As the immune cells including macrophages in the tumor microenvironment (TME) are associated with the response to anti-PD1 therapy, tumor-associated macrophages (TAMs) imaging using nanoparticles can noninvasively provide the immune enrichment status of TME. Herein, the mannosylated-serum albumin (MSA) nanoparticle was labeled with radioactive isotope 68Ga to target the mannose receptors on macrophages for noninvasive monitoring of the TME according to anti-PD1 therapy. RESULTS B16F10-Luc and MC38-Luc tumor-bearing mice were treated with anti-PD1, and the response to anti-PD1 was determined by the tumor volume. According to the flow cytometry, the responders to anti-PD1 showed an increased proportion of TAMs, as well as lymphocytes, and the most enriched immune cell population in the TME was also TAMs. For noninvasive imaging of TAMs as a surrogate of immune cell augmentation in the TME via anti-PD1, we acquired [68Ga] Ga-MSA positron emission tomography. According to the imaging study, an increased number of TAMs in responders at the early phase of anti-PD1 treatment was observed in both B16F10-Luc and MC38-Luc tumor-bearing mice models. CONCLUSION As representative immune cells in the TME, non-invasive imaging of TAMs using MSA nanoparticles can reflect the immune cell enrichment status in the TME closely associated with the response to anti-PD1. As non-invasive imaging using MSA nanoparticles, this approach shows a potential to monitor and evaluate anti-tumor response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gyo Jeong Gu
- grid.31501.360000 0004 0470 5905Macrophage Laboratory, Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyewon Chung
- grid.31501.360000 0004 0470 5905Macrophage Laboratory, Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Bio-MAX Institute, Seoul National University, Seoul, Republic of Korea
| | - Ji Yong Park
- grid.31501.360000 0004 0470 5905Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ranji Yoo
- grid.31501.360000 0004 0470 5905Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.412484.f0000 0001 0302 820X Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyung-Jun Im
- grid.31501.360000 0004 0470 5905Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongyoon Choi
- grid.31501.360000 0004 0470 5905Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Radiation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.412484.f0000 0001 0302 820XDepartment of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- grid.31501.360000 0004 0470 5905Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Radiation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Seok
- grid.31501.360000 0004 0470 5905Macrophage Laboratory, Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Park CS, Kim SH, Yang HY, Kim JH, Schermuly RT, Cho YS, Kang H, Park JH, Lee E, Park H, Yang JM, Noh TW, Lee SP, Bae SS, Han J, Ju YS, Park JB, Kim I. Sox17 Deficiency Promotes Pulmonary Arterial Hypertension via HGF/c-Met Signaling. Circ Res 2022; 131:792-806. [PMID: 36205124 PMCID: PMC9612711 DOI: 10.1161/circresaha.122.320845] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND In large-scale genomic studies, Sox17, an endothelial-specific transcription factor, has been suggested as a putative causal gene of pulmonary arterial hypertension (PAH); however, its role and molecular mechanisms remain to be elucidated. We investigated the functional impacts and acting mechanisms of impaired Sox17 (SRY-related HMG-box17) pathway in PAH and explored its potential as a therapeutic target. METHODS In adult mice, Sox17 deletion in pulmonary endothelial cells (ECs) induced PAH under hypoxia with high penetrance and severity, but not under normoxia. RESULTS Key features of PAH, such as hypermuscularization, EC hyperplasia, and inflammation in lung arterioles, right ventricular hypertrophy, and elevated pulmonary arterial pressure, persisted even after long rest in normoxia. Mechanistically, transcriptomic profiling predicted that the combination of Sox17 deficiency and hypoxia activated c-Met signaling in lung ECs. HGF (hepatocyte grow factor), a ligand of c-Met, was upregulated in Sox17-deficient lung ECs. Pharmacologic inhibition of HGF/c-Met signaling attenuated and reversed the features of PAH in both preventive and therapeutic settings. Similar to findings in animal models, Sox17 levels in lung ECs were repressed in 26.7% of PAH patients (4 of 15), while those were robust in all 14 non-PAH controls. HGF levels in pulmonary arterioles were increased in 86.7% of patients with PAH (13 of 15), but none of the controls showed that pattern. CONCLUSIONS The downregulation of Sox17 levels in pulmonary arterioles increases the susceptibility to PAH, particularly when exposed to hypoxia. Our findings suggest the reactive upregulation of HGF/c-Met signaling as a novel druggable target for PAH treatment.
Collapse
Affiliation(s)
- Chan Soon Park
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.).,Division of Cardiology, Department of Internal Medicine (C.S.P., Y.S.C., H.K., S.-P.L., J.-B.P.)
| | - Soo Hyun Kim
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - Hae Young Yang
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - Ju-Hee Kim
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the German Center for Lung Research (DZL), Germany (R.T.S.)
| | - Ye Seul Cho
- Division of Cardiology, Department of Internal Medicine (C.S.P., Y.S.C., H.K., S.-P.L., J.-B.P.)
| | - Hyejeong Kang
- Division of Cardiology, Department of Internal Medicine (C.S.P., Y.S.C., H.K., S.-P.L., J.-B.P.).,Center for Precision Medicine, Seoul National University Hospital and Seoul National University College of Medicine, Republic of Korea (H.K., S.-P.L.)
| | - Jae-Hyeong Park
- Division of Cardiology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea (J.-H.P.)
| | - Eunhyeong Lee
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - HyeonJin Park
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - Jee Myung Yang
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.).,Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang, South Korea (J.MY.)
| | - Tae Wook Noh
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.)
| | - Seung-Pyo Lee
- Division of Cardiology, Department of Internal Medicine (C.S.P., Y.S.C., H.K., S.-P.L., J.-B.P.).,Center for Precision Medicine, Seoul National University Hospital and Seoul National University College of Medicine, Republic of Korea (H.K., S.-P.L.).,Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.-P.L.)
| | - Sun Sik Bae
- Department of Pharmacology, Pusan National University School of Medicine, Busan, Republic of Korea (S.S.B.)
| | - Jinju Han
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.).,Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea (J.H., Y.S.J., I.K.)
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.).,Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea (J.H., Y.S.J., I.K.)
| | - Jun-Bean Park
- Division of Cardiology, Department of Internal Medicine (C.S.P., Y.S.C., H.K., S.-P.L., J.-B.P.)
| | - Injune Kim
- Graduate School of Medical Science and Engineering (C.S.P., S.H.K., H.Y.Y., J.-H.K., E.L., H.P., J.M.Y., T.W.N., J.H., Y.S.J., I.K.).,Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea (J.H., Y.S.J., I.K.)
| |
Collapse
|
12
|
Alenezi F, Covington TA, Mukherjee M, Mathai SC, Yu PB, Rajagopal S. Novel Approaches to Imaging the Pulmonary Vasculature and Right Heart. Circ Res 2022; 130:1445-1465. [PMID: 35482838 PMCID: PMC9060389 DOI: 10.1161/circresaha.121.319990] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There is an increased appreciation for the importance of the right heart and pulmonary circulation in several disease states across the spectrum of pulmonary hypertension and left heart failure. However, assessment of the structure and function of the right heart and pulmonary circulation can be challenging, due to the complex geometry of the right ventricle, comorbid pulmonary airways and parenchymal disease, and the overlap of hemodynamic abnormalities with left heart failure. Several new and evolving imaging modalities interrogate the right heart and pulmonary circulation with greater diagnostic precision. Echocardiographic approaches such as speckle-tracking and 3-dimensional imaging provide detailed assessments of regional systolic and diastolic function and volumetric assessments. Magnetic resonance approaches can provide high-resolution views of cardiac structure/function, tissue characterization, and perfusion through the pulmonary vasculature. Molecular imaging with positron emission tomography allows an assessment of specific pathobiologically relevant targets in the right heart and pulmonary circulation. Machine learning analysis of high-resolution computed tomographic lung scans permits quantitative morphometry of the lung circulation without intravenous contrast. Inhaled magnetic resonance imaging probes, such as hyperpolarized 129Xe magnetic resonance imaging, report on pulmonary gas exchange and pulmonary capillary hemodynamics. These approaches provide important information on right ventricular structure and function along with perfusion through the pulmonary circulation. At this time, the majority of these developing technologies have yet to be clinically validated, with few studies demonstrating the utility of these imaging biomarkers for diagnosis or monitoring disease. These technologies hold promise for earlier diagnosis and noninvasive monitoring of right heart failure and pulmonary hypertension that will aid in preclinical studies, enhance patient selection and provide surrogate end points in clinical trials, and ultimately improve bedside care.
Collapse
Affiliation(s)
- Fawaz Alenezi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | | | | | - Steve C. Mathai
- Johns Hopkins Division of Pulmonary and Critical Care Medicine, Baltimore, MD
| | - Paul B. Yu
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| |
Collapse
|
13
|
Farrell C, Balasubramanian A, Hays AG, Hsu S, Rowe S, Zimmerman SL, Hassoun PM, Mathai SC, Mukherjee M. A Clinical Approach to Multimodality Imaging in Pulmonary Hypertension. Front Cardiovasc Med 2022; 8:794706. [PMID: 35118142 PMCID: PMC8804287 DOI: 10.3389/fcvm.2021.794706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a clinical condition characterized by progressive elevations in mean pulmonary artery pressures and right ventricular dysfunction, associated with significant morbidity and mortality. For resting PH to develop, ~50-70% of the pulmonary vasculature must be affected, suggesting that even mild hemodynamic abnormalities are representative of advanced pulmonary vascular disease. The definitive diagnosis of PH is based upon hemodynamics measured by right heart catheterization; however this is an invasive and resource intense study. Early identification of pulmonary vascular disease offers the opportunity to improve outcomes by instituting therapies that slow, reverse, or potentially prevent this devastating disease. Multimodality imaging, including non-invasive modalities such as echocardiography, computed tomography, ventilation perfusion scans, and cardiac magnetic resonance imaging, has emerged as an integral tool for screening, classifying, prognosticating, and monitoring response to therapy in PH. Additionally, novel imaging modalities such as echocardiographic strain imaging, 3D echocardiography, dual energy CT, FDG-PET, and 4D flow MRI are actively being investigated to assess the severity of right ventricular dysfunction in PH. In this review, we will describe the utility and clinical application of multimodality imaging techniques across PH subtypes as it pertains to screening and monitoring of PH.
Collapse
Affiliation(s)
- Christine Farrell
- Division of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Aparna Balasubramanian
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Allison G. Hays
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Steven Hsu
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Steven Rowe
- Division of Radiology, Johns Hopkins University, Baltimore, MD, United States
| | - Stefan L. Zimmerman
- Division of Radiology, Johns Hopkins University, Baltimore, MD, United States
| | - Paul M. Hassoun
- Division of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Stephen C. Mathai
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Monica Mukherjee
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
14
|
Li XG, Velikyan I, Viitanen R, Roivainen A. PET radiopharmaceuticals for imaging inflammatory diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
15
|
Cho SG, Kong EJ, Kang WJ, Paeng JC, Bom HSH, Cho I. KSNM60 in Cardiology: Regrowth After a Long Pause. Nucl Med Mol Imaging 2021; 55:151-161. [PMID: 34422125 PMCID: PMC8322215 DOI: 10.1007/s13139-021-00702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/09/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022] Open
Abstract
The Korean Society of Nuclear Medicine (KSNM) is celebrating its 60th anniversary in honor of the nuclear medicine professionals who have dedicated their efforts towards research, academics, and the more comprehensive clinical applications and uses of nuclear imaging modalities. Nuclear cardiology in Korea was at its prime time in the 1990s, but its growth was interrupted by a long pause. Despite the academic and practical challenges, nuclear cardiology in Korea now meets the second leap, attributed to the growth in molecular imaging tailored for many non-coronary diseases and the genuine values of nuclear myocardial perfusion imaging. In this review, we describe the trends, achievements, challenges, and perspectives of nuclear cardiology throughout the 60-year history of the KSNM.
Collapse
Affiliation(s)
- Sang-Geon Cho
- Department of Nuclear Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Eun Jung Kong
- Department of Nuclear Medicine, Yeungnam University Medical Center, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415 Republic of Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Yonsei University Severance Hospital, Seoul, Republic of Korea
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hee-Seung Henry Bom
- 5Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea
| | - Ihnho Cho
- Department of Nuclear Medicine, Yeungnam University Medical Center, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415 Republic of Korea
| |
Collapse
|
16
|
Li S, Ma Y, Yan Y, Yan M, Wang X, Gong W, Nie S. Phosphodiesterase-5a Knock-out Suppresses Inflammation by Down-Regulating Adhesion Molecules in Cardiac Rupture Following Myocardial Infarction. J Cardiovasc Transl Res 2021; 14:816-823. [PMID: 33496888 DOI: 10.1007/s12265-021-10102-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/11/2021] [Indexed: 11/28/2022]
Abstract
Cardiac rupture is a fatal complication of acute myocardial infarction (MI), associated with increased inflammation and damaged extracellular matrix. C57BL/6 J wild type (WT) and Pde5a knockout (Pde5a-/-) mice were selected to establish MI model. The rupture rate of Pde5a-/- mice was significantly reduced (P < 0.01) within 7 days post MI. The cardiac function of Pde5a-/- mice was better than WT mice both at day 3 and 7 post MI. Immunohistochemical staining and flow cytometry showed neutrophils and macrophages were decreased in Pde5a-/- mouse hearts. Inflammatory factors expression such as IL-1β, IL-6, IL-8, Mcp-1, TNF-α significantly decreased in Pde5a-/- mice post MI. Moreover, western blot showed the inhibition of inflammatory response was accompanied by down-regulation of intercellular adhesion molecule-1(ICAM-1) and vascular cell adhesion molecule-1(VCAM-1) in Pde5a-/- mice. Knockout of Pde5a reduced inflammatory cells infiltration by down-regulating the expression of ICAM-1 and VCAM-1, and prevented early cardiac rupture after MI. All authors declare that they have no conflicts of interest. This article does not contain any studies with human participants performed by any of the authors. All applicable international, national, and institutional guidelines for the care and use of animals were followed.
Collapse
Affiliation(s)
- Siyi Li
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Youcai Ma
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Yan Yan
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Mengwen Yan
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Xiao Wang
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Wei Gong
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China. .,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China.
| | - Shaoping Nie
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing, 100029, China. .,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
17
|
Hemnes A, Rothman AMK, Swift AJ, Zisman LS. Role of biomarkers in evaluation, treatment and clinical studies of pulmonary arterial hypertension. Pulm Circ 2020; 10:2045894020957234. [PMID: 33282185 PMCID: PMC7682212 DOI: 10.1177/2045894020957234] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension is a complex disease resulting from the interplay of myriad biological and environmental processes that lead to remodeling of the pulmonary vasculature with consequent pulmonary hypertension. Despite currently available therapies, there remains significant morbidity and mortality in this disease. There is great interest in identifying and applying biomarkers to help diagnose patients with pulmonary arterial hypertension, inform prognosis, guide therapy, and serve as surrogate endpoints. An extensive literature on potential biomarker candidates is available, but barriers to the implementation of biomarkers for clinical use in pulmonary arterial hypertension are substantial. Various omic strategies have been undertaken to identify key pathways regulated in pulmonary arterial hypertension that could serve as biomarkers including genomic, transcriptomic, proteomic, and metabolomic approaches. Other biologically relevant components such as circulating cells, microRNAs, exosomes, and cell-free DNA have recently been gaining attention. Because of the size of the datasets generated by these omic approaches and their complexity, artificial intelligence methods are being increasingly applied to decipher their meaning. There is growing interest in imaging the lung with various modalities to understand and visualize processes in the lung that lead to pulmonary vascular remodeling including high resolution computed tomography, Xenon magnetic resonance imaging, and positron emission tomography. Such imaging modalities have the potential to demonstrate disease modification resulting from therapeutic interventions. Because right ventricular function is a major determinant of prognosis, imaging of the right ventricle with echocardiography or cardiac magnetic resonance imaging plays an important role in the evaluation of patients and may also be useful in clinical studies of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Anna Hemnes
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Andrew J Swift
- University of Sheffield and Sheffield Teaching Hospitals NHS Trust, Sheffield, UK
| | | |
Collapse
|
18
|
Wolf M, Montesi SB. Novel Imaging Strategies in Systemic Sclerosis. Curr Rheumatol Rep 2020; 22:57. [PMID: 32785794 DOI: 10.1007/s11926-020-00926-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW Imaging modalities such as computed tomography, ultrasound, magnetic resonance imaging, and molecular imaging are being used to evaluate for disease in systemic sclerosis (SSc) patients. Here, we review novel imaging strategies to detect organ and vascular complications of SSc and novel imaging techniques for assessing interstitial lung disease and pulmonary hypertension in other conditions that may have further applicability to SSc. RECENT FINDINGS Imaging techniques can be used to identify disease in the lungs, pulmonary vascular system, heart, skin, vascular tissue, and gastrointestinal tract of SSc patients. These show promise in detecting early disease, many without the use of ionizing radiation. Novel imaging techniques in patients with SSc can be used to detect disease in multiple susceptible organs. These imaging strategies have potential for early disease detection, as well as potential for incorporation into clinical trials to accelerate the development of SSc therapies.
Collapse
Affiliation(s)
- Molly Wolf
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02116, USA.,Harvard Medical School, Boston, MA, USA
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02116, USA. .,Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Tang R, Jin Y, Liao J, Chen L, Liu C, Li S, Wang J, Wang T, Lu W. Positron Emission Tomography: A Novel Approach to Detect Pulmonary Artery Hypertension at the Early Stage? Am J Respir Crit Care Med 2020; 202:474-475. [PMID: 32267733 PMCID: PMC7397800 DOI: 10.1164/rccm.202002-0314le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Ruidi Tang
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Yingkang Jin
- Guangzhou Women and Children’s Medical CenterGuangzhou, China
| | - Jing Liao
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Lingdan Chen
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Chunli Liu
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Shaoqiang Li
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Jian Wang
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Tao Wang
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| | - Wenju Lu
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Chinaand
| |
Collapse
|
20
|
Park JB, Paeng JC, Lee SP. Reply to Tang et al.: Positron Emission Tomography: A Novel Approach to Detect Pulmonary Artery Hypertension at the Early Stage? Am J Respir Crit Care Med 2020; 202:475-476. [PMID: 32353249 PMCID: PMC7397794 DOI: 10.1164/rccm.202003-0557le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Jun-Bean Park
- Seoul National University HospitalSeoul, Republic of Koreaand
- Seoul National University College of MedicineSeoul, Republic of Korea
| | - Jin Chul Paeng
- Seoul National University HospitalSeoul, Republic of Koreaand
- Seoul National University College of MedicineSeoul, Republic of Korea
| | - Seung-Pyo Lee
- Seoul National University HospitalSeoul, Republic of Koreaand
- Seoul National University College of MedicineSeoul, Republic of Korea
| |
Collapse
|
21
|
Goncharova EA, Chan SY, Ventetuolo CE, Weissmann N, Schermuly RT, Mullin CJ, Gladwin MT. Update in Pulmonary Vascular Diseases and Right Ventricular Dysfunction 2019. Am J Respir Crit Care Med 2020; 202:22-28. [PMID: 32311291 PMCID: PMC7328315 DOI: 10.1164/rccm.202003-0576up] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elena A. Goncharova
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Center for Pulmonary Vascular Biology and Medicine, and
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Medicine, Alpert Medical School, and
| | - Corey E. Ventetuolo
- Department of Medicine, Alpert Medical School, and
- Department of Health Services, Policy, and Practice, School of Public Health, Brown University, Providence, Rhode Island; and
| | - Norbert Weissmann
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | | | - Mark T. Gladwin
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine
| |
Collapse
|
22
|
Affiliation(s)
- Delphine L Chen
- Seattle Cancer Care AllianceUniversity of WashingtonSeattle, Washington
| |
Collapse
|